AVERY’S DISEASES
OF THE NEWBORN
Ninth Edition
Christine A. Gleason, MD
W. Alan Hodson Endowed Chair in Pediatrics
Professor of Pediatrics
Head, Division of Neonatology
Department of Pediatrics
University of Washington
Seattle Children’s Hospital
Seattle, Washington
Sherin U. Devaskar, MD
Mattel Endowed Executive Chair and Distinguished Professor
Department of Pediatrics
David Geffen School of Medicine
Assistant Vice Chancellor for Children’s Health
University of California, Los Angeles Health System
Physician in Chief
Mattel Children’s Hospital
Los Angeles, California
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899
AVERY’S DISEASES OF THE NEWBORN
ISBN: 978-1-4377-0134-0
Copyright © 2012, 2005, 1998, 1991, 1984, 1977, 1971, 1965, 1960 by Saunders, an imprint
of Elsevier Inc.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies, and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).
Notice
Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treatment
may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating
and using any information, methods, compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the safety of others, including parties
for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the most
current information provided (i) on procedures featured or (ii) by the manufacturer of each product to
be administered, to verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the responsibility of practitioners, relying on their own experience and
knowledge of their patients, to make diagnoses, to determine dosages and the best treatment for each
individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors
assume any liability for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.
Library of Congress Cataloging-in-Publication Data
Avery’s diseases of the newborn. -- 9th ed. / [edited by] Christine A. Gleason, Sherin U. Devaskar.
p. ; cm.
Diseases of the newborn
Includes bibliographical references and index.
ISBN 978-1-4377-0134-0 (pbk. : alk. paper) 1. Newborn infants--Diseases. I. Gleason, Christine A. II.
Devaskar, Sherin U. III. Avery, Mary Ellen, 1927- IV. Title: Diseases of the newborn.
[DNLM: 1. Infant, Newborn, Diseases. WS 421]
RJ254.S3 2012
618.92’01--dc23
2011020667
Acquisitions Editor: Judith Fletcher
Developmental Editor: Dee Simpson
Publishing Services Manager: Anne Altepeter
Associate Project Manager: Jessica L. Becher
Design Direction: Steve Stave
Printed in the United States of America
Last digit is the print number: 9 8
7
6
5 4
3
2
1
CONTRIBUTORS
Steven H. Abman, MD
Professor
Department of Pediatrics
University of Colorado School of Medicine
Director
Pediatric Heart Lung Center
Co-Director
Pulmonary Hypertension Program
The Children’s Hospital
Aurora, Colorado
Amina Ahmed, MD
Pediatric Infectious Disease
Department of Pediatrics
Carolinas Medical Center
Levine Children’s Hospital
Charlotte, North Carolina
Adjunct Clinical Associate Professor
Department of Pediatrics
University of North Carolina
Chapel Hill, North Carolina
Marilee C. Allen, MD
Professor of Pediatrics
Johns Hopkins University School of Medicine
Co-Director
Neonatal Intensive Care Unit Developmental
Clinic
Kennedy Krieger Institute
Baltimore, Maryland
David Askenazi, MD, MSPH
Assistant Professor
Division of Nephrology and Transplantation
Department of Pediatrics
University of Alabama at Birmingham
Birmingham, Alabama
Stephen A. Back, MD, PhD
Associate Professor of Pediatrics and
Neurology
Oregon Health and Science University
Clyde and Elda Munson Professor of
Pediatric Research
Director
Neuroscience Section
Papé Family Pediatric Research Institute
Portland, Oregon
H. Scott Baldwin, MD
Professor of Pediatrics and Cell and
Developmental Biology
Vanderbilt University Medical Center
Chief
Division of Pediatric Cardiology
Co-Director
Pediatric Heart Institute
Monroe Carell Jr. Children’s Hospital
at Vanderbilt
Nashville, Tennessee
Roberta A. Ballard, MD
Professor
Department of Pediatrics and Neonatology
University of California, San Francisco School
of Medicine
San Francisco, California
Eduardo Bancalari, MD
Professor of Pediatrics
Director
Division of Neonatology
University of Miami Miller School
of Medicine
Chief
Newborn Service
Jackson Memorial Hospital
Miami, Florida
Carlton M. Bates, MD
Associate Professor
Department of Pediatrics
University of Pittsburgh School of Medicine
Chief and Program Director
Pediatric Nephrology
Children’s Hospital of Pittsburgh
Rangos Research Building
Pittsburgh, Pennsylvania
Diana W. Bianchi, MD
Natalie V. Zucker Professor of Pediatrics,
Obstetrics and Gynecology
Tufts University School of Medicine
Vice Chair for Research
Department of Pediatrics
Floating Hospital for Children
Boston, Massachusetts
Gil Binenbaum, MD, MSCE
Assistant Professor
Department of Ophthalmology
University of Pennsylvania School of
Medicine
Attending Surgeon
Department of Ophthalmology
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Sureka Bollepalli, MD
Assistant Professor
Department of Pediatrics
University of South Florida Diabetes Center
Tampa, Florida
Sonia L. Bonifacio, MD
Associate Professor of Pediatrics
Division of Pediatric Nephrology
Emory University School of Medicine
Director
Pediatric Hypertension Program
Children’s Healthcare of Atlanta
Atlanta, Georgia
Assistant Adjunct Professor
Department of Pediatrics and Neonatology
University of California, San Francisco School
of Medicine
Co-Director
Neurological Intensive Care Nursery
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Stephen Baumgart, MD
Mitchell S. Cairo, MD
Donald L. Batisky, MD
Professor of Pediatrics
Department of Neonatology
George Washington University School
of Medicine and Health Sciences
Children’s National Medical Center
Washington, DC
Thomas J. Benedetti, MD, MHA
Professor
Department of Obstetrics and Gynecology
University of Washington School of Medicine
Seattle, Washington
Gerard T. Berry, MD
Professor
Department of Pediatrics Harvard Medical
School
Director
Metabolism Program
Division of Genetics
Children’s Hospital Boston
Boston, Massachusetts
Professor of Pediatrics and Medicine
and Pathology
Chief
Division of Blood and Marrow
Transplantation
Department of Pediatrics
New York-Presbyterian Morgan Stanley
Children’s Hospital
Columbia University Medical Center
New York, New York
Katherine H. Campbell, MD, MPH
Fellow
Department of Obstetrics, Gynecology,
and Reproductive Sciences
Yale School of Medicine
New Haven, Connecticut
v
vi
CONTRIBUTORS
Michael Caplan, MD
Chairman
Department of Pediatrics
NorthShore University HealthSystem
Evanston, Illinois
Professor
Department of Pediatrics
University of Chicago Pritzker School
of Medicine
Chicago, Illinois
Stephen Cederbaum, MD
Professor Emeritus
Departments of Psychiatry and Pediatrics
and Human Genetics
University of California, Los Angeles
Attending Physician
Department of Pediatrics
Ronald Reagan UCLA Medical Center
Los Angeles, California
Consulting Physician
Department of Pediatrics
Santa Monica UCLA Medical Center
Santa Monica, California
Sudhish Chandra, MD, FAAP
Medical Director
Department of Neonatology
Neonatal Intensive Care Unit
St. Anthony Medical Center
Crown Point, Indiana
Ming Chen, MD, PhD
Assistant Professor
Department of Pediatrics
University of Michigan
Ann Arbor, Michigan
Nelson Claure, MSc, PhD
Research Associate Professor of Pediatrics
Director
Neonatal Pulmonary Research Laboratory
Department of Pediatrics
Division of Neonatology
University of Miami Miller School
of Medicine
Miami, Florida
Ronald I. Clyman, MD
F. Sessions Cole, MD
Park J. White, MD, Professor of Pediatrics
Assistant Vice Chancellor for Children’s
Health
Director
Division of Newborn Medicine
Washington University School of Medicine
Chief Medical Officer
St. Louis Children’s Hospital
St. Louis, Missouri
Lawrence Copelovitch, MD
Assistant Professor
University of Pennsylvania School of
Medicine
Attending in Nephrology
Department of Pediatrics
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Michael Cunningham, MD, PhD
Professor and Chief
Division of Craniofacial Medicine
Department of Pediatrics
University of Washington
Medical Director
Craniofacial Center
Seattle Children’s Hospital
Seattle, Washington
Alejandra G. de Alba Campomanes,
MD, MPH
Assistant Professor of Ophthalmology
Division of Pediatric Ophthalmology and
Strabismus
University of California, San Francisco
Director
Department of Pediatric Ophthalmology
and Strabismus
San Francisco General Hospital
San Francisco, California
Ellen Dees, MD
Assistant Professor of Pediatrics
Division of Pediatric Cardiology
Monroe Carell Jr. Children’s Hospital
at Vanderbilt
Nashville, Tennessee
Professor
Department of Pediatrics
Senior Staff
Cardiovascular Research Institute
University of California, San Francisco
San Francisco, California
Scott C. Denne, MD
Bernard A. Cohen, MD
Mattel Endowed Executive Chair and
Distinguished Professor
Department of Pediatrics
David Geffen School of Medicine
Assistant Vice Chancellor for Children’s
Health
University of California, Los Angeles Health
System
Physician in Chief
Mattel Children’s Hospital
Los Angeles, California
Professor of Pediatrics and Dermatology
Johns Hopkins University School of Medicine
Director
Pediatric Dermatology
Johns Hopkins Children’s Center
Baltimore, Maryland
Professor of Pediatrics
Indiana University School of Medicine
Riley Hospital for Children
Indianapolis, Indiana
Sherin U. Devaskar, MD
Robert M. DiBlasi, RRT-NPS, FAARC
Respiratory Research Coordinator
Center for Developmental Therapeutics
Seattle Children’s Research Institute
Seattle, Washington
Reed A. Dimmitt, MD, MSPH
Associate Professor of Pediatrics and Surgery
Director
Division of Neonatology and Pediatric
Gastroenterology and Nutrition
University of Alabama at Birmingham
Birmingham, Alabama
Eric C. Eichenwald, MD
Associate Professor
Vice Chair and Division Director
Neonatology
Department of Pediatrics
University of Texas Health Science Center
Texas Children’s Hospital
Houston, Texas
Eli M. Eisenstein, MD
Senior Pediatrician
Department of Pediatrics
Hadassah-Hebrew University Medical Center
Mount Scopus, Jerusalem, Israel
Jacquelyn R. Evans, MD
Medical Director
Newborn/Infant Intensive Care Unit
The Children’s Hospital of Philadelphia
Associate Division Chief
Department of Neonatology
University of Pennsylvania School of
Medicine
Philadelphia, Pennsylvania
Kelly Evans, MD
Fellow
Division of Craniofacial Medicine
Department of Pediatrics
University of Washington
Craniofacial Center
Seattle Children’s Hospital
Seattle, Washington
Diana L. Farmer, MD, FAAP, FACS, FRCS
Professor of Surgery, Pediatrics, and
Obstetrics, Gynecology, and
Reproductive Sciences
Chief
Division of Pediatric Surgery
Vice Chair
Department of Surgery
University of California, San Francisco School
of Medicine
Surgeon-in-Chief
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
CONTRIBUTORS
Patricia Ferrieri, MD
Christine A. Gleason, MD
Donna M. Ferriero, MS, MD
Michael J. Goldberg, MD
Professor
Chairman’s Fund Endowed Chair in Lab
Medicine and Pathology
Division of Infectious Diseases
Department of Pediatrics
University of Minnesota Medical School
Minneapolis, Minnesota
Professor and Interim Chair of Pediatrics
Professor of Neurology
Co-Director
Newborn Brain Research Institute
University of California, San Francisco School
of Medicine
Physician-in-Chief
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Neil N. Finer, MD
Division of Neonatal-Perinatal Medicine
Department of Pediatrics
University of California, San Diego
San Diego, California
Maria Victoria Fraga, MD
Fellow
Division of Neonatology and Pediatrics
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Lydia Furman, MD
Associate Professor of Pediatrics
Case Western Reserve University School
of Medicine
Rainbow Babies and Children’s Hospital
Cleveland, Ohio
Susan Furth, MD, PhD
Associate Professor
Department of Pediatrics
Johns Hopkins University School of Medicine
Associate Professor
Department of Epidemiology
Johns Hopkins Bloomberg School of Public
Health
Baltimore, Maryland
Estelle B. Gauda, MD
Professor
Department of Pediatrics
Division of Neonatology
Johns Hopkins University School of Medicine
Baltimore, Maryland
Bertil Glader, MD, PhD
Professor of Pediatrics (Hematology/
Oncology) and Pathology
Stanford University School of Medicine
Stanford, California
W. Alan Hodson Endowed Chair in Pediatrics
Professor of Pediatrics
Head, Division of Neonatology
Department of Pediatrics
University of Washington
Seattle Children’s Hospital
Seattle, Washington
Clinical Professor
Department of Orthopedics and Sports
Medicine
University of Washington
Director
Skeletal Health Program
Department of Orthopedics
Seattle Children’s Hospital
Seattle, Washington
Fernando Gonzalez, MD
Assistant Professor
Department of Pediatrics
Division of Neonatology
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Sameer Gopalani, MD
Clinical Assistant Professor
Department of Obstetrics and Gynecology
University of Washington School of Medicine
Division of Perinatal Medicine
Swedish Medical Center
Seattle, Washington
P. Ellen Grant, MD
Associate Professor
Department of Radiology
Harvard Medical School
Founding Director
Center for Fetal-Neonatal Neuroimaging
and Developmental Science Center
Chair
Department of Neonatology
Children’s Hospital Boston
Boston, Massachusetts
Carol L. Greene, MD
Professor
Departments of Pediatrics and Obstetrics,
Gynecology, and Reproductive Sciences
Division of Genetics
University of Maryland School of Medicine
Baltimore, Maryland
Susan Guttentag, MD
Associate Professor
Department of Pediatrics
University of Pennsylvania School
of Medicine
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Chad R. Haldeman-Englert, MD
Assistant Professor
Department of Pediatrics
Section on Medical Genetics
Wake Forest University School of Medicine
Winston-Salem, North Carolina
Thomas Hansen, MD
Professor
Department of Pediatrics
University of Washington School of Medicine
Chief Executive Officer
Seattle Children’s Hospital
Seattle, Washington
Anne V. Hing, MD
Associate Professor
Division of Craniofacial Medicine
Department of Pediatrics
University of Washington
Craniofacial Center
Seattle Children’s Hospital
Seattle, Washington
A. Roger Hohimer, PhD
Associate Professor
Department of Obstetrics
Division of Perinatology
Oregon Health and Science University
Portland, Oregon
Margaret K. Hostetter, MD
Professor and Chair
Department of Pediatrics
Yale School of Medicine
New Haven, Connecticut
Andrew D. Hull, MD, FRCOG, FACOG
Professor of Clinical Reproductive Medicine
University of California, San Diego
Director
Maternal Fetal Medicine Fellowship
University of California, San Diego Medical
Center
La Jolla, California
J. Craig Jackson, MD, MHA
Surgeon
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Professor
Department of Pediatrics
Division of Neonatology
University of Washington
Neonatal Intensive Care Unit Medical
Director
Seattle Children’s Hospital
Seattle, Washington
Jean-Pierre Guignard, MD
Lucky Jain, MD, MBA
Salvador Guevara-Gallardo, MD
Honorary Professor of Pediatrics
Lausanne University Medical School
Lausanne, Switzerland
vii
Executive Vice Chairman
Department of Pediatrics
Emory University
Medical Director
Emory Children’s Center
Atlanta, Georgia
viii
CONTRIBUTORS
Vandana Jain, MD
Nanda Kerkar, MD
Halima Saadia Janjua, MD
John P. Kinsella, MD
Associate Professor
Division of Pediatric Endocrinology
Department of Pediatrics
All India Institute of Medical Sciences
New Delhi, India
Pediatric Nephrology Fellow
Department of Pediatrics
Ohio State University
Nationwide Children’s Hospital
Columbus, Ohio
Sandra E. Juul, MD, PhD
Professor
Department of Pediatrics
University of Washington
Seattle, Washington
Satyan Kalkunte, MPharm, PhD
Research Associate
Superfund Basic Research Program
Department of Pediatrics
Women and Infants’ Hospital of Rhode Island
Warren Alpert Medical School of Brown
University
Providence, Rhode Island
Bernard S. Kaplan, MB, BCh
Professor of Pediatrics
University of Pennsylvania School
of Medicine
Attending in Nephrology
Department of Pediatrics
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Roberta L. Keller, MD
Assistant Professor of Clinical Pediatrics
University of California, San Francisco
Director
Neonatal Extracorporeal Membrane
Oxygenation Program
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Thomas F. Kelly, MD
Clinical Professor and Chief
Division of Perinatal Medicine
Department of Reproductive Medicine
University of California, San Diego School
of Medicine
La Jolla, California
Director
Maternity Services
University of California, San Diego Medical
Center
San Diego, California
Steven E. Kern, PhD
Associate Professor of Pharmaceutics,
Anesthesiology, and Bioengineering
University of Utah
Salt Lake City, Utah
Department of Pediatrics
Division of Pediatric Hepatology
Recanati-Miller Transplantation Institute
The Mount Sinai Medical Center
New York, New York
Professor of Pediatrics
Section of Neonatology
University of Colorado School of Medicine
Medical Director
Newborn/Young Child Transport Service
Co-Director
Newborn Extracorporeal Membrane
Oxygenation Service
The Children’s Hospital
Aurora, Colorado
Roxanne Kirsch, MD, FRCPC, FAAP
Cardiac Intensivist
Departments of Anesthesia and Critical Care
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Monica E. Kleinman, MD
Associate Professor of Anesthesia
Department of Pediatrics Harvard Medical
School
Clinical Director
Medical-Surgical Intensive Care Unit
Department of Anesthesia
Division of Critical Care Medicine
Department of Anesthesia, Perioperative,
and Pain Medicine
Medical Director
Critical Care Transport Program
Children’s Hospital Boston
Boston, Massachusetts
Thomas S. Klitzner, MD, PhD
Jack H. Skirball Professor and Chief
Pediatric Cardiology
David Geffen School of Medicine
University of California, Los Angeles
Los Angeles, California
Sarah M. Lambert, MD
Assistant Professor of Surgery in Urology
University of Pennsylvania School of
Medicine
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
John D. Lantos, MD
Professor of Pediatrics
University of Missouri at Kansas City
Director
Children’s Mercy Bioethics Center
Children’s Mercy Hospital
Kansas City, Missouri
Visiting Professor of Pediatrics
University of Chicago
Chicago, Illinois
Tina A. Leone, MD
Assistant Professor of Pediatrics
University of California, San Diego
San Diego, California
Mary Leppert, MB, BCh
Assistant Professor of Pediatrics
Johns Hopkins University School of Medicine
Attending Physician
Neurodevelopmental Medicine
Kennedy Krieger Institute
Baltimore, Maryland
Harvey L. Levy, MD
Professor
Department of Pediatrics
Harvard Medical School
Senior Physician in Medicine and Genetics
Department of Medicine
Children’s Hospital Boston
Boston, Massachusetts
Mark Lewin, MD
Professor and Chief
Pediatric Cardiology
University of Washington School of Medicine
Co-Director
Heart Center
Seattle Children’s Hospital
Seattle, Washington
Karen Lin-Su, MD
Clinical Associate Professor of Pediatrics
Pediatric Endocrinology
Weill Cornell Medical College
New York, New York
Mignon L. Loh, MD
Professor of Clinical Pediatrics
Department of Pediatrics
University of California, San Francisco
Pediatric Hematological Oncologist
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Scott A. Lorch, MD, MSCE
Assistant Professor
Department of Pediatrics
University of Pennsylvania School
of Medicine
Attending Neonatologist
Division of Neonatology and Center
for Outcomes Research
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Ralph A. Lugo, PharmD
Professor and Chair
Department of Pharmacy Practice
Bill Gatton College of Pharmacy
East Tennessee State University
Johnson City, Tennessee
Volker Mai, PhD
University of Florida Microbiology and Cell
Sciences
Emerging Pathogens Institute
Gainesville, Florida
CONTRIBUTORS
Bradley S. Marino, MD, MPP, MSCE
Associate Professor of Pediatrics
University of Cincinnati College of Medicine
Director
Heart Institute Research Core
Attending Physician
Cardiac Intensive Care Unit
Divisions of Cardiology and Critical Care
Medicine
Cincinnati Children’s Hospital Medical
Center
Cincinnati, Ohio
Barry Markovitz, MD, MPH
Dennis E. Mayock, MD
Jeffrey C. Murray, MD
William L. Meadow, MD, PhD
Josef Neu, MD
Ram K. Menon, MD
Maria I. New, MD
Professor
Division of Neonatology
Department of Pediatrics
University of Washington School of Medicine
Seattle, Washington
Professor
Department of Pediatrics
University of Chicago
Chicago, Illinois
Professor of Clinical Anesthesiology and
Pediatrics
University of Southern California Keck
School of Medicine
Director
Critical Care Medicine
Children’s Hospital Los Angeles
Los Angeles, California
Professor of Pediatrics and Molecular
and Integrative Physiology
Director
Division of Endocrinology
Department of Pediatrics
University of Michigan Medical School
Ann Arbor, Michigan
Kerri Marquard, MD
Professor of Pediatric Neurology
Department of Pediatrics
Catholic University
Rome, Italy
Clinical Fellow
Reproductive Endocrinology and Infertility
Department of Obstetrics and Gynecology
Division of Reproductive Endocrinology and
Infertility
Washington University School of Medicine
St. Louis, Missouri
Camilia R. Martin, MD, MS
Assistant Professor of Pediatrics
Harvard Medical School
Associate Director
Neonatal Intensive Care Unit
Director
Cross-Disciplinary Research Partnerships
Division of Translational Research
Beth Israel Deaconess Medical Center
Boston, Massachusetts
Richard J. Martin, MD
Drusinsky-Fanaroff Chair in Neonatology
Professor
Rainbow Babies and Children’s Hospital
Professor of Pediatrics
Case Western Reserve University
Cleveland, Ohio
Katherine K. Matthay, MD
Mildred V. Strouss Professor of Translational
Research
Chief of Pediatric Hematology-Oncology
University of California, San Francisco
Medical Center
Benioff Children’s Hospital
San Francisco, California
Dana C. Matthews, MD
Associate Professor
University of Washington School of Medicine
Director
Clinical Hematology
Pediatric Hematology and Oncology
Seattle Children’s Hospital
Seattle, Washington
Eugenio Mercuri, MD, PhD
Sowmya S. Mohan, MD
Professor
Departments of Pediatrics, Biology, Nursing,
and Epidemiology
University of Iowa
Iowa City, Iowa
Professor of Pediatrics
Division of Neonatology
University of Florida College of Medicine
Gainesville, Florida
Professor of Pediatrics
Director
Adrenal Steroid Disorders Program
Mount Sinai School of Medicine
New York, New York
Annie Nguyen-Vermillion, MD, FAAP
Department of Neonatology
Northwest Permanente, PC
Providence St. Vincent Medical Center
Neonatal Intensive Care Unit
Portland, Oregon
Victoria Niklas, MD
Neonatal-Perinatal Medicine Fellow
Department of Pediatrics
Division of Neonatology
Emory University
Atlanta, Georgia
Associate Professor of Pediatrics
Division of Neonatal Medicine
University of Southern California Keck
School of Medicine
Children’s Hospital Los Angeles
Los Angeles, California
Kelle Moley, MD
Saroj Nimkarn, MD
James P. Crane Professor of Obstetrics and
Gynecology
Division of Reproductive Endocrinology and
Infertility
Vice Chair
Basic Science Research
Washington University School of Medicine
St. Louis, Missouri
Thomas J. Mollen, MD
Associate Medical Director
Infant Breathing Disorder Center
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Jeremy P. Moore, MD
Assistant Professor of Pediatrics
Division of Pediatric Cardiology
Mattel Children’s Hospital
David Geffen School of Medicine
University of California, Los Angeles
Los Angeles, California
ix
Assistant Professor of Pediatrics
Associate Director
Pediatric Endocrinology
Weill Cornell Medical College
New York, New York
James F. Padbury, MD
Oh-Zopfi Professor of Pediatrics and
Perinatal Biology
Vice Chair for Research
Department of Pediatrics
Warren Alpert Medical School of Brown
University
Women and Infants’ Hospital of Rhode Island
Providence, Rhode Island
Marika Pane, MD, PhD
Institute of Neurology
Catholic University
Rome, Italy
Nigel Paneth, MD, MPH
Professor and Chairman
Department of Reproductive Medicine
University of California, San Diego
San Diego, California
University Distinguished Professor
Departments of Epidemiology and Pediatrics
and Human Development
College of Human Medicine
Michigan State University
East Lansing, Michigan
David A. Munson, MD
Thomas A. Parker, MD
Thomas R. Moore, MD
Assistant Professor of Clinical Pediatrics
University of Pennsylvania School of Medicine
Associate Medical Director
Newborn and Infant Intensive Care Unit
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Associate Professor of Pediatrics
Director
Training Program in Neonatal-Perinatal
Medicine
University of Colorado School of Medicine
Aurora, Colorado
x
CONTRIBUTORS
Janna C. Patterson, MD, MPH
Assistant Instructor
Department of Pediatrics
Division of Neonatology
University of Washington
Seattle, Washington
Christian M. Pettker, MD
Assistant Professor
Department of Obstetrics, Gynecology,
and Reproductive Sciences
Yale School of Medicine
New Haven, Connecticut
Lauren L. Plawner, MD
Acting Assistant Professor of Neurology
University of Washington
Pediatric Neurologist
Seattle Children’s Hospital
Seattle, Washington
Dan Poenaru, BSc, MD, MHPE
Adjunct Professor
Department of Surgery
Queen’s University
Kingston, Ontario, Canada
Medical Director
BethanyKids at Kijabe Hospital
Kijabe, Kenya
Brenda B. Poindexter, MD, MS
Associate Professor of Pediatrics
Section of Neonatal-Perinatal Medicine
Indiana University School of Medicine
Indianapolis, Indiana
Michael A. Posencheg, MD
Assistant Professor of Clinical Pediatrics
University of Pennsylvania School of
Medicine
Associate Medical Director
Intensive Care Nursery
Medical Director
Newborn Nursery
Division of Neonatology and Newborn
Services
Hospital of the University of Pennsylvania
Attending Neonatologist
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Sanjay P. Prabhu, MBBS, DCH, MRCPCH,
FRCR
Instructor
Department of Radiology
Harvard Medical School
Director
Advanced Image Analysis Lab
Department of Radiology
Children’s Hospital Boston
Boston, Massachusetts
Katherine B. Püttgen, MD
Assistant Professor
Departments of Dermatology and Pediatrics
Johns Hopkins University School of Medicine
Baltimore, Maryland
Graham E. Quinn, MD, MSCE
Professor of Ophthalmology
Division of Pediatric Ophthalmology
The Children’s Hospital of Philadelphia
University of Pennsylvania School of
Medicine
Philadelphia, Pennsylvania
Tonse N.K. Raju, MD, DCH
Medical Officer
Eunice Kennedy Shriver National Institute of
Child Health and Human Development
National Institutes of Health
Bethesda, Maryland
Gladys A. Ramos, MD
Associate Physician
Department of Reproductive Medicine
Division of Perinatology
University of California, San Diego
San Diego, California
Benjamin E. Reinking, MD
Clinical Assistant Professor
Department of Pediatrics
University of Iowa
Iowa City, Iowa
C. Peter Richardson, PhD
Associate Research Professor
Department of Pediatrics
University of Washington
Associate Research Professor
Department of Pulmonary and Newborn Care
Seattle Children’s Hospital
Principal Investigator
Center for Developmental Therapy
Seattle Children’s Research Institute
Seattle, Washington
David L. Rimoin, MD, PhD
Professor of Pediatrics, Medicine, and
Medical Genetics
David Geffen School of Medicine
University of California, Los Angeles
Director
Medical Genetics Institute
Steven Spielberg Chair
Cedars-Sinai Medical Center
Los Angeles, California
Elizabeth Robbins, MD
Susan R. Rose, MEd, MD
Professor of Pediatrics and Endocrinology
University of Cincinnati
Pediatric Endocrinologist Cincinnati
Children’s Hospital Medical Center
Cincinnati, Ohio
Mark A. Rosen, MD
Professor
Departments of Anesthesia and Perioperative Care and Obstetrics, Gynecology, and
Reproductive Sciences
University of California, San Francisco
Director
Obstetric Anesthesia
University of California, San Francisco
San Francisco, California
Lewis P. Rubin, MPhil, MD
Pamela and Leslie Muma Endowed Chair
in Neonatology
Professor of Pediatrics, Obstetrics and
Gynecology, Pathology and Cell Biology,
and Community and Family Health
University of South Florida
Medical Director
Newborn Service Line
Tampa General Hospital
Tampa, Florida
Inderneel Sahai, MD, FACMG
Assistant Professor
Department of Pediatrics
University of Massachusetts
Chief Medical Officer
New England Newborn Screening Program
Division of Genetics and Metabolism
Massachusetts General Hospital
Department of Pediatrics
Harvard Medical School
Boston, Massachusetts
Sulagna C. Saitta, MD, PhD
Assistant Professor of Pediatrics
Division of Genetics
The Children’s Hospital of Philadelphia
University of Pennsylvania School of
Medicine
Philadelphia, Pennsylvania
Pablo J. Sánchez, MD
Clinical Professor
Department of Pediatrics
University of California, San Francisco
San Francisco, California
Professor of Pediatrics
University of Texas Southwestern Medical
Center
Children’s Medical Center Dallas
Dallas, Texas
Richard L. Robertson, MD
Gary M. Satou, MD
Associate Professor of Radiology
Harvard Medical School
Radiologist-in-Chief
Children’s Hospital Boston
Boston, Massachusetts
Mark D. Rollins, MD, PhD
Associate Professor
Department of Anesthesia and Perioperative
Care
University of California, San Francisco
San Francisco, California
Associate Clinical Professor
David Geffen School of Medicine
University of California, Los Angeles
Director
Pediatric Echocardiography
Co-Director
Fetal Cardiology Program
Mattel Children’s Hospital
Los Angeles, California
CONTRIBUTORS
Richard J. Schanler, MD
Professor
Department of Pediatrics
Hofstra North Shore-LIJ School of Medicine
Hempstead, New York
Associate Chairman
Department of Pediatrics
Chief
Neonatal-Perinatal Medicine
Steven and Alexandra Cohen Children’s
Medical Center of New York
New Hyde Park, New York
Mark S. Scher, MD
Professor of Pediatrics and Neurology
Case Western Reserve University School
of Medicine
Chief of Pediatric Neurology
Rainbow Babies and Children’s Hospital
University Hospitals of Cleveland
Cleveland, Ohio
Mark R. Schleiss, MD
Professor of Pediatrics
Director
Division of Pediatric Infectious Diseases and
Immunology
Associate Chair for Research
Department of Pediatrics
University of Minnesota Medical School
American Legion Endowed Chair in Pediatric
Infectious Diseases
Co-Director
Center for Infectious Diseases and
Microbiology Translational Research
Minneapolis, Minnesota
Thomas D. Scholz, MD
Children’s Miracle Network Professor
of Pediatrics
Director
Division of Pediatric Cardiology
University of Iowa Carver College
of Medicine
Iowa City, Iowa
Andrew L. Schwaderer, MD
Assistant Professor
Department of Pediatrics
Ohio State University
Columbus, Ohio
Istvan Seri, MD, PhD, HonD
Professor and Chief
Division of Neonatal Medicine
Department of Pediatrics
University of Southern California Keck
School of Medicine
Director
Center for Fetal and Neonatal Medicine
Children’s Hospital Los Angeles
Los Angeles, California
Surendra Sharma, MD, PhD
Professor
Department of Pediatrics
Warren Alpert Medical School of Brown
University
Women and Infants’ Hospital of Rhode Island
Providence, Rhode Island
Evan B. Shereck, MD
Endre Sulyok, MD, PhD, DSc
Eric Sibley, MD, PhD
Peter Tarczy-Hornoch, MD, FACMI
Assistant Professor of Pediatrics
Division of Pediatric Hematology and
Oncology
Oregon Health and Science University
Doernbecher Children’s Hospital
Portland, Oregon
Associate Professor of Pediatrics
Stanford University School of Medicine
Stanford, California
Caroline Signore, MD, MPH
Medical Officer
Pregnancy and Perinatology Branch
Eunice Kennedy Shriver National Institute of
Child Health and Human Development
Bethesda, Maryland
Rebecca Simmons, MD
Associate Professor of Pediatrics
Children’s Hospital Philadelphia
University of Pennsylvania School of
Medicine
Philadelphia, Pennsylvania
Jeffrey B. Smith, MD, PhD
Professor
Department of Pediatrics
David Geffen School of Medicine
University of California, Los Angeles
Medical Director
Newborn Nursery
Mattel Children’s Hospital
Los Angeles, California
Lorie B. Smith, MD, MHS
Staff Pediatric Nephrologist
Walter Reed National Military Medical
Center
Bethesda, Maryland
Clara Song, MD, FAAP
Professor of Pediatrics
Faculty
Health Sciences University of Pecs
Institute of Public Health and Health
Promotion
Pecs, Vorosmarty, Hungary
Head and Professor
Division of Biomedical and Health
Informatics
Department of Medical Education and
Biomedical Informatics
Professor
Division of Neonatology
Department of Pediatrics
Adjunct Professor
Computer Science and Engineering
University of Washington
Seattle, Washington
George A. Taylor, MD
John A. Kirkpatrick Professor of Radiology
Department of Pediatrics
Harvard Medical School
Radiologist-in-Chief Emeritus
Children’s Hospital Boston
Boston, Massachusetts
James A. Taylor, MD
Professor
Department of Pediatrics
University of Washington
Seattle, Washington
Janet A. Thomas, MD
Associate Professor
Department of Pediatrics
Section of Clinical Genetics and Metabolism
University of Colorado School of Medicine
The Children’s Hospital
Aurora, Colorado
Assistant Professor of Pediatrics
Division of Neonatal-Perinatal Medicine
The University of Oklahoma Health Sciences
Center
Children’s Hospital at Oklahoma University
Medical Center
Oklahoma City, Oklahoma
George E. Tiller, MD, PhD
Robin H. Steinhorn, MD
Resident Physician
Department of Dermatology
Johns Hopkins Hospital
Baltimore, Maryland
Professor and Division Head
Department of Pediatrics
Children’s Memorial Hospital
Northwestern University Feinberg School
of Medicine
Chicago, Illinois
Frederick J. Suchy, MD
Professor of Pediatrics
Associate Dean for Child Health Research
University of Colorado School of Medicine
Chief Research Officer and Director
The Children’s Hospital Research Institute
The Children’s Hospital
Aurora, Colorado
xi
Regional Chief
Department of Genetics
Southern California Permanente Medical
Group
Los Angeles, California
Mark M. Tran, MD
Michael Stone Trautman, MD
Clinical Professor of Pediatrics
Section of Neonatal-Perinatal Medicine
Indiana University School of Medicine
Riley Hospital for Children
Indianapolis, Indiana
Jeffrey S. Upperman, MD
Associate Professor of Surgery
Department of Pediatric Surgery
Program Director
Pediatric Surgery Fellowship
Children’s Hospital Los Angeles
Los Angeles, California
xii
CONTRIBUTORS
Carmella van de Ven, MA
Department of Pediatrics
Columbia University
Senior Research Staff Associate
Pediatric Blood and Marrow Transplantation
New York-Presbyterian Morgan Stanley
Children’s Hospital
Columbia University Medical Center
New York, New York
Margaret M. Vernon, MD
Assistant Professor
Department of Pediatrics
Division of Cardiology
University of Washington School of Medicine
Children’s Heart Center
Seattle Children’s Hospital
Seattle, Washington
Jon F. Watchko, MD
Professor of Pediatrics, Obstetrics,
Gynecology, and Reproductive Sciences
Division of Newborn Medicine
Department of Pediatrics
University of Pittsburgh School of Medicine
Senior Scientist
Magee-Women’s Research Institute
Pittsburgh, Pennsylvania
Gil Wernovsky, MD
Conrad Taff Professor of Pediatrics and
Nutrition
Harvard Medical School
Mucosal Immunology Laboratory
Boston, Massachusetts
Professor of Pediatrics
Department of Pediatric Cardiology
University of Pennsylvania School of
Medicine
Medical Director
Neurocardiac Care Program
Associate Chief
Department of Pediatric Cardiology
Director
Program Development and Staff Cardiac
Intensivist
The Cardiac Center
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Linda D. Wallen, MD
Klane K. White, MD, MSc
W. Allan Walker, MD
Clinical Professor of Pediatrics
Associate Division Head
Neonatal Clinical Operations
University of Washington
Associate Medical Director
Neonatal Intensive Care Unit
Seattle Children’s Hospital
Seattle, Washington
Sarah A. Waller, MD
Maternal Fetal Medicine Fellow
Department of Obstetrics and Gynecology
University of Washington
Seattle, Washington
Bradley A. Warady, MD
Professor of Pediatrics
University of Missouri at Kansas City
School of Medicine
Senior Associate Chairman
Department of Pediatrics
Chief
Section of Pediatric Nephrology
Director
Dialysis and Transplantation
Pediatric Nephrology
Children’s Mercy Hospitals and Clinics
Kansas City, Missouri
Robert M. Ward, MD
Professor
Department of Pediatrics
Attending Neonatologist
Adjunct Professor
Pharmacology and Toxicology
Director
Pediatric Pharmacology Program
University of Utah
Salt Lake City, Utah
Assistant Professor
Department of Orthopedics and Sports
Medicine
University of Washington
Seattle, Washington
Pediatric Orthopedic Surgeon
Department of Orthopedics and Sports
Medicine
Seattle Children’s Hospital
Seattle, Washington
Calvin B. Williams, MD, PhD
Professor of Pediatrics
Chief
Section of Pediatric Rheumatology
Medical College of Wisconsin
D.B. and Marjorie Reinhart Chair in
Rheumatology
Children’s Hospital of Wisconsin
Milwaukee, Wisconsin
David Woodrum, MD
Professor of Pediatrics
Division of Neonatology
University of Washington School of Medicine
Seattle, Washington
George A. Woodward, MD, MBA
Professor
Department of Pediatrics
University of Washington School of Medicine
Chief
Emergency Medicine
Medical Director
Transport Services
Seattle Children’s Hospital
Seattle, Washington
Dakara Rucker Wright, MD
Pediatric Dermatologist
Johns Hopkins University School of Medicine
Johns Hopkins Children’s Center
Baltimore, Maryland
Jeffrey A. Wright, MD
Associate Professor
Department of Pediatrics
University of Washington
Seattle, Washington
Linda L. Wright, MD
Deputy Director
Center for Research for Mothers
and Children
Director
Global Network for Women’s and Children’s
Health Research
Eunice Kennedy Shriver National Institute of
Child Health and Human Development
National Institutes of Health
Bethesda, Maryland
Christopher M. Young, MD
Fellow
Neonatal-Perinatal Medicine
Department of Pediatrics
Division of Neonatology
University of Florida
Gainesville, Florida
Guy Young, MD
Associate Professor of Pediatrics
University of Southern California Keck
School of Medicine
Director
Hemostasis and Thrombosis Center
Center for Cancer and Blood Disorders
Children’s Hospital Los Angeles
Los Angeles, California
Elaine H. Zackai, MD
Division of Genetics
Department of Pediatrics
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Stephen A. Zderic, MD
Professor of Surgery in Urology
University of Pennsylvania School of
Medicine
John W. Duckett Endowed Chair
The Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Preface
“The neonatal period … represents the last frontier of
medicine, territory which has just begun to be cleared of
its forests and underbrush in preparation for its eagerly
anticipated crops of saved lives.”
Introduction from the first edition of
Schaffer’s Diseases of the Newborn
The first edition of Diseases of the Newborn was published
in 1960 by Dr. Alexander J. Schaffer, a well-known Baltimore pediatrician who coined the term neonatology to
describe this emerging pediatric subspecialty that concentrated on “the art and science of diagnosis and treatment
of disorders of the newborn infant.” Schaffer’s first edition was used mainly for diagnosis, but also included reference to neonatal care practices (i.e., the use of antibiotics,
temperature regulation, and attention to feeding techniques)—practices that had led to a remarkable decrease
in the infant mortality rate in the United States, from 47
deaths per 1000 live births in 1940 to 26 per 1000 in 1960.
But a pivotal year for the new field of neonatology came
3 years later in 1963, with the birth of President John F.
Kennedy’s son, Patrick Bouvier Kennedy, at 36 weeks’
gestation (i.e., late preterm). His death at 3 days of age,
from complications of hyaline membrane disease, accelerated the development of infant ventilators that, coupled
with micro-blood gas analysis and expertise in the use of
umbilical artery catheterization, led to the development
of intensive care for newborns in the 1960s on both sides
of the Atlantic. Advances in neonatal surgery and cardiology, along with further technological innovations, stimulated the development of neonatal intensive care units and
regionalization of care for sick newborn infants over the
next several decades. These developments were accompanied by an explosion of neonatal research activity that led
to improved understanding of the pathophysiology and
genetic basis of diseases of the newborn, which in turn has
led to spectacular advances in neonatal diagnosis and therapeutics—particularly for preterm infants. These efforts
led to continued improvements in the infant mortality rate
in the United States, from 26 deaths per 1000 livebirths
in 1960 to 6.5 per 1000 in 2004. Current research efforts
are focused on decreasing the striking global disparities
in infant mortality rates, decreasing neonatal morbidities,
advancing neonatal therapeutics, and preventing prematurity and newborn diseases. We neonatologists would like
to be put out of business one day!
Dr. Mary Ellen Avery joined Dr. Schaffer for the
third edition of Diseases of the Newborn in 1971. For the
fourth edition in 1977, Drs. Avery and Schaffer recognized that their book now needed multiple contributors
with subspecialty expertise and they became co-editors,
rather than sole co-authors, of the book. In the preface
to that fourth edition, Dr. Schaffer wrote, “We have also
seen the application of some fundamental advances in
molecular biology to the management of our fetal and
newborn patients”—referring to the new knowledge of
hemoglobinopathies. Dr. Schaffer died in 1981, at the age
of 79, and Dr. H. William Taeusch joined Dr. Avery as
co-editor for the fifth edition in 1984. Dr. Roberta Ballard joined Drs. Taeusch and Avery for the sixth edition
in 1991, with the addition of Dr. Christine Gleason for
the eighth edition in 2004. Drs. Avery, Taeusch, and Ballard retired from editing the book in 2009, and became
“editors emeriti.” Dr. Gleason was joined by Dr. Sherin
Devaskar as co-editor for this, the ninth edition.
What’s new and different about this edition? The book
has been completely (and often painfully) revised and
updated by some of the best clinicians and investigators
in their field. Some chapters required more extensive revision than others, particularly those that deal with areas in
which we have benefitted from new knowledge and/or its
application to new diagnostic and therapeutic practices.
This is particularly true in areas such as the genetic basis
of disease, neonatal pain management, information technology, and the fetal origins of adult disease—an area that
is now embedded within many of the chapters of this book.
Some of the book’s sections were reorganized to reflect
our field’s continued evolution. For example, Chapter 52,
Persistent Pulmonary Hypertension, previously in Part X,
Respiratory System, has found a new home in Part XI,
Cardiovascular System. Finally, we’ve added new chapters
that reflect the continued growth and development of our
subspecialty. These include Chapter 4, Global Neonatal
Health; Chapter 29, Stabilization and Transport of the
High-Risk Infant; Chapter 33, Care of the Late Preterm
Infant; Chapter 74, Disorders of the Liver; and Chapter
95, Craniofacial Malformations.
With the incredible breadth and depth of information
immediately available to neonatal caregivers and educators
on multiple internet sites, what’s the value of a textbook?
We, the co-editors of this ninth edition, believe that textbooks such as Diseases of the Newborn and all forms of integrative scholarship, will always be needed—by clinicians
striving to provide state-of-the-art neonatal care, by educators striving to train the next generation of caregivers,
and by investigators striving to advance neonatal scholarship. A textbook’s content is only as good as its contributors
and this textbook, like the previous editions, has awesome
contributors. They were chosen for their expertise and
ability to integrate their knowledge into a comprehensive,
readable, and useful chapter. They did this despite the
demands of their day jobs in the hopes that their syntheses could, as Ethel Dunham wrote in the foreword to the
first edition, “spread more widely what is already known
… and make it possible to apply these facts.” Textbooks
of the future will undoubtedly take advantage of online,
xiii
xiv
PREFACE
interactive publishing technologies, making their content readily accessible and more real-time, with continued
revision and updating. However, in 2011—a full 50 years
after the publication of the first edition of this book—we
continue to find copies of this and other textbooks important to our subspecialty lying dog-eared, coffee-stained,
annotated, and broken-spined in all of the places where
neonatal caregivers congregate. These places, these congregations of neonatal caregivers, are now present in
every country around the world. The tentacles of neonatal
practice and education are spreading—ever deeper, ever
wider—to improve the outcome of pregnancy worldwide.
Textbooks connect us to the past, bring us up to date with
the present, and prepare and excite us for the future. We
will always need them, in one form or another, at our sites
of practice. To that end, we have challenged ourselves to
meet, and hopefully exceed, that need—for our field, for
our colleagues, and for the babies.
We wish to thank key staff at Elsevier—Deidre Simpson, senior developmental editor, and Judith Fletcher,
publishing director, both of whom demonstrated patience,
guidance, and persistence, and Jessica Becher, associate
project manager, for our book. We also wish to thank our
academic institutions and our administrative assistants,
Mildred Hill at the University of Washington and Kristie
Smiley at the University of California, Los Angeles. They
kept us grounded, on track, and basically saved our lives!
We are indebted to our contributors, who actually wrote
the book and did so willingly, enthusiastically, and (for the
most part) in a timely fashion—despite the myriad of other
responsibilities in their lives. And we are deeply grateful
for the support of our families throughout the long, and
often challenging, editorial process. Finally, we thank the
editors emeriti of this book, Drs. Mary Ellen Avery, Bill
Taeusch, and Roberta Ballard, for their enormous contributions to the field of neonatology and to the lives of
babies throughout the world, and their wise influence on
us, the editors of the ninth edition of this text.
Christine A. Gleason
Sherin U. Devaskar
To our parents, Peter and Vera and Sitaram and Santha, who have inspired us
To our husbands, Erik and Uday, who are the wind behind our sails
To our children, Kristen, Lauren, and Erin, and Chirag and Chetan, who are our future
Christine A. Gleason
Sherin U. Devaskar
Editors Emeritis
Mary Ellen Avery, MD
H. William Taeusch, MD
Roberta A. Ballard, MD
P A R T
I
Overview
C H A P T E R
1
Neonatal and Perinatal Epidemiology
Nigel Paneth
EPIDEMIOLOGIC APPROACHES
TO THE PERINATAL AND NEONATAL
PERIODS
The period surrounding the time of birth (the perinatal
period) is a critical episode in human development, rivaling only the period surrounding conception in its significance. During this period, the infant makes the critical
transition from its dependence on maternal and placental
support—oxidative, nutritional, and endocrinologic—and
establishes independent life. The difficulty of this transition is indicated by mortality risks that are higher than
any occuring until old age (Kung et al, 2008) and by risks
for damage to organ systems, most notably the brain, that
can be lifelong. Providers of care in the perinatal period
recognize that the developing human organism cannot
always demonstrate the immediate effects of even profound insults. Years must pass before the effects on higher
cortical functions of insults and injuries occurring during
the perinatal period can be detected reliably. Epidemiologic approaches to the perinatal period must therefore be
bidirectional—looking backward to examine the causes of
adverse health conditions that arise or complicate the perinatal period, and looking forward to see how these conditions shape disorders of health found later in life.
Traditionally the perinatal period was described as from
28 weeks’ gestation until 1 week of life, but the World
Health Organization has more recently antedated the
onset of the perinatal period to 22 weeks’ gestation (World
Health Organization, 2004). For this discussion we will
view the term perinatal more expansively, as including the
second half of gestation—by which time most organogenesis has occurred, but growth and maturation of many systems have yet to occur—and the first month of life. The
neonatal period, usually considered as the first month of
life, is thus included in the term perinatal, reflecting the
view that addressing the problems of the neonate requires
an understanding of intrauterine phenomena.
HEALTH DISORDERS OF PREGNANCY
AND THE PERINATAL PERIOD
KEY POPULATION MORTALITY RATES
Maternal and child health in the population have traditionally been assessed by monitoring two key rates—
maternal mortality and infant mortality. Maternal
mortality is defined by the World Health Organization as the death of a woman from pregnancy-related
causes during pregnancy or within 42 days of pregnancy,
expressed as a ratio to 100,000 live births in the population being studied (World Health Organization, 2004).
Because pregnancy can contribute to deaths beyond 42
days, some have argued for examining all deaths within
1 year of a pregnancy (Hoyert, 2007). When the cause
of death is attributed to pregnancy-related causes, it is
described as direct. When pregnancy has aggravated an
underlying health disorder, the death is termed an indirect
maternal death. Deaths unrelated to pregnancy that occur
within 42 days of pregnancy are termed incidental maternal deaths or sometimes pregnancy-related deaths (Khlat and
Guillaume, 2006). These distinctions are not always easy
to make. Homicide and suicide, for example, are sometimes found to be more common in pregnancy, and thus
might not be entirely incidental (Samandari et al, 2010;
Shadigian and Bauer, 2005).
Since 2003, the U.S. Standard Certificate of Death has
included a special requirement for identifying whether the
decedent, if female, was pregnant or had been pregnant
in the previous 42 days, or from 43 days to 1 year before
the death, thus enhancing the monitoring of all forms of
maternal death (Centers for Disease Control and Prevention, 2003). This addition has added to the number of
recorded maternal deaths.
In most geographic entities, infant mortality (IM) is
defined as all deaths occurring from birth to 365 days of
age in a calendar year divided by all live births in the same
year. This approach is imprecise, because some deaths in
the examined year occurred to the previous year’s birth
cohort, and some births in the examined year may die as
infants in the following year. In recent years, birth-death
linkage has permitted vital registration areas in the United
States to provide IM rates that avoid this imprecision.
The standard IM rate reported by the National Center
for Health Statistics links deaths for the index year to all
births, including those taking place the previous year. This
form of IM is termed period infant mortality. An alternative
procedure is to take births for the index year and link them
to infant deaths, including those taking place the following year; this is referred to as birth cohort infant mortality,
and it is not used for regular annual comparisons because
it cannot be completed in as timely a fashion as period IM
(Mathews and MacDorman, 2008).
Infant deaths are often divided into deaths in the first
28 days of life (neonatal death) and deaths later in the first
year (postneonatal death). Neonatal deaths, which are
largely related to preterm birth and birth defects, tend
to reflect the circumstances of pregnancy. Postneonatal
1
PART I Overview
deaths, when frequent, commonly result from infection,
often in the setting of poor nutrition. Thus in underdeveloped countries, postneonatal deaths predominate; in
industrialized countries the reverse is true. In the United
States, neonatal deaths have been more frequent than
postneonatal deaths since 1921. In recent years, the ratio
of neonatal to postneonatal deaths in the United States has
consistently been approximately 2:1.
Perinatal mortality is a term used for a rate that combines
stillbirths and neonatal deaths in some fashion (World
Health Organization, 2004). Stillbirth reporting before
28 weeks’ gestation is probably incomplete, even in the
United States, where such stillbirths are required to be
reported in every state. Nonetheless, stillbirths continue
to be reported at a levels not much lower those of neonatal
deaths, and our understanding of the causes of stillbirth
remains very limited.
TIME TRENDS IN MORTALITY RATES
IN THE UNITED STATES
Maternal mortality and IM declined steadily through the
twentieth century. By 2000, neonatal mortality was 10%
of its value in 1915, and postneonatal mortality less than
7%. Maternal mortality in this interval declined 74-fold;
the rate in 2000 was less than 2% of the rate recorded
in 1915. The contribution to these changes of a variety
of complex social factors including improvements in
income, housing, birth spacing, and nutrition have been
documented widely, as has the role of ecologic-level public health interventions that have produced cleaner food
and water (Division of Reproductive Health, 1999). Public health action at the individual level, including targeted
maternal and infant nutrition programs and immunization programs, have made a lesser but still notable contribution. Medical care was, until recently, less critically
involved, except for the decline in maternal mortality,
which was highly sensitive to the developments in blood
banking and antibiotics that began in the 1930s. To this
day, hemorrhage and infection account for a large fraction of the world’s maternal deaths (Khan et al, 2006).
A notable feature of the past 50 years is the sharp decline
in all three mortality rates, beginning in the 1960s after a
period of stagnation in the 1950s (Figure 1-1). The decline
began with maternal mortality, followed by postneonatal
and then by neonatal mortality. The contribution of medical care of the neonate was most clearly seen in national
statistics in the 1970s, a decade that witnessed a larger
decline in neonatal mortality than in any previous decade
of the century (Division of Reproductive Health, 1999).
All of the change in neonatal mortality between 1950 and
1975 was in mortality for a given birthweight; no improvement was seen in the birthweight distribution (Lee et al,
1980). The effect of newborn intensive care on mortality
in extremely small babies has been striking. In 1960, 142
white singletons weighing less than 1000 g in the United
States survived to age 1, less than 1% of births of that
weight. In 2005, the survival rate for infants weighing 501
to 999 g was 70%, and the number of survivors at age 1 was
almost 18,000 (Mathews and MacDorman, 2008).
In retrospect, three factors seem to have played critical roles in the rapid development of newborn intensive
MORTALITY RATES IN THE U.S. 1955–2005
50
45
40
35
30
Neonatal mortality rate
per 1000
Postneonatal mortality
rate per 1000
Maternal mortality rate
per 100,000
25
20
15
10
5
0
1955
1957
1959
1961
1963
1965
1967
1969
1971
1973
1975
1977
1979
1981
1983
1985
1987
1989
1991
1993
1995
1997
1999
2001
2003
2005
2
FIGURE 1-1 Neonatal, postneonatal, and maternal mortality, 1955 to
2005.
care programs. These programs that largely accounted for
the rapid decline in birthweight-specific neonatal mortality that characterized national trends in the last third of
the twentieth century. The first factor was the willingness
of medicine to provide more than nursing care to marginal populations such as premature infants. It has often
been noted that the death of the mildly premature son of
President John F. Kennedy in 1963 provided a stimulus to
the development of newborn intensive care, but it should
be noted that the decline in IM that began in the 1970s
was paralleled by a similar decline in mortality for the
extremely old (Rosenwaike et al, 1980). A second factor
was the availability of government funds, provided by the
Medicaid program adopted in 1965, to pay for the care of
premature newborns, among whom the poor are overrepresented. Whereas these two factors were necessary, they
would have been insufficient to improve neonatal mortality had not new medical technologies, especially those supporting ventilation of the immature newborn lung, been
developed at approximately the same time (Gregory et al,
1971).
Advances in newborn care have ameliorated the effects
of premature birth and birth defects on mortality. Unfortunately, the underlying disorders that drive perinatal
mortality and the long-term developmental disorders
that are sometimes their sequelae have shown little tendency to abate. With the important exception of neural
tube defects, whose prevalence has declined with folate
fortification of flour in the United States and programs
to encourage intake of folate in women of child-bearing
age (Mathews et al, 2002), the major causes of death—
preterm birth and birth defects—have not declined, nor
has the major neurodevelopmental disorder that can be
of perinatal origin, cerebral palsy (Paneth et al, 2006).
Progress has come from improved medical care of the
high-risk pregnancy and the sick infant, rather than
through understanding and preventing the disorders
themselves.
The period since 1990 has witnessed much less impressive mortality improvement. While infant, neonatal, and
postneonatal mortality have declined since 1990, in the
1995-2005 decade these rates were at a near standstill
3
CHAPTER 1 Neonatal and Perinatal Epidemiology
TABLE 1-1 U.S. Perinatal Mortality, Morbidity, Interventions, Health Conditions, and Behaviors, 1990 to 2005
1990
1995
2000
8.2
9.2
5.8
3.4
7.5
7.1
7.6
4.9
2.6
6.9
9.8
6.9
4.6
2.3
6.6
10.6%
11.0%
1.9%
2005
Net Change
1990-2005 (%)
Death Rates and Ratios
Maternal mortality ratio (per 100,000 live births)
Infant mortality rate (per 1000 live births)
Neonatal mortality rate (per 1000 live births)
Postneonatal mortality rate (per 1000 live births)
Fetal mortality ratio (per 1000 live births)*
15.1
6.9
4.5
2.3
6.2
+84%
–25%
–22%
–32%
–17%
11.6%
12.7%
+20%
1.9%
1.9%
2.0%
+5%
7.1
7.0
7.2
7.6
+7%
7.0%
1.3%.
6.3
7.3%
1.4%
6.6
7.6%
1.4%
7.1
8.2%
1.5%
7.3
+12%
+15%
+16%
22.6%
20.8%
22.9%
30.3%
+34%
17.7%
3.2%
6.1%
7.9%
13.7%
1.5%
4.2%
9.3%
12.2%
0.9%
3.9%
11.6%
10.7%
0.7%
3.5%
11.4%
–40%
–78%
–42%
+44%
28.0%
32.1%
33.2%
36.9%
+32%
23.3
26.1
31.1
33.8
+45%
70.9
64.6
65.3
66.7
–5.9%
Morbidity Rates
Preterm birth (<37 weeks’ gestation)
per 100 live births
Very preterm birth (<32 weeks’ gestation)
per 100 live births
Extremely preterm birth (<28 weeks’ gestation)
per 1000 live births
Low birthweight (<2500 g) per 100 live births
Very low birthweight (<1500 g) per 100 live births
Extremely low birthweight (<1000 g) per 1000 live
births
Cesarean Section
Per 100 live births
Health Behaviors (per 100 live births)
Cigarette smoking†
Alcohol intake‡
Late or no prenatal care
Inadequate weight gain (<16 lb) at 40 weeks’
gestation§
Unmarried
Multiple Births
Per 1000 live births
Fertility Rate
Per 1000 women aged 15-44 years
*Fetal deaths with stated or presumed period of 20 weeks’ gestation or more.
†1990-2000 smoking data based on 45 states and Washington, D.C.; 2005 data based on 36 states, New York City, and Washington, D.C., which used pre-2003 smoking definitions.
‡1990-2000 alcohol data based on 46 states and Washington, D.C.; 2005 data based on 36 states, New York City, and Washington, D.C., which used pre-2003 drinking definitions.
§1990 weight gain data based on 48 states and Washington, D.C. Remaining data based on 49 states and Washington, D.C.
(Table 1-1). Infant mortality actually rose in 2002, the
first 1 year since 1958. Data from the Vermont Oxford
Neonatal Network encompassing hundreds of neonatal
units show stability in weight-specific mortality rates for
all categories of infants weighing less than 1500 g, beginning in approximately 1995 (Horbar et al, 2002). It appears
that the pace of advancement in newborn medicine and the
expansion of newborn intensive care to previously underserved populations, factors that have exerted a constant
downward pressure on infant mortality since the 1960s,
have lessened greatly in the past decade.
Maternal mortality has actually climbed substantially,
but this is almost certainly the effect of improved reporting (Hoyert, 2007). Two key changes were the implementation of the International Classification of Diseases, Tenth
Revision, in 1999, which was less restrictive in including
indirect maternal deaths, and the introduction of the separate pregnancy question on the U.S. Standard Certificate
of Death in 2003.
The risk of preterm birth has been increasing in recent
years. This increase is mostly noted in moderately preterm
babies, and is likely to reflect increased willingness on the
part of obstetricians to deliver fetuses earlier in gestation who are not doing well in utero, in addition to the
increased prevalence of twins and triplets, who are generally born preterm, resulting from in vitro fertilization. The
small rise in extremely small and preterm babies is likely
to reflect trends toward increased reporting of marginally
viable immature babies as live births rather than stillbirths
(MacDorman et al, 2005).
The cesarean section rate continues its long-term
increase, from 5% in 1970 to 23% in 1990 to 32% in 2005.
The reasons for this unprecedented trend are multifactorial and include pressures from patients, physicians, and
the medical malpractice system. The steady reduction in
smoking during pregnancy is likely to be real, whereas
trends in the self-reporting of alcohol use in pregnancy may
be influenced by societal norms and expectations. Fewer
women seem to have late or no prenatal care in recent
years, but more women have been found to have inadequate pregnancy weight gain at term. A slight increase in
the fertility rate follows a long-term (since approximately
4
PART I Overview
1960) decline in fertility in the United States. One third
of mothers in the United States are now unmarried when
they give birth.
International Comparisons
The United States lags in IM compared with other
developed nations; the United States ranked thirtieth
in the world in IM in 2005 (MacDorman and Mathews,
2009). This surprising finding, in light of more favorable socioeconomic and medical care circumstances in
the United States than in many nations with lower IM,
cannot be attributed to inferior neonatal care. Mortality
rates for low-birthweight infants are generally lower in
the United States than in European and Asian nations,
although mortality at term may be higher. The key difference, however, is that the United States suffers from
a striking excess of premature births. Whereas the U.S.
African American population is especially vulnerable
to premature birth, and especially severe prematurity,
premature birth rates are also considerably higher in
white Americans than in most European populations.
It is likely that the recording of marginally viable small
infants as live births rather than stillbirths is more pronounced in the United States than in Europe (Kramer
et al, 2002). Although this practice makes a contribution
to our higher prematurity and IM rates, it cannot fully
explain them.
Premature birth, fetal growth retardation, and IM are
tightly linked, in every setting in which they have been
studied, to most measures of social class and especially to
maternal education; however, uncovering precisely why
lower social class drives these important biologic differences has been elusive. Factors such as smoking have at
times been implicated, but can only explain a small fraction
of the social class effect. It is unlikely that this situation
will change until a better understanding of the complex
social, environmental, and biologic roots of preterm birth
is achieved.
Health Disparities in the Perinatal Period
In 2005, 55.1% of all U.S. births were to non-Hispanic
white mothers, 23.8% were to Hispanic mothers, 14.1%
were to African American mothers, and 7% were to
mothers of other ethnic groups. Health disparities are
especially prominent in the perinatal period, with African
American IM stubbornly remaining about double that of
white IM in the United States, even as rates decline in
both populations (Table 1-2). Preterm birth is the central contributor to this racial disparity in IM, and the
more severe the degree of prematurity, the higher the
excess risk for African American infants. The risk of birth
before 37 weeks’ gestation was 76% higher in African
American mothers than among non-Hispanic whites in
2005, but the risk of birth before 32 weeks’ gestation was
310% higher. Reduction in IM disparities in the United
States thus requires a better understanding of the causes
and mechanisms of preterm birth. Birth defect mortality
shows a less pronounced gradient by ethnic group, and it
does not contribute in a major way to overall IM disparities (Yang et al, 2006).
The Hispanic paradox is a term often used to describe
the observation that IM is the same or lower in U.S. citizens classified as Hispanic than in non-Hispanic whites,
despite the generally lower income and education levels
of U.S. Hispanics (Hessol and Fuentes-Afflick, 2005).
The IM experience of Hispanic mothers in the United
States reflects the principle that premature birth and low
birthweight are key determinants of IM, because these
parameters are also favorable in Hispanics. Smoking
is much less common among Hispanics in the United
States, but this factor alone does not fully explain the
paradox.
MAJOR CAUSES OF DEATH
Analyzing the cause of death, a staple of epidemiologic
investigation, has limitations when applied to the perinatal period. Birth defect mortality is probably reasonably
accurate, but causes of deaths among premature infants
are divided among categories such as respiratory distress
syndrome, immaturity, and a variety of complications of
prematurity. Choosing which particular epiphenomenon
of preterm birth to label as the primary cause of death is
arbitrary to some extent. Some maternal complications,
such as preeclampsia, are also occasionally listed as causes
of newborn death. However categorized, prematurity
accounts for at least one third of infant deaths (Callaghan
et al, 2006).
Before prenatal ultrasound examination could be used to
estimate gestational age with reasonable accuracy, a high
fraction of neonatal deaths were attributed to low birthweight, but most of these deaths occurred in premature
infants, because premature birth is much more important
as a cause of death than is fetal growth restriction. Extreme
prematurity makes a contribution to IM well beyond its
frequency in the population; 1.9% of births, that occurred
before 32 weeks’ gestation, accounted for 54% of all infant
deaths in 2005.
Following premature birth, another important group
of causes of death is congenital anomalies (Mathews
and MacDorman, 2008). With the exception of folate
supplementation to prevent neural tube defects, there is
no clearly effective primary prevention program for any
birth defect. Pregnancy screening and termination of
severe defects, however, is an option for many mothers,
and there is evidence that this practice contributes to a
reduced prevalence of chromosomal anomalies at birth
(Khoshnood et al, 2004).
The major postneonatal cause of death since the 1970s
in the United States is sudden infant death syndrome. This
cause of death has declined substantially in the United
States in parallel with successful public health efforts to
discourage sleeping in the prone position during infancy
(Ponsonby et al, 2002).
MAJOR MORBIDITIES RELATED
TO THE PERINATAL PERIOD
The principal complications of preterm birth involve
five organs: the lung, heart, gut, eye, and brain. Management of respiratory distress syndrome and its short- and
long-term complications is the centerpiece of neonatal
5
CHAPTER 1 Neonatal and Perinatal Epidemiology
TABLE 1-2 Ethnic Disparities in Key Perinatal Outcomes and Exposures in 2005
Non-Hispanic White
Prevalence rate
African American
Prevalence rate
Hispanic
RR*
Prevalence rate
RR*
Deaths Rates and Ratios
Maternal mortality ratio
(per 100,000 live births)
Infant mortality rate
(per 1000 live births)
Neonatal mortality rate
(per 1000 live births)
Postneonatal mortality rate
(per 1000 live births)
Fetal mortality ratio
(per 1000 live births)†
11.7
39.2
3.4
9.6
0.8
5.7
13.7
2.4
5.8
1.0
3.8
9.1
2.4
3.9
1.0
1.9
4.6
2.4
1.8
1.0
4.8
11.1
2.3
5.4
1.1
11.6%
1.6%
5.6
7.3%
1.2%
5.5
4.5%
3.6%
18.3%
4.1%
18.9
14.0%
3.3%
1.8%
4.6%
3.5%
1.6
2.6
3.4
1.4
2.8
3.3
1.0
1.0
11.6%
1.8%
6.2
6.9%
1.2%
5.9
2.8%
3.8%
1.0
30.4%
26.6%
33.6%
19.7%
1.1
0.7
29.0%
15.5%
1.0
0.6
13.9%
8.5%
0.7
2.9%
0.2
2.2%
9.4%
5.6%
16.3%
2.5
1.7
5.1%
14.2%
2.3
1.5
25.3%
58.3
69.9%
67.2
2.8
1.2
48.0%
99.4
1.9
1.7
38.2
37.5
1.0
22.8
0.6
Morbidity (percent of live births)
Preterm birth
Very preterm birth
Extremely preterm birth
Low birthweight
Very low birthweight
Extremely low birthweight
Pregnancy-associated hypertension
Diabetes in pregnancy
1.2
0.9
1.0
1.1
0.6
1.1
Interventions
(percent of live births)
Cesarean section
Induction of labor
Health Behaviors
Smoking‡
Alcohol intake
Late or no prenatal care§
Inadequate weight gain (<16 lb)
at 40 weeks’ gestation¶
Unmarried
Fertility rate
(women 15-44 years old)
Multiple births
*Relative risk compared with non-Hispanic white (rounded to nearest decimal point).
†Fetal deaths with stated or presumed period of 20 weeks’ gestation or more
‡Based on 36 states, New York City, and Washington, D.C., which used pre-2003 smoking definitions
§Based on 37 states, Washington, D.C. and New York City
¶Based on 49 states, New York City, and Washington, D.C.
medicine. Surgical or medical management of symptomatic patent ductus arteriosus is the major cardiac challenge
in premature infants, and there is limited understanding of
the striking variations, by time and place, of necrotizing
enterocolitis—a disorder that in its most extreme forms
can cause death or substantial loss of bowel function. Retinopathy of prematurity is closely related to arterial oxygen
levels. The epidemic level of this disorder encountered in
the 1950s, when oxygen was freely administered without
monitoring, was a major setback for neonatal medicine
(Silverman, 1980). However, even with more careful management of oxygen, retinopathy of prematurity continues
to occur.
The largest unsolved problem in neonatal medicine
remains the high frequency of brain damage in survivors
of premature birth. The extraordinary decline in mortality rates has not been paralleled by similar declines in rates
of neurodevelopmental disabilities in survivors. The key
epidemiologic feature of cerebral palsy rates in population
registries toward the end of the twentieth century was a
modest overall increase in the prevalence of that disorder,
which was attributable entirely to the increasing number
of survivors of very low birthweight. There are suggestions that this rise may now be leveling (Paneth et al,
2006).
FACTORS AFFECTING PERINATAL
HEALTH
HEALTH STATES IN PREGNANCY
The major causes of neonatal morbidity, prematurity
and birth defects, generally occur in pregnancies free of
antecedent complications. Having a previous birth with
an anomaly or a previous preterm birth raises the risk for
recurrence of the condition. For preterm birth, no other
known risk factor carries as much risk for the mother as
having previously delivered preterm.
6
PART I Overview
More than a quarter of preterm birth is iatrogenic, the
result of induced labor in pregnancies in which the fetus
is severely compromised (Morken et al, 2008). Generally
the reason is preeclampsia with attendant impairments in
uterine blood flow and poor fetal growth, but poor uterine blood flow and impaired fetal growth can also occur
independently of diagnosed preeclampsia; the other major
complication of pregnancy is diabetes, most often gestational but sometimes preexisting. Insulin resistance in the
mother promotes the movement of nutrients towards the
fetus, and typically the infant of the diabetic mother is
large for gestational age. Severe diabetes, however, can be
accompanied by fetal growth retardation.
HEALTH BEHAVIORS
The most carefully studied and well-established health
behavior affecting newborns is maternal cigarette smoking, which has a consistent effect in impairing fetal growth
(Cnattingius, 2004). Infants with growth retardation from
maternal cigarette smoking paradoxically survive slightly
better than do infants of the same weight whose mothers did not smoke, but the net effect of smoking, which
also shortens gestation slightly, is to increase perinatal
mortality. Although the subject is much debated, it has not
been conclusively shown that prenatal maternal smoking
has independent long-term effects on children’s cognitive
capacity (Breslau et al, 2005).
Alcohol is less clearly a growth retardant, but mothers
who drink heavily during pregnancy are at risk of having
infants with the cluster of defects known as fetal alcohol syndrome. Cocaine use in pregnancy is almost surely a severe
growth retardant, and it may affect neonatal behavior, but
the long-term effects of this exposure on infant cognition
and behavior are not as grave as initially feared (Bandstra
et al, 2010).
PERINATAL MEDICAL CARE
In light of the potent effects of medical care on the neonate, it has been important to develop systems of care that
ensure, or at least facilitate, provision of care to neonates
in need. This concept was first promoted by the March of
Dimes Foundation, which in its committee report of 1976
recommended that all hospitals caring for babies be classified as level 1 (care for healthy and mildly sick newborns),
level 2 (care for most sick infants born in the hospital, but
not accepting transfers), or level 3 (regional centers caring for complex surgical disease and receiving transfers)
(The Committee on Perinatal Health, 1976). This concept of a regional approach to neonatal care, with different hospitals playing distinct roles in providing care, was
endorsed by organizations such as American College of
Obstetricians and Gynecologists and the American Academy of Pediatrics and by many state health departments.
Whereas it is important to transfer sick babies to level 3
centers when needed, it is preferable to transfer mothers
at risk of delivering prematurely or of having a sick neonate, because transport of the fetus in utero is superior to
any form of postnatal transport. Birth at a level 3 center
has been shown consistently to produce lower mortality
rates than birth in other levels of care (Paneth, 1992). The
overall system of care, which includes selecting mothers and babies for transfer to other hospitals, is highly
dependent on cooperative physicians and health systems.
Concern has been raised that economic considerations
may threaten ideal systems of regionalization of newborn
intensive care (Bode et al, 2001).
EPIDEMIOLOGIC STUDY DESIGNS
IN THE PERINATAL PERIOD
Epidemiologic studies have contributed substantially to
better understanding the patterns of risk and prognosis
in the perinatal period, to tracking patterns of mortality
and morbidity, to assessing regional medical care, and to
assisting physicians and other providers in evaluating the
efficacy of treatments. A variety of study designs have been
used in this research.
VITAL DATA ANALYSES
Routinely collected vital data serve as the nation’s key
resource for monitoring progress in caring for mothers
and children. All data presented in this chapter’s figures
and tables are derived from the annual counts of births
and deaths collected by the 52 vital registration areas of
the United States (50 states, Washington, D.C., and New
York City) and then assembled into national data sets by
the National Center for Health Statistics. Unlike data collected in hospitals, in clinics, or from nationally representative surveys, birth and death certificates are required by
law to be completed for each birth and death. Birth and
death registration has been virtually 100% complete for
the entire United States since the 1950s. The universality
of this process renders the findings from vital data analyses stable and generalizable. Figure 1-2 illustrates the most
recent nationally recommended standard for birth certificate data collection, which has been adopted by most
states. Birth certificates contain valuable information for
neonatologists. Especially of note are variables such as to
whether the mother or infant was transferred for care (not
shown), breastfeeding plans, alcohol and cigarette smoking histories, and patterns of maternal weight gain and
prenatal care.
The limitations of vital data are well known. Causes
of death are subject to certifier variability and, perhaps
more importantly, to professional trends in diagnostic
categorization. The accuracy of recording of conditions
and measures on birth certificates is often uncertain and
variable from state to state and hospital to hospital; however, the frequencies of births and deaths in subgroups
defined objectively, such as birthweight, are likely to be
valid.
COHORT STUDIES IN PREGNANCY
AND BIRTH
Studies that follow populations of infants over time, beginning at birth or even before birth and continuing to hospital discharge, early childhood, and adult life, are the leading
sources of information about perinatal risk factors for disease and adverse outcomes. As with all observational studies, cohort studies produce associations of exposures and
CHAPTER 1 Neonatal and Perinatal Epidemiology
FIGURE 1-2 U.S. national standard birth certificate, 2003 revision.
7
8
PART I Overview
outcomes whose strength and consistency must be carefully
judged in the light of other biologic evidence, and with
attention to confounding and bias. Collaborations across
centers in assembling such data are highly valuable. One
noteable collaboration is the Vermont-Oxford Network,
which provides continuous information on the frequency
of conditions observed and diagnoses made in hundreds of
U.S. and overseas hospitals, with a particular emphasis on
using these data for improving care (Horbar et al, 2010).
The neonatal network supported by the National Institute
of Child Health and Human Development (NICHD) has
been a rich source of randomized trials and has produced
observations about prognosis based on large samples of
low-birthweight babies (Fanaroff, 2004). These collaborations focus mainly on the period until hospital discharge.
Multicenter cohort studies focusing on diagnosis and
follow-up of brain injury in premature infants—such as
the Developmental Epidemiology Network (Kuban et al,
1999), Neonatal Brain Hemorrhage (Pinto-Martin et al,
1992), and Extremely Low Gestational Age Newborns
(O’Shea et al, 2009) studies—have contributed to our
understanding of the prognostic value of brain injury
imaged by ultrasound in the neonatal period, because
they include follow-up to the age of 2 years or later. Of
particular value have been regional or population-wide
studies of low-birthweight infants with follow-up to at
least school age, among which are included the Neonatal Brain Hemorrhage study from the United States and
important studies from Germany (Wolke and Meyer,
1999), the Netherlands (Veen et al, 1991), the United
Kingdom (Wood et al, 2000), and Canada (Saigal et al,
1990).
Newborn intensive care has been in place long enough
that the first reports of adult outcomes in small infants are
now emerging (Saigal and Doyle, 2008). These reports
paint a picture that is perhaps less dire than anticipated.
From 1959 to 1966, the National Collaborative Perinatal Project assembled data on approximately 50,000 pregnancies in 12 major medical centers and followed them to
age 7 (Niswander and Gordon, 1972). This highly productive exercise, one of whose major contributions was to
show that birth asphyxia is a rare cause of cerebral palsy,
has now been followed by the development of even larger
birth cohort studies starting in pregnancy. For reasons that
are not entirely clear, a sample size of 100,000 has been
adopted in studies in Norway (Magnus et al, 2006), Denmark (Olsen et al, 2001), and the United States (Lyerly
et al, 2009). A major difference from the National Collaborative Perinatal Project is that each of the studies aims
to obtain some degree of national representativeness. The
U.S. National Children’s Study, currently underway, plans
to enroll women in early pregnancy and possibly before
conception and to follow the offspring to age 21 in 105
locations in the United States, selected by a stratified random sampling of all 3141 U.S. counties.
RANDOMIZED CONTROLLED TRIALS
Few areas of medicine have adopted the randomized trial
as wholeheartedly as has newborn medicine. The number
of trials mounted has been large and have created a strong
influence on practice. A notable influence on this field has
been the National Perinatal Epidemiology Unit at Oxford
University, established in 1978, which prioritized randomized trials among their several investigations of perinatal
care practices and other circumstances affecting maternal
and newborn outcomes. The NICHD neonatal research
network was established in 1986, principally to support trials. Hundreds of trials have been mounted by these two
organizations, but many other centers have contributed to
the trial literature.
Trials in pregnancy or in labor have also been supported by the National Perinatal Epidemiology Unit and
by NICHD, who support a network of obstetric centers
to conduct trials in pregnancy and labor, the NICHD
maternal-fetal research network. These trials have often
had important implications for newborns and mothers,
most notably for showing that the risk of preterm birth
can be reduced by administering 17-OH hydroxyprogesterone caproate in midgestation to high-risk women (Meis
et al, 2003).
Most newborn trials have focused on outcomes evident in the newborn period, such as mortality, chronic
lung disease, brain damage visualized on ultrasound
exam, and duration of mechanical ventilation or hospital stay. Recently however, trials extending into infancy
or early childhood that incorporate measures of cognition or neurologic function have been a welcome addition
to the trial arena. In the past few years, such trials have
shown that moderate hypothermia can reduce mortality
and brain damage in asphyxiated term infants (Shankaran
et al, 2005), and that both caffeine for apnea treatment
(Schmidt et al, 2007) and magnesium sulfate administered
during labor can reduce the risk of cerebral palsy (Rouse
et al, 2008).
Trials in which both mortality and later outcome are
combined raise complex methodologic issues. Imbalance
in the frequency of the two outcomes being combined can
result in a random variation in the more common outcome,
overwhelming a significant finding in the other. Precisely
how best to conduct such dual- or multiple-outcome trials
is the subject of discussion and debate in the neonatal and
epidemiology communities.
As the number of trials increases, not all of them sufficiently powered, the methodology for summarizing them
and drawing effective conclusions has become increasingly
important to neonatologists. The terms systematic review
and metaanalysis have firmly entered the research lexicon,
especially the randomized trial literature. The Cochrane
Collaboration is an international organization that uses
volunteers to systematically review trial results in all fields
of medicine. The collaboration, established in 1993, began
in the field of perinatal medical trials. Systematic reviews
of neonatal trials reviewed by the Cochrane Collaboration
are hosted on the website of the NICHD (http://www.
nichd.nih.gov/cochrane).
SUMMARY
The patterns of disease, mortality, and later outcome in the
perinatal period are complex. Some factors are reasonably
stable (e.g., long-term trends in preterm birth and birthweight), whereas others can undergo rapid change (e.g.,
the rates of cesarean section and twinning). The success of
CHAPTER 1 Neonatal and Perinatal Epidemiology
newborn intensive care is well established. No other organized medical care program, targeted at a broad patient
population, has had such remarkable success in lowering
mortality rates in such a short period of time. Much of
that success is due to the evidence-based nature of neonatal practice.
Nonetheless, this success has opened the door to new
problems as survivors of intensive care face the challenges
of the information age. Resource allocations similar to
those that permitted the development of newborn intensive care are now needed to address the educational and
rehabilitative needs of survivors. A hopeful sign is the success of some recently studied interventions in reducing the
burden of brain damage.
On the nontechnologic front, targeted epidemiologic
efforts to address perinatal disorders have yielded progress. Careful study of the circumstances surrounding infant
sleep patterns led to active discouragement of sleeping
in the prone position, which has reduced mortality from
sudden infant death syndrome by half (The Committee
on Perinatal Health, 1976). Observational research, followed by two important randomized trials in Europe, led
to interventions that increased folate intake in women of
child-bearing age and a substantial reduction in the birth
prevalence of neural tube defects (Czeizel and Dudás,
1992; Medical Research Council, 1991).
The population-level study of health events occurring in pregnancy and infancy, their antecedents, and
their long-term consequences have been an important
component to the success of newborn care. Careful selfevaluation through monitoring of vital data and collaborative clinical data, rigorous assessment of new treatments
9
through randomized trials, and alertness to opportunities to implement prevention activities after discovering
important risk factors should continue to guide the care
of the newborn.
ACKNOWLEDGMENTS
The author thanks Ariel Brovont and Kimberly Harris for
assisting in the preparation of the figures and tables.
SUGGESTED READINGS
Division of Reproductive Health: National Center for Chronic Disease Prevention
and Health Promotion: achievements in public health, 1900-1999: healthier
mothers and babies, MMWR 48:849-858, 1999.
Fanaroff A: The NICHD neonatal research network: changes in practice and outcomes during the first 15 years, Semin in Perinatol 27:281-287, 2004.
Horbar JD, Soll RF, Edwards WH: The Vermont Oxford Network: a community
of practice, Clin Perinatol 37:29-47, 2010.
Hoyert DL: National Center for Health Statistics: maternal mortality and related
concepts, Vital Health Stat 33:1-13, 2007.
Khan KS, Wojdyla D, Say L, et al: WHO analysis of causes of maternal death: a
systematic review, Lancet 367:1066-1074, 2006.
MacDorman MF, Mathews TJ: Centers for Disease Control and Prevention
National Center for Health Statistics: behind international rankings of infant
mortality: how the United States compares with Europe, NCHS Data Brief
23:1-8, 2009.
Niswander KR, Gordon M, editors: The Collaborative Perinatal Study of the National
Institute of Neurological Diseases and Stroke: the women and their pregnancies,
Philadelphia, 1972, WB Saunders.
Saigal S, Szatmari P, Rosenbaum P, et al: Intellectual and functional status at school
entry of children who weighed 1000 grams or less at birth: a regional perspective of births in the 1980s, J Pediatr 116:409-416, 1990.
Silverman WA: Retrolental fibroplasias: a modern parable, New York, 1980, Grune
& Stratton.
World Health Organization: International statistical classification of diseases and related
health problems, Tenth Revision, vol 2, ed 2, Geneva, 2004.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
2
Evaluation of Therapeutic
Recommendations, Database Management,
and Information Retrieval
Peter Tarczy-Hornoch
BACKGROUND
At a fundamental level, the practice of neonatology can be
considered an information management problem. The care
provider is combining patient-specific information (history,
findings of physical examination, and results of physiologic
monitoring, laboratory tests, and radiologic evaluation)
with generalized information (medical knowledge, practice guidelines, clinical trials, and personal experience) to
make medical decisions (diagnostic, therapeutic, and management). The Internet has made possible a revolution in
the sharing and disseminating of knowledge in all fields,
including medicine, with continued growth and maturation
of online clinical information resources and tools. Although
medicine remains a quintessentially human endeavor, computers are playing a growing role in information management, particularly in neonatology. Patient-specific and
generalized information (medical knowledge) are becoming increasingly available in electronic form. In the United
States, a growing number of hospitals are adopting electronic medical record systems to manage patient-specific
information, with the approaches ranging from electronic
flow sheets in the intensive care unit to entirely paperless
hospitals. The American Recovery and Reinvestment Act
of 2009 (ARRA; i.e., the federal economic stimulus plan)
has a provision for the investment of $19 billion in health
information technology to motivate physicians to adopt
electronic health records (in the Health Information Technology for Economic and Clinical Health [HITECH] act, a
part of AARA) and $1.1 billion to research the effectiveness
of certain health care treatments. These ARRA provisions
are predicated on the belief that quality, safety, and efficiency of clinical care can be improved through electronic
medical records and through evidence-based practice.
Parallel with and related to the adoption of information
technology is the growth of societal pressures to improve
the quality of medical care while controlling costs. These
changes are beginning to affect the way in which medicine and neonatology are practiced. In turn, it is becoming
important for neonatologists to understand basic principles related to biomedical and health informatics, databases and electronic medical record systems, evaluation
of therapeutic recommendations, and online information
retrieval.
This expansion of information technology in clinical
practice and the concurrent growth of medical knowledge
have great promise in addition to potential pitfalls. One
pitfall that must not be underestimated, and which is as
great a danger today as when Blois (1984) first cautioned
10
against it, is the unquestioning adoption of information
technology: “And, since the thing that computers do is
frequently done by them more rapidly than it is by brains,
there has been an irresistible urge to apply computers to
medicine, but considerably less of an urge to attempt to
understand where and how they can best be used.” A present and real challenge is information overload. Bero and
Rennie (1995) observed, “Although well over 1 million
clinical trials have been conducted, hundreds of thousands remain unpublished or are hard to find and may
be in various languages. In the unlikely event that the
physician finds all the relevant trials of a treatment, these
are rarely accompanied by any comprehensive systematic
review attempting to assess and make sense of the evidence.” The potential of just-in-time information at the
point of care is thus particularly appealing, especially considering that the growth in published literature continues
at an accelerating rate, with a flood of new knowledge
coming from the latest research in genomics, proteomics,
metabolomics, and systems biology. A vision to address
this was articulated by one of the editors of the British
Medical Journal: “New information tools are needed: they
are likely to be electronic, portable, fast, easy to use, connected to both a large valid database of medical knowledge and the patient record” (Smith, 1996). Although
these goals are close to being achieved, there is still progress to be made before this vision is a reality. This chapter
aims to provide an overview of the current progress in this
direction.
BIOMEDICAL AND HEALTH
INFORMATICS
In the 1970s, clinicians with expertise in computers became intrigued by the potential of these tools to
improve the practice of medicine, and thus the field of
medical informatics was born. The importance of this
field addressing the issues of information management
in health care is growing rapidly, as seen in the activities of the American Medical Informatics Association
(AMIA; www.amia.org). Medical informatics can be concisely defined as “the rapidly developing scientific field
that deals with storage, retrieval, and optimal use of biomedical information, data, and knowledge for problem
solving and decision making” (Shortliffe and Blois, 2006).
A more extensive definition can be found at the AMIA
Web site under About AMIA, including professional and
training opportunities. The University of Washington
CHAPTER 2 Evaluation of Therapeutic Recommendations, Database Management, and Information Retrieval
Web site (www.bhi.washington.edu) contains a review of
the discipline (found under History, About Us, Vision).
The field includes both applied and basic research, with
the focus in this chapter being on the applied aspects.
Examples of basic research are artificial intelligence in
medicine, genome data analysis, and data mining (sorting
through data to identify patterns and establish relationships). As our knowledge of the genetic mechanisms of
disease expands and more data about patients and outcomes are available electronically, the role of informatics in medicine will expand, particularly in the field of
neonatology.
The applied focus of the field in the 1960s and 1970s was
data oriented, focusing on signal processing and statistical
data analysis. In neonatology, the earliest applications of
computers were for physiologic data monitoring in the neonatal intensive care unit (NICU). As the field matured in the
1980s, applied work focused on systems to manage patient
information and medical knowledge on a limited basis. Examples are laboratory systems, radiology systems, centralized
transcription systems, and, probably the best-known medical knowledge management system, the database of published medical articles maintained by the National Library
of Medicine known first as MEDLARS, then as MEDLINE
and currently as PubMed (www.ncbi.nlm.nih.gov/pubmed).
For example, neonatologists began to develop tools to aid in
the management of patients in the NICU, such as computerassisted algorithms to help manage ventilators, although the
algorithms have not been successfully deployed on a large
scale in the clinical setting.
As computers and networking became mainstream
in the workplace and home in the 1990s, informatics
researchers began to develop integrated and networked
systems (Fuller, 1992, 1997). With the explosion of information from the Human Genome Project, the intersection between bioinformatics and medical informatics
began to blur, leading to the adoption of the term biomedical informatics. The 1990s saw the development of a
number of important systems. In terms of patient-specific
information retrieval, these systems included integrated
electronic medical record systems that in their full implementation can encompass—in a single piece of easy-to-use
software—interfaces to physiologic monitors; electronic
flow sheets; access to laboratory and radiology data;
tools for electronic documentation (charting), electronic
order entry, and integrated billing; and modules to help
reduce medical errors. The Internet has permitted ready
access and sharing of this information within health care
organizations and limited secured remote access to this
information from home. In terms of patient population
information retrieval, a number of tools were developed
to help clinicians and researchers examine aggregate data
in these electronic medical records to document outcomes
and help to improve quality of care. The Internet, particularly the World Wide Web, has transformed access to
medical knowledge (Fuller et al, 1999). Health sciences
libraries are becoming digital and paper repositories. Journals are available online. Knowledge is now available at
the point of care in ways that were not previously possible
(Tarczy-Hornoch et al, 1997). In 2004, in recognition of
the unfulfilled potential of health care information technology, the Office of the National Coordinator for Health
11
Information Technology (ONC) was established (www.
healthit.hhs.gov) to achieve the following vision:
Health information technology (HIT) allows comprehensive management of medical information and its secure
exchange between health care consumers and providers.
Broad use of HIT has the potential to improve health care
quality, prevent medical errors, increase the efficiency of
care provision and reduce unnecessary health care costs,
increase administrative efficiencies, decrease paperwork,
expand access to affordable care, and improve population
health.
In the upcoming decade, the focus will be shifting from
demonstrating the potential of electronic medical record
and information systems toward implementing them more
broadly to realize their benefit (e.g., the ARRA legislation). Evidence-based medicine is considered by many as
a part of informatics as an approach to the evaluation of
therapeutic recommendations and their implementation
(see later discussion), and it is part of the ARRA and some
approaches to health care reform being proposed in 2009.
Neonatologists have been involved in informatics for a
long time. Duncan (2010) maintains an excellent continually updated bibliographic database on the literature about
computer applications in neonatology. In 1988, as one of
the earlier groups to develop national databases of clinical
care, neonatologists established and expanded the Vermont
Oxford Network (www.vtoxford.org) to improve the quality and safety of medical care for newborn infants and their
families. As part of the activities, the Vermont established
and maintained Oxford Network a nationwide database
about the care and outcome of high-risk newborn infants.
In 1992, Sinclair et al (1992) published one of the earlier
evidence-based textbooks, Effective Care of the Newborn Infant.
DATABASES
In broad terms, a database is an organized, structured collection of data designed for a particular purpose. Thus, a
stack of 3 × 5 cards with patient information is a database,
as is the typical paper prenatal record. Most frequently,
the term database is used to refer to a structured electronic
collection of information, such as a database of clinical trial
data for a group of patients in a study. Databases come in
a variety of fundamental types, such as single-table, relational, and object-oriented.
A simple database can be built using a single table by
means of a spreadsheet program such as Microsoft Excel,
or a database program such as Microsoft Access (Microsoft,
Redmond, Washington). The advantage of such a database
is that it is easy to build and maintain. For an outcomes database in a neonatology unit, each row can represent a patient
and each column represents information about the patients
(e.g., name, medical record number, gestational age, birth
date, length of stay, patent ductus arteriosus [yes/no], necrotizing enterocolitis [NEC; yes/no]). The major limitation
of such a database is that a column must be added to store
the information each time the researcher wants to track
another outcome (e.g., maple syrup urine disease [MSUD]).
This limitation can result in tables with dozens to hundreds
of columns, which then become difficult to maintain. The
challenges can be illustrated with a few examples. The first
12
PART I Overview
example of a challenge is that which results from adding
a new column (e.g., MSUD); one must either review all
records (rows) already in the spreadsheet for the presence
or absence of MSUD or flag all existing records (rows) in
the spreadsheet as unknown for MSUD status. The second
example of a challenge results from the logistics of managing an extremely wide spreadsheet—imagine not adding
the tenth column but the 1000th column.
The majority of databases and electronic medical records
in neonatology are built using relational database software.
To build a simple outcomes relational database that permits easy adding of new outcome measures, one could use a
three-table database design (Figure 2-1). The first table contains all the information for each patient (e.g., name, medical record number, gestation in weeks, birth date, admit
date, discharge date). The second table is a dictionary that
assigns a code number to each diagnosis or outcome being
tracked (e.g., patent ductus arteriosus = 1; NEC = 2; MSUD
= 10234). The third table is the diagnosis-tracking table;
it links a patient number to a particular code and assigns a
value to that code. Adding a new diagnosis to track would
require adding an entry to the diagnosis dictionary table.
To add a diagnosis to a patient, one would add an entry to
the tracking table. For example, Girl Smith (medical record
number 00-00-01) has a diagnosis of NEC. To add the diagnosis, add to the diagnosis table an entry that has a value of
00-00-01 in the medical record column, a value of 2 (the
code for NEC) in the code column, and a value of 2 in the
value column (the code for surgical). Although relational
databases are harder to build, they provide greater flexibility
for expansion and maintenance and thus are the preferred
implementation for clinical databases. They address the
challenges in a simple spreadsheet by tracking dates that
new diagnostic codes were added and by user interfaces that
allow one to easily view only diagnoses present for a given
patient rather than all potential diagnoses for a patient.
The distinction between an NICU quality assessment–
quality improvement (i.e., outcomes) database and an electronic medical record is largely a matter of degree. Some
characteristics typical of a neonatal outcomes database are
data collection and data entry after the fact, limited amount
of data collected (a small subset of the information needed
for daily care), lack of narrative text, lack of interfaces to
laboratory and other information systems, and the episodic
(e.g., quarterly) use of the system for report generation.
Some characteristics typical of an electronic medical record
are real-time (daily or more frequent) data entry, a large
amount of data collected (approximating all the information
needed for daily care in a fully electronic care environment),
narrative text (e.g., progress notes, radiology reports, pathology reports), interfaces to laboratory and other information
systems, and, most important, the use of the system for daily
patient care, including features such as results review, messaging or alerting for critical results, decision support systems (drug dosage calculators, drug-drug interaction alerts,
among others), and computerized electronic order entry.
In the past, the majority of neonatal databases and firstgeneration NICU electronic medical record systems were
FIGURE 2-1 Example of a relational database.
CHAPTER 2 Evaluation of Therapeutic Recommendations, Database Management, and Information Retrieval
developed locally by and for neonatologists. The literature
describing these efforts is available online (Duncan, 2010).
Unfortunately the majority of these systems were never published or publicly documented, and thus a number of important and useful innovations are lost or must be repeatedly
rediscovered. Anybody thinking about building their own
neonatology database would be well advised to review the
existing literature and existing commercial products before
embarking on this path. That said, there is room for improvement of the existing products, and neonatologists continue
to develop their own databases today. With the national push
toward interoperable electronic medical records through
U.S. Department of Health and Human Services Office of
the National Coordinator for Health Information Technology (ONC; since 2004) and ARRA HITECH (since 2009), it
is likely that the market will consolidate into a smaller number of neonatology practice tailored systems that are certified
and that interoperate with the National Health Information
Network (2010). The largest neonatal outcomes database is
the centralized database maintained by the Vermont Oxford
Network with the mission of improving the quality and safety
of medical care for infants and their families. One of the key
activities of the network is their outcomes database, which
involves more than 800 participating intensive care nurseries
both in the United States and internationally collecting data
on approximately 55,000 low-birthweight infants each year.
Other activities of the network are clinical trials, follow-up
of extremely low–birthweight infants, and NICU quality and
safety studies. The focus of the database initially was very
low–birthweight infants (401 to 1500 g), but this focus has
expanded to include data on infants weighing more than
1500 g. Presently the network collects data on more than
two thirds of the very low–birthweight infants born in the
United States. Participants in the network submit data and
in return receive outcome data for their own institution and
comparative data from other nurseries nationwide, including custom reports, comparison groups, and quality management reports. Members also have the ability to participate
in collaborative research projects and collaborative multicenter quality-improvement collaborations. All data except
their own are anonymous for all participants. The network
does have access to both the individual and aggregate data.
The network database is maintained centrally, and data quality monitoring and data entry are centralized. Initially the
process involved paper submission of data by participating
nurseries. Currently a number of the commercial and custom NICU databases and electronic medical record systems
can export their data in the format required by the Vermont
Oxford Network as well as the option to submit data directly
using the custom eNICQ software developed by Vermont
Oxford. Submissions of data are thus a combination of paper
forms and reports generated by commercial and custom software packages.
The database focuses on tracking outcomes. With the
passage of the Health Insurance Portability and Accountability Act of 1996 (HIPAA) and federal regulations governing the confidentiality of electronic patient data, some
of the anonymous demographic data that were collected by
the network in the past have decreased. This change has
grown from concerns that, in combination with identity of
the referring center, these data could be used to uniquely
identify patients, which is a violation of the HIPAA.
13
ELECTRONIC HEALTH RECORD
An electronic health record (EHR; also known as an electronic medical record) is much more complex than an outcomes database, because the system is intended to be used
continually on a daily basis to replace electronically some,
if not all, of the record keeping, laboratory result review,
and order writing that occur in a neonatal intensive care
nursery (or more generically in any inpatient or outpatient
clinical setting). The complexity of this task becomes evident if one imagines that, for a paperless medical record
environment, every paper form in a nursery would need
to be replaced with an electronic equivalent, which is
also true for every paper-based workflow and process.
Organizations are moving in this direction because of a
combination of forces, such as the desire to reduce error
and to control the spiraling costs of health care. These
reasons are addressed at great length in two reports from
the Institute of Medicine (Institute of Medicine Committee on Improving the Patient Record, 1997; Kohn
et al, 2000). These benefits are typically achieved when
information is available electronically (e.g., results of
laboratory tests, radiology procedures, transcription) and
input into the system (e.g., problem lists, allergies), and
when both sets of information are combined and checked
against electronic orders. Only with electronic orders has
it been shown that errors can be reduced and care provider behavior clearly changed. Combining just electronic
laboratory results (e.g., creatinine level) and electronic
order entry (e.g., a drug order), for example, enables one
to verify that drug dosages have been correctly adjusted
for renal failure. This approach would work well in adults,
but in neonates, whose renal function is more difficult to
assess and for whom drug dosage norms depend on gestational age and post-delivery age, additional information
must be entered into the system (e.g., urine output, gestational age), requiring a more sophisticated EHR. Despite
more than one decade of work deploying EHRs, the proportion of acute care hospitals that are members of the
American Hospital Association and have a comprehensive
EHR is remarkably low (1.5%), and computerized order
entry has been implemented in only 17% of hospitals (Jha
et al, 2009).
Results review systems include basic demographic data,
such as name, age, and address from the hospital registration system. These systems require a moderate amount of
work to tie them to the various laboratory, radiology, and
other systems and to train users. The benefits are hard
to quantify, but users typically prefer them to the paper
alternative because of the more rapid access to information. The challenge in moving beyond the results review
level to the integrated system level is that the documentation level and order entry level are essentially prerequisites for the integrated system level, but they have
marginal benefit, particularly given the human and financial costs. Integrated systems require significant work to
implement, including the presence of computers at each
bedside, as well as significant work to train users. The
benefits accrue mainly to the organization, in the form of
reduced costs of filing, printing, and maintaining paper
records and, if providers are forced to enter notes instead
of dictate them, significant savings in transcription costs.
14
PART I Overview
The challenge is that the end users often find that it takes
much longer to do their daily work with electronic documentation. Without moving to electronic order entry, if
not an integrated system, the users do not realize major
day-to-day benefits. The ARRA HITECH provisions
noted earlier essentially are designed to create incentives
for provider adoption of EHRs to overcome this activation barrier.
The benefits start to accrue more clearly at the next
level—electronic order entry. The complexity of implementing and deploying an electronic order entry system
cannot be overstated. Interfaces need to be built with all the
systems that are part of results review in addition to other
systems. Furthermore, a huge database of possible orders
must be created to allow users to pick the right orders.
This database and the menu of choices are needed because
computers are poor at recognizing and interpreting a narrative text typed by a human. Finally and most important,
there is a huge training challenge, because writing orders
electronically is more complex and time consuming than
writing them by hand. The change management issues
become apparent when one considers that typically these
systems take the unit assistant out of the loop; therefore
much of the oversight that can occur at the unit assistant
level does not, or the burden of oversight is borne by the
person entering the orders.
After overcoming the barriers to electronic order entry,
organizations can start to benefit from integrated systems.
For this reason, the trend today is not a stepwise move
from results review to documentation of integrated systems. Instead, organizations are moving from results
review directly to integrated systems. Interestingly, the
technical complexities and the training and usage complexities of integrated systems are not much higher than
those for order entry. Integrated systems add tools to make
life easier for care providers using all the data in the system. As an analogy, an integrated EHR system is like an
office software suite that encompasses a word processor,
a spreadsheet, a slide presentation tool, a graphic drawing
tool, and a database, all of which can communicate with
one another, making it easy to put a picture from the drawing tool or a graph from a spreadsheet into a slide show.
Integrated systems include (1) checking orders for errors,
(2) alerts and reminders triggered by orders or by problems on the problem list or other data in the system, (3)
care plans tied to patient-specific information, (4) charting modules customized to the problem list, (5) charting
and progress notes that automatically import information (e.g., from laboratory tests, flow sheets) and that help
generate orders for the day as the documentation occurs,
(6) modules to facilitate hyperalimentation ordering, and
(7) modules to assist in management. For example it is possible to imagine a system in which reminders for screening
studies (e.g., for retinopathy of prematurity, intraventricular hemorrhage, and brainstem auditory evoked response)
were triggered by gestational age, a problem list, and previous results of screening studies. Similarly, admitting a
neonate at a particular gestational age with a particular set
of problems could trigger pathways, orders, and reminders specific to that clinical scenario. An important caveat
is that all such systems are only as good as the data and
rules put into them. The issues raised in the section on
evaluation of therapeutic recommendations are important
to consider in the context of electronic order entry and
integrated systems.
The EHR market is still relatively young and continually evolving; this is true of products designed specifically
for the NICU and more generic products designed to be
used throughout a hospital or health care system. The
ONC was established in part to address this young marketplace by creating standards and certification bodies. In
particular, the ONC created the Certification Commission for Healthcare Information Technology (CCHIT)
and the Health Information Technology Standards Panel
to begin to bring more standardization to the marketplace.
The CCHCIT examines criteria for certifying systems,
including functionality, security, and interoperability.
Order entry and documentation systems are beginning to
be more widely adopted, however truly integrated systems
are much less broadly implemented. The major reason
for this situation is that the needs of different health care
systems vary significantly, and the existing products are
not flexible enough to meet all these needs in one system.
Furthermore, there is a trend among health care organizations, EHR developers, and vendors to move away from
niche systems tailored to particular subsets of care providers, such as neonatology, and toward a focus on systems
that are generically useful. There are two important drivers behind this trend.
The first and most important reason for adopting a single integrated system is that the benefits of an EHR system begin to accrue only when an entire organization uses
the same one. Consider the following scenario: a woman
receives prenatal care in the clinic of an institution and is
then admitted to the emergency department in preterm
labor. Her infant is delivered in the labor and delivery
department, hospitalized in the neonatal intensive care
nursery, and discharged to an affiliated pediatric follow-up
clinic. In the current era of paper medical records, paper
is used to convey information from one site to the other.
In an integrated EHR system, all the information for both
mother and infant is in one place for all providers to see.
Interoperability is important as well if the care described
crosses organizational boundaries, such as an outpatientfocused health maintenance organization contracting inpatient obstetric care to one hospital system and neonatal or
pediatric care to a children’s hospital in a different health
care system. If a single unified institution were to adopt
niche software tailored to the needs of each site, a provider
caring for the infant might need to access an emergency
department system, an obstetric system, an NICU system,
and an outpatient pediatric system to gather all the pertinent information. Each system would require the user to
learn a separate piece of software. Learning a site-specific
piece of software is a considerably greater burden on care
providers than learning to use a site-specific paper form. If
care crosses organizations and electronic systems are not
interoperable, then care transitions most often remain on
paper.
The second factor driving adoption of integrated systems is economies of scale. The ideal EHR system contains
electronic interfaces that automatically import the system
data from laboratory, pharmacy, radiology, transcription,
integrated electronic orders, error checking, and electronic
CHAPTER 2 Evaluation of Therapeutic Recommendations, Database Management, and Information Retrieval
documentation by care providers. Given that development
of these interfaces, training, and maintenance cost more
than the purchase of the system itself, it is far more cost
effective to install one system with one set of interfaces and
one set of training and maintenance issues than to replicate
the process multiple times.
The neonatal intensive care environment poses some
unique challenges for EHRs. As a result, it is important
to ensure that when health care systems are making decisions about the purchase of an EHR, neonatologists and
other neonatal health care providers are involved in the
process. An excellent source of information about NICU
medical record systems and databases is an article by Stavis
(1999). Neonatologists in the position of helping to select
an EHR system must acquire the necessary background
through reading some basic introductory texts on medical informatics, focusing on EHRs. It is then critical that
they survey other organizations similar to their own to
discover which systems have worked and which ones have
not. For example, the needs of a level III academic nursery
that performs extracorporeal membrane oxygenation are
different from those of a community level II hospital that
does not perform mechanical ventilation. Systems that
work well in teaching hospitals with layers of trainees may
not work well in private practice settings and vice versa.
Most important, when using a medical informatics framework, the neonatologists must develop a list of prioritized
criteria specific to their institution and compare available
products in the marketplace with this list, while also considering the recommendations of the CCHIT as described
earlier.
All end user needs must also be considered. If residents,
nurse practitioners, nutritionists, pharmacists, and respiratory therapists are expected to use the system, their input
must be solicited. Ensuring broad-based input is especially
relevant if the goal is an EHR system into which a lot of
data will be entered by health care providers (e.g., electronic charting, note writing, medication administration
records, order entry). The reason to ensure acceptance by
all users of systems that require data entry is that a significant percentage of systems requiring data entry has
ultimately been unsuccessful because of lack of user acceptance. Unfortunately there is little literature on this issue,
because institutions rarely publicize and publish their failures in this arena, although the situation is beginning to
change. A review of some of these challenges and a theoretical framework for looking at them is provided by Pratt
et al (2004).
The final step in evaluating a potential system is to
develop a series of scenarios and to have potential users
test the scenarios. Evaluating usage scenarios typically
involves visits to sites that have installed the EHR system
under consideration. An example of a scenario might be for
a nurse, a respiratory therapist, a resident, and an attending physician to try to electronically replicate, on a given
system under consideration, the bedside charting, progress
note charting, and order writing for a critically ill patient
who undergoes extracorporeal membrane oxygenation and
then decannulation. The reason for developing and testing
such scenarios is that this approach is the best way to ensure
that aspects of charting, note writing, and documentation
unique to the NICU are supported by the system.
15
EVALUATING THERAPEUTIC
RECOMMENDATIONS
Once all the data about a patient, whether in electronic
or paper form, are in hand, the clinician is faced with the
challenge of medical decision making and applying all that
he or she knows to the problem. It is vital that the clinician understand what is known and what is still uncertain in
terms of the validity of therapeutic recommendations. The
evaluation of new recommendations arising from a variety
of sources, including journal articles, metaanalyses, and systematic reviews, is a critical skill that all neonatologists must
master. Broadly speaking, this approach has been termed
evidence-based medicine. A full discussion of this approach to
evaluation of new approaches in clinical medicine is beyond
the scope of this chapter. Two outstanding sources of information are the works by Guyatt and Rennie (2002) and
Straus et al (2005). An excellent overview of the progress in
evidence-based medicine over the last 15 years is provided by
Montori and Guyatt (2008). An important caveat is that evidence-based medicine is not a panacea. It is not helpful when
the primary literature does not address a particular clinical
situation, such as one that is rare or complex. This approach
also does not necessarily address broader concerns, such as
clinical importance or cost effectiveness, although it sometimes does. There are potential challenges when combining evidence-based medicine with the emerging humanistic
approach to health care, which can heavily weight patient
preferences potentially over the evidence base.
In the early days of medicine, the standard practice was
observation of individual patients and subjective description
of aggregate experiences from similar patients. As the science
of medicine evolved, formal scientific methods were applied
to help to assess possible therapeutic and management interventions. Important tools in this effort are epidemiology,
statistics, and clinical trial design. Currently medicine in general and neonatology in particular are faced with an interesting paradox. For some areas, there is a wealth of information
in the form of randomized controlled clinical trials, whereas
there is scant information to guide clinical practice for others. A wealth of well-designed clinical trials on the use of
surfactant has been published, for example, but there are
essentially no trials addressing the management of chylothorax. One might assume that the practice of medicine reflects
the available evidence, but this is not the case. McDonald
(1996) summarized the problem as follows: “Although we
assume that medical decisions are driven by established scientific fact, even a cursory review of practice patterns shows
that they are not.” A study of 2500 treatments in the British
Medical Journals Clinical Evidence Database (http://clinicalevidence.bmj.com), shows that, as of the summer of 2009,
49% of treatments are of unknown effectiveness, 12% are
clearly beneficial, 23% are likely to be beneficial, 8% are a
tradeoff between beneficial and harmful, 5% are unlikely to
be beneficial, and 3% are likely to be ineffective or harmful.
As a result, neonatologists have a responsibility to identify
what knowledge is available in the literature and elsewhere
and to critically evaluate this information before applying it
to practice. Furthermore, because this information is constantly evolving, practitioners must continually revisit the
underlying literature as it expands (e.g., the recommendations regarding the use of steroids for chronic lung disease).
16
PART I Overview
The evidence-based practice of medicine is an approach
that addresses these issues. It is helpful to consider the
process as involving two steps—the critical review of the
primary literature and the synthesis of the information
offered in the primary literature. Critical review of the
primary literature is an area in which most neonatologists
have significant experience, with journal clubs and other
similar forums. The approach involves systematically
reviewing each section of an article (i.e., background,
methods, results, discussion) and asking critical questions for each section (e.g., for the methods section: Is the
statistical methodology valid? Were power calculations
made? Was a hypothesis clearly stated? Do the methods
address the hypothesis? Do the methods address alternative hypotheses? Do the methods address confounding
variables?). The formal evaluation of each section must
then be synthesized into conclusions. A helpful question
to ask is, “Does this paper change my clinical practice,
and if so, then how?” Additional resources for systematic
review of the primary literature are listed in the Suggested Readings. It is important to note that guidelines
for systematic review of a single article differ according to
whether it describes a preventive or therapeutic trial (e.g.,
use of nitric oxide for chronic lung disease), evaluation
of a diagnostic study (e.g., use of C-reactive protein level
for prediction of infection), or prognosis (e.g., prediction
of outcome from a Score for Neonatal Acute Physiology
score).
The second, and arguably more important, step is to
determine not the effect of one article on one’s practice,
but the overall effect of the body of relevant literature on
one’s practice. For example, if the preponderance of the
literature favors one therapeutic recommendation, then
a single article opposing the recommendation must be
weighed against the other articles that favor it. This task
is complex, and the most complete and formal statistical
approach to combining the results of multiple studies (i.e.,
metaanalysis) requires significant investment of time and
effort. Part of the evidence-based practice of medicine
approach therefore involves the collaborative development
of evidence-based systematic reviews and metaanalyses by
communities of care providers. Within the field of neonatology, Sinclair et al (1992) laid the seminal groundwork
for this approach; their textbook Effective Care of the Newborn remains an important milestone, but it illustrates the
problem of information currency. Because the book was
published in 1992, none of the clinical trials in neonatology in the last decade and a half are included. The Internet has permitted creation and continual maintenance of
up-to-date information by a distributed group of collaborators, lending itself well to the maintenance of a database of evidence-based medicine reviews of the literature.
This international effort is the Cochrane Collaboration,
and the Cochrane Neonatal Review is devoted to neonatology (Cochrane Neonatal Collaborative Review Group). A
limitation of the Cochrane approach is illustrated by the
relatively restricted scope of topics covered at the National
Institute of Child Health and Human Development Web
site (www.nichd.nih.gov/cochrane/cochrane.htm). The
existence of a review requires adequate literature on a
topic and a dedicated and committed clinician to create
and update the review.
It is important to distinguish between these formal
approaches to reviewing the literature (i.e., systematic
literature reviews and metaanalyses) that have specific
methodologies and more ad hoc reviews of the literature.
Evidence-based medicine aggregate resources such as the
Cochrane Collaboration take a more systematic approach,
but review articles published in the literature vary in their
approach. Metaanalyses are easy to distinguish, but systematic reviews versus ad hoc reviews are harder to distinguish. Systematic reviews focus on quality primary
literature (e.g., controlled studies rather than case series
or case reports) and must include (1) a methods section for
the review article that explicitly specifies how articles were
identified for possible inclusion and (2) what criteria were
used to assess the validity of each study and to include or
exclude primary literature articles in the systematic review.
Systematic reviews also tend to present the literature in
aggregate tabular form, even when metaanalyses of statistics of all the articles cannot be done. One commonly
used source of overview information in neonatology—the
Clinics in Perinatology series—is a mix of opinion (written
in the style of a book chapter), ad hoc literature review,
systematic literature review, and metaanalysis. Guidelines
(e.g., screening recommendations for group B streptococcal infection), although based on primary literature review,
are typically neither metaanalyses nor systematic reviews of
the literature. Whereas formal methods are used to derive
conclusions with metaanalyses and systematic reviews,
guidelines are developed frequently instead by consensus
among committee members; this is true of both national
and local practice guidelines. General textbooks of neonatology are typically based on ad hoc literature review that
includes both primary literature and systematic literature
review. When reading overviews of the aggregate state of
current knowledge on a given topic in neonatology, it is
important to keep these distinctions in mind.
Anyone interested in developing evidence-based reviews
on a particular topic should review some of the textbooks
on evidence-based practice listed at the end of this chapter.
Initially, it is a good idea to collaborate with someone who
has experience in systematic review and metaanalysis. The
process consists of the following steps: (1) identifying the
relevant clinical question (e.g., management of bronchopulmonary dysplasia); (2) narrowing the question to a focus
that enables one to determine whether a given article in the
primary literature answers it (e.g., does prophylactic highfrequency ventilation have positive or negative effects on
acute and chronic morbidity—pulmonary and otherwise?);
(3) extensively searching the primary literature (frequently
in collaboration with a librarian with expertise searching the biomedical literature) and retrieving the articles;
(4) critically, formally, and systematically reviewing each
article for inclusion, validity, utility, and applicability; and
(5) formally summarizing the results of the preceding process, including conclusions valid throughout the body of
included primary literature.
ONLINE INFORMATION RETRIEVAL
Because of the rapid growth of biomedical information
and because it changes over time, investigators in informatics and publishers believe that print media will soon
CHAPTER 2 Evaluation of Therapeutic Recommendations, Database Management, and Information Retrieval
no longer be used for sharing and distributing biomedical
information (Smith, 1996; Weatherdall, 1995). Economic
realities will dictate, however, that quality information will
generally come at a price. The medical digital library at the
University of Washington (www.healthlinks.washington.e
du, under Care Provider) serves to illustrate the current
state of the art. The information available is a combination
of locally developed material (e.g., University of Washington faculty writing practice guidelines), material developed
by institutions elsewhere (faculty elsewhere writing such
guidelines), material developed by organizations (e.g., the
neonatal Cochrane Collaboration), and electronic forms
of journals, textbooks, and evidence-based medicine databases. The last (journals, textbooks, databases) generally
are not free, and the University of Washington pays a subscription fee (termed a site license) to provide access to these
resources for their faculty, staff, and students. Libraries
will likely remain the primary source of information, but
will shift their attention from paper to electronic records.
Health sciences libraries can provide invaluable training
in the efficient use of online medical resources, and most
offer training sessions and consultation.
A number of online resources are valuable for neonatologists. For accessing the primary literature, the most valuable resource is the National Library of Medicine’s database
of the published medical literature accessible (PubMed;
www.ncbi.nlm.nih.gov/entrez) along with many other databases accessible from the Health Information Web site
(www.nlm.nih.gov/hinfo.html). The PubMed system is continually being enhanced; therefore it is useful to review the
help documentation online and regularly check the New/
Noteworthy section to see what has changed. One of the
most powerful yet underused tools is the Find Related Link
that appears next to each article listed on PubMed. This
link locates articles that are related to the one selected (Liue
and Altman, 1998). The PubMed system applies a powerful
statistical algorithm with complex weightings to the article
selected to each word in the title, to each author, to each
major and minor keyword (Medical Subject Heading terms),
and to each word in the abstract and then finds statistically
similar articles in the database. In general, this system outperforms novice to advanced health care providers performing a complex search, and it begins to approach the accuracy
of an experienced medical librarian. Another powerful search
tool within PubMed is the Clinical Query (www.ncbi.nlm.
nih.gov/corehtml/query/static/clinical.shtml). This tool
facilitates searches for papers by clinical study category (e.g.,
etiology, diagnosis, therapy, prognosis), focuses on systematic reviews, and performs medical genetics searches—the
last of which is useful in the context of neonatology. All the
major pediatric journals are available online either through a
package at local hospital libraries or by subscription, instead
of or in addition to a print subscription. The value of having an electronic subscription to a journal is that it provides
access to current and past issues. The best free online source
of information on evidence-based practice is the Cochrane
Neonatal Review. Subscriptions to the full Cochrane database can be purchased online as well.
Given the growing role of genetics in health care, and
in particular the importance of genetic diseases in infants,
there are two notable genetics databases that are available
17
free of charge online (in addition to the Medical Genetics
Searches mentioned previously). The first is Online Mendelian Inheritance in Man (www.ncbi.nlm.nih.gov/omim).
This database, which is a catalog of human genes and
genetic disorders, is an online version of the textbook by the
same name. It is a diachronic collection of information on
genetic disorders, meaning that each disease entry chronologically cites and summarizes key papers in the field. The
second is the GeneTests database (www.genetests.org),
which is a directory of genetic testing (what testing is
available on a clinical and research basis, where, and how
one sends a specimen) and a user’s manual (how to apply
genetic testing). The user’s manual section consists of
entries for a growing number of diseases or clinical phenotypes of particular importance. Entries are written by
experts, peer reviewed both internally and externally, subjected to a formal process similar to a systematic review,
and updated regularly online. As of the winter of 2011, the
GeneTests database includes GeneReviews (user’s manual
entries) for 527 diseases, and the directory includes 1189
genetics clinical, 595 laboratories, and information on testing for 2270 diseases (2005 clinically and 265 on a research
basis). An excellent site that maintains links to the majority
of locally developed, neonatology-specific content around
the country is the site maintained by Duncan (2010). In
addition to a database of links to clinical resources around
the country, the site also has a job listing and a database of
the literature on computer applications in medicine.
The clinician must realize that, unlike journals, textbooks, and guidelines, the material on the World Wide
Web (whether accessed from Duncan’s site or based on a
search) is not necessarily subject to any editorial or other
oversight as to what is published; therefore, as stated
by Silberg et al (1997), “caveat lector” (reader beware).
A number of articles and Web sites address criteria
for assessing the validity and reliability of material on a
Web site. (Health on the Net Foundation, www.hon.ch
Mitretek Systems). With caution in mind, a search on
the entire World Wide Web using a sophisticated search
engine (e.g., Google, Google Scholar) can yield valuable information, though search results typically include
a lower proportion of quality resources compared with
curated resources. Google also has a sophisticated statistical algorithm that allows a user to find similar Web pages
after identifying a particular one of interest.
SUGGESTED READINGS
Cochrane Neonatal Collaborative Review Group: Cochrane neonatal review.
Available from www.nichd.nih.gov/cochrane.
Jha AK, DeRoches CM, Campbell EG, et al: Use of electronic health records in
U.S. hospitals, NEJM 360:1628-1638, 2009.
Montori VM, Guyatt GH: Progress in evidence-based medicine, JAMA
300:1814-1816, 2008.
Norris T, Fuller SL, Goldberg HI, et al: Informatics in primary care: strategies in
information management for the healthcare provider, New York, 2002, SpringerVerlag.
Pratt W, Reddy MC, McDonald DW, et al: Incorporating ideas from computer
supported cooperative work, J Biomed Inform 37:128-137, 2004.
Shortliffe EH, Cimino JJ, editors: Biomedical informatics: computer applications in
health care and biomedicine, ed 3, New York, 2006, Springer Science+Business
Media.
Straus SE, Richardson WS, Glasziou P, et al: Evidence-based medicine: how to practice
and teach EBM, ed 3, London, 2005, Churchill Livingstone.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
3
Ethics, Data, and Policy in Newborn
Intensive Care
William L. Meadow and John D. Lantos
PHILOSOPHY
Ethics in the neonatal intensive care unit (NICU), as in
all clinical contexts, starts with the traditional triangular
framework of autonomy (do what the patient, or in this
case the parent, thinks is right), paternalism (do what the
doctor thinks is right), and beneficence and nonmaleficence (do the right thing). These concepts, independent of
context or data, are timeless.
The problem with timeless concepts, of course, comes in
knowing what to do in real time. What exactly is the right
thing? What facts should be brought to bear in the decision?
What weight should each fact be given? And whose opinion
should count in the end? Nonetheless, some applications of
traditional ethical concepts in the NICU are already universally adopted (e.g., avoid futility, do not torture, and intervene when the data provide compelling evidence to do so).
Unfortunately, much of NICU care falls between not
resuscitating 21-week births, obligatory support of 28-week
births, and not performing cardiopulmonary resuscitation
on infants with lethal anomalies. The traditional ethical
solution to medical dilemmas is to ground concerns in context, take data into account, and be sympathetic to patient
preferences when the balance of benefits and burdens is not
clear. The precise problem in the NICU is that the burdens
are real, immediate, long term, and significant—months of
painful procedures such as intubation, ventilation, intravenous catheterization, and any permanent sequelae that
might ensue—whereas the benefits of NICU interventions
are distant, statistical, and unpredictable. Moreover, NICU
success is often viewed as “all or none”—that is, in most of
the NICU follow-up literature a Bayley mental developmental index MDI or psychomotor developmental index
PDI greater than 70 is classified as normal, whereas an MDI
or PDI less than 70 is classified as an adverse outcome.
Faced with a difficult case, it is rare that simply applying
principles will help to devise a solution. Difficult cases are
usually ones in which the principles themselves are in conflict, or their application to the case is ambiguous.
DATA
What kind of information would parents, physicians, or
judges want to know about the NICU? The essential truth
at the intersection of NICU epidemiology and ethics is that
survival depends sharply on gestational age (GA), within relatively precise boundaries. In the United States, as in virtually all the industrialized world, infants born after 27 weeks’
gestation have, from any ethical perspective, no increased
mortality over infants born at term. Consequently, for these
infants, the ethical principle of best interests requires their
resuscitation, in the same way that sick children born at
term deserve resuscitation.
18
Conversely, for infants born before 22 weeks’ gestation,
survival is essentially zero. Consequently, these infants and
their parents deserve our compassion, but not our interventions, on the ethical grounds of strict futility.
In between, spanning roughly one gestational month,
from 23 to 26 weeks, we will require not just data, but
interpretation. First, there is the intriguing finding that
GA-specific mortality for infants resuscitated in this gestational range has more or less reached a plateau. Within
10 to 20 years ago, the “border of viability” was steadily
decreasing, thus improving outcomes for infants in the
gray zone. Not so much, recently, and there is little reason
to think that things will change for infants born in this
gestational range in the foreseeable future.
Nonetheless, epidemiologic progress has been made.
Tyson et al (2008), using the vast data base of the National
Institute of Child Health and Human Development network, attempted to go “beyond gestational age” and quantify additional risk factors for both mortality and neurologic
morbidity in infants born on the cusp of viability. The
analysis revealed that singleton status, appropriate in utero
growth, antenatal steroids, and female gender all improve
the likelihood of survival and intact neurologic outcome,
independent of GA. Tyson’s algorithm, using information
available at the time of birth, significantly improved predictive value and sensitivity of prognostication over GA alone.
However, two problems remain. First, for many infants
the predictive value of the Tyson algorithm is still not very
good—that is, many of the lower-risk patients will still
die, and many of the higher-risk patients will survive. Second, the Tyson algorithm, like GA, ignores a potentially
important feature of NICU care—time. The algorithm
stops accumulating data at the time of birth, and it does
not account for prognostic features that might become
available as the infant’s course unfolds in the NICU. In
theory, making decisions over the course of time offers
two advantages over prognostication in the delivery room.
First, parents often appreciate the opportunity to get to
know their baby as an individual, as opposed to evaluating
anonymous population-based prognostications at the time
of birth. Second, there is valuable information to learn
while the baby is in the NICU.
Two time-sensitive prognostic features have been
evaluated in the context of infants born at the border of
viability—serial illness severity algorithms (Score for Neonatal Acute Physiology [SNAP] scores) and serial that intuitions that the patient would “die before NICU discharge”
(Meadow et al, 2008). Unfortunately, although SNAP
scores on the first day of life have good prognostic power
for death or survival, their power diminishes over time.
Intriguingly, serial intuitions that an individual baby will
die before discharge—offered by medical caretakers for
CHAPTER 3 Ethics, Data, and Policy in Newborn Intensive Care
patients who require mechanical ventilation and for whom
there is an ethical alternative to continued ventilation,
namely extubation and palliative care—are remarkably
accurate in predicting a combined outcome of either death
or survival with neurologic impairment (MDI or PDI <70).
Children with abnormal results from a cranial ultrasound
examination and corroborated predictions of death have a
less than 5% chance of surviving with both MDI and PDI
greater than 70 at 2 years, independent of their gestational
age. The predictive power of these data, acquired over
time during an individual infant’s NICU course, though
not perfect, is greater than any algorithm available at the
time of birth.
What do prospective parents or medical caretakers
consider when they are asked to decide whether or not
to resuscitate their micro-premie? A fascinating insight
has been offered by Janvier et al, (2008), who have done
extensive surveys comparing responses to requests for
resuscitation of sick micro-premies with resuscitation of
comparably sick patients at other ages (from term infants
to 80-year-olds). Consistently, it appears that micropremies are devalued—that is, for comparable likelihood
of survival and comparable likelihood of neurologic morbidity in survivors, more people would let a micro-premie
die first, or at least offer to resuscitate them last. There is
no theory to account for these findings.
Indeed, for male infants born at 23 weeks’ gestation, the
likelihood of intact survival (i.e., neither death in the NICU
nor permanent neurologic morbidity) is less than 10%.
Given that the likelihood of burdensome therapy is 100%,
how can resuscitation be justified? Intriguingly, combining
the outcomes of death in the NICU with abnormal neurologic function in later life may not reflect the emotional
reality of many NICU parents. For these parents, death in
the NICU is not necessarily the worst outcome, because
acknowledging that the baby was too small might be better
consolation than not ever giving the baby a chance at life.
If trying and failing is seen as a positive process, then
the long-term burden of NICU care for parents should
perhaps be calculated not as a function of all live births (the
current practice), but as a function of infants who survive
to discharge. Numerous studies analyzing various populations in several countries have converged on the same surprising observation: the incidence of neurologic morbidity
in NICU survivors is not very different when comparing
infants at 23, 24, 25, and 26 to 27 weeks’ gestation. The
essential epidemiologic difference for infants born in this
gestational range appears to be whether the baby will survive at all. Once the baby leaves the NICU, the risk of
severe morbidity is largely the same; this is true in singlecenter and multicenter studies, in the United Kingdom,
Canada, Europe, and the United States (Johnson et al,
2009; Tyson et al, 2008). Paradoxically, if avoiding survival
with permanent crippling neurologic injury is the driving
force behind resuscitation decisions, it appears that we
should not be worrying about 23- and 24-weekers–rather
we should not be resuscitating 26- or 27-weekers, since
many more of them will survive, and survive with disability. But that seems very odd.
Finally, there is epidemiologic difficulty in assigning value
to morbidity in surviving infants in the NICU. Verrips et al
(2008) have attempted to assess the effects of permanent
19
residual disability for NICU survivors and their immediate
families; they have demonstrated consistently that children
with disabilities and their parents place a much higher value
on their lives, and the quality of those lives, than do either
physicians or NICU nurses. The vast majority of infants
who survive the NICU, even those with significant permanent neurologic compromise, have “lives worth living,” as
judged by the people most affected by those lives.
GA-specific mortality seems to preclude resuscitation
for infants born before 22 weeks’ gestation and require
resuscitation for infants born after 27 weeks’ gestation. In
between, the outcomes are murky, prognostic indices are
imperfect, and sociologic analyses of human behaviors (of
parents and physicians) appear inadequate to develop any
uniform approach that is satisfactory.
POLICY
Neonatologists in the United States are sued for medical
negligence approximately once every 10 years, and the
average U.S. neonatologist will be sued more than once
during his or her career (Meadow et al, 1997). Other than
case-specific failures or oversights, are there any overarching themes that have arisen from medical malpractice cases
in neonatology?
In the United States most malpractice allegations are
state based, as opposed to federal cases. Consequently,
the decisions of lower state courts, or even state supreme
courts, are not binding in any other state, but they can be
informative. The first important legal case in neonatology
in the United States was Miller v HCA (2003). In 1990,
Mrs. Miller came to the Hospital Corporation of America (HCA) in Texas in labor at 23 weeks’ gestation. The
fetus was estimated to weigh 500 to 600 g. No baby born
that size had ever survived at that hospital. Mrs. Miller,
her husband, and the attending physicians agreed that the
baby was previable and that no intervention was indicated.
The baby was born, but a different physician performed
resuscitation, and the infant survived with brain damage.
As a result, the Millers sued the hospital for a breach of
informed consent, and they were awarded $50 million by
a trial jury. The case wound its way to the Texas Supreme
Court, which dismissed the verdict and articulated an
“emergency exception” for physicians—that is, if a Texas
physician finds himself or herself in the emergency position of needing to resuscitate a patient to prevent immediate death, the physician can try to perform resuscitation
without being obligated to obtain consent from anyone.
Whether it would be acceptable for a physician not to perform resuscitation in an emergency was left unarticulated
by the Texas court.
In Wisconsin, the case of Montalvo v Borkovec (2002)
took the legal obligations of neonatologists and parents
to a different place. The case derived from the resuscitation of a male infant born between 234⁄7 and 242⁄7 weeks’
gestation, weighing 679 g. The parents claimed a violation of informed consent, arguing that the decision to use
“extraordinary measures” should have been relegated to
the parents. The Wisconsin Appellate Court disagreed,
holding that “in the absence of a persistent vegetative state,
the right of a parent to withhold life-sustaining treatment
from a child does not exist.” Because virtually no infant is
20
PART I Overview
born in a persistent vegetative state, this decision would
apparently eliminate the ethical possibility in Wisconsin of
a “gray zone” of parental discretion. No other jurisdiction
in the United States has adopted this position. The Wisconsin Appellate Court, like the Texas Supreme Court,
was silent on whether physicians have discretion not to
resuscitate. However, in Texas and Wisconsin, physicians
are apparently not liable if they choose to do so.
A number of other state courts have addressed issues of
treatment or nontreatment. In general, the courts are permissive of physicians who resuscitate infants. If courts are
asked to sanction decisions to allow infants to die, most
will do so only if there is consensus among physicians
and parents, and occasionally ethics committees. Courts
are not eager to punish physicians who treat infants over
parental objections or to empower physicians to stop treatment when parents want it to continue.
BABY DOE REGULATIONS
Of course, state-by-state civil malpractice cases are not
the only administrative means for redrawing the interface
between neonatal medicine and the law. The federal government has contributed as well. The Baby Doe Regulations were one of the first attempts to codify and impose a
federal vision of appropriate ethical behavior in the NICU.
In 1982, a baby with Down syndrome and esophageal
atresia was born in Bloomington, Indiana. At that time, the
standard of care for babies with Down syndrome was shifting. A decade or two earlier, most babies with Down syndrome who survived infancy were institutionalized. If babies
needed surgery for an anomaly like Baby Doe’s atresia, the
parents were given the option. Approximately half chose
surgery and half chose palliative care (Shaw, 1973). Similarly, half of pediatricians thought that palliative care, rather
than surgery, was the better option (Todres et al, 1977). By
the early 1980s, this practice had started to change. More
parents opted for active intervention to save their babies
(Shepperdson, 1983), and more physicians believed that
withholding treatment was medical neglect (Todres et al,
1988). Baby Doe was born into this shifting cultural milieu.
Baby Doe’s parents refused to consent to surgery and
chose palliative care instead. The pediatrician alerted the
state child protection agency, which investigated the case.
At a court hearing, the parents claimed that they were following the advice of their obstetrician and not their pediatrician. The court found that parents only had to follow
the advice of a licensed physician and that, since they were
doing so, they were not neglectful. The court did not take
protective custody. The doctor and hospital appealed. The
Indiana Supreme Court refused to hear the appeal, and the
baby died after 8 days (Lantos, 1987).
These facts became publicly known and led to a national
controversy that eventually reached the Oval Office. President Reagan demanded federal action. It was difficult to
decide which action should be taken, because the federal government has no jurisdiction over child abuse and neglect; it
is the domain of the states. The federal government oversees
civil rights enforcement, however, and the Reagan administration devised a legal strategy that defined not treating
babies with Down syndrome or other congenital anomalies
as discrimination against people with disabilities, rather than
medical neglect. This strategy gave the federal government a
justification for oversight. The new regulations were implemented, and bright red signs containing federal hotline telephone numbers were posted in NICUs across the country.
These signs proclaimed that withholding treatment on the
basis of disability was a federal civil rights violation (Annas,
1984) and that federal investigative squads could review
medical records to determine whether discrimination had
taken place. These regulations were challenged and eventually struck down by the U.S. Supreme Court (Annas, 1984).
A diluted version of the original Baby Doe guidelines
was eventually incorporated into the federal Child Abuse
and Treatment Act (Annas, 1986; Kopelman, 1988). That
law, however, is primarily a funding mechanism to channel federal funds to state child protection agencies; it is
not a regulation that can be enforced for physicians or
hospitals. The Baby Doe regulations do not exist today
and have not existed since 1984; however, they still hold
symbolic power. The mention of Baby Doe strikes fear in
the hearts of pediatricians who lived through the events, in
part because pediatricians had made pediatricians into villains in the societal battle over child protection.
There is a certain irony in the controversy over Baby
Doe regulations. The original goal—to decrease the range
of cases in which withholding treatment of newborns is
permissible—did not need federal input. Progress was
already being made. Many diseases that used to be considered incompatible with life or that were seen as leading to an unacceptable quality of life were being treated
routinely. Metabolic therapies for genetic diseases (phenylketonuria [PKU], hypothyroidism, Gaucher disease)
and surgical therapies for organ dysgenesis (congenital
heart corrections, congenital diaphragmatic hernia (CDH)
repair, extra corporeal membrane oxygenation (ECMO),
dialysis) have been forcefully advanced by medical subspecialists in their corresponding fields (e.g., genetics, cardiac
surgery, general surgery). With few exceptions (hypoplastic left ventricle being one of the best recognized), these
advances have been relatively uncontroversial. Once sufficient data are gathered to demonstrate moderate efficacy,
the innovations are widely and rapidly adopted.
There is still controversy when treatments enable survival but have a high likelihood, or certainty, that survival
will be accompanied by severe neurologic impairment. As
a result, two questions must be asked. First, how severe
will the neurologic impairment be? Second, what is the
likelihood that the child will have the most severe possible
impairment? The prognostic spectrum of Down syndrome
is broad, but few infants with trisomy 21 are severely
impaired. In contrast, almost all infants with trisomy 13
or 18 either die in infancy or are left with profound neurologic impairment. The outcomes for these chromosomal
anomalies can be used to define the spectrum within which
clinical decisions are made. For babies whose outcomes are
likely to be similar to those seen in trisomy 21, it is no longer permissible to withhold life-sustaining treatment. For
babies whose outcomes are likely to be similar to babies
with trisomy 13, it is permissible to withhold or withdraw
life-sustaining treatment and offer palliative care instead.
The calculus becomes more complex in conditions associated with a wider range of outcomes, such as extreme prematurity or high myelomenigocele with hydrocephalus.
CHAPTER 3 Ethics, Data, and Policy in Newborn Intensive Care
The shift in moral standards regarding babies with
Down syndrome was not related to technology, but rather
sociology. The capacity to repair Arnold–Chiari malformation and duodenal atresia existed long before it was
applied to children with myelomeningocele and Down
syndrome. What has changed the mood of the country is
a growing recognition that disability is as much a social
construct as a medical construct, although it is always both
and not one or the other.
BORN ALIVE INFANT PROTECTION ACT
In 2002, the U.S. Congress passed a law called the Born
Alive Infant Protection Act (BAIPA). BAIPA, like the discredited Baby Doe regulations, was an attempt to insert
federal values into medical deliberations. There are some
interesting similarities and distinctions between Baby Doe
and BAIPA. The Baby Doe regulations addressed disability, whereas BAIPA applies to abortion. BAIPA declares
that “for the purposes of Federal law, the words ‘person’,
‘human being’, ‘child’, and ‘individual’ shall include every
infant who is born alive at any stage of development.” As
Sayeed (2005) noted, “The agency arguably substitutes a
nonprofessional’s presumed sagacious assessment of survivability for reasonable medical judgment.” It is unclear
what the implications of this law have been. On the one
hand, all infants born alive before BAIPA were also treated
as human beings; however, that did not necessarily mean
that they received all available life support and resuscitation. After all, patients can have do-not-resuscitate
orders or receive palliative care rather than intensive care.
Still, the purpose of BAIPA seemed to be less about the
treatment of babies and more about restrictions on abortion. Although some authors have expressed concern that
the influence of BAIPA may transform neonatal care of
infants born at, or even below, the threshold of viability,
it appears to have had little measurable effect to date. Partridge et al (2009) surveyed neonatologists in California
and found that they were concerned about the implications
of BAIPA. They write, “If this legislation were enforced,
respondents predicted more aggressive resuscitation
potentially increasing risks of disability or delayed death.”
There have been no cases to date in which BAIPA has been
invoked or in which physicians and hospitals have been
found to be in violation of its requirements.
FUTURE DIRECTIONS
There is no new technology in development that appears
likely to affect outcomes significantly. Consequently, for
infants who receive resuscitation in the delivery room,
birthweight-specific mortality and morbidity are unlikely to
change much in the near future. Nonetheless, three developments may change the way we think about newborns,
and consequently shift the terrain of neonatal bioethics.
HIGH RISK MATERNAL-FETAL
MEDICINE CENTERS
Many children’s hospitals are now developing high-risk,
maternal-fetal medicine centers. The goal of these centers is to identify fetuses at risk—particularly those with
21
congenital anomalies—and to care for those fetuses and
their mothers in centers where there is expertise in fetal
diagnosis, therapy, and neonatal care. The hope is that
such centers will allow more timely, and therefore more
effective, intervention for babies with congenital heart disease, congenital diaphragmatic hernia, or other anomalies.
The medical effectiveness of fetal centers will depend
on two distinct developments. First, on a population basis,
these centers will only be as effective as fetal screening and
diagnosis. The existence of these centers will almost certainly create an expectation and a demand for better fetal
screening. Such screening is likely to include both better
imaging and better screening tests that can be performed
on maternal blood; both will lead to earlier diagnosis of
fetal anomalies. These diagnoses will create more complex
dilemmas for perinatologists and parents who will need
to decide, in any particular case, whether to terminate
the pregnancy, offer fetal therapy, or offer either palliative care or interventions after birth. Ironically, better fetal
diagnosis may increase the likelihood of pregnancy termination, even when postnatal treatment is possible, such as
in hypoplastic left heart syndrome.
Second, the effectiveness of fetal centers will depend on
the effectiveness of fetal interventions. Perhaps surprisingly, other than in utero transfusion for Rhesus disease or
vascular ablation for twin-twin transfusion syndromes—
neither of which are particularly new and neither of which
is performed by pediatric surgeons or pediatricians—there
is little evidence that any fetal intervention has had any
effect on any neonatal outcome. This lack of demonstrated
effectiveness has, thus far, not suppressed the proliferation
of fetal intervention centers. There may be other factors,
including institutional prestige, finances, and recruitment
of “desirable” patients.
EXPANDED NEWBORN SCREENING
In recent years, the number of diseases and conditions that
can be diagnosed through newborn screening has expanded
dramatically. Such screening is under the purview of states,
rather than the federal government, and there is wide variation in the number of tests that are performed. In 1995
the average number of tests per state was five (range: zero
to eight disorders). Between 1995 and 2005 most states
added tests, so that the average number of screening tests
done by 2005 was 24 (Tarini et al, 2006). The expansion of
newborn screening raises three problems. First, even the
most accurate test has false positives. For rare conditions,
the percentage of positive tests that are false positives is
increased. Thus, the more rare conditions that are added
to a newborn screening panel, the more false positives
there will be. False positives are associated with considerable parental anxiety and can lead to potentially dangerous
and unnecessary diagnostic procedures or treatments. Second, expanded newborn screening costs money. Interestingly, the tests themselves are astoundingly inexpensive,
which is why policy makers are tempted to add more to
the panels. However, the follow-up counseling and testing
after positive tests are expensive, and without such followup the screening programs will not work. The Centers for
Disease Control and Prevention has recently expressed
concern about these costs (Centers for Disease Control
22
PART I Overview
and Prevention, 2008). Finally, there is the potential for
discrimination against patients for whom documented
heterozygous carrier status conveys no recognized medical
infirmity, but social or psychological stigma may be real.
There is little funding available to assist or counsel these
patients.
FINANCIAL CONSTRAINTS
The American Academy of Pediatrics guidelines on neonatal resuscitation suggest that resuscitation should be obligatory at 25 weeks’ gestation or greater, optional at 24 weeks’
gestation, and unusual at 23 weeks’ gestation. These recommendations are thought to reflect the best understanding of
both the ethical discussion and the epidemiologic facts surrounding NICU outcomes. The recommendations purport
to reflect the traditional paradigm that data drive policy.
However, particularly in the context of NICU survival for
infants at 23 to 25 weeks’ gestation, there is good evidence
that causation can work in the reverse direction—that is,
policy drives data. In the Netherlands, Canada, and some
parts of Oregon, survival at 25 weeks’ gestation is comparable and comparably good; more than 50% of infants born
at 25 weeks’ gestation will survive to discharge. However,
in the Netherlands, virtually no infant survives birth at 24
weeks’ gestation, whereas in Canada and Oregon the survival rate is 40%. In addition, in Oregon and some parts
of Canada, survival at 23 weeks’ gestation is close to zero,
whereas in other parts of Canada and the United States the
survival rate is 20% or greater. Why? Certainly not because
the Dutch, Canadians, or Oregonians have forgotten how
to resuscitate small infants. Rather, they have chosen not
to. Why not? Perhaps it is non-maleficence–the fear that
survival with permanent neurologic morbidity may be cruel
to the child, the family, or society at large. However, the
incidence of neurologic morbidity in survivors is not different when comparing infants at 23, 24, 25, and 26 to 27
weeks’ gestation. If the fear of a permanent crippling neurologic injury is the driving force, we should not be resuscitating 26 or 27 weekers, since many more of them will
survive, and survive with disability. But that seems odd.
The approach in the Netherlands is consistent; there is
a limited budget and a communitarian ethic. There is a
certain rationale behind spending money on all pregnant
women, instead of 1% of micro-premies. The United
States appears ambivalent–we value individuals over community, are fascinated with high-technology, and claim to
prize our children. On the other hand, we will not spend
money to prevent unwanted teen pregnancy or to provide
visiting nurses for new mothers.
Finally the concept of generational conflict must
be considered. We appear quite comfortable calling
delivery-room resuscitation of 24 weekers “optional,”
based on gestational age alone. It is difficult to imagine the
AMA recommending that resuscitation for 85-year-olds
who come to the emergency department is “optional,”
based on age alone. NICU care is often criticized as “too
expensive.” We ask, “Compared to what?”
Fewer than 5% of infants will be in the NICU for more
than a short stay. The vast majority of these patients will
survive. Only 24,000 infants of 4 million births die each
year in the United States, and half of these will die within
fewer than 7 days. In contrast, nearly 3 million adults will
die in the United States each year and one third of these
will have been admitted to an medical intensive care unit
(MICU) in the 6 months before dying. MICU costs outpace NICU costs by at least 100 to 1.
SUMMARY
Ethical philosophy is a place to start, not a place to finish.
Data are relatively easy to acquire and agree on. Policy
is intriguingly insensitive to data, but that may reflect
social and political realities that exist beyond the NICU—
perceptions of disability, abortion politics, individual versus communitarian emphasis, fascination with technology,
discrimination, publicity, financial constraints—so that an
ethical course of action in one country, one city, or one
family might seem perverse elsewhere.
SUGGESTED READINGS
Annas GJ: The Baby Doe regulations: governmental intervention in neonatal rescue medicine, Am J Public Health 74:618-620, 1984.
Born-Alive Infants Protection Act of 2001: Report together with additional and
dissenting views of the House Committee on the Judiciary, 107th Congress,
1st Session, August 2, 2001. 1-38, 3. (Purpose and Summary).
Centers for Disease Control and Prevention: Impact of expanded newborn screening: United States, 2006, MMWR Morb Mortal Wkly Rep 57:1012-1015, 2008.
Janvier A, Leblanc I, Barrington KJ: The best-interest standard is not applied for
neonatal resuscitation decisions, Pediatrics 121:963-969, 2008.
Johnson S, Fawke J, Hennessy E, et al: Neurodevelopmental disability through
11 years of age in children born before 26 weeks of gestation, Pediatrics
124:E249-E257.
Lantos J: Baby Doe five years later: implications for child health, N Engl J Med
317:444-447, 1987.
Meadow W, Lagatta J, Andrews B, et al: Just in time: ethical implications of serial
predictions of death and morbidity for ventilated premature infants, Pediatrics
121:732-740, 2008.
Miller v HCA, Inc, 118 S.W. 3d 758, 771 (Texas 2003).
Montalvo v Borkovec, 647 N.W. 2d 413 (Wis. App. 2002).
Shepperdson B: Abortion and euthanasia of Down’s syndrome children: the parents’ view, J Med Ethics 9:152-157, 1983.
Todres ID, Guillemin J, Grodin MA, et al: Life-saving therapy for newborns:
a questionnaire survey in the state of Massachusetts, Pediatrics 81:643-649,
1988.
Tyson JE, Parikh NA, Langer J, et al: National Institute of Child Health and
Human Development Neonatal Research Network. Intensive care for extreme
prematurity: moving beyond gestational age, N Engl J Med 358:1672-1681,
2008.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
4
Global Neonatal Health
Linda L. Wright
CHILD MORTALITY
In 2000, the 192 United Nations member states and many
international partners adopted the Millennium Development Goals (MDGs) in response to morbidity and mortality rates in the developing world that were alarmingly
high. These eight international development goals were to
eradicate extreme hunger and poverty; achieve universal
primary education; promote gender equality and empower
women; reduce child mortality; improve maternal health;
combat HIV/AIDS, malaria, and other diseases; ensure
environmental sustainability; and develop a global partnership for development. The agreement included 18 specific
targets and 48 technical indicators to measure progress toward the MDGs between 1990 and 2015 (United
Nations General Assembly, 2001).
Although significant progress has been made toward
achieving many of the MDGs, progress has been uneven,
with huge disparities across the goals and among countries. This disparity is especially true for MDG 5 (i.e.,
improving indicators of maternal health, including maternal mortality during pregnancy or the 42 days following
the end of pregnancy, per 100,000 deliveries) and MDG 4
(i.e., reducing child mortality, expressed as deaths before 5
years per 1000 live births). Regions and countries that have
the highest maternal mortality rates also have the highest
child mortality rates (Table 4-1).
Twenty percent of all deaths in the world are child deaths
(Save the Children, 2007; United Nations Children’s
Fund, 2007), and greater than 99% of deaths occurring
in children aged 5 years or younger are in the developing
world. Of the 136 million babies born in 2007, an estimated 9.7 million died before the age of 5 years (approximately 26,000 per day) (Save the Children, 2007; United
Nations Children’s Fund, 2007). This staggering number
equals approximately half of all U.S. children younger
than 5 years (Save the Children, 2007; U.S. Census
Bureau’s American Community Survey, 2007). The latest
estimates suggest that 8.8 million children died in 2008
(United Nations Children’s Fund, 2008b). The rate of
decline in mortality in children younger than 5 years is
grossly insufficient to meet the MDG 4 goal by 2015, particularly in Sub-Saharan Africa and South Asia (United
Nations Children’s Fund, 2009). At the current rate, the
target of fewer than 5 million annual child deaths will not
be met until 2045. To meet the goal of fewer than 5 million
child deaths in 2015, deaths in children younger than 5
years must be cut in half between 2008 and 2015 (Table
4-2) (United Nations Children’s Fund, 2007, 2009a).
The regions with the highest numbers of child deaths
are Sub-Saharan Africa (which has high fertility rates and
the highest child mortality rates [144 deaths per 1000 live
births], and 4.8 million children [1 in 7] dies before the
age of 5 years) and South Asia (3.1 million deaths [1 in
13] before the age of 5 years) (United Nations Children’s
Fund, 2007). Sub-Saharan Africa accounts for 51% of all
deaths among children younger than 5 years, followed by
Asia with 42% (You et al, 2009).
In 2008, 75% of deaths in children younger than 5
years occurred in only 18 countries, and 40% occurred in
only three countries: India, Nigeria, and the Democratic
Republic of the Congo. Of the 34 countries with mortality rates exceeding 100 per 1000 live births in 2008, all
were in Sub-Saharan Africa, except for Afghanistan (You
et al, 2009). Equally troubling, only 10 of the 67 countries with high mortality rates (at least 40 per 1000 live
births) were on track to meet MDG 4 before the 2009
economic crisis (You et al, 2009). However, a number of
relatively poor countries with low gross national incomes
have made considerable progress in improving survival, including Malawi, Tanzania, Madagascar, Nepal,
Bangladesh (Save the Children, 2007), Eretria, the Lao
People’s Democratic Republic, Mongolia, and Bolivia
(Figure 4-1) (You et al, 2009).
More than 70% of deaths in children younger than 5 years
are caused by newborn problems, pneumonia, and diarrhea.
Pneumonia kills more children than any other illness—
more than HIV, malaria, and measles combined (Figure
4-2). Pneumonia results in death for more than 2 million
children younger than 5 years each year, or approximately
20% of child deaths worldwide. More than 95% of all new
pneumonia cases, representing an estimated 150 million
episodes of pneumonia annually, occur in children younger
than 5 years in developing countries. Sub-Saharan Africa
and South Asia together have more than half the total number of pneumonia cases. Effective prevention strategies
include immunization against measles, whooping cough,
Haemophilus influenzae type b, and Streptococcus pneumoniae;
exclusive breastfeeding and improved nutrition or low
birthweight; zinc supplementation; reducing indoor air
pollution; and prevention and management of HIV infection (Qazi et al, 2008); however, the protection afforded by
immunizations will not prevent neonatal pneumonia. Universal use of simple standardized management guidelines
for identifying and treating pneumonia in communities
and primary health care centers, with the World Health
Organization (WHO) Integrated Management of Childhood Illness (Niessen et al, 2009), may reduce the child
pneumonia fatality rate (Niessen et al, 2009). Sazawal and
Black (1992) suggested that community-based acute respiratory infection case management might reduce mortality
by more than 20% in children younger than 4 years. Failing
prevention, prompt diagnosis and treatment are necessary
to improve pneumonia mortality and morbidity; however,
prompt diagnosis and effective treatment of pneumonia
and hypoxemia are often not available. Radiology, laboratory tests, and pulse oximetry, which can predict response
to antibiotic therapy in cases of severe pneumonia (Fu
et al, 2006), are not available in most first-level (i.e., rural)
23
24
PART I Overview
TABLE 4-1 Countries With the Highest Numbers of Child Deaths Also Have High Rates of Maternal Death
Country
Ranking for Number
of Child Deaths
Number of Child Deaths
Ranking for Number
of Maternal Deaths
Number of Maternal Deaths
India
1
1,919,000
1
136,000
Nigeria
2
1,043,000
2
37,000
DR Congo
3
589,000
4
24,000
Ethiopia
4
509,000
4
24,000
Pakistan
5
473,000
3
26,000
China
6
467,000
9
11,000
Afghanistan
7
370,000
7
20,000
Bangladesh
8
274,000
8
16,000
Uganda
9
200,000
12
10,000
Angola
10
199,000
9
11,000
6,043,000 child deaths
Approximately 60 percent of global total
315,000 maternal deaths
Approximately 60 percent of global total
Sources: Child deaths UNICEF. State of the World’s Children 2007, Table 1; Maternal deaths: World Health Organization, United Nations Children’s Fund and United Nations
Population Fund, Maternal Mortality in 2000: Estimates Developed by WHO, UNICEF and UNFPA.
From Save the Children: State of the world’s mothers, 2007: saving the lives of children under 5.
TABLE 4-2 Global Progress in Reducing Child Mortality is Insufficient to Reach Millennium Development Goal 4*
Average annual rate of reduction (AARR) in the under 5 mortality rate (U5MR) observed for 1990–2006 and required during
2007–2015 in order to reach MDG 4
U5MR
AARR
No. of Deaths per 1,000
Live Births
1990
2006
Observed %
Required %
1990–2006
2007–2015
Progress Toward the
MDG Target
Sub-Saharan Africa
187
160
1.0
10.5
Insufficient progress
Eastern and Southern Africa
165
131
1.4
9.6
Insufficient progress
West and Central Africa
208
186
0.7
11.0
79
46
3.4
6.2
Middle East and North Africa
South Asia
No progress
Insufficient progress
123
83
2.5
7.8
Insufficient progress
East Asia and Pacific
55
29
4.0
5.1
On track
Latin America and Caribbean
55
27
4.4
4.3
On track
CEE/CIS
53
27
4.2
4.7
On track
Industrialized countries/
territories
10
6
3.2
6.6
On track
103
79
1.7
9.3
Insufficient progress
93
72
1.6
9.4
Insufficient progress
Developing countries/
territorries
World
United Nations Children’s Fund: The state of the world’s children 2008: child survival, 2007, New York.
*Progress towards MDG 4, with countries classified according to the following thresholds:
On track: U5MR is less than 40, or U5MR is 40 or more and the average annual rate of reduction (AARR) in under 5 mortality rate observed from 1990 to 2006 is 4.0% or more.
Insufficient progress: U5MR is 40 or more and AARR observed for the 1990-2006 period is between 1.0% and 3.9%.
No progress: U5MR is 40 or more and AARR observed for 1990-2006 is less than 1.0%.
Source: UNICEF estimates based on the work of the interagency Child Mortality Estimation Group.
hospitals, despite high mortality rates in children who
have hypoxemia or HIV. Randomized controlled trials
of parenteral antibiotic treatment in hospitals compared
with home-based treatment have demonstrated the safety
and efficacy of treating pneumonia with oral antibiotics
outside of a hospital setting in older children. New evidence regarding home treatment of severe pneumonia
is changing concepts about the need for hospitalization.
The first randomized trial to compare outcomes of hospital treatment of severe pneumonia, without underlying
complications, with home-based oral antibiotics in Pakistan demonstrated that home-based antibiotics are safe
and effective. Of 2037 children with severe pneumonia
aged 3 to 59 months, randomized to either parenteral
ampicillin for 48 hours followed by 3 days of oral ampicillin or home-based oral amoxicillin for 5 days, there were
equal numbers of failures in the hospitalized group (8.6%)
and in the ambulatory group (7.5%) by day 6. Just 0.2%
children died within 14 days of enrollment and none of
the deaths were considered to be associated with treatment
CHAPTER 4 Global Neonatal Health
25
POVERTY DOES NOT HAVE TO BE A DEATH SENTENCE
FOR CHILDREN UNDER 5
Above-average GNI per capita
Above-average reduction in under-5 mortality
Percent reduction in under-5
mortality (1990–2005)
Below-average GNI per capita
Above-average reduction in under-5 mortality
Bangladesh
Nepal
Malawi
Madagascar
Tanzania
Cameroon
Cambodia
Côte d’Ivoire
Below-average GNI per capita
Below-average reduction in under-5 mortality
South Africa
Equatorial Guinea
Above-average GNI per capita
Below-average reduction in under-5 mortality
GNI per capita (purchasing power parity)
Priority countries for under-5 child survival
A for effort
Failing to save children
FIGURE 4-1 Poverty does not have to be a death sentence for children under 5. (From Save the Children: State of the world’s mothers 2007:
saving the lives of children under 5, 2007.)
WHY CHILDREN DIE
Newborn disorders, pneumonia, and diarrhea account for
73% of all deaths among children under 5
Pneumonia 19%
Newborn
causes 37%
Diarrhea 17%
Preterm 28%
Sepsis/pneumonia 26%
Asphyxia 23%
Congenital 8%
Tetanus 7%
Diarrhea 3%
Other 7%
Malaria 8%
Measles 4%
AIDS 3%
Injuries 3%
Other 10%
Undernutrition is an underlying cause
of 53% of deaths among children
under age 5
FIGURE 4-2 Why children die. (From Save the Children: State of the
world’s mothers 2007: saving the lives of children under 5, 2007.)
allocation (Hazir et al, 2008). A small percentage (approximately 2% to 3%) of severely ill children will still require
early community detection and transport to a hospital for
evaluation of hypoxia, infection, pneumonia, malaria, and
parenteral antibiotics with or without oxygen (Mwaniki
et al, 2009). The WHO has spearheaded efforts to reduce
the global burden of child pneumonia with the Integrated
Management of Childhood Illness, a Global Action Plan
for Control and Prevention of Pneumonia (GAPP) (Qazi
et al, 2008), publication of new comprehensive global,
regional, and national disease burden statistics for pneumococcal and hepatitis b disease, the Global Coalition
against Childhood Pneumonia, and the first World Pneumonia Day on November 2, 2009 (United Nations Children’s Fund, 2009b).
Diarrheal diseases are the second most common cause
of child deaths globally. Worldwide, diarrhea accounts
for 18% of deaths among children younger than 5 years,
or an estimated 1.7 million child deaths every year, and
it accounts for 10% to 80% of growth retardation in the
first few years of life (Baqui and Ahmed, 2006). Exclusive
breastfeeding provided significant protection from diarrheal diseases before the WHO-recommended introduction of complementary feeds at 6 months. Children in
poverty are especially prone to diarrheal diseases after the
introduction of complementary feeding, because diarrhea
is spread by poor hygiene and sanitation facilities; contaminated water, formula, food, or utensils; low rates of
vitamin A supplementation; low zinc intake; and limited
access to rotavirus immunization. Most diarrhea-related
deaths in children are due to dehydration. The WHO
recommendations for oral rehydration therapy (ORT) for
childhood diarrheal diseases have changed. Research has
demonstrated that homemade fluids that contain lower
concentrations of sodium and glucose, sucrose, or other
carbohydrates (e.g., cereal-based solution) can be as effective as ORT. Current recommendations include increased
fluid intake and continued feeding, as well as the use of
zinc and low-osmolarity ORT to prevent and treat diarrheal episodes (Fischer-Walker et al, 2009; World Health
Organization and United Nations Children’s Fund, 2004).
Lower-osmolarity ORT reduces stool output, vomiting,
and unscheduled intravenous therapy (Baqui and Ahmed,
2006). In Sub-Saharan Africa there has been little progress
in diarrhea prevention and treatment in the last decade—
the percentage of children younger than 5 years who
received the recommended treatment increased from 32%
in 2000 to only 38% in 2008 (United Nations Children’s
Fund, 2009). Progress on case management of childhood
26
PART I Overview
pneumonia, diarrhea, and malaria will depend on strengthening the integrated community-based prevention and
treatment of these pervasive childhood diseases within the
health system (United Nations Children’s Fund, 2009).
Undernutrition is another major factor in mortality of
children younger than 5 years; it is responsible for 7% of
the total disease burden in any age group, making it the
highest of any risk factor for overall global burden of disease (Black et al, 2008b). Undernutrition is a contributing factor in more than half of infectious disease deaths
in children (Save the Children, 2007) and is the underlying cause of more than one third of all deaths among
children younger than 5 years (United Nations Children’s
Fund, 2008a). The effects of undernutrition reach beyond
the individual child. Maternal or child undernutrition is
a complex intergenerational problem that includes intrauterine growth restriction, severe wasting, and stunting.
Virtually no progress has been made toward overcoming
undernutrition in the last 25 years (United Nations Children’s Fund, 2008a). Wasting (weight-for-height Z score
less than –2) is associated with acute weight loss. Of the
estimated 55 million children younger than 5 years with
wasting (10% of all children), more than half are in south
central Asia or Sub-Saharan Africa; 19 million children
have severe acute malnutrition or wasting (weight-forheight Z score less than –3) and are in need of urgent
therapeutic feeding (United Nations Children’s Fund,
2008a). Stunting (height-for-age Z score less than –2),
which is more common, indicates chronic restriction of
a child’s potential growth. An estimated 178 million children younger than 5 years have stunting—almost one third
of children in low- to middle-resource settings. Ninety
percent of them (160 million) live in just 36 countries and
represent almost half of the children in those countries.
India’s 61 million children with stunting represent more
than half of all Indian children younger than 5 years and
34% of all children with stunting worldwide (Bhutta et al,
2008a). This urgent problem must be solved, because the
period between birth and 24 months old is critical. If children do not grow appropriately before 2 years old, they are
more likely to be short as adults, have lower educational
achievement and economic productivity, and give birth to
smaller infants who repeat the cycle in the next generation.
Although the problem of undernutrition has been overshadowed by concerns over obesity, there is no evidence
that rapid weight or linear growth in the first 2 years of life
increases the risk of chronic disease in adults (fetal origin
of adult disease), even in children with poor fetal growth
(Victora et al, 2008).
A recent review of interventions that affect maternal
and child undernutrition suggested that counseling about
breastfeeding and supplementation with vitamin A and
increased zinc intake have the greatest potential to reduce
the burden of child morbidity and mortality (Bhutta et al,
2008a). The promotion of breastfeeding has had an effect
on the improved survival of infants and young children,
but its effect on stunting has been negligible. Among populations with inadequate food, food supplements are beneficial with or without educational interventions (increased
height-for-age Z scores by 0.41; 95% confidence interval
[CI], 0.05-0.76) and can reduce stunting and the related
burden of disease. In populations with sufficient food,
complementary feeding education increased height-forage Z scores by 0.25 (95% CI, 0.01-0.49) compared with
controls. Facility management of severe acute malnutrition,
according to WHO guidelines, reduced the case fatality
rate by 55% (relative risk [RR], 0.45; 95% CI, 0.32-0.62),
and observational data suggest that ready-to-use prepared
foods in treatment of severe malnutrition may be effective in community settings as well. Recommended micronutrient interventions for children include strategies for
vitamin A supplementation, zinc supplements to prevent
and treat diarrhea and lower respiratory tract infections,
iron supplements in areas where malaria is not endemic,
and universal promotion of iodized salt (Bhutta et al,
2008a). A subsequent metaanalysis of neonatal vitamin A
supplementation concluded, on the basis of six trials in the
developing world, that there was no evidence for a reduced
risk of mortality and morbidity during infancy and thus
no justification for neonatal vitamin A supplementation
as a public health measure to reduce infant mortality and
morbidity in developing countries (Gogia and Sachdev,
2009). The efficacy of zinc supplementation in reducing
overall mortality in neonates has been questioned as well
(Sazawal et al, 2007). Existing interventions designed to
improve nutrition and prevent related disease may reduce
stunting at 36 months old by as much as 36% and mortality between birth and 36 months old by approximately
25%. However, elimination of stunting will require longterm investment in improved nutrition and interventions
to improve early childhood education, maternal education,
and women’s economic status (Black et al, 2008a). Growth
(length, height, and weight) should be monitored routinely and regularly in view of its importance as a marker
for undernutrition and stunting (Victora et al, 2009).
The nutrition of the children of India and Sub-Saharan
Africa are of greatest concern. India is a concern because
it represents 34% of the world’s children with stunting,
who often start life with intrauterine growth retardation
and as adults are members of families with intergenerational stunting that live on less than $2 per day. In Africa,
neonates are born with normal birthweight, but develop
stunting and wasting because of poverty and civil unrest.
Although exclusive breastfeeding protects the neonate,
providing adequate amounts of nourishing food in early
infancy is critical to the future of children from these two
continents. Among the areas needing further research are
assessments of the effectiveness and cost-effectiveness of a
national health system’s nutritional interventions on stunting rates and weight gain; long-term effects of maternal
nutritional interventions on maternal and child health and
cognitive outcome; research on the reversibility of stunting and cognitive impairment in children aged 36 to 60
months; studies of community-based prevention and treatment strategies for severe acute malnutrition; and studies
of the effectiveness of various zinc delivery strategies.
EPIDEMIOLOGY OF NEONATAL
MORTALITY (BEFORE 28 DAYS)
Developed countries have experienced significant declines
in child mortality in the last 30 years, but only 1% of the
world’s neonatal deaths occur in 39 high-income countries
whose average neonatal mortality rates are 4 to 6 per 1000
27
CHAPTER 4 Global Neonatal Health
TABLE 4-3 Regional or Country Variations in Neonatal Mortality Rates and Numbers of Neonatal Deaths, Showing the Proportion of
Deaths in Children Younger than Age 5 Years
NMR per 1000
Livebirths (range
across countries)
Number (%)
of Neonatal
Deaths (1000s)
Percentage of Deaths in
hildren Aged Younger than
C
5 Years in the Neonatal Period
Percentage Change
in NMR between 1996
and 2005 Estimates*
4 (1–11)
33 (2–70)
42 (1%)
3956 (99%)
63%
38%
–29%
–8%
Africa
44 (9–70)
1128 (28%)
24%
5%
Americas
12 (4–34)
195 (5%)
48%
–40%
Eastern Mediterranean
40 (4–63)
603 (15%)
40%
–9%
Europe
11 (2–38)
116 (3%)
49%
–18%
Southeast Asia
38 (11–43)
1443 (36%)
50%
–21%
Western Pacific
19 (1–40)
512 (13%)
56%
–39%
30 (1–70)
3998 (100%)
38%
–16%
Income groups
High-income countries†
Low-income and middleincome countries
WHO regions
Overall
From Lawn JE, Cousens S, Zupan J: 4 Million neonatal deaths: when? where? why? Lancet 365:891-900, 2005.
*The data inputs cover at least a 5-year period before each set of estimates. Period of change may be assumed to be up to 15 years.
†Thirty-nine countries with NMR data of 54 countries with gross national income per person of US$9386.10.
live births. In contrast, little progress has been made in
reducing maternal and neonatal deaths in the developing
world, where the disparity is large and growing (Lawn
et al, 2005) and the average neonatal mortality rate (deaths
in the first 28 days of life per 1000 live births) in 2000 was
33, with a range of 2 to 70. Together Africa and Southeast Asia account for approximately two thirds of neonatal
deaths (Table 4-3) (Lawn et al, 2005).
More than half of all maternal and newborn deaths occur
at birth or in the first few days after birth, when health coverage is the lowest. Cultural norms, financial constraints,
and small fetuses that are considered nonviable also limit
the reporting of neonatal deaths. As a consequence, the
births of an estimated 51 million children per year go unrecorded in any formal registration system (United Nations
Children’s Fund, 2007). These children are often born in
slums or rural poverty to very young or older mothers who
lack access to education and basic health and reproductive
services, and who also live in countries that have experienced recent political unrest (United Nations Children’s
Fund, 2007). As a result, fewer than 3% of neonatal deaths
occur in countries that have high-coverage vital registration data or recent, reliable data on causes of neonatal death; therefore global analysis is based on estimates
(Lawn et al, 2005) derived from statistical modeling. Until
the middle to late 1990s, estimates of neonatal deaths
were drawn from historical data. More rigorous estimates
using demographic and health surveys of newborn deaths
at a national level were available in 1995 and 2000 (Lawn
et al, 2005; United Nations Children’s Fund, 2008a),
which produced more reliable neonatal mortality rates.
The lack of reliable data from most high-risk countries,
and the complex methods for developing estimates and for
estimating uncertainty, makes even these improved neonatal mortality data inherently uncertain (United Nations
Children’s Fund, 2008a). As a result, data from different
sources are difficult to compare and interpret; this will be
especially true when comparing data from before and after
2008, when the Inter-agency Group for Child Mortality
Estimation (IGME) incorporated a substantial amount of
new data and developed a new method to adjust mortality
related to HIV/AIDS (You et al, 2009).
The latest available data suggest that 3.7 million deaths
(40% of deaths in children younger than 5 years) occur
in the first month and that there are almost as many stillbirths per year worldwide (3.3 million) (Stanton et al,
2006; United Nations Children’s Fund, 2008a; World
Health Organization, 2006). The first hours and days of
a baby’s life are the most critical (Figure 4-3). Every year
2 million babies die on the day they are born (Save the
Children, 2007), representing almost 50% of all neonatal
deaths, and an astounding 75% of neonatal deaths occur
within the first 7 days of life (Murray et al, 2007; Save the
Children, 2007).
As a result, a neonate is approximately 500-fold more
likely to die in the first day of life than at 1 month of age
(United Nations Children’s Fund, 2007). Without a major
reduction in early (7 days) deaths in high-mortality countries, it will be impossible to meet the MDG 4 (Figure 4-4)
(World Health Organization, 2006).
MATERNAL RISK FACTORS
FOR NEONATAL DEATH
Because 40% to 90% of women in low-resource settings deliver their baby in the home without a skilled
birth attendant or access to facility care for themselves or
their newborn, intrapartum complications put the fetus
or neonate at increased risk for death, especially maternal bleeding after the eighth month, hypertensive disorders, obstructed labor, prolonged second-stage of labor or
malpresentation, maternal fever or rupture of membranes
for longer than 24 hours, multiparity, malaria or syphilis,
meconium staining, and maternal HIV (Lawn et al, 2005).
28
PART I Overview
Up to 50%
of newborn deaths are in
the first 24 hours –
2 million deaths
per year
Daily risk of death (per 1,000 survivors)
IN THE FIRST MONTH OF LIFE,THE FIRST DAY AND WEEK ARE MOST RISKY
9
8
7
6
5
4
3
2
1
0
0
7
14
21
Days of life
A child’s risk of dying on the first day of life is about 500 times
greater than their risk of dying when they are 1 month old.
The first hours and days of a baby’s life are critical.
75% of
newborn deaths are in
the first week –
3 million deaths
per year
Mortality rate per 1000 live births
FIGURE 4-3 In the first month of life, the first day and week pose the highest risk. (Modified from ORC Macro: Measure Demographic Health Survey
STAT compiler, 2006. Available at www.measuredhs.com. Accessed April 6, 2006. Based on the analysis by J. Lawn of 38 DHS datasets [2000 to 2004] with
9022 neonatal deaths.)
250
200
150
Late neonatal mortality
Early neonatal mortality
Global mean under-5
mortality rate
Global MDG 4 target
100
50
0
68
28
32
1960
1970
1980
1990
2000
2010
FIGURE 4-4 Progress toward Millennium Development Goal 4 for
child survival showing the increasing proportion of deaths in children
younger than 5 years. (Modified from Lawn JE, Kerber K, EnweronuLaryea C, et al: Newborn survival in low resource settings: are we delivering?
BJOG 116[Suppl 1]:49-59, 2009.)
STILLBIRTHS
Current estimates attribute at least 3.2 million deaths
worldwide to stillbirth—defined as having no signs of life
at delivery in a fetus at 28 weeks’ gestation or greater—
ranging from 5 per 1000 in wealthy countries to 32 per
1000 in South Asia and sub-Saharan Africa (Lawn et al,
2009c; Stanton et al, 2006); however, stillbirths are largely
unrecorded and uncounted. Although differentiating a
macerated stillbirth from a recent stillbirth seems simple,
differentiating a stillbirth from an early neonatal death is
challenging, especially in community settings with weak
health systems and limited access to facility care. The task
requires training in monitoring fetal heart rate and early
signs of life, prompt and appropriate resuscitation, and
emergency caesarian section, as required (McClure et al,
2007). Lack of training of birth attendants in resuscitation
and careful assessment for signs of life, variability in what
is considered the lower limit of viability, gender bias, and
the influence of financial or other burdens with an assignment can result in the misclassification of early deaths as
stillbirths. Cultural norms that discourage the weighing of
dead infants and dictate prompt burial may serve to perpetuate misclassification. Stillbirth is commonly defined as
fetal death within the last 12 weeks of pregnancy (i.e., at
least 28 weeks’ gestation or weighing 1000 g); however,
some community and national standards define stillbirth
as fetal death after 22 weeks’ gestation or weighing 500
g. Because most of these deaths occur in settings where
women have limited access to skilled birth attendants, it is
likely that the current estimates of stillbirth numbers are
too low and that a cause of death may not be established
in the majority of stillbirths that occur in the developing
world. Better data on the number and causes of stillbirth
are urgently needed to prioritize action to reduce avoidable stillbirths in high-mortality settings, where rates are
at least 10-fold higher than in wealthy countries (Stanton
et al, 2006). Stillbirths are included in the mortality tables
of the 2009 Global Burden of Disease for the first time (Lawn
et al, 2009c).
DIRECT CAUSES OF NEONATAL
DEATH
The proportion of neonatal deaths among children
younger than 5 years varies, but the absolute number of
neonatal deaths is determined by the size of the population and the neonatal mortality rate (NMR). Africa and
Southeast Asia represent approximately two thirds of neonatal deaths, with the largest number of newborn deaths in
South Asia and the highest rates of neonatal mortality in
Sub-Saharan Africa, similar to child deaths. India contributes 25% of the world’s neonatal deaths (United Nations
Children’s Fund, 2007).
Three causes of neonatal death are responsible for
approximately 75% of neonatal deaths: prematurity
(28%), sepsis and pneumonia (26%), and asphyxia (23%)
(Save the Children, 2007). Prematurity (birth before 37
weeks’ gestation) and asphyxia (failure to initiate and sustain spontaneous respiration) can result in long-term neurologic injury and cognitive impairment among survivors
CHAPTER 4 Global Neonatal Health
in countries and families that are least able to provide
appropriate care. For every newborn that dies, another 20
suffer birth injury, preterm birth complications, or other
neonatal conditions (United Nations Children’s Fund,
2007). Determining cause-specific perinatal and neonatal
mortality would allow the development of focused interventions and evaluation of their effects on perinatal and
neonatal survival. However, establishing a primary cause
of stillbirth and neonatal death in the 50% (2 million) of
neonates that die in the first day—often in homes without access to health care systems, skilled birth attendants,
or diagnostic techniques—is extremely challenging. To
overcome these problems, verbal autopsy techniques have
been developed to assign a cause of death in such cases,
usually based on an interview with the infant’s mother or
caretaker within 6 months of the child’s death. The cause
of death is assigned by a panel of physicians or compared
to a reference standard from prospective hospital-assigned
causes of death. The diagnostic accuracy of verbal autopsy
techniques to establish neonatal cause of death is limited
because of the lack of standardization of case definitions,
cause of death classifications, methods of assigning cause
of death, and the limited generalizability of hospital reference standards to infants who die in the community
(Edmond et al, 2008; World Health Organization, Development of Verbal Autopsy Standards). In the absence of
critical vital registry data (Setel et al, 2007), global estimates of the causes of neonatal deaths are possible only
through statistical modeling.
The cause-specific distribution of neonatal deaths correlates with the NMR. In high-mortality settings (>45
per 1000 live births), the risk of neonatal death because
of severe sepsis and pneumonia is approximately 11-fold
higher, and the risk of dying because of birth asphyxia is
approximately eightfold higher than the risk in low-mortality countries (<15 per 1000 live births). The proportion
of deaths as a result of prematurity drops in countries with
a high NMR because of the deaths due to infection; however, the risk of death attributable to the complications of
prematurity is still threefold higher than in low-mortality
countries (Lawn et al, 2005). In addition to significant differences in cause-of-death distribution between countries,
there is often substantial variation within countries (Lawn
et al, 2005), especially between urban and rural areas.
CONSEQUENCES OF PRETERM
DELIVERY AND LOW BIRTHWEIGHT
Prematurity and low birthweight are often not distinguished because of the lack of gestational age dating and
failure to weigh all babies at birth. As many as 18 million babies worldwide may be born annually at a low
birthweight (<2500 g). Although low birthweight affects
approximately 15% of births, preterm delivery and low
birthweight may account for up to 60% to 80% of neonatal deaths (Awasthi et al, 2006). South Asia contributes
50% of the world’s babies with low birthweight, because
29% of babies have a low birthweight. In contrast, only
14% of Sub-Saharan African babies have a low birthweight
(United Nations Children’s Fund, 2007).
In developed countries, preterm birth is the leading
cause of morbidity and mortality, and the percentage of
29
deaths attributed to preterm delivery is more than twofold
the percentage of deaths due to asphyxia, sepsis and pneumonia, and congenital anomalies (death from diarrhea
does not occur) (Lawn et al, 2005). The current rate of
12.5% in the United States represents an increase of more
than 30% since 1981 and is almost double the 5% to 9%
frequency in other developed countries. The highest rates
of preterm birth in the United States occur among racial
and ethnic minorities and in older women who conceive
by artificial reproductive technology (Preterm birth: crisis
and opportunity, 2006). Whether the number of preterm
deliveries is rising in the developing world is unknown.
Despite 30 years of research, little is known about the
etiology and prevention of preterm delivery, but improved
management in neonatal intensive care units has increased
the survival of extremely immature fetuses in the developed world. Complex technology is not required to prevent deaths attributable to prematurity and birth asphyxia
in the developing world. Low-cost interventions, including
resuscitation training (Deorari et al, 2001), nutritional and
thermal support through kangaroo-mother care (Charpak
et al, 2005), and exclusive breastfeeding may reduce deaths
attributable to asphyxia and preterm delivery after 34
weeks’ gestation. The WHO has developed an Essential
Newborn Care package, which includes clean delivery
practices, neonatal delivery care (including prompt stimulation and bag-mask resuscitation as required), thermoregulation with skin-to-skin care, early initiation of exclusive
breastfeeding, care of the moderately small baby at home,
and recognition of common illnesses (World Health Organization and Department of Reproductive Health and
Research, 1996). The addition of neonates to the WHO–
United Nations Children’s Fund Integrated Management
of Childhood Illnesses package, which has been adopted by
India, represents a significant resource to improved neonatal survival. The package provides skill-based training with
elements similar to the Essential Newborn Care package,
but it adds immunization and several postpartum home
visits by health workers to help mothers recognize and
manage minor conditions and refer severe cases in a timely
manner (World Health Organization, 2003a). The Global
Alliance to Prevent Prematurity and Stillbirth (GAPPS)
has recently been launched to address stillbirth and prematurity with a comprehensive review of published and
unpublished data on preterm birth and stillbirth research
and interventions. In collaboration with diverse global
partners in science, public health, and policy, GAPPS
plans to advance research on the etiology of preterm delivery and stillbirth, accelerate delivery of low-cost effective
interventions, and raise awareness of the effects of prematurity and stillbirth (www.gapps.org).
NEONATAL SEPSIS AND PNEUMONIA
Neonatal infections are responsible for more than 1 million
of the 3.7 million annual deaths in the developing world.
More than 95% of all deaths from birth to 2 months of age
occur in developing countries. Risk factors include chorioamnionitis, low birthweight, unhygienic delivery, skin
care, cord care, and environments. Ideally, simple preventive strategies such as clean delivery kits, hand washing,
and cord care would be effective, but such data are not
30
PART I Overview
available, and the effectiveness of using chlorhexidine to
prevent community-acquired neonatal sepsis and mortality is still unsettled (Cutland et al, 2009; Mullany et al,
2006). The quality and quantity of data on neonatal deaths
caused by infections in the developing world are extremely
limited. The estimated total is at least 1.6 million annual
deaths (26% for sepsis and pneumonia, excluding tetanus
and diarrhea) (Lawn et al, 2005). The majority of births
and deaths are thought to occur at home without coming
to medical attention (Lawn et al, 2004), because of cultural
norms that prescribe seclusion for mothers and neonates;
lack of trained caretakers and facilities; high out-of-pocket
costs for transport, hospitalization, and medications; and
loss of wages for the mother, the father, and often another
family member. As a result, the current WHO recommendation of 10 to 14 days of inpatient treatment with
broad-spectrum parenteral antibiotics is unavailable or
unacceptable to most families (Zaidi et al, 2005).
Among those babies born in hospitals, the risk of nosocomial infection is threefold to 20-fold higher (6.5 to 38
per 1000) than in industrialized countries because of poor
intrapartum and postnatal infection-control practices
(Zaidi et al, 2005). Many hospitals are overcrowded and
understaffed and lack even basic infection control procedures, despite the guidelines of WHO–United Nations
Children’s Fund Integrated Management of Pregnancy and
Childbirth and the Newborn Problems Handbook: A Guide for
Doctors, Nurses, and Midwives (World Health Organization, 2003a, 2003b; Zaidi et al, 2005). The major pathogens among babies born in a hospital (11,471 isolates) are
Klebsiella pneumonae, other gram-negative rods (Escherichia
coli, Pseudomonas spp., Acinetobacter spp.), and Staphylococcus
aureus (8% to 22%) (Zaidi et al, 2009). Newborns in a hospital often receive empiric therapy with broad spectrum
parenteral antibiotics (imipenem and amikacin), because of
the lack of culture facilities and concerns about resistance.
Data from community settings during the first week of
life are almost nonexistent. A 2009 Pediatric Infectious Disease supplement (Qazi and Stoll, 2009) reviewed evidence
on community-acquired neonatal sepsis in the developing
world from 32 studies published since 1990. The tremendous heterogeneity in studies, suggesting that infections
may be responsible for 8% to 80% of all neonatal deaths
and up to 42% of deaths in the first week, make the data
difficult to interpret. Among neonates from 0 to 60 days
old, rates of clinically diagnosed neonatal sepsis were as
high as 170 per 1000 live births versus 5.5 blood-culture
confirmed sepsis cases per 1000 live births (Thaver and
Zaidi, 2009). Gram-negative rods (Klebsiella spp. [25%],
E. coli [15%]) and S. aureus were the major communityacquired pathogens. Group B streptococcus was relatively
uncommon (7%) in the first week, but group B streptococcus, S. aureus, and nontyphoid Salmonella spp. infection
rates increased to 12% to 14% after the first week. Only
170 isolates, predominantly gram negative, were reported
among home-delivered babies. The authors concluded that
hospital-based and community studies suggest that most
infections in the first week are attributable to gram-negative pathogens that may be environmentally acquired during unhygienic deliveries, rather than maternally acquired.
As with hospital-born infants, empiric therapy with broad
spectrum antibiotics was the norm, based on clinical
diagnosis or on algorithms, because advanced technology
was neither available nor affordable. Because the signs and
symptoms of neonatal sepsis and pneumonia are nonspecific and medical systems are weak, delays in recognition,
referral, and treatment were common and were reflected in
both the high mortality rate (22%) (Bang et al, 2005) and
frequent prescription of broad-spectrum antibiotics.
A recent population-based nested observational study of
community and hospital-born neonates randomized to a
package of neonatal and maternal interventions in Mirzapur, Bangladesh, represents the difficulty of obtaining reliable infection data in the developing world. Of the 239
neonates who died without being enrolled, 59% and 87%
died within the first 2 and 7 days, respectively, and were
thought to be the result of birth asphyxia, prematurity, or
both. Among the 7310 neonates who were assessed at least
once by community health workers, the incidence of early
neonatal sepsis was only 3 in 1000 live births. The 29 positive blood cultures represent an incidence of bacteremia of
only 2.9 cases per 1000 live births; 38% of these cultures
were obtained in the first three postnatal days. Fifty percent
of the organisms were gram negative and 50% were gram
positive; 10 in 15 gram-positive organisms were S. aureus,
and one was group B streptococcus. The case fatality rate
was 13% (2/15) in the gram-positive and 27% in the gramnegative infections. Seventy percent of the isolates were
sensitive to the combination of ampicillin plus gentamicin
or ceftriaxone. The authors noted that the incidence rate
was roughly comparable to reported early-onset neonatal
sepsis in the United States; however, the reported rates are
likely to be low because infants who died early were not
enrolled, parents were not compliant with referrals, and the
intervention package may have prevented some infections.
For the many reasons noted, the current recommendations for hospitalization and parenteral therapy are simply
not feasible in the developing world. Therefore several multicenter trials have been launched in Asia (2009) and (2010)
Africa to test the safety and efficacy of simplified antibiotic
regimens to treat possible serious bacterial infections in 0- to
59-day-old infants in the community or first-level facilities
(S. Qazi, personal communication, 2010). Studies evaluating the effect of prenatal and postnatal home visits by community health workers to improve newborn care practices,
and identification and referral of positive serious bacterial
infection were completed in 2010 in Ghana and Uganda.
Studies are ongoing in Tanzania (R. Bahl, personal communication, 2011). Such research is a priority to guide community management of infections and prevent unacceptably
high neonatal mortality rates in developing countries.
Effective and simple interventions for the prevention and
treatment of neonatal infections exist, but poor coverage of
health services, a shortage of health care providers, access
to referral services, and lack of knowledge on how to implement existing cost-effective interventions at scale in lowresource settings prevent them from reaching community
neonates in the developing world. A methodology developed
by the WHO Department of Child and Adolescent Health
and Development (CAH) provides a systematic method, the
Child Health and Nutrition Research Initiative (CHNRI)
methodology, for setting priorities in health research
investments at any level (institutional to global) (Rudan
et al, 2007, 2008). Applying the CHNRI methodology to
CHAPTER 4 Global Neonatal Health
the prevention and treatment of neonatal infection identified the need for health policy and systems research to
understand the barriers to implementation, effectiveness,
and optimized use of available interventions (Bahl et al,
2009). The need for point-of-care diagnostics for neonatal
pneumonia, hypoxia, bacterial sepsis, and antibiotic resistance is urgent because standard laboratory and radiologic
technology are not available. To clarify the contribution of
vertical transmission to neonatal mortality, sepsis data during the first 3 days of life are also a high priority.
Malaria and HIV infection are threats to neonatal
health in Sub-Saharan Africa. The burden of malaria in
pregnancy is exacerbated by HIV, which increases susceptibility in pregnancy, in addition to reducing the efficacy
of antimalaria interventions and complicating their use
because of potential drug interactions. Important progress
has been made in preventing malaria with intermittent
preventive treatment in pregnancy and insecticide-treated
nets, but coverage of these treatments with funds is still
unacceptably low (Menendez et al, 2007). HIV has devastated Sub-Saharan Africa, but progress is being made.
The latest guidelines are available on the WHO Web site
(www.who.int/hiv). Other useful, sites for current recommendations for treating pregnant women, reducing
mother-to-child transmission, and infant feeding include
http://AIDSinfo.nih.gov (for U.S. guidelines and access to
information on trials and drugs), http://unaidstoday.org,
www.accessdata.fda-gov, and www.cdc.gov/hiv/dhap.htm.
ASPHYXIA
The major causes of stillbirth and early neonatal death
(during the first 7 days after birth) are birth asphyxia
(defined by the WHO as the failure to initiate and maintain
spontaneous respiration), low birthweight, and preterm
delivery. Concerns about identifying stillbirth prevention
strategies, the appropriate timing for such interventions,
misclassification of early neonatal deaths as stillbirths, and
the limitations of verbal autopsy have led to proposals to
use terms that describe the timing of an insult (intrapartum
deaths and intrapartum-related neonatal deaths) and specific
adverse outcome (neonatal encephalopathy) rather than the
term asphyxia (Lawn et al, 2009b, 2009c). Classification of
the timing of death as previable versus antepartum (macerated) or intrapartum (fresh stillbirth) may be possible, even
among the 60 million annual home births; however, the
consequences of intrapartum fetal organ damage caused
by poor oxygenation are often difficult to distinguish
from those associated with infection and trauma; therefore differentiating the specific outcomes associated with
each condition might not be important. Intrapartum fetal
organ damage caused by poor oxygenation may be the final
common pathway for many stillbirths and early neonatal
deaths (Goldenberg and McClure, 2009).
Several studies suggest that improved neonatal resuscitation skills reduce misclassification of stillbirths and improve
neonatal survival (Cowles, 2007; Daga et al, 1992), including a before-and-after study that provided college-educated
Zambian midwives equipment and training in essential
newborn care and resuscitation. The training resulted in a
reduction of stillbirths from 23 to 16 per 1000 births without an increase in neonatal deaths (Chomba et al, 2008),
31
suggesting that resuscitation training of providers can
decrease misclassification of stillbirths and improve neonatal survival.
Of the approximately 136 million babies born every
year, approximately 10% (14 million) require only stimulation at birth to establish regular respiration. As many as
3% to 6% (4 million) require stimulation and basic resuscitation with room air and a self-inflating resuscitation bag
and mask, and less than 1% (1.4 million) require advanced
resuscitation and postresuscitation care (Wall et al, 2009).
Because less than 1% of neonates require advanced resuscitation, and few of those would survive without mechanical ventilation, advanced neonatal resuscitation is not a
priority unless neonatal intensive care is available. The
1997 WHO Basic Newborn Resuscitation: A Practical Guide,
which will be revised and published in 2011, provides
guidelines for resuscitation training that are appropriate
for first-referral level facilities in low-resource settings.
A new educational resuscitation training program, Helping
Babies Breathe, by the American Academy of Pediatrics and
others (Niermeyer, 2009), is designed to support resuscitation training in low-resources settings. It emphasizes
the “golden moment,” provides clear graphics for decision
making in basic resuscitation and hands-on exercises. It is
being rolled out globally by USAID and partners. New
low-cost resuscitation bags and infant resuscitation models
will facilitate hands-on resuscitation training initiatives.
A recent review of resuscitation in low-resource settings describes the available evidence for which newborns
should be resuscitated, when resuscitation should not be
initiated, and when it should be stopped; management of
meconium-stained infants; equipment needed for ventilation during resuscitation; evidence to support resuscitation with room air; evidence of the effects of resuscitation
training in facilities and communities; postresuscitation
management; and considerations for improving neonatal
resuscitation in low- and middle-income countries (Wall
et al, 2009). Improvement will require providing essential newborn care to newborns in all settings and frequent
retraining to maintain resuscitation knowledge and skills.
The authors estimate that systematic implementation of
personnel using standard neonatal and competency-based
training could avert an estimated 192,000 intrapartumrelated neonatal deaths per year and an additional 5% to
10% of deaths as a result of complications of preterm birth
(Lawn et al, 2009; Wall et al, 2009).
Among critical issues neonatal resuscitation are:
• How to deliver neonatal resuscitation in settings with
the highest burden, but the weakest health systems
• How to implement and sustain national vital registries
• How to document the actual number of births
• How to document the number of intrapartum stillbirths
• Whether improved survival is associated with increased
numbers of disabled survivors
• How to improve monitoring of the proportion of infants
requiring resuscitation and their outcome
• How to deliver cost-effective neonatal care, resuscitation training methods, and maintenance of resuscitation skills by different levels of providers in facilities
and communities
• How to determine whether infants should be suctioned,
including those with meconium staining
32
PART I Overview
• Early infant stimulation methods to ameliorate the
effects of perinatal hypoxia
Equally important are methods to improve the quality of care for mothers and neonates, including maternal
and perinatal death reviews, criterion-based audits, and
emergency drills (van den Broek and Graham, 2009). Dissemination of the new Helping Babies Breathe curriculum
represents an important opportunity for implementation
research to improve newborn survival in the developing
world.
PROVIDING A CONTINUUM OF CARE
Early efforts to improve neonatal mortality focused on
high coverage levels of a few simple and cost-effective
interventions in low-resource settings. Because simple
interventions did not decrease neonatal mortality, the
emphasis has subsequently shifted to comprehensive packages of community interventions and to a continuum of
levels of care from home to hospital.
Interventions that improve access to quality health care
systems and can provide training, skilled birth attendants,
transportation, timely emergency obstetric and neonatal care, and early postnatal care are likely to simultaneously reduce stillbirths and early neonatal deaths, as well
as maternal morbidity and mortality; however, they have
only been achieved in the context of research. The proposed components include:
• Empowerment of women
• Increased training of all levels of birth attendants in
essential newborn care and resuscitation
• Emphasis on increased institutional deliveries
ANTENATAL CARE
• Tetanus toxoid immunization
• Nutrition: iodine, iron/folate
(periconceptional)
• Maternal infections: syphilis,
malaria (endemic areas)
• Breastfeeding counseling
• Birth preparedness*
• Danger signs*
Antenatal
Care
Labor &
Delivery Care
INTRAPARTUM CARE
• Clean delivery
• Skilled care at delivery*
• Danger signs*
• Mobilization of communities to identify and transfer
high-risk pregnancies and neonates
• Improved strategies for community treatment of postpartum hemorrhage, eclampsia, and sepsis
• Increased postpartum home visits
• The Safe Childbirth Checklist (World Health Organization, Safe Childbirth Checklist)
The joint statement from the WHO and United Nations
Children’s Fund recommending several early postpartum
visits to deliver effective elements of care to newborns
and their mothers is based on studies in Bangladesh and
Pakistan, where such visits have been associated with
reductions in newborn deaths and improved care practices.
However, postpartum visits in a large-scale communitybased integrated nutrition and health program in Uttar
Pradesh, India, improved care practices but did not reduce
the primary outcome (i.e., neonatal mortality) at the population level (Baqui et al, 2008a).
A 2007 review of the effects of packaged interventions on neonatal health (Haws et al, 2007) found no
true effectiveness trials among 19 randomized controlled trials. No trial targeted women before pregnancy,
and antenatal interventions were largely micronutrient
supplementation. Intrapartum interventions were limited principally to clean delivery, and few increased the
demand for care or improved the delivery of interventions
to large populations. Subsequent trials using existing
human and material resources and documenting external input are limited, but there is an increased emphasis
on rural community-based interventions that could be
improved. The early Bang Gadchiroli trial in Mahrashtra,
India—which achieved a greater than 60% reduction in
IMMEDIATE NEWBORN CARE
• Newborn reuscitation
• Prevention of hypothermia: drying,
warming
• Prevention of hypoglycemia: immediate
breastfeeding
• Prophylactic eye care (areas endemic
for gonorrhea)
Immediate
Newborn
Care
Postpartum Care for
Mother & Newborn
POSTNATAL CARE
• Exclusive breastfeeding
• Clean umbilical cord care
• Maintenance of temperature
• Pneumonia and sepsis
management
• Early postpartum visit*
• Birth spacing*
CONTINUUM OF CARE
FIGURE 4-5 Summary of priority antepartum, intrapartum, and postnatal interventions. (Modified from Bhutta ZA, Darmstadt GL, Hasan BS, et al:
Community-based interventions for improving perinatal and neonatal health outcomes in developing countries: a review of the evidence, Pediatrics 115[Suppl
2]:519-617, 2005.)
CHAPTER 4 Global Neonatal Health
neonatal mortality by intensive training of community
health workers to resuscitate asphyxiated infants, manage
infants with low birthweight, and treat suspected bacterial infections with oral and injectable antibiotics (Bang
et al, 1999, 2005)—has not been replicated in other communities. However, community mobilization training
(women’s support groups with little additional input of
health system strengthening) in rural Nepal between
2001 and 2003 reduced the neonatal mortality rate in
intervention clusters by 30% (adjusted odds ratio, 0.70;
95% CI, 0.53–0.94) to 26.2 per 1000 (76 deaths per 2899
live births) compared with 36.9 per 1000 (119 deaths per
3226 live births) in controls (Manandhar et al, 2004).
More recently, a community-based mobilization and education trial of care of newborn babies in rural India was
associated with improved household care behaviors (early
initiation of breastfeeding, delayed bathing, and skin-toskin care) and a reduction of neonatal mortality (Kumar
et al, 2008). This 30-month, community-based unmasked
cluster randomized trial was conducted through government and nongovernmental organization infrastructures.
The trial provided home care visits (two prenatal and four
postnatal care home care and referral or treatment of sick
neonates by a female community health worker [CHW])
or community-based promotion of care-seeking and
birth or newborn care preparedness through group sessions with female and community mobilizers, in addition
to a comparison arm. Neonatal mortality was reduced
in the home-care arm by 34% (adjusted RR, 0.66; 95%
CI, 0.47–0.93) over the last 6 months of the intervention versus the comparison arm. The community-care
arm documented improved care practices, but no reduction in neonatal mortality. These favorable results were
achieved despite a much lower community health worker
(CHW) density and 30% of the CHW postnatal care visits, compared with the Gadcharoli trial (Bang et al, 2005).
Improvement of the home care service delivery strategy
for essential newborn care is underway in Bangladesh
(Baqui et al, 2008b). Although it appears that communitybased preventive strategies for newborn care can improve
newborn survival and care practices, it is not clear that
government health care workers and CHWs can duplicate these research results (Bhutta and Soofi, 2008).
Because each setting is unique, such efforts are likely to
be improved after local formative research with the communities, CHW, and birth attendants (Bahl et al, 2008).
Key research gaps in community management include:
• How best to create the political will to prioritize community maternal and neonatal health
• How to provide a continuum of care from home to
hospital (effectiveness trials carefully tailored to local
health needs and conducted at scale)
• How to mobilize communities to identify, stabilize, and
transfer at-risk pregnancies and neonates
• What strategies should be used to ensure quality of care
• How to manage birth asphyxia, preterm delivery, and
serious neonatal bacterial infections in the community
Finally, it is important to test whether the community strategies that were effective in rural Southern Asia
will be equally effective in Africa and in urban slums
(Bhutta and Soofi, 2008; Bhutta et al, 2008b; Kumar
et al, 2008).
33
RESOURCES
Some of the key challenges to global health initiatives are
information, communication, and assessment. Although
the data are not always available or consistent, a number
of important resources are available. The United Nations
Children’s Fund reports progress in maternal and child
survival in the The State of the World’s Children, based on
the work of the Inter-agency Group for Child Mortality
Estimation (United Nations Children’s Fund, 2007). The
State of the World’s Children provides summary tables of
basic, health, education, demographics, economics, and
progress indicators with national rankings. The detailed
text discusses when, where, and why mothers and neonates
die, and it documents interventions to improve outcomes.
The 2009 State of the World’s Children emphasizes maternal
health.
The WHO Department of Child and Adolescent
Health and Development is the Secretariat for the Child
Epidemiology Reference Group (CHERG) that quantifies
the burden of child illnesses, supports and disseminates
research to understand the determinants of childhood illnesses, and develops and evaluates interventions of new
delivery strategies and large-scale interventions (Bryce
et al, 2005; World Health Organization, 2009).
A number of other important partnerships publish
current data, including The Countdown to 2015 (United
Nations Children’s Fund, 2005, 2008a), which assembles
and summarizes the latest published data on 68 priority
countries that represent 97% of child and maternal mortality worldwide. A unique feature is data on coverage rates
for interventions that are feasible for universal implementation in poor countries and have been empirically proven
to reduce mortality in mothers, children, and neonates.
The 2008 edition included approaches such as delivery
care and reproductive health services, which can serve as
platforms for delivering multiple, proven interventions to
reduce maternal and neonatal mortality (United Nations
Children’s Fund, 2008a); it is intended to assist policy
makers, development agencies, and donors in making performance-based policy and decisions. An explicit goal is to
hold governments, development partners, and the international health community accountable for the lack of progress (United Nations Children’s Fund, 2008a).
In 2003 a group of technical experts published The
Child Survival Series in The Lancet (United Nations
Children’s Fund, 2007), which went on to become a
unique series of special editions on perinatal health in the
developing world. The series has played a critical role in
drawing attention and resources to improved neonatal
survival, which is important to the future of the developing world.
The comprehensive Disease Control Priorities Project
(DCPP)—a joint effort of the National Institutes of Health
Fogarty International Center, the WHO, and the World
Bank—was launched in 2001 to identify policy changes
and intervention strategies for the health problems of
countries in need. The aim of the DCPP was to generate
knowledge to assist decision makers in developing countries to realize the potential of cost-effective interventions
to rapidly improve the health and welfare of their populations and to detail prevalent investments that are not cost
34
PART I Overview
By disease
22
21-79
Unallocable
Other
Health-sector support
Tuberculosis
Malaria
HIV/AIDS
20
18
18-99
17-90
15-60
14
13-55
12-44
12
10-69 10-90
9-80
7-76
8
8-01
8-10
8-42
8-65
98
10
97
2007 US$ (billions)
16
6-61
6
5-59
6-11
5-47
4
2
07
20
06
05
20
04
20
20
03
02
20
01
20
20
00
99
20
19
19
96
19
95
19
94
19
93
19
92
19
91
19
19
19
90
0
Year
FIGURE 4-6 Development assistance for health from 1990 to 2007, by disease. (Modified from Ravishankar N, Gubbins P, Cooley RJ, et al: Financing of
global health: tracking development assistance for health from 1990 to 2007, Lancet 373:2113-2124, 2009.)
effective. The DCPP published an expanded and updated
second edition that addresses disease conditions, their
burdens and risk factors, strategy and intervention effectiveness and health systems, and financing (Laxminarayan
et al, 2006).
FUNDING
There is no comprehensive system for tracking total
amounts of developmental assistance for health or how
they are spent. However, recent analyses have documented
a fourfold increase in developmental assistance, from $5.6
billion in 1990 to $21.8 billion in 2007. The WHO estimated that the 10-year incremental global costs for universal health coverage of maternal and child health services
ranged from $39.3 billion for a moderate improvement
scenario to $55l.7 billion for a rapid improvement scenario. These projections did not include the cost of health
system reforms, such as recruiting, training, and retaining
a sufficient number of personnel (Johns et al, 2007).
Global assistance rose sharply after 2002 because of
increases in public funding, especially from the United
States, and from increased philanthropic donations and
in-kind contributions from corporations. (The Bill and
Melinda Gates Foundation is the largest single source of
private developmental health assistance.) Donor funding
from the United States for HIV/AIDS has increased from
$300 million in 1996 to $8.9 billion in 2006 (Oomman,
et al, 2007). The proportion of developmental assistance
from United Nations agencies and development banks
decreased between 1990 and 2007 as targeted funding
increased for the Global Alliance for Vaccines and Immunization; Medicines for Malaria, the Global Fund to Fight
AIDS, Tuberculosis and Malaria; and the United States
President’s Emergency Plans for AIDS Relief (PEPFAR). The influx of funds has been accompanied by major
changes in the institutional landscape of global health,
with global health initiatives such as the Global Fund and
the GAVI assuming a central role in mobilizing and channeling global health funds. Nongovernmental organizations have become a major conduit for an increasing share
of developmental assistance (Ravishankar et al, 2009).
The pattern is similar for research and development for
drugs: Global Funds and GAVI HIV/AIDS, tuberculosis,
and malaria initiatives accounted for approximately 80%
of the $2.5 billion that was spent on research and drug
development in 2007 for neglected diseases in developing
countries (Moran et al, 2009). Drugs and vaccines—rather
than diagnostics, platform technologies, or country-specific products—are also funded preferentially (Moran et al,
2009). Research and development in neglected diseases—
such as pneumonia and diarrheal illness, two major causes
of mortality in developing countries–are severely underfunded at less than 6% of the budgeted funding. Increasing
attention has been focused on the large amount of funding
being earmarked for HIV, malaria, and tuberculosis and
CHAPTER 4 Global Neonatal Health
35
TABLE 4-4 The Research Pipeline of Description and Determinants, Discovery, Development, and Delivery
Description and
Determinants
Discovery
Development
Delivery
Research aim
Descriptive epidemiology and understanding
determinants, advancing
definitions
New science for the discovery of mechanisms and
causes of neonatal disease
that provides foundation for developing new
interventions
Developing new or
adapting existing
interventions to
reduce the cost,
increase effect,
improve deliverability
Delivering existing interventions in new ways or in
new settings. Includes
monitoring and evaluation
as feedback to additional
discovery and development science
Types of research
(research
instrument)
Epidemiology
New drugs and vaccines.
Biochemical and genetic
basis for disease
Refining or adapting
existing technology
or drugs
Effectiveness trials or implementation research for
scale-up in health systems
Typical timeline before
impact is seen
Variable
5 to 15 years
5 to 10 years
2 to 5 years
Investment level
Variable
Very high
Moderate
Low to moderate depending
on size of trial and rigor of
evaluation
Probability of major
impact
Variable, if new epidemiology leads directly to
program or intervention
Very high
Moderate
Very high if high impact
intervention and currently
low coverage
Risk of failure
Low
High
Moderate
Low to moderate
Specific examples for
global newborn
health
Cohort studies to better
delineate preterm birth
and term small for gestational age and define
short-and long-term
outcomes
Discovering a marker for
preterm birth amenable
to intervention
Adapting technology for head or
body cooling to be
effective, lower cost,
feasible and safe in
low-income settings
Impact and cost to provide
early postnatal care package using different cadres
of workers in a range of
varying health system contexts especially in Africa
From Lawn JE, Rudan I, Rubens C: Four million newborn deaths: is the global research agenda evidence-based? Early Hum Dev 84:809-814, 2008.
the missed opportunities to save more lives—especially
young lives—at a lower cost by focusing on simpler interventions (Gostin, 2008).
CAREER OPPORTUNITIES
American universities are experiencing an unprecedented
surge in interest in global health. Students and faculty have
become actively engaged in operational research, project
analysis, workforce training, and policy debates. A number
of American universities have made long-term commitments to specific countries in the developing world, including formal opportunities for faculty and residents to work
in targeted countries in low-resource settings. Opportunities range from in-depth experiences to volunteer research
and service projects. The newly launched Consortium of
University for Global Health 2009 survey of 37 universities found that the number of students enrolled in global
health programs in universities across the United States
and Canada doubled in just 3 years and that universities
have established 302 training and education programs in
97 countries. Although there is currently no official “bulletin
board” for international global opportunities at the faculty
level, a number of Web sites offer a range of opportunities
for individuals seeking additional training, career opportunities, and interaction with other global health professionals, including the American Medical Students Association
(www.amsa.org); the Global Health Council career network (careers.globalhealth.org); the United States Agency
for International Development (USAID) (www.usaid.gov);
and the National Institutes of Health Fogarty International
Center (www.nih.fogarty.org). A number of Web sites also
provide research updates, including the WHO, USAID,
Save the Children Newborn Research eUpdates, and Medical
News Today. The Federation of Pediatric Organizations is
working in the areas of international certification, cataloging international rotations, creating a checklist of requirements for international rotations, and creating global
partnerships.
THE WAY FORWARD: DATA,
COLLABORATION, EVALUATION,
AND INVOLVEMENT
There is a consensus in the global research community
regarding the importance of providing a continuum of care
from home to hospital for mothers and neonates and evaluating the effectiveness of packages of interventions that
have proved effective in smaller trials (Madon et al, 2007).
Much attention has been focused on the lack of quality data;
social and cultural limitations; the need for large community randomized trials and their high cost; the lack of coordination of efforts to maximize current data by prioritizing
and strengthening existing programs with proven, low-cost,
high-impact interventions; and the need to systematically
establish research priorities (Lawn et al, 2008, 2009a).
The need for a change in the design, implementation,
and evaluation of programs has received less attention, to
meet the needs of national governments and donors for
rigorous assessment of child survival and health in general.
Victora, Black, and Bryce (2009) emphasize the need for
nationwide improvement and nationwide assessments of
multiple programs, in collaboration with the government
and other concurrent programs. They suggest three initial
36
PART I Overview
steps for an ecologic evaluation platform: (1) develop and
regularly update a district database from multiple sources,
(2) conduct an initial survey to be repeated every 3 years
to measure coverage for proven interventions and health
status, and (3) establish a continuous monitoring system to document provision, use, and quality of interventions at the district level, with mechanisms for prompt
and transparent reporting. Although this ambitious plan
would support the analysis of combinations of interventions and delivery strategies with the ability to adjust for
confounders, there is no precedent for undertaking such
a massive effort. Short of their comprehensive strategy,
there is increasing recognition of the need for national
vital registries and recurrent surveys to provide the basis
for changes in health policy. There is also clear evidence
of the need for a new emphasis on implementation science
and the urgency of strengthening the independent capacity for health research in the developing world (Whitworth
et al, 2008), which will enable collaborators to solve their
own national problems. Finally, everyone has the power
to advocate for political change to support maternal and
child health. Shiffman (2009) emphasizes the need to build
a strong policy community to generate political attention
for global maternal and neonatal health; to develop issue
frames that resonate with politicians to move them to act;
to cultivate strategic alliances within women’s groups, key
ministers, and congressional aides; to link the health of
women and neonates in the developing world with other
problems; and to remember that medical professionals
carry great moral authority because of their expertise and
pursuit of a humanitarian cause, if they choose to exert
their political power in a strategic way.
SUGGESTED READINGS
Baqui A, Williams EK, Rosecrans AM, et al: Impact of an integrated nutrition and
health programme on neonatal mortality in rural northern India, Bull World
Health Organ 86:796-804, 2008.
Bhutta ZA, Ali S, Cousens S, et al: Alma-Ata. Rebirth and revision 6 interventions
to address maternal, newborn, and child survival: what difference can integrated primary health care strategies make?, Lancet 372:972-989, 2008.
Himawan B: State of the world’s mothers 2007: saving the lives of children under 5, 2007,
New York, Save the Children.
Jamison DT, Breman JG, Measham AR, et al: Disease control priorities in developing
countries: a copublication of The World Bank and Oxford University Press, ed 2, New
York, 2006, Oxford University Press.
Madon T, Hofman KJ, Kupfer L, et al: Public health: implementation science,
Science 318:1728-1729, 2007.
Martines J, Paul VK, Bhutta ZA, et al: Neonatal survival: a call for action , Lancet
365:1189-1197, 2005.
Morris SS, Cogill B, Uauy R: Effective international action against undernutrition:
why has it proven so difficult and what can be done to accelerate progress?
Lancet 371:608-621, 2008.
Save the Children: Serious bacterial infections among neonates and young infants
in developing countries: evaluation of etiology and therapeutic management
strategies in community settings, Pediatr Infect Dis J 28:S1-S48, 2009.
United Nations Children’s Fund: Countdown 2015: tracking progress in maternal,
newborn & child survival, the 2008 report, vol 2, New York, 2008a.
United Nations Children’s Fund: The state of the world’s children 2008: child survival,
New York, 2007.
United Nations Children’s Fund: The state of the world’s children 2009: maternal and
newborn health, New York, 2008b.
Complete references used in this text can be found online at www.expertconsult.com
P A R T
I I
Fetal Development
C H A P T E R
5
Immunologic Basis of Placental Function
and Diseases: the Placenta, Fetal
Membranes, and Umbilical Cord
Satyan Kalkunte, James F. Padbury, and Surendra Sharma
Complex yet intricate interactions between maternal and
fetal systems promote fetal growth and normal pregnancy
outcomes. Throughout embryonic development, organogenesis and functional maturation are taking place. This
period of development coincides with a high rate of cellular proliferation and organ development, which creates
critical periods of vulnerability. Adverse factors, disruption,
or impairment during these critical periods of fetal development can alter developmental programming, which can
lead to permanent metabolic or structural changes (Baker,
1998). For example, triggers such as undernutrition can
elicit placental and fetal adaptive responses that can lead
to local ischemia and metabolic, hormonal, and immune
reprogramming, resulting in small for gestational age (SGA)
fetuses. Maternal health, dietary status, and exposure to
environmental factors, uteroplacental blood flow, placental
transfer, and fetal genetic and epigenetic responses likely
all contribute to in utero fetal programming (Figure 5-1).
Adult diseases such as coronary heart disorders, hypertension, atherosclerosis, type 2 diabetes, insulin resistance,
respiratory distress, altered cell-mediated immunity, cancer, and psychiatric disorders are now thought to be a consequence of in utero life (Sallour and Walker, 2003). It is a
matter of considerable interest that, in addition to maternal
predisposing factors, cytokines, hormones, growth factors,
and the intrauterine immune milieu also contribute to in
utero programming. Adaptations of the maternal immune
system exist to modulate detrimental effects on the fetus
and additional mechanisms and factors actively cross the
placenta and induce regulatory T cells in the fetus to suppress fetal antimaternal immunity. These effects persist at
least through adolescence (Burlingham, 2009; Mold et al,
2008). Excessive restraint of maternal immune responses
could lead to a lethal infection in the newborn. On the other
hand, too little modulation of maternal immune response to
the fetal allograft could lead to autoimmune-mediated fetalplacental rejection. Moreover, placental growth resembles
that of a tumor, evading immune surveillance and initiating its own angiogenesis. Therefore a healthy mother with
healthy placentation is critical to healthy fetal outcomes.
MAMMALIAN PLACENTATION
The immune tolerance of the semiallograft fetus and de
novo vascularization are two highly intriguing processes
that involve direct interaction of maternal immune cells,
invading trophoblast cells, and arterial endothelial cells.
Pregnancy is considered an immunologic paradox, in
which paternal antigen-expressing placental cells interact
directly with and coexist with the maternal immune system
(Medawar, 1953). This anatomic distinction of the immunologic interface that arises from hemochorial placentation
that occurs in humans and rodents is distinct from epitheliochorial placentation as seen in marsupials, horses, and
swine or the endotheliochorial placentation seen in dogs
and cats. Understanding the anatomic and physiologic
events that occur during placentation is the key to appreciate the uniqueness of human placentation in the phylogenetic evolution. Typically, in hemochorial placentation,
maternal uterine blood vessels and decidualized endometrium are colonized by trophoblast cells, derived from
trophectoderm of the implanting blastocyst. These cells
come in direct contact with maternal blood and uterine
tissue. A similar phenomenon is evident in murine pregnancy, except the trophoblast invasion is deeper in humans
(Moffett and Loke, 2006). In epitheliochorial placentation,
trophoblast cells of the placenta are in direct contact with
the surface epithelial cells of the uterus, but there is no
trophoblast-cell invasion beyond this layer. In endotheliochorial placentation, the trophoblast cells breach the uterine epithelium and are in direct contact with endothelial
cells of maternal uterine blood vessels.
EMBRYOLOGIC DEVELOPMENT
OF THE PLACENTA
Shortly after fertilization takes place in the ampullary portion of the fallopian tube, the fertilized ovum or zygote
begins dividing into a ball of cells called a morula. As the
morula enters the uterus (by the fourth day after fertilization), it forms a central cystic area and is called a blastocyst
(Figure 5-2). The blastocyst implants within the endometrium by day seven (Moore, 1988).
The blastocyst has two components: an inner cell mass,
which becomes the developing embryo, and the outer cell
layer, which becomes the placenta and fetal membranes.
The cells of the developing blastocyst, which eventually
become the placenta, are differentiated early in gestation
(within 7 days after fertilization). The outer cell layer, the
trophoblast, invades the endometrium to the level of the
decidua basalis. Maternal blood vessels are also invaded.
Once entered and controlled by the trophoblast, these
37
38
PART II Fetal Development
Maternal
Placenta
Fetus
Health status
Uteroplacental
perfusion
Metabolic
adaptation
Diet and
exposure status
Ischemia
Genetics and
epigenetics
Immune status
Hormonal
regulation
Immune
regulation
Placental-fetal
response
Outer cell layer
Inner cell mass
A
Cytotrophoblast
Syncytiotrophoblast
Fetal pole
Fetal Programming
FIGURE 5-1 Fetal programming. Maternal health and the placenta
influence fetal adaptations. Dietary status, exposure to environmental
factors, uteroplacental blood flow, placental transfer, and genetic and
epigenetic changes contribute to the in utero fetal programming.
maternal blood vessels form lacunae, which provide nutrition and substrates for the developing products of conception. The trophoblast differentiates into two cell types,
the inner cytotrophoblast and the outer syncytiotrophoblast (Figure 5-3); the former has distinct cell walls and is
thought to represent the more immature form of trophoblast. The syncytiotrophoblast, which is essentially acellular, is the site of most placental hormone and metabolic
activity. Once the trophoblast has invaded the endometrium, it begins to form outpouchings called villi, which
extend into the blood-filled maternal lacunae or further
invade the endometrium to attach more solidly with the
decidua, forming anchoring villi.
B
FIGURE 5-2 A, The human blastocyst contains two portions: an
inner cell mass, which develops into the embryo, and an outer cell
layer, which develops into the placenta and membranes. B, The outer
acellular layer is the syncytiotrophoblast, and the inner cellular layer is
the cytotrophoblast. (From Moore TR, Reiter RC, Rebar RW, et al, editors:
Gynecology and obstetrics: a longitudinal approach, New York, 1993,
Churchill Livingstone.)
PLACENTAL ANATOMY AND CIRCULATION
At term, the normal placenta covers approximately one
third of the interior portion of the uterus and weighs
approximately 500 g. The appearance is of a flat circular
disc approximately 2 to 3 cm thick and 15 to 20 cm across
(Benirschke and Kaufmann, 2000). Placental and fetal
weights throughout gestation are presented in Table 5-1.
During the first trimester and into the second, the placenta
weighs more than the fetus; after that period, the fetus outweighs the placenta. With the formation of the tertiary villi
(19 days after fertilization), a direct vascular connection
is made between the developing embryo and the placenta
(Moore, 1988). Umbilical circulation between the placenta
and the embryo is evident by 51⁄2 weeks’ gestation. Figure
5-4 demonstrates aspects of the maternal and fetal circulation in the mature placenta. The umbilical arteries from the
fetus reach the placenta and then divide repetitively to cover
the fetal surface of the placenta. Terminal arteries then penetrate the individual cotyledons, forming capillary beds for
substrate exchange within the tertiary villi. These capillaries
then reform into tributaries of the umbilical venous system,
which carries oxygenated blood back to the fetus.
EXAMINATION OF THE PLACENTA
A renaissance in placental pathology has led to a new relevance of the placenta to neonatology and early infant
life, including issues of preterm birth, growth restriction,
and cerebral, renal, and myocardial diseases. The placenta
can give some clues to the timing and extent of important adverse prenatal or neonatal events as well as to the
relative effects of sepsis and asphyxia on the causation of
neonatal diseases. Placental disorders can be noted immediately in the delivery room, and others can be diagnosed
through detailed gross and microscopic examinations over
the ensuing 48 hours. Every placenta should be examined
at the time of birth regardless of whether the newborn
has any immediate problems. Most placentas invert with
traction at the time of delivery, and the fetal membranes
cover the maternal surface. It is important to reinvert the
membranes and examine all surfaces of the placenta and
membranes, looking for abnormalities. Table 5-2 lists
pregnancy complications or conditions that are diagnosable at birth through examination of the placenta.
39
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
Maternal lacunae
Syncytiotrophoblast
Cytotrophoblast
A
Endometrium
Mesenchymal cells
TABLE 5-1 Fetal and Placental Weight Throughout Gestation
Gestational
Age (wk)
Placental
Weight (mg)
Fetal
Weight (g)
14
45
—
16
65
59
18
90
155
20
115
250
22
150
405
24
185
560
26
217
780
28
250
1000
30
282
1270
32
315
1550
34
352
1925
36
390
2300
38
430
2850
40
470
3400
Adapted from Benirschke K, Kaufmann P: Pathology of the human placenta, ed 4,
New York, 2000, Springer-Verlag.
B
Fetal blood vessel
cases of meconium aspiration syndrome, for which legal
questions may arise as to whether the aspiration occurred
before or during labor. If the membranes are deeply
stained, the passage of meconium by the fetus may have
predated onset of labor; therefore aspiration could have
occurred before labor. The umbilical cord should also
be examined for the number of vessels and their insertion into the placenta. Vessels on the fetal surface of the
placenta should be examined for evidence of clotting or
thrombosis.
FUNCTIONS OF THE PLACENTA
C
FIGURE 5-3 A, The cytotrophoblast indents the syncytiotrophoblast
to form primary villi. B, Mesenchymal cells invade the cytotrophoblast
2 days after formation of the primary villi to form secondary villi.
C, Blood vessels arise de novo and eventually connect with blood
vessels from the embryo, forming tertiary villi. (From Moore TR, Reiter
RC, Rebar RW, et al, editors: Gynecology and obstetrics: a longitudinal
approach, New York, 1993, Churchill Livingstone.)
The initial placental examination should include checking the edges for completeness. The membranes and
fetal surface should be shiny and translucent. An odor
may suggest infection, and cultures of the placenta may
be beneficial (Benirschke and Kaufmann, 2000). Greenish discoloration may represent meconium staining or old
blood; placentas with such discoloration should be sent to
the pathologist for complete histologic examination. The
finding of deep meconium staining of the membranes and
umbilical cord suggests that the meconium was passed at
least 2 hours before delivery; this fact may be helpful in
To ensure normal fetal growth and development, the
placenta behaves as an efficient organ of gas and nutrient
exchange and as a robust endocrine and metabolic organ.
Besides mediating the transplacental exchange of gases
and nutrients, the placenta also synthesizes glycogen with
a significant turnover of lactate. Hormones secreted by
the placenta have an important role for the fetus and the
mother. Placental trophoblasts are a rich source of cholesterol and peptide hormones, including human chorionic gonadotrophin (HCG), human placental lactogen,
cytokines, growth hormones, insulin-like growth factors,
corticotrophin-releasing hormones, and angiogenic factors such as vascular endothelial growth factor (VEGF) and
placental growth factor (PlGF). HCG, which is detected
as early as day 8 after conception, is secreted by syncytiotrophoblasts into the maternal circulation, reaches maximal
levels by week 8 of pregnancy and diminishes later during
gestation. HCG is essential to promote estrogen and progesterone synthesis during different stages of pregnancy.
Human placental lactogen mobilizes the breakdown of
maternal fatty acid stores and ensures an increased supply
of glucose to the fetus. VEGF and PlGF are secreted by
trophoblasts and specialized natural killer (NK) cells in the
decidua, and they promote angiogenesis and vascular activity, particularly during early stages of pregnancy when spiral
40
PART II Fetal Development
Umbilical vein
Fetal
circulation
Umbilical arteries
Amniochorionic membrane
Decidua perietalis
Smooth chorion
Intervillous space
Chorionic plate
Mainstem
villus
Amnion
Stump of
mainstem villus
Placental
septum
Decidua
basalis
Anchoring villus
Myometrium
Endometrial
veins
Endometrial
arteries
Cytotrophoblastic shell
Maternal circulation
FIGURE 5-4 Schematic drawing of a section of a mature placenta showing the relation of the villous chorion (fetal part of the placenta) to the
decidua basalis (maternal part of the placenta), the fetal placental circulation, and the maternal placental circulation. Maternal blood flows into the
intervillous spaces in funnel-shaped spurts, and exchanges occur with the fetal blood as the maternal blood flows around the villi. Note that the umbilical arteries carry deoxygenated fetal blood to the placenta, and the umbilical vein carries oxygenated blood to the fetus. In addition, the cotyledons
are separated from each other by decidual septa of the maternal portion of the placenta. Each cotyledon consists of two or more mainstem villi and
their main branches. In this drawing, only one mainstem villus is shown in each cotyledon, but the stumps of those that have been removed are shown.
(From Moore KL: The developing human: clinically oriented embryology, ed 5, Philadelphia, 1993, WB Saunders.)
TABLE 5-2 Pregnancy Conditions Diagnosable at Birth by Gross Placental Examination and Associated Neonatal Outcomes
Pregnancy Conditions
Fetal/ Neonatal Outcomes
Monochorionic twinning
TTT syndrome donor/recipient status, pump twin in TRAP, survivor status after fetal demise,
selective termination, severe growth discordance without TTT
Dichorionic twinning
Less likelihood of survivor brain disease in the event of demise of one fetus
Purulent acute chorioamnionitis
Risk of fetal sepsis, fetal inflammatory response syndrome, cerebral palsy
Chorangioma
Hydrops, cardiac failure, consumptive coagulopathy
Abnormal cord coiling
IUGR, fetal intolerance of labor
Maternal floor infarction
IUGR, cerebral disease
Abruption
Asphyxial brain disease
Velamentous cord
IUGR, vasa previa
Cord knot
Asphyxia
Chronic abruption oligohydramnios syndrome
IUGR
Single umbilical artery
Malformation, IUGR
Umbilical vein thrombosis
Asphyxia
Amnion nodosum
Severe oligohydramnios leading to pulmonary hypoplasia
Meconium staining
Possible asphyxia, aspiration lung disease
Amniotic bands
Fetal limb reduction abnormalities
Chorionic plate vascular thrombosis
Asphyxia, possible thrombophilia
Breus mole
Asphyxia, IUGR
IUGR, Intrauterine growth retardation; TRAP, twin-reversed arterial perfusion; TTT, twin-to-twin transfusion.
artery transformation and trophoblast invasion occurs. In
addition, the placenta is a rich source of estrogen, progesterone, and glucocorticoids. Whereas progesterone maintains a quiescent, noncontractile uterus, it also has a role in
protecting the conceptus from immunologic rejection by
the mother. Glucocorticoids promote organ development
and maturation. Placental transport is another important
function, efficiently transferring nutrients and solutes that
are essential for normal fetal growth. The syncytiotrophoblast covering the maternal villous surface is a specialized
epithelium that participates in the transport of gases, nutrients, and waste products and the synthesis of hormones
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
Anchoring
Villi
Placenta
41
ST
CT
IT
Decidua
ET
NK
ST: Syncytiotrophoblasts
M
T
Myometrium
CT: Columnar trophoblasts
DC
ET: Endovascular trophoblasts
IT: Interstitial trophoblasts
Treg
GC
Spiral
arteries
GC: Giant cells
NK: Natural killer cells
T: T lymphocytes
Treg: Regulatory T cells
M: Macrophages
DC: Dendritic cells
Normal
Preeclampsia/IUGR
FIGURE 5-5 Trophoblast differentiation and spiral artery remodeling. Progenitor trophoblast cells from villi differentiate into syncytiotrophoblasts and the extravillous cytotrophoblasts (EVTs). EVTs migrate out in columns as columnar trophoblasts and anchor the placenta to the decidua.
Further differentiation takes place into invasive or proliferative EVTs. The invasive EVTs invade the decidua known as interstitial trophoblasts, and
some of them fuse to form the multinucleated gaint cells. Endovascular transformation ensues as endovascular trophoblasts migrate into and colonize
the spiral arteries, almost reaching the myometrium. This results in wide-bore, low-resistant capacitance blood vessels as observed in normal pregnancy. In contrast, shallow trophoblast invasion and incomplete transformation of spiral arteries is a common feature of preeclampsia and intrauterine
growth restriction.
that regulate placental, fetal, and maternal systems. The
syncytiotrophoblast layer of the placenta is an important
site of exchange between the maternal blood stream and
the fetus. In addition to simple diffusion, syncytiotrophoblasts facilitate exchange by transcellular trafficking that
utilizes transport proteins such as the water channels (aquaporins). Facilitated diffusion for molecules such as glucose
and amino acids are performed by glucose transporters
(GLUT) and amino acid transporters. In addition, adenosine triphosphate (ATP)-mediated active transport, such as
the Na+, K+-ATPase or the Ca2+-ATPase, besides endocytosis and exocytosis, participates in transplacental exchange
(Hahn et al, 2001; Malassiné and Cronier, 2002; Randhawa
and Cohen, 2005; Siiteri, 2005).
In healthy women who are not pregnant, uterine blood
vessels receive approximately 1% of the cardiac output to
maintain the uterus. During pregnancy, these same vessels must support the rapidly growing and demanding placenta and fetus. This evolutionary challenge is addressed
by remodeling of the spiral arteries, converting them into
large, thin-walled, dilated vessels with reduced vascular
resistance.
TROPHOBLAST DIFFERENTIATION
AND REMODELING OF SPIRAL
ARTERIES
The placental-decidual interaction through invading trophoblasts determines whether an optimal transformation
of the uterine spiral arteries is achieved. Trophoblastorchestrated artery remodeling is an essential feature of
normal human pregnancy. As shown in Figure 5-5, progenitor trophoblast cells from villi differentiate along two
pathways: terminally differentiated syncytiotrophoblasts
and the extravillous cytotrophoblasts (EVTs) that migrate
out in columns and anchor the placenta to the decidua.
From these anchoring layers of EVTs further differentiation takes place into invasive or proliferative EVTs.
The invasive EVTs invade the decidua and shallow parts
of the myometrium and are known as interstitial EVTs.
Thereafter endovascular transformation ensues as invasive
EVTs migrate into and colonize the spiral arteries, almost
reaching the myometrium. These trophoblasts are known
as endovascular EVTs. Insufficient uteroplacental interaction characterized by shallow trophoblast invasion and
42
PART II Fetal Development
incomplete transformation of spiral arteries is a common
feature of preeclampsia and intrauterine growth restriction
(IUGR) (Brosens et al, 1977; Meekins et al, 1994). The
precise period when trophoblast invasion of decidua and
spiral arteries ceases is not clear. Nevertheless it is widely
believed to be completed late in the second trimester.
Although our understanding of the molecular events
underlying spiral artery remodeling in pregnancy remains
poor, efficient trophoblast invasion is an essential feature.
There are two waves of trophoblast invasion that follow
implantation. The first wave is during the first trimester,
when the invasion is limited to the decidual part of the
spiral artery. The second wave is during the late second
trimester involving deeper trophoblast invasion, reaching
the inner third of myometrial segment. The initial invasion of EVTs into the endometrium initiates the decidualization process, which is characterized by replacement of
extracellular matrix, loss of normal musculoelastic structure, and deposition of fibrinoid material. Displacement
of the endothelial lining of spiral arteries by the invading
trophoblasts further results in uncoiling and widening
of the spiral artery, ensuring the free flow of blood and
nutrients to meet the escalating demands of the growing
fetus (Kham et al, 1999; Pijnenborg et al, 1983). A lack of
spiral artery remodeling with shallow trophoblast invasion
has been associated with preeclampsia. During the process of invasion in a normal pregnancy, cytotrophoblasts
undergo phenotypic switching, with a loss of E-cadherin
expression, and they acquire vascular endothelial-cadherin,
platelet-endothelial adhesion molecule-1, vascular endothelial adhesion molecule-1, and α4 and αvβ3 integrins
(Bulla et al, 2005; Zhau et al, 1997). Along with a repertoire
of facilitators for invasion, trophoblasts express a nonclassic major histocompatibility complex (MHC) human leukocyte antigen (HLA) G, which has gained widespread
interest because of providing noncytotoxic signals to uterine NK cells. It still needs to be evaluated whether intrinsic
HLA-G inactivation by polymorphic changes influences
the dysregulated trophoblast invasion seen in preeclampsia
(Hiby et al, 1991; Le Bouteiller et al, 2007).
Although the exact gestational age at which trophoblast invasion ceases is not known, recent studies have
shown that late pregnancy trophoblasts loose the ability
to transform the uterine arteries. Using a novel dual-cell
in vitro culture system that mimics the vascular remodeling events triggered by normal pregnancy serum, we have
shown that first- and third-trimester trophoblasts respond
differentially to interactive signals from endothelial cells
when cultured on the extracellular matrix, matrigel. Term
trophoblasts not only fail to respond to signals from endothelial cells, but they inhibit endothelial cell neovascular
formation. In contrast, trophoblast cells representing firsttrimester trophoblasts with invasive properties undergo
spontaneous migration and promote endothelial cells to
form a capillary network (Figure 5-6).
This disparity in behavior was confirmed in vivo using
a matrigel plug assay. Poor expression of VEGF-C and
VEGF receptors coupled with high E-cadherin expression by term trophoblasts contributed to their restricted
migratory and interactive properties. Furthermore, these
studies showed that the kinase activity of VEGF receptor 2
is essential for proactive crosstalk by invading first-trimester trophoblast cells (Kalkunte et al, 2008b). This unique
maternal and fetal cell interactive model under the pregnancy milieu offers a potential approach to study cell-cell
interactions and to decipher inflammatory components
in the serum samples from adverse pregnancy outcomes
(Kalkunte et al, 2010). One of the inimitable contributors
to trophoblast cell invasion is the specialized NK cell of
the pregnant uterus.
IMMUNE PROFILE AND
IMMUNO VASCULAR BALANCE
DURING PLACENTATION
During pregnancy, trophoblast cells directly encounter
maternal immune cells at least at two sites. One site is the
syncytiotrophoblasts covering the placental villi that are
bathed in maternal blood, and the other is by the invading trophoblasts in the decidua. Although the syncytiotrophoblasts do not express MHC antigens, the invading
trophoblasts express nonclassic HLA-G and HLA-C
and would elicit immune responses in the decidua. The
decidua is replete with innate immune cells including
T cells, regulatory T cells, macrophages, dendritic cells
and NK cells (Table 5-3). Interestingly, NK cells peak and
constitute the largest leukocyte population in the early
pregnant uterus, accounting for 60% to 70% of total lymphocytes. These cells diminish in proportion as pregnancy
proceeds.
PHENOTYPIC AND FUNCTIONAL FEATURES
OF UTERINE NATURAL KILLER CELLS
Peripheral blood NK (pNK) cells constitute 8% to 10%
of the CD45+ population in circulation. All NK cells are
characterized by a lack of CD3 and expression of CD56
antigen. Based on the intensity of CD56 antigen, NK cells
are further divided into CD56bright and CD56dim populations. The presence or absence of FcγRIII or CD16 further differentiates subpopulations of uterine NK (uNK)
cells. Thus the majority of peripheral NK cells are of the
CD56dimCD16+ phenotype (approximately 90%), and
the remaining cells are CD56brightCD16– (approximately
10%). The majority of uterine NK cells (approximately
90%) are CD56brightCD16–. In the uterine decidua, uNK
cell numbers cyclically increase and decrease in tandem
with the menstrual cycle—low in the proliferative phase
(10% to 15%), which amplifies during the early, middle
and late secretory phases (25% to 30%)—falling to a
basal level with menstruation (Figure 5-7) (Kalkunte et al,
2008a; Kitaya et al, 2007).
With successful implantation, the uNK cell population further increases in the decidualized endometrium,
reaches a peak in first-trimester pregnancy, and dwindles
thereafter by the end of the second trimester. The origin
of uNK cells that peak during the secretory phase of the
menstrual cycle and early pregnancy is currently not well
established, and the evidence indicates multiple different
possibilities. These possibilities include recruitment of
CD56brightCD16– pNK cells, recruitment and tissue specific terminal differentiation of CD56dimCD16+ pNK cells,
development of NK cells from Lin–CD34+CD45+ progenitor cells, or proliferation of resident CD56brightCD16–
NK cells. Comparative surface expression of antigens,
natural cytotoxicity receptors, inhibitory receptors, and
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
A
B
C
D
E
F
G
H
I
J
K
L
43
FIGURE 5-6 (Supplemental color version of this figure is available online at www.expertconsult.com.) Differential endovascular activity of first- and thirdtrimester trophoblasts in response to normal pregnancy serum. A representative micrograph of trophoblasts-endothelial cell interactions on matrigel
is shown. Endothelial cells and trophoblasts are labeled with red and green cell tracker respectively, were independently cultured (A to E) or cocultured
(F to I) on matrigel. Capillary-like tube structures were observed with human uterine endothelial cells (HUtECs) (A) and umbilical vein endothelial cells
(HUVECs) (B), but not with first-trimester trophoblast HTR8 cells (C), third trimester trophoblast TCL1 cells (D), and primary term trophoblasts (E).
However, in cocultures, HTR8 cells fingerprint the HUtECs (F) and HUVECs (G), while TCL1 cells (H) and primary term trophoblasts (I) inhibit the
tube formation by endothelial cells (magnification ×4). Panels J to L show the cocultures of HTR8 with HUVECs ( J), HUtECs ( K), and term trophoblasts
with HUVECs (L) at higher magnification (×10). (Reproduced with permission from Kalkunte S, Lai Z, Tewari N, et al: In vitro and in vivo evidence for lack of
endovascular remodeling by third trimester trophoblasts, Placenta 29:871-878, 2008.)
TABLE 5-3 Comparison of Peripheral Blood and Decidual
Immune Cell Profiles
Immune Cells
T cells
Peripheral
blood (%)
Decidua (%)
65-70
9-12
γδT cells
2-5
7-10
Macrophages
7-10
15-20
B cells
7-10
ND
NKT cells
2-5
0.5-1.0
Tregs
2-4
6-10
NK cells
7-12
65-70 (CD56brightCD16-)
ND, Not detected.
chemokines and cytokines on human pNK and uNK cells
are provided in Table 5-4. Furthermore, CD56bright uNK
cells are different from the CD56bright minor population
of pNK cells because of the expression of CD9, CD103,
and killer immunoglobulin-like receptors (KIRs). Despite
being replete with cytotoxic accessories of perforin, granzymes A and B and the natural cytotoxicity receptors
NKp30, NKp44, NKp46, NKG2D, and 2B4 as well as
LFA-1, uNK cells are tolerant cytokine-producing cells at
the maternal-fetal interface (Kalkunte et al, 2008a). The
temporal occurrence around the spiral arteries and timed
amplification of these specialized uNK cells observed during the first trimester implicate its role in spiral artery
remodeling.
NK cell–deficient mice display abnormalities in decidual artery remodeling and trophoblast invasion, possibly
because of a lack of uNK cell–derived interferon γ (Ashkar
et al, 2000). Other studies have shown that unlike pNK
cells, uNK cells are a major source of VEGF-C, Angiopoietins 1 and 2 and transforming grwoth factor (TGFβ1) within the placental bed that decrease with gestational
age (Lash et al, 2006). These observations implicate uNK
cells in promoting angiogenesis. Studies have provided
further evidence that uNK cells, but not pNK cells, regulate trophoblast invasion both in vitro and in vivo through
the production of interleukin-8 and interferon-inducible
44
PART II Fetal Development
LH
P4
E2
NK cell
population
KIR
NKG2D
NKp44
NK
NKp46
NKp30
Inactive
phase
Proliferative
phase
~30%
~50–60%
Secretory
phase
First
trimester
Second
trimester
Term
Menstrual cycle
Pregnancy
FIGURE 5-7 Biologic pattern of natural killer (NK) cells in the human endometrium and the decidua. The uterine NK cell population
characterized by natural cytotoxicity receptors (Nkp30, NKp44, NKp46, NKG2D), killer immunoglobulin-like receptors, and cytolytic machinery
(perforin and granzyme) cyclically increases and decreases in tandem with the hormonal changes during menstrual cycle. With successful implantation, uterine NK cells further increase in the decidua and dwindle thereafter by the end of second trimester. E2, Estradiol; LH, luteinizing hormone;
P4, progesterone.
protein-10, in addition to other angiogenic factors (Hanna
et al, 2006). Recent studies suggest that VEGF-C, a proangiogenic factor produced by uNK cells, is responsible
for the noncytotoxic activity (Kalkunte et al, 2009). As
noted previously, VEGF-C–producing uNK cells support
endovascular activity in a coculture model of capillary tube
formation on matrigel (Figure 5-8). Peripheral blood NK
cells fail to produce VEGF-C and remain cytotoxic. This
function can be reversed by recombinant human VEGFC. Cytoprotection by VEGF-C is related to induction of
the transporter associated with antigen processing 1 and
MHC assembly in target cells. Overall, these findings suggest that expression of angiogenic factors by uNK cells
keeps these cells noncytotoxic, which is critical to their
pregnancy compatible immunovascular role during placentation and fetal development (Kalkunte et al, 2009).
Although uNK cells seem to play a role that is compatible with pregnancy, retention of their cytolytic abilities
suggests their role as sentinels at the maternal-fetal interface in situations that threaten fetal persistence. This facet
of uNK cell function was elegantly demonstrated in animal
models when pregnant mice were challenged with tolllike receptor (TLR) ligands that mimic bacterial and viral
infections. These observations raise an important question
whether uNK cells can harm the fetal placental unit and, if
so, under what conditions?
The antiinflammatory cytokine interleukin (IL)-10
plays a critical role in pregnancy because of its regulatory
relationship with other intrauterine modulators and its
wide range of immunosuppressive activities (Moore et al,
2001). IL-10 expression by the human placenta depends
on gestational age, with significant expression through the
second trimester followed by attenuation at term (Hanna
et al, 2000). IL-10 expression is also found to be poor in
decidual and placental tissues from unexplained spontaneous abortion cases (Plevyak et al, 2002) and from deliveries
associated with preterm labor (Hanna et al, 2006) and preeclampsia (S. Kalkunte et al, unpublished observations).
However, the precise mechanisms by which IL-10 protects
the fetus remains poorly understood. IL-10–/– mice suffer
no pregnancy defects when mated under pathogen-free
conditions (White et al, 2004), but they exhibit exquisite
susceptibility to infection or inflammatory stimuli compared with wild type animals. It is then plausible that in
addition to IL-10 deficiency, a “second hit” such as an
inflammatory insult resulting from genital tract infections, environmental factors, or hormonal dysregulation
during gestation can lead to adverse pregnancy outcomes (Tewari et al, 2009; Thaxton et al, 2009). Our
recent studies provide direct evidence that uNK cells can
become adversely activated and mediate fetal demise and
preterm birth in response to low doses of TLR ligands
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
TABLE 5-4 Phenotypic Characteristics of Surface Markers
and Receptors on Natural Killer Cells
Antigen
CD56
Peripheral blood
Decidua
Dim (>90%)
Bright
CD16
+
–
CD45
+
+
+
CD7
+
CD69
–
+
L-Selectin
–
–/+
NK Receptors
KIR
+
+
NKp30
+
+
NKp44
–*
+
NKp46
+
+
NKG2D
CD94/NKG2A
+
+
–/+
+
+
Chemokine Receptors
CXCR1
+
CXCR2
+
–
CXCR3
–
+
+
CXCR4
+
CX3CR1
+
–
CCR7
–
+
Data from Kalkunte S, Chichester CO, Sentman CL, et al: Evolution of non-cytotoxic
uterine natural killer cells, Am J Reprod Immunol 59:425-432, 2008.
+, Present; –, absent; –/+, variable expression; KIR, killer immunoglobulin receptor;
CXCR, CX-chemokine receptor; CX3CR1, CX3-C–chemokine receptor 1; CCR7, CCchemokine receptor 7.
*Expression seen on activation with interleukin 2.
resulting in placental pathology (Murphy et al, 2005;
Murphy et al, 2009). Moreover, spontaneous abortion is
associated with an increase in CD56dimCD16+ cells and a
decrease in CD56brightCD16– NK cells in the preimplantation endometrium during the luteal phase (Michimata
et al, 2002; Quenby et al, 1999). Therefore a fine balance between maternal activating and inhibitory KIRs
and their ligand HLA-C on fetal cells seems to be maintained in normal pregnancy. Insufficient inhibition of
uNK cells can activate the cytolytic machinery, resulting
in spontaneous abortion, intrauterine growth restriction,
or preterm labor, depending on the timing of the insult
(Varla-Leftherioti et al, 2003). In the setting of IVF, the
implantation failure has been associated with high uNK
cell numbers, but direct evidence for their role in abnormal implantation is not clear (Quenby et al, 1999). Nevertheless current understanding strongly implies that uNK
cells retain the ability to become foes to pregnancy under
the axis of genetic stress and inflammatory trigger.
REGULATORY T CELLS AND PREGNANCY
The existence of regulatory mechanisms that suppress the
maternal immune system was proposed in the early 1950
(Medawar, 1953). For several years, maternal tolerance
toward fetal alloantigens was explored in the context of
Th1/Th2 balance, with Th2 cells and cytokines proposed
45
to predominate over Th1 cellular immune response under
normal pregnancy. Recently the role of specialized T lymphocytes, termed regulatory T cells (Tregs), in tolerogenic
mechanisms has emerged. Tregs are potent suppressors
of T cell–mediated inflammatory immune responses and
prevent autoimmunity and allograft rejection. Tregs act
by controlling the autoreactive T cells that have escaped
negative selection from the thymus, and they restrain the
intensity of responses by T cells reactive with alloantigens
and other exogenous antigens. This unique functional capability to suppress responses to tissue-specific self-antigens
that escape recognition by T cells during maturation is due
to tissue specific expression and alloantigens, particularly in
the epithelial surfaces where tolerance to nondangerous foreign antigen is essential to normal function. This capability
enables Tregs to play a unique role at the maternal-fetal
interface. Tregs are typically characterized by a CD4+CD25+
surface phenotype and expression of the hallmark suppressive transcription factor Foxhead Box P3 (Foxp3+). Their
cell numbers increase in blood, decidual tissue, and lymph
nodes draining the uterus during pregnancy. These cells are
implicated in successful immune tolerance of the conceptus,
mainly by producing IL-10 and TGF-β. Recent evidence
suggests that fetal Tregs also play a vital role in suppressing fetal antimaternal immunity against maternal cells that
cross the placenta (Mold et al, 2008).
In the absence of Tregs the allogeneic fetus is rejected,
suggesting their critical role in normal pregnancy. Unexplained infertility, spontaneous abortion, and preeclampsia
are associated with proportional deficience, functional Treg
deficiency, or both. In the context of pregnancy, the local
milieu, particularly during the first trimester, that includes
hCG, TGF-β, IL-10, granulocyte-macrophage colonystimulating factor, and indoleamine 2,3-dioxygenase expression has now been shown to induce CD4+CD25+ Tregs
with Foxp3 expression with immunosuppressive features.
This induction is thought to occur through the immature
dendritic cells. In addition to immune suppressive and antiinflammatory properties, TGF-β is recognized as inducing
differentiation of naïve CD4 T cells into suppressor T-cell
phenotype, expressing Foxp3, and promoting the proliferation of mature Tregs. In addition to the suppressive effects of
cytokines produced by these cells, contact-mediated immune
suppression by Tregs results from ligation of inhibitory
cytotoxic T-lymphocyte antigen (CTLA-4) and its ability to
induce tolerogenic dendritic cells and influence T-cell production of IL-10 (Aluvihare et al, 2004; Schumacher et al,
2009; Shevach et al, 2009). Therefore the pregnant uterus
may be a natural depot for Tregs.
EPIGENETIC REGULATION
IN THE PLACENTA
The regulation of gene expression is a crucial process that
defines phenotypic diversity. Switching off or turning on
genes as well as tissue-specific variation in gene expression
contributes to this diversity. Besides the genetic make-up (i.e,
sequence) of the individual, the regulation of gene expression
is also influenced by epigenetic factors. Epigenetic changes
include the noncoding changes in DNA and chromatin,
or both, that mediate the interactions between genes and
their environment. Epigenetic regulation generates a wider
46
PART II Fetal Development
DECIDUA
Cytotoxic NK cells
Non-cytotoxic NK cells
CD56
CD56brightCD16
VEGF C
CD16
CD56dimCD16
Intrauterine infections,
Inflammation,
TLR activation,
Loss of IL-10
Promotes endovascular activity
Upregulates TAP-1 and MHC
molecules on trophoblats
Efficient trophoblast invasion
Poor endovascular activity
Trophoblast lysis
Poor trophoblast invasion
Normal pregnancy
Adverse pregnancy
FIGURE 5-8 (See also Color Plate 1.) Angiogenic features of natural killer (NK) cells render them immune tolerant at the maternal-fetal interface.
Vascular endothelial growth factor (VEGF) C–producing noncytotoxic uterine NK cell clones similar to decidual NK cells support endovascular
activity in a coculture of endothelial cells and first-trimester trophoblast HTR8 cells on matrigel. By contrast, cytotoxic uterine NK cell clones similar
to peripheral blood NK cells disrupted the endovascular activity because of endothelial and trophoblast cell lysis. This distinct functional feature
determines whether optimal trophoblast invasion takes place and can result in normal or adverse pregnancy outcomes.
diversity of cell types during mammalian development and
sustains the stability and integrity of the expression profiles
of different cell types and tissues. This regulation is choreographed by changes in cytosine-phosphate-guanine (CpG)
islands of the DNA promoter region by methylation, histone modification, genomic imprinting, and expression of
noncoding RNAs such as micro RNA (miRNA).
Gene-environment interactions resulting in epigenetic
changes in the placenta during the critical window of development can influence fetal programming in utero, with
predisposing health consequences later in life. Using a microarray-based approach to compare chorionic villous samples
from the first trimester of pregnancy with gestational age–
matched maternal blood cell samples, recent studies show
tissue-specific differential CpG methylation patterns that
identify numerous potential biomarkers for the diagnosis
of fetal aneuploidy on chromosomes 13, 18 and 21 (Chu
et al, 2009). Human placentation displays many similarities
with tumorigenesis, including rapid mitotic cell division,
migration, angiogenesis and invasion, and escape from
immune surveillance. Indeed, there are striking similarities
in the DNA methylation pattern of tumor-associated genes
between invasive trophoblast cell lines and first-trimester
placenta and tumors (Christensen et al, 2009). This finding
suggests that a distinct pattern of tumor-associated methylation can result in a series of epigenetic silencing events
necessary for normal human placental invasion and function (Novakovic et al, 2008). Other studies using the placenta as a source suggest that the specific loss of imprinting
because of altered methylation and subsequent gene expression can result in small for gestational age (SGA) newborns.
Moreover, unbalanced expression of imprinted genes in
IUGR placenta when compared with non-IUGR placenta
was observed suggesting a differential expression pattern of
imprinted genes as a possible biomarker for IUGR (Guo
et al, 2008; McMinn et al, 2006).
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
The unique cytokine and hormonal milieu in utero may
influence the trophoblast function and differentiation as
well as immune cell regulation through histone posttranslational modification. In this regard, interferon γ produced
by uNK cells and essential for spiral artery remodeling
fails to induce MHC class II expression in trophoblast
cells because of hypermethylation of regulatory class II
MHC transactivator (CIITA) regions (Morris et al, 2002).
This inability to upregulate classical MHC class II molecules by trophoblasts is essential for maintaining immune
tolerance at the maternal-fetal interface. Moreover, the
transcription factor regulating trophoblastic fusion protein syncytin, which is essential for the syncytialization of
trophoblasts, is regulated by histone acetyl transferase and
histone deacetylase activity (Chuang et al, 2006). miRNAs
are small regulatory RNA molecules that can alter gene
and protein expression without altering the underlying
genetic code. Expression of miRNA is tissue specific, and
several are expressed in the placenta. In placental pathology associated with preeclampsia, there is differential
expression of miRNA (such as miR-210 and miR-182)
compared with normal pregnancy placenta. This finding
suggests that signature differences in placental miRNA
and their detection in maternal serum may potentially be
used as a biomarker for preeclampsia. Because implantation and early placentation is under the regulation of low
oxygen tension, it is possible that miRNA are differentially
expressed under different oxygen levels, as suggested by
recent observations (Maccani and Marsit, 2009; Pineles
et al, 2007).
PLACENTAL DISEASES
The placenta provides a wealth of retrospective information about the fetus and prospective information regarding
the infant. Healthy development of the placenta requires
efficient metabolic, immune, hormonal, and vascular
adaptation by the maternal system as well as the fetus.
Abnormal placentation and placental infections can lead
to maternal or fetal anomalies as seen in preeclampsia,
preterm birth, and SGA, which can have lifelong bearing
on the development and health of infants. Maternal factors such as ascending infections, obesity, hypertension,
genetic predisposition such as gene polymorphism of the
pregnancy-compatible cytokine milieu, and environmental
exposure could also contribute to the placental pathology.
The following sections contain an abbreviated discussion
of the pathogenesis of some of these placenta-associated
disorders.
PREECLAMPSIA
Hypertensive disorders of pregnancy are enigmatic. They
pose a major public health problem and affect 5% to 10%
of human pregnancies. Preeclampsia is clinically associated with maternal symptoms of hypertension, proteinuria, and glomeruloendotheliosis. This disorder is strictly
a placental condition because of its clearance after delivery. It causes morbidity and mortality in the mother, fetus,
and newborn. Pregnancy-associated hypertension is defined
as blood pressure greater than 140/90 mm Hg on at least
two occasions and at 4 to 6 weeks apart after 20 weeks’
47
gestation. Proteinuria is defined by excretion of 300 mg
or more of protein every 24 hours or 300 mg/L or more
in two random urine samples taken at least 4 to 6 hours
apart (ACOG Committee on Practice Bulletins, 2002).
The fetal problems most commonly associated with preeclampsia include fetal growth restriction, reduced amniotic fluid, and abnormal oxygenation (Sibai et al, 2005).
However, the onset of clinical signs and symptoms can
result in either near-term preeclampsia without affecting the fetus or its severe manifestation that is associated
with low birthweight and preterm delivery (Vatten and
Skjaerven, 2004). The heterogeneous manifestation of
this disease is further confounded by preexisting maternal
vascular disease, multifetal gestation, metabolic syndrome,
obesity, or previous incidence of the disease. In addition,
the pathophysiology of the disorder could differ from the
onset before 24 weeks’ gestation and its diagnosis at later
stages of pregnancy:
Abnormal remodeling of spiral arteries and shallow trophoblast invasion are two hallmark features of preeclampsia. Preeclampsia is considered a two-stage disease where
a poorly perfused placenta (stage I) causes the release of
factors leading to maternal symptoms (stage II). However,
it is also now being recognized that the maternal factors
may contribute to programming of stage I of preeclampsia, suggesting that the intrinsic maternal factors stemming from genetic, behavioral, and physiologic conditions
may contribute to placental pathology. Stage I initiated
pathology may be particularly apparent in the oxidative
stress-induced release of causative factors from the poorly
perfused placenta and their effects on the maternal syndrome (Roberts and Hubel, 1999).
Despite a poor mechanistic understanding of placental
pathology leading to preeclampsia, several critical features
are common to this disease. Multiple studies have shown
that reduced vascular activity could be a major factor contributing to preeclampsia. In normal pregnancy, the circulating PlGF levels steadily increase in the first and second
trimesters, peak at 29 to 32 weeks, and decline thereafter.
However, free VEGF remains low and unchanged during this window. Reduced placental expression of VEGF
and PlGF is consistently observed in preeclampsia. Furthermore, preeclampsia is frequently accompanied by
enhanced circulation and placental expression of the antiangiogenic soluble VEGF receptor 1 (sFlt-1), which is a
decoy receptor titrating out VEGFs and PlGF (Levine
et al, 2004; Romero et al, 2008; Thadhani et al, 2004).
A lack of available VEGF and increased sFlt-1 expression has been associated with trophoblast injury. The
soluble form of endoglin (CD105), a coreceptor involved
in TGF-β signaling is reported to enhance the antiangiogenic effects of sFlt-1. Soluble endoglin has been found
to be elevated in the serum of preeclamptic women and is
accompanied by an increased ratio of sFlt-1:PlGF and correlates with the severity of the disease. Soluble endoglin is
thought to inhibit TGF-β1 signaling in endothelial cells
and blocks activation of endothelial nitric oxide synthase
and vasodilatation (Venkatesha et al, 2006). Several recent
studies have suggested an increase in apoptosis within villous trophoblast from preeclampsia and IUGR deliveries
(Allaire et al, 2000; Heazell and Crocker, 2008; Levy et al,
2002). Unlike normal pregnancy, villous placental explants
48
PART II Fetal Development
from preeclamptic placenta have an increased sensitivity and susceptibility to apoptosis on exposure to proinflammatory cytokines, suggesting altered programming
of apoptotic cascade pathway (Crocker et al, 2004; Levy
et al, 2002). It is possible that incomplete spiral artery
transformation resulting in reduced placental perfusion
(stage I) in preeclampsia leads to focal regions of hypoxia
with increase in apoptosis, oxidative stress, shedding of
villous microparticles, and release of antiangiogenic factors such as sFlt-1.64 (Hung et al, 2002; Nevo et al, 2006;
Redman and Sargent, 2000).
Another pathway that may contribute to the etiology of
preeclampsia is unscheduled and excessive activation of the
complement cascade; this is highly likely as a result of the
maternal immune system responding to paternal antigens
and inflammation. However, in normal pregnancy the
placenta expresses complement regulatory proteins such
as DAF, CD55, and CD59 and may control activation of
complement factors (Tedesco et al, 1993). Despite the
positioning of complement inhibitory proteins for protective roles, increasing evidence supports the involvement of
complement activation in the pathogenesis of preeclampsia (Lynch et al, 2008). Interestingly, recent in vitro studies
suggest that hypoxia enhances placental deposition of the
membrane attack complex and apoptosis in cultured trophoblasts (Rampersad et al, 2008). The upstream factors
that trigger complement activation are not yet known.
Recent studies also suggest increased serum levels
of agonistic autoantibodies against angiotensin type 1
receptor (AT-1-AA) in preeclampsia as compared with
healthy women (Zhou et al, 2008). Importantly, studies
from our laboratory have shown that the full spectrum of
preeclampsia-like symptoms can be reproduced in mice
by injecting human preeclampsia serum containing subthreshold levels of AT-1-AA immunoglobulin G, suggesting that pregnancy serum contains some unknown
causative factors. Therefore serum can be used as a blueprint to identify functional biomarkers for preeclampsia
(Kalkunte et al, 2009).
PRETERM BIRTH
Preterm birth is the leading cause of infant morbidity
and mortality in the world. Babies born before 37 weeks’
gestation are considered premature. In the United States,
approximately 12.8% of births are preterm, and the rate
of premature birth has increased by 36% since early 1980s
(Martin et al, 2009). Babies from preterm birth face an
increased risk of lasting disabilities such as mental retardation, learning and behavioral problems, autism, cerebral
palsy, bronchopulmonary dysplasia, vision and hearing
loss, and risk for diabetes, hypertension, and heart disease
in adulthood. The majority of preterm deliveries are due
to preterm labor. Other factors leading to premature birth
are preterm premature rupture of membranes (PPROM),
intervention for maternal or fetal problems, preeclampsia,
fetal growth restriction, cervical incompetence, and antepartum bleeding. Additional risk factors for preterm birth
include stress, occupational fatigue, uterine distention by
polyhydramnios or multifetal gestation, systemic infection such as periodontal disease, intrauterine placental
pathology such as abruption, vaginal bleeding, smoking,
substance abuse, maternal age (<18 or >40 years), obesity,
diabetes, thrombophilia, ethnicity, anemia, and fetal factors such as congenital anomalies and growth restriction.
Activation of the hypothalamic-pituitary-adrenal (HPA)
as a result of major maternal physical or psychological
stress is thought to increase the release of corticotrophinreleasing hormone. In addition to the hypothalamus as a
source of corticotrophin-releasing hormone, placental
trophoblasts, amnion, and decidual cells also express this
hormone during pregnancy. Corticotrophin hormone
regulates the release of adrenocorticotropic hormone
from pituitary and cortisol from adrenal glands, and it
can also influence the activity of matrix metalloproteinases (MMPs). Premature activation of the HPA axis can
eventually stimulate the prostaglandins, ultimately resulting in parturition via activation of proteases. In addition,
activation of the HPA axis promotes the release of estrone,
estradiol, and estriol that can activate the myometrium by
increasing oxytocin receptors, prostaglandin activity, and
enzymes such as myosin light chain kinase and calmodulin, which are responsible for muscle contraction. Concomitantly, progesterone withdrawal is expected with the
raising concentration of myometrial estrogen receptors,
further enhancing estrogen-induced myometrial activation and preterm birth (Dole et al, 2003; Grammatopoulos
and Hillhouse, 1999; McLean et al, 1995).
There is increasing evidence that approximately 50% of
preterm births are associated with infection of the decidua,
amnion, or chorion and amniotic fluid caused by either
systemic or ascending genital tract infection. Both clinical
and subclinical chorioamnionitis are implicated in preterm
birth. Maternal or fetal inflammatory responses to chorioamniotic infection can trigger preterm birth. Activated
neutrophils and macrophages and the release of cytokines
IL-1β, IL-6, IL-8, tumor necrosis factor alpha (TNF-α)
and granulocyte colony-stimulating factor can lead to an
enhanced cascade of signaling activity, causing release of
prostaglandins and expression of various MMPs of fetal
membranes and the cervix. Furthermore, elevated levels
of TNF-α and apoptosis are associated with PPROM.
Non–infection-related inflammation caused by placental
insufficiency and apoptosis can also cause preterm birth.
In addition to augmented inflammatory responses to
infections, pathogenic microbes (e.g. Staphylococcus, Streptococcus, Bacteroides, and Pseudomonas spp.) are thought to
directly degrade fetal membranes by releasing proteases,
collagenases, and elastases, produce phospholipase A2,
and release endotoxin that stimulate uterine contractions
and cause preterm birth (Goldenberg et al, 2000, 2008;
Romero et al, 2006; Slattery and Morrison, 2002).
The innate immune system and trophoblasts during
pregnancy recognize bacterial and viral infections using
TLRs. Placental transcripts for TLRs 1 to 10 have been
detected in human placental tissue, and placental choriocarcinoma cell lines reportedly express TLR-2, TLR-4,
and TLR-9 (Abrahams and Mor, 2005). Studies have demonstrated functionality for TLR-2, TLR-3, and TLR-4
in first- and third-trimester placental tissue (Patni et al,
2007). Decidual expression in humans has demonstrated
functional receptors in term decidua of TLR-1, TLR-2,
TLR-4, and TLR-6 (Canavan and Simhan, 2007). Our
recent studies using mice have shown that extremely small
CHAPTER 5 Immunologic Basis of Placental Function and Diseases: the Placenta, Fetal Membranes, and Umbilical Cord
doses of the TLR-4 ligand lipopolysaccharide can cause
preterm birth or fetal demise in pregnant IL-10–deficient
mice by activating and promoting infiltration of uterine
NK cells into the placenta and inducing apoptosis by
secretion of TNF-α (Murphy et al, 2005, 2009). Similarly,
activation of TLR-3 or TLR-9 has been shown to induce
spontaneous abortion or preterm birth in IL-10–deficient
pregnant mice that is attributed to immune infiltration and
proinflammatory cascade in the placenta (Thaxton et al,
2009).
Decidual hemorrhage leading to vaginal bleeding
increases the risk for preterm birth and PPROM. Increased
occult decidual hemorrhage, hemosiderin deposition, and
retrochorionic hematoma formation is seen between 22
and 32 weeks’ gestation as a result of PPROM and preterm
birth after preterm labor. The development of PPROM in
the setting of abruption could be caused by high decidual
concentration of tissue factors, which eventually generate thrombin. Thrombin activation as measured by serum
thrombin-antithrombin III complex levels are elevated
on preterm birth. Thrombin binds to decidual protease-
activated receptors (PAR1 and PAR2), induces the production of IL-8 in decidua, attracts neutrophils, and promotes
degradation of the fetal membrane MMPs that can result
in PPROM (Lockwood et al, 2005; Salafia et al, 1995).
Polyhydramnios is also a high risk factor for preterm
birth. It was shown recently that exposure of IL-10–
deficient pregnant mice to polychlorinated biphenyls, an
environmental toxicant, can lead to preterm birth with
IUGR. The IUGR was due to increased amniotic fluid
volume (polyhydramnios) and placental insufficiency
caused by poor spiral artery remodeling associated with
reduced expression of water channel aquaporin-1 in the
placenta (Tewari et al, 2009). Increasing evidence also suggests impaired vascular activity because of an increase in
antiangiogenic factors such as sFlt-1 and decreased VEGF
in PPROM and preterm birth (Kim et al, 2003).
INTRAUTERINE GROWTH RESTRICTION
IUGR is used to designate a fetus that has not reached
its growth potential; it can be caused by fetal, placental,
or maternal factors. Disparities between fetal nutritional
or respiratory demands and placental supply can result
in impaired fetal growth. Chromosomal abnormalities
(aneuploidy, partial deletions, gene mutation particularly on the gene for insulin-like growth factors), congenital abnormalities, multiple gestation, and infections
can also result in IUGR. Preterm birth, preeclampsia,
and abruption because of placental ischemia can result in
IUGR. Reduced placental weight with identifiable placental histologic abnormalities (e.g, impaired development or obstruction in uteroplacental vasculature, chronic
abruption, chronic infections, maternal floor infarction,
thrombosis in uteroplacental vasculature or fetoplacental vasculature) are common findings in IUGR. In addition, a single umbilical artery, velamentous umbilical cord
insertion, bilobate placenta, circumvallate placenta, and
placental hemangioma are some of the other structural
anomalies seen in the placenta. Maternal factors such as
nutritional deficiency; severe anemia; pulmonary disease
leading to maternal hypoxemia; smoking; exposure to
49
toxins such as warfarin, anticonvulsants, folic acid antagonists, and caffeine; and pregnancies conceived through
assisted reproductive techniques have a higher prevalence
of IUGR. IUGR results in the birth of an infant who is
SGA. Mortality and morbidity are increased in SGA
infants compared with those who are appropriate for gestational age. SGA infants at birth have many clinical problems that include impaired thermoregulation; difficulty in
cardiopulmonary transition with perinatal asphyxia, pulmonary hypertension, hypoglycemia, polycythemia and
hyperviscosity; impaired cellular immune function; and
increased risk for perinatal mortality. SGA infants in their
childhood and adolescence are at higher risk for impaired
physical growth and neurodevelopment. Adolescents
born SGA at term were reported to have learning difficulties with attention deficits. Cognitive performance is
generally lower in SGA infants at the ages of 1 to 6 years
compared with those whoe are appropriate for gestational
age. Adults who were SGA infants could be at higher risk
for ischemic heart diseases and essential hypertension
(Figueras et al, 2007; Kaijser et al, 2008; Lapillonne et al,
1997; Norman and Bonamy, 2005; O’Keefe et al, 2003;
Spence et al, 2007).
FETAL MEMBRANES AND THEIR
PATHOLOGY
The fetal tissue–derived membrane structure surrounds
the fetus and forms the amniotic cavity. This membrane,
which lacks both vascular and nerve cells, is composed of
an inner layer adjacent to the amniotic fluid and is called
the amnion. The outer layer that is attached to the decidua
is called the chorion. Amnion is composed of inner epithelial cells, and the mesenchymal cell layer is composed of
fibroblast and an outer spongy layer. Intact, healthy fetal
membranes are required for normal pregnancy outcome.
Chorion is composed of an outer reticular cell layer composed of fibroblasts and macrophages and an inner cytotrophoblast layer. The elasticity and strength of these
membranes are maintained by extracellular matrix proteins
such as collagens, fibronectin, laminins, and the activity of
MMP-2 and MMP-9 and their inhibitors until the initiation of parturition when the membranes are susceptible
to rupture. During parturition, when contractions begin
or membranes rupture, MMP activity in the amnion and
chorion increases with a concurrent fall in tissue inhibitors
of metalloproteinases. This change is followed by apoptosis in the amnion epithelial and chorion trophoblast layers
of fetal membrane. Interestingly, some evidence suggests
that fetal membranes have antimicrobial activity and are
known to express TLR-2 and TLR-4, which are pattern
recognition receptors and help in initiating a protective
host response to infection.
The histopathology of amnion and chorion includes
infections, amniotic fluid contaminants, and fetal diseases.
In addition to the membranes, whose infection can lead to
chorioamnionitis, another vulnerable portal for infection
to occur is the placental intervillous space and fetal villi
that provide hematogenous access. Hematogenous sources
of infection are typically associated with inflammation of
villi (villitis) and intervillous space (intervillositis). Viral
pathogens (cytomegalovirus, HIV, herpes simplex virus)
50
PART II Fetal Development
commonly produce hematogenous infection of the placenta in addition to bacteria, spirochetes, fungi, and protozoa (Gersell, 1993; Goldenberg et al, 2000; Lahra and
Jeffery, 2004).
UMBILICAL CORD
The connecting cord from the developing embryo or fetus
to the placenta is the umbilical cord, or funiculus umbilicalis. During prenatal development in humans, the normal umbilical cord contains two umbilical arteries and
one umbilical vein buried within Wharton’s jelly. The
umbilical vein supplies the fetus with oxygenated blood
from the placenta while the arteries return the deoxygenated, nutrient-depleted blood to the placenta. In the fetus,
the umbilical vein branches into the ductus venosus and
another branch that joins the hepatic portal vein. Shortly
after parturition, physiologic processes cause the Wharton’s jelly to swell with the collapse of blood vessels, resulting in a natural halting of the flow of blood. Within the
infant, the umbilical vein and ductus venosus close and
degenerate into remnants known as the round ligament of
the liver and the ligamentum venosum, while the umbilical
arteries degenerate into what is known as medial umbilical
ligaments.
Abnormalities associated with the umbilical cord can
affect both the mother and the child. Pathology of umbilical cord is generally grouped as congenital remnants,
infections, meconium, and masses. Abnormalities that
have clinical significance are nuchal cord, single umbilical artery, umbilical cord prolapse, umbilical cord knot,
umbilical cord entanglement, vasa previa, and velamentous cord insertion. Common intrauterine infections can
result in the umbilical cord being invaded by fetal cells
and bacteria infiltrated from the decidua to amniotic fluid,
or they can elicit fetal inflammatory response. Umbilical
cord inflammation, known as funisitis or vasculitis, poses a
higher risk for development of neurologic compromise in
the fetus. Funisitis is predictive of a lower median Bayley
psychomotor developmental index in infants. Meconium
pigment at high concentrations can damage the umbilical cord by triggering apoptosis of smooth muscle cells.
Vascular necrosis caused by meconium is associated with
oligohydramnios, low Apgar scores, and significant neurodevelopmental delay. Interruption of normal blood flow in
the cord can cause prolonged hypoxia in utero. Clamping
of the umbilical cord within minutes of birth is hospitalbased obstetric practice. A Cochrane review studying the
effects of the timing of umbilical cord clamping in hospitals showed that infants whose cord clamping occurred
later than 60 seconds after birth had a significantly higher
risk of neonatal jaundice requiring phototherapy. However, randomized, controlled studies have shown that
delayed cord clamping in preterm infants reduces the
incidence of intraventricular hemorrhage and late-onset
sepsis. Furthermore, premature clamping can increase the
risk of ischemia and hypovolemic shock, which can lead
to fetal complications (McDonald and Middletone, 2008;
Mercer et al, 2006).
SUGGESTED READINGS
Aluvihare VR, Kallikourdis M, Betz AG: Regulatory T cells mediate maternal
tolerance to the fetus, Nat Immunol 5:266-271, 2004.
Ashkar AA, Di Santo JP, Croy BA: Interferon γ contributes to initiation of uterine
vascular modification, decidual integrity, and uterine natural killer cell maturation during normal murine pregnancy, J Exp Med 192:259-269, 2000.
Baker DJP: In utero programming of chronic disease, Clin Sci 95:115-128, 1998.
Christensen BC, Houseman EA, Marsit CJ, et al: Aging and environmental
exposures alter tissue-specific DNA methylation dependent upon CpG island
context, PLoS Genet 5:e1000602, 2009.
Goldenberg RL, Culhane JF, Iams JD, et al: Epidemiology and causes of preterm
birth, Lancet 371:75-84, 2008.
Hanna J, Goldman-Wohl D, Hamani Y, et al: Decidual NK cells regulate key
developmental processes at the human fetal-maternal interface, Nat Med
12:1065-1074, 2006.
Kalkunte S, Mselle TF, Norris WE, et al: VEGF C facilitates immune tolerance
and endovascular activity of human uterine NK cells at the maternal-fetal
interface, J Immunol 182:4085-4092, 2009.
Moffett A, Loke C: Immunology of placentation in eutherian mammals, Nat Rev
Immunol 6:584-594, 2006.
Murphy SP, Hanna NN, Fast LD, et al: Evidence for participation of uterine
natural killer cells in the mechanisms responsible for spontaneous preterm
labor and delivery, Am J Obstet Gynecol 200:308, 2009.
Paria BC, Reese J, Das SK, et al: Deciphering the cross-talk of implantation:
advances and challenges, Science 296:2185-2188, 2002.
Roberts JM, Hubel CA: Is oxidative stress the link in the two–stage model of preeclampsia? Lancet 354:788-789, 1999.
Slattery MM, Morrison JJ: Preterm delivery, Lancet 360:1489-1497, 2002.
Thadhani R, Sachs BP, Epstein FH, et al: Circulating angiogenic factors and the
risk of preeclampsia, N Engl J Med 350:672-683, 2004.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
6
Abnormalities of Fetal Growth
Rebecca Simmons
Fetal growth and size at birth are critical in determining
mortality and morbidity, both immediately after birth and
in later life. Normal fetal growth is determined by a number of factors, including genetic potential, the ability of the
mother to provide sufficient nutrients, the ability of the
placenta to transfer nutrients, and intrauterine hormones
and growth factors. The pattern of normal fetal growth
involves rapid increases in fetal weight, length, and head
circumference during the last half of gestation. During
the last trimester, the human fetus accumulates significant
amounts of lipid. The birthweight for gestational measurements among populations has been shown to increase
over time; therefore, standards for normal fetal growth
require periodic reevaluation for clinical relevance. These
increases in birthweight for gestational age over time
are attributed to improvements in living conditions and
maternal nutrition and changes in obstetric management.
Variations in fetal growth have been identified in diverse
populations and are associated with geographic locations
(sea level versus high altitude), populations (white, African
American, Latino), maternal constitutional factors, parity,
maternal nutrition, fetal gender, and multiple gestations.
In this chapter, we discuss these factors in greater detail
and critically review the long-term effects of abnormal
fetal growth.
DEFINITIONS
Most approaches for defining fetal growth use gestational
age-based norms. The duration of pregnancy has become
an integral component of prenatal growth assessment,
and all currently prevailing definitions of fetal growth are
specific for gestational age. Assessing the gestational age
accurately, however, can be challenging. Any error in dating will lead to misclassification of the infant, which can
have significant clinical implications. In many instances,
the method of gestational age determination has contributed to variations in the gestational age–specific reference growth curves. For example, some nomograms are
based on approximating the gestational age to the nearest week, whereas others use the completed weeks. The
birthweight charts are also affected by other variables
that may limit their reliability. Many of these variables,
such as fetal gender, race, parity, birth order, parental
size, and altitude, contribute to the normal biologic variations in human fetal growth. There is continuing controversy regarding whether the reference growth charts
should be customized by multiple variables or developed
from the whole population. The customized approach
predicts the optimal growth in an individual pregnancy
and therefore specifically defines suboptimal growth for
that pregnancy. However, it has been argued that such an
approach can lead to a profusion of standards and might
not contribute to improving the outcome of infants who
are small for gestational age (SGA). In recognition of the
utility of a national standard, a population-based reference chart for fetal growth has been developed from all
the singleton births (more than 3 million) in the United
States in 1991 (Alexander, 1996). More recently, a similar national population-based fetal growth chart, which is
also sex specific, has been developed in Canada (Kramer
et al, 2001). There is insufficient evidence about whether
one approach is superior to the other in improving the
perinatal outcome.
There is no universal agreement on the classification of
an SGA infant. Various definitions appear in the medical
literature, making comparisons between studies difficult.
In addition, investigators have shown that the prevalence of fetal growth restriction varies according to the
fetal growth curve used (Alexander et al, 1996). The most
common definition of SGA refers to a weight less than
the 10th percentile for gestational age or birthweight less
than 2 standard deviations (SDs) from the mean. Some
investigators also use measurements less than the 3rd percentile to define SGA. However, these definitions do not
make a distinction among infants who are constitutionally small, growth restricted and small, and not small but
growth restricted relative to their potential. As an example, as many as 70% of fetuses with a weight less than the
10th percentile for gestational age at birth are small simply because of constitutional factors such as female sex or
maternal ethnicity, parity, or body mass index; they are
not at high risk of perinatal mortality or morbidity. In
contrast, true fetal growth restriction is associated with
numerous perinatal morbidities. This has clinical relevance to perinatologists and neonatologists, because many
of the tiniest premature neonates in the neonatal intensive
care units are probably growth restricted. McIntire et al
(1999) reported a threshold of increased adverse outcomes
in infants born with measurements less than the 3rd percentile and suggested that this level of restriction represents a clinically relevant measurement. Other researchers
have found higher rates of neonatal complications when
the 15th percentile of birthweight is used as a cutoff level
(Seeds and Peng, 1998).
There is an important distinction in identifying the fetus
with intrauterine growth restriction (IUGR), and the fetus
that is constitutionally small (i.e., SGA). IUGR is a condition in which a fetus is unable to achieve its genetically
determined potential size and represents a deviation and
a reduction in the expected fetal growth pattern. IUGR
complicates approximately 5% to 8% of all pregnancies
and 38% to 80% of all neonates with low birthweight
(LBW). This discrepancy underscores the fact that no
uniform definition of IUGR exists. Even when a normal
intrauterine growth pattern is established for a population, somewhat arbitrary criteria are used to define growth
restriction.
51
52
PART II Fetal Development
PATTERNS OF ALTERED GROWTH
Neonates with intrauterine growth retardation can be classified as demonstrating either symmetrical or asymmetrical growth. Infants with symmetric IUGR have reduced
weight, length, and head circumference at birth. Weight
(and then length) of infants with asymmetric growth
retardation is affected, with a relatively normal or “headsparing” growth pattern. Factors intrinsic to the fetus in
general cause symmetrical growth restriction, whereas
asymmetric IUGR is often associated with maternal medical conditions such as preeclampsia, chronic hypertension,
and uterine anomalies. Asymmetric patterns generally
develop during the third trimester, a period of rapid fetal
growth. However, now that fetal surveillance is more common, asymmetric growth restriction is often diagnosed in
the second trimester. Furthermore, many extremely premature neonates (<750 g) probably have IUGR.
Factors that are well recognized to limit the growth of
both the fetal brain and body include chromosomal anomalies (e.g., trisomies), congenital infections (toxoplasmosis,
rubella, cytomegalovirus, herpes simplex, malaria, HIV,
and parvovirus), dwarf syndromes, and some inborn errors
of metabolism. Cardiac and renal structural anomalies are
common fetal conditions associated with SGA. These conditions retard fetal growth primarily by impaired cell proliferation. Recognized causes of IUGR are listed in Table 6-1.
Etiologies of Fetal Growth Restriction
The epidemiology of fetal growth restriction varies internationally (Keirse, 2000). In developed countries, the most
frequently identified cause of growth restriction is smoking, whereas in developing countries, maternal nutritional
TABLE 6-1 Causes of Intrauterine Growth Restriction
Genetic
Inheritance, chromosomal abnormalities, fetal
gender
Maternal
constitutional
effects
Low maternal pre-pregnancy weight, low
pregnancy weight gain, ethnicity, socioeconomic status, history of intrauterine growth
restriction
Nutrition
Low pre-pregnancy weight (body mass index),
low pregnancy weight gain, malnutrition
(macronutrients, micronutrients), maternal
anemia
Infections
TORCH infections (toxoplasmosis, rubella,
cytomegalovirus, syphilis)
Decreased
O2-carrying
capacity
High altitude, maternal congenital heart disease, hemoglobinopathies, chronic anemia,
maternal asthma
Uterine and
placental
anatomy
Abnormal uterine anatomy, uterine fibroid,
vascular abnormalities (single umbilical artery, velamentous umbilical cord
insertion, twin-twin transfusion), placenta
previa, placental abruption
Uterine and
placental
function
Maternal vasculitis (system lupus erythematosus), decreased uteroplacental perfusion,
maternal illness (preeclampsia, chronic
hypertension, diabetes, renal disease)
Toxins
Tobacco, ethanol, lead, arsenic
factors (prepregnancy weight, maternal stature) and infections (malaria) are the leading identified causes (Krampl,
2000; Robinson et al, 2000). In addition, in developing
countries there is a direct correlation between the incidence of LBW (<2500 g) and IUGR. In developing countries, the high incidence of infants with LBW is almost
exclusively caused by the incidence of IUGR. Data from
developed countries show the opposite, with rates of LBW
being explained almost exclusively by prematurity rates.
In the United States, the cause of IUGR is identified in
approximately 40% of cases, with the remaining cases
labeled as idiopathic.
Numerous factors have been identified as influencing
size at birth. In a simplified manner, these factors can be
grouped as fetal, placental, or maternal in origin. These
factors will be discussed in detail in the following sections.
Fetal Causes of Growth Restriction
Fetal factors affecting growth include gender, familial
genetic inheritance, chromosomal abnormalities, and dysmorphic syndromes. In one large, population-based study,
the frequency of IUGR among infants with congenital malformations was 22%. The majority of the infants
affected had chromosomal abnormalities. Other studies
have similarly found that fetal growth restriction is more
common among infants with malformations. Fetal gender also influences size, with male infants showing greater
intrauterine growth than female infants (Glinianaia et al,
2000; Skjaerven et al, 2000; Thomas et al, 2000).
Placental Causes of Growth Restriction
In mammals, the major determinant of intrauterine
growth is the placental supply of nutrients to the fetus
(Fowden et al, 2006). In many species, fetal weight near
term is positively correlated to placental weight, as a proxy
measure of the surface area for maternal-fetal transport of
nutrients. Fetal weight near term is positively correlated to
placental weight, and the nutrient transfer capacity of the
placenta depends on its size, morphology, blood flow, and
transporter abundance (Fowden et al, 2006). In addition,
placental synthesis and metabolism of key nutrients and
hormones influences the rate of fetal growth (Fowden and
Forhead, 2004). Changes in any of these placental factors
can, therefore, affect intrauterine growth; however, the
fetus is not just a passive recipient of nutrients from the
placenta. The fetal genome exerts a significant acquisitive
drive for maternal nutrients through adaptations in the
placenta, particularly when the potential for fetal and placental growth is compromised.
Placental maturation at the end of pregnancy is associated with an increase in substrate transfer, a slowing (but not
cessation) of placental growth, and a plateau in fetal growth
near term (Fox, 1997). Abnormalities of placental growth,
senescence, and infarction have been shown to affect fetal
growth. The placentas from pregnancies complicated by
poor fetal growth have a higher incidence of vascular damage
and abnormalities (Pardi et al, 1997). Fetal size and placental growth are directly related, and placentas from pregnancies yielding growth-restricted infants demonstrate a higher
incidence of smallness and abnormality than do those from
CHAPTER 6 Abnormalities of Fetal Growth
pregnancies with appropriately grown infants. The difference in size is seen even in a comparison of placentas associated with growth-restricted infants and those associated
with appropriate for gestational age (AGA) infants of the
same birthweight (Heinonen et al, 2001). Placental growth
in the second trimester correlates with placental weight
and function and, thus, with weight at birth (Godfrey et al,
1996). Clinical conditions associated with reduced placental
size (and subsequent reduced fetal weight) include maternal
vascular disease, uterine anomalies (uterine fibroids, abnormal uterine anatomy), placental infarctions, unusual cord
insertions, and abnormalities of placentation.
Multiple gestations are associated with greater risk
for fetal growth restriction. The higher risk stems from
crowding and from abnormalities with placentation, vascular communications, and umbilical cord insertions.
Divergence in fetal growth appears from approximately
30 to 32 weeks in twin gestation compared with singleton
pregnancies (Alexander et al, 1996; Glinianaia et al, 2000;
Skjaerven et al, 2000). Others have identified differences
in fetal growth between twins and singletons as occurring
earlier in gestation, at approximately 21 weeks’ gestation
(Devoe and Ware, 1995). Larger effects on fetal growth
are seen with increasing number of fetal multiples. Abnormalities in placentation are also more common with multiple gestations (Benirschke, 1995). Monochorionic twins
can share placental vascular communication (twin-twin
transfusion), leading to fetal growth restriction during gestation. Fetal competition for placental transfer of nutrients
raises the incidence of growth restriction and discordance
in growth between fetuses. The rate of birthweights less
than the 5th percentile is higher in monochorionic twins.
Placental growth is restricted in utero because of limitation
in space, leading to a higher incidence of placenta previa in
multiple-gestation pregnancies. In addition, abnormalities
in cord insertions (marginal and velamentous cord insertions) and occurrence of a single umbilical artery are more
frequently found in multiple gestations. The higher incidence of growth restriction in multiple-gestation pregnancies is strongly associated with monochorionic gestations,
the presence of vascular anastomoses, and discordant fetal
growth (Hollier et al, 1999; Sonntag et al, 1996; Victoria
et al, 2001). Placentas of smaller fetuses with discordant
growth are significantly smaller than those of their larger
twin counterparts (Victoria et al, 2001).
Investigators have shown an effect of altitude on fetal
growth, with infants born at high altitudes having lower
birthweights (Galan et al, 2001). Differences in fetal growth
are detected from approximately 25 weeks’ gestation with
pregnancies at 4000 m. In these high-altitude pregnancies, the abdominal circumference is most affected (Krampl
et al, 2000). At tremendously high altitudes, the incidence
of LGA infant births is markedly reduced. In the United
States (and at less severe altitudes), infants born at higher
altitudes are lighter at birth, but those differences are not
pronounced. Interestingly, investigators have shown that
adaptation to high altitude during pregnancy is also possible. Tibetan infants have higher birthweights than infants
of more recent immigrants of ethnic Chinese living at
the same high-altitude (2700 to 4700 m) region of Tibet
(Moore et al, 2001). Tibetan infants also have less IUGR
than do infants born to more recent immigrants to the area.
53
Maternal Causes of Growth Restriction
Maternal health conditions associated with chronic
decreases in uteroplacental blood flow (maternal vascular diseases, preeclampsia, hypertension, maternal smoking) are associated with poor fetal growth and nutrition.
Preeclampsia has been shown to be associated with fetal
growth restriction (Ødegård et al, 2000; Spinillo et al,
1994; Xiong et al, 1999). Investigators have shown that
the extent of growth restriction correlates with the severity and the onset during pregnancy of the preeclampsia.
Ødegård et al (2000) showed that fetuses exposed to preeclampsia from early in pregnancy had the most serious
growth restriction, and more than half of these infants
were born SGA. Chronic maternal diseases (cardiac, renal)
may decrease the normal uteroplacental blood flow to
the fetus and thus may also be associated with poor fetal
growth (Spinillo et al, 1994).
Maternal constitutional factors have a significant effect
on fetal growth. Maternal weight (pre-pregnancy), maternal stature, and maternal weight gain during pregnancy
are directly associated with maternal nutrition and correlate with fetal growth (Clausson et al, 1998; Doctor et al,
2001; Goldenberg et al, 1997; Mongelli and Gardosi,
2000). Numerous studies show that these findings are
often confounded by highly associated cultural and socioeconomic factors. The woman with a previous SGA infant
has a higher risk of a subsequent small infant (Robinson
et al, 2000). Investigators have shown a higher incidence
of SGA infants to be associated with lower levels of maternal education (Clausson et al, 1998). Parity of the mother
also affects fetal size; nulliparous women having a higher
incidence of SGA infants (Cnattingius et al, 1998). A large
population-based study in Sweden found that women who
were older than 30 years, were nulliparous, or had hypertensive disease were at increased risk of preterm and term
growth-restricted infants.
Studies have shown differential fetal growth for women
of diverse ethnicities, with Latina and white women having
higher rates of LGA infants, and African American women
having a higher incidence of SGA infants (Alexander et al,
1999; Collins and David, 2009; Fuentes-Afflick et al,
1998). These gender and ethnic differences in birthweight
become pronounced after 30 weeks’ gestation (Thomas
et al, 2000). Investigators in California have shown that
U.S.-born black women have higher rates of prematurity
and LBW infants than do foreign-born black women.
Other researchers have found that even among women
with low risk of LBW infants (married, age 20 to 34 years,
13 or more years of education, adequate prenatal care,
and absence of maternal health risk factors and tobacco
or alcohol use), the risk of delivering an SGA infant is
still higher for African American women than for white
women (Alexander et al, 1999; Collins and David, 2009).
It is unclear whether these differences in fetal growth are
caused by inherent differences or by differential exposure
to environmental factors.
Maternal nutrition and supply of nutrients to the fetus
affect fetal growth significantly, primarily in developing
countries (Doctor et al, 2001; Godfrey et al, 1996; Neggers
et al, 1997; Robinson et al, 2000; Zeitlin et al, 2001).
Although numerous factors interact with and affect fetal
54
PART II Fetal Development
development, maternal malnutrition is assumed to be a
major cause of IUGR in developing countries. Furthermore, pre-pregnancy weight may be a potential marker
for intergenerational effects on infant weight in developing countries. A woman’s birthweight has been shown to
correlate with her infant’s weight as well as the placental
weight during pregnancy. In the United States, Strauss
and Dietz (1999) report that low maternal weight gain in
the second and third trimesters is associated with a twofold
risk of IUGR, whereas poor maternal weight gain in the
first trimester has no such effect on fetal growth (Strauss
and Dietz, 1999). These investigators also showed that
older women (older than 35 years) and smokers were at
increased risk of IUGR associated with lower weight gains
in late pregnancy.
Teen pregnancy represents a special condition in which
fetal weight is highly influenced by maternal nutrition.
Teen mothers (younger than 15 years) have been shown
to have a higher risk for delivering a growth-restricted
infant (Ghidini, 1996). Teen pregnancies are complicated
by the additional nutritional needs of a pregnant mother,
who is still actively growing, as well as by socioeconomic
status of pregnant teens in developed countries (Scholl and
Hediger, 1995). Maternal nutrition and maternal weight
gain are adversely affected by inadequate or poorly balanced intake in conditions such as alcoholism, drug abuse,
and poverty.
The effects of micronutrients on pregnancy outcomes
and fetal growth have been less well studied. Maternal
intake of certain micronutrients has also been found to
affect fetal growth. Zinc deficiency has been associated with
fetal growth restriction and other abnormalities, such as
infertility and spontaneous abortion (Jameson, 1993; Shah
and Sachdev, 2001). In addition, dietary intake of vitamin
C during early pregnancy has been shown to be associated with an increase in birthweight (Mathews et al, 1999).
Others have shown strong associations between maternal
intake of folate and iron and infant and placental weights
(Godfrey et al, 1996). In developing countries, the effects
of nutritional deficiencies during pregnancy are more
prevalent and easier to detect. Rao et al (2001) have estimated that one third of infants in India are born weighing
less than 2500 g, mainly because of maternal malnutrition.
These investigators have shown significant associations
between infant birthweight and maternal intake of milk,
leafy greens, fruits, and folate during pregnancy.
Although toxins such as cigarette smoke and alcohol
have a direct effect on placental function, they may also
affect fetal growth through an associated compromise in
maternal nutrition. Other environmental toxins (lead, arsenic, mercury) are associated with IUGR and are believed to
affect fetal growth by entering the food chain and depleting
body stores of iron, vitamin C, and possibly other nutrients
(Iyengar and Nair, 2000; Srivastava et al, 2001).
Numerous studies have shown associations between
birthweight and maternal intake of macronutrients and
micronutrients, but the effects of nutritional supplements
used during pregnancy on fetal growth are equivocal
(de Onis et al, 1998; Jackson and Robinson, 2001; Rush,
2001; Say et al, 2003). This finding is underscored by the
results of a recent, large, double-blind, randomized controlled trial including 1426 pregnancies in rural Burkina
Faso (Roberfroid et al, 2008). Pregnant women were randomly assigned to receive either iron and folic acid or the
UNICEF-WHO-UNU* international multiple micronutrient preparation (UNIMMAP) daily until 3 months
after delivery, with the UNIMMAP thereafter in both
groups. Birthweight was increased by 52 g, and length was
increased by 3.6 mm. Unexpectedly, the risk of perinatal
death was marginally significantly increased in the UNIMMAP group (odds ratio, 1.78; 95% confidence interval,
0.95 to 3.32; p = 0.07).
Maternal socioeconomic status and ethnicity have also
been identified as risk factors for IUGR and poor health
outcomes in infants. Numerous investigators have shown
a significant effect of socioeconomic status on birth outcomes, including fetal growth restriction, in both developing and developed countries (Wilcox et al, 1995). In the
United States, low levels of maternal and paternal education, certain maternal and paternal occupations, and low
family income are associated with lower birthweights in
children of African American and white women (Parker
et al, 1994). In a large population-based study in Sweden,
investigators have shown a higher incidence of fetal growth
restriction in association with low maternal education
(Clausson et al, 1998). Researchers have also shown that
rates of compromised birth outcome are higher among
African American women than among Mexican American
and non-Hispanic white women (Collins and Butler, 1997;
Frisbie et al, 1997; Thomas et al, 2000). Some of these
studies also show that the risk of IUGR is higher in women
without medical insurance. In the United States, the incidence of IUGR is significantly higher among African
American women than among white women; this higher
incidence is seen even among African American women
with higher socioeconomic status (Alexander et al, 1999).
In a study in Arizona, the incidence of IUGR was found
to be lower in Mexican American women than in white
women (Balcazar, 1994). Other researchers have shown
that Mexican-born immigrants in California have better
perinatal outcomes (including birthweight) than African
Americans and U.S.-born women of Mexican descent
(Fuentes-Afflick et al, 1998). The reasons for this apparent paradox are unclear, but one postulate is the tendency
of recent immigrants to maintain the favorable nutritional
and behavioral characteristics of their country of origin
(Guendelman and English, 1995). These studies support
the speculation that the differences in fetal growth between
groups do not reflect inherent differences in fetal growth,
but rather stem from inequalities in nutrition, health care,
and other environmental factors (Keirse, 2000; Kramer
et al, 2000).
Smoking
Cigarette smoking is consistently found to adversely affect
intrauterine growth in all studies in which this factor is
considered. In developed countries, cigarette smoking
is the single most important cause of poor fetal growth
(Kramer et al, 2000). The incidence of IUGR in smokers
*United
Nations Children’s Fund, World Health Organization, United Nations
University.
CHAPTER 6 Abnormalities of Fetal Growth
is estimated to be threefold to 4.5-fold higher than in
nonsmokers (Nordentoft et al, 1996). Cigarette smoking has a significant effect on abdominal circumference
and fetal weight, but not on head circumference (Bernstein et al, 2000). Lieberman et al (1994) reported that
cigarette smoking also appears to have a dose-dependent
effect on the incidence of IUGR, with this effect being
seen especially with heavy smoking and smoking during
the third trimester. These investigators have shown that
if women stop smoking during the third trimester, their
infants’ birthweights are indistinguishable from those of
infants born to the normal population. Other researchers
have shown that even a reduction in smoking is associated
with improved fetal growth (Li et al, 1993; Walsh et al,
2001). Numerous potential causes of the effects of smoking on fetal growth have been suggested, including direct
effects of nicotine on placental vasoconstriction, decreased
uterine blood flow, higher levels of fetal carboxyhemoglobin, fetal hypoxia, adverse maternal nutritional intake, and
altered maternal and placental metabolism (Andres and
Day, 2000; Pastrakuljic et al, 1999).
SHORT-TERM OUTCOMES
IUGR alters many physiologic and metabolic functions in
the fetus and neonate that result in a number of morbidities. A large cohort study of 37,377 pregnancies found a
fivefold to sixfold greater risk of perinatal death for both
preterm and term fetuses that had IUGR (Cnattingius et al,
1998; Lackman et al, 2001; Mongelli and Gardosi, 2000).
Predictive factors for perinatal mortality in preterm fetuses
with IUGR reveals that of all antenatal factors examined,
only oligohydramnios and abnormal umbilical artery Dopplers with absent or reversed diastolic flow were predictive
of perinatal mortality (Scifres et al, 2009). Although the
growth-restricted fetus may show symmetric or asymmetric growth at birth, it is unclear whether the proportionality
of the fetus with IUGR truly affects outcomes or is related
to the timing or the severity of the insult. Lin et al (1991)
found that symmetric IUGR resulted in higher levels of
prematurity and higher rates of neonatal morbidity. In contrast, Villar et al (1990) have shown that infants with asymmetric IUGR have higher morbidity rates at birth. They
found that infants with low ponderal index measurements
(which they defined as Weight ÷ Length3) had a higher risk
of low Apgar scores, long hospitalization, hypoglycemia,
and asphyxia at birth than infants with symmetric IUGR.
There is evidence to suggest that infants with asymmetric IUGR show better gains in weight and length in the
postnatal period than symmetrically restricted infants
(May et al, 2001). Other investigators propose that IUGR
represents a continuum, with symmetric IUGR occurring
as the severity of growth retardation increases. Data also
suggest that the more severe the growth restriction, the
worse the neonatal outcomes, including risk of stillbirth,
fetal distress, neonatal hypoglycemia, hypocalcemia, polycythemia, low Apgar scores, and mortality (Kramer et al,
1990; Spinillo et al, 1995).
Fetal growth restriction is associated with intrauterine
demise. Almost 40% of term stillbirths and 63% of preterm stillbirths are SGA (Mongelli and Gardosi, 2000).
Both short-term and long-term effects of abnormalities
55
in SGA fetuses have been described. Perinatal mortality
for intrauterine SGA infants is higher overall than that for
appropriately grown term and preterm infants (Clausson
et al, 1998). The risk of perinatal death is estimated to
be fivefold to sixfold greater for both preterm and term
fetuses with IUGR (Lackman et al, 2001). Overall, intrauterine death, perinatal asphyxia, and congenital anomalies
are the main contributing factors to the higher mortality
rate in SGA infants. The effects of acute fetal hypoxia may
be superimposed on chronic fetal hypoxia, and placental
insufficiency may be an important etiologic factor in these
outcomes. Investigators have described higher incidences
of low Apgar scores, umbilical artery acidosis, need for
intubation at delivery, seizures on the first day of life, and
sepsis in SGA infants (McIntire et al, 1999). The incidence
of adverse perinatal effects correlates with the severity of
the growth restriction, the highest rates of respiratory distress syndrome, metabolic abnormalities, and sepsis being
found in the most severely growth-restricted infants (Spinillo et al, 1995). As previously described, Villar et al (1990)
reported that infants with asymmetric IUGR and low ponderal index measurements had a higher risk of low Apgar
scores, hypoglycemia, asphyxia, and long hospitalization.
Preterm infants with growth abnormalities have a much
higher risk of adverse outcomes. Preterm SGA infants have
a higher incidence of a number of complications, including
sepsis, severe intraventricular hemorrhage, respiratory distress syndrome, necrotizing enterocolitis, and death, than
do normally grown preterm infants (Gortner et al, 1999;
McIntire et al, 1999; Simchen et al, 2000). In addition,
SGA infants have a higher incidence of chronic lung disease at corrected gestational ages of 28 days and 36 weeks.
Neonatal hypoglycemia and hypothermia occur more
frequently in growth-restricted infants (Doctor et al,
2001). These metabolic abnormalities presumably occur
from decreased glycogen stores, inadequate lipid stores,
and impaired gluconeogenesis in the growth-restricted
neonate. Growth-restricted neonates have inadequate
fuel stores and are at increased risk for hypoglycemia during fasting, and these risks are increased in preterm SGA
infants. Infants with IUGR also have a higher incidence of
hypocalcemia, with the incidence correlating strongly with
the severity of growth restriction (Spinillo et al, 1995).
Developmental Outcomes:
Early Childhood
Neurologic outcomes, including intellectual and neurologic function, are affected by growth restriction. Overall,
neurologic morbidity is higher for SGA infants than for
AGA infants. Without identified perinatal events, SGA
infants have a higher incidence of long-term neurologic
or developmental handicaps. Investigators have found the
incidence of cerebral palsy to be greater in IUGR infants
than in a population with normal fetal growth (Blair and
Stanley, 1990; Spinillo et al, 1995; Uvebrant and Hagberg,
1992). SGA infants born at term appear to have double or
triple the risk for cerebral palsy, between 1 to 2 per 1000
live births and 2 to 6 per 1000 live births (Goldenberg
et al, 1998). The rate of cerebral palsy is also higher in
preterm growth-restricted infants than in preterm infants
with appropriate fetal growth (Gray et al, 2001). At 7 years
56
PART II Fetal Development
of age, children whose birth was associated with hypoxiarelated factors had a higher risk for adverse neurologic
outcomes. Infants with symmetric IUGR, or perhaps
more severe restriction, were at higher risk than infants
with asymmetric IUGR. Other researchers have shown
higher rates of learning deficits, lower intelligence quotient scores, and increased behavioral problems in children
with a history of fetal growth restriction, even at 9 to 11
years of age (Low et al, 1992).
LONG-TERM CONSEQUENCES:
THE DEVELOPMENTAL ORIGINS
OF ADULT DISEASE
Programming
The period from conception to birth is a time of rapid
growth, cellular replication and differentiation, and functional maturation of organ systems. These processes are
highly sensitive to alterations in the intrauterine milieu.
The term programming describes the mechanisms whereby
a stimulus or insult at a critical period of development
has lasting or lifelong effects. The “thrifty phenotype”
hypothesis proposes that the fetus adapts to an adverse
intrauterine milieu by optimizing the use of a reduced
nutrient supply to ensure survival; but because this adaptation favors the development of certain organs over that of
others, it leads to persistent alterations in the growth and
function of developing tissues (Hales and Barker, 1992). In
addition, although the adaptations may aid in survival of
the fetus, they become a liability in situations of nutritional
abundance.
Epidemiology
It has been recognized for nearly 70 years that the early
environment in which a child grows and develops can
have long-term effects on subsequent health and survival
(Kermack, 1934). The landmark cohort study of 300,000
men by Ravelli et al (1976) showed that men who were
exposed in utero to the effects of the Dutch famine of 1944
and 1945 during the first half of gestation had significantly
higher obesity rates at the age of 19 years. Subsequent
studies demonstrated relationships among LBW, the later
development of cardiovascular disease (Barker et al, 1989),
and impaired glucose tolerance (Fall et al, 1995) in men in
England. Men who were smallest at birth (2500 g) were
nearly sevenfold more likely to have impaired glucose tolerance or type 2 diabetes than those who were largest at
birth. Barker et al (1993) also found a similar relationship
between lower birthweight and higher systolic blood pressure and triglyceride levels.
Valdez et al (1994) observed a similar association
between birthweight and subsequent glucose intolerance,
hypertension, and hyperlipidemia in a study of young
adult Mexican American and non-Hispanic white men and
women participants in the San Antonio Heart Study. Normotensive individuals without diabetes whose birthweights
were in the lowest tertile had significantly higher rates of
insulin resistance, obesity, and hypertension than subjects whose birthweights were normal. In the Pima Indians, a population with extraordinarily high rates of type 2
diabetes, McCance et al (1994) found that the development
of diabetes in the offspring was related to both extremes
of birthweight. In their study, the prevalence of diabetes in
subjects 20 to 39 years old was 30% for those weighing less
than 2500 g at birth, 17% for those weighing 2500 to 4499
g, and 32% for those weighing 4500 g or more. The risk
of developing type 2 diabetes was nearly fourfold higher
for those whose birthweight was less than 2500 g. Other
studies of populations in the United States (Curhan et al,
1996), Sweden (Lithell et al, 1996; McKeigue et al, 1998),
France (Jaquet et al, 2000; Leger et al, 1997), Norway
(Egeland et al, 2000), and Finland (Forsen et al, 2000) have
all demonstrated a significant correlation between LBW
and the later development of adult diseases.
Studies controlling for the confounding factors of socioeconomic status and lifestyle have further strengthened
the association between LBW and a higher risk of coronary heart disease, stroke, and type 2 diabetes. In 1976, the
Nurses’ Health Study was initiated, and a large cohort of
American women born from 1921 to 1946 established. The
association between LBW and increased risks of coronary
heart disease, stroke, and type 2 diabetes remained strong
even after adjustment for lifestyle factors such as smoking,
physical activity, occupation, income, dietary habits, and
childhood socioeconomic status (Rich-Edwards et al, 1999).
Role of Catch-up Growth
Many studies have suggested that the associations between
birth size with later disease can be modified by body mass
index (BMI) in childhood. The highest risk for the development of type 2 diabetes is among adults who were born
small and become overweight during childhood (Eriksson
et al, 2000). Insulin resistance is most prominent in Indian
children who were SGA at birth, but had a high fat mass at
8 years of age (Bavdekar et al, 1999). Similar findings were
reported in 10-year-old children in the United Kingdom
(Whincup et al, 1997). In a Finnish cohort, adult hypertension was associated with both lower birthweight and
accelerated growth in the first 7 years of life. In contrast,
in two preliminary studies from the United Kingdom,
catch-up growth in the first 6 months of life was not clearly
related to blood pressure in young adulthood, although
birthweight was (McCarthy et al, 2001).
Interpreting the findings of these studies is complicated
by the vague definitions of catch-up growth. The term,
which can encompass either the first 6 to 12 months only
or as much as the first 2 years after birth, usually refers to
realignment of genetic growth potential after IUGR. This
definition allows for fetal growth retardation at any birthweight; large fetuses can be growth retarded in relation
to their genetic potential. However, postnatal factors can
obviously affect infant growth in the first few months of
life. For example, breastfeeding appears to protect against
obesity later in childhood, but breastfed infants usually
exhibit higher body mass during the first year of life than
formula-fed infants. Although it is likely that accelerated
growth confers an additional risk to the growth-retarded
fetus, these conflicting results demonstrate the need for
additional, carefully designed studies to determine how
childhood growth rates affect the later development of
cardiovascular disease and type 2 diabetes.
CHAPTER 6 Abnormalities of Fetal Growth
Size at Birth, Insulin Secretion,
and Insulin Action
The mechanisms underlying the association between size
at birth and impaired glucose tolerance or type 2 diabetes
are unclear. A number of studies in children and adults have
shown that nondiabetic or prediabetic (abnormal glucose
tolerance) subjects with LBW are insulin resistant and thus
are predisposed to development of type 2 diabetes (Bavdekar
et al, 1999; Clausen et al, 1997; Flanagan et al, 2000;
Hoffman et al, 1997; Leger et al, 1997; Li et al, 2001; Lithell
et al, 1996; McKeigue et al, 1998; Phillips et al, 1994; Yajnik
et al, 1995). IUGR is known to alter the fetal development
of adipose tissue, which is closely linked to the development
of insulin resistance (Lapillonne et al, 1997; Widdowson
et al, 1979). In a well-designed case-control study of 25-yearold adults, Jaquet et al (2000) demonstrated that individuals
who were born SGA at 37 weeks’ gestation or later had a
significantly higher percentage of body fat (15%). Insulin
sensitivity, after adjustment for BMI or total fat mass, was
markedly impaired in these SGA subjects. There were no
significant differences between the SGA and control groups
regarding parental history of type 2 diabetes, cardiovascular
disease, hypertension, or dyslipidemia. Of importance when
generalizing the findings to other populations, the causes
of IUGR in these subjects were gestational hypertension
(50%), smoking (30%), maternal short stature (7%), congenital anomalies (7%), and unknown (6%).
The adverse effect of IUGR on glucose homeostasis was
originally thought to be mediated through programming of
the fetal endocrine pancreas. Growth-retarded fetuses and
newborns have been shown to have a reduced population of
pancreatic β cells (Van Assche et al, 1977). LBW has been
associated with reduced insulin response after glucose ingestion in young men without diabetes; however, a number of
other studies have found no effect of LBW on insulin secretion in humans (Clausen et al, 1997; Flanagan et al, 2000;
Lithell et al, 1996). However, none of these earlier studies
adjusted for the corresponding insulin sensitivity, which has
a profound effect on insulin secretion. Jensen et al (2002)
measured insulin secretion and insulin sensitivity in a wellmatched population of 19-year-old, glucose-tolerant white
men whose birthweights were either less than the 10th percentile (i.e., SGA) or between the 50th and 75th percentiles
(controls). To eliminate the major confounding factors,
such as “diabetes genes,” the researchers ensured that none
of the participants had a family history of diabetes, hypertension, or ischemic heart disease. They found no differences between the groups in regard to current weight, BMI,
body composition, and lipid profile. When data were controlled for insulin sensitivity, insulin secretion was found to
be lower by 30%. However, insulin sensitivity was normal
in the SGA subjects. These investigators hypothesized that
defects in insulin secretion precede defects in insulin action,
and that SGA individuals demonstrate insulin resistance
once they accumulate body fat.
Epidemiologic Challenges
The data described in the preceding section suggest that
LBW is associated with glucose intolerance, type 2 diabetes, and cardiovascular disease. However, the question
57
remains whether these associations reflect fetal nutrition
or other factors that contribute to birthweight and the
observed glucose intolerance. Because of the retrospective nature of the cohort identification, many confounding
variables were not always recorded, such as lifestyle, socioeconomic status, education, maternal age, parental build,
birth order, obstetric complications, smoking, and maternal health. Maternal nutritional status, either directly in
the form of diet histories, or indirectly in the form of
BMI, height, and pregnancy weight gain, were usually not
recorded. Instead, birth anthropometric measures were
used as proxies for presumed undernutrition in pregnancy.
Size at Birth Cannot Be Used as a Proxy
for Fetal Growth
Birthweight is determined by the sum of multiple known
and unknown factors, including gestational age, maternal
age, birth order, genetics, maternal pre-pregnancy BMI,
and pregnancy weight gain, plus multiple environmental
factors, such as smoking, drug use, infection, and maternal
hypertension. Some of these determinants may be related
to susceptibility to adult disease, and others may not. Conversely, some prenatal determinants of adult outcomes
may not be related to fetal growth. A good example of how
size at birth may potentially be a proxy for an underlying
causal pathway is the hypothesis that essential hypertension in the adult is caused by a congenital nephron deficit
(Brenner and Chertow, 1993). This study shows that kidney volume is smaller in adults who were thinner at birth,
after adjustment for current body size. In contrast, maternal cigarette smoking is a good example of a prenatal exposure that restricts fetal growth, but to date no association
has been found between cigarette smoking and adverse
long-term outcome in offspring.
Genetics versus Environment
Several epidemiologic and metabolic studies of twins and
first-degree relatives of patients with type 2 diabetes have
demonstrated an important genetic component of diabetes (Vaag et al, 1995). The association between LBW and
risk of type 2 diabetes in some studies could theoretically
be explained by a genetically determined reduced fetal
growth rate. In other words, the genotype responsible
for type 2 diabetes may itself restrict fetal growth. This
possibility forms the basis for the fetal insulin hypothesis,
which suggests that genetically determined insulin resistance could result in insulin-mediated low growth rate in
utero as well as insulin resistance in childhood and adulthood (Hattersley et al, 1999). Insulin is one of the major
growth factors in fetal life, and monogenic disorders that
affect the fetus’s insulin secretion or insulin resistance
also affect fetal growth (Elsas et al, 1985; Froguel et al,
1993; Hattersley et al, 1998; Stoffers et al, 1997). Mutations in the gene encoding glucokinase have been identified that result in LBW and maturity-onset diabetes of
the young. Such mutations are rare, and no analogous
common allelic variation has been discovered, but it is
likely that some variations exist that, once identified, will
help to explain a proportion of the cases of diabetes in
LBW subjects.
58
PART II Fetal Development
There is obviously a close relationship between genes
and the environment. Maternal gene expression can alter
the fetal environment, and the maternal intrauterine environment also affects fetal gene expression. An adverse
intrauterine milieu is likely to have profound long-term
effects on the developing organism that might not be
reflected in birthweight.
Cellular Mechanisms
Fetal malnutrition has two main causes: poor maternal
nutrition and placental insufficiency. In the extensive literature about the fetal origins hypothesis, these two concepts have not been discerned clearly. Such a distinction
is necessary, because maternal nutrition has probably
been adequate in the majority of populations in which the
hypothesis has been tested. Only extreme maternal undernutrition, such as occurred in the Dutch famine, reduces
the birthweight to an extent that could be expected to raise
the risk of adult disease (Lumey et al, 1995). To a lesser
extent but equally important is the LBW in populations
with low resources, resulting in maternal undernutrition.
Overall, in most populations it is reasonable that placental
insufficiency has been a main cause of LBW. The oxygen
and nutrients that support fetal growth and development
rely on the entire nutrient supply line, beginning with
maternal consumption and body size, but extending to
uterine perfusion, placental function, and fetal metabolism. Interruptions of the supply line at any point could
result in programming of the fetus for the future risk of
adult diseases.
The intrauterine environment influences development
of the fetus by modifying gene expression in both pluripotential cells and terminally differentiated, poorly replicating cells. The long-range effects on the offspring (into
adulthood) are determined by which cells are undergoing
differentiation, proliferation, or functional maturation at
the time of the disturbance in maternal fuel economy. The
fetus also adapts to an inadequate supply of substrates (e.g.,
glucose, amino acids, fatty acids, and oxygen) through metabolic changes, redistribution of blood flow, and changes
in the production of fetal and placental hormones that
control fetal growth.
The fetus’s immediate metabolic response to placental
insufficiency is catabolism, consuming its own substrates to
provide energy. A more prolonged reduction in availability
of substrates leads to slowed growth, which enhances the
fetus’s ability to survive by reducing the use of substrates
and lowering the metabolic rate. Slowed growth in late
gestation leads to disproportionate organ size, because the
organs and tissues that are growing rapidly at the time are
affected the most. For example, placental insufficiency in
late gestation can lead to reduced growth of the kidney,
which is developing rapidly at that time. Reduced replication of kidney cells can permanently reduce cell numbers,
because there seems to be no capacity for renal cell division to catch up after birth.
Substrate availability has profound effects on fetal hormones and on the hormonal and metabolic interactions
among the fetus, placenta, and mother. These effects are
most apparent in the fetus of the mother with diabetes.
Higher maternal concentrations of glucose and amino
acids stimulate the fetal pancreas to secrete exaggerated
amounts of insulin and stimulate the fetal liver to produce
higher levels of insulin-like growth factors. Fetal hyperinsulinism stimulates the growth of adipose tissue and
other insulin-responsive tissues in the fetus, often leading
to macrosomia. However, many offspring of mothers with
diabetes with fetal hyperinsulinism are not overgrown by
usual standards, and many with later obesity and glucose
intolerance were not macrosomic at birth (Pettitt et al,
1987; Silverman et al, 1995). These observations suggest
that birthweight is not a good indication of intrauterine
nutrition.
MACROSOMIA
Excessive fetal growth (macrosomia, being large for gestational age) is found in 9% to 13% of all deliveries and can
lead to significant complications in the perinatal period
(Gregory et al, 1998; Wollschlaeger et al, 1999). Maternal factors associated with macrosomia during pregnancy
include increasing parity, higher maternal age, and maternal height. In addition, the previous delivery of an infant
with macrosomia, prolonged pregnancy, maternal glucose
intolerance, high pre-pregnancy weight or obesity, and
large pregnancy weight gain have all been found to raise
the risk of delivering an infant with macrosomia (Mocanu
et al, 2000).
Maternal complications of macrosomia include morbidities related to labor and delivery. Prolonged labor,
arrest of labor, and higher rates of cesarean section and
instrumentation during labor have been reported. In addition, the risks of maternal lacerations and trauma, delayed
placental detachment, and postpartum hemorrhage are
higher for the woman delivering an infant with macrosomia (Lipscomb et al, 1995; Perlow et al, 1996). Complications of labor are more pronounced in primiparous
women than in multiparous women (Mocanu et al, 2000).
The neonatal complications of macrosomia include traumatic events such as shoulder dystocia, brachial nerve
palsy, birth trauma, and associated perinatal asphyxia.
Other complications for the neonate are elevated insulin
levels and metabolic derangements, such as hypoglycemia
and hypocalcemia (Wollschlaeger et al, 1999). In a large
population-based study in the United States, macrosomia
(defined as birthweight greater than 4000 g) was detected
in 13% of births. Of these, shoulder dystocia was noted in
11% (Gregory et al, 1998).
Macrosomia is often not detected during pregnancy
and labor. The clinical estimation of fetal size is difficult and has significant false-positive and false-negative
rates. Ultrasonography estimates of fetal weight are not
always accurate, and there are a wide range of sensitivity estimates for the ultrasound detection of macrosomia. In addition, there is controversy regarding how to
define macrosomia and which ultrasound measurement
is most sensitive in predicting macrosomia. Smith et al
(1997) demonstrated a linear relation between abdominal circumference and birthweight. They showed that the
equations commonly used for estimated fetal weight have
a median error rate of 7%, with greater errors seen with
larger infants. Using receiver operating characteristics
curves to measure the diagnostic accuracy of ultrasound,
CHAPTER 6 Abnormalities of Fetal Growth
O’Reilly-Green and Divon (1997) reported sensitivity
and specificity rates of 85% and 72%, respectively, for
estimation of birthweight exceeding 4000 g. In their
study, the positive predictive value (i.e., a positive test
result represents a truly macrosomic infant) was approximately 49%. Chauhan et al (2000) found lower sensitivity for the use of ultrasound measurement of abdominal
and head circumference and femur length (72% sensitivity), similar to the sensitivity of using clinical measurements alone (73%). Other investigators have shown that
clinical estimation of fetal weight (43% sensitivity) has
higher sensitivity and specificity than ultrasound evaluation in predicting macrosomia (Gonen et al, 1996). In
a retrospective study, Jazayeri et al (1999) showed that
ultrasound measurement of abdominal circumference of
greater than 35 cm predicts macrosomia in 93% of cases
and is superior to measurements of biparietal diameter
or the femur. Other researchers have reported that an
abdominal circumference of more than 37 cm is a better
predictor (Al-Inany et al, 2001; Gilby et al, 2000).
Numerous investigators have also questioned whether
antenatal diagnosis improves birth outcomes in macrosomic infants. Investigators indicate the low rates of specificity of antenatal tests resulting in high rates of false-positive
results (Bryant et al, 1998, O’Reilly-Green and Divon,
1997). Antenatal identification of macrosomia or possible
macrosomia can lead to a higher rate of cesarean section
performed for infants with normal birthweights (Gonen
et al, 2000; Mocanu et al, 2000; Parry et al, 2000). Macrosomia is a risk factor for shoulder dystocia, but the
majority of cases of shoulder dystocia and birth trauma
occur in infants with macrosomia (Gonen et al, 1996).
A retrospective study of infants weighing more than 4200
g at birth showed a cesarean section rate of 52% in infants
predicted antenatally to have macrosomia, compared with
30% in infants without such an antenatal prediction. The
antenatal prediction of fetal macrosomia is also associated
59
with a higher incidence of failed induction of labor and
no reduction in the rate of shoulder dystocia (Zamorski
and Biggs, 2001). Using retrospective data from a 12-year
period, Bryant et al (1998) estimated that a policy of routine cesarean section for all infants with estimated fetal
weight greater than 4500 g would require between 155 and
588 cesarean sections to prevent a single case of permanent
brachial nerve palsy.
SUMMARY
This chapter has described many identified biologic and
genetic factors associated with fetal growth and with
abnormalities of fetal growth. Physicians are limited in the
ability to identify a causative agent in every case. Modification of fetal growth is possible and occurs from diverse
influences such as socioeconomic status, maternal nutrition, and maternal constitutional factors. Abnormal fetal
growth influences acute perinatal outcomes and health
during infancy, childhood, and adulthood. In schools of
public health, students are taught to search “up river” for
solutions to health problems. Solutions for ill health in
adulthood may reside in the identification of methods to
improve the health of the fetus.
SUGGESTED READINGS
Alexander GR, Himes JH, Kaufman R, et al: A United States national reference for
fetal growth, Obstet Gynecol 87:163-168, 1996.
Dahri S, Snoeck A, Reusens-Billen B, et al: Islet function in off-spring of mothers
on low-protein diet during gestation, Diabetes 40:115-120, 1991.
Wilson MR, Hughes SJ: The effect of maternal protein deficiency during pregnancy and lactation on glucose tolerance and pancreatic islet function in adult
rat offspring, J Endocrinology 154:177-185, 1997.
Zeitlin J, Ancel P, Saurel-Cibizolles M, Papiernik E: The relationship between
IUGR and preterm delivery: an empirical approach using data from a European
case-control study, Brit J Obstet Gynecol 107:750-758, 2000.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
7
Multiple Gestations and Assisted
Reproductive Technology
Kerri Marquard and Kelle Moley
EPIDEMIOLOGY OF MULTIPLES
Since the birth of the first in vitro fertilization (IVF) baby
in 1978, the numbers of IVF clinics, ovarian stimulation
cycles, and live births from assisted reproductive technology (ART) have all steadily increased. Between 1998 and
2003, total births in the United States increased by 4%,
while ART births grew by 67% (Dickey, 2007). The growing use of ART, in addition to delayed childbearing until
age-related fertility issues become apparent, has contributed greatly to multiple birth rates. In 2006 ART infants
accounted for 1% of all U.S. births, but represented 17%
of twins and 38% of triplets or greater (Centers for Disease
Control and Prevention et al, 2008; Sunderam et al, 2009).
Three percent of all U.S. births are multiples, yet in 2003
ART multiple live birth rates from fresh nondonor, donor
oocyte, and frozen embryo transfer cycles were 34%, 40%,
and 25%, respectively (Centers for Disease Control and
Prevention et al, 2008).
The assisted conception and spontaneous rates associated with twins, triplets, and higher-order multiples
(HOMs; i.e., four or more fetuses) are as follows. In 2003,
62.7%, 16.3%, and 21% of twins were conceived naturally, from ART, and from non-ART ovulation induction,
respectively. For triplets in the same year, 17.7% were
natural conceptions, 45.4% were from ART, and 36.9%
were from non-ART ovulation induction. HOM rates
from spontaneous conception, ART, and non-ART ovulation induction were 8%, 30%, and 62.4%, respectively
(Dickey, 2007). Given the maternal, perinatal, and neonatal
complications associated with multiples, the goal of infertility treatment is one healthy child. Multifetal pregnancies drastically affect individuals, families, and the public
health system. Of particular importance in both maternal
and fetal outcomes are fetal number and placentation.
et al, 1995) At 10 to 14 weeks’ gestation, ultrasound criteria
correctly diagnosed chorionicity in 99.3% of cases as confirmed by postpartum placental examination and gender
(Carroll et al, 2002). Assessment of zygosity and chorionicity in 237 same-sex twins with physical likeness questionnaires, DNA analysis, and placental inspection accurately
diagnosed 96% of twin pairs (Forget-Dubois et al, 2003).
ZYGOSITY AND CHORIONICITY
Zygosity and placentation affect fetal morbidity and mortality in multifetal pregnancies. Dizygotic twins (DZTs),
which comprise 67% of spontaneous twins, arise from
the fertilization of two separate eggs by different sperm
(Gibbs et al, 2008), and with few exceptions they lead to a
dichorionic diamniotic arrangement in which the placenta
can be separate or fused. A rare case of dizygotic monochorionic (MC) diamniotic (DA) twins has been reported
(Souter et al, 2003). The overall DZT rate varies from 4 to
50 per 1000 worldwide: 1.3% in Japan, 7.1% to 11% in the
United States, 8.1% in India, 8.8% in England and Wales,
and 49% in Nigeria (Table 7-1) (MacGillivray, 1986). Risk
factors for DZT include advancing maternal age, increased
parity, female relatives with DZT, taller height, and larger
body mass index (Hoekstra et al, 2008; MacGillivray, 1986).
The true incidence of monozygotic twins (MZTs) is
difficult to ascertain because of its rarity, inaccuracies in
diagnosis, and lack of confirmatory studies at birth, but
spontaneous MZT rates are estimated to occur in 0.3%
to 0.5% of all pregnancies and in 30% of all twins (Bulmer, 1970; MacGillivray, 1986). Unlike DZT, it is unclear
whether MZT is related to genetics or environment (Bortolus et al, 1999; Hoekstra et al, 2008), although familial
components may play a role (Hamamy et al, 2004). Chorionicity in monozygotic gestations is determined by the
DIAGNOSING ZYGOSITY
AND CHORIONICITY
Determining zygosity and chorionicity is important medically, genetically, and psychosocially for the individual and
family. More immediately, the chorion-amnion arrangement is crucial to antepartum management in cases of one
fetal demise or selective reduction, and because of potential associated problems such as twin-twin transfusion syndrome (TTTS), growth discordance, intrauterine growth
restriction (IUGR), congenital anomalies, and cord accidents. Diagnosing zygosity is possible using ultrasound
markers including the number of placental sites, thickness of dividing membrane, the lambda sign, and fetal
gender in addition to postpartum placental examination,
physical similarity questionnaires, blood type, and DNA
analysis (Hall, 2003; Ohm Kyvik and Derom, 2006; Scardo
60
TABLE 7-1 Twinning Rates per 1000 Births by Zygosity
Country
Nigeria
Monozygotic
Dizygotic
Total
5.0
49
54
African American
4.7
11.1
15.8
Caucasian
4.2
7.1
11.3
England and Wales
3.5
8.8
12.3
India
3.3
8.1
11.4
Japan
3.0
1.3
4.3
United States
From MacGillivray I: Epidemiology of twin pregnancy, Semin Perinatol, 10:4-8, 1986;
and Cunningham FG, Hauth JC, Wenstrom KD, et al, editors: Williams obstetrics,
ed 22, New York, 2005, McGraw-Hill.
CHAPTER 7 Multiple Gestations and Assisted Reproductive Technology
Zygosity
Dizygotic or Monozygotic
Monozygotic
Day of division
0–3 days
0–3 days
4–8 days
8–13 days
Fetal
membranes
2 Amnions,
2 Chorions,
2 Placentas
2 Amnions,
2 Chorions,
1 Placenta
2 Amnions,
1 Chorion,
1 Placenta
1 Amnion,
1 Chorion,
1 Placenta
A
61
B
C
D
FIGURE 7-1 Placentation and membranes based on timing of embryonic division. A, Two amnions, two chorions, and separate placentas
from the division of either a dizygotic or monozygotic embryo within 3 days of fertilization. B, Two amnions, two chorions, and one fused placenta
from the division of either a dizygotic or monozygotic embryo within 3 days of fertilization. C, Two amnions, one chorion, and one placenta from
monozygotic embryonic cleavage, days 4 to 8 after fertilization. D, One amnion, one chorion, and one placenta from a monozygotic embryo splitting,
days 8 to 13 after fertilization. (Modified from Gibbs R, Karlan B, Haney A, et al: Danforth’s obstetrics & gynecology, ed 10, Philadelphia, 2008, Lippincott,
Williams & Wilkins.)
timing of the embryonic division (Figures 7-1 and 7-2)
(Benirschke and Kim, 1973; Hall, 2003). In 18% to 36% of
MZTs, the zygote divides within 72 hours of fertilization
resulting in dichorionic (DC) DA gestation (the placenta
can be separate or fused); 60% to 75% split between days 4
and 8, leading to an MC-DA unit, and 1% to 2% separate
between days 8 and 13, leading to an MC monoamniotic
(MA) pregnancy. Embryonic division after day 13 results in
conjoined twins with an MC/MA placenta (Cunningham
et al, 2005; Gibbs et al, 2008; Hall, 2003).
Although the majority of ART MZTs are MC/DA, any
of the three MZ placental arrangements can transpire after
ART, implying that the timing and mechanism of embryonic splitting are variable (Aston et al, 2008; Knopman et al,
2009). Monozygotic DCDA twins can occur after inner
cell mass (ICM) splitting and atypical hatching in a blastocyst embryo (Meintjes et al, 2001; Van Langendonckt
et al, 2000). In addition, MA twinning may be increased
after IVF (Alikani et al, 2003) and zona pellucida (ZP)
manipulation (Slotnick and Ortega, 1996). Contributing
factors to MZT in ART include ICM damage and other
ZP abnormalities (Hall, 2003).
INCREASE IN MONOZYGOTIC TWINS
WITH ASSISTED REPRODUCTIVE
TECHNOLOGY
The first reported association between ART and MZT
(Yovich et al, 1984) preceded numerous accounts of similar findings. The majority (>90%) of ART twins are dizygotic (Gibbs et al, 2008) secondary to transferring multiple
embryos; however, the rate of MZTs per pregnancy after
fertility treatment is higher (0.9% to 4.9%) (Alikani et al,
2003; Blickstein et al, 2003; Elizur et al, 2004; Knopman et al, 2009; Papaniklaou et al, 2009; Schachter et al,
2001; Sharara and Abdo, 2010; Sills et al, 2000; Vitthala
et al, 2009; Wenstrom et al, 1993) versus the general population (0.3% to 0.5%) (Bulmer, 1970; MacGillivray, 1986).
Several theories to explain the mechanism responsible for
elevated MZTs with ART have been proposed.
AGE
Maternal age affects fertility and reproductive outcomes.
Spontaneous dizygotic twinning increases with advancing maternal age (Bortolus et al, 1999; Bulmer, 1970;
MacGillivray, 1986), but the connection between age
and MZTs is controversial. Some studies reported trends
toward elevated MZT rates in older women (Abusheikha
et al, 2000; Alikani et al, 2003; Bulmer, 1970), whereas
others found no association with increasing maternal age
and MZT (Bortolus et al, 1999; Skiadas et al, 2008), and
one study found that the MZT risk doubles in women
younger than 35 years (Knopman et al, 2009). Overall,
the correlation between age and MZT in ART remains
unclear.
ZONA PELLUCIDA MANIPULATION
The ZP, an acellular protein surrounding the ovum, provides
a species-specific sperm barrier and decreases polyploidy by
inhibiting penetration by multiple sperm (Speroff and Fritz,
2005). Components of both ART and non-ART procedures
are capable of modifying this barrier. Stimulation protocol,
elevated follicle-stimulating hormone or estradiol levels,
and prolonged culture conditions can change ZP thickness
(Loret et al, 1997). In addition, ZP manipulations performed
during IVF all potentially affect MZT risk.
Manipulation of the ZP in IVF occurs via both intracytoplasmic sperm injection (ICSI) and AH (assisted hatching). The injection of one sperm into a mature oocyte
(i.e., ICSI) is most commonly performed for male factor
infertility. AH is achieved with an artificial breach in the
ZP by laser, chemical, or mechanical methods and is indicated in patients with a poor prognosis (Practice Committee of Society for Assisted Reproductive Technology and
Practice Committee of American Society for Reproductive
Medicine, 2008c). Some studies indicate no association
with ICSI or AH and MZT (Behr et al, 2000; Elizur et al,
2004; Knopman et al, 2009; Meldrum et al, 1998; Milki
et al, 2003; Sills et al, 2000), whereas others imply that
62
PART II Fetal Development
Embryo Yolk sac
A
Inner cell
cell mass
Embryo
Amniotic
cavity
B
Yolk sac
Two-cell
embryo
Blastocoel
Amniotic
cavity
Chorionic
cavity
Embryo
C
Yolk sac
FIGURE 7-2 Types of monozygotic placentation. A, Dichorionic diamniotic pregnancy. B, Monochorionic diamniotic pregnancy. C, Monochorionic monoamniotic pregnancy. (Adapted from Hall JG: Twinning, Lancet 362:735-743, 2003; and Benirschke K, Kim CK: Multiple pregnancy, N Engl J
Med 288:1276-1284, 1973.)
ZP manipulation increases the risk of MZT (Alikani et al,
1994; Saito et al, 2000; Schieve et al, 2000; Skiadas et al,
2008; Vitthala et al, 2009), particularly if multiple embryos
undergo hatching before transfer (Alikani et al, 2003;
Schieve et al, 2000). The results of a Cochrane review on
the risk of multiples with AH and ICSI reported increased
multiples with both ICSI and AH, and elevated MZT with
AH versus no manipulation (0.8%) (Das et al, 2009). Alterations in ZP thickness or abnormal ICM splitting or embryo
hatching in both animal (Cohen, 1991; Malter and Cohen,
1989) and human embryos may explain this increase in
MZT after ZP manipulation (Alikani et al, 1994; Cohen,
1991; Malter and Cohen, 1989; Sheen et al, 2001).
BLASTOCYST TRANSFER
After oocyte retrieval and insemination, embryos undergo
intrauterine transfer at either the cleavage stage (day 2 to
3 after retrieval) or the blastocyst stage (day 5 to 6 after
retrieval). Blastocyst-stage embryo transfer produces
higher pregnancy rates (29.4% vs 36%) (Blake et al, 2007)
and may lower overall multiple rates (Frattarelli et al,
2003), but evidence supports the increased incidence of
MZTs (Behr et al, 2000; Chang et al, 2009; Jain et al,
2004; Knopman et al, 2009; Peramo et al, 1999; Practice Committee of American Society for Reproductive
Medicine and Practice Committee of Society for Assisted
Reproductive Technology, 2008a; Sheiner et al, 2001;
Skiadas et al, 2008; Wright et al, 2004) compared with
cleavage-stage embryos. One institution initially noted
increased MZT after blastocyst transfer (5.6% vs 2%;
Milki et al, 2003), but a follow-up study 3 years later
demonstrated similar MZT rates between blastocyst and
day 3 embryos (2.3% vs 1.8%), indicating that changes in
culture media and an experienced embryology team may
affect the rate of MZTs (Moayeri et al, 2007).
Culture media or prolonged culture may influence
MZTs with blastocyst transfer in both human and animal
models. Elevated glucose levels in extended culture and
subsequent glucose-induced apoptotic remodeling of the
ICM (Cassuto et al, 2003; Menezo and Sakkas, 2002), and
ICM splitting in extended culture in murine and human
embryos are both explanations for the phenomenon of
MZTs in blastocysts (Chida, 1990; Hsu and Gonda, 1980;
Payne et al, 2007). Whether MZT rates are elevated with
blastocyst transfer (Behr et al, 2000; Chang et al, 2009;
Jain et al, 2004; Knopman et al, 2009; Peramo et al, 1999;
Practice Committee of American Society for Reproductive
Medicine and Practice Committee of Society for Assisted
Reproductive Technology, 2008a; Sheiner et al, 2001; Skiadas et al, 2008; Wright et al, 2004), or similar to cleavagestage transfer (Frattarelli et al, 2003; Papanikolaou et al,
2006, 2010; Sharara and Abdo, 2010), the explicit role of
culture media remains to be determined. Another environmental factor that may affect MZTs is temperature variation in frozen-thawed embryo cycles. Although minute
evidence links frozen embryo transfers and temperature
fluctuations to MZTs (Belaisch-Allart et al, 1995; Faraj
et al, 2008; Toledo, 2005), most studies show no difference between fresh and frozen-thawed embryos and multiple rates (Blickstein et al, 2003) or MZT rates (Alikani
et al, 2003; Knopman et al, 2009; Sills et al, 2000a, 2000b).
OVARIAN STIMULATION
Human studies of ovarian stimulation with clomiphene
citrate and gonadotropins reveal a higher rate (1.2%) of
MZTs compared with the expected rates in the general
63
CHAPTER 7 Multiple Gestations and Assisted Reproductive Technology
population (Derom et al, 1987). MZT incidences of 1.5%
after ovulation induction with gonadotropins, 0.72% after
IVF, and 0.87% with IVF ICSI-AH suggests that gonadotropins elevate MZTs regardless of ZP manipulation
(Schachter et al, 2001). Ovulation induction medications
may cause uneven hardening of the ZP and atypical blastocyst hatching thereby increasing the chance of MZTs
(Derom et al, 1987).
FETAL COMPLICATIONS ASSOCIATED
WITH MULTIPLES
Singleton pregnancies after assisted conception have
increased complications, including preterm delivery (<37
weeks’ gestation), low birthweight (LBW) (Helmerhorst
et al, 2004; Schieve et al, 2007), prolonged hospital stay
(Schieve et al, 2007), cesarean deliveries, neonatal intensive care unit (NICU) admission, and mortality compared
with spontaneous singletons (Helmerhorst et al, 2004).
Some risk persists in an IVF singleton pregnancy, even
after spontaneous reduction from two to three initial
heartbeats to one heartbeat (Luke et al, 2009). Although
singleton IVF births are associated with morbidity and
mortality, assisted-conception multiple gestations comprise the majority of adverse maternal, perinatal, and neonatal outcomes.
Multiple pregnancies account for a small percentage of
overall live births, but are responsible for a disproportionate amount of morbidity and mortality, largely because of
intrauterine growth restriction and prematurity (Garite
et al, 2004), including 13% of all preterm deliveries, 21%
of all LBW infants, and 25% of all very LBW infants (Robinson et al, 2001). Compared with a singleton pregnancy,
fetal and maternal complications are elevated in twins, and
pregnancies with three fetuses or more have even greater
morbidity and mortality rates (Table 7-2) (ACOG Practice
Bulletin #56, 2004; Albrecht and Tomich, 1996; Elliott and
Radin, 1992; Ettner et al, 1997; Grether et al, 1993; Kiely
et al, 1992; Luke, 1994; Luke and Keith, 1992; Luke et al,
1996; Martin et al, 2003; Mauldin and Newman, 1998;
McCormick et al, 1992; Newman et al, 1989; Seoud et
al, 1992). Higher fetal number correlates with increased
risk of growth restriction, earlier delivery, LBW, neonatal
intensive care unit admission, length of stay, risk of major
handicap and cerebral palsy, and death in first year of life
(ACOG Practice Bulletin #56, 2004; Gardner et al, 1995;
Garite et al, 2004).
The average gestational ages for twin, triplet, and quadruplet deliveries are 35.3, 32.2, and 29.9 weeks, respectively (ACOG Practice Bulletin #56, 2004), corresponding
to NICU admission rates fivefold higher in twins and
17-fold higher in triplets and HOMs (Ross et al, 1999).
Twins have an increased risk of intrauterine fetal demise
(fourfold), intraventricular hemorrhage, sepsis, necrotizing enterocolitis, respiratory distress syndrome and neonatal death (sixfold) versus singletons, and surviving infants
of preterm multifetal pregnancies have higher rates of
developmental handicap (Gardner et al, 1995). A review
of 100 triplet gestations (88 with assisted conception)
revealed that 78% experienced preterm labor (PTL), 14%
delivered before 28 weeks, 5% had congenital anomalies,
and 9.7% died in the perinatal period (Devine et al, 2001).
TABLE 7-2 Morbidity and Mortality by Fetal Number
Characteristic
Twins
Triplets
Average birthweight
2347 g
1687 g
Quadruplets
1309 g
Average gestational
age at delivery
35.3 wk
32.2 wk
29.9 wk
Percentage with
growth restriction
14-25
50-60
50-60
Percentage
requiring admission to neonatal
intensive care unit
25
75
100
Average length of
stay in neonatal
intensive care unit
18 days
30 days
58 days
Percentage with
major handicap
-
20
50
Risk of cerebral
palsy
4 times
more
than
singletons
17 times
more
than
singletons
-
Risk of death by age
1 year
7 times
higher
than
singletons
20 times
higher
than
singletons
-
From ACOG Practice Bulletin #56: Multiple gestation: complicated twin, triplet, and
high-order multifetal pregnancy, Obstet Gynecol 104:869-883, 2004.
Similar to ART singleton versus spontaneous singleton outcomes, ART multiples may have higher morbidity
compared with spontaneous multiples. Assisted-conception
twins are at increased risk for LBW (Luke et al, 2009), preterm delivery (Luke et al, 2009; Nassar et al, 2003), cesarean
delivery (Helmerhorst et al, 2004; Nassar et al, 2003), NICU
admission (Helmerhorst et al, 2004; Nassar et al, 2003; Pinborg et al, 2004), longer length of stay, respiratory distress
syndrome (Nassar et al, 2003), and birthweight discordance
(Pinborg et al, 2004) versus spontaneously conceived twins.
Oftentimes with IVF, because of multiple embryos transferred or MZ splitting, there are multiple heartbeats on an
initial ultrasound examination that ultimately spontaneously
reduce; however, these pregnancies are still at risk for an
adverse outcome. In twin IVF cycles with two initial heartbeats on early ultrasound versus three heartbeats that spontaneously reduced to two heartbeats, pregnancies with three
heartbeats on an early examination had increased rates of
preterm delivery (35%) and LBW (47%) (Luke et al, 2009).
In contrast, other studies suggest comparable outcomes
in assisted conception and spontaneous multiples. Rates
of pregnancy-induced hypertension, gestational diabetes
mellitus (GDM), preterm premature rupture of membranes (PPROM), placenta previa, placental abruption
(Nassar et al, 2003), congenital malformations (Nassar
et al, 2003; Pinborg et al, 2004), and mortality (Pinborg
et al, 2004) were similar in IVF twins versus spontaneous
twins. Similarly, morbidity and mortality in ART triplets
versus spontaneous triplets were comparable regarding the
rates of PPROM, PTL, pregnancy-induced hypertension,
GDM, gestational age at delivery, birthweight, and NICU
admissions (Fitzsimmons et al, 1998).
Besides fetal number, another important factor in pregnancy outcome is placental arrangement. MC multiples
64
PART II Fetal Development
attributed to increased prenatal diagnosis and fetal surveillance (Allen et al, 2001; Heyborne et al, 2005; Rodis
et al, 1997).
TABLE 7-3 Incidence of Twin Pregnancy Zygosity
and Chorionicity With Corresponding Complications
Complication (%)
Type of
Twinning
Dizygotic
Delivery
Incidence
<37 Weeks’ Perinatal
(%)
IUGR Gestation Mortality
67-70
25
40
10-12
Monozygotic
30-33
40
50
15-18
Diamniotic-
dichorionic
18-36
30
40
18-20
Diamniotic-
monochorionic
60-75
50
60
30-40
Monoamnioticmonochorionic
1-2
40
60-70
30-70
From Cunningham FG, Hauth JC, Wenstrom KD, et al, editors: Williams obstetrics,
ed 22, New York, 2005, McGraw-Hill; originally from Manning FA: Fetal biophysical
profile scoring. In Fetal medicine: principles and practices, Norwalk, Conn, 1995,
Appleton & Lange.
IUGR, Intrauterine growth restriction.
experience higher rates of morbidity and mortality, largely
because of placental factors (Table 7-3) (Cunningham
et al, 2005; Gaziano et al, 2000; Hack et al, 2008;
Manning, 1995). When Dube et al (2002) studied different
chorionicity–zygosity groups (monozygotic monochorionic
[MZMC], di zygotic dichorionic [DZDC], and monozygotic dichorionic [MZDC]) they found smaller birthweight,
and IUGR, congenital anomalies, and perinatal death in the
MZMC versus DZDC twins, whereas MZDC and DZDC
risks were similar, implying that poor outcomes are related
more to chorionicity than zygosity. Negative outcomes such
as cerebral palsy, mental retardation, and death, measured
at 1 year of life in MC twins, are elevated (10%) compared
with DC twins (3.7%), the majority of which are caused by
complications from TTTS (Minakami et al, 1999).
Both placental asymmetry and abnormal vascular anastomosis affect fetal morbidity and mortality. IUGR and
growth discordance afflict both DC and MC pregnancies,
but MC twins are more likely to have cord abnormalities, unequal placental distribution, and TTTS (ClearyGoldman and D’Alton, 2008). TTTS, ranging in severity
from oligohydramnios to hydrops and fetal death (ClearyGoldman and D’Alton, 2008; Gaziano et al, 2000) occurs
in 15% to 32% of MC pregnancies (Hack et al, 2008;
Minakami et al, 1999). Mortality rates vary from 22% to
100% (Bajoria et al, 1995; Cleary-Goldman and D’Alton,
2008; Hack et al, 2008), and surviving infants are at risk
for long-standing adverse neurologic outcomes (ClearyGoldman and D’Alton, 2008).
MC-MA twins occur in 1 in 10,000 pregnancies, but
they suffer the highest risk of perinatal morbidity and mortality (Cordero et al, 2006). Similar to other MC twins,
MC-MA twins are susceptible to TTTS, growth discordance, IUGR, preterm delivery, and congenital anomalies,
but they also face the unique complication of cord entanglement. These factors historically account for perinatal
mortality rates of 30% to 70%; however, lower morbidity and mortality rates (8% to 23%) reported in recent
articles (Allen et al, 2001; Cordero et al, 2006; Heyborne
et al, 2005; Rodis et al, 1997; Roque et al, 2003) may be
MATERNAL COMPLICATIONS
Approximately 80% of multiples experience antepartum
complications versus 25% of singletons (Norwitz et al,
2005), and hospitalization for hypertensive disorders,
PTL, PPROM, placental abruption, and postpartum
hemorrhage are elevated sixfold (ACOG Practice Bulletin #56, 2004). Mothers with two or more fetuses are at
increased risk for myocardial infarction, left ventricular
heart failure, pulmonary edema, GDM, operative vaginal or cesarean delivery, hysterectomy, blood transfusion, longer hospital stay, and the three major causes of
maternal mortality: post partum hemorrhage, venous
thromboembolism, and hypertensive disorders (Walker
et al, 2004).
Stratified by fetal number, plurality correlates with
maternal morbidity where quadruplets and other HOMs
experience significantly increased maternal morbidity
versus twins and triplets (Wen et al, 2004). Hypertensive disorders occur in 12% to 20% of twins, triplets, and
quadruplets compared with 6.5% of singletons, HELLP
syndrome increases with higher numbers of fetuses (Day
et al, 2005), and twins with preeclampsia experience more
complications than singletons with preeclampsia (Sibai
et al, 2000). Results of a triplet cohort showed that 96%
had maternal complications, 96% required antenatal hospitalization, one in four were diagnosed with preeclampsia,
and 44% encountered postpartum complications (Devine
et al, 2001).
PSYCHOSOCIAL FACTORS
As fetal number in an assisted conception pregnancy
increases, parents report decreased quality of life, increased
social stigma, increased difficulty meeting material family needs (Ellison et al, 2005; Roca de Bes et al, 2009),
increased depression (Ellison et al, 2005; Olivennes et al,
2005; Sheard et al, 2007), decreased marital satisfaction
(Roca de Bes et al, 2009), increased fatigue (Sheard et al,
2007) and stress (Golombok et al, 2007; Olivennes et al,
2005; Sheard et al, 2007). Part of this challenge lies in the
fact that there are 168 hours in 1 week, but adequately
caring for 6-month-old triplets and household activities
requires 197.5 hours per week (Bryan, 2003). Multiple
fetuses themselves face an increased risk of long-term disabilities that contribute to increased parental fatigue and
depression, and overall siblings of multiples are more at
risk for behavioral issues (Bryan, 2003).
Psychosocial consequences between naturally conceived multiples versus IVF multiples might differ. On
the one hand, parents of IVF multiples can experience
significantly more stress, increased child difficulty (Cook
et al, 1998; Glazebrook et al, 2004), and increased dysfunctional parent-child interactions (Glazebrook et al,
2004) compared with spontaneous multiples. On the
other hand, both ART and non-ART twin conceptions
are more stressful for parents, creating higher levels of
anxiety and depression compared with singletons (Vilska
CHAPTER 7 Multiple Gestations and Assisted Reproductive Technology
et al, 2009). Regardless of conception mode, multiples
potentially have negative psychosocial effects on parents
and families.
COST
Multiple gestations economically influence both the family and society. Preterm delivery, LBW, and postdischarge
hospitalization are increased in multiple fetuses, all of
which have a role in short- and long-term cost (Cuevas
et al, 2005). Annually, neonatal health care consumes $10.2
billion in the Unites States, 57% of which comes from
preterm infants (<37 weeks’ gestation) who comprise less
than 10% of live births (St. John et al, 2000). According to
gestational age, the mean initial hospital charge for infants
born between 26 and 28 weeks’ gestation is approximately
$240,000 compared with approximately $4800 for a term
infant. By birthweight, infants weighing less than 1250
g cost approximately $250,000 compared with infants
weighing more than 2500 g, who cost $5800 (Cuevas
et al, 2005).
ART multiples and their associated comorbidities have a
significant role in health care expenditures. In the United
Kingdom, IVF-induced multiples account for 27% of
pregnancies yearly, but represent 54% of expenditures
(Ledger et al, 2006); this is due largely to IVF triplets
and twins, which are remarkably more costly than IVF
singletons from both neonatal and maternal standpoints.
Estimated maternal cost ratios for IVF singleton:twin
and singleton:triplet are approximately 1:1.94 and 1:3.96,
respectively, and neonatal cost ratios for IVF singleton:twin
and singleton:triplet are 1:16 and 1:109, respectively (Ledger et al, 2006). The cost for IVF triplets from diagnosis
through 1 year of life is tenfold (Ledger et al, 2006), and
IVF twin cost up to 6 weeks postpartum is fivefold versus
IVF singletons (Lukassen et al, 2004). ART singletons are
more costly than spontaneous singletons, possibly because
of increased rates of LBW (Chambers et al, 2007) or the
underlying infertility contributing to these outcomes
(Koivurova et al, 2004). An Australian study comparing
IVF singletons, twins, and HOMs to control counterparts
revealed no significant cost differences between ART
twins and non-ART twins or between ART HOMs and
non-ART HOMs, but combined neonatal-maternal cost
was 57% higher for ART births than for non-ART births
(Chambers et al, 2007).
Because of the extreme economic cost of multiples, one
proposed mechanism to reduce multifetal pregnancies is
single embryo transfer (SET). Elective SET in first-cycle
IVF patients costs less than double embryo transfer (DET)
(Fiddelers et al, 2006; Gerris et al, 2004), and although
SET produced lower live birth rates than DET in an
unselected population (20.8% versus 39.6%) (Fiddelers
et al, 2006), when stratified to less than 38 years age, SET
and DET generate similar live birth rates (Gerris et al,
2004).
DECREASING THE RISK OF MULTIPLES
ART procedures and the rate of multiple pregnancies are
rising concordantly. Although ART triplets and the incidence of HOMs has declined since 1996, ART twin rates
65
are unchanged.2 Primary forms of preventing multiples
include canceling ovulation induction cycles or converting
to IVF, and in IVF cycles limiting the number of embryos
transferred. Worldwide differences exist in medical practice and laws regarding restrictions on the number of
embryos transferred.
The American Society for Reproductive Medicine
(ASRM) and the Society for Assisted Reproductive Technology established transfer guidelines to assist in determining the appropriate embryo number in an attempt to
decrease multiples. Recommended limits are based on
age, prognosis, and embryo stage and further differentiate
between a favorable patient (first IVF cycle, good quality
embryos, number of embryos for potential cryopreservation, successful past IVF cycles) and less favorable conditions. To date there are no embryo transfer guidelines for
frozen embryo cycles. Transfer recommendations by the
ASRM are not legally binding, and they are subject to interpretation or adjustment based on clinical experience and
unique patient instances (Practice Committee of Society for
Assisted Reproductive Technology and Practice Committee of American Society for Reproductive Medicine, 2008b).
Reduction of the number of embryos transferred and the
incidence of triplets or greater in women aged 37 years or
less from 1996 to 2003 may be attributed to a change in the
1998-1999 ASRM guidelines (Stern et al, 2007).
The most important factor involved in creating multiple
fetuses is the number of embryos transferred. Given the
emotional, physical, and financial burden of ART and the
concern that only one embryo might lower pregnancy rates,
multiples fetuses were previously accepted as a known risk
in an effort to ensure a pregnancy. More recently, however, practitioners across the globe are stressing the impact
of multiples and are encouraging SET (Gerris, 2005).
A Swedish study in women aged less than 36 years
undergoing their first or second IVF cycle with two or
more good-quality embryos were randomized to DET or
SET followed by frozen-thawed embryo transfer if unsuccessful. Live birth rates were lower in the SET-alone versus the DET group, but SET followed by frozen-thawed
embryo transfer resulted in a 38.8% live birth rate and a
0.8% multiple rate, compared with a 42.9% live birth rate
and a 33.1% multiple rate in the DET group, showing that
with SET pregnancy rates were acceptable and multiple
pregnancy rates were significantly lower (Thurin et al,
2004). Women aged 36 to 39 years may also be candidates
for SET, because similar live birth rates between SET and
DET and significantly higher cumulative multiple rates
occur in the DET (16.6%) versus SET (1.7%) groups
(Veleva et al, 2006). Superb cryopreservation technique
with frozen embryo transfer after SET in the appropriate patient lowers multiple fetus rates (Gerris, 2005) and
leads to comparable cumulative LBR compared with DET
(Veleva et al, 2009).
Consideration of multiples from non-ART ovulation
induction by controlled ovarian hyperstimulation also
merits discussion. Twenty-two percent of twins, 40% of
triplets, and 71% of HOMs in 2004 were a result of nonART ovulation induction (Dickey, 2009). During gonadotropin stimulation, follicular growth is supervised via
ultrasound examination, and estradiol levels are monitored
in an attempt to minimize overstimulation. Ovulation
66
PART II Fetal Development
induction risk factors for multiples have revealed that
HOMs are positively correlated with gonadotropin dose
and stimulation length, estradiol levels greater than 1000
pg/mL, and seven or more follicles measuring 10 mm or
greater, whereas negative predictors were age less than 32
years, lower body mass index, and a higher number of prior
treatment cycles (Dickey, 2009). Techniques to reduce the
chance of multiple fetuses include minimizing gonadotropin dose, canceling the cycle by discontinuing medications
for excess follicles or high estradiol levels, or converting
to IVF (Dickey, 2009; Nakhuda and Sauer, 2005); however, the specific criteria warranting cycle cancelation are
not uniform between centers (Practice Committee of the
American Society for Reproductive Medicine, 2006).
MULTIFETAL PREGNANCY
REDUCTION
As the rates of ART procedures, multiple fetuses, and
prematurity-related sequelae have increased, so has the
use of selective reduction. Primary prevention of multiple
fetuses by limiting the number of embryos transferred or
canceling an overstimulated ovulation induction cycle is
optimal; however, in reality multifetal pregnancies continue to occur. Multifetal pregnancy reduction (MFPR)
provides another option to enhance overall survival and
decrease the risk of fetal or neonatal morbidity and mortality by decreasing pregnancy loss rates and prematurity
(Evans and Britt, 2005). First developed in the 1980s,
selective termination of one or more fetuses is performed
to reduce the final fetal number. The majority of patients
reduce to twins, followed by singletons; few reduce to triplets (Stone et al, 2008). A discussion of the ethical, medical,
and psychosocial factors involved in MFPR are important
counseling points for any patient undergoing ovulation
induction (Committee on Ethics, 2007).
Improvements in MFPR techniques have enhanced
success rates such that quadruplet or triplets reduced to
twins have equal outcomes compared with natural twins
(Evans and Britt, 2005; Evans et al, 2001). Success rates
correlate with both beginning and ending fetal number
(Evans et al, 2001). The average loss rate in one series of
1000 MFPR cases was 4.7% (Stone et al, 2008). Loss rates
are higher after reducing to a singleton versus reducing
to twins, but twins overall have higher morbidity than do
singletons (Evans and Britt, 2005, 2008; Evans et al, 2001;
Stone et al, 2008). Reduction of twins to singletons may be
considered given a lower loss rate after reduction versus
continuing with twins (Evans et al, 2004). Benefits after
MFPR are apparent in preterm delivery rates, because half
of twins and almost 90% of singletons are delivered full
term, and 95% were delivered after 24 weeks’ gestation
in one series (Stone et al, 2008). Although beneficial in
certain cases, MFPR is not without medical and psychological risk. It might not be an option for some women;
therefore primary prevention should be the focus for
reducing the risk of multiple fetuses.
SUMMARY
Over the last 30 years, advances in ART have helped countless infertile couples achieve a pregnancy. The percentage
of ART live births will likely continue on an upward trend
because of increased accessibility of ART and delayed
childbearing. These factors in addition to ART techniques
will continue to contribute to multiple gestation rates.
Multiple gestations are associated with increased maternal,
fetal, and neonatal complications that generate a medical,
psychological, and economic burden to families and society. Efforts to decrease multifetal pregnancies and prematurity-related sequelae include prevention-based practice
policies and further knowledge regarding the mechanisms
involved with MZT and ART.
SUGGESTED READINGS
ACOG Practice Bulletin #56: Multiple gestation: complicated twin, triplet, and
high-order multifetal pregnancy, Obstet Gynecol 104:869-883, 2004.
Aston KI, Peterson CM, Carrell DT: Monozygotic twinning associated with
assisted reproductive technologies: a review, Reproduction 136:377-386, 2008.
Chambers GM, Chapman MG, Grayson N, et al: Babies born after ART treatment
cost more than non-ART babies: a cost analysis of inpatient birth-admission
costs of singleton and multiple gestation pregnancies, Hum Reprod 22:31083115, 2007.
Dickey RP: The relative contribution of assisted reproductive technologies and
ovulation induction to multiple births in the United States 5 years after
the Society for Assisted Reproductive Technology/American Society for
Reproductive Medicine recommendation to limit the number of embryos
transferred, Fertil Steril 88:1554-1561, 2007.
Ellison MA, Hotamisligil S, Lee H, et al: Psychosocial risks associated with multiple
births resulting from assisted reproduction, Fertil Steril 83:1422-1428, 2005.
Evans MI, Britt DW: Fetal reduction, Semin Perinatol 29:321-329, 2005.
Hack KE, Derks JB, Elias SG, et al: Increased perinatal mortality and morbidity in
monochorionic versus dichorionic twin pregnancies: clinical implications of a
large Dutch cohort study, BJOG 115:58-67, 2008.
Hall JG: Twinning, Lancet 362:735-743, 2003.
Knopman J, Krey LC, Lee J, et al: Monozygotic twinning: an eight-year experience
at a large IVF center, Fertil Steril 94:502-510, 2010.
Practice Committee of the American Society for Reproductive Medicine: Multiple
pregnancy associated with infertility therapy, Fertil Steril 86(Suppl 1):
S106-S110, 2006.
Norwitz ER, Edusa V, Park JS: Maternal physiology and complications of multiple
pregnancy, Semin Perinatol 29:338-348, 2005.
Thurin A, Hausken J, Hillensjo T, et al: Elective single-embryo transfer versus double-embryo transfer in vitro fertilization, N Engl J Med 351:2392-2402, 2004.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
8
Nonimmune Hydrops
Scott A. Lorch and Thomas J. Mollen
An infant with hydrops has an abnormal accumulation
of excess fluid. The condition varies from mild, generalized edema to massive edema, with effusions in multiple
body cavities and with peripheral edema so severe that
the extremities are fixed in extension. Fetuses with severe
hydrops may die in utero; if delivered alive, they may die in
the neonatal period from the severity of their underlying
disease or from severe cardiorespiratory failure.
The first description of hydrops in a newborn, in a
twin gestation, may have appeared in 1609 (Liley, 2009).
Ballantyne (1892) suggested that the finding of hydrops was
an outcome for many different causes, in contrast to the
belief at that time that hydrops was a single entity. Potter
(1943) first distinguished between hydrops secondary to
erythroblastosis fetalis and nonimmune hydrops, by describing a group of infants with generalized body edema who
did not have hepatosplenomegaly or abnormal erythropoiesis. Potter’s description of more than 100 cases of hydrops
included two sets of twins in which one had hydrops and the
other did not, thus presenting the first description of twintwin transfusion syndrome. With the nearly universal use of
anti-D globulin and refinement of the schedule and doses
for its administration, the occurrence of immune-mediated
hydrops has steadily declined, such that later studies found
that immune-mediated causes accounted for only 6% to
10% of all cases of hydrops (Heinonen et al, 2000; Machin,
1989). The reported incidence of nonimmune hydrops in
the general population has been highly variable, ranging
from 6 per 1000 pregnancies in a high-risk referral clinic
in the United Kingdom between 1993 and 1999 (Sohan
et al, 2001) to 1 in 4000 pregnancies (Norton, 1994); other
published rates are 6 per 1000 pregnancies (Santolaya et al,
1992), 1.3 per 1000 pregnancies (Wafelman et al, 1999),
and 1 per 1700 pregnancies (Heinonen et al, 2000). However, all the published studies come from single institutions,
with the at-risk populations ranging from that of a highrisk pregnancy clinic to infants in a neonatal intensive care
unit. No study has monitored all pregnant women in one
geographic area to calculate the true population incidence
of nonimmune hydrops, especially monitoring infants who
died in utero. Geography also affects the incidence; several
causes of nonimmune hydrops, such as α-thalassemia, are
more common in certain areas of the world. Finally, the
incidence of nonimmune hydrops may be rising because of
the more routine use of ultrasound investigation in the late
first trimester of pregnancy (Iskaros et al, 1997).
ETIOLOGY
Nonimmune hydrops has been associated with a wide
range of conditions (Table 8-1). In many of these conditions, edema formation results from one of the following
possible processes:
ll
Elevated central venous pressure, in which the cardiac
output is less than the rate of venous return
ll
ll
ll
Anemia, resulting in high-output cardiac failure
Decreased lymphatic flow
Capillary leak
The actual pathophysiology of hydrops for many of the
conditions in Table 8-1, however, is still not understood.
The most common causes of nonimmune hydrops are
chromosomal, cardiovascular, hematologic, thoracic,
infectious, and related to twinning (Abrams et al, 2007;
Bellini et al, 2009; Wilkins, 1999). As with reported incidence rates, the relative contribution of these causes varies
by study. The studies that focus on early fetal presentation
of hydrops (postconceptional age of less than 24 weeks’
gestation) have found that chromosomal abnormalities,
such as Turner syndrome and trisomies 13, 18, and 21, are
the causes of 32% to 78% of all cases of hydrops (Boyd et
al, 1992; Heinonen et al, 2000; Iskaros et al, 1997; McCoy
et al, 1995; Sohan et al, 2001). For infants whose hydrops
becomes evident after 24 weeks’ gestation, cardiovascular
and thoracic causes are most prevalent, with rates ranging
between 30% and 50% (Machin, 1989; McCoy et al, 1995;
Shan et al, 2001). Studies from Asia have noted a higher
percentage of cases from hematologic causes, probably
because of the higher rates of α-thalassemia in the population (Lin et al, 1991; Nakayama et al, 1999).
The percentage of infants with “idiopathic” hydrops, or
hydrops of unknown etiology, varies from 5.2% to 50%,
depending on the ability of the clinicians to complete their
diagnostic evaluation and the inclusion of fetal deaths in the
analysis (Bellini et al, 2009; Heinonen et al, 2000; Iskaros
et al, 1997; Machin, 1989; McCoy et al, 1995; Nakayama
et al, 1999; Santolaya et al, 1992; Sohan et al, 2001; Wafelman et al, 1999; Wy et al, 1999). Yaegashi et al (1998)
used enzyme-linked immunosorbent assay and polymerase
chain reaction techniques to improve the detection of
parvovirus infection. In both their own institution, and in
eight other series of patients, these investigators found evidence of parvovirus infection in 15% to 19% of all infants
previously diagnosed with idiopathic hydrops. It is likely
that, as there is increased understanding of and testing for
many of the conditions listed in Table 8-1, the number
of infants diagnosed with idiopathic, nonimmune hydrops
will continue to decline.
PATHOPHYSIOLOGY
NORMAL FLUID HOMEOSTASIS
Abnormal body fluid homeostasis is the underlying cause
of edema, whether local or generalized. To understand the
pathogenesis of hydrops, the clinician must consider the
forces underlying normal fluid homeostasis. The regulation of net fluid movement across a capillary membrane
depends on the Starling forces, which were first described
by E.H. Starling in 1896 (Starling, 1896). Flow between
intravascular and interstitial fluid compartments is
67
68
PART II Fetal Development
TABLE 8-1 Conditions Associated With Hydrops Fetalis
Condition Type
Specific Conditions
Condition Type
Specific Conditions
Hemolytic anemias
Alloimmune, Rh, Kell,
α-Chain hemoglobinopathies (homozygous α-thalassemia)
Red blood cell enzyme deficiencies (glucose phosphate isomerase
deficiency, glucose-6-phosphate
dehydrogenase)
Nervous system lesions
Absence of corpus callosum
Encephalocele
Cerebral arteriovenous malformation
Intracranial hemorrhage (massive)
Holoprosencephaly
Fetal akinesia sequence
Other anemias
Fetomaternal hemorrhage
Twin-twin transfusion
Diamond-Blackfan
Pulmonary conditions
Cardiac conditions
Premature closure of foramen ovale
Ebstein anomaly
Hypoplastic left or right heart
Subaortic stenosis with fibroelastosis
Cardiomyopathy, myocardial fibroelastosis
Atrioventricular canal
Myocarditis
Right atrial hemangioma
Intracardiac hamartoma or fibroma
Tuberous sclerosis with cardiac rhabdomyoma
Cystic adenomatoid malformation of
the lung
Mediastinal teratoma
Diaphragmatic hernia
Lung sequestration syndrome
Lymphangiectasia
Renal conditions
Urinary ascites
Congenital nephrosis
Renal vein thrombosis
Invasive processes and storage disorders
Tuberous sclerosis
Gaucher disease
Mucopolysaccharidosis
Mucolipidosis
Supraventricular tachycardia
Atrial flutter
Congenital heart block
Chromosome
abnormalities
Trisomy 13, trisomy 18, trisomy 21
Turner syndrome
XX/XY
Bone diseases
Hemangioma of the liver
Any large arteriovenous malformation
Klippel-Trenaunay syndrome
Idiopathic infantile arterial calcification
Osteogenesis imperfecta
Achondroplasia
Asphyxiating thoracic dystrophy
Gastrointestinal
conditions
Bowel obstruction with perforation and
meconium peritonitis
Small bowel volvulus
Other intestinal obstructions
Prune-belly syndrome
Tumors
Neuroblastoma
Choriocarcinoma
Sacrococcygeal teratoma
Hemangioma or other hepatic tumors
Congenital leukemia
Cardiac tumors
Renal tumors
Maternal or placental
conditions
Maternal diabetes
Maternal therapy with indomethacin
Multiple gestation with parasitic fetus
Chorioangioma of placenta, chorionic
vessels, or umbilical vessels
Toxemia
Systemic lupus erythematosus
Miscellaneous
Neu-Laxova syndrome
Myotonic dystrophy
Cardiac arrhythmias
Vascular malformations
Vascular accidents
Thrombosis of umbilical vein or inferior vena cava
Recipient in twin-twin transfusion
Infections
Cytomegalovirus, congenital hepatitis,
human parvovirus, enterovirus, other
viruses
Toxoplasmosis, Chagas disease
Coxsackie virus
Syphilis
Leptospirosis
Lymphatic abnormalities
Lymphangiectasia
Cystic hygroma
Noonan syndrome
Multiple pterygium syndrome
Congenital chylothorax
Idiopathic
determined by the balance among (1) capillary hydrostatic
pressure, (2) serum colloid oncotic pressure, (3) interstitial hydrostatic pressure or tissue turgor pressure, and (4)
interstitial osmotic pressure, which depends on lymphatic
flow. The Starling equation defines the relationship among
these forces and their net effect on net fluid movement, or
filtration, across a semipermeable membrane (such as the
capillary membrane) as:
Filtration = K[(Pc − Pt ) − R(Op − Ot )]
where K = capillary filtration coefficient, representing the extent of permeability of a membrane to water
and thus describing capillary integrity; Pc = capillary
hydrostatic pressure; Pt = interstitial hydrostatic pressure
or tissue turgor pressure; R = reflection coefficient for a
solute, representing the extent of permeability of the capillary wall to that solute; Op = plasma oncotic pressure
as determined by plasma proteins and other solutes; and
Ot = interstitial osmotic pressure (Figure 8-1).
Although an abnormality of any of the components of
this equation may, in theory, result in the accumulation
of edema fluid, the fetal-placental unit presents a unique
physiologic condition that effectively eliminates two of
the factors, assuming unimpeded fetal-placental flow and
an appropriately functioning maternal-placental interface. Because approximately 40% of fetal cardiac output is
allocated to the placenta, there is rapid transport of water
CHAPTER 8 Nonimmune Hydrops
Ot
Pt
Interstitium
Pc
Op
Capillary
FIGURE 8-1 Starling forces and net effect on fluid homeostasis.
Arrows represent net effect of movement of fluid across the capillary membrane for each factor under normal conditions. Pc, Capillary
hydrostatic pressure; Pt, interstitial hydrostatic pressure or tissue turgor
pressure; Op, plasma oncotic pressure as determined by plasma proteins
and other solutes; Ot, interstitial osmotic pressure.
between the fetus and mother. Any condition resulting in
elevated fetal capillary hydrostatic pressure or low plasma
colloid oncotic pressure would likely cause the net flow of
water from fetal villi in the placenta to the maternal blood
stream, where it can be effectively eliminated. This elimination of fluid would counteract the accumulation of interstitial fluid by the fetus. Although the placenta of a fetus
with hydrops is also edematous, these changes are believed
to occur with, and not before, fetal fluid accumulation.
DERANGEMENTS IN FLUID HOMEOSTASIS
Diamond et al (1932) suggested three possible mechanisms
that might be relevant in infants with hydrops: anemia,
low colloid osmotic pressure with hypoproteinemia, and
congestive heart failure with hypervolemia. Others have
reviewed these potential mechanisms (Phibbs et al, 1974),
which remain among the central hypotheses addressed by
investigators in this area. The causes of hydrops appear
to be multifactorial, with mechanisms that produce elevated central venous pressure (CVP), capillary leakage,
and impaired lymphatic drainage all contributing to its
development.
Infants with alloimmune hydrops (and several of the
nonimmune hydrops conditions as well) have significant
anemia. It has been proposed that anemia leads to congestive heart failure with increased hydrostatic pressure in the
capillaries, causing vascular damage that results in edema.
However, the hematocrit values of infants with and without
hydrops overlap significantly, suggesting that anemia alone
is not the complete explanation. A rapidly lowered hemoglobin concentration results in greater cardiac output to
maintain adequate oxygen delivery. This output results in
higher oxygen demands by the myocardium, which may be
difficult to meet because of the anemia. The hypoxic myocardium can become less contractile and less compliant,
with ventricular stiffness causing increased afterload to the
atria. High-output congestive heart failure may then exist,
resulting in elevated CVP. Raised CVP leads to increased
capillary filtration pressures and impairment of lymphatic
return (Weiner, 1993). In addition, reduced compliance of
a right ventricle may result in flow reversal in the inferior vena cava, which may in turn cause end-organ damage to the liver, with consequent hypoalbuminemia and
portal hypertension enhancing formation of both edema
and ascites. Hydrops has been produced in fetal lambs
69
(Blair et al, 1994) in which the hemoglobin content was
lowered in 12 fetuses through exchange transfusion using
cell-free plasma; six became hydropic. Anemia developed
more rapidly with a higher CVP in fetuses with hydrops
than in the fetuses without hydrops. In the most severely
anemic fetuses, it is probable that decreased oxygen transport causes tissue hypoxia, which in turn increases capillary
permeability to both water and protein. These changes in
capillary permeability also likely contribute to the development of hydrops.
Infants who have erythroblastosis and hydrops seem to
demonstrate a correlation between serum albumin concentration and the severity of hydrops (Phibbs et al, 1974).
Initial therapy after birth, however, tends to rapidly raise
the serum albumin value toward normal, and with diuresis
the albumin concentrations normalize. This finding suggests that hypoalbuminemia may be the result of dilution
rather than the cause of hydrops.
To elucidate the role of isolated hypoproteinemia in
the genesis of hydrops, Moise et al (1991) have induced
hypoproteinemia in sets of twin fetal lambs. One twin
from each set underwent serum protein reduction through
repeated removal of plasma and replacement with normal
saline; the other twin served as the control. Over 3 days,
plasma protein concentrations were reduced by an average
of 41%, with a 44% reduction in colloid osmotic pressure,
in experimental subjects. No fetuses became edematous,
and total body water content values were similar in experimental and control animals. Thus hypoproteinemia alone
was insufficient to cause hydrops fetalis over the course
of the study. Transcapillary filtration probably increased
with hypoproteinemia, but was compensated by lymphatic
return. Human fetuses with hypoproteinemia as a result of
nephrotic syndrome or analbuminemia rarely experience
hydrops, further supporting the hypothesis that hypoproteinemia alone is not sufficient to cause hydrops. Hypoproteinemia may, however, lower the threshold for edema
formation in the presence of impaired lymphatic return or
increased intravascular hydrostatic pressures.
The most commonly diagnosed causes of nonimmune
hydrops that appears in fetuses older than 24 weeks’ gestation are cardiac disorders. Any state in which cardiac output
is lower than the rate of venous return results in an elevated
CVP. Increased CVP raises capillary filtration pressures
and, if high enough, restricts lymphatic return. Both of
these mechanisms may then contribute to interstitial accumulation of fluid. Structural cardiac causes of elevated CVP
include right-sided obstructive lesions and valvular regurgitation. The most common and easily reversible cause of
nonimmune hydrops is supraventricular tachycardia (SVT).
In general, cardiac output rises with heart rate. At the
increasingly high rates seen in SVT, however, cardiac output plateaus and then diminishes. The heart rates observed
with SVT are often associated with decreased cardiac output. Impaired cardiac output results in elevated CVP, which
can give rise to edema through mechanisms discussed previously (Gest et al, 1990). Myocardial hypoxia (most often
caused by severe anemia) and myocarditis (usually infectious) reduce both the contractility and compliance of the
myocardium and can also cause an increase in CVP.
A fourth factor that contributes to hydrops is decreased
lymph flow. If the rate of fluid filtration from plasma
70
PART II Fetal Development
to tissues exceeds the rate of lymph return to the central venous system, then edema and effusions may form.
A structural impediment or increased CVP that opposes
lymphatic return to the heart can impair lymph flow. To
determine the effects of alterations in CVP on lymphatic
return, Gest et al (1992) applied an opposing hydrostatic
pressure to the thoracic duct in fetal lambs; they inserted
catheters into the thoracic ducts of 10 fetuses. Varying the
height of the catheter altered the thoracic duct outflow
pressure. Thoracic duct flow was nearly constant over the
physiologic range of CVP, but sharply decreased at elevated pressures; therefore lymphatic flow may be reduced
or essentially blocked in pathologic states associated with
elevated CVP.
TABLE 8-2 Antenatal Investigation of Fetal Hydrops
Area
Testing
Maternal
History, including:
Age, parity, gestation
Medical and family histories
Recent illnesses or exposures
Medications
Complete blood count and indices
Blood typing and indirect Coombs antibody screening
Hemoglobin electrophoresis
Kleihauer-Betke stain of peripheral blood
Syphilis, TORCH, and parvovirus B19
titers
Anti-Ro and anti-La, systemic lupus
erythematosus preparation
Oral glucose tolerance test
Glucose-6-phosphate dehydrogenase,
pyruvate kinase deficiency screening
Fetal
Serial ultrasound evaluations
Middle cerebral artery peak systolic
velocity
Limb length, fetal movement
Echocardiography
Amniocentesis
Karyotype
Alpha-fetoprotein
Viral cultures; polymerase chain reaction
analysis for toxoplasmosis, parvovirus 19
Establishment of culture for appropriate
metabolic or DNA testing
Lecithin-to-sphingomyelin ratio to assess
lung maturity
Fetal blood sampling
Genetic testing
Complete blood count
Hemoglobin analysis
Immunoglobulin M test; specific cultures
Albumin and total protein measurements
Measurement of umbilical venous pressure
Metabolic testing
PRENATAL DIAGNOSIS
The initial presentation of fetal hydrops varies by report.
Watson and Campbell (1986) found that two thirds of prenatally diagnosed cases were discovered on routine ultrasonographic examinations, and one third was referred for
evaluation because of suspected polyhydramnios. Graves
and Baskett (1984) reported that hydrops was more commonly discovered after referral for polyhydramnios, fetus
large for dates, fetal tachycardia, or pregnancy-induced
hypertension. Despite the underlying cause of hydrops or
the clinical presentation, the prenatal diagnosis is made via
the ultrasonographic finding of excess fluid in the form of
ascites, pleural or pericardial effusions, skin edema, placental edema, or polyhydramnios. Several definitions for
ultrasonographic diagnosis based on quantity and distribution of excess fluid have been proposed. One widely
accepted set of criteria consists of the presence of excess
fluid in any two of the previously listed compartments.
Because this definition is based on the presence of excess
fluid alone, the degree of severity is generally subjective.
Swain et al (1999) outlined a multidisciplinary approach
to the evaluation and management of the mother and fetus
with hydrops. Table 8-2 provides recommendations for the
investigation of fetal hydrops. Patient history should focus
on ethnic background, familial history of consanguinity,
genetic or congenital anomalies, and complications of
pregnancy, including recent maternal illness and environmental exposures. Maternal disorders such as diabetes, systemic lupus erythematosus, myotonic dystrophy, and any
type of liver disease should also be noted. Initial laboratory
investigation includes blood typing and a Coombs’ test to
rule out immune-mediated hydrops. Other blood tests are
a screening for hemoglobinopathies, a Kleihauer-Betke
test to eliminate fetal-maternal hemorrhage, and testing
for the TORCH diseases (i.e., toxoplasmosis, other infections, rubella, cytomegalovirus infection, and herpes simplex), including syphilis and parvovirus B19.
Further evaluation is directed at identifying possible
causes (Forouzan, 1997). Rapid evaluation is necessary
to determine whether fetal intervention is possible and
to estimate the prognosis for the fetus. Many conditions,
such as arrhythmias, twin-twin transfusion, large vascular
masses, and congenital diaphragmatic hernias and other
chest-occupying lesions, are discovered during the initial
ultrasonographic evaluation (Coleman et al, 2002). Middle cerebral artery peak systolic velocity measurement can
Adapted from Swain S, Cameron AD, McNay MB, et al: Prenatal diagnosis and management of nonimmune hydrops fetalis, Aust N Z J Obstet Gynaecol 39:285-290, 1999.
TORCH, Toxoplasmosis, other infections, rubella, cytomegalovirus, and herpes simplex.
aid in detecting the presence of fetal anemia (HernandezAndrale et al, 2004). If the initial ultrasonic examination is
not helpful in identifying a cause, it may be helpful to repeat
it at a later date to reassess fetal anatomy, monitor progression of the hydrops, and evaluate well-being of the fetus.
Fetal echocardiography should also be performed to
evaluate for cardiac malformations and arrhythmia. Amniotic fluid can be obtained for fetal DNA analysis, cultures,
and lecithin-to-sphingomyelin ratio to assess lung maturity. Fetal blood sampling allows for other tests, such as
a complete blood cell count, routine chemical analyses,
DNA analysis, bacterial and viral cultures, metabolic studies, and serum immunoglobulin measurements.
PRENATAL MANAGEMENT
The goals of antenatal evaluation of fetal hydrops depend
on the underlying cause. In diagnoses in which therapy
is futile, the goal is to avoid unnecessary invasive testing
and cesarean section. The prognosis should be discussed
frankly with the parents, who should be given the option
of terminating the pregnancy. If the underlying cause is
CHAPTER 8 Nonimmune Hydrops
amenable to fetal therapy, the risks and benefits of such
therapy, as well as the warning that diagnostic error is possible, should be discussed with the family.
SVT is one of the most common known causes of nonimmune hydrops, and it is the most amenable to treatment
(Huhta, 2004; Newburger and Keane, 1979). Usually the
mother is given antiarrhythmic agents, and the fetus is monitored closely for resolution of the SVT. Digoxin is most commonly administered, although other antiarrhythmics have
been used, such as sotolol or flecainide, because transplacental
transfer of digoxin may be impaired in the setting of hydrops.
In extreme circumstances, such as fetal tachyarrhythmia
refractory to maternal treatment, direct fetal administration
of antiarrhythmic agents via percutaneous umbilical blood
sampling or intramuscular injection, although untested and
highly risky, has met with some success.
If anemia is the cause of hydrops, transfusions of packed
red blood cells may be administered to the fetus. Often
a single transfusion reverses the edema, although serial
transfusions may be necessary. Parvovirus B19 (Anand et
al, 1987) and fetal-maternal hemorrhage are examples of
diagnoses that are amenable to this therapy. Other diagnoses involving anemias that are refractory to transfusions,
such as α-thalassemia, may require neonatal stem cell transplantation. Transfusions should be given, with the use of
ultrasonographic guidance, into the intraperitoneal space
or umbilical vein. Blood instilled into the abdominal cavity is taken up by lymphatics, but elevated CVPs present in
hydropic fetuses may impair this uptake. If uptake of intraperitoneal blood is incomplete, treatment for the hydrops is
less successful; degeneration of the remaining hemoglobin
may create a substantial bilirubin load, necessitating phototherapy or exchange transfusion after the infant is delivered.
Surgery continues to evolve as a promising therapy
for select cases of fetal hydrops (Azizkhan and Crombleholme, 2008; Kitano et al, 1999). Fetal lung lesions such
as congenital cystic adenomatoid malformation (CCAM)
and pulmonary sequestration can, in the most extreme
cases, result in mediastinal shift, pulmonary hypoplasia,
cardiovascular compromise, and hydrops. A recent review
of 36 fetuses with CCAM found higher rates of hydrops
in infants whose mass-to-thorax ratio was greater than
0.56, in whom the lesion had a cystic predominance, or
in whom the hemidiaphram was everted (Vu et al, 2007).
Adzick et al (1998) reported on the outcome of 175 cases
of fetal lung masses, including 134 cases of CCAM and
41 of extralobar pulmonary sequestration. The 76 fetuses
with CCAM lesions without associated hydrops were all
managed expectantly (maternal transport to a high-risk
center, planned delivery near term, and resection in the
newborn period). CCAMs frequently involute and may
disappear before delivery; therefore there were no deaths
in the nonhydropic group of fetuses in this study. Twentyfive fetuses with hydrops were managed expectantly, and
all 25 died before or after preterm labor at 25 to 26 weeks’
gestation. These results highlight the fact that fetuses with
lung lesions leading to hydrops have high mortality rates.
Thirteen fetuses with CCAM and associated hydrops
underwent open fetal resection or lobectomy. Eight survived and were reported as healthy at 1 to 7 years of follow-up. Maternal morbidities related to fetal intervention
ranged from uterine wound infection with dehiscence to
71
mild postoperative interstitial pulmonary edema, which
was treated with diuretics.
High morbidity and mortality rates in severe twintwin transfusion with associated hydrops led to multiple
international trials of laser photocoagulation of interfetal
vascular connections. Although the trials met with varying results, metanalysis involving the three major trials
demonstrates improvement in perinatal and neonatal outcomes. However, the current level of evidence is limited
in the reported effect on neurodevelopmental outcomes
in survivors. A 2008 Cochrane review recommends considering treatment with laser coagulation at all stages of
twin-twin transfusion (Roberts et al, 2008).
Fetal intervention has met with some success in other
diagnoses with associated hydrops. Thoracoamniotic shunts
for large unicystic lesions and pleuroamniotic shunts for
hydrothorax have reportedly enhanced survival in extreme
cases. Similarly, in cases of massive urinary ascites, urinary
diversion via peritoneal shunts has been reported, but with
a poor long-term prognosis (Crombleholme et al, 1990).
However, as with other invasive interventions, there are
potential risks with fetal surgery. A review of the reproductive outcomes of future pregnancies after a pregnancy
complicated by maternal-fetal surgery found a complication rate of 35%: 12% affected by uterine dehiscience,
6% with uterine rupture, 3% requiring hysterectomy; and
9% with hemorrhage requiring transfusion (Wilson et
al, 2004). These longer-term complications suggest that
the potential benefit of fetal surgical intervention must be
balanced by the potential complications of the procedure
experienced by the mother.
In cases in which the cause can be corrected by appropriate care at the time of delivery, such as elimination of
a chorioangioma, and in cases in which no cause can be
ascertained, close observation for fetal demise is the focus
of prenatal management. Many cases of nonimmune
hydrops manifest in the third trimester as preterm labor. It
is difficult to decide whether to attempt tocolysis and delay
delivery so as to allow the potentially beneficial administration of steroids before birth or to deliver the fetus
immediately. If tocolysis is possible, expectant management should include usual biophysical testing, although
fetal decompensation may be difficult to measure. Abnormal fetal heart tracings, oligohydramnios, decreased fetal
movement, and poor fetal tone are all ominous signs. There
is no indication to prolong pregnancy beyond attainment
of a mature lung profile unless available evidence indicates
improvement or resolution of the hydrops.
NEONATAL EVALUATION
Table 8-3 summarizes the diagnostic evaluations recommended for newborn infants with nonimmune hydrops of
unknown cause.
INTENSIVE CARE OF THE INFANT
WITH HYDROPS FETALIS
After successful resuscitation, including intubation,
administration of surfactant, and placement of umbilical
catheters, the clinical management can address both the
cause and the complications of hydrops. Morbidity and
72
PART II Fetal Development
TABLE 8-3 Diagnostic Evaluation of Newborns With
Nonimmune Hydrops
System
Type of Evaluation
Cardiovascular
Echocardiogram, electrocardiogram
Pulmonary
Chest radiograph, pleural fluid examination
Hematologic
Complete blood cell count, differential
platelet count, blood type and Coombs’
test, blood smear for morphologic analysis
Gastrointestinal
Abdominal radiograph, abdominal ultrasonography, liver function tests, peritoneal
fluid examination, total protein and
albumin levels
Renal
Urinalysis, blood urea nitrogen and
creatinine measurements
Genetic
Chromosomal analysis, skeletal radiographs,
genetic consultation
Congenital
infections
Viral cultures or serologic testing, including
TORCH agents and parvovirus
Pathologic
Complete autopsy, placental examination
Adapted from Carlton DP, McGillivray BC, Schreiber MD: Nonimmune hydrops
fetalis: a multidisciplinary approach, Clin Perinatol 16:839-851, 1989.
TORCH, Toxoplasmosis, other infections, rubella, cytomegalovirus, and herpes simplex.
mortality may result from the hydropic state, the underlying conditions giving rise to hydrops, or both. A fetus
with hydrops that is delivered prematurely is subject to the
additional complications of prematurity. If there is massive ascites or pleural effusions, initial resuscitation may
require thoracentesis or peritoneal tap. Because of pulmonary edema, infants with hydrops are susceptible to pulmonary hemorrhage and require high levels of positive
end-expiratory pressure.
RESPIRATORY MANAGEMENT
Virtually all infants with hydrops require mechanical ventilation because of pleural and peritoneal effusions, pulmonary hypoplasia, surfactant deficiency, pulmonary edema,
poor chest wall compliance caused by edema, or persistent
pulmonary hypertension of the newborn. The presence of
persistent pleural effusions may necessitate the placement
of chest tubes. Ascites may also compress the diaphragm
and impair lung expansion. Breath sounds, chest movement, blood gas levels, and radiographs must all be monitored frequently, so that ventilator support can be reduced
in response to improvements in lung compliance and water
clearance. Pneumothoraces and pulmonary interstitial
emphysema remain potential complications as long as ventilator support is continued. Infants who need a prolonged
course of ventilation, particularly those born prematurely,
may develop bronchopulmonary dysplasia. Chronic lung
disease results in a longer and more complicated hospital
course and contributes to the late mortality of hydrops.
FLUID AND ELECTROLYTE MANAGEMENT
A primary goal of fluid management is resolution of the
hydrops itself. Maintenance fluids should be restricted,
with volume boluses given only in response to clear
signs of inadequate intravascular volume. The hydropic
newborn has an excess of free extracellular water and
sodium. Fluids given during resuscitation further increase
the amount of water and sodium that must be removed
during the immediate neonatal period. Initial maintenance
fluids should contain minimal sodium. Serum and urine
sodium levels, urine volume, and daily weights should be
monitored carefully to guide administration of fluids and
electrolytes. Urinary sodium levels may help differentiate
between hyponatremia caused by hemodilution and urinary losses.
CARDIOVASCULAR MANAGEMENT
Shock may be a prominent feature of patients with hydrops.
Hydropic infants may have hypovolemia as a result of capillary leakage, poor vascular tone, and impaired myocardial
contractility from hypoxia or infection, impaired venous
return caused by shifting or compression of mediastinal
structures, or pericardial effusion. Adequate intravascular volume must be maintained, and correctable causes of
impaired venous return should be addressed. Peripheral
perfusion, heart rate, blood pressure, and acid-base status
should be monitored carefully.
CLINICAL COURSE AND OUTCOME
Despite improvements in diagnosis and management,
mortality from nonimmune hydrops remains high.
Reported survival rates for all fetuses diagnosed antenatally with hydrops range from 12% to 24% (Heinonen
et al, 2000; McCoy et al, 1995; Negishi et al, 1997). Higher
survival rates have been reported in infants born alive, but
the highest rates are still only 40% to 50% (Wy et al,
1999). Improved ultrasonography techniques and earlier
testing may actually lead to lower survival rates as hydrops
is diagnosed in more first-trimester infants. These infants
are more likely to have chromosomal abnormalities that
are incompatible with survival, but were previously not
included in populations of hydropic fetuses. The best predictor of survival is the cause of the hydrops and the gestational age of the child at delivery. Highest survival rates
are seen in infants with parvovirus infection, chylothorax,
or SVT. The lowest survival rates are for hydrops from
chromosomal cause, although the figures may be biased
because a significant number of the pregnancies in such
cases are terminated (Heinonen et al, 2000; Sohan et al,
2001). A recent review of 598 patients with nonimmune
hydrops found other risk factors for increased mortality
including younger gestational age, lower 5-minute Apgar
score, and the need for increased respiratory support
(Abrams et al, 2007). A smaller study from Taiwan also
found that lower albumin levels were associated with a
higher mortality rate (Huang et al, 2007).
Interventions to improve outcomes in hydrops are
limited by the rarity of the disease. Carlton et al (1989)
reported on a group of 36 infants with nonimmune
hydrops and noted that 90% of the infants who died within
24 hours had pleural effusions, compared with only 50%
of those who survived. More than one third of the infants
in this study required thoracentesis in the delivery room to
aid in lung expansion. All the infants who lived more than
24 hours were treated with mechanical ventilation and
CHAPTER 8 Nonimmune Hydrops
received supplemental oxygen; they needed ventilation for
an average of 11 days (range, 2 to 48 days). Most hydropic
infants lose a minimum of 15% of their birthweight, and
some lose as much as 30%. Ordinarily, diuresis begins
on the second or third day after birth and continues for
a period of 2 to 4 days. Once the edema has resolved, the
infants have normal levels of circulating protein and eventually recover from their apparent capillary leak syndrome.
No specific management strategies during the neonatal
period, such as the use of high-frequency oscillatory ventilation, have been shown to improve outcome, although
the published studies are powered to detect small survival
differences (Wy et al, 1999).
For infants who survive the immediate neonatal period,
long-term outcomes appear to be excellent. Nonimmune
hydrops by itself does not seem to lead to residual developmental delay. A small study from Japan found that 13
of 19 surviving infants with nonimmune hydrops had normal development at 1 to 8 years (Nakayama et al, 1999).
The six infants with mild or severe delays in this study
had other morbidities, such as extreme prematurity, structural cardiac lesions, or chromosomal anomalies. Thus
73
long-term morbidities from nonimmune hydrops appear
to result from the underlying cause of the hydrops, gestational age at delivery, and complications arising immediately after delivery.
SUGGESTED READINGS
Abrams ME, Meredith KS, Kinnard P, et al: Hydrops fetalis: a retrospective review
of cases reported to a large national database and identification of risk factors
associated with death, Pediatrics 120:84-89, 2007.
Anand A, Gray ES, Brown T, et al: Human parvovirus infection in pregnancy and
hydrops fetalis, N Engl J Med 316:183-186, 1987.
Azizkhan RG, Crombleholme TM: Congenital cystic lung disease: contemporary
antenatal and postnatal management, Pediatr Surg Int 24:643-657, 2008.
Huhta JC: Guidelines for the evaluation of heart failure in the fetus with or without
hydrops, Pediatr Cardiol 25:274-286, 2004.
Bellini C, Hennekam RCM, Fulcheri E, et al: Etiology of nonimmune hydrops
fetalis: a systematic review, Am J Med Genet 149A:844-851, 2009.
Machin GA: Hydrops revisited: literature review of 1414 cases published in the
1980s, Am J Med Genet 34:366-390, 1989.
Swain S, Cameron AD, McNay MB, et al: Prenatal diagnosis and management
of nonimmune hydrops fetalis, Aust N Z J Obstet Gynaecol 39:285-290, 1999.
Complete references used in this text can be found online at www.expertconsult.com
P A R T
III
Maternal Health Affecting Neonatal Outcome
C H A P T E R
9
Endocrine Disorders in Pregnancy
Gladys A. Ramos and Thomas R. Moore
DIABETES IN PREGNANCY
Currently, 17 million people in the United States have a
form of diagnosed diabetes. Alarmingly, the data for 2003
to 2006 indicate that approximately 10.2% (11.5 million)
of women older than 20 years have diabetes. Data indicate that new cases of type 2 diabetes mellitus are occurring at an increasing rate among American Indian, African
American, Hispanic, and Latino children and adolescents
(http://diabetes.niddk.nih.gov/dm). Continued immigration among populations with high rates of type 2 diabetes mellitus and the effects of changes in diet (increases in
number of calories and fat content) and lifestyle (sedentary)
portend marked rises in the percentage of patients with preexisting diabetes who will become pregnant in the future.
There is also an epidemic of childhood obesity currently
under way in the United States, with approximately 23 million (30%) children and youth who are overweight. This
trend will have a profound effect on obstetrics and pediatric practice in the future. Expanded efforts to reach the
populations at risk are necessary if a significant increase in
maternal and neonatal morbidity is to be avoided (Persson
and Hanson, 1998).
Depending on the population surveyed, abnormalities of glucose regulation occur in 3% to 8% of pregnant
women. Although more than 80% of this glucose intolerance arises only during pregnancy (gestational diabetes)
and involves relatively modest episodes of hyperglycemia,
the attendant fetal and newborn morbidity is disproportionate. Compared with weight-matched controls, infants
of diabetic mothers (IDMs) have double the risk of serious birth injury, triple the likelihood of cesarean section,
and quadruple the incidence of admission to a newborn
intensive care unit. Studies indicate that the magnitude of
risk of these maloccurrences is proportional to the level of
maternal hyperglycemia. Therefore, to some extent, the
excessive fetal and neonatal morbidity of diabetes in pregnancy is preventable or at least reducible through meticulous prenatal and intrapartum care.
MATERNAL-FETAL METABOLISM IN NORMAL
PREGNANCY AND DIABETIC PREGNANCY
Normal Maternal Glucose Regulation
With each meal, a complex combination of maternal hormonal actions, including the secretion of pancreatic insulin, glucagon, somatomedins, and adrenal catecholamines,
ensures an ample but not excessive supply of glucose to
the mother and fetus during pregnancy. The key effects
of pregnancy on maternal metabolic regulation are as
follows:
ll
ll
ll
ll
Because the fetus continues to draw glucose from the
maternal bloodstream across the placenta, even during
periods of fasting, the tendency toward maternal hypoglycemia between meals becomes increasingly marked
as pregnancy progresses and fetal glucose demand
grows.
Placental steroid and peptide hormone production
(estrogens, progesterone, and chorionic somatomammotropin) rises linearly throughout the second and
third trimesters, resulting in a progressively increasing
tissue resistance to maternal insulin action.
Progressive maternal insulin resistance requires a significant augmentation in pancreatic insulin production
(more than twofold nonpregnant levels) during feeding
to maintain euglycemia. Twenty-four–hour mean insulin levels are 30% higher in the third trimester than in
the nonpregnant state.
If pancreatic insulin output is not adequately augmented, maternal hyperglycemia and then fetal hyperglycemia result. The severity of hyperglycemia and its
timing depend on the relative inadequacy of insulin
production.
Fetal Effects of Maternal Hyperglycemia
Congenital Anomalies
A major threat to IDMs is the possibility of a life-threatening
structural anomaly. In the normoglycemic pregnancy, the
risk of a major birth defect is 1% to 2%. Among women
with pregestational diabetes, the risk of a fetal structural
anomaly is fourfold to eightfold higher. In a recent cohort
study of 2359 pregnancies in women with pregestational
diabetes, the rate of anomalies was more than doubled.
Major congenital anomalies occurred in 4.6% overall
with 4.8% for type 1 diabetes mellitus and 4.3% for type
2 diabetes mellitus. This is a significant increase over the
expected rate of birth defects in the general population
(approximately 1.5%). Neural tube defects in IDM were
increased 4.2-fold (Figure 9-1), and congenital heart disease by 3.4-fold. Prenatal diagnosis of these anomalies was
accomplished in 65% of neonates (Macintosh et al, 2006).
The typical defects and their frequency of occurrence,
75
76
PART III
Maternal Health Affecting Neonatal Outcome
experience with 105 diabetic patients, finding an overall
malformation rate of 13.3%. A recent study conducted in
the United Kingdom from 1991 to 2000 in patients with
type 1 diabetes mellitus found similar results (Temple
et al, 2002). Adverse outcome was significantly higher in
the poor control group (HbA1c ≥7.5) than in the fair control group (HbA1c <7.5), with a ninefold increase in the
congenital malformation rate (relative risk, 9.2; 1.1 to 79.9)
(Temple et al, 2002). For a woman with an HbA1c value of
less than 7.1%, the risk of delivering a malformed infant was
equivalent or slightly less than that for the normoglycemic
population. However, the anomaly rate rose progressively
with increasing HbA1c, 14% with an HbA1c value of 7.2%
to 9.1%, 23% with an HbA1c value of 9.2% to 11.1%, and
25% with an HbA1c value of greater than 11.2%.
Pathogenesis
FIGURE 9-1 Newborn with caudal regression syndrome, macrosomia, and respiratory distress. The mother had type 1 diabetes and
a glycosylated hemoglobin concentration of 13.5% when first seen for
prenatal care at 12 weeks’ gestation. ( From Creasy RK, Resnik R, editors:
Maternal-fetal medicine: principles and practice, ed 2, Philadelphia, 1989,
WB Saunders.)
TABLE 9-1 Congenital Malformations in Infants of Mothers
With Insulin-Dependent Diabetes
Appropriate
Risk Ratio
Risk (%)
All cardiac defects
18×
8.5
All central nervous system
anomalies
16×
5.3
Anomaly
Anencephaly
13×
–
Spina bifida
20×
–
All congenital anomalies
8×
18.4
Becerra JE, Khoury MJ, Cordero JF, et al: Diabetes mellitus during pregnancy and
the risks for specific birth defects: a population based case-control study, Pediatrics
85:1, 1990.
noted in a prospective study of infants with major malformations, are listed in Table 9-1. The majority of lesions
involve the central nervous and cardiovascular systems,
although other series have reported an excess of genitourinary and limb defects (Cousins, 1991).
There is no increase in birth defects among offspring of
diabetic fathers, nondiabetic women, or women in whom
gestational diabetes develops after the first trimester. These
findings suggest that glycemic control during embryogenesis is a critical factor in the genesis of diabetes-associated
birth defects. In a study by Miller et al (1981), the frequency
of congenital anomalies was proportional to the maternal glycohemoglobin (HbA1c) value in the first trimester
(rate of anomalies 3.4% with HbA1c <8.5%, and 22.4%
with HbA1c >8.5%). Lucas et al (1989) reported a similar
The specific mechanisms by which hyperglycemia disturbs
embryonic development are incompletely elucidated, but
reduced levels of arachidonic acid and myo-inositol and
accumulation of sorbitol and trace metals in the embryo
have been demonstrated in animal models (Pinter et al,
1986). Fetal hyperglycemia may promote excessive formation of oxygen radicals in the mitochondria of susceptible
tissues, leading to the formation of hydroperoxides, which
inhibit prostacyclin. The resulting overabundance of
thromboxanes and other prostaglandins may then disrupt
vascularization of developing tissues. In support of this
theory, the addition of prostaglandin inhibitors to mouse
embryos in culture medium prevents glucose-induced
embryopathy. Furthermore, the addition of dietary antioxidants in the form of high doses of vitamins C and E
decreased fetal dysmorphogenesis to nondiabetic levels in
rat pregnancy and rat embryo culture (Cederberg and
Eriksson, 2005; El-Bassiouni et al, 2005).
Prevention
Because the critical period for teratogenesis is the first
3 to 6 weeks after conception, normal glycemic control
must be instituted before pregnancy to prevent these birth
defects. Several clinical trials of meticulous preconception
glycemic control in women with diabetes have resulted
in malformation rates equivalent to those in the general
population (Fuhrmann et al, 1983). A recent metaanalysis
of these trials demonstrated that the pooled risk of malformations was lower in women with preconception care
compared with those without preconception counseling
(Ray et al, 2001). The threshold level of glycemic control,
as evidenced by the HbA1c value, necessary to normalize a
patient’s risk of congenital anomalies appears to be a nearnormal value. Thus any elevation of the HbA1c above normal increases the risk of teratogenesis proportionately.
Macrosomia
Fetal overgrowth is a major problem in pregnancies complicated by diabetes, leading to unnecessary cesarean sections and potentially avoidable birth injuries. A 1992 study
of birthweights in the previous 20 years indicated that 21%
of infants with birthweights of 4540 g or greater were born
to mothers who were glucose intolerant, a rate clearly disproportionate to the only 2% to 5% of gravidas with some
form of diabetes (Shelley-Jones et al, 1992). Thus the
CHAPTER 9 Endocrine Disorders in Pregnancy
problem of abnormal fetal growth in diabetic pregnancy
remains an important clinical challenge.
Macrosomia is defined variously as birthweight above the
90th percentile for gestational age or birthweight greater
than 4000 g; it occurs in 15% to 45% of diabetic pregnancies. Excessive fetal size contributes to a greater frequency
of intrapartum injury (shoulder dystocia, brachial plexus
palsy, and asphyxia). Macrosomia is also a major factor in
the higher rate of cesarean delivery among diabetic women.
Because the risk of macrosomia is fairly constant for all
classes of diabetes, it is likely that first-trimester metabolic
control has less of an effect on fetal growth than does glycemic regulation in the second and third trimesters.
Growth Dynamics
IDMs with macrosomia follow a unique pattern of in utero
growth compared with fetuses in euglycemic pregnancies. During the first and second trimesters, differences
in size between fetuses born to diabetic and nondiabetic
mothers are usually undetectable with ultrasound measurements. After 24 weeks, however, the growth velocity of the IDM fetus’ abdominal circumference typically
begins to rise above normal (Ogata et al, 1980). Reece et al
(1990) demonstrated that the IDM fetus has normal head
growth, despite marked degrees of hyperglycemia. Landon
et al (1989) have reported that although head growth and
femur growth of IDM fetuses were similar to those of normal fetuses, abdominal circumference growth significantly
exceeded that of controls beginning at 32 weeks’ gestation
(abdominal circumference growth in IDM fetuses is 1.36
cm/week, versus 0.901 cm/week in normal subjects).
Morphometric studies of the IDM newborn indicate
that the greater growth of the abdominal circumference is
caused by deposits of fat in the abdominal and interscapular
areas. This central depositing of fat is a key characteristic
of diabetic macrosomia and underlies the pathology associated with vaginal delivery in these pregnancies. Acker et al
(1986) showed that although the incidence of shoulder dystocia is 3% among infants weighing more than 4000 g, the
incidence in infants from diabetic pregnancies who weigh
more than 4000 g is 16%. Finally, despite our emphasis on
birthweight, this alone may not be a sensitive measure of
fetal growth. Catalano et al (2003) conducted body composition studies on infants born to mothers with diabetes
and found that even when appropriate for gestational age,
these infants have increased fat mass and percent body fat
compared with a normoglycemic control group.
Childhood Effects
Higher growth velocity, begun in fetal life during a pregnancy complicated by diabetes, may extend into childhood
and adult life. Silverman et al (1995) reported follow-up
of IDMs through age 8 years in which half the infants
weighed more than the 90th percentile for gestational age
at birth. By age 8 years, approximately half of the IDMs
weighed more than the heaviest 10% of the nondiabetic
children. The asymmetry index was 30% higher in diabetic
offspring than in the controls by age 8 years. These investigators also showed that offspring with diabetes have permanent derangement of glucose-insulin kinetics, resulting
in a higher incidence of impaired glucose tolerance. In
addition, Dabelea et al (2000) also reported that the mean
77
adolescent body mass index was 2.6 kg/m2 greater in sibling offspring of diabetic pregnancies compared with the
index siblings born when the mother previously had normal glucose tolerance.
Pathophysiology
The pathophysiology of excessive fetal growth is complex
and reflects the delivery of an abnormal nutrient mixture
to the fetoplacental unit, regulated by an abnormal confluence of growth factors. Pedersen (1952) hypothesized that
maternal hyperglycemia stimulates fetal hyperinsulinemia,
which in turn mediates acceleration of fuel utilization and
growth. The features of the abnormal growth in diabetic
pregnancy include excessive adipose deposition, visceral
organ hypertrophy, and acceleration of body mass accretion (Ogata et al, 1980).
Data from the Diabetes in Early Pregnancy project suggest that maternal metabolic control is a critical factor leading to fetal macrosomia (Jovanovic-Peterson et al, 1991). In
this study, in which meticulous glycemic care was maintained
in early pregnancy and beyond, fetal weight did not correlate
significantly with fasting glucose levels. During the second
and third trimesters, however, postprandial blood glucose
levels were strongly predictive of both birthweight and the
overall percentage of macrosomic infants. With postprandial
glucose values averaging 120 mg/dL, approximately 20% of
infants had macrosomia; a 30% rise in postprandial levels
to 160 mg/dL resulted in a predicted percentage of macrosomia of 35%. In contrast, Persson et al (1996) showed
that fasting glucose concentrations account for 12% of the
variance in birthweight and correlated best with estimates of
neonatal fat. Similarly, Uvena-Celebrezze et al (2002) found
that the strongest correlation was between fasting glucose
and neonatal adiposity rather than postprandial measures.
The Pedersen hypothesis (Pedersen, 1977) presumes
that abnormal fuel milieu in the maternal bloodstream is
reflected contemporaneously in the fetal compartment:
Maternal hyperglycemia = Fetal hyperglycemia. Studies by
Hollingsworth and Cousins (1981) have confirmed much
of Pedersen’s hypothesis and note the following features of
normal pregnancy:
ll
ll
ll
Maternal fasting blood glucose levels decline from
approximately 85 mg/dL to 75 mg/dL. Mean blood
glucose also declines.
At night, maternal glucose levels drop markedly as the
fetus continues to draw glucose stores from the maternal circulation.
Postprandial peaks in maternal blood glucose rarely
exceed 120 mg/dL at 2 hours or 130 mg/dL at 1 hour.
In addition, in diabetic pregnancies:
ll
ll
If maternal glucose levels surge excessively after a
meal, the consequent fetal hyperglycemia is accompanied by fetal pancreatic beta-cell hyperplasia and
hyperinsulinemia.
Fetal hyperinsulinemia, lasting only episodically for
1 to 2 hours, has detrimental consequences for fetal
growth and well-being, in that it (1) promotes storage of excess nutrients, resulting in macrosomia, and
(2) drives catabolism of the oversupply of fuel, using
energy and depleting fetal oxygen stores.
78
PART III
Maternal Health Affecting Neonatal Outcome
8
6
Adjusted odds ratio
7
1 hour
Glucose Fasting
category mg/dL
6
1
2
3
4
5
6
7
5
4
3
75
75–79
80–84
85–89
90–94
95–99
100
1 hour
mg/dL
2 hour
mg/dL
106
106–132
133–155
156–171
172–193
194–211
212
90
91–108
109–125
126–139
140–157
158–177
178
3
4
5
6
7
Glucose category
FIGURE 9-2 Adjusted odds ratios for cord C-peptide levels at >90th
percentile at different glucose categories based on a 2-hour oral glucose
challenge test. (Adapted from HAPO Study Cooperative Research Group,
Metzger BE, Lowe LB, et al: Hyperglycemia and adverse pregnancy outcomes,
N Engl J Med 358:1991-2002, 2008.)
ll
5
1 hour
Glucose Fasting
category mg/dL
1
2
3
4
5
6
7
4
3
75
75–79
80–84
85–89
90–94
95–99
100
2 hour
1 hour
mg/dL
2 hour
mg/dL
106
106–132
133–155
156–171
172–193
194–211
212
90
91–108
109–125
126–139
140–157
158–177
178
2
2
1
Fasting
2 hour
Adjusted odds ratio
Fasting
1
2
Episodic fetal hypoxia stimulated by episodic maternal
hyperglycemia leads to an outpouring of adrenal catecholamines, which in turn causes hypertension, cardiac
remodeling, and cardiac hypertrophy.
The Hyperglycemia and Adverse Pregnancy Outcomes
(HAPO) study provided additional evidence in support of
the Pedersen hypothesis. In this well-designed, multicenter
prospective trial, women underwent a 75-g oral glucose
challenge test between 28 and 32 weeks’ gestation. Providers were blinded to test results if the fasting value was
105 mg/dL or less and the 2-hour plasma glucose level was
200 mg/dL or less. The women did not receive any therapeutic intervention. The study found a continuous association between maternal glucose values, cord C-peptide
levels (Figure 9-2) and birthweight above the 90th percentile (Figure 9-3) (HAPO Study Cooperative Research
Group et al, 2008). A followup analysis of the same cohort
evaluating neonatal anthropometric measurements, found
a link between maternal hyperglycemia, cord C-peptide
levels and neonatal adiposity (HAPO Study Cooperative
Research Group et al, 2009). This study suggested that
maternal hyperglycemia results in neonatal adiposity that
is mediated by fetal insulin production (C-peptide level
>90th percentile) (HAPO Study Cooperative Research
Group et al, 2009).
Prevention of Macrosomia
Because macrosomic fetuses are at an increased risk for
immediate complications related to birth injury and for
potential long-term consequences such as late childhood
obesity and insulin resistance, measures for prevention of
macrosomia have been recommended. As described previously, fetal hyperinsulinemia, which acts as a fetal growth
factor, occurs in response to fetal hyperglycemia, which
in turn reflects the maternal hyperglycemic condition.
Therefore, measures that promote consistent maternal
euglycemia may prevent macrosomia. Several prospective trials have shown that strict maternal glycemic control
using insulin and dietary therapy and fastidious blood glucose monitoring can reduce the incidence of macrosomia
1
1
2
3
4
5
6
7
Glucose category
FIGURE 9-3 Adjusted odds ratios for macrosomia at >90th percentile
at different glucose categories based on a 2-hour oral glucose challenge
test. (Adapted from HAPO Study Cooperative Research Group, Metzger BE,
Lowe LB, et al: Hyperglycemia and adverse pregnancy outcomes, N Engl J
Med 358:1991-2002, 2008.)
(Coustan and Lewis, 1978; Langer et al, 1994; Thompson
et al, 1990). Langer et al (1994) compared the outcomes of
diabetic pregnancies managed conventionally (four blood
glucose measurements per day) or intensely (seven blood
glucose measurements per day). Fasting blood glucose
values were maintained between 60 and 90 mg/dL and
2-hour postprandial values at less than 120 mg/dL. Outcomes were compared to nondiabetic control pregnancies. The rate of infants born weighing more than 4000
g was 14% in the conventionally managed group, 7% in
the intensely managed group, and 8% in nondiabetic controls. Similarly, the rate of shoulder dystocia was 1.4% in
the conventionally managed group, 0.4% in the intensely
managed group, and 0.5% in the control group. Thus, like
the reduction of congenital anomalies in mothers with diabetes by means of first-trimester euglycemia, strict glycemic control in the second and third trimesters may reduce
the fetal macrosomia rate to near baseline.
Fetal Hypoxic Stress
As noted previously, episodic maternal hyperglycemia
promotes a fetal catabolic state in which oxygen depletion
occurs. Several fetal metabolic adaptive responses to this
episodic hypoxia occur. For example, the drop in fetal oxygen tension causes stimulation of erythropoietin, red cell
hyperplasia, and elevation in fetal hematocrit. Polycythemia can lead to poor circulation and postnatal hyperbilirubinemia. Profound episodic hyperglycemia in the third
trimester causing severe fetal hypoxic stress has been theorized as the cause of sudden intrauterine fetal demise in
poorly controlled diabetes.
CLASSIFYING AND DIAGNOSING DIABETES
IN PREGNANCY
The classification system for diabetes in pregnancy recommended by White has been replaced by a scheme based
on the pathophysiology of hyperglycemia and developed
by the National Diabetes Data Group (NDDG) in 1979
(Hare and White, 1980). The two types are summarized
79
CHAPTER 9 Endocrine Disorders in Pregnancy
in Table 9-2. This nomenclature is useful because it categorizes patients according to the underlying pathogenesis
of their diabetes—insulin-deficient (type 1) and insulinresistant (type 2 and gestational). One must remember
that the diagnosis of gestational diabetes applies to any
woman who is found to have hyperglycemia during pregnancy. A certain percentage of such women actually have
type 2 diabetes, but the diagnosis cannot be confirmed
until postpartum testing.
Pregestational Diabetes
Patients with type 1 diabetes mellitus typically exhibit
hyperglycemia, ketosis, and dehydration in childhood or
adolescence. Often the diagnosis is made during a hospital admission for diabetic ketoacidosis and coma. Rarely
is the diagnosis of type 1 diabetes mellitus made during
pregnancy. Conversely, it is not unusual for women with
a tentative diagnosis of gestational diabetes to be found
to have overt, type 2 diabetes mellitus after delivery. The
American Diabetic Association has outlined three criteria
for diagnosing type 2 diabetes mellitus in nonpregnant
subjects. They include the finding of a casual plasma glucose of 200 mg/dL or greater, a fasting plasma glucose of
126 mg/dL or greater, or a 2-hour glucose value of 200
mg/dL or greater on a 75-g, 2-hour glucose tolerance test
(GTT). Diagnostic criteria are listed in Box 9-1.
Gestational Diabetes
Gestational diabetes mellitus (GDM) is defined as glucose
intolerance that begins or is first recognized during pregnancy (American Diabetes Association, 2002). Almost
TABLE 9-2 Classification of Diabetes Mellitus
Type
Old Nomenclature
Clinical Features
Type 1
Juvenile-onset diabetes
Insulin-deficient, ketosisprone; virtually all patients
with type 1 diabetes mellitus are insulin dependent
Type 2
Adult-onset diabetes
Insulin-resistant, not
ketosis-prone; few patients
with type 2 diabetes mellitus
are truly insulin dependent
Gestational
—
Occurs during and resolves
after pregnancy; insulin-
resistant; not ketosis-prone
uniformly, GDM arises from significant maternal insulin
resistance, a state similar to type 2 diabetes mellitus. In
many cases, GDM is simply preclinical type 2 diabetes
mellitus unmasked by the hormonal stress imposed by the
pregnancy. Although GDM complicates no more than 5%
to 6% of pregnancies in the United States, the prevalence
of GDM in specific populations varies from 1% to 14%
(American Diabetes Association, 2002). Clinical recognition of GDM is important because therapy—including medical nutrition therapy, insulin when necessary,
and antepartum fetal surveillance—can reduce the welldescribed perinatal morbidity and mortality associated
with GDM.
Traditionally, universal screening for GDM has been
recommended (Metzger, 1991). However, the Fourth
International Workshop-Conference on Gestational
Diabetes and the American College of Obstetricians and
Gynecologists have now indicated that either a risk-factor approach or universal screening can be considered
(American College of Obstetricians and Gynecologists,
2001; Metzger and Coustan, 1998). This recommendation is based on the findings of Sermer et al (1994), who
reported the results of screening Canadian women at 26
weeks’ gestation with the 100-g, 3-hour GTT. They identified several risk factors as significantly increasing the
likelihood of GDM, among which were maternal age of 35
years or more, body mass index higher than 22 kg/m2, and
Asian or other ethnicity than white. Women with one or
no risk factors had a 0.9% risk of GDM, whereas the risk
for those with two to five factors was 4% to 7%. By limiting screening for GDM to patients with more than one
risk factor, these investigators were able to reduce testing
by 34% while retaining a sensitivity rate of approximately
80%, with a false-positive result rate of 13%. Therefore
in patients meeting all criteria listed in Table 9-3 and Box
9-2, it may be cost effective to avoid screening. Currently
a multicenter trial is underway by National Institute of
Child Health and Human Development Maternal Fetal
Medicine Unit Network in which women with GDM
will be randomized to standard therapy versus no therapy
(Landon et al, 2009). This randomized study will address
whether identification and treatment of GDM decrease
perinatal morbidity.
Notwithstanding these findings, multiple studies from
more heterogeneous U.S. populations have demonstrated
TABLE 9-3 Oral Glucose Tolerance Test for Gestational Diabetes
Venous Plasma Glucose Level*
BOX 9-1 C
riteria for the Diagnosis of Type 2
Diabetes Mellitus
ll
ll
ll
Symptoms of diabetes (polyuria, polydipsia, and unexplained weight loss)
and a casual plasma glucose level >200 mg/dL (11.1 mmol/L). Casual is
defined as any time of day without regard to time of last meal, or
Fasting glucose level >126 mg/dL (7.0 mmol/L). Fasting is defined as no
caloric intake for at least 8 hours, or
Two-hour plasma glucose >200 mg/dL (11.1 mmol/L) during a 75-g oral
glucose challenge test.
Adapted from the American Diabetes Assoication: Clinical practice recommendations:
Standards of medical care for diabetes, 2007, Diabetes Care 30:s4-s41, 2007.
Fasting
value
100-g Glucose Load
75-g Glucose Load
mg/dL
mg/dL
mmol/L
mmol/L
95
5.3
95
5.3
1-hr value
180
10.0
180
10.0
2-hr value
155
8.6
155
8.6
3-hr value
140
7.8
—
—
*Test prerequisites: 1-hr, 50-g glucose challenge result >135 mg/dL; overnight fast of
8 to 14 hours; carbohydrate loading for 3 days, including >150 g carbohydrate; seated
and not smoking during the test; two or more values must be met or exceeded; either a
2-hr (75-g glucose) or 3-hr (100-g glucose) test can be performed.
80
PART III
Maternal Health Affecting Neonatal Outcome
BOX 9-2 C
riteria for Low Risk of Gestational
Diabetes*
ll
ll
ll
ll
ll
ll
Age <25 years
Normal prepregnancy body weight
No first-degree relatives with diabetes
Not a member of an ethnic group at high risk for GDM
No history of GDM in prior pregnancy
No history of adverse pregnancy outcome
Data from Metzger BE, Coustan DR: Summary and Recommendations of the Fourth International
Workshop-Conference on Gestational Diabetes Mellitus. The Organizing Committee, Diabetes
Care 21(Suppl 2):B161-B167, 1998.
GDM, Gestational diabetes mellitus.
*Screening for GDM may be omitted only if all criteria are met.
the inadequacy of risk factor based screening of patients
for GDM. Lavin et al (1981) noted that if only those with
risk factors were screened, the percentage of GDM cases
detected was similar to the detection rate in those without risk factors (1.4%). A later study (Weeks et al, 1995),
which assessed the effect of screening only patients with
risk factors, reported that selective screening would have
failed to detect 43% of cases of GDM. Moreover, 28% of
the women with undiagnosed GDM would have required
insulin and had a several-fold higher risk of cesarean section because of macrosomia.
Universal screening should be performed in women in
ethnic groups at a higher risk for glucose intolerance during pregnancy, namely those of Hispanic, African, Native
American, South or East Asian, Pacific Islands, or Indigenous Australian ancestry. For simplicity of administration,
universal screening, with the possible exception of the lowest risk category, is probably best. The universal screening
method for GDM has been shown to result in earlier diagnosis and improved pregnancy outcomes, including lower
rates of macrosomia and a decrease in neonatal admissions
to neonatal intensive care units (Griffin et al, 2000).
The timing of screening for GDM is important. Because
maternal insulin resistance rises progressively during pregnancy, screening too early can miss some patients who will
become glucose intolerant later. Screening too late in the
third trimester can limit the time during which metabolic
interventions can take place. Thus, risk factors for GDM
should be assessed at the initial prenatal visit. Factors that
should lead to a first-trimester glucose challenge test are
listed in Box 9-3. In the remaining patients, screening
should be performed with the use of 50 g of glucose at 26
to 28 weeks’ gestation.
Various threshold levels for the 50-g glucose challenge are in use, including 140 mg/dL, 135 mg/dL, and
130 mg/dL. The sensitivity of the GDM testing regimen
depends on the threshold value used. The most commonly
used threshold, 140 mg/dL, detects only 80% of patients
with GDM and results in requiring a 3-hour oral GTT in
approximately 10% to 15% of patients. Using a challenge
threshold of 135 mg/dL improves sensitivity to more than
90% but increases the number of 3-hour oral GTTs by
42% (Ray et al, 1996). Thus, the clinician encountering
a newborn with multiple stigmata of an IDM, yet whose
mother had a negative diabetes screening test result, should
realize that this result during pregnancy does not rule out
GDM. This is also why every patient who delivered an
BOX 9-3 Indications for the First-Trimester
50-g Glucose Challenge
ll
ll
ll
ll
ll
ll
ll
Maternal age >25 years
Previous infant >4 kg
Previous unexplained fetal demise
Previous pregnancy with gestational diabetes
Strong immediate family history of type 2 or gestational diabetes mellitus
Maternal obesity (>90 kg)
Fasting glucose level >140 mg/dL (7.8 mmol/L) or random glucose reading
>200 mg/dL (11.1 mmol/L)
infant with macrosomia in a prior pregnancy should be
screened early in all subsequent pregnancies.
Definitive diagnosis of gestational diabetes is made with
a GTT. Either 100 g of glucose and 3 hours of testing, or
75 g of glucose and 2 hours of testing can be used. The
diagnostic criteria are shown in Box 9-1. Two or more values must be met or exceeded for the diagnosis of GDM
to be made. A GTT should be performed after overnight
fasting and with modest carbohydrate loading before the
test.
PERINATAL COMPLICATIONS OF DIABETES
DURING PREGNANCY
Fetal Morbidity and Mortality
Perinatal Mortality
Perinatal mortality in diabetic pregnancy has decreased
30-fold since the discovery of insulin in 1922 and the
advent of intensive obstetric and infant care in the 1970s
(Figure 9-4). Improved techniques for maintaining maternal euglycemia have led to later timing of delivery and
have reduced the incidence of iatrogenic respiratory distress syndrome (RDS).
Nevertheless, the currently reported perinatal mortality
rates among women with diabetes remain approximately
twice those observed in nondiabetic women (Table 9-4).
Congenital malformations, RDS, and extreme prematurity account for most perinatal deaths in contemporary
diabetic pregnancy. Figure 9-5 shows the different rates
of RDS in diabetic and euglycemic pregnancies. In the
past decade, fewer intrauterine deaths have been reported,
probably reflecting more careful fetal monitoring. Nevertheless, intrapartum asphyxia and fetal demise remain persistent problems.
Birth Injury
Birth injury, including shoulder dystocia (Keller et al,
1991) and brachial plexus trauma, is more common among
IDMs, and macrosomic fetuses are at the highest risk
(Mimouni et al, 1992). Shoulder dystocia, defined as difficulty in delivering the fetal body after expulsion of the fetal
head, is an obstetric emergency that places the fetus and
mother at great risk. Shoulder dystocia occurs in 0.3% to
0.5% of vaginal deliveries among normal pregnant women;
the incidence is twofold to fourfold higher in women with
diabetes, probably because the hyperglycemia in a diabetic
pregnancy causes the fetal shoulder and abdominal widths
CHAPTER 9 Endocrine Disorders in Pregnancy
60
50
40
30
50
40
30
10
18901922
1926- 1946- 1956- 1966- 1970- 1973- 19761945 1955 1965 1969 1972 1975 1981
TIME PERIODS
FIGURE 9-4 Perinatal mortality rate (percentage) among infants of
diabetic mothers from 1890 to 1981. ( From Creasy RK, Resnik R, editors:
Maternal-fetal medicine: principles and practice, ed 2, Philadelphia,
1989, WB Saunders. Data from Craigin EB, Ryder GH: Obstetrics: a
practical textbook for students and practitioners, Philadelphia, 1916, Lea
and Febiger; DeLee JB: The principles and practice of obstetrics, ed 3,
Philadelphia, 1920, WB Saunders; Jorge CS, Artal R, Paul RH, et al: Antepartum fetal surveillance in diabetic pregnant patients, Am J Obstet Gynecol
141:641-645, 1981; Pedersen J: The pregnant diabetic and her newborn,
ed 2, Baltimore, 1977, Williams and Wilkins; and Williams JW: Obstetrics:
a textbook for the use of students and practitioners, New York, 1925,
D Appleton.)
TABLE 9-4 Perinatal Mortality Rates (No. of Deaths per 100
Births) in Diabetic and Normal Pregnancies
Mortality
Mothers With
Gestational
Diabetes
Mothers With
Preexisting
Diabetes
Healthy
Mothers*
Fetal
4.7
10.4
Neonatal
3.3
12.2
5.7
4.7
Perinatal
8.0
11.6
10.4
*California data for 1986, corrected for birthweight, sex, and race.
to become massive (Nesbitt et al, 1998). This relationship
was investigated by Athukorala et al, (2007), who found
a strong association with fasting hyperglycemia such that
with each 1-mmol increase in the fasting value in the oral
glucose-tolerance test there was an increasing relative risk
(RR) of 2.09 (95% confidence interval [CI], 1.03 to 4.25)
for shoulder dystocia. Although half of shoulder dystocias
occur in infants of normal birthweight (2500 to 4000 g),
the incidence of shoulder dystocia is 10-fold higher (5%
to 7%) among infants weighing 4000 g or more and rises
to 31% for infants whose mothers have diabetes (Gilbert
et al, 1999; see also Chapter 15 for a discussion of complicated deliveries and Chapter 64 for a discussion of the
neurologic consequences of birth injury.)
Neonatal Morbidity and Mortality
For a complete discussion of neonatal morbidity and
mortality, see Chapter 94.
Diabetic
20
10
0
20
0
60
NEW PERINATAL AND
NEONATAL TECHNOLOGY
RATE OF RDS (%)
PERINATAL MORTALITY (percent)
PRE
INSULIN
70 ERA
81
Non-Diabetic
30 31 32 33 34 35 36 37 38 39 40
GESTATIONAL AGE (WEEKS)
FIGURE 9-5 Rate of respiratory distress syndrome (RDS) versus
gestational age. Improved management of maternal glycemic control
permits delaying delivery until after 38 weeks’ gestation, when the risk
of RDS approaches that in nondiabetic pregnancy. ( From Moore TR:
A comparison of amniotic fluid fetal pulmonary phospholipids in normal and
diabetic pregnancy, Am J Obstet Gynecol 186:641-650, 2002.)
Polycythemia and Hyperviscosity
Polycythemia (defined as central venous hemoglobin concentration >20 g/dL or hematocrit >65%) is not uncommon in IDMs and is apparently related to glycemic control.
Widness et al (1990) demonstrated that hyperglycemia is
a powerful stimulus to fetal erythropoietin production,
probably mediated by decreased fetal oxygen tension.
Neonatal polycythemia may promote vascular sludging,
ischemia, and infarction of vital tissues, including the kidneys and central nervous system.
Neonatal Hypoglycemia
Approximately 15% to 25% of neonates delivered from
women with diabetes during gestation will develop hypoglycemia during the immediate newborn period (Alam
et al, 2006). This complication is usually much milder
and less common in the infant of a woman whose insulindependent diabetes is well controlled throughout the entire
pregnancy and who exhibits euglycemia during labor and
delivery. Unrecognized postnatal hypoglycemia can lead
to neonatal seizures, coma, and brain damage; therefore, it
is imperative that the nurseries receiving IDMs have a protocol for frequent monitoring of the infant’s blood glucose
level until metabolic stability is ensured.
Hyperbilirubinemia
The risk of hyperbilirubinemia is higher in IDMs than in
normal infants. There are multiple causes of hyperbilirubinemia in IDMs, but prematurity and polycythemia are
the primary contributing factors. Increased destruction
of red blood cells contributes to the risks of jaundice and
kernicterus. This complication is usually managed using
phototherapy, but exchange transfusions may be necessary
for marked bilirubin elevations.
Hypertrophic and Congestive
Cardiomyopathy
In some infants with macrosomia of mothers with poorly
controlled diabetes, a thickened myocardium and significant septal hypertrophy have been described (Gutgesell
et al, 1976). Although the prevalence of myocardial
82
PART III
Maternal Health Affecting Neonatal Outcome
hypertrophy in IDMs may exceed 30% at birth, almost all
cases have resolved by 1 year of age (Mace et al, 1979).
Hypertrophic cardiac dysfunction in a newborn IDM
often leads to respiratory distress, which may be mistaken
for hyaline membrane disease. IDMs with cardiomegaly
may have either congestive or hypertrophic cardiomyopathy. Echocardiograms show a hypercontractile, thickened
myocardium, often with septal hypertrophy disproportionate to the ventricular free walls. The ventricular chambers
are often smaller than normal, and there may be anterior
systolic motion of the mitral valve, producing left ventricular outflow tract obstruction.
The pathogenesis of hypertrophic cardiomyopathy in
IDMs is unclear, although it is recognized to be associated with poor maternal metabolic control. There is
evidence that the fetal myocardium is particularly sensitive to insulin during gestation, and Susa et al (1979)
reported a doubling of cardiac mass in hyperinsulinemic
fetal rhesus monkeys. The myocardium is known to be
richly endowed with insulin receptors. Recently maternal
insulin-like growth factor-1 (IGF-1), which is elevated in
suboptimally controlled diabetic pregnancies, has been
shown to be significantly elevated among neonates with
asymmetrical septal hypertrophy. Because IGF-1 does not
cross the placenta, it can exert its action through binding
to the IGF-1 receptor on the placenta (Hayati et al, 2007).
Halse et al (2005) noted that B-type natriuretic peptide, a
marker for congestive cardiac failure, is elevated in neonates whose mothers had poor glycemic control during
the third trimester.
IDMs can also have congestive cardiomyopathy without
hypertrophy. Echocardiography shows the myocardium to
be overstretched and poorly contractile (Jaeggi et al, 2001).
This condition is often rapidly reversible with correction of
neonatal hypoglycemia, hypocalcemia, and polycythemia;
it responds to digoxin, diuretics, or both. In contrast, treatment of hypertrophic cardiomyopathy with an inotropic or
diuretic agent tends to further decrease the size of the ventricular chambers and leads to obstruction of blood flow.
Prenatal echocardiogram can identify septal hypertrophy
and other structural abnormalities, but routine fetal echocardiogram in diabetics has not been proved to be costeffective or to improve outcomes (Bernard et al, 2009).
Respiratory Distress Syndrome
Since the 1970s, improved maternal management and
better protocols for timing of delivery have resulted in a
dramatic decline in the incidence of RDS from 31% to
3%. Nevertheless, respiratory dysfunction in the newborn
IDM continues to be a common complication of diabetic
pregnancy; it may be because of surfactant deficiency or
another form of pulmonary distress. Surfactant production
occurs late in diabetic pregnancies. Studies of fetal lung
ion transport in the diabetic rat by Pinter et al (1991) demonstrated decreased fluid clearance and a lack of thinning
of the lung’s connective tissue in diabetic rats compared
with controls. In humans, Kjos et al (1990) noted respiratory distress in 18 of 526 infants delivered after diabetic
gestations (3.4%). Surfactant-deficient airway disease
accounted for fewer than one third of cases, with transient
tachypnea, hypertrophic cardiomyopathy, and pneumonia
responsible for the majority.
As a result, the near-term infant of a mother with poorly
controlled diabetes is more likely to have neonatal RDS
than the infant of a mother without diabetes at the same
gestational age. This circumstance further compounds the
diabetic infant’s metabolic and cardiovascular difficulties
after birth. The fetus without diabetes achieves pulmonary
maturity at a mean gestational age of 34 to 35 weeks. By
37 weeks’ gestation, more than 99% of normal newborn
infants have mature lung profiles as assessed by phospholipid assays. In a diabetic pregnancy, however, it is unwise
to assume that the risk of respiratory distress has passed
until after 38.5 weeks’ gestation (Moore, 2002). Any delivery contemplated before 38.5 weeks’ gestation for other
than the most urgent fetal and maternal indications should
be preceded by documentation of pulmonary maturity
through amniocentesis.
OBSTETRIC COMPLICATIONS
Pregnancy complicated by diabetes is subject to a number
of obstetric disorders, including ketoacidosis, preeclampsia, polyhydramnios, and abnormal labor, at higher rates
than in nondiabetic pregnancy.
Preeclampsia
Preeclampsia is an unpredictable multisystem disorder
in which maternal neurologic, renal, and cardiovascular
status can decline precipitously, which can threaten fetal
health through placental ischemia and abruptio placentae. Preeclampsia is more common among women with
diabetes, occurring two to three times more frequently in
women with pregestational diabetes than in nondiabetic
women (Moore et al, 1985; Sibai et al, 2000). However,
the risk of developing preeclampsia is proportional to the
duration of diabetes before pregnancy and the existence of
nephropathy and hypertension; preeclampsia develops in
more than one third of women who have had diabetes for
more than 20 years. Patients with type 2 diabetes mellitus
without complications have a risk profile similar to that
of patients without diabetes, but the risk of hypertensive
complications is 50% higher in women with evidence of
renal or retinal vasculopathy than in those with no hypertension. The rate of fetal death is higher in women with
diabetes and preeclampsia. In the patient with diabetes
and chronic hypertension, preeclampsia may be difficult to distinguish from near-term blood pressure elevations. The onset is typically insidious and not confidently
recognized until it is severe. Renal function assessment
(creatinine, blood urea nitrogen, uric acid, and 24-hour
urine collection) should be performed each trimester in
women with evidence of pregestational diabetes and vascular disease.
When a patient with diabetes experiences preeclampsia,
she should be evaluated for delivery. If signs of severe disease are present (e.g., blood pressure >160 mm Hg systolic
and 110 mm Hg diastolic, neurologic symptoms, or significant renal dysfunction), delivery should be performed
promptly. In mild cases, the patient may be observed if
the fetal lungs are immature. Preeclampsia after 38 weeks’
gestation, however, is appropriate grounds for initiating
delivery.
CHAPTER 9 Endocrine Disorders in Pregnancy
Polyhydramnios
Polyhydramnios is defined as excess amniotic fluid. The precise clinical definition varies, encompassing the recording
of more than 2000 mL of amniotic fluid at delivery and various measures of amniotic fluid pocket depths as observed
on ultrasonography. In practice, polyhydramnios is usually
diagnosed when any single vertical pocket of amniotic fluid
is deeper than 8 cm (equivalent to the 97th percentile) or
when the sum of four pockets, one from each quadrant of
the uterus (amniotic fluid index), exceeds approximately 24
cm (95th percentile; Moore and Cayle, 1990). The principal cause of hydramnios in diabetic pregnancies is usually poor glycemic control, although fetal gastrointestinal
anomalies (e.g., esophageal atresia) need to be excluded.
A rapid increase in fundal height should prompt a thorough ultrasound examination by a skilled examiner. The
main clinical problems associated with hydramnios are
fetal malposition and preterm labor.
Management of the patient with hydramnios is predominantly symptomatic, focused on improving glycemic
control and preventing premature labor. Enhanced patient
awareness of contractions and the signs and subtle sensations of preterm labor is essential.
MANAGEMENT
Preconception Management
Preconception counseling and a detailed medical risk assessment are recommended for all women with overt diabetes
as well as for those with a history of gestational diabetes in a
previous pregnancy. The significant effects on the maternal
and neonatal complications of diabetic pregnancy cannot
be realized until meticulous preconception metabolic control is achieved in all women contemplating pregnancy.
The important elements to be considered in preconception counseling of patients with diabetes are the patient’s
level of glycemic control; current status of the patient’s
retinal and renal health; and any medications being
taken, especially antihypertensive or thyroid medications.
A realistic assessment of the patient’s risk of complications
during pregnancy, including worsening of renal or ophthalmologic function, should be provided.
Preconception management should lead to a comprehensive program of glucose control. The major goals of
the prepregnancy metabolic program are as follows:
ll
ll
ll
ll
Establishing a regimen of frequent, regular monitoring
of capillary blood glucose levels
Adopting an insulin dosing regimen that results in a
smooth interprandial glucose profile (fasting blood
glucose value 90 to 99 mg/dL, 1-hour postprandial
glucose level of less than 140 mg/dL or 2-hour postprandial glucose level less than 120 mg/dL, no reactions between meals or at night)
Bringing HbA1c level into the normal range
Developing family, financial, and personal resources to
assist the patient if pregnancy complications require
that she lose work time or assume total bed rest
This preconception care has been shown to decrease
congenital anomalies and to result in fewer hospitalizations,
83
fewer infants requiring intravenous glucose after delivery,
and a substantial reduction in total costs (Herman et al,
1999).
Prenatal Metabolic Management
The goals of glycemic monitoring, dietary regulation, and
insulin therapy in diabetic pregnancy are to prevent the postnatal sequelae of diabetes in the newborn—macrosomia,
shoulder dystocia, and postnatal metabolic instability.
These measures must be instituted early and aggressively
if they are to be effective.
Principles of Dietary Therapy
Because women with diabetes have inadequate insulin action after feeding, the goal of dietary therapy is to
avoid single, large meals and foods with a large proportion of simple carbohydrates. Three major meals and three
snacks are prescribed. The use of nonglycemic foods that
release calories into the gut slowly also improves metabolic
control.
Nutritional therapy should be supervised by a trained
professional who performs formal dietary assessment and
counseling at several points during the pregnancy. The
dietary prescription should provide adequate quantity and
distribution of calories and nutrients to meet the needs of
the pregnancy and support achieving the plasma glucose
targets that have been established. For obese women (body
mass index >30 kg/m2), a 30% to 33% restriction in caloric
intake (to 25 kcal per kilogram of actual weight per day or
less) has been shown to reduce hyperglycemia and plasma
triglycerides with no increase in ketonuria. Moderate
restriction of dietary carbohydrate intake to 35% to 40%
of calories has been shown to reduce maternal glucose levels and improve maternal and fetal outcomes (Major et al,
1998). In a nonrandomized study, subjects with low carbohydrate intake (<42% of calories) had lower requirements
for insulin for glucose control and significantly lower rates
of macrosomia and cesarean deliveries for cephalopelvic
disproportion and macrosomia. Recently, a randomized
trial was performed in which 958 women with gestational
diabetes were randomized to usual prenatal care or treatment (Landon, 2009). Women randomized to treatment
underwent nutritional counseling, diet therapy, and insulin if indicated. Among those who underwent treatment
there was lower mean birthweight, neonatal fat mass, rates
of large for gestational age and macrosomic (>4000 g)
infants. There was also a trend toward lower cord C-peptide levels in the treatment group. Maternal outcomes
were significant for lower rates of cesarean delivery, preeclampsia, and shoulder dystocia (Landon et al, 2009).
Principles of Glucose Monitoring
The availability of chemical test strips for capillary blood
glucose measurements has revolutionized the management
of diabetes, and their use should now be considered the
standard of care for pregnancy monitoring. The discipline
of measuring and recording blood glucose levels before
and after meals may have the effect of improving glycemic
control (Goldberg et al, 1986).
Controversy exists as to whether the target glucose levels to be maintained during diabetic pregnancy should be
84
PART III
Maternal Health Affecting Neonatal Outcome
designed to limit macrosomia or to closely mimic nondiabetic pregnancy profiles. The Fifth International Workshop Conference on Gestational Diabetes (Metzger et al,
2007) recommended the following: fasting plasma glucose
less than 90 to 99 mg/dL (5.0 to 5.5 mmol/L) and 1-hour
postprandial plasma glucose less than 140 mg/dL (7.8
mmol/L), or 2-hour postprandial plasma glucose less than
120 to 127 mg/dL (6.7 to 7.1 mmol/L).
The glycemic control profiles from Cousins et al (1980)
were derived from highly controlled studies in which volunteer subjects were fed test meals with specific caloric
content on a rigid schedule. Parretti et al (2001) profiled
normal pregnant women twice monthly, preprandially,
and postprandially during the third trimester. Testing was
conducted with capillary glucose meters, and the women
followed an ad libitum diet. The data demonstrate that
fasting and premeal plasma glucose levels are usually less
than 80 mg/dL and often less than 70 mg/dL. Peak postprandial plasma glucose values rarely exceed 110 mg/dL.
Yogev et al (2004) used a sensor that monitored interstitial
fluid glucose levels to obtain continuous glucose information from pregnant women without diabetes and found
similar results to those of Parretti et al (2001). The range
of normal glucose levels in nondiabetic pregnancy is summarized in Table 9-5.
Postprandial values must be assessed because they have
the strongest correlation with fetal growth (JovanovicPeterson et al, 1991). The Diabetes in Early Pregnancy
Study found that postprandial glucose levels were strongly
predictive of both birthweight and the overall percentage of
macrosomic infants. With postprandial glucose values averaging 120 mg/dL, approximately 20% of infants were macrosomic; a 30% rise in postprandial levels to 160 mg/dL
resulted in a predicted percentage of macrosomia of 35%.
Similar results were reported by de Veciana et al (1995).
Compared with the group who performed preprandial
glucose monitoring, the group performing postprandial
glucose monitoring demonstrated a greater mean change
in the HbA1c value (3.0% vs. 0.6%; p <0.001), lower birthweights (3469 g vs. 3848 g; p = 0.01), and lower rates of
both neonatal hypoglycemia (3% vs. 21%, p = 0.05) and
macrosomia (12% vs. 42%; p = 0.01).
A typical schedule involves performing blood glucose
checks upon rising in the morning, 1 or 2 hours after
breakfast, before and after lunch, before and after dinner,
and before bedtime. The goal of physiologic glycemic control in pregnancy, however, is not met by simply avoiding
hypoglycemia. The data summarized here regarding fetal
macrosomia and postnatal morbidity emphasize the key
role of excessive postprandial excursions in blood glucose
values. Therefore, close attention must be paid to preprandial and postprandial glycemic profiles.
Principles of Insulin Therapy
No available insulin delivery method approaches the precise secretion of the hormone from the human pancreas.
The therapeutic goal of exogenous insulin therapy during
pregnancy is to achieve diurnal glucose excursions similar
to those of nondiabetic pregnant women. Normal pregnant women maintain postprandial blood glucose excursions within a relatively narrow range (70 to 120 mg/dL).
As pregnancy progresses, the fasting and between-meal
blood glucose levels drop progressively lower as a result
of the continual uptake of glucose from the maternal circulation by the growing fetus. Any insulin regimen for
pregnant women must be designed to avoid excessive
unopposed insulin action during the fasting state.
Insulin type and dosage frequency should be individualized. Use of regular insulin before each major meal helps to
limit postprandial hyperglycemia. To provide basal insulin levels between feedings, a longer-acting preparation is
necessary, such as isoprotane insulin (NPH) or insulin zinc
(Lente). Typical subcutaneous insulin dosing regimens are
two thirds of total insulin in the morning, of which two
thirds as intermediate-acting and one third as regular insulin. The remaining third of the total insulin dose is given
in the evening, with 50% as short-acting insulin before
dinner and 50% as intermediate-acting given at bedtime.
The use of an insulin pump for type 1 diabetes mellitus
during pregnancy has become more widespread (Gabbe
et al, 2000). An advantage of this approach is the more
physiologic insulin release pattern that can be achieved
with the pump.
Ultrasonography has also been used to direct insulin
management (Rossi et al, 2000). Kjos et al (2001) showed
that serial normal fetal abdominal circumference measurements can be used to avoid insulin therapy without
increasing neonatal morbidity.
Oral Hypoglycemic Therapy
Historically, insulin has been the mainstay of therapy
for gestational diabetes because of early reports that oral
hypoglycemic drugs are a potential cause of fetal anomalies and neonatal hypoglycemia. Sulfonylurea compounds
are contraindicated during pregnancy because of a high
level of transplacental penetration and clinical reports
of prolonged and severe neonatal hypoglycemia (Zucker
and Simon, 1968). An increased rate of congenital malformations, particularly ear anomalies, has been reported
from a small case-control study (Piacquadio et al, 1991).
TABLE 9-5 Ambulatory Glucose Values in Pregnant Women With Normal Glucose Tolerance
No. of Subjects
Fasting (mg/dL)
Postprandial Level at
60 min (mg/dL)
Parretti et al, 2001
51
69 (57-81)
108 (96-120)
—
Yogev et al, 2004
57
75 (51-99)
105 (79-131)
110 (68-142)*
Study
Postprandial
Peak (mg/dL)
Adapted from Metzger BE, Buchanan TA, Coustan DR, et al: Summary and recommendations of the Fifth International Workshop-Conference on Gestational Diabetes Mellitus,
Diabetes Care 30:S251-S260, 2007.
*The time of the peak postprandial glucose concentration was 70 minutes (range, 44 to 96 minutes).
CHAPTER 9 Endocrine Disorders in Pregnancy
However, when Towner et al (1995) evaluated the frequency of birth defects in patients who took oral hypoglycemic agents during the periconception period, they
noted that first-trimester HbA1c level and duration of
diabetes were strongly associated with fetal congenital
anomalies, but that use of oral hypoglycemic medications
was not.
Glyburide, a second-generation sulfonylurea, has been
shown to cross the placenta minimally in laboratory studies (Elliott et al, 1994) and in a large clinical trial. The
prospective, randomized trial conducted by Langer et al
(2000) compared glyburide and insulin in 404 women with
gestational diabetes and showed equivalently excellent
maternal glycemic control and perinatal outcomes.
Beyond this single, encouraging study, experience with
glyburide during pregnancy is limited (Coetzee and Jackson, 1985; Lim et al, 1997). Chmait et al (2004), reporting
experience with 69 patients with gestational diabetes who
were given glyburide, found a failure rate of 19% (>10%
glucose values above target). Glyburide failure rate was
higher in women receiving a diagnosis earlier in pregnancy
(20 vs. 27 weeks’ gestation; p <0.003) and whose average
fasting glucose in the week before starting glyburide was
higher (126 vs. 101 mg/dL).
Following the publication of the randomized control
trial, several retrospective series have been published
comprising 504 glyburide-treated patients, summarized
recently by Moore (2007). Jacobson et al (2005) performed
a retrospective cohort comparison of glyburide and insulin treatment of gestational diabetes. The insulin group
(n = 268) consisted of those diagnosed in 1999 through
2000 and the glyburide group (n = 236) was diagnosed in
2001 through 2002. Glyburide dosing was begun with 2.5
mg in the morning and increased by 2.5 to 5.0 mg weekly.
If the dose exceeded 10 mg daily, twice daily dosing was
considered. If glycemic goals were not met on a maximum
daily dose of 20 mg, treatment was changed to insulin.
There were no statistically significant differences in gestational age at delivery, mode of delivery, birthweight, large
for gestational age (LGA), or percent macrosomia. The
rate of preeclampsia doubled in the glyburide group (12%
vs 6%; p = 0.02). Women in the glyburide group also had
significantly lower posttreatment fasting and postprandial
blood glucose levels. The glyburide group was also superior in achieving target glycemic levels (86% vs. 63%;
p <0.001). The failure rate (transfer to insulin) was 12%.
The study size, however, was insufficient to detect less
than a doubling of the rate of macrosomia/LGA and a 44%
increase in neonatal hypoglycemia.
Conway et al (2004) reported a retrospective cohort of
75 glyburide-treated patients with GDM. Good glycemic
control was achieved by 84% of the subjects with glyburide, and treatment for 16% was switched to insulin. The
rate of fetal macrosomia was similar between women successfully treated with glyburide and those who received
insulin (11.1% vs. 8.3%; p = 1.0), and mean birthweight
was also similar. Of note, a nonsignificantly higher proportion of infants in the glyburide group required intravenous glucose infusions because of hypoglycemia (25.0%
vs. 12.7%; p = 0.37). Currently there is a growing acceptance of glyburide use as a primary therapy for GDM
(Coustan, 2007).
85
OTHER AGENTS
Metformin is frequently used in patients with polycystic
ovary syndrome and type 2 diabetes mellitus to improve
insulin resistance and fertility (Legro et al, 2007). Metformin therapy has been demonstrated to improve the
success of ovulation induction (Vandermolen et al, 2001)
and may reduce first-trimester pregnancy loss in women
with polycystic ovary syndrome (Jakubowicz et al, 2002).
However, the effects of continuing metformin treatment
during pregnancy are currently being studied. Older studies evaluating the efficacy and safety of the treatment of
pregestational and gestational diabetics with metformin
raised concerns regarding a higher perinatal mortality, a
higher rate of preeclampsia, and failure of therapy (Coetzee and Jackson 1979; Hellmuth et al, 2000). However,
the metformin-treated women were older, more obese,
and treated later in pregnancy.
A more recent cohort study of metformin in pregnancy
by Hughes and Rowan (2006) included 93 women with
metformin treatment (only 32 continued until delivery)
and 121 controls. There was no difference in perinatal
outcomes between the groups. Glueck et al (2002) compared women without diabetes but with polycystic ovary
syndrome who conceived while taking metformin and
continued the agent through delivery (n = 28) to matched
women without metformin therapy (n = 39). Gestational
diabetes developed in 31% of women who did not take
metformin versus 3% of those who did (odds ratio, 0.115;
95% CI, 0.014 to 0.938).
Recently a large randomized controlled trial was performed comparing metformin to insulin for the treatment
of gestational diabetes (Rowan et al, 2008). This study was
powered to detect a 33% increase in composite outcome
(neonatal hypoglycemia, respiratory distress, need for
phototherapy, birth trauma, 5-minute American Pediatric
Gross Assessment Record score of less than 7, or prematurity) in neonates born to mothers treated with metformin.
Seven hundred fifty-one women with gestational diabetes between 20 and 30 weeks’ gestation were randomized to metformin or insulin. Of these, 363 women were
assigned to metformin and 370 were assigned to insulin.
Forty-six percent of women receiving metformin required
the addition of insulin to obtain adequate glycemic control. There were no differences in the rate of the primary
composite outcome. There was a lower rate of severe neonatal hypoglycemia in the metformin-treated group and
no differences in neonatal anthropometric measurements.
There was, however, a higher rate of prematurity in the
metformin-treated group (12.1%) versus the insulin group
(7.6%). A follow-up study is currently under way to assess
the offspring of these women at 2 years of age.
Prenatal Obstetric Management
The overall strategy for managing a diabetic pregnancy in
the third trimester involves two goals: (1) preventing stillbirth and asphyxia and (2) monitoring growth of the fetus
to select the proper time and route of delivery to minimize
maternal and infant morbidity. The first goal is accomplished
by testing fetal well-being at frequent intervals, and the second through ultrasonographic monitoring of fetal size.
86
PART III
Maternal Health Affecting Neonatal Outcome
Periodic Biophysical Testing of the Fetus
A variety of biophysical tests of the fetus are available to
the clinician, including fetal heart rate testing, fetal movement assessment, ultrasound biophysical scoring, and fetal
umbilical Doppler studies. Most of these tests, if applied
properly, can be used with confidence to provide assurance
of fetal well-being while awaiting fetal maturity; they are
summarized in Table 9-6.
Testing should be initiated early enough to avoid significant risk of stillbirth, but not so early that the risk of a
false-positive result is high. In patients with poor glycemic
control or significant hypertension, testing should begin
as early as 28 weeks’ gestation. In lower-risk patients, most
centers begin formal fetal testing by 34 to 36 weeks’ gestation. Counting of fetal movements is performed in all
pregnancies from 28 weeks’ gestation onward.
Assessing Fetal Growth
Monitoring of fetal growth continues to be a challenging
and highly inexact process. Although the current tools,
consisting of serial plotting of fetal growth parameters,
are superior to earlier clinical estimations, accuracy is still
±15%. Single and multiple longitudinal assessments of
fetal size have been attempted.
Calculation of Estimated Fetal Weight
Several polynomial formulas using combinations of head,
abdominal, and limb measurements have been proposed to
predict the weight of the macrosomic fetus from ultrasonography parameters (Ferrero et al, 1994; Tongsong et al,
TABLE 9-6 Tests of Fetal Well-Being
Reassuring
Result
Test
Frequency
Counting
of fetal
movements
Every night
from 28
weeks’
gestation
Ten movements in
<60 min
Comment
Performed in all
patients
Nonstress
test
Twice
weekly
Two heart rate
accelerations
in 20 min
Begin at: 28-34
wk in patients
with insulindependent
diabetes, 36 wk
in patients with
diet-controlled
gestational
diabetes
Contraction
stress
test
Weekly
No heart rate
decelerations
in response
to ≤3 contractions in
10 min
Same as for nonstress test
Ultrasound
biophysical
profile
Weekly
Score of 8 in
30 min
The following
findings are
given 2 points
each:
3 movements
1 flexion
30 sec breathing
2 cm amniotic
fluid
1994). Unfortunately, in such formulas, small errors in
individual measurements of the head, abdomen, and femur
are typically multiplied together. In the obese fetus, the
inaccuracies are further magnified. Bernstein and Catalano (1992) observed that a significant correlation exists
between the degree of error in the ultrasonographically
estimated fetal weight and the percentage of body fat on
the fetus (r = 0.28; p <0.05). Perhaps this problem explains
why no single formula has proved to be adequate in identifying the macrosomic fetus (Tamura et al, 1985).
Shamley and Landon (1994) reviewed the relative accuracy of the various available formulas. Approximately 75%
of the fetal weight predictions were within 10% of actual
birthweight, with sensitivity for detecting macrosomia
varying greatly (11% to 76%). In another study, McLaren
et al (1995) found that 65% of weight estimates based on
a simple abdominal circumference and femur length formula were within 10% of actual weight. A similar accuracy
was achieved with more complex models (53% to 66% of
estimates within the range).
The formula developed by Shepard et al (1982), which
uses biparietal diameter and abdominal circumference, is
readily available in textbooks and is used most commonly
in current ultrasonographic equipment software. The formula has accuracy levels similar to the statistics quoted
previously.
Serial Estimated Fetal Weight Assessments
Because prediction of fetal weight from a single set of measurements is inaccurate, serial estimates showing the trend
of ultrasonographic parameters (typically made every 1.5
to 3 weeks) might theoretically offer a better estimate of
actual weight percentile. A comparison of the efficacy of
serial estimated fetal weight calculations to a single measurement, however, did not show better predictive accuracy. Larsen et al (1995) reported that predictions based on
the average of repeated weight estimates, on linear extrapolation from two estimates, or on extrapolation by a secondorder equation fitted to four estimates were no better than
the prediction from the last estimate before delivery. Similar findings (that a single estimate is as accurate as multiple
assessments) were reported by Hedriana and Moore (1994).
Choosing the Timing and Route of Delivery
Timing of delivery should be selected to minimize maternal
and neonatal morbidity and mortality. Delaying delivery to
as near as possible to the due date helps to maximize cervical ripeness and improve the chances of spontaneous labor
and vaginal delivery. Yet the risks of fetal macrosomia,
birth injury, and fetal death rise as the due date approaches
(Rasmussen et al, 1992). Although earlier delivery at 37
weeks’ gestation might reduce the risk of shoulder dystocia, the higher rates of failed labor inductions and poor
neonatal pulmonary status at this time must be considered.
Therefore an optimal time for delivery of most diabetic
pregnancies is between 39.0 and 40 weeks’ gestation.
Delivery of a diabetic patient before 39 weeks’ gestation
without documentation of fetal lung maturity should be performed only for compelling maternal or fetal reasons. Fetal
lung maturity should be verified in such cases from the presence of more than 3% phosphatidyl glycerol or the equivalent in amniotic fluid as ascertained from an amniocentesis
CHAPTER 9 Endocrine Disorders in Pregnancy
specimen. After 39 weeks’ gestation, the obstetrician can
await spontaneous labor if the fetus is not macrosomic and
results of biophysical testing are reassuring. In patients with
gestational diabetes and superb glycemic control, continued
fetal testing and expectant management can be considered
until 41 weeks’ gestation (Lurie et al, 1992).
Given the previous data, the decision to attempt vaginal delivery or perform a cesarean section is inevitably
based on limited data. The patient’s obstetric history from
previous pregnancies, the best estimation of fetal weight,
the fetal adipose profile (abdomen larger than head), and
results of clinical pelvimetry should all be considered.
A policy of elective cesarean section for suspected fetal
macrosomia (ultrasonographically estimated fetal weight of
greater than 4500 g) would require 443 cesarean deliveries
to avoid one permanent brachial plexus injury (Rouse et al,
1996). Most large series of diabetic pregnancies report a
cesarean section rate of 30% to 50%. The best means by
which this rate can be lowered is early and strict glycemic
control in pregnancy. Conducting a long labor induction
in the patient with a large fetus and marginal pelvis may
increase rather than decrease morbidity and costs.
Intrapartum Glycemic Management
Maintenance of intrapartum metabolic homeostasis is
essential to avoid fetal hypoxemia and promote a smooth
postnatal transition. Strict maternal euglycemia during
labor does not guarantee newborn euglycemia in infants
with macrosomia and long-established islet cell hypertrophy. Nevertheless, the use of a combined insulin and
glucose infusion during labor to maintain maternal blood
glucose in a narrow range (80 to 110 mg/dL) during labor
is a common and reasonable practice. Typical infusion
rates are 5% dextrose in lactated Ringer’s solution at 100
mL/hr and regular insulin at 0.5 to 1.0 U/hr. Capillary
blood glucose levels are monitored hourly in such patients.
For patients with diet-controlled gestational diabetes
in labor, avoiding dextrose in all intravenous fluids normally maintains excellent blood glucose control. After 1 to
2 hours, no further assessments of capillary blood glucose
are typically necessary.
Neonatal Management
Neonatal Transitional Management
One of the metabolic problems common to IDMs is hypoglycemia, which is related to the level of maternal glycemic control over the 6 to 12 weeks before birth. Neonatal
hypoglycemia is most likely to occur between 1 and 5
hours after birth, as the rich supply of maternal glucose
stops with ligation of the umbilical cord and the infant’s
levels of circulating insulin remain elevated. These infants
therefore require close monitoring of blood glucose concentration during the first hours after birth. IDMs also
appear to have disorders of catecholamine and glucagon
metabolism as well as diminished capability to mount normal compensatory responses to hypoglycemia.
In the past, IDMs were treated with glucagon; however, this treatment frequently results in high blood glucose levels that trigger insulin secretion and repeated
cycles of hypoglycemia and hyperglycemia. Current
87
recommendations, therefore, consist of early oral feeding,
when possible, with infusion of intravenous glucose.
Ordinarily, blood glucose levels can be controlled satisfactorily with an intravenous infusion of 10% glucose. If
greater amounts of glucose are required, bolus administration of 2 mL/kg of 10% glucose is recommended. Close
monitoring to correct hypoglycemia while avoiding hyperglycemia and consequent stimulation of insulin secretion is
important.
Breastfeeding
Most authorities prefer to maintain strict monitoring of
newborn IDM glucose levels for at least 4 to 6 hours, which
frequently necessitates admission to a newborn special care
unit. IDMs who are delivered atraumatically and are well
oxygenated, however, can be kept with their mothers while
undergoing close glycemic monitoring for the first 1 to
2 hours of life. This approach permits early breastfeeding,
which may reduce the need for intravenous glucose therapy.
Summary
Intensive management of women with glucose intolerance
during pregnancy has resulted in markedly improved pregnancy outcomes. Despite these advances, care of the IDM
continues to require vigilance and meticulous monitoring
with a full understanding of the quality of the glycemic
milieu in which the infant developed.
DISORDERS OF THE THYROID
INCIDENCE
Thyroid disorders, in general, are more common in
women than in men and represent a common endocrine
abnormality during pregnancy. Both hyperthyroidism and
hypothyroidism in the mother put the infant at risk and
require careful management by the perinatal-neonatal
team. Table 9-7 presents an overview of the approach to
infants who are thought to be at risk for abnormal thyroid function because of maternal thyroid abnormalities.
The most frequently described problem is the syndrome of
postpartum thyroiditis, which has been reported to complicate as many as 5% of all pregnancies. The diagnosis of
thyroid disease in pregnancy is complicated by the natural
changes that occur in immunologic status of the mother
and fetus and that variously affect the assessment of any of
the autoimmune thyroid disorders.
MATERNAL-FETAL THYROID FUNCTION
IN PREGNANCY
Maternal Thyroid Function
Several pregnancy-related physiologic conditions affect
maternal thyroid function, and the appropriate interpretation of thyroid function test results during pregnancy
must take these normal physiologic factors into account.
One important modification that occurs during pregnancy
is the estrogen-dependent increase in thyroid-binding
globulin. This results in an increase in total thyroxine and
total triiodothyronine levels throughout pregnancy. The
88
PART III
Maternal Health Affecting Neonatal Outcome
TABLE 9-7 Approaches for Infants Judged to be at Risk for Abnormal Thyroid Function
Possible Thyroid Abnormality
Cord Blood
Analyses
Assessment at Birth
Assessment at 2-7 Days
of Life
Congenital hyperthyroidism because
mother has any of the following:
Graves’ disease with hyperthyroidism
and may have been treated with PTU,
methimazole, 131I, iodides, or surgery;
history of Graves’ disease; history of
Hashimoto’s disease
T4, TSH, TSAb
Physical examination for intrauterine
growth restriction, goiter, exophthalmos, tachycardia, bradycardia,
size of anterior fontanel, synostosis,
congenital anomalies; determination
of gestational age by dates, ultrasonography during pregnancy, or
Dubovitz examination; neurologic
examination; determination of bone
age (knee); for selected cases: electrocardiogram, auditory and visual
evoked potentials, motor conduction,
velocity tests, skull radiographs
T4, TSH, TSAb (if available)
eterminations:
d
If results normal, give no treatment; observe and repeat T4,
T3, TSH determinations at
7-10 days*; if results indicate
hypothyroidism, repeat T4, TSH;
if results are abnormal, begin
treatment at 7-10 days†; if results
indicate hyperthyroidism, begin
treatment with PTU (8 mg/kg)
and propranolol
Congenital or early childhood hypothyroidism because mother has any of the
following:
Graves disease with excessive PTU
therapy; Hashimoto’s disease; acute
(subacute) thyroiditis; familial genetic
defect in thyroxine synthesis; treatment
with iodides or lithium for nonthyroidal
illness; exposure to 131I while pregnant
T4, TSH,
ThyAb
Same as for hyperthyroidism
T4, TSH, ThyAb determinations:
If results indicate hypothyroidism, perform ultrasound scan to
define presence, size, location of
thyroid tissue; if hypothyroidism
confirmed, begin treatment with
levothyroxine sodium (Synthroid), 0.05 mg/day
Adapted from Creasy RK, Resnik R, editors: Maternal-fetal medicine: principles and practice, ed 2, Philadelphia, 1989, WB Saunders.
I, Radioactive iodine; PTU, propylthiouracil; T4, thyroxine; TSAb, thyroid-stimulating antibody; T3, triiodothyronine; ThyAb, thyroid antibody; TSH, thyroid-stimulating
hormone.
*Graves disease does not develop at 7 to 10 days in children whose mothers receive PTU.
levels of unbound (free) thyroxine (FT4) and free triiodothyronine (FT3), as well as levels of thyroid-stimulating
hormone (TSH), in general remain unchanged. However,
human chorionic gonadotropin, a second factor in pregnancy that may modify thyroid function, has a stimulatory
effect on the thyroid and may transiently effect the FT4,
FT3, and TSH levels in the first trimester and early in the
second trimester. This stimulatory affect of human chorionic gonadotropin rarely causes aberrations of the thyroid
function parameters into the thyrotoxic range (American
College of Obstetricians and Gynecologists, 2001).
Fetal Thyroid Function
The fetal thyroid actively concentrates iodide after 10 weeks’
gestation, releases thyroxine (T4) after 12 weeks’ gestation,
and becomes responsive to pituitary TSH at 20 weeks’ gestation. Although maternal TSH does not cross the placenta,
maternal thyroid hormones and thyrotropin-releasing hormone are transferred to the fetus throughout gestation.
Early studies found that cord blood thyroid function testing
of neonates with congenital thyroid agenesis revealed hormone levels that were 30% of normal (Vulsma et al, 1989),
suggesting a maternal source. Recent studies show that by 4
weeks after conception, small amounts of T4 and triiodothyronine (T3) from the maternal origin are found in the fetal
compartment, with T4 levels increasing throughout gestation. Free T4 levels reach concentrations of biologic significance in the adult by midgestation. Studies using rat models
have demonstrated that this thyroid hormone is important
for corticogenesis early in the pregnancy (Morreale de
Escobar et al, 2004). Transplacental transfer of thyroidstimulating immunoglobulin (TSI) may occur, causing fetal
thyrotoxicosis. Other substances that may be transferred
from the maternal compartment to the fetal compartment
and affect fetal thyroid function are iodine, a radioactive isotope of iodine, propylthiouracil (PTU), and methimazole.
The fetal effects of these agents are reviewed later.
Hyperthyroidism
Hyperthyroidism occurs in approximately 0.2% of pregnancies and results in a significant increase in the prevalence of both low-birthweight delivery and a trend toward
higher neonatal mortality. The most common cause of
thyrotoxicosis (85% of cases) in women of child-bearing
age is Graves’ disease; other causes are acute (or subacute)
thyroiditis (transient), Hashimoto’s disease, hydatidiform
mole, choriocarcinoma, toxic nodular goiter, and toxic
adenoma. Graves’ disease has a peak incidence during the
reproductive years, but patients with the disorder may
actually have remissions during pregnancy, followed by
postpartum exacerbations. The unique feature of these
pregnancies is that the fetus may also be affected, regardless of the mother’s concurrent medical condition. Thyroid
function is difficult to evaluate in the fetus, and the status
of the fetus may not correlate with that of the mother.
Diagnosis
The differential diagnosis of thyrotoxicosis becomes
more difficult during pregnancy because normal pregnant
women may have a variety of hyperdynamic signs and
symptoms—intolerance to heat, nervousness, irritability,
emotional lability, increased perspiration, tachycardia,
and anxiety. Laboratory data are also difficult to evaluate
CHAPTER 9 Endocrine Disorders in Pregnancy
because total serum thyroxin values are normally elevated
during pregnancy as a result of estrogen-induced increases
in thyroxine-binding globulin. Therefore if thyroxinebinding globulin is increased, then T3 resin uptake may
be in the euthyroid to slightly increased range in a patient
who has true hyperthyroidism. Hollingsworth (1989) has
reviewed the assessment of thyroid function tests in nonpregnant and pregnant women, along with the differential
diagnosis of hyperthyroidism during pregnancy.
Pathogenesis of Graves’ Disease
The pathogenesis of Graves’ disease is not completely
understood, but it probably represents an overlapping
spectrum of disorders that are characterized by the production of polyclonal antibodies. It has been appreciated
since the 1960s (Sunshine et al, 1965) that abnormal TSIs,
which appear to be immunoglobulin G, are present in
pregnant women with Graves’ disease and cross the placenta easily to cause neonatal hyperthyroidism in some
infants (McKenzie and Zakarija, 1978). The clinical spectrum of Graves’ disease in utero is broad and may result in
stillbirth or preterm delivery. Some affected infants have
widespread evidence of autoimmune disease, including
thrombocytopenic purpura and generalized hypertrophy
of the lymphatic tissues. Thyroid storm can occur shortly
after birth, or the infant may have disease that is transient
in nature, lasting from 1 to 5 months. Infants born to mothers who have been treated with thioamides may appear
normal at birth, but demonstrate signs of thyrotoxicosis
at 7 to 10 days of age, when the effect of thioamide suppression of thyroxine synthesis is no longer present. The
measurement of thyroid-stimulating antibodies is useful in
predicting whether the fetus will be affected.
Management of the Mother
Because radioactive iodine therapy is contraindicated during pregnancy, treatment of the pregnant woman with
thyrotoxicosis involves a choice between antithyroid drugs
A
B
89
and surgery. The therapeutic goal is to achieve a euthyroid, or perhaps slightly hyperthyroid, state in the mother
while preventing hypothyroidism and hyperthyroidism in
the fetus. Either PTU or methimazole can be used to treat
thyrotoxicosis during pregnancy. Because methimazole
therapy can be associated with aplasia cutis in the offspring
of treated women, and because PTU crosses the placenta
more slowly than methimazole, PTU has become the drug
of choice for use during pregnancy. Ordinarily, thyrotoxicosis can be controlled with doses of 300 mg per day. Once
the disorder is under control, however, it is important to
keep the dose as low as possible, preferably less than 100
mg daily, because this drug crosses the placenta and blocks
fetal thyroid function, possibly producing hypothyroidism
in the fetus.
In women with cardiovascular effects, the use of betablockers may be appropriate to achieve rapid control of
thyrotoxicosis. Because administration of propranolol to
pregnant women has been associated with intrauterine
growth restriction and impaired responses of the fetus to
anoxic stress as well as postnatal bradycardia and hypoglycemia, the doses must be closely controlled. Iodides have
also been used, particularly in combination with betablocking agents, to control thyrotoxicosis. Long-term
iodide therapy presents a risk to the fetus. Because of the
inhibition of the incorporation of iodide into thyroglobulin, a large, obstructive goiter can develop in the fetus. Surgery during pregnancy is best reserved for cases in which
the mother is hypersensitive to antithyroid drugs, compliance with medication is poor, or drugs are ineffective in
controlling the disease.
Effects on the Newborn
Approximately 1% of infants born to mothers with some
level of thyrotoxicosis have thyrotoxicosis (Figure 9-6).
Assessment of fetal risk in utero includes measurement of
TSIs, with the expectation that if the titers are high, there
is a higher risk of thyrotoxicosis. Additional assessment of
the fetus should pay particular attention to elevated resting
C
FIGURE 9-6 A, Hypothyroid 21-year-old mother who experienced Graves’ disease at age 7 years and was treated by subtotal thyroidectomy. She
was given maintenance therapy with daily levothyroxine sodium (Synthroid; 0.15 mg) throughout pregnancy. B, Her infant girl was born at term with
severe Graves’ disease, goiter, and exophthalmos that persisted for 6 months. C, The child was healthy at 20 months old. ( From Creasy RK, Resnik R,
editors: Maternal-fetal medicine: principles and practice, ed 2, Philadelphia, 1989, WB Saunders.)
90
PART III
Maternal Health Affecting Neonatal Outcome
heart rate and poor fetal growth. Daneman and Howard
(1980) reported on the outcome of nine infants with neonatal thyrotoxicosis and noted normal growth, but a high
incidence of craniosynostosis and intellectual impairment.
It may be necessary to treat the asymptomatic mother with
thioamides and propranolol (and thyroid replacement)
during pregnancy to treat the infant and prevent serious
neonatal morbidity and long-term problems.
Mothers with thyrotoxicosis who are taking normal
doses of thioamide can safely breastfeed their infants,
although thioamide appears in breast milk in low amounts.
Currently there does not appear to be any long-term
adverse outcome for infants whose mothers have received
PTU during pregnancy.
HYPOTHYROIDISM
Hypothyroidism complicates about 1 to 3 per 1000 pregnancies. The leading cause of hypothyroidism in pregnancy is Hashimoto’s thyroiditis, which is a chronic
autoimmune thyroiditis characterized by painless inflammation and enlargement of the thyroid gland (Casey and
Leveno, 2006). Other causes of primary hypothyroidism include iodine deficiency, thyroidectomy, or ablative radioiodine therapy for hyperthyroidism. Secondary
causes of hypothyrodism include Sheehan’s syndrome
caused by obstetric hemorrhage leading to pituitary
ischemia, necrosis and abnormalities in all pituitary hormones, lymphocytic hypophysitis, and hypophysectomy
(Casey and Leveno, 2006). Women with over hypothyroidism are at increased risk of pregnancy complications,
such as a higher rate of miscarriage, preeclampsia, placental abruption, growth restriction, and stillbirth (Casey and
Leveno, 2006).
Diagnosis
The finding of a goiter may be associated with cases of
Hashimoto’s thyroiditis or iodine deficiency. The signs
and symptoms of hypothyroidism are usually insidious and
easily confused with those of normal pregnancy including
fatigue, cold intolerance, cramping, constipation, weight
gain, hair loss, insomnia, and mental slowness. The serum
TSH level is an accurate and widely clinically available test
to diagnose hypothyroidism. If the serum TSH is elevated,
a free T4 level should be obtained. In the classic definition
of hypothyroidism, the serum TSH is elevated and the free
T4 is low. Other forms of hypothyroidism have also been
described, including subclinical hypothyroidism, which
is defined as an elevated TSH with a normal free T4, or
hypothyroxinemia defined as a normal TSH but a low free
T4; these have no clinical significance to the mother, but
may be associated with neonatal effects discussed later in
the neonatal neurologic development section.
Management of the Mother
Levothyroxine is the treatment of choice. Adults with
hypothyroidism require approximately 1.7 μg/kg of body
weight and should be initiated on full replacement (www.
thyroidguidelines.org). The goal of therapy is normalization of the TSH level; therefore the TSH is checked at
4- to 6-week increments, and the dose of levothyroxine is
adjusted by 25- to 50-μg increments.
Neonatal Neurologic Development
In humans, early epidemiologic data from iodine-deficient
areas of Switzerland suggested a link between mental retardation in the children of women with abnormal
thyroid function (Gyamfi et al, 2009). Studies performed
by Haddow et al (1999) found that in women with overt,
untreated hypothyroidism, the intelligence quotient (IQ)
points of children aged 7 to 9 years (using the Wechsler
Intelligence Scale IQ test) were 7 points lower in cases
than in controls (p = 0.005). The percentage of children
with IQ scores less than 85 was higher in the cases than in
controls (19% versus 5%; p = 0.007; Haddow et al, 1999).
It has been demonstrated that early transplacental passage of thyroid hormone is important for normal neurodeveloment of the fetus. T3 is made from the conversion
of maternal T4. If maternal T4 levels are low, fetal T3 in
the brain will be low even if the maternal and fetal serum
T3 are normal (Morreale de Escobar et al, 2004). Studies involving rats—which, like humans, are dependent on
maternal thyroid hormone early in development—have
demonstrated that thyroid hormone receptor is present in the brain before neural tube closure, suggesting
a biologic role (Morreale de Escobar et al, 2004). Furthermore in humans, thyroid hormone concentrations
in the cerebral cortex at 20 weeks’ gestation are comparable to those found in adults (Morreale de Escobar et al,
2004). Lavado-Autric et al (2003) have demonstrated that
in iodine-deficient rat pups, there is aberrant neuronal
migration, blurring of the cytoarchitecture, and abnormal
morphology in the somatosensory cortex and hippocampus. Early in human development there is expression of
nuclear thyroid receptors, which are already occupied by
T3, suggesting that normal maternal T4 levels are necessary for normal cortical development (Morreale de Escobar et al, 2004).
Given the increased risk of adverse perinatal and neurodevelopmental outcomes, all women with overt hypothyroidism should be treated in pregnancy. However,
controversy still exists as to whether subclinical hypothyroidism (defined as an elevated TSH level, but normal
free T4) or hypothyroxinemia (defined as a normal TSH
level, but a low free T4) warrant treatment in pregnancy.
In 1969, Man and Jones were the first to evaluate offspring of mothers with hypothyroxinemia and found
that they had lower IQ scores than normal controls or of
those born to mothers with adequately treated hypothyroidism (Gyamfi et al, 2009). Studies performed by Pop
et al (1999) in the iodine-deficient areas of the Netherlands have shown that free T4 levels below the 10th percentile at 12 weeks’ gestation were associated with lower
scores on the Dutch version of the Bayley Scale of Infant
Development at 10 months old. The study included
women with a low free T4 (hypothyroxinemia) and
excluded women with elevated TSH. A follow-up study
on these same infants, tested in both motor and mental
scores at 1 and 2 years of age, found significantly lower
scores in infants born to mothers with low free T4 levels
(Pop et al, 2003). Casey et al (2005) performed a study on
CHAPTER 9 Endocrine Disorders in Pregnancy
Parkland Hospital patients with subclinical hypothyroidism defined as a TSH at 97.5th percentile or higher and
a normal free T4 level. Approximately 2.3% of women
screened (404 women) were identified as having subclinical hypothyroidism; compared with normal controls, they
had a higher incidence of placental abruption (RR, 3.0;
95% CI, 1.1 to 8.2) and preterm birth before 34 weeks’
gestation (RR, 1.8; 95% CI, 1.1 to 2.9) (Casey et al, 2005).
The authors concluded that the reduction in IQ in children born to women with subclinical hypothyroidism may
be caused by prematurity. Based on the available animal
and clinical data, the American Association of Clinical
Endocrinologists, the American Thyroid Association, and
the Endocrine Society recommend universal screening for
all pregnant women. However, the American College of
Obstetricians and Gynecologist (2001) recommend that
screening should be performed only in women who have
risk factors, such as pregestational diabetes, or who are
symptomatic. Universal screening is not recommended
by the American College of Obstetricians and Gynecologist, given that decision and cost effectiveness studies on the effects of such a strategy are currently lacking.
Furthermore, data are lacking regarding therapy dosing,
efficacy, or whether medication should be stopped after
pregnancy in otherwise asymptomatic women with subclinical hypothyroidism and hypothyroxinemia. A multicenter randomized trial is currently underway to examine
whether screening and treatment of hypothyroxinemia
or subclinical hypothyroidism have a long-term effect on
neurodeveloment of offspring (http://clinicaltrials.gov;
study identifier number NCT00388297).
91
SUGGESTED READINGS
Alam M, Raza SJ, Sherali AR, et al: Neonatal complications in infants born to
diabetic mothers, J Coll Physicians Surg Pak 16:212-215, 2006.
American College of Obstetricians and Gynecologists: ACOG Practice Bulletin.
Clinical management guidelines for obstetrician-gynecologists: gestational
diabetes, Obstet Gynecol 98:525-538, 2001.
American Diabetes Association: Expert Committee on the Diagnosis and
Classification of Diabetes Mellitus: Report of the Expert Committee on the
Diagnosis and Classification of Diabetes Mellitus, Diabetes Care 25(Suppl 1):
S5-S20, 2002.
Casey BM, Dashe JS, Wells CE, et al: Subclinical hypothyroidism and pregnancy
outcomes, Obstet Gynecol 105:239-245, 2005.
Casey BM, Leveno KJ: Thyroid disease in pregnancy, Obstet Gynecol 108:12831292, 2006.
Coustan DR: Pharmacological Management of Gestational Diabetes: an overview,
Diabetes Care 30:S206-S208, 2007.
Study Cooperative Research Group HAPO, Metzger BE, Lowe LP, et al:
Hyperglycemia and adverse pregnancy outcomes, N Engl J Med 358:19912002, 2008.
HAPO Study Cooperative Research Group: Hyperglycemia and adverse pregnancy
outcomes: associations with neonatal anthropometrics, Diabetes 58:453-459, 2009.
Landon MB, Thom E, Spong CY, et al: A multicenter, randomized trial of treatment for mild gestational diabetes, N Engl J Med 361:1339-1348, 2009.
Langer O, Conway D, Berkus M, et al: A comparison of glyburide and insulin in
women with gestational diabetes mellitus, N Engl J Med 343:1134-1138, 2000.
Morreale de Escobar, Obregon MJ, Escobar del Rey F: Role of thyroid hormone
during early brain development, Eur J Endocrinol 151(Suppl 3):U25-U37, 2004.
Pop VJ, Browthers EP, Vader HL, et al: Maternal hypothyroxinaemia during early
pregnancy and subsequent child development: a 3-year follow-up study, Clin
Endocrinol (Oxf) 59:282-288, 2003.
Ray JG, O’Brien TE, Chan WS: Preconception care and the risk of congenital
anomalies in the offspring of women with diabetes mellitus:a meta-analysis,
QJM 94:435-444, 2001.
Rowan JA, Hague WM, Gao W, et al: Metformin versus insulin for the treatment
of gestational diabetes, N Engl J Med 358:2003-2015, 2008.
Temple R, Aldridge V, Greenwood R, et al: Association between outcome of pregnancy and glycaemic control in early pregnancy in type 1 diabetes: population
based study, BMJ 325:1275-1276, 2002.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
10
Maternal Medical Disorders of Fetal
Significance: Seizure Disorders,
Isoimmunization, Cancer, and Mental
Health Disorders
Thomas F. Kelly and Thomas R. Moore
A significant spectrum of maternal medical disorders may
complicate pregnancy. Some of these disorders, although
readily manageable in nonpregnant patients, can be lethal
to pregnant women. As a result, two questions arise for
the specialist caring for a pregnant woman with a medical complication. First, is the condition affected by the
patient’s normal adaptations to pregnancy? Second, how
does the medical problem affect the woman and her fetus?
Although many medical conditions during pregnancy can
be managed much like they would be in a nonpregnant
woman, there are usually nuances in care during gestation
to which the obstetrician must be attuned and that will
potentially affect the fetus and neonate.
During pregnancy, any potential medical therapy should
be considered carefully to minimize fetal risk. For example, thalidomide and diethylstilbestrol were prescribed in
the past for morning sickness and recurrent miscarriages,
respectively, on the basis of the reasonable hypotheses that
maternal sedation would decrease nausea and increased
estrogens would support the placenta and reduce the
likelihood of first-trimester loss. Unfortunately, the use
of both of these agents led to significant and tragic congenital anomalies in the offspring. In view of the profusion
of new drugs available today, the importance of assessing
the expected risk-to-benefit ratio for each medication prescribed to a pregnant woman is increasingly important.
An example is phenytoin treatment for maternal seizures.
If there is a less teratogenic alternative that is effective,
it should be prescribed; however, for most women taking
phenytoin, other agents are unable to control their seizures, and the risks to the fetus must be accepted.
The altered pharmacokinetics of many drugs during
pregnancy must be considered, because the dosing of
familiar medications may have to be adjusted if toxicity
is to be avoided. Classic examples are thyroid hormone
replacement (a higher level of thyroid-binding globulin in
pregnancy increases total thyroxin, but leaves the serum
free thyroxin value unchanged) and aminoglycoside antibiotic therapy (increased glomerular filtration in pregnancy
results in lower serum drug levels).
This chapter discusses four maternal conditions that can
influence fetal growth, development, and outcome: seizure
disorders, red blood cell isoimmunization, cancer, and
mental disorders. The basics of management, the potential
effects of pregnancy on the condition, and the effects of
the condition on the mother and fetus are considered.
92
MATERNAL SEIZURE DISORDERS
Epilepsy is the most common major neurologic disorder in
pregnancy. Approximately 18 million women are affected
worldwide, and 40% of those are of childbearing age. The
estimated prevalence in pregnancy is 0.2% to 0.7% (Chen
et al, 2009). The pattern of maternal seizures ranges from
complex partial to generalized tonic clonic (grand mal) and
generalized absence (petit mal) seizures. Physiologically,
seizures arise from paroxysmal episodes of abnormal brain
electrical discharges; when associated with motor activity,
they are termed convulsive.
The effect of pregnancy on the frequency and severity of the seizure disorder has been difficult to ascertain
because of limited prospective data. The International
Registry of Antiepileptic Drugs and Pregnancy (EURAP)
recently reported on more than 1800 patients whose seizure frequency and treatment were recorded. Fifty-eight
percent of patients had no seizures during their pregnancy. When using first-trimester seizure activity as a
reference, 64% had no change in frequency in the second
and third trimester, 6% improved, and 12% deteriorated
(EURAP Study Group, 2006). The only exception was
that tonic-clonic seizures occurred more frequently in
women using oxcarbazepine monotherapy; this has been
confirmed by an Australian registry in which seizures
occurred in 50% of pregnant women with epilepsy who
were receiving therapy. However, in a subset that had no
seizures for 12 months before pregnancy, there was a 50%
to 70% reduced frequency during gestation (Vajda et al,
2008). In patients in whom higher numbers of seizures
occur during gestation, decreased plasma concentrations
of antiepileptic medications have been hypothesized as
causative. The fall in plasma drug levels during pregnancy may be due in part to increased protein binding,
reduced absorption, and increased drug clearance. The
adequacy of prepregnancy seizure control can influence a
patient’s course during gestation. Patients whose seizures
were poorly controlled tended to have more frequent seizures during pregnancy, whereas patient who had no seizures for 2 years before pregnancy had only a 10% chance
of experiencing seizures during gestation. These latter
patients may be candidates for stopping therapy or considering monotherapy if they have previously required
multiple antiepileptic drugs (Schmidt et al, 1983; Walker
et al, 2009).
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
PERINATAL RISK
For reasons that are not clear, women with seizures have
more obstetric complications during pregnancy and a
higher rate of poor perinatal outcomes. Rates of preeclampsia, preterm delivery, small-for-gestational-age
infants, congenital malformations, cerebral palsy, and
perinatal mortality have all been reported as higher in
women with an antecedent seizure disorder (Lin et al,
2009; Nelson and Ellenberg, 1982). Pregnancy outcome
is also greatly influenced by the mother’s socioeconomic
status and age as well as by the prenatal care received.
Earlier publications suggested an increased risk of
congenital malformations in children of mothers with
epilepsy even without prenatal use of antiepileptic drugs
(Bjerkedal, 1982). More recent data appear to refute this,
and the malformation risk apparently correlates with the
number of medications used. A study comparing patients
with epilepsy to matched controls revealed that women
receiving no medication had no increased rate of congenital malformations; however, monotherapy was associated
with and increased risk of embryopathy (odds ratio of 2.8).
Furthermore the frequency was even higher with use of
two or more drugs (odds ratio of 4.2) (Homes et al, 2001).
Recent updates from five international registries have
reported malformation rates ranging from 3.7% to 8.0%
(with monotherapy) and 6% to 9.8% (with polytherapy)
(Meador et al, 2008). Specific malformations include a
fivefold rise in the rate of orofacial clefts (Friis et al, 1986),
an increase in the rate of congenital heart disease, particularly with trimethadione (Friis and Hauge, 1985), and a
3.8% incidence of neural tube defects in fetuses exposed
to valproic acid (Samrén et al, 1997). Facial abnormalities
(e.g., midface hypoplasia) are not specific to any particular
antiepileptic drug; they have been seen with phenytoin,
carbamazepine, and trimethadione. Some antiepileptic
medications can adversely affect postnatal cognitive development. Although conclusive data are lacking, there may
be an increased adverse effect, particularly with valproate
(Meador et al, 2008; Tomson and Battino, 2009).
93
fetal complications. Preconception counseling is preferable and should entail (1) adjusting medication doses into
the therapeutic range, (2) attempting to limit the patient
to one drug if possible, and (3) choosing an agent with the
least risk of teratogenesis. Frank discussion of the various
risks of each agent should be conducted, particularly the
risks associated with valproic acid and trimethadione. Usually if the patient’s disease is adequately controlled with
one agent, it rarely needs to be changed, because the risks
of increasing seizure activity are believed to outweigh the
potential for reducing congenital malformations.
Patients taking antiepileptic medications should also take
folic acid supplements (800 to 1000 μg) before conception,
because inhibition of folate absorption has been proposed
as a teratogenic mechanism, particularly with phenytoin.
During gestation, the anticonvulsant levels should be
checked monthly, and the dose should be adjusted accordingly, particularly with the use of lamotrigine, carbamazepine, and phenytoin (Harden et al, 2009). Although the
evidence is less clear with other agents such as phenobarbital, valproate, primidone, and ethosuximide, serial level
assessment should not be discouraged. Medications should
not be changed unless they prove ineffective at the optimal serum level. If a patient reports greater seizure activity, the serum drug level should be checked immediately.
A common reason for increased seizures is that the patient
is not taking her medication, usually because she fears its
teratogenicity.
Mothers taking phenytoin, phenobarbital, or primidone
may have a higher incidence of neonatal coagulopathy as
a result of vitamin K–dependent clotting factor deficiency.
Although maternal vitamin K supplementation in the third
trimester may be reasonable, there is insufficient evidence
to determine whether it will reduce neonatal hemorrhagic
complications (Harden et al, 2009).
RED BLOOD CELL ISOIMMUNIZATION
The classic features of the fetal hydantoin syndrome are
facial clefting, a broad nasal ridge, hypertelorism, epicanthal
folds, distal phalangeal hypoplasia, and growth and mental
deficiencies; however, these effects also result from the use
of other antiseizure medications (Table 10-1). The postulated cause of this syndrome is the teratogenic action of a
common epoxide intermediate of these medications. The
hydantoin syndrome was found to develop in fetuses with
inadequate epoxide hydrolase activity (Buehler et al, 1990).
This enzymatic deficiency appears to be recessively inherited.
It appears that preconception folic acid supplementation can
reduce the risk of major congenital malformations in women
taking antiepileptic medication (Harden et al, 2009).
Hydrops fetalis, a condition associated with abnormal fluid
collections in various body cavities of the fetus, was first
described in 1892. The causes are many but can be divided
into two categories: immune causes and nonimmune
causes. In immune-mediated hydrops, circulating immunoglobulins lead to the destruction of fetal red blood cells
and hemolytic anemia. Landsteiner and Weiner first elucidated the Rhesus factor (Rh) in 1940. Levine et al, showed
pathogenesis of erythroblastosis to be due to maternal
isoimmunization in 1941. Rh immune globulin was developed in the middle 1960s. Prophylaxis protocols using Rh
immune globulin significantly reduced the incidence of D
isoimmunization, increasing the relative frequency of alloimmunization against atypical red blood cell antigens such
as E, Duffy, and Kell (Figure 10-1). The pathophysiology
of immune-mediated fetal hemolytic disease is similar,
regardless of the blood group antigen involved; therefore
this discussion focuses on the Rh system.
MANAGEMENT
GENETICS
Management of the pregnant patient with epilepsy is based
on keeping her free of seizures. Theoretically, this goal
reduces maternal physical risk and lowers the incidence of
The Rh blood group actually represents a number of
antigens, designated D, Cc, and Ee. The genes for these
antigens, located on the short arm of chromosome 1, are
FETAL HYDANTOIN SYNDROME
94
PART III
Maternal Health Affecting Neonatal Outcome
inherited in a set of three from each parent. The presence
of D determines whether the individual is Rh positive, and
the absence of D (there is no recessive allele, so “d” does
not exist) yields the Rh-negative type. Rarely, a patient
exhibits a Du variant and should be considered D positive.
The combinations of these various antigens occur with different frequencies. For example, prevalence of Cde (41%)
is higher than that of CDE (0.08%) (Lewis et al, 1971).
Although the Rh phenotype is the result of D status, the
various genotype combinations help to predict the zygosity
of an individual. Approximately 45% of Rh-positive individuals are homozygous and therefore will always produce
an Rh-positive offspring; 55% are heterozygous and may
have an Rh-negative child if paired with an Rh-negative
partner.
TABLE 10-1 Clinical Features of the Fetal Hydantoin Syndrome
Craniofacial abnormalities
Broad nasal ridge
Wide fontanel
Low-set hairline
Broad alveolar ridge
Metopic ridging
Short neck
Ocular hypertelorism
Microcephaly
Cleft lip with or without palate
Abnormal or low-set ears
Epicanthal folds
Ptosis of eyelids
Coloboma
Coarse scalp hair
Limb abnormalities
Smallness or absence of nails
Hypoplasia of distal phalanges
Altered palmar crease
Digital thumb
Dislocated hip
Data from Briggs GC, Freeman RK, Yaffe SJ: Drugs in pregnancy and lactation, ed 7,
Baltimore, 2005, Lippincott Williams & Wilkins.
50
45
40
1969
1996
35
There are no sex differences in the frequency of Rh
negativity; however, racial variations are striking. Rh
negativity is common in the Basque population (30% to
35%), but rare in Chinese, Japanese, and North American
Indian populations (1% to 2%). The average incidence in
white populations is approximately 15%. North American blacks have a higher incidence (8%) than African
blacks (4%).
PATHOGENESIS
Before modern blood banking, Rh-negative patients
became immunized from the transfusion of Rh-positive
blood. In the case of atypical red blood cell antigens (e.g.,
Duffy, Kidd), blood transfusion is still a significant cause
of isoimmunization. Currently, fetal transplacental hemorrhage is the primary cause of Rh isoimmunization. Rh
immune globulin prophylaxis protocols have reduced but
not eliminated this problem. Transplacental hemorrhage
of fetal cells into the maternal circulation is surprisingly
common, with 75% of women showing evidence of this
event at some time during gestation (Bowman et al, 1986).
Using sensitive Kleihauer-Betke testing, 0.01 mL or more
fetal cells are found in 3%, 12% and 46% of women in
the first, second and third trimesters, respectively. The
amount of fetal blood is usually small, but approximately
1% of women have 5 mL. In 0.25% of women, 30 mL
or more of fetal cells are noted after delivery. Obstetric
events increase the chance of transplacental hemorrhage
(Box 10-1). As little as 0.3 mL of Rh-positive blood produces immunization, and the risk is dose dependent. ABO
blood group incompatibility between fetus and mother
affords some protection, reducing the risk from 16%
to 2%.
The primary maternal immune response is slow and
can take as long as 6 months to develop. The first appearance of immunoglobulin (Ig) M class anti-D antibodies is
weak; they do not cross the placenta, but are soon followed
by smaller IgG antibodies that are capable of traversing
the placental barrier. Therefore the initial event causing
sensitization rarely results in fetal hemolysis. A second
transplacental hemorrhage leads to the more rapid and
abundant amnestic IgG response that, in the presence of
fetal D-positive cells, can cause significant hemolysis and
fetal anemia.
The severity of hemolytic disease is related to the
maternal antibody titer, the affinity for the red blood
Percent
30
25
20
15
10
5
0
RhD
E
C
Kell
Duffy MNS
Kidd
FIGURE 10-1 Differences in incidence of maternal red cell antibodies over time. (Adapted from Queenan JG, Smith BD, Haber JM, et al:
Irregular antibodies in the obstetric patient, Obstet Gynecol 34:767-770,
1969; and Geifman-Holtzman O, Wojtowycz M, Kosmas E, et al: Female
alloimmunization with antibodies known to cause hemolytic disease, Obstet
Gynecol 89:272-275, 1997.)
BOX 10-1 O
bstetric Events Associated With
Increased Risk of Fetal-Maternal
Hemorrhage
ll
ll
ll
ll
ll
ll
ll
ll
ll
Unexplained vaginal bleeding
Abruptio placentae
External breech version
Amniocentesis
Chorionic villus sampling, placental biopsy
Umbilical cord sampling
Manual removal of the placenta
Abdominal trauma
Ectopic pregnancy
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
cell membrane, and the ability of the fetus to compensate for the red blood cell destruction. Table 10-2 summarizes the severity levels of fetal and neonatal disease
and their incidence. Most cases are mild and result in
normal outcomes; the cord blood is strongly Coombs
positive, but the infants do not exhibit significant anemia
or hyperbilirubinemia. Moderate disease results from
the red blood cell destruction and the greater production of indirect bilirubin. Although the mother is able
to clear this product for the fetus in utero, the neonate
is deficient in the liver glucuronyl transferase enzyme,
leading to the buildup of this water-insoluble molecule.
Albumin carries the indirect bilirubin, but if the binding
capacity is exceeded, diffusion of the bilirubin into the
fatty tissues occurs. Neural tissue is high in lipid content.
Ultimate destruction of neurons can occur, resulting in
kernicterus. Treatment depends on the recognition of
the hyperbilirubinemia and usually entails phototherapy
and possible exchange transfusion in the nursery (see
Chapter 79).
Severe disease occurs when the fetus is unable to produce sufficient red blood cells to compensate for the
increased destruction of these cells. Extramedullary hematopoiesis, which is prominent in the liver, ultimately leads
to enlargement, hepatocellular damage, and portal hypertension. This process is believed to be the etiology of placental edema and ascites. Albumin production diminishes
because of hepatocellular damage and results in anasarca,
giving rise to hydrops fetalis. The theory that hydrops is
due to fetal heart failure no longer holds, as a result of
observations that these infants are neither hypervolemic
nor in failure. The relationship between fetal anemia and
hydrops is variable, but most hydropic fetuses have hemoglobin levels less than 4 g/dL or have a hemoglobin concentration deficit greater than 7 g/dL (Nicolaides et al,
1988).
MANAGEMENT
Management of the sensitized patient, for both Rh and
atypical red blood cell antigens, requires an understanding
of the mechanism of disease and the skill and experience to
TABLE 10-2 Classification of the Severity of Hemolytic Disease
Severity
Description
Mild
Indirect bilirubin result, 16-20
mg/dL
No anemia
No treatment needed
45-50
Fetal hydrops does not develop
Moderate anemia
Severe jaundice with risk of
kernicterus unless treated
after birth
25-30
Fetal hydrops develops in utero
Before 34 weeks’ gestation
After 34 weeks’ gestation
20-25
10-12
10-12
Moderate
Severe
predict its severity. Although management schemes follow
some basic guidelines, successful management requires
access to a blood bank with expertise in antibody typing
and individuals skilled in prenatal diagnostic procedures
(e.g., cordocentesis). Referral to experienced high-risk
centers for the management of this problem is common in
the United States.
MONITORING
At their first prenatal visit, all pregnant patients should
undergo a blood type and antibody screen (indirect
Coombs’ test), which identifies the Rh-negative woman
and screens for the presence of anti-D antibody and other
immunoglobulins that are associated with atypical red
blood cell antigens and capable of causing fetal hemolytic disease (Table 10-3). Any positive result on antibody
screening should be evaluated aggressively to identify the
antibody and quantify its amount by titer. A consultation
with a blood bank pathologist may be necessary for atypical antibodies.
The amount of the anti-D antibody present according
to indirect antibody titer is important. For a titer that is
less than 16 and remains at that level throughout the pregnancy, most centers consider the fetus to be at negligible
risk of hydrops or stillbirth. Each blood bank sets different
standards for this critical titer, which depend on the assay
used. At a titer of 16, the risk is 10%; at 128, it is 75%.
For a woman with a previously affected fetus, no titer is
predictive; therefore management based on its result may
underestimate the severity of fetal disease.
Once the patient has been identified as isoimmunized,
obtaining her obstetric history is important. All of her prior
pregnancies and their outcomes must be documented to
attempt to elucidate the timing and cause of the sensitization and to assess the risk in the current pregnancy. In general, the condition is worse with each pregnancy. In a first
sensitized pregnancy, the risk of hydrops is approximately
10%. More than 90% of patients who have delivered one
hydropic baby will deliver another one subsequently.
Paternal blood typing and Rh genotyping should be performed to calculate the fetal risk of Rh positivity. Given
TABLE 10-3 Examples of Atypical Red Blood Cell Antigens
Associated With Fetal Hemolytic Disease
Incidence (%)
Data from Bowman JM: Maternal blood group immunization. In Creasy R, Resnik R,
editors: Maternal-fetal medicine: principles and practice, Philadelphia, 1984, WB Saunders.
95
Blood Group
System
Antigen
Severity of Hemolytic
Disease
Kell
K
Mild to severe
Duffy
Fya
Mild to severe
Kidd
JKa
JKb
Mild to severe
Mild to severe
MNSs
M
N
S
s
U
Mild to severe
Mild
Mild to severe
Mild to severe
Mild to severe
P
PP
Mild to severe
Public antigens
Yta
Moderate to severe
PART III
Maternal Health Affecting Neonatal Outcome
the high percentage of heterozygosity in Rh-positive
individuals, assays utilizing free DNA in the maternal
circulation are used to determine a potentially D+ fetus.
Accuracy with this technique can approach 97% (Geifman-Holtzman et al, 2006). For atypical blood group
immunization, a history of prior obstetric outcomes plus
transfusions (a significant cause of such antibodies) and
determination of the father’s antigen status are of similar
importance. For example, a woman with an anti-Kell antibody titer of 128 would be at moderate risk of hydrops,
unless the father of the baby were found to be Kell negative. No invasive procedures for isoimmunization should
be performed until the father’s antibody status is established, unless the fetus’s paternity is in question or the
partner is unavailable.
Ultrasonographic screening to identify the prehydropic fetus is notoriously unreliable, in that significantly anemic fetuses may not be grossly hydropic.
However, clues that have been proposed to suggest
impending hydrops are polyhydramnios, skin thickening, early ascites (particularly around the fetal bladder),
and placental thickening. Ultrasonographic measurements of the liver and spleen have been suggested as aids
in predicting anemia in nonhydropic fetuses, but lack
sensitivity and specificity (Bahado-Singh et al, 1998;
Vintzileos et al, 1986).
Antibody titers should be followed monthly to predict which fetus is at risk (in the absence of a history
of a prior infant with hydrops). If the indirect antibody
titer value is less than 16, the development of hydrops
is unlikely. Most centers consider the critical value to
be 16, because above this level, one cannot ensure the
absence of hydrops. When a patient’s antibody titer
value is equal to or greater than 16, more invasive testing is needed. Such testing is usually accomplished with
amniocentesis and, less commonly, through direct fetal
blood sampling.
Amniocentesis is the standard screening technique
for fetal anemia, because amniotic fluid contains hemolytic products excreted from the fetal kidneys and lungs,
including bilirubin. The unconjugated form of bilirubin is
secreted across the respiratory tree and therefore into the
amniotic fluid. The supernatant is analyzed with a spectrophotometer, and the bilirubin peak corresponding to
the level of absorbance (or optical density [OD]) of these
hemoglobin products (450 nm) is quantified. The magnitude of the peak is calculated after subtracting the mean
baseline value surrounding the peak. The difference from
baseline to peak is the ∆OD450 value.
The amount of the increase in the ∆OD450 value correlates reasonably well with severity of hemolysis. In 1961,
Liley developed a graph that has been used to predict the
severity of fetal hemolytic disease using the ∆OD450 value.
The graph is divided into three zones. Zone I represents
the lowest risk and indicates an unaffected fetus, whereas
zone III strongly suggests a severely affected fetus, in
which fetal hydrops and death can ensue if disease is not
treated within the next 7 to 10 days. The Liley curve is
authoritative in the third trimester, but its reliability before
26 weeks’ gestation has been questioned (Queenan et al,
1993). At less than 20 weeks’ gestation, the ∆OD450 value
must be greater than 0.15 or less than 0.09 to be predictive
of severe or mild disease, respectively, with a “gray zone”
between the two values that is nonpredictive (Ananth and
Queenan, 1989). Thus, the clinician must integrate clinical
history and ultrasonography clues for possible impending
hydrops or consider fetal blood sampling in a fetus at risk
between 18 and 25 weeks’ gestation.
Amniocytes can be obtained during the amniocentesis
procedure. Through polymerase chain reaction analysis,
fetal D antigen typing can be obtained as early as 14 weeks’
gestation (Dildy et al, 1996). This process allows the
identification of the fetus that is not at risk for hemolytic
disease caused by maternal antibody isoimmunization. Isoimmunization with atypical red blood cell antigens other
than D can be detected similarly, reducing the amount of
unnecessary invasive procedures on an otherwise unaffected fetus.
A recent advance in ultrasound technology has dramatically altered the assessment of the potentially
affected fetus. Doppler blood flow measurements in the
umbilical vein (Iskarios et al, 1998) and in the middle
cerebral artery have shown to be altered in anemia, with
the latter revealing an elevated peak systolic velocity.
Using a developed reference range with a cutoff of 1.5
multiples of the median, Mari (2000), was able to predict which nonhydropic fetuses had moderate or severe
anemia (Figure 10-2). A prospective multicenter trial
compared the use of middle cerebral artery Doppler
against the “gold standard” amniotic fluid ∆OD450. One
hundred sixty-five fetuses were studied, and almost half
were found to have severe fetal anemia at cordocentesis. Middle cerebral artery Doppler was noted to have a
sensitivity of 88%, a specificity of 85%, and an accuracy
of 85%. Doppler outperformed ∆OD450 analysis using
the Liley curve, with a 9% improvement in the accuracy. Furthermore, it has been suggested that 50% of
invasive procedures could have been averted with the
use of Doppler (Oepkes et al, 2006). Direct ultrasoundguided fetal umbilical blood sampling provides valuable
120
110
MCA peak systolic velocity
96
100
1.5 MoM
90
80
70
Median
60
50
40
30
20
10
15 17 19 21 23 25 27 29 31 33 35 37 39
Gestational age (wks)
FIGURE 10-2 Peak velocity of systolic blood flow in the middle
cerebral artery (MCA) with advancing gestation. The bottom curve
indicates the median peak systolic velocity in the middle cerebral artery,
and the top line indicates 1.5 multiples of the median (MoM; the threshold for significant fetal anemia). (Adapted from Mari G, Hanif F: Fetal
Doppler: umbilical artery, middle cerebral artery, and venous system, Semin
Perinatol 32:254, 2008.)
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
data for the fetus at risk, particularly after 18 weeks’ gestation. Cordocentesis is used most often when the risk
determined from history, indirect antibody titer values,
Liley curve comparisons, or ultrasonographic clues are
significant. Cordocentesis also provides vascular access
if fetal transfusion becomes necessary. Although most
obstetricians are skilled in amniocentesis, cordocentesis is usually performed in tertiary centers by perinatologists. The latter procedure carries a higher risk of
fetal loss and morbidity, and it is associated with a more
significant chance of worsening maternal sensitization
(Bowman and Pollock, 1994).
THERAPY
Fetal transfusion therapy is the mainstay of treatment for
the severely affected but premature fetus. In most centers, for pregnancies beyond 33 weeks’ gestation with
suspected fetal anemia, administration of steroids and
delivery are preferable to an invasive procedure with significant morbidity and mortality. Intraperitoneal transfusions, the primary therapy in the past, are still a useful
treatment. O-negative, tightly packed, irradiated blood is
infused percutaneously into the fetal peritoneal cavity via
an amniocentesis needle with real-time ultrasound guidance. Approximately 10 mL is transfused for each week
of gestation beyond 20 weeks. Red blood cell absorption
occurs promptly through the subdiaphragmatic lymphatics, although it may be erratic in the hydropic fetus. Overall
survival rates with fetuses requiring transfusion approaches
89% (Van Kamp et al, 2005). However, hydrops was associated with a decreased survival rate of 78% (Van Kamp
et al, 2001), with severely hydropic fetuses having a survival rate of 55%. Problems associated with intraperitoneal
transfusions include injury to vascular or intraabdominal
organs and the inability to obtain fetal blood type and
blood count. This procedure should be avoided if possible
in the hydropic fetus, because the resulting higher abdominal pressure can precipitate venous compression and lead
to circulatory collapse.
Direct intravascular transfusions are currently the
first line of treatment. The advantages of this procedure
include assessing the severity of fetal anemia and documenting the fetal blood type. Ultimate success does not
appear to be affected by the presence of hydrops (Ney
et al, 1991). The overall success rate is approximately
85%. Limitations are usually related to the procedure.
The most accessible site usually requires an anterior placenta, in which the cord root can be visualized; posterior
placentation makes the technique more difficult. In addition, risks include bleeding from the cord puncture site,
fetal exsanguination, cord hematoma, rupture of membranes, and chorioamnionitis.
Additional transfusions are required every 2 to 3 weeks
to compensate for the falling hematocrit associated with
fetal growth, the finite life of the transfused red blood cells,
and ongoing hemolysis of the existing fetal erythrocytes.
Ultimately the entire fetal blood supply is replaced with
Rh-negative blood by successive transfusions. A major
neonatal side effect is bone marrow depression, which may
require postnatal transfusions for 1 to 3 months (De Boer
et al, 2008).
97
As the pregnancy proceeds through the third trimester, frequent fetal testing is performed either with nonstress tests and amniotic fluid index or with biophysical
profiles. Delivery is planned for 3 to 6 weeks before term,
usually after demonstration of a mature fetal lung profile.
Preterm delivery may be indicated in a severely affected
fetus, regardless of lung profile, if the risk of transfusion
is deemed to exceed the morbidity of a delivery of a nearterm yet premature baby.
CANCER
PRINCIPLES
Cancer complicating pregnancy is rare, with an estimated frequency of 1 case per 1000 live births. The trend
of delaying childbearing to later maternal age may have
influenced this rate. The sites or types of cancer in pregnancy, in descending order of frequency, are cervical,
breast, ovarian, lymphoma, melanoma, brain, and leukemia (Table 10-4) (Haas et al, 1984; Jacob and Stringer,
1990). Finding a malignancy during gestation poses a
unique set of issues that must be addressed with care.
Will the pregnancy accelerate the malignant process? Are
the accepted therapies appropriate for the mother, and
are they safe for the unborn fetus? Will delay of therapy
adversely affect the mother? Should the pregnancy be terminated, or should the child be delivered prematurely to
maximize treatment of the mother with no resultant risk
to the child?
Few conditions in pregnancy require as meticulous a
multidisciplinary approach as cancer. Oncologists who
are unaccustomed to interacting with a pregnant woman
commonly wish to have the child delivered before giving
definitive cancer therapy, for fear of the teratogenic risks
to the fetus. Therefore input regarding the situation must
be acquired not only from an oncologist but also from the
obstetrician, perinatologist, pediatrician, neonatologist,
and dysmorphologist. The patient and her family must
be involved in decision making, being given information
not only about the risks of the disease and its potential
therapies, but also about the limitations of current knowledge about cancer in pregnancy and the uncertainties of
outcomes.
TABLE 10-4 Cancers that Can Complicate Pregnancy
Site/Type
Incidence (per number of gestations)
Cervix
1:2000 to 1:10,000
Breast
1:3000 to 1:10,000
Melanoma
1:1000 to 1:10,000
Ovary
1:10,000 to 1:100,000
Colorectal
1:13,000
Leukemia
1:75,000 to 1:100,000
Lymphoma
1:1000 to 1:6000
Data from Pentheroudakis G, Pavlidis N, Castiglione M: Cancer, fertility and
pregnancy: ESMO clinical recommendations for diagnosis, treatment and followup,
Ann Oncol 20(Suppl 4):S178-S181, 2009.
98
PART III
Maternal Health Affecting Neonatal Outcome
SURGERY
Patients can undergo indicated surgery during pregnancy safely. The risk of fetal loss does not seem to rise
with uncomplicated anesthesia and surgery. However, if
there are any complications from either the anesthesia
or the operation, the risk of fetal and maternal mortality
increases. For example, fetal loss is rarely related temporally to maternal appendectomy, but is common when the
mother’s appendix has ruptured. The most comprehensive series of pregnant patients undergoing surgery was
collected in Sweden (Mazze and Kallen, 1989). Although
these researchers found a slight increase in the rates of low
birthweight and neonatal death by 7 days of life, the rates
of stillbirth and congenital malformation were similar to
the outcomes expected without surgery. Anesthetic agents
are not believed to be teratogenic.
A few management guidelines help to optimize outcome
for the pregnant patient undergoing surgery. If general
anesthesia is used, the maternal airway must be protected
to avoid aspiration. Gastrointestinal motility is reduced
during pregnancy, and the stomach may contain significant residual contents after many hours without eating.
A left lateral decubitus position on the operating table is
preferred to maximize uteroplacental blood flow. If the
fetus is viable, monitoring of the fetal heart rate should be
performed to assist the anesthesia team in optimizing fetal
status. Preoperative counseling with the patient is important to allow the surgical team to make appropriate choices
in regard to any interventions for fetal distress.
CHEMOTHERAPY
Administering agents that impair cell division in pregnancy
is a concern for both the mother and the care team. Often
the patient is more concerned about this issue than about
the underlying cancer. Cytotoxic chemotherapy should be
avoided in the first trimester because of the high incidence
of spontaneous abortion and the potential teratogenic
effects on the fetus (Table 10-5). For a few agents with
confirmed teratogenic effects (e.g., methotrexate, amethopterin, chlorambucil), chemotherapy must be avoided
during organogenesis. The use of folic acid antagonists in
first-trimester cancer treatment raises the specific problem
of possible induction of neural tube defects, because these
lesions are known to be folate sensitive.
The literature regarding most other chemotherapeutic
agents is limited, consisting of a few collected series; therefore these agents should be used cautiously, with their
potential harm to the fetus balanced against their benefit
to the maternal condition. Little is also known regarding
the long-term outcomes of fetuses exposed to chemotherapeutic agents in utero. The National Cancer Institute in
Bethesda, Maryland, maintains a registry in the hopes of
determining the delayed effects. A small series of fetuses
exposed to chemotherapeutic agents for acute leukemia
revealed normal mental development with follow-up
between 4 and 22 years (Aviles and Niz, 1988).
The risk of teratogenicity does not appear to be higher
with combination chemotherapy than with single-agent
therapy (Doll et al, 1989). Studies performed thus far
involve small numbers of patients, with power insufficient
to show a statistic difference, but there seems not to be
a trend. Low birthweight is found in approximately 40%
of babies whose mothers received cytotoxic drugs during
pregnancy (Nicholson, 1968). Theoretical consequences,
such as bone marrow suppression, immune suppression,
and anemia, could occur in the fetus. As a result, the timing
of chemotherapy should account for the anticipated date
of delivery. Data regarding safety for breastfeeding the
neonate of a mother receiving cancer chemotherapy are
limited. For this reason, the majority of agents are contraindicated in nursing mothers.
TABLE 10-5 Common Chemotherapy Agents and Uses
Class or Drug
Risk Category
Common Uses
Alkylating Agents
Busulfan
Dm
Leukemias
Chlorambucil
Dm
Lymphomas,
leukemias
Cyclophosphamide
Dm
Breast, ovary,
lymphomas,
leukemias
Melphalan
Dm
Ovary, leukemia,
myeloma
Procarbazine
Dm
Lymphomas
5-Fluorouracil
D*
Breast,
gastrointestinal
6-Mercaptopurine
Dm
Leukemias
Methotrexate
Xm
Trophoblastic disease, lymphomas,
leukemias, breast
6-Thioguanine
Dm
Leukemias
Bleomycin
Dm
Cervix, lymphomas
Daunorubicin
Dm
Leukemias
Doxorubicin
Dm
Leukemias,
lymphomas, breast
All-trans-retinoic
acid
X
Leukemias
l-Asparaginase
Cm
Leukemias
Cisplatin
Dm
Ovary, cervix,
sarcoma
Hydroxyurea
D
Leukemias
Prednisone
C*
Lymphomas,
leukemias, breast
Tamoxifen
Dm
Breast, uterus
Paclitaxel
Dm
Breast, ovarian
Vinblastine
Dm
Breast, lymphomas,
choriocarcinoma
Vincristine
Dm
Leukemias,
lymphomas
Antimetabolites
Antibiotics
Other Agents
Data from Neoplastic diseases. In Cunningham FG, MacDonald PC, Gant NF, et al,
editors: Williams obstetrics, ed 20, Stamford, Conn., 1997, Appleton and Lange; and
Briggs GC, Freeman RK, Yaffe SJ: Drugs in pregnancy and lactation, ed 7, Philadelphia,
2005, Lippincott Williams & Wilkins.
*Risk factor D if used in the first trimester.
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
RADIATION THERAPY
The deleterious effects of irradiation on the fetus have
been both theorized and actual. Fortunately, the amount
of concern about the former far exceeds the incidence
of the latter. Irradiation promotes genetic damage and
thus the potential for congenital malformations. The risk
depends on both dose and time (Table 10-6). A dose of less
than 5 rad is believed to be of little consequence (Brent
et al, 1989). If radiation exposure occurs before implantation, the adverse outcomes are usually a small increase in
miscarriage.
The major concern is high-dose radiation (>10 rad)
received during the period of organogenesis (embryonic
weeks 1 to 10). The central nervous system is the most
radiation-sensitive organ, and the complications most
often observed are microcephaly and mental retardation.
Cataracts and retinal degeneration are also seen. After
organogenesis is complete, there is still a risk of central
nervous system abnormalities. However, the sequelae
most often seen are skin changes and anemia. Because of
the highly variable yet potentially grave consequences of
irradiation greater than 10 rad, patients should be counseled accordingly, and termination of pregnancy should be
offered as an alternative if exposure has occurred in the
previable period (Orr and Shingleton, 1983).
There are several other considerations for a pregnant
woman undergoing radiation therapy. First, the dose used
in estimating risk should be the amount that the fetus
actually receives. For example, axillary or neck irradiation for lymphoma involves a lower direct fetal exposure
than direct pelvic irradiation for cervical cancer. The latter
treatment, if given in the second trimester, will likely cause
fetal demise. Second, the magnitude of radiation scatter
to the pelvis must be considered. External shielding does
not prevent internal reflection of the ion beam. Third, the
advancing size of the uterus actually increases the amount
of radiation exposure of the fetus, because of the closer
proximity of the nonpelvic irradiation. Therefore an
8-week-old fetus may actually receive a smaller radiation
dose from supraclavicular irradiation than a 30-week-old
fetus. Fourth, will the fetus concentrate the radiation, and
therefore increase its dose? This is exemplified by the use
of radioactive iodine (131I) for maternal thyroid conditions.
TABLE 10-6 Radiation Dose Thresholds for Deleterious Fetal
Effects
Weeks Since LMP
Fetal Dose
(mGy)
Potential Fetal
Effects
2-4 (preimplantation)
>50-100
Spontaneous abortion,
but generally not
malformation
The actual rad dose is markedly higher in the fetus, because
the fetal thyroid concentrates the iodine.
Diagnostic tests such as radiography may also be associated with radiation exposure for the fetus (Table 10-7). The
doses involved are usually much smaller than those used
for cancer therapy. Nonetheless, the practitioner should
limit the amount of radiographic testing if at all possible.
Inadvertent imaging of a patient who is not known to be
pregnant continues to occur despite sensitive pregnancy
tests, thus creating significant concerns. Indicated radiographs should never be withheld because of pregnancy, but
lead shielding of the patient’s abdomen and careful selection of the type of study should be performed to minimize
the pelvic dose. Usually the amount of fetal exposure is
much less than 5 rad, with no significantly greater risk of
malformations. There does appear to be a slightly higher
incidence of childhood cancer if the fetus is exposed to
doses on the order of 10 mGy (Doll and Wakeford, 1997).
CERVICAL CANCER
Cervical cancer is the most common malignancy found in
pregnancy. The incidence is approximately 1 in 2500 gestations. A Papanicolaou test smear should be performed for
all patients at their first prenatal visit. However, approximately 30% of cervical cancers can be associated with negative cytologic smear results. Although the evaluation for
TABLE 10-7 Estimated Radiation Dose to the Fetus With
Common Diagnostic Imaging Procedures
Test
Fetal Dose (rad)
Radiograph
Upper extremity
<0.001
Lower extremity
<0.001
Upper gastrointestinal series (barium)
0.048-0.360
Cholecystography
0.005-0.060
Lumbar spine
0.346-0.620
Pelvis
0.040-0.238
Hip and femur series
0.051-0.370
Chest (two views)
<0.010
Retropyelography
0.800
Abdomen (kidneys, ureter, bladder
0.200-0.245
Urography (intravenous pyelography)
0.358-1.398
Barium enema
0.700-3.986
Computed Tomography Scan
Head
<0.050
Chest
0.100-0.450
Abdomen (10 slices)
0.240-2.600
Abdomen and pelvis
0.640
2-8 (organogenesis)
>200
Malformations
Pelvis
0.730
8-15
100-1000
Severe mental
retardation
Lumbar spine
3.500
>15
>1000
Mental retardation
Adapted from Doyle S, Messiou C, Rutherford JM, et al: Cancer presenting during
pregnancy: radiological perspectives, Clin Radiol 64:857-871, 2009.
LMP, Last menstrual period.
99
Other
Ventilation perfusion scan
0.06-1.00
From Bentur Y: Ionizing and nonionizing radiation in pregnancy. In: Koren G:
Medication safety in pregnancy and breastfeeding, Philadelphia, 2007, McGraw Hill.
100
PART III
Maternal Health Affecting Neonatal Outcome
an abnormal Papanicolaou test result should not be altered
because of pregnancy, many physicians are reluctant for
fear of cervical hemorrhage. Cervical biopsy remains the
mainstay of diagnosis. The greater vascularity of the cervix during pregnancy predisposes bleeding. An experienced colposcopist may be able to defer actual biopsy in
cases of possible visual findings of a noninvasive process.
However, if cancerous invasion is suspected or if the physician is uncertain of the visual findings, biopsy is necessary. If microinvasive disease is confirmed by biopsy, cone
biopsy is required to rule out frankly invasive disease. This
procedure is undertaken with caution during pregnancy,
because of the associated high rate of bleeding complications and miscarriage. Cervical conization may raise the
risk of incompetence or preterm labor. The assistance of
a gynecologic oncologist is preferred, given these unique
sets of potential consequences. A shallow cone biopsy will
reduce the risk of subsequent cervical weakness.
The therapy for invasive cervical cancer is based on
the stage of disease and the gestational age of the fetus.
Therapy can involve external beam radiation, internal
radiotherapy (brachytherapy), or surgery (Table 10-8). In
most cases, delay of definitive therapy by 4 to 14 weeks
may be acceptable. Pregnancy does not seem to accelerate the growth of the tumor. However, patient counseling is important. In the extremely previable gestation, the
likelihood of achieving a safe gestational age for the fetus
without worsening the stage or spread of the cancer in the
mother must be balanced against parental desires based on
ethical or religious beliefs. Conversely, it might be reasonable to delay definitive therapy until a time when delivery
would not likely result in a long-term disability because of
extreme prematurity.
BREAST CANCER
Breast cancer is the most common malignancy of women,
with approximately 1 in 8 women affected in their lifetimes
(Goldman and O’Hair, 2009). The incidence of breast cancer in pregnancy is estimated to be 10 to 30 per 100,000
pregnancies (Isaacs, 1995). Pregnancy does not seem to
influence the actual course of the disease; however, there
appears to be a higher risk of delay in diagnosis and a trend
toward more advanced stages at diagnosis in pregnant
women than in nonpregnant controls.
The diagnostic procedures for breast cancer should not
be altered during pregnancy. Any suspicious mass should
undergo biopsy. Mammography, although discouraged
for routine screening in pregnancy, can be used safely
if indicated. The amount of radiation is negligible—
approximately 0.01 cGy (Liberman et al, 1994). Mammography imaging may be hampered by physiologic changes
because of pregnancy, and ultrasound examination may
be a useful alternative. Metastatic evaluation may be hampered somewhat because of a reluctance to use bone and
liver scans during pregnancy. Magnetic resonance imaging
can be used safely in the second and third trimesters.
Surgical therapy for breast cancer should not be delayed
because of pregnancy. The risks of mastectomy and axillary node dissection appear to be low (Isaacs, 1995). Radiation therapy is usually not recommended during pregnancy
because of the risk of beam scatter to the pregnant uterus.
If the pregnancy is to continue and the patient has evidence of tumor invasion in the lymph nodes, adjuvant chemotherapy is often given. The timing of delivery should
account for the following factors:
ll When would the fetus have a reasonable chance for survival with a low risk of severe permanent morbidity?
ll Can the number of cycles of chemotherapy be minimized with an earlier delivery? In addition, avoiding delivery just before or just after administration of
chemotherapy is important to reduce the risk of immunosuppression and infection.
ll How long can radiotherapy be delayed without increasing the risk of metastatic spread of the tumor?
Approximately 10% of women treated for breast cancer
become pregnant, the majority within 5 years of diagnosis. Data from small series suggest that pregnancy does
not influence the rate of recurrences or of distal metastasis
(Dow et al, 1994). However, women should be encouraged
to delay childbearing for at least 2 to 3 years, which is the
time of the highest rate of recurrence. Breastfeeding may
be possible in women who have undergone conservative
breast cancer surgery.
TABLE 10-8 Treatment Options for Cervical Cancer in Pregnancy
Gestational Age
(weeks)
Stage I to IIa
Stage IIb to IIIb
<20
4500 cGy
Wide pelvic irradiation
If no spontaneous abortion: modified radical hysterectomy
If spontaneous abortion: brachytherapy
or radical hysterectomy with lymphadenectomy
5000 cGy
Wide pelvic irradiation
If no spontaneous abortion: type II radical
hysterectomy
If spontaneous abortion: brachytherapy or
cesarean section at fetal viability
Subsequent wide pelvic radiation (±5000 cGy)
and brachytherapy (±5000 cGy)
>20
Cesarean section at fetal viability
Subsequent wide pelvic radiation (5000 cGy) and brachytherapy
(5000 cGy) or cesarean radical hysterectomy with lymphadenectomy
Cesarean section at fetal viability
Subsequent wide pelvic radiation (±5000 cGy)
and brachytherapy (±5000 cGy)
Data from Berman ML, Di Saia PJ, Brewster WR: Pelvic malignancies, gestational trophoblastic neoplasia and nonpelvic malignancies. In Creasy RK, Resnik R, editors: Maternalfetal medicine, ed 4, Philadelphia, 1999, WB Saunders.
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
OVARIAN CANCER
Most ovarian cancer occurs in women older than 35
years. Delayed childbearing has been more widely
accepted, exemplified by British birth rates doubling in
women older than 30 years and tripling in women older
than 40 years since 1975 (Palmer et al, 2009), in addition to a twofold increased birth rate among U.S. women
older than 40 years since 1981 (Martin et al, 2009). It
would not be surprising for the rate of ovarian and other
cancers during pregnancy to increase. However, the current estimate of actual ovarian malignancies in pregnancy
is low and estimated to range from 1 in 10,000 to 1 in
50,000 deliveries (Jacob and Stringer, 1990; Palmer et
al, 2009). Whereas most ovarian cancers are epithelial in
origin, borderline epithelial and germ cell tumors (dysgerminomas and malignant teratomas) are more common
in pregnancy.
The widespread use of ultrasonography, particularly
in the first two trimesters, has been helpful in identifying
adnexal masses. Fortunately, most are benign functional
cysts. Actual malignancy is rare and is estimated at 5%
of the ovarian masses found. The risk is higher in nonpregnant females, approaching 15% to 20%. Surgery for
a suspected ovarian mass occurs in approximately 1 per
1000 pregnancies. Most procedures are performed not
for suspected malignancy, but because of concern about
torsion and rupture. The incidence of adnexal torsion
ranges from 1% to 50%, and there appears to be a trend
with increasing rates in masses greater than 6 cm (Yen
et al, 2009). The maximal times of risk of these events
are at the end of the first trimester, when the uterus elevates beyond the true pelvis, and at the time of delivery.
The characterization of an ovarian process can be aided
by ultrasonography or magnetic resonance imaging, but
these modalities are not definitive. Ultrasound scoring
systems that use size and character poorly predict malignancy, but have a better negative predictive value (Lerner
et al, 1994). Although an ovarian cyst, particularly if it is
simple in nature, is likely not malignant, the patient must
be cautioned that histologic diagnosis is more definitive.
Indications for surgical exploration include a complex
mass, a persistent simple cyst 8 cm or larger, or one that
is symptomatic (Leiserowitz, 2006). The optimal time for
laparotomy is in the second trimester. At that time, there
is minimal interference from the gravid uterus and less of
a risk of fetal loss, and the theoretical concerns of teratogenic exposure to anesthetic agents are avoided. Some
patients opt for more conservative management; they
should be counseled that they have a 40% chance of needing urgent intervention with either surgery or percutaneous drainage (Platek, 1995).
If a malignancy is confirmed at the time of laparotomy, treatment and staging are no different than for a
nonpregnant woman. Frozen-section diagnosis, peritoneal washings, omentectomy, and subdiaphragmatic
biopsy are performed. Depending on the cell type and
the stage, treatment can range from removal of the
affected adnexa to complete hysterectomy and bilateral oophorectomy. Chemotherapy may be given during pregnancy if necessary. Fortunately, most epithelial
ovarian cancers found in pregnant women are usually of
101
a lower stage, with 59% of reported cases being stage I
(Palmer et al, 2009).
SURVIVORS OF CHILDHOOD CANCER
Given the improvements of therapy for childhood cancer, a large number of these individuals have survived
into adulthood. Some are unable to conceive because of
high-dose radiation or cytotoxic chemotherapy. The risk
of decreased fertility for patients exposed to pelvic radiation therapy may as high as 32% (Geogeseu et al, 2008).
Those who remain fertile may have concerns regarding
whether their treatment increases the risk of adverse pregnancy outcomes. Although data are limited, female cancer
survivors treated with radiation therapy appear to have
increased risks of premature delivery, low birthweight,
and miscarriage. There is no evidence that female partners
of male cancer survivors treated with radiation have these
excess risks (Reulen et al, 2009).
MENTAL DISORDERS
Pregnancy can be a stressful process. At times, it may
induce a psychotic event. Women experiencing a mental disorder during pregnancy who have no history of a
mood disorder usually exhibit a milder constellation of
symptoms. Serious disorders such as mania and schizophrenia that are antecedent to pregnancy may not be so
benign. In women with all types of mental illness, and in
previously nonaffected women, the postpartum state is
a time of greater maternal risk. Ten percent to 15% of
new mothers experience a depressive disorder (Weissman and Olfson, 1995). Furthermore, there appears to
be an increased perinatal risk with mental disorders and
pregnancy (Table 10-9). Women with preexisting mental
illness have a higher recurrence risk in the puerperium.
Patients with suspected mental illness should be assessed
for substance abuse and thyroid dysfunction. A multidisciplinary approach is advantageous. If the patient’s mental
competency is an issue, the caregiver should obtain legal
assistance to be able to make medical decisions for the
patient.
DEPRESSION
Depression ranks as the fourth leading cause of disability
worldwide, and recognized prevalence appears to be
increasing (Dossett, 2008). A study by Dietz et al (2007)
suggests that the prevalence of depression during pregnancy or postpartum, as defined by onset within 3 to 6
months after delivery, to be approximately 10%.
The predisposing risk factors for depression include
early childhood loss, physical or sexual abuse, socioeconomic deprivation, genetic predisposition, and lifestyle
stress caused by multiple roles (McGrath et al, 1990).
These factors can exaggerate or prolong symptoms and,
if not addressed, can lengthen the duration of depression. The obstetrician must be aware that life events
such as miscarriage, infertility, and complicated pregnancy in patients with risk factors are likely to precipitate
depression; therefore there is a low threshold for diagnosis and treatment of mood alterations in such patients.
102
PART III
Maternal Health Affecting Neonatal Outcome
TABLE 10-9 Impact of Psychiatric Illness on Pregnancy Outcome
Impact on Outcome
Illness
Teratogenic Effects
Obstetric
Neonatal
Treatment Options
Anxiety disorders
N/A
Increased incidence of forceps deliveries, prolonged
labor, precipitate labor, fetal
distress, preterm delivery, and
spontaneous abortion
Decreased developmental
scores and inadaptability;
slowed mental development at 2 years of age
Benzodiazepines
Antidepressants
Psychotherapy
Major depression
N/A
Increased incidence of low
birthweight, decreased
fetal growth, and postnatal
complication
Increased newborn cortisol
and catecholamine levels,
infant crying, rates of
admission to neonatal
intensive care units
Antidepressants
Psychotherapy
ECT
Bipolar disorder
N/A
Increased incidence of low
birthweight, decreased
fetal growth, and postnatal
complication
Increased newborn cortisol
and catecholamine levels,
infant crying, rates of
admission to neonatal
intensive care units
Lithium
Anticonvulsants
Antipsychotics
ECT
Schizophrenia
Congenital malformations, especially
cardiovascular
Increased incidence of preterm
delivery, low birthweight,
small for gestational age,
placental abnormalities and
placental hemorrhage
Increased rates of postnatal
death
Antipsychotics
From ACOG Practice Bulletin: Clinical management guidelines for obstetrician-gynecologists number 92, April 2008 (replaces practice bulletin number 87, November 2007): use of
psychiatric medications during pregnancy and lactation, Obstet Gynecol 111:1001-1020, 2008.
ECT, Electroconvulsive therapy.
Alternatively, perinatal loss experienced by a woman
without predisposing risk factors will probably lead to a
grief reaction or adjustment disorder, which may be misdiagnosed as depression.
Chronic medical conditions that are associated with a
high prevalence of depression and may occur in women
of childbearing age include renal failure, cancer, AIDS,
and chronic fatigue or pain. Antihypertensives, hormones,
anticonvulsants, steroids, chemotherapeutics, and antibiotics can cause depression. Alcoholism and substance
abuse may manifest as depression. Underlying personality
disorders complicate the diagnosis of depression by confusing the clinical situation, in addition to contributing
to the secondary effect of many physicians’ avoidance of
patients suffering from such disorders.
Therapeutic interventions for depression include psychotherapy and medication. Electroconvulsive therapy
has been shown to be an effective and relatively safe treatment in refractory cases (Rabheru, 2001). However, there
remains controversy and some degree of concern because
of published series and case reports suggesting a risk of
fetal cardiac arrhythmias, vaginal bleeding, and premature uterine contractions (Bhatia et al, 1999); therefore it
should be reserved for refractory cases and performed in
a setting with immediate access to obstetric care (Pinette
et al, 2007; Richards, 2007). Treatment of depression
is effective in approximately 70% of cases. Supportive
treatment alone is rarely effective in major depressive
episodes. Most antidepressant medications currently prescribed during pregnancy are selective serotonin reuptake
inhibitors (SSRIs). SSRIs have an advantage over the tricyclic antidepressants by not causing orthostatic hypotension. Unfortunately, and although limited series suggest
that the SSRIs are relatively safe, little is known about
long-term consequences for children exposed to SSRIs in
utero (Altshuler et al, 1996; Chambers et al, 1996; Karasu
et al, 2000). Fluoxetine is the best-studied SSRI in terms of
safety. Alternatively, paroxetine has been associated with
an increased risk of congenital heart defects (Kallen et al,
2007). Although data remain inconsistent, they suggest
avoiding paroxetine as a first-line agent. However, if a certain agent is controlling the patient’s symptoms, it would
seem reasonable not to change medications for the sake of
these concerns.
A recent issue has been raised regarding the use of SSRIs
and persistent pulmonary hypertension of the newborn;
however, the incidence remains low at approximately 10
in 1000 fetuses exposed after 20 weeks’ gestation (Chambers et al, 2006). Currently, there is no consensus regarding the use of SSRIs during breastfeeding. Fluoxetine has
an active metabolite with a long half-life and is found in
higher concentrations in infants (Eberhard-Gran et al,
2006). Short-term neonatal effects have been reported,
including increased crying, decreased sleep, and irritability, particularly with fluoxetine and citalopram. Reasonable guidelines regarding the use of SSRIs and other
psychotropic medications are listed in Table 10-10. The
long-term side effects are currently listed as “unknown”
(Briggs et al, 2005; Dodd et al, 2000). The theoretical concerns are that such drugs may affect the developing central nervous system of the newborn and that abnormalities
may not be readily apparent in the short term. Therefore
SSRIs should be prescribed for a nursing mother only if
the benefit clearly exceeds the risk, and after the patient
has been counseled regarding the potential yet currently
ill-defined risks. Given current medical knowledge, bottle
feeding should be offered as an acceptable alternative if
antidepressants must be used.
CHAPTER 10 Maternal Medical Disorders of Fetal Significance: Seizure Disorders, Isoimmunization, Cancer, and Mental Health Disorders
103
TABLE 10-10 Summary of Current Knowledge of Drug Excretion into Breast Milk, Drug Concentrations in Infant Serum, Adverse Effects
in the Child, and Breastfeeding Recommendations for Different Psychotropic Drugs
Class or Drug
Drug Transfer
into Breast Milk
Infant Plasma
Concentrations
Adverse Effects in the
Child
Breastfeeding
ecommendations
R
SSRIs
Low transfer
Low plasma concentrations
Case reports of adverse
effects in infants exposed
to fluoxetine and citalopram
Compatible with
breastfeeding; however,
fluoxetine and citalopram
may not be drugs of first
choice
TCAs
Low transfer
Low plasma concentrations
(except doxepin)
No suspected immediate
adverse effects observed
(except doxepin)
Compatible with breastfeeding; however, doxepin should be avoided
Other antidepressants
Limited data
Limited data
Limited data
None able to be made
Benzodiazepines
Low transfer
High plasma concentrations
with longer acting drugs
with active metabolites
Case reports of CNS
depression reported for
diazepam
Sporadic use of short-acting
benzodiazepines unlikely
to cause adverse effects
Lithium
Low transfer
Dose received by the infant
is high
Limited data; some reports
of toxicity in the infant
Limited data; however,
breastfeeding should be
avoided
Carbamazepine, sodium
valproate
Low transfer
Low plasma concentrations
Some case reports of various
adverse effects in the
infant
Generally more compatible
with breastfeeding than
lithium
Lamotrigine
High transfer
High plasma concentrations
Limited data
None able to be made
Novel antipsychotics
Moderate transfer
Variable plasma
concentrations
Limited data
None able to be made
Modified from Eberhard-Gran M, Esklid A, Opjordsmoen S: Use of psychotropic medications in treating mood disorders during lactation: practical recommendations, CNS Drugs
20:187-198, 2006.
CNS, Central nervous system; SSRI, selective serotonin reuptake inhibitor; TCA, tricyclic antidepressants.
POSTPARTUM PSYCHOSIS
A severe disorder, postpartum psychosis is fortunately rare,
occurring in 1 to 4 per 1000 births (Weissman and Olfson,
1995). This condition is more worrisome than postpartum
depression, because of the patient’s inability to discern
reality from the periods of delirium. Patients at risk for
postpartum psychosis may have underlying depression,
mania, or schizophrenia. Other risks are younger age and
family history. The recurrence rate is approximately 25%.
The peak onset of symptoms is between 10 and 14 days
after delivery. Recognition of this disorder is extremely
important to the protection of the patient and her family.
SCHIZOPHRENIA
The prevalence of schizophrenia is approximately 1% in
the general population (Myers et al, 1984); it is associated
with delusions, hallucinations, and incoherence. Morbidity due to this mental illness is higher than that due to any
other. There appears to be a genetic component to the
etiology; schizophrenia develops in approximately 10% of
offspring of an affected person. Concordance of schizophrenia in identical twins reaches 65%. There is some
speculation and controversy as to whether low birthweight
(Smith et al, 2001) and obstetric complications (Kendell
et al, 2000) are associated with a higher rate of
schizophrenia.
Because the peak age of incidence is approximately 20
years and women are affected more often than men, it is
unrealistic to assume that obstetricians will never encounter patients with schizophrenia. There appear to be higher
rates of cesarean section and surgical vaginal delivery in
affected patients (Bennedsen et al, 2001b). Children of
women with schizophrenia may have a higher rate of sudden infant death syndrome and congenital malformations
(Bennedsen et al, 2001a). However, it is difficult to ascertain whether these risks are independent of other factors
such as smoking, poor socioeconomic status, and use of
certain medications.
Treatment is achieved primarily through the use of
psychotropic medication. The potential for teratogenesis appears low with the older-generation medications in
the phenothiazine class, but most data for this issue were
derived from the use of lower doses given to patients with
hyperemesis gravidarum. Antipsychotic medication does
cross the placenta. Current recommendations include
avoiding use in the first trimester if possible, the use of
lower doses or higher-potency alternatives, and cessation
of medication 5 to 10 days before delivery (Herz et al,
2000). The use of most antipsychotics in breastfeeding is
associated with an unknown risk (Briggs et al, 2005).
Lithium, used primarily in mania, is associated with a
higher rate of Ebstein anomaly. Although the incidence of
this consequence is low, either discontinuing the medication in the first trimester or continuing its use with careful
counseling is a viable alternative. Fetal echocardiography
should be performed in women who have used lithium in
early pregnancy.
104
PART III
Maternal Health Affecting Neonatal Outcome
SUGGESTED READINGS
ACOG Committee on Practice Bulletin–Obstetrics: ACOG Practice Bulletin:
Clinical management guidelines for obstetrician-gynecologists, number 92,
April 2008. Use of psychiatric medications during pregnancy and lactation,
Obstet Gynecol 111:1001-1020, 2008.
Bent RL: Saving lives and changing family histories: appropriate counseling of
pregnant women and men and women of reproductive age, concerning the risk
of diagnostic radiation exposures during and before pregnancy, Am J Obstet
Gynecol 200:4-24, 2009.
Cohn D, Ramaswamy B, Blum K, et al: Malignancy in pregnancy. In Creasy RK,
Resnik R, Iams JD, editors: Maternal-fetal medicine, ed 6, Philadelphia, 2009,
WB Saunders, pp 885-904.
Moise KJ: Management of rhesus alloimmunization in pregnancy, Obstet Gynecol
112:164-176, 2008.
Walker S, Permezal M, Berkovic S: The management of epilepsy in pregnancy,
BJOG 116:758-767, 2009.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
11
Hypertensive Complications
of Pregnancy
Andrew D. Hull and Thomas R. Moore
Hypertension is the most common medical problem in
pregnancy, affecting 10% to 15% of all pregnant women.
As the third most common cause of maternal mortality
after thromboembolic disease and hemorrhage, hypertension accounts for almost 16% of maternal deaths in the
United States (Berg et al, 2003). Complications arising
from hypertensive disorders have profound effects on the
fetus and neonate and thus are a major source of perinatal
mortality and morbidity. Preeclampsia is also the primary
cause of iatrogenic prematurity.
CLASSIFICATION OF HYPERTENSIVE
DISORDERS OF PREGNANCY
Any discussion of hypertension and pregnancy must begin
with a set of definitions. Although many classifications are
in use worldwide, perhaps one of the more useful comes
from the Report of the National High Blood Pressure
Education Program Working Group on High Blood Pressure in Pregnancy (2000) (Table 11-1). Although this classification scheme appears to be somewhat pedantic, it is of
paramount importance because pregnancy outcome varies
according to the type of hypertension involved. For practical purposes, hypertension in pregnancy can be divided
into the following categories: chronic hypertension, gestational hypertension, and preeclampsia.
Hypertension is defined as a systolic blood pressure of
140 mm Hg or higher or a diastolic pressure of 90 mm
Hg or higher, measured on two separate occasions. Korotkoff phase V (disappearance of sound) is used rather than
Korotkoff phase IV (muffling of sound) to define diastolic
pressure, because Korotkoff IV is poorly reproducible in
pregnancy. The term severe hypertension identifies a population at significantly increased risk for stroke and cardiac
decompensation, and it is defined as a systolic blood pressure of 160 mm Hg or a diastolic pressure of 110 mm Hg
or higher.
CHRONIC HYPERTENSION
Up to 5% of pregnant women have chronic hypertension,
which is diagnosed when hypertension is present before
pregnancy or recorded before 20 weeks’ gestation. However, when hypertension is first noted in a patient after 20
weeks’ gestation, it may be difficult to distinguish chronic
hypertension from pregnancy-induced hypertension or
preeclampsia. In such cases, the precise diagnosis might
not be made until after delivery. Hypertension that is first
diagnosed during the second half of pregnancy and persists
more than 12 weeks postpartum is diagnosed as chronic
hypertension.
Chronic hypertension has an adverse effect on pregnancy
outcome. Women with the disorder are at higher risk for
preterm delivery and placental abruption, and their fetuses
are at risk for intrauterine growth restriction (IUGR) and
demise (Ferrer et al, 2000). Superimposed preeclampsia
complicates up to 50% of pregnancies in women with preexisting severe chronic hypertension (Sibai and Anderson,
1986), and it occurs before 34 weeks’ gestation in 50% of
cases (Chappell et al, 2008). The adverse effects on fetal
and maternal perinatal outcomes are directly related to the
severity of the preexisting hypertension. When chronic
hypertension is secondary to maternal renal disease, the
risks of poor outcome are further increased, with as much
as a tenfold rise in fetal loss rate (Jungers et al, 1997).
Women with untreated severe chronic hypertension are
at increased risk for cardiovascular complications during pregnancy, including stroke (Brown and Whitworth,
1999).
The majority of cases of chronic hypertension seen
in pregnancy are idiopathic (essential hypertension),
but causes of secondary hypertension should always be
sought because pregnancy outcome is worse in women
with secondary hypertension. Renal disease (e.g., chronic
renal failure, glomerulonephritis, renal artery stenosis),
cardiovascular causes (coarctation of the aorta, Takayasu
arteritis), and, rarely, Cushing disease, Conn syndrome,
and pheochromocytoma should be excluded through
physical examination, history, and more detailed testing
if needed.
All patients with chronic hypertension should be evaluated periodically with serum urea, creatinine, and electrolyte measurements, urinalysis, and 24-hour urine
collection for protein and creatinine clearance determinations. Typically this assessment should be performed in
each trimester and more frequently if the patient’s condition deteriorates.
ANTIHYPERTENSIVE TREATMENT OF
CHRONIC HYPERTENSION IN PREGNANCY
Except in cases of severe hypertension, randomized trials have shown that antihypertensive treatment of chronic
hypertension in pregnancy does not improve fetal outcome (Sibai and Anderson, 1986). Rates of preterm delivery, abruption, IUGR, and perinatal death are similar in
treated and untreated women. Therefore treatment is
usually reserved for patients whose hypertension places
them at a significant risk of stroke (systolic blood pressure
of 180 mm Hg or higher or diastolic pressure of 110 mm
Hg or higher). Patients with less severe hypertension who
were taking medications before conception might be able
to discontinue therapy with close surveillance. The risk
of superimposed preeclampsia is not changed by antihypertensive therapy, so its development should be tracked
carefully.
105
106
PART III
Maternal Health Affecting Neonatal Outcome
TABLE 11-1 Classification of Hypertensive Disorders
TABLE 11-2 Drugs Commonly Used to Treat Chronic
of Pregnancy*
Category
Definition
Chronic
hypertension
Hypertension present before pregnancy
or diagnosed before 20 weeks’ gestation, or diagnosed for the first time during pregnancy that persists postpartum
Gestational
hypertension
Transient if blood pressure returns to
normal by 12 weeks after delivery, and
preeclampsia was not diagnosed before
delivery
Chronic if blood pressure does not resolve
by 12 weeks after delivery
Preeclampsiaeclampsia
Usually occurs after 20 weeks’ gestation
Hypertension accompanied by proteinuria
in a woman normotensive before 20
weeks’ gestation
Strongly suspected if nonproteinuric
hypertension is accompanied by
systemic symptoms such as headache,
visual disturbance, abdominal pain, or
laboratory abnormalities such as low
platelet count and elevated liver enzyme
values (HELLP syndrome)
Preeclampsia
superimposed on
chronic hypertension
Preeclampsia occurring in a chronically
hypertensive woman
Adapted from the Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy, Am J Obstet Gynecol 183:S1-S22, 2000.
HELLP, Hemolysis, elevated liver enzymes, and low platelets.
*Hypertension is defined as a systolic blood pressure ≥140 mm Hg systolic or diastolic
blood pressure ≥90 mm Hg.
The choice of antihypertensive agent for use in pregnancy is governed by a desire to adjust blood pressure
without having ill effects on the fetus. Because excessive
lowering of maternal blood pressure below 140 mm Hg
systolic or 90 mm Hg diastolic (140/90 mm Hg) can compromise uterine perfusion, with consequent slowing of
fetal growth, fetal hypoxia, or both, the therapeutic goal is
to maintain maternal pressures at 140 to 155 systolic and
90 to 105 diastolic. The drugs most commonly used in
pregnancy are listed in Table 11-2.
Methyldopa, a centrally acting antihypertensive agent,
formerly was the most widely used drug in this setting.
Many obstetricians remain faithful to the use of this agent
because of extensive clinical and research experience
demonstrating its safety for both mother and fetus during pregnancy (Report of the National High Blood Pressure Education Program Working Group on High Blood
Pressure in Pregnancy, 2000). This agent does not impair
uteroplacental perfusion and has a wide therapeutic margin before side effects are seen. However, methyldopa has
the disadvantage of a rather slow onset of action with prolonged time to therapeutic effect (days), and compliance
with methyldopa therapy may be impeded by side effects
such as sedation in some patients.
Labetalol is a mixed alpha1-adrenergic and beta1- and
beta2-adrenergic blocking agent, and it is the most frequently used alternative to methyldopa. Some pure betablockers have been associated with a significant increase in
the risk of IUGR (e.g., atenolol), and the mixed adrenergic
blockade produced by labetalol is thought to mitigate this
Hypertension in Pregnancy and Their Modes
of Action
Drug
Mode of Action
Methyldopa
Centrally acting antihypertensive
Labetalol
Mixed alpha- and beta-adrenergic blocker
Nifedipine
Calcium channel blocker
Hydralazine
Peripheral vasodilator
Prazosin
Alpha-blocker
unwanted effect (Pickles et al, 1989). Labetalol is also used
intravenously to manage severe hypertension accompanying preeclampsia.
Calcium channel blockers (e.g., nifedipine) are used
mainly as second-line drugs, usually in long-acting,
extended-release forms. Calcium channel blockers appear
to be as effective as methyldopa and labetalol with minimal
fetal side effects (Levin et al, 1994).
Hydralazine, a potent peripheral vasodilator, is frequently used intravenously to treat acute hypertensive
emergencies in pregnancy (blood pressure of >160/110
mm Hg). Its role as an oral agent in the management of
chronic hypertension is limited to a second- or third-line
choice. Long-term use of hydralazine may be associated
with a lupuslike syndrome in some patients.
Prazosin, an alpha-adrenergic blocker, has been used
as a second- or third-line drug in pregnant women whose
hypertension is difficult to control or is severe with an
early onset. This agent appears to be similar in efficacy to
nifedipine in such a setting (Hall et al, 2000).
Although diuretics are used extensively in adults with
hypertension, there appears to be little role for them in the
treatment of chronic hypertension in pregnancy. Diuretics
have been alleged to reduce or prevent the normal plasma
volume expansion seen in pregnancy (Sibai et al, 1984),
an effect that theoretically might impede fetal growth,
although the evidence for this is mixed. Most authorities
restrict the use of diuretics in pregnant patients to those
with cardiac dysfunction or pulmonary edema.
Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers should not be used during pregnancy. In the second and third trimesters, these agents are
associated with malformation of the fetal calvarium, fetal
renal failure, oligohydramnios, pulmonary hypoplasia,
and fetal and neonatal death (Buttar, 1997). Angiotensinconverting enzyme inhibitors appear to be safe when
taken in the first trimester (Steffensen et al, 1998), but a
patient who conceives while taking an angiotensin receptor blocker or angiotensin-converting enzyme inhibitor
should be switched to a safer alternative as soon as possible. Similar precautions apply to the use of angiotensin
receptor blockers in pregnancy.
ANTENATAL FETAL SURVEILLANCE
IN CHRONIC HYPERTENSION
As the third trimester progresses, patients with chronic hypertension are at an increasing risk of slowing of fetal growth
and superimposed preeclampsia. Antenatal surveillance
CHAPTER 11 Hypertensive Complications of Pregnancy
in women with chronic hypertension should include careful screening for signs and symptoms of superimposed
preeclampsia, which constitutes the greatest perinatal risk.
Fetal growth should be followed with serial ultrasonography evaluations (every 3 to 6 weeks). All patients should
perform fetal movement counts from 28 weeks’ gestation
onward, and cases with suspected fetal growth impairment
should be followed with twice-weekly non-stress tests with
amniotic fluid index or weekly ultrasound biophysical profile. Although the optimum interval for these tests is controversial (every 3 to 7 days), and their role is unproven in the
absence of fetal IUGR or other evidence of fetal compromise, most centers begin regular fetal biophysical testing at
32 to 34 weeks’ gestation and continue until delivery.
If fetal growth tapers below expectations (typically sonographic estimated fetal weight [EFW] falls below the tenth
percentile or abdominal circumference much smaller percentile than head), more intensive fetal surveillance is indicated. In cases with IUGR, serial sonography should be
performed at 10- to 21-day intervals with attention paid to
amniotic fluid volume, careful profiling of each biometric
parameter, and cerebral and umbilical Doppler waveforms.
Typical indications for delivery in the setting of IUGR and
hypertension include no growth of the head and abdomen
over a 10-day interval, severe oligohydramnios, biophysical score of less than 6, or reversal of end-diastolic velocity
on the umbilical Doppler waveform. However, individualization of management in these cases is important.
Women with renal impairment and chronic hypertension have a markedly higher risk of poor perinatal outcome
than normotensive women and women with hypertension without renal impairment. In addition, moderate
or severe renal disease (serum creatinine level ≥1.4 mg/
dL) may accelerate the loss of renal function during pregnancy (Cunningham, 1990; Hou, 1999). The incidence of
impaired fetal growth is directly related to the degree of
renal impairment, and women undergoing dialysis are at
particular risk for fetal growth failure, preterm delivery,
and fetal death, even with optimal management. Those
who start dialysis during pregnancy are at the greatest risk,
with only a 50% chance of a surviving infant (Hou, 1999).
GESTATIONAL HYPERTENSION
The diagnosis of gestational hypertension can be made
with confidence only after delivery; it is defined as hypertension occurring in the second half of pregnancy in the
absence of any other signs or symptoms of preeclampsia.
Because a woman with apparent gestational hypertension
at 36 weeks’ gestation can rapidly evolve into preeclampsia
at 39 weeks’ gestation, the diagnosis of gestational hypertension should always evoke caution and vigilance. Only
if the patient’s blood pressure returns to normal postpartum without development of signs of preeclampsia during
the pregnancy should the final diagnosis of gestational
hypertension be applied. During pregnancy, gestational
hypertension is indistinguishable from preeclampsia in
evolution. Therefore all patients with gestational hypertension should be regarded as being at risk for progression
to preeclampsia.
The earlier gestational hypertension is evident, the
greater the risk of preeclampsia. When the diagnosis is made
107
before 30 weeks’ gestation, more than one third will develop
preeclampsia, whereas the risk is less than 10% when the
diagnosis is made after 38 weeks’ gestation. Decisions to
treat patients with gestational hypertension with antihypertensive agents must be carefully considered, given the risk of
concurrent preeclampsia and the lack of evidence supporting improved fetal outcome. Gestational hypertension tends
to recur in subsequent pregnancies and predisposes women
to hypertension in the future (Marin et al, 2000).
PREECLAMPSIA-ECLAMPSIA
Preeclampsia is one of the most enigmatic diseases affecting
humans. Apparently unique to humans, preeclampsia has
proved difficult to simulate in animal experiments. Despite
years of intensive research, the underlying causes of the disease are only recently becoming clearer. It is evident that
the clinical manifestations of preeclampsia arise from vascular endothelial dysfunction that ultimately may involve the
central nervous, renal, hepatic, and cardiovascular systems.
In its full-blown form, preeclampsia can produce a profound
coagulopathy and liver, respiratory, or cardiac failure.
The classic symptom triad of hypertension, proteinuria, and edema defines preeclampsia. Most classifications
of preeclampsia no longer include edema, because this
common finding affects approximately 80% of pregnant
women near term. Preeclampsia is divided into mild and
severe forms (Box 11-1). This distinction is important,
because in the presence of severe disease at any gestational
age, the only appropriate treatment option is delivery,
whereas expectant management may be acceptable in a
woman who has mild disease and is remote from term.
Although the precise etiology of preeclampsia remains
uncertain, numerous factors are associated with elevated
the risk (Table 11-3). Up to 10% of primigravid patients
have mild preeclampsia, and approximately 1% have severe
disease.
PREECLAMPSIA
Etiology
The most widely accepted theory for the pathophysiology
of preeclampsia is based on a model of impaired placental implantation that results in placental hypoperfusion
and hypoxia. The placenta then releases substances into
the maternal circulation that adversely affect endothelial
function, leading to the clinical syndrome of widespread
vascular dysfunction, which is recognized as the syndrome
of preeclampsia (Myers and Baker, 2002). Individual
responses to the process of progressive vascular dysfunction vary in severity and timing in a manner that seems to
have genetic, familial, and immunologic components. For
example, preeclampsia occurring in a first-degree relative
confers a fourfold increase in risk of the disease in siblings
and children (Chesley and Cooper, 1986). Women born
to mothers with preeclampsia have a higher risk. There
is some evidence that the presence of certain genotypes,
such as factor V Leiden and thrombophilia (de Vries
et al, 1997), or metabolic defects such as hyperhomocystinemia secondary to methylenetetrahydrofolate reductase
deficiency (Kupferminc et al, 1999), predispose women to
108
PART III
Maternal Health Affecting Neonatal Outcome
BOX 11-1 F
eatures of Mild and Severe
Preeclampsia
TABLE 11-3 Risk Factors for Development of Preeclampsia
Factor
Relative Risk
MILD
ll Systolic blood pressure ≥140 mm Hg or diastolic pressure of 90 mm Hg
ll Proteinuria ≥300 mg/24 hr
Primigravida
3
Age >40 years
3
African American race
1.5
SEVERE*
ll Systolic blood pressure ≥160 mm Hg or diastolic pressure of 100 mm Hg
ll Proteinuria ≥5 g/24 hr
ll Elevated serum creatinine value
ll Eclampsia
ll Pulmonary edema
ll Oliguria <500 mL/hr
ll HELLP syndrome
ll Intrauterine growth restriction
ll Symptoms suggestive of end-organ involvement: headache, visual disturbance, epigastric or right upper quadrant pain
Family history
5
Chronic hypertension
10
Chronic renal disease
20
Antiphospholipid syndrome
10
Insulin-dependent diabetes mellitus
2
Multiple gestation
4
Modified from ACOG practice bulletin: Diagnosis and management of preeclampsia and
eclampsia. Number 33, January 2002. American College of Obstetricians and Gynecologists,
Int J Gynaecol Obstet 77:67-75, 2002.
HELLP, Hemolysis, elevated liver enzymes, and low platelets.
*Any single feature in the severe definition satisfies criteria for the diagnosis of severe
preeclampsia.
preeclampsia, although a true candidate gene is yet to be
established and probably never will be. Population studies have suggested that women exposed to the antigenic
effects of sperm before conception have a lower rate of
preeclampsia than do women who conceive with lesser
degrees of exposure, although the evidence is inconclusive
(Koelman et al, 2000).
The endothelial dysfunction that characterizes preeclampsia (Roberts, 1999) manifests as greater vascular
reactivity to circulating vasoconstrictors such as angiotensin, reduced production of endogenous vasodilators such
as prostacyclin and nitric oxide (Ashworth et al, 1997),
increased vascular permeability, and an increased tendency
toward platelet consumption and coagulopathy. The end
result is hypertension, proteinuria secondary to glomerular injury, edema, and a tendency toward extravascular
fluid overload with intravascular hemoconcentration.
Predictors
Perhaps one of the most important contributions that
prenatal care makes to maternal and fetal outcomes is the
detection of preeclampsia and the prevention of eclampsia
(Backe and Nakling, 1993; Karbhari et al, 1972). A wide
variety of biochemical and physical tests has been proposed
as screening tools for the early detection of preeclampsia
(Dekker and Sibai, 1991). Most physical tests have been
discredited, and even the most widely used biochemical tests have poor predictive values. Uric acid levels are
elevated in many cases of preeclampsia, but the sensitivity
of the measurement is low (Lim et al, 1998). Early detection of proteinuria is possible with the use of more sensitive tests, such as gel electrophoresis (Winkler et al, 1988),
rather than conventional urinalysis, but such tests do not
lend themselves to routine use. Clinicians should be aware
of the limitations of routine urine testing for detection of
proteinuria, with standard dipstick testing being notoriously inaccurate (Bell et al, 1999).
Doppler ultrasonographic assessment of the vascular
dynamics in the uterine arteries during the second trimester has been proposed as a valuable screening tool in
populations in which obstetric ultrasonography is routine
(Kurdi, 1998). Up to 40% of women who develop preeclampsia have abnormal waveforms, and this finding was
reported to be associated with a sixfold rise in the risk of
preeclampsia (Papageorghiou et al, 2002). Other researchers have obtained less impressive results (Goffinet et al,
2001).
Recently the role of angiogenic factors in the pathophysiology of preeclampsia has been explored. Vascular
endothelial growth factor (VEGF) and placental growth
factor bind to Flt-1 and sFlt-1 receptors and have a critical role in angiogenesis and placental development. The
interactions among FLt-1, VEGF, and placental growth
factor promote angiogenesis and placental vasculogenesis, whereas those among sFLT-1, VEGF, and placental growth factor lead to the inactivation of those proteins
and disordered angiogenesis and endothelial dysfunction.
sFlt-1 levels have been found to be elevated in women with
preeclampsia, and such elevated levels of sFlt-1 precede
the features of clinical preeclampsia. Recent reviews of the
state of the art in this area (Wang et al, 2009; Widmer
et al, 2007) concluded that sFlt-1 and placental growth factor levels were significantly different after 25 weeks’ gestation between women destined to develop preeclampsia
and those with a normal pregnancy course. Measurements
of these factors earlier in pregnancy do not appear to have
the same predictive value. There is no doubt that angiogenic factors are intimately involved in the pathophysiology of preeclampsia, but alterations in their levels do not
seem to be the cause of the disease. Several large studies
are ongoing to further explore the role of these factors and
their potential use in the prediction of disease. On balance, no effective screening test to predict preeclampsia
currently exists, and clinicians are faced with the necessity
of diagnosing the disease early and managing it as adroitly
as possible.
Prevention
If an accurate predictor of preeclampsia could be identified, the next logical step would be the application of a preventive or ameliorative treatment. Unfortunately attempts
to identify an effective treatment have proved equally
CHAPTER 11 Hypertensive Complications of Pregnancy
difficult. Given the recognized association between vascular endothelial dysfunction and preeclampsia (in particular,
vasoconstriction and excessive clotting in the maternal placental arteries), prostaglandin inhibitors have been viewed
as a likely candidate for prophylaxis or treatment. Numerous trials (Duley et al, 2001) have been conducted with lowdose aspirin, based on the idea that the ability of aspirin to
irreversibly inhibit production of the vasoconstrictive prostaglandin thromboxane would promote greater activity of
prostacyclin, a vasodilatory prostaglandin. This ability of
aspirin would help to maintain patency in the maternal
placental vascular bed and limit or prevent the evolution of
preeclampsia. Unfortunately, although a modest reduction
in the frequency of preeclampsia (approximately 15%) was
documented, no improvement in key measures of perinatal
outcome was demonstrable in a metaanalysis of the results
of available studies (Duley et al, 2001).
Calcium supplementation was briefly in vogue as a preventive treatment in the 1990s, on the basis of the known
vasodilatory effect of calcium and impressive results in earlier, small studies (Atallah et al, 2000); however, its worth
was not supported in a metaanalysis (Atallah et al, 2000).
Similarly, it has been suggested that antioxidants may have
a role in preeclampsia prevention, but the only available
trial to date showed mixed results, with improvements in
biochemical indices in women receiving vitamins C and E,
although perinatal outcomes were not different in treated
and untreated groups (Chappell et al, 1999). Of concern
was the finding that women in whom preeclampsia developed despite vitamin therapy had markedly worsened preeclampsia than controls in whom the disease developed.
Therefore, at present, an ideal preventive measure for preeclampsia does not exist.
Antepartum Management
Given the current inability to predict or prevent preeclampsia, clinicians are left to address established disease
and to try to prevent maternal and fetal morbidity. The
division of established preeclampsia into mild and more
severe forms is of great worth in determining management
and minimizing morbidity (see Box 11-1). Mild disease is
generally managed conservatively (bed rest and frequent
fetal and maternal biophysical assessments) until term is
reached or there is evidence of maternal or fetal compromise. The appearance of severe preeclampsia mandates
delivery in all but highly selected cases regardless of gestational age.
Patients with a diagnosis of mild preeclampsia should be
evaluated for signs of maternal or fetal compromise, which
would make their disease severe. Evaluation should include
a 24-hour urine collection to evaluate for proteinuria; full
blood count and platelet measurements; determination
of serum uric acid, blood urea nitrogen, and creatinine
levels; and evaluation of liver transaminases. Fetal size
should be estimated with ultrasonography; the presence of
IUGR (less than the tenth percentile) is a sign of severe
preeclampsia. Patients with mild disease at 37 weeks’ gestation or more should be delivered, because prolonging
pregnancy has no material benefit and increases the risks of
maternal and fetal morbidity. Patients at earlier gestational
stages should be closely monitored with sequential clinical
109
TABLE 11-4 Drugs for Acute Treatment of Hypertension
in Severe Preeclampsia
Drug
Dosage
Hydralazine
1-2 mg test dose
5-10 mg IV followed by 5-10 mg every 20 min as
required, to a total of 30 mg
Labetalol
10-20 mg IV followed by 20-80 mg every 10 min
to a total of 300 mg
Nifedipine
10 mg PO every 10-30 min up to three doses
IV, Intravenous; PO, by mouth.
and laboratory evaluations. Such monitoring often begins
in the hospital and may be continued in an outpatient or
home setting with appropriate supervision. If the clinical
picture deteriorates or term is reached, the baby should
be delivered. There is no evidence that antihypertensive
therapy influences progression of preeclampsia, and its use
may actually be dangerous by masking worsening hypertension. Fetal well-being should be evaluated until delivery by means of kick counts and regular non-stress tests or
modified biophysical profiles.
Patients with severe disease should be delivered. The
only exception to this approach is the diagnosis of severe
preeclampsia, in a patient remote from term (<28 weeks’
gestation), on the basis of proteinuria and transiently
(unsustained) severe hypertension alone. Such patients
may be managed conservatively under close supervision
while antenatal corticosteroids are administered without
adversely affecting maternal or fetal outcome (Sibai et al,
1990). There is no reason for conservative management
in any other circumstance. The patient with severe preeclampsia at less than 24 weeks’ gestation should be
offered termination of the pregnancy; all others should be
delivered by the most expedient means. Cesarean section
should be reserved for obstetric indications.
Severe hypertension requires treatment with fast-acting
antihypertensive agents if stroke and placental abruption are
to be avoided. Intravenous hydralazine is well established as
a first-line drug for this purpose, although there is a growing experience with other agents, including intravenous
labetalol and oral nifedipine (Duley and Henderson-Smart,
2000a) (Table 11-4). The aim of treatment is to lower blood
pressure into the mild preeclampsia range (140/90 mm Hg)
to reduce the risk of stroke and other maternal cardiovascular complications. There is evidence to support the use
of parenteral magnesium sulfate to prevent eclampsia in all
cases of severe disease (Duley et al, 2003).
Severe preeclampsia can manifest as classic disease with
severe proteinuric hypertension, or it can cause atypical
findings such as pulmonary edema or severe central nervous system symptoms, including blindness. More commonly, patients show evidence of microangiopathy leading
to the hemolysis, elevated liver enzymes, and low platelets
(HELLP) syndrome. The full-blown clinical syndrome
of HELLP carries a significant maternal risk. Earlier
reports suggested that the disease carries a grave prognosis (Weinstein, 1982). This suggestion remains true for
florid clinical cases, but most patients now have “laboratory” HELLP and never experience major clinical features
110
PART III
Maternal Health Affecting Neonatal Outcome
of the syndrome because delivery is initiated before their
condition deteriorates to that point.
Preeclampsia and Fetal Risk
Because the only recourse in severe preeclampsia is delivery, the disease has a corresponding effect on prematurity
and its attendant complications. IUGR is not uncommon
in severe preeclampsia, and there may be evidence of progressive deterioration in fetal well-being with worsening
disease. Infants delivered at less than 34 weeks’ gestation
will benefit from antenatal steroid therapy—even as little
as 8 hours of therapy before delivery may have benefit.
Many patients are able to deliver vaginally, but fetal compromise may preclude aggressive induction and mandate
delivery by cesarean section. The incidence of respiratory distress syndrome is lower in infants of mothers with
preeclampsia who are delivered preterm than in those of
age-matched controls without antenatal steroid exposure
(Yoon et al, 1980). Nonetheless, the morbidity of such
infants is greater because of hypoxemic insults received
in utero. Infants born to mothers with preeclampsia may
also have thrombocytopenia or neutropenia, which further
complicates their newborn course (Fraser and Tudehope,
1996).
Intrapartum Management
All women in labor with a diagnosis of preeclampsia should
receive magnesium sulfate as seizure prophylaxis (Box
11-2). Although the absolute risk of seizure is low (1 in
2000 to 3000), the occurrence of seizures is unpredictable,
and the efficacy of magnesium sulfate and margin of safety
has been validated in multiple randomized trials (Duley
et al, 2003). The mechanism of action of MgSO4 in the
prevention of seizures is still unresolved, with various theories being advanced, including peripheral neuromuscular
blockade, membrane stabilization, N-methyl D-aspartate
(NMDA) receptor blocking activity, cerebral vasodilation,
and calcium channel blocking action (Belfort et al, 2006).
Blood pressure should be maintained in the mild preeclampsia range using intravenous antihypertensive agents
(labetalol, hydralazine). Epidural anesthesia is indicated
for pain control and to aid in blood pressure management.
Vaginal delivery should be possible in most cases. Delivery
by cesarean section should be reserved for obstetric indications. Careful attention to fluid balance should be maintained. After delivery, the preeclamptic process should
begin to resolve rapidly.
ECLAMPSIA
Eclampsia is the occurrence of generalized tonic-clonic
seizures in association with preeclampsia. It affects approximately 1 in 2500 deliveries in the United States and may
be much more common in developing countries, affecting as many as 1% of parturients. Up to 10% of maternal
deaths are due to eclampsia (Duley, 1992).
BOX 11-2 M
agnesium Sulfate Therapy
for Prevention of Eclampsia
ll
ll
ll
ll
Bolus 4-6 g IV over 20 min
Continuous infusion 1-2 g/hr
Follow up levels every 6-8 hours to target 4-6 mEq/L
Continue infusion 24 hours after delivery or 24 hours after seizure if seizure
occurs despite magnesium therapy
Most cases of eclampsia occur within 24 hours of delivery.
Almost 50% of seizures occur before the patient’s admission to the labor and delivery department, approximately
30% are intrapartum, and the remainder are postpartum.
There is a considerable drop in the risk of eclampsia by 48
hours postpartum, with seizures occurring in less than 3%
of women beyond that time. Most patients have antecedent features that are suggestive of preeclampsia, although
in some cases eclampsia may occur without warning. If
eclampsia is left untreated, repetitive seizures become
more frequent and of longer duration, and ultimately status eclampticus develops. Maternal and fetal mortality may
be as high as 50% in severe cases, especially if the seizures
occur while the patient is far from medical care.
Randomized controlled trials have demonstrated the
clear superiority of magnesium sulfate for the treatment
of eclampsia over all other anticonvulsants (Duley and
Gulmezoglu, 2002; Duley and Henderson-Smart, 2002b,
2002c). Intravenous magnesium sulfate is given as a 4-g
bolus over 5 minutes followed by a maintenance infusion of
1 to 2 g/hr for 24 hours after delivery. Subsequent seizures
can be treated with further bolus injections. In refractory
cases, second-line treatment with other anticonvulsants
may be required, or the patient may have to be paralyzed
and their lungs ventilated.
Delivery after an eclamptic seizure should take place in
a controlled, careful manner. There is little to be added by
performing an emergency cesarean section (Coppage and
Polzin, 2002). The patient’s condition should be stabilized
first. Vaginal delivery is possible in most cases, although
cesarean delivery may be indicated if the status of the cervix is unfavorable or if fetal compromise is ongoing despite
control of seizures and maternal stabilization. Infants born
to mothers after eclampsia require careful observation
after birth.
SUGGESTED READINGS
ACOG Practice Bulletin: Chronic hypertension in pregnancy, ACOG Committee
on Practice Bulletins, Obstet Gynecol 98(Suppl l):177-185, 2001.
Chappell LC, Enye S, Seed P, et al: Adverse perinatal outcomes and risk factors
for preeclampsia in women with chronic hypertension: a prospective study,
Hypertension 51:1002-1009, 2008.
Myers JE, Baker PN: Hypertensive diseases and eclampsia, Curr Opin Obstet Gynecol
14:119-125, 2002.
Wang A, Rana S, Karumanchi SA: Preeclampsia: the role of angiogenic factors in
its pathogenesis, Physiology 24:147-158, 2009.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
12
Perinatal Substance Abuse
Linda D. Wallen and Christine A. Gleason
Substance abuse during pregnancy has been recognized
as a problem for more than a century. Psychotropic substances, both legal (alcohol, cigarettes, and prescription
drugs such as opioids and benzodiazepines) and illegal
(opioids, amphetamines, cocaine, and marijuana), can
cause obstetric, fetal, and neonatal complications. These
complications include poor intrauterine growth, prematurity, abruptio placenta, fetal distress, spontaneous abortion, stillbirth, fetal (and maternal) cerebral infarctions
and other vascular accidents, malformations, and neonatal
neurobehavioral dysfunction. Although substance abuse
occurs in all socioeconomic classes, illegal drug abuse is
more frequently associated with unhealthy lifestyles, poor
access to prenatal care, untreated health problems, poverty, stress, and psychological disorders. Because of these
socioeconomic confounders as well as the confounders of
polysubstance exposure and the influence of various postnatal environmental factors, it is often difficult to determine the effects of maternal use of one specific drug on the
fetus and newborn. This chapter addresses the epidemiology of perinatal substance use and abuse; the effects of specific drugs on the fetus and newborn; maternal issues and
their effects on the newborn; identification of pregnancies
and babies at risk; neonatal management; and long-term
effects and follow-up. The discussion will focus on abused
substances that are known or suggested to be associated
with significant perinatal and neonatal morbidity: alcohol, tobacco, nicotine, opioids, cocaine, marijuana, and
methamphetamine.
EPIDEMIOLOGY OF PERINATAL
SUBSTANCE EXPOSURE
PREVALENCE
Prevalence rates for perinatal substance exposure have
been determined by using a number of different definitions, survey methods, and drug use detection procedures
(Lester et al, 2004). One of the most comprehensive geographically based prevalence studies on substance use and
abuse by pregnant women was undertaken in California in
the early 1990s by the Perinatal Substance Exposure Study
Group (Vega et al, 1993). In that study, urine was collected
at the time of delivery from more than 30,000 pregnant
women. The prevalence rates for illicit drug use were 3.5%
(overall), 8.8% (tobacco), and 7.2% (alcohol). The authors
concluded that if these results could be extrapolated to the
United States at large, an estimated 450,000 infants per
year (11% of 4 million live births) would be exposed to
alcohol, illicit drugs, or both in the days before delivery.
Rates of perinatal substance exposure have not changed
substantially over the past 20 years, although there is wide
geographic variation. The U.S. Department of Health and
Human Services Pregnancy Risk Assessment Monitoring
System is designed to monitor maternal behaviors and
experiences among women who deliver live-born infants.
Data collected during 2000 to 2003 from 19 states revealed
that during the last 3 months of pregnancy, tobacco use
ranged from 4.9% to 27.5%, and alcohol use ranged from
2% to 8.7%. Data from a 2005-2006 National Survey on
Drug Use and Health revealed that of pregnant women
aged 15 to 44, 4% reported using illicit drugs, 16.5%
reported using tobacco, and 12% reported using alcohol
sometime within the past month (Substance Abuse and
Mental Health Services Administration, 2007). These rates
are significantly lower than the rates reported by women
aged 15 to 44 years who were not pregnant (10% reported
using illicit drugs, 29.5% reported using tobacco, and 53%
reported using alcohol within the past month), confirming that the prevalence of substance use among pregnant
women remains less than among nonpregnant women.
The Maternal Lifestyle Study was developed in the
early 1990s by the National Institute of Child Health and
Human Development (NICHD) and National Institute on
Drug Abuse (NIDA) to follow pregnant women known to
be using opioids or cocaine and to follow their offspring.
The study followed 11,800 pregnant women who received
prenatal care during 1993 to 1995 at four teaching hospital sites. Women were followed from initial presentation
throughout their offspring’s childhood, with a planned
evaluation at 8 to 11 years. Meconium analyses were used to
confirm maternal substance use; there was 66% agreement
between meconium analyses and positive maternal reports.
The prevalence of cocaine and opioid exposure was 10.7%;
98% of cocaine users also used other drugs; and only 2% of
women used cocaine alone (Shankaran et al, 2007).
EPIDEMIOLOGY OF SPECIFIC SUBSTANCES
Nicotine—in the form of cigarettes, smokeless tobacco,
and nicotine replacement patches—remains the substance
used most often during pregnancy. Although cigarette
smoking in the United States has decreased significantly
over the last 20 years, 26% of reproductive-aged women
still smoke, and 15% to 20% of women smoke during their
pregnancies (Andres and Dar, 2000). Women who smoke
during pregnancy are more likely to use opioids, alcohol,
cocaine, amphetamines, and marijuana during pregnancy
than women who do not smoke (Vega et al, 1993). Cigarette smoking has been associated with numerous perinatal complications, often in a dose-dependent fashion.
Smoking has been shown to raise the risk of spontaneous
abortion, stillbirth, fetal growth retardation, prematurity,
perinatal mortality, and sudden infant death syndrome
(Andres and Day, 2000; Kallen, 2001; Lambers and Clark,
1996; Tuthill et al, 1999). Cigarette smoking represents
111
112
PART III
Maternal Health Affecting Neonatal Outcome
the most influential and most common factor adversely
affecting perinatal outcomes. In a 2008, Rogers “estimated
that if all the women in the United States stopped smoking, there would be an 11% reduction in stillbirths and 5%
reduction in neonatal deaths.”
Before 1970, the detrimental effects of alcohol abuse
during pregnancy were believed to be related only to
drunkenness, such as an increased risk for accidents.
There was a widely held belief that the placenta formed
a protective barrier between alcohol and the fetus. This
belief repudiated by studies in the United States (Jones and
Smith, 1973) and France (Lemoine et al, 1967) describing fetal alcohol syndrome. These studies led to the 1989
U.S. federal law requiring that warning labels be placed on
all alcoholic beverage containers regarding alcohol-related
birth defects. Despite this extensive public health campaign
designed to inform and warn women about the dangers
of alcohol consumption during pregnancy, approximately
10% of pregnant women still report using alcohol (Centers for Disease Control and Prevention, 2009). A smaller
percentage of these women are alcoholics, but their infants
are at significantly higher risk for fetal alcohol syndrome
or alcohol-related neurobehavioral disorders compared
with the infants of nonalcoholic pregnant women who use
alcohol.
Opium derivatives have been used as analgesics for
centuries and remain the most effective analgesics available. Opioids of clinical interest are morphine, heroin,
methadone, meperidine, oxycodone, and codeine. Perinatal problems associated with opium were first reported in
the late 1800s. Since the 1950s, heroin use, particularly
among women, has been endemic in most major American cities. Compared with cocaine, marijuana, alcohol,
and tobacco abuse, opioid addiction during pregnancy is
rare (Shankaran et al, 2007). The prevalence of opioid use
among pregnant women is reported to range from 1% to
2% (Vega et al, 1993; Yawn et al, 1994) to as much as
21% in a highly selected group of women (Behnke and
Eyler, 1993; Nair et al, 1994; Ostrea et al, 1992b). One
multicenter study found that the prevalence of opioid
use varied by center and ranged from 1.6% to 4.5% at
the different sites (Lester et al, 2001). In addition, these
centers reported higher rates of opioid use by mothers of
low-birthweight and very low-birthweight infants. Rates
for heroin use are higher in metropolitan areas and cities
and are more concentrated in northeastern and west coast
cities. Opioid abuse is more common in groups of lower
socioeconomic status, and women using opioids during
pregnancy are more likely to use other drugs (Bauer, 1999;
Brown et al, 1998; van Baar and de Graaff, 1994). Investigators have also reported that 93% of women identified as
using opioids and cocaine during pregnancy had also used
a combination of alcohol, nicotine, or marijuana (Bauer,
1999). Of the opioid drugs known to be abused during
pregnancy, heroin and methadone have been studied the
most extensively. Heroin can be ingested through smoking or by the intranasal or intravenous route. Reports
from European countries suggest a trend away from intravenous injection of opioids (Hartnoll, 1994). The use of
noninjectable heroin may reduce the risk of transmission
of human immunodeficiency virus (HIV); however, its
wider use ensures the emergence of new groups of heroin
users for whom the risk of intravenous use is a major
deterrent.
The euphoria-producing effect of cocaine was exploited
extensively in the United States in the late nineteenth and
early twentieth centuries, when the agent was an active
ingredient in a number of widely used over-the-counter
elixirs and tonics. Cocaine use markedly decreased after
the Harrison Narcotic Act of 1914 and the supervening
Comprehensive Drug Abuse Prevention and Control Act
of 1970, which classified cocaine as a schedule II drug
(i.e., one of “high abuse potential with restricted medical
use,” similar to opioids, barbiturates, and amphetamines).
Cocaine’s reputation as a glamour drug, the widely held
misconception that cocaine is not addictive, and the development and marketing of crack, a cheap version of cocaine,
were major factors in the resurgence of drug use. Growing concern regarding the effects of maternal cocaine use
on pregnancy outcomes was one of the reasons that the
U.S. Congress passed the 1986 Narcotics Penalties and
Enforcement Act, which imposed severe penalties on
any person convicted of either possessing or distributing
effects; however, this law did not appreciably alter the fact
that cocaine and other stimulants had become the drugs
of choice for women in the United States. In the 1990s,
studies based on urine toxicology screening reported a
prevalence of cocaine use among pregnant women of 5%
in New York City, 1.1% in a geographic sample in California, and less than 0.5% in private hospitals in Denver,
Colorado (Burke and Roth, 1993). Prevalence increases to
18% when both self-reporting and urine testing are used,
and the highest prevalence rates are reported from studies
using meconium testing.
Amphetamines have surpassed cocaine as the primary
illicit drugs used by pregnant women in many areas of
California and other states. Methamphetamine (or “crystal”) has been the primary form abused, because it can be
produced locally and fairly cheaply. Greater restrictions
on the importing of cocaine have also contributed to resurgence in amphetamine use. Amphetamines have always
been popular among adolescents, especially females, and
accordingly women of child-bearing age are at high risk
for perinatal abuse. A California study of drug-exposed
infants in the social welfare system documented a higher
prevalence of amphetamine use among white pregnant
women than in women of other ethnicities (SagatunEdwards et al, 1995). The Infant Development, Environment, and Lifestyle (IDEAL) study chose four major U.S.
cities with known methamphetamine abusing populations
and reported methamphetamine use in 5.2% of pregnant
women. As in previous reports, these women frequently
used other substances: 25% used tobacco, 22.8% used
alcohol, 6% used marijuana, and 1.3% used barbiturates
(Arria et al, 2006).
According to the 2003 National Survey on Drug
Use and Health, marijuana is the most widely used illegal drug used in the United States, with approximately
14.6 million people reporting that they use the drug. In
a 1986 study, the most frequent age of marijuana use
for women included the childbearing ages, with 50% of
18- to 35-year-old women reporting that they used marijuana at least once and 8% reporting that they used marijuana a minimum of 10 of the past 30 days (Clayton et al,
113
CHAPTER 12 Perinatal Substance Abuse
1986). The 2002-2006 National Household Survey on
Drug Abuse included information on drug use in the last
30 days from over 94,000 women aged 18 to 44 years, of
whom 5017 were pregnant. Marijuana use was reported
in 7.3% of nonpregnant women and 2.8% of pregnant
women with moderate to heavy use (more than five times
per month) in 1.8% of pregnant women compared with
3.7% in nonpregnant women. Marijuana use declined
over the course of pregnancy, from 4.5% in the first trimester to 1.5% in the third trimester (Muhuri and Gfroerer, 2009).
HEALTH POLICY
In the 1980s, with rising cocaine use and the emergence
of crack cocaine, national attention turned to drug use
during pregnancy, with a general public outcry being the
result. Children born to crack addicts were widely believed
to be irrevocably damaged and public opinion was that
mothers should be punished. As the foster care system
became overwhelmed and as evidence to the contrary has
emerged, public opinion regarding prenatal drug use has
shifted more toward maternal treatment and prevention
rather than punishment. As Lester et al (2004) stated, the
initial overreaction of the public “in which drug-exposed
children were characterized as irrevocably and irreversibly damaged” has shifted “to a perhaps equally premature excessive ‘sigh of relief’ that drugs such as cocaine
do not have lasting effects, especially if children are
raised in appropriate environments.” This statement has
led to a change in the public discourse regarding health
policy interventions for substance use and abuse during
pregnancy.
The focus of current policy is to provide appropriate
medical care for substance-using pregnant women, including medical management of their chemical dependency
and programs to decrease substance use during pregnancy.
However, there remains the important step of recognizing
that “the idea that illegal drugs are more harmful to the
unborn fetus than legal drugs is incorrect” (Thompson et
al, 2009). Future research and intervention need to include
programs to educate women of childbearing age of the significant effects of both legal and illegal substance use on
the early-gestation fetus, starting even before pregnancy
may be recognized.
PERINATAL EFFECTS OF SPECIFIC
DRUGS (Table 12-1)
ETHANOL (ALCOHOL)
Pharmacology and Biologic Actions
Alcohol is a mood-altering substance that enhances
the effects of the inhibitory neurotransmitter gamma-
aminobutyric acid and lessens the effect of the excitatory
neurotransmitter glutamate, thus acting as a central nervous system (CNS) depressant or sedative. Alcoholic
beverages come in many forms, and for centuries they
have been consumed for diverse reasons: celebrations,
relaxation, religious ceremonies, and medicinal purposes
(alcohol is an excellent sedative and tocolytic agent). The
alcohol contained in alcoholic beverages is ethanol. Ethanol is absorbed in the digestive tract and into the body
fat and bloodstream. Ethanol is metabolized to acetaldehyde by alcohol dehydrogenase (ADH), primarily in the
liver. ADH is then metabolized to acetate by aldehyde
dehydrogenase (ALDH) and eventually eliminated as
water and CO2. Although acetaldehyde is short-lived, it
can cause significant tissue damage, which is particularly
evident in the liver, where most alcohol metabolism takes
place. Pregnant women have slower rates of alcohol clearance, likely related to hormonal alterations in the activity
of the alcohol-metabolizing enzymes; this leads to slower
clearance of alcohol compared with nonpregnant women
consuming the same amount of alcohol (Shankaran et al,
2007).
Complications of Pregnancy
Heavy drinking carries a higher risk of cardiovascular and
hepatic complications in women compared with men, and
the alcohol-associated mortality rate is also considerably
higher (Smith and Weisner, 2000). These factors alone
can clearly complicate a woman’s pregnancy. In addition,
nutritional deficiencies and poor diet can affect general
health and dentition, which can negatively affect a pregnancy. Alcoholism is a chronic disease that is often progressive and can be fatal. Pregnant alcoholics often have
related medical disorders such as cirrhosis, pancreatitis,
and alcohol-related neurologic problems. These disorders
can affect the health and well-being of their fetus.
TABLE 12-1 Enhanced Risk for Various Events or Processes after Substance Use during Pregnancy*
Event or Process
Ethanol
Cigarettes
Marijuana
Opiates
Cocaine
Malformations
+
–
–
–
–
Amphetamines
–
Abortion/stillbirth
+
+
?
+
+
+
Intrauterine growth restriction
+
+
–
+
+
+
Prematurity
–
+
?
+
+
+
Withdrawal
?
+
+/–
+
–
–
Central nervous system sequelae
+
?
+
?
+
?
Sudden infant death syndrome
+
+
?
+
?
?
Foster care
+
–
–
+
+
+
+, Causes event or process; –, does not cause event or process; ?, not known whether agent causes event or process.
*Although risk is increased, the risk ratio ranges for many from 1 to 2 for these associations.
114
PART III
Maternal Health Affecting Neonatal Outcome
Alcohol affects prostaglandin levels, increasing levels of
its precursors in human placental tissue and thus affecting fetal development and parturition. In fact, researchers have used this knowledge to test the effect of aspirin,
which inhibits alcohol-induced increases in prostaglandin
levels, on reducing alcohol-induced fetal malformations
in a mouse model (Randall, 2001). Specific obstetric complications of heavy drinking may relate to alterations in
prostaglandin levels, including an increased risk for spontaneous abortion, abruptio placenta, and alcohol-related
birth defects such as fetal alcohol syndrome.
Discriminating
Features
short
palpebral
fissures
Associated
Features
epicanthal
folds
low nasal
bridge
flat midface
Fetal Alcohol Syndrome
Fetal alcohol syndrome (FAS) was first described by
Lemoine (1967), a Belgian pediatrician who observed a
common pattern of birth anomalies in children born to
alcoholic mothers in France. This description was followed
by a landmark article by Jones and Smith (1973) reporting
similar features in several children born to alcoholic mothers in the United States.
It is unclear how much alcohol exposure is necessary to
cause fetal teratogenicity, and even high consumption levels do not always result in the birth of a child with FAS
(Abel and Hannigan, 1995). However, a woman with a previous affected child is at increased risk for having a child
with FAS if she consumes alcohol during a subsequent
pregnancy. The adverse effects of alcohol on the fetus are
related to gestational age at exposure, the amount of alcohol consumed, and the pattern of consumption (e.g., binge
drinking), maternal peak blood alcohol concentrations,
maternal alcohol metabolism, and the individual susceptibility of the fetus. Studies show that maternal peak blood
alcohol levels are affected by maternal nutrition, age, body
size, and genetic disposition (Eckardt et al, 1998; Maier
and West, 2001). In addition, various risk factors increase
susceptibility to FAS, including advanced maternal age and
confounding factors such as nonwhite race, poverty, and
socioeconomic status (Abel, 1995; Bagheri et al, 1998; May
and Gossage, 2001). In the United States, the incidence
of FAS is tenfold higher for African Americans living in
poverty than for white middle-class women (Abel, 1995).
Despite the differences in incidence of FAS worldwide,
reports consistently indicate poverty or socioeconomic
status as major determinants of FAS (Abel, 1995; May
et al, 2000).
Features of FAS include characteristic facial dysmorphology (short palpebral fissures, midface hypoplasia,
broad flat nasal bridge, flat philtrum, and thin upper lip;
Figure 12-1), prenatal and postnatal growth deficiency,
and variable CNS abnormalities. Skeletal anomalies,
abnormal hand creases, and ophthalmologic, renal, and
cardiac anomalies have been described in children with
FAS, but less frequently than the facial dysmorphology and CNS abnormalities that include structural brain
defects (e.g., dysgenesis of the corpus callosum and cerebellar hypoplasia), cognitive abnormalities, delayed brain
development, and signs of neurologic impairment, including lifelong behavioral and psychosocial dysfunction. In
1996, the Institute of Medicine further defined the criteria for the diagnosis of FAS and proposed a new term—
alcohol-related neurodevelopmental disorder (ARND).
short nose
minor ear
anomalies
indistinct
philtrum
micrognathia
thin
upper lip
In the Young Child
FIGURE 12-1 Facies in fetal alcohol syndrome. (From Streissguth
AP, Little RE: Alcohol, pregnancy, and the fetal alcohol syndrome, ed 2, unit
5 of Alcohol use and its medical consequences: a comprehensive slide
teaching program for biomedical education. Developed by Project Cash of
the Dartmouth Medical School. Reproduced with permission from MilnerFenwick, Inc., Timonium, Michigan, 1994.)
This term includes structural CNS and cognitive abnormalities in children with confirmed fetal exposure to alcohol. Unlike FAS, a diagnosis of ARND does not require
the presence of facial or other physical abnormalities. In
2000, the American Academy of Pediatrics Committee on
Substance Abuse published these new definitions with an
explanatory drawing (Figure 12-2).
The incidence of FAS in the United States has been estimated to vary from 1.95 to 5 cases per 1000 live births
(Abel, 1995; American Academy of Pediatrics Committee on Substance Abuse and Committee on Children with
Disabilities, 2000; Bertrand et al, 2005; Sampson et al,
1997). FAS is recognized more frequently in the United
States than in other countries and is most common (4.3%)
among women who report heavy drinking (Abel, 1995).
Accurate incidence and prevalence rates of FAS are difficult to obtain because of wide variations in methodologies used for estimation of rates, and because the clinical
diagnosis is often missed in the neonatal period. In fact,
most cases (up to 89%) are not diagnosed until after a
child is age 6 (Centers for Disease Control and Prevention
[CDC], 1997).
FAS is diagnosed from the history and physical findings. No laboratory tests are available for clinical use to
quantify the extent of alcohol exposure during fetal life.
There are also no clinical methods for validating maternal
self-reporting of alcohol use, quantifying the level of fetal
exposure, or predicting future disability after fetal exposure (Jones and Chambers, 1999). Koren et al (2002) have
proposed meconium fatty acid ethyl ester levels as a potential biologic marker for fetal alcohol exposure. Whether
this finding is shown to correlate with childhood outcomes
remains to be studied. Investigators have shown that pediatricians fail to recognize FAS in the newborn and do not
always inquire about alcohol exposure during pregnancy
115
CHAPTER 12 Perinatal Substance Abuse
FAS with confirmed
maternal exposure
FAS without confirmed
maternal exposure
Partial FAS with
confirmed exposure
OR
OR
OR
Alcohol-related
birth defects (ARBD)†
Alcohol-related
neurodevelopmental
disorder (ARND)†
A
B
C
D
E
F
Confirmed
Exposure to
Alcohol
Facial
Anomalies
Growth
Retardation
CNS
Abnormalities
Cognitive
Abnormalities
Birth
Defects
*Adapted from Fetal Alcohol Syndrome; Diagnosis, Epidemiology,
Prevention, and Treatment. 1996;4–5. Letter designations in the
figure indicate the following:
A. Confirmed maternal alcohol exposure indicates a pattern of excessive
intake characterized by substantial, regular intake or heavy episodic
drinking. Evidence of this pattern may include frequent episodes of
intoxication, development of tolerance or withdrawal, social problems
related to drinking, legal problems related to drinking, engaging in
physically hazardous beavior while drinking, or alcohol-related medical
problems such as hepatic disease.
B. Evidence of a characteristic pattern of facial anomalies that includes
features such as short palpebral fissures and abnormalities in the
premaxillary zone (e.g., flat upper lip, flattened philtrum, and flat midface).
C. Evidence of growth retardation, including at least one of the following:
• low birthweight for gestational age
• decelerating weight over time not caused by nutrition
• disproportional low weight to height
D. Evidence of CNS neurodevelopmental abnormalities, including at least
one of the following:
• decreased cranial size at birth
• structural brain abnormalities (e.g., microcephaly, partial or complete
agenesis of the corpus callosum, cerebellar hypoplasia)
• neurological hard or soft signs (as age appropriate), such as impaired
fine motor skills, neurosensory hearing loss, poor tandem gait, poor
eye-hand coordination
E. Evidence of a complex pattern of behavior or cognitive abnormalities that
are inconsistent with developmental level and cannot be explained by
familial background or environment alone, such as learning difficulties;
deficits in school performance; poor impulse control; problems in social
perception; deficits in higher level receptive and expressive language;
poor capacity for abstraction or metacognition; specific deficits in
mathematical skills; or problems in memory, attention, or judgment
F. Birth defects associated with alcohol exposure include:
Cardiac
Atrial septal defects
Ventricular septal defects
Aberrant great vessels
Tetralogy of Fallot
Skeletal
Hypoplastic nails
Shortened fifth digits
Radioulnar synostosis
Flexion contractures
Camptodactyly
Clinodactyly
Pectus excavatum and carinatum
Klippel-Feil syndrome
Hemivertebrae
Scoliosis
Renal
Aplastic, dysplastic,
hypoplastic kidneys
Horseshoe kidneys
Ureteral duplications
Ocular
Strabismus
Retinal vascular
anomalies
Refractive problems
Secondary to small globes
Auditory
Conductive hearing loss
Neurosensory hearing loss
Other
Virtually every malformation has been described in some
patient with FAS. The etiologic specificity of most of these
anomalies to alcohol teratogenesis remains uncertain.
Hydronephrosis
†Alcohol-related
effects indicate clinical conditions in which there is a
history of maternal alcohol exposure, and where clinical or animal
research has linked maternal alcohol ingestion to an observed outcome.
There are two categories, alcohol-related neurodevelopmental disorder
and alcohol-related birth defects, which may co-occur. If both diagnoses
are present, then both diagnoses should be rendered.
FIGURE 12-2 Diagnostic classification of fetal alcohol syndrome and alcohol-related effects. (From the American Academy of Pediatrics Committee on Substance Abuse and Committee on Children with Disabilities: Fetal alcohol syndrome and alcohol-related neurodevelopmental disorders, Pediatrics
106:359. Reproduced with permission from the American Academy of Pediatrics, 2000.)
116
PART III
Maternal Health Affecting Neonatal Outcome
(Stoler and Holmes, 1999). One promising screening tool
is the use of averaged cranial ultrasound images to examine the size and shape of the corpus callosum, which is
typically dysgenic in FAS (Bookstein et al, 2005). Guidelines to aid in the earlier recognition and referral of infants
and children with FAS and fetal alcohol spectrum disorder
have been published recently (Bertrand et al, 2005; Hoyme
et al, 2005). FAS is not a problem just for neonatologists.
Adolescents who were exposed prenatally to alcohol have a
different approach to alcohol than their nonexposed peers,
with an increased risk for earlier use and subsequent alcohol abuse (Baer et al, 2003). One study in rodents suggests
that fetal ethanol exposure increases ethanol intake later
in life by making it smell and taste better (Youngentob
and Glendinning, 2009). Children with FAS continue to
have serious disabilities into adulthood (Streissguth et al,
1991; Streissguth, 1993). Although the facial features and
growth restriction are no longer as distinctive as during
childhood, mental retardation continues to have a significant effect. Adults with FAS have behavior, socialization,
and communication dysfunction, and on average they
function at the second- or third-grade level. A significant
number of FAS patients do not achieve fully independent
living. Earlier recognition and intervention for children
with FAS and its variants may help to minimize eventual
adulthood disabilities and help to prepare adolescents and
young adults with the disorder for independent living
(Bertrand et al, 2005).
Fetal Growth
Intrauterine growth restriction (IUGR) is one of the most
consistent findings of prenatal exposure to alcohol (Hannigan and Armant, 2000). Growth deficit begins in utero
and continues throughout childhood (American Academy
of Pediatrics Committee on Substance Abuse and Committee on Children with Disabilities, 2000). The facial
features and the growth restriction become less noticeable
during adolescence and puberty (Streissguth et al, 1991;
Streissguth, 1993).
CIGARETTE SMOKING AND NICOTINE
Pharmacology and Biologic Actions
Cigarette smoke contains a complex mixture of approximately 4000 compounds, including nicotine and carbon
monoxide, which can adversely affect the fetus (Lester
et al, 2004). In rodents, nicotine releases chemicals in
the reward center of the brain, which likely triggers the
euphoria that smokers experience. Nicotine activates nicotinic acetylcholine receptors, and these receptors remain
depressed for a longer time after their activation stops,
which likely accounts for compulsive smoking (Cohen,
2007). Nicotine crosses the placenta and concentrates in
fetal blood and amniotic fluid, where its levels significantly
exceed maternal blood concentrations (Haustein, 1999).
The serum concentration of cotinine, the primary metabolite of nicotine, is used to quantitate the level of smoking and fetal exposure. Cotinine has a half-life of 15 to 20
hours, and because its serum levels are tenfold higher than
those of nicotine, this substance may represent a better
marker for intrauterine exposure (Lambers and Clark,
1996).
Complications of Pregnancy
Although the exact mechanism of the adverse effects of
smoking on pregnancy is unknown, cigarettes contain
numerous potentially toxic compounds that affect fetal
health in a number of ways. Nicotine and its metabolites
can act as vasoconstrictors, and a study in pregnant rhesus monkeys demonstrated a nicotine-associated decrease
in uterine blood flow (Suzuki et al, 1980), which might
provide a partial explanation for the association between
maternal cigarette smoking and low birthweight. Theories
regarding mechanisms for the adverse effects of smoking on fetal health include direct vasoconstrictive effects
of nicotine on uteroplacental blood flow, the induction of
fetal hypoxia from carbon monoxide production, direct
toxic effects and indirect effects of altered maternal nutritional intake, and altered maternal and placental metabolism (Andres and Day, 2000; Pastrakuljic et al, 1999).
When pregnant women smoke cigarettes, the resulting
increased levels of carbon monoxide cross the placenta
and form carboxyhemoglobin in the fetus, with resulting
hypoxemia (Lambers and Clark, 1996). Supporting this
theory, serum erythropoietin levels are higher in tobacco
smoke–exposed infants at delivery, a finding that is presumed to reflect fetal hypoxia (Beratis et al, 1999; Jazayeri
et al, 1998). In addition to the fetal hypoxia theory, there
have recently been studies demonstrating that nicotine
may act as a developmental neurotoxin targeting nicotinic acetylcholine receptors (Lester et al, 2004; Levin and
Slotkin, 1998) and may disturb protein metabolism during gestation, leading to decreased serum amino acids in
umbilical cord blood (Jauniaux et al, 2001).
Maternal smoking has also been show to affect the
length of gestation in a dose-dependent manner, with a
higher risk of preterm delivery (Jaakkola et al, 2001; Savitz et al, 2001) and a twofold increase in the incidence of
placental abruption (Ananth et al, 1996). Perinatal mortality is increased in pregnant smokers, likely reflecting
the increases in rates of prematurity, placental abruption, and placenta previa in women who smoke. Mothers who smoke during pregnancy commonly continue
to smoke during their infants’ childhood. Asthma and
recurrent otitis media are more common in infants who
are exposed to passive smoking (Ey et al, 1995; Martinez
et al, 1995).
Fetal Growth
The effect of smoking on fetal growth is significant and
dose dependent (Kyrklund-Blomberg et al, 1998; Nordentoft et al, 1996). Studies have shown lower birthweights
associated with levels of nicotine exposure, with a 1-g
reduction in birthweight observed for every microgram
per milliliter increase in maternal serum cotinine level
(Eskenazi et al, 1995; Perkins et al, 1997). Investigators
have shown a dose-dependent relationship between the
amount of smoking and the extent of fetal growth restriction and birthweight reduction (Horta et al, 1997; Jaakkola
et al, 2001; Savitz et al, 2001; Sprauve et al, 1999).
CHAPTER 12 Perinatal Substance Abuse
Both reducing and ceasing cigarette smoking during
pregnancy have been shown to be beneficial and to lead to
improved fetal growth. Lieberman et al (1994) have shown
that if pregnant women stop smoking during the third trimester, their infants’ weights are indistinguishable from
those of a nonsmoking population. Other investigators
report that even a modest reduction in smoking is associated with improved fetal growth (Li et al, 1993; Walsh et
al, 2001).
117
Marijuana, the dried material from the hemp plant Cannabis
sativa, is most often smoked but can be ingested with food.
Its most active ingredient is delta-9-tetrahydrocannabinol (THC), which binds to cannabinoid receptors in the
brain and modifies the release of several neurotransmitters. Its primary biologic effects include euphoria; relaxation; increased heart rate, blood pressure, and appetite;
and impaired coordination, decision-making, short-term
memory, concentration, and learning. Although controversial, there is increasing evidence that regular marijuana
use can cause respiratory difficulties, cognitive impairments, withdrawal, and dependence (Khalsa et al, 2002).
THC crosses the placenta and collects in the amniotic
fluid.
occurring endorphins and enkephalins (Vaccarino and
Kastin, 2000). As modulators of the sympathoadrenal system, endogenous opioids are important during periods of
diverse forms of stress. Activation of these receptors by
the endogenous opioids has physiologic effects, including
analgesia, drowsiness, respiratory depression, decreased
gastrointestinal motility, nausea, vomiting, and alterations
in the endocrine and autonomic nervous systems. Activation of these same endogenous opioid receptors by exogenous opioid drugs has similar clinical effects, producing
euphoria, sleepiness, and decreased sensitivity to pain, as
well as adverse effects such as constipation and nephrotic
syndrome (ACOG, 2005).
The use of opioid drugs can result in the development of
tolerance, physiologic dependence, and addiction. Tolerance leads to a shortened duration of the action of opioids
and a decrease in the intensity of the drug action, followed
by the need for a higher dose to obtain the same clinical effect. Tolerance is believed to result from continued
occupancy of the opioid receptor. Continuous administration of opioids, therefore, leads to the more rapid onset
of tolerance (Anand and Arnold, 1994; Suresh and Anand,
2001). With physiologic dependence, there is a need for
further drug administration to prevent withdrawal symptoms (agitation, dysphoria, temperature instability). Addiction is a more severe form of dependence that involves a
complex pattern of drug-seeking behavior (Christensen,
2008).
Complications of Pregnancy
Complications of Pregnancy
Marijuana use during pregnancy is inconsistently reported
to have effects on birth outcomes. The inconsistency likely
stems from the fact that marijuana is often used in combination with other substances, thus potentiating the risks
for prematurity and low birthweight. Day et al (1991)
reported a higher incidence of meconium-stained amniotic fluid and other pregnancy and delivery complications
after maternal marijuana use, but subsequent studies have
not replicated this finding. The IDEAL study did not
find any relation between marijuana use and altered fetal
growth parameters (Smith et al, 2006). Frequent use of
marijuana was associated with a small decrement in birthweight in a study that did not control for other illicit drug
use (Schempf, 2007).
Obstetric complications associated with maternal use of
opioids include a higher incidence of spontaneous abortion, premature delivery, preterm labor, abruptio placentae, chorioamnionitis, impaired fetal growth, and fetal
distress. In women who use opioids during pregnancy,
the incidence of preterm labor and premature delivery
ranges from 25% to 41% (Chiriboga, 1993; Lam et al,
1992; Little et al, 1990). Maternal opioid use is also associated with higher rates of meconium-stained amniotic
fluid, lower American Pediatric Gross Assessment Record
(Apgar) scores, longer duration of membrane rupture (Gillogley et al, 1990), and increased incidence of syphilis and
HIV infection at birth (Bauer, 1999). The rates of maternal complications of pregnancy are further increased when
drug use is added to infection with HIV. Women infected
with HIV who also use opioids, particularly methadone,
have higher rates of miscarriage, preterm deliveries, and
small-for-gestational-age infants, and more vaginal and
urinary tract infections than women with HIV who do not
use drugs (Mauri et al, 1995).
The etiology of these opioid-associated pregnancy
complications is multifactorial. Maternal lifestyle, malnutrition, infections, and polydrug effects are likely to
result in poor perinatal outcomes, including poor intrauterine growth and prematurity. Because the drug supply
is often episodic, the pregnant addict is subject to episodes of withdrawal and overdose, thereby subjecting the
fetus to intermittent episodes of hypoxia in utero, hindering growth and raising the risk of spontaneous abortion, stillbirth, fetal distress, and prematurity. Infants
born to mothers who are addicted to opioids are more
MARIJUANA
Pharmacology and Biologic Actions
OPIOIDS (INCLUDING PRESCRIPTION PAIN
KILLERS)
Pharmacology and Biologic Actions
Opioids are either drugs derived from the opium poppy or
synthesized compounds that have similar biologic actions.
The prototype opioid is morphine, and all opioids relate to
it. For example, heroin (diacetylmorphine) exerts its effects
by being metabolized to morphine, as does codeine, which
is methylated morphine. Other opioids, such as methadone and oxycodone, are structurally unlike morphine but
share its pharmacologic properties, because they stimulate
similar opioid receptors. Specific opioid receptors (μ, δ, κ)
have been identified in the nervous system and bowel that
are activated by endogenous opioids, such as the naturally
118
PART III
Maternal Health Affecting Neonatal Outcome
likely to be of low birthweight, to be premature, and
to suffer from infection and perinatal asphyxia (Christensen, 2008).
Fetal Growth
Initial reports from studies addressing the effects of maternal opioid use on the fetus suggested that infants exposed
to opioids in utero have a higher incidence of IUGR (Lam
et al, 1992) and smaller head circumference (Bauer, 1999;
Boer et al, 1994). Hulse et al (1997) reported an association with heroin abuse and increased prematurity, low
birthweight, and reduced fetal growth. However, more
recent work controlling for confounding factors has not
demonstrated a significant relationship between opioid
use and prematurity, low birthweight, or IUGR (Minozzi
et al, 2008; Sharpe and Kuschel, 2004).
COCAINE
Pharmacology and Biologic Actions
Cocaine is a highly psychoactive stimulant with a long
history of abuse. A naturally occurring anesthetic of the
tropane family of alkaloids, cocaine is obtained from the
Erythroxylon coca plant, which is indigenous to the mountain slopes of Central and South America. The coca leaf has
been chewed or made into a stimulant tea by the natives of
these areas to decrease fatigue and hunger. The pharmacologic actions of cocaine include inhibition of postsynaptic reuptake of norepinephrine, dopamine, and serotonin
neurotransmitters by sympathetic nerve terminals, thus
allowing higher concentrations of these neurotransmitters.
In adults, cocaine binds strongly to neuronal dopamine
reuptake transporters, thereby increasing postsynaptic
dopamine at the mesolimbic and mesocortical levels and
producing the addictive cycle of euphoria and dysphoria
(Malanga and Kosofsky, 1999). Tryptophan uptake is similarly inhibited, altering serotonin pathways with resultant
effects on sleep.
Cocaine use leads to a sense of well-being, increased
energy, increased sexual achievement, and an intense
euphoria or “high.” The sympathomimetic action can have
potentially devastating physiologic effects on the cardiovascular system. In adults, cocaine has been associated with
cerebral hemorrhage, cardiac arrest, cardiac arrhythmias,
myocardial infarction, intestinal ischemia, and seizures.
Chronic use is associated with anorexia, nutritional problems, and paranoid psychosis and can ultimately result in
neurotransmitter depletion and a “crash,” characterized by
lethargy, depression, anxiety, severe insomnia, hyperphagia, and cocaine craving.
Two forms of cocaine are commonly used—cocaine
hydrochloride and cocaine base, which are either extracted
by organic solvents or precipitated as “crack” through the
use of ammonia and baking soda). Cocaine hydrochloride
is a water-soluble white powder that is used orally, intranasally (“snorting”), or intravenously (“running”). Intravenous users are more likely to have a history of heroin
abuse and often use the drug in combination with heroin
(known as speedballing). Cocaine hydrochloride decomposes on heating and is, therefore, cocaine converted to
the free base for inhalation. “Freebasing” involves extracting cocaine from aqueous solution into an organic solvent
such as ether. Crack, the most widely available form of
freebase, is almost pure cocaine; when smoked, it readily
enters the bloodstream to produce levels similar to those
occurring with intravenous use. Crack cocaine is popular
in urban minority communities, where it may be smoked
in combination with PCP (known as spacebasing). Crack
smoking appears to be particularly reinforcing and is associated with compulsive use, binges, and acceleration of the
addictive process.
Cocaine and some of its metabolites readily cross the
placenta and achieve pharmacologic levels in the fetus
(Schenker et al, 1993). Amniotic fluid may serve as a reservoir for cocaine, and its metabolites and prolong exposure
to vasoactive compounds. The extent to which cocaine or
its metabolites are responsible for aberrant fetal growth,
neurodevelopmental sequelae in exposed infants, and the
range of congenital malformations reported in the literature may be less than suggested by uncontrolled case
reports early in the cocaine-epidemic era. The confounding effects of increased use of multiple drugs, tobacco,
alcohol, nutritional deficits, and decreased use of prenatal care among cocaine users make interpretation of the
causal relationships between gestational cocaine exposure
and intrauterine growth and subsequent neurobehavioral
development difficult (Chiriboga, 1993). These identified
confounders might serve to explain the reported effects
attributed to cocaine in clinical series (Dempsey et al,
1996), although significant effects on the fetus and newborn have been reported in more recent studies that controlled for many confounders (Bada et al, 2002; Shankaran
et al, 2007).
Complications of Pregnancy
Adverse perinatal outcomes associated with cocaine use
are believed to be largely because of the vasoconstrictive
effects of cocaine on uterine blood supply (Woods et al,
1987). An increase in maternal mean arterial blood pressure, a decrease in uterine blood flow, and a transient
rise in fetal systemic blood pressure after an intravenous
cocaine infusion have been described in fetal sheep along
with significant fetal hypoxemia associated with changes in
uterine blood flow (Moore et al, 1986; Woods et al, 1987).
Maternal hypertension and intermittent fetal hypoxia
contribute to the higher risks for abruptio placentae and
IUGR seen in cocaine-exposed infants.
To date, no well-defined cocaine-associated syndrome
has been identified, and the teratogenic potential of
cocaine remains controversial. Earlier reports had suggested that cocaine-exposed infants had a higher rate of
limb reduction anomalies, heart defects, ocular anomalies,
intestinal atresia or infarction, and other vascular disruption sequences. However, the preponderance of more
recent data from multiple studies has failed to demonstrate higher rates of other congenital anomalies among
cocaine-exposed infants (Behnke et al, 2001). Any association between fetal cocaine exposure and malformations
is likely to be confounded by higher rates of maternal
tobacco, marijuana, or alcohol use among the cocaineexposed groups.
CHAPTER 12 Perinatal Substance Abuse
Women who use cocaine during pregnancy are at higher
risk for stillbirths, spontaneous abortions, abruptio placentae, IUGR, anemia and malnutrition, and maternal death
from intracerebral hemorrhage. Cocaine directly stimulates uterine contractions because of its alpha-adrenergic,
prostaglandin, or dopaminergic effects, with resulting
greater risk for fetal distress and premature deliveries.
Abruptio placentae appears to be related to cocaine only
when the drug is used shortly before delivery (Ostrea et
al, 1992b). Pregnant women who use cocaine are also at
high risk for premature labor, low-birthweight infants,
premature rupture of the membranes, and perinatal infections. Cocaine use significantly increases the odds ratio
for prematurity, low birthweight, premature rupture of
membranes, and IUGR (Bada et al, 2002, 2005; Shankaran
et al, 2007) as well as perinatal infections.
Overall, because of the higher risks of premature
delivery, the frequency of respiratory distress syndrome
is greater in cocaine-exposed infants. Cocaine-exposed
infants less frequently require surfactant administration
and intubation for respiratory distress syndrome; however,
the risks of bronchopulmonary dysplasia are similar in
infants who have and those who have not been exposed to
cocaine during gestation (Hand et al, 2001).
Fetal Growth
Infants exposed to cocaine in utero have lower birthweight, smaller birth length, and smaller head circumference (Bada et al, 2002, 2005; Behnke et al, 2001).
Cocaine is hypothesized to reduce fetal growth via vasoconstriction of uteroplacental vessels with consequent
decreased fetal substrate and oxygen delivery (Schempf,
2007). Several studies have shown a dose-response effect
of cocaine exposure on fetal growth. In the Maternal
Lifestyle Study, cocaine-exposed infants were 1 week
younger in gestational age, and after controlling for confounders, cocaine exposure was associated with decrements in birthweight (151 g), length (0.71 cm), and head
circumference (0.43 cm) at 40 weeks’ gestation (Shankaran et al, 2007). After adjusting for the effects of birthweight, gestational age, sex, maternal height, maternal
weight gain, and other drug use, newborns with a high
exposure to cocaine, as measured by radioimmunoassay
of cocaine metabolites in maternal hair, had a disproportionately smaller head circumference even for their
birthweight, resulting in “head wasting” (Bateman and
Chiriboga, 2000).
AMPHETAMINES
Pharmacology and Biologic Actions
Amphetamine (methylphenethylamine) was synthesized in
1887 and introduced in the United States in 1931. The
N-methylated form, methamphetamine (or “crystal”), is
increasingly abused because it readily dissolves in water for
injection and it sublimates (converts directly from a solid
to gas) when smoked (known as ice). The amphetamine
isomers have similar clinical effects and can be distinguished only in the laboratory. Amphetamines were initially marketed for the treatment of obesity and narcolepsy
119
and continue to be used for the treatment of attention
deficit disorders in children. Amphetamines are classified
as schedule II drugs, like cocaine and narcotics. Amphetamines are taken orally, inhaled, or injected. The clinical
effects and toxicity of these agents are often indistinguishable from those of cocaine. The primary difference is in
the duration of action. The psychotropic effects of cocaine
are of a short duration—5 to 45 minutes. The effects of
amphetamines may last from 2 to 12 hours. Methamphetamine exposure has direct and indirect effects on the fetus,
with increases in maternal blood pressure and restrictions
in delivering nutrients and oxygen to the fetus (Smith
et al, 2003). The clinical effects of amphetamines resemble
those of cocaine. Like cocaine, amphetamines are sympathomimetics, and they potentiate the actions of norepinephrine, dopamine, and serotonin. In contrast to cocaine,
amphetamines appear to exert their CNS effects primarily
by enhancing the release of neurotransmitters from presynaptic neurons. Amphetamines can block reuptake of
released neurotransmitters; they can also exert a weaker
direct stimulatory action on postsynaptic catecholamine
receptors.
Complications of Pregnancy
The medical and obstetric complications of amphetamine
use are similar to those described for cocaine use. Amphetamine toxicity has been described as more intense and prolonged than cocaine toxicity. Visual, auditory, and tactile
hallucinations are common, and microvascular damage
has been seen in the brains of chronic users. Amphetamine withdrawal is characterized by prolonged periods of
hypersomnia, depression, and intense, often violent paranoid psychosis. Obstetric complications include a higher
incidence of stillbirth. Methamphetamine use is also associated with an increased incidence of premature delivery
and placental abruption. Methamphetamine users who
stop using earlier in gestation have rebound weight gain,
suggesting that the anorexic effects are limited to continuous use (Smith et al, 2003). Like the pregnancies of
cocaine users, the pregnancies of amphetamine users are
characterized by poor prenatal care, sexually transmitted
diseases, and cardiovascular problems including abruptio
placentae and postpartum hemorrhage. The risk of cerebrovascular accidents is lower in pregnant amphetamine
users than in pregnant cocaine users, but the mechanism
for this difference is not understood.
Perinatal problems associated with maternal amphetamine use include prematurity and IUGR (Smith et al,
2006). Fetal growth restriction, leading to smaller head
circumference and lower birthweight, can result from
the vasoconstrictive effects of norepinephrine or other
vasoactive amines or from diminished maternal nutrient delivery as a consequence of the anorectic effect of
amphetamine. Systemic effects from altered norepinephrine metabolism explain the transient bradycardia and
tachycardia reported in exposed infants. Studies have
failed to show consistent patterns of malformations in
amphetamine-exposed infants, although several studies report cleft lip and cleft palate in association with
amphetamine and methamphetamine exposure during
early gestation (Plessinger, 1998).
120
PART III
Maternal Health Affecting Neonatal Outcome
Fetal Growth
The IDEAL study followed 84 methamphetamineexposed and 1534 unexposed infants (confirmed by
screening for meconium). Both groups included alcohol,
tobacco, and marijuana use, but excluded opioids, PCP,
and LSD (Arria et al, 2006; Smith et al, 2003, 2006).
Cocaine and methamphetamine use occurred together in
13% of the methamphetamine users, and tobacco, alcohol, and marijuana use were also more frequent in the
methamphetamine users. Methamphetamine-exposed
infants were 3.5-fold more likely to be small for gestational age (less than the tenth percentile; 18% incidence).
Methamphetamine contributed significantly to low birthweight, even after correcting for confounders such as
low socioeconomic status, gestational age, and tobacco
exposure.
IDENTIFYING PREGNANCIES AND
BABIES AT RISK
PREGNANCIES
Identification of perinatal substance abuse to intervene and
protect the health and well-being of both mother and child
has been a goal of practitioners for decades, ever since the
scope of the problem became known—particularly for
alcohol. Urine toxicology testing was initially believed to
be the best approach, and the universal approach was used
in many busy labor and delivery units, particularly in urban
centers. In 1994, an American College of Obstetricians
and Gynecologists (ACOG) Technical Bulletin concluded
that urine toxicology testing had limited ability to detect
substance abuse and therefore recommended against universal toxicology screening and for alternative screening
methods. In 2003, the U.S. Congress passed the Keeping Children and Family Safe Act. This law requires each
state (as a condition of receiving federal funds under the
Child Abuse Prevention and Treatment Act) to develop
policies and procedures designed “to address the needs
of infants born and identified as being affected by illegal
substance abuse or withdrawal symptoms resulting from
prenatal drug exposure.” This law included a requirement
that health care providers notify child protective services
regarding prenatal substance exposure, but differed from
the providers’ legal responsibility to report suspected child
abuse or neglect because the former “shall not be construed
to be child abuse” and “shall not require prosecution of the
mother” (Washington State Department of Health, 2009).
Each state was expected to develop their own guidelines
for identification of at-risk pregnancies. A 2004 ACOG
Committee Opinion stated that best practices included
universal screening questions followed by brief interventions or referrals. A number of screening tools have been
developed including tolerance, annoyed, cut down, eyeopener (T-ACE), tolerance, worried, eye-opener, amnesia, K/cut down (TWEAK), and parents, partner, past,
pregnancy (assess for substance abuse, domestic violence)
(4 P’s Plus) (Chang, 2001; Chasnoff et al, 2005; Sokol et
al, 1989). Identification of at-risk pregnancies concentrated on the maxim “if you don’t ask, they won’t tell.”
In 2008, ACOG issued a committee opinion entitled AtRisk Drinking and Illicit Drug Use: Ethical Issues in Obstetric
and Gynecologic Practice, which reaffirmed the 2004 ACOG
TABLE 12-2 Risk Indicators for Gestational Substance Exposure
Maternal
No prenatal care
Precipitous labor
Placental abruption
Repeated spontaneous abortions
Hypertensive episodes
Severe mood swings
Previous unexplained fetal demise
Myocardial infarction or stroke
Newborn
Jittery with normal blood glucose level
Marked irritability
Unexplained seizures or apneic spells
Unexplained IUGR
NEC in an otherwise healthy term infant
Neurobehavioral abnormalities
Signs of neonatal abstinence syndrome
IUGR, Intrauterine growth restriction; NEC, necrotizing enterocolitis.
statement. The opinion stated that “as a result of intensive
research in addiction over the past decade, evidence-based
recommendations have been consolidated into a protocol
for universal screening questions, brief intervention and
referral to treatment.” This approach—in particular, the
use of standard screening questionnaires—was strongly
recommended for obstetricians. Therefore a history of
drug and alcohol use should be routinely included in the
initial contact with every pregnant patient. To be effective,
the history taking must be nonjudgmental and must occur
in the context of other lifestyle questions. When a positive history of use is obtained, intervention should begin
immediately. The person taking the history should be prepared to offer preliminary counseling on risk reduction
and concrete referrals for treatment programs, although
access to drug programs is often restricted, inadequate, or
delayed.
Although the screening questionnaire approach works
well for women who seek prenatal care, it is not useful
for the significantly higher percentage of substance-using
pregnant women who do not seek or actively avoid prenatal care. For these women, it is more helpful to identify
maternal risk indicators for perinatal substance abuse at
the time of delivery. Use of one of the screening questionnaires in addition to drug testing increases the likelihood
of identifying at-risk pregnancies and neonates, allowing
for earlier referral for treatment or specialized interventions. Several of these maternal risk indicators for perinatal
substance abuse were identified in the 2004 ACOG statement (Table 12-2) (American College of Obstetricians and
Gynecologists, 2004).
BABIES
It is important for practitioners to know the difference
between a substance-exposed newborn and a substanceaffected newborn. The Washington State Department of
Health defines them as follows:
ll Substance exposed:
ll Tests positive for substances at birth or
ll Mother tests positive for substances at time of delivery or
ll Is identified by medical practitioner as having been
prenatally exposed to substances
CHAPTER 12 Perinatal Substance Abuse
Substance affected:
ll Has withdrawal symptoms resulting from prenatal
substance exposure or
ll Demonstrates physical and behavioral signs that can
be attributed to prenatal exposure to substances and
is identified by a medical practitioner as affected
For the identification of either of these groups of infants,
assessment of both maternal and newborn risk indicators
(see Table 12-2) is essential. If risk indicators suggest perinatal substance abuse, then consideration should be given
to newborn drug testing. For urine testing, there is a poor
correlation between maternal and newborn tests. The earliest newborn urine will contain the highest concentration of
substances, but the first urination may be missed and urine
output in the first day is often scant. However, some drug
metabolites such as cocaine are present for 4 to 5 days, and
marijuana metabolites may persist for weeks. The disadvantages of newborn urine drug testing are that it primarily reflects substance exposure during the preceding 1 to 3
days, and alcohol is nearly impossible to detect. Meconium
drug testing (at term) reflects substance exposure during
the second half of gestation, has a high sensitivity for opioids and cocaine, and can assess for more drugs than urine
testing. The cost is similar to newborn urine testing, but
it generally takes longer to obtain results. Other newborn
drug tests include hair—which is costly and has a high sensitivity for cocaine, amphetamines, and opioids but not for
marijuana—and umbilical cord segments, which is an evolving technology that is not widely available (Kuschel, 2007).
ll
PREGNANCY MANAGEMENT
Ideally, perinatal substance use or abuse should be identified
by universal screening procedures during prenatal visits; this
gives the practitioner an opportunity to intervene, with the
goal being prevention of significant obstetric and neonatal
complications related to substance abuse. Screening procedures and counseling should include the following topics:
Antepartum
ll Initial screening for hepatitis, HIV, and tuberculosis (if not part of routine prenatal care) and ongoing
screening for sexually transmitted infections
ll Referring to methadone treatment program, if
appropriate
ll Discussing possible drug effects on the fetus and
newborn
ll Discussing contraception and prevention of sexually
transmitted disease
ll Discussing breastfeeding issues related to alcohol
and drug use
Intrapartum
ll Effects of recent drug use on labor and fetal well-being
ll Pain management (women in methadone or alternative opioid treatment programs will be less responsive to opioid pain medications)
ll Intrapartum prophylaxis for HIV, herpes simplex
virus infections
ll Need for social services involvement
Postpartum
ll Breastfeeding issues (related to both drug use and
infection)
ll Contraception and pregnancy prevention including
tubal ligation
ll
ll
121
Support for continuation in, or initiation of, a drug
treatment program
Child Protective Services notification
MATERNAL METHADONE MAINTENANCE
The potential benefits of maternal methadone maintenance are numerous (Kandall and Doberczak, 1999; Ward
et al, 1999) and include the prevention of opioid withdrawal symptoms in the mother, better medical and prenatal care, improved health and growth of the fetus, and
maintenance of opioid levels in the mother to decrease
both the use of illicit drugs and the potential for perinatal
infections. Methadone maintenance programs associated
with comprehensive medical and psychosocial services
for the pregnant woman are of additional benefit. Methadone maintenance has been shown to be associated with
higher birthweight in some but not all studies (Brown
et al, 1998; Kandall and Doberczak, 1999). Detoxification of a pregnant heroin user is infrequently attempted,
because maternal drug withdrawal is believed to be associated with subsequent fetal withdrawal, fetal asphyxia, and
spontaneous abortions (Barr and Jones, 1994). Dashe et al
(1998) reported on a small study of opioid-using pregnant
women undergoing safe maternal detoxification. Close to
60% of the women completed detoxification, but almost
30% resumed opioid use. McCarthy et al (1999) showed
that women who reduced their methadone dose during
pregnancy had infants with higher birthweights than a control group who continued on the same or increased methadone dose throughout pregnancy. In the United States,
most pregnant, narcotic-addicted women are treated with
daily methadone rather than a program of detoxification.
Some authorities, however, have urged the reappraisal and
reevaluation of the benefits of methadone maintenance in
pregnancy (Brown et al, 1998; Hulse and O’Neil, 2001),
and there are recent studies comparing methadone with
buprenorphine in pregnancy (Jones et al, 2008; Minozzi
et al, 2008).
In the Netherlands, women enrolled in a methadone
program had higher rates of prenatal care, which were
associated with higher birthweights and reduced prematurity in the offspring (Soepatmi, 1994). When women
in a methadone maintenance program were enrolled
in an enhanced prenatal care program, their infants’
birthweights were significantly larger than those in the
control group of women receiving regular methadone
maintenance during pregnancy (Chang et al, 1992). Others have shown that higher methadone doses are associated with improved head circumference and increased
gestational age at delivery (Hagopian et al, 1996). Using
a metaanalysis design, Hulse et al (1997) found that low
infant birthweight was associated with heroin use and
that birthweights were improved with methadone treatment during pregnancy. These favorable outcomes are
believed to be to the result of a stable intrauterine environment uncomplicated by periods of intoxication and
withdrawal, as well as less stress and better nutrition in
the mother.
Several investigators have found that neonatal withdrawal symptoms, birthweight, length of pregnancy, and
the number of days infants require treatment for abstinence
do not correlate with maternal methadone dosage (Brown
122
PART III
Maternal Health Affecting Neonatal Outcome
et al, 1998; Finnegan, 1991; Madden et al, 1977; Rosen
and Pippenger, 1976). In contrast, others have reported
a correlation between the severity of neonatal withdrawal
and maternal methadone dose (Dryden et al, 2009; Harper
et al, 1977; Maas et al, 1990; Malpas et al, 1995). Studying maternal and neonatal serum levels of methadone does
not help clarify this dilemma. Investigators have found no
correlation between neonatal serum levels of methadone
and the maternal methadone dose at delivery, the maternal serum levels, or the severity of withdrawal symptoms in
the neonates (Harper et al, 1977; Mack et al, 1991). Other
researchers have reported that neonatal signs of withdrawal
correlate with the rate of decline of the neonatal plasma
level during the first few days of life (Doberczak et al, 1993).
There are no definitive guidelines for methadone doses
during pregnancy, and there is continuing controversy
over the most appropriate dose of methadone maintenance during pregnancy. The divergent findings noted
previously have been used to argue either for weaning a
pregnant woman to a low methadone maintenance dose
or for attempting complete maternal detoxification during
pregnancy. Some authorities believe in maintaining high
methadone doses to keep the mother from “chipping” with
additional street drugs, which would put her at risk for
greater complications of pregnancy and for a higher risk of
infections transmitted by intravenous use of drugs (HIV,
hepatitis) or sexually transmitted disease. High-dose methadone maintenance ranges between 60 and 150 mg/day.
Despite reports of the safe detoxification of pregnant
women, there are still concerns that the fetus is placed
at risk during maternal detoxification. The medical management of pregnant women who are addicted to opioids
remains controversial (Christensen, 2008).
There is a growing body of literature promoting breastfeeding for infants of mothers in methadone maintenance
programs as both beneficial and safe. Studies have shown
that breastfed infants tend to have less need for pharmacotherapy for neonatal abstinence syndrome, despite low
and unpredictable levels of methadone in breast milk and
in infant serum. Maternal serum levels, breast milk levels, and infant serum levels do not correlate with maternal
methadone dose (Jansson et al, 2008).
Buprenorphine (Suboxone, Subutex)
Buprenorphine is an alternative opioid substitute that
was first introduced in France in 1996. It is increasingly
being used with or instead of methadone for the treatment
of opioid addiction, because it has fewer autonomic side
effects than methadone as well as improved compliance
and treatment efficacy. Recent trials have compared methadone with buprenorphine treatment during pregnancy
and found no difference in the neonatal outcome or incidence of withdrawal symptoms (Jones et al, 2008; Minozzi
et al, 2008).
Human Immunodeficiency Virus and Other
Viral Infections
Nationwide, intravenous drug abusers are the second largest risk group for HIV infection. Drug abusers also may
be the primary source of infection for non–drug-using
heterosexuals and children (Chamberland and Dondero,
1987). Seventy-five percent of cases of acquired immunodeficiency syndrome (AIDS) in children are perinatally
acquired. The seropositivity rate varies across the country; the rate among female intravenous drug users in New
York City and northern New Jersey is estimated at 50% to
70%, compared with 5% to 20% in California. Heroin and
cocaine addicts often resort to prostitution to support their
habits, and amphetamine and methamphetamine users
often inject drugs several times daily. Alcohol decreases
sexual inhibition, impairs judgment, and increases the
incidence of unsafe sexual activity. Every infant born to a
substance abuser should be evaluated for HIV infection,
and universal precautions should be observed. The American Academy of Pediatrics Committee on Infectious Diseases (2009) recommends rapid HIV testing of any mother
whose HIV status is not known, with appropriate consent
as required by local law.
Intravenous drug use places the woman at risk for multiple infectious complications, including cellulitis, thrombophlebitis, hepatitis, endocarditis, syphilis, gonorrhea,
and AIDS. In a prospective study undertaken in Canada,
a 5-year incidence of HIV seroconversion was 13.4%; the
rate of conversion associated with injection of heroin or
cocaine was 40% higher in women than in men (Spittal
et al, 2002). Opioid abusers are also less likely to receive
prenatal care or to obtain late prenatal care (Bauer, 1999).
Heroin-addicted mothers are often poorly nourished, and
iron-deficiency anemia is more common in pregnant opioid users than in nonusers. Bauer (1999) found that maternal hepatitis infections were fivefold higher in opioid-using
women than in a control group of nonusers.
Hepatitis C virus (HCV) is another chronic infectious
condition that is spread by parenteral exposure to infected
blood and can be perinatally acquired by the newborn.
The most common risk factors for acquiring infection are
injection drug use, having multiple sexual partners, or having received blood products before 1992. The 2009 American Academy of Pediatrics Red Book (American Academy
of Pediatrics Committee on Infectious Diseases, 2009)
states that “seroprevalence [of hepatitis C] among pregnant women in the United States has been estimated at 1%
to 2%,” and the risk of perinatal transmission averages 5%
to 6% from women who are HCV-RNA positive at the
time of delivery. Maternal coinfection with HIV has been
associated with increased risk of perinatal transmission
of HCV. Antibodies to HCV and HCV RNA have been
detected in colostrum, but the risk of HCV transmission is
similar in breastfed and bottle-fed infants, so breastfeeding
is currently allowed.
NEONATAL MANAGEMENT AFTER
GESTATIONAL SUBSTANCE ABUSE
GENERAL
Although at higher risk for medical complications, the
majority of infants of drug-using women do not require
intensive neonatal care; however, symptomatic infants
often need more nursing care. Physical examination on
admission should document a gestational age assessment,
birthweight, head circumference, and length. Infants
CHAPTER 12 Perinatal Substance Abuse
should be examined carefully for evidence of malformations, dysmorphic facial features, or both. Studies such as
electroencephalography and brain imaging may add diagnostic or prognostic information when physical or neurologic abnormalities are not clearly consistent with drug
exposure, but these procedures are not indicated for most
drug-exposed infants. If indicated by neonatal or maternal
risk indicators, toxicology testing should be performed on
neonatal urine, meconium, or both as soon as possible after
birth. Infants whose mothers were not screened for HIV
should undergo screening for perinatal HIV exposure and
other infections such as hepatitis and syphilis, as clinically
indicated. In some states, rapid testing of the newborn for
HIV is “required by law if the mother refuses to be tested,”
so that appropriate treatment of the infant can be started
before 12 hours of age. Rapid screening detects only HIV1, the most common serotype of HIV in the United States,
but it can miss HIV-2, so measurement of HIV antibodies
should also be performed (American Academy of Pediatrics Committee on Infectious Diseases, 2009).
Cocaine-exposed infants weighing more than 1500 g
have longer hospital stays and increased need for therapies,
procedures, intravenous fluid, and formula feeding. These
infants also undergo more investigations for sepsis, more
neonatal intensive care unit (NICU) admissions, and more
social and family problems delaying discharge (Bada et al,
2002). In the Maternal Lifestyle Study, cocaine-exposed
infants had a higher frequency of infection (odds ratio
[OR], 3.1; 99% confidence interval [CI], 1.8 to 5.4) and
neurologic signs and symptoms (adjusted OR, 1.7; 99%
CI, 1.0 to 2.1). Neurologic signs were highest in the infants
exposed to opioids and cocaine, but remained significantly
increased in infants exposed to cocaine alone. Smoking
also increased the risk for neurologic signs and symptoms
(Shankaran et al, 2007). The association between cocaine
exposure and fetal hypoxic ischemic episodes creates special concerns. Maternal cocaine use exposes infants to a
higher than expected risk of problems with postasphyxial
syndrome, and organ dysfunction from hypoxic-ischemic
injury should be investigated and treated. Feedings in
premature infants with cocaine exposure should be started
cautiously, because premature infants exposed to cocaine
may be at increased risk for necrotizing enterocolitis. In
addition, after controlling for gender, gestational age,
birthweight, maternal parity, ethnicity, and polydrug
use, heavy cocaine use during pregnancy was associated
with a slightly higher risk of subependymal hemorrhage
(Shankaran et al, 2007).
Using the NICU Network Neurobehavioral Scale
(NNNS), subtle differences in behavior were detected
in drug-exposed infants (Lester et al, 2002; Smith et al,
2008). Cocaine-exposed infants showed lower arousal,
and with heavy cocaine use they showed lower regulation and higher excitability than did unexposed infants
(Lester et al, 2002). There were no stress or abstinence
signs associated with cocaine exposure. Marijuana use
was associated with more stress and abstinence signs
and higher excitability scores (Lester et al, 2002).
Low birthweight was also significantly correlated with
poorer regulation and higher excitability. Methamphetamine exposure was also associated with increased
stress signs, especially in first-trimester use. Heavy
123
methamphetamine use was related to lethargy, lower
arousal, and increased physiologic stress (Smith et al,
2008). Neonatal neurologic abnormalities similar to a
mild withdrawal syndrome, consisting of hypertonicity, irritability, and jitteriness, have been reported after
in utero marijuana exposure, but without documented
evidence of long-term sequelae (Cornelius et al, 1995).
Finally, gestational nicotine exposure definitely altered
the newborn neurobehavioral scores and has also been
reported to elevate neonatal abstinence scores (Godding
et al, 2004; Law et al, 2003).
In the Maternal Lifestyle Study, only 100 women were
identified as isolated users of opioids, and a similar number used cocaine and opioids. Transient but dramatic neurobehavioral signs are present in the first week of life as
symptoms of opioid withdrawal (increased irritability, jitteriness, poor feeding, sweating, sneezing; Shankaran et
al, 2007). See the discussion under Neonatal Abstinence
Syndrome.
BREASTFEEDING AND DRUG EXPOSURE
Breastfeeding has the benefit of improved bonding, but
the risks of HIV and continued drug exposure may outweigh this benefit. Women who wish to breastfeed despite
these potential risks should undergo drug monitoring and
sequential HIV antibody testing. Close observation of
mother-infant interactions should be documented in the
infant’s chart. Parenting and childcare skills should be
stressed as part of the discharge education for the mother.
All physician interactions with the family should be documented in detail.
Most illicit drugs of abuse that are of low molecular
weight and lipophilic, are readily excreted in breast milk,
but have varying degrees of bioavailability (Howard and
Lawrence, 1998). Cocaine has been detected in breast
milk and Chasnoff et al described a two-week-old breastfed infant who had clinical signs of cocaine intoxification.
Both the baby’s urine and the mother’s milk contained
cocaine (Chasnoff et al, 1987). A widely-referenced report
of cocaine seizures in a breastfed infant actually resulted
from topical coacine applied to sore nipples, not from
cocaine-laced breast milk (Chaney et al, 1988). Because of
the potential risk of toxicity, breastfeeding is generally discouraged in women who are known abusers of these drugs
and who are not willing to engage in substance abuse treatment and monitoring. Breastfeeding may be supported for
women who are engaged in substance abuse treatment
and who have received good prenatal care with a confirmed period of sobriety prior to delivery. Breastfeeding
by women using methadone is recommended. Concentrations of methadone in human milk are low, and there
are other significant advantages for the mother and infant
(Jansson et al, 2008).
Alcohol use while breastfeeding is not listed as a contraindication by the American Academy of Pediatrics, but
excessive maternal alcohol intake during breastfeeding can
be deleterious for the infant and should be avoided. Smoking in the postnatal period and during breastfeeding also
has deleterious effects on the newborn. Smoking is associated with measurable levels of nicotine and cotinine in
maternal breast milk.
124
PART III
Maternal Health Affecting Neonatal Outcome
NEONATAL ABSTINENCE SYNDROME
Clinical Findings
Classic neonatal withdrawal or abstinence syndrome consists of a wide variety of CNS signs of irritability, gastrointestinal and feeding problems (diarrhea, hyperphagia
or poor feeding), autonomic signs of dysfunction (fever,
sweating, sneezing), and respiratory symptoms (Tables
12-3 and 12-4). These symptoms are most often related to
gestational opioid exposure, but are relatively nonspecific,
with the differential diagnosis including infection, meningitis, hypocalcemia, hyponatremia, intracranial hemorrhage, seizures, and stroke. The signs of neonatal serotonin
syndrome (or selective serotonin reuptake inhibitor
withdrawal) may also mimic the signs of neonatal opioid
abstinence syndrome (Boucher et al, 2008; Moses-Kolko
et al, 2005). The timing of withdrawal signs from specific drug exposures can often be anticipated; for example, heroin withdrawal usually occurs within 24 hours of
birth, whereas methadone withdrawal symptoms typically
TABLE 12-3 Clinical Signs of Neonatal Withdrawal Syndrome
(Narcotic Abstinence Syndrome)
Central nervous system
dysfunction
Excoriation (from frantic
movement)
Hyperactive reflexes
Increased muscle tone
Irritability, excessive crying,
high-pitched cry
Jitteriness, tremulousness
Myoclonic jerks
Seizures
Sleep disturbance
Autonomic dysfunction
Excessive sweating
Frequent yawning
Hyperthermia
Respiratory symptoms
Nasal stuffiness, sneezing
Tachypnea
Gastrointestinal and feeding
disturbances
Inadequate oral intake
Diarrhea (loose, watery,
frequent stools)
Excessive sucking
Hyperphagia
Regurgitation
These signs were abstracted from Finnegan L, Connaughton J, Kron R: Neonatal
abstinence syndrome: assessment and management, Addict Dis 2:141-158, 1975.
begin later, at approximately 48 to 72 hours after birth.
The incidence of neonatal abstinence syndrome (NAS) in
infants of women using heroin or methadone is high, with
wide ranges reported between 16% and 90% (Agarwal
et al, 1999; Boer et al, 1994; Maas et al, 1990; van Baar
et al, 1994), and between 30% and 91% of infants with
signs of NAS receive pharmacologic treatment for NAS
with inpatient stays averaging 3 weeks (Dryden et al, 2009;
Kuschel, 2007). Premature infants generally have milder
signs of withdrawal and often show alternating periods of
hyperactivity and lethargy, with tremors seen less commonly. The mortality rate for these infants is less than 1%
(Boer et al, 1994). Death is rarely associated with withdrawal alone, but usually occurs as a consequence of prematurity, infection, and severe perinatal asphyxia.
A number of evaluation tools are used to assess the severity of opioid withdrawal after birth. The neonatal abstinence
score is a scale based on nursing observations of the severity
of signs of withdrawal (Finnegan et al, 1975) and is the most
widely used scale. The Lipsitz score was developed at the
same time and is simpler to use, with a score greater than 4
indicating withdrawal. Green and Suffet (1981) introduced
the Neonatal Narcotic Withdrawal Index as a rapid physician-based evaluation for neonatal signs of withdrawal. The
use of these scoring systems allows more objective quantification of the severity of the infant’s withdrawal and the
response to treatment. These scoring systems have shown
good interobserver reliability and can improve clinicians’
ability to treat the withdrawing infant appropriately (Anand
and Arnold, 1994; Franck and Vilardi, 1995).
The goal of medical management of opioid withdrawal
is to avoid serious symptoms of NAS, such as seizures, and
to maintain the infant’s comfort while enabling the infant
to feed, sleep, and gain weight in an appropriate manner.
There are a number of reported threshold scores for initiating pharmacologic treatment (Finnegan NAS scores
between 7 and 12) (Kuschel, 2007), but none of these
choices have been examined in a scientific manner. The
decision to begin treatment or to wean treatment should be
influenced by the absolute score and other factors, such as
the infant’s age, comorbidities, other conditions leading to
abnormal behavior, and a daily evaluation of the abnormal
clinical elements observed in the scoring system (Kuschel,
2007). Standard medical practice is to combine both developmental and behavioral methods with pharmacologic
interventions as necessary to control symptoms and signs
of narcotic abstinence.
TABLE 12-4 Neonatal Neurobehavioral Symptoms after Fetal Drug Exposure
Drug
Alcohol
Onset (days)
Peak (days)
0-1
1-2
Duration
Relative Severity
1-2 days
Mild
Likely NICU Admission
No
Symptoms
?
Amphetamine
0-3
—
2-8 wk
Mild
No
Neuro
Cocaine
0-3
1-4
? mo
Mild
No
Neuro
Heroin
0-3
3-7
2-4 wk
Mild to severe
Yes
Neuro, Resp, GI
Methadone
3-7
10-21
2-6 wk
Mild to severe
Yes
Neuro, Resp, GI
SSRI
0-3
1-3
2-10 days
Mild to moderate
No
Neuro, Resp, GI
Tobacco
0-1
1-2
2-3 days
Mild
No
Neuro
GI, Gastrointestinal symptoms including poor weight gain; Neuro, neurobehavioral symptoms; NICU, neonatal intensive care unit; Resp, respiratory symptoms; SSRI, selective serotonin reuptake inhibitor.
CHAPTER 12 Perinatal Substance Abuse
Treatment
Treatment for opioid withdrawal should always begin with
supportive, nonpharmacologic measures such as soothing
including swaddling, rocking, decreased environmental
stimulation, avoiding unnecessary handling and irritation,
and progressing to pharmacologic management only when
medically necessary. Between 30% and 91% of infants
exhibiting signs of NAS will receive pharmacologic treatment (Kuschel, 2007). The mainstay of treatment for
opioid withdrawal is the use of opioids, either alone or in
combination with other medications (American Academy
of Pediatrics Committee on Drugs, 1998; Osborn et al,
2002, 2005). Medication is titrated for each infant according to the severity of the signs of withdrawal and abstinence scoring.
A recent survey in the United States showed that opioids
are the most common medication used for narcotic withdrawal, and phenobarbital is frequently used or added for
polydrug exposure (Sarkar and Donn, 2006). A metaanalysis of seven studies found that opioid treatment reduced
the time to regain birthweight and the duration of supportive care compared with supportive care alone; however, the length of hospital stay was increased (Osborn et
al, 2005). Phenobarbital was also shown to be superior to
diazepam for treating NAS, but small, randomized, controlled trials comparing morphine to phenobarbital for
treatment of NAS suggested that opioids are superior at
decreasing treatment duration and lowering NAS scores
(Ebner et al, 2007; Jackson et al, 2004; Osborn et al, 2002).
Morphine is the opioid most often used in treating
NAS. The only study comparing an oral preparation of
morphine to dilute tincture of opium (DTO), a concentrated morphine solution that also contains alcohol, recommends oral morphine to avoid problems with dilution
of highly concentrated DTO and the unwanted effects of
the alcoholic extracts with various alkaloids (Langenfeld
et al, 2005). A standard starting dose of morphine is 0.04
to 0.05 mg/kg given orally every 3 to 4 hours. This dose
can be increased in increments of 0.05 to 0.1 mg until the
symptoms are controlled. The usual dose for infants experiencing withdrawal at birth ranges from 0.08 to 0.2 mg
every 3 to 4 hours (American Academy of Pediatrics Committee on Drugs, 1998; Anand and Arnold, 1994; Burgos
and Burke, 2009; Levy and Sino, 1993). Higher doses may
be needed to control significant physiologic signs of withdrawal such as diarrhea, pyrexia, hypertension, and significant hypertonicity.
Although preparations of oral morphine are most commonly used in recent studies, methadone has also been
reported as an option for treatment of NAS (Burgos and
Burke, 2009; Kuschel, 2007). There is one retrospective
report of methadone use for neonatal abstinence syndrome
(Lainwala et al, 2005) and more extensive experience with
methadone in the treatment of opioid withdrawal in older
infants and children (Tobias, 2000; Tobias et al, 1990).
Methadone has a long duration of action and can be
administered by either the oral or the parenteral routes.
The initial recommended methadone dose is 0.05 to 0.1
mg/kg followed by 0.025 to 0.05 mg/kg every 4 to 12 hours
until abstinence scores are controlled. The total daily dose
given to control symptoms is then divided into two doses
125
and given every 12 hours. Tobias et al (1990) showed that
methadone could be given every 12 to 24 hours because of
its longer half-life. There were no advantages to methadone over morphine in neonatal abstinence (Lainwala
et al, 2005). In addition, sublingual buprenorphine has
been reported as a treatment for NAS (Kraft et al, 2008).
Phenobarbital has been used for signs of acute opioid
withdrawal. Phenobarbital does not, however, reduce significant physiologic signs of withdrawal, such as diarrhea
and seizures. At higher doses, phenobarbital has also been
shown to impair infant sucking and cause excessive sedation. The doses of phenobarbital used are 5 to 20 mg/kg
in the first 24 hours, followed by 2 to 4 mg/kg every 12
hours; the therapeutic blood level of phenobarbital for
control of opioid withdrawal signs is not known. Combining oral morphine solution (i.e., DTO) and phenobarbital
treatment was found to shorten duration of hospitalization and lessen the severity of withdrawal symptoms, compared with morphine treatment alone (Coyle et al, 2002,
2005). Compared with those treated with morphine alone,
infants treated with morphine and phenobarbital were
more interactive, had smoother movements, were easier
to handle, and were less stressed. Dual treatment resulted
in improved neurobehavioral organization during the first
3 weeks of life, which may indicate a more rapid recovery
from opioid withdrawal (Coyle et al, 2005). In these studies, however, the infants were discharged home on phenobarbital therapy, from which they were slowly weaned
throughout infancy. Some of these infants received phenobarbital for prolonged times.
Clonidine, an α2-adrenergic receptor antagonist, is used
treating opioid withdrawal symptoms in older children
and adults. A recent randomized, controlled trial showed
that adding clonidine to opioid treatment (i.e., DTO)
significantly reduced the median length of therapy with
opioids and the number of infants requiring high dose opioids, and it eliminated infants who met their definition of
treatment failure (Agthe et al, 2009). During hospitalization, there were no significant adverse events (i.e., hypertension, hypotension, bradycardia, desaturations) related
to clonidine use. A German group has also retrospectively
reported their experience using clonidine and chloral
hydrate for NAS compared to a larger group treated with
morphine and phenobarbital; they also reported decreased
duration of treatment, decreased length of stay, and reduction of symptoms in the clonidine group (Esmaeili et al,
2010). Before clonidine is used widely, a larger trial is indicated to investigate appropriate dosing regimens and to
determine long-term safety.
Once medications have been titrated to a level that controls the severity of opioid withdrawal and lowers the NAS
scores, then tapering of the dosage should be started. A
common method is to decrease the opioid dose by 10%
to 20% of the highest dose, with continued surveillance of
NAS scores to assure the infant tolerates the decrease. It
is not unusual to note increased signs of opioid withdrawal
during the medication tapering. The goal of weaning is to
allow the infant to acclimate to a new and lower dose of
medication while ensuring that he or she is comfortable
and consolable and is able to sleep, eat, and gain weight
appropriately. Objective measurements using established
withdrawal scoring systems should be used to determine
126
PART III
Maternal Health Affecting Neonatal Outcome
the rate and efficacy of medication tapering. Most often
opioid medications are weaned every 24 to 48 hours as
long as NAS scores remain low and the infant’s clinical
condition is unchanged (Burgos and Burke, 2009; Coyle
et al, 2002; Jackson et al, 2004). Whether narcotics can
safely be weaned more rapidly in infants has not been
studied. One small study reported good results using oral
morphine on an as-needed basis for elevated individual
withdrawal scores, rather than providing regular doses
of morphine every 3 to 4 hours (Ebner et al, 2007). No
specific protocols for weaning phenobarbital or clonidine
have been reported.
The average length of hospital stay for infants with
NAS who are treated with medications varies from 8 to
78 days, with a stay of 21 to 30 days being common (Coyle
et al, 2002; Ebner et al, 2007; Jackson et al, 2004; Kuschel,
2007). Investigation of outpatient management of detoxification may result in a shorter hospital stay, but with
a more prolonged duration of neonatal treatment (Kuschel,
2007).
LONG-TERM EFFECTS OF PERINATAL
SUBSTANCE ABUSE
non–cocaine-exposed infants at birth, but there were no
differences in weight between ages 1 and 6 years. For
height, cocaine-exposed infants were shorter at birth
through 2 years, but the difference disappeared by age 3,
and cocaine-exposed infants had a smaller head circumference from birth to 1 year old, but this difference subsequently disappeared (Shankaran et al, 2007). A previous
systematic review concluded that there is no consistent
effect of cocaine exposure on physical growth in children
younger than 6 years (Frank et al, 2001). Opioid-exposed
infants also show no difficulties with postnatal growth
through 3 years (Hunt et al, 2008). Infants with IUGR at
birth remained significantly lighter (by 2.1 kg) and shorter
(by 1.8 cm) at 6 years, and they continued to have a smaller
head circumference (by 0.9 cm). There was no interaction
between cocaine exposure and IUGR status (Shankaran et
al, 2007).
There were also no differences in medical diagnoses,
blood pressure, hospitalizations, or overall health status
associated with opioid exposure (Shankaran et al, 2007).
The effect of cocaine on subsequent blood pressure is
unclear. IUGR status at birth was significantly associated
with hypertension after correcting for in utero drug exposures, maternal race, and child’s body mass index.
SUDDEN INFANT DEATH SYNDROME IN
DRUG-EXPOSED INFANTS
NEUROBEHAVIORAL ABNORMALITIES
The incidence of sudden infant death syndrome (SIDS)
is greater in drug-exposed infants, although the increase
appears less significant for cocaine-exposed infants (Chasnoff et al, 1989b; Kandall et al, 1993) than for methadone-exposed or heroin-exposed infants (Bauchner and
Zuckerman, 1990). A metaanalysis of 10 studies demonstrated a 4.1 OR for SIDS among cocaine-exposed infants
(Fares et al, 1997) compared with infants not exposed to
perinatal drugs. However, after data were controlled for
concurrent use of other drugs, the increased risk for SIDS
could not be attributed to intrauterine cocaine alone, but
was believed to be caused by exposure to other illicit drugs
and smoking.
There is a significant relationship between exposure to
tobacco smoke and SIDS. A higher risk for SIDS is reported
in infants exposed to maternal smoking with either antenatal or postnatal exposure (Blair et al, 1996; Mitchell et al,
1993; Schoendorf and Kiely, 1992). Additional evidence
supporting a causal role includes a dose-response relationship (Rogers, 2008). With recent substantial decreases in
SIDS risk (after “back to sleep”), maternal smoking is calculated to account for an increasing proportion of SIDS
risk. Mitchell and Millerad (2006) suggest that one third
of all SIDS deaths might be prevented by eliminating in
utero exposure to maternal smoking. Despite the increased
risks of SIDS among drug- and tobacco-exposed infants,
home apnea monitoring is not indicated in the absence of
other risk factors.
POSTNATAL GROWTH AND MEDICAL
PROBLEMS
There are no documented long-term effects of substance
exposure on postnatal growth. In the Maternal Lifestyle Study, cocaine-exposed infants were lighter than
Detailed studies have identified both neurologic soft signs
and a higher risk of cognitive, behavioral, and psychiatric
problems linked to gestational substance exposure. However, significant confounding factors make it difficult to
attribute specific deficits directly to substance exposures.
No major neurologic deficits in motor development are
found after in utero exposure to cocaine (Shankaran
et al, 2007) or opioids (Hunt et al, 2008). Infants exposed
to cocaine were reported to have lower motor skills at
1-month testing, but they displayed significant improvements over time. Both higher and lower levels of tobacco
use were related to poor motor performance (Shankaran
et al, 2007).
Subtle neurobehavioral abnormalities have been
reported in older studies and more recently using the
NNNS to evaluate infants in the month after birth.
Cocaine-exposed infants manifest a range of neurobehavioral abnormalities that were initially described as drug
withdrawal, but are more likely caused by acute intoxication (Dempsey et al, 1996). Signs are present at birth
and wane as cocaine and the metabolite, benzoylecgonine,
are cleared from plasma. The infants are hypertonic, irritable, and tremulous (Chiriboga, 1993), and they may have
abnormal crying, sleep, and feeding patterns. Tachycardia, tachypnea, and apnea have been noted in two blinded,
controlled studies, with significant elevations in cardiac
output, stroke volume, mean arterial blood pressure, and
cerebral artery flow velocity resolving by day 2, which is
consistent with an intoxicant effect of cocaine (van de Bor
et al, 1990a, 1990b) Cocaine-exposed infants may have
abnormal electroencephalograms or clinical seizures, perhaps the result of toxicity from the metabolite, benzoylecgonine (Konkol et al, 1994); however, neonatal seizures
attributable directly to maternal use of cocaine are rare
(Legido et al, 1992).
CHAPTER 12 Perinatal Substance Abuse
A number of studies suggest that exposure to exogenous
opioids during fetal development may produce lifelong
alterations in the developing brain. Infants who have been
exposed to opioids in utero have a higher risk for fetal
growth retardation and smaller head circumference than
those who have not (Bauer, 1999). In addition, significant
developmental and learning deficits have been described
in both methadone-exposed and heroin-exposed children
(Soepatmi, 1994; van Baar and de Graaff, 1994; van Baar
et al, 1994), and Bunikowski et al (1998) have reported a
higher incidence of abnormalities in intellectual performance, developmental retardation, and neurologic abnormalities in a group of opioid-exposed infants compared
with a control group of infants. However, the treated
infants in the study had an unusually high incidence of
seizures during withdrawal treatment with phenobarbital
alone, as well as a higher incidence of prematurity than the
control infants. These important considerations temper
the findings of that study.
Early neurobehavioral changes have been associated
with maternal methamphetamine use; heavy use was associated with lower arousal, increased lethargy, and increased
CNS stress (Smith et al, 2008). However, a detailed analysis in a subgroup of the same population found that these
same abnormalities on the NNNS were associated with
maternal depression and that prenatal methamphetamine
exposure was not associated with additional neurodevelopmental differences (Paz et al, 2009).
Persisting behavioral, neurologic, and rearing problems
are reported in children exposed to both cocaine and opioids (Chasnoff et al, 1989; Hunt et al, 2008). No significant
differences in mean developmental scores were noted in a
group of children exposed to cocaine plus polydrugs compared to a group without drug exposure (Chasnoff et al,
1992). Other investigators reported no differences between
infants who have and those who have not been exposed
to cocaine in mean cognitive, psychomotor, or language
quotients at age 36 months (Kilbride et al, 2000). A recent
report following a cohort of opioid exposed infants documented lower Bayley mental development index (MDI)
scores at 18 months old, lower Stanford Binet Intelligence
Scale scores at 3 years, and decreased scores on the Vineland Social Maturity Scale and Reynell Language Scale at
3 years (Hunt et al, 2008). However, a similar evaluation
at 3 years showed no mental, motor, or behavioral deficits
after controlling for birthweight and environmental risk
factors (Messinger et al, 2004).
The neurodevelopmental problems among children
exposed to cocaine may occur either from a direct teratogenic drug effect during gestation or from the effects of
the social environment in which the developing infant is
reared. Singer et al (2002) reported that cocaine-exposed
infants are twice as likely to have significant cognitive
but not motor delays at 2 years, and they demonstrated a
downward trend in mean developmental scores by 2 years,
which is consistent with a deleterious effect of the environment, parental stimulation, socioeconomic status, or
possibly other, indirect effects of drugs on the developing
CNS. Other studies have not consistently demonstrated
this association (Frank et al, 2001). In contrast, the Maternal Lifestyle Study, after controlling for covariates, found
no differences in Bayley scores between cocaine-exposed,
127
opioid-exposed, and control infants for the first 3 years
of life (Shankaran et al, 2007), although subtle effects on
cognitive subscales were reported at 3 years (Messinger
et al, 2004).
Other investigators have shown normal development
for opioid-exposed infants during the first 2 years of life
after data have been controlled for socioeconomic status
and birthweight (Bauer, 1999). Using regression analysis,
researchers have shown that the amount of prenatal care
obtained by the mother and the postnatal home environment were more predictive of the infant’s future intellectual performance. Conversely, the amount of maternal
opioid use during pregnancy was not found to be predictive. Ornoy et al (2001) have shown that children exposed
to heroin but adopted at an early age performed better
on intelligence testing than did opioid-exposed infants
who were raised in the homes of their biologic parents.
The same investigators found a higher rate of attention
deficit disorders in children exposed to opioids regardless of home environment, with the highest incidence in
children who were raised in the homes of their biologic
parents. Numerous studies point also to the importance of
the home environment in optimizing child development.
Variable outcomes of these studies may depend on the
amount and type of drug use, or on other covariates such
as nutritional status, poverty, psychosocial problems, and
parental educational level.
With the lack of major neurologic deficits, recent studies have focused on neurologic soft signs, nonfocal signs
with no localized findings that are more often associated
with cognitive deficits, and increased prevalence of attention deficit hyperactivity disorder and behavior problems.
Neurologic soft signs including speech, balance, coordination, and tone were markedly stable over a 1-year period
in 6- to 9-year-olds. Soft signs are a marker for increased
risk of cognitive and psychiatric problems. In the Maternal Lifestyle Study, 23.5% of children exposed to cocaine
had more than two soft neurologic signs, similar to rates
in the comparison group. Soft signs were more common
in infants born weighing less than 1500 g. Both cocaine
and alcohol use significantly increased the incidence of soft
signs in exposed infants weighing less than 1500 g, and this
effect persisted after controlling for other substances used,
birthweight, sex, and race (Shankaran et al, 2007).
Childhood behavior problems were more common in
infants with heavy cocaine exposure than in those with no
or some cocaine exposure. Prenatal tobacco and alcohol
exposure were also significantly associated with behavioral
problems until 7 years, with a significant dose-response
relationship (Shankaran et al, 2007). Evaluation at 7 years
demonstrated behavior problems in cocaine- and substance-exposed infants that suggested direct effects resulting in neurobehavioral dysregulation, which was tracked
through serial assessments beginning at an early age. This
work on a model of early abnormalities predictive of later
behavioral problems may allow for early identification and
possible prevention of later behavioral problems (Lester
et al, 2009).
Follow-up studies have been reported at 9 to 15
years. Cognitive outcomes at 10 years were not abnormal in opioid-exposed children (Shankaran et al,
2007). Cocaine-exposed children were more likely to
128
PART III
Maternal Health Affecting Neonatal Outcome
be referred for special education services in school than
unexposed children, with an estimated additional cost to
the United States of $25,248,384 per year (Shankaran et
al, 2007). Prenatal marijuana exposure is reported to be
associated with deficiencies in executive function in 9 to
12 year olds (Fried and Smith, 2001; Fried et al, 1998),
and recent reports have found that prenatal marijuana
exposure has a significant effect on school-age intellectual performance (Goldschmidt et al, 2004, 2008;
Richardson et al, 2002).
In methamphetamine-exposed infants, neurodevelopmental abnormalities have been described to persist as
late as 14 years (Cernerud et al, 1996). Intellectual capacity does not appear to be diminished among exposed
infants. These children are described as exhibiting disturbed behavior, including hyperactivity, aggressiveness,
and sleep disturbances. Eriksson et al (2000) reported
that neurobehavioral abnormalities appear to be associated with the extent and duration of fetal exposure and
with the severity of head growth restriction. In this study,
children with the most severe problems were those born
to mothers who abused amphetamines throughout pregnancy and were reared in homes with an addicted parent.
Alterations in growth have been reported after prenatal
exposure, with striking gender differences (Cernerud et
al, 1996). Drug-exposed boys in Sweden were taller and
heavier, and girls were smaller and lighter, than national
standards. This finding suggests that fetal amphetamine
exposure affects the onset of puberty and amphetamines
may interfere with neurodevelopment of the adenohypophysis. Children of amphetamine abusers appear to be
at high risk for social problems, including abandonment,
abuse, and neglect. In two Swedish studies, only 22% of
10-year-old children who had been exposed to amphetamine in utero remained in the care of their biologic
mothers, whereas 70% were in foster care (Cernerud et al,
1996; Eriksson and Zetterstrom, 1994).
Considering all the data, it is still difficult to create a
coherent picture and provide a prognosis for a newborn
after gestational substance exposure. A recent report of
volumetric magnetic resonance imaging in thirty five
12-year-old children exposed to cocaine in utero found
smaller total parenchymal volumes, lower cortical gray
matter volumes, and smaller head circumferences with
prenatal substance exposure. The decreases were statistically significant only for prenatal cigarette exposure and
for infants exposed to all substances studied (cocaine,
tobacco, marijuana and alcohol) (Rivkin et al, 2008). As in
other studies, exposure to multiple substances clearly has
detrimental effects on the developing brain.
SUMMARY
The magnitude of observed perinatal outcomes after
illicit maternal substance use pales in comparison to the
established health and developmental risks associated with
tobacco and alcohol exposure (Schempf, 2007). The greatest impact of illicit substance use may be the increased
postnatal risks of neglect, maltreatment, and disruptions
in the home environment. Health policy must be directed
at reducing all these complex factors associated with perinatal substance abuse.
SUGGESTED READINGS
American Academy of Pediatrics Committee on Substance Abuse and Committee
on Children with Disabilities: Fetal alcohol syndrome and alcohol-related
neurodevelopmental disorders, Pediatrics 106:358-361, 2000.
Burgos AE, Burke BL: Neonatal abstinence syndrome, NeoReviews 10:e222, 2009.
Chasnoff IJ, McGourty RF, Bailey GW, et al: The 4P’s Plus screen for substance
use in pregnancy: clinical application and outcomes, J Perinat 25:368-374,
2005.
Jones HE, Martin PR, Heil SH, et al: Treatment of opioid-dependent pregnant
women: clinical and research issues, J Subst Abuse Treat 35:245-259, 2008.
Messinger DS, Bauer CR, Das A, et al: The Maternal Lifestyle Study: cognitive,
motor, and behavioral outcomes of cocaine-exposed and opiate-exposed infants
through three years of age, Pediatrics 113:1677-1685, 2004.
Mitchell EA, Millerad J: Smoking and the sudden infant death syndrome, Rev
Environ Health 21:81-103, 2006.
Moses-Kolko EL, Bogen D, Perel J, et al: Neonatal signs after late in utero exposure to serotonin reuptake inhibitors, literature review and clinical applications,
JAMA 293:2372-2383, 2005.
Rogers JM: Tobacco and pregnancy: overview of exposure and effects, Birth Defects
Res C Embryol Today 84:1-15, 2008.
Schempf AH: Illicit drug use and neonatal outcomes: a critical review, Obstet
Gynecol Surv 62:749-757, 2007.
Shankaran S, Lester BM, Das A, et al: Impact of maternal substance use during
pregnancy on childhood outcome, Semin Fetal Neonatal Med 12:143-150, 2007.
Smith LM, LaGasse LL, Derauf C, et al: The infant development, environment,
and lifestyle study: effects of prenatal methamphetamine exposure, polydrug
exposure, and poverty on intrauterine growth, Pediatr 118:1149-1156, 2006.
Thompson BL, Levitt P, Stanwood GD: Prenatal exposure to drugs: effects on
brain development and implications for policy and education, Nat Rev Neurosci
10:303-312, 2009.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
P A R T
I V
Labor and Delivery
C H A P T E R
13
Antepartum Fetal Assessment
Christian M. Pettker and Katherine H. Campbell
A primary objective of obstetric care is the assessment
and prevention of adverse fetal and neonatal outcomes.
Maternal care is an integral step toward this goal. Optimization of the maternal state, through careful monitoring
and treatment of chronic conditions such as diabetes or
hypertension or acute states like preeclampsia or preterm
labor, is one important facet of care to achieve desirable
perinatal outcomes. Monitoring and management of the
fetus, although a more obvious step toward this goal, are
somewhat less straightforward. Fetal assessment demands
a view into the intrauterine environment, which is somewhat inaccessible. Our ability to gain access to this space
to gauge the needs and health of the fetus has improved
dramatically with the developments in technology, as well
as the increased understanding of fetal physiology over the
past 50 years. As a result, perinatal morbidity and mortality
have decreased substantially (Figure 13-1).
In general, antepartum fetal assessment encompasses the
screening and diagnosis of fetal disorders and fetuses that
are at risk. Selecting appropriate patients at risk for adverse
perinatal events can enhance the prediction of these events,
although some tests may be appropriate even for a low-risk
population. The assessment may allow for certain therapeutic options—often, timely delivery—to prevent fetal
harm. The overall goal of these efforts is to reduce perinatal mortality, although the reduction of morbidities such
as cerebral palsy or preventable birth injury is intertwined
with this objective. In antenatal assessment in the third trimester, the prediction and detection of fetal acidemia and
hypoxemia form a central principle underlying these efforts.
It is important to make the distinction between antepartum and intrapartum fetal assessment. The latter is specifically related to monitoring the fetus during labor. The
nature of labor affords certain advantages (e.g., dilation
allows blood samples from the fetus) and restrictions (the
lack of fluid after rupture of membranes creates difficulties for ultrasound examination) that do not occur in the
antenatal period. As a result, this chapter focuses only on
events and assessment preceding labor.
GENERAL PRINCIPLES
PRINCIPLES OF TESTING
Many of the tests used for antepartum fetal assessment are
screening tests that will lead to further testing allowing for
diagnosis and decision-making; therefore it is important
to note the principles guiding such tests. The outcome,
principally perinatal morbidity and mortality, is a significant burden to both the individual and the overall health
care system. The primary tools for assessment, ultrasound
examination, and fetal heart rate monitoring, are generally easy, safe, and acceptable to patients. Screening has
the potential to allow important and timely interventions,
such as antenatal steroid administration or delivery. The
predominant difficulty with fetal testing comes in the
unproven utility of testing to improve outcomes. Furthermore, some tests, such as the nonstress test (NST), have
high false-positive rates; therefore, when used as a diagnostic test (e.g., to decide on delivery), they can lead to the
overuse of interventions. The specificity and sensitivity of
the tests vary and the critical step to enhancing test performance is patient selection. The utility of fetal surveillance
involves the judicious application of the tests in patients
with specific risk profiles.
FETAL PHYSIOLOGY AND BEHAVIOR
The first trimester (≤14 weeks’ gestation) is mainly a
time of system development and organogenesis. The
hyperplastic enlargement during the first 11 weeks produces standard rates of growth, with deviation being rare.
At the completion of the first trimester, the major organ
systems have developed, allowing the opportunity during
the second trimester to assess for anomalies in development. The second and third trimesters involve maturation
of these systems. Because antenatal fetal assessment is primarily concerned with the prediction or detection of fetal
hypoxemia and acidemia, the integration of the neurologic
and cardiovascular systems, particularly as reflections of
fetal acid-base status, is the cornerstone of this assessment.
We are thus able to monitor the manifestations of hypoxemia and acidemia as shown by neurologic and cardiovascular changes.
TECHNOLOGY
The technology underpinning fetal assessment is ultrasound. Fetal heart rate monitoring during the antepartum
period depends on a Doppler cardiogram; movements of
the fetal heart, in particular the sounds of the valves, are
detected by this monitor. The time between the beats is
translated into a heart rate, which is then graphically represented on a chart over time. This process produces the
129
130
PART IV Labor and Delivery
35
White FMR
Black FMR
Fetal deaths per 1000 births
30
25
20
15
10
5
0
1950 1960 1970 1980 1985 1988 1989 1990 1991 1992 1993 1994 1995 1996
Year
Cumulative risk of fetal death (percent)
A
100
80
60
40
20
0
28–31
32–36
37–39
40–42
42+
Weeks’ gestation
B
FIGURE 13-1 Fetal mortality rate (FMR) in the United States, 1959 to 1996. (Adapted from McIlwaine GM, Dunn FH, Howat RC, et al: A routine
system for monitoring perinatal deaths in Scotland, Br J Obstet Gynaecol 92:9-13, 1985.)
fetal heart rate monitoring strip that becomes the NST or
contraction stress test (CST).
Contemporary ultrasound technology involves a wide
array of features, including B-mode (basic imaging),
M-mode (mapping the movement of structures over time),
pulsed Doppler (demonstrating flow velocity in a particular
area, such as a vessel), color Doppler (showing intensity and
direction of flow through shades of red and blue), and power
Doppler (a more sensitive form of colorized Doppler).
INDICATIONS AND TIMING
Most fetal testing protocols involve a stepwise approach,
and the first step is the selection of the appropriate patient.
Suggested assessments for low-risk pregnancies include
one ultrasound examination for dating and one for the
basic anatomic survey. Prenatal risk assessments for chromosomal disorders, such as first-trimester risk assessment
with maternal serum analysis and fetal nuchal translucency
assessment or the second-trimester maternal quadruple
serum screen, are additional options. Whereas up to 30%
of perinatal morbidity may occur in low-risk patients, routine fetal testing beyond that described previously in a lowrisk pregnancy is an ineffective use of resources.
High-risk pregnancies are those at greater peril for perinatal morbidity and mortality. These pregnancies often
have more justification for targeted or detailed anatomic
ultrasound examinations and for regular assessment of
fetal growth or heart rate assessment. Common conditions requiring increased fetal surveillance are shown in
Box 13-1.
Pregnancy dating should be confirmed at the earliest
possible moment, and fetal anatomic screening is best
accomplished in the second trimester, specifically at 18 to
20 weeks’ gestation, when visualization of the anatomic
features is adequate. However, standards for the timing
of antepartum fetal assessment to survey for fetal compromise do not exist. Certainly assessment with NST or biophysical profiles would have little utility before viability
(approximately 24 weeks’ gestation). Guidelines for initiating fetal testing for specific indications are largely based on
the risk of fetal loss at a particular gestational age.
CHAPTER 13 Antepartum Fetal Assessment
BOX 13-1 T
ypical Indications for Antepartum
Fetal Assessment: High-Risk
Pregnancies
FETAL
Abnormal fetal testing, fetal distress
ll Anatomic anomaly
ll Decreased fetal movement
ll Heart block
ll Intraamniotic infection
ll Intrauterine growth restriction
ll Multiple gestation
ll Oligohydramnios
ll
MATERNAL-FETAL
Abruptio placenta
ll Anemia, fetal (e.g., parvovirus, Rh alloimmunization, NAIT)
ll Abnormal serum screening (low PAPP-A, high MSAFP)
ll Placenta previa, vasa previa
ll Postterm or postdates premature rupture of fetal membranes
ll Threatened preterm delivery
ll
MATERNAL
Advanced maternal age
ll Cardiac disease (severe)
ll Cholestasis of pregnancy
ll Diabetes, gestational
ll Diabetes, pregestational
ll Hemoglobinopathies
ll HIV (receiving medication)
ll Hypertension, chronic
ll Hypertension, gestational
ll History of IUFD
ll Obesity
ll Preeclampsia
ll Pulmonary disease (severe)
ll Renal disease
ll Seizure disorder
ll Substance abuse
ll Systemic lupus erythematosis
ll Thyroid disease
ll Thrombophilia or thromboembolic disease
ll
IUFD, Intrauterine fetal demise; MSAFP, maternal serum alpha-fetoprotein; NAIT, neonatal
alloimmune thrombocytopenia; PAPP-A, pregnancy associated plasma protein
FETAL ASSESSMENT IN LOW RISK
PREGNANCIES
ULTRASOUND: PREGNANCY DATING
The estimated date of delivery is defined at the beginning
of pregnancy based on the best available information,
including menstrual history, ultrasound data, and assisted
reproduction technology. The median duration of a singleton pregnancy is 280 days (40 weeks) from the first
day of the last menstrual period or 266 days (38 weeks)
from the time of ovulation. Term is defined as 37 to 42
weeks’ (259 to 294 days) gestation. Given that the preterm
and postterm periods are associated with increased risks
to the fetus and newborn, pregnancy dating provides an
approximate expectation for the completion of the pregnancy and serves as a basis for the efficient and appropriate
use of fetal surveillance, testing, and treatment. Accurate
pregnancy dating by ultrasound has been associated with
131
reduced diagnoses of growth restriction (Waldenstrom
et al, 1992), reduced use of tocolysis for preterm labor
(LeFevre et al, 1993), and a reduced need to intervene in
postterm pregnancies (Neilson, 2000).
In a spontaneous pregnancy in a woman with regular
cycles and normal menstrual periods, the last menstrual
period is often an accurate way of dating a pregnancy.
Menstrual dating is less accurate in women who are taking oral contraceptives, were recently pregnant, or have
irregular periods or intermenstrual bleeding. In these
cases, and others in whom there is uncertainty, ultrasound
dating in the first trimester is accurate and effective. A
fetal pole may be seen beginning at 5 weeks’ gestation,
and the fetal heartbeat should be visualized at 6 to 8
weeks’ gestation. In the first trimester, measurement of
the crown-rump length is accurate to within 3 to 5 days;
therefore this measurement should take priority in dating
a pregnancy when the timing of the last menstrual period
suggests a gestational age outside this range of variation
(Drumm et al, 1976; Robinson and Fleming, 1975). A
first-trimester ultrasound examination is indicated to confirm an intrauterine pregnancy (i.e., exclude ectopic pregnancy), confirm fetal viability, document fetal number,
estimate gestational age, and evaluate the maternal pelvis
and ovaries.
In the second trimester, ultrasound dating is less accurate, but can nonetheless be helpful. Measurement of
the biparietal diameter (BPD) of the fetal head, the most
accurate parameter, can be accurate to within 7 to 10 days
(Campbell et al, 1985; Waldenstrom et al, 1990). The
BPD is also a parameter of choice because it is less affected
by chromosomal anomalies, in particular Down syndrome
(Cuckle and Wald, 1987). Usually in the second or third
trimesters, several biometric measurements—such as cerebellar distance, femur and humerus length, and abdominal
circumference—are recorded and a computerized algorithm can generate an estimated gestational age.
ULTRASOUND: SECOND AND THIRD
TRIMESTERS
Perinatal ultrasound examination in the second and third
trimesters can be classified broadly into three types: the
basic or standard examination, the specialized (detailed)
examination, and the limited examination. The standard
examination (level I) includes the determination of fetal
number, fetal viability, fetal position, gestational age, placental location, amniotic fluid volume, the presence or
absence of a maternal pelvic mass, and the presence of
gross fetal malformations (American College of Obstetricians and Gynecologists, 2009). Most pregnancies can
be evaluated adequately by this basic examination. If the
patient’s history, physical examination, or basic ultrasound
examination suggest the presence of a fetal malformation,
a specialized examination (level II) should be performed
by a sonographer who is skilled in fetal evaluation. During a detailed ultrasound, which is best performed at 18
to 20 weeks’ gestation, fetal structures are examined in
detail to identify and characterize any fetal malformation.
In addition to identifying structural abnormalities, a specialized ultrasound examination can identify sonographic
markers of fetal aneuploidy. In some situations, a limited
132
PART IV Labor and Delivery
examination may be appropriate to answer a specific clinical question (such as fetal viability, amniotic fluid volume,
fetal presentation, placental location, or cervical length) or
to provide sonographic guidance for an invasive procedure
(such as amniocentesis).
Current debate centers on who should undergo sonographic examination and what type of evaluation these
patients should have. Advocates of routine sonography
cite several advantages of universal ultrasound evaluation,
including more accurate dating of pregnancy and earlier
and more accurate diagnosis of multiple gestation, structural malformations, and fetal aneuploidy (discussed later
in ultrasound section). Opponents of routine sonographic
examination argue that it is an expensive screening test
($100 to $250 for a standard examination) and that the
cost is not justified by published research, which suggests
that routine ultrasound examinations do not significantly
change perinatal outcome (Crane et al, 1994; Ewigman
et al, 1993; LeFevre et al, 1993)
Second-trimester ultrasound examination is indicated
in patients with uncertain dating, uterine size larger or
smaller than expected for the estimated gestational age, a
medical disorder that can affect fetal growth and development (e.g., diabetes, hypertension, collagen vascular disorder), a family history of an inherited genetic abnormality,
or a suspected fetal malformation or growth disturbance
(American College of Obstetricians and Gynecologists,
2009). In the United States, most patients undergo a standard examination at 18 to 20 weeks’ gestation to screen
for structural defects. An understanding of normal fetal
physiology is critical to the diagnosis of fetal structural
anomalies. For example, extraabdominal herniation of
the midgut into the umbilical cord occurs normally in the
fetus at 8 to 12 weeks’ gestation and can be misdiagnosed
as an abdominal wall defect. Placental location should be
documented with the bladder empty, because overdistention of the maternal bladder or a lower uterine contraction
can give a false impression of placenta previa. If placenta
previa is identified at 18 to 22 weeks’ gestation, serial ultrasound examinations should be performed to follow placental location. Only 5% of placenta previa identified in the
second trimester will persist to term (Zelop et al, 1994).
The umbilical cord should also be imaged, and the number
of vessels, placental insertion, and insertion into the fetus
should be noted.
The indications for third-trimester ultrasound examination are similar to that for second-trimester ultrasound.
Fetal anatomy survey examinations and estimates of fetal
weight become less accurate as gestational age increases,
especially in obese women or in pregnancies complicated
by oligohydramnios. However, fetal biometry and an anatomic survey should still be performed, because certain
fetal anomalies, such as achondroplasia, will become evident for the first time later in gestation.
FETAL MOVEMENT COUNTING
Fetal movement (quickening) is typically perceived by the
mother at 16 to 22 weeks’ gestation. Fetal hypoxemia is
typically associated with a reduction in fetal activity; the
fetus is essentially conserving energy and oxygen for vital
activities. A typical procedure for fetal movement counting
consists of having the patient record the interval taken to
feel 10 fetal movements, usually after a meal when the
fetus is more active. If 10 movements are not detected in
1 hour, further testing is often recommended. The data
supporting fetal movement counting are mixed. A large
international cluster randomized trial involving more
than 68,000 patients demonstrated no benefit (Grant et al,
1989), and a Cochrane analysis found insufficient evidence
to support this technique to prevent stillbirth (Mangesi
and Hofmeyr, 2007). Fetal movement counting represents
a low-technology screening test that can be applied easily
to all pregnancies. Although its effectiveness in improving
perinatal outcomes is debatable, it can be used as a costeffective first line strategy
FETAL ASSESSMENT IN HIGH-RISK
PREGNANCIES
CARDIOTOCOGRAPHY
Cardiotocography is the visual representation of fetal
heart rate and uterine contractions. Fetal heart rate has
been recognized as an important indicator of fetal status
since the nineteenth century, with Lejumeau Kergaradec
of Switzerland being credited with the first accounts of
direct fetal auscultation and the uterine soufflé in 1821.
Fetal heart rate monitoring is based on the principle that
the fetal neurologic system, through its afferent and efferent networks, serves as a key mediator to demonstrate fetal
well-being. Oxygenation, acidemia, and other vital functions are monitored by peripheral chemoreceptors and
baroreceptors, which provide input on fetal status through
afferent neurologic networks to the central nervous system (CNS). This information is processed by the CNS,
and signals are conducted through efferent networks to
produce peripheral changes, particularly to the heart via
direct parasympathetic vagal neurons, direct sympathetic
signals, or indirect sympathetic stimulation of catecholamine release. In this way, fetal cardiac activity can be seen
as a surrogate for fetal oxygenation and acid-base status.
For many years, assessment of the fetal heart rate was
limited to the fetoscope, a direct stethoscope attributed to
Adolphe Pinard in 1876. In 1957, Orvan Hess and Ed Hon
at Yale University introduced electronic fetal heart rate
monitoring as a window into the status of the fetus (Hon
and Hess, 1957). This technology relied on direct monitoring through a scalp electrode; only years later would
Doppler technology allow cardiac signals to be detected
noninvasively. Fetal heart rate monitoring became a tool
for fetal assessment as it was recognized that certain fetal
heart rate patterns were associated with fetal compromise
and poor fetal outcomes.
The basic elements of a fetal heart rate strip are baseline, variability, accelerations, and decelerations. A baseline of 110 to 160 beats per minute is normal. Variability is
determined by the irregular fluctuations in amplitude and
frequency in the baseline, and variability of fewer than 6
beats per minute is often abnormal. Accelerations are classified as visually apparent abrupt increases that peak at 15
beats per minute or more above the baseline that last 15
seconds or longer. Fetal movements often coincide with
fetal heart rate (FHR) accelerations. Finally decelerations,
CHAPTER 13 Antepartum Fetal Assessment
133
TABLE 13-1 Interpretation of Antepartum Cardiotocography
Term
Characteristic
Description
Baseline
Definition
Mean fetal heart rate, rounded to increments of 5 beats/min (e.g. 140, 145); need baseline duration
of ≥2 min during a 10-min segment, between periodic or episodic changes, to determine baseline
Bradycardia
<110 beats per minute for >10 min
Tachycardia
>160 beats per minute for >10 min
Definition
Fluctuations of the baseline heart rate; measured from peak to trough
Absent
Undetectable
Minimal
Undetectable to ≤5 beats/min
Moderate
6-25 beats/min
Marked
>25 beats/min
Definition
Abrupt increase ≥15 beats/min lasting ≥15 s
Variability
Acceleration
Deceleration
Prolonged
≥2 min and <10 min (≥10 min is a baseline change)
Definition
Decreases in the fetal heart rate
Variable
Abrupt decrease onset to nadir <30 s; decrease ≥15 beats/min lasting ≥5 s to <2 min
Early
Late
Prolonged
Contractions
Gradual decrease onset to nadir ≥30 s with contraction
Gradual decrease onset to nadir ≥30 s; nadir of deceleration occurring after peak of contraction
Decrease ≥15 beats/min lasting ≥2 min, but <10 min (≥10 min is a baseline change)
Recurrent
Occur with ≥50% of uterine contractions in any 20-min window
Intermittent
Occur with <50% of uterine contractions in any 20-min window
Considerations
Frequency, duration, intensity, and relaxation
Normal
≤5 contractions per 10 minutes averaged over a 30-min window
Tachysystole
>5 contractions per 10 minutes averaged over a 30-min window; should always be qualified as to
the presence or absence of associated FHR decelerations
often classified as early, variable, or late, are decreases in
the fetal heart rate that have specific pathologic and physiologic associations. Although primarily focused on intrapartum monitoring, the 2008 National Institute of Child
Health and Human Development workshop report on
fetal monitoring provides an excellent summary of the
nomenclature and interpretation involved (Table 13-1)
(Macones et al, 2008).
NONSTRESS TEST
A normal result of an NST is defined as a 20-minute fetal
heart rate tracing that contains two heart rate accelerations lasting 15 seconds or longer that peak 15 beats or
more above the baseline. Often this is called a reactive NST
(Figure 13-2). Modifications are made in reference to gestational age. NSTs for fetuses at less than 32 weeks’ gestation are often considered reactive if the acceleration is 10
beats per minute or more above the baseline and lasts for at
least 10 seconds. Furthermore, to account for the periodicity of 20- to 30-minute sleep cycles in the fetus, an NST
that is not reactive over the first 20 minutes may be continued an additional 20 to 40 minutes. A nonreactive NST
or an NST with specific abnormalities (e.g., high or low
baseline, decelerations) should be followed by a biophysical profile (BPP). It is important to note that some abnormal states, such as a fetal CNS abnormality or maternal
drug ingestion, may contribute to a nonreactive NST. In
these cases, ultrasound examination may provide appropriate information to determine the diagnosis or required
management.
Falsely reassuring NSTs occur at a rate of 3 to 5 per
1000 tests, although this does not account for a baseline
rate of unpreventable fetal deaths (Freeman et al, 1982b).
The difficulty with the NST really lies in its lack of specificity for fetal death or compromise; the false-positive rate
may be as high as 50% (Freeman et al, 1982b).
The rather modest false-negative rate is likely because
of the NST being a measurement of short-term hypoxemia. Indeed, longer-term fetal status can be measured
through amniotic fluid assessment, because the amniotic
fluid is correlated with fetal urinary output, which is a
surrogate for renal perfusion. When combined with an
assessment of amniotic fluid level, the false-negative rate
of the NST is reduced to 0.8 per 1000, although a 60%
false-positive rate remains (Miller et al, 1996). Indeed,
when combined with the NST and amniotic fluid assessment—sometimes known as the modified biophysical
profile—reduces the risk of fetal death to negligible levels in high-risk populations (Clark et al, 1989). For these
reasons, the NST combined with amniotic fluid assessment is a modality of choice for monitoring the high-risk
pregnancy.
CONTRACTION STRESS TEST
The CST assesses the fetal heart rate response in the presence of contractions. This test improves on the specificity
and sensitivity of the NST by assessing the fetal response
to stress. In fact, the CST preceded the NST, although
the NST became more favorable because of fewer contraindications, ease of administration, and reduced time and
supervision necessary. Compared with the NST, there is
a much lower incidence of falsely reassuring tests (0.4 per
1000), representing an eightfold reduction in the risk of
fetal loss in one study (Freeman et al, 1982a).
134
PART IV Labor and Delivery
FIGURE 13-2 A reassuring nonstress test. Note two fetal heart rate accelerations exceeding 15 beats/min and lasting at least 15 seconds during
the monitoring period.
FIGURE 13-3 A contraction stress test. The fetal heart rate is plotted above the uterine contraction signal. Note the late deceleration after a
contraction; this is a positive, or abnormal, test result.
Contractions are stimulated by the administration of
intravenous oxytocin or through maternal nipple stimulation. Of course, the CST is contraindicated in patients
in whom contractions should not be provoked, such as
threatened preterm delivery or preterm premature rupture of membranes, prior classical cesarean delivery, or
placenta previa. A minimum of three contractions over
a 10-minute period of continuous fetal heart rate assessment are necessary for a satisfactory test interpretation. An
unsatisfactory test should be followed by continued testing
with a modification of the mode of contraction stimulation. A negative (i.e., normal) test result demonstrates no
late decelerations, whereas a positive test result shows late
decelerations after 50% or more of contractions (Figure
13-3). A positive test result requires immediate further
testing or evaluation, if not delivery. An equivocal test
demonstrates late decelerations with less than 50% of contractions and requires further testing or monitoring. A test
that encompasses a hyperstimulatory contraction pattern
(e.g., five contractions within 10 minutes or contractions
lasting longer than 90 seconds) is also considered equivocal and requires further testing.
ULTRASOUND
Although routine sonography for low-risk pregnant
women is controversial, few would disagree that the benefits far outweigh the costs for high-risk patients. Given
the higher risk for fetal complications such as anatomic
anomalies or growth disturbances, a specialized examination is performed between 18 and 20 weeks’ gestation in
most high-risk pregnancies.
Additional ultrasound modalities are also available,
including fetal echocardiography, three-dimensional (3D)
sonography, and Doppler. Cardiac anomalies are the most
common major congenital defects encountered in the
antepartum period. A four-chamber view of the heart at
the time of fetal anatomy survey at 18 to 20 weeks’ gestation will detect only 30% of congenital cardiac anomalies,
although the detection rate can be increased to approximately 60% to 70% if the outflow tracts are adequately
visualized (Kirk et al, 1994), but this still leaves 30% to
40% of all congenital cardiac anomalies undiagnosed. For
this reason, fetal echocardiography should be performed
by a skilled and experienced sonologist at 20 to 22 weeks’
gestation in all pregnancies at high-risk of a fetal cardiac
CHAPTER 13 Antepartum Fetal Assessment
anomaly; this includes pregnancies complicated by pregestational diabetes mellitus, a personal or family history of
congenital cardiac disease regardless of the nature of the
lesion or whether it has been repaired), maternal drug
exposure (e.g., lithium and paroxitene) (Bérard et al, 2007),
and pregnancies conceived by in vitro fertilization, but not
if the pregnancy was conceived using clomiphene citrate
or ovarian stimulation or intrauterine insemination alone
(Olson et al, 2005).
Compared with standard two-dimensional (2D) ultrasound, 3D ultrasound (or four-dimensional if fetal
movements are included) allows for visualization of fetal
structures in all three dimensions concurrently for the
improved characterization of complex fetal structural
anomalies and for storage of scanned images with 3D
reconstruction at a later date or remote location (telemedicine). Unlike 2D ultrasound, 3D images are greatly
influenced by fetal movements and are subject to more
interference from structures such as fetal limbs, umbilical
cord, and placental tissue. Because of movement interference, visualization of the fetal heart with 3D ultrasound is
suboptimal.
In addition to rapid acquisition of images that can be
later reconstructed and manipulated, 3D ultrasound has
other potential advantages:
ll Surface rendering mode can provide clearer images of
many soft tissue structures. Such images can improve
the diagnosis of certain fetal malformations, especially
craniofacial anomalies (cleft lip and palate, micrognathia, ear anomaly, facial dysmorphism, club foot, finger
and toe anomalies), intracranial lesions, spinal anomalies, ventral wall defects, and fetal tumors.
ll 3D ultrasound may be useful in early pregnancy by providing more accurate measurements of the gestational
sac, yolk sac, and crown-rump length. It may also allow
for a more accurate midsagittal view of the fetus for
measuring nuchal translucency.
ll 3D ultrasound can also be used to measure tissue volume. Preliminary data suggest that the assessment of
cervical volume may predict the risk of cervical insufficiency (Rovas et al, 2005), and measurement of placental volume in the first trimester may predict fetuses
at risk of intrauterine growth restriction (IUGR)
(Schuchter et al, 2001).
Despite these advantages and the fact that 3D ultrasound has been available since the early 1990s, it has yet to
live up to its promises. Although 3D ultrasound is unlikely
to replace standard 2D imaging in the near future, it is a
valuable complementary modality in obstetric imaging. As
the technology improves, it is likely that perinatal ultrasound will evolve to resemble computed tomography and
magnetic resonance imaging.
GROWTH ASSESSMENT
Normal fetal growth is a critical component of a healthy
pregnancy and the subsequent long-term health of the
child. A systematic method of examination of the gravid
abdomen was first described by Leopold and Sporlin
(1894). Although abdominal examination has several limitations, particularly in the setting of maternal obesity,
multiple pregnancy, uterine fibroids, or polyhydramnios,
135
it is safe, free, and well tolerated and may add valuable
information to assist in antepartum management. Palpation is divided into four separate Leopold maneuvers. Each
maneuver is designed to identify specific fetal landmarks
or to reveal a specific relationship between the fetus and
mother. For example, the first maneuver involves measuring fundal height. The uterus can be palpated above the
pelvic brim at approximately 12 weeks’ gestation. Thereafter, fundal height should increase by approximately 1 cm
per week, reaching the level of the umbilicus at 20 to 22
weeks’ gestation. Between 20 and 32 weeks’ gestation, the
fundal height (in centimeters, from the superior edge of
the pubic symphysis) is approximately equal to the gestational age (in weeks) in healthy women of average weight
with an appropriately grown fetus. However, there is a
wide range of normal fundal height measurements. One
study has shown a 6-cm difference between the 10th and
90th percentiles at each week of gestation after 20 weeks
(Belizan et al, 1978). Moreover, fundal height is maximal
at approximately 36 weeks’ gestation, at which time the
fetus drops into the pelvis in preparation for labor and
the fundal height decreases. For these reasons, reliance
on fundal height measurements alone will fail to identify more than 50% of fetuses with IUGR (Gardosi and
Francis, 1999). Serial fundal height measurements by an
experienced obstetric care provider are more accurate than
a single measurement, and they will lead to an improved
diagnosis of fetal growth restriction with reported sensitivities as high as 86% (Belizan et al, 1978).
If the clinical examination is not consistent with the
stated gestational age, an ultrasound examination is indicated to confirm gestational age and to establish a more
objective measure of fetal growth. Ultrasound examination may also identify an alternative explanation for the
discrepancy, such as multiple pregnancy, polyhydramnios,
oligohydramnios, fetal demise, or uterine fibroids.
For many years, obstetric sonography has used fetal
biometry to define fetal size by weight estimation, although
this approach has a number of key limitations. For example, regression equations used to create weight estimation
formulas are derived primarily from cross-sectional data
that rely on infants delivering within an arbitrary period
of time after the ultrasound examination, and they assume
that body proportions (i.e., fat, muscle, bone) are the
same for all fetuses. Moreover, growth curves for healthy
infants from 24 to 37 weeks’ gestation rely on data collected from pregnancies delivered preterm, which should
not be regarded as normal pregnancies and are likely to
be complicated by some element of uteroplacental insufficiency regardless of whether the delivery was spontaneous
or iatrogenic. Despite these limitations, if the gestational
age is well validated, the prevailing data suggest that prenatal ultrasound can be used to verify an alteration in fetal
growth in 80% of cases and exclude abnormal growth in
90% of cases (Sabbagha, 1987).
Sonographic estimates of fetal weight are commonly
derived from mathematical formulas that use a combination of fetal measurements, especially the BPD, abdominal circumference (AC), and femur length (Hadlock et al,
1984). Whereas the BPD may be the most accurate indicator of gestational age in the second or third trimesters,
fetuses gain weight in their abdomen, making the AC the
136
PART IV Labor and Delivery
single most important measurement for fetal size. The AC
is thus given more weight in these formulas. Unfortunately
the AC is also the most difficult measurement to acquire,
and a small difference in the AC measurement will result
in a large difference in the estimated fetal weight (EFW).
The accuracy of the EFW depends on a number of variables, including gestational age (in absolute terms, EFW
is more accurate in preterm or IUGR fetuses than in term
or macrosomic fetuses), operator experience, maternal
body habitus, and amniotic fluid volume (measurements
are more difficult to acquire if the amniotic fluid volume
is low). Although objective, sonographic EFW estimations
are not particularly accurate and have an error of 15%
20%, even in experienced hands (Anderson et al, 2007).
Indeed, a sonographic EFW at term is no more accurate
than a clinical estimate of fetal weight by an experienced
obstetric care provider or the mother’s estimate of fetal
weight if she has delivered before (Chauhan et al, 1992).
Sonographic estimates of fetal weight must therefore be
evaluated within the context of the clinical situation and
balanced against the clinical estimate of fetal weight. Serial
sonographic evaluations of fetal weight are more useful
than a single measurement in diagnosing abnormal fetal
growth. The ideal interval to evaluate fetal growth is every
3 to 4 weeks, with a minimum 10- to 14-day interval necessary to see significant differences. Because of the inherent error in fetal biometric measurements, more frequent
ultrasound determinations of EFW may be misleading.
Similarly, the use of population-specific growth curves,
if available, will improve the ability of the obstetric care
provider to identify abnormal fetal growth. For example,
growth curves derived from a population that lives at high
altitude, where the fetus is exposed to lower oxygen tension, will be different from those derived from a population at sea level. Abnormal fetal growth can be classified as
insufficient (i.e., IUGR) or excessive (fetal macrosomia).
The definition of IUGR has been a long-standing
challenge for modern obstetrics. Distinguishing the
healthy, constitutionally small-for-gestational-age fetus,
defined as an EFW below the 10th percentile for a given
week of gestation, from the nutritionally deprived, truly
growth-restricted fetus has been particularly difficult.
Fetuses with an EFW less than the 10th percentile are not
necessarily pathologically growth restricted. Conversely,
an EFW greater than the 10th percentile does not mean
that an individual fetus has achieved its growth potential,
and such fetuses may still be at risk of perinatal mortality
and morbidity. As such, IUGR is best defined as either an
EFW of less than the 5th percentile for gestational age in a
well-dated pregnancy or an EFW of less than the 10th percentile for gestational age in a well-dated pregnancy with
evidence of fetal compromise, such as oligohydramnios or
abnormal umbilical artery Doppler velocimetry.
Fetal growth restriction has traditionally been classified
into asymmetric or symmetric IUGR. Asymmetric IUGR
is characterized by normal head growth, but suboptimal
body growth, and is seen most commonly in the third trimester. It is thought to result from a late pathologic event,
such as chronic placental abruption leading to uteroplacental insufficiency, in an otherwise uncomplicated pregnancy
and healthy fetus. In cases of symmetric IUGR, both the
fetal head size and body weight are reduced, indicating a
global insult that likely occurred early in gestation. Symmetric IUGR may reflect an inherent fetal abnormality
(e.g., fetal chromosomal anomaly, inherited metabolic disorder, early congenital infection) or long-standing severe
placental insufficiency caused by an underlying maternal
disease (e.g., hypertension, pregestational diabetes mellitus, collagen vascular disorder). In practice, the distinction
between asymmetric and symmetric IUGR is not particularly useful.
Early and accurate diagnosis of IUGR coupled with
appropriate intervention will lead to an improvement in
perinatal outcome. If IUGR is suggested clinically and
by ultrasound examination, thorough evaluations of the
mother and fetus are indicated. Referral to a maternalfetal medicine specialist should be considered. Every
effort should be made to identify the cause of IUGR and
to modify or eliminate contributing factors. Up to 20%
of cases of severe IUGR are associated with fetal chromosome abnormalities or congenital malformations, 25% to
30% are related to maternal conditions characterized by
vascular disease, and a smaller proportion are the result
of abnormal placentation. However, in a substantial number of cases (50% or more in some studies), the cause of
the IUGR will remain uncertain even after a thorough
investigation (Resnik, 2002). Fetal macrosomia is defined
as an EFW (not birthweight) of 4500 g or greater, measured either clinically or by ultrasound, and is independent
of gestational age, diabetic status, or actual birthweight
(American College of Obstetricians and Gynecologists,
2000). Fetal macrosomia refers to a single cutoff EFW;
this should be distinguished from the large-for-gestational
age fetus, which is one in whom the EFW is greater than
the 90th percentile for gestational age. By definition, 10%
of all fetuses are large for gestational age at any given
gestational age. Fetal macrosomia is associated with an
increased risk of cesarean delivery, operative vaginal delivery, and birth injury to both the mother (including vaginal, perineal, and rectal trauma) and the fetus (orthopedic
and neurologic injury) (American College of Obstetricians
and Gynecologists, 2000, 2001; Kjos and Buchanan, 1999;
Magee et al, 1993; O’Sullivan et al, 1973; Widness et al,
1985). Shoulder dystocia with resultant brachial plexus
injury (Erb’s palsy) are a serious consequence of fetal macrosomia and are further increased in the setting of diabetes, because of the increased diameters in the upper thorax
and neck of fetuses of mothers with diabetes.
Fetal macrosomia can be determined clinically, by
abdominal palpation using the Leopold’s maneuvers, or
by ultrasound examination; these two techniques appear to
be equally accurate (Watson et al, 1988). However, EFW
measurements are less accurate in large (macrosomic)
fetuses than in normally grown fetuses, and factors such
as low amniotic fluid volume, advancing gestational age,
maternal obesity, and the position of the fetus can compound these inaccuracies. Clinical examination has been
shown to underestimate the birthweight by 0.5 kg or more
in almost 80% of fetuses with macrosomia (Niswander
et al, 1970). For these reasons, the prediction of fetal macrosomia is not particularly accurate, with a false-positive
rate of 35% and a false-negative rate of 10% (Niswander
et al, 1970; Watson et al, 1988). A number of alternative
sonographic measurements have therefore been proposed
CHAPTER 13 Antepartum Fetal Assessment
350
300
250
99th
200
95th
AFI
in an attempt to better identify the macrosomic fetus,
including fetal AC alone, umbilical cord circumference,
cheek-to-cheek diameter, and upper arm circumference;
however, these measurements remain investigational and
should not be used clinically.
Despite the inaccuracy in the prediction of fetal macrosomia, an EFW should be documented either by clinical
estimation or ultrasound examination in all women at high
risk at approximately 38 weeks’ gestation. Suspected fetal
macrosomia is not an indication for induction of labor,
because induction does not improve maternal or fetal outcomes (American College of Obstetricians and Gynecologists, 2000). However, if the EFW is excessive, an elective
cesarean delivery should be considered to prevent fetal and
maternal birth trauma. Although controversy remains as
to the precise EFW at which an elective cesarean delivery
should be recommended, a suspected birthweight in excess
of 4500 g in women with diabetes or 5000 g in women
without diabetes is a reasonable threshold (American
College of Obstetricians and Gynecologists, 2000, 2001,
2002).
137
Mean
5th
150
1st
100
50
0
16 18 20 22 24 26 28 30 32 34 36 38 40 42
Weeks gestation
FIGURE 13-4 Amniotic fluid index (AFI; in mm) plotted against
gestational age. The lines represent percentiles.
AMNIOTIC FLUID ASSESSMENT
Amniotic fluid plays a key role in the health and development of a growing fetus. Once considered an afterthought
during the ultrasound examination of the fetus, evaluation
of the amniotic fluid is now considered an integral part of
ultrasound evaluation for fetal well-being. Amniotic fluid
serves a number of important functions for the developing
embryo and fetus. It provides cushioning against physical
trauma; creates an environment free of restriction and or
distortion, allowing for normal growth and development
of the fetus; provides a thermally stable environment;
allows the respiratory, gastrointestinal, and musculoskeletal tracts to develop normally; and helps to prevent infection (Hill et al, 1984).
The chorioamnion acts as a porous membrane early
in pregnancy, allowing the passage of water and solutes
across the membrane; there is little contribution from the
small embryo. As the pregnancy progresses into the late
first trimester, the diffusion of fluid across the fetal skin
occurs, increasing the volume of amniotic fluid. In the
second half of the pregnancy, the main sources of amniotic fluid come from fetal kidneys and lungs. The primary
sources for removal of fluid are from fetal swallowing and
absorption into fetal blood perfusing the surface of the placenta. As more fluid is produced than is resorbed by the
fetal-placental unit, the volume of amniotic fluid increases
throughout the first 32 weeks of pregnancy (Figure 13-4).
The volume peaks at approximately 32 to 33 weeks’ gestation, and at this gestational age equal amounts of fluid
are produced and resorbed. After term, the amniotic fluid
declines at a rate of 8% per week (Brace and Wolf, 1989).
Because amniotic fluid plays a critical role in the normal
development of a fetus, the assessment of amniotic volume
is an essential component of the ultrasound evaluation for
fetal well-being. Subjective estimates of the amniotic fluid
volume have been validated, but two ultrasound measurements—amniotic fluid index (AFI) and maximum vertical
pocket—have been developed to quickly and accurately
assess the quantity of amniotic fluid surrounding the fetus.
The AFI is a semiquantitative method for assessing the
amniotic fluid volume with ultrasound. The gravid uterus
is divided into four quadrants using the umbilicus, linea
nigra, and external landmarks (Rutherford et al, 1987).
The deepest amniotic fluid pocket is measured in each
quadrant with the ultrasound transducer perpendicular
to the floor. The four measurements are added together
and the sum is regarded as the AFI. Pockets filled with
umbilical cord or fetal extremities should not be used for
generating the AFI (Hill, 1997). Researchers and clinicians
have used a variety of measurements to define abnormalities in amniotic fluid volume. However, the normal range
of the AFI most commonly used in clinical practice is 5 to
20 cm of fluid. Pregnancies with AFIs less than 5 cm can
be described as having oligohydramnios, and pregnancies
with measurements greater than 20 cm can be described as
having polyhydramnios.
The maximal vertical pocket is another semiquantitative method for assessing the fluid volume. The technique
involves scanning the gravid uterus for the single deepest pocket of amniotic fluid that is free of umbilical cord
and fetal parts and with the transducer perpendicular to
the floor, measuring the pocket of fluid (Manning et al,
1981b). Currently this method is used mostly in multiple
gestation pregnancies in which the AFI is not technically
feasible. Oligohydramnios can be defined as a single measurement less than 2 cm. Polyhydramnios can be defined
as a single measurement greater than 10 cm.
BIOPHYSICAL PROFILE
An NST alone might not be sufficient to confirm fetal
well-being; in such cases, a biophysical profile (BPP) may
be performed. The BPP refers to a sonographic scoring
system performed over a 30- to 40-minute period designed
to assess fetal well-being. The BPP was initially described
for testing postterm fetuses, but has since been validated
for use in both term and preterm fetuses (Manning et al,
138
PART IV Labor and Delivery
1981a, 1985, 1987; Vintzileos et al, 1983, 1987a, 1987b).
Notably, BPP is not validated for use in active labor. The
five variables described in the original BPP were: gross fetal
body movements, fetal tone (i.e., flexion and extension of
limbs), amniotic fluid volume, fetal breathing movements,
and NST (summarized in Table 13-2) (Manning, 1989).
More recently, however, BPP is interpreted without the
NST.
The individual variables of the BPP become apparent
in healthy fetuses in a predictable sequence: fetal tone
appears at 7.5 to 8.5 weeks, fetal movement at 9 weeks,
fetal breathing at 20 to 22 weeks, and FHR reactivity at 24
to 28 weeks’ gestation. Similarly, in the setting of antepartum hypoxia, these characteristics typically disappear in the
reverse order in which they appeared (i.e., FHR reactivity
is lost first followed by fetal breathing, fetal movements,
and finally fetal tone) (Vintzileos et al, 1987a). The amniotic fluid volume, which is composed almost entirely of
fetal urine in the second and third trimesters, is not influenced by acute fetal hypoxia or acute fetal central nervous
system dysfunction. Rather, oligohydramnios (decreased
amniotic fluid volume) in the latter half of pregnancy and
in the absence of ruptured membranes is a reflection of
TABLE 13-2 Fetal Biophysical Profile
Element
Criterion (2 points for each element
satisfied)
Breathing
≥1 episode of breathing movements lasting 30
seconds
Movement
≥3 discrete body or limb movements
Tone
≥1 episode of active extension and flexion of
limbs or trunk
Amniotic fluid
≥1 pocket of amniotic fluid measuring ≥2 cm in
two perpendicular planes
Nonstress test
≥2 fetal heart rate accelerations lasting ≥15 seconds over 20 minutes
chronic uteroplacental insufficiency, increased renal artery
resistance leading to diminished urine output, or both (Oz
et al, 2002); it predisposes to umbilical cord compression,
thus leading to intermittent fetal hypoxemia, meconium
passage, or meconium aspiration. Adverse pregnancy outcome (including nonreassuring FHR tracing, low Apgar
score, and neonatal intensive care unit admission) is more
common when oligohydramnios is present (Bochner et al,
1987; Morris et al, 2003; Oz et al, 2002; Tongsong and
Srisomboon, 1993). Serial (weekly) screening of high-risk
pregnancies for oligohydramnios is important, because
amniotic fluid can become drastically reduced within 24 to
48 hours (Clement et al, 1987).
Although each of the five features of the BPP are scored
equally (2 points if the variable is present or normal, and 0
points if absent or abnormal, for a total of 10 points), they
are not equally predictive of adverse pregnancy outcome.
For example, amniotic fluid volume is the variable that
correlates most strongly with adverse pregnancy events.
The recommended management based on the BPP is summarized in Table 13-3 (Manning, 1989). A score of 8 to
10 out of 10 is regarded as reassuring; a score of 4 to 6 is
suspicious and requires reevaluation, and a score of 0 to 2
suggests nonreassuring fetal testing—previously referred
to as fetal distress (Manning et al, 1981a, 1981b). Evidence
of nonreassuring fetal testing or oligohydramnios in the
setting of otherwise reassuring fetal testing should prompt
evaluation for immediate delivery (Vintzileos et al, 1987a,
1987b).
DOPPLER
Doppler velocimetry shows the direction and characteristics of blood flow, and it can be used to examine the
maternal, uteroplacental, or fetal circulations. Because of
placental capacitance, the umbilical artery is one of the
few arteries that normally has forward diastolic flow, and
it is one of the most frequently targeted vessels during
TABLE 13-3 Interpretation and Management of Biophysical Profile
Perinatal Morbidity or Mortality
Within 1 week (no intervention)
Management
Normal
<1/1000
No intervention
8/8
Normal
—
No intervention
8/10 (abnormal NST)
Normal
—
8/10 (abnormal amniotic
fluid)
Suspect chronic fetal
compromise, renal anomaly,
or rupture of membranes
89/1000
Rule out renal abnormality or rupture
of membranes; consider delivery or
prolonged observation if dictated by
gestational age
6/8 (other)
Equivocal, possible asphyxia
Variable
If fetus is mature, deliver; if immature,
repeat test within 4-6 hours
6/8 (abnormal amniotic
fluid)
Suspect asphyxia
89/1000
Repeat 4-6 hours; consider delivery
4/8
Suspect asphyxia
91/1000
If ≥36 weeks’ gestation or documented
pulmonary maturity, deliver immediately; if not, repeat within 4-6 hours
2/8
High suspicion of asphyxia
125/1000
Immediate delivery
0/8
High suspicion of asphyxia
600/1000
Immediate delivery
Score
Comment
10/10
No intervention
Adapted from Manning FA, Morrison I, Harman CR, et al: Fetal assessment based on fetal biophysical profile scoring: experience in 19,221 referred high-risk pregnancies, Am J
Obstet Gynecol 157:880, 1987.
NST, Nonstress test.
CHAPTER 13 Antepartum Fetal Assessment
pregnancy. Umbilical artery Doppler velocimetry measurements reflect resistance to blood flow from the fetus to
the placenta. Factors that affect placental resistance include
gestational age, placental location, pregnancy complications (placental abruption, preeclampsia), and underlying
maternal disease (chronic hypertension).
Doppler velocimetry of umbilical artery blood flow provides an indirect measure of placental function and fetal
status (Giles et al, 1985). Decreased diastolic flow with
a resultant increase in systolic-to-diastolic ratio suggests
increased placental vascular resistance and fetal compromise. Severely abnormal umbilical artery Doppler velocimetry (defined as absent or reversed diastolic flow) is an
especially ominous observation and is associated with poor
perinatal outcome, particularly in the setting of IUGR
(Ducey et al, 1987; McCallum et al, 1978; Rochelson
et al, 1987; Trudinger et al, 1991; Wenstrom et al, 1991;
Zelop et al, 1996). The overall mortality rate for fetuses
with absent or reversed flow may be near 30% (Karsdorp
et al, 1994). It should be noted that abnormal Doppler
studies are often seen in cases of anatomic anomalies or
chromosomal abnormalities, which should be noted when
managing a case.
The role of ductus venosus and middle cerebral artery
(MCA) Doppler in the management of IUGR pregnancies
is not well defined. As such, urgent delivery should be considered in IUGR pregnancies when the results of umbilical artery Doppler studies are severely abnormal regardless
of gestational age. However, it is unclear how to interpret
these findings in the setting of a normally grown fetus.
For these reasons, umbilical artery Doppler velocimetry
should not be performed routinely on low-risk women.
Appropriate indications include IUGR, cord malformations, unexplained oligohydramnios, suspected or established preeclampsia, and possibly fetal cardiac anomalies.
Umbilical artery Doppler velocimetry has not been shown
to be useful in the evaluation of a variety of high-risk
pregnancies, including diabetic and postterm pregnancies,
primarily because of its high false-positive rate (Baschat,
2004; Farmakides et al, 1988; Landon et al, 1989; Stokes
et al, 1991).
As such, in the absence of IUGR, obstetric management
decisions are not usually made on the basis of Doppler
velocimetry studies alone. Nonetheless, new applications
for Doppler technology are currently under investigation.
A recent application that has proved extremely useful is
the noninvasive evaluation of fetal anemia resulting from
isoimmunization. When a fetus develops severe anemia,
cardiac output increases and there is a decline in blood
viscosity, resulting in an increase in MCA blood flow,
which can be demonstrated by measuring the peak velocity using MCA Doppler velocimetry (Mari et al, 1995).
This demonstration can help the perinatologist to better
139
counsel such patients about the need for cordocentesis and
fetal blood transfusion. Doppler studies of other vessels—
including the uterine artery, fetal aorta, ductus venosus,
and fetal carotid arteries—have contributed considerably
to our knowledge of maternal-fetal physiology, but as yet
have resulted in few clinical applications.
SUMMARY
There are a variety of testing modalities available to the
obstetrician for assessing fetal well-being in the antepartum period, each with specific applications, advantages,
and disadvantages. As such, it is difficult to apply generalized protocols to the assessment of the fetus. A stepwise
approach entails applying the appropriate tests for low-risk
patients and identifying those patients, from the results of
those tests or from historical factors, for whom further
testing is needed. Although many tests, including NST,
fetal weight assessment, and uterine artery Doppler, may
be somewhat nonspecific and may have misleading falsepositive rates, combining those tests with others increases
the specificity. Test results that raise concerns require further investigation or active management.
SUGGESTED READINGS
American College of Obstetricians and Gynecologists (ACOG): Fetal macrosomia,
Washington, DC, 2000, ACOG Practice Bulletin No. 22, ACOG.
American College of Obstetricians and Gynecologists (ACOG): Gestational diabetes,
Washington, DC, 2001, ACOG Practice Bulletin No. 30, ACOG.
American College of Obstetricians and Gynecologists (ACOG): Shoulder dystocia,
Washington, DC, 2002, ACOG Practice Bulletin No. 40, ACOG.
American College of Obstetricians and Gynecologists (ACOG): Ultrasonography in
pregnancy, Washington, DC, 2009, ACOG Practice Bulletin No. 101, ACOG.
Anderson NG, Jolley IJ, Wells JE: Sonographic estimation of fetal weight: comparison of bias, precision and consistency using 12 different formulae, Ultrasound
Obstet Gynecol 30:173-179, 2007.
Baschat AA: Doppler application in the delivery timing of the preterm growthrestricted fetus: another step in the right direction, Ultrasound Obstet Gynecol
23:111-118, 2004.
Brace RA, Wolf EJ: Normal amniotic fluid volume changes throughout pregnancy,
Am J Obstet Gynecol 161:382-388, 1989.
Freeman RK, Anderson G, Dorchester W: A prospective multi-institutional study
of antepartum fetal heart rate monitoring: I. risk of perinatal mortality and
morbidity according to antepartum fetal heart rate test results, Am J Obstet
Gynecol143 :771-777, 1982.
Freeman RK, Anderson G, Dorchester W: A prospective multi-institutional study
of antepartum fetal heart rate monitoring: II. contraction stress test versus
nonstress test for primary surveillance, Am J Obstet Gynecol 143:778-781, 1982.
Macones GA, Hankins GD, Spong CY, et al: The 2008 National Institute of Child
Health and Human Development workshop report on electronic fetal monitoring: update on definitions, interpretation, and research guidelines, Obstet
Gynecol 112:661-666, 2008.
Manning FA, Morrison I, Harman CR, et al: Fetal assessment based on fetal biophysical profile scoring: experience in 19,221 referred high-risk pregnancies,
Am J Obstet Gynecol 157:880-884, 1987.
Manning FA, Morrison I, Lange IR, et al: Fetal assessment based on fetal biophysical profile scoring: experience in 12,620 referred high-risk pregnancies:
I. Perinatal mortality by frequency and etiology, Am J Obstet Gynecol 151:
343-350, 1985.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
14
Prematurity: Causes and Prevention
Tonse N.K. Raju and Caroline Signore
BURDEN OF PRETERM BIRTH
Despite major advances in perinatal medicine, preterm
births (PTBs) in the United States have been increasing
over the past two decades, reaching a high of 12.8% of live
births in 2006 (Martin et al, 2009). This rate amounts to
one PTB occurring at each minute of every day, throughout the year. From the perspective of a societal health care
burden, these data are sobering, because infants born even
1 or 2 weeks before full term suffer higher rates of morbidity and mortality throughout life. Thus even a minimal
increase in the PTB rate has a major effect on the societal
burden of disease. Conversely, even a modest reduction
in the PTB rate can have a major positive effect on the
societal health care burden. In this chapter we focus on the
etiology, prediction, and prevention of PTB.
Preliminary data in the United States for 2007 showed
that among the total number of live births, 2.04% were
very preterm, 1.59% were moderate preterm, and 9.03%
were late preterm (Hamilton et al, 2009). Thus, of all the
PTBs the late PTB stratum was the largest at 71%, the
very PTB stratum was intermediate at 16.1%, and
the moderate PTB stratum was the smallest at 12.6%.
The U.S. 2007 preliminary data also suggest a slight
decline in the total PTB rate to 12.7%, from 12.8% in
2006. The 2007 decline was predominantly among late
PTB, from 9.1% in 2006 to 9.0% in 2007 (Hamilton et al,
2009). However, trends over longer periods need to be
examined to confirm these preliminary findings and to
judge whether the PTB rates in the United States have
leveled or begun to decline.
DEFINITIONS
ETIOLOGY AND RISK FACTORS
The World Health Organization defines preterm birth as
a birth occurring either before 37 completed weeks of
gestation or on or before the 259th day, counting from
the first day of the last menstrual period. Only completed
weeks of gestation are reported; therefore an infant born
6 days after completing 35 weeks of gestation is noted as
35 weeks, not rounded up to 36 weeks. However, one can
precisely denote the exact gestational age by using a superscript for the number of days after the completion of the
gestational week. Thus, the infant in the example can be
noted as being born at 356/7 weeks’ gestation.
PTBs can be categorized into three major gestational
age strata: those occurring at <32 weeks’ (on or before 224
days) gestation, usually referred to as very preterm births;
those occurring between 320/7 and 336/7 weeks’ (between
225 and 238 days) gestation (a group with no specific
name, but can be termed moderate preterm births); and those
occurring between 340/7 and 366/7 weeks’ (239 to 259 days)
gestation, referred to as late preterm births (Figure 14-1). It
is worth noting that the phrase near term is no longer used
to refer to the third group, because the phrase falsely conveys a message that such “borderline” preterm infants are
almost as mature as term infants (Raju et al, 2006).
Individual PTBs can be categorized into two major
groups: (1) spontaneous PTB (accounting for up to 60%
of all PTBs), which occur after the spontaneous onset of
preterm labor, and in some cases, after spontaneous preterm rupture of the fetal membranes before the onset of
labor; and (2) indicated PTB (accounting for about 40% of
PTBs), which arise from interventions by the health care
team to reduce poor outcomes in the presence of specific
medical, surgical, or obstetric conditions and indications
in the mother or fetus.
EPIDEMIOLOGY
In the United States, the PTB rate increased from 10.6% in
1990 to 12.8% in 2006—a 21% increase (Goldenberg et al,
2008; Martin et al, 2009). This increase was part of a general trend in the distribution of gestational age at delivery,
which showed a dramatic, leftward shift by 1 week, such
that the peak modal week (the gestational week at which
most deliveries occurred) shifted from 40 weeks in 1992 to
39 weeks in 2002 (Davidoff et al, 2006). Whereas this shift
has decreased births at 41 weeks’ gestation or later, it has
also led to an increase in the number of all PTBs.
140
SPONTANEOUS PRETERM BIRTHS
In most spontaneous PTBs, the precise cause for the onset
of labor remains unknown; however, a variety of risk factors have been identified (Behrman and Butler, 2007)
(Table 14-1).
BEHAVIORAL AND PSYCHOLOGICAL
CONTRIBUTORS
Behavioral and psychological contributors, especially
when occurring in constellation, tend to increase the risk
for PTB. Some of these factors include excessive consumption of alcohol, smoking, use of cocaine, unfavorable diet,
prolonged and stressful physical labor during pregnancy,
shorter interval between pregnancies, and a variety of psychosocial stressors (Goldenberg et al, 2008).
Sociodemographic and Community
Contributors
Increased risk for PTB has been associated with extremes
of maternal age (<17 or >35 years), unmarried status,
poverty, adverse neighborhood conditions, and lower
educational attainment. The causal pathways for the
CHAPTER 14 Prematurity: Causes and Prevention
141
CATEGORIZATION OF GESTATIONAL
AGE AT BIRTH
First day of
LMP
Day #
1
141
225
239
260
295
Week #
0
20 0/7
32 0/7
34 0/7
37 0/7
42 0/7
Very Preterm
( 224 days)
Moderate Late Preterm
Term
Preterm (239 – 259 days) (260 – 294 days)
Post Term
( 295 days)
(225 – 238 days)
FIGURE 14-1 Categorization of gestational age at birth.
TABLE 14-1 Risk Factors for Preterm Birth*
Category
Specific Variable
Effect Size and Comments
Behavioral and psychological contributors
Tobacco and alcohol
Inconsistent effect; these variables may be confounders with lower socioeconomic status
Other drugs
Cocaine users experience a twofold increase in PTB; other drugs (e.g.,
marijuana) have inconsistent effects
Nutrition
Low prepregnancy weight and lower weight gain increase PTB risk; these
factors may be confounders with other medical conditions
Physical activity, employment
Inconsistent effect of employment per se; but long work hours and stress have
been associated with increased risk of PTBs
Douching
Frequent and prolonged vaginal douching during pregnancy can alter vaginal
flora and enhance the risk for PTB
Stress (chronic, acute, life events)
Emotional stress, living conditions, major stressful life events, discrimination,
and racism, and other forms of stress experiences may act through diverse
mechanistic pathways, affect many body functions, such as the HPA, immune
defense mechanisms, and autonomic nervous system, leading to PTB
Maternal age
U-shaped relationship with maternal age and PTB rate, with increased risk in
women <17 and >35 years; in black women, risk seems to increase by age 30
Marital status
Although unmarried mothers have higher rates of PTB, the protective effects of
marital status on the PTB rate may vary across different ethnic groups
Race and ethnicity
Non-Hispanic black women have high rates of PTB; the causes are likely to
multifactorial
Socioeconomic conditions
Disparities in PTB rates by SES (high rates with low SES) are well documented, although the etiologic pathways for such effects are unclear
Community and neighborhood
Adverse neighborhood conditions influence health outcomes, including PTB
rates, through direct and indirect pathways
Maternal medical illnesses and
conditions
Indicated PTBs may occur with chronic hypertension, hypertensive disorders of
pregnancy, systemic lupus, hyperthyroidism, pregestational diabetes mellitus,
cardiac disease, asthma, renal disorders, and gestational diabetes mellitus
Underweight and low weight gain
Low prepregnancy weight and lower-than-average increase in weight during
pregnancy have been documented with higher rates of PTB
Fetal conditions
In vitro fertilization, assisted reproductive technology, multifetal pregnancy,
congenital anomalies, umbilical cord accidents
Pregnancy conditions
Placenta previa, first-trimester vaginal bleeding, abruption of the placenta,
uterine malformations
Infections
Maternal-fetal infections, bacterial vaginosis
Other
Interpregnancy interval of less than 6 months is estimated to increase PTB
rate by 30% to 60%; family history of PTB is associated with higher rates
of PTB; it is unclear if the effect is caused by genetic factors, environmental
factors, or a combination of both
Lead, air pollution
Exposures linked to higher PTB rates include lead, tobacco smoke, air pollution
(sulfur dioxide, particulates, carbon monoxide), and arsenic in drinking water;
however, interactions between such exposures and confounding factors such
as SES, race, and ethnicity need to be studied
Sociodemographic
contributors
Medical and pregnancy
conditions
Environmental toxins
Gene-environment
interactions
Genetic susceptibility and gene environmental interactions is a rapidly evolving
field; according to some experts, epigenetic mechanisms are the final common pathway to explain diverse sociodemographic, racial, and ethnic factors
and their effects on PTB rates
HPA, Hypthalamopituitary axis; PTB, preterm birth; SES, socioeconomic status.
*For a comprehensive discussion of various factors noted in this table, please see Behrman RE, Butler AS: Preterm birth, causes, consequences, and prevention, Washington, DC, 2007, The
National Academies Press.
142
PART IV Labor and Delivery
association between adverse sociodemographic and community contributors are complex and poorly understood.
It should be noted, however, that these factors are interrelated and often coexist with other medical or obstetric
conditions.
Racial Disparity
African American women are consistently found to have
higher PTB rates than women of other racial and ethnic
groups (16% to 18% versus 5% to 9%; Goldenberg et al,
2008). Black women are also more likely to experience
preterm premature rupture of membranes (PPROM) and
very PTB. The reason for this persistent racial disparity is
unknown.
Previous Preterm Birth
The prior history of a PTB is consistently identified as one
of the strongest predictors of preterm delivery, elevating
the risk to more than twofold the background rate (Mercer
et al, 1999). Risk is further elevated if there is a history of
multiple PTBs, consecutive previous PTBs, and births at
earlier gestational ages (Spong, 2007).
Short Cervix
Among asymptomatic women, cervical length less than
25 mm at 24 weeks’ gestation, as measured by transvaginal ultrasound, in singleton pregnancy is associated with
a sixfold increase in the risk of delivery before 35 weeks’
gestation (Iams et al, 1996). Among women in suspected
preterm labor, a cervical length of less than 15 mm may
discriminate between women who are likely to deliver
preterm and those whose pregnancies will continue (Tsoi
et al, 2003).
Genetic Factors
The risk of PTB is increased in women with a positive family history of PTB. Single nucleotide polymorphisms in a number of maternal and fetal candidate genes
involved in inflammatory pathways have been associated
with PPROM and PTB (Varner and Esplin, 2005). Studies
using genomic and proteomic approaches to further investigate molecular risk factors for PTB are in progress.
Infections
Overt or subclinical intrauterine infection, which might
lead to rupture of the membranes before onset of labor,
has been hypothesized to etiologically contribute to
onset of labor, especially among very PTBs. The most
common route of intrauterine infection is ascension of
vaginal bacteria. Bacterial vaginosis (BV) is a common
vaginal infection characterized by a shift in vaginal flora
toward anaerobic species, accompanied by an abnormally high pH. Risk of PTB is increased up to twofold
in women with BV (Hillier et al, 1995; Meis et al, 1995).
Periodontal disease has also been linked to PTB (Jeffcoat
et al, 2001); however, the biologic pathway is not well
understood.
Multifetal Gestation
Twin and higher-order pregnancies account for 3% of all
births. Multiple pregnancies are at increased risk of preterm delivery, with approximately 40% of twins delivered
spontaneously before 37 weeks’ gestation. Uterine overdistention is believed to be the major etiologic factor in these
births. An additional 20% of twins will develop maternal
or fetal complications leading to indicated PTB. The mean
gestational age at delivery is 35 weeks’ gestation for twins,
32 weeks’ gestation for triplets, and 29 weeks’ gestation for
quadruplets (Elliott, 2005; Martin et al, 2003).
Infertility Treatment
The use of assisted reproductive technology (ART),
in which gametes are manipulated outside the body, is
steadily increasing. In 2006, 41,000 American women
delivered after successful infertility treatment with ART,
accounting for approximately 1% of all live births (Sunderam et al, 2009). The use of ART is strongly associated with multifetal pregnancies, dramatically increasing
the risk for PTBs. However, even singleton pregnancies following ART have a twofold higher risk for PTB
(Jackson et al, 2004), suggesting a complex relationship
between a history of infertility, ART, and reproductive
outcomes. Medical treatment of infertility (i.e., ovulation induction) is also associated with multiple gestation and PTB. In 2003, 21% of twin, 37% of triplet,
and 62% of quadruplet and higher-order multiple births
were attributable to non-ART ovulation induction
(Dickey, 2007).
Maternal Body Habitus: Underweight
and Obesity
Low prepregnancy body mass index (<18.9 kg/m2) is associated with a 1.5- to 2.5-fold increase in the risk of PTB
and has previously been a target of PTB prevention efforts
(Goldenberg et al, 1998; Siega-Riz et al, 1996). Currently
there is an epidemic of overweight and obesity in all U.S.
populations. Maternal obesity is not associated with an
increased risk of spontaneous PTB (Hendler et al, 2005).
However, maternal obesity is a risk factor for a variety of
pregnancy complications, including congenital anomalies,
preeclampsia, and macrosomia, which may result in the
need for indicated PTB (Aly et al, 2010).
Fetal Fibronectin
Fetal fibronectin (FFN) is a glycoprotein involved in the
adherence of the fetal membranes to the uterine wall. The
presence of FFN in the cervicovaginal secretions has been
considered a risk factor for PTB and has been evaluated
for its predictor value of preterm delivery in large cohort
studies. A positive test result for FFN at 22 to 24 weeks’
gestation in asymptomatic women is significantly associated with PTB (Goldenberg et al, 1998); however, because
of its low positive predictive value, it is not a reliable universal screening tool in low-risk women. The most useful
aspect of FFN assessment is its high negative predictive
value: among women with signs and symptoms of preterm
CHAPTER 14 Prematurity: Causes and Prevention
143
labor and a negative FFN, 99% will not deliver over the
next 7 days (Lu et al, 2001).
ischemia, uterine over distension, abnormal allograft reaction, allergy, cervical insufficiency, and hormonal dysregulation (Romero et al, 2006).
INDICATED PRETERM BIRTHS
Allostatic Load
Medical and Pregnancy Conditions
Well-recognized medical and pregnancy-related conditions are noted in Table 14-1. Many of these conditions
are also encountered to a greater extent among women in
lower socioeconomic strata and among those living under
adverse conditions.
Maternal and fetal conditions that can lead to indicated
preterm deliveries include diabetes; hypertension; preeclampsia or eclampsia; maternal cardiovascular and pulmonary disorders; multifetal gestations; uterine structural
disorders; polyhydramnios or oligohydramnios; placenta
previa, abruption, and other uterine hemorrhagic events;
chorioamnionitis and fetal infection; fetal distress; and
congenital anomalies.
Causes for Increasing Preterm Birth
The precise reasons for increasing rates of PTB are unclear.
The contribution of many factors is noted in Table 14-2
(Behrman and Butler, 2007; Raju, 2006; Raju et al, 2006).
Etiologic Pathways
Preterm labor and delivery arise from abnormal activation
of parturition. No single mechanism describes all cases
of PTB; rather, preterm parturition can be viewed as an
obstetric syndrome with multiple etiologies, a long preclinical phase, frequent fetal involvement, and genetic factors
that modify risk. Contributing pathologic processes may
include intrauterine inflammation or infection, uterine
TABLE 14-2 Possible Causes for Increasing Preterm Birth*
Variable
Comments
Increasing maternal age at
first pregnancy
Number of pregnant women 35-39
years nearly doubled between 1993
and 2003
Increasing infertility
treatment and multifetal
gestations
Number of women seeking infertility treatment increased twofold
between 1996 and 2002
Change in practice parameters or guidelines
Improved surveillance and increased
medical interventions to prevent
still births
Increasing cesarean section
rates and decreasing
VBAC
—
Increasing maternal overweight and obesity
Errors in estimating gestational age
of macrosomic fetus
Inaccuracy in estimating
gestational age
Lack of early ultrasound examination
Iatrogenic or nonmedical
reasons
Maternal request for early delivery or
other logistical considerations of
the patient, family or health care
team
VBAC, Vaginal birth after cesarean.
*For a review, please see Goldenberg et al (2008), Raju (2006), and Reddy et al (2007,
2009).
Recent studies of the pathophysiology of and biologic
responses to stress have uncovered a series of causal pathways for adverse outcomes caused by stresses at multiple
levels and from different sources. The biologic responses
to stressors have been collectively called allostatic load, a
summary measure of major perturbations in the autonomic, endocrine, and immune systems. Allostatic load has
been linked to accelerated aging, worsened immunologic
tolerance, ineffective defense systems, and increased cardiovascular morbidity. Abnormal allostatic load during the
entire life span of a woman can potentially modulate her
biologic systems and worsen perinatal outcomes (Groer
and Burns, 2009; Herring and Gawlik, 2007; Latendresse,
2009; Lu and Halfon, 2003; McEwen, 2006; Shannon
et al, 2007). Studies are under way to understand the complex relationship between the effects of sociodemographic
and biologic stressors on organ systems and their influence
on perinatal outcomes.
PREVENTION OF PRETERM BIRTH
Despite intense research efforts, few widely effective strategies for preventing PTB have been identified. Because of
the multifactorial nature of preterm labor and birth, it is
unlikely that any single intervention could serve to prevent
all cases of prematurity. In the United States, efforts have
focused on addressing specific risk factors to prevent PTB
(i.e., secondary prevention), such as treatment of infection,
and interventions to mitigate the morbidity and mortality
of PTB (i.e., tertiary prevention), such as administration
of tocolytic drugs to women in preterm labor (Iams et al,
2008). A better understanding of the complex mechanisms
leading to preterm labor and birth is needed to advance
development of interventions to prevent PTB. In this section, a number of strategies—both encouraging and disappointing in terms of effectiveness—are described.
Progesterone
Progesterone is thought to have antiinflammatory effects
and to be a smooth muscle relaxant that inhibits uterine
contractions. Administration of progesterone has shown
the most promise as an effective intervention for preventing PTB in women with certain risk factors for PTB. In a
recent metaanalysis of 11 trials, progesterone significantly
decreased PTB of singleton gestations less than 34 weeks
in women with a prior spontaneous preterm birth (SPTB)
by 85%, with a number needed to treat of seven (Dodd
et al, 2005). However, progesterone does not prevent PTB
in women carrying twins (Rouse et al, 2007) or triplets
(Caritis et al, 2009). Petrini et al, (2005) estimated that if all
eligible women with a history of prior SPTB were treated
with progesterone, then 10,000 PTBs per year could
be prevented. Progesterone treatment does not appear
to pose a developmental risk to the fetus or infant. In a
follow-up study of children exposed to 7-alpha hydroxyprogesterone caproate versus placebo in utero, there were
144
PART IV Labor and Delivery
no differences in growth or neurodevelopmental measures
between groups at age 4 years (Northen et al, 2007).
Cerclage
Cervical cerclage is a purse-string suture of the cervix
intended to mechanically prevent cervical dilation. A number of investigators have evaluated the use of cerclage to
prevent PTB in women with shortened cervical length, and
they have not demonstrated a significant effect (Berghella
et al, 2004; To et al, 2004). A more recent trial indicated a
significant beneficial effect (adjusted odds ratio, 0.23; 95%
confidence interval, 0.08 to 0.66) of cerclage in women
with a history of previous SPTB and cervical length less
than 15 mm (Berghella et al, 2008; Owen et al, 2009).
Limited Embryo Transfer
Preterm delivery of multifetal pregnancies arising from
ART can be prevented in many cases by limiting the number of embryos transferred to the uterus. The proportion
of in vitro fertilization cycles in which three or more fresh
embryos were transferred declined significantly in the
United States between 1996 and 2002 (92% to 54%; p
<0.001); however, the proportion of single-embryo transfer cycles remained small (2.5% in 2002) (Reynolds and
Schieve, 2006). Recent practice guidelines from the American Society for Reproductive Medicine call for wider
consideration of single-embryo transfer in many clinical
situations (American Society for Reproductive Medicine,
2008).
Multifetal Pregnancy Reduction
Reduction of multifetal pregnancy, or the selective termination of one or more fetuses in a high-order pregnancy,
has been proposed as a means to reduce the risk of prematurity and other pregnancy complications. Although effective at preventing some very PTBs, the procedure carries
an approximately 1% risk of loss of the entire pregnancy
(Stone et al, 2002) and can create profound ethical dilemmas (American College of Obstetricians and Gynecologists, 2007).
Treatment of Vaginal Infections
Despite consistent associations between lower genital tract
infection and PTB, multiple randomized trials have failed
to demonstrate a benefit of screening and antibiotic treatment for vaginitis or genital tract colonization on the PTB
rates (Carey et al, 2000; Iams et al, 2008; Klebanoff et al,
2001; McDonald et al, 2007).
Treatment of Periodontal Disease
PTB has been associated with maternal periodontal disease in a number of studies, and early clinical trials suggested that treatment of periodontitis reduces the risk
of PTB (Jeffcoat et al, 2003; Lopez et al, 2002). On the
other hand, a recent multicenter clinical trial in the United
States found that, whereas periodontal disease improved
in pregnant women randomized to treatment, there were
no differences in rates of PTB, low birthweight, or fetal
growth restriction in the treatment group compared with
controls (Michalowicz et al, 2006).
Tocolytics: Acute and Maintenance
Preterm labor (PTL), is defined as the onset before 37
weeks’ gestation of regular uterine contractions that result
in cervical change. Although numerous pharmacologic and
nonpharmacologic treatments have been aimed at arresting PTL and preventing PTB, none have been shown to
prolong pregnancy for more than 2 to 7 days (Iams et al,
2008). Nevertheless, tocolytic drugs are often used to allow
for the administration of steroids and maternal transfer to
a facility capable of caring for a preterm neonate. Drugs
commonly used to treat PTL are summarized in Table
14-3. Maintenance tocolysis after an acute episode of PTL
has not been shown to prevent PTB (Dodd et al, 2006;
Gaunekar and Crowther, 2004; Nanda et al, 2002).
MANAGEMENT OF PRETERM PREMATURE
RUPTURE OF MEMBRANES
Preterm premature rupture of membranes (PPROM) is
defined as the prelabor rupture of membranes before 37
weeks’ gestation. The period between initial leakage of
TABLE 14-3 Drugs Commonly Used to Treat Preterm Labor*
Agent
Class, Mechanism
Maternal Side Effects
Fetal and Neonatal Effects
Magnesium sulfate
Decreases intracellular calcium,
inhibits contractile response
Flushing, lethargy, nausea, muscle
weakness
Lethargy, decreased tone, respiratory
depression
Terbutaline
β-Mimetic
Tachycardia, hyperglycemia,
myocardial ischemia, pulmonary
edema
Tachycardia, hyperinsulinemia,
hyperglycemia
Nifedipine
Calcium channel blocker
Hypotension, flushing
None reported
Indomethacin
NSAID, prostaglandin synthase
inhibitor
Gastrointestinal upset
Decreased renal function, oligohydramnios, closure of the ductus
arteriosus
Atosiban (not available in
the United States)
Oxytocin inhibitor
Hypersensitivity, injection site
reaction
Increased risk of fetal or infant death
if administered before 28 wk
NSAID, Nonsteroidal antiinflammatory drug.
*For a review, see American College of Obstetricians and Gynecologists (2007), Dodd et al (2006), Gaunekar and Crowther (2004), Nanda et al (2002), Simhan and Caritis (2007),
and Stone et al (2002).
CHAPTER 14 Prematurity: Causes and Prevention
fluid and the onset of labor and delivery is known as the
latency period. Without treatment, the latency period after
PPROM is usually less than 1 week, with an inverse relationship between the gestational age at rupture and length
of the latency period. In a large multicenter trial, a 7-day
course of broad-spectrum antibiotics was shown to prolong the latency period for up to 3 weeks in women who
had experienced PPROM at 24 to 32 weeks’ gestation. In
addition, antepartum antibiotic treatment decreased the
incidence of chorioamnionitis and major morbidity (respiratory distress syndrome, early sepsis, severe intra ventricular hemorrhage, or severe necrotizing enterocolitis)
in the infants (Mercer et al, 1997).
SUMMARY
PTBs are occurring at epidemic proportions in the United
States. With more than 4.3 million live births each year,
a prematurity rate of 12.5% results in more than 537,000
PTBs annually. As described in other chapters, preterm
infants are at greater risk for mortality, and short- and
long-term morbidities compared with term infants. A
recent study from Norway on the adult-age outcomes of
preterm infants reported that even mild prematurity was
associated with significantly elevated risk for long-term
adverse medical, behavioral, psychological, and vocational
outcomes (Moster et al, 2008). In addition to the families’
burden of illness, death, and disability, society bears a large
burden of health care costs related to PTBs. The collective economic burden from prematurity was more than
$26 billion in 2005 (Behrman and Butler, 2007). Given the
complex nature of the risk factors and causal pathways for
PTBs, and the difficulties in diagnosing, preventing, and
treating them, there have been renewed calls in support
of multidisciplinary research to address the problems of
PTBs. The Institute of Medicine of the National Academies of Science issued a major publication providing
specific recommendations and guidelines for research,
surveillance, and education on this topic (Behrman and
Butler, 2007). The U.S. Senate approved the PREEMIE
145
Act on August 1, 2006. This bill called for efforts to reduce
preterm labor and delivery and the risk of pregnancyrelated deaths and complications, and to reduce infant
mortality caused by prematurity. The U.S. House of Representatives passed the bill unanimously on December 9,
2006, and the president signed it into law on December
22, 2006 (March of Dimes Foundation, 2006). The March
of Dimes Foundation initiated the Prematurity Campaign,
designed to educate and inform health care professionals, industry, government agencies, and the general public
about prematurity-related issues. These and other efforts
have begun recently and will take time to yield positive
results. Their collective effect on reducing the burden of
PTBs needs to be monitored on a long-term basis.
SUGGESTED READINGS
Behrman RE, Butler AS: Preterm birth, causes, consequences, and prevention,
Washington, DC, 2007, The National Academies Press.
Dodd JM, Crowther CA, Cincotta R, et al: Progesterone supplementation for
preventing preterm birth: a systematic review and meta-analysis, Acta Obstet
Gynecol Scand 84:526-533, 2005.
Goldenberg RL, Culhane JF, Iams JD, et al: Epidemiology and causes of preterm
birth, Lancet 371:75-84, 2008.
Goldenberg RL, Iams JD, Mercer BM, et al: The preterm prediction study: the
value of new vs standard risk factors in predicting early and all spontaneous preterm births. NICHD MFMU Network, Am J Public Health 88:233-238, 1998.
Iams JD, Romero R, Culhane JF, et al: Primary, secondary, and tertiary interventions to reduce the morbidity and mortality of preterm birth, Lancet 371:164175, 2008.
Owen J, Hankins G, Iams JD, et al: Multicenter randomized trial of cerclage for
preterm birth prevention in high-risk women with shortened midtrimester
cervical length, Am J Obstet Gynecol 201:375, 2009.
Raju TN, Higgins RD, Stark AR, et al: Optimizing care and outcome for latepreterm (near-term) infants: a summary of the workshop sponsored by the
National Institute of Child Health and Human Development, Pediatrics
118:1207-1214, 2006.
Romero R, Espinoza J, Kusanovic JP, et al: The preterm parturition syndrome,
BJOG 113(Suppl 3):17-42, 2006.
Simhan HN, Caritis SN: Prevention of preterm delivery, N Engl J Med 357:
477-487, 2007.
Spong CY: Prediction and prevention of recurrent spontaneous preterm birth,
Obstet Gynecol 110:405-415, 2007.
Varner MW, Esplin MS: Current understanding of genetic factors in preterm
birth, BJOG 112(Suppl 1):28-31, 2005
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
15
Complicated Deliveries: Overview
Sarah A. Waller, Sameer Gopalani, and Thomas J. Benedetti
Historically, child birth was often regarded as a perilous
undertaking. However, over the past century in the United
States, perinatal and maternal mortality have dramatically
fallen with advances in modern obstetric care, such as widespread use of antibiotics, easy access to expedient cesarean delivery, and better understanding of the proper use
of instruments such as forceps and vacuum extraction (Ali
and Norwitz, 2009). Indeed, adverse outcomes are generally uncommon in modern obstetrics, and unlike in the past,
most labor and delivery concludes with a healthy mother
and neonate. Nevertheless, complicated deliveries still exist,
and knowledge of their conduct and sequelae is still required
for the administration of proper maternal and infant care.
In this chapter we will first address the complicated vaginal delivery, with particular attention to neonatal outcomes.
We will then discuss cesarean delivery and vaginal birth
after a prior cesarean delivery (VBAC) and what neonatal
implications these may have. Before discussing complicated
labor and its neonatal effects, it is important to have a brief
understanding of the conduct of normal labor and delivery.
A comprehensive discussion of labor and delivery is beyond
the scope of this chapter, and the interested reader is directed
Williams Obstetrics, ed 23, chapter on normal labor.
The first stage of labor begins with the onset of regular uterine contractions with concomitant cervical dilation and effacement, and it ends with complete cervical
dilation. The first stage is further subdivided into a latent
phase, the length of which is variable and can last for several hours, and an active phase, which usually begins when
the cervix is dilated 3 to 4 cm and is marked by further
rapid, progressive cervical dilation and effacement. Often
the diagnosis of the transition from latent to active phase
labor is retrospective, because the time of onset of active
labor is variable by patient. The second stage begins with
complete cervical dilation and terminates with the expulsion of the fetus from the birth canal. The third stage of
labor concludes with the delivery of the placenta.
Disorders of the conduct of labor are of either protraction, in which cervical dilation or fetal descent occurs
but is at a rate much less than expected, or arrest. Both
disorders are addressed by operative delivery if they are
unresponsive to active medical management; this can be
performed abdominally through cesarean section or vaginally by obstetric forceps or vacuum extraction if the cervix
is fully dilated and specific criteria are fulfilled (see later
discussion on operative vaginal delivery). All these modalities can have neonatal and maternal effects, and the choice
of instrument or mode of delivery must always be selected
taking these potential morbidities into account.
CESAREAN SECTION
Neonatal mortality within 7 days of birth was 17.8 per
1000 live births in 1950; it is currently 3.8 per 1000 (Ali
and Norwitz, 2009). Certainly much of the improvement
146
in neonatal survival stems from advances in neonatal intensive care and resuscitative techniques. Along with improved
technology available to the pediatricians, however, is
the fact that the last 50 years have also seen a dramatic
increase in the universal access to safe cesarean section,
which affords quick and timely fetal delivery. However,
a cause-and-effect relationship between cesarean delivery
and improved neonatal outcome has never been demonstrated, and recent evidence suggests that some current
obstetric cesarean delivery practices may actually cause
harm (Bates, 2009 ). Currently almost one mother in three
is giving birth by cesarean section, a record level for the
United States. Recommendations from the U.S. Department of Health and Human Services Healthy People
2010 recommend a cesarean section rate of less than 15%
for a first pregnancy and 63% for previous cesarean sections. In addition, there are wide variations in cesarean
delivery rates among hospitals in a given state or region,
suggesting that factors other than pregnancy risk factors
may be responsible for the current cesarean delivery rate.
This rise in cesarean delivery has been associated with a
parallel drop in the vaginal operative delivery rate to less
than 10%. These two trends have increased the cost of
childbirth in the United States and are now threatening
many state budgets, because almost 50% of obstetric care
cost is paid with public funds (i.e., Medicaid). In response
as the result of a budget crisis, the state of Washington
reduced hospital payment for uncomplicated cesarean
delivery (DRG 371: Cesarean section) by reimbursing only
the equivalent of a complicated vaginal birth (DRG 372:
Normal delivery with problems), a projected savings of
$2 million per year. Many states are actively working to
safely reduce the rate of cesarean delivery. Projects are
under way to reduce elective delivery before 39 weeks’
gestation, safely promote VBAC and limit the occurrence
of higher-order multiple gestations.
Cesarean section is usually performed through either a
Pfannenstiel or vertical skin incision. The uterine incision
is often made transversely in the lower uterine segment,
because it minimizes intraoperative blood loss and future
risk of rupture during subsequent labor, compared with a
vertical or classical incision. The risk of rupture in future
labor is thought to be 0.5% to 1.0% for a low transverse
incision, compared to 4% to 9% for a classical incision
(Feingold et al, 1988).
Cesarean delivery is also performed for disorders of protraction or arrest in the first stage of labor when conservative measures fail to augment delivery, such as oxytocin or
amniotomy, or in the second stage when assisted or operative vaginal delivery is deemed unfeasible or unsafe.
Accepted obstetric indications for operative delivery are
as follows:
1. Fetal malpresentation (e.g., shoulder or breech)
2. Placenta previa
3. Prior classical uterine incision
CHAPTER 15 Complicated Deliveries: Overview
4. Fetal status not reassuring, remote from vaginal
delivery
5. Higher-order multiple gestation (triplet or greater)
6. Fetal contraindications to labor (alloimmune
thrombocytopenia, neural tube defect)
7. Maternal contraindication to labor (e.g., history of
rectal or perineal fistulas from inflammatory bowel
disease, large lower-uterine segment, cervical leiomyoma preventing vaginal delivery)
8. Maternal choice after counseling regarding risks
versus benefits
COMPLICATED VAGINAL DELIVERY:
OBSTETRIC FORCEPS AND VACUUM
EXTRACTION
DESCRIPTION OF THE OBSTETRIC FORCEPS
Obstetric forceps have been used to facilitate vaginal
deliveries since 1500 bc. Most commonly, the invention is
credited to Peter Chamberlen and his brother of England.
Designed originally as a means of extracting fetuses from
women who were at high risk of dying during childbirth,
forceps now are an alternative to cesarean delivery in
women with a protracted second stage of labor. Originally,
many of these instruments were furnished with hooks and
other accessories of destruction, and they were intended to
save the mother but not the fetus. Over the last 500 years,
the modern instruments in current use have been through
hundreds of modifications, safer techniques have been
established, and the overriding goal now includes delivering an intact, living baby and a healthy mother (Meniru,
1996; O’Grady et al, 2002).
Current obstetric forceps were first devised for practical
use in the sixteenth and seventeenth centuries and were
perfected over the past 300 years into the models in current use. Although there are many variations on the standard blueprint depending on the indication for its use, all
obstetric forceps have a similar design.
Forceps are made of stainless steel and consist of two
blades (each approximately 37.5 cm long, crossing each
other), a lock at the site of crossing, and a handle, whereby
the instrument is grasped by the obstetrician. The long
blades are parallel, divergent, or convergent, depending
on the type of instrument; the lower 8 cm comprises the
shank, which is the part between the blade proper and the
handle, giving a length for the forceps that is sufficient to
be locked easily outside the vagina. The four most common types of lock are the sliding lock, which can articulate anywhere along the shanks of the blade (Kielland and
Barton’s forceps); the English, which is fixed, a double slot
lock; the French, which is a screw lock; and the German,
which is a combination of the English and French. The
part of the forceps that grasps the fetal head is the blade;
this is further divided into the heel, which is the part closest to the lock, and the toe, which is the most distal part of
the blade. The blade can be either fenestrated—meaning
the body of the blade is hollow—or solid to prevent fetal
head compression. A further modification is pseudofenestration, in which a solid blade has a ridged edge, combining
the advantages of easier applicability and less fetal trauma
that a solid blade affords with the ease of traction of a
147
FIGURE 15-1 Simpson forceps: a standard obstetric forceps with
features common to all such instruments.
TABLE 15-1 Types of Obstetric Forceps in Most Common Use
Anatomic
Modification
General Use
Tucker-McClane
Solid blade
Nonmolded vertex
Simpson
Parallel shanks
Molded vertex or
significant caput
Elliot
Convergent shanks
Nonmolded vertex
Laufe
Pseudofenestrated
blade; divergent
shanks
For preterm infants
or EFW <2500 g
English lock; absent
pelvic curve
For rotation of fetal
vertex >45 degrees
Long handles with no
pelvic curvature
For after-coming
head in breech
vaginal delivery
Type
Classic
Rotational
Kielland
Breech
Piper
EFW, Estimated fetal weight
fenestrated blade. Obstetric forceps also possess a rounded
cephalic curve, which accommodates the fetal vertex,
and a pelvic curve that mirrors the maternal pelvic curve
(Figure 15-1).
There are more than 60 different types of obstetric forceps described in the literature, but most of are not used
currently. The forceps used most commonly today are
described in Table 15-1 , along with their indications for
use and the variations in anatomy, which distinguish one
from the other.
INDICATIONS FOR USE OF OBSTETRIC
FORCEPS
To an individual without obstetric training, the use of forceps can be a dangerous and difficult undertaking, fraught
with potential trauma for both the mother and fetus.
It is true that the use of this instrument, if not performed
carefully or appropriately, can have serious consequences.
Nevertheless, with properly trained hands, and a proper
appreciation of its use, forceps can be lifesaving for both
mother and fetus.
The criteria for the safe application of obstetric forceps
are as follows:
1. The cervix must be fully dilated.
2. The position of the fetal vertex must be known.
Forceps should not be applied when the fetal presentation is in doubt.
148
PART IV Labor and Delivery
3. The fetal vertex must be engaged within the maternal pelvis. Often in difficult or challenging labors,
significant caput can lead to the false impression
that fetal station is lower than it actually is. For this
reason, the obstetrician must be confident that the
actual biparietal diameter has passed the pelvic inlet
and that the leading part of the fetal skull is beyond
the level of the ischial spines. In addition, when the
presentation is occiput posterior, the leading point
of the fetal skull may appear to be lower in the pelvis
although the biparietal diameter has not yet passed
through the pelvic inlet; this can also lead to an
erroneous conclusion about fetal station.
4. When the forceps are properly applied, the sagittal
suture must be exactly midway between the blades,
and the lambdoidal sutures should be equidistant
(usually one fingerbreadth) from the edge of the
blade.
If these conditions are not met, application of the forceps should be reconsidered. Furthermore, in 1988 the
American College of Obstetricians and Gynecologists
(ACOG) revised its classification of the type of forceps
delivery, according to the station of the fetal vertex before
forceps application (Ali and Norwitz, 2009), which is as
follows:
1. Outlet forceps—the fetal vertex is visible at the labia
without manually separating them, and the fetal
skull has reached the pelvic floor
2. Low forceps—the leading point of the fetal skull is
greater than 2 cm beyond the ischial spines
3. Mid forceps—the fetal head is engaged and is
beyond the level of the ischial spines; the forceps
should be applied only if cesarean delivery is not
quickly or imminently possible, and the fetus is in
distress; or there should be a high likelihood that
the forceps operation will be successful
4. High forceps—the vertex is not engaged (i.e., not
at the level of the ischial spines or beyond); under
these circumstances, the forceps must never be
applied.
The forceps are further divided into whether they are
rotational (sagittal suture is >45 degrees from the midline)
or nonrotational (sagittal suture <45 degrees from the
midline).
The indications for use of the obstetric forceps are:
1. Maternal exhaustion or inability to push (endotracheal intubation with sedation or paralysis; neuromuscular disease)
2. Fetal heart tracing not reassuring or fetal distress
3. Maternal contraindications to pushing (cardiopulmonary disease, cerebrovascular aneurysm)
4. After-coming head in a vaginal breech delivery
5. Facilitate difficult extraction of the fetal vertex during cesarean delivery
FORCEPS AND POTENTIAL NEONATAL
MORBIDITY
As stated previously, forceps were used for hundreds of
years without regard to fetal survival and were primarily
needed to facilitate or terminate difficult labors for maternal benefit. Today, with the widespread availability of
cesarean delivery, considerations turn to providing the best
neonatal outcome possible; therefore the difficult forceps
deliveries of the past have been abandoned. Nevertheless,
forceps still play a crucial role in modern obstetrics, and if
judiciously used can provide a safer alternative to cesarean
delivery for both mother and baby.
The difficulty in interpreting the obstetric literature, in
regard to neonatal morbidity incurred by forceps, is that
the classification for type of forceps was revised by the
ACOG in 1988; therefore prior studies do not use the same
clinical criteria used currently to select appropriate candidates for forceps use. Furthermore, residency training in
operative vaginal delivery has dramatically decreased over
the past 30 years, potentially increasing fetal risk (Benedetto et al, 2007). Consequently, for proper interpretation
of adverse outcomes, one must look to studies performed
after the ACOG revised the classifications.
The incidence of operative vaginal delivery in the United
States is approximately 5% (1 in 20 deliveries), ranging from 1% to 23% with a 99% success rate (Menacker
and Martin, 2008). The prevalence of forceps use varies
widely by region (highest in the South), and recent estimates show that it accounts for approximately 25% (1:4
ratio with vacuum extractions) of all operative vaginal
deliveries (Benedetto et al, 2006). National statistics show
that of the 1,268,502 deliveries in 12 states (31% of U.S.
deliveries) in 2005, 1% were forceps assisted and 3.7%
were vacuum assisted, for a estimated total national operative vaginal delivery rate of 4.7% (Menacker and Martin,
2008). Unfortunately, the prevalence of low, outlet, and
mid-forceps (the application of forceps when the head is
engaged but the leading point of the skull is more than
+2 cm station) deliveries nationally is not known, nor is the
rate of rotational and nonrotational forceps use. Neonatal
outcome by type of forceps application is also not generally known and is hindered by the fact that even with the
universal ACOG classification scheme, the determination
of fetal station and type of forceps can be subjective and
dependent on the experience and examining skill of the
obstetrician. Moreover the indication for forceps use varies widely by clinical situation, and the neonatal morbidity that can result from a “difficult pull” in a patient with
a transverse arrest with marked fetal asynclitism may be
different from the quick delivery of a 2600-g fetus whose
mother is unable to push, even if both deliveries are by
low-forceps.
Nevertheless, what large studies show that long-term
and short-term neonatal morbidity from outlet or low-
forceps delivery is uncommon? In 2009, Prapas et al (2009)
noted that the rate of vacuum- versus forceps-assisted
deliveries had increased and that different maternal and
neonatal outcomes have been proposed. The aim of their
study was to compare the short-term maternal and neonatal outcomes between vacuum and forceps delivery. They
conducted a medical record review of live born singleton,
vacuum- and forceps-assisted deliveries. Of 7098 deliveries, 374 were instrument assisted, 324 were conducted by
vacuum (86.7%), and 50 were assisted by forceps (13.3%).
The incidence of third-degree lacerations and periurethral hematomas was similar between vacuum and forceps
(3.4% vs. 2% and 0.3% vs. 0%, respectively), whereas perineal hematomas were more common in forceps compared
CHAPTER 15 Complicated Deliveries: Overview
with vacuum application (2% vs. 0.3%, respectively), albeit
not significantly. The rate of neonates with Apgar scores
≤6 at 1 min was significantly higher after forceps compared with vacuum delivery (18% vs. 5.2%, respectively;
p = 0.0003). The rate of neonatal trauma and respiratory
distress syndrome did not differ significantly between
the two groups. The conclusion was that both modes of
instrumental vaginal delivery are safe in regard to maternal
morbidity and neonatal trauma (Prapas et al, 2009).
Alternatively, Benedetto et al, (2007) found that in
healthy women with antenatally normal singleton pregnancies at term, instrument-assisted deliveries are associated with the highest rate of short-term maternal and
neonatal complications. Of the 332 women who underwent an operative vaginal delivery, 201 met study criteria
and were analyzed, with 54% forceps-assisted deliveries.
Maternal complications were mostly associated with forceps-assisted and vacuum-assisted instrumental deliveries
(odds ratio [OR], 6.9; 95% confidence interval [CI], 2.9 to
16.4; and OR, 3.0; 95% CI, 1.1 to 8.8, respectively, versus spontaneous deliveries). Neonatal complications were
also mostly correlated with forceps-assisted and vacuumassisted instrumental deliveries (OR, 3.5; 95% CI, 1.9 to
6.7; and OR, 3.8; 95% CI, 2.0 to 7.4, respectively, versus spontaneous deliveries) (Table 15-2) (Bendetto et al,
2007).
There are few randomized prospective trials specifically
addressing the issue of neonatal morbidity arising from
forceps operations, yet those that exist suggest no significantly increased risk from operative vaginal delivery when
compared with spontaneous birth. Yancey et al (1991)
randomized 364 full-term women at +2 station to elective outlet forceps delivery or unassisted birth. Women
with suspected fetal macrosomia or chorioamnionitis were
excluded. Neonates were examined at birth and 72 hours
of age, and cranial ultrasound examinations were performed by neonatologists 24 to 72 hours after birth. The
spontaneous and forceps-assisted births had no statistically
significant differences in the incidence of scalp abrasions,
facial bruising, cephalhematoma, subconjunctival hemorrhage, or abnormal cranial sonograms (Herbst and Källén,
2008).
Several retrospective, population-based studies have
further examined the issue of potential adverse neonatal
sequelae arising from forceps procedures. Robertson et al
TABLE 15-2 Risk of Intracranial Injury According to Type
of Delivery
Mode of Delivery
Incidence of Intracranial
Injury
Vacuum
1 in 860
Forceps
1 in 664
Combined vacuum and forceps
1 in 256
Cesarean, in labor
1 in 907
Cesarean, not in labor
1 in 2750
Spontaneous vaginal delivery
1 in 1900
Adapted from Towner D, Castro MA, Eby-Wilkens E, Gilbert WM: Effect of mode of
delivery in nulliparous women on intracranial injury, N Engl J Med 341:1709-1714, 1999.
149
(1991) reclassified forceps operations in accordance with the
1988 revised ACOG guidelines and examined neonatal outcomes by type of forceps operation, matched to cesarean section with a second stage of at least 30 minutes, from a similar
station. They found that mid-forceps operations compared
with a cesarean section from a comparable station resulted in
greater neonatal resuscitation requirements, lower umbilical artery pH, and birth trauma, defined as nerve injuries
or fractures. There was no increased risk of trauma from
low forceps procedures compared with cesarean delivery,
although the prevalence of arterial cord pH less than 7.10
was increased (Keith et al, 1988). It must be noted, however,
that this study was not randomized and that an abdominal
delivery group is not necessarily an appropriate control for
operative vaginal delivery. Nevertheless, this large population-based study (from a database of 20,831) emphasizes the
relative safety of outlet and low-forceps procedures, casting
some doubt as to the safety of mid-forceps operations.
The role of mid-forceps delivery in modern obstetrics,
specifically in regard to rotation of 45 degrees or greater,
has incited much controversy. The difficulty lies in the
fact that no randomized trials exist comparing mid-forceps
operations and other modes of delivery, and it is unlikely
that any will be done in the near future given that training in this type of delivery has declined among obstetric
residents in the United States (Kozak and Weeks, 2002;
Learman, 1998).
Hankins et al reported a case-control study of 113 rotational forceps of 90 degrees or greater matched to 167
controls with rotation 45 degrees or less. They found no
significant differences in major injuries, defined as skull
fracture, brachial plexus, facial or sixth nerve palsy, and
subdural hemorrhage. There were also no differences in
the prevalence of cephalhematoma, clavicular fracture, or
superficial laceration. All deliveries were performed with
Kielland forceps by resident staff members, with attending
supervision as required. The prevalence of nerve injury in
these forceps-assisted operations ranged from 2% to 3%;
there were no skull fractures (Lipitz et al, 1989).
The available literature supports the fact that neonatal
morbidity from outlet or low forceps is exceedingly low, and
comparable to spontaneous vaginal delivery. This finding
comes from prospective data and large, population-based
investigations. The evidence concerning neonatal safety
from more advanced forceps operations (mid-forceps and
rotations) shows that there is some degree of increased morbidity from these procedures, although whether this arises
from the operative vaginal delivery itself or from difficult
labor is not currently possible to discern (Meniru, 1996;
O’Grady et al, 2002). In well-trained hands, the benefit of
appropriately selected candidates for mid-forceps or rotational forceps may be justifiable, although the number of
physicians in the United States who are facile and comfortable with attempting these procedures is steadily declining.
VACUUM DELIVERY: INDICATIONS, USES,
AND COMPARISON WITH FORCEPS
PROCEDURES
Operative vaginal delivery for the indications listed previously can also be performed by the vacuum extractor. Vacuum extraction was first described in 1705 by Dr. James
150
PART IV Labor and Delivery
Yonge, an English surgeon, several decades before the
invention of the obstetric forceps. However, it did not
gain widespread use until the 1950s, when it was popularized in a series of studies by the Swedish obstetrician
Dr. Tage Malmström. By the 1970s, the vacuum extractor
had almost completely replaced forceps for assisted vaginal deliveries in most northern European countries, but its
popularity in many English-speaking countries, including
the United States and the United Kingdom, was limited.
By 1992, however, the number of vacuum-assisted deliveries surpassed the number of forceps-assisted deliveries
in the United States, and by the year 2000 approximately
66% of operative vaginal deliveries were by vacuum (Ali
and Norwitz, 2009; Hillier and Johanson, 1994).
The situations that indicate the use of the vacuum and
the requirements that must be fulfilled for its correct use
are identical to those for the obstetric forceps. The device
consists of a metal or plastic cup (flexible or semirigid) that
is applied to the fetal vertex. Care is taken in its application
to ensure that an adequate seal has been created, and that no
maternal soft tissue is trapped between the suction device
and the fetus. Traction is then applied to the fetal head in
the line of the birth canal in an effort to assist delivery. It is
also cautioned that rocking movements or torque to the cup
are not used. The premature infant is a relative contraindication to vacuum application. It is generally advised that
no more than three detachments occur before attempts at
vacuum extraction are abandoned (Ali and Norwitz, 2009).
In a laboratory experiment, Duchon et al (1988) compared the maximum force at suggested vacuum pressures
(550-600 mm Hg) prior to detachments for different types
of vacuum devices. They found that the average force of
traction exerted before detachments ranged from 18 to 20
kg (Benedetto et al, 2007). This result is interesting to bear
in mind when one considers the older data of Wylie who
estimated the average tractive force required for delivery of
infants weighing 15.95 kg for a primigravida and 11.33 kg
for a multipara (Feingold et al, 1988) (Figure 15-2).
The original vacuum device developed in the 1950s by
the Swedish obstetrician Dr. Tage Malmström was a discshaped stainless steel cup attached to a metal chain for
traction. Because of technical problems and lack of experience with this instrument, vacuum devices did not gain
popularity in the United States until the introduction of
the disposable cups in the 1980s. There are two main types
FIGURE 15-2 A soft, bell-shaped vacuum extractor (top) and a rigid,
mushroom-shaped vacuum extractor (bottom). (Courtesy Cooper Surgical.)
of disposable cups, which can be made of plastic, polyethylene, or silicone. The soft cup is a pliable, funnel- or bellshaped cup, which is the most common type used in the
United States. The rigid cup is a firm mushroom-shaped
cup (M cup) similar to the original metal disc-shaped cup
and is available in three sizes.
A metaanalysis of 1375 women in nine trials comparing soft and rigid vacuum extractor cups demonstrated
that soft cups were more likely to fail to achieve a vaginal delivery, because of more frequent detachments (OR,
1.65; 95% CI, 1.19 to 2.29), but were associated with fewer
scalp injuries (OR, 0.45; 95% CI, 0.15 to 0.60) and no
increased risk of maternal perineal injury (Benedetto et al,
2007; Hillier and Johanson, 1994). For example, the risk
of scalp laceration with the rigid Kiwi OmniCup (Clinical Innovations, Murray, Utah) was reported to be 14.1%
compared with 4.5% using a standard vacuum device
(p = 0.006). These and other authors concluded that handheld soft bell cups should be considered for more straightforward occiput-anterior deliveries, and that rigid M cups
should be reserved for more complicated deliveries, such as
those involving larger infants, significant caput succedaneum
(scalp edema), occiput-posterior presentation, or asynclitism.
The vacuum extractor is widely used in the United
States, but is not free of preventing neonatal injury. Other
than superficial scalp lacerations or abrasions, which usually heal without incident, as well as local soft tissue swelling or bruising, the use of the vacuum has been associated
with cephalhematoma and subgaleal hemorrhages.
Cephalhematoma occurs when the force created by the
vacuum results in the rupture of diploic or emissary vessels
between the periosteum and outer table of the skull; this
fills the potential space that exists between the two with
blood. Although cephalhematomas are often cosmetically
alarming, they are limited to traveling along one cranial
bone, because the firm periosteal attachments limit further extravasation of blood across suture lines. Thus large
amounts of blood cannot usually collect in this space, and
serious neonatal compromise from this bleeding is rare.
In a randomized trial of continuous and intermittent vacuum application, Bofill examined factors associated with
increasing the risk of cephalhematoma; they found that
only asynclitism and traction time were independently
related to this complication (Hartley and Hitti, 2005).
There was a clear relationship between increasing time
of vacuum application (up to 6 minutes) and cephalhematoma. Interestingly, Hartley and Hitti did not find a significant independent association of neonatal injury with
continuous versus intermittent vacuum, or with decreasing gestational age or increasing birthweight. Furthermore, the number of detachments was not correlated with
cephalhematoma. These results were further corroborated
by Teng who conducted a prospective observational study
of 134 vacuum extractions and found that only increasing
total duration of vacuum application was associated with
neonatal injury (Feingold et al, 1988). Metaanalysis of randomized trials comparing vacuum to forceps extractions
showed that vacuums are more likely to fail to deliver the
baby and lead to increased rates of cephalhematoma and
retinal hemorrhage (Figure 15-3) (Vacca, 2007).
Subgaleal hemorrhage poses much more of a potential
risk for the neonate; it occurs when emissary veins above
CHAPTER 15 Complicated Deliveries: Overview
the skull and periosteum rupture, with blood dissecting
through the loose tissue underlying the cranial aponeurosis, unimpeded by suture lines. A tremendous amount
of blood, potentially the entire neonatal blood volume
(approximately 250 mL) can fill this space, and thus can
significantly compromise the neonate’s condition (Hillier
and Johanson, 1994). Much of the literature about this rare
complication of vacuum extraction was published in the
1970s and early 1980s, with few recent studies to detail
associated risk factors. Plauche, in his classic paper on
vacuum related neonatal injury, identified only 18 cases
of subgaleal hematomas among 14,276 cases of vacuumassisted births, in contrast to a mean incidence of cephalhematoma of 6% (Keith et al, 1988). These morbidity
151
estimates are derived from data that are approximately 30
to 40 years old; nevertheless, Teng noted an incidence of
cephalhematoma of 8%, and 0.7% for subgaleal hemorrhage in their more recent investigation, which agrees well
with Plauche’s estimates (Herbst and Källén, 2008).
A recent study from Australian investigators evaluated
37 cases of subgaleal hemorrhage at a single tertiary care
center accrued over a period of 23 years, with an estimated
prevalence of 1.54/10,000 total births. The finding was
that this complication occurred most often in primigravidae, and that a large proportion of these infants (89.1%
compared with 9.8% of the general control population)
had an attempted vacuum extraction (Figure 15-4) (Hillier
and Johanson, 1994; Kozak and Weeks, 2002).
Subcutaneous edema
Cephalhematoma
Periosteum
Bone
FIGURE 15-3 A cephalhematoma is a hemorrhage that occurs under the periosteum of the skull and is thus confined to a defined space with limited
capacity for expansion. (Adapted from Gilstrap LC, Cunningham FG, Hankins GDV, et al: Operative obstetrics, ed 2, Stamford, Conn, 2002, Appleton and Lange.)
Skin
Subgaleal
hematoma
Periosteum
Bone
FIGURE 15-4 A subgaleal hematoma spreads along subcutaneous soft tissue planes and has no immediate barrier to expansion, creating the potential for significant neonatal hemodynamic compromise. (Adapted from Gilstrap LC, Cunningham FG, Hankins GDV, et al: Operative obstetrics, ed 2,
Stamford, Conn, 2002, Appleton and Lange.)
152
PART IV Labor and Delivery
It must be kept in mind that the overall neonatal morbidity of the vacuum extractor is low, as ascertained from
the large population-based study by Towner showed that
the risk of intracranial injury is only 1 in 664 (Learman,
1998). Yet as outlined previously, there are definite neonatal risks associated with the use of vacuum extraction. The
U.S. Food and Drug Administration has suggested that
infants delivered with the vacuum have close monitoring
for subgaleal or subaponeurotic hematoma, and that a high
index of suspicion be maintained for this rare complication.
It must be emphasized that the overall risk of adverse
events attributable to the vacuum is extremely low, because
the U.S. Food and Drug Administration estimated five
serious complications per year of recent use during a time
period when 228,354 vacuum deliveries were performed.
However, it is likely that sequelae related to vacuum extraction often go underreported (Menacker and Martin, 2008).
The choice of which instrument to use, forceps or vacuum, is usually determined by the obstetric care provider
depending on the skill level and experience with either
method. There have been several randomized trials exploring this issue (Menacker and Martin, 2008, Meniru, 1996;
O’Grady et al, 2002; Prapas et al, 2009). The Cochrane
Library has pooled the results from 10 randomized trials comparing neonatal morbidity and successful vaginal
delivery between these two devices (Roberts et al, 2002).
This analysis found that the vacuum was more likely than
forceps to fail (OR, 1.69; 95% CI, 1.31 to 2.19), and was
associated with a greater likelihood of Apgar scores less
than 7 at 5 minutes (OR, 1.67; 95% CI, 0.99 to 2.81), cephalhematoma (OR, 2.38; 95% CI, 1.68 to 3.37), and retinal
hemorrhage (OR, 1.99; 95% CI, 1.35 to 2.98). However
the overall serious complication rate was low, and there
was no difference in long-term morbidity between groups
(Johnson and Menon, 2003).
The greatest danger in the use of either vacuum or
forceps comes in the combination of both instruments
together. Towner showed that the use of one instrument
after the other had failed and carried a neonatal intracranial injury risk of 1 per 256, significantly greater than
that of either modality alone (Learman, 1998). This finding was further supported by the work of Gardella et al,
(2001), who matched 11,223 women (a third of whom had
combined instruments, vacuum alone, or forceps alone,
respectively) to an equivalent number of spontaneous vaginal deliveries. These investigators found no statistically
significant difference in intracranial hemorrhage when a
single instrument was compared to spontaneous vaginal
delivery; however, the combined use of both instruments
markedly increased the risk of intracranial hemorrhage, seizures, and low 5-minute Apgar scores (Ron-El
et al, 1981). The overall risk of nerve and scalp injury was
greater when single-instrument delivery was compared
with spontaneous delivery, but the overall incidence of
each complication is rare.
The vacuum extractor is an acceptable instrument if
used judiciously and in the proper circumstances, carrying
an overall minimal risk of serious neonatal complications.
Its safety is comparable to the obstetric forceps, although
it has a higher incidence of cephalhematoma, but a lower
potential for facial nerve injury. The chance of failure is
greater with the vacuum, which could then potentially
tempt the health care provider to subsequently use the forceps. The use of both forceps and vacuum after one instrument has failed carries a higher risk of adverse outcomes,
and it should be undertaken only with an understanding of
the higher likelihood of neonatal morbidity.
SHOULDER DYSTOCIA
Shoulder dystocia is arguably the most dreaded complication in obstetrics. The problem posed by this entity is that
although it is highly anticipated, it is unpredictable and can
appear despite the most cautious measures taken to prevent it. Shoulder dystocia is defined as the delivery of the
fetal head with an impaction of the fetal shoulder girdle
or trunk against the pubic symphysis, making subsequent
delivery either difficult or impossible without performing
auxiliary delivery maneuvers. In some cases the posterior
shoulder may be lodged behind the sacral promontory—a
bilateral shoulder dystocia.
Once shoulder dystocia occurs, a series of maneuvers—which have never been tested in a prospective fashion, because of the sporadic and unpredictable nature of
this complication—are used to resolve it. The first step
is usually the McRoberts maneuver, which consists of
hyperflexing the maternal thighs onto the abdomen. This
maneuver flattens the pubic symphysis and sacral promontory and facilitates delivery of both the anterior and
posterior shoulders. If unsuccessful, this maneuver is usually followed by suprapubic pressure to remove the anterior shoulder from its impacted state behind the pubic
symphysis. If these two maneuvers fail, either rotational
maneuvers or extraction of the posterior fetal arm are usually tried. The Woods-Screw maneuver, or the Rubin’s
rotational maneuver, is used in an attempt to rotate the
infant’s shoulders in an effort to relieve the impaction of
the shoulder against the pubic bone. Alternatively, delivery of the posterior arm can be accomplished by inserting
the operator’s hand into the vagina and grasping the posterior fetal wrist and guiding it across the fetal chest and
through the vaginal introitus. It is often necessary to perform an episiotomy to have sufficient room in the vagina
to accomplish this maneuver. An alternative maneuver
to fetal manipulation is the all-fours position, or Gaskin
maneuver. With this maneuver, the mother is moved from
the lithotomy position to a hands and knees position. Next
the posterior fetal shoulder, which is now at the 12 o’clock
position, is delivered with gentle downward traction. If
the dystocia continues unresolved, the Zavanelli maneuver or cephalic replacement can be performed. After the
fetal head is rotated from occiput transverse to occiput
anterior, it is flexed and pushed back in the birth canal,
and the child was delivered by emergent cesarean section.
McRoberts maneuver, suprapubic pressure, or both will
relieve greater than 50% of instances. Cephalic replacement should be necessary in rare cases. Recent data have
focused on enhanced practitioner training for shoulder
dystocia by means of simulation. It is estimated that almost
50% of currently practicing obstetric birth attendants have
never successfully performed maneuvers other than the
McRoberts maneuver and suprapubic pressure. Draycott
et al (2008) have published data showing that the annual
compulsory training of all attending obstetric physicians
CHAPTER 15 Complicated Deliveries: Overview
in Southmead Hospital in the United Kingdom reduced
the incidence of fetal injury and brachial plexus injury
within 3 years of introduction. These data await confirmation in the United States or other countries.
The prevalence of shoulder dystocia varies depending
on the population studied and the presence of various risk
factors known to predispose women to this obstetric emergency. Estimates range from 0.2% to 1% in a low-risk population to 20% in higher-risk groups (Ali and Norwitz, 2009;
Benedetto et al, 2007; Feingold et al, 1988; Hartley and
Hittii, 2005; Herbst and Källén, 2008). Maternal obesity,
fetal macrosomia, history of prior shoulder dystocia, and
maternal diabetes mellitus are the most common associated
variables, but are not of sufficient prognostic power to be
clinically useful in predicting should dystocia (Ali and Norwitz, 2009; Benedetto et al, 2007; Herbst and Källén, 2008).
Because shoulder dystocia has the potential to cause
significant neonatal morbidity and mortality, efforts have
been made to predict its occurrence; unfortunately, no
clinical guidelines have been clinically tested or proved.
Ultrasound examination is commonly used in patients
with suspected fetal macrosomia or diabetes to detect large
birthweight in infants who might be more likely to suffer
shoulder dystocia. Third-trimester sonographic examination has an accuracy of 10% to 15% in the prediction
of fetal weight, and is thus not highly reliable (Feingold
et al, 1988; Hillier and Johanson, 1994; Keith et al, 1988).
In addition, if ultrasound examination were completely
reliable, the fetal weight cut-off that would prompt an
elective cesarean section has not been determined.
Lipscomb evaluated the deliveries of 227 mother-infant
pairs at their institution with birthweights greater than
4500 g and found a shoulder dystocia rate of 18.5%; therefore the majority of those delivering vaginally did so without any adverse event occurring (Hartley and Hitti, 2005).
In those with a shoulder dystocia, 51.7% infants had a neurologic injury, but at 2 months, all the infants had a normal
neurologic examination. Therefore the occurrence of brachial plexus injury at birth does not necessarily guarantee
permanent neurologic morbidity.
This conclusion has been reinforced by several other
studies. Gherman et al (2006) reviewed 285 cases of shoulder dystocia and found that 77 (24.9%) suffered fatal injury,
most commonly of the brachial plexus (16.8%) or clavicular (9.5%) and humeral (4.2%) fractures. Almost half of
the shoulder dystocias resolved with use of the McRoberts
maneuver alone; the rest required Woods’ maneuver, posterior arm extraction, or the Zavanelli maneuver. The requirement of additional fetal manipulative procedures increased
the risk of humeral fracture only and not clavicular or brachial plexus injury. The incidence of permanent musculoskeletal injury was only 1.4% (Kozak and Weeks, 2002).
A prospective investigation evaluated the natural history
of recovery following a birth-related brachial plexus injury
of infants referred to a tertiary care, multidisciplinary
neurological center. Enrollment required identification
of injury in the newborn period, initial evaluation at the
center between 1 and 2 months of age, and lack of antigravity movement in the shoulder or elbow persisting until
2 weeks of age. In this group of children subject to ascertainment bias (as those injuries resolving before 2 weeks of
age would not have been included in the results), complete
153
neurologic recovery was documented in 66%, and only
14% had persistent, severe weakness (Learman, 1998).
In the best systematic review of brachial plexus injury to
date, the risk of permanent brachial plexus impairment,
if recognizable at birth, was 15% to 20% (Pondaag et al,
2004). Rouse et al (1996) elaborated further on these
concepts in their decision analysis, which showed that if
one chose to perform an elective cesarean section for all
women without diabetes and with sonographically predicted macrosomia (estimated fetal weight >4000 g), 2345
cesarean sections would need to be performed to prevent
one permanent brachial plexus injury. If the 4500-g cutoff were selected, 50% more cesarean deliveries would be
needed to prevent one permanent brachial plexus injury.
In the mother with diabetes, if one chose a cut-off of 4500
g or greater, 443 cesareans would need to be done to prevent one permanent injury (Keith et al, 1988)—a tradeoff
that most practitioners now believe is acceptable. The
conclusions from this decision analysis have been borne
out by several other investigators who have established
that the risk of nerve injury certainly increases with rising
birthweight, but the large number of macrosomic infants
who have a normal, spontaneous vaginal delivery without
sequelae does not justify a policy of elective cesarean for
macrosomia alone in a nondiabetic population (Lipitz et al,
Loucopoulos and Jewewicz, 1982; Menacker and Martin,
2008; Meniru, 1996; O’Grady et al, 2002; Pondaag et al,
2004; Roberts et al, 2002).
Therefore at present there is no universally accepted
method to prevent shoulder dystocia. Studies have shown
that operative vaginal delivery, especially vacuum delivery,
of a fetus suspected to have macrosomia either clinically or
sonographically could increase the risk of shoulder dystocia
(Ron-El et al, 1981). It seems wise to avoid difficult forceps
or vacuum delivery if a patient is thought to have an infant
weighing more than 4000 g, especially if she has diabetes or
a past history of shoulder dystocia. In addition, the ACOG
states that “for pregnant women with diabetes who are suspected of carrying macrosomic fetuses, a planned cesarean
delivery may be a reasonable course of action, depending on
the incidence of shoulder dystocia, the accuracy of predicting
macrosomia, and the cesarean delivery rate within a specific
population” (Ali and Norwitz, 2009; Benedetto et al, 2007).
The documentation of the events surrounding shoulder dystocia are important, as is the discussion of current
status and future status of an infant delivered with a birth
injury after shoulder dystocia. Both obstetric and pediatric providers should debrief the shoulder dystocia event
immediately after the delivery. In the case of fetal injury,
it is optimal for both obstetric and pediatric providers to
discuss the delivery events and subsequent newborn treatment plans with the mother before discharge.
VAGINAL BREECH DELIVERY
Three percent to 4% of all infants at term will be in the
breech presentation at the time of delivery. There are three
main types of breech presentations. The footling breech
has one (single footling) or both (double footling) lower
extremities presenting. The frank breech has both thighs
flexed, but legs extended. The complete breech has both
thighs and legs flexed. The vaginal delivery of a singleton
154
PART IV Labor and Delivery
footling breech carries attendant risks of cord prolapse and
head entrapment, and the consensus among obstetricians
is that this presentation should be delivered operatively
(unless the fetus is a second twin, see later discussion on
Twin Delivery). The frank breech with buttocks presenting has a lower risk of these adverse events occurring, and
thus could potentially deliver vaginally. The complete
breech presentation will convert to frank or footling during labor, and the appropriate management scheme for
delivery depends on which leading fetal part will descend.
The mechanics of vaginal breech delivery are as follows. The frame of reference for the presenting part is
the sacrum (i.e., sacrum anterior, posterior, or transverse).
In the absence of urgent fetal indications, the singleton
breech is allowed to deliver passively with maternal expulsive efforts until the infant has been delivered past the
umbilicus. At this point the legs are gently reduced, and
the trunk and body are gently rotated to bring the sacrum
anteriorly. With the appearance of the scapula below the
maternal symphysis, the arms are then delivered by gently
sweeping them across the chest. Every effort is then made
to keep the neck from extending during the delivery of the
aftercoming head; this is accomplished during delivery of
the body by an assistant exerting suprapubic pressure on
the fetal head to keep it flexed. Once the body has delivered, the delivery of the head is accomplished by either
the Mariceau-Smellie-Veit maneuver, in which one hand
extends along the posterior neck and occiput and applies
pressure to prevent hyperextension, while the other hand
gently applies downward traction against the maxilla to
flex the head forward as the head is delivered, or with Piper
forceps directly applied to the fetal vertex.
The feasibility of vaginal breech delivery and its safety
have been the subject of much debate throughout the past
half century. With the advent of safe, expedient cesarean
delivery in the United States, many obstetricians have
favored the operative approach as the method of choice
for management of the breech presentation at term. The
literature to support this point of view has produced conflicting conclusions, and its interpretation is consequently
difficult. There has been an extensive body of literature
over the past half century examining this issue. Unfortunately, there are only two randomized trials that have
explored the question of which delivery route is best for
the term singleton frank breech fetus (Ali and Norwitz,
2009; Benedetto et al, 2007), but there are several large
retrospective series describing neonatal outcomes with
the vaginal approach, most of which suggest that vaginal delivery in carefully selected patients carries a low
risk of long-term neonatal morbidity and mortality. Diro
et al (1999) evaluated 1021 term singleton breech deliveries occurring at their institution over a 4-year period.
Infants with a clinically adequate pelvis and frank breech
presentation less than 3750 g were allowed a trial of labor.
They found an overall cesarean rate of 85.6%; however,
for women allowed to deliver vaginally, the success rate,
defined as vaginal delivery, was 50% (19 of 38 patients)
for nulliparous women and 75.8% (116 of 153 patients)
for multiparous women. The length of neonatal intensive
care unit (NICU) stay was higher for the group delivered
vaginally (17.4% vs. 12.1%; p = 0.036), but major morbidities between operative and vaginal delivery were not
significantly different (Feingold et al, 1988). Long-term
outcome was not evaluated. Of note, the women in this
cohort had pelvic dimensions evaluated clinically, and not
by x-ray or computed tomography pelvimetry, as has been
performed in other studies.
Norwegian investigators examined their similar policy of
vaginal breech delivery and evaluated maternal and neonatal outcomes in a large cohort of women (Fang and Zelop,
2006). Patients were allowed a trial of vaginal delivery if
they had adequate x-ray pelvimetry, and they were excluded
if they had an estimated fetal weight greater than 4500 g or
a footling presentation. Each vaginal breech was matched
to a term vaginal vertex birth; each cesarean delivery was
matched to the appropriate vertex control; 1212 breech
deliveries were evaluated, 639 (52.7%) of which were vaginal, 172 (11.4%) were intrapartum cesarean section, and
138 (11.4%) were planned cesarean section. Once major or
lethal anomalies and fetal disorders not related to delivery
were excluded, there were no perinatal deaths attributable
to mode of delivery. When births planned vaginally were
compared with operative deliveries, there was an increased
risk of 1-minute Apgar score less than 7 and traumatic morbidity in the vaginal group, but no significant differences in
NICU admissions, 5-minute Apgar scores, or normal neonatal course. The conclusion reached was that short-term
morbidity was worse in the group delivered vaginally, but
long-term outcome was similar between groups. It must be
remembered, however, that this study was not randomized,
and the infants selected for vaginal delivery versus cesarean
section were intrinsically different. In addition, all deliveries occurred in a tertiary care institution.
The truly interesting point raised by these investigators
is that, aside from the issue of cesarean versus trial of labor,
singleton breech infants regardless of mode of delivery
have an increased risk of morbidity compared with their
vertex counterparts. Breech infants had higher incidences
of NICU admissions, eventful courses, hip dislocation,
and traumatic morbidity (soft tissue trauma, fracture, facial
nerve paralysis, and brachial plexus palsy). Thus both the
obstetrician and pediatrician must be aware that the infant
in breech presentation requires careful attention upon
birth for the presence of these potential factors.
Christian evaluated their policy of offering a trial of
vaginal breech delivery to women with an estimated fetal
weight between 2000 and 4000 g and having computed
tomographic pelvimetry documenting adequate pelvic
dimensions (Herbst and Källén, 2008). Of 122 women
evaluated, 85 were judged appropriate by these standards
for vaginal delivery, of which 81.2% had a successful
vaginal delivery. The only indices of neonatal outcome
evaluated were the Apgar score, which was not different
between groups, and neonatal cord pH, which was lower
in a statistically but not clinically significant manner in the
vaginal delivery group.
The largest difficulty in interpreting the large number of
retrospective studies examining the issue of vaginal breech
delivery in the obstetric literature is that even the best
designed reports have relatively small numbers of patients,
and the possibility of a type II or beta error is high. In addition, the groups being compared (planned cesarean section
and vaginal delivery) are different because the patients
are not randomized (Herbst and Källén, 2008; Hillier
CHAPTER 15 Complicated Deliveries: Overview
and Johanson, 1994). Often the women chosen to have a
cesarean section tend to have factors that would place their
neonates at higher risk than those allowed a trial of labor.
A metaanalysis evaluating seven cohort studies and two
randomized trials compared 1825 trial of labor patients
to 1231 elective cesarean patients and found a statistically
significant, but clinically questionable, increased risk with
vaginal breech delivery of 1.10% (Keith et al, 1988).
There is a paucity of randomized trials to specifically
address the role of the vaginal breech delivery in modern obstetrics. Collea et al randomized 208 women with
a singleton frank breech presentation at term to vaginal
delivery or cesarean section; they found a low overall risk
of permanent birth injury or neonatal morbidity in the
vaginal delivery group, although the incidence of neonatal
morbidity was higher in the vaginal delivery group (Ali and
Norwitz, 2009). Of note, a majority of conditions listed
as morbidities (hyperbilirubinemia, meconium aspiration,
mild brachial plexus injury) had resolved by hospital discharge. In addition, decreased neonatal morbidity with
cesarean section was offset by a striking increase in higher
maternal risk in the operative group. It must be remembered that this study was published in 1979, and standards
of maternal and neonatal care have changed dramatically
since then. Until 2001, no additional randomized investigations were available to settle the long raging debate of
the optimal mode of delivery for term singleton breech.
This controversy has recently been clarified by a large,
multicenter, multinational trial that randomized 2088
women at 121 centers in 26 countries to planned vaginal birth or planned elective cesarean section (Benedetto
et al, 2007). Criteria for enrollment were frank or complete term singleton breech with no evidence of fetal macrosomia. The investigation was halted when preliminary
results showed that there was significantly increased neonatal mortality and severe morbidity in the vaginal breech
arm compared with the cesarean arm (5.0% vs. 1.6%). This
conclusion was not altered by the experience of the delivering obstetrician or maternal demographic factors such as
parity and race. Maternal morbidity between both groups
was comparable. The difference in outcome was even more
striking in countries, such as the United States, with a low
national perinatal mortality rate (5.7% vs. 0.4%).
Criticisms of this study are that patients enrolled did not
have computed tomography pelvimetry performed, which
in some institutions is standard practice before considering a vaginal breech delivery. Furthermore, subjects did
not have continuous fetal monitoring, but rather intermittent fetal auscultation every 15 minutes. In addition, the
capability of various centers to perform emergent cesarean sections differs markedly, and this could have potentially affected the neonatal morbidity and mortality rate.
Nevertheless, it is unlikely that another large study will
ever be performed to examine this issue again, and the
ultimate results are difficult to dispute given the excellent study design and adequate sample size. Indeed the
ACOG recently issued a statement that “planned vaginal delivery of a term singleton breech may no longer be
appropriate … patients with a persistent breech presentation at term in a singleton gestation should undergo a
planned cesarean delivery” (Kozak and Weeks, 2002). As
will be discussed later in Multifetal Delivery, this statement
155
does not apply to the vaginal delivery of a nonvertex second twin.
The delivery of the vaginal breech is also an emotional
issue; physicians trained in the art of the vaginal breech
delivery maintain that for an appropriately selected candidate, vaginal breech delivery has acceptable neonatal risk
and has the advantage of sparing the mother significant
operative morbidity. Proponents of cesarean delivery further state that the level of resident training in the art of
the singleton vaginal breech delivery has markedly diminished, with most graduating senior residents having performed few such births. Nonetheless, many practitioners
will be required to assist in vaginal birth of a breech infant
in unplanned situations. The acquisition of skills necessary to competently perform this procedure may need to
be learned and practiced with simulation-based training,
because the opportunities for training in most residency
programs are few.
MULTIFETAL DELIVERY
With the advent of in vitro fertilization and the sophisticated assisted reproductive technologies (ARTs),
the incidence of multifetal pregnancies has dramatically increased, particularly higher-order multiples. The
incidence of twin gestations in patients undergoing in
vitro fertilization is currently more than 30%, and it is
1% to 3% in higher-order multiples. As a result, the frequency of twin gestation in the United States has increased
65% since 1980; twins now account for 3% of births. Of
these, 80% are dizygotic and 20% are monozygotic. This
3% of births accounts for 17% of preterm births and
approximately 25% of infants of low birthweight and very
low birthweight. The perinatal mortality rate of twins is sevenfold that of singletons, of which a small fraction is due to
problems during labor and at delivery. The mode of delivery
for twins is well delineated by several studies, and the issues
surrounding the choice of vaginal birth versus cesarean is
outlined in the following sections (Smith et al, 2005).
TWIN DELIVERY
Vertex-Vertex
It is almost universally accepted that the appropriate
method of delivery is vaginal if both twins are vertex. The
first infant is delivered like a singleton infant. The second
infant is delivered in a similar fashion, but care must be
taken not to rupture membranes before the head is well
engaged, because this may increase the risk of cord accident. Of note, the delivery of the second twin does not
necessarily occur immediately after the first.
Vertex-Nonvertex
The first twin is usually delivered vaginally. The options for
delivery of the second twin are as follows: cesarean section,
breech extraction, or attempts at external cephalic version
and vertex delivery of the second twin if successful. The
subject of the optimal delivery choice for the second twin
has been the subject of much controversy. Many obstetricians claim that cesarean section is the safest approach
156
PART IV Labor and Delivery
to the nonvertex twin, whereas others claim that vaginal
delivery affords equivalent neonatal outcome, sparing the
mother from an unnecessary surgical procedure (Usta et al,
2005). Fortunately there is a large body of evidence in the
literature addressing these issues.
If the vaginal approach is chosen, once the first twin
is delivered the obstetrician inserts a hand into the uterine cavity and, under sonographic guidance if necessary,
finds the feet of the second twin. Once the feet are firmly
grasped, they are brought down into the vagina, and the
membranes are then ruptured. Traction is applied to the
fetus along the pelvic curve; once the body has been delivered through the introitus, delivery of the arms, shoulders,
and aftercoming head proceed in a fashion similar to that
of a singleton breech.
In 2003, Hogle et al performed a metaanalysis to determine whether a policy of planned cesarean or planned
vaginal birth is preferable for twins. They found only four
studies with a total of 1932 infants met their inclusion criteria. There were no significant differences in maternal
morbidity, perinatal or neonatal mortality, or neonatal
morbidity between the two groups. They did find significantly fewer low 5-minute Apgar scores in the planned
cesarean group, principally because of a reduction among
breech first twins. They concluded that, if twin A is vertex,
“there is no evidence to support planned cesarean section
for twins.” In contrast, Smith et al (2005) published a retrospective cohort study of 8073 twin births after 36 weeks
of gestation in Scotland between 1985 and 2001. There
was a death of either twin in two of 1472 (0.14%) deliveries
by planned cesarean and in 34 of 6601 (0.52%) deliveries
by other means (p = 0.05; OR for planned cesarean, 0.26;
95% CI, 0.03 to 1.03). They concluded that planned cesarean may reduce the risk of perinatal deaths of twins at term
by 75% despite the lack of statistical significance in outcomes
between the two groups. The data also suffer from the fact
that 30 of the 36 deaths were in second twins, and there were
no data regarding fetal presentation (Smith et al, 2004).
There are several large cohort studies examining the
issue of feasibility and safety of total breech extraction
of the nonvertex second twin. These studies have almost
unanimously reached the similar conclusion that the neonatal outcome for nonvertex second twins delivered vaginally is similar to the vertex first twin, but is not statistically
different from those second twins delivered by cesarean section, regardless of birthweight or gestational age
(Ali and Norwitz, 2009; Benedetto et al, 2007; Feingold
et al, 1988; Hartley et al, 2005; Herbst and Källén, 2008;
Hillier and Johanson, 1994; Keith et al, 1988). Hartley et al
(2005) conducted a retrospective analysis of birth certificates and fetal and infant death certificates for 5138 twin
pairs selected from those born in Washington State from
1989 to 2001. They concluded that if prompt vaginal
delivery of twin B does not occur, the benefits of vaginal
delivery for twin A might not outweigh the risks of distress
and low Apgar scores in twin B and vaginal plus cesarean
delivery for the mother (Hartley and Hitti, 2005).
Although there are several retrospective studies evaluating the outcome of vaginally born nonvertex second twins,
there is only one randomized trial (Hillier and Johanson,
1994). Rabinovici et al (1987) allocated 60 women with vertex-nonvertex twins to either operative or vaginal delivery.
Maternal morbidity and hospital stay were increased in the
surgical group, but there were no differences in neonatal
outcome. In addition, Acker et al (1982) retrospectively
reviewed 150 nonvertex second twins of all birthweights, 74
delivered by cesarean and 76 by breech extraction, and they
found no mortality in either group and a 3.9% incidence of
low Apgar scores in the breech extraction group, which was
no different from that in the cesarean group. Chervenak
et al (1986) reviewed 76 breech extractions for second
twins, found no morbidity or mortality in the 60% weighing more than 1500 g, and concluded “for birthweights
>1500 gm, routine cesarean for vertex/nonvertex twins
may not be necessary.”
The available body of evidence supports attempts at
vaginal delivery of the nonvertex second twin. Of course
the responsible obstetrician must choose a management
plan most compatible with his or her experience and training. For those not versed in the techniques of successful
vaginal breech extraction, cesarean delivery might be a
more prudent plan. As in the case of the singleton vaginal
breech, simulation training may play a role in the acquisition and maintenance of skills for safe vaginal breech birth.
In addition to abdominal delivery and total breech extraction, there is the option of external cephalic version (i.e.,
attempting to turn a nonvertex fetus to vertex by abdominal
manipulation). Studies have shown that this option is associated with a higher failure rate at successful vaginal delivery and other complications (such as cord accidents and
malpresentations not amenable to vaginal delivery) when
compared with primary breech extraction or cesarean section (Hillier and Johanson, 1994; Learman, 1998).
Nonvertex-Nonvertex
Because of to the theoretical risk of interlocking twins, as
well as the recent data showing the greater morbidity for
the singleton vaginal breech (see the preceding discussion),
cesarean section is the recommended choice for delivery of
the nonvertex-nonvertex presentation.
Monochorionic, Monoamniotic Twins
Monochorionic, monoamniotic twins share a single intra
amniotic space, and thus have a higher risk of cord and
extremity entanglement during the course of delivery. It is
commonly accepted that the optimal mode of delivery is a
planned cesarean section before labor ensues.
HIGHER-ORDER MULTIPLE GESTATIONS
Most perinatologists would suggest cesarean delivery for
triplets and higher-order multiples (Lipitz et al, 1989).
Although this practice is common, the data mandating
operative delivery are far from conclusive.
A Dutch study compared the outcomes of triplets delivered vaginally and abdominally at two institutions (Learman, 1998). One hospital favored cesarean section, whereas
at another trial of labor was offered to all appropriate candidates. The success of vaginal delivery was relatively high
(34 of 39 women [87%]). There was a higher incidence
of neonatal mortality and postdelivery depression (as estimated by Apgar score) in the hospital favoring operative
CHAPTER 15 Complicated Deliveries: Overview
delivery compared with the vaginal delivery group. The
biases inherent in this study are obvious, although the
reported findings have been corroborated by several other
reports from single institutions that offer trial of vaginal delivery to triplet gestations (Menacker and Martin,
2008; Meniru, 1996; O’Grady et al, 2002). Vintzileos et al
(2005) attempted to estimate the risks of stillbirth and
neonatal and infant deaths in triplets, according to mode
of delivery; they used the “matched multiple birth” data
file that was composed of triple births that were delivered in the United States during 1995 through 1998 and
found that 95% of all triplets were delivered by cesarean
delivery. Vaginal delivery (all vaginal) was associated with
an increased risk for stillbirth (relative risk, 5.70; 95%
CI, 3.83 to 8.49) and neonatal (relative risk, 2.83; 95%
CI, 1.91 to 4.19) and infant (relative risk, 2.29; 95% CI,
1.61 to 3.25) deaths. They concluded that cesarean delivery of all three triplet fetuses is associated with the lowest
neonatal and infant mortality rate and that vaginal delivery among triplet gestations should be avoided (Vintzileos
et al, 2005). Most of the data on triplet births consists of
small cohort studies and not randomized trials, and the
possibility of type II or beta errors exists in the interpretation of many of these studies (Feingold et al, 1988; Keith
et al, 1988). Thus delivery of triplet gestations vaginally
while not an unreasonable approach has nearly disappeared from the practice of modern obstetrics in favor of
routine cesarean delivery. Currently quadruplets and other
higher-order multiples are usually delivered by cesarean
section (Ron-El et al, 1981).
VAGINAL BIRTH AFTER CESAREAN:
NEONATAL ISSUES
Cesarean section accounts nationally for one quarter
of all deliveries (Ali and Norwitz, 2009). Surgery carries the maternal risks of increased blood loss, prolonged
hospital stay, and longer recovery period compared with
vaginal delivery. During the 1980s to 1990s, efforts were
made to encourage women to attempt vaginal birth after
a prior cesarean delivery, because success rates vary from
60% to 80% for vaginal delivery, dependent on the indications for the prior cesarean delivery (Benedetto et al,
2007). Whereas VBAC rates remain relatively high in the
United Kingdom at 33% (range, 6% to 64%), the rates
are decreasing rapidly in the United States from a high
rate of 28.3% in 1996 to less than 10 per 1000 deliveries in 2006 (Caughey, 2009; Fang and Zelop, 2006). This
decrease occurred primarily because VBAC can result in
uterine dehiscence, in which the prior scar asymptomatically separates or, more seriously, uterine rupture occurs.
This decrease has significantly affected the United States
cesarean section rate of 31.1% (Caughey, 2009; Fang and
Zelop, 2006). A full discussion of VBAC, studies supporting its safety, and controversies surrounding its feasibility is beyond the scope of this chapter, and the interested
reader is urged to consult Williams Obstetrics, ed 23, for
further details. This discussion instead focuses on neonatal
risks from VBAC, particularly from its most dreaded complication, uterine rupture.
Studies have uniformly shown a risk of uterine rupture with VBAC on the order of 0.5% to 1% (Benedetto
157
et al, 2007; Caughey, 2009; Feingold et al, 1988). A recent
large, retrospective study evaluated 20,095 women with a
history of prior cesarean delivery and found that rupture
risk was 0.16% if the woman elected a repeated, elective
operative delivery; 0.52% if VBAC occurred as a result of
spontaneous labor; 0.77% if labor was induced without
prostaglandins; and 2.5% if labor was induced with prostaglandins (Feingold et al, 1988). Thus VBAC carries the
lowest risk if labor is spontaneous and not augmented.
There are few large, well-designed studies specifically
evaluating neonatal rather than maternal outcomes in
VBAC. Most recently, Kamath et al (2009) performed a
retrospective cohort study of 672 women with one prior
cesarean section undergoing trial of labor. They found that
infants born by cesarean delivery had higher rates of admission to the NICU (9.3% compared with 4.9%) and higher
rates of oxygen supplementation for delivery room resuscitation (41.5% compared with 23.2).
Yap retrospectively evaluated 38,027 deliveries occurring at a single tertiary care institution and found 21 cases
of uterine rupture; 17 occurred after a history of a prior
cesarean delivery (Herbst and Källén, 2008). The two
neonatal deaths that occurred were a result of prematurity (23-week-old fetus) and multiple congenital anomalies; all live born infants were discharged from the hospital
without neurologic sequelae. Thus the ultimate neonatal
outcome despite uterine rupture was favorable. However,
all deliveries occurred in a tertiary care institution with
readily available obstetric anesthesiologists, neonatologists, and obstetricians. Most deliveries after diagnosis of
rupture occurred within 26 minutes.
A third group of investigators retrospectively identified
99 cases of uterine rupture occurring over a period including 159,456 births (Hillier and Johanson, 1994). Thirteen
of these ruptures occurred before the onset of labor. There
were six neonatal deaths, but four of these occurred in
women with uterine rupture at admission, and thus were
never given a trial of labor. There were five cases of perinatal asphyxia, but once again it is not detailed whether
these occurred in women allowed a trial of labor or in
those who had ruptured on presentation to the hospital.
Moreover, many of these women had an undocumented
prior scar, which in some institutions would warrant an
elective repeated cesarean section. The aforementioned
recent study evaluating 20,095 women with a prior cesarean delivery and their subsequent risk of rupture found a
neonatal mortality of 5.5% (Feingold et al, 1988). However, because this was a population based study, it was
not specified whether these deliveries occurred in tertiary
care institutions with the capability of performing emergent operative rescue procedures in the event of uterine
rupture.
Finally Fang et al (2006) reviewed all of the literature
to date in regard to adverse neonatal outcomes and found
that the combined rates of intrapartum stillbirth and neonatal death were not statistically different between trial of
labor and those who elected for repeated cesarean section.
Thus the true neonatal risk of VBAC, especially in the
event of uterine rupture, cannot be precisely estimated at
the current time. There are no studies adequately evaluating long-term outcomes of surviving infants after uterine
rupture (Fang and Zelop, 2006).
158
PART IV Labor and Delivery
It appears that the risk of uterine rupture after a prior
cesarean delivery is low, but this risk increases when labor
is augmented with oxytocin or prostaglandins. It is appropriate to offer women VBAC, but they must be counseled
carefully about the potential risk of uterine rupture. Careful documentation of the informed consent and labor management must be completed. Moreover, VBAC should
ideally occur in a tertiary care institution or in facilities
capable of rapidly performing an emergent cesarean section, because this improves the likelihood of minimizing
adverse neonatal sequelae.
CORD ACCIDENTS
The term cord accident usually refers to adverse events
affecting the fetus that occur as a result of a problem with
the umbilical cord. This heterogeneous term encompasses umbilical cord prolapse, in which the cord delivers
through the cervix and compression by a fetal part results
in a significantly increased risk of asphyxia; it also includes
such entities as cord entanglements or “true knots,” which
can lead to fetal compromise.
The incidence of such events is not clearly known,
because the diagnosis is often one of exclusion. One
large population-based study compared 709 cases of
cord prolapse occurring among 313,000 deliveries to
matched controls and found that low birthweight, male
sex, multiple gestations, breech presentation, and congenital anomalies all increased the risk of umbilical cord
prolapse (Ali and Norwitz, 2009). Not surprisingly, cord
prolapse was associated with a high mortality rate (10%)
that was reduced if cesarean rather than vaginal delivery
was performed.
The standard of care in cases of cord prolapse is to proceed immediately with cesarean section as quickly as possible while an assistant elevates the presenting fetal part
with a vaginal hand to prevent compression of the umbilical cord. It is also of paramount importance to have appropriate pediatric support available at the time of delivery,
because the newborn is likely to be depressed and require
resuscitation.
Cord accident, or in utero compromise, secondary to
entanglement of the umbilical cord as a clinical entity is
difficult to understand. Often in cases of in utero fetal
demise (IUFD), a diagnosis or cause of fetal death is never
found. It is tempting to attribute the demise to an event
that compromises umbilical blood flow to the developing pregnancy. The literature on this subject is scarce.
Hershkovitz et al (2001) identified 841 cases of true knots
from a population of 69,139 deliveries (for a prevalence
of 1.2%) and in a case-controlled study found that grand
multiparity (>10 deliveries), chronic hypertension, history of genetic amniocentesis, male gender, and umbilical
cord prolapse were all independently associated with true
knots of the umbilical cord. The presence of a true knot
was associated with both in utero fetal demise and greater
FIGURE 15-5 (See also Color Plate 2.) A 28-year-old woman was
admitted to the labor and delivery department with an intrauterine
demise. Examination of the fetus shows the cord wrapped tightly around
the torso, leg, and ankle, suggesting cord accident as a cause of death.
No other pathologic abnormalities were found on autopsy. (Courtesy
Thomas R. Easterling.)
likelihood of cesarean delivery (Figure 15-5) (Benedetto
et al, 1994).
SUGGESTED READINGS
American College of Obstetricians and Gynecologists: ACOG Committee Opinion
No. 340: Mode of term singleton breech delivery, 108:235-237, 2006.
American College of Obstetricians and Gynecologists: Fetal macrosomia. ACOG
Practice Bulletin No. 22, Obstet Gynecol 96 , 2000.
American College of Obstetricians and Gynecologists: Operative vaginal delivery.
ACOG Practice Bulletin No. 17, Obstet Gynecol 95 , 2000.
American College of Obstetricians and Gynecologists: Shoulder dystocia. ACOG
Practice Bulletin No. 40, Obstet Gynecol 100:1045-1050, 2002.
American College of Obstetricians and Gynecologists: Vaginal birth after cesarean
section. ACOG Practice Bulletin No. 54, Obstet Gynecol 104:203-212, 2004.
Draycott TJ, Crofts JF, Ash JP, et al: Improving neonatal outcome through practical shoulder dystocia training, Obstet Gynecol 112:14-20, 2008.
Fang YM, Zelop CM: Vaginal birth after cesarean: assessing maternal and perinatal
risks: contemporary management, Clin Obstet Gynecol 49:147-153, 2006.
Hartley RS, Hitti J: Birth order and delivery interval: analysis of twin pair perinatal
outcomes, J Matern Fetal Neonatal Med 17:375-380, 2005.
Johanson RB, Menon BK: Vacuum extraction versus forceps for assisted vaginal
delivery, Cochrane Database Syst Review (2):CD000224, 2000.
Kamath BD, Todd JK, Glazner JE, et al: Neonatal outcomes after elective cesarean
delivery, Obstet Gynecol 113:1231-1238, 2009.
Prapas N, Kalogiannidis I, Masoura S, et al: Operative vaginal delivery in singleton
term pregnancies: short-term maternal and neonatal outcomes, Hippokratia
13:41-45, 2009.
Thorngren-Jerneck K, Herbst A: Low 5-minute Apgar score: a population-based
register study of 1 million term births, Obstet Gynecol 98:65-70, 2001.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
16
Obstetric Analgesia and Anesthesia
Mark D. Rollins and Mark A. Rosen
This chapter introduces some of the scientific background
and clinical techniques used in providing obstetric analgesia and anesthesia. These practices provide substantial
benefit to the patient in labor and are essential for operative delivery. Although the effects of obstetric analgesia and anesthesia on the fetus and neonate are typically
benign, there is potential for significant neonatal effects.
HISTORY OF OBSTETRIC ANESTHESIA
Modern obstetric anesthesia began in Edinburgh, Scotland, on January 19, 1847, when the professor of obstetrics
James Young Simpson used diethyl ether to facilitate child
delivery by anesthetizing a woman with a contracted pelvis.
Morton’s historic demonstration of the anesthetic properties of ether at the Massachusetts General Hospital in
Boston had occurred only 3 months earlier. Fanny Longfellow, wife of Henry Wadsworth Longfellow, was the first
American to receive anesthesia for childbirth, publicly proclaiming in 1847, “This is certainly the greatest blessing of
this age” (Longfellow and Wagenknecht, 1956).
Although anesthesia for surgery was rapidly and widely
accepted, most of Simpson’s contemporaries in the United
States, France, and England were critical of using anesthesia in obstetrics, presenting both medical and religious
arguments in outspoken opposition. However, the public
outcry in support of labor analgesia was vehement. In 1853,
the negative reaction from Thomas Wolsley, editor of The
Lancet, after John Snow administered ether for Queen
Victoria’s eighth child was so strong that court physicians
denied anesthesia had been used. The great debate was
largely settled 4 years later when Victoria delivered her
ninth and last child, and the use of a royal anesthetic was
acknowledged. Although many physicians had remained
opposed, public opinion had changed and women were
requesting labor analgesia from their doctors.
During the second half of the twentieth century, anesthesiologists made significant advances in techniques and
improved safety for delivering labor analgesia. Hingson and Edwards (1943) developed the continuous caudal catheter that preceded development of the epidural
catheter. Apgar (1953) initially proposed a simple neonatal scoring system as a guide for evaluating the effects of
obstetric anesthesia and later as a guide for neonatal resuscitation. Other early pioneers in the emerging specialty of
obstetric anesthesia were Gertie Marx (Marx and Orkin,
1958), John Bonica (1967), and Sol Shnider et al (1963).
These pioneers helped to characterize the normal changes
in maternal physiology related to pregnancy, confirm the
safety and efficacy of obstetric analgesia, determine the
effects of these techniques on uterine blood flow and placental transfer of anesthetic agents, and evaluate the effects
of these techniques and agents on newborn well-being.
ANATOMY OF LABOR PAIN
Contraction of the uterus, dilatation of the cervix, and distention of the perineum cause pain during labor and delivery. Somatic and visceral afferent sensory fibers from the
uterus and cervix travel with sympathetic nerve fibers to
the spinal cord (Figure 16-1). These fibers pass through
the paracervical tissue and course with the hypogastric
nerves and the sympathetic chain to enter the spinal cord
at T10 to L1. During the first stage of labor (cervical
dilation), the majority of painful stimuli are the result of
afferent nerve impulses from the lower uterine segment
and cervix, as well as contributions from the uterine body
causing visceral pain (poorly localized, diffuse, and usually
described as “a dull but intense aching”). These nerve cell
bodies are located in the dorsal root ganglia of levels T10 to
L1. During the second stage of labor (pushing and expulsion), afferents innervating the vagina and perineum cause
somatic pain (well localized and described as “sharp”).
These somatic impulses travel primarily via the pudendal
nerve to dorsal root ganglia of levels S2 to S4. Pain during
this stage is caused by distention and tissue ischemia of the
vagina, perineum, and pelvic floor muscles, associated with
descent of the fetus into the pelvis and delivery. Neuraxial
analgesic techniques that block levels T10 to L1 during
the first stage of labor must be extended to include S2 to
S4 for efficacy during the second stage of labor.
Labor pain can have significant physiologic effects on
the mother, fetus, and the course of labor. Pain stimulates the sympathetic nervous system, elevates plasma
catecholamine levels, creates reflex maternal tachycardia
and hypertension, and reduces uterine blood flow. In addition, changes in uterine activity can occur with the rapid
decrease in plasma epinephrine concentrations associated
with onset of neuraxial analgesia. Oscillations in epinephrine can result in a range of uterine effects from a transient
period of uterine hyperstimulation (Clarke et al, 1994)
to a transient period of uterine quiescence, or conversion of dysfunctional uterine activity patterns associated
with poorly progressive cervical dilation to more regular
patterns associated with normal cervical dilation (Leighton
et al, 1999).
CHANGES IN MATERNAL
PHYSIOLOGY AND THE IMPLICATIONS
During pregnancy, labor, and delivery, women undergo
fundamental changes in anatomy and physiology. These
alterations are caused by changing hormonal activity,
biochemical shifts associated with increasing metabolic
demands of a growing fetus, placenta, and uterus, and
mechanical displacement by an enlarging uterus (Cheek
and Gutsche, 2002; Parer et al, 2002).
159
160
PART IV Labor and Delivery
T 10
11
12
L1
T-10
11
12
L-1
3
4
2
FIGURE 16-1 Parturition pain pathways. Nerves that accompany
sympathetic fibers and enter the neuraxis at the T10, T11, T12, and L1
spinal levels carry afferent pain impulses from the cervix and uterus.
Pain pathways from the perineum travel to S2, S3, and S4 via the
pudendal nerve. (From Bonica JJ: Principles and practice of obstetric
analgesia and anesthesia, Philadelphia, 1967, F.A. Davis Co.)
MATERNAL CIRCULATORY SYSTEM
Hypotension can occur when a pregnant woman is in the
supine position because of compression of the vena cava
by the gravid uterus. Significant aortoiliac artery compression occurs in 15% to 20% of parturients and vena
caval compression is universal, often as early as 13 to 16
weeks’ gestation. Vena caval compression contributes to
lower extremity venous stasis and can cause ankle edema
and varices despite increased collateral circulation. Venous
compression by the gravid uterus diverts some blood
returning from the lower extremities through the internal
vertebral venous plexus, the azygos, and the epidural veins.
This increases the likelihood of entering an epidural vein
with spinal or epidural anesthetic techniques. Anesthetic
interventions that diminish sympathetic tone can further
exacerbate the effects of vena caval compression induced
by supine positioning, potentially causing profound hypotension. Therefore supine positioning is avoided during
anesthetic administration in the second and third trimesters. Significant lateral tilt is used in all operative deliveries and frequently during labor analgesia to help preserve
uterine blood flow and fetal circulation.
Cardiac output increases during pregnancy, reaching an
output 50% greater than the prepregnant state by the third
trimester. During labor, maternal cardiac output increases
during the first and second stages, reaching an additional
40% above prelabor values in the second stage (Robson
et al, 1987). Each uterine contraction results in the autotransfusion of 300 to 500 mL of blood back into the maternal central circulation. The greatest increase in cardiac
output occurs immediately after delivery, when values can
increase as much as 75% above predelivery levels. This
abrupt increase in cardiac output is secondary to the loss
of aortocaval compression, autotransfusion from the contracted uterus, and decreased venous pressure in the lower
extremities (Kjeldsen, 1979).
Physiologic (dilutional) anemia of pregnancy occurs as a
result of a greater increase in plasma volume (45%) than in
red blood cell volume (20%) at term. Average blood loss at
delivery—Approximately 500 mL for vaginal delivery and
1000 mL for cesarean section—is well tolerated because
of this expanded blood volume and autotransfusion (normally in excess of 500 mL) from the contracted uterus after
delivery (Cheek and Gutsche, 2002).
MATERNAL AIRWAY AND RESPIRATORY
SYSTEMS
During pregnancy, the maternal airway has significantly
increased mucosal edema and tissue friability throughout
the pharynx, larynx, and trachea. These changes make
laryngoscopy and intubation more challenging. In addition, the presence of comorbidities such as preeclampsia,
upper respiratory tract infections, and the active pushing
and increased venous pressure during the second stage further exacerbate airway tissue edema (Munnur et al, 2005).
At term, minute ventilation is increased approximately
50%, oxygen consumption is increased by more than 20%,
and functional residual capacity is decreased by 20%. The
combination of these changes (increased oxygen consumption and decreased oxygen reserve) results in a state promoting rapid desaturation during periods of apnea. The
changes in both airway and respiratory physiology during
pregnancy make ventilation and intubation more difficult
and increase the potential for complications.
MATERNAL GASTROINTESTINAL SYSTEM
The gravid uterus displaces the stomach cephalad and
anterior and the pylorus cephalad and posterior. These
changes reposition the intraabdominal portion of the
esophagus into the thorax and result in decreased competence of the esophageal sphincter. Higher progesterone
and estrogen levels further reduce esophageal sphincter
tone. Gastric pressure is increased by the gravid uterus
and by the lithotomy position used during vaginal delivery. Maternal gastric reflux symptoms increase with the
gestational age of the pregnancy and ultimately affect the
majority of parturients (Marrero et al, 1992).
Beyond midgestation, women are at increased risk for
pulmonary aspiration of acidic gastric contents; this is
caused by decreased tone and competence of the lower
esophageal sphincter as well as delayed gastric emptying
with onset of labor or administration of opioids. This risk
has important implications for induction of general anesthesia and airway management by the anesthesiologist, and
it is discussed in detail under General Anesthesia.
UTERINE AND FETAL CIRCULATION
Uterine weight and blood flow increase throughout gestation from approximately 100 mL/min before pregnancy
to approximately 700 mL/min (10% of cardiac output)
at term gestation, with 50% to 80% of the uterine blood
flow perfusing the intervillous space (placenta) and 20% to
50% supporting the myometrium. Uterine vasculature has
limited autoregulation and remains (essentially) maximally
dilated under normal conditions during pregnancy.
CHAPTER 16 Obstetric Analgesia and Anesthesia
Maternal uterine blood flow decreases as a result of
either decreased uterine arterial perfusion pressure or
increased arterial resistance. Decreased perfusion pressure
can result from systemic hypotension secondary to hypovolemia, aortocaval compression, or significant decreases
in vascular resistance from the initiation of neuraxial anesthesia or induction of general anesthesia. Uterine perfusion pressure can also decrease from increased uterine
venous pressure associated with vena caval compression
(e.g., supine position), uterine contractions (particularly
uterine hypertonus by oxytocin hyperstimulation), or
significant increase in intra-abdominal pressure (pushing
during second stage or seizure activity). Stress-induced
endogenous catecholamines and exogenous vasopressors
can increase uterine artery resistance and decrease uterine blood flow. Despite these potential effects, phenylephrine (alpha-adrenergic) is useful for treating maternal
hypotension secondary to neuraxial anesthesia, and it has
been demonstrated to result in less fetal acidosis and base
deficit compared to treatment with ephedrine (primarily
beta-adrenergic) in many clinical trials (Lee et al, 2002a;
Ngan Kee et al, 2009; Smiley, 2009).
ANALGESIC OPTIONS FOR LABOR
AND VAGINAL DELIVERY
The pain of labor is highly variable and described by many
women as severe. Factors influencing the patient’s perception of labor pain include duration of labor, maternal pelvic
anatomy in relation to fetal size, use of oxytocin, parity, participation in childbirth preparation classes, fear and anxiety
about childbirth, attitudes about and experience of pain, and
coping mechanisms. Labor analgesia prevents autonomic
reflex effects that can be deleterious for certain high-risk
patients and their fetuses (e.g., patients with severe preeclampsia, valvular heart disease, myasthenia gravis). The
American College of Obstetricians and the American Society
of Anesthesiologists issued a joint statement indicating that
a maternal request for pain relief is sufficient justification for
administration of analgesics during labor (American College
of Obstetricians and Gynecologists, 2002). Although older
observational studies associate increased cesarean delivery
rates with early administration of epidural analgesia, more
recent randomized studies found no difference in the rate
of cesarean delivery or instrument-assisted vaginal delivery
between women in whom analgesia was initiated early in
labor versus later (Eltzschig et al, 2003; Wong et al, 2005).
Consequently the American Society of Anesthesiology
stated in its 2007 Practice Guidelines, “Neuraxial analgesia
should not be withheld on the basis of achieving an arbitrary
cervical dilation, and should be offered on an individualized
basis. Patients may be reassured that the use of neuraxial
analgesia does not increase the incidence of cesarean delivery” (American Society of Anesthesiologists Task Force on
Obstetric Anesthesia, 2007).
The choice of analgesic method resides primarily with
the patient. The medical condition of the parturient, stage
of labor, urgency of delivery, condition of the fetus, and
availability of qualified personnel are also factors. Many
different techniques are available to alleviate labor and
delivery pain. Analgesia refers to pain relief without loss
of consciousness; regional analgesia denotes partial sensory
161
BOX 16-1 Techniques for Labor Analgesia
Nonpharmacologic analgesia
Systemic medications
Opioid analgesics
Sedatives and anxiolytics
Dissociative analgesics
Inhalation analgesia
Regional analgesia
Epidural
Spinal
Combined spinal-epidural
Paracervical block
Pudendal block
blockade in a specific area of the body, with or without
partial motor blockade. Regional anesthesia is the loss of
sensation, motor function, and reflex activity in a limited
area of the body. General anesthesia results in the loss of
consciousness and the goals for providing general anesthesia typically include hypnosis, amnesia, analgesia, and
skeletal muscle relaxation.
Techniques for labor analgesia must be safe for mother
and fetus and individualized to satisfy the analgesic
requirement and desires of the parturient; they also must
accommodate the changing nature of labor pain and the
evolving, varied course of labor and delivery (e.g., spontaneous vaginal delivery, instrumentally assisted vaginal
delivery, and cesarean delivery). The current approaches
to pain relief are outlined in Box 16-1.
NONPHARMACOLOGIC ANALGESIA
There is a variety of nonpharmacologic techniques for
labor analgesia. These techniques include hypnosis, the
breathing techniques described by Lamaze, acupuncture,
acupressure, the LeBoyer technique (LeBoyer, 1975),
transcutaneous nerve stimulation, massage, hydrotherapy,
vertical positioning, presence of a support person, intradermal water injections, and biofeedback. A metaanalysis
reviewing the effectiveness of a support individual (e.g.,
doula, family member) noted that parturients with a support individual used fewer pharmacologic analgesia methods, had a decreased length of labor, and had a lower
incidence of operative deliveries (Hodnett et al, 2007). In a
2006 retrospective national survey of women’s childbearing experiences, although neuraxial methods of pain relief
were rated as the most helpful and effective, nonpharmacologic methods of tub immersion and massage were rated
more or equally helpful in relieving pain compared with
the use of opioids (Declercq et al, 2006). Although many
nonpharmacologic techniques seem to reduce labor pain
perception, most studies lack the rigorous scientific methodology for the useful comparison of these techniques to
pharmacologic methods.
SYSTEMIC MEDICATIONS
Systemic medications for labor and delivery are widely
used, but are administered with limitations on both dose
and timing because they readily cross the placenta and are
162
PART IV Labor and Delivery
associated with a risk of neonatal respiratory depression in
a dose-dependent fashion. Although the use of systemic
opioid analgesics is common (e.g., fentanyl, meperidine,
morphine, nalbuphine, butorphanol), the use of sedatives
and anxiolytics (e.g., barbiturates, phenothiazine derivatives and benzodiazepines), and dissociative agents (e.g.,
ketamine, scopolamine) is rare.
Opioid Analgesics
Opioids are the most frequently used systemic analgesic,
but for many patients opioids do not provide adequate
analgesia during labor and delivery (Bricker and Lavender, 2002; Olofsson et al, 1996). Opioids are inexpensive,
easy to administer, and do not require a trained anesthesia
provider. However, they have a high rate of maternal side
effects (sedation, respiratory depression, dysphoria, nausea,
pruritus), can decrease fetal heart rate variability and fetal
movements, and carry a potential risk of neonatal respiratory depression and changes in neurobehavior. Systemic
administration of opioids at doses that are safe for mother
and newborn provides some analgesia, but it is not a substitute for the analgesia provided by regional techniques.
Systemic opioids are recommended for administration in
the smallest doses possible with minimization of repeated
dosing to reduce the accumulation of drug and metabolites
in the fetus. Systemic opioids are most useful for patients
with minimal pain, precipitous labor, or contraindications
to neuraxial blockade, such as a coagulopathy.
The opioid analgesics act by binding to opiate receptors
located throughout the central nervous system. Although
this is the main site of action, opioid receptors have also
been identified in other peripheral tissues. Four main types
of receptor have been identified: mu (μ), kappa (κ), delta
(δ), and sigma (σ). The analgesic effects, side effects, and
pharmacodynamic characteristics depend on the receptor affinity of each individual opioid. These receptors are
also responsible for the associated respiratory depression,
sedation, and dysphoria and may affect thermoregulation.
Binding of opiate agonist agents (e.g., meperidine, morphine, and fentanyl) and subsequent receptor activation
alters neural transmission of pain by inhibiting the presynaptic and postsynaptic release of neurotransmitters. The
opioid antagonists (e.g., naloxone, naltrexone) are competitive antagonists and bind with high affinity to the μ receptor more so than the κ or δ opioid receptors. These drugs
do not produce analgesia and are capable of displacing
other agonist drugs from the receptor and reversing their
effects. A third class of agents interacts with the receptors
and results in both agonist and antagonist activity depending on the receptor type. These drugs, with high receptor
affinity, are capable of analgesic effect (e.g., pentazocine,
nalbuphine, and butorphanol).
Opioids differ in pharmacokinetics, pharmacodynamics,
method of elimination, and the presence of active metabolites, but all readily cross the placental barrier through passive diffusion. In order for systemic opioids to effectively
alleviate labor pain, larger doses would be necessary. This
dosing would risk excessive maternal sedation, maternal
respiratory depression, loss of protective airway reflexes,
newborn respiratory depression, and impairment of both
early breastfeeding and neurobehavior. Consequently,
large doses of opioids are avoided, and the doses used routinely for labor analgesia typically do not have significant
adverse effects on either the mother or neonate.
Meperidine remains the most widely used opioid worldwide. Maternal half-life of meperidine is 2 to 3 hours,
with the half-life in the fetus and newborn being significantly greater and more variable at values between 13 and
23 hours (Kuhnert et al, 1979). In addition, meperidine is
metabolized to an active metabolite (normeperidine) that
can significantly accumulate after repeated doses. With
increased dosing and shortened time interval between dose
and delivery, there is greater neonatal risk of decreased
Apgar scores, lowered oxygen saturation, prolonged time
to sustained respiration, abnormal neurobehavior, and
more difficulty initiating successful breastfeeding (Nissen
et al, 1997).
Morphine was used more frequently in the past, but
currently is rarely used. Like meperidine it has an active
metabolite (morphine-6-glucuronide) and a prolonged
duration of analgesia (3 to 4 hours). The half-life is longer
in neonates compared with adults, and it produces significant maternal sedation.
Fentanyl is a synthetic opioid with a short duration of
action (approximately 30 minutes), no active metabolites,
and a ratio of fetal to maternal plasma concentrations of
approximately 1:3. In small intravenous (IV) doses of 50
to 100 μg/hr there were no significant differences in Apgar
scores, respiratory depression, or neurobehavior scoring
compared with newborns of mothers who did not receive
fentanyl (Rayburn et al, 1989a). In addition, a comparison of equianalgesic doses of IV fentanyl compared with
IV meperidine (Rayburn et al, 1989b) demonstrated a
decreased frequency of maternal nausea, vomiting, and
prolonged sedation in the fentanyl group. In addition,
neonates whose mothers received meperidine required
naloxone more often compared with the fentanyl-exposed
infants. There was no difference in the neuroadaptive testing scores of the two groups of infants.
Opioids can be given by intramuscular (IM) or IV
administration. IM injection of opioids is technically easy
but leads to uneven analgesia, the possibility of late respiratory depression, and profound neonatal effects if not
properly timed (Shnider and Moya, 1960). IM administration is associated with a high incidence of neonatal
depression 2 to 4 hours after injection in some of the less
lipid soluble opioids. IV administration is the most widely
used technique to give opioids to a woman in labor, with
effects that are more predictable and doses more easily
timed. However, achievement of a steady blood level of
opiate sufficient to provide analgesia is difficult, with the
parturient frequently suffering underdosage (rarely overdosage). In an effort to improve pain relief and maternal
satisfaction, continuous IV infusion of short-acting opiates
(e.g., alfentanil, remifentanil) or self-administration of IV
opiates is increasingly used for systemic opioid delivery
during labor.
Patient-controlled analgesia (PCA) has the implied
advantage of allowing the patient to titrate her dose to
the minimum required for analgesia with the lowest blood
levels of opiates, resulting in considerably less placental
transfer and fewer side effects with increased patient satisfaction (McIntosh and Rayburn, 1991). Remifentanil, an
CHAPTER 16 Obstetric Analgesia and Anesthesia
ultra-short–acting opioid rapidly metabolized by nonspecific serum esterases, is significantly metabolized by the
fetus with umbilical artery–to–vein ratios of approximately
0.3 (Kan et al, 1998). It can be used effectively for labor
PCA, but it appears difficult to achieve satisfactory analgesia without significant potential of maternal respiratory
depression (Olufolabi et al, 2000; Thurlow and Waterhouse, 2000; Volikas, 2001). In a prospective randomized
controlled trial comparing the effectiveness of epidural
analgesia to a remifentanil PCA with optimized settings,
epidural analgesia was significantly more effective than
PCA in regard to labor pain, but no differences were noted
in neonatal outcome measures (Volmanen et al, 2008).
Although there are individual differences among opioids, all readily cross the placental barrier and exert neonatal effects in typical clinical doses, including decreased fetal
heart rate variability and dose-related neonatal respiratory
depression. All opioids can have maternal side effects,
including nausea, vomiting, pruritus, and decreased gastrointestinal motility and stomach emptying.
Sedatives and Anxiolytics
Sedatives and anxiolytics are administered infrequently
to pregnant patients because they increase risks of sedation and respiratory depression in both mother and
newborn, especially when used with opioids. Sedatives
and anxiolytics were used more frequently in the past to
diminish the adverse motivational-affective component of
labor pain. Examples of such drugs are barbiturates and
benzodiazepines.
Diazepam and midazolam are benzodiazepines used
as anxiolytic agents in obstetrics. They rapidly cross the
placenta, yielding approximately equal maternal and fetal
blood levels within minutes of IV administration (Cree
et al, 1973). In addition, the neonate has a limited ability
to excrete diazepam, so the drug and its active metabolite may persist in significant amounts in the neonate for
up to 1 week (Scher et al, 1972). Diazepam can result
in neonatal hypotonia, lethargy, and hypothermia when
used in large maternal doses (30 mg) (Cohen et al, 1993).
However, when it is used in small doses (2.5 to 10 mg
IV), minimal sedation and hypotonia have been observed
(McAllister, 1980). Midazolam has a shorter duration of
activity, but rapidly crosses the placenta and is associated with neonatal hypotonia in larger doses. The use
of benzodiazepines remains somewhat controversial, but
these agents can reduce maternal anxiety and are useful
for treating convulsions associated with local anesthetic
toxicity or eclampsia. However, all benzodiazepines are
amnestics, and therefore may not be appropriate in many
childbirth situations, depending on the desires of the
parturient.
Dissociative Analgesia
The IM or IV administration of low-dose ketamine
(0.25 mg/kg) produces a state called dissociative analgesia,
which is characterized by analgesia and unreliable amnesia without loss of consciousness or protective airway
reflexes (Galloon, 1976). This state is accompanied by a
dreaming phenomenon, which may be unpleasant but can
163
be minimized by coadministration of benzodiazepines to
improve amnesia. In divided doses totaling less than 1 mg/kg,
ketamine provides adequate analgesia for vaginal delivery
and episiotomy repair. Although these low doses are not
associated with neonatal depression, higher doses are associated with decreased Apgar scores (Akamatsu et al, 1974).
With larger doses, maternal airway protection cannot be
guaranteed, increasing the risk of gastric content aspiration. Ketamine is best reserved for use as a supplement (in
low doses) to other techniques or for situations in which
(1) more reliable and safer agents or techniques are contraindicated or ineffective or (2) when rapid control is required
because the mother’s pain is compromising the fetus (e.g.,
mother is moving uncontrollably, unable to effectively
push, and jeopardizing delivery while the fetal head is presenting during a prolonged fetal heart rate deceleration).
INHALATION ANALGESIA
The use of nitrous oxide is widespread in Canada, Australia,
the United Kingdom and other parts of the world, but its
use in the United States as a labor analgesic is uncommon.
It is usually administered as a 50% mixture with oxygen.
At a 50% concentration (without coadministration of opioids), nitrous oxide is insufficient to cause unconsciousness
or loss of protective airway reflexes. Appropriate equipment and trained personnel are essential to ensure safety
(i.e., limiting the nitrous oxide concentration, avoiding
administration of a hypoxic mixture, avoiding coadministration of other agents). Nitrous oxide is a weak analgesic
but can provide satisfactory pain relief for some parturients. It can be used during the first, second, or third stage
of labor either alone or as a supplement for a regional
block or local infiltration (Rosen, 1971). The use of 50%
nitrous oxide in a supervised fashion is safe and rapid acting, causes minimal maternal cardiovascular or respiratory depression, and does not affect uterine contractility.
The effects of nitrous oxide are rapidly reversed with discontinuation, and it does not cause neonatal depression
regardless of duration of administration (Rosen, 2002a).
Although the use of inhaled halogenated agents such as
sevoflurane in low-inspired concentrations was found to
be safe and effective at reducing labor pain in a small study
(Yeo et al, 2007), barriers to routine use include the need
for specialized vaporizers, scavenging systems, and the lack
of larger studies.
NEURAXIAL (REGIONAL) ANALGESIA
Neuraxial analgesia, including epidural, spinal, and combined spinal-epidural (CSE) techniques, has become the
most widely used method for labor analgesia in the United
States (Bucklin et al, 2005). Neuraxial techniques typically
involve epidural and spinal administration of local anesthetic agents, and often the co-administration of epidural
and spinal opioid analgesics. Other adjuvant agents, such
as clonidine and neostigmine, can decrease the dose of
local anesthetics or opioids required for effective analgesia;
however, they are not routinely used and do not appear to
offer a significant advantage when compared to local anesthetics with or without opioids (Eisenach, 2009; Parker
et al, 2007; Roelants, 2006).
164
PART IV Labor and Delivery
Neuraxial Local Anesthetics
Local anesthetic agents consist of amine moieties linked by
an intermediate chain containing an ester or amide. Local
anesthetics reversibly block nerve impulse conduction via
voltage-gated sodium channels. Their chemical structures
are secondary or tertiary amines, which are weak bases,
marketed as the hydrochloride salts to achieve aqueous
solubility.
The ester-linked local anesthetics (e.g., chloroprocaine,
procaine, tetracaine) are rapidly metabolized by plasma
cholinesterase, decreasing the risk of maternal toxicity and
placental drug transfer (O’Brien et al, 1979). Amide-linked
local anesthetics (e.g., lidocaine, bupivacaine, ropivacaine)
are degraded by P-450 enzymes in the liver. Local anesthetics differ in their onset, peak plasma concentration, potency
and duration based on their lipid solubility, protein binding,
site of injection, and concentration. Vascular absorption of
local anesthetics limits the safe dose that can be administered. Elevated plasma concentrations produce neurologic
toxicity (seizures) or cardiovascular toxicity (myocardial
depression, ventricular arrhythmia). Bupivacaine and
ropivacaine are the most commonly used local anesthetics for labor analgesia. Ropivacaine is a pure amide-linked
S-isomer, unlike bupivacaine, which is a racemic mixture.
Because the R-isomer of bupivacaine can bind strongly to
cardiac sodium channels, ropivacaine can decrease the possibility of severe cardiac toxicity associated with accidental
intravascular injection of large doses of bupivacaine. However, ropivacaine is less soluble than bupivacaine and may be
slightly less potent. Regardless of differences, bupivacaine
and ropivacaine are extremely safe when appropriately used
for epidural or intrathecal administration. An accidental,
large intravascular dose of any local anesthetic can result in
significant maternal morbidity or mortality.
As with all drugs, placental transfer is determined by
molecular size, lipid solubility, protein binding, and maternal
drug concentration. Local anesthetics are weak bases with
high lipid solubility and a low ionized fraction. However,
the lower pH of the fetus has the potential to increase the
fraction of ionized molecules, decrease lipid solubility, and
result in ion trapping. Therefore in an acidotic fetus, higher
concentrations of local anesthetic can accumulate (ion trapping). Increased concentrations of local anesthetics result
in decreased neonatal neuromuscular tone similar to that
seen with magnesium. If a direct intravascular or intrafetal
injection of local anesthetics occurs, significant toxicity and
depression can develop, signified by bradycardia, ventricular
arrhythmia, and severe cardiac depression with acidosis.
Neuraxial Opioids
Although intraspinal (intrathecal) opiates were demonstrated in 1979 to be capable of producing profound analgesia in humans (Behar et al, 1979; Wang et al, 1979), the
epidural injection of opioids as sole agents has proved to
be of limited use for effective labor analgesia. In one study,
high doses of epidural morphine (7.5 mg) provided satisfactory but not excellent analgesia for 6 hours in the first
stage of labor, whereas 2 to 5 mg produced barely satisfactory analgesia in fewer than half the patients (Hughes et al,
1984). Besides the inadequate analgesia and the long onset
time (approximately 1 hour), the side effect of pruritus was
significant. In contrast, when lipid-soluble opioids (e.g.,
sufentanil, fentanyl) are administered in the epidural space
as sole agents, analgesia is rapid and equivalent to that of
systemic administration (Camann et al, 1992), but remains
inferior to that of dilute concentrations of local anesthetics,
and less effective for somatic pain associated with the second
stage of labor. When lipid-soluble opioids are administered
as an adjunctive agent with local anesthetics in the epidural
space, they decrease the total local anesthetic dose required
and lower the minimum local anesthetic concentration
needed to achieve adequate labor analgesia (Buyse et al,
2007; Celleno and Capogna, 1988; Lyons et al, 1997). The
most common maternal side effect of conventional doses of
epidural fentanyl or sufentanil is pruritus. After maternal
epidural administration, the lipid-soluble, poorly ionized
opioid analgesics rapidly enter the fetal circulation. An epidural bolus injection of an opioid results in peak neonatal
depression at 30 to 60 minutes (similar to an IV bolus dose).
Although typical doses used for labor analgesia adversely
affect the neonate, the potential for respiratory depression is
a function of the amount and timing of drug administered.
Subarachnoid (i.e., spinal, intrathecal) injections of fentanyl, sufentanil, meperidine, and morphine as sole agents
are more promising for effective maternal labor analgesia.
Analgesic effects of spinal opioids are more potent than
epidural or systemic administration, but are of limited
duration (2 hours) and are less effective than dilute epidural solutions of local anesthetics for analgesia in the second stage (Honet et al, 1992; Leighton et al, 1989). Spinal
opioid administration is often performed as part of a CSE
technique (discussed in the following section), with fentanyl or sufentanil being the most commonly used agents.
The intrathecal opioid is often combined with a small dose
of local anesthetic (e.g., 2.5 mg bupivacaine), decreasing
the dose of opioid needed and incidence of pruritus (Wong
et al, 2000). Reports of fetal heart rate changes after intrathecal administration of fentanyl or sufentanil (Cohen et al,
1993) may be caused by rapid onset of analgesia and rapid
decrease in circulating catecholamines, with epinephrine
decreasing faster than norepinephrine, resulting in an
unopposed oxytocic effect on the uterus. This effect would
increase uterine tone and decrease uterine blood flow. This
mechanism is speculative, but it is suggested by observed
cases and case reports (Friedlander et al, 1997). Some prospective randomized studies have found no difference in
incidence of fetal bradycardia between epidural administration of local anesthetics and intrathecal opioids administered with the CSE technique (Fogel et al, 1999; Nageotte
et al, 1997). A systematic review of studies comparing intrathecal opioids to other methods of labor analgesia noted an
increase in fetal bradycardia (odds ratio [OR], 1.8; 95% CI,
1.0 to 3.1) and increased maternal pruritus (RR, 29.6; 95%
CI, 13.6 to 64.6), but the risk of cesarean section because
of FHR abnormalities was similar (Mardirosoff et al, 2002).
NEURAXIAL TECHNIQUES
FOR LABOR ANALGESIA
Neuraxial techniques represent the most effective form
of labor analgesia, and they achieve the highest rates of
maternal satisfaction (Declercq et al, 2006). The patient
CHAPTER 16 Obstetric Analgesia and Anesthesia
remains awake and alert without sedative side effects,
maternal catecholamine concentrations are reduced
(Shnider et al, 1983), hyperventilation is avoided (Levinson
et al, 1974), cooperation and capacity to participate actively
during labor are facilitated, and predictable analgesia can
be achieved, superior to the analgesia provided by all other
techniques. Before initiating any neuraxial blockade, anesthesiologists assess the patient’s gestational and health
history, perform a focused physical examination, discuss
the risks, benefits, and alternatives, and obtain consent.
In otherwise healthy parturients, routine laboratory tests
are not required (American Society of Anesthesiologists
Task Force on Obstetric Anesthesia, 2007). Resuscitation equipment and drugs must be immediately available
to manage serious complications secondary to initiation of
epidural or spinal blocks (discussed under Contraindications and Complications of Neuraxial Techniques). During initiation of the neuraxial blockade, mother and fetus
are closely monitored.
EPIDURAL ANALGESIA
Epidural labor analgesia is a catheter-based technique to
provide continuous analgesia during labor. The technique
involves insertion of a specialized needle and catheter
(Figure 16-2) between vertebral spinous processes in the
back, into the epidural space. Most commonly, the needle is inserted at a lumbar space between L1 and L4. The
needle traverses the skin and subcutaneous tissues, supraspinous ligament, interspinous ligament, and the ligamentum flavum, and it is advanced into the epidural space
(Figure 16-3). The tip of the needle does not penetrate the
dura, which forms the boundary between the intrathecal
or subarachnoid space and the epidural space. To locate
the epidural space, a tactile technique called loss of resistance is used. The tactile resistance noted with pressure
on the plunger of an air- or saline-filled syringe dramatically decreases as the tip of the needle is advanced through
the ligamentum flavum (dense resistance) into the epidural space (no resistance), which has an average depth of
approximately 5 cm from the skin. Once the needle is properly positioned, a catheter is inserted through the needle.
165
The catheter remains in the epidural space and the needle
is removed. The catheter is secured with adhesives and
used for intermittent or continuous injections. Once the
catheter is placed, analgesia is achieved by administration
of local anesthetics, opioids, or both (see Neuraxial Local
Anesthetics and Neuraxial Opioids, earlier) and maintained throughout the course of labor and delivery. The
catheter can also be used for operative anesthesia (cesarean
delivery) and postoperative analgesia, when necessary.
After an incremental local anesthetic bolus to initiate
reliable analgesia, local anesthetics are typically infused continuously with similar or lower blood drug levels compared
with repetitive, intermittent boluses of local anesthetics
(Hicks et al, 1988; Rosenblatt et al, 1983). Most importantly,
the possibility of disastrous complications is reduced with
continuous infusions, such as total spinal anesthesia or massive intravascular injections with cardiovascular collapse secondary to large bolus doses of local anesthetics (D’Athis
et al, 1988). If the catheter enters the subarachnoid space
instead, the level of sensory and motor blockade increases
slowly without the sudden onset of complete subarachnoid blockade that can occur with large bolus techniques
(Li et al, 1985). Patient-controlled epidural anesthesia is a
delivery technique allowing the patient to self-administer
small boluses of epidural analgesics with or without a background infusion. Studies comparing patient-controlled epidural anesthesia with continuous infusion technique have
found decreased local anesthetic requirements, less anesthesia provider intervention, equivalent or improved patient
satisfaction, equivalent or decreased motor blockade, and
no significant differences in effects on the fetus or neonate
(Halpern, 2005; van der Vyver et al, 2002).
The choices for local anesthetics for epidural infusion
include dilute solutions of bupivacaine, ropivacaine, lidocaine, or chloroprocaine (Lee et al, 2002b). The concentration and volume of the loading dose, which is administered
before initiating the continuous infusion, and the volume
and concentration of the infusion are highly variable. With
higher concentrations of local anesthetics, the density of
the motor blockade increases. With larger volumes, a
greater dermatomal spread of analgesia is achieved. Most
practitioners routinely use low concentrations of local
A1
A
B
A1
C
D
C1
FIGURE 16-2 Photograph of typical needles and catheters used for neuraxial analgesia and anesthetic techniques. A, Epidural needle
(18-gauge Tuohy) with magnification of tip shown at right (A1). B, Epidural needle (Tuohy) with catheter inserted through needle. C, Spinal
needle (24-gauge Whitacre) with magnification of tip shown at right (C1). D, Spinal needle inserted through epidural needle for use in combined
spinal-epidural technique.
166
PART IV Labor and Delivery
Spinal cord
L1
Filum
terminate
L2
L3
Needle in
subarachnoid
space
Dura
Ligamentum
flavum
Interspinous
ligament
L5
Needle in
epidural
space
S1
S2
SPINAL ANALGESIA
S3
Supraspinous
ligament
Sacrococcygeal
ligament
timing of neuraxial analgesia in labor on operative deliveries in nulliparous women (Marucci et al, 2007). Cesarean
delivery rates (OR, 1.00; 95% CI, 0.82 to 1.23) and instrumental vaginal delivery rates (OR, 1.00; 95% CI, 0.83 to
1.21) were similar in the early neuraxial analgesia and later
control groups. Neonates of patients with early neuraxial
analgesia had a higher umbilical artery pH and received
less naloxone than did patients when using other methods.
The timing and use of neuraxial labor analgesia have differing effects on the length of labor. Compared with opioids
for labor analgesia, the use of epidural analgesia minimally
lengthens the first stage in some women and shortens it in
others, but overall appears to lengthen the second stage by
approximately 15 minutes (Anim-Somuah et al, 2005; Ohel
et al, 2006; Sharma et al, 2004; Wong et al, 2005).
S4
S5
Needle in
caudal canal
FIGURE 16-3 Schematic diagram of lumbosacral anatomy showing
needle placement for subarachnoid, lumbar epidural, and caudal blocks.
(From Rosen MA, Hughes SC, Levinson G: Regional anesthesia for labor
and delivery. In Hughes SC, Levinson G, Rosen MA, editors: Shnider and
Levinson’s anesthesia for obstetrics, ed 4, Baltimore, 2002, Lippincott
Williams and Wilkins, p 125.)
anesthetics and many coadminister an opioid with both
the initial bolus and the infusion (e.g., 2 μg/mL fentanyl)
for its synergistic effect. Dilute solutions of local anesthetics (0.0625% to 0.125% bupivacaine or ropivacaine) minimize the motor blockade and preserve the perception of
pelvic pressure with descent of the fetus.
Effects on the Progress of Labor and Rate
of Operative Delivery
The use of epidural analgesia has been associated with
prolonged labor and increased rates of assisted delivery
and cesarean section. Retrospective studies are difficult
to interpret, because higher pain scores are predictive of
increased labor duration (Wuitchik et al, 1989), and higher
analgesic requirements are predictive of increased rates of
cesarean section (Alexander et al, 2001). A Cochrane review
of the effects of epidural analgesia, including CSE, compared with nonepidural analgesia or no analgesia during
labor (Anim-Somuah et al, 2005) concluded that, although
there was an increased risk of instrumental vaginal birth
(RR, 1.38; 95% CI, 1.24 to 1.53; 17 trials including 6162
women), there was no evidence of a significant difference
in the risk of caesarean delivery (RR, 1.07; 95% CI, 0.93
to 1.23; 20 trials including 6534 women). A recent metaanalysis summarized the results of studies addressing the
Spinal analgesia is typically administered near the time
of anticipated delivery. A small dose of a local anesthetic,
opioid, or both is injected into the subarachnoid space.
This dose of local anesthetic is far lower than that used for
spinal anesthesia for cesarean section, and it has minimal
effects on motor nerve function. Compared with epidural analgesia, it has the benefits of a more rapid onset, a
lower failure rate, and being technically easier and quicker
to perform. It has the significant disadvantage of a finite
effective duration (approximately 90 minutes), but can be
extremely useful for certain circumstances such as forcepsassisted delivery for a woman without epidural analgesia.
COMBINED SPINAL-EPIDURAL ANALGESIA
One variation of the lumbar epidural technique is a CSE
analgesic. After placement of the epidural needle, but
before insertion of the epidural catheter, a longer spinal
needle is passed through the indwelling epidural needle
(see Figure 16-2), puncturing the dura, and a small dose
of local anesthetic or opioid is administered. If opioid
alone is administered and the epidural catheter is placed
but no local anesthetic is given, some analgesia can be
achieved without motor blockade or sympathectomy. This
method can allow the patient to walk safely (i.e., a “walking
epidural”), but the analgesia is limited in efficacy and duration (approximately 2 hours in the first stage) and is rarely
effective for the second stage of labor.
If small doses of local anesthetics are administered
through the spinal needle, segmental analgesia results
more rapidly than with epidural administration of local
anesthetics. Specifically, the onset of analgesia in the sacral
root distribution is much more rapid with CSE compared
with epidural alone. The epidural placement of the catheter allows continuation of the segmental analgesia initiated by the spinal technique. In a metaanalysis of 19 trials
(2658 women) comparing CSE to epidural labor analgesia
(Simmons et al, 2007), CSE had a faster onset of effective analgesia (especially in spread to sacral roots), was
associated with more pruritus, and was associated with
a clinically insignificant lower umbilical arterial pH. No
differences were seen for maternal satisfaction, walking in
labor, type of delivery, maternal hypotension, postdural
puncture headache rate, or need for blood patch.
CHAPTER 16 Obstetric Analgesia and Anesthesia
CONTRAINDICATIONS AND COMPLICATIONS
OF NEURAXIAL TECHNIQUES
Certain conditions contraindicate neuraxial procedures;
these include patient refusal, infection at the needle insertion site, significant coagulopathy, hypovolemic shock,
increased intracranial pressure from mass lesion, and
inadequate provider expertise. Other conditions such as
systemic infection, neurologic disease, and mild coagulopathies should be evaluated on a case-by-case basis. Human
immunodeficiency virus infection is not a contraindication
to regional technique in the pregnant patient (Hughes
et al, 1995).
Infrequent but occasionally life-threatening complications can result from administration of regional anesthesia. The most serious complications are from accidental
IV or intrathecal injections of local anesthetics. A prospective study of 145,550 epidurals in the United Kingdom
noted unintended intravascular injection rates of 1 in
5000 and high spinal rates of 1 in 16,000 (Jenkins, 2005).
An unintended bolus of IV local anesthetic causes dosedependent consequences ranging from minor side effects
(e.g., tinnitus, perioral tingling, mild blood pressure, heart
rate changes) to major complications (e.g., seizures, loss
of consciousness, severe arrhythmias, cardiovascular collapse). The severity depends on the dose, type of local
anesthetic, and preexisting condition of the parturient.
Measures that minimize the likelihood of accidental intravascular injection include careful aspiration of the catheter
before injection, test dosing, and incremental administration of therapeutic doses. If a local anesthetic overdose
occurs, consider using a 20% IV lipid emulsion to bind
the drug and decrease toxicity. Successful resuscitation and
support of the mother will reestablish uterine blood flow;
this will provide adequate fetal oxygenation and allow time
for excretion of local anesthetic from the fetus. The neonate has an extremely limited ability to metabolize local
anesthetics and may have prolonged convulsions if emergent delivery is required (Morishima and Adamsons, 1967;
Ralston and Shnider, 1978).
A “high spinal” (or total spinal) can result from an
unrecognized epidural catheter placed subdural, migration of the catheter during its use, or an overdose of local
anesthetic in the epidural space (i.e., high epidural). Both
high spinals and high epidurals can result in severe maternal hypotension, bradycardia, loss of consciousness, and
blockade of the motor nerves to the respiratory muscles
(Yentis and Dob, 2001).
Treatment of complications resulting from both intravascular injection and high spinal is directed at restoring
maternal and fetal oxygenation, ventilation, and circulation. Intubation, vasopressors, fluids, and advanced cardiac
life support algorithms are often required. In any situation
of maternal cardiac arrest with unsuccessful resuscitation,
the fetus should be delivered by cesarean section if the
mother is not resuscitated within 4 minutes of the arrest.
This guideline for emergent operative delivery increases
the chances of survival for both the mother and neonate
(American Heart Association, 2005; Katz et al, 1986).
In addition, a variety of less severe complications and
side effects can occur with neuraxial blockade. The retrospective rates of inadequate epidural analgesia or
167
inadequate CSE analgesia requiring catheter replacement
were 7% and 3%, respectively, at a U.S. academic center
(Pan et al, 2004). The rate of accidental dural puncture
during epidural catheter placement is 1.5%, and approximately half of these will result in a severe headache (Choi
et al, 2003), which are typically managed with analgesics
or a blood patch if necessary. Hypotension (decrease in
systolic blood pressure greater than 20%) secondary to
sympathetic blockade is the most common complication of
neuraxial blockade for labor analgesia, with rates between
10% and 24% (Brizgys et al, 1987; Simmons et al, 2007).
Prophylactic measures include left uterine displacement
and hydration. Although standards for timing, amount,
and hydration fluid remain controversial, dehydration
should be avoided (Hofmeyr et al, 2004; Kinsella et al,
2000; Ko et al, 2007). Treatment of hypotension consists
of further uterine displacement, IV fluids, and vasopressor administration. Small boluses of either phenylephrine
or ephedrine can be used to treat hypotension. Although
ephedrine (primarily β-adrenergic) was historically used,
phenylephrine (primarily α-adrenergic) is associated with
less fetal acidosis (Lee et al, 2002a; Ngan Kee et al, 2009;
Smiley, 2009). If treated promptly, maternal hypotension
does not lead to fetal depression or neonatal morbidity.
A rise in core maternal body temperature is associated
with labor epidural analgesia and may be influenced by
several factors; these include duration, ambient temperature, administration of systemic opioids, and the presence
of shivering. During the first 5 hours of epidural analgesia,
there is no significant rise in body temperature (Mercier
and Benhamou, 1997). In a retrospective study, temperature
increases approximately 0.10° C per hour, and may reach
38° C in as many as 15% of parturients with a labor epidural compared with 1% without an epidural (Lieberman
et al, 1997). Although Lieberman et al suggest a significant
increase in the rate of newborn evaluation for sepsis (i.e.,
sepsis work-up [SWU]) with epidural analgesia, 67.3% of
the noted SWUs were ordered in infants born to mothers without fever. Kaul et al (2001) found no association
between epidural analgesia and SWU. However, patients
who received epidural analgesia and their neonates had an
increased body temperature at delivery, and approximately
6% of these women had temperatures equal to or exceeding
38° C. Although maternal temperature had no predictive
value for the SWU, Kaul et al (2001) found other risk factors
associated with the SWU, including low birthweight, gestational age, meconium, or respiratory distress requiring intubation at birth, hypothermia at birth, group-B β-hemolytic
streptococcus colonization, and maternal preeclampsia or
hypertension. Although the etiology of the maternal temperature rise remains uncertain, it need not affect the neonatal SWU. Epidural analgesia during labor does not increase
the incidence of neonatal sepsis. Although it is suggested
that the fever is associated with noninfectious inflammatory
activation or altered thermoregulation, the fever is not associated with a change in white blood cell count or with an
infectious process, and treatment is not necessary.
Other potential side effects from neuraxial blockade
include pruritus, nausea, shivering, urinary retention,
motor weakness, low back pain, and a prolonged block.
More serious complications of meningitis, epidural hematoma, and nerve or spinal cord injury are extremely rare.
168
PART IV Labor and Delivery
A retrospective Swedish study of severe neurologic complications from neuraxial blockade included 200,000 obstetric epidurals and 50,000 obstetric spinals. Rates of serious
neurologic events (i.e., neuraxial hematoma or abscess,
nerve or cord damage) were 1:29,000 for obstetric epidural and 1:25,000 for obstetric spinal anesthetic procedures
(Moen et al, 2004).
regional anesthesia is contraindicated (e.g., coagulopathy,
hemorrhage) or if a rapid onset is needed for emergent
deliveries (e.g., fetal bradycardia, uterine rupture). Benefits
of general anesthesia compared with regional anesthesia
include a secure airway, controlled ventilation, rapid and
dependable onset, and potential for less hemodynamic
instability.
PARACERVICAL BLOCK
EPIDURAL ANESTHESIA
A paracervical block is used to provide pain relief during
the first stage of labor in up to 3% of patients (Bucklin et al,
2005). The technique consists of submucosal administration of local anesthetics immediately lateral and posterior
to the uterocervical junction, which blocks transmission of
pain impulses at the paracervical ganglion. Analgesia is not
as profound as with epidural or spinal regional block, and
the duration of analgesia is short (45 to 60 minutes), but
the complications and side effects of epidural analgesia,
such as hypotension, hypoventilation, and motor blockade, are avoided. Complications from systemic absorption
or transfer of local anesthetic can occur. There is also the
possibility of direct fetal trauma or injection. Paracervical block is associated with a 15% rate of fetal bradycardia based on a metaanalysis of studies (Rosen, 2002b).
The mechanism of this phenomenon is unclear, and close
fetal monitoring is warranted. The bradycardia may occur
secondary to decreased uterine blood flow from the vasoconstrictor properties of local anesthetics, greater uterine
activity, or transfer of the local anesthetic across the placenta to the fetus, causing direct toxic effects on the fetal
heart. The bradycardia is usually limited to less than 15
minutes, and treatment is supportive, consisting of lateral
positioning and administration of oxygen.
Epidural anesthesia is an excellent choice for surgical anesthesia when an indwelling, functioning epidural catheter
had been placed for labor analgesia. Epidural anesthesia
provides the ability to titrate the desired level of anesthesia and extend the block time, if needed for a prolonged
procedure. Epidural anesthesia is also ideal for patients in
a nonemergent delivery who would not tolerate the abrupt
onset of a sympathectomy from spinal anesthesia, such
as some patients with cardiac disease. The volume and
concentration of local anesthetic agents used for surgical
anesthesia are larger than those used for labor analgesia.
However, the technique of catheter placement, test dosing, and potential complications are similar. A typical dose
regimen of epidural anesthesia for cesarean delivery could
include 2% lidocaine (20 mL). Typically the anesthesiologist attempts to provide a dense block from the T4 level to
the sacrum. This technique might not completely alleviate the visceral pain and pressure sensation associated with
peritoneal manipulation, and adjuvant drugs are occasionally necessary. IV ondansetron and metoclopramide are
frequently given to decrease nausea and vomiting associated with the operative delivery and hemodynamic effects
from the dense neuraxial blockade (Lussos et al, 1992; Pan
and Moore, 2001). Epidural morphine (3 to 5 mg) is typically given near the end of the procedure to decrease postoperative pain for up to 24 hours (Cohen et al, 1991).
PUDENDAL BLOCK
The obstetrician performs a pudendal block with a transvaginal technique, guiding a sheathed needle to the vaginal mucosa and sacrospinous ligament just medial and
posterior to the ischial spine. The technique primarily
blocks sensation of the lower vagina and perineum, and
it is typically placed just before vaginal delivery. Although
the technique provides analgesia for vaginal delivery or
uncomplicated instrumental delivery, the rate of failure is
high. In many centers, this technique is used when epidural or spinal techniques are unavailable. Complications in
addition to failure include systemic local anesthetic toxicity, ischiorectal or vaginal hematoma, and, rarely, fetal
injection of local anesthetic.
ANESTHESIA FOR CESAREAN
DELIVERY
In the United States, the vast majority of cesarean deliveries are performed with neuraxial anesthesia. It offers the
advantages of less anesthetic exposure to the neonate, has
the benefit of an awake mother at the delivery, allows for
placement of neuraxial opioids to decrease postoperative
pain, and avoids the risks of maternal aspiration and difficult airway associated with general anesthesia. However,
the use of general anesthesia is sometimes required if
SPINAL ANESTHESIA
For the patient without an epidural catheter, spinal anesthesia is the most common regional anesthetic technique
used for cesarean delivery. The block is technically easier
than epidural blockade, more rapid in onset, and more reliable in providing surgical anesthesia from the midthoracic
level to the sacrum (Riley et al, 1995). The risk of profound
hypotension is higher with spinal anesthesia than with epidural anesthesia, because the onset of the sympathectomy
is more rapid. However, this risk can be nearly eliminated
by avoidance of aortocaval compression, prehydration, and
appropriate use of vasopressors. Colloid is significantly
more effective than crystalloid (Ko et al, 2007). Historically, ephedrine was recommended as the vasopressor of
choice, but more recent data confirm that a phenylephrine infusion at the time of spinal placement is effective
at preventing hypotension and is associated with less fetal
acidosis compared to ephedrine (Ngan Kee et al, 2004,
2009). Data also suggest that spinal anesthesia can be used
safely for patients with preeclampsia (Hood and Curry,
1999; Wallace et al, 1995). A typical spinal anesthetic
could consist of bupivacaine (12.5 mg) with morphine
(200 μg) added to decrease postoperative pain. A large variety of other combinations of local anesthetics and opioids
CHAPTER 16 Obstetric Analgesia and Anesthesia
are used. A hyperbaric solution of local anesthetic is often
used to facilitate anatomic and gravitational control of the
block distribution. The medication will flow with the spinal curvature to a position near T4, and a head-down position can enhance the rostral spread of the block if needed.
The duration of a single shot spinal anesthetic is variable,
but normally provides adequate surgical anesthesia for
greater than 90 minutes. In selected circumstances, the use
of a CSE technique offers the advantage of a spinal anesthetic, with rapid onset of a dense block and the ability to
administer additional local anesthetic through the epidural catheter if the procedure lasts for an extended time.
A continuous spinal anesthetic technique with deliberate
subdural catheter placement is a rarely used alternative,
but is sometimes chosen in cases of accidental dural puncture during attempts to place an epidural catheter. This
technique allows the advantage of a titratable, reliable,
dense anesthetic, but carries the risks of high spinal if the
intrathecal catheter is mistaken for an epidural catheter, or
if the provider is unfamiliar with the technique. The rates
of rare complications of meningitis or neurologic impairment from local anesthetic toxicity with the use of a spinal
catheter may be somewhat higher than the other neuraxial
techniques, but they remain unknown. Some data suggest that leaving the spinal catheter in place for 24 hours
decreases the risk of postdural puncture headache (Ayad
et al, 2003; Cohen et al, 1994).
LOCAL ANESTHESIA
Although cesarean delivery can be performed with local
infiltration, it is accompanied with considerable discomfort
to the woman and risks the possibility of local anesthetic
overdose. Most obstetricians are not trained to perform
the technique. However, local infiltration is useful in rare
circumstances, such as acute fetal distress without an available anesthesia provider.
GENERAL ANESTHESIA
General anesthesia is used in obstetric practice for cesarean section typically when regional anesthesia is contraindicated or for emergencies, because of its rapid and
predictable action. The major risks for maternal morbidity
are pulmonary aspiration and failed intubation. Appropriate airway examination, preparation for unanticipated
events, and familiarity with techniques and the algorithm
for difficult intubation (American Society of Anesthesiologists Task Force on Management of the Difficult Airway,
2003) are critical for providing a safe general anesthetic.
After denitrogenation of the lungs (i.e., preoxygenation), general anesthesia is induced by rapid-sequence
administration of an IV induction agent, followed by a
rapidly acting muscle relaxant. The trachea is intubated
with a cuffed endotracheal tube, and a surgical incision is
made after confirmation of tracheal intubation and adequate ventilation. Anesthesia is maintained by administering a combination of inhaled nitrous oxide and a potent
inhaled halogenated agent (e.g., isoflurane), as well as
sedative-hypnotics, opioid analgesics, and additional muscle relaxants if needed. During typical general anesthesia
for cesarean delivery, opioids and benzodiazepines are
169
administered after the baby is delivered, to avoid placental transfer of these agents to the neonate. Before delivery
of the baby, the primary anesthetic for the incision and
delivery is the induction agent, because there is little time
for uptake and distribution of the inhaled agents into the
mother or fetus (Dwyer et al, 1995). If intubation attempts
fail, the operative delivery may proceed if the anesthesiologist communicates that it is possible to reliably ventilate
the mother’s lungs with either facemask or laryngeal mask
airway (American Society of Anesthesiologists Task Force
on Management of the Difficult Airway, 2003).
Induction Agents
Anesthesiologists use a variety of agents to rapidly induce
unconsciousness. Among the most common are thiopental, propofol, etomidate, and ketamine. Each agent represents a different biochemical class, and each has specific
advantages and cardiovascular effects.
Sodium thiopental is a highly lipid-soluble, proteinbound barbiturate that can cause decreased cardiac output
and hypotension and rapidly crosses the placenta. Historically, it was the most common induction agent for cesarean
section under general anesthesia. In a study of healthy volunteers undergoing uncomplicated cesarean section, the
mean umbilical artery (UA)–to–umbilical vein (UV) ratio
of thiopental concentrations was 0.87 (Morgan et al, 1981).
IV administration of an appropriate dose (4 to 6 mg/kg)
renders the patient unconscious within a circulation time
(30 seconds), peaks in the UV blood in 1 minute and in the
UA blood in 2 to 3 minutes, and has no significant clinical
effect on neonatal well-being. However, doses of 8 mg/kg
can result in neonatal depression (Finster et al, 1972), and
higher doses may require cardiorespiratory supportive
techniques until the neonate can excrete the drug. This
elimination may take up to 2 days (Fox et al, 1979). The
lack of neonatal effects is unclear, but may be caused by
first-pass metabolism by the neonatal liver, rapid redistribution into maternal vascular-rich tissue beds, additional
dilution by the fetal circulation, and higher fetal brain
water content.
Propofol is a diisopropylphenol that is available as a 1%
aqueous solution in an oil-in-water emulsion containing
soybean oil, glycerol, and egg lecithin. Like thiopental,
propofol is rapid in onset and can cause significant hypotension with a similar UA:UV ratio of 0.7 (Dailland et al,
1989). Unlike thiopental, propofol is preservative free and
must be drawn up only hours before use. Other differences
are that propofol decreases the incidence of nausea and
vomiting, and it is currently not a controlled substance.
Propofol has not been demonstrated to be superior to
thiopental in maternal or neonatal outcome. Propofol
administration has no significant effect on neonatal behavior scores with induction doses of 2.5 mg/kg, but larger
doses (9 mg/kg) are associated with newborn depression
(Gregory et al, 1990).
Etomidate contains a carboxylated imidazole ring that
provides water solubility in acidic solutions and lipid solubility at physiologic pH. Like thiopental, etomidate has a
rapid onset of action because of its high lipid solubility,
it rapidly crosses the placenta, and redistribution results
in a relatively short duration of action. Unlike thiopental
170
PART IV Labor and Delivery
and propofol, etomidate has minimal effects on the cardiovascular system, but it is painful on injection, can cause
involuntary muscle tremors, has higher rates of nausea and
vomiting, and can increase the risk of seizures in patients
with decreased thresholds. At typical induction doses
(0.3 mg/kg), etomidate administration can cause decreased
neonatal cortisol production (<6 hours), but the clinical
significance remains uncertain (Crozier et al, 1993).
Ketamine, a structural analogue to phencyclidine, is
more lipid soluble and less protein bound than thiopental. It is an analgesic, hypnotic, and amnestic with minimal
respiratory depressive effects. Ketamine is biotransformed
in the liver to active metabolites, such as norketamine. In
contrast to thiopental, sympathomimetic characteristics
of ketamine increase arterial pressure, heart rate, and cardiac output through central stimulation of the sympathetic
nervous system, making it an ideal choice for a patient in
hemodynamic compromise. Doses that are higher than
those appropriate for induction of general anesthesia
(1 mg/kg) can increase uterine tone, reducing uterine
arterial perfusion. No neonatal depression is noted with
typical induction doses (Little et al, 1972). In low doses
(0.25 mg/kg), ketamine has profound analgesic effects, unlike
barbiturates, but has been associated with undesirable psychomimetic side effects (e.g., illusions, bad dreams), which
can be lessened by coadministration of benzodiazepines.
Nitrous Oxide
Inhaled nitrous oxide is often used as part of maintenance
for general anesthesia, because of its minimal effects on
maternal hemodynamics and uterine tone. As a sole agent,
it is insufficient to provide an appropriate level of anesthesia for an operative procedure. It rapidly crosses the
placenta with increasing UV-to–maternal artery ratios of
0.37 in the first 2 to 9 minutes increasing to 0.61 at 9 to
14 minutes (Karasawa et al, 2003). The effects of nitrous
oxide on the neonate are not significant. Additional information about nitrous oxide is found in the previous section
under Inhalation Analgesia.
Inhaled Halogenated Anesthetics
Isoflurane, sevoflurane, desflurane, and halothane are all
halogenated hydrocarbons that differ in chemical composition, physical properties, biotransformation, potencies,
and rates of uptake and elimination. In clinical use, specialized vaporizers deliver these volatile liquid agents, so
that the inhaled concentrations can be carefully titrated by
anesthesiologists because of the relatively profound cardiovascular effects and potential for uterine muscle relaxation. These agents are important components of general
anesthesia for cesarean section, but readily cross the placenta. Without the use of these agents, the incidence of
maternal recall of intraoperative events is unacceptably
high (Schultetus et al, 1986; Tunstall, 1979).
Placental transfer of inhalation agents is rapid because
these are nonionized, highly lipid-soluble substances of
low molecular weight. The concentrations of these agents
in the fetus depend directly on the concentration and
duration of anesthetic in the mother. Clinicians often confuse the use of general anesthesia and the terms fetal distress
and depressed neonate. A depressed fetus will likely become
a depressed neonate, and general anesthesia may be used
because it is the most rapidly acting anesthetic to allow
cesarean delivery. For a healthy fetus, the use of general
anesthesia is not contraindicated. A Cochrane review of
16 studies comparing neuraxial blockade versus general
anesthesia in otherwise uncomplicated cesarean deliveries
found that “no significant difference was seen in terms of
neonatal Apgar scores of six or less and of four or less at
one and five minutes and need for neonatal resuscitation”
(Afolabi et al, 2006). The authors concluded that there was
no evidence to show that neuraxial anesthesia was superior to general anesthesia for neonatal outcome. Recent
experimental animal studies have demonstrated neuronal
apoptosis in the developing brain when a variety of agents
are administered to induce and maintain general anesthesia (Istaphanous and Loepke, 2009; Loepke and Soriano,
2008). Implications for the fetus and neonate from brief
anesthetic exposures are currently unknown because of a
lack of human studies and difficulties extrapolating animal
study methodology to humans.
The induction–delivery interval is not as important in
neonatal outcome as the uterine incision–delivery interval,
during which uterine blood flow may be compromised and
fetal asphyxia may occur. A long induction–delivery time
may result in a neonate who is lightly anesthetized but
not asphyxiated. If excessive concentrations of anesthetic
are given for inordinately long times, neonatal anesthesia,
evidenced by flaccidity, cardiorespiratory depression, and
decreased tone, can be anticipated (Moya, 1966). It cannot
be overemphasized that if the neonatal depression is caused
by transfer of anesthetic drugs, the infant is merely lightly
anesthetized and should respond easily to basic treatment
measures. Treatment should include and focus on effective
ventilation; cardiopulmonary resuscitation is rarely necessary. Ventilation will allow elimination of the inhalation
anesthetic by the infant’s lungs. Rapid improvement of the
infant should be expected. Otherwise, a search for other
causes of depression is imperative. For these reasons, it is
critical that clinicians experienced with neonatal ventilation are present at operative deliveries under general anesthesia in which the time from skin incision to delivery may
be longer (i.e., known percreta, large fibroids), or maternal
condition necessitates an atypical induction and maintenance of anesthesia (e.g., a patient with critical aortic stenosis undergoing an opioid-based induction). A discussion
of the operative and anesthetic plan by the neonatologist,
obstetrician, and anesthesiologist is crucial for optimizing
the outcome of neonates in these situations.
Neuromuscular Blocking Agents
Succinylcholine remains the skeletal muscle relaxant of
choice for obstetric anesthesia, because of its rapid onset
and short duration of action. In doses of 1.5 mg/kg,
appropriate intubating conditions are present within 45
seconds. Because it is highly ionized and poorly lipid soluble, only small amounts cross the placenta. Side effects
include increased maternal potassium levels, myalgias, and
succinylcholine is a known trigger agent for malignant
hyperthermia in susceptible individuals. This depolarizing neuromuscular blocking agent is normally hydrolyzed
CHAPTER 16 Obstetric Analgesia and Anesthesia
in maternal plasma by pseudocholinesterase and usually
does not interfere with fetal neuromuscular activity. If
the hydrolytic enzyme is present either in low concentrations (Shnider, 1965) or in a genetically determined atypical form (Baraka, 1975), prolonged maternal or neonatal
respiratory depression secondary to muscular paralysis can
occur.
Rocuronium is a rapid-acting, nondepolarizing neuromuscular blocker that is an acceptable alternative to succinylcholine. It provides adequate intubating conditions
in approximately 90 seconds at doses of 0.6 mg/kg and in
less than 60 seconds at doses of 1.2 mg/kg (Abouleish et al,
1994; Magorian et al, 1993). Unlike succinylcholine, it
has a much longer duration of action, decreasing maternal
safety in the event the anesthesiologist is unable to intubate or ventilate the patient. It has the benefit of not being
a triggering agent of malignant hyperthermia or elevating
serum potassium levels.
During the operation, nondepolarizing neuromuscular blocking agents can be titrated to improve operating
conditions. Under normal circumstances, the poorly lipidsoluble, highly ionized, nondepolarizing neuromuscular
blockers (i.e., rocuronium, vecuronium, cis-atracurium,
pancuronium) do not cross the placenta in amounts significant enough to cause neonatal muscle weakness (Kivalo
and Saaroski, 1972). This placental impermeability is only
relative, however, and neonatal neuromuscular blockade
can occur when large doses are given (Older and Harris,
1968).
The diagnosis of neonatal depression secondary to
neuromuscular blockade can be made on the basis of the
maternal history (e.g., prolonged administration of neuromuscular blockers, history of atypical pseudocholinesterase), the response of the mother to neuromuscular
blocking drugs, and the physical examination of the newborn. The paralyzed neonate has normal cardiovascular
function and good color, but lacks spontaneous ventilatory movements, has muscle flaccidity, and shows no
reflex responses. The anesthesiologist can place a nerve
stimulator on the neonate and demonstrate the classic signs of neuromuscular blockade (Ali and Savarese,
1976). Treatment consists of ventilatory support until
the neonate excretes the drug, up to 48 hours. Reversal of
nondepolarizing relaxants with cholinesterase inhibitors
171
may be attempted (e.g., neostigmine, 0.06 mg/kg), but
adequate ventilatory support is the mainstay of treatment.
Concomitant administration of an anticholinergic (e.g.,
atropine, glycopyrrolate) is normally necessary to prevent
severe bradycardia from muscarinic side effects of the
increased acetylcholine.
SUMMARY
This chapter serves as a general overview of the changes
in maternal physiology during pregnancy and briefly discusses options and techniques for both labor analgesia and
anesthesia for operative delivery. Its purpose is to allow
the pediatrician and neonatologist a better understanding
of the decisions and concerns of the anesthesiologist and
the implications of his or her interventions. To provide the
best care for mother and child, excellent communication is
required between the obstetrician, pediatrician, anesthesiologist, and nurse. Only by facilitating these lines of communication and obtaining input from each discipline can
patient care and safety become optimized.
SUGGESTED READINGS
Afolabi BB, Lesi FE, Merah NA: Regional versus general anaesthesia for caesarean
section, Cochrane Database Syst Rev (4):CD004350, 2006.
American Society of Anesthesiologists Task Force on Obstetric Anesthesia:
Practice guidelines for obstetric anesthesia: an updated report by the American
Society of Anesthesiologists Task Force on Obstetric Anesthesia, Anesthesiology
106:843-863, 2007.
Anim-Somuah M, Smyth R, Howell C: Epidural versus non-epidural or no analgesia in labour, Cochrane Database Syst Rev (4):CD000331, 2005.
Chestnut DH: Obstetric anesthesia: principles and practice, ed 4, Philadelphia, 2009,
Mosby.
Eltzschig HK, Lieberman ES, Camann WR: Regional anesthesia and analgesia for
labor and delivery, N Engl J Med 348:319-332, 2003.
Hughes SC, Levinson G, Rosen MA, et al: Shnider and Levinson’s anesthesia for
obstetrics, ed 4, Philadelphia, 2002, Lippincott Williams & Wilkins.
Marucci M, Cinnella G, Perchiazzi G, et al: Patient-requested neuraxial analgesia for labor: impact on rates of cesarean and instrumental vaginal delivery,
Anesthesiology 106:1035-1045, 2007.
Simmons SW, Cyna AM, Dennis AT, et al: Combined spinal-epidural versus
epidural analgesia in labour, Cochrane Database Syst Rev (3):CD003401, 2007.
Ngan Kee WD, Khaw KS, Tan PE, et al: Placental transfer and fetal metabolic
effects of phenylephrine and ephedrine during spinal anesthesia for cesarean
delivery, Anesthesiology 111:506-512, 2009.
Wong CA, Scavone BM, Peaceman AM, et al: The risk of cesarean delivery with
neuraxial analgesia given early versus late in labor, N Engl J Med 352:655-665,
2005.
Complete references used in this text can be found online at www.expertconsult.com
P A R T
V
Genetics
C H A P T E R
17
Impact of the Human Genome Project
on Neonatal Care
Jeffrey C. Murray
HISTORY OF THE HUMAN GENOME
PROJECT
Within 50 years of the discovery of the structure of
DNA by Watson and Crick, the Human Genome Project achieved a major milestone with its description of an
almost complete sequence of the approximately 3 billion
nucleotides (A,C,G,T) contained within the human haploid genome. This effort was the result of an early vision
by a group of scientists that with advances in technology
and sequence analysis this task could be achieved (Watson and Cook-Deegan, 1991). The central goal was to
provide a reference sequence of the human genome that
could serve as a framework on which to better understand
disease pathogenesis. Eventually, it was hoped, this would
lead to improved treatments and prevention.
There have been numerous spin-offs from the technology and analytic platforms developed. A wide range of
other organisms have now had their genomes sequenced
from pathogenic viruses to complex plants and animals.
These sequences provide insights into disease pathogenesis that are infectious or immunologic and open new doors
to therapies. Comparing DNA sequences across species to
look for evolutionary conservation has also provided tremendous insights into the protein structure of genes and
the regulatory elements that control the cell type, timing,
and amount for the synthesis of any individual protein.
The nature of how a gene is defined has been substantially
altered by these insights, so that the gene is now recognized as a far more complex structure with elements dispersed sometimes 1 million nucleotides or more from the
protein coding components classically thought of as “the
gene” (Gerstein et al, 2007).
Understanding the genetic relationships between individuals of different ancestral origins has led to the current
effort to describe genetic variation across the human species as an essential aspect of understanding predisposition
to or resistance from disease. Because the identification
that carriers of sickle cell trait had increased resistance to
malaria, or the identification of genetic factors conferring
persistence of lactose tolerance into adult life as a mechanism associated with the advantages of dairy farming (Bersaglieri et al, 2004), it has been evident that individual
genetic variation is an important contributor to health and
disease. The hemoglobin system has provided an evolving
paradigm for our understanding of the molecular nature
of genetic disease beginning in the 1940s (Neel, 1949;
Pauling et al, 1949) and onward to serving as a model for
gene therapy in the modern era. The conception of the
genome project led to wide ranging concerns about ethical, legal, and social aspects and resulted in the development of the Ethical, Legal and Social Issues (ELSI) Project
to investigate these areas and to anticipate future concerns
and problems. Finally, as part of the social and legal component, the work has led to legislation in the United States
and abroad that is designed to protect individuals from discrimination based on their genetic background.
The Human Genome Project has been an international
effort from its beginnings and had critical predecessors in
the human gene mapping (HGM) meetings that had an initial focus on identifying the chromosomal location of normal and disease causing genetic variants. The community
established by the HGM meetings provided an infrastructure that enabled the more comprehensive sequence-based
maps developed in the wake of the HGM meetings. One
early outcome of these meetings and the recognized need
to convert research findings to clinical utility was creation
of Mendelian Inheritance in Man by Victor McKusicks,
which has now evolved into Online Mendelian Inheritance in
Man (www.ncbi.nlm.nih.gov/omim/)—a comprehensive
catalogue of single-gene disorders that is an essential reference tool for learning about the genetic aspects of both
rare and common disease.
The international effort continues with many individual
countries now focusing on genome efforts that are specifically relevant to their own high-risk medical conditions
(e.g., malaria, HIV, hemoglobinopathies). These early collaborative international efforts also created the framework
for one of the greatest successes of the Human Genome
Project—that is, information generated relevant to the
human DNA sequence and its variation should be held
in public trust and there should be open access to DNA
sequence and its annotation. This spirit of open access has,
in turn, led to a stronger community ethic for the sharing
of scientific and medical data that may be a legacy that will
exceed the value of the sequence itself.
There have been many technical and analytic advances
that have enabled the sequence, and more recently its
variation, to be understood and applied. Currently these
advances extend to the characterization of hundreds of
thousands of genetic variants on large populations of individuals. In the last few years this characterization has led to
dramatic advances in the understanding of how common
173
174
PART V
Genetics
genetic variation can contribute to human genetic disease. This chapter will provide some detail on the various
aspects of the Human Genome Project that are of current
relevance.
The interval of more than 50 years between the elucidation of DNA as the information-containing macromolecule and its structural description by Watson and Crick
has also spanned the embedding of medical genetics as an
important specialty in health care. This same time interval
has seen the identification, treatment, and in some cases
prevention of a wide range of disorders that have a strong
genetic component (including Rh incompatibility and
phenylketonuria) and a variety of other neonatal biochemical disorders and the hemoglobinopathies. Discussed in
Chapters 3 and 27 are the details of the current state of
our knowledge of these important advances in prenatal and
neonatal screening. It now seems apparent that the past
focus on adding one disease at a time to the armamentarium
of those that can be studied and treated using genetic tools
will soon give way to a comprehensive analysis available at
the level of the individual genome. Much of the current
effort in human genome work is devoted toward individualized medicine, in which physicians will have the ability to
treat not just a disease but a specific person with a disease
and to account for their genetic and environmental variation in response to therapy or risks associated with prevention. Motulsky (1957) recognized this pharmacogenetic
(or pharmacogenomic when applied globally) approach in
the early days of human genetic study as being essential
to understanding the significant variation in response to
therapeutic agents or toxins. Pharmacogenetics is already
being applied clinically to identify patients at risk for toxic
reactions to some chemotherapeutic agents as well as antiinfectious compounds. Individualization of diet and nutritional approaches is also on the immediate horizon, and
it seems likely that over the next few years there will be
in place high-throughput genotyping platforms that will
provide comprehensive information to the pediatrician
and neonatologist for providing direct alterations and care
beginning in early infancy. These individualized genomewide profiles are already available to early adopters and
are creating new models for how physicians will anticipate
health care needs and outcomes for their patients.
The history of the Human Genome Project, both its
scientific and social basis, has been published widely in
review articles and books. Its central conceit arose in the
middle 1980s, when it became apparent that technology
was advancing so that an undertaking could at least be considered (Watson and Cook-Deegan, 1991). By the early
1990s there were formal projects devoted to its implementation in academic settings (e.g., the U.S. Department of
Energy, the National Institutes of Health [NIH] and the
Medical Research Council in the United Kingdom) as well
as several commercially based entities that had an interest
in gene identification as a mechanism to developing therapeutic products. Besides the primary emphasis on DNA
sequencing, preliminary steps that entailed the development of physical and genetic maps also proved critical to
assembling the DNA sequence of humans into its linear
order on each of the individual chromosomes. These steps
were also necessary for the conversion of DNA sequence
data into useful tools for gene identification and mutation
discovery. The combination of academic and commercial
interests led to a competition that in the end provided the
stimulus to develop large-scale, high-throughput methodologies and the analysis tools that in 2001 enabled the
project to reach one of its primary goals ahead of some of
even the most aggressive predictions for when a complete
human sequence might be in place (Lander et al, 2001;
Venter et al, 2001). These successes and their resulting
technologies and algorithmic advances were based on trials
in less complex organisms (e.g., Escherichia coli, yeast, fruit
fly) and were then quickly applied to model and experimental organisms and to identify the sequences of plants
and animals of commercial and nutritional importance.
At the time of this writing, there are 998 microorganisms
(www.ncbi.nlm.nih.gov/genomes/lproks.cgi), as well as many
higher organisms, whose sequences have been completed.
SCIENCE AND THE HUMAN GENOME
In the years since the description of the primary human
sequence, a deeper understanding of the nature of the DNA
sequence and its regulation has developed. Phenomena that
are still relatively new in 2011 (e.g., copy number variants,
micro-RNA, epigenetic regulation) are important aspects
of how the human genome functions and interacts. There
is doubtless much more to learn about our DNA sequence,
but this chapter will review some current understandings.
There are enormous numbers of highly conserved DNA
sequences outside of traditional DNA-encoded protein
sequences for which function is completely unknown. The
next decades will be, in part, devoted to developing and
understanding how these conserved sequences play a role
in human health and disease. A few current features will
be discussed, with many more evolving almost daily in the
primary literature. The effects of these approaches have
been reviewed recently for neonatology, and this chapter
will expand on those themes (Cotten et al, 2006).
SINGLE NUCLEOTIDE POLYMORPHISMS
Single nucleotide polymorphisms (SNPs) are the workhorses of human genetic variation and indeed are only
a more specific term and characterization of restriction
fragment length polymorphisms (RFLPs), which were
the original DNA variants studied in the human genome.
The advent of the polymerase chain reaction eliminated
the need for RFLP studies. SNPs are specific nucleotide
sites in the human genome, where it is possible to have
one of two different nucleotides, or polymorphisms, at that
position on one of the DNA strands. For example, there
might be either a T or a G at a specific site. These variant sites are common with up to 1% of the human DNA
sequence being potentially variable between any two individuals, resulting in tens of millions of SNPs across the
genome. Most variation is found across all human populations, although some variants appear to be highly population specific. These variants are usually normal, in the sense
that they do not appear to be disease causing, although they
may lie adjacent to DNA changes that do contribute to disease predisposition. Chip-based DNA sequence detection
allows the assay of up to 1 million SNPs simultaneously on
one individual at a cost of a few hundred dollars.
CHAPTER 17 Impact of the Human Genome Project on Neonatal Care
COPY NUMBER VARIANTS
A relatively recent discovery in human genetic variation has
been the importance of copy number variants as contributors to human inherited disorders. Although both small
and large deletion and duplication events of the human
DNA sequence have been known since the 1970s, based on
cytogenetic banding, only recently has the role that these
play in disease been recognized widely. Early copy number variants ranged in length from one to five nucleotides
and proved to be useful in characterizing common genetic
variation used in family linkage studies. In parallel with
these were variants of which the central element might be
15 to 50 nucleotides in length—so-called mini satellites,
which were also used in family-based linkage studies. The
new class of variation is thousands to occasionally hundreds of thousands (or millions) of base pairs in length.
These variations may contain one or multiple genes that
can exist in two, three, or more copies arrayed in tandem
at particular chromosomal positions. When these tandem
arrays of largely identical sequences align themselves during meiosis, there is occasionally a misalignment that can
result in the deletion or duplication of one or more of the
copies. This event in turn can create a range of the number
of copies present from zero to many (Zhang et al, 2009).
When functional genes are contained within the copied
element or functional regulatory elements, the amount of
gene product made may be increased or decreased from
a reference level. Early examples in which copy number
variants contributed to disease include the Di George syndrome 22q- deletion and deletions associated with spinal
muscular atrophy and Charcot-Marie-Tooth disease. New
microdeletion syndromes are now being characterized with
great precision using array-based DNA analysis that is rapidly replacing the traditional karyotype as the first line of
chromosomal analysis. The recognizable syndromes that
are found recurrently, and with an identifiable phenotype
such as 22q-, are complemented by rare deletion or duplication events that result in congenital anomalies, developmental delay, or both, and where their etiologic nature
can be inferred from the normal structure of the parental
chromosomes. Finally, in areas such as autism, it is clear
that these microdeletion duplication events are a major
explanation for the sometimes sporadic, as well as familiar
nature, of these disorders. Their contribution to human
genetic disease is now suggested to be in the vicinity of
10% of all variant-contributed disease, and thus they form
an important class for investigation. Despite their clear
importance, there remains to be resolved many technical
and clinical issues related to their identification and meaning (Aki-Khan et al, 2009).
MITOCHONDRIAL DNA
Mitochondria are the energy-producing organelles present in thousands of copies within each cell. Each mitochondrion has its own genome, distinct from the nuclear
genome and thought to arise from incorporation of bacterial DNA by a eukaryotic cell. Although the mitochondrial
genome is approximately 16,500 bp in length (compared
with the 3 billion bp of the nuclear genome), there is a
wide range of disorders associated with variation in
175
mitochondrial sequence. In addition, because mitochondria reside in the cytoplasm and are not found in sperm,
they have a unique pattern of maternal-only inheritance,
in which mothers pass their mitochondria to all of their
offspring with their daughters in turn passing that on to
subsequent generations and with no passing of mitochondrial DNA from males to their children.
GENE IDENTIFICATION
One of the primary benefits of the reference human DNA
sequence is our ability to move quickly from finding the
location of a gene on a chromosome to identifying the
specifics of that gene and the disease-causing mutations.
Gene discovery can provide immediate clinical benefit in
the form of more accurate diagnoses and risk predictions
and longer-term benefits when gene discovery leads to an
understanding of gene function and physiology that can
be converted into treatment and prevention. Gene mapping approaches to human gene identification have been
in use since the late 1970s, and the first successes were the
identification of single-gene disorders, sometimes termed
monogenic or Mendelian because their inheritance patterns
follow the traditional modes of autosomal dominant, autosomal recessive, and sex linked. Currently there are three
primary methodologies under use for gene identification
(Altshuler et al, 2008). The first involves linkage studies
using large families with genetic disorders or many small
families with the same disorder than can be studied and
their data pooled. These linkage-based approaches can
provide a relatively well-defined chromosomal location for
single-gene conditions and has led to successful gene finding for cystic fibrosis, neurofibromatosis, and hundreds
of additional, mostly rare, conditions. This approach can
also be applied to common but genetically complex traits
for which there are no simple inheritance pattens. This
technique may require many hundreds of small families,
and the resultant gene localization is imprecise. A second
method for gene localization can make use of small chromosome rearrangements, such as balanced translocations
or small deletions or duplications, that result in a phenotype for a known disorder. In these cases, the location of
the chromosomal rearrangement immediately suggests
that a gene at or near that rearrangement is etiologic and
can be used to again directly search for evidence of a specific gene and mutation in that region. A third approach
now in great favor is described next in some detail, and it is
exerting a major influence on disease gene finding.
HAPMAP AND GENOME-WIDE ASSOCIATION
As noted previously, it is particularly challenging to find
genes associated with complex traits that have multiple
genetic and environmental contributors. This heterogeneity creates substantial difficulties in both finding and
confirming that any particular gene plays a role in the
disorder of interest. A new approach, enabled by advances
in technology (i.e., DNA chips) and statistical analysis is
proving to be remarkably successful. This approach takes
the form of the genome-wide association (GWA) study
in which a panel of genetic variants are densely arrayed
on DNA chips and characterized in large case and control
176
PART V
Genetics
populations (Manolio and Collins, 2009). GWA can also
be applied successfully to case-parent approaches, as will
be described in more detail.
The prerequisite to this comprehensive search for variation was the development of the human HapMap (http://
hapmap.ncbi.nlm.nih.gov/). The HapMap project developed a comprehensive understanding of the relationship
of SNPs across multiple human ancestral groups including
Europe, Africa, and Asia (International HapMap Consortium, 2007). HapMap is an essential component of a GWA
study because it provides a comprehensive reference listing of the relationships of SNPs and copy number variants
to enable their careful characterization in case and control
populations. By looking for evidence of DNA sequence
variation or allelic variation in which one allele is found
significantly overrepresented in a case compared with a
control population, there is a strong suggestion that the
etiologic gene and variant lies in the vicinity of the surrogate or marker gene allele. In the last 2 years this approach
has greatly extended the gene discovery process. Because
these common complex traits have a far greater population
impact, they are the ones that hold the greatest promise
for providing information about the common contributors
to pediatric disease. As of this writing approximately 100
associations have been identified (Manolio et al, 2009),
but to date the emphasis has largely been on adult complex disorders (e.g., type 2 diabetes, cardiovascular disease,
mental health disorders such as schizophrenia), with only
a handful applied to pediatric traits. Autism (Wang et al,
2009), asthma (Himes et al, 2009), and cleft lip or palate (Birnbaum et al, 2009) have had new loci identified
with the hope that this will provide insights into their
pathogenesis. One challenge of the GWA study is that at
present, whereas it can identify one or more loci associated with the disease, implementation of a GWA requires
expensive technology and large (usually numbering in the
thousands), well-phenotyped case and control populations.
Thousands of cases and controls must be available to have
sufficient power to detect the small effects seen. A second caveat is that the loci found usually have low relative
risks or odds ratios, so that the clinical effect of any one
identified locus is very small (Hardy and Singleton, 2009).
However, collections of loci can have a combined substantial impact, and even a low odds ratio may identify a new
biologic pathway that could provide great insights into
etiology and treatment (Hirschhorn, 2009). As analytic
approaches improve and as costs drop, it seems likely that
the GWA study will become a common approach to a wide
range of neonatal disorders in which either genetic risks or
pharmacogenetic variation important in drug choice and
dosing will be unraveled.
Besides detecting disease-associated genetic risk factors,
these studies also contribute to knowledge of the genetics of normal trait variation, such as height and skin color.
Substantial advances in this area have also occurred with
more than 50 genes found to have a role in factors such
as height determination (Hirschhorn and Lettre, 2009).
However, although identical twin studies tell us that height
is almost entirely genetically determined across a broad
range of environmental variation, the genetic findings to
date explain only a small amount of the contributors to
height. There is still much to learn in regard to normal
trait variation and the ability to make predictions about a
child’s future physical traits or cognitive behavioral range.
New approaches beyond GWA are still critically needed to
find this large amount of unexplained genetic contributors
(Manolio et al, 2009).
The final, and in some ways ultimate, strategy for
identifying gene variation associated with disease will
be to perform comprehensive genome-wide sequencing.
In the last few years, completely new technologies have
been applied to DNA sequencing, and the costs of DNA
sequencing of a specific individual have dropped by four or
five factors of 10 from the original cost of assembling the
reference human genome. The reference genome was an
amalgamation of many individuals and cost approximately
$3 billion, or $1 per nucleotide of sequence. The goal
is to eventually reduce this cost by $1000 per individual
genome sequenced, which would place it well within the
realm of current medical investigations, such as imaging,
and well below the cost of many therapeutic approaches.
At the time of this writing, several individuals have had
their individual genome sequence reported (Wheeler et al,
2008). There is a newly initiated project under way to
sequence the genome from approximately 1000 individuals around the world to provide a catalog of DNA
sequence variation that can be used as a reference point
for comparisons in the future for normal and disease trait
variation studies.
The high-throughput sequencing projects have an
initial focus on the sequencing of all exons or conserved
elements in the genome that are known to be at higher
risk for containing disease-causing mutations. Nonetheless, mutations can also reside in the intergenic regions
and outside of regions of conservation. Eventually wholegenome sequencing will be required for a comprehensive
view of contributors to human inherited disorders. This
high-throughput sequencing approach, when performed
in sufficient numbers of individuals, can identify disorders
in which there are common variants contributing to the
disease process as well as individually rare variants that also
compose a portion of the disease risk panel. Cystic fibrosis provides an example of this model in that of the single
mutation (the ΔF508) consisting of a three-nucleotide
deletion, which explains approximately 70% of the allelic
variants that result in cystic fibrosis. The remaining 30%,
however, are divided across an additional 20 to 50 that are
relatively common, and then more than 1000 others that
may be rare or family specific. Therefore to identify all the
mutations associated with cystic fibrosis risk, one would
need to perform comprehensive DNA sequencing.
COMPARATIVE GENOMIC HYBRIDIZATION
Comparative genomic hybridization (CGH), or arraybased hybridization, is another technical advance that is
now providing, in many cases, a replacement for traditional
karyotype-based chromosomal analysis. CGH has already
had a substantial influence on prenatal and newborn testing for genetic disease and is discussed in more detail in
Chapter 20. Although these array approaches cannot yet
detect every form of chromosomal abnormality (e.g., balanced translocations are not detected), they are highly successful in detecting major structural whole chromosomal
CHAPTER 17 Impact of the Human Genome Project on Neonatal Care
aneuploidy such as Turner syndrome (45X) or trisomy 21,
and they are also effective in identifying small deletions
and duplications with a high degree of resolution. These
approaches are increasingly becoming a first-line screen
for chromosomal structural abnormalities, with the potential for replacing standard karyotypes almost entirely. The
arrays also benefit from being able to use DNA directly,
so that live cells are not required as they have been for traditional karyotypes. Thus material obtained from even a
deceased infant or fetus can be used in the analysis. Because
the amount of DNA required is also small, minimal DNA
quantity further facilitates its technical application. There
are a variety of competing approaches available for such
CGH. The current generation of array-based tools use
fragments of DNA that are far smaller than prior versions; therefore the resolution is in the range of thousands of nucleotides. This resolution provides detection
of very small deletions or duplication events of potential
etiologic importance and begins to approach the level of
direct sequence comparison. At some point, direct genome
sequencing may replace these tiled-array approaches for
detection of deletions and duplications.
In parallel with this improvement in resolution has been
the recognition that there is no comprehensive catalog of
the normal range of variation for rare deletion and duplication events. When new deletions or duplications are
identified for which there is not a strong prior track record
for their clinical importance (as would be the case for 22qfor example), then the interpretation as to whether they
are causal for disease can be challenging. This challenge
can sometimes be aided by examining parental samples in
which the presence of a de novo deletion or duplication
event may be more strongly indicative of contributing to
a disease etiology than when the event is also identified
in one or more other family members. However, caution
needs to be exercised when such deletion-duplications
are found in other family members, because there can be
a range of penetrance for such abnormalities as well as
other co-contributors that might be necessary for the full
disease phenotype to be expressed (van Bon et al, 2009).
Therefore caution must be exercised in family counseling,
and it is essential to have the most accurate and up-to-date
information on the nature and role of such variants when
using them in a clinical setting. Fortunately these catalogs
of normal variation are becoming available (Shaikh et al,
2009). With these caveats, it is still clear that there is an
enormous amount to learn about how these structural
variants can contribute to disease of the neonate.
PHARMACOGENOMICS
Pharmacogenomics, although originally described in the
1950s as a result of genetic variation associated with commonly recognized enzymes such as G6PD (Motulsky,
1957), is now a highly active area for the clinical application of genome studies. The U.S. Food and Drug Administration has already identified more than a dozen genes for
which characterization of allelic variation of those genes
may be an important component of therapeutic decision
making, including several that have a pediatric application.
Perhaps one of the best understood is the role of thiopurine
methyltransferase and its genetic variation associated with
177
major adverse events associated with chemotherapeutic
agents such as 6-mercaptopurine (Weinshiboum, 2006). It
is now standard care to study pediatric patients preparing
to begin chemotherapy for their risks associated with these
allelic variants that affect thiopurine methyltransferase
drug metabolism. These current pharmacogenetic variants are the beginnings of a much larger group of variants
that will tie individual response to therapeutics and will
eventually result in direct individualized medicine on an
individual basis. In neonatology, many potentially toxic
medications are used routinely; these include indomethacin or ibuprofen for treatment of patent ductus arteriosus, antibacterial and antifungal agents that have potential
serious adverse consequences such as gentamicin, medications to treat pulmonary or systemic hypertension, antiarrhythmic agents, and others for which individual patient
response may be based in part on enzyme allelic variation
in those individuals.
By identifying individual risk beforehand, one can either
adjust medication doses or choose alternative medications
to minimize complications and maximize therapy. In some
cases it may be that larger doses of medication will be
required based on pharmacogenetic variation as well. It is
in the area of this pharmacogenetic variation that perhaps
the greatest advances will come in the application of the
genome project to neonatal care in the next decade.
With the availability of large clinical trials, such as
those overseen by the Neonatal Network and others,
improved understanding of the role of genetic variation in
drug response will almost certainly be obtained. Because
the cost of studies of genetic variation are modest, and
the consequences of using the wrong drug or the wrong
dose potentially devastating, it will be critical for neonatologists and pediatricians to understand the evidence for
which variants can contribute in a proven way to improved
therapeutic approaches. Just as measuring drug levels has
become a routine part of care in many settings, so will testing beforehand for genetics risk identification and dosage
plans also become a part of neonatal care.
INDIVIDUALIZED GENETIC INFORMATION
One of the most dramatic recent advances has been the
application of the SNP-based association technologies
to commercial entities that are now providing direct
consumer testing to interested parties. Currently there
are multiple commercial organizations that, for a fee
of between $100 and $1000, will extract DNA from a
saliva sample and perform high throughout SNP analysis. The results are then provided to the individual with
the whole array of GWA risks that have been published
in peer-review journals. Although currently controversial (Ng et al, 2009), it seems likely that the use of these
technologies will expand in the private or “recreational”
setting, directly in the clinical care setting, and eventually into newborn screening. A challenge for clinicians
is presented when families have obtained this information but have little context for its meaning, and anecdotal
examples of its application to neonates in terms of risk
are already arising. This example is yet another example
of why clinicians need to remain current with the rapidly
advancing genomic capabilities.
178
PART V
Genetics
MICROBIOMES AND THE METAGENOME
As described previously, there have been approximately
1000 bacterial and viral genomes sequenced, as well as
more complicated pathogenic eukaryotic species. These
microbial and viral sequences can be used to establish the
genetic framework of the many organisms that are found
in or on an individual. Because the newborn infant is
largely thought to be sterile at birth, but rapidly acquires
its microbial flora through environmental exposures, an
understanding of the relationships of these many microbial
species and how they participate in maintaining health or
contributing to disease will be critical to an understanding of neonatal health and disease. Humans have tens of
thousands of microbial and viral species contained in or
on their bodies in locations such as the gut, mouth, and
skin (Turnbaugh et al, 2007). A few obvious disease states,
such as necrotizing enterocolitis (Neu et al, 2008) or presumed sepsis, are already under active study for the role in
which microbial communities might have a role in predisposition or disease prevention.
It would seem likely that probiotic therapy at sometime in the future might be targeted to provide a milieu
that would optimize the microbial flora or the neonatal
gut to prevent or limit disease and to assist in nutrition.
Our understanding of the transmittal of microbiomes to
the infant from maternal and environmental sources is
still limited (Palmer et al, 2007), but the capacity of the
Human Genome Project and its DNA sequencing capabilities allows identification of the organisms involved
with a high degree of precision. It is already apparent that
there are many organisms present in and on newborns
that have been previously unknown to science, because
the identification of species has been limited by the ability
to characterize them initially through culture or antigen
characterization. For organisms for which appropriate culture media have not been identified, their existence was
not known until the development of the ability to perform
direct genome sequencing of material collected from the
stool, urine, or elsewhere (DiGiulio et al, 2008).
By bypassing the culture step and sequencing biologic
samples with DNA sequenced directly, it is possible to
establish the identification of new organisms that may
be contributing directly to disease in ways that were not
previously identifiable. This concept of the microbiome
also suggests that in the future—whereas the core DNA
sequence will need to be sequenced only once because it
is relatively unchanging, except in the case of rare somatic
events—the metagenome sequence is constantly evolving based on alterations in factors such as nutrition and
antibiotic exposure. In the pediatric population, it may be
that DNA sequences sampled from the metagenome will
be performed repeatedly as a way of assaying responses to
therapy or risks to disease.
EPIGENETICS
Epigenetic regulation or the modification of DNA
sequences through environmental exposures, such as
altered nutrition or drug exposure, is also an evolving part of the Human Genome Project’s contributions
to our understanding of how genes function and work
(Devaskar and Raychaudhuri, 2007). Many of these epigenetic changes are thought to arise in utero. As a result, a
new field of inquiry into the developmental origins of adult
disease (Joss-Moore and Lane, 2009; Simmons, 2009) is
now expanding to help better understand the consequences
of how DNA can be modified and gene function altered,
whereas the linear DNA sequence remains the same.
Alterations of DNA can include methylation of individual
nucleotides or acetylation or other modifications that take
place in the histones that help to establish the chromosomal and chromatin-based structure of DNA sequences.
The developmental origins of disease hypothesis posits
that in utero changes can be induced by altered exposure
states such as malnutrition, and these exposure states can
imprint on the DNA a modified form that is then transmitted cell to cell and established in the infant, with continuing effects on gene regulation into childhood and
adulthood. For example, if this imprinting results in a
more effective storage capacity for calories, then in adult
life when calories are more readily accessible, adults may
be predisposed to obesity and its consequences. Similarly,
such changes might also regulate cell division and transfer and play a role in childhood leukemias. Because these
effects may occur in utero and in the premature infant, it
seems likely that in the future a better understanding of
our nutritional and drug exposure to infants will be important, which could have direct consequences on the predisposition of neonates to later disease. Although there are
some technologies in place to develop an understanding of
epigenetic modifications and changes, their application in
a clinical setting is still relatively modest.
GENE THERAPY
Although not a direct consequence of the Human Genome
Project, the idea of gene therapy has developed in parallel with our understanding of the human genome. Early
efforts to use direct gene replacement are continuing, and
although many challenges still remain it seems likely that
these forms of therapy will continue to expand and eventually be commonly applied in the pediatric setting. Enzyme
replacement therapy for several biochemical disorders is
now widely available and, for those disorders for which
earlier treatment results in better outcomes, recognition
of such disorders early in life becomes critical. In other
settings direct gene replacement has begun to suggest it
may be both safe and efficacious (Maguire et al, 2009),
opening an important door to future therapies. In parallel with projects that examine gene replacement, there is
also the development of a better understanding of how
related phenomena such as micro RNAs or small interfering RNAs also function. These small interfering RNAs,
for example, may prove to be highly effective therapeutic
agents, and a wide range of pediatric disorders are now
under study to determine whether these can be used in a
therapeutic or preventive sense. The genome sequence is
likely to contribute to an understanding of undiscovered
elements that may have an important role in gene regulation that could be effective in therapeutic settings as well,
and these advances are eagerly anticipated.
Finally, advances in stem cell research and its therapeutic potential have now been enabled by new legislation
CHAPTER 17 Impact of the Human Genome Project on Neonatal Care
that is affording the NIH an opportunity to make stem cell
lines available to investigators for studies using NIH funding (Collins, 2009). More readily available stem cells will
likely increase the pace of research into the use of these
potentially powerful therapeutic agents.
ETHICAL ASPECTS OF HUMAN
GENETICS
The ethical, legal, and social aspects of the Human Genome
Project have been of at least equal importance to the technology developments that have provided the advances in
basic science and translational science described elsewhere
in this chapter. It was Watson’s insight early on in the process of establishing the Human Genome Project to allot
3% to 5% of the NIH Genome Institute budget directly
for ELSI Project activities. The goal was to anticipate the
controversies that might develop and to address them proactively whenever possible. In addition, the ELSI Project
has also been involved in a wide range of concerns about
genetic discrimination. ELSI Project investigators and
their colleagues worked hard to develop model legislation
that could eliminate genetic discrimination in the job and
for health insurance purposes.
The successful passage of the Genetic Information Nondiscrimination Act (Tan, 2009) in the summer of 2008 was
a major milestone at the federal level in providing protection for all citizens from workplace and health insurance
discrimination. While many states had already addressed
these issues, this federal assurance provides a greater level
of national protection that is likely to facilitate the ease with
which individuals enroll in both clinical genetics studies
and research-oriented studies. The second critical aspect
of the ELSI Project is addressing the need to have common forms and formats for enrolling research subjects in
what are now the highly complex and large genetic investigations required to perform GWA studies described elsewhere in this chapter. It was recognized early that obtaining
informed consent for participation in a genetics study was a
constantly moving target, because technologies often outstripped the ability of social scientists to be prepared for
the scale and possibilities of what might come next. These
issues have been dramatically highlighted in recent years
during which the scale of genetic characterization is being
performed on the SNP ChIP assays, and the recognition
that even aggregate data might be able to result in individual identification (Homer et al, 2008) has dramatically
altered the clinical and research landscape for genetics.
GWA studies in particular have been scrutinized in great
depth, and the NIH has detailed guidelines in place for
179
how consent documents should be worded and the information that must be provided to research subjects. Similar oversight is provided for external users of the GWA
data, which, to the benefit of the research community,
have been deposited in readily accessible databases after
appropriate scientific and ethical review. Another area of
influence of the genome project has been on forensic DNA
analysis. Although such a discussion is beyond the scope
of this chapter, the ability to identify family relationships
or ancestral origins has now made DNA analysis a widely
used legal investigative and as a commercial tool. It has
had spectacular successes in freeing convicts whose DNA
evidence has exonerated them from being a participant in
major crimes. DNA analysis is also applied widely to assess
paternity and to help suggest the ancestral origin of an
individual when it might be unknown, as with adoptees or
descendants of slaves.
SUMMARY
The advances in the genome project have now begun
to be evident at the bedside. Individualized or personalized medicine already has applications in pharmacogenetic variation, and risk profiles can also be generated for
common diseases. These risk profiles may one day assist
in anticipatory guidance for common disorders such as
asthma, diabetes, or preterm birth. The microbiome will
also represent an opportunity to better identify disease etiology and to improve therapeutic specificity. As long as
protections against discrimination continue to be aggressively implemented, detailed genetic information will
greatly improve patient care.
SUGGESTED READINGS
Altshuler D, Daly MJ, Lander ES: Genetic mapping in human disease, Science
322:881-888, 2008.
Christensen K, Murray JC: What genome-wide association studies can do for
medicine, N Engl J Med 356:1094-1097, 2007.
Devaskar SU, Raychaudhuri S: Epigenetics: a science of heritable biological c
adaptation, Pediatr Res 61:1R-4R, 2007.
Hardy J, Singleton A: Genome-wide association studies and human disease, N Engl
J Med 360:1759-1768, 2009.
Hirschhorn JN: Genome-wide association studies: illuminating biologic pathways,
N Engl J Med 360:1699-1701, 2009.
Joss-Moore LA, Lane RH: The developmental origins of adult disease, Curr Opin
Pediatr 21:230-234, 2009.
Manolio TA, Collins FS, Cox NJ, et al: Finding the missing heritability of complex
diseases, Nature 461:747-753, 2009.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
18
Prenatal Genetic Diagnosis
Diana W. Bianchi
As a result of the expanding number of prenatal diagnostic tests that are performed on pregnant women, clinicians know a lot about their patients long before they
even touch them. To date, prenatal genetic diagnosis has
focused broadly on detection of fetal structural abnormalities. Whereas in the past the major indication for prenatal
diagnosis was advanced maternal age or a family history
of a single-gene disorder, currently most pregnant women
receive an invasive prenatal diagnosis because of abnormal screening results. This chapter discusses the common
methods of prenatal genetic diagnosis, the information
they convey, and the implications for the newborn.
NONINVASIVE TECHNIQUES
MATERNAL SERUM SCREENING
Maternal serum screening is part of routine obstetric
care worldwide. It is used to identify a high-risk pregnancy in a low-risk population of pregnant women. First-
trimester maternal serum screening consists of measuring
pregnancy-associated plasma protein A and the free beta
subunit of human chorionic gonadotropin (hCG). Secondtrimester maternal serum screening consists of measurement of alpha-fetoprotein (AFP), hCG, unconjugated
estriol (uE3), and inhibin A—proteins that are made by the
fetus or placenta.
Biochemical Screening for Neural
Tube Defects
AFP is one of the major proteins in fetal serum. Its precise
physiologic role is unknown. It can be detected as early
as 4 weeks’ gestation when it is synthesized by the yolk
sac (Bergstrand, 1986). Subsequently, it is produced in the
fetal liver and peaks in the fetal serum between 10 and 13
weeks’ gestation. AFP is then excreted into the fetal urine
or leaks into the amniotic fluid through the skin before
keratinization at 20 weeks’ gestation. It is also present in
cerebrospinal fluid. AFP in maternal serum is exclusively
fetal in origin (Crandall, 1981). Maternal serum AFP
peaks at 32 weeks’ gestation because of greater placental permeability for the protein (Ferguson-Smith, 1983).
For interpretation of results, accurate gestational dating
and knowledge of maternal race, weight, and presence or
absence of diabetes are critical.
Brock and Sutcliffe (1972) observed that there were
markedly increased levels of AFP in the amniotic fluid of
fetuses with anencephaly and open neural tube defects.
Subsequently, it was shown that elevated amniotic fluid
AFP levels were associated with increased maternal serum
AFP (Ferguson-Smith, 1983). The possibility of a screening test for open neural tube defects became apparent.
In the initial collaborative efforts aimed at studying maternal serum AFP, results were expressed as multiples of
180
the median (MoM) to allow comparisons between laboratories. It has become a convention to describe results
greater than 2.5 MoM as abnormally high and less than
0.6 MoM as abnormally low. Both findings require further
investigation.
If the AFP is elevated, the patient is offered an ultrasonographic examination to verify gestational age, determine fetal viability, and diagnose many of the structural
abnormalities that can be associated with an elevated AFP.
Although the AFP test was developed to screen for neural
tube defects, abnormally high results are not specific for
this condition (Box 18-1). If the ultrasonographic examination is unrevealing, the patient undergoes amniocentesis to assay the amniotic fluid for the presence of AFP
and acetylcholinesterase, which are elevated in open spina
bifida (Crandall et al, 1983). Although an elevated AFP is
compatible with a normal diagnosis, a study of 277 infants
with elevations of maternal serum AFP and normal levels
of amniotic fluid AFP revealed a higher incidence of intrauterine growth restriction and non–neural tube anomalies
(Burton and Dillard, 1986).
Aneuploidy Screening
Maternal serum AFP screening has also been used to detect
chromosomally abnormal fetuses since the observation was
made that a low AFP value was more likely in a fetus with
trisomy 18 or 21 than in a normal fetus (Merkatz et al,
1984). Several prospective studies have demonstrated that
expressing risk for Down syndrome as a combined function of maternal age and AFP value, and offering amniocentesis to all women with a risk of 1 in 270 or greater
(the equivalent risk in a 35-year-old woman based on
age alone), makes it possible to detect approximately one
third of otherwise unexpected cases of Down syndrome
in fetuses (Dimaio et al, 1987; Palomaki and Haddow,
1987). Low AFP values are probably caused by decreased
hepatic production in the affected fetus. Although it would
make sense to ascribe this phenomenon to the small size
of the liver, one study found no association between fetal
weight and low AFP values in chromosomally abnormal
fetuses (Librach et al, 1988). The differential diagnosis
of a decreased AFP level is shown in Box 18-2. Because
a low AFP value detects only one third of fetuses with
Down syndrome, a normal AFP value does not rule out
trisomy 21.
Experience with using low maternal AFP levels as a
screen for fetal chromosome abnormalities has led to the
evaluation of many other proteins produced by both the
fetus and placenta. Three of these—uE3, hCG, and inhibin
A—were incorporated into second-trimester maternal
serum screening panels (Wenstrom et al, 1999). Measurements of all four can be combined to improve the sensitivity and specificity of Down syndrome detection. AFP, uE3,
and hCG are only weakly correlated with one another, and
CHAPTER 18 Prenatal Genetic Diagnosis
BOX 18-1 D
ifferential Diagnosis of
Abnormally High Maternal Serum
Alpha-Fetoprotein Levels
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Incorrect gestational dating
Multiple pregnancies
Threatened pregnancy loss
Fetomaternal hemorrhage
Anencephaly
Open spina bifida
Anterior abdominal wall defects
Congenital nephrosis
Acardia
Lesions of the placenta and umbilical cord
Turner syndrome
Cystic hygroma
Renal agenesis
Polycystic kidney disease
Epidermolysis bullosa
Hereditary persistence (autosomal dominant trait)
BOX 18-2 D
ifferential Diagnosis of
Abnormally Low Maternal Serum
Alpha-Fetoprotein Levels
ll
ll
ll
ll
Incorrect gestational dating
Trisomy 18
Trisomy 21
Intrauterine growth restriction
their values are all independent of maternal age (Norton,
1994). Elevations of hCG are the most specific markers
for fetal trisomy 21 (Bogart et al, 1987; Rose and Mennuti,
1993), whereas estriol levels are approximately 25% less
than normal. Pregnancies affected by fetal trisomy 18 also
have reduced levels of uE3 and hCG. Each measurement
is compared with population-specific normal values (i.e.,
MoM), which are then converted to a likelihood ratio
that is expressed as a numeric risk for Down syndrome.
Women whose serum screening results indicate a fetal
Down syndrome risk of greater than 1 in 270 are offered
amniocentesis. Approximately 5% of all serum screen values are calculated to be false-positive results to achieve a
sensitivity of detection of at least 70% for cases of Down
syndrome.
In 1996 the National Health Technology Assessment
Program in the United Kingdom launched a study comparing the performance of first-trimester to second-trimester
screening—the Serum, Urine, and Ultrasound Screening
Study (SURUSS) (Wald et al, 2003). In addition to
the well-validated second-trimester markers, the first-
trimester serum markers (pregnancy-associated plasma
protein A and free beta hCG) and an ultrasound marker,
nuchal translucency (NT), were measured. The results
suggested that combining all measurements in an integrated panel improved the sensitivity of detection of
trisomy 21 to 93% at a fixed false-positive rate of 5%;
this was followed by the largest obstetric clinical trial
ever performed in the United Sates and funded by the
National Institutes of Health—the First and Second
Trimester Evaluation of Risk (FASTER) study (Malone
181
et al, 2005a). In this prospective study of 38,189 pregnant
women, 117 had a fetus affected with Down syndrome.
All study subjects were required to have a first-trimester
NT measurement, as well as first- and second-trimester
serum screening. The results of the study showed that
first-trimester combined serum and sonographic screening was better than conventional second-trimester screening, and that the highest sensitivity was achieved by the
integrated test that combined results from both trimesters.
The excellent results achieved by both of these large-scale
studies resulted in the American College of Obstetricians
and Gynecologists recommendation that all pregnant
women be offered screening for Down syndrome as part
of their routine prenatal care (American College of Obstetricians and Gynecologists, 2007).
ULTRASONOGRAPHIC EXAMINATION
OF THE FETUS
Ultrasonographic examination of the fetus is the best noninvasive method for gestational dating, definition of fetal
anatomy, serial measurements of fetal growth, and evaluation of dynamic parameters such as cardiac contractility,
fetal urine production, and fetal movement. In addition, it
has been suggested that antenatal visualization of the fetus
promotes maternal-infant bonding (Fletcher and Evans,
1983). The advent of antenatal ultrasonography has had
a significant influence on the types of patients who are
admitted to the neonatal intensive care unit.
First Trimester
With the advent of transvaginal scanning and improved
imaging, coupled with advances in first-trimester biochemical screening, fetal ultrasonography during the first
trimester has become routine. A major emphasis of firsttrimester sonography is screening for aneuploidy, using
the following markers: nuchal translucency measurement,
absent nasal bone, cystic hygroma, and abnormal ductus
venosus blood flow.
NUCHAL TRANSLUCENCY MEASUREMENT
Ultrasonographic visualization and quantification of the
normal subcutaneous fluid-filled space at the back of the
fetal neck is known as the nuchal translucency measurement (Said and Malone, 2008). This particular fluid-filled
space is especially well seen in the first-trimester fetus
(10 to 14 weeks’ gestation; Figure 18-1). In normal fetuses,
the maximal thickness of the subcutaneous translucency
between the skin and soft tissue overlying the fetal spine
increases as a function of crown-rump length. Normal
standards have been established for each gestational week.
An enlarged NT measurement is thought to be caused
by abnormal or delayed development of the lymphatic
system, fetal cardiovascular abnormalities, or defects
in the intracellular matrix (Said and Malone, 2008). An
increased NT thickness is associated with a higher risk
of fetal trisomy 21 (Snijders et al, 1998), cardiac defects
in chromosomally normal fetuses (Hyett et al, 1996), and
anomalies such as diaphragmatic hernia, omphalocele,
skeletal defects, Smith-Lemli-Opitz syndrome, and spinal
182
PART V
Genetics
chromosomally normal fetuses (Cicero et al, 2001). The
prevalence of absence of the nasal bone is affected by ethnicity (most common in individuals of African ancestry),
gestational age, and NT thickness. Using standardized
imaging techniques, the nasal bone is absent in 70% of
fetuses with trisomy 21, 55% of fetuses with trisomy 18,
and 34% of fetuses with trisomy 13 (Cicero et al, 2004).
ABNORMAL DUCTUS VENOSUS BLOOD FLOW
FIGURE 18-1 (See also Color Plate 3.) Sagittal ultrasonographic
image of a first-trimester fetus. The nuchal translucency measurement is the distance between the two crosses. A measurement larger
than normal standards for gestational age indicates that the fetus is at
high risk for Down syndrome, congenital heart disease, or both.
(Courtesy Dr. Fergal Malone.)
muscular atrophy (Sonek, 2007). In the largest prospective
study (96,127 pregnant women) to examine the association of NT and aneuploidy, Snijders et al (1998) detected
82.2% of the cases of trisomy 21 at a false-positive rate of
8.3%. The implementation of large-scale, first-trimester
sonographic screening protocols into clinical practice has
emphasized the need for quality assessment, training, and
standardized measurements. Assessment of the risk of fetal
chromosomal abnormalities in the first trimester allows
pregnant women the option of earlier (invasive) diagnostic
testing. The disadvantage of this approach is that earlier
diagnosis of aneuploidy may preferentially identify fetuses
already destined for miscarriage.
CYSTIC HYGROMA
Cystic hygroma manifests sonographically in the first trimester as an enlarged echolucent space that extends along
the entire fetal body (Said and Malone, 2008). What distinguished it from the NT is the presence of characteristic septations. Cystic hygroma in the first trimester is the
single most powerful sonographic marker that is associated
with aneuploidy (Malone et al, 2005b). In the FASTER
trial there were 134 cases of cystic hygroma among 38,167
study subjects (incidence of 1 in 285). In the 134 cases,
67 were associated with chromosome abnormalities,
(51%), 22 were associated with major structural anomalies
(primarily skeletal and cardiac) (34%), and five were associated with fetal death (8%) (Malone et al, 2005b). Therefore pediatricians and neonatologists need to have a high
level of suspicion for anomalies in live born infants with an
in utero history of cystic hygroma.
ABSENT NASAL BONE
In the original description of his eponymous syndrome,
Dr. Langdon Down noted that affected individuals had
a small nose (Down, 1995). In one study, absence of the
nasal bone in the fetal facial profile was noted in 43 of 59
(73%) fetuses with trisomy 21 and in 3 of 603 (0.5%) of
The use of first-trimester Doppler assessment has identified abnormal blood flow velocity patterns across the ductus venosus (Said and Malone, 2008). Blood flows in the
ductus during ventricular systole (the S wave) and diastole
(the D wave) have characteristic forms with high velocity
(Sonek, 2007). This velocity decreases during atrial contraction (the A wave), but in normal fetuses forward blood
flow is maintained. When there is complete cessation or
reversal of forward flow, the A wave is considered to be
abnormal. Abnormal ductal flow is associated with chromosomal abnormalities, cardiac defects, and adverse pregnancy outcomes (Sonek, 2007).
Second Trimester
Within the context of prenatal genetic diagnosis, ultrasonography can be used to detect congenital anomalies.
A malformation is present in 2% to 3% of live births (Nelson and Holmes, 1989). This risk is doubled in twins. Fetal
structures that are normally filled with fluid are especially
well visualized by ultrasonography. In approximately 10%
of infants with anomalies, the central nervous system is
involved (Hill et al, 1983). Ultrasonography is particularly useful in the diagnosis of anencephaly, microcephaly,
encephalocele, and hydrocephalus. By 20 weeks’ gestation,
the fetal facial structures can be examined with this method
for cyclopia, cleft lip, and micrognathia. Nuchal thickening is suggestive of Down syndrome, familial pterygium
coli, and other chromosome abnormalities (Benacerraf
et al, 1987; Chervenak et al, 1983). Fetal cardiovascular
structures may be reliably examined at 20 weeks’ gestation.
The presence or absence of four cardiac chambers, the
dynamic relationships between the cardiac valves and the
locations of the vessels allow diagnoses such as hypoplastic
left heart, double-outlet right ventricle, tricuspid atresia,
tetralogy of Fallot, and Ebstein anomaly. Pericardial effusion and arrhythmias may be similarly observed.
Gastrointestinal anomalies occur in approximately
0.6% of live births, and one third of them are associated
with chromosome abnormalities (Barss et al, 1985). The
decrease in fetal swallowing seen in some cases of bowel
obstruction (from atresia, stenosis, annular pancreas, or
diaphragmatic hernia) may lead to polyhydramnios that
results in a uterine size greater than expected for gestational dates. Although gastroschisis and omphalocele are
readily diagnosed on ultrasonography, they may be confused with each other, and their differing prognoses may
cause considerable parental anxiety (Griffiths and Gough,
1985). Gastroschisis usually occurs as an isolated anomaly;
infants generally do well after surgical repair. The kidneys
are identifiable by 14 weeks’ gestation, but the presence
of perirenal fat and large adrenal glands may obscure the
CHAPTER 18 Prenatal Genetic Diagnosis
diagnosis of renal agenesis (Hill et al, 1983). Renal cysts,
hydronephrosis, and obstructive uropathy are easily visualized. Oligohydramnios is indicative of poor renal function.
Multiple standard curves have been developed for fetal
anthropometric measurements (Elejalde and Elejalde,
1986; Saul et al, 1988). These instruments are particularly
helpful in diagnosing skeletal dysplasias and evaluating
growth restriction. Fetal genitalia may be determined reliably in the second trimester. In addition, ultrasonographic
examination is of benefit in the diagnosis and management
of multiple pregnancies.
Although there have been no documented adverse outcomes related to ultrasound exposure during human pregnancy, the reported experimental biologic effects—altered
immune response, cell death, change in cell membrane
functions, formation of free radicals, and reduced cell
reproductive potential—necessitate judicious use of this
technology. Another concern is the appropriate pediatric
follow-up for prenatally observed conditions with unclear
clinical significance, such as minimal hydronephrosis and
echogenic bowel.
SECOND TRIMESTER GENETIC SONOGRAM
The genetic sonogram is a term that is used to describe the
use of a second-trimester sonographic examination to
adjust the risk of fetal aneuploidy (Breathnach et al, 2007).
The genetic sonogram relies on the identification of major
structural anomalies in association with soft markers—that
is, findings that are normal variants that often resolve in
the third trimester. An example of a soft marker for trisomy
18 is the presence of choroid plexus cysts. Therefore the
presence of choroid plexus cysts, with other soft markers
such as echogenic bowel, single umbilical artery, or both,
in association with an omphalocele or a neural tube defect,
would strongly increase the suspicion of fetal trisomy 18.
NEW IMAGING MODALITIES
New fetal imaging modalites include three-dimentional
ultrasound examination and magnetic resonance imaging
(Lee and Simpson, 2007). Three-dimensional sonography
improves detection of cleft palate, and magnetic resonance
imaging improves assessment of fetal brain anomalies.
CELL-FREE NUCLEIC ACIDS IN MATERNAL
BLOOD
Lo et al (1997) first described the circulation of large
amounts of cell-free DNA in maternal plasma and serum
samples. Since then, knowledge regarding the biology of
fetal cell-free DNA and mRNA in the maternal circulation
has greatly expanded (Maron and Bianchi, 2007). The placenta is the predominant source of the circulating nucleic
acids. To date, two main clinical applications have transitioned from bench to bedside: fetal rhesus D genotyping (Bianchi et al, 2005) and fetal gender determination
(Rijnders et al, 2001). Both have been routinely available
in the United Kingdom and Europe for a number of years
and are just beginning to be available in the United States
(Hill et al, 2010). Fetal gender determination is particularly useful in guiding maternal steroid administration in
183
patients at risk of congenital adrenal hyperplasia, as well as
aiding further management of fetuses at risk for X-linked
disorders. Extensive research is ongoing toward using circulating cell-free fetal nucleic acids for the noninvasive
prenatal diagnosis of aneuploidy and in developing assays
of normal fetal developmental gene expression at different
gestational ages (Maron et al, 2007).
INVASIVE TECHNIQUES
AMNIOCENTESIS
Amniocentesis refers to the removal of up to 20 mL of amniotic fluid from the pregnant uterus. Contained within this
fluid are cellular components (desquamated fetal epithelial
and bladder cells) that serve as sources of chromosomes,
DNA, or enzymes. Most of the cellular elements are nonviable; therefore amniocytes generally require tissue culture under specific conditions to provide enough material
for diagnosis (Gosden, 1983). Herein lies one of the major
disadvantages of the procedure, in that results are received
late in the second trimester after fetal movement has been
perceived by the pregnant woman. In contrast, the amniotic
fluid itself may be assayed biochemically immediately after
being removed for the presence of AFP, acetylcholinesterase,
bilirubin, lecithin, sphingomyelin, or phosphatidylcholine.
The indications for genetic amniocentesis are (1) an
abnormal maternal serum screen result indicating an
increased risk of aneuploidy or neural tube defect,
(2) maternal age of 35 years or older at the time of delivery, because there is an increased risk for fetal chromosome abnormalities, (3) a previous pregnancy that resulted
in a fetus or an infant with chromosome abnormalities,
(4) one parent with a balanced chromosome translocation,
(5) a family history of a child with a neural tube defect,
(6) a family history of a metabolic disorder for which the
enzyme defect is known, (7) maternal history of an X-linked
disorder, (8) a family history of a disorder for which DNA
diagnosis is available, and (9) detection of a sonographic
abnormality that is associated with aneuploidy.
Extensive clinical experience with amniocentesis has
accrued over the past 30 years (Simpson et al, 1976; U.S.
National Institutes of Health, 1976). Historically, institutions in the United States currently quoted a 1% to 2%
incidence of minor complications, such as amniotic fluid
leakage, uterine cramping, and vaginal spotting after the
procedure. The incidence of more serious complications,
such as chorioamnionitis and miscarriage, is 0.25% to
0.5% (Centers for Disease Control and Prevention, 1995).
More recently, however, the rate of fetal loss after secondtrimester amniocentesis was calculated using data from the
FASTER trial (Eddleman et al, 2006). The spontaneous
fetal loss rate at less than 24 weeks’ gestation did not differ statistically significantly between those that did and did
not undergo the procedure. The procedure-related loss
rate after amniocentesis was 0.06%.
Because results of amniocentesis are received relatively late in the pregnancy, the Canadian early and Mid-
Trimester Amniocentesis Trial focused on evaluation of
the procedure when performed between 12 and 15 weeks’
gestation. This study showed a higher fetal loss rate and
a 1.3% risk of fetal clubfoot when amniocentesis was
184
PART V
Genetics
performed between 11 and 13 weeks’ gestation (Canadian
Early and Mid-Trimester Amniocentesis Trial Group,
1998). The rate of clubfoot in the early amniocentesis
group was tenfold the risk in the general population. The
cause of clubfoot is thought to be a disruption of normal
foot development secondary to transient oligohydramnios
(Farrell et al, 1999). For these reasons, early amniocentesis
is not generally recommended.
Decidua
Basalis
Amniotic
Cavity
CHORIONIC VILLUS SAMPLING
The increased use of first-trimester biochemical and ultrasound screening has increased uptake of chorionic villus
sampling as the diagnostic follow-up procedure of choice.
Chorionic villus sampling (CVS) involves the aspiration
of the chorion frondosum between 10 and 11 weeks’ gestation (Figure 18-2). The fact that the procedure is performed early is advantageous, because most women at this
point do not have external manifestations of pregnancy
and have not yet perceived fetal movement. The chorionic
villi are composed of syncytiotrophoblast and mesenchymal core cells that are actively growing and dividing. In
contrast to the dying epithelial cells shed into the amniotic fluid, chorionic villus cells do not require prolonged
culture to provide enough mitoses for a cytogenetic diagnosis. Karyotype results are generally available within
1 week of the procedure. Initially, direct preparations
derived from syncytiotrophoblast were used for analysis,
but the number of apparently mosaic abnormal results
proved unacceptable. Cultured preparations derived from
the cell of the mesenchymal core are more closely related
in embryonic origin to the actual fetus (Bianchi et al,
1993). It is currently recommended that both direct and
cultured preparations be used for cytogenetic analysis.
Mosaicism, defined as the presence of two or more cell
lines carrying different chromosomal constitutions, is a
true biologic (not technical) problem in CVS. In several
large studies, 0.8% to 1.7% of 1000 cases demonstrated
a chromosome abnormality that was present in the villus
but not in the fetus (Hogge et al, 1986; Ledbetter et al,
1992) This finding has led to the observation that postzygotic nondisjunction is more common than was previously
suggested.
The indications for CVS are the same as those for
amniocentesis, with two exceptions. First, neural tube
defects cannot be diagnosed by this procedure, and AFP
or other serum screening is not routinely offered at this
early point in gestation. Second, evidence shows that the
fetomaternal hemorrhage associated with placental biopsy
results in elevated maternal serum AFP immediately after
the procedure (Brambati et al, 1988).
CVS is performed transcervically or transabdominally.
With the transcervical technique, the inherent risks of fetal
and maternal infection appear to be greater, because it is
impossible to sterilize the cervix. Under ultrasonographic
guidance, a flexible catheter is passed through the endocervix and placed into the chorion frondosum. A small segment of placenta is then aspirated into sterile tissue culture
medium, and the catheter is withdrawn (Jackson, 1985).
In contrast, the transabdominal technique uses a needle
to obtain villus material; sterilization of the skin surface
is straightforward (Brambati et al, 1988). With either
Chorion
Laeve
Coelomic
Space
Chorion
Frondosum
FIGURE 18-2 A pregnant uterus containing a fetus at approximately 9 weeks’ gestation. The chorion frondosum, if sampled for
biopsy, can provide fetal cells for chromosome, enzyme, or DNA analysis. (From Jackson LG: First trimester diagnosis of fetal genetic disorders,
Hosp Pract (Off Ed) 20:40, 1985.)
method, approximately 10 to 50 mg of tissue is obtained.
Subsequently, the villi are dissected from maternal decidua
and processed for tissue culture or DNA extraction.
The safety and accuracy of these techniques have been
extensively monitored. A randomized National Institutes
of Health clinical trial compared amniocentesis with CVS
(Rhoads et al, 1989). The Centers for Disease Control
and Prevention and the National Institutes of Health held
a consensus conference to summarize worldwide experience. After adjustment for confounding factors such as
gestational age, the risk of miscarriage after CVS was
found to be on the order of 0.5% to 1.0% (Centers for
Disease Control and Prevention, 1995).
The advantages and disadvantages of CVS are summarized in Box 18-3. For patients at high risk for single-gene
disorders amenable to DNA diagnosis (e.g., cystic fibrosis,
sickle cell anemia, Duchenne muscular dystrophy), CVS
is probably the preferred prenatal diagnostic method.
Alternatively, for patients with relatively low risk (e.g., a
35-year-old woman being tested for chromosome abnormalities), an amniocentesis may be more appropriate.
There have been a few reports of serious maternal sepsis
and transient bacteremia in association with transcervical
CVS (Barela et al, 1986; Silverman et al, 1994).
The 1% incidence of mosaicism in villus samples may
necessitate a further invasive technique, such as amniocentesis or cordocentesis, to confirm or refute diagnoses.
However, the detection of mosaicism in a CVS sample
may identify a fetus at risk for uniparental disomy. Uniparental disomy refers to the inheritance of both copies of
a chromosome pair from a single parent. This inheritance
occurs when a trisomic fetus undergoes rescue and loses
CHAPTER 18 Prenatal Genetic Diagnosis
BOX 18-3 A
dvantages and Disadvantages
of Chorionic Villus Sampling
ADVANTAGES
Performed in first trimester; results available quickly
ll Cells obtained are mitotically active
ll Amount of tissue obtained is preferable for DNA analysis
ll Placental mosaicism is detected
ll
DISADVANTAGES
ll Miscarriage rate slightly higher
ll Increased fetomaternal hemorrhage after procedure
ll Risk of serious maternal infection
ll Risk of fetal limb and jaw malformations
the extra copy of the chromosome. One third of the time,
the fetus will be left with two chromosomes that originated
in a single parent. Uniparental disomy is an important
mechanism in conditions such as Prader-Willi syndrome.
Initially, there was concern regarding the association
between CVS and the risk of limb deficiencies in infants
whose mothers underwent the procedure. The risk of limb
malformations was first suggested by a number of studies describing an increased incidence of transverse limb
anomalies and the hypoglossia-hypodactyly syndrome in
infants whose mothers had undergone CVS (Burton et al,
1992; Firth et al, 1991; Firth et al, 1994; Hsieh et al, 1995).
The overall rate of non–syndrome-related transverse limb
deficiency from 65 centers performing CVS is 7.4 per
10,000 procedures (Centers for Disease Control and Prevention, 1995). This number is reasonably similar to that
in population-based registries that monitor infants with all
limb deficiencies, which give a rate of 5 to 6 per 10,000 live
births (Centers for Disease Control and Prevention, 1995).
Factors likely to influence the rate of limb malformations
include gestational age (risk of limb deficiency is greatest
at or before 9 weeks’ gestation and decreased at or after 10
weeks’ gestation), type of catheter used, and operator experience. The overall risk of limb deficiency appears to be on the
order of 1 per 3000 procedures (Centers for Disease Control
and Prevention, 1995). A higher incidence of hemangioma
has also been suggested in infants born after CVS (Burton
et al, 1995). The most important issue at present, however,
is the fact that relatively few maternal-fetal medicine and
obstetric clinics have personnel who are adequately trained
to perform the procedure (Cleary-Goldman et al, 2006).
CORDOCENTESIS
Percutaneous umbilical blood sampling, or cordocentesis,
was first described as a means of obtaining fetal immunoglobulin M (IgM) measurements in the prenatal diagnosis
of congenital toxoplasmosis (Daffos et al, 1985). Under
continuous ultrasonographic imaging, the insertion site
of the umbilical cord into the placenta is identified. The
umbilical vein is punctured with a 20-gauge needle, the
sample is withdrawn, and the umbilical cord is observed
for signs of hemorrhage. The technique has been used
diagnostically in many clinical settings (Forestier et al,
185
BOX 18-4 Indications for Fetal Blood
Sampling
ll
ll
ll
ll
ll
ll
ll
ll
Thrombocytopenia
Rapid third-trimester diagnosis of chromosome abnormalities*
Immunodeficiency
Congenital infection
Acid-base abnormalities
Metabolic disorders*
Fetal blood type incompatibility*
Hemoglobinopathy*
*Diagnosis can also be made by a DNA-based test performed on any nucleated fetal material,
such as amniocytes.
1988) (Box 18-4). Regarding genetic diagnosis, the lymphocytes are a source of cells for a rapid karyotype; this
is helpful in two situations: (1) when anomalies have been
noted on ultrasonographic examination, but it is too late in
gestation to perform an amniocentesis (antenatal diagnosis of trisomy 13 or 18 influences delivery room management), and (2) for confirmation of a fetal karyotype when
amniocentesis or CVS has shown mosaicism (Gosden
et al, 1988).
SUMMARY
There has been a major shift away from using maternal
age as an indication for prenatal genetic diagnosis. Instead,
the results of biochemical and sonographic screening tests,
which are increasingly being performed in the first trimester, are used to guide the need for amniocentesis and
CVS. Safety considerations and patient preferences have
influenced the emphasis on noninvasive prenatal diagnosis. The results of large-scale clinical trials, such as the
SURUSS and the FASTER study, have shown a high sensitivity and specificity for fetal Down syndrome detection
using noninvasive techniques. Circulating cell-free fetal
nucleic acids in maternal blood are already being used for
noninvasive diagnosis of fetal gender and rhesus D genotype, and many potential opportunities exist to use this
material for future clinical applications.
SUGGESTED READINGS
American College of Obstetrician and Gynecologists: Screening for fetal chromosomal abnormalities, ACOG Practice Bulletin No.77, Obstet Gynecol 109:
217-227, 2007.
Breathnach FM, Fleming A, Malone FD: The second trimester genetic sonogram,
Am J Med Genet C Semin Med Genet 145C:62-72, 2007.
Eddleman KA, Malone FD, Sullivan L, et al: Pregnancy loss rates after midtrimester amniocentesis, Obstet Gynecol 108:1067-1072, 2006.
Malone FD, Canick JA, Ball RH, et al: First-trimester or second-trimester screening, or both, for Down’s syndrome, N Engl J Med 353:2001-2011, 2005a.
Malone FD, Ball RH, Nyberg DA: First-trimester septated cystic hygroma:
prevalence, natural history, and pediatric outcome, Obstet Gynecol 106:288-294,
2005b.
Maron JL, Bianchi DW: Prenatal diagnosis using cell-free nucleic acids in maternal
body fluids: a decade of progress, Am J Med Genet C Semin Med Genet 145C:
5-17, 2007.
Said S, Malone FD: The use of nuchal translucency in contemporary obstetric
practice, Clin Obstet Gynecol 51:37-47, 2008.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
19
Evaluation of the Dysmorphic Infant
Chad R. Haldeman-Englert, Sulagna C. Saitta, and Elaine H. Zackai
Genetic disorders have a major impact on public health,
as indicated by several large epidemiologic studies (Table
19-1) (Hall et al, 1978; McCandless et al, 2004; Scriver
et al, 1973). The latest data indicate that genetic factors
contribute to more than two thirds of the conditions
prompting admission to a children’s hospital (McCandless et al, 2004). Early identification of the genetic nature
of a given condition may then help to appropriately focus
resources for providing better care to these individuals. It
is therefore critical to implement a systematic approach to
evaluating a dysmorphic or malformed infant. This chapter outlines such a general approach.
The clinical geneticist incorporates the following five
essential tools in the evaluation of a child suspected of having a primary genetic disorder:
ll History: prenatal, birth, and medical
ll Pedigree analysis or family history
ll Specialized
clinical evaluation that includes a
detailed dysmorphology examination
ll Comprehensive literature search
ll Focused genetic laboratory analyses (e.g., chromosomes, fluorescence in situ hybridization, DNA
microarray, sequencing)
HISTORY
PRENATAL
A complete gestational history should be generated,
including results of prenatal testing such as maternal
serum screening, ultrasonography, chorionic villus sampling (CVS), and amniocentesis (Box 19-1). The maternal age at conception should be documented, because the
risk of chromosomal anomalies such as nondisjunction
rises with maternal age. It is important to identify prenatal exposures to infection and medications, maternal habits
such as alcohol and drug use, maternal chronic illnesses
such as maternal diabetes, and pregnancy-related complications. An additional significant historical component
involves the presence of abnormal levels of amniotic fluid.
Oligohydramnios (decreased amniotic fluid volume) can
be associated with either a fluid leak or a genitourinary
abnormality, whereas polyhydramnios (excess amniotic
fluid volume) can be seen in fetuses with neuromuscular
disease or gastrointestinal malformations.
It is also important to identify exposure to environmental
agents that might act as teratogens. Teratogens are environmental agents that may cause structural and functional
diseases in an exposed fetus. Each teratogen may have a
characteristic expression pattern, with a specific range of
associated structural anomalies and dysmorphic features.
Effects and the extent of the effects depend on the time
of exposure, duration, and dosage as well as interactions
186
TABLE 19-1 Genetic Disorders in Pediatric Hospital Admissions
Genetic disorders
Montreal
(1973)
Chromosome,
single gene (%)
7.3
Seattle
(1978)
Cleveland
(2004)
4.5
11
Polygenic (%)
29
49
60
Nongenetic
disorders (%)
64
47
29
12,801
4115
5747
Total number of
admissions
Data from Hall JG, Powers EK, McIlvane RT, et al: The frequency and financial
burden of genetic disease in a pediatric hospital, Am J Med Genet 1:417-436, 1978;
McCandless SE, Brunger JW, Cassidy SB: The burden of genetic disease on inpatient
care in a children’s hospital, Am J Hum Genet 4:121-127, 2004; and Scriver CR, Neal
JL, Saginur R, et al: The frequency of genetic disease and congenital malformation
among patients in a pediatric hospital, Can Med Assoc J 108:1111-1115, 1973.
BOX 19-1 Elements of Prenatal History
for the Dysmorphic Infant
MATERNAL HEALTH
Age
Disease: diabetes, hypertension, seizure disorder
MODE OF CONCEPTION
Natural
Assisted reproductive technologies
Fertility medications
In vitro fertilizatioin
Intracytoplasmic sperm injection
Gamete intrafallopian transfer
Artificial insemination
EXPOSURES
Medications
Alcohol
Environmental agents
Infections (gestational age at exposure)
PRENATAL TESTING
Ultrasonography (gestational age performed)
Triple screen
Chorionic villus sampling, amniocentesis, and indications
with maternal and genetic susceptibility factors. In general, more severe effects are typically correlated with
exposure early in the pregnancy and with more extensive
(i.e., higher dose) exposure. The list of well-documented
human teratogens is short and includes such substances as
alcohol, thalidomide, warfarin, trimethadione, valproate,
and hydantoin. If history of an exposure is documented, an
effort should be made to identify the developmental time
and level of exposure. This information is critical, because
CHAPTER 19 Evaluation of the Dysmorphic Infant
Male
Marriage or union
Female
Divorced
Sex unspecified
Consanguinity
Number of children
of sex indicated
Monozygotic twins
187
TABLE 19-2 Dermatoglyphic Patterns Associated With Specific
Dysmorphic Disorders
Dermatoglyphic Pattern
Associated Disorders
Excess arches
Trisomy 13, trisomy 18, Klinefelter syndrome (47,XXY),
deletion 5p (cri du chat), fetal
phenytoin exposure
Excess ulnar loops
Trisomy 21
Excess whorls
Smith-Lemli-Opitz syndrome,
Turner syndrome (45,X), 18q
deletion
3
2
Affected
Dizygotic twins
Nonpenetrant carrier,
may manifest disease
Obligate carrier, will
not manifest disease
?
BOX 19-2 Adjunct Studies in the Evaluation
of the Dysmorphic Infant
ll
ll
ll
ll
ll
ll
ll
ll
ll
Investigation of internal malformation
Assessment of neurologic function
Identification of organ systems involved
Ultrasonography magnetic resonance imaging
Brain imaging
Electroencephalography as indicated
Electromyography as indicated
Prognosis
Treatment and intervention
the counseling and calculation of recurrence risk for a
given malformation are vastly different if environmental
exposures are involved.
BIRTH
Another important component of the gestational history is
obtaining information on fetal activity, size, and position.
Often the mother’s subjective impressions can be further
confirmed by examining obstetric records of the perinatal period. A history of hypotonia may be further supplemented by reports of poor fetal movements and breech
presentation. Perinatal information including gestational
age, fetal position at delivery, the length of labor, type of
delivery, and any evidence of fetal distress, such as passage
of meconium, are all relevant data (Table 19-2). Apgar
scores, the need for resuscitation, birth parameters (weight,
length, and head circumference), any malformations seen
at birth, and all abnormal test results should be noted.
MEDICAL
A full review of the medical issues of the child should
include the baby’s general health, test results, identification
of any chronic medical issues, and need for hospitalization.
Evaluation of growth, review of systems, developmental
assessment, and notation of unusual behaviors can also
provide important clues to a diagnosis.
PEDIGREE ANALYSIS AND FAMILY
HISTORY
A critical part of any genetic evaluation is to obtain the
family history (Box 19-2); this is best accomplished by creating a three-generation pedigree, which is a schematic
II
2
1
I
Proband
Twins of unknown zygosity
1
2
3
Pedigree with
generations and
individuals numbered
Diseased individual
Miscarriage
Sillbirth
No offspring
Adopted into family
Adopted out of family
1
2
Multiple unions
FIGURE 19-1 Symbols commonly used for pedigree notation. (From
Nussbaum RL, McInnes RR, Willard HF, eds: Thompson and Thompson’s
genetics in medicine, ed 7, Philadelphia, 2007, WB Saunders, p 117.)
diagram depicting familial relationships using standard
accepted symbols (Figure 19-1). This formal record can
also be used to summarize positive responses elicited
during the interview. Special attention should be paid to
ethnic origins of both sides of the family, consanguinity,
and any first-degree relatives with similar malformations
to those of the patient being evaluated, also known as the
index case, proband, or propositus. An extended family history
should be used to identify relatives with congenital anomalies, developmental abnormalities, or physical differences.
Often photographs can provide clear objective evidence of
a descriptive history.
Reproductive histories, especially of the parents, should
be elicited. Specifically, questions should be asked about
infertility, miscarriages, and stillbirths. The occurrence of
more than two first-trimester miscarriages increases the
probability of finding a balanced translocation in one parent (Campana et al, 1986; Castle and Bernstein, 1988).
A balanced translocation is a rearrangement of genetic
material such that two chromosomes have an equal
exchange without loss or gain of material. There are typically no associated clinical features with such a rearrangement. However, when chromosomes align to recombine
for meiosis in the sperm or egg, this exchange produces a
risk of unequal distribution and an unbalanced translocation in the resulting fetus. In this case, there would be
aneuploidy for part of a chromosome. It has been estimated that 25% of stillbirths exhibit single or multiple
malformations, and in at least half of these cases there is a
genetic etiology for the malformations. Couples with two
or more pregnancy losses should undergo routine chromosome analysis or karyotyping. When possible, such
analysis should be performed on the stillborn fetus or on
products of conception.
188
PART V
Genetics
Obtaining a formal family history is helpful in discovering information that is often critical to making a diagnosis.
Positive responses may help to discern a Mendelian pattern of inheritance for a given genetic disorder. For example, a disease affecting every generation, with both males
and females involved, such as Marfan syndrome, would
most likely be autosomal dominant. A pattern of X-linked
recessive disease, such as hemophilia, would show affected
males related through unaffected or minimally affected
females; transmission in this pattern should not occur from
father to son.
A congenital malformation can be described as a “morphologic defect of an organ, part of an organ, or larger
region of the body resulting from an intrinsically abnormal
developmental process” (Jones, 2006). The term dysmorphology was introduced by Dr. David Smith in the 1960s
to describe the study of human congenital malformations
(Aase, 1990). This study of “abnormal form” emphasizes a
focus on structural errors in development with an attempt
to identify the underlying genetic etiology and pathogenesis of the disorder.
In a landmark study, Feingold and Bossert (1974) examined more than 2000 children to define normal values for a
number of physical features. These standards were devised
as screening tools to objectively identify children with differences possibly attributable to a genetic disorder. Important measurements include head circumference, inner
and outer canthal distances, interpupillary distances, ear
length, ear placement, internipple distances, chest circumference, and hand and foot lengths. Other graphs and measurements using age-appropriate standards can be found in
compendia such as the Handbook of Physical Measurements
(Hall et al, 2007).
The assessment should begin with newborn growth
parameters that can reflect the degree of any prenatal insult. Measurements such as height, weight (usually
reflecting nutrition), and head circumference should be
plotted on newborn graphs. Gestational age–appropriate
graphs should be used for premature infants. It is often
helpful to express values that are outside the normal range
as 50th percentile for a different gestational age. For example, a full-term baby with microcephaly may have a head
circumference of less than the 5th percentile for 38 weeks.
This can be expressed as a measurement at the 50th percentile for 33 weeks, which imparts the degree of microcephaly more clearly.
A complete physical examination should include assessment of patient anatomy for features varying from usual
or normal standards. This assessment can often provide
clues to embryologic mechanisms. The data obtained
should then be interpreted in regard to normal standards
using comprehensive standard tables that are available
for these purposes. Special attention to familial variants
should be given.
The shape and size of the head and fontanels should be
noted as well as the cranial sutures, with assessment for evidence of craniosynostosis or an underlying brain malformation. Any scalp defects should also be noted. The shape
Interpupillary distance
PHYSICAL EXAMINATION FOR
DYSMORPHOLOGY
in
2.8
cm
7.0
2.6
6.5
2.4
6.0
2.2
5.5
2
5.0
1.8
4.5
1.6
4.0
1.4
3.5
1.2
3.0
1
2.5
)
.5
(1 4)
0
.
4. (1 2)
5 .
3. (1 .0)
0
3. 5 (1 9)
2. (0. )
0 .6
2. (0 .4)
5
1. 0 (0
1.
Inner
canthal
distance
4
5
6
7
8
9
10
cm
1.6
2
2.4
2.6
3.2
3.6
4
in
Outer canthal distance
FIGURE 19-2 Various eye measurements are depicted (top). A indicates the outer canthal distance, B indicates the inner canthal distance,
and C indicates the interpupillary distance (IPD), which is difficult to
measure directly. The IPD can be determined using the graph at the
bottom or with the Pryor formula: IPD = (A − B) 2 + B. (From Feingold M,
Bossert WH: Normal values for selected physical parameters: an aid to syndrome delineation, Birth Defects 10:1-16, 1974.)
of the forehead, appearance of the eyebrows (noting synophrys), and the texture and distribution of hair should be
noted. The spacing of the eyes, or canthal measurements
(Figure 19-2), the interpupillary distances (Figure 19-3;
see also Figure 19-2), palpebral fissure lengths (Figure
19-4), presence or absence of colobomata and epicanthal
folds, and noting whether the palpebral fissures are turned
upward or downward are components of the dysmorphology examination. Examination of the ears should include
a search for preauricular and postauricular pits, tags, and
assessment of the placement (Figure 19-5), length (Figure
19-6), and folding of the ear is important. Ear development
occurs in a temporal frame similar to that of the kidneys,
and external ear anomalies can be associated with renal
anomalies. Evaluation of the nose should cover the shape
of nasal tip, the alae nasi, presence of anteverted nares,
the length of the columella, and patency of the choanae.
189
CHAPTER 19 Evaluation of the Dysmorphic Infant
The mouth and throat are examined for the presence of
a cleft lip or palate; the shape of the palate and uvula are
noted, and the presence of unusual features, such as tongue
deformities, lip pits, frenula, and natal teeth, are recorded.
A small retrognathic or receding chin, which can be a part
of several syndromes or an isolated finding, should be
noted. The neck is inspected for excess nuchal folds or skin
and evidence of webbing. Any bony abnormalities in the
neck should prompt an evaluation of the cervical vertebrae
to confirm cervical and airway stability.
Evaluation of the chest and thorax involves lung auscultation and cardiac examination. Abnormal findings should
prompt a consultation with a cardiologist and appropriate
echocardiographic or invasive studies as needed. External
measurements include determining the internipple distance and its ratio in regard to the chest circumference
(Figure 19-7). The abdominal examination is focused on
determining whether organomegaly is present, a finding
typically associated with an inborn error of metabolism.
The umbilicus should also be examined, with any hernias
and the number of vessels present in the newborn cord
being noted. A two-vessel cord, in which only a single
artery is present, can be associated with renal anomalies.
The genitourinary examination concentrates on determining whether anomalies such as hypospadias, chordee,
cryptorchidism, microphallus, and ambiguous genitalia are
present. These external anomalies may be associated with
internal anomalies involving the upper urinary tract as
34
250
1.3
32
150
Mean
Interpupillary distance
6
75
50
25
2.25
2.00
5
3
1.75
1.50
28
1.2
150
250
1.1
26
1.0
24
0.9
22
0.8
20
4
18
3
0
3
6
9
12
15
18
21
2
3
4
5
6
7
8
9
10
11
12
13
14
1.25
Months
Years
Age
FIGURE 19-3 A nomogram for interpupillary distance at different
ages for both sexes. (From Feingold M, Bossert WH: Normal values for
selected physical parameters: an aid to syndrome delineation, Birth Defects
10:1-16, 1974.)
Inches
%
97
in cm
Palpebral fissure length (mm)
30
16
0.7
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
Age from birth (years)
FIGURE 19-4 A graph of palpebral fissure length from birth to age
16 years for both sexes. (From Hall JG, Allanson JE, Gripp KW, et al:
Handbook of physical measurements, Oxford, 2007, Oxford University
Press.)
FIGURE 19-5 Ear placement.
Using the medial canthi (A and B)
as landmarks, one draws a central
horizontal line and extends it to a
point (C ) on the side of the face. Ears
attached below this line are considered low set.
190
PART V
Genetics
well. The anus is examined for evidence of tags, its placement, and its patency.
The back should be assessed, especially for the shape of
the spine and any associated defects, such as myelomeningocele. These defects prompt further radiologic evaluation
to assess for potential functional limitations. In addition, a
sacral dimple or hair tuft at the base of the spine should be
noted, because either could signify developmental abnormalities in the underlying neural tissue.
Minor anomalies are often manifested in the extremities. Gross differences in the hands and feet include polydactyly (more than five digits), whether the extra digits are
located in a preaxial or postaxial position should be noted,
syndactyly (fusion of the digits), clinodactyly (incurving of the digits), and extremity length, which should be
expressed as a percentile measured on age-appropriate
in cm
7
97
Total ear length
2.5
%
75
50
6
2.25
25
2
5
3
1.75
4
1.50
0
3
6
9
12
15
18
21
2
3
4
5
6
7
8
9
10
11
12
13
14
3
Months
Years
Age
FIGURE 19-6 Graph showing various percentiles for ear length plotted against age. (From Feingold M, Bossert WH: Normal values for selected
physical parameters: an aid to syndrome delineation, Birth Defects 10:1-16,
1974.)
graphs (Figures 19-8 and 19-9). Often these data can provide important clues to a unifying syndrome.
Dermal ridge patterns, or dermatoglyphics, are formed
on the palms and soles early in embryonic life, and they
vary considerably among individuals. This variation can
be inherited and can be influenced by disturbances to the
development of the peripheral limb buds. Environmental
exposures and chromosomal aberrations can greatly affect
the formation of these structures and are reflected by the
dermatoglyphic pattern of an individual. Each of the distal phalanges has one of three basic dermal ridge patterns:
arches, whorls, or loops (Figure 19-10). The predominance of a single pattern can be an associated feature of
a genetic disorder. For example, the occurrence of arches
on eight or more digits is a rare event, but is frequently
encountered in children with trisomy 18 (Box 19-3).
Deltas, or triradii, form at the convergence of three sets
of ridges on the palm. This junction is where the hypothenar, thenar, and distal palmar patterns converge. There
are typically no triradii in the hypothenar area of the palm,
but when patterning is present or is large, a distal triradius arises, which is found in only 4% of normal Caucasian
individuals but in 85% of patients with trisomy 21 (Down
syndrome). A single transverse palmar crease is found in
4% of controls, but in more than half of patients with trisomy 21 (Figure 19-11) and in even greater proportions in
patients with other trisomies. The hallucal area of the foot,
located at the base of the big toe, also has a dermal ridge
pattern, usually a loop or whorl. A simple pattern or open
field in this region is found in less than 1% of controls
but in more than 50% of patients with Down syndrome
(Figure 19-12). This unusual dermal pattern is also associated with hypoplasia of the hallucal pad and a wide space
between the great and second toes in these patients.
An examination of the skin is also important, to look for
phakomatoses or skin manifestations that herald the presence of an underlying disorder. Examples are café-au-lait
spots (associated with neurofibromatosis type I) and ash
leaf spots (associated with tuberous sclerosis and detected
40
38
36
34
32
30
28
26
24
22
20
18
16
14
12
10
8
cm
%
20
97
75
50
25
3
19
Years
Age
FIGURE 19-7 The internipple distance as a percentage of the chest
circumference plotted against age for both sexes. (From Feingold M,
Bossert WH: Normal values for selected physical parameters: an aid to syndrome delineation, Birth Defects 10:1-16, 1974.)
17
6
15
5
13
4
11
9
3
7
2
5
0
3
6
9
12
15
18
21
2
3
4
5
6
7
8
9
10
11
12
13
14
Months
Total hand length
7
%
97
75
50
25
3
0
3
6
9
12
15
18
21
2
3
4
5
6
7
8
9
10
11
12
13
14
Internipple distance as % of
chest circumference
in
Months
Years
Age
FIGURE 19-8 The total hand length plotted against age for both
sexes. (From Feingold M, Bossert WH: Normal values for selected physical
parameters: an aid to syndrome delineation, Birth Defects 10:1-16, 1974.)
97
75
50
25
A
11
10
3
7
6
5
Inches
8
Foot length, females (cm)
9
4
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10
9
8
7
6
97
75
50
25
10
9
3
8
7
6
5
4
3
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
3
Age (years)
191
Inches
29
28
27
26
25
24
23
22
21
20
19
18
17
16
15
14
13
12
11
10
9
8
7
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
Foot length, males (cm)
CHAPTER 19 Evaluation of the Dysmorphic Infant
B
Age (years)
FIGURE 19-9 Total foot lengths
plotted against age for boys (A) and
girls (B). (From Hall JG, Allanson
JE, Gripp KW, et al: Handbook of
physical measurements, Oxford, 2007,
Oxford University Press.)
BOX 19-3 Underlying Mechanisms
of Malformation
SYNDROME
Pathogenetically related pattern of anomalies
SEQUENCE
Pattern of anomalies derived from a presumed or known prior anomaly
or mechanical disturbance
Simple Arch
ASSOCIATION
Nonrandom occurrence of multiple anomalies
FIELD DEFECT
Disturbance of a developmental field leading to a pattern of anomalies
Loop
with the use of a Wood’s lamp). Irregular pigmentation,
such as hypomelanosis of Ito, can be suggestive of chromosomal mosaicism, in which the different skin pigmentation
patterns represent a different, mixed chromosomal composition. Hemangiomas and other skin diseases are also
noteworthy.
Finally a careful neurologic examination with input from
a specialist is often warranted in the child with multiple
anomalies, because the neurologic status is often the most
reliable prognostic indicator. Evaluation of tone, feeding,
unusual movements, and the presence of seizure activity
are critical pieces of diagnostic information.
ADJUNCT STUDIES
Whorl (Spiral)
FIGURE 19-10 Basic fingerprint patterns (dermatoglyphics). (From
Holt SB: The genetics of dermal ridges, Springfield, Ill, 1968, Charles C
Thomas.)
An exhaustive physical examination often reveals differences that require further evaluation for diagnostic, prognostic, and treatment purposes (Box 19-4). Poor feeding
or cyanosis may lead to detection of internal organ malformations on echocardiogram or abdominal ultrasonography. Differences in head shape suggest the need for skull
radiographs, three-dimensional computed tomography,
192
PART V
Genetics
BOX 19-4 P
rocesses Leading to Altered
Form or Structure
DEFORMATION
Abnormal form resulting from mechanical forces
DISRUPTION
Morphologic defect caused by interference with a previously normal
developmental process
DYSPLASIA
Altered morphology because of abnormal organization of cells into a given
tissue
BOX 19-5 Examples of Morphologic
Differences
ll
ll
ll
ll
ll
FIGURE 19-11 The patterns on the hand of a patient with Down
syndrome depicting the palmar crease (D). (From Holt SB: The genetics
of dermal ridges, Springfield, Ill, 1968, Charles C Thomas.)
ll
ll
ll
ll
Malformation
Deformation
Disruption
Dysplasia
Cardiac septal defects, cleft lip
Club foot
Amniotic bands
Localized: hemangioma
Generalized (skeletal): achondroplasia
electroretinograms are needed to predict visual prognosis.
Often there are well-characterized genetic disorders that
have a specific pattern of abnormal findings in these highly
specialized studies. Even when a unifying diagnosis is
reached, there is often variation in the clinical phenotype,
and determining the patient’s prognosis on a system-bysystem basis is typically the most appropriate and accurate
way to proceed.
LITERATURE REVIEW
FIGURE 19-12 The distal sole of the foot of a patient with Down
syndrome, depicting the characteristic “open field” pattern. (From
Holt SB: The genetics of dermal ridges, Springfield, Ill, 1968, Charles C
Thomas.)
or magnetic resonance imaging of the brain. A disproportionality of the limbs prompts a skeletal survey and bone
age measurement.
It is prudent in children with anomalies involving multiple systems to obtain the input of relevant specialists. This
step is often essential to medical decision making and the
planning of interventions. Abnormal neurologic findings
should prompt a consultation with a trained specialist and
interpretation of studies such as head ultrasonography,
brain magnetic resonance imaging, brainstem auditory
evoked responses, and electroencephalogram for seizure
activity. Muscle dysfunction might result in the ordering
of electromyography, muscle ultrasound, or nerve conduction studies. Visual involvement requires a funduscopic
examination by an experienced pediatric ophthalmologist, and sometimes studies of visual evoked responses or
The occurrence of malformations can fit into one of several categories (Box 19-5). The next step toward reaching a diagnosis is to analyze the data generated from the
evaluation and attempt to categorize the findings. A syndrome is a “collection of anomalies involving more than
one developmental region or organ system,” (Aase, 1990).
The word itself means a “running together” or “pattern of
multiple anomalies thought to be pathogenically related.”
Therefore a given congenital anomaly may be an isolated defect in an otherwise normal individual or part of
a multiple malformation syndrome. Furthermore, the primary malformation itself can determine additional defects
through an interrelated cascade of physical and functional
processes; if ensuing malformations are related to one primary defect, factor, or event, a pathogenetic sequence has
occurred. A classic example is the Pierre-Robin sequence
or constellation, consisting of a small recessed jaw, midline
U-shaped cleft palate, and relatively large and protruding
tongue. The primary anomaly is the small jaw, which does
not allow adequate room for the tongue and displaces it
superiorly. The displaced tongue prevents closure of the
palatine shelves, causing the cleft palate. The recurrence
risk with this isolated occurrence is negligible. However,
CHAPTER 19 Evaluation of the Dysmorphic Infant
BOX 19-6 E
lements of Perinatal and Birth
History for the Dysmorphic Infant
ll
ll
ll
ll
ll
ll
ll
ll
Fetal activity
Delivery
Type (e.g., indication for cesarean section)
Gestational age
Fetal presentation
Apgar scores, history of distress, or resuscitation
Growth parameters
Malformations noted
one should also remember that such sequences can also
be part of a larger constellation of findings that does fit
into a syndrome, as Pierre-Robin sequence can when it is
part of velocardiofacial syndrome (caused by a deletion of
chromosome 22q11) or Stickler syndrome (associated with
mutations in the type II collagen gene). The recurrence
risk for an affected individual passing on these particular
syndromes to their children is 50%.
In addition, a cluster of several malformations that are
not developmentally related can occur in a nonrandom
fashion called an association that may appear without characteristic dysmorphic features. One such statistically nonrandom association of defects consists of vertebral defects,
anal atresia, cardiac defects, tracheoesophageal fistula,
esophageal atresia, renal dysplasia, and limb anomalies
(VACTERL). It should be noted that not all features need
to be present and that the extent of involvement of each
system is widely variable. Recently, the CHARGE syndrome (coloboma, heart disease, atresia choanae, retarded
growth and development, genital anomalies, and ear
anomalies/deafness) was considered an association until
the CHD7 gene was identified to cause approximately 60%
of the cases (Lalani et al, 2006).
These associations often manifest as sporadic rather
than familial occurrences. Because they are not clearly
related by a common etiology or pathogenesis, they are
not considered syndromes and do not technically constitute a diagnosis. Instead, they are a recognition of a statistically significant association of features. It is important to
remember that many of these same anomalies can occur as
features of chromosomal aneuploidy or other syndromes.
Syndromic malformations tend to occur in more than one
developmental field. A field defect or complex is a set of
primary malformations in a developmental field that originates from a single or primary abnormality in embryonic
development (see Box 19-5).
When generating a differential diagnosis of malformations that might occur together, the evaluator must also
consider structures that may appear abnormally formed,
but in fact are structures that underwent normal development and then received some insult that distorted their
true form (Box 19-6). For example, a deformation describes
the abnormal form, shape, or position of a part of the body
that was caused by mechanical forces. Examples are clubfoot, hip dislocation, and craniofacial asymmetry; they can
result from intrinsic (embryonic) or extrinsic (intrauterine)
mechanical forces that alter the shape or position of an
organ or part that had already undergone normal differentiation. Deformations are estimated to occur in 2% of births,
193
BOX 19-7 E
lements of Pedigree Analysis
and Family History for the
Dysmorphic Infant
Identification of relatives with:
Congenital anomalies (especially those similar to proband’s)
ll Mental retardation
Photographs (objective evidence)
Parental reproductive history
ll Pregnancy losses (gestational ages)
ll Infertility
Medical histories of primary relatives
Ethnic origin
Consanguinity
ll
and such factors as fetal crowding from the presence of
multiple fetuses and uterine malformations, as well as oligohydramnios, and a face presentation during delivery can
cause them.
Along similar lines, a disruption describes a “morphologic defect of an organ, part of an organ, or larger region
of the body resulting from the extrinsic breakdown of, or
an interference with, an originally normal developmental process,” (Aase, 1990). The classic example of a disruption is entanglement of the fetus in amniotic bands.
Amniotic bands are ribbons of amnion that have ruptured
in utero and cause disruptions of normal developmental
processes in the fetus, either through physical blockage
or interruption of the blood supply or by entangling and
tearing of developing structures. This effect is seen most
often with digits and limbs, and remnants of the bands, or
constriction marks, can frequently be seen at birth. If the
fetus should swallow a band, a cleft palate might result;
the etiology is a very different etiology from that of cleft
palate occurring as a primary malformation. Recurrence
risk counseling of the parent would be very different in
these two scenarios.
Dysplasias occur when there is “an abnormal organization of cells into tissue(s) and its morphologic results,”
(Aase, 1990). Dysplasia tends to be tissue specific rather
than organ specific (e.g., skeletal dysplasia) and can be
localized or generalized.
In summary, structural or morphologic changes identified at birth can occur during intrauterine development
as a result of malformations, deformations, disruptions, or
dysplasia. However, approximately 90% of deformations
undergo spontaneous correction. Malformations and disruptions often require surgical intervention when possible.
Dysplasias are typically not correctable, and the affected
individual experiences the clinical effects of the underlying
cell or tissue abnormality for life (see Box 19-7). Examples
of these entities are listed in Box 19-7.
After the history and physical evaluation are complete, a cross-reference of two or more anomalies is useful to generate a differential diagnosis. When the rest of
the neonate’s physical and history findings are added, the
possibilities can often be narrowed down to a few entities
that may be amenable to diagnostic testing. If multiple
anomalies are present, it is usually best to start with the
least common. As Aase (1990) has stated, “The best clues
are the rarest. The physical features that will be the most
194
PART V
Genetics
helpful on differential diagnosis are those infrequently
seen either in isolation or as part of syndromes. Quite
often, these are not the most obvious anomalies or even
the ones that have the greatest significance for the patient’s
health.” Cross-referencing is usually best accomplished by
using published compendia of malformation syndromes.
These compendia have been supplemented by databases
that are accessible online (i.e., GeneReviews, Online Mendelian Inheritance in Man, and PubMed). The availability of
such tools allows the cross-referenced features to be compared easily with those of other described syndromes that
may include similar malformations. This systematic review
produces a differential diagnosis for the constellation of
features described and identifies references to pertinent
literature.
The recognition of patterns of genetic entities involves
the comparison of the proband with the examiner’s personal experience of known cases and a search of the literature. Multiple anomalies may be causally related, occur
together in a statistically associated basis, or occur together
merely by chance. Diagnosis of a genetic disorder relies
heavily on the ability of the clinician to suspect, detect,
and correctly interpret physical and developmental findings and to recognize specific patterns. Accurate diagnosis
of a syndrome in a child is important to the identification
of major complications and their treatment if possible. It is
also crucial for long-term management of patients and for
parental counseling about recurrence in future offspring.
SPECIALIZED LABORATORY TESTS
In sorting through the array of possibilities listed, the
geneticist uses one other important tool—the availability
of highly specialized cytogenetic and molecular genetic
testing, including:
ll
ll
ll
ll
Karyotype
Fluorescence in situ hybridization
DNA microarray (see Chapter 20)
ll Comparative genomic hybridization
ll Single nucleotide polymorphism or oligonucleotide
arrays
Molecular analysis
The standard karyotype, or analysis of stretched and
stained chromosome preparations usually taken from a
peripheral blood sample, can often confirm a suggested
diagnosis or explain a set of major malformations not classically encountered together. Further description of specific
chromosomal abnormalities is addressed in Chapter 20; it
is sufficient to note that multiple malformation syndromes
can result from large visible chromosome rearrangements
that lead to deletion or addition of material (aneuploidy).
These rearrangements can involve an entire arm of a chromosome or can be submicroscopic, requiring further special testing. Such small deletions can often be detected by
fluorescence in situ hybridization analysis, which is performed using a probe specific for the deleted region.
It has become the standard of care in several centers to
offer more specialized molecular testing, such as a DNA
microarray or individual gene sequencing, as an adjunct
to or instead of karyotype analysis. In general, microarraybased methods are currently focused on detecting copy
number changes (smaller deletions or duplications not
detectable by a karyotype) and can be performed in a targeted or genome-wide fashion (Emanuel and Saitta, 2007).
As more information is made available about the role of
individual gene mutations in newborns with congenital malformations, testing for these mutations is becoming available in diagnostic laboratories. It can be useful
to check with a geneticist or genetics counselor for the
availability of gene mutation testing that may be clinically
available or performed on a research basis. GeneTests is an
internet database of laboratories worldwide that provides
such services.
DIAGNOSIS
There are cases in which, after a detailed examination,
exhaustive literature search, and genetic testing, no unifying diagnosis is evident. Aase (1990), a dysmorphologist,
advises, “Don’t panic! The absence of a diagnosis may be
distressing to the diagnostician and the family, but it is
much less dangerous than the possibility of assigning the
wrong diagnosis with the risk of erroneous genetic and
prognostic counseling and possibly hazardous treatment.”
Therefore, in cases in which there is no clear diagnosis,
prognosis and treatment should be determined according to the organ systems involved and the extent of their
impairment. In addition, when the infant has a severe,
untreatable impairment or the patient’s condition is critical, it may be prudent to offer and obtain consent for a full
postmortem examination by an experienced pathologist.
A skin sample, and sometimes blood, can be taken from
the expired fetus for establishing a cell line or for extracting DNA for future testing. Information gained from
such investigations may often become relevant for family
members, including the parents, allowing one to provide
accurate recurrence risk counseling and perhaps offer prenatal testing of a new pregnancy. Such information can
often help to provide closure for the family as well.
When should a genetics evaluation be considered? The
following clinical situations prompt a further genetic evaluation and counseling by a specialist:
ll
ll
ll
ll
ll
Multiple anatomic anomalies
History of maternal exposure to teratogens
Familial disorders
Increased carrier frequency or ethnic risk
Multiple pregnancy losses
As described previously, if a birth defect is identified in
the presenting patient or proband, especially if the defect
is associated with other anatomic anomalies, short stature,
or developmental delays, the features of a specific genetic
syndrome may be present. A known history of maternal
exposure to a potential teratogen would also be an indication for consultation. Conditions appearing to be familial, a
family history of hereditary disorders involving malformation of a major organ, or major physical differences such as
unusual body proportions, short stature, or irregular skin
pigmentation, would warrant genetic investigation. Mental retardation, blindness, hearing loss, or neurologic deterioration in multiple family members suggests a genetic
etiology. Likewise, a strong family history of cancer or a
defined ethnic risk such as Ashkenazi Jewish heritage and
CHAPTER 19 Evaluation of the Dysmorphic Infant
its association with a higher carrier frequency for TaySachs disease would be an indication for genetic evaluation. The occurrence of multiple pregnancy losses would
also raise the suspicion of a genetically influenced cause and
indicate the need for further investigation and counseling.
SUMMARY
Diseases with underlying genetic bases have significant
effects on health care and its delivery. An appreciation of
these entities, coupled with an organized, systematic evaluation, can help to define the nature of a given disorder
and aid in the development of the optimal plan of treatment and care for the patient.
SUGGESTED READINGS
Aase JM: Diagnostic dysmorphology, New York, 1990, Plenum.
Carey JC, editor: Elements of morphology: standard terminology, Am J Med Genet
149A:1-127, 2009.
Gehlerter TD, Collins FS, Ginsburg D, editors: Principles of medical genetics, ed 2,
Baltimore, Md, 1998, Williams & Wilkins.
195
GeneTests. Available online at www.ncbi.nlm.nih.gov/sites/GeneTests/?db=
GeneTests.
Hall JG, Allanson JE, Gripp KW, et al: Handbook of physical measurements, Oxford,
2007, Oxford University Press.
Hennekam RCM, Krantz ID, Allanson JE, editors: Gorlin’s syndromes of the head and
neck, ed 5, New York, 2010, Oxford University Press.
Jones KL: Smith’s recognizable patterns of human malformation, ed 6, Philadelphia,
2006, WB Saunders.
Nussbaum RL, McInnes RR, Willard HF, editors: Thompson and Thompson’s genetics
in medicine, ed 7, Philadelphia, 2007, WB Saunders.
Online Mendelian Inheritance in Man (OMIM). Available online at
www.ncbi.nlm.nih.gov/omim/.
Rimoin DL, Connor JM, Pyeritz RE, et al: Emery and Rimoin’s principles and practice
of medical genetics, ed 5, New York, 2006, Churchill Livingstone.
Schinzel A: Catalogue of unbalanced chromosome aberrations in man, Berlin, 2001,
Walter de Gruyter.
Spranger J, Benirschke K, Hall JG, et al: Errors of morphogenesis: concepts and
terms, J Pediatr 100:160-165, 1982.
Stevenson RE, Hall JG, editors: Human malformations and related anomalies, ed 2,
New York 2006, Oxford University Press.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
20
Specific Chromosome Disorders
in Newborns
Chad R. Haldeman-Englert, Sulagna C. Saitta, and Elaine H. Zackai
It has been estimated that 3% of newborns have a major
structural anomaly that will affect their quality of life.
Although most of these patients have a single malformation, 0.7% of infants have multiple major malformations.
In an additional 2%, a major anomaly is discovered by the
age of 5 years. Early identification of the genetic nature of
a given condition can aid in treatment and help to identify
resources for providing better health care to these individuals. This chapter will focus on genetic disorders and
syndromes with underlying chromosomal abnormalities
that typically manifest in the newborn period, in addition
to the shift in genetic evaluation and diagnosis with the
advent of newer array-based diagnostic techniques.
HUMAN KARYOTYPE
Chromosomes consist of tightly compacted DNA whose
structure is maintained by association with histones and
other proteins. When treated and stretched, chromosomes from dividing cells can be visualized under the light
microscope as linear structures with two arms joined by a
centromere. The short arm is designated p (petit) and the
long arm is designated q. The ends of the p and q arms are
known as telomeres. Human chromosomes were first visualized in 1956 (Lejeune and Turpin, 1960), and each pair
shows a distinctive size, centromeric position, and staining
or banding pattern after treatment with special dyes, allowing it to be identified and classified. Each chromosome is
identified by a number, in general from largest to smallest, in standard international cytogenetic nomenclature.
This presentation, or karyotype (Figure 20-1), normally
consists of 46 chromosomes, with 22 pairs of autosomes
and one set of sex chromosomes—two X chromosomes for
females (46,XX), and an X and a Y for males (46,XY).
Karyotype analysis is performed in cells undergoing
mitosis, or cell division, in which the chromosomes condense and can be stained and visualized. Thus, cells that
can be stimulated to divide and grow in culture, such as
peripheral blood lymphocytes, skin fibroblasts, and amniocytes, are typically used. Cells from bone marrow and chorionic villi are normally undergoing rapid cell division and
can also be karyotyped successfully. Historically, several
different staining methods have been described. However,
G-banding (Giemsa staining) is the standard cytogenetic
method used. This technique permits a resolution of at
least 400 bands among all the chromosomes and can be
adapted to allow for high-resolution analysis of up to 800
bands to analyze structural rearrangements as small as 5 to
10 million or megabase pairs (Mb).
Gamete formation, either spermatogenesis or oogenesis, is accomplished by a process known as meiosis. In the
first part of meiosis (meiosis I), homologous chromosomes
align as pairs and cross over, exchanging genetic material,
196
also known as recombination. In this stage, reduction division, the recombined pairs separate and the typical diploid content (46 chromosomes) of the cell is reduced by
half to a haploid complement of 23 chromosomes. In the
next stage, meiosis II, the chromosomes separate, similar
to mitosis (Nussbaum et al, 2007). The full diploid state of
the cell will be restored at the time of fertilization.
An imbalance of genetic material, or aneuploidy, occurs
from a net loss or gain of genetic material during sperm or
egg formation or, less commonly, during the initial divisions of the embryo. This missing or extra genetic material
can be small pieces or parts of chromosomes or an entire
chromosome itself. The classic recognizable aneuploidy
syndromes involve trisomy (three copies of a given chromosome) such as those of chromosomes 13, 18, and 21,
or monosomy (only a single copy) of a complete chromosome, such as X. Trisomies in particular can occur from
nondisjunction, a failure of normal chromosome separation. In such cases, a pair of homologues does not separate
in meiosis; one daughter cell receives both homologs of
that pair, and the other cell receives none. This event can
occur in either part of gamete division, meiosis I or meiosis II. Most human meiotic nondisjunction arises during
oocyte formation, specifically in maternal meiosis I. Nondisjunction of meiosis I is especially pronounced in trisomies of the acrocentric chromosomes (13, 14, 15, 21, and
22) and in XXX trisomy (MacDonald et al, 1994; Zaragoza et al, 1994). The occurrence of meiotic nondisjunction increases significantly with maternal age. Therefore
prenatal karyotyping from amniocentesis or chorionic villus sampling is offered to women aged 35 years and older
(Hook and Cross, 1982).
Nondisjunction can also occur in mitosis, with uneven
division of genetic material during early embryonic cell
division. This can result in two cell lines, one trisomic lineage that is potentially viable and one monosomic line. If
this event occurs after the first postzygotic division, cells
with a normal chromosome complement may also exist
with cells containing an aneuploid complement as a mosaic
chromosome constitution.
Partial aneuploidy may result from several mechanisms,
such as rearrangements of material between nonhomologous chromosomes that can occur in the gametes of a balanced translocation carrier. The carrier parent who has no
net loss or gain of genetic material is usually phenotypically
normal; however, the offspring are at increased risk for
an unbalanced rearrangement and its phenotypic consequences. Attention has also been focused on deletion syndromes caused by the loss of genetic material from several
chromosomes (e.g., 1p-, 4p-, 5p-), with a resulting, often
recognizable phenotype. Other microdeletions have been
associated with segmental duplications, or large blocks
of DNA that contain chromosome-specific repetitive
CHAPTER 20 Specific Chromosome Disorders in Newborns
197
FIGURE 20-1 G-banded human male karyotype. The 46 chromosomes are arranged into 23 pairs, each with a specific banding pattern.
sequences (Emanuel and Saikh, 2001). It is thought that
the repeats can mediate misalignment between two
homologs and have been shown to be present in regions
of the genome prone to instability, such as the pericentromeric regions of chromosomes 7q11, 15q11, 17q11,
and 22q11, leading to the phenotypes seen in WilliamsBeuren syndrome, Prader-Willi or Angelman syndrome,
Charcot-Marie-Tooth disease or hereditary neuropathy
with liability to pressure palsies, and DiGeorge or velocardiofacial syndrome, respectively (Emanuel and Saitta,
2007). The use of fluorescent in situ hybridization (FISH)
and chromosome-specific subtelomeric FISH and the
advent of DNA microarrays has allowed the identification
of smaller chromosomal rearrangements that were largely
unrecognized until recently. The use of high-resolution
microarrays in infants with multiple congenital anomalies
has in many cases led to the identification of a specific genotype, with clinical investigations then further defining the
associated phenotype (Bejjani and Shaffer, 2008).
TRISOMIES
DOWN SYNDROME (TRISOMY 21)
Lejeune and Turpin (1960) demonstrated that trisomy of
human chromosome 21 caused the constellation of findings recognized as Down syndrome (Figure 20-2). This
chromosome disorder was the first to be described and is
the most common viable autosomal trisomy, occurring in
approximately 1 in 700 to 800 live births (Hook, 1992).
The vast majority (>90%) occurs secondary to meiotic
nondisjunction, and a pronounced maternal age effect is
encountered. Approximately 3% to 5% of cases are caused
by a translocation, that could be either de novo or passed
from a balanced translocation-carrier parent, that becomes
unbalanced and trisomic in the baby. Typically, the translocated chromosome 21 rearranges with another acrocentric
chromosome, often chromosome 14, resulting in a Robertsonian translocation. Mitotic nondisjunction, or mosaic
Down syndrome, has been demonstrated in approximately
3% of cases as well, with variable features ranging from
normal to a typical Down syndrome phenotype.
Clinical Features
It is the more common occurrence of Down syndrome
in babies of older mothers that led to the use of prenatal karyotyping for advanced maternal age (>35 years) at
the time of conception, with samples typically obtained by
amniocentesis after 15 weeks’ gestation, or chorionic villus
sampling at 10 to 12 weeks’ gestation. Maternal serum analyte testing is used for prenatal screening purposes, with
results showing low alpha-fetoprotein, low unconjugated
estriol, and elevated total human chorionic gonadotropin.
198
PART V
Genetics
FIGURE 20-2 Newborn with Down syndrome (trisomy 21) illustrating some of the characteristic facial features, including upward-slanting
palpebral fissures and a flat facial profile.
Associated ultrasonographic findings for Down syndrome, including a cardiac defect, shortened long bones,
underdeveloped fetal nasal bone, nuchal translucency or
thickening, echogenic small bowel, and duodenal atresia
(“double-bubble” sign), may be seen in 50% to 60% of
fetuses. Most patients with Down syndrome, if not diagnosed prenatally, are usually recognized at birth because of
the typical phenotypic features, which then prompt karyotype analysis.
The constellation of physical findings associated with
Down syndrome consists of brachycephaly, presence of a
third fontanel, upward-slanted palpebral fissures, epicanthal folds, Brushfield spots in the irises, flattened nasal root,
small posteriorly rotated ears with over-folded superior
helices, prominent tongue, short neck with excess nuchal
skin, single palmar creases, brachydactyly, fifth-finger
clinodactyly, exaggerated gap between the first and second
toes, open field hallucal pattern, and hypotonia (see Figure
20-2). Often the physical features conform to an easily distinguishable phenotype, but in some cases, prematurity or
ethnic variations can make a clinical diagnosis less straightforward. An immediate karyotype is indicated to confirm
the diagnosis and as preparation for counseling the family.
Malformations involving many organ systems have been
described in Down syndrome, and whether the diagnosis
is known prenatally or determined in the newborn period,
several clinical investigations are warranted when this
diagnosis is suggested. The most common malformation
is congenital heart disease (seen in approximately 50% of
cases), which may require surgical intervention. Atrioventricular canal defects are often encountered (mean of 40%),
although ventricular septal defects (VSDs), atrial septal
defects (ASDs), tetralogy of Fallot, and patent ductus arteriosus (PDA) are all described in the disorder. An echocardiogram is indicated in all cases, and medical and surgical
interventions of cardiac lesions are routine. Gastrointestinal malformations, especially duodenal atresia (2% to 5%),
in addition to Hirschsprung disease and less frequently
encountered conditions, such as esophageal atresias, fistulas, and webs throughout the tract, have been described. It
is critical to carefully monitor the baby’s feeding and bowel
function before considering discharge from the nursery.
Although growth parameters can be in the range of 10%
to 25% at birth, significantly decreased postnatal growth
velocity is encountered in these patients. Separate growth
curves have been devised for patients with Down syndrome
(Fernandes et al, 2001), because growth retardation involving height, weight, and head circumference has been well
documented. An initial ophthalmologic evaluation is also
indicated in the first few months of life and then annually,
because strabismus, cataracts, myopia, and glaucoma have
been shown to be more common in children with Down
syndrome. In addition, hearing loss of heterogenous origin
is present in approximately half of patients, with middle
ear disease contributing to this problem.
Spinal cord compression caused by atlantoaxial subluxation from ligamentous laxity and subsequent neurologic
sequelae can be a complication of the disorder. Screening
radiographs are typically performed at approximately 3
years of age. Physicians should be vigilant in evaluating the
cervical spine, especially before administration of anesthesia and before an older child’s participation in sports. Other
associated disorders that merit screening are hypothyroidism in approximately 5% of patients, often with the presence of thyroid autoantibodies. Initial evaluation occurs
with newborn screening programs, followed by additional
thyroid-stimulating hormone and free thyroxine levels at 6
months, 12 months, and then yearly thereafter. Bone marrow dyscrasias, such as neonatal thrombocytopenia, and
transient self-resolving myeloproliferative disorders, such
as leukemoid reaction, have been observed in the first year
of life. An elevated rate of leukemia with a relative risk of 10
to 18 times normal up to age 16 years has been described.
Acute nonlymphoblastic leukemia is seen at higher rates in
congenital or newborn cases, but the distribution becomes
similar to that of non–Down syndrome patients after age 3
years. Survival of patients with Down syndrome is shorter
after a diagnosis of acute lymphoblastic leukemia than in
diploid patients (Epstein, 2001).
Patients with Down syndrome demonstrate a wide
range of developmental abilities, with highly variable personalities and behavioral phenotypes as well (Pueschel
et al, 1991). Central hypotonia with concomitant motor
delay is most pronounced in the first 3 years of life, as are
language delays. Therefore immediate and intensive early
intervention and developmental therapy are critical for
maximizing the developmental outcome. A wide range of
intelligence has been described, with conflicting data on
genetic and environmental modifiers of outcome (Epstein,
2001). Seizure disorders occur in 5% to 10% of patients,
often manifesting in infancy.
The most common causes of death in patients with
Down syndrome are related to congenital heart disease,
to infection (e.g., pneumonia) that is thought to be associated with defects in T-cell maturation and function, and
to malignancy (Fong and Brodeur, 1987). Once medical
and surgical interventions for the correction of associated
congenital malformations are complete and successful, the
long-term survival rate is good. However, fewer than half
of patients survive to 60 years, and fewer than 15% survive
CHAPTER 20 Specific Chromosome Disorders in Newborns
199
past 68 years. Neurodegenerative disease with features of
Alzheimer’s disease is encountered in most patients who
are older than 40 years, although frank dementia is not
typical. Men with Down syndrome are almost always
infertile, whereas small numbers of affected women have
reproduced (Epstein, 2001).
In counseling the family of a newborn diagnosed with
Down syndrome, it is important to include the organ systems affected in their baby and the severity of each malformation when defining a prognosis. Above all, the wide
variability of the phenotype should be emphasized, with
a care plan tailored to the needs of the individual patient.
Genetic Counseling
If a complete (full chromosome) or mosaic trisomy 21 is
found, parental karyotypes are generally not analyzed,
because the karyotypes are normal in virtually all cases.
After having one child with Down syndrome, a mother’s
recurrence risk for another affected child is approximately
1% higher than her age-specific risk (Hook, 1992). This
fact is especially significant in younger mothers, whose
age-specific risks are low. If a de novo translocation resulting in Down syndrome is found, the recurrence risk is less
than 1%. If the mother is found to carry a constitutional
balanced Robertsonian translocation, the risk for another
translocation Down syndrome fetus is approximately 15%
at the gestational age when amniocentesis is offered, and
10% at birth. However, if the father is the translocation
carrier, the recurrence risk is significantly smaller, approximately 1% to 2% (Epstein, 2001). Whereas newer arraybased diagnostic techniques will identify the copy number
change associated with the trisomy, structural rearrangements such as Robertsonian translocations are not readily detected. In this situation, a karyotype will provide
information regarding the mechanism of the copy number change, which is needed for accurate recurrence risk
counseling.
EDWARDS SYNDROME (TRISOMY 18)
Trisomy 18 is encountered in 1 in 6000 live births and is
associated with a high rate of intrauterine demise. It is estimated that only 5% of conceptuses with trisomy 18 survive
to birth and that 30% of fetuses diagnosed by second-
trimester amniocentesis die before the end of the pregnancy (Hook, 1992). Findings on prenatal ultrasonography
can raise suspicion for the disorder—growth retardation, oligohydramnios or polyhydramnios, heart defects,
myelomeningocele, clenched fists, and limb anomalies.
Maternal serum screening can show low values for alphafetoprotein, unconjugated estradiol, and total human chorionic gonadotropin, indicating the need for subsequent
karyotype analysis and fetal ultrasonographic monitoring.
Clinical Features
Phenotypic features present at birth consist of intrauterine growth restriction (1500 to 2500 g at term), small
narrow cranium with prominent occiput, open metopic
suture, low-set posteriorly rotated ears, and micrognathia with small mouth. Characteristic clenched hands with
FIGURE 20-3 Newborn with trisomy 18, showing prominent occiput,
characteristic facial appearance, and clenched hands.
overlapping digits, excess of arches on dermatoglyphic
examination, hypoplastic nails, and “rocker-bottom” feet
or prominent heels with convex soles (Figure 20-3) are
also described. Additional malformations encountered in
this syndrome are congenital heart disease (ASD, VSD,
PDA, pulmonic stenosis, aortic coarctation), cleft palate,
clubfoot deformity, renal malformations, brain anomalies,
choanal atresia, eye malformations, vertebral anomalies,
hypospadias, cryptorchidism, and limb defects, especially
of the radial rays.
The prognosis in this disorder is extremely poor, with
more than 90% of babies succumbing in the first 6 months
of life and only 5% alive at 1 year old. Death is caused
by central apnea, infection, and congestive heart failure.
The newborn period is characterized by poor feeding and
growth, typically requiring tube feedings. A few patients
have been described who have survived into childhood
and beyond. Universal poor growth and profound mental retardation with developmental progress stopping at
that of a 6-month-old infant (Baty et al, 1994) have been
documented. Malignant tumors such as hepatoblastoma
and Wilms’ tumor have been described in some of the survivors. In the few patients in whom cardiac surgery was
performed, outcome was not shown to be improved.
Genetic Counseling
The typical estimate of a recurrence risk for trisomy 18
in a future pregnancy is a 1% risk over the maternal age–
specific risk for any viable autosomal trisomy (Hook,
1992). Trisomy occurring from a structural rearrangement, such as a translocation, warrants parental karyotype
analysis before the recurrence risk can be assessed.
PATAU SYNDROME (TRISOMY 13)
It has been estimated that approximately 2% to 3% of
fetuses with trisomy 13 survive to birth, with a frequency
of 1 in 12,500 to 21,000 live births (Hook, 1992). As with
200
PART V
Genetics
also seen. The fingers can be flexed or overlapped and can
show camptodactyly. An increased frequency of nuclear
projections in neutrophils, giving a drumstick appearance
similar to that of Barr bodies, can also be found. This finding would be especially striking in males, in whom Barr
bodies would not be expected.
As with trisomy 18, prognosis for the fetus with trisomy
13 is extremely poor, with 80% mortality in the neonatal
period, and less than 5% of patients surviving to 6 months
old. Mental retardation is profound, and many patients are
blind and deaf as well. Feeding difficulties are typical.
Genetic Counseling
Recurrence risk data suggest that, as with trisomy 18, the
chance that a woman will have a child with any trisomy
after a pregnancy affected by trisomy 13 is rare. The estimated risk is 1% higher than the maternal age–related risk
for the recurrence of any viable autosomal trisomy in a
subsequent pregnancy.
TURNER SYNDROME (45,X)
FIGURE 20-4 Stillborn with trisomy 13. The facial appearance is
that of cebocephaly, which is associated with holoprosencephaly. There
is an extra digit on the ulnar border of the right hand.
other trisomies, amniocentesis performed for advanced
maternal age or indicated by fetal ultrasonographic findings may lead to a prenatal diagnosis of trisomy 13.
Clinical Features
Trisomy 13–associated malformations include congenital
heart disease, cleft palate, holoprosencephaly, renal anomalies, and postaxial polydactyly (Figure 20-4). In addition,
microcephaly, eye anomalies, and scalp defects can suggest
the diagnosis. Brain malformations such as holoprosencephaly are found in more than half the patients, with concomitant seizure disorders. Microcephaly, split sutures,
and open fontanels are encountered. A scalp defect (cutis
aplasia) that can sometimes be mistakenly attributed to a
fetal scalp monitor is specific to the disorder, being found
in 50% of cases. Eye malformations, including iris colobomas and hamartomatous cartilage “islands,” can be seen on
funduscopic examination.
Congenital heart disease is present in approximately
80% of patients, usually VSD, ASD, PDA, or dextrocardia. Limb anomalies, such as postaxial polydactyly, single
palmar creases, and hyperconvex narrow fingernails, are
In early embryogenesis, two active X chromosomes are
required for normal development. Turner syndrome, a
phenotype associated with loss of all or part of one copy
of the X chromosome in a female conceptus, occurs in
approximately 1 in 2500 female newborns. The 45,X
karyotype or loss of one entire X chromosome accounts
for approximately half of the cases. A variety of X chromosome anomalies—including deletions, isochromosomes, ring chromosomes, and translocations—account
for the remainder of the causes. It is important to note that
approximately 0.1% of fetuses with a 45,X complement
survive to term; the vast majority (>99%) is spontaneously
aborted. This fact underscores the requirement for both X
chromosomes during embryonic development. Additional
studies indicate that in approximately 80% of cases, it is
the paternally derived X chromosome that is lost (Willard,
2001).
Clinical Features
There is wide phenotypic variability in patients with
Turner syndrome. Features present at birth include short
stature, webbed neck, craniofacial differences (epicanthal
folds and high arched palate), hearing loss, shield chest,
renal anomalies, lymphedema of the hands and feet with
nail hypoplasia, and congenital heart disease. Typical cardiac defects are bicuspid aortic valve, coarctation of the
aorta, valvular aortic stenosis, and mitral valve prolapse.
Growth issues, especially short stature, are the predominant concern in childhood and adolescence; the mean adult
height of patients with Turner syndrome is 135 to 150 cm
without treatment. Growth hormone therapy, which is
routinely offered starting at approximately 4 to 5 years old,
can lead to an average gain of 6 cm or more in final adult
height (Willard, 2001). Primary ovarian failure caused by
gonadal dysplasia (streak gonads) can result in delay of
secondary sexual characteristics and primary amenorrhea.
Cyclic hormonal therapy is initiated at the age of puberty
to aid the development of secondary sex characteristics
CHAPTER 20 Specific Chromosome Disorders in Newborns
201
and menses as well as to help bone mass. Infertility, related
to gonadal dysplasia, is typical and has been successfully
treated with assisted reproduction techniques and donor
oocytes. It is important to evaluate for structural cardiovascular defects in the patient before pregnancy.
In terms of intellectual development, specific difficulties
with spatial and perceptual thinking lead to a lower performance intelligence quotient; however, this syndrome is
not characterized by mental retardation.
TRIPLOIDY (69,XXX OR 69,XXY)
As its name implies, triploidy is a karyotype containing
three copies of each chromosome. Mechanisms that lead
to this state include fertilization of the egg by two different
sperm (dispermy) and complete failure of normal chromosome separation in maternal meiosis. The vast majority of
triploid fetuses are spontaneously aborted, accounting for
up to 15% of chromosomally abnormal pregnancy losses.
Live births of affected fetuses are rare, and reports of survival beyond infancy are only anecdotal. Mosaicism with
combinations of diploid and triploid cells (mixoploid) has
also been documented. Malformations, including hydrocephalus, neural tube defects, ocular and auricular malformations, cardiac defects, and 3-4 syndactyly of the fingers,
are associated findings. In addition, the placenta is often
abnormal, typically large, and cystic.
DELETION SYNDROMES
In addition to the aneuploid conditions described previously, partial monosomy of a chromosome can lead to a
recognizable pattern of malformations. Three known
syndromes that are associated with the deletion or loss of
genetic material from the short, or p arms of chromosomes
1, 4, and 5 are described. All these syndromes are associated with deletions that involve the loss of many genes
located in a specific region.
CHROMOSOME 1p DELETION
SYNDROME (1p-)
Monosomy for the distal short arm of chromosome 1, or
deletion of 1p36, has been associated with a constellation of clinical findings. A characteristic facies consisting
of frontal bossing, large anterior fontanel, flattened midface with deep set eyes, and developmental delay has been
described (Figure 20-5). Orofacial clefting, hypotonia, seizures, deafness, and cardiomyopathy are also noted.
This deletion syndrome is estimated to occur in approximately 1 in 10,000 live births, and it is the most frequently
occurring subtelomeric deletion. Greater recognition
of the phenotype, the availability of a specific FISH test,
and current widespread use of DNA microarrays will
likely lead to improved diagnosis of this condition. The
majority of deletions arise de novo in the patient, with
approximately 3% being attributable to malsegregation of
a balanced parental translocation. The size of the deletion
varies, from submicroscopic (<5 Mb) to large, cytogenetically visible deletions greater than 30 Mb. A correlation
between the size of the deletion and the severity of clinical
features is suggested.
FIGURE 20-5 Child with deletion of chromosome 1p.
WOLF-HIRSCHHORN SYNDROME (4p-)
Distal deletions of the short arm of chromosome 4 are
associated with a recognizable pattern of malformation.
This syndrome is estimated to occur in 1 in 50,000 births
and has features such as intrauterine growth restriction,
microcephaly, midline structural defects such as cleft lip
and cleft palate, cardiac septal defects, and hypospadias.
The characteristic facial features are described as the
Greek helmet facies, as evidenced by hypertelorism with
epicanthi, a high forehead with a prominent glabella, and a
beaked nose. Prominent, low-set ears are also seen. Hypotonia, failure to thrive, and developmental delay are common, with one third of infants dying in the first year of life.
202
PART V
Genetics
Many patients have lived well into childhood and even into
adulthood, although profound growth and mental retardation is typical and often accompanied by seizures.
Although most deletions are cytogenetically visible on
karyotype analysis, small submicroscopic deletions have
also been described. In cases in which the clinical features
are suggestive but the karyotype is not revealing, further
cytogenetic analysis using specific 4p telomere probes or a
DNA microarray can be diagnostic. More than 80% of 4p
deletions arise de novo in the patient, with minimal risk of
recurrence. In the 10% to 15% of cases resulting from a
translocation, analyzing parental samples is clinically indicated for appropriate recurrence risk counseling.
CRI DU CHAT SYNDROME (5p-)
Partial monosomy of chromosome 5p is seen in approximately 1 in 50,000 live births and is associated with a multiple congenital anomaly syndrome named for the unusual
cry of the affected babies, described as similar to that of a
cat, or cri du chat. The constellation of features associated
with this disorder includes low birthweight, microcephaly,
round face, hypertelorism or telecanthus, downward-slanting palpebral fissures, epicanthi, and broad nasal bridge.
Hypotonia and cardiac defects are also seen, including
ASD, VSD, and tetralogy of Fallot. Early issues include
failure to thrive and pronounced developmental delay.
The unusual cry usually resolves during infancy, and survival into adulthood is possible but is typically associated
with severe mental retardation. Intensive therapy appears
to provide some benefit, and more sensitive measures of
cognition demonstrate clearly better receptive language
skills than expressive language ability. Therefore children
may understand more complex verbal language than their
expressive skills might demonstrate (Cornish et al, 1999).
It is estimated that almost 100 genes are lost when the
putative critical region from 5p15.2 to p15.33 is deleted
(Shaffer et al, 2001; Zhang et al, 2005). Close to 90% of
5p deletions arise de novo in the affected child, incurring
a minimal risk of recurrence (<1%). The remainder arise
from malsegregation of a balanced translocation in a carrier parent, which would be associated with a 10% to 15%
risk of recurrence of an unbalanced karyotype in a future
live born infant. Parental chromosome analysis is indicated
for proper recurrence risk counseling.
DNA MICROARRAYS
Recently, emphasis has been placed on characterizing
variations of the genome that fall in the range between
the single nucleotide and visible chromosomal changes—
submicroscopic structural variants (less than 5 Mb) that
cannot be seen by standard karyotype. An emerging technology rapidly gaining acceptance in various clinical settings involves the use of DNA microarrays (Bejjani and
Shaffer, 2008; Miller et al, 2010). In general, microarraybased methods are focused on detecting copy number
changes, and rearrangements such as balanced translocations or inversions are not detectable using this methodology. Microarray testing can be performed in either
a targeted or genome-wide fashion. Two types of DNA
microarray platforms are used currently: array comparative
genomic hybridization, which can utilize bacterial artificial chromosomes containing large DNA segments as
probes, or small oligonucleotides as DNA probes. Other
approaches use single nucleotide polymorphism arrays that
include probes based on known polymorphisms present
in the human genome. Single nucleotide polymorphism–
based arrays or dense oligonucleotide arrays have the
advantage of detecting gains or losses of shorter stretches
of the genome, because the probes for a given region can
be densely arrayed, and the sensitivity for detecting alterations of that region is greatly enhanced.
With a single test, microarrays can detect genomic
errors associated with disorders that are usually identified by cytogenetic analysis and multiple FISH studies.
Whereas microarray analysis is proving robust and providing an exceptional level of resolution from a diagnostic
perspective, the major difficulty with the current interpretation of the results lies in assigning causality and clinical
significance to the multiple alterations that are detected
in each individual. Toward this end, the availability of
databases with information on normal variation in multiple ethnic populations and testing of unaffected parents
remain standard approaches to discerning whether a copy
number change is likely to cause disease.
SEGMENTAL DUPLICATIONS AND
MICRODELETION SYNDROMES
A greater appreciation of the complexity of the human
genome and its structure has now been afforded with the
completion of the human genome sequence. This work
has focused attention on regions of the genome that are
prone to rearrangement. The presence of such unstable
regions appears to have a significant role in the etiology
of several genetic disorders (Emanuel and Shaikh, 2001;
Emanuel and Saitta, 2007). These disorders result from
inappropriate dosage of crucial genes in a given genomic
segment via either structural mechanisms (deletion or
duplication) or functional mechanisms (imprinting or
uniparental disomy). It has also been demonstrated that
many of these regions of genomic instability have a common element: the presence of large, chromosome-specific
low copy repeats that most likely mediate misalignment
and unequal crossover during recombination, leading to
rearrangements such as a deletion and duplication (Figure
20-6). Many of these large repeat structures are localized
to a single chromosome or within a single chromosomal
band. Examples of such genomic disorders are hemophilia
A (inversion of Xq28), Sotos syndrome (in which a number of patients have a deletion of 5q35), Smith-Magenis
syndrome (deletion of 17p11.2), Charcot-Marie-Tooth
disease (interstitial duplication on 17p12), and the reciprocal deletion of this same region of 17p12, leading to
hereditary neuropathy with liability to pressure palsies and
a small percentage of patients with neurofibromatosis type
I (deletion involving 17q11.2).
In this section, we focus on several deletion syndromes that occur on chromosomes whose underlying
genomic structure contains segmental duplications such
as Williams-Beuren syndrome (involving chromosome
7q11.2), Prader-Willi syndrome or Angelman syndrome
(involving an imprinted region of chromosome 15q11
CHAPTER 20 Specific Chromosome Disorders in Newborns
Normal Recombination Event
A
B
C
D
A
B
C
D
Crossover at LCR-D
A
B
C
D
A
B
C
D
A
Misalignment followed by Recombination
A
B
C
D
A
B
C
D
Crossover between A & D
A
B
A
B
C
DA
B
C
D
AD
Duplication on A
Deletion on B (a case of 22q11 Deletion Syndrome)
B
FIGURE 20-6 A, Alignment of low copy repeats (LCRs) before
exchange. B, Misalignment of LCRs before exchange can result in rearrangement.
through 15q13), and DiGeorge or velocardiofacial syndrome (DGS/VCFS), the most commonly occurring
microdeletion syndrome in humans, involving chromosome 22q11.2. It is important to note that in several of
these microdeletion syndromes, there is the possibility that a reciprocal duplication of the exact same region
may also occur (see Figure 20-6). Typically, the duplication syndromes cause fewer abnormalities and have wide
phenotypic variability, but they are often characterized by
developmental delays or behavioral abnormalities.
In addition, there are newly recognized microdeletion syndromes (e.g., 1q21.1, 3q29, 15q13.3, 16p11.2,
and 17q21) that are rapidly being identified because of
the increased use of DNA microarrays in the clinical and
research settings. These regions are similarly flanked
by segmental duplications, likely predisposing to their
rearrangements. Many of the patients with these newly
recognized rearrangements are not clinically diagnosed
in the newborn period, because they may not have significant anatomic malformations or dysmorphic features that prompt a genetic evaluation; however, they
can present during childhood with variable clinical
findings. Typically there is developmental delay, often
autistic spectrum disorder, or an intellectual disability.
These syndromes are complicated because they have
been reported in unaffected family members, which
proves to be a challenge when ascribing causality to the
rearrangement.
203
When an abnormality is identified by microarray analysis, the genes located in the affected region can be identified. It is then possible to decide what role, if any, the
affected genes have in the observed phenotype. For example, deleted genes could be identified that may predispose
a patient to cancer because of a germline loss of one copy
of a tumor suppressor gene (Adams et al, 2009), resulting in careful surveillance for tumor formation. Therefore
a new genetic paradigm is developing where patients are
initially genotyped (genotype-first), followed by evaluation
of the altered genes in a given region to determine their
possible effect on the future phenotype.
WILLIAMS-BEUREN SYNDROME
(7Q11.2 DELETION)
The estimated incidence of Williams-Beuren syndrome
is 10,000 live births. The phenotype has a variable spectrum, but usually consists of distinctive facies, growth
and developmental retardation, cardiovascular anomalies,
and occasionally infantile hypercalcemia (Figure 20-7).
Babies with Williams-Beuren syndrome usually show
some degree of intrauterine growth restriction with mild
microcephaly. Facial features include epicanthal folds
with periorbital fullness of subcutaneous tissues, flat midface, anteverted nostrils, long philtrum, thick lips, large
open mouth, and stellate irises that may not be discernible
at birth. Most infants have a cardiovascular abnormality;
supravalvular aortic stenosis (SVAS) is the most commonly associated defect, seen in more than 50% of cases.
Pulmonary artery stenosis is also often encountered. It is
interesting to note that isolated SVAS can also exist as a
separate autosomal dominant trait and has been shown to
occur from mutations of the elastin gene that is located
within the deletion region on 7q11.2. Patients with
Williams-Beuren syndrome are typically missing one
copy of the elastin gene.
Hypercalcemia, which is manifested in approximately
10% of patients with this disorder, is severe and persists
through infancy. Umbilical and inguinal hernias are also
associated features. Issues in infancy include feeding and
growth problems, with pronounced irritability and colicky
behavior. Hoarse voice, strabismus, hypertension, and
joint mobility restrictions may develop later in childhood.
In terms of development, the typical mild to moderate
mental retardation can be masked by advanced language
skills, although gross motor and visual-motor integration
skills are especially affected. Attention-deficit disorders are
common, and a characteristic outgoing personality is often
described in affected children.
Many of the classic features of Williams-Beuren syndrome are not clearly discernible in the newborn period,
but the diagnosis should be suggested in any child with
SVAS, hypercalcemia, and facial features consistent with
the disorder. The diagnosis can be confirmed quickly by
performing a DNA microarray or FISH using probes specific for the deleted region of chromosome 7q11.2. Because
the condition is typically sporadic and most deletions arise
de novo, the risk of recurrence in subsequent pregnancies
is minimal. An affected adult, however, would pass on the
condition in an autosomal dominant manner, with a 50%
risk of the disorder in his or her child.
204
PART V
Genetics
A
B
C
D
FIGURE 20-7 Williams syndrome. A, Neonate with a coarse face, periorbital fullness, wide mouth, and thick lips with decreased Cupid’s bow.
B, Neonate profile showing periorbital fullness, flat nasal bridge with full tip, and prominent cheeks. C, This infant has periorbital fullness, flat nasal
bridge, thick lips with decreased Cupid’s bow, pouty lower lip, and low-set, full cheeks. D, Infant profile showing dolichocephaly (increased anteroposterior diameter of head), a higher nasal bridge than in the neonate, full nasal tip, pouty lower lip, long neck, sloping shoulders, and part of pectus
excavatum.
22q11.2 DELETION SYNDROME
A deletion of chromosome 22q11.2 has been identified in
the majority of patients with DiGeorge syndrome, VCFS,
and conotruncal anomaly face syndrome, leading to the
realization that these clinical entities all reflect features
of the same genomic disorder (McDonald-McGinn et al,
1997). The list of findings associated with 22q11.2 deletion is extensive and varies by patient. Estimates indicate
that 22q11 deletion occurs in approximately 1 in 3000
live births (Burn and Goodship, 1996). This disorder is
the most common microdeletion syndrome occurring in
humans and is a significant health concern in the general
population.
The phenotype is characterized by a conotruncal cardiac
anomaly and often aplasia or hypoplasia of the thymus and
parathyroid glands. The majority of patients with a deletion can receive a diagnosis as newborns or infants with
significant cardiovascular malformations, including interrupted aortic arch type B, truncus arteriosus, or tetralogy of Fallot, along with functional T-cell abnormalities
and hypocalcemia. In addition, facial dysmorphia may be
present (Figure 20-8), including hooded eyelids, hypertelorism, overfolded ears, bulbous nasal tip, a small mouth,
and micrognathia. Since the initial report by DiGeorge in
1968, the spectrum of associated clinical features has been
expanded to include anomalies such as palatal anomalies,
CHAPTER 20 Specific Chromosome Disorders in Newborns
A
205
B
FIGURE 20-8 Facial differences associated with 22q deletion.
vascular rings, feeding and swallowing problems, gastroesophageal reflux, renal agenesis, and hypospadias
(McDonald-McGinn et al, 1997). Before advances in the
medical and surgical management of children with complex congenital cardiac disease and immune deficiencies,
this disorder was associated with significant morbidity and
mortality.
Developmental delays or learning disabilities have
been reported in most patients with 22q11.2 deletion,
and a wide range of developmental and behavioral findings have been observed in young children (Emanuel et
al, 2001). In the preschool years, children with a 22q11.2
deletion were most commonly found to be hypotonic and
developmentally delayed with language and speech difficulties. Severe or profound retardation was not seen,
and one third of patients functioned within the average
range.
The vast majority of patients (80% to 90%) have the
same large deletion, approximately 2.4 to 3 Mb, that is
detected by FISH (Figure 20-9) or DNA microarray.
The deletion remains unchanged when inherited from
an affected parent. However, the phenotype can be
widely variable, even within a family. Although smaller
recurrent deletions that are half the size of the common
deletion occur (1.5 Mb), a smaller size does not indicate
milder symptoms, making genotype-phenotype correlations difficult. To date, no explanation for the great
phenotypic variability has been forthcoming, and it is
currently an area of active investigation (Emanuel and
Shaikh, 2001).
Most 22q11 deletions occur as de novo events, with
less than 10% of them being inherited from an affected
parent. The prevalence of these de novo 22q11.2 deletions indicates an extremely high mutation or rearrangement rate within this genomic region that is probably
related to the presence of recombination-permissive
duplicated DNA sequences, segmental duplications,
or low copy chromosome 22–specific repeats in 22q11
FIGURE 20-9 (See also Color Plate 4.) Fluorescence in situ hybridization study of a 22q deletion. (Courtesy Beverly S. Emanuel.)
(Emanuel and Shaikh, 2001; Emanuel and Saitta, 2007;
Shaikh et al, 2007).
NEW MICRODELETION AND
MICRODUPLICATION SYNDROMES
As mentioned previously, several newly identified genetic
syndromes have been described with the increased use of
DNA microarrays (Slavotinek, 2008). These regions are
frequently flanked by segmental duplications, which is
the likely reason for their prevalence in diverse patient
populations, as well as the fact that there are reciprocal
206
PART V
Genetics
rearrangements (deletion or duplication). Almost all
of the patients with these deletions or duplications
were not initially recognized based on their clinical
features, but were instead ascertained by microarray
analysis.
Deletions of 1q21.1 of 1.35 Mb have been seen in
patients with variable presentations including developmental and behavioral abnormalities, mild facial dysmorphia, and microcephaly (Brunetti-Pierri et al, 2008;
Mefford et al, 2008). The 3q29 microdeletion syndrome
is approximately 1.6 Mb and was initially discovered
(Willatt et al, 2005) in patients with mild-to-moderate
mental retardation, microcephaly, and nonspecific facial
dysmorphia. Duplications of this region have also been
described in patients with developmental delays, mental retardation, and microcephaly (Lisi et al, 2008). The
15q13.3 region is distal to the more commonly known
15q11-q13 locus associated with Prader-Willi and Angelman syndromes. Patients with the 1.5-Mb deletion of
15q13.3 have variable phenotypes, but typically manifest
with seizures or abnormal electroencephalograms (Sharp
et al, 2008). A smaller 680-kb deletion has also been
described in a range of patients with neurobehavioral phenotypes (Shinawi et al, 2009). Interestingly, this deletion
is often maternally inherited, suggesting that there might
be an imprinting mechanism involved. A recurrent 550-kb
deletion of 16p11.2 has been discovered in approximately
1% of patients with autism (Weiss et al, 2008), and the
reciprocal duplication associated with attention deficithyperactivity disorder and schizophrenia (McCarthy et al,
2009; Shinawi et al, 2010). 17q21.31 deletions involving
approximately 500 to 650 kb were also found in patients
with developmental delays, learning disabilities, and variable facial dysmorphia (Koolen et al, 2008; Shaw-Smith
et al, 2006).
DISORDERS OF IMPRINTED
CHROMOSOMES
A growing recognition of mechanisms regulating gene
expression has emerged in the last decade. Two of the
most exciting concepts with important clinical correlates are imprinting and uniparental disomy. The term
genomic imprinting implies that a whole region of a chromosome or a group of genes in a given region are subject to a difference in their expression that depends on
whether they reside on the maternally inherited or paternally inherited chromosome. In these cases, a genetic
disorder might manifest according to the parent from
which the genomic region was inherited. The genes
in an imprinted region are not necessarily mutated,
but they are marked such that the cell can distinguish
between the maternal and paternal copies and coordinate expression on the basis of that distinction. At the
molecular level, it appears that differences in the methylation of the DNA, and its replication and regulation
at the transcriptional level, appear to be involved in
this mechanism. This mechanism has become an area
of important research advancements, and there are now
more than 100 known or predicted genes and chromosomal regions thought to be important in human disease
(www.geneimprint.com/site/genes-by-species).
PRADER-WILLI SYNDROME
It has been demonstrated that occasionally, instead of
inheriting one copy of each chromosome from each parent, both copies of a given chromosome can come from
the same parent. This phenomenon, known as uniparental disomy (UPD), is associated with advanced maternal
age. It becomes a significant issue when the involved
chromosome is imprinted or has regions on it that are
imprinted.
Prader-Willi syndrome (PWS) involves the loss of
activity from the paternally derived proximal long arm
of chromosome 15 (15q11). This loss can occur through
deletion or disruption of this region or through maternal
uniparental disomy such that no paternal chromosome
15 is present (Nicholls et al, 1989). Newborns have pronounced central hypotonia, hyporeflexia, and a weak cry.
The poor tone manifests as sucking and swallowing difficulties that can lead to failure to thrive and the need for
feeding tubes in infancy. Facial differences that have been
described include bifrontal narrowing, almond-shaped
eyes, and a small, downturned mouth. Genitalia are often
hypoplastic, with cryptorchidism being common in boys
with this syndrome. The commonly reported small hands
and feet are not usually demonstrated in the newborn.
Strabismus and hypopigmentation relative to the family
are also common.
A history of poor fetal activity during the pregnancy can
often be elicited, especially if the mother has had prior
pregnancies. Consistent with the hypotonia, breech presentation and perinatal insults are found more frequently
than usual. The extreme hypotonia begins to improve in
the first year of life, as does motor development, although
developmental delay is the rule, especially for gross motor
skills and speech. The feeding improves in the first few
years of life and gives way to often uncontrollable hyperphagia and obesity. This issue and other behavioral
problems, including severe temper tantrums and obsessive-compulsive disorder, are encountered throughout
life. The majority of patients manifests mild to moderate
mental retardation. Early diagnosis and preemptive implementation of behavioral therapy are essential components
of the optimal management of these issues.
Deletions of the region critical in Prader-Willi syndrome have been demonstrated in up to 70% of patients.
The deletion can be detected by DNA microarray or FISH
analysis using a probe specific for this region of chromosome 15. A small number of patients have a disruption of
this area as the result of a chromosomal translocation. To
date, no single gene in this region has been implicated
as the cause, but expression of the genes in this region is
under intense investigation. It has been noted, however,
that patients who have Prader-Willi syndrome as a result
of a deletion of the region are more likely to be hypopigmented. This feature has been attributed to deletion of
a gene involved in pigmentation, the so-called P gene
(Rinchik et al, 1993). Recurrence risks are negligible in
cases in which de novo deletions are found and sporadic
occurrence is usually encountered.
Approximately 20% to 25% of patients with PraderWilli syndrome show maternal UPD that can be detected
by means of a molecular assay designed to assess specific
CHAPTER 20 Specific Chromosome Disorders in Newborns
207
FIGURE 20-10 Macrosomic infant with macroglossia and lax abdominal musculature. These findings
are typical of Beckwith-Wiedemann syndrome. (From
Viljoen DL, Jaquire Z, Woods DL: Prenatal diagnosis in
autosomal dominant Beckwith-Wiedemann syndrome, Prenat
Diagn 11:167-175, 1991.)
methylation differences between maternal and paternal alleles. Methylation analysis is abnormal in more
than 99% of affected individuals, but will not determine
whether the cause is attributed to a deletion or maternal
UPD. Further study is required if an abnormal result is
obtained. A maternal age effect has been demonstrated in
UPD cases, and recurrence risks in families without deletions are estimated at 1 in 1000. In addition, this region of
the genome is subject to regulation by imprinting. Large
chromosome-specific segmental duplications are found in
15q11 and have been implicated in mediating the recurrent deletion of this genomic region (Emanuel and Shaikh,
2001; Emanuel and Saitta, 2007).
ANGELMAN SYNDROME
Loss of genetic material from the 15q11 region from
the maternal copy of chromosome 15 is associated with
Angelman syndrome. Clinical features are not evident in
the newborn period and infancy, but include significant
mental retardation, seizures, ataxic gait, tongue thrusting, inappropriate bursts of laughter, and facial differences including protruding jaw, wide mouth, thin upper
lip, and widely spaced teeth. The mental retardation and
hypopigmentation overlap with the features of PraderWilli syndrome, but Angelman syndrome is a distinct
entity.
Seventy percent to 75% of patients have a deletion of
15q11 that is detectable by FISH analysis. A small percentage (3% to 5%) have evidence of paternal isodisomy
(two paternal copies) of the entire chromosome 15, with
no apparent maternal chromosome. Unlike in PraderWilli syndrome, Angelman syndrome has been associated with mutations in a single gene, UBE3A—an enzyme
involved in the ubiquitin pathway of protein degradation,
detected in up to 10% of patients. In addition, mutations
of an imprinting center locus on chromosome 15 are
thought to be associated with 1% to 2% of Angelman phenotypes (Shaffer et al, 2001). Methylation analysis can be
performed and will detect abnormalities in approximately
75% to 80% of patients, because of a deletion or UPD.
If methylation analysis is normal but Angelman syndrome
is still suspected, UBE3A sequence analysis should be
considered. The vast majority of cases result from a sporadic event, and the risk of recurrence can be best evaluated once the genetic mechanism has been determined for
a given patient.
BECKWITH-WIEDEMANN SYNDROME
Beckwith-Wiedemann syndrome affects approximately 1
in 14,000 newborns and manifests as an overgrowth syndrome in the neonatal period. The characteristic findings
are macrosomia, abdominal wall defect, and macroglossia
(Figure 20-10). Affected babies are large for gestational age
with proportionate length and weight. Infants of mothers
with diabetes also manifest with macrosomia, but are more
likely to have a weight disproportionately greater than
length. Advanced bone age is also noted in Beckwith-Wiedemann syndrome. Hemihypertrophy caused by asymmetric growth is common, as is visceromegaly of various
organs, including the spleen, kidneys, liver, pancreas, and
adrenal glands.
Other characteristic features of the syndrome are macroglossia, linear creases of the earlobe with indentations
on the posterior helix, and severe hypoglycemia. Although
the hypoglycemia responds quickly to therapy, it can be
present for several months; therefore recognition of the
condition and immediate therapeutic intervention are critical in these cases. The hypoglycemia resolves spontaneously with age, and the physical diagnostic features also
become less prominent with age, making the diagnosis
more difficult to ascertain.
Equally important is the establishment of routine ultrasonographic surveillance at regular intervals, because
children with Beckwith-Wiedemann syndrome are at
increased risk of malignant tumors, especially Wilms’
tumor. The estimated risk is as high as 8% for patients with
hemihypertrophy. Many centers currently perform ultrasonography at 3-month intervals until the school-age years
(approximately 8 years old). Monitoring of serum alphafetoprotein levels at the same intervals until age 4 years has
proved valuable, as several cases of hepatoblastoma have
also been reported and detected with this adjunct study.
Although most cases of Beckwith-Wiedemann appear to
arise de novo, up to 15% may be familial. In familial cases,
208
PART V
Genetics
the transmission is autosomal dominant, because of mutations of the CDKN1C gene in 40% of familial cases, but
only 5% to 10% of de novo cases. In addition, this region
of the genome (11p15.5) appears to be imprinted such that
the maternal allele is not usually expressed. The insulinlike growth factor type 2 (IGF2) gene is located in this
region and encodes an important factor involved in fetal
growth. Mutations causing overexpression of the paternal
allele or underexpression of the maternal allele can result
in an imbalance of expression leading to the overgrowth
and tumor formation encountered in these patients. Paternal UPD has proved to be a mechanism involved in 10%
to 20% of sporadic cases of Beckwith-Wiedemann syndrome. A method for detecting methylation abnormalities at two distinct genetic loci within 11p15 accounts for
approximately 60% of patients and is related to the overexpression of the IGF2 gene. Therefore all the available
testing methods combined can detect the cause in approximately 85% of the patients with Beckwith-Weidemann
syndrome. The recurrence risk for future affected siblings
or offspring of the proband depends on the specific genetic
abnormality causing the disorder and can range from
low (UPD, methylation abnormality) to as high as 50%
(CDKN1C mutation).
RUSSELL-SILVER SYNDROME
Russell-Silver syndrome presents in neonates with intrauterine growth retardation followed by postnatal growth
deficiency. The head size is usually normal, causing a
relative macrocephaly that may have the appearance of
hydrocephalus. Facial features can include a broad and
prominent forehead, triangular-shaped face with a small
chin, and downturned corners of the mouth. The fingers
can show brachydactyly, camptodactyly, or more commonly fifth-finger clinodactyly. Other concerns involve
limb-length discrepancy, delayed bone age, café au lait
macules, hypospadias in males, developmental delays, diaphoresis, and hypoglycemia during the first 3 years of life.
These patients are often examined by a geneticist as a toddler with growth retardation, proportionate short stature,
and normal head circumference. When evaluating patients
with growth retardation, it becomes important to know
the prenatal and postnatal growth parameters, because
they might provide a clue to the diagnosis of Russell-Silver
syndrome.
The molecular mechanisms underlying the pathogenesis show that Russell-Silver syndrome is likely caused
by abnormalities of imprinted genes. Maternal UPD of
chromosome 7 is present in 7% to 10% of patients, and
the symptoms are likely caused by overexpression of the
maternal GRB10 gene that suppresses activity of various growth factor receptors. In approximately 35% of
the patients, imprinting abnormalities of 11p15.5 occur
because of a loss of the paternally expressed IGF2 gene,
leading to decreased prenatal and postnatal growth. This
finding contrasts with some patients with BeckwithWiedemann syndrome, in whom IGF2 is overexpressed
causing increased growth. To that end, patients with Russell-Silver syndrome do not have a significantly increased
risk for neoplasia compared to patients with BeckwithWiedemann syndrome, and routine cancer surveillance
protocols are typically not recommended.
SUMMARY
This chapter has summarized the rapidly expanding field
of chromosomal and genomic disorders, concentrating
on those that commonly manifest in the newborn. The
widespread development and clinical implementation of
molecular cytogenetic techniques have allowed the identification of subtle rearrangements that were previously
undetectable. These advances now enable the discernment
of new syndromes in which the chromosomal anomaly
may be defined before a characteristic phenotype is recognized. In addition, a greater understanding of the role of
segmental duplications and their effects on human genetic
disorders as well as of the influence of mechanisms that
regulate gene expression, such as imprinting, is emerging.
The tremendous advances in genomics led by the completion of the Human Genome Project and the development
of new molecular diagnostic tools present new challenges
for clinicians to better diagnose, understand, and care for
patients with genetic disorders and their families.
SUGGESTED READINGS
GeneReviews. Available on line at http://www.ncbi.nlm.nih.gov/sites/GeneTests/?
db=GeneTests
Jones KL: Smith’s Recognizable Patterns of Human Malformation, ed 6, Philadelphia,
PA, 2006, WB Saunders.
Online Mendelian Inheritance in Man (OMIM): Available on line at http://www.
ncbi.nlm.nih.gov/omim/
Rimoin DL, Connor JM, Pyeritz RE, Korf BR, editors: Emery and Rimoin’s
Principles and Practice of Medical Genetics, ed 5, New York, NY, 2006, Churchill
Livingstone.
Schinzel A: Catalogue of Unbalanced Chromosome Aberrations in Man, Berlin, 2001,
Walter de Gruyter.
Shaffer LG, Slovak ML, Campbell LJ, editors: An International System for Human
Cytogenetic Nomenclature (ISCN) 2009: Recommendations of the International
Standing Committee on Human Cytogenetic Nomenclature, Basel, 2009, S Karger.
Slavotinek AM: Novel microdeletion syndromes detected by chromosome microarrays, Hum Genet 124:1-17, 2008.
Complete references used in this text can be found online at www.expertconsult.com
P A R T
V I
Metabolic and Endocrine Disorders of the Newborn
C H A P T E R
21
Introduction to Metabolic and
Biochemical Genetic Disease
Stephen Cederbaum
Inborn errors of metabolism or biochemical genetic disorders are one type of genetic disease that may be encountered
in the neonatal period as an acute or more indolent illness.
In these disorders, a mutation in a gene leads to an absent
or defective gene product or enzyme and results in the accumulation of the precursor of the enzyme or a byproduct of
it, a shortage of the product of the enzymatic reaction, or a
combination of both. In reality the effects of many inborn
errors on normal physiology are much more profound, causing many changes in gene expression and normal biochemical function. One example is a case of propionic acidemia
in which interference with the mitochondrial respiratory
apparatus is rarely measured except by the ascertainment
of an elevated blood lactate level, but which may be much
more frequent and represent only the most obvious and easily measured alteration.
Inborn errors of metabolism may be inherited by any
genetic mechanism—autosomal dominant, autosomal
recessive, sex-linked recessive, or through a mutation in the
independently inherited mitochondrial genome (mtDNA),
which leads to a circumstance in which the mother alone
passes the abnormal DNA to all of her children, but the
affected or carrier father to none of his offspring.
Most inborn errors are inherited as autosomal recessive
conditions, with the carrier parents rarely expressing any
obvious metabolic phenotype. A small minority is inherited
in a sex-linked recessive or codominant manner, and they
will be discussed in the context of their particular disease.
Examples would be ornithine transcarbamylase deficiency (a
urea cycle disorder) and Fabry disease (a lysosomal storage
disorder), the latter not appearing in the neonatal period.
When viewed from the perspective of disease mechanism, most genetic disorders, whether single gene or
involving imbalance of chromosomal materials, could be
considered to be inborn errors of metabolism. One or more
changes in the DNA result in either altered gene expression
or expression of a mutated gene, which then lead secondary to an altered product of the reaction or reactions. We
will not use this expansive and grandiose interpretation of
inborn errors, but rather confine ourselves to the more traditional definition described in the first paragraph. Thus
disorders such as cystic fibrosis and spinal muscle atrophy,
considered inborn errors that require broader interpretation, will not be considered.
The advent of newborn screening in the early part of
the 1960s for phenylketonuria (PKU) established a new
paradigm for approaching inborn errors of metabolism and
making the diagnosis and treatment prior to the symptomatic presentation, and hence preventing rather than treating
the condition. This approach has proved to be remarkably
successful for PKU and congenital hypothyroidism, with
few patients becoming mentally retarded. In subsequent
years the menu of tests expanded gradually, but it has
greatly expanded in the last decade with the implementation of expanded newborn screening using tandem mass
spectrometry (MS/MS) technology, which allows for ascertainment of a constellation of disorders and should alter
the probability of, and the diagnostic testing for, inborn
errors when incorporated into the diagnostic algorithms of
the ill newborn. Moreover, the advancing technology permits some of the newborn dried blood spot to be used for a
wider palette of tests, some of which may already be available. The consequences of this expansion have a downside
for the neonatologist and the neonatal intensive care unit
staff members. For a variety of reasons the sick and premature newborn, usually receiving intravenous alimentation
and having immature or damaged organs, is far more likely
to have a false-positive newborn screening test and require
follow-up testing. It is important to recognize that a number of traditionally tested diseases are outside of this class
of disorders, such as hypothyroidism, congenital adrenal
hyperplasia, cystic fibrosis, and hemoglobinopathies.
When considering inborn errors of metabolism, it is
important to consider the molecular basis of mutation.
Large deletions of a gene are certain to eliminate enzymatic
function and any residual gene product. Smaller deletions,
especially if they remove one or more of the in-phase coding triplets, may permit a stable protein to be made and to
function to some extent. Most small deletions, however,
cause the synthesis of unstable and out-of-phase proteins
that do not function and have a short half-life within the
cell. Some single-based change mutations can introduce
a stop codon, causing the synthesis of the polypeptide to
halt abruptly and leave a nonfunctional enzyme. Singlebase changes introducing a new amino acid vary in their
effects from complete loss of activity to a lesser impact,
and finally to having no effect whatsoever. When considering that the mutation test is then all modulated through
the unique genetic background of the individual, there is
no single final phenotype, severity, or time of onset for any
genetic disorder. This variation must be considered when
any diagnosis, genetic or not genetic, is considered.
209
210
PART VI Metabolic and Endocrine Disorders of the Newborn
CLASSIFICATION OF INBORN ERRORS
OF METABOLISM
Each professional uses classification systems to permit effective reasoning as to possible causes of a symptom complex.
A system for understanding inborn errors of metabolism
is shown in Box 21-1. Each group has common characteristics, modes of presentation, types of molecules involved,
and tests that would be applied. Because the demarcation
between the groups is not sharp, other systems can see
them differently. This discussion is restricted to those disorders that may be symptomatic in the newborn period or
in early infancy, whereas many severe disorders would be
unlikely to be associated with neonatal disease and will be
given less emphasis. The disorders more commonly seen
in the neonatal period are listed in Box 21-2.
The first group consists of newborns with progressive
lethargy, poor suck, neurologic deterioration, and often
death. They have inborn errors of amino acids, the urea
cycle, organic acids, or sugar metabolism. This group of
BOX 21-1 C
lassification of Inborn Errors
of Metabolism, 2007
Small molecule disorders
ll Amino acids
ll Organic acids
ll Sugars
Lysosomal storage disorders
ll Mucopolysaccharides
ll Sphingolipids
ll glycolipids
Energy metabolism disorders
ll Oxidation disorders
ll Fatty acid mobilization and metabolism disorders
ll Glycogen storage diseases
Peroxisomal and membrane biogenesis disorders
Carbohydrate-deficient glycoprotein disorders
Cholesterol biosynthetic disorders
Disorders of biogenic amines, folate, and pyridoxine
Transport disorders
Purine and pyrimidine metabolism disorders
Receptor disorders
patients is the product of normal pregnancies and deliveries and becomes ill after 36 hours of life, when the maternal circulation no longer cleanses the accumulating small
molecules from the fetal or infant blood and the offending
metabolites accumulate in intoxicating amounts (Box 21-3).
Examples include maple syrup urine disease, methylmalonic and propionic acidemias, galactosemia, and ornithine
transcarbamylase deficiency. The general characteristics are
given in Box 21-4; they are the disorders for which expanded
newborn screening may lead to earlier detection and a more
rapid diagnosis. These patients’ condition is most likely to
resemble sepsis, and they should be treated with antibiotics.
Most disorders manifesting acutely in the newborn period
will be detected by the newborn screen, with only some urea
cycle disorders and lactic acidoses likely to be missed by
this screening panel. When diagnosed, these conditions are
BOX 21-3 M
etabolic Diseases With
Newborn Coma Secondary to
Toxic Metabolite Accumulation
or Mitochondrial Failure
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
BOX 21-4 C
haracteristics of Small Molecule
Disorders
High levels of metabolites in body fluids
Normal physical phenotype
ll Neonatal presentation
ll Periods of stability and instability
ll Considered to be intoxication disorders
ll Can often be treated by external manipulation
Lysosomal storage disorders (format)
ll Usually born normally
ll Course is progressive, relentless, and indolent
ll Deposition of material seen clinically and microscopically
ll May be deforming
ll Cannot be addressed exogenously
Disorders of energy metabolism
ll Mixed presentation between the first two categories
ll Can be catastrophic at presentation
ll May be present at birth or develop later
ll Usually progressive
ll May cause malformations
ll May have episodes of deterioration
ll Usually not treatable by exogenous means
ll May be tissue specific or preferential
ll
ll
BOX 21-2 C
ommon Types of Inborn Errors
of Metabolism With Newborn
Presentation
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Amino acid disorders
Organic acid disorders
Disorders of ammonia metabolism
Disorders of carbohydrate metabolism
Disorders of gluconeogenesis or hypoglycemia
Disorders of fatty acid oxidation
Primary lactic acidoses (respiratory chain defects)
Disorders of vitamin or metal metabolism
Storage diseases (infrequently)
Peroxisomal disorders
Disorders of sterol metabolism
Congenital defects in glycosylation
Galactosemia
Inborn errors of ammonia metabolism
Maple syrup urine disease
Nonketotic hyperglycinemia
Methylmalonic acidemia with or without homocystinuria
Propionic acidemia
Isovaleric acidemia
Multiple carboxylase deficiency
Glutaric aciduria type 2
Fatty acid oxidation defects
Primary lactic acidosis
Pyruvate dehydrogenase deficiency
Pyruvate carboxylase deficiency
Mitochondrial respiratory chain or electron transport chain defects
CHAPTER 21 Introduction to Metabolic and Biochemical Genetic Disease
treated with dialysis, limitation of protein intake except for
galactosemia, fluid, and caloric support, and some specific
interventions. The association of identifying physical and
laboratory characteristics and various disorders is listed in
Tables 21-1 and 21-2. The individual disorders are discussed
in Chapters 22 and 23. When a diagnosis of a metabolic
disorder appears likely, plasma amino acids, urine organic
acids, plasma acylcarnitine, and plasma carnitine tests should
be repeated, and ammonia and lactate should be determined.
The second major category of inborn errors is the lysosomal storage diseases. This group of disorders results from
defective function of a catabolic hydrolase located in the lysosome that is generally responsible for breaking down complex glycosaminoglycans and sphingolipids that are products
of normal cellular turnover (see Box 21-4). Unlike the small
molecule disorders in which the metabolites are found freely
circulating in the body fluid compartments, these compounds
211
accumulate intracellularly, are not removed by the maternal
circulation, and are present in limited amounts in the body
fluids. They most often cause no apparent symptoms in the
newborn period or early infancy, because the pathologic
metabolites accumulate slowly with time. Exceptions to this
finding are the severe form of α-glucosidase deficiency or
Pompe disease, the neonatal form of α-galactosidase deficiency, or Krabbe disease and galactosialidosis. These findings are discussed in Chapter 23, and some are listed in
Table 21-1. The disorders of mucopolysaccharides and glycolipids lead to the characteristic features pejoratively and
inappropriately referred to as gargoylism, which consist of
an exaggerated eyebrow, coarse-appearing facies, thick skin,
hirsutism, and multiple abnormalities of the bones and joints
seen on a radiograph. Attention to the disorder is often
drawn by the hepatosplenomegaly. The metabolites are synthesized in the body and are not influenced by dietary intake.
TABLE 21-1 Unique or Characteristic Physical Findings in Inborn Errors* (Major Examples)
Finding
Error
Finding
Error
Hepatomegaly
Galactosemia
Glycogen storage diseases
Gluconeogenic defects
Disorders of fatty acid oxidation and
transport
Mitochondrial respiratory or electron
transport chain defects
Hereditary tyrosinemia type 1
Urea cycle defects
Peroxisomal defects
Niemann-Pick disease type C
Congenital defects in glycosylation
Retinitis pigmentosa
Mitochondrial respiratory or electron transport chain defects
Sjögren-Larsson syndrome
Peroxisomal disorders
Abetalipoproteinemia
Optic atrophy or
hypoplasia
Pyruvate dehydrogenase complex deficiency
Mitochondrial disorders
Leigh disease
Peroxisomal disorders
Corneal clouding or
opacities
Mucolipidoses
Mucopolysaccharidoses
Steroid sulfatase deficiency
Cataracts
Galactosemia
Lowe syndrome
Mitochondrial respiratory or electron transport chain defects
Peroxisomal disorders
Congenital defects in glycosylation
Dislocated lens
Methionine synthetase deficiency
Sulfite oxidase deficiency
Bone or limb
deformities or
contractures
Storage, peroxisomal, or connective tissue
disorders
Inborn errors of cholesterol biosynthesis
Thick skin
Mucolipidoses
Gangliosidoses
Mucopolysaccharidoses
Desquamating,
eczematous, or
vesiculobullous
skin lesions
Acrodermatitis enteropathica
Organic acidemias
Early-onset forms of porphyria
Ichthyosis
Gaucher disease type 2
Steroid sulfatase deficiency
Alopecia
Multiple carboxylase deficiency
Steely or kinky hair
Menkes disease
Persistent diarrhea
Glucose galactose malabsorption
Congenital lactase deficiency
Congenital chloride diarrhea
Sucrase isomaltase deficiency
Acrodermatitis enteropathica
Congenital folate malabsorption
Wolman disease
Galactosemia
Hepatosplenomegaly
Gangliosidoses
Niemann-Pick disease type C
Mucopolysaccharidoses
Wolman disease
Ceramidase deficiency
Macrocephaly
Glutaric acidemia type 1
Canavan disease
Microcephaly
Mitochondrial respiratory or electron
transport chain defects
Leigh disease
Methylmalonic acidemia with
homocystinuria
Coarse facial features
Macroglossia
Dystonia or extrapyramidal signs
Macular “cherry red
spot”
*For
Gangliosidosis
Mucolipidoses
Mucopolysaccharidosis type VII
Sialidosis
Galactosialidosis
Pompe disease
Gangliosidoses
Mucopolysaccharidoses
Mucolipidoses
Gaucher disease type 2
Glutaric acidemia type 1
Krabbe disease
Crigler-Najjar syndrome
Biopterin defects
GM1 gangliosidosis
Galactosialidosis
Niemann-Pick disease type A
Tay-Sachs disease (GM2
gangliosidosis)
discussion of specific disorders, see Chapters 22 and 23.
212
PART VI Metabolic and Endocrine Disorders of the Newborn
TABLE 21-2 Characteristic or Unique Laboratory or Diagnostic Testing Outcomes in Inborn Errors (Major Examples)
Outcome
Error
Outcome
Error
Metabolic acidosis
with or without
increased anion
gap
Organic acidemias
Maple syrup urine disease
Fatty acid oxidation defects
Ketothiolase deficiency
Ketogenesis defects
Disorders of pyruvate metabolism
Mitochondrial respirator or electron transport chain defects, including Leigh disease
Galactosemia
Glycogen storage disease type 1
Gluconeogenesis defects
Thrombocytopenia
Organic acidemias
Pearson syndrome
Anemia
Organic acid disorders
Wolman disease
Pearson syndrome
Severe liver failure
Galactosemia
Vacuolated
lymphocytes or
neutrophils
Lysosomal storage disorders
Respiratory alkalosis
Urea cycle disorders
Cardiomegaly
Hyperammonemia
Urea cycle disorders
Methylmalonic acidemia
Organic acidemias
Fatty acid oxidation disorders
Ketosis
Organic acidemias
Maple syrup urine disease
Glutaric acidemia type 2
Ketogenesis defects
Glycogen storage disease type 1
Gluconeogenesis disorders
Pompe disease
Barth syndrome
Fatty acid oxidation defects
Mitochondrial respiratory or electron
transport chain defects
Carbohydrate-deficient glycoprotein
syndrome
Electrocardiographic abnormalities
Pompe disease (short PR interval, large QRS
interval)
Fatty acid oxidation disorders
Mitochondrial respiratory or electron
transport chain defects
Lactic acidosis
Mitochondrial respiratory or electron transport chain defects, including Leigh disease
Pyruvate dehydrogenase complex deficiency
Pyruvate carboxylase deficiency
Organic acidemias
Glutaric acidemia type 2
Fatty acid oxidation defects
Ketogenesis defects
Glycogen storage disease type 1
Gluconeogenesis disorders
Ventricular hypertrophy
Pompe disease
Organic acidemias
Glutaric acidemia type 2
Fatty acid oxidation defects
Mitochondrial respiratory or electron
transport chain defects, including Leigh
disease
Dysostosis multiplex
Gangliosidoses
Mucopolysaccharidoses
Mucolipidoses
Sialidosis
Stippled calcifications of patellae
Peroxisomal disorders
Cholesterol biosynthetic defects
Adrenal calcifications
Wolman disease
Rhizomelica
Rhizomelic chondrodysplasia punctata
Hair abnormalities
Menkes disease
Argininosuccinicaciduria
Basal ganglia lesions
on MRI
Organic acidemias (later in life)
Pyruvate dehydrogenase complex deficiency
Mitochondrial respiratory or electron
transport chain defects, including Leigh
disease
Cerebellar atrophy
or hypoplasia
Pyruvate dehydrogenase complex deficiency
Mitochondrial respiratory or electron transport chain defects, including Leigh disease
Carbohydrate-deficient glycoprotein syndrome
Agenesis of corpus
callosum
Pyruvate dehydrogenase complex deficiency
Pyruvate carboxylase deficiency
Mitochondrial respiratory or electron transport chain defects
Hypoglycemia
Hyperinsulinism
Glycogen storage disease type 1
Gluconeogenesis disorders
Maple syrup urine disease
Glutaric acidemias
Fatty acid oxidation defects
Ketogenesis defects
Galactosemia
Severe liver failure
Mitochondrial respiratory or electron transport chain defects
Lipemia
Glycogen storage disease type 1
Lipoprotein lipase deficiency
Positive urinaryreducing substances
Galactosemia
Hereditary fructose intolerance
Lowe syndrome
Discolored urine
Alkaptonuria
Tryptophan malabsorption
Leukopenia
Organic acidemias
Glycogen storage disease type 1B
Barth syndrome
Pearson syndrome
MRI, Magnetic resonance imaging.
The third important category of metabolic disorders
is insufficient generation of energy by the mitochondrial
machinery. These disorders can be caused by the inability to provide substrates such as glucose in glycogenoses;
the inability to deliver substrate to the site of oxidation,
such as the fatty acid and carnitine transport disorders; the
inability to break down fatty acids in a stepwise fashion
to provide reduced flavin adenine dinucleotide to be oxidized; or the deficient function of the mitochondrial respiratory pathway and energy-generating system itself. These
disorders have characteristics in between those of the
acute, small molecule disorders and the storage disorders
CHAPTER 21 Introduction to Metabolic and Biochemical Genetic Disease
Box 21-5 S
igns and Symptoms of Inborn
Errors in the Newborn
ll
ll
ll
ll
ll
ll
ll
ll
ll
Neonatal catastrophe (life threatening)
Poor suck and feeding
Gastrointestinal problems, vomiting
Respiratory distress
Cardiac failure
Neurologic abnormalities: alertness, tone, seizures
Organomegaly
Ocular abnormalities
Cutaneous changes
(see Box 21-4). They differ from the small molecule disorders in the possible onset immediately at birth or before,
and from storage disorders in the generally normal physical
features with hepatomegaly alone, a regular feature of glycogen storage disorders. The small molecule and energygenerating disorders are discussed in Chapter 22 and the
lysosomal storage disorders are discussed in Chapter 23.
Of the remaining groups that are encountered less
frequently, the peroxisomal biogenesis disorders, carbohydrate-deficient glycoprotein disorders, and SmithLemli-Opitz syndrome (a cholesterol biosynthetic disorder)
are discussed in Chapter 23. Other disorders are too infrequent to be considered in a general neonatology textbook.
Disorders of biogenic amines are discussed in Chapter 22
and in greater depth in Chapter 63 on neonatal seizures,
along with consideration of pyridoxine and folate disorders.
SIGNS AND SYMPTOMS OF INBORN
ERRORS
The limited symptomatic repertoire of the sick newborn is
well established, but it is worth repeating. For this reason,
the first thought when confronting a newborn in deteriorating condition, with lethargy, poor suck, temperature
instability, and neurologic abnormalities, is sepsis (Box
21-5). Most metabolic specialists have never confronted
a sick newborn who has not had a “septic workup” and
who is not receiving standard antibiotics. The issue then
becomes when to perform a metabolic workup. The standard answer is that it should be performed when the neonatologist is concerned that the newborn in extremis does
not fit the pattern that they expect from a child with sepsis or hypoxia. That threshold would vary by individual.
Negative results of tests for infectious agents, a nonconfirmatory white count, hypoglycemia, unexpectedly severe
acidosis, or hyperammonemia could be important triggers. Although dysmorphic features are not characteristic
of inborn errors, there are some that may have subtle or
occasionally pronounced abnormality on a physical examination; they are listed in Box 21-6 and in Table 21-2.
Modern neonatology has one tool that was previously
unavailable: the expanded newborn screen. This screening will diminish the probability of many disorders, and
the newborn screening follow-up hotline should be on the
speed dial of every neonatal intensive care unit. The only
acutely presenting disorders not ascertained by these studies are most hyperammonemias and lactic acidoses. These
test results are available immediately in any tertiary or
213
Box 21-6 M
etabolic Diseases With
Congenital Malformations
or Dysmorphic Features
ll
ll
ll
ll
ll
ll
ll
Cholesterol biosynthetic disorders
Peroxisomal disorders
Glutaric aciduria type 2
Primary lactic acidoses
Congenital defects in glycosylation
Lysosomal storage disorders
Menkes disease
secondary care hospital. With a high level of concern and
near normal levels of lactate and ammonia, the standard
battery of metabolic studies should be performed only when
the index of suspicion for a metabolic disorder is particularly
high and no alternative explanation for the poor condition
of the patient is likely. When deemed necessary, these studies should include plasma amino acids, plasma acylcarnitine
levels, and urinary organic acids. The abnormalities associated with individual disorders are discussed in Chapter 22.
EMERGENCY TREATMENT
An acutely ill child with an inborn error of metabolism is
an emergency, and rapid rescue treatment is mandatory.
When considering a differential diagnosis, special emphasis should be placed on disorders for which there is treatment, as opposed to those for which there is no treatment.
As with all acutely ill patients, supportive care, including
cardiorespiratory, hemodynamic status, fluids, and electrolytes, is the mainstay of treatment. Transfer from institution to institution should not be performed unless the
patient’s condition is stable and there is adequate vascular
access for emergency treatment. Transfer to a tertiary care
center with experience in caring for these children is desirable and should be performed as quickly as possible.
Virtually all disorders require a maximum source of
calories (150 calories/kg is desirable, but 120 calories/kg
is a minimum target) to prevent or diminish catabolism,
and glucose is the most important part of this, especially
in the absence of primary lactic acidemia or acidosis. As
much lipid as is safe should be added to compensate for
the caloric deficit.
Hemodialysis, and especially extracorporeal membrane
oxygenation, will remove most metabolites rapidly, but these
are extreme interventions in a newborn. These interventions should be performed routinely in extreme and symptomatic hyperammonemia (ammonia levels of 400 mol/L
or more), and many would consider dialysis in severe acidosis caused by acids other than lactate or ketones. Once a
diagnosis is established, specific therapy can be initiated. It
is important to emphasize that during this period of generic
therapy, prolonged deprivation of exogenous protein or
amino acids will cause endogenous protein breakdown and
exacerbate the metabolic process. As a result, protein is
added to the intravenous support fluids after 36 to 48 hours,
beginning with 0.25 to 0.5 g/kg/day and increasing gradually to maintenance levels of 1.2 to 1.5 g/kg/day, pending a
definitive diagnosis and assuming that it is tolerated.
214
PART VI Metabolic and Endocrine Disorders of the Newborn
SUGGESTED READINGS
Fernandes J, Saudubray J-M, van den Berghe G, et al, editors: Inborn metabolic diseases: diagnosis and treatmented, ed 4, Germany, 2006, Springer-Verlag.
Online Mendelian Inheritance in Man, OMIM: McKusick-Nathans Institute of
Genetic Medicine, Johns Hopkins University (Baltimore, Md.) and National
Center for Biotechnology Information, National Library of Medicine (Bethesda,
Md.), Accessed {date of download}. Available at www.ncbi.nlm.nih.gov/omim/.
Saudubray JM, Charpentier C: Clinical phenotypes: diagnosis/algorithms. In
Valle D, et al, editors: The metabolic and molecular bases of inherited disease.
Available online at www.ommbid.com, see Chapter 66.
Saudubray J-M, Desguerre I, Sedel F, et al: Classification of inborn errors of
metabolism. In Fernandes JM, Saudubray J-M, van den Berghe G, Walter
JH, editors: Inborn metabolic diseases: diagnosis and treatment, Heidelberg, 2006,
Springer-verlag, pp 1-47.
Valle D, et al, editors: The metabolic and molecular bases of inherited disease. Available
online at www.ommbid.com.
C H A P T E R
22
Inborn Errors of Carbohydrate, Ammonia,
Amino Acid, and Organic Acid Metabolism
Stephen Cederbaum and Gerard T. Berry
The inborn errors of carbohydrate, ammonia, amino acid,
and organic acid metabolism have one factor in common:
all can be associated with acute, life-threatening disease
during the newborn period. The most notable exception
in this broad group of small-molecule disorders is phenylketonuria (PKU). There are often few signs secondary
to classic PKU in the first 6 months of life, underscoring
the importance of newborn screening in establishing the
diagnosis of this disease. Although this chapter will present the most common phenotype for these disorders, special emphasis will be placed on the neonatal presentations.
The fact that a number of the more mild forms can manifest later merely emphasizes the importance of newborn
screening in their mitigation. The disorders that constitute each group are listed in Boxes 22-1 to 22-4. The interrelationships among the major metabolites, biochemical
cycles, and organelle pathways in the most critical facets
of intermediary metabolism are simplified and depicted in
Figure 22-1. The primary lactic acidosis and mitochondrial respiratory chain disorders, as well as the defects in
fatty acid oxidation, are included in the section on inborn
errors of organic acid metabolism.
INBORN ERRORS OF CARBOHYDRATE
METABOLISM
syndrome was a much more common occurrence, associated with unlimited intake of lactose in the proprietary
formula or breast milk. Because death from Escherichia
coli sepsis can occur with only 1 to 2 weeks of exposure to
galactose, the incidence of the disorder in the prescreening era was estimated to be less than 1 in 200,000 births.
Today, even when picked up later after screening, the disorder is rarely fatal because the liver dysfunction is reversible and sepsis is largely prevented.
If patients survive the neonatal period and there is no
diagnosis, the most common initial clinical signs of GALT
deficiency is poor growth, irritability, lethargy, vomiting,
and poor feeding. Jaundice may be present in the first few
weeks of life and can persist. Initially the hyperbilirubinemia may be indirect and is only later associated with an
elevation of the direct component as well. The current
tendency to change formulas can mask the disease if a
non–lactose-containing formula is substituted fortuitously.
With continual lactose ingestion, multiorgan toxicity syndrome ensues, which is associated with liver disease that
can progress to cirrhosis with portal hypertension, splenomegaly, ascites, renal tubular dysfunction, and sometimes
full-blown renal Fanconi syndrome. Anemia, primarily
caused by decreased red blood cell (RBC) survival, and
lethargy; brain edema associated with a bulging fontanel
HEREDITARY GALACTOSEMIA
BOX 22-1 Inborn Errors of Carbohydrate
Metabolism
Galactose-1-Phosphate-Uridyltransferase
Deficiency
ll
The three enzymes of the galactose metabolic pathway
that are responsible for the rapid conversion of galactose
to glucose in the liver after ingestion of dietary lactose
or the breakdown of endogenous galactose-containing
compounds are galactokinase, galactose-1-phosphate uridyl transferase (GALT), and uridine diphosphate (UDP)
galactose-4-epimerase (Figure 22-2). All three enzymes
have been associated with inborn errors of galactose
metabolism (Berry et al, 2006; Fridovich-Keil and Walter,
2008). Although a deficiency of any of the three enzymes
can lead to galactose accumulation in plasma, the term
galactosemia refers to GALT deficiency, the most common
of the three enzyme deficiencies in the newborn period.
The frequency of clinically significant galactosemia is estimated at 1 in 60,000 births in the United States, but would
be higher if the more frequent partial deficiencies are considered. The clinical syndrome of transferase-deficiency
galactosemia has changed since the advent of newborn
screening. In instances of the rapid availability of newborn
screening results (3 to 4 days of life) patients rarely require
hospitalization. In the past, a severe multiorgan toxicity
ll
ll
ll
Hereditary galactosemia
Glycogen storage diseases
Hereditary fructose intolerance
Fructose-1,6-bisphosphatase deficiency
BOX 22-2 Inborn Errors of Ammonia
Metabolism
ll
ll
ll
ll
ll
Ornithine transcarbamylase deficiency
Argininosuccinicaciduria
Citrullinemia
Carbamylphosphate synthetase deficiency
Transient hyperammonemia of the newborn
BOX 22-3 Inborn Errors of Amino Acid
Metabolism
ll
ll
ll
ll
ll
Maple syrup urine disease
Hereditary tyrosinemia type 1
Nonketotic hyperglycinemia
Methionine synthetase deficiency
Phenylketonuria
215
216
PART VI Metabolic and Endocrine Disorders of the Newborn
BOX 22-4 Inborn Errors of Organic Acid
Metabolism
ll
ll
ll
ll
ll
ll
ll
ll
Methylmalonic acidemia
Propionic acidemia
Isovaleric acidemia
Multiple carboxylase deficiency
Glutaric acidemia type 1
Fatty acid oxidation disorders
ll Glutaric acidemia type 2
ll Very-long-chain acyl-CoA dehydrogenase deficiency
ll Medium-chain acyl-CoA dehydrogenase deficiency
ll Short-chain acyl-CoA dehydrogenase deficiency
ll Long-chain 3-hydroxy acyl-CoA dehydrogenase deficiency
ll Carnitine transporter defect
ll Carnitine palmitoyltransferase type I deficiency
ll Carnitine palmitoyltransferase type II deficiency
ll Acylcarnitine translocase deficiency
Defects in ketone metabolism
ll Ketothiolase deficiency
ll Succinyl-CoA: 3-ketoacid-CoA transferase deficiency
ll 3-Hydroxy-3-methylglutaryl-CoA lyase deficiency
Primary lactic acidoses
ll Pyruvate dehydrogenase complex deficiency
ll Pyruvate carboxylase deficiency
ll Phosphoenolpyruvate carboxykinase deficiency
ll Mitochondrial respiratory/electron transport chain defects
ll Barth syndrome
ll Pearson syndrome
ll Leigh disease
CoA, Coenzyme A.
can also occur. Cataracts may be evident in the first few
weeks of life. However, some infants are born with congenital cataracts that are associated with abnormalities of
the embryonal lens; they are central in nature and require
slit-lamp examination for documentation.
After initiation of a lactose-free diet in the newborn
period, the problems related to liver and kidney disease,
anemia, and brain edema usually disappear, unless there has
been severe organ damage such as hepatic cirrhosis. Most
infants begin to grow and develop at a normal rate. However,
patients treated prospectively can manifest long-term complications related to speech defects, delay in language acquisition, learning problems, frank mental retardation, autistic
features, and hypergonadotropic hypogonadism in most of
the females. The cause of these so-called diet-independent
complications is unknown. Patients with galactosemia continue a lactose-restricted diet for their entire lives, although
many lapse as they get older and may not suffer from detectable consequences. A minority of patients with GALT deficiency develop a neurodegenerative condition. One of the
first abnormalities to be detected—albuminuria—reflects
a poorly understood renal glomerular component. This
component develops within 24 to 48 hours of ingestion of
lactose and disappears as quickly after elimination of lactose
from the diet. In addition to hyperbilirubinemia, there may
be mild to severe elevations of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels
and various abnormalities related to renal tubular dysfunction, such as hyperchloremic metabolic acidosis, hypophosphatemia, glucosuria, and generalized aminoaciduria.
Vitreous hemorrhages are newly recognized complications
in the newborn period.
FIGURE 22-1 Intermediary metabolism interactions among the glycolytic, citric acid cycle, mitochondrial respiratory–electron transport chain,
amino acid, organic acid, and urea cycle pathways. Defects in these primarily catabolic pathways are the chief source of inborn errors of metabolism
that involve the small, simple—not the large—complex molecules. ATP, Adenosine triphosphate. (Adapted from Cowett RM, editor: Principles of
perinatal-neonatal metabolism, ed 2, New York, 1998, Springer-Verlag, p 800.)
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
217
FIGURE 22-2 The important reactions in the galactose metabolic pathway are shown in relation to exogenous, via lactose primarily, and endogenous de novo synthesis of galactose. The reactions catalyzed by enzymes that have not been well delineated or are purported to exist are shown by
broken lines. Carbon skeletons exit the galactose pool via galactokinase (GALK)–mediated conversion to galactose-1-phosphate (galactose-1-P), aldose
reductase–mediated conversion to galactitol, and as galactonate. In patients with severe galactose-1-P-uridyltransferase (GALT) deficiency, there is
little or no conversion of galactose-1-P to uridine diphosphate galactose (UDPgalactose). The epimerization of UDPglucose to UDPgalactose by
UDPgalactose-4-epimerase, the utilization of UDPgalactose in the synthesis of glycoconjugates such as glycoproteins, and their subsequent degradation may constitute the pathways of de novo synthesis of galactose. (Adapted from Berry GT, Nissim I, Gibson JB, et al: Quantitative assessment of whole
body galactose metabolism in galactosemic patients, Eur J Pediatr 156:S44, 1997.)
When an infant’s condition is initially diagnosed, either
through the newborn screening program or because of
the recognition of clinical signs, blood galactose levels
may be as high as 5 to 20 µmol/L, and the RBC galactose1-phosphate level is significantly elevated, as are urine
galactitol levels. During this phase of severe hypergalactosemia, positive reducing substances are present in
the urine. After the patient starts a lactose-free diet, the
RBC galactose-1-phosphate levels in patients with classic galactosemia fall, but they never return to the normal
range, remaining mildly elevated for the lifetime of the
patient.
As noted in the newborn screening chapter, a patient’s
condition can be ascertained by elevated plasma galactose levels, reduced levels of GALT, or both. Because
newborn blood spots can be obtained before significant
lactose ingestion occurs, screening by galactose in plasma
alone is unsatisfactory. Many programs screen for GALT
deficiency alone and sacrifice diagnosis of the far less frequent and less lethal kinase and epimerase deficiencies.
Many modern screening follow-up programs eschew the
measurement of RBC galactose-1-phosphate levels and
instead confirm the enzymatic deficiency with a quantitative assay and perform mutation analysis. Mutation
analysis can identify those with a poorer prognosis and
those who are unlikely to exhibit clinical symptoms. The
mutations also provide information for genetic counseling and prenatal diagnosis, with the latter also performed
effectively by enzymatic means. A GALT variant labeled
the Duarte variant, after its place of discovery, is far
more frequent than the more severe mutations and gives
approximately three fourths of the activity of the normal
allele. Compound heterozygotes for this allele (D2) and
a null allele (G) have approximately 25% of normal activity in red blood cells and are not associated with clinical
symptoms.
GLYCOGEN STORAGE DISEASES
The glycogen storage diseases (GSDs) can be divided
into those types that primarily affect the liver and those
that affect striated muscle (Kishnani et al, 2001; Smit
et al, 2006). With some forms, such as GSD type 3 or
debrancher deficiency, both striated muscle and the liver
may be affected. According to European prevalence data,
the overall frequency of GSD is 1 in 20,000 to 25,000
(Kishnani et al, 2001). With the exceptions of GSD type
2 and or Pompe disease, a lysosomal defect, most of the
patients with glycogenosis do not come to clinical attention in the newborn period. With the exception of a form
of phosphorylase kinase deficiency, which is labeled as type
IX and is inherited in a sex-linked recessive manner, all are
inherited in an autosomal recessive manner. Patients with
the three most common forms of GSD—types 1, 3, and
6—have a phenotype that mimics a small-molecule disorder, because glucose homeostasis is affected.
Glucose-6-Phosphatase Deficiency
GSD type 1 is caused by decreased activity of glucose-6phosphatase, the enzyme that is perched at the terminus of
both glycogenolysis and gluconeogenesis. Several different
biochemical abnormalities can result in this phenotype,
now classified as GSD types 1a, 1b, and 1c. The enzyme
that resides on the anticytoplasmic side of internal membrane spaces of the hepatocyte catalyzes the hydrolysis of
glucose-6-phosphate to glucose and phosphate. Impairments in the transport of either glucose-6-phosphate (type
1b–much less frequent than type 1a) or phosphate (type
1c–extremely rare) may cause decreased function of this
enzyme. The frequency of GSD type 1 is estimated to be 1
in 100,000 births, with most cases being type 1a.
This condition infrequently manifests in the neonatal
period, because “demand” feeding or feeding at short
218
PART VI Metabolic and Endocrine Disorders of the Newborn
intervals prevent symptomatic hypoglycemia and hepatomegaly has usually not yet occurred. The major clinical
findings are poor growth and enlarged abdominal girth as
a result of hepatomegaly, and any of the signs that may be
related to hypoglycemia. The major laboratory findings
are fasting hypoglycemia, ketosis, lactic acidosis, hyperlipidemia (i.e., hypertriglyceridemia), and hyperuricemia.
In patients with type 1b disease caused by a defect in the
microsomal transporter of glucose-6-phosphate, there
may be a history of recurrent infections because of neutropenia and defective neutrophil function and inflammatory bowel disease that may be present in the first year
of life. Diagnosis used to be based on hepatic enzyme
analyses, but molecular diagnostic testing is currently
the first choice. The most important aspect of therapy
is the prevention of brain damage from hypoglycemia
and growth failure. The mainstay of therapy is frequent
feedings and restriction of lactose and sucrose (Kishnani
et al, 2001; Smit et al, 2006). The use of continuous nasogastric feedings or uncooked cornstarch, particularly
during the night, has significantly improved the care of
affected children, and although it does not correct all the
biochemical perturbations, it does improve growth and
can prevent hypoglycemic spells. Intercurrent illness with
increased glucose utilization is particularly hazardous and
a careful plan for emergencies is essential. The leukopenia
of type 1b is helped by a regimen of granulocyte colony
stimulating factor.
Lysosomal α-1,4-Glucosidase Deficiency
GSD type 2, or Pompe disease, is a deficiency of the lysosomal enzyme α-glucosidase. The clinical presentation
of patients is most often in the newborn or early infancy
period with symptoms of heart failure. The frequency is
estimated at 1 in 100,000 births. There is usually marked
cardiomegaly with massive biventricular cardiac wall
thickening and a typical abnormal electrocardiogram,
confirming biventricular hypertrophy and with a short
PR interval. Decreased cardiac output can lead to heart
failure and passive congestion. Infants may also have generalized hypotonia because of a skeletal myopathy. There
is increased deposition of glycogen within the lysosomes
of striated muscle. Except in the instance of passive congestion, the liver is not usually enlarged. Diagnosis is
based on enzyme analysis in dried blood spots or leukocytes (Goldstein et al, 2009; Pompe Disease Diagnostic
Working Group et al, 2008; Zhang et al, 2008). Cardiac
transplantation has been performed to prevent death in
infancy. Onset of symptoms as late as adulthood occurs
when the mutations in one or both of the alleles cause less
complete enzyme loss.
In the last several years enzyme replacement therapy
for this disorder has been developed and has had a favorable effect on the outcome. More than half of the patients
with infantile onset, for whom the disease had previously
been fatal within the first year, can now be rescued with
varying degrees of residual disability. Patients with adolescent and adult onset may do better depending on the
speed with which the diagnosis was made, therapy initiated, and the degree of irreversible muscle damage that
has occurred.
HEREDITARY FRUCTOSE INTOLERANCE
Fructose-1,6-Bisphosphate Aldolase B
Deficiency
Hereditary fructose intolerance is a rare disorder caused
by a deficiency of the enzyme fructose-1,6-bisphosphate
aldolase in liver (Steinmann et al, 2001). The enzyme
deficiency results in an impairment in the conversion of
fructose-1-phosphate to glyceraldehyde and dihydroxyacetone phosphate and therefore in the effective metabolism of fructose. The disorder is inherited as an autosomal
recessive trait. Symptoms are triggered by eating fructose
or sucrose, which produces fructose. The signs begin
when juices and fruit are added to the infant diet and are
mitigated by the aversion that the infant develops to these
foods and drinks. The major clinical findings are pallor, lethargy, poor feeding, vomiting, loose stools, poor
growth, hepatomegaly, and any sign that could be related
to hypoglycemia. The major laboratory findings consist
of hypoglycemia; hypophosphatemia; elevations of serum
ALT and AST, including any of the findings that may be
associated with hepatocellular disease; and the presence of
reducing substances in the urine. The liver disease may be
severe. Patients may be jaundiced with hyperbilirubinemia.
There may be bleeding diathesis. In addition to liver disease, renal tubular dysfunction can lead to the renal Fanconi syndrome. The intuition of the physician is crucial
in establishing the diagnosis. Previously an intravenous
fructose tolerance test was performed under controlled
circumstances, to determine whether serum phosphate
and glucose levels decrease and serum AST and ALT values rise after 15 to 30 minutes of fructose administration.
In the past, confirmation depended on enzyme analysis,
but molecular diagnostic testing is more widely available
now and avoids both the risk of provocative testing and the
laborious enzymatic analysis. The treatment consists of
elimination of dietary fructose and sucrose. Many patients
enter adulthood with excellent teeth and an aversion to
foods containing sucrose or fructose.
FRUCTOSE-1,6-BISPHOSPHATASE
DEFICIENCY
Deficiency of the enzyme fructose-1,6-bisphosphatase
results in an inability to hydrolyze fructose-1,6-bisphosphate to fructose-6-phosphate (Steinmann et al, 2001).
It is a rare disorder. This enzyme is indispensable in gluconeogenesis. The main clinical features are hypoglycemia and signs related to glucose deprivation in the central
nervous system (CNS). The symptoms are due primarily to inadequate food intake and not fructose ingestion, although fructose may exacerbate the abnormalities
induced by fasting adaptation. Enlargement of the liver
because of diffuse steatosis may be present only during
periods of fasting and enhanced gluconeogenesis. The
laboratory findings consist of hypoglycemia, ketosis, and
lactic acidosis. The acidosis caused by accumulation of lactic, 3-hydroxybutyric, and acetoacetic acids can be severe
in this disease. Diagnosis depends on enzymatic or mutation analysis. The therapy consists primarily of avoidance
of fasting and the need for gluconeogenesis.
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
Inborn Errors of Ammonia Metabolism
Inborn errors of the urea cycle, because they manifest in
infancy, exemplify the small-molecule weight disorders in
which symptoms occur episodically, are exacerbated by
protein catabolism, and can be treated by the control of
catabolic stimuli, dietary modification, and the enhanced
removal of offending metabolites. Four disorders in particular cause the majority of cases that occur in the newborn period and in infancy; they are carbamyl phosphate
synthetase I deficiency, ornithine transcarbamylase (OTC)
deficiency, argininosuccinate (ASA) synthetase deficiency
(citrullinemia), and ASA lyase deficiency (argininosuccinic acidemia). The other four genetic disorders of the
urea cycle, N-acetylglutamate synthase deficiency, arginase deficiency, ornithine transporter deficiency, and
citrin deficiency, are rare or do not manifest with neonatal hyperammonemia, or both. In aggregate, disorders of
the urea cycle may be as frequent as 1 in 25,000 births or
more. The entire cycle is outlined in Figure 22-3 and will
be important in understanding the therapeutic modalities.
One round through the urea cycle condenses two molecules of toxic ammonia and one molecule of bicarbonate
to form a molecule of urea that is nontoxic and is readily
excreted in the urine. N-Acetylglutamate, the product of
the first enzyme in the cycle, is an obligatory activator of
carbamyl phosphate synthetase. These two reactions and
OTC, which is responsible for citrulline synthesis, occur in
the mitochondrion. The next three reactions occur in the
cytoplasm after citrulline is transported out of the mitochondrion. The transport of ornithine back into the mitochondrion and the shuttling of aspartate to the cytoplasm
are clearly imperative and account for the two other genes
that cause urea cycle defects.
With the exception of arginase deficiency, each of these
enzyme deficiencies has been associated with disease in
the newborn period. Clinical presentation in the newborn
period is similar for all these defects (Brusilow and Horwich, 1995; Leonard, 2006). Almost all the infants are well
in the first 12 to 24 hours of life until they begin to feed
poorly, vomit, hyperventilate, become irritable and lethargic, and become comatose, usually with seizures. When
these diseases are not treated aggressively, they are almost
always fatal. The treatment requires specific therapy to
lower the waste nitrogen burden, including the toxic substance ammonia, and address the increased intracranial
pressure. The severe encephalopathic and life-threatening
features may be related in large part to brain edema.
Chronic hepatomegaly has been reported in patients with
argininosuccinic aciduria, whereas hepatomegaly is evident only during hyperammonemic episodes in the other
urea cycle disorders. Acute hyperammonemia is associated
with transaminase elevation and liver synthetic dysfunction, but these are rarely more than transient. Histologic
examination of the liver shows modest fatty infiltration
and fibrosis. Children with argininosuccinic aciduria can
also manifest a specific abnormality of the hair known as
trichorrhexis nodosa.
The main laboratory finding in the urea cycle defects
(UCDs) is a plasma ammonium elevation. Plasma ammonium values may vary in different laboratories. In general,
however, with automated chemistry testing for ammonia,
219
the normal plasma values in older infants, children, and
adults range between 10 and 35 μmol/L. However, the
normal plasma ammonium value in newborns may occasionally be as high as 110 μmol/L but is usually somewhat
lower. In patients with newborn-onset UCDs who are
acutely ill, the plasma ammonium levels are often higher
than 1000 or 2000 μmol/L. Patients with UCDs usually do
not have metabolic acidosis unless they are in a terminal
state with vascular collapse or respiratory failure. Instead,
the characteristic acid-base abnormality associated with
hyperammonemia is respiratory alkalosis caused by the
effect of ammonia on the respiratory control centers in the
brainstem.
The various UCDs can usually be distinguished on
the basis of the pattern and levels of plasma amino acids.
Because citrulline is the product of the carbamyl phosphate
synthetase type 1 (CPS-I) and OTC reactions and the substrate for ASA synthetase, its value is critical. In newbornonset N-acetylglutamate synthase (NAGS), CPS-I and
OTC deficiencies, plasma citrulline concentrations may be
undetectable and are always low. With OTC deficiency,
there is increased urinary orotate excretion secondary to
carbamyl phosphate accumulation and pyrimidine synthesis. With NAGS or CPS-I deficiency, carbamyl phosphate
production is decreased or absent, and orotate excretion
is decreased. In citrullinemia, the eponymous amino acid
citrulline has markedly elevated concentrations. With
argininosuccinic aciduria, plasma citrulline concentration
is moderately elevated, in the range of 100 to 300 μmol/L,
and can be readily detected during a study of plasma by
amino acid analysis.
Because the ability of infants with these disorders to
excrete waste nitrogen as urea is impaired, therapy is
initially focused on the reduction of nitrogen intake by
decreasing dietary protein and providing essential amino
acids or the ketoacid analogues. This approach theoretically permits adequate growth without an excessive nitrogen load. Excessive protein leads to hyperammonemia, but
too much restriction of protein during long-term therapy
leads to poor growth and can provoke catabolism to maintain essential amino acid levels. Actually this approach
fails when the patient is in a catabolic state and in negative
nitrogen balance, as occurs in the catastrophically ill infant
presenting in the first week of life with massive hyperammonemia. For such an infant with hyperammonemia
and coma, the mainstay of therapy is dialysis treatment.
Hemodialysis (or extracorporeal membrane oxygenation)
is the most effective way of reducing plasma ammonium
levels, because it affords the greatest clearance of ammonia
(Rutledge et al, 1990). Continuous arteriovenous hemofiltration (CAVH) provides a lower clearance rate, but has
the added benefit of continuous use and a lesser likelihood
of major swings in intravascular volume that can exacerbate an already fragile state and cerebral edema. Ammonia
clearance with peritoneal dialysis is only approximately
one tenth that of CAVH and is not recommended for specific UCD therapy in the newborn period. Ammonia is not
cleared effectively by exchange transfusion.
While the intensive care personnel are waiting for
dialysis to be started, alternative waste nitrogen therapy
using intravenous sodium benzoate, sodium phenylacetate (together as Ammonul), and, for patients with
220
PART VI Metabolic and Endocrine Disorders of the Newborn
A
B
FIGURE 22-3 A, Depicted is the nonhomogeneous distribution of enzymes involved in ammonia metabolism in hepatocytes of an acinar sinusoid
as they are linearly distributed from the portal triad to the region of the central vein or terminal hepatic venule. The specific enzymatic reactions are
shown for an individual periportal and perivenous hepatocyte. The glutamine synthetase (GS) and ornithine aminotransferase (OAT) enzyme activities are expressed exclusively in the one- to three-cell layers surrounding the central vein—that is, the region of zone 3 of the liver lobule—whereas
the urea cycle enzymes are concentrated within the periportal hepatocytes. However, the urea cycle enzyme activities are higher in zone 1 immediately surrounding the portal triad than in the middle zone 2. The hepatocytes are shown as squares, the hepatic mitochondria as shaded circles, and the
lining of the space of Disse as the broken lines on either side of the linear array of hepatocytes. Arg, Arginine; ASA, argininosuccinate; Asp, aspartate;
ATP, adenosine triphosphate; Cit, citrulline; CP, carbamylphosphate; CPS-I, carbamylphosphate synthetase type 1; Gln, glutamine; Glu, glutamate;
α-KG, α-ketoglutarate; NH3, ammonia; CO2, CO2 or bicarbonate; Orn, ornithine; P5C, pyrroline 5-carboxylate; P5CDH, pyrroline-5-carboxylate
dehydrogenase. B, The medications sodium phenylacetate and phenylbutyrate and sodium benzoate promote alternative waste nitrogen disposal by
participating in these two mitochondrial reactions. (Adapted from Tuchman M, et al: Hepatic glutamine synthetase deficiency in fatal hyperammonemia after
lung transplantation, Ann Intern Med 127:447, 1997.)
ASA synthetase and ASA lyase deficiencies especially,
arginine hydrochloride should be given (Brusilow, 1991;
Brusilow and Batshaw, 1979; Brusilow et al, 1979). The
medications and the proper dose for a bolus and 24-hour
sustaining infusions of Ammonul are available from Ucyclyd Pharma (Scottsdale, Arizona). This product is rarely
stocked except in pharmacies of tertiary care metabolic
centers. Arginine hydrochloride is thought to be helpful in
NAGS, CPS-I, and OTC deficiencies and is given at a continuous dose of 250 mg/kg per 24 hours. A dose of 600 mg/
kg/day is recommended for ASA lyase particularly and for
citrullinemia. Arginine supplementation the body arginine
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
pool, and in citrullinemia and argininosuccinic acidemia it
increases ornithine to promote synthesis of citrulline and
ASA. ASA contains both waste nitrogen atoms destined for
excretion as urea and has a renal clearance rate equal to
the glomerular filtration rate, provided that it is continuously synthesized and excreted. Accordingly, ASA should
serve as an effective substitute for urea as a waste nitrogen
product. The excretion of citrulline, which contains only
one molecule of ammonia, also exceeds that of ammonia,
although it does not appear to be eliminated as efficiently
as ASA.
Sodium benzoate promotes ammonia excretion when
it is conjugated with glycine to form hippurate, which
is cleared by the kidney at fivefold the glomerular filtration rate (see Figure 22-3).Theoretically, 1 mole of waste
nitrogen is synthesized and excreted as hippurate for each
mole of benzoate administered. The hippurate synthetic
mechanism resides primarily in the hepatic mitochondria
and depends on an intact mitochondrial energy system
for adenosine triphosphate (ATP) synthesis. The glycine consumed in this reaction can be replaced by either
serine or the glycine cleavage pathway. Sodium phenylacetate, as well as sodium phenylbutyrate, which is used
for long-term therapy in the absence of sodium benzoate,
conjugates with glutamine to form phenylacetylglutamine,
which is excreted by the kidney (see Figure 22-3). Sodium
phenylbutyrate is converted to phenylacetate in the liver.
Glutamine contains two nitrogen atoms, whereas glycine
contains one. Two moles of waste nitrogen are removed
for each mole of phenylacetate administered. This acetylation reaction occurs in the kidney and the liver.
The outcome for patients with severe newborn-onset
CPS-I and OTC deficiencies is poor. Sometimes dialysis
therapy cannot rescue severely affected boys with X-linked
OTC deficiency in the first few days of life (Enns et al,
2008). Prospectively administered alternative pathway
therapy in conjunction with high-calorie fluids usually
prevents death and severe hyperammonemia in patients
known from family studies or prenatal diagnosis to be at
risk. This therapy is done best in collaboration with an
expert in the treatment of urea cycle disorders. Even after
institution of successful therapy, the morbidity and mortality rates are high in these severely affected patients and
mental retardation is common in survivors (Brusilow and
Horwich, 1995). There is a significant correlation between
the duration of newborn hyperammonemic coma and the
developmental quotient score at 12 months of age (Msall
et al, 1984). Four of five reported children in whom duration of coma was 2 days or less had normal intelligence
quotient scores, whereas all seven children in whom coma
lasted 5 days or longer were severely mentally retarded.
Currently liver transplantation is recommended for
patients with CPS-I and OTC deficiencies in the newborn
period. The patient should have almost no residual enzyme
activity and grow to a sufficient size for the treatment to be
feasible and safe. Early diagnosis and treatment is important because of the devastating effects of prolonged newborn hyperammonemic coma.
Mutational analysis of DNA is available for all of these
disorders at www.genetests.org. If the lesion in a particular family is known, prenatal diagnosis by means of
direct DNA analysis is also feasible. With the exception
221
of OTC deficiency, all the UCDs are inherited as autosomal recessive traits.
TRANSIENT HYPERAMMONEMIA
OF THE NEWBORN
Transient hyperammonemia of the newborn is a distinct
clinical syndrome that was first identified by Ballard et al
(1978). The disease usually develops in premature infants
during the course of treatment for respiratory distress syndrome. The plasma ammonium level may be enormously
elevated, as high as that found in any of the patients with the
most severe type of UCD. Its onset is usually in the first 24
hours after birth, when the infant is undergoing mechanical ventilatory support. Affected babies can manifest all the
signs associated with hyperammonemic coma. The diagnosis may not be obvious, however, because many of these
same infants are receiving sedatives and muscle relaxants to
optimize therapy of their life-threatening pulmonary disease and an ammonia level is not determined. Important
clues are the absence of deep tendon reflexes, the absence
of normal newborn reflexes, and a decrease or absence of
response to painful stimuli. As with hyperammonemic coma
in UCD, this medical emergency requires dialysis therapy.
The cause of this disease is unknown. The plasma amino
acid levels are similar to those found in CPS-I or OTC
deficiency. Investigators have hypothesized that the disorder may be caused by impairment of hepatic mitochondrial
energy production or shunting of portal blood away from
the liver, such as in patent ductus venosus. The mortality rate in transient hyperammonemia of the newborn is
high. A patient who can be treated early and aggressively
may survive the episode. There is no evidence that any of
the survivors have suffered any further episodes of hyperammonemia, nor has there been any further evidence of
impaired ammonia metabolism.
INBORN ERRORS OF AMINO ACID
METABOLISM
MAPLE SYRUP URINE DISEASE
Branched-Chain 2-Keto Dehydrogenase
Complex Deficiency
Maple syrup urine disease (MSUD) is a rare inborn error
of amino acid metabolism (Chuang et al, 2001; Wendel and de Baulny, 2006). It is inherited as an autosomal
recessive trait and is caused by a deficiency of the enzyme
branched-chain 2-keto dehydrogenase (BCKAD) complex. This enzyme catalyzes the conversion of each of the
3-ketoacid derivatives of the branched-chain amino acids
(BCAAs)—leucine, isoleucine, and valine—into their
decarboxylated coenzyme metabolites within the mitochondria (Figure 22-4). The disease occurs in 1 in 200,000
newborn infants around the world, but in the Mennonite
communities of the United States, the frequency is 1 in
358 because of a founder effect for a point mutation in the
E1α gene. The most common presentation of the disorder occurs in the newborn period. Other forms with later
onset caused by less complete enzyme deficiencies occur,
but they are not the subject of this chapter. The newborn
222
PART VI Metabolic and Endocrine Disorders of the Newborn
FIGURE 22-4 The branched-chain amino acids leucine, isoleucine, and valine are reversibly transaminated to their corresponding 2-keto analogues,
which are substrates for the single decarboxylase (branched-chain 2-keto dehydrogenase) enzyme deficient in maple syrup urine disease. Reduced
activity of isovaleryl coenzyme A (CoA) dehydrogenase, the next enzyme in the leucine degradative pathway, is the cause of isovaleric acidemia. The
immediate precursor of ketones, 3-hydroxy-3-methylglutaryl CoA (HMG-CoA), is the final product of the leucine catabolic pathway. However, its
production more strongly depends on the oxidation of fatty acids as in ketogenesis and in cholesterol biosynthesis. Propionyl CoA, which accumulates
in both propionic and methylmalonic acidemia, can be synthesized from isoleucine, valine, odd-chain fatty acids, cholesterol, methionine, threonine,
and thymine. The adenosyl form of vitamin B12 is the important cofactor in the l-methylmalonyl CoA mutase–catalyzed conversion of
l-methylmalonyl-CoA to the citric acid cycle intermediate, succinyl CoA.
presentation is associated with a severe and catastrophic
illness and usually results in death without specific medical
intervention. Typically the infants are well at birth; only
after 2 or 3 days of ingestion of breast milk or formula
do they begin to manifest poor feeding and regurgitation.
Lethargy becomes evident; the cry may be shrill and high
pitched. There may be hypotonia alternating with hypertonia and opisthotonic posturing. The odor of maple syrup
may be detected in saliva, on the breath, in urine and feces,
and in cerumen obtained from the ear. The babies become
more and more obtunded and eventually lapse into a deep
coma. The anterior fontanel may be bulging. Seizures may
develop. The life-threatening encephalopathic features
may simply be related to brain edema (Chuang et al, 2001).
The clinical biochemical laboratory hallmark is metabolic acidosis. The anion gap may be raised, but not necessarily. There is almost always ketonuria. The plasma
ammonium values are usually normal. The complete
blood count is usually normal. The levels of the plasma
BCAAs leucine, isoleucine, and valine are elevated, with
striking elevation in leucine, the probable toxic metabolite. The normal ranges for plasma leucine, isoleucine,
and valine in the newborn period are 29 to 152 μmol/L,
11 to 87 μmol/L, and 71 to 236 μmol/L, respectively. In
patients who are critically ill, the leucine levels may range
between 1900 and 6900 μmol/L. In the past, almost every
newborn infant who was recognized to have MSUD and
was severely ill was treated with peritoneal dialysis in a tertiary care center. The treatment was clinically successful
in most instances, but it did not allow for as rapid a rate
of reduction in plasma BCAA levels as did hemodialysis or
CAVH (Rutledge et al, 1990).
It is now clear that a nutritional approach works as well
as peritoneal dialysis in newborns with MSUD (Berry
et al, 1991; Chuang et al, 2001; Morton et al, 2002; Parini
et al, 1993; Wendel and de Baulny, 2006). It is best performed with the use of a BCAA-free modified parenteral
nutrition solution for infants and older children with acute
metabolic decompensation, but this is rarely available in
any center that does not have a larger Mennonite population. Protein-free intravenous alimentation augmented
with a low-volume, enteral solution of the 17 nonbranched
chain amino acids for 24 to 48 hours is satisfactory. An insulin drip may also be necessary to curtail the effects of the
catabolic stimulus (Berry et al, 1991; Wendel et al, 1982).
On the basis of the rate of plasma leucine decline, the staff
at Children’s Hospital of Philadelphia have found that this
nutritional therapy is comparable to peritoneal dialysis
when plasma leucine levels are as high as 1908 to 3053
μmol/L. However, CAVH or hemodialysis may achieve
more rapid normalization of the plasma BCAAs and their
corresponding branched-chain ketoacids when the levels of leucine are in the range of 4580 to 6870 μmol/L.
Newborn infants with an odor of maple syrup, a metabolic
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
acidosis, and ketonuria require immediate metabolic consultation assistance and a confirmatory plasma amino acid
quantitation. This is truly a medical emergency with a high
risk of death and permanent brain damage. Most patients
who are successfully treated by 7 days of age do not have
mental retardation. Patients whose diagnosis and therapy
are delayed—and perhaps even in those whose disorder is
diagnosed in the first week of life, but who have suffered
such severe damage because of increased intracranial
pressure—often have substantial decreases in developmental or intelligence quotient, as well as signs compatible
with spastic diplegia or quadriplegia.
Expanded newborn screening is going to lead to a
changed picture for this disorder. In jurisdictions in which
the turnaround time for the program are as fast as 3 to 4
days of life, the patient will likely be less ill and the diagnosis will be known. The outcome will be better because less
risk of permanent damage will have occured.
The mainstay of long-term therapy for patients with
MSUD who survive the newborn period is a special formula
devoid of the BCAAs. The amount of BCAAs necessary to
sustain growth but maintain plasma leucine, isoleucine, and
valine levels in the normal range is supplied with a regular
proprietary formula in limited amounts. In the first week
of life, this amount is approximately 100 mg of leucine per
kilogram of body weight daily and 50 mg/kg daily of both
isoleucine and valine. The BCAA requirements, and thus
the rates of utilization of the BCAAs for protein accretion,
drop rapidly in the first year of life in conjunction with the
decline in growth velocity in young infants. As with PKU,
it is imperative that the administration of the special amino
acid formula be carefully monitored by means of frequent
plasma amino acid quantitations. One of the most common errors made in the treatment of MSUD is the failure
to administer adequate amounts of supplemental isoleucine and valine solutions to maintain normal plasma levels,
because proprietary formulas alone do not always meet the
needs of each growing baby (i.e., adequate amounts of each
BCAA are not supplied by a formula alone). Deficiency of
BCAAs can result in a severe exfoliative rash and anemia.
The rash may mimic that of severe acrodermatitis enteropathica (Giacoia and Berry, 1993).
As with many of the inborn errors of amino acid, organic
acid, and ammonia metabolism, protein administration in
the infant whose disease is controlled and who is in metabolic balance with diet therapy must be discontinued during periods of intercurrent infections because catabolism,
possibly driven by counterregulatory hormones or cytokines, triggers metabolic decompensation through the
release of branched-chain ketoacid from skeletal muscle.
Metabolic decompensation is further exacerbated by poor
nutritional intake. It is imperative that during these times,
adequate calories and fluids be given to prevent the crisis
from escalating into a medical emergency.
In selected instances, a molecular diagnosis of MSUD may
be undertaken; this diagnosis is especially useful in targeted
populations such as the Mennonites. Rarely patients may have
a defect in the enzyme 2(E2) component (Danner et al, 1985)
or, as discussed in the Primary Lactic Acidosis section, in the
enzyme 3(E3) component. The rare patient with MSUD
may respond favorably to high-dose thiamine therapy with a
reduction in BCAAs and a lower need for protein restriction.
223
HEREDITARY TYROSINEMIA TYPE 1 OR
HEPATORENAL TYROSINEMIA
Fumarylacetoacetate Hydrolase Deficiency
Tyrosinemia type 1 is caused by a deficiency of the enzyme
fumarylacetoacetate hydrolase (Tanguay et al, 1995). This
enzymatic reaction is distal in the phenylalanine and tyrosine pathway, and the disorder is actually an inborn error
of organic acid metabolism, with hypertyrosinemia being
a secondary and variable biochemical effect. Tyrosinemia
type 1 is a rare disease inherited in an autosomal recessive manner. The highest incidence is found in those of
French-Canadian ancestry as a result of a founder effect
(De Bracketeer and Larochelle, 1990).
The phenotype is variable. At one extreme, the patient
is in early infancy and has severe, usually fatal disease in
which liver disease dominates the clinical picture. At the
other end of the spectrum is a more chronic phenotype,
and patients exhibit hypophosphatemic rickets related to
renal Fanconi syndrome. These patients usually also have
evidence of liver disease, although milder. Most of the
patients with severe liver disease phenotype do not come
to clinical attention in the newborn period. Careful search,
however, may reveal laboratory findings compatible with
this disease, even in newborn infants. In the phenotype seen
in early infancy, the clinical findings are hepatomegaly, a
bleeding diathesis, and jaundice. Ascites is not uncommon.
The laboratory findings consist of abnormal liver function
and dysfunction tests with increases in serum AST, ALT,
and direct and indirect bilirubin; prolongations of PT and
PTT; and findings related to renal Fanconi syndrome,
such as glycosuria, hypophosphatemia, hypouricemia, proteinuria caused by β2-microglobulin hyperexcretion, and
generalized amino aciduria and organic aciduria.
More specific laboratory abnormalities consist of an
abnormally high serum alpha-fetoprotein level. There may
be an elevation of plasma tyrosine; however, the hypertyrosinemia and a more prominent hypermethioninemia
are caused by a secondary impairment in liver function.
The most important metabolites are those related to the
substrate, fumarylacetylacetic acid, the handling of which
is defective. Increased levels of the diagnostic metabolite
succinylacetone can be detected on urine organic acid
quantitation by gas chromatography–mass spectrometry (GC-MS) in most instances and even more sensitively by an isotope dilution method, and in RBCs by the
δ-aminolevulinic acid dehydratase inhibition assay. As in
all metabolic disorders, the metabolite abnormalities may
not be present or detectable at all times.
Newborn screening methods are likely to alter the phenotype of this disorder. Increasingly, analysis of blood spot
succinylacetone is being added to the expanded newborn
screening panels and will likely lead to preemptive diagnosis and treatment in the majority of cases. The falsenegative rate for this analysis is not known at this time.
Newborn screening by older methods has been performed
in Quebec for many years.
In the past, most of the infants with the hepatic form of tyrosinemia type 1 died in early to late infancy. However, a medical
therapy has been developed that uses the agent 2-(2-nitro4-trifluoromethylbenzoyl)-1,3-cyclohexanedione
224
PART VI Metabolic and Endocrine Disorders of the Newborn
(NTBC), which inhibits p-hydroxyphenylpyruvate dioxygenase (Lindstedt et al, 1992), thus blocking the conversion
of p-hydroxyphenylpyruvate, the transamination product
of tyrosine, to fumarylacetoacetate and thereby retarding
the synthesis and accumulation of succinyl acetate and succinyl acetone. This therapy has been used successfully to
improve liver and renal function as well as to normalize or
improve the various laboratory abnormalities. It is unclear,
however, whether NTBC therapy will eliminate the need
for liver transplantation, which has been the mainstay of
long-term therapy until recently.
One of the most devastating complications for older
infants with tyrosinemia type 1 who have been stabilized
clinically and biochemically is the development of hepatocellular carcinoma. This development is prevalent in
tyrosinemia treated with diet, and most patients who survive infancy or who have a chronic phenotype succumb to
liver cancer. It is recommended that NTBC therapy be
started in infants who are acutely ill while they are waiting
for a liver transplantation, which may be deferred until the
third year of life, while monitoring the liver for development of a hepatoma. Early treatment with NTBC surely
retards the development of tumors, but patients receiving
therapy develop hepatomas and preemptive transplantation, although at a later age. Liver transplantation is still
a highly prevalent therapy. Most liver transplantations
have been successful, and the patients have done well with
immunosuppressive therapy with only minimal persistent evidence of renal tubular dysfunction while eating a
normal diet.
Hepatorenal tyrosinemia has no direct effect on the nervous system, and most patients are free of neurologic findings and mental retardation. Because succinylacetone can
inhibit delta-amino levulinic acid dehydratase, the enzyme
deficient in a rare type of acute intermittent porphyria,
porphyria symptoms have frequently occurred in the preNTBC era and have proved fatal in some instances.
Hepatorenal tyrosinemia is not to be confused with
transient tyrosinemia of the newborn, which is prevalent
in premature infants and is probably the most common
disturbance of amino acid metabolism in man; it is secondary to a delayed maturation of p-hydroxyphenylpyruvate
dioxygenase activity (Levine et al, 1939; Tanguay et al,
1995) or with the hypertyrosinemia secondary to liver disease. Current screening methods are unlikely to detect this
condition that was found frequently in the past.
NONKETOTIC HYPERGLYCINEMIA
Glycine Cleavage Complex Deficiency
Nonketotic hyperglycinemia is a rare defect of glycine
metabolism (Dulac and Rolland, 2006; Hamosh and Johnston, 1995.) and is inherited as an autosomal recessive trait.
The disorder is caused by deficient activity of the glycine
cleavage enzyme complex (GCC), which consists of the
products of four genes. Its frequency is fewer than 1 in
200,000 newborn infants. Although several variant forms
exist, most infants who come to clinical attention in the
newborn period—presumably most patients with this
disease—have a severe, catastrophic illness that mimics the
most acute forms of ammonia, amino acids, or organic acid
metabolism. The infants may be healthy at birth but begin
to show severe hypotonia and seizures after 12 to 36 hours.
They quickly become comatose, and there is a loss of all
the newborn and deep tendon reflexes.
The clinical findings are predominantly those of an acute
encephalopathy. The electroencephalogram (EEG) usually
shows a characteristic pattern of spike and slow waves. The
babies may have hiccups from diaphragmatic spasms. The
main laboratory finding is an elevation of plasma glycine and
a proportionally higher elevation of glycine in the cerebrospinal fluid (CSF). Unfortunately, plasma glycine levels may
be normal in the newborn period. The urine glycine is usually
also elevated. The most common diagnostic criterion is the
raised CSF–plasma glycine ratio. Values greater than 0.08
are considered to be diagnostic, whereas those between 0.04
and 0.08 are highly suspicious of the diagnosis. The amino
acid serine, which is also a product of the defective enzyme
reaction, is depressed in plasma, and there is a corresponding
increase in the glycine-to-serine ratio in body fluids. Prenatal
CNS lesions have been reported (Dobyns, 1989).
The pathophysiology of this brain disease is not well
understood; it is believed that glycine, an active neurotransmitter, may interfere with the activity of specific
chloride channels, thus perturbing the membrane potential
and depolarization of neurons. There also appears to be an
impairment in alpha-motor neuron outflow tract activity,
producing a clinical state that mimics Werdnig-Hoffmann
disease. No acidosis or ketosis is seen in this disease—thus
the name, nonketotic hyperglycinemia. Many of the disorders
of organic acid metabolism, such as methylmalonic acidemia and propionic acidemia, are also associated with elevations of plasma glycine, presumably because of a secondary
impairment in the GCC, and have been referred to in the
past as the ketotic hyperglycinemia syndromes.
Although many therapies have been tried in this disease—
such as protein restriction, benzoate to trap glycine as
the byproduct hippurate, strychnine to affect the lower
motor neuron function, and dextromethorphan to block
N-methyl-d-aspartate receptors—there is no generally
effective treatment. The seizures are variably controlled
and life may be prolonged, but no meaningful improvement occurs in most instances. Many affected babies die in
the newborn period despite medical support with assisted
ventilation, and most others die within 2 to 4 years. A transient form of nonketotic hyperglycinemia has also been
reported in newborns (Luder et al, 1989; Schiffmann et al,
1989). Valproate may result in hyperglycinemia because
of secondary inhibition of the GCC, as is postulated to
explain secondary hyperglycinemia in disorders of organic
acid metabolism, such as propionic acidemia. Milder forms
exist and milder symptoms appear at a later age. This disorder cannot be diagnosed by newborn screening.
METHIONINE SYNTHETASE DEFICIENCY,
OTHER DISORDERS OF HOMOCYSTEINE
REMETHYLATION AND CYSTATHIONINE
SYNTHASE DEFICIENCY
In humans, the essential amino acid methionine is converted to homocysteine, and in the process a methyl
group is transferred to an acceptor molecule from
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
S-adenosylmethionine that serves as a donor for methyl
groups in many different reactions (see Figure 22-5) (Mudd
et al, 1995). Subsequently the homocysteine may either
be completely metabolized through the cysteine pathway
to sulfate or it may be remethylated back to methionine
to maintain the critical levels of methionine. Defective
methionine remethylation or a deficiency in methionine
synthetase leads to a form of homocystinemia (the name
homocystinuria is anachronistic and does not reflect the current reality of ascertainment or testing as well as does homocystinemia). Patients with classic homocystinemia, caused
by a deficiency of the cystathionine beta-synthase enzyme,
rarely manifest signs in early infancy (Mudd et al, 1995).
In contrast, patients with methionine synthetase deficiency
or methionine remethylation defect may come to clinical
attention in the newborn period (Rosenblatt and Fenton,
2001). The patients may have either an abnormality in vitamin B12 metabolism, which can also produce methylmalonic acidemia and homocystinuria, or an isolated defect
in folate metabolism or the methionine synthetase enzyme
(Rosenblatt and Fenton, 2001). In this reaction the methyl
group from 5-methyltetrahydrofolate, which is derived
from 5,10-methylene tetrahydrofolate, is transferred to
methylcobalamin and subsequently to homocysteine.
The clinical findings associated with the methionine
synthetase deficiencies are poor growth and development.
There may be severe cortical atrophy and possible brain
lesions caused by thromboses of the arteries or veins, as
in classic homocystinuria. Infrequently they can manifest
as an acute intoxicating disorder in the newborn period.
The laboratory findings consist of an elevation in plasma
homocysteine values and a normal or decreased methionine value. Often the homocysteine values are not as elevated as in classic cystathionine β-synthetase deficiency. If
there is a defect in folate or vitamin B12 metabolism that
produces secondary impairment in methionine synthetase
activity, there may also be megaloblastic anemia.
Some patients with a defect in cobalamin metabolism
respond to treatment with high doses of intramuscular
hydroxycobalamin. The treatment of methionine synthesis deficiency is a normal protein intake or methionine
supplementation to restore methionine levels in plasma
to normal and to restore CNS pools of methionine, which
may be critical in one-carbon transfer reactions. It is also
possible to retard homocysteine accumulation and restore
methionine levels by administering betaine, which can
enhance remethylation of homocysteine to methionine
through the alternate betaine methyltransferase pathway.
Some investigators have also used pharmacologic doses
of cobalamin, folate, and pyridoxine to stimulate flux
through either the methionine synthetase or the classic
homocysteine pathway. Unfortunately, if infants with this
type of disorder are not treated early, there is usually a
permanent and devastating effect on cognitive and motor
function. Milder and occasionally asymptomatic forms
are known.
Classic homocystinemia caused by cystathionine synthase deficiency can be ascertained by expanded newborn
screening in a minority of cases. The elevated methionine
that is the critical detected metabolite may require several days to become apparent; that is unfortunate because
cystathionine synthetase deficiency is a treatable disorder.
225
Expanded newborn screening has altered the landscape for
several B12 processing defects. Ascertainment of elevated
methylmalonic acid with the C3 acylcarnitine, the C4
dicarboxylacyl carnitine, or both usually allows for earlier diagnosis and treatment, with apparent mitigation of
long-term effects. These disorders are more common than
classical homocystinemia, the latter having a frequency in
most populations of 1 in 200,000 births or less.
PHENYLKETONURIA
Phenylalanine Hydroxylase Deficiency
PKU is the most common inborn error of amino acid
metabolism that can result in mental retardation (Scriver
et al, 2001; Walter et al, 2006). Its frequency is between 1
in 10,000 to 1 in 25,000 births, depending on where in the
United States it is studied. There are areas of the world
such as Ireland, Turkey, and Iran in which it is more frequent, and others such as Finland, Japan, and among Ashkenazi Jews in which it is less prevalent. PKU is caused
by a defect in the activity of the enzyme phenylalanine
hydroxylase, which converts phenylalanine to tyrosine, a
reaction that resides primarily in the liver. Thus in PKU
it is the deficiency of this liver enzyme that results in brain
disease. Because of the paucity of findings in the newborn
period or early infancy, PKU usually was not diagnosed
until late infancy or later until newborn screening was
instituted. It is important for physicians caring for newborns to be aware of the pitfalls of screening, which are
summarized in Chapter 27. This disease, inherited as an
autosomal recessive trait, exemplifies the interaction of a
gene and the manipulatable environment (i.e., diet) in the
expression of a disease (Scriver et al, 2001).
After birth, the baby with PKU who is undergoing normal postnatal catabolism, or ingesting adequate amounts
of breast milk or a proprietary formula, will experience
a gradual and persistent increase in plasma phenylalanine levels. The cutoff value for newborn screening differs by jurisdiction, but both phenylalanine value and the
phenylalanine:tyrosine ratio are considered when calling a
presumptive positive. The diagnosis is rarely missed if testing occurs after 24 hours of age. By 5 to 7 days of age in the
untreated patient, the eponymous phenylketone is found
in urine, and another side product of phenylalanine accumulation, phenylacetic acid, may impart the characteristic
“mousey” odor to urine and the patient. This odor may be
detected when levels of phenylalanine exceed 600 to 900
μmol/L, but it is not detected in every patient. With modern newborn screening programs, the neonatologist will
rarely see a case of PKU in the hospital and will usually be
dealing with a patient who has a preemptive diagnosis and
is treated as an outpatient. Greater detail about the disorder can be obtained in Scriver et al (2001), but a synopsis
will be given here.
In the first 6 months of life, the affected babies may have
difficulty with feeding and vomiting. In some instances,
persistent vomiting has been associated with the diagnosis
of pyloric stenosis, for which corrective surgery has been
performed, perhaps inappropriately. Developmental delay
is usually evident in the second 6 months of life. Patients
may have seizures, sometimes infantile spasms in early
226
PART VI Metabolic and Endocrine Disorders of the Newborn
infancy associated with a hypsarrhythmic EEG pattern.
Persistent elevation of plasma phenylalanine levels greater
than 600 μmol/L may be sufficient to result in mental
retardation.
The mechanism of brain disease in PKU is still
unknown. It may be related to the effect of high phenylalanine levels on the transport of amino acids across the
blood-brain barrier and then into brain neurons or glial
elements. The plasma levels of tyrosine may be decreased,
especially in patients who are not receiving tyrosine supplementation in the special amino acid powder used as a
daily nutrient. Investigators have speculated for many
years that hypotyrosinemia has a role in the CNS deficits,
because tyrosine (only a liver enzyme) cannot be synthesized within the CNS by phenylalanine hydroxylase (a
liver enzyme). Older infants often exhibit long tract findings, such as spastic quadriparesis and spastic quadriplegia.
The untreated infant demonstrates microcephaly acquired
postnatally and may also have severe behavioral problems
in addition to a diagnosis of autism. Typical findings consist of elevated serum plasma phenylalanine levels, normal
or subnormal plasma tyrosine levels, and increased urinary
excretion of phenylpyruvic acid, phenyllactic acid, and
phenylacetic acid (rarely measured routinely, but easily
seen if urine organic acids are studied).
The mainstay of therapy is a low-protein diet and the
use of a special amino acid–containing formula that does
not include phenylalanine. The patient must receive an
adequate amount of phenylalanine from protein in proprietary formulas and later from table foods, which is tracked
by means of a phenylalanine exchange system, to allow for
the normal daily utilization of phenylalanine for protein
synthesis while maintaining plasma phenylalanine levels in
a range as close as possible to normal, but less than 360
μmol/L. As the patient ages, care-givers may have to be
content with levels less than 600 μmol/L. Deviations from
normal plasma levels are believed to be associated with
chronic, perhaps acute, effects on brain function and testing performance; therefore the diet should be maintained
for life. More recently, other forms of therapy have been
used. Preparations of the large neutral amino acids that
compete for the same transporter as phenylalanine may be
given, especially to patients who cannot comply with the
low–natural protein diet, and more importantly the mandatory daily ingestion of the medical food devoid of phenylalanine, but replete with unpalatable amino acids. Positive
effects have been frequently claimed, but few carefully controlled studies are available. The positive effects of this supplement have variously been attributed to competition with
phenylalanine absorption in the gastrointestinal tract, competition for transport into the brain, and repletion of neurotransmitters. A small fraction of patients with full-blown
PKU respond favorably to high doses of tetrahydrobiopterin, the natural cofactor for phenylalanine hydroxylase,
with a substantial fall in plasma phenylalanine levels and
an increased tolerance for natural protein. The more mild
forms of hyperphenylalaninemia are even more responsive.
The therapy is expensive. Injections or implantations of
a reservoir in which there is an enzyme that breaks down
phenylalanine in the body are in the early phases of testing.
The phenylalanine hydroxylase gene has been cloned
and sequenced, and the mutations responsible for most
abnormalities in humans are known. DNA sequencing
and mutational analysis can be used to identify carriers in
families and to provide a scientific rationale during family
counseling.
In rare cases, patients with hyperphenylalaninemia have
severe disease not because of deficiency of the phenylalanine hydroxylase apoenzyme, but because of deficiency of
the active cofactor of this enzyme, tetrahydrobiopterin.
Several defects in the metabolism of biopterin can produce
this type of hyperphenylalaninemia. Patients with these
uncommon types of PKU usually come to attention in the
newborn period because of severe seizure activity. Patients
with biopterin deficiency can have evidence of severe brain
damage despite treatment with a low-phenylalanine diet.
Brain damage may be related to deficiencies of other neurotransmitters whose synthesis also depends on adequate
levels of tetrahydrobiopterin. These various other defects,
such as the dihydropteridine reductase, 6-pyruvoyl tetrahydropterin synthetase, and the guanosine triphosphate
cyclohydrolase deficiencies, can be ascertained by urinary
measurements of neopterin and biopterin in urine. They
are responsive to therapy with neurotransmitter replacement and tetrahydrobiopterin.
If women with PKU are poorly treated during pregnancy, their children may be born with microcephaly and
congenital heart defects. Mental retardation is common.
Adequate treatment before 8 weeks’ gestation is essential,
and control of phenylalanine levels before pregnancy is
most desirable.
INBORN ERRORS OF ORGANIC
ACID METABOLISM
Defects in the catabolism of the BCAAs are responsible
for most of the disorders of organic acid metabolism
(Figure 22-5). Typical examples are methylmalonic, propionic, and isovaleric acidemias. An organic acid is any
organic compound that contains a carboxy functional group
but no α-amino group as in an amino acid. In this section,
we consider the disorders of fatty acid oxidation, ketone
body metabolism, and lactic acid metabolism as well as the
more classic inherited defects in organic acid metabolism.
Until recently, these disorders were diagnosed only after
symptoms have appeared. It is now more common to be confronted with an asymptomatic or early symptomatic patient
and a good idea of the diagnosis. As discussed earlier, acidosis and encephalopathy are usually the hallmarks of these
syndromes when they manifest in the newborn period.
METHYLMALONIC ACIDEMIA
l-Methylmalonyl–Coenzyme
Mutase Deficiency
A
Methylmalonic acidemia, along with propionic acidemia, is thought to be the most common of disorders of
organic acid metabolism (Fenton et al, 2001; Wendel and
de Baulny, 2006). Although more than one enzyme defect
may result in methylmalonic acidemia, all are inherited as
autosomal recessive traits. In California, defects in cobalamin metabolism are more frequent than apoenzyme deficiency, specifically l-methylmalonyl–coenzyme a (CoA)
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
227
FIGURE 22-5 The pathways, metabolites, and vitamin cofactors important in the interconversion of methionine and homocysteine are shown
in this abbreviated scheme of the trans-sulfuration pathway. In a methyl-transfer reaction, homocysteine is converted to methionine; the reaction
is catalyzed by a cobalamin-containing enzyme, 5-methyl-tetrahydrofolate-homocysteine methyltransferase or methionine synthase. An alternate
reaction involves betaine-homocysteine methyltransferase. Therefore vitamin B12 and folate are important vitamins in homocysteine and methionine
metabolism. The 5,10-methylene tetrahydrofolate (THF) derived from dietary folate is converted to 5-methyl THF by the enzyme 5,10-methylene
THF reductase. In classic homocystinuria, the deficient enzyme is the cystathionine β-synthetase, which catalyzes the conversion of homocysteine
to cystathionase, a precursor of cysteine. This pathway is readily operative in the adult, so that cysteine is a nonessential amino acid. In newborn
infants, however, synthesis of cysteine does not occur at the same rate as in adults. Adenosylmethionine-dependent methyl transfer may be extremely
important in the central nervous system. Most of the patients who come to clinical attention in the newborn period have defects in the remethylation
of homocysteine to methionine; therefore these patients may have a severe deficiency of adenosylmethionine in the brain and spinal cord.
mutase (see Figure 22-4). This enzyme is present in mitochondria, and it depends on adenosyl-cobalamin for activity. Impaired function can result from either a mutation of
the l-methylmalonyl-CoA mutase apoenzyme or deficient
availability of the adenosyl form of vitamin B12. The latter
may result from impaired cellular metabolism of vitamin
B12, including defective activity of the enzyme adenosylcobalamin synthetase. Some patients, but not usually newborn
infants with methylmalonic acidemia caused by apoenzyme
deficiency, are responsive to pharmacologic therapy with
vitamin B12. Methylmalonic acidemia, therefore, is one of
the important disorders that can be considered a vitaminresponsive inborn error of metabolism. Because of deficient
activity of l-methylmalonyl-CoA mutase, the substrate
l-methylmalonyl-CoA accumulates in mitochondria and is
subsequently hydrolyzed to methylmalonic acid. Methylmalonic acid is capable of diffusing out of cells in which it
is being produced, so it may be detected in excess in blood,
CSF, and urine of patients with these various forms of
the disease. The precursors of l-methylmalonyl-CoA are
the BCAAs isoleucine and valine, in addition to methionine, threonine, thymine, and odd-chain fatty acids (see
Figure 22-4).
There are many different phenotypes of methylmalonic
acidemia that range from severe, catastrophic newbornonset disease in the first week of life to an almost benign
form that has been detected in adults with partial l-methylmalonyl-CoA mutase deficiency. Most patients in the
newborn period have a severe phenotype. Dialysis therapy
is not effective in some of the patients in the first week of
life with catastrophic illness, possibly because of a delay
in diagnosis and treatment or the breakdown of other
systems, including the heart and circulation. The most
striking presentation is in the second or third day of life.
The baby is usually well at birth, as with UCDs, but then
gradually begins to manifest problems with feeding, vomiting, lethargy, and perhaps seizures. There may be respiratory distress as a symptom of metabolic acidosis. The
important laboratory findings include metabolic acidosis
usually associated with an increased anion gap, ketosis, and
hyperammonemia. The elevation of plasma ammonium
may be as high as in severe newborn-onset hyperammonemic syndromes. Because ketonuria is relatively uncommon
in newborn infants, even in stressed infants with hypoglycemia caused by poor feeding, and because diabetes mellitus is so uncommon in the newborn period, the physician
caring for newborn infants must always consider an inborn
error of organic acid metabolism when confronted with
an acutely ill baby with ketosis. Other laboratory findings
are thrombocytopenia, leukopenia, and anemia caused by
effects of the metabolite on hematopoietic elements in
bone marrow. Plasma amino acid analysis may reveal elevations of several amino acids, such as glycine and alanine.
Secondary carnitine deficiency with elevations of carnitine
esters is expected (Chalmers et al, 1984). Methylmalonic
acid is detected at high levels in urine with GC-MS. Older
patients with methylmalonic acidemia during crisis have
suffered acute pancreatitis and devastating lesions to the
basal ganglia.
The diagnosis can be confirmed with an assay of the
activity of l-methylmalonyl-CoA mutase enzyme in cultured skin fibroblasts. In addition, various other disturbances in vitamin B12 metabolism can be studied by
analyzing skin fibroblasts in culture. It is more common
228
PART VI Metabolic and Endocrine Disorders of the Newborn
to dissect the condition clinically, ascertain the response to
vitamin B12, and use mutation analysis to confirm the exact
site of the genetic lesion.
The treatment of acute disease consists of protein restriction, empirical therapy with vitamin B12 (1 mg/day intramuscularly), intravenous fluids with 10% glucose and sodium
bicarbonate to correct dehydration, electrolyte imbalance
and acidosis, high-calorie feeds via a nasogastric tube, and
often dialysis. The use of carnitine (25 to 200 mg/kg/day
intravenously or orally) is often recommended, but its usefulness as an acute treatment has not been proved. The
treatment of chronic state centers on the judicious use of a
low-protein diet, an amino acid supplement low in the MMA
precursors, carnitine to alleviate free carnitine deficiency,
and appropriate calories and fluid. Vitamin B12 is used only
when a specific and reproducible response is noted.
Most diagnoses are ascertained by expanded newborn
screening. If the results are rapidly available, the outlook for
the neonatal period may be improved. Episodes of decompensation will still occur. Patients who are particularly brittle or who develop renal failure receive either liver, kidney,
or combined liver-kidney transplants, but their efficacy at
curing the primary disease process has not been proved.
PROPIONIC ACIDEMIA
Propionyl Coenzyme A Carboxylase
Deficiency
Propionic acidemia was the first classic defect in organic
acid metabolism to be described in humans (Fenton
et al, 2001; Wendel and de Baulny, 2006). The first
patient, described by Childs et al in 1961, was sick on the
first day of life with severe metabolic acidosis and ketosis.
He responded to massive alkali therapy and survived the
newborn period. He subsequently suffered multiple episodes of metabolic decompensation with ketoacidosis, usually precipitated by infections or protein ingestion. He also
had developmental delay, a seizure disorder, and episodic
neutropenia and thrombocytopenia. He died at 7 years old.
His sister also demonstrated ketosis and metabolic acidosis
in the first week of life, but because of better control of her
disease, there were few severe episodes of decompensation
in the first and second decades of life.
Propionic acidemia, which is caused by a selective
deficiency of propionyl-CoA carboxylase, is inherited
as an autosomal recessive trait. This disorder was originally called ketotic hyperglycinemia, because of elevations
in plasma glycine along with ketosis. The precursors of
propionyl-CoA include the amino acids isoleucine, valine,
methionine and threonine, thymine, and odd-chain fatty
acids (see Figure 22-4). Of the several hundred patients
described with this disease, most clinical presentations
are in the newborn period with poor feeding, vomiting,
lethargy, and hypotonia. The patients commonly have
seizures and hepatomegaly. The metabolic acidosis may
be severe with or without an increase in the anion gap.
Ketosis is usually present, but not always. Patients who
survive may often show choreoathetosis, because of persistent damage to the basal ganglia. Usually episodes of
metabolic decompensation characterized by acidosis and
ketosis can be precipitated by excessive protein intake or
more commonly infection, with the attendant catabolism.
Such episodes can result in permanent neurologic damage. Subsequent findings therefore include developmental
delay, seizures, cerebral atrophy, and EEG abnormalities.
During the acute attacks, leukopenia, thrombocytopenia,
and less rarely anemia, probably caused by suppression
of maturation of bone marrow hematopoietic precursors,
may be exacerbated.
The diagnosis can be confirmed with GC-MS analysis.
The urine has excess concentrations of various propionate metabolites, such as methyl citrate, propionylglycine,
2-methyl-3-hydroxybutyrate, 2-methylacetoacetate, and
several other, rarer compounds. The plasma glycine value
may be elevated, and during acute attacks the plasma
ammonium value is frequently increased. The enzyme
activity of propionyl-CoA carboxylase can be assayed in
white blood cells or extracts of cultured skin fibroblasts
for definitive diagnosis. Numerous mutations have been
described in the two genes that encode the subunits of this
multimeric enzyme.
Therapy consists of a low-protein diet and adequate
calories. As in MMA, there is secondary carnitine deficiency with elevations of propionylcarnitine. The use of
l-carnitine to relieve a deficiency of free carnitine and
promote greater urinary excretion of propionylcarnitine to lower mitochondrial propionyl-CoA levels is not
proved. Because intestinal bacteria can also contribute to
propionate production, antimicrobial therapy with metronidazole has been used during an acute attack and for longterm therapy. In the acutely ill newborn, the immediate
treatment consists of elimination of protein, total parenteral nutrition, administration of an adequate amount of
calories (10% glucose intravenously or a nonprotein formula, or both, via nasogastric tube infusion), administration of alkali to eliminate metabolic acidosis, and platelet
transfusion if warranted by thrombocytopenia. Patients
frequently require hemodialysis, because of severe acidosis
and coma. It is unclear whether administration of sodium
benzoate and sodium phenylacetate, as in the treatment
of hyperammonemia associated with UCDs, is of benefit
to the patient with secondary hyperammonemia. Several
patients with propionic acidemia have undergone liver
transplantation with mixed success. Episodes of acidosis
are often recurrent between 1 and 3 years of age, after
which some ability to modulate the effects of catabolism
occurs.
Most patients with this condition are now ascertained
by expanded newborn screening. If the results are rapidly available, the outlook for the neonatal period may be
improved. Episodes of decompensation will still occur.
ISOVALERIC ACIDEMIA
Isovaleryl–Coenzyme A Dehydrogenase
Deficiency
Isovaleric acidemia is caused by a selective deficiency of
the enzyme isovaleryl-CoA dehydrogenase (Sweetman and
Williams, 2001; Wendel and de Baulny, 2006). It is inherited
as an autosomal recessive trait. There are two major phenotypes; an acute form manifests as catastrophic disease in the
newborn period, and the late-onset type is characterized by
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
chronic, intermittent episodes of metabolic decompensation. The degree of enzyme deficiency and the mutations
differ between the two extreme presentations. In the acute
form, the infants become extremely sick in the first week
of life. There is usually a history of poor feeding, vomiting, lethargy, and seizures. The characteristic sweaty feet or
rancid cheese odor caused by isovaleric acid is noted on the
body or in urine, especially if it is acidic. Metabolic acidosis
is present, usually with an elevated anion gap and ketosis.
There may be secondary hyperammonemia, thrombocytopenia, neutropenia, and sometimes anemia, resulting in
pancytopenia. The babies usually lapse into a coma. Dialysis therapy may be necessary. As with other organic acid
disorders for which an amino acid determines organic acid
production (see Figure 22-4), treatment also consists of
protein restriction, intravenous fluids with glucose, and
perhaps sodium bicarbonate, protein-free formula with
calories via nasogastric tube, glycine (250 mg/kg/day), and
carnitine supplementation (Sweetman and Williams, 2001).
Intravenous l-carnitine may be beneficial. The excretion of
isovaleric acid as the glycine conjugate is highly efficient,
and symptomatic relief can occur rapidly. Glycine is often
available in compounding pharmacies that make their own
IV alimentation solutions.
In the chronic, intermittent form of isovaleric acidemia,
patients have repeated episodes of metabolic decompensation precipitated by infections, primarily or excessive
protein intake. Some of these episodes may mimic Reye
syndrome. The same therapeutic principles are applied as
for the treatment of the neonatal disorder. The mainstay of
long-term therapy is a diet with limited natural protein, a
valine-free amino acid supplement, and long-term administration of glycine (Berry et al, 1988), which enhances the
production of the nontoxic compound isovalerylglycine
and serves to reduce the free levels of isovaleric acid in
body fluids. In addition, carnitine administration can augment the excretion of isovaleryl carnitine (Berry et al, 1988;
Mayatepek et al, 1991). The benefit of carnitine treatment
in the chronic state, however, remains unproved. Some
patients who remain largely asymptomatic are ascertained
through the expanded newborn screening programs.
Newborn screening and the rapid treatment protocols
can alter the outlook for this condition. In the prescreening era, many patients with neonatal onset could not be
saved, and a number of those with the more indolent forms
have been retarded. This situation should change with
preemptive treatment.
The diagnosis can be made from marked elevations of
isovalerylglycine in urine. There is usually also increased
excretion of 3-hydroxyisovaleric acid. The enzyme
isovaleryl-CoA dehydrogenase can be assayed in extracts
of cultured skin fibroblasts. As with many other conditions,
the typical metabolic profile should allow confirmation
directly by mutation analysis bypassing the enzyme assay.
MULTIPLE CARBOXYLASE DEFICIENCY
There are two enzymatic defects leading to deficiency of
the same suite of carboxylase enzymes, holocarboxylase
synthetase deficiency and biotinidase deficiency (Wolf,
2001). All the carboxylase enzymes—propionyl-CoA carboxylase, pyruvate carboxylase, 3-methylcrotonyl-CoA
229
carboxylase, and acetyl-CoA carboxylase—require covalent linkage with biotin for normal activity. In holocarboxylase synthetase deficiency, the enzyme that catalyzes
the covalent linkage of biotin to a lysine residue of these
various carboxylases is deficient. In the second form of the
disorder, the absence of the enzyme biotinidase does not
allow for biotin recycling after a carboxylase enzyme is
degraded, and hydrolyzed biotinidase deficiency does not
usually manifest in the newborn period.
Patients with severe deficiency in holocarboxylase synthetase, however, characteristically are catastrophically ill in
the newborn period. Patients have severe metabolic acidosis
with lactic acidosis and become comatose. As with biotinidase deficiency, administration of biotin is life saving. Holocarboxylase synthetase deficiency is one of the few disorders
of organic acid metabolism for which the administration
of a vitamin in megadoses produces a dramatic turnabout
in clinical and laboratory findings. Diagnosis depends on
GC-MS analysis of urine and the demonstration of markedly increased levels of lactate, 3-methylcrotonylglycine,
and propionate metabolites. In most patients the affinity of
the holocarboxylase synthetase enzyme for biotin is diminished, and for the biochemical disturbances to normalize
or improve, patients need between 10 and 60 mg of biotin
daily. The enzyme deficiency can be confirmed in cultured
skin fibroblasts or by mutation analysis. The disorder is
inherited as an autosomal recessive trait.
Both biotinidase deficiency, ascertained by direct
enzyme assay, and holocarboxylase synthetase deficiency
are detected in the expanded newborn screening panels.
The outcomes should alter because of this earlier diagnosis.
GLUTARIC ACIDEMIA TYPE 1
Glutaryl–Coenzyme A Dehydrogenase
Deficiency
An isolated deficiency of glutaryl-CoA dehydrogenase
causes glutaric acidemia type 1 (GA-1) (Goodman and
Frerman, 2001). Multiple phenotypes are known. In the
most dramatic presentation, which accounts for fewer than
half of the known patients with GA-1, the illness develops
acutely in the first year of life, usually after an infection or
other catabolic event. Acute encephalopathy is followed by
the development of what appears to be a severe form of
extrapyramidal cerebral palsy (Hoffmann et al, 1991). The
affected infants have incurred bilateral damage to the caudate and putamen in the basal ganglia, resulting in an incapacitating dystonic syndrome. Some patients have a slowly
progressive course with developmental delay, hypotonia,
dystonia, and dyskinesia in the first 2 years of life. Other
patients are relatively asymptomatic.
In general, GA-1 is not a disorder that is associated
with acute disease in the newborn period. However, macrocephaly at birth is common (Goodman and Frerman,
2001). The etiology of any of the CNS lesions is unknown,
but toxicity caused by dicarboxylic acid accumulation has
been postulated. Magnetic resonance imaging (MRI) of
the head typically shows bilateral widening of the Sylvian
fissures associated with hypoopercularization, resulting in
the “bat-wing” appearance. Sometimes the fluid accumulation mimics subdural hygromas, and a few patients have
230
PART VI Metabolic and Endocrine Disorders of the Newborn
been noted to have subdural hematomas. The congenital
nature of these findings suggests that GA-1 has its onset
in utero and affects CNS development. Usually infants
do not demonstrate bilateral damage to the basal ganglia
in the newborn period. Diagnosis depends on the demonstration of glutaric acid and 3-hydroxyglutaric acid in
urine and is confirmed by the demonstration of deficiency
of glutaryl-CoA dehydrogenase activity or protein levels
on Western blot analysis in cultured fibroblasts, or more
commonly by mutation analysis.
Newborn screening may be complicated in this disorder. Whereas the majority of patients have positive
findings of elevated C5DC in the newborn acylcarnitine
profile, a minority with severe disease may have normal or
near normal results. These same patients might not show
the characteristic elevation of 3-hydroxyglutarate in urine.
For this reason an initial elevation of C5DC in urine can
and should elicit the close attention of a metabolic specialist who then must help to interpret results of the follow-up
testing.
A rare disorder, GA-1 is especially common in SaulteauxOjibway Indians of Canada (Greenberg et al, 1995) and in
the Old-Order Amish of Lancaster County, Pennsylvania
(Morton et al, 1991). A low-protein diet can help in the
treatment of these patients; acute illnesses, usually viral in
nature, should be treated vigorously with fluids containing glucose and adequate amounts of calories to prevent
or diminish catabolism. Bicarbonate may be necessary to
correct acid-base imbalance. Carnitine has also been used
to reduce mitochondrial glutaryl-CoA levels.
FATTY ACID OXIDATION DISORDERS
Glutaric Acidemia Type 2 or Multiple Acyl
Coenzyme A Dehydrogenase Deficiency
Glutaric acidemia type 2 (GA-2) is characterized by deficiency of multiple acyl-CoA dehydrogenase enzymes
(Goodman and Frerman, 2001; Stanley et al, 2006). All
these dehydrogenase proteins have in common the binding of a protein called electron transfer flavoprotein (ETF).
This protein is responsible for accepting electrons in any
of these oxidative dehydrogenation reactions from the flavin adenine dinucleotide (FAD) cofactor. A mutation in
any of the three genes that encode the protein subunits of
either ETF or the ETF dehydrogenase, which is responsible for further transferring the electrons from ETF to
coenzyme Q10 within the mitochondria leads to this condition. Deficiency of riboflavin, a component of FAD, or
its transport may also lead to this condition. Among the
compounds accumulating in this condition are short-,
medium-, and long-chain fatty acids, glutaric and isovaleric acids, dimethylglycine, and sarcosine.
The three major phenotypes of GA-2 are (1) a newbornonset type with congenital anomalies, (2) a newborn-onset
type without anomalies, and (3) a milder or later-onset
type, sometimes called mild acyl-CoA dehydrogenase deficiency or ethylmalonic adipic aciduria.
Severely affected newborn patients with GA-2 often
have multiple malformations, may be premature, and
are usually ill within the first week of life (Frerman and
Goodman, 2001). The patients demonstrate hypotonia,
encephalopathy, hepatomegaly, hypoglycemia, and metabolic acidosis; often the odor of isovaleric acid is present.
There may be facial dysmorphism consisting of a high
forehead, low-set ears, hypertelorism, and a hypoplastic
midface. The kidneys may be palpably enlarged, associated
with large renal cysts; they may also have rocker-bottom
feet, muscular defects of the inferior abdominal wall, and
anomalies of the external genitalia, including hypospadias
and chordee. Most of the patients with GA-2 and multiple
malformations do not survive the first weeks of life. In
some, the malformations are not so noticeable, and only
renal cysts are identified at autopsy. Some of these patients
have cardiomyopathy. In contrast, some of these infants
can survive the newborn period.
The phenotype in the third form of GA-2 is highly variable. Some patients are relatively free of disease and have
intermittent episodes of vomiting, dehydration, hypoglycemia, and acidosis during childhood or adult life. In some,
there may be hepatomegaly and muscle disease.
Laboratory studies in patients with the newborn-onset
type usually show severe metabolic acidosis with lactate elevation and increased anion gap, mild to moderate
hyperammonemia, and hypoglycemia without a moderate or large ketonuria. The liver function and dysfunction test results may be abnormal, with increases in serum
transaminases and prolongations of PT and PTT. A chest
radiograph may show heart enlargement because of hypertrophic cardiomyopathy. Abdominal ultrasonography or
computed tomography may reveal renal cysts. The diagnosis is made from the characteristic pattern of organic
acid metabolites on urine GC-MS analysis. It consists of
glutarate; ethylmalonate; 3-hydroxyisovalerate; 2-hydroxyglutarate; 5-hydroxyhexanoate; adipic, suberic, sebacic,
and dodecanedioic acids; isovalerylglycine; isobutyrylglycine; and 2-methylbutyrylglycine. The ketones, acetoacetic acid, and 3-hydroxybutyric acid are usually not present
or are only minimally elevated if present, being inappropriate for the degree of fatty acid metabolites that indicate
free fatty acid mobilization and the potential for enhanced
ketogenesis. The renal cystic disease may also be associated
with evidence of impaired renal tubular function. Generalized aminoaciduria may be present. The amino-containing
compound sarcosine may be elevated in serum as well as in
urine. There may be secondary carnitine deficiency, and
abnormal carnitine esters such as isovalerylcarnitine and
butyrylcarnitine can be detected in blood. The ETF and
ETF dehydrogenase deficiency can be detected through
the use of specific antibodies on Western blot analysis
of cultured skin fibroblasts or of functional assays in skin
cells. Mutation analysis in this condition as in others may
be superseding the more cumbersome enzyme studies.
Prenatal diagnosis can be achieved by demonstration of
glutarate in amniotic fluid, from results of dehydrogenase
assays in cultured amniocytes, or by mutation analysis.
Despite much debate, criteria for newborn screening have
not been well established, and many false positive tests
have occurred for this rare disorder.
Although treatment with intravenous glucose, riboflavin, carnitine, and diets low in protein and fat generally
have not been successful for catastrophically ill newborns,
there has been some success in patients with milder or lateronset disease. Riboflavin is suggested to be administered to
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
the newborn with severe disease, but riboflavin at a dose of
100 to 300 mg/day has been effective in only a few older
patients (Gregersen et al, 1982; Harpey et al, 1983). The
rationale for this therapy is that riboflavin, being a precursor of FAD, increases the concentrations of FAD and
allows for better interaction with mutated and defective
ETF or ETF dehydrogenase proteins. Finally, some artificial electron acceptors such as methylene blue have been
used in the newborn period without success.
VERY-LONG-CHAIN, MEDIUM-CHAIN,
AND SHORT-CHAIN ACYLFATTY ACID
OXIDATION DEFECTS
Fatty acids are oxidized in a complex process by which
they are taken up into the cell and transported into the
mitochondrion for beta oxidation and energy production.
There are at least 31 enzymes involved in the process and
defects in the majority have been described. Of these many
disorders, the most frequent are those of carnitine metabolism and transport, and various steps in the beta oxidation
pathways. The presentations are sufficiently different to
bear independent description, but in general they all have
hypoketotic hypoglycemia and energy deficits. Most spare
the CNS.
When severe, the defects have been associated with
coma, hypoglycemia, liver disease, cardiac, and skeletal
myopathy (Roe and Ding, 2001; Stanley et al, 2006). All
of these defects involve abnormalities in enzymes that participate in or facilitate the mitochondrial beta oxidation
of fatty acids. In general the pathophysiology associated
with these disorders has the potential to put the patient
in a life-threatening condition in which there is a state of
catabolism and enhanced liberation of free fatty acids by
adipose stores. The most prevalent disorders will be discussed here.
Medium-Chain Acyl-Coenzyme A
Dehydrogenase Deficiency
The most common of these disorders is the medium-chain
acyl-CoA dehydrogenase (MCAD) deficiency, with a frequency of approximately 1 in 20,000 births in Northern
European populations (Roe and Ding, 2001; Stanley et al,
2006). Most patients’ disease does not manifest until late
infancy, but some with the most severe enzyme deficiencies
manifest in the first days of life and may die without explanation, something recently appreciated with the advent of
expanded newborn screening. It is also a well-known cause
of sudden infant death syndrome having been found in 2%
to 3% of cases in which it has been investigated. The typical
patient is an older infant who, after an infection, experiences
anorexia, vomiting, dehydration, lethargy, and hypoglycemia that may be associated with seizures. Similarly, older
patients have features that mimic those of Reye syndrome
and can die because of brain edema.
In MCAD deficiency, the initial episode is associated
with a high mortality rate. If not diagnosed by newborn
screening, the laboratory studies usually show hypoglycemia and an absence of moderate to large ketones in urine
that would be expected to accompany hypoglycemia. The
plasma ammonium values may be mildly elevated, the liver
231
may be enlarged, and serum ALT and AST levels may be
slightly increased. During an acute episode, urine GC-MS
analysis of organic acids characteristically shows increased
levels of adipic, suberic, and sebacic acids as well as their
unsaturated and hydroxylated analogues. However, this
result is not pathognomonic of a disorder of fat metabolism, because infants who are fed medium-chain triglyceride–enriched formulas also show dicarboxylic aciduria.
Acylcarnitine studies of plasma from patients with MCAD
deficiency demonstrates increased levels of C8, C6, and
C10 species. Urine also contains glycine conjugates such
as suberylglycine and hexanoylglycine. A secondary carnitine deficiency can be present. The MCAD enzyme can be
assayed in cultured skin fibroblasts, but mutation analysis
is the more common means of confirming the diagnosis.
After the MCAD gene was cloned and sequenced, a large
number of more common and private mutations were identified. The most conspicuous gene is R329E, a highly prevalent mutation in people of Northern European ancestry
and known to cause a severe enzyme deficiency. The major
treatment is avoidance of fasting for more than 12 hours,
especially in association with an intercurrent illness. Some
practitioners are more conservative and advocate fasting of
no more than 4 to 6 hours in the first year of life, whether
sick or well. Although not of proven efficacy, many physicians recommend a moderately fat-restricted diet for these
patients. It is thought that once the disease is diagnosed
and preventive programs are instituted, the disease should
not be life threatening or fatal.
With the advent of newborn screening, a larger group of
patients has been found and some more common mutations
in these patients have never been seen in a case ascertained
because of symptoms. Finding these individuals has given rise
to the notion that many patients found by newborn screening are destined never to become symptomatic. Whereas
ascertainment of such mutations can be reassuring, too little
is known about them to allow complete reassurance and full
relaxation of preventive precautions. Newborn screening
also finds individuals heterozygous for a severe mutation
when no other mutation is found. These patients are considered to be biochemically manifesting heterozygotes and are
thought to be at no medical risk from this condition.
Very-Long-Chain Acyl-Coenzyme A
Dehydrogenase Deficiency
Most of the patients with a previous diagnosis of long-chain
acyl-CoA dehydrogenase (LCAD) deficiency (Roe and
Ding, 2001; Stanley et al, 2006) actually had very-long-chain
acyl-CoA dehydrogenase (VLCAD) deficiency (Yamaguchi et al, 1993). Patients with this disease may be ill in the
newborn period because of liver disease with hypoglycemia,
cardiomyopathy, and skeletal myopathy. The membranebound VLCAD—as opposed to the soluble LCAD, whose
specific metabolic role is unknown—is the main enzyme for
initiating oxidation of free fatty acids that are derived from
adipose stores, such as palmitic, stearic, and oleic acids. The
fasting state may include coma. In its absence, however, the
patients may exhibit hypotonia, hepatomegaly, and cardiomegaly. With an acute metabolic decompensation, urine
organic acid analysis may demonstrate dicarboxylic aciduria. There may be a secondary carnitine deficiency with
232
PART VI Metabolic and Endocrine Disorders of the Newborn
increased concentrations of long-chain fatty acids (LCFAs)
bound to carnitine.
VLCAD deficiency can be fatal; sudden death in early
infancy has been reported. Therapy is directed toward
replenishment of glucose, calorie administration, and treatment of any potential brain edema. The myopathy and cardiomyopathy, however, may proceed even in the absence
of fasting. Most investigators suggest a reduction in dietary
fat intake, supplementation in medium-chain triglycerides,
and maintenance of an adequate intake of essential fatty
acids. Diagnosis can be made through enzyme assay in cultured skin fibroblasts, but commonly by mutation analysis.
Expanded newborn screening has expanded our horizons in this disorder as it has for MCAD deficiency. In
addition to patients who may be homozygous for two mild
mutations or compound heterozygous for a mild mutation,
and who are at much reduced clinical risk, heterozygotes
for a pathologic mutation and a wild type gene have been
found and are presumably at no greater risk than others in
the population.
Short-Chain Acyl-Coenzyme A
Dehydrogenase Deficiency
The short-chain acyl-CoA dehydrogenase deficiency is
a disorder mired in controversy (Roe and Ding, 2001;
Stanley et al, 2006). Patients with a severe enzyme deficiency are infrequent, and the majority of them have no
symptoms attributable to the disorder, as judged by the
variability of symptoms in those ascertained symptomatically and the absence of obvious disease in similarly affected
siblings. The situation is further complicated by extensive
mutation analysis, especially in those ascertained through
positive newborn screens. Because of the prevalence of
common polymorphisms that impair but do not eliminate
enzymatic activity, the majority of ascertained individuals
have no symptoms and appear unlikely to develop them.
This problem has led some people to declare the disorder harmless and to pay no attention to it. Other more
sober voices accept it as a benign disease and disorder in
the majority of patients, but recognize the possibility that
a small minority may become symptomatic and treat them
appropriately by warning the parents to react to unusual
and unexpected symptoms. There tends to be some acrimony at meetings from members of the most extreme
positions on either end of the spectrum.
Acylcarnitine analysis on either newborn screen or later
shows elevated C4 carnitine. Organic acid analysis shows
elevated levels of ethylmalonic and methylsuccinic acids,
with higher levels tending to occur with greater enzyme
deficiencies. Diagnosis can be made by assaying the shortchain acyl-CoA dehydrogenase enzyme in cultured skin
fibroblasts and by a fatty acid oxidation profile in these same
cells. Mutation analysis is now the norm for this condition.
Long-Chain 3-Hydroxy Acyl-Coenzyme A
Dehydrogenase Deficiency
The long-chain 3-hydroxy acyl-CoA dehydrogenase deficiency is associated with acute illness, fasting-induced
hypoglycemia, hypoketosis, cardiomegaly, and muscle
weakness (Roe and Ding, 2001; Stanley et al, 2006).
As with VLCFA oxidative abnormalities, some older
patients may have episodes of illness associated with elevated serum creatine phosphokinase levels and myoglobinuria. A few patients have had sensory motor neuropathy
and pigmentary retinopathy. Half the patients do not
survive. Some patients may have severe liver disease with
fibrosis in addition to necrosis and steatosis. Women who
are carriers for this disease may also manifest the HELLP
syndrome when carrying an affected child.
The diagnosis was made in symptomatic individuals and
was suggested by symptoms and demonstration of longer
chain 3-hydroxydicarboxylic acids on urine organic acid
analysis. Enzymatic diagnosis can be made in lymphocytes or in skin fibroblasts. More commonly the diagnosis is confirmed by a combination of clinical biochemical
abnormalities and DNA mutation analysis. The majority
of moderate to severe cases are diagnosed by expanded
newborn screening and follow-up.
Treatment of this disorder has involved frequent highcarbohydrate feedings, dietary fat restriction, and supplementation with uncooked cornstarch. Administration of
medium-chain triglycerides may be helpful. Carnitine and
riboflavin have also been tried without benefit. A high level
of parental vigilance is required to begin therapy quickly
at signs of metabolic decompensation. Liver and neurologic disease can progress despite any intervention. The
outcome of severely affected patients is guarded.
Carnitine Transporter Defect
and Deficiencies of Carnitine
Palmitoyltransferase I and II
and Acylcarnitine Translocase
Carnitine is essential for fatty acid oxidation because transport of LCFAs into mitochondria depends on an adequate
amount of carnitine and the presence of two enzymes that
covalently link carnitine to fatty acid or remove the linkage
and one transporter that carries it across the inner mitochondrial membrane (Roe and Ding, 2001; Stanley et al,
2006). These enzymes are carnitine palmitoyltransferase
I and II and a carnitine translocase. Cellular levels of carnitine in turn depend on a sodium-dependent carnitine
transporter.
Primary carnitine deficiency appears to be associated
with a carnitine transporter defect (Roe and Ding, 2001;
Stanley et al, 2006). Patients with the transporter defect
may present with symptoms in infancy or in childhood, but
rarely in the newborn period. Earliest reports concern the
extended newborn period or early infancy. The disease is
characterized by hypoketotic hypoglycemia, hyperammonemia, elevations of transaminases, cardiomyopathy, and
skeletal muscle weakness. In some of the older patients,
cardiomyopathy may be the presenting sign. The characteristic laboratory finding in this disease is extremely low
plasma carnitine levels. The total carnitine levels are usually less than 10 μmol/L in plasma. A dicarboxylic aciduria
is not usually evident on urine organic acid analysis.
Newborn screening has altered our view of this disorder.
Most cases are ascertained with low carnitine on screening
and are usually diagnosed with mutation analysis and the
failure of carnitine levels to rise in the postnatal period.
Most physicians treat the disorder with carnitine to raise
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
plasma carnitine levels while the evaluation is in progress.
Newborn screening has also ascertained asymptomatic and
affected mothers and has altered our perception of this
condition and its severity. In addition, largely asymptomatic mothers affected with an organic acidemia and low
carnitine levels in the mother and fetus have been detected.
This disorder is the only one in which pharmacologic
administration of carnitine has dramatic effects on the
clinical and laboratory abnormalities. The treatment is
100 to 200 mg/kg/day of l-carnitine. Repletion of plasma
carnitine levels is the benchmark by which the efficacy of
treatment is judged, but the degree of repletion of tissue
levels (skeletal muscle and heart) is inferred but rarely
demonstrated. Treatment is successful in the short, intermediate, and longer terms, but no information on the lifespan of these patients who appear healthy is available.
Carnitine Palmitoyltransferase Type I
Deficiency
Carnitine palmitoyltransferase type I (CPT-I) is responsible for covalently linking LCFAs such as palmitate to
carnitine. Although one patient with deficiency of CPT-I
came to attention in the newborn period, most come to
attention in early to late infancy (Roe and Ding, 2001;
Stanley et al, 2006). The clinical findings are hypoketotic
hypoglycemia, encephalopathy, and hepatomegaly; there is
usually no evidence of cardiomyopathy or skeletal myopathy in CPT-I deficiency. Renal tubular acidosis, which
is caused by impaired distal hydrogen ion secretion, has
rarely been reported. Characteristic laboratory findings
are the absence of dicarboxylic aciduria and high plasma
levels of total carnitine and free carnitine; however, the
plasma acylcarnitine profile is not abnormal. The definitive diagnosis rests on measuring CPT-I enzyme activity
in cultured skin fibroblasts or mutation analysis. Frequent
feeding, reduction of dietary fat, supplementation with
medium-chain triglycerides, and avoidance of fasting all
have been beneficial in the long-term management of
patients with CPT-I deficiency. These patients are now
often found by expanded newborn screening because of an
abnormal ratio of carnitine to the carnitylated long-chain
fatty acids.
Acylcarnitine Translocase Deficiency
Acylcarnitine translocase deficiency is an exceedingly
rare defect. It was initially reported in a male infant
who suffered a cardiac arrest at 36 hours of age in association with fasting stress and ventricular dysrhythmias
(Roe and Ding, 2001; Stanley et al, 2006). Because of
the failure to transport long-chain acylcarnitines across
the intermitochondrial membrane after the synthesis by
CPT-I, the patient had very low total plasma levels of carnitine, most of which was long-chain esterified carnitine;
he also had recurrent episodes of hypoglycemia, vomiting,
gastroesophageal reflux, and mild chronic hyperammonemia as well as severe skeletal myopathy and mild hypertrophic cardiomyopathy. The continuous nasogastric
feeding of a low-fat, high-carbohydrate formula failed to
normalize clinical abnormalities, and the patient died at 3
years of age. At that time, liver failure had also developed.
Pathophysiologic findings in this patient suggested that
accumulation of long-chain acylcarnitine species may be
233
toxic for several organs, including heart, liver, and skeletal
muscle. In addition, the acute development of ventricular
dysrhythmias may be related to accumulation of such species in cardiac tissue. It is often considered to be almost
invariably fatal or very serious, but the probability is that
more mild cases with reduced but not absent transporter
activity have not been found.
Carnitine Palmitoyltransferase
Type II Deficiency
There are two phenotypes of carnitine palmitoyltransferase type II (CPT-II) deficiency (Roe and Ding, 2001;
Stanley et al, 2006). The enzyme is responsible for the
hydrolysis of LCFA bound to carnitine after transport
across the intermitochondrial membrane. Most of the
patients reported with CPT-II deficiency have a mild deficiency of the enzyme and in adulthood exhibit episodes
of muscle weakness and myoglobinuria brought on by
prolonged exercise. The other phenotype is caused by a
more serious enzyme defect and manifests in early infancy.
The first detailed report was of a 3-month-old boy with
hypoketotic hypoglycemia, coma, seizures, hepatomegaly, cardiomegaly, cardiac arrhythmias associated with an
increase in long-chain acylcarnitine levels in tissues, and
the absence of urinary dicarboxylic aciduria. The enzyme
is also expressed in renal tissue and skeletal muscle. Renal
dysgenesis has been noted in three patients (Zinn et al,
1991). Reports of onset in the newborn period associated
with an abnormal physical appearance and MRI (Hug
et al, 1991) and in later infancy have been recorded.
Several mutations have been described. Decreased activity
of the CPT-II enzyme may be demonstrated in cultured
skin fibroblasts.
DEFECTS IN KETONE METABOLISM
After LCFAs are broken down first to medium-chain
and finally to short-chain fatty acids such as acetoacetyl
CoA, they must be converted in the liver to 3-hydroxy3-methylglutaryl CoA (HMG-CoA) before hydrolysis
to acetoacetate, the ketone body used by the body for
energy. Depending on the mitochondrial redox potential—that is, the ratio of the reduced form of nicotinamide adenine dinucleotide (NAD) to its oxidized form
(NADH/NAD+)—some of the acetoacetate is converted
to 3-hydroxybutyrate, and both ketone bodies are transported out of liver mitochondria and hepatocytes into
blood, where they may be used by other tissues, especially brain. Acetoacetyl CoA derived from the last turn of
the beta oxidation spiral together with acetyl CoA forms
HMG-CoA in a reaction catalyzed by HMG-CoA synthetase. Normally, acetoacetyl CoA can also be hydrolyzed
to acetyl CoA by the mitochondrial acetoacetyl-CoA
thiolase. Patients with a deficiency of this thiolase do not
have a defect in ketone body synthesis but, rather, metabolic acidosis associated with excess ketosis (Mitchell and
Fukao, 2008; Stanley et al, 2006).
The clinical features of the thiolase deficiency are variable. Severe acute metabolic decompensation has been
reported in infants, but there are also asymptomatic adults
with the disorder. The episodes are heralded by fasting or
increased protein intake, because isoleucine is a precursor
234
PART VI Metabolic and Endocrine Disorders of the Newborn
of 2-methylacetoacetyl CoA, which is also a substrate
for the mitochondrial acetoacetyl-CoA thiolase enzyme.
Therefore this block leads to a defect in distal catabolism
of isoleucine and in processing of the precursor of ketone
body formation—namely, acetoacetyl-CoA. Cardiomyopathy has been identified in rare patients. The characteristic
urinary metabolite pattern detected on urine organic acid
analysis is the presence of isoleucine metabolites, such as
2-methylacetoacetate, 2-methyl-3-hydroxybutyrate, and
triglylglycine. During acute decompensation, lactate
and the traditional ketone bodies are detected in excess
amounts in the urine. Some older children have been mistakenly identified as having ketotic hypoglycemia because
glycine may be elevated in plasma. Deficiency in the mitochondrial acetoacetyl-CoA thiolase can be demonstrated in
cultured skin fibroblasts and by mutation analysis. Treatment of acute episodes consists of intravenous glucose and
administration of alkali to correct metabolic acidosis, which
may be severe. Long-term therapy involves mild protein
restriction, avoidance of fasting, and prompt attention to
any intercurrent illness or development of ketonuria.
The synthesis of acetoacetate from HMG-CoA depends
on the HMG-CoA lyase enzyme. A deficiency of this
enzyme represents the most profound defect in ketone body
synthesis (Roe and Ding, 2001; Stanley et al, 2006). Approximately one third of patients with this disease are in the first
week of life. In this subgroup of patients, the onset is dramatic and the disease is catastrophic, being characterized by
vomiting, lethargy, coma, seizures, hepatomegaly, hypoglycemia, little or no ketones in urine, and hyperammonemia.
Most of the complications are related to severe effects of
hypoglycemia on the CNS in addition to acidemia, which
may be profound. The characteristic urine metabolites
detected by GC-MS analysis are 3-hydroxy-3-methylglutaric acid, 3-methylglutaconic acid, and 3-hydroxyisovaleric
acid. Only small, inappropriate amounts of acetoacetic acid
and 3-hydroxybutyric acid may be detected. Lactate values
may be elevated during the acute metabolic decompensation. Inherited as an autosomal recessive trait, the HMGCoA lyase deficiency can be demonstrated in cultured skin
fibroblasts or by mutation analysis. Treatment of the acute
episode consists of administration of intravenous glucose
and alkali to correct metabolic acidosis. Most long-term
therapy consists of a high-carbohydrate diet. Some patients
treated with protein-restricted diets, but the most important element in long-term care is the avoidance of fasting.
The last defect to be discussed in the area of disturbances
in ketone body metabolism is the succinyl-CoA 3-ketoacidCoA transferase deficiency (Mitchell and Fukao, 2008;
Stanley et al, 2006). In this disease, the ketone bodies, acetoacetic acid, and 3-hydroxybutyric acid are synthesized
adequately in the liver, but they cannot be metabolized
in the extrahepatic tissues because of the failure of activation of acetoacetate to acetoacetyl CoA by the transferase enzyme. Conversion to a CoA derivative is required
for hydrolysis to acetyl CoA for final metabolism in the
Krebs citric acid cycle. This disorder is rare, and most of
the affected patients in the newborn period do not survive.
Such patients usually exhibit severe ketosis and lactic acidosis. The hallmark of the disease is persistent ketosis, even
after correction of overt metabolic acidosis and the institution of frequent feedings with the avoidance of fasting.
Plasma values of acetoacetate and 3-hydroxybutyrate are
always mildly to moderately elevated, resulting in intermittent ketonuria. The most important aspect of therapy is
the avoidance of fasting, during which acidosis and ketosis
can be overwhelming. The gene that encodes this enzyme
has been cloned, and several mutations have been detected.
PRIMARY LACTIC ACIDOSIS
The term congenital lactic acidosis (CLA) refers to a group of
diseases in which impaired lactate metabolism is caused by a
defect in the mitochondrial respiratory or electron transport
chain (ETC), or the tricarboxylic acid (TCA; Krebs) cycle,
in which there is a primary defect in pyruvate metabolism
that secondarily leads to impaired lactate handling (see Figure 22-1) (De Meirleir et al, 2006; Munnich, 2006; Munnich
et al, 2001; Robinson, 2001). However, there are a limited
number of reports on inborn errors of the TCA cycle. Most
of the information on CLA focuses on mitochondrial ETC,
pyruvate dehydrogenase (PDH) complex, and pyruvate carboxylase (PC) defects, all of which have been reviewed (De
Meirleir et al, 2006; Munnich, 2006; Munnich et al, 2001;
Robinson, 2001) with more detailed information on conditions that must be treated more superficially in this chapter.
Some patients with CLA present have overwhelming
lactic acidosis in the neonatal period. In others, lactate
may be elevated only in CSF, a “cerebral” lactic acidosis
syndrome and present more indolently. Depending on the
nature of the enzyme deficiency, lactate, pyruvate, and alanine levels can be elevated in the blood. The ratio of blood
lactate to pyruvate (L:P ratio) can be helpful in distinguishing the different types of inborn errors. For example, the
L:P ratio is often normal (10 to 20) in PDH complex and
modestly elevated in PC deficiencies, but can be greatly
elevated in an ETC defect. Unlike most of the disorders
previously considered in this chapter, these conditions are
not detectable by newborn screening and become apparent
only when a patient’s symptoms are recognized.
Pyruvate Dehydrogenase Deficiency
Pyruvate Dehydrogenase Complex Deficiency
PDH is a complex of three primary enzymes— plus a
phosphatase, a kinase, and at least one other element of
less known function—that combine as a multimer with a
very large molecular weight and a number of copies of each
enzyme. The first enzymatic step is a decarboxylation reaction catalyzed by a heterodimeric system consisting of the
E-1α subunit, encoded by a gene on the X-chromosome,
and E-1β, which is autosomally encoded as are all the other
subunits in this complex. Defects in all the known genes
have been reported, but mutations in the X-linked E-1α
outnumber all others by far and may represent as much as
25% of known causes in patients with CLA.
Severe PDH deficiency sometimes manifests in the neonatal period with profound lactic acidosis, elevated blood
lactate and pyruvate, elevated plasma alanine, and congenital anomalies of the brain noted on MRI, including absent
or underdeveloped corpus callosum, heterotopic migration deficits, and a somewhat typical dysmorphic appearance. Typically, the L:P ratio is normal and distinguishes
it from disorders of the mitochondrial respiratory chain.
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
The patients are hypotonic and respirator dependent and
have a poor prognosis. The diagnosis is rarely known
immediately, and the standard intravenous support of high
glucose exacerbates the metabolic acidosis and worsens the
outlook for the patient. The diagnosis is inferred when all
the clinical biochemical data are collated and can be confirmed by an enzymatic deficiency in lymphocytes or cultured skin fibroblasts or by mutation analysis of the E-1α
gene in particular. The enzyme assay is difficult in the
most experienced hands, and it is difficult to understand
the high residual activity that is often recorded.
The majority of patients are more indolent on clinical
presentation, with developmental delay that may resemble
Leigh syndrome, and they may have a modest elevation of
lactate, with pyruvate being the most telling biochemical
marker of the disease. This group of patients often respond
well biochemically to a high-fat and low-carbohydrate
diet. Fat, as acetyl CoA, enters the energy pathway after
the block, whereas glucose must traverse the PDH reaction to provide all but minimal energy generation.
Other defects of the PDH complex including two
other subunits, the activating and deactivating enzymes,
and a subunit X of unknown function are rare and usually result in chronic psychomotor retardation syndrome
in late infancy and childhood. The E3 subunit defect
causes a unique syndrome, because the subunit is important in the PDH complex, the BCKAD complex, and the
α-ketoglutarate dehydrogenase complex. Therefore these
patients have multiple deficiencies involving the BCAA
as in MSUD, as well as Krebs cycle metabolites that are
indicative of a block in the TCA cycle. Most of the patients
are later than the newborn period and have severe progressive neurodegenerative disease. The key laboratory
findings are elevations of lactic acid in blood, BCAAs in
plasma, and detection of α-ketoglutarate in urine by urine
organic acid analysis. PDH phosphatase deficiency is a
rare cause of congenital lactic acidosis. Other than the E3
deficiency, other defects in PDH are responsive, at least
biochemically to the high-fat, low-carbohydrate diet.
Pyruvate Carboxylase Deficiency
Pyruvate carboxylase is an enzyme that is involved in gluconeogenesis and that adds bicarbonate to pyruvate to
form oxaloacetate, a compound also involved in replenishing intermediates of the tricarboxylic acid cycle. There
are two main types of PC deficiency (De Meirleir et al,
2006; Robinson, 2001). Type A is characterized by lactic
acidosis in the newborn period and delayed development.
However, the disease is of a chronic nature. In type B,
the catastrophic form of the disorder, the infant is acutely
ill, usually in the first week of life, with encephalopathy,
severe metabolic acidosis with lactic acidosis, and hyperammonemia (De Meirleir et al, 2006; Munnich et al, 2001;
Robinson, 2001). The mortality rate in this form is high.
As discussed earlier, PC is a biotin-containing enzyme.
Most of the patients with a type B form of PC deficiency
have been of French or English origin. Unlike patients
with a type A defect, in whom the blood L:P ratio is normal because both lactate and pyruvate are comparably elevated, patients with the type B defect often have an elevated
L:P ratio. Because of the importance of the PC product
235
oxaloacetate in providing adequate cellular levels of aspartate, citrulline metabolism in the urea cycle is defective,
leading to elevations of plasma citrulline and plasma ammonium concentrations. Although PC is also an important
enzyme in gluconeogenesis, hypoglycemia has not been
commonly reported. The liver may be enlarged. There is
no effective treatment for PC deficiency when it is associated with progressive neurodegeneration. The gene that
encodes the PC subunits, of which four combine to make an
active enzyme, has been cloned and sequenced. An expanding number of mutations have been identified and provide a
basis for prenatal diagnosis. PC deficiency may be detected
in cultured skin fibroblasts or in liver biopsy samples.
Phosphoenolpyruvate Carboxykinase
Deficiency
Phosphoenolpyruvate carboxykinase enzyme also functions
in gluconeogenesis, and there are two forms in liver, one
in the cytosol and the other in the mitochondrial compartment. It is an exceedingly rare disorder. Patients do not
usually come to attention until childhood with hypotonia,
failure to thrive, hepatomegaly, lactic acidosis, and hypoglycemia. Although there is no adequate experience in identifying the optimal therapy for these patients, it is reasonable
to assume that frequent feedings and avoidance of fasting
are important in avoiding severe metabolic imbalance.
Mitochondrial Respiratory Chain
or Electron Transport Chain Defects
Oxidative phosphorylation is the key process performed
by the mitochondria of cells. Any inborn error of metabolism that involves the tightly coupled and regulated process
of mitochondrial energy metabolism may have profound
effects on health and disease, because oxidative phosphorylation is the process by which we convert nutrients into
energy. The various derivatives of the nutrients, such as
pyruvate and fatty acids, are converted to CO2 in mitochondria. The energy derived from such controlled chemical
combustion is harnessed by allowing the reducing equivalents (in the form of NADH or the reduced form of flavin
adenine dinucleotide [FADH2], which are derived from such
metabolism) to combine with oxygen to form water, and in
the process the synthesis of ATP is coupled to the orderly
flow of electrons down the respiratory chain components.
The important components in the mitochondrial respiratory chain are complex 1 (NADH dehydrogenase), complex
2 (ETF dehydrogenase), complex 3 (cytochromes b, c1) and
the terminal complex in this chain, and complex 4, which is
cytochrome c oxidase (COX) (Shoffner, 1995). In addition,
there is a complex 5, or ATP synthetase, and an adenine
nucleotide translocase, which permits transport of adenosine
diphosphate into and ATP out of the mitochondria. Complex 2 is involved primarily in fatty acid oxidation and oxidation of succinate derived from the Krebs cycle, because the
reducing equivalents extracted from fatty acids, glutaric acid,
and succinate flow from ETF into complex 2. The polypeptides that compose these various complexes are derived
from both the nuclear genes and the genes on mitochondrial
DNA (mtDNA). Except for complex 2, mtDNA is important in production of the subunits of all the respiratory chain
236
PART VI Metabolic and Endocrine Disorders of the Newborn
complexes. CLA involving the ETC components has been
associated with both nuclear and mtDNA mutations. In addition to the actual complex components, there are many more
genes responsible for the assembly of various subunits into
the functional complexes, all of which are encoded in nuclear
DNA (nDNA). There are estimates that defects in any one of
more than 500 genes can result in an ETC deficiency.
On the basis of molecular diagnostic testing, the oxidative phosphorylation diseases can be divided into the following four genetic groups (Shoffner, 1995):
ll
ll
ll
ll
Group 1: nDNA mutations
Group 2: mtDNA point mutations
Group 3: mtDNA deletions and duplications
Group 4: unidentified genetic defects
The relationship between phenotype and mtDNA mutations
is not straightforward, probably because of the phenomenon
of heteroplasmy. Mitochondria with their unique mtDNA
are inherited solely from the mother. Random segregation
of mitochondria having mtDNA mutations leads to heteroplasmy and, ultimately, a variable concentration of defective
mitochondria within cells and among tissues (Wallace et al,
1988). Much of our understanding of the detailed molecular
mechanisms that contribute to or produce the ETC gene
disturbances concern the mtDNA mutations, although the
application of molecular methodology and increasingly
inexpensive mass sequencing is allowing more definition
of the nDNA disorders that compose the majority of the
ETC disease seen in newborn and infancy periods. With the
exceptions of the neurogenic muscle weakness, ataxia, and
retinitis pigmentosa (NARP caused predominantly by mutations at position 8993 of the mitochondrial genome), only a
minority of the mtDNA defects actually are known to manifest in the newborn period (Wong, 2007). Other examples
of syndromes caused by mtDNA mutations are the MELAS
(mitochondrial encephalopathy, lactic acidosis, and strokelike—episodes position 3243), MERRF (myoclonic epilepsy
with ragged-red—fiber position A8344G), Leber hereditary
optic neuropathy (Wallace et al, 1988), and sporadic deletion–duplication syndromes such as Pearson syndrome (Di
Donato, 2009). The diseases that affect young infants are
benign infantile mitochondrial myopathy, cardiomyopathy,
or both; lethal infantile mitochondrial disease; lethal infantile cardiomyopathy; subacute necrotizing encephalomyopathy (SNE) or Leigh disease; Pearson syndrome; Alpers’
disease; and the most dramatic form, with presentation often
in the first few days of life, during which an acid-base disturbance dominates the clinical picture (Carrozzo et al, 2007;
Gibson et al, 2008). The hallmarks of mitochondrial disease
are almost always multisystem involvement and unambiguous lactate acidemia or acidosis. Although some debate exists
as to whether lactate elevation is present in all mitochondrial
disorders that present in life, it is a virtual requirement for
such a diagnosis in newborns and early infants.
The nuclear encoded mitochondrial DNA depletion
syndromes, which may include fatal hepatopathy, are
caused by mutations in the SUCLG1, SUCLA2, MPV17,
RRM2B, PEO1, TP, POLG1, DGUOK, and TK2 genes.
The ribosomal translational defects (EFG1 and EFTμ
genes) are the relatively new groups of mitochondrial diseases (Di Donato, 2009; DiMauro and Schon, 2008). Both
are part of the group I genetic disease types.
The diagnostic tools to unravel these disorders have proliferated recently. We assess lactate in the CSF after lumbar
puncture, lactate (among other metabolites) in the brain by
magnetic resonance spectroscopy (MRS) during an MRI,
more readily obtained in a sick newborn than previously, and
DNA technology that has become faster and less expensive
and includes full sequencing of the mitochondrial genome.
Older and still useful technologies such as muscle biopsy
with histologic analysis by light and electron microscopy,
mitochondrial complex assay on either fresh (preferred, but
infrequently available) or flash-frozen tissue and skin fibroblast studies are still available. In fatal cases, a rapid autopsy
and proper preservation of tissue specimens are essential.
Finally, a newer and increasingly frequently diagnosed family of disorders includes mitochondrial depletion syndromes
caused by a number of genetic defects in the mtDNA synthesis apparatus or the availability of nucleotides. A less
frequent, newly recognized family of disorders are the coenzyme Q biosynthetic disorders, diagnosed reliably only by
coenzyme Q10 levels in muscle, and not in plasma.
Although the subject of much debate, there is no credible evidence that mitochondrial disease is treatable, with
the obvious exception of a small minority caused by coenzyme Q deficiency. Given the inability to identify these
patients quickly, it is reasonable to treat acutely ill patients
with presumed mitochondrial disorders with high doses of
coenzyme Q. However, indiscriminant use of this cofactor
is to be discouraged.
Benign Infantile Mitochondrial Myopathy,
Cardiomyopathy, or Both
Benign infantile mitochondrial myopathy is associated
with congenital hypotonia and weakness at birth, feeding
difficulties, respiratory difficulties, and lactic acidosis. In
this poorly understood, developmental-like disorder, only
skeletal muscle appears to be affected, and histochemical
analyses show a COX deficiency that returns to normal
levels after 1 to 3 years of age. An nDNA mutation in a
gene important in a fetal isoform of an ETC polypeptide
specific for muscle oxidative phosphorylation was hypothesized to be the cause of this problem. A developmental
switch from the defective fetal gene to the adult form may
be responsible for the gradual improvement. It was thought
to be the only example of a developmental defect in oxidative phosphorylation that is probably nuclear encoded
and in which the treatment is only supportive during the
early newborn period to prevent death from respiratory
disease. However, a recent study suggests that the etiology
may be a maternally inherited, homoplasmic m.14674T>C
mt_tRNAGlu: mutation (Horvath et al, 2009).
The form also associated with cardiomyopathy may be a
variant of the benign isolated myopathy and involves striated muscle in both skeletal and cardiac muscle. It manifests
in the newborn period with lactic acidosis and a cardiomyopathy that improves during the first year of life. The exact
gene defect is unknown. More attention must be paid to
these two disease entities, because with early optimal medical care, affected infants may have an excellent prognosis.
Lethal Infantile Mitochondrial Disease
Infants with lethal infantile mitochondrial disease are
severely ill in the first few days or weeks of life or in
the extended newborn period. They exhibit hypotonia,
CHAPTER 22 Inborn Errors of Carbohydrate, Ammonia, Amino Acid, and Organic Acid Metabolism
237
CASE STUDY
A boy was born to nonconsanguinous, healthy parents after a full-term gestation.
Biventricular hypertrophy with a predominant right-sided component had been
observed on fetal ultrasonography at 24 weeks’ gestation. He was delivered by
cesarean section because of variable decelerations, and he emerged without
meconium staining or passage. Respiratory distress developed shortly after birth.
An echocardiogram showed concentric right ventricular hypertrophy with elevated
right ventricular pressure (70 mm Hg). Initial laboratory studies showed acute
metabolic acidosis (blood lactate >18 mmol/L; arterial pH, 6.91).
His weight was 2.36 kg (10th percentile), his length was 46 cm (5th percentile),
and his head circumference was 31 cm (5th percentile). He had a right ventricular
heave and an intermittent fourth heart sound. There was no liver enlargement.
Muscle bulk was normal, and deep tendon reflexes were intact. Metabolic
investigations showed persistent arterial lactic acidemia (range, 3 to 11 µmol/L)
with increased pyruvate (range, 0.02 to 0.44 µmol/L). The L:P ratio ranged from
25 to 290. The activities of the respiratory chain complexes II, III, and IV were
normal in cultured skin fibroblasts. A left quadriceps muscle biopsy was used for
histochemical analysis and to prepare a 10% extract for respiratory chain studies.
Histochemical analysis showed no ragged-red fibers with the modified Gomori
trichrome stain, but a diffusely weak response to staining for cytochrome c oxidase (COX, ETC complex 4). Biochemical analysis showed markedly reduced COX
activity, contrasting with normal activities of other complexes. Direct sequencing
of the three COXs and all 22 transfer RNA genes of mtDNA, all the COX-assembly
muscle weakness, failure to thrive, and severe lactic acidosis. Death often occurs by 6 months of age and almost
always is associated with overwhelming lactic acidosis.
Skeletal muscle shows lipid and glycogen accumulation and abnormally shaped mitochondria on electron
microscopic examination. Hepatic dysfunction may be a
prominent finding in these patients. Generalized proximal
renal tubular dysfunction may occur, leading to the renal
Fanconi syndrome. The ETC defects reported in these
patients include defects in complexes 1, 3, and 4. Original
reports concerned infants with a phenotype resembling
severe Werdnig-Hoffman disease with COX deficiency
and renal Fanconi syndrome.
Lethal Infantile Cardiomyopathy
Defects of the mitochondrial respiratory chain have varied
neonatal presentations and are commonly caused by isolated COX deficiency (Shoffner, 1995). Clinical features of
COX deficiency reflect involvement of one or more tissues
and include encephalopathy, myopathy, cardiomyopathy,
liver disease, and nephropathy (Munnich et al, 2001). The
heterogeneous clinical features are due, in part, to the dual
genetic control of COX. The three catalytic subunits—
COX I, COX II, COX III—are encoded by mtDNA,
whereas the remaining subunits (COX IV to COX VIII)
are encoded by nDNA. In addition, the proper assembly of
COX requires several nDNA-encoded proteins. The various syndromes caused by COX deficiency are only partially
understood at the genetic level. Most infants with isolated
complex IV deficiency are likely to have nDNA gene defects
in COX assembly genes, including SURF1, SCO2, COX15,
COX10, SC01, and LRPPRC (Di Mauro and Schon, 2008).
Subacute Necrotizing Encephalomyelopathy
or Leigh Disease
Probably because of a failure to recognize the clinical
signs, infants with SNE or Leigh disease usually come to
clinical attention after the newborn period. This disease is
nuclear genes known to harbor pathogenic mutations (SURF1, SC01, SC02,
COX10, and COX15), and the two other nuclear ancillary genes (COX11 and
COX17 ) showed no mutations. Southern blot analysis showed no deletions, but
results were inconclusive regarding mtDNA depletion. However, sequencing of
the two genes known to be associated with the myopathic (Saada et al, 2001) and
the hepatocerebral (Mandel et al, 2001) mtDNA depletion syndromes (TK2 and
dGK ) did not show any mutations.
A series of echocardiograms documented biventricular, hypertrophic, nonobstructive cardiomyopathy. The patient died on the seventh hospital day after a
sudden episode of hemoglobin desaturation. An autopsy was performed 4 hours
after death. The right ventricle was found to be thickened and enlarged, with
relative sparing of the left side. The pulmonary vasculature of the lungs showed
hypertrophy that extended to the most distal vessels. Routine microscopic findings in skeletal muscle were normal, and the CNS was without microscopic or
macroscopic abnormalities.
This baby had a fatal syndrome defined clinically by prenatal cardiomyopathy
and severe pulmonary hypertension in the newborn period. The syndrome was
caused by isolated ETC complex 4 deficiency. The pathophysiology centered on
the heart and lungs. How much disease was caused by the involvement of other
organs remains unclear. This case is an example of a mitochondrial metabolic disorder, but without a specific molecular genetic cause. A more extensive evaluation
by current methods might or might not have revealed a diagnosis.
characterized as a progressive neurodegenerative disorder
with severe hypotonia, seizures, extrapyramidal movement
disorders, optic atrophy, and defects in automatic ventilation or respiratory control (Finsterer, 2008; Leigh, 1951).
It is clear that there are many causes of SNE. As discussed
earlier, PDH complex deficiency (PDHE1 alpha-subunit
and PDHX1) can lead to Leigh disease. Patients with
defects in the ETC have also been reported to have findings compatible with SNE. The mtDNA mutation causing NARP is an example. Reported nuclear gene defects
include ETC complex I (NDUFS1), complex II (SDHA),
complex IV (SURF1, COX15), coenzyme Q10 biosynthetic
defects, mitochondrial ribosomal translational defects, and
SUCLA2. Many neuropathologists believe that the diagnosis of SNE depends on an analysis of CNS tissue at
autopsy. However, MRI characteristically shows bilateral
symmetrical lesions of the basal ganglia as occurs in other
mitochondrial disorders.
There is no effective treatment for this disease, unless
the cause is a specific inability to synthesize coenzyme Q10.
It is possible that most of the patients with Leigh disease
have disturbances in nuclear-encoded genes. Although,
as discussed later, the NARP lesion caused by a group 2
mtDNA mutation is one important cause in early infancy.
Clearly, this is not one disease entity, because the specific
neuropathologic findings for SNE have also been reported
in a patient with Menkes’ disease, in which there is a secondary ETC complex 4 deficiency, because copper is an
important metal cofactor of COX.
The following clinical findings have been noted in infants
with SNE: optic atrophy, ophthalmoplegia, nystagmus,
respiratory abnormalities, ataxia, hypotonia, spasticity,
seizures, developmental delay, psychomotor retardation,
myopathy, and renal tubular dysfunction. Some patients
may manifest hypertrophic cardiomyopathy, liver dysfunction, and microcephaly. The neuropathologic lesions
include demyelination, gliosis, necrosis, relative neuronal
sparing, and capillary proliferation in specific brain lesions.
238
PART VI Metabolic and Endocrine Disorders of the Newborn
There are lesions of the basal ganglia, which are bilaterally
symmetrical, as well as of the brainstem, cerebellum, and
the cerebral cortex to a lesser degree. Commonly, elevation
in blood lactate is only slight to moderate, as well as intermittent, in this diverse group of patients. In some instances,
lactate values may be elevated only in the CSF. The most
commonly reported biochemical abnormalities are deficiencies in COX, complex 4 NADH dehydrogenase, or complex
1 and PDH. In a few rare patients, the abnormality in oxidative phosphorylation has been reported to be secondary
to NARP mutation. This involves a T:C transition at base
pair 8993 of the adenosine triphosphatase (ATPase) 6 gene,
changing a leucine to a proline at position 156 in the ATPase
6 polypeptide. Investigators have speculated that defects
such as those in COX and the NADH dehydrogenase, when
associated with neuropathology of Leigh disease, are caused
by nuclear gene mutations and not mtDNA gene defects
such as the NARP point mutation in the ATPase 6 gene.
The diseases in group 3 (see earlier), exemplified by the
Kearns-Sayre and chronic progressive external ophthalmoplegia syndromes, are genetic but not familial and are caused
by mtDNA deletions or duplications that are spontaneous
mutations. These disorders do not usually come to clinical
attention in infancy. The only example of such a mutation
manifesting in early infancy is the Pearson syndrome. This
disorder is systemic and primarily affects the hematopoietic
system and pancreas function. The characteristics are severe
macrocytic anemia with varying degrees of neutropenia and
thrombocytopenia. Bone marrow examination shows normal cellularity, but extensive vacuolization of erythroid and
myeloid precursors, hemosiderosis, and ringed sideroblasts.
This disease of the bone marrow can lead to death in infancy.
However, patients who are able to recover or who benefit
from aggressive therapy may demonstrate other signs of this
systemic disorder in late infancy or childhood, such as poor
growth, pancreas dysfunction, mitochondrial myopathy,
lactic acidosis, and progressive neurologic damage.
Barth Syndrome
Barth syndrome is an X-linked disorder associated with
cardiomyopathy, skeletal muscle disease, and neutropenia
(Yen et al, 2008). Skeletal muscle shows abnormal mitochondrial morphology. Important laboratory findings
include decreased plasma-free carnitine, increased urinary
excretion of 3-methylglutaconate on GC-MS analysis of
urine organic acids, and decreased levels of serum cholesterol in early infancy. Positional cloning identified a gene
for this disorder on Xq28 that encodes for a phospholipid
remodeling enzyme, cardiolipin acyl transferase. Several
mutations have been identified. It has been hypothesized
that the organic acid 3-methylglutaconate accumulates
because of defective mitochondrial transport. Patients
must be supported from birth to early infancy. It is possible
that if severe cholesterol deficiency can be avoided, affected
infants may survive and may be relatively free of cardiomyopathy during childhood. Of diagnostic importance, not all
patients with 3-methylglutaconic aciduria have Barth syndrome. A few have isolated leucine-dependent 3-methylglutaconyl-CoA hydratase deficiency or Costeff syndrome,
but most have ill-defined mitochondropathies.
Unidentified Genetic Defects
A number of diseases are believed to be caused by mitochondrial respiratory chain problems, but the specific mutations
remain unknown. These disorders constitute the group 4
mutations, or the disorders of unknown inheritance (Shoffner, 1995). Alpers disease was once one such example. It has
also been called progressive infantile poliodystrophy. Infants and
children with this progressive disease experience progressive cerebral cortical damage, sometimes also involving the
cerebellum, basal ganglia, and brainstem; in some, liver disease may progress to cirrhosis. The neuropathologic lesions
consist of spongiform or microcystic cerebral degeneration,
gliosis, necrosis, and capillary proliferation Seizures are
prominent, including myoclonus. Laboratory abnormalities
include abnormal NADH oxidation or complex 1 defects,
impaired pyruvate handling, PDH complex deficiency,
TCA cycle malfunction, and decreased mitochondrial cytochrome a + a3 content. This disorder is caused by mutations in one or more gene defects associated with mtDNA
depletion, such as the autosomal recessive disease caused by
mutations in the DNA polymerase gamma 1 (POLG1) gene
and can usually be defined by mutation analysis.
Early Lethal Lactic Acidosis
In an unknown fraction of patients with primary disturbances in mitochondrial oxidative phosphorylation or ETC
defects, massive lactic acidosis develops within 24 to 72
hours after birth. Commonly the condition is untreatable,
because it is relentless and unresponsive to alkali therapy.
Dialysis is a remedy but not a cure. Often, affected infants
have no obvious organ damage early in the course or evidence of malformations; this is also true for infants with the
PDH complex deficiency, which is probably a more common cause of overwhelming acidosis in the first week of
life. In addition, acidemia per se can easily cause the coma
or impaired cardiac contractility that may be encountered.
Some infants have survived with aggressive therapy.
The care of babies with these different forms of severe
lactic acidosis almost always brings an ethical dilemma to
the forefront for physicians and nurses of the neonatal
intensive care unit as well as for the babies’ families. To
further complicate the issues, enzymatic and molecular
analyses usually are not immediately available. The disease
in most patients probably remains idiopathic, and no DNA
mutation, nuclear or mitochondrial, will be identified
without extensive, expensive, and extremely inconvenient
and Herculean efforts. A rigid approach to care is impractical and unwise. Decisions regarding management must
be individualized, because the mitochondrial dysfunction
and resultant pathophysiology can vary among infants.
SUGGESTED READINGS
Blau N, Hoffmann GF, Leonard J, et al, editors: Physicians guide to the treatment and
follow-up of metabolic diseases, Heidelberg, 2006, Springer.
Fernandes J, Saudubray JM, Van den Berghe G, et al: Inborn Metabolic diseases:
diagnosis and treatment, Heidelberg, 2006, Springer.
Scriver CR, Beaudet AL, Sly WS, et al: The metabolic and molecular bases of inherited
disease. ed 8, New York, 2001, McGraw-Hill. Updated material is available
online at www.ommbid.com.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
23
Lysosomal Storage, Peroxisomal,
and Glycosylation Disorders and SmithLemli-Opitz Syndrome in the Neonate
Janet A. Thomas, Carol L. Greene, and Gerard T. Berry
Lysosomal storage diseases (LSDs), peroxisomal disorders, congenital disorders of glycosylation (CDGs), and
Smith-Lemli-Opitz (SLO) syndrome are single-gene disorders, most of which demonstrate autosomal recessive
inheritance. The combined incidence of LSDs has been
reported to be 1 in 1500 to 8000 live births in the United
States, Europe, and Australia (Fletcher, 2006; Meikle et al,
2006; Staretz-Chacham et al, 2009; Stone and Sidransky,
1999; Wenger et al, 2003; Winchester et al, 2000). The
incidence of peroxisomal disorders is estimated to be
more than 1 in 20,000. The most current estimate for
SLO syndrome is 1 in 20,000, and a similar frequency of
1 in 20,000 is estimated for the congenital disorders of
glycosylation.
These four categories of metabolic diseases involve molecules important in cell membranes and share overlapping
clinical presentations. Clinical presentations are heterogeneous, with a broad range of age at presentation and severity of symptoms. All are chronic and progressive. Age of
onset varies from prenatal to adulthood, and severity can
range from severe disability and early death to nearly normal lifestyle and life span. For each condition, interfamilial variability is greater than intrafamilial variability. The
genetic and clinical characteristics of conditions in these
categories that can manifest in the neonatal period (except
Pompe disease, which is addressed in Chapter 22) are also
summarized in Tables 23-1 to 23-3.
Important presentations that should lead the neonatologist to consider these disorders in the differential diagnosis
are as follows:
1. In utero infection—hepatosplenomegaly and hepatopathy, possibly with extramedullary hematopoiesis
2. Nonimmune hydrops fetalis, ichthyotic or collodion skin, or both
3. Neurologic only—early and often difficult to control seizures, hypertonia or hypotonia, with or
without altered head size and with or without eye
findings
4. Coarse facial features with bone changes, dysostosis
multiplex, or osteoporosis
5. Dysmorphic facial features with or without major
malformations
6. Rarely, known family history or positive prenatal
diagnosis
Only for the last three presentations are these conditions
likely to be considered early in the differential diagnosis.
Most babies with these conditions are born to healthy,
nonconsanguineous couples with normal family histories,
and these disorders are usually considered late, if at all, as
in Case Study 1.
LYSOSOMAL STORAGE DISORDERS
Lysosomes are single-membrane–bound intracellular
organelles that contain enzymes called hydrolases. These
lysosomal enzymes are responsible for splitting large molecules into simple, low-molecular-weight compounds,
which can be recycled. The materials digested by lysosomes
and derived from endocytosis and phagocytosis, are separated from other intracellular materials by the process of
autophagy, which is the main mechanism whereby endogenous molecules are delivered to lysosomes. The common
element of all compounds digested by lysosomal enzymes
is that they contain a carbohydrate portion attached to a
protein or lipid. These glycoconjugates include glycoproteins, glycosaminoglycans, and glycolipids.
Glycolipids are large molecules with carbohydrates
attached to a lipid moiety. Sphingolipids, globosides, gangliosides, cerebrosides, and lipid sulfates all are glycolipids. The different classes of glycolipids are distinguished
from one another primarily by different polar groups at
C1. Sphingolipids are complex membrane lipids composed
of one molecule each of the amino alcohol sphingosine,
a long-chain fatty acid, and various polar head groups
attached by a β-glycosidic linkage. Sphingolipids occur in
the blood and nearly all tissues of the body, the highest
concentration being found in white matter of the central
nervous system (CNS). In addition, various sphingolipids are components of the plasma membrane of practically all cells. The core structure of natural sphingolipids
is ceramide, a long-chain fatty acid amide derivative of
sphingosine. Free ceramide, an intermediate in the biosynthesis and catabolism of glycosphingolipids and sphingomyelin, composes 16% to 20% of normal lipid content of
stratum corneum of the skin. Sphingomyelin, a ceramide
phosphocholine, is one of the principal structural lipids of
membranes of nervous tissue.
Cerebrosides are a group of ceramide monohexosides
with a single sugar, either glucose or galactose, and an
additional sulfate group on galactose. The two most common cerebrosides are galactocerebroside and glucocerebroside. The largest concentration of galactocerebroside
is found in the brain. Glucocerebroside is an intermediate in the synthesis and degradation of more complex
glycosphingolipids.
Gangliosides, the most complex class of glycolipids,
contain several sugar units and one or more sialic acid residues. Gangliosides are normal components of cell membranes and are found in high concentrations in ganglion
cells of the CNS, particularly in nerve endings and dendrites. GMI is the major ganglioside in brain of vertebrates.
239
240
PART VI Metabolic and Endocrine Disorders of the Newborn
CASE STUDY 1
C.J. was a 2200-g girl, born to a 24-year-old mother (third pregnancy, second
viable child) after a 32-week gestation, by cesarean section performed for fetal
distress. Pregnancy was complicated by the finding on ultrasonography of fetal
hydrops and ascites and possible hepatosplenomegaly at 24 weeks’ gestation.
Fetal blood sampling showed a hematocrit of 31% and elevations of γ-glutamyltransferase and aspartate transaminase values. Results of viral studies were
negative, and chromosomes were normal. At delivery, the infant was limp and
blue with a heart rate of 60 beats/min. Physical examination and chest radiograph
showed marked abdominal distention, hepatosplenomegaly, multiple petechiae
and bruises, a bell-shaped thorax, generalized hypotonia, talipes equinovarus,
contractures at the knees, a large heart, and hazy lung fields with low volumes.
Disseminated intravascular coagulopathy and evidence of liver disease developed
rapidly, with elevated aspartate transaminase, γ-glutamyl-transferase, and
increasing hyperbilirubinemia. The patient’s condition was maintained with a
ventilator and treatment with antibiotics for possible sepsis.
Results of evaluations for bacterial and viral agents were negative. Metabolic
studies, including ammonia, lactate, very long chain fatty acids (VLCFAs), and
urine amino and organic acids, yielded unremarkable measurements. The white
blood cells were noted to have marked toxic granularity consistent with overwhelming bacterial sepsis or metabolic storage disease.
The patient experienced continued cardiorespiratory deterioration, had
bilateral pneumothoraces and pneumopericardium, and died on the third day
of life. Consent for autopsy was obtained from the family. A standard autopsy
was performed and showed the presence of large, membrane-bound vacuoles
within hepatocytes, endothelial cells, pericytes, and bone marrow stromal
cells, which are typical of a metabolic storage disorder. Similar cells were
also found within the placenta. There was no evidence of an infectious cause.
Unfortunately, because a lysosomal storage disorder was not considered as
a possible cause at the time of death, no frozen tissue or cultured fibroblasts
were available to pursue the diagnosis. As a result of efforts by a research
laboratory and the recurrence of disease in the couple’s subsequent pregnancy,
a diagnosis of β-glucuronidase deficiency, or mucopolysaccharidosis type VII,
was confirmed.
CASE STUDY 2
M.E. was born by normal spontaneous vaginal delivery, at full term according to
dates based on early ultrasonography, with weight of 2.2 kg, length of 45 cm, and
head circumference of 31.5 cm. On the basis of physical examination, gestational
age was assessed as 36 weeks. A heart murmur was noted, and investigation
showed the presence of a small ventricular septal defect with no hemodynamic
significance. Submucous cleft palate was noted. Examination for dysmorphic features showed simple, posteriorly rotated ears, mild epicanthic folds, micrognathia,
and unilateral simian crease. Tone was moderately decreased. Irritability and
severe feeding problems were noted, and gavage feeding was required; growth
was poor despite adequate calories. The results of a karyotype analysis were
normal, and the results of studies for velocardiofacial syndrome were negative.
Vomiting developed, and further evaluation showed no acidosis, hypoglycemia,
or hyperammonemia. Liver-associated values and cholesterol level were normal,
as were results of studies of amino acids, organic acids, and acylcarnitine profile.
Vomiting became more severe and did not respond to elemental formula, and
pyloric stenosis was detected. Feeding problems persisted after successful
s urgical correction. Delivery of more than 140 kcal/kg by gavage was poorly
tolerated, but resulted in weight gain; however, length and head growth remained
poor.
SLO syndrome was suggested despite the normal cholesterol value obtained
on analysis in the hospital laboratory. Studies performed in a specialized laboratory showed the 7- and 8-dehydrocholesterol values to be elevated and the
cholesterol value decreased. Cholesterol supplementation led to some improvement in behavior and feeding. A decrease to 110 kcal/kg/day was tolerated
without worsening of growth, and weight for height gradually returned to normal.
A review of records confirmed that the pregnancy had been accurately dated by
ultrasonography at 10 weeks’ gestation, confirming that M.E. was small for gestational age and microcephalic at birth, with subsequent growth typical for SLO
syndrome. The incorrect assessment of gestational age as 36 weeks on examination was found to result from a failure to appreciate the effect of hypotonia on the
findings for gestational age. The family was counseled about autosomal recessive
inheritance, including the availability of prenatal diagnosis.
CASE STUDY 3
H.K. was born at term to healthy parents by cesarean section performed for
breech presentation after an otherwise uncomplicated pregnancy. Hypotonia and
dysmorphic features were noted in the delivery room, including inner epicanthic
folds, flat occiput, large fontanels, shallow orbital ridges, low nasal bridge,
micrognathia, redundant skin folds at the neck, and unilateral simian crease.
Brushfield spots were present. Investigation of a heart murmur revealed patent
ductus arteriosus and a small atrial septal defect. There was mild hepatomegaly
but normal liver function, no acidosis, and no hypoglycemia. Suck was poor, and
gavage feeding was required.
Gangliosides function as receptors for toxic agents, hormones, and certain viruses, are involved in cell differentiation, and they can also have a role in cell-cell interaction by
providing specific recognition determinants on the surface
of cells.
Ceramide oligosaccharides (i.e., globosides) are a family of cerebrosides that contain two or more sugar residues, usually galactose, glucose, or N-acetylgalactosamine.
Glycosaminoglycans and oligosaccharides are essential
constituents of connective tissue, parenchymal organs,
cartilage, and the nervous system.
Karyotype was normal and there was no evidence of trisomy 21 in blood in 50
interphase cells examined. The option of skin biopsy to search further for evidence of
mosaicism for trisomy 21 was considered. Thyroid function values were normal. Urine
amino and organic acid values were normal, as was the acylcarnitine profile. Plasma
VLCFA analysis showed elevation consistent with a diagnosis of Zellweger syndrome,
along with a typical increase in pipecolic acid value and impaired capacity for
fibroblast synthesis of plasmalogens. The baby died at 3 months of age, and autopsy
showed polymicrogyria and small hepatic and renal cysts. The family was counseled
about autosomal recessive inheritance, including the availability of prenatal diagnosis.
Glycosaminoglycans, also called mucopolysaccharides, are
complex heterosaccharides consisting of long sugar chains
rich in sulfate groups. The polymeric chains are bound to
specific proteins (core proteins). Glycoproteins contain
oligosaccharide chains (long sugar molecules) attached
covalently to a peptide core. Glycosylation occurs in the
endoplasmic reticulum and Golgi apparatus. Most glycoproteins are secreted from cells and include transport
proteins, glycoprotein hormones, complement factors,
enzymes, and enzyme inhibitors. There is extensive diversity in the composition and structure of oligosaccharides.
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
241
CASE STUDY 4
M.J. had hypotonia at birth after an uncomplicated pregnancy. Minor dysmorphic
features were noted, including high nasal bridge, large ears, and inverted nipples.
Feeding difficulties were significant, and growth was poor. Findings on head ultrasonography were unremarkable, as were those of head magnetic resonance imaging, although the radiologist questioned whether the cerebellum might be slightly
small. Results of a karyotype analysis were normal. Hypothyroidism, discovered
on newborn screening, was promptly treated and closely monitored. There was no
acidosis or hypoglycemia, and liver enzyme values were normal; results of amino
and organic acid analyses and acylcarnitine profile were all normal.
The baby was discharged on a diet providing 130 kcal/kg/day. On follow-up,
growth remained poor, and development was severely delayed. At 6 months of
age, she was admitted to the hospital for an episode of acutely altered mental
status and low blood pressure. Mild acidosis, borderline elevations of lactate and
ammonia, and significant elevation of liver enzymes all resolved over the course
The degradation of glycolipids, glycosaminoglycans, and
glycoproteins takes place especially within lysosomes of
phagocytic cells, related to histiocytes and macrophages, in
any tissue or organ. A series of hydrolytic enzymes cleaves
specific bonds, resulting in sequential, stepwise removal of
constituents such as sugars and sulfate, and degrading complex glycoconjugates to the level of their basic building blocks.
Lysosomal storage diseases most commonly result when an
inherited defect causes significantly decreased activity in one
of these hydrolases. Other causes are failure of transport of an
enzyme, substrate, or product. Whatever the specific cause,
incompletely metabolized molecules accumulate, especially
within the tissue responsible for catabolism of the glycoconjugate. Additional excess storage material may be excreted in
urine. The mechanisms of cellular dysfunction and damage
in the majority of LSDs remain unknown. Various hypotheses have been offered, such as a pivotal disturbance in the
normal process of autophagy (Ballabio and Gieselmann,
2009; Kiselyov et al, 2007). In this pathophysiologic construct, endoplasmic reticulum membrane engulfment of cellular components, such as mitochondrial derivatives targeted
for destruction, is perturbed. As a consequence, deleterious
pathways become activated, leading to unwanted ubiquitination of targeted molecules and apoptosis.
Lysosomal storage diseases are classified according to
the stored compound. Clinical phenotype depends partially on the type and amount of storage substance. There
are more than 50 different LSDs and a significant fraction,
approximately 20 LSDs, may have manifestations in the
newborn infant (Staretz-Chocham et al, 2009). The disorders selected for discussion in this chapter are all known to
manifest in the neonatal period.
CLINICAL PRESENTATIONS
Table 23-1 summarizes the clinical characteristics of the
neonatal presentations of lysosomal storage disorders.
Niemann-Pick A Disease (Acute,
Sphingomyelinase Deficient)
Etiology
Niemann-Pick A disease is caused by a deficiency of sphingomyelinase. Sphingomyelinase catalyzes the breakdown
of sphingomyelin to ceramide and phosphocholine, and
of the hospital stay. Cardiac ultrasonography showed mild ventricular dysfunction,
which also resolved. Amino and organic acid values were normal, as was the
acylcarnitine profile. Urine oligosaccharide levels showed an unusual pattern, and
urine mucopolysaccharide values were normal.
At 2 years of age, developmental delay remained marked, and hypotonia persisted with reflexes absent. The creatinine phosphokinase level was normal, but
liver function values were again abnormal. Because mitochondrial disease was
suspected, the patient was scheduled for liver biopsy, but clotting values were
abnormal. A congenital disorder of glycosylation was suspected, and a transferrin
assay confirmed the diagnosis. A review of neonatal records revealed a comment
from a neurology consultant about the unusual distribution of fat on the buttocks
and thighs of M.J. as a neonate. The family was counseled about autosomal
recessive inheritance, including availability of prenatal diagnosis.
its deficiency results in sphingomyelin storage within
lysosomes. Cholesterol is also stored, suggesting that its
metabolism is tied to that of sphingomyelin. Sphingomyelin normally composes 5% to 20% of phospholipid in
liver, spleen, and brain, but in these disorders it can compose up to 70% of phospholipids. Patients with NiemannPick A disease usually have enzyme activity less than 5%
of normal.
Clinical Features
Clinical features of this disorder may appear in utero or up
to 1 year of age. Affected infants usually have massive hepatosplenomegaly (hepatomegaly greater than splenomegaly), constipation, feeding difficulties, and vomiting with
consequent failure to thrive. Patients eventually appear
strikingly emaciated with a protuberant abdomen and thin
extremities. Neurologic disease is evident by 6 months of
age, with hypotonia, decrease or absence of deep tendon
reflexes, and weakness. Loss of motor skills, spasticity,
rigidity, and loss of vision and hearing occur later. Seizures
are rare. A retinal cherry-red spot is present in about half of
cases, and the electroretinographic findings are abnormal.
Respiratory infections are common. The skin may have
an ochre or brownish yellow color, and xanthomas have
been observed. Radiographic findings consist of widening
of medullary cavities, cortical thinning of long bones, and
osteoporosis. In the brain and spinal cord, neuronal storage is widespread, leading to cytoplasmic swelling together
with atrophy of cerebellum. Bone marrow and tissue biopsy
samples may show foam cells or sea-blue histiocytes, which
represent lipid-laden cells of the monocyte-macrophage
system. Similarly, vacuolated lymphocytes or monocytes
may be present in peripheral blood. Tissue cholesterol levels may be threefold to tenfold of normal, and patients may
have a microcytic anemia and thrombocytopenia. Death
occurs by 2 to 3 years of age.
Niemann-Pick C Disease
Etiology
Niemann-Pick C disease is caused by an error in the intracellular transport of exogenous low-density lipoprotein
(LDL)–derived cholesterol, which leads to impaired esterification of cholesterol and trapping of unesterified cholesterol in lysosomes. The incidence may be higher than
242
TABLE 23-1 Lysosomal Storage Disorders in the Newborn Period: Genetic and Clinical Characteristics of Neonatal Presentation
Neurologic
Facies Findings
Distinctive
Features
Eye
Findings
Cardiovascular
Dysostosis Hepatomegaly/
Findings
Multiplex Splenomegaly Defect
Gene Location
Molecular
Findings
Ethnic
Predilection
Onset
NiemannPick A
disease
Early
Frontal
infancy
bossing
Difficulty feeding,
apathy, deafness,
blindness,
hypotonia
Brownish-yellow Cherry-red
skin, xanthomas
spot (50%)
–
–
++/+
Sphingomyelinase
deficiency
NiemannPick C
disease
Birth to
3 mo
Normal
Developmental
delay, vertical
gaze paralysis,
hypotonia, later
spasticity
–
–
–
–
+/++
Abnormal cholesterol NPC1 gene at 18q11 Increased in
esterification
accounts
French
for >95% of cases;
Canadians of
HE1 gene mutaNova Scotia
tions may account
and
for remaining
Spanish
cases
Americans in
the southwest
United States
Gaucher
disease
type 2
In utero
to 6
mo
Normal
Congenital
Poor suck and
ichthyosis,
swallow, weak
collodion skin
cry, squint, trismus,
strabismus,
opsoclonus,
hypertonic,
later flaccidity
–
–
–
+/++
Glucocerebrosidase
deficiency
1q21; large number Panethnic
of mutations
known; five mutations account for
approximately
97% of mutant
alleles in the Ashkenazi population,
but approximately
75% in the nonJewish population
Krabbe
disease
3-6 mo
Normal
Irritability, tonic
spasms with light
or noise stimulation, seizures,
hypertonia, later
flaccidity
Optic atrophy
–
–
–/–
Galactocerbrosidase
deficiency
14q 24.3-q32.1; >60 Increased in
mutations with
Scandinavian
countries
some common
and in a large
mutations in speDruze kindred
cific populations
in Israel
Coarse
Poor suck, weak
Gingival
cry, lethargy,
hypertrophy,
exaggerated startle,
edema, rashes
blindness, hypotonia, later spasticity
Cherry-red
spot (50%)
–
+
+/+
β-Galactosidase
deficiency
3 pter-3p21; hetero- Panethnic
geneous mutations;
common mutations in specific
populations
–
Hepatomegaly in
50%, splenomegaly less
common
Lysosomal acid
ceramidase
8p21.3-22; 9 disease- Panethnic
causing mutations
identified
GM1 ganglio- Birth
sidosis
Farber disease
type I
2 wk to 4 Normal
mo
Increased CSF
protein
Grayish opaci- Occasional
Progressive psychoJoint swelling
fication
motor impairment,
with nodules,
surroundseizures, decreased
hoarseness,
ing retina
reflexes, hypotonia
lung disease,
in some
contractures,
patients,
fever, granulosubtle
mas, dysphagia,
vomiting,
cherry-red
increased CSF
spot
protein
ASM gene at
1:40,000 in
11p15.1-p15.4
Ashkenazi Jews
3 of 18 mutawith carrier
tions account for
frequency of
approximately
1:60
92% of mutant
alleles in the Ashkenazi population
PART VI Metabolic and Endocrine Disorders of the Newborn
Disorder
Normal
Farber
disease
type IV
(neonatal)
Birth
Normal
Congenital
sialidosis
In utero Coarse, Mental retardation,
to birth
edema
hypotonia
Galactosialidosis
In utero Coarse
to birth
Mental retardation,
Ascites, edema,
Cherry-red
occasional deafness,
inguinal hernias,
spot,
hypotonia
renal disease,
corneal
telangiectasias
clouding
Wolman
disease
First
weeks
of life
Mental deterioration
Vomiting, diarrhea, steatorrhea, abdominal
distention, failure to thrive,
anemia, adrenal
calcifications
Infantile
sialic acid
storage
disease
In utero Coarse,
to birth
dysmorphic
Mental retardation,
hypotonia
Ascites, anemia,
diarrhea, failure
to thrive
I-cell
disease
In utero Coarse
to birth
Mental retardation,
deafness
Gingival hyperCorneal
plasia, restricted
clouding
joint mobility,
hernias
Mental retardation,
hypotonia
—
Severe corneal
clouding,
retinal
degeneration, blindness
Hernias
Variable
corneal
clouding
Normal
Nodules not consistent findings
Mucolipidosis Birth to
type IV
3 mo
Normal
Mucopolysaccharidosis type
VII
Variable Mild to severe
coarse- mental retardation
ness
In utero
to
childhood
Joint swelling with Normal
nodules, hoarsemacula,
ness
corneal
opacities
–
Corneal opacities
(1/3)
–
–
Neonatal ascites, Corneal
inguinal hernias,
clouding
renal disease
–
HSM less common
than in type I
8p21.3-p22
Panethnic
++/++
Unknown
Panethnic
NEU 1 gene (sialidase) at 6p21
Panethnic
–
+
+/+
Neuraminidase
deficiency
Cardiomegaly
progressing
to failure
+
+/+
Absence of a pro20q13.1
tective protein
that safeguards
neuraminidase and
beta-galactosidase
from premature
degradation
–
–
–
+/+
Lysosomal acid lipase 10q23.2-q23.3; vari- Increased in Iradeficiency
ety of mutations
nian Jews and
identified
in non-Jewish
and Arab
populations of
Galilee
–
Congestive
heart failure
+
+/+
Defective transport
SLC17A5
of sialic acid out of
gene at 6q
the lysosome
Valvular
disease,
congestive heart
failure, cor
pulmonale
++
+++/+++
–
–
–/–
Variable
++
Variable
Panethnic
Panethnic
Lysosomal enzymes Enzyme encoded by Panethnic
two genes; α and β
lack mannosesubunits encoded
6-PO4 recognition
marker and fail
by gene at 12p; γ
to enter the lysosubunit encoded
some (phospho-
by gene at 16p
transferase
deficiency,
3-subunit complex
[α2 β2 γ2])
Unknown; some
patients with
partial deficiency
of ganglioside
sialidase
MCOLN1 gene
Increased in
at 19p13.2Ashkenazi
13.3 encoding
Jews
mucolipin; two
founder mutations
accounting for
95% of mutant
alleles in Ashkenazi population
β-Glucuronidase
deficiency
GUSB gene at
7q21.2-q22;
heterogeneous
mutations
–, Not seen; +, typically present, usually not severe; ++, usually present, and moderately severe; +++, always present, usually severe; CSF, cerebrospinal fluid; HSM, hepatosplenomegaly.
Panethnic
243
Birth to
9 mo
(≤20
mo)
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
Farber disease types
II and III
244
PART VI Metabolic and Endocrine Disorders of the Newborn
1 in 150,000 births (Wraith et al, 2009). Cell lines from
patients can be divided into two complementation groups,
NPC1 and NPC2, corresponding to different genes (Millat
et al, 2001). In each group, the primary defect is abnormal cholesterol esterification, but the enzyme responsible
for cholesterol esterification—acetyl coenzyme A (CoA)
acetyltransferase (ACAT)—is not deficient. The storage
of sphingomyelin is secondary. It has been suggested that
the defect is in transport of cholesterol out of the lysosome, making cholesterol unavailable to ACAT (Natowicz
et al, 1995). Sphingomyelinase activity appears normal or
elevated in most tissues, but is partially deficient (60% to
70%) in fibroblasts from most patients with this disorder.
Storage of sphingomyelin in tissues is much less than in
Niemann-Pick A or B disease and is accompanied by additional storage of unesterified cholesterol, phospholipids,
and glycolipids in the liver and spleen. Only glycolipids
are increased in the brain.
Clinical Features
The age of onset, clinical features, and natural history of
Niemann-Pick C disease are highly variable. Onset can
occur from birth to 18 years of age. Fifty percent of children with onset in the neonatal period have conjugated
hyperbilirubinemia, which usually resolves spontaneously
but is followed by neurologic symptoms later in childhood.
In the severe infantile form, hepatosplenomegaly is common, accompanied by hypotonia and delayed motor development. Further mental regression is usually evident by
the age of 1 to 1.5 years, in association with behavior problems, vertical supranuclear ophthalmoplegia, progressive
ataxia, dystonia, spasticity, dementia, drooling, dysphagia,
and dysarthria. Seizures are rare. Foam cells and sea-blue
histiocytes may be found in many tissues. Neuronal storage with cytoplasmic ballooning, inclusions, meganeurites,
and axonal spheroids are also seen. Death may occur in
infancy or as late as the third decade of life. Niemann-Pick
C disease can also manifest as fatal neonatal liver disease,
often misdiagnosed as fetal hepatitis. Patients with mutations in the NPC2 gene (HE1) may have remarkable features consisting of pronounced pulmonary involvement
leading to early death caused by respiratory failure (Millat
et al, 2001).
Gaucher Disease Type 2
(Acute Neuropathic)
Etiology
Three types of Gaucher disease have been defined. Type 1,
the nonneuropathic form, is the most common and is distinguished from types 2 and 3 by the lack of CNS involvement. Type 1 disease most commonly manifests in early
childhood, but may do so in adulthood. Type 2 disease, the
acute neuropathic form, is characterized by infantile onset
of severe CNS involvement. Type 3 disease, the subacute
neuropathic form, is also late in onset with slow neurologic progression. Almost all types of Gaucher disease are
caused by a deficiency of lysosomal glucocerebrosidase and
result in storage of glucocerebroside in visceral organs;
the brain is affected in types 2 and 3. Although there is
significant variability in clinical presentation among
individuals with the same mutations, there is a clear correlation between certain mutations and clinical symptoms
involving the CNS (Beutler and Grabowski, 2001). The
enzyme splits glucose from cerebroside, yielding ceramide
and glucose. A few patients with Gaucher disease type 2
have a deficiency of saposin C, a cohydrolase required by
glucocerebrosidase.
Clinical Features
Typically, the age of onset of Gaucher disease type 2 is
approximately 3 months, consisting of hepatosplenomegaly
(splenomegaly predominates) with subsequent neurologic
deterioration. Hydrops fetalis, congenital ichthyosis, and
collodion skin, however, are well-described presentations
(Fujimoto et al, 1995; Ince et al, 1995; Lipson et al, 1991;
Liu et al, 1988; Sherer et al, 1993; Sidransky et al, 1992). In
a review of 18 cases of Gaucher disease manifesting in the
newborn period, Sidransky et al (1992) found that eight
of the patients had associated dermatologic findings and
six patients had hydrops. The etiology of the association
of such findings and Gaucher disease is unclear, although
the enzyme deficiency appears to be directly responsible
(Sidransky et al, 1992). Ceramides have been shown to be
major components of intracellular bilayers in epidermal
stratum corneum, and they have an important role in skin
homeostasis (Fujimoto et al, 1995). Therefore Gaucher
disease should be considered in the differential diagnosis
for infants with hydrops fetalis and congenital ichthyosis.
For the subset of patients in the prenatal period or at birth,
death frequently occurs within hours to days, or at least
within 2 to 3 months.
Krabbe Disease (Globoid Cell
Leukodystrophy)
Etiology
The synonym for Krabbe disease, globoid cell leukodystrophy, is derived from the finding of large numbers of multinuclear macrophages in cerebral white matter that contain
undigested galactocerebroside. Disease is caused by a deficiency of lysosomal galactocerebroside β-galactosidase,
which normally degrades galactocerebroside to ceramide
and galactose. Deficiency of the enzyme results in storage of galactocerebroside. Galactocerebroside is present
almost exclusively in myelin sheaths. Accumulation of
the toxic metabolite psychosine, also a substrate for the
enzyme, has been postulated to lead to early destruction
of oligodendroglia. Impaired catabolism of galactosylceramide is also important in pathogenesis of the disease.
Clinical Features
Age of onset ranges from the first weeks of life to adulthood. The typical age of onset of infantile Krabbe disease is
between 3 and 6 months, but there are cases of early onset in
which neurologic symptoms are evident within weeks after
birth. Symptoms and signs are confined to the nervous system; no visceral involvement is present. The clinical course
has been divided into three stages. In stage I, patients who
appeared relatively normal after birth exhibit hyperirritability, vomiting, episodic fevers, hyperesthesia, tonic spasms
with light or noise stimulation, stiffness, and seizures.
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
Peripheral neuropathy is present, but reflexes are increased.
Stage II is marked by CNS deterioration and hypertonia
that progresses to hypotonia and flaccidity. Deep tendon
reflexes are eventually lost. Patients with stage III disease are
decerebrate, deaf, and blind with hyperpyrexia, hypersalivation, and frequent seizures. Routine laboratory findings
are unremarkable except for an elevation of cerebrospinal
fluid protein. Cerebral atrophy and demyelination become
evident in the CNS, and segmental demyelination, axonal degeneration, fibrosis, and macrophage infiltration are
common in the peripheral nervous system. The segmental
demyelination of peripheral nerves is demonstrated by the
finding of decreased motor nerve conduction. The white
matter is severely depleted of all lipids, especially glycolipids, and nerve and brain biopsies show globoid cells. Death
from hyperpyrexia, respiratory complications, or aspiration
occurs at a median age of 13 months.
GM1 Gangliosidosis
Etiology
Infantile GM1 gangliosidosis is caused by a deficiency in
lysosomal β-galactosidase. The enzyme cleaves the terminal galactose in a β linkage from oligosaccharides, keratan
sulfate, and GM1 ganglioside. Deficiency of the enzyme
results in storage of GM1 ganglioside and oligosaccharides.
Clinical severity correlates with the extent of substrate
storage and residual enzyme activity. The same enzyme is
deficient in Morquio disease type B.
Clinical Features
Age of onset ranges from prenatal to adult. Infantile or
type 1 GM1 gangliosidosis may be evident at birth as coarse
and thick skin, hirsutism on the forehead and neck, and
coarse facial features consisting of a puffy face, frontal
bossing, depressed nasal bridge, maxillary hyperplasia,
large and low-set ears, wide upper lip, moderate macroglossia, and gingival hypertrophy. These dysmorphic
features, however, are not always obvious in the neonate.
A retinal cherry-red spot is seen in 50% of patients, and
corneal clouding is often observed. Shortly after birth,
or by 3 to 6 months of age, failure to thrive and hepatosplenomegaly become evident, as does neurologic involvement with poor development, hyperreflexia, hypotonia,
and seizures. Cranial imaging shows diffuse atrophy of the
brain, enlargement of the ventricular system, and evidence
of myelin loss in white matter.
The neurologic deterioration is progressive, resulting
in generalized rigidity and spasticity and sensorimotor
and psychointellectual dysfunction. By 6 months of age,
skeletal features are present, including kyphoscoliosis and
stiff joints with generalized contractures, and striking bone
changes are seen—vertebral beaking in the thoracolumbar region, broadening of shafts of the long bones with
distal and proximal tapering, and widening of the metacarpal shafts with proximal pinching of four lateral metacarpals. Tissue biopsy samples demonstrate neurons filled
with membranous cytoplasmic bodies and various types of
inclusions as well as foam cells in the bone marrow. Death
generally occurs before 2 years of age. A severe neonatalonset type of GM1 gangliosidosis with cardiomyopathy has
also been described (Kohlschütter et al, 1982).
245
Farber Lipogranulomatosis
Etiology
Farber lipogranulomatosis results from a deficiency of
lysosomal acid ceramidase. Ceramidase catalyzes the degradation of ceramide to its long-chain base, sphingosine,
and a fatty acid. Clinical disease is a consequence of storage of ceramide in various organs and body fluids.
Clinical Features
Four types of Farber lipogranulomatosis can manifest in
the neonatal period. Type I, classic disease, is a unique
disorder with onset from approximately 2 weeks to 4
months of age. Patients exhibit hoarseness progressing to
aphonia, feeding and respiratory difficulties, poor weight
gain, and intermittent fever caused by granuloma formation, and swelling of the epiglottis and larynx. Palpable
nodules appear over joints and pressure points, and joints
become painful and swollen. Later, joint contractures and
pulmonary disease appear. Liver and cardiac involvement
can occur, and patients can have a subtle retinal cherryred spot. Severe and progressive psychomotor impairment
can occur, as can seizures, decreased deep tendon reflexes,
hypotonia, and muscle atrophy. Affected patients die in
early infancy, usually from pulmonary disease.
Type 2, or intermediate, Farber lipogranulomatosis manifests from birth to 9 months of age as joint and
laryngeal involvement and nodules. Death occurs in early
childhood. Type 3 disease (mild) manifests slightly later,
from approximately 2 months to 20 months of age, with
survival into the third decade. Clinically types 2 and 3 are
both dominated by subcutaneous nodules, joint deformity, and laryngeal involvement. Liver and pulmonary
involvement may be absent. Two thirds of patients have
a normal intelligence quotient. Type 4, or neonatal visceral, Farber lipogranulomatosis manifests at birth as
hepatosplenomegaly caused by massive histiocyte infiltration of the liver and spleen, with infiltration also in the
lungs, thymus, and lymphocytes. Subcutaneous nodules
and laryngeal involvement may be subtle. Death occurs by
6 months of age.
In all types of Farber lipogranulomatosis, tissue biopsy
samples show granulomatous infiltration, foam cells, and
lysosomes with comma-shaped, curvilinear tubular structures called Farber bodies. Cerebrospinal fluid protein may
be elevated in patients with type 1 disease.
Sialidosis
Etiology
Sialidosis is caused by a deficiency of neuraminidase, which
is responsible for the cleavage of terminal sialyl linkages
of several oligosaccharides and glycopeptides. The defect
results in multisystem lysosomal accumulation of sugars
rich in sialic acid.
Clinical Features
Type I sialidosis is characterized by retinal cherry-red
spots and generalized myoclonus with onset generally
in the second decade of life. Type II is distinguished from
type I by the early onset of a progressive, severe phenotype with somatic features. Type II is often subdivided
246
PART VI Metabolic and Endocrine Disorders of the Newborn
into juvenile, infantile, and congenital forms. Congenital
sialidosis begins in utero and manifests at birth as coarse
features, facial edema, hepatosplenomegaly, ascites, hernias, and hypotonia, and occasionally frank hydrops fetalis. Radiographs demonstrate dysostosis multiplex and
epiphyseal stippling. Delayed mental development is
quickly apparent. The patient may have recurrent infections. Severely dilated coronary arteries, excessive retinal
vascular tortuosity, and an erythematous macular rash may
also be features of this disease (Buchholz et al, 2001). Most
patients are stillborn or die before 1 year of age. Age of
onset for the infantile form of sialidosis ranges from birth
to 12 months. Clinical features are coarse facial features,
organomegaly, dysostosis multiplex, retinal cherry-red
spot, and mental retardation. Death occurs by the second
or third decade. In both types of sialidosis, vacuolated cells
can be seen in almost all tissues, and bone marrow foam
cells are present.
Wolman Disease
Etiology
Galactosialidosis
Wolman disease is caused by lysosomal acid lipase deficiency, which is an enzyme involved in cellular cholesterol
homeostasis and responsible for hydrolysis of cholesterol
esters and triglycerides. The result of enzyme deficiency is
defective release of free cholesterol from lysosomes, which
leads to upregulation of LDL receptors and 3-hydroxy-3methylglutaryl-CoA reductase activity. De novo synthesis
of cholesterol and activation of receptor-mediated endocytosis of LDL then occur, leading to further deposition of
lipid in lysosomes. The result is the accumulation of cholesterol esters and triglycerides in most tissues of the body,
including the liver, spleen, lymph nodes, heart, blood vessels, and brain. An extreme level of lipid storage occurs in
cells of the small intestine, particularly in the mucosa. In
addition, neurons of the myenteric plexus demonstrate a
high level of storage, with evidence of neuronal cell death,
which may account for prominence of gastrointestinal
symptoms (Wolman, 1995).
Etiology
Clinical Features
Galactosialidosis results from a deficiency of two lysosomal
enzymes, neuraminidase and β-galactosidase. The primary
defect in galactosialidosis has been found to be a defect in
protective protein–cathepsin A, an intralysosomal protein
that protects the two enzymes from premature proteolytic
processing. The protective protein has catalytic and protective functions, and the two functions appear to be distinct. Deficiency of enzymes results in the accumulation of
sialyloligosaccharides in tissue lysosomes and in excreted
body fluids.
Clinical presentation of Wolman disease is within weeks
of birth, with evidence of malnutrition and malabsorption,
including symptoms of vomiting, diarrhea, steatorrhea,
failure to thrive, abdominal distention, and hepatosplenomegaly. Adrenal calcifications may be seen on radiographs,
and adrenal insufficiency appears. The presence of adrenal
calcifications in association with hepatosplenomegaly and
gastrointestinal symptoms is strongly suggestive of Wolman disease. Later, mental deterioration becomes apparent. Laboratory findings include anemia secondary to
foam cell infiltration of the bone marrow and evidence of
adrenal insufficiency. The serum cholesterol level is normal. Death usually occurs before 1 year of age.
Clinical Features
Galactosialidosis has been divided into three phenotypic
subtypes based on age at onset and severity of clinical
manifestations. Most cases occur in adolescence and adulthood, but early infantile and late infantile presentations
occur. Patients develop early infantile galactosialidosis
between birth and 3 months of age with ascites, edema,
coarse facial features, inguinal hernias, proteinuria, hypotonia, and telangiectasias, and, occasionally, frank hydrops
fetalis. Patients subsequently demonstrate organomegaly,
including cardiomegaly progressing to cardiac failure, psychomotor delay, and skeletal changes, particularly in the
spine. Ocular abnormalities can occur, including corneal
clouding and retinal cherry-red spots. Death occurs at an
average age of 8 months, usually from cardiac and renal
failure. Galactosialidosis can be a cause of recurrent fetal
loss or recurrent hydrops fetalis.
Late infantile galactosialidosis manifests in the first
months of life as coarse facial features, hepatosplenomegaly, and skeletal changes consistent with dysostosis multiplex. Cherry-red spots and corneal clouding may also be
present. Neurologic involvement may be absent or mild.
Valvular heart disease is a common feature, as is growth
retardation, partially because of spinal involvement and
often in association with muscular atrophy. Early death is
not a feature of the late infantile form. Vacuolated cells in
blood smears and foam cells in bone marrow are present in
all forms of galactosialidosis.
Infantile Sialic Acid Storage Disease
Etiology
Infantile sialic acid storage disease is caused by a defective lysosomal sialic acid transporter that is responsible for
efflux of sialic acid and other acidic monosaccharides from
the lysosomal compartment. The defective transporter
results in greater storage of free sialic acid and glucuronic
acid within lysosomes and increased sialic acid excretion.
Clinical Features
Infantile sialic acid storage disease often manifests at birth
as mildly coarse features, hepatosplenomegaly, ascites,
hypopigmentation, and generalized hypotonia. Mild dysostosis multiplex may be seen on radiographs. Failure
to thrive and severe mental and motor retardation soon
appear. Cardiomegaly may be present. Corneas are clear,
but albinoid fundi have been reported (Lemyre et al,
1999). Vacuolated cells are seen on a tissue biopsy sample,
and electron microscopy demonstrates swollen lysosomes
filled with finely granular material. CNS changes include
myelin loss, axonal spheroids, gliosis, and neuronal storage.
Death occurs in early childhood. Infantile sialic acid storage disease can also manifest as fetal ascites, nonimmune
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
fetal hydrops, or infantile nephrotic syndrome (Lemyre
et al, 1999).
I-Cell Disease (Mucolipidosis Type II)
Etiology
In normal cells, targeting of enzymes to lysosomes is mediated by receptors that bind a mannose-6-phosphate recognition marker on the enzyme. The recognition marker
is synthesized in a two-step reaction in the Golgi complex.
It is the enzyme that catalyzes the first step of this process, uridine diphosphate–N-acetylglucosamine: lysosomal
enzyme N-acetylglucosaminyl-1-phosphotransferase, that
is defective in I-cell disease. As a result, the enzymes lack
the mannose-6-phosphate recognition signal, and the
newly synthesized lysosomal enzymes are secreted into
the extracellular matrix instead of being targeted to the
lysosome. Consequently, multiple lysosomal enzymes are
found in plasma in 10- to 20-fold their normal concentrations. Affected cells, especially fibroblasts, show dense
inclusions of storage material that probably consists of
oligosaccharides, glycosaminoglycans, and lipids; these
are the inclusion bodies from which the disease name is
derived. This disorder is found more frequently in Ashkenazi Jews, because of a putative founder effect.
Clinical Features
I-cell disease can manifest at birth as coarse features, corneal clouding, organomegaly, hypotonia, and gingival
hyperplasia. Birthweight and length are often below normal. Kyphoscoliosis, lumbar gibbus, and restricted joint
movement are often present, and there may be hip dislocation, fractures, hernias, or bilateral talipes equinovarus.
Dysostosis multiplex may be seen on radiographs. Severe
psychomotor retardation, evident by 6 months of age,
and progressive failure to thrive occur. The facial features
become progressively more coarse, with a high forehead,
puffy eyelids, epicanthal folds, flat nasal bridge, anteverted
nares, and macroglossia. Linear growth slows during the
first year of life and halts completely thereafter. The skeletal involvement is also progressive, with development of
increasing joint immobility and claw-hand deformities.
Respiratory infections, otitis media, and cardiac involvement are common complications. Death usually occurs
in the first decade of life because of cardiorespiratory
complications.
Mucolipidosis Type IV
Etiology
Although mucolipidosis type IV is associated with a partial deficiency of the lysosomal enzyme ganglioside sialidase, a deficiency of mucolipin 1 (TRPML1), a member
of the transient receptor potential TRMPL subfamily
of channel proteins, is the cause of the disorder (Bargal
et al, 2000; Sun et al, 2000). Mutations in the MCOLN1
gene result in lysosomal storage of lipids such as gangliosides, plus water-soluble materials such as glycosaminoglycans and glycoproteins in cells from almost all
tissues.
247
Clinical Features
The age of onset for mucolipidosis type IV ranges from
infancy to 5 years. Presenting features are corneal clouding (may be congenital), retinal degeneration, blindness,
hypotonia, and mental retardation. Survival of affected
patients into the fourth decade of life has been reported
(Chitayat et al, 1991). Cytoplasmic inclusions are noted
in many cells, including those in conjunctiva, liver, and
spleen, as well as fibroblasts.
Mucopolysaccharidosis Type VII
(Sly Disease)
Etiology
Sly disease is a member of a group of lysosomal storage
disorders that are caused by a deficiency of enzymes catalyzing the stepwise degradation of glycosaminoglycans.
Skeletal and neurologic involvement is variable. There is
a wide spectrum of clinical severity among the mucopolysaccharidoses and even within a single enzyme deficiency.
Most of these disorders manifest in childhood, but type VII
is included in this chapter because of its well-recognized
neonatal and infantile presentations. Sly disease is caused
by β-glucuronidase deficiency and results in lysosomal
accumulation of glycosaminoglycans, including dermatan
sulfate, heparan sulfate, and chondroitin sulfate, causing
cell, tissue, and organ dysfunction.
Clinical Features
Sly disease can manifest as a wide spectrum of severity.
Patients with the early-onset or neonatal form may have
coarse features, hepatosplenomegaly, moderate dysostosis
multiplex, hernias, and nonprogressive mental retardation.
Corneal clouding is variably present. Frequent episodes of
pneumonia during the first year of life are common. Short
stature becomes evident. Granulocytes have coarse metachromic granules. A severe neonatal form associated with
hydrops fetalis, and early death has been recognized frequently. Milder forms of the disease with later onset are
also known.
DIAGNOSIS, MANAGEMENT,
AND PROGNOSIS
Growing recognition of lysosomal storage disorders in
the neonate has led to expansion of the spectrum of possible clinical presentation in the newborn period. Diagnostic tools and options for treatment also continue to
advance. For example, efforts are currently underway to
develop newborn screening for mucopolysaccharidoses
(Whitley et al, 2002), with the goal to offer treatment with
enzyme infusion or bone marrow transplantation (BMT)
to affected babies (Vogler et al, 1999). The state of New
York has implemented newborn screening for Krabbe disease using dried blood spots. The test uses a tandem massspectrometry–based enzyme analysis (Li et al, 2004a).
This test has resulted in a fairly large number of positive
newborn screens for Krabbe disease, most of which appear
to be false positives, including enzyme perturbations that
are not linked with clinical disease (Duffner et al, 2009).
248
PART VI Metabolic and Endocrine Disorders of the Newborn
As a consequence, an expert advising panel, the Krabbe
Consortium of New York State, has been generated to
establish standardized clinical evaluation guidelines. The
goal is to help physicians determine which infant with a
positive newborn screen may express disease and require
treatment, such as hematopoietic stem cell transplantation in early infancy. The neonatologist is urged to work
closely with appropriate experts to explore diagnostic
and treatment protocols on an individual basis. Larger
panels of multiplex testing for various other LSDs are
in the testing stages (Li et al, 2004b) and some states in
the United States are poised to begin implementing LSD
newborn screening. Currently a federal advisory committee actively reviews and makes recommendations to
the U.S. Secretary of Health and Human Services about
the introduction of new newborn screening tests in the
United States, with the aim of vetting proposed tests for
need, cost effectiveness, and availability of effective and
timely therapy.
Recognizing lysosomal storage disorders in the newborn
period can be difficult, because they often mimic more
common causes of illness in newborns, such as respiratory
distress, nonimmune hydrops fetalis, liver disease, and sepsis. The initial step in the diagnosis of these disorders is
to consider them in the differential diagnosis of a sick or
unusual-appearing newborn. At times the phenotype may
suggest a specific diagnosis, such as respiratory distress and
painful, swollen joints in Farber lipogranulomatosis or gastrointestinal symptoms, hepatosplenomegaly, and adrenal
calcifications in Wolman disease. Subtle dysmorphic features, coarsening of features, and radiographic evidence
of dysostosis multiplex are also strong indications that
lysosomal storage disorders should be considered. Routine laboratory findings are often normal or nonspecific.
Affected infants do not have episodes of acute metabolic
decompensation. Anemia and thrombocytopenia may be
seen because of bone marrow involvement. Vacuolated
cells may be found in peripheral blood, but the absence
of this finding does not exclude lysosomal storage disease.
Elevated cerebrospinal fluid protein is seen in Krabbe disease and Farber lipogranulomatosis type I.
Nonimmune hydrops fetalis deserves special mention.
The physician must consider LSDs as the cause of nonimmune hydrops fetalis or unexplained ascites in the affected
newborn infant. The following LSDs are potential causes:
sialidosis type II, MPS types VII and IV, ISSD, Salla disease, galactosialidosis, Gaucher disease type II, GM1 gangliosidosis, I-cell disease, Niemann-Pick disease types A
and C, Wolman disease, and Farber’s disease (StaretzChacham et al, 2009). The mechanisms of edema are
unclear. Furthermore, not all of the 13 LSDs routinely
appear in the neonatal period.
Directed analysis of urine is helpful for conditions in
which characteristic metabolites are excreted in urine.
One- or two-dimensional electrophoresis or thin-layer
chromatography can detect excess excretion of urine glycosaminoglycans, oligosaccharides, or free sialic acid, but
all urinary tests for the diagnosis of lysosomal storage
disorders can have false-negative results. Examination of
bone marrow or other tissues may demonstrate storage
macrophages in Gaucher disease and in Niemann-Pick
disease types A and C. Small skin or conjunctival biopsy
specimens may demonstrate storage within lysosomes in
most of these disorders.
Definitive diagnosis for all lysosomal storage disorders,
except for Niemann-Pick C disease, is confirmed by enzymatic assays in serum, leukocytes, fibroblasts, or a combination of these. The diagnosis of Niemann-Pick C disease
requires measurement of cellular cholesterol esterification
and documentation of a characteristic pattern of filipincholesterol staining in cultured fibroblasts during LDL
uptake. Analysis of DNA mutations may be helpful for the
diagnosis of Niemann-Pick C disease, Gaucher disease,
and some other conditions, and it will become increasingly
available for other conditions. An imperfect genotype-
phenotype correlation impedes the use of mutation analysis as a prognostic tool. In addition, prenatal diagnosis
is available for most lysosomal storage disorders through
the use of enzyme assays performed on amniocytes or
chorionic villus cells or measurements of levels of stored
substrate in cultured cells or amniotic fluid. As mutation
analysis becomes more prevalent, it will increasingly substitute for biochemical and enzymatic methods.
These conditions must also be considered in the dying
infant, and the neonatologist must be prepared to request
the appropriate samples for diagnosis at the time of death.
In surviving patients, treatment and management must be
considered. All the lysosomal storage disorders are chronic
and progressive conditions for which there is no curative
treatment. Gene transfer therapy holds promise, but is not
currently available for lysosomal storage disorders. With
few exceptions, current standard medical management
is supportive and palliative. Patients must be continually
reassessed for evidence of disease progression and associated complications. These complications manifest at variable ages and can include hydrocephalus, valvular heart
disease, joint limitation, and obstructive airway disease.
For several disorders, particularly neonatal Gaucher
disease and Niemann-Pick C disease, splenectomy may be
indicated to improve severe anemia and thrombocytopenia. This procedure enhances the risk of serious infections,
and it can accelerate the progression of disease at other
sites. Patients with Krabbe disease may have significant
pain of radiculopathy and spasms, and alleviation of that
pain is important for the patient’s comfort. The administration of glutamic acid transaminase inhibitor, vigabatrin,
has been used in a small number of patients with Krabbe
disease, because part of the pathology may involve a secondary deficiency of γ-aminobutyric acid (Barth, 1995).
Low-dose morphine has also been reported to improve
the irritability associated with this disorder (Stewart et al,
2001).
Enzyme replacement therapy with imiglucerase (Cerezyme), a recombinant enzyme, is available for Gaucher
disease. Although enzyme replacement therapy has successfully reversed many of the systemic manifestations of
the disease, it has been suggested that enzyme replacement therapy should not be given to patients with Gaucher disease type 2 who already have severe neurologic
signs, because no substantial improvement has been demonstrated to occur in the neurologic symptoms of patients
treated (Erikson et al, 1993; Gaucher disease, 1996).
Bone marrow transplantation has been tried for a variety of lysosomal storage disorders. The rationale for the
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
procedure is that circulating blood cells derived from
the transplanted marrow become a source of the missing
enzyme. Results of bone marrow transplantation in disorders of glycosaminoglycans show that after successful
engraftment, leukocyte and liver tissue enzyme activity
normalizes, organomegaly decreases, and joint mobility increases. Skeletal abnormalities stabilize but do not
improve. Whether brain function can be improved in
patients with CNS disease remains questionable. Some
patients maintained their learning capability or intelligence quotient, but others continued to deteriorate. Clinical experience and studies in animal models indicate that
BMT before the onset of neurologic symptoms can prevent or delay the occurrence of symptoms, whereas there
is no clear benefit if transplantation is performed when
symptoms are already present (Hoogerbrugge et al, 1995).
BMT in patients with nonneuropathic Gaucher disease can
result in complete disappearance of all symptoms; however,
the procedure is associated with significant risks (Hoogerbrugge et al, 1995) that must be balanced against lifelong
enzyme replacement therapy. Currently it is unclear to
what extent patients with the neuropathic types of Gaucher disease (types 2 and 3) would benefit from transplantation; therefore it is generally not recommended.
BMT has also been attempted in a small number of
patients with infantile Krabbe disease, Farber lipogranulomatosis, and Niemann-Pick A disease. The outcome after
transplantation for these few patients has been poor, with
continued disease progression and death. Krivit et al (2000)
reported successful long-term bone marrow engraftment
in a patient with Wolman disease that resulted in normalization of peripheral leukocyte lysosomal acid lipase
enzyme activity. The patient’s diarrhea resolved; cholesterol, triglyceride; and liver function values normalized; and the patient attained developmental milestones.
Lysosomal storage diseases are not all equally amenable
to BMT, and the use of BMT as a treatment modality for
most lysosomal storage disorders remains uncertain. In a
small number of cases, BMT has been performed in utero
after prenatal diagnosis showing an affected infant, and
experimental protocols are available for families who wish
to pursue this option.
The goal of therapy for a dietary protocol proposed for
the treatment of Wolman disease is reduced accumulation of storage material in intestine and phagocytes. The
diet, which should be started as soon as the diagnosis of
Wolman disease is suggested, consists of (1) discontinuing breastfeeding or feeding with a formula containing
triglycerides and cholesterol esters and (2) keeping the
infant on a fatty ester–free diet (Wolman, 1995). The
diet should include all necessary vitamins, including fat-
soluble vitamins. In addition, daily smearing of the skin of
a different extremity with a small amount (10 to 50 μL) of
sunflower or safflower oil or preferably soy, canola, flax,
cod liver, or algal oil is required for preventing essential
fatty acid deficiency, which complicates the restricted diet
(Wolman, 1995). The absorption of fatty acids through
skin spares the gastrointestinal tract from accumulation
and is associated with the formation of phospholipids and
triglycerides (Wolman, 1995). Preliminary results of this
approach suggest that treatment appears to halt disease
progression.
249
CONGENITAL DISORDERS
OF GLYCOSYLATION
ETIOLOGY
Previously called carbohydrate-deficient glycoprotein syndromes, CDGs are a large and increasing family of genetic
diseases resulting from deficient glycosylation of glycoconjugates, mainly glycoproteins and glycolipids. Most
extracellular proteins, such as serum proteins (e.g., transferrin and clotting factors), most membrane proteins, and
several intracellular proteins (e.g., lysosomal proteins), are
glycosylated proteins. Glycosylation, the addition of sugar
chains (glycans) to proteins, occurs in every human cell and
serves a number of functions, including aiding in correct
folding of the nascent protein, participating in cell adhesion phenomena, protecting against premature proteolytic
destruction, and modifying biologic function (Grünewald,
2007). The glycans are defined by their linkage to the protein N-glycan or O-glycan. N-Glycosylation consists of
the assembly of a glycan on and in the endoplasmic reticulum and its attachment to a particular asparagine of target
proteins, followed by remodeling of this glycan mainly in
Golgi (Jaeken and Matthijs, 2007); therefore it is a twopart process of assembly and processing. O-Glycosylation
consists of assembly of a glycan and its attachment to a
serine or threonine of a target protein, or the attachment
of a monosaccharide (mannose, fructose, or xylose) to one
of these amino acids. No processing pathway is present in
O-glycosylation (Jaeken and Matthijs, 2007). Combined
N- and O-glycosylation defects and lipid glycosylation
defects have also been described (Jaeken and Matthijs,
2007).
Given the ubiquitous occurrence of glycoproteins and
the number of apparent genes involved in glycosylation
(more than 200; approximately 1% of the human genome),
it is not surprising that the number of described CDG
defects is increasing rapidly and clinical manifestations
are diverse (Jaeken, 2006; Jaeken et al, 2008; Morava et
al, 2008b). Currently, 21 disorders in protein N-glycosylation, 12 in protein O-glycosylation, five in both protein
N- and O-glycosylation, and two in lipid glycosylation
have been described (Jaeken, 2006; Jaeken and Matthijs, 2007; Marklová and Albahri, 2007). The number of
described disorders is expected to continue to grow.
The disorders of N-glycosylation have been divided
into two primary categories. CDG-I disorders result from
defects in N-glycan assembly (designated CDG-Ia to
-Im). On isoelectrofocusing of serum transferrin, the most
widely used screening test for N-glycosylation disorders, a
type I pattern is observed. This pattern is characterized by
a decrease of anodal fractions and an increase of disialotransferrin and asialotransferrin. A type II pattern, showing an increase of trisialofractions, monosialofractions, or
both is seen in CDG-II defects, which represent defects in
N-glycan processing (designated CDG-IIa-IIf).
O-Glycosylation defects have been found to be causative
in a number of muscular dystrophies with reduced glycosylation of α-dystroglycan. These disorders, collectively
referred to as α-dystroglycanopathies, encompass previously
described disorders, such as Walker-Warburg syndrome,
muscle-eye-brain disease, Fukuyama congenital muscular
250
PART VI Metabolic and Endocrine Disorders of the Newborn
dystrophy, and limb-girdle muscular dystrophy (Mercuri et al, 2009; Topaloglu, 2009). Six known or putative glycosyltransferase genes have been identified in
these disorders (Godfrey et al, 2007). In general, this is
a heterogeneous group of autosomal recessive disorders
with a wide spectrum of clinical severity, and it shares
the common pathologic feature of hypoglycosylated
α-dystroglycan (Godfrey et al, 2007). α-Dystroglycan is
a major component of the dystrophin-associated glycoprotein complex that forms a link between the actin-
associated cytoskeleton and extracellular matrix. It is a
highly glycosylated peripheral membrane protein that
binds many of its extracellular matrix partners through
its carbohydrate modifications (Godfrey et al, 2007). In
the dystroglycanopathies, these modifications are either
absent or reduced, resulting in decreased binding of
ligands (Barresi and Campbell, 2006). O-Glycosylation
defects have also been described in hereditary multiple
exostoses syndrome, familial tumoral calcinosis, Schneckenbecken dysplasia, spondylocostal dysostosis type 3,
Peters plus syndrome, and the progeria variant of EhlersDanlos syndrome (Grünewald, 2007; Jaeken et al, 2008;
Jaeken and Matthijs, 2007).
Combined N- and O-glycosylation defects are important because they appear to affect trafficking in the glycosylation machinery (Grünewald, 2007). The disruption
of multiple glycosylation pathways is caused by mutations
of the conserved oligomeric Golgi (COG) complex. This
large complex spanning eight subunits plays a key role in
protein transport between the endoplasmic reticulum and
Golgi and within the Golgi complex (Grünewald, 2007;
Jaeken, 2006). COG7 deficiency, first described in two siblings with poor intrauterine growth, dysmorphic features,
encephalopathy, cholestatic liver disease, and perinatal
asphyxia, shows a partial combined N- and O-glycosylation defect caused by decreased transport of CMP-sialic
acid and UDP-galactose into Golgi and reduced activity
of two glycosyltransferases involved in the galactosylation
and sialylation of O-glycans (Jaeken, 2006). An autosomal
recessive cutis laxa syndrome has recently been found to
be also associated with a combined glycosylation defect
(Morava et al, 2008a).
Two disorders of lipid glycosylation have been
described. Amish infantile epilepsy was the first identified
and is caused by a defect of lactosylceramide α-2,3 sialyltransferase (GM3 synthase; Jaeken, 2006). This enzyme
catalyzes the initial step in the biosynthesis of most complex gangliosides from lactosylceramide (Jaeken and
Matthijs, 2007). The defect causes accumulation of lactosylceramide associated with decreased gangliosides of
the GM3 and GD3 series (Jaeken, 2006). Glycosylphosphatidylinositol deficiency is the second disorder in glycolipid glycosylation described, but the first reported
genetic defect in glycosylation of the glycosylphosphatidylinositol (GPI) anchor (Jaeken and Matthijs, 2007).
Glycosylphosphatidylinositol-anchored proteins have
heterogeneous functions as enzymes or adhesion molecules. Finally, the term CDG-x indicates syndromes with
strong evidence for a glycosylation defect, but in which
the defective gene has yet to be identified (Jaeken and
Matthijs, 2007). This clinically heterogeneous group is
also growing rapidly.
CLINICAL FEATURES
The phenotypic spectrum of CDG defects is extremely
broad and ranges from mild to severe disease and from a
single-organ system to multisystem disease. Clinical features alone are insufficient to define the CDG subtype.
CDG should be considered a possible diagnosis in any
unexplained clinical condition, but especially in multiorgan disease with neurologic involvement. Discussion of
the clinical presentation of all forms of CDG is beyond the
scope of this chapter (Table 23-2).
CDG-Ia is the classic and most common presentation.
The basic defect in CDG-Ia is a deficiency of the enzyme
phosphomannomutase, which is required for the early
steps of protein glycosylation (Jaeken, 2006; Jaeken et al,
2001; van Schaftingen and Jaeken, 1995). Patients with
CDG-Ia at birth exhibit dysmorphic features consisting of
a high nasal bridge, prominent jaw, large ears, and inverted
nipples, feeding difficulties, and subsequent growth failure, hypotonia, lipocutaneous abnormalities (including
prominent fat pads on the buttocks), and mild to moderate
hepatomegaly. The clinical progression of this disorder is
divided into four stages. In stage I—the infantile, multisystem stage—patients show evidence of multisystem involvement, including variable strokelike episodes, thrombotic
disease, liver dysfunction, pericardial effusions and cardiomyopathy, proteinuria, and retinal degeneration. The
coagulopathy likely stems from the number of clotting
and anticlotting proteins that are N-linked glycoproteins.
Mental retardation, peripheral neuropathy, and decreased
nerve conduction velocities are observed. Strabismus and
alternating esotropia are present in almost all patients, and
retinitis pigmentosa and abnormalities of the electroretinogram are present in most. Cranial imaging shows varying
degrees of cerebral, cerebellar, and brainstem hypoplasia.
Electroencephalogram results are usually normal. Liver
biopsy samples typically show steatosis and fibrosis, and
multicystic changes in kidneys have been noted.
Stage II, the childhood stage, is characterized by ataxia
and mental retardation. Skeletal abnormalities may
become more prominent, consisting of contractures,
kyphoscoliosis, pectus carinatum, and short stature. Stage
III, generally occurring in the teenage years, is characterized primarily by lower extremity atrophy. Adulthood, or
stage IV, is characterized by hypogonadism. In general,
patients have an extroverted disposition and happy appearance. Approximately 20% of patients die during the first
year of life because of severe infection, liver failure, or cardiac insufficiency.
Patients with CDG-Ib are unique among patients with
these disorders. They may have vomiting, diarrhea, hypoglycemia, and liver disease (coagulopathy, hepatomegaly,
hepatic fibrosis). In addition, they often have a proteinlosing enteropathy (also seen in CDG-Ih; Jaeken and Matthijs, 2007). Development is normal. The remaining forms
of CDG-I and CDG-II are similar in presentation to type
Ia. Additional features include seizures, normal cerebellar
development, delayed myelination, optic atrophy, blindness, frequent infections, hypoventilation and apnea, and
further dysmorphic features such as adducted thumbs,
high-arched palate, coarse facies, widely spaced nipples,
and low-set ears.
251
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
TABLE 23-2 Common Congenital Disorders of Glycoprotein
Types of CDG
Findings
Ia
Ib
Ic
Id
Ie
IIa
IIb
x
Enzyme
defect
Phosphomannomutase
Phosphomannose
isomerase
α1,3Glucosyltransferase
α1,3Mannosyl
transferase
Dol-P-Man
synthase
GlcNAc
transferase 2
Glucosidase I
Unknown
Dysmorphic
features
+
+/–
+/–
+/–
+
+
+
+
Psychomotor
retardation
+
–
+
+
+
+
+
+
Hypotonia
+
+/–
+
+
+
+
+
+
Cerebellar
hypoplasia
+
–
+/–
–
+/–
–
–
–
+/–
+/–
+/–
+
+
+
+
–
–
Strabismus
Optic atrophy
Cortical
blindness
–
–
–
Seizures
Eye findings
Strabismus,
esotropia
Liver disease
+
+
–
–
+
+
+
–
Coagulopathy
+
+
+
–
+
+
+
–
Multiorgan
involvement,
peripheral
neuropathy,
subcutaneous fat
distribution,
inverted
nipples,
strokelike
episodes,
cardiomyopathy,
ataxia,
microcephaly,
hypothyroidism
Proteinlosing
enteropathy, cyclic
vomiting,
diarrhea,
hypoglycemia
Microcephaly,
feeding
difficulties,
ataxia
Stereotype
behavior,
frequent
infections,
ventricular septal
defect,
widely
spaced
nipples,
delayed
myelination
Early death,
generalized
edema,
hypoventilation,
apnea,
demyelinating
polyneuropathy
Leukocyte
adhesion
deficiency
syndrome
type II
(guanosine
diphosphate–
fucose
transporter);
phenotype:
elevated
peripheral
leukocytes,
absence
of CD 15,
Bombay
blood
group
phenotype,
failure to
thrive,
recurrent
infections,
short arms
and legs,
simian
crease
Other
Microcephaly,
reduced
responsiveness,
adducted
thumbs
Microcephaly,
delayed
myelination
Adapted from Westphal V, Srikrishna G, Freeze H: Congenital disorders of glycosylation: Have you encountered them? Genet Med 2:329-337, 2000.
+, Present; –, absent; +/–, occasionally present.
The phenotypic presentation of dystroglycanopathy
is extremely variable. At the severe end of the spectrum
are individuals with Walker-Warburg syndrome, muscleeye-brain disease, and Fukuyama congenital muscular dystrophy. These conditions are characterized by congenital
muscular dystrophy with severe structural brain and eye
abnormalities and death typically before the age of 1 year
(Godfrey et al, 2007; Jaeken, 2006; Jaeken and Matthijs,
2007). Toward the more mild end of the spectrum are
individuals in adulthood with limb-girdle muscular dystrophy with no brain or eye involvement (Godfrey et al, 2007;
Jaeken and Matthijs, 2007). Intermediate phenotypes lie
between the two extremes.
The presence of carbohydrate-deficient transferrin in
serum and cerebrospinal fluid is a distinctive biochemical
feature of CDG. Laboratory findings are often nonspecific
(e.g. elevated liver function studies or hypoalbuminemia),
but concentrations of plasma glycoproteins such as α1antitrypsin, thyroxin-binding globulin, and transferrin are
frequently abnormal (Marklová and Albahri, 2007). Clotting factors such as factors V, XI, II, X, antithrombin III,
proteins C and S, and thyroid hormones (triiodothyronine,
thyroxine, and reverse T3) are also frequently decreased
(Marklová and Albahri, 2007; Morava et al, 2008b). Abnormal clotting is an important indicator of a glycosylation
disorder (Morava et al, 2008b).
252
PART VI Metabolic and Endocrine Disorders of the Newborn
DIAGNOSIS
CDG should be considered in newborns with several of
the following features:
ll Neurologic signs, including hypotonia, hyporeflexia, or seizures
ll Ophthalmic signs, including abnormal eye movements, cataracts, glaucoma, optic nerve atrophy, or
retinitis pigmentosa
ll Hepatic and gastrointestinal signs such as ascites
or hydrops, hepatomegaly, diarrhea, and proteinlosing enteropathy
ll Endocrinologic signs, including hyperinsulinemic
hypoglycemia and hypothyroidism
ll Signs of renal or cardiac disease
ll Congential muscular dystrophy
ll Congenital joint contractures
ll Dysmorphic
features, microcephaly, structural
brain anomalies, or abnormal skin findings
Isoelectrofocusing of serum transferrin is the screening method of choice, but only to detect defects of
N-glycosylation. Confirmatory enzyme assays or molecular studies are required to pinpoint the specific defect.
Urine oligosaccharide analysis, Bombay blood phenotype, serum apoC-III screening, or membrane bound
sialyl-LewisX antigen may be helpful in distinguishing
subclasses of CDG II disease (Marklová and Albahri,
2007). If suspicion of CDG remains, further structural
analysis of the lipid-linked oligosaccharide (LLO) and the
peptide-protein N-linked oligosaccharide should follow,
if available (Marklová and Albahri, 2007). Patients with
CDG can often be identified through neonatal screening for congenital hypothyroidism, because of an associated thyroid-binding globulin deficiency and an increased
thyroid-stimulating hormone level. Prenatal diagnosis
by transferrin isoelectric focusing is not reliable. Prenatal diagnosis is possible in all types of CDG for which the
molecular defect is known (Grünewald, 2007). The vast
majority of CDG disorders are autosomal recessive disorders or presumed autosomal recessive disorders; hereditary multiple exostoses syndrome is autosomal dominant.
TREATMENT AND MANAGEMENT
The treatment and management for most types of CDGs
are primarily supportive and palliative. There is no curative or corrective treatment. In infancy, evidence of multisystem involvement and the resulting complications must
be treated promptly. There is substantial mortality in the
first years of life because of severe infection or vital organ
failure (Grünewald, 2007; Jaeken, 2006). The exception is
treatment of CDG-Ib with oral mannose therapy. In this
disorder, oral mannose effectively bypasses the impaired
pathway and allows glycosylation to continue (Freeze,
1998; Jaeken, 2006; Jaeken and Matthijs, 2007). Therapy
improves the protein-losing enteropathy and liver disease
(Grünewald, 2007). Oral fucose therapy has also been
used in patients with CDG-IIc, a GDP-fucose transporter
defect (Grünewald, 2007). Therapy appears to improve
the fucosylation of glycoproteins and to improve control
of recurrent infections, but it has no effect on neurologic
complications of the disorder (Grünewald, 2007).
PEROXISOMAL DISORDERS
DISORDERS OF PEROXISOME BIOGENESIS
Peroxisomes are single-membrane–bound cellular organelles that contain no internal structure or DNA and are
characterized by an electron-dense core and a homogeneous
matrix. Peroxisomes are found in all cells and tissues except
mature erythrocytes, and they are in highest concentration in
the liver and kidneys. They are formed by growth and division of preexisting peroxisomes and are randomly destroyed
by autophagy. Their half-life is 1.5 to 2 days. Peroxisomal
proteins are encoded by nuclear genes, synthesized in cytosol, and imported posttranslationally into the peroxisome.
The import of proteins into the peroxisome is mediated by
receptors and requires adenosine triphosphate hydrolysis.
Peroxisomes contain enzymes that use oxygen to oxidize
a variety of substrates, thereby forming peroxide. The peroxide is decomposed within the organelle by the enzyme
catalase to water and oxygen. This process protects the cell
against peroxide damage through compartmentalization
of peroxide metabolism within the organelle. Peroxisomes
can also function to dispose of excess reducing equivalents
and may contribute to thermogenesis, producing heat
from cellular respiration (Gould et al, 2007).
More than 50 enzymes have been found within peroxisomes (Gould et al, 2001). The proteins have multiple
functions, both synthetic and degradative (Wanders et al,
2001). The primary synthetic functions are plasmalogen
synthesis and bile acid formation. Plasmalogens constitute
5% to 20% of phospholipids in cell membranes and 80%
to 90% of phospholipids in myelin. They are involved in
platelet activation and may also protect cells against oxidative stress. Degradative functions include (1) β-oxidation of
VLCFA (≥C23), fatty acids (down to C8 to C6), long-chain
dicarboxylic acids, prostaglandins, and polyunsaturated fatty
acids; (2) oxidation of bile acid intermediates, pipecolic acid
and glutaric acid (intermediates in lysine catabolism), and
phytanic acid; (3) deamination of d- and l-amino acids,
(4) metabolism of glycolate to glyoxylate; (5) polyamine degradation (spermine and spermidine); and (6) ethanol clearance.
At least 16 conditions caused by single peroxisomal enzyme
deficiencies have been confirmed (Wanders et al, 2007).
Peroxisomal disorders constitute a clinically and biochemically heterogeneous group of inherited diseases that result
from the absence or dysfunction of one or more peroxisomal
enzymes. Conditions in which multiple peroxisomal enzymes
are affected can result from a disturbance of biogenesis or
the organelle. Pathophysiology apparently involves either
deficiency of necessary products of peroxisomal metabolism
or excess of unmetabolized substrates. Disorders with similar biochemical defects may have markedly different clinical
features, and disorders with similar clinical features may be
associated with different biochemical findings. General features of peroxisomal disorders, each of which can manifest or
be evident in the newborn period, are as follows:
ll Dysmorphic craniofacial features
ll Neurologic dysfunction, primarily consisting of
severe hypotonia, possibly associated with hypertonia of the extremities and seizures
ll Hepatodigestive dysfunction, including hepatomegaly, cholestasis, and prolonged hyperbilirubinemia
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
Rhizomelic shortening of the limbs, stippled calcifications of epiphyses, renal cysts, and abnormalities in neuronal migration may also be seen.
Peroxisomal biogenesis disorders are composed of at
least 12 complementation groups (Matsumoto et al, 2001).
All involve defects in proteins targeted to the organelle.
Genes and proteins required for peroxisomal biosynthesis
are referred to as peroxins and are encoded by PEX genes.
The PEX genes responsible for disease in most human
patients are known, and more than 50% of patients with
peroxisomal biogenesis disorders have mutations in PEX1
(Moser, 2000; Steinberg et al, 2006). This section We discusses the peroxisomal disorders that can manifest in the
newborn period.
Zellweger syndrome is the prototype of neonatal peroxisomal disease. It is a disorder of peroxisome biogenesis
caused by failure to import newly synthesized peroxisomal
proteins into the peroxisome. The proteins remain in the
cytosol, where they are rapidly degraded. In this condition, peroxisomes are absent from liver hepatocytes or
exist as “ghosts.” Neonatal adrenoleukodystrophy and
infantile Refsum disease are also disorders of peroxisome
biogenesis in which, as in Zellweger syndrome, disruption of function of more than one peroxisomal enzyme
is demonstrable. A few residual peroxisomes, however,
may be seen in the liver. These disorders represent a continuum of clinical severity. Rhizomelic chondrodysplasia
punctata is caused by a defect in a subset of peroxisomal
enzymes. In this disorder, liver peroxisomes are demonstrable and normal in number, but their distribution and
structure are abnormal.
253
There is circumstantial evidence that in utero elevations
of VLCFAs may be key to congenital CNS abnormalities.
Powers and Moser (1998) proposed that VLCFAs and
phytanic acid that accumulate in these peroxisomal disorders are incorporated into myelin and cell membranes, and
that alteration of normal constituents of the membrane
adversely affects membrane function. Specifically, these
investigators suggest that abnormal constituents accelerate
cell death and impede neuronal migration, accounting for
the conspicuous CNS abnormalities in disorders of peroxisomal biogenesis.
To date, four disorders of peroxisomal fatty acid
β-oxidation have been defined: acetyl-CoA oxidase deficiency, d-bifunctional protein deficiency, peroxisomal
thiolase deficiency, and 2-methylacyl-CoA racemase deficiency (Wanders et al, 2007). The clinical presentation of
the first three disorders resembles that of biogenesis disorders. Individuals with the fourth disorder have a late-onset
neuropathy.
Clinical Presentations
Table 23-3 summarizes the clinical features of disorders of peroxisome biogenesis that can present in the
neonate.
Zellweger Syndrome
Zellweger syndrome is most often evident at birth, with
affected babies having dysmorphic facial features including large fontanels, high forehead, flat occiput, epicanthus, hypertelorism, upward-slanting palpebral fissures,
TABLE 23-3 Disorders of Peroxisomal Biogenesis in the Newborn Period
Feature
Zellweger Syndrome
Neonatal
Adrenoleukodystrophy
Infantile Refsum
Disease
Rhizomelic Chondrodysplasia Punctata
Onset
Birth
Birth to 3 mo
Birth to 6 mo
Birth
Facies
High forehead, large
fontanels, upward-
slanting palpebral fissures,
hypoplastic supraorbital
ridges, epicanthic folds,
micrognathia, abnormal
ears
Milder features of
Zellweger syndrome
Epicanthic folds, midface
hypoplasia, low-set ears
Depressed nasal bridge,
hypertelorism,
microcephaly
Neurologic
findings
Weakness, hypotonia,
seizures, psychomotor
retardation, sensorineural
hearing loss
Hypotonia, seizures,
slow psychomotor development and
neurodegeneration
Mild hypotonia, normal early
development followed
by degeneration, ataxia,
sensorineural hearing loss
Severe psychomotor
retardation
Ophthalmologic
findings
Cataracts, glaucoma,
corneal clouding, retinitis
pigmentosa, optic nerve
dysplasia, Brushfield spots
Retinopathy
Retinitis pigmentosa
Cataracts
Other findings
Hepatomegaly, multicystic kidneys, congenital
heart disease, growth
failure, chondrodysplasia
punctata
Impaired adrenal function
Hepatomegaly, anosmia,
diarrhea
Severe shortening of
proximal limbs, joint
contractures, ichthyosis
Diagnosis
↑ plasma VLCFA, phytanic
acid, pipecolic acid, and
bile acid intermediates,
↓ plasmalogens
Same as for Zellweger
syndrome
Same as for Zellweger
syndrome
↑ phytanic and pipecolic
acids, ↓ plasmalogens,
normal VLCFA and bile
acid intermediates
VLCFA, Very-long-chain fatty acid; ↑, elevated; ↓, reduced.
254
PART VI Metabolic and Endocrine Disorders of the Newborn
hypoplastic supraorbital ridges, abnormal ears, severe
weakness and hypotonia, hepatomegaly, multicystic
kidneys, and congenital heart disease. Seizures, feeding difficulties, and postnatal growth failure soon manifest. Ophthalmologic examination may detect cataracts,
corneal clouding, glaucoma, optic atrophy, retinitis pigmentosa, and Brushfield spots. Somatic sensory evoked
responses and electroretinograms are abnormal. Hearing
assessment often shows an abnormal brainstem auditory
evoked response consistent with sensorineural hearing
loss. Skeletal radiographs demonstrate epiphyseal stippling, and cranial imaging shows leukodystrophy and
neuronal migration abnormalities. Hepatic cirrhosis and
severe psychomotor retardation occur later. Laboratory
analysis may demonstrate abnormal liver function values,
hyperbilirubinemia, or hypoprothrombinemia. Death usually occurs within the first year of life, the average life span
being 12.5 weeks.
Neonatal Adrenoleukodystrophy
Clinically, neonatal adrenoleukodystrophy is similar to,
but less severe than, Zellweger syndrome. Differences
include less dysmorphology, absence of chondrodysplasia punctata and renal cysts, and fewer neuronal and gray
matter changes. Patients with neonatal adrenoleukodystrophy may have striking white matter disease, however,
and often show degenerative changes in adrenal glands.
They also have slow psychomotor development followed
by neurodegeneration that usually begins before the end
of the first year of life. Disease progression is slower than
that observed in Zellweger syndrome, and longer survival is usual, to an average of approximately 15 months
of age.
Infantile Refsum Disease
Patients with infantile Refsum disease also have relatively
mild dysmorphic features, such as epicanthic folds, midface
hypoplasia with low-set ears, and mild hypotonia. Early
neurodevelopment is normal, possibly up to 6 months of
age, but then slow deterioration begins. Later, sensorineural hearing loss (100%), anosmia, retinitis pigmentosa,
hepatomegaly with impaired function, and severe mental
retardation are evident. Patients learn to walk, although
their gait may be ataxic and broad based. Diarrhea and
failure to thrive may also be seen. Chondrodysplasia punctata and renal cysts are absent. Neuronal migration defects
are minor, and adrenal hypoplasia occurs. The life span of
patients with infantile Refsum disease ranges from 3 to 11
years.
Rhizomelic Chondrodysplasia Punctata
Patients with rhizomelic chondrodysplasia punctata at
birth have facial dysmorphia, microcephaly, cataracts,
rhizomelic shortening of extremities with prominent
stippling, and coronal clefting of vertebral bodies. The
chondrodysplasia punctata is more widespread than in
Zellweger syndrome and may involve extraskeletal tissues.
Infants with this disorder have severe psychomotor retardation from birth onward and severe failure to thrive. In
addition, patients may have joint contractures, and 25%
experience ichthyosis. Neuronal migration is normal. Life
span is usually less than 1 year.
DISORDERS OF PEROXISOMAL β-OXIDATION
d-Bifunctional protein deficiency is more rare than peroxisomal biogenesis disorders and results in a phenotype
similar to Zellweger syndrome. In general, children have
severe CNS involvement consisting of profound hypotonia, uncontrolled seizures, and failure to acquire any significant developmental milestones. Children are usually
born full term without evidence of intrauterine growth
restriction. Dysmorphic features, similar to those seen in
Zellweger syndrome, are notable in most children. In most
cases, neuronal migration is disturbed with areas of polymicrogyria and heterotopic neurons in the cerebrum and
cerebellum. Death generally occurs before 1 year of age,
but survival to at least 3 years of age is possible.
Acetyl-CoA oxidase deficiency is less common. Patients
exhibit global hypotonia, deafness, and delayed milestones
with or without facial dysmorphic features. Patients may
demonstrate early developmental gains, but then show
regression of skills. Retinopathy with extinguished electroretinograms, failure to thrive, hepatomegaly, areflexia,
and seizures have also been reported.
One patient with peroxisomal thiolase deficiency has
been described (Goldfischer et al, 1986). The child had
marked facial dysmorphia, muscle weakness, and hypotonia. She demonstrated no psychomotor development
during her 11 months of life. Autopsy showed renal cysts,
atrophic adrenal glands, minimal liver fibrosis, hypomyelination in cerebral white matter, foci of neuronal heterotopia, and a sudanophilic leukodystrophy (Goldfischer
et al, 1986).
X-linked adrenoleukodystrophy is the most common
peroxisomal disorder, because of altered function of a
membrane transport protein ABCD1 that affects metabolism of VLCFAs, and does not usually present in the neonatal period. However, three patients with contiguous
deletions involving the ABCD1 gene have exhibited a phenotype similar to peroxisomal biogenesis disorders (Corzo
et al, 2002).
DIAGNOSIS, MANAGEMENT, AND
PROGNOSIS OF PEROXISOMAL DISORDERS
The key to diagnosing peroxisomal disease is a high index
of suspicion. Peroxisomal disorders should be considered in newborns with dysmorphic facial features, skeletal abnormalities, shortened proximal limbs, neurologic
abnormalities (including hypotonia or hypertonia), ocular abnormalities, and hepatic abnormalities. Babies with
abnormal visual, hearing, or somatosensory evoked potentials should also be considered for these diagnoses.
Peroxisomal disorders are not associated with acute metabolic derangements or abnormal routine laboratory tests.
Measurements of VLCFAs, phytanic acid, pipecolic acid,
bile acid intermediates, and plasmalogens are required for
diagnosis. Zellweger syndrome is associated with elevations
of VLCFAs, phytanic acid, pipecolic acid, and bile acid
intermediates, and a decrease in plasmalogen synthesis.
Neonatal adrenoleukodystrophy and infantile Refsum disease have similar biochemical findings; however, the defect
in plasmalogen synthesis and the degree of VLCFA accumulation are less severe. Laboratory findings in rhizomelic
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
chondrodysplasia punctata are elevations of phytanic and
pipecolic acids, a decrease in plasmalogen, and normal
levels of VLCFAs and bile acid intermediates. Therefore
screening that uses only levels of VLCFAs fails to detect
rhizomelic chondrodysplasia punctata. d-Bifunctional
protein deficiency is associated with deficient oxidation of
C23:0 and pristanic acid, leading to elevations of pristanic
acid and, to a lesser extent, phytanic acid. This deficiency
results in an elevated pristanic acid-to-phytanic acid ratio,
which is generally not elevated in peroxisomal biogenesis
disorders. Abnormal VLCFA and elevation of varanic acid
(an intermediate metabolite in β-oxidation) are also seen.
Accumulation of bile acid intermediates is a variable finding. Abnormalities in phytanic acid and plasmalogens are
age dependent. The elevation of phytanic acid might not
be demonstrable in young infants, and reduction in red
blood cell plasmalogen levels may not be evident in children older than 20 weeks (Gould et al, 2007). A liver biopsy
may be a useful adjunct diagnostic tool to assess for the
presence or absence and structure of peroxisomes. Definitive diagnoses for all types of peroxisomal disease require
cultured skin fibroblasts for measurement of VLCFA
levels and their β-oxidation and, as needed, assay of the
peroxisomal steps of plasmalogen synthesis, phytanic acid
oxidation, subcellular localization of catalase, enzyme
assays, and immunocytochemistry studies. Prenatal diagnosis with a variety of methods is available (Steinberg et al,
2005). DNA diagnosis is possible for most patients, but is
“challenging for the Zellweger syndromes spectrum since
12 PEX genes are known to be associated with this spectrum of peroxisomal biogenesis disorders” (Steinberg et al,
2006). DNA study for deletions also has a role in diagnostic
evaluations in some cases before counseling for recurrence
risk, as demonstrated by the neonatal presentation of cases
with deletion of the ABCD1 gene on the X chromosome
(Corzo et al, 2002). The complexities of prenatal diagnosis
emphasize the need for studies of tissue on the proband to
determine the potential for and appropriate strategies to
offer in prenatal diagnosis for any family.
One of the more interesting recent developments in
peroxisomal disease is the preliminary work on combined
liquid chromatography-tandem mass spectroscopy application for blood spot–based newborn screening. It is targeted at conditions with abnormal VLCFAs, especially
X-linked adrenoleukodystrophy, that typically manifest in
childhood and for which presymptomatic therapy may alter
the course of the usual progressive disease. The method
is promising, but still under investigation (Hubbard et al,
2006).
Treatment for all peroxisomal disorders in the newborn
period remains supportive. These disorders are chronic,
progressive diseases with no currently available curative therapy. Setchell et al (1992) described the effects
of administration of primary bile acids on liver function
in a 6-month-old infant with Zellweger syndrome. The
effects included normalization of serum bilirubin and liver
enzyme levels and a decrease in hepatic inflammation, bile
duct proliferation, and canalicular plugs. The patient also
showed an improvement in growth and neurologic function (Setchell et al, 1992).
Martinez (1992) reported the use of docosahexaenoic acid ethyl ester in two patients with neonatal
255
adrenoleukodystrophy. Both patients had an increase
in erythrocyte omega fatty acid levels and plasmalogens accompanied by significant improvement in clinical parameters, including alertness, motor performance,
vocabulary, and visual evoked responses. Martinez et al
(2000) also reported on the effects of docosahexaenoic
acid supplementation in 13 patients with generalized peroxisomal disorders. The effects were normalization of
blood docosahexaenoic acid levels, increased plasmalogen
concentrations, decreased plasma VLCFAs, and improvement to near normal of liver enzymes. Although patients
with severe neonatal Zellweger syndrome presentation did
not benefit, patients with milder peroxisomal biogenesis
disorders—in which clinical course is more variable—
experienced improvement in vision, liver function, muscle
tone, and social contact. Three patients showed normalization of brain myelin, and myelination improved in three
others (Martinez et al, 2000).
In addition, a “triple” dietary approach consisting of
oral administration of ether lipids, decreased phytanic
acid intake, and the oral administration of glyceryl trioleate and glyceryl trierucate (Lorenzo’s oil) leads to biochemical improvement and may have value in patients
with mild forms of peroxisomal biogenesis defects (Gould
et al, 2007). Although treatment protocols are available for
infants affected with disorders of peroxisomal biogenesis,
improvement in long-term outcome remains limited in
all but the mildly affected patients. However, treatment
involving diet and bone marrow transplant, when appropriate, has been demonstrated to significantly improve the
outcome of some patients affected with adrenoleukodystrophy (Moser et al, 2001).
SMITH-LEMLI-OPITZ SYNDROME
ETIOLOGY
Smith-Lemli-Opitz syndrome is a well-recognized autosomal recessive malformation syndrome, with an estimated incidence ranging from 1 in 10,000 to 70,000 in
various populations (Porter, 2008; Yu and Patel, 2005).
Because of the identification of an underlying biochemical defect, SLO syndrome has been reclassified as an
inborn error of metabolism. In 1993, it was discovered
that SLO syndrome is caused by a defect in cholesterol
biosynthesis that results in low levels of cholesterol and
elevated levels of 7-dehydrocholesterol (7DHC) and its
isomer, 8-dehydrocholesterol (8DHC). Patients have
markedly reduced activity of the enzyme 7DHC reductase, the enzyme responsible for conversion of 7DHC to
cholesterol (Porter, 2008; Salen et al, 1995; Waterham
and Clayton, 2006), which is located on chromosome 11
(Kelley and Hennekam, 2000; Waterham and Clayton,
2006). Cholesterol is a major lipid component of cellular
membranes such as myelin, and it is an important structural component of lipid rafts, which play a major role in
signal transduction (Porter, 2008; Yu and Patel, 2005). In
addition, bile acids, steroid hormones, neuroactive steroids, and oxysterols are all synthesized from cholesterol
(Merkens et al, 2009; Porter, 2008). The possible role of
these various pathways in the etiology of SLO syndrome
is still being defined (Merkens et al, 2009; Porter, 2008).
256
PART VI Metabolic and Endocrine Disorders of the Newborn
The cause of the clinical phenotype of SLO syndrome may
be related to deficient cholesterol, deficient total sterols,
the toxic effects of either 7DHC or compounds derived
from 7DHC, or a combination of these factors (Porter,
2008; Yu and Patel, 2005). A single underlying pathologic
mechanism is unlikely (Porter, 2008).
The pivotal connection between cholesterol and SLO
syndrome involves development and differentiation of the
vertebrate body plan, although additional metabolic effects
have not been ruled out. Among those proteins exerting
decisive influence on patterning during embryogenesis
are the Hedgehog proteins. One variant, Sonic hedgehog
(Shh), becomes covalently linked to cholesterol at the protein’s amino-terminal signaling domain. This linkage is
needed to restrict the locus of action of Shh to the region
of plasma membrane. Although it was originally suggested
that failure to modify Shh could account for multiplicity
of structural abnormalities in patients with SLO syndrome
(Farese and Herz, 1998), the process is thought to be more
complex and to involve other signaling proteins such as
Patched (PTCH) and Smoothened (SMO) (Farese and
Herz, 1998; Kelley and Hennekam, 2000; Porter, 2008).
CLINICAL FEATURES
Recognition of the biochemical defect in SLO syndrome
provided the diagnostic test required to recognize the most
mild and most severe cases, substantially expanding the
clinical spectrum of the condition. Classic SLO syndrome
is often evident at birth; affected patients have microcephaly and facial dysmorphism, including bitemporal narrowing, ptosis, epicanthic folds, anteverted nares, broad
nasal tip, prominent lateral palatine ridges, micrognathia,
and low-set ears. Other features are 2- to 3-syndactyly of
toes (found in 95% of patients), small proximally placed
thumbs, and occasionally postaxial polydactyly and cataracts. Males usually have hypospadias, cryptorchidism, and
a hypoplastic scrotum, but may have ambiguous or female
genitalia. Pyloric stenosis, cleft palate, pancreatic anomalies, Hirschsprung disease, and lung segmentation defects
have also been reported. Hypotonia progressing to hypertonia and moderate to severe mental deficiencies are also
present. Feeding difficulties and vomiting are common
problems in infancy. Irritable behavior and shrill screaming may also pose problems during infancy. Older children frequently have hyperactivity, self-injurious behavior,
sleep difficulties, and autistic characteristics. Cranial imaging studies show (and autopsy confirms) defects in brain
morphogenesis, including hypoplasia of frontal lobes, cerebellum, and brainstem, dilated ventricles, irregular gyral
patterns, and irregular neuronal organization.
Historically, approximately 20% of patients die within
the first year of life, although others may survive for
more than 30 years. Life expectancy appears to correlate
inversely with the number and severity of organ defects
and with the kinds and numbers of limb, facial, and genital abnormalities (Kelley and Hennekam, 2000; Tint et al,
1995). Developmental outcomes are also highly variable,
ranging from severe mental retardation to normal intelligence. Development of treatment protocols for SLO
syndrome may contribute to improvements in prognosis,
but improved recognition of more mildly affected patients
may explain the increasing reports of SLO syndrome in
patients with mild mental retardation or normal intelligence (Jezela-Stanek et al, 2008). Testing for SLO syndrome has been suggested for all patients with idiopathic
intellectual impairment, behavioral anomalies, or both,
when associated with nonfamilial two- and three-toe syndactyly and failure to thrive (Jezela-Stanek et al, 2008).
DIAGNOSIS
The diagnosis of SLO syndrome is based on findings of
elevated levels of 7DHC and 8DHC. Plasma cholesterol
levels are usually but not always low; as many as 10% of
patients at all ages have normal cholesterol levels (Kelley
and Hennekam, 2000). In addition, the standard method
for analysis of cholesterol in most hospital laboratories identifies 7DHC and 8DHC as cholesterol. Therefore most laboratories report normal cholesterol levels
in patients who have low cholesterol, but elevations of
7DHC and 8DHC sufficient to bring the total level into
the “normal” range (Kelley and Hennekam, 2000; Porter,
2008). The difference between mild and more severe disease appears to be one of degree; the enzyme defect is
more severe and the block is more complete in patients
with severe disease (Kelley and Hennekam, 2000; Tint et
al, 1995). Clinical severity in SLO syndrome correlates
best with either reduction in absolute cholesterol levels
or the sum of 7DHC plus 8DHC expressed as a fraction
of total sterol (Waterham and Clayton, 2006). Recently,
maternal apolipoprotein E (ApoE) genotype was implicated in phenotype heterogeneity (Witsch-Baumgartner
et al, 2004). Maternal ApoE2 genotypes were associated
with a severe SLO syndrome phenotype, whereas ApoE
genotypes without the E2 allele were associated with a
milder phenotype (Witsch-Baumgartner et al, 2004; Yu
and Patel, 2005).
Confirmation of diagnosis via molecular analysis is
available. Genotype-phenotype correlation, however, is
relatively poor (Jezela-Stanek et al, 2008; Porter, 2008).
Prenatal diagnosis is possible. A mother carrying an
affected fetus may have an abnormally low unconjugated
estriol value. Ultrasonography detects many but not all
affected fetuses, and biochemical analysis of amniotic fluid
and of chorionic villus samples is accurate and unambiguous in most cases (Kelley and Hennekam, 2000). A direct
correlation between the level of 7DHC in amniotic fluid
and clinical severity has been demonstrated; however, a
similar correlation does not exist for the level of cholesterol (Yu and Patel, 2005). When molecular mutations are
known, analysis may be useful.
TREATMENT
There are two goals of therapy for SLO syndrome: to
increase the level of cholesterol in plasma and other body
fluids and to lower the level of 7DHC. Treatment consists of providing exogenous cholesterol, in the form of
either dietary cholesterol or cholesterol suspension, to
replenish body stores of cholesterol and downregulate the
patient’s endogenous cholesterol synthesis, thus decreasing the amount of 7DHC produced. A goal of cholesterol supplementation of 20 to 60 mg/kg per day was
CHAPTER 23 Lysosomal Storage, Peroxisomal, and Glycosylation Disorders and Smith-Lemli-Opitz Syndrome in the Neonate
initially advocated, but doses higher than 300 mg/kg per
day have been used without adverse outcome (Irons et al,
1995; Kelley and Hennekam, 2000). In infants with SLO
syndrome, the use of breast milk should be encouraged
because it supplies approximately 133 mg/L of cholesterol
(Irons et al, 1995).
Providing bile acids to facilitate adequate absorption of dietary cholesterol is controversial, especially
because fat malabsorption is unusual and certain bile
acids can decrease tissue uptake of cholesterol (Kelley and
Hennekam, 2000). Dietary cholesterol supplementation
appears to restore both adrenal and bile salt deficiencies
(Yu and Patel, 2005). Steroid replacement therapy may
still be needed during times of stress or illness. Use of
3-hydroxy-3-methyl-CoA (HMG-CoA) lyase inhibitors
such as lovastatin has also been suggested as a mechanism
to decrease levels of 7DHC. However, because the results
of animal studies suggest that downregulation of cholesterol synthesis might not decrease 7DHC, and because of
concern about a decrease in the synthesis of other essential
isoprenoid compounds, HMG-CoA reductase inhibitors
are not routinely used in therapy (Kelley and Hennekam,
2000). Furthermore, this treatment could be theoretically
detrimental in patients with SLO syndrome with little
or no enzyme activity (Yu and Patel, 2005). Statins may
also impair dietary cholesterol absorption (Merkens et al,
2009). In addition, a retrospective study of simvastatin use
in patients with SLO syndrome failed to demonstrate a
positive effect on anthropometric measures or behavior
(Haas et al, 2007).
Although many questions remain about optimal therapy and outcomes, therapeutic interventions appear to
increase plasma cholesterol levels, decrease 7DHC levels, and improve irritability, behavior, and growth (Elias
et al, 1997; Irons et al, 1995; Kelley and Hennekam, 2000;
Waterham and Clayton, 2006). Parents reported children to be more alert, active, and happier during therapy.
Therapy was well tolerated. Unfortunately, a study of
14 patients with SLO syndrome indicated that cholesterol
supplementation had hardly any effect on developmental
progress (Sikora et al, 2004). Treatment probably does
not significantly change sterol levels in brain which are
dependent on de novo cholesterol synthesis because of
limited ability of cholesterol to cross the blood-brain barrier (Porter, 2008; Waterham and Clayton, 2006). Direct
delivery of cholesterol to the CNS by low-pressure catheter infusions has been proposed, but not tested (Yu and
Patel, 2005). Gene therapy, the use of neuroactive steroids, and inhibition of glycosphingolipids are also being
investigated as possible therapeutic options in SLO syndrome (Merkens et al, 2009).
257
For maximal benefit, it has been suggested that treatment should begin prenatally, because SLO syndrome has
many features that are consistent with in utero involvement of the disease process. Antenatal supplementation by
fetal intravenous and intraperitoneal transfusions of fresh
frozen plasma were shown to increase fetal cholesterol in
one patient (Irons et al, 1999). Treatment should otherwise
begin as soon as possible after birth or as soon as the diagnosis is confirmed. Patients with severe SLO syndrome
may need gavage or gastrostomy feeding for management
of reflux and gastrointestinal dysmotility, and many have
protein allergies and require elemental formulas. Growth
is often a problem, but the temptation to overfeed must be
avoided because overfeeding would contribute to feeding
problems and could not rescue intrauterine growth restriction in severe SLO syndrome (Kelley and Hennekam,
2000).
SUGGESTED READINGS
Barth PG: Sphingolipids. In Fernandes J, Saudubray J-M, van den Berghe G,
editors: Inborn Metabolic Diseases: Diagnosis and Treatment, ed 2, Berlin, 1995,
Springer-Verlag, pp 375-382.
Beutler E, Grabowski GA: Gaucher Disease. In Scriver CR, Beaudet AL, Sly WS,
Valle D, et al, editors: The Metabolic and Molecular Bases of Inherited Disease, ed 8,
New York, 2001, McGraw-Hill, pp 3635-3668.
Fletcher JM: Screening for lysosomal storage disorders: a clinical perspective,
J Inherit Metab Dis 29:405-408, 2006.
Gould S, Raymond G, Valle D: The peroxisome biogenesis disorders. In Valle D,
Beaudet AL, Vogelstein B, et al: The Metabolic and Molecular Bases of Inherited
Disease, ed 8, New York, 2001, McGraw-Hill, pp 3181-3217.
Grünewald S: Congenital disorders of glycosylation: rapidly enlarging group of
(neuro)metabolic disorders, Early Hum Devel 83:825-830, 2007.
Jaeken J: Congenital disorders of glycosylation. In Fenandes J, Saudubray JM, van
den Berghe G, et al: Inborn Metabolic Diseases: Diagnosis and Treatment, ed 4,
Heidelberg, 2006, Springer Medizin Verlag, pp 523-530.
Jaeken J, Matthijs G, Carchon H, et al: Defects of N-glycan synthesis. In Scriver
CR, Beaudet AL, Sly WS, et al: The Metabolic and Molecular Bases of Inherited
Disease, ed 8, New York, 2001, McGraw-Hill, pp 1601-1622.
Meikle PJ, Grasby DJ, Dean CJ, et al: Newborn screening for lysosomal storage
disorders, Mol Genet Metab 88:307-314, 2006.
Moser HW, Smith KD, Watkins PA, et al: X-linked adrenoleukodystrophy. In
Valle D, Beaudet AL, Vogelstein B, et al: The Metabolic and Molecular Bases of
Inherited Disease, ed 8, New York, 2001, McGraw Hill, pp 3257-3301.
Porter FD: Smith-Lemli-Opitz syndrome: pathogenesis, diagnosis, and management, Eur J Hum Genet 16:535-541, 2008.
Staretz-Chacham O, Lang TC, LaMarca ME, et al: Lysosomal storage disorders in
the newborn, Pediatrics 123:1191-1207, 2009.
Stone DL, Sidransky E: Hydrops fetalis: lysosomal storage disorders in extremis,
Adv Pedatr 46:409-440, 1999.
Wanders R, Barth P, Heymans H: Single peroxisomal enzyme deficiencies. In Valle
D, Beaudet AL, Vogelstein B, et al: The Metabolic and Molecular Bases of Inherited
Disease, ed 8, New York, 2001, McGraw Hill, pp 3219-3256.
Waterham HR, Clayton PT: Disorders of cholesterol synthesis. In Fenandes J,
Saudubray JM, van den Berghe G, Walters JH, editors: Inborn Metabolic
Diseases: Diagnosis and Treatment, ed 4, Heidelberg, 2006, Springer Medizin
Verlag, pp 414-415.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
24
Skeletal Dysplasias and Connective
Tissue Disorders
David L. Rimoin and George E. Tiller
The skeletal dysplasias, or osteochondrodysplasias, are
disorders of the development and growth of cartilage and
bone. The connective tissue disorders involve abnormalities of the cells’ supporting and connecting structures in
the matrix. In one series of 126,316 deliveries monitored
over 15 years, the incidence of skeletal dysplasias was
2.14 in 10,000 (Rasmussen et al, 1996). With the growing
use and accuracy of ultrasonography for prenatal care, a
greater number of osteochondrodysplasias and connective
tissue disorders are diagnosed prenatally.
The skeletal dysplasias have been classified into
37 groups on the basis of radiologic criteria (Superti-Furga
and Unger, 2007). Other classifications vary according to
molecular, clinical, pathologic, and radiolographic criteria
and may be confusing. For example, osteogenesis imperfecta (OI) can be classified as either a skeletal dysplasia
or a connective tissue disorder. This chapter focuses on
several of the more common skeletal dysplasias (see Table
24-1 for an expanded list) and connective tissue disorders
that manifest prenatally or perinatally, but the discussion
is not exhaustive. The osteochondrodysplasias have been
reviewed extensively elsewhere (Cohen, 2006; SupertiFurga et al, 2001; Unger et al, 2007).
There are a large number of different connective tissue molecules, including collagens (over two dozen types),
elastin, fibrillin (two types), and microfibril-associated
glycoproteins. These molecules are components of tissues such as bone, cartilage, skin, vascular media, tendon,
ligaments, and basement membrane in many organs. The
heritable disorders of connective tissue are varied, may
be very dissimilar clinically, and may manifest in utero or
at any age postnatally. Those that may manifest at birth
include the infantile (neonatal) form of Marfan syndrome,
congenital contractural arachnodactyly (Beals syndrome),
cutis laxa, Ehlers-Danlos syndrome, and Menkes disease.
CLINICAL SPECTRA OF DISORDERS
WITH COMMON MOLECULAR BASES
The number of clinically distinguishable skeletal dysplasias and connective tissue disorders is extensive. With
advances in molecular knowledge, several different dysplasias have been recognized to have mutations in the
same genes. In some of these disorders, clinical similarities
noted previously suggested a common etiology. One such
clinical spectrum includes achondroplasia, hypochondroplasia, severe achondroplasia with developmental delay
and acanthosis nigricans (SADDAN), and thanatophoric
dysplasia, all of which are caused by mutations in the
fibroblast growth factor receptor 3 (FGFR3) gene (Bellus
et al, 1995; Shiang et al, 1994; Vajo et al, 2000; Wilcox
et al, 1998). Another spectrum of disorders includes Stickler syndrome, Kniest dysplasia, spondyloepimetaphyseal
258
dysplasia, spondyloepiphyseal dysplasias, hypochondrogenesis, achondrogenesis type II, and recessive multiple
epiphyseal dysplasia, all of which are caused by mutations
in the gene for collagen type II, COL2A1 (Spranger et al,
1994; Winterpacht et al, 1993). With other disorders, the
common etiology is not as obvious clinically: diastrophic
dysplasia, atelosteogenesis type II, and achondrogenesis
type 1B are all caused by mutations in the diastrophic
dysplasia sulfate transporter (DTDST) gene (Bonafe and
Superti-Furga, 2007; Hastbacka et al, 1994, 1996; SupertiFurga et al, 1996). The obverse is also evident, wherein a
specific clinical entity (e.g., multiple epiphyseal dysplasias)
may be caused by a mutation in one of several genes—a
concept known as genetic heterogeneity.
APPROACH TO DIAGNOSIS
An early and precise diagnosis is important for prognosis,
optimal immediate- and long-term management, accurate
genetic counseling about recurrence risk, and identification of other possibly affected family members or disease
carriers. An example is the group of disorders with punctate calcifications (“stippling”) in epiphyses, called chondrodysplasia punctata. There are more than three types, each
of which has a different cause and mode of inheritance:
autosomal recessive, X-linked recessive, and X-linked
dominant (see Table 24-1). As in any uncommon genetic
condition, multiple factors may be required to arrive at
the correct diagnosis: a complete physical examination,
three-generation family history, radiologic studies, and
biochemical or molecular tests.
Most skeletal dysplasias cause short stature, which can be
proportionate or disproportionate. The disproportion may
be evident as a short-limbed or short-trunk form of dwarfism. If the limbs are affected, there may be segmental shortening of the upper arms and thighs (rhizomelia), forearms
and legs (mesomelia), or hands and feet (acromelia). Most
skeletal dysplasias that manifest at birth involve short limbs.
Accurate measurements of length (on a firm surface), arm
span, and head and chest circumferences must be plotted on
standard growth curves, with calculation of upper and lower
body segment ratios to objectively assess disproportion.
Other skeletal characteristics can give important clues
for specific disorders:
ll Children with achondroplasia and thanatophoric dysplasia have large heads (macrocephaly). Cloverleaf skull
deformity is present in some forms of thanatophoric
dysplasia.
ll A relatively long chest is seen in asphyxiating thoracic
dystrophy.
ll In achondroplasia the hand is short and the fingers
form a trident configuration. In diastrophic dysplasia,
there are distinctive “hitchhiker” thumbs.
259
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
TABLE 24-1 Skeletal Dysplasias Manifesting Prenatally or Perinatally
Dysplasia
Skeletal Features
Nonskeletal
Features
Radiographic
Features
Inheritance;
Gene
Comments
Lethal
Achondrogenesis
type IB
Soft cranium;
round face; short,
round chest; very
short limbs
Polyhydramnios
Poorly ossified calvarium;
ribs short with fractures
(beading); nonossified
vertebrae; small pelvis;
short broad femurs with
metaphyseal spikes, short
broad tibiae, and fibulae
AR;
DTDST
(diastrophic
dysplasia sulfate
transporter)
Same gene as diastrophic dysplasia
and atelosteogenesis II
Achondrogenesis
type II, hypochondrogenesis
Large head, flat face
with cleft palate;
short trunk;
very short limbs
(micromelia)
Fetal hydrops;
distended abdomen;
Lack of vertebral mineralization; short limbs (all segments); enlarged cranium
with normal ossification
AD; COL2A1
(type II collagen)
Same gene as for
spondyloepiphyseal
dysplasia congenita,
spondyloepimetaphyseal dysplasia,
Stickler syndrome,
Kniest syndrome
Asphyxiating thoracic dystrophy
Normal face; narrow, long chest;
variable limb
shortening
Lethal pulmonary
insufficiency
Normal calvarium and vertebrae; very short ribs with
anterior cupping; short
limbs with wide proximal femoral metaphyses;
premature ossification of
proximal femoral epiphysis
AR
Survivors may have
renal disease; possibly a variant of
short-rib polydactyly
III
Type I
Flat face with cleft
palate, micrognathia; very
narrow chest;
very short limbs
(rhizomelic) with
equinovalgus
deformities; joint
dislocations
Prematurity; stillbirth
Flat vertebrae with coronal
and sagittal clefts, scoliosis,
short ribs (11 pairs),
small pelvis with enlarged
sacrosciatic notch, short
limbs, “drumstick” humeri
and femurs, absent fibulas,
short metacarpals triangular first metacarpals,
dislocated knees
AD; filamin B
Filamin B mutations
also seen in boomerang dysplasia,
Larsen syndrome,
and spondylocarpotarsal syndrome
Type II
Cleft palate, narrow
chest, short limbs
with dislocations,
equinovarus
deformities, gap
between first and
second digits
Laryngeal stenosis;
patent foramen
ovale
Occasional coronal and sagittal vertebral clefts; short
ribs; normal sacrosciatic
notch; short “dumbbell”
humeri and femurs, small
fibulas; large second and
third metacarpals; small
round midphalanges
AR; DTDST
(diastrophic
dysplasia sulfate
transporter)
Same gene as for
diastrophic dysplasia, achondrogenesis
type IB, and some
forms of multiple
epiphyseal dysplasia
(MED)
Campomelic
dysplasia
Large cranium;
small face with
flat nose bridge,
small chin (cleft
soft palate);
small, narrow
chest; bowed
thighs and legs,
with dimple on
leg
Polyhydramnios,
congenital
cardiac abnormalities, female
external genitalia
in XY males
Large dolichocephalic
calvarium with shallow
orbits; short and wavy ribs,
often 11 pairs; hypoplastic
scapula; small, flat vertebrae; tall, narrow pelvis;
relatively long, thin limbs
with bent femurs and short
tibiae
AD (most are new
mutations);
SOX9
Chondrodysplasia punctata,
rhizomelic type 1
(RCDP1)
Face flat; very flat
nasal bridge and
tip; proximal
shortening of
limbs
Cataracts; joint
contractures;
ichthyosiform
erythroderma
Wide coronal vertebral
clefts; short humeri and
femurs; stippled epiphyses of long bones, pelvis
and periarticular areas;
trapezoid ilia
AR; PEX7 (peroxisome biogenesis
factor 7)
Atelosteogenesis
Short-rib
polydactyly
Survivors may live
a few years, with
severe growth and
mental retardation; biochemical
abnormalities are
decreased RBC
plasmalogens and
increased phytanic
acid
Heterogeneous
ciliopathies
Continued
260
PART VI Metabolic and Endocrine Disorders of the Newborn
TABLE 24-1 Skeletal Dysplasias Manifesting Prenatally or Perinatally—cont’d
Nonskeletal
Features
Radiographic
Features
Inheritance;
Gene
Hydropic appearance, round flat
face, micrognathia, extremely
narrow chest,
very short limbs,
postaxial polydactyly
Cardiac, renal, anal
malformations
Normal calvarium; very
short, horizontal ribs; flat,
wide intervertebral disc
spaces; small pelvis; short
limbs with lateral and
medial metaphyseal spurs
AR; DYNCH2H1
(dynein heavy
chain 1B for
type III only)
Types II and IV
Hydropic; short
face, flat nose,
CLP; low-set
ears; narrow
chest, protuberant abdomen;
moderately short
limbs
Cardiac, renal,
respiratory malformations
Very short, horizontal
ribs; normal pelvis and
vertebrae; short limbs with
round metaphyses; premature epiphyseal ossification; polydactyly
AR; NEK1 (never
in mitosis gene
A-related kinase
1 for type II
only)
Thanatophoric
dysplasia
Large cranium,
proptosis, flat
nasal bridge, narrow chest, very
short limbs (all
segments)
Polyhydramnios,
hydrocephalus,
brain anomalies,
congenital cardiac abnormalities
Large calvarium, short base,
small foramen magnum,
cloverleaf skull (type 2);
short, splayed, cupped ribs;
small, very flat, U-shaped
vertebrae; short, small,
flat pelvis; short, bowed
limbs; metaphyseal flare
with spike
AD (most are new
mutations);
FGFR3 (fibroblast growth
factor receptor
3)
Same gene as for
achondroplasia,
hypochondroplasia,
SADDAN
Achondroplasia
Large cranium;
frontal bossing,
flat nose bridge,
short neck;
slightly narrow
chest; proximal
limb shortening,
short trident
hands; short
proximal and
middle phalanges; joint laxity;
thoracolumbar
kyphosis
Hypotonia: delayed
motor milestones; spinal
stenosis causes
spinal compression; small
foramen magnum can cause
hydrocephalus
and apnea
Large calvarium, small foramen magnum, short base;
diminished lumbosacral
interpedicular space,
short pedicles; short ribs
with anterior cupping;
short humeri and femurs;
relatively long fibulas;
metaphyseal flare; small
iliac wings
AD (most are new
mutations);
FGFR3
Same gene as for
hypochondroplasia, SADDAN,
and thanatophoric
dysplasia
Chondrodysplasia punctata,
X-linked
recessive
Hypoplasia of the
distal phalanges;
severe hypoplasia
of nose; short
stature
Cataracts; hearing
loss; congenital ichthyosis,
anosmia, and
hypogonadism (in
contiguous gene
deletion patients)
Distal phalangeal hypoplasia; stippled epiphyses of
long bones; paravertebral
stippling
XLR; ARSE
(arylsulfatase E)
Usually milder than
X-linked dominant
form; variable
clinical severity, with
neonatal death to
longevity and diagnosis in adulthood
Chondrodysplasia
punctata,
X-linked dominant (ConradiHunermann
syndrome)
Asymmetric rhizomesomelia
Congenital cataracts; ichthyosis;
patchy alopecia
Stippled epiphyses of
long bones; paravertebral stippling; tracheal
calcifications
XLD; ESP
(3β-hydroxy
Δ8-Δ7 sterol
isomerase)
Severe form of disease;
usually lethal in
males; females vary
from stillborn to
mild (diagnosis in
adulthood); elevated
8(9)-cholestenol
Diastrophic
dysplasia
Normal cranium;
cleft palate;
micrognathia;
normal chest at
birth; very short
limbs; thumbs
proximally placed
and adducted
(hitchhiker
thumb); severe
equinovarus
of feet; limited
movement of
many joints
Cystic masses in
auricles (cauliflower ears)
during infancy;
deafness caused
by lack or fusion
of ossicles;
narrow external
auditory canal
Premature ossification of rib
cartilage; narrow L1-L5
interpedicular spaces;
scoliosis; short limbs; disproportionately short ulna
and fibula (mesomelia);
broad flared metaphyses;
ovoid first metacarpals;
variable symphalangism of
proximal interphalangeal
joints
AR; DTDST
(diastrophic
dysplasia sulfate
transporter)
Same gene as atelosteogenesis II,
achondrogenesis
type IB, and some
forms of multiple
epiphyseal dysplasia
(MED); intrafamilial
variability; normal
life span if tracheomalacia or scoliosis do not impair
respiratory function;
normal intelligence
Dysplasia
Skeletal Features
Types I and III
Comments
Nonlethal
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
261
TABLE 24-1 Skeletal Dysplasias Manifesting Prenatally or Perinatally—cont’d
Nonskeletal
Features
Radiographic
Features
Inheritance;
Gene
Large cranium; flat
face with large
eyes, flat nasal
bridge, cleft palate; short limbs
with proximal
shortening (more
severe in lower
limbs), enlarged
joints, flexion
contractures
Infancy: tracheomalacia; childhood: myopia
and retinal
detachment,
hearing loss,
delayed motor
development,
normal intelligence
Frontal and maxillary hypoplasia with shallow orbits;
slightly short ribs; flat vertebrae with coronal clefts;
small pelvis with irregular
acetabular roof; short
limbs with broad, flared
metaphyses (dumbbell),
lateral bowing of femurs
and tibiae; slightly short
and broad tubular bones of
hands and feet; epiphyses
at knees not ossified
AD; COL2A1
(type II
collagen)
Same gene as for
spondyloepiphyseal
dysplasia congenita,
spondyloepimetaphyseal dysplasia,
Stickler syndrome,
hypochondrogenesis, achondrogenesis
type II
Flat face, cleft palate, short limbs
Infancy: tracheomalacia; childhood: myopia
and retinal
detachment,
hearing loss, normal intelligence
Frontal and maxillary
hypoplasia, flat vertebrae,
small pelvis with irregular
acetabular roof, short
limbs, normal hands and
feet
AD; COL2A1
(type II
collagen)
Same gene as for
spondyloepimetaphyseal dysplasia,
Stickler syndrome,
hypochondrogenesis, achondrogenesis type II, Kniest
syndrome
Dysplasia
Skeletal Features
Kniest syndrome
Spondyloepiphyseal dysplasia
congenita
Comments
AR, Autosomal recessive; AD, autosomal dominant; RBC, red blood cell; CLP, cleft lip with or without cleft palate; XLR, X-linked recessive; XLD, X-linked dominant.
Clubfeet may occur in diastrophic dysplasia, Kniest
dysplasia, spondyloepiphyseal dysplasias, and OI
type II.
ll Postaxial polydactyly occurs in short-rib polydactyly
and asphyxiating thoracic and chondroectodermal
dysplasias. Occasionally, preaxial polydactyly can also
occur in the short-rib dysplasias.
ll Multiple joint dislocations can manifest at birth in
Larsen syndrome, Ehlers-Danlos syndrome type VII,
atelosteogenesis, and Desbuquois syndrome.
The presence of extraskeletal abnormalities may provide
additional clues to diagnosis, as follows:
ll Cleft palate may occur in campomelic, Kniest, spondyloepiphyseal, short-rib polydactyly (Majewski), atelosteogenesis types I and II, hypochondrogenesis, and
diastrophic dysplasias.
ll Congenital cataracts are frequent in some forms of
chondrodysplasia punctata.
ll Congenital cardiac defects occur in short-rib polydactyly dysplasias and Ellis van Creveld syndrome.
If the infant or fetus dies, specimens of cartilage and
skin fibroblasts should be obtained for histochemical tests,
biochemical assays, and molecular analysis; these can be
used to make or confirm diagnoses and permit accurate
future prenatal diagnosis. Even if the molecular or enzymatic basis of the condition is not understood at the time,
the tissue may be useful in the future. If photographs and
skeletal radiographs were not obtained premortem, they
should be obtained postmortem.
CLINICAL TESTING
Presentation
Radiographs of the entire skeleton, including the skull,
hands, feet, and lateral spine, are essential for accurate
diagnosis. Atlases dedicated to skeletal dysplasias are
essential for this purpose (Lachman, 2006; Spranger et al,
2002), even to the experienced radiologist or neonatologist. Ultrasound images of the brain, heart, and kidneys
may be helpful if anomalies in those organs are suspected.
Detailed family history and measurements of family members may be helpful; more mildly affected members might
have gone without a diagnosis. Molecular investigations
may be necessary to arrive at the proper diagnosis; given
their complexity, such analyses should be considered after
consultation with a clinical geneticist.
OI type II (perinatal lethal type) is estimated to affect
1 in 20,000 to 60,000 infants. Affected infants may be
born prematurely, with low birthweight and disproportionately short stature. The limbs are short and bowed
with extra, circular skin creases; the hips are abducted
and flexed. The head is soft and boggy, and minimal calvarial bone can be felt. The sclerae are dark blue and the
chest is narrow. The infant cries with handling because
there are many fractures at different stages of healing.
Sixty percent of affected babies are stillborn or die during the first day of life, and 80% die by 1 month. With
the growing use of ultrasonography, affected fetuses
may be detected in the early second trimester because of
ll
DISORDERS OF BONE FRAGILITY
OSTEOGENESIS IMPERFECTA TYPES II AND III
OI is characterized by increased bone fragility. There are
classically four major clinical types: types II and III are the
most severe, manifesting prenatally and perinatally (Byers,
2002; Steiner et al, 2005). However, fractures at birth
can occur in OI type I. Further heterogeneity in OI has
recently been described.
262
PART VI Metabolic and Endocrine Disorders of the Newborn
A
B
FIGURE 24-1 (See also Color Plate 5.) Osteogenesis imperfecta type II. A, A 20-week fetus. The limbs are angulated and deformed from multiple
fractures. B, Radiograph of fetus (20 weeks’ gestation) showing an absence of ossification in the calvarium, short telescoped or crumpled humeri and
femurs, and short and wavy ribs with fractures.
short and bowed or angulated limbs and narrow thoraces
(Figure 24-1).
OI type III (progressive deforming type) can manifest prenatally, perinatally, and in the first 2 years of life.
Prenatal and perinatal clinical features resemble those in
OI type II, but are less severe (Figure 24-2), and perinatal death is not uncommon. If not present at birth,
fractures and deformations of the limbs develop in the
first and second years. The highest prevalence of fractures in OI, up to 200, occur in type III. Extremely short
stature, with adult heights of 92 to 108 cm, can result
from microfractures in growth plates. The head may be
large because the calvarium is soft with a large anterior
fontanel. The sclerae may be blue initially, but are white
by puberty. The head assumes a triangular shape, with a
bossed, broad forehead and a tapered, pointed chin. Later
in childhood, dentinogenesis imperfecta and hearing loss
may develop. Severe kyphoscoliosis may occur, leading to
cardiopulmonary compromise, which is the major cause
of early death.
Radiolographic Features
Radiographs show the femurs in OI type II to be short,
broad, and “telescoped” or “crumpled.” The tibiae are
short and bowed or angulated, and the fibulae may be
thin (see Figure 24-1, B). There is minimal to no calvarial
mineralization. The acetabulae and iliac wings may be
somewhat flattened. The ribs are short, wavy, and thin or
broad, with “beading” from callus formation at fetal fracture sites.
In OI type III, the femurs are short and deformed, but
not crumpled as in OI type II (see Figure 24-2, B and C).
The other long bones are thinner than usual, with healing fractures incurred in utero, bowing, and deformations.
The calvarium is undermineralized with a large anterior
fontanel, and there are many Wormian bones (small
islands of bone in the suture spaces; see Figure 24-2, D).
The ribs are thin and gracile.
Etiology
OI is most commonly caused by mutations in one of the
two genes for type I collagen (COL1A1 and COL1A2),
the predominant protein building block of bone. More
clinically severe forms of OI are the result of qualitatively
abnormal collagen synthesis, rather than decreased production (Byers, 2002).
Inheritance
A fetus or infant with OI type II or III is usually the result
of a spontaneous dominant-acting gene mutation, but
there is a small risk of recurrence (approximately 6%) in
subsequent siblings because of parental somatic or gonadal
mosaicism. The parent is usually asymptomatic but may
have minimal manifestations, such as short stature. Most
cases of OI are inherited as autosomal dominant traits,
although rare recessive forms have been shown to be caused
by mutations in the cartilage-associated protein CRTAP
(Barnes et al, 2006), prolyl 3-hydroxylase (LEPRE1), and
cyclophylin B (van Dijk et al, 2009).
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
A
263
B
C
D
FIGURE 24-2 Osteogenesis imperfecta type III. A, Neonate with normal face, short neck, slightly short limbs. B, Radiograph shows that the
calvarium is undermineralized with Wormian bones. C, Radiograph shows the upper limbs, which have bowed humeri and callus in the ulnae.
D, Radiograph shows lower limbs with moderately short, thick femora, and angulated tibiae and fibulae. (Courtesy Paige Kaplan, Children’s Hospital of
Philadelphia, Philadelphia, Penn.)
Differential Diagnosis
Other lethal skeletal dysplasias may have similar abnormalities to those in OI type II and may be difficult to
distinguish by prenatal ultrasonography; however, in experienced hands they can be differentiated based on several
ultrasound findings. Krakow et al (2008) did a retrospective analysis of 1500 prenatally diagnosed cases of skeletal
dysplasias. The three most common prenatal-onset skeletal dysplasias were osteogenesis imperfecta type 2, thanatophoric dysplasia, and achondrogenesis 2, accounting for
almost 40% of cases. Postnatal radiographs clearly reveal
distinctive differences among thanatophoric dysplasia,
campomelic dysplasia, achondrogenesis, and perinatal
hypophosphatasia, among others.
Management
If the diagnosis of OI is made prenatally, cesarean delivery
has not been shown to decrease fracture rate or improve
survival rate of severely affected fetuses (Cubert et al,
2001). Those severely affected with OI II are not expected
to survive the neonatal period. In OI type III, the neonate
needs careful handling to minimize pain and prevent further fractures. Analgesia alleviates pain. Consideration can
264
PART VI Metabolic and Endocrine Disorders of the Newborn
be given to treatment with bisphosphonates (using intravenous pamidronate), which increase bone density, reduce
the frequency of fractures and pain, possibly prevent short
stature and deformations, and permit ambulation (Aström
et al, 2007). It is prudent to treat only severely affected
children in whom the clinical benefits outweigh potential
long-term effects.
Handling an Infant With Osteogenesis
Imperfecta
When changing the diapers of an infant with OI, place a
hand behind the buttocks with the forearm supporting the
legs. Similarly, when the infant is lifted the buttocks, head,
and neck must be supported. The infant can be laid on a
pillow to be carried. To transport the infant, an infant seat
that reclines as much as possible and allows easy placement
or removal should be used. The seat can be padded with
egg crating or 1-inch foam. A layer of foam can be placed
between the seat’s harnesses and the child for extra protection. The car seat must always be placed in the back seat.
Sling carriers and “umbrella” strollers should not be used
for infants with OI because they do not give sufficient leg,
head, and neck support.
PERINATAL HYPOPHOSPHATASIA
Presentation
Perinatal hypophosphatasia is a lethal condition characterized by short, deformed limbs, a soft skull, blue sclerae,
and undermineralization of the entire skeleton, so that
many bones cannot be visualized and may seem absent
on radiography. In the skull, only the base can be visualized radiologically. There may be rachitic changes and
fractures. Seizures that are responsive to pyridoxine may
occur. There is polyhydramnios during pregnancy, and
death can occur in utero. The disorder affects approximately 1 in 100,000 live births; neonatal death is common
(Whyte, 2000).
Radiolographic Features
The radiologic features of perinatal hypophosphatasia
include polyhydramnios (prenatal); underossification,
especially of the calvarium and long bones (with marked
variability); small thoracic cavity; short, bowed limbs;
spurs in the middle portion of the forearms and lower legs;
and dense vertebral bodies.
Etiology
Mutations in the ALPL gene are responsible for deficiency
of the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP), thus causing perinatal hypophosphatasia. The serum alkaline phosphatase (ALP) value is low.
Serum values of inorganic pyrophosphate and pyridoxal
5′-phosphate (putative natural substrates for TNSALP)
may be elevated, and urinary phosphoethanolamine is elevated (Mornet and Nunes, 2007).
TNSALP acts on multiple substrates: the essential function of TNSALP is in osteoblastic bone matrix
mineralization. TNSALP hydrolyzes inorganic pyrophosphate to phosphate, thought to be critical in promoting
osteoblastic mineralization. If TNSALP is deficient, there
is extracellular accumulation of inorganic pyrophosphate,
which inhibits hydroxyapatite crystal formation and mineralization of the skeleton. TNSALP is also needed for
delivery of pyridoxal-5-phosphate into cells where it is a
cofactor (vitamin B6).
Inheritance
Perinatal hypophosphatasia is inherited as an autosomal
recessive trait, with a 25% recurrence risk in future pregnancies. Prenatal diagnosis is optimized through the use
of ultrasonography, assay of TNSALP activity in amniocytes, DNA mutation analysis if the previously affected
infant’s mutation was known, or a combination of these
methods.
Differential Diagnosis
Differential diagnoses are osteogenesis imperfecta type II
and achondrogenesis.
Management
Treatment is primarily supportive and directed toward
minimizing pain and discomfort. Clinical trials with bonetargeted human recombinant enzyme replacement therapy
are underway.
FGFR3 SPECTRUM
ACHONDROPLASIA
Presentation
Achondroplasia is the most common of the nonlethal
chondrodysplasias; it affects 1 in 25,000 live births. It
is characterized by short stature with short limbs, particularly rhizomelic (proximal) and acromelic (hands)
shortening with trident hand configuration, large head
with frontal prominence (“bossing”), flat nasal bridge
and midface, long narrow trunk, joint laxity, and development of thoracolumbar kyphosis (“gibbus”) in infancy
(Figure 24-3).
The foramen magnum and cervical spinal canal may
be narrow and can cause compression of the spinal cord.
Standards have been published for foramen magnum size
in achondroplasia (Hecht et al, 1985). Compression of
the lower brainstem and cervical spinal cord can lead to
hypotonia, central apnea, retardation, quadriparesis, and
(rarely) sudden death (Pauli et al, 1984). Perinatal or infantile death can occur, but is unusual. Infants often sleep with
their neck hyperextended, and symptoms can be exaggerated by neck flexion (Danielpour et al, 2007).
Radiolographic Features
The calvarium is large with a relatively small foramen
magnum and a short base. The lateral cerebral ventricles may be large, but hydrocephalus is not a common
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
A
C
265
B
FIGURE 24-3 Achondroplasia. A, Infant with achondroplasia has macrocephaly and
proximal limb shortening (rhizomelia). B, Infant with achondroplasia exhibits frontal bossing
and flat nasal bridge. C, Neonatal film of achondroplasia illustrates a large skull, a somewhat
narrow chest, short vertebral bodies with a lack of lumbar interpediculate flare, and rhizomelia. (B, Courtesy Charles I. Scott, AI DuPont Institute, Wilmington, Del.)
complication. The proximal long bones (humeri and
femurs) are short, including the femoral neck. Fibulae
are longer than tibiae. There is metaphyseal flaring. The
hand is short with a trident configuration of the fingers,
with short proximal and middle phalanges. Vertebrae are
small and cuboid with short pedicles, and there may be
anterior beaking of the first or second lumbar vertebrae;
there is lack of flare of the interpedicular distance in the
lumbar vertebrae. The pelvis has squared iliac wings
(“elephant ear” appearance), a narrow greater sciatic
notch, and flat acetabular roofs. Compression of the cervical cord, if present, can be ascertained with magnetic
resonance imaging cerebrospinal fluid flow studies in
flexion and extension.
266
PART VI Metabolic and Endocrine Disorders of the Newborn
Etiology
The cause of achondroplasia is a mutation of the gene for fibroblast growth factor receptor 3 (FGFR3). The FGFR3 protein is a membrane-spanning tyrosine kinase receptor, which
may form dimers with other gene family members FGFR1,
FGFR2, and FGFR4. The heterodimers serve as receptors for
several fibroblast growth factors (Cohen, 2006). More than
97% of persons with achondroplasia have a mutation in the
transmembrane domain of the FGFR3 gene, in which glycine
is substituted by arginine (Gly380Arg; Shiang et al, 1994).
The same gene is mutated at different sites in hypochondroplasia, thanatophoric dysplasia, SADDAN, Muenke craniosynostosis, and Crouzon craniosynostosis syndrome with
acanthosis nigricans (Vajo et al, 2000). Histopathologic examination demonstrates a defect in the organization and maturation of the cartilage growth plates of long bones because
of differing degrees of constitutive activation of the receptor.
and given a copy of the guidelines for health supervision
of children with achondroplasia issued by the American
Academy of Pediatrics (Trotter et al, 2005).
THANATOPHORIC DYSPLASIA
Presentation
Thanatophoric dysplasia is one of the most common lethal
dysplasias (Unger et al, 2007), occurring in 1 in 45,000
births. It is characterized by extremely short limbs, long
narrow trunk, large head with bulging forehead, prominent eyes, flat nasal bridge, wide fontanel, and occasionally
cloverleaf skull deformity (Figure 24-4). It is differentiated
into types I and II on the basis of radiologic features. Death
occurs in the neonatal period from respiratory insufficiency. Polyhydramnios is common during pregnancy.
Inheritance
Radiographic Features
The inheritance pattern in achondroplasia is autosomal dominant. Approximately 80% of cases are sporadic occurrences
in a family, representing new mutations. Cases may be associated with advanced paternal age, with molecular confirmation
that the new mutations are of paternal origin. Affected individuals are fertile, and achondroplasia is transmitted as a fully
penetrant autosomal dominant trait, meaning that each person who inherits the mutant gene will manifest the condition.
Femurs are short, flared at the metaphyses with a medial
spike, and are bowed (type I) or straight (type II); other
long bones are also short and bowed (see Figure 24-4).
The calvarium is large with a short base and small foramen magnum; cloverleaf skull is sometimes present in
type I and is severe in type II. Vertebrae are strikingly flat
(platyspondyly) with a U- or H-shape in anteroposterior
projection and uniform interpediculate narrowing. Ribs
are short, cupped, and splayed anteriorly (Lachman, 2006).
Differential Diagnosis
Differential diagnoses are SADDAN (Vajo et al, 2000)
hypochondroplasia (Bellus et al, 1995).
Management
The infant with achondroplasia is often hypotonic; together
with the large head, the hypotonia leads to delayed motor
milestones. Development of thoracolumbar kyphosis
may be exacerbated by unsupported sitting before truncal muscle strength is adequate; therefore infants should
not be carried in flexed positions (including soft slingcarriers and umbrella strollers). Rear-facing car safety seats
should always be used. Most infants lose their kyphosis and
develop lumbar lordosis when they begin walking.
Hydrocephalus may occasionally develop during the first
2 years, so the head circumference and body length should
be carefully measured and plotted on standard achondroplasia growth charts (Trotter et al, 2005). Routine imaging of
the skull and brain is not recommended; however, development of hyperreflexia, hypotonia, or apnea may herald the
development of clinically significant cord compression. Surgical decompression at the foramen magnum or the upper
cervical spine may prevent neurologic damage, although
most patients usually gain motor milestones late but spontaneously, because the foramen grows faster than the cord.
The upper airway in individuals with achondroplasia
is small, often leading to obstructive apnea, snoring, and
chronic serous otitis media beyond infancy. Treatment
may consist of tonsillectomy, adenoidectomy, and placement of myringotomy tubes. Parents should be counseled
about the clinical and hereditary aspects of the disorder
Etiology
Thanatophoric dysplasia represents the severe end of
the FGFR3 spectrum. In thanatophoric dysplasia type I,
the most common mutation in the extracellular domain
is a substitution of arginine at position 248 by cysteine
(Arg248Cys), but other mutations have been described
throughout the gene. In all studied cases of thanatophoric
dysplasia type II, there is a substitution of lysine at position
650 by glutamate (Lys650Glu; Wilcox et al, 1998).
Inheritance
All cases of thanatophoric dysplasia, as with most cases of
achondroplasia and hypochondroplasia, occur sporadically
and result from new autosomal dominant mutations. Nevertheless, there may be a small risk of recurrence to siblings
of a sporadic case, possibly caused by gonadal mosaicism.
Differential Diagnosis
Differential diagnoses are OI types II and III, achondroplasia (severe), achondrogenesis, and hypochondrogenesis.
Management
If the condition is diagnosed prenatally, the couple should
receive genetic counseling and anticipate neonatal death.
If the diagnosis is suggested after delivery and radiographically confirmed, management is solely supportive,
with death from pulmonary insufficiency usually occurring
within hours to days.
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
267
B
A
C
FIGURE 24-4 (See also Color Plate 6.) Thanatophoric dysplasia. A, Neonate with thanatophoric dysplasia has a large head, narrow chest, short
limbs, extra creases on the limbs, short hands with trident fingers, and angulated abducted thighs. B, Neonate with thanatophoric dysplasia has a face
with a bossed forehead, flat nose bridge, short neck, very short limbs with extra creases, and trident fingers. C, Radiograph of an infant with thanatophoric dysplasia demonstrates a large calvarium, short ribs with anterior splaying, flat vertebral bodies (platyspondyly), and short bowed femurs with
medial metaphyseal spike. (B, Courtesy Montreal Children’s Hospital, Montreal, Quebec, Canada.)
COL2A1 SPECTRUM
SPONDYLOEPIPHYSEAL DYSPLASIA
CONGENITA
from the spinal (spondylo-) and growth plate (epiphyseal)
involvement. Congenita indicates that the condition is present from birth.
Presentation
Radiolographic Features
Spondyloepiphyseal dysplasia congenita (SEDC) manifests with shortened neck, trunk and limbs, normal-sized
hands and feet, flat facial profile, and occasional cleft palate and clubfoot (Unger et al, 2007). The name is derived
Radiolographic features include ovoid or pear-shaped vertebral bodies in infancy, with platyspondyly more evident
at a later age; odontoid hypoplasia evident in early childhood; midface hypoplasia; retrognathia; mild rhizomelia
268
PART VI Metabolic and Endocrine Disorders of the Newborn
A
D
C
B
FIGURE 24-5 (See also Color Plate 7.) Spondyloepiphyseal dysplasia congenita. A, A 2-month-old infant demonstrating short neck, trunk, and
limbs. Note the flat facial profile and normal size of hands and feet. B, Anteroposterior radiograph reveals platyspondyly and short chest. C, Lateral
radiograph reveals platyspondyly. D, Upper limb radiograph reveals rhizomelia, mesomelia, and a normal-sized hand.
and mesomelia (Figure 24-5); absent ossification of the
os pubis; apparent decreased bone age caused by epiphyseal involvement; and development of coxa vara, variable
kyphosis, and scoliosis in childhood
Etiology
SEDC is caused by mutations in the gene for type II collagen (COL2A1), the predominant protein building block
of cartilage. Mutations in COL2A1 are also responsible for
Kniest dysplasia, some forms of spondyloepimetaphyseal
dysplasia and Stickler syndrome, and the perinatal lethal
disorders achondrogenesis type II and hypochondrogenesis (Spranger et al, 1994; Tiller et al, 1995; Winterpacht
et al, 1993).
Inheritance
SEDC is inherited in an autosomal dominant pattern.
Offspring of affected individuals are at 50% risk for inheriting the disorder. Recurrence risk for unaffected parents is approximately 6%, because of parental gonadal
mosaicism.
Differential Diagnosis
Differential diagnoses are the milder form of hypochondrogenesis and Morquio syndrome.
Management
Neonates may require intubation because of upper airway compromise. Care must be given when manipulating
the cervical spine (as in endotracheal intubation), because
of odontoid hypoplasia. C1-C2 fusion may be required
in early childhood to stabilize the cervical spine. Annual
hearing screens are recommended during childhood.
Regular ophthalmologic evaluation (semiannually before
school age) is essential to detect early development of retinal detachment and to manage myopia. Osteoarthritis is
a common feature in early adulthood, often requiring hip
arthroplasty.
ACHONDROGENESIS
II–HYPOCHONDROGENESIS
Presentation
The severe end of the COL2A1 spectrum manifests with
fetal hydrops and maternal polyhydramnios, severe short
trunk and limbs, and fetal or neonatal death caused by pulmonary hypoplasia.
Radiolographic Features
Radiolographic features include prenatal polyhydramnios,
a large calvarium with normal ossification, midface hypoplasia, retrognathia, platyspondyly with underossification
of the vertebral bodies (achondrogenesis II), short chest
with a protuberant abdomen, marked shortening of all
tubular bones (Figure 24-6), and small iliac wings.
Etiology
Achondrogenesis II–hypochondrogenesis is caused by
mutations in the gene for type II collagen (COL2A1), the
predominant protein building block of cartilage (Mortier
et al, 2000).
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
A
FIGURE 24-6 Achondrogenesis type II. A, This
20-week fetus demonstrates small chest and short
limbs. B, This radiograph demonstrates poor ossification of vertebral bodies and short limbs.
B
Inheritance
All cases of achondrogenesis II–hypochondrogenesis
are caused by spontaneous dominant-acting mutations
in COL2A1. Recurrence risk has been reported as high
as 6%, because of parental gonadal mosaicism (Forzano
et al, 2007).
Differential Diagnosis
Differential diagnoses are achondrogenesis type I and
osteogenesis imperfecta types II and III.
Management
If the condition is diagnosed prenatally, the couple should
receive genetic counseling and anticipate neonatal death. If
the diagnosis is suggested after delivery and radiographic
confirmation is obtained, management is solely supportive,
with death from pulmonary insufficiency usually occurring
within hours to days.
DTDST SPECTRUM
DIASTROPHIC DYSPLASIA
Presentation
Newborns with diastrophic dysplasia exhibit limb
shortening, cystic ear swelling, hitchhiker thumbs
(developing several days after birth), spinal deformities
(especially cervical kyphosis), and contractures of the
large joints (Figure 24-7). Clubfoot and ulnar deviation
269
of the fingers may also be present. On occasion the
disease can be lethal at birth, but most individuals survive the neonatal period (Bonafe and Superti-Furga,
2007).
Radiolographic Features
The most characteristic clinical and radiologic feature
is the proximally-placed hitchhiker thumb, with ulnar
deviation of the fingers. Cervical kyphosis is a frequent
finding. Long bones are moderately shortened and thick,
with mild metaphyseal flaring, rounding of the distal
femur, and bowing of the radius and tibia. Severe talipes
equinovarus may be present. Iliac wings are hypoplastic,
with flat acetabular roofs. The chest can be narrow, bell
shaped, or both. Narrowing (lack of flare) of the interpedicular distance in the lumbar spine is reminiscent of
achondroplasia.
Etiology
Diastrophic dysplasia is caused by mutations in the diastrophic dysplasia sulfate transporter gene (DTDST),
which also cause the lethal disorders achondrogenesis type 1B and atelosteogenesis type 2, as well as a
rare recessive form of multiple epiphyseal dysplasia.
The gene product is a sulfate-chloride exchanger of
the cell membrane (Superti-Furga et al, 1996); this
affects incorporation of sulfate into proteoglycans
(mucopolysaccharides), especially chondroitin sulfate
B–containing proteoglycans, which are prevalent in
cartilage.
270
PART VI Metabolic and Endocrine Disorders of the Newborn
A
C
B
FIGURE 24-7 Diastrophic dysplasia. A, This infant has prominent eyes, small chin, slightly narrow chest, proximally placed angulated thumbs,
and short limbs. B, Neonate profile showing small chin, swollen ears, and short neck. Note the proximally placed, angulated thumb. C, View of the
neonate’s hand shows the proximally placed angulated thumb and mild syndactyly. (Courtesy Paige Kaplan, Children’s Hospital of Philadelphia, Philadelphia, Penn.)
Inheritance
Radiolographic Features
Diastrophic dysplasia is inherited in an autosomal recessive pattern. Siblings of affected individuals are at 25% risk
for inheriting an abnormal allele from both carrier parents.
The long bones are extremely short, with square, globular,
or triangular shapes and medial spikes in the metaphyses of
the femurs (Figure 24-8). The calvarium and vertebrae are
poorly ossified (type IB), and the ribs are short.
Differential Diagnosis
Differential diagnoses are atelosteogenesis type II (part of
the DTDST spectrum), spondyloepiphyseal dysplasia, and
arthrogryposis.
Etiology
Management
Inheritance
Mechanical ventilation may be required because of small
chest circumference and a floppy airway. Maintenance of
joint mobility and proper positioning through physical
therapy is essential. Serial casting and or surgical correction
of clubfeet may be required. Cervical kyphosis can impede
endotracheal intubation and can result in cord compression, but may resolve spontaneously during infancy.
Achondrogenesis 1B is inherited in an autosomal recessive pattern. Siblings of affected individuals are at 25%
risk for inheriting an abnormal allele from both carrier
parents.
ACHONDROGENESIS 1B
Presentation
Achondrogenesis type IB is characterized by short stature,
extremely short limbs, a relatively large head with a round
face, short nose, small mouth, soft skull, and very short
neck (Borochowitz et al, 1988). Polyhydramnios during
pregnancy, premature delivery, and hydrops are common.
The affected infant is stillborn or dies within hours of
birth.
Achondrogenesis type 1B is caused by mutations in the
DTDST gene (see Diastrophic dysplasia, earlier).
Differential Diagnosis
Differential diagnoses are atelosteogenesis type II (part
of DTDST spectrum), achondrogenesis type II, and
hypochondrogenesis.
Management
If the condition is diagnosed prenatally, the couple should
receive genetic counseling and anticipate neonatal death. If
the diagnosis is suggested after delivery and radiographic
confirmation is obtained, management is solely supportive,
with death from pulmonary insufficiency usually occurring
within hours to days.
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
A
B
OTHER SKELETAL DYSPLASIAS
CAMPOMELIC DYSPLASIA
271
FIGURE 24-8 A and B, Achondrogenesis type IB. Cervical, thoracic, and lumbar vertebral bodies are not ossified,
the sacrum is not ossified, the ribs are short, and the limbs
are extremely short with medial femoral metaphyseal spikes.
(Courtesy Elaine Zackai, Children’s Hospital of Philadelphia,
Philadelphia, Penn.)
chest with thin, wavy ribs (with only 11 pairs); and scapular hypoplasia.
Presentation
Etiology
Campomelic dysplasia is characterized by short stature
(birth length of 35 to 49 cm), large dolichocephalic skull,
large anterior fontanel, high forehead, flat face, widely
spaced eyes with short palpebral fissures, low-set ears, cleft
soft palate, micrognathia, relatively long and slender thighs
and upper arms, short bowed legs with dimples in the midshaft (in most cases), narrow chest, and kyphoscoliosis
(Figure 24-9). Sex reversal or ambiguous genitalia affects
75% of the chromosomal males; there may be internal
and external genital abnormalities (from mild anomalies
to complete sex reversal) in XY males (Meyer et al, 1997;
see Figure 24-9, A and B). Absence of the olfactory bulbs
and tracts as well as heart and renal malformations may
occur. Death, usually in infancy, results from pulmonary
hypoplasia, tracheomalacia, or cervical spinal instability.
Survivors are usually globally developmentally delayed.
A few more mildly affected people without bowed limbs
have been reported (Unger et al, 2008).
Campomelic dysplasia is caused by mutations in or near
the SRY-related HMG-Box gene 9 (SOX9; Tommerup,
1993). SOX9, with homology to the SRY gene, is a transcription factor involved in both bone formation and testis
development.
Radiolographic Features
The most characteristic finding is midshaft angulation (campomelia) of the femurs, although it is not a
constant finding. Other features include hypoplastic,
undermineralized cervical vertebrae and thoracic pedicles; narrow iliac wings with dislocated hips; brachydactyly; clubfeet; anterior bowing of tibia; bell-shaped
Inheritance
Campomelic dysplasia is an autosomal dominant trait.
Most cases are new sporadic occurrences in a family; recurrence caused by gonadal mosaicism has been reported
(Smyk et al, 2007).
Differential Diagnosis
Differential diagnoses are osteogenesis imperfecta types II
and III, diastrophic dysplasia, kyphomelic dysplasia, thanatophoric dysplasia, and spondyloepiphyseal dysplasia congenita (severe).
Management
Survival beyond the newborn period is rare; therefore support is primarily directed toward comfort measures. In survivors, care must be given to the cervical spine, which may
be unstable. Chromosomal studies to determine gender
and pelvic ultrasonography to examine internal genitalia
272
PART VI Metabolic and Endocrine Disorders of the Newborn
A
B
C
FIGURE 24-9 Campomelic dysplasia. A, 46,XY female 22-week-old fetus with normal head, long philtrum, micrognathia, low-set ears, mild
narrowing of chest, proximally placed thumbs, and bowed or angulated lower limbs resembling those of osteogenesis imperfecta type II but less shortened. The external genitalia are female. B, Neonate with the long-limb form of the disorder has a relatively large head, micrognathia, narrow chest,
and bowing of lower limbs with characteristic dimpling of lower leg. C, Radiograph shows the narrow chest, the relatively long, thin limb bones with
bowing of the femurs and tibiae, and a long, narrow pelvis. (Courtesy Paige Kaplan, Children’s Hospital of Philadelphia, Philadelphia, Penn.)
may be performed. Cleft palate may be repaired in those
able to feed orally, and clubfeet may require casting or surgical correction.
CONNECTIVE TISSUE DISORDERS
CONGENITAL (NEONATAL, INFANTILE)
MARFAN SYNDROME
Presentation
Infants with neonatal congenital Marfan syndrome (cMS)
have a long, thin body and can have an aged appearance
because of a lack of subcutaneous tissue and wrinkled,
sagging skin (Morse et al, 1990; Figure 24-10). The craniofacial features include dolichocephaly, deep-set eyes
with large or small corneas (and occasionally cataracts),
high nasal bridge, high palate, small pointed chin with a
horizontal skin crease, and large simple or crumpled ears.
The fingers and toes are long and thin (arachnodactyly).
Some joints are hyperextensible, and others have flexion
contractures causing clubfoot, dislocated hips, or adducted
thumbs. Infants tend to exhibit hypotonia with low muscle mass. Lenses are usually not subluxated at birth. The
most important cause of morbidity and mortality is severe
cardiovascular disease, which affects almost every neonate
with cMS—namely, mitral and tricuspid valve prolapse
and insufficiency and aortic root dilatation. The ascending
aorta may be dilated and tortuous. Many infants die in the
first year of life from congestive heart failure. Survivors
have chronic hypotonia and contractures, are unable to
walk, and require many surgical procedures.
Radiolographic Features
Radiolographic features include pectus deformity, spontaneous pneumothorax, dural ectasia, aortic root dilatation,
and mitral valve prolapse. Many of these features may not
be present in the newborn period.
Etiology
Congenital MS is caused by mutations in the gene encoding fibrillin 1 (FBN1; Dietz et al, 1991). Fibrillin is a glycoprotein associated with microfibrils, which form linear
bundles in the matrices of many tissues, such as aorta, periosteum, perichondrium, cartilage, tendons, muscle, pleura,
and meninges. There are two regions in FBN1 in which
many mutations causing cMS occur; these lie among exons
24 to 27 and exons 31 and 32 (Dietz, 2009). Molecular
analysis does not yield mutations in all cases.
Inheritance
Marfan syndrome is an autosomal dominant disorder.
Most neonates with cMS are sporadic occurrences within
a family (Dietz et al, 1991; Morse et al, 1990). However,
there is one well-documented neonate with cMS whose
father had classic Marfan syndrome except for average
height (Lopes et al, 1995).
Differential Diagnosis
Differential diagnoses are congenital contractural arachnodactyly (CCA), autosomal recessive cutis laxa, and
Loeys-Dietz syndrome.
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
273
B
A
C
FIGURE 24-10 Congenital Marfan syndrome. A, Neonate with long, thin trunk and limbs (particularly the feet), lack of adipose tissue, and multiple skin creases giving an aged appearance. The ears are large and simple, and the chin is small with a horizontal crease. There are flexion contractures at the joints. B, The neonate’s face shows laxity of skin, typical horizontal chin crease, and a pointed chin. The fingers are long with adduction
contractures of the thumbs, which extend past the edge of the palm, and floppy wrists. C, Lateral view of the neonate’s head showing simple, large
ears and redundant skin on the neck. (Courtesy Paige Kaplan, Children’s Hospital of Philadelphia, Philadelphia, Penn.)
Management
Patients require annual ophthalmologic and cardiac evaluation throughout childhood. Cardioselective beta-blockers,
such as atenolol, are often implemented at the first signs of
aortic root dilatation. The angiotensin II antagonist losartan has also shown promise in this regard (Brooke et al,
2008). Children should be screened for the development
of scoliosis.
CONGENITAL CONTRACTURAL
ARACHNODACTYLY (BEALS SYNDROME)
Presentation
CCA (Beals syndrome) is characterized by a thin, wasted
appearance with minimal muscle and fat mass (similar
to neonatal Marfan syndrome). Distinctive features
include arachnodactyly with contractures of the large
and small joints (Figure 24-11, A), as well as crumpled,
overfolded helices of the external ear. Cardiovascular
involvement is usually limited to mitral valve prolapse,
but aortic root dilatation may occasionally develop
(Godfrey, 2007).
Radiolographic Features
Features are nonspecific and include elongated proximal
phalanges; contractures of digits, ankles, knees, and hips;
thin, gracile tubular bones; and gradual development of
kyphoscoliosis.
Etiology
CCA is caused by mutations in the fibrillin 2 gene.
274
PART VI Metabolic and Endocrine Disorders of the Newborn
Inheritance
CCA is inherited in an autosomal dominant manner, with
many patients representing the result of spontaneous
mutations. Offspring of affected individuals are at 50%
risk for inheriting the condition. Gonadal mosaicism has
been described in CCA (Putnam et al, 1997).
Differential Diagnosis
caused by mutations in the gene for type III collagen,
COL3A1. The arthrochalasia type is caused by mutations
in either gene for type I collagen (COL1A1 or COL1A2),
which result in loss of the N-proteinase cleavage site of the
protein.
Inheritance
Proper nutrition is essential to ensure adequate weight
gain. Joint contractures respond to physical therapy, but
occasionally surgical release may be required. Surveillance
for development of spinal curvature and aortic root dilatation, although rare, are essential throughout childhood.
Most types of EDS are inherited as autosomal dominant
traits. Each child of an affected person has a 50% chance
of inheriting and manifesting the disorder, although there
can be marked intrafamilial variability (Malfait and de
Paepe, 2005). One form of the arthrochalasia type, also
referred to as dermatosparaxis, is inherited in an autosomal
recessive pattern, and is caused by deficiency of procollagen N-peptidase. The kyphoscoliotic form (formerly type
VI) is also inherited in an autosomal recessive pattern and
is caused by a deficiency of lysyl hydroxylase, an enzyme
that aids in crosslinking of collagen fibrils.
EHLERS-DANLOS SYNDROMES
Differential Diagnosis
Presentation
Differential diagnoses are cMS, congenital contractural
arachnodactyly, and Larsen syndrome.
Differential diagnoses are cMS, cutis laxa, and distal
arthrogryposis.
Management
The Ehlers-Danlos syndromes (EDSs) are a clinically and
genetically heterogeneous group of connective tissue disorders, which are characterized by varying degrees of joint
and skin hypermobility, excessive bruising, abnormal wound
healing, and fragility of tissues (Steinmann et al, 2002;
Wenstrup and de Paepe, 2008). This group of disorders was
reclassified in 1997 (Beighton et al, 1998). The classic type
(formerly type I) and the arthrochalasia type (formerly type
VII) are the most likely to manifest in the newborn period.
Type I is often characterized by premature delivery of an
affected fetus as a result of a rupture of the fragile amniotic
membranes. The infant may be floppy and in the breech
position. There may be joint laxity and joint instability. In
type VII, the major involvement is in the ligaments and
joint capsules. Large and small joints are hypermobile and
dislocatable; severe congenital dislocation of hips occurs.
In vascular (formerly type IV) EDS, the greatest danger
is to the pregnant affected woman, for whom there is a
high risk of uterine and arterial rupture. Although there
is a 50% risk that the fetus will be affected, the problems
of blood loss and prematurity are more important in the
newborn period than the disorder itself.
Radiolographic Features
Radiolographic features are dependent on the particular
type of EDS. Congenital hip dislocation may be evident
on plain films. Hydronephrosis, bladder diverticula, and
spontaneous pneumothorax may occur occasionally. Aortic dilatation and arterial aneurysms may be evident by
echocardiography and other imaging modalities, but only
occur in patients of school age or older.
Etiology
Mutations in two of the genes for type V collagen (COL5A1
and COL5A2) are demonstrable in some cases of classic
EDS (Malfait and de Paepe, 2005). The vascular type is
Management
Trauma should be avoided because of skin fragility. Effective closure of surgical wounds is challenging because of
a tendency for dehiscence (Wenstrup and Hoechstetter,
2004).
CUTIS LAXA
Presentation
Cutis laxa is a genetically heterogeneous disorder, meaning
that mutations in several different genes may be responsible for the phenotype. As such, the presentation can be
highly varied. Infantile forms may exhibit loose, furrowed
skin, a large anterior fontanelle, hypotonia, hernias, and
congenital hip dislocation (see Figure 24-11, B) (Kaler,
2005; van Maldergem et al, 2011).
Radiolographic Features
Radiolographic features are in part dependent on the
genetic form of the disorder. Nonspecific features include
a large anterior fontanel, congenital hip dislocation, and
hernias. The X-linked form may exhibit occipital horns.
Arterial tortuosity, aortic root dilatation, and cortical and
cerebellar anomalies may be seen in some forms, as well as
gastrointestinal and urinary tract diverticula.
Etiology
The relatively mild, autosomal dominant form of cutis
laxa is caused by mutations in the elastin gene, ELN.
The X-linked recessive form (occipital horn syndrome)
is caused by mutations in the ATP7A gene (allelic with
Menkes syndrome). Autosomal recessive forms may be
caused by mutations in the fibulin 4 (FBLN4) and fibulin
CHAPTER 24 Skeletal Dysplasias and Connective Tissue Disorders
A
275
B
FIGURE 24-11 A, Congenital contractural arachnodactyly (Beals syndrome). This infant has a long, thin trunk and limbs, contractures of joints,
and crumpled ears. B, Infant with cutis laxa. (Courtesy Montreal Children’s Hospital, Montreal, Quebec, Canada.)
5 (FBLN5) genes or the A2 subunit of the V-ATPase gene
(ATP6V0A2). Biochemical clues as to the etiology in a
particular patient may include decreased serum copper
and ceruloplasmin (X-linked form), and abnormal serum
sialotransferrin isoelectric focusing in cases caused by
ATP6V0A2 mutations.
Inheritance
Because cutis laxa is genetically heterogeneous, modes of
inheritance include autosomal dominant, autosomal recessive, and X-linked recessive. The latter two modes are usually
responsible for forms with neonatal and infantile presentation.
Differential Diagnosis
Differential diagnoses are EDS, Menkes syndrome, gerodermia osteodysplastica, and de Barsy syndrome.
Management
Serious childhood complications include developmental
delay, pulmonary emphysema, aortic root dilatation, and
arterial tortuosity. Annual ophthalmologic and cardiac
examinations are essential, and referral to special education programs may be indicated.
MENKES SYNDROME
Presentation
Menkes syndrome often appears in the newborn period with
nonspecific neurologic manifestations. Typically, developmental delay is evident in the first 2 to 3 months of life, with
failure to thrive, seizures, and severe ocular manifestations.
Changes in the appearance of the hair include hypopigmentation, brittleness, patchy alopecia, and twisted shafts
seen by light microscopy (i.e., pili torti; Figure 24-12).
Early death is common and may occur in infancy (Kaler,
2010). Serum copper and ceruloplasmin concentrations
are low, and the plasma dopamine-to-norepinephrine ratio
may be elevated (Goldstein et al, 2009).
Radiolographic Features
Features may evolve during infancy and may include bladder diverticula (seen on bladder ultrasound and voiding
cystourethrogram [VCUG]), tortuous vessels (on echocardiogram, magnetic resonance imaging with contrast),
gastric polyps (on upper GI), metaphyseal spurring, osteopenia, and Wormian bones on plain radiographs (Lachman, 2006; see Figure 24-12).
Etiology
Menkes syndrome is caused by mutations in a coppertransporting adenosine triphosphatase gene, ATP7A (Kaler
et al, 1994). This enzyme takes part in the final processing of a number of copper-dependent enzymes, including dopamine beta-hydroxylase, tyrosinase, lysyl oxidase,
superoxide dismutase, and cytochrome c oxidase. As a
result, several physiologic processes and cellular functions
are impaired, including collagen cross-linking, pigment
production, and neurotransmission (Goldstein et al, 2009).
Inheritance
Menkes syndrome is an X-linked recessive disorder; therefore only males are affected. Female carriers may exhibit
pili torti in some hair shafts because of lyonization (Moore
276
PART VI Metabolic and Endocrine Disorders of the Newborn
FIGURE 24-12 (See also Color Plate 8.)
Menkes syndrome. A, Note blonde hair,
fair complexion, and epicanthal folds in this
11-month-old Hispanic boy. B, Note multiple Wormian bones near the occiput.
A
and Howell, 1985). Sons born to carrier females have a
50% risk for manifesting the disease.
Differential Diagnosis
Differential diagnoses are cutis laxa (the occipital horn
form is allelic), EDS, neonatal cMS, biotinidase deficiency,
mitochondrial myopathies, nutritional copper deficiency,
and organic aciduria.
Management
Early diagnosis allows for parenteral copper supplementation therapy (Kaler et al, 2008), but this is not effective in all patients. Patients should be monitored for the
development of seizures, as well as a propensity for bone
fragility, poor wound healing, and vascular fragility leading to excessive bleeding, hemorrhagic strokes, and
B
subdural hematomas. Bladder diverticula may result in
urinary retention and urinary tract infections and should
be surgically corrected. Patients are at risk for moderate to
severe developmental delay, and they should be referred to
infant stimulation and early intervention programs.
SUGGESTED READINGS
Aström E, Jorulf H, Söderhäll S: Intravenous pamidronate treatment of infants with
severe osteogenesis imperfecta, Arch Dis Child 92:332-338, 2007.
Brooke BS, Habashi JP, Judge DP, et al: Angiotensin II blockade and aortic-root
dilation in Marfan’s syndrome, N Engl J Med 358:2787-2795, 2008.
Kaler SG, Holmes CS, Goldstein DS, et al: Neonatal diagnosis and treatment of
Menkes disease, N Engl J Med 358:605-614, 2008.
Trotter TL, Hall JG: The Committee on Genetics: Health supervision for children
with achondroplasia, Pediatrics 116:771, 2005.
Wenstrup RJ, Hoechstetter LB: Ehlers-Danlos syndromes. In Cassidy SB,
Allanson JE, editors: Management of Genetic Syndromes, ed 2, New York, 2004,
Wiley-Liss, pp 211-224.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
P A R T
V
II
Care of the Healthy Newborn
C H A P T E R
25
Initial Evaluation: History and Physical
Examination of the Newborn
Jeffrey B. Smith
The central focus of this chapter is the medical evaluation
of the apparently well newborn in the first few days after
birth in a hospital setting. Obtaining the history, performing the physical examination, and judging the significance
of risk factors and findings all require skill and experience.
For brevity, we mostly refer to the person evaluating the
newborn as a pediatrician. In some institutions, the routine
evaluation is performed by a general pediatrician, family
practitioner, or pediatric nurse practitioner, and the neonatologist enters the well baby nursery only as a consultant. In other institutions, the neonatologist has direct
responsibility for patients in the normal nursery in addition to the neonatal intensive care unit (NICU). In either
setting, the neonatologist should develop and maintain
proficiency in the evaluation of the normal newborn, a task
that requires an approach and set of skills somewhat different than the evaluation of the NICU patient, who typically has a known symptom or high-risk condition. In the
normal nursery, the vast majority of newborns are healthy
and will not require medical intervention; therefore a primary goal is to identify the small minority with problems
that have the potential to cause serious morbidity if not
detected in a timely fashion. This goal includes identifying psychosocial and medical problems. For the healthy
majority, the evaluation provides the basis for appropriate
parental reassurance and education. For this reason, the
emphasis of the discussion in this chapter is on common
problems, variations of normal, and subtle abnormalities.
Some symptoms and findings that suggest significant illness are also described, but the discussion in this chapter
is not intended to provide a complete differential diagnosis or guide to management. Physical findings pertinent
to specific diseases and organ systems are described with
greater detail in other sections throughout the book. Evaluation of the dysmorphic infant is described in Chapter
19. Laboratory studies helpful in the routine care of the
healthy newborn are discussed in Chapter 26.
HISTORY
The medical history of the newborn begins with pertinent information about the mother’s past medical and
pregnancy history, the current pregnancy, and the family. Information about the current pregnancy, labor, and
delivery is central at the time of delivery, but the infant’s
postnatal history becomes progressively more important in
the subsequent hours and days. An outline of basic components of the prenatal and newborn history is presented in
Table 25-1. The history is potentially gathered from multiple sources, including records of prenatal outpatient visits
and laboratory studies; the records of the mother’s current
and prior hospitalizations; the delivery record; newborn
records created by nurses and other personnel; direct communications from the obstetrician, midwife, and nurses;
and interviews with the mother and other family members.
Most of the prenatal history is collected and recorded by
members of the obstetric team. Systems should be in place
to ensure that the pediatrician responsible for the newborn
is directly informed about high-risk conditions in a timely
manner, but the pediatrician has an independent responsibility to review the information available in the maternal
record. From a potentially large amount of information,
a key task for the pediatrician is to efficiently identify and
highlight the portions that are relevant to the newborn’s
current situation and the task at hand.
To ensure that important information is not overlooked,
a systematic approach to the collection and recording of
the history is essential. Structured data systems, preferably
electronic, can help to ensure that essential information is
not missed. However, information gathering must be prioritized appropriately, because the relative importance of
specific parts of the history depends on the clinical situation. For example, the physician paged to attend the emergency delivery of a fetus in distress should focus information
gathering in the few minutes available on what is directly
relevant to preparations for resuscitation. At that moment,
it is not necessary to know the results of prenatal testing
of the mother for hepatitis B. For the stable infant sent to
room-in with the mother and for the critically ill infant
admitted to the NICU, it becomes important to obtain
this information in the next few hours, so that hepatitis
B vaccine can be administered within the recommended
12 hours after birth if the mother does not have a documented negative test result for hepatitis B infection.
For the healthy newborn, history gathering at the time
of the initial encounter after birth will emphasize the prenatal history (including maternal and family history), the
delivery and neonatal transition, the initiation of feeding,
and any symptoms or parental concerns that have manifested since birth. A major goal of the initial evaluation is
277
278
PART VII Care of the Healthy Newborn
TABLE 25-1 Components of the Prenatal and Newborn History
Category
Typical Components of the History
Maternal identification
Name, medical record number, age, gravidity, parity, estimated gestational age
Maternal medical history
Allergies
Significant past illnesses, hospitalizations, and surgeries
Chronic illnesses, especially diabetes
Chronic medications
Psychiatric history
Previous pregnancies
Dates, routes of delivery, complications, and outcomes
Breastfeeding history
Family history
General health status and history of family members
Congenital anomalies, metabolic disorders, hearing impairment
Food and other allergies of parents and siblings
Psychosocial history
Family structure, care of previous children
Availability of support for parents (extended family, others)
Living accommodations, access to transportation
Language, racial or ethnic group, parental education level
Smoking, alcohol, drugs of abuse
Family strife, domestic violence
Current pregnancy
Estimated gestational age (and how determined)
Singleton or multiple fetuses
Fertilization history (e.g., assisted reproduction)
Prenatal care (when started, number of visits, provider)
Maternal blood type and screening for unusual isoimmune antibodies
Screening for group B streptococcal colonization
Other prenatal screening for infectious diseases*
Glucose tolerance test results
Results of maternal drug screening, if applicable
Occupational or other exposure to teratogens
Sexual contact with high-risk group
Genetic screening tests
Ultrasound studies and amniocentesis
Complications, hospitalizations
Prescription and nonprescription medications; herbal remedies
Breastfeeding plans; plans for well-child care after discharge
Labor
Spontaneous, induced, augmented
Duration of first and second stages
Rupture of membranes: spontaneous versus artificial, duration
Amniotic fluid: clear, meconium stained, bloody, foul smelling
Signs of infection: fever, increased white blood cell count, uterine tenderness, tachycardia
Antibiotics during labor and the indication
Other medications during labor
Fetal monitoring abnormalities
Delivery and delivery room stabilization
Route and presentation
Indication for cesarean section, vacuum, or forceps, if applicable
Stabilization and resuscitation measures; time to spontaneous breathing
Apgar scores
Placental abnormalities, if noted
Cord blood gases, if obtained
Ease versus difficulty of transition
Medications administered (erythromycin eye drops, vitamin K)
Abnormalities noted on initial examination
Postnatal history
Nursing or parental concerns
Growth parameters (weight, length, head circumference, and percentiles)
Vital signs, daily weights
Glucose checks
Activity and alertness
Breastfeeding (frequency, duration, quality of latch)
Bottle feeding (amount, frequency, reason)
Voiding, wet diapers
Meconium and transitional stools
Procedures (lumbar puncture, circumcision)
Results of laboratory tests and imaging studies
*Prenatal screening performed in accordance with medical recommendations and local regulations commonly include tests for group B Streptococcus, hepatitis B, syphilis, gonorrhea,
and for immunity to rubella; it may also include tests for chlamydia, tuberculosis, HIV, and hepatitis C.
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
to identify risk factors for problems that may develop in
the next few days, such as early-onset neonatal sepsis or
exacerbated hyperbilirubinemia. Identification of psychosocial risk factors is also an important goal, and it remains
important throughout the hospitalization. At the time of
the predischarge evaluation, the key goal is to determine
whether the infant can be discharged home safely. The
prenatal and perinatal history will have already been documented and reviewed, so information-gathering for the
predischarge evaluation will focus on the interval history,
mostly collected from the nursing records and the parents.
Because of the frequency and potential morbidity of
early onset neonatal sepsis, the presence or absence of risk
factors for sepsis should be assessed as part of the initial
evaluation of every newborn. Historical risk factors for
early onset sepsis include prolonged rupture of the fetal
membranes (18 hours or more), a maternal body temperature of 38° C or higher, uterine tenderness, foul-smelling
or purulent amniotic fluid, an elevated maternal white
blood cell count or left shift, and fetal or maternal tachycardia. Maternal colonization with group B streptococci is
also considered a risk factor unless adequate intrapartum
prophylaxis was administered or the fetal membranes were
intact until delivery by cesarean section (American Academy of Pediatrics, 1997). Poor tolerance of labor, manifested by an unexplained need for resuscitation or a slow
transition, can be a nonspecific symptom of sepsis.
Whereas gathering the history and performing the physical examination are distinct activities, described by convention in separate parts of a note, they are not performed
in isolation. Knowledge of specific concerns, events, and
risk factors in the history should prompt a more focused or
detailed examination of the relevant body region or organ
system than otherwise might be done. Conversely, specific
questioning prompted by physical examination findings
will often elicit information that was not volunteered in
the earlier routine history gathering, such as a family history of a specific anomaly. When a note is written, the history, the physical examination, and laboratory studies are
the data upon which the current assessment and plan of
care are based. Once documented, the assessment and plan
also become part of the patient’s ongoing history.
PHYSICAL EXAMINATION
Physical examination requires skills whose refinement
can continue to reward and challenge the pediatrician
and neonatologist throughout a professional lifetime. No
matter how experienced the practitioner, every examination represents an opportunity to add to or refine one’s
knowledge of significant abnormalities and of the wide
range of variation of common, benign conditions. Nevertheless, the examiner knows that most newborns are in fact
healthy; therefore one of the practical challenges of performing the routine newborn examination is to maintain a
high level of vigilance and thoroughness throughout every
examination.
The examination of the healthy newborn entails a complete physical examination in the sense that all parts of the
body are examined. However, no actual examination or
its write-up can be complete in the sense of exhaustively
exploring and explicitly documenting all possible findings.
279
The degree of detail that is appropriate is a matter of judgment that will vary with the presenting situation and the
findings discovered during the examination. Distinct subtypes of the newborn examination include examinations in
the delivery room, at admission, and at discharge from the
normal nursery, in addition to examinations initiated in
response to specific concerns.
EVALUATION AT BIRTH
At the time of birth, attention focuses on the initiation of
air breathing and cardiorespiratory stability. The assessment of successful adaptation or a need for resuscitation
is described in Chapter 28. The infant whose condition
remains unstable or who has major anomalies that are
apparent on initial inspection will be transferred to an
NICU for further evaluation and management.
DELIVERY ROOM DISPOSITION
The infant whose condition stabilizes after delivery, with
or without intervention, is evaluated to determine whether
the infant can remain with the mother. This examination
centers on assessment of the adequacy of the cardiorespiratory transition, and it also includes a basic inspection for
congenital anomalies such as imperforate anus or other
problems that may indicate a need for admission to an
NICU or observation nursery. Passage of a thin catheter
through both nostrils and into the stomach can be done
in the delivery room to rule out choanal and esophageal
atresia. For routine deliveries, the delivery room discharge
examination is often done by the obstetrician or nursemidwife. If the pediatrician is present, the examination at
this time can be expanded to serve as the nursery admission exam.
NURSERY ADMISSION EXAMINATION
For babies not requiring admission to an NICU or observation unit, this examination is done after the infant
completes transition, and usually by 24 hours after birth.
The purpose of a complete physical examination is to
efficiently detect problems that, if present, are inapparent or may soon develop. This examination is the main
focus of this chapter. The initial physical examination of
the patient admitted to the NICU is similar, except that
examination in the NICU should be initiated immediately after admission, and parts of the NICU examination
may need to be modified or delayed because of physiologic instability or limited accessibility. Whether in the
nursery or the NICU, the identification of historical risk
factors or the detection of symptoms or abnormalities
requires that the basic examination be expanded to focus
additional attention on the areas relevant to the differential diagnosis.
TARGETED OR PROBLEM-DIRECTED
EXAMINATION
When the physician is called to evaluate an infant because
of specific symptoms or concerns, the examination will
naturally focus on aspects relevant to those issues. Because
280
PART VII Care of the Healthy Newborn
the nonspecific nature of many symptoms in the infant
often implies a large differential diagnosis, even the targeted examination will often need a wide focus. The daily
follow-up examination can be regarded as a variety of
targeted examination, guided by the infant’s overall condition. For healthy infants remaining in the hospital for
several days after birth, areas of active attention always
include the infant’s neurologic and cardiorespiratory stability, hydration status, feeding and elimination behavior,
and jaundice.
NURSERY DISCHARGE EXAMINATION
The discharge examination is similar in scope to the wellbaby admission examination, but with a slightly altered
emphasis, based on the additional information provided by
the period of observation in the hospital, and by the goal
of determining whether the infant is ready for routine care
at home. The discharge examination is still a complete
physical examination that encompasses the entire body.
However, it is not necessary for the discharge examination to duplicate all portions of the admission examination, provided that it was performed by the same person
or specific aspects of the admission examination were
sufficiently well documented by a trusted colleague. For
example, it is unnecessary to repeat a search for physical
anomalies that do not change with time, such as examination of the oral cavity for cleft palate, or to check again
for red retinal reflexes if they were already found to be
normal. Nevertheless, it is often more efficient to repeat
the entire examination than to verify the completeness of
an earlier examination by another. When the hospital stay
is short, a combined admission-discharge examination is
appropriate.
EVALUATION OF GESTATIONAL AGE
If the obstetric estimate of the gestational age is uncertain
or appears unreliable, the gestational age of the newborn
infant can be estimated based on physical examination criteria. No individual feature is a reliable guide to the gestational age, but scoring systems that use multiple features of
physical and neuromuscular maturity have been evaluated
extensively (Amiel-Tison, 1968; Dubowitz et al, 1970).
The New Ballard Score is probably the most widely used
in contemporary practice (Ballard et al, 1991). Detailed
descriptions and a video demonstration of this examination are available at www.ballardscore.com.
ENVIRONMENT OF EXAMINATION
The environment in which the examination is performed
can significantly affect the reliability of the examination
via effects on the examiner or on the baby. The examiner
must be aware of the limitations produced by a suboptimal environment and adjust the approach to the examination to compensate, arrange for the infant to be moved, or
defer selected parts of the examination when appropriate.
Important environmental considerations include lighting,
room temperature, and the levels of background noise and
other distractions. An often overlooked source of distraction is a situation that forces the examiner into a physically
awkward or uncomfortable position, such as tall examiner
bending over a low bed or examining table. If possible, the
infant’s clinical state and tolerance for handling can also
have a major effect on the conduct of the examination. The
wise examiner will postpone the examination of a hungry,
crying infant if possible, and try to return when the infant
is fed and calm.
The healthy infant is typically examined in a warmer
bed in the delivery room or in a bassinet in the mother’s
room or in the nursery. An open warmer bed provides the
best access to the patient, allowing the infant to be kept
warm while completely undressed during the examination. The presence of a warmer bed in the delivery room
adds to the advantages of performing the nursery admission examination soon after delivery, but often this is not
practical. Most examinations of the healthy newborn are
done with the infant in a bassinet, starting with the infant
dressed in at least a shirt and hat, and wrapped in blankets.
An adequate well-baby examination can be performed
under these conditions, but the sequence of the examination should be modified to minimize the time the infant
is fully undressed. The examiner must take extra care to
ensure that the examination is complete and the entire skin
surface is visualized at some point during the examination.
Performing the examination in the presence of the parents allows the examiner to show how the infant responds
to handling, and to demonstrate immediately any findings that require explanation or reassurance. Watching
the examination may stimulate the parents to ask questions that might otherwise not occur to them until later,
and it provides the physician an immediate opportunity
for further education. On the other hand, interruptions
from parents and other family members can interfere with
the examiner’s train of thought, risking inadvertent distraction and omissions. In large, busy nurseries, it may be
more practical for the pediatrician to examine a series of
infants in the nursery and then report the results of the
examination to each set of parents afterward. Specific findings should be demonstrated to the parents at that time, if
appropriate. Regardless of whether the parents are present
for the examination, the results of the examination should
be communicated promptly. Parents may be anxious about
findings that the pediatrician believes are of little consequence, and vice versa. Prompt and sensitive communication of the examination findings helps to build parents’
trust, whereas delayed or poor communication can undermine the parents’ relationship with the physician and the
hospital.
CLINICAL APPROACH TO THE NEWBORN
EXAMINATION
An important challenge of the newborn examination is the
need to maintain a high level of vigilance and thoroughness, while projecting an attitude of comfortable reassurance appropriate to the reality that the overwhelming
majority of newborns seen in the well-baby nursery are in
fact healthy. Although the eyes and fingers of the experienced examiner will detect many abnormalities by pattern recognition alone, even the most expert examiner can
miss important findings unless a disciplined, systematic
approach is used. However, the desire to ensure that no
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
essential aspect of the examination is missed or slighted
needs to be balanced with the infant’s limited tolerance
for handling. It may seem easier to avoid omissions if the
examiner always performs the examination in the same
sequence, but a rigid approach that fails to adjust to the
state and activity of the individual infant may yield a suboptimal examination that is inefficient and unnecessarily
stressful for infant, parent, and examiner.
The physical examination of the newborn includes measurements of the weight, length, and head circumference,
which must be compared with standardized growth data
(Figure 25-1) to determine whether the infant is small
(<10th percentile), appropriate, or large (>90th percentile) for gestational age (Battaglia and Lubchenco, 1967).
The body temperature, heart rate, and respiratory rate
are typically recorded at regular intervals by nurses, but
the pediatrician should also consciously evaluate the heart
and respiratory rate at the time of examination. The blood
pressure is not routinely measured in healthy newborns,
but the blood pressure should be checked in all four
extremities if the history or examination suggest a problem
with the circulation. Of the standard physical examination
techniques, the most important in the examination of the
newborn is observation. Palpation and auscultation are
also important, whereas percussion is of relatively limited
use. The routine examination also includes specific physical maneuvers for examining the hips and for eliciting a
variety of reflex responses. A stethoscope, an ophthalmoscope, and a tape measure are the only pieces of equipment
generally needed. A source of light for transillumination is
helpful for specific purposes. Pulse oximetry can enhance
early detection of critical congenital heart disease, but further study is needed to determine whether this procedure
should become a standard of care in the routine assessment
of the neonate (Mahle et al, 2009).
Sensitivity to the infant’s state and responses can greatly
facilitate the examination and make it less stressful for the
infant and less time-consuming for the examiner. The
newborn infant generally responds more slowly to a stimulus than does an older child or adult, and the response
5.5
50
4
97
90
55
50
10
3
50
45
10
3.5
3
3
97
90
50
10
3
40
2.5
Head circ (cm)
Birthweight (kg)
4.5
Length (cm)
90
5
tends not to remain localized to the area of the stimulus.
For example, a gentle touch to the face or extremity of
a sleeping infant, or merely loosening a blanket, typically results in some limited initial movement of the area
touched, followed with a slight delay by a wave of movement that spreads to involve the whole body, and which
may be accompanied by partial arousal. If the examiner
waits a few seconds for this wave of response to fully subside before proceeding to move or touch the infant again,
the infant will often settle and remain asleep. With a series
of such gentle, well-spaced interactions, the examiner can
often complete important portions of the examination
without waking the infant. If, however, the examiner provides a new stimulus soon after the first, before the secondary movements have finished spreading and while the
infant is partially aroused, the new stimulus is likely to
reinforce the initial stimulation and provoke full awakening and a crying.
The examination should begin with a deliberate pause
to observe the infant in an undisturbed state. Before the
infant is moved or undressed, the examiner should actively
observe the infant and mentally note as much information as possible, including the state of alertness or sleep,
color of visible portions of the skin and mucous membranes, respiratory rate and any audible sounds or signs of
increased work of breathing, posture, spontaneous activity, and quality of cry. Throughout the examination, the
examiner should alternate between focusing attention on
specific elements of the examination (e.g., listen for a heart
murmur, closely scrutinize a scalp lesion) and maintaining a global awareness of the infant’s overall responses and
activity. The experienced examiner’s general impression
of whether the patient is sick or healthy is an important
part of the evaluation.
If the infant is sleeping or in a quiet alert state, the examiner will usually begin by gently uncovering the chest.
Depending on the type of clothing the infant is wearing, it
may be best at first to lift the shirt or gown just enough to
slide the stethoscope underneath. Because the heart sounds
and a soft murmur, if present, can be obscured easily by
60
97
2
1.5
0
32
34
36
38
Weeks
40
42
44
281
35
30
25
32
34
36
38
Weeks
40
42
44
FIGURE 25-1 Fetal-infant growth
chart for 32 to 44 weeks’ gestation.
( Redrawn from Fenton TR: A new
growth chart for preterm babies: Babson
and Benda’s chart updated with recent
data and a new format. BMC Pediatr
3:13, 2003. The complete chart for 22
to 50 weeks’ gestation can be downloaded
from http://members.shaw.ca/
growthchart.)
282
PART VII Care of the Healthy Newborn
crying, the precordial area is usually auscultated first. If the
infant remains quiet, the clothing can be removed further,
and the stethoscope can be moved to other locations on
the chest to expand the area of auscultation for breath and
heart sounds, and then to the abdomen to listen for bowel
sounds. If the infant is already crying or awakens and starts
crying vigorously, auscultation can be deferred in favor of
other parts of the examination. Auscultation of the chest
can be performed later, after the infant has become quiet.
If the infant is lightly dressed and is being examined on
an open warming bed, all clothing can be removed after
the initial auscultation of the chest and abdomen; this
allows the entire body to be easily observed throughout the
examination with a systematic head-to-toe approach. If the
infant is in an open crib, he or she may become cold and
upset if undressed fully at the beginning of the examination. In this environment, it is preferable to allow the infant
to remain at least partially clothed for as long as possible.
For example, start by examining the head, and after replacing the infant’s hat, open the shirt to finish examining the
anterior chest and abdomen, closing but not buttoning
or snapping the shirt while moving to examine the hips,
genitalia, and lower extremities, and finally removing the
shirt to examine the upper extremities and back. However,
proceeding in this way requires extra attention on the part
of the examiner to ensure that no parts of the examination
are missed.
An experienced examiner can accomplish a thorough
examination of the newborn more quickly than a novice,
largely because of the acquired skill in handling the infant
and minimizing wasted or redundant motions. Although
there is no substitute for experience, the trainee’s skill will
develop more rapidly if close attention is paid to developing a fluid approach to the examination as a whole. The
following paragraphs provide one example; each experienced examiner will develop his or her own preferred
sequence. Further details of specific parts of the examination are described in subsequent sections.
After the initial moment of observation and auscultation
of the anterior chest and abdomen, begin examining the
head. Using both hands, gently encircle and palpate the
entire scalp while inspecting parts of the scalp that are visible in this position (the back of the head and neck will be
inspected later). Gently turn the head to one side (noting
any limitation of range of motion), and inspect that side of
the head, including the ear, and then the side of the neck.
Roll the infant’s head gently while working the fingers
around the neck to the other side, palpating the neck and
clavicular areas and applying gentle traction on the skin as
needed to open the neck creases to allow full visualization.
After reaching the other side of the neck, inspect the ear
and side of the head. Next, proceed to the face, attending
first to overall features of shape and symmetry and then
focusing sequentially on the skin, eyes, nose, mouth, and
oral cavity.
With the shirt open in front, inspect and palpate the
anterior chest. Observe the respiratory pattern, noting the
presence or absence of retractions. Additional auscultation
of the chest may be done at this time, if not completed
earlier. Open the diaper, and inspect and palpate the abdomen and umbilicus. Palpate the femoral pulses. One may
examine the genitalia and perineum at this time, or wait
until after the examination of the hips and lower extremities, described next.
Inspect the lower extremities. Check the range of
motion of the hips and perform the Barlow and Ortolani
maneuvers. Palpate the legs, sliding the examiner’s hands
from the hips down to the ankles. The examiner should
place the thumbs on the soles of the feet, with the fingers
around the back of the ankles. From this position, the
examiner can smoothly check the alignment of the feet,
elicit the plantar grasp, and then slide the thumbs up along
the outer edge of the soles to elicit Babinski’s reflex. Next,
lift and abduct the legs into a frog-leg position to provide a
full view of the perineum and anus. Inspect the genitalia by
gently retracting the labia majora in females, or depressing the skin at the base of the penis in males. Inspect and
palpate the scrotum and testes. After this, the diaper can
be refastened.
Attention next turns to the upper extremities. The shirt
should be removed completely to allow unobstructed observation of the overall shape, symmetry, and movements of
the arms and hands. With the infant supine, turn the head to
elicit the asymmetric tonic neck reflex on each side. Palpate
the whole arm gently, starting with one of the examiner’s
hands on each of the baby’s shoulder’s, and then slide down
to the baby’s hands, noting any swelling or discontinuities.
Inspect the hands, fingers, nails, and palms. If the infant’s
hand is tightly fisted, do not attempt to pry the fingers open.
Instead, gently flex the wrist to 90 degrees, which will cause
the fingers to relax naturally. Inspect the palms, and then
elicit the palmar grasp reflex. Without releasing the baby’s
hands, one can then perform the pull-to-sit maneuver. The
examiner places his or her hand behind the infant’s head
and neck to provide support as the infant is gently lowered
back toward the bed. When the infant’s head and shoulders
are a few inches from the bed, the examiner drops his or her
hand rapidly to elicit the Moro reflex.
Next, place the hands on either side of the chest, under
the arms at the shoulders, and raise the baby to an upright
position, noting the strength and tone of the shoulder
muscles. Lower the infant, still in an upright position, and
try to elicit the supporting and stepping reflexes. Turn the
infant to a prone position, suspended on the examiner’s
hand. Observe the infant’s posture and tone, and elicit the
incurvation response. Inspect the infant’s back, from the
vertex of the head down to the sacrum (pulling the diaper
down, if needed). Gently place the infant back in the crib,
and dress the infant. The red reflex examination can be
performed at this time, or at any suitable moment when
the eyes are open spontaneously.
The preceding description assumes routine findings
throughout. The examiner should always be prepared to
deviate from his or her preferred sequence in response to
changes in the infant’s state or if abnormalities are found
that require a more extended examination of particular
features. However, it remains important for the examiner
to complete the entire examination and not become distracted by a prominent finding. On occasion, portions of
an examination need to be deferred; this is more common
in the NICU, because of patient instability, than in the
well-baby nursery. In either environment, any limitations
of the examination should be clearly documented, with the
need for reexamination listed explicitly in the plan of care.
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
SKIN
The entire skin surface should be inspected during the course
of the examination, with attention to the color, moisture,
temperature, texture, and elasticity of the skin, the pattern
and depth of skin creases, and the presence and character of
any local alterations or lesions. The skin appendages—nails
and hair—are inspected along with the skin, with attention
to the color, size, shape, and any lesions of the nails, and to
the growth pattern, color, texture, and distribution of scalp
and body hair. Lighting must be adequate and consistent.
Natural light is best, although not always available. Phototherapy lights must be turned off during the examination.
Typically the skin in different regions of the body is examined sequentially during the course of the routine examination, but if abnormalities are seen, it can be helpful to
reexamine the infant in a fully undressed state and in a warm
environment, so that all parts of the body can be directly
compared. The size, shape, location, distribution, and time
course of lesions are all important for differential diagnosis.
Skin color is a composite of the infant’s basic skin pigmentation, the adequacy of perfusion, the amounts of oxygenated and deoxygenated hemoglobin in the local circulation,
and, at times, internal staining of skin by extravasated blood
or pigmented molecules such as bilirubin. The skin can also
be stained externally by in utero exposure to meconium
or postnatally by foreign substances applied to the skin.
The newborn is typically much more pink or red than the
mother, because of the healthy infant’s high hemoglobin
level. With excessive hemoglobin levels, as in polycythemia
or twin-twin transfusion, a deeply red or purple-red color
(plethora) is evident. Pallor may be caused by anemia or
poor perfusion. Central oxygenation is usually best evaluated by observing the color of the tongue and oral mucous
membranes, because the color of lips can be misleading.
Visual detection of central cyanosis requires approximately
5 g of desaturated Hb per 100 mL of blood, and may not
be apparent in the presence of significant anemia (Snider,
1990). Cyanosis of the perioral area, hands, and feet (acrocyanosis), which is common in the first 24 hours after birth
or if the infant is cold, is due to relatively poor perfusion
of those areas, resulting in increased O2 extraction by the
tissues and an increase in the concentration of deoxyhemoglobin. Because skin color is dependent on perfusion, it can
be a sensitive indicator of systemic perfusion. However, the
infant’s peripheral color can vary markedly and rapidly with
activity and the local environmental temperature. Transient mottling of skin of the extremities and trunk (cutis
marmorata) is common in newborn infants in response to
cold. If mottling does not resolve with warming, other conditions should be considered, including hypertension and
hypothyroidism. Harlequin color change is a striking but
infrequently observed transient asymmetry of color and
perfusion, in which one side of the body is vasodilated, with
a clear line of demarcation at the midline.
The yellow-orange color of jaundice caused by unconjugated bilirubin becomes obvious if serum bilirubin levels
are sufficiently high, but it can be difficult to detect at more
moderate levels. Typically, jaundice first becomes apparent
in the face and then progresses distally as bilirubin levels
continue to rise. The detection of mild-to-moderate jaundice is easier if the infant’s foot is lifted so that the face and
283
foot can be seen side by side in the same visual field, with
the foot providing a built-in control for the infant’s basic
skin pigmentation. This is particularly helpful in the black
or brown infant, in whom mild jaundice is more difficult to
detect than in the white infant, and in the Asian infant whose
skin tones can exaggerate the appearance of mild jaundice.
A transient period of generalized erythema may be noted
a few hours after birth, usually by the parents, often during
a bath or with vigorous crying, resolving within minutes to
an hour. Sometimes called erythema neonatorum, it appears
to be associated with successful completion of neonatal
transition of the circulation, and it rarely recurs with the
same intensity after the first episode (Fletcher, 1998).
Skin creasing is affected by both development and movement. Creasing increases if movement is constrained, as in
the fetus near term. An absence of creasing on the palms
or soles of a term or near-term neonate can be caused by
a prolonged lack of movement secondary to a neuromuscular disorder. Edema stretches the skin and obscures normal skin folds and creases. Edematous skin pits with gentle
pressure. The distribution of edema is affected by gravity
and thus changes with position. In contrast, lymphedema
(as in Turner syndrome) tends to accentuate the creases,
and is less affected by gravity (Fletcher, 1998).
Injuries caused by iatrogenic trauma in utero or at delivery are common; they include scars from injury by an
amniocentesis needle, minor lacerations and abscesses from
fetal scalp electrodes or fetal blood gas sampling, scalpel
lacerations during cesarean section, and ecchymoses and
abrasions from forceps or vacuum. Trauma can also occur
from pressure on the fetus during labor, particularly over
bony prominences. Such pressure may be involved in the
development of subcutaneous fat necrosis, an uncommon
condition hypothesized to be caused by hypoxic injury
to fat and manifested by firm, subcutaneous nodules and
plaques (Cohen, 2008). The lesions usually resolve spontaneously after several months, but can become inflamed
and fluctuant and are occasionally associated with the late
development of significant hypercalcemia. Aplasia cutis
congenita is a focal lesion characterized by congenital
absence of some or all layers of skin (Figure 25-2) and can
FIGURE 25-2 Aplasia cutis congenita. Lesions are usually single,
but in this case there are two adjacent lesions near the vertex—one
bullous and one membranous. (Reprinted from Rudolph AJ: Atlas of the
newborn, vol 4, Hamilton, Ont, Canada, 1997, BC Decker, p 30.)
284
PART VII Care of the Healthy Newborn
BOX 25-1 C
ommon and Uncommon Causes
of Skin Lesions in Neonates
PUSTULAR, VESICULOPUSTULAR, AND VESICULOBULLOUS
LESIONS
ll Common or benign: erythema toxicum neonatorum, transient neonatal
pustular melanosis, miliaria crystallina, miliaria rubra, sucking blisters,
neonatal acne (benign cephalic pustulosis)
ll Infectious: herpes simplex, varicella, staphylococcal pustulosis, bullous
impetigo, congenital candidiasis, syphilis, scabies
ll Chronic or recurrent: epidermolysis bullosa, mastocytosis, epidermolytic
hyperkeratosis, acropustulosis of infancy
ll Positive Nikolsky’s sign: epidermolysis bullosa, staphylococcal scalded skin
syndrome
ll Other: incontinentia pigmenti
NODULES AND PLAQUES
Common or benign: milia, Ebstein’s pearls, Bohn’s nodules, sebaceous
hyperplasia
ll Yellow: sebaceous nevus, juvenile xanthogranuloma
ll Brown or black: congenital pigmented nevus, epidermal nevus
ll Other: subcutaneous fat necrosis, dermoid cyst, fibroma, infantile myofibromatosis, hamartomas, malignant tumors, leukemia
ll
PAPULOSQUAMOUS AND SCALING LESIONS
Common or benign: physiologic desquamation
ll Healthy infant: atopic dermatitis, contact dermatitis, seborrheic dermatitis,
local candida dermatitis, psoriasis
ll Ill infant: acrodermatitis enteropathica, Langerhans cell histiocytosis,
syphilis
ll Other: ichthyosis syndromes, collodion baby, harlequin baby
ll
EROSIONS AND ULCERATIONS
Common or benign: sucking blisters, traumatic injury (e.g., scalp electrode,
diaper erosions, reaction to adhesives)
ll Other: aplasia cutis congenita, herpes simplex, epidermolysis bullosa, toxic
epidermal necrolysis
ll
ALTERED PIGMENTATION
Common or benign: Mongolian spots, transient neonatal pustular
melanosis, isolated café au lait macules
ll Increased pigmentation: Mongolian spots, transient neonatal pustular
melanosis, café au lait macules, lentigines, incontinentia pigmenti
ll Decreased pigmentation: ash leaf macule, nevus depigmentosus,
piebaldism, albinism
ll Purpuric or erythematous: petechiae, dermal hematopoiesis (“blueberry
muffin” lesions), neonatal lupus erythematosus
ll
VASCULAR AND LYMPHATIC LESIONS
Common or benign: Nevus simplex or salmon patch, petechiae on the
presenting part, small hemangioma
ll Other vascular: complicated hemangioma, vascular malformation, port-wine
stain
ll Lymphatic: cystic hygroma, lymphangioma, lymphedema
ll
Data from Cohen B: Pediatric dermatology, ed 3, Philadelphia, 2005, Mosby; Eichenfield LF, Frieden IJ, Esterly NB, editors: Neonatal dermatology, (See also Color Plate 9.) ed 2, Philadelphia,
2008, Elsevier Saunders; and Fletcher MA: Physical diagnosis in neonatology, Philadelphia,
1998, Lippincott-Raven.
occur sporadically or in association with chromosomal
defects or other malformations (Kos and Drolet, 2008).
It occurs most often on the scalp near the vertex and can be
mistaken for a traumatic injury.
Some common and uncommon skin conditions that
may be seen in the newborn are outlined in Box 25-1. Fortunately, most of the skin findings encountered during the
routine examination are due to a relatively few common
FIGURE 25-3 (See also Color Plate 9.) Erythema toxicum neonatorum
with erythematous macules, wheals, and pustules. Pustules predominate in
this example. At times, patchy or confluent areas of erythema occur without pustules. (Reprinted from Eichenfield LF, Frieden IJ, Esterly NB, editors:
Neonatal dermatology, ed 2, Philadelphia, 2008, Elsevier Saunders, p 88.)
conditions, mostly benign, that are described in the following paragraphs. If unusual or unfamiliar lesions that
fall outside the examiner’s “comfort zone” are observed,
consultation with an appropriate subspecialist should be
arranged.
Erythema toxicum neonatorum, or erythema toxicum,
is the most common skin rash in the newborn, occurring
in up to 70% of term infants (Howard and Frieden, 2008;
Lucky, 2008). Lesions can be present at birth, but in most
cases the lesions are first noted at 1 to 2 days. The lesions
are seen predominately on the face and trunk, but can
appear anywhere on the body except the palms and soles.
New lesions may continue to appear for approximately
1 week while older lesions resolve. The typical lesion is an
isolated elevated erythematous papule or pustule 1 to 2 mm
in diameter, surrounded by an irregular area of erythema
1 to 3 cm in diameter (Figure 25-3). When extensive, the
lesions can occur in clusters or become nearly confluent.
Diagnosis can usually be made by appearance alone, but a
scraping of the pustule will reveal an almost pure infiltrate
of eosinophils if confirmation is needed in atypical cases.
Transient neonatal pustular melanosis is a self-limited
process with lesions that evolve through three distinct
phases (Howard and Frieden, 2008; Lucky, 2008; Figure
25-4). Initially, a superficial vesicopustule appears, which
after rupturing leaves a fine collarette of scale around the
unroofed pustule, which is without erythema. The final
stage is a hyperpigmented macule that gradually disappears. The condition is most common in African American
infants. In some cases, only the second or third stages are
seen at birth, the initial stage presumably having occurred
in utero.
Miliaria crystallina is the result of superficial obstruction
of the sweat ducts, producing small, crystal-clear vesicles
that resemble water droplets (Figure 25-5). It is mostly seen
in warm climates or febrile infants. The vesicles are fragile
and can be removed by wiping the skin with a soft damp
cloth (Howard and Frieden, 2008; Lucky, 2008). Miliaria
rubra, also called heat rash or prickly heat, results from sweat
duct obstruction deeper in the epidermal layer, and is more
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
A
C
285
B
D
FIGURE 25-5 (See also Color Plate 10.) Miliaria crystallina. The tiny,
clear vesicles resemble water droplets, with no signs of inflammation.
(Reprinted from Rudolph AJ: Atlas of the newborn, vol 4, Hamilton, Ont,
Canada, 1997, BC Decker, p13.)
common after the first week. Occasionally miliaria rubra
will look sufficiently pustular as to mimic lesions caused by
staphylococcal, candidal, or herpes simplex.
Sucking blisters and calluses result from vigorous sucking on a hand or forearm in utero, and they can cause a
tense, fluid-filled blister, which when ruptured forms an
erosion or a callus. The lesion is most often solitary, but
may be bilateral, and is without inflammation. A sucking
pad or callus may develop on the lips postnatally as a result
of vigorous and frequent nursing.
Neonatal cephalic pustulosis, or neonatal acne may
occasionally be seen at birth, but more typically appears
later, with a mean age of onset of 2 to 3 weeks (Howard
and Frieden, 2008; Lucky, 2008). It is characterized by
inflammatory, erythematous papules and pustules located
primarily on the cheeks with extension over the face and
FIGURE 25-4 Transient neonatal
pustular melanosis. The first stage
consists of small superficial pustules,
without inflammation (A). Collarettes
of scale, the second stage, may be
seen at birth without evident pustules
(B), or may develop postnatally after
pustules have ruptured (C). Small
hyperpigmented macules remain in
the final stage (D), gradually fading
over weeks or months. (Reprinted from
Eichenfield LF, Frieden IJ, Esterly NB,
editors: N
eonatal dermatology, ed 2,
Philadelphia, 2008, WB Saunders, p 89.)
FIGURE 25-6 (See also Color Plate 11.) Neonatal cephalic pustu
losis (neonatal acne). Small, red papules and pustules are seen on the
cheeks and forehead, with some extension into the scalp. Comedones
are absent. (Reprinted from Eichenfield LF, Frieden IJ, Esterly NB, editors:
Neonatal dermatology, ed 2, Philadelphia, 2008, WB Saunders, p 90.)
into the scalp (Figure 25-6). It can be difficult to distinguish clinically from miliaria rubra, but both are benign
conditions and biopsy is not warranted.
Infectious causes of skin lesions are relatively uncommon in the immediate newborn period, but often need to
be considered in the differential diagnosis. Superficial skin
infections of Staphylococcus aureus are rarely present at birth,
but can develop within the first few days. Thrush and candidal diaper dermatitis, common later but not immediately
after birth, usually pose no diagnostic difficulty. Congenital
candidiasis, which is uncommon, typically manifests within
the first day with a papulovesicular eruption that progresses
to pustules, followed by crusting and desquamation (Figure
25-7). Lesions can be widespread and may appear on any
part of the body, including the palms and soles (Carder,
286
PART VII Care of the Healthy Newborn
FIGURE 25-7 (See also Color
Plate 12.) Congenital candidiasis. The
rash may be a diffuse, erythematous
pustular eruption (A) or have diffusely
distributed but distinct pustules (B). In
premature infants, a diffuse scaldlike erythematous dermatitis may be seen (not
shown). (Reprinted from Eichenfield LF,
Frieden IJ, Esterly NB, editors: Neonatal
dermatology, ed 2, Philadelphia, 2008,
Elsevier Saunders, p 214.)
A
B
A
A
B
FIGURE 25-9 (See also Color Plate 14.) Desquamation on the palms
(A) and soles (B) of an infant with congenital syphilis. (Reprinted from
Rudolph AJ: Atlas of the newborn, vol 4, Hamilton, Ont, Canada, 1997,
BC Decker, p 108.)
B
FIGURE 25-8 (See also Color Plate 13.) Herpes simplex. A, The first
signs of herpes infection in this neonate were eroded vesicles at the
corner of the mouth. B, Herpetic vesicles on the face, scalp, and ear of
an infant with respiratory distress and hepatitis. (Reprinted from Cohen B:
Pediatric dermatology, ed 3, Philadelphia, 2005, Mosby, p 36.)
2008; Darmstadt et al, 2000). Neonatal herpes simplex is
unlikely in the first few days after birth, unless the fetal
membranes were ruptured for many days, but it must be
considered whenever vesicles are seen in the newborn. The
skin lesions of herpes simplex start as small, 2- to 4-mm
vesicles on an erythematous base, become pustular after 1
to 3 days, and develop an eschar (Friedlander and Bradley, 2008; Figure 25-8). The mucosal lesions are usually
shallow ulcerations. The “blueberry muffin” skin lesions of
congenital rubella and cytomegalovirus, caused by dermal
hematopoiesis, are unlikely to be seen as an isolated finding.
Affected infants typically have multiple stigmata of congenital infection including growth retardation, microcephaly,
and hepatosplenomegaly. Cutaneous findings of congenital
syphilis are present in only a minority of infected infants,
but they classically involve the palms, soles, and perioral
and anogenital areas (Dinulos and Pace, 2008; Howard and
Frieden, 2008). The rash is highly variable, taking papulosquamous, vesiculobullous, macular erythematous, annular, and polymorphous forms. Desquamation limited to the
palms and soles, with no rash or peeling elsewhere, is suggestive of congenital syphilis (Figure 25-9).
Milia, Ebstein’s pearls, and Bohn’s nodules are epidermal inclusion cysts (Lucky, 2008; Figure 25-10). Milia are
tiny epidermal inclusion cysts seen primarily on the face
and scalp in small numbers; they are smooth, firm, white
papules with no associated erythema. They may be present at birth or appear somewhat later, and they generally
resolve spontaneously within a few months. Larger inclusion cysts, which usually occur singly, are called pearls; the
foreskin and the ventral surface of the penis and scrotum
are common locations. Epidermal inclusion cysts located
on the palate are called Ebstein’s pearls, and those on the
alveolar ridge are called Bohn’s nodules.
Sebaceous hyperplasia occurs mostly on the face, especially on the nose and upper lip. It is due to hypertrophy
of the sebaceous glands caused by androgenic hormonal
stimulation in utero, and it gradually resolves over several
weeks. It is characterized by sheets of smooth, yellowwhite papules with the regular spacing of involved follicles
and no surrounding erythema (Figure 25-11).
Physiologic desquamation occurs in many full-term
infants. These infants will have some dry skin with fine
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
287
A
FIGURE 25-11 (Supplemental color version of this figure is available
online at www.expertconsult.com.) Sebaceous hyperplasia. Sheets of
tiny, white-yellow follicular papules, without inflammation, are seen on
the nose. (Reprinted from Eichenfield LF, Frieden IJ, Esterly NB, editors:
Neonatal dermatology, ed 2, Philadelphia, 2008, Elsevier Saunders, p 87.)
B
C
FIGURE 25-10 Epidermal inclusion cysts. Milia are most commonly seen on the face (A), but can occur anywhere on the body.
Ebstein’s pearls occur on the midline of the hard palate, most commonly
near the junction with soft palate (B). Bohn’s nodules are found along
the gum margins and on the lateral palate (C). Dental lamina cysts
(not shown) are similar inclusions located on the crest of the alveolar
ridge. (Reprinted from Fletcher MA: Physical diagnosis in neonatology,
Philadelphia, 1998, Lippincott-Raven, p124 [A]; and Eichenfield LF, Frieden
IJ, Esterly NB, editors: Neonatal dermatology, ed 2, Philadelphia, 2008,
Elsevier Saunders, pp 503-504 [B and C].)
desquamation at 1 to 2 days of age, particularly on the
hands and feet. A more exaggerated variety of physiological desquamation is common in postmature infant (born at
greater than 41 6⁄7 weeks post-menstrualage). These infants
are often born with dry, thickened skin that cracks and peels
extensively and then normalizes spontaneously over the
course of approximately 1 week. The condition is usually
not difficult to distinguish from the rare inherited disorders
of cornification (ichthyosis) that manifest in the neonatal
period (Irvine and Paller, 2008). These disorders include
Harlequin ichthyosis (caused by mutations in the ABCA12
gene) and “collodion babies” who appear encased in a
thickened, shiny skin that resembles collodion, a phenotype
associated with a number of different ichthyotic conditions.
Mongolian spots (dermal melanocytosis) are macular
areas of a slate grey, blue-grey, blue-black, or deep brown
color. The distinctive appearance is due to the presence of
melanocytes located in the dermis, instead of their typical
site at the dermal-epidermal junction. The spots are most
commonly located on the lower back and buttocks, but can
occur elsewhere. Mongolian spots in this location—which
are common in East Asian, East African, Native American,
and Polynesian infants—tend to fade over several years,
whereas similarly appearing lesions that occur elsewhere
on the body may never resolve (Gibbs and Makkar, 2008;
Lucky, 2008). Dermal melanocytosis in the area of the first
and second divisions of the trigeminal nerve is called the
nevus of Ota. The nevus of Ito is a similar lesion occurring on the neck, upper back, and shoulders in the area of
the posterior supraclavicular and lateral brachial cutaneous
nerves. Unlike Mongolian spots, these nevi do not become
less pigmented with time, and malignant melanoma or
malignant nevi can develop within them, though rarely.
Café au lait macules are tan-brown macules that can
occur anywhere on the body. They are common as an isolated finding in the newborn and are generally benign, but
can be markers of other conditions (Gibbs and Makkar,
2008). The presence of six or more café au lait macules
greater than 5 mm in diameter is considered presumptive
evidence of neurofibromatosis type 1. Multiple large café
au lait macules with irregular borders may be a manifestation of the McCune-Albright syndrome.
The salmon patch (or nevus simplex) is a benign vascular
lesion consisting of erythematous macules or patches frequently seen at the nape of the neck (“stork bite”), on the
eyelids and glabella (“angel’s kisses”), and somewhat less
frequently on the nose and upper lip (Lucky, 2008; Figure
25-12). The lesions are caused by dilated capillaries in the
upper dermis, with normal overlying skin. Those on the
face usually fade or resolve completely within 1 to 2 years,
but 25% to 50% of those on the neck persist throughout
life (Lucky, 2008).
After vaginal delivery of a healthy infant, it is not unusual
or concerning to see petechiae on the presenting part (i.e. the
part of the fetus closest to the pelvic inlet of the birth canal
at the onset of labor) or on other areas of the body subjected
to localized pressure during delivery. A tight nuchal cord
can cause extensive ecchymoses and petechiae on the entire
head. Widespread petechiae are abnormal, however, and are
suggestive of thrombocytopenia or platelet dysfunction.
Hemangiomas are soft, pink-red, compressible vascular
tumors composed of proliferating endothelial cells. Small
288
PART VII Care of the Healthy Newborn
A
FIGURE 25-13 This infant with the Sturge-Weber syndrome has
a port-wine stain in the distribution of the ophthalmic division of
the facial nerve. (Reprinted from Cohen B: Pediatric dermatology, ed 3,
Philadelphia, 2005, Mosby, p 49.)
B
FIGURE 25-12 (See also Color Plate 15.) Salmon patches are commonly seen on the glabella, eyelids, nose, or upper lip, either singly or in
all these locations (A), and on the nape of the neck (B). (Reprinted from
Eichenfield LF, Frieden IJ, Esterly NB, editors: Neonatal dermatology, ed 2,
Philadelphia, 2008, Elsevier Saunders, p. 95.)
hemangiomas are recognized at or shortly after birth in
1% to 3% of healthy term infants and become apparent in
10% of all infants by 1 month old (Cohen, 2005). Hemangiomas are more common in female infants by 2:1 to 9:1 as
compared with males. The incidence is much higher (22%
to 30%) in preterm infants weighing less than 1000 g, but
only slightly higher (15%) in those weighing 1000 to 1500
g (Amir et al, 1986; Enjolras and Garzon, 2008). Infantile
hemangiomas typically undergo a period of growth beyond
that of surrounding tissues for 6 to 12 months, typically
followed by spontaneous involution. Approximately 25%
regress by 2 years of age, 40% to 50% regress by 4 years
of age, 60% to 75% regress by 6 years of age, and 95%
regress by adolescence (Cohen, 2005). Before the hemangioma becomes obvious, careful examination may reveal a
precursor lesion manifesting as telangiectasias surrounded
by an area of pallor or as pale, erythematous, or bruiselike macules and patches. In contrast, vascular malformations are nonproliferative lesions, usually present at birth.
The Kasabach-Merritt phenomenon occurs in association with specific types of large vascular tumors that cause
platelet trapping and severe thrombocytopenia, and is not
associated with the true hemangioma of infancy (Enjolras
and Garzon, 2008). In the rare Klippel-Trenaunay syndrome, capillary malformation and varicose veins are associated with overgrowth of an affected limb.
Port-wine stains are capillary malformations evident at
birth as pink or red patches that grow proportionately with
the child and persist throughout life (Enjolras and Garzon,
2008). The initial pink-red color typically changes to a
deeper red or purple hue with age. Approximately 10% of
port-wine stains that involve the area supplied by the ophthalmic (V1) branch of the trigeminal nerve (Figure 25-13)
are associated with seizures, arterial brain malformations,
and ocular abnormalities that constitute the Sturge-Weber
syndrome (Enjolras et al, 1985).
HEAD
The head is mainly examined by inspection and palpation.
The overall size, shape, and features of the skull should be
noted. The head circumference (maximal occipital-frontal
circumference) should be measured in every neonate and
plotted on an appropriate growth chart (see Figure 25-1).
The measurement of the head circumference is subject to
error and can be significantly affected by molding of the skull
during labor, so the head circumference should be measured
again if the result appears discordant with the visual examination or with the infant’s weight and length, and repeated
after molding has resolved. The soft tissue of the scalp should
be examined for swelling, ecchymoses, and other evidence
of injury because of the forces of labor, and for iatrogenic
injuries including those from application of a vacuum device,
289
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
placement of scalp electrodes, fetal blood sampling, and lacerations with a scalpel during cesarean section.
The quantity of scalp hair present in the newborn is
highly variable, but abnormalities in the distribution, texture, and patterning of the hair are potentially informative.
The hair usually forms a single whorl near the vertex, but
double whorls occur in approximately 5% of newborns.
Abnormal placement or the presence of more than two
whorls may be a marker of abnormal brain development
(Smith and Gong, 1974).
The infant skull is composed of several bony plates, separated by sutures and fontanels. This structure allows the
skull to deform during labor (Figure 25-14). The entire
surface of the skull should be palpated to identify the location and size of the major fontanels and assess for discontinuities. The soft tissue over a fontanel should normally be
flat; a raised or bulging fontanel suggests that intracranial
pressure is increased. It is common to find a palpable discontinuity at a suture, because of vertical displacement of
a skull bone relative to its neighbor as a result of molding.
In some cases, one bone can truly override the other. Such
discontinuities at a suture must be distinguished from the
step-off of a displaced skull fracture. Premature fusion of
one or more of the cranial sutures, or craniosynostosis,
results in a variety of abnormal skull shapes, depending on
which sutures are involved (Fletcher, 1998; Volpe, 2008).
Fusion of the saggital suture produces a narrow skull elongated in the anterior-posterior dimension (scaphocephaly
or dolichocephaly). Fusion of the coronal sutures causes a
widened skull that is shortened in the anterior-posterior
dimension (brachycephaly). Unilateral closure of either a
coronal or lambdoid suture causes an oblique deformity
(frontal or occipital plagiocephaly, respectively). Closure
of a metopic suture produces a triangular skull with a
prominent, narrow forehead (trigonocephaly). Depending on the timing of the fusion, the skull shape may be
abnormal at birth or become visibly deformed later. The
fused suture typically has a palpable elevation or ridge,
which must be distinguished from displacement of a normal suture caused by molding. In the absence of craniosynostosis or overriding, normal mobility at a suture can
be verified by gently applying alternating pressure to the
bones on either side of the suture line.
Caput succedaneum is a diffuse edematous swelling of
the scalp caused by pressure during delivery that results in
fluid accumulation external to the periosteum. The caput
is boggy, has diffuse edges, is not limited by suture lines,
A
B
and most commonly is located over the vertex. It is usually
present at birth and resolves over several days. In contrast,
a cephalohematoma is caused by hemorrhage under the
periosteum; it forms a distinct, firm bump laterally that
does not cross suture lines (Figure 25-15). The cephalohematoma may not appear until several hours after birth,
and it often increases in size over the first 12 to 24 hours.
It typically remains palpable for 2 to 3 weeks, during which
time it may develop a calcified rim. Unlikely a cephalohematoma, a subgaleal hematoma is not confined by the
coronal
suture
posterior
fontanel
(lambda)
lambdoid
suture
mastoid
fontanel
sphenoidal fontanel
A
posterior
fontanel
parietal
tuberosity
sagittal
suture
anterior
fontanel
(bregma)
coronal
suture
B
metopic
suture
FIGURE 25-14 The major sutures and fontanels of the newborn
skull. (Reprinted from Fletcher MA: Physical diagnosis in neonatology,
Philadelphia, 1998, Lippincott-Raven, p 175.)
FIGURE 25-15 (See also Color Plate
16.) Anterior (A) and posterior (B) views
of a large cephalohematoma under the
periosteum of the right parietal bone.
(Reprinted from Fletcher MA: Physical
diagnosis in neonatology, Philadelphia,
1998, Lippincott-Raven, p 185.)
290
PART VII Care of the Healthy Newborn
periosteum and can involve massive blood loss. Depending on the volume of blood that has accumulated, it can
be palpated as a firm or fluctuant mass with poorly defined
edges that may extend onto the neck or forehead. Large
subgaleal hematomas are uncommon, but potentially life
threatening.
Craniotabes (or craniomalacia) is a softening of the
skull, most commonly involving the parietal bones near
the vertex. Gentle pressure on the involved bone produces a sudden collapse, with recoil when the pressure
is released, similar to the way a ping-pong ball collapses
when squeezed. It has usually been attributed to localized
bone resorption or interference with ossification caused by
prolonged pressure on the fetal skull from the maternal
pelvis, but it may be associated with maternal vitamin D
deficiency in some populations (Yorifuji et al, 2008).
A localized defect in the skull can occur in association with
aplasia cutis congenita. A small soft-tissue mass or bulge at
the occiput or in the midline of the forehead near the bridge
of the nose may be caused by an encephalocele protruding
through a small defect in the skull. Large encephaloceles
and major neural tube defects will be obvious if present.
Hydranencephaly, if suspected, can be detected by transillumination of the skull.
FACE
The face and facial expressions should be observed throughout the course of the entire examination. In addition, there
should be a moment when attention is directed to the overall arrangement and proportion of the features of the face as
a whole. It is helpful to view the face from the front, in profile, and looking down from the top of the head. Important
characteristics to note include the symmetry and expressiveness of the face, both at rest and during crying, and the
relative sizes, spacing, and orientation of the features. After
this overview, specific attention should be paid to the ears,
eyes, eyebrows, eyelids, nose and nasal passages, lips, palate,
and mouth. Suggested abnormalities should be described as
precisely as possible. Measurements of the facial features
are not part of the routine newborn examination, but comparison with reference nomograms can be helpful if dysmorphism is suspected (see Chapter 19).
EARS
The external ear is a relatively common site of minor
anomalies. The external ear develops from the first and
second branchial arches, from which the six hillocks of His
grow, migrate, and fuse to form the pinna. Most of the
pinna derives from the second branchial arch. The third
arch contributes the tragus, antitragus, and anterior wall
of the canal (Figure 25-16). The position and orientation
of each ear should be noted, as well as the size, shape, and
structure of the helix, and any signs of trauma. The ear
is palpated to assess the firmness and recoil of the cartilage and to detect any masses or abnormalities of texture.
The skin near the pinna is examined for skin tags, pits, and
sinus tracts. Minor variations in the shape of the ear and
ear lobe are common, and they usually have only cosmetic
significance. A lop ear is characterized by downward folding of the superior helix caused by underdevelopment of
Helix
Triangular fossa
Antihelix
Tragus
External
auditory
meatus
Concha
Lobule
Antitragus
FIGURE 25-16 Anatomy of the external ear. (Reprinted from Fletcher
MA: Physical diagnosis in neonatology, Philadelphia, 1998, Lippincott-Raven,
p 285.)
the upper third of the pinna; this needs to be distinguished
from folding caused by positioning in utero. A cup ear is a
protruding ear with an excessively concave concha. Microtia implies a severely dysplastic and malformed ear, and
it is often associated with abnormalities of the middle ear
and other malformations.
Assessment of hearing by observation of behavioral
responses has been essentially replaced in the routine
examination by automated testing of hearing before hospital discharge (American Academy of Pediatrics, 2007;
US Preventive Services Task Force, 2008). Otoscopy to
inspect the eardrum is not part of the routine examination
in the immediate newborn period. The eardrum is more
horizontal in neonates than in adults, lying almost parallel
to the external canal. Movement of the eardrum is more
difficult to detect than in older infants, and visualization of
the eardrum is often impeded by the presence of amniotic
debris, vernix, or blood in the ear canal. Moreover, findings typical of otitis media in older infants (dullness of the
tympanic membrane, decreased light reflex and translucence, and diminished mobility) may be present in healthy
newborns.
EYES
As part of the inspection of the face, the size, spacing,
and orientation of the eyes, lids, and eyebrows should
be assessed. In microphthalmos the entire eye is small,
whereas microcornea can be an isolated finding in an
otherwise normal eye. Hypertelorism implies that the
spacing between the bony orbits is excessively wide. In
telecanthus, the inner canthi are displaced laterally, giving a false impression of hypertelorism. Hypotelorism is
often associated with holoprosencephaly, Trisomy 13, and
other genetic abnormalities. The palpebral fissures are
downslanting if the medial canthi are higher than the lateral canthi, and upslanting if the lateral canthi are higher.
Ptosis can be difficult to detect reliably in the neonate
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
unless it is severe or unilateral. It may be congenital or
caused by trauma or inflammation. Transient episodes
of gaze disconjugation are common in otherwise healthy
newborns, but persistent eye deviation requires follow-up.
Obstruction at both ends of the lacrimal sac produces a
mucocele (or dacryocele), which appears as a bluish subcutaneous mass that can be mistaken for a hemangioma or an
encephalocele. Mucoceles can sometimes extend into the
nasal passages and cause respiratory distress.
The routine examination of sclera, cornea, and internal
portions of the newborn eye is often limited by the presence of significant eyelid edema in the first few days after
birth. If the corneas and conjunctivae cannot be adequately
visualized during spontaneous eye opening or with gentle
lid retraction, it is normally preferable to defer further
examination of the eyes until the swelling recedes, rather
than to attempt to retract the lids forcibly. A coloboma
is a congenital defect in the formation of the eye, which
can affect any or all of the external or internal structures
of the eye. Colobomas of the iris, most frequently located
inferomedially and producing a keyhole-shaped pupil, are
the most common type visible on external examination. As
part of the routine examination, the lower lid of each eye
should be retracted so that the entire pupil is visible, if not
the entire cornea. If a coloboma of the lid or iris is seen,
ophthalmologic evaluation of internal structures of the
eye is advisable. Cloudy corneas represent glaucoma until
proven otherwise and require prompt ophthalmologic
evaluation even if obvious enlargement of the cornea and
globe (buphthalmos) is not present. Small subconjunctival
hemorrhages are common in newborns, particularly after
vaginal delivery, and do not indicate trauma unless other
findings are present.
An examination of the red reflex of the eyes should be
performed in all neonates before discharge from the nursery and at subsequent health supervision visits (American
Academy of Pediatrics, 2008). Anything that interferes
with the transmission of light through the normally transparent parts of the eye on its path to the fundus and back to
the examiner will result in an abnormality of the red reflex.
Problems potentially detected by the red reflex examination include cataracts, aqueous and vitreous opacities, and
retinal abnormalities including tumors and chorioretinal
colobomas. High-refractive errors and strabismus may
also produce abnormalities or asymmetry of the red reflex.
Dark spots in the red reflex, a markedly diminished reflex,
the presence of a white reflex, or asymmetry of the reflexes
are indications for immediate referral to an ophthalmologist experienced in neonatal evaluation (American Academy of Pediatrics, 2008). The red reflex examination is to
be rated as normal when the reflections of the two eyes
viewed both individually and simultaneously are equivalent in color, intensity, and clarity, and there are no opacities or white spots within the area of either or both red
reflexes (American Academy of Pediatrics, 2008). The red
reflex examination is vital for early detection of visionthreatening and potentially life-threatening abnormalities,
including retinoblastoma. Unfortunately, the sensitivity of
the routine red reflex examination for retinoblastoma is
low, and most retinoblastomas are first detected by family
members rather than pediatricians, despite routine screening (Abramson et al, 2003).
291
NOSE
The nose should be inspected for deformities, with attention to the size, shape, and symmetry of the various components of the nose, including the nostrils, columella, alae
nasi, and the bridge, root, and tip. Asymmetries of the nose
caused by in utero compression are common and need to
be distinguished from malformations. Passage of a catheter beyond the nasopharynx via each nare, usually done
in the delivery room, rules out choanal atresia but does
not ensure that the size of the nasal passages is adequate
for normal breathing. The quality and volume of air flow
through each nostril can be evaluated by listening with the
bell of a stethoscope or by observing the deflection of a wisp
of cotton placed under the nostril. With the mouth closed,
the infant should be able to breath comfortably via each
nostril separately; this can be assessed by briefly occluding each nostril with the examiner’s fingertip. If unilateral
atresia or stenosis is present, the infant will exhibit signs
of distress when the patent nostril is occluded. Flaring of
the alae nasi may occur as the sole or initial symptom of
mild respiratory distress, or it may accompany grunting
and retractions.
MOUTH AND ORAL CAVITY
The lips, perioral area, and oral cavity should be inspected
both at rest and during crying. The shape of the philtrum
should be evaluated when the mouth is relaxed, because
stretching of the upper lip during crying can give a false
impression of a flat philtrum, which is suggestive of fetal
alcohol syndrome. Perioral cyanosis is common and
benign in normal infants in the immediate newborn period,
whereas cyanosis of the tongue and mucous membranes
is always abnormal and requires immediate investigation.
The mucous membranes will be moist and shiny with saliva
if the infant is well hydrated. Excessive oral secretions can
be caused by esophageal atresia or impaired swallowing.
An excessively short frenulum that restricts protrusion of
the tongue can be a cause of feeding difficulty.
In the routine examination, the oral cavity is usually
inspected without the use of a tongue depressor or other
instruments. Epstein’s pearls, Bohn’s nodules, and dental lamina cysts (Figure 25-10) are common findings that
should be indicated to the parents as benign. With patience
and some adjustment of the head position, the entire palate and much of the pharynx can often be visualized. The
tonsils are normally inconspicuous in neonates. The uvula
is short but highly mobile. The soft palate elevates during
crying if cranial nerves (CNs) IX and X are intact.
Inspection of the oral cavity is supplemented by palpation with a gloved finger to assess the shape and integrity of
the palate, and to feel for natal teeth and masses. Although
clefting of the lip and anterior palate will be obvious at
a glance, an isolated cleft of the posterior palate may be
missed unless deliberately sought by palpation. Eliciting
the sucking reflex allows the strength and coordination
of sucking to be assessed. In the routine examination of a
vigorous, alert infant, elicitation of a gag reflex is unnecessarily upsetting to the infant and is normally avoided. The
gag reflex should be tested if the infant is neurologically
depressed or has difficulty swallowing.
292
PART VII Care of the Healthy Newborn
NECK
It is usually convenient to combine palpation of the neck
for lymphadenopathy and masses with palpation of the
clavicles. The entire skin surface of the neck should be visualized and palpated, while turning the head and retracting
the skin to open the neck creases and folds. The range of
motion of the head and neck is evaluated at the same time.
Congenital muscular torticollis at birth is commonly but
not invariably accompanied by a palpable fibrous tumor
(fibromatosis colli) in the shortened sternocleidomastoid
muscle. Nonmuscular causes of torticollis include tumors of
the posterior fossa or cervical spine and malformations of
the cervical spine. A short neck, low hairline at the back
of the head, and restricted mobility of the upper spine
are characteristic features of the Klippel-Feil syndrome.
Redundant skin or a webbed neck may be seen in Trisomy
21 and in Turner and Noonan syndromes. Cystic hygromas are soft, fluctuant masses that transilluminate and are
usually unilateral. Branchial cleft cysts or sinuses are also
found laterally, from the level of the mastoid to the center of the sternocleidomastoid muscle. Thyroglossal duct
cysts are located in the midline high in the neck or under
the chin. Further investigation is needed if the larynx or
trachea are displaced from the midline, or if enlargement
of the thyroid gland is suspected.
CHEST WALL
Although evaluation of the heart and lungs is a central focus
of the examination of the chest, the skin, soft tissue, and
bony structures of the thorax should not be neglected. The
position of the nipples and the presence of any accessory
nipples should be noted. The definition and stippling of
the areola and the size of the breast bud are developmental
features helpful as part of scoring for gestational age estimation. Transient galactorrhea occurs in approximately
5% of term neonates (Madlon-Kay, 1986). Variations in
the shape of the xiphoid process are common, and parents
can be reassured that a prominent or bifid xiphoid is benign
and will usually become much less apparent as the infant
grows. A mildly depressed sternum (pectus excavatum) or
protuberant one (pectus carinatum) is usually of no clinical
consequence. A small, bell-shaped chest in an infant with
respiratory distress may reflect lung hypoplasia or a disorder of skeletal growth. An increase in the anterior-posterior
diameter of the chest (barrel chest) may reflect an increase in
the intrathoracic volume caused by air trapping from meconium aspiration or pneumothorax. Palpation of the chest
wall may reveal irregularities or tenderness, and crepitus
may be felt at the site of a fractured clavicle or rib. Crepitus
can also be caused by dissection of air into the subcutaneous
tissue from a pneumothorax or pneumomediastinum, but
this is an unlikely occurrence in an asymptomatic infant.
LUNGS AND RESPIRATION
The respiratory examination begins with observing the
color of the tongue and mucous membranes, respiratory
rate, breathing pattern, and work of breathing. Respiratory problems are unlikely to be found in an infant who is
centrally pink and breathing comfortably at a relaxed rate.
In the normal newborn, the abdomen expands smoothly
with each contraction of the diaphragm, while the chest
moves inward slightly.
The respiratory rate of the newborn infant is highly
variable when the infant is awake, changing with activity such as feeding and crying. Tachypnea during sleep is
more clearly associated with respiratory problems than is
tachypnea during awake states. During a routine examination of the healthy infant, it is not necessary to measure the
exact respiratory rate, as long as it is clearly within a normal range in an asymptomatic infant, but the respiratory
rate should be determined if tachypnea or an unusually
slow rate is seen. Because short pauses and brief periods
of rapid breathing are common in normal newborns, accurate measurement of the respiratory rate requires counting
for a full minute, preferably when the infant is asleep or
at least not crying. During crying, the quality and vigor
of vocalization are assessed, and the infant is observed for
changes in color and perfusion. Central cyanosis that only
appears during crying may be caused by cardiac or respiratory disease and requires further evaluation. Cyanosis that
resolves during crying may be due to choanal atresia or
stenosis, apnea, or hypoventilation.
The classic symptoms of respiratory distress are nasal
flaring, grunting, and retractions. Nasal flaring and mild
grunting are common in the immediate postnatal period,
but in the healthy newborn they should resolve within
15 to 20 minutes after birth. Increasing respiratory distress
caused by decreasing lung compliance is typically reflected
in a progression from nasal flaring, or mild tachypnea,
or both; to nasal flaring plus mild or intermittent grunting; and then to flaring, grunting, and increasingly severe
retractions. The respiratory rate generally decreases as
the work or effort of breathing increases, as indicated by
the development of grunting and increasing retractions.
When respiratory distress is mild, intermittent grunting at
a slower respiratory rate may alternate with periods of mild
tachypnea. As grunting becomes more severe, the expiratory phase becomes increasingly prolonged. The length
of the grunt, rather than its loudness, correlates with the
severity of distress. Intermittent mild grunting can be misinterpreted by parents as crying. The rhythm of grunting
and its occurrence at the end of expiration are key features
that help to distinguish it from other vocalizations. Retractions require a forceful inspiratory effort and decreased
lung compliance, and they may be absent or less prominent
than expected in an infant with neuromuscular depression.
Nasal congestion, airway obstruction, and airway secretions can produce sounds that are audible without a stethoscope. Noisy or congested nasal breathing and intermittent
sneezing not associated with upper respiratory infection
is common in the first few days after birth. A hoarse cry
suggests an abnormality affecting the vocal cords. Because
intubation of vigorous infants born through meconiumstained amniotic fluid is no longer routine (Halliday and
Sweet, 2001), hoarseness or stridor caused by vocal cord
trauma in healthy term infants is less common than previously. Inspiratory stridor is due to narrowing or partial
obstruction of the upper airway. The presence and loudness of the stridor depends on respiratory effort as well
as the extent of airway narrowing, so that stridor worsens with forceful inspiration during crying. Stridor during
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
crying in an infant with no respiratory distress when quiet
is often due to tracheolaryngomalacia, and it is usually
benign. Stridor that is present during quiet breathing or
present during both inspiration and expiration suggests
the presence of a more significant airway obstruction that
requires further evaluation.
In the routine examination of the infant who is observed
to be centrally pink and breathing comfortably in room
air, brief auscultation of the chest of a sleeping or quiet
infant is usually sufficient to ensure that the breath sounds
are clear, and that air entry is adequate and equal bilaterally. Sounds are not well localized in the neonate, and the
infant might not remain quiet for long, so attention to the
quality of the breath sounds is usually more helpful than
attempting to compare multiple sites. Detection of abnormal lung sounds including crackles, wheezes, and rhonchi
requires further assessment. If more detailed examination is indicated, auscultate over the four major quadrants
anteriorly, on the sides, and on the upper and lower back
bilaterally. Diaphragmatic hernia manifesting in the neonatal period usually causes significant respiratory distress,
but rarely a small diaphragmatic hernia is detected by the
presence of bowel sounds in the chest in an asymptomatic
infant. Spontaneous cough, which is abnormal in neonates,
is most commonly caused by infection or aspiration.
Percussion of the chest, rarely done as part of routine
examination of newborn, can be useful for estimating the
position of the upper margin of the liver. Percussion can
also be used to detect a large effusion or lung consolidation,
but infants with these conditions will have other symptoms
of respiratory distress, so the diagnosis will rely on imaging
studies and not the physical examination. Transillumination
can be useful for supporting a rapid diagnosis of pneumothorax in a distressed infant, but it is not reliable for detecting a small pneumothorax that produces minimal symptoms.
Respiratory symptoms are sensitive but nonspecific
indicators of illness in the newborn, because alterations
in respiration (including apnea) can accompany illness of
many different etiologies. Common causes of subtle or
mild respiratory distress detected in the routine evaluation include retained fetal lung fluid (transient tachypnea
of the newborn), spontaneous pneumothorax, neonatal
sepsis, pneumonia, meconium or amniotic fluid aspiration,
and congenital heart disease. Any infant with respiratory
distress should be transferred to an NICU or observation
nursery for further evaluation, monitoring, and treatment.
CARDIOVASCULAR
The evaluation of the cardiovascular system during the
routine examination has two major goals: to assess the
current status of the circulation and to detect signs of
congenital heart disease, particularly the critical, ductaldependent forms that can produce rapid clinical deterioration in the newborn period. Although detection of heart
disease is important, abnormal circulatory findings in the
newborn are more often secondary to other problems,
including sepsis, hypovolemia, anemia, and hypoglycemia.
The cardiovascular system undergoes marked changes
after birth involving the transition to air breathing, the
progressive decrease of pulmonary vascular resistance, and
the closure of the ductus arteriosus. These changes affect
293
the physical examination of healthy infants and those with
congenital heart disease; therefore the time after birth is
always an important consideration in the interpretation of
the examination.
Physical examination of the cardiovascular system
is not limited to examination of the heart and pulses. It
also requires attention to the infant’s general behavior
and activity, to respiratory symptoms, to the color of the
tongue and oral mucosa, and to the temperature, color and
perfusion of all regions of the body. These features will
usually be inspected at different times during the course
of the routine examination, but they should be reevaluated
during the heart and chest examination if cardiovascular
abnormalities are suspected.
Important features of the pulses are the rate, rhythm, volume and character. The heart rate of the resting well newborn averages 120 to 130 beats/min, with high variability.
Transient sinus tachycardia during vigorous crying is common, but persistent tachycardia with a rate greater than 160
beats/min suggests a need for further investigation. A low
resting heart rate caused by sinus bradycardia (80 to 100
beats/min) during sleep is common in healthy full-term
infants. Isolated premature beats can be noted occasionally in
otherwise healthy infants and are almost always benign. The
femoral pulses in a normal neonate are not easy to palpate,
and they are easily obliterated with pressure. They usually
receive a grade of 2 on the traditional scale of 0 to 4. A patent ductus arterious will not cause bounding pulses until the
pulmonary vascular resistance has dropped enough to allow
significant left-to-right shunting. Uniformly weak pulses
suggest a low output state, usually accompanied by signs of
poor perfusion. If a decrease or delay of the femoral relative
to the brachial pulses is detected, measurement of the blood
pressure in all four extremities can reveal a gradient in the
blood pressure caused by coarctation of the aorta. However,
normal pulses and four-limb blood pressures do not rule out
a coarctation if the ductus arteriosus remains open.
The precordial area and heart are examined by inspection, palpation, and auscultation. A precordial impulse can
be visible in normal newborns, especially during activity,
but visible prominence of the precordial area together with
a palpably increased cardiac impulse suggest cardiomegaly or a hyperdynamic state. Displacement of the cardiac
impulse to the right suggests dextrocardia or a shift in the
mediastinum. Thrills are rarely palpable in the newborn.
Auscultation of the heart should be performed with specific questions in mind, with attention attuned sequentially
for heart sounds, clicks, murmurs, and other abnormal
sounds. Most sounds and murmurs in the newborn are
relatively high-pitched, so the diaphragm is usually used
initially. The bell can be used for further evaluation of lowpitched sounds, if needed. The first heart sound, produced
by closure of the mitral and tricuspid valves, is usually single
in newborns and best heard in the precordial area. The second sound, produced by closure of the aortic and pulmonary
valves, is usually best heard at the left upper sternal border.
With focused attention, slight splitting of the second sound
and its variation with respiration can be appreciated. Third
and fourth heart sounds are abnormal in the newborn. Systolic ejection clicks may be heard in some normal neonates
in the first several hours after birth, but ejection clicks are
abnormal after this period (Johnson, 1990).
294
PART VII Care of the Healthy Newborn
The timing, location, intensity, radiation, quality, and
pitch are important characteristics of heart murmurs that
can help to distinguish physiologic from pathologic murmurs. With repeated examination, physiologic heart murmurs may be detected in as many as 60% of infants during
the first 48 hours (Johnson, 1990). These murmurs are
most commonly systolic ejection murmurs that are transient and soft (grade I or II), and they are usually attributed
to flow through a closing ductus arteriosus or to increasing flow across the pulmonic valve as pulmonary vascular
resistance drops. Vibratory systolic murmurs resembling
Still’s murmur can be heard in some newborns. A murmur detected on a routine newborn examination that is not
clearly physiologic needs further evaluation.
Detection of a suspicious murmur is the most common
reason for further evaluation in an otherwise asymptomatic newborn, but absence of a murmur does not rule out
congenital heart disease. Examples include transposition
of the great arteries and atrioventricular (AV) canal defect.
A decrease or loss of a murmur can be an ominous sign
that accompanies clinical deterioration associated with
ductal closure in an infant with ductal-dependent pulmonary or systemic blood flow.
Pathologic murmurs detected in the first few hours of
life are usually caused by obstruction of ventricular outflow such as aortic stenosis or pulmonic stenosis; they are
crescendo-decrescendo murmurs, usually grade II or III.
This type of murmur may also be caused by subaortic stenosis associated with hypertrophic cardiomyopathy in a
macrosomic infant of a mother with diabetes. Murmurs
associated with defects that produce left-to-right shunting usually appear after a few days, when the pulmonary
vascular resistance has dropped sufficiently. Pansystolic
murmurs (which include and obscure the first heart sound)
that are heard soon after birth are most commonly caused
by AV valve insufficiency, whereas left-to-right shunting
through a ventricular septal defect produces a pansystolic
murmur that typically appears only after 1 to 2 days. Diastolic murmurs are rare in newborns. A continuous murmur in a neonate usually represents an aortopulmonary
communication or an arteriovenous fistula. The location
of an extrathoracic arteriovenous fistula may be revealed by
auscultation of a bruit, most commonly in the head or liver.
ABDOMEN
Examination of the abdomen begins with observation of
the configuration, fullness, and movement with respiration of the abdominal wall. Major abdominal wall abnormalities such as omphalocele, gastroschisis, prune belly
syndrome, and bladder extrophy will be obvious on initial inspection in the delivery room and may be diagnosed
prenatally. Although a large omphalocele will never be
missed, a small one may produce only a slight widening of
the umbilicus and proximal umbilical cord (Figure 25-17).
If such an omphalocele is not detected in the delivery
room before the umbilical cord is clamped and cut, the
intestine within it may be damaged. The abdomen of the
neonate ranges from flat to moderately protuberant, with
substantial variation depending on feeding and the passage
of gas and meconium. A markedly distended abdomen suggests the possibility of significant ascites, a large mass, or
FIGURE 25-17 A small omphalocele that could be injured if the cord
were to be clamped too close to its insertion. (Reprinted from Rudolph AJ:
Atlas of the newborn, vol 4, Hamilton, Ont, Canada, 1997, BC Decker, p 109.)
an intestinal obstruction. A proximal obstruction such as
esophageal or duodenal atresia does not cause abdominal
distension, however. A sunken or scaphoid abdomen may
be seen in the infant with respiratory distress caused by a
diaphragmatic hernia. The umbilicus should be inspected
for meconium staining, for signs of infection, and for the
rare occurrence of pallor and edema or the visible discharge
of urine caused by a patent urachus. At 1 or 2 days after
birth, slight redness of the periumbilical skin is common,
because of irritation from the cord clamp, and needs to be
distinguished from an omphalitis or cellulitis. Counting of
the umbilical vessels is best done in the delivery room, on
the freshly cut cord.
If the infant is asleep or resting quietly, it is prudent
to auscultate for bowel sounds before proceeding to palpate the abdomen. Palpation should be initially gentle and
superficial, to detect any signs of tenderness and the presence of an enlarged liver or spleen. Tenderness must be
distinguished from the tendency of the infant to stiffen the
abdominal muscles in reaction to the touch of the examiner’s fingers, which are usually colder than the infant’s
skin. If the examiner’s fingers remain in gentle contact
within the same spot, the temperature difference will disappear in a moment, and the baby will usually relax and
allow the examiner to proceed without a struggle. In the
healthy infant, the liver edge may be at or slightly above
the right costal margin, or palpable 1 to 2 cm below. The
spleen is rarely palpable unless it is enlarged. Gentle palpation of the lower abdomen can detect an enlarged bladder,
which is the most common cause of a midline abdominal
mass in neonates. Deep palpation to detect small masses
or enlargement of the kidneys is most easily done soon
after birth, before the infant has fed much, and when the
infant is quiet. However, a satisfactory examination can be
done even in a crying infant by keeping the fingers in position, and gradually increasing the depth of palpation each
time the infant briefly relaxes the abdominal muscles while
taking a breath between cries. It is helpful to support the
flank with one hand while palpating for the kidney with
the other, or to palpate with the thumb while supporting
the flank with fingers of the same hand. Percussion of the
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
abdomen is not particularly helpful in routine examination, but it can sometimes help to define the boundaries of
an enlarged liver or bladder.
GENITALIA AND PERINEUM
Brief inspection after delivery is usually sufficient to
identify the infant as male or female. The evaluation and
management of the infant with ambiguous genitalia is discussed in Chapter 92. In both male and female infants, a
soft swelling or bulge in the inguinal area may be due to an
inguinal hernia. The bulge typically appears or increases in
size during crying and is easily reduced with gentle pressure when the infant relaxes. The perineum is inspected to
locate the anus and assess the tone of the anal sphincter.
Absence of a normal anal opening should be detected as
part of the initial evaluation in the delivery room. However, external observation of an apparently normal anus
does not guarantee internal patency of the anus, which is
best confirmed by the normal passage of meconium.
The genitalia are mainly examined by inspection, supplemented by palpation, with the infant in a supine, frogleg position. In the newborn male, the foreskin normally
covers the entire head of the penis, which is adherent to
the glans. The urethral opening is usually hidden by the
foreskin and need not be visualized if the foreskin is intact.
The foreskin is typically incomplete if hypospadias is present, which allows the abnormal position of the urethral
opening to be identified easily. Congenital chordee, a ventral angulation of the head of the penis, may accompany
hypospadias or occur in isolation. Chordee can be missed
unless the examiner straightens the penis by gently retracting the skin along the shaft towards the base of the penis.
In infants who have a generous pad of subcutaneous fat at
the base of the penis, this maneuver also helps to avoid a
false impression that the penis is short. Dribbling of urine
or a weak stream, if observed, is suspicious for bladder
dysfunction or urethral obstruction. The scrotal sac and
inguinal areas are palpated to locate the testes and assess
their size. The scrotal rugae usually appear at approximately 36 weeks’ gestation and cover the entire scrotum
at term. Enlargement of the scrotal sac is most commonly
caused by a hydrocele. Transillumination can help to distinguish a hydrocele from swelling because of congenital
testicular torsion or other masses. Bowel sounds may be
audible in a scrotum enlarged by an inguinal hernia.
During the inspection of the female genitalia, the examiner must gently retract the labia majora laterally to allow
full visualization. The sizes and positions of the labia
minora, clitoris, urethra, and vaginal opening should be
noted. Relative prominence of the labia minora is normal
in preterm infants. Partial labial fusion and an increase in
the size of the clitoris may represent virilization caused
by congenital adrenal hyperplasia or related endocrine
abnormalities. The posterior fourchette should be at least
1 cm from the anal opening. Enlargement of the uterus
because of hydrometrocolpos may produce a protruding
perineal mass. Vaginal tags and mucoid vaginal discharge
are common at birth, resulting from exposure to maternal
estrogen. A slightly bloody vaginal discharge (pseudomenses) caused by hormonal withdrawal is common in healthy
females during the first week after birth.
295
BACK
The back is inspected for asymmetry or abnormal positioning of the shoulders, ribs, and hips. Major neural tube defects
and large masses such as a large sacrococcygeal teratoma
will be detected prenatally or on initial inspection in the
delivery room. In the routine examination, the lumbosacral
area should be inspected carefully for the presence of deep
or unusual dimpling of the skin over the sacrum, for sinus
tracts, for unusual tufts of hair, and for small masses such
as a lipoma or hemangioma, any of which may be associated with spina bifida occulta or tethering of the spinal cord.
The spine is inspected for straightness and palpated for the
integrity and alignment of the posterior spinous processes.
MUSCULOSKELETAL SYSTEM
Routine assessment of the musculoskeletal system begins
with plotting the height, weight, and head circumference
on appropriate growth charts. Detailed measurements of
the limbs and trunk can be made if growth appears disproportionate, but they are not part of the routine examination.
The infant’s posture, muscle mass, and movements should
be observed. As the different parts of the body are surveyed
during the course of the examination, any limitation in the
normal range of joint motion and any localized swelling or
tenderness should be noted. Major limb or skeletal malformations will usually be appreciated on initial inspection in the
delivery room, but minor anomalies such as supernumerary
digits, syndactyly, or nail hypoplasia can be missed if a disciplined approach is not taken. Lack or restriction of normal
movement of an extremity can be caused by trauma, most
commonly a fractured humerus or clavicle, or by an intrinsic abnormality of the joint or limb. Although they generally
require no treatment, fractures of the clavicle are sufficiently
common that the clavicles should be specifically examined in
every newborn. Crepitus and tenderness at the site of a clavicle fracture may be more easily detected if the examiner palpates a clavicle with one hand while using the other to elevate
and rotate the ipsilateral shoulder.
Deformations caused by in utero positioning are not
always easy to distinguish from malformations on an initial examination. Mild inward bowing of the lower legs
and feet is common in newborns. If an inward-turning
foot can be brought easily to a neutral position, a clubfoot
deformity is unlikely, and the angulation of the foot can be
expected to normalize spontaneously.
EXAMINATION OF THE HIPS
Assessment for developmental dysplasia of the hip (DDH)
is an important component of the examination of every
newborn (American Academy of Pediatrics, 2000; Sewell
et al, 2009). Frank dislocations, partial dislocations, and
instability of the humoral head in the socket can be detected
by examination in the newborn period. Nevertheless, DDH
is not always detectable at birth. Statistical risk factors for
DDH include a positive family history, female sex, and
breech presentation. In addition to the Ortolani and Barlow
maneuvers, the examination also involves a focused inspection for indirect clues related to the possible presence of
DDH.
296
PART VII Care of the Healthy Newborn
During general inspection of the infant, attention should
be paid to the resting posture and spontaneous movements;
any asymmetry or unusual positioning of the legs should be
noted. Asymmetry of the gluteal or femoral skin folds may
be a sign of unilateral hip dislocation. A unilateral dislocation may also produce an apparent inequality of leg length,
seen either with the legs in extension (Thomas’ sign) or with
the feet flat on the bed and the knees bent (Galeazzi’s sign).
A restricted range of motion, particularly abduction, is a clue
that may detect either unilateral or bilateral dislocations.
The dislocation of the femoral head during a Barlow
maneuver or its relocation during an Ortolani maneuver
produces what is called a clunk that must be distinguished
from high-pitched clicks of the hip that are common and
benign. Clunks and clicks are perceived by palpation and
are not actually audible. The key element defining the clunk
is a distinct sensation of abrupt movement of the femoral
head as it passes over the rim of acetabulum and drops into
or out of the socket. A dislocated or dislocatable hip has
the distinctive clunk, whereas a subluxable hip is characterized by a feeling of looseness or sliding without a distinct clunk (American Academy of Pediatrics, 2000). Both
maneuvers are performed with the infant supine, starting
with the legs held in neutral rotation and the hips flexed to
90 degrees but not more. Each hip is examined separately.
For the Ortolani maneuver, the index and middle fingers of
the examiner’s hand are placed along the greater trochanter with the thumb placed along the inner thigh near the
knee. The other hand stabilizes the pelvis. The examiner
gently abducts the hip by rotating the thumb outward while
lifting anteriorly with the fingers. A distinct sensation of
movement is felt when a posteriorly dislocated hip relocates
during abduction. For the Barlow maneuver, the index and
middle fingers are also positioned along the greater trochanter, but the examiner’s hand is rotated so that the base of the
thumb is on top of the knee. The maneuver is performed
by adducting the leg until the knee is in the midline, and
then applying gentle pressure to the knee in a downward
direction along the adducted femur. A clunk is felt if this
maneuver induces the femoral head to exit the acetabulum
posteriorly. Minimal force is used in either maneuver.
The examination is considered positive for DDH if a
clunk of dislocation or reduction is elicited during the Barlow or Ortolani maneuvers, in which case prompt referral
to an orthopedist is strongly recommended. The examination is considered equivocal if the Barlow and Ortolani test
results are negative, but warning signs such as asymmetric
creases, apparent or true leg length discrepancy, or limited
abduction are found. In this case, the recommended next
step in evaluation is a follow-up examination of the hips by
a pediatrician at 2 weeks (American Academy of Pediatrics,
2000).
NEUROLOGIC EXAMINATION
The neurologic system is assessed during the course of the
general newborn examination and by performing some
specific maneuvers to elicit neonatal reflexes. Because the
healthy infant’s responses vary with the state of alertness,
and because the infant’s tolerance for prolonged examination is limited, eliciting a perfect response to each maneuver should not be expected. Fortunately, there is sufficient
redundancy in the routine examination that it is not difficult in most cases for the examiner to be satisfied that
the infant is neurologically normal. When the examination is not fully reassuring, repeating selected parts of the
examination at a later time may be more helpful in clarifying findings than attempting an extended examination
at one time. The newborn screening examination includes
assessment of the infant’s alertness, spontaneous activity, posture, muscle tone and strength, head control, and
responses to manipulation and handling.
The overall assessment of alertness, tone, and activity
is one of the most important components of the newborn
examination. Diminished alertness, tone, or spontaneous
activity are sensitive but nonspecific indicators of illness
that are much more likely the result of other causes, such as
neonatal sepsis, than a specific neurologic abnormality. The
typical healthy newborn is easily awakened from sleep and
remains alert through the remainder of the routine examination, shifting among states of quiet alertness, active alertness, and crying. The examiner’s initial attempt to quietly
auscultate the breath and heart sounds of a sleeping infant is
often foiled by the infant’s prompt arousal and crying. The
healthy newborn demonstrates a vigorous cry when upset,
but is able to self-console or to be consoled with holding,
sucking, or feeding. An infant who is stuporous or difficult
to arouse is clearly abnormal and needs further evaluation,
as does an infant who is unusually irritable or inconsolable.
The typical newborn will be rather alert for several hours
after delivery, but may then provoke parental concern by
becoming relatively sleepy and uninterested in feeding for
the remainder of the first 24 hours. As long as the infant
continues to be easily arousable and the examination result
remains otherwise normal, parents can be reassured that
the infant will likely begin feeding much more vigorously
on the second day. Decreases in tone or alertness occurring
1 day or more after birth in a previously vigorous infant
are not normal and require prompt investigation for sepsis
or other problems, including inborn errors of metabolism
that can cause progressive symptoms starting a few days
after birth because of accumulation of toxic metabolites.
Because the newborn cannot respond to verbal questions
or commands, assessment of the infant’s sensory functions
in the routine examination depends on observation of the
strength and quality of the infant’s movements in response
to handling during the examination and to specific local
stimulation, such as the elicitation of the palmar and plantar grasp reflexes and the rooting and sucking reflexes.
Assessment of vision in the routine newborn examination
is limited to observing pupillary constriction and a blink
response to light, which are subcortical responses, and to
observing the infant’s visual attentiveness. The healthy
term newborn is expected to be able to visually fixate on
and follow the examiner’s face, but failure to observe this
response during the course of the routine examination is
common, because it can take more time and patience to
elicit than are available. Hearing can be evaluated behaviorally by observing the infant’s responses to the ringing of
a bell and other sounds. However, behavioral testing can
only detect profound, bilateral hearing loss, and it has been
effectively replaced in the routine newborn examination
by automated hearing tests (American Academy of Pediatrics, 2007; US Preventive Services Task Force, 2008).
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
The assessment of motor function relies on the observation of the infant’s posture and spontaneous movements,
observation of the infant’s general responses to stimulation and handling, and the elicitation of specific reflexes.
The healthy term newborn normally maintains a resting
posture with elbows, hips, and knees strongly flexed. The
scarf sign, forearm recoil, square window of the wrist, the
heal-to-ear maneuver, and popliteal angle are measures of
tone and flexibility commonly scored in the gestational age
assessment (Ballard et al, 1991; Dubowitz et al, 1970). The
active tone and strength of upper extremity muscle groups
are routinely assessed by eliciting the palmar grasp and
by arm traction during the pull-to-sit maneuver. Lower
extremity strength and tone are assessed during the general examination of hips and feet, and by testing for the
supporting reaction, stepping reflex, Babinski’s reflex, and
ankle clonus. The neck flexors may be evaluated during the
standard pull-to-sit maneuver, or by lifting the shoulders
to pull the baby to a sitting position. The neck extensors
can be evaluated by tilting the infant forward from a sitting
position, or evaluated along with truncal tone by suspending the infant in a prone position with the examiner’s hand
under the chest (ventral suspension maneuver).
When observing the infant’s movements, the examiner
should note any asymmetry and pay attention to qualitative
characteristics such as the relative smoothness versus jerkiness or tremulousness of movement. Unusual jitteriness may
be a symptom of hypoglycemia or hypocalcemia, especially
in infants of mothers with diabetes, infants that are small
or large for gestational age, or infants who were exposed to
opiates or other drugs in utero and are experiencing withdrawal. Passive restraint of an extremity should inhibit jittery movements, but will not stop the rhythmic contractions
of clonic seizure activity. In addition to clonic movements,
signs of neonatal seizures can include tonic posturing,
repetitive stereotyped movements of the face or extremities, tonic horizontal eye deviation or nystagmoid jerking,
staring or blinking, apnea, and unexplained changes in heart
rate or blood pressure. Although it is uncommon to observe
an actual seizure during the routine newborn examination,
the pediatrician or neonatologist is frequently called to evaluate an infant because of concern about unusual movements
seen by a parent or nurse. In the otherwise healthy newborn
who is fully alert and responsive, jerky or abrupt movements
that are evoked by stimulation (e.g., startles), that can be
suppressed by passive restraint (e.g. jitteriness), and that
are not accompanied by autonomic changes or changes in
alertness are unlikely to be seizures. Transient episodes of
disconjugate gaze are also not unusual in normal newborn
infants, particularly when the infant is entering or awakening from sleep.
Neonatal Reflexes
The neonatal or primitive reflexes frequently tested during routine examination of the newborn include the Moro
reflex, the asymmetrical tonic neck reflex, truncal incurvation (Galant reflex), the palmar and plantar grasp reflexes,
the Babinski’s reflex, and the placing and stepping reflexes.
The Moro reflex can be elicited following the pull-tosit maneuver, by lowering the infant until there is only a
slight space between the neck and bed, and then allowing
297
the infant to fall back suddenly. Alternatively, the Moro
response can be elicited by the “drop” method: the examiner lifts the baby completely off the bed, supporting the
head and trunk with both hands and keeping the baby
supine, and then rapidly lowers the baby by approximately
4 to 8 inches. The complete Moro reflex involves a quick
bilateral abduction of the arms and extension of the forearms with full opening of the hands, followed by smoother
and slower return of the hands toward the midline, with
curling of the fingers. The startle reflex is similar to the
Moro reflex, but without full extension or hand opening,
and may occur spontaneously or be evoked by a sudden
noise or movement.
The position of the neck affects the tone of the extremities via the asymmetrical tonic neck reflex. This should be
kept in mind during observation of the infant’s spontaneous movements, because it can cause a false impression
of asymmetry if the position of the neck is not taken into
account. To test the asymmetric tonic neck reflex, turn the
infant’s head 90 degrees to one side for 15 seconds, keeping the infant lying on the back with the shoulders horizontal. In the complete response, the ipselateral arm and
leg will extend and the contralateral arm and leg will flex,
producing the “fencing” posture. The test is then repeated
with the head turned to the other side. Observation of
the complete response is reassuring, but its absence is not
necessarily abnormal, because partial and unidirectional
responses are common in normal newborns. However, an
unusually sustained or exaggerated response is abnormal.
The truncal incurvation reflex is elicited with the infant
held in ventral suspension by stroking lightly down the
back on one side, and then the other. The normal response
is for the infant to curve the spine strongly, concave toward
the stimulated side.
The supporting, placing, and stepping reflexes are elicited with the infant held upright. The supporting reaction is
elicited by lowering the infant vertically until both feet touch
the surface of the bed or table. A positive response, usually seen after a slight delay, is partial extension at the hips
and knees, as though the infant is attempting to stand and
support his or her weight. The stepping reaction is tested
by lowering the infant so that one foot touches the surface,
with the infant tilted slightly forward. The infant should flex
that leg and extend the other, as though taking a step. The
response can sometimes be sustained for several alternating
steps. The placing reaction is elicited by lifting the infant to
bring the dorsum of one foot in contact with the underside
of a table or bassinet edge. In a positive response, the infant
lifts the foot up and places it on the top surface.
Babinski’s reflex, which is normal in neonates, consists of dorsal flexion of the big toe and spreading of the
other toes in response to stroking the foot laterally. Firm
pressure on the sole of the foot, in contrast, will elicit a
plantar grasp. The deep tendon reflexes are usually not
elicited during routine examination of the well newborn,
but they are helpful as part of a more complete examination if neurologic abnormalities are suspected. The pectoralis, biceps, brachioradialis, thigh adductor, crossed
adductor, knee jerk, and ankle jerk reflexes are the most
readily elicited (Volpe, 2008). Ankle clonus may be elicited by quickly dorsiflexing the foot, which in a healthy
term infant should produce no more than approximately
298
PART VII Care of the Healthy Newborn
five beats of alternating extension and flexion with rapidly
decaying intensity.
Brachial Plexus Injury
Although fortunately not common, brachial plexus injury
is one of the more frequent neurologic abnormalities found
in the otherwise healthy newborn, occurring in about
0.5 to 2 per 1000 live births. Often there is a history of
difficult delivery because of shoulder dystocia. Brachial
plexus injury can occur in isolation or in conjunction with
fractures of the clavicle or humerus. Typical findings in
Erb’s palsy are an inability to abduct and externally rotate
the shoulder, flex the elbow, and supinate the forearm
because of injury to C5-C6 (Volpe, 2008). If C7 is involved,
wrist and finger extension are also weak (Figure 25-18).
A distal brachial plexus injury involving C8-T1 causes
weakness of wrist and finger flexion.
Cranial Nerves
Observation and maneuvers during the routine examination provide at least partial assessment of all the cranial
nerves (Volpe, 2008) except for the olfactory nerve (CN I),
which is not routinely tested. Visual attentiveness, the ability to fix and follow, eyelid closing in response to light, and
the pupillary light reflex require the optic nerve (CN II).
The pupillary light reflex also tests the oculomotor nerve
(CN III). Extraocular movements are controlled by CNs
III, IV, and VI. Vertical gaze is difficult to assess in neonates, but horizontal gaze in both directions can usually
be verified by observing spontaneous eye movements or
by the rotation test. The examiner performs the rotation
test by turning in place while holding the infant upright
and supporting the back of the head. If vestibular functions (CN VIII) are intact, the infant will turn the head
in the direction of rotation or, if the head is restrained,
turn the eyes in that direction. Eliciting rooting or a facial
grimace in response to touching the face tests the sensory
portion of the trigeminal nerve (CN V). The corneal reflex
can be tested by touching the cornea with a wisp of sterile
cotton, but this is not part of the routine newborn examination. CNs V, VII, IX, X, and XII are all involved in normal sucking and swallowing. The motor portion of CN V
controls the muscles of mastication, which are involved in
the jaw-closing phase of the suck and are assessed during
elicitation of the suck when the infant bites down on the
examiner’s finger. Although not part of the routine examination, the masseter or jaw-jerk reflex can be elicited by
placing the forefinger of one hand on the infant’s relaxed
chin and tapping it with the forefinger of the other hand.
FIGURE 25-18 Hand and arm position in an infant with Erb’s palsy
involving C5, C6, and C7. (Reprinted from Fletcher MA: Physical diagnosis in neonatology, Philadelphia, 1998, Lippincott-Raven, p 450.)
The facial nerve (CN VII) is required for pursing of the
lips in sucking, as well as normal facial expression and tone.
Compression of the facial nerve by in utero positioning or
by forceps injury are common causes of unilateral facial
weakness in the newborn period (Figure 25-19). CNs IX
and X are needed for normal swallowing and the gag reflex.
CN XII is involved in the milking action of the tongue
during sucking and swallowing. Taste, mediated by
CN VII (anterior two thirds of the tongue) and IX (posterior third) is not evaluated in the routine examination.
Sternocleidomastoid function (CN XI) is assessed by evaluating head flexion and lateral rotation. Because head control is often poor in normal newborns, it can be difficult to
detect an abnormality unless it is unilateral. However, an
asymmetric abnormality of a sternocleidomastoid is caused
more commonly by torticollis than dysfunction of CN XI.
The vestibular portion of CN VIII is assessed by the rotation test, as noted previously, or by the doll’s eye maneuver. The auditory portion of CN VIII is best assessed using
brainstem auditory evoked responses, rather than behavioral responses to auditory stimulation.
CHAPTER 25 Initial Evaluation: History and Physical Examination of the Newborn
A
C
299
B
FIGURE 25-19 (See also Color
Plate 17.) Unilateral facial weak
ness in two infants. There is mild
facial asymmetry with flattening of
the nasolabial folds at rest (A and C)
and more obvious asymmetries of
the grimace and eye closing during
crying (B and D). The weakness is on
the infant’s left side in A and B, and
on the infant’s right side in C and D.
(Reprinted from Fletcher MA: Physical
diagnosis in neonatology, Philadelphia,
1998, Lippincott-Raven, p 457.)
D
SUGGESTED READINGS
Books
Cohen B: Pediatric Dermatology, ed 3, Philadelphia, 2005, Mosby.
Eichenfield LF, Frieden IJ, Esterly NB, editors: Neonatal Dermatology, ed 2,
Philadelphia, 2008, Saunders.
Fletcher MA: Physical Diagnosis in Neonatology, Philadelphia, 1998, LippincottRaven.
Rudolph AJ: Atlas of the Newborn, Hamilton, Ontario, 1997, B.C. Decker.
Volpe JJ: Neurology of the Newborn, ed 5, Philadelphia, 2008, Saunders.
Guidelines and Policy Statements
Early detection of developmental dysplasia of the hip: clinical practice guideline.
Committee on Quality Improvement, Subcommittee on Developmental
Dysplasia of the Hip. American Academy of Pediatrics, Pediatrics 105:896-905,
2000.
Hospital stay for healthy term newborns: policy statement. American Academy of
Pediatrics, Pediatrics 125:405-409, 2010.
Red reflex examination in neonates, infants, and children: Joint policy statement.
American Academy of Pediatrics Section on Ophthalmology, American
Association for Pediatric Ophthalmology and Strabismus, American Academy
of Ophthalmology, American Association of Certified Orthoptists, Pediatrics
122:1401-1404, 2008.
Universal screening for hearing loss in newborns: US Preventive Services Task
Force recommendation statement, Pediatrics 122:143-148, 2008.
Online Resources
Lehmann CU, Cohen BA: Dermatlas: an online collaborative education tool, The
Internet Journal of Dermatology 1, 2002. Available at http://dermatlas.med.jhmi.
edu/derm/.
Ballard JL, Khoury JC, Wedig K, Wang L, Eilers-Walsman BL, Lipp R: New
Ballard Score, expanded to include extremely premature infants, J Pediatr
119:417-423, 1991. Descriptions and video demonstrations by JL Ballard.
Available at www.ballardscore.com.
Pediatric NeuroLogic Exam, Available at http://library.med.utah.edu/pedineurologic
exam/html/home_exam.html. A tutorial with video demonstrations and descriptions
by PD Larsen and SS Stensaas.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
26
Routine Newborn Care
James A. Taylor, Jeffrey A. Wright, and David Woodrum
Two central paradoxes underlie the care of healthy newborn infants. First, although birth is perhaps the oldest and
most natural of all human processes, the infant mortality
rate has been extraordinarily high until recently. The second paradox in providing newborn care is that neonates
are at once the healthiest and most vulnerable patients in
medicine. Recent medical history is replete with examples
of the pendulum swinging too far in each direction around
these paradoxes. The promotion of scheduled feeding using
infant formulas rather than breastfeeding is an example of
the medicalization of neonatal care. Conversely, recent
resistance to treatments that prevent uncommon but disastrous conditions represent a denial of the benefits provided
by medical care.
Therefore optimal care of a normal neonate is an
attempt to balance these competing forces. Systems of
care should be designed to support the concept that newborn infants are extraordinarily healthy and require little
intervention beyond promotion of breastfeeding. Interventions for which there is evidence that the benefits far
outweigh the risks should be provided as unobtrusively
as possible. Simultaneously, while promoting natural
care for these newborns, health care providers need to be
vigilant for the early identification of neonates who are at
risk for conditions such as dehydration, sepsis, and severe
hyperbilirubinemia.
The goal of this chapter is to provide an evidence base
for the promotion of healthy newborn care by parents, the
rationale for monitoring full-term neonates for various
conditions, a risk-benefit analysis of common treatments,
and the significance of common prenatal and postnatal
findings. Rather than providing a comprehensive prescription for the care of these infants, it is hoped that the reader
will integrate the information provided in this chapter
with expert opinion and clinical experience to determine
the proper care of healthy newborns.
INITIAL ASSESSMENT
Timing of the initial assessment of a full-term newborn
is dependent on the condition of the infant and parental
preference. In most instances a health care professional
who is present at the birth will make a general appraisal of
the infant and alert the child’s provider if there is an acute
problem necessitating an immediate evaluation. Usually
the neonate will be healthy and the assessment can be
timed so as not to interfere with breastfeeding, bonding
with the family, and routine care.
Before examining a healthy newborn, the mother’s
medical history should be reviewed to identify issues
that would affect the care or prognosis of the infant. For
example, a history of diabetes in the mother would lead to
glucose testing in the neonate. Maternal drug use should
be assessed for possible teratogenic effects, possibility of
300
symptoms of withdrawal in the infant, and compatibility
with breastfeeding. It is important to review the pregnancy
history and focus on estimated gestational age, the results
of screening for genetic conditions, and the results of prenatal ultrasound examinations. Perinatal events such as
type of delivery, length of time that membranes were ruptured, and Apgar scores should also be reviewed. Finally,
it is critical to review the mother’s social history to insure
that the newborn will be raised in a nurturing environment
and to identify high-risk situations for which interventions
are needed before or shortly after discharge from the newborn nursery.
The results of several laboratory tests commonly performed on pregnant women will determine the need for
treatment or monitoring during the newborn nursery stay.
These tests include maternal HIV, hepatitis B surface
antigen status, and syphilis. The mother’s blood type, Rh
status, and antibody test results are useful in identifying
newborns with an increased risk of hyperbilirubinemia.
It is important to note the results of testing for maternal
colonization with group B Streptococcus (GBS) and the type
and timing of antenatal antibiotic prophylaxis in mothers
with a positive test result for GBS.
The infant’s weight, length, and head circumference
should be measured shortly after birth and plotted on a
standardized chart. Although the most common reason
for a significant discrepancies among weight, height, and
head circumference percentiles is an inaccurate measurement, a valid discrepancy warrants close clinical observation or testing. Glucose testing may be indicated for
newborns found to be small or large for gestational age. If
the estimated gestational age of the infant is inconsistent
with the growth parameters a formal evaluation using a
Dubowitz-Ballard assessment of gestational age may be
helpful.
When examining a newborn infant for the first time,
the initial focus is directed toward an overall assessment
of the child’s health. Observation and auscultation of the
chest allows for detection of an irregular heart rate, murmur, or acute lung condition such as pneumothorax. The
heart rate and respiratory rate can be measured. Normal
values for heart and respiratory rate in a newborn infant
are 100 to 160 beats/min and 35 to 60 breaths/min. Evaluation of skin color may be useful for identifying cyanotic
congenital heart disease or pulmonary conditions in a
neonate. If uncertainty exists about the presence of cyanosis, oxygen saturation can be measured quickly with
a pulse oximeter. The newborn’s tone, general posture,
and movement should be assessed; abnormalities may be
indicative of an acute or chronic central nervous system
problem or sepsis.
There are several purposes for the rest of physical examination, including maternal reassurance and education
about normal variations. Asymptomatic conditions can be
CHAPTER 26 Routine Newborn Care
220
BOX 26-1 C
linical Signs Compatible With
Hypoglycemia
200
ll
180
ll
ll
Plasma glucose (mg/dl)
160
ll
ll
140
ll
120
100
ll
*
ll
(35)
80
(55)
(51)
(69)
(51)
ll
(26)
(55)
(40)
(49)
ll
(52)
40
6
68
–1
6
–9
2
–7
8
–4
4
–2
97
4
73
3
49
2
25
1
12
0
Age in hours
FIGURE 26-1 Predicted plasma glucose values during the first week
of life in healthy term neonates appropriate for gestational age. (Adapted
from Srinivasan G et al: Plasma glucose values in normal neonates: a new look.
J Pediatr 109:114, 1986.)
diagnosed, such as some types of congenital heart disease
or developmental dysplasia of the hips. Finally, the examination is a useful screen for rare but serious conditions.
ROUTINE TESTING
GLUCOSE
The fetal blood glucose level is approximately 80% of the
maternal level. After birth and separation of the infant from
its major energy supply, the infant’s glucose falls by an
average factor of 0.5. Over the next several hours, it gradually increases to a level approaching that of older infants
(Figure 26-1). Critical factors involved in this normal
adaptive process include transient inhibition of the infant’s
insulin secretion and an increase in the counter regulatory
hormones, including growth hormone, cortisol, epinephrine, and glucagon (Polin et al, 2004). The end result is the
promotion of liver glycogen breakdown, gluconeogenesis,
and tissue lipolysis. Given this normal sequence of events,
there is no reason to routinely screen the blood glucose
in infants who are products of uncomplicated pregnancies, labor, and delivery. Clinical scenarios that might be
expected to alter the normal sequence of events and indicate the need for early glucose screening include:
ll
ll
ll
Infants with midline facial anomalies, which might be
markers for pituitary deficiency
Infants demonstrating hepatomegaly, suggesting glycogen storage disease
Any infant showing clinical signs of hypoglycemia (Box
26-1)
The treatment approach to confirmed hypoglycemia
depends on the glucose level, the presence of symptoms,
or both (see Chapter 94).
( ) Number of samples
* Mean & 95% confidence interval
20
ll
Poor feeding
Lethargy
Hypotonia
Irritability
Tremor
Seizure-like activity
Apnea
(52)
60
ll
301
A pregnancy history of maternal treatment with
β-agonists or intrapartum intravenous glucose
Infants of diabetic mothers
Infants demonstrating intrauterine growth restriction
Premature infants
Infants delivered after in utero or intrapartum fetal
distress
NEWBORN METABOLIC SCREENING
Newborn screening for metabolic disorders began in
1962 when 29 states participated in a trial of testing for
phenylketonuria. With the implementation of screening
programs, criteria were proposed for determining which
conditions should be screened. It was recommended that
only disorders that were important health problems be
included in screening programs. The condition should
be detectable before the onset of significant symptoms.
Importantly, a specific treatment to prevent adverse clinical consequences from the disorder should be available,
and the screening program for the condition should be
cost effective (Tarini, 2007). Based on these criteria, conditions such as congenital hypothyroidism and congenital
adrenal hyperplasia were slowly added to newborn screening tests in many states, and subsequently conditions such
as sickle cell disease were added. Although there is no specific treatment for sickle cell disease, there was evidence
that the use of a newborn screening program to identify
infants with the disorder led to early initiation of penicillin treatment, which resulted in fewer deaths from sepsis
than when disease was identified at the onset of symptoms
(Vichinsky et al, 1988). Given the demonstrable effectiveness of early identification, sickle cell disease met criteria
for newborn screening.
The advent of tandem mass spectrometry in the 1990s
revolutionized newborn metabolic screening. With this
technology it is possible to test for a multitude of conditions using a small sample of blood. In 1995, the average
number of conditions included in state-mandated screening programs was eight; by 2005 this had increased to 19,
with some states testing for up to 46 conditions. Unfortunately, this increase in newborn screening has been
controversial. Some of the conditions included do not
meet the established criteria for screening, in that there
are no known effective treatments, and in some cases it
is not known whether the targeted condition always leads
to disease. In addition, with increasing numbers of tests
come increasing numbers of false-positive results, with the
302
PART VII Care of the Healthy Newborn
resulting parental anxiety and potential for the overuse of
medical services (Tarini et al, 2006).
In an attempt to define a rational list of disorders for
which newborn screening is appropriate, the American
College of Medical Genetics used an iterative process to
identify 29 “core conditions” that should be included in
mandatory screening programs (Watson et al, 2006). As of
2009, approximately 25 of these conditions were included
in the screening panels of at least 48 states (National Newborn Screening and Genetics Resource Center, 2009). The
most common disorders included in newborn metabolic
screens in the United States are congenital hypothyroidism (1 case per 3000 to 4000 infants) and sickle cell disease
(American Academy of Pediatrics [AAP] Newborn Screening Task Force, 2000). The incidence of phenylketonuria
is approximately 1 in 15,000. For many of the core conditions for which screening is recommended, incidence rates
are in the range of 1 in 100,000 to 1 in 200,000 (Kaye et al,
2006). For some disorders, the incidence rate is unknown.
The expansion in newborn screening programs presents
a challenge to health care providers. It is difficult to remain
knowledgeable about all of the conditions that are screened,
the incidence of the various conditions, and their natural
histories and treatments. Fortunately, many states have
informative Web sites providing up-to-date information
for parents and health care professionals. In addition, the
American College of Medical Genetics maintains online
information for health care practitioners, including synopses of conditions commonly included in screening programs
and appropriate management of infants with a positive
result (accessible at http://www.acmg.net/resources/
policies/ACT/condition-analyte-links.htm).
HEARING SCREEN
Newborn hearing screening has become nearly universal in the United States. Thirty states plus Guam, Puerto
Rico, and the District of Columbia have established mandatory early hearing screening programs, and 17 states plus
Guam, Puerto Rico, and the District of Columbia require
all health insurers to cover the test (National Conference
of State Legislatures, 2009). Newborn hearing screening
and early intervention is endorsed by the AAP (AAP Joint
Committee on Infant Hearing, 2007), and the U.S. Preventive Services Task Force (2008). Many nurseries now
use a two-step screen, first using automated otoacoustic emissions screening followed by auditory brainstem
response done in those who fail the otoacoustic emissions
screening.
Before universal screening, there were questions
about the utility of newborn hearing screening, including whether the false-positive rate creates more harm
than the benefit of detecting a small number of infants
with hearing loss (approximately 1 in 1000 newborns)
and whether early intervention is effective (Keren et al,
2002). The specificity of a two-step screening program
for hearing loss approaches 0.99. However, even with this
high specificity, there at least six false-positive screens for
every true positive screening result, because of the low
rate of hearing loss (Nelson et al, 2008). The effect of
false-positive screens has been minimally investigated.
In one study, at 6 months of age, parents of newborns
with false-positive screens continued to express worries
about their child’s hearing after subsequent testing confirmed normal hearing. Fortunately, using standardized
measures, there was no evidence of an increase in general
anxiety these parents compared with parents of babies
with normal newborn hearing screens (van der Ploeg
et al, 2008).
False-negative screens are also a concern, because the
equipment used is designed for screening and because
most auditory brainstem response screening is designed
to detect moderate or greater hearing loss. As a result,
there is a chance that the screen will be falsely negative
in approximately 2% of newborns (Johnson et al, 2005),
and pediatricians should continue surveillance of hearing
status during childhood.
The results of recent studies have shown improved reading and communication skills in hearing-impaired children
identified during the period of universal newborn hearing
screening. Furthermore, hearing-impaired children who
enroll in treatment programs during the first 3 months of
life have better language outcomes at school age (McCann
et al, 2009; Vohr et al, 2008). Investigators in a Belgian
study found that a cause for hearing loss can be determined
in approximately half of children identified by newborn
screening, of which 60% have genetic origins and 19%
have cytomegalovirus infection (Declau et al, 2008).
It is challenging to ensure that all children with positive newborn screening test results have confirmatory
audiologic testing and begin treatment by 3 to 6 months
of age. The number of patients lost to follow-up remains
problematic in many areas, and services such as amplification may take time to arrange. Efforts must be made to
improve these problems for hearing screening programs
to be maximally effective.
PRENATAL ULTRASOUND SCREENING FOR
BIRTH DEFECTS
Second-trimester ultrasound screening for fetal anomalies
has become increasingly routine. Major fetal organ system abnormalities can, for the most part, be identified and
referred for appropriate fetal and neonatal management.
There are, however, a number of ultrasound findings that
have a variable natural history, may or may not be markers for serious conditions, and do not always result in a
definitive prenatal workup. These findings often do not fit
within the pediatric lexicon, and they can present a challenge to the pediatrician regarding parent counseling and
determining management in the neonatal period.
Central Nervous System Findings
Choroid plexus cysts are found in 1% to 3% of second-trimester fetal ultrasound examinations. They are transient,
functionally benign in nature, and resolve spontaneously
before term. If one or more choroid plexus cysts are found
in isolation on prenatal ultrasound examination, no adverse
effect on fetal growth and development has been noted.
Therefore, without other risk factors, no further evaluation is needed in an infant with this isolated finding and a
benign prenatal course. Choroid plexus cysts are believed
to be a “soft marker” for aneuploidy (particularly trisomy
CHAPTER 26 Routine Newborn Care
18) when associated with other fetal anomalies or with
maternal risk factors such as maternal age. In such situations, begin an appropriate prenatal evaluation, such as
karyotyping (DiPietro et al, 2006; Lopez and Reich, 2006;
Sohaey, 2008b).
Agenesis of the corpus callosum is reported to occur
in 0.01% to 0.7% of unselected postnatal populations.
Aneuploidy has been reported in 10% to 20% of children with this prenatal ultrasound finding. Major organ
system abnormalities are reported to occur in up to 60%
of such fetuses. Notably, when absence of the corpus callosum is an isolated fetal ultrasound finding, the reported
rate of a relatively normal developmental outcome ranges
from 50% to 75%. Well-conducted, long-term studies are
lacking. Postnatal follow-up for infants with a history of
a finding of agenesis of the corpus callosum on prenatal
ultrasound examination should include, at a minimum,
close long-term clinical assessment (Chadie et al, 2008;
Fratelli et al, 2007; Woodward, 2008).
Mild ventriculomegaly is a relatively uncommon fetal
ultrasound finding that may be a soft marker for aneuploidy, fetal infection, or other central nervous system
abnormalities. As such, it is recommended that serial
imaging studies be undertaken, in some cases including a
more extensive workup. In the presence of a benign fetal
assessment, most infants appear to do reasonably well
after delivery. Close pediatric developmental followup and
serial imaging studies are important to consider (Leitner et
al, 2009; Melchiorre et al, 2009; Sohaey and Filipek, 2008).
Cardiac Findings
Echogenic cardiac focus is an incidental ultrasound finding in 3% to 4% of healthy fetuses. Notably, there is
an increased incidence of twofold to threefold in Asian
populations. It is said to be a soft marker for chromosomal abnormalities when associated with other screening
abnormalities. If the results of a physical examination of
a newborn are normal and there are no other ultrasound
findings, no further evaluation is needed (Borgida et
al, 2005; Koklanaris et al, 2005; Ouzounian et al, 2007;
Sohaey, 2008b).
Gastrointestinal Findings
Echogenic bowel, when noted to be present during a second-trimester ultrasound examination and determined to
be grade 0 or 1 (i.e., less echogenic than bone) is a normal
variant. Density greater than that of bone (grade 2 to 3)
is abnormal and is potentially a marker for cystic fibrosis,
trisomy 21, gastrointestinal anomalies, or in utero infection. Isolated echogenic findings are considered benign
with a good prognosis and require no special workup or
postnatal management (Al-Kouatly et al, 2001; Patel et al,
2004; Sohaey, 2008a).
Cholelithiasis is an uncommon third-trimester fetal
ultrasound finding that needs to be differentiated from
hepatic calcification. Cholelithiasis is a benign condition
requiring no special evaluation or treatment. An imaging
examination at 1 year of age for a child with this prenatal
finding may be helpful in documenting expected resolution (Agnifili et al, 1999; Sohaey, 2008b).
303
Hepatic calcifications are uncommon fetal ultrasound
findings; they are often isolated, single and, in a low-risk
pregnant patient, of no significance. When numerous,
hepatic calcifications may be markers for fetal aneuploidy, infection, meconium peritonitis, hepatic tumor,
or vascular insult. Twenty percent are associated with
some form of fetal pathology. Neonatal management
depends on the prenatal workup and the clinical presentation in the newborn period (Oh, 2008; Simchen et al,
2002).
Urinary Tract Findings
Mild fetal pelviectasis is one of the more common abnormalities detected by second-trimester ultrasound examination, with a reported incidence of 0.5% to 5% in unselected
pregnant populations. Diagnostic criteria vary, but generally include a second-trimester renal pelvis diameter of
4 to 10 mm and 7 to 10 mm during the third trimester.
Some authors consider mild fetal pelviectasis to be a soft
marker for trisomy 21. When mild fetal pelviectasis is an
isolated finding, the prognosis is good and the condition
usually resolves either in utero or during early childhood.
In a meta-analysis, it was reported that 11% of children
with a history of mild fetal pelviectasis demonstrated postnatal pathology. Authors of a prospective cohort followup study reported uropathy in 18% of the patients. There
is a lack of consensus as to postnatal management, with
some authorities recommending close clinical assessment
and others recommending follow-up renal ultrasound
examinations at approximately 1 week and 1 month of life
(Coelho et al, 2007; Lee et al, 2006; Sohaey and Arnold,
2008).
CAR SEAT TRIALS
The observation that preterm infants had episodes of
hypoxia when monitored in car seats led the AAP to recommend in 1991 that preterm infants should be observed
and monitored for apnea, bradycardia, or oxygen desaturation in their car safety seat before hospital discharge—the
so-called car seat challenge (AAP Committee on Injury
and Poison Prevention and Committee on Fetus and Newborn, 1991, 1996; Bull et al, 1999). In the United States,
the car seat challenge expanded to include late preterm
infants, most of whom did not have respiratory problems
during their newborn hospital stay. It has been reported
that 25% of late preterm infants do not fit securely into
standard car safety seats, and 12% of healthy late preterm
infants have apneic or bradycardic events in their car seats
(Merchant et al, 2001).
The authors of a Cochrane Review questioned whether
car seat trials actually prevent morbidity or mortality, and
whether there are adverse effects of not passing this test,
such as prolonging the hospital stay or creating parental
anxiety. Their review did not disclose any randomized trials, and they concluded that “it is unclear whether undertaking a car seat challenge is beneficial or indeed whether
it causes harm” (Pilley and McGuire, 2006). Since then,
there has been one randomized trial in healthy term
infants comparing car seats to car beds, and no differences were found in rates of oxygen desaturation or apneic
304
PART VII Care of the Healthy Newborn
events (Kinane et al, 2006). Further study is needed to
determine whether car seat trials in late preterm neonates
are warranted.
ROUTINE AND COMMON MEDICAL
TREATMENTS
PREVENTION OF OPHTHALMIA
NEONATORUM AND CONJUNCTIVITIS
Approximately15% to 20% of babies will develop conjunctivitis in the first few weeks of life. Conjunctivitis can
be caused by a sexually transmitted bacteria, normal skin
or nasopharyngeal flora, or chemical irritation (Krohn
et al, 1993). In addition, eye discharge can be caused by
obstruction of the nasolacrimal duct rather than from the
conjunctivitis. The most worrisome infection is that from
Neisseria gonorrhea, which can be invasive to the cornea in
a matter of hours and lead to blindness. Despite effective
preventive measures known since the 1880s, there are still
thousands of children blinded by this infection worldwide
each year.
Most states in the United States have laws or regulations
that require administration of topical antibiotic ointment
to the conjunctivae of babies within a few hours of birth.
This practice has been effective in reducing the cases of
blindness caused by gonococcal conjunctivitis. It is moderately effective in preventing conjunctivitis caused by
chlamydia.
Parents may question the need to expose all babies to
eye medication, especially if the mother has been tested
and found to be without gonorrhea or chlamydia. Some
countries have stopped routine administration of eye
prophylaxis. In those countries, an increase in infection,
primarily caused by chlamydia, has been noted. With
informed consent, parents can opt out of eye prophylaxis
for their newborn.
There is lay literature recommending the instillation
of colostrum or breast milk into the eyes of babies to
prevent or treat conjunctivitis. Although colostrum has
antimicrobial action, its efficacy has not been adequately
studied.
Povidone-iodine solution has been shown to be more
effective and cause less irritation than erythromycin ointment. It is also less expensive, but is not yet approved
for this use by the U.S. Food and Drug Administration
(Isenberg et al, 1995). Whether instillation of povidoneiodine in the eyes may affect newborn thyroid screens—as
has been reported with use of this solution on umbilical
cord stumps—is not clear (Lin et al, 1994).
VITAMIN K
Vitamin K is necessary for biologic activation of several
human proteins, most notably coagulation factors II,
VII, IX, and X. Because placental transfer is limited, cord
blood levels of vitamin K1 (phylloquinone) are 30-fold
lower than maternal levels. Intestinal bacteria synthesize
menaquinone (vitamin K2), which has 60% of the activity of phylloquinone. However, neonates have a decreased
number of bacteria in their gut that manufacture vitamin
K2; levels of this form of vitamin K are not found in the
livers of infants until they are 2 to 3 months old. Therefore newborn infants are deficient in vitamin K at birth and
are at risk for significant bleeding. Fortunately intramuscular vitamin K rapidly activates clotting factors, greatly
decreasing this risk.
Three presentations of vitamin K–deficient bleeding
(VKDB) have been described. Early VKDB manifests in
the first 24 hours after birth. It is not prevented by postnatal administration of vitamin K, and it usually occurs in
infants born to mothers who are taking medications that
inhibit vitamin K. Common medications that inhibit vitamin K include many anticonvulsants, isoniazid, rifampin,
warfarin, and some antibiotics such as cephalosporins.
Early VKDB is frequently serious because of intracranial
and intraabdominal hemorrhage. It is estimated that in
neonates at risk for early VKDB, the incidence is as high
as 12% (Van Winckel et al, 2009). Classic VKDB occurs
in infants during the first week of life. Although the clinical presentation is often mild, blood loss can be significant, and intracranial hemorrhages have been reported.
Although estimates vary, the incidence of classic VKDB in
the absence of vitamin K supplementation is likely 0.25%
to 1.7% (AAP Committee on Fetus and Newborn, 2003).
Late VKDB occurs in infants between the ages of 2 and 12
weeks and is usually severe. The mortality rate from late
VKDB is approximately 20%, and 50% of children with
this disorder have intracranial hemorrhages. Late VKDB is
associated with exclusive breastfeeding. Human milk contains only 1 to 4 μg/L of vitamin K, whereas commercially
available formula contains 50 μg/L or greater. In neonates
who do not receive supplemental vitamin K, the incidence
of late VKDB is estimated at 4.4 to 7.2 in 100,000 (or 1 in
15,000 to 1 in 20,000; Van Winckel et al, 2009). Vitamin
K administered shortly after birth is effective in preventing
classic and late VKDB. Since 1961 the recommended dose
of vitamin K for term infants born in the United States
has been 1 mg given intramuscularly. However, the results
of a study suggesting an association between intramuscular vitamin K given at birth and childhood cancer created
controversy regarding this practice (Golding et al, 1990,
1992). The results of subsequent studies have indicated
conclusively that there is no increased risk for solid tumors
in children given intramuscular vitamin K; however, the
possibility that there is a slightly increased risk for leukemia cannot be excluded (Puckett and Offringa, 2000).
Because of concerns regarding an increased risk of childhood cancers, a switch to oral vitamin K occurred in some
countries, but not in the United States. It is apparent that
a single oral dose of vitamin K has an efficacy similar to an
intramuscular dose in preventing classic VKDB, but offers
less protection against late VKDB. Repeated doses of an
oral vitamin K preparation until an infant is 8 to 12 weeks
old increases the efficacy of this route of administration.
However, it is not clear that even multiple doses of an oral
formulation of vitamin K are as effective as a single intramuscular dose, given at birth. In a multinational review,
rates of late VKDB in infants receiving various regimens
of oral vitamin K were mostly in the range of 1.2 to 1.8
in 100,000 compared with 0 cases in 325,000 children
receiving an intramuscular dose (Cornelissen et al, 1997).
The oral regimens assessed included a dose at birth of
1 mg. Reported rates of VKDB in infants who received
CHAPTER 26 Routine Newborn Care
2 mg orally at birth, with doses repeated subsequently, are
lower but still somewhat higher than in neonates treated
with intramuscular vitamin K (Busfield et al, 2007; Von
Kries et al, 2003). Early data from the Netherlands, where
infants received 1 mg orally at birth and 25 mcg daily for
up to 12 weeks, suggested that this regimen was as efficacious as an intramuscular dose (Cornelissen et al, 1997).
However, in a subsequent study from the Netherlands, the
rate of late VKDB was 3.2 in 100,000 in a group of infants
with undiagnosed biliary atresia who had been treated with
this dosing schedule (van Hasselt et al, 2008). Unrecognized cholestatic liver disease is a significant risk factor for
VKDB. Finally, no cases of late VKDB were found among
396,000 Danish infants who received an oral dose of 2 mg
of vitamin K at birth and 1 mg weekly until the age of 3
months (Hansen et al, 2003).
Risks from intramuscular vitamin K include pain at
the injection site and the possibility of a serious medication error. The risks of a significant complication from
the injection are probably negligible; in one study, zero
significant complications were reported after 420,000
injections (Von Kries, 1992). In the United States, oral
administration is complicated by the lack of an oral vitamin K preparation being licensed for newborns. In some
settings, infants have received the intramuscular preparation orally; however, tolerance may be a problem, and the
efficacy of this preparation when given orally might not
be comparable to the oral formulations used in Europe.
In addition, compliance with repeated doses of oral vitamin K in infants may be suboptimal. Finally, it is unknown
whether the use of repeated administration of oral vitamin
K in the dose range of 1 to 2 mg each week is associated
with an increased risk of childhood cancers.
For parents who have questions regarding the best
method to prevent classic and late VKDB, the clinician is
advised to discuss the pros and cons of intramuscular versus oral vitamin K. If the parents choose oral administration, a dose of 2 mg of vitamin K should be given shortly
after birth, with subsequent doses until the infant is at least
4 weeks old if he or she is breastfed. In a policy statement,
the AAP suggests that, if an oral vitamin K formulation
becomes licensed for use in the United States, a dose of
2 mg can be given at birth and repeated at 1 to 2 weeks
of age and at 4 weeks of age for neonates whose parents
decline intramuscular vitamin K (AAP Vitamin K Ad Hoc
Task Force, 1993).
CIRCUMCISION
Neonatal circumcision is a polarizing issue for health
care professionals and parents. Those who favor routine
circumcision highlight health benefits such as decreased
urinary tract infections (UTIs), reduced risk of penile cancer, and possibly lower rates of sexually transmitted diseases, including HIV (Schoen, 2008). Those who oppose
the procedure indicate that the number of circumcisions
needed to be performed to prevent one of these outcomes
(i.e., the number needed to treat [NNT]) is large, the risks
of the procedure balance out these benefits, circumcision
leads to loss of sexual sensation, and subjecting a neonate to a painful procedure without clear benefits may be
unethical (Andres, 2008).
305
It is clear that circumcision reduces the risk of UTI by
approximately 10-fold; however, given the low incidence
of UTI in male infants, 100 boys need to be circumcised
to prevent one UTI. Similarly, although circumcision has
been shown to prevent penile cancer, it is an extremely
rare condition and the NNT is about 900 (Christakis et
al, 2000). There has been recent interest in circumcision
as a method for preventing HIV. The results of studies in
three African countries indicate that circumcision reduces
the risk of HIV infection by 56%. Because the incidence
of HIV in these countries is high, the calculated NNT is
approximately 72 (Mills et al, 2008). Given the same efficacy, the NNT to prevent one case of HIV infection in
Canada, where the incidence is 13 in 100,000, or in the
United States, where the incidence is 23 in 100,000, is
greater than 5000 (Andres, 2008; Hall et al, 2008).
Circumcision is generally a safe procedure. Although
some increased bleeding is reported after 1% of circumcisions, the rate of significant complications is approximately 0.2% (Christakis et al, 2000; Gee and Ansell,
1976; Wiswell and Geschke, 1989). Bleeding, sometimes
requiring suturing of a vessel, is the most common significant complication, followed by penile injury and infection. Infection is more common after a circumcision using
a Plastibell rather than a Gomco clamp; hemorrhage is
reportedly similar after either technique (Gee and Ansell,
1976).
Circumcision is an uncomfortable experience for the
neonate. Small amounts of sucrose solutions can be offered
to the baby for soothing. Pain from the actual surgery can
be significantly decreased with the use of a dorsal penile
nerve block or ring block. In one study, 65% of infants
who had received a dorsal nerve block had no or minimal
response to the initial clamping of the foreskin (Taeusch
et al, 2002).
A poor cosmetic outcome can be caused by removal
of too little foreskin. It has been estimated that 1-9.5%
of circumcisions are redone because of parental concern
regarding the appearance. In a prospective study, among
boys younger than 3 years who had been circumcised using
either a Plastibell or with a Mogen clamp, the glans was
fully exposed in only 35.6%. However, in older circumcised males the glans was fully exposed in more than 90%
(Van Howe, 1997). This finding suggests that parents of
a circumcised infant should be counseled that the vast
majority of properly done circumcisions will lead to an
acceptable cosmetic appearance over time.
In the United States, the Gomco clamp is the most
commonly used apparatus for performing circumcisions,
followed by the Plastibell and Mogen clamp (Stang and
Snellman, 1998). The use of the Mogen clamp leads to
shorter procedures and, reportedly, less pain and bleeding than the other techniques (Kurtis et al, 1999; Taeusch
et al, 2002). However, less foreskin is removed with the
Mogen clamp than with the other two techniques (Alanis
and Lucidi, 2004).
HEPATITIS B VACCINE
The implementation of routine hepatitis B immunization during infancy has been associated with a dramatic
decrease in the incidence of this infection. Between 1990
306
PART VII Care of the Healthy Newborn
(before routine vaccination of infants) and 2004 the overall
incidence of acute hepatitis B in the United States declined
by 75%, and by 94% among children and adolescents
(Centers for Disease Control and Prevention, 2005). The
Centers for Disease Control and Prevention and the AAP
recommend that the initial dose of the three-dose hepatitis
B immunization series be given in the newborn nursery;
however, this recommendation is far from being followed
universally. In 1999, it was estimated that 54% of newborns in the United States received a dose of hepatitis B
vaccine at birth. However, at that time a recommendation
was made to suspend birth dosing until vaccines were made
that did not contain thimerosal (a preservative containing
mercury). Even after a thimerosal-free hepatitis B vaccine was produced, many newborn nurseries in the United
States did not immediately resume newborn immunization programs. This issue, combined with the reluctance
of some parents to have their newborns immunized, has
led to continued suboptimal rates of administration of the
birth dose of hepatitis B vaccine (Clark et al, 2001). Among
infants born in the United States between 2003 and 2005,
it was estimated that 50.1% received a dose of the vaccine
by the age of 3 days (Centers for Disease Control and Prevention, 2008).
There are at least two advantages of providing the first
dose of hepatitis B vaccine during the newborn nursery
stay. First, newborns who receive a dose at birth are more
likely to complete their hepatitis B immunization series
on time than those who receive a first dose later (Yusuf
et al, 2000). Second, because a dose of hepatitis B vaccine
given within 12 hours of birth can prevent vertical transmission of hepatitis B infections in 75% to 90% of cases,
early provision of immunization serves as a safety net in
cases where there has been an error in identifying a mother
who is positive for hepatitis B surface antigen (Centers for
Disease Control and Prevention, 2005).
The main disadvantage of providing a dose of hepatitis
B vaccine during the nursery stay is that it can complicate documentation of hepatitis B immunization status in
a child by increasing the number of vaccination providers.
There is no evidence that administration of a dose of hepatitis B vaccine at birth leads to more evaluations for sepsis
because of adverse events related to the immunization.
ONGOING CARE
UMBILICAL CORD CARE AND SINGLE
UMBILICAL ARTERY
Umbilical cord care recommendations vary from “dry cord
care” to the use of dyes and cleansing with alcohol, soap
and water, or antiseptics. Concern over the possible toxic
effects of dye and antiseptics led many hospitals in the
United States to adopt the dry cord care method of cord
care. Unfortunately this method may be responsible for
causing an increase in the risk for omphalitis (Janssen et
al, 2003; Simon and Simon, 2004). In addition, the results
of a randomized trial in Nepal indicate that cord care
using topical chlorhexidine reduces the risk of developing
omphalitis; however, that population may be at higher risk
compared with those born in the United States (Mullany
et al, 2006).
Because omphalitis is rare, and its more severe complication necrotizing fasciitis is even rarer, large trials are
needed to determine which cord care regimen is best for
preventing these complications. Until such trials are conducted, there is no clear advantage of one regimen over
another. Providers caring for newborns need to keep these
diagnoses in mind and encourage parents to report redness
around the umbilical cord stump.
A single umbilical artery is detected in about 4 out of
1000 births. There is an association of the single umbilical
artery with a number of congenital anomalies, including
renal or genitourinary malformations, cardiac malformations, and chromosomal anomalies such as Down syndrome. In this era of near universal use of prenatal fetal
ultrasonography, any associated anomalies are usually
discovered (Deshpande et al, 2009; Johnson and Tennenbaum, 2003). Unless there are abnormalities noted on
physical examination, there is no need to repeat diagnostic
ultrasound examinations after birth in a child with a single
umbilical artery.
BREASTFEEDING
There is voluminous evidence that the optimal feeding for
normal neonates is human milk provided via the mother’s
breast. From this most preferred feeding there is a continuum to least preferred feeding that includes: maternal
milk given by an artificial method, donor human milk
(although this is rarely used by most health care facilities),
and commercially available infant formula. It is incumbent
on health care professionals and health care systems to
vigorously promote the optimal feeding for normal newborns. This promotion includes education to prospective
mothers, knowledge of the characteristics of human milk
and the normal course of lactation, organization of health
care facilities to optimize the initiation of breastfeeding
after delivery, early recognition of suboptimal breastfeeding and interventions to correct problems, and adoption of
a parsimonious list of contraindications to breastfeeding.
Nutritional Composition of Human Milk
Although the composition varies among mothers, by age of
the child, and even within a feeding, the nutritional components of mature human milk can be summarized (Picciano,
2001). The caloric content of human milk is approximately
0.67 Kcal/mL (20 Kcal/ounce). Approximately 50% of
these calories are provided by the lipid components.
Importantly, human milk contains omega-3 fatty acids.
Lactose is the principal carbohydrate, providing approximately 40% of the calories. The protein in human milk
is divided into two categories, casein and whey, that are
provided in a 40:60 ratio. The sodium content of human
milk is low, averaging approximately 7 mEq/L. With the
exception of vitamins D and K, the amounts of minerals
and micronutrients available are all adequate for optimal
infant growth. The evolutionary advantage afforded by the
relatively low concentrations of vitamin D and vitamin K
in human milk is unclear.
Healthy newborns receive surprisingly little breast milk
in the first few days of life. Average intake of colostrum
during the first day of life is approximately 10 mL/kg and
CHAPTER 26 Routine Newborn Care
20 mL/kg during the second 24 hours of life (Casey et al,
1986; Evans et al, 2003). Maternal milk production dramatically increases during the period from 36 to 96 hours
of life; this increase in quantity is accompanied by a change
from colostrum to mature milk (Casey et al, 1986; Saint et
al, 1984). The increase in milk production is perceived by
the mother as breast fullness. In one study, mothers noted
that their breasts were noticeably fuller when their infants
were an average of 53 hours old. This finding was closely
correlated with the mean age (58 hours) when infants
transferred greater than 15 mL of milk from the breast
(Dewey et al, 2003).
Given the low volume of milk provided initially, neonates have a decrease in weight and an increase in serum
sodium during the first few days of life (Marchini and
Stock, 1997). Average maximal weight loss in breastfed
infants is 5% to 7% of birthweight and occurs between
48 and 72 hours of life (Macdonald et al, 2003; Marchini
and Stock, 1997; Rodriguezet al, 2000). With the onset of
copious production of mature milk, neonates begin to gain
weight and their serum sodium levels fall (Marchini and
Stock, 1997). Infants fed human milk regain their birthweight, on average, by the age of 8.3 days; 97.5% have
regained their birthweight by 21 days of age (Macdonald
et al, 2003).
Health Benefits of Human Milk
There is copious research on numerous health benefits for
infants who are breastfed. The list of benefits that have been
found include: decreased incidence of conditions such as
gastrointestinal infections, lower respiratory tract disease,
otitis media, hypertension, obesity, diabetes, allergies, and
asthma; improved cognitive development; and reduced
risk of sudden infant death syndrome (Hoddinott et al,
2008). Unfortunately, because it is unethical to conduct
individual randomized controlled trials on breastfeeding,
most of these benefits have been documented in observational studies. Such studies are subject to numerous problems that might bias the results toward the null hypothesis
or an overestimation of the benefits (Kramer et al, 2009).
Perhaps the biggest problem with observational studies on
breastfeeding is that there is significant confounding by
socioeconomic status, for which it is difficult to adequately
account in analyses.
The best evidence of the health benefits of breastfeeding comes from a large trial conducted in Belarus. Health
care professionals at participating hospitals were randomly
assigned to receive training on promoting breastfeeding
using the Baby-Friendly Hospital Initiative (BFHI) developed by the World Health Organization and the United
Nations Children’s Fund (BFHI is further described in a
following section) or to continue to provide standard care.
A total of 17,046 mother-infant dyads were enrolled in the
study (Kramer et al, 2001). All participating infants were
born at 37 weeks’ gestation or later and weighed more
than 2500 g; all the mothers intended to breastfeed their
infants. The rate of any breastfeeding at 3 months old was
significantly higher among infants born in BFHI hospitals
than at control sites (43% and 6%, respectively). Rates of
breastfeeding were also significantly higher at BFHI locations at 6 and 12 months of age. Most pertinent, rates of
307
BOX 26-2 B
aby-Friendly Hospital Initiative:
Ten Steps to Successful
Breastfeeding
1. Maintain a written breastfeeding policy that is routinely communicated to
all health care staff.
2. Train all health care staff in the skills necessary to implement this
policy.
3. Inform all pregnant women about the benefits and management of
breastfeeding.
4. Help mothers initiate breastfeeding within 1 hour of birth.
5. Show mothers how to breastfeed and how to maintain lactation, even if
they are separated from their newborns.
6. Give infants no food or drink other than breast milk unless medically
indicated.
7. Practice “rooming-in”—allow mothers and infants to remain together 24
hours a day.
8. Encourage unrestricted breastfeeding (i.e., on demand).
9. Give no pacifiers or artificial nipples to breastfeeding infants.
10. Foster the establishment of breastfeeding support groups and refer mothers to them on discharge from the hospital or clinic.
gastrointestinal infection and atopic dermatitis were significantly lower in babies who had been born at BFHI hospitals. These data provide strong evidence for a significant
beneficial effect of breastfeeding.
Among children enrolled in the original study who were
assessed at 6.5 years of age, the verbal intelligence quotient
of those born at BFHI hospitals was 7.5 points higher than
those born at control sites (Kramer et al, 2008). Whether
this finding was related to the components in the breast
milk or from the physical act of breastfeeding is unclear;
regardless of the mechanism, the effect is profound. In
other follow-up studies, no difference between the two
groups of children were found in the rates of asthma,
allergy, or obesity (Kramer et al, 2007); however, the
design of the study provides a conservative estimate of the
effects of breastfeeding.
Promotion of Breastfeeding
Given the demonstrable improvements in outcomes,
health care providers should promote breastfeeding as the
preferred method of feeding newborn infants and facilitate
its initiation during the newborn nursery stay. As a comprehensive program, implementation of the 10 steps of the
BFHI (Box 26-2) has been shown to significantly increase
the rates of breastfeeding (Kramer et al, 2001). There is
evidence to support the efficacy of each of the 10 separate
steps, although for some of the steps, such as excluding
pacifiers, the evidence is contradictory (Cramton et al,
2009; O’Connor et al, 2009).
From a practical standpoint there are several evidencebased interventions during the newborn nursery stay
that increase the rate or prolongation of breastfeeding.
These interventions include the use of frequent demand
feedings as opposed to a rigid feeding schedule, early
skin-to-skin contact between mother and infant, professional advice on breastfeeding techniques, and exclusion
of commercial formula from discharge packs (Anderson
et al, 2003; Britton et al, 2007; Donnelly et al, 2000;
Renfrew et al, 2000).
308
PART VII Care of the Healthy Newborn
Breastfeeding Problems
The vast majority of difficulties with breastfeeding are
related to a delay in transfer of adequate quantities of
human milk to the infant. Delayed lactogenesis occurs in
20% to 30% of mothers; however, in most instances the
primary problem is related to infant breastfeeding behaviors in the first few days of life. Although the clinical correlate is not well defined, weight loss of greater than 10%
of birthweight is considered excessive in breastfed infants.
Studies on breastfed neonates indicate that approximately
10% of infants lose more than 10% of their birthweight
during the first few days of life (Dewey et al, 2003).
It is important to identify mother-infant dyads who are at
risk for breastfeeding problems, so that early interventions
can prevent excessive weight loss. Mothers with previous
breast surgery, particularly breast reduction, are at increased
risk of primary insufficient lactation. Flat or inverted nipples
can also make breastfeeding more difficult. Prolonged labor
and cesarean delivery have been associated with delayed
onset of milk production (Dewey et al, 2003).
The adequacy of breastfeeding behaviors can be
assessed during the newborn nursery stay using scoring
systems such as the Infant Breastfeeding Assessment Tool.
Low scores on this measure during the first day of life are
moderately predictive of excessive weight loss in the neonate (Dewey et al, 2003). Decreased numbers of voids and
stools in the newborn are also helpful in identifying children with breastfeeding problems, but this information is
most useful after day 3 of life.
Supplementation of Breastfeeding
It is usually unnecessary to provide any nutrition or fluid to
full-term breastfed infants beyond human milk. Dextrose
water or commercial formula may be needed in neonates
with hypoglycemia who are not responsive to breastfeeding.
Supplementation may also be indicated in newborns who
have lost more than 10% of birthweight or have decreased
urine and stool output. Supplementation should be considered a temporary intervention, and its provision should not
interfere with the onset of successful breastfeeding.
Contraindications to Breastfeeding
The few absolute contraindications to breastfeeding
include maternal HIV infection, untreated tuberculosis in
the mother, evidence of current cocaine use or antimetabolite drugs in the mother, and galactosemia in the neonate
(Gartner et al, 2005). There are a myriad of other drugs for
which there is concern regarding long-term neurodevelopmental outcomes in the infant. Selective serotonin reuptake inhibitors are commonly used to treat depression and
anxiety in young women. Among drugs in this category,
sertraline and paroxetine are thought to be the safest for
use in breastfeeding mothers, whereas fluoxetine and citalopram are believed to have the most potential for toxicity
in the neonate (Field, 2008). Overall there have been few
adverse effects noted with use of any of these drugs, and
generally the potential risks associated with these medications are thought to be outweighed by the benefits of
breastfeeding (Field, 2008). Similarly, although methadone is detectable in the breast milk of women receiving
this medication, serum levels in neonates are low and
unlikely to cause a significant effect (Jansson et al, 2008).
Hepatitis C virus RNA has been found in the milk of
mothers infected with this virus. Despite this finding, transmission of infection via breastfeeding has not been documented. Maternal hepatitis C infection is not considered
a contraindication to breastfeeding (Gartner et al, 2005).
BOTTLE FEEDING
Commercial formula that provides adequate nutrition,
vitamins, and minerals is available for infants of mothers who do not wish to breastfeed their infants or when
breastfeeding is contraindicated or impossible. There are
three major categories of formula used in neonates: cow
milk–based, soy, and hydrolyzed formula. Of these, cow
milk–based formula is used most commonly. The main
carbohydrate in cow milk–based formula is lactose. Soy
formulas were developed for infants with a possible cow’s
milk allergy. Because the main carbohydrate in soy formulas is sucrose or corn syrup, soy formula can be used
in neonates with possible galactosemia. Protein hydrolysate formulas were initially developed for use in infants
who are highly intolerant to cow’s milk protein (Kleinman, 2009). These formulas are purported to lead to fewer
allergies in babies and children than does cow milk–based
formula, but the evidence for this is limited (Osborn and
Sinn, 2006). All extensively hydrolyzed formulas are lactose free (Kleinman, 2009). These formulas are indicated
in infants with definitive evidence of cow’s milk protein
allergy because 10% to 14% of them also have a soy allergy
(Bhatia and Greer, 2008).
Standard preparations of formulas available for use in
healthy term neonates provide 0.67 Kcal/mL. Most formulas are fortified with 10 to 12 mg/L of iron; however,
some low-iron cow milk–based formulas are available. It is
recommended that all formula-fed newborns receive the
iron-fortified products. Vitamin D at the concentration of
approximately 400 IU/L is provided in all the commercially available formulas (Kleinman, 2009).
Mothers who elect to bottle-feed report feeling unsupported for their decision by health care professionals, and
up to 50% feel pressured to breastfeed (Lakshman et al,
2009). Although the benefits of breastfeeding should be
provided to mothers who have not decided how to feed
their babies, the role of health care providers is to support the decision of those who have elected to bottle-feed.
It is also important to provide practical education about
bottle-feeding to these parents; this is frequently neglected
in many newborn nurseries (Lakshman et al, 2009).
Newborns who are bottle-fed can feed ad libitum beginning shortly after birth. Average formula intake in term
newborns during the first day of life is 15 to 20 mL/kg and
40 to 45 mL/kg during the second day. During the nursery stay, neonates who are formula fed typically lose less
weight than breastfed infants (Dollberg et al, 2001).
ANTICIPATORY GUIDANCE
A primary duty of providers of newborn care is to ensure
that parents of new infants have the knowledge and skills
to provide for normal growth and development. Parents
CHAPTER 26 Routine Newborn Care
who are taught about normal newborn development and
behavior have more realistic expectations about the work
involved and look upon their child with more fondness.
Conversely, before discharge from the newborn nursery,
it is important to assess the parents’ ability to provide a
safe and nurturing environment for the neonate. Parents
showing concerning behaviors, possibly leading to abuse
or neglect, should have supervision and interventions
to help them, possibly leading to termination of parental rights (Davidson-Arad et al, 2003; Wattenberg et al,
2001).
There are several major challenges for parents of normal newborns: sleep deprivation, learning to calm a crying infant, significant life changes, and the new worries
that come with being responsible for a totally dependent
being. Postpartum depression is more common and of
longer duration than previously thought, and it occurs
in at least 10% of mothers. This condition is related to
sleep deprivation and has major and long-lasting effects
on infant homeostasis and development (Chaudron,
2003).
Anticipatory guidance should be given to help prepare
new parents for the common tasks of newborn care and to
educate them about the many normal variations in newborn behavior. Learning how to soothe a baby is one of
the first needed parenting tasks. Providers can help by giving suggestions to reduce crying and to better cope with
infants who are more sensitive and harder to soothe (Barr
et al, 2009).
Most parents have questions about feeding, elimination, bathing, cord care, genital care, jaundice, and common rashes. There are numerous checklists of educational
topics that can be overwhelming to new parents. In addition, learning styles can vary, with some preferring written materials and while others preferring audio-visual
materials or hands-on demonstration. Ideally, education
should be targeted toward the topics of interest and with
the appropriate materials for learning style (Dusing et al,
2008).
Mothers are often not in a good learning state in the
immediate postpartum period because of pain, postpartum hormonal changes, and the stress of being in a hospital. However, there is heightened receptivity to change
during this period, so attempts to teach or make lifestyle
changes (e.g., smoking cessation) may be more effective.
There is some evidence that providing parental education
using tools such as interactive video and computers may
be superior to traditional teaching (Snowdon et al, 2009;
Trepka et al, 2008).
Given the obstacles to providing meaningful education
during the nursery stay, it is probably better for practitioners to focus on a few key points of anticipatory guidance
rather than reciting a litany of instructions. There is also
a philosophical choice in deciding whether to emphasize
the overall health of a newborn or to concentrate on prevention or identification of illness. There is little evidence
for the efficacy of most anticipatory guidance provided to
parents during the newborn nursery stay. A notable exception is the advice to put infants to sleep in the supine position (described in the following section). There is also
emerging evidence that education about the normality of
inconsolable crying in infants helps parents cope with this
309
stressful situation, and it could reduce the risk of shaken
baby syndrome (Barr et al, 2009).
Sleep Position
With the exception of immunizations, no child health
intervention in the past two decades has resulted in a
larger decrease in postneonatal infant mortality than
the “Back to Sleep” campaign. The remarkable change
in the predominant sleep position of infants from prone
to supine has led to a 30% to 50% reduction in the rate
of sudden infant death syndrome (SIDS) in the United
States (AAP Task Force on Sudden Infant Death Syndrome, 2005). A multipronged effort including brochures,
public service announcements, and education provided by
health care professionals was used to affect the change in
sleep position (Willinger et al, 2000). Obviously education provided to parents during the newborn nursery stay
is a crucial determinant of the sleep position of an infant.
In addition to providing education, there is evidence that
parents model sleep position for their babies after how
they saw nurses and physicians place their neonate in the
bassinet in the newborn nursery (AAP Task Force on
Sudden Infant Death Syndrome, 2005; Colson and Joslin,
2002). Therefore it is crucial that neonates are placed on
their backs to sleep in the newborn nursery. In addition,
there is an additive effect of both physicians and nurses
recommending the supine sleep position (Willinger et al,
2000).
In addition to supine position, there are other factors
related to the sleep environment that can effect the risk of
SIDS in a newborn infant. It is recommended that infants
sleep on firm surfaces and without excessive bedding such as
pillows. Many experts also recommend against co-sleeping
between parents and infants; however, this topic is controversial and the evidence is somewhat contradictory. Similarly, although use of a pacifier has been found to reduce
the risk of SIDS, there is a reluctance to recommend these
devices because of concerns about reducing breastfeeding (AAP Task Force on Sudden Infant Death Syndrome,
2005; Willinger et al, 2000).
DISCHARGE AND FOLLOW-UP
For infants born in the United States at 35 weeks’ gestation or later, the average length of the initial hospital stay
is 48 to 52 hours (Datar and Sood, 2006; Kuzniewicz et
al, 2009; Paul et al, 2009). Because approximately 50% of
infants born by vaginal delivery are discharged before the
age of 48 hours, and because up to 40% of those born by
cesarean delivery are discharged before 72 hours of age,
a large proportion of neonates are discharged before the
age of 3 to 4 days, when bilirubin levels typically peak and
breastfeeding is well established (Paul et al, 2006). It is recommended that infants discharged before 48 hours have
a follow-up appointment with a provider within 48 hours
(AAP Committee on Fetus and Newborn, 2004). This
follow-up can be accomplished either by a visit to a health
care provider or via a home nursing visit.
Risk factors for readmission after an initial hospital
stay of less than 48 to 72 hours include gestational age
less than 39 weeks (and especially less than 37 weeks),
310
PART VII Care of the Healthy Newborn
primiparous mother, and Asian race (presumably because
of an increased risk of hyperbilirubinemia) (Burgos et al,
2008; Grupp-Phelan et al, 1999; Liu et al, 1997; Paul et
al, 2006). Consideration of a longer nursery stay is suggested for infants with one or more of these risk factors.
In addition, early discharge is not recommended for term
newborns who have not voided, passed at least one stool,
or demonstrated adequate breastfeeding (AAP Committee
on Fetus and Newborn, 2004). However, there is little evidence to support these recommendations.
COMMON PROBLEMS DURING
THE NURSERY STAY
HYPOTHERMIA AND HYPERTHERMIA
Upon leaving the womb, a newborn is immediately
challenged with maintaining a normal body temperature. If a neonate is not quickly dried at birth, he or
she may lose up to 1° C body temperature per minute.
Healthy term babies are able to increase heat production through glycogenolysis and nonshivering thermogenesis for minutes to a few hours, depending on
environmental conditions (Aylott, 2006). Babies typically have a decline in body temperature during the first
hour of life with a gradual increase during the following 12 hours (Li et al, 2004). By the second day of life,
the infant’s body temperature becomes more stable, but
heat loss can occur again with bathing or other stresses
(Takayama et al, 2000).
Being too cold or too hot causes metabolic stress to the
newborn, so efforts to maintain a steady and neutral thermal environment should be provided. The best practice is
to dry the baby immediately after delivery and place the
infant skin-to-skin with the mother. Although the AAP
and the American College of Obstetricians and Gynecologists jointly recommend keeping infants’ core temperatures within the narrow range of 36.5° to 37° C, in one
study of healthy term newborns the average temperature
was 36.5° C, with a normal range from 36.0° to 37.9° C
(Takayama et al, 2000). Thin babies tend to have lower
body temperatures, and heavier babies tend to have higher
body temperatures. Hypothermia should be managed by
placing the baby skin-to-skin with a parent or under a radiant warmer.
Standard practice at most nurseries is to measure axillary
temperatures, probably because of reports in the 1960s
and 1970s of perforations caused by rectal thermometers;
however, axillary temperatures may not always accurately
reflect core temperature (Hutton et al, 2009).
An elevated body temperature at birth generally reflects
the intrauterine temperature and is not usually a sign of
sepsis (Baumgart, 2008). Isolated hyperthermia during
labor is associated with neonatal encephalopathy, occurring in approximately 1 in 2000 births (Blume et al, 2008).
After the first 3 to 4 days of life, increased temperatures
are most likely caused by dehydration from suboptimal
breast milk supply (Maayan-Metzger et al, 2003). A single
increased temperature in an otherwise normally behaving newborn is not a strong predictor of infection, but has
been reported as a sign of intracranial hemorrhage (Fang
et al, 2008).
ELIMINATION
Voiding
Approximately 15% of healthy newborns void at the time
of delivery, and 95% void by 24 hours of age. The cause
of delayed voiding is likely a consequence of stress on the
infant during labor and delivery (Vuohelainen et al, 2007;
2008), which is a protective mechanism for the baby. Normally no intervention is needed once homeostatic adaption to extrauterine life is stable.
The differential diagnosis of delayed voiding (defined as
no urine output by 24 to 48 hours of age) includes renal
and postrenal causes. With the frequent use of prenatal
ultrasound examination, it is unusual that a significant
renal anomaly is discovered because of a delay in voiding.
Most infants with bilateral renal agenesis have other findings, such as oligohydramnios or Potter sequence. Unilateral renal agenesis does not usually give symptoms of
decreased urine output. Renal vascular thrombosis can also
cause anuria, and babies with this condition are usually ill.
Severe cystic kidney disease can involve urinary outflow
obstruction. The diagnosis of cystic kidneys is usually
made after the newborn period, or it is found incidental to
evaluation of other anomalies and not because of delayed
voiding.
Postrenal causes of delayed voiding include neuropathic
bladder dysfunction and anatomic obstruction of urinary
flow by anomalies in ureters, the bladder, or the urethra.
Persistent or recurrent bladder distention after catheterization is found with occult lower spinal cord anomalies.
Presacral teratoma or other tumors can cause compression
and urinary blockage as well. In male infants, there is the
possibility of posterior urethral valves. Physical findings of
loose abdominal skin or musculature and a distended bladder suggest this diagnosis.
In a healthy-appearing newborn with a normal result on
prenatal ultrasound examination, the absence of enlarged
kidneys or palpable suprapubic mass, allowing up to
72 hours for a spontaneous first void, will avoid excessive
testing. In fussy neonates, infants with other genitourinary
abnormalities, enlarged kidneys, or distended bladder,
testing should begin immediately. Ultrasound examination of bladder, kidneys, and posterior urethra is often
diagnostic.
Normal newborns have decreased renal concentrating
ability and excessive extracellular free water at birth. As a
result, neonates will continue to void despite low intake of
fluids. This process is normal, with excess fluid volumes
being intrinsically protective against dehydration. Conversely, delayed voiding is not indicative of dehydration in
the first 72 hours of life.
Defecation
Similar to the first void, the first passage of meconium
occurs by an average of 7 hours of age. One third of newborns pass meconium before their first feeding. Late preterm newborns tend to pass meconium later than term
infants, and 32% of preterm infants do not pass meconium
in the first 2 days of life. In the first few days of life, intake
is not well correlated with meconium output. However,
CHAPTER 26 Routine Newborn Care
the number of wet and soiled diapers reflects adequacy of
breast milk production on day 4 of life. Fewer than four
soiled diapers on day 4 correlates with inadequate milk
production (Nommsen-Rivers et al, 2008). By 2 weeks
of age, breastfed infants pass feces more frequently than
bottle-fed infants; they also have larger variability in time
between bowel movements (Sievers et al, 1993).
Because 99.7% of healthy newborns pass meconium by
34 hours of age, those who are delayed beyond that time
deserve extra vigilance during examination to avoid missing obstructions, such as an imperforate anus (Metaj et al,
2003). A baby with abdominal distention or vomiting and
delayed stooling deserves evaluation for a possible gastrointestinal tract obstruction.
JAUNDICE
There are few conditions in newborn infants that create as
much controversy and clinician and parental angst as does
hyperbilirubinemia. Since the discovery of phototherapy in
1956 and its integration into medical care in the 1960s, the
standard management of neonatal jaundice in the United
States has gone through three distinct phases. Until the
early 1990s, clinicians visually monitored full-term neonates during their 2- to 5-day newborn nursery stay and
obtained serum bilirubin levels on those with significant
jaundice. Phototherapy was begun when the total bilirubin was 15 mg/dL, and an exchange transfusion was indicated if the level rose to 20 mg/dL (Watchko et al, 1983).
The wisdom of this approach was challenged by several
significant events. First, there was a growing awareness of
the increase in costs associated with the prolonged hospitalization of a healthy infant for phototherapy to treat
a relatively modest level of hyperbilirubinemia. Second,
Kemper et al (1989, 1990) conducted a series of studies
documenting that mothers of neonates who received phototherapy were at risk of overmedicalizing their children.
Finally and most significantly, informal and formal reviews
of data on jaundice in full-term newborns, without hemolytic disease, revealed that the risk of kernicterus in such
infants was extraordinarily low (Newman and Maisels,
1992; Watchko et al, 1983).
Based on this evidence, a “kinder and gentler” approach
to the management of hyperbilirubinemia in term infants
was advocated, leading to the AAP practice parameter in 1994 (AAP Provisional Committee for Quality
Improvement and Subcommittee on Hyperbilirubinemia,
1994). Under this guideline, phototherapy for a healthy,
72-hour-old, full-term newborn was not definitively recommended unless the serum bilirubin was 20 mg/dL or
greater. Unfortunately, publication of this guideline corresponded to a shortening of the nursery stay by term
infants to as little as 24 hours. Therefore infants were
discharged home before their levels of bilirubin peaked
at 3 to 4 days of life, and there were numerous reports of
infants with extremely high bilirubin levels and a general
impression that the incidence of kernicterus was increasing. In retrospect, it does not appear that the incidence
of kernicterus increased, but case reports and anecdotal
evidence led to significant consternation by clinicians,
parents, and quality assurance organizations (Burke et al,
2009).
311
Because newborns are usually discharged well before
bilirubin levels reach their peak, it is clear that predictive
models were needed to assess risk in newborns who are
discharged early. Bhutani et al (1999) developed a nomogram based on data from neonates in whom serum bilirubin levels were measured multiple times. Infants who had
initial bilirubin levels above the 95th percentile for any
time period were significantly more likely to have “significant hyperbilirubinemia” detected in subsequent bilirubin
measurements. These data were used in the development
of the 2004 AAP practice guideline (AAP Subcommittee on Hyperbilirubinemia, 2004b). With this iteration
of the guideline, clinicians are provided hourly guidance
on levels of bilirubin for which phototherapy or exchange
transfusions are indicated. Separate curves for low-risk,
medium-risk, and high-risk neonates have been developed.
Internet-based tools are available that provide the appropriate management for an infant with a specific bilirubin at
a specific hour of life.
Although the AAP bilirubin nomogram provides useful
information, there are some caveats. First, in the study by
Bhutani et al (1999), 39.5% of infants with an initial bilirubin level greater than the 95th percentile had significant
hyperbilirubinemia at subsequent testing. Therefore more
than 60% of such infants had less severe hyperbilirubinemia when retested. The nomograms for medium- and
high-risk infants are based almost exclusively on expert
opinion rather than actual data. Given these limitations, it
is important for the clinician to determine management of
a jaundiced neonate on an individual basis, remaining cognizant of the infant’s medical condition, social situation,
and parental preferences.
There are numerous neonatal conditions that increase
the risk for hyperbilirubinemia (Dennery et al, 2001); chief
among these is hemolysis secondary to maternal antibodies to red blood cell antigens. Thankfully, hemolysis secondary to antibodies to Rh factor is rare because of proper
management of Rh-negative mothers. Because there is no
way to prevent hemolysis from an ABO incompatibility,
it may be useful to test cord blood from neonates born to
mothers with blood type O for blood type and the presence of antibodies on their red cells (i.e., Coombs’ test) at
birth. The increase in bilirubin secondary to ABO compatibility is highly variable, even with a positive direct
Coombs’ test. Some infants will have an early and dramatic rise in serum bilirubin and evidence of hemolysis,
whereas in others no effect can be detected clinically. In
addition to ABO incompatibility, some women will have
antibodies to minor red cell antigens that can usually be
diagnosed prenatally. In most instances the increases in
bilirubin associated with antibodies against minor antigens are mild.
Other neonatal conditions that are risk factors for
hyperbilirubinemia include bruising secondary to birth
trauma and polycythemia. Decreased intake of breast milk
can lead to decreased passage of stool. Because intestinal
bacteria break down conjugated bilirubin to the unconjugated form, a decrease in stooling can lead to increased
reabsorption of this unconjugated bilirubin (enterohepatic
circulation). Breastfeeding is a significant risk factor for
hyperbilirubinemia particularly when intake is limited. The
propensity for developing significant jaundice is variable in
312
PART VII Care of the Healthy Newborn
different racial groups. Asian and American Indian infants
are at the highest risk for significant hyperbilirubinemia
(Dennery et al, 2001). Finally, late preterm infants are at
significantly increased risk for significant hyperbilirubinemia and kernicterus.
Traditionally, visual assessment has been used to judge
whether a newborn infant has significant jaundice. This
method of assessment is moderately accurate, but may be
sufficient to rule out the need for serum bilirubin testing in
many full-term newborns with no risk factors for jaundice
(Moyer et al, 2000; Riskin et al, 2008). Transcutaneous
bilirubinometers offer a noninvasive method for screening for hyperbilirubinemia. Depending on the technology and brand used, these instruments generally provide
estimates of transcutaneous bilirubin (TcB) that correlate
well with serum values. However, in practice it is not clear
whether categorical estimates of the severity of neonatal
jaundice using the transcutaneous meter are actually better than a visual assessment (Kaplan et al, 2008). Because
the TcB may be lower or higher than the serum level in
actual clinical practice, it should be considered as a screening tool only. Serum bilirubin testing remains the standard
on which management decisions are based. Transcutaneous testing may be warranted for infants with risk factors
for significant hyperbilirubinemia, even in the absence of
significant jaundice.
Unless levels are high enough to require an exchange
transfusion, phototherapy is effective for treating an infant
with significant hyperbilirubinemia. Although repeated
measurements of direct bilirubin are not cost effective,
one assessment is helpful before or immediately after initiating phototherapy to rule out direct hyperbilirubinemia
(Newman et al, 1991). An assessment of the potential for
hemolysis as the etiology of the elevated bilirubin, possibly including a review of maternal and infant blood type,
direct Coombs’ test, hematocrit, reticulocyte count, and
red cell morphology may also be useful.
There is no conclusive evidence as to whether continuous phototherapy leads to more rapid reduction in
serum bilirubin levels than does intermittent treatment
(AAP Subcommittee on Hyperbilirubinemia, 2004b; Lau
and Fung, 1984). Unless bilirubin levels are approaching exchange transfusion levels, it is probably reasonable
to discontinue treatment for several minutes to 1 hour at
frequent intervals to allow parents to feed and hold their
baby. Serial bilirubin measurements are needed to determine the adequacy of therapy and to determine when phototherapy can be discontinued. A rebound bilirubin level
obtained 24 hours after discontinuation of phototherapy
may be helpful in some clinical situations.
RESPIRATORY COMPLICATIONS
The term or late preterm fetus accomplishes the transition from dependency on the placenta to the newborn cardiorespiratory system, for the most part, without incident.
After birth, pulmonary blood flow increases, fetal shunts
reverse and begin to close, spontaneous breathing effort is
initiated, and fetal lung fluid is cleared. Effective cardiorespiratory function, as represented by an absence of respiratory distress (nasal flaring, grunting, chest wall retractions,
a respiratory rate of greater than 60 per minute) and an
oxygen saturation in the middle 90s should be established
by several hours of age (Levesque et al, 2000; O’Brien
et al, 2000).
This normal sequence of events fails to occur in 2% to
8% of infants born at 34 weeks; gestation or later (Farchi et al, 2009; Hansen et al, 2008; Yoder et al, 2008).
It is important to keep in mind that initial presenting symptoms are relatively nonspecific. Agrawal et al
(2003) studied a large number of consecutive births in an
attempt to determine the frequency and nature of different early-onset respiratory disorders and found that more
than half did not meet specific diagnostic criteria. When
confronted with a neonate with early onset respiratory
symptoms, the most important diagnostic considerations
include:
ll
ll
ll
ll
ll
ll
ll
ll
Complex structural cardiac system anomalies; incidence
estimated to be between 0.2% and 0.3%, often but not
always identified by in utero imaging studies
Diaphragmatic hernia, incidence estimated to be
between 0.04% and 0.08%, commonly identified by
second-trimester ultrasound (de Buys Roessingh and
Dinh-Xuan, 2009)
Respiratory distress syndrome (RDS); incidence estimated to vary between 0.45% and 2.4%, depending on
the population studied; risk increased in the late preterm and infants delivered by cesarean section, particularly if accomplished before labor (Jain et al, 2009; Tita
et al, 2009; Yoder et al, 2008)
Persistent pulmonary hypertension of the newborn;
incidence of 0.1% to 0.3%; often occurs in association
with other acute respiratory conditions; questionable
increased risk with maternal selective serotonin reuptake inhibitor treatment (Andrade et al, 2009; Chambers et al, 2006; Konduri and Kim, 2009)
Meconium aspiration syndrome; incidence reported to
vary between 2% and 9% of infants delivered through
meconium-stained amniotic fluid (7% to 20% of all
deliveries); risk increased in infants delivered after 40
weeks’ gestation, intrapartum distress, or both (Bhutani, 2008; Liu and Harrington, 2002)
Spontaneous pneumothorax; incidence between 0.1%
and 0.8%; infants born by cesarean delivery may be
at increased risk (Benterud et al, 2009; Zanardo et al,
2007)
Transient tachypnea of the newborn (TTNB); incidence variable between 0.3% and 3.9%; risk factors
include late prematurity and cesarean section; initial
diagnosis sometimes difficult to differentiate from
pneumonia and early RDS (Guglani et al, 2008; Jain et
al, 2009; Tita et al, 2009; Yoder et al, 2008)
Pneumonia; incidence difficult to determine, with one
recent retrospective report estimated rate at 0.3%; risk
factors include maternal chorioamnionitis and prolonged ruptured membranes; sometimes difficult to
differentiate from RDS and/or TTNB (Yoder et al,
2008)
A review of the maternal history—particularly pregnancy, labor, and delivery—can provide useful diagnostic
information. For example, the results of a second-trimester
ultrasound examination could reveal the possibility of
a cardiac defect or diaphragmatic hernia. A positive
CHAPTER 26 Routine Newborn Care
maternal GBS test result without adequate treatment,
prolonged rupture of amniotic membranes, or evidence
of chorioamnionitis suggests the possibility of pneumonia. For infants with respiratory distress born by cesarean section before the onset of labor, a diagnosis of RDS
should be considered and assessment of gestational age
should be performed. Finally, TTNB is a diagnosis of
exclusion; it is prudent to rule out other causes before it
is considered as the cause of respiratory distress in a term
neonate.
In most cases minimal initial diagnostic efforts for
a term newborn with unsuspected respiratory distress
should include a chest radiograph and assessment of the
arterial oxygen saturation. The results of these studies, in
combination with maternal history, should provide information helpful to: (1) establish initial management, such
as the need for supplemental oxygen, continuous monitoring, or both; (2) determine the need for further work-up or
treatment, possibly including an echocardiogram, laboratory testing, and treatment for possible sepsis; or (3) give
a referral for further specialty consultation, intensive care,
or both (in severe cases).
CARDIOVASCULAR ISSUES
Congenital heart disease is a relatively common condition in newborns, with an estimated incidence of 81 cases
per 10,000 live births (Reller et al, 2008). Ventricular
septal defect (VSD) is by far the most common defect,
accounting for more than 30% of all cases. The increasing
accuracy of prenatal ultrasound examination has greatly
improved the early diagnosis of complex congenital heart
disease. The results of population-based reviews indicate
that the sensitivity of routine prenatal ultrasound examination in identifying selected congenital defects is as high as
70% and ranges as high as 85% for hypoplastic left heart
(Chew et al, 2007; Rasiah et al, 2006). For mothers at high
risk of delivering a newborn with congenital heart disease,
the use of fetal echocardiography is helpful for delineating
the anatomy and significance of specific lesions. However,
many of the most common defects, particularly VSD, are
not typically detected prenatally.
In the absence of a prenatal diagnosis, detection of congenital heart disease is by physical examination. Sequential examinations are most helpful. At birth, many babies
have loud murmurs that are thought to be from either a
closing ductus arteriosus or tricuspid regurgitation (Silberbach and Hannon, 2007). These murmurs are transient and not indicative of disease. Conversely, murmurs
associated with VSDs may not be heard for several days
when the pressures on the right side of the heart have
dropped enough to permit a significant shunting of blood
from left to right. Although the ratio of pathologic to
benign murmurs is higher in newborns than in older children, most of the murmurs heard during the newborn
nursery stay in a healthy neonate are not clinically significant. Characteristics that increase the likelihood that
a murmur signifies the presence of congenital heart disease include an intensity of 3/6 or greater, a harsh quality,
occurrence during all of systole or into diastole, and being
heard best at the lower sternal border or right upper
border (Mackie et al, 2009). In a healthy newborn, the
313
most common presentation of congenital heart disease is
a somewhat harsh systolic murmur that is heard best at
the lower left sternal border in an asymptomatic infant,
indicative of a VSD.
In addition to auscultation, it is helpful to assess a newborn with a murmur for dysmorphic features and other
anomalies, because these findings increase the likelihood
that the murmur is indicative of congenital heart disease.
It is important to evaluate the adequacy of femoral pulses
to rule out coarctation of the aorta. Femoral pulses may
be difficult to palpate in a neonate; if there is uncertainty,
upper and lower extremity blood pressures can be measured. Although it is not a good screening tool, measurement of oxygen saturation may be helpful in diagnosing
a cyanotic lesion in a child with a murmur or other signs
of heart disease, particularly because hypoxia is frequently
difficult to detect in newborns (Mahle et al, 2009). Chest
radiographs and electrocardiograms are usually of limited value in evaluating healthy newborns with murmurs
(Mackie et al, 2009; Oeppen et al, 2002).
Full-term neonates frequently have alterations in cardiac rhythm and rate. Heart rates in full-term infants can
range as high as 200 beats/min or as low as 80 beats/min.
These values are usually indicative of normal variation and
are not clinically meaningful unless there are other signs of
illness or if there is a lack of variability in rate with stimulation or attempts at calming the newborn. Arrhythmias
are also not uncommon, occurring in approximately 1% of
newborns (Oeppen et al, 2002). By far, the most common
arrhythmia in a healthy-appearing, full-term newborn is
from premature atrial contractions (Larmay and Strasburger, 2004; Southall et al, 1981). These contractions are
almost always benign and are usually transient. If there is
concern about an irregular rhythm in a newborn, an electrocardiogram can be obtained. With premature atrial
contractions, the irregular beat is initiated by a P wave.
Although the QRS complex may be widened, it is always
be preceded by the P wave. In most cases no further workup is needed. Cardiology consultation may be warranted if
premature atrial contractions are persistent or if widened
QRS complexes are seen on an electrocardiogram.
POSSIBLE NEONATAL SEPSIS
Group B Streptococcus Screening
and Intrapartum Antibiotics
Since the 1970s, GBS has been a major cause of neonatal
sepsis (Schuchat, 1998). The implementation of intrapartum antibiotic prophylaxis (IAP) to prevent early-onset
GBS disease in neonates was associated with an 80%
decrease in the rate of infection (Phares et al, 2008). The
currently recommended strategy to prevent GBS disease
is to obtain rectovaginal cultures on all pregnant women
at 35 to 37 weeks’ gestation and to administer penicillin or
ampicillin during labor to those colonized with the bacteria. In situations where the mother’s GBS status is unknown
before the onset of labor, IAP is advised for those with certain risk factors for neonatal infection (Box 26-3; Schrag
et al, 2002). A third strategy may become available with
the development of rapid strep testing, which can be done
intrapartum. Overall, it is estimated that IAP can reduce
314
PART VII Care of the Healthy Newborn
BOX 26-3 R
isk Factors for Group B
Streptococcus Sepsis in
Newborns
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Delivery before 37 weeks’ gestation
Maternal fever during labor (body temperature >38.5° C)
Prolonged rupture of membranes (>18 hours)
Chorioamnionitis (maternal fever >38.5° C, tender uterus, fetal tachycardia)
Prior child with group B Streptococcus disease
Group B Streptococcus infection during pregnancy (urinary tract infection or
bacteremia)
Young maternal age
African American race
Hispanic ethnicity
Meconium-stained amniotic fluid
Newborn low absolute neutrophil count (e.g., immature:total ratio >0.2)
the risk of early-onset GBS disease by 83% (Ohlsson and
Shah, 2009). The overall rate of early-onset disease in the
United States is approximately 0.3 to 0.4 cases per 1000
live births (Centers for Disease Control and Prevention,
2009; Van Dyke et al, 2009).
Unfortunately, despite an effective strategy to prevent
the infection, the rate of GBS disease in the United States
increased between 2003 and 2006 (Centers for Disease
Control and Prevention, 2009). Much of this increase
was seen in African American infants. Although the rate
of infection was 2.8-fold higher in premature infants than
term newborns, 72% of the cases of GBS disease during this period were in full-term neonates. In addition,
although 93% of mothers of infants in this series with
GBS had been screened, IAP was administered to only
20%. Finally, although GBS cultures are highly accurate,
there is always the possibility of a false-negative screen. In
one review, 61% of infants with GBS disease were born
to mothers with a negative test result before delivery (Van
Dyke et al, 2009). These statistics highlight the need for
continued vigilance for signs of GBS infection in term
newborns by health care providers, even in an era of surveillance and IAP.
Guidelines for prevention of GBS sepsis indicate that
neonates are adequately treated only if IAP is administered
at least 4 hours before delivery (Schrag et al, 2002). There
is no clinical evidence to support this recommendation; it
is based on expert opinion and is used to provide a margin
of safety. However, there is evidence that penicillin given
2 hours before delivery is 90% effective in preventing
GBS sepsis, and IAP provided less than 2 hours prior to
delivery may be less effective (Illuzzi and Bracken, 2006).
Blood levels are higher in the neonate than in the mother,
even 30 minutes after a dose of penicillin, but the clinical
importance of this has not been studied (Colombo et al,
2006).
Term infants whose mothers have received IAP for a
positive GBS screen at least 4 hours before delivery can be
safely discharged at 24 hours of age if there are no signs
or symptoms of infection (Ohlsson and Shah, 2009). One
area of consternation is how long to observe term newborns when antibiotics were not given more than 4 hours
before delivery. In the current guidelines it is recommended that such infants be monitored in the hospital for
at least 48 hours after birth. The pressure for early newborn discharge has led some to compromise this period
of observation. Most infants with sepsis manifest it early
in life—many at birth, and nearly all by 12 hours of age
(Escobar et al, 2000). Among a group of 172 term infants
with documented early-onset GBS infection, 95% had
presenting symptoms within 24 hours after delivery. More
importantly, among the 33 neonates in this study that had
GBS infection despite IAP, 31 had signs of infection before
24 hours of age (93.9%; 95% confidence interval, 79.8%
to 99.3%; Bromberger et al, 2000). These data suggest,
but do not definitively indicate, that some term newborns
born to GBS positive mothers who received IAP less than
4 hours before delivery can be discharged by 48 hours of
age. This decision is best made on an individual basis considering all risk factors, examination of the baby, vital sign
stability, the results of any available laboratory tests, and
parental wishes.
Assessment of Term Newborn for
Possible Sepsis
The decision to remove a baby from the care of his or
her parents during the period of initial homeostasis, and
bonding should not be done without significant concern for the well-being of the infant. These decisions
can have permanent effects on the parent-child relationship (Paxton and Nyington, 2001; Pearson and Boyce,
2004). Conversely, neonatal sepsis is such a dangerous
and potentially rapidly progressive condition that any
suggestive signs and symptoms cannot be ignored. It
is between these extremes that the provider dwells and
must make decisions often based on uncertainty and
vague information.
The strongest predictor of sepsis is whether the baby is
ill. This predictor can be somewhat challenging to determine given the limited repertoire of behaviors in newborns.
Neonates typically have erratic temperature, poor feeding,
periods of lethargy, and crying, all of which can also be
symptoms of infection. Some of the earliest signs of infection include resting tachypnea or tachycardia, especially
with reduced heart rate variability and transient decelerations (Griffin et al, 2005). This level of early identification
requires continuous monitoring which, although routinely
used on premature infants, is not routinely performed
on term newborns. Because the infection entry portal is
often the lungs, increased respiratory rate or increased
work of breathing are the most common symptoms noted
(Andersen et al, 2004). Fever is not a common symptom
in an infected newborn, and usually babies with high temperatures are found to have other causes, especially after
24 hours of age (Maayan-Metzger et al, 2006). Decreased
skin perfusion or mottling and decreased or increased tone
are also worrisome signs.
GBS is not the only cause of sepsis in newborns. Coliforms were the dominant cause in the 1950s, and current
causes may involve a natural cycle in the dominant bacteria causing sepsis. There are also sporadic cases of sepsis
caused by enterococci, Serratia spp., and gram-negative
bacteria such as Escherichia coli, Proteus spp., and Klebsiella
spp. Although the decision of whether to evaluate or treat
a neonate for possible sepsis cannot be informed only by
CHAPTER 26 Routine Newborn Care
the GBS status of the mother, and because other causes of
infections are rare, most guidelines focus on GBS sepsis
risk.
In general, the physical examination is as good as laboratory testing in identifying ill newborns (Escobar et al,
2000). Laboratory tests can aid in the prediction that a
baby is ill, but testing is also viewed as invasive by parents,
so it is often helpful to carefully explain the tests and how
the results will be used to make decisions. There is no one
single test that has shown superiority, and opinion on testing varies. Among the most useful tests are the absolute
neutrophil count, immature-to-total neutrophil ratio, and
the procalcitonin level (Carrol et al, 2002; van Rossum
et al, 2004). The C-reactive protein or erythrocyte sedimentation rate may help, but have lower predictive values
(Galetto-Lacour et al, 2003).
The decision to start testing or treating a term newborn
for suspected sepsis should be multivariate and include:
(1) risk factors during pregnancy, labor, and delivery (see
preceding section); (2) current age in hours of the newborn; (3) presence of concerning signs and symptoms; and
(4) physical examination. One single finding, such as an
elevated temperature in an otherwise normally behaving
newborn, is not usually sufficient evidence to start intravenous antibiotics. If the status of a term newborn is not
rapidly changing, the use of close observation and periodic reassessment of risk factors and findings may prevent
unnecessary testing or treatment.
315
SUGGESTED READING
Burke BL, Robbins JM, Bird TM, et al: Trends in hospitalizations for neonatal
jaundice and kernicterus in the United States, 1988-2005, Pediatrics 123:
524-532, 2009.
Bhutani VK, Johnson L, Sivieri EM: Predictive ability of a predischarge hourspecific serum bilirubin for subsequent significant hyperbilirubinemia in
healthy term and near-term newborns, Pediatrics 103:6-14, 1999.
Dewey KG, Nommsen-Rivers LA, Heinig MJ, et al: Risk factors for suboptimal
infant breastfeeding behavior, delayed onset of lactation, and excess neonatal
weight loss, Pediatrics 112:607-619, 2003.
Nommsen-Rivers LA, Heinig MJ, Cohen RJ, et al: Newborn wet and soiled diaper
counts and timing of onset of lactation as indicators of breastfeeding inadequacy, J Hum Lact 24:27-33, 2008.
Escobar GJ, Li DK, Armstrong MA, et al: neonatal sepsis workups in infants
>/=2000 grams at birth: a population-based study, Pediatrics 106:256-263,
2000.
Kramer MS, Aboud F, Mironova E, et al: Breastfeeding and child cognitive
development: new evidence from a large randomized trial, Arch Gen Psychiatry
65:578-584, 2008.
Kramer MS, Chalmers B, Hodnett ED, et al: Promotion of Breastfeeding
Intervention Trial (PROBIT): a randomized trial in the Republic of Belarus,
JAMA 285:413-420, 2001.
Kuzniewicz MW, Escobar GJ, Newman TB: Impact of universal bilirubin
screening on severe hyperbilirubinemia and phototherapy use, Pediatrics 124:
1031-1039, 2009.
Lee RS, Cendron M, Kinnamon DD, et al: Antenatal hydronephrosis as a predictor
of postnatal outcome: a meta-analysis, Pediatrics 118:586-593, 2006.
Phares CR, Lynfield R, Farley MM, et al: Epidemiology of invasive group B streptococcal disease in the United States, 1999-2005, JAMA 299:2056-2065, 2008.
Van Dyke MK, Phares CR, Lynfield R, et al: Evaluation of universal antenatal
screening for group B streptococcus, N Engl J Med 360:2626-2636, 2009.
Tarini BA, Christakis DA, Weich HG: State newborn screening in the tandem
mass spectrometry era: more tests, more false-positive results, Pediatrics 118:
448-456, 2006.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
27
Newborn Screening
Inderneel Sahai and Harvey L. Levy
Newborn screening is directed primarily at disorders in
which the clinical complications develop postnatally. In
metabolic diseases, these complications result from biochemical abnormalities that appear after birth, when the
infant is no longer protected by fetal-maternal exchange.
For example, the infant with phenylketonuria (PKU) has a
normal blood phenylalanine level at birth, but within a few
hours demonstrates hyperphenylalaninemia. The infant
with congenital hypothyroidism (CH) is also protected
in utero, most likely from placental transfer of maternal
thyroxine (T4). If the hyperphenylalanemia in PKU is not
controlled by diet or the hypothyroidism in CH is not corrected by supplemental T4, the infant begins to show signs
of developmental delay and subsequently becomes mentally retarded. If therapy begins during the first weeks of
life, mental retardation in both disorders is prevented.
PKU was the first metabolic disorder known to benefit from dietary therapy. This fact was established by the
middle 1950s. By the late 1950s, it was evident that the
diet could prevent mental retardation if initiated in the
neonatal period. Detecting PKU in all affected infants at
that early age, before irreversible brain damage occurred,
then became the challenge. This challenge entailed neonatal screening for a biochemical marker of the disease.
In 1962, Guthrie developed a simple bacterial assay for
phenylalanine that required only a small amount of whole
blood soaked into filter paper (Guthrie and Susi, 1963).
Therefore infants in newborn nurseries could be routinely
tested for PKU in blood specimens obtained by lancing
the heel and blotting the drops of blood onto a filter paper
card. This filter paper blood specimen (dried blood spot
specimen) could be mailed to a central laboratory for PKU
testing. An increased concentration of phenylalanine in
the specimen indicated PKU in the infant.
By the middle 1960s, many states had established routine
newborn screening programs for PKU using the Guthrie
method. Infants with PKU were identified in larger numbers than anticipated and were showing normal development while receiving treatment (O’Flynn, 1992). The
success of PKU screening led to the addition of tests for
other metabolic diseases, including galactosemia, maple
syrup urine disease (MSUD), and homocystinuria. These
additional tests could be performed on the same blood
specimen obtained for PKU screening. Over time, a test
was added for the endocrine disorder congenital hypothyroidism, followed by screening tests for sickle cell disease,
congenital adrenal hyperplasia (CAH), biotinidase deficiency, cystic fibrosis, and others.
In 1990, tandem mass spectrometry (MS/MS) was first
applied to the Guthrie specimen, opening a new era in newborn screening (Levy, 1998; Millington et al, 1990). This
technology allowed for the accurate detection of numerous biochemical markers with a single assay, thereby making redundant several assays traditionally used in screening
316
for metabolic disorders. Furthermore, it enabled analysis of
biomarkers not detectable by previous methodologies, thus
greatly expanding the spectrum of conditions identifiable in
the neonate (Levy and Albers, 2000). By the early 1990s,
Naylor (Chace and Naylor, 1999) and Rashed et al (1995)
began using this technology to routinely screen neonates for
more than 20 biochemical disorders with high specificity
and an extremely low rate of false-positive results. Currently
most programs in the United States and screening programs
in Europe and elsewhere have integrated MS/MS into newborn screening, and many are screening for more than 50
individual conditions (http://genes-r-us.uthscsa.edu).
In order to standardize screening panels nationwide,
the American College of Medical Genetics has recommended a panel of 29 core conditions for screening (Table
27-1). An additional 25 secondary conditions were listed
for which test results could be reported (American College
of Medical Genetics, 2006). These secondary conditions,
revealed in the course of screening for the 29 core conditions, are not well known or documented treatment is
unavailable.
Molecular diagnostic techniques (i.e., DNA analysis)
are also commonly used in newborn screening, predominantly as secondary tests for diseases such as cystic fibrosis or medium-chain acyl-CoA dehydrogenase deficiency
(MCADD). Molecular testing substantially improves the
positive predictive value of a primary screening result that
is based on metabolite testing only (Ranieri et al, 1994;
Wilcken et al, 1995; Ziadeh et al, 1995). With the initiation of pilot screening for severe combined immunodeficiency syndrome wherein the markers measured are DNA
molecules (i.e., T cell receptor excision circles), the role of
molecular technology has extended into primary marker
analysis (Baker et al, 2009; Vogt, 2008). The use of molecular testing in screening will most likely expand further
with advances in DNA technology.
Screening for neuroblastoma, the most common solid
tumor of childhood, was formerly being performed in
Japan and some European programs measuring vanillylmandelic acid and homovanillic acid in urine collected
from infants (Sawada, 1993). However, this screening was
abandoned because it failed to identify individuals with
poor-prognosis neuroblastoma, instead identifying infants
who had a form of neuroblastoma that either spontaneously regressed or could be effectively treated after clinical
detection (Woods et al, 2003).
SCREENING PROCEDURE
SPECIMEN
The blood specimen is generally obtained from the heel
of the infant. This simple sampling method, conceived
and introduced by Guthrie and Susi (1963), has had an
CHAPTER 27 Newborn Screening
317
TABLE 27-1 Core Disorders Recommended for Screening by American College of Medical Genetics
Disorder
Acronym
Primary Marker
Beta-ketothiolase deficiency (mitochondrial acetoacetyl CoA thiolase deficiency)
BKT
C5:1/C5OH
Cobalamin defects A, B
CBL (A,B)
C3
Isovaleric acidemia*
IVA
C5
Glutaric aciduria I
GA-I
C5DC
3-Hydroxy 3-methylglutaryl-CoA lyase deficiency*
HMG
C5OH/C5-3M-DC
Multiple carboxylase deficiency*
MCD
C3/C5OH
3-Methylcrotonyl-CoA carboxylase deficiency
3MCC
C5OH
Methylmalonic aciduria (mutase)*
MMA
C3
Propionic acidemia*
Fatty acid oxidation defects
PA
C3
Carnitine uptake defect (carnitine transporter defect)
CUD
C0
Long-chain hydroxyacyl-CoA dehydrogenase deficiency*
LCHAD/D
C16OH/C18:1OH
Medium-chain acyl-CoA dehydrogenase deficiency
MCAD/D
C8
Trifunctional protein deficiency*
TFP
C16OH/C18:1OH
Very-long-chain acyl-CoA dehydrogenase deficiency
Amino acid disorders
VLCAD/D
C14:1/C14
Argininosuccinic aciduria (argininosuccinate lyase deficiency)*
ASA
ASA
Citrullinemia I (argininosuccinate synthase deficiency)*
CIT-I
Citrulline
Phenylketonuria
PKU
Phenylalanine
Maple syrup urine disease*
MSUD
Leucine
Homocystinuria
HCY
Methionine
Tyrosinemia type I
TYR-I
Tyrosine
Biotinidase deficiency
BIOT
Biotinidase activity
Galactosemia*
GALT
Total galactose, GALT activity
Congenital adrenal hyperplasia*
CAH
17-Hydroxyprogesterone
Congenital hypothyroidism
CH
T4, TSH
Sickle cell anemia
HbSS
Hb variants
Sickle cell disorder
HbS/C
Hb variants
Hemoglobin S/β-thalassemia
HbS/betaTh
Hb variants
Cystic fibrosis
CF
Immunoreactive trypsinogen
Hearing
HEAR
Metabolic Disorders Detected Using Tandem Mass Spectrometry
Organic acid disorders
Other Metabolic Disorders
Endocrine Disorders
Hemoglobin Disorders
Other Disorders
GALT, Galactose-1-phosphate uridyl transferase; T4, thyroxine; TSH, thyroid-stimulating hormone.
*Can manifest acutely in the first week of life.
enormous effect on newborn screening. The specimen is
easily obtained and easily and inexpensively sent by mail
to a central testing facility. There are no complications
in obtaining the specimen from the newborn, contrary
to early fears that its collection would lead to infection or
result in excessive bleeding.
SPECIMEN COLLECTION PROCEDURE
The blood specimen should be obtained from the lateral
or the medial side of the heel (Figure 27-1). Blood should
be applied to only one side of the filter paper card, but it
should saturate each circle on the card. Contamination of
the filter paper specimen with iodine, alcohol, petroleum
jelly, stool, urine, milk, or a substance such as oil from
the fingers can adversely affect the results of the screening tests. In addition, exposure to heat and humidity can
inactivate enzymes and produce false results. The specimen should be dried in air at room temperature for at least
3 hours before being placed in an envelope.
Specimens are sometimes collected in capillary tubes, by
venipuncture of a dorsal vein or from a central line, and
then spotted on filter paper. There is little or no substantial difference in analyte levels between blood collected
318
PART VII Care of the Healthy Newborn
FIGURE 27-1 Hatched areas at medial and lateral sides of the heel in
this drawing of the sole indicate the proper sites for a heel stick in the
newborn.
directly from the heel and that collected by any of these
other methods (Lorey and Cunningham, 1994). However,
there is the danger of introducing amino acids from total
parental nutrition solutions given through a central line
into blood collected from this line, resulting in a falsepositive increase in amino acids or interference in some
molecular assays by the heparin from the line. In general,
it is preferable that blood for screening be spotted on filter
paper directly from the heel.
TIMING OF COLLECTION
Newborn screening encompasses a gamut of conditions,
each with its own ideal screening period during which
there is the greatest chance of diagnosing the disorder and
before onset of symptoms. As a result, it is worth noting
that recommendations on the timing of specimen collection, although appropriate for the majority, may not be
ideal for all conditions on the screening panel. In congenital adrenal hyperplasia, in which the symptoms can
manifest within the first week of life, the optimal time for
collection of the specimen is within 24 to 48 hours after
birth. Formerly there was concern that with the newborn screening specimen collected early, often during
the first day of life, some infants with metabolic disorders
or CH might not have a sufficient degree of abnormality
for identification. However, MS/MS methodology with
its improved sensitivity and specificity has considerably
improved the reliability of screening for metabolic conditions in early specimens (Chace and Naylor, 1999) and
using thyroid-stimulating hormone (TSH) as the primary
marker for CH, or as a second-tier test when T4 is the
primary marker, has similarly allowed early screening for
CH to be reliable.
Specimen collection timings vary around the world.
In Europe and Australia, most screening specimens are
collected within 48 to 72 hours, whereas specimens in
the United Kingdom are not collected until the infant is
5 to 8 days old (U.K. Newborn Screening Progam Centre, 2008). In the United States, most screening specimens
are collected within 24 to 72 hours after birth. The specimen should be obtained from every newborn infant before
nursery discharge or by the third day of life, whichever is
first. In infants whose initial specimen was obtained within
the first 24 hours of life, as may happen with the practice of
early nursery discharge, a second blood specimen should
be obtained no later than 7 days of age to be certain that a
diagnosis is not missed.
Special circumstances require specific attention to newborn blood specimen collection. Premature infants or
those with very low birthweight, as well as infants who are
sick and those in neonatal intensive care units (NICUs),
are at a risk of unreliable screening owing to factors such
as the unique physiology of the infant, therapeutic interventions, and a focus on critical activities in caring for the
very sick neonate. Consequently, a single specimen is inadequate for screening in this subpopulation, and additional
specimens should be collected for retesting. Serial screening with collection of three specimens—upon admission
to the NICU, between 24 and 48 hours of life, and at discharge or at 28 days of life, whichever is sooner—has been
proposed as an adequate and efficient protocol for this
population (Clinical and Laboratory Standards Institute,
2009). In addition, some programs recommend screening
every month until discharge for babies who continue to
remain in the NICU.
A blood specimen should be collected from any infant
who is being transferred to a different hospital or to a
NICU, regardless of age. The first specimen should be
collected before transfer, and a second specimen at the
receiving hospital by 4 days of age. This dual collection
policy covers the infant from whom a newborn specimen
might not have been obtained in the turmoil that frequently accompanies the transfer of neonates.
In a newborn who is to receive a blood transfusion, a
screening specimen should be collected before transfusion,
and a second specimen should be collected 2 days after the
transfusion. In addition, a third screening specimen should
be obtained 2 months after the transfusion, when most of
the donor red blood cells (RBCs) have been replaced. This
practice ensures reliable testing for analytes present in
RBCs, if a pretransfusion specimen has not been obtained.
Newborn screening tests are usually performed in a
centralized state, provincial, or regional laboratory. In a
regional program, the specimens may be received by the
state program and then delivered to the regional state
or private laboratory, or they may be sent directly to the
regional laboratory. In either case, the individual state programs serve as the state data and follow-up centers.
SCREENING TESTS
The testing procedure begins with the punching of small
discs (each usually 3 mm in diameter) from the screening
specimen. These small discs are then analyzed by various
methodologies for the individual markers being sought.
Amino acids and acylcarnitines, the markers for the majority of screened metabolic conditions, are simultaneously
measured by MS/MS (Rinaldo et al, 2004). MS/MS is
CHAPTER 27 Newborn Screening
superior in terms of accuracy of measurement of the individual analytes when compared with alternate methods
originally used for screening amino acids, such as bacterial
assays or fluorometric techniques.
Immunoassays, including fluoroimmunoassay and
enzyme-linked immunosorbent assays, are used to test for
endocrinopathies such as congenital hypothyroidism and
CAH, for infectious diseases such as congenital toxoplasmosis and human immunodeficiency virus (HIV) seropositivity, and for cystic fibrosis. Hemoglobin electrophoresis
of blood eluted from the filter paper disc is used for sickle
cell disease screening. An enzyme assay is often used to
screen for galactosemia and is always used to screen for
biotinidase deficiency. Molecular assays are applied to
quantify T cell receptor excision circles in dried blood to
screen for severe combined immunodeficiency syndrome.
More commonly, molecular assays are used to identify
known mutations in certain disorders as a second-tier test.
Several platforms, such as DNA microarrays and microsphere-based assays, can multiplex several molecular and
immunological assays for high-throughput screening and
are being used by screening programs (Dobrowolski et al,
1999; Green and Pass, 2005; McCabe and McCabe, 1999).
SECONDARY TESTS
An abnormal finding on a newborn screening test is not
diagnostic of a disorder. Abnormalities in the newborn
specimen can be transient or artifactual. Accordingly,
when an abnormality is identified, the original specimen is
retested for the analyte that was abnormal. Additional tests
can be performed by the screening laboratory to substantiate the finding and improve the specificity of screening
(Matern et al, 2007; Rinaldo et al, 2006).
In screening for congenital hypothyroidism, many programs initially measure T4 in the original newborn blood
specimen. Specimens in which a low T4 level is found are
further tested for an increased level of TSH, which would
indicate congenital hypothyroidism. A normal TSH level
would suggest transiently low T4, a common finding in
premature infants, or T4-binding globulin deficiency.
Some screening programs have adopted screening protocols in which the primary analysis is for TSH, and T4
is measured as a second-tier test in specimens with high
concentrations of TSH. Similarly, in screening for galactosemia an elevated galactose measurement in a specimen
can trigger the analysis of galactose-1-phosphate uridyltransferase (GALT) enzyme activity as a second-tier test.
Second-tier molecular testing is also performed in some
screening laboratories. For example, in screening for cystic
fibrosis, an initial out-of-range primary marker prompts
DNA analysis to identify several specific pathogenic mutations (Comeau et al, 2004; Rock et al, 2005; Wilcken
et al, 1995). Screening programs following this two-tiered
immunoreactive trypsinogen-DNA approach can identify
up to 99% of patients with cystic fibrosis and report a positive predictive value ranging between 1/9.5 to 1/25 (Grosse
et al, 2004). Molecular assays to detect disease-causing
mutations are currently used as second-tier tests for several other disorders, and their use is likely to expand with
advances in DNA technology. These uses include testing
for the prevalent c.985A>G mutation in MCADD, the
319
predominant E474Q mutation in long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD; Dobrowolski et al, 1999), and a panel of several GALT mutations
in galactosemia screening.
The final interpretation of the screening results is
based on the primary analysis and, if available, the results
of second-tier testing. However, it is important to realize that screening is not intended to be diagnostic; abnormal screening results must be supported by confirmatory
investigations. These studies require additional specimens
and are performed by clinical laboratories or sometimes by
the screening laboratory.
PHYSICIAN CONTACT FOR ABNORMAL
RESULTS
Table 27-2 indicates disorders or other reasons for abnormal screening results, sorted according to the primary analyte usually used to screen for the condition. For example, a
low T4 level together with an elevated TSH concentration
indicates congenital hypothyroidism, and a marked elevation of 17-hydroxyprogesterone (17-OHP) indicates the
likelihood of CAH. An elevation of an acylcarnitine could
indicate an organic acid or fatty acid oxidation disorder.
Any infant for whom such a screening result is reported
should be evaluated by the primary care provider as soon
as possible to facilitate the next steps towards the confirmation and management of the disorder. However, several
conditions screened for are extremely rare, and primary
health care professionals might not have sufficient information available to be able to direct appropriate intervention in screen-positive infants. To overcome the challenge,
readily accessible, one- to two-page explanations of the
possible disorders represented by the abnormality and the
recommended confirmatory tests, known as ACT sheets
and confirmatory algorithms (Kaye et al, 2006), are available on the Web site of the American College of Medical
Genetics (www.acmg.net/resources/policies/ACT/condi
tion-analyte-links.htm).
Although all specimens with a metabolite concentration
that crosses its threshold are considered screen-positive,
all screen-positive results are not associated with the same
likelihood of being associated with a disorder. Most infants
with a positive screening result that is only mildly abnormal are less likely to have a disorder (see later discussion
of false-positive results) than are infants with analyte concentrations that are several fold above the cutoff. Applying a uniform approach for all positive results in terms of
urgency of intervention or battery of tests suggested can
result in unnecessary parental anxiety and medical costs.
However, if recommendations for further action and workup are customized in accordance with the potential significance of the abnormality, both parental anxiety and costs
associated with false-positive results can be reduced. To
achieve this goal, some programs subcategorize positive
screening results. The New England Newborn Screening
Program uses primary marker concentrations, second-tier
analyses, biomarker profiles for markers analyzed by MS/
MS, and acuity of the likely disorder to subcategorize outof-range screening results (Sahai et al, 2007). The primary
care providers are supplied with category-based, customized fact sheets when a positive screening result is reported
320
PART VII Care of the Healthy Newborn
TABLE 27-2 Disorders or Other Reasons for Abnormal Screening Results Sorted by Primary Marker Analyzed
Marker
Disorders
Possible Causes of False Positives
Markers Analyzed Using Tandem Mass Spectrometry
Free carnitine (C0)
Low
CUD
Poor feeding, maternal CUD
Free carnitine (C0)
High
CPT-I
Carnitine supplementation
Propionylcarnitine (C3)
High
MMA, PA, CBL (A,B)
Hemolysis,* maternal biotin deficiency, carrier of
associated disorders
Butyrylcarnitine (C4)
High
SCADD/EE/IBDD
Hypoglycemia from other causes,* FIGLU elevation
Tigylcarnitine (5:1)
High
BKT deficiency
VLBW neonate
Isovalerylcarnitine (C5)
High
IVA
Antibiotics containing pivalic acid, IVA/MBCD
carrier, VLBW neonate, neonate receiving total
parental nutrition, FAS hemoglobin profile*
Glutarylcarnitine (C5DC)
High
GA-I
MCADD carrier, twin or multiple births*
3-Methylglutarylcarnitine (C5-3M-DC)
High
HMG
Severe respiratory distress, neonates receiving
ECMO*
Hydroxyisovalerylcarnitine (C5OH)
High
3MCC
Maternal MCC, maternal biotin deficiency
Octanoylcarnitine (C8)
High
MCADD
MCADD carrier, MCT supplementation
Tetradecenenoylcarnitine (C14:1)
High
VLCADD
Carrier
Hydroxyhexadecanoylcarnitine (C16OH)
High
LCHADD/TFP
Carrier
Hexadecanoylcarnitine (C16)
High
CPT-II
Severe hemolysis*
Arginine (Arg)
High
ARG
Hyperalimentation
Argininoscuccinic acid (ASA)
High
ASA
—
Citrulline (Cit)
Low
OTC/CPS/NAGS
Poor feeding*
Citrulline (Cit)
High
CIT-I
Hyperalimentation
Phenylalanine (Phe)
High
PKU
Hyperalimentation, specimen contaminated with
artificial sweetener*
Leucine (Leu)
High
MSUD
Hyperalimentation, hydroxyprolinemia
Methionine (Met)
High
HCY
Hyperalimentation
Tyrosine (Tyr)
High
TYR-I
Prematurity, transient immaturity of enzyme
Biotinidase activity (Bio)
Low
BIOT
Exposure of specimen to heat, improper drying
Total galactose (T-Gal)
High
GALT
GALE deficiency, GALK deficiency, contamination
with milk/cream
Activity GALT
Low
GALT
Exposure of specimen to heat
17-Hydroxyprogesterone (17-OHP)
High
CAH
Physiologic stress (seen commonly in NICU babies),
VLBW, EDTA in specimen
Markers analyzed by assays other than
tandem mass spectrometry
T4 / TSH
Low/High
CH
Neonates in NICU, maternal thyroid medications
Immunoreactive trypsinogen (IRT)
High
CF
VLBW neonate, NICU
T cell receptor excision circles (TREC)
Low
SCID
Other immunodeficiencies, Di George syndrome,
heparin in specimen*
BIOT, biotinidase deficiency; BKT, beta-ketothiolase deficiency; CAH, congenital adrenal hyperplasia; CBL, cobalamin defect; CF, cystic fibrosis; CH, congenital hypothyroidism;
CIT-I, citrullinemia I; CPT, carnitine palmitoyltransferase deficiency; CUD, carnitine uptake defect; ECMO, extracorporeal membrane oxygenation; EDTA, ethylenediamine
tetraacetic acid; EE, ethylmalonic encephalopathy; FIGLU, formiminoglutamic; GA-I, glutaric aciduria I; GALE, uridine diphosphate galactose-4-epimerase; GALK, galactokinase;
GALT, galactosemia; HCY, homocystinuria; HMG, 3-hydroxy 3-methylglutaryl-CoA lyase deficiency; IBDD, Isobut yryl-CoA dehydrogenase deficiency; IVA, isovaleric acidemia;
LCHADD, long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency; MBCD, 2-methylbutyryl-CoA dehydrogenase deficiency; MCC, methylcrotonyl-CoA carboxylase;
MCT, medium-chain triglycerides; MSUD, maple syrup urine disease; MMA, methylmalonic aciduria; MCADD, medium-chain acyl-CoA dehydrogenase deficiency; NICU, neonatal
intensive care unit; PA, propionic acidemia; PKU, phenylketonuria; SCADD, Short-chain acyl-CoA dehydrogenase; SCID, severe combined immune deficiency; TFP, trifunctional
protein deficiency; TYR-I, tyrosinemia type I; VLBW, very low birthweight; VLCADD, very-long-chain acyl-CoA dehydrogenase deficiency.
*Associations observed in the New England Newborn Screening Program.
(Sahai and Eaton, 2008). These sheets include information on disorders associated with the marker, estimated
likelihood of being affected, clinical presentations of likely
disorders, factors contributing to false positives, and recommendations for further management. The follow-up
recommendations can range from immediate admission to
a hospital, where further evaluation and therapy for the
illness can be initiated without delay, to simply repeating
the filter paper analysis on a sample collected a few days
later. Other programs approach this problem differently,
but with the same goal in mind, providing the primary care
providers with the information needed to put the result in
the appropriate context for the family.
Any infant for whom an abnormal screening result is
reported should be seen as soon as possible and evaluated
with a careful history and physical examination. When
specific guidelines based on the individual results are not
provided by the screening program, and the infant is ill or
CHAPTER 27 Newborn Screening
the likely disorder manifests acutely within the first few
days of life (see Table 27-1), a specialist should be contacted. The infant may need to be admitted to the hospital
where further evaluation and therapy for the illness can be
initiated without delay.
If the infant is active and alert with good feeding and
shows no abnormal signs on initial evaluation, and the suspected disorder does not require immediate attention, a
second filter paper blood specimen can be obtained and
sent to the screening laboratory for repeated testing, or
confirmatory testing can be performed on a less urgent
basis. In many cases, confirmatory testing or referral to a
specialist is required only if the second test indicates the
presence of a disorder. However, the follow-up of an initial positive screening result can vary. In some programs,
more specific confirmatory testing is the first response to
a presumptive positive newborn screen, with a less intense
time frame for individuals in whom the level of suspicion
is lower.
The physician should contact the screening laboratory
when an infant whose screen has been reported as normal
or whose screening results have not yet been reported has
symptoms that suggest a metabolic disorder. The screening laboratory can check the results in the infant’s newborn specimen. If the testing has been completed and the
newborn specimen is retained in storage, the laboratory
may wish to recover the specimen and repeat the tests. The
physician should also contact the screening laboratory for
the results of repeated tests and inform the family of the
results as soon as possible. If the second result is normal,
the duration of the family’s anxiety may be shortened.
SCREENED DISORDERS
Following are brief summaries of the categories of the
most common disorders detected by newborn screening.
There is no attempt to describe any of the disorders in
detail or their rare variants.
METABOLIC DISORDERS
Amino Acid Disorders
The amino acid disorders are caused by an enzymatic
defect in the catabolic pathway of amino acids, with consequent accumulation of specific amino acids above the
block. Screening relies on the detection of these elevated
amino acids in the newborn specimen. The clinical manifestations may be a result of the toxic effects of the accumulating amino acid and metabolites produced by alternate
pathways, a deficiency of the products of the normal pathway, or both. PKU is the best-known example of an amino
acid disorder and is the paradigm for screened disorders in
the newborn.
In addition to PKU, MSUD and homocystinuria are
historically significant in the context of screening, because
they are among the original metabolic conditions for
which screening was performed before the introduction of
MS/MS and therefore the expansion of screening. Screening for other amino acid disorders, such as the urea cycle
defects and tyrosinemia type I, became possible only with
the advent of MS/MS technology.
321
In PKU, the cardinal screening feature is an increased
level of phenylalanine. PKU should always be identified
by newborn screening. If untreated, patients with PKU
experience severe mental retardation and other neurologic
abnormalities. The average incidence of the disorder is
approximately 1 in 12,000 live births. With screening by
MS/MS, PKU can reliably be identified as early as the first
day of life (Chace et al, 1998). Not all infants with an elevation of phenylalanine have PKU. Mild elevations can indicate benign mild hyperphenylalaninemia. Liver disease,
such as that associated with galactosemia, tyrosinemia type
I or citrin deficiency, can also produce increased phenylalanine. Therefore treatment for PKU should never be
started on the basis of a positive screening test result alone.
The dietary therapy is complicated and can be hazardous
to an infant who does not have PKU. If the screening test
reveals a marked elevation of phenylalanine (≥6 mg/dL)
and the metabolic profile (including reduced tyrosine) is
supportive of PKU, the infant should be referred directly
to a metabolic center for confirmatory testing and prompt
consideration of dietary treatment. If the screening level of
phenylalanine is only slightly increased, retesting a second
specimen before initiating a complete diagnostic work-up
may suffice.
The primary indicator for MSUD in the newborn blood
specimen is an increase in leucine. To improve the specificity for MSUD of an abnormal leucine measurement,
some screening programs also report the ratio between
leucine and a reference amino acid or alloisoleucine analysis as a second-tier test (Matern et al, 2007). The average
incidence of classic MSUD is 1 in 185,000. MSUD can
be a fulminant disease associated with severe ketoacidosis, vomiting, and lethargy, and it can progress rapidly to
coma and death. Consequently, the finding of a substantially increased leucine level in the newborn blood specimen should prompt an immediate telephone call from the
screening program to the attending physician. If the infant
is ill, he or she should be transported immediately to an
NICU at a medical center with a metabolic specialist. Confirmatory plasma and urine specimens should be obtained,
and emergency therapy should be initiated. Plasma amino
acid analysis in an infant with MSUD will show marked
increases in leucine, isoleucine, and valine as well as alloisoleucine (the branched-chain amino acids). The urine
specimen will test strongly positive for ketones and will
contain large quantities of the branched-chain ketoacids
and amino acids. The characteristic odor reminiscent of
maple syrup, which appears earliest in cerumen and only
later in urine, will probably be detected on a cotton-tipped
swab inserted in the infant’s ear. Milder variants of MSUD
can be missed by newborn screening (Bhattacharya et al,
2006). A newborn with the intermediate variant might not
have a blood leucine elevation, or the increase may be so
mild as to be below the cutoff value. In the intermittent
variant, the blood leucine concentration is normal in the
newborn period, becoming elevated only in later infancy
or childhood during acute metabolic episodes precipitated
by febrile illness or surgery.
Individuals with homocystinuria are clinically normal at
birth but, if untreated, may develop ectopia lentis (dislocation of the lens), thromboembolism, osteoporosis, and
mental retardation. The worldwide frequency of all forms
322
PART VII Care of the Healthy Newborn
of homocystinuria has been estimated at 1 in 344,000, but
may be considerably higher (Skovby et al, in press). The
newborn blood screening marker for detecting homocystinuria is an increased level of methionine. Homocysteine
can be measured as a second-tier analysis to improve specificity (Matern et al, 2007). The diagnosis of homocystinuria may be missed if the blood methionine concentration
is not elevated at the time the newborn specimen is collected (Whiteman et al, 1979). Reducing the cutoff value
for methionine can substantially increase the frequency of
identified infants (Peterschmitt et al, 1999), but may also
result in an increased number of false-positive results.
A high methionine level alone is not diagnostic of homocystinuria. Liver disease of a nonmetabolic nature can
produce a strikingly high methionine level, as can isolated
hypermethioninemia (methionine-S-adenosyltransferase
[MAT] I/III deficiency), a metabolic disorder that may be
benign. Two additional rare disorders also produce hypermethioninemia: glycine-N-methyltransferase deficiency
associated with liver disease (Luka et al, 2002; Mudd et al,
2001) and S-adenosylhomocysteine hydrolase deficiency,
which may result in developmental delay and hypotonia
(Baric et al, 2004). Furthermore, transient hypermethioninemia may occur in newborn infants. Confirmation
of the disorder requires quantitative amino acid analyses
of plasma and urine. In the infant with homocystinuria,
homocystine is usually detectable in plasma and urine,
plasma total homocysteine is increased as is methionine,
and cystine is reduced. In isolated hypermethioninemia,
methionine is markedly increased in plasma, but there
is no detectable homocystine in plasma or urine and the
plasma cystine concentration is normal. Hypermethioninemia secondary to liver disease owing to tyrosinemia type
I, or to nonspecific liver disease, is usually accompanied by
increased tyrosine.
The three urea cycle disorders currently screened by
MS/MS analysis are citrullinemia, argininosuccinic acidemia, and arginase deficiency. Citrullinemia and argininosuccinic acidemia produce hyperammonemia, often in the
neonatal period, accompanied by poor feeding, tachypnea,
lethargy, and vomiting. Respiratory alkalosis is characteristic. Severe hyperammonemia in the newborn is a medical
emergency and should trigger prompt consultation with
a metabolic specialist or referral to an NICU at a metabolic center. Discontinuation of protein and the provision of intravenous fluids with high caloric content are the
first steps to take. L-Arginine or L-citrulline, as well as
the “scavenger drugs” sodium phenylbutyrate and sodium
benzoate, may be administered. Hemodialysis might be
required to control the neurotoxic hyperammonemia,
which can lead to irreversible brain damage, coma, and
death. It is hoped that with early identification through
newborn screening, patients with urea cycle disorders will
be protected by presymptomatic therapy in the neonatal
period. Arginase deficiency can also present acutely with
hyperammonemia as described earlier, although more
frequently it manifests as developmental delay and spastic
diplegia in childhood with a milder degree of hyperammonemia (Crombez and Cederbaum, 2005).
Tyrosinemia type I is an amino acid disorder that can
be diagnosed by finding an elevation of succinylacetone
in MS/MS analysis (Allard et al, 2004). However, the
preanalytic processing required for succinylacetone is
more involved than that required for the amino acids and
acylcarnitines. As a result, succinylacetone is not currently
measured by all screening programs. These programs may
rely on elevations of tyrosine for identification of this disorder. Unfortunately, moderate elevations of tyrosine that
are transient occur frequently in neonates, especially those
who have low birthweights and are sick, necessitating frequent requests for repeated screening with virtually no
detection of tyrosinemia type I. Unfortunately moderate
transient elevations of tyrosine occur frequently in neonates, especially those who have low birthweights and are
sick, necessitating frequent requests for repeated screening. Virtually no cases of tyrosinemia type I have been
detected based on elevated tyrosine because almost all
infants with tyrosinemia type I have had normal tyrosine
levels when screened (Frazier et al, 2006). Consequently,
the newborn detection of tyrosinemia type I by a tyrosine marker alone is ineffective. Tyrosinemia type I leads
to liver and renal tubular disease and can later result in
hepatocellular carcinoma. It is treated with administration
of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC; Orfadin) and a diet low in phenylalanine
and tyrosine. Tyrosinemia types II and III are identified by
an increased level of tyrosine in newborn screening. Tyrosinemia type II can result in mental retardation, painful
hyperkeratoses, and keratoconjunctivitis, and it is treated
by a low tyrosine-phenylalanine diet. Tyrosinemia type
III seems to be benign, although developmental delay has
been reported occasionally (Ellaway et al, 2001).
Medium-Chain Acyl-CoA Dehydrogenase
Deficiency and Other Fatty Acid Oxidation
Disorders
The fatty acid oxidation disorders include those in which
the long-chain fatty acids cannot traverse the mitochondrial membranes to be oxidized within the mitochondrial
matrix (i.e., the carnitine palmitoyltransferase disorders)
and those that constitute defects in fatty acid oxidation per
se. In either category, the problem is the inability to fully
oxidize fatty acids. Fatty acid oxidation is essential to supply energy as adenosine triphosphate via the Krebs cycle
and as ketones in the presence of a low supply of glucose.
The disorders involving defective transport concern carnitine, whereas those with defective oxidation are named
according to the enzyme that is deficient (see Table 27-1).
The clinical consequence of these disorders is fasting intolerance resulting in hypoketotic hypoglycemia, lethargy,
hyperammonemia, metabolic acidosis, hepatomegaly, and
sometimes sudden death. Cardiomyopathy is an additional feature of very-long-chain acyl-CoA dehydrogenase
deficiency (VLCADD) and long-chain hydroxyacyl CoA
dehydrogenase deficiency (LCHADD). Each fatty acid
disorder is associated with a specific or almost specific
acylcarnitine pattern on MS/MS analysis.
The most common fatty acid oxidation disorder is
MCADD. Tragically, before newborn screening was available, this disorder was often diagnosed only retrospectively
after a sudden unexplained death, usually when postmortem
examination revealed a fatty liver. This devastating outcome and a frequency of 1:15,000 to 1:20,000 (Zytkovicz
CHAPTER 27 Newborn Screening
et al, 2001), comparable with that of PKU, made MCADD
the primary reason for the addition of MS/MS technology
to newborn screening. To reduce the rate of false-positive
results in screening for MCADD, some programs have
added molecular testing of the c.985A>G MCAD mutation
as second-tier screening. Because this mutation occurs in
as many as 90% of persons with MCADD, this additional
analysis of a newborn blood specimen with an elevation
of the octanoylcarnitine marker for MCADD substantially
improves the predictive value of the screening abnormality
(Zytkovicz et al, 2001).
The treatment for fatty acid oxidation disorders is
avoidance of fasting with high-carbohydrate, low-fat feedings and, of critical importance, prompt attention to acute
illnesses in which vomiting occurs. Carnitine supplementation may be beneficial. Medium-chain triglycerides (i.e.,
MCT oil) is given for the long-chain disorders VLCADD
and LCHADD. Any infant with a fatty acid oxidation disorder should be evaluated at a metabolic center. Most of
these disorders are treatable, but screening enables early
diagnosis and genetic counseling for the family even when
early treatment may not be effective, such as in neonatal carnitine palmitoyltransferase II deficiency (CPT-II)
(Albers et al, 2001a).
Organic Acid Disorders
Organic acid disorders are a heterogenous group of disorders with a combined frequency of approximately 1 in
50,000 (Zytkovicz et al, 2001). Many of them can be identified through MS/MS screening (see Table 27-1). The
marker for this disease group, as for the fatty acid oxidation disorders, is an abnormal acylcarnitine pattern. If a
screening result suggests an organic acidemia, a metabolic
specialist should be consulted immediately. The major
organic acid disorders identified in newborn screening are
propionic acidemia, the methylmalonic acidemias, and isovaleric acidemia.
The organic acidemias can manifest in the neonatal
period with a life-threatening, sepsis-like picture of feeding difficulties, lethargy, vomiting, and seizures. Metabolic
acidosis virtually always accompanies this presentation,
and hyperammonemia is common. In this situation, protein administration should be discontinued and replaced
by administration of intravenous fluids with high caloric
content and carnitine. The hyperammonemia rarely
requires specific treatment. The effects of early diagnosis
and treatment on the clinical and neurologic development
of individuals affected by an organic acid disorder are currently under investigation (Albers et al, 2001b).
Galactosemia
Galactosemia typically manifests in the neonatal period
as failure to thrive, vomiting, and liver disease (Hughes
et al, 2009). Death from bacterial sepsis, usually caused by
Escherichia coli, occurs in a high percentage of untreated
neonates (Levy et al, 1977). The average incidence of the
disorder is 1 in 62,000.
Some screening programs use a metabolite assay for
total galactose (galactose and galactose-1-phosphate) to
detect galactosemia. Other programs screen the newborn
323
specimen with a specific enzyme assay for activity of
GALT, undetectable in severe galactosemia. The enzyme
assay identifies only galactosemia, whereas the metabolite
assay also identifies other galactose metabolic disorders,
such as deficiencies of galactokinase and epimerase. Severe
neonatal liver disease and portosystemic shunting caused
by anomalies in the portal system can also increase the
galactose level.
The most rapid confirmatory test for a positive result in
galactosemia screening is urine testing for reducing substance. In almost all cases of severe galactosemia, this test
produces a strongly positive reaction. Galactosemia with
residual GALT activity, however, may be accompanied by
a negative or only slightly positive result for urine reducing
substance. If the urine contains reducing substance and the
infant has clinical signs of galactosemia (e.g., poor feeding,
jaundice, hepatomegaly), a blood specimen for confirmatory testing should be collected, and milk feeding (breast
or formula) should be discontinued with substitution of a
nonlactose formula such as soy or elemental. The infant
should then be referred immediately to a pediatric metabolic center, where confirmatory testing should include
the measurement of RBC galactose-1-phosphate and an
enzyme assay for RBC GALT activity (Levy and Hammersen, 1978). Molecular testing for GALT mutations
may also be performed. Approximately 70% of the patients
with galactosemia carry the Q188R mutation (Elsas and
Lai, 1998). Variant galactosemia is produced by mutations
such as S135L. The N314D mutation produces the benign
Duarte variant.
If the urine test is negative for reducing substance, the
newborn screening result is most likely to be false-positive or to indicate a benign GALT enzyme variant (e.g.,
Duarte variant). Nevertheless, urine-reducing substance
may be absent in infants with clinically significant variants
of galactosemia. Consequently, follow-up testing should
be performed for all infants with an initial positive galactosemia screening result.
Biotinidase Deficiency
Biotin recycling is necessary for the maintenance of sufficient intracellular biotin to activate carboxylase enzymes.
Biotinidase is a key enzyme in biotin recycling. Lack of
biotinidase activity results in reduced carboxylase activities and an organic acid disorder known as multiple carboxylase deficiency (Wolf and Heard, 1991). The clinical
features of the disorder are developmental delay, seizures,
hearing loss, alopecia, and dermatitis. The developmental
delay and seizures usually manifest at 3 to 4 months of age.
Death during infancy has also been reported.
Initiating biotin therapy in early infancy, when the disorder is presymptomatic, seems to prevent all the features
of biotinidase deficiency. For this reason, a screening test
has been developed and added to newborn screening in
a number of newborn screening programs throughout
the world (Hart et al, 1992). The frequency of identified newborns in these programs has a wide range, from
1:30,000 to 1:235,000. The average frequency is approximately 1 in 70,000. Almost all identified infants have
been asymptomatic and have remained normal with biotin treatment.
324
PART VII Care of the Healthy Newborn
ENDOCRINE DISORDERS
Congenital Hypothyroidism
Congenital hypothyroidism is the most common disorder identified by routine newborn screening. It is found
in 1:3000 to 1:5000 screened infants (Dussault, 1993).
The major clinical features of untreated congenital hypothyroidism are growth retardation and delayed cognitive
development leading to mental deficiency. If treatment
with pharmacologic doses of T4 is initiated early, growth
and mental development are normal.
Two screening approaches are used (Pass and Neto,
2009). One method is primary screening for low T4 with
secondary screening for high TSH. The second method is
primary screening for high TSH levels. Either procedure
reliably identifies congenital hypothyroidism. Nevertheless, affected infants can be missed with either approach.
This situation may be due to a lack of the identifying
marker abnormality at the time of specimen collection.
Specifically, the T4 level during the first 24 hours of life in
an affected infant might not yet be sufficiently decreased
for identification because of persistence of maternally
transmitted T4. Moreover, in the premature infant with
congenital hypothyroidism, it might take 2 weeks or more
for a TSH elevation to develop (Larson et al, 2003).
The reported false-positive rates of screening for congenital hypothyroidism range from approximately 0.05%
to as high as 4% (Pass and Neto, 2009). Infants with falsepositive results have transiently low T4 or elevated TSH
levels. Many of those with low T4 levels are premature
infants with a normal TSH concentration or infants with
perinatal stress and elevated TSH levels. To avoid missing
congenital hypothyroidism, screening programs require a
second blood specimen from each of these infants. In addition to false-positive results, a low T4 level with a normal
TSH value can result from benign T4-binding globulin
deficiency (Dussault, 1993; Mandel et al, 1993) or hypothyroidism secondary to pituitary deficiency.
Infants with a positive screening test result should not
be labeled as having congenital hypothyroidism until diagnostic testing confirms the disorder. This is especially
true if the TSH concentration reported by the screening
program is normal. If congenital hypothyroidism is confirmed, however, administration of T4 should be started
without delay to prevent irreversible brain damage.
Congenital Adrenal Hyperplasia
CAH caused by steroid 21-hydroxylase deficiency occurs in
1:16,000 to 1:20,000 births (White, 2009). Infants with the
salt-losing form of CAH can rapidly become hyperkalemic
and die precipitously, often without a specific diagnosis.
The clinical diagnosis may be suspected in the newborn
girl because of ambiguous genitalia. However, the diagnosis is usually not suspected in boys and in girls with atypical forms of CAH in which ambiguous genitalia may not
occur. Infant girls with ambiguous genitalia might not be
recognized as having CAH if the ambiguity is not obvious,
or they could be misassigned as boys if the ambiguity is
advanced. Because accurate gender assignment and initiation of hormone therapy as soon as possible are critical to a
favorable prognosis in CAH, newborn screening is important in leading to early diagnosis and prompt therapy with
pharmacologic doses of hydrocortisone. Consequently,
testing for CAH has been incorporated into routine newborn screening in most programs.
Screening is based on identifying elevated levels of
17-OHP, the preferred substrate for 21-hydroxylase, and
is usually measured using an immunoassay. Unfortunately,
compared with other neonatal screening assays, the specificity of the CAH assay is low and false-positive results in
newborn screening for CAH are relatively common, with
the rate often as high as 0.5%. The finding may be due to
a truly increased 17-OHP, as in perinatal stress and early
specimen collection (within the first 24 hours of life), or
to cross-reacting steroids, such as in prematurity and low
birthweight (al Saedi et al, 1996). Cross-reacting steroids
are produced by residual fetal adrenal cortex or result from
decreased metabolic clearance by an immature liver.
A decidedly increased level of 17-OHP suggests CAH,
and the infant should be referred to a pediatric endocrinologist for management. A second blood specimen
is usually requested from infants found to have slight to
moderately increased 17-OHP. If the infant shows signs
of illness or has ambiguous genitalia, serum electrolytes
should be measured. If these results indicate hyponatremia
and hyperkalemia, the infant should be hospitalized without delay, and the electrolyte imbalance should be corrected immediately. Pediatric endocrinology consultation
should also be sought. It may be possible to improve the
positive predictive value by second-tier screening using
DNA-based methods or liquid chromatography followed
by tandem mass spectrometry, but currently these methods are not widely used by newborn screening programs.
SICKLE CELL DISEASE
In most newborn screening programs in the United
States, the blood specimen is routinely tested for hemoglobin abnormalities. The major goal of this testing is to
identify infants with sickle cell disease so that they can
be given penicillin prophylaxis to prevent pneumococcal
septicemia. Additional benefits of early detection are early
referral to a comprehensive sickle cell program and early
education and genetic counseling for parents (Smith and
Kinney, 1993). Unfortunately, the long-term complications are not yet preventable.
Sickle cell screening is usually performed by means of
hemoglobin electrophoresis of blood eluted from a disc
of the Guthrie specimen. This procedure identifies sickle
cell disease, sickle cell trait, and several other abnormal
hemoglobins. Other than sickle cell disease, most of these
abnormalities are benign. It is especially critical to differentiate the common and benign sickle cell trait from the
much rarer sickle cell disease (homozygosity for S hemoglobin). For example, sickle cell disease affects approximately 1 in 600 African American persons, whereas sickle
cell trait (carrier status for S hemoglobin) is present in 1 in
12. Infants with sickle cell trait do not have complications
and should not be stigmatized as having sickle cell disease.
When sickle cell disease is confirmed, penicillin prophylaxis should be initiated as soon as possible, and the
infant should be referred to a sickle cell disease center or
CHAPTER 27 Newborn Screening
hematologist. The combination of screening and careful
follow-up has been highly effective in preventing pneumococcal sepsis in infants with sickle cell disease.
CYSTIC FIBROSIS
The frequency (1:2000 to 1:3000) and severity of cystic
fibrosis explain its inclusion in routine newborn screening. As with sickle cell disease, therapy that can prevent
the ultimate complications of cystic fibrosis is not yet available. However, early and usually presymptomatic diagnosis through screening leads to early nutritional therapy,
pancreatic enzyme replacement, and antibiotic prophylaxis
for pulmonary infection. Data from newborn screening
suggest better growth, prevention of vitamin deficiency in
early infancy, and some advantage in terms of pulmonary
status later in life in children identified by screening (Farrell et al, 2001; McKay and Wilcken, 2008; Southern et al,
2009). Other benefits of newborn screening are identifying the parents’ genetic potential for producing additional
children with cystic fibrosis and, through presymptomatic
identification, allowing the family to avoid months or years
of delay in the correct diagnosis of a child with chronic
respiratory problems or poor growth (Farrell and Mischler, 1992).
The analyte marker in newborn screening for cystic
fibrosis is increased immunoreactive trypsinogen (IRT).
Transient increases in IRT are common in healthy newborns as a result of perinatal stress or for unknown reasons. Consequently, the rate of false-positive results in
cystic fibrosis screening using only IRT is relatively high.
To reduce this rate, screening programs have adopted
a second-tier DNA analysis for a panel of cystic fibrosis
mutations when the specimen has increased IRT (Ferec
et al, 1995). Despite this two-tiered approach to screening, a substantial number of infants who do not have cystic fibrosis must undergo a sweat test or complete DNA
sequencing of the cystic fibrosis gene before the diagnosis
can be eliminated.
SPECIFIC ISSUES IN NEWBORN
SCREENING
CRITERIA FOR NEWBORN SCREENING
Under the auspices of the World Health Organization,
Wilson and Jungner (1968) published a set of criteria for
screening for disease that have generally been accepted as
required for population screening. The ten criteria state
that (1) the condition be an important health problem, (2)
there be accepted treatment, (3) facilities for diagnosis and
treatment be available, (4) there be a recognizable latent
or early symptomatic stage, (5) there should be a suitable
test, (6) the test should be acceptable to the population, (7)
the natural history of the condition should be understood,
(8) there should be a policy prescribing whom to treat, (9)
the cost of case finding should be economically balanced
in relation to medical care as whole, and (10) case-finding
should be a continuing process.
These criteria were developed at a time when newborn
screening was in its beginning stages and with screening
for adult disorders in mind. For example, regarding to the
325
first criterion, Wilson and Jungner recognized that the
term important is relative; whereas diabetes was prevalent,
treatment might not influence outcome while PKU was
rare, but it warranted screening because of the serious consequences that would be prevented by early discovery and
treatment.
There is a question as to the current relevance of the
Wilson-Jungner criteria for newborn screening. Ideally, all
the criteria should be applied to newborn screening. However, advances in technology have caused this application
to be questioned (Green and Pollitt, 1999; Levy, 1999). As
an example, screening using MS/MS allows detection of
serious disorders for which there may not be acceptable
or agreed upon therapy, or in which the natural history is
largely unknown, challenging the criterion that the disorder included in screening have acceptable treatment. How
is this dilemma resolved? The answer is not yet available.
It is hoped that the experience and findings from expanded
newborn screening will be used to develop a new set of
criteria that will apply to newborn screening. These criteria will likely retain the essence of the Wilson-Jungner
compilation, but with important modifications that could
be applied to any new screening venture.
FALSE-POSITIVE RESULTS
The majority of positive results in newborn screening,
particularly when this result is only mildly or moderately abnormal, are not due to a disorder. Unfortunately,
because screening is primarily based on a quantitative
measure, false-positive results must be addressed. Metabolite concentrations vary among individuals, and the distribution curves of the markers from affected and unaffected
populations are expected to be different (Figure 27-2).
In screening, a value that separates the two distribution
curves is established as a cutoff.
The cutoff values of the quantitative biomarkers are
established by the individual screening laboratories, and
they can vary among the different laboratories because
of variations in the testing technology. In general, the
cutoff value should be set such that it is greater than the
99th percentile of the concentration in normal neonates
and less than the 5th percentile of the concentration in
affected neonates (Rinaldo et al, 2006). However, for disorders that are extremely rare, the population of affected
neonates may be so small that establishing an appropriate cutoff becomes a challenge. In such cases, the laboratory may empirically set a cutoff at 3 to 4 SD from the
population mean and adjust the values with experience to
minimize the false positives without compromising the
sensitivity. Specimens in which the concentration crosses
the established cut-off are considered screen-positive.
The metabolite concentration in a majority of unaffected
individuals is below the cutoff value, but in a small proportion it crosses this threshold. The positive screening
results that are not caused by a disorder are considered
false positives. Because of some degree of overlap in the
distribution curves of the affected and unaffected populations, these false positives cannot be entirely eliminated
without compromising the sensitivity of screening. Furthermore other physiologic factors, such as immaturity of
enzymes or stress and therapeutic interventions, can skew
326
PART VII Care of the Healthy Newborn
Normal Population
Affected
Unaffected
TP
FN
FP
TN
Test Positive
Test Negative
Number of Neonates
True Negatives
Sensitivity [TP] / [TP FN]
Specificity [TN] / [TN FP]
Positive Predictive Value [TP] / [TP FP]
False Positives
Affected
False Negatives
True
Positives
Mean
(Normal)
Cut-Off
Mean
(Affected)
Analyte Concentration
FIGURE 27-2 Distribution of quantitative markers measured in screening. For most conditions, the distribution in the unaffected or normal
population overlaps that in the affected individuals. The number of false positives (FP), false negatives (FN), true positives (TP), and true negatives
(TN) depends on the established cutoff.
the concentrations of certain metabolites and lead to falsepositive screening results. Currently, indicators measured
using immunoassays (e.g.,17-OHP in CAH and T4 in congenital hypothyroidism) or enzymatic activity (low GALT
activity in galactosemia) are associated with the highest
false-positive rates.
The false-positive results are more common in preterm
and low-birthweight infants than in full-term infants. For
example, up to 85% of preterm infants have transiently
low T4 levels (Paul et al, 1998). Transient increases in
17-OHP are another common abnormality in infants who
are preterm or have low birthweights or have experienced
perinatal stress (Pang and Shook, 1997). In addition, transient tyrosinemia is commonly observed in preterm and
low-birthweight infants, although it can also occur in fullterm infants (Levy et al, 1969).
Artifacts produced in the collection or transport of the
Guthrie specimen account for some false-positive results.
As mentioned in the discussion of the specimen collection procedure, collection of the specimen from a central
line can result in mixing with amino acids in total parental
nutrition solution and a false increase of amino acids in the
specimen. Contamination with milk (or any drink containing milk) can result in a false elevation in galactose and the
mistaken suspicion of galactosemia. Prolonged exposure
to heat can reduce the activity of GALT in the specimen
and produce a false impression of galactosemia when the
enzyme assay is used to screen for this disorder. This error
is common during the summer, especially when the specimens remain in a mailbox for a period of time. Some factors known to be associated with false positives are shown
in Table 27-2 (Sahai et al, 2009).
With the substitution of MS/MS for the traditional bacterial or specific assays, such as those for PKU and MSUD,
the number of false-positive results is distinctly lower. For
PKU screening, the false-positive rate is reported at 0.05%
compared with 0.23% by earlier methods (Levy, 1998). In
addition, with MS/MS the multiplexed approach allows
for a profile of analytes rather than a single analyte level
(e.g., phe/tyr ratio vs. only a phenylalanine level for PKU
identification), further improving the specificity of screening (Chace et al, 1998; Schulze et al, 1999). Therefore
the average positive predictive value for primary markers analyzed by MS/MS, previously reported to be 8%
to 10% (Schulze et al, 2003; Wilken et al, 2003), can be
improved substantially if the marker is evaluated in context
with other metabolites that are screened or when a tiered
approach is applied (Frazier et al, 2006; Sahai et al, 2007).
Second-tier assays, such as molecular assays for cystic
fibrosis or secondary immunoassays for hypothyroidism,
are commonly performed to reduce the false-positive rates
for primary markers analyzed by immunoassays or enzymatic assays. Nevertheless, false-positive results cannot be
entirely eliminated; therefore it is important to reassure
the parents that not every abnormal result of newborn
screening inevitably implies a disorder and that transient
or nonspecific abnormalities are common. Although all
infants with an abnormal screening result must undergo
repeated testing, the families should be informed that an
initial positive result might have no medical implications.
This approach can alleviate excessive anxiety and prevent
unnecessary diagnostic procedures and treatment.
INCREASED DETECTION BY SCREENING
For certain disorders screened by MS/MS, many more
infants are identified than the expected numbers based
on clinical identification. The greatest increases are in
the fatty acid oxidation disorders, including short-chain
acyl-CoA dehydrogenase deficiency SCADD, MCADD,
CHAPTER 27 Newborn Screening
VLCADD, and carnitine deficiency, and in three organic
acid disorders (glutaric acidemia type I, 3-methylcrotonylCoA carboxylase deficiency, and 3-ketothiolase deficiency;
Wilken et al, 2003).
Although some of the excess detection by screening
could represent cases that were symptomatic but not diagnosed clinically, it is likely that the greater part of the excess
represents infants with benign or milder forms of the disorders who would not come to clinical attention. Notably,
Spiekerkoetter et al (2003) identified a frequent mutation in asymptomatic patients with VLCADD detected
by newborn screening, and Ensenauer et al (2004) found
a common, mild, and perhaps asymptomatic mutation in
patients with isovaleric acidemia identified by screening.
MISSED CASES
Infants with congenital hypothyroidism, PKU, intermittent MSUD, glutaric acidemia, tyrosinemia I, and other
screened disorders have been undetected by newborn
screening. Laboratory or program errors were reported as
the most common cause of these missed cases (Holtzman
et al, 1974). In some instances, a specimen was never collected, such as when infants were transferred to another
hospital. However, in screening for a multitude of disorders, each with its own biomarker that varies with time
and physiologic states, an occasional affected neonate may
have normal biomarker concentrations in the newborn
specimen simply because the timing of collection was not
ideal for the particular condition. Therefore physicians
must exercise clinical judgment and not fall into the trap
of excluding a diagnosis because an infant has presumably
been screened. Specific testing for metabolic and endocrine disorders should be performed in any infant or child
with symptoms that suggest the presence of such a disorder, regardless of the assumed or actual newborn screening
result.
THE FUTURE
Many factors are impinging on newborn screening, including rapidly advancing technology, new and increasingly
available therapeutic approaches to previously untreatable
disorders, and family support groups and influential citizens or legislators who are heavily invested in individual
disorders that may or may not be ready for inclusion as
screening tests. To address these pressures, a committee
convened by the American College of Medical Genetics
327
and the Maternal and Child Health Division of the U.S.
Department of Health and Human Services meets regularly to study newly proposed newborn screening tests
(and those currently recommended) to judge their suitability for inclusion in the screening panels. One new candidate for screening severe combined immunodeficiency
has passed through a careful vetting process and has been
recommended for inclusion in the standard panel. Other
candidate conditions being considered are the lysosomal
storage disorders and adrenoleukodystrophy, for which
enzyme or early bone marrow therapies are available and
are being studied, Fragile X syndrome, and the SmithLemli-Opitz syndrome. These disorders will be judged on
the basis of frequency, severity, availability of preemptive
therapies, and the cost and robustness of the screening test
itself. Some disorders have been included in isolated state
panels on the basis of the political influences mentioned
above.
Finally, there is the issue of retention of blood spots for
future study. Despite multiple safeguards to protect the
identity and anonymity of individuals, parents and civil
libertarians are concerned that retention of these blood
spots poses a threat to the privacy of individuals and that
the specimens should be destroyed. If this view prevails,
a resource of great value in the development of new and
more effective tests, and one that is increasingly recognized as the avatar of personalized medicine, will be lost.
SUGGESTED READINGS
American College of Medical Genetics: Newborn Screening Expert Group.
Newborn screening: toward a uniform panel and system, Genet Med, 2006.
Clinical and Laboratory Standards Institute: Newborn screening for preterm, low birth
weight and sick newborns: approved guideline. CLSI document I/LA31-A, Wayne,
Penn, 2009, Clinical and Laboratory Standards Institute.
Dobrowolski SF, Banas RA, Naylor EW, et al: DNA microarray technology for
neonatal screening, Acta Paediatr Suppl 88:61-64, 1999.
Kaye CI, Accurso F, La Franchi S, et al: Newborn screening fact sheets, Pediatrics
118:e934-e963, 2006.
Matern D, Tortorelli S, Oglesbee D, et al: Reduction of the false-positive rate in
newborn screening by implementation of MS/MS-based second-tier tests: the
Mayo Clinic experience (2004-2007), J Inherit Metab Dis 30:585-592, 2007.
Pass KA, Neto EC: Update: newborn screening for endocrinopathies, Endocrinol
Metab Clin North Am 38:827-837, 2009.
Rashed MS, Ozand PT, Bucknall MP, et al: Diagnosis of inborn errors of metabolism from blood spots by acylcarnitines and amino acids profiling using automated electrospray tandem mass spectrometry, Pediatr Res 38:324-331, 1995.
Sahai I, Marsden D: Newborn screening, Crit Rev in Cli Lab Sci 46:55-82, 2009.
Smith J, Kinney T: Clinical practice guidelines, quick reference guide for clinician:
sickle cell disease: screening and management in newborns and infants, Am
Fam Physician 48:95-102, 1993.
Wilken B, Wiley V, Hammond J, et al: Screening newborns for inborn errors of
metabolism by tandem mass spectrometry, N Engl J Med 348:2304-2312, 2003.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
28
Resuscitation in the Delivery Room
Tina A. Leone and Neil N. Finer
The transition from fetal to neonatal life is a dramatic and
complex process involving extensive physiologic changes
that are most obvious at the time of birth. Individuals who
care for newly born infants must monitor the progress of
the transition and be prepared to intervene when necessary. In the majority of births this transition occurs without a requirement for any significant assistance. However,
when the need for intervention arises, the presence of providers skilled in neonatal resuscitation can be life saving.
Each year approximately 4 million children are born in
the United States (Martin et al, 2008) and more 30-fold
as many are born worldwide. It is estimated that approximately 5% to 10% of all births will require some form of
resuscitation beyond basic care, making neonatal resuscitation the most frequently practiced form of resuscitation
in medical care. Throughout the world approximately 1
million newborn deaths are associated with birth asphyxia
(Lawn et al, 2005). Whereas early effective newborn resuscitation will not eliminate all early neonatal mortality, such
intervention will save many lives and significantly reduce
subsequent morbidities.
Attempts at reviving nonbreathing infants immediately after birth have occurred throughout recorded time
with references in literature, religion, and early medicine. Although the organization and sophistication have
changed, the basic principle and goal of initiating breathing
have remained constant. During the last 20 years, attention has focused on the process of neonatal resuscitation.
Resuscitation programs in other areas of medicine were
initiated in the 1970s in an effort to improve knowledge
about effective resuscitation and provide an action plan for
early responders. The first such program was focused on
adult cardiopulmonary resuscitation (Kattwinkel, 2006).
These programs then began increasing in complexity and
becoming more specific to different types of resuscitation
needs. With the collaboration of the American Heart Association and the American Academy of Pediatrics, the Neonatal Resuscitation Program (NRP) was initiated in 1987
and was designed to address the specific needs of the newly
born infant. The NRP textbook (Kattwinkel, 2006) now
includes specific recommendations for the preterm infant.
Various groups throughout the world also provide resuscitation recommendations that are more specific to the practices in certain regions. An international group of scientists,
the International Liaison Committee on Resuscitation
meets on a regular basis to review available resuscitation
evidence for all the different areas of resuscitation and puts
forth a summary of its review (Chamberlain, 2005).
The overall goal of the NRP is similar to other resuscitation programs, in that it intends to teach large groups
of individuals of varying backgrounds the principles of
resuscitation and provide an action plan for providers.
Similarly, a satisfactory end result of resuscitation would
be common to all forms of resuscitation, namely to provide
328
adequate tissue oxygenation to prevent tissue injury and
restore spontaneous cardiopulmonary function. However,
when comparing neonatal resuscitation with other forms
of resuscitation, several distinctions can be noted. First,
the birth of an infant is a more predictable occurrence
than most events that require resuscitation in an adult,
such as an arrhythmia or a myocardial infarction. Although
not every birth will require resuscitation, it is more reasonable to expect that skilled individuals can be present
when the need for neonatal resuscitation arises. It is possible to anticipate with some accuracy which newborns will
more likely require resuscitation based on perinatal factors
and thus allow time for preparation. The second distinction of neonatal resuscitation compared with other forms
of resuscitation involves the unique physiology involved
in the normal fetal transition to neonatal life. The fetus
exists in the protected environment of the uterus where
temperature is closely controlled, the lungs are filled with
fluid, continuous fetal breathing is not essential, and the
gas exchange organ is the placenta. The transition that
occurs at birth requires the newborn infant to increase
heat production, initiate continuous breathing, replace the
lung fluid with air, and significantly increase pulmonary
blood flow so that gas exchange can occur in the lungs.
The expectations for this transitional process and knowledge of how to effectively assist the process help to guide
the current practice of newborn resuscitation.
FETAL TRANSITION TO
EXTRAUTERINE LIFE
The complete transition from fetal to extrauterine life is
complex and much more intricate than can be discussed in
these few short paragraphs, but a basic knowledge of these
processes will contribute to the understanding of the rationale for resuscitation practices. The key elements necessary for a successful transition to extrauterine life involve
changes in thermoregulation, respiration, and circulation.
In utero the fetal core temperature is approximately 0.5° C
greater than the mother’s temperature (Gunn and Gluckman, 1983). Heat is produced by metabolic processes and
is lost over this small temperature gradient through the
placenta and skin (Gilbert et al, 1985). After birth the temperature gradient between the infant and the environment
becomes much greater and heat is lost through the skin
by radiation, convection, conduction, and evaporation.
The newborn infant must begin producing heat through
other mechanisms such as lipolysis of brown adipose tissue
(Dawkins and Scopes, 1965). If heat is lost at a pace greater
than it is produced, the infant will develop hypothermia.
Preterm infants are at particular risk because of increased
heat loss through immature skin, a greater surface area–
to–body weight ratio, and decreased brown adipose tissue
stores.
329
CHAPTER 28 Resuscitation in the Delivery Room
The fetus lives in a fluid-filled environment, and the
developing alveolar spaces are filled with lung fluid. Lung
fluid production decreases in the days before delivery
(Kitterman et al, 1979), and the remainder of lung fluid
is reabsorbed into the pulmonary interstitial spaces after
delivery (Bland, 1988). As the infant takes the first breaths
after birth, a negative intrathoracic pressure of approximately 50 cm H2O is generated (Vyas et al, 1986). The
alveoli become filled with air, and with the help of pulmonary surfactant the lungs retain a small amount of air
persisting at the end of exhalation, which is known as the
functional residual capacity (FRC). Although the fetus makes
breathing movements in utero, these efforts are intermittent and are not required for fetal gas exchange. Continuous spontaneous breathing is maintained after birth by
several mechanisms, including the activation of chemoreceptors, the decrease in hormones that inhibit respirations,
and the presence of natural environmental stimulation.
Spontaneous breathing can be suppressed at birth for
several reasons, most critical of which is the presence of
acidosis secondary to compromised fetal circulation. The
natural history of the physiologic responses to asphyxia
and acidosis has been described by researchers evaluating
animal models. Geoffrey Dawes described the breathing
response to acidosis in different animal species (Dawes,
1968). He noted that when pH was decreased, animals
typically had a relatively short period of apnea followed
by gasping. The gasping pattern then increased in rate
until breathing ceased again for a second period of apnea.
The physiologic effects that occur with worsening acidosis
are noted in Figure 28-1. Dawes also noted that the first
period of primary apnea could be reversed with stimulation, whereas the second period, secondary or terminal
apnea, required assisted ventilation to ultimately establish
spontaneous breathing. The first sign of improvement was
noted to be an increase in heart rate. Further recovery
was noted when the newborn begins gasping again. The
secondary period of apnea varies in duration depending
on the duration of asphyxia and degree of acidosis. In the
clinical situation, the exact timing of onset of acidosis is
generally unknown; therefore any observed apnea may be
either primary or secondary. This finding is the basis of
the resuscitation recommendation that stimulation may be
attempted in the presence of apnea, but if not quickly successful, assisted ventilation should be initiated promptly.
Without the presence of acidosis, a newborn may also
develop apnea because of recent exposure to respiratory
suppressing medications such as narcotics, anesthetics,
and magnesium. These medications cross the placenta
when given to the mother and may depress the newborn’s
respiratory drive, depending on the time of administration
and dose.
Fetal circulation is unique because gas exchange takes
place in the placenta. In the fetal heart, oxygenated blood
returning via the umbilical vein is mixed with deoxygenated blood from the superior and inferior venae cavae, and
it is distributed differentially throughout the body. The
most oxygenated blood is directed toward the brain, and
the most deoxygenated blood is directed toward the placenta. Thus blood returning from the placenta to the right
atrium is preferentially streamed via the foramen ovale
to the left atrium and ventricle and then to the ascending
RESPIRATORY EFFORT
HEART RATE
AORTIC BLOOD PRESSURE
CENTRAL VENOUS PRESSURE
CARDIAC OUTPUT
BLOOD FLOW, HEAD & HEART
BLOOD FLOW, BODY
BLOOD FLOW, LUNGS
BRAIN DAMAGE
TIME
pH
7.4
7.1
7.0
6.7
ASPHYXIA
RESUSCITATION
FIGURE 28-1 The sequence of cardiopulmonary changes with
asphyxia and resuscitation. Time is on the horizontal axis. Asphyxia
progresses from left to right; resuscitation proceeds from right to left.
Units of time are not given. If there is complete interruption of respiratory gas exchange, the entire process of asphyxia from extreme left to
right could occur in approximately 10 minutes. It could take much longer with an asphyxiating process that only partly interrupts gas exchange
or does so completely, but only for repeated brief periods. With resuscitation, the process reverses, beginning at the point to which asphyxia
has proceeded. (Adapted from Dawes G: Foetal and neonatal physiology,
Chicago, 1968, Year Book; and Avery GN: Neonatology, Philadelphia,
1987, JB Lippincott.)
aorta, providing the brain with the most oxygenated blood.
Fetal channels, including the ductus arteriosus and foramen ovale, allow most blood flow to bypass the lungs with
their intrinsically high vascular resistance. As a result, pulmonary blood flow is approximately 8% of the total cardiac output. In the mature postnatal circulation, the lungs
must receive 100% of the cardiac output. When the lowresistance placental circulation is removed after birth, the
infant’s systemic vascular resistance increases while the
pulmonary vascular resistance begins to fall as a result of
pulmonary expansion, increased arterial and alveolar oxygen tension, and local vasodilators. These changes result in
a dramatic increase in pulmonary blood flow. The average
fetal oxyhemoglobin saturation as measured in fetal lambs
is approximately 50% (Nijland et al, 1995), but ranges in
different sites within the fetal circulation between values of
20% and 80% (Teitel, 1988). The oxyhemoglobin saturation rises gradually over the first 5 to 15 minutes of life
to 90% or greater as the air spaces are cleared of fluid.
A diagram of the blood flow patterns in the fetus and normally transitioning newborn is shown in Figures 28-2 and
28-3. In the face of poor transition secondary to asphyxia,
330
PART VII Care of the Healthy Newborn
FIGURE 28-2 Fetal circulation. Oxygenated blood leaves the
placenta by way of the umbilical vein (vessel without stippling). The
blood flows into the portal sinus in the liver (not shown), and a variable
portion of it perfuses the liver. The remainder passes from the portal
sinus through the ductus venosus into the inferior vena cava, where it
joins blood from the viscera (represented by the kidney, gut, and skin).
Approximately half of the inferior vena cava flow passes through the
foramen ovale to the left atrium, where it mixes with a small amount of
pulmonary venous blood. This relatively well-oxygenated blood (light
stippling) supplies the heart and brain by way of the ascending aorta.
The other half of the inferior vena cava stream mixes with superior vena
cava blood and enters the right ventricle (blood in the right atrium and
ventricle has little oxygen, which is denoted by heavy stippling). Because
the pulmonary arterioles are constricted, most of the blood in the main
pulmonary artery flows through the ductus arteriosus (DA), so that the
descending aorta’s blood has less oxygen (heavy stippling) than blood
in the ascending aorta (light stippling). (From Avery GN: Neonatology,
Philadelphia, 1987, JB Lippincott.)
meconium aspiration, pneumonia, or extreme prematurity, the lungs might not be able to develop efficient
gas exchange; therefore the oxygen saturation might not
increase as expected. In addition, in some situations the
normal reduction in pulmonary vascular resistance does
not fully occur, resulting in persistent pulmonary hypertension and decreased effective pulmonary blood flow with
continued right-to-left shunting through the aforementioned fetal channels. Right-to-left shunting will lead to
persistent hypoxemia and potentially to significant newborn illness requiring intensive care until the circulatory
pattern adjusts to extrauterine life. The circulatory pattern
associated with poor transition is noted in Figure 28-4.
FIGURE 28-3 Circulation in the normal newborn. After expansion
of the lungs and ligation of the umbilical cord, pulmonary blood flow
increases and left atrial and systemic arterial pressures increase while
pulmonary arterial and right heart pressures decrease. When the left
atrial pressure exceeds right atrial pressure, the foramen ovale closes
so that all of the inferior and superior vena cava blood leaves the right
atrium, enters the right ventricle, and is pumped through the pulmonary
artery toward the lung. With the increase in systemic arterial pressure
and decrease in pulmonary arterial pressure, flow through the ductus
arteriosus becomes left to right, and the ductus constricts and closes.
The course of the circulation is the same as in the adult. (From Avery
GN: Neonatology, Philadelphia, 1987, JB Lippincott.)
ENVIRONMENT AND PREPARATION
The environment in which the infant is born should facilitate the transition to neonatal life as much as possible and
should be able to readily accommodate the needs of a
resuscitation team when necessary. Hospitals vary in the
approach to the details of how to prepare for resuscitation. For example, some hospitals have a separate room
designated for resuscitation where the infant will be taken
after birth, whereas others have the delivery room adjacent
to the neonatal intensive care unit (NICU) and the infant
is resuscitated in the NICU if necessary. Hospitals may
bring all the necessary equipment into the delivery room
when resuscitation is expected or have every delivery room
equipped for any resuscitation. Wherever the resuscitation
will take place, a few key elements must be considered.
The room should be warm enough to prevent excessive
newborn heat loss, bright enough to assess the infant’s
clinical status, and large enough to accommodate the necessary personnel and equipment to care for the baby.
When no added risks to the newborn are identified, the
term birth frequently occurs without the attendance of a
CHAPTER 28 Resuscitation in the Delivery Room
331
TABLE 28-1 Risk Factors for Neonatal Resuscitation
Maternal
Factors
Fetal Factors
Intrapartum Factors
Maternal
hypertension
Preterm
delivery
Opiates in labor
Maternal
infection
Breech
presentation
Rupture of membranes >18
hours
Multiple
gestation
(preterm)
Shoulder
dystocia
Meconium-stained amniotic
fluid
Nonreassuring fetal heart
rate patterns
Emergency cesarean section
Prolapsed cord
Data from Aziz K et al: Ante- and intra-partum factors that predict increased need for
neonatal resuscitation, Resuscitation 79:444-452, 2008.
FIGURE 28-4 Circulation in an asphyxiated newborn with
incomplete expansion of the lungs. Pulmonary vascular resistance is
high, pulmonary blood flow is low (normal number of pulmonary veins),
and flow through the ductus arteriosus is high. With little pulmonary
arterial flow, left atrial pressure decreases below right atrial pressure, the
foramen ovale opens, and vena cava blood flows through the foramen
into the left atrium. Partially venous blood goes to the brain via the
ascending aorta. The blood of the descending aorta that goes to the
viscera has less oxygen than that of the ascending aorta (heavy stippling),
because of the reverse flow through the ductus arteriosus. Therefore the
circulation is the same as in the fetus, except that there is less well-oxygenated blood in the inferior vena cava and umbilical vein. (From Avery
GN: Neonatology, Philadelphia, 1987, JB Lippincott.)
specific neonatal resuscitation team. However, it is recommended that one individual be present who is responsible
only for the infant and can quickly alert a neonatal resuscitation team if necessary. Even the best neonatal resuscitation triage systems will not anticipate the need for
resuscitation in all cases. Using a retrospective risk assessment scoring system, 6% of newborns requiring resuscitation were not be identified based on risk factors (Smith et
al, 1985). Similarly, a recent review found that when a riskbased determination of neonatal resuscitation team attendance at deliveries was used, 22% of infants at attended
deliveries required at least assisted ventilation (Aziz et al,
2008). These investigators found that the most significant
risk factors were preterm birth, emergency cesarean section, and meconium-stained amniotic fluid. Other significant risk factors for the need for resuscitation are listed
in Table 28-1. Antenatal determination of risk allows the
resuscitation team to be present for the delivery and to be
more thoroughly prepared for the situation.
The composition of the neonatal resuscitation team will
vary tremendously among institutions. Probably the most
important factor in how well a team functions is how the
group has prepared for the delivery. Preparation involves
both the immediate tasks of readying equipment and personnel for an individual situation as well as the more broad
institutional preparation of training team members and
providing appropriate space and equipment. We believe
that when there is a strong suspicion that the newborn
infant will be born in a compromised state, a minimum
of three team members should be present, including one
member with significant previous experience leading
neonatal resuscitations. Each team member is assigned
tasks that are performed on a regular basis. The leader
is expected to ensure that the appropriate interventions
are performed and that they are performed well. All team
members are encouraged and expected to speak up if a
problem is noticed or if they believe an alternate course
would be beneficial. It seems logical that teams that regularly work together and divide tasks in a routine manner
will have a better chance of functioning smoothly during
a critical situation. Institutions can facilitate team readiness with regular review of practices and mock codes or
simulator training to practice uncommon scenarios. In our
institution, University of California San Diego (UCSD),
video-taped resuscitations are reviewed twice monthly
with representatives from all disciplines involved in the
resuscitation team (Carbine et al, 2000); this is done as a
quality-assurance procedure and allows for ongoing identification of areas needing improvement. In addition, this
practice provides an opportunity for education and discussion about potential solutions to repetitive problems
of newborn resuscitation. UCSD also instituted a supplemental training program for pediatric trainees to obtain
experience in a preclinical situation (Garey et al). These
training sessions allow adequate time to review scenarios
in detail, and trainees are given the opportunity to prepare
and operate the equipment and practice procedures on an
individualized basis. Others have used simulators to provide additional resuscitation training (Halamek, 2008). All
of these training elements help to prepare teams for future
resuscitations.
332
PART VII Care of the Healthy Newborn
When preparing for a resuscitation that will occur in
the immediate short-term period, additional aspects must
be addressed. The most important element in preparing
for the resuscitation is the discussion that occurs among
the team members who will participate. This discussion,
known as a prebrief, is an essential part of improving communication during resuscitation and involves reviewing the
clinical information, reviewing the available resources and
introducing the individuals who will participate, assigning tasks to individuals, and making equipment as readily available as possible. A discussion of the overall plan
will help team members know what to expect and how to
address problems that could occur. We have formalized
this process by creating a prebrief checklist for the team
to review while preparing for the delivery. Checklists are
considered one of the most important methods of preventing medical errors. A briefing after the resuscitation (i.e.,
a postbrief) allows team members to address positive and
negative aspects of the resuscitation. This briefing is an
important element in the preparation for future resuscitations, because team members can immediately identify
how to improve the care being provided. An example of
our current checklist with prebrief and postbrief documentation is shown in Figure 28-5. To better train the
staff in these techniques, UCSD instituted training in
crew resource management, a methodology developed for
training air crews from the late 1970s that evolved from a
careful evaluation of the role of human error in air crashes
(Cooper et al, 1980).
ASSESSMENT
Immediately after birth the infant’s condition is evaluated by general observation and measurement of specific
parameters. Typically a healthy newborn will cry vigorously and maintain adequate respirations. The color will
transition from blue to pink over the first 2 to 5 minutes,
the heart rate will remain in the range of 140 to 160 beats/
min, and the infant will demonstrate adequate muscle tone
with some flexion of the extremities. The overall assessment of an infant who is having difficulty with the transition to extrauterine life will often reveal apnea, bradycardia,
hypotonia, and cyanosis or pallor. After the initial steps of
resuscitation, interventions are based mainly on the evaluation of respiratory effort and heart rate, which need to be
assessed continually throughout the resuscitation. Heart
rate can be monitored by auscultation or palpation of the
cord pulsations, with auscultation being the more reliable
method (Owen and Wyllie, 2004). The continuous monitoring of this parameter gives providers an ongoing indication of the effectiveness of interventions.
The use of more sophisticated monitoring devices, such
as a pulse oximeter or electrocardiographic leads, can be
helpful during resuscitation. A pulse oximeter can provide
the resuscitation team with a continuous audible and visual
indication of the newborn’s heart rate throughout the various steps of resuscitation while allowing all team members
to perform other tasks. In addition, the pulse oximeter can
be used as a more accurate measure of oxygenation than the
evaluation of color alone, which is an unreliable measure
of the infant’s oxygen saturation (O’Donnell et al, 2007).
Whenever interventions beyond brief positive-pressure
ventilation by mask are required, a pulse oximeter should
be considered. Electrocardiographic monitoring may be
helpful, but at this point has been infrequently evaluated in
the delivery room and does not provide information about
oxygenation.
The overall assessment of a newborn was quantified by
Virginia Apgar in the 1950s with the Apgar score (Apgar,
1953). The score describes the infant’s condition at the
time it is assigned and consists of a 10-point scale, with
a maximum of 2 points assigned for each of the following categories: respirations, heart rate, color, tone, and
reflex irritability. Table 28-2 shows the detailed components of the score. Although the components of the score
include items that are assessed for determining interventions, the score itself is not used to determine the need for
interventions. The score was initially intended to provide
a uniform, objective assessment of the infant’s condition
and was used as a tool to compare different practices, especially obstetric anesthetic practices. Despite the intent of
objectivity, there is often disagreement in score assignment among various practitioners (O’Donnell et al, 2006).
Low scores are associated with increased risk of neonatal
mortality (Casey et al, 2001), but have not been predictive
of neurodevelopmental outcome (Nelson and Ellenberg,
1981). Interpreting the score when interventions are being
provided may be difficult, and current recommendations
suggest that clinicians should document the interventions
used at the time the score is assigned (American Academy of Pediatrics Committee on Fetus and Newborn and
American College of Obstetricians and Gynecologists and
Committee on Obstetric Practice, 2006).
INITIAL STEPS: TEMPERATURE
MANAGEMENT AND MAINTAINING
THE AIRWAY
In the first few seconds after birth, all infants are evaluated
for signs of life and a determination of the need for further
assistance is made. This evaluation is done both formally
as described in the NRP and informally as the initial care
providers observe the infant in the first few moments of
life. When the determination is made that further assistance and formal resuscitation are necessary, the infant is
then placed on a radiant warmer and positioned appropriately for resuscitation to proceed. Appropriate positioning
includes placing the infant supine on the warmer in such a
way that care providers have easy access. In addition, the
head should be in a neutral or “sniffing” position to facilitate maintenance of an open airway. Frequently the oropharynx contains fluid that can be removed by suctioning
with a standard bulb syringe.
An infant born through meconium-stained amniotic
fluid is at risk for aspirating meconium and developing
significant pulmonary disease, known as meconium aspiration syndrome, which may also be accompanied by persistent pulmonary hypertension. For many years, routine
management of all infants with meconium-stained amniotic fluid included suctioning of the mouth and pharynx
once the head was delivered, but before the shoulders
were delivered, and endotracheal intubation and tracheal
suctioning in an attempt to remove any meconium from
the trachea and prevent the development of meconium
CHAPTER 28 Resuscitation in the Delivery Room
333
FIGURE 28-5 Delivery room resuscitation checklist form.
TABLE 28-2 The Apgar Score
Feature
Evaluated
0 Points
1 Point
2 Points
Heart rate
(beats/min)
0
<100
>100
Respiratory
effort
Apnea
Irregular, shallow, or
gasping respirations
Vigorous
and crying
Color
Pale, blue
Pale or blue extremities
Pink
Muscle tone
Absent
Weak, passive tone
Active
movement
Reflex
irritability
Absent
Grimace
Active
avoidance
aspiration syndrome. Routine suctioning of the mouth and
nose before delivery of the shoulders has been shown to
be of no benefit in decreasing the incidence of meconium
aspiration syndrome, ventilation for meconium aspiration
syndrome, or mortality (Vain et al, 2004). Recognizing
that intubation may not be necessary for all infants and
that the procedure may be associated with complications, a
more selective approach was evaluated in randomized controlled trials (Wiswell et al, 2000). A metaanalysis of studies that have evaluated this question supported the notion
that universal endotracheal suctioning does not result in
a lower incidence of meconium aspiration syndrome or
improve mortality compared with selective endotracheal
334
PART VII Care of the Healthy Newborn
suctioning (Halliday, 2000). The likelihood that an infant
with meconium-stained amniotic fluid will develop meconium aspiration syndrome is increased in the presence of
fetal distress. The selective approach to endotracheal suctioning requires a quick evaluation of the infant after delivery. If the infant is vigorous with good respiratory effort,
normal heart rate, and normal tone, the steps of resuscitation proceed as usual. However, if the infant is not vigorous and has poor respiratory effort, a heart rate less than
100 beats/min, or decreased tone, rapid endotracheal intubation for suctioning is currently indicated, although this
has not been evaluated in a standardized manner. When
endotracheal intubation for suctioning is performed, the
resuscitation team must continue to adequately monitor
the infant and may need to proceed quickly to providing
assisted ventilation if bradycardia is present.
Whereas temperature control is important for all newborn infants, it is particularly important for the extremely
preterm infant. Preterm infants are commonly admitted
to the NICU with core temperatures well below 37° C.
In a population-based analysis of all infants younger than
26 weeks’ gestation, more than one third of these preterm infants had an admission temperature of less than
35° C (Costeloe et al, 2000). More disturbing is the fact
that these infants with hypothermia on admission survived
less often than those with admission temperatures greater
than 35° C. Admission temperatures can be improved in
preterm infants by immediately covering the infant’s body
with polyethylene wrap before drying the infant (Vohra et
al, 2004). With this approach the infant’s head is left out
of the wrap and is dried, but the body is not dried before
wrap application. Other measures for maintaining infant
temperatures include performing resuscitations in a room
that is kept at an ambient temperature of approximately
27° to 30° C, using radiant warmers with servo-controlled
temperature probes placed on the infant within minutes of
delivery, and the use of accessory prewarmed mattress or
heating pads for the tiniest infants. Hats are routinely used
as a method of decreasing heat loss, but they have not been
shown to be consistently effective (McCall et al, 2008).
It is important to note that as a required safety feature,
radiant warmers substantially decrease their power output
after 15 minutes of continuous operation at full power. If
this decrease in power is unrecognized, the infant will be
exposed to much less radiant heat. By applying the temperature probe and using the warmer in servo-controlled
mode, the temperature output will adjust as needed and
the power will not automatically decrease.
ASSISTING VENTILATION
As the newborn infant begins breathing and replaces the
lung fluid with air, the lungs become inflated and an FRC
is established and maintained. With inadequate development of FRC, the infant will not be able to oxygenate
and will develop bradycardia if inadequate FRC is prolonged. Providing assisted ventilation when the infant’s
spontaneous breathing is inadequate is probably the most
important step in newborn resuscitation. The indications for providing assisted ventilation (positive-pressure
ventilation) include apnea or bradycardia of less than 100
beats/min. Positive-pressure ventilation can be delivered
noninvasively with a pressure delivery device and a face
mask or invasively with the same pressure delivery device
and an endotracheal tube. Pressure delivery devices can
include self-inflating bags, flow-inflating or anesthesia
bags, and t-piece resuscitators, each with its own advantages and disadvantages. The self-inflating bag is easy
to use for inexperienced personnel and will work in the
absence of a gas source. However, the self-inflating bag
requires a reservoir to provide nearly 100% oxygen and
does not consistently provide adequate positive end expiratory pressure (PEEP). These devices can deliver very
high pressure if not used carefully. Although they have
pressure blow-off valves, these valves do not always open at
the target blow-off pressures (Finer et al, 1986). An anesthesia bag or flow-inflating bag requires a gas source for
use, allows the operator to vary delivery pressures continuously based on the felt compliance, but requires significant
practice to develop expertise. However, using a test lung
and intermittent airway occlusion, experienced anesthesiologists were unable to recognize the increased resistance
from an airway obstruction using only their hands (Spears
et al, 1991). A t-piece resuscitator is easy to use, requires a
gas source, and delivers the most consistent levels of pressure, but requires intentional effort to vary the pressure
levels (Hoskyns et al, 1987). The flow-inflating bag and
t-piece resuscitator allow the operator to deliver continuous positive airway pressure (CPAP) or PEEP relatively
easily (Bennett et al, 2005; Finer et al, 2001).
A level of experience is required to perform assisted
ventilation using a face mask and resuscitation device,
and this is especially true for an infant with extremely low
birthweight. It is important to maintain an open airway
for pressure to be transmitted to the lungs. The procedure
of obtaining and maintaining an open airway includes at
minimum clearing fluid with a suction device, holding the
head in a neutral position, and sometimes lifting the jaw
slightly anteriorly. The face mask must make an adequate
seal with the face in order for air to pass to the lungs effectively. No device will adequately inflate the lungs if there is
a large leak present between the mask and the face. Wood
et al (2008) measured face mask leaks of more than 55%
when participants were evaluated, providing positive pressure to manikins at baseline. The amount of the leak was
able to be decreased to approximately 30% with specific
instruction (Wood et al, 2008). Until recently there were
no masks that were small enough to provide an adequate
seal over the mouth and nose for the smallest infants.
Such masks are now readily available and facilitate bagmask resuscitation of small infants. Signs that the airway is
open and air is being delivered to the lungs include visual
inspection of chest rise with each breath and improvement
in the clinical condition, including heart rate and color.
The use of a colorimetric carbon dioxide detector during
bagging will allow confirmation that gas exchange is occurring by the observed color change of the device, or it will
alert the operator of an obstructed airway by not changing
color (Leone et al, 2006). Airway obstruction is common
in the preterm infant during positive-pressure ventilation
immediately after birth (Finer et al, 2009). It is important
to remember that these devices will not change color in
the absence of pulmonary blood flow, as occurs with inadequate cardiac output. At times, multiple maneuvers are
CHAPTER 28 Resuscitation in the Delivery Room
required to achieve an open airway, such as readjusting the
head and mask positions, choosing a mask of more appropriate size, and further suctioning of the pharynx. Alternate methods of providing an open airway include the use
of a nasopharyngeal tube (Lindner et al, 1999), a laryngeal
mask airway device (Grein and Weiner, 2005), or an endotracheal tube.
The amount of pressure provided with each breath
during assisted ventilation is critical to establishing lung
inflation and therefore adequate oxygenation. Although
it is important to provide adequate pressure for ventilation, excessive pressure can contribute to lung injury.
Achieving the correct balance of these goals is not simple
and is an area of resuscitation that requires more study.
A single specific level of inspiratory pressure will never
be appropriate for every baby. Initial inflation pressures
of 25 to 30 cm H2O are probably adequate for most term
infants. The NRP textbook (Kattwinkel, 2006) recommends initial pressures of 20 to 25 cm H2O for preterm
infants. The first few breaths may require increased
pressure if lung fluid has not been cleared, as occurs
when the infant does not initiate spontaneous breathing. Newborn infants with specific pulmonary disorders such as pneumonia or pulmonary hypoplasia also
frequently require increased inspiratory pressure. It has
been shown that using enough pressure to produce visible chest rise is associated with hypocarbia on admission
blood gas evaluation (Tracy et al, 2004), and excessive
pressure may decrease the effectiveness of surfactant
therapy (Bjorklund et al, 1997). It may be possible to
establish FRC without increasing peak inspiratory pressures by providing a few prolonged inflations (3 to 5
seconds of inspiration), although the use of prolonged
inflations has not been associated with better outcomes
than conventional breaths during resuscitation (Lindner
et al, 2005). Choosing the actual initial inspiratory pressure is less important than continuously assessing the
progress of the intervention. A manometer in the circuit
during assisted ventilation provides the clinician with
an indication of the administered pressure, although if
the airway is blocked this pressure is not delivered to the
lungs. The volume of air delivered to the lungs seems to
be more important than the absolute pressure delivered
in the development of lung injury. Tidal volume can be
monitored with respiratory function monitors that are
placed in the respiratory circuit (Schmolzer et al, 2010).
The most critical component of continued assessment is
evaluation of the infant’s response to the intervention. If
the condition of the infant does not improve after initiating ventilation, then the ventilation is most likely inadequate. In our experience the two most likely reasons for
inadequate ventilation are a blocked airway and insufficient inspiratory pressure. The occluded airway can
be noted using a colorimetric carbon dioxide device as
described previously, and frequently it can be corrected
with changes in position or suctioning while inadequate
pressure is corrected by adjusting the ventilating device.
The use of continuous pressure throughout the breathing cycle seems to be beneficial for establishing FRC and
improving surfactant function (Hartog et al, 2000; Michna
et al, 1999; Siew et al, 2009); this is accomplished during
assisted ventilation with the use of PEEP or with CPAP when
335
additional inspiratory pressure is not needed. In the absence
of PEEP, a lung that has been inflated with assisted inspiratory pressure will lose on expiration most of the volume that
had been delivered on inspiration. This pattern of repeated
inflation and deflation is frequently thought to be associated
with lung injury. In preterm infants, a general approach of
using CPAP as a primary mode of respiratory support in
NICUs has been associated with a low incidence of chronic
lung disease (Ammari et al, 2005; Avery et al, 1987; Van
Marter et al, 2000; Vanpee et al, 2007). Recently, te Pas and
Walther (2007) evaluated a ventilation strategy that included
the use of a I-piece resuscitator and a nasopharyngeal tube
to deliver a prolonged breath followed by CPAP, compared
with the use of a self-inflating bag to deliver positive pressure
ventilation when needed without any CPAP provided until
infants reached the NICU. Infants treated with the prolonged breath and CPAP required less endotracheal intubation and had lower rates of bronchopulmonary dysplasia. The
use of CPAP compared with endotracheal intubation and
mechanical ventilation as the initial mode of respiratory support was evaluated in the Continuous Positive Airway Pressure or Intubation at Birth (COIN) trial (Morley et al, 2008),
but the intervention did not begin until after 5 minutes of
life and therefore cannot inform the decision about the use
of these therapies as immediate resuscitative interventions.
The recently completed Surfactant Positive Airway Pressure
and Pulse Oximetry Trial (SUPPORT Study Group 2010.
Available at http://clinicaltrials.gov/ct2/show/NCT002333
24?term=neonatal+respiratory+support&rank=15) evaluated
the use of CPAP versus intubation and surfactant use in the
first hour of life as the primary respiratory support for preterm infants. These trials suggest that CPAP is beneficial for
improving some short-term outcomes associated with prematurtiy. Although these trials do not evaluate these therapies specifically in the resuscitation period, the respiratory
support provided shortly after birth should probably mimic
the support provided in the NICU after delivery.
If assisted ventilation is necessary for a prolonged period
of time or if other resuscitative measures have been unsuccessful, ventilation must be provided by a more secure
device such as an endotracheal tube. If it has been difficult to maintain an open airway while ventilating via a
face mask, an appropriately placed endotracheal tube will
provide a stable airway. This stability will allow more
consistent delivery of gas to the lungs and therefore provide for the ability to establish and maintain FRC. At this
time intubation is required for administering surfactant,
and it can be used to administer other medications such
as epinephrine if necessary for resuscitation. Finally, for
non-vigorous infants born through meconium-stained
amniotic fluid, intubation is performed for suctioning of
the airway.
The intubation procedure is often critical for successful resuscitation, requires a significant amount of skill
and experience to perform reliably, and may be associated with serious complications. The placement of a
laryngoscope in the pharynx often produces vagal nerve
stimulation, which leads to bradycardia. A photograph of
the desired view of the larynx is shown in Figure 28-6.
Assisted ventilation must be paused for the procedure,
which if prolonged can lead to hypoxemia and bradycardia. Intubation has been shown to increase blood pressure
336
PART VII Care of the Healthy Newborn
TRANSITIONAL OXYGENATION
AND OXYGEN USE
FIGURE 28-6 (See also Color Plate 18.) View of the glottis and vocal
cords as the laryngoscope is gently lifted. (From American Heart Association, American Academy of Pediatrics: Kattwinkel J, editor: Neonatal resuscitation textbook, ed 5, Elk Grove Village, Ill., 2006, American Academy of
Pediatrics.)
and intracranial pressure (Kelly and Finer, 1984). Trauma
to the mouth, pharynx, vocal cords, and trachea are all
possible complications of intubation. Performing the
intubation procedure when the infant already has bradycardia and hypoxemia can lead to further decline in heart
rate and oxygenation (O’Donnell et al, 2006). In addition,
hypoxia and bradycardia are more likely when intubation
attempts are prolonged beyond 30 seconds. Therefore it
is most appropriate to make an attempt to stabilize the
infant with noninvasive ventilation before performing
the procedure, limiting each attempt to 30 seconds or
less (Lane et al, 2004), and allow time for the infant to
recover with noninvasive ventilation between attempts.
If misplacement of the endotracheal tube in the esophagus goes unrecognized, the infant may experience further
clinical deterioration. Clinical signs that the endotracheal
tube has been correctly placed in the trachea include auscultation of breath sounds over the anterolateral aspects
of the lungs (near the axilla), mist visible in the endotracheal tube, chest rise, and clinical improvement in heart
rate and color or oxygen saturation. The use of a colorimetric carbon dioxide detector to confirm intubation
significantly decreases the amount of time necessary to
determine correct placement of the endotracheal tube
from approximately 40 seconds to less than 10 seconds
(Aziz et al, 1999; Repetto et al, 2001); this is the primary
method of determining endotracheal tube placement.
Successful placement of the endotracheal tube is not
always easy and is sometimes not possible. Airway anomalies may make alternate methods of airway management
necessary. A laryngeal mask airway is one such alternative that has been described for use in patients with Pierre
Robin sequence or other airway anomalies (Yao et al,
2004). Some practitioners have reported using the laryngeal mask airway for all positive-pressure delivery after
birth, with success noted in infants as small as 1.2 kg (Gandini and Brimacombe, 1999). Administration of medications including surfactant and epinephrine through this
device has undergone preliminary investigations (Chen
et al, 2008; Trevisanuto et al, 2005).
It is critical to remember that fetal arterial oxygen levels
are much lower than newborn arterial oxygen levels. The
transition from fetal to newborn levels does not take place
instantaneously. Using pulse oximetry in the immediate
newborn period, several investigators have established that
this transition with an ultimate oxygen saturation level
greater than 90% takes 5 to 15 minutes to occur in infants
who do not otherwise require resuscitation. For term and
preterm nonresuscitated infants, the median interquartile
range (IQR) of oxygen saturation at 3 minutes was 76%
(64% to 87%) and at 5 minutes was 90% (79% to 91%;
Kamlin et al, 2006). Expected oxygen saturation levels are
slightly lower for preterm compared with term and for
infants delivered via cesarean section compared with those
delivered vaginally (Rabi et al, 2006). Oxygen saturation
levels measured in preductal sites are 5% to 10% higher
than those measured from postductal sites for approximately 15 minutes of life (Mariani et al, 2007). A great deal
of variability occurs in the saturation values among different healthy individuals during the first 5 minutes of life,
but a resuscitation team can expect that there be a steady,
albeit slow, increase in levels over several minutes. If values
are below a threshold at different time points or not progressively increasing, intervention should be considered.
The use of pure oxygen for ventilation became routine
practice in resuscitation simply because it seemed logical
that oxygen would be beneficial. However, the recognition that oxygen could also be toxic led some investigators to question this previously well accepted practice. The
toxicity of oxygen is anticipated when the cellular antioxidant capacity is impaired, as occurs during the reperfusion
phase following an hypoxic-ischemic insult. After animal
studies showed the potential harmful effects of oxygen
(Poulsen et al, 1993; Rootwelt et al, 1992), clinical trials
were conducted to evaluate the effects of oxygen use during resuscitation of depressed infants. Several worldwide
trials have compared the use of pure (100%) oxygen with
room (ambient) air (21% oxygen) as the initial ventilating
gas for asphyxiated newborns. These trials found that air
was as successful as oxygen in achieving resuscitation, and
infants resuscitated with air had a shorter time to initiate
spontaneous breathing and less evidence of oxidative stress
(Ramji et al, 2003; Saugstad et al, 1998; Vento et al, 2001,
2003). Metaanalyses of several of the trials indicated that
infants resuscitated with air had a lower risk of mortality than those resuscitated with pure oxygen (Rabi et al,
2007; Tan et al, 2005). These trials have been criticized
because they were not all strictly randomized, and some
sites were in developing countries; however, metaanalysis
of the strictly randomized trials, which were mostly done
in European centers, demonstrates significant benefit for
survival with the use of air compared with pure oxygen
(Saugstad et al, 2008).
The preterm infant may be more susceptible to any
harmful effects of excessive oxygen exposure, because
of decreased antioxidant enzyme capacity. Some of the
infants in the previous oxygen trials were preterm, but
few weighed less than 1000 g. Review of the outcomes for
preterm infants (<37 weeks’ gestation) demonstrated an
CHAPTER 28 Resuscitation in the Delivery Room
even greater reduction in death for those initially resuscitated with room air compared with oxygen (Saugstad
et al, 2005). NICUs generally attempt to reduce oxygen
toxicity by limiting the amount of oxygen administered to
newborns using an upper limit for oxygen saturation and
adjusting supplemented oxygen levels to maintain saturation levels within that limit. The unlimited use of oxygen
during resuscitation exposes the preterm infant to higher
oxygen saturation levels than would routinely be accepted
in the NICU. Several small trials of oxygen use during
resuscitation of preterm infants have been performed in
the last 3 years. Among 42 preterm infants younger than
32 weeks’ gestation treated with either pure oxygen for
5 minutes or a targeted oxygen strategy beginning with
21% oxygen, all infants were provided some level of
supplemental oxygen (Wang et al, 2008). Infants initiated on pure oxygen had higher pulse oximeter saturation (Spo2) levels during transition, but did not have any
differences in heart rate, need for intubation, or survival.
The infants provided air from the start of resuscitation
all required an increase in inspired oxygen to obtain the
specified oxygen saturation targets. Using 30% versus
90% oxygen at the start of resuscitation and adjusting the
concentration based on the clinical status of the infant,
Escrig et al (2008) found that infants initially receiving
30% oxygen had lower overall exposure to oxygen without any adverse effects. The group receiving 30% oxygen
received increased oxygen concentrations up to approximately 55% at 5 minutes, but both groups received similar oxygen concentrations after 4 minutes of life, with a
level of approximately 35% by 15 minutes of life (Escrig
et al, 2008). In a more recent study, these investigators
also reported a decrease in the incidence of bronchopulmonary dysplasia in infants initially receiving 30% oxygen
compared with 90% oxygen (Vento et al, 2009). Finally,
the Room Air versus Oxygen Administration during
Resuscitation (ROAR) study was a blinded, randomized
controlled trial in 106 infants at 32 weeks’ gestation or less
comparing three oxygen strategies: one group received
100% throughout (HOB), one received 100% initially
(MOB), and one received 21% (LOB) initially; the last
two groups had the oxygen concentration titrated to keep
Spo2 at 85% to 92%. Spo2 levels were below the specified target range 61% of the time in the LOB group and
were above the target range 49% of the time in the HOB
group (p <0.001; Rabi et al, 2008). Larger trials of preterm infants treated with different oxygen strategies are
necessary to determine the long-term effects of resuscitation with different oxygen concentrations. Oxygen use in
the first minutes of life could affect survival and common
neonatal morbidities associated with prematurity and free
radical disease, such as neurodevelopmental impairment,
retinopathy of prematurity, bronchopulmonary dysplasia,
and necrotizing enterocolitis.
The use of oxygen concentrations between 21% and
100% requires compressed air and a blender. Different
organizations throughout the world have provided differing recommendations on the use of oxygen for newborn resuscitation. The most recent review of evidence
for the updated NRP guidelines suggest that resuscitation could begin with either air or blended oxygen; if
blended oxygen is unavailable, air is preferred to pure
337
oxygen. The suggested goal for oxygen saturation values
is the IQR at each minute (Kattwinkel et al, 2010). Our
approach has been to begin with 40% oxygen and adjust
slowly, attempting to mimic the gradual transition in
Spo2 values that occur in healthy newborns transitioning
from fetal oxygenation levels with an expected increase
in Spo2 from the fetal level of approximately 50% to a
target of 85% to 90% by 7 to 8 minutes, an increase of
approximately 5% per minute. Figure 28-7 demonstrates
continuous physiologic data during the first 10 minutes
of life for an infant resuscitated using a targeted oxygen
strategy.
ASSISTING CIRCULATION
In newborn infants, the need for resuscitative measures
beyond assisted ventilation is extremely rare. Additional
circulatory assistance can include chest compressions,
administration of epinephrine, and volume infusion. In a
large urban delivery center with a resuscitation registry,
0.12% of all infants delivered received chest compressions,
epinephrine, or both during 1991 to 1993 (Perlman and
Risser, 1995), and 0.06% of all infants delivered received
epinephrine during 1999 to 2004 (Barber and Wyckoff,
2006). Ventilation remains the most critical priority in
neonatal resuscitation; however, if adequate ventilation
is provided for 30 seconds, and bradycardia with a heart
rate less than 60 beats/min persists, chest compressions are
initiated. Further attention to ventilation with the use of
increased pressures or intubation may be required. Chest
compressions are preferably provided with two thumbs
on the sternum while both hands encircle the chest with
(Menegazzi et al, 1993). The chest is then compressed in a
3:1 ratio coordinated with ventilation breaths.
Further circulatory support may be necessary if adequate chest compressions do not result in an increase in
heart rate after 30 seconds. Epinephrine is then indicated
as a vasoactive substance, which increases blood pressure
by α-receptor agonism, improves coronary perfusion pressure, and increases heart rate by β-receptor agonism. Intravenous administration of epinephrine is more likely to be
effective than endotracheal administration. The intravenous dose of 0.01 to 0.03 mg/kg (0.1 to 0.3 mL/kg of a
1:10,000 solution) is currently recommended. Early placement of an umbilical venous catheter is critical to delivering
intravenous epinephrine quickly enough to be effective. In
order to place an umbilical catheter as quickly as possible,
it is necessary to have the equipment readily available and
to begin the procedure as soon as possible. This could be
accomplished by the lead resuscitator assigning the task of
placing the catheter as soon as chest compressions are initiated. If there is any prenatal indication that substantial
resuscitation will be required, the necessary equipment for
umbilical venous catheter placement should be prepared
before delivery as completely as possible. Epinephrine
can be given by endotracheal tube, but the efficacy of this
delivery method is not as certain; therefore an increased
dose (0.05 to 0.1 mg/kg) is recommended. Epinephrine
doses can be repeated every 3 minutes if heart rate does not
increase. Excessive epinephrine can result in hypertension,
which in preterm infants may be a factor in the development of intraventricular hemorrhage. However, the risks
PART VII Care of the Healthy Newborn
200.0
A
Pulse rate
150.0
100.0
bpm
338
50.0
0.0
102.0
B
65.0
Sat
Oxygen saturation
83.5
46.5
50.0
0.0
0.0
Percent
FiO2
28.0
100.0
2.5
5.0
7.5
Minutes
FIGURE 28-7 Tracing of physiologic data during transition. The pulse rate, oxygen saturation, and oxygen concentration (FiO2) measured
within 30 seconds of birth are displayed here. The pulse oximeter is placed on the infant’s right hand as soon as possible. Point A shows when the
pulse oximeter begins working. Point B shows the oxygen saturation (SpO2) at 3 minutes (approximately 80%). This infant was initially treated with
40% oxygen, and the concentration was adjusted to achieve SpO2 of 85% to 90% by 5 minutes of life. When the oxygen is weaned down to 21% at
approximately 7.5 minutes of life, the SpO2 decreases and oxygen concentration is increased again.
are balanced by the benefit of successful resuscitation in an
infant who might not otherwise survive.
If the infant has not responded to all the prior measures,
a trial of increasing intravascular volume should be considered, which involves administering crystalloid or blood.
Situations associated with fetal blood loss are also frequently associated with the need for resuscitation. These
situation include placental abruption, cord prolapse, and
fetal maternal transfusion. Some of these clinical circumstances will have an obvious history associated with blood
loss, whereas others might not be readily evident at the
time of birth. Signs of hypovolemia in the newborn infant
are nonspecific, but include pallor and weak pulses. Volume replacement requires intravenous access, for which
emergent placement of an umbilical venous catheter is
essential. Any infant who has signs of hypovolemia and
has not responded quickly to other resuscitative measures should have an umbilical venous line placed and a
volume infusion administered. The most common, and
currently recommended, fluid for volume replacement is
isotonic saline. A trial volume of 10 mL/kg is given initially and repeated if necessary. If a substantial blood loss
has occurred, the infant may require infusion of red blood
cells to provide adequate oxygen-carrying capacity. This
infusion can be accomplished emergently with noncrossmatched, O-negative blood, with blood collected from
the placenta or with blood drawn from the mother, who
will have a compatible antibody profile with her infant
at the time of birth. Because not all blood loss is obvious and resuscitation algorithms usually discuss volume
replacement as a last resort of a difficult resuscitation, the
clinician needs to keep a high index of suspicion for significant hypovolemia so that action can be taken to correct the
problem as promptly as possible. Therefore in situations
where the possibility for hypovolemia is known before
birth, it would be wise to prepare an umbilical catheter
and an initial syringe of isotonic saline and discuss with
the blood bank and the delivering physicians the possibility that non-crossmatched blood may be required.
SPECIFIC PROBLEMS ENCOUNTERED
DURING RESUSCITATION
NEONATAL RESPONSE TO MATERNAL
ANESTHESIA AND ANALGESIA
Medications administered to the mother during labor can
affect the fetus by transfer across the placenta and direct
action on the fetus or by adversely affecting the mother’s
condition, and therefore altering uteroplacental circulation and fetal oxygen delivery. The most commonly discussed complication of intrapartum medication exposure
is perinatal respiratory depression after maternal opiate
administration. The fetus can develop respiratory depression from the direct effect of the drug. Naloxone has been
used during neonatal resuscitation as an opiate receptor
antagonist to reverse the effects of fetal opiate exposure.
Naloxone chloride (0.1 mg/kg per dose, intravenously,
intramuscularly, or endotracheally) can be considered
if the newly born infant does not develop spontaneous
respirations after adequate resuscitation and the mother
has received an opiate analgesic during labor, provided
CHAPTER 28 Resuscitation in the Delivery Room
the opiate exposure is not chronic. Infants of mothers
who have a chronic opiate exposure can potentially have
a sudden withdrawal syndrome, including seizures if they
receive a narcotic antagonist. It is also critical that assisted
ventilation be provided as long as spontaneous respirations
are inadequate. It should be noted that the administration of a narcotic antagonist is never an acutely required
intervention during neonatal resuscitation, because these
infants can be treated with assisted ventilation.
CONDITIONS COMPLICATING
RESUSCITATION
When resuscitation has proceeded through the described
steps without improvement in the infant’s clinical condition, other problems should be considered. Some of these
problems may be modifiable with interventions that could
improve the course of the resuscitation. For example, an
unrecognized pneumothorax could prevent adequate pulmonary inflation and if under tension could impair cardiac
function. If the pneumothorax is recognized and drained,
gas exchange and circulation can be improved. Some congenital anomalies that were not diagnosed antenatally
make resuscitation more difficult. Congenital diaphragmatic hernia is difficult to recognize on initial inspection of the infant, but can cause significant problems with
resuscitation. The abdominal organs are displaced into
one hemithorax, and the lungs are unable to develop normally; this will cause ventilation to be difficult. If the intestines are displaced into the thorax and mask ventilation
is provided, the intestines will become inflated, making
ventilation more difficult. If the congenital diaphragmatic
hernia is known before delivery or a presumptive diagnosis
is made in the delivery room, the baby should be intubated
early to prevent intestinal inflation. An orogastric suction
tube should also be placed to decompress the inflated
intestines.
Many other congenital anomalies that can lead to a difficult resuscitation will be more visibly obvious when the
baby is born. For example, hydrops fetalis occurring for
any reason can be associated with difficult resuscitation.
Although most cases are diagnosed on fetal ultrasound
examination before delivery, severe hydrops would be visible on examination with skin edema and abdominal distention. Frequently peritoneal and pleural fluid will need
to be drained to achieve adequate ventilation. Abdominal
wall defects such as gastroschisis require special attention
in the delivery room to ensure that the exposed bowel is
covered in plastic, to prevent excessive fluid loss, and is
protected from twisting or trauma. An orogastric suction
tube is also important to decompress the stomach and limit
the chances of further vomiting. If assisted ventilation is
necessary, early intubation as opposed to mask ventilation
should be considered to prevent gastric distention. Infants
with neural tube defects should also have any exposed
tissues protected with a covering. In addition, all efforts
should be made to avoid pressure on the defect.
Fetal congenital high airway obstruction can cause
particularly difficult resuscitation. If a significant airway
obstruction is diagnosed antenatally, an ex utero intrapartum treatment (i.e., EXIT) procedure can be planned. This
procedure allows for establishing a stable airway before
339
clamping the umbilical cord, which maintains placental
function until the airway is secure. An airway obstruction
may not necessarily be diagnosed before delivery and can
cause difficult resuscitation. The therapy will vary depending on the cause of obstruction. An alternate airway (oral or
nasopharyngeal) can be helpful if endotracheal intubation is
not possible, as can occur with micrognathia. Tracheal suctioning can be attempted if a tracheal plug is suspected. In
extreme situations of airway obstruction, an emergency cricothyroidotomy may be attempted.
Birth trauma of any kind has been documented to occur
in approximately 2.6% of all deliveries in the United States
(Moczygemba et al, 2010). The most serious injuries are
the variety of head injuries that can occur, including subgaleal hemorrhage and intracranial hemorrhage. Subgaleal
hemorrhage is more often associated with vacuum-assisted
delivery; it and is important to recognize because of the
rapid blood loss that can occur into this soft tissue space.
Intracranial hemorrhages such as subdural hematomas can
occur, although many such injuries are mild and can be
found incidentally after uncomplicated vaginal delivery.
Spinal cord injuries after birth are extremely rare, but can
be severe with long-lasting functional limitations. Brachial
plexus and other peripheral nerve injuries may be noticed
in the delivery room and evaluated shortly after birth, but
they should not interfere with the resuscitation efforts.
Pressure injuries from forceps-assisted deliveries can be
noted on examination and evaluated as necessary in accordance with the location of the injury. Fractures and lacerations can occur and need to be evaluated after the newborn
infant has transitioned adequately.
LIMITS OF VIABILITY
Neonatal intensive care has increased survival at lower
gestational ages, resulting in changes in the definition of
viability over time. There is a variety of opinion among
practitioners regarding the lower limit of gestational age
when intensive care should be offered. The NRP states that
not providing intensive care is appropriate at less than 23
weeks’ gestation or 400 g birthweight. When the outcome
is uncertain, parents of the infant are frequently included
in the decision-making process. Making an informed decision about providing intensive care should be done with
the best information available. One of the most useful
resources to determine the likelihood of survival without
severe disabilities has been provided by Tyson et al (2008)
using the National Institute of Child Health and Human
Development Neonatal Research Network data. Using
information regarding the gestational age, birthweight,
sex, number of fetuses, and antenatal steroid exposure, a
calculation of the risk of mortality and neurodevelopmental impairment can be made. These predictive data that
were compiled from the participating centers of the Neonatal Research Network, a group of mostly academic centers, may currently be the best national data available for
predicting outcome, but they do not necessarily represent
the outcome at any single unit or of any individual baby.
When there is the possibility of delivery at a very preterm
gestational age at a small, inexperienced center, all efforts
should be made to transport the mother to an experienced
inborn center.
340
PART VII Care of the Healthy Newborn
CARE AFTER RESUSCITATION
Infants who survive a significant resuscitation require special attention in the hours to days that follow. Frequent
complications immediately following resuscitation include
hypoglycemia, hypotension, and persistent metabolic acidosis. In addition, infants with evidence of hypoxic ischemic encephalopathy may benefit from mild therapeutic
hypothermia (Azzopardi et al, 2009; Jacobs et al, 2007;
Shankaran et al, 2005). Mild therapeutic hypothermia, in
which the core body temperature is kept at 33.5° C, has
been extensively evaluated and has been effective at reducing death or impairment in infants with moderate to severe
hypoxic ischemic encephalopathy. The decreased temperature is thought to decrease the secondary injury that
occurs after an hypoxic-ischemic insult. Hypothermia can
be accomplished with both whole-body and head cooling,
although clinical trials of whole-body cooling more effectively achieved a reduction in adverse outcomes. The therapy is most beneficial when initiated as quickly as possible
after an insult, with beneficial effects noted when treatment was initiated within 6 hours of birth. The timing of
insult in relation to the time of birth is not always obvious,
making it difficult to know the actual timing of initiating
therapy after the insult.
Mild hypothermia therapy should be considered when
an infant has required a significant resuscitation after
birth, Apgar scores are low (especially a 5-minute score
less than 5), fetal or neonatal acidosis are documented on
cord or newborn blood gases, and signs of encephalopathy
are apparent. In addition, the history of a significant event
likely to cause a hypoxic-ischemic insult should trigger a
thorough evaluation of the newborn to determine whether
hypothermia therapy is indicated. Hypothermia therapy
is not currently available at all institutions; however, all
delivery services must be able to recognize the indications
for therapy, so that a transfer can be initiated as quickly as
possible, if necessary.
In infants born without a heart rate or any respiratory
effort, if resuscitation is performed to the full extent without any response, discontinuation is recommended after
10 minutes. From a review of 13 years of data from a
database including 81,603 deliveries, Haddad et al (2000)
found that survival with an Apgar score of 0 at 1 minute
occurred in 1.26 of 1000 delivered infants without major
malformations. Of 33 infants assigned Apgar scores of 0 at
both 1 and 5 minutes, 67% died before hospital discharge
(Haddad et al, 2000). A recent review of the available literature for infants with Apgar scores of 0 at 10 minutes
found that 94% of infants either died or were severely
handicapped, whereas 3% were mild or moderately handicapped (Harrington et al, 2007).
The transition from fetal to neonatal life is a critical time
in an individual’s life and is an opportunity for care providers to have significant effect on the outcome of infants who
need assistance. The need for neonatal resuscitation, even
when no signs of encephalopathy are recognized, increases
the risk that children will have lower scores on intelligence
quotient tests at school age (Odd et al, 2009). This risk
arises most likely because resuscitation is a marker for a
prior insult. However, a well-performed resuscitation
could be critical for a successful recovery. Neonatal care
providers have an obligation to ensure that this process is
performed as well as possible and that the techniques of
resuscitation are evaluated in an objective manner to promote continued improvement.
SUGGESTED READINGS
Apgar V: A proposal for a new method of evaluation of the newborn infant, Curr
Res Anesth Analg 32:260-267, 1953.
Carbine DN, Finer NN, Knodel E, et al: Video recording as a means of evaluating
neonatal resuscitation performance, Pediatrics 106:654-658, 2000.
Costeloe K, Hennessy E, Gibson AT, et al: The EPICure study: outcomes to
discharge from hospital for infants born at the threshold of viability, Pediatrics
106:659-671, 2000.
Kamlin CO, O’Donnell CP, Davis PG, et al: Oxygen saturation in healthy infants
immediately after birth, J Pediatr 148:585-589, 2006.
Morley CJ, Davis PG, Doyle LW, et al: Nasal CPAP or intubation at birth for very
preterm infants, N Engl J Med 358:700-708, 2008.
Saugstad OD, Ramji S, Soll RF, et al: Resuscitation of newborn infants with 21%
or 100% oxygen: an updated systematic review and meta-analysis, Neonatology
94:176-182, 2008.
Shankaran S, Laptook AR, Ehrenkranz RA, et al: Whole-body hypothermia for
neonates with hypoxic-ischemic encephalopathy, N Engl J Med 353:1574-1584,
2005.
Siew ML, te Pas AB, Wallace MJ, et al: Positive end-expiratory pressure enhances
development of a functional residual capacity in preterm rabbits ventilated
from birth, J Appl Physiol 106:1487-1493, 2009.
te Pas AB: Walther FJ. A randomized, controlled trial of delivery-room respiratory
management in very preterm infants, Pediatrics 120:322-329, 2007.
Teitel DF: Circulatory adjustments to postnatal life, Semin Perinatol 12:96-103,
1988.
Tyson JE, Parikh NA, Langer J, et al: Intensive care for extreme prematurity: moving beyond gestational age, N Engl J Med 358:1672-1681, 2008.
Vohra S, Roberts RS, Zhang B, et al: Heat Loss Prevention (HeLP) in the delivery
room: a randomized controlled trial of polyethylene occlusive skin wrapping in
very preterm infants, J Pediatr 145:750-753, 2004.
Wiswell TE, Gannon CM, Jacob J, et al: Delivery room management of the apparently vigorous meconium-stained neonate: results of the multicenter, international collaborative trial, Pediatrics 105(1 Pt 1):1-7, 2000.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
P A R T
V
III
Care of the High-Risk Infant
C H A P T E R
29
Stabilization and Transport
of the High-Risk Infant
George A. Woodward, Roxanne Kirsch, Michael Stone Trautman, Monica E. Kleinman,
Gil Wernovsky, and Bradley S. Marino
NEONATAL TRANSPORT MEDICINE
INTRODUCTION
Regionalization of medical care has improved the ability
to centralize resources and improve patient outcomes. For
optimal coordinated care, however, adequate medical transport needs to be developed and continually refined to enable
delivery of patients to regional centers, and for specialized
care to be available for and delivered to patients in distress.
For centers that serve as a basic or specialized service, there
will be times when subspecialty care is required. For those
who deliver subspecialty care, there will also be times for
most organizations when transfer to a similar level organization may be required for reasons such as capacity or extraordinary care. For hospitals that do not have birthing centers
as part of their facility, the patient population, the acuity of
the arriving patients, and potentially the ultimate morbidity
and mortality of those patients depend on skilled, efficient,
and quality neonatal transport. Although transfers to neonatal centers in the 1960s and early 1970s often occurred in an
ad hoc manner, such as in a police car or an ambulance (i.e.,
a converted hearse) with variably skilled providers, transfer
programs today offer a more sophisticated level of care. This
level of care, however, is not consistent throughout cities,
nationwide, and internationally. Many transport services
and centers have grown around individual center needs,
without clear attention to coordination and regionalization
of services. Competitive systems, often located in similar
areas or vying for similar patient populations, have resulted
in the duplication of services at the ground and air levels,
and at times increased risk and cost to patients and providers
as part of the efforts to increase patient volume and revenue.
When considering transport of neonatal patients, several situations can occur. Intrafacility transport may be
required for specialty services within a particular institution. Interfacility transport between lower and higher levels of service capability can also occur, as well as between
relatively equivalent levels of service because of capacity
or other issues. Transported patients may be of high acuity, relatively stable, or in various stages of convalescent
care. Each type of transport requires anticipatory planning,
appropriate staffing, adequate modalities (e.g., transport
vehicles), and strong relationships between referring and
receiving providers, in addition to skilled, qualified, and
certified transport personnel. As noted in the next section,
transfer agreements can help to minimize inefficiencies and
enable rapid approval and eventually transport of patients.
This chapter will review considerations and requirements for neonatal transport; discuss issues involved in
transport team operation, including equipment, personnel,
mode of transport, and medical-legal issues; and present
general and specific topics, including quality improvement
opportunities, that might be encountered in a neonatal
transport system (American Academy of Pediatrics et al,
2007; Cornette, 2004; Woodward et al, 2007).
REGIONALIZATION OF NEONATAL CARE,
TRANSFER AGREEMENTS, AND BACK
TRANSPORT
The concept of regionalization of neonatal care and transport developed from the formation of neonatal stations in
the 1920s and 1940s (Oppenheimer, 1996). These stations
were located within certain area hospitals, where additional resources were allocated to provide care for premature infants. One consequence of the formation of these
stations was the development of equipment and protocols
to transport premature infants from other area hospitals to
those specialized areas to receive care.
In the 1960s and 1970s, as interest in neonatal care grew,
so did the number of hospitals offering services for premature infants. To help optimize the care being delivered, the
March of Dimes produced Toward Improving the Outcome
of Pregnancy in a 1976 report (Committee on Perinatal
Health, 1976). The report stratified maternal and neonatal
care into levels based on their complexity, and it proposed
the referral of high-risk patients to centers with sufficient
personnel and resources to provide care. The goal was to
create standard definitions so that comparisons of health
outcomes, resource utilization, and costs among regional
institutions could be made. High-risk maternity patients
would be able to actively participate in selecting a delivery
service, and businesses would be able to select appropriate
health care resources for their employees. The subsequent
March of Dimes publication in 1993 Toward Improving the
Outcome of Pregnancy: The 90s and Beyond reiterated the
importance of regionalized care and further delineated
care levels (Committee on Perinatal Health, 1993). The
concepts of regional care were adopted and incorporated
into the Guidelines for Perinatal Care (American Academy
341
342
PART VIII Care of the High-Risk Infant
of Pediatrics Committee on Fetus and Newborn and Bell,
2007; Woodward et al, 2007). Whereas the original driving
forces for regionalization in the 1970s were the shortage
of trained personnel to care for low birthweight (LBW)
infants and the economic expense to maintain these skills,
during the late 1980s and 1990s, technology and clinical
expertise disseminated outside the regional tertiary centers, resulting in proliferation of the number of intermediate care neonatal intensive care units (NICUs). This
proliferation has blurred many of the original distinctions
between various care systems. Whether driven by third
party payers or other factors, with various interpretations
and applications of what “regional care” means, the results
have been the creation of a variety of care options (Lainwala et al, 2007).
Limited space and cooperative longitudinal care planning in some tertiary care units have created the necessity
for patient transport back to a unit with less acuity or fewer
resources once their critical condition has resolved or stabilized (Attar et al, 2005; Donovan and Schmitt, 1991;
Lynch et al, 1988). Regionalization guidelines should support the return to the community for patients who no longer need the highest care level. Patient selection for back
transport (transport back to referring service, hospital, or
a hospital closer to the patient’s home) and care in another
facility should match the capabilities and expertise of the
community hospital (Stark and American Academy of
Pediatrics Committee on Fetus and Newborn, 2004).
Although third-party payers often drive decision making,
transport relationships develop between various institutions
either by formalized transfer and preferred provider agreements or by historical and personal relationships (Attar
et al, 2005). Transfer agreements can help to define the
roles, understanding, and expectations between institutions
and the transport service; they also help frequently to detail
reimbursement issues. These agreements set the expectations for participating facilities, with the ultimate goal of the
timely movement of patients from one facility to another
2007; Woodward et al, 2007). Whether optimal care can
be provided in some smaller NICUs or outside of a tertiary
unit is frequently debated. Nonetheless, there is a trend in
the United States and elsewhere toward the centralization
of perinatal care (Howell et al, 2002; Wall et al, 2004).
Several investigators have shown that mortality was lowest for deliveries of infants with very LBW that occurred
in hospitals with tertiary care NICUs (Chien et al, 2001;
Phibbs et al, 2007; Rautava et al, 2007; Robertson et al,
1994). Cifuentes et al (2003) supported the idea that whenever possible, women in early preterm labor should be
moved to the regional hospital rather than transferring
the infant after birth. The study was insufficiently powered, however, to make a recommendation regarding the
difference between regional and large community NICUs
(Cifuentes et al, 2003). In trying to discern which LBW
infant might be better cared for in tertiary regional care
centers, Vieux et al (2006) showed that the risk factors for
requiring high levels of intensive care were low gestational
age, twin pregnancy, maternal hypertension, antepartum
hemorrhage, infection, and male gender, whereas antenatal
steroid therapy and premature rupture of membranes were
protective factors against requiring intensive care. Regional
care concerns can apply to the neonates with extremely
LBW and to the late preterm population (Khashu et al,
2009). Investigators trying to describe the optimal neonatal unit caring for very preterm infants in Europe were
unable to produce a consensus as to what the ideal neonatal
unit size and patient volume should be (Van Reempts et al,
2007). Given the wide variability in levels of neonatal care
and the inability to predict premature delivery, neonatal
transport will continue to be a dynamic process.
TRANSPORT COMMUNICATIONS
When developing and maximizing transport capabilities, a key concept is the centralization of communication. Although a telephone call from a referring provider
to a receiving provider might be the most efficient way to
receive advice or notify a receiving provider of a potential
transport, there are more centralized and effective means
of communication. First, a centralized, easily recalled,
advertised, and monitored (24 hours per day, 7 days per
week) communication system should be available. Identifying and publishing a centralized access number for immediate access to the transport system or receiving center
personnel are imperative for optimal communication and
efficiency (Southard et al, 2005). Anyone who has transported or referred patients to systems without centralized
access understands the challenges in working through
operators, unit clerks, multiple providers, and often multiple services to enable a singular transport. This process
is time consuming and often frustrating for the referring
provider, and it is often time that could be better spent in
direct assessment and care of the patient, rather than on the
telephone with repetitive informational transfer. Ideally a
centralized transport communication center would enable
a referring provider to make a single call to a single number and immediately receive all the services that they might
require. Those services include appreciation and recognition of the need for transfer, identification of appropriate
hospital and facility, review of medical issues, and determination of required services (i.e., personnel, equipment, time
of response) needed for transport. Also included would be
simultaneous awareness of need for transport by the personnel responsible for arrangement of particular modes of
transport, verification of bed capacity, and any other logistic items that may need to take place prior to the transport.
A telephone call made to an individual provider requesting
transport requires a sequential process from data gathering
through acceptance for admission and eventual transport
service or modality identification and dispatch. Centralized
access to a communication center allows all those functions
to occur simultaneously, enabling more rapid transport
response and appropriate involvement of all those required
for the management of the particular patient (American
Academy of Pediatrics Committee on Fetus and Newborn
and Bell, 2007; Woodward et al, 2007).
MEDICAL SUPERVISION
A key requirement for any system is to have appropriately skilled and immediately available medical command
physicians (American Academy of Pediatrics Committee
on Fetus and Newborn and Bell, 2007; Woodward et al,
2007). A medical command physician should be literate
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
and expert in the medical area of concern. In most cases
involving neonatal transport, this provider should be a
neonatologist. There may be instances, however, when
the referring or receiving physicians may request or desire
additional medical expertise. For example, a cyanotic newborn with congenital heart disease may be temporarily
improved or stabilized well by the referring neonatologist
and have additional stabilization direction provided by the
receiving medical command physician; however, invaluable
additional management and planning might be added by a
partnering cardiac intensive care physician. A communication or transfer center can allow for multiple providers to
be linked together during an initial referral call to allow the
highest level advice to be presented and discussed among
the providers. These telephone calls should also include
the transport personnel so that the providers have the
background information and care plans delivered directly.
MODE OF TRANSPORT
Once the transport referral has been made, ideally through
a communication center, and discussions have been started
with the medical command physician, the transport can
take place. Decision on mode of transport is an important
component to consider at this juncture. In general, ground
transport allows door-to-door service between facilities,
enables a well-lighted environment, provides space for
several providers as well as the patient and a family member, and is efficient in urban and short-range transfers.
As the distance for transfer increases, and the need for
decreased out-of-hospital time becomes more important,
consideration of air transport becomes important depending on acuity and patient disease process. In general, air
transport is most expeditious over approximately 50 miles,
with helicopter being favored between 50 and 150 miles
and airplane being effective for transports greater than 150
miles. The decision regarding mode of transport is influenced by several issues, which include available modes of
transport, staffing and medical expertise of the providers
involved in those particular modes, current and projected
patient condition (which includes preliminary identification of the underlying disease process), consideration of
stability or illness progression during the projected transport timeframe, capabilities of the referring facility and
personnel, and the urgency of intervention and definitive
placement of the patient. Other issues that influence the
process include geographic and weather constraints, distance and duration of transport, and provider perception
about the transport process. In addition to the potential
risks of ground transport, which include traffic accidents,
vibration, noise, and other issues, air transport creates concerns for accident, vibration, gravitational forces during
acceleration and deceleration, excessive noise, temperature variations, and decreased humidity with altitude. Air
transport also includes issues related to altitude physiology that can affect patients with respiratory issues or air
trapping and their air-containing equipment (e.g., endotracheal tube cuffs, laryngeal mask airways; Wilson et al,
2008; Woodward et al, 2002, 2006). Dalton’s Law recognizes that ambient oxygen decreases as altitude increases;
therefore there may be a need for pressurization and augmentation with increased FiO2. Boyle’s Law states that
343
as altitude increases, the volume of a gas also increases,
and the barometric pressure is inversely related to the volume of the gas. This law is potentially a serious issue for
patients with an enclosed gas collection, such as a simple or
developing pneumothorax.
TRANSPORT PERSONNEL, EDUCATION,
AND TEAM COMPOSITION
Awareness of the capabilities of the transport system
and of the personnel involved in transport is imperative
in decisions regarding mode of transport. Although it is
ultimately the responsibility of the referring physician to
identify the appropriate mode and personnel for transport,
per Emergency medical Treatment and Active Labor Act
(EMTALA), opportunities exist for tertiary care and referral centers to help to educate the referral providers regarding optimal transport planning and usage (Bolte, 1995;
Woodward, 1995). In general, issues influencing transport
decisions include current level of care, urgency for a different level of medical capability or equipment, current
providers of care, stability of the patient, options available to the provider and patient, and efficiency and quality of the transport process. Ideally, these issues are the
determinants of appropriate transfer; however, referring
providers are often overwhelmed by the severity or acuity of the patient and their primary desire may be to have
the patient removed from their facility as quickly as possible. The providers may decide on a transport or transfer
process based solely on speed of transport process rather
than quality of care. It is imperative for the receiving and
tertiary care centers to educate the referring population
on the importance of stabilization, initiation and quality
of initial response, transport options, and definitive care to
maximize potential patient outcome. When examining the
transfer of patients, providers should ask a simple question:
“Are we trying to deliver the patient to tertiary care, or are
we trying to deliver tertiary care to the patient?” In most
high-functioning transport and referral centers, the latter
is true. The referring physician should expect to have tertiary care advice and direction delivered at the moment of
the referral telephone call and continued throughout the
transport process (American Academy of Pediatrics Committee on Fetus and Newborn and Bell, 2007; Woodward,
1995; Woodward et al, 2007). At no time should the care
delivered to the patient decrease in sophistication after a
referral call is made. If a transfer places the infant in a suboptimal environment or with providers who are not competent to address the infant’s current or potential medical
needs, the patient might receive suboptimal care and the
referring provider is liable for arranging and providing
inappropriate care.
When considering the team composition of the transport providers, it is important to consider the quality of
the personnel, their expertise and experience, and their
ability to work in the transport environment (King and
Woodward, 2002a; King et al, 2001, 2007). There are
many variations of transport teams in the United States
and abroad. These teams can be composed of physicians,
nurse practitioners, nurses, respiratory therapists, paramedics, and other health care providers. In the United
States, medical (pediatric or specialty) trainees are often
344
PART VIII Care of the High-Risk Infant
used as primary providers in the transport environment.
Regardless of the formal educational background of an
individual, there are several criteria that must be met to be
optimally effective in the transport environment. First, the
provider must have adequate certification, be licensed for
the care they deliver, and be able to provide the assessments
and interventions that the patient currently or potentially
requires during the transport process. For example, a neonatal retrieval service must be able to manage acute and
critical airways in the neonatal population, both at a referring hospital and during the transport. It is interesting that
those same providers might not be credentialed to provide
those skills within their home hospital; however, they have
been certified to provide them in the ambulance environment. In general, this must be done under the auspices of
a physician’s care, which may be from an accompanying
physician or via online medical control (real-time medical advice during the transport process). Transport care
also often involves offline medical control, which involves
the use of protocols developed by medical personnel. It
is important to recognize that the transport timeframe is
somewhat limited; therefore the personnel might not need
to have the longitudinal or differential diagnosis expertise
of a fully trained neonatologist. However, these personnel
must have the acute care assessment abilities and intervention skills of an experienced neonatal expert. In addition to
training the onsite personnel, it is imperative to have medical command physicians understand the opportunities
and limitations of the transport services and environment
and to understand the risks and challenges that referring
personnel can potentially encounter with situations and
patients who exceed their own personal or their facility’s
management abilities. The physicians must have significant
awareness of the transport environment to understand the
limitations of potential interventions. It is also imperative
that medical command positions have superb communication skills, not only at the referring physician level but also
within the transport team. Medical command physicians
will need to be able to effectively and efficiently communicate with providers from different disciplines.
QUALITY IMPROVEMENT
Transport medicine offers an opportunity to identify areas
for potential improvement within the inpatient arena, in
the transport system, and at the referring facilities (Browning Carmo et al, 2008; Chen et al, 2005; Lim and Ratnavel, 2008; McPherson et al, 2008; Ramnarayan, 2009).
This glimpse into medical sophistication and capability is
one that is privileged and should be used to identify educational opportunities rather than prompt judgmental
and critical review. Education by referring physicians and
transport teams can have a significant effect on the quality and outcome of patient care and the volume of future
referrals. Ideally, once a referral call is made, a receiving
physician or medical command physician will direct the
care so that the job of the transport team is to verify that
an appropriate working diagnosis has been made and that
adequate stabilization procedures have been performed.
The team can verify that the interventions are secure and
then transport the patient in a stable fashion. Systems that
do not gather adequate information or offer appropriate
advice, or in which the referring facilities do not follow
that advice or choose not to perform needed interventions, put the patient at risk by having a delay in care of
initial interventions, prolonging the transport, and delaying delivery of definitive care. The transport team that has
invested several hours at a bedside, stabilizing a newborn
with medical or surgical issues, may be spending time in
a facility that is not ideal, has a limited number of skilled
personnel, and has minimal backup, thus prolonging the
transport process and potentially putting that individual
patient at risk (Chen et al, 2005; Haji-Michael, 2005).
However, the patient and the system are at risk because the
valuable resource of specialized neonatal transport personnel is not available for another patient.
Ideally, care delivery would be the same at referral and
receiving centers; the development of practice guidelines
can be helpful in that regard. Guidelines that are evidence
based, developed by regional and local experts, and disseminated to referring locations and transport teams will
help to standardize and enable consistent care across variable locations. It is necessary, however, to assess and reassess the quality of the guidelines and the competency of
their use to ensure optimal results from this process. Even
in the best of hands, near-miss or realized adverse events
may happen. It is clear that identification of those events,
discussion with families (where appropriate), and root
cause analysis are imperative. Several studies have examined adverse events in transported patients. Ligtenberg
et al (2005) noted that one third of patients had an adverse
event, and 50% of those resulted from not following the
advice of the medical command physician. Of that group,
70% of events were avoidable and 30% were logistical.
In a review of the London Neonatal Transfer Service,
Lim and Ratnavel (2008) noted that 36% of their patients
had greater than or equal to one adverse event, and two
thirds of those were due to human error; half of those
occurred before the team arrived at the referral center, and their major etiologies included preparation and
communication.
A neonatal transport service must determine if maternal
transport is part of their purview. It is clear that preterm
infants who are born at outside hospitals and require transfer to tertiary care centers or who are born at one tertiary
care center and require transfer to another have worse
outcomes that include increased mortality and morbidities such as intraventricular hemorrhage (IVH) and other
medical issues (Baskett and O’Connell, 2009; Janse-Marec
and Mairovitz, 2004; Jony and Baskett, 2007; O’Brien et al,
2004; Ohara et al, 2008). Developing appropriate criteria for transporting women in preterm labor may help to
direct the optimal use of resources required with maternal transfer. Identifying the appropriate time to transport
women in preterm labor, as well as those with other preterm medical issues, will help in developing appropriate
criteria and limit resource utilization for maternal transfers. It is important to note that significant work regarding
neonatal and maternal transport and regionalization of care
in the United States, United Kingdom, Australia, Europe,
and other areas has already been accomplished. There are,
however, multiple opportunities to improve transport on
an individual and regional basis in these developed countries and in other developing countries.
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
TRANSPORT ADMINISTRATION
As a hospital develops and optimizes a neonatal transport
program, experts in transport medicine are integral to
the success of this program (American Academy of Pediatrics Committee on Fetus and Newborn and Bell, 2007;
Woodward et al, 2007). A quality medical director and
program director, often a nurse or respiratory therapist,
are essential to understand the potentially complicated and
challenging environment of transport medicine. These
leaders should be instrumental in identifying expectations,
roles, and responsibilities for the process; this includes
access through a communication center and developing and
disseminating referral center expectations. These expectations include stabilization and preparation of the patient
before transport, making an appropriate decision to transfer, choosing the appropriate transport process and destination, obtaining consent from the family for transport,
discussing plans for stabilization and intervention with the
medical command physician, and initiating that plan as
able. Referral centers must be able to be direct when they
believe that the suggested interventions are inappropriate
or beyond their scope. The referring team also needs to
be available to participate in transition care to the transport team and the receiving service. The receiving and
transport team responsibilities include being immediately
available for case discussion, having the ability to rapidly
accept the patient if capacity is available, offering clear and
concise expert recommendations, ensuring preparation of
the environment and the staff both for the transport and
arrival of the patient, organizing additional diagnostics and
interventions, and providing available and accessible neonatal advice throughout the process. Most important, the
team needs to ensure that appropriate skills and therapy are
available and delivered throughout the process, from referral call through definitive placement, and ensure seamless
transition at each point of care. The team needs to communicate well with referring and the patient’s physicians
and document their advice, interventions and activities in
a clear, concise fashion to enable appropriate patient and
provide protection for the transport service.
TRANSPORT EXPERTISE
As transport medicine has developed over the past 30 to
40 years, there has been an increase in the quality and volume of transport research, as well as organizations that
are discipline and process specific. Multiple disciplines
have organizations with transport focus and expertise,
including the American Academy of Pediatrics Section on
Transport Medicine, which includes executive committee
representation from neonatology, pediatric critical care,
and emergency medicine physicians, as well as a broad
membership from other subspecialties and disciplines.
The American Academy of Pediatrics Transport Section
published its most recent Guidelines for Neonatal and
Pediatric Transport in 2007; they also host a listserv at
[email protected], which is a national repository of information and a conduit to others in the transport arena for transport-related issues.
There are many keys to success in transport medicine.
These keys include integrity, professionalism, preparation,
345
anticipation, education, competency, critical evaluation of
current and new practices, the ability to critically assess the
quality of care being delivered at every point along the way,
and looking for opportunities to improve access, efficiency,
and delivery of care. An area that is vital for appropriate
transport care is the assurance of competency of the providers (American Academy of Pediatric Committee on Fetus
and Newborn and Bell, 2007). As noted, providers can come
from many different backgrounds and bring different skills
from their clinical and training experience. It is imperative
for the transport system and leadership to ensure that the
skills required are present in their personnel. This responsibility includes initial education, continuing education,
and competency assessment. A team may decide to have
individuals who are experts in defined areas of care, such as
a physician who manages airways and the pneumothorax,
a nurse who manages intravenous access and medication
delivery, and a respiratory therapist who is responsible for
airway and ventilatory support, whereas another team may
choose to have all team members competent in all skills.
Specific procedures may be useful or needed in transport
that are not as common in the hospital environment or to
the hospital-based providers (e.g., laryngeal mask airway;
Trevisanuto et al, 2005). However the team is structured,
there must be clear and concise guidelines for ongoing
training and assessment of the personnel. High-fidelity
simulation models offer additional opportunities to assess
and potentially improve technical and cognitive capabilities
(LeFlore and Anderson, 2008). The American Academy
of Pediatrics Guidelines for Air and Ground Transport of
Neonatal and Pediatric Patients (available at www.aap.org)
is an invaluable resource that outlines required education
and competencies for each level and discipline.
The true quality of a transport service is sometimes difficult to determine, because many teams and services have
been developed independently and are not part of larger
regional systems. It is difficult to compare and contrast
systems in different areas for which different patient populations are transported (Berge et al, 2005; Cornette and
Miall, 2006; Craig, 2005; Doyle and Orr, 2002; Khilnani
and Chhabra, 2008; Van Reempts et al, 2007).
One avenue for potential standardization of the transport
environment is by assessment and accreditation by the Commission on Accreditation of Medical Transport Systems,
which was initiated in 1990 as a direct response to the number of air medical accidents in the 1980s. This certification is
voluntary in some areas and required in others, and it serves
to assure providers, stakeholders, and the public of adherence to quality of care and transport safety standards. The
Commission on Accreditation of Medical Transport Systems is an independent organization, supported by 16 member organizations, which publishes standards and arranges
reviews of interested air and ground transport programs.
Further information can be found at http:www.camts.org.
TRANSPORT SAFETY
Safety of the transport system and for its providers is paramount and must be assessed and ensured before transporting any patient. Vehicles must be safe and meet the
standards for air or ground transport, the personnel must
be appropriate, licensed, and competent, and the patients
346
PART VIII Care of the High-Risk Infant
must be managed in the most appropriate and professional
fashion. In addition, the logistics of travel must include
a safe environment, including helmets and fire-retardant
suits for those who fly in helicopters, three-point restraints,
and appropriate ambulance seating arrangements. There
should not be an occasion when providers put themselves
at risk by being unrestrained or being in an area where
unsecured debris or inappropriately placed equipment
may damage a provider or a patient. Adherence to rules
and regulations of air and ground transport is imperative
as well (Clawson, 2002; Greene, 2009; King and Woodward, 2002b; Levick et al, 2006; National Highway Traffic
Safety Administration, 2009).
It is imperative to recognize that there is risk with both
air and ground transport. The air transport industry has
seen an acute spike in tragic and fatal air accidents (Greene,
2009; National Transportation Safety Board Accident
Database, 2009). This increase has caused the industry, and
the U.S. government, to critically investigate these issues
and offer suggestions to improve transport safety (Federal
Aviation Administration Helicopter Safety Initiative, 2011;
10.5-year U.S. HEMS Safety, 2008). Requirements such
as duty hours for pilots, weather restrictions, flight under
instrument, flight rules with terrain avoidance equipment,
and night vision goggles can to help minimize the risk for
these transports. Although ground ambulances are used
much more frequently, and the risk of injury and death is
evident, the fatality rate is lower in ambulance accidents
than it is in aircraft accidents (Becker, 2003; Becker et al,
2003; King and Woodward, 2002b). It is necessary, however, that the vehicles be maintained and operated in a safe
manner. Many systems do not allow ambulances to exceed
posted speed limits and use lights and sirens only as a way
to identify an emergency response, not to enable the vehicle to circumvent or ignore standard traffic laws (Clawson,
2002). Appropriate equipment for ambulances is required
as well, and the July 2009 statement by the American College of Surgeons Committee on Trauma regarding appropriate equipment for ambulances (also published as an
American Academy of Pediatrics’ policy statement) should
be reviewed by all transport systems (American College of
Surgeons Committee on Trauma, 2009; American College
of Surgeons Committee on Trauma et al, 2009).
It is evident from the transport and pediatric literature
that patient outcomes are improved with specialty providers. There have been multiple studies to examine this
particular issue (Belway et al, 2006; Mullane et al, 2004).
Perhaps the most compelling is the study by Orr et al
(2009), which examined transports provided by variable
providers within the same system. This study compared
outcomes in patients whose care was delivered by specialized pediatric critical care teams with those whose care was
delivered by general providers. Both teams had the same
medical command oversight, equipment, and modalities.
Patient outcomes were worse for those whose care was not
delivered by specialty teams, and much improved for those
who were. One challenge, however, with transport teams
is that differentiation of medical resources, such as a neonatal specialty team, likely means that there may be a scarcity and potential need for rationing of those resources.
It is possible to develop teams with a variety of personnel
with complementary cognitive and procedural skill sets
and work toward appropriate triage of transport requests
to ensure the proper level of onsite skill provision. There
have been multiple attempts to develop triage tools for
pediatric and neonatal care providers, including the Mortality Index for Neonatal Transport, the Modified Clinical
Risk Index for Babies, and the Risk Score for Transported
Patients, which are noted in the bibliography (Broughton
et al, 2004a, 2004b; Markakis et al, 2006).
FAMILY-CENTERED CARE
Transport team research has shown that family-oriented
care, as in other areas of the medical systems, is an important component of transport (Woodward and Fleegler,
2000, 2001). Families who have been formally surveyed
appreciate the opportunity to participate in the care of
their child. In neonatal transport, however, there are times
when there are two patients who may require care in two
disparate locations. A mother who has had a cesarean section and has delivered an acutely ill child in need of care in
a higher facility is one such example. It is required that the
mother be in one facility and the child be in another. The
father may be conflicted regarding accompanying his new
child on the transport or staying with the child’s mother.
Transport teams need to be sensitive to the challenges and
opportunities for the family and include them in the process when possible. It is evident that when parents attend
or accompany transport team members on critical care
transports, they are not there to assess the medical skill set
of the provider, but to provide support to their child. It is
also a great opportunity for the transport team to demonstrate to the family that their patient is in focused, professional, caring, and capable hands.
MEDICAL LEGAL ISSUES
There are many medical legal issues in transport medicine,
as elsewhere in the medical system (Hedges et al, 2006;
Williams, 2001; Woodward, 2003). The Health Insurance
Portability and Accountability Act (HIPAA) is a required
component of transport planning and delivery. Discussion
of patients should not take place in a public area or via public
communication airways where non–patient-related personnel or bystanders could overhear information about a specific patient. As noted earlier, a requirement of EMTALA
is that the referring physician chooses the appropriate
mode of transport and ensures that the transport process
and receiving hospital are appropriate for the particular
patient. Patients cannot be transferred if they are unstable
and the ability to further stabilize them is available at the
initial site of care. If a patient must be transferred for care
while in an unstable condition—a frequent scenario for critically ill patients who need care not available at the referring
institution—consent must be obtained from the family,
which acknowledges their understanding of the potential
risks and benefits of the process. In practice, there are often
patients in unstable condition who are transferred from
lower to higher levels of care, because the level of care that
can be provided at the referring or initial center is not optimal for the child. This reason is appropriate for transfer as
compared with transferring patients because of financial or
other economic drivers. The medical liability for transport
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
is a shared process. Before the referring center calls regarding the patient and the center of referral has accepted care,
the entire medical responsibility lies with the referring provider. Once the receiving team has accepted the patient and
offered advice, medical liability is a shared process. The
referring physician maintains the majority of the liability, as
well as medical control of the patient, throughout the process until the transport team has left the referring hospital. It
is important to recognize that most transport teams and personnel do not have privileges at referring hospitals and are
working under the guidance and supervision of the referring
physician team. Transport teams that act independently, or
referring physicians who are not available when the transport team arrives, put both the referring provider and the
transport team at risk if there is disagreement or inappropriate care delivered to the patient. There will be times, however, when there is disagreement regarding the optimal care
to be delivered. For example, a child with a hypoplastic left
heart syndrome and an open ductus arteriosus may be given
a low-dose prostaglandin infusion and be in stable condition.
A transport team can insist on intubation before a long air
transport, whereas the referral physician might believe that
intubation is not required and that it poses a risk to that
particular patient. This situation can be challenging, and
it must be handled appropriately. It is never appropriate to
have obvious provider conflict occur at a patient’s bedside,
especially in front of family members. The appropriate way
to handle a situation that cannot be easily mitigated is to
involve the medical command physician with a telephone
call to the referring physician in a discussion at a peer-topeer level. Transport teams have been known to comply
with the wishes of the referring providers to not perform
advanced procedures at the referring hospital, only to perform those procedures in the ambulance, which is a much
less desirable location. Ideally, all disagreements and considerations of different therapies are discussed in a collegial
fashion in the appropriate environment to enable the safe
care and transport of the child to the receiving center. As
noted previously, documentation of all information received
and advice offered is imperative. If there is future review or
challenge to the care delivered during transport, as with any
other care delivered in the hospital, clear and appropriate
documentation should stand alone as an excellent defense. In
addition, many centers use recorded (i.e., digital, tape, other
retrievable recording process) for their intake and advice
lines; this is another way to review, educate, and ensure that
appropriate information is delivered in an effective communication style. The use of recorded lines with frequent
review, for educational and quality assurance purposes, can
be invaluable. Review with legal advisors can help to define
the length of time the recorded materials should be maintained for quality improvement or patient record addendum.
PATIENT CARE DURING TRANSPORT
EXTREME PREMATURITY
The most effective method of transporting premature
infants is within the mother as a maternal transport. By
delivering at a referral hospital, the premature infant
is exposed to all the variability of the extrauterine environment, with the added complexity of having to be
347
transported to a facility capable of providing the level of
care the infant needs. Most neonatal transport teams are
regarded as extensions of the NICU. The team initiates
and provides much of the same level of complex neonatal
care as the receiving hospital, but in a changing environment. It is this changing environment that poses unique
challenges for both patient and caregiver. These issues
would be amplified in the case of a disaster with care and
transfer required for premature infants (Gershanik, 2006).
Limit of Viability
The incidence of premature births has been increasing
in the United States. Of the approximate 4.1 million live
births occurring annually, 520,000 (12.7%) infants are
born premature (Ananth et al, 2009; Martin et al, 2008).
Although some premature infants require minimal care
to survive, infants with very LBW often test the resources
and skills of the most experienced care provider. The longterm outcome for many of these small infants is often
unknown for months to years after leaving the hospital. In
some instances, the long-term results have not been ideal
(Donohue et al, 2009; Tyson and Saigal, 2005). Several
studies of extremely premature infants born outside of tertiary care centers show increases in morbidity and mortality (Cifuentes et al, 2002; EXPRESS Group et al, 2009;
Phibbs et al, 2007; Rautava et al, 2007). Human viability is
currently limited by the physiology of pulmonary development and its ability to exchange gases. Currently this limit
appears to be at approximately 22 to 24 weeks’ gestation
(Pignotti and Donzelli, 2008). Unfortunately, for transport
teams and the care givers at referring hospitals, it is difficult
to determine which infants born at the margins of viability
should be resuscitated and provided with aggressive neonatal care and which should be allowed to die (American
Academy of Pediatrics Committee on Fetus and Newborn
and Bell, 2007; Buchanan, 2009). These decisions are best
made collaboratively with the family, transport team members, and the referring and receiving physicians (Ahluwalia et al, 2008; Gunderman and Engle, 2005; Tyson et al,
1996) and may ultimately result in a patient transport, even
when the likelihood for survival is minimal.
Thermoregulation
Problems in neonatal thermoregulation continue to be
a major contributor to neonatal morbidity and mortality
worldwide and can be especially problematic in neonatal
transport (World Health Organization, 1996). During
transport, neonates often cross into and out of multiple
different environments with wide temperature and humidity variations. Although a normal term infant is capable of
a significant homoeothermic response by using their sympathetic nervous system to vasoconstrict peripherally, thus
placing their normal layer of insulating white fat between
the body’s core and the exposed skin, preterm infants lack
the subcutaneous white fat insulation to protect their core
temperature. Moreover, term infants use brown fat as a
source for nonshivering thermogenesis. Preterm infants,
although as sensitive to temperature as term infants, lack
sufficient brown fat to sustain a response when exposed to
a cold environment (Baumgart, 2008). The consequences
348
PART VIII Care of the High-Risk Infant
of hypothermia can include hypoglycemia, metabolic
acidosis, intraventricular hemorrhage, persistent pulmonary hypertension, and hypoxemia (Bartels et al, 2005).
Silverman et al (1958) demonstrated that using a higher
incubator temperature, even without additional humidity
resulted in improved premature survival rates.
Additional measures such as chemical gel packs and
polyethylene occlusive skin wrapping help in maintaining
the temperature of an infant with very LBW (Vohra et al,
2004). For all newborns, an equally important condition to
avoid is hyperthermia.
Although elevated temperatures in neonates occur with
increased metabolic rates, prolonged seizures, dehydration, or infection, the most common cause of neonatal
hyperthermia is high ambient air temperature and humidity (Baumgart, 2008). In a review of a subset of patients
referred for a trial of hypothermia for hypoxic ischemic
encephalopathy, patients with an elevated body temperature had poorer neurologic outcomes than those with normal body temperatures (Laptook et al, 2008; Yager et al,
2004). Humidity also has an extremely important role
in temperature control of LBW infants, especially those
receiving mechanical ventilator support. The importance of
delivering humidified gas to neonates receiving mechanical
ventilation is widely acknowledged (Sousulski et al, 1983).
Ventilation with dry gases affects the airway epithelium in
very LBW infants, and it can result in hypothermia secondary to their large surface area–to–body mass ratio and
their relatively large respiratory minute volume (Fassassi
et al, 2007). There is a linear correlation between incubator temperature and the humidity generated by heated
humidity systems (Fassassi et al, 2007). Whereas ventilator
complications can be reduced and thermoregulation can
be improved by providing exogenous heat and humidity
to the gases, active heated humidification systems are used
infrequently during neonatal transport. Passive hygroscopic heat and moisture exchangers have been used for
short-term conventional mechanical ventilation and with
some types of high-frequency ventilation (Fassassi et al,
2007; Schiffmann, 1997; Schiffmann et al, 1999). Humidifying the incubator and the ventilator system helps in thermoregulation while transporting very LBW neonates.
Surfactant
Surfactant replacement has had a significant effect on newborn intensive care. Numerous systematic reviews have
demonstrated the benefit of surfactant administration in
reducing oxygen needs and ventilation requirements, as well
as ventilator complications such as pneumothorax and pulmonary interstitial emphysema (Horbar et al, 1993; Liechty
et al, 1991; Schwartz et al, 1994). Extremely LBW infants
benefit from both prophylactic (defined as within 10 to
30 minutes of birth) and rescue surfactant administration
(within 12 hours of birth) (Kendig et al, 1991; Soll, 2000).
Infants given prophylactic surfactant appear to have fewer
complications, including death, pneumothorax, and bronchopulmonary dysplasia (Soll and Morley, 2001), supporting
its use during transport (Riek, 2004). Surfactant administration can be complicated by airway obstruction and right
mainstem bronchus or esophageal instillation, justifying the
American Academy of Pediatrics Committee on Fetus and
Newborn recommendations that “preterm and term neonates who are receiving surfactant should be managed by
nursery and transport personnel with the technical and clinical expertise to administer surfactant safely and deal with
multisystem illness” (Engle, 2008). While investigations
are still ongoing as to the optimal manner of surfactant
administration, future technology might eventually allow
for surfactant to be delivered without airway intubation.
Currently, aerosolized surfactant has yet to be shown to
be superior to endotracheal tube administration (Berggren
et al, 2000; Mazela et al, 2007). Although there are concerns about the composition variability of animal-derived
surfactants, use of a synthetic surfactant such as Lucinactant creates other concerns. Lucinactant requires a special
warming cradle to convert it from a gel to a liquid before
administration, adding a level of difficulty to its administration during neonatal transport (Kattwinkel, 2005; Moya
et al, 2005). Smaller community hospitals might not stock
surfactant in their pharmacies, making it necessary for the
transport team to carry it as part of their medical supplies.
Earlier forms of surfactant came as lyophilized powder and
could be reconstituted when needed. Currently, the most
frequently used surfactant preparations in the United States
are derived from either bovine or porcine lung extracts.
These surfactants require refrigeration and are usually carried by transport teams in small containers cooled by gel
packs. The same precautions used in the delivery room or
the NICU apply to its use in transport. Checklists should
be used to avoid inadvertently leaving this lifesaving therapy
behind when the transport team is dispatched.
Before surfactant administration, proper endotracheal
tube position must be confirmed either clinically or radiographically. Surfactant administration is generally done
using transport team protocols, which usually mirror the
package inserts. After administering surfactant, monitoring
pulmonary compliance and adjusting the patient’s ventilator support will occasionally extend the time of a transport
to avoid clinical complications, such as pneumothorax or
endotracheal tube occlusion, during the transfer process.
Often by the time the transport team arrives at the receiving hospital, significant clinical improvement has occurred.
Surfactant has a role in other disease processes, such as
meconium aspiration, sepsis, and pulmonary hemorrhage,
during which a patient’s endogenous surfactant is depleted
or inactivated, thus creating a condition of secondary surfactant deficiency (Chinese Collaborative Study Group
for Neonatal Respiratory Diseases, 2005; Finer, 2004;
Wiswell et al, 2002).
HYPOXIC RESPIRATORY FAILURE
Meconium Aspiration Syndrome,
Persistent Pulmonary Hypertension
of the Newborn, Inhaled Nitric Oxide on
Transport, and Extracorporeal Membrane
Oxygenation Referrals
Hypoxic respiratory failure describes a heterogeneous
group of neonatal disorders that have in common impaired
oxygenation and the need for assisted ventilation. The
most frequently observed conditions are respiratory
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
distress syndrome, meconium aspiration syndrome, and
persistent pulmonary hypertension of the newborn
(PPHN), which can occur individually or in combination.
Interfacility transport of the newborn with hypoxic respiratory failure is potentially hazardous, with a high risk for
patient deterioration and complications of therapy, such as
pneumothorax.
Assisted ventilation during neonatal transport can be
accomplished with a variety of devices, although not all
modes of mechanical ventilators have been modified for
use in a mobile setting. Newborns with milder degrees of
illness may be managed successfully with continuous positive airway pressure (Murray and Stewart, 2008). Infant
transport ventilators range from simple time-cycled,
pressure-limited machines to more sophisticated devices
with flow sensitivity and the ability to synchronize. Certain
high-frequency ventilators have been successfully adapted
for the transport environment (see later).
Inhaled nitric oxide (iNO) is approved for use in term
and near-term newborns with hypoxic respiratory failure
with clinical or echocardiographic evidence of pulmonary hypertension. Persistent PPHN can exist in isolation or be secondary to another insult, such as sepsis or
meconium aspiration syndrome. Administration of iNO to
newborns with PPHN reduces the need for extracorporeal
membrane oxygenation (ECMO; Clark et al, 2000; Lowe
and Trautwein, 2007). With its availability to community
NICUs, iNO is often initiated before transport to a tertiary care center. Once iNO is administered, abrupt cessation of therapy can result in rapid clinical deterioration
from rebound pulmonary hypertension, so it is essential
to continue iNO if already initiated (Kinsella et al, 1995).
Transport teams may also consider initiation of iNO
therapy for term and near-term newborns with hypoxic
respiratory failure. If the referring facility does not have
the capability to perform echocardiography, transport staff
should carefully consider the possibility of congenital heart
lesions that could be worsened by the use of iNO, including total anomalous venous return and lesions dependent
on right-to-left ductal flow such as critical aortic stenosis.
The rationale for initiating iNO during transport is
to reduce pulmonary vasoreactivity and improve stability during transport. However, there have been no prospective studies to determine whether this practice affects
patient outcome. INO during transport has been delivered by a number of different systems, such as the aeroNOX (Aeronox Technology Corporation, Quezon City,
Philippines) iNOvent, and INOmax DS (INO Therapeutics, New Jersey) transport systems (Kinsella et al, 1995,
2002; Lutman and Petros, 2008; Tung, 2001). The use of
iNO in combination with high-frequency ventilation in
non-ECMO centers can complicate the transfer process.
If the transport team does not have the capability to provide mobile high-frequency ventilation, it is recommended
that the referring hospital perform a trial of conventional
mechanical ventilation before the arrival of the transport
team to establish that the infant can tolerate the transition.
Both high-frequency jet ventilation (Bunnell ventilators [Bunnell Inc., Utah]) and high-frequency flow interrupter ventilation (Bird ventilators [Viasys, California])
have been configured and used with nitric oxide during
transport (Honey et al, 2007; Mainilli et al, 2007). The
349
high-frequency oscillator (SensorMedics 3100A [Viasys,
California]) that is commonly used in many NICUs is
impractical for ground transport and is not configured for
helicopter or fixed wing transport. However, even with
these technologies, 30% to 40% of critically ill neonates
improve only temporarily with iNO therapy and will ultimately require higher care levels (Fakioglu et al, 2005;
Kinsella et al, 2002).
In patients with pulmonary or cardiac failure that is
unresponsive to maximal medical therapy, conventional
ECMO is often used as a bridge therapy to allow either
the lungs and heart to recover. Ideally, centers without
ECMO capability have prospective criteria to guide the
transfer of newborns before the need for ECMO cannulation. In some cases, the patient’s condition is so unstable
that conventional transport cannot be conducted safely.
Selected programs have the capability to provide mobile
ECMO, during which patients are cannulated for ECMO
before transport to the referring institution. Mobile
ECMO can also benefit patients already receiving ECMO
at a tertiary facility who are in need of advanced quaternary
therapies, such as heart or heart-lung transplants (Wagner
et al, 2008; Wilson et al, 2002). The resources and skill
set necessary to safely and consistently perform ECMO
in the transport environment have, by their complexity,
restricted the number of transport programs with mobile
ECMO capabilities (Coppola et al, 2008).
NEUROLOGIC ISSUES
Perinatal Depression and Therapeutic
Hypothermia
Hypoxic ischemic encephalopathy affects approximately
1 to 2 in 1000 term neonates (du Plessis and Volpe, 2002).
Several early studies from the 1950s and 1960s suggested
that hypothermia might be an effective therapy for the
asphyxiated neonates, but there was no systemic long-term
follow-up for these patients to confirm its effectiveness
until a series of animal studies suggested that hypothermia was an effective neuroprotective therapy (Gunn et al,
1997, 1998). Several randomized trials of therapeutic
hypothermia for term human infants have demonstrated
a reduction in the combined outcome of death and neurodevelopmental disability when cooled, compared with a
control population (Gluckman et al, 2005; Shankaran et al,
2005). However, the efficacy of hypothermia may diminish as the time from insult to cooling increases. Although
clinical trials and current guidelines suggest that cooling
should be initiated within 6 hours of insult, animal studies demonstrated that hypothermia is most effective if it
is started as soon as possible (Gunn et al, 1997, 1998). For
patients with a delay in referral or who are being transported from a distant center, waiting to start hypothermic
therapy until arrival at the receiving center potentially
places the patient at a significant disadvantage. The initial
larger clinical trials did not address these issues in regard
to critical care transport, but several case (Anderson et al,
2007), pilot (Eicher et al, 2005), and single-center studies have demonstrated the feasibility of controlled hypothermia during transport to significantly shorten the time
to therapy initiation (Zanelli et al, 2008). Several new
350
PART VIII Care of the High-Risk Infant
large-scale hypothermia trials that have been recently
completed or are nearing completion include initiation
of hypothermia before arrival at the referral center. The
Total Body Hypothermia for Neonatal Encephalopathy
Trial (the Toby Trial) compared standard intensive care
plus total body cooling with standard intensive care for
72 hours. Recruited patients from referring hospital were
assessed by the transport team, who performed amplitude
integrated electroencephalograms and obtained consent if
the patient was eligible and less than 6 hours of age. The
patients randomized to cooling had gel packs applied if
needed to cool the body to rectal temperature between 33°
and 34° C (Azzopardi et al, 2009). The Australian Cooling
Trial for Hypoxic-Ischemic Encephalopathy achieves the
temperature range for referral study patients by turning
off the normal heating systems and applying gel packs (at
10° C) around the infant’s head and chest, until the rectal
temperature is reduced to 33° to 34° C (Jacob et al, 2008).
While hypothermia is still being evaluated as a therapeutic tool to manage hypoxic ischemic encephalopathy, the
current management trend for term and near-term infants
being considered for hypothermia is to delay actively
warming the patient at a referral hospital until there is a
consultation with a receiving hospital that offers therapeutic hypothermia. If criteria are met, further cooling could
be achieved by placing wrapped disposable cooling packs
next to the trunk and head with continuous monitoring
of rectal temperature. Once the rectal temperature stabilizes, the infant is transferred to a transport incubator with
the heater turned off, and the transport team applies cooling packs or a blanket as needed to maintain a rectal temperature of 33° to 34° C during transport. Just as careful
temperature monitoring during hypothermia is necessary
for the safe transport of these patients, observational studies suggest that the avoidance of hyperthermia is critically
important. In an observational study, Laptook et al (2008)
noted an increase in mortality or disability in asphyxiated neonates who had elevated skin or esophageal temperatures. As additional studies are completed, therapeutic
hypothermia may enter common clinical and transport
practices. When combined with other therapeutic interventions, hopefully therapeutic hypothermia will have a
greater effect on the consequences of perinatal asphyxia.
Neonatal Seizures
The highest rate for seizures in pediatrics occurs during
the neonatal period, with an incidence of 2 to 3.5 in 1000
live births (Cowan, 2002); however, there is considerable
variability in treatment regimens among pediatric hospitals
in the United States (Blume et al, 2009). Phenobarbital,
phenytoin, or midazolam have not been shown to significantly improve seizure management or reduce morbidity
and mortality (Booth and Evanse, 2004; Carmo and Barr,
2005; Painter et al, 1999). Despite the evidence of limited
efficacy, phenobarbital remains the first line of therapy
to treat neonatal seizures (Painter et al, 1999; Sankar and
Painter, 2005). Before initiating therapy, the transport
team needs to consider the diverse causes for neonatal seizures. Metabolic disorders such as hypoglycemia, hypocalcemia, and inborn errors of metabolism, cerebral vascular
events including intraventricular hemorrhage and stroke,
bacterial and viral meningitis, infectious and developmental abnormalities can all manifest as or with neonatal
seizures (Silverstein and Jensen, 2007). Having initiated
treatment, the transport team needs to remain vigilant for
potential changes in the patient’s clinical status either as a
response to therapy or from the primary disease process.
Attention to the basics of airway, breathing, and circulation should continue throughout the transport, because
the major anticonvulsants used to treat neonatal seizure
all have potential side effects, most commonly respiratory depression and hypotension. Controversy remains
as to whether to treat all forms of neonatal seizures,
although treatment seems prudent in the transport environment (Bartha et al, 2007). In animal models, seizures
create neural, biochemical, and structural changes that
have long-term cognitive and behavioral consequences
(Thibeault-Eybalin et al, 2009). Similar neurologic findings have been suggested in human neonates. Both of the
large-scale clinical hypothermia trials for hypoxic ischemic
encephalopathy noted poorer prognosis for children with
hypoxic ischemic encephalopathy with seizures (Gluckman
et al, 2005; Shankaran et al, 2005). Glass et al (2009) compared magnetic resonance imaging findings and the presence of seizures in a group of newborns with suggested
hypoxic ischemic encephalopathy. Adjusting for the magnetic resonance imaging results, patients with seizures were
more likely to have long-term neurologic issues than those
without seizures (Glass et al, 2009). The ideal anticonvulsant agent would reliably stop clinical and electrographic
seizures with minimal adverse effects (Thibeault-Eybalin
et al, 2009). Several trials of new antiepileptic medications
are underway. Topirmante, levetiracetam, bumetanide,
and zonisamide have all been used to treat neonatal seizures, but there have been no randomized trial to demonstrate their effectiveness compared with current therapies.
Trials with levetiracetam and bumetanide are reportedly ongoing. Further work is necessary to develop more
effective and safer antiepileptic drugs so that, along with
potentially neuroprotective strategies, the vulnerable and
immature brain can be protected. Several trials and studies
have been proposed to hopefully elucidate a better treatment strategy (Clancy, 2006).
STABILIZATION AND TRANSPORT
OF THE NEONATE WITH CONGENITAL
HEART DISEASE
Transport of the neonate with congenital heart disease
(CHD) follows the general guidelines for the transport of
any critically ill neonate. However, neonates with complex
CHD often need therapeutic intervention, requiring the
support of multiple subspecialty consultants, including the
pediatric cardiothoracic surgeon and pediatric cardiologist
(Allen et al, 2003; Stark et al, 2000). The care of the neonate
with CHD generally necessitates transport to a specialized
center (Castaneda et al, 1989; Penny and Shekerdemian,
2001). The preoperative care of the patient affects postoperative outcomes and mortality (Mahle and Wernovsky,
2000; Mahle et al, 2000; Robertson et al, 2004; Simsic
et al, 2007; Wernovsky et al, 1995, 2000). With the increasing use of fetal ultrasound examination to diagnose CHD
antenatally, the ability to intervene immediately at delivery
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
has become increasingly expected. In a single tertiary care
center, 53% of neonates with CHD had a prenatal diagnosis (Dorfman et al, 2008), with an additional 38% obtaining
a diagnosis before discharge from the newborn nursery.
The aspects of stabilization of the neonate with CHD
either diagnosed by prenatal ultrasound or suggested by
postnatal clinical examination include initial resuscitation,
airway management, vascular access, a judicious use of
supplemental oxygen, prostaglandin E1 (PGE1) therapy,
inotropic support, and communication with cardiac specialty services for timely transport and intervention.
Neonates with Prenatally Diagnosed
Congenital Heart Disease
Significant advances in the diagnosis and treatment of
neonates with critical CHD across the last decade have
altered the timing of intervention, the types of interventions available, and the way in which CHD is diagnosed.
With increasing numbers of patients with prenatal diagnosis (Berkley et al, 2009; Friedman et al, 2002; Jone and
Schowengerdt, 2009; Khoshnood et al, 2005), fewer neonates may have their initial presentation of CHD in the
emergency department. In a single-center review, prenatal diagnosis accounted for 53% of presentations of CHD
(Dorfman et al, 2008). Another European center reported
that 51% of transports with CHD had a prenatal diagnosis (Bouchut, 2008). In addition, a single-center cohort of
critical CHD showed 44.3% to have had a prenatal diagnosis (Schultz et al, 2008). Clearly, prenatal diagnosis of
CHD has increased across the past decade.
Neonates with Suspected Congenital
Heart Disease
In a single tertiary care center, Dorfman et al (2008) found
that 38% of neonates received a diagnosis with CHD in
the newborn nursery before discharge, and 8% of patients
presented with CHD after initial hospital discharge at 4 to
27 days of life. The most common nursery findings
prompting diagnosis of heart disease in the nursery were
an isolated murmur, cyanosis, or both (Berkley et al, 2009;
Table 29-1). In a single emergency department review of
patients younger than 5 months with undiagnosed cardiac
disease, the most common presentation was congestive
TABLE 29-1 Presenting Symptoms of Patients Diagnosed
Postnatally
Murmur
Cyanosis
Respiratory distress
Shock
Arrhythmia
Other
Multiple symptoms
Diagnosis
in Nursery,
n = 73 (%)
Diagnosis
After Discharge,
n = 16 (%)
38
32
7
4
3
3
14
25
0
19
38
0
6
13
Modified from Dorfman AT, Marino BS, Wernovsky G, et al: Critical heart disease in
the neonate: presentation and outcome at a tertiary care center, Pediatr Crit Care Med
9:193-202, 2008.
351
heart failure, shock, and cyanosis; if restricting the findings to a neonatal population, shock and profound cyanosis were the presenting symptoms (Savitsky et al, 2003).
Another study showed that 45% of critical CHD cases
(CHD with signs of shock, end organ dysfunction, or in
cardiac arrest) were diagnosed postnatally, particularly
left-sided obstructive lesions (Schultz et al, 2008). This
finding is consistent with other centers showing a preponderance of left-sided obstructive lesions (excluding hypoplastic left heart syndrome) in those diagnosed postnatally
(Friedberg et al, 2009).
The cyanotic neonate who fails the hyperoxia test without an obvious pulmonary etiology, who has an equivocal
result on the hyperoxia test but has other signs or symptoms of CHD, or who is in shock within the first 3 weeks
of life is highly likely to have complex CHD. Furthermore,
pulse oximeter measurement of the postductal (probe on
the foot) oxygen saturation has been found to be specific for
the detection of CHD, particularly when combined with
findings on the clinical examination (de-Wahl Granelli
et al, 2009; Meberg et al, 2008, 2009; Reich et al, 2008; Sendelbach et al, 2008; Thangaratinam et al, 2007; Valmari,
2007). Although debate is ongoing as to the appropriateness of routine pulse oximetry as a screening tool (Sendelbach et al, 2008), the detection of saturations of less than
95% has a sensitivity of 72% to 77%, specificity rates over
99%, and false-positive rates of 0.2% to 0.6% in largevolume studies (Meberg et al, 2008; Valmari, 2007). These
neonates are likely to have heart lesions that depend on
blood flow through a patent ductus arteriosus to contribute
to either systemic or pulmonary blood flow, or improvement of intercirculatory mixing. PGE1 given as continuous infusion will reopen the ductus arteriosus. In babies
with ductal-dependent pulmonary blood flow, hypoxemia
is lessened as the ductus opens, and the resultant metabolic acidosis will resolve. Babies with systemic flow that
is dependent on the ductal connection to provide flow to
the descending aorta will have congestive heart failure, low
cardiac output, or shock, which is unlikely to be treatable
by standard measures without reopening the ductus arteriosus. Patients with transposition of the great arteries will
have improved intercirculatory mixing with a patent ductus arteriosus (Wernovsky, 2008; Wernovsky and Jonas,
1998). Interventions such as supplemental oxygen, airway
management, volume resuscitation, inotropic therapy, and
prostaglandin infusion require careful thought and consideration in the child with CHD.
Considerations of Therapies in the
Neonate with Congenital Heart Disease
The neonatal resuscitation algorithm is still applicable
in the presence of CHD (Johnson and Ades, 2005), but
should be modified in certain circumstances. In presentations with hypoxemia that is unresponsive to supplemental
oxygen, congestive heart failure, or shock, simultaneous
attention is devoted to the basics of neonatal advanced life
support and to assurance of a patent ductus arteriosus.
A stable airway must be maintained, allowing for adequate alveolar oxygenation and ventilation. In critically
ill neonates with CHD presenting with severe cyanosis
or circulatory collapse, intubation should be performed if
352
PART VIII Care of the High-Risk Infant
possible after premedication with sedation and neuromuscular blockade (discussed subsequently). Reliable venous
access is important, and arterial monitoring is helpful for
ongoing assessment of blood pressure, acid-base status,
and gas exchange. Volume resuscitation, inotropic support, and correction of metabolic acidosis may be required
to maximize cardiac output and tissue perfusion. Blood
glucose and ionized calcium should be checked and treated
to achieve normal range for age. An evaluation for sepsis is
typically performed simultaneously, and empiric antibiotic
therapy is initiated while evaluation continues.
Supplemental Oxygen
Supplemental oxygen is a potent pulmonary vasodilator
and systemic vasoconstrictor, and it can adversely affect
the physiology in neonates with a single ventricle, as well as
those with two ventricles with an unrestrictive ventricular
septal defect or great vessel communication (see Chapter 55,
Congenital Heart Disease). In these babies, the ratio of
pulmonary vascular resistance to systemic vascular resistance will determine the proportion of blood flow to each
vascular bed. The oxygen-induced pulmonary vasodilation
can decrease pulmonary vascular resistance and increase
pulmonary blood flow at the expense of systemic blood
flow, thus reducing systemic output. Titrating oxygen via
nasal cannula or face mask to a target peripheral saturation of 75% to 85% usually corresponds to adequate blood
flow in both the pulmonary and systemic systems. In the
setting of normal hemoglobin, cardiac output, and oxygen
consumption, these oximetry values will provide adequate
oxygen delivery. Higher oxygen saturations are typically
not necessary and importantly may in fact ultimately result
in decreased oxygen delivery to the peripheral tissues.
Considerations for intubation and mechanical ventilation
are discussed Intubation and Ventilation, later.
Prostaglandin E1 Therapy
In the instance of antenatally diagnosed CHD, ductal
dependency (either for systemic or pulmonary blood flow)
is often already determined. Whenever possible, PGE1
infusions should be prepared ahead of delivery and started
promptly in a peripheral intravenous catheter. The dose
of PGE1 varies according to the timing of the diagnosis
and the degree of ductal closure that is present upon discovery (or suspicion) of the cardiac lesion. In most cases,
PGE1 at 0.01 to 0.025 μg/kg/min via intravenous infusion
is adequate for stabilization. PGE1 may be given via a central venous catheter or peripheral intravenous catheter, or
in situations where intravenous access cannot be obtained,
via an umbilical arterial catheter or intraosseous line.
The dose is usually titrated down once the patent ductus
arteriosus has been demonstrated echocardiographically.
However, doses as high as 0.1 μg/kg/min may be required
in the newborn with significant ductal constriction or in
the neonate who at 1 to 2 weeks of life presents with a
closing, closed, or severely restrictive ductus arteriosus.
As in the prenatally diagnosed neonate, once therapeutic effect is achieved, the dose can generally be decreased
without loss of therapeutic effect. Prostaglandin response
is often immediate if ductal patency is important to the
hemodynamics of the infant. Failure to respond may mean
that the initial diagnosis of ductal dependent CHD is
incorrect, the ductus is unresponsive to PGE1 (which may
occur in older infants), or that there is no ductus arteriosus
present.
On rare occasion, the neonate may have progressive
instability after initiating PGE1 therapy. This important
diagnostic finding strongly suggests a congenital heart
defect with obstructed blood flow out of the pulmonary
veins or the left atrium. These lesions include (1) hypoplastic left heart syndrome with restrictive foramen ovale
or intact atrial septum, (2) other variants of mitral atresia
with a restrictive foramen ovale, (3) transposition of the
great arteries with intact ventricular septum and restrictive
foramen ovale, and (4) total anomalous pulmonary venous
return with obstruction of the common pulmonary vein.
If the neonate clinically deteriorates despite PGE1
therapy, urgent echocardiography with plans for potential
interventional cardiac catheterization or cardiac surgery
need to be made. Early recognition of this deterioration
despite PGE1 requires prompt transfer to a cardiac unit
(Penny and Shekerdemian, 2001). Controversy exists as to
whether PGE1 should be continued in these rare instances;
it has generally been the authors’ practice to continue the
infusion at the usual dose. More important than the decision to continue or discontinue PGE1 is that the lack
of response to PGE1 in a child with suspected CHD is
a marker for rare forms of CHD that do not respond to
medical management and require urgent surgical or catheter intervention.
Although PGE1 is critical in the management of most
neonates with CHD, there are a number of potential adverse
effects associated with PGE1 continuous infusion that must
be anticipated, particularly in the premature or LBW infant,
in which they occur more commonly. The most common
adverse effects include hypotension (caused by vasodilation), apnea, rash, and fever (Kramer et al, 1995; Lewis
et al, 1981). Other less common side effects include seizures, gastric outlet obstruction, cortical hyperostosis, and
leukocytosis (Arav-Boger et al, 2001; Teixeira et al, 1984).
Although high-dose PGE1 infusions may occasionally
be required, most often neonates are in stable condition
receiving a low-dose infusion. The resultant vasodilation
and hypotension is less common and usually treatable with
fluid resuscitation rather than inotropic support (Kramer
et al, 1995). A separate intravenous catheter is typically
necessary for the purpose of volume administration and
should be considered prior to transport. A 5 to 10 mL/kg
bolus of normal saline, lactated Ringer’s solution, or 5%
albumin will generally normalize the blood pressure in
cases of hypotension related to PGE1. Serum ionized calcium levels should be checked and normalized if low. If
hypotension is refractory to fluid administration, an alternative cause of hypotension should be considered (e.g.,
a restrictive ductus, pericardial effusion, myocardial dysfunction, sepsis), and dopamine (3 to 5 μg/kg/min) may be
given to offset the vasodilatory effects of PGE1.
If apnea and hypotension occur, they will usually manifest during the first hours of administration, but may occur
at any time during the infusion. This occurrence mandates
the need for ongoing cardiorespiratory monitoring even
after stabilization on PGE1 infusion. Furthermore it has
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
been shown that, although the side effect profile (particularly apnea) increases with increasing doses and lower
weight, the clinical response is not dose dependent. In
other words, infants can be maintained safely on a lowdose PGE1 infusion until definitive care is achieved and
thereby potentially avoid the significant side effects of
apnea and hypotension. Certainly, PGE1 therapy alone
does not require mechanical ventilation without the presence of significant or recurrent apnea (Browning et al,
2007; Kramer et al, 1995) or other clinical states requiring mechanical ventilation. The use of low-dose PGE1
(<0.015 μg/kg/min) for duct-dependent lesions is unlikely
to cause apnea requiring mechanical ventilation, and it is
safe to transport the infant without establishing an artificial airway in most cases (Carmo and Barr, 2005).
When starting PGE1 infusion, it is prudent to remeasure arterial blood gases and reassess vital signs and perfusion within 15 to 30 minutes of initiation. Doing so allows
effectiveness to be assessed and any adverse events to be
addressed promptly.
Intubation and Ventilation
Although the initiation of PGE1 alone does not require the
neonate to be intubated, the presence of profound hypoxemia, respiratory distress, or hemodynamic instability may
demand airway intervention. In most cases, intubation
should be performed after premedication with sedation
(narcotic or benzodiazepine), preferably with neuromuscular blockade. Although intubation can be performed
without sedation and neuromuscular blockade, there are
physiologic reasons to use these agents for the intubation
of a neonate with CHD. First, the catecholamine surge
that will occur with a nonsedated intubation may result
in significant dysrhythmias in the at-risk myocardium.
Second, vagally mediated bradycardia from hypoxemia,
hypercapnia, or laryngeal stimulation may lead to asystole
in these neonates, who have little reserve. Finally, sedation and neuromuscular blockade will reduce total body
oxygen consumption, raising the mixed venous oxygen
saturation and improving oxygen delivery. Premedication
with atropine (0.02 mg/kg) can blunt the vagal effects of
laryngoscopy. Fentanyl (1 to 2 μg/kg) or midazolam (0.05
to 0.1 mg/kg) for sedation may be given, with titration of
dose to effect. Chest wall rigidity may occur with low-dose
fentanyl (more likely if given rapid push) and may require
neuromuscular blockade for adequate ventilation.
In most cases, the neonate is preoxygenated with 100%
FiO2, which can be down-titrated to achieve an acceptable oxygen saturation given the underlying CHD after
intubation. In an effort to maintain a balanced circulation,
ventilation should attempt to achieve normocarbia (Pco2,
35 to 40 mm Hg). In the single-ventricle patient (i.e., a
patient in whom the systemic and pulmonary circulations
are dependent on a single-ventricular pumping chamber)
or the neonate with unrestrictive ventricular or great vessel communications, significant adjustments in ventilation
will alter pulmonary vascular resistance, and therefore may
have an effect on both pulmonary and systemic blood flow.
Hyperventilation in the neonate decreases pulmonary vascular resistance, thereby increasing pulmonary blood flow
potentially at the expense of systemic blood flow.
353
Although intubation and ventilation may be necessary interventions, it should be noted that preoperative
mechanical ventilation is a risk factor for mortality or
poor outcome after surgery for CHD in multiple studies
(Gottlieb et al, 2008; Robertson et al, 2004; Simsic et al,
2007; Tabbutt et al, 2008).
Inotropic Therapy
The neonate with CHD discharged from the nursery
before diagnosis may be experiencing congestive heart failure or circulatory collapse. These infants, while urgently
requiring the opening of the ductus arteriosus to provide
either systemic or pulmonary blood flow, may also require
inotropic therapy to recover from the adverse effects of a
myocardium that has decompensated in the face of high
afterload (obstructive left-sided lesions requiring ductal
patency for systemic blood flow) or hypoxemia (obstructive right-sided lesions requiring ductal patency for pulmonary blood flow). Appropriate volume status should be
achieved in conjunction with the institution of inotropic
therapy and secure access for its delivery. The choice of
inotropic agent remains largely practitioner dependent.
Sympathomimetic amines are the most commonly used
inotropic agents. They can be endogenous (dopamine and
epinephrine) or synthetic (dobutamine and isoproterenol).
Dopamine is a norepinephrine precursor and stimulates
dopaminergic, β1-adrenergic, and α-adrenergic receptors
in a dose-dependent manner. Dopamine improves myocardial contractility, which will increase stroke volume. This
increase leads to improved cardiac output and higher mean
arterial pressure, with resultant increased urine output.
There is a low incidence of side effects at doses less than
10 μg/kg/min. Dobutamine is an analogue of dopamine;
it stimulates β1-adrenergic receptors predominantly, with
relatively weak β2-adrenergic receptor and α-adrenergic
receptor activity. No definitive benefit has been found in
the use of dopamine versus dobutamine, although dopamine is more likely to increase the systemic blood pressure
in the short term. No differences in outcomes or mortality were found between the two inotropes (Subhedar and
Shaw, 2003). Both drugs are generally initiated at 3 to 5
μg/kg/min. Epinephrine may also be used in neonates with
hypotension and hemodynamic deterioration. Epinephrine has α1-, α2-, β1-, and β2-adrenergic effects. Few trials have been done to recommend epinephrine over other
agents. A randomized controlled trial comparing epinephrine and dopamine in LBW infants found equal efficacy
in treating hypotension (Valverde et al, 2006). The recommended starting dose is 0.03 to 0.05 μg/kg/min. Isoproterenol stimulates β1- and β2-adrenergic receptors. It
has greater chronotropic effect and stronger vasodilatory
effect because of the β2 stimulation. It needs to be started
at low dose and titrated to effect, because of the strong
chronotropic effect. Chronotropic effects will precede
inotropic effects in a responsive heart and can produce
tachyarrhythmias. Recommended starting dose is 0.01 to
0.05 μg/kg/min.
The adverse effects of the sympathomimetic amines
include tachycardia, atrial and ventricular arrhythmias, and peripheral vasoconstriction causing increased
afterload. Tachycardia will increase myocardial oxygen
354
PART VIII Care of the High-Risk Infant
consumption, whereas arrhythmias and vasoconstriction
decrease cardiac output.
Vascular Access
Umbilical venous catheters (UVCs) and umbilical arterial
catheters (UACs) are useful in the stabilization, transport,
and preoperative and postoperative management of the
neonate with CHD. However, the placement of these lines
is not without risk. Beyond the infectious risks, newborns
with CHD are at a higher risk for thromboembolic events
because of their underdeveloped clotting mechanisms,
small vessel lumens, and low flow states. There is also an
ongoing risk of systemic embolization of air and particulate
matter in babies with an intracardiac right-to-left shunt.
There has been much controversy surrounding the optimal placement of a UAC in premature infants, although
there are scant data regarding full-term infants with CHD.
A Cochrane Database review suggested that “high” catheters were associated with a decreased risk of vascular complications with no significant increase in adverse sequelae
(Barrington, 2000; Hermansen et al, 2005). Although there
is variation in definition, in general “high” catheters are
above the level of the diaphragm and below the level of the
left subclavian artery (approximately T6 to T10); “low”
catheters are below the renal arteries but above the aortic bifurcation (approximately L3 to L5). Levels between
high or low are associated with increased risks of complications, as are placements below the L5 level (Hermansen
et al, 2005). Although transient placement of a UAC in this
area will generally be well tolerated, long-term placement
should be avoided if possible. The UVC should be placed,
if possible, at the inferior vena cava/right atrial junction
or in the atria. It might not be necessary to obtain ideal
placement of either the UAC or UVC for stabilization
and transport, and manipulation of lines and reconfirmation can delay transport and stabilization to the tertiary
care center. The risk-benefit relation for the use of umbilical lines in the neonate with ductal dependent circulation
has not been well delineated. The need for central access
should be judged based on the clinical status of the neonate
and stability for transport.
Transport of the Neonate
With Congenital Heart Disease
Once the neonate with suspected or known CHD has
been sufficiently resuscitated and stabilized, they should
be transferred to an institution that provides subspecialty
care in pediatric cardiology and pediatric cardiothoracic
surgery. In a single tertiary center study of neonates presenting with CHD, 157 of 190 patients required at least
one surgical or catheter intervention with a median of
3 days of life at time of surgery (Dorfman et al, 2008).
To optimize management, early communication with the
specialist center is vital. Successful transport involves two
phases: referring hospital staff to the transport team and
subspecialists, and transport staff to the accepting hospital
staff. The need for this precise and thorough communication between respective teams cannot be overemphasized.
Whenever possible, the pediatric cardiologist, neonatologist, or intensivist at the accepting hospital should be
included in formulating the transport management plan
while the neonate is still at the referring hospital. This
procedure will help to guide the timing and urgency of
the transport, line placement, and recommendations for
airway management and supplemental oxygen. Often the
patient with a duct-dependent lesion will improve greatly
with the institution of prostaglandin therapy and may not
need to be rushed to the cardiac referral center as an emergent case.
Similar supports and treatments need to be put into
place before transport of any critically ill neonate or
infant. Secure vascular access should be obtained, with a
port available for volume resuscitation that is not running
the inotropic support or PGE1 infusion, to avoid interruption of this medication. Intubated patients should have the
airway position recorded and secured and have a nasogastric or orogastric tube for decompression. The intubated
infant should remain unfed, and medication administration and fluids should be given intravenously. Appropriate
sedation should be used, as well as maintenance of normothermia and avoidance of hyperthermia, to minimize
oxygen consumption.
Evaluation of acid-base status, oxygen delivery, temperature, serum glucose, and calcium should take place before
transport and should be corrected when possible. Neonates with conotruncal anomalies are at particular risk for
hypocalcemia, because they have an increased frequency
of 22q11 deletion syndrome (Gallot et al, 1998), which
may have associated thymic and parathyroid hypoplasia.
Furthermore, the neonatal myocardium may be more
dependent on calcium for inotropy than the adult. Ionized
calcium levels less than 1 mg/dL may have a significant
negative effect on contractility and should be considered
for treatment with calcium gluconate (50 to 100 mg/kg)
or calcium chloride (0.1 mEq/kg of elemental calcium or
0.01 g/kg) intravenously, via a central catheter if possible.
In patients with mixing lesions, single ventricle or in
those with an unknown diagnosis, supplemental oxygen
should be used and titrated to maintain pulse oximeter oxygen saturation of 75% to 85% for “balance” of circulation
between pulmonary and systemic systems and to provide
adequate oxygen delivery, because higher oxygen saturations may actually be deleterious, as noted previously. The
accepting pediatric cardiologist can aid with lesion-specific
advice and guidelines to optimize in-transport care.
Hypotension is a late finding of shock in neonates. Earlier, more sensitive signs, of impending decompensation
include persistent tachycardia despite adequate intravascular volume and temperature control, poor tissue perfusion,
and metabolic acidosis. Treatment of shock should occur
before transport during the stabilization phase of management, although the patient in profound shock and with
significant acidosis at presentation may require significant
time for resolution of organ dysfunction and perfusion and
clearance of acidosis. Finally, failure to respond to prostaglandin, assuming adequate dose and delivery, should
promote prompt transfer to a cardiac unit able to care for
lesions with obstruction to pulmonary venous outflow.
Before leaving the referring hospital, hemodynamic status
(capillary refill, heart rate, systemic blood pressure, and
acid-base status) needs to be reassessed and communicated
to the accepting hospital.
CHAPTER 29 Stabilization and Transport of the High-Risk Infant
Summary of Cardiac Transport
Stabilization and transport of the neonate with known or
suspected critical CHD affect their preoperative condition, potentially contributing to short-term mortality risk
and long-term morbidity. One key to stabilization is the
recognition of potential or confirmed CHD, appropriate
resuscitation, airway management where indicated, stable
vascular access, judicious oxygen utilization, PGE1, and
inotropic support when necessary. Early consultation with
a center specializing in pediatric cardiac care, for advice
and timing of transport, will aid the initial care and stability of the infant on transport. Accurate, detailed information must be communicated among the referring hospital,
the transport team, and the accepting hospital.
SURGICAL EMERGENCIES
Because most births occur in hospitals without an NICU or
neonatal surgical abilities, the need for surgical evaluation
or intervention is a common reason for interfacility transport. Whereas some surgical conditions are relatively not
urgent in nature, there are several diagnoses that represent
truly life-threatening conditions for which stabilization
and transport require expertise and specialized resources.
Congenital Diaphragmatic Hernia
Advances in ultrasound technology have resulted in the
prenatal diagnosis of up to 60% of fetuses affected by congenital diaphragmatic hernia (Gallot et al, 2007). A recent
systematic review showed that outcome was improved for
newborns with a prenatal diagnosis or born in a tertiary
care center (Logan et al, 2007b). Newborns with undiagnosed CDH, especially those delivered at a community
hospital, are at high risk for complications. Before transport, critically ill infants with suspected or known CDH
should undergo tracheal intubation and placement of a
large-bore nasogastric or orogastric tube to continuous
suction, because many have received bag-mask ventilations
and swallowed air can quickly travel beyond the pylorus
and distend the intrathoracic intestinal contents (GrisaruGranovsky et al, 2009; Logan et al, 2007b). Whenever possible, the same principles of management that are used in
the tertiary care center should be used during transport:
limitation of peak airway pressures and use of low tidal volumes to avoid ventilator-induced lung injury, judicious use
of sedation, avoidance of chemical paralysis to permit spontaneous breathing, and maintenance of adequate systemic
blood pressure with the use of fluid therapy and inotropes
(Logan et al, 2007a). Although there is still controversy
over the role of extracorporeal membrane oxygenation for
newborns with CDH, transport to a high-volume center
with ECMO capabilities should be strongly considered
(Grushka et al, 2009; Morini et al, 2006).
Abdominal Wall Defects
The proper management of a newborn with gastroschisis
or omphalocele is critical during the first several hours of
life, and delivery in a tertiary care center has been associated with improved outcome (Quirk et al, 1996). In
355
addition to the need for initial resuscitation and cardiorespiratory support, the correct treatment of the exposed
bowel or sac may improve the infants’ chances of a successful repair and long-term intestinal function.
The mean gestational age for newborns with gastroschisis is 36.6 weeks, and many affected infants are small for
gestational age (Baerg et al, 2003; Lausman et al, 2007). As
with other mildly premature and growth-restricted infants,
patients with gastroschisis are at risk for hypothermia and
hypoglycemia. Heat loss is exacerbated by the large surface area of the exposed intestines, which can also serve
as a significant source of fluid loss. Prevention of heat and
fluid losses can be accomplished by placing the lower part
of the infant’s body, including the intestines, into a transport bag (i.e., bowel bag or Lahey bag) before placement
of the infant into the heated transport isolette. Significant
fluid losses can occur through the exposed mucosa, and the
patient may require aggressive fluid replacement (120 to
150 mL/kg/day). The use of antibiotics should be considered if risk factors for sepsis are present and reviewed with
the pediatric surgeon.
Infants with gastroschisis are at risk for intestinal vascular compromise, because the vascular pedicle containing the arterial supply and venous drainage from the
bowel must pass through the relatively small abdominal
wall defect. Transport personnel must closely monitor the
appearance of the bowel to detect signs of venous congestion or ischemia. Transporting the infant in the lateral
position, with support of the exposed intestines to avoid
tension or torque, is recommended. The use of intestinal pulse oximetry has been described for monitoring the
bowel for ischemia through a transparent silo, but has not
been studied as a tool during interfacility transport (Kim
et al, 2006). Vascular compromise of the intestine is a surgical emergency, and communication with the receiving
facility is essential to coordinate urgent intervention.
The transport of an infant with omphalocele has similar
considerations, although unless the sac has ruptured there
is significantly less risk of heat and fluid loss. Infants with
an omphalocele are more likely than those with gastroschisis to have other birth defects (e.g., CHD). Furthermore,
infants with giant omphaloceles often have respiratory
insufficiency caused by diaphragmatic dysfunction, pulmonary hypoplasia, or both, and they may require ventilatory
assistance.
All infants with gastroschisis or omphalocele require
placement of a large-bore nasogastric or orogastric tube
due to functional ileus or intestinal obstruction, as may
occur with associated stenoses or atresias. In general,
cannulation of the umbilical vessels is not recommended
unless other methods for vascular access are not successful.
Esophageal Atresia and
Tracheo Esophageal Fistula
Esophageal atresia, with or without tracheo esophageal
fistula, is typically diagnosed within the first day of life
because of increased secretions, poor feeding, and respiratory distress. General transport considerations include
placement of a large-bore sump-type tube for continuous
aspiration of the proximal esophageal pouch, positioning (prone with the head of bed elevated), and respiratory
356
PART VIII Care of the High-Risk Infant
support as indicated. Direct aspiration of secretions into
the trachea may occur with either a proximal or distal tracheo esophageal fistula. Transport providers should be
aware that infants with a distal tracheo esophageal fistula
(type C), characterized by the presence of air in the intestinal tract, are at risk for gastric and intestinal insufflation
via the fistula when receiving positive-pressure ventilation.
Bag-mask ventilation and continuous positive airway pressure should be avoided. If the infant requires endotracheal
intubation, the endotracheal tube should be positioned as
close to the carina as tolerated in an effort to position the
distal tip beyond the fistula and minimize direct inflation
of the distal esophageal segment with pressurized gas. In
extreme cases, gastric rupture with pneumoperitoneum
has been reported, requiring emergency paracentesis, laparotomy, or both (Maoate et al, 1999).
Midgut Volvulus
Malrotation with midgut volvulus can be a catastrophic
event resulting in intestinal ischemia and shock, and it
represents a surgical emergency in the neonate. The
most common clinical presentation of midgut volvulus is
bilious vomiting, which is a nonspecific sign of intestinal
obstruction. Expeditious evaluation of the newborn with
bilious vomiting is essential to facilitate prompt surgical
intervention in the event that midgut volvulus is identified,
to prevent progression of vascular insufficiency to actual
intestinal necrosis. An upper gastrointestinal series is the
radiologic test of choice to diagnose malrotation and midgut volvulus, although some practitioners have reported
success with the use of ultrasound examination to identify
the relationship of the superior mesenteric vessels (Lampl
et al, 2009; Shew, 2009).
An infant with suspected midgut volvulus should be rapidly transported to a facility with pediatric radiology and
surgical capabilities. Care of the infant with suspected midgut volvulus during interfacility transport is primarily supportive and includes circulatory support with intravenous
fluid repletion, correction of metabolic abnormalities, and
gastric decompression with a large-bore nasogastric or
orogastric tube.
Necrotizing Enterocolitis
Necrotizing enterocolitis (NEC) is common in premature
infants, and approximately 30% of newborns with NEC
will require surgical intervention in the form of laparotomy or peritoneal drain placement (Guthrie et al, 2003).
The clinical presentation of NEC is typically nonfocal and
often mimics signs of systemic sepsis. Clinical and radiologic findings that are specific to NEC include abdominal
distention and discoloration of the abdominal wall, dilated
or thickened bowel loops, pneumatosis intestinalis, portal
venous gas, and free intraperitoneal air.
Infants with suspected NEC should be transported to
a facility with pediatric surgical capabilities. Care during
transport is primarily supportive and includes intravenous fluids, administration of broad-spectrum antibiotics,
correction of metabolic abnormalities, and gastric decompression. Respiratory failure is common because of disordered control of breathing and elevation of the diaphragm
from abdominal distension.
Meningomyelocele
Although most newborns with neural tube defects are
diagnosed prenatally because of an elevated AFP and are
therefore delivered in a tertiary care center, the unexpected birth of an infant with a meningomyelocele may
be an indication for neonatal transport. For purposes of
transport, the infant should be placed in the prone position and the spinal defect should be covered with moist
sterile dressings as well as some form of plastic wrap to
maintain moisture. The lesion can be covered with a
moistened Telfa dressing and then loosely encircled with a
Kerlix “donut,” with the entire defect covered with a sterile drape. This dressing can be moistened as indicated during the transport process (Jason and Mayock, 1999). Avoid
the use of latex gloves during care of these patients. If the
skin covering the defect is disrupted, there is an increased
risk of infection, and empiric antibiotics should be considered. Infants with meningomyelocele may or may not have
accompanying hydrocephalus at birth; approximately 25%
of affected patients will require shunting in the immediate
newborn period, with up to 85% eventually undergoing
shunt placement (Bowman et al, 2001).
SUGGESTED READINGS
Browning Carmo KA, Barr P, West M, et al: Transporting newborn infants with
suspected duct dependent congenital heart disease on low-dose prostaglandin
E1 without routine mechanical ventilation, Arch Dis Child Fetal Neonatal Ed
92:F117-F119, 2007.
Das UG, Leuthner SR: Preparing the neonate for transport, Pediatr Clin N Am
51:581-598, 2004.
Dorfman AT, Marino BS, Wernovsky G, et al: Critical heart disease in the neonate:
presentation and outcome at a tertiary care center, Pediatr Crit Care Med 9:
193-202, 2008.
Engle WA: American Academy of Pediatrics Committee on Fetus and Newborn:
Surfactant-replacement therapy for respiratory distress in the preterm and term
neonate, Pediatrics 121:419-432, 2008.
King BR, Woodward GA: Procedural training for pediatric and neonatal transport
nurses: Part I: - training methods and airway training, Pediatr Emerg Care
17:461-464, 2001.
Orr RA, Felmet KA, Han Y, et al: Pediatric specialized transport teams are associated with improved outcomes, Pediatrics 124:40-48, 2009.
Phibbs CS, Baker LC, Caughey AB, et al: Level and volume of neonatal intensive care and mortality in very-low-birth-weight infants, N Engl J Med 356:
2165-2175, 2007.
Polin RA, Randis TM, Sahni R: Systemic hypothermia to decrease morbidity of
hypoxic-ischemic brain injury, J Perinatol 27:S47, 2007.
Silverstein FS, Jensen FE: Neurological progress: neonatal seizures, Ann Neurol
62:112, 2007.
Thangaratinam S, Daniels J, Ewer AK, et al: Accuracy of pulse oximetry in screening for congenital heart disease in asymptomatic newborns: a systematic review,
Arch Dis Child Fetal Neonatal Ed 92:F176-F180, 2007.
Woodward GA, Insoft RM, Kleinman ME, editors: Guidelines for air and ground
transport of neonatal and pediatric patients, Elk Grove Village, Ill, 2007, American
Academy of Pediatrics.
Woodward GA, King BR, Garrett AL, et al: Prehospital care and transport medicine. In Fleisher G, Ludwig S, Henretig F, editors: Textbook of pediatric emergency medicine, ed 5, Philadelphia, 2006, Lippincott, Williams, and Wilkins,
pp 93-134.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
30
Temperature Regulation of the
Premature Neonate
Stephen Baumgart and Sudhish Chandra
The human neonate is a homeothermic mammal. Even the
smallest premature infants respond adaptively to changes
in their environment. Response, however, may be insufficient to maintain core body temperature and can render
preterm babies functionally poikilothermic, even in moderately temperate environments. Morbidity (e.g., poor
brain and somatic growth) and mortality rates increase
when core body temperature is permitted to decline much
below 36° C (96.8° F), and moderate to severe hypothermia below 31° C (88.7° F) results in precipitous declines in
heart rate and blood pressure.
COLD STRESS
Evaporative heat loss is widely as regarded the most stressful cooling event upon birth. Severe nonevaporative heat
loss is also problematic for several reasons. First, a baby’s
exposed body surface area is much larger than an adult’s,
relative to metabolically active body mass (Table 30-1).
Especially for the extremely low birthweight infant, the
heat-dissipating area is fivefold to sixfold greater proportionate to that of the adult. Second, the baby’s small size
presents a much smaller heat sink to store thermal reserve.
Finally, the radius of curvature of the body is less than in
the adult, resulting in a thinner protective boundary layer
of warm, still air.
Aside from these geometric considerations, characteristics of the premature infant’s skin contribute to the
problem of excessive heat loss. The skin and subcutaneous
fascia provide little insulation against the flow of heat from
the core to the surface. Moreover, the lack of a keratinized epidermal barrier exposes infants to vastly increased
evaporative heat loss.
Finally, premature infants may not induce effective thermogenesis in response to cold stress. Shivering and nonshivering thermogenesis are compromised by low brown
fat stores. Furthermore, the presence of hypoxia (common
with preterm birth) seriously reduces nonshivering thermogenesis by reducing mitochondrial oxidative capacity.
PHYSICAL ROUTES OF HEAT LOSS
CONVECTION
Convective heat loss in newborns occurs when ambient
air temperature is less than the infant’s skin temperature.
Convective heat loss includes natural convection (passage
of heat from the skin to the ambient still air) and forced
convection, in which mass movement of air over the infant
conveys heat away from the skin. The quantity of heat lost
is proportional to the difference between air and skin temperatures, and to air speed. The effect of forced convection
in disrupting the microenvironment of warm, humid air
layered near an infant’s skin usually is not appreciated in
the nursery, where drafts, air turbulence, and consequently
heat loss can occur within the relatively protective environment of an incubator.
EVAPORATION
Passive transcutaneous evaporation of water from a newborn’s skin (insensible water loss) results in the dissipation
of 0.58 Kcal/mL of latent heat. As shown in Figure 30-1,
transcutaneous water loss increases exponentially with
decreasing body size and gestation. The tiniest premature
baby, who is least able to tolerate cold stress, can incur
evaporative heat loss in excess of 4 Kcal/kg/hr (water loss
of approximately 7 mL/kg/hr). Evaporation is enhanced
by low vapor pressure (i.e., high air temperature and low
relative humidity) and air turbulence. The highest evaporative losses occur on the first day of life. During the first
week of life in infants born at 25 to 27 weeks’ gestation,
evaporative heat losses may be higher than radiant losses
(Hammarlund et al, 1986).
RADIATION
Radiant heat loss constitutes the transfer of heat from an
infant’s warm skin, via infrared electromagnetic waves,
to the cooler surrounding walls that absorb heat. Radiant heat loss is proportional to the temperature gradient
between the skin and surrounding walls. An infant’s posture may affect radiant heat loss by increasing or reducing
the exposed radiating surface area. In a moderately humid
environment (relative humidity approximately 50%),
babies experience an ambient temperature (termed operant temperature) determined 60% by wall temperature and
40% by air temperature.
CONDUCTION
Conductive heat loss to cooler surfaces in contact with an
infant’s skin depends on the conductivity of the surface
material and its temperature. Usually babies are nursed on
insulating mattresses and blankets that minimize conductive heat loss.
PHYSIOLOGY OF COLD RESPONSE
AFFERENTS
Homeothermic response to a cold environment begins
with the sensation of temperature. Traditional physiology identifies two temperature-sensitive sites: the hypothalamus and the skin. Sensation of cold by neonatal skin
triggers a cold-adaptive response long before core sensors
357
358
PART VIII Care of the High-Risk Infant
7
TABLE 30-1 Body Surface Area-to-Body Mass Ratio
Adult
70
Surface
Area
(m2)
Ratio
(cm2/kg)
1.73
250
Premature infant
1.5
0.13
870
Very premature infant
0.5
0.07
1400
in the hypothalamus become chilled. Some investigators
conjecture that neonatal cold reception resides primarily
in the skin, whereas warm reception resides in the hypothalamus. Both sensors are probably integrated, because
cold sensory response is inhibited by core sensor hyperthermia and vice versa. Peripheral skin cold sensation is
teleologically important, because early detection of heat
loss from the skin aids in the infant’s timely response for
maintaining core temperature.
CENTRAL REGULATION
Integration of multiple skin temperature inputs probably
occurs in the hypothalamus; however, no single control
temperature seems to exist. Under different environmental
conditions, temperature of the skin can fluctuate 8° to 10° C,
and temperature of the hypothalamus may vary ±0.5° C.
There are also diurnal temperature fluctuations, variations with general sympathetic tone, and blunted regulation with asphyxia, hypoxemia, and other central nervous
system defects. Premature infants can regulate core temperature near 37.5° C (99.5° F), whereas term infants may
respond by maintaining 36.5° C (97.7° F). Because important thermoregulatory processes are triggered by deviations of as little as 0.5° C at any temperature-sensitive site,
environmental temperature homeothermy is important.
EFFERENTS
The effector limb of the neonatal thermal response is
mediated primarily by the sympathetic nervous system,
although infant behavior may also be involved. The earliest maturing response is vasoconstriction in deep dermal
arterioles, resulting in reduced flow of warm blood from
the infant’s core into the exposed periphery. In addition,
reducing blood flow effectively places a layer of insulating fat between the warm core tissue compartment and the
cooler exposed skin surface in the term infant. Reduced
fat content in babies with low birthweight diminishes this
effective insulating property. Vasoconstriction nevertheless remains the newborn’s first line of thermally insulating defense, and the response is present even in the most
premature infant.
Brown fat constitutes a second sympathetic effector
organ that provides a metabolic source of nonshivering
thermogenesis. (Babies do not shiver like adults to generate heat, because muscle constriction-relaxation fibers are
not yet myelinated.) Brown fat located in axillary, mediastinal, perinephric, and other regions of the newborn is
particularly enervated and equipped with an abundance
of mitochondria to hydrolyze and reesterify triglycerides
IL 28.04e1.73(Wt)
r 0.90
p 0.001
6
Insensible water loss, ml/kg/hr
Body
Weight
(kg)
5
4
3
2
1
0
0
0.4
0.8
1.2
1.6
2.0
2.4
Weight, kg
FIGURE 30-1 Exponential increase in evaporative water loss from
skin of infants with very low birthweight who were nurtured under
radiant warmers. (Adapted from Baumgart S, et al: Fluid, electrolyte and
glucose maintenance in the very low birthweight infant, Clin Pediatr
21:199-206, 1982.)
and to oxidize free fatty acids. In the term infant, these
reactions are exothermic and may increase metabolic rate
by twofold or more. Preterm babies, however, have little
brown fat and may not be capable of more than a 25%
increase in metabolic rate despite the most severe cold
stress (Hull, 1966).
Finally, recent evidence suggests that control of voluntary muscle tone, posture, and increased motor activity
with agitation may serve to augment heat production in
skeletal muscle via glycogenolysis and glucose oxidation.
Clinical observations of infant posture, behavior, and skin
perfusion and measurements of skin and core temperature
gradient may ultimately provide the most useful guidelines
for assessing infant comfort during incubation.
MODERN INCUBATION: INCUBATORS
AND RADIANT WARMERS
The lifesaving requirement of an appropriate thermal
environment was demonstrated conclusively by Day et al
(1964) and further defined by Silverman et al (1966). Minor
changes in heat balance create an oxygen and energy cost,
inducing an increased metabolic rate that can be met only
by increased ventilation or increased inspired oxygen and
appropriate cardiovascular response to provide oxygen
delivery to activated tissues.
THERMAL NEUTRAL ZONE
The thermal neutral zone is a narrow range of environmental temperatures within which an infant’s metabolic rate
is minimal and normal body temperature is maintained.
359
CHAPTER 30 Temperature Regulation of the Premature Neonate
°F
°C
TABLE 30-2 Mean Temperature Needed to Provide Thermal
36
Baby weighing 1 kg at birth
Neutral thermal environment
96
95
Neutrality for a Healthy Baby Nursed Naked in
Draft‑Free Surroundings of Uniform Temperature
and Moderate Humidity during the Days or Weeks
after Birth
35
94
93
Operative Environmental Temperature*
Birthweight (kg)
34
92
91
90
35° C
34° C
33° C
32° C
For 10
days
After 10
days
After 3
weeks
After 5
weeks
1.5
For 10
days
After 10
days
After 4
weeks
2.0
For 2
days
After 2
days
After 3
weeks
For 2 days
After 2
days
1.0
33
32
Baby weighing 2 kg at birth
89
0
5
10
15
20
25
30
35
Age in days
FIGURE 30-2 The range of temperature needed to provide neutral
environmental conditions for a baby lying naked on a warm mattress in
draft‑free surroundings of moderate humidity (50% saturation) when
mean radiant temperature is the same as air temperature. The hatched
areas show the average neutral temperature range for a healthy baby
weighing 1 or 2 kg at birth. Optimal temperature probably approximates
the lower limit of neutral range as defined here. Approximately 1° C
should be added to these operative temperatures to derive the
appropriate neutral air temperature for a single‑walled incubator when
room temperature is less than 27° C (80° F), and more should be added
if room temperature is significantly less. ( Adapted from Hey EN, Katz G:
The optimum thermal environment for naked babies, Arch Dis Child
45:328-334, 1970.)
When thermally neutral, infants regulate temperature
through vasomotor tone alone without regulatory changes
in metabolic heat production. A range of critical environmental temperatures relevant to modern incubators was
clearly articulated by Hey and Katz (1970; Figure 30-2,
Table 30-2). Below this range, an increase in the infant’s
minimal metabolic rate was observed; therefore the thermal
neutral temperature range was defined as the optimal incubator operating (operant) temperature. Several important
considerations in regulating incubator temperature were
included in these studies: (1) incubator wall temperature was
maintained identical to air temperature, (2) relative humidity was controlled near 50%, and (3) the environment was
maintained in a steady state, uninterrupted by turbulence
or invasion of the incubator’s enclosed perimeter.
Rigid application of older air temperature recommendations should be modified. Many modern incubators
incorporate a double-walled design that results in lower
radiant heat loss to colder incubator walls encountered in
single-walled designs. As a result, slightly cooler air temperature may be required. In addition, many nurseries do
not humidify incubators artificially, fearing the occurrence
of condensation (“rain-out”) resulting in bacterial colonization, particularly around door openings. Finally, it is
important to recognize that the incubator’s steady state
temperature control is frequently interrupted for nursing and medical procedures that require opening doors
to care for the infant. More than 1 hour may be required
to recover prior steady state conditions after such procedures; therefore the thermal neutral zone must be redefined in practical terms. Silverman et al (1966) used a
modified concept of the thermal neutral zone to simplify
>2.5
Data from Hey E: Thermal neutrality, Br Med Bull 31:72, 1975.
*To estimate operative temperature in a single‑walled incubator, subtract 1° C from
incubator air temperature for every 7° C by which this temperature exceeds room
temperature.
clinical application. Reasoning that infants sense environmental temperature first on the skin, electronic negativefeedback (servo-controlled) regulation of the incubator
heater in response to skin temperature was introduced.
These authors demonstrated minimum metabolic expenditure near 36.5° C (97.7° F) abdominal skin temperature as measured by a shielded thermistor in a less rigidly
defined incubator environment. The importance of frequently checking core temperatures (axillary or rectal)
must be emphasized before delegating the infant’s environment to thermostatic control. In addition, Chessex et al
(1988) have demonstrated that incubator temperature can
vary by more than 2° C when skin temperature servo control rather than air temperature control is used, obviating
homeothermic environmental conditions.
Finally, with the modern use of open radiant warmer
beds (improving access to the critically ill premature infant
without interrupting heat delivery) skin temperature servo
control is the only practical method for approximating
the thermal neutral zone (Malin and Baumgart, 1987).
The extension of infant warming to include extremely low
birthweight, critically ill premature babies have generated
new problems for determining a universally accepted optimal environment.
PARTITIONING INFANT HEAT LOSSES
AND HEAT GAINS
Wheldon and Rutter (1982) demonstrated the special
problems encountered in incubating infants with very
low birthweight in a convection-warmed, closed-hood
incubator environment (Figure 30-3). Figure 30-3, A,
demonstrates the thermal balance achieved by a series of
12 infants (mean weight, 1.58 kg). Heat losses to radiation (R), convection (C), and evaporation (E) are modest,
and their sum (Σ) is balanced by the infant’s metabolic
heat production (M). Used in this fashion, the incubator
reduces physical heat losses such that the infant’s minimal
metabolism (larger than any single avenue of heat loss)
360
PART VIII Care of the High-Risk Infant
A
Incubator, 1.58 kg
40
Heat losses
3.0
Heat gains
2.0
1.0
20
Kcal/kg/hr
Heat (watts/m2)
30
10
0
C
E
C
R
M
S
M
2.0
FIGURE 30-4 Partitional calorimetry in 10 critically ill premature
newborns with low birthweight who were nursed under open radiant
warmers. C, Convection; E, evaporation; M, infant’s metabolic heat
production; R, radiation. (Adapted from Baumgart S: Radiant heat loss vs.
radiant heat gain in premature neonates under radiant warmers, Biol Neonate 57:10-20, 1990.)
20
B
Incubator, 1.08 kg
40
30
Heat (watts/m2)
E
1.0
R
10
20
10
0
C
C
R
E
M
10
20
FIGURE 30-3 A, Partition of heat losses and gains in 12 premature
infants nursed within incubators. B, Partition in an infant weighing 1.08
kg at birth. Heat losses to radiation (R), convection (C ), and evaporation
(E ) are modest, and their sum (Σ ) is balanced by the infant’s metabolic
heat production (M ). (Adapted from Wheldon AE, Rutter N: The heat
balance of small babies nursed in incubators and under radiant warmers, Early
Hum Dev 6:131-143, 1982.)
delicately balances minimal physical heat losses within the
controlled thermal environment.
In contrast, a subject with very low birthweight (1.08 kg)
is evaluated in Figure 30-3, B. As the incubator servo control increases warming power to accommodate massive
evaporative heat loss, convective loss becomes a net gain
(negative histogram bar). Radiant loss is diminished by
warm walls inside the incubator. These conditions differ
strikingly from those discussed previously: the incubator
truly warms the infant rather than modestly attenuating convective heat loss, and evaporative heat loss vastly
exceeds the infant’s metabolism. The small infant’s body
temperature is balanced, therefore, between opposing
physical parameters of evaporative and convective heat
transfer. Metabolism plays a secondary role.
The modern use of radiant warmers that are servo controlled to maintain infant abdominal skin temperature
between 36.5° and 37° C (97.7° to 98.6° F) also demonstrates the opposition of physical forces described earlier.
Figure 30-4 demonstrates the heat balance partition for 10
critically ill premature infants (mean weight, 1.39 kg) nursed
on open radiant warmer beds (Baumgart, 1990). Because
ambient room air temperature is 5° to 10° C cooler than
air inside an incubator, convective heat loss is nearly double
the infant’s metabolic heat production. Evaporation adds
to the net physical heat loss. In addition, small amounts of
heat are lost to conduction and radiation (to cooler room
walls). The infant’s metabolism provides only one third of
the energy required to maintain body temperature, with the
majority of heat supplied by the servo-controlled radiant
heat source injecting heat transdermally through skin blood
flow. In this instance, radiant warming (not convection as
in the incubator discussed earlier) delicately balances the
infant’s physical temperature environment. Wheldon and
Rutter (1982) demonstrated similar results in their studies.
HYBRID INCUBATOR-RADIANT
WARMER TECHNOLOGY
A more promising new development in commercially
available incubators is a hybrid design, combining these
two separate warming modes in one device. Manufacturers in the United States and Germany (Air-Shields
[Hatboro, Pennsylvania], Hill-Rom [Batesville, Indiana],
Drager [Lubeck, Germany], and General Electric, Ohmeda [Laurel, Maryland]) have launched such products. In
the incubator mode, movable plastic walls enclose the tiny
premature infant, providing servo-controlled air warming,
while the overhead radiant warmer is incorporated into the
roof of this design but remains off. In the radiant warmer
mode, the plastic walls are dropped down, and the radiant
warmer rises overhead on a motorized pylon and rapidly
turns on to maintain servo-controlled skin temperature
during infant care procedures. The infant is not moved,
and no plastic barrier is placed between the infant and the
radiant heat source when turned on in this mode. The
CHAPTER 30 Temperature Regulation of the Premature Neonate
servo control algorithms, and the integrity of the plastic
enclosure in incubator mode, are critical to the performance of such devices, and the utility of these products
(e.g., survival, quality of life) is still unproved. Published
data are reviewed in the following sections.
VERSALET INCUWARMER
Greenspan et al (2001) tested nine premature lambs, randomized at delivery, to receive incubation from a conventional radiant warming bed (Resuscitaire) with subsequent
transfer into an incubator C550 Isolette or from the
hybrid Versalet 7700 Care System (Air-Shields, Hatboro,
Pennsylvania) in both the warmer bed and the incubator
modes. Deep central and surface temperatures, heart rate,
blood pressure, and blood gas determinations were measured during warming in the radiant warmer bed mode
(Versalet) or on the radiant warmer bed Resuscitaire and
then during transition to the incubator mode (Versalet or
Isolette), and then back to the warmer bed, and bed mode.
The animals conditions all remained clinically stable
throughout the entire transfer protocol on both arms of
the study. Despite careful planning, loss of temperature
probe data occurred when probes became unattached in
the control group during transfers from one device to the
other. There were no significant differences in recorded
temperatures or in pH and blood gasses in either group.
Compared with the standard warming techniques currently used in neonatal intensive care units (NICUs; separate warmer bed for resuscitation and stabilization with
transfer as soon as possible into an incubator device), the
Versalet provided similar thermal and cardiovascular stability without adverse physiologic events during transition
to different modes of warming. The authors state that the
contribution of this hybrid device to the ease of management and improved outcomes in humans needs to be evaluated in a clinical trial (Greenspan et al, 2001; Sherman
et al, 2006). No such trial has yet been conducted, and the
Versalet is currently not in production (Jay Greenspan,
personal communication).
GIRAFFE OMNIBED
As a radiant warmer, the Giraffe OmniBed (General Electric, Ohmeda Division, Laurel, Maryland) evenly heats the
infant’s mattress with a curved reflector surface designed
to distribute heat to the baby and its bed surface without
overwarming bedside caregivers. Babies are warmed uniformly, regardless of their position on the bed platform
surface, which can be rotated 360 degrees, accommodating
intravenous and ventilator tubing and attached wire leads.
The same platform tilts up to 12 degrees in Trendelenburg
or reverse Trendelenburg positions. Three-sided access
from drop-down walls in the radiant warmer mode facilitates procedures such as diaper and bedding changes, blood
sampling, starting intravenous lines, performing tracheal
intubation, administering medications, creating radiographs, and conducting ultrasound examinations without
interrupting warming. A stable thermal environment in
either the incubator or radiant warmer mode eliminates
the stress of moving premature babies, such as when performing chest tube insertion or other surgical procedures
361
performed in the NICU and outside the operating room. In
one of several industry sponsored studies, Leef et al (2001)
reported that infants were handled significantly less with
the Giraffe OmniBed, especially when converted to incubator mode (from 6.9 handling events per hour maximum
on a standard radiant warmer bed device to 1.6 times per
hour on an OmniBed-closed). These authors concluded
that the OmniBed is conducive to providing developmentally appropriate care—that is, medically fragile newborns
are not exposed to a variety of visual, auditory, and tactile
stimuli that would not occur otherwise within the mother’s
womb. Consequences of such stimulation are unknown,
although it seems reasonable to avoid excessive handling
and inappropriate touches because of documented physiologic effects of procedural handling (Gressens et al, 2002).
In a second industry-sponsored study (Gaylord et al,
2001), there were no differences found in mean skin temperature among the four tested conditions in premature
neonates (R = radiant warmer configuration of OmniBed;
transition R to C = convection-warmed closed OmniBed
and transition C to R). Mean heart rate, respiratory rate,
blood pressure, and oxygen saturation were not statistically
different among the four test conditions. These authors
conclude that the Giraffe OmniBed provided thermal and
physiologic stability across bed states, eliminating the risk
of infant mishap as a result of bed transfer.
When transforming the Giraffe OmniBed from incubator to warmer bed and back, the closed-convection heat
partition adapts to form a uniform open-radiant heating
configuration with sequential alterations of air warming
temperature, fan power, and radiant heat delivered while
displaying all equipment and baby parameters in one control panel. For example, when returning to the closedconvection mode, the retracting radiant warmer pylon
immediately disconnects electrical power to the warming
element and opens a mechanical air vent to cool the reflector hood, avoiding overheating the infant upon descent.
In closed-convection configuration, bidirectional airflow through a double wall construction provides a stably
enclosed thermal environment. When either door port is
opened, an air curtain minimizes infant heat loss.
Light and sound levels are carefully controlled within
the OmniBed to promote infant health and development
(Lynam, 2003). An alarm light easily visible to caregivers
remains out of infant’s view. The WhisperQuiet mode
limits sound to create the most quiet and soothing environment possible. Alarm speakers are deflected to minimize any noise experienced by the baby. An in-bed scale
further reduces infant handling.
In addition, servo-regulated humidification is supplied
within the closed-incubator condition and can be set to a
determined relative humidity between 70% and 80%, which
is optimal to avoid excessive insensible water loss and electrolyte disturbances often experienced by premature neonates with extremely low birthweight in the first week of life
when incubated dry. One recent non–industry-sponsored
report of a clinical series compared the use of initial stabilization of babies with extremely low birthweight (<1000 g)
under a radiant warmer followed by conventional incubation—dry versus use of humidity control in OmniBeds.
The authors demonstrated that humidification improved
care by decreasing fluid intake, with more stable electrolyte
362
PART VIII Care of the High-Risk Infant
balance, and growth velocity (Kim et al, 2010). The authors
did not address the risk-benefit issue of humidification and
infection.
The Giraffe Humidifier immerses a heating element in
a reservoir of sterile, distilled water. Water temperature
at equilibrium ranges 52° to 58° C, which is bactericidal
to most organisms thriving at temperatures of 20 to 45° C
(most human pathogens). As an added safety measure
against reservoir contamination, water is boiled off the
immersion element as the humidified air is passed inside
the infant’s compartment. Sterile humidity is created in a
vapor state, with no airborne droplets. In a third industrysponsored study by Lynam and Biagotti (2002), humidified OmniBeds (in vitro, air control mode at 35° C, and
humidified to 65% relative humidity) were cultured after
investigator inoculation with reservoir contamination
with four waterborne pathogens over a 4-week incubating
period. No infant environment culture revealed growth of
any pathogen. The authors concluded that there is no concern for an increased risk of infection to an infant when the
reservoir is filled daily with sterile distilled water and the
bed is routinely cleaned, according to their protocol.
LESS CONVENTIONAL TECHNIQUES
PLASTIC HOODS
Plastic hoods comprise rigid body shields sometimes used
as miniature incubator hoods placed over infants on open
radiant beds. Hoods are usually made of plastic (1 to 3 mm
thick) and can obstruct the delivery of radiant heat to the
baby’s skin when placed between an infant and the radiant
warming element. The plastic can form a heat sink, warming to 42° to 44° C, but disrupts the radiant warmer’s servo
control mechanism and is a poor strategy. In addition, such
devices might not diminish insensible water loss, especially when used without humidity. Use with humidity—a
technique often referred to as swamping, which has never
been validated—encourages bacterial colonization with
water born pathogens.
PLASTIC BLANKETS
Alternatively, a thin, flexible plastic wrap or polyethelene
plastic “blanket” was introduced in 1968 for covering premature infants under radiant warmers (Baumgart, 1984;
Baumgart et al, 1981, 1982a, 1982b). The blanket prevents
convective heat loss and is thin enough to transmit almost
completely radiant heat from the warmer. The flexible plastic molds closely around the infant’s body (with care taken
to avoid skin adhesion), conserves evaporation, and forms a
microenvironment near the baby’s body. The plastic acts as
a mechanical barrier to prevent convective turbulence from
disrupting this microenvironment. A two-thirds reduction
in insensible water (and evaporative heat) loss under radiant
warmers is prevented, resulting in less servo-controlled radiant
heat delivery required to maintain body temperature in even
the smallest infants. By moderating heat losses, oxygen consumption is reduced by approximately 10%. This reduction
exactly matches the oxygen consumption cost of using radiant
warmers reported by LeBlanc (1982). Risks of plastic blankets include sticking to immature skin, causing masceration
and accidental airway obstruction in patients without intubation. In rare cases, partial detachment of the skin thermistor
probe can result in life-threatening hyperthermia. Diligence
is required to avoid these complications. The risk for abnormal bacterial colonization has not been evaluated.
A polyethylene plastic bag was evaluated by Vohra et al
(1999) to promote temperature maintenance during delivery room resuscitation under radiant warmers and during
subsequent transport of infants to the NICU. A significantly higher admission rectal temperature and survival
was reported for infants at less than 28 weeks’ gestation.
A larger, randomized trial of this technique was performed,
confirming the prevention of heat loss with a polyethylene
bag but not the improvement in mortality (Vohra et al,
2004). Recently another large, randomized trial was performed to compare polyethylene caps with polyethylene
bags; it found that both methods were more effective than
conventional treatment in improving NICU admission
temperature for infants at less than 29 weeks’ gestation
(Trevisanuto et al, 2010).
SEMIPERMEABLE POLYURETHANE SKIN
A semipermeable, semiocclusive polyurethane dressing
(Tegederm [3m Corporation, Maplewood, Minnesota] or
Opsite [Smith & Nephew, London, United Kingdom]) can
be applied like an artificial skin to shield premature infants
with very low birthweight. These breathing polyurethane
plastics do not cause skin breakdown and are often used
to dress central venous catheter sites. Temperature probe
detachment is less likely to occur because of skin adherence.
These dressings can also be tailored to avoid airway obstruction. Insensible water loss may be reduced more than 30%
with these dressings, and after carefully removing them the
skin’s naturally developing keratin moisture barrier may
be preserved. Porat and Brodsky (1993) and Bhandari et al
(2005) demonstrated that an adherent polyurethane layer
over the torso and extremities of infants with very low birthweight improved fluid and electrolyte balance, reduced the
occurrences of patent ductus arteriosus and intraventricular hemorrhage, and improved survival. Further study is
required to demonstrate the safety of this technique.
PETROLEUM OINTMENTS
Another occlusive dressing for the skin of preterm infants
with low birthweight during the first week of life is the application of Aquaphor (Biersdorf Co, Hamburg, Germany).
Although early results were encouraging for preserving skin
integrity and perhaps controlling excessive dehydration in
dry incubators, the findings of a multicenter randomized
trial were not so laudable (Edwards et al, 2004). Anecdotal
reports suggest that bacterial infections may actually be
more common with use of this technique. Clinicians should
exercise critical judgment before adopting this practice.
KANGAROO CARE
Kangaroo care connotes warming by skin-to-skin contact with the mother or father, where premature infants
are held naked between the axilla or the breasts, mimicking a kangaroo’s pouch. First implemented in Bogota,
CHAPTER 30 Temperature Regulation of the Premature Neonate
Columbia, this method was popularized for nonintubated
premature infants in Scandinavian and European countries
in the 1980s. Early studies suggested a significant reduction in early mortality and morbidity in premature infants
weighing less than 1.5 kg and who are nursed by kangaroo
care. Behavioral studies demonstrated more stable sleep
patterns, less irritability at 6 months of age, and more eye
contact with caregivers in infants nursed with kangaroo
care. Kangaroo care has been shown to promote a thermal-neutral metabolic response and temperature stability in stably growing premature babies. Moreover during
kangaroo care, infants with bronchopulmonary dysplasia
have better oxygenation, and other infants show less periodic breathing and reduced apnea. In the modern nursery,
kangaroo care be initiated during mechanical ventilation
in uncomplicated patients. The infant should be placed
between breasts with maximum skin contact and should
be covered with a blanket to avoid outward convective and
evaporative heat losses. After initial sessions of 30 minutes
to 1 hour with careful intermittent temperature monitoring, periods up to 4 hours may be achieved successively.
Kangaroo care integrates the family into the neonatal
intensive care team. No adverse reports have been published, and use in many nurseries is on the rise.
SPECIAL CASES
EXTREMELY LOW-BIRTHWEIGHT INFANTS
Case Presentation
A baby boy (234⁄7 weeks’ gestation, weighing 510 g) was
born limp, cyanotic, with no respiratory effort, and a heart
rate less than 100 beats/min. The baby was resuscitated
under a radiant warmer and dried, and a stocking cap was
placed over the head. The warmer’s temperature probe
recorded a low skin temperature because it did not attach
well to skin. The radiant warmer was on full power to
compensate for this low probe reading. After resuscitation, the baby was transported to the NICU in an incubator preheated to a temperature of 35° C. On arrival,
the baby was weighed quickly and placed onto a preheated
radiant warmer bed. Skin probe (servo control) temperature registered 33° C, with a rectal temperature of 35° C
and axillary temperature of 34° C. The radiant warmer
set point was targeted at 37° C, and temperatures were
monitored every 15 minutes. A plastic blanket was used to
minimize heat loss. Heated and 100% humidified air from
a respiratory pack was run underneath the blanket. next,
the baby was prepared with iodine solution and draped
completely for umbilical catheterization (a 30-minute
procedure). Over the first 2 hours of life, rectal temperature increased only to 34.5° C. The baby was transferred
into a preheated and humidified incubator regulated by
a skin probe set at 37° C. Two light bulbs (250 W) were
positioned over the incubator hood for supplemental heat
and for warming during any procedure requiring opening
the incubator (e.g., serial weight determinations). Over
the next 12 hours the baby had difficulty maintaining temperature despite incubator air temperature running near
a set temperature of 37° C, and sometimes greater than
38.5° C.
363
Discussion
Temperature maintenance of an extremely premature
infant should be part of resuscitation from the time of
delivery. Despite radiant warming in the delivery room
and convective incubation during transport, this baby had
hypothermia on admission to the NICU. Low admission
temperature correlates with increased mortality rates in
these infants. They are born wet and prone to excessive
transepidermal evaporative and convective heat losses. In
addition, heat loss can be exacerbated by suboptimal radiant heating; blowing noncontrolled warm, humidified air
under plastic blankets, which is probably not as effective
as the still air envelope conserved by the blanket; and the
pressure for performing procedures with surgical drapes
that block radiant heat delivery.
Quick drying, proper placement directly under the
radiant heater at birth, and covering the head are small
but important steps of temperature resuscitation. Other
techniques might be considered from birth, such a use of
a plastic bag described by Vohra et al (1999) or a plastic
blanket or polyurethane drape during umbilical catheterization. Finally, transferring infants into incubators before
adequately rewarming them under a radiant warmer can
prolong thermal recovery in hypoxic subjects who are
incapable of generating enough metabolic heat to recover.
The use of light bulbs to provide supplemental radiant heat
in incubators attests to the incubator’s intrinsic operating
power deficiency (i.e., seeking to reduce heat loss, rather
than rewarm cold babies). Moreover, such supplemental
radiant heat sources are not controlled, produce more
radiation in the visible light range, and are inefficient and
potentially dangerous for promoting burns if placed too
close to the infant’s skin.
There are few data describing the best rate of rewarming. Cold-stressed babies should probably be nursed
under a servo-controlled radiant heat source, and heat
loss should be minimized. During rewarming the infants
should be closely monitored, and heat delivery should
be servo controlled. A rate of approximately 0.5° to 1° C
per hour seems reasonable, but it might not be achievable. Profound cardiovascular and electrolyte disturbances characterize rewarming. Alternatively, a preheated
and humidified incubator can be used to rewarm infants,
although the rate may be slower. The incubator air or skin
servo control temperature can be set 1° C higher than the
baby’s temperature and gradually increased until a normal
core temperature is achieved. Hybrid incubators that can
also be used as radiant warmer beds are available and are
helpful in rewarming scenarios.
NEONATAL FEVER
Problem of Temperature Elevation
There is no universally accepted definition of neonatal
fever. Craig (1963) defined neonatal pyrexia as a rectal (core
body) temperature greater than 37.4° C; however, other
investigators accept temperatures up to 37.8°C as normal.
Between 1% and 2.5% of all newborns admitted to the
nursery develop fever, judged by rectal or axillary temperatures and depending on the limits chosen. Fever is an
inconsistent and infrequent sign of sepsis (fewer than 10%
364
PART VIII Care of the High-Risk Infant
of febrile neonates have culture-proven sepsis), and temperature elevation may be seen with several other clinical
entities.
Mechanisms Producing Neonatal Fever
Mechanisms producing neonatal fever are understood
incompletely and result from disturbances in the complex
interactions between heat conservation and heat dissipation mechanisms. Fever can occur when immunogenic
pyrogens (commonly prostagladin [PG E2]) leads to
upward displacement of the normal thermal set-point in
the hypothalamus, leading to activation of heat conservation and physiologic heat-generating responses. Generally
heat conservation starts with peripheral vasoconstriction
and is followed by thermogenesis (generally nonshivering in neonates) while the new set-point is achieved. It is
important to note that newborn infants of different animal
species react in peculiar ways to different known pyrogens.
Human newborns can have severe bacterial infections
without increased body temperature.
In addition to pyogenic mechanisms of febrile response
in newborns, other phenomena can lead to elevated of
body temperature. Newborn infants have poor heat dissipation mechanisms (absence of sweating); therefore
exposure to excess heat or insulation (e.g., excessive swaddling) can quickly increase their core temperature. Such
overheating commonly occurs when term babies are
nursed in uncontrolled incubators or under radiant warmers. Temperature elevation can also occur with increased
infant metabolic rate such as that seen with skeletal muscle rigidity and status epilepticus. Another cause of temperature elevation is occasionally observed in healthy,
breast-feeding newborn infants on the third to fourth
day of life, and it is believed to result from dehydration
caused by insufficient milk production. Finally, there are
more recent reports of an increased incidence of neonatal
fever in infants of mothers receiving epidural analgesia.
The mechanisms for temperature elevation in these latter
instances are unknown.
Determining the Cause of Fever
Sepsis is an uncommon cause of fever. Paradoxically, neonates with sepsis more frequently have hypothermia as
well. However, sepsis is probably the most treatable lifethreatening illness occurring in febrile newborn infants,
especially those with temperature elevations to greater
than 38° to 39° C, who are more likely to have bacteremia,
purulant menigitis, and pnemonia. Most neonatal febrile
episodes are noted in first day of life (54% in one series);
however, any fever occurring on the third day of life and
body temperature greater than 39° C have both been
correlated with a significantly higher chance of bacterial
disease. Severe temperature elevation is also associated
with viral disease, particularly herpes simplex encephalitis; therefore work-ups for sepsis in these infants should
include lumbar puncture.
Hyperthermia has been reported in tiny premature
infants as a complication of improper use of shielding
devices under either convection-warmed incubator or radiant warmer conditions. When incubated, babies should
always have a strictly monitored and controlled source of
heat. Fever secondary to overheating, particularly associated with incubators, is more common in equatorial and
tropical countries.
Dehydration is an infrequently recognized cause of fever
in the newborn period. Dehydration occurring in healthy
term infants between the third and fourth day of life was
noted previously, and it is probably the result of inadequate
milk intake. Dehydration fever is commonly seen in large
breastfed babies whose milk intake is poor and who may
be exposed to high environmental temperatures during the
summertime or in tropical areas. Body temperature can
range between 37.8° and 40° C. Rehydration leads to resolution of fever and is key to the diagnosis of dehydration fever.
In two reports (Lieberman et al, 1997; Pleasure and Stahl,
1990), fever was more common in neonates born to mothers
receiving epidural analgesia during labor when compared
with those without analgesia (7.5% versus 2.5% and 14.5%
versus 1%, respectively). One of these reports (Lieberman
et al, 1977) observed more frequent sepsis evaluations and
antibiotic use in the offspring of women receiving epidural analgesia. With the increasing use of epidural analgesia during labor, recognizing epidural neonatal fever is an
important consideration when evaluating a febrile neonate.
Unusual and uncommon causes of neonatal fever
include neonatal typhoid fever and congenital malaria,
which should be considered in immigrant populations or in
third-world countries. An increase in unexplained neonatal
fevers was associated with the introduction of routine hepatitis B vaccination versus historical controls (Lewis et al,
2001), but this was not confirmed in a subsequent, large
prospective clinical study (Lewis et al, 2001). In addition,
temperature elevations may be seen with hypothalamic or
other central nervous system malformations or masses.
Subarachnoid or other intracranial hemorrhages may also
be associated with temperature elevation. On rare occasions, neonatal spinal neurenteric cyst can manifest with
long-lasting neonatal fever and should be considered in the
differential diagnosis of acute myelopathy with persistent
fever in infancy (greater than 3 weeks’ duration). The presence of myelopathy will help to establish this diagnosis.
Management
The clinical problem is that fever may be the only indication of severe bacterial disease. The relevant perinatal
history should be evaluated for risk factors mitigating a
laboratory evaluation, presumptive treatment for infection, or both. Furthermore, signs that are suggestive
of sepsis (e.g., diminished activity, irritability, seizures)
should be considered. All neonates with fever should be
evaluated for hydration, weight loss, and foci of infection
(i.e., cellulitis, septic arthritis or osteomyelitis, omphalitis, and the presence of colonized foreign bodies, such as a
central venous line).
However, febrile neonates without clinical history or
any signs of infection present a challenge with insufficient
data in the literature regarding appropriate management.
An infant’s environment should be examined for overheating. In breastfeeding, infants with fever at 3 to 4 days of
age and excessive weight loss, dehydration fever should be
considered and treated to establish this diagnosis. Mothers
CHAPTER 30 Temperature Regulation of the Premature Neonate
receiving epidural analgesia often manifest shivering with
their temperature rise, and they experience a rapid defervescence after discontinuing the epidural infusion. Recognition of this pattern may avoid unnecessary sepsis
evaluations in neonates with early fever.
THERAPEUTIC HYPOTHERMIA
FOR BRAIN PROTECTION FOLLOWING
NEONATAL ASPHYXIA
Since 2005, therapeutic hypothermia has been considered the standard of care for a highly selected population
of near-term and term infants suffering hypoxic-ischemic
events after birth, or after medically witnessed cardiopulmonary arrest and resuscitation. Two randomized studies
suggest that cerebral cooling with either whole body cooling to a core temperature of 33.5° C (considered moderate
hypothermia) or selective head cooling to approxmately
10° C (with mild whole-body cooling to 34° C) reduces
the risk for death or moderate to severe neurologic injury
from approxiately two thirds, to less than half (Gluckman
et al, 2005; Shankaran et al, 2005). In the United States,
the National Institutes of Health Institute of Child Health
and Human Development Experts Panel Workshop held
in May 2005 emphasized using standardized protocols
adapted from these randomized trials for hypothermia
treatment, and it recommended continual follow-up until
school age to develop and better refine therapy for treating moderate to severe clinical neonatal encephalopathy
observed in term or near-term neonates (Higgins et al,
2006).
The Children’s National Medical Center (CNMC) has
adapted the Neonatal Network’s whole-body hypothermia
protocol (Shankaran et al, 2005). In addition, CNMC provides continuous electroencephalogram (EEG) neurologic
monitoring as part of a protocol, without consideration of
the EEG as a criteria for initiating cooling. A full montage video-EEG (using a modified International 10-20
system accepted for neonates) is recorded by computer for
120 hours after birth to include cooling and rewarming
recovery. EEG data are displayed on a centralized monitoring station and reviewed at least daily for every patient
treated for hypothermia. Amplitude-integrated EEG is
also reviewed for characterization of background pattern
and for detection of seizures that can contribute to evolving brain injury and are treated aggressively. Automated
seizure detection software is confirmed on raw EEG signals by an electrophysiology neonatal neurologist who is
integrated into the program. To receive cooling therapy,
infants must be at 34 weeks’ gestation or greater, arrive for
treatment within 6 hours of birth, and have experienced
a hypoxic-ischemic event. Evidence of hypoxic-ischemic
injury includes: resuscitation being required at delivery, an
umbilical vessel blood gas pH of 7.00 or less, or having a
significant base deficit of at least −16. Also qualifying for
therapy are blood gas disturbances within the first hour of
life, with a pH of 7.01 to 7.15 and a base deficit of −10 to
−15.9, along with an ominous perinatal history (e.g., fetal
heart rate decelerations, umbilical cord prolapse or rupture, uterine rupture, severe maternal trauma preceding
birth, abruption of the placenta, maternal life-threatening
event requiring cardiopulmonary resuscitation). Signs of
365
moderate to severe neonatal encephalopathy must also
be present to qualify for hypothermia intervention: lethargy, complete stupor, diminished or completely absent
spontaneous activity and/or aberrant muscle tone, weak
or absent sucking and Moro reflexes with pupils fixed and
constricted or unresponsive and dilated, fixed flexion or
extension posturing of extremities, or a clinically observed
seizure. Infants with such symptoms persisting for several
days have an approximately 60% risk for death before hospital discharge, whereas a majority of survivors experience
moderate to severe life-long neurodevelopmental disabilities (e.g., cerebral palsy, deafness, blindness, mental retardation, or recurrent seizures as in epilepsy).
Infants meeting these criteria are quickly examined at
admission and then placed supine onto a water-filled cooling blanket that is precooled to 5° C (41° F). CNMC uses
a Blanketrol II device (Cincinnati Sub-Zero, Cincinnati,
Ohio) that is used commonly in the emergency department to reduce high fevers and in the operating room
to promote hypothermia before and during reconstructive heart surgery. An esophageal temperature probe is
placed into the distal third of the esophagus to monitor
the infant’s core temperature, and the thermostatic controller in the water mattress’ cooling unit is set to 33.5° C.
A second, larger pediatric-size blanket is also attached in
parallel to the cooling system and is suspended at the bedside as a “sail” heat capacitor. Water circulates through
both blankets exposed to both the baby and the room air
temperature to diminish continuously monitored temperature fluctuations in esophageal temperature (less than
±0.5° C). Although CNMC uses a warmer bed platform as
a crib, the overhead warmer is not turned on during the
cooling period. Abdominal wall skin temperature is monitored with a surface probe that is available with the warmer
bed (in monitor-only mode). Temperatures of the esophagus, skin, and axilla are thus monitored and recorded every
15 minutes for the first 4 hours of cooling, every hour for
the next 8 hours, and every 4 hours during the remaining 72-hour period of hypothermia. CNMC’s electronic
medical record (Cerner) provides hourly esophageal
temperature recording. After 72 hours, the set-point of
the controller on the cooling system is increased by 0.5°
C increments per hour to promote gradual rewarming.
The Neonatal Network reported febrile rebound and
seizures during rewarming (Shankaran et al, 2005); however, CNMC has not experienced this with continuous
EEG monitoring over 24 to 48 hours throughout rewarming and a subsequent recovery period. After 6 hours, the
esophageal probe and cooling blankets are removed, and
anterior abdominal wall skin temperature is then regulated using the radiant warmer’s servomechanism set at
36° to 36.5° C (i.e., warmer is turned on). The purpose
of rewarming slowly is to avoid rapid shifts in electrolytes
(calcium and potassium in particular), cardiac arrhythmias,
and in rewarming hyperthermia, because fever promotes
further brain injury. Infants otherwise receive routine neonatal intensive care, with continuous monitoring of vital
signs. Mild sinus bradycardia (80 to 90 beats/min) and
small decreases in mean arterial blood pressure (less than
20 mm Hg) are commonly observed and treated easily with
volume and pressor infusions, according to CNMC’s standard institutional guidelines for babies developmentally.
366
PART VIII Care of the High-Risk Infant
Frequent blood samples are monitored for glucose regulation (infants are restricted to 4 to 6 mg/kg/min of dextrose
infusion), coagulopathy (in particular treating fibrinogen
levels less than 150 mg/dL and platelet levels less than
80,000 per millimeter), and major organ failure (e.g., electrolytes, blood urea nitrogen, creatinine, liver enzymes).
CNMC’s clinical experience over the past 3 years in
more than 98 babies meeting strict criteria for therapeutic
hypothermia has been commensurate with that reported
from the Neonatal Network (Shankaran et al, 2005). Target esophageal temperature was achieved at 33.5 ± 0.5° C
(range 33° to 34° C) in 30 to 60 minutes without a major
circulatory mishap at CNMC. Continuous EEG monitoring has been instructive for intervening seizure activity
(observed on EEG in approximately one quarter of infants
shortly after admission). CNMC electrophysiologists
observed general improvements in background voltages
and patterns on EEG recordings during and after hypothermia. Specifically, improvement of background activity, appearance of sleep wake cycling, and disappearance
of seizures has been observed at the time of rewarming
(El-Dib et al, 2007). The Neonatal Network observed seizures emerging frequently during rewarming, hence the
precaution in re-warming more slowly. CNMC is presently acquiring and reviewing infant neurologic and neuron-developmental follow-up data on a 6-month schedule
through the first 2 years of life. CNMC has also made the
clinical observation that the water mattress felt warm to
the touch (i.e., warmer than the examiner’s hand) during
most of the cooling period (Baumgart et al, 2007). Median
ambient temperature in CNMC’s NICU during October
2007 (23.1° C; range, 20.9° to 25.4° C) was usually less
than both the blanket water and baby temperatures. No
infant had acidemia during cooling. Temperature gradients suggest that whole-body cooling is achieved through
surface cooling from skin exposed to the ambient environment, and not actually by heat loss into the water-blanket.
CNMC observers believe that the water matress simply
incubates infants at a lower temperature. Except during the first 30 minutes, the blanket more often provided
warmth to maintain esophageal temperature at 33.5° C.
Whole-body cooling might also be provided by regulating an incubator air temperature or a radiant heater to
maintain esophageal temperature. It has been important to
CNMC’s outreach education program to emphasize that
referring (i.e., resuscitating) institutions should perform
cardio pulmonary resuscitation under warm conditions to
facilitate cardiac response according to Neonatal Resuscitation Program guidelines. Cold resuscitation is not advocated until after careful clinical evaluation of a stabilized
patient has been performed at CNMC to meet therapeutic
cooling criteria.
SUMMARY
The premature newborn with very low birthweight is
extremely vulnerable to harsh fluctuations in physical
environment. These infants require frequent assessments
of skin, core, and air temperature and relative humidity to
design an optimal strategy for thermal regulation. In caring for smaller babies, heat replacement is often required,
and refinement of techniques to accomplish replacement
without inducing hyperthermia is needed. The use of
therapeutic hypothermia in highly selective cases at risk
for brain injury is no longer considered novel.
SUGGESTED READINGS
Baumgart S: Partitioning of heat losses and gains in premature newborn infants
under radiant warmers, Pediatrics 75:89-99, 1985.
Bell EF, Rios GR: Air versus skin temperature servo control of infant incubators,
J Pediatr 103:954, 1983.
Bell EF, Rios GR: A double‑walled incubator alters the partition of body heat loss
of premature infants, Pediatr Res 17:135-140, 1983.
Bell EF, Weinstein MR, Oh W: Heat balance in premature infants: Comparative
effects of convectively heated incubator and radiant warmer, with and without
plastic heat shield, J Pediatr 96:460-465, 1980.
Bruck K: Heat production and temperature regulation. In Stave U, editor: Perinatal
physiology, New York, 1978, Plenum Publishing, pp 455-498.
Dawkins MJ, Hull D: The production of heat by fat, Sci Am 213:62-67, 1965.
Day RL, Caliguiri L, Kaminski C, et al: Body temperature and survival of premature infants, Pediatrics 34:171-181, 1964.
Knauth A, Gordin M, McNelis W, et al: Semipermeable polyurethane membrane
as an artificial skin for the premature neonate, Pediatrics 83:945-950, 1988.
Marks KH, Lee CA, Bolan CD, et al: Oxygen consumption and temperature control of premature infants in a double‑wall incubator, Pediatrics 68:93-98, 1981.
Mayfield SR, Bhatia J, Nakamura KT, et al: Temperature measurement in term and
preterm neonates, J Pediatr 104:271-275, 1984.
Okken A, Blijham C, Franz W, et al: Effects of forced convection of heated air on
insensible water loss and heat loss in preterm infants in incubators, J Pediatr
101:108-112, 1982.
Scopes JW: Thermoregulation in the newborn. In Avery CB, editor: Neonatology,
pathophysiology and management of the newborn, ed 2, Philadelphia, 1981, JB
Lippincott, pp 171-181.
Yager JY, Armstrong EA, Jaharus C, et al: Preventing hyperthermia decreases brain
damage following neonatal hypoxic-ischemic seizures, Brain Res 1011:48-57,
2004.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
31
Acid-Base, Fluid, and Electrolyte
Management
Michael A. Posencheg and Jacquelyn R. Evans
FLUID AND ELECTROLYTE BALANCE
Maintenance of fluid and electrolyte balance is essential
for normal cell and organ function during intrauterine
development and throughout extrauterine life. Pathologic conditions in the newborn often lead to disruption
of the complex regulatory mechanisms of fluid and electrolyte homeostasis. Therefore a thorough understanding
of the physiologic changes in neonatal fluid and electrolyte homeostasis and the provision of appropriate fluid and
electrolyte therapy based on the principles of developmental fluid and electrolyte physiology are among the cornerstones of modern neonatal intensive care.
DEVELOPMENTAL CHANGES AFFECTING
FLUID AND ELECTROLYTE BALANCE
IN THE FETUS AND NEONATE
Developmental Changes in Body
Composition and Fluid Compartments
Dynamic changes occur in body composition and fluid distribution during intrauterine life, labor and delivery, and
the early postnatal period. Thereafter the rate of change
in body composition and fluid distribution gradually
decreases, with more subtle changes taking place especially
after the first year of life (Friis-Hansen, 1961).
Changes during Intrauterine Development
In early gestation, body composition is characterized by
a high proportion of total body water (TBW) and a large
extracellular compartment (Brans, 1986; Friis-Hansen,
1983). As gestation advances, rapid cellular growth, accretion of body solids, and fat deposition result in gradual
reductions in TBW content and extracellular fluid volume while the intracellular fluid compartment increases
(Figure 31-1) (Friis-Hansen, 1983). In the 16-week-old
fetus, TBW represents approximately 94% of total body
weight, and approximately two thirds of the TBW is distributed in the extracellular and one third in the intracellular compartment. By term, TBW contributes only 75%
of body weight, and almost half of this volume is located
in the intracellular compartment. Therefore infants born
prematurely have TBW excess and extracellular volume
expansion compared with their term counterparts, with
the majority of the expanded extracellular volume being
distributed in the interstitium (Brace, 1992).
Changes during Labor and Delivery
Additional and more acute changes in TBW and its distribution occur during labor and delivery. Arterial blood
pressure rises several days before delivery in response
to increases in catecholamine, vasopressin, and cortisol
plasma concentrations and translocation of blood from the
placenta into the fetus. This rise in arterial blood pressure,
along with changes in the fetal hormonal milieu and an
intrapartum hypoxia-induced increase in capillary permeability, results in a shift of fluid from the intravascular to
the interstitial compartment. This fluid shift results in an
approximately 25% reduction in circulating plasma volume in the human fetus during labor and delivery (Brace,
1992). The postnatal increase in oxygenation and changes
in vasoactive hormone production then restore capillary
membrane integrity and favor absorption of interstitial
fluid into the intravascular compartment. The ensuing
gradual movement of fluid from the expanded interstitial
space into the bloodstream aids in maintaining intravascular volume during the first 24 to 48 hours postnatally,
when oral fluid intake may be limited. However, prematurity, pathologic conditions, or both can disrupt this delicate process and interfere with the physiologic contraction
of the extracellular fluid (ECF) compartment in the immediate postnatal period.
In the fetus, body composition and fluid balance depend
on the electrolyte and water exchange between mother,
fetus, and amniotic space (Brace, 1986). Antenatal events can
have significant effects on postnatal fluid balance. Maternal indomethacin treatment or excessive administration of
intravenous fluids during labor can result in neonatal hyponatremia with expanded extracellular water content (Rojas
et al, 1984; vd Heijden et al, 1988). Placental insufficiency
or maternal diuretic therapy can impair fetal hydration,
leading to decreases in extracellular volume, urine output,
and amniotic fluid volume (Van Otterlo et al, 1977). The
timing of cord clamping after delivery is another important
factor significantly affecting total circulating blood volume
and extracellular volume in the neonate. Immediate cord
clamping does not allow for placental transfusion, but if the
cord clamping is delayed only 3 to 4 minutes after delivery
with the newborn positioned at or below the level of the
placenta, up to 25 to 50 mL/kg of blood is transfused into
the neonate, representing an approximately 25% to 50%
increase in the total blood volume (Linderkamp, 1982; Yao
and Lind, 1974). The onset of infant respiratory effort after
birth also increases placental transfusion regardless of the
infant’s position (Philip and Teng, 1977).
Changes in the Postnatal Period
In the first few days and weeks after birth, the most important effects on the pace of further changes in body composition, TBW content, and its distribution are exerted by
gestational and postnatal ages, the presence or absence of
pathologic conditions, the immediate environment, and
the type of nutrition. In normal conditions in the first few
367
368
PART VIII Care of the High-Risk Infant
FIGURE 31-1 Total body water (TBW) content and its distribution between the extracellular fluid (ECF) and intracellular fluid (ICF)
compartments in the human fetus, newborn, and infant from conception
until 9 months of age. (Data represent average values from Friis-Hansen
B: Body water compartments in children: changes during growth and related
changes in body composition, Pediatrics 28:169-181, 1961.)
days after birth, the postnatal increase in capillary membrane integrity favors absorption of the interstitial fluid
into the intravascular compartment. The ensuing rise in
circulating blood volume stimulates the release of atrial
natriuretic peptide from the heart, which in turn enhances
renal sodium and water excretion (Sagnella and MacGregor, 1984) resulting in an abrupt decrease in TBW and
attendant weight loss. Although it is generally accepted
that this postnatal weight loss is primarily due to the contraction of the expanded ECF compartment (Cheek et al,
1961), some water loss from the intracellular compartment
can also occur, particularly in infants with extremely low
birthweight (ELBW) and increased transepidermal water
losses (Costarino and Baumgart, 1991; Sedin, 1995).
Healthy term newborns lose an average of 5% to 10% of
their birthweight during the first 4 to 7 days of life (Brace,
1992); thereafter they establish a pattern of steady weight
gain. Because preterm infants have an increased TBW content and extracellular volume, they lose an average of 15%
of their birthweight during transition (Shaffer and Meade,
1989) and, depending on the degree of prematurity and
associated pathologic conditions, these neonates only regain
their birthweight by 10 to 20 days after birth (Figure 31-2).
Physiology of the Regulation of Body
Composition and Fluid Compartments
Although human cells have the ability to adjust their intracellular composition, ultimate regulation of the intracellular volume and osmolality relies on the control of the
extracellular compartment. Therefore the human body
must be able to monitor the volume and osmolality of
the extracellular compartment and to correct the changes
resulting from its interaction with the environment.
Regulation of the Intracellular Solute
and Water Compartment
The major intracellular solutes are the cellular proteins
necessary for cell function, the organic phosphates associated with cellular energy production and storage, and
FIGURE 31-2 Postnatal changes in body weight (expressed in percent
of birthweight), extracellular fluid volume (estimated by the bromide
dilution method), and sodium balance (defined as the difference
between sodium intake and urinary sodium excretion). (From Shaffer SG,
Weismann DN: Fluid requirements in the preterm infant, Clin Perinatol
19:233-250, 1992.)
the equivalent cations balancing the phosphate and protein anions (MacKnight and Leaf, 1977). Potassium is the
major intracellular cation, and sodium is the major extracellular cation. The energy derived from the concentration
differences for sodium and potassium between the intracellular and extracellular compartments is used for cellular
work. Because changes in osmolality of the extracellular
compartment are reflected as net movements of water in or
out of the cell, regulation of ECF concentration ultimately
controls the osmolality and size of the intracellular compartment (MacKnight and Leaf, 1977). This physiologic
principle must be kept in mind by the neonatologist managing sick term and preterm neonates with disturbances
of sodium homeostasis. Rapid changes in serum sodium
concentration and thus in extracellular osmolality directly
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
369
affect the osmolality and size of the intracellular compartment and can lead to irreversible cell damage, especially in
the central nervous system.
Regulation of the Intracellular-Extracellular
Interface: The Interstitial Compartment
In the healthy term neonate, hydrostatic and oncotic pressures are well balanced, with both being approximately half
those in the adult (Sola and Gregory, 1981). In normal physiologic conditions, movement of fluid across the capillary
is determined by the direction of the net driving pressure
([PC − PT] − [πP − πT]) and the water and protein permeability characteristics of the capillary wall (Figure 31-3).
At the arterial end of the capillary, intracapillary hydrostatic pressure (PC) is high, and plasma oncotic pressure
(πP) is relatively low, resulting in a net movement of fluid
out of the capillary. As filtration of relatively protein-poor
fluid continues along the capillary, plasma oncotic pressure rises and intracapillary hydrostatic pressure drops;
therefore on the venous side, fluid moves from the interstitium into the capillary, so that much of the filtered fluid
is reabsorbed at the end of the capillary bed. The fluid
remaining in the interstitium (arterial-venous side of the
capillary) is drained by the lymphatic system. Interstitial
hydrostatic (PT) and oncotic (πT) pressures remain virtually
unchanged along the capillary bed. However, pathologic
conditions readily disturb the delicate balance between the
hydrostatic and oncotic forces, leading to an expansion of
the interstitial compartment at the expense of the intravascular compartment. The increased interstitial fluid volume
(edema) then further affects tissue perfusion by altering
the normal function of the extracellular-intracellular interface. Box 31-1 summarizes the mechanisms for conditions
resulting in interstitial edema formation in the neonate.
There are also some important developmentally regulated
differences between the newborn and adult relating to the
pathogenesis of edema formation. Capillary permeability
to proteins is increased during the early stages of development (Brace, 1992; Gold and Brace, 1988). Because
neonatal capillary permeability is further increased under
pathologic conditions (see Box 31-1), protein concentration in the interstitial compartment may approach that of
the intravascular space, favoring further intravascular volume depletion and interstitial volume expansion. When
sick neonates are treated with frequent albumin boluses,
much of the infused albumin leaks into the interstitium,
creating a vicious cycle of intravascular volume depletion
and edema formation. If the cycle is not interrupted, it can
result in the formation of anasarca, which has an extremely
poor prognosis. In summary, the sick neonate has a limited
capacity to maintain appropriate intravascular volume and
to regulate the volume and composition of the interstitium.
The ensuing intravascular hypovolemia and edema formation result in vasoconstriction and disturbances in tissue
perfusion and cellular function with further impairments
in the regulation of extracellular volume distribution.
Regulation of the Extracellular Solute
and Water Compartment
The regulation of the volume and osmolality of the extracellular compartment ensures the integrity of the circulation and maintains the osmolality of the extracellular
FIGURE 31-3 Filtration and reabsorption of fluid along the capillary
under physiologic conditions.
BOX 31-1 M
echanisms for Conditions
Causing Interstitial Edema
in the Neonate
CONDTIONS FAVORING FLUID ACCUMULATION IN THE
INTERSTITIAL SPACE BY CAUSING A DYSEQUILIBRIUM
BETWEEN FILTRATION AND REABSORPTION OF FLUID
BY THE CAPILLARIES
Increased hydrostatic pressure
Elevated capillary hydrostatic pressure
Increased cardiac output
Venous obstruction
Decreased tissue hydrostatic pressure
Conditions associated with changes in the properties of the interstitial gel
(edematous states, effects of hormones including prolactin)
Decreased oncotic pressure gradient
Decreased capillary oncotic pressure
Prematurity, hyaline membrane disease
Malnutrition, liver dysfunction
Nephrotic syndrome
Increased interstitial oncotic pressure is usually the result of increased capillary
permeability
Elevation of the filtration coefficient
Increased capillary permeability
Organs with large-pore capillary endothelium (liver, spleen)
State of maturity (preterm infants > term newborns > adults)
Production of pro-inflammatory cytokines (sepsis, anaphylaxis, hypoxic
tissue injury, tissue ischemia, ischemia-reperfusion, soft tissue
trauma, extracorporeal membrane oxygenation)
Increased capillary surface area
Vasodilation
CONDITIONS ASSOCIATED WITH DECREASED LYMPHATIC
DRAINAGE
Decreased muscle movement
Neuromuscular blockade and/or heavy sedation
Central and/or peripheral nervous system disease
Obstruction of lymphatic flow
Increased central venous pressure
Scar tissue formation (bronchopulmonary dysplasia)
Mechanical obstruction (dressings, high mean airway pressure in mechanically ventilated newborns)
Modified from Costarino AT, Baumgard S: Neonatal water metabolism. In Cowett RM, editor:
Principles of perinatal/neonatal metabolism, New York, 1991, Springer Verlag, pp 623-649.
370
PART VIII Care of the High-Risk Infant
compartment within 2% of the osmolar set point between
275 and 290 mOsm (Robertson and Berl, 1986). Blood
pressure and serum sodium concentration (i.e., osmolality)
are monitored by baroreceptors and osmoreceptors, respectively. The effector limb of the regulatory system consists
of the heart, vascular bed, kidneys, and intake of fluid in
response to thirst. The latter part of the effector system is
inactive in critically ill term and preterm neonates whose
fluid intake is completely controlled by caregivers. By regulating the function of the effector organs, several hormones
have a role in the control of the extracellular compartment,
including the renin-angiotensin-aldosterone system, vasopressin, atrial natriuretic peptide, brain (B-type) natriuretic
peptide, bradykinin, prostaglandins, and catecholamines.
Because volume changes in the extracellular compartment
must be reflected by similar changes in the intravascular
volume for effective operation of the regulatory mechanisms, regulation of the extracellular compartment relies
on intact cardiovascular function and on the integrity of
the capillary endothelium (Robertson and Berl, 1986). For
example, under physiologic conditions, an increase in the
extracellular volume is reflected by an increase in the circulating plasma volume, leading to rises in blood pressure
and renal blood flow. The ensuing increase in glomerular
filtration and urine output returns the extracellular volume
to normal. In critically ill neonates, however, the capillary
leak and reduced myocardial responsiveness resulting from
immaturity and underlying pathologic conditions limit the
increase in the circulating blood volume when extracellular
volume expands. Thus, especially in sick preterm infants,
blood pressure may rise only transiently, and renal blood
flow may remain low after volume boluses as fluid rapidly
leaks into the interstitium. Inappropriate central regulation
of vascular tone results in vasodilatation, further decreasing effective circulating blood volume and compromising
tissue perfusion; this leads to impaired gas exchange in the
lungs, resulting in hypoxia with further increases in capillary leak. Unless interrupted by appropriate therapeutic
measures, a vicious cycle with further deterioration readily
occurs in the sick neonate.
Maturation of Organs Regulating Body
Composition and Fluid Compartments
The heart, kidneys, skin, and endocrine system play the most
important roles in the regulation of extracellular (and thus
intracellular) fluid and electrolyte balance in the neonate.
Immaturity of these organ systems, especially in infants with
very low birthweight (VLBW), results in a compromised
regulatory capacity, which must be noted when estimating
daily fluid and electrolyte requirements in these patients.
Maturation of the Cardiovascular System
There is a direct relationship between gestational maturity
and the ability of the neonatal heart to respond to acute
volume loading (Baylen et al, 1986). The blunted Starling response of the immature myocardium results from
its lower content of contractile elements and incomplete
sympathetic innervations (Mahony, 1995). Because central
vasoregulation and endothelial integrity are also developmentally regulated (Brace, 1992; Gold and Brace, 1988),
an appropriate effective intravascular volume is seldom
maintained in a critically ill preterm infant. Since regulation of the extracellular volume requires the maintenance
of an adequate effective circulating blood volume, the
immaturity of the cardiovascular system contributes to the
limited capacity of sick preterm infants to effectively regulate the total volume of their extracellular compartment.
Maturation of Renal Function
The kidney has a crucial role in the physiologic control of
fluid and electrolyte balance. It regulates extracellular volume and osmolality through the selective reabsorption of
sodium and water, respectively. Immaturity of renal function renders preterm infants susceptible to both excessive
sodium and bicarbonate losses (El-Dahr and Chevalier,
1990). In addition, the inability of the preterm infant to
respond promptly to a sodium or volume load results in
a tendency toward extracellular volume expansion with
edema formation. Because prenatal steroid administration
accelerates maturation of renal function (van den Anker et
al, 1994), preterm infants treated with steroids in utero have
a better capacity to regulate their postnatal ECF contractions. During the first few weeks of life, hemodynamically
stable but extremely immature infants produce dilute urine
and may develop polyuria because of their renal tubular
immaturity. As tubular functions mature, their concentrating capacity gradually improves from the 2nd to 4th week
of life. However, it takes years for the developing kidney to
reach the concentrating capacity of the adult kidney.
Maturation of the Skin
Although term infants have a well-developed cornified
layer of the epidermis, extremely immature neonates have
only two or three cell layers in the epidermis (CartridgePatrick and Rutter, 1992). Because of the lack of an effective barrier to diffusion of water through the immature
skin, transepidermal free water losses in the immature
infant may be extremely high during the first few days
postnatally. Gestational age, postnatal age, the pattern of
intrauterine growth, and environmental factors have crucial roles in the magnitude of transepidermal free water
losses (Figure 31-4). Although skin cornification rapidly
increases, even in the extremely immature infant during
the first few days after birth, full maturation of the epidermis does not occur for more than 28 days of age (Sedin,
1995). Chronic intrauterine stress (Hammarlund et al,
1983) and prenatal steroid treatment (Aszterbaum et al,
1993) also enhance maturation of the skin.
Increases in free water losses through the immature skin
of the VLBW infant can result in early postnatal hypertonic dehydration with rapid changes in intracellular volume and osmolality. In many organs, especially the brain,
these abrupt changes in intracellular volume and osmolality
can lead to cellular dysfunction and ultimately cell death.
Maturation of End-Organ Responsiveness
to Hormones Involved in the Regulation
of Fluid and Electrolyte Balance
Several hormones, including but not limited to the reninangiotensin-aldosterone system, vasopressin, atrial natriuretic peptide, and brain (B-type) natriuretic peptide,
directly regulate the volume or composition of the extracellular compartment. These hormones exert their effects
Transepidermal Water Loss
(g/m2/h)
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
60
50
40
3
30
5
20
14
10
0
28
21
7
s)
ay
(d
lA
a
at
n
st
Po
ge
0
1
26 28 30 32 34 36 38 40
Gestational Age (weeks)
FIGURE 31-4 Transepidermal water loss in relation to gestational
age during the first 28 postnatal days in infants who are appropriate for gestational age. There is an exponential relationship between
transepidermal water loss and gestational age, the water loss being
higher in preterm infants than in term infants. Transepidermal water
loss is also significantly affected by postnatal age, especially in the immature preterm infant. The measurements were performed at an ambient
air humidity of 50% and with the infants calm and quiet. (From Hammarlund K, Sedin G, Stromberg B: Transepidermal water loss in newborn
infants. VIII: Relation to gestational age and postnatal age in appropriate and
small for gestational age infants, Acta Paediatr Scand 72:721-728, 1983.)
mainly by altering renal sodium and water excretion and
by inducing changes in systemic vascular resistance and
myocardial contractility. Other hormones, including the
prostaglandins, bradykinin, and prolactin, modulate the
actions of many of the regulatory hormones.
Renin-Angiotensin-Aldosterone System
Decreases in renal capillary blood flow stimulate renin
secretion, which in turn initiates the production of angiotensin. Angiotensin induces vasoconstriction, increased
tubular sodium and water reabsorption, and the release of
aldosterone (Riordan, 1995). Aldosterone increases potassium secretion and further enhances sodium reabsorption in the distal tubule; therefore the primary function
of this system is to protect the volume of the extracellular compartment and maintain adequate tissue perfusion
(Bailie, 1992). However, its effectiveness in the neonate
is somewhat limited by the decreased responsiveness of
the immature kidney to the sodium- and water-retaining
effects of these hormones (Sulyok et al, 1985). Vasodilatory and natriuretic prostaglandins generated in the
kidney (Gleason, 1987) are the main counterregulatory
hormones balancing the renal actions of the renin-angiotensin-aldosterone system. Therefore, when prostaglandin
production is inhibited by indomethacin, the unopposed
vasoconstrictive and sodium-retentive actions of the activated renin-angiotensin-aldosterone system contribute to
the development of the drug-induced renal failure in the
preterm infant (Gleason, 1987; Seri, 1995; Seri et al, 2002).
Vasopressin (Antidiuretic Hormone)
Vasopressin has its major effect in maintaining the osmolality of the extracellular compartment. Vasopressin selectively raises free water reabsorption in the kidneys and
371
results in blood pressure elevation (Elliot et al, 1996).
Plasma levels of vasopressin are markedly elevated in the
neonate, especially after vaginal delivery, and its cardiovascular actions facilitate neonatal adaptation (Pohjavuori and
Raivio, 1985). The high vasopressin levels are in part also
responsible for the diminished urine output of the healthy
term neonate during the first day of life. Under certain
pathologic conditions, the dysregulated release of, or the
end-organ unresponsiveness to, vasopressin significantly
affects renal and cardiovascular functions and electrolyte
and fluid status in the sick preterm and term infant. In
the syndrome of inappropriate secretion of antidiuretic
hormone (SIADH), an uncontrolled release of vasopressin occurs in sick preterm and term infants, with resulting water retention, hyponatremia, and oliguria. In the
syndrome of diabetes insipidus (DI), the lack of pituitary
production of vasopressin or renal unresponsiveness to
vasopressin results in polyuria, with increased thirst and
hypernatremia.
Atrial Natriuretic Peptide
Via its direct vasodilatory and renal natriuretic actions,
atrial natriuretic peptide (ANP) regulates the volume of
the extracellular compartment in the fetus and neonate
in a fashion opposite to that of the renin-angiotensin-
aldosterone system (Iwamoto, 1992; Needleman and
Greenwald, 1986; Seymour, 1985). ANP has a direct
inhibitory effect on renin production and aldosterone
release (Christensen, 1993).
The stretch of the atrial wall caused by an increase in
the circulating blood volume is the most potent stimulus
for the release of this hormone. Plasma levels of this hormone are high in the fetus (Claycomb, 1988). There are a
few specific conditions in which the actions of atrial natriuretic peptide are directly relevant for the neonatologist.
For example, the hormone is involved in the regulation
of both the fluid shifts during labor (Brace, 1992) and the
extracellular volume contraction during postnatal transition (Kojima et al, 1987; Ronconi et al, 1995; Rozycki and
Baumgart, 1991; Tulassay et al, 1987). Furthermore, the
oliguric effects of positive end-expiratory pressure ventilation are due in part to a decrease in atrial natriuretic
peptide secretion (Christensen, 1993) along with the
enhanced release of vasopressin (El-Dahr and Chevalier,
1990).
Brain (or B-type) Natriuretic Peptide
Similar to ANP, brain natriuretic peptide (BNP) is a hormone secreted by cardiac myocytes. Specifically, BNP is
thought to be secreted by cardiac ventricular myocytes in
response to increases in wall tension. Both of these peptides cause natriuresis, diuresis, and vasodilation while
inhibiting the renin-angiotensin-aldosterone system
(Gemelli et al, 1991; Holmes et al, 1993; Kojima et al,
1989). BNP levels increase rapidly after birth, with levels on the first day of life up to 20-fold higher than those
at birth (Mir et al, 2003; Yoshibayashi et al, 1995), and
correlate with the downward trend in pulmonary arterial
pressure in the days after birth, unlike ANP (Ikemoto et
al, 1996). BNP levels continue to fall during the first week
of life (Koch and Singer, 2003; Mir et al, 2003). Infants
and children with congestive heart failure have an elevated
372
PART VIII Care of the High-Risk Infant
BNP level, which is associated with decreased ejection
fraction and increased heart failure score (Mir et al, 2002).
In this way, BNP can be used as a surrogate for changes in
ventricular volume from either pulmonary hypertension
or ventricular overload. Clinical researchers have begun
to investigate the utility of this peptide in diagnosing and
managing cardiac disorders in neonates. The best-studied
association to date is the relationship between BNP and
a patent ductus arteriosus (PDA). Multiple studies have
described a positive correlation between BNP level and
a hemodynamically significant PDA (Choi et al, 2005;
Czernik et al, 2008; Flynn et al, 2005; Sanjeev et al, 2005).
BNP levels were associated with larger ductal size and
degree of shunting, especially in infants older than 2 days
(Flynn et al, 2005). Furthermore, significant correlations
have been seen between BNP level and left atrial to aortic
root diameter (Choi et al, 2005; Czernik et al, 2008) and
diastolic flow velocity in the left pulmonary artery (Choi
et al, 2005). BNP levels decline significantly with either
medical or surgical therapy of a hemodynamically significant PDA (Choi et al, 2005; Sanjeev et al, 2005). The
biologically inactive fragment N-terminal pro-B-type
natriuretic peptide (NT-proBNP) has also been studied
regarding its association with PDA with similar results
(Farombi et al, 2008). NT-proBNP has a longer halflife (60 versus 20 minutes) than BNP and has also been
associated with sepsis (Farombi et al, 2008). BNP has also
been associated with other forms of cardiac dysfunction
in neonates. Increases in BNP levels have been described
in infants with persistent pulmonary hypertension of the
newborn over the first 4 days of life, correlating with the
degree of tricuspid regurgitation on an echocardiogram
(Reynolds et al, 2004). BNP measurements can also be
useful in differentiating infants in respiratory distress who
have a cardiac etiology for their disease from those who
do not (Ko et al, 2008; Reynolds et al, 2004). The usefulness of this test is limited by the variability of levels in the
first few days of life as well as the variety of assays available to measure BNP levels. Its utility may ultimately lie
in repeated measures in the same patient over time using
the same assay and following trends.
Prostaglandins
Prostaglandins have a well-documented, counterregulatory role for the renal vascular and tubular effects of
renin-angiotensin-aldosterone and vasopressin (Bonvalet
et al, 1987). The inhibition of these actions of prostaglandins by indomethacin results in clinically important and
sometimes detrimental renal vascular and tubular effects
in the preterm infant. The actions of prostaglandins modulating the effects of the other regulatory hormones of
neonatal fluid and electrolyte homeostasis are less well
studied.
Prolactin
Prolactin plays a permissive role in the regulation of fetal
and neonatal water homeostasis (Coulter, 1983; Pullano
et al, 1989). High fetal plasma prolactin levels contribute
to the increased tissue water content of the fetus. Interestingly, postnatal prolactin levels remain high in the preterm
neonate until approximately the 40th postconceptional
week (Perlman et al, 1978).
MANAGEMENT OF FLUID AND ELECTROLYTE
HOMEOSTASIS
General Principles of Fluid and Electrolyte
Management
Fluid and electrolyte management is the cornerstone of
neonatal intensive care, and appropriate management
requires an understanding of the previously outlined physiologic principles and careful monitoring of key clinical
data. Requirements vary substantially from infant to infant
and in the same infant over time; therefore intakes must
be individualized and frequently reassessed. The primary
goals are to maintain the appropriate ECF volume, ECF
and intracellular fluid osmolality, and ionic concentrations.
Assessment of Fluid and Electrolyte Status
Maternal conditions during pregnancy, drugs and fluids
administered to the mother during labor and delivery, and
specific fetal and neonatal conditions all affect early fluid
and electrolyte balance. Excessive administration of free
water or oxytocin use in the mother can result in hyponatremia in the neonate. Maternal therapy with indomethacin,
angiotensin-converting enzyme inhibitors, furosemide,
and aminoglycosides can all adversely affect neonatal renal
function. A newborn’s history of oligohydramnios or birth
asphyxia may also alert the clinician to the possibility of
abnormal renal function. In young infants, altered skin turgor, sunken anterior fontanel, and dry mucous membrane
are not sensitive indicators of dehydration, but tachycardia, hypotension, and metabolic acidosis may be seen when
intravascular volume is moderately to severely affected. In
addition, edema usually occurs early when there is volume
overload. Serial measurements of body weight, intake and
output, and serum electrolytes will usually provide the
most precise and accurate information regarding overall fluid status. Appropriate fluid balance in the first few
days after birth is associated with a urine output of 1 to 3
mL/kg per hour and a weight loss of 5% to 10% in term
infants and 10% to 15% in preterm infants. In critically ill
infants and in situations of altered homeostasis, additional
clinical data that may help in diagnosis and management
include blood urea nitrogen, serum and urine osmolarity or
specific gravity, urine electrolytes and serum bicarbonate,
along with close monitoring of blood pressure and heart
rate. The frequency of monitoring depends on the extent
of immaturity and the severity of the fluid and electrolyte
disturbance and of the underlying pathologic condition.
Water Homeostasis and Management
Water Losses
Free water losses occurring through the skin and the
respiratory tract are considered insensible losses, whereas
the sensible water losses are composed of the amounts
lost through urine and feces. Urine output is the most
important source of sensible water loss. Extremely preterm infants without systemic hypotension or renal failure usually lose 30 to 40 mL/kg/day of water in the urine
on the first postnatal day and approximately 120 mL/
kg/day by the third day. In stable, more mature preterm
infants born after the 28 weeks’ gestation, urinary water
373
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
loss is approximately 90 mL/kg/day on the first postnatal
day and 150 mL/kg/day by the third day (Coulthard and
Hey, 1985). Because of their renal immaturity, preterm
neonates have a tendency to produce dilute urine, thereby
increasing their obligatory free water losses.
Normal water losses in the stool are less significant,
amounting to approximately 10 mL/kg/day in term infants
and 7 mL/kg/day in preterm infants during the first postnatal week (Sedin, 1995). Water losses in the stool increase
thereafter and are influenced by the type of feeding and
the frequency of stooling.
Gestational age, postnatal age, and environmental
factors determine the amount of daily insensible water
losses through the skin (see Figure 31-4). During the
first few postnatal days, transepidermal water losses may
be 15-fold higher in extremely premature infants born at
23 to 26 weeks’ gestation than in term neonates (Sedin,
1995). Although the skin matures rapidly after birth, even
in extremely immature infants, insensible losses are still
somewhat higher at the end of the first month than in the
term counterparts. Prenatal steroid exposure is associated
with substantially less insensible water loss (IWL) in premature infants (Aszterbaum et al, 1993; Omar et al, 1999;
Sedin, 1995).
Among environmental factors, ambient humidity has the
greatest effect on transepidermal water loss. In extremely
immature neonates, a rise in the ambient humidity of the
incubator from 20% to 80% decreases the transepidermal
water loss by approximately 75% (Sedin, 1995). However,
the use of an open radiant warmer more than doubles transepidermal water losses (Flenady and Woodgate, 2003).
Applying a plastic heat shield while the infant is under the
warmer can decrease transepidermal water loss by 30%
to 50% (Costarino et al, 1992). At low ambient humidity, phototherapy increases transepidermal water losses by
approximately 30%. However, one study showed that if the
ambient humidity is at least 50% in the incubator, regular
phototherapy lamps will not increase the transepidermal
water loss in infants older than 28 weeks’ gestation (Sedin,
1995). On the other hand, halogen spotlight phototherapy
increases transepidermal water loss in premature infants
by 20% despite constant skin temperature and relative
humidity (Grunhagen et al, 2002). Newer light-emitting
diode phototherapy devices are not associated with significant transepidermal water loss or changes in cerebral
hemodynamics (Bertini et al, 2008). Other factors that
increase IWL include activity, airflow, elevated body, and
environmental temperature as well as skin breakdown and
skin or mucosal defects (e.g., gastroschisis, epidermolysis
bullosa).
Insensible water losses from the respiratory tract depend
mainly on the temperature and humidity of the inspired gas
mixture and on the respiratory rate, tidal volume, and dead
space ventilation. In a healthy term newborn, the water
loss through the respiratory tract is approximately half the
total IWL if the ambient air temperature is 32.5°C and
the humidity is 50% (Sedin, 1995). However, in infants
undergoing mechanical ventilation there will be no insensible losses through the respiratory tract if the ventilator
gas mixture is humidified at body temperature.
Extraordinary losses are also seen in the neonate requiring intensive care. The most commonly encountered
extraordinary water losses occur when a nasogastric tube
is placed under continuous suction (to be discussed later
in this chapter). Large losses may also occur in association
with chest tubes, other drains, ostomies, and fistulas as well
as with emesis or diarrhea.
Management of Water Requirements
Appropriate management requires estimating any existing deficits or surpluses, calculating ongoing maintenance
needs because of usual sensible and insensible losses and
growth and additional needs as a result of extraordinary
losses. The infant’s prenatal history, birthweight, gestational age, and need for mechanical ventilation and the
environment in which the infant is to be cared for should
be considered when determining initial fluid and electrolyte needs. Frequent reevaluations are necessary. The most
useful parameter for monitoring fluid balance is the weight
of the baby, as rapid changes in weight will reflect changes
in water balance. Serial weights can be used to estimate the
IWL using the following formula:
IWL = Fluid intake − Urine output + Weight loss or
IWL = Fluid intake − Urine output − Weight gain
It is reasonable to initiate fluid volume based on the sum
of an allowance for sensible water loss of 30 to 60 mL/
kg/day and the estimated IWL. Figure 31-4 shows usual
IWL ranges by gestational and postnatal age. Factors previously outlined that predictably affect IWL should be
considered when prescribing fluids. Prevention of excessive IWL rather than replacement of increased IWL is
associated with fewer complications in the preterm neonate and usually can be achieved by modifying the infant’s
environment. See Table 31-1 for usual maintenance fluid
administration based on birthweight. These numbers are
guidelines for initial management only; the approach must
subsequently be individualized based on laboratory values
and other clinical data.
It is important to remember that the TBW excess and
extracellular volume expansion of preterm infants implies
that their negative water and sodium balance during the
first 5 to 10 postnatal days (see Figure 31-2; Shaffer and
Meade, 1989) represents an appropriate adaptation to extrauterine life and should not be compensated for by increased
fluid administration and sodium supplementation. If this
principle is not followed and a positive fluid balance (i.e.,
weight gain) is achieved during the transitional period,
preterm infants are at higher risk of a more severe course
of respiratory distress syndrome (Shaffer and Weismann,
1992) and a higher incidence of patent ductus arteriosus
(Bell et al, 1980), congestive heart failure (Bell et al, 1980),
TABLE 31-1 Estimated Maintenance Fluid Requirements
Fluid Requirements (mL/kg/day)
Birthweight (g)
<750
750-1000
1000-1500
>1500
Day 1
Day 2
Day 3-6
≥ Day 7
100-140
100-120
80-100
60-80
120-160
100-140
100-120
80-120
140-200
130-180
120-160
120-160
140-160
140-160
150
150
374
PART VIII Care of the High-Risk Infant
pulmonary edema (Shaffer and Weismann, 1992), necrotizing enterocolitis (Bell et al, 1979), and bronchopulmonary dysplasia (Oh et al, 2005; Van Marter et al, 1990).
Infants with ELBW and others with anticipated fluid
problems should be weighed daily or twice daily. Serum
sodium levels should be measured every 4 to 8 hours
untilstabilized, usually by 3 to 4 days after birth, and urine
output should be recorded and reviewed every 6 to 8 hours.
Once data are available, fluids should be increased if weight
loss is greater than 1% to 2% per day in term infants and
2% to 3% per day in preterm infants, if urine output is
low, if urine specific gravity is rising, or if serum sodium
concentration is rising. Overall, expected and appropriate
weight loss in the first week of life is up to 10% in term
infants and up to 20% in preterm infants. Conversely, fluids should be decreased if weight is not falling appropriately and serum sodium is decreasing. The goal is to reach
140 to 160 mL/kg/day of fluids by 7 to 10 days to allow for
adequate caloric intake.
Treatment of Fluid Overload
Fluid overload commonly occurs in sick neonates, often
because of the use of fluid bolus administration for hypotension. The diagnosis is based on weight gain, edema,
and often hyponatremia. Overhydration can sometimes be
prevented by the use of blood transfusions or dopamine
instead of colloid or crystalloid, if appropriate, for blood
pressure support. In addition to reducing the need for volume boluses, dopamine may facilitate the process of extracellular volume contraction via its renal and hormonal
effects (Seri, 1995). Once overhydration has occurred,
management is usually effected by 10% to 20% decrements of total daily fluid intake while carefully monitoring clinical and laboratory signs to ensure maintenance of
adequate intravascular volume as well as normal glucose
and electrolyte status while the ECF contraction occurs.
Treatment of Dehydration
Dehydration may be suspected based on clinical signs.
The total water deficit may be estimated by using weight
changes, calculating total inputs and outputs, and following serial sodium levels. Free water deficit (or excess) can
be calculated as:
H2 O deficit (or excess) (L) ={[0.7 × BW (kg) ] ×
}
[Na (mEq/L) ] current
−1
[Na (mEq/L) ] desired
In this formula, 0.7 × BW is the estimation of TBW.
When dehydration is diagnosed, correction should
generally occur over 24 hours, with half correction over
8 hours and the remainder over the next 16 hours. Longer
correction times are indicated when dehydration is accompanied by moderate to severe hypernatremia. Treatment is
best approached by considering separately the fluid resuscitation requirements, fluid to replace current deficits and
ongoing losses, and maintenance requirements, because
the volume of the fluid, the composition, and the rate of
replacement differ for each. Resuscitation of the intravascular volume to restore blood pressure and perfusion
should be provided with boluses of isotonic saline (0.9%
saline), and this volume is included in the initial half correction. The remaining deficit replacement volume should
TABLE 31-2 Free Water Content (as Volume %) of Common
Intravenous Solutions at Normal and High Serum
Sodium Concentrations*
Serum Sodium Concentration
145 mEq/L
Intravenous
Fluid
D5W
0.2% saline
0.45% saline
0.9% saline
Lactated
Ringer’s
solution
195 mEq/L
Isotonic
(%)
Water
(%)
Isotonic
(%)
Water
(%)
0
22
50
100
86
100
78
50
0
14
0
17
39
79
68
100
83
61
21
32
Modified from Molteni KH: Initial management of hypernatremic dehydration in the
breastfed infant, Clin Pediatr 33:731-740, 1994.
*Note that isotonic saline provides 21% free water when given to a patient with a
serum sodium concentration of 195 mEq/L and therefore will induce undesirable
decreases in serum sodium concentration when used for volume resuscitation in the
severely dehydrated hypernatremic neonate.
TABLE 31-3 Approximate Electrolyte Composition of Body
Fluids (mEq/L)
Body Fluid
Sodium
Potassium
Chloride
Gastric
Small intestine
Bile
Ileostomy
Diarrhea
20-80
100-140
120-140
45-135
10-90
5-20
5-15
5-15
3-15
10-80
100-150
90-130
80-120
20-115
10-110
be with fluid of appropriate sodium content based on the
serum sodium (see Sodium and Potassium Homeostasis
and Management, later), usually 0.2% or 0.45% saline.
Free water contents of the common intravenous fluids are
listed in Table 31-2. Usual maintenance fluids and electrolytes must also be provided. Ongoing urine losses should
be replaced volume for volume every 4 to 6 hours with
a solution tailored to the urine’s electrolyte concentration (usually 0.45% normal saline). Extraordinary losses
caused by tubes, drains, emesis, diarrhea, and ostomies
should always be sought in the dehydrated infant and also
accounted for in fluid management. The composition of
this latter replacement solution depends on the electrolyte
concentration of the fluid loss. The most common extraordinary loss, gastric fluid, contains significant sodium and
chloride. See Table 31-3 for approximate electrolyte compositions of body fluids.
Potassium replacement (usually by adding 20 to 40 mEq
potassium per liter of replacement fluid) should not begin
until adequate urine output is established. For additional
details of fluid correction and sodium management, see the
discussion under Management of Hypernatremia later in
this chapter.
Sodium and Potassium Homeostasis
and Management
Serum sodium values should be kept between 135 and 145
mEq/L. Sodium chloride supplementation at 1 to 2 mEq/
kg/day should be started in preterm and sick term neonates
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
only after completion of the postnatal extracellular volume contraction, usually after the first few days of life or
after more than 5% of birthweight is lost (Hartnoll et al,
2001). In general, as long as the infant’s fluid balance is
stable, maintenance sodium requirements do not exceed
3 to 4 mEq/kg/day, and providing this amount usually
ensures the positive sodium balance necessary for adequate
growth. Extreme prematurity and pathologic conditions
associated with delayed transition or disturbance of fluid
and electrolyte balance may significantly alter the infant’s
daily sodium requirement. For example, although the preterm infant has a limited ability to excrete a sodium load
(Hartnoll, 2003), some of the most immature infants may
have sodium requirements of as much as 6 to 8 mEq/kg/
day because of the decreased capacity of their kidneys
to retain sodium. Neonates recovering from an acute
renal insult and preterm infants with immature proximal
tubule functions who are in a state of extracellular volume
expansion (Ramiro-Tolentino et al, 1996) may need daily
sodium bicarbonate supplementation to compensate for
their greater renal bicarbonate losses.
Hyponatremia
Hyponatremia (serum sodium <130 mEq/L) represents
a deficit of sodium in relation to body water content and
may be caused by either total body sodium deficit or free
water excess. In both situations, total body water may be
decreased (hyponatremia with volume contraction), normal, or increased (hyponatremia with volume expansion).
Hyponatremia may be hypotonic, isotonic, or hypertonic.
To initiate effective treatment, it is important to attempt
to determine the primary cause of the hyponatremia and
whether there is associated volume expansion or contraction. The most common cause of hyponatremia in the sick
neonate is excessive administration or retention of free
water. In these situations the total body sodium content is
normal, and the appropriate treatment is restriction of free
water intake and not administration of sodium. In situations of true sodium deficit, the deficits can be estimated
by assuming 70% of total body weight as the distribution space of sodium. The formula for calculating sodium
deficit is:
Na+ deficit (or excess) (mEq) ≈ 0.7 × kg ×
[ (Na+ ) desired − [ (Na+ ) actual ]
In most situations of depletional hyponatremia, the
sodium deficit should be replaced on a schedule that provides two thirds replacement in the first 24 hours and the
remainder in the next 24 hours. Frequent measurements
of serum electrolytes are needed to ensure that the correction is occurring appropriately. If the serum sodium concentration is less than 120 mEq/L, regardless of whether
the hyponatremia is due to free water overload or total
body sodium deficit, then correction of the serum sodium
concentration up to 120 mEq/L is recommended with
administration of 3% saline solution (513 mEq of sodium
per liter). This correction should be done over 4 to 6
hours, depending on the severity of hyponatremia (Avner,
1995) and using the above formula. Although rapid intravenous bolus administration of 4 to 6 mL/kg of 3% saline
solution has been effective in children with seizures or
375
coma (Sarnaik et al, 1991), rapid and complete correction of low serum sodium concentration in adults with
chronic hyponatremia has been shown to be associated
with pontine and extrapontine myelinolysis. Once the risk
of acute central nervous system symptoms has been minimized and serum sodium concentration has reached 120
mEq/L, complete correction of hyponatremia should be
performed more slowly over the next 48 hours. In patients
with asymptomatic hyponatremia whose serum sodium
concentration exceeds 120 mEq/L, hypertonic infusions
are not indicated. Additional therapy should be directed
at fluid restriction if the hyponatremia is dilutional or
sodium repletion if the hyponatremia is depletional. The
use of 5% dextrose in water with 0.45% to 0.9% saline is a
reasonable replacement fluid for depletional hyponatremia
once the sodium is above 120 mEq/L. More stable infants
with chronic sodium losses can also be corrected with
enteral sodium chloride. Figure 31-5 summarizes the clinical evaluation and therapy of neonates with hyponatremia.
Hypernatremia
Hypernatremia (serum sodium >145 mEq/L) reflects a
deficiency of water relative to total body sodium and is most
often a disorder of water rather than sodium homeostasis.
Hypernatremia does not reflect the total body sodium content, which can be high, normal, or low depending on the
cause of the condition. Hypernatremia can also be associated with hypovolemia, normovolemia, or hypervolemia
(Box 31-2). If hypernatremia is primarily due to changes
in sodium balance, it can result from pure sodium gain
or, more commonly, sodium gain coupled with a lesser
degree of water accumulation or, rarely, water loss. The
hypernatremia-induced hypertonicity causes water to shift
from the intracellular to the extracellular compartment,
resulting in intracellular dehydration and the relative preservation of the extracellular compartment. This shift is the
main reason that neonates with chronic hypernatremic
dehydration often do not demonstrate overt clinical signs
of intravascular depletion and dehydration until late in the
course of the condition.
Compared with other organs, the central nervous
system has a unique adaptive capacity to respond to the
hypernatremia-induced hypertonicity, leading to a relative preservation of neuronal cell volume. The shrinkage
of the brain stimulates the uptake of electrolytes such as
sodium, potassium, and chloride (immediate effect) as
well as the synthesis of osmoprotective amino acids and
organic solutes (more delayed response). These idiogenic
osmols aid in maintaining normal brain cell volume during
longer periods of hyperosmolar stress (Trachtman, 1991).
As long as hypernatremia develops rapidly (within hours),
as in accidental sodium loading, a relatively rapid correction of the condition improves the prognosis without
raising the risk of cerebral edema formation. Intracellular
fluid accumulation does not occur because the accumulated electrolytes (sodium, potassium, and chloride) are
rapidly extruded from the brain cells, and cerebral edema
is unlikely. In these cases, reducing serum sodium concentration by 1 mEq/L per hour (24 mEq/L per day) is
appropriate (Adrogue and Madias, 2000).
However, because of the slow dissipation of idiogenic
osmols over a period of several days (Adrogue and Madias,
376
PART VIII Care of the High-Risk Infant
HYPONATREMIA
1. Exclude “pseudohyponatremia” (hyperlipidemia, hyperproteinemia)
2. Exclude hypertonic hyponatremia (↑ ECF osmolality due primarily to hyperglycosemia)
3. Evaluate ECF volume status (clinical and laboratory indicators)
Hypovolemic
↓↓ Total body sodium
↓ Total body water
Urine [Na]
<20 mEq/L
Urine [Na]
>20 mEq/L
Euvolemic
± Total body sodium
↑ Total body water
Hypervolemic
↑ Total body sodium
↑↑ Total body water
Urine [Na] usually
>20 mEq /L
Urine [Na]
<20 mEq/L
Urine [Na]
>20 mEq/L
Extrarenal losses:
1. Gastrointestinal
(vomiting, diarrhea,
drainage tubes,
fistulas)
2. Pleural effusions,
ascites
3. Ileus
4. Necrotizing
enterocolitis
Renal losses:
1. Diuretics
2. Osmotic diuresis
3. Contraction alkalosis
4. Mineralocorticoid
deficiency
5. Mineralocorticoid
unresponsiveness
6. Fanconi syndrome
7. Bartter’s syndrome
8. Obstructive uropathy
1. Glucocorticoid,
thyroid
2. Excess ADH
Edema-forming states:
1. Congestive heart
failure
2. Liver failure/cirrhosis
3. Nephrosis syndrome
4. Indomethacin therapy
Renal failure:
1. Acute
2. Chronic
Volume expansion
Volume expansion
Water restriction
Sodium and
water restriction
Sodium and
water restriction
FIGURE 31-5 Flow diagram for the clinical evaluation and therapy of neonates with hyponatremia. ADH, Antidiuretic hormone; ECF, extracellular
fluid. (Modified from Avner ED: Clinical disorders of water metabolism: hyponatremia and hypernatremia, Pediatr Ann 24:23-30, 1995.)
2000), in cases of chronic hypernatremia or in cases in
which the time frame is unknown, the hypernatremia
should be corrected more slowly, at a maximum rate of
0.5 mEq/L per hour (12 mEq/L per day). If correction is
performed more rapidly in these cases, the abrupt fall in
the extracellular tonicity results in the movement of water
into the brain cells, which have a relatively fixed hypertonicity because of the presence of the osmoprotective molecules. The result is the development of brain edema with
deleterious consequences (Adrogue and Madias, 2000;
Molteni, 1994).
In the breastfed term neonate, hypernatremia most commonly develops because of dehydration caused by inadequate breast milk intake (Molteni, 1994), but may also
be caused by high sodium levels in maternal breast milk.
Reduction in breastfeeding frequency has been shown to
be associated with a marked rise in the sodium concentration of breast milk (Neville et al, 1991). A vicious cycle
can ensue in which the infant sucks poorly, breast milk
production drops, sodium concentration rises, and the
infant becomes increasingly dehydrated, hypernatremic,
and lethargic. Recognition may be delayed because these
infants may appear quiet and content initially. Because this
process is chronic, usually occurring over 7 to 14 days,
signs of extracellular volume contraction are less prominent until the development of the full clinical presentation consisting of lethargy, irritability, abnormal muscle
tone with or without seizures, and cardiovascular collapse
BOX 31-2 C
onditions Causing
Hypernatremia
HYPOVOLEMIC HYPERNATREMIA
Inadequate breast milk intake
Diarrhea
Radiant warmers
Excessive sweating
Renal dysplasia
Osmotic diuresis
EUVOLEMIC HYPERNATREMIA
Decreased Production of Antidiuretic Hormone
Central diabetes insipidus, head trauma, central nervous system tumors (craniopharyngioma), meningitis, or encephalitis
Decrease or Absence of Renal Responsiveness
Nephrogenic diabetes insipidus, extreme immaturity, renal insult, and medications such as amphotericin, hydantoin, aminoglycosides
HYPERVOLEMIC HYPERNATREMIA
Improperly mixed formula
NaHCO3 administration
NaCl administration
Primary hyperaldosteronism
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
377
with renal failure. This presentation can be associated with
serious central nervous system morbidity from both the
hypertonicity (sagital or other venous sinus thrombosis,
subdural capillary hemorrhage, white matter injury) and
inappropriately rapid rehydration therapy (brain edema,
myelinolysis).
In the extremely immature neonate, hypernatremia
most commonly occurs from excessive transepidermal
free water losses. The condition usually develops rapidly,
within 24 to 48 hours after birth. The diagnosis is based on
the attendant decrease in body weight and clinical signs of
extracellular volume contraction. Prevention of this condition has been successful in the majority of immature neonates by frequent monitoring of serum electrolyte levels,
appropriate adjustments of free water intake, and the use
of humidified incubators (Modi, 2004).
The central and nephrogenic forms of diabetes insipidus
are much less commonly encountered and result in hypernatremia because of the lack of production of and renal
responsiveness to ADH, respectively. Hypernatremia can
also develop in response to excessive sodium supplementation, mainly in the sick neonate receiving repeated volume
boluses for cardiovascular support. In these cases, clinical
signs of edema, increased body weight, and the history of
volume boluses help to establish the diagnosis.
And the amount of free water required to decrease
serum sodium by 12 mEq/L over a 24-hour period when
hypernatremia is severe is calculated as:
Treatment of Hypernatremia
Percentage of free water content =
1 − (IV fluid sodium / Serum sodium)
Thorough analysis of the medical history and the changes
in clinical signs, laboratory findings, and body weight
usually aid in determining the major etiologic factor in
hypernatremia and thus the appropriate treatment. In the
critically ill infant, the cause of the serum sodium abnormality may be multifactorial however, and the treatment
less straightforward. Although some cases of hypernatremia are a result of sodium excess with normal or high
TBW, most cases in neonates are due to hypernatremic
dehydration. Treatment of this condition is generally
divided into two phases: the emergent phase where the
intravascular volume is restored, usually by administration
of 10 to 20 mL/kg of isotonic saline, and the rehydration
phase, where the sum of the remaining free water deficit
and usual maintenance needs are administered evenly over
at least 48 hours.
The free water deficit can be calculated as:
H2 O deficit
(or excess) (L) = [0.7 × BW (kg) ] ×
{
}
1 −[NA+ (mEq/L) ] current /[NA+ (mEq/L)] desired
In this formula, (0.7 × BW) is the estimation of TBW.
It is important to note that the amount of free water
required to decrease the serum sodium by 1 mEq/L
is 4 mL/kg with moderate hypernatremia, but only
3 mL/kg when the hypernatremia is as high as 195 mEq/L
(Molteni, 1994). Therefore the amount of free water
required to decrease serum sodium by 12 mEq/L over a
24-hour period when hypernatremia is moderate is cal
culated as:
Free water required = Current weight (kg) ×
4mL/kg × 12mEq/L or
Free water required = Current weight ×
48 mL/kg /day
Free water required = Current weight × 36 mL / kg / day
The free water contents of the common intravenous
fluids are listed in Table 31-2. In most mild to moderate hypernatremic states, during the rehydration phase,
replacement fluids of 5% dextrose in 0.2% normal saline
(31 mEq/L) or 0.45% normal saline (77 mEq/L) are
appropriate. Infants with serum sodium levels greater than
165 mEq/L should initially be given 0.9% saline to avoid
sudden drops in serum sodium concentration. When the
serum sodium is greater than 175 mEq/L, however, normal saline will be hypotonic compared with the patient’s
serum. In these instances of severe hypernatremia, an
appropriate amount of 3% saline (513 mEq/L) should
be added to the intravenous (IV) fluid so that the sodium
concentration is approximately 10 to 15 mEq/L less than
the serum sodium level (Rand and Kolberg, 2001). The
relative free water content of an IV solution for a specific
patient with sodium perturbations can be calculated using
the formula:
Serum electrolytes should be monitored every 2 to 4
hours until the desired rate of decline in serum sodium
concentration is established. At this point, the frequency
of the laboratory measurements can be relaxed to every
4 to 6 hours until the serum sodium concentration is less
than 150 mEq/L. The speed of correction of hypernatremia depends on the rate of its development. This approach
provides a reasonable chance that the serum sodium concentration will gradually decrease to the normal range over
2 to 4 days. Except in cases of acute massive sodium overload, the goal should be to drop the serum sodium concentration at a rate no greater than 1 mEq/L/h. A slower pace
of correction of 0.5 mEq/L/h is prudent in patients with
hypernatremia of chronic or unknown duration to avoid
iatrogenic central nervous system sequelae.
Once serum sodium concentration, urine output, and
renal function are normal, the patient should receive standard maintenance fluids, either intravenously or orally,
depending on his or her condition. At this time, electrolyte
status must still be monitored for an additional 24 hours
to ensure that complete recovery has occurred. Hyperglycemia and hypocalcemia commonly accompany hypernatremia. The use of insulin to treat the hyperglycemia is
not recommended, because it can increase brain idiogenic
osmol content. Hypocalcemia should be corrected with
appropriate calcium supplementation.
Potassium Homeostasis and Management
Serum potassium should be kept between 3.5 and 5
mEq/L. In the early postnatal period, neonates, especially
immature preterm infants, have higher serum potassium
concentrations than older persons. The etiology of the
relative hyperkalemia of the newborn is multifactorial and
378
PART VIII Care of the High-Risk Infant
involves developmentally regulated differences in renal
function, Na+,K+-ATPase activity (Vasarhelyi et al, 2000),
and hormonal milieu. Exposure to prenatal steroids in premature infants is associated with a decreased incidence of
hyperkalemia, believed to be due to improved renal function (Omar et al, 2000).
In general, potassium supplementation should be started
only after urine output has been well established, usually
by the third postnatal day. Supplementation should be
started at 1 to 2 mEq/kg/day and increased over 1 to 2
days to the usual maintenance requirement of 2 to 3 mEq/
kg/day. Some preterm infants may need more potassium
supplementation after the completion of their postnatal
volume contraction, because of their increased plasma
aldosterone concentrations, prostaglandin excretion, and
disproportionately high urine flow rates. Most term and
preterm neonates will require potassium supplementation
if they are receiving diuretics.
Hypokalemia
Hypokalemia in the neonate is usually defined as a serum
potassium level of less than 3.5 mEq/L. Hypokalemia
can occur from potassium loss due to diuretics, diarrhea,
renal dysfunction, or nasogastric drainage from inadequate
potassium intake or from intracellular movement of potassium in the presence of alkalosis. Except in patients receiving digoxin, hypokalemia is rarely symptomatic until the
serum potassium concentration is less than 2.5 mEq/L.
Electrocardiogram (ECG) manifestations of hypokalemia
include flattened T waves, prolongation of the QT interval, or the appearance of U waves. Severe hypokalemia can
result in cardiac arrhythmias, ileus, and lethargy.
Treatment of Hypokalemia
Hypokalemia is treated by slowly replacing potassium
either intravenously or orally, usually in the daily fluids.
Rapid administration of potassium chloride is not recommended, because it is associated with life-threatening
cardiac dysfunction. In extreme emergencies, potassium
can be given as an infusion over 30 to 60 minutes of not
more than 0.3 mEq/kg potassium chloride. If hypokalemia
is secondary to alkalosis, the alkalosis should be corrected
before considering increasing the potassium intake.
Hyperkalemia
Hyperkalemia in the neonate is defined as a serum potassium level greater than 6 mEq/L in a nonhemolyzed specimen. It is important to understand that most of the body’s
potassium is contained within cells; therefore serum potassium levels do not accurately reflect total body stores.
However, a serum potassium greater than 6.5 to 7 mEq/L
can be life threatening, even if stores are normal or low,
because of its effect on cardiac rhythm. ECG manifestations of hyperkalemia include peaked T waves (the earliest
sign), a widened QRS configuration, bradycardia, tachycardia, supraventricular tachycardia, ventricular tachycardia, and ventricular fibrillation. Because pH affects the
distribution of potassium between the intracellular and
the extracellular space, serum potassium levels rise during
acidosis, which may occur acutely. The clinician should
be aware of the potential for life-threatening arrhythmias
to occur in infants with chronic lung disease on diuretics
and potassium supplements who develop a sudden respiratory deterioration with acidosis. Another common cause of
hyperkalemia is renal dysfunction, of particular concern in
very preterm and asphyxiated infants. In addition, infants
who have suffered intraventricular hemorrhage or tissue
trauma and those with intravascular hemolysis often have
hyperkalemia caused by the release of potassium during
breakdown of red blood cells. Finally, hyperkalemia may
be one of the earliest manifestations of congenital adrenal
hyperplasia.
Treatment of Hyperkalemia
When hyperkalemia is diagnosed, all potassium intake
should be discontinued and the ECG should be monitored.
Table 31-4 presents medications used in management of
significant hyperkalemia. Calcium gluconate stabilizes
cardiac membranes and alkali therapy, insulin, and inhaled
albuterol (Singh et al, 2002) all rapidly enhance cellular
uptake of potassium and can acutely drop serum potassium
levels, but not decrease total body potassium. Intravenous
furosemide and rectally administered sodium polystyrene
sulfonate (Kayexalate) enhance potassium excretion and
will lower total body stores, but they require several hours
to take effect. Dialysis or exchange transfusion may be
used when the hyperkalemia is life threatening and these
measures do not result in improvement.
CLINICAL CONDITIONS ASSOCIATED WITH
FLUID AND ELECTROLYTE DISTURBANCES
Extreme Prematurity
Infants born between 23 and 27 weeks’ gestation, or with
a birthweight of <1000 g (i.e., ELBW), are at particular
risk for acute abnormalities of both fluid and electrolyte
status in the immediate postnatal period. Their transepidermal water loss is much higher than that in more mature
preterm neonates (see Figure 31-4), and it is difficult for
their water balance to be maintained unless the excessive
losses are prevented. It should be kept in mind that such
an infant, when cared for in an open warmer without the
use of a plastic heat shield, may lose up to 150 to 300 mL/
kg/day of free water through the skin during the first 3 to 5
days of life. Infants whose mothers received antenatal glucocorticoids often have fewer problems because prenatal
glucocorticoids enhance maturation of the epidermis and
result in increases in urine output and fractional excretion
of sodium (Ali et al, 2000; Omar et al, 1999).
Although the IWL primarily affects the extracellular
volume, the intracellular compartment ultimately shares
the loss of free water as osmotic pressure in the extracellular compartment rises. As water leaves the cells, intracellular osmolality rises and cell volume diminishes. In the
central nervous system, as described earlier, these changes
stimulate the generation of idiogenic osmols, resulting in
selective increases in intracellular osmolality and a tendency toward normalization of neuronal cell volumes.
This protective mechanism has significant clinical implications for the rate at which hypernatremia should be corrected, and the decrease in serum sodium concentration
should not exceed 12 mEq/L/day, especially in the infant
in whom the hypernatremia has been chronic (>12 hours).
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
379
TABLE 31-4 Medications Used for Treatment of Hyperkalemia
Medication
Dosage
Onset
Length
of Effects
Calcium
gluconate
100 mg/kg IV over
2-5 min
Immediate
30 min
Protects the myocardium from toxic
effects of potassium; no effect on total
body potassium
Can worsen digoxin
toxicity
Sodium
bicarbonate
1-2 mEq/kg
Immediate
Variable
Shifts potassium intracellularly; no
effect on total body potassium
Maximum infusion:
mEq/min in emergency
situations
Tromethamine
3-5 mL/kg
Immediate
Variable
Shifts potassium intracellularly; no
effect on total body potassium
—
Insulin plus
dextrose
Insulin 0.1-0.15 U/
kg IV plus dextrose
0.5 g/kg IV
15 to 30 min
2 to 6 h
Shifts potassium intracellularly; no
effect on total body potassium
Monitor for
hypoglycemia
Albuterol*
0.15 mg/kg every
20 min for three
doses then 0.15-0.3
mg/kg
15 to 30 min
2 to 3 h
Shifts potassium intracellularly; no
effect on total body potassium
Minimum dose, 2.5 mg
Furosemide
PO: 1-4 mg/kg/dose
1-2 times/day
IV: 1-2 mg/kg/dose
given every 12-24
hours
15 min to 1 h
4h
Increases renal excretion of potassium
—
Kayexalate
1 g/kg PR every 6
hours
1 to 2 h
(rectal route
faster)
4 to 6 h
Removes potassium from the gut in
exchange for sodium
Use with extreme caution in neonates, especialy preterm; contains
sorbital; may be associated with bowel necrosis
and sodium retention
Mechanism of Action
Comments
and Cautions
IV, Intravenous; PO, by mouth or gastric tube; PR, per rectum.
*From Singh BS, Sadiq HF, Noguchi A, et al: Efficacy of albuterol inhalation in treatment of hyperkalemia in premature neonates, J Pediatr 141:16, 2002.
Because serum sodium concentration is a reliable clinical indicator of extracellular tonicity, monitoring of this
parameter every 6 to 12 hours during the first 2 to 3 postnatal days coupled with daily (or twice daily) measurements of body weight provide valuable information and
appropriate guidance for the fluid and electrolyte management of the extremely immature preterm neonate. This is
especially true in the absence of prenatal steroid exposure
and if the infant is not cared for in a humidified incubator.
Serum osmolality should be directly measured in patients
in whom calculated serum osmolality is more than 300 to
320 mOsm/L.
Table 31-1 shows suggested maintenance fluid requirements by birthweight and postnatal day of life. Because
immature neonates in an incubator with an ambient air
humidity of 50% to 80% require significantly less free
water and less frequent measurements of serum electrolyte
and osmolality (Sedin, 1995), open radiant warmers should
be used only for critically ill, extremely labile preterm
infants requiring frequent hands-on medical management.
In these cases, the use of a protective plastic heat shield can
help decrease excessive evaporative losses, and total daily
fluid intake, similar to that used for infants in incubators,
may be started at approximately 100 mL/kg/day containing 5% to 10% dextrose in water with close monitoring
for dehydration. Daily fluid intake is then increased by 10
to 30 mL/kg/day every 6 to 12 hours if the serum sodium
concentration rises from the baseline, the goal being
to keep serum sodium concentration below 145 to 150
mEq/L. As skin integrity improves during the course of
the 2nd to 3rd days, serum sodium concentration starts to
fall. At this time, a significant stepwise limitation of total
fluid intake is obligatory to allow a complete contraction
of the extracellular volume to occur and to minimize the
possibility of free water overload with its attendant risks
for the development of ductal patency, pulmonary edema,
and worsening lung disease.
Potassium chloride supplementation may be started as
soon as urine output has been established and the serum
potassium concentration is less than 5 mEq/L. Extremely
premature infants are at risk for the development of both
oliguric and nonoliguric hyperkalemia, so the serum
potassium concentration should be monitored closely, and
supplementation should be discontinued if warranted by
changes in serum potassium values or in renal function.
Critically ill, extremely immature neonates often receive
excess sodium with volume boluses, medications, and the
maintenance infusion of their arterial lines (Costarino et
al, 1992). Therefore extra sodium supplementation usually
should not be started during the first few postnatal days, to
prevent a rise in total body sodium concentration and thus
in extracellular volume, which will hinder the appropriate
postnatal diuresis.
Many critically ill preterm infants retain their originally high extracellular volumes, even when sodium and
water intakes are restricted, and such neonates also tend
to lose more bicarbonate in the urine. Interestingly, proximal tubular bicarbonate reabsorption may be appropriate
even in the VLBW infant despite the immaturity of their
renal functions, as long as extracellular volume contraction
380
PART VIII Care of the High-Risk Infant
takes place (Ramiro-Tolentino et al, 1996). Therefore the
presence of the extracellular volume expansion appears to
be an important factor in the renal bicarbonate wasting in
these infants. The diagnosis of functional proximal tubular
acidosis in such cases should not rely solely on the finding
of an alkaline urine pH, because the distal tubular function
is usually mature enough to acidify the urine once serum
bicarbonate has decreased to its new threshold. Provided
that liver function is normal, daily supplementation of
bicarbonate in the form of sodium acetate, potassium acetate, or both normalizes blood pH and serum bicarbonate
in these infants and also increases urine pH, aiding in the
diagnosis. Once extracellular volume contraction occurs,
these neonates generally achieve a positive bicarbonate
balance (Ramiro-Tolentino et al, 1996), and supplementation becomes unnecessary.
Other general guidelines in the fluid and electrolyte
management of the immature preterm infant during the
1st week of life are (1) daily calculation of fluid balance
and estimation of sodium balance; (2) daily measurements
of body weight, serum electrolytes, blood urea nitrogen,
creatinine, and plasma glucose; and (3) testing of urine
samples for glucose, and osmolality or specific gravity.
The frequency of testing and the addition of other tests,
including the measurement of serum albumin concentration and osmolality, depend on the clinical status, severity
of underlying disease, and fluid and electrolyte disturbance
of the individual patient.
Respiratory Distress Syndrome
There is a well-established relationship between fluid and
electrolyte imbalance and respiratory distress syndrome.
Surfactant deficiency results in pulmonary atelectasis,
elevated pulmonary vascular resistance, poor lung compliance, and decreased lymphatic drainage. In addition,
preterm infants have low plasma oncotic and critical pulmonary capillary pressures and suffer pulmonary capillary
endothelial injury from mechanical ventilation, oxygen
administration, and perinatal hypoxia (Dudek and Garcia,
2001; Sola and Gregory, 1981). These abnormalities alter
the balance of the Starling forces in the pulmonary microcirculation, leading to interstitial edema formation with
further impairment in pulmonary functions.
In the presurfactant era, an improvement in pulmonary
function occurred only during the 3rd to 4th postnatal
day. This improvement was usually preceded by a period
of brisk diuresis characterized by small increases in glomerular filtration rate and sodium clearance and a larger
rise in free water clearance (Costarino and Baumgart,
1991). Although the exact mechanism for this diuresis is
not known, it is likely that improving endogenous surfactant production and capillary integrity promoted the
recovery of the pulmonary capillary endothelium and lymphatic drainage. The ensuing changes in Starling forces
then favored reabsorption of the hypotonic interstitial
lung fluid into the circulation, and a delayed physiologic
diuresis took place.
Currently, with the routine use of surfactant and antenatal steroids, the pulmonary compromise and its consequences are less severe. However, because significant
improvements in lung function take place only after the
majority of the excess free water is excreted (Costarino
and Baumgart, 1991), daily fluid intake should still be
restricted to allow the extracellular volume contraction to
take place. If this principle is not followed and a positive
fluid balance occurs, preterm infants with respiratory distress syndrome are at higher risk for a more severe course
of acute lung disease and have a higher incidence of patent
ductus arteriosus, congestive heart failure, and necrotizing
enterocolitis as well as a greater severity of the ensuing
bronchopulmonary dysplasia.
Antenatal administration of steroids and postnatal use
of surfactants have clearly altered the course and clinical
presentation of respiratory distress syndrome (Ballard and
Ballard, 1995; Kari et al, 1994). Antenatal steroid administration accelerates maturation of organs including those
involved in the regulation of fluid and electrolyte balance
(Ballard and Ballard, 1995), whereas the use of exogenous
surfactant decreases pulmonary capillary leak and edema
formation (Carlton et al, 1995). Furthermore, surfactant administration does not alter the rate and timing of
ductal closure (Reller et al, 1993), although it may affect
the pattern of shunting through the ductus arteriosus in
the acute period (Kaapa et al, 1993; Kluckow and Evans,
2000). Thus, these interventions generally enhance extracellular volume contraction and aid in the stabilization of
fluid and electrolyte homeostasis in preterm neonates with
respiratory distress syndrome. However, maintenance of
a negative water and sodium balance during the first few
days of life remains the cornerstone of fluid and electrolyte
management in these infants (Tammela, 1995; Van Marter
et al, 1990).
On the basis of the events in the pathophysiology of
pulmonary edema formation in these infants, the use of
furosemide has long been suggested to promote a negative
fluid balance and to directly inhibit pulmonary epithelial
transport processes involved in edema formation in the
lungs (Green et al, 1988; Yeh et al, 1984). However, furosemide induces only short-term improvements in pulmonary function in these patients, and no beneficial effects on
long-term morbidity or mortality have been documented.
Moreover, prophylactic use of the drug during the first
postnatal days can lead to intravascular volume depletion
with hypotension, tachycardia, and decreased peripheral
perfusion as well as to acute and chronic disturbances in
serum electrolytes and thus osmolality (Green et al, 1988;
Shaffer and Weismann, 1992; Yeh et al, 1984). Furthermore, continued administration may be associated with
an increased incidence of patent ductus arteriosus (Green
et al, 1983). Therefore the use of furosemide during this
period should be restricted to patients with oliguria of renal
origin whose intravascular volume appears to be adequate.
Chronic Lung Disease or
Bronchopulmonary Dysplasia
Low gestational age and birthweight, lack of antenatal
steroid administration, severe respiratory distress syndrome with oxygen toxicity, volutrauma and barotrauma,
air leak, inflammation, patent ductus arteriosus, and insufficient nutrition are among the known etiologic factors
for the development of bronchopulmonary dysplasia. In
addition, a high fluid and salt intake during the first weeks
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
of life has been shown to increase the incidence and severity of chronic lung disease. Specifically, higher fluid intake
and lack of appropriate weight loss in the first 10 days
of life are associated with significantly higher risk for
bronchopulmonary dysplasia, even after controlling for
other known risk factors such as those listed previously
(Oh et al, 2005). Therefore careful fluid and electrolyte
management during the first weeks of life, allowing for
the appropriate degree of weight loss, is of great importance in decreasing the incidence and severity of this condition (see Chapter 48).
Patent Ductus Arteriosus
A PDA has been associated with an increase in morbidity
and mortality in epidemiological studies, especially in the
infant with ELBW. However, trials aimed at prophylactic
or therapeutic closure of the ductus have not demonstrated
improvements in these outcomes. Several conditions,
including hypoxemia, unstable cardiovascular status, metabolic acidosis, increases in extracellular volume, inflammatory mediators, and ductal prostaglandin synthesis,
have been recognized to prolong patency of the ductus
arteriosus (Hammerman, 1995; see Chapter 54). Accordingly, clinical management aimed at preventing the occurrence of ductal patency involves interventions that keep
the cardiovascular status and oxygenation stable, minimize
inflammation, restrict fluid intake, and maintain low levels
of local prostaglandin synthesis by the administration of
indomethacin (Clyman, 1996). Pharmacologic ductal closure with indomethacin is normally indicated during the
first 2 postnatal weeks, because ductal sensitivity to prostaglandins rapidly diminishes thereafter (Clyman, 1996;
Van Overmeire et al, 2000). Ibuprofen has been offered
as an alternative inhibitor of cyclooxygenase, the enzyme
responsible for prostaglandin synthesis, because of its
equivalent efficacy in closing the symptomatic PDA with
fewer adverse effects (Ohlsson et al, 2008). Specifically,
ibuprofen may cause less renal dysfunction (Van Overmeire et al, 2000) and cerebral vasoconstriction (Mosca
et al, 1997; Patel et al, 2000) when used in this manner.
However, ibuprofen is not recommended for prophylactic
use because it has an adverse effect on renal function in
this setting and does not reduce grade III and IV IVH as
indomethacin has been proven to do (Shah and Ohlssen,
2006). For more details, see Chapter 61.
Under physiologic circumstances in the immediate postnatal period, renal prostaglandin production is increased
to counterbalance the renal actions of vasoconstrictor and
sodium- and water-retaining hormones released during
labor and delivery (Bonvalet et al, 1987; Gleason, 1987).
Compared with the renal function of the adult kidney
in euvolemia, the neonatal kidney is more dependent on
the increased production of vasodilatory and natriuretic
prostaglandins, rendering it more sensitive to the vasoconstrictive and sodium- and water-retaining actions of
cyclooxygenase inhibition. In the preterm infant, indomethacin administration has been shown to have clinically
significant, although mostly transient, renal side effects
because of decreased prostaglandin production through
inhibition of cyclooxygenase. In the indomethacin-treated
neonate, the unopposed renal vasoconstriction and sodium
381
and water reabsorption leads to decreases in renal blood
flow and glomerular filtration rate and to increases in
sodium and free water reabsorption. These side effects
occur despite the diminishing left-to-right shunt through
the closing ductus. Characteristic clinical findings include
a rise in serum creatinine level, oliguria, and hyponatremia (Cifuentes et al, 1979). Hyponatremia occurs because
the free water retention caused by the unopposed renal
actions of high plasma vasopressin levels is out of proportion to the sodium retention induced by angiotensin and
noradrenaline. This pattern of renal response can most
likely be explained by the fact that in a preterm infant,
the function of the distal tubule is more mature than that
of the proximal tubule (Lumbers et al, 1988), leading to
an expanded but somewhat hypotonic extracellular space.
Therefore fluid management of the preterm infant receiving indomethacin must focus on maintaining an appropriately restricted fluid intake and avoiding extra sodium
supplementation. As the prostaglandin inhibitory effects of
indomethacin diminish following the last dose, renal prostaglandin production returns to normal, and the retained
sodium and excess free water are usually rapidly excreted,
especially with the improvement in the cardiovascular status as the ductal shunt decreases.
Because furosemide increases prostaglandin production, the drug has been hypothesized to attenuate the
renal side effects of indomethacin if the intravascular volume is judged to be adequate (Yeh et al, 1982). However,
furosemide administration can increase the incidence of
ductal patency (Green et al, 1983), so routine use of the
drug might not be prudent in preterm infants treated with
indomethacin, because of the theoretical risk of reopening
the ductus arteriosus (Seri, 1995). Furthermore, the concomitant use of furosemide and indomethacin has been
shown to worsen renal function, as evidenced by increases
in serum creatinine and worsening hyponatremia without
increasing overall urine output (Andriessen et al, 2009).
As a result, furosemide should not be used early in life
when the PDA is also being actively managed. In cases of
hemodynamic instability or to avoid the potential effect of
furosemide on ductal closure, dopamine infusion can be
used to support the cardiovascular status and attenuate the
indomethacin-induced oliguria (Cochran et al, 1989; Seri
et al, 1984, 1993). However, there is controversy regarding the efficacy and clinical significance of this intervention (Baenziger et al, 1999; Fajardo et al, 1992; Seri, 1995).
Growing Premature Infant With Negative
Sodium Balance
The 2- to 6-week-old growing preterm neonate who does
not have significant chronic lung disease and is not undergoing diuretic treatment may have hyponatremia (serum
sodium concentration 125 to 129 mEq/L) because of a relative sodium deficiency (Sulyok et al, 1979, 1985). Despite
the low total body sodium and high activity of sodium
retaining hormones, these infants continue to lose sodium
in the urine mainly because of immature renal function.
Although the infant is usually in a positive sodium balance,
it is insufficient to compete with the increased sodium
demand because of growth. The treatment of this condition is to provide extra sodium supplementation in the
382
PART VIII Care of the High-Risk Infant
form of sodium chloride (usually 2 to 4 mEq/kg/day) to
keep serum sodium values greater than 130 mEq/L.
Shock and Edema
In the uncompensated phase of shock, blood pressure is
low; cardiac output may be low, normal, or high; effective circulating blood volume is usually decreased; transcapillary hydrostatic pressure is elevated; and capillary
integrity and lymphatic drainage are impaired, resulting
in edema formation and increased interstitial compliance.
The latter condition further enhances fluid accumulation
in the interstitium. The changes in the effective circulating blood volume also trigger the release of antidiuretic
hormones, including catecholamines, renin-angiotensinaldosterone, and vasopressin, resulting in the retention of
sodium and free water. The specific cause of shock (e.g.,
infection, asphyxia, myocardial insufficiency, hypovolemia) may independently contribute to this chain of events,
further compromising fluid and electrolyte balance. In
affected infants, treatment is directed at normalizing tissue perfusion and oxygen delivery by restoring effective
intravascular volume, cardiac output, and renal function
with the use of vasopressor and inotropic support, as
well as with the judicious use of volume expanders while
monitoring blood pressure, cardiac output, and changes
in organ blood flow (see Chapter 51). In shock refractory
to these therapies, early initiation of low-dose glucocorticoid and mineralocorticoid replacement may help to break
the vicious cycle by improving capillary integrity and thus
effective circulating blood volume, and by potentiating
the cardiovascular response to vasopressors and inotropic
agents (Seri et al, 2000).
Syndrome of Inappropriate Anti-Diuretic
Hormone Secretion
SIADH may be associated with birth asphyxia, intracerebral hemorrhage, respiratory distress syndrome, pneumothorax, and the use of continuous positive-pressure
ventilation (El-Dahr and Chevalier, 1990; Leake, 1992).
The syndrome is characterized by oliguria, free water
retention, decreased serum sodium concentration and
serum osmolality, increased urine concentration, and
weight gain caused by edema formation. However,
because the urinary concentrating capacity of the newborn is limited, a less than maximally diluted urine satisfies the diagnosis of SIADH in the presence of the other
symptoms. The treatment is based on fluid and sodium
restriction despite the oliguria and hyponatremia, as well
as on appropriate circulatory and ventilatory support. The
clinician must remember that total body sodium is normal, but TBW is elevated in such an infant, and that it is
particularly dangerous to treat the hyponatremia caused
by free water retention with large amounts of sodium.
Because of their more immature renal functions, infants
with extreme LBW during the first few weeks of life usually do not exhibit the full-blown syndrome despite their
sometimes excessively high plasma vasopressin levels
(Aperia et al, 1983).
Diminished vasopressin secretion or complete unresponsiveness of the renal tubules to vasopressin results
in polyuria, dilute urine production, and increased serum
osmolality (Leake, 1992), otherwise known as diabetes
insipidus (DI). This condition is not common in neonates,
but can occur in association with central nervous system
injury or disease, such as in meningitis or cerebral hemorrhage affecting the pituitary gland (central DI) or in an
inherited form (nephrogenic DI). The treatment of neonates with this condition consists of allowing for adequate
free water intake and the use of desmopressin.
Surgical Conditions
Surgery has a major effect on metabolism, fluid balance,
and electrolyte balance in the newborn. Preterm infants
with acute or chronic lung disease are especially sensitive
and respond to the procedure with significant catabolic
responses, increases in capillary permeability with the
attendant shift of fluid into the interstitial space, and retention of sodium and free water (John et al, 1989). The retention of sodium and free water is secondary to the decrease
in effective circulating blood volume and to the increased
plasma levels of sodium- and water-retaining hormones,
including catecholamines, renin-angiotensin-aldosterone,
and vasopressin.
Preoperative management has a significant effect on
outcome and should be aimed at maintaining adequate
effective circulating blood volume as well as cardiovascular and renal function. In preterm infants who have
evidence of absolute or relative adrenal insufficiency
(Watterberg, 2002), the provision of stress doses of steroids may be necessary. In the postoperative period,
maintenance of the integrity of the cardiovascular system
through the judicious use of volume expanders and pressor support, meticulous replacement of ongoing surgical
and nonsurgical fluid and electrolyte losses, close monitoring, and intense and effective communication between
the neonatal and surgical teams are essential to ensure
a successful outcome. As capillary integrity improves,
reabsorption and excretion of the expanded interstitial
fluid volume occurs, with normalization in the secretion
of hormones regulating fluid and electrolyte balance. At
this time, the provision of maximized nutritional support
becomes essential to restore the anabolic state and growth
of the infant.
The most commonly encountered surgical water losses
occur when a nasogastric tube is placed under continuous suction to provide relief for the gastrointestinal tract
in conditions such as necrotizing enterocolitis and postoperative management after abdominal surgery. Because
these losses may be substantial, they should be monitored and a portion of it should be replaced every 6 to
12 hours to maintain appropriate water and electrolyte
balance. However, free water retention often develops
after surgery; therefore full replacement of the nasogastric free water loss is not usually recommended. The
composition of the replacement solution depends on the
electrolyte concentration of the fluid loss. Gastric fluid
usually contains 50 to 60 mEq/L of sodium chloride,
and therefore, 0.45% sodium chloride with potassium
is normally used as the fluid of choice for replacement.
See Table 31-3 for estimated electrolyte compositions
of body fluids.
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
ACID-BASE BALANCE
PHYSIOLOGY OF ACID-BASE BALANCE
REGULATION
Like adults, newborns must maintain their extracellular pH, or hydrogen ion concentration, within a narrow
range. A normal pH is essential for intact functioning of
all enzymatic processes and, therefore, the intact functioning of all organ systems of the body. Newborns are
subjected to many stresses that can affect their acid-base
balance. In addition, neonates, especially if they are premature, have a limited ability to compensate for acid-base
alterations; therefore acid-base disturbances are common
in the neonatal period. An understanding of the principles
of acid-base regulation is essential for proper diagnosis and
treatment of these disturbances.
In healthy humans, the normal range of ECF hydrogen ion concentration is 35 to 45 mEq/L. Because pH is
defined as the negative logarithm of hydrogen ion concentration (pH = −log[H+]), these values of hydrogen ion
concentration correspond to a pH range of 7.35 to 7.45.
Acidosis is a downward shift in pH to less than 7.35, and
alkalosis is an upward shift in pH to more than 7.45. Alterations in normal pH are resisted by complex physiologic
regulatory mechanisms. The main systems that maintain
pH are the body’s buffer systems, the respiratory system,
and the kidneys. Some of these systems respond immediately to sudden alterations in hydrogen ion concentration,
whereas others respond more slowly to changes but maintain the overall balance between acid and base production,
intake, metabolism, and excretion over the long term.
The physiologic regulatory systems that respond immediately to changes in acid-base balance include the various
intracellular and extracellular buffers as well as the lungs.
A buffer is a substance that can minimize changes in pH
when acid or base is added to the system. The extracellular
buffers, which include the bicarbonate-carbonic acid system, phosphates, and plasma proteins, act rapidly to return
the extracellular pH toward normal. The intracellular buffers, which include hemoglobin, organic phosphates, and
bone apatite, act more slowly and require several hours to
reach maximal capacity.
The most important extracellular buffer is the plasma
bicarbonate–carbonic acid buffer system, in which the acid
component (carbonic acid [H2CO3]) is regulated by the
lungs, and the base component (bicarbonate [HCO3−]) is
regulated by the kidneys. The buffer equation is:
H + + HCO3− ↔ H2 CO3 ↔ H2 O + (CO2 )d
where (CO2)d represents the dissolved carbon dioxide. At
equilibrium, the amount of (CO2)d exceeds that of H2CO3
by a factor of 800:1; therefore for practical purposes,
(CO2)d and H2CO3 can be treated interchangeably. The
fact that CO2 excretion can be controlled by the respiratory system markedly improves the efficiency of this buffer
system at physiologic pH. The enzyme carbonic anhydrase allows rapid interconversion of H2CO3 to H2O and
CO2. If the hydrogen ion (H+) concentration increases for
any reason, hydrogen combines with HCO3−, driving the
buffer reaction toward greater production of H2CO3 and
383
CO2. Carbon dioxide crosses the blood-brain barrier and
stimulates central nervous system chemoreceptors, leading
to increased alveolar ventilation and decreased concentration of extracellular CO2. This respiratory compensation
begins within minutes after a pH change and is complete
within 12 to 24 hours. A similar compensation occurs in
response to a decrease in H+ concentration, leading to
decreased alveolar ventilation and a resultant increase in
extracellular CO2.
The relationship of the two components of the bicarbonate–carbonic acid buffer system to pH is expressed by
the Henderson-Hasselbalch equation:
pH = pK + log
[HCO3− ]
[H2 CO3 ]
Because H2CO3 is in equilibrium with the dissolved CO2
in the plasma, and because the amount of dissolved CO2
depends on the partial pressure of CO2, the equation can
be modified as:
pH = pK + log
[HCO3− ]
× PaCO2
0.03
Both the original equation and the modified equation are
clinically difficult to use; therefore the modified Henderson-Hasselbalch equation can be rewritten as the Henderson equation without logarithms for easier clinical use.
[H+ ] = 24 ×
PaCO2
[HCO3− ]
This last equation clearly indicates the clinically most
important aspect of acid-base regulation by the bicarbonate-carbonic acid buffer system, that the change in
the ratio of Paco2 to HCO3− concentration, and not in
their absolute values, determines the direction of change
in H+ concentration and thus in pH. The status of the
plasma bicarbonate-carbonic acid buffer system can be
monitored easily by serial blood gas measurements, making understanding of this buffer system important in
clinical care.
The physiologic regulation system that responds more
slowly to changes in acid-base balance is the renal system. There must be a long-term balance between net acid
increase caused by intake and production and net acid
decrease caused by excretion and metabolism. Although
infant formula and protein-containing intravenous fluids
have small amounts of preformed acid, most of the daily
acid load is derived from metabolism. A large amount of
the acid produced is in the form of the volatile H2CO3
that can be excreted in the lungs. Nonvolatile or fixed
acids are also produced, which must be excreted through
the kidneys. Nonvolatile acids normally are sulfuric acid
produced in the metabolism of the amino acids methionine and cysteine as well as smaller contributions from
phosphoric acid, lactic acid, hydrochloric acid, and incompletely oxidized organic acids. In addition to the excretion of nonvolatile acids, however, the kidneys have a role
in long-term acid-base regulation by controlling renal
HCO3− excretion.
384
PART VIII Care of the High-Risk Infant
Two regions of the kidney act to achieve urinary acidification—the proximal tubule and the collecting tubule.
The proximal tubule acidifies the urine by two mechanisms. The first mechanism is by the reabsorption of any
HCO3− already present in the blood that is being constantly
filtered through the glomeruli. The proximal tubule reabsorbs 60% to 80% of all filtered HCO3− and performs this
role through an exchange of Na+ for H+ across the luminal membrane of the proximal tubular cells via the Na+/
H+ exchanger. The excreted H+ combines with filtered
HCO3−, producing H2CO3 through the activity of carbonic anhydrase in the cellular brush border. The H2CO3
is then quickly converted to CO2, which crosses into the
tubular cell, where HCO3− is regenerated and reabsorbed
back into the blood stream, probably in exchange for chloride (Cl–). The regenerated H+ ion reenters the cycle at the
Na+/H+ exchanger.
The second mechanism by which the proximal tubule
acidifies urine is by the production of ammonia (NH3).
Inside the tubular cell, NH3 is produced by the deamination of glutamine. The NH3 is secreted into the tubular
lumen, where it combines with and traps free H+ to form
ammonium (NH4+).
The remaining urinary acidification occurs mostly in the
collecting tubule. H+ secretion in this region of the kidney is sufficient to combine with or titrate any remaining
filtered HCO3− or any filtered anions, such as phosphate
and sulfate. Hydrogenated phosphate and sulfate anions
produce the titratable acid of the urine. The collecting
tubule also takes up NH3 from the medullary interstitium
and secretes it into the urine, where again it can combine
with and trap H+ as NH4+. This urinary NH4+ can act as
a cation and be excreted with urinary anions such as Cl−,
PO4−, and SO4−, thereby preventing loss of cations such as
Na+, Ca++, and K+. Total acid secretion in the kidney can
be represented by
Titratable acid + NH4 + − HCO3−
and under normal conditions should equal the net production of acid from diet and metabolism that is not excreted
in the form of CO2 through the lungs.
In adults, the steady state for renal compensation for
respiratory alkalosis is reached within 1 to 2 days and
for respiratory acidosis within 3 to 5 days. Newborns
are able to compensate for acidemia through the previously described renal mechanisms, although the renal
response to acid loads is limited, especially in premature
infants born before 34 weeks’ gestation. Reabsorption of
HCO3− in the proximal tubule and distal tubular acidification are also decreased, with a fairly rapid gestational age-
dependent maturation of these functions after birth (Jones
and Chesney, 1992).
To accomplish the tight regulation of pH necessary
for survival, H+ ions generated in the form of the volatile acid H2CO3 are excreted by the lungs as CO2. H+
ions generated in the form of nonvolatile acids are buffered rapidly by extracellular HCO3− and more slowly
by intracellular buffers. HCO3− is then replenished by
the kidneys via the reabsorption of much of the filtered
HCO3− and by the excretion of H+ in the urine as NH4+
and titratable acids.
DISTURBANCES OF ACID-BASE BALANCE
IN THE NEWBORN
General Principles
The evaluation of the acid-base status in a newborn is one
of the most common laboratory assessments made in the
neonatal intensive care unit. The status of this system can
be monitored with blood gas measurements and should be
the starting point for the evaluation of any acid-base disorder. In the blood gas measurement, the pH and Paco2
levels are directly measured; from these, the HCO3− level
and base excess or deficit are calculated.
The whole blood buffer base, defined as the sum of the
HCO3− and non-HCO3− buffer systems, is another important blood gas value used in evaluating acid-base disturbances. The difference between the observed whole blood
buffer base of any blood gas sample and the expected normal buffer base of that sample is called the base excess or
base deficit. The base excess and base deficit give an accurate measure of the amount of strong acid or base, respectively, that would be needed to titrate the pH back to
normal once the respiratory contribution of the acid-base
disturbance is also corrected. For example, a base excess
of 10 mEq/L indicates that there is an additional 10 mEq
of base per liter (or loss of 10 mEq of H+ per liter) that is
contributing to the acid-base abnormality. Conversely, a
base deficit of 10 mEq/L indicates there is relatively more
acid (or less base) in the ECF than expected after accounting for the effect of Pco2 on pH.
Acid-base disorders are classified according to their
cause as being either metabolic or respiratory. Metabolic
acidosis occurs as a result of the accumulation of increased
amounts of nonvolatile acid or decreased amounts of
HCO3− in the ECF. Metabolic alkalosis occurs as a result
of increased amounts of HCO3− in the ECF. Respiratory
acidosis is caused by hypoventilation and decreased excretion of volatile acid (CO2), whereas respiratory alkalosis
is caused by hyperventilation and increased excretion of
volatile acid (CO2).
Acid-base disorders are also classified according to the
number of conditions causing the disorder. When only
one primary acid-base abnormality and its compensatory
mechanisms occur, the disorder is classified as a simple
acid-base disorder. When a combination of simple acid-base
disturbances occurs, the patient has a mixed (or complex)
acid-base disorder. Because secondary physiologic regulatory mechanisms often compensate for the alteration in
pH caused by primary disturbances, it is sometimes difficult to differentiate simple from mixed disorders or even
a simple disorder from its resulting compensation. One
important principle that allows the determination of primary acid-base disturbance is that the compensatory regulatory mechanisms do not completely normalize the pH.
Nomograms, such as the one shown in Figure 31-6,
can help in the diagnosis of the primary disturbance. The
nomogram describes the 95% confidence limits of the
expected compensatory response to a primary abnormality in either Paco2 or HCO3−. Table 31-5 summarizes
the expected respiratory and metabolic compensatory
mechanisms for primary acid-base disorders (Brewer,
1990). If the compensation in a given patient differs from
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
that predicted in Figure 31-6 or Table 31-5, the patient
either has not had enough time to compensate for a simple
acid-base disturbance or has a mixed acid-base disorder.
Furthermore, the complete correction of an acid-base disturbance occurs only when the underlying process responsible for the abnormality has been treated effectively.
For identification of the primary disturbance, the analysis of blood gas values must be considered in light of the
patient’s history and physical findings and with an understanding of expected compensatory responses. Further
laboratory evaluation is indicated if the problem is not
immediately obvious or if the response to therapy is not
as expected. The evaluation of the acid-base disturbance
should always involve efforts to determine the underlying cause of the disturbance, because adequate treatment
requires correction of the underlying disorder, if possible.
Transitional Physiology after Birth
Arterial Plasma Bicarbonate (mmol/Liter)
As part of a discussion of normal physiology, it is important to understand the in utero environment just before
delivery of the newborn and its effects on neonatal
60
56
52
48
44
40
36
32
28
24
20
16
12
8
4
0
100 90
120
110
80
70
60
50
40
35
Metabolic
Alkalosis
Chronic
Respiratory
Acidosis
25
7.2
Metabolic Acidosis
Metabolic acidosis is a common problem, particularly in
the critically ill newborn. Metabolic acidosis occurs when
the drop in pH is caused by the accumulation of acid other
than H2CO3 by the ECF, resulting in loss of available
HCO3−, or by the direct loss of HCO3− from body fluids. Patients who have metabolic acidosis are divided into
those with an elevated anion gap and those with a normal
anion gap.
The anion gap reflects the unaccounted acidic anions
and certain cations in the ECF. The unmeasured anions
normally include the serum proteins, phosphates, sulfates,
and organic acids, whereas the unaccounted cations are the
serum potassium, calcium, and magnesium. Thus, in clinical practice, the anion gap is estimated using the formula
−
Anion gap =[Na + ] serum − ( [Cl ] serum +[HCO3− ] serum )
Normal
15
Acute
Respiratory
Alkalosis
7.3
10
Chronic
Resp.
Alkalosis
Metabolic
Acidosis
7.1
acid-base analysis shortly after birth. Hyperventilation of
pregnancy is a known phenomenon with corresponding
maternal PCO2 levels of approximately 31 to 34 mm Hg
(Thorp and Rushing, 1999). This relative respiratory
alkalosis in the mother is compensated for by a corresponding metabolic acidosis in the mother and, therefore,
fetus. As a result, umbilical arterial blood gases have a
normal pH range of 7.20 to 7.28 with a corresponding base deficit ranging from 2.7 ± 2.8 mEq/L (SD) to
8.3 ± 4.0 mEq/L (SD) (Riley and Johnson, 1993; Sykes
et al, 1982). In other words, a mild metabolic acidosis in the
newborn shortly after birth can be expected and explained
by normal physiology.
20
Acute
Respiratory
Acidosis
7
30
7.4
7.5
PCO (mm Hg)
2
7.6
7.7
7.8
Arterial Blood pH
FIGURE 31-6 Acid-base nomogram illustrating the 95% confidence
limits for compensatory responses to primary acid-base disorders. (From
Cogan MG, Rector FC Jr: Acid-base disorders. In Brenner BM, Rector FC Jr,
editors: The kidney, Philadelphia, 1986, WB Saunders.)
The normal range of the serum anion gap in newborns
is 8 to 16 mEq/L, with slightly higher values in very premature newborns. Accumulation of strong acids because of
increased intake, increased production, or decreased excretion results in an increased anion gap acidosis, whereas loss
of HCO3− or accumulation of H+ results in a normal anion
gap acidosis. A decrease in serum potassium, calcium, and
magnesium concentrations, an increase in serum protein
concentration, or a falsely elevated serum sodium concentration can also result in an increased anion gap in the
absence of metabolic acidosis. In clinical practice, although
TABLE 31-5 Expected Compensatory Mechanisms Operating in Primary Acid-Base Disorders
Acid-Base Disorder
Primary Event
Compensation
Rate of Compensation
↓ Pco2
For 1 mEq/L ↓ [HCO3–], Pco2 ↓ by 1-1.5 mm Hg
Metabolic acidosis
Normal anion gap
↓ [HCO3–]
For 1 mEq/L ↓ [HCO3–], Pco2 ↓ by 1-1.5 mm Hg
↑ Acid production
↑ Acid intake
↓ Pco2
Respiratory acidosis
Acute (<12-24 h)
Chronic (3-5 days)
↑ Pco2
↑ Pco2
↑ [HCO3
↑ [HCO3–]
For 10 mm Hg ↑ Pco2, [HCO3–] ↑ by 1 mEq/L
For 10 mm Hg ↑ Pco2, [HCO3–] ↑ by 4 mEq/L
Respiratory alkalosis
Acute (<12 h)
Chronic (1-2 days)
↓ Pco2
↓ Pco2
↓ [HCO3–]
↓ [HCO3–]
For 10 mm Hg ↑ Pco2, [HCO3–] ↑ by 1-3 mEq/L
For 10 mm Hg ↓ Pco2, [HCO3–] ↓ by 2-5 mEq/L
Increased anion gap
Metabolic alkalosis
385
↑ [HCO3–]
↑ Pco2
–]
Modified from Brewer ED: Disorders of acid-base balance, Pediatr Clin North Am 37:430-447, 1990.
For 1 mEq/L ↑ [HCO3–], Pco2 ↑ by 0.5-1 mm Hg
386
PART VIII Care of the High-Risk Infant
BOX 31-3 C
ommon Causes of Metabolic
Acidosis
INCREASED ANION GAP
Lactic acidosis caused by tissue hypoxia
ll
Asphyxia, hypothermia, shock
ll Sepsis, respiratory distress syndrome
ll Inborn errors of metabolism
ll Congenital lactic acidosis
ll Organic acidosis
ll Renal failure
ll Late metabolic acidosis
ll Toxins (e.g., benzyl alcohol)
ll
NORMAL ANION GAP
Renal bicarbonate loss
ll Bicarbonate wasting caused by immaturity
ll Renal tubular acidosis
ll Carbonic anhydrase inhibitors
ll Gastrointestinal bicarbonate loss
ll Small bowel drainage: ileostomy, fistula
ll Diarrhea
ll Extracellular volume expansion with bicarbonate dilution
ll Aldosterone deficiency
ll Excessive chloride in intravenous fluids
ll
BOX 31-4 Inborn Errors of Metabolism
Associated With Metabolic
Acidosis
ll
ll
ll
ll
Primary lactic acidosis
Organic acidemias
Pyruvate carboxylase deficiency
Pyruvate hydroxylase deficiency
ll
ll
ll
Galactosemia
Hereditary fructose intolerance
Type I glycogen storage
disease
a serum anion gap value greater than 16 mEq/L is highly
predictive of the presence of lactic acidosis and a value less
than 8 mEq/L is highly predictive of the absence of lactic
acidosis, an anion gap value between 8 and 16 mEq/L cannot be used to differentiate between lactic and nonlactic
acidosis in the critically ill newborn (Lorenz et al, 1999).
Therefore if the anion gap is within this high normal range
and lactic acidosis is suggested, measuring serum lactate is
indicated.
An increased anion gap metabolic acidosis in the newborn is most commonly caused by lactic acidosis secondary
to tissue hypoxia, as seen in asphyxia, hypothermia, severe
respiratory distress, sepsis, and other severe neonatal illnesses. Other important but much less common causes of
an increased anion gap metabolic acidosis in the neonatal
period are inborn errors of metabolism, renal failure, and
intake of toxins (Box 31-3). Box 31-4 lists inborn errors of
metabolism that can manifest as increased anion gap metabolic acidosis in the newborn period.
In the syndrome of late metabolic acidosis of prematurity, first described in the 1960s, otherwise healthy premature infants at several weeks of age demonstrated mild
to moderate increased anion gap acidosis and decreased
growth. All the infants were receiving high-protein
cow’s milk formula, and they demonstrated higher net
acid excretion compared with controls. This type of late
metabolic acidosis is rarely seen, probably because of the
use of special premature infant formulas and changes in
regular formulas with decreased casein-to-whey ratios and
lower fixed acid loads.
A normal anion gap metabolic acidosis most commonly
occurs in the newborn as a result of HCO3− loss from the
extracellular space through the kidneys or the gastrointestinal tract. Hyperchloremia develops with the HCO3− loss
because a proportionate rise in serum chloride concentration must occur to maintain the ionic balance or to correct
the volume depletion in the extracellular compartment.
The most common cause of normal anion gap metabolic
acidosis in the preterm newborn is a mild, developmentally regulated, proximal renal tubular acidosis with renal
HCO3− wasting. In infants with this disorder, the serum
HCO3− usually stabilizes at 14 to 18 mEq/L in the early
postnatal period. The urinary pH is normal once the
serum HCO3− falls to this level, because the impairment
in proximal tubular HCO3− reabsorption is not associated with an impaired distal tubular acidification of similar
magnitude (Jones and Chesney, 1992). The diagnosis of
this temporary cause of acidosis can be established by the
recurrence of a urinary alkaline pH when serum HCO3−
is raised above the threshold after HCO3− or acetate supplementation. Even term newborns have a lower renal
threshold for HCO3−, with normal plasma HCO3− levels
in the range of 17 to 21 mEq/L. In most infants, plasma
HCO3− increases to adult levels over the first year as the
proximal tubule matures. Other common causes of normal
anion gap metabolic acidosis seen in neonatal intensive
care units are gastrointestinal HCO3− losses, often caused
by increased ileostomy drainage, diuretic treatment with
carbonic anhydrase inhibitors, and dilutional acidosis with
rapid expansion of the extracellular space through the use
of non-HCO3− solutions in the hypovolemic newborn.
The presence of metabolic acidosis in the newborn should
be suggested by the clinical presentation and the history of
predisposing conditions, including perinatal depression,
respiratory distress, blood or volume loss, sepsis, and congenital heart disease associated with poor systemic perfusion or cyanosis. Metabolic acidosis is confirmed by blood
gas measurements. The cause of metabolic acidosis is often
readily discernible from the history and physical examination. Specific laboratory evaluation of electrolytes, renal
function, lactate, and serum and urine amino acids may
be undertaken, depending on the diagnosis that is suggested clinically. Figure 31-7 shows a simple flow diagram
outlining an approach to diagnosis of metabolic acidosis
in the newborn. It is important to remember that infants
might not manifest an increased anion gap in the setting
of lactic acidosis and thus require the direct measurement of lactate when a lactic acidosis is suspected (Lorenz
et al, 1999).
The morbidity and mortality of metabolic acidosis
depend on the underlying pathologic process, the severity of
the acidosis, and the responsiveness of the process to clinical
management. By far, the most important intervention for an
infant with a metabolic acidosis is to identify the pathologic
process contributing to the acidosis and to take measures
to correct it. The administration of base, such as sodium
bicarbonate, as supportive therapy for metabolic acidosis is
unproven in its efficacy (Aschner and Poland, 2008).
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
ANION GAP
Increased (16)
Urinary ketones
Moderate-large
Absent-small
Organic acidemias
Lactic acidosis
· Congenital
· Hypoxia
· Poor tissue
Normal (16)
Blood ammonia
Normal
RTA
· Transient
· Permanent
Increased
Urea cycle
defects
perfusion
FIGURE 31-7 Diagnostic approach of increased anion gap and
normal anion gap metabolic acidosis in the newborn. Infants with
lactic acidosis may not have an increased anion gap and lactate should be
measured directly if suspected by the history and physical examination.
RTA, Renal tubular acidosis. (Adapted from Lorenz JM, Kleinman LI,
Markarian K, et al: Serum anion gap in the differential diagnosis of metabolic
acidosis in critically ill newborns, J Pediatr 135:751-755, 1999.)
The use of base administration is supported by findings
on the beneficial cardiovascular effects of sodium bicarbonate in preterm newborns with an arterial pH of less than
7.25 and term newborns with an arterial pH of less than
7.30 (Fanconi et al, 1993). The use of sodium bicarbonate
in this study was associated with an increase in myocardial
contractility and a reduction in afterload, albeit transient.
However, there is concern for harm associated with the
administration of base, including increased mortality and
intraventricular hemorrhage (Papile et al, 1978; Simmons
et al, 1974; Usher, 1967), increased cerebral blood volume
regardless of rate of administration (van Alfen-van der
Velden et al, 2006), and decreased intracellular pH with
cellular injury (Lipshultz et al, 2003).
If therapy with base is warranted, the clinician has three
options: sodium bicarbonate, sodium (or potassium) acetate, and tromethamine. Sodium bicarbonate is the most
widely used buffer in the treatment of metabolic acidosis
in the neonatal period. Bicarbonate should not be given if
ventilation is inadequate, because its administration results
in an increase in Paco2 with no improvement in pH and an
increase in intracellular acidosis. Therefore sodium bicarbonate should be administered slowly and in diluted form
only to newborns with documented metabolic acidosis and
adequate alveolar ventilation. Once a blood gas measurement has been obtained, the dose of sodium bicarbonate
required to fully correct the pH can be estimated with the
use of the following formula:
Dose of NaHCO3 (mEq) = Base deficit (mEq/L) ×
Body weight (kg) × 0.3
Sodium bicarbonate is thought to be confined mostly
to the ECF compartment. Although there are controversies regarding the actual bicarbonate space in humans,
the 30% of total body weight in the formula represents
its estimated volume of distribution in the neonate. Most
clinicians would use half of the calculated total correction dose for initial therapy to avoid overcorrection of
387
metabolic acidosis. Subsequent doses of sodium bicarbonate are then based on the results of additional blood gas
measurements.
When clinicians are faced with a chronic metabolic
acidosis caused by a prematurity-related proximal renal
tubular acidosis with bicarbonate wasting, many choose
to replace these losses over time. In this instance, either
sodium or potassium acetate can be used as an alternative
to sodium bicarbonate. Sodium acetate is a conjugate base
of a weak acid (acetic acid) with a pKb of 9.25. pKb is a
measure of the strength of a base, which depends on its
base dissociation constant. It has been shown in one study
to be an effective alternative to sodium bicarbonate in correcting this type of acid-base abnormality when added to
parenteral nutrition (Peters et al, 1997). The median doses
of acetate used in this randomized controlled trial were
2.6 mmol/kg/day on day 4 of life and 4.1 mmol/kg/day on
day 8 of life. Infants randomized to acetate had an increased
base excess and pH and increased Pco2, and they received
less bicarbonate boluses compared with control infants.
In certain clinical situations, tromethamine can be used
as an alternative buffer to sodium bicarbonate. The theoretical advantages of tromethamine over sodium bicarbonate in the treatment of metabolic acidosis of the newborn
include its more rapid intracellular buffering capability, its
ability to lower Paco2 levels directly, and the lack of an
increase in the sodium load (Schneiderman et al, 1993).
Tromethamine lowers Paco2 by covalently binding H+ and
thus shifting the equilibrium of the reaction
H+ + HCO3− ↔ H2 CO3 ↔ H2 O + CO2
to the left, resulting in a decrease in CO2 and an increase
in HCO3−. Because the end-product (chelated tromethamine) is a cation that is excreted by the kidneys, oliguria is
a relative contraindication to the repeated use of this buffer. Tromethamine administration also has been associated with the development of acute respiratory depression,
most likely secondary to an abrupt decrease in Paco2 levels
as well as from rapid intracellular correction of acidosis in
the cells of the respiratory center (Robertson, 1970). Furthermore, because hypocapnia is associated with decreases
in brain blood flow and a higher incidence of white matter
damage, especially in the immature preterm neonate, close
monitoring of Paco2 is of paramount importance when
tromethamine is being used. Finally, when large doses of
tromethamine are administered, hyponatremia (Seri et al,
1998b), hypoglycemia, hyperkalemia, an increase in hemoglobin oxygen affinity, and diuresis followed by oliguria
can occur. Because the tromethamine solution is hyperosmolar, and because rapid infusion of tromethamine
can also lower blood pressure and intracranial pressure
(Duthie et al, 1994), slow infusion is recommended. The
suggested initial dose is 1 to 2 mEq/kg or 3.5 to 6 mL/kg
given intravenously using the 0.3 M solution, with the
rate of administration not exceeding 1 mL/kg/min. Once
a blood gas measurement has been obtained, the dose of
tromethamine required to raise the pH can be estimated
using the formula
Dose of tromethamine (mL) = Base deficit (mEq/L) ×
Body weight (kg)
388
PART VIII Care of the High-Risk Infant
Finally, during the correction of metabolic acidosis, particular attention should be paid to ensuring an appropriate potassium balance. Because potassium moves from the
intracellular to the extracellular space in exchange for H+
when acidosis occurs, the presence of a total body potassium deficit might not be appreciated during metabolic
acidosis. Hypokalemia may become evident only as the pH
increases and potassium returns to the intracellular space.
Furthermore, intracellular acidosis cannot be completely
corrected until the potassium stores are restored. Therefore close monitoring of serum electrolytes and potassium
supplementation are important during the correction of
metabolic acidosis in the sick newborn.
Respiratory Acidosis
Respiratory acidosis occurs when a primary increase
in Paco2 develops secondary to impairments in alveolar ventilation that result in an arterial pH of less than
7.35. Primary respiratory acidosis is a common problem
in newborns, and causes include hyaline membrane disease, pneumonia owing to infection or aspiration, patent
ductus arteriosus with pulmonary edema, chronic lung
disease, pleural effusion, pneumothorax, and pulmonary
hypoplasia. The initial increase in Paco2 is buffered by the
non-HCO3− intracellular buffers without noticeable renal
compensation for at least 12 to 24 hours (see Table 31-5).
Renal metabolic compensation reaches its maximum levels
within 3 to 5 days, and its effectiveness in the newborn is
influenced mainly by the functional maturity of proximal
tubular HCO3− transport.
Management of respiratory acidosis is directed toward
improving alveolar ventilation and treating the underlying disorder. For sick newborns, adequate ventilation must
often be provided by mechanical ventilation. In severe
respiratory acidosis, tromethamine can be used to raise
pH, because it lowers CO2 levels. Tromethamine, however, produces only a transient decrease in Paco2, and toxic
doses would quickly be reached if it were used to buffer all
the CO2 produced by metabolism over a sustained period.
Therefore tromethamine should be used only as a temporizing measure in severe respiratory acidosis until alveolar
ventilation can be improved.
Metabolic Alkalosis
Metabolic alkalosis is characterized by a primary increase
in the extracellular HCO3− concentration sufficient to
raise the arterial pH above 7.45. In the newborn, metabolic alkalosis occurs when there is a loss of H+, a gain
of HCO3−, or a depletion of the extracellular volume with
the loss of more chloride than HCO3−. It is important to
understand that metabolic alkalosis generated by any of
these mechanisms can be maintained only when factors
limiting the renal excretion of HCO3− are also present.
Metabolic alkalosis can result from a loss of H+ from the
body, from either the gastrointestinal tract or the kidneys,
that induces an equivalent rise in the extracellular HCO3−
concentration. The most common causes of this type of
metabolic alkalosis in the newborn period are continuous
nasogastric aspiration, persistent vomiting, and diuretic
treatment. Less common causes of H+ losses are congenital
chloride-wasting diarrhea, certain forms of congenital
adrenal hyperplasia, hyperaldosteronism, posthypercapnia, and Bartter syndrome.
Metabolic alkalosis can also result from a gain of
HCO3−, such as occurs during the administration of buffer
solutions to the newborn. In the past, a metabolic alkalosis
was intentionally created when sodium bicarbonate or tromethamine was used to maintain an alkaline pH to decrease
pulmonary vasoreactivity in infants with persistent pulmonary hypertension, a practice not recommended anymore.
Currently, iatrogenically produced metabolic alkalosis
is primarily unintentional and due to chronic excessive
administration of HCO3−, lactate, citrate, or acetate in
intravenous fluids and blood products. Because excretion
of HCO3− is normally not limited in the newborn, metabolic alkalosis resulting from HCO3− gain alone should
rapidly resolve after administration of HCO3− is discontinued. However, if the alkalosis is severe and urine output
is limited, inhibition of the carbonic anhydrase enzyme by
the administration of acetazolamide may enhance elimination of HCO3−.
Metabolic alkalosis can also result from a loss of
ECF containing disproportionately more chloride than
HCO3−—the so-called contraction alkalosis. During the
diuretic phase of normal postnatal adaptation, preterm and
term newborns retain relatively more HCO3− than chloride (Ramiro-Tolentino et al, 1996). The obvious clinical
benefits of allowing this physiologic extracellular volume
contraction to occur, especially in the critically ill newborn, clearly outweigh the clinical importance of a mild
contraction alkalosis that develops after recovery. No specific treatment is needed in such cases, because with the
stabilization of the extracellular volume and renal function
after recovery, acid-base balance rapidly returns to normal. Contraction alkalosis due to other causes, however,
may require treatment.
For metabolic alkalosis to persist, factors limiting the
renal excretion of HCO3− must be present. The kidneys
are usually effective in excreting excess HCO3−, but this
ability can be limited under certain conditions, such as
decreased glomerular filtration rate, increased aldosterone production, and the more common clinical situation of volume contraction–triggered metabolic alkalosis
with potassium deficiency. In the last condition, there is
a direct stimulation of Na+ reabsorption coupled with
H+ loss in the proximal tubule, and an indirect stimulation of H+ loss in the distal nephron by the increased
activity of the renin-angiotensin-aldosterone system.
Contraction alkalosis responds to administration of
saline to replace the intravascular volume in conjunction
with additional potassium supplementation to account
for renal potassium wasting. In the other disorders,
however, the primary problem of reduced glomerular
filtration rate or elevated aldosterone must be treated
for the alkalosis to resolve.
One of the most commonly encountered clinical scenarios of chronic metabolic alkalosis actually occurs
in the form of a mixed acid-base disorder in a preterm
infant with chronic lung disease on long-term diuretic
treatment. Such a newborn initially has a chronic respiratory acidosis that is partially compensated for by renal
HCO3− retention. Prolonged or aggressive use of diuretics
CHAPTER 31 Acid-Base, Fluid, and Electrolyte Management
can lead to total-body potassium depletion and contraction of the extracellular volume, thus exacerbating the
metabolic alkalosis. By stimulating proximal tubular Na+
reabsorption and thus H+ loss, distal tubular H+ secretion,
and renal ammonium production, the diuretic-induced
hypokalemia contributes to the severity and maintenance
of the metabolic alkalosis. Furthermore, metabolic alkalosis per se worsens hypokalemia, because potassium moves
intracellularly to replace hydrogen as the latter shifts into
the extracellular space. Although the serum potassium
concentration may be decreased, the serum levels in the
newborn do not accurately reflect the extent of total-body
potassium deficit because potassium is primarily an intracellular ion, with approximately 98% of the total body
potassium being in the intracellular compartment. In
addition, the condition is often accompanied by marked
hypochloremia and hyponatremia. Hyponatremia occurs
in part because sodium shifts into the intracellular space
to compensate for the depleted intracellular potassium.
If the alkalosis is severe, alkalemia (pH >7.45) can supervene and result in hypoventilation. In this situation, potassium chloride, and not sodium chloride supplementation,
reverses hyponatremia and hypochloremia, corrects hypokalemia and metabolic alkalosis, and increases the effectiveness of diuretic therapy. Because chloride deficiency
is the predominant cause of the increased pH, ammonium
chloride or arginine chloride also corrects the alkalosis.
These agents do not affect the other electrolyte imbalances such as the hypokalemia, so they should not be the
only therapy given.
It is important to keep ahead of the potassium losses in
infants receiving long-term diuretic therapy, rather than
to attempt to replace potassium after intracellular depletion has occurred. Because the rate of potassium repletion
is limited by the rate at which potassium moves intracellularly, correction of total body potassium deficits can
require days to weeks. In addition, there is also a risk of
acute hyperkalemia if serum potassium levels are driven
too high during repletion, particularly in newborns in
whom an acute respiratory deterioration may occur, with
worsened respiratory acidosis and the subsequent movement of potassium from the intracellular to the extracellular space. The routine use of potassium chloride
supplementation and close monitoring of serum sodium,
chloride, and potassium levels are therefore recommended
during long-term diuretic therapy to prevent these common iatrogenic problems.
389
Respiratory Alkalosis
When a primary decrease in Paco2 results in an increase in
the arterial pH beyond 7.45, respiratory alkalosis develops.
The initial hypocapnia is acutely titrated by the intracellular buffers, and metabolic compensation by the kidneys
returns pH toward normal within 1 to 2 days (see Table
31-5). Interestingly, respiratory alkalosis is the only simple
acid-base disorder in which, at least in adults, the pH can
completely be normalized by the compensatory mechanisms (Brewer, 1990). The cause of respiratory alkalosis
is hyperventilation, which in the spontaneously breathing
newborn is most often caused by fever, sepsis, retained
fetal lung fluid, mild aspiration pneumonia, or central
nervous system disorders. In the neonatal intensive care
unit, the most common cause of respiratory alkalosis is
iatrogenic secondary to hyperventilation of the intubated
newborn. Because findings suggest an association between
hypocapnia and the development of periventricular leukomalacia (Okumura et al, 2001; Wiswell et al, 1996)
and chronic lung disease (Garland et al, 1995) in ventilated preterm infants, avoidance of hyperventilation during resuscitation and mechanical ventilation is of utmost
importance in the management of sick preterm newborns.
The treatment of neonatal respiratory alkalosis consists of
the specific management of the underlying process causing
hyperventilation.
SUGGESTED READINGS
Aschner JL, Poland RL: Sodium bicarbonate: basically useless therapy, Pediatrics
122:831, 2008.
Avner ED: Clinical disorders of water metabolism: hyponatremia and hypernatremia, Pediatr Ann 24:23, 1995.
Brewer ED: Disorders of acid-base balance, Pediatr Clin North Am 37:430, 1990.
El-Dahr SS, Chevalier RL: Special needs of the newborn infant in fluid therapy,
Pediatr Clin North Am 37:323, 1990.
Hartnoll G: Basic principles and practical steps in the management of fluid balance
in the newborn, Semin Neonatol 8:307, 2003.
Lorenz J: Fluid and electrolyte therapy in the very low-birthweight neonate,
Neoreviews 9:e02, 2008.
Modi N: Management of fluid balance in the very immature neonate, Arch Dus
Child Fetal Neonatal Ed 89:F108, 2004.
Oh W, Poindexter BB, Perritt R, et al: Association between fluid intake and weight
loss during the first ten days of life and the risk of bronchopulmonary dysplasia
in extremely low birthweight infants, J Pediatr 147:786, 2005.
Sedin G: Fluid management in the extremely preterm infant, In Hansen TN,
McIntosh N, editors: Current topics in neonatology, London, 1995, WB Saunders,
pp 50-66.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
32
Care of the Extremely Low-Birthweight
Infant
Eric C. Eichenwald
In the past two decades, the field of neonatology has
experienced significant progress in medical care and
improvement in overall patient survival. Advancement in
technology, greater use of prenatal glucocorticoids and
neonatal surfactant replacement therapy, better regionalization of perinatal and high-risk neonatal care, and a
more comprehensive understanding of the physiology
of the immature infant have all contributed to dramatic
increases in survival of very preterm infants. Care of premature infants with birthweights between 1000 and 1500 g
has become almost routine in most neonatal intensive care
units (NICUs) in the United States.
The most recent challenge in neonatology is the care of
extremely low-birthweight (ELBW) infants (birthweight
<1000 g), sometimes referred to colloquially as micropremies. These infants present one of the greatest medical and
ethical challenges to the field. Although they represent a
small percentage of overall births and NICU admissions,
ELBW infants are often the most critically ill and at the
highest risk for mortality and long-term morbidity of any
NICU patient. They also contribute disproportionately to
overall hospital days and consume a large percentage of
NICU personnel time, effort, and costs of care. Care of
these infants is in constant evolution, as a result of new
discoveries in both basic and clinical research as well as to
growing clinical experience. This chapter will review some
of the special challenges in and practical aspects of the
management of the ELBW infant. The reader is referred
to specific chapters throughout the text for a more comprehensive review of specific problems and conditions.
Several studies have shown increased survival in the
smallest and most premature infants over the past decade
(Figures 32-1 and 32-2) (Fanaroff et al, 2007; Horbar et al,
2002; Lemons et al, 2001). See Chapter 1 for a complete
discussion of these changes.
Improved survival has not been accompanied by a change
in the incidence of several major morbidities among survivors (Tables 32-1 and 32-2). Little to no change in the
frequency of severe intracranial hemorrhage, periventricular white matter injury, necrotizing enterocolitis, or
bronchopulmonary dysplasia was observed in the Eunice
Kennedy Shriver National Institute of Child Health and
Human Development cohort of infants born in institutions
participating in the Neonatal Research Network in 19951996 compared with and 1997-2002 (Fanaroff et al, 2007).
Although the Vermont–Oxford NICUs reported a reduction in the incidence of severe intracranial hemorrhage
in the early part of the 1990s, the incidence has remained
static into the late 1990s (Horbar et al, 2002). With the
current trends in survival and in hospital morbidity, the
absolute number of extremely premature infants who survive to NICU discharge and are diagnosed with a major
morbidity in the neonatal period is increasing. A significant
percentage of these infants continues to suffer from neurodevelopmental and neurosensory disability into childhood
(Hack et al, 2005; Marlow et al, 2005; Wood et al, 2000).
100
EPIDEMIOLOGY
The percentage of babies born preterm in the United
States has risen slowly over the past two decades (Behrman and Stith Butler, 2007). In the year 2007, preterm
births (<37 weeks’ gestation) accounted for 12.7% of all
births, with approximately 1% of births occurring before
28 weeks’ gestation. There is a significant racial disparity
in the incidence of extreme preterm birth, with the African American ELBW birth rate being nearly double that of
the Hispanic and non-Hispanic white populations. Associated with this increase in frequency of preterm births is the
greater availability of assisted reproductive technologies.
These technologies result in a higher incidence of LBW
infants, because of the higher frequency of multiple gestations (Schieve et al, 2002). Twins, triplets, and higher-order
multiple gestations currently represent almost a quarter all
LBW deliveries in the United States (Martin et al, 2002)
and contribute to the ELBW population. Multiple gestations add to the potential morbidity of extremely premature
birth because of a higher frequency of intrauterine growth
restriction and other medical complications of pregnancy.
390
Mortality %
80
1991
1996
1997-2002
2008
60
40
20
0
501-750
751-1000
1001-1250
1251-1500
Birthweight (grams)
FIGURE 32-1 Mortality in very low birthweight infants receiving care
in the National Institute of Child Health and Human Development
Eunice Kennedy Shriver Neonatal Research Network Centers from
1991 to 2002 and the Vermont-Oxford Network in 2008. (From Eichenwald EC, Stark AR: Management and outcomes of very low birth weight,
N Engl J Med 358:1700-1711, 2008. Data from Lemons J, Bauer C, Oh
W, et al: Very low birth weight outcomes of the National Institute of Child
Health and Human Development Neonatal Research Network, January 1995
through December 1996, Pediatrics 107:1-8, 211, 2001; Fanaroff AA, Stoll
BJ, Wright LL, et al: Trends in neonatal morbidity and mortality for very low
birthweight infants, Am J Obstet Gynecol 196:147.e1-147.e8, 2007; and
Horbar JD, Carpenter J, Kenny M, et al, editors: Vermont Network 2008
very low birth weight database summary, Burlington, Vt, 2009, Vermont
Oxford Network.)
391
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
PERINATAL MANAGEMENT
Extremely premature infants born in perinatal centers for
high-risk infants, especially those with a high volume of
such infants, have better short-term outcomes than infants
transferred to such centers after birth (Arad et al, 1999;
Bartels et al, 2006; Chien et al, 2001; Cifuentes et al, 2002;
Phibbs et al, 2007; Shah et al, 2005; Towers et al, 2000).
Therefore, if clinically feasible, the pregnant woman who
seems likely to deliver an extremely premature infant
should be transferred to a high-risk perinatal center for
the expertise in both obstetric and neonatal management.
Upon arrival, the expectant mother should be evaluated
for factors that may have predisposed to preterm labor
and assessed for the status of the fetal membranes and
the presence or absence of chorioamnionitis. In addition,
best obstetric estimate of gestational age (by date of last
100%
menstrual period and early ultrasonographic dating, if
available), ultrasonographic assessment of fetal size and
position, and the presence of other medical or obstetric
complications (preeclampsia, placenta previa, abruptio placentae) should be documented. Specimens for rectovaginal cultures to detect the presence of group B streptococci
should also be obtained on admission (Schrag et al, 2002),
and treatment with penicillin or ampicillin (or vancomycin
for the patient with a severe penicillin allergy) should be
initiated until culture results are available. There is widespread agreement that prenatal glucocorticoids should be
offered to any woman in whom delivery at 24 to 34 weeks’
gestation threatens; treatment at earlier gestational ages is
controversial and of unclear benefit. Although unlikely to
arrest labor for an extended period, tocolytic agents should
be considered for women with preterm uterine contractions without evidence of chorioamnionitis.
Premature rupture of the fetal membranes (PROM)
occurs in 30% to 40% of women who deliver prematurely
(Mazor et al, 1998). If PROM is diagnosed without evidence of chorioamnionitis, consideration should be given
Mortality %
80%
TABLE 32-1 Survival and Selected Morbidities
60%
for Very Low Birthweight (501-1500 g) Infants Born
in NICHD Neonatal Research Network Sites
(1995-1996 Compared with 1997-2002)
40%
0%
1995-1996*
n = 4438%
1997-2002†
n = 18,153%
Survival without morbidity
84
85
Bronchopulmonary dysplasia
70
70
Home in supplemental
oxygen
23
22
Necrotizing enterocolitis
15
11
7
7
12
12
5
3
Survival
20%
501-750g
751-1000g 1001-1250g 1251-1500g 501-1500g
Intact Survival
Survived with Morbidity
Died
FIGURE 32-2 Proportion of very low birthweight infants who
died, and the proportion who survived with short-term complications
(bronchopulmonary dysplasia, severe intraventricular hemorrhage,
necrotizing enterocolitis, or a combination of these disorders, or with
no complications from 1997 to 2002 at the Eunice Kennedy Shriver
National Institute of Child Health and Human Development Neonatal
Research Network sites. (From Eichenwald EC, Stark AR: Management
and outcomes of very low birth weight, N Engl J Med 358:1700-1711,
2008. Data from Fanaroff AA, Stoll BJ, Wright LL, et al: Trends in neonatal morbidity and mortality for very low birth weight infants, Am J Obstet
Gynecol 196:147.e1-147.e8, 2007.)
Severe intraventricular
hemorrhage
Periventricular white matter
injury
Late-onset sepsis
NICHHD, National Institute of Child Health and Human Development.
*Lemons J, Bauer C, Oh W, et al: Very low birth weight outcomes from the National
Institute of Child Health and Development Neonatal Research Network, January 1995
through December 1996, Pediatrics 107:1-18, 2001.
†Fanaroff AA, Stoll BJ, Wright LL, et al: Trends in neonatal morbidity and mortality
for very low birth weight infants, Am J Obstet Gynecol 196:147, 2007.
TABLE 32-2 Survival With Selected Morbidities for Very Low Birthweight Infants: NICHD Neonatal Research Network, 1997-2002*
Birthweight
501-750 g
n = 4046%
751-1000 g
n = 4266%
1021-1250 g
n = 4557%
1251-1500 g
n = 5284%
Overall survival
55
88
94
96
Overall
65
43
22
11
BPD alone
42
25
11
4
Severe IVH
5
6
5
4
NEC alone
3
3
3
2
10
4
2
<1
Survived With Morbidity
BPD and severe IVH
BPD, Bronchopulmonary dysplasia; IVH, intraventricular hemorrhage.
*Data from Fanaroff AA, Stoll BJ, Wright LL, et al: Trends in neonatal morbidity and mortality for very low birth weight infants, Am J Obstet Gynecol 196:147, 2007.
392
PART VIII Care of the High-Risk Infant
to the use of prophylactic antibiotic therapy (ampicillin or
erythromycin) for the mother. Several studies have shown
that such therapy prolongs the latency period, reduces
the incidence of chorioamnionitis and endometritis, and
improves neonatal outcome (Gibbs and Eschenbach, 1997;
Mazor et al, 1998; Mercer et al, 1997). Tocolytic and antibiotic therapy in the setting of PROM may prolong latency
by 48 to 72 hours in many extremely preterm pregnancies
in which delivery threatens, allowing the administration of
a complete course of glucocorticoids to the mother.
PRENATAL CONSULTATION
If possible, all parents who are at risk for delivery of an
extremely premature infant should meet in consultation
with a neonatologist before the infant’s birth, preferably
jointly with a perinatologist caring for the mother (Finer
and Barrington, 1998). There are several goals of this consultation. First, the neonatologist and perinatologist should
inform the parents about the proposed management of the
pregnancy and delivery of the infant, including a discussion of the advantages of administration of glucocorticoids
to the mother, the possible need for cesarean delivery, and
delivery room care and resuscitation of the baby. Second,
the parents should be informed about the potential risks of
both the extent of prematurity and the proposed therapeutic interventions (Tables 32-3 and 32-4). Third, the neonatologist should investigate the parents’ beliefs and attitudes
about delivery of an extremely premature infant and the
potential for long-term morbidity. This outline for prenatal
consultation is especially important if delivery is expected at
the borderline of viability (23 to 24 weeks’ gestation).
Data are sparse about the process and results of prenatal
consultation with parents of an extremely preterm infant.
Some studies suggest that significant incongruity exists
among the attitudes of obstetricians, nurses, neonatologists,
and parents about active delivery room resuscitation and
treatment of extremely premature infants (Streiner et al,
2001; Zupancic et al, 2002). Reuss and Gordon (1995) have
shown that the obstetric judgment of fetal viability was
associated with an 18-fold increase in survival for infants
with a birthweight less than 750 g, indicating that obstetric
attitudes and decisions influence outcome. Obstetricians
and nurses tend to overestimate, and parents tend to underestimate, mortality and morbidity in premature infants
TABLE 32-4 Incidence of Specific Interventions and Outcomes
of Extremely Low Birthweight Infants (<1000 g)
in the Vermont–Oxford Network, 2008
TABLE 32-3 Major Problems of Extremely Low Birthweight
Birthweight Category
Infants
Short-Term
Problems
Long-Term
Problems
Respiratory distress
syndrome
Air leaks
Bronchopulmonary
dysplasia
Apnea of prematurity
Chronic lung disease
Reactive airway
disease
Feeding intolerance
Necrotizing
enterocolitis
Growth failure
Growth failure
Failure to thrive
Inguinal hernias
Immunologic,
infection
Immune deficiency
Perinatal infection
Nosocomial infection
Respiratory syncytial
virus
Central nervous
system
Intraventricular
hemorrhage
Periventricular white
matter disease
Cerebral palsy
Neurodevelopmental
delay
Hearing loss
Ophthalmologic
Retinopathy of
prematurity
Blindness, retinal
detachment
Myopia
Strabismus
Cardiovascular
Hypotension
Patent ductus
arteriosus
? Hypertension in
adulthood
Pulmonary hypertension
System
Respiratory
Gastrointestinal,
nutritional
Renal
Hematologic
Endocrine
Water, electrolyte
imbalance
Acid-base disturbances
Iatrogenic anemia
Frequent transfusions
Anemia of prematurity
Transient hypothyroxinemia
?Cortisol deficiency
? Impaired glucose
regulation
? Increased insulin
resistance
Outcome
Survival (%)
501 to 750 g
n = 9963
751 to 1000 g
n = 12,341
58
85
Chronic lung disease
at 36 weeks (%)
66
39
Discharge home in
oxygen (%)
38
20
Eye examination performed (%)
66
82
None (%)
25
51
Stages 1-2 disease (%)
47
40
≥Stage 3 disease (%)
27
8
Laser, cryotherapy (% treated)
15
5
Cranial ultrasound
examination
91
95
No intraventricular
hemorrhage (%)
54
69
Grade 1-2 (%)
23
19
≥Grade 3 (%)
23
12
Patent ductus arteriosus (%)
63
53
Indomethacin, ibuprofen
treatment (%)
61
52
Surgical ligation (%)
23
12
Nosocomial infection (%)
39
26
Necrotizing enterocolitis (%)
13
10
Survival without major
morbidity (%)
12
38
Respiratory Morbidity
Retinopathy of Prematurity
Intraventricular Hemorrhage
Data from Horbar JD, Carpenter J, Kenny M, Michelle J, editors: Vermont Oxford
Network 2008 very low birth weight database summary, Burlington, Vt, 2009,
Vermont Oxford Network.
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
(Doron et al, 1998; Haywood et al, 1994). Parents report
being more in favor of intervening regardless of gestational age or condition at birth compared with health care
professionals (Streiner et al, 2001). In a survey by Ballard
et al (2002), the majority of neonatologists responded that
they would respect parents’ wishes about resuscitation of a
borderline viable infant. In practice, when parental preferences about active resuscitation are known before delivery of an extremely preterm infant, neonatologists report
that they would alter their management of the infant in
the delivery room accordingly after assessment of the baby
after birth (Doron et al, 1998), indicating the important
role for prenatal consultation. However, parental attitudes
likely are strongly influenced by physician recommendations about aggressive resuscitation conveyed during the
prenatal consultation. This consultation is further complicated by significant variability in physician’s opinions
about the benefits and burdens of providing intensive care
at the borders of viability (De Leeuw et al, 2000). Physician attitudes may be influenced by fear of litigation for
not actively resuscitating an extremely premature infant
even if parental choices are made clear, as well as local,
regional, and national norms (Partridge et al, 2009).
The earliest gestational age at which resuscitation should
be initiated remains controversial among neonatologists
(Finer et al, 1999), making firm recommendations for the
actual content of prenatal consultation difficult (Kaempf
et al, 2006). Most studies that describe the outcome of
premature infants are based on birthweight rather than
gestational age and thus confound the effects of extremely
preterm birth with those of intrauterine growth restriction, but both parents and physicians are usually faced with
making decisions based on the anticipated gestational age
at delivery. However, the likelihood of survival without
serious sequelae may be influenced by factors other than
gestational age. Using prospectively collected data from
the Eunice Kennedy Shriver National Institute of Child
Health and Human Development Neonatal Research Network on infants born at 22 to 25 weeks’ gestation between
1998 and 2003, Tyson et al (2008) presented an analysis of
some of these factors. In a multivariate analysis, exposure to
antenatal glucocorticoids, female sex, singleton gestation,
and higher birthweight (in 100-g increments) were each
associated with a decrease in the risk of death or of survival
with neurodevelopmental impairment. The reduced risk
was similar to the risk for infants with an additional week
of gestational age. Other data from several studies suggest
that in terms of survival and potential for long-term disability, encouragement of active management is appropriate
for pregnancies expected to deliver at 25 weeks’ gestation
or later (Piecuch et al, 1997; Wood et al, 2000). The risk
of death and severe morbidity is significantly higher before
25 weeks’ gestation (El-Metwally et al, 2002). Mortality
approaches 80% for infants delivered at 23 weeks’ gestation, with the majority of survivors suffering from neurodevelopmental sequelae (Vohr et al, 2000; Wood et al, 2000).
These data should be shared with parents during prenatal
consultation, and appropriate guidance should be given for
decision making about the pregnancy, delivery, and resuscitation of the infant. Some have argued that, because of
the risk of long-term disability associated with extreme
preterm birth and the difficulty of making an informed
393
decision immediately before delivery, parents should be
given the opportunity to have these issues discussed during
routine prenatal care (Harrison, 2008).
GENERAL PRINCIPLES OF CARE
SPECIFIC TO EXTREMELY
LOW-BIRTHWEIGHT INFANTS
FIRST HOURS
The first few hours after admission to the NICU are critical for the ELBW infant, although there are significant
variations in practice among hospitals and practitioners.
Careful adherence to details in the delivery room and during the first few hours after birth is essential to help avoid
some of the immediate and long-term complications of
the ELBW infant. All NICUs should have a consistent
approach to the initial care of these fragile infants in the
delivery room and upon admission to the NICU. A suggested treatment guideline for the first few hours after
birth is presented in Table 32-5, and screening guidelines
for common complications are shown in Table 32-6.
DELIVERY ROOM
At delivery, strict attention to maintenance of body temperature by means of rapid, gentle drying of the infant and
the use of adequate heat sources is paramount to avoid cold
stress. Use of a polyethylene occlusive skin wrap or bag
immediately after delivery may also help to prevent initial
evaporative heat losses (Vohra et al, 1999). Many ELBW
infants need some form of assisted ventilation immediately
after birth. If positive-pressure ventilation is required, it
should be provided with low inspiratory pressure to prevent overdistention of the lungs, which can result in air
leak and other lung injury, and adequate positive endexpiratory pressure (PEEP) to maintain lung volume by
use of a flow-inflating bag, or optimally, a T-piece resuscitator that allows for more consistent delivery of inspiratory pressure and PEEP. Hyperventilation and hyperoxia
also should be avoided; many units use continuous oxygen
saturation monitoring in the delivery room, with supplemental oxygen titrated to maintain oxygen saturations in
the range of 85% to 93%. The use of room air for resuscitation of these infants has been proposed to protect them
from hyperoxia and damage to the lungs by oxygen free
radicals. This issue is under active investigation (Lefkowitz,
2002), but most centers use blended oxygen for initial
resuscitation, with an initial starting Fio2 between 0.4 and
0.6 (see also Chapter 28.)
Considerable practice variation exists in the use and type
of ventilatory support and timing of surfactant administration for the ELBW infant after birth. Some centers
routinely intubate all ELBW infants in the delivery room
for respiratory support and prophylactic surfactant administration (Egberts et al, 1997; Kendig et al, 1998). Soll
(2009) has reported on a metaanalysis of eight randomized
trials of the use of natural surfactant as prophylaxis versus as a rescue strategy for established respiratory distress
syndrome (RDS) in the ELBW infant. Administration of
exogenous surfactant before 15 minutes of age resulted in
a reduction in the rates of neonatal mortality, air leak, and
394
PART VIII Care of the High-Risk Infant
TABLE 32-5 Treatment Guidelines for Initial Management of Extremely Low-Birthweight Infants
Time after Birth
Guideline
Time after Birth
Delivery room
Ensure good thermoregulation
“Gentle” ventilation as required
Avoid hyperventilation and hyperoxia
Administer surfactant (if prophylaxis
approach)
Initiate NCPAP (if early CPAP approach)
First 24 to 48 Hours
NICU admission
Obtain weight measurement
Administer surfactant within first hour
(if rescue approach)
Establish vascular access:
Peripheral intravenous catheter
Umbilical arterial catheter
Umbilical venous catheter (central,
double-lumen)
Start intravenous fluids as soon as possible
with dextrose and amino acid solution
Limit evaporative water losses (humidified
incubator)
Minimize stimulation
Avoid hyperventilation and hyperoxia
Maintain target oxygen saturations
between 88% and 93%
Obtain specimens for complete blood
count with differential, blood culture,
blood glucose measurement
Give antibiotics as indicated
Give parents information about their child
Guideline
Cardiovascular
Monitor blood pressure, give vasopressors
as required
Maintain vigilance for presence of patent
ductus arteriosus
Obtain echocardiogram as indicated
Respiratory
Give additional surfactant doses as indicated
Maintain low tidal volume ventilation
Avoid hyperventilation and hyperoxia
Extubate infant and start on continuous
positive airway pressure when possible
Fluid management
Obtain weight every 12 to 24 hours
Monitor serum electrolyte, blood glucose,
and calcium concentrations every
4 to 8 hours
Limit evaporative water losses
Administer skin care
Hematologic
Obtain second blood count
Administer transfusion support as indicated
Monitor bilirubin, give phototherapy as
indicated
Infection
Consider discontinuing antibiotics if blood
culture results are negative at 48 hours
Nutrition
Start amino acid solution, parenteral
nutrition
Neurologic
Minimize stimulation
Perform screening head ultrasonography
Social
Arrange to meet with family
CPAP, Continuous positive airway pressure; NCPAP, nasal continuous positive airway pressure; NICU, neonatal intensive care unit.
the combined outcome of bronchopulmonary dysplasia
(BPD) or death compared with a selective rescue approach.
However, no trials comparing prophylactic administration
with early rescue administration of exogenous surfactant
(within the first 30 to 60 minutes) have been performed
(see Chapter 46). In some centers, ELBW infants are
observed briefly after birth, and if respiratory distress
develops, they are intubated and ventilated for transfer to
the NICU. Exogenous surfactant is given within 1 hour
of birth, after endotracheal tube position and the presence
of RDS are confirmed on chest radiograph. This strategy
assures uniform distribution of surfactant to both lungs
and allows initial stabilization and more intensive monitoring of the infant during its administration. Recent data
suggest that many spontaneously breathing, extremely premature infants can be managed successfully with continuous positive airway pressure (CPAP) started in the delivery
room (Aly et al, 2005b; Lindner et al, 1999; Morley et al,
2008; SUPPORT Study Group, 2010). Routine use of
CPAP immediately after delivery may obviate the need
for intubation in infants with a gestational age of 24 weeks
or more, and increasing experience with this approach has
been shown to improve its success (Aly et al, 2005b; Finer,
2006). According to one report, mechanical ventilation
was avoided in approximately one third of infants with a
gestational age of 25 weeks or less, and in nearly 80% of
infants with a gestational age of 28 weeks or more using
this approach (Aly et al, 2005b). Whereas aggressive use of
early CPAP in the delivery room avoids intubation in some
ELBW infants, it remains unclear whether it improves
longer term respiratory outcomes (Morley et al, 2008;
SUPPORT Study Group, 2010).
If intubation is required, both oral and nasal routes for
endotracheal tube placement are equally effective and have
similar complication rates (Spence and Barr, 2002). Preference is usually institution specific, although successful
placement of a nasal endotracheal tube requires more time.
ADMISSION TO THE NEONATAL
INTENSIVE CARE UNIT
All infants should be weighed upon admission; frequent
determination of subsequent weights is a valuable tool in
managing fluid and electrolyte balance. In many centers,
ELBW infants are initially placed under a radiant warmer
for easier access (i.e., for surfactant administration and
catheter placement). Because of the high transepidermal
fluid losses in these infants, intravenous solutions containing 5% to 10% dextrose should be started as quickly as
possible after admission, and efforts should be made to
ameliorate evaporative water losses by increasing the relative humidity surrounding the infant. A plastic heat shield
used in concert with the radiant warmer may decrease transepidermal water losses; alternatively, a polyethylene tent
with an infusion of warmed humidified air may be used.
Several studies suggest that fluid management is improved
by using a humidified incubator instead of a radiant
warmer, because of lower water losses (Gaylord et al, 2001;
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
TABLE 32-6 Recommended Screening for Common
Complications of Extremely Low-Birthweight
Infants
Complication
Screening
IVH
HUS on days 1-3; repeat on days 7-10
Germinal matrix
hemorrhage
Repeat HUS weekly until findings
normal
Intraventricular
hemorrhage
Repeat HUS every 3 to 7 days until
stable or resolved
IVH with ventricular dilation or intraparenchymal
bleeding
Repeat HUS every 3 to 7 days until
stable or resolved
Periventricular white
matter disease
HUS at day 30; repeat at 36 weeks of
postmenstrual age or at discharge
Consider magnetic resonance imaging
if HUS findings are equivocal
ROP
Perform OE examination at 4 to 6
weeks of postnatal age
Repeat every 2 weeks if no ROP
Repeat weekly if ROP present
Repeat twice weekly for prethreshold
disease or rapidly progressive ROP
Audiology screening
Hearing screen no earlier than
34 weeks of postmenstrual age, but
before discharge home
Consider measurement of resistive
indices for progressive ventricular
dilatation
HUS, Head ultrasonography; IVH, intraventricular hemorrhage; OE, ophthalmologic
examination; ROP, retinopathy of prematurity.
Meyer et al, 2001). Exposure to high humidity may raise
the rate of skin colonization with gram-negative organisms, although no increase in the rate of nosocomial infections was observed in several randomized trials comparing
incubators with radiant warmers (Flenady and Woodgate,
2002). However, temperature regulation for the smallest infants in the first few days may be more difficult in
an incubator than in a servo-controlled radiant warmer,
because of rapid drops in air temperature as the incubator
doors are opened to care for the infant (Meyer et al, 2001;
see also Chapter 30).
VASCULAR ACCESS
Close monitoring of blood pressure, arterial blood gases,
and serum chemistries during the first few days after birth
is required in most sick ELBW infants; therefore it is
advantageous to insert an umbilical arterial catheter for
reliable access in infants who require assisted ventilation.
Infusion of half-normal saline with 0.5 unit of heparin per
milliliter at a low rate (0.5 to 1 mL/h) is usually enough to
maintain catheter patency. Although using a saline solution rather than a dextrose-water solution in the umbilical
arterial line may complicate fluid and electrolyte management, this disadvantage is offset by the advantage of reliable
measurement of blood glucose levels, which are frequently
required, without disturbing the infant. Placement of a
central umbilical venous catheter (tip at the inferior vena
cava–right atrial junction) at the same time the umbilical
arterial catheter is placed also provides the clinician with
reliable venous access for infusion of fluids, medications,
395
and blood products. Use of a double-lumen umbilical
venous catheter often obviates insertion of a peripheral
intravenous line over the first few days after birth and
helps to preserve intravenous sites and skin integrity.
The length of time that umbilical catheters are left in place
varies by hospital. In most centers, umbilical lines are generally discontinued after 7 to 10 days because of the potential
for catheter-related infection and vascular complications,
although it is unusual for arterial access to be needed beyond
a few days after birth. Before the umbilical venous catheter
is removed, it is advisable to insert a percutaneous central
venous catheter with its tip at the junction of the superior or
inferior vena cava and the right atrium, dedicated to infusion
of parenteral nutrition (see Nutritional Management, later).
This catheter helps to maintain intravenous access for nutritional purposes without raising the risk of catheter-related
infections and reduces the need to establish and maintain
peripheral intravenous lines (Janes et al, 2000; Parellada
et al, 1999). The incidence of complications of percutaneous central catheters, including catheter-related infections,
is lower if a limited number of NICU personnel insert and
maintain the lines.
SKIN CARE
The skin of an infant born at 23 to 26 weeks’ gestation is
extremely immature and is ineffective as an epidermal barrier. Poor epidermal barrier function in the extremely preterm infant leads to disturbances in temperature regulation
and water balance as well as breakdown in skin integrity,
which can increase the risk of infection. The stratum corneum, which is responsible for epidermal barrier function,
does not become functionally mature in the fetus until
approximately 32 weeks’ gestation (Rutter, 2000). However, acceleration of the maturation process occurs after
birth, so most extremely premature infants have a mature
epidermal barrier by approximately 2 weeks of postnatal
age. Until that time, full-thickness skin injury can occur in
the ELBW infant from seemingly innocuous causes, such
as local pressure from body positioning, removal of adhesives, and prolonged exposure to products containing alcohol or iodine. Such injury can lead to larger transepidermal
water losses, an even greater risk of nosocomial infection,
and significant scarring.
Because of these risks, preservation of skin integrity should
be incorporated into the care of the extremely preterm
infant (Table 32-7). Limited use of adhesives and extreme
care upon their removal, frequent repositioning of the infant
to avoid pressure points on the skin, and use of soft bedding
or a water mattress are the minimum requirements. Hydrocolloids (e.g., DuoDerm, ConvaTech, U.S.) applied to the
baby in areas where adhesive tape may come in prolonged
contact with the skin (i.e., umbilical catheter or endotracheal
tube fixation) to prevent the direct application of tape to the
baby may be useful, because it is more easily removed than
standard adhesive tape. Polyurethane adhesive dressings
(Tegaderm 3m, St. Paul, Minnesota) or hydrogels (Vigilon,
Bard Medical, Covington, Georgia) may also be used to protect areas of skin friction and superficial wounds.
Prophylactic application of preservative-free emollient
ointments (Aquaphor, Beiersdorf AG, Hamburg, Germany)
to protect the skin of the ELBW infant has been studied
396
PART VIII Care of the High-Risk Infant
TABLE 32-7 Practical Guidelines for Skin Care of Extremely
Low-Birthweight Infants
Interventions
Guidelines
Adhesive application
Increase adhesive tack by applying to dry,
clean skin surface
Avoid alcohol for skin cleansing
Use smallest amount of tape possible
Use a hydrocolloid or pectin-based layer
on the skin, before application of heavy
adhesive
Avoid using adhesive over areas of skin
breakdown
Avoid adhesive bonding agents (e.g., benzoin)
Use hydrophilic gel or pectin-based
adhesives preferentially
Adhesive removal
Avoid adhesive removers and solvents
Use a warm, wet cotton ball to periodically
saturate hydrogel adhesives (avoid
overdrying and oversaturation)
Facilitate removal of adhesive with mineral
oil, petrolatum, and emollients if
reapplication is not necessary
Emollient
application
Infants born at <27 weeks’ gestation may
benefit from emollient use
Avoid multidose containers (e.g., large jars)
Use nonperfumed, nonirritating
hydrophilic emollients
Recognize potential for emollients to
interfere with adhesive and conductive
properties of monitoring devices
Emollient removal
Wipe off gently with a soft cloth or gauze
if site is contaminated
Avoid repeated attempts to thoroughly
cleanse the skin (undesired friction effect)
Remove emollients before attaching
thermistors or other monitoring devices
Data from Hoath S, Narendran V: Adhesives and emollients in the preterm infant,
Semin Neonataol 5:289-296, 2000.
(Edwards et al, 2004; Lane and Drost, 1993; Nopper et al,
1996). Several small studies demonstrated smaller transepidermal water losses, improved skin condition, and lower
risk of suspected or proven nosocomial infection with prophylactic application of emollient ointments. However, one
report documented an increase in the rate of systemic yeast
infections in a single NICU coincident with a change to use
of prophylactic emollients in ELBW infants, who returned
to baseline after such use was discontinued (Campbell et al,
2000). In the largest study to date, infants randomized to
prophylactic emollient had better skin integrity during the
first month after birth, but had a higher rate of nosocomial
bacterial infection compared with a control group with no
emollient use (Edwards et al, 2004). Given these concerns,
routine use of prophylactic emollients is not recommended;
however, selective use in ELBW infants at risk for significant skin breakdown may be effective as an adjunct to other
types of local skin care already described.
MECHANICAL VENTILATION AND NASAL
CONTINUOUS AIRWAY PRESSURE
A high percentage of ELBW infants require some level of
assisted ventilation to survive. Data from animal models of
RDS indicate that positive-pressure ventilation with large
tidal volumes damages pulmonary capillary endothelium,
alveolar and airway epithelium, and basement membranes.
This mechanical damage results in leakage of fluid, protein, and blood into the airways, alveoli, and interstitial
spaces, leading to inhibition of surfactant activity and further damage to the lungs. These data suggest that a ventilator strategy that avoids large changes in tidal volume may
reduce ventilator-induced lung injury in ELBW infants, an
important management goal. The optimal mode, timing,
and application of ventilatory support used in the initial
management of the ELBW infant to meet this goal remain
controversial. However, the objectives of all strategies of
assisted ventilation in the ELBW infant should be similar—(1) to provide the lowest level of ventilatory support
possible that will both support adequate oxygenation and
ventilation and prevent atelectasis and (2) to try to reduce
acute and chronic lung injury secondary to barotrauma,
volutrauma, and oxygen toxicity (Clark et al, 2000). Data
also suggest that targeting oxygen saturations (88% to
93%) lower than those used historically in ELBW infants
who are receiving supplemental oxygen may protect the
lung from oxidative injury and result in better outcomes
(Askie et al, 2003).
CONTINUOUS POSITIVE AIRWAY PRESSURE
In a classic study comparing the incidence of BPD between
NICUs, Avery et al (1987) reported that the NICU with
the lowest incidence used CPAP more frequently and
more aggressively than the other units. Van Marter et al
(2000) confirmed these observations.
Theoretically, early CPAP protects the immature lung
from injury caused by positive-pressure tidal breaths, by
preserving surfactant function, increasing alveolar volume and functional residual capacity, enhancing alveolar
stability, and improving ventilation-perfusion matching.
It also may stimulate the growth of the immature lung.
Some NICUs now use CPAP (delivered via nasal prongs
or nasopharyngeal tube at 5 to 7 cm H2O) as the initial
mode of assisted ventilation for ELBW infants, starting
almost immediately in the delivery room (Lindner et al,
1999). CPAP used this way has been reported in retrospective studies to decrease the need for mechanical ventilation
(Gittermann et al, 1997; Poets and Sens, 1996), the need for
surfactant treatment, and the incidence of BPD (Aly, 2001;
Aly et al, 2005b; de Klerk and de Klerk, 2001; Lindner
et al, 1999). In one prospective study (Morley et al, 2008),
infants with a gestational age of 25 to 28 weeks who were
breathing spontaneously, but required ventilatory assistance at 5 minutes after birth, were randomly assigned to
treatment with nasal CPAP or intubation and mechanical ventilation. In the infants assigned to CPAP, 56% did
not require intubation, and surfactant use was halved.
Although respiratory outcomes at 36 weeks postmenstrual
age were equivalent in the two study groups, a greater
number of infants who were assigned to initial treatment
with CPAP had pneumothorax (9% versus 3%). Alternatively, CPAP may be used after prophylactic or rescue
surfactant therapy is given during a brief period of intubation and positive-pressure ventilation (Booth et al, 2006;
Verder et al, 1999). Observational studies of this approach
show that approximately one fourth of infants born before
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
27 weeks’ gestation do not require a subsequent course of
mechanical ventilation and are less likely to develop BPD
(Booth et al, 2006; Dani et al, 2004).
CPAP can be delivered by a conventional mechanical
ventilator with continuous flow, a variable flow device
that adjusts flow through the respiratory cycle, or via
“bubble” CPAP, in which a tube is immersed in water
to the desired depth to generate CPAP with continuous
gas flow bubbling through the immersed tube. Animal
data suggest that bubble CPAP improves lung volume
and gas exchange compared with conventional CPAP
(Pillow et al, 2007), perhaps secondary to effects of the
higher intranasal pressures generated by the bubbling
water (Kahn et al, 2008). Other data suggest that work
of breathing and thoracoabdominal asynchrony may be
lessened with variable flow devices (Liptsen et al, 2005).
Whether these differences are clinically relevant remains
to be elucidated.
CONVENTIONAL MECHANICAL VENTILATION
Most NICUs continue to use pressure-limited, timecycled conventional ventilators for the initial respiratory
management of the ELBW infant requiring mechanical
ventilation. Synchronized intermittent mandatory ventilation (SIMV) remains the preferred mode of conventional
ventilation for the ELBW infant. In the premature infant,
SIMV, in which the inspiratory cycle is synchronized with
the patient’s own effort, is better than conventional IMV in
terms of oxygenation, ventilation, work of breathing, and
blood pressure variability (Cleary et al, 1995; Hummler et al,
1996; Jarreau et al, 1996). Technologic limitations in the
ability of some ventilators to synchronize breaths with
very weak inspiratory efforts may prevent use of SIMV in
the smallest infants. With further advancement in ventilator design, other modes of patient-triggered ventilation,
including volume guarantee, assist control, and pressure support, have become available to clinicians. These
designs generally allow ventilation at lower pressures than
conventional SIMV. These modes of ventilation are under
investigation but have not been proved to produce better
pulmonary outcomes than SIMV in ELBW infants (Donn
and Sinha, 1998; Herrera et al, 2002), although one report
suggested a shorter time to extubation in infants babies
managed with pressure-support ventilation (Reyes et al,
2006).
Currently accepted conventional ventilatory strategies
in the ELBW infant stress the avoidance of excessive tidal
volumes by limiting peak inspiratory pressures and provision of adequate PEEP to maintain lung volume (Thome
et al, 1998). This strategy helps to prevent repeated cycles
of atelectasis and lung overdistention, a risk factor for
ventilator-induced lung injury (Dreyfuss and Saumon,
1998). Hyperventilation (Paco2 <35 mm Hg) has been
associated with a higher risk for the development of BPD
(Garland et al, 1995; Van Marter et al, 2000) and neurodevelopmental sequelae in ELBW infants (Wiswell et al,
1996).
In response to this suggestion of worse pulmonary outcome in hyperventilated infants, a strategy of minimal
ventilation, or permissive hypercapnia, has been proposed for conventional ventilation in ELBW infants. In
397
the largest study to date of this ventilatory strategy, Carlo
et al (2002) randomly assigned 220 infants with birthweights between 501 and 1000 g to receive either minimal
ventilation (target Paco2 >52 mm Hg) or routine ventilation (target Paco2 <48 mm Hg). No difference in the
rate of BPD, death, or other major short-term morbidities was seen between the two groups. Potential risks of
a high Paco2 include increases in both cerebral perfusion
and pulmonary vascular resistance and lower pH. Without
clear data to support the benefit and safety of higher levels of hypercapnia, most centers target their conventional
ventilatory strategy to maintain Paco2 between 45 and 55
mm Hg in the first several days of mechanical ventilation
in the ELBW infant.
In infants whose lung disease prevents extubation in
the first several days after birth, changes in lung dynamics over the first 1 to 2 weeks often necessitate a change
in ventilatory strategy. In the early stages of chronic lung
disease, increased airway resistance and decreased lung
compliance may require higher values for mean airway
pressure, peak inspiratory pressure, PEEP, and inspiratory time than are usually used in initial ventilatory management. Once the need for more prolonged mechanical
ventilation is established, many centers tolerate higher
target Paco2 values in an attempt to limit further ventilator-induced lung injury.
HIGH-FREQUENCY VENTILATION
Considerable interest has been generated over the past
15 years in the application of high-frequency ventilation
(HFV) in newborns who have respiratory failure, because
this technique allows ventilation with small tidal volumes.
Results of studies using HFV in animal models of RDS
have been promising in the prevention of lung injury, but
results of clinical studies of this ventilatory technique have
not. Despite many clinical trials, controversy continues
to surround the indications for HFV in ELBW infants,
whether HFV is more effective than other modes of ventilation for RDS, whether HFV reduces adverse outcomes
(specifically BPD), and whether HFV is more likely to
have significant long-term complications than conventional mechanical ventilation.
Early trials of HFV before surfactant replacement
therapy demonstrated no pulmonary advantage of HFV
over conventional mechanical ventilation and suggested
an increase in rates of air leak and intracranial abnormalities in the HFV-treated infants (HIFI Study Group,
1989). Later trials in ELBW infants who were treated
with surfactant also failed to demonstrate any reduction in the incidence of BPD in the HFV-treated infants
(Johnson et al, 2002; Rettitz-Volk et al, 1998; Thome
et al, 1999).
A rigorously controlled trial of HFV as the primary
mode of assisted ventilation compared with conventional
mechanical ventilation was the first to suggest a small
advantage to the early use of HFV in reduction of BPD or
death without an increase in the incidence of short-term
complications in ELBW infants. Courtney et al (2002)
compared high-frequency oscillatory ventilation (HFOV)
with synchronized IMV in a randomized trial that enrolled
500 ELBW infants. These investigators found a small but
398
PART VIII Care of the High-Risk Infant
significant decrease in the incidence of BPD in survivors,
requirement of fewer doses of exogenous surfactant, and
shorter time to successful extubation in the HFOV-treated
group. No differences in other complications of prematurity were observed between the two groups.
These results suggest that, when used in experienced
hands according to strict protocol, HFV confers some
protection from lung injury in ELBW infants. It remains
unclear whether this same advantage is gained when HFV
is used in usual clinical circumstances in less experienced
centers and whether the incidence of other complications
may be affected by mode of ventilation. Some NICUs with
the most experience with HFV use it routinely as the initial mode of ventilation for ELBW infants. Most centers
continue to use conventional ventilation with low tidal volumes and reasonable ventilation goals as the initial mode of
ventilation for ELBW infants, reserving HFV for infants
in whom conventional ventilation and surfactant fail. This
latter practice seems advisable, given the potential risks of
HFV, including inadvertent lung overdistention, impaired
cardiac output, and increased central venous pressure that
may lead to intracranial hemorrhage.
POST-EXTUBATION CPAP FOR RESPIRATORY
DISTRESS SYNDROME AND APNEA
CPAP is commonly used in ELBW infants after extubation to stabilize functional residual capacity and reduce
the frequency of apneic spells after the lung disease has
improved. All ELBW infants should be extubated as soon
as they have recovered from acute RDS and should be
given a trial of CPAP to protect them from further ventilator induced lung injury. When used in combination
with methylxanthine therapy, CPAP decreases the need
for reintubation because of progressive respiratory distress or apnea (Davis et al, 2003). Methylxanthine (aminophylline or caffeine) treatment before extubation has
also been shown to lower the incidence of apnea and the
need for reintubation in premature infants. Many ELBW
infants benefit from prolonged use of CPAP by nasal
prongs after extubation, especially those with frequent
or severe episodes of apnea and bradycardia resistant to
methylxanthine. Some centers currently provide assisted
ventilation through nasal prongs (nasal intermittent positive pressure ventilation) to avoid intubation (Khalaf et al,
2001); this method may be more successful than conventional CPAP in avoiding postextubation failure in some
infants (Bhandari et al, 2009). However, the smallest and
least mature infants may need prolonged mechanical ventilation because of frequent, severe apneic spells that are
unresponsive to other therapies.
ADJUNCTIVE THERAPIES TO PREVENT
BRONCHOPULMONARY DYSPLASIA
Vitamin A Supplementation
ELBW infants have low stores of vitamin A. Because of
the role of vitamin A in promoting lung healing, its deficiency has been linked to a higher risk for development
of BPD. In a large multicenter trial, vitamin A supplementation (5000 IU given intramuscularly three times
per week for 4 weeks) reduced biochemical evidence of
vitamin A deficiency and decreased the incidence of BPD
by 12% without adverse effects in ELBW infants who
required mechanical ventilation or supplemental oxygen
at 24 hours of age (Tyson et al, 1999). Given these efficacy data and apparent safety, many NICUs choose to
administer vitamin A supplementation as described previously to all ELBW infants starting at 24 hours after
birth. However, widespread acceptance of this therapy
has been limited by concerns over the need for thrice
weekly intramuscular injections and its associated pain
and stress to the infant.
Caffeine
In a prospective, masked, randomized trial involving
infants with a birthweight of 500 to 1250 grams, the initiation of treatment with caffeine citrate (20 mg/kg loading
dose, followed by 5 mg/kg/day) in the first 10 days after
birth decreased the rate of BPD (a secondary outcome), as
compared with placebo (36% versus 47%) without adverse
effects (Schmidt et al, 2006). The composite primary outcome of death, cerebral palsy, cognitive delay, deafness or
blindness at 18 to 22 months of age was also reduced in
the subjects randomized to caffeine (Schmidt et al, 2007).
The effect of caffeine on the incidence of bronchopulmonary dysplasia may be secondary to less ventilator-induced
lung injury in the caffeine-treated infants, because shorter
duration of positive pressure ventilation and supplemental oxygen therapy was associated with its use (Schmidt
et al, 2006). These results suggest that caffeine should be
administered routinely in ELBW infants in the first 10
days after birth, even if they continue to require mechanical ventilation.
Inhaled Nitric Oxide
Inhaled nitric oxide (iNO) may improve the pulmonary
outcome in some VLBW infants, through mechanisms
that are thought to involve decreased pulmonary vascular
resistance or improved ventilation-perfusion matching,
bronchodilatation, antiinflammatory effects, promotion of
lung remodeling in response to injury, or normalized surfactant function. iNO used as rescue therapy for ELBW
infants with severe hypoxic respiratory failure does not
improve the pulmonary outcome or survival, and it may
be associated with increased mortality or an increased
incidence of intraventricular hemorrhage (Van Meurs et
al, 2005). Results of trials of iNO involving premature
infants with less severe lung disease who were at risk for
BPD are mixed. In the Nitric Oxide for the Prevention of
Chronic Lung Disease Trial (NO-CLD) study, a multicenter trial involving ventilator-dependent infants with a
birthweight of 500 to 1250 g, treatment with iNO started
at 7 to 14 days and continued for an average of 23 days of
increased survival without BPD as compared with placebo
(Ballard et al, 2006). In contrast, in a multicenter trial of
iNO, treatment started before 48 hours of age and continued for 21 days (20 ppm for 3 to 4 days, then weaned
to 10 ppm, 5 ppm, and 2 ppm for 7 days each) in infants
with a birthweight of 500 to 1250 g who continued to
require mechanical ventilation, only iNO-treated infants
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
with a birthweight greater than 1000 g had a pulmonary
benefit (Kinsella et al, 2006). Treated infants in this latter
study were also less likely to have ultrasonic evidence of
brain injury than were control infants. Long-term followup from the latter two studies suggests that iNO is safe
and may improve long term pulmonary outcomes (Hibbs
et al, 2008; Watson et al, 2009). iNO use for the prevention of BPD is not currently approved by the United
States Food and Drug Administration. Routine use of
iNO for the prevention of BPD should be discouraged
until additional studies are available to define dosing and
duration of therapy.
Systemic Corticosteroids
Inflammation also plays an important role in the pathogenesis of BPD; therefore pharmacologic doses of
systemic corticosteroids have been widely used for prevention and treatment of BPD in ELBW infants. Several
studies have examined early (<96 hours of age) administration of corticosteroids, usually dexamethasone, to prevent the development of BPD in infants at risk (Garland
et al, 1999; Rastogi et al, 1996; Stark et al, 2001). A metaanalysis of studies in which corticosteroids were used to
prevent rather than treat established BPD suggested that
early corticosteroid treatment in ELBW infants results
in more rapid extubation and a lower incidence of BPD
(Halliday et al, 2002), although no effect on mortality
was observed. However, a higher incidence of short-term
complications, including hyperglycemia, hypertension,
poor growth, and intestinal perforations and bleeding,
was observed in the corticosteroid-treated infants (Garland et al, 1999; Stark et al, 2001; Watterberg et al, 2004).
More importantly, long-term follow-up data suggest that
exposure to corticosteroid for prevention or treatment of
BPD raises the risk of neurological sequelae in treated
infants, including poor head growth, cerebral palsy,
and developmental impairment (American Academy of
Pediatrics, 2010).
The apparently higher risk of long-term sequelae without an effect on overall mortality has tempered enthusiasm for systemic corticosteroid treatment to prevent or
treat BPD in ELBW infants. The possible role of postnatal corticosteroids in the prevention of BPD in selected
infants, such as those exposed to chorioamnionitis, was
suggested by a trial of early treatment with hydrocortisone (Watterberg et al, 2004). The risk of impaired neurodevelopment may be lower with hydrocortisone than
dexamethasone (Rademaker et al, 2007; Watterberg et al,
2007), and some centers are substituting hydrocortisone
for dexamethasone if postnatal corticosteroids are used.
However, it seems prudent to avoid routine use of systemic corticosteroids in ELBW infants for the prevention or treatment of BPD until further data are available
(American Academy of Pediatrics, 2010).
NUTRITIONAL MANAGEMENT
Provision of adequate nutrition is central to effective care
of ELBW infants (see Chapters 66 and 67). These infants
are born with limited nutrient reserves, immature pathways for nutrient absorption and metabolism, and higher
399
nutrient demands. In addition, medical conditions associated with extreme prematurity both alter requirements for
and complicate the adequate delivery of nutrients. The
goals of nutritional management of the ELBW infant are
preservation of endogenous body stores, achievement of
postnatal growth similar to intrauterine weight accretion
and body composition, and maintenance of normal physiologic and metabolic processes concomitant with minimizing complications and side effects. However, few ELBW
infants are able to meet these goals despite the use of central parenteral nutrition and caloric supplementation of
enteral feedings. As a result, significant growth failure is
commonplace (Berry et al, 1997; Ehrenkranz et al, 1999;
Martin et al, 2009).
Enteral Nutrition
Medical problems of ELBW infants sometimes preclude
initiation of enteral feedings for several days to weeks.
However, the structural and functional integrity of the
gastrointestinal tract depends on the provision of enteral
feedings. Withholding enteral feedings at birth imposes
risks for all the complications of luminal starvation, including mucosal thinning, flattening of the villi, and bacterial
translocation. Early initiation (within the first few days
after birth) of low volumes of milk (10 to 20 mL/kg/day,
preferably with expressed breast milk; trophic feedings,
or “gut priming”) has been studied in several small trials
in premature infants (McClure and Newell, 2000; Schanler et al, 1999a). Trophic feedings are not meant to give
the infant significant nutrition, rather to promote continued functional maturation of the gastrointestinal tract.
Documented benefits of trophic feedings include higher
plasma concentrations of gastrointestinal hormones, a
more mature gut motility pattern, lower incidence of cholestasis, increased calcium and phosphorus absorption, and
improved and earlier tolerance of enteral feedings. Trophic feedings have not been associated with a higher risk
of necrotizing enterocolitis or other adverse outcomes;
therefore there is no clinical advantage to delaying initiation of feedings in the medically stable ELBW infant. (See
also Chapter 66.)
Feeding intolerance, indicated by gastric residuals that
exceed 25% to 50% of the volume fed, abdominal distention, or microscopic blood in the stool, is common in
ELBW infants and may be difficult to differentiate from
early stages of necrotizing enterocolitis. Feeding intolerance may preclude the advance of enteral nutrition for days
to weeks, complicating nutritional management and prolonging the need for parenteral nutrition. Numerous feeding strategies to avoid episodes of feeding intolerance have
been used in ELBW infants, including slow increase in
enteral volume (<10 mL/kg/day), use of dilute rather than
full-strength milk, continuous versus bolus tube-feeding,
and use of prokinetic agents. None of these feeding strategies has been found to be clearly superior, although bolus
feedings may decrease episodes of gastric residuals compared with continuous tube feedings and may allow a more
rapid advance to full enteral volumes as well as promote
better growth.
Episodes of feeding intolerance also are reduced in
ELBW infants fed human milk rather than specialized
400
PART VIII Care of the High-Risk Infant
formulas for premature infants (Schanler et al, 1999b).
Other benefits of giving human milk in ELBW infants are
a more rapid advance to full enteral volumes and its positive immunologic effects, with an associated reduction in
the risk of necrotizing enterocolitis and late-onset sepsis.
Data also suggest that neurodevelopmental outcome may
be improved in ELBW infants fed expressed breast milk
(Vohr et al, 2006). Human milk must be fortified with
calcium, phosphorus, sodium, protein, and other minerals to provide adequate nutrition in the ELBW infant. In
addition to commercially available human milk fortifiers,
which increase the caloric density to approximately 24
calories per ounce, human milk can be fortified further to
higher caloric densities with medium chain triglycerides,
glucose polymers, and added protein.
Premature infants who are fed fortified human milk
may grow more slowly than infants fed premature formulas (Schanler et al, 1999b), perhaps because of the variability in fat and caloric content of pumped breast milk or
changes in the nutrient composition of human milk with
fortification that affect fat absorption. Despite the potential for slower growth in infants fed human milk, its use
should be strongly encouraged in ELBW infants because
of the immunologic and other nutritional benefits. Further
research on how best to fortify human milk is necessary to
promote the best rate of growth in ELBW infants. (See
also Chapters 65 and 66.) In addition, even after recommended enteral dietary intakes are reached, many ELBW
infants continue to have a cumulative energy and protein
deficit, which in part explains their later growth failure
(Ehrenkranz et al, 1999; Embleton et al, 2001; Martin
et al, 2009).
Early Parenteral Nutrition
Protein losses in ELBW infants receiving a glucose infusion alone begin immediately after birth and can approach
1.5 g/kg/day in the first 24 to 72 hours. Fortunately, these
losses can be offset by early administration of an amino
acid solution, even at low caloric intakes. Several studies have demonstrated the safety of early administration
(within 24 hours after birth) of an amino acid solution,
with no abnormal elevations of ammonia or blood urea
nitrogen even in the most immature infants (Rivera
et al, 1993; Van Goudoever et al, 1995). To prevent early
protein deficit, ELBW infants should be given a source of
parenteral protein as soon as possible after birth (see also
Chapter 67). In one study, ELBW infants who received 3
g or more of protein per day at 5 days of age or less were
less likely to have a weight below the 10th percentile at 36
weeks postmenstrual age and suboptimal head growth at
18 months of age when compared with ELBW infants who
received less protein supplementation after birth (Poin
dexter et al, 2006).
In most centers, parenteral nutrition is used exclusively
during the first few days after birth and then gradually
reduced as enteral feedings are introduced. Longer
duration of parenteral nutrition is associated with the
development of a number of complications, including
cholestasis, osteopenia, and sepsis. For example, the risk
of an episode of late-onset sepsis in premature infants is
22-fold higher if parenteral nutrition is continued for
more than 3 weeks compared with 1 week or less (Stoll
et al, 2002b).
MANAGEMENT AND PREVENTION
OF INFECTION
Bacterial and fungal infections are an important cause of
illness and death among ELBW infants. In addition to the
immediate morbidity and mortality, local and systemic
inflammation caused by infections may increase the risk
for development of other complications of prematurity,
including BPD and brain injury. ELBW infants are frequently exposed to perinatal and delivery complications
that raise their risk of early-onset (<72 hours) infections.
The need for prolonged intravenous access, exposure to
parenteral nutrition, and mechanical ventilation also subject the ELBW infant to a high risk of late-onset (>72
hours) nosocomial infections. The frequent infections
seen in the ELBW population are related to immaturity
of both humoral and cellular immunity (see Chapter 36).
In addition to the judicious use of antimicrobial therapy,
environmental controls, nursery surveillance, and modulation of the immature immune response have been proposed as possible interventions to prevent infections in
extremely premature infants.
Early-Onset Infections
The incidence of early-onset bacterial infections in very
LBW (VLBW) infants is approximately 1% to 2%, with
a mortality of approximately 40% to 50% (Stoll et al,
2002a; see also Chapter 39). The major risk factor for the
development of perinatally acquired bacterial infections is
PROM with chorioamnionitis, which frequently complicates premature deliveries. However, a significant percentage of extremely premature births may be associated with
intrauterine infection before membrane rupture (Goldenberg et al, 2000). In one study, 41% of premature infants
with early-onset sepsis were born less than 6 hours after
membrane rupture (Stoll et al, 2002a).
Because the clinical signs of perinatally acquired infection are nonspecific, the index of suspicion and the concern about the possibility of intrauterine infection should
always be high in the presence of premature birth. All
ELBW infants, except for those delivered for maternal
indications with no labor, should be evaluated for infection at birth by means of a complete blood count with differential and blood culture, and empiric antibiotic therapy
with ampicillin and an aminoglycoside should be initiated.
A white blood cell count less than 5000 cells/μL, a ratio
of immature to total neutrophils ratio greater than 0.2 to
0.3, and neutropenia (absolute neutrophil count less than
1000 cells/μL) are all suggestive of infection, but may also
be seen in infants with other conditions, including maternal preeclampsia and hypertension. The duration of initial antibiotic therapy depends on the results of the blood
culture, blood counts, the clinical course, and the perinatal history. If the blood culture is negative for bacterial
growth at 48 hours and the infant has improved clinically,
consideration should be given to discontinuing antibiotics.
Prolonged exposure to antibiotic therapy increases the likelihood of colonization with multiple antibiotic-resistant
401
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
organisms, the development of fungemia, and necrotizing
enterocolitis (Cotten et al, 2009); therefore it should be
reserved for infants with documented infection or a very
high index of suspicion of infection based on clinical or
historical factors.
The distribution of pathogens causing early-onset sepsis
in VLBW infants has changed, likely because of increased
use of intrapartum antibiotics for prevention of group B
streptococcal infections and treatment of preterm PROM
(Puopolo and Eichenwald, 2010). As the rate of infections
from group B streptococci has diminished with intrapartum antibiotic prophylaxis, the proportion of documented infections from gram-negative organisms has risen
(Table 32-8; Stoll et al, 2002a). In an additional change
of the epidemiology of early-onset infections in premature
infants, possibly related to increased use of intrapartum
antibiotics, the frequency of infections owing to ampicillin-resistant Escherichia coli strains has increased in some
centers (Bizzarro et al, 2008; Joseph et al, 1998; Stoll et al,
2002a). Current recommendations for empiric antibiotic
therapy for ELBW infants at risk of early-onset sepsis have
not changed, but continued surveillance of the epidemiology and antibiotic resistance patterns of isolates within
individual units is warranted. In infants with severe illness
that may be caused by sepsis, broadening initial antibiotic coverage to include a third-generation cephalosporin
should be considered.
LATE-ONSET INFECTIONS
Nosocomial infection is a common though preventable
complication of intensive care of the ELBW infant. The
incidence of late-onset sepsis in ELBW infants who survive beyond 3 days of age is 25% to 50%, depending on
gestational age and birthweight, with the median age at
onset of the first episode approximately 2 weeks (Fanaroff
et al, 1998; Stoll et al, 2002b). The overall mortality rate is
approximately 20% but may be as high as 80%, depending
on the organism causing sepsis. The risk of neurodevelopmental impairment is increased by approximately 1.5-fold
in ELBW survivors of nosocomial bloodstream infections
(Stoll et al, 2004). Risk factors for the development of
late-onset sepsis include prolonged hyperalimentation and
lipid use, the presence of a central venous catheter, longer
duration of mechanical ventilation, and delay in initiation
of enteral feedings (Stoll et al, 2002b). These practices and
procedures are common events that may be unavoidable in
the care of ELBW infants. However, large variations have
been observed among NICUs in the rate of late-onset infections in premature infants (Brodie et al, 2000; Stoll et al,
2002b), suggesting that individual NICU practices may
affect the incidence of nosocomial infections.
The most common cause of late-onset infections
in ELBW infants is coagulase-negative Staphylococcus
(CoNS). The most significant risk factor for the development of CoNS infection is the use of a fat emulsion (e.g.,
Intralipid) infusion (Freeman et al, 1990). Infection with
CoNS is almost never fatal but is associated with significant morbidity, such as prolonged ventilator use and hospital stay (Gray et al, 1995). Other organisms that cause
late-onset infections in ELBW infants are associated with
a much higher morbidity and mortality. The distribution
TABLE 32-8 Distribution of Pathogens among 84 Cases
of Early-Onset Sepsis*
Organism
No.
%
Gram-negative:
51
60.7
Escherichia coli
37
44.0
Haemophilus influenzae
7
8.3
Citrobacter
2
2.4
Other
5
6.0
31
36.9
Group B streptococci
9
10.7
Viridans streptococci
3
3.6
Other streptococci
4
4.8
Listeria monocytogenes
2
2.4
Coagulase-negative staphylococci
9
10.7
Other
4
4.8
Gram-positive:
Fungi: Candida albicans
2
84
Total
2.4
100
Data from Stoll B, Hansen N, Fanaroff A, et al: Changes in pathogens causing early
onset sepsis in very low birth weight infants, N Engl J Med 347:240-247, 2002.
*Occurring in 5447 infants born between September 1, 1998, and August 31, 2000.
TABLE 32-9 Distribution of Pathogens Associated With the First
Episode of Late-Onset Sepsis*
Organism
No.
%
Gram-positive:
922
70.2
Coagulase-negative staphylococci
629
47.9
Staphylococcus aureus
103
7.8
43
3.3
30
2.3
Enterococcus spp.
Group B streptococci
Other
Gram-negative:
117
8.9
231
17.6
Escherichia coli
64
4.9
Klebsiella spp.
52
4.0
Pseudomonas spp.
35
Enterobacter spp.
33
2.5
Serratia spp.
29
2.2
Other
Fungi:
Candida albicans
27
18
1.4
160
12.2
76
5.8
Candida parapsilosis
54
4.1
Other
30
2.3
Total
1313
100
Data from Stoll B, Hansen N, Fanaroff A, et al: Late-onset sepsis in very low birth
weight neonates: experience of the NICDH Neonatal Research Network, Pediatrics
110:285-291, 2002.
*In National Institute of Child Health and Development Neonatal Research Network
institutions, September 1, 1998, through August 31, 2000.
of pathogens associated with the first episode of late-onset
sepsis among 1313 infants in a cohort of VLBW babies
over a 2-year period is shown in Table 32-9.
Presenting features of late-onset sepsis include increased
apnea, feeding intolerance, abdominal distention, guaiacpositive stools, increased respiratory support, and lethargy and hypotonia (Fanaroff et al, 1998). Because these
402
PART VIII Care of the High-Risk Infant
symptoms are nonspecific, ELBW infants are frequently
evaluated for infection and treated with empiric antibiotic therapy. In one study, use of both vancomycin and
antifungal therapy was inversely related to birthweight;
approximately three fourth of infants with a birthweight
less than 750 g was treated with vancomycin during their
hospital stay, and approximately one third was treated with
antifungals (Stoll et al, 2002b). Central catheters should
be removed immediately to ensure adequate treatment of
infants in whom sepsis is diagnosed, except for that caused
by CoNS (Benjamin et al, 2001).
Endotracheal tube colonization with multiple organisms
is common in infants who require prolonged mechanical
ventilation. In general, such colonization should not be
treated with antibiotics unless there is evidence of pneumonia or significant inflammation indicative of tracheitis.
Prospective surveillance of common isolates and antimicrobial resistance patterns within individual NICUs can
help to guide empiric antibiotic therapy in ELBW infants
being evaluated and treated for presumed sepsis. However,
indiscriminate use of broad-spectrum antibiotics in the
absence of true infection can alter antimicrobial resistance
patterns (Goldmann et al, 1996), raising the risk of lateonset infections and complicating therapy.
Prevention of Nosocomial Infection
Because of the frequency and potential severity of lateonset sepsis in ELBW infants, several strategies to prevent
infection have been proposed. Using these practices as a
guideline, Horbar et al (2001) observed a decrease in the
CoNS infection rate from 22% to 16.6% over a 2-year
period in VLBW infants in six study NICUs. Other investigators have confirmed that changes in practice, primarily
surrounding the use and care of central venous catheters,
can reduce the overall burden of late onset infections in
individual units (Aly et al, 2005a; Kilbride et al, 2003).
Some NICUs routinely screen ELBW infants by stool
or respiratory secretion cultures for the presence of multiple antibiotic-resistant organisms, which would necessitate isolation (Gregory et al, 2009). Clusters of infections
with unusual organisms should prompt surveillance cultures of infants, potential NICU environmental sources,
and NICU staff (Foca et al, 2000). Restriction of broadspectrum antibiotic use by hospital policy or treatment
guidelines may limit the local spread of resistant organisms
(Goldmann et al, 1996). Strict adherence to hand hygiene
before and after every patient contact and avoidance of
overcrowding within NICUs also help to decrease the
incidence of infection. The use of alcohol-based hand gels
at the bedside may improve compliance with hand hygiene
(Harbarth et al, 2002).
In addition to practice and environmental controls, prophylactic use of antibiotics and modulation of the immune
response of ELBW infants have been studied as methods
to reduce the incidence of late-onset sepsis. Low-dose
vancomycin given continuously via hyperalimentation
solutions (Baier et al, 1998; Spafford et al, 1994), intermittently via peripheral vein (Cooke et al, 1997), or vancomycin lock of the central venous catheter (Garland et al,
2005) has been shown to reduce the incidence of CoNS in
premature infants at risk. Concern about the emergence
of vancomycin-resistant organisms and the low mortality
associated with CoNS infections has prevented widespread
use of this approach. Prophylactic fluconazole given for 6
weeks lowered the incidence of fungal colonization and
invasive disease in ELBW infants without associated
complications or the emergence of resistant organisms
(Kaufman et al, 2001, 2005; Manzoni et al, 2007). Such
an approach might be advisable for NICUs with a high
incidence of fungal infections in their ELBW population,
but more study is needed to define any potential short- and
long-term risks. Approaches that have been used with success in immunocompromised adult patients, such as antiseptic-impregnated central catheters, are promising but
have not yet been studied adequately in premature infants.
Prophylactic intravenous administration of polyclonal
immunoglobulin (IVIG) to prevent late-onset sepsis has
been studied extensively in premature infants. Several trials have shown a decrease in the incidence of documented
sepsis by a small but significant amount in premature
infants treated with prophylactic IVIG and no effect on
mortality or other complications of prematurity (Lacy and
Ohlsson, 1995). The costs associated with this therapy
to achieve the small decrease in infection rates, as well as
the increased exposure to blood products, have limited
its use; it is unclear whether selective prophylactic IVIG
treatment of ELBW infants at highest risk for sepsis is
warranted. However, IVIG therapy in addition to antibiotic therapy may be of benefit in reducing mortality in
infants with established sepsis (Jenson and Pollock, 1997).
Development of more targeted polyclonal or monoclonal
γ-globulin preparations for specific organisms that cause
sepsis in premature newborns may alter the use of IVIG in
the future (Lamari et al, 2000; Weisman et al, 2009).
Another promising strategy under investigation for modulation of the immature immune response to help prevent
infections in premature infants is treatment with hemopoietic colony-stimulating factors, including granulocyte
colony-stimulating factor and granulocyte-macrophage
colony-stimulating factor (Modi and Carr, 2000). Most
studies of these factors have been conducted in neutropenic, small-for-gestational-age infants or infants delivered
to women with preeclampsia. Treatment with granulocyte
colony-stimulating factor in neutropenic infants resulted in
an increase in neutrophil counts and reduced the incidence
of sepsis (Kocherlakota and La Gamma, 1998). Prophylactic
treatment with granulocyte-macrophage colony-stimulating
factor in premature infants with normal neutrophil counts
prevented the development of neutropenia in episodes of
sepsis, but it remains unclear whether this type of therapy
to address cellular immune deficiency in ELBW infants will
reduce the incidence of infection without additional complications (Miura et al, 2001; Modi and Carr, 2000).
NEUROSENSORY COMPLICATIONS
The major neurosensory complications associated with
extreme premature birth are intraventricular hemorrhage,
periventricular white matter injury, and retinopathy of
prematurity. Although the incidence of severe intraventricular hemorrhage has fallen with improvements in management and increased antenatal steroid use, it remains
a major cause of brain injury with consequent abnormal
CHAPTER 32 Care of the Extremely Low-Birthweight Infant
neurodevelopment. Pharmacologic approaches to its prevention after birth have been generally unsuccessful. Prophylactic indomethacin reduces the incidence of severe
intraventricular hemorrhage, but does not improve longterm neurodevelopment (Schmidt et al, 2001).
Periventricular white matter injury is the predominant form of brain injury in extremely preterm infants
and correlates strongly with the development of cerebral
palsy. Its pathogenesis is poorly understood, and no specific neuroprotective strategy is known. In some infants,
cerebral blood flow and oxygen delivery measured with
near infrared spectroscopy varies during variations of
blood pressure considered to be in the normal range,
and this lack of autoregulation of cerebral blood flow
may lead to ischemic white matter injury (Evans, 2006).
Whether aggressive treatment of hypotension in ELBW
infants prevents or may lead to subsequent brain injury
is uncertain, probably because blood pressure, which is
easily measured, does not correlate well with systemic or
cerebral blood flow (Fanaroff et al, 2006; Limperopoulos
et al, 2007). A higher rate of white matter injury occurs
in the setting of maternal or neonatal infection, or with
elevated proinflammatory cytokines in amniotic fluid or
cord blood, suggesting that inflammation has a role in
the pathogenesis (Viscardi et al, 2004). Advanced magnetic resonance imaging techniques in infants with white
matter injury show disturbances in cerebral growth, with
reduced volume of both gray and white matter (Inder
et al, 2005). These observations might serve to explain
the motor and cognitive dysfunction often seen in infants
with white matter injury.
Retinopathy of prematurity, a vascular proliferative disorder that affects the incompletely vascularized retina of
preterm infants, is a major cause of blindness in these children. Severe retinopathy is 18-fold more likely to develop
in infants delivered at less than 25 weeks’ gestation compared with 28 weeks’ gestation (Fanaroff et al, 2007). Periods of hyperoxia owing to exposure to excessive inspired
oxygen concentration contribute to the development of
retinopathy (Saugstad, 2006); however, the optimal target range of oxygen saturation is not known. Because fetal
hemoglobin shifts the hemoglobin oxygen saturation curve
to the left, oxygen saturations greater than 95% may be
associated with arterial oxygen tension greater than 80 mm
Hg, possibly excessive for the ELBW infant. Conversely,
oxygen saturation that is too low can increase the risk of
injury to the brain or other end organs (Deulofuet et al,
2006).
Adjusting inspired oxygen concentration to target lower
oxygen saturations in extremely preterm infants may
decrease the rate of severe retinopathy. In a prospective
observational study, ELBW infants treated in centers with
a restrictive approach to oxygen delivery (i.e., saturation
alarm limits of 70% to 90%) had less retinopathy requiring cryotherapy (6.3 versus 27.7 %) than did those in units
with a liberal approach (i.e., alarm limits of 88% to 98%),
and neurodevelopmental outcome at 1 year of age was
similar (Tin et al, 2001). In two other studies with historical controls, the incidence of severe retinopathy decreased
after oxygen saturation alarm limits were lowered from
87%-97% to 85%-93% for infants born at 28 weeks’ gestation and less until they reached 32 weeks postmenstrual
403
age (Chow et al, 2003; Vanderveen et al, 2006). In a
masked randomized trial comparing two different target
oxygen saturation ranges (85% to 89% compared with
91% to 95%) in infants born between 24 and 27 weeks,
ROP occurred less frequently in survivors assigned to the
lower oxygen saturation group. However, death before
discharge occurred more frequently in the lower oxygen
saturation group, suggesting that the best target range
for oxygen saturation remains unclear (SUPPORT Study
Group, 2010).
DEVELOPMENTAL AND PARENTAL CARE
ELBW infants are particularly vulnerable to the potentially noxious stimuli of the NICU environment, including light, noise, frequent disturbances, and painful
procedures. The ELBW infant reacts to the noisy and
well-lit environment of many NICUs with greater variability of blood pressure, ventilatory requirements, and
oxygen saturation as well as behavioral disorganization, which may have both short- and long-term effects
on outcome (Jacobs et al, 2002). Modification of the
NICU environment to limit exposure of ELBW infants
to such stresses—by lowering ambient light and reducing noise, clustering caregiving periods and procedures
to allow periods of uninterrupted sleep, and using positioning aids to promote containment—is an intuitive part
of their care. Newer NICU designs, transitioning from
open common rooms to private room settings, may also
facilitate a better environment for vulnerable infants and
enhance parental involvement. These environmental and
developmental interventions in the NICU can improve
physiologic stability and some short-term outcomes in
preterm infants, including decreased severity of BPD
and shorter length of hospital stay. It remains unclear
whether individualized, developmentally supportive
care or other developmental interventions started in the
NICU improve long-term outcome in ELBW infants
(Symington et al, 2009).
In addition to environmental modifications, NICUs
should promote parental involvement with infants, even
when they are critically ill. Open family presence guidelines and encouragement of parental caregiving when
appropriate may help parents to bond with their baby.
Many NICUs have embraced a philosophy of familycentered care, in which a stronger bond is forged with the
families of NICU patients by encouraging collaboration
among family members and care providers in policy and
program development, professional education, and aspects
of the delivery of care (Dunn et al, 2006). Skin-to-skin
(kangaroo) care, in which the infant is placed unclothed on
the mother or father’s bare chest, was originally developed
in nonindustrialized countries to maintain temperature
regulation in premature infants. It is now used in many
NICUs to promote parental attachment. Skin-to-skin care
may have a positive effect on infant state organization and
respiratory patterns, increase the rate of infant weight gain,
improve maternal milk production, and have long-term
benefits in infant development and parents’ perceptions of
their babies (Feldman et al, 2002). Skin-to-skin care can
be initiated in ELBW infants within the first 2 weeks after
birth, when they are more medically stable.
404
PART VIII Care of the High-Risk Infant
FUTURE DIRECTIONS
This chapter presents some of the special needs of the
ELBW infant. The medical care of the ELBW infant is
a complex combination of knowledge of developmental
physiology, evidenced-based interventions, and clinical
experience. Wide variability in approaches to care of these
infants exists among practitioners and NICUs, as does
variability in outcomes. Nevertheless, NICUs involved
in treating ELBW infants should develop a coherent
approach to the medical and ethical aspects of their care.
Future research should focus on identifying best practices
to narrow the variability in approach to care and with the
goal to prevent long-term disability. As more of these tiny
infants survive, it is the responsibility of neonatologists to
stay abreast of clinical improvements and the short- and
long-term consequences of established and newly proposed medical interventions to provide the best care for
these vulnerable infants and to keep parents informed and
involved.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
33
Care of the Late Preterm Infant
Sowmya S. Mohan and Lucky Jain
Had he been alive today, Patrick Bouvier Kennedy would
have been hailed as a triumph of neonatal care—after all,
he was the son of the former United States President, John
F. Kennedy and former First Lady Jacqueline B. Kennedy.
Born prematurely at 34 weeks gestation, he would have
been aptly labeled as a late preterm neonate; however,
based on his birth weight (2.1 kg) and gestational age,
few would have predicted the outcome he had then, were
he born in 2009. But then, in 1963, little was available
to the clinician for the management of hyaline membrane
disease—no routine use of neonatal ventilators, no device to
provide airway positive pressure, no surfactant, and no antenatal steroids. He died two days after his birth; the New
York Times obituary said that “the battle for the Kennedy
baby was lost because medical science has not advanced far
enough.”
(Jain and Carlton, 2009)
With more than 4 million live births per year (Martin
et al, 2007), the United States has one of the highest
birth rates among industrialized countries; it also has the
stigma of having a disproportionately high prematurity
rate. Decades of efforts to reduce preterm births have
not affected this formidable problem. In recent years the
problem has been highlighted by the rise in births between
34 and 366⁄7 weeks’ gestation, a group referred to as late
preterm infants (Figure 33-1). Late preterm infants have a
checkered history, having been passed off as nothing more
than “near term” infants, yet being feared as the “quick to
spiral down group” when they develop respiratory distress
syndrome or other complications. As the number of late
preterm infants has grown, so has the awareness of their
unique set of problems, such as delayed neonatal transition, wet lung syndrome, hypothermia, hypoglycemia, and
hyperbilirubinemia (Figure 33-2). Although not unique to
this population, these complications have sufficient differences in their manifestations and management, prompting
the editors to add an entirely new chapter to this textbook
devoted to the health issues of late preterm infants. In
Chapter 14, the obstetric issues and epidemiology related
to prematurity are addressed. This chapter focuses on the
special considerations applicable to the clinical course and
management of late preterm infants.
There has been a shift in the distribution of births
away from term and post term and toward earlier gestational ages (Davidoff et al, 2006). This shift has resulted
in a disproportionately high rate of premature births with
estimates of up to 12.7% of live births being premature
(Martin et al, 2007)—defined as <37 completed weeks’
gestation or <260 days, counting from the first day of the
last menstrual period (Raju, 2006). Within this group of
premature babies, up to 75% are classified as late preterm
infants (Adamkin, 2009). Although the reasons for such
a high number of late preterm births are multifactorial,
higher rates of induced deliveries, cesarean births, and
efforts to reduce stillbirths may have contributed to the
increase.
Late preterm babies currently account for up to one
third of all neonatal intensive care unit (NICU) admissions
in the United States (Angus et al, 2001), adding strain to
the overburdened system of health care delivery, particularly in community hospitals and rural areas. These admissions range from short stays, for problems such as transient
tachypnea of the newborn (TTNB), to more complicated
or extended NICU stays for problems such as persistent
pulmonary hypertension of the newborn (PPHN). With
the average NICU stay costing up to $3500 per day, the
economic impact of caring for the late preterm baby can
be significant. For example, in 1996 the State of California
alone could have saved $49.9 million in health care costs
by preventing non–medically indicated deliveries between
34 and 37 weeks’ gestation (Gilbert et al, 2003). In addition to the expense of the initial hospitalization, the cost of
caring for a late preterm baby can also be compounded by
the increased incidence of hospital readmissions and the
long-term care issues related to persistent problems. The
effects of the increasing number of late preterm births create a societal burden in lost productivity, as parents take
extended leave from work to be with their fragile newborns. More importantly, there may be lasting effects with
neurodevelopmental delays extending into early school
age. Because a significant proportion of brain growth
occurs during the last 6 weeks of gestation (Adams-Chapman, 2006), late preterm infants are vulnerable to neuronal injury and disruption of normal brain development.
Whereas more longitudinal studies are needed, preliminary studies show that late preterm infants are more likely
to have a diagnosis of developmental delay within the first
3 years of life, require special needs preschool resources,
and have more problems with school readiness (Morse
et al, 2009).
Given their large numbers, the overall socioeconomic
effects of the late preterm births can be significant. Strategies are required that can reduce the preventable fraction of late preterm births and work toward reducing the
morbidity in others, when continuation of the pregnancy
is deemed harmful to the fetus or the mother. This chapter explores the pathophysiology of the major morbidities
that affect late preterm infants and discusses the unique
challenges faced by clinicians in the management of these
conditions.
DEFINITION
Late preterm birth is an accepted term used for infants
born between 34 and 366⁄7 weeks’ gestation (see Figure
33-1) (Raju et al, 2006). This group of infants was initially
referred to as near term, but the misleading implication of
405
406
PART VIII Care of the High-Risk Infant
Late
preterm
First day of last
menstrual period
Day #
1
239
Early
term
259 260 274
BOX 33-1 C
haracteristics of Late Preterm
Infants
294
ll
ll
Week # 0/7
34
0/7
36
6/7
37 38
0/7 6/7
41
6/7
Term
Preterm
Post term
FIGURE 33-1 Definitions of late preterm and early term. (Adapted
from Engle WA, Kominiarek MA: Late preterm infants, early term infants,
and timing of elective deliveries, Clin Perinatol 35:325-341, 2008.)
CLINICAL OUTCOMES
Temperature
instability
Full term
Near term
Hypoglycemia
Intravenous
infusion
Respiratory
distress
ll
Late preterm infants—defined as born at 34 to 366⁄7 weeks’ gestation
Physiologically immature with limited compensatory responses to extrauterine environment compared to term infants
Greater risk than term infants for mortality and morbidities such as:
ll Temperature instability
ll Hypoglycemia
ll Respiratory distress
ll Apnea
ll Jaundice
ll Feeding difficulties
ll Dehydration
ll Suspected sepsis
Adapted from Engle WA, Tomashek KM, Wallman C: “Late preterm” infants: a population at
risk, Pediatrics 120:1390-1401, 2007.
physiological jaundice, late neonatal sepsis (Raju, 2006),
thermoregulation issues, hypoglycemia, feeding difficulties (Dudell and Jain, 2006; Escobar et al, 2006; Fuchs and
Wapner, 2006), and risk of injury to the developing brain,
which can lead to neurodevelopmental problems. These
problems account for a substantially higher number of
NICU admissions (see Figures 33-2 and 33-3).
PATHOPHYSIOLOGY AND CLINICAL
COURSE
Clinical
jaundice
0
10
20
30
40
50
60
(%)
FIGURE 33-2 Graph of clinical outcomes in near-term (35 to 366⁄7
weeks) and full-term infants as percentage of patients studied. (Adapted
from Wang ML, Dorer DJ, Fleming MP, et al: Clinical outcomes of nearterm infants, Pediatrics 114:372-376, 2004.)
maturity has prompted the name change to late preterm
(Box 33-1). This notion is further validated by recent studies showing that term infants born at 37 to 38 weeks’ gestation have higher morbidity and mortality than those born
at 39 weeks’ gestation (Hansen et al, 2008; Madar et al,
1999; McIntire and Leveno, 2008; Shapiro-Mendoza et al,
2008); this has prompted the use of early term to describe
births at 37 to 38 weeks’ gestation.
The term late preterm and the gestational age limits were
established by a panel of experts convened by the National
Institutes of Health and the National Institute of Child
Health and Human Development in 2005. While developing these criteria, the group considered many factors,
including the obstetric guidelines that consider 34 weeks
to be a maturational milestone. Beyond 34 weeks’ gestation, surfactant is generally considered to be adequate
and antenatal steroids are not offered to mothers with
anticipated delivery (Raju et al, 2006). Unlike the smaller,
more typical premature infant, late preterm infants appear
mature because of their larger size, but have a higher incidence of transient tachypnea of the newborn (McIntire
and Leveno, 2008; Wang et al, 2004), respiratory distress
syndrome (RDS) (Clark, 2005; Wang et al, 2004), PPHN
(Roth-Kleiner et al, 2003), respiratory failure, prolonged
Although many of the diseases discussed in this section are
not specific or unique to late preterm infants and are being
covered in other chapters in this book, it is important to
understand and recognize them as part of the unique challenge of caring for late preterm newborns.
RESPIRATORY
Several studies have consistently shown that late preterm
infants have higher respiratory morbidity and mortality
compared with full-term infants. Many late preterm infants
develop respiratory distress soon after birth (sustained distress for more than 2 hours after birth accompanied by
grunting, flaring, tachypnea, retractions, or supplemental oxygen requirement), which studies show occurs more
often in late preterm infants than in term newborns (28.9%
versus 4.2%, respectively) (Wang et al, 2004). In addition,
within the early term and late preterm groups, infants born
at 37 weeks’ gestation are fivefold more likely, and babies
born at 35 weeks’ gestation are ninefold more likely, to
have respiratory distress compared with babies born at 38
to 40 weeks’ gestation (Escobar et al, 2006). For each gestational week of age, infants delivered by elective cesarean
section tend to do worse (Figure 33-4). In fact, Madar et
al (1999) found that the incidence of respiratory distress
was significantly increased with every week of gestation
less than 39 weeks: 30 in 1000 infants born at 34 weeks’
gestation developed respiratory distress, 14 in 1000 born at
35 weeks’ gestation, and 7.1 in 1000 born at 36 weeks’ gestation. The etiology of respiratory distress is diverse and
includes transient tachypnea of the newborn, RDS, persistent pulmonary hypertension, and apnea. Not surprisingly, of the affected babies, the incidence of respiratory
407
CHAPTER 33 Care of the Late Preterm Infant
30000
Number of patients
25000
20000
15000
10000
5000
0
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
Estimated gestational age (wks)
3.5
3.3
PVD
ECD
EmCD
45
40
35
3.0
2.5
30
Percent of infants
Respiratory morbidity, % (95% CI)
50
FIGURE 33-3 Distribution of
neonatal intensive care unit (NICU)
admissions by gestational age,
highlighting the contribution made
by late preterm and early preterm
infants. Data were obtained from a
large consortium of NICUs under
a common management. (Adapted
from Clark RH: The epidemiology of
respiratory failure in neonates born at an
estimated gestational age of 34 weeks or
more, J Perinatol 25:251-257, 2005.)
25
20
15
10
5
2.0
1.5
1.0
0.5
0
34
35
36
37
38
39
40
41
42
43
GA stratum, wk
FIGURE 33-4 Respiratory morbidity in late preterm and early term
infants and the impact of mode of delivery. ECD, Elective cesarean
delivery; EmCD, emergency cesarean delivery; PVD, planned vaginal
delivery. (Adapted from De Luca R, Boulvain M, Irion O, et al: Incidence of
early neonatal mortality and morbidity after late-preterm and tern cesarean
section, Pediatrics 123:e1064-e1071, 2009.)
distress requiring mechanical ventilation corresponded to
the degree of prematurity: 3.3% of late preterm infants
born at 34 weeks’ gestation, 1.7% at 35 weeks’ gestation,
and 0.8% at 36 weeks’ gestation (Figure 33-5) (McIntire
and Leveno, 2008).
Whereas respiratory issues often tend to be transient in a
vast majority of these neonates, some develop into PPHN
or severe hypoxic respiratory failure requiring additional
therapies such as nitric oxide, high frequency ventilation,
and extracorporeal membrane oxygenation (ECMO; Heritage and Cunningham, 1985; Keszler et al, 1992). Studies
have shown that pulmonary hypertension is more likely in
preterm infants (born at 34 to 37 weeks’ gestation) who
develop RDS than in similar infants born at 32 weeks’ gestation. Such predisposition is attributed to a developmental
increase in smooth muscle in the walls of pulmonary blood
1.7
0.8
0.3
0
34 wks
35 wks
36 wks
39 wks
FIGURE 33-5 Percentage of infants born at late preterm gestations
who require mechanical ventilation. (Adapted from McIntire DD, Leveno
KJ: Neonatal mortality and morbidity rates in late preterm births compared
with births at term, Obstet Gynecol 111:35-41, 2008.)
vessels. PPHN is associated with increased pulmonary vascular resistance that eventually leads to right-to-left shunting by means of fetal pathways and ventilation-perfusion
mismatching (Dudell and Jain, 2006). Management of
neonates who develop significant pulmonary hypertension
can be challenging, given the self propagated nature of
hypoxia-induced pulmonary vasoconstriction. Treatment
options include exogenous surfactant (shown to be effective if used earlier in the disease course; Dudell and Jain,
2006), inhaled nitric oxide (selectively lowers pulmonary
vascular resistance and decreases extrapulmonary right-toleft shunting) (Kinsella et al, 1992, 1993), high-frequency
ventilation, and ECMO.
A review of the Extracorporeal Life Support Organization Neonatal Registry from 1989 to 2006 by Dudell and
Jain (2006) found that 14.5% of the ECMO patients during that time period were late preterm infants and had a
mean gestational age of 35.3 weeks. Interestingly, affected
408
PART VIII Care of the High-Risk Infant
Alveolus
AMILORIDE
Na
Na, K
Cl
ENaC
NSC
Paracellular
Na, K, Cr
H2O
ENaC
HSC
CNGC
Na, K
AQP5
ENaC ENaC
HSC NSC
Na Na, K H2O
AQP5
T1 cell
Na
Interstitium
K
Na, K-ATPase
Cl
CFTR
Paracellular
Na, K, Cl
T2 cell
CT
CLC
OUABAIN
Na
K
Na, K-ATPase
Pulmonary capillary
FIGURE 33-6 Epithelial sodium (Na) absorption in the fetal lung near birth. Na enters the cell through the apical surface of both ATI and
ATII cells via amiloride-sensitive epithelial Na channels (ENaC), both highly selective channels (HSC) and nonselective channels (NSC), and cyclic
nucleotide gated channels (seen only in ATI cells). Electroneutrality is conserved with chloride movement through cystic fibrosis transmembrane
conductance regulator (CFTR) or through chloride channels (CLC) in ATI and ATII cells, and/or paracellularly through tight junctions. The increase
in cell Na stimulates Na-K-ATPase activity on the basolateral aspect of the cell membrane, which drives out three Na ions in exchange for two K
ions, a process that can be blocked by the cardiac glycoside ouabain. If the net ion movement is from the apical surface to the interstitium, an osmotic
gradient would be created, which would in turn direct water transport in the same direction, either through aquaporins or by diffusion. (From Jain L:
Respiratory morbidity in late-preterm infants: prevention is better than cure! Am J Perinatol 25:75-78, 2008.)
infants were more likely to require ECMO secondary to
hypoxic respiratory failure or RDS instead of aspiration
syndromes, which is the primary insult for term infants
requiring ECMO (Dudell and Jain, 2006). In addition, late
preterm infants were older at cannulation (likely because
most of these infants are asymptomatic at birth, but gradually develop an increasing oxygen requirement with subsequent development of PPHN), had a longer duration of
ECMO support, and were more likely to have intraventricular hemorrhage and other neurologic complications
than term infants. Overall survival rate was significantly
lower (74%) for late preterm infants compared with term
infants (87%) (Dudell and Jain, 2006).
Why is it that, even in situations in which amniotic
fluid testing shows a mature surfactant profile, late preterm infants are at risk for developing respiratory distress?
Part of the answer lies in the delay in clearing fetal lung
fluid. Throughout much of gestation, fetal lungs actively
secrete fluid into alveolar spaces via a chloride secretory
mechanism. This process can be blocked by inhibitors of
Na-K-2Cl co-transport. The fluid that accumulates in the
developing lung plays a critical role by providing a structural template that prevents the collapse of the developing
lung and promotes its growth. At the time of delivery, the
lung epithelium becomes integral in the process of switching from placental to pulmonary gas exchange (Bland, 2001;
Jain, 1999). For effective gas exchange to occur in the lungs,
alveolar spaces must be cleared of excess fluid, and pulmonary blood flow must be increased to match ventilation
with the perfusion that is taking place. If either the ventilation or perfusion is inadequate, the infant will have a difficult time transitioning and will develop respiratory distress.
In addition, during fetal development, many abnormalities
can occur and interfere with the normal production of this
lung fluid. Some problems during development include
pulmonary artery occlusion, diaphragmatic hernia, and
uterine compression of the fetal thorax from chronic leak
of amniotic fluid. All these conditions inhibit normal lung
development and growth (Jain and Eaton, 2006).
Although a small role in the clearance of this fluid can be
attributed to Starling forces and “vaginal squeeze” (Bland,
2001; Jain, 1999), amiloride-sensitive sodium transport by
lung epithelial cells through epithelial sodium channels
(ENaCs) has emerged as a key event in the transepithelial movement of alveolar fluid (Figure 33-6) (Bland, 2001;
Jain et al, 2001). Research has shown that these ENaCs
orchestrate the clearing of fluid from the fetal lungs, and
disruption of their function has been implicated in several
disease processes affecting the newborn, including transient tachypnea of the newborn and hyaline membrane
disease. The late preterm infant is more susceptible to
these problems, in part because ENaC expression is developmentally regulated and peak expression in the alveolar
epithelium is achieved only at term gestation, which leaves
the preterm infant with lower expression of these channels, thus reducing their ability to clear fetal lung fluid
after birth (Smith et al, 2000).
High doses of glucocorticoids have been shown to
stimulate transcription of ENaCs in several sodium transporting epithelia and in the lung (Tomashek et al, 2007).
In the alveolar epithelia, glucocorticoids were found to
induce lung sodium reabsorption in the late gestation fetal
lung (Tomashek et al, 2007). In addition to increasing
transcription of sodium channel subunits, steroids increase
the number of available channels, by decreasing the rate
at which membrane-associated channels are degraded, and
increase the activity of existing channels. Glucocorticoids
have also been shown to enhance the responsiveness of
lungs to β-adrenergic agents and thyroid hormones (Venkatesh and Katzberg, 1997).
In addition to problems with lung fluid clearance,
several other factors may contribute to the overall burden of respiratory morbidity (Hansen et al, 2008; Kolas
et al, 2006; Levine et al, 2001; Morrison et al, 1995;
Roth-Kleiner et al, 2003; Villar et al, 2007). Given
the shortcomings clinicians face in accurate estimation of gestational age, elective induction and cesarean
section may have increased the burden of iatrogenic
CHAPTER 33 Care of the Late Preterm Infant
prematurity. In an attempt to minimize the occurrence
of iatrogenic RDS in light of the increasing frequency
of elective cesarean sections—commonly performed
between 37 and 40 weeks’ gestation (Hales et al, 1993)—
fetal lung maturity testing was recommended before
elective cesarean sections. Because of the risks and complications associated with amniocentesis, this testing is
done infrequently (Dudell and Jain, 2006), especially in
light of recent studies showing that even late preterm
infants and some early term infants born by cesarean section before the onset of labor have respiratory distress
despite having mature surfactant profiles. This finding prompted the American College of Obstetrics and
Gynecologists (2002) to recommend scheduling elective
cesarean section at 39 weeks or later or waiting for the
onset of spontaneous labor, but unfortunately factors
related to the convenience of scheduled elective cesarean section deliveries for both families and providers will
continue to influence the timing of elective cesarean section (Dudell and Jain, 2006).
GASTROINTESTINAL
Nutrition
Feeding problems are one of the primary reasons for delay
in the discharge of late preterm infants (Adamkin, 2006).
Late preterm infants often have poor coordination of
sucking and swallowing because of neuronal immaturity,
decreased oromotor tone, and inability to generate adequate intraoral pressures during sucking (Engle et al, 2007;
Kinney, 2006; Polin et al, 2003; Raju et al, 2006). Breastfeeding has also been shown to be more difficult for early
term or late preterm infants compared with term infants
(Raju, 2006). These problems can lead to poor caloric
intake and dehydration.
These problems are compounded by the variations
in practice and nutritional management of these infants,
given the paucity of published studies in this regard. Recent
studies have shown that issues such as hypoglycemia and
poor feeding contributed to 27% of all late preterm babies
requiring intravenous fluids, compared with only 5% of
their term counterparts (Wang et al, 2004). In the face of
poor enteral intake, TPN is indicated and can become an
important therapy in the care of the late preterm infant,
but is often delayed in anticipation of a quick recovery
(Adamkin, 2006). The challenge then becomes providing adequate nutrition to support growth and equate the
energy expenditure that can occur when the infant faces
issues such as hypothermia, sepsis, and respiratory distress,
which are often seen in late preterm infants. Studies show
that the energy expenditure of nongrowing low-birthweight infants (birthweight less than 2500 g) is 45 to 55 cal/
kg/day (Adamkin, 2006). These calories come from several
sources in the TPN including amino acids and lipids.
Late preterm infants are more adept at handling amino
acids, allowing the protein content in TPN to be started
at 2 g/kg/day. With a protein intake of 2.5 to 3 g/kg/day
(with adequate caloric intake), a late preterm infant can
achieve weight gain similar to a term infant fed human
milk (Adamkin, 2006). More controversial is the use
of intravenous lipids in late preterm infants. Of the late
409
preterm infants with respiratory distress or disease, there
are two subgroups: infants with parenchymal lung disease
without increased pulmonary vascular resistance (PVR)
and those with signs of PPHN or increased PVR (Adamkin, 2006). The concern over the use of lipids in the late
preterm infant with lung disease stems from adult studies
showing that failure to clear infused lipids has an adverse
effect on gas exchange in the lungs (Greene et al, 1976).
Contrary to those findings, preterm neonates randomized
to different lipid infusion rates did not demonstrate any
effect on alveolar-arterial oxygen gradient, arterial blood
pH, or oxygenation when randomly assigned to modest
doses of lipids (0.6 to 1.4 g/kg/day) over the first week of
life (Adamkin, 2006). The other argument for restricted
use of lipids in late preterm infants specifically addresses
the infants with increased PVR and respiratory disease.
The concern is that the high polyunsaturated fatty acid
content of lipid emulsions (with excess omega 6-linoleic
acid) feeds into the arachidonic acid pathways, leading to
synthesis of prostaglandins and leukotrienes, which can
increase vasomotor tone and result in hypoxemia (Adamkin, 2006). Despite the lack of firm evidence for the effects
of lipid emulsions in infants with severe respiratory failure with or without pulmonary hypertension, the recommendation is that infants with respiratory disease, but not
increased PVR, should receive adequate lipids to prevent
essential fatty acid deficiency; in infants with elements of
PPHN, lipids should be avoided during the critical stages
of their illness (Adamkin, 2006). Because of these issues
and other concerns about parenteral nutrition (e.g., difficult in optimizing nutrition, the need for intravenous
access and the potential for infiltrates or infection, risk
of cholestatic jaundice with prolonged use of parenteral
nutrition) enteral feeds should be started as soon as clinically possible while the infant is slowly weaned from the
parenteral nutrition.
In general, nutritional experts recommend that 34- and
35-week late preterm infants receive nutrient-enriched
(22 kcal/oz) milk, whereas older 36- and 37-week late
preterm infants with an uncomplicated neonatal course
be fed unfortified milk after discharge (Adamkin, 2006).
These nutrient-enriched formulas have a higher protein
content (1.9 versus 1.4 g/dL), increased energy (22 versus
20 kcal/oz), additional calcium, phosphorous, zinc, trace
elements, and vitamins compared with standard formulas
(Adamkin, 2006). This enrichment becomes essential to
the late preterm infant who was born at 34 to 35 weeks’
gestation or the older group of late preterm infants who
had a difficult NICU course, where the goal is to compensate for earlier deprivation of adequate nutrition and
allow for somatic and brain growth during the first year
of life. These issues notwithstanding, these nutritional
guidelines are not always followed, leading to variability
in nutritional practices by providers. One study showed
that, although nearly 46% of late preterm infants were discharged home with recommendations to use formula that
contained more than 20 kcal/oz, this practice recommendation had a broad range of followers (4% to 72%) (Adamkin, 2006). The issue becomes more pressing in the late
preterm infant with chronic conditions, such as bronchopulmonary dysplasia, that are often associated with growth
failure caused by inadequate nutrient intake.
410
PART VIII Care of the High-Risk Infant
BOX 33-2 Causes of Hypoglycemia in the Late Preterm Infant
TRANSIENT HYPOGLYCEMIA IN THE LATE PRETERM INFANT
Maternal Conditions
ll Glucose infusion in the mother
ll Preeclampsia
ll Drugs: tocolytic therapy, sympathomimetics
ll Infant of diabetic mother
Neonatal Conditions
ll Prematurity
ll Respiratory distress syndrome
ll Twin gestation
ll Neonatal sepsis
ll Perinatal hypoxia-ischemia
ll Temperature instability: hypothermia
ll Polycythemia
ll Specific glucose transporter deficiency
ll Isoimmune thrombocytopenia, Rh incompatibility
PERSISTENT HYPOGLYCEMIA IN THE LATE PRETERM INFANT
Endocrine Disorders
ll Pituitary insufficiency
ll Cortisol deficiency
ll Congenital glucagon deficiency
Inborn Errors of Metabolism
Carbohydrate metabolism: glycogen storage disease, galactosemia, fructose1,6-diphoshatase deficiency
ll Amino acid metabolism: maple syrup urine disease, propionic academia,
methylmalonic academia hereditary tyrosinemia
ll Fatty acid metabolism: acyl-coenzyme dehydrogenase defect, defects in
carnitine metabolism, beta-oxidation defects
ll Defective glucose transport
ll
From Garg M, Devaskar SU: Glucose metabolism in the late preterm infant, Clin Perinatol 33:853-870, 2006. Reprinted with permission.
For mothers who choose to breastfeed their late preterm infant, it can often be more challenging compared
with nursing a full-term infant. The challenge often lies
in initiating and establishing breastfeeding because these
infants are sleepier; have less stamina; have more difficulty maintaining body temperature; have problems with
latching, sucking, and swallowing; and have more respiratory instability than full-term infants (Adamkin, 2006).
Despite these obstacles, mothers should still be encouraged to provide breast milk given the numerous proven
benefits of breast milk. In fact, recent studies have shown
that the advantages of breast milk feeding for premature
infants might be even greater than those for term infants
(Adamkin, 2006).
Hypoglycemia
Hypoglycemia is defined as low circulating glucose concentrations, but the actual neonatal threshold value is still
debated. A physiologic definition was established, based
on abnormal electroencephalograms at glucose levels that
were lower, with a glucose level less than 45 mg/dL being
considered hypoglycemia (Koh et al, 1988). Hypoglycemia
is often missed in late preterm infants, mainly because of
the early transition of these infants to the well baby nursery or the mother’s room in an effort to triage the limited
number of acute care beds in the NICU and to allow the
mother to bond with her new baby (Garg and Devaskar,
2006). However, developmental immaturity is associated
with multiple problems, including decreased glycogen
stores and feeding difficulties, both of which can lead to
hypoglycemia (Box 33-2). Not surprisingly, the incidence
of hypoglycemia in preterm infants is threefold greater than
in full-term infants (Wang et al, 2004). In addition, severe
hypoglycemia is a well-known risk factor for neuronal cell
death and adverse neurodevelopmental outcomes (Garg
and Devaskar, 2006). Therefore, if the hypoglycemia is not
recognized and treated in a timely manner with intravenous
fluids or feedings, the infant can develop neurodevelopmental abnormalities because the compensatory mechanisms for protecting the brain from hypoglycemia are not
fully developed (Cornblath and Ichord, 2000; Cornblath
et al, 2000; Rozance and Hay, 2006; Vannucci and Vannucci, 2001). Therefore early recognition, diagnosis, and
treatment of hypoglycemia are crucial to the late preterm
infant’s long-term outcome. It is recommended that institutions develop protocols for routine testing of blood sugars
in late preterm infants. One can use existing serum glucose
screening protocols for infants at high risk for hypoglycemia (i.e., small for gestational age, large for gestational
age, infant of a diabetic mother). If none are available,
the following is recommended: glucose checks between
1 and 2 hours after birth, followed by testing before the
next three consecutive feeds and then before alternate
feedings for the remainder of the first 24 hours. If the
blood sugar is less than 40 to 45 mg/dL, the hypoglycemia
protocol should be followed for management.
Hypoglycemia is not a problem in utero, because the
fetus receives a steady supply of glucose primarily by maternal transfer through the placenta. Once the baby is delivered, this constant supply of glucose is abruptly stopped and
the infant has to rely on glucose production primarily via
hepatic glycogenolysis and gluconeogenesis (Halamek et al,
1997). After birth, the baby experiences a surge in catecholamines, glucagon, and corticosteroids, which play a key
role in maintaining a euglycemic state. The increase in catecholamines leads to a surge in glucagon concentration and
a decline in circulating insulin concentrations, which both
contribute to maintaining a normal serum glucose level.
Glucose levels are also affected by the unregulated insulin
production by the immature pancreatic β cells (Garg and
Devaskar, 2006). As a result, the late preterm newborn can
experience significant hypoglycemia secondary to developmentally immature hepatic enzyme systems for gluconeogenesis, glycogenolysis, and hormonal dysregulation (Engle
et al, 2007; Garg and Devaskar, 2006; Raju et al, 2006).
The neonatal glucose requirement is 6 to 8 mg/kg/min,
which is a higher value than that observed in adults (3 mg/
kg/min) (Bier et al, 1977). This demand for glucose
increases if the late preterm infant has coexisting conditions such as sepsis, birth asphyxia, or cold stress (Greisen and Pryds, 1989; Halamek et al, 1997; Halamek and
CHAPTER 33 Care of the Late Preterm Infant
Stevenson, 1998). Treatment options for hypoglycemia
in the late preterm infant include establishing early feeds
(supplementing with formula if quantity of breast milk is
insufficient), glucose infusion through intravenous fluids,
hydrocortisone, glucagon, epinephrine, diazoxide, and
octreotide (Garg and Devaskar, 2006). The treatment
choice for the late preterm infant is based on the underlying cause of the hypoglycemia. Regardless of the etiology of the hypoglycemia, it is important to have constant
monitoring of the glucose levels until they stabilize and the
baby is tolerating adequate nutrition.
Hyperbilirubinemia
Hyperbilirubinemia is the most common clinical condition requiring evaluation and treatment in the late
preterm newborn and the most common cause for readmission during the first postnatal week of life (Bhutani
et al, 2004; Brown et al, 1999; Escobar et al, 2005; Maisels
and Kring, 1998). Studies show that late preterm infants
are more likely than term infants to be rehospitalized
for jaundice (4.5% versus 1.2% in term infants) (Escobar
et al, 2005).
In general, neonatal hyperbilirubinemia in late preterm
infants is more prevalent, more pronounced, and more
protracted than in their term counterparts. Important risk
factors for severe jaundice are summarized in Figure 33-7,
A. A study by Newman et al (1999) showed that infants
born at 36 weeks’ gestation have an eightfold increase in
the risk of developing a total serum bilirubin concentration greater than 20 mg/dL (343 μmol/L) when compared
with those born at 41 weeks’ gestation or later. Part of the
reason for this increased risk is the immature hepatic metabolic pathways for bilirubin and the overall immaturity
of gastrointestinal function and motility. The decreased
ability for hepatic uptake and conjugation puts the late
preterm infant at increased risk of elevated serum bilirubin levels, and the jaundice then becomes more prolonged,
prevalent, and severe (Bhutani and Johnson, 2006). In
addition, late preterm infants are at increased risk of kernicterus at bilirubin levels equal to or lower than that of
term infants (Bhutani and Johnson, 2006). Kernicterus is
a devastating, chronic, and disabling condition characterized by the tetrad of choreoathetoid cerebral palsy, neural hearing loss, palsy of vertical gaze, and dental enamel
hypoplasia (Watchko, 2006).
Whereas the need for universal predischarge bilirubin
testing in neonates is debated, it is generally accepted that
late preterm infants are at higher risk and should not be
included with term infants. The current recommendation
from the American Academy of Pediatrics from 2004 for
the management of hyperbilirubinemia recommends that
all newborns be assessed for their risk of developing hyperbilirubinemia by using predischarge total serum bilirubin
or transcutaneous bilirubin measurements (Kuzniewicz
et al, 2009). The effectiveness of this policy was studied by
Kuzniewicz et al (2009), and they concluded that universal
bilirubin screening, whether using transcutaneous bilirubin or total serum bilirubin for measurements, was associated with increased identification of newborns needing
phototherapy and a significantly lower incidence of severe
hyperbilirubinemia (see Figure 33-7, B and C).
411
This finding underscores the need for close monitoring
of late preterm infants, particularly breastfed infants whose
mothers may not have a proper milk supply before being
discharge home. Early discharges should be avoided until
proper feeding has been established, and early follow-up
should be arranged.
INFECTIOUS DISEASES
Late preterm infants are also more susceptible to infections because of their immunologic immaturity. The
timing of the infection categorizes them as congenital
(acquired before delivery), early onset (usually acquired
during delivery and presenting within the first 72 hours),
or late onset (often acquired in the hospital and presents
after 72 hours of life). Congenital infections are commonly
attributed to rubella, cytomegalovirus, herpes simplex
virus, and HIV (Benjamin and Stoll, 2006). The severity
of the infection and its effect on the late preterm infant
depends on the stage of pregnancy at which the maternal infection occurred. With both herpes simplex virus
and HIV, maternal-infant transmission is more frequent
if a mother has a primary infection at the time of delivery, whereas maternal viral load is the main risk factor for
HIV transmission from mother to newborn (Benjamin and
Stoll, 2006).
Early-onset sepsis is almost always caused by perinatally
acquired infections. In most cases, the late preterm infant
is initially colonized by exposure to various organisms in
the maternal genital tract including group B Streptococcus
(GBS), Escherichia coli, and Candida spp. Additional risk
factors for developing sepsis are prolonged rupture of
membranes (greater than 18 hours), maternal fever, and
chorioamnionitis (Centers for Disease Control and Prevention, 2002).
The third group—the late-onset sepsis—may be caused
by perinatally or postnatally acquired organisms, but usually is a consequence of nosocomial transmission. The
most common organisms are gram-negative rods, but sepsis could be caused by Staphylococcus aureus, Enterobacter
spp., or Candida spp. Although the mortality rate is low for
late preterm infants, infections increase the risk of complications and often involve longer hospital stays (Benjamin
and Stoll, 2006).
In addition, research shows that late preterm infants
undergo testing for sepsis more often than term infants
(36.7% versus 12.6%; odds ratio, 3.97; 95% confidence
interval, 1.82 to 9.21; p = 0.00015) and receive antibiotics
more often and for a longer duration (7-day course 30%
versus 17% in term infants) (Wang et al, 2004). Other
studies show that the need for a sepsis evaluation increases
with decreasing gestational age; 33% were evaluated for
possible sepsis at 34 weeks’ gestation compared with 12%
at 39 weeks’ gestation (p <0.01), of which only 0.4% of
infants had culture-proven sepsis (McIntire and Leveno,
2008). This higher frequency of screening late preterm
newborns for sepsis compared with term infants may be
multifactorial. First, records show that one third of all
preterm deliveries occur after prolonged premature rupture of membranes, which can put the newborn at a significantly higher risk of infection. In addition, this higher
rate of sepsis workups in the late preterm baby may be
412
PART VIII Care of the High-Risk Infant
A
IMPORTANT RISK FACTORS FOR SEVERE HYPERBILIRUBINEMIA
• Predischarge total serum bilirubin (TSB) or transcutaneous bilirubin (TcB) measurement in
the high-risk or high-intermediate-risk zone
• Lower gestational age
• Exclusive breastfeeding, particularly if nursing is not going well and weight loss is excessive
• Jaundice observed in the first 24 h
• Isoimmune or other hemolytic disease (e.g., G6PD deficiency)
• Previous sibling with jaundice
• Cephalohematoma or significant bruising
• East Asian race
HOURLY PROGRESSION OF TOTAL SERUM BILIRUBIN (TSB) LEVELS:
RISK CATEGORIES
25
428
20
342
15
257
10
171
5
Infants at lower risk (38 wk and well)
Infants at medium risk (38 wk risk factors or 35–37 6/7 wk and well)
Infants at higher risk (35–37 6/7 wk risk factors)
0
mol/L
TSB, mg/dL
B
85
0
Birth
24 h
48 h
72 h
96 h
5d
6d
7d
Age
C
GUIDELINES FOR PHOTOTHERAPY IN HOSPITALIZED INFANTS 35 WEEKS’ GESTATION
Note that these guidelines are based on limited evidence and that the levels shown are
approximations. The guidelines refer to the use of intensive phototherapy, which should be
used when the TSB level exceeds the line indicated for each category.
• Use total bilirubin. Do not subtract direct-reacting or conjugated bilirubin.
• Risk factors are isoimmune hemolytic disease, G6PD deficiency, asphyxia, significant lethargy,
temperature instability, sepsis, acidosis, or an albumin level of 3.0 g/dL (if measured).
• For well infants at 35 to 37 6/7 weeks’ gestation, on can adjust TSB levels for intervention
around the medium-risk line. It is an option to intervene at lower TSB levels for infants closer
to 35 weeks’ gestation and at higher TSB levels for those closer to 37 6/7 weeks’ gestation.
• It is an option to provide conventional phototherapy in the hospital or at home at TSB levels
of 2 to 3 mg/dL (35–50 mol/L) below those shown, but home phototherapy should not be
used in any infant with risk factors.
FIGURE 33-7 Hyperbilirubinemia. A, Risk factors. B, Hourly progression of total serum bilirubin levels. C, Management guidelines. (Adapted
from Maisels MJ, Bhutani VK, Bogen D et al: Hyperbilirubinemia in the newborn infant > or = 35 weeks’ gestation: an update with clarifications, Pediatrics
124:1193-1198, 2009.)
a reflection of a standard protocol used for admissions
to the NICU or due to their clinical presentation (e.g.,
respiratory distress, hypothermia, hypoglycemia), which
could be a sign of sepsis or a reflection of the infants’
immaturity.
THERMOREGULATION
Because of their relatively smaller size, the late preterm
infant is susceptible to periods of hypothermia or cold stress.
Unfortunately, as for other problems discussed earlier, this
may be difficult to assess if the late preterm newborn has
413
CHAPTER 33 Care of the Late Preterm Infant
been sent to the mother’s room or is not closely observed.
Usually cold stress will manifest as tachypnea or apnea,
poor feeding, poor color caused by peripheral vasoconstriction, and metabolic acidosis. Hypothermia and its
related consequences can delay the respiratory transition
and exacerbate hypoglycemia; these signs and symptoms
may also be misinterpreted as possible sepsis, which then
leads to unnecessary interventions and workups.
The reason that late preterm infants are particularly
more susceptible to temperature instability is because of
their physiologic immaturity of thermoregulation, which
in turn is dependent on three main things: the amount of
brown adipose tissue, white adipose tissue, and body surface area (Engle et al, 2007; Martin et al, 2006; Polin et
al, 2003). Nonshivering thermogenesis is controlled by the
hypothalamic ventromedial nucleus through the sympathetic nervous system, which releases the neurotransmitter norepinephrine. The norepinephrine then causes the
brown adipose tissue to liberate free fatty acids, which are
eventually oxidized and produce heat (Engle et al, 2007;
Martin et al, 2006; Polin et al, 2003). Late preterm infants
have decreased stores of brown adipose tissue and the hormones responsible for brown fat metabolism (i.e., prolactin, norepinephrine, triiodothyronine, and cortisol). These
hormones peak at term gestation and the late preterm
infant misses those last few weeks of in utero development
(Engle et al, 2007; Polin et al, 2003). In addition to the
decreased stores of hormones leading to thermogenesis,
late preterm infants also have problems with hypothermia
because of a decreased amount of white adipose tissue,
which leads to less insulation, and their smaller size. The
late preterm infant’s relatively smaller size, compared with
term infants, leads to an increased ratio of surface area to
body weight, which allows for greater heat loss to the environment (Engle et al, 2007; Martin et al, 2006; Polin et
al, 2003). Appropriate monitoring and triaging of the late
preterm infant who is susceptible to temperature instability can avoid unnecessary morbidity, workups, interventions, and prolonged hospitalizations.
NEURODEVELOPMENTAL
During pregnancy, the infant’s lungs and brain are
among the last organs to mature and are therefore more
prone to injury. Not surprisingly, research has shown
that even healthy near-term or late preterm infants are
at risk for developmental delays through the first 5 years
of life (Raju, 2006). During the final few weeks of gestation, many aspects of brain maturity are still in progress.
These aspects include maturing oligodendroglia, increasing neuronal arborization and connectivity, maturation
of neurotransmitter systems, and continued brain growth
that accounts for a 30% increase in brain size during the
last few weeks of gestation (Figure 33-8) ( Jain and Raju,
2006). At 34 weeks’ gestation, the brain weighs only 65%
of the weight of a 40-week term infant (Billiards et al,
2006; Kinney, 2006). Researchers have shown that the
brain of a late preterm infant is still immature and continues to grow until 2 years of age, when it reaches 80%
of adult brain volume. In addition, the cerebral cortex is
still smooth and the gyri and sulci are not fully formed,
and myelination and interneuronal connectivity is still
20 weeks
35 weeks
40 weeks
Near-term
FIGURE 33-8 The immaturity of the laminar position and dendritic
arborization of neurons, as demonstrated by Golgi drawings, in the
cerebral cortex in the late preterm infant at 35 weeks’ gestation is striking in comparison with neurons at mid-gestation (20 weeks) and at term
(40 weeks). (From Kinney HC, Armstrong DD: Perinatal neuropathology.
In Graham DI, Lantos PE, editors: Greenfield’s neuropathology, ed 7,
London, 2002, Arnold, pp 557-559.)
incomplete. Multiple insults during this critical phase of
neuronal and glial maturation cause white and grey matter injury, especially in the thalamic region and the periventricular white matter (Kinney, 2006). These events can
be correlated subsequently to delayed development and
special education needs; therefore it is important to start
early developmental follow-up, anticipatory guidance, and
interventions for infants born at 32 to 36 weeks’ gestation
(Chyi et al, 2008).
HOSPITALIZATION OF THE LATE PRETERM
INFANT
Based on the need for close monitoring and management
of the various medical problems identified in late preterm
infants, they are more likely than a full-term infant to
require admission to an intensive care unit. Despite this
finding, individual hospitals and nurseries follow different
criteria regarding which infants to admit to the NICU,
an intermediate care unit, or an observation area. Some
routinely admit all infants <35 weeks, gestation to the
NICU, whereas others do so on an individual basis. The
most common reasons for admission include temperature
instability, jaundice, respiratory distress, dehydration, poor
feeding, and hypoglycemia (Vachharajani and Dawson,
2009; Wang et al, 2004). Studies have shown that 88% of
infants born at 34 weeks’ gestation, 12% born at 37 weeks’
gestation, and 2.6% born at 38 to 40 weeks’ gestation were
admitted to the NICU (Engle and Kominiarek, 2008).
Other studies have shown similar rates, and the overall
trend was that the late preterm infant had significantly
higher rates of NICU admission than the 39-week infants
(McIntire and Leveno, 2008). In addition, the duration of
hospitalization for the late preterm infant is inversely proportional to the baby’s gestational age, which means that
late preterm infants require longer hospitalization after
birth than their term counterparts. On average, infants are
hospitalized for 6 to 11 days at 34 weeks’, 4 to 6 days at
35 weeks’, and 3 to 4 days at 36 weeks’ gestation (Escobar
414
Early Neonatal
Mortality
(1-7 days)
Infant Mortality
(1-365 days)
Gestational
Age (weeks)
Mortality
Rate
Risk
Ratio
Mortality
Rate
Risk
Ratio
34
7.2
25.5
12.5
10.5
35
4.5
16.1
8.7
7.2
36
2.8
9.8
6.3
5.3
37
0.8
2.7
3.4
2.8
38
0.5
1.7
2.4
2.0
39
0.2
0.8
1.2
1.2
From Young PC, Glasgow TS, Li X, et al: Mortality of late-preterm (near-term)
newborns in Utah, Pediatrics 119:e659-e665, 2007.
et al, 2005; Gilbert et al, 2003; Khashu et al, 2009; McIntire and Leveno, 2008; Phibbs and Schmitt, 2006; Vachharajani and Dawson, 2009).
MORTALITY
Preterm birth is consistently recognized as the most pressing public health problem in perinatology by both clinicians
and researchers, given its overall contribution to infant
mortality. It also contributes substantially to neurocognitive, pulmonary, and ophthalmologic morbidity (Kramer
et al, 2000). On review of infant birth and death files from
1995 to 2002 in the United States, Tomashek et al (2007)
compared the overall and cause-specific mortality rates
between singleton late preterm infants and term infants.
They found that, despite significant declines since 1995 in
mortality rates for late preterm and term infants, the infant
mortality rate in 2002 was threefold higher in late preterm
infants than in term infants (7.9 versus 2.4 deaths per 1000
live births); early, late, and postneonatal mortality rates
were sixfold, threefold, and twofold higher, respectively.
Another study by Young et al (2007) found that in a large
cohort from Utah, the relative risk of death increased for
every decreasing week in gestational age less than 40 weeks
(Table 33-1). In addition, a large study involving 133,022
infants born at 34 to 40 weeks’ gestation found that neonatal mortality rates were significantly higher for late preterm infants (1.1, 1.5, and 0.5 per 1000 live births at 34, 35,
and 36 weeks, respectively, compared with 0.2 per 1000
live births at 39 weeks’ gestation p <0.001) (McIntire and
Leveno, 2008). In 2005, the U.S. infant mortality rate for
late preterm infants was 7.3 versus 2.43 per 1000 live births
in term neonates. Surprisingly, the mortality rate is 30%
higher even for infants born between 37 and 39 weeks’
gestation (early term) (Mathews and MacDorman, 2008).
These studies and statistics again emphasize the fact that
infants born just a few weeks early are at a much greater
risk of morbidity and mortality than those born at term
gestation. However, perinatal data collected over similar
periods also reveal a remarkable decline in stillbirth rate
(Figure 33-9). Perinatologists argue that this reduction in
fetal demise is directly related to close monitoring of the
fetus and early intervention (delivery) when needed.
10
Infant deaths
9
9
Late preterm births
8
8
7
7
Stillbirths
6
6
0
infant deaths per 1000 births
at Late Preterm and Early Term Gestations
10
Stillbirths and
TABLE 33-1 Mortality (rate per 1000 live births) in Infants Born
Percent of late preterm births (34–36 weeks)
PART VIII Care of the High-Risk Infant
0
1990
1995
2000
2005
Year
FIGURE 33-9 Trends in late preterm birth, stillbirth, and infant
mortality in the United States, 1990 to 2004. The left axis shows
trends in stillbirth and infant mortality rates; the right axis shows trends
in late preterm births (34 to 36 weeks). Late preterm birth rates are
shown per 100 live births; stillbirth rates per 1000 total births, and
infant death rates per 1000 live births. (Adapted from Ananth CV, Gyamfi
C, Jain L: Characterizing risk profiles of infants who are delivered at late preterm gestations: does it matter? Am J Obstet Gynecol 199:329-331, 2008.)
RECOMMENDATIONS
ADMISSION CRITERIA
With more than 80% of all deliveries occurring in community hospitals (Jain and Raju, 2006), many of which have a
relatively small number of deliveries, and health care teams
that might not always be equipped to assess and manage
the needs of a late preterm infant, it becomes increasingly important to establish safeguards for the screening,
identification, and appropriate triage of these patients.
A subcommittee on the American Academy of Pediatrics
Committee on the Fetus and Newborn (Engle et al, 2007)
outlined recommendations for the care of the late preterm newborn. Based on these guidelines and the potential complications associated with late preterm infants, we
recommend that all infants born before 35 weeks’ gestation weighing less than 2300 g should be admitted to a
transitional nursery where the infant can be monitored
closely until there has been adequate time to assess the
baby’s vital signs, feeding abilities, and thermoregulation, among other issues, before sending the baby to the
mother’s room. Sicker neonates who require intensive care
obviously will need to be admitted to higher levels of care.
In addition, each nursery should establish guidelines for
frequency of monitoring vital signs, assessment for sepsis
and use of antibiotics, and the use of supplemental oxygen. It is also important to determine a threshold (based
on comfort level, staff training, and available resources) for
transferring the newborn to a tertiary care center when the
disease process associated with the late preterm infant continues to progress or worsen. Box 33-3 shows the recommendations for admission, management, and discharge of
the late preterm infant. This list is not all-inclusive and was
designed to be used as a guideline, and not as a replacement for good clinical judgment.
CHAPTER 33 Care of the Late Preterm Infant
BOX 33-3 A
dmission and Discharge Criteria
and Management of Late Preterm
Infants
ADMISSION CRITERIA
Admit all infants born before 35 weeks’ gestation or weighing less than
2300 g at birth
ll They should not to be sent to their mother’s rooms in the first 24 hours until
stable, unless arrangements can be made to provide transitional care and
close monitoring in the mother’s room.
ll
HOSPITAL MANAGEMENT
Physical examination on admission and discharge
ll Determination of accurate gestation age on admission examination
ll Vital signs and pulse oximeter check on admission, followed by vital signs
every 3 to 4 hours in the first 24 hours, and every shift thereafter
ll Caution against use of oxyhoods with high Fio2; consider transfer to NICU or
tertiary care center if Fio2 exceeds 0.4
ll A feeding plan should be developed. Formal evaluation of breastfeeding
and documentation in the record by care givers trained in breastfeeding at
least twice daily after birth
ll Serum glucose screening per existing protocols for infants at high risk of
hypoglycemia
ll
DISCHARGE CRITERIA
Discharge should not be considered before 48 hours after birth
ll Vital signs should be within normal range for the 12 hours preceding
discharge
ll Respiratory rate less than 60 breaths/min
ll Heart rate of 100 to 160 beats/min
ll Axillary temperature of 36.5° to 37.4° C measured in an open crib with
appropriate clothing
ll Passage of one stool spontaneously
ll Adequate urine output
ll Twenty-four hours of successful feeding: ability to coordinate sucking,
swallowing, and breathing while feeding
ll If weight loss is greater than 7% in 48 hours, consider further assessment
before discharge
ll Risk assessment plan for jaundice for infants discharged within 72 hours
of birth
ll No evidence of active bleeding at circumcision site for at least 2 hours
ll Initial hepatitis B vaccine has been given or an appointment scheduled for
its administration
ll Metabolic and genetic screening tests have been performed in accordance
with local or hospital requirements
ll The late preterm infant has passed a car seat safety test
ll Hearing assessment is performed and results documented in the medical
record; follow-up if necessary has been arranged
ll Parents have been trained and demonstrate competency in caring for the
infant
ll Family, environmental, and social risk factors have been assessed; when
risk factors are present, discharge should be delayed until a plan for future
care has been generated
ll Identification of a physician with a follow-up visit arranged for 24 to 48
hours after discharge with a possibility of additional visits initially until the
infant can demonstrate a consistent pattern of weight gain
ll
Adapted from Engle WA, Tomashek KM, Wallman C: “Late Preterm” Infants: a population at
risk, Pediatrics 120: 1390-1401, 2007.
Fio2, Fractional concentration of oxygen in inspired gas; NICU, neonatal intensive care unit.
DISCHARGE CRITERIA
Because of the morbidities and risk factors associated with late preterm babies, they should not be discharged before 48 hours after birth. Before discharge,
415
while the baby is still in the NICU, the following are
recommended:
1. Vital signs should be within normal range for at least
12 hours preceding discharge; this includes respiratory rate less than 60 breaths/min, heart rate of 100
to 160 beats/min, and axillary temperature 36.5 to
37.5° C in an open crib with appropriate clothing.
2. There should be documentation of passage of at
least one stool spontaneously.
3. Adequate urine output should be accompanied by
educating the parents about ways to assess the adequacy of output and appropriate interventions if the
urine output appears to decrease, with at least 24
hours of successful feeding with adequate coordination of sucking, swallowing, and breathing during
feedings.
4. Weight loss should not exceed 7% of birthweight in
the first 48 hours of life.
5. Serum or transcutaneous bilirubin check—a transcutaneous bilirubin higher than 12 mg/dL should
warrant a serum bilirubin check, which will then
be stratified into risk category by using a bilirubin
nomogram. Parents should also be educated on
what to look for and what to do if their baby appears
jaundiced.
6. Hearing screen, a car seat test, and metabolic and
genetic screening tests should have been performed
in accordance with state, local, and hospital protocols; if the baby is circumcised, there should be no
bleeding at the site for at least 2 hours.
7. Hepatitis B vaccine should be given or an appointment should be made for its administration.
8. Parents should be educated about umbilical cord
and skin care, identification of common signs and
symptoms of illness, sleeping patterns and positions, instructions on using the thermometer and
parameters for normal measurements, and instructions regarding responses to an emergency (i.e.,
CPR training before discharge).
If these guidelines (or other criteria outlined as standard
of care) are not met, we recommend considering postponing discharge until the baby has been observed for a longer
period of time and the issues have been resolved.
When additional risk factors are present (e.g., twin or
multiple gestation, teenage mother), discharge should be
delayed until an appropriate care plan has been generated. In addition, when indicated, it may be appropriate to
arrange a nursing home health visit for closer monitoring,
but this should not be used to replace the due diligence
that must be done while the baby is in the hospital and the
appropriate and timely follow-up with a pediatrician.
Follow-up after Discharge
After discharge from the hospital, the majority of the medical care a newborn baby receives occurs in two main settings: the primary care physician’s office and the emergency
department. To avoid fragmented care by multiple emergency department visits and to allow for an early assessment of the baby, it is recommended that the late preterm
baby should be brought for a checkup by their pediatrician
within 24 to 48 hours after discharge from the hospital.
416
PART VIII Care of the High-Risk Infant
In addition, because of their increased risk for developmental delays, these infants should be monitored closely
to ensure that all milestones are achieved appropriately
and that early intervention (e.g., physical therapy, occupational therapy, speech therapy) is in place if needed. Early
developmental testing can also be useful in determining
any cognitive delays, which can then be addressed with
individualized educational programs.
Readmission to the Hospital
The late preterm infant is susceptible to many of the problems of smaller preterm infants. Because of the multiple
factors discussed throughout this chapter, the close observation and intensity of management provided to smaller
premature infants is often lacking. In addition, there are
often more lenient criteria for discharge, which sets them
(and their parents) up for failure and eventual readmission to the hospital. Tomashek et al (2007) looked at late
preterm infants who were discharged early (<2 days after
birth) from the hospital and found that 4.3% of late preterm and 2.7% of term infants were either readmitted or
had an observational hospital stay.
Recent data suggest that the most frequent causes of
emergency department visits by this subgroup of premature infants after being discharged home are dehydration,
feeding problems, respiratory distress, apnea, fever, infection, and jaundice (Jain and Cheng, 2006). Whether they
are evaluated in the emergency department or a pediatrician’s office, it is important to have a lower threshold for
readmitting these infants because of their vulnerability to
serious complications. Their fragility is evidenced by their
higher rate (4.4%) of rehospitalization than term infants
(2%; Escobar et al, 2005). In addition, studies show that
34- to 36-week infants who were never admitted to the
NICU (or if admitted, their NICU stay lasted less than
24 hours) had nearly a threefold and 1.3-fold higher risk
of readmission compared with term infants, respectively
(Escobar et al, 1999, 2005, 2006). Because late preterm
infants are at a much higher risk for rehospitalization, they
need close follow-up after discharge to assess breastfeeding and nutrition and to monitor for jaundice.
BOX 33-4 U
nanswered Questions
and Future Directions
EPIDEMIOLOGY, TRENDS, ETIOLOGY, AND PREVENTION
OF LATE PRETERM BIRTHS
ll Is there a subset of preventable late preterm births?
ll What is the effect of continuing such pregnancies (preterm rupture of
membranes, preterm labor, medically indicated birth) on perinatal outcomes?
ll Is there a role for antenatal steroids in late preterm gestation pregnancies
threatened with preterm delivery?
ll Should singleton and multiple gestation late preterm deliveries be treated
differently?
CLINICAL MANAGEMENT AND OUTCOMES OF LATE PRETERM
INFANTS
ll Can standardized “care paths” for late preterm infants improve outcomes?
ll Optimal discharge strategies that minimize readmissions and other
problems after discharge
ll Strategies for management of rapidly progressive and/or severe morbidities
in late preterm infants
ll Standardized approach to central nervous system imaging and postdischarge follow-up.
LONGTERM OUTCOMES
Prospective studies of long-term outcomes of symptomatic and asymptomatic late preterm births to determine which infants should have long-term
follow-up
ll
the understanding of the spectrum of morbidities in late
preterm infants; they have also formed the basis for current strategies to standardize management and optimize
outcomes.
SUGGESTED READINGS
Definition, Epidemiology, and Background
Davidoff MJ, Dias T, Damus K, et al: Changes in the gestational age distribution
among U.S. singleton births: impact on rates of late preterm birth, 1992 to
2002, Semin Perinatol 30:8-15, 2006.
Engle WA, Tomashek KM, Wallman C: Late-preterm” infants: a population at
risk, Pediatrics 120:1390-1401, 2007.
Raju TN, Higgins RD, Stark AR, et al: Optimizing care and outcome for latepreterm (near-term) infants: a summary of the workshop sponsored by the
National Institute of Child Health and Human Development, Pediatrics
118:1207-1214, 2006.
Pathophysiology and Clinical Course
FUTURE RESEARCH
Since 1981, the number of premature births in the United
States has increased by 30%. The majority of this increase
is attributable to the increased number of late preterm
infants (Pulver et al, 2009)—a unique and high-risk subgroup of premature infants. The identification and care of
this group and their unique challenges have only recently
been recognized as a high-priority area of research in the
field of neonatology. There are many questions still to be
answered regarding the care and outcomes associated with
late preterm infants (Box 33-4). Topping this list of priorities is the need to know which babies are better off being
delivered early, and for the rest, what can be done to take
pregnancy as close to term as possible. Because answers
to many issues related to preterm birth still remain elusive, clinicians have, rightly so, focused on ways to improve
the postnatal management of these premature babies.
New studies, albeit mostly retrospective, have added to
Jain L: Alveolar fluid clearance in developing lungs and its role in neonatal transition, Clin Perinatol 26:585-599, 1999.
McIntire DD, Leveno KJ: Neonatal mortality and morbidity rates in late preterm
births compared with births at term, Obstet Gynecol 111:35-41, 2008.
Wang ML, Dorer DJ, Fleming MP, et al: Clinical outcomes of near-term infants,
Pediatrics 114:372-376, 2004.
Mortality
Kramer MS, Demissie K, Yang H, et al: The contribution of mild and moderate
preterm birth to infant mortality. Fetal and Infant Health Study Group of the
Canadian Perinatal Surveillance System, J Am Med Assoc 284:843-849, 2000.
Tomashek KM, Shapiro-Mendoza CK, Davidoff MJ, et al: Differences in mortality
between late-preterm and term singleton infants in the United States, 19952002, J Pediatr 151:450-456, 2007.
Longterm Outcome
Chyi LJ, Lee HC, Hintz SR, et al: School outcomes of late preterm infants: special
needs and challenges for infants born at 32 to 36 weeks gestation, J Pediatr
153:25-31, 2008.
Kinney HC: The near-term (late preterm) human brain and risk for periventricular
leukomalacia: a review, Semin Perinatol 30:81-88, 2006.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
34
Pharmacokinetics, Pharmacodynamics,
and Pharmacogenetics
Robert M. Ward, Steven E. Kern, and Ralph A. Lugo
Dynamic changes in growth and physiologic maturation
in newborns create unique complexities in drug therapy
that affect absorption, distribution, metabolism, and elimination. Pharmacologic studies during this period of rapid
growth and physiologic maturation reveal different patterns of change in kinetics, some related to postnatal age
and others to postmenstrual age. These differences also
provide information about varied patterns of maturation
in drug metabolizing enzymes and pathways of elimination during early infancy. Therapeutic drug monitoring is
used for a few drugs in neonatology, such as phenobarbital and gentamicin, when concentrations correlate with
desired effects as well as adverse effects. Dosage adjustments to reach desired concentrations can be estimated at
the bedside using simple calculations. Pharmacogenetics
and pharmacogenomics of enzymes and receptors explain
many variations among individuals in their responses to
drugs. The immature, preterm newborn adds another level
of complexity to drug metabolism, because of variability in
the timing of the expression of these enzymes.
PRINCIPLES OF NEONATAL
THERAPEUTICS
A thorough understanding of factors that affect drug
concentrations helps to provide accurate, effective drug
therapy and to identify the causes of therapeutic failure in neonates. Many important factors are not chosen
consciously in a therapeutic plan, but have tremendous
impact on its effectiveness. Because pharmacokinetics and
pharmacodynamics in newborns follow the same general
principles that govern drug actions in patients of any age,
the diagnosis, drug selection, and administration needed
to achieve a therapeutic goal must consider the effects
of absorption, distribution, metabolism, and excretion
on the dose-exposure relationship. When applied to the
newborn, these principles should adjust for several unique
physiologic and pharmacologic features of these immature
patients, as outlined in Box 34-1 and discussed in detail
here.
DIAGNOSIS
Effective treatment begins with an accurate diagnosis and
assessment of clinical signs and symptoms. Although this
principle applies to all areas of therapeutics, treatment in
newborns presents special diagnostic challenges because
the small size and fragility of such nonverbal patients may
preclude useful, but inordinately invasive, diagnostic procedures. For example, many small immature newborns
with chronic lung disease are treated for bronchospasm
based on the findings of decreased air entry associated
with desaturation and abnormal breath sounds. Relief of
these symptoms with aerosolized bronchodilators may be
interpreted as confirmation of the diagnosis. Although
this interpretation may be correct, increased humidity or
movement of the endotracheal tube bevel away from a pliable tracheal wall during an aerosol treatment may also
produce the improvement. Therefore evaluation of any
ineffective therapy should include reconsideration of the
original diagnosis, just as conclusions about why a therapy
succeeded should be made with a degree of skepticism.
ABSORPTION
Although most drug therapy for acute problems in the
intensive care setting involves intravenous administration
to ensure drug delivery to the site of action, this route is
not always reliable in newborns (Gould and Roberts, 1979;
Roberts, 1984). In a critical care setting, drugs should be
infused toward the patient as close as possible to the site of
vascular access. If a drug is injected away from the infant
into a catheter with a slow infusion rate, the drug may
reach the patient too slowly to achieve effective concentrations. Infusion solution filters may also prevent effective
drug delivery by blocking large molecules, such as amphotericin, by direct adsorption of the drug to the filter, or
by allowing a heavier drug to settle in the filtration chamber and mix slowly with the infusion solution. For drug
therapy in which the driving force for tissue penetration is
a concentration gradient between the circulation and the
tissue (e.g., in meningitis), sustained low drug concentrations might be therapeutically suboptimal.
Intramuscular administration of drugs to newborns is
generally suboptimal, but might be used when there is difficulty establishing intravenous access. Absorption of drugs
from an intramuscular injection site is directly related to
muscle blood flow, which is usually reduced in patients
experiencing hypothermia or shock, when intravenous
access can be difficult, but intramuscular doses are unlikely
to be absorbed effectively. Furthermore, intramuscular
administration of drugs can sclerose tissue and cause sterile abscesses or create large intramuscular collections of
the drugs, which are then absorbed slowly, producing a
“depot effect” in which serum concentrations rise slowly
over a prolonged period. Intramuscular administration of
drugs, especially for multiple doses, should be avoided in
newborns. Although oral administration of drugs is preferred for treatment of chronic illnesses in newborns, this
route is not well studied, particularly in acutely ill premature infants. Studies in adults show that less drug is usually
absorbed from the stomach than from the intestinal tract
because of the smaller surface area and differences in pH.
Many newborns experience gastroesophageal reflux associated with delayed gastric emptying, which might also alter
drug bioavailability. Delayed gastric emptying postpones
417
418
PART VIII Care of the High-Risk Infant
BOX 34-1 P
harmacologic Principles and
Pitfalls in Management of the
Very Low-Birthweight Infant
I. Diagnosis
A. Limited diagnostic procedures
II. Absorption
A. Intravenous
1. Drug injection away from patient
2. Uneven mixing of drugs and intravenous fluids
3. Delayed administration because of low flow
4. Part of the dose discarded with tubing changes
B. Intramuscular
1. Poor perfusion limits absorption
2. Danger of sclerosis or abscess formation
3. Depot effect
C. Oral
1. Poorly studied
2. Affected by delayed gastric emptying
3. Potentially affected by reflux
4. Passive venous congestion may occur with chronic lung disease,
decreasing absorption
III. Distribution, affected by
A. Higher (85%) total body water (versus 65% in adults)
B. Lower body fat—that is, approximately 1% body weight
(versus 15% in term infants)
C. Low protein concentration
D. Decreased protein affinity for drugs
IV. Metabolism
A. Half-life prolonged and unpredictable
B. Total body clearance decreased
C. Affected by nutrition, illness, and drug interaction
D. Affected by maturational changes
V. Excretion: decreased renal function, both glomerular filtration rate and
tubular secretion
reaching peak serum drug concentrations and prolongs the
absorption phase, which reduces the peak concentration.
If the total absorbed dose is reduced, the area under the
concentration time curve (AUC) will decrease along with
drug exposure. Passive venous congestion of the intestinal
tract from elevated right atrial pressures decreases drug
absorption in adults and may do so in premature infants
with severe bronchopulmonary dysplasia complicated by
cor pulmonale (Peterson et al, 1980). The administration
of medications to newborns in small volumes of formula
or during continuous gastric feedings may also alter drug
absorption by binding to proteins, lipids, carbohydrates, or
minerals in the feeding. When enteral drug therapy fails,
possible effects of feeding patterns on drug absorption and
action should be considered.
DISTRIBUTION
In pharmacokinetics, distribution is the partitioning of
drugs among various body fluids, organs, and tissues. The
distribution of a drug within the body is determined by
several factors, including organ blood flow, pH and composition of body fluids and tissues, physical and chemical properties of the drug (e.g., lipid solubility, molecular
weight, ionization constant), and the extent of drug binding to plasma proteins and other macromolecules (Plonait
and Nau, 2004; Ward and Lugo, 2005).
Important differences among premature infants, children, and adults affect the distribution of drugs. Total body
water varies from 85% in premature newborns to 75%
in term newborns to 65% in adults (Friis-Hansen, 1961,
1971). Conversely, body fat content varies from 0.7% or
less in extremely premature newborns to approximately
12% in term newborns (Friis-Hansen, 1971; Ziegler
et al, 1976). These differences change the distribution of
many drugs, especially polar, water-soluble drugs such as
the aminoglycosides. Protein binding of drugs in the circulation is decreased in the premature newborn because
of a smaller total amount of circulating protein and lower
binding affinity of the protein itself (Aranda et al, 1976).
With rare exceptions, only the free (not bound to protein)
drug molecules cross membranes, exert pharmacologic
actions, and undergo metabolism and excretion. Clinical
measurements of serum or plasma drug concentrations
usually reflect total circulating drug concentrations, which
consist of both free and protein-bound drug. Thus, even
when total circulating drug concentrations in the newborn
may be low by adult standards, the free drug concentrations may be equivalent or even higher than those in the
adult because of decreased protein binding in the newborn.
METABOLISM
Many drugs require metabolic conversion before elimination from the body. Biotransformation of a drug usually
produces a more polar, less lipid-soluble molecule that can
then be eliminated rapidly by renal, biliary, or other routes
of excretion. Drug biotransformation is classified into two
broad categories: nonsynthetic (phase I) reactions, which
include oxidation, reduction, and hydrolysis; and synthetic
or conjugation (phase II) reactions, which include glucuronidation, sulfation, and acetylation. Multiple forms of
phase I and phase II enzymes exist to perform these metabolic functions. Although the liver is considered the major
organ responsible for drug biotransformation, many other
organs also contribute to drug metabolism.
For many drugs in the newborn, the half-life is prolonged and total body clearance is decreased compared
with older children and adults. Important variations occur,
however, among drug classes and among individuals that
prevent broad generalizations. Glucuronide conjugation
of bilirubin by uridine 5′-diphospho-glucuronosyltransferase (UGT) 1A1 is usually low at birth unless this enzyme
has been induced in utero through maternal exposure to
drugs, cigarette smoke, or other inducing agents (Maurer
et al, 1968). In contrast, conjugation through sulfation is
usually active at birth. Various factors after birth, such
as nutrition, illness, and drug interactions, may hasten
or retard the maturation of enzymes and organs responsible for drug metabolism in the newborn. Maturational
changes in hepatic blood flow, drug transport into hepatocytes, synthesis of serum proteins, protein binding of
drugs, and biliary secretion—alone and in combination—
confound accurate predictions about drug metabolism
after birth, leading to empiric dose adjustments (Morselli
et al, 1980). Because smaller and more immature newborns
are now surviving, many of these factors that were studied
in larger, more mature neonates must be reassessed in this
less mature population.
CHAPTER 34 Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics
The primary types of enzymes involved in phase I reactions are cytochromes P450 (CYPs). CYPs are microsomal,
mixed-function oxidases that catalyze chemical changes
within a molecule, such as hydroxylation, methylation,
demethylation, addition of oxygen, or removal of hydrogen (Leeder and Kearns, 1997). There are thousands of
different CYP450 enzymes found in plants and animals on
land and in water. CYPs with more than 40% of the same
amino acid sequence are grouped as a numbered family
(e.g., CYP3). Those with greater than 67% polypeptide
homology belong to the same subfamily (e.g., CYP3A), and
specific genes are denoted with a number (e.g., CYP3A5).
The amino acid sequence of these enzymes determines
the tertiary structure that creates a hydrophobic pocket
with selective binding for chemicals and drugs. This substrate specificity creates groups of drugs, often with similar
structure and function that are metabolized by the same
CYP. In the preterm newborn, CYPs develop at different
rates and in different patterns. Maturation correlates best
with postmenstrual age for some CYPs and with postnatal age for others. Some CYPs do not reach adult activity
until several years of age, whereas others develop activities
twice that of the adult during childhood; therefore generalizations about patterns of maturation for CYPs are seldom valid. In addition, single nucleotide polymorphisms
(SNPs) or substitutions in the DNA sequence for a CYP
may reduce its metabolizing activity or completely eliminate it if the polypeptide cannot be formed.
The phase II conjugation enzymes, such as the UGTs,
have several forms with different substrate specificity (e.g.,
UGT1A1 for bilirubin, UGT2B7 for morphine), although
they are not as absolutely selective as the CYPs (Leeder
and Kearns, 1997). Some phase II enzymes complement
each other at birth for conjugation of specific drugs, such
as acetaminophen that is conjugated primarily by sulfotransferases at birth and less so by UGT. Moreover, by
adolescence, UGT has become the predominant conjugation pathway for acetaminophen.
EXCRETION
Drug elimination from the body can occur through several
mechanisms, including renal excretion, biliary excretion,
transcutaneous loss, gastrointestinal loss, and pulmonary
exhalation. Renal excretion is one of the most important
pathways for elimination of metabolized and unchanged
drug. Neonatal renal function is diminished in absolute
terms and when normalized to body weight or surface
area. The neonatal glomerular filtration rate averages 30%
of the adult rate per unit surface area. Glomerular function
rises steadily after birth, whereas tubular function matures
more slowly, causing a glomerular and tubular imbalance
(Aperia et al, 1981). The postnatal increase in glomerular function reflects greater cardiac output, reduced renal
vascular resistance, redistribution of intrarenal blood flow,
and changes in intrinsic glomerular basement membrane
permeability (Morselli et al, 1980). The dynamics of neonatal renal function markedly influence drug excretion.
The rate of change of renal function and its susceptibility to hypoxemia, nephrotoxic drugs, and underperfusion
prevent accurate predictions of drug elimination rates in
newborns, which often must be measured empirically.
419
Some drugs, such as nafcillin and spironolactone, have
metabolites that must be eliminated through biliary excretion. Drugs that are conjugated within the liver may also
be excreted through bile, enter the intestinal tract, and
undergo deconjugation and enterohepatic recirculation,
similar to bilirubin. Although biliary excretion is not well
studied in newborns, clinical conditions such as parenteral
nutrition– associated cholestasis suggest that it may be
highly variable among specific patients and conditions.
Transporters play important roles in removing drugs
and preventing drug absorption. Organic anion transporter polypeptides provide facilitated transport of anions
in many tissues, including the kidney and liver. Permeability glycoprotein is an efflux transporter that belongs to the
adenosine triphosphate–binding cassette–multiple drug
resistance family of transporters. Permeability glycoprotein prevents the absorption of many compounds across
the intestinal wall or into the brain, where it functions as a
significant portion of the blood brain barrier.
PHARMACOGENETICS
AND PHARMACOGENOMICS
The human genome project described the protein structures of many enzymes involved in drug metabolism and
identified genetic variants in the proteins that alter activity, many of which are produced by a change in a single
nucleotide to create SNPs. As discussed in the previous
sections, genetic variation in drug metabolizing enzymes
and in drug transporters can have a significant influence
on the relative activity of these systems within a particular individual. Knowledge of pharmacogenetic and pharmacogenomic factors that can effect pharmacokinetics,
particularly with a relationship to drug metabolism and
elimination, has been important for understanding ways
to avoid unanticipated drug effects in adults and to some
extent older children. In neonates, however, this knowledge
is only beginning to develop. Moreover, because many of
these drug-metabolizing systems that are subject to pharmacogenetic variation continue to mature during the first
few years of life, it can be difficult to determine whether
genetic variability or physiologic maturation are contributing to differences in drug metabolism by neonates. What
should be appreciated from this limited knowledge, however, is that drug metabolism is often reduced in neonates,
and scaling of drug dosage by simple body weight or allometrically with an exponent will not fully compensate for
differences in clearance that exist in this newborn population. Clearance often varies several-fold among adults, and
the same degree of variation is emerging among neonates
whether caused by maturation of the expression of these
enzymes by gestational age or induction of protein synthesis after birth.
One of the most prominent phase 1 metabolizing
enzymes, CYP3A4, illustrates several important aspects of
CYPs. CYP3A4 exists as a fetal form at birth (CYP3A7)
that decreases over the first months of life as it is replaced
by the adult form, CYP3A4, by 1 year of age (Lacroix
et al, 1997). However, these changes are not linear, which
confounds making accurate dosing adjustments by age.
The maturation of metabolic activity of phase I enzymes to
adult activity varies among the specific enzymes, requiring
420
PART VIII Care of the High-Risk Infant
months for some and years for others (Kearns et al, 2003).
During the fetal–neonatal period some CYPs increase
in activity, but the most rapid increase in activity usually
occurs after birth, regardless of the gestational age at birth
(Koukouritaki et al, 2004). Drug-drug interactions influence activity of CYPs as well. Macrolide antibiotics, such as
erythromycin and clarithromycin, and the azole antifungal
drugs, such as fluconazole, inhibit the activity of CYP3A4
(Leeder and Kearns, 1997). These drugs can reduce clearance for drugs that are a substrate for CYP3A4, such as
fentanyl or midazolam, and lead to toxicity without dosage adjustments. Conversely, inducers of CYP3A4 activity,
such as rifampin and phenobarbital, can increase clearance
of drugs that are substrates and reduce the concentration
and effectiveness.
Large interindividual variation occurs for most of these
enzymes and is complicated by inherited differences in
activity. An SNP in the DNA for one of these enzymes
is designated with an asterisk, such as CYP2C9*1, and
can alter the protein structure enough to reduce or even
inactivate its enzymatic activity. SNPs have been identified for most CYPs, especially CYP2D6, one of the first
recognized inherited variations in activity for debrisoquine
(Leeder and Kearns, 1997). Ethnic variations in these
SNPs help to predict when activity is likely to be reduced
or increased. For example, several inactivating SNPs of
CYP2D6 have been recognized, indicating that 50% or
more of Asians lack enough active CYP2D6 to metabolize codeine to morphine for a more potent analgesic effect
(Ingelman-Sundberg, 2005). Study of the maturational
changes in the pharmacokinetics of drugs metabolized by
specific CYPs expands current knowledge of how these
enzymes mature. A better understanding of the rates and
patterns of maturation of different CYPs will help to guide
appropriate dosage adjustments to improve drug therapy
for newborns.
PHARMACOKINETIC PRINCIPLES
Pharmacokinetics describes the time course of changes in
drug concentrations within the body. Although rates of
change are often described with differential equations,
useful concepts at the bedside are emphasized in this section. More detailed mathematical discussions of pharmacokinetics can be found elsewhere (Buxton, 2006; Rowland
and Tozer, 2010).
COMPARTMENT
In pharmacokinetics, compartment refers to fluid and tissue
spaces into which drugs penetrate. These compartments
may or may not be equivalent to anatomic or physiologic fluid volumes. In the simplest case, the compartment may correspond to the vascular space and equal the
volume of a real body fluid (i.e., blood). Large or highly
polar molecules can be confined to this central compartment until they are eliminated by excretion or metabolism. Many drugs, however, diffuse reversibly out of the
central compartment into tissues or other fluid spaces,
generically referred to as peripheral or tissue compartments.
Such compartments are seldom sampled directly, but their
involvement in kinetic processes may be recognized from
the graphic or mathematical description of the kinetics
of a drug. The number of exponential terms necessary to
adequately describe the kinetic profile of a drug designates
the number of compartments involved, recognizing that
many more compartments may exist. For appropriate clinical application, rarely are more than three compartments
required to describe a drug’s pharmacokinetics.
APPARENT VOLUME OF DISTRIBUTION
The apparent volume of distribution might be better
termed volume of dilution, because it is a mathematical
description of the volume (L or L/kg) that dilutes a dose
(mg or mg/kg) to produce the observed circulating drug
concentration (mg/L or μg/mL). (To simplify cancellation
of units, concentrations are expressed as mg/L, which is
the same as μg/mL, the more conventional unit for drug
concentrations.)
Concentration (mg/L) =
Dose (mg/kg)
Apparent volume of distribution (L/kg)
For many drugs, the volume of distribution does not correspond to a specific physiologic body fluid or tissue—
hence the term apparent. In fact, the volume of distribution
for drugs that are bound extensively in tissues may exceed
1.0 L/kg, a physiologic impossibility that emphasizes the
arithmetic, nonphysiologic nature of the apparent volume
of distribution. The determination of distribution volume
is described later.
FIRST-ORDER KINETICS
Removal of most drugs from the body can be described by
first-order (exponential or proportional) kinetics, in which
a constant proportion or percentage of a drug is removed
per unit of time (e.g., 50% in one half-life interval), rather
than a constant amount per unit of time. For drugs exhibiting first-order kinetics, the higher the concentration,
the greater the amount removed during an interval of
time. The following equations describe the concentration
(C) of a drug whose first-order kinetics have a constant
rate k (hour−1), at time t, and an initial concentration of C0
achieved after administration of a dose.
In differential equation form, the change in C with time is
dC
= − kC
dt
The solution to this differential equation gives the exponential form, which describes C at time t:
Ct = C0 e − kt
If this equation is transformed using the natural logarithm
(ln), it results in:
ln Ct = ln C0 − kt
The last equation fits the equation of a straight line, so that
a graph that plots ln Ct versus t has an intercept of ln C0 at
t = 0 and a slope of –k, the rate constant for the change in
concentration; this can be used to calculate the half-life and
to estimate appropriate dosages. Multiple rate constants in
CHAPTER 34 Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics
1000
10
DISTRIBUTION PHASE (ALPHA)
(Distribution + Elimination)
Log serum concentration
Plasma Concentration
800
400
200
T1/2
421
ELIMINATION PHASE (BETA)
(Elimination)
1
Slope = β/2.303
.1
Slope = α/2.303
100
0
30
60
90
120
150
180
Minutes after Dose
FIGURE 34-1 Apparent single-compartment, first-order plasma drug
disappearance curve illustrating graphic determination of half-life from
best-fit line of serial plasma concentrations.
more complex equations are distinguished with the letter k
and numbered subscripts or with Greek letters.
HALF-LIFE
The drug half-life (t1/2) is the time required for a drug concentration to decrease by 50%. Half-life is a first-order kinetic
process because the same proportion, 50%, of the drug is
removed during equal periods. Half-life can be determined
mathematically from the elimination rate constant k as:
Natural logarithm 2 0.693
t1/2 =
=
k
k
Figure 34-1 illustrates a graphical method for determination of half-life. Drug concentrations measured serially
are graphed on semilogarithmic axes, and the best-fit line
is determined either visually or by linear regression analysis. In this illustration of first-order kinetics, the concentration decreases 50% (from 800 to 400) during the first
hour and decreases another 50% (from 400 to 200) during
the second hour. Thus the half-life is 1 hour. More drug
is removed during one half-life at higher concentrations,
although the proportion removed remains constant. The
exponential equation for this graph is:
C = 800e − 0.693t
where k = 0.693/h and C0 = 800, allowing a mathematical calculation of half-life using the equation described
previously:
0.693
0.693
t1/2 =
= 1 hour
=
-1
0.693/h
k (hour )
MULTICOMPARTMENT, FIRST-ORDER
KINETICS
The rate of removal of many drugs from the circulation is
often biphasic. An initial rapid decrease in concentration is
the distribution (α) phase, often lasting 15 to 45 minutes,
which is followed by a sustained slower rate of removal,
the elimination (β) phase. Such biphasic processes are best
visualized from semilogarithmic graphs of concentration
versus time. When such semilogarithmic graphs show
.01
0
120
240
Minutes after dose
FIGURE 34-2 Multicompartment serum drug disappearance curve.
kinetics that best fit two straight lines, the kinetics are
described as biexponential, or reflective of a drug that shows
two-compartment first order pharmacokinetics (Figure
34-2). Two exponential terms are needed to describe the
change in concentration over time, as:
C = Ae − αt + Be − βt
In this equation, the rate constant for distribution is
designated α to discriminate it from the rate constant for
terminal elimination (β), where A and B are the time = 0
intercepts for the lines describing distribution and elimination, respectively. Division by 2.303 converts logarithms
to natural logarithms.
After an intravenous dose, drug loss from the vascular
space during the distribution phase occurs through both
distribution and elimination (see Figure 34-2). The rate
constant of distribution (α) can be determined by plotting
the difference between the total amount of drug lost initially
and the amount of drug lost through elimination (Greenblatt and Koch-Weser, 1975). This determination produces
the line with the steeper slope (equal to α/2.303) below the
serum concentration graph in Figure 34-2. The single slope
of the distribution phase and of the terminal elimination
phase does not imply that distribution or elimination occurs
through a single process. The observed rates usually represent the summation of several simultaneous processes, each
with differing rates, occurring in various tissues.
When the time course of drug elimination is observed
for prolonged periods, a third rate of elimination, or γ
phase, may also be observed and is usually attributed to
elimination of drug that has reequilibrated from deep tissue compartments back into the plasma. Such kinetics are
designated three-compartment first-order pharmacokinetics. The kinetics of a drug are expressed with the smallest
number of compartments that accurately describe its concentration changes over time.
APPARENT SINGLE-COMPARTMENT,
FIRST-ORDER KINETICS
When a semilogarithmic graph of concentration versus
time reveals a single slope with no distribution phase, the
kinetics are characterized as apparent one-compartment,
422
PART VIII Care of the High-Risk Infant
BOX 34-2 D
rugs That Demonstrate Saturation
Kinetics With Therapeutic Doses
in Newborns
ll
ll
ll
ll
ll
ll
FIGURE 34-3 Representation of saturation, or zero-order (serum
concentration–dependent), and first-order (serum concentration–
independent) pharmacokinetics.
first-order (see Figure 34-1). Such kinetics can occur when
a drug remains entirely within the vascular space or central compartment or when a drug passes rapidly back and
forth between the circulation and peripheral sites until it is
metabolized or excreted by first-order kinetics. The adjective apparent is used because careful study often shows that
distribution occurs even though the kinetic curve has only a
single slope. Single-compartment kinetics implies that the
drug rapidly and completely distributes homogeneously
throughout the body, which rarely occurs clinically.
In many pharmacokinetic studies in newborns, blood
samples are not obtained early enough to allow calculation
of the distribution phase, and the kinetics are described
as single-compartment. If sampling begins after the distribution phase, the concentration time points may fit a
single-compartment, first-order model, which determines
the elimination rate constant (β). The kinetics cannot be
assumed, however, to fit a single-compartment model
from such a limited study. The more clinically limited but
accurate approach to kinetic analysis, noncompartmental
analysis, makes no assumptions about the number of compartments (Rowland and Tozer, 2010).
ZERO-ORDER KINETICS
Some drugs demonstrate zero-order kinetics, in which a
constant amount of drug, rather than a constant proportion or percentage, is removed per unit of time. This relationship can be expressed as:
dC
= −k
dt
It is important to understand when zero-order kinetics
occurs, how to recognize it, and how it affects drug concentrations. Zero-order kinetics is sometimes referred
to as saturation kinetics, because it can occur when excess
amounts of drug completely saturate enzymes or transport systems so that they metabolize or transport only a
constant amount of drug over time. Zero-order processes
produce a curvilinear shape in a semilogarithmic graph of
concentrations versus time (Figure 34-3). When drug concentrations are high from a drug overdose or the pathway
for elimination is impaired as in renal dysfunction, kinetics
Caffeine
Heparin
Theophylline
Furosemide
Indomethacin
Phenytoin
may become zero-order initially and be followed by firstorder kinetics at lower concentrations. For drugs exhibiting zero-order kinetics, small increments in dose can cause
disproportionately large increments in serum concentration. Certain drugs administered to newborns exhibit
zero-order kinetics at therapeutic doses, and concentrations and must be recognized for their potential accumulation (Box 34-2).
NONCOMPARTMENTAL ANALYSIS
Noncompartmental analysis is based on describing drug
exposure measured by the AUC without any assumptions
about the pattern of elimination or number of compartments. Central to this analysis is the determination of drug
clearance from the dose and the AUC:
Cl = Dose / AUC
If the dose is administered intravenously, then noncompartmental analysis allows the direct determination of drug
clearance using this relationship. Estimation of the elimination half-life is generally done using the slope of the log
transformed concentration measurements made during
the end of a pharmacokinetic study. With an estimation of
clearance and elimination rate, the apparent distribution
volume can also be estimated from:
Cl = V × K
Thus noncompartmental pharmacokinetics provides a
simple means to assess fundamental pharmacokinetic
parameters, which may be useful for dosing patients when
detailed knowledge of the complete pharmacokinetic profile is not needed.
POPULATION PHARMACOKINETICS
Most pharmacokinetic studies in newborns are limited
by the amount of blood that can be removed safely for
sampling. To determine the kinetics safely, population
pharmacokinetic approaches are valuable, particularly
because they can accommodate unbalanced study designs
for drug sampling. The population approach describes
the concentration-versus-time profile for all the patients
enrolled in a given study simultaneously, estimating population parameters that describe the general pharmacokinetic profile of the complete study group and patient
specific parameters that define the individual patients in
the study. The population approach can use fewer samples
taken from each patient, if the samples are taken at different specified times over the complete time profile of
423
CHAPTER 34 Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics
interest for the study analysis. For example, one group
of 28- to 32-week premature infants might have samples
drawn at 1, 4, and 12 hours, whereas another group of 28to 32-week premature infants might be sampled at 0.5, 2,
and 8 hours. The concentrations from these two groups
of similar patients are then analyzed in aggregate to provide information during both distribution and elimination
phases, thus describing the kinetics with a limited volume
of blood sampled from each patient.
Furthermore, the population approach allows for the
investigation of patient covariates of interest that might
explain differences seen within the population of patients
enrolled in the trial. Typical covariates such as gestational
age, gender, and disease conditions can also be assessed for
their contribution to differences seen between subjects in a
clinical study. These covariates can be helpful for gaining a
better understanding of factors that may alter the pharmacokinetics of infants that might otherwise be considered similar.
TARGET DRUG CONCENTRATION STRATEGY
Drug treatment of newborns commonly uses the target
drug concentration strategy (Box 34-3), in which drug therapy corrects a specific problem by producing an effective
concentration of free drug at a specific site of action (Rowland and Tozer, 2010). The target site of drug action is usually inaccessible for monitoring concentrations. A specific
concentration or range of circulating concentrations is correlated with the effective concentration at the site of action,
which provides a “therapeutic” concentration range.
The requirements for effective and accurate application of the target drug concentration treatment strategy in
adults have been discussed by Spector et al (1988). When
applied to newborns, these requirements highlight the
special problems of drug therapy in these patients and the
special circumstances in which clinical drug concentration
monitoring is appropriate. Some of these requirements are
as follows:
ll An available analytic procedure for accurate measurement of drug concentrations in small volumes of blood
ll A wide variation in pharmacokinetics among individuals with the knowledge that population-based kinetics
do not accurately predict individual kinetics
ll Drug
effects are proportional to plasma drug
concentrations
ll A narrow concentration range between efficacy and
toxicity (narrow therapeutic index)
ll Constant pharmacologic effect over time, in which tolerance does not develop
ll Clinical studies that have determined the therapeutic
and toxic drug concentration ranges
THERAPEUTIC DRUG MONITORING
Box 34-3 illustrates the basic assumptions of therapeutic
drug monitoring—that total plasma drug concentrations
correlate with dose, circulating unbound drug concentrations, unbound drug concentration at the site of action.
Clinical measurements of drug concentrations usually
include both bound and unbound drug, and the active
portion is the portion that is unbound (see Distribution,
earlier). The two broad indications for monitoring drug
BOX 34-3 T
arget Drug Concentration
Strategy
Drug
dose
↔ Plasma
total drug
concentration
↔
Plasma
unbound drug
concentration
↔
Target site
unbound drug
concentration
↔
Desired
pharmacologic
effect
Data from Shenier LB, Tozer TN: Clinical pharmacokinetics: the use of plasma
concentrations of drugs. In Melmon KL, Morrelli HF, editors: Clinical pharmacology:
basic principles in therapeutics, ed 2, New York, 1978, Macmillan, p 71.
concentrations are attainment of effective concentrations and avoidance of toxic concentrations. As Kauffman
(1981) has indicated, drug concentration ranges are not
absolute reflections of effective therapy. Patient response,
not a specific drug concentration range, is the endpoint of
therapy.
Although concentrations of aminoglycoside antibiotics, such as gentamicin, are monitored frequently in newborns, toxicity is rare (McCracken, 1986). Because of the
limited evidence of toxicity in newborns, it is more important to measure aminoglycoside concentrations to achieve
effective concentrations for treatment of culture-proven
infections than to avoid toxicity. In newborns with serious therapeutic problems, measurement of serum drug
concentrations should be used to achieve effective concentrations and to avoid toxicity. When the desired concentration range and kinetic parameters are known, doses may
be estimated to reach that concentration with single bolus
doses or bolus doses followed by continuous infusions.
PHARMACOKINETIC-BASED DOSING
The following equations can be used both to guide dosing
and to derive kinetic parameters for individual patients.
Dose = ΔC × Vd = (Cdesired − Cinitial ) × Vd
mg/kg = (mg/L) × (L/kg) = (mg/L) × (L/kg)
where C = concentration, and Vd = volume of distribution.
This equation may be used to estimate dosage changes
needed to increase or decrease concentration. For the first
dose, the starting concentration is zero; afterward, the
calculation of distribution volume should use the change
(Δ) in concentration from the preceding trough to the peak
associated with that dose. To reach a desired concentration rapidly, a loading dose can be administered followed
by a sustaining infusion. The equation for calculation of
infusion doses to maintain a constant concentration is
shown:
Infusion rate = k × Vd × C
(mg/kg) (min−1 ) = min−1 × (L/kg) × (mg/L)
where C = concentration, Vd = volume of distribution, and
k = rate constant of elimination.
Steady state is reached when tissue concentrations are
in equilibrium and the amount of drug removed equals
the amount of drug infused. The time needed to reach a
steady state depends on the elimination half-life. Whereas
the time is not shortened by the administration of a loading dose, a loading dose can allow the patient to achieve
424
PART VIII Care of the High-Risk Infant
and trough concentrations after the first dose); therefore the shorter the dosing interval–to–half-life ratio, the
higher the drug accumulation. As noted during infusions,
the length of time required to reach steady-state concentrations depends primarily on the elimination half-life, not
the dosing interval.
CLEARANCE
FIGURE 34-4 Representation of multiple dosing with accumulation
of serum drug levels to steady-state concentration.
therapeutic concentrations rapidly that can be maintained
with an infusion during steady state.
It is important to consider that if drug clearance
decreases, the steady state concentration during an infusion will increase proportionally. In addition, the half-life
increases and the rate constant decreases. Because concentrations are not measured for most of the drugs administered by continuous infusion in the neonatal intensive care
unit (NICU), it is important to adjust dosages for factors
that reduce clearance, such as kidney or liver dysfunction
or reduced kidney or liver blood flow, to avoid high and
toxic concentrations.
REPETITIVE DOSING AND THE “PLATEAU
PRINCIPLE”
During the typical course of drug therapy, drug doses are
administered before complete elimination of previous
doses, and the drug accumulates in the body (Rowland
and Tozer, 2010). During repeated administration, the
peak and trough levels after each dose increase for a time.
Steady-state, or plateau, concentrations are reached when
the amount of drug eliminated equals the amount of drug
administered during each dosing interval. During repetitive dosing, the steady-state concentrations achieved are
related to the half-life, dose, and dosing interval relative
to the half-life (Buxton, 2006; Rowland and Tozer, 2010).
Figure 34-4 illustrates a hypothetical concentration-time
curve for a drug with a half-life of 4 hours administered
orally every 4 hours, so that the dosing interval corresponds
to one half-life. Several important principles of pharmacokinetics are illustrated in this figure; the mathematics are
described in detail elsewhere (Buxton, 2006). Drug concentrations rise and fall with drug administration (absorption) and elimination. For dosing intervals of one half-life,
accumulation is 88% complete after the third dose, 94%
complete after the fourth dose, and 97% complete after
the fifth dose. At steady state, the peak and trough concentrations between doses are the same after each dose. If
a drug is administered with a dosing interval equal to one
half-life, the steady-state peak and trough concentrations
are twofold those reached after the first dose. If the dosing
interval is shortened to half of a half-life, the concentration decreases less before the next dose, more total drug is
administered per day, and the steady-state peak and trough
concentrations are considerably higher (3.4-fold the peak
Clearance of drugs, as for creatinine, describes the volume
of blood from which all the drug is removed per unit of
time. Clearance is proportional to organ blood flow and
the intrinsic capacity of organs to metabolize or remove
drug from the circulation. In its simplest form, clearance
is proportional to the flow to a single organ (Q) and to
the arterial-venous difference in drug concentrations compared with the amount of drug in the arterial circulation,
expressed as
C
− Cvenous
CL = Q × arterial
Carterial
Total body clearance usually reflects the combined clearance of multiple organs with different enzyme activities and
different rates of blood flow. Clearance can be measured by
the rate of appearance of drug outside the body (similar to
urinary creatinine clearance) or by the rate of disappearance
of drug from the circulation compared with the circulating
concentration. For calculations, clearance (CL) is defined as
the dose divided by the area under the plasmaconcentration
versus AUC and by the rate of drug input per Css average,
where rate of input is the dosing interval (τ) and Css is the
steady-state concentration. For a drug administered by continuous infusion, this is simply the infusion rate (mg/kg)hr−1
divided by Css as follows:
Dose Dose/τ Infusion rate
CL =
=
=
AUC
Css
Css
Once clearance is known, this equation can be rearranged to solve for the dose necessary to achieve any
desired steady state concentration:
Infusion rate = Css × CL
−1
−1
(mg/kg) h = (mg/L) × (mL/kg) h
Clearance changes significantly for some drugs during fetal and infant development because the activity of
metabolic enzymes increases with advancing gestational
and postnatal age. Values for clearance and volume of distribution at different stages of preterm development are
available for a few drugs and can be used to estimate the
doses needed to achieve and maintain therapeutic concentrations associated with desired clinical responses.
Studies of the analgesic fentanyl illustrate the developmental changes in its kinetics and how they can be used
to calculate dosages to reach and maintain concentrations
associated with effective analgesia. Analgesia has been associated with a serum fentanyl concentration of 1 to 2 ng/mL
(Santeiro et al, 1997). If analgesic treatment is initiated with
a continuous infusion of fentanyl, five half-lives are needed
to reach a steady state. The fentanyl half-life ranges from
3 hours in term newborns to 12.7 hours in premature newborns (Koehntop et al, 1986; Santeiro et al, 1997). Because
of this prolonged half-life, the patient may be inadequately
CHAPTER 34 Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics
TABLE 34-1 Fentanyl Development Kinetics
Gestational Age (wk)
Clearance at 0-47 Hours
after Birth (mL/[min·kg])
29
9.6
33
11.4
37
13.2
41
15.0
Data from Saarenmaa E, Neuvonen PJ, Fellman V: Gestational age and birth weight
defects of plasma clearance of fentanyl in newborn infants, J Pediatr 136:767-770, 2000.
treated for a long time, unless a loading dose is administered to reach an effective concentration more rapidly. In
general, the initiation of analgesic treatment and increases
in infusion doses of analgesics should begin with a loading
dose based on the estimated volume of distribution in the
central compartment (circulation) and desired concentration. The use of a loading dose shortens the time to reach
higher effective analgesic concentrations, but also increases
the likelihood of toxicity, as has been reported with digoxin.
Limited data are available regarding the gestational age–
related changes in fentanyl clearance, but two studies show
that it increases with advancing gestational age (Koehntop
et al, 1986; Santeiro et al, 1997) and with increasing age
after birth (Gauntlett et al, 1988; Santeiro et al, 1997). The
linear graph of clearance versus gestational age from 38
neonates who began treatment within 47 hours after birth
was used to derive mean rates of clearance at different gestational ages, as shown in Table 34-1.
Other investigators studied single-dose fentanyl kinetics
during anesthesia and found an apparent central volume of
distribution of fentanyl in neonates of 1.45 L/kg (Koehntop
et al, 1986). Note that this distribution volume is smaller
than the steady-state volume of distribution of 5.1 L/kg,
also calculated after a single dose of fentanyl (Koehntop
et al, 1986). In turn, the apparent steady-state volume of
distribution after a single bolus dose of a lipophilic drug is
usually smaller than that associated with continuous drug
infusions, during which tissues throughout the body become
saturated with drug. The steady-state distribution volume
for fentanyl during continuous infusions was calculated as
17 L/kg (Santeiro et al, 1997). It should be noted that because
fentanyl is highly lipid soluble, it distributes rapidly from
the central compartment into the peripheral tissue compartment. This large distribution volume likely reflects the
period during the infusion when the drug is leaving the circulation to penetrate peripheral tissues, such as fat. Because
it may take 15 to 60 hours to achieve a steady-state concentration (five half-lives) after a fentanyl infusion is begun or
the infusion rate is increased, a patient may need repeated
bolus doses to maintain effective plasma concentrations in
the central compartment. The best approach is to repeat
the calculated loading dose until the desired clinical effect is
achieved. This also illustrates why, for sedation specifically,
dosing should be adjusted to achieve the desired clinical
effect. Clearance calculations, however, can guide the starting doses to achieve effective sedation, as illustrated later.
The kinetic parameters for fentanyl in premature infants
reported by Koehntop et al (1986) can be used to calculate a
loading and infusion dose to reach a fentanyl concentration
425
of 2 ng/mL, which is considered an analgesic concentration
(Saarenmaa et al, 2000). Fentanyl analgesic concentration is
estimated for a premature newborn at a gestational age of
33 weeks (note that ng/mL is equivalent to μg/L) as follows:
C (μg/L) =
Loading dose (μg/kg)
Vdcentral (L/kg)
Loading dose = 2 µg/L × 1.45 L/kg
= 2.9 µg/kg
[1]
[2]
Infusion rate (µg/kg × h) = C (µg/L) × CL (mL/kg × h)
= 2 µg/L × 11.4 mL/kg/min ×
1 L/1000 mL × 60 min/h
= 1.4 µg/kg × h
[3]
Two studies have observed rises in fentanyl clearance
with increasing postnatal age (Gauntlett et al, 1988; Santeiro et al, 1997). This postnatal rise in clearance of fentanyl
likely relates either to maturation of cytochrome P450 3A4
(the enzyme responsible for fentanyl metabolism) activity or to increased hepatic blood flow after birth, because
fentanyl has a high hepatic extraction rate. For drugs like
fentanyl with a high hepatic extraction ratio, the rate-limiting factor in clearance is the flow of blood to the liver
(Saarenmaa et al, 2000). Some researchers have observed
that increased intraabdominal pressure reduces fentanyl
clearance, which is likely caused by reduced hepatic blood
flow (Gauntlett et al, 1988; Koehntop et al, 1986). Clinical changes known to increase or decrease fentanyl clearance should be used to adjust starting dosages, but dosing
should be adjusted primarily for the desired clinical effect.
MODELING AND SIMULATIONS
Pharmacokinetic modeling and simulations can be used to
evaluate the effects of important developmental changes
and disease processes upon pharmacokinetic parameters
for distribution volume, elimination rate, and clearance.
These models can identify clinical situations and conditions
when doses are likely to require modification. Mathematical simulations create theoretical pharmacokinetic profiles
for patients after a dose using the range of pharmacokinetic parameters determined from a patient population.
These can then be calculated for 100 to 1000 hypothetical patients to define the expected range of concentrations
that are likely after a dose. For drugs, such as antiinfectives
with which serum concentrations have been correlated
with effectiveness, this provides estimates of how large a
dose is needed to reach effective concentrations. A recent
study of fluconazole kinetics in newborns illustrates the
application of this process (Wade et al, 2008).
PHARMACOKINETIC PRACTICAL
EXAMPLES
ESTIMATED DOSE ADJUSTMENTS
Gentamicin and phenobarbital can be used to illustrate
the practical application of the principles of pharmacokinetics and therapeutic drug monitoring already discussed.
426
PART VIII Care of the High-Risk Infant
The calculations can be performed with standard arithmetic calculators and provide close enough estimates of the
kinetics for drugs with a long half-life to adjust dosages at
the bedside.
GENTAMICIN
Assume that optimal gentamicin concentrations are
Peak = 6 to 10 µg/mL
Trough = 0.5 to 2 µg/mL
After the fourth dose (2.5 mg/kg) of gentamicin to an
edematous premature newborn, the peak concentration
was 5.0 μg/mL; 18 hours later, the trough was 2.5 μg/mL.
It appears that the distribution volume is greater than
anticipated, because the peak concentration is lower
than expected, and the half-life is longer than anticipated
because the trough is higher than expected. The time of
drug administration and blood sampling were confirmed
(an important step), so the half-life is 18 hours, because the
concentration decreases 50% from 5.0 to 2.5 μg/mL in 18
hours, assuming that the kinetics are linear and first-order.
Vd (mL/kg) =
=
Dose (mg/kg)
Δ C (μg/mL) × 1 mg/1000 μg
2.5 mg/kg × 1000 μg/mg
(5.0 − 2.5) (μg/mL)
= 1000 mL/kg
To ensure a trough concentration of 2.0 μg/mL or
less, doses are administered every two half-lives or every
36 hours. When two half-lives have passed after the fourth
dose, the gentamicin concentration should be approximately 1.25 μg/mL (50% of 2.5 μg/mL). Increasing the
concentration from the 1.25 μg/mL (trough) to greater
than 6 μg/mL (peak) requires a concentration difference
of 4.75 μg/mL or greater. With a distribution volume
of 1000 mL/kg, a dose of 4.75 mg/kg should raise the
concentration from a trough of 1.25 μg/mL to a peak of
6.00 μg/mL. In one half-life, this concentration will
decrease to 3.0 μg/mL, and in two half-lives or 36 hours
to 1.5 μg/mL. An additional 4.75-mg/kg dose will raise the
peak concentration to 6.25 μg/mL, which will fall to 3.12
μg/mL in one half-life and to 1.6 μg/mL in two half-lives.
The variation between the peak and trough concentrations
after the last dose is within the measurement error for gentamicin and should achieve the optimum concentrations
defined previously.
the phenobarbital concentration was checked daily. The
results were as follows:
ll 24 hours: 40 μg/mL
ll 48 hours: 31 μg/mL
ll 72 hours: 25 μg/mL
ll 96 hours: 21 μg/mL
The maintenance dose (7 mg/kg) was resumed immediately after the 21 μg/mL concentration was measured and
produced a peak concentration of 30 μg/mL after administration of the dose. These concentrations and doses can be
used to calculate the volume of distribution and a dose to
maintain the phenobarbital concentration between 20 and
30 μg/mL as follows:
Dose (mg/kg)
Vd (L/kg) =
Δ C (μg/mL = mg/L)
=
=
7.0 (mg/kg)
(30 − 21) (mg/L)
7 mg/kg
9 mg/L
= 0.78 L/kg
Half-life can be determined from inspection, because
the concentration decreased from 50 to 25 μg/mL in
72 hours; therefore it should take 72 hours for the concentration to decrease by one half-life from 30 to 15 μg/mL.
The concentration will decrease approximately 5 μg/mL
every 24 hours, or one third of a half-life. Dividing the halflife into fractions is an approximation because it estimates
the change in concentration as linear rather than exponential. To be more accurate, the concentration decreases
59% in half of one half-life. Although this approximation
violates certain principles of pharmacokinetics, it allows
estimation of the change in concentration for each one
third of a half-life as one third of the change during one
half-life. Therefore the concentration decreases approximately 5 μg/mL in 24 hours. The following approach can
be used to estimate the daily phenobarbital dose needed to
return the concentration to 30 μg/mL, a change in concentration of 5 μg/mL:
Dose (mg/kg)
Δ C (mg/L) =
Vd (L/kg)
5 mg/L =
Dose (mg/kg)
0.78 L/kg
3.6 mg / kg = Dose (mg/kg)
PHENOBARBITAL
DRUG-INDUCED ILLNESS
Seizures that were difficult to control developed in a 3.6-kg
asphyxiated newborn. Seizures continued after two 20-mg/kg
phenobarbital doses until an additional 10-mg/kg dose
was administered. A maintenance dose of 7 mg/kg per day
was started 24 hours after the loading doses were administered. At 10 days, the child was increasingly somnolent.
The phenobarbital level measured in a blood specimen
taken 2 hours after administration of the oral maintenance
dose was 50 μg/mL. Additional doses were withheld, and
The extensive exposure of newborns to drugs in the NICU
is not benign. Neonates in the NICU have higher rates
of medication errors and potential adverse error rates
(91 in 100 admissions) than do neonates in other areas
of the hospital, and physician orders are responsible for
74% of errors and 79% of potential errors (Kaushal et al,
2001). Physician reviewers judged more than 90% of these
errors to be preventable, and 16% were potentially lifethreatening or fatal (e.g., heparin and digoxin overdoses).
CHAPTER 34 Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics
Similar problems have been observed worldwide (Kunac
et al, 2009). The causes of this drug-related morbidity and
mortality are complex. Pharmacologic studies in pediatric
patients are difficult because of a variety of problems ranging from ethics to study design (Ward and Green, 1988).
The difficulty of studying therapeutics in the newborn has
created a situation in which a plethora of drugs is administered with a paucity of pharmacologic data. For the smaller
and more immature newborns who are now surviving, gestational age–appropriate pharmacologic data for efficacy,
dose-response, and kinetics for most drugs remain limited
and need further study (Ward and Kern, 2009).
Furthermore, drug-induced illness is seldom considered
in newborns. Failure to recognize drug-induced illness in
the newborn often leads to further pharmacologic treatment as the first approach to correct unrecognized druginduced problems. This fact may reflect an expectation that
drug therapy is usually effective and safe. Prudent management of newborns must recognize and weigh the potential
benefits of unstudied drug therapy against potential druginduced adverse effects, morbidity, and mortality. Some
examples from the history of drug-induced mortality and
morbidity in newborns should serve as a reminder of how
more harm than good may accrue from uncontrolled or
unstudied drug therapy in the NICU.
LESSONS FROM CHLORAMPHENICOL
Chloramphenicol was released for use in the 1940s, and
reports of its efficacy for treatment of Salmonella spp.
infections included pediatric patients. The manufacturer recommended doses of 50 to 100 mg/kg per day for
patients weighing 15 kg or less. When Sutherland (1959)
reported three cases of sudden death in newborns treated
with high doses of chloramphenicol (up to 230 mg/kg per
day), the drug was considered “well tolerated and nontoxic.” Later the same year, Burns et al (1959) reported
the disturbing results of a controlled trial of the following
four prophylactic treatment regimens for newborn sepsis:
(1) no treatment, (2) chloramphenicol alone, (3) penicillin and streptomycin, and (4) penicillin, streptomycin,
and chloramphenicol. The groups that received chloramphenicol (100 to 165 mg/kg per day), in regimens 2 and 4,
had overall mortality rates of 60% and 68%, respectively,
whereas groups receiving regimens 1 and 3 had mortality
rates of 19% and 18%, respectively. The deaths of these
newborns demonstrated the stereotyped sequence of symptoms and signs caused by chloramphenicol, designated the
gray syndrome, which consisted of abdominal distention
with or without emesis, poor peripheral perfusion and
cyanosis, vasomotor collapse, irregular respirations, and
death within hours of the onset of these symptoms. Weiss
et al (1960) attributed the gray syndrome in newborns to
high concentrations of chloramphenicol secondary to its
prolonged half-life in newborns who received dosages of
more than 100 mg/kg per day, which are usually used in
older children. They recommended maximum doses of
50 mg/kg per day in term infants younger than 1 month,
half that dose for premature infants, and careful monitoring of chloramphenicol blood concentrations.
The discovery and explanation of chloramphenicol
toxicity in newborns illustrate several important aspects
427
of neonatal pharmacology. Because chloramphenicol was
considered well tolerated in older children and adults, it
was regarded as nontoxic for newborns. Chloramphenicol
was so effective in newborns that higher doses were used
without pharmacokinetic study. Higher doses were administered to newborns despite recognition that its clearance
required glucuronide conjugation, which was known to
be immature in newborns. The unexpected finding that
chloramphenicol in doses of 100 to 165 mg/kg per day
could be lethal to newborns was demonstrated because the
study conducted by Burns et al (1959) included appropriate control groups. Because the mortality rate from the
most effective antibiotic treatment regimen was equivalent
to that of no antibiotic treatment, these investigators discontinued prophylactic use of antibiotics in the nursery.
Similar pharmacologic comparisons are needed for
other drugs used in newborns. Additional thoughtful consideration should be given to clinicians‘ response to therapeutic failure. Fewer drugs and lower doses may be safer
and more effective than additional drugs in higher dosages.
REDUCTION AND PREVENTION
OF MEDICATION ERRORS
IN NEWBORN CARE
Drug treatment is one of the most common approaches
used in the care of sick newborns. At Primary Children’s
Medical Center 35-bed NICU in Salt Lake City, Utah,
with an average of 785 patient-days per month, patients
receive an average of 8700 (range 6990 to 11,290) doses
of medications, pharmacy-formulated intravenous solutions, and aerosols each month (unpublished observations,
1994). These doses are usually prepared by pharmacists
and administered by nurses, respiratory therapists, and
(rarely) physicians. In such a large and complex system
that produces so many drug treatments per month, errors
are virtually inevitable despite several levels of prospective
and redundant reviews by nurses, pharmacists, and NICU
unit secretaries involved in the drug treatment process.
At Primary Children’s NICU, the medication error rate
averages 0.04% for nurses and pharmacists and 0.07% for
physicians (unpublished observations, 1994). Many errors
are inconsequential, whereas others have serious adverse
effects. Medication errors incur significant costs, ranging from the obvious ones such as direct patient injury,
prolonged hospital stays, and additional corrective treatments to the more subtle costs associated with monitoring
and regulation of medication use within hospitals (ASHP,
1995).
In a study of 393 malpractice claims reported to the
Physician Insurers Association of America, the secondmost common cause of malpractice claims was drug errors
(Physician Insurers Association of America, 1993). Among
16 medical specialties with two or more claims, pediatric
practice ranked sixth in the number of claims, yet it had
the third highest average cost per indemnity. The medications most frequently involved in all claims were antibiotics, glucocorticoids, narcotic or non-narcotic analgesics,
and narcotic antagonists. In pediatric practice, the medications most frequently involved were vaccines (diphtheria-pertussis-tetanus) and bronchodilators (theophylline).
In the Physician Insurers Association of America review
428
PART VIII Care of the High-Risk Infant
(Physician Insurers Association of America, 1993), the five
most common causes of drug errors were as follows:
ll Incorrect doses
ll Medications that were inappropriate for the medical
condition
ll Failure to monitor for drug side effects
ll Failure of communication between physician and
patient
ll Failure to monitor drug levels
The primary opportunity for prevention of these five
most common errors rests with the prescribing physician.
Additional information and time for communication and
documentation may be needed.
Prescriptions and drug orders are a means of communicating, but clinicians often devote too little attention
to making them legible, clear, and unambiguous (ASHP
guidelines on preventing medication errors in hospitals,
1993). Physicians should keep the following recommendations in mind to ensure that their medication orders communicate more effectively:
ll Write instructions in full rather than with abbreviations.
ll Avoid vague instructions (e.g., “take as directed”).
ll Specify exact dose strengths.
ll Avoid abbreviations of drug names (e.g., MS could
mean morphine sulfate or magnesium sulfate).
ll Avoid U as an abbreviation for units, because the U may
be mistaken for a 0 (zero).
ll Avoid trailing zeroes (e.g., 5.0 mg).
ll Use leading zeroes (e.g., 0.5 mg).
ll Minimize the use of verbal orders.
ll Ensure that prescriptions and signatures are legible,
even if it means printing the name that corresponds to
the signature.
The process for ordering, preparing, dispensing, and
administering medications in an ICU with acutely ill
patients is often complicated and may contribute directly
to errors. The frequency of those errors, however, may be
reduced in almost every NICU. Although complex and
expensive computerized systems may help reduce medication errors, caregivers can take steps that are completely
within their control to reduce medication errors without
waiting for changes in the entire pharmacy process within
the hospital (ASHP guidelines on preventing medication
errors in hospitals, 1993).
DRUG EXCRETION IN BREAST MILK
The excretion of drugs in breast milk remains a source of
confusion and concern for many physicians and families.
Newer analytic techniques and more thorough pharmacokinetic studies have improved the available data in this area
of neonatal pharmacology. The available data regarding
drug exposure of the newborn through human milk have
been organized, in decreasing levels of concern, from drugs
that are associated with adverse effects on the infant during nursing to those that are of concern pharmacologically
to those that have not been associated with problems during nursing. The list of drugs clearly contraindicated during nursing is surprisingly short (American Academy of
Pediatrics, Committee on Drugs, 2001). On average, the
breastfeeding infant receives approximately 2% to 3% of a
maternal dose through milk. Drugs that are organic bases
or are lipid soluble may reach higher concentrations in
milk than in maternal serum.
SUMMARY
The extensive drug exposure of the sick newborn in the
NICU is dangerous because of the frequency of adverse,
sometimes fatal, drug reactions. Unfortunately, in the rapidly changing fetus and newborn, drug therapy is often
empiric because of a lack of gestational age–appropriate
kinetic data. Methods appropriate for the study of therapeutics in newborns present unique difficulties, but a
review by Ward and Green (1988) may provide assistance
for investigators. Drug therapy of newborns requires practical application of the principles of pharmacokinetics and
pharmacodynamics—which describe the processes of drug
absorption, distribution, metabolism, and excretion—to
the estimation and individualization of dosages.
SUGGESTED READINGS
American Academy of Pediatrics: Committee on Drugs: The transfer of drugs and
other chemicals into human breast milk, Pediatrics 108:776-789, 2001.
ASHP guidelines on preventing medication errors in hospitals: Am J Hosp Pharm
50:305-314, 1993.
Buxton ILO: Pharmacokinetics and pharmacodynamics: the dynamics of drug
absorption, distribution, action, and elimination, In Brunton LL, Lazo JS,
Parker KL, editors: Goodman and Gilman’s the pharmacological basis of therapeutics, ed 11, New York, 2006, McGraw-Hill, pp 1-39.
Ingelman-Sundberg M: Genetic polymorphisms of cytochrome P450 2D6
(CYP2D6): clinical consequences, evolutionary aspects and functional diversity,
Pharmacogenomics J 5:6-13, 2005.
Kaushal R, Bates DW, Landrigan C, et al: Medication errors and adverse drug
events in pediatric inpatients, J Am Med Assoc 285:2114-2120, 2001.
Kearns GL, Abdel-Rahman SM, Alander SW, et al: Developmental pharmacology—
drug disposition, action, and therapy in infants and children, N Engl J Med
349:1157-1167, 2003.
Kunac DL, Kennedy J, Austin N, et al: Incidence, preventability, and impact of
Adverse Drug Events (ADEs) and potential ADEs in hospitalized children in
New Zealand: a prospective observational cohort study, Paediatr Drugs 11:153160, 2009.
Lacroix D, Sonnier M, Moncion A, et al: Expression of CYP3A in the human
liver—evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth, Eur J Biochem 247:625-634, 1997.
Leeder JS, Kearns GL: Pharmacogenetics in pediatrics. Implications for practice,
Pediatr Clin North Am 44:55-77, 1997.
McCracken GH: Aminoglycoside toxicity in infants and children, Am J Med
80(Suppl 6B):172-178, 1986.
Rowland M, Tozer TN: Clinical pharmacokinetics and pharmacodynamics: concepts and
applications, Baltimore, 2010, Williams and Wilkins.
Spector R, Park GD, Johnson GF, et al: Therapeutic drug monitoring, Clin
Pharmacol Ther 43:345-353, 1988.
Wade KC, Wu D, Kaufman DA, et al: Population pharmacokinetics of fluconazole
in young infants, Antimicrob Ag Chemo 52:4043-4049, 2008.
Ward RM, Lugo RA: Drug therapy in the newborn, In MacDonald MG, Seshia
MMK, Mullett MD, editors: Avery’s neonatology: pathophysiology and management
of the newborn, ed 6, Philadelphia, 2005, Lippincott Williams and Wilkins, pp
1507-1556.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
35
Neonatal Pain and Stress: Assessment
and Management
Dennis E. Mayock and Christine A. Gleason
Relief of human suffering is one of the most important
goals of all health care providers. Advances in neonatology have significantly improved neonatal morbidity and
mortality, but pain, discomfort, and stress remain sad
realities for babies in the neonatal intensive care unit
(NICU). Assessing, managing, and trying to limit these
clinical realities, particularly while caring for critically
ill neonates, are challenging and increasingly controversial. Fortunately there has been considerable clinical and
laboratory research and much clinical dialogue aimed at
developing the best clinical practices in this problematic
arena. This chapter describes the developmental biology,
history, and public policies that have informed and shaped
current clinical practices; it also summarizes relevant clinical and basic research regarding clinical assessment tools
and both pharmacologic and nonpharmacologic management approaches. Finally, future directions in this field are
discussed.
HISTORY AND DEVELOPMENT
OF PUBLIC POLICIES
ll
ll
ll
ll
ll
HISTORICAL TIMELINE: NEONATAL PAIN
MANAGEMENT
Like all challenging medical issues, it is important to know
the history and current practice of neonatal pain management to understand what direction research should take in
the future. Management of neonatal pain and stress serves as
an excellent example of this philosophy; therefore a brief history of neonatal pain management follows, beginning with
the isolation of morphine from the opium poppy, which has
been used to treat pain since approximately 3500 bc.
ll 1806: Morphine (named for Morpheus, god of dreams)
isolated from opium poppy by Friedrich Serturner (a
pharmacist) and used to treat pain
ll 1960s to 1980s: Infants believed to be too immature to
feel pain; adverse effects of anesthetics feared; Liverpool method (pancuronium only, no anesthesia) used
widely for patent ductus arteriosus (PDA) ligation in
premature infants
ll 1985: Landmark paper published by Anand et al (1985)
describing adverse physiologic effects of Liverpool
method and improved outcomes using anesthesia for
PDA ligation; anesthesia and postoperative pain medication begin to become more widely used in neonatal care
ll 1987: Joint statement issued by the American Academy
of Pediatrics (AAP) and American Society of Anesthesiologists (ASA) regarding safety of (and necessity for)
operative anesthesia and postoperative analgesia for
neonates, regardless of their age or maturity
ll 1990s: Use of morphine infusions in the NICU expands
from operative and postoperative pain relief to include
ll
ll
ll
ll
ll
preemptive sedation, particularly during mechanical
ventilation
1999: Pilot study (NOPAIN) suggesting that neurologic
outcome is improved if preemptive morphine infusions
are used in a neonate receiving mechanical ventilation
2000: AAP and Canadian Pediatric Society issue joint
statement regarding neonatal pain management; primarily directed at surgical anesthesia and postoperative
and procedural pain assessment and relief (American
Academy of Pediatrics Committees on Fetus and Newborn and on Drugs, Sections on Anesthesiology and on
Surgery et al, 2000)
2001: International evidence-based group develops
consensus guidelines for prevention and management
of neonatal pain, most of which are pharmacologically
based
2003: The Joint Commission issues mandate regarding
pain assessment and management in all hospitalized
patients, including neonates
2004: Cochrane Review (Stevens et al, 2004) supports
use of oral sucrose for procedural analgesia
2004: NEOPAIN trial results published (Neurologic
Outcomes and Pre-emptive Analgesia in Neonates), concluding that preemptive morphine infusion did not
improve outcomes in neonates receiving ventilation
and did not relieve procedural pain
2005: Cochrane Review (Bellù et al, 2005) states “There
is insufficient evidence to recommend routine use of
opioids in mechanically-ventilated newborns.”
2005: Lee et al (2005) conclude that “fetal perception
of pain is unlikely before the 3rd trimester” and “little
or no evidence addresses the effectiveness of direct fetal
anesthetic or analgesic techniques.”
2006: Cochrane Review (Shah et al, 2006) recommends
breastfeeding or oral breast milk for neonatal procedural pain
2009: More than 40 infant pain assessment tools
available around the world; most were developed for
research and have not been validated for clinical use
2009: First national practice guidelines developed
in Italy for procedural pain management for NICU
patients
DEVELOPMENT OF PUBLIC POLICY
Since the late 1980s, in response to a public outcry regarding the recognition and management of pain in hospitalized patients, mandates have been promulgated by the
U.S. Department of Health and Human Services, The
Joint Commission, and other professional organizations.
The initial public policy statement regarding pain management for neonates undergoing surgical interventions was
issued as a joint communication by the AAP and the ASA
429
430
PART VIII Care of the High-Risk Infant
(American Academy of Pediatrics Committees on Fetus
and Newborn and on Drugs, Sections on Anesthesiology
and on Surgery, 1987; American Society of Anesthesiologists, 1987). This statement made it clear that anesthesia
and analgesia could be given relatively safely to neonates
despite their age or cortical immaturity. A second influential document was issued by the Acute Pain Management Guideline Panel of the U.S. Agency for Health Care
Policy and Research (Acute Pain Management Guideline
Panel, 1992; Agency for Health Care Policy and Research
Pain Management Guideline Panel, 1992). This statement
unequivocally endorsed the need for pain management in
neonates. The result of the publication of these two documents was the initiation of research studies into the prevention and amelioration of neonatal pain.
The most influential of these documents came from
The Joint Commission. The 2003 accreditation standards
required health care providers to look across the continuum of life, including the neonatal period, at the complex
nature of the pain experience so as to create new foundations for care (Joint Commission International Accreditation Standards for the Care Continuum, 2003). However,
these guidelines do not provide specific instructions for
assessing or managing pain in neonates. Neonatal caregivers needed to assess and treat perceived neonatal pain
and discomfort, but had little research-based evidence on
which to base their assessment and therapy. Organizations
have commissioned interdisciplinary teams to incorporate
regulatory directives and results of scientific investigation
into institutional practice guidelines and standards for care
(Anand, 2001; Howard et al, 2008a, 2008b; Lago et al,
2009). These guidelines include a patient’s right to regular
and systematic assessments of pain, interventions to relieve
pain, evaluation of effectiveness of interventions, attention
to long-term pain management needs, deleterious effects
of unmanaged pain, and educational needs of families and
staff members who provide care (American Pain Society,
2006; Bell, 1994; Carrier and Walden, 2001; Howard
et al, 2008a, 2008b; Joint Commission on Accreditation of
Healthcare Organizations, 2004; Lago et al, 2009).
As a result of these public policy initiatives and regulations, new tools for pain assessment and innovative
methods to treat pain have been developed and evaluated.
However, many old questions remain unanswered and new
concerns have been raised. Neonates can suffer both acute
and chronic pain. Treatment protocols for acute pain may
not be appropriate for chronic pain, because the origin
and resultant physiologic status can be quite different.
Long-term use of narcotics and other drugs leads to drug
tolerance and the need to wean slowly to avoid drug withdrawal. The drugs themselves, as well as drug tolerance
and drug withdrawal, may all contribute to adverse effects
on brain development and neurodevelopmental outcomes.
RECENT SURVEYS OF CLINICAL PRACTICE
Surveys of clinical practice have shown that attention to neonatal pain management has improved over the last decade,
but they also demonstrate that much more needs to be done.
This concern is evidenced by three recent surveys from
NICUs. Prestes et al (2005) surveyed four Brazilian units in
October 2001 prospectively. They found that, of 91 neonates
admitted to the NICU, there was documentation of systemic
analgesia in only 25% of 1025 patient days. No specific
drug was administered during arterial, venous, and capillary
sticks, lumbar punctures, or intubation. Only 9 of 17 surgical patients received any postoperative analgesia (Prestes et
al, 2005). Taylor et al (2006) surveyed 10 NICUs in North
America to determine their use of postoperative analgesia.
Data were collected for 25 consecutive postoperative admissions. Nurses documented pain assessments for 88% of the
infants, whereas physicians documented pain assessments
for only 9%. The study concluded that “documentation
of postoperative pain assessment and management in neonates was extremely variable” and called for development of
evidence-based guidelines for postoperative care and education of professional staff. Carbajal et al (2008) prospectively
studied 430 infants admitted to tertiary NICUs in Paris
between September 2005 and January 2006. Each neonate
averaged 115 total procedures (75 of which were considered
to be painful) and 16 procedures per day (10 of which were
considered to be painful). Of a total of 42,413 painful procedures, 2.1% were performed with pharmacologic therapy;
18.2% with nonpharmacologic therapy; 20.8% with pharmacologic, nonpharmacologic, or both; and 79.2% without
specific analgesia. All three of these recent survey studies
found that despite current knowledge of pain assessment and
treatment methods, most infants still are not provided any
specific treatment to alleviate pain and discomfort
ONTOGENY AND DEVELOPMENT
OF PAIN AND STRESS RESPONSES
The sensory system of the neonate, especially the preterm
infant, is immature (Figure 35-1). Afferent input from both
noxious and non-noxious stimuli terminate in the dorsal
horn of the spinal cord in a diffuse manner on multiple
cells; this results in the inability to distinguish between
noxious and non-noxious stimuli and limits the care provider’s ability to correctly interpret the infants’ behavioral
response. In the neonatal rat, separation of sensory input is
not completed until 3 to 4 weeks after birth (approximately
1 to 2 years in humans; Beggs et al, 2002); this prevents the
newborn from differentiating touch from painful sensory
input. The responses of the infant are therefore nonspecific. With repeated painful exposures, infants may lose any
discriminatory ability and develop hypersensitive states for
long periods of time. This hypersensitivity persists even if
non-noxious stimuli are introduced (Evans, 2001; Jennings
and Fitzgerald, 1998). The responses are less synchronized
in the immature central nervous system are also noted in
the immature central nervous system because of underdeveloped myelination and slower synaptic transmission as
manifested in longer and more variable latencies (Fitzgerald, 2005; Jennings and Fitzgerald, 1998).
Neuronal connections within the cortex appear to form at
approximately 22 weeks’ gestation (Kostovic and JovanovMilosevic, 2006), suggesting that higher cortical level pain
processing may be limited despite the presence of a behavioral response (Fitzgerald and Walker, 2009). In addition,
the neonate lacks sufficient descending modulatory control,
thereby limiting their ability to benefit from endogenous
control over noxious stimuli compared with adults (Fitzgerald and Koltzenburg, 1986; Hathway et al, 2006).
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
431
Excitatory
interneuron
4
2
Lamina II
neuron
A-fibre
input
Late
embryonic
Inhibitory
interneuron
Epidermis
Dermis
A-fibre central terminal fields
C-fibre central terminal fields
5
Descending
input
Excitatory
interneuron
C-fibre
input
1
Lamina II
neuron
A-fibre
input
Newborn
Inhibitory
interneuron
Peripheral nerve
Spinal
cord
DRG
Epidermis
Dermis
N52
3
trkA
Descending
input
Excitatory
interneuron
C-fibre
input
Lamina II
neuron
Adult
Inhibitory
interneuron
Epidermis
Dermis
trkA N52
IB4
FIGURE 35-1 1, In early postnatal life, descending fibres are present, but inhibitory and excitatory influences are weak or absent. The connections
gradually strengthen, becoming fully functional at the end of the third postnatal week. 2, A-fibres are the first primary afferents to enter the dorsal
horn gray matter and are present during the last few embryonic days. Their distribution is diffuse with exuberant, more superficial projections gradually retracting over the first 3 postnatal weeks. C-fibres are present in the dorsal horn during late embryonic stages, but only enter the gray matter 2 to
3 days before birth. Unlike A-fibres, they project to topographically appropriate regions in lamina II of the spinal cord as soon as they enter. C-fibre
synaptic connectivity is present, although weak at the time of birth, with connections strengthening over the first 2 postnatal weeks. 3, At birth the
majority (approximately 80%) of dorsal root ganglion (DRG) neurons express the nerve growth factor (NGF) receptor trkA. Over the first postnatal
week, this population reduces, with approximately half of these neurons losing their trkA expression and beginning to express receptors for glial cell
line-derived neurotrophic factor (GDNF) (identifiable as the IB4-binding population). 4, The balance of excitation and inhibition in the superficial
dorsal horn develops postnatally, through changes in both local interneuron circuitry and descending fibres. A-fibre input is stronger in the neonate
and weakens as the influence of C-fibre input increases. 5, Primary afferent innervations of the skin occurs earlier than central projections. By late
embryonic stages, primary afferents of all classes have reached the skin and innervate through the dermis into the epidermis. These projections die
back during the immediate perinatal period to leave the full adult situation of dermal innervations present soon after birth. (From Beggs S, Fitzgerald
M: Development of peripheral and spinal nociceptive systems. In Anand KJ, Stevens BJ, McGrath PJ, editors: Pain in neonates and infants, ed 3, Philadelphia,
2007, Elsevier, p 15.)
432
PART VIII Care of the High-Risk Infant
Noxious stimuli in adults result in the release of inflammatory and trophic factors that activate and sensitize nociceptors in the injured tissue. Such noxious stimuli lead to
nociceptive afferent input to the central nervous system,
exciting nociceptive circuits in the spinal cord, brainstem, thalamus, somatosensory cortex, cingulate cortex,
and amygdale (Tracey and Mantyh, 2007; Woolf and Ma,
2007). However, noxious stimuli in infants do not evoke
similar patterns of central nervous system activity as noted
in adults (Fitzgerald, 2005). The response to noxious
stimuli is more diffuse and less spatially focused in infants.
Studies in rats have demonstrated major alterations in neuronal circuitry with maturation, appearing to mirror that
seen in humans but in a compressed time period. These
animal data may parallel developmental changes that have
been noted in humans (Fitzgerald, 2005). Local tissue
injury resulting from repeated heel sticks and invasive procedures triggers increased proliferation of nerve endings in
surrounding tissues, particularly when this damage occurs
early in gestation. As a result, scars (e.g., from heel sticks,
old intravenous sites) and surrounding tissues can remain
hypersensitive well beyond the neonatal period (Jennings
and Fitzgerald, 1998; Reynolds and Fitzgerald, 1995). Pain
assessment and management are thus most challenging in
preterm infants who are exposed to the stressful NICU
environment for long periods (Johnston and Stevens,
1996).
In summary, the immature nervous system in preterm infants lacks the ability to discriminate consistently
between noxious and non-noxious stimuli, reacts often
with similar behavior to a variety of stimuli, lacks the ability to modulate pain responses, and does not consistently
manifest signs or symptoms that allow care providers to
accurately assess the infant’s level of pain and discomfort.
Furthermore, infants cannot verbally report their level of
pain. This later challenge remains a significant obstacle to
accurate assessment of pain in the newborn and determination appropriate treatment.
ASSESSMENT OF NEONATAL PAIN
AND STRESS
DEVELOPMENT OF BEHAVIORAL RESPONSES
TO PAIN
Tools for assessment of neonatal pain and stress must be
based on an understanding of the normal development of
behavioral responses to pain and stress—an infant is not
a small adult. Behavioral responses to noxious stimuli in
infants are not always predictable because of immaturity of the central nervous system; therefore assessment
of pain and response to therapeutic intervention can be
similarly unpredictable. Significant structural and functional changes occur in pain pathways during development, many of which continue to take place after birth (see
later discussion of ontogeny and development of pain and
stress responses). When assessing pain and discomfort in
infants, especially those born prematurely, it is important
to remember that the patients are nonverbal and cannot
say where and how badly they hurt. Indeed, even if they
could verbalize such information, their ability to accurately localize and describe the pain would be limited as
discrimination between inputs (tactile or nociceptive) is
immature. Moreover, their behavioral response repertoire
is also limited and is not always predictable. These limitations make the use of assessment tools problematic, especially when trying to determine whether there has been a
response to a drug or other intervention.
INFANT PAIN SCORES
Over the past two decades, more than 40 infant pain scales
have been developed (Ranger et al, 2007). Most of these
scales were developed for use in clinical trials to assess efficacy of various pharmacologic therapies and have not been
validated for general clinical use. However, the use of these
scales in clinical practice has led to better health care provider recognition that procedural pain and stress are common. Moreover, it is assumed that this pain and distress
can be ameliorated by appropriate use of pharmacologic
and nonpharmacologic approaches. However, assessment
and treatment of chronic pain and discomfort in infants
remains problematic. The pain instruments developed for
assessment of acute pain were not designed or validated for
the chronic pain and discomfort associated with mechanical ventilatory support, or for use in paralyzed or neurologically compromised infants (Anand, 1998). Furthermore,
painful stimuli may be processed at the cortical level in
infants without producing detectable behavioral changes
(Slater et al, 2008). Therefore the clinician is left to decide
whether the touted benefits of opiate sedation during
mechanical ventilation in newborns outweigh the possible
adverse effects, both acute and long term. The importance
of selecting a valid, reliable, and practical pain assessment
tool is evident from the current literature. These tools
assess the response to noxious stimuli by categorizing the
behavioral or physiologic reactions of the infant, or a combination of both (Grunau and Craig, 1987). The behavioral
responses can include limb movements, muscle tone, crying, and characteristic facial expressions. The physiologic
measures can include heart rate, oxygen saturation, and
respiratory rate. Of note, specific facial expression changes
have been determined to be the most reliable indicators
of pain (brow bulge, eye squeeze, nasolabial furrow, taut
lips, and open mouth) whereas crying is the least reliable
indicator (Grunau and Craig, 1987; Guinsburg et al, 1997;
Stevens et al, 1993). Use of facial expression changes can
be challenging when the infant’s face is partially covered
with adhesives to secure tubes and lines, or with a phototherapy mask in place.
A recent publication lists the current available pain
assessment tools that can be used in neonates (Stevens
et al, 2007). The most commonly used tools are listed in
Table 35-1.
Many pain assessment tools are available; however, they
have not been systematically reviewed, and there is no specific gold standard for comparison. Because each instrument requires training and practice for optimal use, care
providers should choose one that best fits the infant population being assessed to allow for consistency. The optimal
approach to pain management should include reducing the
frequency of painful procedures, reducing environmental
stressors, facilitating neurologic developmental, determining the best technique to minimize the pain and stress
433
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
TABLE 35-1
Infant Pain Scales
Measure
Age Level
Indicators
Pain Type
Psychometric Properties
Unidimensional Behavioral Measures of Infant Pain
Baby facial action coding
system (Rosenstein and
Oster, 1988)
Term infants
Facial actions based on adaptation from adult work
Procedural
Interrater reliability (r = 0.65-0.85)
Infant body coding system
(Craig et al, 1993)
Preterm infants
(32 wk GA) to
term infants
Movement of hand, foot, arm,
leg, head, torso
Procedural
Interrater reliability (r = 0.83), face
validity, content validity
Neonatal facial coding
system (Grunau and
Craig, 1987)
Preterm infants
(>25 wk
GA) to term
infants
Brow bulge, eye squeeze, nasolabial furrow, open lips, horizontal mouth, vertical mouth,
lips pursed, taut tongue, chin
quiver, tongue protrusion
Procedural
Interrater reliability (r = 0.88),
content validity, face validity,
construct validity, convergent
validity, feasibility
Multidimensional Pain Measures in Infants
Children’s and infants’
postoperative pain scale
(Büttner and Finke,
2000)
Birth to 4 years
Crying, facial expression, posture of the trunk, posture of
the legs, motor restlessness
Prolonged
(postoperative)
Interrater reliability (<3 yr old
subsample; r = 0.64-0.77), internal
consistency (<1 yr old subsample;
r = 0.96), content validity, construct and concurrent validity
demonstrated in older subsample
Modified behavioral pain
scale (Taddio et al,
1995a)
2-6 months
Facial expression, cry, body
movement
Procedural
Interrater reliability (ICC = 0.95),
internal consistency (r = 0.55-0.66),
test-rest reliability (r = 0.95), content
validity, construct validity (p <0.01),
concurrent validity (r = 0.68-0.74)
Composite Pain Measures in Infants
Clinical scoring system
(Barrieret al, 1989)
1-7 months
Infant sleep during the preceding
hour, facial expression, cry,
motor activity, excitability, flexion, sucking, tone, consolability
Prolonged
(postoperative)
Interrater reliability (r = 0.79-0.88),
content validity, discriminant
validity (p <0.0001)
Modified postoperative
comfort score (Guinsburg et al, 1998)
<32 wk GA
(postnatal age
of 12-48 h)
Sleep, facial expression, sucking,
hyperreactivity, agitation,
hypertonicity, toe or finger
flexion, consolability
Prolonged
(mechanical
ventilation)
Convergent validity in bedside (p
<0.0001) and laboratory video
coding (p = 0.02), content validity,
divergent validity shown between
placebo and analgesic samples (p
< 0.05)
Echelle Douleur Inconfort
Nouveau-né (Debillon
et al, 2001)
26-36 wk GA
Facial expression, movement,
sleep, consolability
Prolonged
(postoperative)
Interrater reliability (r = 0.59-0.74),
content validity, construct validity
(p <0.000)
Modified postoperative
comfort score (Guinsburg et al, 1998)
Preterm infants
Sleep, facial expression, activity, tone, consolability, cry,
sociability
Prolonged
(postoperative)
Content validity, discriminant validity (p <0.0001)
Neonatal pain, agitation and sedation scale
(Hummel et al, 2008)
<28-35 wk GA
(age correction for
prematurity)
Crying, irritability, behavior
state, facial expression, extremities, tone, vital signs (heart
rate, respiratory rate, blood
pressure, oxygen saturation)
Prolonged
(mechanical
ventilation or
postoperative)
Preliminary reliability and validity
testing in progress
Pain assessment tool
(Hodgkinson et al, 1994)
27 wk GA to full
term
Posture, tone, sleep pattern,
expression, color, cry, respirations, heart rate, oxygen
saturations, blood pressure,
nurse perception
Prolonged
(postoperative)
Interrater reliability (r = 0.85),
content validity, convergent validity (r = 0.38), concurrent validity
(r = 0.76)
Scale for use in newborns
(Blauer and Gerstmann,
1998)
24-40 wk GA
Central nervous system state,
breathing, movement, tone,
face, heart rate, mean blood
pressure
Procedural
(excluded
postoperative)
Beginning indications of reliability,
content validity, discriminant
validity (p < 0.05-0.01)
Distress scale for ventilated
newborn infants (Sparshott, 1996)
—
Crying, requires increased
oxygen, increased vital
signs, expression, sleeplessness (Krechel and
Bildner, 1995)
Neonates 32-60
wk GA
Facial expression, body movement,
color, heart rate, blood pressure,
oxygenation, temperature
Crying, requires increased
oxygen, increased vital signs,
expression, sleeplessness
—
Face validity, content validity (Duhn
and Medves, 2004)
Prolonged
(postoperative)
Interrater reliability (r = 0.72),
content validity, concurrent validity (r = 0.49-0.73), discriminant
validity (p <0.0001), concurrent
validity for first 24 h (ICC = 0.340.65; McNair et al, 2004)
Continued
434
PART VIII Care of the High-Risk Infant
TABLE 35-1 Infant Pain Scales—cont’d
Measure
Age Level
Indicators
Pain Type
Psychometric Properties
Neonatal infant pain scale
(Lawrence et al, 1993)
Preterm and full
term
Facial expression, cry, breathing
patterns, arm movement, leg
movement, state of arousal
Procedural
Interrater reliability (r = 0.92-0.97),
internal consistency (0.87-0.95),
content validity, concurrent validity (r = 0.53-0.83)
Pain assessment in neonates scale GA (HudsonBarr et al, 2002)
26-47 wk GA
Facial expression, cry, breathing
patterns, extremity movement,
state of arousal, oxygen saturation, increased heart rate
Procedural
Content validity, concurrent validity
(r = 0.93)
Modified infant pain scale
(Bucholz et al, 1998)
4-30 wk old
Sleep during evaluation, facial
expression, quality of cry,
spontaneous motor activity,
excitability and responsiveness to stimulation, finger and
toe flexion, sucking, overall
tone, consolability, sociability,
change in heart rate, change
in blood pressure, fall in
oxygen saturation
Prolonged preceding hour
Interrater reliability (postoperative;
r = 0.85), content validity, convergent validity in dichotomous
rating (p <0.0001)
Premature infant pain profile (Stevens et al, 1996)
Term and preterm neonates
GA, behavioral state, heart
rate, oxygen saturation, brow
bulge, eye squeeze, nasolabial
furrow
Procedural
Interrater reliability (ICC = 0.930.96), intrarater reliability (ICC
= 0.94-0.98), internal consistency
(alpha = 0.59-0.76), content validity, construct validity (in preterm
neonates, p = 0.0001-0.02; in
term neonates, p <0.02), construct
validity in clinical setting (p
<0.0001), interrater reliability (r
= 0.94-0.98; Ballantyn et al 1999),
concurrent validity with cry duration (Johnston et al, 1999)
Modified from Stevens BJ et al: Assessment of pain in neonates and infants. In Anand KJ, Stevens BJ, MaGrath PJ, editors: Pain in neonates and infants, ed 3, Philadelphia, 2007,
Elsevier, pp 70-75.
GA, Gestational age.
associated with procedures, delegating responsibility for
pain assessment and treatment to the bedside nurse staff,
and using a balanced multimodal approach to pain control
(Allegaert et al, 2009).
BEDSIDE NONINVASIVE NEUROIMAGING
TO EVALUATE PAIN AND STRESS
Several new modalities are being evaluated for their capability to help health care providers recognize pain in
neonates. Near infrared spectroscopy might be a helpful technology. In a study of 29 infants between 26 and
36 weeks’ gestation at birth, cortical activation occurred
over both somatosensory cortices during unilateral tactile
and painful stimuli (Bartocci et al, 2006). Amplitude-integrated electroencephalography also can be used to detect
cortical activation in neonates (Toet and Lemmers, 2009).
These and other new technologies will be needed to first
recognize and then continually assess and manage pain and
discomfort in our fragile patients.
LONG-TERM CONSEQUENCES
OF NEONATAL PAIN AND STRESS
Untreated pain and stress has been linked to adverse longterm outcomes. Acute effects include elevations of cortisol, catecholamines, and lactate, hypertension, tachycardia,
respiratory instability, glucose instability, and changes
in cerebral blood flow. Chronic pain can affect growth,
immune function, recovery, and length of hospitalization.
In addition, a growing body of evidence has drawn attention to the potential deleterious effects of repeated handling, stress, and pain on long-term memory, social and
cognitive development, and neural plasticity (Anand, 1998;
Anand et al, 1987, 1989; Evans, 2001; Jennings and Fitzgerald, 1998; Pokela, 1994; Porter et al, 1999; Taddio et al,
1995a). Extrapolation of information from older children
and adults can be inappropriate for our most vulnerable
infants (Berde et al, 2005; Howard, 2003; Walker, 2008).
Pain and stress may alter neurodevelopment (Abdulkader
et al, 2008; Anand et al, 1987, 2000). Pain suffered during
neonatal intensive care has been associated with adverse
long-term outcomes, such as altered pain perception to
subsequent immunizations after circumcisions without
anesthesia (Taddio et al, 1997), abnormal cortisol responses
to stress in later infancy (Grunau et al, 2007), and altered
pain responses in childhood (Grunau et al, 1998). There
is concern that such hormonal changes might lead to the
development of cardiovascular disease and type 2 diabetes
in adulthood (Kajantie et al, 2002; Rosmond and Björntorp, 2000). As a result, appropriate use of sedation and
analgesia might be beneficial to infants requiring intensive
care support; however, a clear delineation of the benefits
still requires further study.
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
CLINICAL PAIN AND STRESS
MANAGEMENT STRATEGIES
SURGICAL ANESTHESIA
The recognition that surgical intervention in infants
results in dramatic physiologic and metabolic changes
similar to those noted in adults with significant pain has
changed current practice. It is well recognized that infants
perceive pain, and surgical intervention without anesthesia and analgesia leads to increased morbidity and excess
mortality; however, experience obtained in older children
and adults cannot be extrapolated to immature patients.
Providing general anesthesia in infants requires an intimate knowledge of the developmental status and function
of each organ system. A complete review of this subject is
beyond the scope of this chapter. Interested readers are
referred to A Practice of Anesthesia for Infants and Children
by Spaeth and Kurth (2009) or Anesthesia for Infants and
Children by Brett et al (2006).
FETAL SURGERY
The fetus has increasingly become a candidate for surgical
intervention to repair fetal anomalies such as congenital
cystic adenomatoid malformation of the lung, sacrococcygeal teratoma, and myelomeningocele, and to treat fetal
disorders such as severe anemia secondary to hemolytic
diseases. Many fetal interventions are accomplished using
general maternal anesthesia, and therefore fetal anesthesia,
but certain procedures are attempted with maternal analgesia and local anesthesia. Thus, during such fetal surgical interventions, the fetus requires consideration for pain
management to attenuate fetal physiologic and hormonal
stress responses (Fisk et al, 2001; Giannakoulopoulos
et al, 1999). Anand et al (1987) summarized the available
evidence regarding fetal and neonatal nociceptive activity in 1987 and put all care providers on notice that late
gestation fetuses and newborns have intact cortical and
subcortical centers necessary for pain perception and demonstrate physiologic responses to painful stimuli similar to
evidence in adult subjects. A recent review of the literature
supports the contention that the fetus perceives pain (Lee
et al, 2005). The International Association for the Study of
Pain supported the view that the human fetus is capable of
reacting to nociceptive stimuli during the second trimester
(Anand, 2006).
NEONATAL SURGERY
Selection of anesthetic agents to be used in neonates must
account for the developmental status and function of each
organ system and the potential adverse and toxic effects
of specific anesthetic agents. Animal models have demonstrated that anesthetic agents can be both neuroprotective
and neurotoxic in the immature brain. Frequently used
anesthetics act by two principal mechanisms, either by
decreasing excitation via N-methyl-d-aspartate (NMDA)
receptors (e.g., ketamine, nitrous oxide) or by increasing
inhibition via gamma-aminobutyric acid receptors (e.g.,
benzodiazepines, barbiturates, propofol, etomidate, isoflurane, enflurane, halothane). In the immature rat brain,
435
drugs that act by either of these two mechanisms can
induce widespread neuronal apoptosis when given during
the period of synaptogenesis (Ikonomidou et al, 1999; Ishimaru et al, 1999). The applicability of these findings to the
human infant has been questioned (Soriano and Anand,
2005); however, recent data suggest that some toxicity may
occur in human infants (Wilder et al, 2009).
Regional anesthesia for neonatal surgery is commonly
used for minor interventions. As more experience with
regional anesthesia for major surgical procedures is gained,
such practice is expanding. Bösenberg (1998) presented
results of the use of epidural analgesia for major neonatal
surgical interventions in 240 infants weighing between 900
and 5800 g and reported that all infants had effective intraoperative analgesia with only two complications. Advantages of regional anesthesia may include less postoperative
apnea, lees need for intubation, and better postoperative
pain control. Sethna and Suresh (2007) reviewed this subject in depth recently.
POSTOPERATIVE PAIN MANAGEMENT
STRATEGIES
Pain management after surgical intervention, like acute
pain management, requires knowledge of the developmental status and function of each organ system and the
potential adverse and toxic effects of specific analgesic
agents. Many neonates remain intubated for mechanical ventilation after major surgery. Analgesia can be provided via intravenous administration of an analgesic agent,
either by intermittent bolus dosing or by continuous infusion. Another option is continuation of regional analgesia
(Bösenberg, 1998). Regional analgesia appears to be effective for pain control, but sedation may also be required
in active or vigorous infants (Frumeinto et al, 2000).
Regional anesthesia, when used alone, may also reduce the
incidence of postoperative apnea in preterm infants (Craven et al, 2003).
Research-based evidence of appropriate postoperative pain management in neonates is limited. Taylor et al
(2006) surveyed 10 NICUs regarding their postoperative
pain assessment and management practices; they found
that pain assessment documentation was extremely variable. Nursing documentation was done for most infants,
whereas few physicians documented any assessment. Most
infants were treated with opioids, benzodiazepines, or
both, and some infants (7%) received no analgesia despite
recent major surgery (Taylor et al, 2006). Van der Marel
et al (2007) evaluated the use of rectal acetaminophen as
an adjuvant treatment to continuous morphine infusion
in postoperative neonates and could not demonstrate
any additional analgesia effect. Recent evidence suggests
that neonates require significantly less morphine to control pain and discomfort than older infants do, based on
monitoring of pain scale data. Bouwmeester et al (2003a,
2003b) determined that neonates required less morphine
for postoperative pain control and that the dose requirement increased with age. Both studies found that morphine, given by bolus or continuous infusion, was equally
effective. The later study (2003b) found that mechanical ventilation decreased morphine metabolism and
clearance.
436
PART VIII Care of the High-Risk Infant
The Association of Pediatric Anesthetists of Great Britain and Ireland commissioned guideline development
for pain management after surgery and painful medical procedures (Howard et al, 2008a, 2008b). The level
of evidence from the literature was determined and the
recommendations were graded, allowing for better interpretation. These guidelines provided evidence-based
recommendations and listed best clinical practice points
when published evidence was insufficient to make formal
recommendations.
MECHANICAL VENTILATION
The use of mechanical ventilation in neonates with respiratory failure is a fairly common practice in the United
States, although decreasingly less than in the 1980s and
1990s. In older children and adults who require mechanical ventilation, sedation is routinely provided—most often
with opiates (Gélinas et al, 2004). However, pharmacologic sedation is used less frequently in the adult intensive
care unit because of concern regarding adverse cognitive
outcomes and longer duration of ventilator support (Izurieta and Rabatin, 2002; Oderda et al, 2007). Extrapolation
of evidence from past studies in adult patients led to the
routine use of opiate sedation in neonates during mechanical ventilation (Kahn et al, 1998), with limited information
regarding safety and efficacy (Anand et al, 2004; Simons
et al, 2003b). As noted previously, the ability to assess and
treat discomfort and pain is limited, and preemptive use of
pharmacologic sedation during mechanical ventilation in
newborns, especially preterm infants, remains controversial (Anand and Hall, 2007).
Mechanical ventilation in neonates is associated with an
increase in hormonal stress responses, including increased
cortisol and catecholamine levels (Guinsburg et al, 1998;
Quinn et al, 1998). In the past, infants who appeared
uncomfortable while receiving ventilatory support demonstrated asynchronous respiratory effort (i.e., “fighting” the
ventilator), compromised gas exchange, and altered stress
responses (Dyke et al, 1995). Furthermore, pain and stress
in newborns receiving mechanical ventilation have been
associated with decreased pulmonary compliance, atelectasis, and intrapulmonary shunting (Bolivar et al, 1995).
More recently, with the introduction and use of surfactant
replacement therapy and synchronized ventilatory technology, some of the problems with fighting the ventilator
have been eliminated (Claure and Bancalari, 2009; Keszler, 2009).
Clinical studies from the 1990s demonstrated that opiate treatment prevented these adverse effects on neonatal
ventilation and reversed the previously described hormonal stress changes (Quinn et al, 1993; Saarenmaa et al,
1999). Opiate sedation has been demonstrated to decrease
stress scores in newborns who receive mechanical ventilation (Orsini et al, 1996; Quinn et al, 1993, 1998). In fullterm infants receiving mechanical ventilation, the severity
of respiratory failure as assessed by the oxygenation index
directly correlated with the need for analgesia and sedation
(Aretz et al, 2004).
A small randomized trial of routine morphine infusion in preterm infants receiving mechanical ventilation
concluded that morphine lacked a “measurable analgesic
effect” and there was “absence of a beneficial effect on poor
neurological outcome” (Orsini et al, 1996). The larger
clinical trial that followed (i.e., NEOPAIN) reported
no beneficial effect of preemptive morphine infusions in
ventilated preterm infants and an increased incidence of
severe intraventricular hemorrhage in preterm infants
born at 27 to 29 weeks’ gestation and receiving morphine
(Anand et al, 2004). Bhandari et al (2005) found that morphine infusions had no beneficial effects in preterm infants
receiving mechanical ventilation. Additional bolus doses
of morphine resulted in worse respiratory outcomes and
longer requirement for ventilatory support. None of these
controlled clinical trials provide evidence that routine
narcotic sedation during mechanical ventilatory support
in neonates is beneficial. In addition, the well-recognized
and common adverse effects of narcotic exposure (e.g.,
hypotension, feeding intolerance, respiratory depression)
are mentioned infrequently in sedation and pain treatment protocols in neonates receiving mechanical ventilation. Future trials have been deemed unethical if they
involve withholding sedation from a group of infants. One
approach to this dilemma would be to minimize the use of
ventilatory support as much as possible.
A recent Cochrane Review evaluated the effects of opioid analgesics on pain, duration of mechanical ventilation,
mortality, growth, and development in neonates requiring mechanical ventilation (Bellù et al, 2008). The authors
found no differences in mortality, duration of mechanical
ventilation, and short- and long-term neurodevelopmental
outcomes. Preterm infants given morphine took longer to
achieve full enteral feeding. If morphine sedation prolongs
ventilatory support needs and time to full enteral feeds,
then an increase in the risk of complications related to the
use of venous lines (bloodstream infections) and parenteral nutrition (cholestasis) should be expected (Menon
et al, 2008). Hällström et al (2003) studied risk factors for
necrotizing enterocolitis in premature infants and found
that the duration of morphine use was the strongest predictor for development of severe necrotizing enterocolitis. The Cochrane Review’s overall conclusion regarding
the use of sedation during mechanical ventilation was that
“there is insufficient evidence to recommend routine use
of opioids in mechanically ventilated newborns” (Bellù
et al, 2008). Menon and McIntosh (2008) came to a similar
conclusion in their recent review.
PROCEDURES
Infants undergoing intensive care suffer through many
painful procedures, often several times each day. Although
new pharmacologic and nonpharmacologic treatment
strategies to decrease or eliminate some of this pain and
stress have been developed, there is still a need to develop
better management techniques. An article by D’Apolito
(2006) reviews this issue in detail and outlines where
knowledge remains limited.
BLOOD SAMPLING AND MONITORING
Heel sticks are routinely performed to obtain blood samples in neonates. The most appropriate method for relieving pain from a heel stick is yet to be determined. The heel
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
should be warmed to aid blood sampling. Eutectic mixture
of local anesthetics (EMLA) cream does not relieve the
pain of a heel lance (Stevens et al, 1999b; Taddio et al,
1998). Shah et al (1997) and Larsson et al (1998a, 1998b)
demonstrated that neonates experiencing venipuncture
had lower pain scores than those who underwent heel stick
for blood sampling. In selected neonates, venipuncture
should be used preferentially over heel stick.
The pain of arterial puncture can be decreased by infiltrating the site with 0.1 to 0.2 mL of 0.5% or 1% lidocaine using the smallest-gauge needle possible (Franck
and Gregory, 1993). Buffering the lidocaine with sodium
bicarbonate is recommended to decrease the burning
caused by lidocaine. In addition, EMLA may reduce the
pain of arterial puncture.
TRACHEAL INTUBATION
The use of premedication to minimize the pain and stress
of intubation has been demonstrated to benefit neonates
(Simons et al, 2003a). However, concerns about rapid
medication availability, ability to maintain the airway, and
the ability to provide ongoing ventilatory support continue
to cause controversy (Carbajal et al, 2007). Premedications
typically include atropine, narcotics for sedation, and muscle relaxants. Atropine abolishes vagal bradycardia. Narcotics attenuate the increases in arterial blood pressure.
Muscle relaxants attenuate the increases in intracranial
pressure. Combinations decrease the time and number of
attempts needed to intubate the infant.
When considering the use of medications for intubation, several questions need to be asked:
ll Does the infant have adequate vascular assess?
ll What is the urgency of intubation need?
ll Is the infant known to have a difficult airway?
ll When was the last feeding?
ll Can the infant be preoxygenated while avoiding gastric
distension?
If the decision is made to use medications for intubation,
typical doses include:
ll Atropine 0.02 mg/kg fast intravenous (IV) push
ll Fentanyl 2 μg/kg slow IV push
ll Vecuronium 0.1 mg/kg IV push
Alternative medications may include succinylcholine
(1 to 2 mg/kg IV push) or rocuronium (1 mg/kg IV push).
The investigational agent sugammadex can reverse
rocuronium-induced neuromuscular blockade in less than
under 2 minutes. Sugammadex forms a 1:1 complex with
steroidal nondepolarizing neuromuscular blockers in the
plasma. Although approved for use in Europe, sugammadex is not currently available in the United States; it might
render succinylcholine unnecessary. Rocuronium could be
administered and if intubation is unsuccessful, paralysis
could be immediately reversed with sugammadex (de Boer
et al, 2007).
Propofol has been used as a premedication for intubation. Being a single agent, it is easier and faster to prepare
than three separate drugs. Propofol is a hypnotic agent
without anesthetic properties. If combined with remifentanil, there is no need for paralysis in older children (Batra
et al, 2004); however, propofol is painful when injected
in small veins, and extremely painful if it extravasates.
437
A major advantage is continued spontaneous breathing
during the intubation procedure. The dose is 2.5 mg/kg
IV; this dose might need to be repeated. Concerns over
the use of propofol for intubation in neonates include
minimal experience in neonates, uncertain pharmacokinetics and duration of action, and compatibility with precutaneously inserted central catheters lines.
Because intubation can raise both blood pressure and
intracranial pressure, a short-acting benzodiazepine,
such as midazolam, can be beneficial for infants with stable cardiovascular function. Fentanyl can be used as an
alternative for infants with compromised cardiovascular
function (McClain and Anand, 1996). Any infant who is
pharmacologically paralyzed during mechanical ventilation should receive adequate sedation. In addition, the
infant should receive pain medication if pain is suggested
by the infant’s condition or because of the procedures
being performed.
CIRCUMCISION
The AAP Circumcision Policy Statement (American
Academy of Pediatrics Task Force on Circumcision, 1999)
states that analgesia must be provided to infants undergoing circumcisions. EMLA cream, dorsal penile nerve block,
and subcutaneous ring block are all possible options. The
AAP reports that subcutaneous ring block may provide
the best analgesia (American Academy of Pediatrics Task
Force on Circumcision, 1999). Subcutaneous ring block
has been found to be more effective than EMLA or dorsal
penile nerve block in other studies (Lander et al, 1997).
Dorsal penile nerve block has been found to be more effective than EMLA, but this method is not always available
(Lee and Forrester, 1992).
EMLA has been established as superior to placebo for
pain relief during circumcision (Benini, 1993; Taddio
et al, 1997). An effective method for applying EMLA in
preparation for circumcision is to apply one third of the
dose to the lower abdomen, extend the penis upward gently, pressing it against the abdomen, and then apply the
remainder of the dose to an occlusive dressing placed over
the penis. This dressing is then taped to the abdomen so
that the cream surrounds the penis. Another method is to
apply the cream and then place plastic wrap around the
penis in a tubelike fashion to direct the urine stream out
and away from the cream.
Acetaminophen is ineffective for the management of
severe pain associated with the circumcision procedure,
but it provides some analgesia in the postoperative period.
Acetaminophen has been found to decrease pain 6 hours
after circumcision (Howard et al, 1994).
OTHER INVASIVE PROCEDURES
Placement of a central venous catheter requires topical
anesthesia with EMLA or infiltration of the skin with lidocaine. In addition, a parenteral opioid, such as morphine or
fentanyl, is typically required. Consideration should also
be given to regional blocks for central line placement.
The pain of a lumbar puncture is compounded by both
the needle puncture and the distress caused by the body
position required for the procedure. EMLA has been
438
PART VIII Care of the High-Risk Infant
shown to decrease the pain of lumbar puncture in children (Halperin et al, 1989). Chest tube insertion requires
an intravenous opioid, adequate local analgesia (lidocaine),
or both.
NONOPIOID ANALGESICS
Administering 10 mg/kg may be inadequate for pain
control, because this dose is based on antipyretic doseresponse studies. The maximum recommended daily dose
is 75 to 90 mg/kg for infants, 45 to 60 mg/kg for term and
preterm neonates more than 32 to 34 weeks of postconceptual age, and 25 to 40 mg/kg/day for preterm neonates
28 to 32 weeks of postconceptual age (Berde and Sethna,
2002; Morris et al, 2003).
Rectally administered acetaminophen has a longer halflife, but absorption is highly variable because it depends
on the individual infant and placement of the suppository.
It should also be noted that the suppository may contain
all of the drug in its tip and should be divided lengthwise
if a partial dose is desired. The analgesic effect of acetaminophen may be additive when the agent is administered with opioids. This coadministration may enable a
decrease in the opioid dose and therefore in corresponding opioid side effects; however, demonstration of this
potential benefit awaits further study (van der Marel
et al, 2007).
Nonsteroidal Antiinflammatory Drugs
(Indomethacin, Ibuprofen)
OPIOID ANALGESICS
PHARMACOLOGIC ANALGESIA
INTERVENTIONS
The severity of the pain, etiology, available administration
routes, and consideration of potential side effects should
all be evaluated during selection of an analgesic. Once
medication administration has begun, careful monitoring for side effects can decrease potential adverse events
related to administration of pain medications to infants.
A key component of effective pain management is reassessment after a painful intervention, although this is difficult
to do with limited pain assessment tools.
Nonsteroidal antiinflammatory drugs (NSAIDs) inhibit
prostaglandin synthesis by inhibiting the action of cyclooxygenase enzymes. Cyclooxygenase enzymes are responsible
for the breakdown of arachidonic acid to prostaglandins.
NSAIDs have many physiologic effects, including sleep
cycle disruption, increased risk of pulmonary hypertension, cerebral blood flow alterations, decreased renal
function by decreasing glomerular filtration rate, alteration in thermoregulatory control, and changes in platelet
function. Moreover, development of the central nervous,
cardiovascular, and renal systems is dependent on prostaglandins. These adverse effects are particularly worrisome
for neonates and infants; however, these drugs are used
frequently in the NICU for pharmacologic closure of the
PDA. Aside from effects on renal and perhaps mesenteric
circulation, which are difficult to separate from the PDA,
no clear side effects have been reported.
Acetaminophen
Acetaminophen is the most widely administered analgesic
in patients of all ages. Acetaminophen inhibits the activity
of cyclooxygenase in the central nervous system, decreasing
the production of prostaglandins, and peripherally blocks
pain impulse generation (Arana et al, 2001). Neonates are
able to form the metabolite that results in hepatocellular
damage (Arana et al, 2001); however, it is inappropriate to
withhold acetaminophen in newborns because of concerns
of liver toxicity. The immaturity of the newborn’s cytochrome P-450 system may actually decrease the potential
for toxicity by reducing production of toxic metabolites
(Collins, 1981).
Current recommendations are for less frequent oral
dosing (every 8 to 12 hours in preterm and term neonates),
because of slower clearance times, and higher rectal dosing because of decreased absorption (Arana et al, 2001;
van Lingen et al, 1999). Typical oral doses for acetaminophen are 10 to 15 mg/kg every 6 to 8 hours for term neonates and 10 to 15 mg/kg every 4 to 6 hours for infants.
Opioids are believed to provide the most effective treatment for moderate to severe pain in patients of all ages.
There is a wide range of interpatient pharmacokinetic
variability. Opioid dosing depends on the severity of the
pain as well as the age and clinical condition of the infant.
Opioids should be used in infants younger than 2 months
only in a monitored setting such as an intensive or intermediate care unit (Yaster et al, 2003). Some clinicians propose a more conservative recommendation, restricting the
use of opioids to monitored settings for any infant younger
than 6 months.
Morphine
Morphine remains the standard for pain management in
neonates, although not necessarily because it has been
shown to be the most effective analgesic. Morphine is
metabolized in the liver by uridine diphosphate glucuronyltransferase into two active metabolites: (1) morphine6-glucuronide (M6G), a potent opiate receptor agonist,
and (2) morphine-3-glucuronide (M3G), a potent opiate receptor antagonist. Both metabolites and some
unchanged morphine are excreted in the urine. The predominant metabolite in preterm and full-term neonates
is M3G. Because of slow renal excretion, the metabolites
can accumulate substantially over time (Bouwmeester
et al, 2003a, 2003b; Saarenmaa et al, 2000). There is
a potential for late respiratory depression because of a
delayed release of morphine from less well-perfused tissues
and the sedating properties of the M6G metabolite (Anand
et al, 2000).
Because the predominant metabolite of morphine in
infants is M3G, a potent opiate receptor antagonist, using
the lowest dose possible to achieve the needed analgesia
should be considered. Escalating morphine doses will
also increase the levels of M3G in the infant, interfering with the goal of adequate analgesia. Doses as low as
1 to 5 μg/kg/h can provide adequate analgesia, minimizing the risk of accumulation of high M3G levels with
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
the metabolite’s prolonged half-life (Bouwmeester et al,
2003a, 2003b).
Clearance or elimination of morphine and other opioids
is prolonged in infants, because of the immaturity of the
cytochrome P-450 system at birth. The rate of elimination
and clearance of morphine in infants 6 months and older
approaches that in adults. Chronologic age seems a better
indicator than gestational age of how an infant metabolizes
opioids (Scott et al, 1999; Yaster et al, 2003).
Infants are at greater risk for opioid-associated respiratory depression because of their immature responses to
hypoxia and hypercarbia. There is an increase in unbound
or free morphine and M6G available to reach the brain as a
result of the reduced concentration of albumin and alpha1
acid glycoproteins (Houck, 1998).
Hypotension, bradycardia, and flushing constitute
the response to the histamine release and rapid intravenous administration of morphine. Histamine release may
cause bronchospasm in infants with chronic lung disease,
although this is not commonly seen (Anand et al, 2000).
Morphine sedation may result in extended need for ventilatory support in neonates (Anand et al, 1999; Bhandari
et al, 2005).
Dosing recommendations currently reflect the wide
range of interpatient pharmacokinetic variability. In the
past, 0.03 mg/kg of morphine IV was suggested as a starting dose in infants not receiving ventilation (Acute Pain
Management Guideline Panel, 1992), whereas 0.05 to 0.1
mg/kg of morphine IV was recommended as an appropriate starting dose in infants receiving ventilation. Recently,
much lower doses have been recommended (Anand et al,
2008; Bouwmeester et al, 2003b, 2004; Lynn et al, 2000;
Saarenmaa et al, 2000). Titration to the desired clinical
effect is required in adjusting both the dose and the frequency of administration. Furthermore, it is important to
continually assess need and responses so that dosing can be
adjusted both up and down (Allegaert et al, 2009). As the
use of morphine for analgesia and sedation in neonates is
explored further, it is becoming clear that some of the risks
may outweigh the potential benefits (Allegaert et al, 2009;
Anand et al, 2008; Black et al, 2008; Carbajal et al, 2005;
Nandi et al, 2004; Ng et al, 2003; Ranger et al, 2007).
Fentanyl
Fentanyl is 80- to 100-fold more potent than morphine
and causes less histamine release, making it a more appropriate choice for infants with hypovolemia, hemodynamic
instability, or congenital heart disease. Another potential
clinical advantage of fentanyl is its ability to reduce pulmonary vascular resistance, which can be of benefit for infants
who have undergone cardiac surgery, have persistent pulmonary hypertension, or need extracorporeal membrane
oxygenation (Anand et al, 2000). Bolus doses of fentanyl
must be administered over a minimum of 3 to 5 minutes
to avoid chest wall rigidity, a serious side effect observed
after rapid infusion. Chest wall rigidity, which can result
in difficulty or inability to ventilate, can be treated with
naloxone or a muscle relaxant such as pancuronium or
vecuronium.
Fentanyl is highly lipophilic. It has a quick onset and
relatively short duration of action. Because of fentanyl’s
439
short duration of action, it is typically used as a continuous
infusion for postoperative pain. In infants 3 to 12 months
of age, total body clearance of fentanyl is greater than that
of older children, and the elimination half-life is longer
because of its increased volume of distribution (Singleton et al, 1987). Fentanyl has been demonstrated to have
a prolonged elimination half-life in infants with increased
abdominal pressure (Gauntlett et al, 1988; Koehntop et al,
1986). Because of tachyphylaxis, continuous infusions of
fentanyl are often increased to maintain constant levels of
sedation and pain management. Infusion dosing can reach
substantial levels requiring prolonged withdrawal.
A rebound transient increase in plasma fentanyl levels
is a phenomenon known to occur after discontinuation of
therapy in neonates. It is a result of the accumulation of
fentanyl in fatty tissues, which can prolong its effects after
continued use; therefore caution must be exercised in the
use of repeated doses or a continuous infusion.
Oral Opioids
Oral methadone can be used to wean infants from long-term
opioid use. Methadone is widely used in neonates and children, although there are limited data regarding its efficacy
and pharmacokinetics in this population (Chana and Anand,
2001; Suresh and Anand, 1998). The respiratory depressant effect of methadone is longer than its analgesic effect.
Methadone is metabolized slowly, and it has a long half-life.
Codeine is prescribed at 0.5 mg to 1 mg/kg orally every
4 hours as needed. Scarce data are available to recommend use of codeine in neonates. Most pharmacies supply
acetaminophen and codeine in a set formula, consisting of
acetaminophen (120 mg) and codeine phosphate (12 mg
per 5 mL) with alcohol (7%). The dose prescribed is limited by both the appropriate dose of codeine and the safe
dose of acetaminophen. This combination is not recommended in neonates.
Oxycodone dosing is 0.05 mg/kg to 0.15 mg/kg orally
every 4 to 6 hours as needed. No data are available to recommend oxycodone in neonates. The liquid form is not
universally available.
MIXED OPIOID AGONIST-ANTAGONIST
DRUGS
Nalbuphine is a mixed agonist-antagonist opioid receptor drug; therefore its administration in infants of opioidaddicted mothers may precipitate withdrawal. This agent
is equianalgesic with morphine. Nalbuphine has a ceiling
effect for analgesia. Additional studies are needed regarding the safety and efficacy of nalbuphine use in infants. It
is not recommended for use in neonates, although it may
be useful during opioid drug withdrawal (Jang et al, 2006).
LONG-TERM CONSEQUENCES OF NEONATAL
OPIOID EXPOSURE
Experimental Animal Studies
Perinatal and neonatal opioid exposure in experimental animals is associated with both short- and long-term
adverse neurologic effects that should make clinicians ask
440
PART VIII Care of the High-Risk Infant
whether the use of such medications with questionable
benefits should be used at all. Data from previous studies
suggested that perinatal narcotic exposure restricts brain
growth, induces neuronal apoptosis, and alters behavioral pain responses later in life (Handelmann and DowEdwards, 1985; Hu et al, 2002; Kirby et al, 1982; Seatriz
and Hammer, 1993). One area of particular concern to
clinicians is the developing cerebral circulation, which
is extremely vulnerable to physiologic perturbations
and the effects of drugs (Volpe, 1998). Cerebrovascular
effects of drug exposure early in development can have
lifelong consequences, including increased risk for stroke
(Barker, 2000; Craft et al, 2006; Hanson et al, 2004).
The acute effects of exogenous narcotics, including morphine, on the developing cerebral circulation have been
described in piglets and include modulation of prostaglandin-induced pial artery dilation during hypoxia,
alteration in endothelin production, and increases in
endothelin A receptor mRNA expression (Armstead
et al, 1990, 1996; Van Woerkom et al, 2004). Endogenous opioids are important regulators of cerebrovascular
tone and angiogenesis (Blebea et al, 2000; Gupta et al,
2002; Pasi et al, 1991; Poonawala et al, 2005). Exposure
to morphine in fetal sheep and neonatal rats permanently
alters cerebrovascular control mechanisms (Mayock et al,
2005, 2006). We have also shown permanent neurobehavioral and neuropathologic changes in a rodent model
of neonatal stress and morphine exposure (Boasen et al,
2009; McPherson et al, 2007; Vien et al, 2009). These
and other animal studies demonstrate short- and longterm effects of neonatal morphine exposure, which is not
surprising because opioid receptor-mediated signaling
likely has a role in several aspects of early brain development (Durrmeyer et al, 2010). However, the clinical
relevance of these animal studies regarding the long-term
effects of neonatal opioids is difficult because of species
differences in the timing of brain development, the development of opiate receptors and major neurotransmitter
systems, and the pharmacokinetics of administered opioids. Overall, it is difficult but necessary to simulate the
premature infant’s NICU experience in an animal model
(Durrmeyer et al, 2010).
Clinical Studies
Clinical studies addressing the short- and long-term
effects of prolonged opiate use in neonates are limited.
The few that exist are contradictory and confounded by
illness severity. Bergman et al (1991) described reversible
encephalopathic changes in neonates receiving long-term
sedative and narcotic infusions. MacGregor et al (1998)
demonstrated no adverse neurodevelopmental outcomes
in a small group of newborns who received morphine for
a median of 5 days. Roźe et al (2008) presented 5-year
neurodevelopmental outcomes in very low-birthweight
(VLBW) infants exposed to prolonged sedation or analgesia (defined as greater than 7 days of sedative or opioid
drugs). They found that exposed VLBW infants had more
severe or moderate disability at 5 years (42%) compared
with those not exposed (26%), but after adjusting for gestational age and propensity score (as a way to ensure that
treatment effects are only compared between infants who
are equally likely to receive that treatment), the association
was no longer significant. Preterm infants (23 to 32 weeks’
gestation at birth) evaluated at 36 weeks postconceptual
age in the NEOPAIN study already demonstrated neurobehavioral abnormalities if exposed to morphine during
ventilatory support (Rao et al, 2007).
TOPICAL AND LOCAL ANESTHETICS
Lidocaine reduces the pain and stress of venipuncture
and IV catheter placement (Larsson et al, 1998a, 1998b;
Long et al, 2003). EMLA cream has been demonstrated
to reduce the pain of circumcision. Place the cream on
the area where anesthesia is desired and then cover it
with an occlusive dressing for 1 hour before the procedure. Longer application times provide deeper local
anesthetic penetration, but can lead to toxicity. There
is a slight risk of methemoglobinemia with the use of
EMLA cream in infants and patients who are G6PD deficient. A rare occurrence, methemoglobinemia can occur
when hemoglobin is oxidized by exposure to prilocaine.
EMLA should not be used in patients with methemoglobinemia or in infants younger than 12 months who are
also receiving methemoglobinemia-inducing drugs, such
as acetaminophen, sulfonamides, nitrates, phenytoin,
and class I antiarrhythmics. (Refer to Table 35-2 for recommended maximum doses of EMLA cream by age and
weight.) A study of 30 preterm infants found that a single
0.5-g dose of EMLA applied for 1 hour did not lead to a
measurable change in methemoglobin levels (Taddio et
al, 1995b). A systematic review concluded that EMLA
diminishes the pain during circumcision. Limited efficacy was noted with pain from venipuncture, arterial
puncture, and percutaneous venous line placement.
EMLA was not found to diminish pain from heel lancing
(Taddio et al, 1998). Oral sucrose or glucose may be as
effective as EMLA for venipuncture (Abad et al, 2001;
Gradin et al, 2002).
SEDATIVES
Benzodiazepines
Benzodiazepines such as lorazepam and midazolam are
sedatives which activate gamma-aminobutyric acid receptors and should not be used in place of an appropriate
EMLA Cream: Recommended Maximum Dose
by Age and Weight
TABLE 35-2
Body
Weight
(kg)
Maximum
Total
EMLA
Dose (g)
Maximum
Application
Area (cm2)
Maximum
Application
Time (h)
Birth to
3 mo
or <5
1
10
1
3-12
mo
and
>5 kg
2
20
4
Age
Data from Taketomo CK, Hodding JH, Kraus DM, editors: Pediatric dosage handbook,
2001-2002, ed 8, Hudson, Ohio, Lexi-Comp Inc, p 595.
EMLA, Eutectic mixture of local anesthetics.
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
pain medication, because this class of medication has
no analgesic effect. For painful procedures, an analgesic
must be used in conjunction with the benzodiazepine.
Benzodiazepines are administered to decrease irritability
and agitation in infants and to provide sedation for procedures. In ventilated infants, benzodiazepines can help
to avoid hypoxia and hypercarbia from breathing out of
sync with the ventilator although, as noted for opioids,
new synchronized infant ventilators make this clinical
problem less likely. When given as continuous infusions,
dosing often escalates rapidly to maintain apparent sedation resulting in need for prolonged weaning. Use of such
medications has been associated with abnormal neurologic movements in both preterm (Lee et al, 1994) and
term infants (Chess and D’Angio, 1998). In rats, prenatal
exposure to diazepam results in long-term functional deficits and atypical behaviors (Kellogg et al, 1985); exposure
of 7-day-old mice to diazepam induces widespread cortical and subcortical apoptosis (Bittigau et al, 2002); and
midazolam potentiates pain behavior, sensitizes cutaneous
reflexes, and has no sedative effect in newborn rats (Koch
et al, 2008). Whether these data can be extrapolated to
human infants is unknown, but clinicians have reason to
be concerned and should use these drugs with caution in
the NICU.
Dexmedetomidine
Dexmedetomidine is a potent and relatively selective α2
adrenergic receptor agonist indicated for the short-term
sedation of patients in intensive care settings, especially
those receiving mechanical ventilatory support. The drug
is administered by either bolus doses for short procedural
sedation (1 to 3 μg/kg) or continuous intravenous infusion (0.25 to 0.6 μg/kg/h). Because dexmedetomidine
does not produce significant respiratory depression, it has
been used for procedural interventions in spontaneously
breathing infants (Barton et al, 2008; Chrysostomou et al,
2009). As neonatologists become more familiar with dexmedetomidine, its use may increase (O’Mara et al, 2009);
however, short- and long-term safety and effectiveness
information needs to be assessed in infants, as has been
initiated in adults (Pandharipande et al, 2007; Riker et al,
2009).
NONPHARMACOLOGIC ANALGESIC
INTERVENTIONS
Nonpharmacologic interventions for prevention or relief
of neonatal pain and stress are numerous and widely publicized in the medical literature. These interventions have
been used either as the sole method of pain control or
in combination with pharmacologic interventions. No
one would argue with the statement that neonatal intensive care is associated with stress, pain, and discomfort.
Because opioid analgesia and sedation have not been
proved to be efficacious and may possibly be harmful,
alternative methods of pain and stress relief need to be
evaluated for efficacy and safety. A variety of approaches
have been investigated. As stated clearly by Golianu et al
(2007), “These therapies may optimize the homeostatic
mechanisms of the infant, thereby mitigating some of
441
the adverse consequences of untreated pain, as well as
facilitating healthy physiologic adaptions to stress.” However, widespread adoption of specific techniques is not
consistent.
Nonnutritive sucking with pacifiers reduces pain
responses to heel prick, injections, venipuncture, and circumcision procedures (Sexton and Natale, 2009; Shiao
et al, 1997; South et al, 2005). Infant massage has been
demonstrated to decrease plasma cortisol and catecholamine levels in preterm infants (Acolet et al, 1993; Kuhn
et al, 1991).
Maternal skin-to-skin contact (also termed kangaroo
care) is associated with greater physiologic stability and
reduced responses to acute pain (Bergman et al, 2004;
Fohe et al, 2000; Gray et al, 2000; Johnston et al, 2003;
Ludington-Hoe and Swinth, 1996). Kangaroo care can
decrease Neonatal Infant Pain Scale scores after vitamin K
injections (Kashaninia et al, 2008). Maternal rocking has
been shown to diminish neonatal distress (Jahromi et al,
2004). Breastfeeding reduces the physiologic and behavioral responses to acute pain and stress in neonates and has
been recommended as the first line of treatment (Osinaike
et al, 2007; Shah et al, 2006).
The Neonatal Individualized Developmental Care and
Assessment Program (NIDCAP) systematically changes
a protocol-based model of nursing care to a relationshipbased approach (Als et al, 1994). There is a significant body
of empiric evidence that use of the NIDCAP approach
improves the clinical and neurodevelopmental outcomes
of preterm infants (Als and Gilkerson, 1997; Brown and
Heermann, 1997; Kleberg et al, 2002; Westrup et al,
2000; Wielenga et al, 2007), but recent metaanalyses of
published studies could not demonstrate any improvement
in long-term outcomes (Jacobs et al, 2002; Symington and
Pinelli, 2006).
Another approach is multisensory stimulation of
preterm infants undergoing painful procedures. This
approach entails simultaneous gentle massage, soothing
vocalizations, eye contact, smelling a perfume, and sucking
on a pacifier. This technique was associated with analgesia
and calming of the infants in several reports from one unit
(Bellieni et al, 2001, 2002, 2007).
Music therapy may reduce the behavioral and physiologic responses to acute procedural pain (Hartling et al,
2009).
Oral sucrose (versus intragastric) reduces pain behavior
in preterm and term infants and is used widely (Stevens et
al, 1997, 2004). The mechanism of oral sucrose analgesia
is believed to be the sweet taste stimulation of endogenous
opioid release (Shide and Blass, 1989). Of all methods
and techniques discussed, oral sucrose has been the most
widely used. As more data regarding the limitations of
pharmacologic treatment are published, consideration of
nonpharmacologic interventions will likely become more
important and commonplace.
IATROGENIC DRUG DEPENDENCE,
TOLERANCE, WITHDRAWAL
A clear distinction must be made between opioid or benzodiazepine dependence, tolerance, and addiction. Physical dependence is demonstrated by the need to continue
442
PART VIII Care of the High-Risk Infant
the administration of the drug to prevent signs or symptoms of physical withdrawal. Tolerance is a reduction in
the drug effects after repeated administration, or the need
to increase the dose to achieve the same clinical effect.
Addiction is compulsive drug-taking behavior (Gutstein
and Akil, 2001). Infants are not capable of becoming psychologically addicted, but they clearly develop tolerance
and dependence.
DRUG WEANING CONSIDERATIONS
Baseline pain and withdrawal scores should be obtained
before beginning the drug weaning process, and infants
should be reassessed every 2 to 4 hours for signs of withdrawal. In addition, when an opioid dosage is being
tapered, the infant should be assessed for the presence of
pain a minimum of every 4 hours. If an infant is receiving both an opioid and a benzodiazepine, it is prudent
to taper and stop only one class of medication at a time.
Typically, a weaning schedule is 10% of the total initial dose every other day. Many patients can tolerate a
relatively large initial decrease in dose, but subsequent
decreases may need to be smaller. Environmental stressors should be eliminated or reduced whenever possible.
It should be noted that the potential onset of withdrawal
symptoms varies according to the half-life of the opioid
or benzodiazepine and the half-life of active metabolites,
which may be much longer than that of the parent compound (Tobias, 2000).
PALLIATIVE CARE
Palliation, as defined in the New Oxford American Dictionary, is “making a disease (or its symptoms) less severe or
unpleasant without removing the cause.” Most deaths
in neonatal intensive care units occur after withdrawal
of supportive measures, when the burden of care is
extreme and the likelihood of recovery is remote (Cook
and Watchko, 1996; Wall and Partridge, 1997). Traditionally, initiation of palliative care occurs when further
medical interventions are no longer curative and death
of the infant is expected. However, the AAP and the
World Health Organization both support palliative care
that starts early in the course of an illness and works with
other therapies to prolong life. “The goal of palliative
care is the achievement of the best quality of life for the
patients and their families, consistent with their values,
regardless of the location of the patient” (World Health
Organization, 1998). Both the AAP (American Academy
of Pediatrics Committee of Bioethics and Committee on
Hospital Care, 2000) and the World Health Organization (1998) have provided guidelines for palliative care
for infants that emphasize optimizing quality of life until
death occurs and eliminating of the notion of euthanasia. Such care is an active process that includes familycentered care with attention to physical, emotional, and
spiritual issues (Papadatou, 1997). Several good reviews
of neonatal end-of-life care have been published (De
Lisle-Porter and Podruchny, 2009; Institute of Medicine
of the National Academies, 2003; Moro et al, 2006). Box
35-1 provides a comprehensive listing of the many aspects
of end-of-life care that should be considered for the dying
neonate.
Parents and other family members should be actively
involved in palliative care. Palliative treatments focus on
relief from clinical symptoms such as dyspnea, pain, agitation, vomiting, and seizures, in addition to maintaining
dignity and providing warmth. Palliative care should also
include attention relieving psychological stress in caregivers such as loneliness, depression, anxiety, grief, and
separation from family and loved ones. Family members
should be prepared for the end of life. They need to be
provided with a description of the dying process that
includes mention of the potential for gasping respiratory
efforts and an estimate of the length of the dying process.
Opportunities to create memories for the family should
be offered, such as pictures, footprints, locks of hair, hats,
or blankets.
Consideration of to the place of death is important.
Should it occur in the hospital or at home with hospice
care? Unfortunately, many deaths occur in busy NICUs
with little privacy. Placement of the infant and the family
on a postpartum ward with other parents and their healthy
infants is also suboptimal. Ideally, for deaths likely to occur
in the hospital, the infant and family should be placed in a
private room with enough space for all family members to
be present to support the parents in their grieving process.
Bereavement and follow-up discussions should be
planned with the family. Care providers should discuss
autopsy, discuss follow-up discussion times, and provide
ongoing bereavement support to the family for as long as
needed.
SUMMARY
Recognition and treatment of pain and discomfort in
the neonate remain challenging issues. Despite significant progress in the understanding of human neurodevelopment, pharmacology, and more careful attention to
the care of sick infants, there is still have much to learn.
Because protecting and comforting these patients are
important, and because external regulatory forces have
required intervention to minimize distress, what is known
about adult patients must be applied to infants. Some good
has come from these endeavors, but errors have been made
along the way.
It is important to minimize pain and distress in these
patients to avoid more aggressive interventions. Such care
will minimize the risks of adverse effects on neurodevelopment. Learning to provide good care without doing
harm should be the goal. Nonpharmacologic methods
of pain and distress control should be explored further.
When pharmacologic intervention is necessary for pain
control, use the least amount of drug that controls the
pain. Escalation of drug doses may, in fact, be adding to
the problem.
As newer techniques and medications are introduced to
clinical practice, it must be demonstrates that such additions achieve the goal of pain control. It must also be demonstrates that neonatal interventions are safe over the lives
of the patients. Better tools are needed to help optimize
the outcomes for infants.
CHAPTER 35 Neonatal Pain and Stress: Assessment and Management
443
BOX 35-1 Guideline for End-of-Life Care of the Neonate
I. Patient population
Any infant in the NICU diagnosed with a medical condition incompatible with
life.
II. Purpose
The guideline is for use by the bedside nurse in the NICU when faced with
caring for an infant at the end of life. It provides the bedside nurse with
alternative ways to encourage parents to perform normal parenting
activities, such as bathing, holding, and picture taking with their infant,
in the time surrounding his death. These activities may be the only, and
sometimes last, opportunity the mother and father will have to parent
their infant.
III. Background and rationale
Many NICUs have policies in place relating to bereavement and postmortem
care of the neonate, but few have policies or guidelines on palliative
care (Sudia-Robinson, 2003). Many nurses have not received sufficient
training to support families or themselves through this bereavement
process. Many nursing programs lack or have only recently added
curricula regarding end-of-life care (Romesberg, 2004). This lack of
protocols, along with the limited education available to professionals, serves to decrease the chance that infants and their families will
receive the end-of-life care they deserve. Palliative care consists of
three components: (1) pain and comfort management, (2) assistance
with end-of-life decision making, and (3) bereavement support (March
of Dimes, 2010).
IV. Procedure
A. Inclusion criteria: any infant in the NICU with a diagnosis
of nonviability or a fatal condition should be considered for
inclusion in this guideline. These medical conditions may
fall into three categories: (1) newborns born at the edge of
viability with extremely low birthweight (<500 g) and <24
weeks’ gestational, (2) newborns with complex or multiple
congenital anomalies, and (3) newborns not responding to
intensive care interventions and experiencing deterioration
despite medical efforts to save them (Catlin and Carter,
2002). For these infants, the focus of care should change
from curative to palliative.
B. Predeath: any intervention should focus on supporting the
parents during their infant’s end-of-life care. Some nurses
may be uncomfortable with their own feelings and have
trouble communicating with parents at this time. Communication, however, is vital, and the words spoken should serve
to validate the infant’s life and death (Capitulo, 2005). Communication also helps the parents understand their infant’s
condition and offers them the opportunity to make choices
concerning the extent of their participation in his end-of-life
care (Workman, 2001).
1. Communication
a. The following are suggestions for successful communication techniques to use with parents:
(1) Active listening
(2) Open communication
(3) Total presence
(4) Uninterrupted time for parents to express their
wishes
(5) Nonverbal gestures such as touch
b. Refer to the infant by name
c. Inform parents of what to expect while their infant is
dying (see, hear, smell, feel)
d. Inform parents of their options for disposition of their
infant’s remains
(1) Transfer to morgue
(2) Pick up by mortuary
e. Use language that does not confuse the parents.
(1) Use definite words such as death, die, and dying
instead of euphemisms such as not doing well or
passing away
(2) Words such as good, stable, or better could be misinterpreted to mean the infant could improve or survive
2. Environment
a. Do not restrict the number of visitors
b. Provide privacy and comfort
(1) Low lights
(2) Decreased noise and activity
3. Religion
a. Ask parents about any religious preference
b. Notify clergy of the parents’ choice
c. Offer baptism, blessing, anointing, or prayer
4. Parental activity
a. Provide a stuffed animal
b. Keep the family involved
(1) Decisions (medical and physical care)
(2) Handprints, footprints
(3) Kangaroo care, holding
(4) Bathing
(5) Dressing
(6) Changing diaper
(7) Any other parenting acts
(8) Taking pictures of the infant and family members at
any time during this process
C. Active dying: the time period when the infant is dying
offers special challenges for both the nurse caring
for the dying infant and his family members. Pain
management for the infant during the extubation and
dying process is of extreme importance. Parents must
be informed of what they may see, hear, and experience during this time. The family and infant should be
offered a comfortable, private area for this process.
1. Pain management
a. Maintain intravenous access
b. Medicate the infant appropriately
c. Assess pain routinely and frequently after the
withdrawal of life support
2. Communication
a. Inform the parents what to expect (see, hear,
smell, feel)
b. Inform the parents that their baby may not die
immediately after removal of the endotracheal
tube
c. Be available for any questions or concerns the
parents may have
3. Environment
a. Do not restrict the number of visitors
b. Provide privacy and comfort
(1) Low lights
(2) Decreased noise and activity
4. Religion
a. If ceremonies or rituals have not been performed
before now, offer any of the following:
(1) Clergy of choice
(2) Blessing, baptism, anointing, orprayer
b. Parental activity
(1) Offer the opportunity to hold the baby during
extubation
(2) Encourage the parents to notify the nurse if
they believe their baby is in pain and needs
assistance
Continued
444
PART VIII Care of the High-Risk Infant
BOX 35-1 Guideline for End-of-Life Care of the Neonate—cont’d
D. After death: once the infant has died, creating lasting
memories is integral to the healing of the family. Regardless of whether the family participates in their infant’s
death care, no steps should be eliminated. When an
infant dies, there are few, if any, memories of their time
together, and parents will cherish any memento of their
infant’s life (Jansen, 2003). It is the nurse’s responsibility
to help ensure that as many memories as possible are
created (Capitulo, 2005).
1. Communication
a. Offer the parents opportunities to be involved with
all aspects of after-death care.
b. If parents decline, respect their decision, but gently
remind them that this will be their only chance to
perform these tasks.
c. If parents decline, offer them the opportunity to
help the nurse with the infant’s care.
d. Reassure the family that the nurse is available at
any moment if assistance is required.
2. Environment
a. A room with a bed and chairs for family and friends
to be alone with their infant should be provided.
b. Complimentary snacks and beverages should be
made available.
c. Family time with the infant should be unrushed
and unlimited.
3. Parental activities
a. Encourage the parents to hold and dress their
infant.
b. Offer the parents the opportunity to bathe their
infant if this has not already been done.
c. Offer the parents the opportunity to have family
pictures taken at this time.
4. Memory makers: memory boxes should contain
items that have meaning and have been a part of the
infant’s hospitalization. Objects that seem trivial to
others will have profound significance for the family.
They will be the only concrete evidence to the parents and family that their infant was alive. The nurse
needs to be creative. There is no second chance to
create memories for the family (Catlin and Carter,
2002).
a. Identification band
b. Name card
c. Blood pressure cuff
d. Oxygen saturation probe
e. Thermometer
f. Respiratory paraphernalia
g. Measuring tape with infant’s length marked and
name and date written on it
h. Polaroid or digital photographs pictures
i. Stuffed animal
j. Pacifier
k. Diaper
l. Lock of hair
m. Handprints, footprints
n. Swaddling blanket (do not discard blanket if soiled)
o. Clothing (include postmortem gown, hat, socks,
T-shirt; do not wash if soiled)
p. Ring
q. Mold of hand, foot, or both
r. Cord clamp
s. Phototherapy mask
t. Dried washcloth and soap from bath, placed in a
small plastic bag to preserve soap fragrance
u. Black mourning (remembrance) pin
5. Final departure of parents: ensure that the parents do
not leave with “empty arms”
a. Memory box
b. Stuffed animal
c. Baby’s blanket
d. Pictures
e. Baby’s belongings
V. Nursing responsibilities
A. Prepare the infant’s body for transportation to the hospital
morgue per hospital protocol.
B. Complete the necessary paperwork.
C. Ensure that all tasks are completed per hospital policy.
Adapted from De Lisle-Porter M, Podruchny AM: The dying neonate: family-centered end-of-life care, Neonatal Netw 28:75-83, 2009.
NICU, Neonatal intensive care unit.
SUGGESTED READINGS
Allegaert K, Veyckemans F, Tibboel D: Clinical practice: analgesia in neonates,
Eur J Pediatr 168:765-770, 2009.
American Academy of Pediatrics Committees on Fetus and Newborn and on
Drugs, Sections on Anesthesiology and on Surgery; Canadian Paediatric
Society Fetus and Newborn Committee: Prevention and management of pain
and stress in the neonate, Pediatrics 105:454-461, 2000.
Anand KJ, Anderson BJ, Holford NH, et al: Morphine pharmacokinetics and
pharmacodynamics in preterm and term neonates: secondary results from the
NEOPAIN trial, Br J Anaesth 101:680-689, 2008.
Anand KJ, Hall RW, Desai N, et al: Effects of morphine analgesia in ventilated
preterm neonates: primary outcomes from the NEOPAIN randomised trial,
Lancet 363:1673-1682, 2004.
Carbajal R, Lenclen R, Jugie M, et al: Morphine does not provide adequate analgesia for acute procedural pain among preterm neonates, Pediatrics 115:14941500, 2005.
Carbajal R, Roussit A, Danan C, et al: Epidemiology and treatment of painful
procedures in neonates in intensive care units, J Am Med Assoc 300:60-70, 2008.
De Lisle-Porter M, Podruchny AM: The dying neonate; family-centered end-oflife care, Neonatal Netw 28:75-83, 2009.
Durrmeyer X, Vutskits L, Anand KJ, et al: Use of analgesic and sedative drugs in
the NICU: Integrating clinical trials and laboratory data, Pediatr Res 67:117127, 2010.
Fitzgerald M: The development of nociceptive circuits, Nature Rev Neurosci 6:507520, 2005.
Howard R, Carter B, Curry J, et al: Quick reference summary of recommendations
and good practice points, Pediatr Anesth 18(Suppl 1):4-13, 2008b.
Lago P, Garetti E, Merazzi D, et al: Guidelines for procedural pain in the newborn.
For the Pain Study Group of the Italian Society of Neonatology, Acta Paediatr
98:932-939, 2009.
Stevens BJ, Riddell RR, Oberlander TE, et al: Assessment of pain in neonates and
infants, In Anand KJ, Stevens BJ, McGrath PJ, editors: Pain in neonates and
infants, ed 3, Philadelphia, 2007, Elsevier, pp 67-90.
Complete references used in this text can be found online at www.expertconsult.com
P A R T
I X
Immunology and Infections
C H A P T E R
36
Immunology of the Fetus and Newborn
Calvin B. Williams, Eli M. Eisenstein, and F. Sessions Cole
Understanding the contribution of the newborn infant’s
immunologic response to neonatal disease requires a
review of the complex immunologic environment of pregnancy and the developmentally regulated changes in fetal
and neonatal immunity. The contrasting functions of the
fetal, neonatal, and maternal immunologic responses (i.e.,
preservation of fetal well-being as an allogenic graft versus
adequate immunologic protection in a nonsterile extrauterine environment) are regulated by a host of incompletely
understood developmental and genetic mechanisms. The
diversity and importance of these mechanisms are suggested by the heterogeneity and frequency of the infectious problems encountered in newborns. Differences in
immunologic responsiveness between adults and newborns should not be considered defects or abnormalities.
Just as the ductus arteriosus, a cardiopulmonary necessity
in the intrauterine environment, closes at different rates in
different infants, human fetal and newborn infant immunologic response mechanisms are developmentally and
genetically programmed to change from graft preservation
to identification and destruction of invading pathogens at
different rates.
MATERNAL AND PLACENTAL
IMMUNOLOGY
Although pregnancy is a natural process that is essential
for the propagation of all mammalian species, from an
immunologic perspective it poses a great dilemma. The
fetus represents a hemiallogenic graft that expresses
paternal antigens, which under ordinary circumstances
would be rejected as foreign tissue. The fact that such
rejection ordinarily does not occur cannot be explained
by systemic suppression of the maternal immune system.
Most studies of maternal immune function during pregnancy have not shown significant abnormalities (AagaardTillery et al, 2006) and are consistent with the clinical
observation that pregnant women are not at increased
risk for opportunistic infection. Furthermore, maternal
acceptance of the fetus does not occur exclusively as a
result of physical separation between immunogenic fetal
and maternal tissues. Trophoblastic tissue expressing
paternal antigens comes into direct contact with maternal
immune cells, and fetal cells can readily be detected in
maternal peripheral blood (Price et al, 1991). Conversely,
maternal leukocytes and somatic cells are present in significant numbers in the fetus and may persist into adult
life in a phenomenon known as microchimerism (Bianchi
et al, 1996; Lo et al, 1996; Maloney et al, 1999). As a
result, mechanisms must exist whereby antigen-specific
responses against paternal antigens either are not initiated
or are specifically suppressed. Beginning with the seminal
studies of Peter Medawar more than 50 years ago, significant progress has been made in unraveling the immunologic mechanisms underlying maternal-fetal tolerance
(Seavey and Mosmann, 2008; Trowsdale and Betz, 2006).
Currently it appears that no single factor can explain the
commensal immunologic relationship that exists between
mother and fetus throughout pregnancy. Rather, several
distinct but complementary innate and adaptive immune
mechanisms contribute to this process.
Before fertilization, both systemic and local immunologic changes occur in the endometrium to favor acceptance of the fetal allograft. For example, progesterone
induced by ovulation can reduce the proliferation of
T cells and inhibit antigen-presenting cells (Ehring et al,
1998; Miyaura and Iwata, 2002). In addition, as discussed
later, T cells with immunosuppressive properties accumulate cyclically in the endometrium during normal menses.
Semen has contrasting immunomodulatory effects on
the female reproductive tract and immune system, respectively; however, seminal plasma induces expression of proinflammatory cytokines, including granulocyte-monocyte
colony-stimulating factor (GM-CSF), interleukin (IL)
6, IL-8, and the chemokine monocyte chemotactic protein 1 by the uterine endometrium (Sharkey et al, 2007).
These molecules help to induce a local inflammatory cell
infiltrate containing macrophages, neutrophils, and dendritic cells, which may help to break down endometrial
mucin and thereby facilitate adherence of the blastocyst
to the endometrium (Dekel et al, 2010). GM-CSF also
has embryotrophic effects that promote fetal growth and
viability. Knockout mice lacking GM-CSF have normal
implantation rates, but have 15% to 25% smaller litters
(Robertson et al, 1999). However, semen contains immunosuppressive factors that teleologically are necessary to
inhibit initiation of an immunologic reaction against sperm
antigens. Therefore seminal plasma can induce secretion
of the immunosuppressive cytokine IL-10 by monocytic
cell lines (Denison et al, 1999) and is rich in transforming
growth factor (TGF) β-family cytokines (Tremellen et al,
1998). TGF-β1 is essential for inducing local differentiation of certain forms of immunoregulatory T cells (Chen
et al, 2003; Keskin et al, 2007).
445
446
PART IX Immunology and Infections
Inflammatory mediators and cytokines produced by
uterine tissues have a key role in embryonic implantation.
Prostacyclin is expressed by uterine tissues by day 5 of gestation and is required for blastocyst adherence and subsequent placental decidualization (Lim et al, 1999). The
IL-6 family of cytokines, which includes leukemia inhibitory factor (LIF), IL-11, and IL-6 itself, is also essential
for placental implantation. This family of cytokines is
defined by their receptors, which share a common signaling component, GP130, in addition to a cytokine-specific
alpha chain. LIF is expressed by uterine endometrium,
beginning during the mid-secretory phase of the menstrual cycle (Cullinan et al, 1996), probably in response to
progesterone (Eijkemans et al, 2005). Implantation does
not occur in LIF-deficient mice (Stewart, 1992). Interleukin-11, a cytokine that possesses hematopotic and antiinflammatory properties, has also been shown to be essential
for implantation, albeit at a later stage than LIF. Placenta
of mice deficient in the alpha chain of the IL-11 receptor fail to undergo decidualization, resulting in infertility
(Robb et al, 1998). IL-6 itself is expressed by endometrial
epithelial and stromal cells, mainly during the secretory
phase of menses (Perrier d’Hauterive et al, 2004). Reduced
expression of IL-6 mRNA has been noted in secretoryphase endometrium of women with recurrent miscarriages
(Jasper et al, 2007). This finding was attributed to a possible role of IL-6 in facilitating placental decidualization
and trophoblast development. In summary, an orchestrated series of specific proinflammatory and tolerogenic
mechanisms support implantation and early fetal allograft
survival.
STRATEGIES FOR IMMUNOLOGIC EVASION
AT THE MATERNAL-FETAL INTERFACE
One possible way in which the hemiallogeneic fetus might
avoid rejection is to evade detection by the maternal
immune system. This approach clearly is not the entire
explanation for tolerance of the fetal allograft. Indeed, fetal
antigenic maturity in eliciting a maternal immunologic
response has been well documented (Beer and Billingham,
1976; Billington, 1987). The view that maternal tolerance is
an active process is further supported by observations that,
at least in mice, maternal T lymphocytes specific for paternal antigens in particular are activated during pregnancy. In
addition, the fact that paternally derived tumor cells are not
rejected by pregnant female mice supports a central role for
antigen-specific systemic immune inhibition (Tefuri et al,
1995); therefore, tolerance appears to be at least in part a
metabolically active process. Nevertheless, several unique
features of the maternal-fetal interface undoubtedly contribute to attenuation of antifetal immunity (Table 36-1).
One such mechanism is altered expression of major
histocompatibility complex (MHC) proteins. MHC class
II molecules are necessary for CD4+ T cell responses,
which contribute both effector and regulatory functions
needed for graft rejection. MHC class II molecules are
not ordinarily constitutively expressed, except by certain antigen-presenting cells; however, proinflammatory
stimuli such as interferon (IFN)-γ are capable of inducing
MHC class II expression on most cell types. In contrast,
fetal trophoblastic tissues do not express MHC class II
TABLE 36-1 Selected Molecules of Immunologic Significance
at the Fetal-Maternal Interface
Molecule
MHC class II
Expression
Pattern
Functional
Consequence
Not expressed by
trophoblast, either
constitutively or
inducibly
Impaired CD4+ T cell
activiation
MHC Class I Molecules
HLA-A,
HLA-B
Not expressed by
trophoblast
Impaired CD8+ T cell
activation
HLA-C
Expressed by
trophoblast during
1st trimester
Modulation of NK cell
function
HLA-G
Expressed by
trophoblast
Inhibition of T cell and
dendritic cell function; modulation of
T cell and dendritic
cell function
Fas ligand
(CD178)
Expressed by placental
decidual and glandular epithelium and by
fetal trophoblast
Induces leukocyte
apoptosis
PDL-1
Expressed on placental
deciduas basalis
Immune inhibition
Cofactor for induction
of regulatory T cells
Galectins
Expressed by trophoblast early in
pregnancy
Induction of T cell
apoptosis
HLA, Histocompatibility leukocyte antigen; MHC, major histocompatibility complex;
NK, natural killer; PDL, programmed cell death ligand.
proteins, either in the resting state or in response to IFN-γ
(Peyman et al, 1992). MHC class II gene silencing in trophoblasts is at least partially the result of the transcriptional repression of class II transactivator, a necessary
transcription factor for class II gene expression (Murphy
and Tomasi, 1998; Peyman et al, 1992).
More importantly, human trophoblasts also do not
express conventional MHC class I human leukocyte antigen (HLA) A or HLA-B molecules. This fact undoubtedly contributes to reduced alloantigenic recognition at
the fetal-maternal interface. Human trophoblasts express
HLA-C, principally during the first trimester of pregnancy
(King et al, 1996) and two nonclassic HLA molecules—
HLA-E and HLA-G. The role of the latter in pregnancy
has been studied extensively. HLA-G is a nonpolymorphic
molecule encoded by a single gene that encodes seven
alternatively spliced transcripts and exists as a protein in
both membrane-bound and soluble forms. Its expression
is primarily limited to fetal tissue, but it can be detected
on some adult tissues, and its expression can be induced
on other cells in response to infection, inflammation, or
malignant transformation (Carosella et al, 2008). Unlike
classical MHC molecules, HLA-G does not have a significant role in stimulating CD8+ T cells via the T cell receptor complex. Rather, the principal function of HLA-G
molecules expressed by the trophoblast appears to be modulating the activity of natural killer (NK) cells. HLA-G
has also been shown to have other immunomodulatory
properties, including inhibition of cytotoxic T cell activity,
inhibition of alloproliferative responses by CD4+ T cells,
CHAPTER 36 Immunology of the Fetus and Newborn
and modulation of dendritic cell maturation and function
(Carosella et al, 2008). These data reveal that the unique
MHC class I molecule expression pattern on fetal trophoblast constitutes an intricate mechanism for orchestrating
the activity of immune cells.
Several additional interesting molecules expressed
at the maternal-fetal interface confer unique immunologic properties to this environment. Fas ligand (FasL)
is expressed in both maternal and fetal components of
the uteroplacental unit throughout gestation. Activated
T cells express the Fas receptor, which delivers an apoptotic (death) signal when bound by FasL. Implicit in
these observations is the hypothesis that expression of
FasL limits the reciprocal migration of activated fetal
and maternal T cells. This idea is strengthened further
by experiments examining the homozygous matings
between generalized lymphoproliferative disease (gld)
mice, which make a nonfunctional FasL. Pregnant generalized lymphoproliferative disease (gld) mice demonstrate
leukocyte infiltration and necrosis at the decidual-placental border, with many resorption sites and small litters (Hunt et al, 1997). Progesterone-induced blocking
factor is an immunomodulatory molecule released in
response to progesterone by trophoblastic cells (Anderle
et al, 2008). Its properties include indirectly suppressing NK cell function and inducing bias of CD4+ T cells
toward Th2-type cytokine secretion (Szekeres-Bartho
and Wegmann, 1996). The galectins represent a family
of molecules expressed at the maternal-fetal interface
with immunoregulatory properties. These glycoproteins
regulate the immune response by recognition of cell surface glycans on immune cells. Galectins are expressed in
human placenta primarily by the syntrophoblast early in
pregnancy (Than et al, 2009). Upon cell surface contact,
galectins downregulate the cellular immune response, in
part by inducing programmed cell death (apoptosis) of T
lymphocytes (Liu and Rabinovich, 2010).
A series of murine investigations points to a role for
local metabolic factors in suppressing maternal T cell
alloresponses in preventing spontaneous fetal loss. When
monocytes are induced to differentiate into macrophages
in vitro using macrophage colony-stimulating factor,
they become inhibitors of T cell proliferation rather than
activators. This inhibition was shown to be the result
of selective degradation of tryptophan by the inducible
enzyme indoleamine 2,3-dioxygenase (IDO). Serum
tryptophan levels fall during pregnancy, possibly in
response to IDO expression by syncytiotrophoblast cells,
which make the enzyme as early as 7.5 days after coitus.
Given the localization of IDO to the maternal-fetal interface and the immunosuppressive effects of tryptophan
depletion, it seemed plausible to investigators that this
pathway had a role in survival of the fetal allograft. This
hypothesis was supported by experime nts with either
normal or recombinase activating gene knockout mice,
which lack T and B cells, together with the IDO inhibitor
1-methyl tryptophan. Normal mice carrying allogenic
fetuses lose all their concepti by 11.5 days after coitus
when they are treated with the inhibitor. In contrast, the
delivery of healthy litters in RAG-deficient mice treated
with 1-methyl-tryptophan demonstrates an immunologic basis for the observation (Munn et al, 1998). More
447
recently, an additional tolerogenic mechanism influenced
by tryptophan metabolism has been suggested by studies of the indirect influence of the IDO pathway on differentiation and activation of regulatory T lymphocytes
(Baban et al, 2009; Chen et al, 2008). However, the lack
of fetal rejection observed in female, genetically deficient
IDO mice suggests that IDO may be necessary, but not
sufficient, in maintaining tolerance during pregnancy
(Baban et al, 2004).
Another possible mechanism through which the maternal immune system might be rendered relatively indifferent to the presence of fetal alloantigens concerns antigen
presentation within the uterine environment. Dendritic
cells (DCs) are a type of antigen-presenting cell that is critical for cellular and humoral immune responses. Collins
et al (2009) recently reported that the migration of DCs
from decidual tissue to draining uterine lymph nodes is
impaired compared with myometrial DCs. Therefore placental entrapment of DCs might represent a way by which
presentation of fetal antigens to the maternal immune system is retarded, thereby potentially inhibiting initiation of
allogenic immune responses.
NK cells found in the placental decidua are referred to
as decidual NK cells (dNK cells) to distinguish them from
NK cells found in maternal blood and those found in the
nonpregnant uterine endometrium (Yagel, 2009). These
cells comprise 70% of decidual lymphocytes, express a
CD56+CD16– phenotype (Manaster et al, 2008), and
have a unique gene expression profile which indicates that
dNK cells are a unique cell population (Koopman et al,
2003). Furthermore, despite expressing high amounts of
granzyme and other molecules necessary for mounting a
cytotoxic response, these cells exhibit reduced cytotoxicity compared with peripheral blood NK cells (Kopcow
et al, 2005) and were noncytotoxic to trophoblastic cells.
In contrast to NK cells found in peripheral blood and tissues that have important innate immune functions, including surveillance against infection and tumor cells, it is not
certain whether dNK cells have any role in immunity during pregnancy. Rather, they have been shown to have an
essential role in promoting trophoblast invasion into the
decidua and placental vascularization. These cells accomplish these tasks by secreting cytokines with effects on the
placental vasculature including IFN-γ, chemokines that
affect trophoblastic growth, and angiogenic factors including vascular endothelial growth factor, placental growth
factor, angiopoietin-2, and NKG5 (Hanna et al, 2006).
To facilitate activation of dNK cells to perform their
essential tissue remodeling functions, while at the same
time avoiding overactivation that could possibly lead to
cytotoxic damage to the trophoblast, their activity must be
tightly modulated by a series of interactions between several activating and inhibitory receptors and their respective ligands (Manaster and Mandelboim, 2010). The
nonclassic class I MHC molecules are among these dNK
cell receptor ligands. For example, a principal physiologic
ligand of β2-microglobulin–associated HLA-G is the
inhibitory receptor leukocyte immunoglobulin (Ig)-like
receptor 1 (Ponte et al, 1999). Free heavy chain HLA-G
also binds to this receptor in a noninhibitory fashion, and
this interaction could represent a feedback mechanism to
avoid overinhibition (Gonen-Gross, 2010).
448
PART IX Immunology and Infections
HLA-C is a major ligand for the different killer immunoglobulin-like receptors (KIRs), some of which are activating and others which are inhibitory. Amino acid allotypes
at position 80 of HLA-C confer preferential binding to
different forms of KIR, which in turn may result in clinically significant differences in dNK cell activation. Various combinations of maternal KIR and fetal HLA-C genes
have been shown to confer differential risk for preeclampsia, as a result of differences in dNK cell-mediated placental vascular changes (Hiby et al, 2004).
A final innate immune condition necessary for successful
completion of pregnancy is the absence of placental activation of complement. Fetal death occurs in mice that are
genetically deficient in the complement regulatory protein
Crry, accompanied by placental complement deposition
and inflammation (Xu et al, 2000). Although there is no
direct homolog of Crry in humans, complement inhibition
has been shown to be essential for normal pregnancy in a
murine model of the antiphospholipid syndrome, an autoimmune condition characterized by thrombosis, thrombocytopenia, and recurrent fetal loss. In this model, fetal
injury occurs as a result of placental inflammation initiated by local dysregulation of complement proteins. Both
complement activation and fetal loss can be prevented
administering anticoagulants with complement-inhibitory
properties such as heparin, but not by anticoagulants lacking complement-binding properties (Girardi et al, 2003,
2004). Some but not all human clinical interventional studies using anticoagulants with complement-binding properties to prevent fetal loss in the antiphospholipid syndrome
have shown positive results (Di Nisio et al, 2005).
CYTOKINE SECRETION BY T CELLS
DURING PREGNANCY
Bias in T cell cytokine secretion toward Th2-type cytokines and away from Th1-type cytokines has been
proposed as an immunologic condition necessary for
maintaining healthy pregnancy. Excessive exposure to
Th1-type cytokines is clearly detrimental to the fetus.
The Th1 cytokines IFN-γ and tumor necrosis factor-α
have been shown to inhibit the growth of trophoblasts and
to inhibit embryonic and fetal development (Haimovici
et al, 1991). Th1 cytokines terminate normal pregnancy
when injected into pregnant mice (Chaouat et al, 1990).
Th2-type cytokines contribute to implantation as discussed previously, and to maintenance of pregnancy as
suggested by LIF expression throughout pregnancy by
placental leukocytes, decidua, and chorionic villous cells
(Sharkey et al, 1999). LIF production by maternal T cells
from healthy pregnancies is also positively associated with
IL-4 production and inversely with IFN-γ (Piccinni et al,
1998). These studies have been interpreted to support the
notion that the balance between Th1 and Th2 cytokines
is important in maintaining pregnancy. However, it has
been indicated that mice lacking all Th2-type cytokines
(deleted by multiple genetic mutations) undergo normal
allogenic pregnancies with normal litter sizes (Fallon
et al, 2002). Based on this result, some investigators have
concluded that Th1/Th2 cytokine shifts is an epiphenomenon that is irrelevant to maternal-fetal tolerance (Trowsdale and Betz, 2006).
ROLE OF REGULATORY T CELLS
IN PREGNANCY
The mechanism whereby adaptive allogenic responses
against paternal antigens are actively suppressed during
pregnancy was largely unexplained until the discovery of a
distinct lineage of T lymphocytes with dominant immunosuppressive properties (Brunkow et al, 2001). This type of
regulatory T (Treg) cell is characterized by expression of a
lineage-specific transcription factor, Foxp3. Upon activation via their antigen-specific T cell receptor, Treg cells
are capable of suppressing immune effector cells, including dendritic cells and T effector cells through a variety
of mechanisms (Tang and Bluestone, 2008), thereby preventing a fatal form of autoimmune disease throughout life
(Kim et al, 2007). The ontogeny of Treg cells is discussed
more fully in the section on adaptive immunity.
The possible role of Treg cells during pregnancy was
first investigated in mice. These experiments disclosed
that Treg cell numbers are expanded in peripheral lymphoid tissues, and this expansion is not dependent on the
presence of alloantigen. Particularly high numbers of
Treg cells are found at the maternal-fetal interface, where
they comprise 30% of all CD4+ T cells. Treg cells are not
required for pregnancy in syngeneic mice, in which the
fetus does not display any foreign paternal antigens. In contrast, in allogenic pregnancy in which the fetus expresses
paternal antigens, Treg cell depletion resulted in pregnancy failure (Aluvihare et al, 2004). In another series of
experiments examining an abortion-prone strain of mice,
allogeneic pregnancy loss could be prevented by adoptive
transfer of Treg cells from healthy mice (Zenclussen et al,
2005). These data provide a plausible mechanism whereby
the adaptive immune system can actively suppress cellular
immune responses directed against paternal alloantigens.
Another line of evidence from studies performed in mice
provides additional indirect support for an essential role
of Treg cells in maintaining fetal tolerance in mice. Programmed death-1 (PD-1) is an inhibitory receptor expressed
on activated lymphocytes. There are two programmed death
ligands (PDLs) for the PD-1 receptor, PDL-1 and PDL-2.
PDL-1 is expressed on the decidua basalis, the portion of the
placenta abutting the fetal trophoblast, and by Treg cells.
Blockade of PDL-1 using antibodies in allogenic pregnancy
results in T cell–mediated fetal loss, indicating an essential
role for this molecule in preventing rejection of the fetal
allograft (Wang et al, 2008). This effect of anti–PDL-1 antibody is abrogated when Treg cells are depleted. Furthermore, in PDL-1–deficient mice, suppressive activity of Treg
cells was found to be impaired (Habicht et al, 2007). Finally,
an independent series of experiments has demonstrated an
important role for PD-1–PDL-1 interaction in Treg cells
(Krupnick et al, 2005). These data support a critical role for
PD-1–PDL-1 interactions in generating functional Treg cell
populations necessary for successful allogenic pregnancy.
Studies of Treg cells in humans further support an essential role for this cell type in initiating and maintaining
healthy pregnancy. Treg cells are increased in the peripheral
blood of women during the late follicular and luteal phases
of the menstrual cycle. This increase is believed to occur in
response to estradiol (Polanczyk et al, 2004; Tai et al, 2008).
Soluble factors in seminal fluid may contribute to further
CHAPTER 36 Immunology of the Fetus and Newborn
local expansion of these cells at the time of copulation
(Robertson et al, 2009). Human Treg cells selectively migrate
to the placental decidua under the influence of soluble factors including human chorionic gonadotropin (Schumacher
et al, 2009). Most but not all studies have shown that Treg
cell numbers are increased in peripheral blood throughout
pregnancy (Mjosberg et al, 2009; Somerset et al, 2004).
Alterations in Treg cells have been associated with certain pathologic conditions during pregnancy. Although
preeclampsia does not appear to be associated with reduced
numbers of Treg cells in peripheral blood, the ratio of
Treg cells relative to CD4+ T cells bearing proinflammatory phenotypic markers, such as Th17 cells, is abnormal
in preeclamptic women (Santner-Nanan et al, 2009). In
addition, toxoplasma infection in pregnant mice is associated with reduced numbers of Treg cells in peripheral
lymphoid tissues (Ge et al, 2008). This observation raises
the possibility that embryopathy associated with prenatal
infection is caused in part by the maternal immune system,
because of the breakdown of maternal-fetal tolerance as a
result of altered Treg cell differentiation.
A fascinating permutation of the role of Treg cells during
pregnancy concerns the effects of these cells on the developing fetal immune system. As already noted, fetal lymphoid
tissues are microchimeric—that is, they contain small but
significant numbers of maternally derived cells. Because not
all maternal antigens are inherited, it would be expected that
the fetal immune system would reject these cells. Recently it
was demonstrated that maternal cells expressing noninherited maternal antigens (NIMAs) are tolerated by the fetal
immune system as a result of active suppression by fetal Treg
cells (Mold et al, 2008). In this study, Treg cell numbers
were not found to be increased in the fetal thymus, but were
markedly increased in number in fetal lymphoid tissues. It is
therefore likely that these cells represent an induced rather
than a natural Treg cell population, which differentiates in
peripheral lymphoid tissues under the influence of TGF-β.
It was also shown in this study that small numbers of these
NIMA-specific Treg cells persist into childhood, but can be
reactivated using appropriate stimuli.
This property of Treg cells has important clinical implications in the transplantation setting. Bone marrow and solid
organ transplants are known to be more successful when
either the donor or recipient expresses NIMA (Burlingham,
2009; Dutta et al, 2009; van Rood et al, 2009). This finding
is explained by restimulation of dormant, NIMA-specific
Treg cells by transplanted cells expressing these antigens,
thereby contributing to active tolerance of the graft. In
addition to their potential relevance for developing future
clinical strategies for improving transplantation outcomes
while reducing the need for pharmacologic immune suppression (Burlingham, 2009), these findings also show that
some of what is learned by the fetal immune system in utero
is retained into childhood, and perhaps throughout life.
DEVELOPMENTAL FETAL-NEONATAL
IMMUNOLOGY
The newborn infant, especially the preterm infant, is at
increased risk for developing a considerable spectrum of
opportunistic infections, including Candida spp., herpes simplex, and cytomegalovirus. Developmental and
449
genetic differences between adults and infants in immunologic responsiveness account for much of this enhanced
susceptibility (Adkins et al, 2004; Levy, 2007), although
under certain circumstances neonates can mount adultlevel responses (Marchant et al, 2003; Sarzotti et al, 1996).
Research has focused on the molecular, cellular, and
functional definitions of these differences. This discussion focuses on those developmental aspects of innate and
adaptive immunity known to be important for fetal or neonatal responsiveness to infection.
INNATE IMMUNITY
During fetal adaptation from the sterile intrauterine environment to neonatal colonization of mucosal surfaces, the
innate immune system shields the newborn from infection while helping to orchestrate the acquisition of protective adaptive immune responses (Levy, 2007). These
innate mechanisms include protective barriers like the
vernix caseoa, which contains antimicrobial peptides and
fatty acids (Tollin et al, 2005), developmentally controlled
regulation of toll-like receptor signaling (Fusunyan et al,
2001; Lotz et al, 2006), expression of acute phase reactants
(Jokic et al, 2000; Levy 2007) and complement proteins,
and alterations in neutrophil and monocyte function (Forster-Waldl et al, 2005; Levy et al, 2004, 2006a, 2006b).
Importantly, functional maturation of innate immunity
allows for colonization with commensal organisms while
reducing potentially dangerous inflammatory responses.
Complement
The complement system, a principal component of the
innate immune response, consists of more than 40 plasma,
cell surface, and regulatory proteins that interact dynamically to regulate multiple physiologic functions, including resistance to pyogenic infections, interaction between
adaptive and innate immunity, and elimination of immune
complexes, products of inflammatory injury, and apoptotic self cells (Carroll, 2004; Walport, 2001a; Zipfel
and Skerka, 2009). Components of the complement system recognize and lyse bacteria, opsonize microorganisms nonspecifically, release anaphylatoxins, solubilize
immune complexes, and induce B‑cell proliferation and
differentiation.
Activation of the complement cascade can occur via
three pathways—classic, lectin, or alternative. The activation steps in these pathways have been reviewed recently
(Thiel, 2007; Zipfel and Skerka, 2009). Several characteristics of the complement cascade are important for
the fetal–neonatal immunologic response. First, whereas
the specificity of classic pathway activation results from
interaction of antigens with antibodies of several isotypes,
activation of the alternative and lectin pathways is antibody-independent and can be initiated by structures such
as endotoxin and polysaccharides frequently expressed by
pathogenic organisms. For the fetus or infant who has not
yet produced antigen‑specific IgG for immunologic recognition, the alternative and lectin pathways may be critical for triggering the effector functions of the complement
cascade (Kielgast et al, 2003; Simister, 2003; Stossel et al,
1973; Zilow et al, 1997). Second, the enzymatic activation
450
PART IX Immunology and Infections
of the complement cascade permits rapid amplification of
its functions: deposition of a single immunoglobulin molecule or C3b fragment can generate enzymatic cleavage
of thousands of later‑acting components and thus multiple
complement activities (Carroll, 2004; Walport, 2001a,
2001b; Zipfel and Skerka, 2009). In addition, the alternative pathway can be amplified via a positive feedback activation mechanism, because C3b, an activation product of
the alternative pathway C3 convertase, is a component of
this convertase (Janssen et al, 2006). Because of the importance of antibody‑independent recognition for the immunologic responsiveness of the fetus and infant, the positive
amplification loop of the alternative pathway is critical for
rapid generation of complement effector functions without specific immunologic recognition. Third, the continuous activation of the alternative pathway requires rigorous
regulation in the fetus to avoid tissue damage during organ
remodeling (Zipfel et al, 2007). Finally, the contributions
of the lectin pathway to fetal–neonatal complement activation and fetal well-being are still under investigation.
Complement activation via any of the three pathways
occurs in four main steps: initiation of complement activation, C3 convertase activation and amplification, C5
convertase activation, and terminal pathway activity or
the assembly of the membrane attack complex (Zipfel
and Skerka, 2009) (Figure 36-1). Although the classic and
lectin pathways may be triggered by recognition of ligands
via complement activating soluble pattern recognition
molecules (e.g., IgG or IgM antibodies, mannan-binding
lectin [MBL], or the ficolins), the alternative pathway
is spontaneously and constantly activated on biological
surfaces in plasma and in most or all other body fluids
(Pangburn and Muller-Ederhard, 1984). The spontaneous activation of the alternative pathway readily initiates
amplification and requires rigorous regulation (Zipfel et al,
2007). Complement activation results in assembly of the
first enzyme in the cascade, C3 convertase, via early‑acting
components of the classical (C1, C4, and C2), alternative
(factor B, factor D, and C3), or lectin (MBL, L-ficolin,
M-ficolin, H-ficolin, MBL-associated serine proteases
[MASP-1, -2, -3]), and a smaller MBL-associated protein
(MAp19) pathway. C3 convertase is assembled via highly
specific and limited proteolysis. Proteolytically activated
components form specific enzymatic complexes composed
of classical (C2b and C4b), alternative (C3bBb), or lectin (C2b and C4b) pathway components, which proteolytically cleave C3 into C3a and C3b (see Figure 36-1).
These two endopeptidases (C3bBb and C4bC2b) have
identical substrate specificities; each cleaves the single
peptide bond Argine77–Serine78 of the alpha chain of
C3 (Janssen et al, 2006). The rates of formation and dissociation of both C3 convertases are regulated by multiple
soluble (e.g., factor H, factor I, C4b‑binding protein) and
membrane‑associated proteins (e.g., membrane cofactor protein, delay accelerating factor) (Zipfel and Skerka,
2009). During the first phase of complement activation,
small (8 to 10 kD) peptides are released by proteolytic
cleavage from the second (C2a), third (C3a), and fourth
(C4a) components of complement. These fragments have
anaphylatoxin and antimicrobial activities and can establish chemotactic gradients for effector cells (Nordahl et al,
2004). If C3b is deposited close to the site of generation
and surface-bound convertases are formed, complement
cascade activation can be amplified (Gros et al, 2008). If
C3b fragments coat microbial or apoptotic surfaces, they
opsonize the particles for phagocytosis. Although C3b
deposition on the surface membrane of intact self cells can
be prevented by regulators that block further complement
activation, C3b deposition on microbial surfaces or on
modified self cells that lack such regulators amplifies complement activation (Zipfel and Skerka, 2009). The role of
non–inflammation-inducing, C3b-mediated removal of
Alternative
pathway
Classical
pathway
C3
C1
Initiation
C3
Lectin
pathway
MASP
C4 and C2
C3 convertase
C3bBb
C3a
C5
convertase
Inflammation
C5a
Terminal pathway
C4bC2b
C3b
C3bBbC3b
Opsonization
C4bC2bC3b
C5
C5b
C5b–C9
Terminal complement complex (lysis)
FIGURE 36-1 Summary of complement activation via the classic, alternative, and lectin pathways. (From Zipfel PF, Skerka C: Complement regulators
and inhibitory proteins, Nat Rev Immunol 9:729-740, 2009.)
CHAPTER 36 Immunology of the Fetus and Newborn
cells during fetal organ remodeling has not been explored
in detail.
If complement pathway activation continues beyond C3
cleavage, the binding of a second C3b molecule to either
C3 convertase generates the alternative (C3bBbC3b)
or classical or lectin (C4bC2bC3b) C5 convertases (see
Figure 36-1). Both enzymes proteolytically cleave C5 to
C5a and C5b. C5a is a powerful anaphylatoxin, and the
C5b fragment can initiate activation of the terminal pathway (Ward, 2009). The C5a-induced inflammation and
the C5b-initiated terminal pathway seem to be separately
regulated (Zipfel and Skerka, 2009).
Binding of C5b to either of the active C5 convertases
initiates directed, nonenzymatic assembly of the terminal
pathway components C5, C6, C7, C8, and C9 to form the
membrane attack complex (see Figure 36-1). The assembly and conformational changes of these soluble, hydrophilic proteins generate pores composed of lipophilic,
membrane-inserting complexes (the membrane attack
complex) that ultimately cause cell lysis by disrupting the
osmotic gradient between the intracellular and extracellular environments (Morgan, 1999).
Studies of fetal and neonatal complement have focused
on quantification of serum concentrations of individual
components, examining maternal–fetal transport of these
proteins, assessing specific effector functions of the classical
and alternative pathways, and investigating contributions
of complement activation to common neonatal diseases.
In humans, Gitlin and Biasucci (1969) reported detectable
concentrations of C3 (1% of adult levels) and C1 inhibitor
(20% of adult levels) by immunochemical methods as early
as 5 to 6 weeks’ gestation. By 26 to 28 weeks’ gestation,
both C3 and C1 inhibitor concentrations increased to 66%
of adult levels. Since these studies, multiple investigators
have demonstrated that functionally and immunochemically measured classical and alternative pathway protein
concentrations in cord blood increase with advancing gestational age and that they are only 50% to 75% of adult
concentrations at full‑term gestation (Sonntag et al, 1998;
Wolach et al, 1997). Although cord blood lectin pathway
component concentrations are lower than those in older
children and adults, the correlation between MBL and
gestational age has not been consistently observed (Hilgendorff et al, 2005; Kielgast et al, 2003; Kilpatrick et al,
1996; Lau et al, 1995; Swierzko et al, 2009; Thiel et al,
1995). The important roles of complement regulatory
proteins, decay‑accelerating factor, membrane cofactor
protein, and CD59 have also prompted examination of the
ontogeny of these proteins in the human fetus (Simpson
et al, 1993).
On the basis of studies of genetically determined, structurally distinct complement variants in maternal and cord
serum, no transplacental passage from mother to fetus
of C3, C4, factor B, or C6 has been observed (Colten
et al, 1981; Propp and Alper, 1968). The presence of
detectable amounts of C2 and C1 inhibitor in cord blood,
but not in the sera of mothers with genetic deficiencies
of these proteins, suggests that fetal–maternal transport of
these components does not occur.
Regulation of complement effector functions in the
fetus and newborn infant has not been as extensively examined. Opsonization of invading microorganisms without
451
specific immunoglobulin recognition requires activation
of the alternative or lectin pathways. For infants born
prematurely or without organism‑specific maternal IgG,
alternative or lectin pathway activation provides a critical mechanism for triggering complement effector functions (Maruvada et al, 2008; Super et al, 1989; Swierzko
et al, 2009). For example, Stossel et al (1973) demonstrated
opsonic deficiency in 6 of 40 cord sera examined because
of decreased factor B concentrations, despite normal C3
and IgG levels. The functional contribution of the classic
pathway to neonatal effector functions has been assessed
through the use of cord blood–mediated opsonophagocytosis by adult polymorphonuclear leukocytes of group B
streptococci type Ia (Edwards et al, 1983). This serotype
may be opsonized by classical pathway components in the
absence of specific antibodies and thus permits evaluation
of the function of classical pathway activation. In 8 of 20
neonatal sera examined, decreased bactericidal activity was
detected and correlated with significantly lower functional
activity of C1q and C4. These studies did not determine
whether this decrease was mediated by an inhibitor of
function or by an intrinsic change in functional activity
of these components in neonatal sera. The contribution
of the lectin pathway to immunosusceptibility of the newborn infant has been suggested by studies of both MBL
concentrations and pathway activity (Kielgast et al, 2003;
Kilpatrick et al, 1996; Sumiya et al, 1991; Super et al, 1989;
Swierzko et al, 2009; Thiel et al, 1995). Complement regulatory proteins (e.g., C4b-binding protein and factor H)
also contribute to neonatal immunosusceptibility as suggested by the failure of neonatal serum to stop invasion by
group B streptococci and Escherichia coli into human brain
microvascular endothelial cells (Maruvada et al, 2008).
The importance of the terminal complement component
C9 for cytolysis of multiple isolates of E. coli was suggested
by in vitro experiments in which killing of E. coli by neonatal serum samples was limited by C9, but not by other
classical pathway components (Lassiter et al, 1992, 1994).
Although lower serum concentrations of classical, alternative, and lectin pathway complement proteins can contribute to enhanced susceptibility of infants to systemic
infection, other complement functions important for fetal
and neonatal well‑being, but not related to antimicrobial
response, can contribute to reduced capacity to activate the
classical and alternative pathways. For example, reduced
serum concentration of C4b‑binding protein (8% to 35%
of pooled adult plasma levels), which is a critical regulator of classical pathway C3 convertase activity, has been
noted in fetal and neonatal sera (Fernandez et al, 1989;
Malm et al, 1988; Melissari et al, 1988; Moalic et al, 1988).
Lower C4b‑binding protein concentration increases the
functional anticoagulant activity of protein S, with which
it complexes and thereby contributes to decreased coagulation function of the fetus and newborn. Consideration
of functions besides immunologic effector functions may
be important in furthering the current understanding of
the developmental regulation of complement component
production.
The contribution of complement activation to tissue
injury has been investigated in several common neonatal
diseases, including neonatal hypoxic ischemic encephalopathy, necrotizing enterocolitis, meconium aspiration
452
PART IX Immunology and Infections
syndrome, and intrauterine growth restriction and fetal
loss (Girardi and Salmon, 2003; Girardi et al, 2006; Lassiter, 2004; Mollnes et al, 2008; Schlapbach et al, 2008;
Schultz et al, 2005). Concern has also been raised that
unregulated complement activation can occur in selected
infants who undergo extracorporeal membrane oxygenation therapy (Johnson, 1994; Kozik and Tweddell, 2006).
Complement activation is an important regulator of
multiple functions of the host immunologic response. Further study of the fetus and newborn infant will be aimed at
understanding the developmental and genetic regulation
of immunologic and nonimmunologic functions of this
important group of plasma and cell surface proteins.
Natural Killer Cells
NK cells are a component of the innate immune response
and represent approximately 10% to 15% of all peripheral
blood lymphocytes. NK cells are present in the spleen,
lungs, and liver, but rarely are found in lymph nodes and
thoracic duct lymph (Cerwenka and Lanier, 2001). Interestingly, NK cells are the major type of lymphocyte found
in the maternal decidual tissue, where they represent up
to 70% of all lymphocytes (King et al, 1996). NK cells are
distinguished from other lymphocytes by their morphology, function, and expression of distinct surface molecules.
Expression of the cell surface markers CD16 (FγRIII) and
CD56 (nerve cell adhesion molecule-1) can be used to
identify the NK population by analytical flow cytometry.
Mature NK cells appear larger and more granular than T
or B cells (Cooper et al, 2001) Mature NK cells are also
distinguished by the presence of both activating and inhibitory receptors that are used to selectively identify and kill
virally infected cells and tumors (Biassoni et al, 2001). NK
receptors recognize MHC class I molecules on target cells,
resulting in signals that suppress NK cell function. Target
cells that are deficient in or lack MHC class I activate NK
cell function, resulting in the release of lysosomal granules. These granules contain serine proteases, perforin, and
TGF-β, which disrupt the target cell membrane and induce
an inflammatory response. Studies show significantly lower
NK cell activity in fetuses and neonates compared with
adults (Georgeson et al, 2001; Kadowaki et al, 2001).
NK cells are derived from a common hematopoietic
progenitor that retains T and B cell developmental potential (Boos et al, 2008). NK cells first make their appearance in fetal liver as early as 6 weeks’ gestation. Committed
CD34+CD56– NK progenitors have been identified in the
fetal thymus, bone marrow, and liver. In humans, there
are several subsets of NK cells with distinct cell surface
markers and function. For example, the CD56high CD16–
subset is activated by IL-2 and also expresses CCR7 and
CD62L, which allows these cells to traffic through lymph
nodes (Di Santo, 2006). This subset is similar to NK cells
that develop in the mouse thymus through an IL-7- and
GATA3-dependent pathway and, in both species, is characterized by low cytotoxicity and enhanced cytokine production (Boos et al, 2008; Vosshenrich et al, 2006). In the
human neonate, the NK population is immature; only half
of all NK cells express CD56, and the NK cytolytic activity
is lower (Dominguez et al, 1993). This functional reduction
in NK activity has been proposed as a factor that contributes
to the severity of neonatal herpes simplex virus infections.
Profound defects in NK cell activity result in familial hemophagocytic lymphohistiocytosis, a disease characterized by
fever, hepatosplenomegaly, cytopenia, hyperferritinemia,
and hemophagocytosis. Familial hemophagocytic lymphohistiocytosis arises from mutations in genes that encode proteins involved in the granule-exocytosis pathway and is a fatal
disorder without bone marrow transplantation (Jordan and
Filipovich, 2008; Orange, 2006).
NK cell receptors are fundamentally different from
the T cell receptor (TCR) and B cell receptor (BCR).
NK receptor gene expression does not require gene segment rearrangement, and the receptors are not clonally
distributed. Instead, NK cells use an array of stimulatory
and inhibitory receptors to regulate their cytolytic functions (Lanier, 2008). A cluster of 10 or more genes encoding KIRs was located on human chromosome 19q13.4
(Biassoni et al, 2001; Lanier, 1998). Each of these type I
glycoproteins recognizes a different allelic group of HLAA–, HLA-B–, HLA-C–, or HLA-G– encoded proteins,
and each KIR is expressed by only a subset of NK cells.
Another family of immunoglobulin-like receptor genes
termed ILT is present near the KIR locus at 19q13.3.
These receptors are not as restricted as the KIRs and
bind multiple HLA class I molecules. A third inhibitory
receptor gene locus has been identified on chromosome
12p12-p13. These genes encode a C-type lectin inhibitory heterodimeric receptor called CD94/NKG2 that
binds HLA-E. Importantly, those KIRs, ILT receptors, and CD94/NGK2 molecules with long cytoplasmic
tails and two immunoreceptor tyrosine-based inhibitory
motifs (ITIMs) function as inhibitory receptors. Upon
phosphorylation, the two ITIMs recruit and activate
the Src homology domain 2 (SH2)-containing phosphatases, which turn off the kinase-driven activation cascade
(Ravetch and Lanier, 2000). The KIR family member
KIR2DL4 is distinct from other KIRs in structure and
distribution. KIR2DL4 binds HLA-G, has a single ITIM
in the cytoplasmic tail and a lysine in the transmembrane
region, which allows association with adaptor proteins.
This inhibitory receptor was found on all dNK cells in the
placenta at term, but not on circulating maternal NK cells.
This observation suggests that expression of KIR2DL4 is
induced during pregnancy (Rajagopalan and Long, 1999).
Other KIR or members of the C-type lectin superfamily
serve as activating receptors (Moretta et al, 2001). These
receptors lack the long cytoplasmic tail of the inhibitory
receptors and therefore do not contain ITIMs. Instead,
they have a charged amino acid in the transmembrane
region that allows the receptor to associate with the adaptor molecule DAP12 (Lanier et al, 1998). This adaptor
contains an immunoreceptor tyrosine-based activation
motif (ITAM) that allows these receptors to activate NK
cells. The physiologic role of these HLA class I–specific
activating receptors remains unknown. Another group
of NK cell–activating receptors has been identified and
termed natural cytotoxicity receptors (Moretta et al, 2001).
These proteins (NKp46, NKp30, NKp44) are immunoglobulin superfamily members with little similarity to one
another or to other NK receptors. They are highly specific for NK cells and appear to interact with non-HLA
molecules.
CHAPTER 36 Immunology of the Fetus and Newborn
453
CD244 (2B4) is a member of the signaling lymphocyte activation molecule family of receptors expressed on
all human NK cells (Ma, 2007). Upon interaction with
the ligand CD48 on target cells, NK signaling proceeds
via interactions between the ITSM (switch motif) in the
cytoplasmic tail of CD244 and one of two SH2 domaincontaining adaptor proteins, SAP and EAT-2. SAP interactions result in activation, as evidenced in humans with
X-linked lymphoproliferative disease, which is caused
by loss-of-function mutations in the SAP linker. In the
absence of SAP, interactions with EAT-2 may be inhibitory (Lanier, 2008).
infants and systemic neutropenia motivated several investigators to give neutrophil replacement therapy to neutropenic, infected infants (Christensen et al, 1980). Although
this approach has been successful in some cases, the results
have not been uniformly beneficial (Cairo, 1987; Cairo
et al, 1984; Menitove and Abrams, 1987; Stegagno et al,
1985). Similarly, the use of granulocyte CSF or GM-CSF
to treat infants with suspected infection has generally not
shown a reduction in mortality (Carr et al, 2003). This heterogeneity emphasizes the importance of individualizing
immunologic interventions for the developmental stage of
the infant and the invading microorganism being treated.
Polymorphonuclear Neutrophils
Monocytes and Macrophages
As observed for T and B lymphocytes, neonatal polymorphonuclear neutrophils (PMNs) are present at early stages
of gestation, but their functional capacities are different
from those of adult PMNs. Progenitor cells that are committed to maturation along granulocyte or macrophage
cell lineages (granulocyte-macrophage colony‑forming
units) are detectable in the human fetal liver between 6
and 12 weeks’ gestation in proportions comparable to
those observed in adult bone marrow (Christensen, 1989).
Human fetal blood has detectable granulocyte-macrophage colony‑forming units from 12 weeks’ gestation to
term (Christensen, 1989; Liang et al, 1988). Although
these progenitor cells are detectable in the fetus and newborn infant, developmental differences between adult and
mature neonatal PMNs have been demonstrated—in signal transduction, cell surface protein expression, cytoskeletal rigidity, rolling adhesion, microfilament contraction,
transmigration oxygen metabolism, intracellular antioxidant mechanisms, and extracellular trap formation (Carr,
2000; Henneke and Berner, 2006; Hill, 1987; Levy, 2007;
Ricevuti and Mazzone, 1987; Yost et al, 2009). The severity of functional differences correlates with the maturity of
the infant and begins to decrease within the first few weeks
after birth (Carr, 2000).
Besides intrinsic differences in PMN function, induction of specific functions and maturation of these cells
are developmentally regulated by the availability in the
microenvironment of specific inflammatory mediators and
growth factors (Christensen, 1989; Vercellotti et al, 1987).
For example, an activation product of the fifth component of complement, C5a, is a chemoattractant at sites of
inflammation. Low concentrations of C5 in neonatal sera
might not permit establishment of chemoattractant gradients at sites of inflammation in newborns comparable
to those in adults. Differences between adult and fetal–
neonatal PMN functions may thus reflect intrinsic cellular
differences required for fetal well‑being and differences in
the availability or activity of substances that regulate PMN
function.
The recognition that systemic bacterial infection in
newborns is frequently accompanied by profound neutropenia prompted the investigation of neutrophil kinetics
in infected infants (Christensen et al, 1980, 1982; Santos,
1980). These studies have suggested diverse, developmentally specific regulatory mechanisms required for mobilization of the neutrophil response to infection. The lack of
neutrophil precursors in bone marrow aspirates of infected
Cells committed to phagocyte maturation (granulocyte or
monocyte-macrophage) are detectable in the human fetal
liver by 6 weeks’ gestation and in peripheral fetal blood
by 15 weeks’ gestation. Unlike granulocytes, whose tissue
half‑life is hours to days, macrophages migrate into tissues and reside for weeks to months. In a tissue‑specific
fashion, these cells regulate availability of multiple factors,
including proteases, antiproteases, prostaglandins, growth
factors, reactive oxygen intermediates, and a considerable
repertoire of cytokines.
The importance of macrophages in the neonatal
response to infectious agents has been documented in
multiple studies. For example, increased antibody response
and protection from lethal doses of Listeria monocytogenes
were induced in newborn mice by administration of adult
macrophages (Lu et al, 1979). Functional differences in
chemotaxis, phagocytosis, and toll-like receptor signaling between adult and neonatal cells have been observed
and most likely result from both intrinsic fetal–neonatal
monocyte-macrophage characteristics, such as reduced
IL-12p70 production, and from nonmacrophage factors
(e.g., decreased production of the lymphokine IFN-γ)
(English et al, 1988; Kollmann et al, 2009; Levy, 2007;
Stiehm et al, 1984; van Tol et al, 1984). Inducible expression of individual complement proteins by lipopolysaccharide (LPS), a constituent of gram‑negative cell walls,
has also been shown to differ between adult and neonatal
monocyte–macrophages (Strunk et al, 1994; Sutton et al,
1986). This difference suggests that, although signal transduction mediated by LPS, LPS‑induced transcription, and
accumulation of mRNAs, which direct the synthesis of the
third component of complement and factor B, are comparable in adult and neonatal cells, a translational regulatory
mechanism does not permit these important inflammatory
proteins to be synthesized by LPS‑induced neonatal cells.
This observation emphasizes the fact that fetal–neonatal
monocytes–macrophages can have functions developmentally distinct from those of adult cells. For example, in
utero production of growth factors and removal of senescent cells during tissue remodeling may be critical to fetal
development (Kannourakis et al, 1988). Concurrent induction of these functions and immunologic effector functions
in fetal monocyte–macrophages would potentially elicit
nonspecific inflammation in actively remodeling tissues.
Besides having antibacterial functions, neonatal monocyte–macrophages contribute to tissue‑specific regulation of the microenvironment in individual organs. For
454
PART IX Immunology and Infections
example, considerable attention has focused on the contributions of these cells to antioxidant defenses and to
regulation of protease-antiprotease balance. Because
of the importance in tissue injury and repair, tissue and
injury‑specific treatment by appropriately targeted and
primed monocyte–macrophages may provide therapeutic
options for treating a spectrum of problems, from oxygen
toxicity in the lung to hemorrhage in the brain.
ADAPTIVE IMMUNITY
Lymphocytes play multiple critical roles in the adaptive
immune responses. Major lymphocyte lineages identified
by cell surface and functional criteria include T, B, and
NK cells (Kawamoto and Katsura, 2009). All three types
develop from CD34+CD38dim hematopoietic stem cells
found in the fetal liver and bone marrow (see Figure 36-1).
CD34 is also expressed on early committed progenitors,
which differentially express other markers usefully in lineage determination. Some early T cell progenitors retain
myeloid potential, suggesting a revision of the classic
model base on the presence of a common lymphoid progenitor (Bell and Bhandoola, 2008; Wada et al, 2008). The
process of lymphocyte differentiation is best viewed as a
progressive narrowing of differentiation potential based
on the sequential expression of specific transcriptional
regulators (Mansson et al, 2010).
T Lymphocytes
T lymphocytes or T cells develop in the thymus, which is
formed from the third branchial cleft and the third or fourth
brachial pouch. Thymic lobes are generated when tissue
from these sites moves caudally to fuse in the midline. Each
lobe can be divided into three regions based on structure
and function: the cortex, the corticomedullary junction,
and the medulla. The thymic cortex is composed of specialized epithelial cells that express MHC class I and class
II molecules and mediate the early stages of T cell maturation. This complex developmental process (Figure 36-2)
begins when multipotent CD1a–, CD5–, CD34+, and
Transcription
factors
Ikaros
Surface
receptors Notch-1
HSC
CD38low stem cells enter the thymus at the corticomedullary junction and migrate into the outer cortex (Awong et al,
2009; Res et al, 1996). Maturation continues as cells
migrate back through the cortex toward the corticomedullary junction. The first committed T cells express low
levels of CD4 and show DNA rearrangement at the TCR
δ gene locus. These events are followed by low levels of
CD8 expression and by rearrangement at the TCR-β
locus, which generates a pre-TCR on the cell surface
(Spits et al, 1998). The pre-TCR is composed of several
polypeptides, including a TCR-β chain and the invariant pre-Tα chain. Expression of the pre-TCR serves as
an important developmental checkpoint (beta selection;
von Boehmer and Fehling, 1997). Cells that do not successfully rearrange their TCR-β chain cannot express the
pre-TCR and die by apoptosis (Falk et al, 2001). The
remaining cells generate antigen-specific receptors when
the TCR-α chain replaces pre-Tα. These intermediate- to late-stage progenitors also express both the CD4
and CD8 coreceptors and are called double positives. The
minor population of δγT cells follows a similar developmental pattern.
Small double-positive cells (CD4+, CD8+, TCRlow)
constitute approximately three fourths of all thymocytes,
reflecting the critical and rate-limiting developmental
stage that follows. Further maturation requires that the
unique TCR on each thymocyte interact with self-peptide–MHC complexes expressed on the surface of thymic
epithelial cells. Several factors control the outcome of this
interaction, including the strength and timing of the TCR
signal generated and the nature of the antigen-presenting
cell (Hogquist et al, 2005). The TCR and associated CD3
signaling complex translate subtle differences in the affinity
of ligand binding into cell fate decisions through induced
conformational changes in the CD3 signaling complex
(Dave, 2009; Gil et al, 2005; Hayes, 2005), differential
utilization of mitogen-activated protein kinase signaling
pathways (Sohn et al, 2008; Sugawara et al, 1998), and
subcellular compartmentalization of mitogen-activated
protein kinase proteins (Daniels et al, 2006). Heterogeneity among thymic epithelial cells and the peptides that they
HES-1
GATA-3
Sox4, HEB, NFATc
E2A, STAT5
c-Kit, IL-7Rc
DN1
DN2
TCF-1-Lef-1, NF-B, p53
Frizzled-receptor,
Death-receptor, pre-TCR
DN3
DN4
TCR
DP
SP
CD4CD8
or
CD4CD8
-selection
Hematopoletic
stem cells
CD44CD25 CD44CD25 CD44CD25
Commitment
CD44CD25
CD4CD8
SP
Pre-TCR
TCR
checkpoint
checkpoint
FIGURE 36-2 Transcription factors and checkpoints in thymic selection. TCR, T cell receptor. (From Wu L, Strasser A: “Decisions, decisions”: betacatenin-mediated activation of TCF-1 and Lef-1 influences the fate of developing T cells, Nat Immunol 2:823-824, 2001.)
455
CHAPTER 36 Immunology of the Fetus and Newborn
present also contributes to the complexity of the selection
process (Klein et al, 2009).
In the affinity-based model of thymic selection (see
Figure 36-2), thymocytes that are unable to interact with
thymic cortical epithelial cells fail to generate a survival
signal and die by apoptosis, the default pathway for most
developing cells. Weak interactions generate survival
signals and continued development of positive selection.
Studies in mice show that the result of positive selection
is a thymocyte population containing a high frequency
of cells that are broadly cross-reactive for multiple MHC
alleles (alloreactivity) and for multiple peptides (Huseby
et al, 2005). For many positively selected cells, subsequent
high-avidity interactions with bone marrow–derived antigen-presenting cells in the thymic medulla result in activation-induced apoptosis or negative selection (Gong et al,
2001). Negative selection effectively culls from the positively selected pool of cells with cross-reactive TCR and is
the primary mechanism for the elimination of self-reactive
T cells (Huseby et al, 2003). For a small subset of thymocytes, high-affinity interactions with an agonist ligand
can also induce a transcription factor, Foxp3, that is associated with regulatory T cell development rather than cell
death (Jordan et al, 2001; Relland et al, 2009). Following
avidity-based selection, mature T cells that express either
the CD4 or CD8 coreceptor and a single TCR heterodimer are found in the thymic medulla just before the egress
into the peripheral circulation. In the human embryo, the
first naive, mature T cells appear at approximately 11 to
12 weeks of embryonic development. Thymopoiesis continues for many years thereafter, and integrity of this
process is essential for a healthy immune system. Thymectomy early in life results in a substantial loss of naive
T cells and in an oligoclonal memory T cell compartment
(Prelog et al, 2009; Sauce et al, 2009). The decay process
is accelerated by chronic CMV infection, resulting in an
immunosenescent T cell phenotype similar to that seen in
elderly individuals and associated with increased morbidity
and mortality (Wikby et al, 2006).
The final phase of T cell development is independent
of the thymus and involves peripheral (lymph nodes,
spleen- and gut-associated lymphoid tissue) homeostatic
mechanisms. These poorly understood events control
the expansion of clones recognizing specific antigens and
the development of T cell memory. Peripheral repertoire
selection begins with migration of lymphocytes from the
blood into lymphatic tissue. Naive T cells express the
adhesion molecule L-selectin (CD62L) that interacts with
peripheral-node addressins, a group of sialomucins present on high endothelial venules. Lymphocyte attachment,
coupled with the shear forces produced by blood flow,
results in rolling of the lymphocytes along the endothelial
cell surface. Signals mediated by CC-chemokine receptor
7 (CCR7) molecules on the surface of rolling lymphocytes
result in their tight binding to endothelial cells via integrins and in transendothelial migration (Forster et al, 2008).
After entering the lymph nodes, movement of lymphocytes is also controlled by chemokines and their receptors
(Worbs et al, 2007). CCR7 signaling directs cells to the
T cell areas, whereas CXCR5-mediated signaling controls
movement to B cell follicles (Reif et al, 2002). After activation, changes in chemokine receptor expression control
the mobilization and function of lymphocytes within the
lymph nodes.
T cell activation requires a complex molecular cascade
that results in reorganization of signaling molecules in the
membrane into an “immunological synapse” and in signal
transduction (Bromley et al, 2001). Many of the important
biochemical events in this process have been described.
TCR engagement by an appropriate MHC–peptide ligand
results in phosphorylation of components of the CD3
complex. The CD3 complex is composed of the αβTCR
and γ, δ, ε and ζ chains. These later molecules all contain
specific amino acid sequences called ITAMs, which serve
as molecular targets for the tyrosine kinases fyn and lck.
The ζ chain is thought to be the most critical component
and is found as a homodimer. Each ζ chain contains three
ITAMs, which will bind the tyrosine kinase Zap-70 when
sequentially phosphorylated. Appropriate phosphorylation
of ζ results in a downstream cascade that involves the tyrosine phosphorylation of multiple cellular substrates including phospholipase Cγ1, the guanine nucleotide exchange
factor Vav, and the adaptor protein Shc. Ultimately the
genetic program of the cells is altered, leading to the transcription of genes for cytokines, cytokine receptors, and
transcription factors (Cantrell, 2002).
After activation, CD4+ T cells differentiate into Th1,
Th2, or Th17 effector cells or become induced Treg cells
(Figure 36-3) (Murphy and Reiner, 2002; Zhu et al, 2010).
Each of these CD4+ T cell types is defined by prototypical transcription factors and by secretion of a characteristic
profile of cytokines in response to antigenic stimulation.
The cytokine milieu in the local environment during antigen presentation is a primary factor influencing the developmental fate of a naive T cell following activation. IL-12
promotes Th1 cell development by a signal transducer and
activator of transcription 4–dependent mechanism and
nTreg
Thymus
Foxp3
CD4
CD8
Foxp3
Naive T
IL-12
T-bet
Runx3
Th1
IFN-, IL-2
Autoimmune disease
Anti-tumor immunity
iTreg
IL-2, RA,
TGF-
Foxp3
IL-6, TGF- Th17
IL-4
RORt
Th2
GATA-3
IL-17
Autoimmune disease
Microbial immunity
IL-4, 5, 10, 13
Parasite infection
Allergy
FIGURE 36-3 Cytokines and transcription factors associated with
T cell differentiation. IFN, Interferon; IL, interleukin; iTreg, inducible
regulatory T cell; nTreg, natural regulatory T cell; TGF, transforming
growth factor. (From Sakaguchi S, Yamaguchi T, Nomura T, et al:
Regulatory T cells and immune tolerance, Cell 133:775-787, 2008.)
456
PART IX Immunology and Infections
results in expression of the transcription factor T-bet. Th1
cells secrete IL-2, IFN-γ, lymphotoxin alpha, and tumor
necrosis factor-α. Th1 responses are generally proinflammatory. Th1 cytokines act synergistically to lyse virally
infected cells and activate antigen-presenting cells as well
as granulocytes. In addition to being the signature cytokine of Th1 cells, IFN-γ also promotes the development of
the Th1 phenotype and inhibits the development of Th2
cells (Lighvani et al, 2001). Human neonatal T cells are
biased against Th1 polarization relative to adult T cells
(Levy, 2007). This bias has been linked to reduced IL12(p35) gene expression by dendritic cells (Goriely et al,
2001, 2004) and IL-4–dependent apoptosis of Th1 cells
after reexposure to antigen (Li et al, 2004). Poor Th1
function is associated with impaired killing of intracellular pathogens and a reduction in vaccine responsiveness
(Levy, 2007).
IL-4–mediated activation of the transcription factor
STAT6 is required for Th2 cell development (Kaplan
et al, 1996). STAT6 activation drives expression of another
transcription factor, GATA3, which promotes Th2 cell
differentiation. Th2 cells produce IL-4, IL-5, IL-10, and
IL-13. IL-4 induces immunoglobulin heavy-chain class
switching to the IgE isotype and promotes the development of the Th2 phenotype. IL-5 is an eosinophil growth
factor. Thus, Th2 cells are thought to promote antibody
production and the allergic response by multiple mechanisms (Ouyang et al, 2001). Human fetal T cells are biased
toward Th2 polarization (Prescott et al, 1998), and their
abundant representation in the neonate contributes to the
Th2 skewing of neonatal responses (Adkins et al, 2004).
The cytokines TGF-β and IL-6 activate STAT3, which
induces the expression of the transcription factor RORγt
and Th17 cell differentiation (Littman and Rudensky,
2010). Th17 cells, which are named for their secretion
of the cytokine IL-17A and related family members, are
proinflammatory cells associated with the clearance of
intracellular pathogens and protection from infection by
a number of bacterial and fungal species (Weaver et al,
2007). Abnormalities in Th17 cell development and function are also linked to immunodeficiency and autoimmunity. For example, humans with mutations in molecules
affecting Th17 cell differentiation have the hyper-IgE
syndrome (Al Khatib et al, 2009; de Beaucoudrey et al,
2008), and genes associated with Th17 cell differentiation
are associated with increased susceptibility to Crohn’s disease (Brand, 2009).
Activation of naive T cells requires a second signal, and
the most potent costimulatory molecule is CD28. The
ligands for CD28 are B7-1 and B7-2, which are present on
the surface of antigen-presenting cells. Cyotoxic T-lymphocyte antigen 4 (CTLA-4), a molecule expressed only
on activated T cells, also binds B7-1 and B7-2 and functions as a negative regulator of T cell activation. Experiments with knockout mice demonstrate that CD28 is the
major costimulatory receptor and that signaling through
CTLA-4 attenuates CD28-dependent responses (Alegre
and Thompson, 2001; Subudhi et al, 2005). A maturational delay in the development of costimulatory function
that contributes to reduced T cell responses in the neonatal period has been suggested (Orlikowsky et al, 2003),
and the expression of costimulatory molecules is further
suppressed by treatment with dexamethasone (Orlikowsky
et al, 2005).
Control of the clonal proliferation seen in a primary
immune response involves a number of inhibitory mechanisms, including Fas/FasL–mediated cell death, ligation
of the cell surface molecule PD-1, and regulatory T cell–
mediated suppression (Haribhai et al, 2007; Lettau et al,
2009; Riley, 2009; Sakaguchi et al, 2008). These mechanisms reset the peripheral immune system and thereby
maintain adequate clonal diversity. A small number of
activated T cells survives, and these cells differentiate into
memory cells (Wakim and Bevan, 2010). Memory cells
are defined by function (i.e., more rapid response) and by
expression of certain cell surface markers. The transcription factors T-bet and eomesodermin influence memory
development, probably by controlling expression of IL-7R
and IL-15R (Intlekofer et al, 2005; Pearce et al, 2003).
These receptors are linked to cell survival. Early memory
T cells are L-selectinlow, CD45ROhigh, and CD44high. Late
memory cells can become L-selectinhigh, which is a surface marker seen in naive T cells. The ultimate number of
memory cells has been shown in mice to reflect the initial
antigenic load and clonal burst size. Memory T cells allow
the secondary response to be more rapid and more potent,
characteristics that form the basis for vaccinations. The
memory phenotype is not seen early in life, but increases
with age. The precise mechanisms that generate and maintain memory T cells have not been determined, although it
is known that the persistence of CD8+ memory T cells in
mice does not depend on the persistence of specific antigen (Surh and Sprent, 2002; Williams and Brady, 2001).
In one study, preterm and term infants developed comparable memory T cell responses after vaccination (Klein
et al, 2010).
Immunocompetent T cells capable of responding to
foreign lymphocytes in the mixed lymphocyte reaction
are found in the fetal liver at 5 weeks’ gestation. Before
8 weeks’ gestation, lymphocytes are not detectable in the
fetal thymus. After 8 weeks, lymphoid follicles, T lymphocytes, and Hassall’s corpuscles can be identified. By 12 to
14 weeks’ gestation, T lymphocytes can be found in the
fetal spleen (Timens et al, 1987). By 15 to 20 weeks’ gestation, the fetus has readily detectable numbers of peripheral
T lymphocytes. The first T cell proliferative responses
that occur are to mitogens and can be measured at approximately 12 weeks’ gestation. By 15 to 20 weeks’ gestation,
antigen-specific responses can be detected (Adkins, 1999;
Garcia et al, 2001). Fetal growth has been shown to influence T, B, and NK lymphocyte counts by 3% to 6% per
week increase of gestational age (Duijts et al, 2009). Maturation of immune responses continues throughout the first
year of life, a fact that affects vaccination schedules (Gans
et al, 1998). A low level of thymopoiesis has been shown to
continue into adulthood (Kennedy et al, 2001).
Regulatory T Cells
Natural regulatory T cells develop as a distinct lineage
in the thymus dedicated to maintaining self tolerance
(Sakaguchi et al, 2008). Induced Treg cells arise in the
periphery from conventional CD4+ T cells activated in
the presence of TGF-β (Curotto de Lafaille and Lafaille,
CHAPTER 36 Immunology of the Fetus and Newborn
2009). Expression of the forkhead-winged helix transcription factor Foxp3 ultimately identifies these cell types
and is essential for the acquisition of suppressive effector function (Zheng and Rudensky, 2007). Treg cells are
required for the maintenance of immunologic tolerance,
as illustrated by the autoimmunity that arises after neonatal thymectomy and by the fatal autoimmune lymphoproliferative disease that develops shortly after birth in
mice and humans deficient in Foxp3 (Bennett et al, 2001;
Brunkow et al, 2001; Chatila et al, 2000; Sakaguchi et al,
1995; Wildin et al, 2001).
Treg cells have risen to the forefront of research in
immunology because of their essential role in maintaining
immunologic tolerance. Preclinical animal models have
demonstrated that adoptive transfer of Treg cells can prevent or cure diabetes, experimental allergic encephalomy
elitis (multiple sclerosis), inflammatory bowel disease, lupus,
arthritis, and graft-versus-host disease (Hori et al, 2002;
Kohm et al, 2002; Morgan et al, 2005; Mottet et al, 2003;
Scalapino et al, 2006; Tang et al, 2004; Tarbell et al, 2004;
Taylor et al, 2002). In humans, removal of Treg cells in
vitro enhances the proliferation of T cells in response to
self-antigens (Danke et al, 2004). Defects in Treg cell function and number have been described in a number of different human autoimmune diseases, including diabetes,
multiple sclerosis, rheumatoid arthritis, and juvenile idiopathic arthritis (Baecher-Allan and Hafler, 2006). Clinical
trials using the adoptive transfer of Treg cells after allogenic
hematopoietic stem cell transplantation as therapy for graftversus-host disease have begun in Germany and are planned
in the United States (Roncarolo and Battaglia, 2007).
B Lymphocytes
B cells are lymphocytes that upon activation give rise
to terminally differentiated, immunoglobulin-secreting
plasma cells. Immunoglobulins form the humoral arm of
the immune system and provide the main form of protection against many pathogens. B cell development can be
divided into embryonic and adult phases. The embryonic
phase begins in the fetal liver at the same gestational age as
T cell development, follows a similar time course, and uses
similar developmental strategies. The adult phase occurs
in the bone marrow and continues throughout life. In both
phases, B cell progenitors are selected at key developmental checkpoints for the presence of a functional BCR
rearrangement and for the absence of BCR self-reactivity.
Those progenitors with self-reactive receptors are either
eliminated or generate new antigen receptors by continued
gene segment rearrangements that are not self-reactive.
Both B cell development and peripheral B cell homeostasis
require signal transduction through the BCR. Although
the B cell compartment is well formed before birth, diversification of the antibody repertoire and several important
antibody responses are not developed until long after the
neonatal period (Hardy and Hayakawa, 2001; Rohrer et al,
2000; Rolink et al, 2001; Yankee and Clark, 2000).
B cell development begins before 7 weeks’ gestation in the human fetal liver, and pre-B cells in various
stages of maturation are seen by 8 weeks’ gestation. By 8
to 10 weeks’ gestation, CD34+ hematopoietic stem cells
are also found in the bone marrow. The early phase of
457
B cell development, like T cell development, is antigen
independent. The first committed progenitors (pro/preB-I) express CD34, terminal deoxynucleotidyl transferase (TdT), recombinase-activating genes (RAG) and
CD19 (Li et al, 1993). They lack immunoglobulin gene
rearrangement (Allman et al, 1999). The next lineage is
marked by heavy chain (H) diversity to joining (DH to
JH) gene segment rearrangements followed by variable
to diversity/joining segment rearrangement. Terminal
deoxynucleotidyl transferase is used during this process
to insert nucleotides between the segments to create
additional diversity at a region that encodes the VH portion of the antibody-binding site. Late cells in this stage
express an invariant surrogate light chain (Kitamura et al,
1992). The normal light chain genes remain in germline
configuration.
The first developmental checkpoint in B cell development requires expression of surrogate light chain together
with Ig-α and Ig-β on the cell surface as the pre-BCR,
marking the transition to the pre–BII stage. Expression of
the pre-BCR is essential for positive selection of cells that
have a functional μH chain rearrangement and for expansion of the pre–B cell lineage (Gong and Nussenzweig,
1996). Cells with a nonfunctional H chain rearrangement
or with H chains that assemble poorly with surrogate light
chain are unable to progress. VL to JL rearrangement takes
place next, and the newly expressed polymorphic L chain
protein replaces the surrogate light chain. The completed
BCR is antigen-specific and contains the mIgM molecule.
A second developmental checkpoint occurs here, and those
B cells with self-reactive receptors are eliminated or edited
(Hartley et al, 1991). Only 10% to 20% of the immature
B cells survive this negative selection and migrate to the
spleen, which they enter through the terminal branches of
the central arterioles. Once in the spleen, they rapidly differentiate into mIgM+, mIgD+, B220+ mature B cells that
enter the recirculating pool of B lymphocytes (Hardy and
Hayakawa, 2001).
When mature B cells contact antigen through the
BCR, a signal is transduced that promotes further growth
and differentiation into surface Ig+ memory cells and
plasma cells (Calame, 2001). BCR signaling involves
activation of the tyrosine kinases Syk and Btk, which are
also involved in pre-BCR signal transduction. Certain
mutations in Btk result in a failure of pre-BCR signaling and lead to X-linked agammaglobulinemia. Many B
cell responses use the ability of B cells to capture antigen at vanishingly low concentrations, process the antigen into small peptide fragments, and then present the
antigen to Th cells in the context of MHC class II molecules. Antigen-specific T cells then form conjugates
with antigen-presenting B cells. The T cell membrane
protein gp39 interacts with CD40 on B cells and triggers
B clonal expansion, cytokine responsiveness, and isotype
switching.
B lymphocytes with surface IgM are first found in the
human fetal liver at 9 weeks’ gestation and in the fetal
spleen at 11 weeks’ gestation (Owen et al, 1977; Timens
et al, 1987). Antigen‑specific antibody production can be
detected in the human fetus by 20 weeks’ gestation. Fetal
spleen cells can synthesize in vitro IgM and IgG by 11 and
13 weeks’ gestation, respectively.
458
PART IX Immunology and Infections
TABLE 36-2 Characteristics and Functions of Immunoglobulins
Molecular Weight (kD)
Serum
Concentration (g/L)
Serum Half-life (Days)
Neutralization
Opsonization
IgG1
150
10
21
++
+++
IgG2
150
5
21
++
(+)
IgG3
170
1
7
++
++
IgG4
150
0.5
21
++
+
IgA1
160
3
7
++
+
IgA2
160
0.5
7
++
+
IgM
900
2
5
+
+
IgD
180
0.03
3
–
–
IgE
190
0.00003
3
–
–
Adapted from Mix E, Goertsches R, Zett UKL: Immunoglobulins: basic considerations, J Neurol 253:V9-V17, 2006.
Ig, Immunoglobulin; NK, natural killer.
Immunoglobulins
Immunoglobulin G
Immunoglobulins are a heterogeneous group of proteins
that are detectable in plasma and body fluids and on the
surfaces of mucosal barriers and B lymphocytes. Although
these proteins have multiple, diverse functions, they are
classified as a family of proteins because of their capacity
to act as antibodies—that is, to recognize and bind specifically to antigens. The rapid advances in understanding
molecular structure and regulation, genetic diversity, and
differences in functions of immunoglobulins have been
reviewed recently (Bengten et al, 2000; Mix et al, 2006).
The functions of immunoglobulins relevant to fetal and
neonatal immunity are summarized in Table 36-2.
There are five known classes of immunoglobulins:
IgG, IgM, IgA, IgE, and IgD. Human IgM circulates as
a pentamer or hexamer and IgA as a dimer. Multimers are
formed in association with an additional J-chain (or joinchain). Functions of individual immunoglobulin classes
are different but overlapping (see Table 36-2) (Mix et al,
2006). The prototype immunoglobulin molecule consists
of a pair of identical heavy (H) chains that determine the
immunoglobulin class in combination with a pair of identical light (L) chains (Figure 36-4). Disulfide bonds and
electrostatic forces link the chains. Each immunoglobulin
molecule contains two N-terminal, identical domains with
antigen‑binding activity (Fab). The Fab fragment consists
of variable (VH and VL) and constant (CH and CL) regions
on both the H and L chains. The Fab domains function
to bind antigenic epitopes via complementarity determining regions. A third fragment (i.e., F crystalline [Fc]) is
devoid of antibody activity, but mediates immunoglobulin
effector functions (Table 36-3). The principal functions of
the Fc fragment include: receptor-mediated phagocytosis
(IgG1/3 and IgA), cytotoxicity (IgG1/3), release of inflammatory mediators (IgE), receptor-mediated transport
through mucosa (IgA and IgM) and placenta (IgG1/3),
and complement activation (IgG1/3 and IgM). The five
different isotype classes of human immunoglobulins (IgG,
IgM, IgA, IgD, and IgE) are defined structurally by differences in the Fc fragments. Within isotypes, there are four
IgG subclasses (IgG1, IgG2, IgG3, and IgG4) and two IgA
subclasses (IgA1 and IgA2).
IgG is the most abundant immunoglobulin class in human
serum and accounts for more than 75% of all antibody
activity in this compartment. Its monomeric form circulates in plasma, has a molecular mass of approximately
155 kD, and in adults constitutes approximately 45% of
total body IgG in the extravascular compartment. The
human conceptus is able to produce IgG by 11 weeks’
gestation (Gitlin and Biasucci, 1969; Martensson and
Fudenberg, 1965). The importance of the contribution of
IgG to immunologic function is illustrated by the clinical
problems encountered in individuals with genetically disrupted IgG production (hypogammaglobulinemia). These
patients have recurrent infections if they do not receive
treatment with immunoglobulin replacement therapy
(Yong et al, 2008).
Several investigators have observed that infants in whom
group B streptococcal sepsis develops have low concentrations of type‑specific IgG, which has prompted attempts
at acute or prophylactic treatment with immunoglobulin
replacement therapy (Stiehm et al, 1987). Although successful in some trials, replacement therapy in newborns has
not proved as efficacious as in individuals with genetically
determined hypogammaglobulinemia (Cohen-Wolkowiez
et al, 2009; Noya and Baker, 1989). This difference might
be caused in part by fetal and neonatal IgG synthesis being
regulated by both developmental and genetic mechanisms
(Cates et al, 1988; Lewis et al, 2006).
The kinetics of IgG placental transport suggest both
passive and active transport mechanisms (Saji et al, 1999;
Simister, 2003). Because IgG transport begins at approximately 20 weeks’ gestation, preterm infants are born
with lower IgG concentrations than are their mothers or
infants born at term. The full‑term infant has a complete
repertoire of adult, maternal IgG antibodies. Thus, provided that relevant maternal IgG has been transported to
the fetus, newborns are generally immune to many viral
and bacterial infections (e.g., measles, rubella, varicella,
group B streptococcus, and E. coli) until transplacentally
acquired antibody titers decrease to biologically nonprotective concentrations at 3 to 6 months of age (Pentsuk and
van der Laan, 2009). The regulation of IgG production in
CHAPTER 36 Immunology of the Fetus and Newborn
Epithelial
Transport
Complement Activation
Placental
Transport
Sensitization for
Killing by NK Cells
Sensitization of Mast
Cells and Basophils
Strong classic, alternative
–
+++
++
+
Classical, alternative
–
+
–
–
Strong classical, alternative
–
++
++
+
Alternative
–
(+)
–
–
Alternative
+++
–
–
–
Alternative
+++
–
–
–
Strong classical
+
–
–
–
Alternative
–
–
–
–
–
–
–
–
+++
Idiotopes
H
L
V
re ari
gi ab
on le
V
s
Idiotopes
s
s
L
s
s
Constant
region CH 3
s
s
s
s
Constant
region CH 4
Carbohydrates
Hinge
s s
s s
s
s
s
s
s
s
Constant
region CH 2
s
s
s
Constant
region CH 2
s
s
s
t
an
st C L
on n
C gio
re
s
s
V
re ari
gi ab
on le
V
s
Constant
region CH 3
s
s
C
re on
gi sta
on n
C t
s
t
an 1
st H
on C
C ion
g
re
Light chain
Heavy chain
re Con
gi s
on ta
C nt
H
1
e
bl L
ria n V
Va gio
re
s
s
Paratope with 3
complementaritydetermining
regions (CDRs)
s
s
Carbohydrates
Constant
region CH 4
s
e
bl H
ria n V
Va gio
re
N-terminal
459
C-terminal
FIGURE 36-4 Structure of monomeric immunoglobulin. (From Mix E, Goertsches R, Zett UK: Immunoglobulins: basic considerations, J Neurol
253(Suppl 5):V9-V17, 2006.)
preterm infants has been a topic of study for more than
60 years (Ballow et al, 1986; Bauer et al, 2002; Bonhoeffer
et al, 2006; Dancis et al, 1953; Meffre and Salmon, 2007;
Schroeder et al, 1995). Although adults with antibody deficiency syndromes have a higher rate of infections when IgG
concentrations are less than 300 mg/dL, the serum IgG
concentrations of many preterm infants decrease to less
than 100 mg/dL apparently without consequences. These
observations suggest that preterm infants have additional
immunologic protective mechanisms or that the protective
capacity of humoral immunity is not accurately assessed by
serum IgG concentrations alone in preterm infants. Studies also suggest that reduced endogenous secretion of IgG
by newborns may result from reduced ability of neonatal
B cells to undergo immunoglobulin isotype switching,
because of decreased or ineffective expression of the ligand
for the B cell surface protein CD40 on activated cord
blood T cells (Brugnoni et al, 1994; Fuleihan et al, 1994).
460
PART IX Immunology and Infections
TABLE 36-3 Severe Combined Immunodeficiency Classification
Mechanisms
Mutated
Genes
Premature cell death
ADA
AR
T, B, NK
Defective cytokinedependent survival
signaling
γc
X-L
T, NK
Defective VDJ
rearrangement
Defective pre-TCR
and TCR signaling
Inheritance
Affected
Cells
multiple effector functions of this cascade. Second, its pentameric structure provides conformational flexibility to
accommodate multivalent ligand binding. Third, because
of its localization in the vascular compartment and its high
efficiency in complement activation, IgM has a prominent
role in clearance from serum of invading microorganisms.
Immunoglobulin A
JAK3
AR
T, NK
IL7RA
AR
T
RAG1 or
RAG2
AR
T, B
Artemis
AR
T, B
CD3 δ,
ξ, ε
AR
T
CD45
AR
T
Adapted from Cavazzana-Calvo et al: Gene therapy for severe combined
immunodeficiency: are we there yet? J Clin Invest 117:1456-65, 2007.
AR, Autosomal recessive; NK, natural killer; TCR, T cell receptor; X-L, X-linked.
IgG functions in host defenses in several ways. It can
neutralize a variety of toxins in plasma by direct binding.
As described previously in the section on complement,
after antigen binding IgG can activate the complement
cascade via interaction with early‑acting, classical pathway complement components. The Fc portion of IgG can
interact with cell surface receptors on mononuclear phagocytes and polymorphonuclear leukocytes and thereby
promote clearance of immune complexes and phagocytosis of particles or microorganisms—a process known as
opsonization. Finally, the presence of IgG on specific target
cell antigens (e.g., tumors or allogeneic transplant tissues)
can mediate antibody‑dependent cellular cytotoxicity, a
mechanism through which lymphocyte subpopulations
bearing Fc receptors can acquire the capacity to recognize
non–self-antigens.
Immunoglobulin M
IgM represents approximately 15% of normal adult
immunoglobulin. IgM circulates in serum as a pentamer
of disulfide‑linked immunoglobulin molecules joined by
a single cross‑linking peptide (J-chain). The size of IgM
(molecular mass >900 kD) restricts its distribution to the
vascular compartment. Although the antibody‑binding
affinity of monomeric IgM is low, the multivalent structure of the molecule provides high pentameric antibody
avidity. IgM synthesis has been detected in the human conceptus at 10.5 weeks’ gestation (Rosen and Janeway, 1964).
Because maternal‑fetal transport of IgM does not occur,
elevated (>20 mg/dL) concentrations of IgM in the fetus
or newborn may be suggestive of intrauterine infection or
immunologic stimulation (Alford et al, 1969; Stiehm et al,
1966). However, because it is technically difficult to distinguish IgM molecules with specificity for individual organisms, diagnosis of infections by analysis for specific IgM
antibody remains of limited usefulness.
IgM is important for fetal and neonatal host defenses for
several reasons. First, the IgM molecule is the most efficient of any immunoglobulin isotype in activation of the
classical pathway of complement. As a result, it can trigger
Although IgA accounts for approximately 10% of serum
immunoglobulins, it is detectable in abundance in all external secretions (Brandtzaeg, 2010). In serum, IgA is present as a monomer (molecular weight, 160 kD), whereas
in secretions it exists as a dimer (molecular weight, 500
kD) attached to a J chain identical to that found in IgM.
In addition to this structural difference, mucosal IgA is
attached to an additional protein called the secretory component. This protein is a proteolytic cleavage fragment of
the receptor involved in the secretion of polymeric IgA
onto mucosal surfaces and into bile. Secretory IgA produced locally on mucosal surfaces by plasma cells is thus
readily distinguishable from serum IgA. Although the
amount of IgA produced daily is not rigorously quantified,
it is estimated to exceed immunoglobulin production of all
isotypes combined. Despite its relative abundance, unlike
IgM and IgG, IgA cannot activate the classical pathway
of complement nor effectively opsonize for phagocytosis
particles or microorganisms.
Although IgA is detectable on the surface of human fetal
B cells at 12 weeks’ gestation, adult concentrations of serum
and secretory IgA are not achieved until approximately
10 years of age. Because serum IgA is not transplacentally
transferred in significant amounts, IgA is almost undetectable in cord blood. Colostrum‑derived secretory IgA
can provide a source of IgA in both gastrointestinal tract
and other secretions for the newborn infant (Brandtzaeg,
2010). Unlike other immunoglobulin isotypes, amino acid
sequences of the hinge region of the IgA‑2 subclass confer
partial resistance to bacterial proteases. IgA is thus more
resistant than other immunoglobulin isotypes to proteolytic effects of gastric acidity. Although considerable
investigation suggests that passive immunization with IgA
occurs with breastfeeding in humans, the overall importance of IgA in host defenses is currently not well characterized (Bailey et al, 2005).
Immunoglobulin E
The serum concentration of IgE is undetectable by
standard immunochemical techniques and accounts for
approximately 1/10,000 of the immunoglobulin in adult
serum. It circulates in the monomeric form (molecular
weight, 190 kD). Structurally, IgE lacks a hinge region. It
is produced by most lymphoid tissues in the body, but in
greatest amounts in the lung and gastrointestinal tract. It
is not secreted, and its appearance in body fluids generally
occurs only with induction of inflammation. IgE cannot
activate complement or act as an effective opsonin. Its bestknown role is as a mediator of immediate hypersensitivity
reactions. Specifically, antigen‑specific IgE triggers mast
cell degranulation with resultant bronchoconstriction, tissue edema, and urticaria via interactions with IgE receptors
on the mast cell surface. Because of the presence of IgE in
lung secretions and its potential importance in mediating
CHAPTER 36 Immunology of the Fetus and Newborn
allergic pulmonary and gastrointestinal reactions, considerable interest has been recently focused on the use of
serum IgE concentrations to identify premature infants at
risk for developing reactive airways disease or in the diagnosis of gastrointestinal hypersensitivity reactions (Bazaral
et al, 1971; Herz, 2008; Jarrett, 1984; Kagan, 2003).
Immunoglobulin D
Although IgD is found in trace quantities in adult human
serum and has neither complement‑activating activity
nor the capacity to opsonize particles or microorganisms,
approximately 50% of cord blood lymphocytes exhibit IgD
on their cell surface (Colten and Gitlin, 1995; Haraldsson et al, 2000; Preud’homme et al, 2000). These pre–B
lymphocytes express surface IgM and IgD simultaneously.
Because of its wide distribution on B cells, IgD may have
an important role in primary antigen recognition for the
fetus and newborn infant.
Immunoglobulin Therapy
Because of low serum immunoglobulin concentrations, a
concurrent greater susceptibility to infection in preterm
infants, and successful use of immunoglobulin replacement in patients with genetically based hypogammaglobulinemia, several investigators have proposed prophylactic,
intravenous administration of immunoglobulin (IVIG)
to prevent antibody deficiency and nosocomial infection
(Baker et al, 1992; Clapp et al, 1989; Cohen-Wolkowiez
et al, 2009; Fanaroff et al, 1994; Kinney et al, 1991; Lewis
et al, 2006; Magny et al, 1991; Weisman et al, 1994). Immunoglobulin therapy in very low-birthweight neutropenic
infants with bacterial sepsis and shock has been tried with
some success, when the potential benefits might outweigh
the immunomodulatory properties of IVIG (Christensen
et al, 2006). However, well-designed, placebo-controlled
studies have failed to show consistent benefit of this strategy using different preparations of immunoglobulin, different treatment groups, and different dosage regimens
(Ohlsson and Lacy, 2004a). These results suggest that
serum immunoglobulin concentrations do not predict
immunoglobulin function and immunologic response as
accurately in the newborn infant as in the older child or
adult.
In infants with sepsis, the results of immunoglobulin
therapy have been difficult to interpret because of the complexities in study design associated with enrolling acutely ill
infants and the small numbers of enrolled infants (Ohlsson
and Lacy, 2004b). A recent metaanalysis of randomized,
controlled studies that examined the effects of polyvalent
immunoglobulin for treatment of sepsis or septic shock in
newborn infants (12 trials, 710 newborn infants) suggested
a reduction in mortality (Kreymann et al, 2007). However,
heterogeneous study quality led the Cochrane reviews to
suggest that there is still insufficient evidence to recommend
routine use of this strategy. The availability of results from
the large cohort recruited for the International Neonatal
Immunotherapy Study (www.npeu.ox.ac.uk/inis) should
provide a more definitive recommendation (INIS Study
Collaborative Group, 2008). Other interventions including
administration of colony-stimulating factors, antistaphylococcal antibodies, probiotics, glutamine supplementation,
461
recombinant human protein C, and lactoferrin have been
reviewed recently (Cohen-Wolkowiez et al, 2009).
Besides prophylactic or acute treatment of systemic bacterial infection, the therapeutic scope of monoclonal and
polyclonal immunoglobulin therapy has expanded over the
last 10 years. For example, although controversy still exists
concerning specific populations of infants for whom prophylaxis is appropriate, administration of anti–respiratory
syncytial virus monoclonal antibody has become standard of care for prevention of respiratory syncytial virus
(RSV) infection in high-risk infants and for management
of nosocomial outbreaks of RSV (Fitzgerald, 2009; Halasa
et al, 2005; Kurz et al, 2008; Wu et al, 2004). In addition,
administration of intravenous IgG is a safe and effective
immunomodulatory strategy for reducing the need for
exchange transfusion in hemolytic disease of the newborn
if the total serum bilirubin is rising despite intensive phototherapy (American Academy of Pediatrics Subcommittee
on Hyperbilirubinemia, 2004; Smits-Wintjens et al, 2008;
Walsh and Molloy, 2009). Intravenous immunoglobulin
is also indicated for treatment of immune thrombocytopenia (Bussel and Sola-Visner, 2009). The administration directly into the ocular vitreous of antibody against
vascular endothelial growth factor to bind and inactivate
vascular endothelial growth factor has been shown in early
studies to reduce the need for laser therapy for retinopathy of prematurity (Mintz-Hittner and Best, 2009). The
historically promising use of oral immunoglobulin for
preventing necrotizing enterocolitis has not proved effective in a recent Cochrane review of three trials (n = 2095
infants) (Foster and Cole, 2004). In view of the continued
expansion of neonatal diseases that are potentially treatable with immunoglobulin, the use of immunoglobulin
therapy must continue to be evaluated not only in terms of
immediate benefits, but also in terms of long-term effects
on immunologic function.
SPECIFIC IMMUNOLOGIC
DEFICIENCIES
The most common reason for increased immunologic
susceptibility to infection in newborns, besides prematurity, is iatrogenic immunosuppression caused by administration of corticosteroids for treatment or prevention
of bronchopulmonary dysplasia. Although pulmonary
and neurodevelopmental benefits have been attributed
to this therapy (Abman and Groothius, 1994; Cummings
et al, 1989), caution has developed concerning possible
long-term, adverse neurodevelopmental effects of steroid
administration (Bancalari, 2001; Barrington, 2002; Committee on the Fetus and Newborn, 2002). Although the
mechanisms that lead to steroid-induced amelioration of
pulmonary disease have not been elucidated completely,
the pulmonary inflammatory response as measured by the
concentrations of the anaphylatoxin C5a, leukotriene B4,
IL-1, elastase‑α1‑proteinase inhibitor, and the number
of neutrophils has been shown to be attenuated in infants
receiving steroid treatment (Groneck et al, 1993). Whereas
shorter courses of steroids can reduce side effects, including immunosusceptibility, the availability of nebulized steroids may provide effective antiinflammatory therapy with
minimum toxicity (Cole et al, 1999).
462
PART IX Immunology and Infections
There are more than 130 recognized primary immunodeficiency diseases (Notarangelo, 2010). The physician should attempt to differentiate infants with specific
genetically regulated immunologic deficiencies from those
with developmentally regulated, environmentally induced,
or infection-related susceptibility to microbial invasion
(Rosen, 1986; Rosen et al, 1984). Documenting a full
family history during an antenatal visit can be helpful in
identifying relatives removed by as many as two to four
generations with histories suggestive of primary immunodeficiency disease, as most arise from single-gene defects.
Genetic testing is widely available for many primary
immunodeficiency diseases, although de novo mutations
are common (Notarangelo, 2010). Several single-gene
defects resulting in blocks in T and B cell development are
shown in Figure 36-5. Several disorders arising from these
defects can manifest in the first months of life.
SEVERE COMBINED IMMUNODEFICIENCY
Severe combined immunodeficiencyt (SCID) is a rare category of diseases that affect development of both B cells
and T cells. The estimated frequency is between 1 per
70,000 and 1 per 100,000 live births (Buckley et al, 1997;
Stephan et al, 1993). Before identifying the genes involved
in producing the SCID phenotype, investigators classified
patients with the disorder by analyzing the number, cell
surface proteins, and function of circulating lymphocytes.
The most common subgroups are T-B- SCID, T-B+SCID,
and adenosine deaminase deficiency.
T-B+ SCID accounts for more than half of all SCID
cases, and both X-linked and autosomal recessive forms
exist (Fischer, 2000). Approximately 80% of T-B+ SCID
myeloid
precursor
cases involve males with the X-linked form, which results
from deleterious mutations in the common gamma chain
(γc) shared by the IL-2, IL-4, IL-7, IL-9, and IL-15 cytokine receptors (see Figure 36-5) (Noguhi et al, 1993).
Defects in the γc protein abrogate the development of T
cells and NK cells and lead to B cell dysfunction despite
normal B cell numbers. IL-7 receptor signaling is essential for T cell development within the thymus, and the
deficiency of this cytokine accounts for the absence of T
cells (Cao et al, 1995; DiSanto et al, 1995; Peschon et al,
1994). IL-15 is implicated in NK cell development, and
reductions in IL-4 receptor signaling are thought to contribute to poor B cell function seen in γc deficient patients
(Mrozek et al, 1996). Nevertheless, the precise mechanisms involved in the NK cell developmental defect and
the B cell functional defect are largely unknown.
The less common autosomal recessive form of
T-B+SCID is primarily due to mutations in the Janus-associated tyrosine kinase JAK3 (Macchi et al, 1995; Russell
et al, 1995). This kinase is expressed in cells of hematopoietic lineage and associates with γc as part of the cytokine
receptor signaling cascade. The T-B+NK- immunophenotype of these patients is identical to that seen in γc deficiency. Rarely, patients with autosomal recessive T-B+
SCID have NK cells. In two such patients, a defect was
found in the IL-7Rα chain (Puel et al, 1998). This finding was predicted by the phenotype of the IL7R knockout
mouse, and the data imply that the IL-7 receptor is not
essential for NK cell development.
Clinically, the X-linked and autosomal recessive forms
of T-B+ SCID are indistinguishable. Affected infants
often appear healthy at birth. In the neonatal period
a morbilliform rash, probably the result of attenuated
PERIPHERY
pro NK
NK
coronin-1A
THYMUS
AK2
HSC
Orai-1
Stim1
CLP
pro T
JAK-3
IL-7R
CD3 , ,
TAP1,2
Tapasin
ZAP70
CD8
RAG-1,2
Artemis
Cernunnos
DNA ligase 4
DNA-PKcs
CD4
MHC cl. II def.
mature B
CD40, CD40L,
pre B
immature B
pro B
AID, UNG,
RAG-1,2
TACI, ICOS,
Artemis, Cernunnos
CD19
DNA ligase 4, DNA-PKcs
switched B cell
BTK, mu heavy chain, 5,
Ig, Ig, BLNK, LRRC8
FIGURE 36-5 Blocks in T and B cell development associated with severe combined immunodeficiency and other primary immunodeficiencies. NK,
Natural killer. (From Notarangelo LD: Primary immunodeficiencies, J Allergy Clin Immunol 125:S182-S194, 2010.)
CHAPTER 36 Immunology of the Fetus and Newborn
graft‑versus‑host disease from transplacental passage of
maternal lymphocytes, may be the only symptom of SCID
(Rosen, 1986). Over the first several months of life, as
acquired maternal antibody levels drop, failure to thrive
and undue susceptibility to infection become universal
features. Intractable diarrhea, pneumonia, and persistent thrush, especially oral thrush, constitute the triad of
findings most frequently seen in infants with this disease
(Stephan et al, 1993).
Mutations in the gene encoding adenosine deaminase
(ADA), an enzyme in the purine salvage pathway, account
for approximately 20% of all cases of SCID. Mutations
in another enzyme in the purine salvage pathway, purine
nucleoside phosphorylase, are found in approximately
4% of patients with SCID (Fischer, 2000; Fischer et al,
1997). The ADA-deficient phenotype is variable, and neonatal onset, delayed onset, and partial forms have been
described (Santisteban et al, 1993). In neonatal-onset
ADA deficiency, patients have a profound T, B, and NK
cell lymphopenia, with a clinical presentation similar to
T-B+ SCID (Hirschhorn, 1990). Although ADA is normally present in all mammalian cells, life-threatening disease in ADA deficiency is limited to the immune system.
Less severe manifestations include flaring of the costochondral junction as seen on the lateral chest radiograph
(termed rachitic rosary) and pelvic dysplasia. Other findings
associated with ADA deficiency include hepatic and renal
dysfunction, deafness, and cognitive problems (Fischer,
2000; Rogers et al, 2001). Patients who lack ADA accumulate deoxy-adenosine triphosphate in red blood cells and
lymphocytes, and the concentration correlates with disease severity. The ADA substrates, adenosine and deoxyadenosine, are found at increased levels in the serum and
urine. Studies in ADA-deficient mice suggest that developing thymocytes are particularly sensitive to these metabolic derangements and that the few T cells that mature
have signaling defects (Blackburn et al, 2001). The precise
mechanism by which the accumulation of ADA substrates
results in the immunologic pathology seen in humans
remains unclear (Yamashita et al, 1998).
The remaining 20% of SCID patients lack mature T
and B cells, but have functional NK cells (T-B-NK+).
These patients exhibit an autosomal recessive pattern of
inheritance and have defects in the VDJ recombination
machinery (Schwarz et al, 1996). Somatic recombination
of variable, diversity, and joining DNA segments is a critical step in the development of B and T cells. A rearranged
IgM heavy chain and TCR-β chain form components
of the pre–B and pre–T receptors, which provide essential survival signals for developing lymphocytes. In the
absence of recombination, these lymphocyte precursors
do not receive a survival signal, die, and produce the T-BSCID phenotype. NK cells do not require somatic cell
recombination and survive. In the recombination process,
DNA cleavage is mediated by RAG1 and RAG2, which
are the recombination-activating proteins that recognize
specific sequences flanking V, D, and J segments. The two
proteins act in concert to introduce a double-strand DNA
break and leave hairpin-sealed coding ends. Recruitment
of a DNA-dependent protein kinase and other components of the general DNA repair machinery completes the
process.
463
Mutations in the genes encoding both RAG1 and RAG2
have been identified in patients with T-B-NK+ SCID
(Cornero et al, 2000) and in some patients with Omenn’s
syndrome (Cornero et al, 2001). Omenn’s syndrome
is clinically characterized by failure to thrive, diarrhea,
erythroderma, alopecia, hepatosplenomegaly, and lymphadenopathy. This concurrence of RAG mutations implies
that factors other than RAG mutations also contribute to
the Omenn’s syndrome phenotype. Finally, a small subset
of T-B-NK+ SCID patients also have increased sensitivity to ionizing radiation, which suggests mutations that
affect the DNA repair mechanism (Fischer, 2000). This
supposition is strengthened by murine and equine SCID
in which specific defects in the DNA-dependent protein
kinase involved in VDJ recombination and DNA repair
have been identified.
The diagnosis of SCID is often suggested by an opportunistic or unusually severe infection in the setting of
profound lymphopenia (<1000 lymphocytes per mm3)
(Gennery and Cant, 2001). Only 10% of patients with
SCID have lymphocyte counts in the normal range. T cells
detectable in peripheral blood of affected infants shortly
after birth may be either maternal T cells or circulating
thymocytes. The thymus gland is not seen on chest radiographs. Histologically, the gland is composed of islands
of endodermal cells that have not become lymphoid and
contain no identifiable Hassall’s corpuscles. Lymphocyte
subpopulation analysis using monoclonal antibodies to
cell surface markers and analytical flow cytometry is the
most important confirmatory test. As noted previously,
each phenotypic pattern suggests a specific diagnosis and
molecular defect. Other useful tests include measurements
of ADA and purine nucleoside phosphorylase activity in
red blood cells and isohemagglutinins as a marker of specific IgM production. Quantitative immunoglobulin levels
are not particularly helpful in the diagnosis of neonatal
SCID, because most IgG is maternal in origin and IgA and
IgM levels are often low in the neonatal period. Once the
immunophenotype has been established, a precise molecular diagnosis should be obtained and genetic counseling
should be provided. Females carrying deleterious mutations in the IL2RG gene (X-linked SCID) can be identified by nonrandom inactivation of the X chromosome in
lymphocytes. Prenatal diagnosis is available by gene identification from a chorionic villous biopsy. In the absence of
definitive prenatal genetic testing, all newborn siblings of
patients with known SCID, and infant male first cousins of
patients with known or suspected X-linked SCID, should
be considered at high risk for this disorder and investigated promptly after birth.
An effective strategy for detecting newborn infants
with SCID with statewide newborn screening was first
implemented in Wisconsin (Routes et al, 2009). The test
is based on the detection of T cell recombination excision circles by PCR and also identifies patients with lymphopenia resulting from other causes (Baker et al, 2009;
Chan and Puck, 2005). When a fetus at risk for a genetic
form of SCID is identified, treatment should begin in the
delivery room and should be coordinated with antenatal
diagnostic interventions. Specifically, cord blood samples
should be obtained for white blood cell count and differential, lymphocyte subset determinations, karyotype (if not
464
PART IX Immunology and Infections
performed antenatally), mitogen stimulation studies, and
immunoglobulin measurements. Because the majority of
children with genetic SCID do not become ill within the
1st week of life, care in an incubator should be provided,
and staff members should observe strict handwashing
technique.
SCID is a pediatric emergency and is invariably fatal if
untreated. Most untreated patients die in the 1st year of
life. Treatment of SCID begins with aggressive antibiotic
and antiviral therapy for infections, intravenous immunoglobulin replacement, and prophylaxis for Pneumocystis
carinii infection. Besides greater susceptibility to opportunistic infections, these infants are also susceptible to development of graft‑versus‑host disease, either before birth
as a result of engraftment of maternal T lymphocytes or
after birth as a result of the engraftment of T lymphocytes
present in transfused blood products (Pollack et al, 1982;
Thompson et al, 1984). Therefore infants in whom SCID
is suspected should receive only irradiated blood products
and should not be given live viral vaccines.
Good and colleagues performed the first successful bone
marrow transplantation (BMT) in a SCID patient (Gatti
et al, 1968). Allogeneic BMT, preferably from an HLAmatched sibling, is now the standard treatment for most
types of SCID (Buckley, 2000). In these transplants, conditioning regimens are not required for engraftment. Recipients usually become chimeric, with only T and NK cells
of donor origin. B cell function is frequently deficient and
many patients continue to require monthly therapy with
IVIG. Survival rates for this type of BMT exceed 90%.
When no HLA-identical donor is available, haploidentical
transplants have been used successfully, although overall
survival is lower with such transplants (Fischer, 2000). The
rates of engraftment after haploidentical transplantation of
T-B-NK+ SCID patients are low; this is likely due to host
NK cell function, and some form of preconditioning may
be indicated in this subset of patients. In addition to BMT,
ADA deficiency can be treated with enzyme replacement.
This treatment involves weekly injections of ADA coupled
to polyethylene glycol. Response, consisting of decreasing
deoxyadenosine triphosphate levels and increasing T cell
numbers, is seen in most patients within weeks. Finally,
gene therapy based on vector-mediated transfer of therapeutic gene into autologous hematopoietic stem cells has
been used to treat patients with ADA and γc SCID and
patients with chronic granulomatous disease. Results in
more than 30 patients showed engraftment and clinical
benefit, but were limited by transient expression and by
insertional mutagenesis (Aiuti and Roncarolo, 2009).
WISKOTT‑ALDRICH SYNDROME
Wiskott‑Aldrich syndrome (WAS), another form of
primary immunodeficiency, is characterized by severe
eczema, thrombocytopenia, increased risk of malignancy, and susceptibility to opportunistic infection.
Other manifestations, which may be present in the newborn period, include petechiae and bruises, bloody diarrhea, and hemorrhage after procedures. In infants with
any of these clinical findings, abnormal low mean platelet
volume in a complete blood count report is an important
clue to possible WAS. WAS is inherited as a sex‑linked
recessive trait (Rosen, 1986). In untreated cases, children survive longer than infants with SCID (median
survival, 5.7 years). T lymphocytes in affected patients
are decreased in number and diminished in function.
Reductions in platelet size and thrombopoiesis are also
noted. Affected children can be treated with BMT, and
the 5-year survival rate after HLA-identical sibling BMT
is approximately 90% (Filipovich et al, 2001). Although
transplantation corrects the T cell defects, thrombocytopenia persists. As for children with SCID, all blood
products given to children with WAS should be irradiated before administration to avoid T cell engraftment
and graft‑versus‑host disease.
The gene responsible for the WAS was cloned in 1994
(Derry et al, 1994). It is composed of 12 exons and encodes
a 502 amino acid cytoplasmic protein (WAS) expressed
in all hematopoietic cells. Evaluation by flow cytometry
for expression of this protein by lymphocytes can be used
to diagnose WAS. Other mammalian WAS family members include a more widely expressed N-WAS and three
WAVE/Scar isoforms. These proteins have multiple
domains and are important regulators of actin polymerization (Symons et al, 1996); they have a common carboxyterminal region through which they activate Arp2/3, an
actin-nucleating complex involved in actin assembly and
cytoskeletal structure. The amino termini are distinct
and allow the proteins to couple Arp2/3 activation to a
wide variety of different intracellular signals. More than
340 mutations in WAS have been described, which have
profound effects on cell motility, signaling, and apoptosis
(Rengan and Ochs, 2000).
DiGEORGE SYNDROME
The embryologic anlage of the thymus gland and the parathyroid gland is the endodermal epithelium of the third
and fourth pharyngeal pouches. When normal development of these structures is disturbed, thymic and parathyroid hypoplasia can occur (Rosen, 1986). Infants with
this disorder, DiGeorge syndrome, can exhibit abnormalities of calcium homeostasis during the neonatal period
(hypocalcemia and tetany) and variable T cell deficits,
which appear to depend on the presence and number of
small, normal‑appearing ectopic thymic lobes. In addition,
these infants have congenital, conotruncal cardiac defects,
low‑set ears, midline facial clefts, hypomandibular abnormalities, and hypertelorism.
The availability of methodology for performing gene
dosage studies and more refined cytogenetic techniques
(fluorescence in situ hybridization) has permitted the
description of a contiguous gene syndrome that includes
the DiGeorge phenotype (Hall, 1993). The DiGeorge syndrome is known to result from varying sized deletions on
chromosome 22q11 in more than 90% of patients (Markert, 1998; Shprintzen, 2008). This deletion has been linked
to several other diagnostic labels, including velocardiofacial (Shprintzen) syndrome, Cayler syndrome, and Opitz
G/BBB syndrome. Collectively, these are referred to as
the 22q11 deletion syndromes. The cardiac anomalies associated with the 22q11 deletion syndrome are variable, but
usually involve the outflow tract and the derivatives of the
branchial arch arteries. These defects include interrupted
CHAPTER 36 Immunology of the Fetus and Newborn
aortic arch type B, truncus arteriosus, and tetralogy of Fallot (Carotti et al, 2008).
Children with the 22q11 deletion syndrome also exhibit
a higher incidence of receptive-expressive language difficulties, cognitive impairment, and behavioral problems
including psychotic illness (Jolin et al, 2009; Scambler,
2000). The cardiovascular defects have been shown to be
the result of haploinsufficiency of TBX1 (Merscher et al,
2001).
In the nursery, identification of infants with congenital conotruncal abnormalities or unexplained, persistent
hypocalcemia should prompt consideration of this syndrome. Thymic implants can partially correct the immunologic deficits (Markert et al, 2007).
IMMUNIZATION
MATERNAL IMMUNIZATION
Immunization before or during pregnancy has been effective in preventing several specific neonatal infections
including diphtheria, pertussis, tetanus, hepatitis B, and
rabies (ACOG Technical Bulletin Number 160, 1993;
Hackley, 1999; Immunization Practices Advisory Committee, 1991; Stevenson, 1999). For example, in developing
countries, immunization during pregnancy with tetanus
toxoid is a cost‑effective method for preventing neonatal
tetanus and for providing up to 10 years of protection for
infants (Gill, 1991; Schofield, 1986; Vandelaer, 2003). The
benefits are substantial for both mother and infant from
induction before or during pregnancy of maternal IgG
antibody that can be transferred to the fetus and protect
both the fetus and mother against postpartum morbidity
and mortality (Healy and Baker, 2006; Insel et al, 1994;
Linder and Ohel, 1994). However, immunization during
pregnancy is biologically distinct from immunization of
nonpregnant individuals. Vaccine epitopes can be shared
with vital fetal or placental tissues; therefore vaccination
can lead to unanticipated maternal or fetal morbidity.
Maternal immunization can induce an antibody response
in the fetus, as has been demonstrated with tetanus toxoid
(Gill et al, 1983) and thereby induce potentially undesirable immunologic side effects (e.g., immunologic unresponsiveness or tolerance) in the infant.
Nevertheless, the increase in availability of potentially
protective, transplacentally transferred IgG through active
maternal vaccination prompted the Institute of Medicine
to recommend the establishment of a program of active
immunization to control early-onset and late-onset group
B streptococcal disease in both infants and mothers (Institute of Medicine and National Academy of Sciences, 1985).
Efforts to develop safe, effective vaccines for protection
from group B streptococcal infections have encountered
the same difficulties with immunogenicity and safety
observed in the development of other vaccines that induce
protection from polysaccharide‑encapsulated organisms
(Baker et al, 1988; Noya and Baker, 1992). The availability
of conjugate vaccines, which include group B streptococcal
polysaccharide antigens covalently linked either to tetanus
toxoid or to a protein in the membrane of group B streptococci (beta C protein) (Madoff et al, 1994; Wessels et al,
1993; Yang et al, 2007) have shown some promise.
465
The indications for active vaccination during pregnancy
rest on assessment of maternal risk of exposure, the maternalfetal-neonatal risk of disease, and the risk from the immunizing agents. (ACOG Technical Bulletin Number 160, 1993;
Hackley, 1999; Stevenson, 1999). In general, immunization with live viral vaccines during pregnancy is not recommended (Box 36-1). Preferably, immunizations with live viral
vaccines are performed before pregnancy occurs. However,
rare instances may occur in which live viral vaccine administration is indicated. For example, if a pregnant woman travels to an area of high risk for yellow fever, administration of
that vaccine might be indicated because of the susceptibility of the mother and the fetus, the probability of exposure,
and the risk of the mother and fetus from the disease. More
common examples in the United States include influenza
and polio virus vaccination. If a chronic maternal medical
condition would be adversely affected by influenza, active
immunization may be indicated during pregnancy. Similarly,
if imminent exposure to live polio virus in an unprotected
woman is anticipated, live oral polio virus vaccine may be
used during pregnancy. If immunization can be completed
before the anticipated exposure, inactivated polio virus
vaccine can be given. A summary of recommendations for
immunizations during pregnancy is provided in Box 36-1.
For the pediatrician, maternal immunization represents an important preventive intervention. Breastfeeding
does not adversely affect immunization, and inactivated or
killed vaccines pose no special risk for mothers who are
breastfeeding or for their infants. Maternal immunizations with vaccines against polysaccharide‑encapsulated
BOX 36-1 Summary of Recommendations
for Immunization during Pregnancy
LIVE VIRUS VACCINES
Influenza (LAIV)—contraindicated
ll Measles—contraindicated
ll Mumps—contraindicated
ll Rubella—contraindicated
ll Yellow fever—safety not established (travel to high‑risk areas only)
ll Varicella—contraindicated
ll
INACTIVATED VIRUS VACCINES
Hepatitis A—consider if high risk of exposure
ll Influenza—recommended
ll Rabies—consider if indicated
ll
INACTIVATED AND RECOMBINANT BACTERIAL VACCINES
Cholera—to meet international travel requirements
ll Meningococcal polysaccharide vaccine—consider if indicated
ll Plague—selective vaccination of exposed persons
ll Typhoid—safety not established
ll Pneumococcal polysaccharide vaccine—consider if indicated
ll Tetanus‑diphtheria—consider if indicated
ll Hepatitis B—consider if indicated
ll Meningococcal conjugate vaccine—safety not established
ll
POOLED IMMUNE SERUM GLOBULINS
ll
ll
Hepatitis A—postexposure prophylaxis
Measles—postexposure prophylaxis
Adapted from Centers for Disease Control and Prevention: Guidelines for Vaccinating
Pregnant Women (May, 2007). Available at www.cdc.gov/vaccines/pubs/preg-guide.htm.
LAIV, Live attenuated influenza vaccine.
466
PART IX Immunology and Infections
organisms (e.g., Haemophilus influenzae type b and group B
streptococci) can decrease morbidity and mortality from
these diseases during the first 3 to 6 months of the infant’s
life (Amstey et al, 1985; Baker et al, 1988; Colbourn et al,
2007; Walsh and Hutchins, 1989). The benefit of decreasing the risks of development of hepatocellular carcinoma,
cirrhosis, and chronic active hepatitis from perinatal transmission of hepatitis B through prenatal screening and
active and passive immunization of the infant is substantial (Arevalo and Washington, 1998). The implications of
maternal vaccination during pregnancy for preterm infants
have not been studied.
The anthrax attacks of September 2001 have raised
the possibility of the need for preexposure or postexposure immunization programs that may include pregnant
women. Currently available data tend to suggest that
the anthrax vaccine licensed since 1970 in the United
States has no detrimental effects on pregnancy and does
not increase adverse birth outcomes, although caution is
advised when administering this vaccine during the first
trimester (Inglesby, 2002; Ryan et al, 2008; Wiesen and
Littell, 2002). A second infectious agent that might be
used in a bioterrorist attack is variola virus (smallpox).
Although eradicated in 1977, this virus remains a concern
because of its lethality, especially in pregnancy (Enserink,
2002; Suarez and Hankins, 2002). Pregnancy is a contraindication to smallpox immunization (Wharton et al, 2003);
however, if an intentional release of smallpox virus should
occur, pregnant women should be immunized because
of their high risk of mortality if unprotected (Suarez and
Hankins, 2002).
INFANT IMMUNIZATION
Advances in understanding the developmental regulation
of immunity have suggested that immunization during
the neonatal period offers important advantages (Lawton,
1994). The recommendations of the American Academy
of Pediatrics for immunization of infants can be found
at http://aapredbook.aappublications.org/resources/IZ
Schedule0-6yrs.pdf (Anonymous, 2002; Committee on
Infectious Diseases et al, 2002). Immunization against
H. influenzae type b should be considered for infants who
are discharged from intensive care nurseries at or after 2
months of age. For the preterm infant, different clinical
approaches to immunization are used by practitioners,
including decreasing the dose of immunogen, postponing
the first immunization until a corrected age of 2 months, or
waiting for an arbitrary weight to be achieved by the infant
(e.g., 4.5 kg). However, the American Academy of Pediatrics recommends administering full‑dose diphtheria, tetanus, and pertussis immunization beginning at 2 months
of age. These recommendations (i.e., that no correction
needs to be made for prematurity when initiating routine
immunization in preterm infants) have been supported by
longitudinal evaluation of serum antibody response in preterm infants (Bernbaum et al, 1989; Conway et al, 1993).
The Advisory Committee on Immunization Practices
(ACIP) of the United States Public Health Service recommends universal immunization of infants to protect
against perinatal transmission of the hepatitis B virus
(HBV) and chronic HBV infection (Mast et al, 2005). This
recommendation has been endorsed by the Committee on
Infectious Diseases of the American Academy of Pediatrics. Chronic HBV infection occurs in approximately 90%
of infected infants and is associated with hepatocellular
carcinoma and cirrhosis leading to end-stage liver disease.
All medically stable infants born to women with a negative test result for hepatitis B surface antigen (HBsAg) and
weighing more than 2000 g at birth should begin a hepatitis immunization schedule in the newborn period before
hospital discharge. Only single-antigen hepatitis B vaccines should be used for the birth dose (Mast et al, 2005).
All infants regardless of gestational age at the time of
birth, and whose mothers have a positive test result for
HBsAg, should receive passive immunization with hepatitis B immunoglobulin and active immunization with
a single-antigen HBV vaccine within 12 hours of birth.
Repeated vaccinations are given at 1 to 2 months and
again at 6 months of age. For premature infants weighing less than 2000 g and born to HBsAg-positive mothers, the birth dose is not counted as part of the vaccine
series. Special efforts should be made to complete the
hepatitis B vaccination schedule within 6 to 9 months in
populations of infants with high rates of childhood hepatitis B infection (Peter, 1994). For premature infants with
birthweights of less than 2000 g born to HBsAg‑negative
women, vaccination can be delayed until just before discharge. These infants do not need routine serologic testing
for anti-HBsAg after the third dose of vaccine. The infant
whose mother has a negative test result for HBsAg, but has
received active or passive immunization during pregnancy
because of exposure to hepatitis B, should receive no treatment as long as the mother was HBsAg‑negative at the
time of birth. For current HBV vaccinations recommendations, the reader is directed to the Centers for Disease
Control and Prevention Web site: www.cdc.gov/hepatitis/
HBV/VaccChildren.htm.
In 1999, the U.S. Agency for Toxic Substances and
Disease Registry raised concern about the possibility that
administration of thimerosal-containing vaccines, including the hepatitis B vaccines, might lead to exposure to mercury that exceeded federal guidelines (Clark et al, 2001).
Since March 2000, HBV vaccines used in the United
States have not contained thimerosal. These yeast‑derived
recombinant HBV vaccines contain 10 to 40 μg/mL of
HBsAg protein, have excellent safety records, induce
minimum adverse reactions, and are highly immunogenic
(Greenberg, 1993). Multivalent vaccines are available, but
are not recommended for the birth dose. Guidelines for
hepatitis immunization of infants have been developed for
implementation for term and preterm infants. However,
vaccine responses in the extremely preterm infant whose
mother is HBsAg‑positive, a population seen with increasing frequency because of the coincidence of intravenous
drug abuse and carriage of HBsAg, have not been studied.
Short-term and long-term neonatal immunologic protection after fetal exposure to maternally administered
vaccines against anthrax or smallpox, the mass use of which
might be prompted by a bioterrorist attack, has not been
studied. Similarly, guidelines for neonatal immunization
against these agents are not currently available. Before
the eradication of smallpox, smallpox vaccine was routinely administered to older infants and children. No data
CHAPTER 36 Immunology of the Fetus and Newborn
or experience is available on immunization of newborn
infants with anthrax vaccine. For postexposure prophylaxis in newborn infants, both chemotherapy (amoxicillin,
ciprofloxacin, or doxycycline) and immunization should
be considered. Clinicians faced with decisions concerning
immunization strategies for pregnant women and newborn infants after a bioterrorist attack should consult the
Web site for the Centers for Disease Control and Prevention (www.cdc.gov).
SUGGESTED READINGS
Chase NM, Verbsky JW, Routes JM: Newborn screening for T-cell deficiency,
Curr Opin Allergy Clin Immunol 10:521-525, 2010.
D’Argenio DA, Wilson CB: A decade of vaccines: Integrating immunology and
vaccinology for rational vaccine design, Immunity 33:437-440, 2010.
Koga K, Mor G: Toll-like receptors at the maternal-fetal interface in normal
pregnancy and pregnancy disorders, Am J Reprod Immunol 63:587-600, 2010.
467
Leber A, Teles A, Zenclussen AC: Regulatory T cells and their role in pregnancy,
Am J Reprod Immunol 63:445-459, 2010.
Levy O: Innate immunity of the newborn: basic mechanisma and clinical correlates,
Nat Rev Immunol 7:379-390, 2007.
M’Rabet L, Vos AP, Boehm G, et al: Breast-feeding and its role in early development of the immune system in infants: consequences for health later in life,
J Nutr 138:782S-1790S, 2008.
Mor G, Cardenas I: The immune system in pregnancy: a unique complexity, Am J
Reprod Immunol 63:425-433, 2010.
PrabhuDas M, Adkins B, Gans H, et al: Challenges in infant immunity: implications for responses to infection and vaccines, Nat Immunol 12:189-194, 2011.
Trowsdale J, Betz AG: Mother’s little helpers: mechanisma of maternal-fetal tolerance, Nat Immunol 7:241-246, 2006.
Willems F, Vollstedt S, Suter M: Phenotype and function of neonatal DC, Eur J
Immunol 39:26-35, 2009.
Zaghouani H, Hoeman CM, Adkins B: Neonatal immunity: faulty T-helpers and
the shortcomings of dendritic cells, Trends Immunol 30:585-591, 2009.
Zhu J, Yamane H, Paul WE: Differentiation of effector CD4 T cell populations,
Annu Rev Immunol 28:445-489, 2010.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
37
Viral Infections of the Fetus and Newborn
and Human Immunodeficiency Virus
Infection during Pregnancy*
Mark R. Schleiss and Janna C. Patterson
Viral Infections of the
Fetus and Newborn
Viral infections of the fetus and newborn infant are common and underrecognized. Given the urgency of identifying invasive bacterial disease in the neonate, the
identification of viral infections is often relegated to a matter of secondary importance. However, identifying viral
infections can also be a matter of great urgency, because
antiviral agents are available for many of the most common infections. Accordingly, an appropriately high index
of clinical suspicion of a neonatal viral infection can be
lifesaving. Moreover, identification of viral disease in the
newborn period may be of great prognostic significance,
particularly for neurodevelopmental issues. Making a
diagnosis of a neonatal viral infection can help to direct
and focus the anticipatory management of the child’s pediatrician during the course of well-child care. This chapter
reviews the epidemiology, pathogenesis, diagnosis, and
short- and long-term clinical management of many of the
more common viral infections encountered in neonates.
One of the great challenges in the evaluation of viral disease in the newborn is the ascertainment of the timing of
acquisition of infection. Some infections can be acquired
either in utero or in the early postnatal period. In this chapter, congenital infection is defined as any infection acquired in
utero. Perinatal infections are defined as those acquired intrapartum, typically during the labor and delivery process. Postnatal infections are acquired in the post-partum period and are
defined as infections acquired after delivery through the first
month of life. In some situations, correctly identifying the
timing of acquisition of infection can have substantial consequences, not only for the management of the infant, but for
the long-term prognosis. Figure 37-1 outlines the most common timing of acquisition of neonatal viral infections with
an emphasis on the relative importance of the many viruses
that can be acquired congenitally, perinatally, or postnatally.
There is considerable overlap across categories for some viral
infections that can be transmitted at any of these time points,
and this will be considered on a pathogen-by-pathogen basis.
GENERAL DIAGNOSTIC APPROACH
Clinicians caring for newborns have long recognized
that there are some common clinical manifestations that
suggest the presence of a congenital or perinatal viral
*In writing this chapter, the authors excerpted portions of the chapters Identification, Evaluation, and Care of the Human Immunodeficiency Virus–Exposed Neonate by
Karen P. Beckerman and Viral Infections of the Fetus and Newborn by Erica S. Pan,
F. Sessions Cole, and Peggy Sue Weintrub from the previous edition of this book.
468
infection. These manifestations include evidence of intrauterine growth restriction, hydrops fetalis, hepatomegaly,
splenomegaly, pneumonitis, bone lesions, rashes, and
hematologic abnormalities (Box 37-1). Because the incidence of congenital viral infections is high (Alpert and
Plotkin, 1986), it is appropriate for clinicians to have a high
index of suspicion in any newborn with signs or symptoms.
However, caution should be taken in efforts to lump all
neonatal viral infections into a single diagnostic category.
An unfortunate example of this manner of thinking is the
continued use of the terminology TORCH titers in the diagnostic approach to a symptomatic neonate. The TORCH
acronym, first coined by Nahmias et al (1971), stands for
toxoplasmosis, other infections, rubella, cytomegalovirus
(CMV), and herpes simplex virus (HSV). Numerous variants of this acronym have been suggested over the past
four decades (Ford-Jones and Kellner, 1995; Kinney and
Kumar, 1988; Ronel et al, 1995; Tolan, 2008). Unfortunately numerous clinical laboratories continue to offer the
“TORCH panel,” typically consisting of serologic tests for
toxoplasmosis, HSV, CMV, and rubella. This acronym has
outlived its usefulness (Lim and Wong, 1994), and should
be discarded from clinical parlance, based on the following
considerations:
ll Measurements of immunoglobulin G antibody titers
virtually always simply reflect transplacental maternal
antibody and provide little information of relevance
to the infant’s infection status. With the exception of
the identification of antibodies to Treponema pallidum,
which is always of interest and significance, antibodies
against the other members of the TORCH panel are of
little diagnostic significance.
ll Congenital and perinatal infection can occur with HSV
and CMV, even in the face of preconception maternal
immunity. Thus the finding of antibody in a TORCH
titer is neither diagnostic of infection nor reassuring in
regard to protection against that infection.
ll Highly sensitive virologic and molecular tools are available to identify virtually all pathogenic viruses. These
tools include standard culture and nucleic acid identification techniques, typically based on polymerase
chain reaction (PCR) amplification of viral nucleic
acids. Such studies can facilitate rapid pathogen-specific diagnosis; therefore the diagnosis of neonatal
viral disease should depend on diagnostic virology, not
serology.
ll Most importantly, the use of the TORCH acronym
vastly underemphasizes the great diversity of viral
pathogens that have been associated with infection in
the newborn. A list of the myriad of viral pathogens
that have been reported to cause congenital infection
and disease is included in Box 37-2. A discussion of
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
Prenatal
Perinatal/intrapartum
Cytomegalovirus
Parvovirus B19
Varicella-zoster virus
Rubella
LCMV*
HSV**
Parechovirus
EBV***
HHV 6, 7
HSV
Hepatitis B, C
Enterovirus
Varicella-zoster virus
Cytomegalovirus
Adenovirus
Parechovirus
Postnatal
Respiratory
syncytial virus
Enterovirus
Rotavirus
Cytomegalovirus
Varicella-zoster
virus
Hepatitis
Adenovirus
Influenza
* Lymphocytic choriomeningitis virus
** Herpes simplex virus
*** Epstein Barr virus
FIGURE 37-1 Relative importance of neonatal viral infections
related to timing of acquisition of infection. Viruses listed in declining relative order of importance relative to prenatal, perinatal (intrapartum), and postnatal timing of typical infection. Some neonatal virus
infections (e.g., cytomegalovirus) can be substantial causes of disease
whether acquired during gestation or postpartum, whereas others (e.g.,
respiratory syncytial virus) are typically acquired in the postnatal period.
EBV, Epsteinn–Barr virus; HSV, herpes simplex virus; HHV, human
herpes virus; LCMV, Lymphocytic choriomeningitis virus
BOX 37-1 C
linical Features Commonly
Associated With Congenital Viral
Infections in Neonates
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Intrauterine growth restriction
Nonimmune hydrops fetalis
Echogenic bowel (prenatal ultrasound)
Hepatosplenomegaly
Jaundice (>20% direct-reacting bilirubin)
Hemolytic anemia
Purpura, ecchymoses and petechiae
Skeletal defects (“celery-stalking”)
Microcephaly and hydrocephaly
Intracranial calcification
Neuronal migration defects
Pneumonitis
Myocarditis
Cardiac abnormalities
Chorioretinitis
Keratoconjunctivitis
Cataracts
Glaucoma
many of the more unusual viral causes of congenital
infection is beyond the scope of this chapter, but this
table underscores the diversity of agents associated with
fetal infection. A recent travel history or recent emigration might suggest consideration of some of these more
unusual agents.
Rather than rely on a large battery of serologic tests,
the clinician can usually narrow the differential diagnosis
of a suspect neonatal or congenital viral infection with
the history and physical examination, followed by the use
of focused, specific diagnostic studies. Important questions include: What was the overall health of the mother
during her pregnancy? What is her age and marital status (e.g., young, unmarried women have a higher risk of
469
BOX 37-2 V
iral Pathogens Reported to
Cause Congenital Infections
Adenoviridae
Adenovirus serogroup 3
Arenaviridae
ll Lymphocytic choriomeningitis virus
ll Lassa fever virus
Bunyaviridae
ll Bunyamwera serogroup (Cache Valley virus)
ll La Crosse encephalitis virus
Flaviviridae
ll Hepatitis C virus
ll Japanese encephalitis virus
ll West Nile virus
ll St. Louis encephalitis virus
ll Yellow fever virus
ll Dengue virus
Hepadenoviridae
ll Hepatitis B virus
Herpesvirinae
ll Herpes simplex viruses 1 and 2
ll Varicella zoster virus
ll Cytomegalovirus
ll Epstein–Barr virus
ll Human herpesviruses 6 and 7
Orthomyxoviruses and paramyxoviruses
ll Influenza
ll Measles
Parvoviridae
ll Human parvovirus B19
Picornaviridae
ll Poliovirus
ll Coxsackievirus
ll Enteric cytopathic human orphan virus
ll Parechovirus
ll Hepatitis A virus
Retroviridae
ll Human T-lymphotropic viruses 1 and 2
ll HIV
Togavirinae
ll Mumps
ll Western equine encephalitis virus
ll Venezuelan equine encephalitis virus
ll
acquiring primary genital HSV infection). What is her
immunization history? Has she had chickenpox? Did she
have other common childhood viral infections? What
part of the world is she from? Are there potential animal
exposures (e.g., exposure to cat litter or consumption of
undercooked meat might suggest toxoplasmosis; exposure to rodents might suggest lymphocytic choriomeningitis virus)? Does she have other children and, if so,
what are their ages, overall health status, and histories
of group day care attendance? Have there been recent
illnesses in the household? What time of year is it? (For
example, respiratory syncytial virus and enterovirus
infections have characteristic seasonality in temperate
zones.) What are her occupational exposures? Did she
have any symptomatic infectious illnesses during pregnancy? The answers to these types of questions, considered in the context of the infant’s physical examination,
can direct the next steps in establishing a definitive etiologic diagnosis.
470
PART IX Immunology and Infections
TABLE 37-1 Clinical Findings in Selected Congenital and
erinatal Infections That Suggest a Specific
P
Diagnosis
Congenital
Infection
Findings
Rubella
Cataracts, cloudy cornea, pigmented retina;
petechiae with “blueberry muffin” rash;
bone defects with longitudinal bands of
demineralization (“celery stalking”); cardiovascular malformations (patent ductus
arteriosus, pulmonary artery stenosis);
sensorineural hearing loss; hydrops
Cytomegalovirus
Microcephaly with periventricular calcifications; chorioretinitis; petechiae with
thrombocytopenia; jaundice; sensorineural
hearing loss; bone abnormalities; abnormal
dentition, hypocalcified enamel
Herpes simplex
virus
Skin vesicles, keratoconjunctivitis, acute
central nervous system findings (seizures),
hepatitis, pneumonitis
Parvovirus B19
Hydrops, ascites, hepatomegaly, ventriculomegaly, hypertrophic myocardiopathy,
anemia
Varicella zoster
virus
Limb hypoplasia, dermatomal scarring in
cicatricial pattern, gastrointestinal tract
atresia
Lymphocytic
choriomeningitis virus
Hydrocephalus, chorioretinitis, intracranial
calcifications
Some of the classic presentations of the more common
perinatal viral infections are reviewed in Table 37-1. It
should be noted that there can be considerable overlap of
these clinical features across the different infectious categories listed; for example, the “blueberry muffin” rash
of congenital rubella syndrome may be indistinguishable
from that of congenital CMV, and both syndromes can
include sensorineural deafness. The presence of brain calcifications is similarly nonspecific, and this finding should
always suggest a differential diagnosis that includes CMV,
toxoplasmosis, lymphocytic choriomeningitis virus, and
the recently described parechoviruses. Because neuroradiologic studies cannot reliably distinguish these entities,
definitive diagnostic virology is necessary. Specific viral
pathogens, their basic virology, the clinical manifestations
of diseases they cause in the newborn, management strategies, and prospects for prevention are considered on a
pathogen-specific basis in the remainder of this chapter.
HERPESVIRIDAE
Currently there are eight recognized human herpesviruses, which are subdivided into three categories based on
aspects of viral biology, pathogenesis, and clinical presentations. These categories are the α-herpesviruses, consisting of HSV-1, HSV-2, and varicella-zoster virus (VZV);
the β-herpesviruses, which include CMV and the roseola
viruses HHV-6 and HHV-7; and the γ-herpesviruses,
which include Epstein–Barr virus (EBV) and Kaposi’s sarcoma herpesvirus (KSHV), reviewed in Schleiss (2009).
Remarkably, all of these agents have been implicated in
varying degrees as causes of clinically important congenital
and perinatal infections, although these associations are
less well studied for the γ-herpesviruses.
HERPES SIMPLEX VIRUS INFECTIONS
HSV-1 and HSV-2 are highly related viruses. Although
classically HSV-1 has been identified as a cause of oral
infections (gingivostomatitis and pharyngitis), and HSV-2
has been implicated as the most common virus associated
with genital herpes, in recent years these distinctions have
become blurred. The greatest risk for the newborn is in
the context of a first-time episode of maternal genital HSV
infection occurring during pregnancy. The entity of neonatal herpes is reviewed in the following section, along
with current management approaches for this infection.
VIROLOGY, EPIDEMIOLOGY, AND CLINICAL
MANIFESTATIONS OF HSV DISEASE
HSV-1 and HSV-2 demonstrate a high degree of similarity, both at the molecular level and in their clinical manifestations. The degree of genetic relatedness of these two
viruses is approximately 45%, and the genome structures
and morphology of the virion (virus particle) are virtually
identical (Kieff et al, 1972). HSV-1 and HSV-2 are both
acquired predominantly at mucosal surfaces and require
intimate contact for transmission. After primary infection
in epithelial cells, intraaxonal trafficking of viral DNA to
the dorsal route ganglia results in the establishment of
latency. The latent state is characterized by the cessation
of virtually all gene transcription, except for the latencyassociated transcript (LAT), which is expressed in the dorsal route ganglia even as the virus maintains a quiescent
state (Steiner et al, 2007; Taylor et al, 2002). The function of the latency-associated transcript is unknown, but it
might involve a novel RNA-mediated mechanism, because
there does not appear to be a protein product associated
with the transcript (Umbach et al, 2008). After a number of triggers, including ultraviolet radiation, stress, and
immunosuppression, the virus reactivates at the level of the
dorsal route ganglia and initiates a cascade of viral transcription that leads to the production of infectious virus,
which can traffic via the axon to the cutaneous surface or
ocular surface, producing lesions (Toma et al, 2008). The
recrudescence of HSV lesions, usually manifest as vesicular or ulcerative lesions at the site of primary infection, can
in turn lead to person-to-person transmission, including
maternal-fetal and maternal-infant transmission. Importantly, clinically evident lesions need not be present for
person-to-person transmission of infection to susceptible
individuals, because asymptomatic or subclinical shedding
of virus is well documented, particularly in the setting of
genital herpes.
A wide variety of disease syndromes are associated with
primary and recurrent HSV infection. Classically HSV-1
has been described as causing disease “above the belt,”
whereas HSV-2 is associated with disease “below the belt.”
The finding of HSV-2 antibodies in seroprevalence studies
is generally viewed as indicative of genital herpes, and in
the pregnant patient it can be considered to be diagnostic of
genital herpes. The most common disease associated with
HSV infection is herpetic gingivostomatitis, characterized
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
typically by perioral and intraoral lesions involving the
pharyngeal mucosa. HSV infection of the oropharynx may
present in adolescence as herpetic pharyngitis (McMillan
et al, 1993); it can be indistinguishable from other causes
of pharyngitis. Other common manifestations of HSV
infection include primary cutaneous infection, which can
manifest as herpes gladiatorum or as a herpetic whitlow
(Johnson, 2004; Wu and Schwartz, 2009). In children with
atopic dermatitis, cutaneous HSV infection can be associated with eczema herpeticum, a serious illness that can be
associated with systemic symptom (Bussman et al, 2008).
All of these cutaneous manifestations of HSV could put
a newborn at risk for acquisition of infection if care is not
taken to protect the infant. HSV encephalitis is unlikely to
be transmitted person-to-person; it is the most common
sporadic cause of viral encephalitis in North America, and
it can be associated with primary infection or reactivation
of latent infection. It is most commonly associated with
HSV-1 (Baringer, 2008).
The most important manifestation of HSV disease is
maternal genital herpes. Genital herpes can be associated
with either HSV-1 or HSV-2. HSV is the most common cause of genital ulcerative disease in the developed
world, and the prevalence has increased steadily in recent
year (Fleming et al, 1997). Genital herpes is characterized by blisters, ulcers, or crusts on the genital area, buttocks, or both. Typically, symptomatic disease manifests
with a mixture of vesicles, ruptured vesicles with resulting ulcers, and crusted lesions. Systemic flulike symptoms
such as headache, fever, and swollen glands can accompany
an outbreak of genital herpes, particularly during primary
infection. Other symptoms include dysuria, urinary retention, vaginal or penile discharge, genital itching, burning
or tingling, and groin sensitivity. Genital lesions vary in
number, are painful in nature, and if untreated persist for
up to 21 days (Whitley et al, 1998). It has been recognized
in recent years that many individuals with genital herpes are asymptomatic and unaware of their status (Wald
et al, 2000). Therefore a negative maternal history of HSV
should not dissuade the clinician from considering the
possibility of neonatal herpes in an infant with compatible
signs and symptoms. Patients with recurrent symptomatic
episodes continue to shed virus in between episodes, even
after lesions have healed and crusted over (Leone, 2005).
This information has important implications for the continued evolution of the HSV epidemic in the United States.
Because individuals with asymptomatic genital herpes shed
virus frequently in the absence of lesions, all HSV-2 seropositive individuals are probably at risk for transmitting
infection in setting of sexual activity, labor and delivery,
and intimate contact.
NEONATAL HERPES
For the neonatologist, the most important category of
HSV-associated disease is that of neonatal herpes. In
the United States, the reported range of neonatal herpes
ranges from 1 in 2500 births to 1 in 8000 births (Corey and
Wald, 2009; Kimberlin, 2005; Whitley, 2004). Approximately 70% to 85% of neonatal herpes simplex infections
are caused by HSV-2 (Kimberlin et al, 2001). The majority
of cases occur in infants born to women who were recently
471
infected, rather than to women with histories of recurrent
genital herpes. Primary infection late in pregnancy poses
a higher risk of transmission to the infant than do primary
infections occurring before or early in pregnancy, suggesting that the evolution of a maternal antibody response
confers some measure of protection for the infant (Brown
et al, 2003; Caviness et al, 2008a). Primary genital herpes
infection in a pregnant mother results in an attack rate of
33% to 50% for her infant, whereas recurrent maternal
infection results in a 1% to 3% attack rate (Arvin, 1991;
Brown et al, 1997; Prober et al, 1987). Overall, it is estimated that approximately 22% of pregnant women have
genital herpes (HSV-2 seropositivity), and 2% of women
will acquire HSV during pregnancy (Brown et al, 2005).
Approximately 90% of these women are undiagnosed,
because they are asymptomatic or have other subtle symptoms that are incorrectly attributed to other vulvovaginal
disorders.
Strategies for preventing neonatal HSV infection can
be optimized by identifying women with genital lesions at
the time of labor for cesarean delivery, prescribing antiviral suppressive therapy as appropriate (see Treatment and
Outcomes, later), and minimizing invasive intrapartum
procedures for women in whom the diagnosis of genital
herpes cannot be excluded. Rarely, cases of intrauterine
infection have been described; these are often associated
with overwhelming primary maternal infection and usually result in fetal demise. On occasion, placentitis is also
observed (Baldwin and Whitley, 1989; Chatterjee et al,
2001; Florman et al, 1973; Hutto et al, 1987; Vasileiadis
et al, 2003). Although intrauterine transmission is possible, the vast majority of HSV infections in newborns are
acquired intrapartum, related to the presence of virus in
the maternal genital track. Neonatal acquisition of HSV
from an individual other than the mother, such as from a
recurrent oropharyngeal lesion, is unusual (Light, 1979;
Linnemann et al, 1978; Yeager et al, 1983). Neonatal HSV
has been described as a complication of ritual circumcision (Rubin and Lanzkowsky, 2000). Approximately 85%
of infants with neonatal HSV acquire infection from the
maternal genital tract at the time of delivery, but only
15% to 30% of mothers who give birth to infants with
neonatal HSV infection have a known history of genital
HSV (Whitley, 1988). Intrapartum interventions that have
the risk of penetrating fetal skin, such as scalp electrode
monitoring, increase the risk of transmission to the infant
(Golden et al, 1977; Parvey and Ch’ien, 1980).
Neonatal herpes can have devastating long-term consequences, making early recognition of paramount importance. Most infants with perinatal or postnatal HSV
infection are normal at birth. Illness typically develops
after 3 days of age; therefore the presence of skin lesions,
oral ulcers, and other signs and symptoms in the first 72
hours of life should suggest diagnoses other than HSV.
Premature infants appear to be at greater risk, possibly
because of reduced transplacental transfer of protective
antibody. Approximately 40% to 50% of affected infants
are less than 36 weeks in gestational age (Whitley, 1988).
Although a history of maternal cervical, vaginal, or labial
lesions should be sought when neonatal HSV is being considered in the differential diagnosis, overt herpetic disease
in the maternal genital tract is evident in approximately
472
PART IX Immunology and Infections
• Sepsis syndrome
• Hepatitis, DIC, pneumoia
• Skin vesicles may be absent
• High mortality even with therapy
• Survivors may be free of
CNS disease
• Disease limited to skin,
eyes, mucous membranes
• May progress to CNS or
disseminated disease
• Frequent cutaneous
recurrence in first year of
life – increased risk of
poor neurodevelopmental
outcome
Disseminated
disease
SEM disease
CNS disease
• Skin vesicles are absent
in ~ of cases
• Severe seizures common
• High incidence of CNS
imaging abnormalities
and neurologic sequelae
FIGURE 37-2 Characteristic presentations of neonatal herpes
simplex virus (HSV) infection. Approximately 45% of neonatal HSV
manifests as skin, eye, or mucous membrane disease (SEM disease); 25%
as disseminated disease; and 30%, central nervous systems (CNS) disease. Characteristic features of each subtype of neonatal HSV are listed
(arrows). Disease may span categories; for example, infants with SEM
disease may progress to disseminated or CNS disease, and infants with
CNS disease may develop skin vesicles later in hospital course, although
up to one-third of infants with CNS disease never have cutaneous
manifestations.
one third of patients (Overall, 1994). In the remaining two
thirds, infection is presumably via asymptomatic maternal
genital tract shedding of virus.
Infection can manifest in newborns in one of three
forms: disease limited to the skin, eye, or mucous membrane disease (SEM); disease involving the central nervous
system (CNS); or disseminated HSV infection, frequently
manifesting as a sepsislike syndrome, with pneumonia,
hepatitis, and viremia (Figure 37-2) (Kimberlin, 2005;
Whitley, 2004). There can be overlap in these syndromes;
for example, an infant with disseminated disease may initially have only skin lesions. The relative proportion of
infants with disseminated disease has been declining in
recent years, probably because earlier recognition and
treatment of SEM disease have resulted in more timely
intervention with antiviral therapy. Disseminated disease
usually begins toward the end of the 1st week of life. Skin
vesicles may be an early sign, but they are entirely absent
in almost half of patients. The scalp should be inspected
carefully, particularly near the site of insertion of fetal
scalp electrodes, because such lesions are easy to overlook.
Systemic symptoms, although initially insidious in onset,
progress rapidly. Poor feeding, lethargy, and fever may
be accompanied by irritability or seizures if the CNS is
involved. These symptoms are followed rapidly by jaundice, hypotension, disseminated intravascular coagulation,
apnea, and shock. This form of disease is indistinguishable
at its onset from both neonatal enterovirus infection and
bacterial sepsis. HSV infection should be considered in the
differential diagnosis of infants who have fever during the
first 2 weeks of life, because fever can herald the onset of
systemic disease. Localized disease may begin somewhat
later, with most cases appearing in the 2nd to 3rd weeks
of life. When the CNS is the primary site of infection, the
skin or eyes may be involved: up to one third of infants
with neonatal CNS disease will never have skin lesions
during their clinical course. The infants are lethargic, irritable, and tremulous, and seizures are common and difficult to control.
Other less common but potentially localized or disseminated findings are keratoconjunctivitis, chorioretinitis, and
pneumonitis, which can manifest as a focal infiltrate or as
diffuse bilateral disease. Supraglottitis, intracranial hemorrhage, aseptic meningitis, and fulminant liver failure have
been described (Abzug and Johnson, 2000; Erdem et al,
2002; Greenes et al, 1995; Kohl, 1994, 1999; Schlesinger
and Storch, 1994). Less common presentations of neonatal
herpes include hydrops fetalis (Anderson and Abzug, 1999)
and laryngitis (Vitale et al, 1993).
Diagnosis
The cornerstone of the diagnosis of neonatal HSV is virologic detection; HSV serology is of little use. HSV-1 and
HSV-2 are both easily recovered by culture of clinical
samples. In disseminated disease, virus is present in blood,
conjunctivae, respiratory secretions, and urine; it is also
present in the central nervous system in approximately half
of patients. In SEM disease, the virus can usually be found
at the site of disease (i.e., within a vesicle). Definitive microbiologic diagnosis requires growth of the virus in tissue culture, or detection of viral nucleic acid by PCR. HSV can
also be presumptively identified by immunofluorescence
of infected cells using HSV-specific antibodies; such tests
are commercially available and should be used in lieu of the
outdated and insensitive Tzanck smear. Viral culture has the
added advantage of allowing detection in the clinical virology laboratory of other agents that can mimic HSV disease
in neonates, such as enteroviruses (see Enteroviruses later),
because most diagnostic viral culture systems will support
the growth of a variety of pathogens. When neonatal herpes
is suggested, viral cultures of the throat, conjunctiva, blood,
stool or rectum, and urine should be obtained, as should
scrapings of vesicular, pustular, and ulcerative skin lesions.
Of these sites, skin and conjunctival HSV cultures have the
highest yield (Kimberlin et al, 2001).
All infants with presumed neonatal HSV disease should
undergo lumbar puncture, even if SEM disease is the only
observed clinical manifestation. In some infants, CNS
infection may be present but subclinical, and the finding
of HSV DNA in the cerebrospinal fluid (CSF) has important therapeutic and prognostic implications. Blood should
be sent for viral blood culture or PCR, because viremia is
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
a common finding in neonatal HSV infection (Diamond
et al, 1999; Stanberry et al, 1994). Usually if the CNS is
involved in the setting of neonatal HSV disease, evaluation of the CSF reveals a lymphocytosis, red blood cells,
normal or high protein level, and low or normal glucose
level. In addition to viral culture, CSF, blood, skin lesions,
and other specimens should be analyzed by PCR for the
presence of HSV genome (Ryan and Kinghorn, 2006).
Caution should be taken with interpretation of a negative
CSF PCR result: many infants with neonatal HSV disease will not have CNS involvement, so a negative CSF
PCR considered in isolation does not exclude the diagnosis of neonatal HSV. PCR of DNA extracted from the
dried newborn blood spot has been reported as a way to
retrospectively identify HSV infection in young infants
(Lewensohn-Fuchs et al, 2003). Some experts recommend
obtaining a second CSF specimen for evaluation at the end
of antiviral therapy, and it has been reported that persistence of HSV DNA may be a poor prognostic factor and
an indication for continuing antiviral therapy (Kimberlin
et al, 1996b; Malm and Forsgren, 1999; Mejías et al, 2009).
In disseminated disease, transaminase elevations consistent
with hepatocellular injury are typically present; in severe
disease, fulminant hepatitis with hepatic necrosis may be
observed. Infants with CNS disease should undergo neuroradiographic imaging with computed tomography (CT) or
magnetic resonance imaging (MRI). Early in the course of
illness, imaging may demonstrate nonspecific lack of graywhite matter junction differentiation and general signs of
encephalitis. Later CT findings include dilated ventricles,
parenchymal echogenicity, cystic degeneration, and intracranial calcifications (O’Reilly et al, 1995), whereas MRI
images demonstrate a variable appearance (Vossough
et al, 2008). Neonatal HSV-2 encephalitis can be multifocal or limited to only the temporal lobes, brainstem, or
cerebellum. Deep gray matter structures are involved and
hemorrhage is observed in more than half of patients. In
approximately 20% of patients, lesions are seen only by
diffusion-weighted imaging. In 40% of patients, watershed
distribution ischemic changes are also observed in addition to areas of presumed direct herpetic necrosis. In contrast to the classical description in older patients, neonatal
HSV encephalitis is not usually confined to the temporal
lobes. Electroencephalography should also be considered
in all infants with CNS involvement or with disseminated
disease, to evaluate for seizures. Electroencephalography
findings will be abnormal in approximately 80% of such
patients (Kimberlin et al, 2001).
Treatment and Outcomes
The cornerstone of the treatment of neonatal HSV disease
is the nucleoside analog known as acyclovir. The development of acyclovir in the 1980s was a watershed event in the
management of HSV infection. The use of acyclovir and
other antivirals is summarized in Table 37-2. Although
acyclovir had an efficacy similar to a previously used antiviral agent (vidarabine) in a controlled trial, acyclovir has
emerged as the drug of choice because of its greater ease
of administration and its highly favorable toxicity profile.
The current recommendation of the American Academy of
Pediatrics (AAP) Committee on Infectious Diseases is that
473
neonatal HSV infections should be treated with high-dose
acyclovir at a dose of 60 mg/kg/day intravenously, divided
into three doses given every 8 hours, for either 14 days
for infants with SEM disease or 21 days for infants with
disseminated or CNS disease. Support for this recommendation was derived from a comparison of the results from
an earlier study using standard-dose acyclovir (30 mg/kg/
day) for 10 days (Kimberlin et al, 2001). There is no role
for oral acyclovir in the management of neonatal HSV,
and no role for topical acyclovir in neonatal SEM disease.
Herpetic keratoconjunctivitis should receive topical ophthalmic antiviral therapy along with parenteral treatment.
In addition to antiviral therapy, appropriate supportive
care is essential, with anticipatory management targeting
complications of neonatal HSV disease such as seizures,
pneumonitis, and hepatic insufficiency. A beneficial role
of intravenous immunoglobulin (IVIG) has been inferred
from animal models of neonatal HSV disease, in which
passive antibody is clearly beneficial (Bravo et al, 1996),
but the absence of controlled studies precludes any recommendations in infants.
Even with timely institution of antiviral therapy, the
prognosis following neonatal HSV infection is guarded.
The mortality rates in infants with localized CNS disease
range from 4% to 14% with antiviral therapy, and most
survivors have long-term neurologic sequelae. Risk factors
for increased morbidity and mortality from CNS infection include prematurity and seizures upon initiation of
therapy (Kimberlin et al, 2001; Kohl, 1999). Infants with
disseminated disease have a high mortality rate without
antiviral therapy; 80% die, and most survivors have serious
neurologic sequelae (Whitley, 1988). Intravenous antiviral
therapy has decreased mortality of disseminated disease
to approximately 30%, and approximately 80% of surviving infants have a normal neurologic outcome (Corey and
Wald, 2009). Antiviral therapy has decreased the mortality
of neonatal CNS infection from approximately 50% to 6%,
but more than 70% of survivors have sequelae (Corey and
Wald, 2009; Engman et al, 2008; Freij and Sever, 1988).
Lethargy at initiation of therapy has been associated with a
higher mortality rate in neonates with disseminated HSV
infection (Kimberlin et al, 2001). Infants with skin involvement often have recurrent crops of skin vesicles for several
years. In an infant younger than 6 months, readmission to
the hospital for diagnostic evaluation and administration
of intravenous acyclovir is appropriate when cutaneous
recurrences are observed. It is postulated that recurrent
cutaneous lesions may be associated with subclinical CNS
reactivation, and it has been proposed that treatment of
these recurrences with acyclovir will improve long-term
outcome.
Management of asymptomatic neonates potentially
exposed to HSV in the birth canal is controversial. This
situation sometimes arises when a maternal perineal lesion
is discovered after vaginal delivery. Some experts recommend neonatal surface cultures and administration of
prophylactic antivirals, but there is little evidence to support this approach. The most important variable informing clinical management is probably the maternal history.
Term infants exposed to HSV in the birth canal and born
to women with long-standing histories of recurrent genital
herpes are at low risk (<1%) of infection and, if the infant is
474
PART IX Immunology and Infections
TABLE 37-2 Commonly Used Antiviral Agents in Neonatology Practice
Dose, Route of Administration,
Duration of Therapy
Antiviral Agent
Indication
Acyclovir
Neonatal HSV
60 mg/kg/day, dosing every 8 h IV; 21 days for
disseminated infection or CNS disease; 14
days for SEM disease
Comments
Monitor CBC twice weekly; adjust dosage for
renal insufficiency
Oral suppression
following neonatal
HSV
Efficacy for improving neurodevelopmental outcomes unproven; 10 to 20 mg/kg/dose twice
daily; duration of therapy, 12 months
Neutropenia observed in 1⁄2 to 2⁄3 of infants;
lesions while receiving suppressive therapy
should suggest acyclovir-resistant strain
VZV infection
15 mg/kg every 8 h for minimum of 5 to 7 days;
longer courses may be needed for severe
end-organ disease (pneumonia, hepatitis)
VZV postexposure
prophylaxis
10 mg/kg PO four times per day; treat for at
least 7 days (from 7 days after the earliest
exposure until 14 days after the last exposure)
Use in conjunction with VariZIG
Trifluridine 1%
HSV ophthalmic
disease
Apply as eye drops; 1 drop every 2 h to affected
cornea while awake; maximum 9 drops per day
Manage in consultation with ophthalmologist
Ganciclovir
Congenital CMV;
acquired CMV
12 mg/kg/day, dosing every 12 h IV; duration of
therapy is 6 weeks for prevention of hearing
loss; shorter courses of therapy (14 to 21 days)
are reasonable for serious end-organ disease
Efficacy against CMV associated hearing loss in
controlled trial; benefits of shorter courses of
therapy unknown; neutropenia observed in
63% of patients in controlled trial; adjust dose
for renal insufficiency; consider G-CSF if continued therapy desired in setting of neutropenia
Valganciclovir
Congenital CMV
32 mg/kg/day divided twice daily; no data available
for duration or efficacy of oral formulation;
CASG is currently performing a controlled
clinical trial of 6 months oral suppression
Valine ester (prodrug) of ganciclovir; similar
toxicity profile as GCV; long-term suppressive
therapy not well studied in infants; theoretical
concerns of carcinogenesis, gonadal toxicity
Lamivudine
Hepatitis B, HIV
For children 3 months to 16 years of age,
recommended dose is 4 mg/kg, up to 150 mg
per dose, twice daily
Chronic hepatitis B infection; also used for
HIV therapy
Interferon α2b
Hepatitis B,
hepatitis C
3 to 6 million international units/m2 3 times
weekly; up to 24 months duration; combined
with oral ribavirin for hepatitis C
Chronic hepatitis B infection; no data in
neonates; chronic hepatitis C infection when
administered with ribavirin; systemic side
effects (fever, flu-like symptoms, anorexia);
leucopenia; thyroid autoantibodies
Pegylated interferon
α2b
Hepatitis B,
hepatitis C
1.5 μg/kg once per week; no information on
dosing in children <2 yr old
Chronic hepatitis B; administer in conjunction
with ribavirin for hepatitis C; systemic side
effects (fever, flulike symptoms, anorexia);
leucopenia; thyroid autoantibodies; side effects
less common with pegylated formulations
Adefovir
Hepatitis B
Not recommended in children <10 years of age;
0.3 mg/kg once daily PO in children 2 to 6 yr
old has favorable pharmacokinetics
Chronic hepatitis B infection; no safety or
efficacy data in infants or young children
Ribavirin (oral)
Hepatitis C
15 mg/kg/day PO; no information on dosing in
children <2 years of age
Hemolytic anemia; teratogenic in animal
models
Ribavirin (aerosol)
Respiratory syncytial
virus
Standard ribavirin aerosol therapy is 6 g per
300 mL water for 18 h daily; short-duration
therapy, 6 g per 100 mL water given for a
period of 2 h 3 times per day
Not indicated for use with mechanical
ventilator; conjunctivitis; bronchospasm
Amantadine
Influenza A
FDA-approved dosage for children 1 to 9 yr
old for treatment and prophylaxis is 4.4 to
8.8 mg/kg/day, not to exceed 150 mg/day in
children <9 yr old
Not approved for use in children <1 yr old;
influenza B resistant; influenza A; H1N1
resistant
Rimantidine
Influenza A
Administered in one or two divided doses at a
dosage of 5 mg/kg/day, not to exceed 150 mg/
day in children <9 yr old
Not approved for use in children <1 yr old;
influenza B resistant; influenza A H1N1
resistant
Oseltamivir
Influenza A,
influenza B
For children <15 kg, recommended dose is 30
mg PO twice daily for 5 days (treatment) or
10 days (prophylaxis)
Not typically recommended in children <1 yr
old; FDA-approved guidelines for emergency use of oseltamivir in pediatric patients
younger than 1 year in 2009 in response to
H1N1 influenza pandemic
Zanamivir
Influenza A, influenza B
Recommended dose of zanamivir for treatment
of influenza is two inhalations (one 5-mg blister per inhalation for a total dose of 10 mg)
twice daily (approximately 12 h apart)
Not recommended in children <5 yr old
CASG, Collaborative Antiviral Study Group; CBC, complete blood cell count; CNS, central nervous system; FDA, U.S. Food and Drug Administration; G-CSF, granulocyte colonystimulating factor; HSV, herpes simplex virus; IV, intravenous; PO, by mouth; SEM, skin, eye, or mucous membranes; VariZIG, varicella-zoster immunoglobulin; VZV, varicellazoster virus.
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
asymptomatic, observation without antiviral therapy is sufficient. If the maternal HSV history is unclear, the infant
is premature, or other obstetric complications or risk factors are present (e.g., prolonged rupture of membranes,
maternal fever, signs or symptoms of chorioamnionitis,
fetal scalp electrode monitoring), then empiric antiviral
therapy is warranted. After the appropriate specimens are
collected and sent for viral culture and PCR analysis, the
infant should receive parenteral acyclovir (60 mg/kg/day)
pending the results of diagnostic virology studies. There
is no role for topical or oral formulations of acyclovir in
this setting.
Similarly controversial is the question of whether empiric
acyclovir therapy should be administered to neonates who
are readmitted for evaluation of febrile illness in the first
30 days of life. Standard practice in most children’s hospitals is to rule out sepsis in this setting by administering
broad-spectrum antibiotics pending the result of diagnostic cultures of blood, urine, and spinal fluid. Some experts
recommend the use of empiric acyclovir in this setting
(Long, 2008), citing data that indicate that the prevalence
of neonatal HSV infection is similar to that of invasive bacterial infection in this setting, and that infants may exhibit
fever and sepsis-like syndrome as the only manifestations
of HSV infection (Caviness et al, 2008b). Other experts
recommend a more selective approach, based on analysis
of history, risk factors, and laboratory and radiographic
analyses, such as liver function tests and chest radiograph
(Kimberlin, 2008). Additional studies will be required
to ascertain whether acyclovir should be included in the
empiric antimicrobial regimen of all neonates who are
evaluated for fever in the first few weeks of life.
In addition to its critical role in the management of the
acute clinical syndromes of neonatal HSV infection, acyclovir may be of benefit when administered as long-term
suppressive therapy in the first 6 months of life. The Collaborative Antiviral Study Group (CASG) has reported
results of controlled studies evaluating long-term oral
suppressive acyclovir therapy after neonatal HSV aimed
at both prevention of recurrent skin lesions and neurologic sequelae. The observation driving this study is that
recurrent skin lesions (more than three episodes) within
the first 6 months of life predict an adverse neurologic
prognosis, possibly because recurrent skin lesions are a
surrogate marker for subclinical reactivation events in the
CNS (Whitley, 1991). Phase I and II trials demonstrated
fewer cutaneous recurrences in the treatment group, but
did not have enough subjects to analyze the effect on
neurologic outcomes (Kimberlin et al, 1996a). Whether
reducing cutaneous recurrences in the 1st year of life will
result in improved neurodevelopmental outcome remains
unknown (Gutierrez and Arvin, 2003). Given the favorable safety profile of acyclovir, the use of long-term suppression after an episode of neonatal herpes is probably an
appropriate management strategy, and it is likely to reduce
the need for repeat hospitalization when lesions reappear.
Any theoretical benefit of acyclovir must be considered
against the potential risk of neutropenia, which has been
reported in 20% to 50% of neonates receiving long-term
acyclovir therapy. Although readily reversible with discontinuation of drug, this concern necessitates frequent monitoring of the absolute neutrophil count. The emergence of
475
acyclovir-resistant HSV strains is also a potential concern
(Kimberlin et al, 1996a; Oram et al, 2000). That long-term
oral suppressive therapy may be beneficial for improving
long-term neurodevelopmental outcomes owing to neonatal HSV infection was suggested by a 2-year pilot study of
oral suppressive therapy in a cohort of 16 infants (Tiffany
et al, 2005). In this uncontrolled study, all children were
independently mobile, free of seizures, and had normal
vision and speech development at the time of final neurodevelopmental assessment.
Prospects for Prevention
Even with prompt recognition and therapy, the risk of
long-term morbidity with neonatal HSV infection, particularly neurodevelopmental morbidity, remains significant.
Ideally, prevention of neonatal HSV infection would be
the best approach to solving the problem of HSV-induced
long-term neurologic injury. Recent developments in the
approach to HSV infections among women of childbearing age include consideration of maternal screening programs and the use of antiviral agents in women at risk for
transmission of infection. The availability of reliable kits
that allow specific serodiagnosis of HSV-1 and HSV-2
infections has enabled the identification of asymptomatic
women with genital herpes who can be counseled appropriately during pregnancy (Sauerbrei and Wutzler, 2007).
Although routine seroscreening of pregnant women is not
currently considered a standard of care, screening in some
instances, including situations where high maternal anxiety
exists, seems justified. There is no evidence that routine
administration of suppressive antivirals during pregnancy
can improve outcomes or reduce disease in newborns
(Sheffield et al, 2006), although this approach has become
common in many obstetric practices (Hollier and Wendel,
2008). One benefit of suppressive antiviral therapy during
pregnancy for a woman with a history of genital herpes may
be a reduced likelihood of cesarean section, predicated on
the assumption that suppressive therapy will prevent reactivation of HSV (Brown et al, 2003; Hollier and Wendel,
2008). Strategies for preventing neonatal herpes can also
target prevention of intrapartum transmission by caesarean section. If a mother has active genital herpes simplex
infection at the time of delivery, and if the membranes are
either intact or have been ruptured for less than 4 hours,
both the AAP and the American College of Obstetricians
and Gynecologists (ACOG) recommend cesarean delivery. The role of intrapartum antivirals or immunoglobulin
has not been studied in this setting.
Even with careful histories and meticulous physical
examination during labor and delivery, many at-risk deliveries cannot be predicted or identified, because so many
HSV-2–seropositive women are asymptomatic, do not
know that they have genital herpes, and may unknowingly
shed virus at the time of delivery. Infants in whom HSV
infection is known or highly suspected should be put in
isolation with contact precautions, and skin lesions should
be covered. Finally, any health care provider with active
herpetic whitlow or other skin lesions should not have
direct patient care responsibilities for neonates.
Ultimately, prevention of neonatal HSV infection could
be conferred by the development of a vaccine. Several
476
PART IX Immunology and Infections
phase I, II, and III clinical trials are ongoing to investigate
the utility of various HSV vaccines to prevent genital infections (Stanberry and Rosenthal, 2005). A double-blind,
randomized trial of an HSV-2 glycoprotein-D-subunit
vaccine given with alum adjuvant and 3-O-deacylatedmonophosphoryl lipid A, was reported in a study of individuals whose regular sexual partners had a history of
genital herpes. The primary end point was the occurrence
of genital herpes disease. The vaccine was efficacious in
women who were seronegative for both HSV-1 and HSV2, but it was not efficacious in women who were seropositive for HSV-1 and seronegative for HSV-2 at base line.
The vaccine had no efficacy in men, regardless of serostatus (Stanberry et al, 2002). The basis for this discrepancy
remains unexplained, but may relate to anatomic differences in how primary infections are established in men and
women, or sex-related differences in immune response. It
remains to be studied what effect widespread HSV-2 vaccination might have on the incidence or severity of neonatal HSV infection.
VARICELLA-ZOSTER VIRUS
Varicella-zoster virus (VZV) is a member of the
α-herpesvirus subfamily of the Herpesviridae. Like the
related HSV-1 and HSV-2, VZV can infect neurons where
it can establish latent infection. Primary varicella infection, commonly known as chickenpox, usually results in a
fever and a characteristic vesicular exanthem. The illness
often includes other systemic symptoms, such as headache and malaise. Reactivation from latency, which can
occur decades after the primary VZV infection, is usually
referred to as zoster or shingles. Zoster is characterized by a
painful vesicular rash in a dermatomal distribution, and in
older patients can lead to postherpetic neuralgia.
The neonatologist may encounter consequences of
maternal VZV infection in two different clinical presentations. In congenital varicella, VZV is transmitted to the
fetus in the first or second trimester of pregnancy, where
it can produce a number of teratogenic consequences
(Auriti et al, 2009; Laforet and Lynch, 1947; Smith and
Arvin, 2009; Srabstein et al, 1974). In contrast, neonatal
varicella occurs in the setting of primary maternal varicella
acquired late in the third trimester, and the affected infant
can exhibit symptoms and signs in the neonatal period.
This section reviews both of these presentations of VZVrelated disease in infants.
Epidemiology of Maternal and Perinatal
Varicella-Zoster Virus
Before the advent of routine childhood vaccination against
chickenpox, it was estimated that the VZV seroprevalence in women of childbearing age was greater than 95%,
because of the formerly ubiquitous nature of this infection. In this era, primary VZV infections during pregnancy
occurred with a frequency of 5 to 7 per 10,000 pregnancies
in the United States (Balducci et al, 1992; Brunell, 1992).
The major concern in the setting of primary maternal
VZV infection is the risk for congenital varicella syndrome
(CVS). It is not yet clear whether the risk for CVS has
been ameliorated by the widespread use of VZV vaccine.
For pregnant women with primary varicella infection, the
transmission rate to the fetus is estimated to be approximately 25%. Only a subset of infected fetuses exhibit
symptomatic disease. Approximately 100 cases of CVS
have been reported in the literature (Sauerbrei and Wutzler, 2000). The risk of symptomatic intrauterine VZV
infection after maternal varicella occurring during the first
20 weeks of pregnancy is approximately 1% to 2% (Enders
et al, 1994; Paryani and Arvin, 1986; Pastuszak et al, 1994;
Siegel, 1973). Symptomatic disease appears to be more
common in female infants (Sauerbrei and Wutzler, 2000).
CVS does not appear to occur in the setting of maternal
zoster. A prospective study of infants born to 366 mothers
with a clinical history of zoster during pregnancy found no
infants with CVS (Enders et al, 1994).
Maternal infection in the third trimester is not associated with CVS, presumably because this falls outside
the time frame when teratogenicity can occur. Maternal
infection just before or after delivery poses a high risk for
neonatal varicella. Before the advent of VZV vaccination,
neonatal varicella was encountered much more commonly
that CVS. For infants in which maternal illness begins 5
days or less before delivery or up to 2 days after delivery,
the infant attack rate is 17% to 31% (Brunell, 1992; Feldman, 1986; Meyers, 1974). Because the incubation period
for varicella is between 10 and 21 days, cases beginning
in the first 10 days of life are considered to have been
acquired in utero.
Pathogenesis and Clinical Manifestations
As noted, CVS is acquired from a maternal primary varicella infection that occurs during the first or second trimester. The virus is thought to be transmitted transplacentally
during the viremia that precedes or accompanies the rash
of chickenpox. Ascending infection from cervical infection
has been proposed as a potential mechanism of transmission, but is probably much less common (Sauerbrei and
Wutzler, 2000). Some of the congenital malformations
associated with CVS may be a consequence of zosterlike
virus reactivation events in the infected fetus rather than
the direct effects of the primary viral infection. This explanation is supported by the common finding of unusual
cicatricial rashes in dermatomal distributions in the newborn with CVS. Other clinical manifestations include
asymmetric muscular atrophy with limb hypoplasia, low
birthweight, neurologic abnormalities (cortical or spinal
cord atrophy, seizures, microcephaly, encephalitis, Horner
syndrome), and ophthalmologic abnormalities (chorioretinitis, microphthalmia, atrophy, and cataracts; Brunell,
1992; Feldman, 1986; Sauerbrei and Wutzler, 2000). Gastrointestinal abnormalities are reported in 15% to 23% of
cases; findings include duodenal stenosis, dilated jejunum,
small left colon, intestinal atresia or bands, and hepatic calcifications (Alkalay et al, 1987; Jones et al, 1994). Immature fetal cell–mediated immune response may explain the
short latency period and the inadequate protection from
the consequences of episodes of reactivation in utero (Higa
et al, 1987; Kustermann et al, 1996). Pathology reports
have noted destruction of neural tissue with residual dystrophic calcifications, chronic active inflammation in nonneural tissues surrounding viral inclusions, and evidence of
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
chronic placental villitis (Bruder et al, 2000; Petignat et al,
2001; Qureshi and Jacques, 1996).
Maternal varicella near term or immediately postpartum
can lead to neonatal varicella. Neonatal varicella is usually
caused by maternal chickenpox acquired during the last 3
weeks of pregnancy. Infection may be transmitted perinatally by transplacental viremia (most cases) or by ascending
infection from the birth canal; it can also be treated postnatally by the aerosol route or direct contact with infectious lesions. Serious postnatal infection acquired from
maternal varicella via breastfeeding has not been reported.
Neonatal varicella may develop despite the administration of varicella-zoster immunoglobulin (VariZIG) to the
infant at birth. Transplacentally transmitted infections
occur in the first 10 to 12 days of life, whereas chickenpox
after that time is most likely acquired by postnatal infection. The clinical presentation differs markedly for cases
in which maternal rash began 5 days or more before delivery from those in which maternal illness occurred from 5
days before to 2 days after delivery. When maternal varicella is noted more than 5 days before delivery, neonatal
disease usually begins within the first 4 days of life and is
typically mild. In contrast, neonatal varicella in an infant
whose mother develops varicella from 5 days before until
2 days after delivery has a high risk of morbidity and mortality (Isaacs, 2000; Tan and Koren, 2006). In this second
group, neonatal disease typically begins between 5 and 10
days after delivery, and a fatal outcome has been reported
in 23% to 30% of cases (Brunell, 1966; Sauerbrei and
Wutzler, 2001). When the disease appears in this setting,
it closely resembles varicella in the immunodeficient or
immunosuppressed host. Recurrent crops of skin vesicles
develop over a prolonged period. Typical presenting signs
are fever, hemorrhagic rash, and visceral dissemination
with involvement of the liver, lung, and brain. Secondary
bacterial infection may occur.
Prospective studies of the long-term outcomes of CVS
have not been performed, and there is probably a wide
continuum of disease. Among the 96 infants reviewed
by Sauerbrei and Wutzler (2000), 14 (15%) had clinical
signs of zoster during early infancy; these infants had been
exposed to varicella between 8 and 24 weeks’ gestation.
Some experts consider early zoster infections as one criterion for diagnosis of CVS. Between 1% and 2% of infants
without clinical evidence of CVS, but whose mothers had
chickenpox in the second and third trimesters, develop
zoster in the first few weeks of life; if this occurs, consideration should be given to ophthalmologic evaluations to
rule out the possibility of CVS (Enders et al, 2004; Sauerbrei and Wutzler, 2000). Overall mortality rates for CVS
are estimated at 30%. Deaths occur in the first few months
of life, usually secondary to severe pulmonary disease (Pastuszak et al, 1994; Sauerbrei and Wutzler, 2000).
Diagnostic Studies
Prenatal diagnosis—by means of quantification of varicella-specific IgM on fetal blood obtained by cordocentesis or through PCR analysis of chorionic villi, fetal blood,
and amniotic fluid—has been attempted (Cuthbertson
et al, 1987; Kustermann et al, 1996; Mouly et al, 1997).
Although the yield of PCR on amniotic fluid or fetal
477
blood is higher than that of serologic analysis of fetal IgM
or viral cultures, large prospective studies of the correlation of such findings with clinical outcomes have not been
performed. Reported prenatal ultrasonographic findings
include polyhydramnios, hydrops, progressive intrauterine growth restriction, microcephaly, limb hypoplasia,
and liver hyperechogenicities (Petignat, 2001; Pretorius
et al, 1992). Abnormal ultrasonographic findings might
not develop in a fetus for at least 5 weeks after maternal
infection (Kerkering, 2001).
Serologic studies can be performed postnatally on infants
(Paryani and Arvin, 1986). Suspected congenital infection
with varicella can be confirmed by the finding of persistent
VZV IgG beyond the presumed duration of passive transfer
of maternal antibodies (at least 6 to 7 months). Detection
of fetal IgM can also confirm infection, but it is less useful
because approximately one fourth of infants reported with
classic CVS have positive VZV IgM titer values (Enders
et al, 1994; Sauerbrei and Wutzler, 2000). Scrapings of skin
lesions, as with herpes simplex infections, can show large
multinucleated cells when stained with Wright or Giemsa
stain (Tzanck smears), but this procedure is no longer
recommended, because highly sensitive direct fluorescent
antibody (DFA) tests are readily available and are more sensitive and specific (Chan et al, 2001; Coffin and Hodinka,
1995). VZV can also be detected from skin lesions by PCR
(Leung et al, 2010). Although VZV can be detected by DFA
staining of cells or PCR of DNA extracted from skin and
visceral lesions, these tests in infants with suspected congenital infection are often extremely low yield compared
with infants and children who have acquired varicella infection. Neuroradiographic demonstration of intracranial calcifications has been reported with CVS, but this is not a
common finding (Kerkering, 2001).
Treatment
No controlled studies examining the effect of antiviral
therapy to prevent or treat congenital varicella syndrome
have been conducted. Some experts have recommended
oral acyclovir for pregnant women with varicella, especially during the second and third trimesters (AAP
Committee on Infectious Diseases, 2010a). Intravenous
treatment with acyclovir for the infected pregnant woman
is recommended for patients with serious complications of
varicella, particularly pneumonia. Anecdotal observations
suggest a potential effect of acyclovir on the progression of
eye disease in CVS (Sauerbrei and Wutzler, 2000). However, there are no recommendations for the use of acyclovir or immunoglobulin for treatment or prevention of
CVS. For neonatal varicella, treatment with intravenous
acyclovir, 60 mg/kg/day divided into doses every 8 hours,
is recommended, particularly for infants at the highest risk
of adverse outcomes (i.e., the infant born to a woman who
develops varicella from 5 days before until 2 days after
delivery; see Table 37-2).
Prevention
If a susceptible pregnant woman has a significant exposure to varicella, administration of VariZIG to her and
her newborn infant should be considered seriously. These
478
PART IX Immunology and Infections
BOX 37-3 C
andidates for VariZIG* after
Significant Exposure† to VaricellaZoster Infection in Perinatology
and Neonatology Practice
ll
ll
ll
ll
Pregnant women without evidence of immunity
Newborn infant whose mother developes chickenpox within 5 days before
delivery or within 48 hours after delivery
Hospitalized premature infant (≥28 weeks’ gestational age) whose mother
has no history for chickenpox or serologic evidence of prior infection
Hospitalized preterm infants (<28 weeks’ gestational age or birthweight
≤1000 g) regardless of the maternal history of varicella or varicella-zoster
virus serostatus
*Varicella-zoster immunoglobulin (VariZIG) is given intramuscularly at a recommended dose
of 125 units per 10 kg body weight, up to a maximum of 625 units. If VariZIG is unavailable,
immunoglobulin (400 mg/kg, intravenously) can be substituted.
†Significant exposures include: household exposure; face-to-face indoor play; face-to-face
contact in hospital setting with infectious staff member, patient, or visitor; intimate contact
(hugging or touching) with patient with active zoster lesions.
recommendations are summarized in Box 37-3. Infants of
mothers in whom varicella develops from 5 days before
to 2 days after delivery should receive VZIG as soon as
possible (AAP Committee on Infectious Diseases, 2010a).
VariZIG is given intramuscularly at a recommended dose
of 125 units per 10 kg bodyweight, up to a maximum of
625 units. If VariZIG is unavailable, IVIG (400 mg/kg)
can be substituted. Passive immunoprophylaxis has been
shown to prevent chickenpox in exposed older children
(Brunell et al, 1969), but does not always prevent neonatal disease (Reynolds et al, 1999). Approximately 50%
of exposed infants treated with immunoglobulin can still
develop varicella, but the disease is often attenuated, and
approximately 10% have severe disease (Hanngren et al,
1985). For healthy term infants exposed postnatally to varicella, including infants whose mother’s rash began more
than 48 hours after delivery, VariZIG is not generally indicated. VariZIG is not indicated for an infant whose mother
has zoster. Breastfeeding is not contraindicated.
Follow-up of infants exposed to VZV and treated with
VariZIG can include consideration of serologic testing
(enzyme immunoassay, latex agglutination, or indirect fluorescent antibody staining for IgG) to determine whether
asymptomatic infection has elicited immune protection.
Some experts recommend repeated administration of
VariZIG after a repeated exposure of an infant in whom
varicella did not develop more than 3 weeks after administration of the initial dose of VariZIG, although the risk to
these infants is less well defined. Infants receiving VariZIG
should also be placed in respiratory isolation for 28 days
or until discharge, because administration of VariZIG can
prolong the incubation period.
In the event of a significant varicella exposure in a
nursery situation, infants whose mothers have no history of chickenpox and who have undetectable antivaricella antibody titers should be considered candidates for
VariZIG. All exposed infants less than 28 weeks of gestational age or birthweight less than 1000 g, regardless of
maternal history, should receive VariZIG (see Box 37-3)
(AAP Committee on Infectious Diseases, 2010a). The
decision to use VariZIG in the premature infant greater
than 28 weeks’ gestation should be predicated on maternal
history of chickenpox or serologic evidence of protection.
As of late 2010, VariZIG is available under an investigational new drug protocol and can be obtained by calling FFF Enterprises (800-843-7477; www.fffenterprises.
com/Products/VariZIG.aspx).
Acyclovir has also been recommended by some experts
for postexposure prophylaxis in the setting of a nursery
outbreak of VZV (Hayakawa et al, 2003; Shinjoh and
Takahashi, 2009). A suggested dose is 10 mg/kg by mouth,
four times per day for 7 days. All exposed health care professionals without evidence of immunity should be excused
from patient contact from day 8 to 21 after exposure to an
infectious patient, or to day 28 if the individual has received
VariZIG (AAP Committee on Infectious Diseases, 2010a).
The best means of prevention is to follow current recommendations for universal varicella immunization of all
children at 12 to 15 months of age, as well as vaccinating all susceptible adolescents and adults at high risk of
exposure to varicella. VZV vaccine is available in three
formulations: a monovalent formulation for children and
young adults, a combination vaccine given with the measles-mumps-rubella vaccine in children and young adults,
and a monovalent formulation for adults older than 60
years for prevention of herpes zoster (shingles). There is
no evidence that CVS occurs after exposure to varicella
vaccine during pregnancy. From March 17, 1995, through
March 16, 2005, 981 women were enrolled in a pregnancy
registry for women exposed to varicella vaccine (Shields
et al, 2001; Wilson et al, 2008). Pregnancy outcomes were
available for 629 prospectively enrolled women. Among
the 131 live births to VZV-seronegative women, there was
no evidence of congenital varicella syndrome. Nonetheless
it is recommended that adolescents and women of childbearing age should avoid pregnancy for at least 1 month
after immunization.
CYTOMEGALOVIRUS
CMV infection is ubiquitous in the general population and
generally produces few if any symptoms in the immunocompetent infant, child, or adult. The mild nature of primary infection in most persons belies the severe nature of
CMV-induced illness in those with impaired, suppressed,
or immature immune systems, including infected newborns. Among the perinatally acquired viral infections
in the developed world, CMV imposes the largest economic burden and produces the greatest long-term neurodevelopmental morbidity. This section focuses on the
epidemiology, pathogenesis, diagnosis, and therapeutic
management of maternal, congenital, and perinatal CMV
infections.
Epidemiology
In retrospect, the first description of congenital CMV disease was in 1904, when Ribbert observed the large inclusion-bearing cells that represent the typical histopathologic
finding of CMV end-organ disease in a stillborn infant.
In 1920 a viral cause was proposed for the “cytomegaly”
seen in tissue sections of these inclusion-bearing cells
(Goodpasture and Talbot, 1921), and it would be several
more decades before the ubiquitous nature of this virus
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
Recurrent infection
0.5% to 3%
transmission
rate
Primary infection
Congenital
cytomegalovirus infection
85% to 90%
asymptomatic
at birth
30% to 50%
transmission
rate
10% to 15%
symptomatic
at birth
Asymptomatic
Symptomatic
10% to 15% of
asymptomatic
infants have
sequelae
60% to 90% of
symptomatic
infants have
sequelae
Sequelae
• Sensorineural hearing loss
• Mental retardation
• Seizure disorders
• Cerebral palsy
• Visual deficits
• Developmental delay
FIGURE 37-3 Profiles of congenital cytomegalovirus epidemiology, infection, and outcome. Transmission rates to the fetus are highest in the setting of primary maternal infection (up to 50%), although
0.5% to 3% of women with preconception immunity may nonetheless
transmit cytomegalovirus (CMV) because of reinfection or reactivation
of latent infection. Among all infants with congenital CMV infection,
regardless of maternal immune status during pregnancy, approximately
10% to 15% have symptoms or signs at birth (e.g., microcephaly, chorioretinitis, hepatosplenomegaly, petechiae, purpura, thrombocytopenia,
hepatitis, seizures, pneumonitis). Symptomatic infants have the highest
risk of neurodevelopmental sequelae, although any infant with congenital CMV is potentially at risk for sequelae. Among asymptomatic congenitally infected infants with sequelae, the most common manifestation
is sensorineural hearing loss, which may not be present at birth.
and the depth and breadth of the pathology it produces
would be elucidated. In the developed world, CMV transmission occurs in 0.5% to 2% of all live births, making
it the most common congenital viral infection (Demmler,
1991). Approximately 40,000 infants are born with CMV
infection every year in the United States. Consequently,
congenital CMV has a much greater impact than other
more commonly recognized causes of birth defects in
newborns. More than 8000 children per year will be permanently disabled by congenital CMV infection (Figure
37-3). This number is higher than those affected by other,
better known childhood conditions such as Down syndrome, fetal alcohol syndrome, and spina bifida (Ross
et al, 2006). Rates of congenital CMV infection tend to parallel those of maternal seropositivity and vary substantially
across populations (Mustakangas et al, 2000). Although
the lifetime risk of acquiring CMV infection is high,
approaching 90% by the eighth decade of life (Staras et al,
2006), seroprevalence is substantially lower among women
of childbearing age. Seronegative women are therefore at
risk for acquiring primary infections during pregnancy.
Primary infections pose an increased risk of transmission
to the fetus, and possibly a higher risk of sequelae. Every
year in the United States, approximately 27,000 seronegative women acquire a primary CMV infection (Colugnati
et al, 2007).
479
Seroprevalence rates for CMV vary significantly globally and are generally inversely correlated with socioeconomic status. CMV seroprevalence is greater in childhood
in developing countries. In developed countries, seroprevalence tends to be higher in blacks and Hispanics,
low-income groups, and persons without higher education. Young maternal age, single marital status, and
non-Caucasian race are associated with higher rates of
congenital CMV infection. Women with increased occupational exposure to young children (including daycare
providers) appear to be at elevated risk for primary CMV
infections (Adler, 1989; Pass et al, 1986, 1990; Stagno and
Britt, 2006; Stagno and Cloud, 1994). Health care providers in contrast are not at increased risk for acquisition of a
primary CMV infection (Dworsky et al, 1983).
Pathogenesis
Morphologically and at the genome level, CMV is the
largest pathogen (Schleiss, in press). There are approximately 160 known CMV genes; although based on its coding potential, CMV has the potential to encode more than
250 open reading frames. The mechanisms by which CMV
injures the fetus are complex and involve a complex interplay of viral gene products, maternal immune response,
and placental biology. CMV encodes genes modifying the
cell cycle, cellular apoptosis mechanisms, inflammatory
responses, and evasion of host immune responses. The
pathogenesis of fetal injury in the setting of congenital
CMV infection is incompletely understood. The pathogenesis of disease associated with acute CMV infection has
been attributed to lytic virus replication, with end-organ
damage occurring either secondary to virus-mediated cell
death or from pathologic host immune responses targeting virus-infected cells (Britt, 2008; Schleiss, in press).
Factors that contribute to fetal injury include the timing of infection relative to the gestational age of the fetus
(Pass et al, 2006), the maternal immune status (Fowler
et al, 1992), the extent of associated placental injury (Fisher
et al, 2000), the magnitude of the viral load in the amniotic fluid (Lazzarotto et al, 2000), the induction of host
genes occurring in response to infection (Challacombe
et al, 2004), and possibly the genotype of the particular strain
of CMV infecting the fetus (Arav-Boger et al, 2006). The
relative contribution of maternal, placental, and fetal compartments in the pathogenesis of disease remains incompletely defined. Much of the injury that CMV produces in
the newborn may be caused by placental insufficiency and
not by viral infection of the fetus (Schleiss, 2006a). Delivery of oxygen, substrate, and nutritional factors to the fetus
is impaired for a CMV-infected placenta. Moreover, CMV
infection of the placenta might contribute to intrauterine
growth restriction and fetal injury via induction of proinflammatory cytokines and modulation of normal trophoblast gene expression (Chan and Guilbert, 2005; Chou
et al, 2006; La Torre et al, 2006; Maidji et al, 2007; Yamamoto-Tabata et al, 2004). Identified as a site for major
pathogenic virus-induced injury, the placenta is increasingly considered to be a target for novel therapeutic interventions, such as CMV hyperimmune globulin for women
with primary CMV infection during pregnancy (La Torre
et al, 2006).
480
PART IX Immunology and Infections
Congenital CMV infection appears to be pathologically
more severe in the setting of primary maternal infection,
which confers a 40% to 50% risk of intrauterine transmission during gestation, versus the 0.5% to 2% transmission risk in women with preconceptional immunity
(Fowler et al, 2003). Congenital CMV infection in previously immune mothers appears to be related to reinfection with new strains of virus, with variations in epitopes of
virally encoded proteins that may correlate with decreased
maternal immune (Boppana et al, 1999, 2001). Many of
the pathologic manifestations of congenital CMV infection that reflect visceral organ involvement (hepatitis and
pneumonitis) are also observed in immunocompromised
adults with disseminated CMV disease. The developing fetal brain is also highly susceptible to CMV-induced
injury. CNS injury may be attributable to interference
with important cellular functions of the stem and progenitor cells in the brain. The pathogenesis of CMV infection
in the CNS seems to be strongly related to perturbations
in neural migration, neural death, cellular compositions,
and the immune system of the brain (Cheeran et al, 2009).
In infants with severe symptomatic congenital CMV infection, histopathologic evidence of viral dissemination is
commonly found in the brain, ear structures, retina, liver,
lung, kidney, and endocrine glands (Bissinger et al, 2002).
Distinctive features include large cells with large nuclei
containing oval inclusions separated from the nuclear
membrane by a clear zone, which gives them a characteristic “owl’s eye” appearance. Inclusion-bearing epithelial
cells have been described in the semicircular canals, vestibular membrane, cochlea, and other structures of the ear.
In addition, temporal bone anomalies with cochlear, vestibular, and auditory canal defects have been noted in association with hearing loss in affected infants (Davis, 1969;
Myers and Stool, 1968).
Clinical Presentation, Sequelae,
and Prognosis
Prenatal ultrasonography provides clues to the possible
diagnosis of fetal CMV infection. Findings include intrauterine growth restriction, microcephaly, ventriculomegaly, periventricular calcifications, echogenic bowel,
polyhydramnios, pleural effusion, pericardial effusion,
hepatosplenomegaly, intrahepatic calcifications, pseudomeconium ileus, and placental enlargement (Guerra et al,
2008; Nelson and Demmler, 1997). Fetal hydrops is also a
common finding (Sampath et al, 2005). However, the sensitivity of ultrasound to detect congenital CMV infection is
poor given that the majority of congenitally infected infants
are asymptomatic. In a study of 600 pregnant women with
primary CMV infection, abnormal ultrasound findings
were detected in 51 of 600 (8.5%) pregnancies and in 23
of 154 (14.9%) fetuses in which congenital infection was
documented. The positive predictive value of an abnormal
ultrasound finding that predicted symptomatic congenital
infection in women with primary CMV infection was only
35.3% when fetal infection status was unknown, compared
with 78.3% when congenital CMV infection was confirmed (Guerra et al, 2000, 2008).
Signs and symptoms are apparent at birth in 10% to
15% of all children with congenital CMV infection. Table
37-1 outlines the clinical manifestations of symptomatic
CMV infection. Each year in the United States, approximately 40,000 babies are born with congenital infection;
of these, 4000 to 6000 have symptomatic disease (see Figure 37-3) (Sharon and Schleiss, 2007). Infection in the
symptomatic infant can involve any organ and manifests
along a spectrum from mild illness to severe disseminated
multiorgan system disease. The mortality rate of symptomatic congenital CMV disease in the 1st year of life is
estimated to be greater than 10% (Boppana et al, 1992).
Clinical features include jaundice, hepatosplenomegaly,
lethargy, respiratory distress, seizures, and petechial rash.
Infants with symptomatic disease are often premature and
small for gestational age. A wide spectrum of disease can
be observed, including hemolysis, bone marrow suppression, hepatitis, pneumonitis, enteritis, and nephritis. Bone
abnormalities have been described (Alessandri et al, 1995).
Common laboratory abnormalities include thrombocytopenia, anemia, abnormal levels of liver enzymes (particularly elevated transaminases), and elevated conjugated
bilirubin levels. End-organ involvement is often inferred
on a clinical basis; it can be confirmed by isolation of the
virus or viral DNA from tissue or by the histopathologic
demonstration of characteristic inclusion bodies, positive
in situ hybridization with CMV gene probes, or immunofluorescence using appropriate CMV-specific antibodies
from tissue biopsies.
Of particular concern are the CNS pathologies observed
with symptomatic congenital CMV infection. These
pathologies include meningoencephalitis, calcifications,
microcephaly, neuronal migration disturbances, germinal
matrix cysts, ventriculomegaly, and cerebellar hypoplasia
(Cheeran et al, 2009). CNS disease is usually characterized
by at least one of the following signs and symptoms: lethargy, microcephaly, intracranial calcifications, hypotonia,
seizures, hearing deficit, or an abnormal eye examination
finding, such as chorioretinitis or optic atrophy. The clinical finding of microcephaly implicates the CNS as a site
of infection with CMV, and abnormal findings on cranial
imaging studies corroborate its involvement (Figure 37-4).
Long-term neurodevelopmental disabilities are observed
in 50% to 90% of children who are symptomatic at birth.
In contrast, long-term neurodevelopment injury is strikingly less likely in congenitally infected infants who are
asymptomatic at birth: when it does occur, it is typically
limited to hearing deficits. If there is a control for sensorineural hearing loss (SNHL), the intellectual development
of asymptomatic congenitally infected infants appears to
be normal (Conboy et al, 1986, 1987; Stagno et al, 1982b).
Among symptomatic congenitally infected infants, longterm sequelae can include microcephaly, hearing loss,
motor deficits (paresis or paralysis), cerebral palsy, mental
retardation, seizures, ocular abnormalities (chorioretinitis, optic atrophy), and learning disabilities (Cheeran et al,
2009; Sharon and Schleiss, 2007).
The incidence of hearing loss among children with congenital CMV infection ranges from 10% to 15% of those
who are asymptomatic at birth to up to 60% of those who
are symptomatic as newborns (Pass, 2005). SNHL can
be progressive and fluctuating in both asymptomatic and
symptomatically congenitally infected infants (Rosenthal
et al, 2009). Among asymptomatic congenitally infected
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
A
B
C
D
481
FIGURE 37-4 Magnetic resonance image abnormalities in infants with congenital cytomegalovirus (CMV) infection with neurologic
manifestations. A, T1 axial flair image of neonate with congenital CMV diagnosed with CNS malformation in utero by prenatal ultrasound, demonstrating severe hydrocephalus and cortical dysplasia and polymicrogyria. B, Axial T1 image of infant with symptomatic congenital CMV infection
demonstrating ventriculomegaly and periventricular enhancement (arrow). C and D, T1 flash axial (C) and sagittal (D) images of a symptomatic,
congenitally infected infant demonstrating ventriculomegaly, polymicrogyria, and porencephalic cyst (arrow).
infants, SNHL tends to be high frequency in nature; it
ranges in severity from a unilateral, mild hearing deficit
to severe, bilateral, profound deafness. CMV-induced
SNHL is a dynamic, evolving lesion; it may be present
at birth or can appear later in childhood. Delayed-onset
hearing loss usually occurs before 4 years of age (Dahle et
al, 2000; Fowler et al, 1997; Rivera et al, 2002; Williamson
et al, 1992), but has been reported to evolve and progress
through 6 years of age and beyond. The pathogenesis of
the SNHL is incompletely understood. SNHL appears to
be related to an inflammatory labyrinthitis, possibly potentiated by the fact that the labyrinth is a site of immune
privilege, allowing virus to persist in this compartment
long after it has been cleared from the systemic compartment (Schleiss and Choo, 2006). Although some temporal
bone and cochlear abnormalities have been described in
a small case series of hearing-impaired infants with congenital CMV infection (Bauman et al, 1994), a more comprehensive analysis indicated that CMV-associated SNHL
was not associated with structural abnormalities, such as
enlarged vestibular aqueduct syndrome (Pryor et al, 2005).
Logistic regression analysis of the clinical and laboratory
parameters observed in 180 children with symptomatic
congenital CMV infection showed that the presence of
petechiae and intrauterine growth restriction were independently associated with the development of hearing loss
(Rivera et al, 2002). A retrospective review of symptomatic congenitally infected infants identified neurologic and
radiologic sequelae in 81% of affected patients. There
was a significant correlation between the severity of the
initial pure-tone audiometry and the development of progressive hearing loss, in addition to a significant correlation between a less severe final pure-tone audiometry and
the presence of cerebral palsy (Madden et al, 2005). All
congenitally infected infants, regardless of the results of
functional hearing assessment at birth, should be monitored prospectively for SNHL by an audiologist using ageappropriate diagnostic tools. For severe SNHL, cochlear
implantation has been used with success (Lee et al, 2005;
Yoshida et al, 2009).
Diagnosis and Infant Assessment
Congenital CMV infection is best diagnosed by detection
of virus, either through culture techniques or via PCR, in
samples collected within the first 2 to 3 weeks of life. The
timing of these samples is important because subsequent
viral isolation may represent neonatal infection acquired
in the birth canal or after exposure to breast milk (Schleiss,
2006b). Urine and saliva are the clinical samples of choice
for viral culture. Specimens are typically inoculated onto
a monolayer of human fibroblasts. The high viral titer in
482
PART IX Immunology and Infections
these samples usually allows virus detection by cytopathic
effect within 1 to 3 days; however, 2 to 4 weeks may be
required (Revello and Gerna, 2002). Rapid virus isolation
can be achieved using monoclonal antibodies to CMVspecific early protein with low-speed centrifugation of the
clinical isolate onto the monolayer of fibroblasts growing
on cover slips inside shell vials. This “shell vial” assay has a
high sensitivity and specificity, and it allows the confirmation of diagnosis within 24 hours of inoculation (Gleaves
et al, 1984; Rabella and Drew, 1990).
PCR can readily amplify CMV DNA from various clinical samples, including urine, cerebrospinal fluid, blood,
plasma, saliva, and biopsy material (Revello and Gerna,
2002). If primer selection and amplification conditions
are carefully chosen, PCR can yield results comparable
to a standard tissue culture test. Obvious advantages of
PCR over culture are the small sample requirement, the
short time requirement for test results, and the ability to
use frozen specimens for diagnosis. The magnitude of the
systemic viral load in the congenitally infected infant can
be a predictor of neurodevelopmental prognosis. In some
studies, the magnitude of viral DNA in blood (DNAemia)
assessed by quantitative PCR correlated with an increased
risk of long-term sequelae, including SNHL (Lanari et
al, 2006). Quantitative PCR assays, particularly of blood,
should be ordered in the evaluation of congenitally infected
infants and may be useful in making decisions about which
infants to treat with antiviral therapy.
Serodiagnosis of congenital CMV is problematic. In
congenital CMV infection, antibody production by the
infected fetus begins in utero; however, serodiagnosis of
congenital infection is complicated by the presence of
maternal IgG antibodies that cross the placenta. Although
a negative antibody titer in infant and maternal sera provides sufficient evidence to exclude the diagnosis of congenital CMV infection, positive titers in the newborn by
no means confirm congenital infection. The presence
of IgM antibodies to CMV in cord or neonatal blood is
highly specific and in principle represents fetal antibody
response, but many IgM commercial detection assays
cross-react with IgG antibodies. Therefore a positive IgM
titer has limited sensitivity in diagnosing congenital CMV
infection and should not take the place of viral culture or
PCR (Revello et al, 1999b).
A recent development in molecular diagnostics for congenital CMV infection has been the use of dried blood
spots (DBSs) as a source of CMV DNA for PCR-based
detection (Barbi et al, 2006; Scanga et al, 2006; Yamagishi
et al, 2006). DBS screening is amenable to long-term storage, so diagnosis can be made retrospectively, even after
several years. The test might be useful in the future for
implementation of widespread population-based newborn
screening for congenital CMV infection (Dollard et al,
in press; Kharrazi et al, 2010). However, recent evidence
from a large multicenter study suggests that DBS is not sufficiently sensitive for diagnosis of congenital CMV compared with detection in other bodily fluids such as saliva
and urine (Boppana et al, 2010). An alternative approach
would be to analyze the DBSs obtained from infants with
failed newborn hearing screens, because congenital CMV
infection can be identified in approximately 3% of such
cases (Choi et al, 2009). This approach would allow early
detection and more timely intervention; however, it would
fail to identify those congenitally infected infants who pass
the newborn hearing screening but later develop SNHL.
Cranial ultrasonography, head CT, and brain MRI
are used to detect brain lesions associated with congenital CMV infection. CNS anomalies can be detected in
some infants in utero. Fetal MRI can also detect abnormalities, including microcephaly and cortical anomalies,
even when the ultrasound is normal; this appears to be
the preferred modality for diagnosis of fetal CNS involvement (see F
igure 37-4; Benoist et al, 2008; Doneda et al,
2010). Because the finding of CNS disease is a potential
harbinger of permanent sequelae, diagnostic CNS imaging is warranted in all suspected cases of congenital infection (Ancora et al, 2007; Boesch et al, 1989; Boppana
et al, 1997; Kylat et al, 2006). Any of the standard imaging modalities is valuable in assessing CNS involvement.
Ultrasound, because of its convenience, is an appropriate
initial study and is particularly valuable and sensitive in
detecting periventricular calcifications and lenticulostriate
vasculopathy associated with mild to moderate ventricular
dilatation. MRI provides important additional information, particularly the presence of associated polymicrogyria, hippocampal dysplasia, and cerebellar hypoplasia (de
Vries et al, 2004); therefore the staged sequential use of
ultrasound and MRI is probably the preferred approach to
CNS imaging in this setting.
Careful initial hearing evaluation and longitudinal
monitoring for SNHL is required in all infants with documented congenital CMV infection, given that this complication is the most common late sequela of congenital
CMV infection. Early recognition of SNHL and institution of appropriate interventions (speech–language therapy, centers for deafness education, and cochlear implants)
can markedly improve the developmental, social, and language skills of a child with hearing impairment. Any child
born with congenital CMV infection, whether symptomatic or not, deserves careful and recurring evaluation for
cognitive delays, visual impairment, or motor disabilities.
Follow-up evaluation should include a multidisciplinary
team approach involving a pediatric infectious diseases
specialist, pediatric otolaryngologist, and child behavioraldevelopmental specialist, in addition to a physical therapist, ophthalmologist, and neurologist as needed.
Treatment: Antiviral Intervention in the
Newborn and the Pregnant Patient
Experience with the use of antiviral therapies against CMV
in immunosuppressed patients, particularly solid organ and
hematopoietic stem cell recipients and patients with HIV
infection, is considerable (Boeckh and Ljungman, 2009;
Razonable, 2005). Several studies in recent years have
extended this experience to infants and children and have
clearly indicated a benefit of antiviral therapy against CMV
infection in several settings. In a phase 3, randomized, nonblinded controlled trial of ganciclovir for newborns with
congenital CMV disease (Kimberlin et al, 2003), a group
of infants with virologically confirmed congenital CMV
received a 6-week course of ganciclovir (6 mg/kg every
12 hours). The primary endpoint was improved hearing
or retention of normal hearing. Significant differences
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
between treated and untreated groups were noted, including a statistically higher likelihood of normal or improved
hearing at 6 months of age in treated infants compared
to controls. Although almost two thirds of the treated
infants had neutropenia, it was reversible when antiviral
therapy was halted. In a follow-up assessment, infants with
symptomatic congenital CMV involving the CNS who
received intravenous ganciclovir therapy had fewer developmental delays at 6 and 12 months, using the Denver
Developmental Screening test, compared with untreated
infants (Oliver et al, 2009). Because the use of ganciclovir
can promote improved hearing and neurodevelopmental
outcomes, antiviral therapy should be considered for all
infants with congenital CMV infection with any evidence
of CNS involvement (microcephaly, abnormal CNS imaging study, positive CSF for CMV DNA, chorioretinitis, or
evidence of SNHL).
Ganciclovir should also be used in any infant with severe
or life-threatening end-organ CMV disease, whether
acquired via congenital infection or via a postnatal route
(see Table 37-2) (Schleiss and McVoy, 2004). CMV retinitis in the congenitally infected infant can represent a
particularly problematic management issue; it has been
reported that up to 6 months of antiviral therapy may be
required to control chorioretinitis in the symptomatic congenitally infected infant (Shoji et al, 2010). All infants with
documented congenital infection should have an ophthalmologic evaluation. If chorioretinitis is present, it should
be managed in consultation with an ophthalmologist and
infectious diseases expert. Although ganciclovir improves
neurodevelopmental outcomes for symptomatic infants,
it is not clear whether ganciclovir holds the promise of
improving outcomes in infants with asymptomatic congenital CMV infection. Because some of these infants are
at risk to progress to SNHL, clinical trials are warranted to
explore whether antiviral therapy could prevent development of hearing loss. Other investigational studies including the assessment of the therapeutic potential in infants
of the prodrug of ganciclovir, valganciclovir, are needed.
The potential role of valganciclovir in long-term (suppressive) therapy of CMV in congenitally infected infants
is currently under investigation. The CASG is currently
conducting a clinical trial of 6 weeks versus 6 months of
valganciclovir (www.casg.uab.edu; Nassetta et al, 2009). A
number of case reports and small case series have reported
the safety, tolerability, and possibly efficacy of long-term
oral therapy with valganciclovir in the setting of congenital CMV infection (Amir et al, 2010; Hilgendorff et al,
2009; Lombardi et al, 2009; Yilmaz Çiftdogan and Vardar,
2011), but the uncontrolled nature of these reports precludes any recommendations for oral therapy at this time.
The prospect of treating the pregnant patient to prevent transmission of CMV to the fetus is an area of active
investigation. Ganciclovir has demonstrated teratogenic
risk in some studies (Schleiss and McVoy, 2004); although
this has never been demonstrated in humans, it has limited
research in this area. A case report of the use of oral ganciclovir in a pregnant liver transplant patient did not show
any evidence of teratogenicity (Pescovitz, 1999). Ganciclovir has been demonstrated to cross the placenta, and therefore could theoretically be used to treat CMV infection
in utero (Brady et al, 2002). An observational study of 20
483
women with 21 fetuses, with confirmed congenital CMV
infection treated with oral valacyclovir, demonstrated placental transfer of valacyclovir with measurable concentrations in the amniotic fluid and a subsequent reduced viral
load in the fetal blood (Jacquemard et al, 2007). There have
been several case reports of treatment of congenital CMV
infection in utero with oral, parenteral, or intraamniotic
ganciclovir with varying degrees of success (Miguelez
et al, 1998; Puliyanda et al, 2005; Revello et al, 1993,
1999a). Although it is probably safe, prenatal treatment of
fetal CMV infection with ganciclovir is currently not supported by the available data; further study with a randomized controlled trial is needed.
Passive immunization with CMV human immunoglobulin (HIG) has been studied for the in utero treatment and
prevention of congenital CMV infection. CMV HIG is a
pooled, high-titer immunoglobulin preparation derived
from donors with high levels of CMV antibody. Nigro
et al (2005) completed a prospective study of CMV HIG
for the treatment of pregnant women with primary CMV
infection, including some women with confirmed fetal
CMV (Nigro et al, 2005). The women were enrolled in the
therapy group if they had an amniocentesis and confirmed
congenital CMV infection, as evidenced by a positive
PCR in the amniotic fluid; they were enrolled in prevention group if they did not have an amniocentesis. In the
therapy group, only 1 in 31 of the treated mothers delivered an infant with congenital CMV disease, compared to
7 in 14 mothers who were not treated with HIG. In the
prevention group 6 of 37 mothers receiving HIG delivered infants with congenital CMV, compared to 19 of 7
mothers who did not receive treatment. Overall, there was
a statistically significant reduction of risk for congenital
CMV infection with HIG therapy. In a subsequent study,
three fetuses treated with HIG had resolution of their
ultrasonographically detected cerebral abnormalities; in
contrast the two untreated fetuses had persistence of their
cerebral abnormalities (Nigro et al, 2008). In addition to
fetal effects, CMV HIG has been demonstrated to affect
the placenta. In pregnancies treated with HIG, significant
reductions in placental thickness have been demonstrated
(La Torre et al, 2006). The reduction in placental thickness with HIG treatment suggests that at least part of the
beneficial effect of treatment is mediated at the level of the
placenta (Schleiss, 2006a). Randomized controlled trials of
HIG for the treatment and prevention of congenital CMV
infection are needed. Until such data are available, clinicians could consider treatment with CMV HIG in pregnant patients with confirmed fetal CMV infection.
Natal Acquisition of CMV Infection:
Implications for the Premature Infant
In addition to congenital infection, CMV can produce
disease in the newborn infant after natal acquisition; this
can occur via one of three mechanisms: (1) transmission
in the birth canal during vaginal delivery after exposure
to infectious cervicovaginal secretions, (2) through ingestion of breast milk, and (3) via blood transfusion. Of these
potential mechanisms, the most common is via breast
milk (Schleiss, 2006c), with transmission in the birth canal
occurring less commonly. It has long been recognized that
484
PART IX Immunology and Infections
CMV is shed by the cervix (Montgomery et al, 1972) and
excreted in the breast milk of seropositive women (Hayes
et al, 1972; Stagno, 1982a). The risk of CMV transmission in infants who are breastfed by seropositive women
shedding virus in their breast milk has been reported to
be between 58% and 69% (Dworsky et al, 1983; Stagno
et al, 1980). CMV infection acquired in the postnatal period in healthy term infants by this route typically
is asymptomatic, only rarely producing any morbidity.
There is no convincing evidence that acquisition of CMV
via breast milk leads to any adverse neurodevelopmental
sequelae (Kurath et al, 2010). In a study of CMV transmission through breastfeeding, all the infants who acquired
CMV infection had normal neurodevelopment at a mean
follow-up of 51 months of age (Vollmer et al, 2004).
Although the safety of breastfeeding has been established in term infants in CMV-seropositive women,
controversy exists regarding the safety of breastfeeding
low-birthweight, premature infants. Studies in low-birthweight and very low-birthweight (VLBW) preterm infants
yield conflicting results regarding to the risk of developing symptomatic infection after breast milk acquisition of
CMV (Schleiss, 2006c). In one study of VLBW premature
infants (<32 weeks’ gestation, <1500 g) exposed to CMV
via breast milk, virus transmission occurred in 33 of the 87
exposed infants (Maschmann et al, 2001). Approximately
half of these infants were ill and exhibited symptoms such
as hepatopathy, neutropenia, thrombocytopenia, and sepsislike deterioration. Conflicting reports of early postnatal
CMV infection in preterm infants in highly immune populations have suggested that symptomatic CMV infection
acquired by breast milk is rare (Mussi-Pinhata et al, 2004;
Kurath et al, 2010). Proposed efforts to reduce the infectivity of breast milk from seropositive mothers have included
freezing breast milk at –20°C, Holder pasteurization, and
short-term pasteurization (Hamprecht et al, 2004). Of
these methods, freezing is the most studied and most likely
to maintain the salutary immunologic properties of breast
milk. Some experts recommend freeze-thawing all breast
milk before feeding the VLBW premature infant if the
mother is known to be CMV seropositive, or if her CMV
serostatus is unknown. Although freezing breast milk may
lower the incidence of postnatally acquired CMV infection, it does not entirely eliminate the risk (Maschmann
et al, 2006). It is presumed that VLBW premature infants
are at increased risk because they possess fewer transplacentally acquired antibodies against CMV than do term
babies, and thus are more likely to develop disease upon
infection. It has recently been shown that treating the
VLBW infant with IVIG appeared to reduce the likelihood of transmission of CMV by breast milk (Capretti
et al, 2009), although this is not currently considered to be
an indication for the use of IVIG in the premature infant.
Further evidence is necessary to make recommendations
regarding what, if any, interventions are appropriate in
low-birthweight, preterm infants receiving breast milk
from CMV-seropositive mothers.
CMV can be transmitted through blood transfusion,
and transfusion-associated infections were at one time
a major problem in the neonatal intensive care setting
(Adler, 1986; Adler et al, 1983, 1984; Prober et al, 1981).
Two approaches are currently used to decrease the risks
of transfusion-associated CMV, leukocyte reduction, and
directed transfusion of CMV-negative blood products
(Lamberson et al, 1988). Although leukocyte reduction has
had a dramatic effect on the risk of transfusion-associated
CMV, reports are conflicting in the literature regarding
the question of whether this intervention is completely
effective at eliminating the risk of transfusion-transmitted
CMV (Allain et al, 2009; Fergusson et al, 2002; Vamvakas, 2005). A recent survey of the American Association of Blood Banks physician membership revealed that
65% of those responding believed that leukocyte-reduced
and CMV-negative blood components were equivalent in
their ability to prevent transfusion-associated transmission of CMV (Smith et al, 2010). Leukoreduction should
be adequate for preventing the overwhelming majority of
transfusion-associated CMV infections in the newborn
intensive care unit. However, despite the American Association of Blood Banks survey results on attitudes toward
leukocyte-reduced blood products, fetuses and neonates
are more likely to receive CMV-negative products compared with other groups receiving transfusions, and additional studies are warranted to ascertain what risk may
exist for premature infants receiving leukocyte-reduced
products from CMV-seropositive donors.
Prevention
One important strategy for addressing the problem of
congenital CMV is the education of women of childbearing age about the risks of transmission and strategies for
prevention. Child care providers (including daycare workers, special education teachers, and therapists) appear
to have a higher risk of occupational exposure to CMV,
because of extensive contact with infants and young children (Adler, 1989; Pass et al, 1986, 1990). Education of
the potential occupational risk in this group is essential. In
contrast to these child care providers, health care workers
who appropriately use routine infection control practices
are not at increased risk of CMV acquisition. CMV infections in pregnant women are typically clinically “silent.”
Like most healthy individuals, more than 90% of pregnant
women with primary CMV infections have no symptoms.
When symptoms occur, they are nonspecific and vague,
often described as a flulike syndrome. Potential manifestations include fever, fatigue, headache, myalgia, lymphadenitis, and pharyngitis, but these are the exception and
not the rule. Because most maternal CMV infections are
asymptomatic, a major goal is education of all women of
childbearing age on hygienic practices (ACOG Practice
Bulletin, 2002; Jeon et al, 2006; Ross et al, 2006). Hygienic
strategies are important because the saliva and urine of
infected children are significant sources of CMV infection
among pregnant women. Strategies include washing hands
whenever there is contact with a child’s saliva or urine, not
sharing food, utensils, or cups, and not kissing a child on
the mouth or cheek (Anderson et al, 2007; Cannon and
Davis, 2005). It is also essential that women become better educated on the importance of CMV infection. A survey of women in 2005 showed that only 14% of women
knew what CMV was, but most believed that preventative measures for an infection that could harm an unborn
baby would generally be acceptable (Ross et al, 2008). The
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
effectiveness of educating pregnant women on methods to
prevent CMV transmission has been demonstrated (Adler
et al, 1996). In a study in which seronegative mothers with
a child in group day care were instructed on measures to
prevent CMV transmission, pregnant mothers had a significantly lower rate of CMV infection when compared
with nonpregnant mothers attempting conception (Adler
et al, 2004). In addition, a study in France recently demonstrated a lower CMV seroconversion rate after counseling
pregnant women on hygienic measures (Vauloup-Fellous
et al, 2009).
Prenatal maternal screening for CMV antibodies is controversial. Because women who are CMV-immune can
be reinfected with new strains that can then be transmitted to the fetus, with subsequent sequelae (Boppana et al,
1999, 2001), the finding of a positive preconception titer
for CMV IgG antibody may provide a false sense of reassurance and decrease a pregnant patient’s motivation to
engage in careful hygienic practices. A recent study evaluated three screening strategies and suggested that universal maternal screening for CMV could be a cost-effective
strategy if a treatment were available that could achieve a
47% reduction in disease (Cahill et al, 2009).
Ultimately the control of congenital CMV could be
realized by the development of an effective vaccine. No
CMV vaccines are currently licensed; however, because
of the enormous economic impact of congenital CMV,
the Institute of Medicine has identified a CMV vaccine
as the highest-level priority for new vaccine development
(excluding HIV vaccines) for the United States (Stratton
et al, 2001). The CMV envelope glycoprotein B has been
the most studied subunit vaccine candidate for this purpose,
because it is a target of neutralizing antibody in all CMVseropositive individuals. Results of a phase II, placebo-controlled, randomized, double-blind trial of the recombinant
CMV envelope glycoprotein B with MF59 adjuvant were
published recently (Pass et al, 2009). In this study, three
doses of the CMV vaccine or placebo were administered
at 0, 1, and 6 months to healthy women within 12 months
postpartum. Women in the vaccine group were less likely
than the placebo group to become infected with CMV (p =
0.02) with 18 of 225 women becoming infected with CMV
in the vaccine group, compared to 31 o 216 in the placebo
group. This report is the first to document efficacy of any
CMV vaccine in a clinical trial and should help to accelerate the pace of future vaccine development and testing.
Other clinical trials include evaluation of live, attenuated
CMV vaccines and the testing of subunit vaccines targeting other key proteins involved in the humoral and cellular
immune responses to CMV infection (Schleiss, 2008).
HUMAN HERPESVIRUS 6 AND 7
Human herpesvirus (HHV) 6 was isolated in tissue culture in 1986 from peripheral blood leukocytes of patients
with both lymphoproliferative disorders and HIV infection (Bernstein and Schleiss, 1996; Schleiss, 2009). The
virus was eventually shown to have a tropism for T cells,
and molecular studies revealed homology with CMV.
These findings suggested that the virus belonged to the
family Herpesviridae. For several years after discovery, its
role in disease was unclear, but it is now known to be the
485
major etiologic agent of roseola infantum (exanthem subitum) and has been implicated in other clinical syndromes.
HHV-6 is a prototypical β-herpesvirus, with a doublestranded DNA genome contained within an icosahedral
capsid, surrounded by an outer envelope. HHV-6 is subclassified as either variant A or B, based on differences in
nucleotide sequence, restriction enzyme profile, and reactivity with monoclonal antibodies. HHV-6B is the subtype
typically associated with exanthem subitum (Yamanishi et
al, 1988). HHV-6 and HHV-7 are ubiquitous in nature,
and typically cause infection in the first 2 years of life.
HHV-7 is highly related to HHV-6 and, like HHV-6,
is responsible for roseola infantum (Tanaka et al, 1994).
HHV-7 is a β-herpesvirus, structurally and molecularly
similar to CMV and HHV-6. It was first isolated from
CD4+ T cells of a healthy individual. The high degree of
homology with HHV-6 creates difficulty in interpretation of serologic assays, which are largely investigational
in nature and not generally useful for clinical practice,
because there is considerable cross-reactivity of antibodies
between HHV-6 and HHV-7 proteins. As with HHV-6,
infection with HHV-7 appears to be ubiquitous, although
infection appears to be acquired somewhat later in life than
is HHV-6 (Caserta et al, 1998; Suga et al, 1997). Approximately 40% to 45% of children have antibodies to HHV-7
by 2 years of age, and 70% of children are seropositive
by 6 years of age. Like other β-herpesviruses, HHV-7 can
be found in the saliva, suggesting a route for person-toperson transmission. Based on its identification in cervical secretions, HHV-7 also has the potential for perinatal
transmission (Okuno et al, 1995).
It is of interest to examine the evidence that suggests
that HHV-6, like CMV, can cross the placenta and infect
the fetus in utero, given the extensive molecular similarities between these two viruses. In one early study, HHV-6
DNA was detected by PCR in approximately 25% of pregnant seropositive women at the time of delivery, and in
approximately 1% of cord blood samples, suggesting the
possibility of congenital transmission (Dahl et al, 1999).
Examination of 305 cord blood samples in another study
identified HHV-6 DNA by PCR in 1.6% of infants (Adams
et al, 1998). Interestingly, this potential vertical transmission rate of 1% to 2% is similar to congenital infection rates
commonly reported for CMV. Congenital HHV-6 transmission was first definitively reported in a study of 5638
cord bloods; 57 samples (1%) had HHV-6 DNA by PCR,
but none had HHV-7 (Hall et al, 2004). Of note, these
infections were all asymptomatic, and the HHV-6 genome
variant in one third of congenital infections was HHV6A,
in contrast to the more common variant HHV6B, which is
encountered in virtually all postnatal infections. Although
the rate of congenital HHV-6 transmission at approximately 1% is highly similar to that observed for congenital CMV, the mechanisms of transmission are different. It
has been shown that vertical transmission of HHV-6 most
often occurs (90% of cases) because of the germline passage
of chromosomally integrated HHV-6 (Hall et al, 2008).
The mode of inheritance of HHV-6 genome in this form
of vertical transmission appears to be exclusively maternal (Hall et al, 2010). HHV-6 is unique among the human
Herpesviridae regarding its capacity to integrate into the
host chromosome and mediate germline transmission of
486
PART IX Immunology and Infections
viral genome from mother to fetus. Remarkably, HHV-6
is the first functional virus of any type reported to be
passed through the human germline. The clinical consequences of such transmission, and the differences in germline transmission of viral genome and the more common
postnatal acquisition of HHV-6 in early childhood, remain
unknown. That HHV-6 genome appears to integrate into
the telomere—a chromosomal component important in
cellular aging and in cancer—suggests that there may be
interesting long-term consequences associated with vertical germline transmission of this virus (Arbuckle et al,
2010; Nacheva et al, 2008) that remain to be elucidated.
HHV-6 and HHV-7 DNA has been found to be present in cervical secretions, placenta, and peripheral blood
mononuclear cells of pregnant women (Caserta et al, 2007).
Whether intrapartum transmission of these viruses can
occur in the birth canal during delivery remains unknown.
HHV-6 DNA can also be found in breast milk. Perinatal
transmission via this mechanism has been postulated (Joshi
et al, 2000), but has not been demonstrated.
KAPOSI’S SARCOMA HERPESVIRUS
AND EPSTEIN–BARR VIRUS
In 1994 a novel herpesvirus, HHV-8 or KSHV, was identified in patients with AIDS-associated Kaposi’s sarcoma
(KS; Chang et al, 1994). This virus was assigned to the
γ-herpesvirus family of the Herpesviridae, based on its
molecular and sequence similarity to the other prototypical γ-herpesvirus, Epstein–Barr virus. Subsequent studies
have linked KSHV to both AIDS-associated KS and the
endemic forms of KS that are prevalent in elderly Mediterranean men. Little is known about routes of transmission of KSHV in nonpregnant patients, let alone whether
it can be transmitted perinatally. The clinical significance
of acquiring primary KSHV infection outside of the HIV
setting is also unclear. Serologic evidence from sexually
transmitted disease clinics suggests that sexual contact is
a risk factor for acquiring KSHV. The epidemiology of
primary HHV-8 infection appears to vary considerably
worldwide. Indeed, the routes of acquisition of infection
and mechanisms responsible for person-to-person transmission remain uncertain. After the virus was initially
discovered, the unique role it seemed to play in inducing
malignant disease in HIV-infected patients suggested that
the primary route of transmission of HHV-8 was through
sexual contact, particularly among gay men. However,
more recent evidence suggests that other routes of infection exist, including transmission by saliva (Pica and Volpi,
2007).
A recent cross-sectional study of the seroprevalence of
HHV-8 in children and adolescents in the United States
indicated a prevalence of approximately 1% (Anderson
et al, 2008). There appears to be considerable regional variation in prevalence in the United States. In a population
of children in south Texas, the seroprevalence was 26%,
strongly suggesting that nonsexual modes of transmission
predominate (Baillargeon et al, 2002). In Sub-Saharan
Africa, prevalence in children is even higher, approaching
60% in some studies (Sarmati, 2004). There are reports of
infections in infants that suggest possible vertical transmission, but congenital infection has not been demonstrable
by PCR techniques (Sarmati et al, 2004). HHV-8 can also
be transmitted by blood transfusion (Hladik et al, 2006);
this observation would suggest that transplacental transmission is at least theoretically feasible. In a study of 89
KSHV-seropositive women, 13 mothers (14.6%) had
KSHV DNA detected in their peripheral blood mononuclear cells; 2 of 89 samples drawn at birth from infants
born to these mothers had KSHV DNA detectable within
their peripheral blood mononuclear cells. These findings
suggest that KSHV can be transmitted perinatally, but
infrequently (Mantina et al, 2001). As serologic and nucleic
acid–based diagnostics tests become more widely available,
a better assessment of the worldwide seroepidemiology of
HHV-8 infection and an increased understanding of its
modes of transmission will be achievable.
Most primary infections with HHV-8 are probably
asymptomatic, although the clinical course of primary
symptomatic HHV-8 infection in immunocompetent
children has been described (Andreoni et al, 2002). In this
study, fever and rash were noted with primary infection.
The rash first appeared on the face and gradually spread
to the trunk, arms, and legs. It initially consisted of discrete red macules that blanched with pressure and eventually became papular. An upper respiratory tract infection
appeared as a secondary symptom in most children, and a
lower respiratory tract infection appeared as a secondary
symptom in one third of symptomatic children. Additional
information on the epidemiology and modes of transmission of this pathogen, particularly in the prenatal and
intrapartum period, is needed.
There is a minimal amount of information available
about prenatal and perinatal modes of transmission of
EBV. EBV is the causative agent of infectious mononucleosis and is associated with nasopharyngeal carcinoma,
Burkitt’s lymphoma, and lymphoproliferative disease in
immunocompromised patients (Schleiss, 2009). Primary
EBV infection during pregnancy appears to be rare. In a
prospective study, susceptibility to EBV infection in 1729
pregnant women was evaluated by screening for EBV
antibodies. Fifty-eight subjects (3.4%) had no detectable
EBV antibody and were presumably susceptible. Of the
54 women who agreed to participate in this study, none
acquired EBV antibody during pregnancy (Le et al, 1983).
It is not clear whether transplacental passage of EBV in
seropositive pregnant women occurs. Because EBV can be
acquired by blood transfusion, such a mode of transmission
is feasible. It has been postulated that high-titer antibodies cross the placenta and protect the fetus from hematogenous transmission of virus in women who reactivate EBV
during pregnancy (Purtilo and Sakamoto, 1982). A solitary
case report describes the occurrence of severe EBV disease
in a premature infant, born at 28 weeks’ gestation, who
was examined on the 42nd day of life with hepatosplenomegaly, hemolytic anemia, thrombocytopenia, and atypical lymphocytosis (Andronikou et al, 1999). In another
study, the potential for EBV vertical transmission from a
seropositive mother to her child was evaluated in 67 pregnant women by nested PCR (Meyohas et al, 1996). Two of
67 neonates were positive for EBV DNA, suggesting that
mother-to-child transmission of free EBV or of maternal
EBV-infected cells can occur during pregnancy in the setting of latent EBV infection, but no clinical consequences
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
of these putative congenital infections were reported. In
a study of six pregnant women with evidence of primary
EBV infection, four pathologic births were observed: one
spontaneous abortion, two premature babies, one of whom
died, and one stillborn with multiple malformations (Icart
et al, 1981). However, the assessment of primary infection
was drawn solely from the serologic profile (including the
presence of anti–early antigen antibodies) and the relationship of EBV infection to adverse pregnancy outcome was
undefined, and it was unclear whether intrauterine EBV
infection was present. In another study, placentas and some
fetuses were studied in five cases of pregnancy interruption caused by maternal infectious mononucleosis in early
gestation (Ornoy et al, 1982). Decidual lesions, consisting
of perivasculitis and necrotizing deciduitis, were noted,
and endovasculitis, perivasculitis, and occasional vascular
obliteration were found in villi, as well as mononuclear and
plasma cell infiltrates. Two fetal hearts exhibited evidence
of myocarditis. These observations suggested a pathogenic
role for placental and fetal EBV infection, but no direct
virologic evidence of EBV was obtained, and these observations have not been duplicated. Accordingly a recent
review concluded that maternal EBV infection did not
impose a serious threat to pregnancy outcome (Avgil and
Ornoy, 2006) and that no strong evidence for teratogenicity or fetal injury existed.
HUMAN PARVOVIRUS B19
Parvovirus B19, a small, single-stranded DNA virus, is the
only member of the parvovirus family that causes human
disease. The virus was identified in 1975 (Cossart et al,
1975) and was first linked to a disease in 1981—aplastic
crisis in children with sickle cell anemia (Pattison et al,
1981). Primary infection with parvovirus B19 is commonly
known as fifth disease or erythema infectiosum; it is classically described as a childhood exanthem with a “slapped
cheek” appearance (Anderson et al, 1984). The agent is
less ubiquitous than other common childhood exanthematous illnesses, such as HHV-6; therefore many individuals
reach adulthood with no prior evidence of infection, placing women of childbearing age at potential risk. Considerable interest in the role of this virus in hydrops fetalis
(nonimmune) and fetal aplastic crisis has evolved since the
first cases of fetal death associated with maternal parvovirus B19 infection were reported in the 1980s (Brown et al,
1984; Kinney et al, 1988).
Epidemiology
Parvovirus B19 infection is global in nature. It is common in childhood and continues at a low rate throughout
adult life. One study identified an annual seroconversion
of 1.5% in women of childbearing age unrelated to their
occupation (Koch and Adler, 1989). The peak incidence of
erythema infectiosum is in the late winter and early spring.
Periodic epidemics at intervals of a few years are typical.
The virus is spread by respiratory droplet (Anderson and
Cohen, 1987), by blood products (especially pooled clotting factor concentrates; Jordan et al, 1998), and transplacentally (Ergaz and Ornoy, 2006). Approximately 50%
to 80% of adults in the United States are seropositive
487
for human parvovirus B19 (Anderson, 1987; Vyse et al,
2007). A significant proportion of childbearing women
are thus susceptible to infection (Markenson and Yancey,
1998; Yaegashi et al, 1998). Preconception seroprevalence
to parvovirus B19 ranges from 24% to 84% (Ergaz and
Ornoy, 2006). During pregnancy the risk of acquiring parvovirus B19 infection is low, ranging from 0% to 16.5% in
different studies (Ergaz and Ornoy, 2006). The risk of primary maternal infection is higher during epidemics, with
reported seroconversion rates ranging between 3% (Kerr
et al, 1994) and 34% (Woernle et al, 1987).
It is estimated that one fourth to half of maternal parvovirus infections result in transmission of infection to the
fetus (Alger, 1997; Gratacos et al, 1995; Koch et al, 1998).
The vast majority of pregnancies are unaffected (Berry
et al, 1992; Sheikh et al, 1992). The risk of adverse fetal
outcome is increased if maternal infection occurs during
the first two trimesters of pregnancy (Skjoldebrand-Sparre
et al, 2000), particularly before 20 weeks’ gestation. There
are conflicting reports regarding the prognosis once fetal
infection has been established. A longitudinal study of fetal
morbidity and mortality in more than 1000 women with
primary parvovirus B19 infection in pregnancy demonstrated a risk of fetal hydrops of 3.9% and a risk of fetal
death of 6.3%; fetal death was observed only if maternal infection occurred before the 20th week of gestation
(Enders et al, 2004). A recent retrospective analysis of
intrauterine parvovirus B19 infection at a single site suggested that the rate of adverse fetal outcome is much higher
than previously appreciated, with fetal hydrops and demise
occurring in greater than 10% of pregnancies (Beigi et al,
2008), although the total number of cases reported in this
series was low; this primarily represented a referral population to a tertiary care center.
Pathogenesis
The most common mode of transmission of parvovirus B19
is via a respiratory route. Typically, once the virus establishes infection, viremia occurs, followed by mild systemic
symptoms such as fever and malaise. Viremia is short-lived,
lasting only 1 to 3 days, and the characteristic immunemediated rash develops 1 to 2 weeks later. Once the rash
appears, an individual is no longer infectious. Arthropathy caused by parvovirus B19 is common; it is observed
more frequently in adults with primary infection than in
children. It typically manifests late in the course of illness
with acute onset of arthralgias or frank arthritis involving
the hands, knees, wrists, and ankles. The symptoms usually
subside within 1 to 3 weeks, although approximately 20%
of affected women have persistent or recurring arthropathy for months to years (Woolf et al, 1989).
Potential pathogenic mechanisms involve the recognized affinity of parvovirus B19 for progenitor erythroid
cells of bone marrow. The blood group P antigen is a main
cellular receptor for parvovirus B19, and it is found on red
blood cells and on placental trophoblast cells (Jordan et
al, 2001). The P antigen is also expressed on fetal cardiac
myocytes, enabling parvovirus B19 to infect myocardial
cells (Rouger et al, 1987) leading to myocarditis (von Kaisenberg et al, 2001). Myocarditis induced by parvovirus
B19 can contribute to high-output cardiac failure, and the
488
PART IX Immunology and Infections
myocardial inflammation and subendocardial fibroelastosis may also contribute to fetal hydrops (Morey et al,
1992). Fetal infection most likely occurs hematogenously
via the placenta during maternal viremia. Parvovirus B19
infection in utero causes a pronormoblast arrest, which
leads to fetal anemia, nonimmune hydrops, and sometimes
progressive congestive heart failure (Ergaz and Ornoy,
2006; Kinney et al, 1988). The fetus is especially susceptible to adverse consequences of red blood cell infection,
secondary to the intrinsic short fetal erythrocyte life span
and rapidly expanding blood volume, especially during the
second trimester. It has also been postulated that parvovirus B19 infection leads to cytotoxicity and subsequent
anemia by inducing apoptosis of infected red blood cells
(Yaegashi et al, 1999, 2000). The NS1 protein of parvovirus B19 induces cell death by apoptosis in erythroidlineage cells by a pathway that involves caspase 3, whose
activation may be a key event during parvovirus-induced
cell death (Moffatt et al, 1998). The NS1 protein also has
a key role in the arrest of infected cells at the G1 phase of
the cell cycle prior to apoptosis induction (Chisaka et al,
2003). In addition to erythroid precursors and myocytes,
other organs appear to be involved in fetal parvovirus B19
infection. Fetal brain infection has been reported. Neuropathologic findings in the infected hydropic fetus include
perivascular calcifications, primarily in the cerebral white
matter, as well as multinucleated giant cells. Viral DNA
has been demonstrated in the brain and liver (Isumi et al,
1999). Data also suggest that the maternal cell–mediated
immune response at the placental level contributes to the
pathogenesis of congenital infection. In one study, placentas from women whose pregnancies were complicated by
parvovirus B19 infection had increased infiltration of CD3
T cells and elevated levels of interleukin 2 (Jordan et al,
2001).
Clinical Spectrum
Parvovirus B19 infection causes erythema infectiosum, or
fifth disease, in normal hosts, aplastic crisis in patients with
hemolytic disorders, and chronic anemia in immunocompromised hosts. A substantial proportion of infected adult
women may also have arthropathy in association with parvovirus B19 infection (Woolf et al, 1989). Maternal symptoms have been present in up to two thirds of documented
cases of nonimmune hydrops fetalis associated with parvovirus B19 infection (Yaegashi et al, 1998).
The major clinical presentation of parvovirus B19 infection in the fetus is hydrops fetalis. Various estimates suggest that human parvovirus B19 infection contributes
from 10% to 27% of cases of nonimmune hydrops fetalis
(Essary et al, 1998; Markenson and Yancey, 1998; Yaegashi et al, 1994). Nonimmune hydrops fetalis was the
main complication in 0.9% to 23% of pregnancies among
proven maternal infections with parvovirus B19 (Ergaz and
Ornoy, 2006). A risk of 7.1% for hydrops fetalis has been
described for pregnant women who acquire parvovirus B19
infection between 13 and 20 weeks’ gestation (Enders et
al, 2004). Parvovirus B19 does not appear to have a major
role in intrauterine fetal demise in the absence of hydrops
fetalis (Riipinen et al, 2008). In addition to hydrops fetalis, in recent years there have been increasing numbers of
case reports of neurologic and ophthalmologic anomalies
associated with fetal parvovirus B19 infections. Although
some studies undertaken to examine the association
between fetal infection and congenital anomalies failed
to reveal any associations (Kinney et al, 1988; Mortimer
et al, 1985), parvovirus B19 has increasingly been recognized as a cause of neuronal migration defects (Pistorius et
al, 2008). The role of parvovirus B19 in neurodevelopmental injury has not been fully explored. There have been at
least three case reports of fetal encephalopathy associated
with in utero infection with parvovirus B19 (Alger, 1997).
Parvovirus B19 has recently been recognized as a cause of
CNS injury in older children, including encephalitis, meningitis, stroke, and peripheral neuropathy (Douvoyiannis
et al, 2009). Consideration should be given to performing
brain imaging studies in infants with symptomatic in utero
parvovirus B19 infection. Such infants need careful neurodevelopmental follow-up. Few data are available regarding
long-term outcomes of infants infected in utero. Two prospective studies in the United Kingdom of approximately
300 congenitally exposed infants found the risk of major
congenital or developmental abnormality to be less than
1% (Miller et al, 1998).
Parvovirus B19 has been implicated in some cases of
congenital anemia (Heegaard and Brown, 2002). In one
series of 11 children with a diagnosis of Diamond-Blackfan syndrome, 3 of 11 bone marrow aspirates revealed
evidence of parvovirus B19 DNA. All three of these children, but none of the parvovirus B19 PCR-negative cases,
underwent spontaneous remission (Heegaard et al, 1996).
In light of these data, all infants undergoing evaluation for
congenital anemias should be evaluated for the possibility
of parvovirus B19 infection.
Laboratory Evaluation
In primary care, the diagnosis of human parvovirus B19
infection is most commonly made clinically through recognition of the characteristic rash. Serologic confirmation is necessary in high-risk situations, such as after a
significant exposure of a pregnant woman to a child with
erythema infectiosum. Both radioimmunoassays and
enzyme-linked immunosorbent assays are available for
detection of human parvovirus B19–specific IgG and IgM
antibodies (Kinney and Kumar, 1988). Presence of anti–
parvovirus B19 IgM in fetal blood or amniotic fluid may
confirm fetal infection, but may be detected in only one
fifth of infected fetuses (Torok et al, 1992). False-positive
results of parvovirus B19 IgM testing have been reported,
including cross-reactions with anti-rubella IgM (Dieck
et al, 1999).
Monitoring of the pregnant patient with a primary parvovirus B19 infection is an important clinical problem.
In the context of a human parvovirus B19 infection in a
symptomatic, pregnant woman, elevated or rising weekly
measurements of maternal alpha-fetoprotein suggest fetal
infection, and rising concentrations may be a marker for an
increased risk for hydrops fetalis (Carrington et al, 1987).
However, some studies have failed to demonstrate any
association between the magnitude of the elevation of the
α-fetoprotein and the severity of fetal anemia (Simms et al,
2009). Serial fetal ultrasonographic evaluations of infected
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
pregnant women are recommended to evaluate and follow
for fetal hydrops or intrauterine demise. The findings of
echogenic bowel, ascites, pleural or pericardial effusion,
or scalp edema are considered to be important markers
of fetal infection and pathology. Middle cerebral artery
Doppler to evaluate for fetal anemia may be another useful
prospective surveillance tool, because fetal anemia can be
detected using this technique before fetal hydrops is evident (Feldman et al, 2010). Other techniques to diagnose
fetal parvovirus B19 infection have been studied and used,
but most are not readily available. Virus can be cultured
from tissue in suspension cultures of bone marrow cells
from persons with hemolytic anemias, but it is difficult to
isolate; therefore this method is not feasible for prenatal
or postnatal diagnosis. Electron microscopy and histology
have permitted visualization of parvovirus in fetal blood,
ascitic fluid, tissue, and amniotic fluid, but the utility and
sensitivity of these evaluations have not been well studied
(Markenson and Yancey, 1998). A number of commercial
PCR assays are available, and these can be performed on
serum, amniotic fluid, or fetal tissue. Presumptive diagnosis can also be made based on finding IgM antibody in the
maternal and fetal blood. PCR for parvovirus B19 DNA or
in situ hybridization studies can be performed using maternal blood, amniotic fluid, cord blood, or fetal tissues. The
detection of B19 DNA in maternal blood appears to have
the best diagnostic sensitivity for identifying maternal B19
infection, and new-generation EIA and IgG avidity assays
appear to hold promise for improved serodiagnosis during
pregnancy (Enders et al, 2006, 2008).
Treatment
Spontaneous resolution of fetal hydrops with normal neonatal outcome has been reported in approximately one
third of cases (Humphrey et al, 1991; Rodis et al, 1998a;
Sheikh et al, 1992). Because two thirds of fetuses do not
recover without intervention, fetal transfusion is usually
recommended (Boley and Popek, 1993; Brown et al, 1994).
The earlier fetal transfusion is attempted, the more likely
it is to be successful. Cordocentesis allows precise assessment of the magnitude of fetal anemia, which can then
be corrected by blood transfusion, typically using packed
red blood cells. Using this approach, outcomes have been
favorable in most reported series, even among severely
anemic fetuses. In one report, packed red cell transfusion
was performed in 30 patients with fetal anemia (hemoglobin values ranging from 2.1 to 9.6 g/dL). The overall
survival rate was 83.8% (Schild et al, 1999). In a report
of 13 cases with severe hydrops fetalis who received intrauterine transfusion, 11 of 13 survived (84.6%), whereas all
the nontransfused fetuses with severe hydrops fetalis died
(Enders et al, 2004).
Prognosis
Mortality rates for fetal hydrops resulting from all nonimmune causes continue to exceed 50%, even with current aggressive therapies and intensive care (Huang et al,
2007; Wy et al, 1999). Hydrops secondary to parvovirus
B19 seems to have a better outcome than that from other
causes (Enders et al, 2004; Ismail et al, 2001). Although
489
few long-term prospective studies of infants born to mothers with documented primary parvovirus B19 infection
have been conducted, most report normal developmental
outcome. One case-control study involving approximately
200 mother–infant pairs found no differences in frequency
of developmental delay between infants born to women
with confirmed primary parvovirus B19 infection during
pregnancy and infants born to mothers with evidence of
preconceptional immunity (Rodis et al, 1998b). Other
studies have also suggested a favorable long prognosis
in children born to women with primary parvovirus B19
infections during pregnancy (Miller et al, 1998).
Prevention
If a pregnant woman has a significant exposure to an infectious case of parvovirus B19, counseling should be provided
regarding the potential risk of infection. Anti–parvovirus
IgM and IgG serologic analyses and serum PCR should
be performed; if they show evidence of primary infection, then serial fetal ultrasonographic evaluations should
be performed. Postexposure passive immunization with
immunoglobulin is not currently recommended because
the period of maternal viremia has passed by the time the
diagnosis of acute parvovirus B19 infection is made (Boley
and Popek, 1993). Although high-dose IVIG has been
used to attempt to prevent hydrops fetalis during pregnancy in the setting of acute infection (Selbing et al, 1995),
treatment with this modality in the pregnant woman or the
neonate has not been shown to improve fetal outcomes;
therefore it is not routinely recommended. Human IgG
monoclonal antibodies with potent neutralizing activity
have been generated, and these are suggested as candidates
for the development of immunotherapeutic approaches
for individuals chronically infected with parvovirus B19
virus or for acutely infected pregnant women (Gigler et al,
1999), but these interventions are not commercially available. There has been limited progress in the development
of a candidate parvovirus B19 vaccine. Phase 1 studies of a
recombinant vaccine based on baculovirus-produced capsids have been conducted, and this vaccine was found to
have a favorable safety and immunogenicity profile (Bansal
et al, 1993). Efforts are under way to research and develop
vaccines to prevent parvovirus B19 infections using other
expression systems (Lowin et al, 2005).
Pregnant health care providers should be counseled
about the potential risks to their fetus from parvovirus
B19 infections and should, at a minimum, wear masks and
use standard droplet precautions when caring for immunocompromised patients with chronic parvovirus B19
infection or patients with parvovirus B19–induced aplastic
crises. Some hospitals exclude pregnant health care providers from caring for these high-risk patients, but this
issue remains controversial.
RUBELLA
Rubella virus is an enveloped, single-stranded, positive
sense RNA virus belonging to the family Togaviridae.
Although togaviruses are typically vector-borne infections,
rubella is the notable exception, being transmitted instead
by respiratory droplet. Humans are the only known natural
490
PART IX Immunology and Infections
host for rubella virus. Rubella infection, commonly known
as the German measles, usually results in a mild illness with
an accompanying exanthem in adults and children; however, rubella produces serious consequences in pregnant
patients, in whom fetal infection can lead to serious anomalies. An ophthalmologist named Norman Gregg offered
the first description of congenital rubella syndrome (CRS)
in 1941 while investigating an epidemic of neonatal cataracts (Gregg, 1941). Not until the global pandemic of 1964
to 1965, however, were the multiple teratogenic manifestations of CRS fully appreciated and the permanent
neurodevelopmental consequences for newborns fully recognized. The capacity to grow the virus in tissue culture
led rapidly to the development of a vaccine and subsequent
a reduction in the incidence of CRS in the United States
and other developed countries. However, CRS is still
encountered in the developing world, and unfounded concerns about the safety of measles-mumps-rubella vaccine
have set the stage for potential reemergence of these diseases (Omer et al, 2009). Therefore a working knowledge
of CRS remains highly relevant to the care of newborns.
EPIDEMIOLOGY
Since the development of rubella vaccine in 1969, the
incidence of rubella in the United States has decreased
dramatically. The annual incidence of rubella cases has
dropped 99%, from 58 per 100,000 population in 1969
to less than 0.5 per 100,000 population in 1997 to 1999
(Danovaro-Holliday et al, 2001). CRS cases in the United
States have demonstrated a similar dramatic decline, and
in 2004 the Centers for Disease Control and Prevention
(CDC) concluded, against the background of few or no
reports of rubella activity from the 50 states and the virtual
absence of reported CRS, that endemic rubella had been
eliminated from the United States (Centers for Disease
Control and Prevention, 2005; Plotkin, 2006; Reef et al,
2006). The persistence of rubella in Latin America serves
as the source for a small number of importation cases of
rubella that are recognized in the United States annually (Castillo-Solórzano et al, 2003). In parts of the world
where routine rubella immunization is unavailable or not
used, rubella and CRS continue to be common. Rubella
is still an important and potentially preventable cause of
birth defects globally, with more than 100,000 cases of
CRS occurring annually in developing countries.
Maternal rubella infection that occurs in the period of
time from 1 month before conception through the second
trimester of pregnancy may be associated with transmission of infection and, depending on the timing of infection, disease in the infant. The frequency of congenital
infection after maternal rubella with a rash is 70% to 85%
if infection occurs during the first 12 weeks of gestation,
30% to 54% during the first 13 to 16 weeks of gestation,
and 10% to 25% at the end of the second trimester (Miller
et al, 1982; South and Sever, 1985). The classic findings of
congenital rubella are most typically associated with the
onset of maternal infection during the first 8 weeks of gestation (Miller et al, 1982). Both the risk of fetal infection
and severity of disease decline after the first trimester, and
the risk of any teratogenic effect is extremely low after 17
weeks’ gestation (Lee and Bowden, 2000).
PATHOGENESIS
Rubella virus is transmitted via respiratory droplets. Once
the oral or nasopharyngeal mucosae are infected, viral
replication occurs in the upper respiratory tract and nasopharyngeal lymphoid tissue. The virus then spreads contiguously to regional lymph nodes and hematogenously to
distant sites. Fetal infection is believed to occur as a consequence of maternal viremia. The mechanism by which
rubella infection of the fetus leads to teratogenesis has not
been fully determined, but the cytopathology in infected
fetal tissues suggests necrosis, apoptosis, or both, as well
as inhibition of cell division of precursor cells involved in
organogenesis (Atreya et al, 2004; Lee and Bowden, 2000).
The rubella replicase protein p90 interacts with a cellular cytokinesis-regulatory component (i.e., the citron-K
kinase) in the process leading to tetraploidy and cell cycle
arrest. In tissue culture, a number of unusual manifestations of rubella replication have been observed, including
mitochondrial abnormalities and disruption of the cytoskeleton. Characteristic markers of apoptosis such as DNA
fragmentation, nuclear chromatin condensation, and
annexin V staining can be observed following rubella infection in cell culture. Cytoplasmic inclusions have also been
reported in some cell lines. Microarray analysis following
rubella infection in cell culture demonstrated upregulation
of cytokines and interferon, suggesting that the induction
of inflammatory responses may serve as another possible
mechanism of injury induced by rubella (Adamo et al,
2008). Fetuses infected with rubella demonstrate cellular
damage in multiple sites and a noninflammatory necrosis
in target organs, including eyes, heart, brain, and ears (Lee
and Bowden, 2000).
CLINICAL SPECTRUM
The peak incidence of endemic rubella is in the late winter
and early spring months. Up to 50% of primary infections
are asymptomatic. The period of maximal communicability extends from a few days before until 7 days after onset
of the rash. Often a prodrome of mild systemic symptoms
precedes the rash by 1 to 5 days. Viremia can be detected
as early as 9 days before the onset of rash. Lymphadenopathy, which can precede a rash, often is present in the posterior auricular or suboccipital region. The rash classically
begins on the face and spreads caudally to the trunk and
extremities. Symptoms generally last up to 3 days, and the
incubation period ranges from 14 to 21 days. Transmission by breastfeeding in a case of postpartum acquisition
of infection has been described (Klein et al, 1980). Typical
illness in adults and children with acquired rubella infection consists of an acute generalized maculopapular rash,
fever, and arthralgias, arthritis, or lymphadenopathy. Conjunctivitis is also common. Encephalitis (1:5000 cases) and
thrombocytopenia (1:3000 cases) are complications.
Infants with congenital rubella are usually born at term,
but often are small for gestational age. The most common
isolated sequela is hearing loss (Miller et al, 1982; Ueda
et al, 1979). The next most common findings are heart
defects, cataracts, low birthweight, hepatosplenomegaly,
and microcephaly. The triad of deafness, cataracts, and
congenital heart disease constitutes the classic syndrome.
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
In addition, systemic illness can occur and be characterized by purpura, hepatosplenomegaly, jaundice, pneumonia, and meningoencephalitis; 45% to 70% of infants have
cardiac lesions, including patent ductus arteriosus, peripheral pulmonic stenosis, and valve abnormalities (Reef et al,
2000; Schluter et al, 1998). Recently, an extensive review
of published reports of cardiovascular disease in the setting
of congenital rubella syndrome demonstrated that branch
pulmonary artery stenosis is the most commonly identified
isolated lesion (Oster et al, 2010). A variety of other signs
may be present. Additional ocular findings are pigmentary retinopathy, microphthalmia, and strabismus. The
skin lesions have been described as resembling a blueberry
muffin and represent extramedullary dermal hematopoiesis; an identical rash can be observed in congenital CMV
infection (Avram et al, 2007; Bowden et al, 1989; Brough
et al, 1967; Mehta et al, 2008).
The clinical manifestations of CRS vary to some extent
depending on the timing of fetal infection. In a prospective
study following pregnant women with confirmed rubella
infection by trimester, a full range of rubella-associated
defects (including congenital heart disease and deafness)
were observed in nine infants infected before the 11th
week. Thirty-five percent of infants (9 of 26) infected
between 13 and 16 weeks’ gestation had deafness alone
(Miller et al, 1982). Cataracts typically occur secondary
to maternal rubella infection occurring before day 60 of
pregnancy; heart disease is found almost exclusively when
maternal infection is before the 80th day (i.e., first trimester). Disease manifestations that may have their onset after
birth (late-onset disease) include: a generalized rash with
seborrheic features that may persist for weeks, acute or
chronic interstitial pneumonia, abnormal hearing resulting from presumed labyrinthitis, central auditory imperception, and progressive rubella panencephalitis (Franklin
and Kelley, 2001; Phelan and Campbell, 1969; Reef et al,
2000; Sever et al, 1985).
A higher than expected incidence of autoimmune diseases, such as thyroid disorders and diabetes mellitus, have
also been reported years after the diagnosis of congenital rubella (Forrest et al, 2002; Gale, 2008; McEvoy et al,
1988; Reef et al, 2000). Infants with late-onset disease have
demonstrated immunologic abnormalities, including dysgammaglobulinemia or hypogammaglobulinemia (Hancock et al, 1968; Hayes et al, 1967; Soothill et al, 1966).
Hyper-IgM syndrome has also been described in association with autoimmune disease in a child with CRS (Palacin
et al, 2007). Other studies have demonstrated dysfunction
of cellular immune responses in children with CRS (Fuccillo et al, 1974; South et al, 1975; Verder et al, 1986). The
pathogenesis of these rubella-related syndromes is poorly
understood. Psychiatric disturbances, including some with
features of autism spectrum disorders, have been observed
decades after CRS (Hwang and Chen, 2010).
LABORATORY EVALUATION
It is common obstetric practice to screen all pregnant
women for rubella antibodies, and because of the devastating consequences of CRS, this is a reasonable policy
to continue, even in the setting of eradication of endemic
rubella in the United States. Women who are exposed to
491
rubella should be screened for evidence of previous immunity; if none is found, they should be tested for rubella IgG
and IgM antibodies. A positive IgM titer or a rise in paired
IgG titers is indicative of recent infection. Women with
such findings should also be evaluated to try to determine
the likely gestational age at time of infection in order to
assess the potential risk to the fetus.
The laboratory diagnosis of congenital rubella can be
made definitively only during the 1st year of life, unless
the virus can be recovered later from an affected site, such
as the lens. Diagnosis can be made with any one of the following four criteria:
1. Positive anti-rubella IgM titer, preferably determined
with enzyme immunoassays, but indirect assays are
acceptable
2. A significant rise in rubella IgG titer between acute and
convalescent measurements 2 to 3 weeks apart or the
persistence of high titers longer than expected from
passive maternal antibody transfer
3. Isolation of rubella virus cultured from nasal, blood,
throat, urine, or CSF specimens (throat swabs have the
best yield)
4. Detection of virus by reverse transcriptase PCR in specimens from throat swabs, CSF, or cataracts obtained
from surgery (Centers for Disease Control and Prevention, 2001)
An infected infant can excrete the virus for many months
after birth despite the presence of neutralizing antibody
and, thus, may pose a hazard to susceptible individuals. In
rare cases the virus can be recovered after 1 year of age.
An exception to this rule is the cataract, in which the virus
can remain for as long as 3 years. In late-onset disease, the
virus can also be found in affected skin and lung.
Other laboratory findings are thrombocytopenia, hyperbilirubinemia, and leukopenia. Radiographic findings
include large anterior fontanel, linear areas of radiolucency
in the long bones (i.e., celery stalking), increased densities
in the metaphyses, and irregular provisional zones of calcification (Chapman, 1991; Reed, 1969). The radiographic
changes seen in rubella are not pathognomonic of the
disease, but resemble those seen in other congenital viral
infections, including congenital cytomegalovirus infection
(Alessandri et al, 1995).
TREATMENT AND PROGNOSIS
There is no specific therapy for congenital rubella. Initially the infant may need general supportive care, such as
administration of blood transfusion for anemia or active
bleeding, seizure control, and phototherapy for hyperbilirubinemia. Long-term care requires a multidisciplinary
approach consisting of occupational and physical therapy,
close neurologic and audiologic monitoring, and surgical interventions as needed for cardiac malformations and
cataracts.
The consequences of fetal rubella infection may not be
evident at birth. In one study of 123 infants with documented congenital rubella, 85% of cases were not diagnosed until after discharge from the nursery (Hardy, 1973).
Communication disorders, hearing defects, some mental
or motor retardation, and microcephaly by 1 to 3 years of
age were among the major problems that were discovered
492
PART IX Immunology and Infections
after the newborn period. A predisposition to inguinal
hernias was also noted. Longitudinal studies of somatic
growth show that most infants with congenital rubella
remain smaller than average throughout infancy, but grow
at a normal rate. Stunting of growth was more common
after rubella infection in the first 8 weeks of pregnancy
than after later infection. Even in the absence of mental
retardation, neuromuscular development is commonly
abnormal. A study of neurodevelopmental outcomes in 29
affected children without mental retardation found that
25 had other abnormalities; hearing loss, difficulties with
balance and gait, learning deficits, and behavioral disturbances were found in more than half of the affected children (Desmond et al, 1978).
PREVENTION
There is no effective antiviral therapy against congenital
rubella infection, so the most useful practice is to ensure
that women who are considering pregnancy are immune.
The Advisory Committee on Immunization Practices recommends screening of all pregnant women for rubella
immunity and postpartum vaccination of those who are
susceptible (Centers for Disease Control and Prevention,
2001). Immunity to rubella appears to confer almost complete protection against CRS. Rare cases of documented
subclinical maternal reinfection with rubella have been
reported (Morgan-Capner et al, 1991; Saule et al, 1988).
In rare cases, maternal reinfection can lead to CRS (Banerji
et al, 2005). Approximately 20 cases of CRS after maternal
reinfection have been reported in the literature, and none
have caused symptomatic CRS when the known reinfection
occurred after 12 weeks’ gestation (Bullens et al, 2000).
Live attenuated rubella virus vaccine is safe and effective, although the duration of immunity is uncertain. It is
typically administered in the United States in a trivalent
formulation in combination with measles-mumps-rubella
vaccine or in a quadrivalent combination with measlesmumps-rubella-varicella vaccine. The vaccine is recommended for children at 12 to 15 months of age and at 4
to 5 years of age. It is also recommended for women of
childbearing age in whom results of both a hemagglutination inhibition antibody test and a pregnancy test are
negative. Although no cases of symptomatic congenital
rubella infection have been reported as a consequence
of vaccination during pregnancy in the more than 500
cases monitored, vaccination is not recommended during
pregnancy because of the theoretical hazard to the fetus
(Josefson, 2001; Nasiri et al, 2009; Tookey, 2001). A mild
rubella-like illness is sometimes seen after immunization,
with arthralgia occurring 10 days to 3 weeks after injection. If a woman is found to be susceptible, vaccine should
be administered during the immediate postpartum period
before discharge. Breastfeeding is not a contraindication
to postpartum immunization. Immunization in the postpartum period has rarely produced polyarticular arthritis,
neurologic symptoms, and chronic rubella viremia (Tingle
et al, 1985).
The problem of management of the pregnant woman
who is exposed to rubella or who contracts the disease
should be resolved after the known risks are weighed.
If serum antibody is detectable at the time of exposure,
the fetus is probably protected. If no antibody is detectable, additional serum samples at 2 to 3 weeks after exposure and again at 4 to 6 weeks after exposure should be
obtained. These samples can be run concurrently with the
first serum to ascertain whether infection has occurred
(i.e., seroconversion). There is no evidence that administration of immunoglobulin prevents rubella infection or
viremia. Moreover, administering immunoglobulin may
provide an unwarranted sense of security, because infants
with congenital rubella have been born to women who
received immunoglobulin shortly after exposure. Regardless of these limitations, administration of immunoglobulin may be considered if the pregnancy is not terminated.
Immunoglobulin can reduce the likelihood of fetal infection, but will not eliminate the risk; therefore the CDC
does not routinely recommend the use of immunoglobulin
in a pregnant woman for postexposure prophylaxis unless
she does not wish to terminate the pregnancy under any
circumstances (Centers for Disease Control and Prevention, 2001). Decisions about the termination of pregnancy
should be made only after maternal infection has been
proved and should also account for the risk of rubella-associated damage to the fetus, which is highest when maternal
infection occurs during the first 8 weeks of pregnancy.
LYMPHOCYTIC CHORIOMENINGITIS
VIRUS
Lymphocytic choriomeningitis virus (LCMV) is a member of the family Arenaviridae. Rodents are the primary
reservoir, particularly mice and hamsters. Like other arenaviruses, LCMV has a bisegmented negative-strand RNA
genome. The S segment encodes for the virus nucleoprotein and glycoprotein, whereas the L segment encodes
for the virus polymerase (L) and Z protein. Susceptible
rodents are infected asymptomatically in utero, can harbor chronic infection, and excrete virus in urine, feces,
saliva, nasal secretions, milk, and semen for life. Typically
mice remain asymptomatic, although hamsters may demonstrate viremia and viruria with variable symptoms (Jahrling and Peters, 1992). Sequelae of human exposure to
LCMV range from asymptomatic infection to nonspecific,
flulike symptoms; a proportion of infections have neurologic manifestations. LCMV was first described as a cause
of congenital infection in England in 1955 in a 12-dayold infant (Komrower et al, 1955) and later in the United
States (Barton and Mets, 2001; Barton et al, 1993, 2001;
Larsen et al, 1993). Because LCMV has only recently been
recognized as a source of congenital infection, it is likely
underdiagnosed (Jamieson et al, 2006).
EPIDEMIOLOGY
Human seroprevalence ranges between less than 1% and
10% worldwide and varies extensively with geographic
region (Ambrosio et al, 1994; Childs et al, 1991; Marrie
and Saron, 1998; Stephensen et al, 1992). One study noted
a higher prevalence in women (Marrie and Saron, 1998).
Lower socioeconomic status and older age are associated
with higher seroprevalence. Studies conducted in the
1940s through the 1970s found that approximately 8% to
11% of cases of aseptic meningitis and encephalitis were
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
associated with LCMV infection (Meyer et al, 1960; Park
et al, 1997b). In temperate climates, human exposure is
more common during the fall and winter, when rodents
move indoors. Outbreaks have been reported in laboratory personnel working with hamsters and mice (Dykewicz
et al, 1992; Hinman et al, 1975; Vanzee et al, 1975).
Multiple outbreaks associated with pet hamsters have also
been reported in the United States (Biggar et al, 1975;
Maetz et al, 1976) and Europe (Brouqui et al, 1995; Deibel
et al, 1975); however, congenital infection is relatively
rare. A total of 54 cases have been diagnosed worldwide
since the first case in 1955, and 27 of those occurred in
the United States (Barton and Mets, 2001; Greenhow and
Weintrub, 2003). There has been an increased recognition of congenital LCMV in recent years; 34 of the cases
described in a review were reported since 1993 (Jamieson
et al, 2006). The true frequency of congenital LCMV
infection is unknown, because there is no active surveillance. As with other congenital infections, there may be
a wide spectrum of disease, including asymptomatic and
subclinical or nonspecific infections.
PATHOGENESIS
Humans acquire LCMV infection from aerosolized particles, bites, or fomite contact with virus excreted from
rodents (Jahrling and Peters, 1992). Human-to-human
horizontal transmission by organ transplantation has
been documented (Fischer et al, 2006). In this report,
abdominal pain, altered mental status, thrombocytopenia, elevated aminotransferase levels, coagulopathy, graft
dysfunction, renal failure, seizures, and either fever or
leukocytosis were variably present within 3 weeks after
transplantation. Seven of the eight recipients died, 9
to 76 days after transplantation. The pathogenesis of
LCMV infection is poorly understood, although it is
likely an immunopathologic process mediated by the
host CD8+ T-cell response (Craighead, 2000). It is also
postulated that the high rate of spontaneous mutations
that arise during LCMV replication allows both variability in pathogenicity and a mechanism of escape from
humoral response during the initial phase of infection
(Ciurea et al, 2001).
Like other arenaviruses, LCMV replicates either at the
site of infection or in corresponding lymph nodes; this
localized replication is followed by viremia. It is thought
that during the viremic stage, the virus travels to parenchymal organs and the CNS. Pathologic findings include
lymphocytic infiltration and extramedullary hematopoiesis. In two congenitally infected infants for whom
neuropathology results were available, cerebromalacia,
glial proliferation, and perivascular edema were reported
(Barton et al, 1993). In adult mice inoculated intracranially with LCMV, subsequent viral proliferation in the
ependyma, meninges, or both have been described, with
injury mediated by CD8+ T cells (Doherty et al, 1990).
This viral infiltration, along with the host inflammatory
response, can lead to aqueductal stenosis and subsequent
hydrocephalus. Studies in a murine model suggest that
cytokine-chemokine cascades mediate much of the neuropathology observed in the setting of CNS infection
(Christensen et al, 2009).
493
CLINICAL SPECTRUM
It is estimated that asymptomatic or mild LCMV infections occur in approximately one third of patients infected;
however, the classic presentation of LCMV infection is
a nonspecific, flulike, or mononucleosis-like illness that
is often biphasic. Symptoms are fever, malaise, nausea,
vomiting, myalgias, headache, photophobia, pharyngitis,
cough, and adenopathy. After defervescence and resolution of these constitutional symptoms, a second phase of
CNS disease may develop. Neurologic manifestations
occur in approximately one fourth of infectious episodes
and vary from aseptic meningitis to meningoencephalitis.
Transverse myelitis, Guillain-Barré syndrome, and deafness have also been reported. Other manifestations are
pneumonitis, arthritis, myocarditis, parotitis, and dermatitis. Recovery may take months, but usually occurs without
sequelae (Craighead, 2000).
When LCMV infection occurs during pregnancy,
maternal symptoms typically appear during the first and
second trimesters, but only 50% to 60% of mothers of
infants diagnosed with LCMV congenital infection recall
having symptoms (Wright et al, 1997). Known maternal exposure to rodents is reported in approximately one
fourth to one half of cases (Barton and Mets, 2001). Usually, exposed women are from rural settings, come from
lower socioeconomic settings with substandard housing
conditions, or have pet rodents in the home (i.e., hamsters).
The complete spectrum of disease secondary to congenital LCMV infection is still uncertain, although
chorioretinitis and hydrocephalus are the predominant
characteristics reported among children diagnosed with
congenital LCMV infection. Chorioretinitis is present in
more than 90% of cases. Other ocular findings are chorioretinal scars, optic atrophy (usually bilateral), nystagmus,
esotropia, exotropia, leukocoria, cataracts, and microphthalmia (Barton and Mets, 2001; Brezin et al, 2000; Enders
et al, 1999). Some ophthalmologic findings resemble those
described in the lacunar retinopathy of Aicardi syndrome
(Wright et al, 1997), a finding of interest insofar as Aicardi
syndrome can, like congenital CMV or LCMV infection,
produce intracranial calcifications. A wide range of neurologic defects are described, including microencephaly,
encephalomalacia, chorioretinitis, porencephalic cysts,
neuronal migration disturbances, periventricular infection,
and cerebellar hypoplasia (Bonthius et al, 2007). Congenital LCMV should be considered in the differential diagnosis of neonatal hydrocephalus (Schulte et al, 2006).
Most infants are born at term, and birthweights are generally appropriate or large for gestational age. Thirty-five
percent to 40% of infants reported with congenital LCMV
have had microcephaly or macrocephaly at birth (Barton
and Mets, 2001). Systemic symptoms are rare, although
hepatosplenomegaly and jaundice have been noted. Other
individual case report findings are pes valgus, dermatologic
findings consistent with staphylococcal scalded-skin syndrome (Wright et al, 1997), spontaneous abortion (Biggar
et al, 1975), and intrauterine demise secondary to hydrops
fetalis (Enders et al, 1999; Meritet et al, 2009). It is important to note that, because systemic symptoms are typically
minimal at birth, the diagnosis of congenital LCMV infection often is not considered until an affected infant is a few
494
PART IX Immunology and Infections
months of age, when microcephaly, macrocephaly, visual
loss, or developmental delay may be noted.
LABORATORY EVALUATION
Serology is the most reliable and feasible method to diagnose LCMV. In most reports, the diagnosis was established by testing of the infant’s serum, CSF, or both; in
some, maternal serum testing was the key. Testing all
three fluids provides the most information. Because of the
low baseline population seroprevalence, positive titers for
LCMV are much more useful for diagnosis than detection
of antibodies to microbes such as CMV and toxoplasmosis. There is a commercially available immunofluorescent
antibody test that detects both IgM and IgG for LCMV.
It has better sensitivity than the complement fixation
and neutralizing antibody tests that also have been used
(Lehmann-Grube et al, 1979; Lewis et al, 1975). Complement fixation titers generally do not rise until more
than 10 days after onset of infection, but immunofluorescent antibody results may be positive within the first
few days of illness (Deibel et al, 1975). The CDC also has
an enzyme-linked immunosorbent assay test for IgM and
IgG; it may be more useful for diagnosis in an older child
because it can detect increased IgG later than and persistent IgG for longer than the immunofluorescent antibody
test. Some studies have found antibody as late as 30 years
after suspected exposure. Virus can be cultured in Vero
cell lines or inoculated into newborn mice, but use of
these methods is uncommon. Reverse transcriptase–PCR
has been used in serum and CSF to diagnosis LCMV and
as a surveillance tool; it may become more available in the
future (Enders et al, 1999; McCausland and Crotty, 2008;
Park et al, 1997a).
Information about routine laboratory data in patients
with a diagnosis of congenital LCMV infection is minimal, but thrombocytopenia and hyperbilirubinemia have
been reported. CSF findings are variable. Up to one half
of cases demonstrate a mild increase in white blood cell
count (up to 64 cells/μL in one case series of 18 infants),
the serum protein concentration may be normal or mildly
elevated, and the serum glucose concentration may be
normal or mildly decreased (Wright et al, 1997). Among
infants reported in whom neuroradiographic imaging was
performed, 89% (17 of 19) had hydrocephalus or periventricular calcifications. Flattened gyri, lissencephaly,
and schizencephaly have been reported (Barton and Mets,
2001), compatible with a role for LCMV in fetal neuronal
migration defects (Bonthius et al, 2007).
TREATMENT
Ribavirin has been used for management of other arenavirus infections and inhibits LCMV growth in vitro (Géssner and Lother, 1989). Although novel approaches are
being used to develop antivirals against LCMV and other
pathogenic arenaviruses (de la Torre, 2008), there are currently no recommendations for the use of antiviral agents
against these viruses. In the outbreak associated with solid
organ transplantation, one affected recipient received ribavirin and reduced levels of immunosuppressive therapy
and survived (Fischer et al, 2006).
PROGNOSIS
Because congenital LCMV infections have been recognized relatively recently and the existing data come from
case reports, there may be a wider spectrum of disease
than is currently appreciated. The proportion of asymptomatic infected infants is unknown. For the 25 infant
cases reported, estimated mortality rates are 16% to 35%
between birth and 21 months of age (Barton and Mets,
2001; Wright et al, 1997). Among infants who survived,
84% (32 of 38) have neurologic sequelae, including spastic quadriparesis, mental retardation, developmental delay,
seizures, and visual loss (Barton and Mets, 2001). Sensorineural hearing loss is less common and has been reported
in only two infants (Barton and Mets, 2001; Wright
et al, 1997). Some ophthalmologists suggest that among
patients with developmental delay and visual loss consistent with chorioretinitis, LCMV congenital infection may
be underdiagnosed. A study in Chicago of prospectively
diagnosed patients with chorioretinitis and patients in a
home for severely mentally retarded children with chorioretinal scars found six children with elevated LCMV titers
and normal toxoplasmosis, CMV, rubella, and HSV titers
(Mets et al, 2000). Two children with chorioretinal scars
and elevated LCMV titers have been reported in France
(Brezin et al, 2000).
PREVENTION
Public health officials and clinicians should be aware that
(1) wild, laboratory, and pet rodent exposure can lead to
intrauterine infection with LCMV virus and (2) congenital infection has been associated with potentially devastating ophthalmologic and neurologic sequelae. Pregnant
women need to be educated about the potential risks of
exposure to infected rodent excreta and instructed to avoid
rodents and rodent droppings whenever possible. Obstetricians and neonatologists should seek a history of pet or
wild rodent exposure for counseling purposes and to aid in
the evaluation of infants with unexplained CNS pathologies. The diagnosis of LCMV should be considered in all
unexplained cases of infant hydrocephalus. There are currently no vaccines approved by the U.S. Food and Drug
Administration (FDA) for the prevention of arenavirus
disease, although candidate multivalent arenavirus vaccines capable of providing T cell–mediated protection
against a variety of pathogenic arenaviruses are currently
in development (Botten et al, 2010).
ENTEROVIRUSES
Enteroviruses are single-stranded, positive-sense RNA
viruses. These viruses belong to the family Picornaviridae
(pico means very small in Spanish). The enteroviruses of
humans include polioviruses 1, 2, and 3, coxsackieviruses
A and B (named after Coxsackie, New York the city where
these viruses were first identified and characterized), and
the echoviruses (echo is an acronym for enteric cytopathic
human orphan). Of these, poliovirus infection has historically been responsible for the greatest morbidity in infants.
Severe, often fatal poliovirus disease used to occur with
great frequency in infants infected in the perinatal period,
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
with a high incidence of residual paralysis in survivors
(Bates, 1955; Cherry, 2005). Fortunately, poliovirus infections have become rare in the developed world, because
of the widespread implementation of effective immunizations. However, neonatal diseases associated with coxsackieviruses and echoviruses, for which there are no vaccines,
remain common and can be associated with serious morbidity and occasional mortality. Typically acquired from
a maternal source, these agents are associated with a wide
range of clinical syndromes in the neonatal intensive care
unit, including CNS infection, myocarditis, and a sepsislike syndrome. Enteroviruses are also responsible for a
large number of hospital readmissions for evaluation of
febrile syndromes in infants younger than 2 months.
EPIDEMIOLOGY
Enterovirus infections are seasonal, occurring most commonly during the summer and autumn in temperate
climates. The incidence varies from year to year, with
outbreaks sometimes caused by a single coxsackievirus
or echovirus serotype and sometimes by several (Sawyer
et al, 1994). In older children, enteroviruses are transmitted by the fecal-oral route and are typically associated with a variety of febrile syndromes, including febrile
exanthematous syndromes, aseptic meningitis, pneumonia
with or without pleural effusion, and myocarditis. Disease
in newborn infants is relatively uncommon, but reflects the
frequency of infection in the general population (Krajden
and Middleton, 1983). Enteroviruses may also be associated with nosocomial outbreaks. Nursery and obstetric
clinic outbreaks of both coxsackievirus B (Bhambhani et al,
2007; Brightman et al, 1966; Rantakallio et al, 1970) and
echovirus infections (Chen et al, 2005; Jankovic et al, 1999;
Nagington et al, 1978) have been reported and associated
with severe, and sometimes fatal, illnesses.
Enteroviral infections account for at least one third of
neonatal febrile admissions for suspected sepsis and for
between half and two thirds of all admissions during peak
enteroviral season (Byington et al, 1999; Dagan, 1996).
Neonatal aseptic meningitis is also frequently caused by
enteroviral infections. In a review of neonatal meningitis
seen over a 15-year period in Galveston, Texas, enterovirus was the most common cause of meningitis in newborn
infants older than 7 days of age (Shattuck and Chonmaitree, 1992). Enteroviruses, along with other viruses, have
been implicated as a potential cause of sudden infant death
syndrome (SIDS), possibly from myocarditis or pulmonary infection (Grangeot-Keros et al, 1996; Shimizu
et al, 1995), but this association has been controversial. In
one study of SIDS victims, a comprehensive assessment
was undertaken to attempt to identify potential viral infection of the myocardium. Overall, 62 SIDS victims and 11
controls were studied. Enteroviruses were detected in 14
(22.5%), adenoviruses in 2 (3.2%), Epstein–Barr viruses in
3 (4.8%), and parvovirus B19 in 7 (11.2%) cases, whereas
control group samples were completely negative for viral
nucleic acid (Dettmeyer et al, 2004). However, an evaluation of histopathologic features and PCR analysis from 24
SIDS cases failed to demonstrate any association with viral
infection (Krous et al, 2009), leaving this putative association unclear.
495
ETIOLOGY AND PATHOGENESIS
Neonates can acquire enteroviral infections secondary to in utero transmission, intrapartum transmission
during labor and delivery, or postnatally. Intrauterine
infections appear to occur via transplacental spread, secondary to maternal viremia, and this mode of transmission appears to be responsible for up to 22% of cases of
neonatal enteroviral infection (Kaplan et al, 1983; Modlin,
1986). Enteroviruses have been implicated as a cause of
fetal demise (Johansson et al, 1992; Konstantinidou et al,
2007; Nielsen et al, 1988). Intrapartum or postnatal transmission is more common than transplacental transmission.
The dominant mode of transmission of serious neonatal
infection is through contact with maternal blood, fecal
material, or vaginal or cervical secretions, most likely during or shortly after delivery (Hawkes and Vaudry, 2005;
Jenista et al, 1984). After acquisition of infection, viremia
ensues in the infant, leading to a variety of end-organ diseases. Virus strain or serotype appears to be a factor in
predicting the severity of illness, as does the quantity of
the inoculum. The presence or absence of transplacental
maternally derived antibody also dictates the severity of
disease in the infected infant. Severe disease occurs in the
absence of type-specific antibody, a scenario that is more
likely if the maternal infection is acquired near the time of
delivery. Breastfeeding appears to provide a relative degree
of protection against acquisition of infection in neonates
(Sadeharju et al, 2007).
One of the most comprehensive analyses of the incidence of neonatal enterovirus disease came from a prospective study in Rochester, N.Y. This study demonstrated
that approximately 13% of all newborns tested positive for
enterovirus from throat or stool cultures during a typical
season (June to October; Jenista et al, 1984). Although
this result represented a remarkably high incidence, 79%
of these infections were asymptomatic. The most common clinical findings in the 21% of symptomatic infections were lethargy and fever. These infants were typically
admitted to hospital for an evaluation to rule out sepsis,
making enterovirus infection a more common reason for
hospitalization owing to an infectious disease than group B
streptococcus, herpes simplex virus, and cytomegalovirus
infections combined.
It appears that any of the non–polio enteroviruses can
cause disease in the newborn infant. A variety of clinical
syndromes are associated with certain enteroviruses. A retrospective chart review of 24 neonatal enteroviral infections in Toronto, Canada, found that 10 infants died, 12
had aseptic meningitis, and 5 had myocarditis (Krajden and
Middleton, 1983). Of the 24 isolates, 7 were echovirus, 15
were coxsackievirus B, one was coxsackievirus A, and one
was nontypable. In infants requiring hospitalization with
acute enterovirus disease, coxsackievirus B is associated
primarily with myocarditis and aseptic meningitis (Kibrick
and Benirschke, 1958). Recent reports have identified coxsackievirus B1 as an emerging cause of life-threatening
myocarditis and other severe, fatal syndromes in neonates
(Verma et al, 2009; Wikswo et al, 2009). Echoviruses
are associated with severe nonspecific febrile illnesses
with disseminated intravascular coagulation (Nagington
et al, 1978), aseptic meningitis (Cramblett et al, 1973), or
496
PART IX Immunology and Infections
hepatitis (Modlin, 1980). With both coxsackievirus and
echovirus, nonspecific febrile illnesses, with or without
an exanthem, are commonly observed. Enterovirus 71 is
particularly notable for its etiologic role in epidemics of
severe neurologic diseases in children (Chen et al, 2010).
Nursery-based outbreaks of this neurovirulent enterovirus
have also been described (Huang et al, 2010).
Except for transplacental infections, the portal of entry
for enterovirus is via the oral or respiratory route. After
replication in the pharynx and the gastrointestinal tract,
virus seeds the tonsils, cervical and mesenteric nodes, and
Peyer’s patches. The pathogenesis of enterovirus disease
stems from the ensuing viremia, which can lead to infection of the heart, CNS, liver, pancreas, adrenal glands, skin,
mucous membranes, and respiratory tract. This feature of
enterovirus replication and dissemination helps to explain
the diverse disease manifestations that may be observed
following infection. The humoral immune response is
associated with recovery from systemic and end-organ
infection, although virus may continue to be shed in the
stool for several weeks. Failure to clear enteroviral infection, particularly from the CNS, should suggest an underlying humoral immunodeficiency, although this would not
typically be observed in the neonatal period, when maternally derived Ig is present in the newborn (McKinney
et al, 1987; Misbah et al, 1992). Depending on the extent of
disease, extensive end-organ pathology may be observed.
In coxsackievirus B infections, myocardial necrosis, and
inflammation may be seen that are patchy or diffuse, with
extensive infiltration by lymphocytes, mononuclear cells,
histiocytes, and polymorphonuclear leukocytes. Similar
infiltrates are seen in the meninges in both coxsackievirus
and echovirus aseptic meningitis. Brainstem encephalitis
can be observed with enterovirus 71 infection, often in
association with pulmonary edema (Wang and Liu, 2009).
When liver or adrenal glands are involved, there is usually
extensive hemorrhage as well as inflammation and necrosis; such fatal fulminant infections are often associated
with echovirus 11 (Mostoufizadeh et al, 1983).
CLINICAL SPECTRUM
When neonatal disease is acquired vertically from the
mother, the infant is typically asymptomatic at birth,
although premature delivery is more common. The
mother may be febrile at this time or may have a history of
recent high fevers and gastrointestinal symptoms. Fever,
anorexia, and vomiting develop in the baby after an incubation period of 1 to 5 days. The onset of illness occurs in
the first week of life in more than 50% of affected infants
(Krajden and Middleton, 1983). At that point, the clinical evolution depends on the infecting virus and the extent
of end-organ involvement. In most instances, the disease
is mild and self-limited. Symptomatic infections may be
characterized by rash, aseptic meningitis, hepatitis, and
pneumonia. A review of 29 infants younger than 2 weeks
with enteroviral infections reported that 5 of 29 infants
had severe multisystem disease, and all survived (Abzug
et al, 1993). In another study in Salt Lake City, 1779 febrile
infants younger than 90 days and undergoing evaluation
for sepsis were enrolled; 1061 had enterovirus testing, and
214 (20%) were enterovirus positive (57% from blood, and
74% from CSF). The mean age of infants with enterovirus infection was 33 days; 91% were admitted, and 2%
required intensive care (Rittichier et al, 2005).
These observations underscore the generally benign
and self-limited nature of neonatal enterovirus disease;
however, morbidity can be substantial in severe disseminated disease. A viral sepsis syndrome—characterized by
disseminated intravascular coagulation, refractory hypotension, and death—may occur in the setting of severe
disease. Typically these severe infections are acquired in
the immediate perinatal period. The mother is commonly
symptomatic and may have been empirically treated with
broad-spectrum antibiotics for possible chorioamnionitis.
A maternal history of suspected chorioamnionitis in the
absence of positive bacterial cultures should suggest this
association, particularly during the typical “enterovirus
season” observed in temperate climates. If myocarditis
is present, congestive heart failure is often severe. Some
infections, particularly those with echoviruses, are characterized by a rampant and overwhelming hepatitis (Modlin,
1980). Others exhibit primarily pulmonary disease or gastrointestinal involvement including diarrhea and necrotizing enterocolitis (Lake et al, 1976). Intracranial bleeding
ranging from small to massive, severe hemorrhage has
also been reported as a complication of neonatal enteroviral infection (Abzug, 2001; Abzug and Johnson, 2000;
Swiatek, 1997). Other rarely associated findings include
disseminated vesicular rash, dermal hematopoiesis, and
hemophagocytic syndrome (Barre et al, 1998; Bowden
et al, 1989; Sauerbrei et al, 2000). The severity of CNS
infection is similarly variable. Enteroviruses, particularly
enterovirus 71, can produce overwhelming meningoencephalitis, sometimes with cranial nerve signs. It is more
common, however, to see moderate or mild meningitis
characterized by temporary irritability, lethargy, fever, and
feeding difficulty.
Acquired postnatal enteroviral disease in infants is
characterized primarily by high fever, irritability, lethargy, or poor feeding. One fourth of infected infants
develop diarrhea or vomiting with or without an erythematous maculopapular rash. Conjunctivitis has also
been observed. Respiratory tract symptoms are less common (Dagan, 1996). As noted, there is significant overlap
between enteroviral and bacterial neonatal infections, and
the two syndromes are difficult to distinguish; therefore
many febrile infants will be admitted to the hospital and
treated with broad-spectrum antibiotics and, if the CNS
is involved, with acyclovir for possible bacterial sepsis and
neonatal HSV infection. Such treatment seems unavoidable, except in circumstances in which enterovirus infection can be diagnosed rapidly and definitively. Duration of
illness varies from less than 24 hours to longer than 7 days,
but generally lasts 3 to 4 days.
LABORATORY EVALUATION
Viral culture from stool or rectal swab, nasopharyngeal
swab, blood, buffy coat, urine, or CSF represents the
gold standard diagnosis. Stool or rectal swab cultures can
remain positive for several weeks following the initial
infection, underscoring the importance of fecal-oral transmission in the epidemiology of these infections. Recently,
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
reverse transcriptase PCR assays have become standardized and are available from many commercial and reference laboratories. Serologic tests for enteroviruses have
been reported, but are less useful than culture and PCR
(Swanink et al, 1993).
Laboratory evaluation is predicated on the clinical syndrome and the end organs involved. Infants with aseptic meningitis typically have moderate CSF pleocytosis,
which can be either lymphocytic or polymorphonuclear,
but may lack pleocytosis even in the setting of documented CNS infection (Seiden et al, 2010). Accordingly,
during periods of active enterovirus infection in the community, CSF should be sent for PCR even if a pleocytosis
is absent. Thrombocytopenia, elevated transaminase levels, hyperbilirubinemia, hyperammonemia, hematologic
abnormalities consistent with disseminated intravascular
coagulation, anemia, peripheral leukocytosis, and abnormal chest radiographs are among other potential laboratory findings. When myocarditis is a diagnostic possibility,
echocardiogram and electrocardiogram are indicated and
may reveal diminished LV function, or dysrhythmias.
Liver biopsy may be warranted in cases of fulminant
hepatic failure (Abzug, 2001).
TREATMENT
The cornerstone of treatment of neonatal enteroviral disease is supportive care. Myocarditis and heart failure can
be treated with inotropic support, diuretics, aggressive
fluid management, and other supportive measures. Disseminated intravascular coagulation should be treated with
blood products and other supportive measures as indicated.
There is no evidence that steroids are of benefit. IVIG has
been reported anecdotally to treat neonatal enteroviral
infections with varying degrees of success (Kimura et al,
1999; Valduss et al, 1993). Only one randomized trial has
systematically studied its use in 16 neonates with severe
enteroviral infection; nine of these infants were randomized to receive IVIG, at a dose of 750 mg/kg. Decreased
viremia and viruria along with faster resolution of irritability, jaundice, and diarrhea was demonstrated in patients
administered IVIG with high titers of neutralizing enteroviral-specific antibodies. However, there were no significant differences in other major clinical outcomes, such as
duration of hospitalization, fever, and symptoms of acute
illness between treatment and control groups (Abzug et al,
1995). To attempt to augment the enterovirus type–specific antibody level in the setting of symptomatic neonatal
disease, maternal plasma transfusion has been attempted
(Jantausch et al, 1995; Rentz et al, 2006), based on the
rationale that neonates typically acquire infection from
their mothers in the peripartum period. Although reports
of success using IVIG for treatment of neonatal enterovirus infections is largely based on anecdotal reports in
limited numbers of patients, its use should be considered
for severely symptomatic infections with life-threatening
end-organ disease.
The antiviral drug pleconaril has been developed specifically to treat picornavirus infections (enteroviruses
and rhinoviruses). A small case series of infants with
severe enteroviral hepatitis suggested a beneficial effect
(Aradottir et al, 2001). Another small case series indicated
497
that five of six neonates with severe enteroviral infection
who were treated with pleconaril survived, with minimal
or no sequelae (Rotbart and Webster, 2001). However,
a multicenter study of pleconaril treatment for enteroviral meningitis in children younger than 12 months
conducted by the CASG and sponsored by the National
Institutes of Health demonstrated no significant differences in duration of positivity by culture or PCR, hospitalization, or symptoms comparing the treatment and
placebo groups (Abzug et al, 2003). Pleconaril did appear
to have an effect on enteroviral meningitis in adults, promoting more rapid resolution of illness (Desmond et al,
2006), but the role of this drug in infants and children
remains undefined. The drug continues to be investigational and is only available through the CASG, which is
currently comparing pleconaril with placebo in severe
symptomatic neonatal enteroviral infections (www.clini
caltrials.gov).
SHORT- AND LONG-TERM PROGNOSIS
Prognostic factors for severe neonatal disease include peripartum maternal illness, earlier age of onset of neonatal
disease, absence of serotype-specific antibody, and absence
of fever and irritability (Abzug et al, 1993). All these
risk factors are most consistent with vertical intrauterine enteroviral infection rather than postnatally acquired
infection. The highest mortality rates are associated with
the combination of severe hepatitis, coagulopathy, and
myocarditis. Severe hepatitis caused by enteroviral infection is associated with mortality rates ranging from 30%
to 80% (Abzug, 2001; Modlin, 1986). By the time disseminated intravascular coagulation has developed, the prognosis is grave. Prothrombin time longer than 30 seconds
was a risk factor for death in one retrospective case review
(Abzug, 2001).
Few long-term follow-up studies have been published,
but the available information suggests that infants who
survive severe enteroviral neonatal disease have a complete recovery in most instances. Outcomes of 6 of 11
survivors with follow-up ranging from 9 to 48 months
reported normal growth and no residual medical problems or liver dysfunction (Abzug, 2001). The long-term
prognosis following CNS infection is unclear. A number
of early studies of infants younger than 3 months with
aseptic meningitis suggested that there may be some
impairment of intellectual development in comparison
with carefully selected control groups (Farmer et al, 1975;
Sells et al, 1975). However, in a series of nine children
with enteroviral meningitis and nine matched controls
evaluated for sequelae at approximately 4 years of age, no
differences in mean intelligence quotient, head circumference, detectable sensorineural hearing loss, or intellectual functioning were detected. Receptive language
functioning of the meningitis group was significantly less
than that in control subjects (Wilfert et al, 1981). A similar case-control follow-up study of 33 subjects, who were
compared with siblings used as controls, reported no neurodevelopmental sequelae (Bergman et al, 1987). Older
children with enterovirus 71 infection involving the CNS
are at risk for significant neurodevelopmental sequelae
(Chang et al, 2007).
498
PART IX Immunology and Infections
PREVENTION
Anecdotal reports of the use of IVIG in nursery outbreaks to
prevent further horizontal transmission of enteroviral infection produce conflicting results (Carolane et al, 1985; Kinney et al, 1986; Nagington et al, 1983). Because there are
multiple non–polio enteroviral serotypes that cause clinical
disease, development of anti-enteroviral immunization is
conceptually difficult, although vaccines are in development
for enterovirus 71 (Zhang et al, 2010). Standard contact
precautions should be used for the treatment of hospitalized
infants with known or suspected enteroviral infections.
HUMAN PARECHOVIRUS
Recently two viruses formerly classified with the echoviruses, echovirus 22 and 23, were shown to comprise a
separate genus within the Picornaviridae family, the genus
Parechovirus; these viruses are now referred to as human
parechoviruses types 1 and 2. A total of 10 distinct parechoviruses have now been recognized (Drexler et al, 2009;
Harvala et al, 2010; Pajkrt et al, 2009). These viruses have
been recognized recently as significant neonatal pathogens
(Harvala et al, 2010). The primary site of parechovirus replication is believed to be the respiratory and gastrointestinal
tract. Replication in the gastrointestinal tract is associated
with prolonged shedding of infectious virus in the feces.
As a result, fecal-oral transmission, as with enteroviruses,
appears to be the predominant route of infection. Parechovirus infections may also be acquired via a respiratory
route, with subsequent virus shedding detectable in respiratory secretions. Viremia leads to secondary seeding of
other organs and systemic symptoms. Neonates with parechovirus infection, particularly parechovirus 3, have a clinical presentation similar to infants with severe enteroviral
disease, with a sepsislike illness in severe cases (VerboonMaciolek et al, 2008a; Wolthers et al, 2008). Hepatitis is
also a prominent feature of neonatal infection. The most
frequent signs are fever, seizures, irritability, rash, and
feeding problems. It has been postulated that parechovirus 3 is a newly emerging infection, and the relative lack
of maternal antibody may predispose the neonate to more
severe disease (Harvala et al, 2010). Parechovirus can also
cause aseptic meningitis and encephalitis in the neonate,
with the predominate site of injury being the periventricular white matter (Gupta et al, 2010; Levorson et al, 2009;
Verboon-Maciolek et al, 2008b). Parechovirus RNA has
also been found at autopsy in children younger than 2 years
(Sedmak et al, 2010), including from infants with otherwise
unexplained deaths. Management is similar to that for neonatal enterovirus disease—namely, supportive care.
The other virus classified in the Parechovirus genus
is Ljungan virus (Tolf et al, 2009). This virus was first
described in 1998 and was found to cause type 1 diabetes–like symptoms and myocarditis in bank voles; it also
appears to be endemic in other wild rodent populations.
The virus does not appear to have a role in the development of diabetes in humans (Tapia et al, 2010). This zoonotic virus has recently emerged as a recognized cause of
fetal infection (Niklasson et al, 2007, 2009b). Ljungan
virus infection appears in particular to be associated with
severe CNS abnormalities, including hydrancephaly and
hydrocephalus (Niklasson et al, 2009a). A study from Sweden identified a strong epidemiologic association between
small rodent abundance and the incidence of intrauterine
fetal death in humans. Ljungan virus antigen was detected
in this study in half of the intrauterine fetal death cases
tested (Niklasson et al, 2009).
HEPATITIS VIRUSES
There are six distinct viruses known to cause viral hepatitis—hepatitis A, B, C, D (delta agent), E, and G. Hepatitis G is of importance for the pregnant patient, although
only B and C are of major importance in the newborn. The main features of each virus type are listed in
Table 37-3 (Jonas, 2000; Koff, 2007; Krugman, 1992).
Hepatitis A virus (HAV) is passed by fecal-oral transmission and is a rare cause of neonatal disease, although
it has been nosocomially transmitted in the setting of a
neonatal intensive care unit. Hepatitis E virus is similar to
hepatitis A in its mode of transmission and clinical manifestations, except for an increased mortality in pregnant
women infected with hepatitis E (Aggarwal et al, 2009;
Teshale et al, 2010). There are no data regarding perinatal
transmission of hepatitis E. Hepatitis D virus may cause
only coinfection or super-infection with hepatitis B virus.
Its only clinical significance is that hepatitis B infection
may become more severe when hepatitis D virus is present. Perinatal transmission has been described (Ramia and
Bahakim, 1998), but is uncommon.
Hepatitis G virus (HGV), also known as GB virus type C,
has been associated with acute and chronic hepatitis and
usually is noted as a coinfection with hepatitis B or C. This
virus has a tropism for lymphocytes and may influence
the course and prognosis in HIV-seropositive patients
(Reshetnyak et al, 2008). Perinatal transmission can occur
in 60% to 80% of infants born to HGV-viremic mothers, but there has not been any report of clinical hepatitis
attributed to the HGV infection in this setting (Ohto et al,
2000; Wejstal et al, 1999; Zanetti et al, 1998; Zuin et al,
1999). The clinical significance and effects of HGV infection are still poorly understood. Hepatitis B and C viruses
are both transmitted vertically and among the viral hepatitis are of the greatest important to the care of newborns.
HEPATITIS B
The issues that are most important to neonatologists are
the frequency with which hepatitis B virus (HBV) is transmitted to infants at the time of birth, the short-term and
long-term consequences of these infections, the importance of greater surveillance for maternal carriage of HBV,
the role of antiviral therapy to prevent transmission and
disease progression, and the availability of effective HBV
immunoprophylaxis (Arfaoui et al, 2010; Krugman, 1988).
Incidence
In certain parts of the world and among certain ethnic
groups, as many as 7% to 10% of all infants acquire hepatitis B infections at the time of birth, and almost all of
these infections become chronic. In the United States, it
has been estimated that approximately 20,000 infants are
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
499
TABLE 37-3 Viral Hepatitis Types A, B, C, D, E, and G: Comparison of Clinical, Epidemiologic, Immunologic, and Therapeutic Features
Feature
Hepatitis A
Hepatitis B
Hepatitis C
Hepatitis D
(Delta)
Hepatitis E
Virus
Hepatitis A
virus (HAV)
Hepatitis B
virus (HBV)
Hepatitis C
virus (HCV)
Hepatitis D virus
(HDV)
Hepatitis E
virus (HEV)
Hepatitis G virus
(HGC), (GB
virus type C
[GBV-C])
Family
Picornavirus
Hepadnavirus
Flavivirus
Satellite
Calicivirus
Flavivirus
Genome
RNA
DNA
RNA
RNA
RNA
RNA
Incubation period
15-40 days
50-180 days
1-5 months
2-8 weeks
2-9 weeks
Unknown
Usual
Rare
Rare
Food- or
water-borne
No
Usual
Yes
Sexual contact
No
Usual
Yes
Sexual contact
less common
No
Usual
Only with HBV
Sexual contact less
common
Usual
No
Unknown
Water-borne
transmission
in developing
countries
No
Usual
Yes
Sexual contact;
probably less
common
No
No cases
reported
Yes
Yes
Yes
Yes
Yes
Yes
No
No cases
reported
Yes
Yes; controversial
Yes
Yes
No
Yes
Yes
Yes
No
No
Yes
No
No
No
No
No
No
No
No
No
Mode of transmission
Oral (fecal)
Parenteral
Perinatal
Other
Sequelae
Carrier state
Chronic disease
Interventions
Immunoglobulin
Vaccine
Antiviral therapy
Hepatitis
G/GBVC
Modified from Krugman S: Viral hepatitis: A, B, C, D, and E: prevention, Pediatr Rev 13:245-247, 1992.
born annually to mothers who are chronic HBV carriers
(Mast et al, 1998). Since 1990 the incidence of acute hepatitis B in the United States has declined dramatically, with
the largest declines occurring in children younger than 15
years (98%) and in young adults 15 through 24 years of age
(90%). The frequency of transmission depends primarily
on the prevalence of the hepatitis B carrier state among
women of childbearing age. Hepatitis B is a carcinogenic
virus; perinatal acquired infection can lead to chronic liver
failure and hepatic carcinoma in adult life (Balistreri, 1988;
Beasley and Hwang, 1984; Chan and Sung, 2006).
The incidence of neonatal hepatitis B infection depends
on the timing of infection and the overall prevalence of
the disease in the population under study. Women with
acute hepatitis B infection during the first or second trimester rarely transmit the virus to their infants (Krugman,
1988; Stevens, 1994). Transplacental transfer has been
described, but appears to be rare. The carriage rate for
hepatitis B surface antigen (HBsAg) varies from 0.1% in
the United States and Europe to 15% in Taiwan and parts
of Africa, with intermediate rates in Japan, South America,
and Southeast Asia. Transmission rates among immigrant
women in Western countries appear to parallel the rates in
their countries of origin (Krugman, 1988).
The most important route of transmission is transmission that occurs during labor and delivery. The likelihood
of transmission is great if symptomatic acute disease is
present (60% to 70% transmission; Gerety and Schweitzer,
1977). Infants of hepatitis B e antigen (HBeAg)–positive
mothers have an 80% to 90% chance of becoming HBsAg
carriers (Lee et al, 1978; Okada et al, 1976). The risk of
an infant acquiring hepatitis B if born to an HBsAg-positive but HBeAg-negative mother is 5% to 20%. Chronic
neonatal infection occurs in less than 10% of infants of
HBeAg antigen–negative mothers (Krugman, 1988).
Although HBsAg has been found in breast milk, breastfeeding does not appear to have any influence on the rate
of transmission (Beasley et al, 1975). The World Health
Organization currently recommends that all mothers who
are hepatitis B positive breastfeed their infants, and that
their infants be immunized at birth.
Etiology and Pathogenesis
HBV is a DNA virus that localizes primarily in hepatic
parenchymal cells but circulates in the bloodstream, along
with several subviral antigens, for periods ranging from a
few days to many years. Several distinct genotypes have been
identified, and these subtypes show biologic variability in
transmission and disease progression (Magnius and Norder,
1995; Schaefer et al, 2009). Transplacental leakage of HBeAgpositive maternal blood is a potential source of intrauterine
infection (Lin et al, 1987). During either acute or persistent
viremia in the mother, the virus itself or viral antigens may
rarely cross the placenta and cause intrauterine infection, but
more commonly infection occurs perinatally during labor or
delivery (Chisari and Ferrari, 1995; Xu et al, 2001). The finding of hepatitis antigens in the newborn might not indicate
the presence of infection, but rather the passive transfer of
antigen only; therefore antigen tests should be interpreted
cautiously in this setting. Hepatitis B infection of placental
trophoblast has been documented and is compatible with
a transplacental route of infection in some circumstances
(Bai et al, 2007). Most infants born to mothers infected
with HBV have a negative test result for HBsAg at birth,
but in the absence of prophylaxis are at risk for becoming
500
PART IX Immunology and Infections
TABLE 37-4 Licensed Monovalent and Combination Hepatitis B Vaccines
Monovalent Vaccines
Clinical Scenario
Recombivax Hepatitis
B Dose, μg (mL)
Enginerix-B
Dose, μg (mL)
Combination Vaccines
Twinrix*
Comvax†
Pediatrix‡
Newborns born to HBsAg-negative mothers
5 (0.5)
10 (0.5)
NA
NA
NA
Newborns born to HBsAg-positive mothers§
5 (0.5)
10 (0.5)
NA
NA
NA
Infants, children, and adolescents <20 yr old
5 (0.5)
10 (0.5)
NA
5 (0.5)
10 (0.5)
Adults >20 yr old
10 (1.0)
20 (1.0)
20 (1.0)
NA
NA
*Twinrix is a combination of Engerix-B (20 mg) and hepatitis A vaccine, licensed for use in people 18 years of age and older in a three-dose series at a 0-, 1-, and 6-month schedule.
†Comvax is a combination of Recombivax HB (5 μg) and Haemophilus influenzae type b (PRP-OMP) recommended for use at 2, 4, and 12 to 15 months of age. This vaccine should not
be administered at birth.
‡Pediatrix is a combination of diptheria and tetanus toxoids and acellular pertussis (i.e., DTaP), inactivated poliovirus, and hepatitis B (Engerix B, 10 mg). It is recommended for use at 2, 4,
and 6 months of age, but should not be administered at birth, before 6 weeks of age, or after 7 years of age.
§Hepatitis B immunoglobulin (0.5 mL) should be adminstered simultaneously with vaccination.
HBsAg-positive during the first 3 months of life, suggesting
that transmission is primarily peripartum (Krugman, 1988,
1992; Mulligan and Stiehm, 1994; Shapiro, 1993).
Infants with hepatitis B infection do not show clinical or
chemical signs of disease at birth. Without immunoprophylaxis, the usual pattern is the development of chronic
antigenemia with mild and often persistent enzyme elevations, beginning at 2 to 6 months of age (Mulligan and
Stiehm, 1994). Less commonly the infection becomes
clinically manifest, with jaundice, fever, hepatomegaly,
and anorexia, followed by either recovery or chronic active
hepatitis. Rarely, fulminant hepatitis is seen and can be
fatal (Delaplane et al, 1983).
Laboratory tests are essential in the diagnosis of hepatitis
B infection. Evaluations of serum enzymes and of bilirubin
reflect the extent of liver damage. Several helpful serologic
tests identify the virus involved (Krugman, 1988). HBsAg
appears early, usually before liver disease is found; it persists in those who become chronic carriers or disappears in
the 5% of infants who resolve the infection. HBeAg and
anti-HBe testing can be used to assess infectivity; although
they are not as frequently used for serodiagnosis, they are
useful markers of the likelihood of perinatal transmission. PCR and other nucleic acid detection techniques are
important in confirming diagnosis, assessing viral load,
and monitoring the response to therapy.
3. Routine immunization of children and adolescents who
have not been immunized previously
4. Immunization of nonimmunized adults at increased
risk of infection
For infants, the three-dose vaccination schedule should
be initiated in the neonatal period or by 2 months of age
(Table 37-4). Four doses can be given if a birth dose is
given and a combination vaccine is used to complete the
series. Vaccination can be delayed until just before hospital
discharge in preterm (birthweight less than 2000 g) infants
born to HBsAg-negative mothers.
All infants born to HBsAg-positive women should
receive both active and passive immunization within 12
hours of birth. The doses and recommended options for
administration of the hepatitis B vaccines that are currently
licensed in the United States are provided in Table 37-4. If
maternal HBsAg status is unknown, it should be checked
immediately, and the infant should receive the first dose
of HBV vaccine immediately. Infants born to women who
test positive, or to women in whom the HBsAg is unknown,
should be given hepatitis B immunoglobulin as soon as
possible within 1 week after birth. The highest immunization failure rates have been observed in infants of HBeAgpositive women and those infected in utero (Farmer et al,
1987; Tang et al, 1998). It is recommended that infants
born to HbsAg-positive mothers undergo postimmunization testing for anti-HBS and HbsAg 3 to 9 months after
completion of the series, for identification of breakthrough
carriers and of infants who may benefit from revaccination
(AAP Committee on Infectious Diseases, 2010a).
Prevention
Treatment
The primary goal of prevention strategies for hepatitis B
is to prevent chronic infection and chronic liver disease. In
the United States over the past two decades, a comprehensive immunization strategy has been implemented consisting of four components:
1. Universal immunization of all infants beginning at
birth
2. Prevention of perinatal infection through routine
screening of all pregnant women and appropriate
immunoprophylaxis of infants born to HBsAg-positive
women (or women whose HBsAg status is unknown)
There is no therapy for acute hepatitis B infection. Interferon (IFN) α and antiretroviral drugs such as lamivudine, an antiretroviral drug that blocks HBV polymerase,
and adefovir, a reverse transcriptase inhibitor, have been
approved for treatment of chronic hepatitis B in children, but success is widely variable (Giacchino and Cappelli, 2010). Three therapeutic agents for chronic HBV
infection in children have been approved in the United
States, including standard IFN-α, lamivudine, and adefovir (Kurbegov and Sokol, 2009). These antiviral therapies
are summarized in Table 37-2. IFN-α appears to be the
Clinical Spectrum and Laboratory
Evaluation
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
most effective (approximately 30% HBeAg seroconversion; 10% HBsAg seroconversion), although benefits are
primarily observed in children with alanine aminotransferase levels more than twofold the upper limit of normal.
The virologic response rates for lamivudine mirror those
of IFN-α (23% to 31% HBeAg seroconversion) with
easier administration and a more favorable safety profile,
but lower HBsAg seroconversion (2% to 3%) and high
rates of drug resistance. Adefovir demonstrates a favorable safety profile and is less likely to select for resistance
than lamivudine, but virologic response was limited to
adolescent patients and was lower than that of lamivudine
(16% HBeAg seroconversion; <1% HBsAg seroconversion). Entecavir and tenofovir, both approved therapies
for adults with chronic HBV infection, are in trials for
use in children. Some experts recommend “watchful waiting” of children, because current therapies are only 30%
effective at best. Young children are often believed to be
immune tolerant of hepatitis B infection (i.e., they have
viral DNA present in serum, but normal transaminase
levels and no evidence of active hepatitis). These children
should have transaminases and viral load monitored, but
are not typically considered to be candidates for antiviral
therapy.
Prognosis
Most long-term follow-up studies have shown that children vaccinated at birth have high levels of protection
until at least 5 years of age. Approximately 5% to 10% of
infants born to HbeAg-positive mothers become chronic
HBV carriers despite combined active and passive immunoprophylaxis with hepatitis B immunoglobulin and HBV
vaccine (Kato et al, 1999). Failure of immunoprophylaxis
may be associated with the level of maternal viremia and
specific HBV genetic variants (Ngui et al, 1998). Infants
who become infected with HBV perinatally have a 90%
risk of chronic infection, and 15% to 25% of those with
chronic infection die of HBV-related liver disease (primarily hepatocellular carcinoma) as adults. There is some
evidence that the risk of carcinoma correlates with specific
hepatitis B genotypes (Sherman, 2010).
HEPATITIS C
In 1989, hepatitis C virus (HCV) was found to be the main
cause of non-A, non-B, parenterally transmitted hepatitis;
subsequently, HCV has been found to account for a significant portion of the cases of sporadic acute and chronic
hepatitis (Choo et al, 1989; Reyes et al, 1990; Weiss and
Persing, 1995). Hepatitis C virus is a small, single-stranded
RNA virus that is a member of the family Flavivirus; this
family consists primarily of vector-borne infections, such as
St. Louis encephalitis virus and West Nile virus. Hepatitis
C virus is an exception in this family because it is not transmitted by insect vectors. Seven genotypes are described,
with significant biologic differences in regard to disease
progression and responsiveness to therapy (Klenerman et
al, 2009). Vertically transmitted hepatitis C infection in
infants is associated with a higher rate of chronic hepatitis,
but less liver injury compared with adult HCV infections
(Tovo et al, 2000).
501
Incidence
The seroprevalence for anti-HCV antibody in pregnant
women ranges from 0.7% to 4.4% worldwide (Conte et al,
2000). In the United States, seroprevalence of hepatitis C
decreased during the 1990s and has remained low and stable since then. The overall seroprevalence is estimated to
be 1.3%, with a seroprevalence in pregnant women ranging from 1% to 2% (AAP Committee on Infectious Diseases, 2010a). Forty percent to 50% of women with HCV
have no identified known risk factors for infection (Bortolotti et al, 1998). Estimates from the 1990s suggested that
the vertical transmission rate of HCV is approximately 5%
to 11% in HIV-negative mothers, and ranges from 10%
to 20% from mothers coinfected with HIV (Hillemanns
et al, 2000; Palomba et al, 1996; Polywka et al, 1997; Tajiri
et al, 2001). More recent studies, however, suggest a lower
rate of vertical transmission, ranging from 2.7% to 3.6%
(Ferrero et al, 2003; Syriopoulou et al, 2005). One study
has shown, in a multivariate analysis, that higher risk of
vertical transmission is related more to maternal use of
injection drugs than to HIV infection itself, although
the mechanism for this finding is still unclear (Resti et al,
2002). The risk of transmission correlates with maternal
viremia, and transmission appears to be rare in the absence
of viremia. The risk of HCV infection from transfused
blood after the advent of HCV screening is estimated to
be less than 1 in 1 million units transfused.
Etiology and Pathogenesis
HCV is transmitted less efficiently than HBV by sexual
contact. Risk factors for HCV infection include transfusion, intravenous drug use, frequent occupational exposure
to blood products, and household or sexual contact with
an infected person (Weiss and Persing, 1995). In children,
perinatal transmission is the most common route of infection (Mohan et al, 2010). Preparations of IVIG contaminated with HCV were reported between April 1993 and
February 1994, but since that time, routine screening for
HCV with PCR and application of a viral inactivation
process during manufacturing have been implemented to
reduce the risk of transmission (Schiff, 1994).
Perinatal transmission is the leading cause of childhood HCV infection (Mohan et al, 2010). Transmission
of HCV from mother to child is thought to occur either
in utero or at the time of delivery. Viral genotypes and
infection and replication in maternal peripheral blood
monocytes can also affect the ability of the virus to infect
the fetus or newborn (Azzari et al, 2000; Zuccotti et al,
1995). Perinatal transmission is confined almost always
to women with detectable HCV ribonucleic acid (RNA)
in the peripheral blood by the PCR, but all children born
to women with anti-HCV antibodies should be tested for
HCV. Data regarding the correlation of HCV RNA titer
in the mother with the risk of vertical transmission are
conflicting, although most studies have reported an association (Conte et al, 2000; Lynch-Salamon and Combs,
1992; Ohto et al, 1994; Resti et al, 1998; Tajiri et al, 2001).
Maternal peripheral blood mononuclear cell infection by
HCV, membrane rupture of longer than 6 hours before
delivery, and procedures exposing the infant to maternal
502
PART IX Immunology and Infections
blood infected with HCV during vaginal delivery are
associated with an increased risk of transmission. Internal
fetal monitoring is also a risk factor for transmission (Mast
et al, 2005). Maternal coinfection with HCV and human
immunodeficiency virus, maternal history of intravenous
drug use and of HCV infection of the sexual partner of
the mother are also risk factors for transmission (Fiore
and Savasi, 2009; Indolfi and Resti, 2009). Transmission
rates as high as 25% have been reported in HCV-infected,
HIV-positive women. The effect of vaginal delivery versus cesarean section on transmission rates is unclear (Gibb
et al, 2000; Paccagnini et al, 1995); current recommendations do not support the practice of cesarean delivery in
women with HCV infection.
Transmission of HCV via breast milk has not been demonstrated conclusively, although HCV can be found in
breast milk and colostrum (Gurakan et al, 1994; Zimmermann et al, 1995). Studies comparing breastfed and bottlefed infants born to HCV-infected mothers have not shown
a statistically significant difference in vertical transmission
(Lin et al, 1995; Resti et al, 1998). Accordingly, maternal
HCV infection is not a contraindication to breastfeeding (Mast, 2004); however, it may be prudent for mothers
who are HCV-infected and who choose to breastfeed to
consider abstaining from breastfeeding if their nipples are
cracked and bleeding.
Clinical Spectrum and Laboratory
Evaluation
Acquired HCV infection typically causes jaundice in one
third of cases and significant increases in alanine aminotransferase (ALT) in almost all persons infected. Most
neonates perinatally infected with HCV demonstrate little in the way of clinical symptomatology; they may have
elevated liver ALT either transiently or intermittently.
The increases in ALT values, when tested, are most commonly noted between 3 and 6 months of age. HCV-RNA
PCR results may be negative initially at birth or within the
first few days of life, but typically become positive by 1 to
2 weeks of age and remain so until at least 5 years of age.
The highest sensitivity of PCR is reported after 1 month
of age (Polywka et al, 2006; Thomas et al, 1997). Confirmatory anti-HCV IgG serology should be delayed until
exposed infants are at least 15 to 18 months old, when at
least 99% will have cleared maternal antibody (Dunn et
al, 2001). Before 18 months of age, only a positive HCV
PCR result can confirm diagnosis of neonatal infection;
positive anti-HCV IgG results simply reflect passively
acquired maternal antibody. IgM assays are not available
or reliable for perinatal diagnosis of HCV. Liver ultrasonographic findings are usually normal or may consist
of a mild diffuse increase in echogenicity. Liver biopsies,
when performed, typically demonstrate mild to moderate
chronic persistent hepatitis (Palomba et al, 1996; Tovo
et al, 2000).
Treatment
Interferon and ribavirin are both approved by the FDA to
treat adults and children with chronic hepatitis C. Antiviral agents are summarized in Table 37-2. Only 10% to
25% of adults treated with interferon have a sustained
remission of disease (Bonkovsky and Woolley, 1999).
Treatment with a combination of interferon and ribavirin achieves remission in closer to half of treated adults
(Cornberg et al, 2002). Randomized controlled trials
indicate that patients treated with pegylated interferons
(so called because they are formulated and stabilized with
polyethylene glycol), both as dual therapy with ribavirin
and as monotherapy, experience higher sustained viral
response rates than do those treated with nonpegylated
interferon (Shepherd et al, 2004). Data on the use of these
agents in infants and children are limited, although the use
of IFN-α2b in combination with ribavirin was recently
approved by the FDA (Palumbo, 2009). In a case series of
four pediatric patients treated with interferon for 1 year,
viremia decreased to undetectable levels during treatment in all four patients, but two patients became viremic
again once the treatment was stopped (Tovo et al, 2000).
There are significant genotype-dependent differences in
responsiveness to antiviral therapy; patients with genotype
1 had the lowest levels of sustained virologic response, and
patients with genotype 2 or 3 had the highest (Palumbo,
2009; Shepherd et al, 2004). The use of IFN-α2b in combination with ribavirin provides a much more favorable
sustained virologic response in children with HCV genotype 2/3 (84%) than in those with HCV genotype 1 (36%;
González-Peralta et al, 2005). For genotype 1 hepatitis C
treated with pegylated interferons combined with ribavirin, it has been shown that genetic polymorphisms near
the human IL28B gene, encoding interferon lambda 3, are
associated with significant differences in response to the
treatment (Ge et al, 2009; Thomas et al, 2009). Interferon
therapies are available only in parenteral formulations and
are associated with significant side effects; therefore further research into its utility in pediatric HCV infection is
needed.
Infants and children with persistent elevations in liver
transaminases should be referred to a pediatric gastroenterologist for evaluation and management. The necessity
for, as well as the frequency of, screening tests of liver
function has not been established.
Prognosis
In 60% to 80% of HCV-infected adults, chronic hepatitis
occurs, and in one third of HCV-infected adults, chronic
HCV infection leads to cirrhosis or liver failure within 20
to 30 years after infection (Iwarson et al, 1995; Leone and
Rizzetto, 2010). In patients with cirrhosis, the incidence
of hepatocellular carcinoma is 2% to 5% per year. There
are limited data regarding long-term follow-up in HCVinfected infants, but existing knowledge indicates that
most children remain viremic until at least 5 to 6 years of
age, and most develop chronic, persistent hepatitis. Studies monitoring infected children beyond 6 years of age
have not been completed, but they are at risk for cirrhosis
and hepatocellular carcinoma. Transient hepatitis C viremia with subsequent resolution has been reported (Padula
et al, 1999; Ruiz-Extremera et al, 2000; Zanetti et al, 1995).
All the infants who have been followed for an average of 3
to 4 years have been reported to have normal growth and
development (Tovo et al, 2000).
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
503
Prevention
Although development of an HCV vaccine is a major public health priority (Houghton and Abrignani, 2005), there
is currently no vaccine available. Like all other infants,
infants with HCV should receive routine hepatitis B
immunization. In addition, they should receive hepatitis
A vaccination at 2 years of age. Parents should be advised
to avoid unnecessary administration of medicines known
to be hepatotoxic. Standard precautions are recommended
for the hospitalized infant.
ADENOVIRUS
Adenoviruses are so named because they were originally
recovered from human adenoidal tissue; they are mediumsized (90 to 100 nm), nonenveloped icosahedral viruses
composed of a nucleocapsid and a double-stranded linear
DNA genome. They are the largest of the nonenveloped
viruses. There are 53 described adenovirus serotypes in
humans.
EPIDEMIOLOGY AND CLINICAL
MANIFESTATIONS
Adenoviruses are responsible for a wide variety of clinical syndromes, including conjunctivitis, respiratory track
disease, and gastroenteritis. Adenovirus causes 5% to
10% of upper respiratory infections in children, and many
infections in adults as well. No clear seasonality has been
described for adenovirus infections.
ADENOVIRUS INFECTIONS IN NEWBORNS
There are a limited number of reports of adenovirus infection in young infants, but published case series indicate
that adenovirus can cause serious, life-threatening disease
in the neonate. A review of neonatal adenovirus infection (Abzug and Levin, 1991) identified several characteristic historical features, including prolonged rupture
of membranes, history of maternal illness, vaginal mode
of delivery, and onset of illness within the first 10 days of
life. Serotypes 2, 3, 7, 11, 13, 19, 21, 30, and 35 have been
implicated (Abzug and Levin, 1991; Andiman et al, 1977;
Matsuoka et al, 1990; Osamura et al, 1993; Pinto et al,
1992; Sun and Duara, 1985). Clinical findings in various
case reports and case series have included lethargy, fever
or hypothermia, anorexia, apnea, hepatomegaly, bleeding, and progressive pneumonia (Figure 37-5). Laboratory
abnormalities include thrombocytopenia, coagulopathy,
and hepatitis. Acquisition of infection from the mother via
vaginal delivery is the presumed mode of transmission in
most cases, although transplacental spread has also been
implicated.
Although most neonatal adenovirus infection is believe
to be acquired in the birth canal, congenital infections
have been described, presumably because of transplacental transmission. A wide range of fetal pathologies including pleural effusion, hepatitis, myocarditis, meningitis,
and CNS abnormalities have been described (Baschat
et al, 2003; Meyer et al, 1985; Rieger-Fackeldey et al,
2000). Adenovirus has been implicated as a potential cause
FIGURE 37-5 (See also Color Plate 19.) Postmortem histologic analysis
from an infant who died from disseminated adenovirus infection at
2 weeks of age. Hematoxylin and eosin stain of lung demonstrating
inflammatory infiltrates (arrow) and intranuclear inclusions (arrowhead).
This infant had a viral sepsis syndrome characterized by hepatic failure,
disseminated intravascular coagulation, and pneumonitis from adenovirus infection presumed to have been acquired intrapartum.
of chorioamnionitis and premature birth (Tsekoura et al,
2010; Van den Veyver et al, 1998).
PREVENTION AND INTERVENTION
Intravenous ribavirin has been administered to a neonate with disseminated adenovirus infection undergoing
extracorporeal membrane oxygenation, with evidence of
viral clearance within 48 hours of initiating therapy (Aebi
et al, 1997). Adenovirus vaccines were at one time available
for use in military personnel (Gaydos and Gaydos, 1995;
Tucker et al, 2008), but are not currently in production.
Adenoviruses are hardy and resistant to inactivation by
physical and chemical methods that kill most viruses, adding to the challenge of hospital infection control during
outbreaks of infection.
RESPIRATORY VIRUSES
Although relatively uncommon, any one of the respiratory viruses can cause symptomatic respiratory disease in
newborn infants. The association has been described for
rhinoviruses, adenoviruses, parainfluenza viruses, influenza virus, and respiratory syncytial virus. Adenovirus,
rhinovirus, and parainfluenza virus infections are generally characterized by mild rhinorrhea in neonates. All of
these viruses, however, can cause clinical symptoms indistinguishable from those of bacterial infection, leading to
increased diagnostic testing and empiric antibiotic treatment. Influenza virus infections are usually mild, but in
the absence of maternally transmitted antibody, they can
be life threatening, with severe pneumonia, hypoxia, and a
504
PART IX Immunology and Infections
prolonged course. During the H1N1 influenza pandemic
of 2009 to 2010, pregnant women were at uniquely high
risk for severe influenza (Jamieson et al, 2009), and infections were described in neonates (Sert et al, 2010).
The most extensive nursery outbreaks, however, have
been caused by respiratory syncytial virus (RSV; Hall et al,
1979; Wilson et al, 1989). Because of the importance that
RSV plays in the newborn nursery, this virus is considered
in greater detail in the following section.
RESPIRATORY SYNCYTIAL VIRUS
RSV is the major cause of viral pneumonia and bronchiolitis in infants and children. In temperate climates, it causes
large annual epidemics during the winter and early spring
months (typically ranging from November through April).
During these months, RSV appears to be responsible for
up to 20% of all pediatric hospital admissions (Hall et al,
2009). Nosocomial infections are frequent during these
times, and illness among hospital staff members is a major
factor in its spread from infant to infant. Several nursery
outbreaks have been described. In one of these outbreaks,
cultures were obtained prospectively so that a full picture of
the virus’s pathogenicity and epidemiology could be drawn
(Hall et al, 1979). Twenty-three of 66 infants hospitalized
for 6 days or more were infected. Virtually all infants were
symptomatic. Clinical manifestations included pneumonia,
upper respiratory infection, apneic spells, and nonspecific
signs. Pneumonia and apnea were seen almost exclusively
in infants older than 3 weeks, and nonspecific signs were
most commonly observed in younger infants. Four (17%)
infants died, two unexpectedly, during the course of infection. The spread of infection in the unit was difficult to
interrupt; infants in isolettes did not seem to be protected
against acquisition of the infection. Eighteen of the 53
nursery personnel were infected during the outbreak; 83%
of the infected nursery providers were symptomatic. Of
particular importance is the observation that RSV infection, both in term and in preterm infants, is commonly
associated with a new onset of apnea (Bruhn et al, 1977).
Boys are at greater risk than girls for serious RSV disease.
RSV-associated apnea has been the probable explanation for some case reports of deaths attributed to SIDS
(Eisenhut, 2006). RSV lower respiratory track disease can
be slow to resolve. After discharge from initial hospitalization, risk factors for infant rehospitalization secondary to
RSV infection include premature gestational age, chronic
lung disease, siblings in daycare or school, chronologic
age of less than 3 months, and exposure to tobacco smoke
(Carbonell-Estrany and Quero, 2001).
Diagnosis can be confirmed by DFA staining of nasopharyngeal or tracheal aspirate, nasopharyngeal swab, or
other respiratory secretions. Culture of RSV can require 3
to 5 days and can be used if DFA staining is not available.
PCR is also available for the rapid diagnosis of infection.
Treatment and prevention of RSV infection in infants
attracted considerable attention during the 1990s because
of the clinical and economic impacts of these infections
(Groothuis, 1994; Kinney et al, 1995; Levin, 1994; Meissner, 1994). Considerable debate has ensued concerning the
efficacy, safety, and potential effect on health care workers of ribavirin therapy, so its use remains controversial
BOX 37-4 G
uidelines for Prophylaxis in
Preterm Infants at Start of RSV
Season
PROPHYLAXIS RECOMMENDED
Infants born at ≤28 weeks, 6 days of gestation during first RSV season*
ll Infants born at 29 weeks, 0 days through 31 weeks, 6 days of gestation if
less than 6 months of age at beginning of RSV season
ll Infants and children less that 24 months of age with chronic lung disease
requiring medical therapy (e.g., supplmental oxygen, bronchodilators,
diuretics, corticosteroids) within the 6 months before RSV season
ll Infants born at 32 weeks, 0 days through 34 weeks, 6 days of gestation if
one of the following two risk factors are present:
ll Attending child care
ll Sibling younger than 5 years of age
ll
PROPHYLAXIS CONSIDERED
Infants with congenital anomalies of the airways
ll Infants younger than 24 months of age with hemodynamically significant
congenital heart disease, as manifest by congestive heart failure, pulmonary hypertension, and cyanosis
ll Infants with severe neuromuscular disease
ll Infants with immune deficiencies
ll Infants with cystic fibrosis
ll
RSV, Respiratory syncytial virus.
*Earliest date to consider initiation of prophylaxis depends upon geographic location: July 1
for southeast Florida, September 15 for north central and southwest Florida, November 1 for
most other areas of the United States.
(Meissner, 2001; Ventre and Randolph, 2004; Wald and
Dashefsky, 1994). Treatment consists of nebulization of
ribavirin by a small-particle aerosol generator supplied by
the manufacturer into an oxygen hood, tent, or mask from
a solution containing 20 mg of ribavirin per milliliter of
water (see Table 37-2). The aerosol has been administered
on various schedules for 3 to 5 days (e.g., 12 to 20 h/day).
The efficacy of ribavirin in this setting remains unclear.
Some long-term benefit on recurrent wheezing may be
realized (Ventre and Randolph, 2004). Corticosteroids are
not effective in the treatment of RSV.
Prevention efforts have focused on passive and active
immunization. Standard immunoglobulin has not been
shown to be efficacious for prevention of RSV infection
in high-risk infants (Meissner et al, 1993). The efficacy
of monthly prophylactic administration of RSV-specific
immunoglobulin (750 mg/kg or 150 mg/kg) in 249 infants
with cardiac disease or bronchopulmonary dysplasia was
examined in a multicenter trial (Groothuis, 1994). In the
high-dose (750 mg/kg) group, there were fewer lower respiratory tract infections, hospitalizations, days in hospital, and
days in the intensive care unit as well as less use of ribavirin. Subsequently a mouse monoclonal antibody was developed, which provides similar protection against RSV (i.e.,
palivizumab). Unlike anti-RSV immunoglobulin, which is
a pooled human blood product, the monoclonal antibody
does not confer the theoretical risk of acquiring bloodborne pathogens. Palivizumab is also substantially easier
to administer because it is an intramuscular injection. Recommendations by the AAP for the use of palivizumab are
summarized in Box 37-4. Additional preventive measures
for high-risk infants include eliminating or minimizing
exposure to tobacco smoke, avoiding crowds and situations
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
GASTROINTESTINAL VIRUSES
The most important of the viruses that cause diarrhea from
the perspective of the neonatal nursery are the rotaviruses.
This important group of viruses, with at least four serotypes, is responsible for a large proportion of significant
and sometimes severe diarrhea in infants 6 to 24 months of
age (Cohen, 1991; Greenberg et al, 1994; Haffejee, 1991;
Taylor and Echeverria, 1993). Recent evidence suggests
that rotavirus extracts an unexpectedly significant burden of disease on infants as young as 2 to 3 months of age
(Clark et al, 2010). Nursery-acquired infections are common; surprisingly, such infections appear to be benign in
most infants. Two studies performed in nurseries in Sydney, Australia, and in London found that 30% to 50% of
5-day-old babies shed the virus (Chrystie et al, 1978; Murphy et al, 1977). However, more than 90% of the infected
infants were asymptomatic. The remaining symptomatic
infants had loose stools and vomiting, but this figure was
only slightly higher than the proportion of uninfected
infants with the same symptoms. The full scope of rotavirus disease in the newborn and in the premature infant
remains to be fully defined. The recent advent and licensure of several rotavirus vaccines should have a significant
effect on both nosocomial and community-acquired rotavirus disease (Bernstein, 2009).
Human Immunodeficiency
Virus Infection During
Pregnancy
CHANGING EPIDEMIOLOGY OF THE HIV/AIDS
PANDEMIC
From 1980, when previously healthy, young, gay men
first began to have complaints of fever, malaise, weight
loss, lymphadenopathy, and malaise, to the identification in 1983 of 36 cases in Kinshasa, Zaire (Congo) of
acquired immunodeficiency syndrome (AIDS) by a team
of representatives from the CDC and National Institutes
of Health, the scope of HIV infection has evolved into a
global pandemic. The Joint United Nations Program on
140
Rate (per 100,000 population)
in which exposure to infected individuals cannot be controlled, careful hand hygiene education of parents, vaccinating against influenza beginning at 6 months of age, and
restricting participation in child care during the RSV season whenever feasible. The prospects for infant or maternal immunization with live attenuated and protein subunit
vaccines are under active investigation and development
(Crowe, 2001; Fretzayas and Moustaki, 2010; Piedra, 2000).
Nosocomial spread of RSV and other respiratory viruses
can be minimized by emphasis on hand washing by care
providers between contacts with patients. Without additional special precautions, an attack rate of approximately
26% has been observed (Madge et al, 1992). Use of standard contact precautions, such as cohort nursing and the
use of gowns and gloves for all contacts with RSV-infected
children, can reduce the risk of nosocomial RSV infection
to 9.5% (Madge et al, 1992).
120
100
80
505
American Indian/Alaska Native
Asian
Black/African American
Hispanic/Latino*
Native Hawaiian/Other Pacific Islander
White
Mutiple races
60
40
20
0
Males
Females
FIGURE 37-6 Estimated rates of diagnoses of HIV infection among
adults and adolescents, by sex and race/ethnicity, in 2008 for 37 states
with confidential name-based HIV infection reporting. Estimated rates
resulted from statistical adjustment that accounted for reporting delays,
but not for incomplete reporting. (From Centers for Disease Control and
Prevention: HIV Surveillance Report 20, 200, June 2010. Available at
www.cdc.gov/hiv/topics/surveillance/resources/reports. Accessed November 24,
2010.)
HIV/AIDS estimated that, as of the end of 2008, up to 35
million individuals were living with HIV/AIDS globally.
Relevant to this chapter, women accounted for 60% of all
adults living with HIV (Christie et al, 2008).
Currently, national and international surveillance for
HIV infection and the resulting illness of AIDS is routine.
In the United States, CDC surveillance data for 2005 to
2008 revealed an 8% increase in the absolute number of
newly diagnosed HIV infections, whereas the overall rate
of new infections was stable. Women currently account
for 25% of the HIV-infected U.S. population, with African American women having the highest rate of diagnosed
infection among women (Figure 37-6). In Florida, an HIV
prevalence study among pregnant women from 1998 to
2007 found that black women were 11-fold more likely than
white women to have HIV infection (Salihu et al, 2010).
The incidence of HIV infection is challenging to measure
and has not been measured directly. Based on a relatively
new antibody test that can distinguish recent (median, 156
days) seroconversion from longstanding infection, it is
estimated that there were 13,000 to 15,000 HIV infections
among U.S. women in 2006 (Hall et al, 2008).
Estimates of HIV prevalence in pregnant women worldwide are challenging to obtain and may be underestimated,
because those who consent to testing and counseling
are more likely to be HIV negative. For example, in an
urban antenatal clinic in Zambia, 25% of women accepting HIV testing were found to be HIV positive, whereas
anonymous sampling of infants’ cord blood demonstrated
a prevalence of 29% among women who refused testing
(p <0.0001 for a difference in HIV prevalence between the
two groups; Stringer et al, 2005). In Zimbabwe, HIV prevalence among antenatal clinic attendees has been declining, although rates remain high: 32.1% in 2000 and 23.9%
in 2004 (Mahomva et al, 2006). Infection rates in antenatal
clinics in KwaZulu-Natal, South Africa, reached 37.5%
(Ramogale et al, 2007). In the country of South Africa
alone, there are an estimated 220,000 pregnant women
living with HIV infection. Most African cities exhibit an
HIV prevalence rate in the 20% to 40% range, whereas
506
PART IX Immunology and Infections
estimates in India are markedly lower—closer to 1% to
2% (Dandona et al, 2008; Lionel et al, 2008).
A markedly disproportionate burden of disease is spread
among a few countries as evidenced by this summary statement in a 2008 World Health Organization report: “Close
to 90% of all pregnant women living with HIV in low and
middle-income countries live in 20 countries and 75% are
concentrated in 12 countries” (World Health Organization, 2008). The high prevalence of HIV infection among
women living in settings where antiretroviral medications
are not universally available results in many cases of pediatric HIV infection. Mother-to-child transmission accounts
for 90% of HIV infections in children. A recent estimate
is that a staggering 2.1 million children younger than 15
years are infected with HIV (World Health Organization,
2008).
In addition to the expansive societal and economic cost
directly resulting from maternal HIV infection, there is a
ripple effect of HIV infection on prevalence and morbidity of other infections. Tuberculosis and malaria are now
common opportunistic infections of HIV in low-income
countries. Herpes and HIV are considered “overlapping epidemics” (Corey et al, 2004), and the interplay is
complex. Reactivation of HSV increases HIV RNA levels in plasma, and mucocutaneous shedding of HIV-1 is
greater during mucocutaneous replication of HSV-2. Less
publicized but more morbid effects are seen on the risk
for puerperal sepsis, which is increased in HIV-infected
women. A study in Zimbabwe demonstrated an 11-fold
increase in risk for puerperal sepsis among HIV-infected
women compared with HIV-negative women (Zvandasara
et al, 2006). The same trend is exhibited in European settings; the European Collaborative Study found a fourfold
increased risk for puerperal sepsis among HIV-infected
women in 14 HIV reference centers across five countries
(Fiore et al, 2004). Clearly the effect of HIV is multidimensional, and interdisciplinary cooperation is essential to
effect change.
PATHOGENESIS OF HIV DISEASE
Transmission of the Virus
Transmission of HIV infection requires the exchange of
bodily fluids. Numerous studies have demonstrated that
casual household or community contacts are not associated with transmission or acquisition of HIV/AIDS. The
three general mechanisms by which the virus can be passed
from one individual to another are sexual, parenteral, and
perinatal.
Different types of exposure to infection are associated
with different risks of infection (Royce et al, 1997). One
sexual transmission occurs for every 100 to 1000 exposures. Male-to-female transmission risk is about 10-times
greater than female-to-male transmission risk. Needlestick injury from an infected source carries a transmission
risk of approximately 0.3% (Panlilio et al, 2005), with the
risk from needle sharing being significantly greater. The
risk of transmission through blood transfusion is greater
than 90%, although this rarely occurs in the United States
because of current screening methods used by blood
banks.
Mother-to-Child Transmission
Transfer of virus from maternal to fetal or neonatal tissues
is thought to occur during one of three discrete periods—
antepartum (before the onset of labor), peripartum (during labor and delivery), and during breastfeeding. Studies
of early viral dynamics in infected neonates suggest that
the peripartum period is the time of highest risk (Mofenson, 1997). The risk of vertical transmission is heightened
by premature rupture of membranes, high maternal viral
load, and low maternal CD4 counts (Ioannidis and Contopoulos-Ioannidis, 1999; Mofenson et al, 1999). Without
antiretroviral treatment, the risk of vertical transmission
is estimated to be 15% to 20% in Europe, 15% to 30%
in the United States, and 25% to 35% in Africa (Volmink
et al, 2007). With highly active antiretroviral therapy, the
risk of vertical transmission in the United States has been
reduced to 1% to 5% (Coovadia, 2009).
Transmission through breast milk carries a 5% to 20%
risk of transmission, largely dependent on the viral load
and CD4 count of the mother (World Health Organization, 2008). A Kenyan study that randomized infants of
HIV-infected mothers to breast versus formula feeding
found that breastfeeding increased the risk of HIV transmission by 16% (Nduati et al, 2000b).
Prevention of Mother-to-Child Transmission
The cornerstone of preventing mother-to-child transmission of HIV is knowledge of the mother’s HIV status.
Updated recommendations from the CDC in 2006 (Branson et al, 2006) reinforced the importance of antenatal
testing by stating that:
1. HIV screening should be included in the routine panel
of prenatal screening tests for all pregnant women.
2. HIV screening is recommended after the patient is
notified that testing will be performed, unless the
patient declines (i.e., opt-out screening).
3. Separate written consent for HIV testing should not
be required; general consent for medical care should
be considered sufficient to encompass consent for HIV
testing.
4. Repeated screening in the third trimester is recommended in certain jurisdictions with elevated rates of
HIV infection among pregnant women (described as
one diagnosed HIV case per 1000 pregnant women per
year).
Research has shown that HIV testing rates are highest
when included in standard testing for pregnant women
both in high-income and low-income countries. A study
done in Birmingham, Alabama, found that HIV testing
rates increased from 75% to 88% (p <0.001) subsequent to
the 1999 Institute of Medicine recommendation to make
HIV testing a routine part of antenatal care (Stringer
et al, 2001). Similarly, in Zimbabwe, testing rates increased
from 65% to 99.9% (p <0.001) after implementation of an
opt-out HIV testing approach in antenatal clinics (Chandisarewa et al, 2007).
HIV testing rates in most low-income countries are
lower than those seen in Zimbabwe, but modest gains are
being made. Globally, HIV testing in low- and middleincome countries rose from 10% of pregnant women
attending antenatal clinics in 2004 to 18% in 2007 (World
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
Health Organization, 2008). As antiretroviral therapy
becomes more readily available and the effects of this therapy on vertical transmission become more widely known,
the incentive to perform HIV testing among pregnant
women should increase.
ll
Maternal Therapy
ll
The introduction of potent antiretroviral therapies in the
middle 1990s markedly reduced deaths caused by AIDS in
most regions of the developed world. Antiretroviral therapies have not only reversed AIDS mortality trends in the
developed world; they have also dramatically altered patterns of HIV transmission from mothers to children. The
results of the Pediatric AIDS Clinical Trial Group Study
076 lead to a profound change in the approach to medical care for HIV-positive pregnant women (Connor et al,
1994). This prospective, placebo-controlled, randomized
trial demonstrated that zidovudine prophylaxis during
pregnancy, delivery, and early infancy reduced the rate of
vertical HIV transmission from 25% to 8%. Less than 2
years later, additional antiretroviral agents became available
for treatment of HIV infection. It also became possible to
monitor viral load via plasma HIV-1 RNA assays. These
monitoring and treatment tools created the potential for the
well-controlled management of HIV infection in pregnant
women seen today, where it is common to see viral loads of
less than 1000 copies per milliliter and vertical transmission
risk as low as 1%. Numerous studies have been conducted
in low- and middle-income countries as well, demonstrating the benefit of antiretroviral therapy during pregnancy
for prevention of vertical transmission of HIV (Dorenbaum
et al, 2002; Ioannidis et al, 2001; Namukwaya et al, 2010).
Current guidelines exist for antiretroviral therapy in
pregnant women (Panel on Treatment of HIV-Infected
Pregnant Women and Prevention of Perinatal Transmission, 2010; available at http://aidsinfo.nih.gov/Content
Files/PerinatalGL.pdf). Antiretroviral therapy is recommended for all HIV positive pregnant women during the
latter trimesters of their pregnancy, with reassessment of
the need to continue antiretroviral therapy after birth of
the infant. If a woman is already receiving antiretroviral
therapy that successfully suppresses viremia, it is recommended to continue the same regimen but find substitutes
for any teratogenic drug (e.g., efavirenz). If the woman has
a new HIV diagnosis, is antiretroviral therapy–naïve, and
does not require antiretroviral therapy for her own health,
therapy should be initiated for prophylaxis against vertical transmission based on the parameters in Box 37-5. If
the mother is symptomatic from her infection or has a history of antiretroviral therapy use, the Perinatal Guidelines
should be referenced for additional recommendations
(Panel on Treatment of HIV-Infected Pregnant Women
and Prevention of Perinatal Transmission, 2010).
Internationally, women in countries with the highest
HIV prevalence often have less 25% intrapartum antiretroviral therapy coverage (Figure 37-7). United Nations
goals target an 80% coverage rate of antiretroviral therapy in pregnant women, but drug availability still varies
widely between countries. Rapid increases in antiretroviral availability in some African countries during 2004 to
2007 prove that such services can be provided quickly and
507
BOX 37-5 T
reatment Recommendations for
HIV-Infected, Antiretroviral-Naive
Asymptomatic Pregnant Women
ll
ll
ll
Perform HIV antiretroviral drug resistance testing before initiating combination antiretroviral drug therapy and repeat after initiation of therapy if viral
suppression is suboptimal.
Prescribe a combination antiretroviral drug prophylaxis regimen (i.e., at
least three drugs) for prophylaxis of perinatal transmission.
ll Consider delaying initiation of antiretroviral prophylaxis until after first
trimester is completed.
ll Avoid use of efavirenz or other potentially teratogenic drugs in the first
trimester and drugs with known adverse potential for the mother (e.g.,
combination stavudine/didanosine).
ll Use zidovudine as a component of the antiretroviral regimen when feasible.
ll If the woman has CD4 count >250 cells/mm3, use nevirapine as a component of therapy only if the benefit clearly outweighs the risk, because
of an increased risk of severe hepatic toxicity.
Although the use of zidovudine prophylaxis alone is controversial, consider
it if the woman has plasma HIV RNA level <1000 copies/mL on no therapy.
Continue antiretroviral prophylaxis regimen during intrapartum period (zidovudine given as continuous infusion during labor while other antiretroviral
agents are continued orally).
Evaluate the need for continuing the combination regimen postpartum;
discontinue the combination regimen unless the woman has indications for
continued therapy. If regimen includes a drug with a long half-life, such as
NNRTI, consider stopping NRTIs at least 7 days after stopping NNRTI.
Adapted from Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission: [Perinatal Guidelines] Recommendations for use of antiretroviral drugs in
pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV
transmission in the United States, May 24, 2010, pp 1-117. http://aidsinfo.nih.gov/ContentFiles/
PerinatalGL.pdf. Accessed November 22, 2010.
effectively in low-income countries given adequate political and financial support (Figure 37-8) (World Health
Organization, 2008).
Antiretroviral Drug Resistance
Resistance to available antiretroviral drug therapy has
become an increasingly important problem requiring continual reevaluation of antiretroviral therapy regimens for
women and prophylactic regimens against perinatal transmission in both mother and infant. Drug resistance in the
context of longitudinal maternal antiretroviral therapy is
beyond the scope of this chapter, but current recommendations for appropriate drug regimens can be accessed in the
Perinatal Guidelines (Panel on Treatment of HIV-Infected
Pregnant Women and Prevention of Perinatal Transmission, 2010). It is important to conduct resistance testing
before antiretroviral therapy initiation in women or infants,
with the exception of the recommended 6 weeks of postnatal
zidovudine for infants described in the following section).
Selection of an appropriate combination drug regimen is
best undertaken with the advice of a clinician specializing in
the care of HIV-infected pediatric or adult patients.
In low- and middle-income countries, a single dose of
nevirapine is often used to prevent vertical transmission of
HIV (Guay et al, 1999). Whereas single-dose nevirapine is
the most widespread approach to prophylaxis against perinatal transmission of HIV because of cost, limited availability of other drugs, and ease of administration, issues of
508
PART IX Immunology and Infections
80% to 100%
50% to 80%
25% to 50%
10% to 25%
Less than 10%
Data not available
High income areas
FIGURE 37-7 Coverage of antiretroviral agents to prevent mother-to-child transmission of HIV in low- and middle-income countries: 2007.
(From World Health Organization: Towards universal access: scaling up priority HIV/AIDS interventions in the health sector. Progress Report 2008.
Available at www.who.int/hiv/pub/2008progressreport/en. Accessed November 22, 2010.)
Estimated number of pregnant women
living with HIV
% of pregnant women living with HIV
receiving antiretrovirals to reduce the risk of
mother-to-child transmissjion of HIV, 2007
100%
250,000
UNGASS target for 2010
Number of women
200,000
90%
80%
70%
60%
150,000
50%
100,000
40%
30%
50,000
20%
10%
a
In
di
i
pi
a
hi
o
Et
al
aw
M
bi
a
Za
m
Ke
ny
a
da
ga
n
U
U
ni
te
d
R
ep
ub
lic
So
ut
h
N
Af
ig
e
ric
ria
of
Ta
nz
an
ia
M
oz
am
bi
qu
e
0%
a
0
UNGASS: United Nations General Assembly Special Session on HIV/AIDS in 2001
The bar indicates the uncertainty range around the estimate.
FIGURE 37-8 Percentage of pregnant women living with HIV receiving antiretroviral agents for preventing mother-to-child transmission
of HIV in the 10 countries with the highest estimated number of pregnant women living with HIV: 2007. (From World Health Organization:
Towards universal access: scaling up priority HIV/AIDS interventions in the health sector. Progress Report 2008. Available at www.who.int/hiv/
pub/2008progressreport/en. Accessed November 22, 2010.)
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
nevirapine resistance are paramount. One study published
in 2010 was terminated early because of the significant difference in nevirapine resistance and associated treatment
failure in infants exposed to single-dose perinatal nevirapine prophylaxis (Palumbo et al, 2010). Another study from
Uganda found 100% resistance to nevirapine in 6-monthold HIV-infected infants who had received 6 weeks of
daily nevirapine in addition to single-dose nevirapine
prophylaxis perinatally. Resistance patterns were less
prevalent in infants who had received only the single-dose
perinatal prophylaxis (16% at 6 months; p=0.005; Church
et al, 2008). Appropriate drug regimens for prophylaxis
against perinatal transmission in the face of viral mutation
and resistance patterns will likely remain a moving target
in high-, middle- and low-income countries.
Delivery Method
In 1999, two studies (one randomized clinical trial and one
metaanalysis) showed a decreased risk of vertical transmission of HIV with cesarean section before rupture of
membranes or onset of labor (European Mode of Delivery Collaboration, 1999; International Perinatal HIV
Group, 1999). This evidence was compelling enough for
the ACOG to issue new guidelines in August 1999 recommending that all HIV positive pregnant women be offered
elective cesarean at 38 completed weeks of gestation. The
current ACOG recommendations, updated in 2000 and
again in December 2010, recommend that all HIV-positive
pregnant women with plasma viral loads greater than 1000
copies per milliliter be “counseled regarding the benefits
of an elective cesarean delivery” in addition to a zidovudine infusion 3 hours before the operation (Jamieson et al,
2007). Currently, no study has answered whether elective
cesarean delivery further decreases the risk of transmission
in women with undetectable viral loads or in those receiving antiretroviral therapy.
Infant Feeding
In the United States, where replacement feeding (i.e.,
infant formula) is affordable and safe, it is recommended
that HIV-positive mothers avoid breastfeeding entirely
to minimize the estimated risk of 9% to 14% of postnatal HIV transmission through breast milk. Cell-associated
HIV has been detected in human breast milk even among
women receiving highly active antiretroviral therapy.
Globally, there is less clarity on best practices for infant
feeding. When formula feeding is not affordable, feasible, acceptable, sustainable, and safe, then breastfeeding
appears to be best for all infants of HIV-infected mothers.
Current World Health Organization guidelines updated in
2010 recommend that mothers known to be HIV-infected
either breastfeed and receive antiretroviral interventions
or avoid all breastfeeding. The guidelines further clarify
that feeding recommendations should be formulated at a
national or subnational level based on local epidemiology
and socioeconomic and infrastructure realities rather than
trying to modify feeding recommendations for each individual mother (World Health Organization, 2010).
Data from a randomized controlled trial in Zambia
showed that abrupt weaning at 4 months after birth versus
509
gradual weaning at the mother’s time of choice (median,
16 months) yielded no difference in HIV-free survival at
24 months of age (Kuhn et al, 2008). Among children who
became infected, early weaning increased mortality. This
finding contrasts with data from Kenya that showed an
increased risk of HIV transmission in breastfed infants at
24 months of age (37% risk of HIV infection in breastfed versus 21% risk in formula-fed infants; Nduati et al,
2000a). Most vertical transmission occurred during the first
6 months of breastfeeding, and the overall risk of transmission from breastfeeding was 16%. The 2-year all-cause
mortality was similar between the two groups in this study.
Safety of breastfeeding in low-resource settings can be
enhanced further by antiretroviral therapy for mothers,
breastfeeding infants, or both. A synthesis of related studies
was reviewed by Coovadia (2009). The mild-borne transmission of HIV-1C (MASHI) study in Botswana (Thior
et al, 2006), the Six Week Extended-Dose Nevirapine study
in India, Ethiopia, and Uganda (Six Week Extended-Dose
Nevirapine Study Team et al, 2008), the post-exposure
prophylaxis of infants (PEPI) study in Malawi (Kumwenda
et al, 2008), and the Mitra study in Tanzania (Kilewo et al,
2008) have all assessed the utility of prophylactic antiretroviral agents in breastfeeding infants. There was variation in
drug choice and duration, but most of these studies showed
decreased vertical transmission in breastfeeding infants on
some form of antiretroviral drug. A larger effect on vertical
transmission, however, is observed from continuation of
maternal antiretroviral therapy throughout the breastfeeding period. Maternal antiretroviral therapy in low-resource
settings has decreased the vertical transmission rates during breastfeeding to 1% to 5% (Coovadia, 2009).
Evaluation and Treatment of
HIV-Exposed Infants
Infant Testing
Infants born to HIV-infected mothers should have an initial serum evaluation by HIV-DNA or RNA PCR assay.
This test is most sensitive if performed initially at 2 weeks
of age; however, some physicians also do this testing in
the first few days after birth to rule out in utero infection,
particularly when the situation is high risk. High-risk situations would include unknown maternal HIV status with
concerning history or known maternal HIV infection without prophylactic antiretroviral treatment. A negative test
result in the first few days after birth requires confirmatory
testing at a later time. Table 37-5 shows the recommended
testing and evaluation schedule for an HIV-exposed infant
(Havens et al, 2009).
If a mother’s HIV status is unknown at the time of delivery, a rapid antibody test should be performed on mother
or baby. Test results should be available within a few
hours. If this test result is positive, prophylactic antiretroviral therapy should be initiated in the infant within 12
hours of birth, even in the absence of a confirmatory test.
Presumptive HIV-negative status of the infant is achieved
with two negative tests results, one at 2 weeks old or later
and the other at 4 weeks old or later. These are nucleic
acid amplification tests (NAATs) and can be either DNA
or RNA PCR. A definitive HIV-negative status is achieved
510
PART IX Immunology and Infections
TABLE 37-5 Evaluation and Treatment of the Infant Exposed to HIV-1 (Birth to 18 Months of Age), in Addition to Routine Pediatric Care
and Immunization
Infant Age
Action*
Birth
History and physical examination†
X
Assess risk of other infections
X
14 days
4 wk
6 wk
8 wk
4 mo
12-18 mo
X
X
X
X
Antiretroviral prophylaxis‡
X
X
X
X
Recommend against breastfeeding
X
X
X
X
Hemoglobin or complete blood cell count
X
HIV-1 DNA PCR or RNA assay¶
‖
X**
X§
X§
X
††
X
Initiate PCP prophylaxis‡‡
Enzyme immunoassay for antibody to HIV-1§§
X
From Havens PL et al: Evaluation and management of the infant exposed to HIV-1 in the United States, Pediatrics 123:175, 2009.
PCR, polymerase chain reaction; PCP, Pneumocystis jirovechii pneumonia;
*See text (in Havens et al, 2009) for detailed discussion of each action. If the infant has a diagnosis of HIV-1 infection during this period, laboratory monitoring and immunizations
should follow guidelines for treatment of pediatric HIV infection.
†Review maternal health information for possible exposure to coinfections. Frequency of examinations is determined, in part, by frequency of visits for immunizations in infancy.
‡Antiretroviral prophylaxis is initiated as soon as possible after birth, but certainly within 12 hours. Zidovudine prophylaxis is continued for 6 weeks.
§Checked at 4 weeks by some experts and rechecked at 8 weeks if the week 4 hemoglobin level is significantly low.
¶All HIV-1–exposed infants should undergo virologic testing for HIV-1 with HIV-1 DNA PCR or RNA assays at 14 to 21 days of age and, if results are negative, repeated at 1 to 2
and 4 to 6 months of age to identify or exclude HIV-1 infection as early as possible. Any positive test result at any age is promptly repeated to confirm the diagnosis of HIV-1 infection.
‖HIV-1 DNA PCR or RNA assay testing in the first few days of life allows identification of in utero infection and might be considered if maternal antiretroviral agents were not
administered during pregnancy or in other high-risk situations. A negative test result at this age requires repeated testing to exclude HIV-1 infection.
**If HIV-1 RNA or DNA testing of the newborn infant was not performed shortly after birth, or if such test results were negative, diagnostic testing with HIV-1 NAAT is delayed
until 14 to 21 days of age, because the diagnostic sensitivity of virologic assays increases rapidly by the age of 2 weeks. A negative test result at this age requires repeated testing
to exclude HIV-1 infection. Presumptive uninfected indicates negative nucleic acid amplification test (NAAT) result at ≥14 days and ≥4 weeks (1 month) of age; definitive uninfected
indicates negative NAAT result at ≥1 and ≥4 months of age.
††No NAAT is needed at 8 weeks of age if previous test results at 2 and 4 weeks of age were negative. A single negative NAAT result at 8 weeks identifies a presumptively uninfected
infant.
‡‡Infants with HIV-1 infection should be given PCP prophylaxis until 1 year of age, at which time infants are reassessed on the basis of age-specific CD4+ T-lymphocyte count and
percentage thresholds. Infants with indeterminate HIV-1 infection status should receive prophylaxis starting at 4 to 6 weeks of age until they are deemed to be presumptively or
definitively uninfected with HIV-1. Prophylaxis is not recommended for infants who meet criteria for presumptive or definitive lack of HIV-1 infection; therefore an NAAT at 2
and 4 weeks of age allows avoidance of PCP prophylaxis if both results are negative.
§§Many experts confirm the absence of HIV-1 infection with a negative HIV-1 antibody assay result at 12 to 18 months of age.
when a negative NAAT test obtained at 4 weeks old or later
is added to a negative NAAT results from 4 months old
or later (AAP Committee on Infectious Diseases, 2010b).
Many clinicians will also obtain an antibody test at 12 to
18 months after birth. By this time, an HIV-negative infant
should have cleared any passively acquired maternal antibodies and have a negative antibody test result, a process
referred to as seroreversion. If the infant’s antibody test is
positive at this time, an NAAT test should be repeated in
addition to close clinical evaluation for signs of infection.
Infant Treatment
Infants exposed to HIV should receive 6 weeks of prophylactic zidovudine (Table 37-6). It is important that families be given the entire quantity of medication at the time
of hospital discharge to avoid challenges for compliance
resulting from difficulty in filling a prescription (e.g., limited availability of the proper drug formulation, insurance
coverage issues).
If the mother has a positive antibody test and received
no intrapartum prophylactic zidovudine, some experts
would consider early initiation of additional infant antiretroviral medication. However, a careful analysis of the
pros and cons of potential toxicity versus decreased transmission risk is imperative. As stated by the Committee on
Pediatric AIDS, “If infant prophylaxis with antiretroviral
drugs in addition to ZDV is being considered, decisions
and choice of antiretroviral drugs should be determined
in consultation with a practitioner who is experienced in
care of infants with HIV infection,” (American Academy
of Pediatrics Committee on Pediatric AIDS, 2008).
If the infant has a positive NAAT test result before 6
weeks of age, antiretroviral treatment should be initiated
immediately. As stated in Red Book Online (AAP Committee on Infectious Diseases, 2010b), “Initiation of antiretroviral therapy is recommended for all HIV-1-infected
infants as soon as infection is confirmed, regardless of
clinical, immunologic, or virologic parameters.” Before
initiation, however, it is recommended that infants have
resistance testing, because infants can acquire resistant
virus from their mothers. Extensive guidelines exist for
selecting an appropriate combination drug regimen (available at: http://aidsinfo.nih.gov/ContentFiles/PediatricG
uidelines.pdf). A specialist in the treatment of pediatric
HIV infection should also be consulted. Although it was
once thought that treatment with antiretroviral therapy
should be delayed until an infant had signs of illness, it is
now known that infected infants have a decreased mortality when treatment is started early. Several studies in
South Africa have found that early antiretroviral therapy
slowed disease progression, and one study found a 76%
reduction in early infant mortality and a 75% reduction
in HIV disease progression when antiretroviral therapy
was initiated early (Violari et al, 2008).
CHAPTER 37 Viral Infections of the Fetus and Newborn and Human Immunodeficiency Virus Infection during Pregnancy
511
TABLE 37-6 Intrapartum Maternal and Neonatal Zidovudine Dosing for Prevention of Mother-to-Child Transmission of HIV
Dosing
Duration
Maternal Intrapartum
2 mg/kg IV over 1 hour, followed by continuous infusion of 1 mg/kg/h
Onset of labor until delivery
Neonatal
>35 weeks’ gestation: 2 mg/kg by mouth*,† (or 1.5 mg/kg IV) started within 6-12 h of delivery, then every 6 h
Birth to 6 wk
30-35 weeks’ gestation: 2 mg/kg by mouth (or 1.5 mg/kg IV) started within 6-12 h of delivery, then every 12 h,
advanced to every 8 h at 2 weeks of age
Birth to 6 wk
<30 weeks’ gestation: 2 mg/kg by mouth (or 1.5 mg/kg IV) started within 6-12 h of delivery, then every 12 h,
advanced to every 8 h at 4 weeks of age
Birth to 6 wk
From Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission: [Perinatal Guidelines] Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States, May 24, 2010; pp 1-117. http://aidsinfo.nih.gov/
ContentFiles/PerinatalGL.pdf. Accessed November 22, 2010.
IV, Intravenous.
*Zidovudine dosing of 4 mg/kg given every 12 h has been used for infant prophylaxis in some international perinatal studies. Although there are no definitive data to show equivalent
pharmacokinetic parameters or efficacy in preventing transmission, a regimen of zidovudine 4 mg/kg by mouth twice daily instead of 2 mg/kg by mouth four times daily may be
considered when there are concerns about adherence to drug administration to the infant.
†A simplified zidovudine dosing regimen has been developed for use in low resource settings. This regimen consists of 10 mg by mouth twice daily for infants weighing less than 2.5
kg at birth and 15 mg twice daily for infants weighing more than 2.5 kg at birth. This regimen could be considered for infants in higher resource settings born after 35 weeks’ gestation if simplicity in zidovudine dosing and administration is of prime importance.
Prophylaxis with trimethoprim-sulfamethoxazole against
opportunistic infections should also be initiated in any
infant with possible HIV infection at 6 weeks of age and
continued until at least 1 year of age. With a presumptive HIV-negative diagnosis, prophylaxis can be deferred.
Although the occurrence of opportunistic infections has
declined with more widespread access to testing and
antiretroviral therapy, Pneumocystis jirovechii pneumonia remains a common serious opportunistic infection in
HIV-1–infected infants, responsible for one third of pediatric AIDS diagnoses (AAP Committee on Infectious Diseases, 2010b). A Cochrane review that evaluated a study
in HIV-positive Zambian children found a 33% reduction
in mortality among children receiving cotrimoxazole prophylaxis versus placebo (Grimwade and Swingler, 2006).
Side Effects of Antiretroviral Therapy
Exposure in Infants
Antiretroviral therapy has been overwhelmingly successful in the prevention of vertical transmission of HIV, but
few human studies have assessed drug safety and toxicity
in infants (Thorne and Newell, 2007). Most antiretroviral
drugs are pregnancy category B or C; however, efavirenz
was changed to a D classification in 2005 when an association with infants with neural tube defects was seen (De
Santis et al, 2002; Fundarò et al, 2002).
A few case reports of mitochondrial toxicity (hyperlactatemia, some with additional motor or cognitive symptoms)
in HIV-uninfected, antiretroviral therapy–exposed infants
have come from Europe (Blanche et al, 1999; Noguera et
al, 2004; Tovo et al, 2005). Some of the symptoms resolved
or improved with time. However, large cohort studies in
the United States have not shown excess death owing to
mitochondrial dysfunction, although they might be underpowered to detect mild dysfunction.
One study done in Malawi showed a hypersensitivity reaction, primarily consisting of a rash, in 16 of 852
(1.9%) infants treated with nevirapine for 28 weeks. This
reaction was not seen in the control group, which received
only 1 week of zidovudine and lamivudine (Chasela et al,
2010).
Studies are inconsistent regarding the risk of prematurity in antiretroviral therapy–exposed infants. European
studies have shown an increased risk of prematurity in
infants born to mothers receiving antiretroviral therapy
regimens containing a protease-inhibitor drug (European
Collaborative Study and Swiss Mother and Child HIV
Cohort Study, 2000; Thorne et al, 2004). Some U.S. studies have not confirmed this association; however, a U.S.
study confirmed a significantly increased risk of very low
birthweight in infants born to women receiving proteaseinhibitor–containing antiretroviral therapy versus other
combination antiretroviral therapy (adjusted odds ratio,
3.56) (Tuomala et al, 2002).
Several studies have identified anemia in infants exposed
to prophylactic zidovudine; typically it is mild and transient, but occasionally severe anemia develops (Connor
et al, 1994; Taha et al, 2002; Wiktor et al, 1999). Slightly
longer term effects were seen in a French study in which
decreased levels of platelets, lymphocytes, and neutrophils
were persistent in the treatment group after adjusting for
several factors, including maternal CD4+ count and prematurity (Le Chenadec et al, 2003). Two European studies
have found neutropenia in children exposed to antiretroviral therapy up to 8 years of age (Bunders et al, 2005;
European Collaborative Study, 2004). These effects are
of uncertain clinical significance, but as prenatal and postnatal antiretroviral therapy exposure increases in duration
and number of drugs, studies to assess both subtle and significant sequelae in exposed infants will be crucial.
Special Case of HIV-Exposed
Uninfected Infants
An increasing area of research is focused on HIV-exposed,
uninfected (HIV-EU) infants. HIV-EU infants may be at
increased risk of death and morbidity. One study demonstrated a fivefold increased risk of death to the infant
512
PART IX Immunology and Infections
after the death of an HIV-infected mother (Newell et al,
2004). Other studies have found no increase in morbidity
among HIV-EU infants when compared with unexposed
infants (Taha et al, 2000). Whereas these findings may
simply reflect different levels of background disease burden among vulnerable children when their mother is ill or
dead, there may be an additional immunologic effect on the
infant from the mother’s HIV infection. In rural Kenya,
a study of tetanus antibody levels found a 22% reduction
in cord:maternal ratio of tetanus antibodies after adjusting for maternal vaccination and other factors in infants
born to HIV-positive mothers. An even greater reduction in infant antibody titers was seen with maternal HIV
and malaria coinfection (Cumberland et al, 2007). A large
study in Zambia found that HIV-EU infants had twice the
risk of death or severe morbidity when the mother’s CD4+
T-cell counts were low even when controlling for maternal mortality, separation due to maternal hospitalization,
lower birthweight, and other factors (Kuhn et al, 2005).
Another study documented an increase in mortality and
morbidity (e.g., respiratory infections) in HIV-EU infants
over community baseline in Latin America and the Carribean (Mussi-Pinhata et al, 2007). All these studies are,
however, limited by lack of a control group of unexposed
infants in the same study setting. Early evidence supporting the possibility of impaired neonatal immunity in HIVEU infants needs further investigation.
CONCLUSION
Pediatric HIV infection has become a disease of the developing world, where more than 90% of HIV-exposed and
more than 98% of HIV-infected children live. Without
therapy, morbidity and mortality are high for all HIVinfected individuals and are worse for children. Exposed,
uninfected children face the virtual certainty of becoming orphans by their tenth birthdays and a 50% chance of
some day becoming infected.
HIV transmission is preventable, whether sexual, vertical, or parenteral. Progression to AIDS and death is also
preventable. Although the arrest of all HIV transmission
remains an unfulfilled goal, potent antiretroviral combination therapy has dramatically decreased AIDS deaths and
virtually eliminated vertical transmission of the virus in the
United States and other high-income countries. Vigilance
in researching sustainable prevention measures and continued advocacy at national and international levels can
arrest HIV transmission worldwide.
SUGGESTED READINGS
Branson BM, Handsfield HH, Lampe MA, et al: Revised recommendations for
HIV testing of adults, adolescents, and pregnant women in health-care settings,
MMWR Recomm Rep 55:1, 2006.
Centers for Disease Control and Prevention: HIV Surveillance Report 20, 2008.
Available at www.cdc.gov/hiv/topics/surveillance/resources/reports/. Accessed
November 24, 2010.
Cheeran MC, Lokensgard JR, Schleiss MR: Neuropathogenesis of congenital
cytomegalovirus infection: disease mechanisms and prospects for intervention,
Clin Microbiol Rev 22:99-126, 2009.
Coovadia H: Current issues in prevention of mother-to-child transmission of HIV1, Curr Opin HIV AIDS 4:319, 2009.
Corey L, Wald A: Maternal and neonatal HSV infections, N Engl J Med 361:13761385, 2009.
Corey L, Wald A, Celum CL, et al: The effects of herpes simplex virus-2 on HIV-1
acquisition and transmission: a review of two overlapping epidemics, J Acquir
Immune Defic Syndr 35:435, 2004.
Ergaz Z, Ornoy A: Parovivus B19 in pregnancy, Reprod Toxicol 21:421-435, 2006.
Fiore S, Savasi V: Treatment of viral hepatitis in pregnancy, Expert Opin
Pharmacother 10:2801-2809, 2009.
Hall CB, Weinberg GA, Iwane MK, et al: The burden of respiratory syncytial virus
infection in young children, N Engl J Med 360:588-598, 2009.
Hall CB, Caserta MT, Schnabel KC, et al: Transplacental congenital human herpesvirus 6 infection caused by maternal chromosomally integrated virus, J Infect
Dis 201:505-507, 2010.
Hawkes MT, Vaudry W: Nonpolio enterovirus infection in the neonate and young
infant, Paediatr Child Health 10:383-388, 2005.
Kimberlin DW: Advances in the treatment of neonatal herpes simplex infections,
Rev Med Virol 11:157-163, 2001.
Lee JY, Bowden DS: Rubella virus replication and links to teratogenicity, Clin
Microbiol Rev 13:571-587, 2000.
Nassetta L, Kimberlin D, Whitley R: Treatment of congenital cytomegalovirus
infection: implications for future therapeutic strategies, J Antimicrob Chemother
63:862-867, 2009.
Palumbo P, Lindsey JC, Hughes MD, et al: Antiretroviral treatment for children
with peripartum nevirapine exposure, N Engl J Med 363:1510, 2010.
Panel on Antiretroviral Therapy and Medical Management of HIV-Infected
Children: Guidelines for the use of antiretroviral agents in pediatric HIV infection,
2010, pp 1-219. Available at http://aidsinfo.nih.gov/ContentFiles/PediatricGui
delines.pdf. Accessed November 22, 2010.
Panel on Treatment of HIV-Infected Pregnant Women and Prevention of
Perinatal Transmission: Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States, 2010, pp 1-117. Available at http://
aidsinfo.nih.gov/ContentFiles/PerinatalGL.pdf. Accessed November 22, 2010.
Plotkin SA: The history of rubella and rubella vaccination leading to elimination,
Clin Infect Dis 43(Suppl 3):S164-S168, 2006.
Prevention of respiratory syncytial virus infections: Indications for the use of palivizumab and update on the use of RSV-IGIV. Committee on Infectious Diseases
and Committee of Fetus and Newborn, Pediatrics 102:1211-1216, 1998.
Smith CK, Arvin AM: Varicella in the fetus and newborn, Semin Fetal Neonatal
Med 14:209-217, 2009.
Stagno S: Breastfeeding and the transmission of cytomegalovirus infections, Ital J
Pediatr 28:275-280, 2002.
Thorne C, Newell ML: Safety of agents used to prevent mother-to-child transmission of HIV: is there any cause for concern? Drug Saf 30:203, 2007.
Violari A, Coton MF, Gibb DM, et al: CHER Study Team: Early antiretroviral
therapy and mortality among HIV-infected infants, N Engl J Med 359:2233,
2008.
Volmink J, Siegfried NL, van der Merwe L, et al: Antiretrovirals for reducing the
risk of mother-to-child transmission of HIV infection, Cochrane Database Syst
Rev 1:CD003510, 2007.
World Health Organization: Towards universal access: scaling up priority HIV/AIDS
interventions in the health sector. Progress Report 2008. Available at www.who.int/
hiv/pub/2008progressreport/en/. Accessed November 22, 2010.
World Health Organization: Guidelines on HIV and infant feeding 2010. Principles
and recommendations for infant feeding in the context of HIV and a summary of
evidence. 2010. Available at www.who.int/child_adolescent_health/documents/
9789241599535/en/index.html.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
38
Toxoplasmosis, Syphilis, Malaria,
and Tuberculosis
Pablo J. Sánchez, Janna C. Patterson, and Amina Ahmed
TOXOPLASMOSIS
Toxoplasmosis is the disease state that results from infection with the obligate protozoan parasite Toxoplasma gondii. T. gondii is a coccidian that is ubiquitous in nature, and
the cat is the definitive host. The organism exists in the
following three forms:
1. An oocyst that is shed in cat feces from sporozoites
formed within the cat’s intestinal tract
2. A tachyzoite or endozoite that is the proliferative
form and was formerly referred to as a trophozoite
3. A tissue cyst that has an intracystic form termed
cystozoite or bradyzoite.
Nonfeline mammals or birds ingest infective oocysts
from contaminated soil. Tissue cysts then accumulate in
the organs and skeletal muscle of these animals. The possible routes of transmission from animal to human are direct
contact with cat feces, ingestion of undercooked meat containing infective cysts, and ingestion of fruits or vegetables
that have been in contaminated soil. Congenital infection
results from placental infection and subsequent hematogenous spread to the fetus.
EPIDEMIOLOGY
Toxoplasmosis is a worldwide medical problem. High
prevalence of infection has been documented in Europe,
Central and Latin America, and parts of Africa. However,
seroprevalence rates differ considerably from one country
to another, from one region of a country to another, and
even from one ethnic group to another in the same region.
These widely disparate seroprevalence rates among different adult populations throughout the world have been
explained by differences in eating and sanitation practices
that contribute to acquisition of infection. Eating undercooked or raw meat or unwashed raw fruits and vegetables,
drinking unpasteurized goat’s milk, working with meat,
having three or more kittens, and even certain climactic
conditions have been associated with higher risks of infection (Jones et al, 2009).
Among women of childbearing age in the United States,
the prevalence of antibody to T. gondii varies from approximately 3% to 30%, depending on the region of the country
(Boyer and McAuley, 1994; Remington et al, 2001). The
lowest seroprevalence rates have been found in the Mountain and Pacific states, and the highest rates have been seen
in the Northeastern and Southeastern United States. Seroprevalence rates for pregnant women seem to be decreasing, although regional and ethnic differences persist.
The prevalence of congenital infection in Massachusetts and New Hampshire has been documented to be 0.08
per 1000 births through immunoglobulin (Ig) M screening of newborn blood specimens collected on filter paper
(Guerina et al, 1994). This finding compares with a rate
of 3 to 10 per 1000 live births in Paris and Vienna, where
maternal seroprevalence rates of approximately 70% and
40%, respectively, are observed. In Massachusetts, a casecontrol study involving 14 years of newborn screening for
congenital toxoplasmosis found that the mother’s birth
outside the United States, particularly in Cambodia and
Laos, as well as the mother’s educational level and higher
gravidity were strongly predictive of congenital infection
(Jara et al, 2001). With approximately 4 million live births
annually in the United States, there are an estimated 400 to
4000 babies born each year with congenital toxoplasmosis
(Feldman et al, 2010).
NATURAL HISTORY
Infection of the fetus occurs as a consequence of maternal primary infection during pregnancy or, rarely, just
before conception (Villena et al, 1998b). Reactivation of
latent T. gondii infection during pregnancy does not lead
to fetal infection, except among immunocompromised
women such as those infected with the human immunodeficiency virus (HIV) or those undergoing chemotherapy
(Bachmeyer et al, 2006; Dunn et al, 1997; European Collaborative Study, 1996; Langer, 1983; Mitchell et al, 1990;
O’Donohoe et al, 1991; Remington et al, 2001). Under
these circumstances, however, the risk is low. In addition,
maternal reinfection can result in congenital toxoplasmosis
(Gavinet et al, 1997; Hennequin et al, 1997). Acute maternal infection, which is usually acquired early in pregnancy,
can lead to fulminant fetal infection resulting in stillbirth,
nonimmune fetal hydrops, preterm birth, and perinatal
death (Wong and Remington, 1994); however, chronic
T. gondii infection rarely has been associated with sporadic
abortion (Remington et al, 1964).
Infection of the fetus occurs transplacentally during
maternal parasitemia. Placental infection is an important
intermediary step, and up to 16 weeks may elapse between
placental infection and subsequent infection of the fetus.
This time delay has been termed the prenatal incubation
period (Remington et al, 2001). Congenital toxoplasmosis has
occurred in twins and triplets (Couvreur et al, 1991; Sibalic
et al, 1986; Wiswell et al, 1984). The clinical manifestations
are usually similar in monozygotic twins, whereas discrepancies in clinical findings are common in dizygotic twins.
Approximately 40% of infants born to mothers who
acquired toxoplasmosis during pregnancy are infected with
T. gondii. The rate of vertical transmission varies according to the trimester in which the mother became infected,
with fetal infection rates increasing as pregnancy advances
(Dunn et al, 1999; Remington et al, 2001; Wong and
Remington, 1994). Specifically, when maternal infection
513
514
PART IX Immunology and Infections
occurs in the first trimester, 15% of infants are infected,
whereas maternal infection in the second and third trimesters results in transmission rates of 30% and 60%, respectively. The severity of clinical manifestations is greatest,
however, when maternal infection is acquired early in
pregnancy. Maternal infection in the first trimester results
in severe disease in as many as 40% of infected fetuses,
and in still birth or perinatal death in an additional 35% of
infants. Approximately 15% of newborns have subclinical
disease; however, maternal infection in the third trimester
is rarely if ever associated with severe fetal disease or still
birth, and approximately 90% of infants in such situations
have subclinical infection.
Postnatally, transmission of T. gondii can occur from
transfusion of blood or blood products or from transplantation of organ or bone marrow from a seropositive donor
with latent infection. Although the organism has been
detected in human milk, transmission by breastfeeding has
not been documented.
The majority of newborns with congenital toxoplasmosis
lack clinical signs of infection, although thorough evaluation may demonstrate eye or neurologic abnormalities in
approximately 20% of cases. Clinically apparent disease is
present in approximately 10% to 25% of infected infants
(Alford et al, 1969, 1974; Guerina et al, 1994). The clinical manifestations of toxoplasmosis are often indistinguishable from those seen with other congenital infections, such
as cytomegalic inclusion disease and congenital syphilis.
Approximately one third of infants have a generalized form
of the disease that principally involves organs of the reticuloendothelial system. The abnormalities include temperature
instability, hepatosplenomegaly, jaundice, pneumonitis,
generalized lymphadenopathy, rash, chorioretinitis, anemia,
thrombocytopenia, eosinophilia, and abnormal cerebrospinal fluid (CSF) indices (Table 38-1; Boyer and McAuley,
1994; Eichenwald, 1960). The other two thirds of infected
infants principally manifest neurologic disease.
Central nervous system involvement is the hallmark of
congenital T. gondii infection (Diebler et al, 1985; McAuley et al, 1994; Remington et al, 2001). Chorioretinitis,
intracranial calcifications, and hydrocephalus are the most
characteristic findings, occurring in approximately 86%,
37%, and 20% of symptomatic infants, respectively (see
Table 38-1) (Boyer and McAuley, 1994; Eichenwald, 1960;
Remington et al, 2001). This constellation of findings has
been referred to as the classic triad of congenital toxoplasmosis; its presence should alert the clinician to the diagnosis.
Intracranial calcifications may be single or multiple, but
typically are generalized and located in the caudate nucleus,
choroid plexus, meninges, and subependyma (Müssbichler,
1968); they also may occur periventricularly, as in cytomegalovirus infection. They are visualized best by computed
tomography (CT), but are often detected on ultrasonography as well. Intracranial calcifications may resolve with
appropriate antimicrobial therapy (McAuley et al, 1994).
Hydrocephalus may be the only manifestation of disease;
it results from the extensive periaqueductal and periventricular vasculitis with necrosis that causes obstruction
of the ventricular system. Ventriculoperitoneal shunting is often required (Martinovic et al, 1982; McAuley et
al, 1994). Abnormalities of the CSF are common; characteristically, they consist of lymphocytic pleocytosis
TABLE 38-1 Clinical Findings Among Infants With Congenital
Toxoplasmosis
Infants with Findings (%)
Neurologic
Disease*
(108 Cases)
Generalized
Disease†
(44 Cases)
Chorioretinitis
94
66
Abnormal cerebrospinal fluid
55
84
Anemia
51
77
Convulsions
50
18
Intracranial calcification
50
4
Jaundice
29
80
Hydrocephalus
28
0
Fever
25
77
Splenomegaly
21
90
Lymphadenopathy
17
68
Hepatomegaly
17
77
Vomiting
16
48
Microcephaly
13
0
Diarrhea
6
25
Cataracts
5
0
Eosinophilia
4
18
Abnormal bleeding
3
18
Hypothermia
2
20
Glaucoma
2
0
Optic atrophy
2
0
Microphthalmia
2
0
Rash
1
25
Pneumonitis
0
41
Finding
Adapted from Remington JS, McLeod R, Thulliez P, Desmonts G: Toxoplasmosis. In
Remington JS, Klein JO, editors: Infectious diseases of the fetus and newborn infant, ed 5,
Philadelphia, 2001, WB Saunders, p 246.
*Infants with otherwise undiagnosed central nervous system diseases in the first year
of life.
†Infants with otherwise undiagnosed non-neurologic diseases during the first 2 months
of life.
and a markedly elevated protein content. Microcephaly,
when present, indicates severe brain injury. Hypothermia
and hyperthermia may occur secondary to hypothalamic
involvement. T. gondii has been detected in the inner ear
and mastoid, with the associated inflammation resulting in
deafness. An ascending flaccid paralysis with myelitis has
also been reported (Campbell et al, 2001).
Chorioretinitis secondary to congenital toxoplasmosis
can manifest at any age. It usually manifests as strabismus
in infants. Defects in visual acuity are more common in
older children. Typically the eye lesion consists of a focal
necrotizing retinitis that is often bilateral with involvement of the macula and even the optic nerve. Complications include blindness, iridocyclitis, and cataracts (Arun
et al, 2007; Phan et al, 2008).
Other less common manifestations of congenital toxoplasmosis are nonimmune hydrops fetalis, myocarditis,
nephrotic syndrome, and immunoglobulin abnormalities
with both hypergammaglobulinemia and hypogammaglobulinemia described. Bony abnormalities consisting of
metaphyseal lucencies similar to those seen in congenital
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
syphilis have also been reported (Milgram, 1974). A variety
of endocrine abnormalities may occur, including hypothyroidism, diabetes insipidus (Oygur et al, 1998; Yamakawa
et al, 1996), precocious puberty, and growth hormone
deficiency.
DIAGNOSIS
Isolation of T. gondii from body fluids and tissues provides
definitive evidence of infection. The organism can be isolated from placenta, amniotic fluid, fetal blood obtained
by cordocentesis, umbilical cord blood, infant peripheral
blood, and CSF by means of intraperitoneal and subcutaneous inoculation into laboratory mice (Foulon et al,
1999a; Remington et al, 2001; Wong and Remington,
1994). Mouse inoculation may require as long as 4 to 6
weeks for demonstration of the parasite. Although it is not
a practical method, isolation of the organism should be
attempted whenever possible. It is available at the Toxoplasma Serology Laboratory, Palo Alto Medical Foundation (860 Bryant Street, Palo Alto, California 94301;
telephone: 415-326-8120). In addition, tissue culture has
been used to isolate T. gondii from amniotic fluid.
Histopathologic examination of the placenta and tissues obtained at postmortem examination or by biopsy
from stillborns or infants should be performed because the
specimens may demonstrate the presence of tachyzoites.
In addition, tachyzoites have been demonstrated in CSF,
ventricular fluid, and aqueous humor by specialized staining techniques.
Polymerase chain reaction (PCR) analysis has been used
successfully to detect T. gondii DNA in amniotic fluid, placenta, CSF, brain, urine, and fetal and infant blood (Foulon
et al, 1999a; Fricker-Hidalgo et al, 1998; Grover et al, 1990;
Guy et al, 1996; Hohlfeld et al, 1994; Jenum et al, 1998;
Romand et al, 2001). PCR performed on amniotic fluid
obtained by amniocentesis has become the preferred method
of confirming in utero infection (Kasper et al, 2009). Falsenegative results have been reported, however, and interlaboratory variability in performance of PCR assays has been
documented (Guy et al, 1996; Romand et al, 2001). PCR
performed on neonatal CSF is recommended for the evaluation of possible central nervous system involvement.
Serologic assays for measurement of antibodies to
T. gondii in serum and body fluids are the most widely used
methods of diagnosing congenital toxoplasmosis (Boyer,
2001; Boyer and McAuley, 1994; Dannemann et al, 1990;
Foudrinier et al, 1995; Guerina et al, 1994; Lappalainen
et al, 1993; Madi et al, 2010; Naessens et al, 1999; Naot
et al, 1991; Pinon et al, 2001; Rabilloud et al, 2010;
Remington et al, 1985; Robert-Gangneux, 2001; Robert-
Gangneux et al, 1999a, 1999b; Villena et al, 1999; Wong and
Remington, 1994). These tests are commercially available at
the Toxoplasma Serology Laboratory. The more commonly
used tests that detect T. gondii-specific IgG antibodies are
the Sabin-Feldman dye test, which is considered the gold
standard but requires live organisms, indirect immunofluorescent antibody test, IgG enzyme-linked immunosorbent
assay (ELISA), direct agglutination, and IgG avidity test.
In addition, a differential agglutination test has been
developed as a confirmatory test to differentiate acute
from chronic maternal infection. This test compares the
515
IgG serologic titer obtained with the use of formalinfixed tachyzoites (HS antigen) with those obtained with
acetone- or methanol-fixed tachyzoites (AC antigen). The
latter preparation contains stage-specific T. gondii antigens
that are recognized by IgG antibodies only during early
infection. An additional assay to assist in ruling out maternal infection acquired in the first 3 months of pregnancy
is the IgG avidity test performed by the ELISA technique.
This test is based on the principle that, although the antibody-binding avidity or affinity for an antigen is initially
low after primary antigenic stimulation, IgG antibodies
that are present from previous antigenic stimulation are
usually of high avidity. Therefore a high-avidity result in
the first trimester would exclude an infection acquired in
the previous 12 weeks. Finally, an enzyme-linked immunofiltration assay has been developed that allows discrimination between IgG antibodies of maternal origin and IgG
synthesized by the fetus as well as identification of antibody subtypes in infected neonates (Zufferey et al, 1999).
Tests that detect T. gondii-specific IgM are (1) the double-sandwich IgM ELISA, which has a sensitivity of 75%
to 80% and a specificity of 100% (Guerina et al, 1994);
(2) the IgM immunosorbent agglutination assay, which
is the most sensitive test but should not be performed on
umbilical cord blood, because even small quantities of
maternal IgM antibodies contaminating the specimen will
yield a false-positive result (Boyer and McAuley, 1994);
and (3) the IgM immunofluorescent antibody test. The last
test is not recommended because it has a much lower sensitivity than either the IgM ELISA or IgM immunosorbent
agglutination assay and it has poor specificity secondary to
rheumatoid factors and antinuclear antibodies, contributing to false-positive results. Other tests that are still being
investigated include a T. gondii-specific IgA ELISA and IgA
immunofiltration assay; a T. gondii-specific IgE immunofiltration assay; and IgG, IgM, and IgA immunoblotting tests.
Because the majority of adults with acquired T. gondii
infection are asymptomatic, evaluation of the pregnant
woman and fetus is usually prompted by either seroconversion or an elevated maternal Toxoplasma spp. IgG titer
(Couvreur et al, 1988; Daffos et al, 1988; Montoya et al,
2008). The latter may reflect chronic or past infection;
therefore the acuity of the maternal infection is determined
serologically with the HS-AC differential agglutination
test where agglutination titers to formalin-fixed tachyzoites (HS antigen) are compared with titers against acetoneor methanol-fixed tachyzoites (AC antigen). In general, an
acute pattern demonstrates high AC and HS titers, while
a nonacute pattern demonstrates high AC titers and low
HS titers. This method can differentiate an acute from a
remote infection in pregnant women, whereas IgM and
IgA antibodies detectable by ELISA or ISAGA are elevated
for prolonged periods. If recent maternal infection is documented by an acute pattern on the HS-AC test, seroconversion, or rising IgG antibody titers, the fetus should be
evaluated by ultrasonography, and amniotic fluid should
be tested for specific Toxoplasma spp. DNA with PCR.
PCR has supplanted the need for cordocentesis, and a positive result confirms fetal infection (Hohlfeld et al, 1994).
Postnatally, serologic testing of paired maternal and infant
sera should be performed at a reliable laboratory that will
include assays for Toxoplasma spp. IgG and IgM antibodies.
516
PART IX Immunology and Infections
Neonatal serum for Toxoplasma spp. IgA determination by
ELISA also should be considered, because it may yield the
only positive result in some infants.
Subinoculation of placental tissue, amniotic fluid, and
umbilical cord blood into mice should be considered. If
results of these tests suggest possible infection, the newborn
should be evaluated fully with complete blood cell count
and platelet determination, liver function tests, CSF evaluation (including tests for IgG and IgM antibodies and PCR;)
(Wallon et al, 1998), cranial ultrasound or CT imaging of
the head, ophthalmologic examination, and hearing evaluation. The presence of neonatal IgM antibody in serum or
CSF, or a positive PCR result for blood or CSF, indicates
congenital infection. In addition, at-risk infants should
undergo serologic follow-up to detect rising serum IgG
titers during the 1st year of age or persistence of IgG antibody beyond 12 to 15 months of age, when maternal IgG
antibody has disappeared (Robert-Gangneux et al, 1999a,
1999b). Uninfected infants show a continuous decline in
T. gondii IgG titer with no detectable IgM or IgA antibodies.
Low IgG titers and an HS-AC differential agglutination test that indicate remote maternal infection do not
require further evaluation of the mother or infant unless
the mother is infected with HIV. Because fetal infection
has occurred during chronic T. gondii infection in HIVinfected pregnant women, their infants should be evaluated
serologically at birth for evidence of congenital infection.
It has been suggested that HIV-infected pregnant women
who have low CD4+ T lymphocyte counts and who are
seropositive for T. gondii antibody receive prophylaxis to
prevent fetal infection (Beaman et al, 1992; Wong and
Remington, 1994). However, insufficient data currently
are available to recommend that such therapy be given
routinely for this indication. Nevertheless, if such women
previously have had toxoplasmic encephalitis, prophylaxis
with pyrimethamine, sulfadiazine, and leucovorin (folinic
acid) should be considered (Masur et al, 2002).
THERAPY
It is currently recommended that fetuses and infants
younger than 1 year who are infected with T. gondii
receive specific therapy effective against this congenital
pathogen, even if they have no clinical signs of disease
(Couvreur et al, 1988; Daffos et al, 1988; Foulon et al,
1999b; Friedman et al, 1999; Gilbert et al, 2001; Hohlfeld
et al, 1989; Koppe et al, 1986; McAuley et al, 1994; McGee
et al, 1992; McLeod et al, 2009; Peyron and Wallon, 2001;
Roizen et al, 1995; Vergani et al, 1998; Wallon et al, 1999;
Wong and Remington, 1994). On the basis of comparison
with untreated historical controls, outcome is improved
substantially by neonatal treatment. The effectiveness of
maternal and fetal treatment is less clear. Spiramycin has
been used in pregnant women with acute toxoplasmosis
to reduce transplacental transmission of T. gondii. If fetal
infection is confirmed after the 17th week of pregnancy,
however, treatment with pyrimethamine, sulfadiazine,
and folinic acid is recommended. Prenatal treatment of
congenital toxoplasmosis is believed to reduce the clinical severity of infection in the newborn while shifting the
disease to a more subclinical form. This effect in turn may
ameliorate the long-term neurologic complications that
are commonly seen among infants who have clinical manifestations in the neonatal period. A recent metaanalysis
of the effectiveness of prenatal treatment of toxoplasmosis infection found no evidence that such treatment significantly decreased clinical manifestations of disease in
infected infants (Thiebaut et al, 2007).
Neonatal treatment has also resulted in reductions in
sensorineural hearing loss and neurodevelopmental and
visual handicaps. Table 38-2 shows the recommended
guidelines for the treatment of congenital toxoplasmosis.
In infants with congenital toxoplasmosis, the treatment
consists of pyrimethamine, sulfadiazine, and folinic acids
(Boyer and McAuley, 1994; McAuley et al, 1994; McLeod
et al, 1992; Remington et al, 2001). The actual duration
of therapy is not known, although prolonged courses of at
least 1 year are preferred. Currently most experts recommend combined treatment until the patient is 1 year old
(Remington et al, 2001; Villena et al, 1998a, 1998b).
Complete blood cell counts and platelet determination
must be monitored closely while the patient is receiving
therapy, because granulocytopenia, thrombocytopenia,
and megaloblastic anemia can occur. These parameters
usually improve once a higher dose of folinic acid is administered or pyrimethamine and sulfadiazine are discontinued
temporarily. The indications for adjunctive therapy with
corticosteroids such as prednisone (0.5 mg/kg twice per
day) are CSF protein concentration 1 g/dL or higher and
chorioretinitis that threatens vision; corticosteroid treatment is continued until either condition resolves. Current
therapies are not effective against encysted bradyzoites and
therefore might not prevent reactivation of chorioretinitis
and neurologic disease.
PROGNOSIS
Maternal toxoplasmosis acquired during the first and
second trimesters has been associated with still birth
and perinatal death secondary to severe fetal infection in
approximately 35% and 7% of cases, respectively. Among
infants born with congenital toxoplasmosis, the mortality rate has been reported to be as high as 12%. In addition, infants with congenital toxoplasmosis are at high risk
for ophthalmologic, neurodevelopmental, and audiologic
impairments, including mental retardation (87%), seizures
(82%), spasticity and palsies (71%), and deafness (15%)
(Eichenwald, 1960; Hohlfeld et al, 1989; Koppe et al,
1986; McAuley et al, 1994). Of neonates with subclinical
infection, long-term follow-up reveals eye or neurologic
disease in as many as 80% to 90% by the time they reach
adulthood (Couvreur and Desmonts, 1962; Couvreur et al,
1984; McLeod et al, 2000; Saxon et al, 1973; Wilson et al,
1980). Data from the United States National Collaborative Treatment Trial show that treatment of neonates with
congenital toxoplasmosis early and for 1 year resulted in
more favorable outcomes than were reported for untreated
infants or infants who were treated for only 1 month.
PREVENTION
Pregnant women whose serologic status for T. gondii is
negative or unknown, as well as women who are attempting to conceive, should be educated on the prevention
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
517
TABLE 38-2 Treatment Guidelines for Toxoplasmosis
Condition
Therapy
Dose (Oral Unless Specified)
Duration
Pregnant woman with acute
toxoplasmosis
Spiramycin for first 21 wk of
gestation or until term if fetus
not infected*
1 g every 8 h without food
Until fetal infection documented
or excluded at 21 wks; if fetal
infection documented, replaced
with pyrimethamine, leucovorin, and sulfadiazine (see below)
Pyrimethamine (if fetal infection
confirmed after 18th week of
gestation or if infection acquired
in last few weeks of gestation) and
Loading dose: 100 mg/day in two
divided doses for 2 days followed by
50 mg/day
Until delivery
Sulfadiazine* and
Loading dose: 75 mg/kg/day in two
divided doses (maximum, 4 g/d) for
2 days; then 100 mg/kg/day in two
divided doses (maximum, 4 g/day)
Until delivery
Leucovorin†
10-20 mg/d
Until delivery
Pyrimethamine and
Loading dose: 2 mg/kg/day for 2 days;
then 1 mg/kg/day for 2 or 6 months;
then 1 mg/kg/day on Mon, Wed,
and Fri each week
≥1 yr
Congenital Toxoplasma gondii
infection in infant
≥1 yr
Sulfadiazine* and
100 mg/kg/day in 2 daily divided doses
Leucovorin (folinic acid)†
10 mg 3 times weekly
Corticosteroids (prednisone)‡
≥1 yr
1 mg/kg/daily in 2 daily divided doses
Until resolution of elevated (≥1
g/dL) CSF protein or active chorioretinitis that threatens vision
Data from Boyer KM, McAuley B: Congenital toxoplasmosis, Semin Pediatr Dis 5:42, 1994; and Remington JS, McLeod R, Thulliez P, Desmonts G: Toxoplasmosis. In Remington
JS, Klein JO, editors: Infectious diseases of the fetus and newborn infant, ed 5, Philadelphia, 2001, WB Saunders, p 293.
CSF, Cerebrospinal fluid.
*Available only on request from the U.S. Food and Drug Administration (301-827-2127; fax, 301-927-2475).
†Monitor blood and platelet counts weekly; adjust dosage for megaloblastic anemia, granulocytopenia, or thrombocytopenia.
‡When signs of inflammation or active chorioretinitis have subsided, dose can be tapered and eventually discontinued; use only in conjunction with pyrimethamine, sulfadiazine,
and leucovorin.
of congenital toxoplasmosis through avoidance of at-risk
behaviors that may expose them to cat feces or encysted
bradyzoites in raw meat (Centers for Disease Control and
Prevention, 2000; Eskild et al, 1996; Foulon et al, 2000;
Jones et al, 2001; Wilson and Remington, 1980). Such
women should be taught to wear gloves when changing
cat litter boxes or gardening and to wash hands after such
activities. Daily changing of cat litter will also decrease
the chance of infection, because oocysts are not infective during the first 1 to 2 days after passage. In addition,
feeding cats commercially prepared foods rather than
undercooked meats or wild rodents reduces the likelihood
of their becoming infected and capable of transmitting the
infection to a pregnant woman. Oral ingestion of T. gondii
can be prevented by either cooking meat to well done,
smoking it, or curing it in brine, and by washing kitchen
surfaces that come into contact with raw meat. Vegetables
and fruits should be washed, and hands and kitchen surfaces should be cleaned after handling fruits, vegetables,
and raw meat. Flies and cockroaches may serve as transport hosts for T. gondii, so their access to food must be
prevented.
Routine serologic screening of women during pregnancy has been an effective means of prevention in such
countries as France and Austria, where the incidence of
congenital toxoplasmosis is high. No such screening is
currently recommended in the United States. However,
high-risk women, including those who are immunocompromised, should be screened early in pregnancy.
Neonatal screening for IgM antibody has also been advocated so that asymptomatic infants can be detected and
treated before neurologic symptoms develop (Peterson
and Eaton, 1999). This strategy, however, has been hampered by the lack of readily available and reliable IgM test
kits. Moreover, such screening will not detect the approximately 25% of infected infants who lack anti-Toxoplasma
spp. IgM antibody. Further study involving cost analyses
is needed to define the best preventive strategy for congenital toxoplasmosis in specific populations, regions, and
countries.
SYPHILIS
Syphilis is caused by infection with the spirochete
Treponema pallidum. In adults, this spirochete is transmitted through sexual contact, but infants acquire the
infection from their mothers, either in utero or during delivery. The history of syphilis epidemics is one
of intermittent peaks. During the 1930s and 1940s in
the congenital syphilis clinic of the Harriet Lane Home
(Baltimore, Md.), 60 to 80 infants and children attended
each week for arsenic therapy. Many more were lost to
follow-up before completing their 2- to 3-year course
of treatment. It was unusual if fewer than three or four
new examples were discovered in the general outpatient
department in the course of 1 week. Then, for several
decades, the frequency of new cases of congenital syphilis declined.
518
PART IX Immunology and Infections
INCIDENCE AND EPIDEMIOLOGY
After years of declining incidence, congenital syphilis is
again on the rise. Recent Centers for Disease Control and
Prevention (CDC) surveillance data show a 23% increase
in congenital syphilis cases from 2005 to 2008 as shown in
Figure 38-1 (Centers for Disease Control and Prevention,
2010a). This increase is directly linked to a rise in primary
and secondary syphilis in women from 2004 to 2007. In
2008, there was a total of 431 congenital syphilis cases in
the U.S. Half of these cases were in infants born to black
mothers, primarily in the South. Approximately 30% of
the mothers of these infected infants did not receive prenatal care. When mothers did receive prenatal care and their
infants still became infected, 27% were screened <30 days
before delivery (likely resulting in a false negative test) and
24% screened positive but were not treated (Centers for
Disease Control and Prevention, 2010a). An outbreak was
also reported among Pima Indians in Arizona in 2007-2009
in which a total of 106 cases were identified, including six
congenital cases, two of which resulted in stillbirth (Centers for Disease Control and Prevention, 2010b). A similar
epidemic was seen in Alabama with a peak of 238 cases in
2006 with the largest increase seen in heterosexual women
(Centers for Disease Control and Prevention, 2009).
Worldwide more newborns are affected by congenital
syphilis than by any other neonatal infection (Schmid,
2004). Countries such as Ethiopia, Swaziland, and Mozambique have prevalence rates of maternal syphilis as high as
12% to 13% (World Health Organization, 2007). There
are an estimated 2 million pregnancies affected annually,
and most women infected with syphilis within 1 year of
their pregnancy will transmit the infection to their infant.
Adverse outcomes in these pregnancies are severe: 17% to
40% result in stillbirth, 10% to 23% in neonatal death,
and 10% to 30% result in congenital syphilis infection
(World Health Organization, 2007).
The global problem of congenital syphilis is further
confounded by the high prevalence of HIV, which reaches
25% to 40% in some African cities. Mothers who are coinfected with syphilis and HIV may be less likely to respond
to penicillin treatment, which increases the risk of congenital syphilis in the fetus (Lukehart et al, 1988). In addition,
HIV-infected mothers with untreated syphilis may be at
increased risk of transmitting HIV to their fetus secondary
to a placentitis that allows the virus to pass from maternal to fetal circulation (Pollack et al, 1990). In one study
of HIV-infected women from Tanzania, coinfection with
syphilis doubled the risk of still birth (Kupka et al, 2009).
The extent of negative synergy between these two infections will become increasingly clear as research progresses.
PREVENTION
Screening for syphilis in pregnancy is the primary method
of preventing congenital syphilis. The American Academy
of Pediatrics and the American Congress of Obstetricians
and Gynecologists in Guidelines for Perinatal Care (2002)
recommend screening all pregnant women at the first prenatal visit, after exposure to an infected partner, and at
delivery. Reinforcing this recommendation is the Reaffirmation Recommendation Statement by the U.S. Preventive
Services Task Force (2009) that “Grade A evidence” is present for screening all pregnant women for syphilis infection.
As summarized by Miller and Karras (2010) in reference
to the latest CDC surveillance data, “the increase in the
50
8
40
6
30
4
20
2
10
0
P&S rate (per 100,000 females)
CS rate (per 100,000 live births)
CS rate
P&S syphilis rate among females
0
1995
1997
1999
2001
2003
Year
2005
2007
* CS rates from 1995 to 2006 were calculated using yearly live birth data as denominators.
Rates for 2007 and 2008 were calculated using live birth data for 2006. Available at http://
www.cdc.gov/nchs/births.htm.
† P&S syphilis rates were calculated using bridged race population estimates for 2000-2007
based on 2000 U.S. Census counts. Available at hhtp://wonder.cdc.gov/wonder/help/
bridged-race.html.
FIGURE 38-1 Congenital syphilis (CS) rate among infants less than 1 year old and the rate of primary and secondary syphilis (P&S) among females
10 years or older. National Electronic Telecommunication System for Surveillance, United States, 1995-2008. (Data from Centers for Disease Control
and Prevention: Congenital syphilis: United States, 2003-2008, MMWR Morb Mortal Wkly Rep 59:413, 2010.)
519
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
congenital syphilis rate, the substantial burden of primary
and secondary syphilis among black women in the south,
and the high case/fatality ratio associated with congenital syphilis require that congenital syphilis prevention be
given high priority in areas with high syphilis morbidity
and evidence of heterosexual syphilis transmission.”
ETIOLOGY AND PATHOGENESIS
The organism responsible for syphilis is Treponema pallidum.
This delicate, corkscrew-shaped, flagellated, highly motile
spirochete is almost identical in appearance to Treponema
pertenue, which causes yaws. Because Treponema spp. enter
the fetal bloodstream directly, the primary stage of infection is completely bypassed. There is no chancre and no
local lymphadenopathy. Instead, the liver, the immediate
target of the invasion, is flooded with organisms that then
penetrate all the other organs and tissues of the body to a
lesser degree. Other sites of invasion include skin, mucous
membranes of the lips and anus, bones, and the central nervous system. If fetal invasion has taken place early, the lungs
may be heavily involved in a characteristic pneumonia alba,
but this condition is usually life threatening. Treponema
spp. may be found in almost any other organ or tissue of
the body, but seldom cause inflammatory and destructive
changes in loci other than the ones named previously.
Under the microscope, the tissue alterations consist of
nonspecific interstitial fibrosis with or without evidence of
low-grade inflammatory response in the form of round cell
inflammation. Necrosis follows fairly regularly in bone,
but only rarely in other tissues. Localization and gumma
formation are not common in the neonate; however, extramedullary hematopoiesis in the liver, spleen, kidneys, and
other organs can be seen.
Syphilis in infants is acquired primarily by transplacental
transmission, which can occur at any time during pregnancy
but ordinarily occurs during 16 to 28 weeks’ gestation.
Fetuses infected early may die in utero or are at high risk
for significant neurodevelopmental morbidity. The usual
outcome of a third-trimester infection is the birth of an
apparently normal infant who becomes ill within the first
few weeks of life. Virtually all infants born to women with
primary or secondary infection have congenital infection,
but approximately half are clinically symptomatic. As hospitals and insurers strive to cut costs, early discharge of new
mothers has hindered identification of congenitally infected,
asymptomatic infants whose mother’s infections occurred
late in the third trimester and whose syphilis serologic test
results are not yet positive at the time of delivery (Dorfman and Glaser, 1990). It is critical that at-risk infants have
a source of primary health care capable of tracking both
maternal and infant syphilis status (Chhabra et al, 1993;
Zenker and Berman, 1991). Early latent infection results in
a 40% infant infection rate, and late latent infection results
in a 6% to 14% infant infection rate (Wendel, 1988).
CLINICAL PRESENTATION
Congenital syphilis is challenging to diagnose in neonates because 60% of infected infants are asymptomatic.
Early presentation is typically within the first 4 weeks
after birth and is characterized by the signs and symptoms
summarized in Table 38-3. Persistent rhinitis (“snuffles”)
was estimated to occur in two thirds of patients in the early
literature, but is now less prevalent (Ingall et al, 2006).
Prematurity and low birthweight is seen in 10% to 40% of
infants (Saloojee et al, 2004). Additional diagnoses associated with congenital syphilis include nonimmune hydrops,
nephrotic syndrome, and myocarditis.
Cutaneous lesions can appear at any time from the
2nd week after birth and onward. These copper-colored
lesions can be either sparse or numerous; round, oval, or
iris-shaped; and circinate or desquamative (Figure 38-2).
Even more characteristic than their appearance is their
distribution, which most frequently includes the perioral,
perinasal, and diaper regions. Palms and soles are also
involved, but the rash is soon replaced there by diffuse
reddening, thickening, and wrinkling. In heavily infected
infants, the rash may become generalized. Mucocutaneous junctions become involved in typical fashion. The
lips become thickened and roughened and tend to weep.
TABLE 38-3 Clinical Features of Congenital Syphilis in the
Neonatal Period
Feature
Prevalence (%)
Hepatomegaly with or without splenomegaly
33-100
Radiographic bone changes
75-100
Lymphadenopathy
50
Blistering skin rash
40
Respiratory distress
34
Jaundice
33
Pseudoparalysis of Parrot
12
Fever
16
Bleeding
10
Adapted from Remington JS, Klein JO, Wilson CB, et al, editors: Infectious diseases of
the fetus and newborn infant, ed 6, Philadelphia, 2006, Saunders, p 572 and Saloojee H,
Velaphi S, Goga Y, et al: The prevention and management of congenital syphilis: an
overview and recommendations, Bulletin of the World Health Organization 82, 2004.
FIGURE 38-2 Congenital syphilis with desquamation over the hand.
(From American Academy of Pediatrics Committee on Infectious Diseases:
Syphilis: clinical manifestations images. Red Book Online Visual
Library, 2009, American Academy of Pediatrics. Available at http://
aapredbook.aappublications.org/cgi/content/figsonly/2009/1/3.129. Accessed
November 19, 2010.)
520
PART IX Immunology and Infections
Radial cracks appear that traverse the vermillion zone up
to and a bit beyond the mucocutaneous margins of the lips.
These cracks are the beginnings of the radiating scars that
may persist for many years as rhagades. Similar mucocutaneous lesions involve the anus and vulva, but in these
locations, the white, flat, moist, raised plaques known as
condylomata are also encountered, although less frequently.
Radiographs of the bones show characteristic osteochondritis and periostitis in 80% to 90% of infants with
symptomatic congenital syphilis (Figure 38-3). In most
cases, the bone lesions are asymptomatic, but in a few they
are severe enough to lead to subepiphyseal fracture and
epiphyseal dislocation with an extremely painful pseudoparalysis of one or more extremities. Approximately 20% of
asymptomatic, congenitally infected infants have metaphyseal changes consistent with congenital syphilis. Radiographic alterations include an unusually dense band at the
epiphyseal ends, below which is a band of translucency
whose margins are at first sharp but that later become serrated, jagged, and irregular. The shafts become generally
more opaque, but spotty areas of translucency throughout
may give them a moth-eaten look. The periosteum of the
long bones becomes more thickened. Epiphyses separate
because the dense end plate breaks away from the shaft by
fracture through the subepiphyseal zone of decalcification.
Pseudoparalysis of Parrot can occur because of bone pain
from these changes.
Signs of visceral involvement include hepatomegaly,
splenomegaly, and general glandular enlargement. Palpable
epitrochlear nodes are not pathognomonic, but are highly
suggestive of congenital syphilis. The liver may be greatly
enlarged, firm, and nontender. Associated with this finding
may be jaundice, which appears in the 2nd or 3rd weeks of
life, is seldom intense, and does not persist for many days.
Anemia, probably indicative of bone marrow infection and
hematopoietic suppression, may become severe. Lesions in
the gastrointestinal tract and pancreas can occur and produce distention and delay in the passage of meconium.
Clinical signs of central nervous system involvement seldom appear in the newborn infant, although one third to
FIGURE 38-3 The radiograph displays the characteristic “celery
stalking” and widening of the metaphases in long bones found in
untreated congenital syphilis. (From American Academy of Pediatrics
Committee on Infectious Diseases: Syphilis: clinical manifestations images.
Red Book Online Visual Library, 2009, American Academy of Pediatrics.
Available at http://aapredbook.aappublications.org/cgi/content/figsonly/
2009/1/3.129.)
half of those infected suffer such involvement. One study
by Beeram et al (1996) found no difference in leukocyte
values or protein between the CSF of symptom-free infants
born to syphilis-seropositive mothers (positive rapid plasmin reagin [RPR] test result confirmed by positive fluorescent treponemal antibody-absorbent [FTA-ABS]) and
symptom-free control infants for whom sepsis for associated sepsis risk factors had been ruled out. In addition,
only 2 of the 329 infants born to syphilis-infected mothers with no or inadequate treatment had a positive venereal disease research laboratory ( VDRL) test result on their
CSF specimen. Another study of neonatal CNS invasion by
T. pallidum found the sensitivity and specificity of CSF
VDRL to be 71% and 92%, respectively (Sanchez et al,
1993). Overall, the added value of CSF evaluation in
asymptomatic infants appears marginal, because few cases
of congenital syphilis are diagnosed based on CSF abnormalities in the absence of other laboratory or clinical findings. However, the CDC and the European International
Union against Sexually Transmitted Infections guidelines
still recommend that infants with confirmed disease (e.g.,
those with increasing quantitative titers) or infants who
have a normal physical examination result but with an
inadequately treated, syphilis-infected mother have a CSF
evaluation for VDRL, cell count, and protein (Centers for
Disease Control and Prevention, 2006; French et al, 2009).
Further research may inform future guideline development.
DIAGNOSIS
The suggestion of congenital syphilis infection should
be spurred by a positive maternal antibody test (RPR or
VDRL). If the mother’s treponemal test (treponema pallidum particle agglutination assay [TP-PA] or FTA-ABS)
is also positive, a RPR or VDRL test should be performed
on the infant’s serum per current Red Book recommendations by the American Academy of Pediatrics (2009). Cord
blood is not a reliable testing source for neonatal infection
as false-positive RPR rates of 10% and false-negative RPR
rates of 5% have been reported (Rawstron and Bromberg,
1991). However, a serum-positive antibody test alone does
not confer a diagnosis of congenital syphilis, because of
transplacental transfer of maternal nontreponemal and
treponemal IgG antibodies. Because IgG is transferred
across the placenta, its finding in the baby’s serum simply means that the mother either currently has or has had
syphilis. She may have been successfully treated during
pregnancy and yet still has antibodies in her blood, or she
may not have received treatment at all and still has not
passed the disease on to her fetus. Conversely, if an antibody test is done on the infant’s serum because of clinical
suspicion in the absence of a positive maternal antibody
test, the infant’s antibody test can also be falsely negative
if the mother acquired infection late in pregnancy, because
she may not have had time to form antibodies herself or
has not passed them to her fetus.
IgM does not cross the placenta, so the presence of
specific IgM antibodies in the infant is generally diagnostic. Whereas the IgM immunoblot appears to be the
best available method to detect T. pallidum-specific IgM in
neonates, the CDC (2006) does not identify this as a commercially available and recommended test (Herremans
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
et al, 2010). Treponemal tests on infant serum are not
necessary. Comparison of maternal and infant nontreponemal serologic titers at delivery and followed over time
are most informative for infant diagnosis. These titers
should be accessed via the same test and in the same laboratory to ensure valid comparisons. If the infant titer is
fourfold higher than the mother’s titer, it is considered
highly suggestive of congenital infection. Additional criteria for treatment in the neonate include inadequate maternal treatment in a mother with syphilis and the presence of
clinical, laboratory, or radiographic evidence in the infant.
521
Evaluation of an infant with a concerning physical
examination result and maternal profile should include
darkfield or fluorescent antibody test of skin lesions or
mucous discharge. In addition, a complete blood cell count
with differential should be obtained, and CSF studies for
cell count, protein and VDRL are recommended. If clinically indicated, long-bone radiographs can be diagnostic.
Clearly the evaluation for congenital syphilis is complex, and serial investigations may be necessary for definitive diagnosis. An algorithm for evaluation is shown in
Figure 38-4.
Reactive maternal RPR/VDRL
Nonreactive maternal
TP-PA or FTA-ABS
False-positive
reaction: no further
evaluation
Reactive maternal TP-PA
or FTA-ABSa
Infant RPR/VDRL
nonreactive;
Treatment (1)
Infant RPR/VDRL less than
4 times maternal
RPR/VDRL
Infant physical
examination normal
a
b
c
d
Maternal
treatment
before
prenancyb
Maternal penicillin
treatment during
pregnancy and
more than 4 weeks
before delivery
No evaluation
or treatment
No evaluation;
Treatment (2)
Infant RPR/VDRL
reactive
Infant RPR/VDRL at least
4 times maternal
RPR/VDRL
Infant physical
examination abnormal
Maternal treatment:
none; or undocumented;
or 4 wk or less before
delivery; or nonpenicillin
drug; or maternal
evidence of reinfection
or relapse (fourfold or
greater increase in titers)
Evaluatec
Treatment (3)
Evaluated
Normal
evaluation
Abnormal, not done, or
incomplete evaluation
Treatment
(2)
Treatment (3)
RPR indicates rapid plasma reagin (test); VDRL, Venereal Disease Research Laboratory (test); TP-PA, Treponema pallidum particle
agglutination (test); FTA-ABS, fluorescent treponema antibody absorption (test).
Test for human immunodeficiency virus (HIV) antibody. Infants of HIV-infected mothers do not require different evaluation or
treatment.
Women who maintain a VDRL titer 1:2 or less (RPR 1:4 or less) beyond 1 year after successful treatment are considered serofast.
Evaluation consists of complete blood cell (CBC) and platelet count; cerebrospinal fluid (CSF) examination for cell count, protein,
and quantitative VDRL. Other tests as clinically indicated: long-bone and chest radiographs, neuroimaging, auditory brainstem
response, eye examination, liver function tests.
CBC, platelet count; CSF examination for cell count, protein, and quantitative VDRL; long-bone radiography.
TREATMENT:
(1) If the mother has had no treatment, undocumented treatment, treatment 4 weeks or less before delivery or evidence of
reinfection or relapse (fourfold or greater increase in titers) AND the infant’s physical examination is normal, THE treat infant
with a single intramuscular (IM) injection of benzathine penicilin (50 000 U/kg). If these criteria are not met, no treatment is
required. In both scenarios, no additional evaluation is needed.
(2) Benzathine penicillin G, 50 000 U/kg, IM, 1 dose.
(3) Aqueous penicillin G, 50 000 U/kg, IV, every 12 hours (1 week of age or younger), every 8 hours (older than 1 week), or procaine
penicillin G, 50 000 U/kg, IM, single daily dose, 10 days.
FIGURE 38-4 Algorithm for evaluation and treatment of infants born to mothers with reactive serologic tests for syphilis. (From American Academy
of Pediatrics Committee on Infectious Diseases: Syphilis, Red Book Online, 2009, American Academy of Pediatrics. Available at http://aapredbook.aappublication
s.org/cgi/content/full/2009/1/3.129. Accessed November 19, 2010.)
522
PART IX Immunology and Infections
TABLE 38-4 Recommended Treatment of Pregnant Patients With Syphilis
Stages of Syphilis
Drug (Penicillin)
Route
Dose (Units)
Early (<1 yr duration)
Primary, secondary, or early latent
HIV antibody-negative
Recommended: benzathine
IM
2.4 million single dose; possibly repeat in 1 wk
HIV antibody-positive*
Recommended: benzathine
IM
2.4 million single dose; possibly repeated weekly for 3 wk
IM
2.4 million weekly for 3 wk
IV
3-4 million every 4 hr for 10-14 days
IM
2.4 million daily for 10-14 days
Alternative: penicillin desensitization
Latent (>1 yr duration)†
Recommended: benzathine
Alternative: penicillin
desensitization
Neurosyphilis
Recommended: aqueous
Alternative:
procaine‡
From Remington JS, Klein JO, Wilson CB, et al, editors: Infectious diseases of the fetus and newborn infant, ed 6, Philadelphia, 2006, WB Saunders, p 567.
IM, Intramuscular; IV, intravenous.
*With normal cerebrospinal fluid findings, if performed.
†Lumbar puncture to exclude neurosyphilis is recommended for HIV-antibody–positive patients.
‡Probenecid, 500 mg orally, four times per day for 10 to 14 days, should also be prescribed.
TREATMENT AND LONG-TERM OUTCOMES
Treatment of all pregnant women infected with syphilis
is recommended. Guidelines for maternal treatment are
found in Table 38-4 and are tailored to the stage of disease
(e.g., primary, secondary, latent). Treatment of an infected
infant requires 10 days of intravenous or intramuscular
penicillin. Specific dosing and treatment regimens are outlined in Table 38-5, followed by long-term follow-up recommendations in Table 38-6. Notably, if a single dose is
missed during the treatment of an infected infant, restarting the entire series is recommended.
Late manifestations of infection in untreated infants,
even if initially asymptomatic, can occur years after birth.
Pathognomonic presentations include the Hutchinson
triad (interstitial keratitis, eight cranial nerve deafness and
Hutchinson’s [peg-shaped] teeth), mulberry molars (first
lower molar with many small cusps), and Clutton’s joints
(synovitis with hydrarthrosis, tenderness, and limited
range of motion) (Fiumara et al, 1970). Mental retardation
can also be a feature of late, untreated congenital syphilis
(Brosco et al, 2006).
CONGENITAL MALARIA
Malaria is a parasitic disease of epidemic proportion. An
estimated 200 to 500 million cases occur worldwide each
year, resulting in 2 to 3 million deaths. The greatest burden
of disease occurs in Sub-Saharan Africa, although malaria
is increasingly recognized as a significant public health
problem in Asia and Oceania. In areas of high transmission,
mortality is concentrated largely among young children
and pregnant women. Malaria is caused by four Plasmodium
species: P. falciparum, P. vivax, P. ovale, and P. malariae.
Of these, P. falciparum is the major cause of morbidity and
mortality. Humans typically acquire infection through the
bite of the Anopheles spp. mosquito. Transmission may also
occur through blood transfusion or vertically from the
mother to fetus, resulting in congenital malaria.
Despite the prevalence of malaria in the developing
world, it has historically been believed that congenital
malaria occurs infrequently in areas of high transmission
(Bruce-Chwatt, 1952; Covell, 1950; McGregor, 1984). As
of 1995, only 300 cases of congenital malaria had been
reported in the literature (Balatbat et al, 1995), largely
from outside of malarious areas. The rarity of vertical
transmission has been attributed to the effectiveness of
the placenta as a barrier against passage of maternal parasitized red blood cells as well as the protective effect of
maternally derived antibodies in the fetus and newborn.
Recent reports, however, suggest that congenital malaria
is no longer a rare occurrence in endemic areas and has
likely been underrecognized and underreported (Akindele
et al, 1993; Falade et al, 2007; Fischer, 1997, Ibhanesebhor, 1995; Menendez and Mayor, 2007; Uneke, 2007a,
2007b).
Although endemic malaria has been eliminated from
the United States, approximately 1200 cases of malaria
are reported to the CDC annually, almost exclusively in
travelers and immigrants from endemic countries. From
1966 to 2005, 81 cases of congenital malaria were reported
to the National Malaria Surveillance System of the CDC
(Lesko et al, 2007). The majority of infants were born to
foreign-born women. As an increasing number of people
travel to and emigrate from malarious areas, the number
of cases of malaria (and consequently congenital malaria)
will continue to increase. The lack of familiarity with this
disease in the United States renders it a diagnostic and
therapeutic challenge for clinicians, with delays in diagnosis potentially leading to significant morbidity and
mortality (Griffith et al, 2007). It is therefore critical to
maintain a high index of suspicion for congenital malaria
in the evaluation of infants born to women from endemic
countries.
DEFINITION
Congenital malaria is defined as malaria acquired by the
fetus or newborn from the mother, either in utero or at
parturition, but there exists no consensus on the application of this definition. Most commonly, congenital malaria
is defined as the presence of Plasmodium spp. parasites
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
TABLE 38-5 Recommended Treatment of the Newborn With
Syphilis
Penicillin
Dosage
Aqueous penicillin G
50,000 units/kg every 12 hours during first
7 days of life and every 8 hours thereafter for a total of 10 days
or
Penicillin G, procaine
50,000 units/kg/day, intramuscular, in a
single dose every day for 10 days
From Centers for Disease Controls and Prevention: Sexually transmitted diseases treatment
guidelines, 2006. Available at www.cdc.gov/std/treatment/2006/congenital-syphilis.htm.
Accessed November 17, 2010.
TABLE 38-6 Follow-up after Treatment or Prophylaxis for
Congenital Syphilis
Patient Category
Follow-up Procedures
Infants with diagnosis of congenital
syphilis
RPR testing every 2-3 mo until negative or
decreased fourfold; if RPR titer is stable
or increasing after 6-12 mo after treatment, reevaluate and treat again; perform
treponemal antibody test after age of 15
mo; if initial CSF analysis is abnormal or
infant shows signs of CNS disease, repeat
CSF evaluation every 6 mo until normal;
with abnormal CSF not due to intercurrent illness on retesting, treat again;
careful developmental evaluation, vision
testing, and hearing testing are indicated
Infants who received
treatment in utero
or at birth because
of maternal syphilis
RPR testing at birth and then every 3
mo until result is negative; treponemal
antibody test after age of 15 mo
Women who
received treatment
for syphilis during
pregnancy
RPR testing as often monthly until delivery, then every 6 mo until negative result
obtained or titer decreased fourfold;
repeated treatment any time there is a
fourfold rise in RPR titer
From Remington JS, Klein JC, Wilson CB, et al, editors: Infectious diseases of the fetus
and newborn infant, ed 6, Philadelphia, 2006, Saunders, 2006, p 572.
CSF, Cerebrospinal fluid; RPR, rapid plasmin reagin.
in the peripheral blood in the first 7 days of life (Covell,
1950; Menendez and Mayor, 2007; Moran and Couper,
1999; Sotimehin et al, 2008; Uneke, 2007a). This definition is applicable in areas of high malaria transmission
where, among older infants, it would be difficult to distinguish congenitally acquired from mosquito-acquired
disease. Outside endemic areas, where postnatal transmission can be reasonably excluded, clinical onset of disease
often does not occur until after the 1st week of life, and
age-specific criteria are not useful for the diagnosis of
congenital malaria. It is likely that because of the delay
in clinical presentation, many cases of congenital malaria
in endemic areas are misclassified as being acquired from
mosquitoes.
Alternate applications of the definition of congenital
malaria include the detection of parasites in peripheral
blood or in umbilical cord blood (Fischer, 1997) within the
first 24 hours of life (Larkin and Thuma, 1991). It has been
argued that congenital malaria should be distinguished
523
from cord blood parasitemia, which is not uncommon in
some endemic areas and frequently clears without involving the peripheral circulation. Reinhardt (1978) found parasites in thick smears of umbilical cord blood in 22% of 19
infants born to women in the Ivory Coast, but the peripheral blood smears were negative for all of the infants. Similarly, 4% of 1009 infants born to Tanzanian women had
parasites in cord blood, but parasitemia was detected on
peripheral smear in only 2 of 11 infants (McGregor, 1984).
Parasitemia detected shortly after birth may also resolve
without evolving into clinically symptomatic disease. In a
recent multicenter study, the correlation between maternal, placental, umbilical cord, and peripheral parasitemia
was evaluated in 1875 mother-baby pairs in Nigeria. Thick
and thin blood smears were obtained within 4 hours of
birth, and smear-positive (cord or peripheral) neonates
were retested on days 2, 3, and 7 of life, with treatment
for those with symptoms or persistent parasitemia. The
overall prevalence of congenital malaria was 5.1%, with
parasitemia detected in 19% of infants born to mothers
with peripheral parasitemia, 21% of those born to mothers
with placental malaria, and 45% of those with positive cord
smears. Spontaneous clearance of parasitemia occurred in
62% of infants before day 2, whereas 33% were symptomatic within 3 days of birth (Falade et al, 2007). It has now
been well established that (1) maternal and placental parasitemia are important risk factors for congenital malaria,
(2) a correlation between umbilical cord parasitemia and
neonatal parasitemia exists, and (3) congenital malaria can
occur in the absence of symptoms (Mukhtar et al, 2006;
Sotimehin et al, 2008; Uneke, 2007a). However, the definition of congenital malaria has yet to be standardized to
facilitate a better understanding of the epidemiology of
this disease and the outcomes of prevention measures.
EPIDEMIOLOGY
Congenital malaria has traditionally been considered a
rare consequence of malaria in pregnant women living
in endemic areas. Covell (1950) published a review of
cases of congenital malaria reported in areas with rates
of placental parasitemia ranging from 5% to 74%, and
rates of maternal malaria ranging from 1% o 68%. The
prevalence of congenital malaria—defined as parasitemia
detected in the first 7 days of life—was estimated to be
0.3% (16 of 5324 births) among immune mothers. Subsequent reports supported the observed low frequency
of congenital malaria—defined as umbilical cord parasitemia or parasitemia in the first 24 hours of life—among
indigenous populations (Bruce-Chwatt, 1952; Cannon,
1958; MacGregor, 1984; Williams and McFarlane, 1970).
These observations have been cited repeatedly in the literature to support the notion that congenital malaria is
an uncommon occurrence in endemic areas despite the
high prevalence of maternal and placental malaria. More
recent reports, however, suggest that the prevalence of
congenital malaria was underestimated, with rates from
endemic and nonendemic areas ranging from 8% to
33% (Desai et al, 2007; Menendez and Mayor, 2007). In
Zambia during a season of heavy malaria transmission,
incidence rates for congenital malaria ranged from 4%
to 15% (Nyirjesy et al, 1993). A survey of seven sites in
524
PART IX Immunology and Infections
Sub-Saharan Africa indicated a prevalence of congenital
malaria—defined as umbilical cord blood parasitemia—
ranging from 0% to 23% (Fischer, 1995). Congenital
malaria, defined as neonatal parasitemia, was detected in
15.3% and 17.4% of neonates born in two sites in Nigeria (Mukhtar et al, 2006; Runsewe-Abiodun et al, 2006).
The apparent increase in the frequency of congenital
malaria has been attributed to increasing resistance of
P. falciparum to antimalarial drugs resulting in increased
maternal parasitemia, increased virulence of the parasite,
and reduced transmission of antibody from mother to
newborn because of malaria chemoprophylaxis administered to pregnant women. It is equally likely that congenital malaria was previously underreported because of
the difficulty in differentiating congenital malaria from
postnatally acquired malaria in neonates, or it was underdetected because of low parasite densities in newborns.
It is evident that congenital malaria is not an uncommon
occurrence in areas of high transmission. The variability
in prevalence of disease may be attributed to differences
in the definition of congenital malaria or the methods
used for parasite detection. The inconsistency may also
represent true environmental differences with differences
in levels of maternal immunity.
In contrast to the rarity of congenital malaria in indigenous populations, Covell (1950) found the prevalence
of congenital malaria among nonimmune populations
(i.e., Europeans residing in or visiting endemic areas) to
be approximately 7%. It was postulated that congenital
malaria is more common in infants born to these women
because of lower levels of malaria-specific maternal antibodies being transmitted to the fetus. Most published
reports of congenital malaria describe infants born in
nonendemic countries whose mothers emigrated from
malarious areas to areas free of malaria. These women presumably developed a recrudescence of disease because of
waning immunity from lack of continued exposure
In the United States, the occurrence of congenital
malaria is well documented because the country has been
free of indigenous disease since the 1950s. From 1950
to 1991, 49 cases of congenital malaria were reported in
the literature (Hulbert, 1992), and additional cases were
reported during the next 15 years (Balatbat et al, 1995;
Baspinar et al, 2006; D’Avanzo et al, 2002; Gereige and
Cimino, 1995; Starr and Wheeler, 1998; Viraraghavan and
Jantausch, 2000). In an updated review, Lesko et al (2007)
tabulated 81 cases reported to the CDC between 1966 and
2005. Almost all the cases were among infants whose mothers were foreign born, suggesting that congenital malaria
is primarily a health problem of recent immigrants rather
than of U.S.-born travelers to malaria-endemic countries.
Forty-four women (54%) had emigrated from Asia, 27
(33%) from South or Central America, and 7 (9%) from
Africa. Until 1979, one to two cases were reported annually (Malviya and Shurin, 1984). An abrupt rise to 16 cases
around 1981 (Figure 38-5) correlated with an increase in
the total number cases of malaria that occurred as a result
of a large influx of refugees and immigrants from Southeast Asia, with 15 of the 16 infants being born to mothers
from that region (Quinn et al, 1982).
Congenital malaria occurs with all Plasmodium species.
Among the 107 cases of congenital malaria reported by
Covell (1950), mostly from Africa, 64% were infected with
P. falciparum, 32% by P. vivax, and 2% by P. malariae.
Although P. falciparum remains the predominant pathogen
in Sub-Saharan Africa, P. vivax may account for a larger
proportion of cases in Asia. Among 27 cases reported in
Thailand between 1981 and 2005, 82% of were cause by
P. vivax (Wiwanitkit, 2006). P. malariae is less frequently a
causative agent, with fewer than 10 cases reported worldwide since 1950 (de Pontual et al, 2006). Concurrent infection with P. malariae and P. vivax has been documented
(MacLeod et al, 1982). In the United States, the predominant Plasmodium species causing congenital malaria
12
Number of cases
10
8
6
4
2
19
66
19
68
19
70
19
72
19
74
19
76
19
78
19
80
19
82
19
84
19
86
19
88
19
90
19
92
19
94
19
96
19
98
20
00
20
02
20
04
0
Year
FIGURE 38-5 Number of cases of congenital malaria reported to the National Malaria Surveillance System per year, 1966-2005. (From Lesko CR,
Arguin PM, Newman RD: Congenital malaria in the United States: a review of cases from 1966 to 2005, Arch Pediatr Adolesc Med 161:1062-1067, 2007.)
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
reflects the countries of origin of the mothers, as noted
previously. In Hulbert’s (1992) review of 49 cases, 82% of
infections were caused by P. vivax. In the updated review
by Lesko et al (2007), the predominant infecting species
remained P. vivax (81%), although all four species were
represented.
PREGNANCY AND MALARIA
Malaria in pregnancy is an immense public health problem
in the developing world, with substantial effects on maternal, fetal, and infant health. It is well established that both
the frequency of disease and density of parasitemia are
higher in pregnant women compared with nonpregnant
women (Coll et al, 2008; Desai et al, 2007; Rogerson et al,
2007). In Sub-Saharan Africa, 25 million pregnant women
are at risk for P. falciparum infection every year (Desai
et al, 2007). Among 20 studies conducted between 1985
and 2000, the median prevalence of maternal malaria
infection (defined as peripheral or placental infection) in
all was 28% (Steketee et al, 2001). Thus, one in four pregnant women in areas of stable transmission in Africa have
evidence of malaria infection at the time of delivery. The
increased susceptibility likely represents a combination of
immunologic and hormonal changes combined with the
unique ability of infected erythrocytes to sequester in the
placenta (Rogerson et al, 2007). The risk is indisputably
greater for primigravidae, with primigravidae having a
twofold to fourfold increased risk of placental malaria compared with multigravidae (Desai et al, 2007; McGregor,
1984; Uneke, 2008).
The clinical features of P. falciparum malaria in a pregnant woman depend to a large degree on her immune status, which in turn is determined by her prior exposure to
malaria. In pregnant women with little or no preexisting
immunity, such as women from nonendemic countries or
travelers to malarious areas, infection is associated with
high risks of severe disease with significant maternal and
perinatal mortality. In contrast, women residing in areas
of stable malaria transmission usually have a high level of
immunity to malaria. Infection may be frequently asymptomatic and therefore unsuspected or undetected, but it
is associated with placental parasitization, with consequent effects on maternal and fetal outcomes. The most
significant consequence of pregnancy-associated malaria
is maternal anemia. It is estimated that in Sub-Saharan
Africa between 200,000 and 500,000 pregnant women
develop anemia as a result of malaria, and that up to 10,000
maternal anemia-related deaths are a consequence of
P. falciparum parasitemia (Uneke, 2008). Malaria in pregnancy also has potentially devastating effects on the fetus
and newborn, including spontaneous abortion, still birth,
premature delivery, congenital infection, and neonatal
death (Coll et al, 2008; Fischer, 2003). The most notable
consequence is low birthweight (LBW), likely because of
fetal growth restriction. In areas of high transmission in
Africa, the risk of LBW approximately doubles if women
have placental malaria, with the greatest effect in primigravidae (Desai et al, 2007). Pregnancy-associated malaria
is believed to be responsible for 30% to 35% of LBW
infants and for 75,000 to 200,000 infant deaths each year
(Coll et al, 2008).
525
TRANSMISSION
The timing and mechanism of transmission of Plasmodium spp. parasites from the mother to the fetus is not
well understood. Postulated mechanisms include maternal transfusion into fetal circulation either during pregnancy or at delivery, or direct penetration of parasitized
red blood cells through the chorionic villi or through
premature separation of the placenta. In utero transmission is supported by the finding of malarial parasites in
fetal tissues at autopsy (Mertz et al, 1981), by umbilical
cord blood parasitemia (McGregor, 1984) and the onset
of clinical signs of malaria within hours of birth (Brandenburg and Kenny, 1982; Covell, 1950; Gereige and
Cimino, 1995). Antenatal transmission is also suggested
by findings from Malawi, where approximately 50% of
newborns with cord blood parasitemia were infected with
parasites of a different genotype than their mothers at the
time of delivery (Fischer, 2003). Alternatively, clinical
findings in infants with congenital malaria may be delayed
for several weeks after birth, suggesting infection at parturition. It is generally understood that the placenta acts
as an effective barrier to prevent the transfer of malaria
parasites from maternal into fetal circulation, supporting
transmission at parturition as the most likely mechanism.
Vertical transmission of malaria probably does not occur
as a result of transplacental passage of exoerythrocytic
parasites. More likely, transmission occurs by transfusion
of parasitized maternal erythrocytes through a breach
in the placental barrier that may occur either prematurely during pregnancy or during labor. Transmission
of malaria by breastfeeding is not known to occur. The
fate of the Plasmodium spp. parasite is unclear after it is
transmitted to the fetus. As discussed previously, parasites
detected in umbilical cord blood or shortly after birth may
be cleared spontaneously, resulting in no disease manifestation. Alternatively, parasitemia may be maintained and
proliferate until multiplication permits the development
of clinical disease.
CLINICAL PRESENTATION
The clinical picture of overt congenital malaria is detailed
in cases reported outside of endemic areas (Harvey et al,
1969; Hindi and Azimi, 1980; Hulbert, 1992; Lesko et al,
2007). The manifestation of disease, although occasionally noted within hours of birth (Brandenburg and Kenny,
1982; Gereige and Cimino, 1995), is typically delayed until
the infant is several weeks old. In the classic review of 49
infants with congenital malaria reported in the United
States between 1950 and1992, the mean age at onset of
symptoms was 5.5 weeks, with 96% of infants presenting
between 2 and 8 weeks of age (Hulbert, 1992). Among
cases reported to the CDC from 1966 to 2005 (Lesko et al,
2007), the median age of symptom onset for 81 infants was
21.5 days for all species combined (Figure 38-6). Infants
infected with P. malariae were significantly older at symptom onset (mean, 53 days) compared with those infected
with P. vivax or P. falciparum.
The prolonged interval between birth and onset of clinical manifestations may be explained by transmission late in
pregnancy or at delivery, such that multiple erythrocytic
526
PART IX Immunology and Infections
18
16
14
Number of cases
12
10
8
6
4
2
0
<1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21
Age at symptom onset, wk
FIGURE 38-6 Age in weeks at symptom onset of infants with reported congenital malaria, United States, 1966-2005. (From Lesko CR, Arguin PM,
Newman RD: Congenital malaria in the United States: a review of cases from 1966 to 2005, Arch Pediatr Adolesc Med 161:1062-1067, 2007.)
life cycles are required to produce clinically evident disease. Alternatively, the delay may be attributed to the presence of transplacentally acquired maternal antimalarial
antibodies. When such antibodies are present in sufficient
concentrations, as in infants born to immune mothers,
parasitic replication can be prevented or attenuated, and
clinical signs can be mild, delayed, or even absent. The
presence of a high concentration of fetal hemoglobin in
newborns may also promote resistance to multiplication
of parasites. Among infants born to mothers with low or
nonexistent immunity, parasitic replication is more likely
uninhibited, and clinical signs of malaria may supervene.
Preterm infants, who do not benefit from passive immunity, can manifest clinical signs earlier than full-term
infants. In a review of premature neonates with congenital
malaria, 4 of 5 infants received a diagnosis in the 1st week
of life (Ahmed et al, 1998), although the prompt medical
evaluation afforded these infants may have facilitated earlier detection.
The clinical features of congenital malaria are nonspecific and often resemble those of bacterial or viral sepsis
and other congenital infections. Fever is almost uniformly
present, although without the classic paroxysmal pattern
described for malaria beyond the neonatal period. Hulbert (1992) noted fever in all 44 infants for whom clinical information was available. In the cases reported from
1966 to 2005, fever was reported in 70 of 81 cases (86%)
(Lesko et al, 2007). Hepatomegaly and splenomegaly suggestive of a transplacentally acquired infection are found
in a substantial portion of infants (Table 38-7). Anemia
(often hemolytic), thrombocytopenia, and hyperbilirubinemia are the most commonly reported laboratory findings. Additional signs, symptoms, and laboratory findings
are listed in Table 38-7.
TABLE 38-7 Frequency of Symptoms, Signs, and Laboratory
Findings Among 81 Infants With a Diagnosis of Congenital Malaria
(United States, 1966-2005)
Symptoms, Signs, and
Laboratory Findings
Fever
Anemia
Splenomegaly
Hepatomegaly
Thrombocytopenia
Jaundice
Irritability
Anorexia
Vomiting
Cough
Diarrhea
Lethargy
Hemolysis
Pallor
Hyperbilirubinemia
Failure to thrive
Seizures
Dyspnea
Purpura
Tachycardia
Monocytosis
Infants, No. (%)*
70 (86)
28 (36)
25 (31)
16 (20)
12 (15)
11 (14)
8 (10)
8 (10)
8 (10)
6 (7)
3 (4)
3 (4)
3 (4)
3 (4)
2 (3)
2 (3)
2 (3)
1 (1)
1 (1)
1 (1)
1 (1)
From Lesko CR, Arguin PM, Newman RD: Congenital malaria in the United States: a
review of cases from 1966 to 2005, Arch Pediatr Adolesc Med 161:1062-1067, 2007.
*Percentages do not total 100% because each case can have more than one symptom,
sign, or laboratory finding.
In endemic areas, the traditional belief has been that
congenital malaria is rare and that when it occurs the
infant is typically asymptomatic and develops no clinical features. The lack of symptoms has been attributed to
transplacentally acquired antibodies from the mother as
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
well as the protective effects of high levels of fetal hemoglobin. Depending on the region, spontaneous clearance
of peripheral parasitemia has been documented in 87% to
100% of neonates (Lesko et al, 2007; Mukhtar et al, 2006).
Larkin and Thuma (1991) found peripheral parasitemia
within 24 hours of age in 19 of 51 newborns (65%), but
only 7 had clinical signs of disease. Because all 19 newborns received antimalarial therapy, it is unknown how
many would have manifested disease if untreated. More
recently, Falade et al (2007) noted spontaneous clearance
of parasitemia in 62% of 95 neonates before day 2 of life.
Of the remaining infants, 34% were symptomatic within
3 days of birth, with fever and refusal to eat being the
most common signs of disease. When active surveillance
for malaria was conducted in newborns being evaluated
for possible bacterial sepsis in Nigeria, 16 of 203 (8%)
neonates had parasitemia, and 10 (5%) met the definition
of congenital malaria (Ibhanesebhor, 1995). Predominant
features of disease included fever, respiratory distress,
anemia, and hepatomegaly. In another area in Nigeria, of
202 neonates less younger than 1 week who were admitted for evaluation of sepsis, 71 (35%) were diagnosed
with congenital malaria (Ekanem et al, 2008). Fever was
the most common symptom and was present in 93% of
infants. Refusal to feed and jaundice were reported in
approximately 33%. These observations suggest that, as
in infants diagnosed outside endemic areas, the clinical
presentation of congenital malaria in endemic areas does
not differ significantly from bacterial sepsis. Because the
clinical symptoms of congenital malaria may be indistinguishable from that of neonatal sepsis, it is suggested that
screening for malaria be included as part of routine investigation of newborns with fever in areas of high malaria
transmission (Ekanem et al, 2008; Runsewe-Abiodun
et al, 2006).
DIAGNOSIS
Diagnostic tests for malaria include blood smears, rapid
antigen detection tests (RDTs) and PCR. Definitive diagnosis of congenital malaria is based on the microscopic
demonstration of parasites on stained thick and thin blood
films. Thick blood smears test for the presence of parasites
by concentration of red blood cells, whereas thin blood
smears allow species identification and quantification
of parasitemia. In cases of suggested congenital malaria,
specimens for smears should be obtained from both the
infant and the mother. If test results from the initial set
of smears are negative, additional sets should be obtained
every 12 to 24 hours; three sets are generally considered
sufficient for diagnostic evaluation. Response to therapy
may also be measured by clearance of parasitemia on blood
films.
RDTs are based on the immunochromatographic detection of parasite-specific antigens circulating in the bloodstream. Many RDTs are commercially available outside the
United States, and, depending on the antigens targeted,
the tests may detect only P. falciparum or all Plasmodium
species. RDTs are simple to use, do not require specialized training or facilities, and offer a useful alternative to
microscopy in situations where reliable microscopic diagnostics are not readily available. However, the tests have
527
demonstrated mixed results in multiple trials, and sensitivity remains a problem, especially at low parasite densities. Information regarding the sensitivity of these tests is
limited for neonatal or congenital malaria. In the evaluation of an RDT for the diagnosis of congenital malaria
in Nigeria, parasitemia was detected by microscopy in 21
of 192 newborns (10.9%) at 0 to 3 days of age, whereas
the RDT result (OptiMAL [Flow Inc, Portland, Oregon])
was negative in all infants, including those diagnosed by
microscopy. Whether the poor performance of the test
was due to low-level parasitemia or suboptimal state of the
parasites, the RDT was determined not to be useful for the
diagnosis of congenital malaria (Sotimehin et al, 2007). It
is recommended that microscopy be conducted in parallel with RDTs because low level parasitemia may not be
detected or, if parasitemia is detected, for species identification and determination of parasite density.
The major advantage of PCR for the diagnosis of malaria
is its ability to detect low level parasitemia and identify
parasites to a species level. Currently PCR is used mainly
to confirm positive blood smears, particularly when the
results of the smear are not definitive or there is a mixed
species infection. PCR may detect DNA from circulating
nonviable parasites after treatment, resulting in difficulty
differentiating an active infection from a recently cleared
infection. Although PCR is a highly sensitive alternative
to microscopy, the infrastructure and expertise required
preclude its use in malaria-endemic areas and many health
care settings in the United States.
As with malaria in general, the diagnosis of congenital
malaria outside of endemic areas is often delayed because
of nonspecific features and lack of clinical suspicion.
Among the 81 cases reviewed by Lesko et al (2007), a
median length of delay of 8.5 days was noted for 15% of
the infants. Occasionally the diagnosis is made incidentally.
In all four cases of congenital malaria reported by Quinn
et al (1982), Plasmodium spp. parasites were noted by hematology technicians on routine smears performed for blood
cell counts. Maternal history of recent travel to or emigration from an endemic area may suggest the diagnosis, but
is often obscured by the lack of clinical or laboratory findings in the mother. Lesko et al (2007) found that, of the
mothers for whom a history was available, 67% reported
having fever during pregnancy, and 26% reported a diagnosis of malaria during pregnancy. Maternal blood films
were performed after either symptomatic illness or malaria
diagnosis in the infant. Overall only 42% of women had
parasitemia detected, although it is not clear whether
an adequate number of smears were conducted for each
patient. As a result, lack of peripheral parasitemia in the
mother of an infant with suspected congenital malaria does
not exclude the diagnosis.
Further confounding the early recognition of disease
in the infant is the potentially prolonged lapse between
malaria exposure in the mother and transmission of infection to the infant. P. vivax and P. ovale may remain dormant in the liver, especially if the infected individual did
not receive therapy for the exoerythrocytic stage, which
can cause a delayed relapse of malaria in travelers or immigrants. P. malariae can persist for 20 to 40 years before
clinical symptoms or demonstrable parasitemia appear
(D’Avanzo et al, 2002). Congenital malaria has been
528
PART IX Immunology and Infections
reported in an infant whose mother lived in the United
States for 5 years before delivery and had no signs or
symptoms or malaria for more than 20 years (Harvey et al,
1969). In North Carolina, congenital P. malariae infection
was reported in a 10-week-old infant who was born to a
mother who had emigrated from the Democratic Republic
of Congo 4 years before delivery (D’Avanzo et al, 2002). In
a review by Lesko et al (2007), the median duration from
the mother’s last exposure to delivery was 9.5 months. The
time elapsed since exposure was longest for those with
P. malariae infection, ranging from 2 to 12 years.
Recognizing that congenital malaria is an exceptional
occurrence in the United States, it is still important to
include malaria in the differential diagnosis of fever
in infants born to mothers who have been exposed to
malaria, even if the exposure is remote and even if the
woman is asymptomatic. Of 11 infants with congenital malaria in the United States born to women known
to have parasitemia at or shortly after delivery, only
five underwent testing by blood smears, and all five had
negative test results at the time of delivery (Lesko et al,
2007). Data are insufficient to determine the overall risk
of an infant developing congenital malaria when born to
a woman at risk for parasitemia or identified with parasitemia at birth. Consequently, the evaluation of infants
born outside endemic areas to women with epidemiologic
risk factors for parasitemia should be individualized. In
malaria endemic areas, and as a public health measure,
it has been recommended that blood smears should be
checked as part of the evaluation of neonates with fever
born to mothers who have had fever within a few weeks of
delivery (Uneke, 2007a).
TREATMENT
The management of malaria consists of supportive care
and antimalarial therapy. Information regarding treatment of congenital malaria is limited, and recommended
chemotherapy is similar to that of noncongenital infections. The treatment regimen is based on the infecting
species, the possibility of drug resistance, and the severity
of disease. For mild infections caused by P. vivax, P. ovale,
and P. malariae or chloroquine-sensitive P. falciparum,
chloroquine orally (10 mg base/kg initially followed by
5 mg base/kg 6, 24, and 48 hours later) is recommended.
Treatment with primaquine is not necessary for congenitally acquired P. vivax or P. ovale infection because, like
transfusion-associated malaria, congenital infection does
not involve the exoerythrocytic phase.
The treatment of congenital malaria due to chloroquine-resistant P. falciparum is poorly defined. In older
children, three treatment options currently recommended
are: (1) oral quinine plus either tetracycline, doxycycline,
or clindamycin; (2) atovaquone-proguanil; or (3) mefloquine. For the treatment of congenital malaria, oral quinine sulfate and trimethoprim-sulfamethoxazole for 5
days was recommended by Quinn et al (1982), who used
the regimen to treat a 1-month-old infant. Ahmed et al
(1998) used a similar regimen for the treatment of an
infant born at 28 weeks’ gestation to a mother from Zaire.
Other regimens used successfully in neonates include oral
quinine sulfate and pyrimethamine-sulfadoxine (Gereige
and Cimino, 1995) and intravenous quinine hydrochloride
followed by oral quinine (Airede, 1991). Intravenous quinine is no longer available in the United States. Because of
the rarity of congenital malaria in the United States, the
changing pattern of resistance, and the potential toxicity
associated with drugs used for therapy, current treatment
recommendations should be sought from the Malaria
Branch of the CDC (www.cdc.gov/malaria). For health
care professionals, assistance with management of malaria
is also available 24 hours a day through the CDC Malaria
Hotline (770-448-7788).
Severe malaria occurs most commonly with P. falciparum
infection and is characterized by one or more of the following: (1) parasitemia greater than 5% of red blood cells, (2)
central nervous system or other end-organ involvement,
(3) shock, (4) acidosis, (5) severe anemia, or (6) hypoglycemia. Management of severe malaria involves parenteral
treatment in an intensive care setting. Until recently, the
only parenteral therapy available in the United States was
quinidine gluconate. Quinidine is more cardiotoxic than
quinine and should be administered with continuous cardiac monitoring. Exchange transfusion may be warranted
when parasitemia exceeds 10% or if there are complications at lower parasite densities.
The efficacy of treatment should be monitored by examining blood smears (i.e., malaria smears) every 12 hours
until negative for malaria parasites. Response to therapy
with chloroquine for non-P. falciparum malaria is usually
favorable (Brandenburg and Kenny, 1982; Dowell and
Musher, 1991; Hindi and Azimi, 1980). It has been suggested that infants born to mothers with parasitemia at
delivery should be treated presumptively for congenital
malaria (Lesko et al, 2007). Data are insufficient to determine the risk of an infant developing congenital malaria
when born to a mother with parasitemia. Although there is
evidence from endemic areas that parasitemia detected at
or shortly after delivery may clear spontaneously, the clinical relevance of this observation in nonendemic areas is
unclear. It is recommended that physicians judge each case
individually, considering factors such as access to medical care and reliability of follow up in deciding whether to
treat infants presumptively.
PROGNOSIS
Malaria during pregnancy is likely an underappreciated
risk factor for increased infant morbidity and mortality in
endemic areas. In a review of studies published between
1985 and 2000, Steketee et al (2001) determined population-attributable risks for maternal malaria of 3% to 8% for
infant mortality. It was estimated that 75,000 to 200,000
infant deaths annually are associated with malaria during
pregnancy, although what proportion of these are related
to congenital malaria is unknown. Outside endemic areas,
the short-term outcome of congenital malaria has been
favorable. Most infants respond rapidly to therapy with
clearance of parasitemia. There were no reports of death
or adverse outcomes in the 49 cases reported from 1950 to
1992 or in the 81 cases reported to the CDC from 1966 to
2005 (Hulbert, 1992; Lesko et al, 2007). It is unclear, however, whether the outcomes are due to an overall favorable
prognosis or reporting bias.
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
PREVENTION
The prevention of congenital malaria is based on a pregnant woman’s avoidance of exposure and use of chemoprophylaxis. Malaria infection in pregnant women is more
common and more severe than in nonpregnant women.
Malaria also increases the risk for adverse pregnancy outcomes, including prematurity, abortion, and still birth.
Nonimmune pregnant women are at the highest risk for
these adverse outcomes. Based on these observations, the
CDC advises women who are pregnant or likely to become
pregnant to avoid travel to areas with malaria transmission. If such travel is unavoidable, consultation with an
infectious disease or malaria expert is advised. The use of
mosquito netting, mesh screens on windows, insecticides,
and mosquito repellents can decrease potential exposure to malaria parasites. For pregnant women traveling
to areas where there is no chloroquine-resistant P. falciparum malaria, prophylaxis with chloroquine is advised.
The safety of chloroquine for the fetus when used at the
recommended doses for malaria prophylaxis is well established (MacLeod et al, 1982). For travel to areas where
chloroquine resistance has been reported, mefloquine is
the only medication that is currently recommended for
prophylaxis during pregnancy. Atovaquone-proguanil is
not recommended during pregnancy because of insufficient information on potential adverse effects. Doxycycline is contraindicated because of adverse effects on the
fetus caused by a related drug, tetracycline, which include
dysplasia and discoloration of teeth and inhibition of bone
growth. Health care professionals caring for women who
cannot take the recommended antimalarial agent should
contact the CDC Malaria Hotline (770-488-7788).
Pregnant women originally from areas where malaria is
endemic but who are now living in nonendemic areas may
be only partially immune. When traveling to their countries of origin, they should be considered nonimmune and
thus should receive the same recommendations as nonimmune women.
The burden of malaria among pregnant women in
endemic areas is well recognized, and prevention and control strategies for areas of high P. falciparum transmission
are aimed at reducing maternal and infant mortality. The
World Health Organization has proposed a three-pronged
approach: (1) intermittent preventative treatment with an
effective antimalarial agent at scheduled antenatal visits,
(2) insecticide-treated nets, and (3) effective case management of clinical infection. A metaanalysis of more recent
intervention trials suggests that successful prevention of
these infections reduces the risk of severe maternal anemia
by 38%, LBW by 43%, and perinatal mortality by 27%
among paucigravidae women (Desai et al, 2007). Unfortunately, the full implementation of antenatal malaria prevention efforts is burdened by the challenges associated
with health care delivery in the developing world.
CONGENITAL TUBERCULOSIS
Tuberculosis remains one of the deadliest communicable
diseases worldwide. More than 2 billion people, equivalent to one third of the world’s population, are infected
with Mycobacterium tuberculosis. Each year, an estimated
529
9 million new tuberculosis cases are identified, and 2 million people die from the disease. The greatest burden
of disease is in developing countries, where tuberculosis
remains a major public health threat. While case rates
have declined steadily in the United States and Europe,
the corresponding numbers have increased dramatically in
the former Soviet Union and Sub-Saharan Africa, in part
fueled by the epidemic of human immunodeficiency virus
(HIV).
Despite the prevalence of tuberculosis worldwide, congenital tuberculosis occurs rarely, with fewer than 400 cases
reported in the English-language literature. The majority
of reports describe infants born in low-burden countries to
mothers who have emigrated from high-burden countries.
Although some reports originate from countries where
tuberculosis is endemic, it is likely that congenital tuberculosis is underrecognized and underreported in these areas,
largely because of the nonspecific clinical features of disease and the limited diagnostic capability.
In the United States, 10,000 to 15,000 cases of tuberculosis are reported annually. Although the overall rate
of tuberculosis infection has been declining steadily since
1992, the proportion of cases in foreign-born populations
has increased. More than 50% of cases reported in 2008
occurring among the foreign born. With increased global
mobility and the epidemiologic trends of tuberculosis, it
is likely that tuberculosis and congenital tuberculosis will
continue to be observed in developed countries. The nonspecific features of congenital tuberculosis and the mortality associated with untreated disease underscore the
importance of maintaining a high index of suspicion for
tuberculosis in pregnant women and young infants.
DEFINITION
Transmission of M. tuberculosis from the mother to the
neonate can occur in utero, intrapartum, or postpartum.
Although congenital infection is classically considered the
result of in utero infection of the fetus, the term congenital
tuberculosis has historically referred to infection acquired
either in utero or intrapartum. The infection can be transmitted by direct spread to the fetus from the placenta via
the umbilical vein or by aspiration or ingestion of infected
amniotic fluid, either in utero or intrapartum.
Beitzke (1935) proposed diagnostic criteria to distinguish congenital tuberculosis from postnatally acquired
tuberculosis. The criteria required that the infant have
proven tuberculosis lesions and one of the following: (1)
a primary hepatic complex as evidence of dissemination
of the tubercle bacilli via the umbilical vein or (2) in the
absence of a primary complex, the presence of tuberculous lesions in the first few days of life or the exclusion
of postnatal infection by separation of the infant at birth
from the mother and other potential sources of infection.
These criteria were developed before the introduction
of chemotherapy, when infant mortality with congenital
tuberculosis was high and diagnosis was largely based on
autopsy findings. It is difficult and no longer practical to
apply Beitzke’s criteria. The demonstration of a primary
hepatic complex with liver and regional node involvement
requires an open surgical procedure; a percutaneous liver
biopsy may demonstrate caseating granulomas, but the
530
PART IX Immunology and Infections
primary complex will seldom be identified. Cantwell et al
(1994) proposed revised criteria that are more applicable
to current practice and improve diagnostic sensitivity. To
meet the criteria, the infant must have proven tuberculous lesions and at least one of the following: (1) lesions
in the first week of age, (2) a primary hepatic complex or
caseating hepatic granulomas, (3) tuberculous infection
of the placenta or maternal genital tract, or (4) exclusion or postnatal transmission by thorough investigation
of contacts. Whereas the distinction between congenital tuberculosis and postnatally acquired disease may be
relevant for academic or epidemiologic purposes, it does
not affect the management, treatment, or prognosis of
disease.
EPIDEMIOLOGY
From 1953 through 1984, the incidence of tuberculosis in
the United States declined steadily, reaching a nadir of 9.4
cases per 100,000 population. From 1985 through 1992,
there was a 20% increase in the total number of cases. The
resurgence of disease was attributed to multiple factors,
including the HIV epidemic, increased immigration, and
a decline in public health funding for tuberculosis control.
With the availability of antiretroviral therapy for HIV and
fortification of public health measures, the epidemiologic
trend was reversed. Since 1992, the case rates for tuberculosis have decreased annually to a case rate of 4.2 per
100,000 in 2008. Despite the decrease in the total burden
of disease, tuberculosis continues to disproportionately
affect the foreign-born and racial and ethnic minorities. In
2008, 59% of all cases of tuberculosis in the United States
occurred in foreign-born persons (CDC, 2008).
The current epidemiology of tuberculosis in pregnancy
is not well delineated. With the resurgence of tuberculosis
in the 1980s, the largest increase in the incidence of disease
occurred in the 25- to 44-year age group, and the number
of cases among women of childbearing age rose by 40%
(Cantwell et al, 1994). In 1991, almost 40% of tuberculosis cases in minority women occurred in those between 15
and 35 years of age (Smith and Teele, 1995). These trends
persist through the present time, thus placing women of
childbearing age, especially those who are foreign born,
and their newborns at continued risk.
The risk of congenital tuberculosis in infants born to
women with tuberculosis is unknown. Blackall (1969)
reported only three cases among infants born to 100 mothers with tuberculosis. Ratner et al (1951) identified no cases
among infants born to 260 mothers with the disease. In a
study of 1369 infants separated at birth from their tuberculous mothers and placed in foster care, only 12 became
tuberculin-positive during 4 years of observation, and in
all 12 cases there was a source of infection in the postnatal environment (Smith and Teele, 1995). The low incidence of congenital tuberculosis is in part attributable to
the high likelihood of infertility in women who have endometrial tuberculosis (Balasubramanian et al, 1999). However, in areas with high rates of tuberculosis transmission,
neonates may be undiagnosed or underreported, and the
incidence of congenital infection or vertical transmission
remains unknown.
Fewer than 400 cases of congenital tuberculosis have
been reported in the literature, with the majority being in
the prechemotherapy era (Laartz et al, 2002). Hageman
et al (1980) reported two cases of congenital tuberculosis
and reviewed another 24 reported in the English-language
literature since the introduction of isoniazid (INH) in
1952. In the subsequent 30 years, more than 30 additional
cases of neonates with congenital tuberculosis have been
described (Abughali et al, 1994; Cantwell et al, 1994; Chen
and Shih, 2004; Doudier et al, 2008; Grover et al, 2003;
Hatzistamatiou et al, 2003; Laartz et al, 2002; Manji et al,
2001; Mazade et al, 2001; Nicolaidou et al, 2005; Pejham
et al, 2002; Saitoh et al, 2001). The more recent reviews
are cited in Table 38-8. Not surprisingly, the majority of
infants were born to foreign-born mothers living in nonendemic areas.
PATHOPHYSIOLOGY
In pregnant women, tuberculous bacillemia can result
in dissemination of infection to the placenta, the endometrium, or the genital tract. Genital tuberculosis that
occurred before pregnancy may be asymptomatic, but
often results in sterility, thus likely accounting for the low
overall frequency of congenital infection. Vertical transmission may occur in one of three ways: (1) hematogenous
spread from the infected placenta via the umbilical vein, (2)
in utero aspiration or ingestion of amniotic fluid infected
from the placenta or endometrium, or (3) ingestion of
infected amniotic fluid or secretions from maternal genital
lesions during delivery. The hematogenous route and in
TABLE 38-8 Reviews of Cases of Congenital Tuberculosis Cases Reported in the English-Language Literature in the Era of Chemotherapy
Years Cases
Reported
No. of
Cases
Hageman
et al, 1980
1952-1980
26
NR
2 of 14
Respiratory distress, fever,
hepatomegaly
46 (12)
Cantwell
et al, 1994
1980-1994
31
Median 24 (range
1 to 84)
0 of 9
Hepatosplenomegaly,
respiratory distress, fever
38 (22)
Abughali
et al, 1994
1952-1994
58
NR
1 of 19?
Respiratory distress,
hepatomegaly, fever
45 (14)
Laartz
et al, 2002
1994-2002
16
Mean 17.4 (range,
1 to 60)
1 of 4
Respiratory distress,
hepatomegaly, fever
20
Reference
NR, Not reported; TST, tuberculin skin test.
Age at Clinical
Presentation (d)
No. of Infants
With Reactive TST
Common Symptoms
Mortality (%)
(With Treatment)
531
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
utero aspiration each probably account for approximately
half of the cases of congenital tuberculosis.
Tuberculous bacilli have been demonstrated in the
decidua, amnion, and chorionic villi of the placenta. It is
unlikely that the fetus can be infected directly from the
mother without the presence of a caseous lesion in the placenta, although massive involvement of the placenta does
not always result in congenital tuberculosis. When a tubercle ruptures into the fetal circulation, bacilli in the umbilical vein can infect the liver, forming a primary focus with
involvement of periportal lymph nodes. The bacilli also
may pass through the liver and right ventricle and into the
lung, or they can enter the left ventricle via the foramen
ovale and pass into the systemic circulation. The organisms
in the lung remain dormant until after birth, when oxygenation and circulation result in their multiplication and
the subsequent development of a primary pulmonary focus.
Alternatively, if the caseous lesion in the placenta ruptures
directly into the uterine cavity and infects the amniotic
fluid, the fetus can inhale or ingest the bacilli, leading to
primary foci in the lung, intestine, or middle ear. Pathologic
examination of tuberculosis in the fetus and newborn usually demonstrates disseminated disease, with the liver and
lungs being principally involved. In Siegel’s study (1934) of
38 postmortem cases, the lungs were involved in 97%, the
liver in 82%, and the spleen in 76% of the infants. Other
sites described are the gastrointestinal tract, kidneys, adrenal glands, and skin (Agrawal and Rehman, 1995; Hageman
et al, 1980; Sood et al, 2000). It is not always possible to
determine whether sites represent multiple primary foci or
are secondary to primary lesions in the lung or liver. The
only lesion in the neonate that is unquestionably associated
with congenital infection is a primary complex in the liver;
all others may be acquired congenitally or postnatally.
M. tuberculosis infection acquired in utero or perinatally
may be indistinguishable from postpartum infection. Postnatal acquisition of M. tuberculosis acquired by airborne
inoculation, either from the mother or another contagious
adult in the infant’s environment, is the most common
route of infection of the neonate. In addition, postnatal
infection can occur from ingestion of infected breast milk
from a mother with a tuberculous breast abscess. In the
absence of a breast abscess, transmission of tuberculosis
via breast milk has not been documented. The distinction
between congenital tuberculosis and postnatally acquired
disease may be important for academic or epidemiologic
purposes, but the management, treatment, and prognosis
of the disease processes are the same.
CLINICAL PRESENTATION
The clinical presentation of congenital tuberculosis is
neither distinctive nor specific. Manifestations of disease
resemble those of neonatal sepsis or other congenital infections. The affected infant is commonly born prematurely
(Amodio et al, 2005; Davis et al, 1960; Foo et al, 1993;
Katumba-Luenya et al, 2005; Premkumar et al, 2008; Wanjari et al, 2008). A retrospective cohort study from Mexico
of infants born to 35 mothers with pregnancies complicated by tuberculosis demonstrated an approximately
twofold risk of prematurity compared with newborns of
mothers without tuberculosis (Figueroa-Damian and
Arredondo-Garcia, 2001). Clinical signs may be evident
shortly after birth, but typically do not appear until 2
to 4 weeks of age (see Table 38-8). Among the 29 cases
reviewed by Cantwell et al (1994), the median age of presentation was 24 days. In an updated review of 16 cases
reported since 1994, the mean age at presentation was
slightly younger at 17.4 days (Laartz et al, 2002).
Before the availability of INH, congenital tuberculosis
was almost uniformly fatal. Notable signs included failure
to thrive, jaundice, and central nervous system involvement.
In the post-INH era, the most commonly described features of disease are respiratory distress, hepatomegaly with
or without splenomegaly, and fever (Abughali et al, 1994;
Cantwell et al, 1994; Hageman et al, 1980). Additional findings are listed in Table 38-9. Although it is important to
evaluate for meningitis in an infant with suspected congenital tuberculosis, central nervous involvement occurs in fewer
than 50% of cases (Hageman et al, 1980; Starke, 1997).
Otitis media with aural discharge has been described as the
presenting sign of congenital tuberculosis (Ng et al, 1996;
Senbil et al, 1997), accompanied by regional lymphadenopathy (Gordon-Nesbitt and Rajan, 1973; Hatzistamatiou
et al, 2003; Figure 38-7) or facial palsy (Pejham et al, 2002).
It is presumed that the infection is due to the accumulation
of infected amniotic fluid in the eustachian tube, either in
utero or at birth. Cutaneous manifestations of congenital
tuberculosis include papular, pustular, or vesicular lesions
often surrounded by erythema (Al-Katawee et al, 2007;
Azimi and Grossman, 1996; Hageman et al, 1980; Loeffler
et al, 1996; Sood et al, 2000). Biopsy of the lesions is often
confirmatory, demonstrating granulomatous inflammation
and the presence of acid fast bacilli (AFB) on tissue stain
(Hageman et al, 1980; Loeffler et al, 1996). A unique case
of congenital tuberculosis involving the spine was recently
reported in India (Grover et al, 2003).
TABLE 38-9 Clinical Signs of Congenital Tuberculosis in
58 Infants
Sign
No. of Patients
% of Patients
Respiratory distress
44
76
Hepatomegaly with or
without splenomegaly
38
65
Fever
33
57
Lymphadenopathy
19
33
Poor feeding
18
31
Lethargy, irritability
16
30
Abdominal distention
15
26
Failure to thrive
9
15
Ear discharge
9
15
Rash
5
9
Abnormal funduscopic
findings
4
7
Jaundice
4
7
Seizure
3
5
Bloody diarrhea
3
5
Ascites
3
5
Adapted from Abughali N, Van Der Kuyp F, Annable W, et al: Congenital tuberculosis,
Pediatr Infect Dis J 13:738-741, 1994.
532
PART IX Immunology and Infections
FIGURE 38-7 Cervical and suboccipital tuberculous lymphadenitis
in a 6-day-old premature infant. (From Hatzistamatiou Z, Kaleyias J,
Ikonomidou U, et al: Congenital tuberculous lymphadenitis in a preterm infant
in Greece, Acta Paediatr 92:392-394, 2003.)
DIAGNOSIS
The timely diagnosis of congenital tuberculosis requires
a high index of suspicion. Clinical signs of disease in the
neonate are nonspecific, and disease in the mother may be
unsuspected, contributing to further delay in diagnosis.
The diagnosis of congenital tuberculosis should be considered in any neonate with suspected infection who is unresponsive to conventional antimicrobial therapy. Evaluation
for suspected disease should include a tuberculin skin test
(TST), chest radiography, lumbar puncture, and mycobacterial culture of appropriate specimens. Biopsy specimens
of affected tissue, either from the infant or the mother,
and the placenta have been confirmatory in several case
reports (Abughali et al, 1994; Cantwell et al, 1994; Chou,
2002; Hageman et al, 1980; Laartz et al, 2002; Loeffler
et al, 1996).
The TST is the most commonly used diagnostic test
for tuberculosis. The test uses 5 tuberculin units of purified protein derivative injected intradermally on the volar
surface of the forearm. The reaction is measured 48 to 72
hours later as millimeters of induration. As many as 10% to
40% of immunocompetent children with culture-proven
tuberculosis do not initially react to a TST. Host factors
such as young age and immunocompromised state can
also decrease the sensitivity of the TST. Specificity may
be compromised by cross reactivity with bacille-CalmetteGuérin vaccine or with environmental nontuberculous
mycobacteria. The TST result is usually negative in neonates with congenital or perinatal tuberculosis, either secondary to immature cell-mediated immunity or because
of overwhelming disease (see Table 38-8). Hageman et al
(1980) found that only 2 of 14 infants who underwent skin
testing had positive test results; on repeated testing, seven
infants subsequently demonstrated positive tuberculin skin
tests, the earliest being at 6 weeks of age, almost 4 weeks
after presentation with clinical signs. Similarly, results of
TSTs performed in 9 of 29 patients described by Cantwell
et al (1994) were all negative, with results of subsequent
testing being positive in 2 of the 9 infants. Among the 16
infants with congenital tuberculosis recently reviewed by
Laartz (2002), three of four infants tested had nonreactive
TST results.
Recent advances in diagnostic tools for tuberculosis
include whole-blood interferon γ release assays (IGRAs),
which are immunologically based tests that measure interferon γ production from lymphocytes in response to antigens that are fairly specific to M. tuberculosis. The two
types of assays currently available include the Quantiferon
Gold (Cellestis, Valencia, California) and the enzymelinked immunosorbent spot assay. Advantages of these
tests include lack of cross-reactivity with bacille-CalmetteGuérin vaccine and most nontuberculous mycobacteria.
The correlation between IGRAs and TSTs is variable, and
negative results do not definitively exclude tuberculosis.
Published experience with the use of IGRAs in children is
limited, and the negative predictive value of these tests in
this population is unclear. Although IGRAs are endorsed
by the CDC for use in circumstances in which a TST
is indicated (Mazurek et al, 2010), the tests are not recommend for use in children younger than 5 years of age
(American Academy of Pediatrics, 2009). Data on the use
of IGRAs in newborns is limited to case reports, and these
assays should not be substituted for TSTs in the evaluation
of congenital tuberculosis.
Given the frequency of respiratory distress in infants
with congenital tuberculosis, it is not surprising that chest
radiograph findings are frequently abnormal at first examination. Typically, a nonspecific parenchymal infiltrate is
noted, although a miliary pattern representative of disseminated disease is occasionally observed (Agrawal and
Rehman, 1995; Airede, 1990; Nemir and O’Hare, 1985;
Pal and Ghosh, 2008; Polansky et al, 1978; Singh et al,
2006; Figure 38-8). Sixteen of 26 patients (62%) reviewed
by Hageman et al (1980) had abnormal radiographic findings on presentation; seven had a miliary pattern and nine
had nonspecific changes. Radiographic abnormalities
developed subsequently in four additional infants. Among
the 29 cases reviewed by Cantwell et al (1994), 23 infants
(79%) had chest radiographic abnormalities, the majority being nonspecific infiltrates. Cavitation secondary to
progressive pulmonary involvement has been reported
(Cunningham et al, 1982). CT imaging of the chest may
demonstrate adenopathy suggestive of tuberculosis or confirm miliary disease (Das et al, 2008; Singh et al, 2006).
An ultrasound or CT image of the abdomen may reveal
enlargement of the liver, spleen, or both, possibly with
areas of abscesses (Amodio et al, 2005; Berk and Sylvester,
2004; Grover et al, 2003; Senbil et al, 1997). Congenital
tuberculosis involving the spine was identified by radiographs and confirmed by magnetic resonance imaging in
India (Grover et al, 2003) (Figure 38-9).
Microbiologic confirmation of disease in the neonate
should be sought using specimens from multiple sites. For
infants and children unable to expectorate sputum, gastric
aspirates are considered the specimens of choice. Additional sources for culture include endotracheal aspirate,
bronchial washing, middle-ear discharge, and lymph node
tissue. CSF should be analyzed and cultured, although isolation of M. tuberculosis from CSF is uncommon (Abughali et al, 1994; Hageman et al, 1980). Traditionally the
detection of mycobacterial organisms by smear or culture
has been considered difficult, because children have paucibacillary disease relative to adults. With three morning
gastric aspirates collected appropriately in hospitalized
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
FIGURE 38-8 Miliary tuberculosis in a neonate with congenital
tuberculosis. (From Singh M, Kothur K, Dayal D, et al: Perinatal tuberculosis a case series, J Trop Pediatr 53:135-138, 2006.)
FIGURE 38-9 Saggital magnetic resonance image on the left demonstrates destruction of T9 to T11 vertebral bodies with the collapse of
T10, leading to a kyphotic deformity causing cord compression. A large
prevertebral collection is also seen. The image on the right demonstrates
destruction with collapse of L5 and S1 vertebral bodies. (From Grover
SB, Pati NK, Mehta R, et al: Congenital spine tuberculosis: early diagnosis by
imaging studies, Am J Perinatol 20:150, 2003.)
533
children with a clinical diagnosis of tuberculosis, only
40% of children had positive cultures (Starke and TaylorWatts, 1989). In comparison, cultures of aspirates from
infants evaluated at the same institution had a 75% yield
(Starke and Taylor-Watts, 1989; Vallejo et al, 1994).
The improved diagnostic yield in infants likely reflects
more widely disseminated and progressive disease, with
higher bacillary loads. Hageman et al (1980) found positive
cultures of M. tuberculosis in 10 of 12 gastric aspirates, 3 of 3
liver biopsy specimens, 3 of 3 lymph node specimens,
and 2 of 4 bone marrow biopsy specimens. Among the
31 cases reviewed by Cantwell et al (1994), noninvasive
procedures and biopsy were useful for the diagnosis of congenital malaria in the majority infants (Table 38-10). More
recent reports confirm the high yield of cultures from a
variety of specimens in neonates (Berk and Sylvester, 2004;
Chou, 2002; Mazade et al, 2001; Premkumar et al, 2008;
Wanjari et al, 2008). Histologic examination of tissue may
suggest the diagnosis before culture results are available.
Biopsy of skin lesions, lymph nodes, and the liver have suggested the diagnosis in several cases by demonstration of
granulomas or acid-fast bacilli on staining before culture
results are available (Berk and Sylvester, 2004; Davis et al,
1960; Hageman et al, 1980).
Whereas M. tuberculosis can require 7 to 21 days for
growth by standard culture technique, results from techniques such as PCR may be available within 48 hours.
A comparison of PCR, AFB smear, and culture with clinical
diagnosis in children found a sensitivity of 60% and a specificity of 97% (Smith, 2002). Although PCR has been useful
for diagnosing congenital tuberculosis in a few case reports,
it is not sensitive enough to preclude obtaining specimens
for culture. Isolation of M. tuberculosis is still important to
determine susceptibilities and to optimize treatment.
The mother of a newborn in whom congenital tuberculosis is suspected is often asymptomatic or has subclinical disease. In the series of congenitally infected infants reported
by Hageman et al (1980), the majority of mothers did not
have a diagnosis until after the disease became apparent in
their infants. Cantwell et al (1994) found that 50% of the
mothers of infected infants were not ill at the time their
newborns exhibited clinical signs of disease. Evaluation of
the mother should include a TST, a chest radiograph, and,
if the radiograph is consistent with tuberculosis disease,
collection of sputum for microbiological confirmation. It
is not unusual for the mother to have extrapulmonary disease such as meningitis or peritonitis (Laartz et al, 2002;
Naouri, 2005), and evaluation may need to be extended to
identify such sites if pulmonary disease is not discovered.
In mothers with no clinical evidence of disease, endometritis should be considered. Pathologic examination and
culture of the placenta (if available) or endometrial biopsy
can confirm the diagnosis of genital transmission (Asensi
et al, 1990; Balasubramanian et al, 1999; Cantwell et al,
1994; Cooper et al, 1985; Niles, 1982; Surve et al, 2006).
In several case reports, the diagnosis of maternal tuberculosis was solely and ultimately made by endometrial biopsy
and culture (Pejham et al, 2002). In addition, culture of
amniotic fluid should be performed. All mothers with
tuberculosis should be tested for HIV infection and, if the
mother is seropositive, the infant should be evaluated for
perinatally acquired HIV infection.
534
PART IX Immunology and Infections
TREATMENT AND MANAGEMENT
The successful management of congenital tuberculosis
depends on early recognition and treatment of disease.
In suspected cases, treatment should not be delayed until
results of cultures or other diagnostic tests are available.
TABLE 38-10 Results of Diagnostic Procedures Performed on
29 Infants With Congenital Tuberculosis Reported from 1980 to
1994
Type of
Specimen
Acid-Fast
Smear*
Mycobacterial
Culture
Smear or
Culture
Gastric aspirate
8/9
8/9
9/11
Endotracheal
aspirate
7/7
7/7
7/7
Ear discharge
2/2
1/1
2/2
Cerebrospinal
fluid
1/2
1/2
1/2
Urine
0/2
0/2
0/2
Peritoneal fluid
1/1
1/1
1/1
Bronchoscopic
specimen
1/1
1/1
1/1
14/19
11/12
16/21
Lymph node
7/8
6/6
7/8†
Liver
4/6
1/2
4/6†
Skin
1/3
1/1
1/3
Lung
1/1
1/1
2/2
Bone marrow
—
1/1
1/1
Ear
1/1
1/1
1/1
Biopsy specimen
Adapted from Cantwell MF, Shehab ZM, Costello AM, et al: Brief report: congenital
tuberculosis, N Engl J Med 330:1051, 1994.
*Results expressed as number of positive results per number of patients tested.
†All biopsy specimens of lymph node and liver that tested negative on smear and
culture showed histopathologic changes consistent with tuberculosis (i.e., giant cell
transformation of granulomas, with or without caseation).
Multiple drug therapy for an extended duration has long
been recognized as the standard of care for tuberculosis.
Because of the rarity of the condition, clinical trials have
not been conducted to establish the optimal treatment
regimen for congenital tuberculosis. It is assumed that the
regimens used for older infants and children are safe and
effective for the treatment of neonates with congenital
tuberculosis. Consultation with a pediatric infectious disease specialist or tuberculosis expert is advised.
Until susceptibility results are known, infants with
proven or suspected tuberculosis should be treated with
a four-drug regimen consisting of INH rifampin (RIF),
pyrazinamide (PZA), and ethambutol (Table 38-11).
Some experts would recommend administration of three
drugs (INH, RIF, PZA) if antimicrobial resistance is not
suspected in the mother, because either she or the source
case are known to have a susceptible strain or she has no
risk factors for resistant M. tuberculosis. The adjunctive
use of corticosteroids is recommended for the treatment
of tuberculosis meningitis based on decreased mortality
and morbidity demonstrated in adults and children (Girgis
et al, 1991). Supplementation with pyridoxine, although
not routinely recommended for otherwise healthy older
children, should be provided to breastfeeding infants
receiving INH. If the M. tuberculosis isolate is determined
to be susceptible, the regimen can be narrowed to three
drugs (INH, RIF, PZA) for the first 2 months of initial
treatment, and subsequently to 2 drugs (INH, RIF) to
complete the continuation phase of treatment. Once the
infant is discharged to home, directly observed therapy
is recommended to ensure adherence and to prevent
relapse.
The optimal duration of treatment for infants with congenital tuberculosis is unknown. The typical duration of
treatment for susceptible M. tuberculosis is 6 months for
pulmonary disease, pulmonary disease with hilar adenopathy, or hilar adenopathy alone (American Academy of
TABLE 38-11 Commonly Used Drugs for Treatment of Tuberculosis in Infants, Children, and Adolescents
Daily Dose
(mg/kg)
Twice per Week
Dose (mg/kg)
Tablets (100,400 mg)
20-25
50
Scored tablets
(100, 300 mg)
Syrup 10 mg/mL
10-15†
20-30
Pyrazinamide
Scored tablets
(500 mg)
30-40
50
Rifampin
Capsules
(150, 300 mg)
Syrup formulated
capsules
10-20
10-20
Drugs
Dose Forms
Ethambutol
Isoniazid
Maximum Dose
Adverse Reactions
2.5 g
Optic neuritis (usually reversible),
decreased red-green color discrimination, gastrointestinal tract
disturbances, hypersensitivity
300 mg daily
Mild hepatic enzyme elevation,
hepatitis,† peripheral neuritis,
hypersensitivity
Diarrhea and gastric irritation
caused by vehicle in the syrup
900 mg twice per week
2g
Hepatotoxic effects, hyperuricemia,
arthralgia, gastrointestinal tract
upset
600 mg
Orange discoloration of secretions
or urine, staining of contact
lenses, vomiting, hepatitis,
influenza-like reaction, thrombocytopenia, pruritus; oral contraceptives may be ineffective
From American Academy of Pediatrics Committee on Infectious Diseases: Section 3. Summaries of Infectious Diseases. Tuberculosis. In Pickering LK, editor: 2009 red book: report of
the committee on infectious diseases, ed 28, Elk Grove Village, Ill, 2009, American Academy of Pediatrics, p 688.
†When isoniazid in a dose exceeding 10 mg/kg/d is used in combination with rifampin, the incidence of hepatotoxic effects may be increased.
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
Pediatrics, 2009). For extrapulmonary disease, the duration is extended to 9 to 12 months, and for drug-resistant
M. tuberculosis the duration may be extended even further
to prevent failure or relapse. Most experts would treat
infants with congenital tuberculosis for 9 to 12 months
because of the decreased immunocompetence of neonates
(Starke, 1997).
Although there is a fair amount of data to support the
safety of INH, data on the safety and pharmacokinetics of
other agents are limited. Careful monitoring for signs and
symptoms of hepatitis and other adverse effects of drug
therapy is recommended. Routine determination of serum
transaminases in children is indicated with severe tuberculosis (e.g., military or meningitis), for those with concurrent liver or biliary disease, or for those receiving other
potentially hepatotoxic drugs. Routine monitoring of liver
function also should be considered for neonates with congenital tuberculosis given the paucity of data on adverse
effects of anti-tuberculosis agents in this age group (Patel
et al, 2008). The risks of optic neuritis with ethambutol
should be considered when this agent is used, and vision
should be monitored periodically.
The management of infants born to mothers who have
latent tuberculosis infection (LTBI) or tuberculosis disease is outlined in Figure 38-10. Recommendations are
based on the categorization of infection in the mother and
the potential risk of transmission of tuberculosis to the
infant (American Academy of Pediatrics, 2009). Infants
born to mothers with potentially contagious tuberculosis
should be evaluated for congenital tuberculosis. Separation of the infant and mother is necessary only in cases in
which the mother is highly infectious at the time of delivery. The mother with latent tuberculosis infection is not
contagious. To prevent reactivation disease in the mother
and subsequent exposure of the infant, the mother should
receive treatment with INH for LTBI. In addition, latent
tuberculosis infection in the mother may be a marker for
contagious tuberculosis within the household, and it is recommended that all household members and close contacts
of the mother be evaluated for tuberculosis.
Breastfeeding is not contraindicated in women with
LTBI. The breast milk of a woman with tuberculosis does
not contain tubercle bacilli. For women with tuberculosis
who are potentially infectious and separated from the newborn, breast milk may be manually expressed and fed to
the infant. Once the mother is noninfectious or the infant
is receiving therapy, breastfeeding can be resumed. The
exception, however, is the mother with an active tuberculous breast lesion. In this situation the breast milk may
be pumped and discarded until resolution of the lesion
(Efferen, 2007).
PROGNOSIS
The prognosis for congenital tuberculosis was dismal in
the prechemotherapy era, the diagnosis often being only
made at autopsy. Although the survival rate subsequently
improved, mortality remained approximately 50% secondary to delayed diagnosis. In a review of 26 cases reported
between 1952 and 1980, 12 (46%) patients died, 9 of whom
were untreated but with a diagnosis made at autopsy. The
subsequent reviews demonstrate a decrease in case fatality
535
(see Table 38-8), with earlier diagnosis and treatment.
Timely diagnosis and initiation of anti-tuberculosis therapy are critical for a favorable outcome.
PREVENTION
Prevention of congenital tuberculosis requires the treatment and prevention of disease in women of childbearing
age. Risk factors for acquiring tuberculosis infection or
progressing to disease should be assessed at prenatal visits,
and women identified as being high risk for LTBI or progression to disease should undergo tuberculin skin testing
as recommended by the CDC (Centers for Disease Control, 2000). Women who are TST positive should undergo
evaluation for active disease. The treatment of active
tuberculosis during pregnancy is considered standard, and
early treatment has been shown to improve maternal and
neonatal outcome (Figueroa-Damian and ArredonondoGarcia, 1998). While not without potential complications, treatment during pregnancy is less of a hazard to
a pregnant woman and her fetus than tuberculosis itself.
The treatment of LTBI during pregnancy is somewhat
more controversial. Some experts advocate treatment during pregnancy, whereas others support a delay in therapy
until weeks to months after delivery. Although there is no
demonstrated teratogenic potential for the use of INH,
there is concern that pregnant and postpartum women are
more vulnerable to INH-related hepatotoxicity (Franks
et al, 1989).
Most children with pulmonary tuberculosis, especially
those younger than 10 years, have paucibacillary disease
and often have little to no cough. Isolation of the hospitalized pediatric patient is directed at accompanying adult
contacts who may be source cases and potentially contagious. Visitation of the hospitalized pediatric patient
should be restricted to adults in whom contagious tuberculosis has been excluded. Hospitalized children with
negative sputum AFB smears (if obtained) require standard precautions, assuming that contagious tuberculosis
has been excluded in the visitors. Airborne precautions
are recommended for the following pediatric patients: (1)
children and adolescents with adult-type cavitary disease,
(2) extensive pulmonary infection, (3) those with smears
positive for AFB, and (4) congenitally infected neonates
undergoing endotracheal intubation (American Academy
of Pediatrics, 2009).
Compared with older children, neonates likely have a
higher concentration of bacilli in their sputum. As noted
previously, AFB smears on tracheal aspirates and other
specimens are frequently positive in this population compared with older children. Transmission of tuberculosis
from congenitally infected neonates to health care workers and other hospitalized infants has been reported and
is likely related to aerosolization of bacilli during respiratory manipulation (Crockett et al, 2004; Laartz et al,
2002; Lee et al, 1998; Mouchet et al, 2004). Neonates suspected of having congenital tuberculosis should be placed
in respiratory isolation if intubated or if undergoing any
procedure with the potential for aerosolization of infected
sputum. Exposed infants, visitors, and health care workers should undergo evaluation for tuberculosis infection
or disease.
536
PART IX Immunology and Infections
Mother: Tuberculin skin test positive*
Symptomatic infant †
Asymptomatic infant
Maternal chest x-ray
normal; no active
disease
Maternal chest
x-ray abnormal †
Mother with clinical or
radiographic evidence of
contagious TB †
Maternal evaluation consistent with TB?
• TST, chest x-ray
• Lumbar puncture
• AFB culture: gastric aspirate,
endotracheal aspirate, CSF
• Head CT scan or MRI
• Multi-drug treatment (see text)
• Airborne isolation
• Consult infectious disease specialist
Yes
No ‡
Maternal treatment for TB
2 wk, sputum AFB
negative, and strain not
multi-drug resistant
Maternal treatment none
or 2 wk, sputum AFB
positive, or strain is
multi-drug resistant
No infant evaluation or therapy required
• Separate infant from mother until she is non-contagious (treatment 2wk) or infant on appropriate TB drug(s)
• Evaluate infant for congenital tuberculosis:
• TST; chest x-ray
• AFB culture: gastric aspirate, endotracheal aspirate
• Consider lumbar puncture and AFB culture of CSF
• Consult infectious disease specialist; notify local health department
Evaluation consistent with congenital tuberculosis?
No
• INH for 3–4 months
• If maternal TB strain is multi-drug resistant, consider
BCG vaccine
• Follow-up tuberculin skin test at 3–4 months:
• Negative test: stop INH
• Positive test:
• Reassess for TB as in symptomatic infant
• If no other evidence of TB, INH for 9 months
Yes
• Lumbar puncture if not previously done
• Head CT scan or MRI
• Multi-drug treatment (see text)
FIGURE 38-10 Management of infants born to mothers with a positive tuberculin skin test (TST) result. BCG, Bacille Calmette-Guérin; CSF,
cerebrospinal fluid; CT, computed tomography; INH, isoniazid; MRI, magnetic resonance imaging. (Adapted from American Academy of Pediatrics:
Tuberculosis. In Pickering LK, editor: 2003 Red book: report of the committee on infectious diseases, ed 26, Elk Grove Village, Ill, 2003, American Academy of Pediatrics, p 642.)
*Household contacts should have a TST and further evaluation for contagious tuberculosis (TB). Consult local health department. The mother should
receive treatment for latent tuberculosis infections. All persons with TB should be tested for human immunodeficiency virus (HIV) infection.
†Acid-fast bacillus (AFB) culture of amniotic flid and placenta, if available; placenta for histopathologic examination.
‡Includes mother with chest radiographic findings consistent with old, healed TB.
CHAPTER 38 Toxoplasmosis, Syphilis, Malaria, and Tuberculosis
SUGGESTED READINGS
Toxoplasmosis
Beaman MH, Luft BJ, Remington JS: Prophylaxis for toxoplasmosis in AIDS, Ann
Intern Med 117:163-164, 1992.
Boyer K: Diagnostic testing for congenital toxoplasmosis, Pediatr Infect Dis J
20:59-60, 2001.
Centers for Disease Control and Prevention: CDC recommendations regarding
selected conditions affecting women’s health: Preventing congenital toxoplasmosis, MMWR Morb Mortal Wkly Rep 49(RR-2):59-68, 2000.
Feldman DM, Timms D, Borgida AF: Toxoplasmosis, parvovirus and cytomegalovirus in pregnancy, Clin Lab Med 30:709-720, 2010.
Jones JL, Dargelas V, Roberts J, et al: Risk factors for Toxoplasma gondii infection
in the United States, Clin Infect Dis 49:878-884, 2009.
Kasper DC, Sadeghi K, Prusa AR, et al: Quantitative real-time polymerase chain
reaction for the accurate detection of Toxoplasma gondii in amniotic fluid, Diagn
Microbiol Infect Dis 63:10-15, 2009.
Koppe JG, Loewer-Sieger DH, DeRoever-Bonnet H: Result of 20‑year follow-up
of congenital toxoplasmosis, Lancet 1:254-256, 1986.
Masur H, Kaplan JE, Holmes KK, et al: Guidelines for preventing opportunistic
infections among HIV-infected persons: 2002 recommendations of the U.S.
Public Health Service and the Infectious Diseases Society of America, MMWR
Morb Mortal Wkly Rep 51(RR-8):5, 2002.
McLeod R, Kieffer F, Sautter M, et al: Why prevent, diagnose and treat congenital
toxoplasmosis?, Mem Inst Oswaldo Cruz 104:320-344, 2009.
Rabilloud M, Wallon M, Peyron F: In utero and at birth diagnosis of congenital
toxoplasmosis: use of likelihood ratios for clinical management, Pediatr Infect
Dis J 29:421-425, 2010.
Syphilis
American Academy of Pediatrics Committee on Infectious Diseases: Syphilis, Red
Book Online: 2009. Available at http://aapredbook.aappublications.org/cgi/
content/full/2009/1/3.129. Accessed November 17, 2010.
Beeram MR, Chopde N, Dawood Y, et al: Lumbar puncture in the evaluation of
possible asymptomatic congenital syphilis in neonates, J Pediatr 128:125-129,
1996.
Centers for Disease Control and Prevention: Sexually Transmitted Diseases
Treatment Guidelines, 2010. MMWR 59:1-110, 2010. Accessed
537
Centers for Disease Control and Prevention: Congenital syphilis: United States,
2003-2008, MMWR 59:413-417, 2010.
Herremans T, Kortbeek L, Notermans DW: A review of diagnostic tests for congenital syphilis in newborns, Eur J Clin Microbiol Infect Dis 29:495-501, 2010.
Ingall J, Sanchez P, Baker C: Syphilis, In Remington JS, Klein JO, Baker C, et al:
Infectious diseases of the fetus and newborn infant, ed 6, Philadelphia, 2006, WB
Saunders, pp 545-580.
World Health Organization Department of Reproductive Health and Research:
The global elimination of congenital syphilis: rationale and strategy for
action: 2007. Available at www.who.int/reproductivehealth/publications/rtis/
9789241595858/en/index.html. Accessed November 17, 2010.
Malaria
Desai M, ter Kuile FO, Nosten F, et al: Epidemiology and burden of malaria in
pregnancy, Lancet Infect Dis 7:93-104, 2007.
Hulbert TV: Congenital malaria in the United States: report of a case and review,
Clin Infect Dis 14:922-926, 1992.
Lesko CR, Arguin PM, Neman RD: Congenital malaria in the United States: a
review of cases from 1966 to 2005, Arch Pediatr Adolesc Med 161:1062-1067,
2007.
Menendez C, Mayor A: Congenital malaria: the least known consequence of
malaria in pregnancy, Semin Fetal Neonatal Med 12:207-213, 2007.
Tuberculosis
Abughali N, Van Der Kuyp F, Annable W: Kumar ML: Congenital tuberculosis,
Pediatr Infect Dis J 13:738-741, 1994.
Cantwell MF, Shehab ZM, Costello AM, et al: Brief report: congenital tuberculosis, N Engl J Med 330:1051-1054, 1994.
Hageman J, Shulman S, Schreiber M, et al: Congenital tuberculosis: critical reappraisal of clinical findings and diagnostic procedures, Pediatrics 66:980-984,
1980.
Laartz BW, Narvarte HJ, Hold D, et al: Congenital tuberculosis and management
of exposures in a neonatal intensive care unit, Infect Control Hosp Epidemiol
23:573-579, 2002.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
39
Neonatal Bacterial Sepsis
Patricia Ferrieri and Linda D. Wallen
NEONATAL BACTERIAL SEPSIS
Throughout pregnancy, the fetus is protected from bacterial and viral infections by the chorioamniotic membranes,
the placenta, and various antibacterial factors, that are
poorly described in amniotic fluid. It is thought that some
subclinical infections of the fetus, amniotic fluid, membranes, or placenta may contribute to the onset of preterm
labor and the delivery of preterm infants. There are several
mechanisms by which bacteria can reach the fetus or newborn and initiate infection. Maternal blood stream infections, caused by bacteria such as Listeria monocytogenes and
Mycobacterium tuberculosis, can reach the fetus and cause
infection. Bacteria such as group B streptococcus (GBS)
can be acquired from the vagina, cervix, or fecal contamination of the birth canal through either ruptured or intact
membranes, leading to amnionitis, intrauterine pneumonitis, and premature delivery (Ancona et al, 1980; Ferrieri,
1990; Larsen and Sever, 2008; Payne et al, 1988). Finally,
infection can occur via aspiration of birth canal contents or
colonization of mucosal surfaces during passage through
the birth canal, leading to pneumonia, followed by bacteremia and sepsis after 1 day or later; this may be the mode
by which Neisseria gonorrhoeae, Escherichia coli, and GBS are
acquired.
Early-onset bacterial sepsis remains a major cause of
neonatal morbidity and mortality, although the sepsisassociated death rates per 100,000 live births have declined
significantly from 2001-2011. Much of this decline in
mortality is because of the introduction of intrapartum
antibiotic prophylaxis in pregnant women during labor
and delivery (Centers for Disease Control and Prevention, 2007, 2009; Schrag et al, 2002; Schrag and Stoll,
2006). Mortality rates in infected premature infants and
very immature infants are significantly higher than in term
infants. Major improvements in neonatal intensive care
and early identification and recognition of infected infants
have all contributed to reduced mortality rates in the newborn period.
PATHOGENESIS OF EARLY-ONSET
NEONATAL BACTERIAL INFECTIONS
There are multiple portals through which bacteria can
enter and infect the newborn. The primary portals of entry
appear to be the respiratory tract, as suggested by the high
frequency of acute respiratory distress and pneumonia,
which occurs in infants with early-onset disease. Acquisition via the placenta is suggested in some instances by the
presentation of high-grade bacteremia and severe sepsis
clinically apparent at the time of birth in the presence of
intact membranes in infants born via cesarean section. The
primary maternal event in this sequence, leading to infection of the fetus and newborn infant, is colonization of the
maternal genital tract with organisms such as the GBS.
538
Bacteria that reside in the cervix, vagina, or rectum can
ascend into the amniotic cavity through intact or ruptured
membranes and lead to chorioamnionitis. Bacteria can initially spread into the choriodecidual space and can occasionally cross intact chorioamniotic membranes. Although
organisms recovered from the amniotic sac in the mother
are usually polymicrobial and include such organisms as
the GBS, group D enterococcus, aerobic gram-negative
bacteria, and anaerobes such as Bacteroides spp. (Gibbs
et al, 1980), a single organism causing bacterial sepsis is
the rule in de novo sepsis of the newborn infant. Genital
microplasmas are at times recovered from women as well as
Chlamydia spp., but the precise pathogenic role is unclear
(Krohn et al, 1995; Pankuch et al, 1984). Ureaplasma spp.,
and Chlamydia spp. can be isolated from infants’ respiratory tract after birth; these two plus Mycoplasma hominis
can be recovered from the respiratory tract after birth, but
they are not associated with sepsis syndrome.
Although many microorganisms recovered from the
amniotic cavity are thought to induce spontaneous preterm labor, and possibly premature rupture of membranes,
the exact mechanisms by which this may occur are debatable. Clinical or subclinical chorioamnionitis can incite a
marked inflammatory response with the release of cytokines that can contribute to the onset of preterm labor and
premature rupture of membranes. Other risk factors for
clinical intraamniotic infection include young maternal
age, prolonged labor, prolonged rupture of membranes
(≥18 hours), internal scalp fetal monitoring, the presence of urinary tract infections, and a history of bacterial
vaginosis (Newton et al, 1989; Soper et al, 1989). Despite
inherent antibacterial properties in amniotic fluid, these
may not be sufficient to overcome a large bacterial inoculum, because of rapid multiplication of bacteria during a
prolonged labor or the absence of type-specific maternal
antibodies for various pathogens (Ferrieri, 1990).
Infants who immediately display signs of respiratory
distress and after birth undoubtedly have onset of infection before or during labor and delivery. Particularly with
hypoxia in utero, the infant may gasp and inhale contaminated amniotic fluid, leading to pneumonia, blood stream
infection, sepsis, and a severe systemic response syndrome.
Infants who display such signs at birth or within a short
time after birth have the highest mortality rates. Infants
who have an initial asymptomatic period after birth may
display symptoms gradually as the organisms multiply in
the lung and in the blood. An example of another invasive site of entry is the scalp lesion created by a monitoring device, which becomes contaminated in the setting
of amniotic fluid infected with the GBS. An overarching
mechanism for continued bacteremia is the absence of sufficient local and systemic host defenses, such as adequate
complement levels or type-specific immunity against the
invading microorganism (Ferrieri, 1990).
CHAPTER 39 Neonatal Bacterial Sepsis
The inflammatory cascade is initiated by activation of
macrophages by bacterial cell wall constituents, toxins,
or enzymes. A number of proinflammatory cytokines can
be released, such as interleukin (IL) 6, IL-8, and tumor
necrosis factor-α (TNF-α). These cytokines can alter vascular permeability and vascular tone, decrease myocardial
contractility, activate clotting systems, increase pulmonary
vascular resistance, and activate other phagocytic cells,
such as polymorphonuclear leukocytes (PMNs). Ideally,
proinflammatory and antiinflammatory cytokines would
be balanced; however, this is usually not the case and
the bacteria persist with subsequent consequences. It is
common in newborn infants and, particularly in preterm
infants, to have dissemination of bacteria to other organs
such as the meninges, kidneys, and bone.
EPIDEMIOLOGY OF EARLY-ONSET
BACTERIAL INFECTIONS
Before the availability and use of antibiotics in the late
1940s and 1950s, there were few survivors of neonatal bacterial sepsis, contributing to the high perinatal mortality
rate of that period. There have been changes in the types
of bacteria responsible for neonatal infection over the
years. In the 1930s and 1940s, the group A streptococcus
was a prominent cause of neonatal sepsis; this organism is
now rather rare (Bizzarro et al, 2005). In the 1950s, nursery outbreaks of Staphylococcus aureus infections appeared
across North America and Europe, prompting changes in
techniques of hygiene and encouraging the development
and use of penicillinase-resistant antibiotics (Bizzarro
et al, 2005). In the 1960s, Escherichia coli became the most
common cause of bacterial sepsis, followed in the 1970s
by the GBS. Even in an era of intrapartum antibiotic prophylaxis of GBS colonized mothers, the GBS remains the
most common bacterial pathogen in neonatal centers of
North American and Europe, followed by E. coli (Schrag
et al, 2006). An update on neonatal sepsis at Yale, between
1989 to 2003, revealed an overall decrease in sepsis caused
by both GBS and E. coli (Bizzarro et al, 2005). Regional
differences exist, however, and must be considered before
attempting to apply epidemiologic data to individual perinatal units. For example, Listeria monocytogenes is a frequent
isolate in some western European countries, and S. aureus
is found commonly in Germany and Scandinavia (PosfayBarbe and Wald, 2009).
The incidence of early-onset bacterial infection is variable and ranges from one to five per 1,000 live births; however, it is clear that the incidence has declined as a result
of intrapartum antibiotic therapy (Centers for Disease
Control and Prevention, 2007, 2009). Recent data from
the CDC revealed a downward trend from 2000 to 2003
(from 0.52 to 0.31 case per 1000 live births), followed by
an increase from 2003 to 2006 (from 0.31 to 0.40 case per
1000 live births) (Centers for Disease Control and Prevention, 2009). Stratified by race, the incidence increased significantly among black infants from 2003 to 2006 (from
0.53 to 0.86 case per 1000 live births), whereas the incidence among white infants did not change significantly
(from 0.26 to 0.29 case per 1000 live births). When stratified by gestational age, the average incidence of early-onset
GBS disease among preterm infants during 2003 to 2006
539
was 2.8-fold higher among black infants compared with
white infants (1.79 case versus 0.67 case per 1000 live
births). It is of interest that both preterm black and white
infants had increases in early-onset disease from 2003 to
2006 that were not statistically significant. Early-onset
disease among full-term white infants was stable during
2003 to 2006, whereas term black infants had a significant
increase of the incidence during this period, from 0.33 to
0.7 case per 1000 live births.
The overall rates of late-onset GBS disease remained
stable from 2000 to 2006 (0.36 case versus 0.30 case per
1000 live births) (Centers for Disease Control and Prevention, 2009). No overall incidence trend was observed from
2003 to 2006. When stratified by race, late-onset disease
incidence among black infants decreased significantly by
42% from 2005 to 2006 (0.95 case versus 0.55 case per
1000 live births). Between 2003 and 2006 there were no
significant trends among black or white infants.
Infants described with early-onset sepsis frequently have
one or more identifiable risk factors (Dutta et al, 2010).
Prematurity is considered the single greatest risk factor for
early-onset bacterial infections. Because it is accepted that
ELBW infants have impairment of host defenses, and since
preterm birth may be associated with low-grade chorioamnionitis, it is not surprising that the attack rates for infection by pathogens such as GBS are 26- to 30-fold higher
in preterm infants, compared to term newborn infants,
with an associated high mortality. Other risk factors for
early-onset sepsis are maternal age, health and nutrition,
colonization with well-known pathogens (e.g., GBS), and
rupture of membranes for longer than 18 hours (Schuchat
et al, 2000). Neonatal susceptibility to GBS infection is
increased with deficiencies in circulating levels of GBS
type-specific antibody and complement, which is further
heightened by any element of neutrophil dysfunction, as
may be seen in the more premature infants (Ferrieri, 1990;
Foxman, 2007; Makhoul et al, 2009; Nandyal, 2008).
BACTERIAL PATHOGENS IN EARLYONSET INFECTIONS
GROUP B STREPTOCOCCAL INFECTIONS
Since the early 1930s when Rebecca Lancefield reported
her grouping system for hemolytic streptococci, group
A streptococcus (Streptococcus pyogenes) was widely acknowledged as the major pathogen associated with puerperal
sepsis. GBS was initially thought to be a commensal until
1938, when Frye reported seven cases of GBS-associated
puerperal fever with three deaths (Eickhoff et al, 1964).
Before the 1960s, GBS was not recognized frequently as a
cause of human disease. However, in the late 1960s GBS
emerged as the leading cause of neonatal sepsis in newborn
infants (Bizzarro et al, 2005; Zaleznik et al, 2000). Before
the era of maternal intrapartum prophylaxis, GBS had a
reported national incidence of approximately two per 1000
live births and was associated with approximately 50%
mortality in the newborn infant. As mentioned previously,
over the past decade with the introduction of antibiotic
maternal prophylaxis, there has been a significant decrease
in the incidence of GBS to its current rate of approximately
0.32 per 1000 live births for early-onset disease.
540
PART IX Immunology and Infections
Transmission of GBS from Mothers
to Infants
In the United States, approximately 20% to 35% of pregnant women are asymptomatic carriers of GBS in the
genital and gastrointestinal tract during pregnancy and at
the time of delivery (Ferrieri, 1990; Ferrieri et al, 2004b;
Zaleznik et al, 2000). The prevalence of GBS colonization during pregnancy varies. Among women who were
positive for GBS between 26 and 28 weeks’ gestation,
only 65% remain colonized at term, whereas 8% of those
with negative prenatal cultures were positive for GBS at
term (Ancona et al, 1980; Zaleznik et al, 2000). Treatment of GBS-colonized women during pregnancy only
temporarily eradicates the organism, and most women
are recolonized within several weeks. At birth, 50% to
65% of infants who were born to GBS-colonized mothers
have positive GBS cultures from mucous membranes and
skin (external ear canal, throat, umbilicus, and anal or rectal sites) (Shet and Ferrieri, 2004). Before the introduction of intrapartum antibiotic prophylaxis, approximately
1% to 2% of colonized infants developed GBS infection, and the overall incidence of neonatal GBS infection was approximately two per 1000 live births in the
United States. With intrapartum prophylaxis, approximately 60% to 80% of GBS cases occur in infants born to
women with negative antenatal GBS screens (Verani and
Schrag, 2010).
A small number of GBS-infected infants acquired
their bacteremia because of hematogenous transmission
through the placenta. In these situations, the mother commonly displays signs and symptoms of chorioamnionitis,
although it may occur in the absence of maternal symptoms (Baker and Edwards, 1995).
Detection of GBS colonization has been emphasized
since approximately 1996; studies to determine the optimal sites of sampling have been key to the effectiveness
of intrapartum prophylaxis. GBS resides in the genitourinary and gastrointestinal tracts, where large numbers
of gram-negative bacteria are also present. The majority of colonization studies have revealed high rates of
both rectal and vaginal colonization with GBS (Ancona
et al, 1980; Hickman et al, 1999; Zaleznik et al, 2000).
The use of the selective broth enrichment medium that
inhibits the growth of gram-negative enteric bacilli and
other normal flora can increase culture sensitivity for
GBS to greater than 90%. The most widely used selective medium is Todd-Hewitt broth with either gentamicin or colistin and nalidixic acid. As recommended in a
2002 publication from the Centers for Disease Control
and Prevention, the optimal time for performing antenatal cultures is between 35 and 37 weeks’ gestation,
and the highest culture yield is obtained when both the
lower vaginal area and anal or rectal sites are sampled
(Schrag et al, 2002).
Epidemiologic studies of GBS-colonized women have
shown that those with heavy colonization (3+ to 4+) are
more likely to transmit GBS to their infants (Ancona et al,
1980). The colonization of GBS in pregnant women may
be long standing, intermittent, or transient. There is a definite association between GBS colonization and other risk
factors for neonatal sepsis; these include preterm labor,
preterm delivery, premature rupture of membranes, prolonged rupture of membranes, and maternal fever.
In the past few years, rapid diagnostic tests to detect
GBS colonization in pregnant women have included realtime polymerase chain reaction (PCR); compared with
broth enrichment cultures, there is an approximately 10%
to 15% increased sensitivity (P. Ferrieri, unpublished
data). The advantages of PCR detection of maternal vaginal or rectal colonization are the rapid turnaround time, as
well as the increased sensitivity. Although more expensive
than traditional culture-based detection assays, the results
are available 1 to 2 days sooner. The argument that this
does not provide semiquantitative data on the degree of
GBS colonization in the mother is moot, because even low
grade colonization in pregnant women is a risk factor for
neonatal GBS sepsis. However, women with heavy (3+ to
4+) colonization, determined by semiquantitative assessment of vaginal or rectal cultures, are more likely to pass
the microorganism to their infants (Ancona et al, 1980).
Chemoprophylaxis and Intrapartum
Antibiotics
Prevention is of key importance in decreasing invasive
GBS disease. The challenge has been to widely promulgate
screening cultures in pregnant women. Revised guidelines
from the CDC were published in 2002 and presented
only the culture screening based approach for prevention
and chemoprophylaxis, rather than the two preventive
approaches published in 1996: a culture screening-based
and a risk-based approach (Schrag et al, 2002). The riskbased approach involved the use of antibiotics based solely
on the presence of antenatal or intrapartum risk factors
such as maternal fever, preterm labor or premature rupture of membranes (<37 weeks’ gestation); prolonged
rupture of membranes (≥18 hours); history of a previous
newborn infant with GBS disease; and GBS bacteruria
during pregnancy. Challenges to the implementation of
the CDC guidelines, such as failure to seek prenatal care
and the use of suboptimal laboratory culture techniques
continue in certain populations. Data for the United States
as a whole show a decrease in the incidence of early-onset
GBS concurrent with the implementation of maternal
GBS screen and intrapartum antibiotic prophylaxis guidelines. The current estimate for the overall United States
population for early-onset GBS disease is 0.32 per 1000
live births (Van Dyke et al, 2009).
Intrapartum Antibiotic Prophylaxis
GBS is sensitive to penicillin, which is the drug of choice
because of its narrow spectrum; the alternative is ampicillin. If a mother is allergic to penicillin but not at high
risk for anaphylaxis, the use of cefazolin has been proposed
(Schrag et al, 2002). When patients are at high risk for
anaphylaxis in which the GBS is known to be susceptible to
clindamycin and erythromycin, they can receive either of
these drugs intravenously (IV) until delivery. When GBS
is resistant to clindamycin or erythromycin or the antibiotic susceptibility is unknown, vancomycin given every
12 hours IV until delivery is the current recommendation
(Schrag et al, 2002). In the United States, GBS exhibits
CHAPTER 39 Neonatal Bacterial Sepsis
considerable resistance against erythromycin (5% to 32%)
and clindamycin (3% to 21%) (Castor et al, 2008). It is
therefore important to have antibiotic testing done on the
group B streptococcal isolates from pregnant women. For
laboratories performing PCR on maternal vaginal or rectal
cultures, it is recommended that the swabs be placed in
a selective enrichment broth containing inhibitory antibiotics (either colistin and nalidixic acid or gentamicin and
nalidixic acid) against gram-negative bacteria. If the PCR
result is positive, the broth culture can be subcultured and
antibiotic testing can be pursued. Because of the higher
sensitivity of PCR, compared with the selective brothenrichment culture, the organism will not grow in 10% to
15% of occasions.
Group B Streptococcal Sepsis in Neonates
The majority of infections in newborn infants occur within
the first week of life and are designated as early-onset disease (Table 39-1). Late-onset infections occur in infants
7 days or older, with the majority of these infections
appearing in the first 3 months of life. Although chemoprophylaxis has led to a significant decrease in the incidence of early-onset GBS disease, there is no evidence that
chemoprophylaxis prevents late-onset disease (CohenWolkowiez et al, 2009; Hamada et al, 2008; Jordan et al,
2008). Young infants with early-onset invasive GBS disease usually have pneumonia, sepsis, often; less often they
have meningitis, osteomyelitis, or septic arthritis (Koenig
and Keenan, 2009). The frequency of meningitis, osteomyelitis, or septic arthritis is higher among infants with
late-onset disease.
There are nine antigenically distinct GBS serotypes,
based on their capsular polysaccharide analysis (types
Ia, Ib, II to VIII) and a proposed new type, IX (Diedrick
et al, 2010; Henrichsen et al, 1984; Slotved et al, 2007).
In the United States and Western Europe, types Ia, II,
and III account for the majority of isolates from infants
TABLE 39-1 Manifestations of Early-Onset and Late-Onset
Group B Streptococcal Disease
Early-Onset
Disease
Late-Onset
Disease
Age at onset
Birth through day
6 of life
Day 7 to
3 months
Symptoms
Respiratory
distress, apnea
Irritability, fever,
poor feeding
Findings
Pneumonia, sepsis
Sepsis, meningitis,
osteoarthritis
Maternal obstetrical
complications
Frequent
Uncommon
Mode of transmission
Vertical, in utero,
or intrapartum
Nosocomial,
horizontal
Predominant serotypes
Ia, III, V*
III, Ia, V*
Effect of intrapartum
antibiotic prophylaxis
recommended by the
Centers for Disease
Control and Prevention
Reduces incidence
by 85%-90%
No effect
Characteristic
*In decreasing order of frequency
541
with early-onset disease (Diedrick et al, 2010; Zaleznik
et al, 2000). However, recent studies in the United States
have demonstrated that serotypes Ia, III, and V, the latter emerging in recent years (Diedrick et al, 2010; Elliott
et al, 1998; Harrison et al, 1998), account for the majority (70-75%) of early-onset invasive disease in newborn
infants and parturient women (Diedrick et al, 2010;
Zaleznik et al, 2000). Late-onset GBS disease in infants
is dominated by serotype III followed by serotypes Ia and
V (Shet and Ferrieri, 2004). A polysaccharide capsule is
considered the most important virulence factor (Cieslewicz et al, 2005; Kasper et al, 1996; Paoletti et al, 1997,
2001); however, the role of surface localized GBS proteins (Ferrieri et al, 2004a; Johnson and Ferrieri, 1984;
Lindahl et al, 2005; Smith et al, 2004) in pathogenesis and
immune protection has gained favor (Maione et al, 2005;
Tettelin et al, 2005).
The remaining GBS isolates from invasive disease consist primarily of types Ib and II, but types IV, VI, VII, and
VIII compose a small fraction. Type IV GBS represented
between 0.4% and 0.6% of colonizing GBS isolates (Diedrick et al, 2010), but it was relatively uncommon for type
IV isolates to be found in invasive GBS (Ferrieri et al,
2008; Puopolo and Madoff, 2007). Recent studies in the
United Arab Emirates, Turkey, and Zimbabwe showed
large proportions of type IV GBS among their isolates
(Amin et al, 2002; Diedrick et al, 2010; Ekin and Gurturk,
2006; Moyo et al, 2002). In Zimbabwe it was the fourth
most common serotype, comprising 4.6% of the colonizing and invasive isolates (Moyo et al, 2002). Serotypes VII
and VIII are uncommon in Western countries.
Infected infants have low levels of type-specific antibody
to the infecting GBS serotype (Baker and Edwards, 2003).
Vaccines against the common GBS serotypes have been
shown to elicit a specific antibody response in humans
(Baker and Edwards, 2003; Kasper et al, 1996; Paoletti
et al, 1997, 2001). Sera from these vaccinated individuals protected against GBS challenge in neonatal mice,
thereby showing the potential of vaccines to prevent invasive neonatal GBS disease in infants (Paoletti et al, 1997).
The prospect of a multivalent GBS vaccine, with or without conjugated GBS surface localized proteins, makes the
study of the common GBS serotypes important because of
the possibility of serotype replacement or capsular switch
(Cieslewicz et al, 2005; Lipsitch, 1999; Maione et al, 2005;
Tettelin et al, 2005).
ESCHERICHIA COLI INFECTIONS
Historically, E. coli has been the second-most common
pathogen causing sepsis and meningitis in newborn infants.
The antigenic structure of E. coli is complex, composed
of approximately 150 somatic or cell wall O antigens, 50
flagellar H antigens, and approximately 80 capsular K
antigens. However, a limited number of K antigen E. coli
strains cause meningitis, and approximately 80% of the
strains causing meningitis and 40% of the strains causing
bacteremia or sepsis express K1 (Mulder et al, 1984). The
capsular K1 polysaccharide antigen is highly homologous
to the capsular antigen of group B Neisseria meningitidis.
Because a high percentage of women may have bacteriuria
with strains of E. coli that express the K1 antigen or are
542
PART IX Immunology and Infections
colonized with it at the time of delivery, it is surprising
that E. coli sepsis or meningitis is not more common. It
has been estimated that disease occurs in 1 in 100 to 200
infants colonized by K1 E. coli. Surveillance data from the
National Institute of Child Health and Human Development Neonatal Research Network, a consortium of 16
U.S. academic neonatal centers, revealed that in the era
of widespread implementation of antibiotic prophylaxis,
E. coli sepsis increased from 3.2 to 6.8 cases per 1000 live
births. This increase was observed in the 1998-2000 era
and persisted in 2002 to 2003. Approximately 85% of E.
coli infections in very low-birthweight (VLBW) infants
were ampicillin resistant (Stoll et al, 2002). However, most
evidence suggests that intrapartum antibiotic prophylaxis has not been associated with a concomitant adverse
impact of increasing E. coli or other non-GBS bacterial
causes. Among preterm infants, however, the incidence of
E. coli and ampicillin-resistant E. coli infections increased
significantly (Bizzarro et al, 2005). A recent retrospective
case-control study between 1997 and 2001 concluded that
exposure to intrapartum antibiotic prophylaxis therapy in
mothers did not increase the odds of invasive, early-onset
E. coli infection. In fact, among full term infants, exposure
to 4 hours or greater of intrapartum antibiotic therapy was
associated with decreased odds of early-onset E. coli infection (Schrag et al, 2006).
LISTERIA MONOCYTOGENES INFECTIONS
Listeria monocytogenes is a small, facultatively anaerobic,
gram-positive motile bacillus that produces a narrow zone
of beta hemolysis on blood agar plates, and can be confused with GBS unless a careful Gram stain, a catalase
reaction, and other tests are performed. Most disease is
due to three primary serotypes: 1a, 1b, and 4b. The last
serotype has been described in most outbreaks of listeriosis
(Posfay-Barbe and Wald, 2009). Most cases of listeriosis
appear to be food borne, including those acquired by pregnant women.
Foods that can be contaminated by L. monocytogenes
include raw vegetables such as cabbage, raw milk, fish,
poultry, processed chicken, beef, and hot dogs (Schlech,
2000). Transmission to the fetus occurs through either a
hematogenous (transplacental) route or via an ascending
infection through the birth canal. Frequently, infections
with Listeria spp. early in gestation result in abortion;
later in pregnancy, infection with Listeria spp. can result
in premature delivery of a stillborn or infected newborn.
Approximately 70% of Listeria-infected women deliver
before 35 weeks’ gestation. Illness in the mother may be
undetected because of vague influenza-like illnesses that
may not come to medical attention. In approximately half
of perinatal cases, illness in the mother has preceded delivery by 2 days to 2 weeks. At autopsy of stillborn infants or
of those who die in the perinatal period, granulomas may
be found throughout such organs as the liver and lungs,
and infection is widely disseminated, including involvement of the meninges. Treatment of Listeria spp. infection
or bacteremia during pregnancy can prevent infection in
the fetus (Kalstone, 1991). Like GBS infection, Listeria
spp. infection may have either an early-onset or late-onset
presentation. Epidemics of neonatal Listeria spp. infection
have been described after ingestion of contaminated foods
such as cheese or coleslaw. The first clearly documented
food borne (coleslaw) outbreak of listeriosis was in 1981
from the Eastern Maritimes in Canada (Schlech, 2000);
it was associated with a fatality rate of 27%. There are
reports of repeated abortions in women with colonization in the gastrointestinal tract, and cold-enrichment
cultures can be performed to try to detect fecal carriage
in such women. However, cold enrichment cultures are
inferior to selective media for Listeria spp. in isolating the
organism from various foods or stool specimens. Rapid
antigen tests based on nucleic acid amplification are not
in common use in clinical diagnostic laboratories. There
is no current vaccine for Listeria spp. infection, but preventive measures have included the surveillance programs
from the U.S. Department of Agriculture, prohibiting
the sale of contaminated meats. Between 1996 and 2006,
the incidence of Listeria spp. infections declined by 36%;
however, an outbreak of disease in 2002 related to contaminated turkey meat led to 54 illnesses, eight deaths,
and three fetal deaths in nine states (Posfay-Barbe and
Wald, 2009).
MISCELLANEOUS BACTERIAL PATHOGENS
Bacteria responsible for early-onset neonatal sepsis have
changed dramatically over time. There are regional differences in the organisms commonly responsible for
early-onset sepsis. In addition to the organisms mentioned
previously, other bacterial pathogens associated with earlyonset bacteremia or sepsis in newborn infants are Enterococcus spp., viridans group Streptococcus spp., Klebsiella spp.,
Enterobacter spp., Haemophilus influenzae (typeable and
nontypeable), S. aureus, Streptococcus pneumoniae, group A
streptococcus and other beta-hemolytic streptococci, and
coagulase-negative staphylococci.
CLINICAL SIGNS OF BACTERIAL SEPSIS
There is great variability in the clinical presentations of
infants with early-onset bacterial sepsis (Box 39-1). Most
infants exhibit respiratory distress in the first 12 hours of
life, frequently immediately after birth. In these infants,
the progression may be rapid with cardiovascular instability, shock, and death. Presentation within the first 12
hours of life suggests that the infection with pneumonia and bacteremia occurred at or near the time of birth
BOX 39-1 C
ommon Clinical Signs
of Neonatal Sepsis
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Abnormal neurologic status: irritability, lethargy, poor feeding
Abnormal temperature: hyperthermia or hypothermia
Apnea
Bleeding problems: petechiae, purpura, oozing
Cardiovascular compromise: tachycardia, hypotension, poor perfusion
Cyanosis
Gastrointestinal symptoms: abdominal distention, emesis, diarrhea
Jaundice
Respiratory distress: tachypnea, increased work of breathing, hypoxemia
Seizures
CHAPTER 39 Neonatal Bacterial Sepsis
543
Bacterial Infection Present
YES
NO
POSITIVE
True Positive
TP
False Positive
FP
Positive Predictive Value
(TP)/(TPFP)
NEGATIVE
False Negative
FN
True Negative
TN
Negative Predictive Value
(TN)/(FNTN)
Sensitivity
(TP)/(TPFN)
Specificity
(TN)/(FPTN)
Laboratory
Test Result
FIGURE 39-1 Diagram of the relationships among sensitivity, specificity, positive predictive value, and negative predictive value. FN,
Number with infection incorrectly diagnosed as healthy by the test. FP, number of healthy infants incorrectly diagnosed as infected by the test; TN,
number of healthy infants correctly diagnosed as not infected by the test; TP, number of infants with infection correctly diagnosed by the test.
or during the immediate postnatal period. Infants with
hypoxia in utero may gasp, inhaling contaminated amniotic fluid and setting the stage for early-onset pneumonia,
bacteremia, and sepsis.
The signs of early-onset infection may be subtle, with
tachypnea suggesting “wet lung disease” or may be more
overt with grunting, flaring, and subcostal and intercostal retractions. Because the signs of sepsis can be relatively
nonspecific, such as poor feeding and increased sleepiness, they can be overlooked. In newborn or intermediate
or intensive care nurseries, one must be attuned to subtle
abnormal findings in newborn infants. The clinical signs
of neonatal sepsis include hyperthermia or hypothermia,
respiratory distress, apnea, cyanosis, jaundice, hepatomegaly, abdominal distention, feeding abnormalities, and neurologic abnormalities. Autopsy findings in preterm infants
with fatal early-onset GBS infection suggested that surfactant deficiency respiratory distress syndrome was common
(Payne et al, 1988).
LABORATORY TESTING
There are many laboratory tests that have been evaluated for infants with possible sepsis, and the results must
be interpreted with caution, assessing the sensitivity and
specificity of a particular test as well its positive and negative predictive accuracy (Figure 39-1). The sensitivity
of a test is defined as the proportion of individuals with
proven or probable sepsis in whom the result is abnormal; the specificity is the proportion of healthy or noninfected infants in whom the result is normal. Ideally, a test
would have a high sensitivity and a high specificity, but
this is rarely achievable. High sensitivity is the most desirable characteristic when dealing with serious and treatable
diseases such as neonatal sepsis. Because sepsis is generally treated with antibiotic agents that have a low toxicity,
diagnostic tests do not need to have a high specificity, but
should have a high sensitivity, which will allow sepsis to
be excluded. A positive predictive accuracy is the probability that an infant with an abnormal laboratory result
is infected; a negative predictive value is the probability
that infection with a normal or negative result is free of
infection. The more sensitive the test, the greater its negative predictive value; the more specific a test, the higher its
positive predictive value.
MICROBIOLOGIC CULTURES
Previously, cultures of superficial body sites in newborn
infants (external auditory canal, gastric aspirate, umbilicus, and nasopharynx) were used to identify bacterial
pathogens when more specific blood culture results were
negative. Most centers no longer use surface cultures to
make clinical decisions regarding either the institution or
the discontinuation of antibiotic therapy, because surface
cultures are of limited value in predicting the etiology
of bacterial sepsis in newborn infants (Evans et al, 1988;
Shenoy et al, 2000). Examination of gastric aspirates by
Gram stain as a screening mechanism has also lost favor,
as has examination for PMNs. Although the presence of
increased numbers of PMNs may represent amnionitis and
an inflammatory response, these cells may reflect a maternal origin and have no specificity in predicting bacterial
sepsis for the infants (Vasan et al, 1977).
Blood Cultures
The gold standard for detection of bacteremia in newborn
infants with suspected sepsis is a positive blood culture.
With the introduction of newer blood culture detection
instruments that are semiautomated and examined for
the presence of growth by CO2 production by the internal computer of the instrument every minute, the sensitivity of detecting positive blood cultures has increased.
Another variable that influences the sensitivity of detection of bacteremia is the volume of blood obtained and
placed in the culture bottles. Ideally 1 to 3 mL of blood
from infants should be obtained, but this not always possible in very small infants. Most positive blood cultures are
detected within 24 to 48 hours using the new technology
(Garcia-Prats et al, 2000). However, the use of intrapartum antibiotics for prophylaxis in mothers with either GBS
colonization or suspected amnionitis on account of any
cause can reduce the ability to detect bacteremia in newborn infants. In a term infant who was asymptomatic at the
initiation of antibiotic therapy, it may be reasonable to stop
antibiotic administration if the blood cultures remain negative after 48 hours. However, the decision to discontinue
treatment with antibiotics should include the assessment of
results of other laboratory tests used for sepsis screens, and
should not solely rely on a negative blood culture. When
544
PART IX Immunology and Infections
TABLE 39-2 Predictive Values of Components of the White Blood Cell Count and of C-Reactive Protein
Predictive Value (%)
Laboratory Result
Sensitivity (%)
Specificity (%)
Positive
Negative
ANC <1750 cells/mm3
38-96
61-92
20-77
96-99
ANC <10% (≈5580 cells/mm3 for term)
48
73
4
98
I/T ratio ≥ 0.2
90-100
30-78
11-51
98
I/T ratio ≥ 0.3
35
89
7
98
CRP > 1mg/dL
70-93
78-94
27
100
Adapted from Gerdes JS: Diagnosis and management of bacterial infections in the neonate, Pediatr Clin North Am 51:939-959, 2004.
ANC, Absolute neutrophil count; CRP, C-reactive protein; I/T, immature to total neutrophils (ratio), calculated by the number of immature neutrophils (bands, metamyelocytes, and
myelocytes) divided by the total number of neutrophils (immature plus mature).
the suspicion of sepsis is high, clinicians should consider
continuing antibiotic therapy for a complete course despite
negative blood cultures (Ottolini et al, 2003).
Urine Cultures
The frequency of positive urine cultures in infants with
early-onset sepsis is relatively low, and it is rare to find bacteriuria in infants with negative blood culture results (DiGeronimo, 1992). Infants with late-onset sepsis tend to have
a higher rate of positive urine cultures (Visser and Hall,
1979). In the era of widespread intrapartum antibiotic
prophylaxis in the mother, positive urine cultures may be
obscured because of excretion of antibiotics in the urine of
newborn infants. When pyelonephritis is found in newborn
infants, it likely represents metastatic seeding of the kidney
during a bout of bacteremia. In the first 72 hours of life,
because the yield from urine cultures is low, it is not generally recommended to obtain these specimens. However, in
the older newborn infant, a urine sample collected by an
aseptic technique (urinary catheter or suprapubic bladder
aspiration) is an important part of the sepsis workup.
Cerebrospinal Fluid
Lumbar punctures are deferred in infants with any instability or uncorrected bleeding disorders. The details of examination of cerebrospinal fluid and the diagnostic approach
for examining cerebrospinal fluid will be discussed under
the section on bacterial meningitis.
WHITE BLOOD CELL COUNT AND
NEUTROPHIL INDICES
Normal white blood cell (WBC) counts range from 9000
to 30,000 cells/mm3 at the time of birth, and differences
in the site of sampling can affect these values. The absolute neutrophil count (ANC), the absolute band count of
immature neutrophils, and the ratio of immature neutrophils to total neutrophils (I/T) are regarded as more useful than total leukocyte counts in the diagnosis of neonatal
sepsis.
The lower limit or total neutrophil count rises to 7200
cells/mm3 by 12 hours of age, and then declines to approximately 1720 cells/mm3 by 72 hours of age (Manroe et al,
1979; Schmutz et al, 2008). Postnatally the absolute band
count also undergoes similar changes, with peak values of
1400 cells/mm3 at 12 hours of age, and then declines. In contrast, the I/T ratio is maximum at birth and then declines to
a value of 0.12 beyond 72 hours of age (Manroe et al, 1979;
Schelonka et al, 1994). However, in VLBW infants there
is a greater reference range for the total neutrophil counts
(Manroe et al, 1979; Mouzinho et al, 1994). There are no
significant differences in I/T ratio or absolute immature
neutrophil counts in VLBW infants. There is considerable
overlap of the cutoff values of the ANC, absolute band count,
and I/T ratio between healthy and infected newborns.
There are a number of clinical conditions that affect the
total neutrophil count. Prolonged crying, meconium aspiration syndrome, maternal fever, and asphyxia are all associated with an increase in the total neutrophil count, and
there may be an increase in the total immature neutrophil
forms, as well as an increased I/T ratio. Maternal hypertension is associated with a decrease in total neutrophils.
At high altitudes, a higher upper limit of neutrophil values
occurs (Schmutz et al, 2008).
In approximately two thirds of infants with sepsis, the
total neutrophil count is abnormal. Neutropenia is the best
predictor of sepsis, whereas neutrophilia does not correlate well. The absolute neutrophil band count is not a sensitive marker of sepsis, but has a relatively good predictive
value and specificity. The I/T ratio is considered to have
the best sensitivity of all of the neutrophil indices (Table
39-2) (Gerdes, 2004).
PLATELET COUNTS
Approximately 25% to 30% of infants exhibit thrombocytopenia at the time of diagnosis of sepsis, and this frequency
increases during the course of infection. Accelerated platelet destruction and possibly depressed production caused
by bacterial products on the bone marrow are the underlying mechanisms for thrombocytopenia in infected infants.
Disseminated intravascular coagulation may be seen in
some infants with severe sepsis.
ACUTE PHASE REACTANTS AND
ERYTHROCYTE SEDIMENTATION RATE
A number of acute phase reactants have been studied to
help identify infants with likely sepsis. Most biochemical
markers currently in use are derived from components of
CHAPTER 39 Neonatal Bacterial Sepsis
the complex inflammatory response to an invading pathogen. These markers have been studied during the past
20 years and continue to be under investigation. Of issue
is the availability of tests for these inflammatory mediators
when investigating an infant with possible sepsis, and the
value of these tests in assisting with the early diagnosis of
sepsis. It is known that some proinflammatory cytokines
peak rapidly, within 1 to 4 hours after a sepsis stimulus
(Lam and Ng, 2008). C-reactive protein (CRP) rises to a
maximum at 12 to 24 hours, and procalcitonin (PCT) rises
at 4 hours, peaks at 6 hours, and plateaus 8 to 24 hours after
a stimulus. CRP is induced by proinflammatory cytokines.
Among the early markers are the proinflammatory mediators, such as IL-1β, IL-6, the chemokine IL-8, TNF-α,
and interferon gamma; these activate host defenses against
bacterial and other infecting agents, whereas antiinflammatory mediators such as IL-4, IL-10, and transforming
growth factor β1 (TGF-β1) are important in regulating
and limiting the inflammatory response, thus preventing
an excessive reaction that could lead to organ damage and
tissue cell death (Mehr and Doyle, 2000). Survival should
be considered a fine balance between the proinflammatory
cytokines and the antiinflammatory mediators. Because of
the limitations of other assessment markers of potentially
infected neonates, such as WBC counts and band counts,
the advantages of having a surrogate marker readily available are apparent.
CRP is probably the most studied acute phase reactant
in neonatal sepsis (Benitz et al, 1998; Jaye and Waites,
1997; Lam and Ng, 2008; Weitkamp and Aschner, 2005).
Monitoring of CRP levels has been widely promulgated as
a way to diagnose neonatal infection and to adjust the duration of antibiotic therapy in infants with suspected versus
proven sepsis (Benitz et al, 1998; Ehl et al, 1997; Gerdes,
2004; Philip and Mills, 2000). Depending on the laboratory, a CRP value of 1 to 8 mg/dL is considered the upper
limit of normal; it is important to know the different cutoff
values in the laboratory supporting one’s neonatal units.
Theoretically, results can be available within 30 minutes.
CRP is produced by the liver in response to stimulation
by the proinflammatory cytokine IL-6, which is produced
by both T and B cells (Lam and Ng, 2008; Weitkamp and
Aschner, 2005). Because exposure of the host to bacterial
products results in a substantial and rapid increase in IL-6
concentrations, it appears that IL-6 is potentially a more
useful marker than CRP during the early phase of an infection. In one study, the IL-6 concentration had a sensitivity of 89% versus 60% for CRP at the onset of clinical
suspicion of a neonatal infection (Lam and Ng, 2008).
The negative predictive values of IL-6 are much higher
than those for CRP (Lam and Ng, 2008), and this was also
found in cord blood IL-6 levels, where the sensitivity of
detection was high. This finding can be useful in deciding
which infants do not require antibiotic therapy and those
in whom antibiotics can be discontinued after a relatively
short course.
In a recent study, the high-sensitivity CRP (hsCRP)
measurement has been shown to provide increased sensitivity for detecting neonatal infection (Edgar et al, 2010).
Not all diagnostic laboratories can provide hsCRP in a
timely fashion. In addition, the optimum diagnostic cutoff
levels for CRP and hsCRP are debatable.
545
A recent study examined the combination of hsCRP,
serum soluble intercellular adhesion molecule 1 (slSAM1), soluble E-selectin (sE-selectin), and serum amyloid A,
individually and in combination, for the diagnosis of sepsis in a neonatal intensive care unit (Edgar et al, 2010).
In this study, all four measurements had some diagnostic
value for neonatal infection; however, s1SAM-1, hsCRP,
and sE-selectin demonstrated the highest negative predictive value individually (sISAM, 84%; hsCRP, 79%; and
sE-selectin, 74%) (Edgar et al, 2010). Use of a combination of these measurements enhanced the diagnostic value,
with sensitivities of 90.3% and a negative predictive value
of 91.3% (Edgar et al, 2010). However, the application
of this set of diagnostic markers is not available for most
facilities, and more investigative work is needed to confirm
their role in excluding early-onset infection.
In another study, early markers (IL-6, TNF-α, IL-8,
interferon gamma, CRP, IL-18, the antiinflammatory
cytokine IL-10, the I/T ratio, and PCT, a later marker of
infection) were studied for use in detection of early-onset
sepsis in 123 newborn infants (Bender et al, 2008). This
study concluded that IL-6 combined with PCT was a fair
measure of evaluating early-onset infection, and that the
traditional I/T ratio was almost as efficient as IL-6. Combining an early marker such as IL-6 and an I/T ratio may
reduce the number of diagnostic tests and nonconclusive
values. The combined value of IL-6 and PCT at the first
blood drawing had a sensitivity of 71% and specificity of
88% (Bender et al, 2008).
There are many studies of procalcitonin in the literature, and most have concluded that procalcitonin is superior to CRP levels in the early diagnosis of neonatal sepsis
(Lam and Ng, 2008). PCT is the precursor of calcitonin,
normally synthesized in the C-cells of the thyroid gland.
PCT is induced by systemic inflammation and bacterial
sepsis and is produced by such cells as hepatocytes, nephrons, and monocytes. The physiologic function of PCT is
unknown. In bacterial infections, plasma PCT concentrations increase from 0.001 to 0.01 ng/mL (baseline range) to
values ranging from 1 to 1000 ng/mL. PCT concentrations
rise much faster than CRP (6 to 8 hours versus 48 hours
for maximum levels). In healthy newborn infants, plasma
PCT concentrations increase gradually after birth, reaching peak levels at approximately 24 hours of age (range, 0.1
to 20 ng/mL) and then decrease to normal values less than
0.5 ng/mL by 48 to 72 hours of age (Stocker et al, 2010).
Various studies on the use of PCT as a marker of neonatal sepsis have yielded contradictory results regarding
its application to clinical decision making for both diagnosis and adjustment of the length of antibiotic therapy.
In a recent study of 121 newborn infants with suspected
early-onset sepsis, serial PCT determinations allowed
shortening of the duration of antibiotic therapy (Stocker
et al, 2010). However, there are currently few institutions
or intensive care units that are applying PCT as a regular
measurement for diagnosis or for adjustment of length of
antibiotic therapy. Larger studies are needed to determine
the true value of PCT in diagnosis and therapy.
To date, studies on the role of various cytokine determinations in assisting with diagnosis and treatment of
early-onset sepsis are intriguing, but have not translated
to widespread use. Other molecular technologies are also
546
PART IX Immunology and Infections
being studied to diagnose neonatal sepsis by the rapid
identification and differentiation of gram-negative and
gram-positive bacterial bloodstream infections. PCR,
using universal bacterial primers, targets conserved
regions of the 16SrRNA gene common to all bacteria, but
not found in other organisms (Dutta et al, 2009). In one
study, universal primer PCR was performed in newborn
infants with clinically suspected sepsis. PCR was performed before starting antibiotic therapy, and repeated
at 12, 24, and 48 hours after starting drug therapy (Dutta
et al, 2009). The sensitivity, specificity, and positive and
negative predictive values of universal primer PCR were
96.2%, 96.3%, 87.7%, and 98.8% respectively. Results
of testing in two patients were blood culture positive,
but 0-hour PCR negative, and results in 7 patients were
0-hour PCR positive, but blood culture result was negative. Of the patients with a result of 0-hour PCR positive,
7 remained positive at 12 hours but none remained positive at 24 and 48 hours after starting antibiotic therapy.
Although universal bacterial primer PCR may be a useful
test for diagnosing an early episode of culture-proven sepsis, it cannot be used for diagnosis if the patient has been
exposed to 12 hours or more of antibiotic therapy (Dutta
et al, 2009). Much larger studies are certainly required
before this assay can be recommended for routine clinical
use in newborn infants suspected of sepsis.
PREVENTION
INTRAPARTUM ANTIBIOTIC PROPHYLAXIS
There were initial concerns about adverse effects of implementing the CDC consensus strategies, but these have
proved unwarranted. The primary risks considered were
maternal anaphylaxis from administered antibiotics, and
these were unfounded. The possibility of the emergence
of infections in mothers and infants caused by antibiotic
resistant organisms (e.g., E. coli) was addressed in part
by studies conducted by the Neonatal Network of the
National Institute of Child Health and Human Development. In 2002, they reported a change in the pathogens
causing early-onset sepsis, with an increase in sepsis caused
by E. coli from 3.2 to 6.8 per 1000 live births (Stoll et al,
2002). Most of the E. coli isolates were resistant to ampicillin. Mothers of infants with ampicillin-resistant E. coli
infections were more likely to have received intrapartum
antibiotic prophylaxis than were those with ampicillin-sensitive strains (Stoll et al, 2002). In another study comparing a period of no prophylaxis to periods of risk-based and
universal screening-based prophylaxis, no change in the
incidence of infection with ampicillin-resistant organisms
was observed overall or among VLBW infants (Puopolo
and Eichenwald, 2010). However, an increased proportion
of infections was caused by ampicillin-resistant organisms.
Mothers of infants with ampicillin-resistant infections
were also more likely to have been treated with ampicillin
(Chen et al, 2005). A 10-year study of the effect of intrapartum antibiotic prophylaxis on GBS and E. coli sepsis in
Australasia revealed a steady decline in early-onset GBS
infection, but also a trend to decreasing early-onset E. coli
sepsis in all infants and a stable rate for this infection in
VLBW infants (Daley et al, 2004).
INTRAPARTUM MANAGEMENT
OF PARTURIENTS
The universal GBS screening strategy recommended by
the CDC is done at 35 to 37 weeks’ gestation. Antepartum antibiotic therapy is recommended for women with
a previous infant with invasive GBS disease, a positive
GBS screening culture result during pregnancy, unknown
GBS status (culture either not performed or incomplete or
results unknown), and any of the following features: delivery before 37 weeks’ gestation, rupture of chorioamniotic
membranes for greater than 18 hours, intrapartum temperature of 100.4° F (38° C) or higher, and GBS bacteriuria during pregnancy.
Women who are not allergic to penicillin can be given
penicillin G (5,000,000 units as a loading IV dose followed by 2.5 million units every 4 hours until delivery) or
ampicillin (2g IV as a loading dose followed by 1g every
4 hours until delivery). For women who are allergic to
penicillin, the recommendation is to determine, ideally
during prenatal care, whether the patient is at high risk
for anaphylaxis (i.e., history of immediate hypersensitivity
reactions). Women who are not at high risk for anaphylaxis can receive cefazolin (2g IV and 1g IV every 8 hours
until delivery). Women who are at high risk for anaphylaxis whose GBS isolate is not resistant to clindamycin or
erythromycin can receive clindamycin (900 mg IV every
8 hours until delivery) or erythromycin (500 mg IV every
6 hours until delivery) (Schrag et al, 2002). Women with
clindamycin-erythromycin–resistant GBS isolates can
receive vancomycin (1g IV every 12 hours until delivery).
INTRAVENOUS IMMUNE GLOBULIN FOR
PREVENTION OF EARLY-ONSET SEPSIS
Premature infants are susceptible to early-onset sepsis
because of diminished transplacental transfer of immunoglobulins and decreased synthesis of immunoglobulin (Ig)
G. An intravenous infusion of immunoglobulin (IVIg) will
increase the typically low levels of serum immunoglobulin
in preterm infants, and it has been proposed to improve
immune function that may lead to improved clinical outcome (Wynn et al, 2009). Clinical trials examining the
possible benefit of IV-Ig began over 15 years ago (Chirico
et al, 1987; Conway et al, 1990; Haque et al, 1986). The
very premature infants are the group most likely to receive
and potentially benefit from these adjuvant treatments
and preventative interventions. A Cochrane metaanalysis
showed a reduction in mortality of newborn infants treated
with IV-Ig for what were subsequently proved to be bacterial infections (relative risk, 0.55; 95% confidence interval,
0.31 to 0.98) (Ohlsson and Lacy, 2010). However, a review
of the studies included in the metaanalysis revealed the
inclusion of few very premature infants. A large international, placebo-controlled, double-blind, randomized clinical trial has recently been undertaken in approximately
5000 newborn infants to examine the benefit of polyclonal
IV-Ig treatment for proven or suspected sepsis (Wynn
et al, 2010). However, because randomization will allow
the inclusion of neonates of all gestational and postnatal
ages with suspected or proven infection, the subgroup
sample sizes may leave the issue of clinical efficacy for
CHAPTER 39 Neonatal Bacterial Sepsis
early- and late-onset sepsis in the different gestational
age groups unanswered. A metaanalysis that included
almost 4000 preterm (<37 weeks’ gestation) or low birthweight (<2500 g) infants showed a modest reduction in the
development of sepsis after IV-Ig prophylaxis in preterm
neonates (Wynn et al, 2010). Currently available data do
not support the suggested immunologic enhancements
expected for IV-Ig in very premature neonates. There is
continued interest in the potential of IV-Ig as treatment
or prophylaxis for infection in very premature infants, but
large trials are needed to provide convincing data.
Other immunomodulating factors such as granulocyte
colony-stimulating factor (G-CSF) have been studied to
both prevent and treat neonatal sepsis. An earlier metaanalysis showed that G-CSF administration improved mortality in all neonates and in the subgroups of neutropenic and
preterm infants (Bernstein et al, 2001). A recent Cochrane
metaanalysis concluded that prophylaxis with G-CSF does
not significantly reduce mortality in all infants, although
in premature infants with neutropenia (ANC < 1750) mortality may be improved (Carr et al, 2009). It is possible
that combination therapies using IV-Ig and other immunomodulating factors, such as granulocyte-macrophage
colony-stimulating factor (GM-CSF), G-CSF, and complement-containing blood products, may lead to improved
immune function in these highly susceptible infants; however, these studies are not currently being conducted.
DIAGNOSTIC APPROACH TO NEONATES
WITH SUSPECTED SEPSIS
Obviously, all symptomatic newborn infants must be carefully evaluated for the possibility of bacterial sepsis and
treated with antibiotics, if necessary. Although the presence of various risk factors should increase the suspicion
of sepsis, the absence of risk factors in a symptomatic
infant cannot be dismissed. In adjusting to postnatal life,
some infants exhibit abnormal signs transiently, such as
tachypnea, before becoming asymptomatic. However, any
infant who has other findings or is still symptomatic with
only one finding by 6 hours of life should have a diagnostic evaluation performed with a complete blood cell count
(CBC) and differential, a blood culture, and as appropriate, a lumbar puncture and a chest radiograph. Antibiotic
therapy can be stopped when the physical findings are normal, the clinical suspicion of sepsis is low, and the screening results for sepsis, including the blood culture, remain
negative. If the blood culture result is positive, the lumbar
puncture findings abnormal, or there are clinical signs of
sepsis, then the infant should be treated with an appropriate course of antibiotics (Figure 39-2).
Management of the asymptomatic infant with risk factors for sepsis is more controversial. Escobar et al (2000)
retrospectively reviewed 2785 newborns with birthweight
greater than 2000 g and who were evaluated for sepsis
after birth. Asymptomatic status was associated with a significantly decreased odds ratio for infection. Low absolute
neutrophil count and meconium-stained amniotic fluid
were associated with an increased risk of infection. The
highest infection rates occurred in infants whose mother
had a clinical diagnosis of “definite” chorioamnionitis,
but chorioamnionitis was significantly associated with an
547
increased risk of neonatal infection only in infants whose
mothers had not received intrapartum antibiotics (Escobar
et al, 2000).
Current practice management for the infant with no
signs or symptoms of sepsis, but with risk factors such as
maternal chorioamnionitis or incomplete GBS maternal
prophylaxis, includes performing a limited sepsis evaluation (CBC with differential and blood culture). If the WBC
count, absolute neutrophil count, and the I/T ratio are all
normal, then the infant is usually observed in the hospital
and discharged at approximately 48 hours. If the screening
tests are abnormal, the infant undergoes a complete sepsis
evaluation with subsequent antibiotic therapy initiated.
Recent CDC and American Academy of Pediatrics
guidelines for the prevention of early-onset neonatal GBS
sepsis have a slightly different approach to management of
the at-risk asymptomatic infant (see Figure 39-2; Verani
and Schrag, 2010). If there are signs of sepsis in the newborn, then a full sepsis workup and antibiotic therapy are
recommended. If there is maternal chorioamnionitis, a
limited sepsis evaluation and antibiotic are recommended.
For infants whose mother received greater than 4 hours
of antibiotic therapy, it is recommended that the infant be
observed in the hospital for approximately 48 hours. For
infants whose mother did not receive more than 4 hours
of antibiotic prophylaxis, a 48 hour in-hospital observation
is recommended. If the infant is less than 37 weeks’ gestation, or the duration of rupture of membranes is greater
than or equal to 18 hours, then a limited evaluation and
observation in the hospital for approximately 48 hours are
recommended.
TREATMENT
ANTIMICROBIAL THERAPY
The choice of antibiotic administration for an infant with
suspected early-onset sepsis depends on the predominant
bacterial pathogens and the antibiotic susceptibility profiles for the microorganisms causing early-onset disease
in a particular geographic region. Any decision to discontinue antimicrobial therapy should be based on the level
of suspicion for sepsis at the time treatment was begun,
the culture results, laboratory test results, and the clinical
behavior and course of the infant. If sepsis is highly suspected in an infant, antibiotics should be considered for a
full course even if the culture results are negative.
Empiric therapy for early-onset sepsis generally consists of combinations of antibiotics effective against grampositive (e.g., GBS, L. monocytogenes) and gram-negative
pathogens (e.g., E. coli). The two most commonly used
combinations are (1) ampicillin with an aminoglycoside, usually gentamicin, and (2) ampicillin with a third-
generation cephalosporin, usually cefotaxime. Cefotaxime
has minimal toxicity and is well tolerated by newborn
infants. However, the third-generation cephalosporins, as
well as vancomycin, a glycopeptide antibiotic, have been
associated with the development of vancomycin-resistant
enterococci and, in the case of cephalosporins, with the
induction of various β-lactamase producing gram-negative
bacteria, including extended-spectrum β-lactamase producing organisms (Bryan et al, 1985). These latter bacteria
548
PART IX Immunology and Infections
Signs of neonatal sepsis?
yes
Full diagnostic evaluation*
Antibiotic therapy†
yes
Limited evaluation¶
Antibiotic therapy†¶
no
Routine clinical care††
yes
Observation for 48 hours††§§
yes
Observation for 48 hours††
yes
Limited evaluation¶
Observation for 48 hours¶¶
no
Maternal chorioamnionitis?§
no
GBS prophylaxis indicated for
mother?**
yes
Mother received 4 hours of
penicillin, ampicillin or cefazolin IV?
no
37 weeks AND duration of
membrane rupture <18 hours?
no
Either <37 weeks OR duration of
membrane rupture 18 hours?
* Includes CBC with differential, platelets, blood culture, chest radiograph (if respiratory abnormalities are
present), and LP (if patient stable enough to tolerate procedure and sepsis is suspected).
†
Antibiotic therapy should be directed towards the most common causes of neonatal sepsis including GBS
and other organisms (including gram negative pathgoens), and should take into account local antibiotic
resistance patterns.
§
Consultation with obstetric providers is important to determine the level of clinical suspicion for chorloamnionitis. Chorioamnionitis is diagnosed clinically and some of the signs are non-spedific.
¶
Includes blood culture (at birth), and CBC with differential and platelets. Some experts recommend a CBC
with differential and platelets at 6-12 hours of age.
** GBS prophylaxis indicated if one or more of the following: (1) mother GBS positive at 35-37 weeks’
gestation, (2) GBS status unknown with one or more intrapartum risk factors including <37 weeks’ gestation,
ROM 18 hours or T 100.4F (38.0C), (3) GBS bacteriuria during current pregnancy, (4) history of a
previous infant with GBS disease.
††
If signs of sepsis develop, a full diagnostic evaluation should be done and antibiotic therapy initiated.
§§
If 37 weeks’ gestation, observation may occur at home after 24 hours if there is a knowledgeable
observer and ready access to medical care.
¶¶
Some experts recommend a CBC with differential and platelets at 6-12 hours of age.
FIGURE 39-2 Algorithm for recommended management of newborns at risk for group B streptococcal disease. (Adapted from Verani JR, Schrag SS:
Group B streptococcal disease in infants: progress in prevention and continued challenges, Clin Perinatol 37:375-392, 2010.)
are resistant to all β-lactam antibiotics and frequently
are resistant to other antibiotics, but not to meropenem.
Another disadvantage of the cephalosporin antibiotics is
the lack of effectiveness against enterococci or L. monocytogenes. L. monocytogenes is usually treated with ampicillin and an aminoglycoside until the blood culture result is
negative and the infant has shown an improved outcome.
In infants with bacteremia and sepsis caused by GBS, gentamicin is frequently combined with ampicillin or penicillin, although there are no data to suggest that the addition
of the aminoglycoside improves outcome. However, it
is common practice to use the combination of these two
drugs during the first few days of therapy and then to continue the full course of therapy with ampicillin or penicillin alone.
When the likelihood of infection is very low, the antibiotic therapy should be stopped. In most hospitals using
modern blood culture instrumentation, 48 hours should
be considered sufficient to determine whether a blood culture result is negative, assuming that no antibiotics were
being given when the culture was obtained (Garcia-Prats
et al, 2000). Blood culture bottles with antimicrobial binding resins are in common use in microbiology laboratories,
enhancing the ability to demonstrate positive blood cultures. Infants with proven bacteremia, but without meningitis, are commonly treated for 7 to 10 days. The use
of antibiotics with nephrotoxicity (i.e., aminoglycosides)
should be monitored using appropriate drug levels.
IMMUNOLOGICAL THERAPIES FOR
TREATMENT OF EARLY-ONSET SEPSIS
Various adjunctive therapies have been proposed to
improve the immune status of the infant in an attempt to
mitigate the mortality of neonatal sepsis. These included
IV-Ig, granulocyte transfusions and G-CSF or GM-CSF
treatment. Overall, there are insufficient data to recommend the routine use of any of these therapies for the
treatment of sepsis in newborn infants. However, mortality in cases of subsequently proven infection was reduced
in the IV-Ig–treated infants, and a large randomized controlled trial of IV-Ig use was recently completed (Ohlsson
and Lacy, 2010). Clinical studies also did not show that
granulocyte transfusions provided a significant benefit
to neonates with culture-proven early-onset infections
(Vamvakas and Pineda, 1996). However, in infants with
CHAPTER 39 Neonatal Bacterial Sepsis
overwhelming sepsis, severe neutropenia, and depletion of
bone marrow neutrophil stores, treatment with granulocyte transfusions improved survival (Cairo et al, 1992).
Using GM-CSF and G-CSF to enhance the quantity
and quality of neutrophils has also been studied in human
neonates (Carr et al, 2009). Although these agents have
induced circulating numbers of neutrophils and appeared
to be relatively safe, they have not significantly reduced
neonatal sepsis mortality (Wynn et al, 2009). In two metaanalyses, subgroup analysis showed a significant reduction
in mortality in the group of infants with systemic infection
and neutropenia (Bernstein et al, 2001; Carr et al, 2009).
Future studies may prove that selective use of these therapies is beneficial in specific subgroups of septic infants.
NEONATAL BACTERIAL MENINGITIS
Neonatal bacterial meningitis is ominous because of its
mortality and morbidity, and it is associated with the
same pathogens that cause bacterial sepsis, with GBS
and E. coli accounting for approximately 70% of all cases,
and L. monocytogenes accounting for an additional 5% in
the first week of life. On occasion, it is possible to isolate
Streptococcus pneumoniae and Haemophilus influenzae, and
in infants who are older than 1 week residing in neonatal
intensive care units, coagulase-negative staphylococci are
the most common isolates. The underlying pathogenesis
of bacterial meningitis is a seeding of the meninges during
a bacteremic phase in the infant. Studies in neonatal rats
have shown that high-grade bacteremia is more likely to
lead to bacterial meningitis than lower grade bacteremia.
GBS meningitis (with a mortality approaching 30% and
a morbidity of 50%) usually presents as late-onset disease,
and the most common GBS serotype identified is III (Levent et al, 2010). In a recent paper by Ansong et al (2009),
GBS meningitis complicated 22 in 145 (15%) episodes of
early-onset GBS sepsis and 13 in 23 (57%) of episodes of
late-onset GBS sepsis. GBS meningitis can occur in the
presence of negative blood cultures, and 20% of infants in
this study had negative blood cultures (Ansong et al, 2009).
Approximately 80% of all serotypes of E. coli that cause
meningitis in newborn infants possess the K1 or capsular
antigen. The K1 capsular polysaccharide antigen is considered one of the primary virulence factors of this capsular type of E. coli, because antibody against K1 antigen
has been shown to be protective in neonatal rat models
of infection. Mortality rates for neonatal E. coli meningitis
vary from 20% to 30% in some centers and 50% to 60%
in others (Dodge, 1994).
549
instability of temperature regulation, vomiting, respiratory
distress, and apnea. A bulging fontanel may be seen, but
this is usually a late manifestation. Seizures are frequently
observed and can be caused by either direct central nervous system inflammation or by metabolic abnormalities
such as hypoglycemia or hyponatremia.
DIAGNOSIS
The gold standard for diagnosis of meningitis is the analysis of the cerebrospinal fluid, including the WBC count,
glucose and protein levels, Gram stain, and culture. The
interpretation of cerebrospinal fluid cell counts in newborn infants may be difficult (Garges et al, 2006; Greenberg et al, 2008; Polk and Steele, 1987; Unhanand et al,
1993). During the first week of life, the cerebrospinal fluid
WBC count slowly decreases in full-term infants, but may
remain high or even increase in premature infants. There
is no change in cerebrospinal fluid WBC counts or protein content with gestational age, but there is a significant
decrease with postnatal age (Mhanna et al, 2008). The
cerebrospinal fluid cell counts, protein, and glucose concentrations from healthy infants may overlap with those
from infants with meningitis, and from 1% to 10% of
infants with proven meningitis have normal results on a
cerebrospinal fluid analysis (Hristeva et al, 1993; Garges
et al, 2006; Greenberg et al, 2008). Finally, approximately
30% of all infants with positive results of cerebrospinal
fluid cultures for bacteria have negative blood culture
results (Garges et al, 2006; Wiswell et al, 1995).
A Gram stain of cerebrospinal fluid must be examined
carefully for every infant with suspected meningitis. The
stains for approximately 20% of newborns with proven
meningitis are reported as showing “no bacteria seen.”
Although an increase is expected in neutrophils with bacterial meningitis, one may see a predominance of lymphocytes within a conversion to PMNs. With L. monocytogenes,
a mononuclear cellular response is found in examination of
the cerebrospinal fluid. In clinical care units, it is routine to
repeat the cerebrospinal fluid examination and culture 2 to 3
days after the initiation of antibiotic therapy. This examination is especially important if the patient has not responded
clinically and is experiencing seizures or continued fever.
At times it is difficult to eradicate the organism from the
cerebrospinal fluid, and consideration can be given to
examining the inhibitory and bactericidal concentrations in
cerebrospinal fluid. It is especially important to repeat the
cerebrospinal fluid examination before stopping antibiotics
in patients with more complicated courses, and for enteric
gram-negative bacterial meningitis.
PATHOLOGY AND CLINICAL
MANIFESTATIONS
THERAPY
At autopsy, infants who die of meningitis have purulent
exudates of the meninges and the surfaces of the ventricles
associated with inflammation. Historically, hydrocephalus
and a noninfectious encephalopathy were demonstrated
in approximately 50% of infants who died of bacterial
meningitis.
The signs and symptoms of neonatal meningitis are not
easy to distinguish from those of sepsis. The most common presenting symptoms are lethargy, feeding problems,
Infants with bacterial meningitis are frequently ill and
should be monitored in intensive care units when the critical needs can be met with aggressive management. These
patients may require mechanical ventilation, complex fluid
management to attenuate the effects of cerebral edema
and effects of secretion of inappropriate anti-diuretic hormone (ADH), seizure control, vasopressor support, and
cardiopulmonary monitoring. The choice of appropriate
antibiotic therapy is based on several factors, including
550
PART IX Immunology and Infections
the achievable cerebrospinal fluid levels of drugs that have
in vitro efficacy against the microorganism. In the case of
gram-positive bacteria, the use of penicillin and ampicillin
will achieve 10- to 100-fold higher concentrations than the
minimal inhibitory concentrations needed to inhibit the
bacteria, and there is rapid sterilization of the cerebrospinal fluid. In contrast, aminoglycosides, such as gentamicin
and tobramycin, achieve only 40% of peak serum levels
and may not achieve minimal inhibitory concentrations
more than those equal to or slightly greater than found in
vitro for gram-negative bacteria.
In many intensive care units, ampicillin and gentamicin are recommended for the initial therapy for neonatal
meningitis. An alternative regimen of ampicillin and cefotaxime can also be used, recognizing the potential for the
introduction of cephalosporin-resistant gram-negative
isolates in the unit. Although used in the past, neither
intrathecal nor intraventricular administration of antibiotics has been found to improve the morbidity or mortality
of gram-negative meningitis (Shah et al, 2008). Once the
microorganism has been identified and the antibiotic susceptibility results are available, either a single drug or a
combination of drugs found to be effective in vitro should
be used. Usually penicillin or ampicillin is used for GBS
meningitis; in the first few days of therapy, dual therapy
with an aminoglycoside may be used. For L. monocytogenes, it is common to treat with ampicillin with or without gentamicin. Ampicillin or cefotaxime with or without
an aminoglycoside can be used for infection with gramnegative enteric bacteria. The precise length of antibiotic
therapy depends on the rapidity of response and sterilization of cerebrospinal fluid. In general, continue therapy
for approximately 2 weeks after sterilization of the cerebrospinal fluid, or a minimum of 2 weeks for gram-positive
meningitis and a minimum of 3 weeks for gram-negative
meningitis. In difficult situations, therapy may be required
for as long as 4 to 6 weeks.
It may be prudent to repeat a cerebrospinal fluid examination and culture after initiation of therapy, especially if
the clinical response is less than satisfactory. If organisms
are seen on gram-stained smears of the fluid, modification
of the therapeutic regimen should be considered. In general, approximately 3 days or more are required for an antibiotic regimen to sterilize the cerebrospinal fluid in infants
with gram-negative meningitis. In infants with gram-positive meningitis, sterilization is usually seen within 36 to
48 hours. Neuroimaging should be considered to exclude
parameningeal foci and abscess formation and to assist in
assessing the infant’s prognosis.
PROGNOSIS
Complications from neonatal meningitis include brain
abscess, communicating or noncommunicating hydrocephalus, subdural effusions, ventriculitis, deafness, and
blindness. Generally the severity of complications is
related to severity of the disease during the early neonatal period. It is imperative to follow hearing competency
and to examine these infants for prolonged periods after
recovery. The infant who has experienced meningitis may
appear relatively healthy at the time of discharge, and only
after careful follow-up do perceptual difficulties, reading problems, or signs of brain damage become apparent.
Approximately 40% to 50% of survivors have some evidence of neurologic damage, with severe damage being
obvious in 11%. Infants who survive neonatal meningitis
should have regular audiology, language, and neurologic
evaluations until they enter school (Edwards et al, 1985;
Stevens et al, 2003).
SUGGESTED READINGS
Ansong AK, Smith PB, Benjamin DK, et al: Group B streptococcal meningitis:
cerebrospinal fluid parameters in the era of intrapartum antibiotic prophylaxis,
Early Hum Dev 85:S5-S7, 2009.
Bender L, Thaarup J, Varming K, et al: Early and late markers for the detection of
early-onset neonatal sepsis, Dan Med Bull 55:219-223, 2008.
Castor ML, Whitney CG, Como-Sabetti K, et al: Antibiotic resistance patterns in
invasive group B streptococcal isolates, Infect Dis Obstet Gynecol 727505:2008,
2008.
Centers for Disease Control and Prevention: Trends in perinatal group B streptococcal disease: United States, 2000-2006, MMWR Morb Mort Wkly Rep
58:109-112, 2009.
Diedrick MJ, Flores AE, Hillier SL, et al: Clonal analysis of colonizing group B
Streptococcus, serotype IV, an emerging pathogen in the United States, J Clin
Microbiol 48:3100-3104, 2010.
Edgar JD, Gabriel V, Gallimore JR, et al: A prospective study of the sensitivity, specificity and diagnostic performance of soluble intercellular adhesion molecule
1, highly sensitive C-reactive protein, soluble E-selectin and serum amyloid A
in the diagnosis of neonatal infection, BMC Pediatr 10:22, 2010.
Ferrieri P, Baker SJ, Hillier SL, et al: Diversity of surface protein expression
in group B streptococcal colonizing and invasive isolates, Indian J Med Res
119:191-196, 2004a.
Garges HP, Mood MA, Cotton CM, et al: Neonatal meningitis: what is the correlation among cerebrospinal fluid cultures, blood cultures, and cerebrospinal
fluid parameters? Pediatrics 117:1094-1100, 2006.
Jordan HT, Farley MM, Craig A, et al: Revisiting the need for vaccine prevention
of late-onset neonatal group B streptococcal disease: a multistate, population
based analysis, Pediatr Infect Dis J 27:1057-1064, 2008.
Koenig JM, Keenan WJ: Group B streptococcus and early-onset sepsis in the era of
maternal prophylaxis, Pediatr Clin North Am 56:689-708, 2009.
Lam HS, Ng PC: Biochemical markers of neonatal sepsis, Pathology 40:141-148,
2008.
Levent F, Baker CJ, Rench MA, et al: Early outcomes of group B Streptococcal
meningitis in the 21st century, Pediatr Infect Dis J 29:1009-1012, 2010.
Puopolo KM, Eichenwald EC: No change in the incidence of ampicillin-resistant,
neonatal, early-onset sepsis over 18 years, Pediatrics 125:e1031-1038, 2010.
Verani JR, Schrag SJ: Group B streptococcal disease in infants: progress in prevention and continued challenges, Clin Perinatol 37:375-392, 2010.
Wynn JL, Neu J, Moldawer LL, et al: Potential of immunomodulatory agents for
prevention and treatment of neonatal sepsis, J Perinatol 29:79-88, 2009.
Wynn JL, Seed PC, Cotton CM: Does IVIg administration yield improved
immune function in very premature neonates? J Perinatol 30:635-642, 2010.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
40
Health Care–Acquired Infections
in the Nursery
David A. Munson and Jacquelyn R. Evans*
For decades, nosocomial or health care–acquired infection (HAI) has been considered by many as an unavoidable
problem associated with prolonged stays in an intensive
care nursery. Immature skin and immune systems, pathologic skin flora, and necessary invasive interventions all
put the premature neonate at high risk for HAI. But some
centers have clearly shown that, despite these risk factors, HAIs ranging from catheter-associated infections
to acquisition of respiratory viruses can largely be prevented (Andersen, 2005; Bloom et al, 2003; Kilbride et al,
2003). There is a direct financial cost to the health care
system associated with infections acquired while in the
hospital, but more importantly there is growing evidence
that the deleterious effects of the inflammatory response
to infection may adversely affect long-term outcomes,
including increased rates and severity of bronchopulmonary dysplasia and neurodevelopmental impairment
(Adams-Chapman and Stoll, 2001, 2006; Gonzalez et al,
1996; Hintz et al, 2005; Jobe and Ikegami, 1996; Leviton
et al, 1999; Rojas et al, 1995; Shah et al, 2008; Stoll et al,
2004).
The growing numbers of surviving very low-birthweight (VLBW) infants magnifies the impact of this
problem. Clinicians must make efforts to minimize the
exposure of newborns to known risk factors for infection.
Continuous surveillance and monitoring of endemic HAI
rates and patterns of responsible pathogens are necessary
to establish a reference point in each nursery and facilitate
early identification of epidemics. Prevention of infections
in the neonatal patient requires an expanding skill set in
addition to organized hospital-wide initiatives. Familiarity with quality improvement concepts, standardization
of practice, and hospital programs such as antimicrobial
stewardship are all essential to preventing harm in the
smallest patients.
DEFINITIONS
The Centers for Disease Control and Prevention (CDC)
provides the following surveillance definition for an HAI:
illness associated with a pathogen or its toxins that is not
present or incubating at the time of admission to the intensive care unit (Horan et al, 2008). As simple as this definition seems, it does leave some room for confusion for the
clinician caring for the neonatal population. Infections that
manifest early in the 1st week of life are typically related
to perinatal risk factors and vertical transmission from the
mother. HAIs are more often related to patient colonization and environmental risk factors. Unfortunately, there
*Acknowledgment: In writing this chapter, the authors excerpted significant portions of the chapter Nosocomial Infections in the Nursery by Ira Adams-Chapman and
Barbara Stoll from the previous edition of this book.
is no specific postnatal age that distinguishes maternally
transmitted infections from HAIs (Baltimore, 1998), and
it may be difficult to differentiate a late-onset, perinatally
acquired infection from an HAI. Most sources define HAIs
as infections occurring after 3 days of age (Baltimore, 1998;
Stoll et al, 2002a). The CDC defines HAI as any infection
that occurs after admission to the neonatal intensive care
unit (NICU) and that was not transplacentally acquired
(Garner et al, 1988). Other authorities suggest that HAI
be defined as any infection occurring more than 5 to 7 days
after birth (Baltimore, 1998).
The majority of HAIs in the neonatal population are
bloodstream infections associated with an intravascular
device. Criteria for central line–associated bloodstream
infections (CLABSIs), as defined by the CDC, are (1) isolation of a recognized pathogen from one blood culture
specimen or of a skin commensal from two blood culture
specimens; (2) one or more clinical signs of infection,
such as temperature instability, apnea, and bradycardia;
and (3) the presence of an intravascular device at the time
the culture specimen is collected (Horan and Gaynes,
2004). Ventilator-associated pneumonias are difficult to
diagnose in the NICU, but can contribute to morbidity, especially in patients with evolving lung disease.
Although the use of Foley catheters is low in the NICU
compared with other intensive care units, the acquisition
of urinary tract infections while in the NICU is still a
problem.
A summary of definitions of important HAIs in infants
is provided in Box 40-1. The prevention of the spread of
respiratory viruses is another area that demands attention, especially in a unit caring for infants with significant
bronchopulmonary dysplasia. The NICU, as with other
intensive care units, must remain vigilant in the control
of bacteria resistant to antibiotics such as methicillinresistant Staphylococcus aureus (MRSA) and vancomycinresistant enterococcus (VRE). As community-acquired
MRSA is becoming more prevalent, it is becoming more
difficult to determine the origin of MRSA in a neonatal
patient.
INCIDENCE
WELL BABY (HEALTHY NEWBORN) NURSERY
Accurate rates of HAI in the well baby nursery are difficult
to ascertain because there is no systematic reporting or
surveillance network for this issue; however, the incidence
appears to be low. Some researchers estimate rates of less
than 1 per 100 patients discharged (Baltimore, 1998).
Typical risk factors for acquiring a HAI, such as invasive
procedures and presence of an intravascular device, are
uncommon in the healthy newborn population. Discharge
551
552
PART IX Immunology and Infections
from the hospital within 48 hours of birth and rooming-in
practices have helped further to decrease the risk of exposure in modern mother-baby units. Early discharge may
also limit surveillance efforts, because patients may be
discharged home before they become symptomatic from
HAIs (Goldmann, 1989).
NEONATAL INTENSIVE CARE UNIT
The majority of neonatal HAIs occur in term and preterm infants hospitalized in special care nurseries. It is
difficult to determine and compare reports of endemic
HAI rates in different NICU populations. Any comparison of surveillance data between institutions is limited by
differences in patient demographics (e.g., birthweight,
BOX 40-1 D
efinitions of Nosocomial
Bacteremia and Pneumonia for
Patients Younger than 12 Months,
from the Centers for Disease
Control and Prevention
NOSOCOMIAL BLOODSTREAM INFECTIONS
Recognized pathogen isolated from blood culture and pathogen is not
related to infection at another site.
ll Must have at least one of the following clinical symptoms: fever (body
temperature >38°C, rectal), hypothermia (body temperature <37°C, rectal),
apnea, bradycardia, and a skin commensal isolated from two blood culture
specimens on separate occasions.
ll
PNEUMONIA
Chest radiograph with new or progressive infiltrate, cavitation, consolidation, or pneumatoceles
ll Worsening gas exchange and three of the following:
ll Temperature instability
ll White blood cell count <4000 or >15,000 with ≥10% bands
ll New-onset purulent sputum, change in character of sputum, increased
respiratory secretions, or increased suctioning requirements
ll Physical examination findings consistent with increased work of breathing or apnea
ll Wheezing, rales, or rhonchi
ll Cough
ll Bradycardia or tachycardia
ll
URINARY TRACT INFECTION
Clinical symptoms (fever [body temperature >38°C, rectal], hypothermia
[body temperature <37°C, rectal], apnea or bradycardia, dysuria, lethargy or
vomiting) and positive urine culture (≥100,000 microorganisms/mL) with no
more than two species identified
ll May also qualify as a UTI if the above symptoms are present without
another cause and has at least one of the following
ll Positive dipstick for leukocyte esterase, nitrate, or both
ll Pyuria
ll Organisms seen on Gram stain of unspun urine
ll At least two urine cultures with the same uropathogen with ≥100 colonies/mL
ll ≤100,000 colonies/mL of a single uropathogen if the patient is receiving
an effective antimicrobial agent
ll Physician diagnosis of UTI
ll Physician institutes appropriate therapy for a UTI
ll
Modified from Horan TC, Andrus M, Dudeck MA: CDC/NHSN surveillance definition of health
care-associated infection and criteria for specific types of infections in the acute care setting,
Am J Infect Control 36:3091-332, 2008.
UTI, Urinary tract infection.
gestational age distribution, underlying severity of disease, birth in the hospital versus elsewhere), back transport policies, and the use of different definitions for
HAI.
Among a cohort of 6215 VLBW infants (weight <1500 g)
who were monitored by the National Institute of Child
Health and Human Development (NICHD) Neonatal
Research Network, 21% of those who survived beyond
3 days of age had at least one episode of late-onset sepsis (Stoll et al, 2002b). There was significant variability
among the participating centers, with rates ranging from
10.6% to 31.7% at individual sites (Stoll et al, 2002b).
Moreover, rates were inversely related to birthweight and
gestational age, ranging from 43% for infants weighing
401 to 750 g at birth to 7% for infants weighing 1251 to
1500 g at birth. These data are similar to those reported
by Brodie et al (2000). In their study, 19% of 1354 infants
weighing less than 1500 g at birth had HAIs, and the rates
were highest for infants at the lowest birthweights (39%,
<750 g; 27%, 750 to 999 g; 10%, 1000 to 1499 g). A point
prevalence survey conducted by the 29 level II to level
IV nurseries participating in the Pediatric Prevention
Network revealed a prevalence of 11.4% for HAI (Sohn,
2001).
The CDC National Healthcare Safety Network
(NHSN) was developed in 2005 and replaced the National
Nosocomial Infections Surveillance system that reported
national data before that time. Some investigators believe
that reporting overall incidence rates may be misleading, because of the wide variations in practice and patient
populations in different units; therefore the NHSN system
monitors device-associated HAI rates by using an approach
that accounts for variability in device use and length of hospital stay (Edwards et al, 2008; Emori et al, 1991; Gaynes
et al, 1996). These data are also stratified by birthweight
categories and expressed as incidence density per 100 or
1000 patient-days. These two adjustments modify the relative risk on the basis of the severity of the illness and the
duration of exposure to the risk factor. A number of states
in the United States have mandatory reporting of HAIs
through the NHSN, and the number of participating
medical centers is growing. The NHSN data are similar to
institutional and collaborative epidemiologic data reported
elsewhere. Rates of device-associated infections are presented in Table 40-1. These rates remained constant in the
1990s (Gaynes et al, 1996; National Nosocomial Infections
Surveillance System Report, 2000), but appear to have
dropped considerably in the 2007 report (Edwards et al,
2008), suggesting that preventive strategies for CLABSIs
may be having an effect.
ANATOMIC SITES OF INFECTION
WELL BABY NURSERY
HAIs in the well baby nursery are most commonly superficial, involving the skin, mouth, or eyes. These infections include omphalitis, pustules, abscesses, and bullous
impetigo (Goldmann, 1989). Nursery epidemics of diarrhea caused by bacterial and viral enteropathogens have
been reported, but they occur infrequently (Goldmann,
1989).
CHAPTER 40 Health Care–Acquired Infections in the Nursery
NEONATAL INTENSIVE CARE UNIT
National and institutional surveillance data demonstrate
that bloodstream infections are responsible for the majority of HAIs among NICU patients (Gaynes et al, 1996;
National Nosocomial Infections Surveillance System
Report, 2000; Sohn et al, 2001). The remaining cases
involve the respiratory tract, eye, ear, nose, throat, or
gastrointestinal tract (Figure 40-1). Surveillance data for
meningitis are limited. Reports suggest that late-onset
meningitis may be underdiagnosed in the high-risk population of VLBW infants (Stoll et al, 2002b). There are
widespread differences in clinical practice regarding the
inclusion of a lumbar puncture with cerebrospinal fluid
TABLE 40-1 Rate of Device-Associated HAIs
Birthweight (g)
NHSN 2008
CLABSI per 1000
catheter days
NHSN 2008
VAP per 1000
ventilator days
<750
3.7
2.6
751-1000
3.3
2.1
1001-1500
2.6
1.5
1501-2500
2.4
1.0
>2500
2.0
0.9
Modified from Edwards JR, Peterson KD, Andrus ML, et al: National Healthcare
Safety Network (NHSN) Report, data summary for 2006 through 2007, issued
November 2008. National Healthcare Safety Network Facilities, Am J Infect Control
36:609-626, 2008. Level III NICU pooled data.
CLABSI, Central line–associated bloodstream infection; HAI, health care–acquired
infection NHSN, National Healthcare Safety Network; VAP, ventilator-associated
pneumonia.
553
analysis in the evaluation of a neonate with possible sepsis.
This variation in practice has been challenged by recent
data suggesting that generally accepted cerebrospinal fluid
parameters are not sensitive or specific for meningitis in
the neonate (Smith et al, 2008). Although early onset meningitis remains a rare event, clinicians need to consider
including a lumbar puncture in the initial evaluation of a
neonate with signs and symptoms of infection.
RISK FACTORS
Some risk factors for acquiring an HAI are reflections of
the patient population and are difficult or impossible to
modify. Other risk factors are directly related to the level
of supportive care associated with intensive care medicine
and may be difficult yet possible to modify. Risk factors for
HAI include lower gestational age and low birthweight;
invasive procedures; invasive devices, especially intravascular catheters and endotracheal tubes; parenteral nutrition
and intravenous lipids; colonization of skin, gastrointestinal tract, and airway with invasive organisms; selected
drugs administered to the neonate; and issues surrounding nursery staffing, nursery crowding, and hand washing
(Box 40-2) (Gaynes et al, 1996; Kawogoe et al, 2001; Perlman et al, 2007; Stoll et al, 1996, 2002a; Stover et al, 2001;
Suara et al, 2000). Minimizing exposure to known risk factors for infection is important to reduce the rate of HAIs
in the nursery.
The risk of developing an HAI is inversely related to
gestational age and birthweight (Gaynes et al, 1996, Kawogoe et al, 2001; National Nosocomial Infections Surveillance System Report, 2000; Stoll et al, 1996, 2002a; Stover
1000 g
1501–2500 g
BSI (49%)
BSI (32%)
EENT (21%)
SST (6%)
EENT (8%)
SSI (1%)
GI (7%)
PNEU (16%)
Other (13%)
GI (11%)
SST (10%)
SSI (3%)
Other (10%)
PNEU (13%)
(N = 3,987)
(N = 3,547)
1001–1500 g
>2500 g
BSI (45%)
BSI (36%)
EENT (13%)
EENT (14%)
SST (7%)
SSI (1%)
GI (10%)
PNEU (12%)
Other (11%)
SST (9%)
GI (5%)
SSI (7%)
Other (12%)
PNEU (18%)
(N = 1,881)
(N = 3,764)
FIGURE 40-1 Distribution of health care–acquired infections by site of infection. (Data from Gaynes RP, Edwards JR, Jarvis WR, et al: Nosocomial
infections among neonates in high-risk nurseries in the United States, Pediatrics 98:357-361, 1996.)
554
PART IX Immunology and Infections
BOX 40-2 R
isk Factors for Acquiring Health
Care–Acquired Infections in the
Neonatal Intensive Care Unit
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
ll
Prematurity
Low birthweight
Invasive device
Intravascular device (CVC, PAL, PICC, PIVC, umbilical catheter)
Mechanical ventilator
Urinary catheter
Ventriculoperitoneal shunt
Medications
Histamine2-blocking agents
Steroids
Others
Prolonged administration of hyperalimentation
Intralipid administration
Delayed enteral feedings
Feeding with formula rather than human milk
Inadequate nursery staffing and overcrowding
Poor compliance with handwashing
CVC, Central versus catheter; PAL, percutaneous arterial line; PICC, peripherally inserted
central catheter; PIVC, percutaneous intravenous catheter.
et al, 2001; Suara et al, 2000). Previous reports estimate
that there is a 3% lower risk of acquiring an HAI with
each 500-g increment in birthweight (Goldmann, 1989).
Infants with birthweights less than 1000 g have twice the
rate of nosocomial bloodstream infections than do infants
with birthweights greater than 1000 g (Brodie et al, 2000;
Geffers et al, 2008; Gaynes et al, 1996; National Nosocomial Infections Surveillance System Report, 2000; Stoll
et al, 2002a; Stover et al, 2001). Severity of illness scores
may be more predictive than birthweight alone, because
they reflect physiologic stability and the cumulative need
for intervention and invasive therapies (Goldmann, 1989;
Gray et al, 1995).
The use of any type of invasive device increases the risk
for infection. The most common invasive devices used in
the nursery are intravascular catheters, mechanical ventilators, ventriculoperitoneal shunts, and urinary catheters. In general, the risk rises as the duration of exposure
lengthens. Compared with adult patients, neonates are at
higher risk for catheter-related bloodstream infections
and at lower risk for ventilator-associated pneumonia and
urinary tract infections (Langley et al, 2001; National
Nosocomial Infections Surveillance System Report,
2000). These patterns correlate with the frequency with
which these invasive devices are used in the neonatal
patient population.
Prolonged duration of mechanical ventilation is the
primary risk factor for development of hospital-acquired
pneumonia. Contamination of respiratory equipment—
especially with gram-negative organisms that thrive in
moist environments, such as Acinetobacter, Pseudomonas,
and Flavobacterium spp.—frequently leads to colonization
of the respiratory tract. There is some evidence that aspiration of gastric and oropharyngeal secretions around the
uncuffed endotracheal tube may occur, and this could be a
mechanism for ventilator-associated pneumonia (Farhath
et al, 2008). The various closed-system suctioning devices
may decrease the risk of iatrogenic contamination during
suctioning. Defining a ventilator-associated pneumonia in
the neonatal population remains a challenge. Patients in
the NICU do not often meet all of the parameters defined
by the CDC (see Box 40-1), but acute worsening of pulmonary function in an infant with a ventilator could represent a new-onset pneumonia. Collaboration among the
largest NICUs is needed to better define this entity in the
neonatal population.
Intravascular devices commonly used in the neonatal
population are peripheral intravenous catheters (PIVs),
umbilical catheters, peripherally inserted central catheters (PICCs), surgically placed central venous catheters
(CVCs) and percutaneous arterial catheters. Regardless
of the type of device used, the rate of catheter-related
bloodstream infections is directly related to the number
of days the catheters are in place and inversely related to
the gestational age and birthweight of the patients (Gaynes
et al, 1996; Kawogoe et al, 2001; National Nosocomial
Infections Surveillance System Report, 2000; Stoll et al,
2000a; Suara et al, 2000). Coagulase-negative Staphylococcus (CONS) remains the primary pathogen associated with
catheter-related bloodstream infections (Garland et al,
2001).
PIVs are the most commonly used device for vascular
access in neonates. In adults, the removal of such catheters after 72 hours is recommended. Data in neonates are
insufficient to recommend elective removal of PIVs after
72 hours, because studies have not shown a clear correlation between the higher colonization rate noted after
72 hours and an increased rate of catheter-related bloodstream infection (Oishi, 2001; Pearson, 1996). Further
study is needed to answer this question.
Data comparing the infection rates of the various types
of intravascular catheters are limited. Theoretically the
risk should be lower for tunneled catheters, because the
Dacron cuff proximal to the exit site of a surgically placed
catheter can inhibit the migration of organisms into the
catheter tract (Mermel et al, 2001). Adult data suggest that
tunneled catheters have lower infection rates than nontunneled catheters; however, a report of 79 surgical neonates
requiring tunneled lines showed a rate of infection of 9.9
per 1000 catheter days (Klein et al, 2003), comparable to or
worse than reported rates of PICC line infections in other
NICU populations. This issue needs further study in the
neonatal population, especially because PICCs are being
used with greater frequency for long-term vascular access
in neonates (Stoll et al, 2002a). Some recent trials comparing PIVs to PICC lines for the provision of parenteral
nutrition have challenged the notion that the presence of
a central line increases the risk of infection above the risk
that exists simply with the use of peripheral catheters. It
appears that the use of a PICC line decreases the risk of
complications associated with PIVs without increasing the
rate of infection, at least in the initial weeks of life (Ainsworth et al, 2007). There is certainly much work to be
done to determine the ideal access for the different populations cared for in the NICU.
The use of intravenous lipid emulsions increases the
risk for infection (Freeman et al, 1990). Lipid emulsions
decrease the flow rate through the intravenous catheter and potentiate the growth and proliferation of some
microorganisms. Lipid emulsions can interfere with host
555
CHAPTER 40 Health Care–Acquired Infections in the Nursery
defense mechanisms by impairing the function of neutrophils and reticuloendothelial macrophages (Freeman et al,
1990; Langevin et al, 1999; Nugent, 1984). Extrinsic contamination has been reported but rarely occurs in the
United States (Hernandez-Ramos et al, 2000). Freeman et al
(1990) reported that the use of a lipid emulsion was positively and independently predictive for the development of
CONS bacteremia. Infants who demonstrated CONS sepsis were 5.8-fold more likely to have been exposed to lipid
emulsions. Administration of such emulsions has also been
linked to a higher risk for HAI with Candida and Malassezia
spp. in neonates (Long and Keyserling, 1985; Redline et al,
1985; Saiman et al, 2000).
Histamine-blocking agents and postnatally administered
corticosteroids are the medications most commonly associated with an increased risk of HAIs among newborns.
It is hypothesized that the reduced gastric pH associated
with the use of histamine2-blocking agents promotes
bacterial overgrowth and invasion of pathogenic bacteria
(Beck-Sague et al, 1994; Stoll et al, 1999). Dexamethasone
has been used in ventilator-dependent preterm infants to
facilitate weaning from the ventilator and to minimize
the risk of chronic lung disease. The use of dexamethasone has decreased in the last decade in VLBW infants
secondary to concerns of spontaneous bowel perforation
and adverse effects on growth and neurodevelopmental
outcome (O’Shea et al, 1999; Stark et al, 2001, Vohr et
al, 2000). However, there is renewed interest in a role for
steroids after the first week of life, especially in infants who
have demonstrated a high risk for developing chronic lung
disease (Doyle et al, 2005, 2006, 2007). Because the use
of dexamethasone has been associated with an increased
risk of infection in VLBW infants (Stoll et al, 1999; Yeh et
al, 1997), neonatologists will need to include this concern
in any risk-benefit analysis of the use of steroids in their
patients.
Nursery design and staffing influence the risk of infection. Overcrowding and larger workloads decrease compliance with hand washing and raise the risk of HAI
(Archibald et al, 1997; Fridkin et al, 1996; Harbarth et al,
1999; Robert et al, 2000; Vicca, 1999). Inadequate numbers of staff and the use of temporary or inexperienced
staff members both adversely affect the rate of infection.
The adverse effects of inadequate staffing and overcrowding were demonstrated by Hayley and Bregman (1982),
who showed a relationship between nurse-to-patient ratio
and colonization of patients with MRSA. Fridkin et al
(1996) found that patient-to-nurse ratio was an independent predictor of development of a catheter-related bloodstream infection. Furthermore, strategic nursery design
and improvement in nursing staffing correlate with lower
rates of HAIs (Gladstone et al, 1990).
DISTRIBUTION BY PATHOGEN
The predominant pathogens responsible for nosocomial
bloodstream infections have changed over time. Goldmann (1989) proposed that these trends are explained by
changes in the neonatal intensive care patient population
and advancing technology. S. aureus was the most common nosocomial pathogen in the 1950s and 1960s. In
the 1960s and 1970s, gram-negative organisms emerged
TABLE 40-2 Distribution of Pathogens Responsible
for Bloodstream Infections
Pathogen
No.
%
Coagulase-negative
staphylococci
3833
51.0
Staphylococcus aureus
563
7.5
Group B streptococci
597
7.9
Enterococcus spp.
467
6.2
Candida spp.
518
6.9
Escherichia coli
326
4.3
Other Streptococcus spp.
205
2.7
Enterobacter spp.
219
2.9
Klebsiella pneumoniae
188
2.5
Data from Gaynes RP, Edwards JR, Jarvis WR, et al: Nosocomial infections among
neonates in high-risk nurseries in the United States, Pediatrics 98:357-361, 1996.
as the predominant pathogens; globally, these organisms
remain the most important pathogens responsible for
HAIs in the nursery (Stoll, 2001). National surveillance
data in the United States indicate that CONS is currently the most common nosocomial pathogen (Gaynes
et al, 1996; National Nosocomial Infections Surveillance
System Report, 2000) (Table 40-2). In the United States,
the distribution of pathogens has not changed significantly over the past decade (Gaynes et al, 1996; Stoll et
al, 1996, 2002a) (Box 40-3). Among a cohort of infants
with birthweights less than 1500 g with late-onset infections, the NICHD Neonatal Research Network reported
that gram-positive organisms were responsible for 70%
of cases, gram-negative organisms for 18%, and fungi for
12% (Stoll et al, 2002a). These findings are similar to those
of a 10-year retrospective analysis of pathogens in a single
center, in which gram-positive organisms caused 57% of
late-onset infections (Karlowicz et al, 2000). The distribution of infecting pathogens did not differ with birthweight
or timing of infection (Stoll et al, 2002a). More recent
articles evaluating interventions and mechanisms of infection confirm the continued predominance of CONS in
neonatal line infections in the United States (Garland
et al, 2008) as do review articles (Curtis and Shetty, 2008).
For the clinician, understanding the specific colonization and resistance patterns in the individual NICU is perhaps more important than being aware of national trends.
The emergence of nosocomial pathogens with antimicrobial resistance is a concern. In patients of all ages, reports
estimate that 50% to 60% of HAIs are caused by resistant organisms (Jones, 2001; Weinstein, 1998). Organisms showing patterns of increasing antibiotic resistance
of importance to NICU patients are VRE, MRSA, and
multidrug-resistant gram-negative organisms (Bizzarro
and Gallagher, 2007).
GRAM-POSITIVE BACTERIA
CONS is the most common endemic nosocomial pathogen
in neonates (Brodie et al, 2000; Garland et al, 1996, 2008;
Gray et al, 1995; National Nosocomial Infections Surveillance system report, 2000; Stoll et al, 2002a). The majority
of CONS infections are bloodstream infections. Reported
556
PART IX Immunology and Infections
BOX 40-3 C
ommon Pathogens Causing
Health Care–Acquired Infections
GRAM-POSITIVE ORGANISMS
Coagulase-negative staphylococci
ll Staphylococcus aureus
ll Enterococcus spp.
ll Group B Streptococcus
ll
GRAM-NEGATIVE ORGANISMS
Escherichia coli
ll Klebsiella spp.
ll Pseudomonas spp.
ll Enterobacter spp.
ll Serratia spp.
ll Haemophilus spp.
ll Acinetobacter spp.
ll Salmonella spp.
ll
FUNGAL ORGANISMS
Candida albicans
ll Candida parapsilosis
ll Malassezia furfur
ll
VIRAL ORGANISMS
Respiratory syncytial virus
ll Influenza
ll Varicella zoster
ll Rotavirus
ll Enterovirus
ll
Data from Gaynes RP, Edwards JR, Jarvis WR, et al: Nosocomial infections among neonates
in high-risk nurseries in the United States, Pediatrics 98:357-361, 1996; and Stoll BJ, Hansen
N, Fanaroff AA, et al: Late-onset sepsis in very-low-birth-weight neonates: the experience of
the NICHD Neonatal Research Network, Pediatrics 110:285-291, 2002.
incidence ranges from 51% to 78% among VLBW infants
(Gray et al, 1995; Isaacs et al, 1996; Stoll, 1996, 2002a).
Gray et al (1995) reported a cumulative incidence of 17.5
episodes of CONS sepsis per 100 patient-days and an incidence density of 6.9 episodes of CONS sepsis per 1000
patient-days.
Known risk factors for CONS infection are low birthweight, lower gestational age, use of central venous catheters, prolonged administration of hyperalimentation, use
of intravenous lipid emulsions, postnatal administration
of corticosteroids, and prolonged hospital stay (Brodie
et al, 2000; Freeman et al, 1990; Goldmann, 1989; JohnsonRobbins et al, 1996). CONS is the pathogen most commonly associated with catheter-related infections, partially
because it produces a capsular polysaccharide adhesin
(poly-N-succinyl glucosamine), which enhances its ability
to adhere to intravascular devices. Although some studies
suggest that prophylactic use of vancomycin reduces the
risk of CONS catheter-related infections, this practice is
not recommended because of the serious risk of encouraging antibiotic-resistant organisms, especially VRE and
staphylococci. The use of a vancomycin lock, where vancomycin is instilled into the catheter and then removed
after a specified dwell time, has more promise in terms of
balancing the risk of systemic exposure and prophylaxis
against contamination of the catheter lumen. Garland
et al (2005) demonstrated a reduction in the rate of CLABSI
from 17.8 to 2.4 per 1000 catheter days using this technique. Although promising, several units have achieved
comparable reductions without the use of this kind of
intervention. Consequently, the role of an antibiotic lock
will likely find its place in certain circumstances or in populations that have demonstrated persistently elevated risks
for infection.
Enterococci are responsible for both endemic and epidemic HAIs in the NICU. Use of central venous catheters,
prolonged hospital stay, and prior antibiotic use are recognized risk factors for colonization with these organisms.
The gastrointestinal tract is often the primary source of
infection; however, the pathogens are typically spread
via the hands of health care workers or through environmental contamination. The widespread use of antibiotics
has led to the emergence of VRE. There are published
guidelines to prevent the spread of VRE, which include
hand washing, isolation, barrier precautions, and cohorting of infected patients (Gross and Pujat, 2001; Hospital
Infection Control Practices Advisory Committee, 1995).
Educational programs to limit the indiscriminate use of
antibiotics have been effective in decreasing the spread of
VRE (Goldmann et al, 1996; Isaacs, 2000).
S. aureus has caused epidemics in well baby nurseries and
in NICUs. In the nursery, the major reservoirs for staphylococci are colonized or infected infants. These infants
transmit the organism to health care workers, who subsequently infect other infants. The skin, nares, and umbilicus
are the most common sites of colonization. Unfortunately,
routine surveillance cultures are not useful because colonization rates correlate poorly with infection rates. Studies in adult patients have shown that many patients who
demonstrate S. aureus bacteremia were colonized with the
identical strain at the time of admission to the hospital, suggesting that some of the infections with S. aureus are community acquired rather than hospital acquired (von Eiff et
al, 2001). Similarly, health care workers can be colonized
with a community-acquired strain of S. aureus, which they
then transfer to vulnerable infants. Importantly, there has
been a significant increase in methicillin-resistant strains
of S. aureus causing infections in the NICU. A review of
the National Nosocomial Infections Surveillance data
from 1995 to 2004 indicated a 308% increase in the incidence of MRSA over that 10-year time frame. As of the
2004, MRSA accounted for 34% of S. aureus infections in
the reported data (Lessa et al, 2009). Consequently, when
covering for a possible S. aureus infection, it is critical to
select an antibiotic that is effective against methicillinresistant strains.
Group B streptococcus (GBS) remains an important cause
of late-onset infection in neonates, but do not have a clear
role as an HAI. Intrapartum antibiotic treatment to prevent vertical transmission of GBS from a colonized mother
to her infant has led to a decrease in early-onset GBS disease. In contrast, the incidence of late-onset GBS disease
has remained unchanged, presumably because prophylaxis
does not eradicate colonization of the genital tract or the
environment.
GRAM-NEGATIVE ORGANISMS
Gram-negative organisms are a particularly important
cause of nosocomial bloodstream infections, pneumonia,
and meningitis because they generally cause severe disease.
CHAPTER 40 Health Care–Acquired Infections in the Nursery
Escherichia coli is the most common gram-negative pathogen. Other gram-negative organisms responsible for HAI
are Klebsiella, Pseudomonas, Enterobacter, Acinetobacter, Serratia, Haemophilus, and Salmonella spp. (see Box 40-3).
The attributable mortality is much higher for gram-negative infections than for gram-positive infections. Stoll et al
(2002a) reported that infants with gram-negative infections
had a 3.5-fold higher risk of death. Karlowicz et al (2000)
found that gram-negative infections were associated with
fulminant death within 48 hours of a positive blood culture
result in 69% of cases. Pseudomonas spp. appear to be particularly virulent, causing death in 42% to 75% of infected
neonates (Karlowicz et al, 2000; Leigh et al, 1995; Stoll et
al, 2002a). A more recent study by Makhoul et al (2005)
and colleagues in Israel confirms that gram-negative infections are associated with a substantially higher mortality.
FUNGAL ORGANISMS
Fungal infections are discussed in detail in Chapter 41.
Invasive fungal infection is estimated to occur in 1% to 4%
of VLBW infants and the incidence in extremely lowbirthweight (ELBW) infants has been reported to be significantly higher. The risk may even approach 20% in infants
with a birthweight less than 750 g (Bartels et al, 2007;
Benjamin et al, 2006; Clerihew et al, 2006; Fridkin et al,
2006; Makhoul et al, 2002, 2007; Rodriguez et al, 2006;
Stoll et al, 2002a). Rates and predominant fungal species
vary among clinical centers. The smallest and most premature infants appear to be at the highest risk, particularly
when they are exposed to broad-spectrum antibiotics or
long courses of antibiotics. Other identified risk factors are
prolonged mechanical ventilation, prolonged use of central venous catheters, prior use of lipid emulsions, and the
use of histamine type 2 antagonists (Benjamin et al, 2001;
Long and Keyserling, 1985; Makhoul et al, 2002; Saiman
et al, 2000).
Efforts at preventing nosocomial fungal infection have
focused on the use of prophylactic fluconazole for VLBW
infants. This strategy has been shown to decrease the rate
of invasive disease in randomized trials, but the baseline
rates in the control arms in some studies have been high
(Kaufman et al, 2001). Consequently it has been difficult to
determine how generalizable the findings are. Furthermore,
metaanalysis has not demonstrated an associated improvement in mortality (Clerihew et al, 2007). A number of
retrospective studies describing the implementation of fluconazole prophylaxis in individual units have also suggested
a reduction in invasive fungal disease (Aghai et al, 2006; Bertini et al, 2005; Dutta et al, 2005; Healy et al, 2005; Manzoni
et al, 2006; Uko et al, 2006). Although no study has found an
increase in fluconazole-resistant fungus, resistance remains
a concern with any prophylactic strategy. Limiting a prophylactic strategy to units with a high rate of invasive candidal disease and to patients with the highest risk will likely
ultimately provide the greatest benefit with the lowest risk.
VIRAL ORGANISMS
Viral organisms that cause HAI in the NICU include
respiratory syncytial virus (RSV), rhinovirus, metapneumovirus, influenza, varicella, rotavirus, and enterovirus.
557
Isolated infections generally result from contact with
infected caregivers or family members. Nursery epidemics
may occur in addition to isolated individual cases.
Respiratory Syncytial Virus
Careful hand washing is the most effective measure to
prevent RSV infection. Recommendations for preventing
RSV epidemics in the inpatient setting include cohorting
of patients, barrier precautions, and careful hand washing (Goldmann, 2001; Hall, 2000; Karanfil et al, 1999;
Mlinarić-Galinović and Varda-Brkić, 2000; Snydman et al,
1988). Rapid testing to detect the virus in nasal washings
facilitates efforts to cohort infected patients, which has
been shown to be an effective control measure (Madge
et al, 1992; Snydman et al, 1988). RSV is a fastidious organism capable of surviving on inanimate objects for prolonged
periods; therefore some authorities advocate the use of
gowns and gloves, because of the increased risk of contamination through casual contact between the patient and the
environment (Goldmann, 2001; Hall, 2000; Karanfil et al,
1999; Mlinarić-Galinović and Varda-Brkić, 2000). Several
case reports have described the use of palivizumab, an RSV
monoclonal antibody, to control nosocomial outbreaks;
however, the efficacy of administering monthly injections
to hospitalized patients has not been critically or systematically evaluated (Cox et al, 2001; Macartney et al, 2000).
Preterm infants born before 32 weeks’ gestation and those
with chronic lung disease remain at high risk for RSV
infection after hospital discharge; therefore the American
Academy of Pediatrics (AAP) recommends that all highrisk infants (<32 weeks’ gestation, chronic lung disease,
or asymptomatic acyanotic congenital heart disease, such
as a patent ductus arteriosus or ventricular septal defect)
receive up to five doses of palivizumab during the RSV season (American Academy of Pediatrics, 1998, 2009a; Meissner et al, 1999). These guidelines were updated in 2003
to also include patients with cyanotic congenital heart disease. They have been modified again in 2009 to clarify the
risk factors for the 32- to 35-week gestational age group
(American Academy of Pediatrics, 2009a).
Influenza
Influenza is spread primarily via airborne transmission.
Hand washing and immunization of health care workers
are the primary tools to prevent nosocomial spread of this
virus (Nichol and Hauge, 1997). Most infection control
guidelines recommend that every health care worker wear
a mask during contact with infected patients. Although
several drugs are available for the prophylaxis and treatment of influenza, they have not been studied in newborns and cannot be recommended at this time (Meissner,
2001). At-risk infants should receive the influenza vaccine
during the winter months once they reach the age of
6 months.
With the appearance of the novel H1N1 influenza
strain, the season for possible flu exposure has increased
substantially. In addition, newborns are thought to be in a
high-risk population if they are born to mothers who are
actively infected. Guidelines for minimizing risk to the
infant have been offered by the CDC. Pregnant women
558
PART IX Immunology and Infections
with symptoms that are consistent with H1N1 influenza
should receive treatment as soon as possible. If tolerated,
the mother should wear a mask during labor and delivery.
After the delivery, the infant should be cared for away from
the mother until he or she has received 48 hours of treatment and has become afebrile and able to control cough and
secretions. The infant should be assisted in lactation if she
intends to breastfeed, because the breast milk itself is not
thought to be a means of viral transmission (CDC, 2009).
Varicella
Potential exposures and epidemics of nosocomial varicella
have been reported, but true nosocomial infection in the
NICU is rare. The index case is typically an asymptomatic
infected health care worker or family member who has had
contact with a susceptible infant before the onset of clinical
disease in the health care worker or family member. The
incubation period lasts 14 to 16 days, and infected persons
are contagious 24 to 48 hours before the appearance of the
rash. The relative risk of infection varies according to the
intensity of the exposure and the presence of maternal antibody in the infant. The majority of transplacental antibody
transfer occurs during the third trimester; therefore most
extremely preterm infants are born before this process is
complete. ELBW infants may be at risk even if their mothers have a documented titer of varicella antibodies. The
AAP recommends administration of varicella zoster immunoglobulin (VZIG) to any newborn whose mother shows
signs of varicella infection within 5 days before delivery
or within 2 days after delivery. If the exposure is postnatal, whether via a family member or a health care worker,
an exposed preterm infant born before 28 weeks’ gestation
or weighing less than 1000 g birthweight should receive
immunoprophylaxis regardless of the mother’s varicella
history or serologic status. A postnatally exposed preterm
infant born after 28 weeks’ gestation should receive VZIG
only if the mother lacks clinical or serologic evidence of
prior disease (American Academy of Pediatrics, 2009b).
Airborne and contact precautions are recommended for
all infants with active varicella disease for at least 5 days
after vesicles appear and until all lesions have crusted.
Airborne and contact precautions should be used for any
exposed susceptible patient from 8 to 21 days after the
exposure. Administration of VZIG potentially prolongs
the incubation period; therefore all exposed infants who
receive this product should be isolated for up to 28 days
after the exposure. An infant born to a mother with active
varicella should be isolated for 21 days, or for 28 days if the
infant received VZIG. The infants should also be isolated
from the mother until all of her lesions have crusted.
Rotavirus
Although rare, epidemics of rotavirus diarrhea may occur
in the nursery (Herruzo et al, 2009; Jain and Glass, 2001;
Lee et al, 2001; Widdowson et al, 2000); they are primarily caused by inadequate hand washing and cross-contamination between patients. Standard and contact precautions
should be followed throughout the duration of the illness.
Some patients have prolonged fecal shedding of low concentrations of the virus; therefore some infection control
experts recommend contact precautions for the duration of
the hospitalization of such patients. Rotavirus is an important cause of diarrhea in older infants and should be suspected with any apparent epidemic of diarrhea. There are
now two live virus vaccines available for the prevention of
rotavirus infection, but their use in the NICU remains controversial because of concerns about the possibility of illness
occurring in immunocompromised infants via spread of the
vaccine related virus. Currently the AAP recommends the
administration of rotavirus vaccine at the time of discharge
or after discharge (American Academy of Pediatrics, 2009c).
Enterovirus
There are numerous serotypes of enteroviruses, including
polioviruses, Coxsackie viruses A and B, echovirus, and
nonassigned subtypes (Chambon et al, 1999). Enterovirus infections have been described among neonates in the
well baby nursery and the neonatal intensive care setting
(Chambon et al, 1999; Isaacs et al, 1989; Sizun et al, 2000;
Takami et al, 2000, Wreghitt et al, 1989). Both individual
cases and epidemics can occur. The seasonal distribution
of enterovirus infections in neonates mirrors what is seen
in the community.
The clinical presentation associated with enteroviral
infection is variable. Many patients are asymptomatic;
however, several case reports describe overwhelming
infection with multisystem organ dysfunction and death
(Jankovic et al, 1999; Keyserling, 1997). Clinical manifestations of severe disease include meningoencephalitis,
hepatic dysfunction, disseminated intravascular coagulation, myocarditis, pneumonitis, gastroenteritis, and muscle
weakness (Abzug et al, 1993; Isaacs et al, 1989; Keyserling,
1997; Wreghitt et al, 1989). The severity of disease and
the likelihood of death are often more pronounced in perinatally acquired cases than in nosocomially acquired cases,
presumably related to the lack of maternal antibody present in the neonate (Isaacs et al, 1989; Modlin et al, 1981).
Infection control measures during an enteroviral epidemic include hand washing with alcohol-based preparations or antimicrobial soaps and cohorting of infected
patients. Surveillance cultures of specimens from the throat
and rectum may be helpful in identifying asymptomatic
colonized infants. Blood and cerebrospinal fluid cultures
should be obtained from any patient with clinical symptoms
of disease. Polymerase chain reaction analysis is helpful in
making a rapid diagnosis (Nigrovic, 2001). No antiviral
agents are currently available to treat enteroviral infections
in newborns (Nigrovic, 2001). Although commercially
available intravenous immunoglobulin preparations have
high levels of neutralizing antibodies to common enterovirus serotypes, there is no clear evidence that administration of immunoglobulin alters the process or outcome of
enteroviral infection (Abzug et al, 1993; Dagan et al, 1983;
Keyserling, 1997); therefore the use of these products
remains controversial in patients with enteroviral infection.
EPIDEMICS
Numerous nursery epidemics have been reported. Common sources for infection are contaminated equipment
(i.e., breast pump, thermometer, ventilator), environmental
CHAPTER 40 Health Care–Acquired Infections in the Nursery
reservoirs, soaps, and lapses in hand hygiene practices
(Focca et al, 2000; Hervas et al, 2001; Hoque et al, 2001;
Jeong et al, 2001; Jones et al, 2000; McNeil et al, 2001;
Reiss et al, 2000; van den Berg et al, 2000; Wisplinghoff
et al, 2000; Yu et al, 2000). Clinicians must have a high
index of suspicion to detect nursery outbreaks, especially
when clusters of infections with unusual pathogens occur
in a nursery. Continuous surveillance and monitoring of
the endemic infection rates are crucial for determining
when there has been a significant change in the baseline
pattern of infection for a given nursery. Modern molecular
technology facilitates identification and tracking of specific
strains of bacteria to help determine the common source
of infection (Chambon et al, 1999; Jeong et al, 2001; Jones
et al, 2000; Takami et al, 2000).
Epidemics must be identified promptly, and immediate control measures must be instituted. Efforts should
be made to identify the causal agent and the mode of
transmission. Surveillance of staff members may be necessary, even if they are asymptomatic. Reinforcement of
hand-washing policies is of utmost importance. Cohorting
infected patients may be helpful in limiting the spread of
organisms in the nursery. In an effort to minimize the risk
of cross-contamination between patients, staff members
caring for colonized or infected infants should not care for
infants who are not infected or colonized.
INFECTION CONTROL
The CDC has developed a two-tiered approach to infection control. Standard precautions should be used with
all patient contact regardless of the underlying diagnosis
or infectious status. These precautions consist of universal precautions (designed to prevent blood and body fluid
contamination) and body substance precautions (designed
to prevent contamination with moist substances). Transmission-based precautions are necessary when a patient is
infected with a known or suspected pathogen that is associated with a high risk of contamination via airborne or
droplet transmission or contact with the skin or contaminated surfaces (Garner, 1996).
The routine use of gowns is not an effective measure
to decrease the endemic HAI rate in the nursery (Garner,
1996; Goldmann, 1989, 1991). Gowns should be used in
specific circumstances in which the risk for contamination is high or the infant is being held. Gowns should be
changed after each patient encounter.
Visitation policies are not restrictive in most modern
nurseries. Various regulatory agencies have developed
guidelines for sibling visitation (Box 40-4) (American
Academy of Pediatrics, 1985). Infection and colonization
rates have not risen with use of the current standards. Siblings should be screened for infection or recent exposures
before visiting.
PREVENTION OF HEALTH
CARE–ACQUIRED INFECTIONS
Several intensive care nurseries have described their success in decreasing the rate of central line–associated infections (Andersen et al, 2005; Bloom et al, 2003; Kilbride
et al, 2003). Until recently it was considered implausible
559
BOX 40-4 G
uidelines for Sibling Visitation
in the Nursery
ll
ll
ll
ll
Siblings should not have been exposed to known communicable diseases
(e.g., varicella).
Siblings should not have fever or symptoms of acute illness (i.e., upper
respiratory infection or gastroenteritis).
Children should be supervised by their parents or a responsible adult during
the entire visit.
Children should be prepared in advance for the visit.
Adapted from the American Academy of Pediatrics Committee on the Fetus and Newborn:
Postpartum (neonatal) sibling visitation, Pediatrics 76:650, 1985.
by many that HAIs could be eliminated completely. But
the challenge that faces neonatologists, nurses, respiratory therapists, physical therapists, clerks, environmental
service staff, and anyone else who affects the care of the
neonate now must be framed by the following question:
can we get to zero? Much is known about the mechanisms
of catheter-related infections and the factors that increase
risk. Effective prevention strategies focus on modifying
the known risks, such as standardization of procedures
that relate to central line care, strategic nursery design,
adequate staffing, hand-washing compliance, minimization of catheter days, and promotion of enteral nutrition,
especially with human milk (Adams-Chapman and Stoll,
2002; Goldmann, 1989; Horbar et al, 2001). Care “bundles” incorporating a group of interventions aimed at standardizing central line care can be effective if a particular
intensive care unit is committed to implementing them.
One hundred percent compliance with adequate hand
hygiene is required to eliminate the spread of viruses and
bacteria, particularly those that are drug resistant. Monitoring, surveillance, and benchmarking of the HAI rates in
the nursery are also critical components of any prevention
program (Box 40-5). There is power in simply knowing
how an individual unit compares to others, and there is
empowerment in knowing that preventing HAIs is possible (Schulman et al, 2009).
HAND HYGIENE
Historic Perspective
Hand washing has clearly been shown to be the most
effective and least expensive means of preventing the
spread of HAI; however, compliance with this simple
practice is poor (Jarvis, 1994; Larson, 1999; Pittet, 2000;
Pittet et al, 2000). Historically, the benefits of hand washing have been known since the early nineteenth century.
Labarraque, a French pharmacist, was one of the first to
demonstrate that cleansing the hands with solutions containing lime or soda could be used as disinfectants and
antiseptics (Boyce et al, 2002). The issue was further elucidated by Ignaz Semmelweis in the 1850s, who noted
that the mortality and puerperal infection rates were
higher among women receiving care from physicians
than in those receiving care from midwives. He postulated that the puerperal fever in these patients was caused
by “cadaverous particles” transmitted from the autopsy
suite on the hands of the students and physicians. After
560
PART IX Immunology and Infections
BOX 40-5 P
rinciples for the Prevention of
Health Care–Acquired Infections
in the NICU
ll
ll
ll
ll
ll
ll
ll
ll
Observe recommendations for standard precautions with all patient contact.
Observe recommendations for transmission-based precautions as indicated.
ll Gowns
ll Gloves
ll Masks
ll Isolation
Use good nursery design and engineering.
ll Appropriate nurse-to-patient ratio
ll Avoidance of overcrowding and excessive workload
ll Readily accessible sinks, antiseptic solutions, soaps, and paper towels
ll Maintain hand-washing practices.
ll Improving hand-washing compliance
ll Washing of hands before and after each patient encounter
ll Appropriate use of soap, alcohol-based preparations, or antiseptic solutions
ll Alcohol-based antiseptic solution at each patient’s bedside
ll Emollients provided for nursery staff
ll Education and feedback for nursery staff
Minimize risk of contamination of CVCs.
ll Maximal sterile barrier precautions during CVC insertion
ll Local antisepsis with chlorhexidine gluconate
ll Minimal entries into the line for laboratory tests
ll Aseptic technique when entering the line
ll Minimal CVC days
ll Sterile preparation of all fluids to be administered via a CVC
Provide meticulous skin care.
Encourage early and appropriate advancement of enteral feedings.
Provide education and feedback for nursery personnel.
Perform continuous monitoring and surveillance of HAI rates in the NICU.
CVC, Central venous catheter; HAI, health care–acquired infection; NICU, neonatal intensive
care unit.
he instructed physicians to cleanse their hands with a
chlorine solution between patients, the maternal infection and mortality rates dropped dramatically (Boyce
et al, 2002; Semmelweis, 1983). This intervention represents the first clinical evidence suggesting that cleansing contaminated hands with an antiseptic agent was
more effective than washing them with plain soap and
water and that hand antisepsis could reduce the spread of
contagious disease via the hands of health care workers
(Boyce et al, 2002).
Guidelines for Hand Hygiene Practices
in the Neonatal Intensive Care Unit
Clinical Indications for Hand Hygiene
Hand hygiene techniques are effective in decreasing
the colonization rate of resident and transient flora and
have been shown to reduce cross-contamination among
patients. Several studies have shown that the hands of
health care workers become contaminated during routine
patient care activities, including “clean” procedures such
as lifting patients and checking vital signs, as well as during contact with intact skin (Casewell and Phillips, 1977;
Pittet et al, 1999). Attempts have been made to stratify
the type of activity with the likelihood of contamination,
but they have not been validated by quantitative bacterial contamination analysis. Direct patient contact and
BOX 40-6 R
ecommendations for Hand
Hygiene Practices in the NICU
INDICATIONS FOR HAND HYGIENE
Wash hands with a nonantimicrobial soap or an antimicrobial soap and
water when hands are visibly soiled or contaminated with proteinaceous
material.
ll If the hands are not visibly soiled, then alcohol-based, waterless antiseptic
agents are strongly preferred for routine decontamination of hands in all
other clinical situations.
ll Alcohol-based, waterless antiseptic agents should be available at each
patient area and other convenient locations, and in individual pocket-sized
containers for health care providers.
ll Antimicrobial soaps can be considered in settings with few time constraints
and easy access to hand hygiene facilities.
ll Decontaminate hands after contact with intact patient skin (i.e., checking
pulse or lifting).
ll Decontaminate hands after contact with body fluids or excretions, mucous
membranes, nonintact skin, or wounds.
ll Decontaminate hands before applying sterile gloves or inserting a central
intravascular catheter.
ll Decontaminate hands before inserting indwelling urinary catheters or other
invasive devices not requiring surgical procedures.
ll Decontaminate hands after removing gloves.
ll Decontaminate hands before caring for patients with severe neutropenia or
severe immunosuppression.
ll Decontaminate hands after contact with inanimate objects in the immediate vicinity of the patient.
ll
RECOMMENDED TECHNIQUES FOR HAND HYGIENE
When using a waterless antiseptic agent, apply enough of the product to
cover all surfaces of the hands and fingers, and rub hands together until
they are dry. Each manufacturer has guidelines for the volume to be used; in
general, enough should be applied so that it takes 15 to 25 seconds to dry.
ll When using a nonantimicrobial or antimicrobial soap, wet hands, apply 3
to 5 mL of solution to the hands, and rub for at least 15 seconds. Be sure to
cover all surfaces of the hands and fingers. Rinse hands with warm water,
and dry thoroughly. Foot pedals, automatic faucets, or towel barriers should
be used to turn off the water.
ll
Adapted from Boyce JM, Pittet D, et al: Guidelines for hand hygiene in health-care settings:
recommendations of the Healthcare Infection Control Practices Advisory Committee and the
HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force, Infect Control Hosp Epidemiol
23(Suppl 12):S33-S40, 2002.
NICU, Neonatal intensive care unit.
respiratory tract care seem to be particularly associated
with contamination (Pittet et al, 1999). Organisms such
as RSV, S. aureus, and gram-negative bacilli are able to
survive on inanimate objects, so changing diapers or
holding an infant infected with one of these organisms,
and even touching items in the infant’s environment, may
result in contamination (Boyce et al, 2002; Goldmann,
1989; Hall, 2000).
Recommendations concerning indications and techniques for hand hygiene are summarized in Box 40-6
(Boyce et al, 2002; Garner and Favero, 1986; Larson,
1995). Current recommendations strongly support the
use of waterless alcohol-based preparations as the primary agents for hand hygiene, except when the hands
are soiled with organic material (Boyce et al, 2002). The
bottom line is that hands need to be cleaned before and
after every patient contact. Until there is 100% compliance with this simple intervention, HAIs will continue to
be a problem.
CHAPTER 40 Health Care–Acquired Infections in the Nursery
561
TABLE 40-3 Antimicrobial Spectrum and Characteristics of Hand Hygiene Antiseptic Agents
Group*
GramPositive
Bacteria
GramNegative
Bacteria
Mycobacteria
Fungi
Viruses
Alcohols
+++
+++
+++
+++
+++
Fast
Optimum concentration 60%90%, no persistent activity
Chlorhexidine (2%
and 4% aqueous)
+++
+++
+
+
+++
Intermediate
Persistent activity, rare allergic reactions
Iodine compounds
+++
+++
+++
++
+++
Intermediate
Causes skin burns, usually too
irritating for hand hygiene
Iodophors
+++
+++
+
++
++
Intermediate
Less irritating than iodine
Phenol derivatives
+++
+
+
+
+
Intermediate
Activity neutralized by nonionic surfactants
Triclosan
+++
++
+
−
+++
Intermediate
Acceptability varies
Quaternary ammonium compounds
+
++
−
−
+
Slow
Used only in combination with
alcohols, ecologic concerns
Speed of
Action
Comments
Reprinted with permission from Boyce JM, Pittet D, et al: Guidelines for hand hygiene in health-care settings: recommendations of the Healthcare Infection Control Practices Advisory Committee and the HICPAC/SHEA/APIC/IDSA Hand Hygiene Task Force, Infect Control Hosp Epidemiol 23(Suppl 12):S33-S40, 2002.
+++, Excellent; ++, good, but does not include the entire bacterial spectrum; +, fair; −, no activity or not sufficient.
*Hexachlorophene is not included because it is no longer an accepted ingredient of hand disinfection.
Preparations for Hand Hygiene
Washing the hands with soap causes suspension and
mechanical removal of microorganisms and dirt from the
hands. Hand disinfection refers to the same process, but
with the use of an antimicrobial product to kill or inhibit
microorganisms. Table 40-3 compares properties of the
various hand hygiene agents.
The cleansing activity of plain soap results from its
detergent properties (Boyce et al, 2002). Hand washing with soap alone reduces bacterial colonization of the
hands, but has no significant antimicrobial activity and is
ineffective at removing pathogenic flora. Soaps containing antimicrobial agents are typically used in the NICU
environment.
The antimicrobial activity of alcohol stems from its
ability to denature proteins (Boyce et al, 2002). Because
proteins are not readily denatured in the absence of
water, solutions containing 50% to 80% alcohol are most
effective (Boyce, 2000; Boyce et al, 2002). Alcohol has
a rapid onset of action and reduces bacterial colonization, but has no residual activity. The efficacy of alcoholbased products is influenced by the type of alcohol used,
concentration, contact time, volume used, and whether
the hands are wet when the product is applied (Boyce
et al, 2002; Mackintosh and Hoffman, 1984). When used
in adequate amounts, alcohol is usually more effective
than other hand hygiene products (Boyce, 2000; Boyce
et al, 2002).
Chlorhexidine gluconate is a cationic bis-biguanide
whose antimicrobial activity is caused by attachment and
disruption of cytoplasmic membranes (Boyce et al, 2002).
The onset of action is slower than the alcohol-based
preparations; however, its major benefit is the persistent
antimicrobial activity, which may last up to 6 hours after
application (Boyce et al, 2002; Larson, 1995). Varying percentages of this product have been added to other hand
hygiene preparations, especially the alcohol-based preparations, to confer greater residual activity.
Hexachlorophene is a bisphenol compound with bacteriostatic properties. Its activity is due to its ability to inactivate essential enzymes systems. It has good activity against
S. aureus, but weak activity against gram-negative bacteria,
fungi, and Mycobacterium tuberculosis. Hexachlorophene
also has residual activity. Hexachlorophene was used for
routine bathing of newborn infants until 1972, when
the U.S. Food and Drug Administration (FDA) warned
against its use because of an increased occurrence of cystic
degeneration of the cerebral white matter in infants who
had been bathed in the 3% solution (Shuman et al, 1975).
This product should be considered only in term infants
during a severe outbreak of S. aureus. Most experts recommend diluting the material 1:4 with water to decrease the
risk of systemic absorption.
Iodine and iodophors penetrate the cell walls of organisms, impairing protein synthesis and altering the cellular membranes (Boyce et al, 2002). The amount of iodine
present determines the level of antimicrobial activity.
Combining iodine with polymers (i.e., povidone or poloxamer) increases the solubility, promotes sustained release
of iodine, and decreases skin irritation. The activity of this
product is affected by pH, exposure time, temperature, the
presence of organic (blood or sputum) or inorganic material, and the concentration of iodine.
Compliance
Despite the fact that the benefits of hand washing have been
reported since the nineteenth century, compliance with
hand-washing protocols remains unacceptably low. The
overall compliance rate is approximately 40% (Pittet, 2000).
Reported barriers to compliance with hand hygiene recommendations include skin irritation, poor accessibility of sinks
or cleansing agents, greater priority for patient needs, insufficient time, heavy workload and understaffing, and lack of
information (Box 40-7). A common misconception is that
using gloves obviates the need for adequate hand hygiene.
562
PART IX Immunology and Infections
BOX 40-7 F
actors Influencing Compliance
With Hand Hygiene Practices
OBSERVED RISK FACTORS FOR POOR ADHERENCE
TO RECOMMENDED PRACTICES
ll Physician status (rather than a nurse)
ll Nursing assistant status (rather than a nurse)
ll Working in an intensive care unit
ll Working during the week compared with the weekend
ll Wearing gowns and gloves
ll Automated sink faucet
ll Activities with high risk of contamination
ll High numbers of indications for hand hygiene per hour of patient care
SELF-REPORTED FACTORS FOR POOR ADHERENCE
TO RECOMMENDED PRACTICES
ll Agents are irritating or drying
ll Inconvenient location of sinks and supplies
ll Inadequate supplies (e.g., soap, paper)
ll Too busy
ll Patient needs take priority
ll Perceived low risk of acquiring infection
ll Gloves obviate the need for hand hygiene
ll Lack of knowledge of guidelines and recommendations
ll Disagreement with recommendations
Data from Pittet D: Improving compliance with hand hygiene in hospitals, Infect Control Hosp
Epidemiol 21:381-386, 2000; and Boyce JM: Using alcohol for hand antiseptics: dispelling old
myths, Infect Control Hosp Epidemiol 21:438-441, 2000.
Leakage and contamination of gloves have been reported
(Boyce et al, 2002; Larson, 1995). Disposable single-use
gloves should be removed after each patient encounter, and
hands should be washed before and after their use.
Hand hygiene is extremely cost effective. The additional
hospital charges associated with a single HAI almost equal
the yearly hand hygiene budget. Pittet et al (2000) estimated the cost of a hand hygiene intervention program
to be less than $57,000 per year. Assuming that only 25%
of the observed decrease in infections was attributable to
improved hand hygiene practices, they estimated a savings
of $2,100 for every infection averted.
PREVENTION OF CENTRAL LINE–ASSOCIATED
BLOODSTREAM INFECTIONS
The care and maintenance of central venous catheters may
affect the risk of catheter-related bloodstream infection.
Before insertion of an intravascular device, attempted sterilization of the skin insertion site is of utmost importance.
The recommended technique for skin antisepsis is to use
two consecutive 10-second applications or a single 30-second application of the selected antibacterial agent. Sterile
water rather than alcohol should be used to remove antiseptics from the skin. Povidone-iodine (10%) solution is
commonly used for skin antisepsis; however, data suggest
that chlorhexidine gluconate is more effective at decreasing skin colonization and subsequent CLABSIs in adults,
as well as colonization of peripheral intravenous catheters
in neonates (Darmstadt and Dinulos, 2000; Garland et al,
1995).
Hub colonization and repeated entry into the line for
administration of medications or collection of specimens
for laboratory studies are both associated with an increased
risk for infection. Salzman et al (1993) conducted a prospective study in which surveillance cultures of catheter
hubs were performed three times per week, and blood cultures and hub cultures were conducted for any suspected
episode of sepsis. These investigators found that 54% of
28 episodes of CLABSI were preceded by or coincided
with colonization of the catheter hub with the same pathogen. This mechanism has been confirmed in the neonatal
population by Garland et al (2008). Mahieu et al (2001b)
reported that catheter manipulation for blood sampling
and fluid or tubing changes raised the risk of colonization.
Despite the lack of large randomized controlled trials, the
available data suggest that the use of heparin in intravenous fluids is associated with a lower risk of bacterial colonization and thrombosis (Appelgren et al, 1996; Mahieu et
al, 2001b). It is hypothesized that heparin prevents bacteria from adhering to the catheter and that the preservatives
in heparin preparations have some limited antibacterial
properties.
Various products have been designed to decrease
the risk of catheter-related infection in adult patients.
Although data on their use are promising, many of the
currently available products are not specifically designed
for the neonate, and their safety and efficacy have not
been tested in this patient population. Antiseptic- or
antibiotic-impregnated catheters have been shown to
significantly decrease the risk of CLABSI in adults
(Elliott, 2000; Marin et al, 2000; Pai et al, 2000; Tennenberg et al, 1997; Veenstra et al, 1999). Pierce et al (2000)
reported that pediatric patients randomly assigned to the
use of a heparin-bonded device had a lower incidence
of CLABSI than controls (4% versus 33%; p < 0.005).
Studies are in progress to evaluate the efficacy of antibiotic lock therapy in neonates. Johnson et al (1994)
treated a small group of pediatric patients with CLABSIs
with antibiotic lock therapy for 10 days. They reported
that 10 of 12 catheters were salvaged. There are limited
data describing the use of antibiotic lock therapy in neonates. Garland et al (2002) randomly assigned 82 VLBW
infants to saline control or vancomycin antibiotic lock
therapy of catheters in an effort to prevent CLABSI.
These researchers found a significantly lower incidence
density of catheter-related infections, no evidence of
toxicity, and no cases of VRE in the neonates receiving
antibiotic lock therapy.
UMBILICAL CORD CARE
Umbilical cord infections remain a significant cause of
neonatal mortality in developing nations, although not
in the United States (Zupan and Garner, 2000). In clinical practice, there are numerous approaches to umbilical cord care in the healthy term infant because the
available data are insufficient to recommend the use of
a specific agent or regimen (Zupan and Garner, 2000).
Most authorities agree that the umbilical cord should
be separated from the placenta with the use of good
aseptic technique. Subsequently, some experts recommend “natural drying,” but others support the use of
an antiseptic agent such as alcohol, silver sulfadiazine,
chlorhexidine, triple dye, or gentian violet (Darmstadt
CHAPTER 40 Health Care–Acquired Infections in the Nursery
and Dinulos, 2000; Hsu et al, 1999; Pezzati et al, 2002;
Zupan and Garner, 2000). Hexachlorophene and iodine
are used sparingly because of concerns about their systemic absorption and toxicity. Studies have shown that
antiseptic products decrease umbilical cord colonization.
Unfortunately, this decrease has not clearly resulted in a
lower incidence of umbilical cord infections. In a metaanalysis that included “no intervention” as an alternative, Zupan and Garner (2000) were unable to determine
which regimen was superior. In general, they found that
time to cord separation was prolonged when antiseptics
were used, but there were no significant differences in
the incidence of infection or death with the use of a particular agent for cord care.
SKIN CARE
The skin of VLBW preterm infants is immature and functions as an ineffective barrier to prevent transepidermal
loss of water and invasion of bacteria. The stratum corneum (the outermost layer of skin) has both mechanical
and chemical properties that decrease the risk of infection
(Darmstadt and Dinulos, 2000). This layer of skin matures
at approximately 32 weeks’ gestation. In a prematurely
born neonate, the maturation process is accelerated and
is usually complete by 2 to 4 weeks after birth (Darmstadt
and Dinulos, 2000).
Unfortunately, there is no consensus on the most effective skin care practices for VLBW infants (Baker et al,
1999; Munson et al, 1999). Neonatologists had hoped
that the application of a topical emollient would protect
the developing epidermal layer, reduce the risk of infection, and prevent transepidermal water loss. Several studies have documented that such a practice decreases water
loss; however, there was no significant reduction in HAI
in VLBW infants randomly assigned to receive routine application of one emollient product (Aquaphor) in
a study performed by Edwards et al (2001). Infants randomly assigned to emollient therapy had more nosocomial
bloodstream infections, particularly with CONS. This difference was most evident in infants with birthweights of
501 to 750 g.
The efficacy of other topical agents warrants further
study. Efforts to prevent traumatic injury to the skin of
VLBW infants include the use of transparent dressing over
bony prominences, semipermeable barriers between the
skin and adhesive tape, and water-activated electrodes and
the avoidance of agents such as tincture of benzoin, Mastisol, and adhesive removers (Adams-Chapman and Stoll,
2002; Darmstadt and Dinulos, 2000; Hoath and Narendran, 2000).
MANAGEMENT OF HEALTH
CARE–ACQUIRED INFECTIONS
CATHETER-RELATED BLOODSTREAM
INFECTIONS
Management of neonates with CLABSIs is problematic
because of the limited intravenous access in most patients,
as well as the lack of consensus among clinicians. Current
recommendations for adults regarding catheter removal
563
with associated CLABSI suggest the removal of nontunneled central venous catheters associated with bacteremia
or fungemia, unless the pathogen is CONS (Mermel et al,
2001). A patient with a catheter-related infection caused
by CONS should undergo catheter removal if culture
results are persistently positive or if the patient’s condition is unstable (Benjamin et al, 2001; Karlowicz et al,
2000).
Benjamin et al (2001) retrospectively reviewed data
on infants with CLABSIs and compared outcomes in
patients in whom catheters were removed at the onset
of infection with those in whom catheters remained in
place. Forty-six percent (59 of 128) of infants in whom
catheter sterilization was attempted had complications,
compared with 8% (2 of 25) of those in whom catheters
were removed. In particular, infants with gram-negative infections were more likely to have complications
if their catheters remained in place. A study of infants
with CLABSIs caused by CONS found no difference
in the complication or mortality rate in patients in
whom removal of the catheter was delayed (Karlowicz
et al, 2000). However, patients with a CONS infection
related to their central line were more likely to have persistently positive culture results when their lines were
not removed with the first positive culture (43% versus
13% for immediate catheter removal). The attempt to
retain the catheter was never successful if culture results
remained positive for more than 4 days. Additional prospective randomized trials are needed to validate these
observational data.
ANTIBIOTIC AND ADJUNCTIVE THERAPIES
Antibiotic therapy that is effective against a cultureproven or suspected pathogen is the primary treatment
for HAIs. As a general rule, antibiotic choice should initially cover a broad spectrum of pathogens and should
then be narrowed as soon as possible to cover the specific bacteria identified once sensitivities are known, or
it should be discontinued if infection is not proved and
is not likely. One should consider coverage for Pseudomonas spp. or other resistant gram-negative organisms
in patients with a rapid clinical deterioration (Karlowicz
et al, 2000; Stoll et al, 2002a). However, empiric broadspectrum antibiotic use should be limited as much as
possible to avoid complications of resistant bacterial and
fungal infections.
A metaanalysis of the prophylactic use of intravenous
immunoglobulin (IVIG) in preterm neonates found only a
3% reduction in HAI and no reduction in mortality (Modi
and Carr, 2000; Ohlsson and Lacy, 2001a). The benefit
in patients with culture-proven sepsis remains unclear,
and IVIG is therefore not recommended for routine use
in such patients (Modi and Carr, 2000). Some authorities speculate that the benefit of IVIG would be greater
if products containing high concentrations of specific
antibodies against pathogens frequently responsible for
neonatal infections were developed (Hill, 2000; Ohlsson
and Lacy, 2001b; Weisman et al, 1994). Hemopoietic
colony-stimulating factors (granulocyte and granulocytemacrophage) are effective in raising the neutrophil count,
but have not consistently decreased HAI rates or mortality
564
PART IX Immunology and Infections
(Carr et al, 2003; Modi and Carr, 2000). Studies are being
conducted in adults to evaluate the efficacy of administering various cytokine preparations known to modulate the
inflammatory response.
CONCLUSION
Interventions to reduce HAI are urgently needed. Infants
with HAIs have significantly longer hospital stays (79 versus 60 days; p < 0.001) and higher hospital costs (Gray et
al, 1995; Mahieu et al, 2001a; Pittet et al, 1994; Stoll et
al, 2002a). The higher costs are primarily caused by daily
hospital charges and pharmaceutical fees (Mahieu et al,
2001a). The attributable cost of a nosocomial infection has
been estimated to be approximately $40,000 per adult case
and $1200 per neonatal case (Mahieu et al, 2001a; Pittet et
al, 1994). The magnitude of these costs is significant, especially when considering the growing number of surviving
VLBW infants, who are at the highest risk for acquiring
an HAI. Moreover, infants who experience an HAI are
significantly more likely to die than are those who remain
uninfected (Stoll et al, 1996, 2002a).
A variety of new therapeutic alternatives is currently
being investigated. Clinicians must continue to focus
on developing effective prevention strategies, including strict hand-washing policies, minimal use of invasive
devices, promotion of enteral nutrition surveillance of
infection patterns, and education for the nursery staff
members. Quality improvement approaches can be
effective in implementing the necessary practices within
a given unit.
SUGGESTED READINGS
Bloom BT, Craddock A, Delmore PM, et al: Reducing acquired infections in the
NICU: observing and implementing meaningful differences in process between
high and low acquired infection rate centers, J Perinatol 23:489-492, 2003.
Clerihew L, Austin N, McGuire: Prophylactic systemic antifungal agents to
prevent mortality and morbidity in very low birth weight infants, Cochrane
Database Syst Rev 4:CD003850, 2007.
Curtis C, Shetty N: Recent trends and prevention of infection in the neonatal
intensive care unit, Curr Opin Infect Dis 21:350-356, 2008.
Kilbride HW, Wirtschafter DD, Powers RJ, et al: Implementation of evidencebased potentially better practices to decrease nosocomial infections, Pediatrics
111(4 Pt 2):e519-e533, 2003.
Meissner HC, Long SS: American Academy of Pediatrics Committee on Infectious
Diseases and Committee on Fetus and Newborn: revised indications for the
use of palivizumab and respiratory syncytial virus immune globulin intravenous
for the prevention of respiratory syncytial virus infections, Pediatrics 112:14471452, 2003.
Perlman SE, Saiman L, Larson EL: Risk factors for late-onset health care-associated bloodstream infections in patients in neonatal intensive care units, Am J
Infect Control 35:177-182, 2007.
Parellada JA, Moise AA, Hegemier S, et al: Percutaneous central catheters and
peripheral intravenous catheters have similar infection rates in very low birth
weight infants, J Perinatol 19:251-254, 1999.
Schulman J, Wirtschafter DD, Kurtin P: Neonatal intensive care unit collaboration
to decrease hospital acquired bloodstream infections: from comparative performance reports to improvement networks, Pediatr Clin N Am 56:865-892, 2009.
Stoll BJ, Adams-Chapman I, et al: Abnormal neurodevelopmental outcome of
ELBW infants with infection, Pediatr Res Suppl 53:2212, 2003.
Stoll BJ, Hansen NI, Adams-Chapman I, et al: National Institute of Child Health
and Human Development Neonatal Research Network: neurodevelopmental
and growth impairment among extremely low-birth-weight infants with neonatal infection, J Am Med Assoc 292:2357-2365, 2004.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
41
Fungal Infections in the Neonatal
Intensive Care Unit
Margaret K. Hostetter
EPIDEMIOLOGY
Invasive fungal infection occurs in approximately 1% to
2% of all infants admitted to U.S. neonatal intensive care
units (NICUs) (Stoll et al, 1996), and the incidence rises
dramatically with decreasing gestational age. Among the
fungi, Candida spp. is the dominant pathogen, with infections evenly distributed among C. albicans and non–C. albicans species (C. parapsilosis, C. orthopsilosis, C. metapsilosis,
C. glabrata, C. guilliermondii, C. krusei, and C. lusitaniae).
Candida spp. are the third most common cause of lateonset sepsis in the NICU, with a fatality rate more than
sevenfold greater than that of Staphylococcus epidermidis, the
most common pathogen (Benjamin et al, 2000; Saiman
et al, 2001). Other fungi encountered include Aspergillus
fumigatus, A. flavus, Malassezia furfur, M. pachydermatis.
The yeast Cryptococcus neoformans and the fungi Histoplasma
capsulatum, Blastomyces dermatitidis, and Coccidioides immitis
are rarely if ever seen in the NICU.
The most important risk factor for fungal infection is
gestational age. In a survey of 2847 infants from six different nurseries, the incidence of candidemia in infants
weighing less than 800 g (7.55%) was 25-fold that of the
infant weighing more than 1500 g (Figure 41-1) (Saiman
et al, 2000). This latter group acquires blood-borne candidal infection in association with congenital anomalies,
especially those of the gastrointestinal tract (Rabalais et al,
1996). Candidemia carries a mortality rate exceeding 25%
in most studies (Chapman and Faix 2000; Weese-Mayer
et al, 1987).
Colonization with ubiquitous fungal species occurs in
at least 25% of very low-birthweight infants (Baley et al,
1986), and both the amount of Candida spp. in the gastrointestinal tract (Pappu-Katikaneni et al, 1990) and colonization at sites such as endotracheal tubes (Rowen et al,
1994) have been correlated with increased risk of invasive
disease caused by Candida spp. Prospective studies correlating colonization by other fungal genera (e.g., Aspergillus,
Malassezia spp.) with risk of invasive disease have not been
done.
Apart from colonization and gestational age, other host
factors that contribute to the susceptibility of the infant
in the NICU to fungal infection include 5-minute Apgar
scores less than 5, and an age-dependent immunocompromised state ascribable to reduced numbers of T cells,
impaired phagocyte number and function, and reduced levels of complement (Marodi et al, 1994; Rebuck et al, 1995;
Witek-Janusek et al, 2002; Zach and Hostetter, 1989). Concomitants of nursery care that are thought to increase the
risk of fungal infections include length of stay greater than
1 week, indwelling central venous catheters, abdominal surgery, parenteral nutrition, intralipids, H2 (histamine) receptor antagonists, endotracheal intubation, and prolonged
use of broad-spectrum antimicrobials, especially third-
generation cephalosporins (Cotten et al, 2006; Saiman et
al, 2001; Saiman et al, 2000). Other centers have identified
associations with systemic steroids and catecholamine infusions in retrospective studies and with topical petrolatum in
a prospective case-control study (Benjamin et al, 2000; Botas
et al, 1995; Campbell et al, 2000).
Interestingly, a number of variables appear not to associate with candidal colonization, including the use of
antibiotics in the mother, premature rupture of the membranes, the infant’s gender, the use of antimicrobial agents
other than third-generation cephalosporins in the infant,
surgical procedures, or frequency of intubation (Saiman
et al, 2001). Although ≈5% of NICU staff members carry
C. albicans on the hands and 19% carry C. parapsilosis, there
is no correlation with site-specific rates of infant colonization (Saiman et al, 2001).
This chapter will place major emphasis on infections
caused by Candida spp. and other fungi, as well as the
approach to diagnosis, treatment, and management of
infants with fungal infection.
INFECTIONS CAUSED BY CANDIDA
SPECIES
CONGENITAL CANDIDIASIS
Appearing within the first 24 hours of life in both full-term
and premature infants, congenital candidiasis, a very rare
entity, manifests as a deeply erythematous skin rash in the
setting of pronounced neutrophilia, with white blood cell
counts often rising to 50,000 or more, and infrequently
Candida spp. funisitis (see Local Infections, later). In the fullterm infant, there are no invasive consequences, and desquamation typically ensues within 2 to 3 days. In contrast, the
condition is life threatening in the premature infant (Dvorak
and Gavaller, 1966; Johnson et al, 1981) and is distinguished
by a pustular rash, hazy infiltrates reminiscent of respiratory distress syndrome on chest radiograph, and frequently
positive blood cultures. The premature infant is thought to
acquire the organism from inhaling infected amniotic fluid.
Diagnosis in both premature and full-term infants
requires the visualization of the organism on Gram stain
from a bullous lesion or an opened pustule. On rare
occasions, the placenta has yielded the diagnosis. Treatment for the full-term infant requires only the full-body
application of topical antifungal creams containing either
nystatin or azoles, such as miconazole or clotrimazole. In
the premature infant, initiating parenteral amphotericin B
deoxycholate at a dose of 1 to 1.5 mg/kg is mandatory,
but respiratory involvement typically heralds death despite
anti-fungal therapy.
565
566
PART IX Immunology and Infections
8
Incidence (%)
7
6
5
4
3
2
1
0
800
801–1000
1001–1500
1500
Birthweight in grams
FIGURE 41-1 Incidence of candidemia related to birthweight in
grams. (Data from Saiman L et al: Risk factors for candidemia in Neonatal
Intensive Care Unit patients. The National Epidemiology of Mycosis Survey
study group, Pediatr Infect Dis J 19:319-324, 2000.)
LOCAL INFECTIONS WITH CANDIDA SPECIES
Diaper Dermatitis
Diaper dermatitis manifests as an erythematous, erosive
dermatitis of the perineal region, typically with pustular
satellite lesions beyond the borders of the rash. Predisposing factors include systemic antibiotics, glucosuria, and
wet diapers. Care must be taken to differentiate this tractable condition from invasive fungal dermatitis (see Invasive Fungal Dermatitis, later). The infection responds well
to topical antifungal ointments.
Funisitis
Infection of the umbilical cord with Candida spp., although
rare, is an indicator of chorioamniotitis and carries a poor
prognosis, especially in the premature infant (Qureshi
et al, 1998). A minority (16%) of infants in one study had
associated congenital candidiasis. Intrauterine contraceptive devices or cervical cerclage were reported in 16% of
the mothers.
Urinary Tract Infection
Isolation of Candida spp. from a catheterized specimen or
suprapubic aspiration, as opposed to a bagged sample, is
a reliable indicator of infection, although asymptomatic
colonization of urinary catheters, stents, or nephrostomy
tubes can be difficult to distinguish from true infection
(Lundstrom and Sobel, 2001).
The presence of candiduria in infants in the NICU is
associated with renal candidiasis—the latter manifested by
cortical abscesses or fungal mycelia in the collecting system (“fungus balls”)—almost half the time and may be a
cause of frank obstruction (Bryant et al, 1999). Therefore,
in contrast to older children or adults, the finding of candiduria in an infant in the NICU should at least prompt
blood cultures and renal imaging. If blood cultures prove
to be positive, a full evaluation for disseminated candidiasis
should be undertaken (see Systemic Infection, later).
Because of the high prevalence of associated upper tract
disease, imaging of the kidneys by ultrasonography should
occur after isolation of the organism from a sterile urine
specimen. Approximately one half of the patients who
eventually develop upper tract manifestations will display
them on the first ultrasound (Bryant et al, 1999). Therefore follow-up imaging is recommended both to ensure
the clearance of fungal mycelia, if present, and to monitor
for later development of this complication. Unfortunately,
no standard interval for monitoring has been proposed;
however, in the infant with persistent funguria or candiduria, a single negative ultrasound should not be considered definitive.
Removal of a colonized urinary catheter may suffice for
treatment in a patient without pyuria or systemic symptoms. Disease confined to the lower tract is best addressed
with azoles (e.g., fluconazole, 4 to 6 mg/kg/day). Upper
tract disease requires parenteral amphotericin B in systemic doses (1 to 1.5 mg/kg/day). Liposomal amphotericin
B may not be an acceptable alternative. The particles in at
least one liposomal preparation (Abelcet) appear to be too
large to penetrate the adult kidney (Hell et al, 1999), and
this author has seen a premature newborn whose persistent
candiduria failed to resolve until treatment was changed to
amphotericin B deoxycholate.
PERITONITIS
Candida spp. peritonitis typically develops as a consequence
of bowel perforation or rarely as a complication of peritoneal dialysis. In the former situation, multiple organisms
such as gram-negative rods and enterococci may also be
involved, and the neonate is at risk for sepsis with any one
of them (Johnson et al, 1980). Peritonitis associated with
a peritoneal dialysis catheter usually occurs as an isolated
process, and the outcome is much better.
Spontaneous intestinal perforation associated with
Candida spp. peritonitis with or without sepsis has been
described within 7 to 10 days of birth in infants weighing
less than 1000 g, typically in the absence of necrotizing
enterocolitis (Adderson et al, 1998; Holland et al, 2003;
Meyer et al, 1991; Mintz and Applebaum, 1993). Hallmark
clinical findings include bluish discoloration of the abdomen and a gasless pattern on abdominal film. A substantial
proportion of these infants will have systemic candidiasis,
although Staphylococcus epidermidis can also be seen. In a
small study of seven patients (Holland et al, 2003), deficiency of the muscularis propria was found in six.
Diagnosis requires visualization of the organism on a
Gram stain of peritoneal fluid sterilely obtained or culture
of the organism from the same source. Isolation of Candida spp. from the peritoneal fluid should always prompt a
search for bowel perforation, by either radiology or surgical exploration, depending upon the clinical circumstances.
Treatment of Candida spp. peritonitis caused by necrotizing enterocolitis or bowel perforation requires surgical
evaluation, supportive therapy, and direct address of all
contaminating microorganisms in the peritoneal fluid and
the bloodstream. The typical regimen may include ampicillin and an aminoglycoside for enterococci and gramnegative rods, clindamycin for anaerobes, and systemic
antifungal therapy, most likely with amphotericin B. The
isolation of Candida spp. from peritoneal dialysate can be
treated with removal of the catheter and a short course
(7 to 10 days) of amphotericin B therapy in a dose of
CHAPTER 41 Fungal Infections in the Neonatal Intensive Care Unit
0.3 to 0.5 mg/kg/day. The catheter can typically be reinserted within 24 to 48 hours, once the Gram stain is free
of yeast cells.
SYSTEMIC INFECTION
CANDIDEMIA ASSOCIATED WITH CENTRAL
VENOUS CATHETERS
The association between prematurity and blood-borne
candidal infections has been recognized for 25 years (Baley
et al, 1984; Johnson et al, 1984). Over this same period
of time, the incidence of candidemia has escalated from
25 to 123 cases per 10,000 NICU admissions (Kossoff
et al, 1998; Saiman et al, 2000). The median time of onset is
approximately 30 days of age (Baley et al, 1984). In a large
multicenter study, colonization of the gastrointestinal tract
preceded candidemia in 43% of cases (Saiman et al, 2000).
A variety of nonspecific clinical findings may be associated with this presentation of candidal disease, including
respiratory decompensation, feeding intolerance, temperature instability, or mild thrombocytopenia. It is unclear
whether the latter manifestation relates more to the use of
heparin in vascular catheters or to the presence of Candida
spp. in the blood stream.
Isolation of Candida spp. from a blood culture should
never be regarded as a contaminant and should prompt
an immediate search for evidence of dissemination, which
occurs in approximately 10% of premature newborns with
candidemia (Noyola et al, 2001; Patriquin et al, 1980).
A thorough evaluation includes ophthalmologic examination and ultrasonography of the heart, venous system,
and abdomen. When lumbar puncture is performed, 10%
to 50% of infants with candidemia may have associated
meningitis (Benjamin et al, 2006; Faix, 1984); in one prospective study, almost 50% of extremely low-birthweight
(ELBW) infants with Candida spp. meningitis (13/27) had
negative blood cultures (Benjamin et al, 2006).
Numerous studies have shown that central venous catheters should be removed within 24 hours after identification of yeasts in the blood culture (Karlowicz et al, 2000);
in particular, removal of the central venous catheter within
3 days is associated with a significantly shorter median
duration of candidemia (3 versus 6 days) and a reduced
mortality rate (0% versus 39%). In at least one study of
candidemia, delayed removal of central venous catheters
was associated with neurodevelopmental impairment at 18
to 22 months (Benjamin et al, 2006). Many experts recommend routine echocardiograms for patients with catheterassociated candidemia to look for thrombi before removal
of the catheter. However, even with the prompt removal
of the catheter and the institution of appropriate antifungal therapy, a substantial proportion of infants may exhibit
prolonged candidemia lasting 1 to 3 weeks (Chapman and
Faix, 2000).
DISSEMINATED CANDIDIASIS
Mortality rates approach 30% for this dreaded complication of candidal infection. C. albicans is the leading
pathogen. Organ involvement is most common in the
vascular tree at catheter sites (15.2%), followed by the
567
kidneys (7.7%) (Noyola et al, 2001; Patriquin et al, 1980).
Eye involvement occurs in approximately 6% of infants.
Thrombi within the vascular bed may be particularly difficult to eradicate with antifungal therapy; infants with
right atrial thrombi may benefit from atriotomy (Foker
et al, 1984). Other sites less frequently involved include
the liver, spleen, and skeletal system. In infection of the
bones and joints in premature newborns, Candida spp. are
typically the second most likely pathogen, preceded by
Staphylococcus aureus (Ho et al, 1989).
ANTI-FUNGAL THERAPY FOR SYSTEMIC
INFECTION
As is true of most medications used in the neonatal ICU,
dosing recommendations for antifungal therapies have
not undergone rigorous testing in this patient population.
With that caveat in mind, it is important to discuss practice guidelines for this difficult clinical problem. Amphotericin B is the standard antifungal therapy for treatment
of systemic neonatal fungal infection. The drug binds to
ergosterol in the membrane of fungi, facilitating membrane leakage. Rapid institution of parenteral amphotericin B deoxycholate in doses of 1.0 to 1.5 mg/kg/day, given
by intravenous infusion over 2 to 6 hours, is the therapy
of choice for systemic infection, including catheter-associated candidemia and disseminated candidiasis. No more
than 24 hours should elapse before the infant is receiving
a dose of 0.7 to 1.0 mg/kg/day. Some experts recommend
instituting empiric antifungal therapy in acutely thrombocytopenic ELBW infants (Benjamin et al, 2003). Dose
adjustment for renal dysfunction is necessary only if serum
creatinine increases significantly during therapy. Amphotericin B has poor cerebrospinal fluid penetration; therefore
accompanying meningitis should prompt the addition of
5-flucytosine in doses of approximately 12.5 to 37.5 mg/kg
per dose given by mouth every 6 hours (in patients with
normal renal function). Peak serum concentrations of this
drug should be kept between 40 and 60 μg/mL to avoid
bone marrow suppression or hepatotoxicity. Fluconazole
may also be added to amphotericin B for meningeal penetration, if 5-flucytosine is unavailable or cannot be used.
Infants being treated with amphotericin B who experience a twofold rise in creatinine, which is evidence of
renal tubular compromise or renal tubular acidosis, may
benefit from use treatment with one of the liposomal
amphotericin preparations. AmBisome has been used in
neonates at well-tolerated doses of 5 to 7 mg/kg per dose
every 24 hours intravenously, infused over 2 hours (JusterReicher et al, 2003; Weitkamp et al, 1998), although the
failure of many liposomal preparations to penetrate renal
parenchyma can militate against fungal clearance from this
organ.
With prompt removal of an offending central venous
catheter, and no evidence of dissemination, the duration
of therapy for catheter-associated candidemia is typically
10 to 14 days after the blood culture becomes negative
(Donowitz and Hendley, 1995). Disseminated candidiasis, including Candida spp. meningitis, requires at least 3
weeks or more of parenteral therapy; the course is typically
completed when all foci have been eradicated. Most infectious disease experts will use fungicidal doses of parenteral
568
PART IX Immunology and Infections
amphotericin B or a liposomal preparation for the entire
course. Azoles (such as fluconazole at 6 mg/kg intravenously at varying dosing intervals) are antifungal agents
that interfere with ergosterol synthesis by inhibiting C-14
alpha demethylase, a cytochrome P450 enzyme. Use of an
azole to complete the latter part of an antifungal course
must account for the inability of the infant’s immune system to compensate for the fungistatic activity of the azoles.
Azoles such as itraconazole and posaconazole are preferable to fluconazole for aspergillus and zygomycetes, but
no studies have been done to recommend neonatal dosing
guidelines.
Although not a first-line antifungal medication, echinocandins such as caspofungin, which interrupt biosynthesis
of β-(1,3)-D-glucan, an integral part of the fungal cell wall,
have also been used in doses of 1 to 2 mg/kg/day to treat
invasive candidal disease in the newborn in several case
reports; this drug is particularly helpful for species such
as Candida glabrata, Candida krusei or Candida lusitaniae,
which may have decreased susceptibility or de novo resistance to amphotericin B (Odio et al, 2004; Saez-Llorens
et al, 2009).
ANTI-FUNGAL PROPHYLAXIS
Five randomized, controlled trials comparing intravenous
fluconazole (3 mg/kg/day) with placebo or no treatment in
very low-birthweight (VLBW) or ELBW infants for 4 to
6 weeks (Cabrera et al, 2002; Kaufman et al, 2001; Kicklighter et al, 2001; Manzoni et al, 2007; Parikh et al, 2007)
met criteria for analysis in Cochrane reviews (Clerihew
et al, 2007). Kaufman and Manzoni reported significantly
lower incidence of invasive fungal infection, whereas there
was no difference in treated versus untreated infants in
other studies (Cabrera et al, 2002; Kicklighter et al, 2001;
Parikh et al, 2007). The only study to evaluate neurologic
outcomes found no difference in neurologic impairment at
16 months (Kaufman et al, 2001). Fluconazole prophylaxis
did not create a significant difference in the risk of death
before discharge in any of the five studies or in a metaanalysis (Clerihew et al, 2007). No study documented clinically
significant adverse effects of fluconazole or the emergence
of fluconazole resistance.
One study from a single center compared nonrandomized fluconazole prophylaxis in 2002 to 2006 with
an untreated, retrospective cohort (2000 to 2001) and
reported that invasive candidiasis decreased from 0.6% to
0.3%, although the proportion of invasive disease caused
by non–C. albicans species increased from 26% to 41% after
the introduction of fluconazole prophylaxis (Healy et al,
2008). Interestingly, fluconazole prophylaxis in this study
was extended to several infants with birthweights greater
than 1000 g, if risk factors (e.g., maternal HIV infection,
intestinal abnormalities) were present.
The finding that prophylactic fluconazole reduces the
incidence of invasive fungal infection must be interpreted
with caution (Clerihew et al, 2007, 2008):
1. The incidence of invasive fungal infection in the
placebo groups (Kaufman et al, 2001; Manzoni et al,
2007; Parikh et al, 2007) was significantly higher
(13% to 16%) than in other large cohort studies of
VLBW or ELBW infants in the United States (6%
to 7%) or the United Kingdom (1% to 2%).
2. Fluconazole prophylaxis may have impaired the
microbiologic isolation of some fungal species and
led to underdiagnosis of infection in the treatment
group.
3. Six years after the introduction of fluconazole prophylaxis, one study reported that non–C. albicans
species with relatively reduced susceptibility to the
azoles were the most common causes of invasive
fungal infection (Parikh et al, 2007). This study did
not detect a significant effect of fluconazole prophylaxis in reducing invasive candidal disease.
Based on these results and cautionary notes, infants
weighing less than 1000 g who receive third-generation
cephalosporins and central venous catheters may be the
best group to be evaluated for a statistically significant
benefit of fluconazole prophylaxis in preventing invasive
fungal infection, neurologic complications, and death
before hospital discharge. Study durations less than 6 years
may be insufficient to detect the emergence of fluconazole
resistance.
INFECTIONS ASCRIBABLE TO OTHER
FUNGI
INVASIVE FUNGAL DERMATITIS
Invasive fungal dermatitis typically manifests in the infant
weighing less than 1000 g who displays macerated or
bruised lesions that are contaminated with fungal species.
In the initial report, three of seven confirmed cases had
C. albicans, C. parapsilosis, C. tropicalis, Trichosporon beigelii,
Curvularia spp., or Aspergillus niger and A. fumigatus were
cultured from the remainder (Rowen et al, 1995). Among
cases considered “probable,” seven of eight had C. albicans.
Systemic complications including fungemia, meningitis,
or infection of the urinary tract occurred in four of seven
confirmed cases and seven of eight probable cases. More
cases than controls had postnatal steroids and prolonged
hyperglycemia. Disseminated infection occurred in 69%,
all ascribable to Candida spp.
Diagnosis requires a skin biopsy specimen demonstrating fungal invasion beyond the stratum corneum or a positive potassium hydroxide preparation of skin scrapings;
growth of the identical organism from an otherwise sterile
site (blood, cerebrospinal fluid, or urine obtained via supra
pubic aspiration) is confirmatory. Treatment requires
systemic doses of amphotericin B in the range of 0.7 to
1.0 mg/kg/day; in infants who do not develop systemic
infection, oral therapy with fluconazole or topical antifungal creams may suffice. Oral therapy is not advisable for
pathogens like Aspergillus spp., and repeated skin biopsies
may be necessary to define duration of therapy.
LINE INFECTIONS CAUSED BY LIPOPHILIC
ORGANISMS
The species Malassezia furfur and Malassezia pachydermatis are lipophilic organisms commonly carried on the skin,
even in patients without tinea versicolor (Marcon and
CHAPTER 41 Fungal Infections in the Neonatal Intensive Care Unit
Powell, 1992). Cutaneous colonization can infect hyperalimentation fluids or parenteral lipid formulations. Infants
typically exhibit mild but nonspecific signs: respiratory
decompensation, glucose intolerance, or thrombocytopenia (Dankner et al, 1987; Stuart and Lane, 1992). Diagnosis requires isolation of the organism from blood by
growth on fungal medium overlaid with olive oil, because
Malassezia spp. will not grow in the absence of lipids (Marcon et al, 1986). Removal of the intravascular catheter
usually suffices for therapy, although some experts recommend the addition of amphotericin B in dosages of 0.5 mg/
kg/day for 7 days.
MISCELLANEOUS FUNGAL INFECTIONS
Aspergillus species
Although rarely seen in neonates, systemic infection with
Aspergillus spp. suggests severe immune system compromise, such as DiGeorge Syndrome or myeloperoxidase deficiency (Chiang et al, 2000; Marcinkowski et al,
2000). Disseminated disease has occurred in premature
newborns without additional immunologic abnormalities (Rowen et al, 1992). Diagnosis requires isolation of
the fungus from a normally sterile tissue site or visualization by Gomori-methenamine silver stain on a biopsy
specimen of infected tissue. Of note, a commercially
available enzyme-linked immunosorbent assay for diagnosis of aspergillosis on serum specimens had an 83%
rate of false positive results in premature newborns (Siemann et al, 1998). Treatment requires systemic amphotericin B in doses of 1.0 to 1.5 mg/kg/day. Fungistatic
therapies such as the triazoles are not recommended for
aspergillosis.
569
Trichosporon beigelii
In a cluster of five neonatal cases of infection caused by
T. beigelii, a yeast found ubiquitously in soil, no common source was identified (Fisher et al, 1993). Two of
three premature infants infected with this organism died.
Resistance to achievable concentrations of amphotericin B
complicates therapy.
SUGGESTED READINGS
Benjamin DK Jr, Stoll BJ, Fanaroff AA, et al: Neonatal candidiasis among extremely low birth weight infants: risk factors, mortality rates, and neurodevelopmental outcomes at 18 to 22 months, Pediatrics 117:84-92, 2006.
Clerihew L, Austin N, McGuire W: Prophylactic systemic antifungal agents to
prevent mortality and morbidity in very low birth weight infants, Cochrane
Database Syst Rev 4:CD003850, 2007.
Cotten CM, McDonald S, Stoll B, et al: The association of third-generation cephalosporin use and invasive candidiasis in extremely low birth-weight infants,
Pediatrics 118:717-722, 2006.
Faix RG: Systemic Candida infections in infants in intensive care nurseries: high
incidence of central nervous system involvement, J Pediatr 105:616-622, 1984.
Juster-Reicher A, Flidel-Rimon O, Amitay M, et al: High-dose liposomal amphotericin B in the therapy of systemic candidiasis in neonates, Eur J Clin Microbiol
Infect Dis 22:603-607, 2003.
Karlowicz MG, Hashimoto LN, Kelly RE Jr, et al: Should central venous catheters
be removed as soon as candidemia is detected in neonates? Pediatrics 106:E63,
2000.
Noyola DE, Fernandez M, Moylett EH, et al: Ophthalmologic, visceral, and cardiac involvement in neonates with candidemia, Clin Infect Dis 32:1018-1023,
2001.
Odio CM, Araya R, Pinto LE, et al: Caspofungin therapy of neonates with invasive
candidiasis, Pediatr Infect Dis J 23:1093-1097, 2004.
Rowen JL, Atkins JT, Levy ML, et al: Invasive fungal dermatitis in the < or = 1000gram neonate, Pediatrics 95:682-687, 1995.
Saiman L, Ludington E, Pfaller M, et al: Risk factors for candidemia in Neonatal
Intensive Care Unit patients. The National Epidemiology of Mycosis Survey
study group, Pediatr Infect Dis J 19:319-324, 2000.
Saiman L, Ludington E, Dawson JD, et al: Risk factors for Candida species
colonization of neonatal intensive care unit patients, Pediatr Infect Dis J 20:
1119-1124, 2001.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
P A R T
X
Respiratory System
C H A P T E R
42
Lung Development: Embryology, Growth,
Maturation, and Developmental Biology
Maria Victoria Fraga and Susan Guttentag
The primary function of the lung is to accomplish exchange
of oxygen and carbon dioxide to accommodate the needs
of aerobic cellular respiration. The oxygen consumption of
the adult human ranges from 250 mL/min at rest to 5500
mL/min at peak exercise (Warburton et al, 2000). To
accommodate these metabolic needs, a large surface area,
and a thin alveolar-capillary membrane are required to
enable efficient diffusion of oxygen more so than carbon
dioxide. Ultimately the zone of gas exchange will attain a
surface area of 50 to 100 m2 and a volume of 2.5 to 3.0 L in
the adult human; therefore a primary goal of lung organogenesis is to expand the lung surface area to meet these
needs. A second goal of lung organogenesis is to minimize
the diffusing distance from alveolus to red blood cell, coordinating the development of an extensive capillary network
with a thin, expansive alveolar epithelial surface. A third
goal of lung development is production of a protective
aqueous barrier overlying the delicate alveolar epithelium
while mitigating the effects of the surface tension generated by this barrier, specifically alveolar collapse, through
the production of a surface active agent or surfactant.
The trachea, airways, and alveoli are in constant contact
with the external environment. Consequently with every
inhalation, epithelial surfaces encounter large numbers of
microorganisms and potentially toxic particles and gases.
Lung organogenesis must also incorporate mechanisms
for clearance of microorganisms and allergens that may
result in epithelial infection or injury. Similarly the lung
must defend against nonparticulate gases that are potentially harmful. Oxygen, although critical to cellular function, can be the source of harmful reactive oxygen species
and inhaled pollutants similarly require detoxification.
The appropriate development and maintenance of these
lung functions are critical to the health and survival of
newborn infants. This chapter focuses on developmental
aspects of each function that place the premature neonate
at increased risk for lung injury and disease.
KEY EVENTS IN LUNG DEVELOPMENT
Lung formation begins early in human gestation (by day
25), and growth extends well into childhood (Burri, 2006;
Kumar et al, 2005). Lung development can be organized
into stages (embryonic, pseudoglandular, canalicular, saccular, and alveolar), although the timing of these stages is
somewhat imprecise and considerable overlap can occur.
Figure 42-1 shows a timeline of fetal and postnatal lung
development.
DEVELOPMENT OF AIRWAYS AND GAS
EXCHANGE SURFACES
The initial phase of lung development, the embryonic
phase, is marked by the formation of the lung bud and
initial branching of presumptive airways. The lung bud is
first recognizable as a laryngotracheal groove of the ventral
foregut at 25 days’ gestation. Within a few days the groove
closes so that the only remaining lumenal attachment to
the foregut is in the region of the developing hypopharynx and larynx. The lung bud, consisting of epithelium and
surrounding mesenchyme, then begins the first of a series
of dichotomous divisions that give rise to the conducting
airways and five primordial lung lobes (two left and three
right). Tracheoesophageal fistulas, tracheal atresia, and
tracheal stenosis result from errors in separation of the
laryngotracheal groove, whereas failure of partitioning of
the lung bud can result in pulmonary agenesis, most typically of the right lung.
Branching continues into the pseudoglandular stage of
lung development. By 7 weeks’ gestation, the trachea and
the segmental and subsegmental bronchi are evident. By
the end of 16 weeks’ gestation, all bronchial divisions are
completed. It is important to remember that, although the
conducting airways will certainly enlarge as the fetus and
newborn grow (airway diameter and length increase twofold to threefold between birth and adulthood), large airway branching ceases after 16 weeks’ gestation.
Closure of the pleuroperitoneal folds is a critical event
of the pseudoglandular phase, reaching completion by
7 weeks’ gestation and resulting in separation of the thoracic cavity from the peritoneal cavity. Failure of pleuroperitoneal closure results in a diaphragmatic defect and
continuity between these cavities. At 10 weeks’ gestation,
when the midgut returns to the peritoneal cavity from the
umbilical cord, abdominal contents are free to pass into
the thoracic cavity and restrict the space into which the
lung grows. The resulting congenital diaphragmatic hernia leads to pulmonary hypoplasia of the lung ipsilateral to
the diaphragmatic defect as bowel and solid viscera migrate
into the thorax. Pulmonary hypoplasia can also extend to
the contralateral lung as the mediastinum shifts because of
accumulating abdominal viscera in the thorax.
571
572
PART X
Respiratory System
Lung growth
Late alveolarization
Microvascular maturation
Bulk alveolarization
Saccular
Canalicular
Pseudoglandular
Embryonic
0
5
10
15
20
25
30
35 40 wk
1 yr
2 yr
3 yr
FIGURE 42-1 Human lung development. Timeline of major events in human fetal and postnatal lung development.
The canalicular phase is marked by completion of the
conducting airways through the level of the terminal
bronchioles, and the development of the rudimentary gas
exchange units that are no longer invested with cartilaginous support. The acinus is the gas exchange unit of the
lung and encompasses a respiratory bronchiole and all of
its associated alveolar ducts and alveoli. A terminal bronchiole with all its associated acinar structures constitutes
a lobule. Branching of these distal airspaces continues on
a more limited basis during the canalicular phase, finally
achieving a total of 23 airway subdivisions.
Evolution of the relationships between the airspaces,
capillaries, and mesenchyme acquires greater significance
during the saccular phase of lung development (24 to
38 weeks’ gestation), enabling an alveolocapillary membrane sufficient to participate in gas exchange (0.6 μm)
by approximately 24 weeks. Beyond this point, the efficiency of gas exchange is determined by the available surface area. Lengthening and widening of the terminal sacs
expands the gas exchange surface area. Each saccule consists of smooth-walled airspaces with thickened interstitial
spaces containing a double capillary network. These will
give rise to two to three alveolar ducts, further expanding
the available surface area. Expansion of these rudimentary
gas exchange units continues well into the 3rd trimester
of human gestation; therefore the human lung is not fully
mature structurally, even at term delivery.
Postnatal lung development can be subdivided into additional stages (Burri, 2006). True alveoli become evident as
early as 36 weeks’ gestation, initiating the alveolar phase
of lung development. The development of primary alveoli
is followed by a further expansion of the gas-exchange
surface area through the formation of septae or secondary crests (see Alveolarization, later). Postnatal alveolarization extends from term through 1 to 2 years of age. An
initial phase of bulk alveolarization occurs within the first
6 months postnatally, with a more modest addition of secondary alveoli through the remainder of this period. The
alveoli of the infant lungs are different from adult alveoli.
These immature secondary alveoli contain a double capillary bed, whereas adult alveoli are invested by a single
capillary bed. Microvascular maturation, the next phase of
postnatal lung development, occurs between the first few
postnatal months of life through 3 years of age (see Development of the Pulmonary Vasculature, later).
There is considerable controversy regarding when the
lung ceases to add alveoli. Estimates have ranged from as
early as 2 years to as late as 20 years old in humans; this is
further complicated by the observation that alveolar expansion can occur in response to pneumonectomy in adult
animals and humans. The acquisition of alveoli after the
maturation of the microvasculature has been termed late
alveolarization. This activity has been most often demonstrated in subpleural regions of the lung and likely invokes
mechanisms similar to secondary crest formation.
The addition of alveoli is not the only means for
expanding the surface area of the lung. While alveolarization wanes over the first 3 years of life in the human,
growth of the lung continues to expand the gas exchange
surface. Between 2 years of age and adulthood, lung tissue
expands with lung volume roughly proportionately to the
increase in bodyweight of the child. Thus, because of the
combined processes of prenatal lung development, postnatal lung development, and lung growth, there is tremendous potential for expansion of the gas exchange surface
area that is developmentally programmed into the fetal
lung to account for the growing needs of the infant, child,
and adult for aerobic cellular respiration. The extent to
which these developmental mechanisms can be harnessed
after premature birth, with or without superimposed lung
injury, is a topic of active investigation.
COMPOSITION OF AIRWAYS AND ALVEOLI
As branching morphogenesis proceeds, the epithelium lining the successive generations of airways and alveoli gives
rise to specialized cells that participate in gas exchange,
surfactant production, mucociliary clearance, detoxification, and host defense. Differentiation proceeds in a centrifugal fashion from proximal to distal airspaces, lagging
behind branching. Temporal and contextual signals foster
the regionalization of epithelial cell types.
Proximal Airways
The airway epithelium is tall and columnar, decreasing to
a more cuboidal appearance more distally (Jeffrey, 1998;
Snyder et al, 2009). The endodermal epithelial lining cells
of the trachea and bronchi partition into four cell types:
undifferentiated columnar, ciliated, secretory-goblet, and
basal cells. Ciliated cells critical to the process of mucus
clearance are first apparent between 11 and 16 weeks’ gestation and become less prevalent in more distal airways.
Three types of secretory cells—those with largely mucous
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
granules, those with serous granules, and some with both
types of granules—can be seen as early as 13 weeks’ gestation. The number of mucin-producing goblet cells in
airways peaks at mid-gestation in the fetus and declines
into adulthood. Finally, immature basal cells expressing
epidermal keratin have been noted as early as 12 weeks’
gestation. Basal cells have a critical role in regenerating
injured large airway epithelium (see Stem and Progenitor
Cells in the Lung, later).
Cartilaginous support of the tracheobronchial tree begins
and proceeds in a centrifugal fashion beginning in the
primitive trachea at 4 weeks, reaching the main bronchi by
10 weeks, and proceeding to the most distal terminal bronchioles by approximately 25 weeks’ gestation. Cartilaginous
investment of airways is complete by the 2nd month postnatally. Submucosal glands are found in the interstitium
between the cartilaginous tissue and surface epithelium, and
they have a major role in airway host defense. Submucosal
gland development can be characterized by five stages: (1)
epithelial budding and invasion of the lamina propria, (2)
development of a lumen, (3) initiation of tube branching,
(4) dichotomous branching, and (5) repeated dichotomous
branching. By comparison, the airways of infants and children contain relatively more submucous glands than do
adults. The glands are lined by mucous cells proximally
and serous cells more distally, the latter comprising 60% of
the total epithelial cell content of the glands. Serous cells
secrete water, electrolytes, and proteins with antimicrobial,
antiinflammatory, and antioxidant properties, whereas the
mucous cells produce primarily mucins. In addition to this
host defense role, submucosal glands also contain a population of basal cells that respond to injury of the airway by
replenishing the airway epithelium.
Muscular investment of the airways begins as early as 6
to 8 weeks gestation as smooth muscle cells are identifiable
around the trachea and large airways. Fetal airway smooth
muscle is innervated and able to contract during the first
trimester. It is also responsive to methacholine challenge
that is reversible with β-adrenergic agonists. Muscularization increases through fetal life and childhood such that
there is an increase in the amount of smooth muscle relative to airway size compared with adults. Furthermore,
there is a rapid increase in bronchial smooth muscle immediately after birth, whether born at term or prematurely.
An additional airway cell deserves mention because of
its role in a wide variety of pediatric diseases. Pulmonary
neuroendocrine cells (PNECs) are found throughout the
airways, often in innervated clusters known as pulmonary
neuroendocrine bodies (NEBs) located at branch points in
the bronchial tree (Cutz et al, 2007). Although they arise
from foregut endoderm, the cell of origin is distinct from
other epithelial components of the lung. Solitary PNECs
are sensitive to stretch- and hypoxia-mediated secretion,
producing both amines (i.e., serotonin) and peptides (i.e.,
bombesin) that are important in regulating bronchial tone.
Pathologic conditions recently associated with PNECs
and NEBs, most often characterized by hyperplasia,
include bronchopulmonary dysplasia, disorders of respiratory control (congenital central hypoventilation syndrome
and sudden infant death syndrome), cystic fibrosis, and
pulmonary hypertension. Neuroendocrine hyperplasia of
infancy is a rare form of interstitial lung disease of infancy
573
associated with expansion of the number of PNECs and
NEBs, yet little is known about the mechanism of disease.
Distal Airways
The bronchiolar epithelium differs from the more proximal airway epithelium. In addition to being more cuboid
in appearance, the epithelium contains progressively fewer
ciliated cells and goblet cells, which are ultimately absent
from the terminal bronchioles. Instead, the nonciliated,
secretory Clara cell is found in increasing numbers and
density down the conducting airways, such that the Clara
cell is the most abundant cell of the terminal bronchiole
(Jeffrey, 1998). Clara cells are first evident by 16 to 17
weeks’ gestation, initially exhibiting large glycogen stores
that are replaced by secretory granules. Between 23 and 34
weeks’ gestation, there is a dramatic increase in Clara cell
numbers in distal bronchioles. Clara cells are critical to the
host defense and detoxification functions of the lung. This
specialized cell produces the highest levels of cytochrome
P-450 and flavin monooxygenases in the lung. While critically important in detoxification, these enzymes participate in the bioactivation of procarcinogens as well, placing
the Clara cell in a precarious position as a primary target
of toxic metabolites. The Clara cell also has an important
role in immunoregulation in the distal airways. Important
host defense products of the Clara cell include Clara cell
secretory protein (CCSP or CC10), surfactant proteins A,
and D, leukocyte protease inhibitor, and a trypsinlike protease. The function of Clara cell SP-B in airways is less
certain, especially because Clara cells do not produce a
mature 8-kDa SP-B protein, but may also contribute to
host defense. The secretion of antiproteases from Clara
cells suggests that they modulate the protease-antiprotease
balance in the distal lung.
Alveolar Epithelium
During the 4th through 6th months of gestation, the epithelial cells lining the acini begin to differentiate further
(Mallampalli et al, 1997). The cuboidal epithelial cells
accumulate large glycogen stores and develop small vesicles containing loose lamellae. The large glycogen pools
provide a ready source of substrate required for the production of increasing amounts of surfactant phospholipids,
and they decrease in size as surfactant production advances
in the fetal lung. In cells destined to become type 2 cells,
lamellar bodies become larger, more numerous, and more
densely packed with surfactant phospholipids and proteins,
whereas those cells destined to become type 1 cells, upon
losing their relationship to mesenchymal fibroblasts, lose
the prelamellar vesicles and become progressively thinner, thereby adopting a phenotype more suitable for gas
exchange. Alveolar type 1 and 2 cells are readily identified
early in the saccular stage of fetal lung development. There
remains considerable controversy regarding the origin of
type 1 cells. In culture, these cells demonstrate slow turnover, with a doubling time estimated to be between 40 and
120 days, suggesting that they are functionally terminally
differentiated in vivo. Furthermore, in response to epithelial denudation occurring with lung injury, type 2 cells proliferate to reestablish epithelial continuity, and then lose
574
PART X
Respiratory System
Lamellar body
Alveolus
Lung
Monolayer–multilayer film
Tubular
myelin
Adsorption
Nucleus
Lipid vesicle
Turnover
Clearance
Secretion
Recycling
A
Lamellar
body
Synthetic
SP-B, SP-C
Alveolar
macrophage
Alveolar
type II cell
Multivesicular body
B
2 microns
FIGURE 42-2 Surfactant life cycle. A, Schematic diagram depicting the life cycle of surfactant. B, Electron micrograph of a type II alveolar
epithelial cell showing the prominent lamellar bodies near the apical surface. (A, Reprinted with permission from Whitsett J, Weaver T: Hydrophobic surfactant proteins in lung function and disease, N Engl J Med 347:2141–2148, 2002. Copyright 2002 Massachusetts Medical Society. All rights reserved.)
phenotypic features such as lamellar bodies and acquire
markers of type 1 cells, suggesting that rapid repopulation
of type 1 cells requires a type 2 cell intermediary.
There is increasing appreciation for the alveolar type
1 cell as more than a passive membrane for gas exchange
(Williams, 2003). The large surface area and small cytoplasm/nucleus ratio provides for a thin alveolocapillary
membrane to facilitate gas exchange. However, this large
surface area also provides a large absorptive surface in the
lung. The presence of water and ion channels, some distinct from those in type 2 cells, facilitates the maintenance
of a relatively dry alveolus. Type 1 cells may also regulate
cell proliferation locally, signal macrophage accumulation,
and modulate the functions of local peptides, proteases and
growth factors.
Although most notable for its role in surfactant production, the alveolar type 2 cell provides additional important
functions in the alveolus (Fehrenbach, 2001). Alveolar type
2 cells are local progenitor cells, as mentioned previously.
Like type 1 cells, alveolar type 2 cells contain specialized
ion and water channels as well as ion pumps in both the apical and basal membranes that contribute to the movement
of water and ions across the epithelium (see Static Stretch:
Fetal Lung Fluid Protection, later). Type 2 cells contain
and secrete important antioxidants (superoxide dismutases
1, 2, and 3, glutathione) and molecules of innate host
defense (SP-A, SP-D, lysozyme) to participate in detoxification and sterilization of the alveolar microenvironment.
More recently, it is becoming clear that alveolar type 2
cells may also play a part in exacerbating alveolar pathology.
The type 2 cell participates in the coagulation-fibrinolysis
cascade through the production of fibrinogen, urokinasetype plasminogen activator, and tissue factor, especially
under pathologic circumstances. Type 2 cells are increasingly recognized as a source of cytokine and chemokine
production in the lung, as well as growth factors that can
promote fibrosis. Finally, cross-talk between epithelial
cells, cell matrix, interstitial cells, and local inflammatory
cells can foster the resolution of injury and inflammation
or prolong lung remodeling after injury with detrimental
effects, such as lung destruction and fibrosis. Although
previously heralded as the defender of the alveolus, the
alveolar type 2 cell have a much more complex role in
alveolar health and disease.
SURFACTANT
Pulmonary surfactant is essential to alveolar health. A
thin layer of liquid is constantly secreted into the alveolar
lumen to protect the delicate alveolar epithelium. The surface tension generated by this aqueous layer opposes alveolar inflation and promotes alveolar collapse at the end of
expiration, because of Laplace’s law, which states that the
collapsing pressure on the alveolus is directly proportional
to the surface tension while inversely proportional to the
radius of the alveolus. The film of pulmonary surfactant
at the air-liquid interface lowers surface tension as alveolar surface area decreases, thereby preventing end-expiratory atelectasis, maintaining functional residual capacity,
and lowering the force required for subsequent alveolar
inflations.
Pulmonary surfactant is a complex mixture of phospholipids, neutral lipids, and proteins that is synthesized,
packaged, and secreted by alveolar type 2 cells (Zuo et al,
2008). The life cycle of surfactant is depicted in Figure
42-2. Storage of surfactant occurs in the lamellar body, a
lysosome-derived membrane-bound organelle that undergoes regulated secretion in response to a variety of stimuli,
including stretching. In the alveolus, surfactant phospholipids transition through an extracellular storage form—
tubular myelin. Phospholipid and protein components are
recycled out of the surfactant monolayer at the air-liquid
interface and taken back into the alveolar type 2 cell, where
they can be repackaged into lamellar bodies. Alternatively,
alveolar macrophages are able to engulf and degrade surfactant components.
The predominant surfactant phospholipid is saturated
dipalmitoylphosphatidylcholine, with the remaining phospholipids consisting of monounsaturated phosphatidylcholine, phosphatidylglycerol and other phospholipids
(Table 42-1). Dipalmitoylphosphatidylcholine is the only
surface active component of lung surfactant capable of
lowering surface pressure to nearly zero. The presence of
unsaturated phospholipids and other lipid components like
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
TABLE 42-1 Composition of Pulmonary Surfactant
Component
Lipid
Percentage (by Weight)
90
Saturated phosphatidylcholine
45
Unsaturated phosphatidylcholine
25
Phosphatidylglycerol
Other phospholipids
Neutral lipids
Protein
5
5
10
10
Surfactant proteins
5
Serum proteins
5
cholesterol enables the monolayer to remain fluid at body
temperature during the respiratory cycle. Phospholipid
content in the fetal lung increases with advancing gestation because of increased activity of enzymes responsible
for phospholipid synthesis within alveolar type 2 cells. The
expression and activity of enzymes of the choline incorporation pathway, the predominant pathway for surfactant
phospholipid synthesis, are developmentally regulated and
induced by hormones. The inductive hormones that have
direct clinical relevance are glucocorticoids and agents that
increase intracellular cyclic adenosine monophosphate
(cAMP) such as the β-adrenergic agonist (and tocolytic)
terbutaline.
Surfactant contains a group of specific proteins with
importance to surfactant function and host defense. The
four surfactant proteins SP-A, -B, -C, and -D are subdivided based on their physical characteristics into either
hydrophobic (SP-B and -C) or hydrophilic (SP-A and -D)
proteins. The hydrophobic surfactant proteins play a major
role in the surface-active properties of surfactant, whereas
the primary roles of the hydrophilic surfactant proteins are
in host defense, immunomodulation, and surfactant clearance and metabolism (see Surfactant, earlier).
Together, the hydrophobic proteins facilitate the mobilization of surfactant phospholipid from tubular myelin to
the surface monolayer, promote spreading of phospholipids in the surfactant film, and assist in film stability at
end-expiration (Zuo, 2008). SP-B plays a central role in
alveolar health because of its critical function in surfactant
homeostasis. It is a secretory protein that exhibits strong
association with membranes, unlike SP-C, which contains
a membrane-spanning domain and covalently attached
fatty acids (palmitate) that render it integral to phospholipid membranes (Conkright et al, 2001). Both SP-B and
SP-C are synthesized as large precursor proproteins that
undergo extensive posttranslational processing as they pass
through the secretory pathway, ultimately reaching the
lamellar body. SP-B is essential for the process of lamellar body formation, and the alveolar type 2 cells of infants
with inherited deficiency of SP-B are devoid of lamellar
bodies. Because the lamellar body is where SP-C processing is completed, infants with inherited deficiency of SP-B
are also deficient in mature SP-C, instead accumulating
a larger, nonfunctional precursor of SP-C. Therefore
patients with inherited deficiency of SP-B, despite having
relatively normal surfactant phospholipid profiles, make a
575
pulmonary surfactant with poor surface tension properties because of the combined defects in SP-B and SP-C.
Conversely, because SP-C does not have either a direct
nor indirect role in SP-B protein processing, animals
with SP-C deficiency have normal SP-B, normal lamellar bodies, and relatively normal surfactant function, and
they exhibit no perinatal lethality because of surfactant
dysfunction.
Like the enzymes of surfactant phospholipid production, SP-B and SP-C exhibit developmental and hormonal
regulation of expression (Mendelson, 2000). In human
fetuses, SP-C mRNA is detected as early as 12 weeks’ gestation and SP-B mRNA by 14 weeks’ gestation, but the
mature proteins are not detectable in fetal lung tissue until
after 24 weeks’ gestation. SP-B protein is not detectable in
amniotic fluid until after 30 weeks’ gestation, increasing
toward term (Pryhuber et al, 1991), because of developmental regulation of posttranslational events in the proteolytic processing of proSP-B and proSP-C (Guttentag,
2008). Consequently, infants delivered prematurely have
reduced levels of both surface active components of surfactant, phospholipid, and hydrophobic surfactant proteins,
because of the developmental regulation of surfactant
proteins and the enzymes of phospholipid production in
alveolar type 2 cells. The rate of type 2 cell differentiation,
and secondarily surfactant production by the fetal lung, is
modulated by levels of endogenous corticosteroids and is
accelerated by administering antenatal glucocorticoid to
women in preterm labor. The response of the surfactant
system to glucocorticoid involves all the lipid and protein
components, and it occurs primarily through increased
gene expression, thus representing precocious maturation
mimicking the normal developmental pattern. Endogenous
thyroid hormones, prostaglandins, and catecholamines
also have stimulatory effects on type 2 cell maturation and
clearance of lung fluid at birth. Certain proinflammatory
cytokines (e.g., tumor necrosis factor [TNF]-α and transforming growth factor [TGF-β]) inhibit surfactant production in experimental systems and may downregulate
surfactant in conditions such as sepsis and inflammation.
A partial list of hormones capable of inducing or inhibiting
lung maturation is presented in Table 42-2.
DEVELOPMENT OF THE PULMONARY
VASCULATURE
The pulmonary vasculature consists of the vascular supply
to the acini and the bronchial circulation (Hislop, 2005).
During early fetal life, the airways act as a template for
pulmonary blood vessel development. The earliest pulmonary vessels form de novo in the tissue that surrounds
the lung bud by differentiation of mesenchymal cells into
endothelial cells and then capillaries, a process known as
vasculogenesis. Mesodermal cells within the mesenchyme
investing the developing lung tube differentiate into
endothelial cells, proliferate, organize into chords, and
develop a central lumen. As each new airway buds into the
mesenchyme, a new plexus forms and adds to the pulmonary circulation, thereby extending the arteries and veins.
By 5 weeks’ gestation, a capillary network surrounds each
bronchus and circulation of blood between the right ventricle and the left atrium via this network is evident.
576
PART X
Respiratory System
TABLE 42-2 Hormonal Regulators of Lung Maturation
Inducers
Inhibitors
Glucocorticoids
(cortisol)
Major endogenous
modulator of alveolar
development and
surfactant production
β-Adrenergic agonists
(epinephrine),
cAMP
Increase surfactant
production and secretion,
especially during labor
and delivery
Thyroid hormones
(T3, T4)
Enhance glucocorticoid
effects on lipid synthesis
Retinoic acid
May interact with
glucocorticoids to regulate
surfactant phospholipid
and protein production
Bombesin-related
peptides,
parathyroid
hormone–related
protein
May contribute to surfactant
lipid synthesis
Protein kinase
C activators
(proinflammatory
cytokines)
Inhibit surfactant protein
gene transcription during
infection, inflammation
TGF-β family
Inhibit type 2 cell maturation
during early gestation and
with inflammation
TNF-α
Inhibit SP-B and SP-C
gene transcription during
infection
Insulin
Inhibit surfactant protein
gene transcription
in infants of poorly
controlled diabetic women
Dihydrotestosterone
Delayed type 2 cell
maturation in males
By the canalicular stage of lung development, continued branching of the airways is accompanied by thinning
of the epithelium. At this stage, new blood vessels form
from preexisting vessels, a process known as angiogenesis.
By comparison, angiogenesis is initiated by endothelial
cell proliferation and sprouting from established vessels,
resulting in the extension of a vascular network into undervascularized regions. Vasculogenesis is the primary mode
of pulmonary vascular development until 17 weeks’ gestation, when all preacinar airways and their accompanying vessels are present, whereas angiogenesis becomes the
predominant mode of vascular development in the later
stages of lung development. Although originally thought
to be sequential processes, it is generally accepted that vasculogenesis occurring in the periphery and more central
angiogenesis occur concurrently during lung development
(deMello et al, 1997). Interconnections between vascular
networks arising from both angiogenesis and vasculogenesis increase in the saccular phase of lung development.
In the human lung, a second circulatory system, the
bronchial circulation, arises from the dorsal aorta supplying systemic blood. The bronchial vasculature develops
after the pulmonary circulation, with bronchial vessels first
apparent by 8 weeks’ gestation. The network of bronchial
vessels is extensive, with bronchial arteries demonstrated
as distal as the alveolar ducts in the adult respiratory tree.
The inappropriate branching of bronchial vessels from the
dorsal aorta is implicated in the formation of bronchopulmonary sequestration, a space-occupying lung malformation that can result in hypoplasia of the ipsilateral lung.
Vasculogenesis and angiogenesis are the primary mechanisms of vascular development throughout intrauterine
life. The human lung at term contains only a small portion of the adult number of alveoli, and the airspaces walls
are represented by a thick primary septum consisting of a
central layer of connective tissue surrounded by two capillary beds, each of them facing one alveolar surface (Burri,
2006). As alveolar architecture changes with the appearance of secondary septa, or secondary crests, folding of
one of the two capillary layers occurs within the secondary
septa. This double capillary network is not present in the
adult lung. Microvascular maturation involves fusion of
the double capillary network into a single capillary system.
The expansion of surface area and lumenal volume compresses the interstitium, bringing the capillary networks
in close proximity to potential air spaces and thereby promoting both alveolar surface area expansion and capillary
bed fusion. Interestingly, by the third postnatal week during which lung volume increases by 25%, there is a concomitant 27% decrease of the interstitial tissue volume
that is believed to promote focal microvascular fusions.
Subsequently, there is preferential growth of fused areas
that continues until 3 years of age.
Lastly, it is well known that lung volume increases
approximately 23-fold between birth and young adulthood, and capillary volume expands 35-fold. It has been
recently shown that this increase in capillary volume
occurs by insertion of new capillary meshes in the absence
of capillary sprouting. This new concept in capillary network growth has been named intussusceptive microvascular
growth, and involves the formation of transluminal tissue
pillars that then expand, resulting in increased capillary
surface area (Burri, 2006).
Muscularization can be detected early in development
of the pulmonary arteries (Hislop, 2005). Initially the
muscular investment of the vasculature is derived from the
migration of bronchial smooth muscle cells from adjacent
airways. Muscularization of preacinar and resistance arteries of the pulmonary vasculature begins in the canalicular
stage and continues through the remainder of gestation.
This second phase of smooth muscle cells investing pulmonary vessels develops from the surrounding mesenchyme.
Fibroblasts in close proximity to developing arteries alter
their cellular shape and begin to express α-smooth muscle
actin, a marker of smooth muscle cells. A third phase of
vascular muscularization has been described, largely in the
distal lung, in which capillary endothelial cells undergo
a process of endothelial-mesenchymal transition that
encompasses endothelial cell division, separation, and
migration from the endothelial layer and expression of
smooth muscle cell markers.
Muscularization of pulmonary arteries normally extends
to the level of the terminal bronchiole and is minimal to
absent in blood vessels surrounding respiratory bronchioles. Abnormal extension of smooth muscle along arterioles supplying acinar structures occurs in infants dying
from persistent pulmonary hypertension of the newborn
and in severe bronchopulmonary dysplasia.
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
577
TABLE 42-3 Composition of Human Fetal Lung Fluid Compared With Other Body Fluids
Component
Lung Fluid
Interstitial Fluid
Plasma
Amniotic Fluid
Sodium (mEq/L)
150
147
150
113
Potassium (mEq/L)
6.3
4.8
4.8
7.6
Chloride (mEq/L)
157
107
107
87
Bicarbonate (mEq/L)
3
25
24
19
pH
6.27
7.31
7.34
7.02
Protein (g/dL)
0.03
3.27
4.09
0.10
MECHANISMS OF LUNG
DEVELOPMENT
Fetal and postnatal lung development depend on several
key developmental processes: branching morphogenesis
to promote branching of the lung bud into the surrounding mesenchyme, static and cyclic stretching of the lung
that assist in promoting lung branching, alveolarization to
enhance the expansion of the gas exchange surface area,
and vasculogenesis and angiogenesis to ensure that the
developing epithelial surface area is invested with a similarly extensive vascular supply.
BRANCHING MORPHOGENESIS
Branching morphogenesis is the fundamental mechanism
of lung development. Branching is mediated by the accelerated growth of epithelial cells lateral to branch points with
concomitant growth arrest at the branch point (Affolter
et al, 2009). This process requires extensive communication between the cells lining the tubular lung bud and cells
contained within the invested mesenchyme. Classic tissue
recombination experiments in which mesenchyme from
proximal airways was transplanted to distal airways (and
vice versa) indicate that the mesenchyme has an important
inductive role in dictating the branching pattern and cell
fate of the expanding epithelium. More recently, studies
of mouse lung development indicate that three modes of
branching—domain branching, planar bifurcation, and
orthogonal bifurcation—are the basic mechanisms that
characterize the complex three-dimensional development
of the respiratory tree through the pseudoglandular phase
(Metzger and Krasnow, 1999). These routines occur repetitively during lung development and appear to be set by a
genetic clock dictating when side branches form, a bifurcation program determining when a branch bifurcates, and a
rotator function controlling the planar orientation of the
bifurcation.
STRETCH AND MECHANOTRANSDUCTION
The role of physical factors in modulating lung size is
well established; normal lung growth requires adequate
space in the chest cavity and appropriate tonic and cyclic
distending forces. Genetic defects that compromise the
thoracic skeleton and space-occupying lung masses like
congenital cystic adenomatoid malformations are associated with pulmonary hypoplasia because of the restriction
of intrathoracic space. Denervation of the diaphragm to
eliminate fetal breathing movements is also associated
with pulmonary hypoplasia, as is the manipulation of fetal
lung fluid volume.
Static Stretch: Fetal Lung Fluid Production
Fetal lung fluid is a product of the epithelial lining of
the developing lung (Wilson et al, 2007), averaging 4 to
6 mL/kg/h. Because of the resistance imparted by laryngeal abduction, fluid accumulates to a total volume of 20
to 30 mL/kg during gestation, providing end-expiratory
pressure of approximately 2.5 cm H2O pressure (Kitterman, 1996). The composition of fetal lung fluid is distinct from both amniotic fluid and plasma, as shown in
Table 42-3. The increased chloride content of fetal lung
fluid compared with serum is the result of active chloride
secretion by the tracheal and distal pulmonary epithelium,
largely because of the chloride channel CLC-2/CLCN2.
Fetal lung fluid secretion can be enhanced by prolactin,
KGF, PGE2 and PGF2, whereas it is inhibited by a variety
of mediators, including β-adrenergic agonists, vasopressin,
serotonin, and glucagon.
Fetal lung fluid is an essential component of lung development, but it presents a significant obstacle to the transition to air breathing upon delivery. Three important
events must occur to decrease the amount of fetal lung
fluid and its potential effect on alveolar surface tension:
absorption, bulk removal, and maturation of pulmonary
surfactant. The transition to air breathing must be coupled with a conversion from a secretory pulmonary epithelium to one that is absorptive. Enhanced sodium transport
across the alveolar epithelium is in part responsible for this
change. Much evidence suggests that induction of components of the epithelial sodium channels (ENaC) around
the time of birth is a major factor in promoting sodium
transport with water passively following the movement of
sodium. Absence of the α-subunit of ENaC in transgenic
mice is perinatal lethal because of the failure of fetal lung
fluid clearance. Induction of ENaC components occurs at
a transcriptional level in response to changes in extracellular matrix components, glucocorticoids, aldosterone, and
oxygen. By comparison, agents that increase intracellular
cAMP levels (i.e., β-agonists, phosphodiesterase inhibitors,
and cAMP analogues), while not increasing the number
of sodium channels, increase the probability of a channel
being open to sodium transport. In addition, glucocorticoid and thyroid hormones have important roles in priming the lung epithelium to be responsive to the actions of
β-adrenergic agonists on sodium transport across lung epithelia near term. Water channels consisting of aquaporins
578
PART X
Respiratory System
are also induced during the late fetal period to facilitate
fluid movement, but their contribution is unclear because
of the perinatal survival of mice in which aquaporin 5 or
both aquaporins 5 and 1 were absent.
Conversion to an absorptive surface is not sufficient to
minimize the fetal lung fluid at the time of term delivery.
The absence of uterine contractions is associated with an
increased incidence of retained fetal lung fluid in infants
delivered by cesarean section without the benefit of labor.
On delivery of the head and neck, continued uterine contractions on the fetal thorax promote expulsion of bulk
fluid from the fetal lung. However, animal studies have
shown that the magnitude of the benefit of thoracic compression during labor is modest (Bland, 2001). The primary mechanism by which labor facilitates clearance
of lung fluid is through hormonal effects on fluid clearance, especially through catecholamine-induced changes
in the ENaC opening. The onset of air breathing, associated with increased intrathoracic negative pressure,
assists in the clearance of residual fetal lung fluid into the
loose interstitial tissues surrounding alveoli. Fluid is then
reabsorbed via lymphatics and pulmonary blood vessels.
It is generally accepted that the amount of residual liquid in the lung after complete transition is approximately
0.37 mL/kg body weight.
Cyclic Stretch: Fetal Breathing Movements
Fetal breathing is an essential stimulus for lung growth
(Kitterman, 1996). Fetal breathing is readily detectable
as early as 10 weeks’ gestation. Fetal breathing occurs
for 10% to 20% of the time at 24 to 28 weeks’ gestation, increasing to 30% to 40% after 30 weeks’ gestation.
Originating from the diaphragm, fetal breathing is erratic
in frequency and amplitude, and it changes throughout
gestation. The volume of fluid moved is small and insufficient to be cleared from the trachea. Respiratory rates
range from 30 to 70 breaths/min, and periods of apnea
of up to 2 hours have been recorded. Sustained periods
of fetal breathing increase in duration with advancing
gestation. The frequency of fetal breathing varies with
sleep state (inhibited during quiet sleep) and exhibits
diurnal variation, with the lowest rates recorded early
in the morning. Fetal breathing is hormonally responsive and the inhibition of fetal breathing with the onset
of labor is attributed to the action of increased circulating prostaglandins. Maternal medications can influence
the frequency of fetal breathing movements. Central
nervous system stimulants are associated with increased
fetal breathing (i.e., caffeine, amphetamines), whereas
depressants are associated with decreased fetal breathing
(i.e., anesthetics, narcotics, ethanol). Maternal smoking is
associated with reduced fetal breathing, largely because
of increased fetal hypoxemia. Animal studies have shown
that permanent cessation of fetal breathing, regardless of
the insult, is associated with impaired fetal lung growth.
However, the effects of short-term alterations in fetal
breathing frequency and amplitude on fetal lung development are unknown. Together, constant distention from
the production and retention of fetal lung fluid, and episodic cyclic fetal breathing, are important mechanisms
for lung growth during fetal life.
ALVEOLARIZATION
Branching morphogenesis is the primary developmental
program that establishes the conducting airways of the
lung, but it is important to remember that alveolarization
is the developmental program that will establish the large
surface area involved in gas exchange (Galambos and Demello, 2008). This process will result in a 20-fold increase in
surface area between birth (with between 0 and 50 million
alveoli) and adulthood (>300 million alveoli). Primitive saccules develop low ridges (primary septa) that subdivide the
saccule into an alveolar duct containing primary alveoli and
outpouchings between the ridges (secondary septa/crests)
that establish secondary alveoli (Figure 42-3). Regions
destined for secondary septation exhibit increased elastin
deposition (Mariani et al, 1997), and elastin localizes to the
tips of the secondary crests as they form. Septae contain a
connective tissue core separating two capillary membranes,
suggesting that the septum is formed by the folding of a
capillary on itself, as mentioned previously. Septation also
leads to the development of the pores of Kohn, allowing
gaseous continuity between acini. The process of alveolarization is poorly understood, but is receiving much attention because of observations that infants who die after
severe bronchopulmonary dysplasia exhibit alveolar simplification with little evidence of secondary septation. It
remains unclear to what extent the process of alveolarization is disrupted by preterm birth and whether this developmental program can be resurrected after preterm birth
in the absence or presence of lung injury.
INTERDEPENDENCE OF ALVEOLAR AND
VASCULAR DEVELOPMENT
Recent evidence suggests that the pulmonary capillary
bed actively promotes normal alveolar development and
contributes to the maintenance of alveolar structures
throughout life (Thebaud and Abman, 2007). The observation that combined abnormalities in the airways and
vasculature occur in bronchopulmonary dysplasia supports
this hypothesis. Intraacinar arteries and veins continue to
develop after birth by angiogenesis as long as alveoli continue to increase in number and size. This process may
be reciprocal because vascular growth around the distal
airspaces suggests an inductive influence from the alveolar
epithelial cells as well.
MOLECULAR BASIS FOR LUNG DEVELOPMENT
The developmental processes that contribute to lung
organogenesis are under the regulation of interdependent
signaling pathways mediated by secreted growth factors
that are themselves under the control of large networks
of transcription factors controlling gene expression. Gene
regulatory networks common to other organs that depend
on branching morphogenesis during development, most
notably in the kidney and mammary gland, are also found
in the lung. Selected regulatory networks are highlighted
in the following sections to illustrate these systems. For
more detailed reading, additional references are provided
(Kaplan, 2000; Kumar et al, 2005; van Tuyl and Post,
2000; Warburton et al, 2000;).
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
Conducting airway
Primary septa
1. Wrinkling of primary septa
3. Transformation of conducting
airways into respiratory airways
2. Outgrowth of
secondary septa
FIGURE 42-3 Development of primary versus secondary alveoli.
The development of secondary alveoli from primary alveoli allows for
expansion of the total surface area available for gas exchange. Continued
growth of primary septa results in a wrinkled appearance that is followed
by elastin deposition at points where secondary septae, also known
as secondary crests, will form by an in-folding of the epithelium that
pulls underlying capillaries into the septum. Finally the more proximal
epithelium becomes differentiated into alveolar epithelial cells capable
of participating in gas exchange. Shaded cells represent bronchiolar
epithelial cells.
GROWTH FACTORS IN LUNG DEVELOPMENT
The initiation of branching morphogenesis is the result
of the interplay of signals between the developing lung
epithelial tube and its surrounding mesenchyme. Central
to this process is the family of fibroblast growth factors
(FGFs) that are produced and secreted by mesenchymal
cells, and bind to receptors on the plasma membrane of
epithelial cells, establishing a system of mesenchymalepithelial cross talk. In particular, the growth factor
FGF10 secreted by mesenchymal cells binds to the receptor FGFR2b on nearby epithelial cells. This signal will be
strongest in the closest epithelial cells because of a gradient that develops with diffusion of the secreted FGF10.
Binding of ligand to receptor results in signal transduction
within the epithelial cell that induces expression of a group
of genes via the mitogen-activated protein kinase pathway.
One of the genes induced by FGF10/FGFR2b signaling,
Sprouty 2 (Spry2), inhibits the mitogen-activated protein
kinase pathway, resulting in inhibition further FGF10/
FGFR2b signaling. Therefore a signal propagated by the
mesenchyme has an effect that is subsequently dampened
within the epithelial cell. Examination of genes expressed
early in the formation of a branch point in response to
FGF10 revealed a large number of genes that regulate
cell adhesion, cytoskeleton, and cell polarity—all essential elements of cell migration (Lu et al, 2005). Surprisingly, bud initiation was not associated with the expression
of genes that promote proliferation, which appears to
be more important for branch elongation. Still, animals
expressing reduced FGF10 develop pulmonary hypoplasia
with reduced numbers of large airways (Abler et al, 2009;
Ramasamy et al, 2007). Furthermore, increased FGF10
signaling during fetal lung development in mice by intrapulmonary injections of recombinant FGF10 produced
cystic structures with epithelial characteristics dependent
579
on the location of the injection: proximally with Clara cells,
distally with alveolar type 2 cells (Gonzaga et al, 2008).
These data provide strong evidence that FGF10 signaling
has diverse responses—from initiation of branching to differentiation of epithelial cells—depending on the temporal
or spatial context of signaling.
Like branching morphogenesis, lung vascular development is a complex and highly organized process that
requires multiple vascular signaling molecules to interact
in a specific temporospatial sequence. Vascular endothelial growth factor (VEGF) is a critical growth factor in
angiogenesis and vasculogenesis. The expression of the
VEGF ligand by epithelial cells and VEGF receptors by
endothelial cells of the developing human fetal lung reinforces the interdependence of the airspace and vascular
development. The expression of VEGF mRNA and protein is localized to the epithelial cells at the distal tips of
developing lung branches, and expression levels increase
with time (Galambos and Demello, 2007; Hislop, 2005).
VEGF gene expression is induced in epithelial cells by the
hypoxic environment of the growing fetal lung through
the actions of the oxygen-sensing hypoxia-inducible factor (HIF) family of transcription factors. From the single
VEGF gene, five different VEGF protein isoforms can
be produced, although VEGFA (VEGF165) is the most
studied. Each isoform has different affinities for each
of the three VEGF receptors (Flt-1/VEGFR1, Flk-1/
KDR/VEGFR2, and Flt-4/VEGFR3). Vascular endothelial cells express primarily VEGFR-1 and VEGFR-2,
whereas VEGFR-3 is on the plasma membrane of the
lymphatic endothelium. VEGF receptors are expressed
on the plasma membrane endothelial cells surrounding
the developing airways from very early in gestation, and
expression of VEGFR2/Flk1 is considered the earliest
marker of an endothelial progenitor cell. In vitro and in
vivo experiments have shown that VEGFA induces endothelial cell proliferation and migration, both key elements
of vascular sprouting, as well as tube formation through
interactions with VEGFR2. VEGFR1 appears to have
more importance in transforming primitive endothelial
tubes into more stable vascular networks, in part by reducing endothelial proliferation through downregulation of
VEGF production. The embryonic lethality of animals
with reduced VEGF expression attests to the critical
importance of VEGF and VEGFR signaling to vascular
development in the fetus, though not limited to the developing pulmonary vasculature.
TRANSCRIPTION FACTORS IN LUNG
DEVELOPMENT
The ligand-receptor interactions important to branching
morphogenesis and pulmonary vascular development are
in part determined by the actions of transcription factors
on facilitating or reducing gene expression. Transcription
factors are also critical in the differentiation of the lung
epithelium from the most rudimentary lung bud out to the
type 1 and 2 alveolar epithelial cells. The more important
transcription factors in epithelial cell determination in the
lung include the Gli, hepatocyte nuclear factor (HNF),
GATA, and Hox families of transcription factors (Kumar
et al, 2005; Warburton et al, 2000).
580
PART X
Respiratory System
The most important transcription factor in the lung is
thyroid transcription factor-1 (TTF-1), a product of the
Nkx2.1 gene. TTF-1 is considered a master regulator
of lung development, as transgenic mice null for Nkx2.1
exhibit complete absence of lung branching. However,
Nkx2.1 also has a prominent role in establishing cell fate
proximally to distally along the branching lung epithelium.
TTF-1 expression is detected as early as 11 weeks’ gestation in humans, and continues to be expressed by epithelial
cells at the distal tips of the branching lung epithelium,
ultimately becoming more restricted to Clara cells and
alveolar type 2 cells. TTF-1 is critical for the expression
of genes that are unique to differentiated epithelium, such
as CC10 expression in Clara cells and surfactant proteins
in alveolar type 2 cells. DNA binding sites for TTF-1
are found in the promoter regions of all four surfactant
proteins, CC10, and Nkx2.1 itself, creating a positive
feedback loop for sustained TTF-1 expression. TTF-1
function is highly dependent on phosphorylation of critical amino acids, although it remains unclear which kinase
is involved in this process (DeFelice et al, 2003). Interestingly, VEGFA expression is reduced in animals unable to
phosphorylate TTF-1, providing another important link
between epithelial and vascular development. TTF-1 is
itself regulated by other transcription factors that bind
to the promoter region of the Nkx2.1 gene, specifically
HNF-3β and GATA-6. Therefore the ability of networks
of transcription factors to bind to gene regulatory elements
of DNA in a coordinated fashion during fetal lung development enables the temporospatial expression of growth
factor networks that foster branching morphogenesis,
the process of differentiation that ultimately gives rise to
the approximately 40 cell types that constitute the human
lung, as well as critical coordination of both epithelial and
vascular development.
OTHER TOPICS IN LUNG
DEVELOPMENT
DEVELOPMENT OF PULMONARY HOST
DEFENSE
The adult human lung takes in approximately 7 L/min of
air contaminated with a variety potential pathogens that
can cause epithelial injury. The continuous exposure of
the epithelial surface of the conducting airway to inhaled
pathogens requires the presence of an efficient innate
immune response system to prevent infections. The proximal and distal airway epithelia have a major role in clearing
pathogens by secreting antimicrobial as well as antiinflammatory molecules. Components of mucociliary clearance
appear as early as 11 weeks’ gestation with the differentiation of ciliated epithelial cells and the expression of mucus
in mucus-secreting goblet cells within the epithelium and
submucosal glands, as discussed previously. The number of goblet cells peaks in mid-gestation (30% to 35%
of total airway epithelial cells), decreasing toward term to
levels lower than in adults, and then increasing after birth.
The postnatal increase in goblet cells occurs after preterm
birth as well, giving premature infants a greater number
of goblet cells than in term infants. Therefore premature
infants are prone to more mucus production in smaller
airways than are term infants, and premature infants have
relatively fewer ciliated cells to assist in mobilization of
secretions.
A number of microbial defense molecules are produced
and secreted by epithelial cells into the airways (Bartlett et
al, 2008). They include lysozyme, C-reactive protein, lactoferrin, collectins, β-defensins, and the only human member of the cathelicidin family, hCAP-18/LL-37 (Hiemstra,
2007). Two lung collectins were originally identified as
surfactant-related proteins—the hydrophilic surfactant
proteins SP-A and SP-D. Although originally identified as products secreted by epithelial cells lining airways
(Clara cells) and alveoli (type 2 cells), SP-A and SP-D have
been found in other sites associated with epithelial surfaces exposed to the external environment (Haagsman et
al, 2008). They interact with microorganisms, inflammatory cells, and leukocytes to facilitate clearance of microorganisms from the airspace, and they modulate allergic
responses.
The basis for the interactions of the lung collectins
with microbes and antigens centers on the binding of
sugars by the carbohydrate recognition domains of these
proteins (Wright, 2005). Both collectins bind a variety of
fungi and Pneumocystis carinii, and they have an important
role in inhibiting a variety of respiratory viruses, including influenza A and respiratory syncytial virus. Differences
in the structure of the carbohydrate recognition domain
provide SP-A and SP-D with altered affinities for different
sugar molecules, allowing complementary functions and
improving the diversity of microbial interactions. Interactions with gram-negative organisms frequently depends on
the ability of SP-A and SP-D to bind lipopolysaccharide,
whereas the mechanism of SP-A interactions with grampositive organisms, including group B beta-hemolytic
streptococci, are not as clear.
The lung collectins also modulate the functions of a
variety of immune cells, including macrophages, neutrophils, eosinophils, and lymphocytes (Wright, 2005). In
addition to opsonizing microorganisms, functions of the
lung collectins include stimulating chemotaxis of macrophages and neutrophils, enhancing cytokine production
by macrophages and eosinophils, attenuating lymphocyte
responses by inhibiting T cell proliferation, and modulating the production of reactive oxygen and nitrogen species
used in killing microorganisms.
Like the hydrophobic surfactant proteins, SP-A and
SP-D exhibit both developmental and hormonal regulation of expression (Mendelson, 2000). In human fetuses,
SP-A mRNA is undetectable before 20 weeks’ gestation,
and SP-A protein is first detectable in amniotic fluid by
30 weeks’ gestation, increasing toward term (Pryhuber
et al, 1991). In humans, SP-A gene expression is induced
by cAMP and glucocorticoids, although the response to
glucocorticoids in biphasic, showing attenuation at higher
doses. Retinoids, insulin, and growth factors such as TGFβ and TNF-α inhibit SP-A gene expression. Like SP-A,
SP-D levels in human lung are low during the second trimester (Dulkerian et al, 1996), are detectable in amniotic
fluid, and increase toward term (Whitsett, 2005). Levels
of both SP-A and SP-D increase markedly in the first days
after preterm birth.
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
DEVELOPMENT OF DETOXIFICATION
SYSTEMS
Although essential to cellular processes, oxygen concentrations beyond the physiologic limits may be hazardous to
cells. The lung is particularly susceptible to reactive forms
of oxygen and free radicals, because it is the organ with the
highest exposure to atmospheric oxygen. The fetal lung is
exposed to oxygen tensions of 20 to 25 mm Hg in utero,
and the transition to air breathing is associated with a fourfold to sevenfold increase in oxygen tension, presenting a
significant oxidant stress. Oxygen free radicals arise from
endogenous production through metabolic reactions, or
by exogenous exposure, from air pollutants and cigarette
smoke, and they can result in lung injury from oxidation of
proteins, DNA, and lipids. Therefore it is imperative for
the lung to develop an antioxidant detoxification system,
and for this system to be functional upon the fetal transition to air-breathing.
Oxygen free radicals are highly toxic substances. Superoxide, produced by the reduction of molecular oxygen by the
addition of an electron, is formed by all cells and occurs in
particularly high concentrations in phagocytic cells to facilitate the killing of microorganisms. Hydrogen peroxide is
generated from the transfer of a single electron to superoxide, and hydroxyl radicals are generated from the interaction
of hydrogen peroxide with superoxide. The free electrons of
free radicals interact with membrane lipids, resulting in lipid
peroxidation, with sulfhydryl and other groups on exposed
amino acids in proteins and with DNA causing direct damage.
These lipid, protein, and nucleic acid modifications damage
airway and alveolar epithelial cells as well as capillary endothelial cells, leading to altered epithelial integrity, interstitial
and airspace edema, and infiltration of inflammatory cells.
Increased reactive oxygen species levels have also been implicated in initiating inflammatory responses through the activation of transcription factors such as NFκB and AP-1, signal
transduction pathways, and chromatin remodeling that foster the expression of proinflammatory mediators (Rahman
et al, 2006).
Oxygen can have additional effects beyond the direct
toxicity of free radicals. Oxygen levels regulate the activity of plasminogen activator inhibitor-1 and other protease-antiprotease systems within the airspaces, thereby
modulating the destructive effects of proteases elaborated
from inflammatory cells. Oxygen can also regulate cellular
growth responses by altering the secretion of growth factors and DNA synthesis, generally through oxygen-sensing transcription factors, such as the HIF family.
Antioxidants attenuate the effects of oxygen free radicals
in the lungs, and in the lung there are both nonenzymatic
and enzymatic antioxidants. The major nonenzymatic
antioxidants are gluthatione (GSH), vitamins C (ascorbate)
and E (primarily α-tocopherol), β-carotene, and uric acid.
Enzymatic antioxidants include superoxide dismutases 1,
2, and 3, catalase, and a variety of peroxidases. Animal
studies indicate that many of the antioxidant enzymes
are induced before term delivery, and limited data suggest that the same is true of human fetuses (Weinberger
et al, 2002). Premature animals fail to induce antioxidant
enzymes in response to oxidative lung injury (Thibeault,
2000). Therefore preterm infants are significantly more
581
compromised in antioxidant defenses and, because of the
need for oxygen in the treatment of respiratory distress
syndrome, they are more susceptible to oxygen toxicity.
NOVEL CONCEPTS IN LUNG
DEVELOPMENT
STEM AND PROGENITOR CELLS IN THE LUNG
The ability of lung epithelium to replace cells damaged
from normal aging or injury has become the focus of recent
attention (Rawlins et al, 2008; Warburton et al, 2008).
Stem cells are undifferentiated and have an unlimited
capacity for self-renewal. Asymmetric divisions allow for
self-renewal through one daughter cell while enabling the
other daughter cell to become more terminally differentiated. Progenitor cells are more committed, and although
capable of self-renewal, they have more restricted cell fates.
Thus far, three cell types in the lung have been identified
as progenitor cells, having the capacity for self-renewal and
for replacement of a variety of specialized lung epithelial
cells. Basal cells in large airways and submucosal glands,
identified by the expression of Trp63/p63 and cytokeratin
5, are able to self-renew and give rise to ciliated and secretory cells. Clara cells in smaller airways, identified by the
expression of CC10/Scgb1a1, seem to be more committed
progenitor cells because they are able to self-renew or differentiate into ciliated cells. A subset of Clara cells at the
bronchoalveolar duct junction are both CC10/Scgb1a1
and SP-C positive, and thus have the potential to produce
either Clara cells or alveolar type 2 cells. The most limited
lung epithelial progenitor cell is the alveolar type 2 cell,
which divides rapidly to reestablish epithelial continuity in
damaged alveoli and then transdifferentiates into alveolar
type 1 cells. The mesenchymal progenitor cells that provide for vascular and muscular components of the developing lung have been studied less. Candidate cells have been
identified that give rise to endothelial cells in the process
of vasculogenesis and airway smooth muscle cells along the
branching lung epithelial tubes.
Evidence for the existence of stem-progenitor cells in
the lung is strong, but limited largely to mouse models of
lung development and lung repair; therefore extrapolation
to humans should be done with caution. The capacity for
self-renewal is tantalizing, but it means that such cells have
a risk for autonomous growth such as cancer. Controversy
exists around the potential to harness these populations of
cells as a means for correcting errors in lung development
(pulmonary hypoplasia), genetic diseases of the lung (cystic fibrosis), and abnormal repair of injured lungs (bronchopulmonary dysplasia).
EPITHELIAL TO MESENCHYMAL TRANSITION
Fibrosis often occurs as the result of severe injury to the
lung and has historically been a component of bronchopulmonary dysplasia (BPD). Emerging evidence suggests
that expansion of fibroblast populations in fibrotic lesions
is not simply the result of increased proliferation of local
fibroblast populations, rather the transformation of epithelial cells into mesenchymal components, a process
known as epithelial-mesenchymal transition (EMT) (Guarino
582
PART X
Respiratory System
TABLE 42-4 Potential Effects of Premature Birth on Lung Development and Maturation
Event
Effect of Preterm Birth
Potential Consequences
Development of
conducting airways
Branching: no effect; completed to the level of the respiratory
bronchi by 24 wk gestation
Tone: increased secondary to lung disease in part reflecting
developmental deficiency of nitric oxide
None
Increased airways resistance
Alveolarization
Variable depending on timing of delivery and severity of lung
disease; may also be compromised by excess glucocorticoids
Reduced lung growth and lung surface area
with increased alveolar size; impaired
pulmonary function
Development of
alveolocapillary
membrane
Minimal, reaches adult diameter by 24 wk gestation;
glucocorticoids induce precocious thinning
Gas exchange largely dependent upon surface
area, not alveolocapillary diameter
Type II cell
differentiation
Variable immaturity and deficient surfactant production depending
on timing of delivery; improved with antenatal glucocorticoids
Developmental deficiency of surfactant
content and composition results in RDS
Type I cell differentiation
Variable depending on timing of delivery; cells develop from
type II cells
Gas exchange largely dependant upon surface
area
Hydrophobic surfactant
proteins (SP-B, SP-C)
Variable depending on timing of delivery and other factors,
such as infection, that can impair gene transcription
High alveolar surface tension; RDS
Hydrophilic surfactant
proteins (SP-A, SP-D)
Variable depending on timing of delivery; both proteins appear
relatively late in third trimester
Comprised host defense: ability to clear
microorganisms from airways, alveolar
space, or both; impaired ability to modulate
inflammatory responses
Clara cell differentiation
Variable depending on timing of delivery; these cells appear in
middle 2nd trimester, but antioxidant products appear late in
3rd trimester
Impaired antioxidant and antimicrobial
defenses; may contribute to chronic lung
disease and pneumonia
Induction of antioxidant
systems
Variable depending on timing of delivery; expression of
antioxidants occurs late in third trimester
Lung injury from oxidant stress exacerbated
by need for increased oxygen; may contribute to chronic lung disease
Mucociliary clearance
Variable depending on timing of delivery; goblet cells decrease
in number toward term
Increased mucous production may obstruct
small airways
Development of the
pulmonary capillary
bed
Variable depending on timing of delivery in parallel to alveolar
development
Variable degrees of impaired gas exchange
commensurate with impaired alveologenesis and any superimposed lung injury;
pulmonary hypertension
Pulmonary arteries
Variable depending on presence and severity of associated lung
disease
Pulmonary hypertension associated with
chronic lung disease
Fetal lung liquid
Fluid loss: variable effects depending on magnitude and duration
of fluid loss (i.e., prolonged premature rupture of membranes) as
well as timing of delivery
Fluid retention: variable effects depending on timing of delivery,
because hormone surges near term and in labor promote
reabsorption before delivery
Pulmonary hypoplasia
Transient tachypnea of the newborn
Fetal breathing
movements
Variable depending on timing of delivery, but also depending on
maternal exposure to substances that reduce fetal breathing
movements
Unlikely to have effects in preterm infants
unless coexisting conditions severely limit
fetal breathing
Respiratory drive
Variable depending on timing of delivery
Apnea of prematurity
RDS, Respiratory distress syndrome.
et al, 2009; Thiery and Sleeman, 2006). EMT is an essential part of gastrulation and the development of cardiac
valves. Neural crest cells are the best model system for
EMT, because these epithelial cells must lose their local
attachments and travel long distances before locating their
final niche and differentiating accordingly. The evidence
is clear that the changes in cell phenotype characteristic
of EMT—from sedentary, interconnected epithelial cell
expressing epithelial marker proteins to mobile, proliferative mesenchymal cell expression markers of fibroblasts
and secreting collagen and other components of extracellular matrix—are under the control of local growth factors.
In the lung, the most prominent growth factor in EMT
associated with pulmonary fibrosis is TGF-β, a growth
factor essential for normal branching morphogenesis during early lung development.
It remains uncertain whether EMT has a similarly
important role in lung development as it does in cardiac
development, and whether EMT alone is important to
the abnormal repair response to lung injury. EMT has
the potential to reduce epithelial populations, resulting in
lung destruction, while expanding mesenchymal fibroblast
pools and enhancing local matrix deposition that reduces
lung elasticity. Many of the models of EMT are derived
from lung cancer cell lines or mouse models, limiting their
applicability to humans. However, improved understanding of the processes controlling EMT could lead to novel
therapies for limiting or reversing pulmonary fibrosis after
CHAPTER 42 Lung Development: Embryology, Growth, Maturation, and Developmental Biology
lung injury. Furthermore, given the potential for EMT to
contribute to lung destruction, a more common feature
of the “new BPD,” early events in EMT may be particularly important targets of preventive therapy in premature
infants.
ROLE OF miRNA IN LUNG DEVELOPMENT
AND MATURATION
Regulation of normal development and consequences of
abnormal development are at the heart of understanding
the implications of preterm birth and developing potential protective lung therapies. The central tenet of DNA
to RNA to protein is being challenged by new epigenetic
mechanisms—histone modifications, modification of DNA
and RNA, silencing RNA, and micro RNA (miRNA)—for
regulating gene expression. The evidence that miRNA has
an important role in normal fetal development is strong
(Nana-Sinkam et al, 2009). miRNAs are small, noncoding
RNAs (generally 19 to 25 nucleotides long) found within
cells that target genes for RNA degradation or inhibition
of protein synthesis. There are more than 500 recognized
miRNA, some of which are particularly enriched in lung
cell populations. MiRNAs are generated from a process
that begins in the nucleus. Long primary miRNA are transcribed, processed, and exported from the nucleus, followed by further cytoplasmic maturation before the final
miRNA is able to interact with regions of messenger RNA,
usually in the 3ˈ untranslated region of RNA that extends
beyond the RNA sequence used for protein translation.
Emerging evidence indicates that miRNA is essential
for normal lung development, because targeted deletion
of Dicer, a key enzyme in miRNA processing, results in
abnormal airway development and excessive apoptosis in
the lungs. The miR-17-92 cluster of miRNA is highly
expressed in embryonic mouse lung, decreasing into adulthood as lung development progresses. Altered expression
of the miRNA cluster miR-17-92 suggests a primary role
for these miRNA in maintaining a population of undifferentiated lung epithelium during lung development.
Because of their role in regulating developmental and
pathologic processes, miRNAs are increasingly seen as targets for therapeutic interventions. Obstacles to miRNAs as
therapeutic agents are similar to obstacles encountered in
other gene therapies, including mode of delivery, cell and
tissue specificity, and the potential for off-target effects.
SUMMARY
Lung branching morphogenesis is coordinated with pulmonary vascular development to provide a large surface
area and thin alveolocapillary membrane for adequate gas
583
exchange in the transition to air breathing and to meet the
needs of a growing infant and child. Although maturation
occurs late in fetal lung development, it is similarly critical
in the transition to air breathing. Although the fetal lung
developmental program requires an array of transcription
factors, hormones, and growth factors promoting branching morphogenesis, lung growth is equally dependent on
intact neural input to modulate fetal breathing, stability of an appropriately sized thorax, and the presence of
adequate lung and amniotic fluid. Furthermore the maturation of the host defense and detoxification systems minimize the effects of increased oxygen tension and exposure
to potential pathogens accompanying the transition to air
breathing. Premature birth affects all these functions as
illustrated in Table 42-4. Bronchopulmonary dysplasia is
the net result of multiple injuries to the underdeveloped
lungs of premature newborns that compromise postnatal
growth and development, and thus impairing function.
Integrated approaches to therapy that reflect the interdependency of these lung functions have the most promise
for minimizing the effects of premature birth on childhood
and, ultimately, adult lung function.
SUGGESTED READINGS
Burri PH: Structural aspects of postnatal lung development: alveolar formation and
growth, Biol Neonate 89:313-322, 2006.
Galambos C, Demello DE: Regulation of alveologenesis: clinical implications of
impaired growth, Pathology 40:124-140, 2008.
Hislop A: Developmental biology of the pulmonary circulation, Paediatr Respir Rev
6:35-43, 2005.
Jeffrey PK: The development of large and small airways, Am J Respir Crit Care Med
157:S174-S180, 1998.
Kaplan F: Molecular determinants of fetal lung organogenesis, Mol Genet Metab
71:321-341, 2000.
Kitterman JA: The effects of mechanical forces on fetal lung growth,, Clin Perinatol
23:727-740, 1996.
Kumar VH, Lakshminrusimha S, El Abiad MT, et al: Growth factors in lung development, Adv Clin Chem 40:261-316, 2005.
Mallampalli RK, Acarregui MJ, Snyder JM: Differentiation of the alveolar epithelium in the fetal lung. In McDonald JA, editor: Lung growth and development,
vol 100, New York, 1997, Marcel Dekker, pp 119-162.
Mendelson CR: Role of transcription factors in fetal lung development and surfactant protein gene expression, Annu Rev Physiol 62:875-915, 2000.
Thibeault DW: The precarious antioxidant defenses of the preterm infant, Am J
Perinatol 17:167-181, 2000.
Warburton D, Schwarz M, Tefft D, et al: The molecular basis of lung morphogenesis, Mech Dev 92:55-81, 2000.
Zuo YY, Veldhuizen RA, Neumann AW, et al: Current perspectives in pulmonary surfactant–inhibition, enhancement and evaluation, Biochim Biophys Acta
1778:1947-1977, 2008.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
43
Control of Breathing
Estelle B. Gauda and Richard J. Martin
The developmental aspects of respiratory control are of
considerable interest to physiologists and clinicians for a
multitude of reasons. The transition from fetal to neonatal life requires a rapid conversion from intermittent fetal
respiratory activity not associated with gas exchange to
continuous breathing upon which gas exchange is dependent. In addition, the circuitry that regulates respiratory
control serves as a unique link between the maturing lung
and brain. The term infant has the repertoire of respiratory
and cardiovascular responses that meets his or her metabolic demands during wakefulness, sleeping, crying, and
feeding even though the breathing pattern may be punctuated with apneic pauses and sighs (augmented breaths),
and the neurocircuitry that controls breathing is not completely developed. In some term infants who appear well,
developmental abnormalities in the neurocircuitry and
neurochemistry that regulate respiratory rhythmogenesis
can lead to profound disorders of breathing, placing them
at increased risk for sudden death. Infants who have died
of sudden infant death syndrome (SIDS), and those infants
with central congenital hypoventilation and Rett syndrome, have provided a window of opportunity for careful epidemiologic, genetic, neurochemical, and anatomic
study that has allowed researchers to better understand
which genes regulate the development of the respiratory
system and how environmental factors in fetal and early
neonatal life may adversely affect normal development.
The infant who is born prematurely has also provided
a unique opportunity to observe the natural developmental trajectory of respiratory control. Breathing in the most
premature infants is akin to fetal breathing, which is episodic, punctuated by periods of disturbingly long apneic
pauses interspersed with frequent periods of hyperventilation and sighs (augmented breaths). The purpose of fetal
breathing movements is not gas exchange, rather it is lung
development by inducing sufficient stretch of the chest
wall. For the infant who is born prematurely, the frequent
apneic events and hypoventilation associated with oxygen
desaturations and bradycardia are of significant concern.
The purpose of this chapter is to present what is currently
known about the neuroanatomy, neurocircuitry, and neurochemistry that controls breathing during development
to better understand how infants breathe, why premature
infants have apnea of prematurity, why term infants have
apnea of infancy, and how genetic errors and environmental influences modify mechanisms that control breathing
during fetal and early neonatal life.
ANIMAL MODELS OF CONTROL
OF BREATHING
Much of our understanding of basic mechanisms that
lead to a stable respiratory pattern is derived from animal
studies, both adult and newborn. Earlier studies used the
584
newborn pig, dog, and cat, as well as fetal and newborn
sheep models, to better understand developmental physiology, and much has been gained from these models. However, a more detailed understanding of the neuroanatomy,
neurocircuitry, and neurochemistry has been obtained
using in vitro models from newborn rats and mice. Of particular relevance, the stage of respiratory development of
the rat born at term is similar to that of the human infant
born at 25 to 29 weeks’ gestation; therefore the newborn
rodent model is applicable to breathing in the premature
infant. Studies using either of two in vitro models (brainstem slices that include the area that contains the “pacemaker cells mediating rhythmogenesis” and the isolated
brainstem spinal cord preparation from fetal and newborn
rodents) have led to an explosion of information about all
aspects of maturation of respiratory control within the last
decade (Richter and Spyer, 2001). Because the stability and
viability of the in vitro preparations are best when tissues
are used from late embryonic or early postnatal rodents
(within first week of postnatal life), data from these in vitro
models are relevant to respiratory control during early
development.
OVERVIEW OF RESPIRATORY
CONTROL
The leading hypothesis regarding autonomic control of
breathing is that respiratory rhythm is generated from
a core group of synaptically coupled excitatory neurons
in the brainstem located in the pre-Bötzinger complex
(PBC). These neurons depolarize during the three phases
of respiration: inspiration, postinspiration, and expiration.
The timing of the respiratory cycle is controlled by inhibitory interneurons that discharge during specific phases of
the respiratory cycle. Collectively this complex network of
neurons is called the central pattern generator (Duffin, 2004;
Smith et al, 2009).
Inspiratory, postinspiratory, and expiratory neurons depolarize with each phase of the respiratory cycle coincident
with activity motoneurons innervating the muscles of
respiration (Figure 43-1). Whereas inspiration is always
associated with phrenic nerve activity and contraction of
the diaphragm, the first phase of expiration (E1), known as
postinspiratory activity, can be associated with phrenic nerve
activity, and the second phase of expiration (E2) is not (see
Figure 43-1).
With development, the intrinsic properties and neurotransmitter profiles of respiratory-related neurons and
synaptic inputs from (1) higher brain centers (frontal and
insular cortex, hypothalamus, reticular activating system
and amygdala, (2) mechanoreceptors in the lungs and
upper airways, (3) peripheral chemoreceptors in the carotid
body, and (4) central chemoreceptors on the ventral medullary surface modify the excitability of respiratory-related
CHAPTER 43 Control of Breathing
neurons. In addition, the intrinsic “sensing” properties of
mechanoreceptors and chemoreceptors are also changing.
Last, the integrated respiratory output is also dependent
on the strength of the synapse between the premotor respiratory neurons and the respiratory motoneurons innervating the diaphragm, intercostals and muscles of the upper
airway, and the corresponding neuromuscular junctions
(Figure 43-2). The effect of development on respiratory
motoneurons and the neuromuscular junction will not be
discussed in depth in this chapter; for further discussion
of this topic the reader is referred to two excellent reviews
(Greer and Funk, 2005; Mantilla and Sieck, 2008).
55
Vl
mV
70
1
Insp.N.
3 Exp.N.
Post-Insp.N.
2
Ve
INSP
Output
phrenic
Cycle phases
PI
MUSCLES OF RESPIRATION
EXP
aug dec
E1
2s
585
E2
FIGURE 43-1 Simultaneous triple recordings of electrical activities
of the main populations of respiratory neurons: 1, an inspiratory neuron
(large spikes, middle tracing); 2, a postinspiratory neuron (small spikes,
middle tracing); 3, an expiratory neuron (top tracing) and the phrenic
nerve (bottom tracing). Note the augmenting (aug) activity of the
phrenic nerve during inspiration and the decrementing (dec) activity of
the phrenic nerve during the postinspiratory (PI) phase of the respiratory cycle. The postinspiratory phase is the first phase of expiration (E1)
and is associated with phrenic nerve activity. In the second phase of
expiration (E2), the phrenic nerve is silent. (Reproduced from Richter DW,
Spyer KM: Studying rhythmogenesis of breathing: comparison of in vivo and in
vitro models, Trends Neurosci 24:464-472, 2001.)
The muscles of respiration include the pump muscles
(diaphragm, intercostal muscles, and abdominal muscles)
and muscles of the upper airway (alae nares, pharyngeal
muscles, and laryngeal muscles). Upper airway muscles
modulate the rate of inspiratory and expiratory airflow.
During the inspiratory phase of the respiratory cycle the
diaphragm, external intercostals (in infants), and posterior cricoarytenoid (laryngeal dilator) contract. During
the postinspiratory phase, the diaphragm and the thyroarytenoid (laryngeal constrictor) contract. Diaphragmatic
and thyroarytenoid postinspiratory activity is common
in newborns, both human (Eichenwald et al, 1993) and
animals (England et al, 1985). Because the chest wall of
newborn infants, particularly premature infants, is highly
compliant, diaphragmatic and laryngeal postinspiratory
activity retards expiration to maintain functional residual
ParabrachialKölliker-Fuse
complex
Bötzinger
complex
Nucleus of the
solitary tract
PreBötzinger
complex
Rostral VRG
Caudal VRG
Nucleus
ambiguus
Ventral
respiratory
column
Medullary
raphe
Arcuate
nucleus
FIGURE 43-2 Schematic illustrating the anatomic relationship between the regions in the human brainstem that compose the respiratory network. These regions include specialized neurons in the dorsolateral pons (parabrachial and Kölliker–Fuse nuclei) nucleus of the solitary
tract (nTS), and ventral respiratory column. The ventral respiratory column is organized rostrocaudally extending from the level just below the facial
nucleus to the C1 level of cervical cord. The ventral respiratory column consists of the Bötzinger complex, pre-Bötzinger complex, and the rostral and
caudal ventral respiratory groups (VRGs). Vagal motor neurons of the nucleus ambiguus innervate the laryngeal muscles. The medullary raphe, arcuate
nucleus, located just underneath the ventral medullary surface, contains neurons that depolarize in response to hypercapnia and hypoxia. The retrotrapazoid nucleus (not shown) are CO2/H+ chemosensors, and is located rostrally below the facial nucleus on the ventral medullary surface. (Modified with
permission from Benarroch EE: Brainstem respiratory control: substrates of respiratory failure of multiple system atrophy, Mov Disord 22:155-161, 2007.)
586
PART X
Respiratory System
capacity. This postinspiratory activity is often audible,
known as grunting, and is heard in infants with low lungvolume disease states, such as surfactant deficiency and
atelectasis. As outlined previously, the second phase of
expiration (E2) is often quiescent regarding muscle activity; the diaphragm relaxes and the lungs recoil. During
forced expiration often seen during exercise and significant hypercapnia, the internal intercostals and abdominal
muscles may contract during E2. Brainstem nuclei that are
involved in the control of upper airway muscles include
the upper airway motoneurons of the nucleus ambiguous,
the dorsal motor nucleus of the vagus, and the hypoglossal
nucleus (Jordan, 2001; Nunez-Abades et al, 1992). Projections from respiratory-related neurons synapse onto these
upper airway motoneurons, thereby regulating the activity
of these nerves and muscles during the respiratory cycle.
RESPIRATORY CENTER
NEUROANATOMY
As shown in the anatomic illustration (see Figure 43-2)
and the schematic (Figure 43-3), the respiratory-related
neurons are located in three main areas in the brainstem:
(1) the dorsal respiratory group within the nucleus tractus solitarii, (2) the ventral respiratory column (VRC) that
extends from the facial nucleus to the ventrolateral medulla
at the spinal-medullary junction, and (3) the pontine respiratory group within the dorsolateral pons (Alheid and
McCrimmon, 2008). The VRC can be subdivided into a
rostral part, involved in rhythmogenesis, and a caudal part
involved in pattern formation. The rostral VRC contains
both the rostral ventral respiratory group (VRG), which
contains a large proportion of bulbospinal inspiratory neurons that project directly to the phrenic and external intercostal motoneurons, and the caudal VRG, which contains
bulbospinal expiratory neurons that project to abdominal
and internal intercostal motoneurons. Bulbospinal neurons are neurons that originate in the medulla and synapse
onto motoneurons in the spinal column. Of importance,
within the rostral VRC are two areas that are essential to
formation of respiratory rhythm: the Bötzinger complex,
and PBC. The Bötzinger complex contains propriobulbar
expiratory neurons that provide strong inhibitory inputs
onto inspiratory and expiratory bulbospinal neurons in the
VRC. Propriobulbar neurons are neurons originating in
the brainstem that send projections to other neurons in the
brainstem. The PBC contains a core group of synaptically
coupled excitatory neurons that have pacemaker properties, similar to the pacemaker cells in the AV node of the
heart. These pacemaker cells are rostral to the nucleus
SARs
RARs
C-fiber
Inspiratory neurons
Expiratory neurons
Peripheral
arterial
chemoreceptors
Laryngeal
chemoreflex
CO2/H chemosensors
RONTINE
PONTINE
RESPIRATORY
GROUP
(phase switching)
Caudal nucleus tractus solitarii (nTS)
Dorsal Respiratory Group
Inspiratory neurons
VENTRAL RESPIRATORY COLUMN (VR Column)
Facial
nucleus
Rostral (rhythm generating)
BötzC
expiratory
neurons
PBC
pacemaker neurons
inspiratory &
expiratory
Caudal (pattern forming)
Ventral Respiratory Group VRG
Rostral VRG (rVRG)
Caudal VRG
(cVRG)
RTN/pFRG
CO2/H
Bulbopspinal
neurons
Bulbopspinal neurons synapse on to respiratory motoneurons
(phrenic, intercostals, upper airway, abdominals)
FIGURE 43-3 Simplified schematic illustrating the major pontine-medullary brainstem network controlling respiration and afferent and
efferent projections. The pontine respiratory group contains two nuclei that send neuronal connections to respiratory-related neurons in the ventral
respiratory column (VRC). The pontine respiratory group mediates phase switching between inspiration and expiration. Within the nucleus tractus
solitarii (nTS) are the inspiratory neurons of the dorsal respiratory group with projections to motoneurons in the spinal column. The nTS also receive
monosynaptic inputs from vagally mediated reflexes in the lung and upper airways (including slowly adapting receptors [SARs], rapidly adapting
receptors [RARs], and C-fibers receptors), laryngeal chemoreceptors, and peripheral arterial chemoreceptors. Projections from second-order neurons
in the nTS then synapse on to neurons in the rostral and caudal VRC. The VRC extends from the level of the facial nucleus to C1 in the cervical
spinal cord. The rostral VRC is involved in respiratory rhythmogenesis and contains the expiratory neurons of the Bötzinger complex (BötzC) and the
pacemakers cells of the pre-Bötzinger complex (PBC). The BötzC and PBC contain propriobulbar neurons that project to inspiratory neurons in the
rostral ventral respiratory group (VRG) and expiratory neurons in the caudal VRG. The BötzC also contains bulbospinal neurons that synapse on to
phrenic motoneurons in the spinal cord, whereas the PBC only contains propriobulbar neurons. Neurons in the VRG are responsible for shaping the
respiratory pattern and receive inputs from second-order neurons in the nTS and from rhythm-generating neurons in the BötzC and PBC. Bulbospinal neurons from the DRG, BötzC, rVRG, and cVRG synapse onto motoneurons that control the activity of the muscles of respiration.
CHAPTER 43 Control of Breathing
ambiguous, have both intrinsic inspiratory and expiratory
bursting properties, and are essential to maintaining respiratory rhythm (Smith et al, 1991). Progressive destruction
of the PBC disrupts rhythmogenesis, leading to death in
an animal model (Ramirez et al, 1998).
Another important group of neurons are those within
the retrotrapezoid nucleus (RTN) located along the ventral
medullary surface beneath the facial nucleus (see Figure
43-3). These neurons have chemosensitive properties and
depolarize in response to increasing CO2 and decreasing
pH, and they are presumed to synapse on rhythm and
pattern generating neurons in the VRC (Guyenet et al,
2008). All these neuronal groups and networks for rhythmogenesis are present in newborn animals born at term
(e.g., sheep, cats, pigs) or born prematurely (e.g., rodents)
in which rhythmogenesis is well established before birth.
Episodic spontaneous fetal breathing movements occur
in human fetuses as early as 10 weeks’ gestation (de Vries
et al, 1985). In rodents, respiratory rhythmogenesis is first
detected at embryonic day 15 in rats and day 17 in mice
(Thoby-Brisson and Greer, 2008). The emergence of this
respiratory related activity in rats is coincident with the
characteristic expression of neurokinin 1 receptors of the
PBC (Thoby-Brisson and Greer, 2008).
In summary, respiratory rhythm and inspiratory-
expiratory patterns emerge from dynamic interactions
between: (1) excitatory neuron populations in the PBC and
rostral VRG, which are active during inspiration and form
the inspiratory motor output; (2) inhibitory neuron populations in the PBC that provide inspiratory inhibition within
the network; and (3) inhibitory neuron populations in the
Bötzinger complex, which are active during expiration and
provide expiratory inhibition within the network and to
phrenic motor neurons (see Figure 43-3) (Smith et al, 2009).
NEUROCHEMISTRY MEDIATING
RESPIRATORY CONTROL
Glutamate is the major neurotransmitter mediating excitatory synaptic input to brainstem respiratory neurons and
respiratory premotor and motor neurons through binding
to the α-amino-3-hydroxy-5-methylisoxazole-4-propionic
acid kainite (Bonham, 1995) and metabotropic glutamate receptors (Pierrefiche et al, 1994). GABA (gamma-
aminobutyric acid) and glycine are the two major inhibitory
neurotransmitters mediating inhibitory synaptic input in
the respiratory network; they have a key role in pattern generation and termination of inspiratory activity (Haji et al,
2000). GABA (via GABAA receptors) and glycine (via glycine receptors mediate fast synaptic inhibition via activation
of chloride channels (Bianchi et al, 1995). GABAB receptors, which are metabotropic G-protein coupled receptors, have a greater role in inhibiting respiratory rhythm in
adult mammals (Kerr and Ong, 1995). Throughout development, glutamate always functions as an excitatory neurotransmitter; however, it is not the case that GABA and
glycine are always inhibitory neurotransmitters. During
early development, GABA and glycine mediate excitatory
neurotransmission in many neuronal networks, including
the respiratory network (Putnam et al, 2005). The expression of K± chloride cotransporters (KCC2 and NKCC2)
reduces the intracellular chloride concentrations that
587
change the effect of GABA binding to GABAA receptors
from depolarizing during late embryonic and early postnatal to hyperpolarizing with maturation (Rivera et al, 1999).
NEUROMODULATION OF RESPIRATORY
PATTERN AND RHYTHM
The baseline excitatory and inhibitory influences mediated
by glutamate and GABA–glycine, respectively, on major
neuronal networks are further altered by many endogenously released neuromodulators that shape and fine
tune respiratory pattern and rhythm throughout development, as outlined in Table 43-1. For example, acetylcholine, substance P, cholecystokinin, cholecystokinin (CCK)
and thyrotropin-releasing hormone all exert an excitatory
drive, whereas opioids and somatostatin exert an inhibitory drive on respiratory-related neurons. Dopamine,
adenosine, serotonin, and norepinephrine can have excitatory and inhibitory influences depending on the specific
receptors that the neuromodulator binds. Neurons within
the PBC that are the kernel of rhythmogenesis are also distinctly identified by being immunopositive for the glutamate transporter, NK1, μ-opioid, and GABAB-receptors.
Although these cells are primarily excitatory and release
glutamate, they can be modulated by synaptic inputs that
release substance P, opioids, and GABA (Doi and Ramirez,
2008). Recently these rhythmogenic neurons have been
found to produce, and be excited by, brain-derived neurotrophic factor (Bouvier et al, 2008).
GENETIC MUTATIONS AFFECTING
BIOGENIC AMINES
Some neuromodulators may be more critical for supporting respiratory rhythmogenesis than others. By identifying the genetic mutations that are associated with marked
abnormalities in respiratory control, a better understanding of the key role of several neuromodulator systems has
been elucidated. For example, serotonergic neurons in the
caudal medullary raphe nuclei have extensive projections
to phrenic and hypoglossal motoneurons, the nucleus tractus solitarii (nTS), the RTN, and the PBC (Pilowsky et al,
1990; Voss et al, 1990). Thus, as reviewed by Kinney et al
(2009), the serotonergic system has a significant influence
on the modulation and integration of diverse homeostatic
functions. Medullary serotonergic neurons are also CO2
sensitive (Richerson et al, 2001). In genetically modified
mice that do not develop medullary serotonergic neurons,
CO2 sensitivity is reduced by 50% (Hodges et al, 2008).
Individuals with Prader–Willi syndrome, who may exhibit
breathing abnormalities at birth) have mutations in the
Necdin gene associated with abnormalities in the brainstem
serotonergic system (Zanella et al, 2008a). Mice lacking
the Necdin gene also have abnormal brainstem serotonergic neurochemistry (Zanella et al, 2008b). In some infants
who have died of SIDS, disruptions of the brainstem serotonergic system have been identified (Kinney et al, 2009;
Paterson et al, 2006b). Abnormalities in norepinephrine
production also disrupt normal breathing. Rett syndrome
is an X-linked disorder with mutations in the methyl CpG
binding protein 2 (MECP2) gene. Affected individuals have
severe respiratory disturbances that can be fatal. Genetically
588
PART X
Respiratory System
TABLE 43-1 Neurotransmitters and Neuromodulators That Mediate Respiratory Rhythm
Neuromodulator
Glutamate
Receptor
Subtype
Source of the Endogenous
Ligand
NMDA, AMPA
GluR
Excitatory or
Inhibitory
on Respiratory
Rhythm
Comment
Excitatory
Major excitatory neurotransmitter
Ach
M3
PAG, LC, X
Excitatory
NE
α1-adrenergic
LC
Excitatory
Serotonin
5-HT2A2B,
5-HT3, 5-HT4
Raphe
Excitatory
Dopamine
Likely D1
PVN, hypothalamus
Excitatory
ATP
P2X2
Ventral medulla; CO2/H+ cells in
the RTN
Excitatory
Adenosine
P2Y1
Ventral medulla
Excitatory
Substance P
NK1
nTS, NA
Excitatory
CCK
CCK1
nTS, raphe
Excitatory
TRH
TRH-R, R2
raphe
Excitatory
GABA
GABAA
GABAB
Glycine
GlyR
NE
α2-adrenergic
Pons
Dopamine
D4
PVN, hypothalamus
Adenosine
A1, A2
Ubiquitous from metabolism of
ATP that increases during hypoxia
Inhibitory
Contributes to respiratory depression
at baseline (A1), and mediates HVD
Opioid
μ, δ, κ
nTS, PBN, PVN, raphe
Inhibitory
Prominent inhibitory effect during
early development
PDGF
PDGF-β
nTS,
Inhibitory
Contributes to HVD
Inhibitory
Major inhibitory neurotransmitter
(can be excitatory during fetal life)
Inhibitory
Can be excitatory during fetal life
Inhibitory
Data combined from Doi A, Ramirez JM: Neuromodulation and the orchestration of the respiratory rhythm, Respir Physiol Neurobiol 164:96, 2008; and Simakajornboon N,
Kuptanon T: Maturational changes in neuromodulation of central pathways underlying hypoxic ventilatory response, Respir Physiol Neurobiol 149:273, 2005.
Ach, Acetylcholine; ATP, Adenosine triphosphate; CCK, cholecystokinin; GABA, gamma-aminobutyric acid; HVD , hypoxic ventilatory depression; LC locus ceruleus; NA, nucleus
ambiguous; NE, norepinephrine; nTS, nucleus tractus solitarii; PAG, periaqueductal gray; PBN, parabrachial nucleus; PDGF, platelet-derived growth factor; PVN, paraventricular
nucleus; RTN, retrotrapezoid nucleus; TRH, thyrotropin-releasing hormone; X, vagal nucleus.
modified mice that lack the Mecp2 gene have reduced levels
of norepinephrine and serotonin in the medulla and have
breathing patterns similar to humans with Rett syndrome
(Roux et al, 2008). Pharmacologic treatment to increase
brain norepinephrine and serotonin levels stabilizes breathing and prolongs the life of these mice (Roux et al, 2007).
PERIPHERAL MECHANORECEPTOR
INPUTS THAT MODULATE
RESPIRATORY PATTERN AND
RHYTHM
The nTS in the brainstem (see Figure 43-3) is where sensory afferents that are transmitting pressure signals from
the upper airway, volume signals from the lung, and chemical signals from the blood and cerebrospinal fluid, synapse
onto second-order neurons that send projections either
monosynaptically or polysynaptically to phrenic motor
neurons via respiratory-related bulbospinal neurons.
VAGALLY MEDIATED REFLEXES
Essentially all bronchopulmonary reflexes that modify
depth and duration of inspiration and expiration are mediated through the vagal nerve. The vagal nerve has both
myelinated and unmyelinated fibers. Myelinated vagal
afferent fibers are activated via (1) slowly adapting stretch
receptors (SARs), which are activated by volume and stretch
of the lung (mediating the Breuer-Hering Reflex), or
(2) rapidly adapting receptors (RARs), which are activated in response to inhaled irritants (e.g., ammonia, cigarette smoke) and large inflations or deflations of the lung
(Kubin et al, 2006). Changes in respiratory timing of inspiration and expiration are the respiratory response to activation of SARs, whereas sighing (i.e., augmented breaths)
and coughing are the characteristic ventilatory responses
to activation of RARs. Unmyelinated vagal afferents, specifically C fibers, are activated by a multitude of chemical
stimuli, including CO2 and capsaicin, in addition to lung
edema and elevated temperature. Rapid shallow breathing
and apnea are the characteristic ventilatory responses to
the activation of C-fibers in the airways.
BREUER–HERING REFLEX DURING
DEVELOPMENT
The duration of inspiration and expiration is greatly influenced by lung inflation, and the most well-characterized
bronchopulmonary reflex is the pulmonary stretch reflex
mediated through SARs, discovered by Josef Breuer
in 1868. In adult cats, Breuer showed that expansion of
the lungs reflexively inhibits inspiration and promotes
CHAPTER 43 Control of Breathing
expiration, and that deflation of the lungs promotes inspiration and inhibits expiration (Widdicombe, 2006). In the
nTS, these afferents monosynaptically synapse on to second-order neurons called pump cells and inspiratory-β neurons. These second-order neurons then send projections to
respiratory-related bulbospinal neurons in the VRC. Bulbospinal neurons synapse on the phrenic motoneurons in
the cervical spinal cord. SARs also influence the activity of
a subset of propriobulbar VRC neurons that are involved
in rhythmogenesis (for review, see Kubin et al, 2006).
The Breuer–Hering (B-H) reflex does not appear to be
important in regulating fetal breathing movements, because
vagotomy performed in fetal sheep has little effect on the
incidence, frequency, or amplitude of these breathing movements (Hasan and Rigaux, 1992). However, the B-H reflex
is important in establishing continuous breathing and adequate gas exchange at birth (Wong et al, 1998). The reflex
maintains functional residual capacity in newborns and
infants, because vagotomy within 48 hours of birth results
in respiratory failure associated with marked atelectasis in
newborn sheep (Lalani et al, 2001; Wong et al, 1998). Vagal
innervation in utero was initially thought to be necessary
for the development of surfactant (Alcorn et al, 1980), but
this belief has recently been challenged by new findings
from Gahlot et al (2009). These studies showed that vagal
denervation performed at 110 to 113 days’ gestation (term
gestation, 147 days) in fetal sheep had no effect on alveolar
architecture, number of type II cells, morphology of lamellar bodies, or the level of surfactant proteins A and B and
total phospholipids in lung tissue (Gahlot et al, 2009).
In humans, the contribution of the B-H to tidal breathing is determined by occluding the airway at either end
expiration, where the next occluded inspiratory effort is
prolonged and expiratory effort is shortened, or end inspiration, where the next occluded expiratory effort is prolonged and inspiratory effort is shortened. The inspiratory
and expiratory time of the occluded effort is compared to
the inspiratory and expiratory time of the preceding nonoccluded breath to determine the percentage increase or
decrease of the inspiratory or expiratory times. With this
technique, the B-H reflex has been shown to contribute
significantly to tidal breathing in infants, which is strongest at birth and then decreases during the 1st year of life
(Rabbette et al, 1994). It is reasoned that the strength of the
B-H reflex is inversely related to gestational and postnatal
age because of the excessively compliant chest wall in newborns, which collapses at lung volumes less than functional
residual capacity. With decreasing lung volumes during
expiration, the B-H deflation reflex will become activated
and thereby shorten expiratory time and prolong inspiratory time. Several factors increase the strength of the B-H
reflex, including premature birth (De Winter et al, 1995;
Kirkpatrick et al, 1976), prone sleeping position (Landolfo
et al, 2008) and active sleep (Hand et al, 2004) and respiratory distress syndrome (RDS) (Rabbette et al, 1994).
RAPIDLY ADAPTING RECEPTORS DURING
DEVELOPMENT
Lung deflation, mechanical stimulation, and chemical irritants also stimulate vagal afferents of RARs causing augmented breaths and increased mucous production
589
(Widdicombe, 2006). RAR afferents synapse on RAR cells
and inspiratory Iγ neurons throughout the nTS (Kubin
et al, 2006). RAR cells receive monosynaptic excitatory
inputs from vagal afferents, which are excited by irritants,
particularly ammonia. RAR cells then send projections to
the pons respiratory group and bulbospinal inspiratory
and expiratory neurons. Stimulation of RARs as a result
of lung deflation monosynaptically activates inspiratory Iγ
neurons in the nTS, which then send axonal projections to
bulbospinal neurons (Kubin et al, 2006).
RARs are particularly important in restoring lung inflation in premature and term infants, because the excessive
compliance of the chest wall in newborn infants causes low
lung volumes during tidal breathing. RARs become active
at low lung volumes, leading to threshold activation of
vagal afferents that results in augmented breaths, restoring lung volume. The frequency of augmented breaths
is inversely related to gestational age, with premature
infants having the greatest number (Alvarez et al, 1993),
and the characteristic pattern of the augmented breath
differs between newborns and adults. Augmented breaths
in infants have a biphasic pattern with two large inspiratory efforts in succession, whereas in adults only one large
inspiratory effort is seen. Augmented breaths in preterm
and term infants are also relatively larger than those in
adults; immediately after the augmented breath, preterm
and term infants often hypoventilate or have apnea. In
contrast, ventilation often increases after the augmented
breath in adults (Qureshi et al, 2009). The increased frequency of augmented breaths and the hypoventilation and
apnea after augmented breaths in premature infants suggest that peripheral arterial chemoreceptor inputs may
have a greater influence on respiration in infants than in
adults. Peripheral arterial chemoreceptors reflexively alter
ventilation in response to acute changes in arterial CO2
and O2 tensions (discussed later). In response to an augmented breath, Pao2 rapidly increases and PaCO2 rapidly
decreases, which reduces the excitatory input from peripheral arterial chemoreceptors, leading to hypoventilation or
apnea. Peripheral arterial chemoreceptors are also key in
inducing augmented breaths, because carotid sinus nerve
denervation in animals is associated with decreased frequency of augmented breaths (Matsumoto et al, 1997). As
a result, increased activity of RARs during lung deflation
and increased sensitivity of peripheral arterial chemoreceptors in early development both likely contribute to the
increased frequency and ventilatory consequences of augmented breaths in premature infants.
C-FIBER RECEPTORS AND RESPONSES
DURING DEVELOPMENT
Pulmonary and bronchial C-fiber receptors are unmyelinated vagal fibers located throughout the respiratory tract,
extending from the nose to the lung parenchyma. Pulmonary C-fibers are accessible from the pulmonary circulation, whereas bronchial C-fibers are accessible from the
bronchial circulation and have similar sensitivity to various stimuli (Coleridge and Coleridge, 1984). C-fibers are
activated by a variety of substances: inflammatory mediators, capsaicin, lobeline, and phenylbiguanidine. Capsaicin
and phenylbiguanidine are often used experimentally to
590
PART X
Respiratory System
identify vagal afferents as C-fibers and characterize stimulus-response profiles. C-fiber simulation induces central
and local effects: cough, apnea, and laryngospasm, followed
by rapid shallow breathing, bradycardia, and hypotension
mediated by the central reflex pathways. Bronchoconstriction, increased mucous secretion, and bronchial and nasal
vasodilation are mediated by local or axon reflexes (Carr
and Undem, 2003). The central effects involve transmission of impulses to interneurons in the central nervous system, which influences the activity of autonomic or somatic
efferent nerves. The local, direct effects are mediated by
the release of neuropeptides, particularly substance P,
from C-fiber endings. By far the most common respiratory
response from C-fiber stimulation is reflex apnea characterized by prolongation of expiratory time from excitation
of postinspiratory neurons and continuous firing of central
expiratory neurons (Coleridge and Coleridge, 1984). Central integration of pulmonary C-fiber afferent information
also occurs in the subnucleus of the nTS. Unlike the relay
pathways that have been carefully delineated for the SARs,
and to a lesser extent for the RARs, the specific secondorder neurons in the nTS that are activated in response
to bronchopulmonary C-fiber stimulation have not been
completely identified. It is likely, however, that the central integration of pulmonary C-fiber stimulation occurs
within the nTS, because the interruption of synaptic transmission within the subnucleus of the nTS ablates reflex
response of pulmonary C-fibers from intraatrial injections
of phenylbiguanidine (Bonham and Joad, 1991).
In newborns, the stimulation of pulmonary C-fibers by
chemical stimulants causes bronchoconstriction and apnea
(Frappell and MacFarlane, 2005). Capsaicin-induced
apneic response and the sensitivity of the reflex was greatest in newborn rat pups younger than 10 postnatal days
(Wang and Xu, 2006). Bronchopulmonary C-fibers are
also stimulated by acidosis, adenosine, reactive oxygen
species, hyperosmotic solutions, and lung edema. Furthermore, inflammatory mediators in the local environment
sensitize C-fibers to other stimuli (Lee and Pisarri, 2001).
Pulmonary C-fiber–mediated respiratory inhibition may
be causing persistent apnea beyond term postconceptional
age in infants born at the limit of viability who have significant lung disease (Eichenwald et al, 1997). As proposed by
Lee and Pisarri (2001), C-fiber activation may also account
for the increased frequency of apnea observed in infants
with viral infections, especially caused by respiratory syncytial virus (Pickens et al, 1989).
LARYNGEAL REFLEXES
Receptors that respond to changes in upper airway pressure and chemical compounds are abundantly distributed
throughout the laryngeal mucosa. These receptors can
be slowly adapting, rapidly adapting irritant receptors, or
C-fibers. Water receptors that are simulated by hyposmolarity and low chloride content may also be involved.
Stimulation of upper airway mechanoreceptors and chemoreceptors modifies activity of upper airway muscles as
well as the pattern and timing of diaphragmatic activity.
The upper airway reflex that mediates significant cardiorespiratory effects that occur in newborns is the laryngeal
chemoreflex (LCR). The LCR is one of the most potent
defensive reflexes protecting the respiratory tract from
inadvertent aspiration (Harding et al, 1978). These receptors are stimulated by liquid in the airway, which induces
coughing, swallowing, and arousal in mature models. However, the response in immature models is apnea followed
by hypoventilation, laryngeal constriction, and swallowing.
In addition to respiratory inhibition, bradycardia, peripheral vasoconstriction, and redistribution of blood flow also
occurs. Of importance, the associated apnea and bradycardia can be life threatening in newborns (Boggs and Bartlett,
1982; Sasaki et al, 1977; Thach, 2001; Wetmore, 1993),
and in newborn infants baseline hypoxemia enhances the
severity of the apnea and bradycardia induced by the LCR
(Wennergren et al, 1989). Afferent fibers for this reflex
travel in the superior laryngeal nerve, a branch of the vagus.
These afferents synapse onto neurons in the nTS which
then send (1) excitatory projections to motoneurons of the
recurrent laryngeal nerve in the nucleus ambiguous, causing constriction of the thyroarytenoid muscle (laryngeal
constrictor) resulting in laryngospasm; (2) inhibitory projections to phrenic motoneurons in the cervical spinal cord,
inhibiting diaphragmatic contraction resulting in apnea;
and (3) an excitatory pathway to cardiac vagal neurons in
the nucleus ambiguous causing bradycardia. However, the
circuitry from second-order neurons in the nTS to cardiac
vagal afferent neurons has not been demonstrated clearly.
The LCR occurs in the fetus and likely functions to prevent
aspiration of amniotic fluid, which contains approximately
half the chloride content of pulmonary fluid (Bland, 1990;
Reix et al, 2007). With premature birth, the reflex may be
involved in the apnea and bradycardic responses associated
with feeds and gastroesophageal reflux that reaches the
larynx or nasopharynx. Whether the immature response is
still present in term infants or how the maturation of the
reflex is affected by premature birth has not been determined. Because of its profound inhibitory cardiorespiratory effects, stimulation of the LCR may be an important
reflex that is operative in some SIDS cases and infants with
acute life-threatening events (Duke et al, 2001; Gauda et al,
2007; Richardson and Adams, 2005; Thach, 2001).
CHEMICAL CONTROL OF BREATHING
(CO2/H+ and O2)
In air-breathing animals, respiratory rhythmogenesis is
primarily driven by the level of Pco2 in the blood and cerebrospinal fluid and, to a lesser extent, by oxygen tension.
In fact, for every increase of 1mm Hg in Pco2, ventilation
will increase by 20% to 30%. Specialized chemosensitive
cells in the brainstem depolarize in response to changes
in CO2/H+; they drive breathing through synaptic inputs
to respiratory-related neurons (Spyer and Gourine, 2009).
Although peripheral arterial chemoreceptors in the carotid
body also depolarize in response to increasing CO2/H+,
these receptors are primarily responsible for modifying breathing in response to changes in oxygen tension
(reviewed later). As a result of careful anatomic, physiologic, neurochemical, and genetic studies, the location
and the development of central chemoreceptors and some
of the genetic factors that drive the development of these
receptors in health and disease have been determined.
Several groups of neurons in the brainstem, specifically in
CHAPTER 43 Control of Breathing
the medullary raphe, RTN, nTS, locus ceruleus, and the
fastigial nucleus are responsive to CO2/H+ in vitro and in
vivo; therefore they are characterized as chemosensitive.
The demonstration that (1) focal acidification either with
acetazolamide (inhibits carbonic anhydrase) or local elevations of CO2 in these regions in either nonanesthetized
or anesthetized adult animals increases ventilation, and
(2) focal ablation or disruption of the region inhibits the
ventilatory response, is essential for a region to be identified as chemosensitive (Feldman et al, 2003). Although
several regions are CO2/H+ sensitive, there appears to be
some specificity in the contribution of each of the regions
in chemical control of breathing wakefulness and sleep
(Feldman et al, 2003). The greatest density of CO2/H+–
sensitive neurons in the brainstem is in serotonergic cells of
the raphe of the caudal medulla and glutamatergic neurons
of the RTN, located just below the ventral medullary surface. The serotonergic neurons in the caudal raphe project
to phrenic motoneurons, where they modulate neuronal
plasticity in response to hypoxia (Feldman et al, 2003).
The RTN receives afferent polysynaptic excitatory inputs
from peripheral arterial chemoreceptors cartoid body
(Takakura et al, 2006) and sends projections to neurons in
the VRC, including the PBC (Guyenet et al, 2008). In animals older than 7 postnatal days, the activity of the RTN
depends on intrinsic pH sensitivity and synaptic drive. The
parafacial respiratory group may be the precursor to the
RTN, and these neurons have intrinsic bursting properties starting at embryonic day 19 (Guyenet et al, 2008).
In humans the arcuate nucleus in the medulla (not to be
confused with the arcuate nucleus in the hypothalamus)
is believed to be the homologous chemosensitive region,
as described in animals based on the following findings:
(1) the arcuate nucleus is located along the ventral medullary
surface, (2) it contains a large population of glutamatergic
neurons and a smaller population of serotonergic neurons
(Paterson et al, 2006a), (3) it depolarizes in response to
hypercapnia (Gozal et al, 1994), and (4) the absence of the
arcuate nucleus in a human infant was associated with lack
of CO2 sensitivity during life (Folgering et al, 1979).
DEVELOPMENT OF CENTRAL CO2/H+
SENSITIVITY
In fetal sheep, hypercapnia causes an increase in the depth
of fetal breathing movements, with no change in inspiratory or expiratory time. In humans, maternal exposure to
CO2 also increases fetal breathing (Ritchie and Lakhani,
1980). Ventilatory responses to CO2 are present immediately after birth in most mammalian species, and CO2
sensitivity increases with maturation. However, in the
newborn rat, CO2 sensitivity is robust during the first several days of postnatal life; it declines markedly in the next
2 weeks of life and then gradually increases to reach adult
levels by the end of the 3rd week (Stunden et al, 2001).
Premature and term infants tested at 2 days of postnatal
age have modest ventilatory responses to 2% and 4% CO2,
although the strength of the ventilatory response is less in
the more immature infants (Frantz et al, 1976). In premature and late preterm infants, CO2 sensitivity increases with
postnatal age, reaching a mature response within 4 weeks
of postnatal development (Figure 43-4) (Frantz et al, 1976;
591
FIGURE 43-4 The relationship between ventilatory sensitivity to
carbon dioxide and gestational age (top panel), postnatal age (middle
panel), and the concentration of inspired oxygen (bottom panel). (From
Rigatto H, Brady J, Verduzco RT: Chemoreceptor reflexes in preterm infants:
the effect of gestational and postnatal age on the ventilatory response to inhaled
carbon dioxide, Pediatrics 55:614-620, 1975; and Rigatto H, Verduzco RT,
Cates DB: Effects of O2 on the ventilatory response to CO2 in preterm infants,
J Appl Physiol 39:896-899, 1975.)
Rigatto et al, 1975a, 1975c). Similar to the response in the
fetus, the increase in ventilation is predominately due to an
increase in tidal volume and not respiratory rate. Using the
increase in inspiratory effort against an occluded airway as
an indicator of central respiratory drive, when exposed to
2% and 4% CO2, the increase in CO2 sensitivity with postnatal development is due to an increase in central respiratory drive in human infants (Frantz et al, 1976). Premature
infants with apnea of prematurity have reduced ventilatory
responses to CO2 compared with control infants at the
same postconceptional age (Durand et al, 1985; Gerhardt
and Bancalari, 1984). This finding suggests that infants with
apnea of prematurity have reduced central respiratory drive
to breathe when compared with infants who do not have
apnea of prematurity at the same postconceptional age.
It is unknown whether maturation of synaptic inputs from
chemosensitive neurons to respiratory related neurons in
the brainstem, or maturation of intrinsic properties of chemosensitive neurons, accounts for the increase in CO2 sensitivity with early postnatal development. Although such
592
PART X
Respiratory System
studies of human infants are impossible, data from studies
performed in neonatal rats show that intrinsic responses of
chemosensitive neurons in the nTS and locus ceruleus are
already mature at birth. It is less clear whether there is a
developmental increase in the sensitivity of chemosensitive
neurons in the medullary raphe, an increase in the number
of chemosensitive neurons in the RTN, or both (Putnam
et al, 2005). However, within the first several weeks of postnatal development in the rat, the size of brainstem neurons
changes, and both dendritic arborization in the nTS and
astrocyte proliferation increase. Astrocytes contribute substantially to the pH of the extracellular milieu surrounding
chemosensitive neurons (Putnam et al, 2005). It is likely
that all these morphologic and neurochemical changes
within and between neurons and astrocytes in the brainstem contribute to maturation of CO2 sensitivity within the
early weeks of postnatal development.
MATURATION OF PERIPHERAL CO2/H+
CHEMOSENSITIVITY IN THE CAROTID BODY
The arterial chemoreceptors in the carotid body, located at
the bifurcation of the carotid artery, are primarily responsible for reflex control of ventilation in response to changes
in arterial oxygen tension. Specialized cells within the
carotid body also depolarize in response to changes in blood
CO2/H+, reflexively increasing ventilation in response to
acidosis and hypercapnia and reflexively decreasing ventilation in response to hypocapnia. Histologically, the
carotid body chemoreceptors consist of (1) type I or glomus cells, similar to presynaptic neurons, that are chemosensitive and contain neurotransmitters and autoreceptors;
(2) postsynaptic afferent nerve fibers from the carotid sinus
nerve, which oppose glomus cells (Gonzalez et al, 1994),
contain neurotransmitters and postsynaptic receptors,
and have cell bodies in the petrosal ganglion; (3) type II
cells similar to glial cells, which are not chemosensitive;
(4) microganglion cells that express cholinergic traits
(Gauda et al, 2004); and (5) blood vessels and sympathetic
fibers innervating these vessels. The commissural nucleus
of the nTS is the primary target for afferents from peripheral arterial chemoreceptors. Although these afferent processes contain glutamate, dopamine, and substance P, it
is glutamate, binding to both NMDA and non-NMDA
receptors on second-order neurons in the nTS, that is
responsible for chemical transmission of excitatory inputs
from the peripheral arterial chemoreceptors (Vardhan et al,
1993). These second-order neurons then send tonic excitatory projections to CO2 sensitivity neurons in the RTN
(see Development of Central CO2/H+ Sensitivity, earlier),
bulbospinal neurons in the dorsal respiratory group, and
VRG that synapse onto respiratory motoneurons.
In order to separate the contribution of peripheral arterial chemoreceptors from that of central chemoreceptors
on ventilatory control, studies performed in animals have
either perfused the carotid body separately from the systemic circulation or directly measured the neuronal output
from the carotid body (in vivo or in vitro) in response to
changes in arterial gas tension. With these approaches, it
has been determined that peripheral chemoreceptors sense
changes in arterial CO2/H+ more rapidly than do central
chemoreceptors (Smith et al, 2006).
Sensitivity of the peripheral arterial chemoreceptors to
CO2 increases with postnatal development in newborn
animals (Carroll et al, 1993); however, in human infants,
the contribution of peripheral arterial chemoreceptors to
ventilatory response to CO2 is difficult to delineate. Inferences can be made from the ventilatory response that is
seen within a few seconds of exposure to a particular concentration of CO2, O2, or both, because the response time
of the peripheral arterial chemoreceptors is faster than that
of the central chemoreceptors. What is inferred is that,
after the first 2 days after birth, peripheral arterial chemoreceptors in newborns are highly responsive to changes
in Pco2. Hypoventilation and apnea are frequently seen
in newborn infants after a sigh or augmented breath, in
which the Pco2 is rapidly reduced. This response is secondary to a high-gain system that is regulated at the level
of the peripheral arterial chemoreceptors. In premature
infants, the Pco2 apneic threshold is close to the Pco2 that
mediates regular breathing (Khan et al, 2005). The level
of Pco2, however, increases the response of the peripheral
arterial chemoreceptors to any level of arterial O2 tension
(Pao2), and this O2-CO2 interaction at the carotid body
increases with development (Carroll et al, 1993).
MATURATION OF HYPOXIC
CHEMOSENSITIVITY
The contribution of afferent inputs from peripheral arterial chemoreceptors during maturation is more easily
accessed by determining the acute change in ventilation in
response to changes in oxygen tension. This determination
is often made according to hypoxic gas exposure in newborn animals; an acute increase in ventilation within 30
seconds of hypoxic exposure is a measure of the strength of
the peripheral arterial chemoreceptors. Whereas hypoxic
gas exposure is occasionally used in human infants to test
peripheral arterial chemoreceptor function, the reduction
in ventilation in response to short acute exposure to hyperoxia (Dejours test) is more frequently performed to assess
the strength of the reflex during postnatal development.
Although the peripheral arterial chemoreceptors can
respond to lower than baseline Pao2 in the fetus, peripheral
arterial chemoreceptors do not contribute significantly to
fetal breathing, and functioning peripheral arterial chemoreceptors are not necessary for continuous breathing
to occur at birth (for a historical overview, see Walker,
1984). However, studies using newborn animals (e.g., cats,
dogs, rats, sheep, pigs) show that carotid body denervation
shortly after birth destabilizes breathing, often leading to
persistent apneic periods and death of the animal within
days to weeks (Gauda and Lawson, 2000). Therefore it
is believed that trophic factors from peripheral arterial
chemoreceptors acting on central mechanisms that control breathing during early postnatal development are the
key to stable rhythmogenesis throughout life. The critical
period for these trophic influences appears to be within the
first 2 weeks of postnatal development (Gauda et al, 2007).
Exposures during this critical period of development that
can lead to lifelong alterations in chemoreceptor function
include environmental exposure to the extremes of oxygen tension (chronic hypoxia and hyperoxia), intermittent
hypoxia (associated with apnea of prematurity) (Carroll,
CHAPTER 43 Control of Breathing
2003; Gauda et al, 2007), nicotine exposure (Gauda et al,
2001; Hafstrom et al, 2005), and maternal separation in
male rats (Genest et al, 2004).
Acute insensitivity to changes in oxygen tension associated with birth and then a gradual increase in hypoxic
chemosensitivity during the first 2 to 3 weeks of postnatal
development are two phases that affect the contribution of
peripheral arterial chemoreceptors on breathing. Although
oxygen tension less than 25 mm Hg stimulates the carotid
body in exteriorized fetal sheep, peripheral arterial chemoreceptors are not functional at any level of hypoxic exposure
for the first several days after birth in most mammalian species, including human infants (term and preterm). It is speculated that the resetting of peripheral arterial chemoreceptors
occurs at birth because of the increase in arterial oxygen tension during the transition from fetal to neonatal life. After
birth, there is a gradual increase in hypoxic chemosensitivity
that occurs during the first 2 weeks of postnatal life (Rigatto
et al, 1975b). Mechanisms accounting for the increase in
hypoxic chemosensitivity of peripheral arterial chemoreceptors with maturation in most mammalian species have
been reviewed recently by Gauda et al (2009). Although premature infants may have reduced peripheral chemoreceptor responses soon after birth, by 2 weeks of postnatal age
they often have enhanced peripheral arterial chemoreceptor
influences on eupneic breathing. Furthermore, premature
infants who have a greater frequency of apnea and periodic breathing have a greater reduction in ventilation when
exposed to short bouts of hyperoxia (Dejours test) (Rigatto
and Brady, 1972), suggesting a greater influence of peripheral arterial chemoreceptors on eupneic breathing in this
infant population (Al Matary et al, 2004; Nock et al, 2004).
GENETIC REGULATION OF CENTRAL
AND PERIPHERAL CHEMORECEPTOR
DEVELOPMENT
Development of central and peripheral chemoreceptors is
genetically regulated by the expression of the transcription
factor PHOX2B. PHOX2B is a homeobox gene located
on chromosome 4 that is specifically expressed in limited
types of neurons involved in autonomic processes (Dauger
et al, 2003). Its expression is required for the development
of the carotid body, nTS, and catecholaminergic neurons,
and it is expressed in chemosensitive glutamatergic neurons in the RTN that receive polysynaptic inputs from
peripheral arterial chemoreceptors (Guyenet et al, 2008).
Mutations in the PHOX2B gene affect the development of
key structures that regulate chemical control of breathing.
Congenital central hypoventilation syndrome (CCHS) is
characterized by impaired ventilatory responses to CO2
and hypoxia and other abnormalities of autonomic control
(Berry-Kravis et al, 2006). More than 90% of individuals
with CCHS have mutations in the PHOX2B gene (for a
review, see Amiel et al, 2009; Dubreuil et al, 2009.
NEONATAL HYPOXIC VENTILATORY
DEPRESSION
Although the peripheral arterial chemoreceptors function
to increase ventilation in response to hypoxia, minute ventilation significantly declines after 2 to 3minutes of hypoxic
593
exposure. This decline is commonly referred to as hypoxic
roll-off, hypoxic ventilatory decline, or hypoxic ventilatory depression. Hypoxic ventilatory depression occurs in individuals
at all ages, but it is most pronounced in the fetus and newborn (Bissonnette, 2000). Whereas the hypoxic ventilatory
decline is usually still above baseline ventilation in mature
models, the hypoventilatory response in newborns is usually below baseline ventilation and is often associated with
apnea. Mechanisms accounting for hypoxic respiratory
depression are most well characterized in the fetal animals in which the central brainstem nuclei mediating this
response are located in the pons. Transverse section of the
upper pons results in a sustained hyperventilatory response
to hypoxia in fetal and newborn sheep (Gluckman and
Johnston, 1987). Hypoxia activates expiratory neurons in
the ventrolateral pons, and chemical blockade of this area
blocks the hypoxic respiratory depression in newborn rats
(Dick and Coles, 2000).
Several neuromodulators have been implicated in mediating hypoxic ventilatory decline, including norepinephrine,
adenosine, GABA, serotonin, opioids, and platelet-derived
growth factor (see Table 43-1) (Simakajornboon and
Kuptanon, 2005). All these neuromodulators have been
shown to contribute to the ventilatory depression in newborns, but particular attention has been given to adenosine.
Degradation of intracellular and extracellular ATP is the
main source of extracellular adenosine, which then mediates its cellular effects by binding to A1, A2a, A2b, and A3
adenosine receptors. In response to hypoxia, brain adenosine levels can increase 2.3-fold in fetal sheep (Koos et al,
1994), and 100-fold in rats in response to ischemia (Winn
et al, 1981). Nonspecific adenosine receptor blockers,
particularly caffeine and methylxanthine, are commonly
administered to premature infants to increase central
respiratory drive, and aminophylline inhibits hypoxic ventilatory depression in newborn infants (Darnall, 1985).
A1-adenosine inhibitory receptors are found on respiratory related neurons (Bissonnette and Reddington, 1991).
Specific A1-adenosine receptor agonists depress phrenic
output in a reduced brainstem spinal cord preparation,
whereas A1-adenosine receptor blockers reverse this inhibitory effect (Dong and Feldman, 1995). In fetal sheep, the
hypoxic respiratory depression appears to be mediated by
excitatory A2a receptors, because blockade of A2a receptors eliminates hypoxic ventilatory roll-off in conscious
newborn sheep. Xanthine (e.g., caffeine, aminophylline)
blocks both A1 and A2a adenosine receptors; therefore
the effectiveness of methylxanthine in stabilizing ventilation and decreasing the frequency of apnea in premature
infants may be directly altering the ventilatory response
to hypoxia as well as nonspecifically increasing respiratory
drive (see Apnea of Prematurity, later).
SLEEP STATE AND BREATHING
Sleep state has a profound influence on breathing in the
fetus and newborn, and most disorders of breathing that
affect the young and old are worse during sleep. Active
sleep (AS) and quiet sleep (QS) in infants is equivalent to
rapid eye movement (REM) and non-REM sleep, respectively in older children and adults. Breathing during AS
is mostly driven by inputs from the reticular activating
594
PART X
Respiratory System
system with less influence from Pco2, whereas breathing
during QS is driven by chemical control. Similar to REM
sleep in adults, AS is associated with paralysis of striated
muscles. Although this paralysis may be necessary to prevent acting out dreams, paralysis of striated muscles that
are involved in breathing can be problematic for the newborn. AS and REM sleep are characterized by rapid eye
movements, increase in cerebral blood flow and metabolic
rate, and irregular breathing because of inhibition of the
intercostals and upper airway dilating muscles. The discoordination between chest wall muscles and the diaphragm
during active sleep causes paradoxic breathing: the chest
wall moves in during inspiration with the abdomen moving
outward. The more compliant the chest wall, the greater
propensity for paradoxic breathing, which is common in
the most immature infants. In addition, during inspiration, intrathoracic pressure becomes more negative, and
this “suction pressure” causes narrowing or collapse of the
compliant upper airway, particularly pharyngeal structures,
leading to upper airway obstruction. Paradoxic breathing movements seen on physical examination or detected
on inductive plethysmography are often interpreted as a
sign of upper airway obstruction. During QS, breathing is
characterized by smooth, regular breaths of consistent frequency and depth associated with tonic and phasic activity
of the muscles of respiration that are in phase with each
other. The chest wall and the abdomen move outward during inspiration, whereas they move inward during expiration. AS and QS can be reliably assessed at 30 to 32 weeks’
gestation (Curzi-Dascalova et al, 1993). At this gestation,
premature infants spend 80% of their sleep time in SA.
This time decreases to 50% by term, and in adulthood
REM sleep accounts for only 20% of sleep time. Sleep
state in normal infants also modifies the time to arousal
and the ventilatory responses. The time to arousal upon
exposure to a hypoxic, somatosensory or auditory stimuli
is greater in AS compared with QS in infants during the
first 6 months of life (Horne et al, 2005). Of interest, the
arousal latency in response to hypoxic stimulus in AS is
longer in preterm infants at 2 to 5 weeks postnatal age than
that of term infants at the same postnatal age (Verbeek
et al, 2008). The level of oxygen desaturation at the time
hypoxic arousal occurs is similar in the two sleep states
(Richardson et al, 2007). Respiratory pauses and periodic
breathing are more common during AS in both term and
premature infants, but the ventilatory response to CO2
is greater during QS (Cohen et al, 1991). Because of the
complexity of the ventilatory response to hypoxia and the
frequent occurrence of arousals induced by hypoxic exposure, assessing the affect of sleep state on the ventilatory
response to hypoxia in newborns is more difficult. Other
than the clear difference in arousal in response to hypoxic
stimulus between the two sleep states, differences between
sleep states on other respiratory parameters are more variable (Richardson et al, 2007).
Although most disorders of breathing, such as obstructive sleep apnea, become more severe during sleep, the
breathing disorder that is most significantly influenced by
sleep state in the newborn is CCHS. As noted previously,
CCHS is characterized by abnormalities in central chemoreception and chemical control of breathing; therefore,
with maturation, as the frequency of QS increases, so does
the severity of the disorder. During QS, CO2 sensitivity
is markedly impaired in affected individuals, and exposure
to hypercapnia does not significantly increase minute ventilation (Paton et al, 1989). In infants who died of SIDS,
their sleep state during their terminal event cannot be
known. However, adequate arousal mechanisms are key
in preventing respiratory failure and death, and impaired
arousal mechanisms are hypothesized to be causative in
SIDS. Prone sleeping position increases the percent of QS
(Horne et al, 2002), and QS is associated with increased
time to hypoxic arousal in human infants (Richardson et al,
2007). A hypoxic microenvironment from rebreathing
with defected arousal and autoresuscitative mechanisms is
hypothesized to have occurred in infants who have died
of SIDS in the face-down (i.e., prone) sleeping position
(Patel et al, 2001). Therefore sleep state can have a significant influence on control of breathing during health and
disease, especially in the newborn infant.
This chapter has outlined the neurocircuitry and neurochemistry of the respiratory network along with its synaptic inputs that undergo significant maturation during the
newborn period. Because these pathways are less developed in premature infants, premature infants have apnea of
prematurity, which often requires active therapeutic intervention and can delay hospital discharge. At preterm gestational ages, the components of the respiratory network,
similar to other developing organ systems, are plastic and
uniquely vulnerable to pathologic processes. Therefore
the episodes of intermittent hypoxemia and bradycardia
that accompany apnea of prematurity may be a cause of
acute and chronic morbidities in this high-risk population.
What follows is a discussion of the clinical presentation of
physiology and pathophysiology of breathing seen in premature infants and therapeutic interventions.
APNEA OF PREMATURITY
Respiratory pauses are universal features of preterm birth
and are most prominent in infants of the lowest gestational age. There is no consensus as to when a respiratory pause can be defined as an apneic episode. It has been
proposed that apnea be defined by its duration (e.g., longer than 15 seconds) or accompanying bradycardia and
desaturations. However, even the 5- to 10-second pauses
that occur in periodic breathing can be associated with
bradycardia or desaturation. It should be emphasized that
periodic breathing—ventilatory cycles of 10 to 15 seconds
with pauses of 5 to 10 second—is a normal breathing pattern that should not require therapeutic intervention. It
is thought to be the result of dominant peripheral chemoreceptor activity responding to fluctuations in arterial
oxygen tension. Episodic bradycardia and desaturation in
preterm infants is almost invariably secondary to apnea or
hypoventilation (Figure 43-5) (Martin and Abu-Shaweesh,
2005). The rapidity of the fall in oxygen saturation after a
respiratory pause is directly proportional to baseline oxygenation, which is related to lung volume and severity of
lung disease.
Apnea is classified traditionally into three categories
based on the absence or presence of upper airway obstruction: central, obstructive, and mixed. Central apnea is characterized by total cessation of inspiratory efforts with no
CHAPTER 43 Control of Breathing
DECREASED RESPIRATORY DRIVE
Apnea,
hypoventilation
Increased
vagal tone
Decreased O2
delivery
Bradycardia
Desaturation
Carotid
body
FIGURE 43-5 Schematic representation of the sequence of the events
whereby apnea results in various combinations of desaturation and
bradycardia. (From Martin RJ, Abu-Shaweesh JM: Control of breathing and
neonatal apnea, Biol Neonate 87:288-295, 2005.)
evidence of obstruction. In obstructive apnea, the infant
tries to breathe against an obstructed upper airway, resulting in chest wall motion without airflow through the entire
apneic episode. Mixed apnea consists of obstructed respiratory efforts, usually following central pauses. The site of
obstruction in the upper airways is primarily in the pharynx, although it also may occur at the larynx and possibly at
both sites. It is assumed that there is an initial loss of central
respiratory drive, and its recovery is accompanied by a delay
in activation of upper airway muscles superimposed on a
closed upper airway (Gauda et al, 1987). Mixed apnea typically accounts for more than 50% of long apneic episodes,
followed in decreasing frequency by central and obstructive
apnea. Purely obstructive spontaneous apnea in the absence
of a positional problem is probably uncommon. Because
standard impedance monitoring of respiratory efforts via
chest wall motion cannot recognize obstructed respiratory
efforts, mixed versus obstructive apnea is frequently identified by the accompanying bradycardia or denaturation.
THERAPEUTIC APPROACHES FOR
APNEA OF PREMATURITY
Presentation of apnea can reflect a nonspecific alteration
in either the environment (e.g., thermal) or general well
being of preterm infants. For example, neonatal sepsis can
manifest as an increase in frequency or severity of apnea,
and the underlying cause must be treated. Studies using
a rat pup model suggest that the systemically released
cytokine interleukin-1β binds to its receptor on vascular
endothelial cells at the blood-brain barrier. This binding induces synthesis of prostaglandin E2, which induces
respiratory depression in the brainstem (Hofstetter et al,
2007). These studies provide new insight into mechanisms
whereby sepsis often manifests as apnea of prematurity.
Anemia, presumably via decreased oxygen delivery, is also
frequently implicated as a cause of apnea, although transfusion of packed red cells is of variable benefit for apnea of
prematurity.
595
CONTINUOUS POSITIVE AIRWAY PRESSURE
Continuous positive airway pressure (CPAP) at 4 to 6 cm
H2O is a relatively safe and effective therapy. Because longer episodes of apnea frequently involve an obstructive
component, CPAP appears to be effective by splinting the
upper airway with positive pressure and decreasing the risk
of pharyngeal or laryngeal obstruction (Miller et al, 1985).
CPAP also benefits apnea by increasing functional residual
capacity, thereby improving oxygenation status. At a higher
functional residual capacity, time from cessation of breathing to desaturation and resultant bradycardia is prolonged.
High-flow nasal cannula therapy has been suggested as
an equivalent treatment modality to allow CPAP delivery
while enhancing infant mobility. Although this approach
is used widely, its efficacy for apnea of prematurity has not
been studied in depth. Noninvasive ventilatory strategies,
using a nasal mask to deliver intermittent positive pressure,
avoid the need for full ventilatory support in some infants.
For severe or refractory episodes, endotracheal intubation and mechanical ventilation may be needed. Minimal
ventilator settings should be used to allow for spontaneous
ventilatory efforts and to minimize the risk of barotrauma.
XANTHINE
Methylxanthine has been the mainstay of pharmacologic
treatment of apnea of prematurity for several decades.
Both theophylline and caffeine are used and have multiple physiologic and pharmacologic mechanisms of action.
Xanthine therapy appears to increase minute ventilation,
improve CO2 sensitivity, decrease hypoxic depression of
breathing, enhance diaphragmatic activity, and decrease
periodic breathing. The likely major mechanism of action
is through competitive antagonism of adenosine receptors.
Adenosine acts as an inhibitory neuroregulator in the central nervous system via activation of adenosine A1 receptors
(Herlenius et al, 1997). In addition, activation of adenosine
A2A receptors appears to excite GABAergic interneurons,
and released GABA may contribute to the respiratory inhibition induced by adenosine (Mayer et al, 2006).
Methylxanthine has some well-documented acute adverse
effects. Toxic levels can produce tachycardia, cardiac dysrhythmias, feeding intolerance, and seizures (infrequently),
although these effects are seen less commonly with caffeine at the usual therapeutic doses. Mild diuresis is caused
by all methylxanthines. The observation that xanthine
therapy causes an increase in metabolic rate and oxygen
consumption of approximately 20% suggests that caloric
demands can be increased with this therapy at a time when
nutritional intake already is compromised.
A recent large, international, multicenter clinical trial
was designed to test short- and long-term safety of caffeine therapy for apnea of prematurity. In the neonatal
period, caffeine treatment was associated with a significant
reduction in the postmenstrual ages at which both supplemental oxygen and endotracheal intubation were needed
(Schmidt et al, 2007). Of even greater interest was the
significant decrease in cerebral palsy and cognitive delay
in the caffeine-treated group (Schmidt et al, 2007). This
finding raises interesting questions regarding possible
mechanisms underlying this beneficial effect of caffeine on
596
PART X
Respiratory System
Xanthines
Less apnea
Less BPD
Altered CNS
neurotransmission
(adenosine,
GABA)
Less hypoxemic events
?
?
Downregulation
of cytokine release
from immune cells
?
Protection against
hypoxia-induced
PVL and
ventriculomegaly
?
Improved
neurodevelopmental outcome
FIGURE 43-6 Multiple proposed mechanisms are demonstrated whereby xanthine therapy for apnea of prematurity improves neurodevelopmental
outcome. These outcomes include functional changes in neurotransmitters in the brain, a decrease in hypoxemic episodes that accompany apnea,
especially in the presence of BPD, a proposed protective effect of adenosine receptor inhibition on hypoxia induced white matter injury, and the
benefit of adenosine receptor blockade on cytokine mediated lung or brain injury. (Modified from: Abu-Shaweesh JM, Martin RJ: Neonatal apnea: what’s
new? Pediatr Pulmonol 43:937-944, 2008.)
neurodevelopmental outcome (Figure 43-6). These beneficial effects include the observation in animal models that
loss of the adenosine A1 receptor gene is protective against
hypoxia-induced loss of brain matter (Back et al, 2006) and
a potential benefit of caffeine on immune mechanisms that
mediate lung and brain injury (Chavez-Valdez et al, 2009).
RELATIONSHIP TO GASTROESOPHAGEAL
REFLUX [GER]
GER is often incriminated as a cause for neonatal apnea.
Despite the frequent coexistence of apnea and GER in
preterm infants, investigations of the timing of reflux
in relation to apneic events indicate that they are rarely
related temporally. When these events coincide, there is
no evidence that GER prolongs the concurrent apnea (Di
Fiore et al, 2005). Although physiologic experiments in
animal models reveal that reflux of gastric contents to the
larynx induces reflex apnea, there is no clear evidence that
treatment of reflux affects the frequency of apnea in most
preterm infants. Therefore pharmacologic management of
reflux with agents that decrease gastric acidity or enhance
gastrointestinal motility generally should be reserved for
preterm infants who exhibit signs of emesis or regurgitation of feedings, regardless of whether apnea is present.
Therapy for such infants should begin with nonpharmacologic approaches, such as thickened feeds, because acid
suppression therapy has been shown to increase the risk
of lower respiratory infection in infants (Orenstein et al,
2009). Recent data indicate considerable differences of
opinion among neonatologists, pediatric gastroenterologists, and pediatric pulmonologists regarding diagnosis
and management of this problem (Golski, 2010).
RESOLUTION AND CONSEQUENCES
OF NEONATAL APNEA
Apnea of prematurity generally resolves by 36 to 40 weeks’
postconceptional age; however, apnea frequently persists
beyond this time in more immature infants. Available
data indicate that cardiorespiratory events in such infants
return to the baseline normal level at 43 to 44 weeks’
postconceptional age (Ramanathan et al, 2001). In other
words, beyond 43 to 44 weeks’ postconceptional age, the
incidence of cardiorespiratory events in preterm infants
does not significantly exceed that in term infants. The
persistence of cardiorespiratory events may delay hospital
discharge for a subset of infants. In these infants, apnea
longer than 20 seconds is rare; rather they exhibit frequent bradycardia to less than 70 to 80 beats/min with
short respiratory pauses (Di Fiore et al, 2001). The reason
that some infants exhibit marked bradycardia with short
pauses is unclear, but available data suggest a vagal phenomenon and benign outcome. For a few of these infants
home cardiorespiratory monitoring, until 43 to 44 weeks’
postconceptional age, is offered in the United States as
an alternative to a prolonged hospital stay. The apparent
lack of a relationship between persistent apnea of prematurity and SIDS has significantly decreased the practice
of home monitoring, with no increase in the SIDS rate.
Infants born prematurely experience multiple problems
during their time in the neonatal intensive care unit, and
many of these conditions can contribute to poor neurodevelopmental outcomes. For example, a history of prior
hyperbilirubinemia has been associated with persistent
apnea of prematurity in preterm infants and animal models (Amin et al, 2005; Mesner et al, 2008). The problem
of correlating apnea with outcome is compounded by the
fact that nursing reports of apnea severity may be unreliable, and impedance monitoring techniques will fail to
identify mixed and obstructive events. Despite these reservations, available data suggest a link between the number
of days that apnea and assisted ventilation were recorded
during hospitalization and impaired neurodevelopmental
outcome (Janvier et al, 2004). A relationship has also been
shown between delay in resolution of apnea and bradycardia beyond 36 weeks’ corrected age and a higher incidence
of unfavorable neurodevelopmental outcome (Pillekamp
et al, 2007). Finally, a high number of cardiorespiratory
events recorded after discharge via home cardiorespiratory
CHAPTER 43 Control of Breathing
597
INTERMITTENT EPISODIC HYPOXIA
?
?
?
?
Acute morbidity
Cardiovascular
Respiratory instability
Neurodevelopmental
[e.g., retinopathy
instability
[e.g., sleep disordered
disability
of prematurity]
[e.g., hypertension]
breathing]
FIGURE 43-7 Proposed acute and longer-term morbidities that might be a consequence of intermittent hypoxic episodes in early
postnatal life. (Modified from Martin RJ, Wilson CG: What to do about apnea of prematurity, J Appl Physiol 107:1015-1016, 2009.)
monitoring appear to correlate with less favorable neurodevelopmental outcome (Hunt et al, 2004). Future studies
might focus more on the incidence and severity of desaturation events, because techniques for long-term collection
of pulse oximeter data are now more advanced. Furthermore, it is likely that recurrent hypoxia is the detrimental feature of the breathing abnormalities exhibited by
preterm infants. Figure 43-7 summarizes proposed morbidities that might be attributable to intermittent hypoxic
episodes in early life. Recurrent episodes of desaturation
during early life and resultant effects on neuronal plasticity related to peripheral and central respiratory control
mechanisms may serve as an important future direction for
study. Ongoing investigation into the genetic background
of infants with CCHS may also serve to enhance our
understanding of the problem of apnea in preterm infants
(Berry-Kravis et al, 2006).
SUGGESTED READINGS
Abu-Shaweesh JM, Martin RJ: Neonatal apnea: what’s new? Pediatr Pulmonol
43:937-944, 2008.
Alheid GF, McCrimmon DR: The chemical neuroanatomy of breathing, Respir
Physiol Neurobiol 164:3-11, 2008.
Bianchi AL, Denavit-Saubie M, Champagnat J: Central control of breathing in
mammals: neuronal circuitry, membrane properties, and neurotransmitters,
Physiol Rev 75:1-45, 1995.
Gauda EB, Carroll JL, Donnelly DF: Developmental maturation of chemosensitivity to hypoxia of peripheral arterial chemoreceptors: invited article, Adv Exp
Med Biol 648:243-255, 2009.
Kinney HC, Richerson GB, Dymecki SM, et al: The brainstem and serotonin in
the sudden infant death syndrome, Annu Rev Pathol 4:517-550, 2009.
Martin RJ, Wilson CG: What to do about apnea of prematurity, J Appl Physiol
107:1015-1016, 2009.
Nock ML, DiFiore JM, Arko MK, et al: Relationship of the ventilatory response to
hypoxia with neonatal apnea in preterm infants, J Pediatr 144:291-295, 2004.
Richter DW, Spyer KM: Studying rhythmogenesis of breathing: comparison of in
vivo and in vitro models, In TRENDS in Neurosciences 24:464-472, 2001.
Schmidt B, Roberts RS, Davis P, et al: Long-term effects of caffeine therapy for
apnea of prematurity, N Engl J Med 357:1893-1902, 2007.
Thach BT: Maturation and transformation of reflexes that protect the laryngeal
airway from liquid aspiration from fetal to adult life, Am J Med 111:69S-77S,
2001.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
44
Pulmonary Physiology of the Newborn
Robert M. DiBlasi, C. Peter Richardson, and Thomas Hansen
LUNG MECHANICS
AND LUNG VOLUMES
The lungs possess physical, or mechanical, properties that
resist inflation, such as elastic recoil, resistance, and inertance. The dynamic interaction between these properties
determines the effort that must be exerted during spontaneous breathing and the resting and extreme values for the
volume of gas in the lung.
ELASTIC RECOIL
The lung contains elastic tissues that must be stretched for
lung inflation to occur. Hooke’s law requires that the pressure needed to inflate the lung must be proportional to
the volume of inflation (Figure 44-1). Conventionally, volume of inflation is plotted on the y-axis, and the distending
pressure is plotted on the x-axis. In this way, the constant
of proportionality is volume divided by pressure, or lung
compliance. Throughout the range of tidal ventilation,
the relationship between pressure and volume is linear. At
higher lung volumes, as the lung reaches its elastic limit
(i.e., total lung capacity), this relationship plateaus, making
the pressure-volume relationship nonlinear.
The lungs and the chest wall function as a unit (the
respiratory system) coupled by the interface between the
parietal and visceral pleura. Therefore, respiratory system compliance (CRS) can be partitioned into lung compliance (CL) and chest wall compliance (CCW), where
CRS = 1/CL + 1/CCW. The tendency for the lung to collapse inward at end-exhalation is balanced by the outward
recoil of the chest wall resulting in a negative (subatmospheric) intrapleural pressure. The functional residual
capacity (FRC) is the volume of gas in the lungs when the
elastic forces of these two structures reach equilibrium
(Greenspan et al, 2005). Inflation of the respiratory system above FRC requires a positive distending pressure
that must overcome the elastic recoil of both the lung
(transpulmonary pressure; alveolar-intrapleural pressure)
and the chest wall (transthoracic pressure; intrapleural
pressure-atmospheric pressure) (Gappa et al, 2006). Deflation below FRC requires an active expiratory maneuver.
Residual volume (RV) is defined as the volume of air that
cannot be expired with a forced deflation.
As depicted in Figure 44-1, the relative compliance
of the lung of the newborn is similar to that of the adult
(Krieger, 1963). However, the infant’s chest wall is composed primarily of cartilage, whereas in adults the chest
wall is completely ossified and therefore, CCW is greater
in infants than adult (see Figure 44-1) and pleural pressure is only slightly subatmospheric. Measurements of
lung and chest wall compliance suggest that the newborn
should have a lower percent RV and a lower percent FRC
598
than the adult. In fact, the percent FRC in the newborn is
equal to the adult’s, and the infant’s percent RV is slightly
greater. This seeming paradox exists because FRC and RV
are measured while the infant is breathing, and predictions from the pressure volume curves assume that there
is no air movement and passive relaxation of all respiratory
muscles (Bryan and England, 1984).
Establishment of the FRC is vital to the role of maintaining adequate lung mechanics and gas exchange. The
highly compliant newborn chest wall exerts very little outward distending pressure, and thus the lung is more prone
to collapse at end-exhalation (Hülskamp et al, 2005. Data
suggest three mechanisms by which the newborn can limit
expiratory flow and increase the intrapulmonary pressure
to maintain a normal FRC during spontaneous breathing:
(1) by increasing expiratory resistance through laryngeal
adduction (glottic narrowing), (2) by maintaining inspiratory muscle activity throughout expiration, and (3) by initiating high breathing frequencies to limit the expiratory
time (Magnenant et al, 2004; te Pas et al, 2008).
RESISTANCE
Resistance to gas flow arises because of friction between gas
molecules and the walls of airways (i.e., airway resistance)
and because of friction between the tissues of the lung and
the chest wall (i.e., viscous tissue resistance). Airway resistance represents approximately 80% of the total resistance
of the respiratory system, and tissue resistance and inertial
forces account for the remaining 20% (Polgar and String,
1966). In the newborn, nasal resistance represents nearly
half the total airway resistance; in the adult, it accounts for
about 65% of the airway resistance (Polgar and Kong, 1965).
Gas flows only in response to a pressure gradient (Figure
44-2). During laminar flow, the pressure difference needed
to move gas through the airway is directly related to the
flow rate times a constant—airway resistance. During turbulent flow, however, this pressure is directly proportional
to a constant times the flow rate squared. Gas flow becomes
turbulent at branch points in airways, at sites of obstruction,
and at high flow rates. Turbulence occurs whenever flow
increases to a point that Reynolds’ number exceeds 2000.
This dimensionless number is directly proportional to the
volumetric flow rate and gas density, and it is inversely proportional to the radius of the tube and gas viscosity. Obviously, turbulent flow is most likely to occur in the central
airways where volumetric flow is high, rather than in lung
periphery where flow is distributed across a large number
of airways. Both types of flow exist in the lung, so the net
pressure drop is calculated as follows (Pedley et al, 1977):
2
ΔP = ( K 1 × V̇ ) + (K2 × ˙V )
(1)
CHAPTER 44 Pulmonary Physiology of the Newborn
A
599
B
FIGURE 44-1 This is an idealized plot of volume as a function of distending pressure for the lung, chest wall, and respiratory system (lung plus
chest wall) of an adult (A) and an infant (B). These curves are derived by instilling or removing a measured volume of gas from the lung and allowing
the respiratory system to come to rest against a shuttered airway. At this point only elastic forces are acting on the respiratory system, and airway
pressure is equal to alveolar pressure. Intrapleural pressure can be measured using an esophageal balloon. Because airway pressure is equal to alveolar
pressure, the distending pressure for the lung can be measured as airway pressure – intrapleural pressure. The distending pressure for the chest wall is
intrapleural pressure – atmospheric pressure, and the distending pressure for the respiratory system is airway pressure – atmospheric pressure. Compliance is
the change in volume divided by the change in distending pressure. The shaded area is the resting intrapleural pressure at functional residual capacity.
Lung volumes depicted include residual volume ( RV), functional residual capacity ( FRC), and total lung capacity ( TLC).
LAMINAR FLOW
Pin
Pout
∇
P = Pin - Pout
•
∇
P = V x K1
TURBULENT FLOW
Pin
Pout
∇
P = Pin - Pout
•
P = V2 x K2
FIGURE 44-2 Gas flow ( V̇ ) through tubular structures occurs only in
the presence of a pressure gradient (Pin > Pout). For laminar flow, P is
directly proportional to V̇ .
∇
Δ P = V̇ ×
8×L×μ
π × r4
In this case, the constant of proportionality (K1) is directly related to the
length of the airway (L) and the viscosity of the gas (μ) and indirectly proportional to the fourth power of the radius of the airway (r). For turbulent
flow, ΔP is proportional to V̇ . The constant of proportionality (K2) is
directly proportional to the length of the airway and the density of the gas
and inversely proportional to the fifth power of the radius of the airway.
It is possible to take advantage of the differences between
laminar and turbulent flow to determine the site of airway obstruction in the lung. If obstruction to gas flow
is in the central airways, turbulent flow is affected the
most. Because turbulent gas flow is density dependent,
allowing the patient to breathe a less dense gas (such as
helium mixed with oxygen) reduces the resistance to gas
flow. If the site of obstruction is peripheral, the mixture of helium and oxygen does not appreciably affect
resistance.
Inflation of the lung increases the length of airways and
might therefore be expected to increase airway resistance;
however, lung inflation also increases airway diameter.
Because airway resistance varies with the fourth to fifth
power of the radius of the airway, the effects of changes
in airway diameter dominate, and resistance is inversely
proportional to lung volume (Rodarte and Rehder, 1986).
Similarly, airway resistance is lower during inspiration
than during expiration because of the effects of changes in
intrapleural pressure on airway diameter. During inspiration, pleural pressure becomes negative, and a distending
pressure is applied across the lung. This distending pressure
increases airway diameter as well as alveolar diameter and
decreases the resistance to gas flow. During expiration, pleural pressure increases and airways are compressed. Collapse
of airways is opposed by their cartilaginous support and by
the pressure exerted by gas in their lumina. During passive
expiration, these defenses are sufficient to prevent airway
closure. When intrapleural pressure is high, during active
expiration, airways may collapse, and gas may be trapped
in the lung. This problem may be accentuated in the small
preterm infant with poorly supported central airways.
INERTANCE
Gas and tissues in the respiratory system also resist accelerations in flow. Inertance is a property that is negligible
during quiet breathing and physiologically significant only
at rapid respiratory rates.
DYNAMIC INTERACTION
Compliance, resistance, and inertance all interact during spontaneous breathing (Figure 44-3). This interaction is described
by the equation of motion for the respiratory system:
(
)
1
(2)
P (t) = V [t] ×
+ (V̇ [t] × R) + (V̇ [t] × I )
C
600
PART X
Patm
=
Respiratory System
>>
=
<<
= Palv
–100
–80
Slope = –1/RC
•
Mid-volume Mid-volume
Volume
V(t)
(mL/sec)
Flow
Pleural
pressure
–60
–40
C=
Volume
Pressure
–20
Ptot
Pel
Pfr
0
Ptot
Pfr
Pel
FIGURE 44-3 Gas flows from the atmosphere into the lung only if
atmospheric pressure (Patm) is greater than alveolar pressure (Palv). At
end exhalation, when Patm equals Palv, there is no gas movement in or
out of the lung. During a spontaneous inspiration, the diaphragm contracts, the chest wall expands, and the volume in the intrathoracic space
increases. As a result, pleural pressure (Ppl) decreases relative to Patm, and
a gradient is created between Ppl and Palv, distending the lung, increasing
alveolar volume, and decreasing Palv. A gradient is also created between
Patm and Palv, and gas flows from the atmosphere into the alveolar space.
The rate of gas flow increases rapidly, reaches a maximum (peak flow),
then decreases as the alveolus fills with gas and Palv approaches Patm. At
peak inspiration, Palv equals Patm, and lung volume is at its maximum, as
is Ppl. The curved solid line connecting end expiration to end inspiration is
the total driving pressure for inspiration (Ptot). The dotted line represents
the pressure needed to overcome elastic forces alone (Pel). The difference
between the two lines is the pressure dissipated overcoming flow resistive
forces (Pfr). During exhalation, this cycle is reversed.
25
P (t) = (V [t] × 1/C) and C = V (t) /P (t)
(4)
This series of equations and Figure 44-3 demonstrate that
at points of no gas flow (end expiration and end inspiration), only elastic forces are operating on the lung. During
inflation or deflation of the lung, however, both elastic and
resistive forces are important.
Although the solution to the equation of motion for the
respiratory system is beyond the scope of this discussion, the
behavior of the respiratory system during passive exhalation
is a special situation for which a solution can be obtained
relatively easily using the occlusion technique (Lesouef
et al, 1984; McIlroy et al, 1963). Before a passive exhalation
maneuver, the infant is given a positive pressure breath, and
the airway is occluded—invoking the Hering-Breuer reflex
and a brief apnea. Airway pressure is measured, and the
occlusion is released. Expired gas flow is measured using a
pneumotachometer and integrated to volume; flow is then
plotted as a function of volume (Figure 44-4, A). During a
passive exhalation, there are no external forces acting on
15
A
10
5
0
V(t) (mL)
30
Vo
24
V(t)
(mL)
18
12
Pressure (cm H2O)
5.0
5.0
Compliance (mL/cm H2O)
Resistance (cm H2O/mL/sec) 0.05
Time Constant (sec)
0.25
63%
6
V(t) = Vo x e
86%
95%
–t/RC
98%
99%
0
0
where P(t) is the driving pressure at time, t, V[t] is the
lung volume above FRC, C is the respiratory system compliance, V̇[t] is the rate of gas flow, R is the resistance of
the respiratory system, V̇[t] is the rate of acceleration of
gas in the airways, and I is the inertance of the respiratory
system. If I is neglected, the equation simplifies to:
(
)
1
(3)
P (t) = V [t] ×
+(V̇ [t] ×R)
C
At times of zero gas flow (end expiration and end inspiration), the equation further simplifies to:
20
B
0.50
0.75
1.0
1.25
Time (sec)
FIGURE 44-4 A, Plot of flow of gas out of the lung versus volume of
gas remaining in the lung V(t) for a passive exhalation. Flow of gas out
of the lung is negative by convention. After an initial sharp increase,
flow decreases linearly as the lung empties. Static compliance of the
respiratory system is obtained by dividing the exhaled volume by the
airway pressure at the beginning of the passive exhalation. Resistance
is calculated from the slope of the flow-volume plot −(1/RC) and the
compliance. This technique has the advantage of not requiring measurements of pleural pressure and being relatively unaffected by chest wall
distortion. B, V(t) is plotted as a function of time for a passive exhalation. The graph is an exponential with the equation
V (t) = Vo × e−t / RC
V0 is the starting volume, and e is mathematical constant (roughly 2.72).
For this example, the time constant of the respiratory system (Trs) is
roughly 0.25 second. Calculations show that when exhalation persists
for a time equal to one time constant (t = 0.25 sec = 1 × Trs), 63% of the
gas in the lung is exhaled. For t = 2 Trs, 86% of the gas is exhaled; for
3 Trs, 95%; for 4 Trs, 98%; and for 5 Trs, 99%. If expiration is interrupted before a time t = 3Trs, gas is trapped in the lung.
the respiratory system (P[t] = 0), so the equation of motion
simplifies to a first-order differential equation:
(
)
1
V [t] ×
+ (V̇ [t] × R) = 0
C
Rearranging yields the linear equation
(
)
1
(5)
V̇ (t) =−
× V (t)
[RC]
where the slope is −1/RC, which can be determined using
a linear regression of V̇ (t) versus V(t).
CHAPTER 44 Pulmonary Physiology of the Newborn
This equation states that during passive exhalation, flow
plotted against volume is a straight line with slope −1/(RC).
The quantity RC has the units of time and is termed the respiratory system time constant (Trs). Trs defines the rate at which
the lung deflates during a passive exhalation (see Figure 44-4).
Time constants affect the rate of lung inflation in the same
manner in which they affect lung deflation (see Chapter 45).
WORK OF BREATHING
The work of breathing is a reflection of the amount of
energy required to overcome the elastic and resistive elements of the respiratory system and move gas into and
out of the lung during spontaneous breathing. Work of
breathing is defined as the cumulative product of distending pressure and the given volume displaced during inhalation or exhalation (Figure 44-5):
WOB = ∫ PdV
C
Tidal volume (change in mL)
Expiration
20
D
Compliance line
B
10
Inspiration
FRC
A
where P is pleural pressure in time relative to pleural pressure at end exhalation and V is the volume in time relative
to the volume at end exhalation.
The work of breathing required to ventilate the lungs of
normal newborns is approximately 10% of that required in
adults (McIlroy and Tomlinson, 1955). However, infants
have been shown to have a higher oxygen cost and lower
mechanical efficiency associated with the work of breathing than adults (Thibeault et al, 1966). In healthy infants,
the majority of the work of breathing is being done by the
diaphragm during inhalation. Approximately one third of
the total inspiratory work of breathing is related to overcoming the resistance to gas flow in the airways (Mortola
et al, 1982). Exhalation is usually passive due to potential energy stored in the lung and the chest wall at endinhalation but may become active as expiratory resistance
increases or lung volumes decrease below FRC.
MEASUREMENTS OF RESPIRATORY
SYSTEM MECHANICS
TLC
E
601
5
Tidal pressure
(change in cm H2O)
RV
FIGURE 44-5 Pressure-volume loop showing the compliance line
(AC, joining points of no flow); work done in overcoming elastic resistance (ACEA), which incorporates the frictional resistance encountered during expiration (ACDA); work done in overcoming frictional
resistance during inspiration (ABCA); and total work done during the
respiratory cycle (ABCEA), or the entire shaded area. (Figure and legend
taken from Wood BR: Physiologic principles, In Goldsmith JP, Karotkin EH,
editors: Assisted ventilation of the neonate, ed 4, Philadelphia, 2003, WB
Saunders, pp 15-30.)
Respiratory system mechanics are objective measurements used to determine the severity of lung disease,
changes in pathophysiology, response to therapeutic
interventions, and progression of lung growth and development. Table 44-1 shows mechanics values for well and
sick newborn infants. Lung mechanics in infants are typically measured during spontaneous breathing (dynamic)
and by applying maneuvers during passive breathing conditions (static).
As mentioned previously, the mechanical behavior of the
respiratory system (chest wall and lung) can be decoupled
effectively by measuring pleural pressure. Because measuring pleural pressure is not feasible, esophageal pressure
measured in the distal third of the esophagus, using an
air-filled catheter attached to a pressure transducer, can be
used to estimate pleural pressure. Thus, transpulmonary
pressure is estimated by the difference between airway
pressure and esophageal pressure and is useful in measuring lung and chest wall mechanics.
Dynamic compliance takes advantage of gas flow transiently being equal to zero at end inspiration and end expiration (see Figure 44-3). Dynamic compliance is calculated
by dividing the change in volume between these two points
in time by the concomitant change in distending pressure.
Static compliance is the compliance measured when the
infant is completely passive and can be estimated using an
inspiratory hold at end inhalation during assisted ventilation (McCann et al, 1987).
TABLE 44-1 Lung Volumes and Mechanics of Well and Sick Neonates
Measurements
Units
Normal
RDS
BPD
4-6
4-7
Tidal volume
mL/kg
5-7
FRC
mL/kg
25-30
20-33
20-30
Compliance
mL/cm H2O
1-2
0.3-0.6
0.2-0.8
Resistance
cm H2O/L/sec
25-50
60-160
30-170
FRC, Functional residual capacity.
Data from Choukroun et al, 2003; Cook et al, 1957; Gerhardt and Bancalari, 1980; McCann et al, 1987; Polgar and Promadhat, 1971; Polgar and String, 1966; Reynolds and Etsten, 1966.
602
PART X
Respiratory System
Another estimate of static respiratory system compliance requires instilling a known volume of gas into the
lung, then measuring airway pressure at equilibrium in
the absence of airflow and respiratory muscle activity (see
Figure 44-1). This single occlusion technique is used to
measure compliance during the passive exhalation maneuver described previously (see Figure 44-4). In the normal
infant, it is generally assumed that dynamic compliance is
equal to static compliance. However, in infants that are
tachypneic or who have elevated airway resistance, the
dynamic compliance may underestimate the static compliance of the lung (Katier et al, 2006).
As was mentioned earlier, measurements of compliance
are affected by lung size. For example, if a 5 cm H2O distending pressure results in a 25-mL increase in lung volume in a newborn, calculated lung compliance is 5 mL/cm
H2O. In an adult, the same 5 cm H2O distending pressure
increases the lung volume by roughly 500 mL, and calculated compliance is 100 mL/cm H2O. Although the calculated lung compliances are different, the forces needed to
carry out tidal ventilation are similar (i.e., lung function is
normal in both circumstances). This example points out
that if lung compliances are to be compared, they must be
corrected for size. This is usually performed by dividing
compliance by resting lung volume to get specific compliance. For the newborn, resting lung volume is roughly
100 mL, so specific compliance is 0.05 mL/cm H2O/mL
lung volume. For the adult, resting lung volume is nearly
2000 mL, so specific compliance is 0.05 mL/cm H2O/mL
lung volume—identical to that of the newborn. Thus, one
might expect an infant born small for gestational age to
have low lung compliance and normal specific compliance.
Lung compliance changes with volume history, meaning that it decreases with fixed tidal volumes and increases
after deep breaths that recruit air spaces that may have
been poorly ventilated or collapsed. The periodic sigh
in spontaneous breathing is typically associated with an
increase in lung compliance and in oxygenation (Frappell and MacFarlane, 2005). However, sighs in premature
infants are often followed by apnea and hypoventilation
that could lead to destabilization in infants affected with
lung disease (Qureshi et al, 2009).
Many respiratory disorders result in nonhomogeneous
increases in small airway resistance in the lung (see Table
44-1). Therefore, if lung compliance remains relatively
uniform, the product of resistance and compliance (Trs)
varies throughout the lung. During lung inflation, units
with normal resistance have the lowest Trs and fill rapidly. Units with high resistance have a longer Trs and fill
more slowly. At rapid respiratory rates when the duration
of inspiration is short, only those lung units with a short
Trs are ventilated. In effect, the ventilated lung becomes
smaller. As discussed earlier, as the lung becomes smaller,
its measured compliance decreases. Therefore, in infants
with ventilation inhomogeneities, dynamic lung compliance decreases as respiratory rate increases. This decrease
in lung compliance with increasing respiratory rate is
termed frequency dependence of compliance, and it is suggestive of inhomogeneous small airway obstruction.
Resistance of the total respiratory system can be measured using the passive exhalation technique described
previously (see Figure 44-4), or it can be calculated from
measurements of distending pressure, volume, and flow
(see Figure 44-3). Points of equal volume are chosen during inspiration and expiration. The gas flow and the distending pressure are measured at each point. The pressure
needed to overcome elastic forces should be the same for
inspiration and expiration and therefore cancel out. Total
resistance, consequently, is equal to distending pressure
at the inspiratory point minus distending pressure at the
expiratory point, divided by the sum of the respective
inspiratory and expiratory point gas flows. Investigators
have calculated compliance and resistance by measuring distending pressure, gas flow, and volume (see Figure
44-3), then fitting these measurements to the equation
of motion (see Equation 3), using multiple linear regression techniques, and solving for the coefficients 1/C and R
(Bhutani et al, 1988).
The forced oscillation technique is used to estimate
RRS in spontaneously breathing subjects (Goldman et al,
1970). A loudspeaker is used to generate a sinusoidal pressure wave (PRS) to the infant’s nose via a face mask, with
the mouth occluded, and the resulting gas flow is measured
using a pneumotachograph. The PRS / V̇ relationship,
called impedance (Z), is expressed as an amplitude ratio,
called modulus (|Z|), and a phase shift (Φ) of both signals (Desager et al, 1996). Applying the simple RLC lung
model described earlier in Equation 2 yields:
ZRS = RRS + j[ωIRS − 1/ωCRS ]
where j = √−1 and Φ = 2π (frequency). The imaginary term
on the right in the equation is termed reactance. At low
frequencies, 1/ωCRS >> ωIRS, and Φ is negative. At high
frequencies, the inertance term predominates, and Φ is
positive. At an intermediate frequency, called resonant
frequency, the effects of compliance and inertance are
equal and cancel each other. The forced oscillations can
be superimposed on spontaneous breaths (Jackson et al,
1996), making this technique suitable for infants. Forced
oscillations methods can also be used with more complex
lung models separating airway impedance from tissue
impedance (Peslin et al, 1972).
FRC is measured by inert gas dilution techniques
(helium dilution) or inert gas displacement (nitrogen washout) (Figure 44-6). Both of these techniques measure gas
that communicates with the airways. The total volume of
gas in the thorax at end expiration (thoracic gas volume
[TGV]) can be measured using a body plethysmograph and
applying Boyle’s law. This technique measures all gas in the
thorax—even trapped gas that is not in contact with the airways. Obviously, FRC measured by inert gas dilution is less
than TGV if significant volumes of trapped gas are present.
ALVEOLAR VENTILATION
The tissues of the body continuously consume O2 and
produce CO2 (Figure 44-7). The primary function of the
circulation is to pick up O2 from the lungs and deliver it
to the tissues, then to pick up CO2 from the tissues and
deliver it to the lungs. The exchange of O2 and CO2 with
the blood occurs within the alveolar volume of the lungs.
The alveolar volume acts as a “large sink” from which O2
is continuously extracted by the blood and to which CO2 is
continuously added. This mechanism for acquiring O2 from
CHAPTER 44 Pulmonary Physiology of the Newborn
A
603
B
FIGURE 44-6 A, Measurement of functional residual capacity (FRC)
by helium dilution. At end exhalation, the infant breathes from a bag
containing a known volume (Vbag) and concentration of helium (Hei)
in oxygen. The gas in the infant’s lungs dilutes the helium oxygen
mixture to a new concentration (Hef): FRC = Bag volume × (HeI − Hef)/
Hef. B, Measurement of thoracic gas volume (TGV) using a plethysmograph. The infant breathes spontaneously in a sealed body plethysmograph. At end exhalation, the airway is closed with a shutter. As the
infant attempts to inspire against the shutter, the volume of the thorax
increases and airway pressure decreases. The increase in volume of the
thorax can be measured from the change in the pressure inside of the
plethysmograph (Pbox). By Boyle’s law: P × TGV = (P − ΔP) × (TGV
+ ΔV), where P is atmospheric pressure, (P − ΔP) is airway pressure
during occlusion, and (TGV + ΔV) is thoracic volume during occlusion.
Therefore, TGV = (P − ΔP) × ΔV/ΔP. Because ΔP is small compared
with P, this can be simplified to TGV = P × ΔV/ΔP.
the atmosphere and excreting CO2 into the atmosphere is
the alveolar ventilation (Slonim and Hamilton, 1987).
The alveolar volume of the lung includes all lung units
capable of exchanging gas with mixed venous blood: respiratory bronchioles, alveolar ducts, and alveoli. Because the
conducting airways do not participate in gas exchange,
they constitute the anatomic dead space (Vd). At end exhalation, the FRC is the sum of the volume of gas in the
alveolar volume and in the anatomic dead space. During
normal breathing, the amount of gas entering and leaving
the lung with each breath is the tidal volume (Vt):
VT × respiratory rate (RR) = minute ventilation (V̇)
Part of each Vt is wasted ventilation because it moves
gas in and out of the Vd. Therefore, alveolar ventilation
( V̇A ) can be expressed as:
(6)
V̇A = (VT − VD ) × RR
Alveolar ventilation is an intermittent process, whereas
gas exchange between the alveolar space and the blood
occurs continuously. Because arterial O2 and CO2 tensions (Pao2 and Paco2) are roughly equal to the O2 and
CO2 tensions within the alveolar space, these fluctuations
in breathing could result in intermittent hypoxemia and
hypercarbia. Fortunately the lung has a large buffer—the
FRC. The FRC is four to five times as large as the Vt;
therefore, only a fraction of the total gas in the lung is
exchanged during normal breathing. This large buffer
continues to supply O2 to the blood during expiration and
acts as a sump to accept CO2 from the blood, so alveolar O2 and CO2 tensions (PaO2 and PaCO2) change little
throughout the ventilatory cycle.
FIGURE 44-7 Schematic showing coupling of alveolar ventilation to
tissue oxygen consumption.
Alveolar ventilation is linked tightly to metabolism.
When alveolar ventilation is uncoupled from the body’s
metabolic rate, hypoventilation or hyperventilation results.
During hypoventilation, less O2 is added to the alveolar space than is removed by the blood, and less CO2 is
removed from the alveolar space than is added by the blood.
As a result, PaO2 decreases and PaCO2 increases. The
net result of hypoventilation is hypoxemia and hypercapnia. Administering supplemental O2 increases the quantity
of O2 in each breath delivered to the alveolar space, and
it may prevent arterial hypoxemia. For example, suppose
a 1-kg male infant has a Vt of 6 mL, an anatomic Vd of
2 mL, and a respiratory rate of 40 breaths/min. His alveolar ventilation is 160 mL per minute ([6 mL − 2 mL] × 40/
min). If he breathes room air (21% O2), he delivers 33.6
mL of O2 to the alveolar space every minute (160 mL/min
× 0.21). If he maintains the same Vt but breathes only 20
times per minute, his alveolar ventilation decreases to 80
mL per minute, only 16.8 mL of O2 (80 mL/min × 0.21) is
delivered to the alveolar space each minute, and his PaO2
and Pao2 decrease. If he is allowed to breathe 50% O2,
O2 delivery to the alveolar space increases to 40 mL per
minute (80 mL/min × 0.50), and both his PaO2 and Pao2
increase. Because O2 administration has no effect on the
accumulation of CO2, it does not prevent hypercapnia.
Hyperventilation delivers more O2 to the alveolar space
than can be removed by the blood and removes more CO2
than can be added by the blood. As a result, PaO2 increases
and PaCO2 decreases.
604
PART X
Respiratory System
Measurements of alveolar ventilation and anatomic
Vd in the infant rely on the relationship between CO2
production (VCO2 ), V̇A , and PaCO2. The mathematical
expression of this relationship states (Cook et al, 1955):
V̇CO2
FA CO2 =
(7)
V̇A
FACO2 is the fraction of CO2 in total alveolar gas, or
FA CO2 = PACO2/(PB − 47)
(8)
Pb is the barometric pressure, and 47 mm Hg is the vapor
pressure of water at body temperature.
Therefore,
V̇A =
[ V̇CO2 × (PB − 47) ]
PA CO2
(9)
If minute ventilation ( V̇) is measured, dead space ventilation ( V̇D ) is calculated as:
(10)
V̇D = V̇ − V̇A
and Vd is calculated by dividing by the respiratory rate.
This method measures the anatomic Vd in the lung. As
seen in the next section, portions of some gas exchanging
units in the lung can also function as Vd; therefore, the total
Vd, or the physiologic Vd, may be greater than the anatomic
Vd. Physiologic Vd is calculated by substituting PaCO2 into
Equation 9 for PaCO2. When PaCO2 = PaCO2, all the Vd
is anatomic Vd, and the gas-exchanging units are all functioning normally. As physiologic Vd increases, however,
PaCO2 increases relative to PaCO2. Therefore, the difference between PaCO2 and PaCO2 (the aA.DCO2) is a measure of efficiency of gas exchange in the lung.
For clinical purposes, V̇CO2 in Equation 9 is assumed
to be a constant so that V̇A is proportional to 1/PaCO2.
Thus, increased PaCO2 means that alveolar ventilation has
decreased; decreased PaCO2 means that alveolar ventilation has increased.
VENTILATION-PERFUSION
RELATIONSHIPS
Under ideal circumstances, ventilation and perfusion of
the lung are evenly matched ( V̇/Q̇ = 1) , both in the lung
as a whole and in each individual air space. The air spaces
receive O2 from the inspired gas and CO2 from the blood.
O2 is transported into the blood, while CO2 is transported
to the atmosphere. Even though V̇/Q̇ is 1, CO2 and O2 are
exchanged in the lung at the same ratio at which they are
exchanged in the tissues: A little less CO2 is transported
out than O2 is transported in, so the respiratory exchange
ratio R equals 0.8. If there is no diffusion defect, the gas
composition of the air spaces and the blood comes into
equilibrium. N2 makes up the balance of dry gas. The sum
of partial pressures of all gases in the air spaces must equal
atmospheric pressure. The ideal alveolar gas composition
is PO2 = 100, PCO2 = 40, PN2 = 573, and PH2O = 47 (all
in mm Hg) at an atmospheric pressure of 760 mm Hg.
The ideal arterial blood composition is the same. Therefore, differences between alveolar and arterial gas composition under ideal circumstances are all zero. Knowing
the values for PaCO2 and inspired gas, ideal alveolar gas
composition can be calculated from the alveolar gas equations (Farhi, 1966):
PAO2 = PI O2 − PACO2 × [FI O2 + (1 − FI O2 ) / R] (11)
where PI O2 = FI O2 × (PB − PH2 O)
(12)
PA N2 = FI N2 × [PACO2 × (1 − R) / R + (PB − PH2 O) ] (13)
Under normal circumstances, and certainly in the presence
of lung disease, this ideal situation is not the case; some air
spaces receive more ventilation than perfusion, and others
receive more perfusion than ventilation. A reduction of ventilation may occur because of atelectasis, alveolar fluid, or
airway narrowing. Reduced ventilation in one part of the
lung may cause increased ventilation elsewhere. A reduction
of perfusion may occur if air spaces are collapsed or overdistended or because of gravitational effects, and increased
perfusion may occur in congenital heart disease. As with
ventilation, reduced perfusion in one part of the lung may
cause increased perfusion in other regions. If an air space
is relatively overventilated (high V̇/Q̇ ), its gas composition
trends toward that of inspired gas, which in the case of room
air is PO2 = 150 mm Hg and PCO2 = 0 mm Hg. If an air
space is relatively underventilated (low V̇/Q̇ ), its gas composition tends toward that of mixed venous blood, which is
PO2 = 40 mm Hg and PCO2 = 46 mm Hg. What counts is
the V̇/Q̇ ratio, not absolute values of V̇ or Q̇ (West, 1986).
To understand V̇/Q̇ imbalance, it is common to view
the lung as a three-compartment model (Figure 44-8):
V̇/Q̇ = 0 (Figure 44-8, A); V̇/Q̇ = 1 (Figure 44-8, B); and
V̇/Q̇ = infinity (Figure 44-8, C). The O2 saturation of
blood in each compartment depends on the PO2 and the
O2 dissociation curve. For illustrative purposes, in a badly
diseased lung, 50% of ventilation goes to V̇/Q̇ = 1 and
50% to V̇/Q̇ = infinity , whereas 50% of perfusion goes to
V̇/Q̇ = 1 and 50% to V̇/Q̇ = 0. Perfusion of V̇/Q̇ = 0 causes
venous admixture, whereas ventilation of V̇/Q̇ = infinity
causes alveolar Vd. The mixed alveolar gas composition
is easily calculated as the mean. For mixed arterial blood,
the PO2 must be read from the O2 dissociation curve, but
because the CO2 dissociation curve is fairly linear, the values for CO2 are easily calculated as the mean. The abnormalities in distribution of V̇ and Q̇ have created an Aa.DO2
= 70, aA.DCO2 = 23, and aA.DN2 = 32 mm Hg (see Figure
44-8). The Aa.DO2 is greater than the sum of the other two
because the O2 dissociation curve is not linear. Of course,
the situation in most lungs is not as extreme as the one illustrated. From this illustration, however, it can be seen that:
1. Open low V̇/Q̇ units produce increased Aa.DO2,
significant hypoxemia, and increased aA.DN2, but
because they are poorly ventilated and have a PCO2
close to the ideal value, they do not change the
aA.DCO2 significantly.
2. High V̇/Q̇ units produce increased Aa.DO2 without
hypoxemia and increased aA.DCO2, but because they
are poorly perfused and have a PN2 close to the ideal
value, they do not change the aA.DN2 significantly.
For the calculation of Aa.DO2 and aA.DN2, it is customary
to calculate the ideal alveolar gas composition for O2 and
N2 from the alveolar gas equations and use these values
with those measured for arterial PO2 and PN2. This
CHAPTER 44 Pulmonary Physiology of the Newborn
where Q̇va/Q̇t = venous admixture, CO2 = oxygen content,
ċ = pulmonary capillary, a = arterial, and v = mixed venous
blood. For practical application, Cv O2 is calculated from a
constant a v .O2 difference, which does introduce an error.
If an infant breathes 100% O2 for 15 minutes, most N2
is washed out of the lung, and the PO2 in open low V̇/Q̇ Q
units becomes so high that associated blood is 100% saturated with O2. The remaining venous admixture is attributed to true right-to-left shunt ( Q̇s/ Q̇t). If an infant has the
total venous admixture Q̇va/ Q̇t measured while breathing
room air, then true shunt ( Q̇s/ Q̇t) measured while breathing 100% O2, the venous admixture caused by open low
V̇/Q̇ units ( Q̇o/ Q̇t) can be calculated as the difference. The
venous admixture caused by open low V̇/Q̇ units can also be
calculated from the aA.DN2 (Markello et al, 1972):
PAO2 = 125
PACO2 = 20
PAN2 = 568
A
B
C
Q̇o
Q̇t
PAO2 = 55
PACO2 = 43
PAN2 = 600
FIGURE 44-8 Three-compartment model of the lung with
V̇
V̇
V̇
= O (A) ,
= 1 (B) ,
= infinity (C)
Q̇
Q̇
Q̇
The inspired gas is room air, and B is the ideal compartment. The sum
of alveolar gas partial pressures is always 713 mm Hg. SO2 is oxygen
saturation in capillary blood. PaO2 is read from the oxygen dissociation
curve for a saturation of 86%. By calculated differences, Aa.DO2 is 70
mm Hg, aA.DCO2 is 23 mm Hg, and aA.DN2 is 32 mm Hg.
emphasizes that part of the Aa.DO2 and aA.DN2 responsible for hypoxemia. For aA.DCO2, both an arterial and
mixed alveolar sample are required.
In the newborn, a fourth compartment in the model is
important. A significant part of the venous return may be
shunted from right to left at the foramen ovale, ductus arteriosus, pulmonary arteriovenous vessels, or lung mesenchyme
without airway development, thus adding mixed venous to
mixed arterial blood. This substantially increases the Aa.DO2
but has little effect on aA.DCO2 and no effect on aA.DN2. The
last mentioned is because there is no significant exchange of
N2 in the body, so venous and arterial PN2 are the same. The
effect on aA.DCO2 is small because venous PCO2 is only
slightly higher than arterial. From this analysis, it can be seen
that hypoxemia is produced by a true right-to-left shunt and
open low V̇/Q̇ units. Diffusional problems are not thought
to be important in the newborn. Hypoxemia may be modeled as a venous admixture, the part of mixed venous blood,
expressed as a fraction of cardiac output, that when added to
blood equilibrated with an ideal lung would produce the measured arterial oxygen saturation. It is calculated as follows:
Q̇va
Q̇t
=
Cc O2 − Ca O2
Cc O2 − Ca O2
605
(14)
=
PAN2 − PAN2
PON2 − PAN2
(15)
where Pon2 is the Pn2 in the units (see Figure 44-8), PaN2
is measured, and PaN2 is the ideal value calculated from
the alveolar gas equation. In newborns with a significant
value for true shunt, this value really represents venous
admixture as a fraction of effective pulmonary blood flow
( Q̇o/ Q̇ c .). A better estimate for Q̇o/ Q̇t can be obtained
from simple arithmetic (Corbet et al, 1974):
(
)
Q̇Va
(
)
1−
Q̇t
Q̇o
Q̇o
)
=
× (
Q̇o
Q̇t
Q̇ċ
(16)
1−
Q̇ċ
The true right-to-left shunt can then be estimated without
100% O2 breathing using the equation:
Q̇s Q̇va Q̇o
=
−
(17)
Q̇t
Q̇t
Q̇t
The normal values for the various indices of ventilationperfusion imbalance in normal newborn infants are shown
in Table 44-2.
HEART-LUNG INTERACTION
EFFECTS OF THE LUNG ON THE HEART
There exists considerable potential for the lung to affect
the heart. Because they share the thoracic cavity, changes
in intrathoracic pressure accompanying lung inflation are
transmitted directly to the heart. In addition, all of the
blood leaving the right ventricle must traverse the pulmonary vascular bed, so changes in pulmonary vascular resistance may greatly affect right ventricular function.
EFFECTS OF CHANGES IN INTRATHORACIC
PRESSURE ON THE HEART
Negative Intrathoracic Pressure
During spontaneous inspiratory efforts, the chest wall and
diaphragm move outward, intrathoracic volume increases,
and intrathoracic pressure decreases (Figure 44-9, A). The
606
PART X
Respiratory System
TABLE 44-2 Indices of Ventilation‑Perfusion Imbalance in the
Normal Newborn Breathing Room Air
Aa.DO2
mm Hg
Q̇va
Q̇t
aA.DN2
mm Hg
Q̇o
Q̇s
Q̇t
Q̇t
Newborn
25
0.25
Adult
10
0.07
10
0.10
0.15
1
7
0.05
0.02
1
aA.DCO2
mm Hg
Adapted from Nelson NM: Respiration and circulation after birth. In Smith CA,
Nelson NM, editors: The physiology of the newborn infant. Springfield, Ill, 1976,
Charles C Thomas.
heart also resides within the thoracic cavity and is subject
to the same negative intrathoracic pressure during inspiration. With a decrease in intrathoracic pressure, the heart
increases in volume, and the pressure within its chambers
decreases relative to atmospheric pressure. Analogous to the
lung, when the pressure within the heart decreases, blood is
literally sucked back into the heart from systemic veins and
arteries. On the right side of the heart, the phenomenon
serves to increase the flow of blood from systemic veins into
the right atrium, increasing right ventricular preload and
ventricular output. On the left side of the heart, ventricular
ejection is impaired. During systole, the left ventricle must
overcome not only the load imposed by the systemic vascular resistance, but also the additional load imposed by the
negative intrathoracic pressure (McGregor, 1979).
In infants with normal lungs, spontaneous respiratory
efforts result in relatively small swings in pleural pressure
(2 to 3 mm Hg) that have little effect on the pressure within
the heart. With airway obstruction or parenchymal lung
disease, however, swings in pleural pressure can be much
greater (5 to 20 mm Hg), and systemic arterial pressure may
fluctuate as much as 5 to 20 mm Hg depending on where
in the respiratory cycle ventricular systole occurs. In older
children with asthma or some other form of airway obstruction, these fluctuations in blood pressure constitute pulsus
paradoxus and are indicative of severe airway obstruction.
Positive Intrathoracic Pressure
During positive-pressure ventilation, the lung inflates
and pushes the chest wall and diaphragm outward (Figure
44-9, B). This outward push generates a pressure in the
thoracic space that is greater than atmospheric pressure.
The magnitude of the increase (relative transmission of
airway pressure to the pleural space) is determined by the
volume of lung inflation (which, in turn, is determined by
the airway pressure and lung compliance) and by the compliance of the chest wall and diaphragm. If the lung is compliant and the chest wall rigid, little airway pressure is lost
inflating the lung, but considerable pressure is generated
in the thoracic cavity as the lung attempts to push the rigid
chest wall outward. In this instance, intrathoracic pressure
(intrapleural pressure) is much greater than atmospheric
and in fact nearly equal to airway pressure. If the lung
is poorly compliant and the chest wall highly compliant,
most of the airway pressure is dissipated trying to inflate
the lungs, and little is transmitted to the thoracic cavity.
The effects of positive intrathoracic pressure on the heart
are opposite to those of negative intrathoracic pressure.
A
B
FIGURE 44-9 A, Negative intrathoracic pressure increases the
volume of the heart and decreases the pressure within the chambers.
This facilitates return of blood from the superior vena cava (SVC) and
inferior vena cava ( IVC) to the right atrium ( RA) and impedes ejection
of blood from the left ventricle ( LV ) into the extrathoracic aorta. B,
Positive intrathoracic pressure decreases the volume of the heart and
increases pressure within its chambers. This impedes blood return to the
right atrium and augments ejection of blood from the left ventricle.
The heart is compressed by the lungs and chest wall, and
blood is squeezed out of the heart and the thoracic cavity.
Return of blood from systemic veins is impaired, and right
ventricular preload and output decrease. If the increase in
intrathoracic pressure coincides with ventricular systole,
the effect is to augment left ventricular ejection and reduce
the load on the left ventricle.
In the infant undergoing positive-pressure ventilation,
the degree to which lung inflation compromises venous
return is related to the relative compliances of the lung
and chest wall. If the infant’s lung is poorly compliant
and the chest wall is compliant, as in hyaline membrane
disease, there is little effect of lung inflation on venous
return. If the infant’s lung is normally compliant but tight
abdominal distention prevents descent of the diaphragm,
intrathoracic pressure increases dramatically during positive pressure ventilation, and venous return and cardiac
output can be impaired. This mechanism may help explain
the circulatory instability of infants after repair of gastroschisis or omphalocele. A similar situation may arise in the
preterm infant with pulmonary interstitial emphysema
and massive lung overinflation. In these infants, the heart
is tightly compressed between the hyperinflated lungs, the
other structures of the mediastinum, and the diaphragm.
Venous return may be severely limited and venous pressures so increased that massive peripheral edema often
accompanies the reduction in cardiac output.
Although the effects of increased pleural pressure on the
right atrium are detrimental, the effects on the left ventricle may be extremely beneficial (Niemann et al, 1980).
During cardiopulmonary resuscitation, the chest wall is
compressed against the lung, and intrathoracic pressure
increases. Because the left ventricle is in the thorax, left
ventricular pressure increases as well. A gradient is created,
favoring flow of blood out of the ventricle and thorax and
into the extrathoracic systemic circulation. Between chest
compressions, elastic recoil causes the chest wall to pull
CHAPTER 44 Pulmonary Physiology of the Newborn
A
C
B
FIGURE 44-10 A, Effects of lung inflation on extra-alveolar vessels.
B, Effects of lung inflation on alveolar vessels. C, Effect of lung volume
on pulmonary vascular resistance ( PVR; solid line). Inflation is from
residual volume ( RV) to functional residual capacity ( FRC) to total
lung capacity ( TLC). Dashed line represents alveolar vessels; dotted line
represents extraalveolar vessels.
away from the lung and heart, decreasing pleural pressure,
favoring return of venous blood, and priming the heart for
the next chest compression. A similar phenomenon may
result in augmentation of systemic pressure when ventilator breaths coincide with ventricular systole.
EFFECT OF LUNG INFLATION ON
PULMONARY VASCULAR RESISTANCE
The pulmonary interstitium comprises three different
interconnected connective tissue compartments, each containing a different element of the pulmonary circulation
(Fishman, 1986). The first—the perivascular cuffs—consists
of a sheath of fibers that contain the preacinar pulmonary
arteries, lymphatics, and bronchi. The second consists of
the intersegmental and interlobular septa and contains pulmonary veins and additional lymphatics. The third connects these two within the alveolar septa and contains the
majority of the pulmonary capillaries. The first and second
compartments represent the extra-alveolar interstitium,
whereas the third represents the alveolar interstitium. The
perivascular cuffs are surrounded by alveoli and expand
during lung inflation (Figure 44-10, A). As a result, pressure within each cuff decreases, distending extra-alveolar
blood vessels and decreasing their resistance to blood flow.
The alveolar interstitium lies between adjacent alveoli and
contains the majority of gas-exchanging vessels in the lung.
These vessels are exposed to alveolar pressure on both sides
and during lung inflation (Figure 44-10, B) are compressed
so that their resistance to blood flow increases.
Therefore, during lung inflation (Figure 44-10, C ), the
resistance in extra-alveolar vessels decreases, whereas resistance in alveolar vessels increases. As a result, the overall
pulmonary vascular resistance decreases initially, with lung
inflation reaching its nadir at FRC, and then increases with
further inflation.
If transition from intrauterine life to extrauterine life
is to be successful, after birth all of the right ventricular
output must traverse the pulmonary vascular bed. To
some extent, this adaptation is facilitated by a reduction
in pulmonary vascular resistance that occurs with inflation
607
of the lungs (see Figure 44-10) to a stable FRC. Inflation
of the lung beyond FRC increases pulmonary vascular
resistance. If care is not taken during positive pressure
ventilation, it is possible to inflate the lung to the point
that alveolar vessels close and blood flow through the
lung is impaired. When this occurs, either cardiac output
decreases or the blood bypasses the lung via the foramen
ovale or ductus arteriosus. Clinically, this is manifest as
circulatory insufficiency from impaired right ventricular
output or hypoxemia from right-to-left shunting of blood,
or both.
EFFECTS OF THE HEART ON THE LUNG
Pulmonary Edema
Pulmonary edema is the abnormal accumulation of water
and solute in the interstitial and alveolar spaces of the lung
(Bland and Hansen, 1985; Staub, 1974). In the lung, fluid is
filtered from capillaries in the alveolar septa into the alveolar interstitium (Figure 44-11, A) and then siphoned into
the lower pressure extra-alveolar interstitium. The extraalveolar interstitium contains the pulmonary lymphatics,
and under normal conditions, they remove fluid from the
lung so that there is no net accumulation in the interstitium. Pulmonary edema results only when the rate of fluid
filtration exceeds the rate of lymphatic removal. There are
only three mechanisms by which this can occur (Figure
44-11, B): (1) the driving pressure for fluid filtration (filtration pressure) increases, (2) the permeability of the vascular bed (hence, the filtration coefficient Kf) increases, or (3)
lymphatic drainage decreases.
Increased Driving Pressure
Filtration pressure can be increased by increased intravascular
hydrostatic pressure, decreased interstitial hydrostatic pressure, decreased intravascular oncotic pressure, or increased
interstitial oncotic pressure (Figure 44-11, C and D). By far
the most common cause of increased filtration pressure is
increased intravascular hydrostatic pressure (Box 44-1). In
the newborn, intravascular hydrostatic pressure increases
with increased left atrial pressure from volume overload or a
number of congenital and acquired heart defects. In the preterm and term newborn, evidence suggests that alterations
in pulmonary blood flow that are independent of any change
in left atrial pressure may also influence fluid filtration in
the lung. Preterm infants with patent ductus arteriosus and
left-to-right shunts exhibit signs of respiratory insufficiency
before they develop any evidence of heart failure, and experiments performed in newborn lambs show that fluid filtration
in the lung can be increased by increasing pulmonary blood
flow without increasing left atrial pressure (Feltes and Hansen, 1986). In the newborn with a reduced pulmonary vascular bed, either from lung injury or from hypoplasia, cardiac
output appropriate for body size may represent a relative
overperfusion to the lung and can result in increased fluid
filtration. This phenomenon has been invoked to explain the
lung edema that often complicates the course of the infant
with bronchopulmonary dysplasia.
The exact cause of pulmonary edema that accompanies severe hypoxia or asphyxia in the newborn is still a
608
PART X
Respiratory System
B
A
C
D
FIGURE 44-11 A, In the lung, fluid is continuously filtered out of vessels in the microcirculation into the interstitium and then returned to the
intravascular compartment by the lymphatics. Only when the rate of filtration exceeds the rate of lymphatic removal can fluid accumulate in the
interstitium. Spillover of fluid into the alveolar space occurs only when the interstitial space fills or when the alveolar membrane is damaged. B, Fluid
flows out of vessels at a flow rate (Q̇f ) that is equal to the driving pressure for fluid flow (ΔP) times the filtration coefficient (Kf): (Q̇f ) = Kf × ΔP.
Kf can be thought of as the relative permeability of the vascular bed to fluid flux. Kf in the normal lung is a small number so that despite a driving
pressure of roughly 5 mm Hg, the net rate of fluid filtration is approximately 1 to 2 mL/kg per hour. C, The driving pressure for fluid flow out of the
microvascular bed represents a balance of two sets of pressures. Within the blood vessel, hydrostatic pressure tends to push fluid out of the vessel into
the interstitium. This pressure is partially opposed by a smaller hydrostatic pressure within the interstitium pushing fluid back into the blood vessel.
Within the blood vessel, there also exists a discrete oncotic pressure that results predominantly from intravascular albumin that tends to draw fluid
from the interstitium back into the blood vessel. This pressure is partially opposed by an interstitial oncotic pressure tending to draw fluid from the
blood vessel into the interstitium. D, The intravascular hydrostatic pressure must be less than pulmonary artery pressure (Ppa) for blood to flow into
the microvascular bed and greater than left atrial pressure (Pla) for blood to flow out. Intravascular pressure within the microvascular bed is roughly
equal to 0.4 (Ppa − Pla) + Pla. The interstitial hydrostatic pressure is roughly equal to alveolar pressure. The intravascular oncotic pressure can be
calculated from the plasma albumin concentration. The interstitial oncotic pressure is roughly two thirds of the intravascular oncotic pressure. The
balance of these pressures favors filtration out of the vessel (in the normal lamb, this pressure is roughly 5 mm Hg).
BOX 44-1 Increased Intravascular
Hydrostatic Pressure
INCREASED LEFT ATRIAL PRESSURE
Intravascular volume overload
Overzealous fluid administration
Overtransfusion
Renal insufficiency
Heart failure
Left-sided obstructive lesions
Left-to-right shunts
Myocardiopathies
INCREASED PULMONARY BLOOD FLOW
Normal pulmonary vascular bed
Patent ductus arteriosus
Increased cardiac output
Reduced pulmonary vascular bed
Bronchopulmonary dysplasia
Pulmonary hypoplasia
controversial issue. Data suggest that it is the result of
increased filtration pressure and not the result of any alteration in permeability. Heart failure accounts for some of
the increased filtration pressure following severe asphyxia.
In addition, there may be some element of pulmonary
venous constriction. Finally, there is evidence that hypoxia
and acidosis may redistribute pulmonary blood flow to a
smaller portion of the lung and result in relative overperfusion and edema, similar to that seen with anatomic loss
of vascular bed (Hansen et al, 1984).
Several investigators have suggested that upper airway
obstruction may cause pulmonary edema by decreasing
interstitial hydrostatic pressure relative to intravascular
hydrostatic pressure. Other data suggest, however, that
with airway obstruction, vascular pressures decrease with
intrapleural pressure in such a way that filtration pressure
remains unchanged (Hansen et al, 1985).
Hypoproteinemia in infants results in a decrease in
intravascular oncotic pressure. Its effects on filtration pressure, however, are blunted by the simultaneous decrease in
protein concentration in the interstitial space of the lung.
As a result, edema is unlikely to occur unless hydrostatic
pressure also increases (Hazinski et al, 1986).
Increased Permeability
Another possible mechanism for increased fluid filtration in the lung is a change in the permeability of the
microvascular membrane to protein—high-permeability
CHAPTER 44 Pulmonary Physiology of the Newborn
pulmonary edema. In this form of edema, the sieving properties of the microvascular endothelium are altered so that
Kf increases and patients may develop pulmonary edema
despite relatively normal vascular pressures (Albertine,
1985). Furthermore, even small changes in vascular pressures can result in a dramatic worsening of pulmonary status. High-permeability pulmonary edema usually implies
either direct or indirect injury to the capillary endothelium of the lung. Direct injuries result from local effects
of an inhaled toxin such as oxygen. Indirect injuries imply
that the initial insult occurs elsewhere in the body and that
the lung injury occurs secondarily. An example of indirect lung injury is sepsis: Neutrophils activated by bacterial toxins attack endothelial cells in the lung and increase
permeability to water and protein (Brigham et al, 1974).
Indirect injuries usually involve bloodborne mediators,
such as leukocytes, leukotrienes, histamine, or bradykinin.
Alveolar overdistention can also cause high-permeability
pulmonary edema, presumably by direct injury of the pulmonary vascular bed. This type of vascular injury probably
accounts for some of the edema that accompanies diseases
such as hyaline membrane disease and bronchopulmonary
dysplasia, in which maldistribution of ventilation results in
areas of alveolar overdistention (Carlton et al, 1990).
Decreased Lymphatic Drainage
In the normal lung, the rate of lung lymph flow is equal
to the net rate of fluid filtration, and as long as lymphatic
function can keep up with the rate of fluid filtration, water
does not accumulate in the lung. Although lymphatics can
actively pump fluid against a pressure gradient, studies
show that this ability is limited and that lung lymph flow
varies inversely with the outflow pressure (pressure in the
superior vena cava). Several groups of investigators have
demonstrated that, in the presence of an increased rate of
transvascular fluid filtration, the rate of fluid accumulation
in the lung is substantially greater if systemic venous pressure is increased (Drake et al, 1985). Recent data suggest
that the ability of the lymphatics to pump against an outflow pressure is impaired in the fetus and newborn. In fact,
in fetal lambs, lymph flow ceases at an outflow pressure of
roughly 15 mm Hg (Johnson et al, 1996). This explains
why pulmonary edema often complicates the course of
infants with bronchopulmonary dysplasia and cor pulmonale and explains the particular problem of edema with
pleural effusions complicating the postoperative course of
patients following cavopulmonary shunts.
More recently, investigators have shown that the ability
of the lymphatics to pump can be affected by other mediators with pumping increased by α-adrenergics and certain
leukotrienes and impaired by nitric oxide, β-adrenergics
(Von Der Weid, 2001), and products of hemolysis (Elias
et al, 1990).
CONGENITAL PULMONARY
LYMPHANGIECTASIS
Congenital pulmonary lymphangiectasis is a rare form of
pulmonary lymphatic dysfunction that can be characterized into two groups: (1) cases associated with congenital
heart disease and (2) cases not associated with congenital
609
heart disease. The cardiac anomalies may include hypoplastic left heart syndrome, total anomalous pulmonary
venous drainage, and pulmonary stenosis, including
Noonan syndrome (France and Brown, 1971). The group
that does not include associated cardiac anomalies may be
of early or late onset and has a wide spectrum of severity.
In some individuals, the lesion is asymptomatic, whereas in
others it can lead to severe respiratory failure, usually in
the first hours after birth but sometimes during the first
weeks or months of life. Most infants with this condition
die early in the neonatal period. Pulmonary lymphangiectasis has been reported twice as often in males and has been
seen in families (Scott Emaukpor et al, 1981).
Usually the radiologist is the first to suggest the diagnosis after observing dilated lymphatic vessels and sometimes
small accumulations of pleural fluid on the chest radiograph. The older infant may have no symptoms or mild to
moderate tachypnea with various degrees of hypoxemia. If
pleural fluid has accumulated, examination of the fluid is
important. If the infant has received milk feedings, pleural
fluid is chylous, and if not, there is an elevation in mononuclear cells and moderate protein of up to about 4%. These
findings in the absence of fever or other signs of systemic
illness are diagnostic of impaired lymphatic drainage.
Lung biopsy is probably not indicated and can be hazardous because once the distended lymphatic channels are
severed, they can leak fluid for weeks. Only if the diagnosis
is in doubt is open lung biopsy appropriate.
In noncardiac associated diffuse lymphangiectasis, only
supportive treatment is available. The long-term prognosis depends on the severity of the lesion, but this form of
pulmonary lymphangiectasis is compatible with asymptomatic life as an adult (Wohl, 1989).
SYMPTOMS OF PULMONARY EDEMA
As discussed previously, fluid filtered into the alveolar
interstitium ordinarily moves rapidly along pressure gradients into the extra-alveolar interstitium where it is removed
by the lymphatics. A delay in this process at birth can result
in clinical transient respiratory distress (see Chapter 47).
The extra-alveolar interstitium has a large storage capacity. Fluid does not begin to spill over into the alveoli and
airways until total lung water is increased more than 50%,
unless the alveolar membrane is damaged. Therefore, the
first signs and symptoms of pulmonary edema are related
to the presence of extra fluid in the interstitial cuffs of
tissue that surround airways. As fluid builds up in these
cuffs, airways are compressed, and the infants develop
signs of obstructive lung disease. The chest may appear
hyperinflated, and auscultation reveals rales, rhonchi, and
a prolonged expiration. Early in the course, chest radiographs reveal lung overinflation and an accumulation of
fluid in the extra-alveolar interstitium—linear densities of
fluid that extend from the hilum to the periphery of the
lung (the so-called sunburst appearance) and fluid in the
fissures. With more severe edema, fluffy densities appear
throughout the lung as alveoli fill with fluid (Figure 44-12).
Heart size may be increased in infants with edema from
increased intravascular pressure. Initially, infants present
with increased Paco2 secondary to impaired ventilation.
Later PaO2 decreases secondary to ventilation-perfusion
610
PART X
Respiratory System
mismatching and alveolar flooding. In adults, a ratio of
protein concentration in tracheal aspirate to that in plasma
greater than 0.5 may help to differentiate permeability
pulmonary edema from high-pressure pulmonary edema
(Fein et al, 1979).
(Rowe and Avery, 1966). Fedrick and Butler (1971) judged
massive pulmonary hemorrhage to be the principal cause
of death in about 9% of neonatal autopsies.
TREATMENT OF PULMONARY EDEMA
Pulmonary hemorrhage usually occurs between the 2nd
and 4th days of life in infants who are being treated with
mechanical ventilation. It has been associated with a wide
variety of predisposing factors, including prematurity,
asphyxia, overwhelming sepsis, intrauterine growth retardation, massive aspiration, severe hypothermia, severe Rh
hemolytic disease, congenital heart disease, and coagulopathies. It is often associated with central nervous system
injury, such as asphyxia or intracranial hemorrhage. Cole
et al (1973) studied a group of infants with pulmonary
hemorrhage to determine the clinical circumstances under
which the illness occurred as well as the hematocrit and
protein composition of fluid obtained from lung effluent
and arterial or venous blood. Their results indicated that
the lung effluent was, in most cases, hemorrhagic edema
fluid and not whole blood (i.e., as indicated by hematocrit values significantly lower than those of whole blood).
In addition, they did not find that coagulation disorders
initiated the condition but probably served to exacerbate
it in some cases. They postulated that the important precipitating factor was acute left ventricular failure caused by
asphyxia or other events that might increase the filtration
pressure and so injure the capillary endothelium of the
lung. Thus, pulmonary hemorrhage may be considered as
the extreme form of high-permeability pulmonary edema.
Pulmonary edema following central nervous system
injury probably results from increased hydrostatic pressure
and some increase in vascular permeability (Malik, 1985).
With the massive sympathetic discharge that accompanies central nervous system injury, left atrial pressure
increases, and pulmonary arteries and veins constrict. As
a result, microvascular pressure increases dramatically and
causes dramatic damage to the microvascular endothelium,
increasing its permeability to proteins and red blood cells.
In infants with overwhelming sepsis and endotoxin production, increased microvascular permeability is apparent in
the pulmonary circulation as well, undoubtedly contributing to the massive pulmonary hemorrhage sometimes seen
in this group of infants. Pulmonary hemorrhage also has
Treatment of pulmonary edema is directed at relieving
hypoxemia and lowering vascular pressures. Hypoxemia
should be treated with the administration of oxygen and,
if necessary, positive pressure ventilation. Positive end
expiratory pressure frequently improves oxygenation in
individuals with pulmonary edema by improving ventilation-perfusion matching within the lung. Available evidence suggests that positive pressure ventilation does not
reduce the rate of transvascular fluid filtration in the lung
(Woolverton et al, 1978). Optimal treatment of pulmonary edema requires correction of the underlying cause. In
infants with patent ductus arteriosus (see Figure 44-12) or
other heart disease amenable to surgery, this is often easily accomplished. In cases of permeability edema or edema
from nonsurgical heart defects, correction of the underlying cause may not be possible. In these instances, the only
remaining option is to lower vascular pressures (even in
permeability edema, lowering vascular pressures lowers
the rate of fluid filtration and may also improve lymphatic
function). This can be accomplished by lowering circulating blood volume by use of diuretics and fluid restriction,
by improving myocardial function with the use of digitalis
or other inotropic agents, or in severe cases by using a systemic vasodilator to reduce afterload and lower vascular
pressures directly.
More recent data suggest that clearance of fluid from
the alveolar space may be accelerated by β-adrenergic
agents (Frank et al, 2000) and by dopamine (Saldias et al,
1999). Whether these agents will have any clinical efficacy
remains to be determined.
PULMONARY HEMORRHAGE
Landing (1957) described pulmonary hemorrhage in 68%
of lungs of 125 consecutive infants who died in the 1st
week of life; massive pulmonary hemorrhage was found in
17.8% of neonatal autopsies at the Johns Hopkins Hospital
FIGURE 44-12 A, Preterm infant
with a large patent ductus arteriosus
and pulmonary edema. B, The same
infant 24 hours after the ductus arteriosus was closed by surgical ligation.
A
Etiology and Pathogenesis
B
CHAPTER 44 Pulmonary Physiology of the Newborn
been described occasionally in the presence of a large patent ductus arteriosus, with a left-to-right shunt that results
in high flow and high pressure injurious to the vascular bed.
Pulmonary hemorrhage is also associated with surfactant replacement therapy. Presumably the hemorrhage
results from the rapid increase in pulmonary blood flow
that accompanies improved lung function after surfactant
therapy. The contribution of the patent ductus arteriosus
to this increased blood flow remains to be determined. A
meta-analysis suggests that surfactant replacement may
be associated with an increased risk of pulmonary hemorrhage. This risk, however, is still extremely small compared
to the known benefits of surfactant replacement (Pappin
et al, 1994; Raju and Langenberg, 1993).
Diagnosis
Infants with any of the conditions mentioned previously
should be observed carefully for possible pulmonary hemorrhage. Particular note should be made of any occurrence
of blood-stained fluid from endotracheal tube aspirates,
especially if repeated suctioning shows an increase in the
amount of hemorrhagic fluid. The infant’s chest radiograph may show the fluffy appearance of pulmonary edema
in addition to the underlying pathology, and the infant
may have increased respiratory distress. Frank pulmonary
hemorrhage, when it occurs, is an acute emergency, and
the fluid has the appearance of fresh blood being pumped
directly from the vascular system, although hematocrit
values of the fluid are at least 15 to 20 points lower than
the hematocrit of circulating blood, in keeping with hemorrhagic pulmonary edema.
Treatment
Effective treatment of pulmonary hemorrhage requires
(1) clearing the airway of blood to allow ventilation; (2) use
of adequate mean airway pressure, particularly end expiratory pressure; (3) resisting the temptation to administer
611
large volumes of blood because in most cases the infant has
not had a large loss of volume, and thus, administration
of excessive volume exacerbates the increase in left atrial
pressure and hemorrhagic pulmonary edema; rather, red
cell replacement should be done as a slow administration
of packed cells after the infant’s pulmonary status has been
stabilized; and (4) evaluation of the possibility of coagulopathy and administration of vitamin K and platelets, if
appropriate.
SUGGESTED READINGS
Choukroun ML, Tayara N, Fayon M, Demarquez JL: Early respiratory system
mechanics and the prediction of chronic lung disease in ventilated preterm
neonates requiring surfactant treatment, Biol Neonate 83:30-35, 2003.
Cook CD, Cherry RB, O’Brien D, et al: Studies of respiratory physiology in the
newborn infant: I. Observations on normal premature and full term infants,
J Clin Invest 34:975-982, 1955.
Corbet AJ, Ross JA, Beaudry PH, et al: Ventilation-perfusion relationships as
assessed by aADN2 in hyaline membrane disease, J Appl Physiol 36:74-81, 1974.
Frappell PB, MacFarlane PM: Development of mechanics and pulmonary reflexes,
Respir Physiol Neurobiol 149:143-154, 2005.
Gappa M, Pillow JJ, Allen J, et al: Lung function tests in neonates and infants with
chronic lung disease: lung and chest-wall mechanics, Pediatr Pulmonol 41:291317, 2006.
Greenspan JS, Miller TL, Shaffer TH: The neonatal respiratory pump: a developmental challenge with physiologic limitations, Neonatal Netw 24:15-22, 2005.
Hülskamp G, Pillow JJ, Stocks J: Lung function testing in acute neonatal respiratory disorders and chronic lung disease of infancy: a review series, Pediatr
Pulmonol 40:467-470, 2005.
Katier N, Uiterwaal CS, de Jong BM, et al: Passive respiratory mechanics measured
during natural sleep in healthy term neonates and infants up to 8 weeks of life,
Pediatr Pulmonol 41:1058-1064, 2006.
Lesouef PN, England SJ, Bryan AC: Passive respiratory mechanics in newborns
and children, Am Rev Respir Dis 129:552-556, 1984.
McCann EM, Goldman SL, Brady JP: Pulmonary function in the sick newborn
infant, Pediatr Res 21:313-325, 1987.
McIlroy MB, Tomlinson ES: The mechanics of breathing in newly born babies,
Thorax 10:58-61, 1955.
Qureshi M, Khalil M, Kwiatkowski K, Alvaro RE: Morphology of sighs and their
role in the control of breathing in preterm infants, term infants and adults,
Neonatology 96:43-49, 2009.
Complete references and supplemental color images used in this text can be found online at
www.expertconsult.com
C H A P T E R
45
Principles of Respiratory Monitoring
and Therapy
Eduardo Bancalari and Nelson Claure
RESPIRATORY MONITORING
One of the most important aspects of newborn intensive
care is monitoring of cardiorespiratory function, ventilation, and oxygenation. This can be accomplished by
bedside monitoring devices and laboratory analysis. The
information provided by these monitoring techniques is an
essential tool in the diagnosis and treatment of respiratory
problems in the newborn.
BREATHING FREQUENCY, APNEA, AND HEART
RATE MONITORING
Transthoracic impedance is the standard method used
to monitor neonatal respiration. This technique is based
on changes in the thorax’s electrical impedance caused by
changes in gas volume during respiration measured by surface electrodes. Transthoracic impedance is mainly used to
monitor breathing frequency and to detect apnea (Olsson
and Victorin, 1970).
Breathing is detected when the change in impedance
exceeds a set threshold. A low threshold decreases sensitivity for apnea, and small disturbances such as cardiogenic
oscillations can be falsely considered as breathing. Conversely, a high threshold may lead to false apnea alarms
during shallow breathing. Monitors can automatically
adjust this threshold or the impedance amplitude, or use
sophisticated methods for detecting breathing or apnea.
Transthoracic impedance is not reliable in assessing absolute tidal volume, but it can be used to assess relative
changes.
This technique is more effective in detecting apneas of
central origin than obstructive apnea because the latter
can produce internal displacement of gas volumes that still
change impedance. More specific techniques are used for
diagnosis and classification of apnea, including thermistors
and CO2 monitors to detect gas exhalation and respiratory
inductance plethysmography that measures expansion of
the chest and abdomen. Transthoracic impedance monitors can also measure heart rate and are used to detect
brady- and tachycardia. More sophisticated devices or
built-in algorithms are utilized to detect cardiac rhythm
anomalies.
VENTILATION MONITORING
The basic clinical determination of the adequacy of ventilation in the mechanically ventilated neonate consists of
assessment of chest expansion, breathing frequency and
auscultation. This can be complemented by monitoring
tidal volume (VT) and minute ventilation utilizing flow
sensors available in most neonatal ventilators. This allows
for monitoring of spontaneous breathing, assessment of
612
respiratory system mechanics and detection of excessive or
insufficient VT, hypoventilation, and gas trapping (Becker
and Donn, 2007).
These flow sensors are either mainstream (connected
between the endotracheal tube and the ventilator circuit)
or built into the ventilator. In small infants, mainstream
flow sensors have better accuracy than those built into
the ventilator because VT and flow are only a fraction of
the gas volumes compressed in the circuit and the circulating bias flow (Cannon et al, 2000; Chow et al, 2002).
Although mainstream flow sensors are usually small and
typically have a dead space volume <1 mL, they can induce
rebreathing of exhaled gases and affect CO2 elimination in
small preterm infants (Claure et al, 2003; Figueras et al,
1997).
BLOOD GAS MONITORING
Determination of arterial blood gas status is key to the
management of respiratory failure and lung disease in
the neonate. Measurements of oxygen (Pao2) and carbon
dioxide (Paco2) tension in arterial blood are considered
the reference standard by which the efficacy and adequacy
of ventilation and oxygenation are assessed.
In order to obtain repeated arterial blood samples, placement of invasive catheters is required. In neonates, umbilical artery catheters (UACs) are commonly used during the
acute phases of respiratory failure. Samples obtained from
a UAC are the most accurate when proper sample handling and laboratory procedures are followed. However,
there are important issues that must be considered before
their placement and during their use to ensure that the
risk-to-benefit ratio remains favorable. UAC have risks
during placement and use, including perforation of the
vessels, formation of thrombi and emboli, vasospasm and
infection. UAC lines with the tip above the celiac plexus
are associated with fewer complications than those below
the renal or mesenteric artery (Barrington, 2000).
Alternatively, percutaneous lines in the radial, ulnar, or
dorsalis pedis artery are used when placement of a UAC is
not possible. When an invasive line is not available, arterial
punctures to the radial, ulnar, temporal, posterior tibialis,
or dorsalis pedis artery are done to obtain blood samples.
These latter results should be interpreted carefully because
the procedure frequently disturbs the infant and alters
blood gases. An alternative is to obtain blood samples from
the peripheral capillary bed of the medial or lateral plantar surface. These are obtained after warming of the area,
which produces hyperemia or “arterialization.” These
samples provide a gross estimate of arterial blood gases,
and they should also be interpreted cautiously (Courtney
et al, 1990). Erroneous estimation usually occurs due to
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
contamination by venous blood or air. This procedure can
also disturb the infant and alter the basal status. Errors in
blood gas measurement are often technique related. Contamination by an air bubble can reduce Pco2 and alter
the Po2 in the direction of the air’s oxygen tension. Contamination by fluids can reduce Po2 and Pco2 while also
affecting pH. Because blood cell metabolism continues,
reducing the time from sampling to analysis and cold temperature storage and transport attenuate its effects.
During conditions such as transport or when the turnaround time is important, bedside portable blood analysis
devices have proven to be most effective (Murthy et al,
1997). Another alternative measurement method is that of
indwelling electrodes inserted through a UAC that provide a continuous stream of blood gas information (RaisBahrami et al, 2002).
TRANSCUTANEOUS BLOOD GAS
MONITORING
Transcutaneous measurement of O2 (TcPo2) and CO2
(TcPco2) tensions provide a noninvasive estimate of Pao2
and Paco2, respectively. Local hyperperfusion of the skin
induced by heating creates a skin-electrode unit under the
transcutaneous sensor, and electrochemical measurements
in an electrolyte solution within this unit determine the
partial pressure of O2 and CO2.
During measurement of TcPo2, oxygen diffusing from
the capillaries to the skin is reduced by the electrode, and
the resulting electrical current is proportional to the Po2
of the capillary bed. Because skin metabolism continues,
local hyperemia is needed to maintain skin perfusion sufficiently high that the measured TcPo2 is not affected by
the skin’s O2 consumption. For this reason, accuracy of
TcPo2 depends on electrode temperature with improved
accuracy at or above 43° C (Huch et al, 1976). Under adequate conditions, TcPo2 correlates with Pao2 (Peabody et
al, 1978). However, conditions such as arterial hypotension and acidosis often result in underestimation due to
insufficient skin perfusion (Versmold et al, 1979).
During TcPco2 measurement, CO2 molecules diffusing from the capillary bed change the pH of the electrolyte
solution in the skin-electrode unit. This changes the electric potential between a reference and the electrode that
is then translated into TcPco2. Metabolism in the skin
produces CO2, and if the local perfusion is not adequate,
the measured values can exceed those of Paco2. TcPco2
measurements can also be affected by conditions affecting
peripheral perfusion (Peabody and Emery, 1985) and tend
to overestimate Paco2 in hypercapnia as local perfusion
decreases (Martin et al, 1988).
Recent reports have demonstrated improved TcPco2
measurement accuracy in preterm infants of <29 weeks’
gestation, but inconsistent precision due to large betweenpatient variability (Aliwalas et al 2005; Bernet-Buettiker
et al, 2005; Tingay et al, 2005). TcPco2 measurements
have also been shown to be tightly correlated to capillary
Pco2. Although capillary blood gases may not be the optimal reference, these are often the only available method
for long-term monitoring because indwelling lines are
available only during the acute phase of respiratory failure. In preterm infants, TcPco2 may reduce the need for
613
blood sampling and the number of painful punctures, but
the main benefit is the ability to monitor continuously.
For this reason, TcPco2 is commonly used as an adjunct
to standard blood gas sampling to provide information on
trends and respiratory stability. This is particularly useful in the management of invasive ventilatory support
where close tracking of the effects of ventilator changes
is required.
The thin epidermal skin layer of the neonate has a relatively low metabolism. Nonetheless, some transcutaneous
monitors include a metabolic correction factor. Measurements of TcPo2 and TcPco2 require a period of stabilization after sensor application until skin perfusion increases.
A tight seal around the skin-electrode unit is required for
accuracy. Similar to blood gas sampling, an air bubble
transiently lowers TcPco2 and shifts TcPo2 toward the
partial pressure of O2 in room air until the O2 is reduced.
The transcutaneous electrode is usually applied on the
thorax or the thigh. The need for high electrode temperature combined with the premature infant’s skin sensitivity
requires frequent change of the application site to avoid
thermal injury. Transcutaneous measurements have an
intrinsic delay with respect to changes occurring in the
arterial blood.
END-TIDAL CARBON DIOXIDE MONITORING
Monitoring of the partial pressure of CO2 in end-tidal
gases (PetCo2) is based on the assumption that gases measured at the airway opening at the end of exhalation represent alveolar gases and that these match the arterial levels.
PetCo2 is obtained by infrared sensors placed mainstream
or by side-stream gas sampling. PetCo2 measurements are
dependent on tidal volume size because the exhaled gas has
to carry alveolar gas. The accuracy of PetCo2 is affected by
lung disease with an increased arterial to alveolar CO2 gradient that results in underestimation of Paco2 (Sivan et al,
1992). For this reason, PetCo2 is more often used in term
or near-term neonates and pediatric patients who require
CO2 monitoring for reasons other than lung disease.
ARTERIAL OXYGEN SATURATION BY PULSE
OXIMETRY
Estimation of the oxygen saturation in arterial blood
(Sao2) by pulse oximetry (Spo2) is based on the differences in the rates of light absorption between oxygenated
and deoxygenated or reduced hemoglobin (Hb) in the red
and infrared regions of light. Deoxygenated Hb absorbs
more red light and less infrared light than oxygenated Hb.
As Sao2 increases, the ratio of the absorption of red light
to that of infrared light decreases. It is assumed that in
the circulation, changes in this ratio can only be produced
by pulsating arterial blood. In neonates, Spo2 has been
shown to correlate well with measured saturation in arterial samples (Hay et al, 1989).
The absorption by pulsatile blood is only a small fraction
of the light absorbed by tissue and venous blood. Thus,
changes in pulse amplitude or patient movement that disrupt the optical pathway from transmitter to receiver side
of the probe, or produce venous blood fluctuations, can
affect Spo2 accuracy, although newer techniques have
614
PART X
Respiratory System
reduced the effect of the latter (Hay et al, 2002). The
accuracy of Spo2 is also affected by conditions such as low
perfusion or by inappropriate placement such as excessive
tightening of the probe (Bucher et al, 1994).
Data indicate reliable detection of hypoxemia spells by
pulse oximetry (Bohnhorst et al, 2000; Hay et al, 2002).
Nonetheless, some hypoxemia episodes detected by pulse
oximetry are considered artifactual because of their temporal association with infant movement. However, increased
infant activity leading to heart rate, lung volume, and ventilation changes has been shown to trigger hypoxemia (Bolivar
et al, 1995; Dimaguila et al, 1997) with increased frequency
during periods when the infants are awake compared to
periods of active or quiet sleep (Lehtonen et al, 2002).
OXYGEN THERAPY
PRINCIPLES
Neonates presenting with acute respiratory failure or with
some degree of respiratory distress suffer abnormalities of
gas exchange that almost invariably result in hypoxemia.
Depending on the severity and duration of hypoxemia and
the metabolic demands for oxygen, this can lead to reduced
O2 availability and tissue hypoxia. Hypoxemia in the neonate can result from reduced alveolar oxygen content, low
ventilation-perfusion ratio, reduced diffusion capacity, and
extrapulmonary right-to-left shunts.
The most common form of respiratory therapy for the
neonate with hypoxemia consists of oxygen supplementation. The increased fraction of inspired oxygen (Fio2)
increases the alveolar O2 tension (PAo2) in both welland partially ventilated areas of the lung. The resulting
increase in the alveolar-arterial O2 gradient (A-aDo2) in
part compensates for the conditions producing hypoxemia
mentioned earlier. The proportion of neonates requiring supplemental O2 increases with lower gestational ages
because premature birth is associated with many of the factors that contribute to hypoxemia.
The primary goal of oxygen therapy is to maintain adequate O2 availability to the tissues, especially to the central
nervous system and the heart, and to improve an incomplete hemodynamic adaptation to extrauterine life evident
by persistently elevated pulmonary vascular resistance and
patency of the ductus arteriosus. These goals, however,
need to be attained without inducing the side effects of O2
toxicity on the eye, brain, and other organs that are common in the premature infant.
Normal Sao2 for room-air-breathing term or healthy preterm infants is reportedly greater than 93%, with Pao2 levels
above 70 mm Hg (Fenner et al, 1975; O’Brien et al, 2000;
Richard et al, 1993). Maintenance of such oxygenation levels in premature infants with lung disease and immaturity
would almost invariably require high inspired O2 levels.
Earlier strategies to “normalize” oxygenation with use of
high Fio2 in this population resulted in high rates of retinopathy of prematurity (ROP) and blindness (Campbell,
1951; Cross, 1973; Crosse and Evans, 1952). Conversely,
strict curtailment of supplemental O2 regardless of the oxygenation level was associated with increased rates of neurologic damage and death (Avery, 1960; Bolton and Cross,
1974; Patz et al, 1952).
The preterm infant is at risk for O2-induced injury
because of an immature antioxidant system that is unable
to balance the oxidative effects of O2 radicals. In the past,
severe neonatal lung injury was only partly attributed to
exposure to high Fio2. However, animal experiments
have showed that lung damage was caused by high alveolar O2 independent of Pao2 (Miller et al, 1970; Northway
et al, 1967; Taghizadeh and Reynolds, 1976). In preterm
infants, hyperoxia has been linked to neurologic damage
and impairment (Ahdab-Barmada et al, 1980; Collins et al,
2001; Haynes et al, 2003). For this reason, when supplemental O2 is administered to hypoxemic neonates, oxygenation is continuously monitored to avoid hyperoxemia.
METHODS OF ADMINISTRATION
In neonates, supplemental O2 is usually administered by
means of a head box, mask, nasal cannula, nasal continuous
positive airway pressure (CPAP), or a mechanical ventilator. In mechanically ventilated infants or in infants receiving nasal CPAP, supplemental O2 is administered by the
mixture of air and O2 in the ventilator or CPAP device. In
all four methods, verifying that correct mixing in the airO2 blender produces the desired Fio2 (by means of an O2
analyzer) is recommended.
A head box, the least invasive of these methods, is generally used for infants who only need supplemental O2.
Depending on the size of the infant, a minimum flow is
required to flush exhaled gases and to minimize entrainment of ambient air. Gas warming and humidification is
recommended to avoid drying of the airways and secretions as well as to avoid convective heat losses.
Nasal cannulas deliver a constant flow of the air-O2
mixture to the nostrils. The actual Fio2 is determined by
the delivered flow and the infant’s inspiratory flow. With
increasing flows the actual inspired O2 concentration
approaches the mixture delivered by the cannula, whereas
higher inspiratory flows, in larger infants or during periods of increased demands, reduce the actual inspired O2 by
entraining more room air (Walsh et al, 2005). The actual
Fio2 during oral breathing has not been determined. Cannula flows >1 lpm can produce positive pressure at the
nose at levels similar or above typical CPAP levels (Locke
et al, 2003), which can become very high if the prongs fit
tightly in the nostrils. Gas conditioning is recommended
to avert drying of the nose and the risk of mucosal damage (Kopelman and Holbert, 2003). Nasal cannulas have
gained popularity because they are flexible and facilitate
access to and mobility of the infant.
TREATMENT STRATEGIES
The effects of the introduction of continuous monitoring of
oxygenation in the care of preterm infants in relation to ROP
have been inconsistent (Bancalari et al, 1987; Grylack, 1987;
Yamanouchi et al, 1987). Data showing that ROP severity
was associated with duration of hyperoxemia emphasized the
importance of monitoring and curtailing hyperoxemia (Flynn
et al, 1987). However, infants with severe ROP were also
found to spend considerable periods of time in hypoxemia.
Continuous monitoring of arterial oxygen saturation
by pulse oximetry (Spo2) has become an important and
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
standard component of neonatal intensive care. The use of
Spo2 has recently been extended to the delivery room for
titration of Fio2 during resuscitation (Dawson et al, 2009;
Escrig et al, 2008). Although the optimal range of Spo2 in
premature infants has not been fully defined, existing data
suggest deleterious effects of hyperoxemia, with higher rates
of ROP and worse respiratory course in neonatal centers
that have tolerant policies toward high Spo2 levels (Anderson et al, 2004; Tin et al, 2001). Observational data indicate
that more restrictive policies toward high Spo2 can reduce
rates of ROP (Chow et al, 2003; Wright et al, 2006). Tolerance of high Spo2 levels in convalescent infants beyond
the neonatal period has been proposed to improve growth
and neurologic outcome, as well as to stop the progression
of threshold ROP. However, clinical trials showed minimal
benefits that were outweighed by deterioration in lung function caused by the additional O2 required to maintain higher
Spo2 (Askie et al, 2003; STOP-ROP Study Group, 2000).
Implementation of policies to curb hyperoxemia should
also consider the potential deleterious effects of insufficient oxygenation. Hypoxemia can increase patency of the
ductus arteriosus (Noori et al, 2009; Skinner et al, 1999) as
well as increase the resistance of the pulmonary vasculature
and airways, particularly in infants with established lung
disease (Abman et al, 1985; Cassin et al, 1964; Halliday
et al, 1980; Tay-Uyboco et al, 1989; Teague et al, 1988).
Observational data showed that policies targeting lower
Spo2 ranges were associated with better outcomes (Chow
et al, 2003; Wright et al, 2006). However, it is unknown how
closely such ranges were actually maintained and is possible
that the side effects of lower Spo2 levels were not detected
because infants were maintained above those ranges most of
the time. Those findings should be interpreted with caution
in face of recent evidence from a large blinded randomizedcontrolled trial showing increased mortality when targeting
a lower Spo2 range (Carlo et al, 2010).
The use of pulse oximetry to avoid hyperoxemia and
hypoxemia must be done in the context of the sigmoidshaped oxygen dissociation curve between Pao2 and Spo2.
Although Spo2 levels in the upper range can be associated
with a wide range of Pao2 levels (Brockway and Hay, 1998),
data indicate that a Spo2 threshold of 93% or 94% can
avoid most Pao2 values >80 mm Hg (Bohnhorst et al, 2002;
Castillo et al, 2008; Hay et al, 1989; Poets et al, 1993). On
the other end, most Spo2 values <80% or 85% are associated with Pao2 <40 mm Hg. Thus, a moderate and stepwise
increase in Fio2 when Spo2 decreases below this threshold
can adequately correct hypoxemia and is unlikely to produce hyperoxemia unless the additional oxygen is excessive or prolonged unnecessarily. When Fio2 is increased in
response to a hypoxemia spell, Spo2 should be continuously
observed, and Fio2 should be weaned as the spell ends.
Policies of oxygenation monitoring should clearly identify both intended range and the alarm limits of Spo2.
Although in many centers they coincide, these ranges serve
different purposes. The intended range usually defines a
prescribed basal level of oxygenation to be maintained by
the staff, whereas the alarm limits usually define specific
conditions that require immediate intervention.
In practice, the use of the low and high Spo2 alarms differs. The low Spo2 alarm, typically set between 85% and
88%, is generally used to detect acute hypoxemia. Setting
615
Spo2 alarms below these levels is often aimed at ensuring
that the caregiver will respond only to the most severe
events, thereby indirectly avoiding overuse of supplemental O2. The high Spo2 alarm is primarily used to avoid
hyperoxemia and is typically set between 93% and 95%.
The high Spo2 alarm level is quite important because it
has been shown to be closely linked to the actual mean
Spo2 observed in preterm infants receiving supplemental
O2 (Hagadorn et al, 2006).
Observational data have shown that preterm infants
spend only half the time within the intended range of
Spo2, with the remaining 30% of the time above and 20%
of the time below this range (Hagadorn et al, 2006; Laptook et al, 2006). Setting Spo2 alarm limits within 2% of
the intended range produces some improvement, but not
strikingly so. Staff compliance to Spo2 alarms plays an
important role, with the prescribed high Spo2 alarm level
being frequently altered by the staff (Clucas et al, 2007).
Insufficient staff education and communication can also
influence the maintenance of Spo2 within the intended
range. However, a recent survey showed that only a third
of the caregivers were aware of and could identify their
center’s oxygenation policies (Nghiem et al, 2008).
Policies for oxygen supplementation for preterm infants
who reach term-corrected postmenstrual ages (e.g., 36
weeks) vary significantly between centers (Ellsbury et al,
2002). Centers with policies that target higher Spo2 have
greater proportions of infants on supplemental O2, and
many of those infants could be off O2 if lower Spo2 levels
are tolerated or if their actual needs are frequently evaluated (Walsh et al, 2004). This is also the case during the
discharge period as well as during home oxygen therapy.
Many preterm infants present with hypoxemia spells, and
these are more frequent in infants with evolving chronic
lung disease (Bolivar et al, 1995; Dimaguila et al, 1997; Garg
et al, 1988). These spells often require a transient increase
in Fio2, but a delayed response can prolong the hypoxemia
spell, whereas a delayed weaning of Fio2 after hypoxemia
ends can induce hyperoxemia. It is also evident that caregivers often tolerate or maintain high Spo2 levels with the
purpose of reducing the frequency of the spells or attenuating their severity (Claure et al, 2009, 2011). However, this
is not truly effective and may actually increase the risk of
ROP (McColm et al, 2004). Neonatal center policies should
clearly define the response of the caregiver to these events
to minimize both excessive and inadequate oxygenation.
Currently, automated systems to adjust the inspired
oxygen for maintenance of Spo2 within a set range are
being developed and tested (Claure et al, 2001, 2009, 2011;
Urschitz et al, 2004). Reports indicate improvements in
terms of maintenance of an oxygenation range as well as
reductions in hyperoxemia, exposure to supplemental O2,
and staff workload with these automated systems. Large
trials are necessary to determine the effects of this form of
Fio2 control on short- and long-term neonatal outcomes.
At the present time, most clinical effort is focused on avoiding the extreme high and low ranges of Spo2. Forthcoming
are the findings of multicenter trials being conducted to
compare the effects of maintaining Spo2 within different
ranges in terms of ophthalmic, neurologic, and respiratory
outcome. The information obtained from these trials will
further refine the oxygen management strategies.
616
PART X
Respiratory System
NONINVASIVE RESPIRATORY
SUPPORT
Because of the severe complications associated with invasive mechanical ventilation, there has been a persistent
search for less invasive alternatives to support infants with
respiratory failure. This is especially relevant in the small
preterm infant, who is more susceptible to acute complications and chronic pulmonary sequelae. Alternatives have
ranged from transdermal oxygenation to lung volume
maintenance by sternal traction. The strategies that have
shown to be most clinically effective are nasal continuous
positive airway pressure and, more recently, nasal intermittent positive pressure ventilation. These strategies can
be used to support infants with respiratory failure in lieu of
invasive mechanical ventilation.
NASAL CONTINUOUS POSITIVE AIRWAY
PRESSURE
CPAP was introduced in 1971 by George Gregory to stabilize lung volume in preterm infants with respiratory distress syndrome (RDS) (Gregory et al, 1971). Initially, the
constant pressure was delivered through a mask or head
box and later was applied through nasal prongs. With the
introduction of surfactant therapy, more infants were intubated and treated with mechanical ventilation, and the use
of nasal CPAP declined for a period of several years; in the
past decade, however, it has been reintroduced as a safer
alternative to mechanical ventilation.
Physiologic Effects
Nasal CPAP (N-CPAP) prevents alveolar collapse and
stabilizes functional residual capacity, thereby reducing
pulmonary shunts and resulting in improved oxygenation.
N-CPAP also stabilizes large and small airways and can
decrease the work of breathing and obstructive apnea episodes. By preventing alveolar closure and reopening with
each breath, N-CPAP can also preserve surfactant.
Indications
Nasal CPAP is used primarily in infants with RDS who
because of surfactant deficiency have decreased functional
residual capacity (FRC). N-CPAP is also effective in the
transition period immediately after birth while reabsorption of fetal lung fluid and establishment of FRC is occurring. N-CPAP is also effective in reducing apneic episodes
and in stabilizing respiratory function after extubation
(Locke et al, 1991; Martin et al, 1977; Miller et al, 1985).
Nasal CPAP is also used in infants with increased pulmonary blood flow and pulmonary edema secondary to heart
lesions with left-to-right shunting such as patent ductus
arteriosus (PDA). Nasal CPAP can also be used effectively
in infants with airway obstruction.
Devices for Nasal CPAP
Nasal CPAP has been applied using a variety of systems
and devices. Most of the evidence suggests that best results
are obtained using double short nasal prongs (De Paoli
et al, 2002). The devices used to generate the pressure can
be a water column such as the one used for “bubble CPAP”
or an adjustable valve at the end of a continuous-flow system or a variable-flow system. Although there are data suggesting that some of these systems are superior to others, in
clinical practice the stability and permeability of the nasal
interface is probably the most important factor determining the success or failure of this form of respiratory support. Recently, nasal cannulas have been used as a method
for generating nasal CPAP. The limitation of this method
is that it is difficult to control and measure the actual pressures and inspired oxygen concentrations that are delivered
to the infant because both depend on the gas flow and the
leaks around the cannulas and through the mouth.
Clinical Application
As mentioned earlier, the major indication for N-CPAP use
in preterm infants is RDS. There is considerable debate as
to the ideal time to start N-CPAP and about the population
of infants in which it should be used. In recent years most
centers have used N-CPAP soon after birth in infants who
have sufficient respiratory drive. The exceptions are infants
who are depressed at birth or those who have severe respiratory failure and are intubated to administer surfactant.
The success rate with this strategy depends on the maturity
of the infant and the severity of the respiratory failure, but
more than 50% of preterm infants who are not depressed
at birth can be managed successfully with N-CPAP, avoiding the use of invasive ventilation (Ammari et al, 2005;
Dani et al, 2004; Kamper and Ringsted, 1990; Reininger
et al, 2005). The sooner N-CPAP is started, the better the
results are in infants with RDS (Gittermann et al, 1997;
Hegyi and Hiatt, 1981; Jonsson et al, 1997; Verder et al,
1999). It has been suggested that the early use of N-CPAP
instead of invasive ventilation could lead to a reduction
in the incidence of bronchopulmonary dysplasia (BPD).
However, two large prospective randomized trials addressing this question did not show that early use of N-CPAP
reduces BPD (Finer et al, 2010; Morley et al, 2008).
The other frequent indication for N-CPAP is to stabilize respiratory functions after weaning from mechanical ventilation and extubation (Andreasson et al, 1988;
Engelke et al, 1982; Higgins et al, 1991). Although it is not
clear whether the use of N-CPAP in this situation reduces
the need for reintubation, it clearly prevents a deterioration of respiratory function (Davis and Henderson-Smart,
1999; Peake et al, 2005).
The other indication for N-CPAP is to reduce the incidence of apneic episodes in preterm infants, in which it has
been shown to be very effective (Martin et al, 1977; Miller
et al, 1985).
The levels of N-CPAP that are used in clinical practice
range from 3 to 8 cm H2O, but there are few data to justify
a specific level. In practice, the level of N-CPAP used is
determined by the severity of the lung disease, reflected
by the inspired oxygen concentration and the degree of
lung expansion in the chest radiograph. The more severe
the disease is, the higher the level of N-CPAP that is used.
As the respiratory condition improves and the oxygen
requirement decreases the level of N-CPAP is reduced to
3 to 4 cm H2O before removing the prongs.
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
Complications
617
The complications of N-CPAP can be related to the pressure that is applied or to the interface with the nose. The
application of excessive pressure can cause overdistention
and alveolar rupture with pulmonary interstitial emphysema and pneumothorax. It can also reduce venous return,
increase pulmonary vascular resistance, and reduce cardiac
output. Overdistention of the lung can also reduce compliance and induce hypoventilation.
When the nasal prongs are too large or are applied with
too much pressure over the nasal septum, they can produce erosions or pressure necrosis that sometimes makes it
impossible to continue the N-CPAP. Avoiding these complications and keeping the nasal prongs in place is a task
that requires considerable time and skill. During N-CPAP
there is a risk of gas being pushed into the stomach.
A nasogastric catheter is often used to avoid accumulation
of gas and gastric distention.
A/C or PSV. Internal flow sensors are available in many
new ventilators. However, data on their use in neonatal
noninvasive ventilation are lacking, and it is unlikely that
small infants are able to produce flows large enough to be
detected by the internal sensors.
Devices that alternate between two levels of positive airway pressure (Bi-PAP) are also available. In these devices
the increase in pressure is achieved by an increase in circulating flow instead of a valve. For this reason, in Bi-PAP
the increase in pressure is small compared to that in conventional ventilators, and it is achieved over a relatively
long time. Synchronization of the increase in airway pressure with the infant’s inspiration using the Graseby capsule is available in some countries.
Another approach has been that of applying high-frequency ventilation nasally, but it is unclear what proportion of the oscillating pressure is transmitted to the infant’s
airways.
NONINVASIVE VENTILATION
Clinical Application
Noninvasive forms of respiratory support have been introduced with the aim of avoiding the use of invasive ventilation in infants in respiratory failure. N-CPAP is effective
in improving gas exchange by stabilizing lung volume and
the airways. However, it is not fully effective in maintaining ventilation in infants with weak inspiratory effort and
inconsistent respiratory drive.
Intermittent positive pressure ventilation via nasal
devices was among the original forms of support used
in preterm infants in respiratory failure (Llewellyn et al,
1970). Recently, it has been reintroduced in neonatal care
for indications including respiratory distress and apnea and
to facilitate weaning from invasive mechanical ventilation.
In infants with apnea, the cycling positive pressure at the
upper airway may produce an intermittent stimulus that
prevents or attenuates the duration of breathing pauses of
central origin. In infants with central apnea, resumption of
breathing following ventilator cycles has been described,
and the reduction of apnea in comparison to N-CPAP is
more striking among infants with more frequent apnea
spells (Bisceglia et al, 2007; Lin et al, 1998; Ryan et al,
1989).
Noninvasive ventilation can improve gas exchange and
ventilation compared to N-CPAP in infants with respiratory insufficiency during the first hours after birth as well
as during weaning from mechanical ventilation (Bisceglia
et al, 2007; Moretti et al, 1999). In contrast, infants with
mild respiratory distress who are stable on N-CPAP and
are able to maintain adequate gas exchange only reduced
their spontaneous breathing effort when receiving noninvasive ventilation (Aghai et al, 2006; Ali et al, 2007;
Chang et al, 2011). In the preterm infant, the chest wall
is excessively compliant, and the breathing effort is in part
dissipated by an inward motion of the chest. Hence, the
reduced breathing effort with noninvasive ventilation can
also be explained by a reduction in chest distortion (Ali
et al, 2007; Kiciman et al, 1998).
In spite of applying cycles of relatively small positive
pressure, Bi-PAP can improve CO2 elimination and oxygenation (Migliori et al, 2005) compared to N-CPAP.
Noninvasive application of high-frequency ventilation was
also shown to be useful as a rescue for avoidance of intubation in a group of infants failing N-CPAP (van der Hoeven
et al, 1998).
In addition to the effects on apnea, ventilation, and
breathing effort, it is possible that the increase in mean
airway pressure during noninvasive ventilation can better
maintain lung volume than N-CPAP alone. It is also possible that CO2 removal is improved during noninvasive
ventilation by clearance of exhaled gases from the upper
airway.
The use of noninvasive ventilation to achieve a reduction in the need for intubation or the duration of invasive
mechanical ventilation is aimed at reducing the associated
Devices for Noninvasive Ventilation
The devices used for neonatal noninvasive positive pressure ventilation consist of conventional time-cycled pressure-limited ventilators that utilize the same circuits and
gas conditioning devices used for invasive ventilation, with
the main difference being that in lieu of the endotracheal
tube, positive pressure is applied through the same interfaces used for N-CPAP.
Noninvasive positive pressure can be delivered in the
intermittent mandatory ventilation mode (N-IMV) where
the positive pressure cycles are delivered at fixed intervals.
In some ventilators, ventilator cycles can be synchronized
to the neonate’s inspiration to provide nasal synchronized
IMV (N-SIMV). Synchronized noninvasive ventilation
can also be provided in the assist/control (N-A/C) and
pressure support (N-PSV) modes to assist every spontaneous inspiration.
The most commonly reported method for synchronization in noninvasive ventilation is the Graseby pressure
capsule placed on the abdomen (Barrington et al, 2001;
Bhandari et al, 2007; Friedlich et al, 1999; Khalaf et al,
2001). Mainstream flow sensors have been used for synchronization in noninvasive ventilation, but they are likely
to require frequent sensitivity adjustments to avoid autocycling due to variable gas leaks around the interface and the
mouth. This is particularly important during noninvasive
618
PART X
Respiratory System
risks of lung injury. This is obviously more relevant in the
smaller and more immature infants in whom N-CPAP
more frequently fails. In infants with RDS, nasally delivered IMV has not consistently reduced the rate of intubation compared to N-CPAP (Kugelman et al, 2007; Meneses
et al, 2011), but it has been shown to facilitate early extubation after surfactant administration (Bhandari et al, 2007).
In some of these trials, noninvasive ventilation reduced
BPD appreciably, but this was not a consistent finding.
The possible beneficial effects of nasal ventilation during
the initial respiratory failure still need to be confirmed.
During weaning from mechanical ventilation, adequate
maintenance of lung volume is often achieved by N-CPAP.
However, many infants fail because of insufficient ventilation resulting from central apnea, a weak respiratory pump,
or poor lung mechanics due to the underlying lung disease.
Noninvasive ventilation has consistently been shown to be
an effective way to reduce extubation failure (Barrington
et al, 2001; Friedlich et al, 1999; Khalaf et al, 2001; Moretti
et al, 2008), mainly by reducing apnea and improving gas
exchange. Smaller infants and those with poor lung function at extubation were more likely to benefit from nasal
ventilation than larger infants or infants with better lung
function (Khalaf et al, 2001).
Ventilator cycles delivered at fixed intervals can fall
toward the end of spontaneous inspiration or during exhalation and disturb the infant’s breathing pattern, whereas
delivery of the ventilator cycle when the upper airway
is patent may improve transmission of the pressure and
reduce the risk of gas being pushed into the esophagus.
Both nonsynchronized and synchronized modes of nasal
ventilation have been shown to be more effective than
N-CPAP, but data are lacking on the superiority of synchronization in terms of efficacy or safety. Data on the
most effective mode, frequency, and duration of the cycle
and, most importantly, peak pressures during noninvasive
ventilation are also lacking. Until more data are available,
a conservative approach with relatively low ventilator settings and in ranges near those used for intubated infants
recovering from RDS or near extubation is recommended.
This is particularly important in setting peak pressures,
because VT monitoring is not available during noninvasive
ventilation.
Potential Drawbacks
The risks for gastrointestinal complications observed with
N-CPAP may be greater during noninvasive ventilation
because of the additional positive pressure. An earlier
report indicated an increased rate of gastrointestinal complications (Garland et al, 1985), but more recent clinical
trials have not confirmed this.
Because of the additional pressures, the risks for pneumothorax and pulmonary interstitial emphysema may also
be increased. Although there are no data on these side
effects, caution should be exercised and high peak pressures
or ventilator rates should be avoided. The risks for nasal
damage and obstruction often observed during N-CPAP
are also present in noninvasive ventilation. Proper application and maintenance of the nasal interface and avoidance
of excessive force on the nasal septum are important to
avoid these complications.
INVASIVE MECHANICAL VENTILATION
Because of the high incidence of respiratory failure in the
neonate, mechanical ventilation has been one of the main
therapies responsible for the progress in neonatal critical care. This is especially relevant in the small preterm
infant who, besides lung immaturity, has a soft chest wall
and poor central respiratory drive, making the need for
mechanical ventilation very common.
INDICATIONS
The decision to initiate invasive mechanical ventilation
in the newborn is very important because of the serious
complications associated with this mode of therapy and
because in smaller infants, it is often difficult to wean them
from respiratory support. There is considerable variation
between different centers in the criteria used to initiate
mechanical ventilation. Most often, this decision is based
on the gestational age of the infant, the severity of the
respiratory failure, and the disease process that is underlying the respiratory failure. This is also done considering
the alternatives available to support the infant’s respiratory function. The experience of the team and the outcomes of infants exposed to mechanical ventilation in each
institution should also be an important consideration. In
units with vast experience and good outcomes, ventilation
may be used more liberally, whereas in units with limited
experience and high rates of complications, other alternatives should be considered before embarking on invasive
ventilation.
The initiation of mechanical ventilation is usually based
on the clinical condition of the infant and the evaluation
of arterial blood gases. In the preterm infant, mechanical ventilation is frequently started because of recurrent
episodes of apnea and hypoxemia that require some intervention to recover. In more immature infants, ventilation is often begun in the delivery room because of severe
respiratory depression and bradycardia not responsive to
stimulation. The other common indication for mechanical
ventilation is when levels of Paco2 rise rapidly, indicating
alveolar hypoventilation. Although there are no specific
levels of Paco2, most centers initiate mechanical ventilation when Paco2 rises acutely above 55 to 65 mm Hg and
the pH decreases below 7.25 to 7.20.
The introduction of positive pressure ventilation is associated with complications and seldom results in improved
lung function. In fact, intermittent positive pressure ventilation (IPPV) frequently results in further deterioration
in pulmonary function due to the negative effects of high
inspired oxygen concentrations, ventilation-associated
infections, and overdistention of the lung. The effectiveness of mechanical ventilation is primarily due to the
support of the infant’s failing respiratory pump and the
reduction in the work of breathing. One exception to this is
the infant with RDS in whom the positive airway pressure
produces recruitment of distal air spaces with improvement in ventilation-perfusion matching and gas exchange.
In other instances, mechanical ventilation is started
because of hypoxemia not responsive to continuous positive airway pressure. Although there are no set levels of
Pao2 or Fio2 to start ventilation in preterm infants with
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
RDS, the initiation of mechanical ventilation is frequently
associated with the decision to administer exogenous
surfactant. The indications for surfactant vary between
institutions, but there is good evidence that early administration of surfactant results in better outcomes, and therefore, in an infant with RDS, surfactant is usually given
when the inspired oxygen concentration required to maintain acceptable O2 saturation levels increases above 30% to
40%. If the infant has hypercapnia or clinical signs of significant distress and impending failure, ventilation may be
started earlier. In many centers surfactant is given as prophylaxis to all infants below certain gestational age, usually
26 to 28 weeks. After the infant becomes more stable, he
or she is extubated to nasal CPAP.
In full-term infants, the indication for mechanical ventilation is usually more conservative because these infants
are better able to cope with increased work of breathing.
The indication also varies depending on the underlying
cause of the respiratory failure. For example, in an infant
with respiratory failure due to a congenital diaphragmatic
hernia, ventilation is usually started immediately after
birth, whereas in an infant with a congenital pneumonia or
meconium aspiration, a more conservative approach can
be taken, and ventilation may not be started until there
is evidence of rising Paco2 and hypoxemia requiring
inspired oxygen concentrations up to 40% to 60%. The
clinical evaluation of the infant is critical in deciding
whether invasive ventilation should be started.
INSPIRED GAS CONDITIONING
During normal breathing, inspired gas is heated and
humidified in the nasal passages and airways. By the time
it reaches the distal airways, it is fully saturated with water
vapor at core body temperature, that is, 100% relative
humidity at 37° C for an absolute humidity of 44 mg H2O
per liter of gas. The isothermal saturation point region in
the respiratory tract, where inspired gas equilibrates with
core body temperature and is fully saturated, is near the
main bronchi. As inspiratory flow increases, this point
moves distally into the airways.
The nose and airways function as heat and moisture
exchangers. There, a portion of heat and water added to
the inspired gas is recovered during exhalation, with the
net loss depending on the temperature and relative humidity of the gas. This requires continuous replenishment of
water to the aqueous mucosal layer by the airway epithelium and loss of heat. In contrast to ambient air at 22° C
and 50% saturated (absolute humidity of 9. 7 mg H2O per
liter of gas) medical gases are typically colder (15° C or
less) and dry (<2% relative humidity) and therefore require
more heat energy and humidity. Hence, all forms of respiratory support where medical gases are used require conditioning of the inspired gas. This is a key component
of mechanical ventilation because the endotracheal tube
(ETT) bypasses the nose and upper airway.
Inadequate conditioning of the inspired gas can increase
water and heat loss (Fonkalsrud et al, 1980). Exposure to
dry and cold inspired gas can also produce inflammation of
the airway epithelium and increase the risk of airway damage (Marfatia et al, 1975; Todd et al, 1991). Insufficient
humidification can also affect the mucociliary transport
619
system, reducing clearance of secretions, pathogens, and
foreign particles as well as increase the risk of airway blockage by mucus plugs (Fonkalsrud et al, 1975). These effects
are likely to be more striking in small infants, infants with
impaired thermoregulation, and those who are fluid and
energy limited. In small preterm infants, a few minutes
of mechanical ventilation with inadequately conditioned
gases can increase airway resistance and reduce lung compliance (Greenspan et al, 1991). Inadequate gas conditioning has also been associated with increased risk for air leaks
and augmented need for O2 (Tarnow-Mordi et al, 1989).
The standard method for conditioning of the inspired
gases consists of a heater/humidifier (H/H) device and
heated breathing circuits. Dry and cold medical gases are
heated in the H/H chamber to 37° C to increase the water
carrying capacity to 44 mg per liter of gas at 100% relative
humidity. The gas travels through the ventilator circuit,
where it is heated to 39° C to prevent condensation. The
gas temperature decreases as it travels through the ETT,
and gas is delivered at approximately 37° C and near 100%
saturated at the distal end.
Many conditions can affect gas conditioning. Air temperature in an incubator or radiant warmer above the 39° C set
point can inadvertently reduce heating at the ventilator circuit and result in condensation. To avoid this, the temperature probe can be insulated or place outside the incubator
or radiant warmer. If gas heating at the ventilator circuit is
not adequate, low ambient temperatures and low circulating
gas flows can also produce condensation. In general, presence of water condensate in ventilator circuit and minimal
consumption of the humidifier water indicate inadequate
conditioning of the gas. Water condensation can occur in
the ETT and connector when incubator temperature is low
and result in water droplets being pushed into the airway.
Humidity of the inspired gas can also be increased by
water nebulization. In contrast to the size of the water
vapor molecules (0.0001 μm), the size of aerosolized water
particles ranges between 0.5 and 5 μm, which can potentially transport virus or bacteria. Thus, water nebulization
is not an efficient or safe method, particularly for prolonged use.
Gas conditioning is also recommended during nasal
CPAP or nasal ventilation and oxygen head box or nasal
cannula use because the nasal passages and airways may
not achieve adequate conditioning of the cold and dry
medical gases. These gases are typically heated to 32° C,
but heating at or above room temperature may be sufficient. On the other hand, passing the gas through a water
bath may not produce a sufficient gain in humidity because
of the low water carrying capacity of cold gas. Thick and
dry nasal mucus and airway secretions are observed when
gas conditioning is insufficient. Lack of conditioning may
also affect body temperature and water losses.
CONVENTIONAL POSITIVE PRESSURE
VENTILATION
Principles
Neonatal positive pressure ventilation can be described in
its basic form as the cycled application of two distinct levels of positive pressure at the infant’s airway via an ETT.
620
PART X
Respiratory System
5 lpm
5 lpm
Flow
Flow
0
0
20 mL
20 mL
VT
VT
0
Low bias flow
High bias flow
0
25 cm H2O
Ti
Te
PAW
25 cm H2O
PIP
PEEP
PAW
0
FIGURE 45-1 Time-cycled pressure-limited ventilation. In this
example, recordings of flow, tidal volume ( VT), and airway pressure
( PAW) show that ventilator cycles, occurring at intervals determined by
Te, increase PAW from the PEEP level to PIP during the set Ti. The
increase in pressure with respect to the alveolar pressure drives gas into
the lung to achieve VT. The inspiratory flow, which determines the rate
of lung inflation, peaks initially and subsequently declines to zero as the
lung is inflated.
The positive end-expiratory pressure (PEEP) provides a
continuous distending pressure to the lung and is aimed
at maintaining lung volume. This is important because the
ETT bypasses the upper airway mechanism that normally
prevents lung volume loss by active closure of the glottis. The positive pressure applied at the airway opening is
intermittently increased to a predetermined peak inspiratory pressure (PIP) during a set inspiratory time (Ti). The
rise in airway pressure produces a gradient with respect to
the alveolar pressure that drives a tidal volume (VT) of gas
into the lung. This form of ventilation is known as timecycled pressure-limited ventilation.
Neonatal ventilators utilize a constant flow of conditioned gas, also known as bias flow, through the breathing circuit to produce positive pressure. A controlled
obstruction by a valve at the expiratory port of the ventilator produces PEEP, and the intermittent increase to PIP
at intervals determined by the set expiratory time (Te) as
shown in Figure 45-1.
The circulating gas flow determines the profile of the
airway pressure by modulating the rise to the set PIP. The
airway pressure rise is faster and PIP is reached earlier at
higher circulating flow rates. A fraction of this bias flow is
driven into the infant’s airways during tidal inflation. The
infant’s inspiratory flow, which indicates how fast is the
lung being inflated, is in part determined by the profile
of the airway pressure. The rapid rise in airway pressure
produces a rapid increase in the infant’s inspiratory flow
0
FIGURE 45-2 Effect of circulating bias flow. Recordings of flow,
tidal volume ( VT) and airway pressure ( PAW) show how increasing bias
flow rates in the ventilator circuit change the profile of PAW with a more
rapid rise toward the peak pressure. This produces a higher peak flow
that indicates faster lung inflation. An insufficient bias flow rate does not
permit generating the desired peak pressure with each ventilator cycle
and results in a smaller VT.
to a high peak inspiratory flow that decays as the lung is
inflated. In this pattern of rapid lung inflation, most of the
VT is delivered early in the inspiratory phase, as shown
in Figure 45-2. In contrast, a slow rising airway pressure
produces a slower inflation and a smaller peak inspiratory
flow. A low bias flow may not be sufficient to reach the
set PIP within a fixed Ti and in consequence produce a
smaller VT, also shown in Figure 45-2. In most neonatal
ventilators, the bias flow is constant during both the inspiratory and expiratory phases, whereas other ventilators
self-adjust the flow necessary to produce a desired profile.
In other ventilators, the caregiver can set a maximal flow
to be delivered by the ventilator to the circuit during the
cycle, which also modifies the airway pressure profile.
The bias flow should be sufficient to sustain the PEEP
level when the infant’s spontaneous respiratory flow
demands increase. Otherwise, it may create an inspiratory load that the infant has to overcome with each breath.
This is noted as a decline in PEEP during spontaneous
inspiration toward zero pressure, or it can even become
negative. The bias flow is also responsible for removal
of exhaled gases, and an insufficient bias flow may cause
rebreathing. On the other hand, a very high bias flow produces an almost square inspiratory airway pressure waveform that increases the velocity of lung inflation. This
may have undesirable consequences because the lung will
be expanded much faster than during a normal physiologic inflation. In general, circulating bias flow rates for
ventilated preterm infants are set between 5 and 8 liters
per minute, with higher circulating flow rate settings to
accommodate gas leaks around the airway or the demands
of larger neonates.
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
5 lpm
Respiratory System Mechanics during
Positive Pressure Ventilation
The neonate’s respiratory system mechanical properties
(i.e., compliance and airway resistance) determine its time
constant as their product. The respiratory time constant,
which is a measure of the time to achieve equilibrium
between the applied pressure and the alveolar pressure,
varies with different lung diseases and their severity.
The compliance of the respiratory system (CRS) is a
measure of the recoil pressure that opposes expansion volume and is determined by the compliance of lung (CL) and
the chest wall. In the neonate, and most particularly in the
preterm infant, CRS is mainly determined by the compliance of lung because the chest wall is highly distensible.
Lung diseases characterized by lung restriction, such as
respiratory distress syndrome, where the increased lung
recoil is indicated by a low CRS and a short time constant,
are quite prevalent in preterm infants. In addition, they
have a respiratory pump that is often too weak to produce
the pressure required to achieve an adequate VT, and thus
they require mechanical ventilation.
During positive pressure ventilation, a decreased CRS is
characterized by a smaller VT for a given peak pressure
and a relatively brief duration of inflation. As shown in
Figure 45-3, the shorter time constant is illustrated by a
shorter duration of inflation with an earlier return of the
inspiratory flow to zero. This marks the point when the
airway and alveolar pressures are at equilibrium. At this
point, the PIP equals the lung’s recoil pressure, with CRS
as the ratio between VT and the applied pressure. When
CRS decreases, prolonging Ti is ineffective, and a higher
PIP is required to maintain VT constant.
The resistance of the airways opposes the flow of gas,
which dissipates part of the driving pressure during lung
inflation. In diseases characterized by increased airway
resistance, this attenuates the infant’s inspiratory flow and
results in a slower inflation rate. The longer time constant
indicates the increased time required for alveolar pressure
to rise and equilibrate with the applied pressure. When
airway resistance increases, the set Ti may not be long
enough to achieve VT, as shown in Figure 45-4. Although
setting a longer Ti may be reasonable, this should be
done cautiously because the increased airway resistance
also attenuates the expiratory flow and prolongs the time
required to achieve complete exhalation. A long Ti combined with a Te that does not allow complete exhalation
will result in gas trapping.
Modes of Conventional Ventilation
Modes of conventional neonatal ventilation are generally
classified according to the parameter controlled by the ventilator in each cycle as well as by the timing and duration
of the cycle. Ventilator cycles can be pressure or volume
controlled depending on whether the ventilator targets a
set peak pressure or volume, respectively. Ventilator cycles
can be delivered at fixed intervals regardless of the timing with respect to the infant’s spontaneous breathing or
they can be delivered in synchrony with the spontaneous
inspiration. The duration of the inspiratory phase of each
ventilator cycle can be constant or it can adapt to the time
621
End of inflation
Flow
0
20 mL
Reduced CRS
VT
0
CRS 0.66 mL/cm H2O
CRS 0.33 mL/cm H2O
25 cm H2O
PAW
0
FIGURE 45-3 Effect of lung compliance. In this example, recordings of flow, tidal volume ( VT ), and airway pressure ( PAW) show the
effects of a decrease in lung compliance (CL ). A reduction in CL results
in a proportional decrease in VT for the same driving PAW. The effect
is also noted by a shorter time constant indicated by an earlier return of
the inspiratory flow to zero marking the end of lung inflation.
required to complete lung inflation, deliver the set volume,
or to reach the end of spontaneous inspiration.
Intermittent Mandatory Ventilation
The time-cycled pressure-limited (TCPL) ventilation
mode described earlier and illustrated in Figure 45-1 has
been most commonly known as intermittent mandatory
ventilation (IMV). This mode can be classified as pressure
controlled because ventilator cycles deliver the PIP set
by the caregiver. These cycles are of constant inspiratory
duration, that is, Ti, and are delivered at fixed intervals,
that is, Te is determined by the set ventilator frequency.
For many years IMV has been one of the most commonly
used modes of neonatal ventilation during the acute as well
as the more chronic phases of neonatal respiratory disease.
In IMV, the increase in airway pressure due to PIP in
each ventilator cycle produces a given lung inflation during the set Ti. If Ti is too short, complete lung inflation
may not be achieved, whereas a long Ti that maintains a
pressure plateau does not achieve a larger VT and instead
622
PART X
Respiratory System
Increased resistance
5 lpm
End of inflation
5 lpm
End of inflation
Flow
Flow
0
0
End of exhalation
10 mL
20 mL
Volume plateau
VT
VT
0
0
25 cm H2O
Ti
PAW
0
FIGURE 45-4 Effect of airway resistance. In this example, recordings of flow, tidal volume ( VT), and airway pressure ( PAW) show the
effects of an increase in airway resistance. The increase in resistance
results in a decrease in inspiratory flow that does not permit delivery of
VT in the set Ti. The prolonged time constant is noted by the increased
time required to achieve full inflation and to complete exhalation.
produces an inspiratory hold while the lung is kept inflated
as illustrated in Figure 45-5. In infants with respiratory
failure Ti is usually set in the range of 0.25 and 0.4 second. Caution should be exercised when setting ventilator cycles of longer Ti or high ventilator frequencies that
could result in gas trapping due to an insufficient Te to
allow full exhalation as shown in Figure 45-6. This is particularly important when the infant’s time constant is long
because of a high airway resistance or when the ventilator
frequency results in Te <0. 5 seconds.
During IMV, total minute ventilation results from the
ventilation produced by the ventilator and the contribution of the infant’s spontaneous breathing effort. During
clinical use, PIP is usually adjusted to maintain VT between
3 and 5 mL/kg of body weight which is considered adequate for infants with lung disease. Ventilator frequency is
adjusted depending on the infant’s ability to contribute to
minute ventilation and maintain sufficient gas exchange.
PaCO2 levels between 40 and 50 mm Hg are considered
adequate, but higher levels are often accepted in infants
with chronic ventilator dependency.
Although the management of IMV is relatively simple,
there are some important limitations. With IMV, cycles
are often delivered out of synchrony with the infant’s inspiration (Greenough et al, 1983a, 1983b). Asynchronous
ventilator cycles delivered toward the end of the infant’s
25 cm H2O
Long Ti
Short Ti
PAW
0
Pressure plateau
FIGURE 45-5 Inspiratory duration. Recordings of flow, tidal volume ( VT), and airway pressure ( PAW) illustrate the effects of inspiratory
time ( Ti). A Ti that prolongs the inspiratory phase beyond the time
needed to achieve inflation does not increase VT and only keeps the
lung expanded. A Ti of insufficient duration does not permit achieving
full inflation, as indicated by an inspiratory flow that does not return to
zero and leads to a decrease in VT.
inspiration can prolong or increase lung inflation. IMV
cycles delivered during the infant’s exhalation can prolong its duration and in some cases elicit active exhalation
against the positive pressure. Reports suggest associations
between asynchrony and the occurrence of air leaks and
intraventricular bleeding (Greenough and Morley, 1984;
Greenough et al, 1984; Perlman et al, 1983, 1985; Stark
et al, 1979). In older IMV ventilators, VT cannot be measured, and PIP is adjusted based on the expansion of the
chest, which does not permit determination of hypoventilation and gas trapping and assessment of spontaneous
ventilation.
Synchronized Intermittent Mandatory
Ventilation
Synchronized intermittent mandatory ventilation (SIMV)
is for the most part similar to IMV except that ventilator
cycles are delivered in synchrony with the onset of spontaneous inspiration. In SIMV, ventilator management is
similar to that of IMV during the different phases of respiratory failure. SIMV was rapidly accepted and has become
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
5 lpm
Flow
0
Exhalation
ends
Interrupted
exhalation
20 mL
VT
0
Gas
trapping
25 cm H2O
PAW
0
Ti Te
Ti Te
FIGURE 45-6 Gas trapping. Recordings of flow, tidal volume ( VT),
and airway pressure ( PAW) illustrate the potential for gas trapping at
high ventilator rates. As the ventilator rate increases, Te becomes insufficient to complete exhalation where every exhalation is interrupted by
a new ventilator cycle. This produces a volume of gas trapped in the
airways and alveoli and a reduction in VT.
one of the most common modes of neonatal ventilation.
During SIMV, spontaneous breathing is continuously
monitored by the ventilator, and the first spontaneous
inspiration during a time window of constant duration
triggers a ventilator cycle. These time windows are opened
consecutively, and their duration is similar to the interval
between cycles in IMV. If no spontaneous inspiration is
detected by the time the window elapses, a backup ventilator cycle is delivered. In this manner the number of ventilator cycles the infant receives every minute is the same
as in IMV, but the interval between them is not constant.
Different techniques have been incorporated into neonatal ventilators to achieve synchronization, including the
Graseby abdominal pressure capsule, esophageal balloons,
transthoracic impedance, airway pressure changes, and
623
flow sensors. The last are used by most neonatal ventilators for synchronization and VT monitoring.
SIMV produces a greater and more consistent VT
in comparison to IMV because of the increased transpulmonary pressure in ventilator cycles that are in synchrony with spontaneous inspiration (Bernstein et al,
1994; Hummler et al, 1996). Thus, lower PIP and slightly
shorter Ti are often sufficient because the infant’s inspiratory effort contributes to generating the VT. As lung
disease subsides, the infant’s lung mechanics improve,
and spontaneous breathing effort becomes more consistent, PIP is gradually adjusted to maintain a VT between
3 and 5 mL/Kg. During acute respiratory failure, a ventilator SIMV frequency ranging between 40 and 60 b/m
is usually required to maintain Paco2 between 40 and
50 mm Hg. At this frequency, SIMV is likely to assist
almost all spontaneous breaths. During weaning, SIMV
frequency is gradually reduced as spontaneous unassisted
breathing can better maintain acceptable Paco2 levels.
In infants with chronic lung disease, Paco2 levels between
50 and 65 mm Hg are often tolerated for weaning of the
ventilator settings as long as respiratory acidosis is not
observed. The relative advantages of SIMV in comparison to IMV include faster weaning and a shorter ventilator dependency with improved respiratory outcome
in the smaller infants (Chen et al, 1997; Bernstein et al,
1996).
As the SIMV frequency is weaned, the spontaneous
breathing effort must be able to sustain most of the ventilation, thus requiring a greater breathing effort. It has
been suggested that the work of breathing in unassisted
spontaneous breaths between SIMV cycles is counterproductive and can potentially lead to diaphragmatic fatigue.
Conversely, it has also been suggested that exposure to
fewer ventilator cycles may reduce the risk of lung injury
and improve diaphragmatic fitness toward an eventual
extubation. Failure to synchronize leads the ventilator to
cycle at the backup IMV rate. Delayed cycling prolongs
the duration of inspiration and can disrupt the breathing
pattern (Beck et al, 2004), whereas autocycling produce
asynchrony between ventilator and infant.
Assist-Control Ventilation
In A/C, every spontaneous inspiration is assisted by a synchronous ventilator cycle. If the ventilator does not detect
spontaneous breaths because of either apnea or very shallow breathing, it provides an IMV rate with the same PIP
as the assisted breaths. A/C can also be used during the
acute and weaning phases of respiratory failure.
A/C is primarily managed by adjustment of PIP to
maintain VT within an adequate range. As the respiratory failure resolves with improving lung mechanics and
stronger spontaneous breathing effort, PIP is gradually
weaned as VT remains within an acceptable range. In the
more immature infants, the respiratory drive is not consistent, and such infants have frequent apnea. Thus, the
“controlled” or backup IMV rate becomes more relevant
in the maintenance of ventilation during apnea.
In infants with respiratory failure, the Ti required to
achieve lung inflation usually ranges between 0.25 and
0.4 seconds. Longer Ti is not recommended because
624
PART X
Respiratory System
of the potential for gas trapping with ventilator cycles
delivered at the infant’s often high breathing frequency.
A long Ti can also disrupt spontaneous breathing (Beck
et al, 2004; Dimitriou et al, 1998; Upton et al, 1990).
Some ventilators provide flow cycling where Ti terminates at the end of the spontaneous inspiration or at the
end of lung inflation. The advantages of A/C include a
reduction in spontaneous breathing effort relative to IMV
or SIMV because in contrast to these modes, A/C assists
every inspiration (Bernstein et al, 1994; Kapasi et al,
2001). It has been suggested that assisting every spontaneous inspiration in A/C prevents diaphragmatic fatigue
and can reduce the duration of weaning and ventilator
dependency compared to IMV (Baumer, 2000; Beresford
et al, 2000; Chan and Greenough, 1993; Donn et al, 1994).
However, A/C has not been shown to be more effective
than SIMV (Chan and Greenough, 1994; Dimitriou
et al, 1995).
In A/C, PIP should be adjusted to avoid a large VT
that can produce hyperventilation, whereas the backup
rate should be just sufficient to prevent hypoventilation
during apnea. Autocycling is a problem in A/C because
neonatal ventilators do not have a limit on cycling frequency and therefore can produce hyperventilation or gas
trapping.
Pressure Support Ventilation
In PSV, the ventilator also provides a positive pressure
breath with every spontaneous inspiratory effort. The
start of the support pressure is in synchrony with the onset
of the infant’s inspiration and termination of the support
pressure occurs toward the end of spontaneous inspiration
or when lung inflation is completed, that is, flow cycling.
The use of PSV is aimed at unloading the respiratory pump
with support pressure that helps overcome the elastic and
resistive loads imposed by the preterm infant’s underlying lung disease. PSV can be used as a stand-alone mode
with an IMV backup rate for apnea or in some ventilators
as an adjunct to SIMV to assist the spontaneous breaths,
whereas the SIMV cycles with a higher peak pressure are
used to maintain lung volume.
Although PSV can be used during both the acute and
weaning phases of respiratory failure, it is most commonly
used during weaning. A consistent respiratory drive is
required for the use of PSV as a stand-alone mode. For
this reason, a backup IMV rate is recommended particularly in preterm infants to prevent hypoventilation. During
PSV, gas leaks around the ETT can produce autocycling
and extend the duration of the cycle. To avoid the risk of
hyperventilation or gas trapping, trigger and termination
sensitivity levels must be adjusted and limits on cycle duration must be set.
When PSV is used as an adjunct to SIMV, PSV can
reduce the spontaneous breathing effort compared to
SIMV alone even with support pressures set at a fraction of
the peak pressures of the SIMV cycles (Gupta et al, 2009;
Osorio et al, 2005; Patel et al, 2009). More importantly, by
reducing the need for high SIMV rates, PSV can facilitate
weaning in comparison to SIMV alone in preterm infants
of birthweight (BW) less than 1000 g (Reyes et al, 2006).
In that study PSV set at 30% to 50% of the peak pressure
of the SIMV cycles were sufficient to maintain acceptable
Paco2 levels with significantly lower SIMV rates.
Volume-Targeted Ventilation
In volume-targeted ventilation, automatic adjustments
to the peak positive pressure or the duration of ventilator cycle are done to maintain a target VT. Volume targeted ventilation modes have been proposed as means to
reduce ventilator-associated lung injury caused by ventilation with excessive or insufficient tidal volumes during
conventional pressure-controlled ventilation. A number of
volume-targeted modes are available in neonatal ventilators. These differ in the timing of the adjustment or in the
duration of the mechanical cycle, that is, whether it occurs
as the cycle is delivered to the infant or in the subsequent
cycle. These modes also differ in the volume parameter
that is controlled—whether it is the tidal volume received
by the infant or the volume delivered by the ventilator to
the circuit, and whether this is measured during the inspiratory phase or during exhalation.
Volume-Controlled Ventilation
In volume-controlled ventilation (VC), the ventilator
delivers a set volume of gas into the ventilator circuit in
each cycle. VC cycles are delivered in the IMV, SIMV, or
A/C modes described earlier. The time required to deliver
the set volume depends on the ventilator flow rate, which
can be constant during the cycle or variable with an initial
peak followed by a gradual decline. The flow continues
until the set volume is delivered. The cycle ends before the
set volume is delivered if its duration exceeds the set Ti or
the airway pressure exceeds the set PIP. In small infants,
most of the volume delivered by the ventilator is compressed in the circuit, and the actual VT delivered to the
infant is only a fraction. Some ventilators use algorithms
to correct the measured volume by the compressed volume
to estimate true VT.
VC has been proposed as a strategy to facilitate weaning
and reduce the complications of positive pressure ventilation. Clinical trials showed that compared to pressure limited ventilation in the A/C mode, VC reduced the weaning
time and the duration of mechanical ventilation in infants
weighing at least 1200 g at birth (Sinha et al, 1997). Weaning was also faster with VC in infants of birthweight <1000
g, but the total duration of mechanical ventilation did not
change and respiratory outcome did not differ significantly
(Singh et al, 2006).
Pressure-Regulated Volume-Controlled
Ventilation
In pressure-regulated volume-controlled ventilation
(PRVC), the peak pressure of pressure-controlled ventilator cycles is adjusted from one cycle to the next to maintain
a target volume. The targeted volume can be that delivered by the ventilator or the actual estimated or measured
VT. During PRVC, gas leaks around the ETT can produce
overestimation of volume during the inspiratory phase and
consequently lead to inappropriate reductions in peak
inspiratory pressure.
Compared to conventional IMV, PRVC in A/C mode was
effective in reducing the duration of mechanical ventilation
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
and the incidence of intraventricular hemorrhage (IVH) in
infants with RDS of BW <1000 g (Piotrowski et al, 1997).
These advantages can be attributed, in addition to volume
targeting, to synchronized delivery of PRVC cycles. These
advantages were not evident when PRVC was compared to
SIMV (D’Angio et al, 2005).
Volume Guarantee Ventilation
In volume guarantee ventilation (VG), the peak pressure
of each ventilator cycle is adjusted to maintain a target VT
based on exhaled VT measured in previous cycles. Measurement of exhaled VT is aimed at circumventing the
effects of gas leaks around the ETT during the inspiratory
phase. VG can be used in combination with A/C, PSV,
SIMV, or IMV modes.
VG was proposed as a potential alternative to avoid
both extremes of VT and to achieve a consistent weaning
of peak pressure. The stability of VT and gas exchange in
infants with RDS can be improved by VG when combined
with A/C or PSV as noted by fewer breaths with too small
or large VT and less hypocapnia (Abubakar and Keszler,
2001; Cheema et al, 2007; Herrera et al, 2002; Keszler
and Abubakar, 2004). In infants with RDS or who had
received surfactant, VG achieved the proper reduction in
PIP, but this was dependent on setting a lower target VT
than the VT attained with the pressure-controlled modes
(Abd El-Moneim et al, 2005; Abubakar and Keszler, 2001;
Cheema and Ahluwalia, 2001; Herrera et al, 2002; Nafday
et al, 2005; Olsen et al, 2002). However, it must be noted
that the smaller target VT was in some cases not sufficient,
resulting in increased Pco2 and spontaneous breathing
effort (Herrera et al, 2002).
VG was proposed as a means to attenuate hypoxemia
spells triggered by hypoventilation during periods of agitation and decreased compliance in preterm infants. VG
reduced the duration of the hypoxemia spells compared to
SIMV, but only when the target VT was increased which
resulted in a considerable increase in airway pressure
(Polimeni et al, 2006).
New and Experimental Modes
Proportional Assist Ventilation
In proportional assist ventilation (PAV), the ventilator
pressure is increased in proportion to the measured volume, flow, or both generated by the infant’s inspiratory
effort. This achieves a perceived reduction of the elastic
and resistive loads imposed by lung disease that commonly
prevent the infant from producing an adequate VT. The
proportionality factors by which the positive pressure
increases in relation to volume or flow are the elastic (volume proportional) and resistive (flow proportional) gains.
The elastic and resistive gain factors must be individualized to each infant’s lung compliance and airway resistance. Elastic gain factors that exceed the lung elastance
(inverse of compliance) can lead to a runaway increase in
pressure, whereas excessive resistive gain factors can produce oscillations in airway pressure. For this reason, PAV
devices provide peak pressure and volume limits. Because
PAV amplifies only the spontaneous breathing effort, a
backup IMV rate is needed to prevent hypoventilation
during apnea.
625
By compensating for the loads induced by lung disease,
PAV reduced the breathing effort in infants recovering from RDS (Musante et al, 2001). Compared to conventional modes that deliver a constant pressure during
inspiration, PAV produced similar ventilation with lower
ventilator and transpulmonary pressures (Schulze et al,
1999, 2007).
Neurally Adjusted Ventilatory Assist
Neurally adjusted ventilatory assist (NAVA) is a mode
where the ventilator pressure is adjusted in proportion to
the electrical activity of the crural diaphragm measured by
esophageal electrodes. NAVA was developed to improve
the coupling of the infant’s inspiration and the ventilator by
overcoming conditions that delay or limit pneumatic triggering of the ventilator cycle. A study in preterm infants
approaching extubation showed NAVA can produce comparable gas exchange with lower pressures than PSV, but
no physiologic effects of the better synchrony, including
breathing effort, were noted. This study showed the feasibility of noninvasive NAVA after extubation (Beck et al, 2009).
Data are lacking on the effects of different NAVA
proportionality factors between diaphragmatic electrical activity and ventilator pressure within the same infant
or between infants, because electrical activity of the diaphragm cannot be normalized. A backup ventilator rate is
needed to prevent hypoventilation during apnea.
Targeted Minute Ventilation
Targeted minute ventilation (TMV) is an experimental
mode where the ventilator rate is adjusted to maintain
minute ventilation at a target level. If minute ventilation
exceeds or decreases below the target level, the ventilator
rate is reduced or increased, respectively. If spontaneous
breathing can maintain a normal minute ventilation, the
ventilator rate is reduced. In preterm infants recovering
from RDS, TMV reduced the ventilator rate to half without impairing gas exchange compared to SIMV. Although
these infants were able to sustain their ventilation for long
periods, at times they required increased ventilator rates
(Claure et al, 1997).
Mandatory minute ventilation (MMV) is a form of
targeted minute ventilation where the ventilator rate is
turned off when spontaneous breathing maintains ventilation. If minute ventilation falls below the target level, a
constant ventilator rate of VC cycles is provided. In nearterm infants ventilated for reasons other than lung disease,
MMV weaned the rate and reduced the mean airway pressure compared to SIMV (Guthrie et al, 2005).
Adaptive backup ventilation is a form of backup support.
In this mode, a ventilator rate is provided during apnea
as well as during the occurrence of hypoxemia detected
by Spo2. In preterm infants recovering from RDS, this
hybrid backup mode reduced the incidence and duration
of hypoxemia spells compared to a backup ventilator rate
for apnea alone (Herber-Jonat et al, 2006).
HIGH-FREQUENCY VENTILATION
Because of the association between pulmonary overdistention, lung injury, and the development of BPD, the
possibility of achieving gas exchange with very small tidal
626
PART X
Respiratory System
volumes has been of great interest to neonatologists for
many years. This led to the development of instruments
that can generate changes in airway pressure at rates in
excess of 10 Hertz (600/min) with the aim of producing
ventilation with very small tidal volumes, usually much
smaller than the dead space.
Gas Transport during High-Frequency
Ventilation
During conventional ventilation, gas exchange occurs
by introducing of fresh gas into the distal airspaces with
each inspiration. In contrast, during high-frequency ventilation (HFV), the volume of fresh gas delivered by each
cycle is very small and does not reach the most distal portions of the lung. Therefore, different mechanisms must
explain alveolar ventilation and gas exchange. These
include bulk flow into the more proximal portions of
the lung, enhanced mixing of gas within the conducting
airways, and out-of-phase movement between different
regions of the lung that have different time constants.
There is also enhanced diffusion of gas in large and
medium-sized airways due to asymmetrical velocity profiles during inspiration and expiration. Finally, there is
molecular diffusion in the more distal air spaces that
moves the different gas molecules from areas of higher
to lower concentrations. These mechanisms have been
mostly explored in adult lung models, and therefore it
is not clear how well they apply to the immature or sick
neonatal lung.
Devices for HFV
Several types of high-frequency (HF) ventilators have
been used in the neonate, including jet ventilators, oscillators, and flow interrupters.
Jet ventilators generate a high-velocity gas flow that is
injected through a small-diameter tube that opens into the
airway connector. Expiration is passive, and the cycling
rate is determined by an electrically operated valve that
opens and closes the jet at a predetermined rate and timing. The high velocity of the gas injected into the airway
produces a Venturi effect that pulls additional gas from
the ventilator circuit. This is known as “gas entrainment.”
Tidal volume is determined primarily by the driving pressure of the gas, the inspiratory time, and the resistance of
the injection port. Because expiration is passive during
high-frequency jet ventilation (HFJV), there is a risk of gas
trapping and lung overdistention (Bancalari et al, 1987).
This risk is higher when the time constant of the respiratory system is increased by airway obstruction and when
large tidal volumes are delivered. Because of this, jet ventilators are used at lower rates (4 to 10 Hz). Jet ventilators
are used in combination with conventional ventilators that
provide PEEP and may also provide conventional positive
pressure cycles.
Oscillatory ventilators use a piston or a membrane
driven by an electromagnetic force and connected to the
ventilator circuit. The mean airway pressure is determined
by the gas flow through the circuit and a variable resistance, whereas the tidal volume is generated by the size
of the excursion of the piston or membrane. With these
devices, the expiratory phase is active because during expiration, airway pressure falls below baseline. This reduces
the risk of gas trapping. Flow interrupters are a hybrid
between jets and oscillators. They produce airway pressure
changes by interrupting the gas source at very high rates
using a standard ventilator circuit rather than an injection
cannula. They are relatively simple and are usually offered
as an additional mode in conventional ventilators. Most
do not have enough power to generate sufficient tidal volumes to effectively ventilate large infants or infants with
very stiff lungs.
Clinical Experience
The indications for HFV vary widely among different centers. Although some use HFV routinely as a primary mode
of support, most centers use HFV as rescue when conventional ventilation has failed. This includes preterm infants
who require increasing ventilator settings to maintain Co2
elimination and oxygenation within acceptable limits and
those with evidence of pulmonary interstitial emphysema.
In larger infants, HFV is also indicated in situations where
conventional ventilation is not sufficient to maintain
acceptable Co2 elimination or oxygenation, and especially
in infants with persistent pulmonary hypertension secondary to hypoplastic lungs due to congenital diaphragmatic
hernia or oligohydramnios or to severe lung disease due to
meconium aspiration or pneumonia.
HFV in RDS
The clinical results with the use of HFV in infants with
RDS have been inconsistent. Whereas some studies have
shown better outcomes such as increased survival with no
BPD (Courtney et al, 2002), others have not shown differences (Johnson et al, 2002). Some studies have suggested
a higher risk of air leaks (Thome et al, 1999), and others
higher incidence of intracranial hemorrhage with HFV
(HIFI Study Group, 1989; Moriette et al, 2001).
Many of the earlier studies were performed before exogenous surfactant was available, and therefore the results are
not applicable to the present situation where most preterm
infants are exposed to antenatal steroids and, when indicated, receive exogenous surfactant. A meta-analysis of the
most recent trials suggests a possible advantage for HFV in
the outcomes “BPD at 36 weeks” or “death or BPD” at 36
weeks corrected age. However, when the results were analyzed using adjustments for heterogeneity between trials,
the beneficial effect disappeared (Thome et al, 2005). The
results of this meta-analysis revealed a significant increase
in the risk of air leaks and a trend for higher risk of IVH
grades III and IV with HFV. When the results of HFV
were compared with conventional ventilation, including only studies where HFV was used with high-volume
strategies and conventional ventilation with an optimized
low positive pressure and tidal volume strategy, there were
no significant differences in any of the outcomes (Bollen
et al, 2007; Thome et al, 2005). Because of the inconsistency of the results and the lack of solid evidence of benefits of HFV over conventional ventilation in infants with
RDS, the selection of one modality over the other is mostly
based on individual preference.
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
HFV in PPHN
The clinical evidence with the use of HFV in infants with
PPHN is not solid and comes from a few, relatively small
trials. Some of these studies suggested a decreased need for
extracorporeal membrane oxygenation (ECMO) in infants
with PPHN treated with HFV compared to those managed with IPPV (Clark et al, 1994). However, other studies
have not shown clear advantage of HFV over conventional
management (Kinsella et al, 1997; Rojas et al, 2005). HFV
may offer some advantage over conventional ventilation in
infants with hypoplastic lungs secondary to congenital diaphragmatic hernia (Desfrere et al, 2000).
Other Indications
HFV has also been used in infants with bronchopleural
fistulas in an attempt to reduce the amount of gas leak into
the pleural space (Gonzalez et al, 1987; Walsh and Carlo,
1989). It can also be used during bronchoscopy or during
airway surgery because in contrast to conventional ventilation, it can produce adequate gas exchange with a partially
open airway and large gas leaks (Nutman et al, 1989).
Side Effects of HFV
Because during HFV the tidal volumes are extremely
small, when HFV is used in unstable lungs there is a possibility of progressive loss of lung volume and atelectasis.
For this reason it is necessary to use mean airway pressures
that are usually higher than those used during conventional IPPV. In fact there is evidence that during HFV the
use of an open lung strategy utilizing recruiting maneuvers
and high mean airway pressures produces better outcomes
than utilizing lower pressures (Thome et al, 2005). As a
result of these higher pressures, it is likely that HFV may
negatively influence cardiovascular function (Osborn and
Evans, 2003; Trindade et al, 1985; Truog and Standaert,
1985; Weiner et al, 1987) and may also explain the higher
incidence of pneumothorax reported in some studies with
HFV (Thome et al, 2005).
Because of the effectiveness of HFV in enhancing alveo
lar ventilation it is easy to drive Paco2 to very low values within a very short period of time. For this reason it
is important to monitor Paco2 values closely, especially
when HFV is initiated or when settings are changed.
Because of the very short inspiratory and expiratory times,
it is extremely important to maintain the airway as patent as possible, ensuring a correct position of the endotracheal tube. Any obstruction will produce a decrease in
pressure transmission and in tidal volume and can lead to
gas trapping. This is even more important with jet ventilation. During jet ventilation, it is also critical to ensure
proper humidification of the inspired gas to prevent airway
damage that can be produced by the high velocity of gas
injected into the airway.
Ventilator Settings during HFV
The ventilator settings during HFV are simpler to adjust
than with conventional ventilation. The mean airway pressure determines the lung volume and is adjusted to achieve
627
better ventilation-perfusion ratio and oxygenation. This
is done considering the severity and type of lung disease.
Most infants with RDS are managed with mean airway
pressures between 8 and 15 cm H2O. Higher levels may
be necessary in some cases with severe lung disease and
poorly compliant lungs.
The tidal volume is determined by the delta pressure,
and this, in combination with the frequency, determines
the Co2 elimination. In contrast to conventional ventilation, during HFV the tidal volume is reduced as the
frequency increases because the shorter times for inspiration and expiration prevent the equilibration of pressures
between the ventilator circuit and the distal portions of
the lung. Therefore, during HFV a reduction in rate may
result in larger tidal volumes, more Co2 elimination, and
lower Paco2 levels.
During HF oscillation or with flow interrupters the
frequencies used range from 8 to 15 Hz, whereas during
jet ventilation lower frequencies between 4 and 10 Hz are
used.
The pressure transmission to the distal airways is greatly
influenced by the resistance of the tube and the airway, so
only a small fraction of the delta pressure generated by the
ventilator is transmitted to the terminal airspaces. For this
reason, the delta pressure during HFV represents more of
a relative value used to adjust the ventilator than the real
pressure change in the distal airways.
WEANING FROM MECHANICAL VENTILATION
Weaning from IPPV is extremely difficult in very immature infants who have poor inconsistent respiratory drive,
a weak respiratory pump, and immature and many times
damaged lungs. This combination of factors explains why
they frequently become ventilator dependent for long
periods of time.
For many years, infants who were ventilated received
most of their minute ventilation from the ventilator and
were not allowed to have effective spontaneous ventilation.
In recent years this has changed, and ventilators are used as
an assist support, preserving the patient’s respiratory drive
and effort. This has been possible by the introduction of
synchronized patient-triggered ventilation (PTV) and has
resulted in better outcomes and shorter times on mechanical ventilation.
Because of the high rate of complications associated with
prolonged mechanical ventilation, weaning should start
as soon as respiratory function is stabilized. The order in
which the different ventilator parameters are decreased is
determined by the relative risk of complications associated
with each of them. With the possibility of measuring VT,
it has become much simpler to define the appropriate PIP
that is required to generate an adequate VT of 3 to 5 mL/kg
body weight. As lung compliance improves PIP can be
reduced to keep VT within this range.
The level of PEEP is usually kept between 4 and 8 cm
H2O depending on the type of underlying lung disease and
the level of oxygenation and Fio2 requirement. The PEEP
level is decreased gradually as the oxygenation improves
until a level of 4 to 5 cm H2O is reached, and this level is
maintained until extubation. The inspired oxygen concentration is adjusted according to the level of arterial oxygen
628
PART X
Respiratory System
tension or O2 saturation measured by pulse oximetry.
The ideal ranges of oxygenation have not been defined,
but most clinicians accept saturations between 88% and
95% in preterm infants and up to 98% in term infants. In
infants with evidence of pulmonary hypertension, higher
levels are targeted to prevent pulmonary vasoconstriction.
If VT measurement is not available, the reduction in PIP
is based on the observation of chest movement, the degree
of aeration on chest radiograph, and Paco2 levels. The
ventilator rate is adjusted depending on the type of ventilation strategy being used. When the infant is ventilated with
synchronized modes such as A/C or pressure support (PS),
the infant determines the rate of the mechanical breaths
so that the set rate is only the backup that the ventilator
will provide when the infant’s own rate falls below that
level. Therefore, this rate is only relevant when the infant
becomes apneic or hypoventilates. When the infant is controlled, the rate in the ventilator is not determined by the
infant, and the adjustment in mechanical rate is based on
the arterial Pco2 level.
During weaning, it is advisable to do gradual changes
and adjust one parameter at a time to evaluate the response
of the infant to each change. With the availability of continuous oxygen and Co2 monitoring, it is not always necessary to wait for results of arterial gas measurement to
change ventilator settings, and the weaning can proceed
faster.
Synchronized Patient-Triggered
Ventilation
The use of patient-triggered synchronized ventilation has
become common practice in neonatal units. Most randomized trials comparing PTV with nonsynchronized ventilation have shown reduction in the duration of mechanical
ventilation in infants treated with synchronized modes
(Greenough, 2001). This may be because, during PTV,
the infant retains more control of ventilation, and the
effectiveness of the mechanical and spontaneous breaths is
enhanced by the summation of the ventilator positive pressure and the negative pressure generated by the respiratory
muscles.
A/C and SIMV are the most common modes of synchronized ventilation in the neonate. It has been suggested
that assisting each spontaneous inspiration in A/C may
avoid respiratory muscle fatigue and facilitate weaning.
However, the duration of weaning has not been consistently shorter with A/C than with SIMV (Beresford et al,
2000; Chan and Greenough, 1994). On the other hand, a
randomized trial comparing the use of pressure support
ventilation as an adjunct to SIMV to assist every spontaneous inspiration revealed a faster weaning and shorter duration of ventilation compared with SIMV alone in preterm
infants (Reyes et al, 2006).
Volume Monitoring and Targeting
The continuous monitoring of VT allows rapid weaning
of PIP as the mechanical conditions of the lung improve.
This can be achieved by a manual decrease of PIP as VT
increases, or automatically by using volume-targeted
ventilation where weaning is achieved automatically
independent of the clinician, who only sets the VT targeted
by the ventilator. Evidence from randomized trials using
volume-targeting strategies suggest that faster weaning
from mechanical ventilation can be achieved, although the
results have not been entirely consistent (Singh et al, 2006;
Sinha et al, 1997).
Nasal CPAP and Nasal Ventilation
After extubation, the infant is exposed to a number of
mechanical impediments that explain the frequent need for
reintubation in the smaller preterm infants. These include
upper airway damage and retained secretions leading to
obstruction and atelectasis, loss in lung volume due to
poor respiratory effort, and a highly compliant chest wall.
For these reasons, the use of continuous positive airway
pressure applied through the nose can significantly reduce
the deterioration that occurs frequently after extubation.
Surprisingly, despite this improvement in respiratory
function, the need for reintubation has not been consistently shown to be reduced by the use of N-CPAP after
extubation (Davis and Henderson-Smart, 2003).
In contrast with N-CPAP, the use of nasal ventilation
after extubation has been shown to significantly reduce
extubation failure (Davis et al, 2001; De Paoli et al, 2003).
Although these studies have included small numbers of
infants, the effects have been consistent. This is a promising therapeutic alternative that needs further evaluation
and the development of suitable equipment to provide
synchronized noninvasive support.
Respiratory Stimulants
Respiratory stimulants such as aminophylline and caffeine have been shown to be effective to increase respiratory center activity in preterm infants and to decrease the
incidence of severe apneic episodes. These drugs have also
been shown to facilitate successful weaning from mechanical ventilation and decrease the need for reintubation. For
this reason most preterm infants receive a loading dose
of caffeine or aminophylline before extubation, and they
are maintained on these stimulants at least during the
first days after extubation while they are also maintained
on N-CPAP or nasal ventilation (Henderson-Smart and
Davis, 2003).
Permissive Hypercapnia
Tolerance of higher carbon dioxide levels may reduce the
need for support and reduce the duration of ventilation.
However, the results of clinical trials have been inconsistent. Whereas initial trials suggested faster weaning from
mechanical ventilation in the group with higher Co2 levels (Carlo et al, 2002; Mariani et al, 1999), a more recent
study showed no benefit in duration of ventilation and a
possible increase in mortality and central nervous system
(CNS) impairment in infants with higher Paco2 (Thome
et al, 2006). Although these results shed doubt on the benefits of high Co2 levels in premature infants during the
acute stages of their clinical course, in infants with chronic
lung disease it is necessary to tolerate high Co2 levels to
wean them from mechanical ventilation.
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
Dead Space Reduction
The large anatomical dead space in preterm infants in addition to the instrumental dead space can also delay weaning
from mechanical ventilation (Figueras et al, 1997).
Continuous tracheal gas insufflations (CTGI) pumped
through small capillaries to the distal end of the endotracheal
tube to produce a continuous washout can reduce arterial
CO2 and shorten the weaning process (Dassieu et al, 2000).
This method is limited by the need for special ventilators and
endotracheal tube. Continuous washout of the flow sensor
by a controlled gas leak can also improve CO2 elimination
and facilitate weaning (Claure et al, 2003; Estay et al, 2010).
Extubation from IMV versus Endotracheal
CPAP
For many years infants on mechanical ventilation were
extubated only after they had tolerated some time on
CPAP via endotracheal tube. This practice was changed
after a small, simple trial demonstrated that infants extubated from a low IMV rate had better success rates than
those kept on CPAP for 6 hours before extubation (Kim
and Boutwell, 1987).
Weaning from High-Frequency Ventilation
Infants ventilated with HFV are frequently switched to
conventional ventilation before extubation. However,
infants can be extubated directly from HFV following
similar steps to those used to wean from conventional ventilation. The reduction in mean airway pressure (MAP) is
done after oxygenation and lung expansion is estimated by
chest radiographs while the Fio2 is adjusted to maintain
the desired oxygenation levels.
The oscillatory amplitude is gradually reduced in
response to the levels of Paco2. If the MAP is weaned
to low levels before the infant has spontaneous respiratory effort, this can lead to hypoventilation, loss in lung
volume, and atelectasis. For this reason, it is advisable to
lower the delta pressure and allow the CO2 to rise and
stimulate spontaneous respiration before the MAP is lowered below 10 or 8 cm H2O.
Prediction of Successful Extubation
It is difficult to decide the best time for extubation in ventilated infants. It is obvious that many infants remain intubated for longer than necessary. This is evidenced by the fact
that many infants who are accidentally extubated tolerate it
well without needing further respiratory support. Various
tools have been evaluated to predict successful extubation.
These include measurement of lung mechanics, minute ventilation, inspiratory effort strength and ability to cope with
mechanical loads, and stability of respiratory pattern during
periods when the ventilator cycling is stopped (Kamlin et al,
2006). Some of the tools have been shown to predict with
some accuracy successful extubation, but these tests have not
been widely accepted in neonatal clinical practice.
The decision to extubate an infant is usually based on
the level of inspired oxygen and ventilator support that the
infant is requiring to maintain acceptable arterial blood
629
gas levels. In general terms, if an infant needs less than
30% to 40% oxygen, a ventilator rate less than 15 per
minute, and peak airway pressures below 15 cm H2O and
keeps acceptable blood gases, most clinicians attempt extubation. The lower the gestational age, the more likely it is
that the infant will not tolerate extubation and will require
reintubation. In most cases this failure is because of poor
respiratory effort or severe apneic episodes.
Automated and Computer-Assisted
Weaning
In the targeted minute ventilation mode described earlier,
the ventilator rate is automatically reduced during periods of
consistent spontaneous breathing where minute ventilation
is maintained at or above the target level. In a study of preterm infants recovering from RDS, this experimental mode
reduced the ventilator SIMV rate set by the clinical team by
half while arterial blood gases remained unchanged (Claure
et al, 1997). A similar reduction in rate was observed in nearterm infants without lung disease when supported by mandatory minute ventilation, a mode where the ventilator rate
is turned off if minute ventilation exceeds a set level or delivers a set rate of volume-controlled breaths when minute
ventilation decreases below this level (Guthrie et al, 2005).
Preterm infants often need supplemental O2, which
increases their risk for eye and lung injury, particularly
when exposure to oxygen is prolonged. In these infants,
hyperoxemia is induced by an excessive Fio2, and therefore it is modifiable by appropriate weaning. Automated
weaning of supplemental oxygen was achieved by systems
developed to adjust Fio2 in a continuous manner. Systems
of automated Fio2 control have been shown to be as or
more effective than routine or dedicated manual control
in maintaining oxygenation within a desired range, with
most of the improvement due to significant reductions in
hyperoxemia (Claure et al, 2009, 2011).
Ventilator management involves adjustments of several parameters that affect the infant’s ventilation and
gas exchange. Computerized algorithms for ventilator
management have been proposed to achieve efficient
and consistent weaning. In infants with RDS, ventilator
management assisted by one of these algorithms led to
improvements in gas exchange and avoided unnecessary
increases in ventilator settings (Carlo et al, 1986). Data
indicated that during routine care, hypoxemia and hypercapnia were more diligently corrected than hyperoxia and
hypocapnia, whereas computer-assisted management was
similarly effective in correcting both extremes. The potential benefits of computer-assisted weaning on long-term
outcome have not been explored.
ACUTE COMPLICATIONS
OF RESPIRATORY SUPPORT
PULMONARY GAS LEAKS
Some of the most serious complications of mechanical ventilation are pulmonary gas leaks. These include pulmonary
interstitial emphysema (PIE), pneumothorax, pneumomediastinum, pneumopericardium, pneumoperitoneum, and
intravascular gas.
630
PART X
Respiratory System
involved areas in the lung. If PIE is unilateral, this can be
accomplished by positioning the infant with the involved
side down (Swingle et al, 1984). Mechanical ventilation
using short inspiratory times (0.1 to 0.2 seconds), low
inflation pressures, and small tidal volumes can reduce the
gas leak (Meadow and Cheromcha, 1985). Unfortunately,
it is difficult to maintain oxygenation and ventilation while
using low volumes. A multicenter controlled trial found
that HFJV allowed the use of lower peak and mean airway
pressures in infants with PIE than did rapid-rate conventional ventilation and led to more rapid improvement in
PIE (Keszler et al, 1991).
Pneumothorax
FIGURE 45-7 Pulmonary interstitial emphysema. A grossly hyperinflated lung with coarse radiolucencies extending from the pleura to the
hilum. These radiolucencies represent gas bubbles in the perivascular
and peribronchial interstitial cuffs.
Pulmonary Interstitial Emphysema
PIE is the result of rupture of air spaces from overdistention. Although it may be seen in infants breathing spontaneously it is much more common in preterm infants
undergoing mechanical ventilation and in infants with
evidence of pulmonary infection. Once alveolar rupture
occurs, gas is forced from the air spaces into the connective
tissue sheaths surrounding airways and vessels and into
the interlobular septa containing pulmonary veins. The
air follows a track along these sheaths to the hilum of the
lung, producing the characteristic radiographic appearance of PIE (Figure 45-7). The PIE occupies space within
the lung parenchyma, decreasing lung compliance. Gas
trapped within the interstitial cuffs compresses airways and
increases airway resistance. In addition, gas in the interstitial space impairs lymphatic drainage, increasing interstitial fluid. This leads to significant deterioration in gas
exchange with increase in Paco2 and decrease in Pao2.
The main cause of PIE is airspace overdistention and
rupture. Therefore, minimizing pulmonary overdistention
should reduce the risk of PIE. Shorter inspiratory times
also decrease the incidence of PIE by reducing overdistention and avoiding an inspiratory hold (Heicher et al, 1981;
OCTAVE Study Group, 1991).
PIE is a serious complication of mechanical ventilation. Infants with PIE have a significantly increased risk of
developing chronic lung disease as well as higher mortality
rates (Gaylord et al, 1985; Powers and Clemens, 1993).
Management
Because the gas leak can behave like a check valve, gas trapping occurs, resulting in further alveolar overdistention
and rupture. Therefore, the first step in treatment must be
to interrupt this cycle by putting to rest the more severely
Pneumothorax can occur spontaneously in healthy infants
because of the very high transpulmonary pressures produced at birth. However, the incidence is much higher
in the presence of underlying lung disease and in infants
exposed to mechanical ventilation. Pneumothorax develops in 5% to 10% of spontaneously breathing infants with
hyaline membrane disease. Although positive pressure
ventilation increases the risk dramatically, treatment with
surfactant has markedly lowered this risk.
Pneumomediastinum occurs when gas tracks through
the perivascular and peribronchial cuffs to the hilum and
then ruptures into the mediastinum. From there, gas can
rupture into the pleural space, producing a pneumothorax. If the leak is large, it produces a tension pneumothorax that collapses the lung and results in severe hypoxia
and hypercapnia. In addition, by compressing mediastinal
structures, venous return to the heart may be impeded,
resulting in circulatory collapse.
Diagnosis
Because of the severe consequences of a tension pneumothorax, it is important to diagnose it as early as possible. In
the spontaneous breathing infant, pneumothorax usually
produces tachypnea, grunting, pallor, and cyanosis. The
cardiac sounds may be shifted away from the side of the
pneumothorax, and the affected hemithorax and abdomen may appear to be bulging. In the infant on positive
pressure ventilation, signs are frequently more dramatic,
with sudden onset of hypoxemia and cardiovascular collapse (Ogata et al, 1976). Transillumination of the chest
is positive over the affected side, and the pneumothorax is
confirmed by chest radiograph that shows gas in the pleural space and partial collapse of the lung with shift of the
mediastinum to the opposite side (Figure 45-8).
Management
A small pneumothorax in a spontaneously breathing infant
may be observed closely until spontaneous resolution
occurs. Infants with larger symptomatic pneumothoraces and all infants receiving positive pressure ventilation
require thoracostomy and pleural tube placement. The
tube may be inserted at the midaxillary line and directed
anteriorly, or placed in the second intercostal space in the
midclavicular line and directed toward the diaphragm so
that the tip lies between the lung and the anterior chest
CHAPTER 45 Principles of Respiratory Monitoring and Therapy
FIGURE 45-8 Tension pneumothorax. The lung on the involved
side is collapsed, and the mediastinum is shifted to the opposite side,
with bulging of the pleura into the intercostals spaces.
wall. When placing the tube, the operator must avoid
puncturing the lung, especially when a trocar, rather than
a curved hemostat, is used to direct the tube. It also is
possible to drain pneumothoraces using pigtail catheters
that are placed percutaneously into the pleural space. This
technique produces less trauma but may not be effective
when large gas leaks are present. The thoracostomy tube
is connected to a water seal with 10 to 20 cm H2O negative pressure and is left in place until it ceases to drain.
The negative pressure should be discontinued, and the
tube should be left under the water seal for 12 to 24 hours
before removal.
Pneumopericardium
Pneumopericardium results from direct tracking of interstitial gas along the great vessels into the pericardial sac.
Gas under tension in the pericardium impairs atrial and
ventricular filling, decreases stroke volume, and ultimately
decreases cardiac output and systemic blood pressure.
Infants present with increasing cyanosis, decreased heart
sounds, and decreased systemic blood pressure and pulse
pressure. The chest radiograph is diagnostic, showing gas
surrounding the cardiac silhouette (Figure 45-9). Needle
aspiration alleviates the acute symptoms, but because
recurrence rate is high, continuous tube drainage is necessary in most cases.
Pneumoperitoneum
Pneumoperitoneum results from dissection of air from the
mediastinum along the sheaths of the aorta and vena cava
into the peritoneal cavity. Infants with this condition present sudden abdominal distention and a typical abdominal
radiograph. Occasionally, the pneumoperitoneum may
be large enough to cause respiratory embarrassment by
compromising descent of the diaphragm and may require
631
FIGURE 45-9 Pneumopericardium. A thin rim of pericardium is visible and clearly separated from the heart by gas within the pericardial sac.
drainage. This cause of peritoneal free gas must be distinguished from a primary gastrointestinal perforation.
Intravascular Gas
It has been suggested that the intravascular gas is pumped
under high pressure through the pulmonary lymphatics
into the systemic venous circulation (Booth et al, 1995).
The intravascular gas results in immediate cardiovascular
collapse and is often diagnosed when gas is seen in vessels
or in the heart chambers when a chest radiograph is taken
to determine the cause of cardiovascular collapse. Intravascular gas is usually a fatal complication.
AIRWAY COMPLICATIONS
Prolonged endotracheal intubation can produce airway
damage and subglottic stenosis. The risk is increased by a
too snugly fitting endotracheal tube, prolonged duration
of intubation, and traumatic intubation. Some infants may
require tracheostomy and even surgery to repair the stenosis. Inadequate humidification of the inspired gas can
produce necrotizing tracheobronchitis, a necrotic inflammatory process involving the trachea and main bronchi.
Infants present with acute respiratory deterioration due to
airway obstruction, hyperexpansion on chest radiograph,
and poor chest movement. Emergency bronchoscopy may
be necessary to relieve airway obstruction.
Atelectasis frequently occurs during prolonged ventilation and after extubation from mechanical ventilation.
This can also be secondary to airway injury due to trauma
produced by suction catheters and by inadequate conditioning of the inspired gas.
VENTILATOR-ASSOCIATED PNEUMONIA
Ventilator-associated pneumonia is a very common
complication of mechanical ventilation at any age,
and it is likely to occur even more frequently in the
632
PART X
Respiratory System
immune-compromised premature infant. Unfortunately,
it is not commonly diagnosed in this population because of
the lack of specific criteria and the difficulty of differentiating pneumonia from other acute and chronic pulmonary
pathologies observed previously in infants requiring prolonged mechanical ventilation. Despite these limitations, it
should be suspected any time there is deterioration in lung
function with radiographic changes suggestive of pneumonia. Changes in the amount and quality of the secretions
obtained from the airway and colonization with pathogens
is another indication of possible pulmonary infection that
may require antibiotic therapy. Some evidence indicates
that a large proportion of ventilated premature infants
have aspiration of gastric contents into their airways. This
is more common in fed infants and those receiving methylxanthines (Farhath et al, 2006). The consequences of this
are not clear, but this could further contribute to the risk
of pulmonary infection and chronic lung damage.
SUGGESTED READINGS
Avery ME: Recent increase in mortality from hyaline membrane disease, J Pediatr
57:553-559, 1960.
Bancalari A, Gerhardt T, Bancalari E, et al: Gas trapping with high-frequency ventilation: jet versus oscillatory ventilation, J Pediatr 110:617-22, 1987.
Bancalari E, Flynn J, Goldberg RN, et al: Influence of transcutaneous oxygen
monitoring on the incidence of retinopathy of prematurity, Pediatrics 79:663669, 1987.
Beck J, Reilly M, Grasselli G, et al: Patient-ventilator interaction during neurally
adjusted ventilatory assist in low birth weight infants, Pediatr Res 65:663-668,
2009.
Becker MA, Donn SM: Real-time pulmonary graphic monitoring, Clin Perinatol
34:1-17, 2007.
Bollen CW, Uiterwaal CS, van Vught AJ: Meta-regression analysis of highfrequency ventilation vs conventional ventilation in infant respiratory distress
syndrome, Intensive Care Med 33:680-688, 2007.
Carlo WA, Finer NN, Walsh MC, et al: Target ranges of oxygen saturation in
extremely preterm infants. SUPPORT Study Group of the Eunice Kennedy
Shriver NICHD Neonatal Research Network, N Engl J Med 362:1959-1969,
2010a.
Chang HY, Claure N, D’ugard C, et al: Effects of synchronization during nasal
ventilation in clinically stable preterm infants, Pediatr Res 69:84-89, 2011.
Chow LC, Vanderhal A, Raber J, et al: Are tidal volume measurements in neonatal
pressure-controlled ventilation accurate, Pediatr Pulmonol 34:196-202, 2002.
De Paoli AG, Davis PG, Lemyre B: Nasal continuous positive airway pressure
versus nasal intermittent positive pressure ventilation for preterm neonates: a
systematic review and meta-analysis, Acta Paediatr 92:70-75, 2003.
Finer NN, Carlo WA, Walsh MC, et al: Early CPAP versus surfactant in extremely
preterm infants, N Engl J Med 362:1970-1979, 2010.
Gregory GA, Kitterman JA, Phibbs RH, et al: Treatment of the idiopathic respiratory-distress syndrome with continuous positive airway pressure, N Engl J Med
284:1333-1340, 1971.
Hay WW Jr, Rodden DJ, Collins SM, et al: Reliability of conventional and new
pulse oximetry in neonatal patients, J Perinatol 22:360-366, 2002.
Keszler M, Abubakar K: Volume guarantee stability of tidal volume and incidence
of hypocarbia, Pediatr Pulmonol 38:240-245, 2004.
Moretti C, Giannini L, Fassi C, et al: Nasal flow-synchronized intermittent positive pressure ventilation to facilitate weaning in very low-birthweight infants:
unmasked randomized controlled trial, Pediatr Int 50:85-91, 2008.
Morley CJ, Davis PG, Doyle LW, et al: Nasal CPAP or intubation at birth for very
preterm infants, N Engl J Med 358:700-708, 2008.
Reyes ZC, Claure N, Tauscher MK, et al: Randomized, controlled trial comparing
synchronized intermittent mandatory ventilation and synchronized intermittent mandatory ventilation plus pressure support in preterm infants, Pediatrics
118:1409-1417, 2006.
Walsh M, Engle W, Laptook A, et al: Oxygen delivery through nasal cannulae to
preterm infants: Can practice be improved? National Institute of Child Health
and Human Development Neonatal Research Network, Pediatrics 116:857861, 2005.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
46
Respiratory Distress in the Preterm Infant
J. Craig Jackson
INTRODUCTION
This chapter on the causes of respiratory distress in the
preterm infant will focus primarily on respiratory distress
syndrome (RDS) also known as hyaline membrane disease and on surfactant replacement therapy. However, the
chapter also includes discussion of two less frequent causes
- pneumonia/sepsis and pulmonary hypoplasia. Other
causes of respiratory distress that are more commonly seen
in term infants, such as transient tachypnea of the newborn
(TTNB), are discussed in Chapter 47. Table 46-1 lists
causes of respiratory distress in newborns of all gestational
ages and demonstrates the relative frequency of each diagnosis. This table provides a useful differential diagnosis
for respiratory distress in preterm neonates, particularly if
polycythemia and hypoglycemia are included. More than
half of extremely low birthweight newborns will have some
type of respiratory distress (Figure 46-1). In that population respiratory distress syndrome is by far the most common diagnosis (50.8%), followed by transient tachypnea
of the newborn (4.3%) and pneumonia/sepsis (1.9%). In
higher birthweight preterm newborns, the incidence of any
type of respiratory distress is much lower, but in symptomatic babies, the incidence of RDS and pneumonia/sepsis is
lower, whereas TTNB is more common. Compared to the
three diagnoses featured in Figure 46-1, the incidence of
other causes of respiratory distress (see Table 46-1) is very
low in preterm infants. However, these less common diagnoses should be considered in preterm infants with respiratory distress who follow an atypical clinical course.
RESPIRATORY DISTRESS SYNDROME
During breathing (either spontaneous or assisted), shear
stresses in the alveoli and terminal bronchioles occur due
to the repetitive reopening of collapsed alveoli and the
overdistention of open alveoli (Nilsson et al, 1978). These
forces can quickly damage the fragile lung architecture,
leading to leakage of proteinaceous debris into the airways
(i.e., hyaline membranes). This debris (Figure 46-2) may
impair the function of what little surfactant is present,
leading to a downward spiral that may end in respiratory
failure and death if not interrupted.
If supportive therapy is successful, the repair phase
begins during the 2nd day after birth with the appearance
of macrophages and polymorphonuclear cells. Debris is
phagocytosed and the damaged epithelium is regenerated.
Edema fluid in the interstitium is mobilized into lymphatics, leading to the “diuretic” phase of RDS characterized
by high urine output.
With uncomplicated RDS, the patient improves by the
end of the first week after birth. However, infants born
at <1250 g and larger newborns needing high concentrations of oxygen and positive-pressure ventilation for severe
RDS may develop inflammation and inappropriate repair
of the growing lung, leading to emphysema and fibrosis
(see Chapter 48).
PURPOSE OF SURFACTANT
Surface tension is generated from molecular attractive
forces within a liquid that oppose spreading; it is the reason
that water ‘beads up” on a clean surface. If you try to inflate
a bubble under water with a straw (Figure 46-3, A), the
RISK FACTORS
The main risk factor for RDS, by far, is prematurity (see
Figure 46-1). Other factors that increase the risk of RDS
include perinatal asphyxia, maternal diabetes, lack of labor,
absence of antenatal steroid administration to the mother,
male gender, and White race. The central feature of RDS
is surfactant deficiency due to lung immaturity, commonly
a result of premature birth or delayed lung maturation
associated with maternal diabetes or male gender. Surfactant dysfunction can also be caused by perinatal asphyxia,
pulmonary infection, or excessive fetal lung liquid due to
delivery without labor.
PATHOPHYSIOLOGY OF RDS
Because alveoli with insufficient (or dysfunctional) surfactant are unstable and tend to collapse, patients with
RDS develop generalized atelectasis, ventilation-perfusion
mismatching, and subsequent hypoxemia and respiratory
acidosis.
TABLE 46-1 Causes of Respiratory Distress
Respiratory distress syndrome
Transient tachypnea of newborn
Pneumonia/sepsis
Meconium aspiration syndrome
Congenital cardiac malformation
Chromosomal disorder/multiple congenital anomalies
Spontaneous pneumothorax
Perinatal asphyxia
Pulmonary hemorrhage
Persistent pulmonary hypertension
Diaphragmatic hernia
Apnea of prematurity
Pulmonary hypoplasia
Pulmonary dysplasia
Hydrothorax
Postsurgical diaphragmatic palsy
46%
37%
5%
2%
2%
1.4%
1.2%
1.1%
1.0%
0.8%
0.8%
0.6%
0.3%
0.2%
0.2%
0.2%
100%
Data from Rubaltelli FF, Dani C, Reali MF, et al: Acute neonatal respiratory distress
in Italy: a one-year prospective study, Acta Paediatr 87:1261-1268, 1998.
633
634
PART X
Respiratory System
spreading of the water’s surface and enlargement of its surface area will be opposed by its surface tension (T). According to Laplace’s law, the pressure (P) required to inflate the
bubble is proportional to T divided by the radius of the
bubble (r). If you simultaneously try to inflate two interconnected bubbles, the smaller one will deflate into the
larger one, because of its smaller radius (Figure 46-3, B).
Surfactants are surface active materials that lower surface tension; a detergent is a type of surfactant which
causes water to spread out on a surface rather than beading
up. The surface tension of clean water resists the creation
60.0%
50.0%
of bubbles, but the addition of detergent allows bubbles
to form. Because the surfactant properties of detergents
do not persist, all of the small bubbles eventually collapse
into the large ones, and finally even the large ones collapse.
Lung surfactant has the miraculous property of reducing surface tension as the size of the bubble decreases.
In clean water with high surface tension, the pressure
required to keep open a bubble as the radius decreased
would be greater than the pressure keeping open a larger
bubble, so the smaller bubbles would eventually collapse
into larger ones (or small alveoli would collapse into larger
1.9%
4.3%
Pneumonia/sepsis
TTNB
RDS
0.84%
Incidence
40.0%
9.2%
30.0%
50.8%
20.0%
32.7%
0.28%
10.0%
0.0%
6.1%
6.6%
1000g
1000-1500g
1500-2500g
Birthweight
FIGURE 46-1 Incidence of respiratory problems in preterm newborns. (Data from Rubaltelli FF, Dani C, Reali MF, et al: Acute neonatal respiratory
distress in Italy: a one-year prospective study, Acta Paediatr 87:1261-1268, 1998.)
A
A
100 m
200 m
A
B
FIGURE 46-2 Photomicrographs of lung tissue from experimental animal with RDS, flash frozen during inflation. Note liquid-air interface
(arrows). The alveolar debris forms hyaline membranes. A, Low power. B, Higher power. (From Jackson JC, MacKenzie AP, Chi EY, et al: Mechanisms for
reduced total lung capacity at birth during hyaline membrane disease in premature newborn monkeys, Am Rev Respir Dis 142:413-419, 1990. American Thoracic Society.)
CHAPTER 46 Respiratory Distress in the Preterm Infant
ones), as shown in Figure 46-3, B. However, if the bubbles
are lined with good-quality surfactant, the surface tension
falls quickly as the radius gets smaller because the surfactant molecules become crowded during deflation (Figure
46-4). When the radius is very small, the surface tension
P
P
r
A
r2
r1
B
T
T
T
FIGURE 46-3 Effect of surface forces generated by inflating
bubbles under water. A, Single bubble of radius (r) resists inflation and
thus requires pressure (P) to overcome the surface tension (T). B, If the
surface tension is the same in two bubbles of unequal size, the smaller
one will collapse into the larger one, because of the Laplace relationship,
P = T/r (small r requires larger pressure to stay inflated).
P
P
r
T
T
r
Expiration
FIGURE 46-4 Surfactant molecules on surface crowding during
deflation. (From Jackson JC: Respiratory distress syndrome (figure 212–1).
In: Osborn LM, DeWitt TG, First LR, Zenel JA, editors: Pediatrics, Philadelphia, 2005, Elsevier Mosby.)
635
falls almost to zero, and the pressure required to keep
the smaller bubble open is negligible. Thus, it does not
collapse.
During inflation, as the radius of the bubble increases,
surface tension increases even faster. This means that the
amount of pressure in the larger bubbles will be higher
than in smaller ones; in the lung where bubbles are interconnected, this pressure difference will cause flow to the
smaller alveoli, thus keeping all the alveoli about the same
size.
The cumulative result of the alveoli remaining open
during deflation is a nonlinear pressure-volume relationship. The lung with sufficient surfactant retains gas during
expiration, compared to rapid and almost complete loss of
gas in the surfactant-deficient lung (Figure 46-5, A). During inflation, more pressure is required to achieve similar
tidal volume (Figure 46-5, B), because of poor compliance (∆V/∆P) from having to reopen collapsed alveoli. For
instance, to achieve a tidal volume of 5 mL/kg, an infant
with RDS may require a pressure increase of 25 cm H2O;
dividing the volume change, ∆V, by pressure change, ∆P,
we calculate that the compliance is only 0.25 mL/kg/cm
H2O, which is about one third of normal.
In the absence of adequate amounts of functional surfactant in the newborn lung, there is widespread alveolar collapse with overdistention of open alveoli (Figure
46-6). Because reopening collapsed alveoli requires high
pressure, the spontaneously breathing newborn with surfactant deficiency must generate highly negative intrathoracic pressure. Clinically, this is manifested by retractions
of the respiratory muscles during inspiration. Newborns
may also attempt to prevent alveolar collapse by grunting. This partial closure of the glottis during expiration
helps maintain an end-expiratory pressure that may keep
unstable alveoli open.
A consequence of widespread alveolar collapse is intrapulmonary shunting of blood past atelectatic lung, without the opportunity for blood in pulmonary capillaries
to pick up oxygen from, or deliver carbon dioxide to, the
alveoli. In addition, lungs that are poorly inflated have
widespread collapse of pulmonary vessels, leading to pulmonary hypertension. The elevated pulmonary artery
pressures lead to right-to-left shunting of unoxygenated
Healthy
With surfactant
Without surfactant
Volume
Volume
V
P
RDS
V
P
A
Pressure
B
Pressure
FIGURE 46-5 Effect of surface forces on pressure-volume relationships. During deflation (A), the lungs with surfactant retain gas even at very
low pressures, because of falling surface tension as the alveoli get smaller. The alveoli without surfactant collapse as the alveoli get smaller. During
inflation of RDS lungs (B), the starting lung volume (functional residual capacity) is lower, and much more pressure is required during inflation,
compared to healthy lungs.
636
PART X
Respiratory System
O
N
O
O P O
O
O
O
O
FIGURE 46-7 The main ingredient of lung surfactant, dipalmitoylphosphatidylcholine (DPPC).
A
for low viscosity for optimal spreading and redistribution
along the smallest airways with the need for a stable and
low surface tension.
Surfactant phospholipids are assembled in the type II
pneumocytes of the lung epithelium into lamellar bodies in
the form of bilayered membranes (Figure 46-9). Surfactantassociated proteins SP-B and SP-C are essential for the
transition to a monolayer at the air-liquid interface. The
molecular structure of the hydrophobic SP-B is complex
and it interacts with the phospholipid monolayer as shown
in Figure 46-10. Its absence is associated with fatal neonatal
respiratory failure. Surfactant-associated proteins SP-A and
SP-D are hydrophilic and have roles in immune defense.
SP-A is involved in reuptake and reuse of secreted surfactant
(see Chapter 42).
CLINICAL SIGNS OF RDS
B
FIGURE 46-6 Histology of RDS. Scanning electron micrograph of
lung frozen during inflation with air, in premature monkey healthy premature monkey (A) compared to one with RDS (B). Lungs affected by
RDS have collapsed alveoli full of liquid and proteinaceous debris, with
overdistended terminal airways. (From Jackson JC, Truog WE, Standaert
TA, et al: Effect of high-frequency ventilation on the development of alveolar
edema in premature monkeys at risk for hyaline membrane disease, Am Rev
Respir Dis 143:865-871, 1991. American Thoracic Society.)
blood across the patent ductus arteriosus to the descending
aorta (see Chapter 52).
ORIGIN AND COMPOSITION OF SURFACTANT
Pulmonary surfactant is composed of approximately
90% lipids and 10% proteins. The main phospholipid
in surfactant is dipalmitoylphosphatidylcholine (DPPC),
also known as lecithin. It is surface active because of its
hydrophilic head and hydrophobic tails (Figure 46-7).
However, DPPC by itself does not adsorb efficiently at
the air-liquid interface and is in the form of a gel at body
temperature. The presence of some unsaturated phospholipids and cholesterol helps to make it more fluid (Mingarro et al, 2008).
The remainder of surfactant’s ingredients is shown in
Figure 46-8. One of them, phosphatidylglycerol (PG), is
sometimes used as a marker of lung maturation; it interacts
with the hydrophobic surfactant proteins to improve biophysical activity. Even minor components of pulmonary
surfactant play important roles; for instance, free fatty
acids improve the stability of the interfacial film, especially
after repeated compression. The composition of surfactant is complex because it has evolved to balance the need
The cardinal clinical signs of RDS are tachypnea, grunting,
and increased work of breathing (Box 46-1). The newborn
respiratory rate is elevated in an attempt to increase the
exchange of oxygen and carbon dioxide, but with exhaustion, the rate may decline or even stop. Grunting is used to
create positive pressure in the lungs to reduce the collapse of
air sacs. Signs of increased work of breathing include nasal
flaring and retraction of respiratory muscles, especially the
intercostal and subcostal muscles. Because the ribcage in
premature infants is so flexible, the sternum may deeply
retract during inspiration. Cyanosis results from inadequate
oxygenation, and pallor from acidosis due to poor elimination of carbon dioxide. The combination of increased work
of breathing, cyanosis, and acidosis causes lethargy and disinterest in feeding, and eventually apnea. Rather than progressing through these signs during the first hours of life,
newborns with intrapartum asphyxia or extreme prematurity may present with apnea immediately following birth.
On auscultation, breath sounds may be distant or shallow from the fast inspiratory rate and low tidal volume,
and fine inspiratory rales may be heard due to reopening of moist, collapsed air sacs. The onset of symptoms is
always within hours after birth and, in severe cases, may
occur with the first few breaths after delivery. In general,
the respiratory distress from RDS tends to get worse over
the first 1 to 3 days after birth, and then usually improves
gradually over a few days (although the natural course may
be interrupted by exogenous surfactant therapy).
Differential Diagnosis
Clinical improvement during first 12 hours after birth
suggests TTNB, and onset after the first 24 hours suggests pneumonia and sepsis. Tachypnea without increased
work of breathing suggests cyanotic heart disease; the
diagnosis of obstructed pulmonary veins from total
637
CHAPTER 46 Respiratory Distress in the Preterm Infant
Phosphatidylglycerol
Other
phospholipids
SP-A
5%
SP-B
2%
SP-C
2%
SP-D
1%
8%
Neutral
Lipids
8%
8%
6%
Unsaturated
Phosphatidylcholine
20%
Saturated
Phosphatidylcholine
50%
FIGURE 46-8 Composition of surfactant. (From Jobe AH, Ikegami M: Biology of surfactant, Clin Perinatol 28:655-669, 2001, WB Saunders.)
Alveolar type II cell
Alveolar type I cell
SP-B
SP-A
Lamellar body
Tubular myelin
Surfactant layer
SP-C
Air space
Alveolar fluid
Alveolar macrophage
FIGURE 46-9 Assembly of surfactant. (Republished and adapted from
Hawgood S, Clements JA: Pulmonary surfactant and its apoproteins, J Clin
Invest 86:1-6, 1990. The American Society for Clinical Investigation.)
anomalous venous return is occasionally confused with
RDS. Hypoventilation without increased work of breathing suggests a central nervous system problem such as
intracranial hemorrhage or asphyxia. Asymmetric breath
sounds may be due to pneumothorax (which is a complication of RDS), congenital diaphragmatic hernia, or unilateral pleural effusion. Meconium staining of amniotic fluid
suggests the possibility of meconium aspiration syndrome,
but this is rare in premature infants—green-stained amniotic fluid in this population is more likely to be due to
infection or to bile refluxed into the esophagus because of
intestinal obstruction, rather than meconium.
LABORATORY FEATURES OF RDS
Initially, the arterial blood gases or oxygen saturation
monitor may show only hypoxemia or desaturation. The
Paco2 may be normal because of tachypnea, but is almost
FIGURE 46-10 Structure and interactions of surfactant and proteins.
(From Pérez-Gil J: Molecular interactions in pulmonary surfactant films, Biol
Neonate 81:6-15, 2002. Karger AG Basel.)
BOX 46-1 Clinical Signs of RDS
Tachypnea
Grunting
Increased work of breathing
Nasal flaring
Retraction of respiratory muscles (intercostal, subcostal, sternal)
Cyanosis
Pallor
Lethargy
Disinterest in feeding
Apnea
638
PART X
Respiratory System
always elevated. Later, as the infant tires, the Paco2 will
rise further and cause respiratory acidosis. With imminent
respiratory failure, there may be metabolic acidosis due
to inadequate oxygen delivery to tissues, and from poor
peripheral perfusion due to respiratory acidosis.
Differential diagnosis: Extremely elevated Paco2 within
minutes of birth suggests pulmonary hypoplasia, tension pneumothorax, congenital diaphragmatic hernia, or
obstruction of the airways due to debris or an anatomic
cause. The tachypneic, cyanotic newborn with low Paco2
may have transient tachypnea of the newborn or cyanotic
congenital heart disease. A positive blood culture suggests
pneumonia and sepsis. Low blood glucose (<40) suggests
symptomatic hypoglycemia, and high hematocrit (>65)
suggests symptomatic polycythemia.
RADIOGRAPHIC FEATURES OF RDS
The classic radiographic findings of RDS include a reticulogranular (i.e., ground-glass) pattern and air bronchograms (Figure 46-11). The lungs are diffusely and
homogeneously dense because of widespread collapse of
alveoli. The appearance is reticular (i.e., netlike) because
the small airways are open (black) and surrounded by interstitial and alveolar fluid (white); in severe cases, the lungs
may appear complete white on the film. Air bronchograms
are commonly seen because the large airways beyond the
second or third generation are more visible than usual as
a result of radiodensity from engorged peribronchial lymphatics and fluid-filled or collapsed alveoli. Another cardinal feature is low lung volume (e.g., the diaphragms are at
the eighth rib level or higher) due to widespread alveolar
collapse and low functional residual capacity.
Differential diagnosis: Normal or high lung volumes,
especially with prominent interstitial fluid pattern, suggest
TTNB. In this case, there are coarse white lines (engorged
lymphatics and interstitial water) radiating from the hilum
rather than the crisp black lines (air bronchograms) of RDS.
Other causes of a coarse (rather than diffuse) fluid pattern
include pneumonia with sepsis, and obstructed pulmonary
venous drainage due to total anomalous pulmonary venous
return. An abnormal cardiac silhouette or size should suggest congenital heart disease, and asymmetry of the lungs
suggests pneumothorax, congenital diaphragmatic hernia,
or lung anomaly. Very low lung volumes, especially with
pneumothorax, may indicate pulmonary hypoplasia.
INITIAL DIAGNOSTIC EVALUATION
FOR POSSIBLE RDS
The initial diagnostic evaluation (Box 46-2) is shaped by
the differential diagnoses listed in Table 46-1, and by the
urgency to intervene quickly for serious but infrequent
conditions such as bacterial pneumonia and sepsis, hypoglycemia, and polycythemia. It is often helpful to obtain
both anteroposterior and lateral radiographs for the initial
evaluation. If congenital heart disease is suspected on clinical grounds, an echocardiogram is indicated.
TREATMENT OF PATIENTS WITH RDS
The treatment of patients with RDS is outlined in
Box 46-3. All patients with RDS need the basics of warmth,
hydration, and nutrition appropriate for the degree of prematurity, as described in other chapters. Because pulmonary edema contributes to surfactant dysfunction in RDS,
it is important to avoid excessive intravenous (IV) fluid
administration.
Newborns with significant tachypnea (e.g., more than
60 breaths per minute) or increased work of breathing
BOX 46-2 Initial Diagnostic Evaluation
for Possible RDS
Arterial or capillary blood gas
Blood glucose
Complete blood count
Blood culture
AP and lateral chest radiograph
Echocardiogram (if clinically indicated)
BOX 46-3 Treatment of Established RDS
FIGURE 46-11 Radiograph of RDS. Note low lung volumes and
reticulogranular pattern. (From Welty S, Hansen TN, Corbet A: Respiratory distress in the preterm infant. In Taeusch HW, Ballard RA, Gleason
CA, editors: Avery’s diseases of the newborn, ed 8, Philadelphia, 2005,
Elsevier.)
Warmth by radiant warmer or incubator
Hydration at approximately 60–80 mL/kg/d
Nutrition
Initially D5W or D10W (with protein, if possible)
NPO if respiratory rate over 60 or moderate/severe work of breathing
Gavage feeds if stable
Consider parenteral nutrition if enteral feeds are delayed
Antibiotics if at risk for pneumonia and sepsis
Supplemental oxygen
Oxygen saturation monitoring, with appropriate target for infants at risk for
retinopathy of prematurity
Exogenous surfactant
CPAP or mechanical ventilation, as needed
CPAP, Continuous positive airway pressure.
CHAPTER 46 Respiratory Distress in the Preterm Infant
(moderate or severe) often do not have the energy required
for oral feeding, and there is some risk of aspiration if nipple feeding is attempted. Initially, IV fluids and nothing
by mouth (NPO) status may be appropriate, with consideration of small gavage tube gastric feedings if the baby
is otherwise stable. Parenteral nutrition may be indicated
because of the increased caloric expenditures associated
with work of breathing. Antibiotics should be considered
unless the risk of pneumonia and sepsis is negligible.
The techniques of respiratory support are described in
other chapters. See Chapter 45 regarding risks and benefits of different modes of respiratory therapy.
RESPIRATORY COMPLICATIONS OF RDS
Air leak complications occur in patients with RDS because
of the asymmetry of alveolar inflation and the sheer stresses
in terminal bronchioles, leading to dissection of air into the
interstitium (causing pulmonary interstitial emphysema)
and through the visceral pleura (causing pneumothorax).
The former can be seen in up to 50% of patients with
RDS, and the latter in 5% to 10%, even in those treated
with exogenous surfactant.
Hemorrhagic pulmonary edema (i.e., pulmonary hemo
rrhage) occurs more frequently in the most premature
infants, probably due to left ventricular failure and excessive left-to-right flow through a patent ductus arteriosus,
with resultant disruption of the pulmonary capillaries
(Cole et al, 1973). It may also be related to insufficient
attention to reducing mechanical ventilator settings after
lung compliance improves following exogenous surfactant
treatment. Onset is typically at 1 to 3 days of age, with
sudden respiratory deterioration associated with pink or
red frothy fluid in the ET tube, and widespread white-out
on chest radiograph. Treatment considerations include
increased positive end-expiratory pressure (PEEP); there
is also evidence that exogenous surfactant may be beneficial (Pandit et al, 1995).
Bronchopulmonary dysplasia (BPD), also known as
chronic lung disease, is probably due to abnormal lung
repair following lung injury from RDS (see Chapter 48).
PREVENTION OF RDS
Box 46-4 outlines the key measures for prevention and
treatment of RDS.
Antenatal Steroids
Antenatal steroids were shown in 1972 to be effective
in reducing the risk of RDS (Liggins and Howie, 1972).
A consensus panel convened by the National Institutes of
BOX 46-4 Prevention of RDS
Antenatal steroids
Prevention of asphyxia
Continuous positive airway pressure
Exogenous surfactant
Surfactant, then CPAP
CPAP, Continuous positive airway pressure.
639
Health concluded that “antenatal corticosteroid therapy
is indicated for women at risk of premature delivery with
few exceptions and will result in a substantial decrease in
neonatal morbidity and mortality, as well as substantial
savings in health costs” (NIH Consensus Panel, 1995).
They recommended that the treatment be used from 24
to 34 weeks’ gestation without limitation by gender or
race, and whether or not surfactant therapy was available.
Although the beneficial effects were found to be greatest if treatment was begun more than 24 hours before
delivery, there was also a benefit when given for less than
24 hours.
A metaanalysis in 2006 (Roberts and Dalziel, 2006)
reviewed 21 published trials and concluded that antenatal
corticosteroids did not increase a mother’s risk of death,
chorioamnionitis, or puerperal sepsis. Treatment was
associated with a 31% reduction in neonatal death, 34%
reduction in RDS, 46% reduction in cerebroventricular
hemorrhage, and 54% reduction in necrotizing enterocolitis (all significant at p <0.05 or less).
Although there is now widespread consensus on use
of antenatal steroids, many issues remain controversial,
including the type of corticosteroid to use; the dose, frequency, and timing of use; and the route of administration.
A metaanalysis in 2008 (Brownfoot et al, 2008) reviewed
10 trials and found that antenatal dexamethasone appears
to decrease the incidence of intraventricular hemorrhage
but possibly increases the rate of NICU admissions, compared with betamethasone. Oral antenatal dexamethasone
was found in one study to increase the incidence of neonatal sepsis compared to intramuscular drug (Egerman
et al, 1998). More research is needed to optimize antenatal
steroid administration.
Because the effectiveness appears to wane if antenatal
steroids are given more than 1 week before premature
delivery, several trials have been conducted to determine
whether one or more repeat doses at weekly intervals was
beneficial. Although a metaanalysis (Crowther and Harding, 2007) found modest improvements in occurrence and
severity of RDS, there were concerning findings of smaller
birthweight and head circumference, indicating that there
is insufficient evidence to recommend repeated doses
(Bonanno and Wapner, 2009).
Prevention of Asphyxia
Prevention of asphyxia may decrease the incidence and
severity of RDS, because asphyxia leads to hypoxemia
and acidosis, which reduce surfactant synthesis. Leakage of fluids from capillaries into alveoli may also impair
surfactant function. Thus, delivery by cesarean section
should be considered for signs of fetal distress if the fetus
is deemed to be viable, or if the fetus is in the breech
presentation during labor. Compliance with consensus
neonatal resuscitation techniques outlined by the American Academy of Pediatrics and American Heart Association (Kattwinkel, 2006) is critical, especially because
premature infants are at much higher risk for needing
intervention at birth. Maternal transfer to a center experienced in management of premature infants, if it can be
accomplished safely, is associated with improved neonatal outcome.
640
PART X
Respiratory System
Continuous Positive Airway Pressure
Gregory et al (1971) first introduced continuous positive
airway pressure (CPAP) for newborns with RDS, primarily by endotracheal tube. Since then, a variety of devices
have been developed, including short nasal prongs that do
not add much to the work of breathing (De Paoli et al,
2002). Avery et al (1987) reported that centers that used
early nasal CPAP for RDS had a lower incidence of chronic
lung disease.
The goals of CPAP are to prevent end-expiratory alveolar collapse, reduce the work of breathing, and better
match ventilation to perfusion. If started in the delivery
room, it may help the newborn establish functional residual capacity, in addition to stabilizing the chest wall and
reducing airway resistance. Furthermore, adequate expansion of the lungs at birth improves pulmonary blood flow.
Some have speculated that prophylactic use of CPAP in
the delivery room might make intubation for exogenous
surfactant unnecessary (De Klerk and De Klerk, 2001),
and that avoidance of intubation and mechanical ventilation may lower the risk of BPD (see Chapter 48).
The COIN Trial randomized 25- to 28-weeks’ gestation
newborns who were breathing spontaneously at 5 minutes
of age either to CPAP or to prophylactic intubation with
early extubation to CPAP if possible. The investigators
found that 46% of the CPAP group eventually required
intubation anyway during the first 5 days, and the rate
of pneumothorax was 9% compared to only 3% in those
intubated routinely (p <0.001) (Morley et al, 2008). This
suggests that routine intubation may be safer than immediate CPAP in the extremely premature infant, but it also
demonstrated that some patients in this extremely highrisk population do well with early CPAP. Success with
CPAP, especially in this population, requires considerable
experience and skill (Bohlin et al, 2008).
Recently, some centers have begun using humidified
high-flow nasal cannulas in infants with RDS (Lampland,
2009) to achieve the benefits of positive airway pressure
with fewer of the perceived disadvantages of CPAP (challenges in keeping the nasal prongs in the nares, easier handling of the patient, and less risk of pressure necrosis of
the nasal septum). However, the pressure generated from
this therapy, although proven to produce clinical effect,
is variable, unpredictable, and unregulated, and the commercially available systems are not approved by the U.S.
Food and Drug Administration (FDA) for this indication.
It should only be used by practitioners aware of the balance of risks and benefits, and those prepared to recognize
and treat pneumothorax and respiratory failure.
The use of CPAP following tracheal administration of
exogenous surfactant is discussed later in the section on
timing of surfactant administration.
Exogenous Surfactant
Historical Summary
The development of exogenous surfactant for treatment
of RDS is one of the most important advances in the history of newborn medicine. This history is well told elsewhere (e.g., Halliday, 2008). The key milestones include
von Neergaard’s discovery in 1929 that surface tension
contributes to lung recoil, Gruenwald’s demonstration
in 1947 that lungs of stillborn infants have high surface
tension, Prattle’s speculation in 1955 that absence of surfactant active material contributes to RDS, Clement’s
description in 1957 of surfactant dysfunction in experimental animals, and Avery and Mead’s demonstration
in 1959 that RDS in human infants is due to surfactant
deficiency. There was an increase in research interest and
funding for RDS treatment after U.S. President Kennedy’s son, born at 34 weeks’ gestation, died of RDS in 1963.
In 1972, Enhorning and Robertson used natural surfactant
to delay the progression of RDS in preterm rabbits, and in
1980 Fujiwara et al demonstrated the first successful use
of exogenous surfactant in human infants. By 1990, exogenous surfactant was widely used throughout the developed
world, and many large clinical trials have been conducted
since then to refine and improve surfactant treatment and
prevention of RDS. Compared to standard therapy without surfactant, a metaanalysis of 13 randomized controlled
trials suggests that animal-derived exogenous surfactant
reduces the risk of pneumothorax by 58%, pulmonary
interstitial emphysema by 55%, mortality by 32%, and the
combined outcome of BPD or death by 17% (Seger and
Soll, 2009).
Types of Surfactant Available
A list of commonly used surfactant preparations can be
found in Table 46-2. The first generation of commercially
available artificial surfactants (e.g. Exosurf [colfosceril])
was composed mainly of DPPC and did not have SP-B
or SP-C. However, a metaanalysis of 11 randomized controlled trials showed that natural surfactants were faster
acting than artificial surfactants with lower incidence of
pneumothorax and mortality (Soll and Blanco, 2001).
TABLE 46-2 Surfactant Preparations and Their Sources
Brand Name
Generic Name
Constituents
Nonprotein Synthetic Surfactants
Adsurf
Pumactant
(ALEC)
DPPC, PG
Exosurf
Colfosceril palmitate
DPPC
Protein-Containing Animal Surfactants
Curosurf
Poractant α
Porcine lung tissue
Alveofact
SF-RI 1
Bovine lung lavage
BLES
Bovine lipid
extract surfactant
Bovine lung lavage
Infasurf
Calactant CLSE
Bovine (calf) lung lavage
Surfacten
Surfactant-TA
Bovine lung homogenate
Survanta
Beractant
Bovine lung tissue
Peptide-Containing Synthetic Surfactants
Venticute
rSP-C surfactant
DPPC, POPG, PA, rSP-C
Surfaxin
Lucinactant
DPPC, POPG, PA, KL4
Adapted from Sinha S, Moya F, Donn SM: Surfactant for respiratory distress syndrome: are there important clinical differences among preparations? Curr Opin Pediatr
19:150-154, 2007.
ALEC, Artificial lung-expanding compound; DPPC, dipalmitoylphosphatidylcholine;
PG, phosphatidylglycerol; rSP-C, recombinant human SP-C; POPG, palmitoyl-oleoyl
phosphatidylglycerol; PA, palmitic acid.
CHAPTER 46 Respiratory Distress in the Preterm Infant
Clinical trials comparing the available natural surfactants have been inconclusive. Speer et al (1995) showed no
differences in a small study, whereas Baroutis et al (2003)
showed differences in days of intubation, oxygen use, and
duration of hospitalization, but not survival. A comparison of Infasurf and Survanta by Bloom and Clark (2005)
was stopped because of lack of statistical significance in
the primary outcome variable. Ramanathan et al (2004)
compared Curosurf and Survanta, finding that the former
was less costly because of fewer doses, but there were no
important clinical differences in outcome.
A new generation of synthetic surfactants is under
development to lower cost and because of theoretical
concerns about immunologic or infectious complications
from animal-derived surfactant. These new products have
synthetic peptides or proteins, such as KL4 in lucinactant,
which mimics the actions of natural surfactant-associated
proteins SP-B and SP-C. Lucinactant has been shown
to be superior to older synthetic surfactants (Sinha et al,
2005), but its superiority to animal surfactants has not
been conclusively proven (Moya et al, 2005). In addition,
there are uncertainties regarding the metabolic fate of
lucinactant and its component chemicals, and there may be
risks from the requirement to convert the lucinactant gel
into liquid by using a special warming cradle immediately
before instillation (Engle et al, 2008); the authors added
that “efforts to develop more effective and safer surfactant
formulations continue to be warranted because of concerns with animal-derived surfactants for transmission of
microbes, exposure to animal proteins and inflammatory
mediators, susceptibility to inactivation, and inconsistent
content.”
Surfactant Selection
The important issue for clinical providers and managers is
which surfactant to stock in their pharmacy, because of the
expense of the preparations and the need to standardize
dosing guidelines. Compared to first-generation synthetic
surfactants, current studies strongly support use of natural
surfactants because of reduced risk of air leak and more
rapid response to treatment. Among natural surfactants
that are commercially available, no differences in longterm outcomes have been conclusively proven. However,
poractant alfa (Curosurf) when given as prophylaxis with
an initial dose of 200 mg/kg appears to have some advantages when compared to beractant (Survanta) and calfactant (Infasurf), such as fewer repeat doses, quicker oxygen
weaning, and potentially less cost. The benefit may be
related to the larger amount of phospholipid in each dose.
For hospitals currently using beractant and calfactant, the
advantages of poractant alfa must be considered in light of
the logistical issues and potential drug errors when a hospital changes from one preparation to another, especially
when there may be better products on the horizon.
routinely intubated. The treatment appeared feasible and
safe, but the sample size (23) was too small to prove that
the approach was beneficial compared to surfactant instillation via endotracheal tube during the first minutes after
delivery.
Prophylactic surfactant generally refers to the administration of doses within the first 15 minutes after birth. To
avoid giving an unnecessary, expensive medication and subjecting a newborn to the risks of tracheal intubation, prophylactic endotracheal surfactant should ideally be given
only to patients who would have eventually developed RDS
and met treatment criteria for surfactant anyway. However, in practice, the clinician can only select those patients
at highest risk and for whom clinical trials indicate that the
benefits of prophylactic surfactant outweigh the risks. One
such population is the baby born at <30 weeks’ gestation
whose mother was not given antenatal steroids (Table 46-3).
In the highest risk populations, a metaanalysis of eight
clinical trials suggests that prophylactic surfactant reduces
mortality (by 39%), frequency and severity of RDS, pneumothorax (by 38%), and the combined outcome of BPD
and death, compared to infants who received placebo or
rescue surfactant (Soll and Morley, 2001). However, the
criteria for selecting the high risk infants remain controversial. Table 46-3 provides one practical approach for the
clinician who must make a decision in the delivery room
as to whether to begin surfactant immediately or to wait.
However, some centers manage even 25-week gestational
newborns with initial CPAP, with only one third eventually needing surfactant (Aly et al, 2004). A randomized
study of 27 to 31 weeks’ gestation newborns with signs of
early RDS demonstrated that infants intubated for surfactant during the first hour of life followed immediately
by extubation and CPAP had a lower rate of subsequent
ventilation, incidence of pneumothorax, and chronic lung
disease compared to newborns treated with early CPAP
alone (Rojas, 2009). This observation may be especially
important for hospitals not equipped to provide mechanical ventilation for this population.
Early rescue surfactant (usually defined as treatment
at 1 to 2 hours after birth) may be indicated for infants of
<30 weeks’ gestation at the first signs of RDS. The clinical trials showing superiority of prophylactic surfactant
over rescue (Soll and Morley, 2001) compared prophylaxis to late rescue (needing mechanical ventilation and
>40% Fio2 and typically treated 4 to 6 hours after birth).
TABLE 46-3 Sample Algorithm for When to Begin Exogenous
Surfactant
When to Give First
Dose of Surfactant
Populations to Consider
Prophylactically, within
15 minutes after birth
<26 weeks’ gestation
and
26 to 30 weeks if (a) no antenatal steroids or (b) needs intubation anyway
Early rescue, preferably in
first 60 minutes after birth
<30 weeks’ gestation at first signs
of RDS
Treatment of established
RDS, preferably started
within 12 hours after
birth
All newborns with established RDS,
regardless of gestational age, if
they need ventilator and at least
30–40% O2
Timing of Surfactant Administration
In theory, surfactant would ideally be given with the first
breath. This concept was evaluated by Kattwinkel et al
(2004) by delivering the head of the infant, suctioning
the nasopharynx, and then instilling calfactant into the
airway before delivery of the shoulders. CPAP was initiated immediately after delivery, but the trachea was not
641
642
PART X
Respiratory System
One clinical trial showed clinical advantages of prophylactic compared to early-rescue surfactant (Kattwinkel
et al, 1993) but a historically controlled study showed that
aggressive use of CPAP led to a large reduction in the need
for any surfactant at all and was associated with lower rates
of BPD and other morbidity (De Klerk and De Klerk,
2001). Furthermore, immediate intubation in the delivery
room may be associated with significant procedure morbidity, including apnea and bradycardia. Thus, there is no
compelling evidence favoring prophylactic over early rescue surfactant for preterm infants at the time of this writing, except in those patients at highest risk (see Table 46-3)
such as extremely preterm infants whose mothers did not
receive antenatal steroids (Sweet and Halliday, 2009).
Late rescue surfactant: Clinical trials have firmly established the benefits of surfactant compared to no surfactant in preterm infants with established RDS (Engle et al,
2008). However, as noted previously, prophylactic or early
rescue surfactant is more beneficial than late rescue in the
highest-risk populations. In lower-risk populations (e.g.,
near-term infants) it is likely that the late rescue approach
offers a better balance of benefit, risk of intubation, and
cost efficiency than treatment before or at the earliest
onset of RDS. Entry criteria in clinical trials of rescue
surfactant compared to no surfactant usually required that
RDS be severe enough to require mechanical ventilation
and at least 30 to 40% supplemental oxygen. Many of the
trials were conducted before nasal CPAP was widely used,
so the indications for intubating and administering surfactant in infants tolerating CPAP are unknown. However,
those with a requirement for very high concentration of
supplemental oxygen (e.g., >80%) or with sustained severe
hypercarbia (e.g., Paco2 >60 torr) will likely benefit.
Combining surfactant therapy with CPAP: Because of
advances in use of CPAP since the first clinical trials of
surfactant, some have advocated immediate extubation
to CPAP after surfactant dosing (Verder et al, 1999) to
avoid complications from the endotracheal tube (including
excessive tidal volumes and airway inflammation that may
lead to BPD). Stevens et al (2007) reviewed six randomized controlled clinical trials of this treatment approach
in infants with clinical signs of RDS. The “early rescue/
CPAP” group was intubated for early surfactant therapy
followed by immediate extubation to nasal CPAP. In comparison to the late rescue group, early rescue/CPAP was
associated with a 33% reduction in need for mechanical
ventilation, 48% reduction in air leak syndrome, and 49%
reduction in incidence of BPD. They also noted that initiation of surfactant treatment at an Fio2 of less than 0.45
was associated with lower incidence of air leak, compared
to waiting until the Fio2 was higher.
Methods of Surfactant Dosing
Animal studies suggest that surfactant distribution is better distributed when administered as a bolus rather than
by infusing it slowly over several minutes (Fernandez et al,
1998). The package inserts of many surfactant preparations
recommend moving the infant into multiple positions for
better distribution of surfactant, but there are no clinical
trials supporting one particular method. Therefore, surfactant should be given as quickly as tolerated, and with
the least disruptive infant positioning.
Exogenous surfactant is generally given via an endotracheal tube. However, tracheal intubation is potentially
risky, and sometimes difficult to accomplish, so administration through a laryngeal mask airway (LMA) may be
considered. In a pilot study of 8 preterm infants (body
weight range 880 to 2520 g) receiving nasal CPAP for
RDS, the LMA administration approach appeared to be
safe and potentially effective (Trevisanuto et al, 2005).
Number of Surfactant Doses
and Dosing Intervals
A metaanalysis of two clinical trials that compared one versus multiple doses of animal-derived surfactant suggests a
49% reduction in incidence of pneumothorax and a trend
toward a 37% reduction in mortality when multiple doses
are used (Soll and Ozek, 2009). There are no data to suggest that dosing should continue once the patient’s ventilator and oxygen requirement are at minimal levels, or
beyond four doses. The interval between doses is usually at
least 6 hours, and most protocols recommend discontinuation of dosing after 48 hours. In an attempt to reduce the
incidence of BPD, clinical trials of surfactant administration at several days of age are underway.
Clinical Care after Dosing
Because natural surfactants may work quickly, the clinician must be prepared after dosing to immediately lower
the Fio2 while carefully monitoring the pulse oximeter.
The tidal volume, as measured by the ventilator and/or by
careful observation of chest wall movement, may gradually increase, resulting in a need to lower inspiratory pressures to avoid air leak syndrome, lung injury, and possibly
pulmonary hemorrhage. Blood gases should be monitored
by transcutaneous monitors and/or intermittent sampling. PEEP should be maintained but may be reduced if
the starting levels at dosing were high, given that functional residual capacity increases shortly after surfactant
administration.
A poor response to exogenous surfactant may occur
because the patient does not have surfactant deficiency but
rather, lung hypoplasia, pneumonia, or congenital heart
disease. Other causes for a lack of response may be poor
distribution of the surfactant, such as administration down
the right stem bronchus due to malposition of the endotracheal tube, plugging of the tube, or malposition of the
tube in the esophagus. A less likely reason is an inadequate
dose of surfactant.
The rapid improvement in lung compliance after exogenous surfactant therapy may lead to excessive pulmonary
blood flow from left-to-right shunting from a patient ductus arteriosus (Raju and Langerberg, 1993). It is uncertain
whether early and aggressive intervention to close the ductus with medication or surgery will reduce the risk of pulmonary hemorrhage.
PULMONARY HYPOPLASIA
DEFINITION AND INCIDENCE
One or both lungs of newborns with pulmonary hypoplasia are smaller than normal, including reduced numbers of lung cells, airways, blood vessels, and alveoli.
CHAPTER 46 Respiratory Distress in the Preterm Infant
Pulmonary hypoplasia is the cause of respiratory distress
at birth in only 0.3% of newborns with respiratory symptoms (Rubaltelli et al, 1998), but it is commonly fatal,
especially in preterm infants. The incidence is about
1 per 1000 live births, and 90% are associated with congenital anomalies or pregnancy complications (Churg
et al, 2005). There is a continuum of severity of pulmonary hypoplasia—from negligible to severe—and therefore all but the most severe cases are difficult to diagnose.
This is especially true when attempting to diagnose prenatally by imaging studies, but is also true during clinical
assessment of the newborn, and even after postmortem
examination of the lungs.
When considering the causes of pulmonary hypoplasia,
it is useful to categorize them into associated conditions,
each with representative diagnoses, as in Table 46-4. This
chapter on respiratory distress in preterm infants focuses
on the problem of pulmonary hypoplasia associated with
oligohydramnios from preterm premature rupture of
membranes (PPROM) because this cause of pulmonary
hypoplasia occurs almost exclusively in preterm infants.
When PPROM occurs at 15 weeks’ gestation, the incidence
TABLE 46-4 Categories of Conditions Associated
With Pulmonary Hypoplasia
Category
Representative Diagnoses
Restriction of thoracic
space
Diaphragmatic hernia or eventration
Intrathoracic mass
Congenital cystic adenomatoid
malformation
Bronchogenic cyst
Extralobar sequestration
Thoracic neuroblastoma
Pleural effusions
Chylothorax
Hydrothorax
Oligohydramnios
Renal
Bilateral renal agenesis or dysplasia
Bladder outlet obstruction
(posterior urethral valves)
Nonrenal
Prolonged preterm rupture of
membranes
Skeletal anomalies
Chondroectodermal dysplasia
Osteogenesis imperfecta
Thanatophoric dwarfism
Hydrops fetalis
Rh isoimmunization
Neuromuscular and
central nervous system
anomalies
Fetal akinesia
Anencephaly
Arnold-Chiari malformation
Cardiac anomalies
Hypoplastic right or left heart
Pulmonary stenosis
Ebstein’s anomaly
Abdominal wall defects
Omphalocele
Gastroschisis
Syndromes
Trisomy 13, 18, 21
Larsen’s
Cerebrocostomandibular
Jarcho-Levin
Roberts
Lethal multiple pterygium
Adapted from Churg AM, Myers JL, Tazelaar H, Wright J, editors: Thurlbeck’s pathology of the lung, ed 3, New York, 2005, Thieme.
643
of pulmonary hypoplasia is 80% and at 19 weeks it is 50%,
whereas after 26 weeks it is near zero (Rothschild et al,
1990). Other conditions associated with pulmonary hypoplasia are covered in other chapters.
PATHOLOGY
The easiest method of defining pulmonary hypoplasia
during postmortem exam is to calculate the ratio of lung
weight to body weight. At 28 weeks’ gestation, a ratio of
0.015 is at the 5th percentile, whereas at 35 weeks’, the
5th percentile is at a ratio of 0.012. The lung-to-body
weight ratio may be artificially elevated if the lungs are
wetter than usual from edema, hemorrhage, inflammation,
or lymphangiectasia—this may lead to the false conclusion that the patient does not have pulmonary hypoplasia.
Conversely, the ratio may be artificially low if the body is
heavier than usual because of renal cystic disease, hydrops,
ascites, tumors, hydrocephaly, and so forth. Therefore,
a better method for postmortem diagnosis of pulmonary
hypoplasia is to measure the lung volume by inflating the
lung with fixative at physiologic pressure and then measuring the displacement of fluid when the lung is immersed.
Pulmonary hypoplasia assessed by this method is defined
as lung volume less than the 5th percentile of standards
for each gestational age (Churg et al, 2005). This method
facilitates comparison of postmortem to in utero estimates
of lung volume made during antenatal imaging. However,
low lung volume does not necessarily correlate with deficiency of lung structure and function. A postmortem technique more physiologically relevant than lung volume is
the radial alveolar count, which is proportional to alveolar surface complexity (Churg et al, 2005) and thus gas
exchange surface area; however, this procedure is complex
and time-consuming.
Impairment of lung development before 16 weeks causes
reduced airway branching, reduced cartilage development,
reduced acinar complexity and maturation, delayed vascularization, and delayed thinning of the air-blood barrier.
Impairment after 16 weeks typically causes reduced acinar
complexity and maturation. These outcomes are predictable, given the time in gestation when these structures are
developing (Figure 46-12, A).
Because the growth of lung blood vessels parallels
the development of the airways, pulmonary hypoplasia
causes decreased total size of the pulmonary vascular bed,
decreased number of vessels per unit of lung tissue, and
increased pulmonary arterial smooth muscle. This last
phenomenon accounts for persistent pulmonary hypertension after birth.
CLINICAL SIGNS
The preterm newborn with pulmonary hypoplasia often
presents with immediate signs of respiratory distress and
cyanosis indistinguishable from those in the newborn with
severe respiratory distress syndrome (see earlier section
on clinical signs of RDS). However, respiratory failure
from severe pulmonary hypoplasia often becomes apparent within minutes of birth, whereas respiratory failure
from RDS usually progresses over the first few hours
after birth. The thorax may appear small or bell-shaped
644
PART X
Respiratory System
and, if oligohydramnios was severe, there may be flattening of the face and deformation (such as contractures of
the extremities). Hypercarbia may be severe on the earliest blood gas measurement, despite aggressive mechanical ventilation. The hypoxemia from surfactant deficiency
and lung immaturity may be compounded by right-to-left
shunting of deoxygenated blood due to pulmonary hypertension, leading to severe desaturation. If there is also a
tension pneumothorax, there may be asymmetry of breath
sounds or malposition of heart sounds, as well as impaired
cardiac output from impaired venous return to the thorax.
bronchograms, as with RDS, but the lung volumes may
be smaller and the diaphragms higher than in RDS. With
severe pulmonary hypoplasia, the thorax may appear bellshaped, and early pneumothorax is common.
TREATMENT
After birth, treatment for pulmonary hypoplasia is similar
to that provided to patients with RDS, including assisted
ventilation and exogenous surfactant. Even with cautious ventilation, however, tension pneumothoraces are
common and are often the proximate cause of death, so
the neonatal team should be prepared for urgent needle
decompression of the chest and insertion of a chest tube.
Permissive hypercapnia is appropriate, and high-frequency
ventilation may be necessary for adequate ventilation and
removal of very high arterial Pco2 levels. If pulmonary
RADIOGRAPHIC SIGNS
Because lung immaturity and surfactant deficiency accompany pulmonary hypoplasia, particularly in preterm
infants, the lungs may be radiographically dense with air
cell lineage
lung cell fate
Embryonic
Pseudoglandular
Canalicular
Saccular
Alveolar
lung bud
airway branching
capillarization
sac complexity
alveoli
Type I & Type II cells
surfactant production
tracheo-esophageal
septation
neuroendocrine, basal
ciliated & secretory cells
cartilage & smooth muscle
0.0
4.0
8.0
Human Weeks Gestation
16.0
26.0
0.0
9.0
12.0
Mouse Days Gestation
16.5
17.5
A
36.0
Birth
Birth
2.0
Postnatal-Years
5.0
Postnatal-Days
30.0
Total Lung Volume—Gestational
Age Ratio
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
B
Nonsurvivor
Survivor
C1
C2
FIGURE 46-12 A, Stages of lung development. B, Measurement of fetal lung volume by magnetic resonance imaging and outcomes after delivery.
C, Lung growth after occlusion of the fetal trachea for 6 days: (C1) before tracheal occlusion and (C2) after tracheal occlusion. (A from Kimura J,
Deutsch GH: Key mechanisms of early lung development, Pediatr Dev Pathol 10:335-347, 2007. Allen Press, Inc. C from Kohl T, Geipel A, Tchatcheva K, et
al: Life-saving effects of fetal tracheal occlusion on pulmonary hypoplasia from preterm premature rupture of membranes, Obstet Gynecol 113:480-483, 2009,
Lippincott Williams and Wilkins.)
CHAPTER 46 Respiratory Distress in the Preterm Infant
hypertension is present, as it commonly is, inhaled nitric
oxide for pulmonary vasodilation is logical, but clinical
trials with sufficient sample size to prove better survival
are lacking (Chock et al, 2009). Extracorporeal membrane
oxygenation may be appropriate for larger preterm infants
if the pulmonary hypoplasia, surfactant deficiency, and
pulmonary hypertension are expected to improve within a
few days. However, it is very difficult to select appropriate
candidates with pulmonary hypoplasia for extracorporeal
membrane oxygenation, and it is often unsuccessful with
severe disease or in combination with other major lifethreatening malformations.
PRENATAL DIAGNOSIS
An accurate prenatal test for pulmonary hypoplasia is
important because it may affect the obstetric management.
It would be particularly helpful to be able to discriminate
lethal from nonlethal pulmonary hypoplasia, especially
before 24 weeks’ gestation when termination of the pregnancy may be an option. This is an easier task than quantifying the degree of fetal pulmonary hypoplasia, which—as
noted previously—is challenging even for the pathologist.
A metaanalysis of clinical parameters for prediction of
fatal pulmonary hypoplasia following prelabor rupture
of membranes before 28 weeks’ gestation concluded that
gestational age at rupture was a better predictor of pulmonary hypoplasia than latency or oligohydramnios (van
Teeffelen et al, 2010).
In 1992, ultrasound measurement of the fetal chest circumference was shown to be helpful in predicting fatal
pulmonary hypoplasia (Ohlsson et al, 1992), whereas
in 2002 the best prediction was achieved by combining
clinical, biometric, and Doppler parameters (Laudy et al,
2002). These parameters included thoracic, cardiac, and
abdominal circumference, the largest vertical amniotic
fluid pocket, and pulsed Doppler measurements of the
arterial pulmonary branches. Nonsurvivors with pulmonary hypoplasia due to fetal urinary anomalies were noted
to have lower in utero lung volumes as determined by fetal
MRI, adjusted for gestational age (Zaretsky et al, 2005),
but there was no clear separation between the survivors
and nonsurvivors (Figure 46-12, B). Furthermore, there
was considerable overlap of the confidence intervals before
26 weeks’ gestation, which limits the usefulness of MRI
assessment of fetal volume for prenatal counseling. Threedimensional ultrasound measurements of lung volume
had better diagnostic accuracy for predicting pulmonary
hypoplasia, compared to two-dimensional measurements
of thoracic/heart area ratio (Gerards et al, 2008). Although
there will continue to be advances in fetal imaging to assess
lung volume, it will likely remain difficult to differentiate
lethal from nonlethal pulmonary hypoplasia.
The management of the preterm pregnancy with premature rupture of membranes near the limit of fetal viability is beyond the scope of this chapter. However, a recent
review article by Waters and Mercer (2009) offers a management algorithm that illustrates the areas of controversy.
For the patient who desires conservative management of
premature rupture before the limit of viability, the algorithm recommends serial assessments for signs of infection or labor, as well as interval ultrasound examinations to
645
watch for the development of pulmonary hypoplasia. If this
occurs, antenatal antibiotics and corticosteroids should be
considered if more aggressive management is desired.
PRENATAL TREATMENT
Treatment of oligohydramnios with amnioinfusion with
saline has been proposed to improve fetal survival by
increasing the latency period (the interval between PROM
and delivery) and thus the gestational age at delivery.
In a small study, it appeared to reduce the incidence of
pulmonary hypertension from about 50% to 10% with
PPROM in the 20th week of gestation (De Carolis et al,
2004). However, the procedure restores adequate amniotic fluid volume in only a minority of patients in which
it is performed, and there are procedure-related complications such as chorioamnionitis and placental abruption
(Tan et al, 2003). When it is successful, the benefit may
come from restoration of back-pressure from the amniotic
sac fluid to the lungs, stimulating fetal lung growth and
development during the critical canalicular stage between
16 and 26 weeks’ gestation.
Based on this same logic, there have been efforts to
reverse pulmonary hypoplasia in utero by fetal tracheal
occlusion. In a case report of PPROM at 16 weeks’ gestation, fetoscopic tracheal balloon occlusion was performed
at 27 6⁄7 weeks (Kohl et al, 2009). Within 6 days, the fetal
lung volume as measured by MRI increased from 13 to
70 mL (Figure 46-12, C) and the lung blood flow normalized. The fetus was delivered at 28 6⁄7 weeks using the EXIT
procedure (ex utero–intrapartum treatment), had no signs
of pulmonary hypertension, and was discharged home at 8
weeks of age. This approach has been used for fetuses with
congenital diaphragmatic hernia (Done et al, 2008), but
with mixed results. Whether this technique proves to be
valuable for PPROM is uncertain, but the restoration of
lung size in only 6 days suggests that pulmonary hypoplasia may be more reversible than previously assumed.
PNEUMONIA
INCIDENCE
The incidence of pneumonia/sepsis in preterm infants
with birthweight 1500 to 2500 g is only 0.28%, whereas in
patients with birthweight <1000 g the incidence is severalfold higher at 1.9% (see Figure 46-1) (Rubaltelli et al, 1998).
If only newborns with respiratory distress are considered,
the overall incidence (in mostly term infants) is 5% (see
Table 46-1)—the third most likely cause after RDS (46%)
and TTNB (37%). The data in Figure 46-1 indicate that
the incidence of pneumonia in newborns with respiratory
distress who are <1000 g, 1000 to 1500 g, and 1500 to 2500 g
is 4%, 2%, and 1%, respectively. The NICHD Neonatal
Research Network reported an incidence of blood culture–
proven early-onset sepsis (<72 hours after birth) in newborns of birthweight <1500 g of 1.9% during 1991–1993
(Stoll et al, 1996) and 1.5% during 1998–2000 (Stoll et al,
2003). In the earlier period, group B streptococcus was
the most frequent pathogen (31%) followed by Escherichia
coli (16%) and Haemophilus influenzae (12%). In the more
recent period, the most common pathogens were E. coli
646
PART X
Respiratory System
(44%), group B streptococcus (11%), coagulase-negative
staphylococcus (11%), viridans streptococci and other
streptococci (8%), H. influenzae (8%), Citrobacter (2%),
Listeria monocytogenes (2%), and Candida albicans (2%).
CLINICAL SIGNS
The clinical signs of pneumonia after preterm birth are
often indistinguishable from the more common problem
of RDS. Bacterial pneumonia is usually accompanied by
sepsis because newborns are frequently unable to confine
bacteria to the lung, and therefore some infants will exhibit
clinical signs of sepsis or shock, including poor perfusion
and hypotension, in addition to respiratory failure. Many
premature infants with pneumonia also have surfactant
deficiency from RDS, further obscuring the diagnosis of
pneumonia.
LABORATORY AND RADIOGRAPHIC SIGNS
Blood cultures will be positive in some premature newborns with pneumonia, but the presence of maternal antibiotics in the blood of the newborn reduces confidence
in a negative result. Leukopenia, increased percentage of
immature granulocytes, and elevated inflammatory markers such as C-reactive protein increase the likelihood of
sepsis/pneumonia, but with poor positive predictive value.
Tracheal aspirate culture (but not Gram stain) obtained
immediately after placement of an endotracheal tube may
help with diagnosis and guide therapy, especially when the
blood culture is negative (Booth et al, 2009).
Premature newborns with pneumonia will have pulmonary infiltrates on chest radiograph, but the radiographic
appearance is difficult to distinguish from RDS (although
classically in term newborns it has a more coarse and wet
appearance). Because newborns are unable to localize pulmonary infection, lobar infiltrates are rarely an indication
of pneumonia—plugging of airways with secretions is
more likely.
TREATMENT
Antibiotics directed at the most common organisms (see
earlier discussion) should be started immediately when
pneumonia in the preterm infant is suspected. Ampicillin
and gentamicin are reasonable choices, to be administered
for 48 hours pending culture results. Empiric vancomycin
has the disadvantage of promoting the emergence of vancomycin-resistant organisms, and a delay in treating coagulase-negative staphylococcal bacteremia until cultures
are positive is rarely of consequence to the patient. If the
blood culture is negative, and the mother has been pretreated with antibiotics, a longer course of antibiotics (e.g.,
5 to 7 days) may be prudent, especially if there are laboratory abnormalities such as elevated C-reactive protein and
a clinical course suggestive of sepsis or pneumonia.
Because it is so difficult to tell which preterm infants
with respiratory distress have pneumonia and which have
the more common problems of RDS or TTNB, one
approach is to empirically treat with antibiotics all premature newborns with respiratory symptoms. The likelihood
of infection in newborns <1000 g with respiratory distress
is about 4% (see Incidence, earlier), and so 24 extremely
premature newborns will be needlessly treated for every
one who will benefit. This number can be reduced by
avoiding empiric antibiotics at birth for newborns who
are prematurely delivered for maternal indications, such as
hypertension. Both PROM and premature labor increase
the risk of infection.
Among the possible adverse consequences of unnecessary empiric antibiotics are interference with the colonization of the intestinal tract with nonpathogenic bacteria,
selection of antibiotic-resistant bacteria, and fungal infection. On the other hand, the development of bronchopulmonary dysplasia is associated with inflammation from
chorioamnionitis (Speer, 2009), and so antibiotics even in
the absence of frank pneumonia may be beneficial.
SUGGESTED READINGS
Engle WA: American Academy of Pediatrics Committee of Fetus and Newborn:
Surfactant-replacement therapy for respiratory distress in the preterm and term
neonate, Pediatrics 121:419-432, 2008.
Halliday HL: Surfactants: past present and future, J Perinatol 28:S47-S56, 2008.
Lampland AL, Plumm B, Meyers PA, et al: Observational study of humidified highflow nasal cannula compared with nasal continuous positive airway pressure,
J Pediatr 154:177-182, 2009.
Rojas MA, Lozano JM, Rojas MX, et al: Colombian Neonatal Research Network:
Very early surfactant without mandatory ventilation in premature infants
treated with early continuous positive airway pressure: a randomized, controlled trial, Pediatrics 123:137-142, 2009:2009.
Seger N, Soll R: Animal derived surfactant extract for treatment of respiratory
distress syndrome, Cochrane Database Syst Rev 2:CD007836, 2009.
Sinha S, Moya F, Donn SM: Surfactant for respiratory distress syndrome: are there
important clinical differences among preparations, Curr Opin Pediatr 19:150154, 2007.
Soll R, Ozek E: Multiple versus single doses of exogenous surfactant for the prevention and treatment of neonatal respiratory distress syndrome, Cochrane Database
Syst Rev 1:CD000141, 2009.
Sweet DG, Halliday HL: The use of surfactants in 2009, Arch Dis Child Educ Pract
Ed 94:78-83, 2009.
Waters TP, Mercer BM: The management of preterm premature rupture of the
membranes near the limit of fetal viability, Am J Obstet Gynecol 201:230-240,
2009.
Complete references used in this text can be found online at www.expertconsult.com
C H A P T E R
47
Respiratory Failure in the Term Newborn
Thomas A. Parker and John P. Kinsella
The evaluation and management of respiratory failure in
the term newborn poses unique challenges and remains
one of the most vexing problems facing clinicians in the
newborn intensive care unit. Although some of the pathophysiologic features of respiratory failure in the term infant
are similar to the premature newborn condition, several
disorders occur more commonly in the term newborn
(e.g., meconium aspiration) and are often made more difficult to evaluate and manage because of cardiac and pulmonary vascular abnormalities that complicate the clinical
course. Indeed, the traditional perspective of categorizing
hypoxemia and respiratory failure in the term newborn as
cardiac, pulmonary vascular, or due to air-space (lung) disease is insufficient. For example, the syndrome of persistent pulmonary hypertension of the newborn (PPHN) is
defined by severe pulmonary vasoconstriction leading to
suprasystemic pulmonary artery pressure with extrapulmonary right-to-left venoarterial admixture across the fetal
channels of the oval foramen and the arterial duct. However, PPHN rarely occurs without concomitant parenchymal lung disease and disturbances in cardiac performance.
In this chapter we present an algorithm for evaluation
of the term newborn with hypoxemia and respiratory failure, review the syndrome of PPHN, and discuss common
causes of respiratory failure in the term newborn.
EVALUATION OF THE TERM
NEWBORN WITH HYPOXEMIA/
RESPIRATORY DISTRESS
One of the most anxiety-provoking experiences for many
clinicians (particularly those in training) is the initial evaluation and management of a term newborn with hypoxemia/respiratory distress. Traditional textbooks provided
a wealth of information about individual conditions once
identified. However, there are few sources designed to
guide the clinician in an ordered fashion through a comprehensive diagnostic evaluation. In this section, we propose
an approach to the evaluation of the hypoxemic newborn
that may be useful in clarifying the etiology of hypoxemia/respiratory distress and in determining the proper
sequence of diagnostic and therapeutic interventions.
HISTORY
Marked hypoxemia in the newborn can be caused by
parenchymal lung disease with V/Q mismatch or intrapulmonary shunting, pulmonary vascular disease causing
extrapulmonary right-to-left shunting (PPHN), or anatomic right-to-left shunting associated with congenital
heart disease. Evaluation should begin with the history and
assessment of risk factors for hypoxemic respiratory failure. Relevant history may include the results of prenatal
ultrasound studies. Lesions such as congenital diaphragmatic hernia (CDH) and congenital cystic adenomatoid
malformation are diagnosed prenatally with increasing
frequency. Although many anatomic congenital heart
defects can be diagnosed prenatally, vascular abnormalities
(e.g., coarctation of the aorta, total anomalous pulmonary
venous return) are more difficult to diagnose with prenatal
ultrasound. A history of a structurally normal heart by fetal
ultrasonography should be confirmed by echocardiography in the newborn with cyanosis (see later).
Other historical information that may be important in
the evaluation of the cyanotic newborn includes a history
of severe and prolonged oligohydramnios causing pulmonary hypoplasia. Also important is a history of prolonged
fetal bradyarrhythmia and/or tachyarrhythmia and marked
anemia (caused by hemolysis, twin-twin transfusion, or
chronic hemorrhage) that may cause congestive heart failure, pulmonary edema, and respiratory distress. Maternal
illness (e.g., diabetes mellitus), medication use (e.g., aspirin
or medications containing nonsteroidal antiinflammatory
drugs causing premature constriction of the ductus arteriosus, association of Ebstein’s malformation with maternal
lithium use), and illicit drug use may contribute to acute
cardiopulmonary distress in the newborn. Risk factors for
infection that cause sepsis/pneumonia should be considered, including premature or prolonged rupture of membranes, fetal tachycardia, maternal leukocytosis, uterine
tenderness, and other signs of intraamniotic infection.
Events at delivery may provide clues to the etiology of
hypoxemic respiratory failure in the newborn. For example, if positive-pressure ventilation is required in the delivery room, the risk of pneumothorax increases. A history
of meconium-stained amniotic fluid, particularly if meconium is present below the cords, is the sine qua non of
meconium aspiration syndrome. Birth trauma (e.g., clavicular fracture, phrenic nerve injury) or acute fetomaternal
or fetoplacental hemorrhage may cause respiratory distress
in the newborn (Box 47-1).
PHYSICAL EXAMINATION
The initial physical examination provides important clues
to the etiology of cyanosis. Marked respiratory distress
in the newborn (retractions, grunting, nasal flaring) suggests the presence of pulmonary parenchymal disease with
decreased lung compliance. However, it is important to
recognize that upper airway obstruction (e.g., Pierre Robin
sequence or choanal atresia) and metabolic acidemia also
can cause severe respiratory distress. In contrast, the newborn with cyanosis alone or cyanosis plus tachypnea (i.e.,
nondistressed tachypnea) typically has cyanotic congenital
heart disease, most commonly transposition of the great
vessels (TGV) or idiopathic PPHN.
647
648
PART X
Respiratory System
BOX 47-1 N
eonatal Respiratory Failure:
History and Risk Factor
Assessment
PRENATAL
Prenatal ultrasound study results
History of oligohydramnios and duration
History of fetal brady/tachyarrhythmia
Maternal illnesses, drugs , medications
History of fetal distress
Risk factors for infection
DELIVERY
History of positive pressure ventilation in DR
Meconium stained amniotic fluid
Hemorrhage
Birth trauma
Low Apgar score
The presence of a heart murmur in the first hours of life
is an important sign in the newborn with cyanosis or respiratory distress. In this setting, it is unusual for the common left-to-right shunt lesions (patent ductus arteriosus,
atrial septal defect, ventricular septal defect) to produce
an audible murmur because pulmonary vascular resistance
(PVR) remains high and little turbulence is created across
the defect. A murmur that sounds like a ventricular septal
defect in the first hours of life is most commonly caused
by tricuspid regurgitation (associated with PPHN or an
ischemic myocardium).
The response to supplemental oxygen can also provide
important clues to the pathophysiology of hypoxemic respiratory failure in the term newborn (Boxes 47-2 and 47-3).
BOX 47-2 Physical Examination
RESPIRATORY DISTRESS (RETRACTIONS, GRUNTING, NASAL
FLARING)
Suggests lung parenchymal disease (compliance), upper airway disease or
metabolic acidemia
NO SIGNIFICANT RESPIRATORY DISTRESS (TACHYPNEA ALONE)
Suggests hypoxemia caused by cyanotic heart disease without lung disease
BOX 47-3 A
cute Response to Supplemental
Oxygen (High Fio2 by Hood, Mask)
MINIMAL OR TRANSIENT CHANGE IN SA o2
Cyanotic heart disease, PPHN
MARKED IMPROVEMENT IN SA o2
Parenchymal lung disease, CHD with ductal-dependent systemic blood flow
TABLE 47-1 Role of Pulse Oximetry in Evaluation of Neonatal
Hypoxemic Respiratory Failure
Preductal Sao2 =
postductal Sao2
1. Intrapulmonary shunt: PVR < SVR
2. Cyanotic congenital heart disease with
L→R PDA:
Ductal-dependent Qp: pulmonary
atresia/stenosis, tricuspid atresia,
Ebstein’s anomaly
3. PPHN:R→L shunt at PFO: PVR >
SVR, ductus closed
Preductal Sao2 >
postductal Sao2
1. PVR > SVR with R→L PDA: PPHN:
MAS, RDS, CDH
2. Ductal-dependent Qs: HLHS, IAA,
coarctation
3. Anatomic PV disease: alveolarcapillary dysplasia