Ped Core
Ped Core
Ped Core
SECTION EDITORS
Jane Balint, MD
Director, Intestinal Support Service
Pediatric Gastroenterology, Hepatology and Nutrition
Nationwide Childrens Hospital
Columbus, OH
Elizabeth Bobo, MS, RD, LDN, CNSD
Clinical Dietitian, Gastroenterology and Nutrition
Nemours Childrens Clinic
Jacksonville, FL
Steve Plogsted, PharmD, BCNSP
Clinical Pharmacy Specialist
Nutrition Support Pharmacist
Nationwide Childrens Hospital
Columbus, OH
Jane Anne Yaworski, MSN, RN
Clinical Nurse Specialist/Nutrition Support Service/Intestinal Care Center
Childrens Hospital of Pittsburgh
Pittsburgh, PA
MANAGING EDITOR
Nina D. Seebeck
The American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) is a scientific society whose members are health care professionalsphysicians, dietitians, nurses, pharmacists, other allied health professionals, and researchersdedicated to assuring that every patient receives optimal
nutrition care.
A.S.P.E.N.s mission is to serve as a preeminent, interdisciplinary, nutrition society dedicated to patient-centered clinical practice worldwide through
advocacy, education, and research in specialized nutrition support.
NOTE: This publication is designed to provide accurate authoritative information in regard to the subject matter covered. It is sold with the understanding that the publisher is not engaged in rendering medical or other professional advice. Trademarked commercial product names are used only
for education purposes and do not constitute endorsement by A.S.P.E.N.
This publication does not constitute medical or professional advice, and should not be taken as such. To the extent the information published herein
may be used to assist in the care of patients, this is the result of the sole professional judgment of the attending health professional whose judgment is
the primary component of quality medical care. The information presented herein is not a substitute for the exercise of such judgment by the health
professional.
All rights reserved. No part of this may be used or reproduced in any manner whatsoever without written permission from A.S.P.E.N. For information write: American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.), 8630 Fenton Street, Suite 412, Silver Spring, MD 20910-3805; (301)
587-6315, www.nutritioncare.org, email: [email protected].
Contents
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Reviewers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
Foreword . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xv
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xvii
Acknowledgments & Dedication. . . . . . . . . . . . . . . . . . . . . . . xix
PART I
INTRODUCTORY AND BASIC PHYSIOLOGY
1. Mechanics of Nutrient Intake. . . . . . . . . . . . . . . . . . . . . . . 3
Mark R. Corkins, MD, CNSP, SPR, FAAP
Carol G. McKown, DDS, MS
Anna M. Dusick, MD
8. Fat-Soluble Vitamins . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
Winston Koo, MBBS, FACN, CNS
May Saba, PharmD, BCNSP
Mirjana Lulic-Botica, BSc, BCPS
Judith Christie, RN, MSN
PART II
AGE-SPECIFIC NUTRITION FOR GROWTH
ANDDEVELOPMENT
Jane P. Balint, MD
Seema Mehta, MD
Robert J. Shulman, MD
4. Fats. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Peggy R. Borum, PhD
6. Minerals. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Winston Koo, MBBS, FACN, CNS
Mirjana Lulic-Botica, BSc, BCPS
May Saba, PharmD, BCNSP
Letitia Warren, RD, CSP
iii
iv
CONTENTS
PART III
DISEASE STATES AND NUTRITION
18. Developmental Delay . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Kathleen J. Motil, MD, PhD
PART IV
NUTRITION CARE OF THE PEDIATRIC PATIENT
33.Assessment of Nutrition Status by Age
and Determining Nutrient Needs. . . . . . . . . . . . . . . . . 409
Liesje Nieman Carney, RD, CNSD, LDN
Jennifer Blair, MA, RD, CSP, LDN
Contributors
Brian S. Carter, MD
Professor of Pediatrics
Vanderbilt University Medical Center
Nashville, TN
Jane P. Balint, MD
Director, Intestinal Support Service
Pediatric Gastroenterology, Hepatology and Nutrition
Nationwide Childrens Hospital
Columbus, OH
vi
CONTRIBUTORS
CONTRIBUTORS
John A. Kerner, MD
Pediatric Gastroenterology, Hepatology and Nutrition
Stanford University
Palo Alto, CA
Samuel A. Kocoshis, MD
Professor of Pediatrics
University of Cincinnati College of Medicine
Director, Nutrition and Small Bowel Transplantation
Cincinnati Childrens Hospital Medical Center
Cincinnati, OH
vii
viii
CONTRIBUTORS
Susan R. Rheingold, MD
Assistant Professor of Pediatrics
Division of Oncology
The Childrens Hospital of Philadelphia
Philadelphia, PA
Robert Murray, MD
Nationwide Childrens Hospital
Center for Healthy Weight and Nutrition
Columbus, OH
CONTRIBUTORS
ix
Reviewers
xi
xii
REVIEWERS
REVIEWERS
xiii
xiv
REVIEWERS
Foreword
Preface
xvii
xix
PART I
INTRODUCTORY AND
BASIC PHYSIOLOGY
Mark R. Corkins, MD, CNSP, SPR, FAAP, Carol G. McKown, DDS, MS, and Anna M. Dusick, MD
Learning Objectives
CONTENTS
Background. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Appetite . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Mastication/Dentition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Feeding Development. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
3
3
4
5
Swallowing
Feeding Skills in the First Year
Feeding Skills in the Second Year
Dysphagia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Feeding Teams . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Background
Appetite
eat some foods just because they taste good. The reward of
taste stimulates feeding even in the absence of a true energy
deficit.4
There are critical periods of infant development that
hinge upon exposures to new tastes and textures. Genetic
input affects the sense of taste itself, including the strength
of response to sweet, salty, bitter, and sour. 5 Studies show
that breastfed infants have greater willingness to try
new tastes2,6; this is believed to be from their exposure
to various flavors in breast milk. Early exposure to tastes
determines taste and food preferences before the child
develops neophobia. 2 Neophobia is the developmental stage
between 18 months to 2 years of age when children resist
trying new foods. 2,5 The more a child is exposed to a taste,
the more likely it is to be accepted as a preferred taste. 5,6
Studies indicate that it may take 5 to 10 exposures before
a food becomes an accepted taste. 6 Also, there appears to
be a critical period for beginning solid foods, due to their
texture, which is before 10 months of age. Northstone et al
reported on long-term feeding problems related to acceptance of the taste and texture of foods.7 Feeding difficulties
in children identified by 9,360 parents were found to occur
more often in those children whose parents delayed initiating solid foods until after 10 months of age.7
Biological preferences are also altered by modeling
that children see when observing their parents and peers.2,5
Eating with others influences the feeding behaviors and the
food preferences that children ultimately develop.6
Appetite control is centered in the hypothalamus of the
brain and integrates information from multiple sources.4,8
The general level of appetite is influenced by the amount
of the hormone, leptin, produced by the bodys adipose
tissue.4,9 The liver also sends appetite-influencing signals
to the brain, primarily through the energy products fat and
glucose and the level of adenosine triphosphate (ATP) in
the liver cells.8 The insulin level produced by the pancreas
in response to serum glucose levels influences the general
appetite level as well.4,8 Leptin and insulin suppress inhibition, or drive the appetite, via the hypothalamus.10
The first stage in appetite is the cephalic phase, which
is a biological response to feeding cues. This concept was
first presented by Ivan Pavlov. Pavlov demonstrated that
he could condition dogs to salivate at the ringing of a bell
by developing an association with feedings.11 A variety of
studies have shown that visual, olfactory, gustatory, tactile,
and auditory inputs stimulate processes in the salivary
glands, gastrointestinal (GI) tract, pancreas, and cardiovascular and renal systems.12 These responses are quick
and prepare the body for the digestion and absorption of
2010 A.S.P.E.N. www.nutritioncare.org
Mastication/Dentition
Age of Eruption
610 mo
812 mo
913 mo
1016 mo
1319 mo
1418 mo
1622 mo
1723 mo
23 y
Age of Eruption
67 y
67 y
67 y
78 y
78 y
89 y
910 y
1011 y
1012 y
1012 y
1112 y
1112 y
Feeding Development
Swallowing
The process of swallowing is divided into 3 distinct
physiological phases. These phases are the oral phase, the
pharyngeal phase, and the esophageal phase. Each phase
depends on the correct anatomy and neurophysiology of
the muscles for feeding and appropriate ventilation.
The oral phase consists of the preparatory stage and the
movement of food being propelled to the back of the mouth.
Infants take most liquids into the mouth already on the
back third of the tongue; this changes as they develop more
skill in the front of the mouth (tongue, jaw, and lips), and
develop teeth. Purees and solids are propelled to the back of
the mouth by the tongue. (Later in infant development the
lips are used for assisting with eating during the oral phase.
Skills such as taking purees off a spoon or drinking through
a straw will occur in the last half of the first year.)
The pharyngeal phase consists of the liquid or food
coming to the back of the mouth, the pharynx, and reflexively being swallowed. The soft palate and uvula lift so that
liquid or food will not pass into the nasopharynx. The larynx
closes by muscle contractions and the downward movement
of the epiglottis. Respiration ceases briefly (deglutition
apnea) and, after the food has started its descent, expiration
then inspiration will occur. In the infant, the swallow can
occur at any part of the respiratory cycle and it is variable.15
The upper esophageal sphincter relaxes so that the food
can enter the esophagus in the beginning of the esophageal
phase. The cricopharyngeus muscle relaxes at the upper
end of the esophagus and food travels to the stomach by
peristalsis. Successful swallowing depends on all of these
structures, muscles, and neurological and respiratory
systems working in coordinated millisecond timing.16,17
Anatomical or physiological abnormalities with these
systems will affect feeding effectiveness and efficiency.
suck patterns. 30 Some infants, who had significant limitations in oral feeding attempts due to a high level of illness
or respiratory disorders, can display aversion to attempts at
oral feeding. In others, a persistent rapid breathing rate can
interfere with establishing an efficient feeding rhythm.
Evaluation of the infant who is not feeding appropriately can be performed by a physician, nurse, occupational
therapist, or speech therapist trained in, and experienced
with, infant feeding. The evaluation will include a medical
history, developmental history, and a neurological examination as well as an oromotor assessment including feeding or
feeding attempt. A multidisciplinary approach is required.
Pediatric specialists in gastroenterology, neurology, rehabilitation, development, and others may be needed. The
evaluation, under the direction of a physician, may include
radiological testing such as a feeding study, esophagram, or
studies for reflux or gastric emptying. Neurological studies
of the central nervous system may be needed for the diagnosis of neurodevelopmental abnormalities. Studies of
respiratory function and sufficiency of ventilation may be
needed to provide optimal respiratory support for feeding.
It is important to determine the safety of feeding and
ensure that the infant is not aspirating as a result of the
underlyingproblems. 31
Sometimes, despite extensive evaluations, the etiology
of the feeding difficulties is never discovered. There is also a
belief that in some situations the initiating organic cause has
resolved but the resulting behavior has become established.
Once the cause and extent of poor feeding is understood,
a treatment plan can be undertaken to work on oral feedings, and/or to provide supplemental feedings for the safety
and growth of the infant. Supplemental feedings can consist
of nasogastric feedings or feedings into the jejunum. Such
treatment plans should be drafted with the parents and a
team of medical and therapy providers.
Dysphagia
Feeding Teams
a child psychologist;
a speech and/or occupational therapist;
a dietitian; and
a social worker.
For the feeding team to be successful, the parents must
clearly identify their goals and be willing to make changes,
and to work with the team members. Each professional
must also be willing to work with team members and to
support the parents and patient during the process. Setting
clear goals with the team and reviewing those goals is an
important part of a feeding team program.
For example, Abby is a young toddler who has
aversiveness to eating because of severe respiratory problems caused by prematurity. Her respiratory status has
significantly improved and she needs only occasional bronchodilators. She is gastrostomy fed and is very sensitive to
the rate of feedings. Her mother has cut back on her feedings so Abby is also failing to thrive. There are many goals
for the team members, and progress may vary in these goals.
The family hopes to shorten feeding times and increase
Abbys oral feedings so she can eventually stop gastrostomy
feedings. These are appropriate goals for the child and the
team members; however, these are long-term goals and the
progress is incremental and variable. The parents, with the
teams help, determine the daily schedule for feeding, and
continue in the routine of feedings. The physician and dietitian may work on a short-term goal of improving Abbys
weight gain while trying to shorten her feeding times. The
therapists may start by improving her ability to stay at the
table at mealtime and touch food for short time periods.
The dietitian and therapists may use a chaining technique
to select foods that are similar to foods Abby likes so as to
slowly broaden her food choices. At each follow-up visit
with the team, the goals are reassessed and the plan of care is
revised based on the childs progress. 39 Referral to a feeding
team is generally made by the childs primary care provider
and reviewed by the coordinator for appropriateness to the
team.
References
10
12. Mattes RD. Physiologic responses to sensory stimulation by food: nutritional implications. J Am Diet Assoc.
1997;97(4):406413.
13. McDonald RE, Avery DR, Dean JA. Dentistry for the Child and
Adolescent. 8th ed. St. Louis, MO: Mosby; 2004:176178.
14. Kronfeld R, Schour I. Chronology of the human dentition. J
Am Dent Assoc. 1939;26:1832.
15. Kelly BN, Huckabee ML, Jones RD, Frampton CMA. The
first year of human life: Coordinating respiration and nutritive swallowing. Dysphagia. 2007; 22(1):3743.
16. Bosma JF. Development of feeding. Clin Nutr. 1986;5(5):
210218.
17. Derkay SD, Schechter GL. Anatomy and physiology of
pediatric swallowing disorders. Otolaryngol Clin North Am.
1998;31(3):397404.
18. Weber F, Wollridge MW, Baum JD. An ultrasonographic study
of the organisation of sucking and swallowing by newborn
infants. Dev Med Child Neurol. 1986;28(1)1924.
19. Tamura Y, Horikawa Y, Yoshida S. Co-ordination of tongue
movements and peri-oral muscle activities during nutritive
sucking. Dev Med Child Neurol. 1996;38(6):503510.
20. Wolff PH. The serial organization of sucking in the young
infant. Pediatrics. 1968;42(6):943956.
21. Qureshi MA, Vice FL, Taciak VL, Bosma JF, Gewolb IH.
Changes in rhythmic suckle feeding patterns in term infants
in the first month of life. Dev Med Child Neurol. 2002;44(1):
3439.
22. McGowan JS, Marsh RR, Fowler SM, Levy SE, Stallings VA.
Developmental patterns of normal nutritive sucking in infants.
Dev Med Child Neurol. 1991;33(10):891897.
23. Ayano R, Tamura F, Ohtsuka Y, Mukai Y. The development
of normal feeding and swallowing: Showa University study of
the feeding function. Int J Orofacial Myology. 2000;26:2432.
24. Gesell A, Ilg FL. Feeding Behavior in Infants. A Pediatric
Approach to the Mental Hygiene of Early Life. Philadelphia, PA:
J.B. Lippincott Co; 1937.
25. Gisel EG. Effect of food texture on the development of chewing
of children between six months and two years of age. Dev Med
Child Neurol. 1991;33(1):6979.
26. Sheppard JJ, Mysak ED. Ontogeny of infantile oral refluxes
and emerging chewing. Child Dev. 1984;55(3):831843.
Jane P. Balint, MD
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Gastric Motility. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .12
Small Bowel Motility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Effect of Intake on Motor Activity. . . . . . . . . . . . . . . . . . . . 13
Consistency
Nutrient Content
Caloric Density
Osmolarity
Learning Objectives
Background
12
Gastric Motility
Gastric emptying can be affected by a variety of characteristics of the ingested material as well as the rapidity of
transit through the GI tract. With this, the time for gastric
emptying can vary from 1 to 4 hours. The effect of nutrient
composition on motor activity has been more clearly delineated for the stomach than the small bowel.
Consistency
Liquids will empty from the stomach more quickly than
solids. In turn, clear liquids will empty more rapidly than
full liquids, with non-nutritive liquids emptying the fastest,
leaving the stomach in about 20 minutes.11,20 The viscosity
of the feeds will also affect the speed of gastric emptying,
with lower viscosity material emptying more rapidly. Thus
both soluble and insoluble fibers mixed with the feedings
will slow emptying.21,22 In addition to delaying gastric
emptying, soluble fibers can prolong transit through the GI
tract by prolonging the duration of the fed pattern of small
bowel motor activity.22
Nutrient Content
The response to the type of nutrient ingested is complex.
There are both hormonal and neural responses to food that
regulate motor activity of the stomach and small intestine.
These are interrelated in that neural activity can affect
the release of some GI hormones. Once chyme starts to
pass through the pylorus, there is feedback via vagal afferents from receptors in the duodenum that regulate gastric
emptying. These receptors include those that detect fat,
amino acids, glucose, pH, and osmolarity.23 A component
of the response is mediated by release of gastrin, cholecystokinin, pancreatic polypeptide, glucagon-like peptide 1,
and peptide YY.4,24,25 It has been demonstrated that carbohydrates will empty from the stomach before proteins, with
fats emptying the most slowly.4,26,27 Fats reaching the distal
ileum inhibit both gastric emptying and small intestinal
motility, a mechanism known as the ileal brake.28 Complex
carbohydrates that reach the distal small intestine can
also stimulate the ileal brake.1 In addition to the actual
nutrient content, the pH will affect gastric emptying as well
with more acidic material emptying more slowly from the
stomach.29
13
Caloric Density
Increased calorie content of the ingested food or liquid will
result in slower emptying from the stomach.21,30 It has also
been demonstrated that a higher calorie content meal will
result in a longer fed pattern in the small bowel leading to a
longer transit time through the small intestine. 31
Osmolarity
Finally, solutions that have a higher osmolarity will empty
more slowly from the stomach than those that are lower in
osmolarity.27
While each of these factors individually can influence
gastric emptying in the older infant, child, and adult, the
same has not been found to be true in premature infants.
In one study of 25 to 30 weeks gestation infants, it was
demonstrated that altering osmolality, caloric density, or
the volume of the feeding separately did not change gastric
emptying. However, when the volume of the feeding was
increased in combination with decreasing the osmolality,
gastric emptying was more rapid. 32 Additionally, across age
groups in clinical practice, these factors can clearly overlap
as nutrient content is closely tied to caloric density. For
example, a meal or formula higher in fat will likely be higher
in calories.
14
Dumping
Very rapid gastric emptying, or dumping, is relatively
uncommon in pediatrics. 35 It can occur as a result of surgery,
particularly involving the pylorus, or with damage to the
vagal nerve such as during a fundoplication or cardiac
surgery. Symptoms include nausea, abdominal distention,
cramping, diarrhea, and vasomotor changes associated with
swings in glucose. Some management strategies are the opposite of those suggested for use in delayed emptying. One can
try to take advantage of normal physiology by increasing the
fat and protein content of the diet and increasing the fiber
content of the diet, including the addition of pectin and guar
gum.36 Rather than mixing liquids with the meal, liquids
should be taken separately from solids. On the other hand,
some of the same strategies recommended for treatment of
delayed gastric emptying may also be helpful in too rapid
emptying, specifically smaller more frequent meals as well
as continuous drip or post-pyloric feedings. Pharmacologic
therapeutic options are limited. Acarbose, an alpha-glucosidase inhibitor, has been used to treat dumping syndrome
as it delays the hydrolysis of carbohydrates with resultant
delayed absorption of glucose38,39 Octreotide, an analogue of
somatostatin, can be beneficial as one of its specific actions is
to delay gastric emptying.40,41
bowel. There are some data to indicate that amoxicillinclavulonic acid may promote small bowel motility.42 In
more severe cases, octreotide may be beneficial. This somatostatin analogue has a wide range of effects on the GI tract,
including induction of phase III MMC when given in small
doses subcutaneously.43
References
15
16
34. De Ville K, Knapp E, Al-Tawil Y, Berseth CL. Slow infusion feedings enhance duodenal motor responses and gastric emptying
in preterm infants. Am J Clin Nutr. 1998;68:103108.
35. Di Lorenzo C, Ciamarra P. Pediatric gastrointestinal motility.
In: Schuster MM, Crowell MD, Koch KL, eds. Schuster Atlas
of Gastrointestinal Motility in Health and Disease. 2nd ed.
Hamilton, Ontario: BC Decker, Inc; 2002:411428.
36. Karamanolis G, Tack J. Nutrition and motility disorders. Best
Pract Res Clin Gastroenterol. 2006;20:485505.
37. Karamanolis G, Tack J. Promotility medications now and
in the future. Dig Dis. 2006;24:297307.
38. Ng DD, Ferry RJ, Kelly A, et al. Acarbose treatment of
postprandial hypoglycemia in children after Nissen fundoplication. J Pediatr. 2001;139:877879.
39. Zung A, Zadik Z. Acarbose treatment of infant dumping
syndrome: extensive study of glucose dynamics and long-term
follow-up. J Pediatr Endocrinol Metab. 2003;16:905915.
Contents
Digestion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Lactose
Starch
Absorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Malabsorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Metabolism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Learning Objectives
Digestion
Lactose
Lactose is the primary carbohydrate present in human
milk and most infant formulas.2 Lactose is hydrolyzed by
the enzyme lactase into the monosaccharides glucose and
galactose (Figure 3-1).
17
18
Starch
Formula-fed infants are exposed to a variety of other carbohydrates in infancy including starches. The simplest form
of starch is amylose, a linear polymer of glucose molecules
linked by -1,4 glycosidic bonds. Amylopectin, a plant
starch, is the major form of carbohydrate in the diet. Structurally it is similar to amylose, but for every 20 to 30 glucose
units there are -1,6 branch points.11
Digestion of starch begins with intraluminal digestion
by salivary and then pancreatic amylases. Amylase hydrolyzes starch at the internal -1,4 bonds (Figure 3-1). It is not
active against those bonds located next to the -1,6 bonds or
those at the reducing end of the starch molecule. Amylase
cleaves amylose and amylopectin into maltotriose, maltose,
and -limit dextrans (Figure 3-1).12
Salivary amylase, secreted by the salivary glands, is
present in preterm infants. Amylase activity increases with
gestational age, rising rapidly after birth and approaching
adult values by 6 months to 1 year of age. Salivary amylase
is inactivated at pH < 4.5 In newborns, however, salivary
amylase tends to remain active because they have poorly
acidified stomachs.13 Pancreatic amylase, secreted by the
pancreas, is present at low levels in preterm and full-term
infants. Activity begins to increase at 4 to 6 months of age and
reaches adult values by 1 to 2 years of age (Figure 3-4).2
19
Further digestion of the disaccharides and oligosaccharides occurs at the level of enterocytes. Glucoamylase, like
lactase, is located in the brush border of the small intestine.
It cleaves -1,4 bonds of primarily non-branching glucose
polymers (ie, amylose) and non-reducing ends of polysaccharides (Figure 3-1). Short chains (ie, < 10 glucose units)
are more easily digested by glucoamylase than longer-chain
units.14
Glucoamylase activity is detectable by 20 weeks gestation. At the beginning of the third trimester, the activity
level is about half that at 36 to 38 weeks gestation. 5 In addition, it is also present in low levels in the colon only early
in fetal life. 5 Therefore, in newborn infants, glucoamylase
is the primary enzyme for complex carbohydrate digestion
since pancreatic amylase activity is low. Young infants and
older children have similar glucoamylase activity levels
which are approximately half those in adults.15
Sucrase-isomaltase is a disaccharidase also found in the
brush border of the small intestine. At 20 weeks gestation,
sucrase-isomaltase activity is almost half to three-quarters that of term newborns and adults. 5 Similar to lactase,
sucrase-isomaltase activity is highest in the proximal small
intestine. Activity remains high over the course of an individuals life. Sucrase-isomaltase is cleaved into sucrase and
isomaltase by pancreatic proteases. 5 Sucrase hydrolyzes
sucrose into the monosaccharides glucose and fructose
(Figure 3-1). Isomaltase cleaves the -1,6 glycosidic bonds
of amylopectin (Figure 3-1). 5 A genetic deficiency of
sucrase-isomaltase known as congenital sucrase-isomaltase
deficiency is an autosomal recessive disorder. All patients
with this deficiency lack sucrase and have varying degrees
of isomaltase activity.16
2010 A.S.P.E.N. www.nutritioncare.org
20
Absorption
Dietary carbohydrates are absorbed in the form of the monosaccharides glucose, galactose, and fructose. Active transport
of glucose and galactose is present by 10 weeks gestation. 5
The absorption rate of glucose increases through gestation
and continues to increase postnatally into adulthood. Galactose absorption rate reaches adult values between 4 to 8 years
of age.8
Access into enterocytes occurs via carrier molecules.
Glucose and galactose are actively transported via the
sodium-glucose linked transporter (SGLT1) located at the
brush border. Glucose and galactose entry is coupled to the
entry of sodium along its electrochemical gradient. This
electrochemical gradient is maintained via the sodium-potassium-adenosine triphosphatase (Na+-K+ ATPase) located
at the basolateral surface. SGLT1 has binding sites for both
glucose and sodium. Two sodium molecules are absorbed
for every glucose molecule. Once both sites are occupied, the
transporter translocates across the brush border membrane
and releases the glucose and sodium into the enterocyte.11
The sodium-linked transport of glucose provides the basis
for adding glucose or starches to oral rehydration solutions.
Passive absorption of glucose also can occur across the brush
border.8
Fructose transport across the brush border membrane
occurs passively down its concentration gradient. The
fructose transporter, GLUT5, is not sodium dependent.
Transport of glucose, galactose, and fructose across the
basolateral membrane also occurs by facilitated transport
via the sodium-independent transporter, GLUT2.11 Recently
GLUT2 has been recognized in the brush border membrane,
and glucose transported by SGLT1 promotes the activation
of GLUT2 already in the apical membrane and rapid insertion of GLUT2 from vesicles.17 This observation explains the
enhancement of fructose absorption in the proximal intestine
in the presence of equimolar amounts ofglucose.17
Malabsorption
Metabolism
References
21
Fats
Peggy R. Borum, PhD
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Different Types of Fat
Essential Fatty Acids
Different Functions of Adipose Tissue
Importance of Dietary Fat in Infants
Digestion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Absorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
Metabolism During Normal Physiology. . . . . . . . . . . . . . . . 26
Synthesis of Fatty Acids
Beta-Oxidation of Long-Chain Fatty Acids inMitochondria
Beta-Oxidation of Medium-Chain Fatty Acids inMitochondria
Steps of Beta-Oxidation
Beta-Oxidation of Very Long-Chain Fatty Acids inPeroxisomes
Omega-Oxidation of Fatty Acids
Learning Objectives
Introduction
FATS
23
24
2
and -6 fatty acids are required in the human diet and are
termed essential fatty acids (EFAs).1 The simplest -6 fatty
acid is linoleic acid (LA). Humans can convert LA to arachidonic acid (ARA). The simplest -3 fatty acid is -linolenic
acid (-LA). Humans can convert -LA to eicosapentaeonic
acid (EPA), and EPA very slowly to docosahexaenoic acid
(DHA). The same enzyme pool is used to metabolize LA and
-LA to their respective longer-chain metabolites. The more
prevalent fatty acid is metabolized preferentially. Thus a diet
high in the -6 fatty acid LA will preferentially metabolize
it over the -3 fatty acid -LA. ARA (20-carbon polyunsaturated -6) and EPA (20-carbon polyunsaturated -3)
are released from membranes and further metabolized to a
cascade of eicosanoids. Although it is not quite that simple,
eicosanoids from -6 fatty acids generally are considered
pro-inflammatory and those from -3 fatty acids are considered weakly anti-inflammatory. The docosanoid products
of DHA also have anti-inflammatory actions. 3 Fatty acids
and eicosanoids can change the abundance of transcription
factors which in turn regulate gene expression. Thus, the
fatty acid composition of the diet influences the composition of the cell membranes which influences the availability
of appropriate substrate to make either pro- or anti-inflammatory fatty acid products. Traditionally low intake of -3
and -6 fatty acids associated with an increase in an -9
fatty acid known as Mead acid in plasma has been used
to diagnose EFA deficiency. A more recent method uses
red blood cells and measures two additional parameters.4
The emerging field of investigation known as lipidomics
measures all small molecular weight lipids in a sample and
will likely provide the nutrition community with improved
methods for specific EFA evaluation.
With the industrialization of Western society, dietary
intake of EFAs has changed, reflecting the trend away from
grains to more processed foods high in fat. Intake of LA
relative to -LA has increased significantly. Currently the
ratio of LA to -LA in the diet is 14:1 in contrast to the 2:1
or 1:1 ratio that is usually recommended.2 Interestingly,
the change in dietary fat intake follows a similar temporal
pattern to the increase in inflammatory bowel disease incidence in the pediatric population. 5
FATS
the last trimester, and most of that is DHA. The brain also
grows rapidly during the last trimester and requires a large
amount of DHA for the synthesis of myelin. At birth, the
brain of the human is at about 50% of its adult size and
continues to grow rapidly until it reaches near adult size
at about 2 years of age. Thus, the extremely preterm infant
born at 24 or 25 weeks gestation requires a large amount of
EFAs, and -3 fatty acids in particular, to achieve the type
of growth usually experienced by a fetus in utero during the
same period postconception.6,7
Regulation of gene expression by nutrients is critically
important in preterm neonates. Regulation of gene expression by -3 fatty acids involves multiple complex processes
including regulation of the transcription factors sterol regulatory element-binding proteins (SREBPs) and peroxisome
proliferator-activated receptors (PPARs) that modulate
many critical steps in metabolism.8,9
The two most abundant long-chain PUFAs in the brain
are DHA and ARA. DHA is concentrated in the prefrontal
cortex and in some retinal cells.10 Inadequate intake of -3
fatty acids not only results in decreased brain DHA but also
increased brain -6 fatty acid concentration.11,12 Worldwide
there are a variety of enteral products that vary in their
supply of -3 fatty acids and in DHA in particular. However
none of them provide the amount of EFAs that would accumulate in the fetal brain during the last trimester of gestation
without additional supplementation. Thus, the current diet
provided to these infants appears to be deficient in DHA.
Although it is not known how these infants would develop
if fed to sufficiency, preterm infants fed increased amounts
of DHA have been reported to have improved visual acuity
and mental development.13,14 A recent multicenter prospective, randomized, double-blind placebo-controlled trial
in Italy compared 580 healthy term neonates receiving
20mg of liquid DHA to 580 healthy term infants receiving
placebo with a focus on developmental milestones. The
infants receiving DHA were able to sit without support
1week earlier, but there was no difference between groups
for hands-and-knees crawling, standing alone, and walking
alone.15 It is unclear how these data from healthy term
neonates relate to preterm neonates. In infants being fed
parenterally, the challenge to provide adequate DHA and
ARA is even greater with currently available lipid emulsions
approved for use in this population.
Digestion
Because dietary lipids are hydrophobic, they must be hydrolyzed and emulsified to very small micelles before they can
be absorbed by the intestine. Lingual and gastric lipases
25
26
Absorption
Bile acids play an important role in solubilizing the products of lumenal lipolysis and facilitating their transfer to
the absorptive epithelium. Fatty acids and monoglycerides
have enough solubility in the lumenal contents that they
can diffuse to the brush border and be absorbed.16
The intestinal epithelium is the site of significant
metabolic activities. The absorbed 2-monoglycerides are
re-acylated to triglycerides. Some 1-monoglycerides in the
emulsion are absorbed and hydrolyzed to fatty acids and
glycerol. The glycerol released within the intestinal epithelium is reutilized for triglyceride synthesis. Long-chain fatty
acids (LCFAs) are esterified to triglycerides, secreted into
the lymphatics as chylomicrons, and enter the blood via the
thoracic duct. Free short- and medium-chain fatty acids are
absorbed into the hepatic portal vein.16
FATS
27
Patients with high plasma levels of triglycerides are sometimes given heparin (assuming that it will stimulate lipolysis
of triglycerides) and carnitine (assuming it will stimulate
-oxidation of the released fatty acids). Definitive data to
confirm these assumptions in patients on special nutrition
support are yet to be obtained. In addition, it should be
remembered that supplemented heparin and carnitine each
have a variety of effects on the bodys metabolism.
Steps of Beta-Oxidation
28
in the body both during health and disease. Twelve children with pediatric short bowel syndrome who developed
parenteral nutrition-associated liver disease received parenteral -3 fatty acids while awaiting liver transplant. They
showed restoration of liver function to the point that 9 of
the children were no longer considered for liver transplant.
The remaining 3 children received a liver transplant with no
complications attributable to the -3 emulsion. 33 In another
study, 18 infants who developed cholestasis while receiving
a parenteral emulsion high in -6 fatty acids were switched
to an emulsion high in -3 fatty acids and compared to
21 historical controls who had similar symptoms and had
been maintained on the -6 fatty acid emulsion. Patients
receiving the -3 fatty acids experienced a reversal of
cholestasis 6.8 times faster when the data were adjusted for
baseline bilirubin concentration, gestational age, and diagnosis of necrotizing enterocolitis. The -3 fatty acid cohort
had had 2 deaths and no liver transplants and the historical
control cohort had 7 deaths and 2 liver transplantations. 34
Parenteral lipid emulsions containing fish oil are still
not approved for use in children or available in the United
States and Canada.
FATS
1. In order to maintain health and prevent a home parenteral nutrition patient from becoming overweight, the
optimal nutrition support prescription:
A. Restricts fat to an absolute minimum while
providing a generous amount of glucose.
B. Provides adequate calories including a fatty acid
blend with appropriate chain length, chain saturation, and ratios of LA, -LA, ARA, EPA, and DHA.
C. Provides 80% of the calories recommended for oral
intake.
D. Provides all the fat calories as the essential fatty
acids LA and -LA.
2. The cellular site(s) for fatty acid oxidation in the body
is (are):
A. Mitochondria
B. Peroxisomes
C. Cytoplasm
D. Mitochondria, peroxisomes, and cytoplasm
3. The essential fatty acid needs of a critically ill 24-week
neonate in the neonatal intensive-care unit requires
dietary:
A. LA and -LA
B. -LA and DHA
C. LA and ARA
D. LA, -LA, ARA, and DHA
See p. 487 for answers.
References
29
30
33. Diamond IR, Sterescu A, Pencharz PB, Kim JH, Wales PW.
Changing the paradigm: omegaven for the treatment of liver
failure in pediatric short bowel syndrome. J Pediatr Gastroenterol Nutr. 2009;48(2):209215.
34. Gura KM, Lee S, Valim C, et al. Safety and efficacy of
a fish-oil-based fat emulsion in the treatment of parenteral nutrition-associated liver disease. Pediatrics.
2008;121(3):e678e686.
35. Riserus U. Fatty acids and insulin sensitivity. Curr Opin Clin
Nutr Metab Care. 2008;11(2):100105.
36. Assy N, Nassar F, Nasser G, Grosovski M. Olive oil consumption and non-alcoholic fatty liver disease. World J Gastroenterol.
2009;15(15):18091815.
Richard A. Helms, PharmD, Emma M. Tillman, PharmD, Anup J. Patel, MD, and John A. Kerner, MD
Learning Objectives
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Ontogeny of the Gastrointestinal Tract
inRelation to Protein. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Protein Digestion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Protein Absorption. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Digestion and Absorption of Whey and Casein . . . . . . . . .
Amino Acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
31
31
32
33
34
35
Introduction
32
neurocrine pathways.2,3 Selected gastrointestinal regulatory peptides involved with the ontogeny of the GI tract as
it relates to proteins are outlined in Table 5-1. All of these
peptides are present by the end of the first trimester in the
fetus, but adult levels may not be present until term.4 These
peptides that function as hormones are released in response
to feeding. The release of some of these hormones is limited
in the newborn compared to the adult.4,5
There are significant differences in the ontogeny of
the two main enzymes secreted by the gastric mucosa. In
the human, even though both pepsin and gastric lipase are
located at the same site (ie, in the chief cells of the gastric
mucosa), the enzymes have different developmental
patterns.6 Pepsin activity and output are much lower in
infants than adults.7,8 In contrast, gastric lipase activity and
output are equal in infants and adults.7,8
The ontogeny of the brush border amino-oligopeptidases as well as the dipeptide and amino acid transport
systems parallel that of the carbohydrate enzymes. Aminooligopeptidases are first detected immunohistochemically
by 10 to 16 weeks gestation. By 28 to 30 weeks gestation,
enzyme levels are approximately one-half of the values
found in term infants. Therefore, all aspects of intestinal
Protein Digestion
Sources
Mechanisms
Actions
Salivary glands
Brunner glands
Paneth cells
Hormone
Paracrine
Gastrointestinal tract
Liver
Hormone
Autocrine
Paracrine
Enteroglucagon
Ileum
Colon
Hormone
Neurotensin
Ileum
Central nervous system
Hormone
Neurocrine
Vasoactive Intestinal
Polypeptide
All tissues
Neurocrine
Pancreatic Polypeptide
Pancreas
Hormone
Motilin
Hormone
Peptide YY
Gastrointestinal tract
Central nervous system
Hormone
Paracrine
Neurocrine
Reprinted from Gilger MA. Normal gastrointestinal function. Table 342-1. In: McMillan JA, Feigin RD, DeAngelis C, Jones MD, eds. Oskis Pediatrics. 4th ed.
Copyright 2006 with permission from Lippincott Williams & Wilkins.
2010 A.S.P.E.N. www.nutritioncare.org
stimulates both gastric acid and pepsin production and secretion, which initiates protein digestion in the stomach.10
The protein denaturation within the stomach does not
appear to be critical because patients with a more neutral
gastric pH do not have impaired protein digestion. However,
the amino acids that are produced from protein digestion in
the stomach do assist in releasing cholecystokinin (CCK)
or pancreozymin.12 CCK has a role in protein digestion by
helping to release pancreatic digestive enzymes, stimulate
gallbladder contraction, and relax the sphincter of Oddi.11,12
In contrast, the hormone secretin promotes pancreatic
secretion of bicarbonate-rich fluid to help establish a favorable pH.10
The pancreatic digestive enzymes are secreted in their
inactive proenzyme form, similar to the secretion of pepsin
as pepsinogen in the stomach. The pancreas secretes both
endopeptidases (trypsinogen, chymotrypsinogen, and
proelastase) and exopeptidases (procarboxypeptidase A
and B) into the proximal small intestine.11 The activated
endo- and exopeptidases hydrolyze the proteins at peptide
bonds within the polypeptide chains to form oligopeptides
and at the carboxyl terminal to form single amino acids,
respectively.911
Bile acids and trypsinogen together cause the release
of enterokinase, a brush border enzyme, which converts
trypsinogen into its active form trypsin. Trypsin then
converts the remaining pancreatic peptidases into their
respective active forms (chymotrypsin, elastase, and
carboxypeptidase A and B), and assists in forming more
trypsin from trypsinogen (see Figure 5-1).9,10,13 Overall, the
products of lumenal protein digestion are about 70% oligopeptides and 30% free amino acids.9
Enterokinase and trypsin have been detected at 26
and 28 weeks gestation, respectively. Enterokinase levels at
time of birth are about 10% of adult levels except in those
rare infants with congenital enterokinase deficiency.9,14
Trypsin activity in duodenal juice of the premature infant
is slightly less than in the full-term infant but does increase
in response to food as it does in full-term infants. Infants
have been found to have decreased trypsin activity in
duodenal fluid compared to older children.15 This reduced
trypsin activity has been demonstrated to increase over the
first 4 months of life, possibly resulting from their relative
increased enteral protein intake.9 However, the significance
of these differences is unclear given that protein digestion
and absorption is fairly efficient even in preterm infants. It
is estimated that term infants digest and absorb about 80%
to 85% of lumenal proteins while estimates for adults range
from 95% to 98%.9,11
33
Reprinted from Wahbeh GT, Christie DL. Basic aspects of digestion and
absorption. In: Wyllie R, Hyams JS, eds. Pediatric Gastrointestinal and Liver
Disease: Pathophysiology, Diagnosis, Management. 3rd ed.
Copyright 2006 with permission from Elsevier.
Protein Absorption
34
Reprinted from Roy CC, Silverman A, Alagille D. Pediatric Clinical Gastroenterology. Malabsorption syndrome. p. 307. (Modified and published with
permission from Ahnen DJ. Protein digestion and assimilation. In Yamada T, et al. Textbook of Gastroenterology, Philadelphia. 1991. Lippincott.)
Copyright 1995 with permission from Elsevier.
The major proteins in milk are whey and casein. Unmodified cows milk is approximately 18% whey and 82% casein.
When this unheated casein-predominant protein enters
the acidic environment of the stomach, it forms a relatively
hard curd of casein and minerals that can be difficult to
digest. In contrast, human milk protein is approximately
60% to 70% whey and 30% to 40% casein.20,21 Human milk
forms a very small, soft curd in acid. Some infant formula
companies have developed whey-predominant formulas by
combining equal amounts of demineralized whey protein
35
Amino Acids
Conditionally Essential
Non-Essential
Histidine*
Cyst(e)ine
Alanine
Isoleucine
Taurine
Aspartic Acid
Leucine
Tyrosine
Asparagine
Lysine
Glutamic Acid
Methionine
Glutamine
Phenylalanine
Glycine
Threonine
Proline
Tryptophan
Serine
Valine
Arginine
36
37
The amino acids of the urea cycle are considered nonessential (Figure 5-5). Snyderman50 found no evidence that
arginine is essential in preterm infants. However, early
amino acid solutions with relatively lower concentrations of
arginine resulted in hyperammonemia in infants receiving
these formulas as part of PN. 51 Higher concentrations of
arginine reversed the finding of hyperammonemia. 52 Acute
renal failure patients given large doses of protein with
little supplemental arginine also present with hyperammonemia, and can be reversed with the supplementation
of arginine. 53 Treatment of urea cycle disorders generally
involves the supplementation of low-dose essential amino
acids, andarginine.
38
lower concentrations in plasma in premature infants. Lipoprotein concentrations rise rapidly after birth in response
to high dietary fat intake, while albumin and TTR increase
toward adult values in the first months to year of life. Acute
phase proteins, such as C-reactive protein (CRP), appear to
be induceable even in prematurity, and are used to monitor
inflammatory response to infection or metabolic stress.
Reduction in visceral protein occurs in preterm infants,
and response to protein and energy intake can be assessed
through monitoring changes in serum concentrations of
albumin, TTR, and RBP. 57
The production of glutathione in the liver is essential;
however, other tissues can produce the tripeptide. Glutathione is an important sulfhydryl-reducing agent that
protects cells from oxygen free radicals. 58 It is synthesized
from glutamate, cysteine, and glycine, and studies have
shown cysteine is the rate-limiting substrate in glutathione
production. It is dependent in two distinct steps on cysteine
production via the transsulfuration pathway (principally in
the liver), or from dietary intake. The tripeptide also appears
to be important in the transport of amino acids, and in the
synthesis of leukotrienes via the enzyme gamma-glutamyl
transpeptidase. 59,60
The metabolism of amino acids is an important function of the liver. Several enzymes and enzyme systems, such
as the transsulfuration pathway and phenylalanine hydroxylase, both predominantly located in the liver, are responsible
for the metabolism of the essential amino acids methionine
and phenylalanine, respectively. Enzyme activity is reduced
in prematurity and early infancy, rendering the products
of these enzyme systems as conditionally essential amino
acids.
Approximately one-third of amino acids entering the
liver from portal blood are used for protein synthesis.61 The
remainder may be used in energy production, or gluconeogenesis, with perhaps only a third of dietary amino acids
entering to the peripheral blood. This explains why plasma
amino acids do not fluctuate substantially in the postprandial period. One group of amino acids that are released at
higher relative concentrations from the liver in the postprandial period are the branched-chain amino acids.62 These
are leucine, isoleucine, and valine. The branched-chain
amino acids are preferentially metabolized in the periphery
as these amino acids and arginine stimulate insulin release
and muscle protein synthesis.63 All is reversed in the postabsorptive period. As insulin concentrations fall, the
muscle then provides the principal gluconeogenic substrate,
alanine, to the liver for production of glucose in the fasting
period.
39
Protein has multiple functions; it is essential for cell structure, maturation, remodeling, and growth. Besides being
utilized for energy, amino acids and proteins serve as precursors that are essential for many biological processes.67
When protein is consumed, it is extensively broken
down in the GI tract to amino acids, which can then enter
cells or continue to circulate in plasma. Once in the cell,
amino acids can be combined by peptide linkages to form
small peptides (such as glutathione), they can be substrates
for protein synthesis, or they can function as individual
amino acids in the urea cycle. Specific amino acids can act
as substrates, regulators, transporters, and precursors to
neurotransmitters and hormones.67
In the cell, protein can be utilized for energy or it can be
stored. There is a continuous flux of proteins being broken
down. The carbon chain of the amino acids can be utilized
for energy, and free amino acids can be released back into the
plasma to maintain plasma amino acid concentrations. 67
After synthesis within the cell, many proteins are
Amino acids
Substrates for protein synthesis
Regulators of protein turnover
Regulators of enzyme activity (allosteric)
Precursor of signal transducer
Methylation reactions
Neurotransmitters
Ion fluxes
Precursor of physiologic molecules
Nitrogen transport
Oxidation and reduction
Precursor of conditionally indispensable amino acids
Gluconeogenic substrate and fuel
Proteins
Enzymatic catalysis
Transport
Messenger / signals
Movement
Structure
Storage / sequestration
Immunity
Growth; differentiation; gene expression
Example
40
34
2.53
22.5
1.52
11.5
0.81.5
41
References
42
18. Motil KJ. Meeting protein needs. In: Tsang RC, Zlotkin SH,
Nichols BL, Hansen JW, eds. Nutrition During Infancy: Principles and Practice. 2nd ed. Cincinnati, OH: Digital Education
Publishing, Inc; 1997:83103.
19. Philipps AF, Sherman MP. Neonatal nutrition and gastrointestinal function. In: Rudolph CD, Rudolph AM, Hostetter
MK, Lister GE, Siegel NJ, eds. Rudolphs Pediatrics. 21st ed.
New York, NY: McGraw-Hill; 2003.
20. Hansen JW, Boettcher JA. Human milk substitutes. In: Tsang
RC, Zlotkin SH, Nichols BL, Hansen JW, eds. Nutrition
During Infancy: Principles and Practice. 2nd ed. Cincinnati,
OH: Digital Education Publishing, Inc; 1997:441466.
21. Schanler, RJ. Nutritional composition of human milk and
preterm formula for the premature infant. UpToDate Web site.
http://www.uptodate.com/home/index.html. September 7,
2007. Accessed December 19, 2008.
22. Lnnerdal B, Cederblad A, Davidsson L, Sandstrm B.
The effect of individual components of soy formula and
cows milk formula on zinc bioavailability. Am J Clin Nutr.
1984;40(5):10641070.
23. Brock, JH. Lactoferrin in human milk: its role in iron absorption and protection against enteric infection in the newborn
infant. Arch Dis Child. 1980 Jun;55(6):417421.
24. Lnnerdal B. Digestibility and absorption of protein in
infants. In: Rih NC, ed. Protein Metabolism During Infancy.
New York, NY: Raven Press; 1994:5365.
25. Lindberg T, Ohlsson K, Westrm B. Protease inhibitors and
their relation to protease activity in human milk. Pediatr Res.
1982 Jun;16(6):479483.
26. Davidson LA, Lonnerdal B. Fecal alpha 1-antitrypsin
in breast-fed infants is derived from human milk and is
not indicative of enteric protein loss. Acta Paediatr Scand.
1990;79(2):137141.
27. Rose WC. The amino acid requirements of adult man. Nutr
Abstr Rev. 1957;27:631647.
28. Nasset ES, Gatewood VH. Nitrogen balance and hemoglobin
of adult rats fed amino acid diets low in L- and D-histidine. J
Nutr. 1956;53:163176.
29. Kopple JD, Swendseid ME. Evidence that histidine is an
essential amino acid in normal and chronically uremic man.
J Clin Invest. 1975;55:881891.
30. Kopple JD, Swendseid ME. Effect of histidine intake on plasma
and urine histidine levels, nitrogen balance and N-methylhistidine excretion in normal and chronically uremic men. J Nutr.
1981;111:931942.
31. Snyderman SE, Boyer A, Roitman E, Holt LE Jr, Prose
PH. The histidine requirement of the infant. Pediatrics.
1963;31:786801.
32. Heird WC, Dell RB, Helms RA, et al. Evaluation of an amino
acid mixture designed to maintain normal plasma amino acid
patterns in infants and children requiring parenteral nutrition.
Pediatrics. 1987;80:401408.
33. Heird WC, Hay W, Helms RA, Storm MC, Kashyap S, Dell,
RB. Pediatric parenteral amino acid mixture in low birth
weight infants. Pediatrics. 1988;81(1):4150.
43
52. Thomas DW, Sinatra FR, Hack SL, Smith TM, Platzker ACG,
Merritt RJ. Hyperammonemia in neonates receiving intravenous nutrition. J Parenter Enteral Nutr. 1982;6:503506.
53. Motil KJ, Harmon WE, Grupe WE. Complications of essential amino acid hyperalimentation in children with acute renal
failure. J Parenter Enteral Nutr. 1980;4:3235.
54. Laidlaw SA, Kopple JD. Newer concepts of the indispensable
amino acids. Am J Clin Nutr. 1987;46:593605.
55. Raiha NCR, Suihkonen J. Development of urea-synthesizing enzymes in human liver. Acta Pediatr Scand.
1968;57:121124.
56. Raiha NCR, Lindros KO. Development of some enzymes
involved in gluconeogenesis in human liver. Ann Med Exp
Biol. 1969;47:146150.
57. Helms RA, Dickerson RN, Ebbert ML, Christensen ML,
Herrod HG. Retinol-binding protein and prealbumin: useful
measures of protein repletion in critically ill, malnourished
infants. J Pediatr Gastroenteral Nutr. 1986;5:586592.
58. Beutler E. Nutritional and metabolic aspects of glutathione.
Annu Rev Nutr. 1989;9:287302.
59. Meister A. On the enzymology of amino acid transport.
Science. 1973;180:3339.
60. Svartz J, Hallin E, Soderstorm M, Hammarstrom S. Identification of regions of leukotriene C4 synthase which direct the
enzyme to its nuclear envelope localization. J Cell Biochem.
2006;98;15171527.
61. Yamamoto H, Aikawa T, Matsutaka H, Okuda O, Ishikawa
E. Interorganal relationships of amino acid metabolism in fed
rats. Am J Physiol. 1974;226;14281433.
62. Munro HN. Fifth annual Jonathan E. Rhoads lecture. Metabolic integration of organs in health and disease. J Parenter
Enteral Nutr. 1982;6:271.
63. Rocha DM, Falona GR, Unger RH. Glucagon-stimulating activity of 20 amino acids in dogs. J Clin Invest.
1972;51:2346.
64. Raiha NCR, Kekomaki MP. Studies on the development of
ornithine-keto acid amino transferase activity in the liver.
Biochem J. 1968;108:521524.
65. Edelman CM, Wolfish NM. Dietary influence on renal maturation in premature infants. Pediatr Res. 1968;2:421422.
66. Brodehl J, Gellissen K. Endogenous renal transport of
free amino acids in infancy and childhood. Pediatrics.
1968;42:395404.
67. Protein metabolism. In: Guyton AC, ed. Textbook of
Medical Physiology. Philadelphia, PA: W.B. Saunders Co;
1991:765770.
68. Wu PYK, Edwards N, Storm MC. The plasma amino
acid pattern of normal term breast-fed infants. J Pediatr.
1986;109:347349.
69. American Academy of Pediatrics, Committee on Nutrition.
Protein. In: Kleinman RE. Pediatric Nutrition Handbook. 6th
ed. Elk Grove Village, IL: American Academy of Pediatrics;
2009:325341.
70. Scrimshaw NS. Effect of infection on nutritional status. Proc
Natl Sci Counc Repub China B. 1992;16:4664.
44
71. Poindexter BB, Langer JC, Dusick AM, Ehrenkranz RA. Early
provision of parenteral amino acids in extremely low birth
weight infants: relation to growth and neurodevelopmental
outcome. J Pediatr. 2006;148:300305.
72. Boehm KA, et al. Assessing the validity of adjusted urinary
urea nitrogen as an estimate of total urinary nitrogen
in three pediatric populations. J Parenter Enteral Nutr.
1994;18(2):172176.
73. Lopez et al. Estimation of nitrogen balance based on a sixhour urine collection in infants. J Parenter Enteral Nutr.
1986;10(5):517518.
74. Helms RA, Mowatt-Larssen CA, Boehm KA, et al. Urinary
nitrogen constituents in the postsurgical preterm neonate
receiving parenteral nutrition. J Parenter Enteral Nutr.
1993;17:6872.
75. Strom MC, Helms RA. Normalizing plasma amino acid levels
in pediatric patients requiring parenteral nutrition. Nutr Clin
Pract. 2007;22:194203.
76. Helms RA, Christensen ML, Muer EC, Storm MC. Comparison of a pediatric versus standard amino acid formulation
in preterm neonates requiring parenteral nutrition. J Pediatr.
1987;110:466470.
77. Beck R. Use of a pediatric parenteral amino acid mixture in a
population of extremely low birth weight neonates: frequency
and spectrum of direct bilirubinemia. Am J Perinatol. 1990
Jan;7(1):8486.
78. Forchielli ML, Gura KM, Sandler R, Lo C. Aminosyn PF
or TrophAmine: which provides more protection from
cholestasis associated with total parenteral nutrition? J Pediatr
Gastroenterol Nutr. 1995;21:374382.
79. Pratt CA, Garcia MG, Poole RL, Kerner JA. Life-long total
parenteral nutrition versus intestinal transplantation in children with microvillus inclusion disease. J Pediatr Pharmacol
Ther. 2001;6:498503.
80. Fitzgerald KA, MacKay MW. Calcium and phosphate solubility in neonatal parenteral nutrient solutions containing
TrophAmine. Am J Hosp Pharm. 1986 Jan;43(1):8893.
Minerals
Winston Koo, MBBS, FACN, CNS, Mirjana Lulic-Botica, BSc, BCPS, May Saba, PharmD, BCNSP, and Letitia Warren, RD, CSP
CONTENTS
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Biochemistry and Physiology
Sources
Dietary Requirements
Absorption and Excretion
Metabolism
Deficiency States
Excess Intake and Adverse Effects
Calcium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
Biochemistry and Physiology
Sources
Absorption, Excretion, and Metabolism
Deficiency States
Excess Intake and Adverse Effects
Phosphorus. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50
Biochemistry and Physiology
Sources
Absorption, Excretion, and Metabolism
Deficiency States
Excess Intake and Adverse Effects
Magnesium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
Biochemistry and Physiology
Sources
Absorption, Excretion, and Metabolism
Deficiency States
Excess Intake and Adverse Effects
Learning Objectives
Overview
Biochemistry and Physiology
Calcium (Ca) is the most abundant mineral in the body and
together with phosphorus (P) forms the major inorganic
constituent of bone. Magnesium (Mg) is the fourth most
abundant cation and is the second most common intracellular electrolyte in the body. The total body content of
Ca, P, and Mg in the skeleton is about 99%, 85%, and 60%,
respectively.
Less than 1% of the total body content of Ca, P, and
Mg is in the circulation. However, disturbances in serum
concentrations of these minerals are associated with disturbances of physiological function. These are manifested by
numerous clinical symptoms and signs. Acute changes in
the serum concentrations likely reflect adaptive changes and
do not reflect the state of tissue store, although chronic and
severely lowered serum concentrations of these minerals
usually reflect the presence of a deficiency state.
From a clinical perspective, the skeleton has the dual
function of providing both structural and mechanical
support as well as being a reservoir to maintain mineral
45
46
Sources
Natural foods and a variety of foods and beverages fortified
with Ca are good sources for these minerals. Any food that
provides 20% or more of the daily recommended intake per
serving for a specific nutrient is considered to be high in
that nutrient.2 Dietary sources of minerals, including Ca
fortified foods and beverages, are preferred to the use of
supplements alone because the range of nutrients and the
establishment of good dietary habits are enhanced by the
use of dietary sources. Nutrient interactions also may be
less and tolerance may be greater for minerals provided by
food sources.
Mineral supplements are freely available. However, use
of supplements does not compensate for poor food choices
and inadequate diet. Furthermore, the Food and Drug
Administration (FDA) regulates dietary supplements under
a different set of regulations than drug products (prescription and over-the-counter). As a result, specific contents of
dietary supplements and contaminants are not as rigorously
monitored. 3
Calcium*
mg/d
Phosphorus
mg/d
Magnesium
mg/d
06 mo*
712 mo*
13 y
48 y
Males
913 y
1418 y
Females
913 y
1418 y
Pregnancy
1418 y
Lactation
1418 y
UL
210
270
500 (2500)
800 (2500)
100
275
460 (3000)
500 (3000)
30
75
80 (65)
130 (110)
1300 (2500)
1300 (2500)
1250 (4000)
1250 (4000)
240 (350)
410 (350)
1300 (2500)
1300 (2500)
1250 (4000)
1250 (4000)
240 (350)
360 (350)
1300 (2500)
1250 (3500)
400 (350)
1300 (2500)
1250 (4000)
360 (350)
Dietary Requirements
MINERALS
Metabolism
Maintaining mineral homeostasis requires a complex interaction of hormonal and non-hormonal factors; adequate
functioning of various body systems, in particular the
renal, gastrointestinal, skeletal, and endocrine systems; and
adequate dietary intake. When assessing changes in serum
concentrations of these minerals, it is vital to understand
there is an interrelationship among them. For example,
hypomagnesemia may cause hypocalcemia, due to the
decreased action of parathyroid hormone (PTH). Serum
Ca and P have a reciprocal relationship, and hypoalbuminemia can cause hypocalcemia secondary to decreased Ca
binding, while ionized calcium (unbound calcium) concentration remains normal.
At the intestine-kidney-bone axis, intake of minerals
may interact with other nutrients including protein, sodium,
potassium, vitamin D, iron, zinc, and copper. There may be
significant effects on intestinal absorption, renal excretion,
or metabolism of the minerals or on these other nutrients.
Direct regulation on this axis by PTH, 1,25-dihydroxyvitamin D, and fibroblast growth factor-23, and indirect
regulation by growth-regulating hormones including sex
hormones, also significantly affect growth and mineralization of the skeleton, and maintenance of normal circulating
concentrations of these minerals. A negative effect on
mineral metabolism is possible, particularly if minerals and
other nutrients stated above are ingested in large amounts
as dietary supplements. Thus, it is vital to manage the cause
of the abnormalities in addition to providing symptomatic
treatment to the abnormal circulating concentration of the
minerals.
Deficiency States
Low dietary intake for Ca is common in older children. Less
than 40% of boys and < 30% of girls 6 years or older receive
the recommended daily adequate intake for Ca. Preoccupation with being thin is common in adolescents, especially
among females, as is the misconception that all dairy foods
are fattening. Many children and adults are unaware that
low-fat milk contains at least as much Ca as does whole
milk. A list of foods relatively high in Ca is shown in Table
6-2. Low Ca intake places children at risk for fractures, and
both Ca and vitamin D deficiency are factors in the development of rickets in infants and young children. 8,9
47
48
Calcium
(1 mmol = 40 mg)
Sources
Calcium is present in many dietary sources with dairy
products having the best bioavailability (Table 6-2) and
accounting for > 70% of dietary Ca intake in the United
States.2,5 Non-fat and reduced fat dairy products contain the
same amount of Ca as regular dairy products. Some food
sources (eg, fruit juices, fruit drinks, tofu, and cereals) are
fortified with various Ca compounds that are well absorbed.
Calcium is present in human milk in relatively constant
amounts between 200 to 250 mg/L. Various Ca salts are
added to cows milk formulas for term infants to provide
at least 60 mg/100 kcal. This is about twofold higher than
the Ca density in human milk.13 Calcium fortification of
formulas for small preterm infants may be four- to sixfold
higher than human milk.
Oral supplements such as calcium compounds
containing carbonate and citrate are the most common
although preparations containing other anions are available.
Naturally occurring products (eg, oyster shell marketed as
calcium supplement) contain high levels of lead, mercury,
and other potentially toxic contaminants.14 Common
parenteral supplements include calcium gluconate and
calcium chloride but other compounds also are available.
The proportion of elemental calcium by weight varies with
the calcium compound and is 40% for carbonate, 38% for
tribasic calcium phosphate, 21% for citrate, 13% for lactate,
9% for gluconate, and 6.4% for glubionate.14,15
MINERALS
49
Deficiency States
Risk factors for a deficient state include increased need, such
as the rapidly growing infant, or the presence of disease
states that affect Ca digestion, absorption, or metabolism.
Physically active females, particularly those with secondary
menstrual disorders, may have lower Ca absorption and
decreased bone formation, and adequate Ca intake is important to their bone health.26 Lactose-intolerant individuals
may be at risk for Ca deficiency, not due to an inability to
absorb Ca, but rather from the avoidance of dairy products. 2
Drinking milk with a meal and other dietary options (eg,
choosing aged cheeses (such as Cheddar and Swiss) which
contain little lactose, yogurt which contains live active
cultures that aid in lactose digestion, or lactose-reduced
and lactose-free milk) may allow increased Ca intake. Strict
vegans27 should include adequate amounts of non-dairy
sources of Ca in their daily diets or likely will need a Ca
supplement to meet the recommended Ca intake.
Dietary Ca deficiency is a risk factor for fractures8
although it is usually not associated with clinical or biochemical manifestations unless the Ca intake is extremely low.
Hypocalcemia does not usually occur due to low Ca intake
alone but occurs with concomitant deficiency of vitamin
D or Mg or associated medical problems. Severe hypocalcemia may result in clinical signs and symptoms that may
be subtle and vary with maturity of the individuals. Preterm
infants often manifest with non-specific symptomatology
that may include irritability, jitteriness or lethargy, feeding
poorly with and without feeding intolerance, abdominal
distention, apnea, cyanosis, and seizures. These features
may be confused with manifestations of hypoglycemia,
sepsis, meningitis, anoxia, intracranial bleeding, and
narcotic withdrawal. The degree of irritability of the infant
does not appear to correlate with serum Ca values. In more
mature individuals, other symptoms and signs may include
2010 A.S.P.E.N. www.nutritioncare.org
50
Phosphorus
(1 mmol = 31 mg)
Sources
Phosphorus is ubiquitous in natural foods and is present
in both organic and inorganic forms. Various phosphate
salts used in food processing for non-nutritive functions
(eg, moisture retention, smoothness, and binding) provide
significant contributions to dietary P intake. Cola soft
drinks contain phosphoric acid as the acidulant and provide
about 50 mg of P per 12 oz serving.2 Phosphorus intake
fromsoft drinks can be substantial when multiple beverages
are consumed.
Dairy and meat products have high P density. Plants,
nuts, and seeds also are significant sources of P. Animal or
synthetic protein has more bioavailable phosphorus than
soy or grain-based protein. Phosphorus in plants (beans,
peas, cereals, nuts) is present in the poorly digestible
phytic acid. However, some phytate P is absorbed from the
combined effect of food phytases, colonic bacteria enzymes,
MINERALS
51
Deficiency States
52
Magnesium
(1 mmol = 24 mg)
Sources
Magnesium is ubiquitous in foods but the Mg content of
foods varies substantially. Green leafy vegetables are rich
in Mg because chlorophyll is the Mg chelate of porphyrin.
Unpolished grains and nuts also have high Mg content,
whereas meats and milk have intermediate levels. Refined
foods generally have the lowest Mg content.40,41 Water is a
variable source of Mg depending on the source of ground
water. Typically, hard water has higher concentrations
of Mg salts.2,41 Human milk contains adequate amounts of
Mg and infant formulas are mandated to contain at least
6mg/100 kcal.13 Supplements containing various Mg salts
are freely available in oral forms. The proportion of elemental
magnesium by weight is 60.3% for oxide; 28% for carbonate;
16% for citrate; ~12% for chloride, acetate, and lactate;
9.7% for sulfate; 6.8% for phosphate; 6.4% for ascorbate;
and 5.4%for gluconate.15 Magnesium sulfate is available in
parenteral form. Mg supplements may contribute a substantial portion of daily intake and are used as a basis for the
derivation of tolerable upper intake levels.2
MINERALS
alcohol intake, as well as elevated serum Mg or Ca, depletion of potassium and phosphate, and metabolic acidosis
inhibit Mg and Ca reabsorption, leading to increased urine
excretion of both cations.43
Deficiency States
Magnesium deficiency is usually associated with malabsorption, increased losses from the gut or kidney, or during
refeeding of severe and chronically malnourished individuals. Chronic therapy with loop diuretics, cisplatin, and
tacrolimus are among the increasing list of medications
that result in increased renal Mg excretion and predisposition to Mg deficiency.43 There are also increasing reports of
patients with heritable defects of Mg transport that lead to
Mg wasting and deficiency states.44
The typical deficit required to produce symptomatic hypomagnesemia is approximately 0.5 to 1 mmol
(1224 mg)/kg of body weight.45 Hypomagnesemia is
usually associated with a significant Mg deficit. These
individuals often are at risk for concurrent hypocalcemia,
hypokalemia, hypophosphatemia, and possible disturbance of acid-base status. The loss of other nutrients such
as zinc from the gastrointestinal secretion also may be
considerable. Symptoms and signs of hypomagnesemia,
which often coexists with hypocalcemia, may be indistinguishable. Prolonged dietary Mg deprivation in human
adults leads to personality change, tremor, muscle fasciculations, spontaneous carpopedal spasm, and generalized
spasticity as well as hypomagnesemia, hypocalcemia, and
hypokalemia.45 In infants, acute complications associated with clinical manifestations include seizure, apnea,
cyanosis and hypoxia, electrocardiographic changes,
bradycardia, and hypotension.
53
54
5. All of the following statements associated with magnesium deficit are correct except for:
A. May be associated with chronic gastrointestinal
losses
B. May result in fecal fat loss
C. May be associated with deficiency of other
nutrients
D. May result in hypocalcemia
E. May be asymptomatic
6. Which of the following statements about hypermagnesemia is incorrect?
A. Can occur with drinking hard water
B. Can result in cardiac arrhythmia
C. Can cause respiratory failure
D. Usually occurs only with pharmacologic doses of
magnesium
E. Can decrease muscle tone
See p. 487 for answers.
References
MINERALS
27. Key TJ, Appleby PN, Rosell MS. Health effects of vegetarian
and vegan diets. Proc Nutr Soc. 2006;65:3541.
28. Shannon MT, Wilson BA, Stang CL. Health Professionals Drug
Guide. Stamford, CT: Appleton & Lange; 2000.
29. Jellin JM, Gregory P, Batz F, Hitchens K. Pharmacists Letter/
Prescribers Letter Natural Medicines Comprehensive Database.
3rd ed. Stockton, CA: Therapeutic Research Facility; 2000.
30. U.S. Food and Drug Administration. Information for Healthcare Professionals: Ceftriaxone (marketed as Rocephin
and generics). http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/
ucm084263.htm#main. Accessed November 20, 2009.
31. Nordin BEC. Phosphorus. J Food Nutr. 1988;45:6275.
32. Takeda E, Taketani Y, Sawada N, Sato T, Yamamoto H. The
regulation and function of phosphate in the human body.
Biofactors. 2004;21:345355.
33. Tenenhouse HS. Regulation of phosphorus homeostasis by
the type IIa Na/phosphate cotransporter. Annu Rev Nutr.
2005;25:197214.
34. Specker BL, Lichtenstein P, Mimouni F, Gormley C, Tsang
RC. Calcium-regulating hormones and minerals from birth
to 18 months of age: a cross-sectional study. II. Effects of sex,
race, age, season, and diet on serum minerals, parathyroid
hormone, and calcitonin. Pediatrics. 1986;77:891896.
35. Freiman I, Pettifor JM, Moodley GM. Serum phosphorus
in protein energy malnutrition. J Pediatr Gastroenterol Nutr.
1982;1:547550.
36. Lotz M, Zisman E, Bartter FC. Evidence for a
phosphorus-depletion syndrome in man. N Engl J Med.
1968;278:409415.
37. Elin RJ. Assessment of magnesium status. Clin Chem.
1987;33:19651970.
55
Winston Koo, MBBS, FACN, CNS, Judith Christie, RN, MSN, May Saba, PharmD, BCNSP,
Mirjana Lulic-Botica, BSc, BCPS, and LetitiaWarren, RD, CSP
CONTENTS
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
Digestion, Absorption, and Metabolism. . . . . . . . . . . . . . . 57
Dietary Reference Intake. . . . . . . . . . . . . . . . . . . . . . . . . . . 57
Increased Demands and Predisposition toDeficiency
States . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
Management of Deficiency State andSupplementation . 59
Specific Nutrients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Thiamin (Vitamin B1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Riboflavin (Vitamin B2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
Niacin (Vitamin B3) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Vitamin B6. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
Folate (Vitamin B9). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
Vitamin B12 (Cobalamin) . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
Vitamin C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
Pantothenic Acid (Vitamin B5) . . . . . . . . . . . . . . . . . . . . . . 69
Biotin (Vitamin B7). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
Choline . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
56
Learning Objectives
Overview
57
These dietary micronutrients are released with the digestion of foods and then absorbed in free form or as small
molecular complexes. Absorption is usually by active transport at low nutrient intake and by passive diffusion at high
intake. Absorption occurs mainly at the jejunum except for
cobalamin which is absorbed only at the ileum under physiological conditions. Some absorption occurs at the stomach
(niacin) and proximal colon (riboflavin, biotin).11 Postabsorptive transport usually occurs as an enzyme complex or
bound to proteins. Erythrocytes may serve both as transporter and storage source. Tissue uptake is usually specific
to each nutrient. Some of these essential nutrients have
interrelated metabolism and function. The bioavailability
of water-soluble vitamin supplements is generally similar to
or better than that from dietary sources when tested under
the same conditions.1,2,5,6,9
Table 7-1 Content Per Unit Dose of Multivitamin Preparations for Use with Parenteral Nutrition*
Ingredient
M.V.I.
Pediatric
M.V.I. Adult
(for ages 11 y)
M.V.I. 12
(no vitamin K)
10
10
0.7
10
7
200
1
5
10
150
1
5
10
none
1.2
1.4
17
1
140
1
80
5
20
6
3.6
40
6
600
5
200
15
60
6
3.6
40
6
600
5
200
15
60
* Data from manufacturer product insert (Hospira, Inc., Lake Forest, IL).
Lyophilized powder reconstituted immediately prior to use.
Dual vial liquid formulation. Vial one or lower chamber of unit vial contains fat-soluble vitamins A, D, E, and K; and water-soluble vitamins B1, B2, B6, C,
niacinamide, and dexpanthenol. Vial two or upper chamber of unit vial contains: biotin, folic acid, and vitamin B12.
2010 A.S.P.E.N. www.nutritioncare.org
58
Table 7-2 Dietary Reference Intake of Water-Soluble Essential Micronutrients in Pediatric Populations1,2
Life
Stage
06 m*
712 m*
13 y
48 y
Males
913 y
1418 y
Females
913 y
1418 y
Pregnancy
1418 y
Lactation
1418 y
Highest
intake
UL
Thiamin
(B1) mg
Riboflavin
(B2) mg
Niacin
(B3) mg
B6
(Pyridoxine)
mg
Folate
(B9) mcg
B12
(cobalamin)
mcg
Vitamin
C mg
Pantothenic
acid (B5) mg*
Biotin
(B7) mcg*
Choline
mg*
0.2
0.3
0.5
0.6
0.3
0.4
0.5
0.6
2
4
6 (15)
8 (20)
0.1
0.3
0.5 (30)
0.6 (40)
65
80
150 (300)
200 (400)
0.4
0.4
0.9
1.2
40
50
15 (400)
25 (650)
0.2
0.2
2
3
0.7
0.7
8
12
125
150
200 (1000)
250 (1000)
0.9
1.2
0.9
1.3
12 (20)
16 (30)
1.0 (60)
1.3 (80)
300 (600)
400 (800)
1.8
2.4
45 (1200)
75 (1800)
4
5
20
25
375 (2000)
550 (3000)
0.9
1.0
0.9
1.0
12 (20)
14 (30)
1.0 (60)
1.2 (80)
300 (600)
400 (800)
1.8
2.4
45 (1200)
65 (1800)
4
5
20
25
375 (2000)
400 (3000)
1.2
1.4
18 (30)
1.9 (80)
600 (800)
2.6
80 (1800)
30
450 (3000)
1.4
1.6
17 (30)
2.0 (80)
500 (800)
2.8
35
550 (3000)
11 mg
at >
51 y
NA
11 mg
at
> 70 y
NA
77 mg
at
5170 y
3.9 mg
at
1930 y
625 mcg
at
5170 y
36.8 mcg
at
1455 y
NA
115
(1800)
656 mg at
5170 y
NA
NA
NA
NA
NA
* AI, Adequate intake = observed or experimentally determined estimates of nutrient intake by a group or groups of healthy people. AI is the only reference
level provided for infants < 12 months, and for pantothenic acid, biotin, and choline at all life stages and genders.
RDA, Recommended dietary allowance is calculated as + 2 standard deviations of the estimated average requirement or in its absence, a coefficient of
variation of 10% to 15% for each standard deviation is assumed for a life stage and gender group.
UL, Tolerable upper intake level = highest level of daily nutrient intake that is likely to pose no risk of adverse health effects to almost all individuals in the
general population.
Upper limit for each life stage is indicated in parenthesis.
NA = not available.
Highest intake = highest reported daily mean intake from both food and supplements at 95th percentile for any life stage or gender group.
nutrients and increasing data on the type and bioavailability of specific nutrients from naturally occurring dietary
sources, fortification of foods and drinks, and supplements.
For infants, DRI of each essential micronutrient is
based on adequate intake (AI): the average daily intake of
a specific nutrient in healthy infants fed principally human
milk during the first and second 6 months after birth. For
ages beyond 1 year, several sets of reference values are used
and include AI, estimated average requirement (EAR), and
recommended dietary allowance (RDA). Establishing AI
depends on the availability of the content of the specific
essential micronutrient in food-composition data, largescale epidemiologic studies to determine the dietary and
any supplement intake of the nutrient in group/s of healthy
people of both genders, and at the specific life stage. These
data in turn allow the determination of EAR which is a
daily nutrient intake value estimated to meet the requirement of half the healthy individuals, and RDA which is the
2010 A.S.P.E.N. www.nutritioncare.org
Requirements for many water-soluble vitamins generally increase with growth, pregnancy, lactation, physical
exertion, fever, and conditions associated with increased
metabolic needs. Tobacco and alcohol also affect the needs
for these nutrients. More than 15% of in-school youths in the
United States are reported to be current cigarette smokers12
59
60
Biological
Half-life
Major
Interactions
with Other
Nutrients
Interactions
with Drugs
Deficiency
Major Clinical
Manifestations
DeficiencyBiochemical
Erythrocyte (RC), Blood
(WB), Plasma (P)
Toxicity
Thiamin (B1)
~30 mg
918 d
Not available
Excess alcohol
& caffeine,
diuretics
Beriberi: wet,
dry. Wernicky
Encephalopathy
RC or WB Thiamin
Rapid injection:
anaphylactoidlike reaction
Riboflavin
(B2)
Very low
Minutes
Folate,
pyridoxine,
niacin
Ariboflavinosis:
pharyngitis,
cheilosis, angular
stomatitis,
glossitis,
seborrheic
dermatitis
Niacin (B3)
Not
available
45 min
Riboflavin,
pyridoxine
Anticholinergic,
tricyclic
antidepressant,
phenothiazines,
phenytoin,
probenecid,
thiazide diuretics,
doxorubicin
Isoniazid
B6
(Pyridoxine)
160 mg
assume
muscle has
80%
1228 mg,
liver has
~50%
25 d
Riboflavin,
niacin, folate,
zinc
Isoniazid, L-dopa
Erythrocyte
~8 wk
B12
Antifolate
medications
B12
(cobalamin)
23 mg,
liver has
50%+
6d
Folate
Vitamin C
2g
840 d
Glutathione,
tocopherol,
flavanoid, iron,
copper
Not
available
Bile acid
sequestrants,
H2 receptor
antagonist,
proton pump
inhibitors
Aspirin, warfarin,
proton pump
inhibitors
Folate
Not
available
Not
available
Biotin
Not
available
2h
Avidin binds
(postingestion biotin
elimination)
Not
available
Choline
Not
available
43 h
Not
available
Seizures
P pyridoxal phosphate,
RC Aspartate & alanine
transferase saturation
Pantothenic
acid
Folate
pyrophosphate (TPP);
RC transketolase activity
with TPP
RC Riboflavin; RC
erythrocyte glutathione
reductase activity
coefficient with FAD
stimulation; 24 h urine
riboflavin
Not
available
Seizures,
neuromotor
disorder,
developmental
delay,
megaloblastic
anemia
Macrocystic
megaloblastic
anemia,
Nervous system
involvement
Scurvy
Vasodilatory,
nausea &
vomiting,
hepatitis,
neurovisual
disturbance,
glucose
intolerance
Peripheral
sensory
neuropathy
RC folate, P
Homocysteine
Exacerbates
B12 deficient
neuropathy
P B12, P & U
Methylmalonic acid & P
Homocysteine
Cyanocobalamin
worsens Lebers
optic atrophy,
use hydroxy
cobalamin
Nausea,
abdominal
cramps, and
diarrhea
Not available
ascorbate
Gastrointestinal
and nervous
system
Seizures,
developmental
delay, rash,
alopecia
Non-specific
Photo-oxidation
of some amino
acids
No consistent changes in
RC, WB or P
U biotin,
3-hydroxyisovaleric acid
Not available
Hypotension,
cholinergic, fishy
odor
and phosphatidylcholine
Specific Nutrients
Thiamin (Vitamin B1)
Biochemistry and Physiology
Chemically, thiamin consists of substituted pyrimidine and
thiazole rings linked by a methylene bridge. Thiamin exists
mainly in various interconvertible phosphorylated forms:
thiamin monophosphate (TMP), thiamin triphosphate
(TTP), and ~80% as thiamin pyrophosphate (TPP). TPP,
the coenzyme form of thiamin, is involved in 2 main types
61
Source
Thiamin in the diet includes free thiamin, phosphorylated
thiamin, and protein-phosphate complexes. Dietary sources
include enriched, fortified, or whole grain rice; pasta and
cereals; pork; eggs; yeast; legumes; and nuts. Thiamin is lost
during processing to white flour, from milling of brown rice,
and during cooking.
Thiamin in fortified foods and pharmaceutical preparations are usually thiamin salts: thiamin hydrochloride
and thiamin mononitrate. Thiamin supplement is available
in oral and injectable forms as thiamin salts or as part of
multivitamin preparations. Lipid-soluble thiamin derivatives called allithiamins are also available and may be better
absorbed at higher intakes.9
Deficiency State
Thiamin deficiency can occur with inadequate intake
particularly during PN without added water-soluble vitamins,24 or inadequate absorption, and excessive loss from
multiple causes. A state of severe depletion may occur in
less than 3 weeks from a strict thiamin-deficient diet but the
2010 A.S.P.E.N. www.nutritioncare.org
62
most common cause of thiamin deficiency in affluent countries is alcoholism.16,17 Alcohol abuse is often associated
with poor dietary intake of many essential nutrients, and
decreases the absorption and phosphorylation of thiamin.
Patients with end-stage organ failure especially of the liver
are at risk for deficiency of thiamin and multiple nutrients. A number of inborn errors of metabolism have been
described in which clinical improvements can be documented following administration of pharmacologic doses of
thiamin, such as thiamin-responsive megaloblastic anemia
with ringed sideroblasts. The latter is part of a clinical triad
including diabetes mellitus and sensorineural deafness,
associated with an autosomal recessive defect in thiamin
transporter.15
The classic clinical syndromes associated with
deficiency of thiamin include Beriberi or Wernickes
encephalopathy. Beriberi is traditionally classified as dry
or wet form. Dry beriberi is characterized by a symmet
rical peripheral neuropathy with progressive weakness,
muscle wasting, difficulty walking, and ataxia and is accompanied by paresthesia and loss of deep tendon reflex. Wet
beriberi is secondary to cardiomyopathic congestive cardiac
failure and edema. Infantile beriberi is characterized by
shock at 2 to 3 months in a breastfed child with or without
a preceding history of weak cry and poor feeding. It generally occurs in the exclusively breastfed infant whose mother
has a subclinical thiamin deficiency.25 Wernickes encephalopathy is characterized by altered consciousness as well
as the triad of ophthalmoplegia, nystagmus, and ataxia. It is
generally seen in adults with alcohol abuse and malnutrition
although it has been reported in infants and children. 25,26
Thiamin deficiency from feeding of unfortified soy
infant formula due to manufacturing error has been
reported.27, 28 Clinical presentation occurred between 2
and 12 months. Initial manifestations were non-specific
and included vomiting, lethargy, irritability, abdominal
distention, diarrhea, respiratory symptoms, developmental
delay, and failure to thrive. Respiratory and gastrointestinal
infections were noted in many of the reported cases. Classic
manifestation of ophthalmoplegia or death also occurred in
several infants.
Laboratory investigations include abnormal magnetic
resonance imaging at the frontal lobes, basal ganglia, periaqueductal region, thalami, and the mammillary bodies,
while magnetic resonance spectroscopy demonstrates
a characteristic lactate peak.28 Biochemical diagnosis of
thiamin deficiency is by low TPP in erythrocyte and whole
blood29 or by the transketolase activation test. 30 The latter
measures the whole blood or erythrocyte transketolase
2010 A.S.P.E.N. www.nutritioncare.org
Supplementation
Prophylactic use of thiamin supplements may be warranted
in malabsorption disorders. Occasional reports of anaphylactic reaction with parenteral administration of thiamin
has been noted but toxic effects of thiamin excess have not
been studied systematically.1
Source
More than 90% of dietary riboflavin is consumed as a
complex of food protein with FAD and FMN and lesser
amounts as free vitamin and traces of glycosides and esters.
Animal protein (meat, dairy, and eggs) as well as green
vegetables and fortified cereals are abundant sources.4 Riboflavin may be synthesized by colonic bacteria but the extent
of its contribution to human needs is not known. 33
Deficiency State
Riboflavin deficiency is often accompanied by deficiencies of one or more of the other B vitamins. Chronically
limited dietary meat or dairy intake (including infants after
weaning) is a specific risk factor for riboflavin deficiency.
Riboflavin deficiency is also found in protein energy malnutrition states, such as Kwashiorkor and anorexia nervosa,
and in patients with other risk factors that are common to all
water-soluble micronutrients. Symptoms and signs of mild
deficient state can be non-specific. The more characteristic
features of severe deficiency state of ariboflavinosis include
pharyngitis, cheilosis, angular stomatitis, glossitis (magenta
tongue), and seborrheic dermatitis involving nasolabial
folds, flexural area of extremities, and the genital areas.
Diagnosis of riboflavin deficiency is by low erythrocyte riboflavin quantified by high-performance liquid
chromatography and low 24-hour excretion of riboflavin.
Erythrocyte glutathione reductase activity coefficient
from in vitro stimulation by FAD increased in the ranges of
>40%, 20% to 40%, and < 20% are considered as deficient,
low, and acceptable levels of riboflavin status. Biochemical
deficiency has been reported in infants receiving short-term
phototherapy. 36
Supplementation
Excess riboflavin turns urine yellow although there are no
demonstrated functional or structural adverse effects in
vivo after excess riboflavin intake. This is in part because of
the limited absorption of ingested riboflavin. Nevertheless,
it is theoretically possible that riboflavin increases photosensitivity to ultraviolet irradiation and excess riboflavin
63
Source
Unlike most of the water-soluble vitamins, nicotinamide
can be synthesized in the liver and kidney from tryptophan. This process requires adequate amounts of riboflavin,
pyridoxine, and iron and is highly variable depending on
multiple other factors. For instance, tryptophan is used also
for protein synthesis which takes priority over NAD and
NADP synthesis. An estimated average of 60 mg of tryptophan produces 1 mg of niacin or niacin equivalent. However,
dietary intake of niacin is needed to meet the daily requirements. Good sources of niacin include yeast, meats, poultry,
red fish (eg, tuna and salmon), cereals (especially fortified
cereals), legumes, and seeds. Lesser amounts are found in
milk, green leafy vegetables, coffee, and tea. In corn and
wheat, niacin may be bound to sugar molecules as glycosides which significantly decrease niacin bioavailability. 38
64
Deficiency State
Several conditions uniquely predispose to niacin deficiency.
These include carcinoid syndrome in which tryptophan is
preferentially oxidized to 5-hydroxytryptophan and serotonin; prolonged treatment with isoniazid which competes
with the pyridoxine-derived coenzyme required in the
tryptophan-niacin pathway; and Hartnups disease, an autosomal recessive disorder that interferes with the absorption
of tryptophan.
The classic manifestation of severe niacin deficiency is
pellagra, an Italian term meaning rough skin. It is characterized by the triad of diarrhea, dermatitis, and dementia. 39
Gastrointestinal manifestations include glossitis, angular
stomatitis, chelitis, and diarrhea in about 50% of patients.
Skin lesions begin as painful erythema in sun-exposed areas.
Vesicle or bullae formation may occur upon re-exposure to
sun and the skin eventually becomes rough, hard, and scaly.
Hair and nails tend to be spared. Neuropsychiatric manifestations include insomnia, fatigue, nervousness, irritability,
apathy, and memory impairment. Dementia and death may
occur in untreated cases.
Niacin status is determined by a decreased concentration of NAD relative to NADP in erythrocytes, and low
levels of excretion in 24-hour urine niacin and its metabolite N1-methyl-nicotinamide and its 2-pyridone derivative.
Supplementation
Niacin, as a supplement or pharmacologic agent used
primarily in the treatment of hyperlipidemia, can result
in clinical side effects. Both forms of niacin may result
in similar side effects although nicotinamide results in
less vasodilatory effects. Nicotinic acid as low as 30 mg
daily is associated with vasodilatory effects including
flushing, burning, tingling, and itching sensation, and
2010 A.S.P.E.N. www.nutritioncare.org
Vitamin B6
Biochemistry and Physiology
Vitamin B6 comprises a group of 6 related compounds: pyridoxal (PL), pyridoxine (PN), pyridoxamine (PM), and their
respective 5-phosphates (PLP, PNP, and PMP). The major
forms in animal tissues are PLP and PMP; plant-derived
foods contain primarily PN and PNP, sometimes in the
form of a glucoside.
PLP is a coenzyme for a multitude of enzymes involved
in amino acid metabolism including aminotransferases,
decarboxylases, racemases, and dehydratases. It is required
for the conversion of tryptophan to both niacin and the
neurotransmitter serotonin; from homocysteine to cysteine,
dopa to dopamine as well as the synthesis of the inhibitory
neurotransmitter gamma-aminobutyric acid. Pyridoxine
is a coenzyme for -aminolevulinate synthase, the ratelimiting first step in heme synthesis.
Source
Foods rich in pyridoxine include fruits and nuts (bananas,
cantaloupe, walnuts), plants (green leafy vegetables, broccoli, peas, carrots, rice husks, brown rice, maize, wheat germ,
yeast), and animal products (eggs, chicken, fish, beef), as
well as fortified cereals. Microbial synthesis of B6 is possible
but the extent to which its contribution to the physiological
need is not known.
Deficiency State
Isolated deficiency of pyridoxine is rare because its metabolism is intimately involved with multiple other nutrients
including riboflavin, niacin, zinc, and folate. Special considerations for predisposition to lower plasma PLP may include
therapy with isoniazid and L-dopa (react with carbonyl
group of PLP); acetaldehyde but not ethanol decreases
net PLP formation and may compete with PLP for protein
binding. However, the extent to which these situations
increase B6 requirements is not known.
Clinical manifestations of the B6 deficient state
include seizures in infants fed milk formulas without
fortification by pyridoxine from error in the manufacturing41,42 and abnormal electroencephalogram in adults
from experimental B6 deficiency.43 Both are reversed with
reintroduction of B6. Intractable seizures during infancy
and childhood may respond to large doses of pyridoxine and
sometimes only to pyridoxal phosphate. The latter form may
be a result of mutations in the PNPO gene for pyridox(am)
ine 5-phosphate oxidase and present as neonatal epileptic
encephalopathy.44 Other clinical manifestations of B6 deficiency are non-specific and may include microcytic anemia,
glossitis, seborrheic dermatitis, and depression.
Plasma PLP is probably the best indicator of B6 status
because it appears to reflect tissue stores.45 Erythrocyte
aspartate and alanine aminotransferase saturation by PLP
or tryptophan metabolites is also a useful indicator of relative B6 status.
65
Supplementation
High-dose pyridoxine supplement of 2 to 6 g daily for 2 to
40 months46 or chronic intake even at doses < 500 mg/d47
may result in peripheral sensory neuropathy.
Source
Natural sources include fresh green vegetables, liver, yeast,
and some fruits. Folic acid is the only water-soluble vitamin
mandated as part of prenatal supplement for the prevention
of neurotube defects. The generally lower intake of vegetables
and fruits contributes to the potentially inadequate intake
of folate. Significantly improved folate status as indicated
by population survey of red cell and serum folate has been
reported since the Food and Drug Administration (FDA)
in 1998 required the addition of folic acid to all enriched
breads, cereals, flours, corn meal, pasta products, rice, and
other cereal grain products sold in the United States.49
66
derivatives) are hydrolyzed by conjugase enzymes to monoglutamate forms, then enter various cells by membrane
carrier or folate-binding protein-mediated system. The folate
transporter derives from the SLC19 gene family of solute
carriers and shares structural homology with the thiamin
transporter.15 Monoglutamates, mainly 5-methyl-tetrahydrofolate, are metabolized in the liver and other tissues to
polyglutamate derivatives by the enzyme folylpolyglutamate synthetase. Polyglutamates are the forms retained in
various tissues or found in blood or bile and are the forms
needed for function as a coenzyme in single-carbon transfer
reactions.
Folate catabolism involves cleavage of intracellular and
circulatory polyglutamates to the monoglutamate form.
Approximately two-thirds of the folate in plasma is protein
bound and albumin accounts for ~50% of the bound folate.
Folates freely filter through the glomeruli, are secreted,
and reabsorbed by the renal tubules. The bulk of the urine
excretory products are folate cleavage products mainly
in the monoglutamate form. There is extensive enterohepatic circulation of folate. 50 The estimated total body folate
content is between 12 and 28 mg, of which ~50% is in the
liver.
Deficiency State
Patients receiving folate antagonists such as methotrexate
and other drugs that have antifolate activity including
pyrimethamine, trimethoprim, triamterene, trimetrexate,
and sulfasalazine require monitoring for folate status to
ensure adequate dietary intake or folate supplementation. Several inborn errors in folate metabolism (MTHF
reductase deficiency associated with mutations of alleles on
chromosome 1)51 as well as the presence of autoantibody to
folate receptor (cerebral folate deficiency)52 predispose the
affected individual to folate deficiency. Serum and red cell
folate concentration are low in the former and normal in
the latter. In cerebral folate deficiency, the transfer of folate
from plasma to cerebrospinal fluid is low and cerebrospinal
fluid MTHF concentration is low.
Clinical features of folate deficiency may begin during
infancy and include retarded psychomotor development,
poor social contact, decelerating head growth, hypotonia,
ataxia, dyskinesia, irritability, visual and hearing deficiency,
and seizures. Some neurological manifestations may be
reversible with folinic acid supplement. Symptomatology
from anemia may be present.
Deficiency states are confirmed with low red cell folate
which reflects tissue folate store. Plasma folate reflects
concurrent folate balance. Plasma total homocysteine
2010 A.S.P.E.N. www.nutritioncare.org
Supplementation
Excessive folate intake may precipitate or exacerbate
neuropathy in vitamin B12 deficient individuals.
Source
Cobalamin is found in animal foods including meat, fish,
poultry, cheese, milk, and eggs. Cereal and soy milks are
fortified with varied amounts of B12 . Vitamin B12 content
in breast milk is dependent on maternal diet and mature
milk has lower content than colostrum. In the United States
and Canada, both cyanocobalamin and hydroxocobalamin
are available commercially although cyanocobalamin is
most commonly used in supplements and pharmaceuticals.
Vitamin B12 can be synthesized by intestinal bacteria but
its contribution to body pool is not known but likely to be
limited.
67
Deficiency State
Supplementation
Short-term studies up to 4 months indicate that oral B12
supplement has similar efficacy to intramuscular treatment
for B12 deficiency. 59,60 B12 fortification of foods to improve
the populations B12 status55 has been advocated. Intranasal
delivery of B12 is possible but the bioavailability is ~10% of
intramuscular preparation. Periodic parenteral administration of high-dose B12 (1-5 mg) to patients with pernicious
anemia (lack of intrinsic factor) supports the lack of adverse
effects at high doses.
The use of cyanocobalamin is contraindicated in B12
deficient individuals at risk for Lebers optic atrophy, a
genetic disorder caused by chronic cyanide (present in
tobacco smoke, alcohol, and some plants) intoxication,
because the latter may increase the risk of irreversible optic
atrophy. Hydroxocobalamin, a cyanide antagonist, 61 can be
used instead of cyanocobalamin.1
Vitamin C
Biochemistry and Physiology
68
Source
Almost 90% of vitamin C in the typical diet comes from
fruits and vegetables. Citrus fruits, tomatoes, tomato juice,
and potatoes are major sources. Other sources include
brussel sprouts, cauliflower, broccoli, strawberries, cabbage,
and spinach. Vitamin C content of foods can vary with
growing condition, season, stage of maturity, cooking practice, and storage time prior to consumption. 22
Deficiency State
In developed countries, population surveys show serum
vitamin C concentrations are significantly lower in smokers
and low-income persons, presumably reflecting the increased
oxidative stress of smokers and low dietary consumption of
vitamin C foods.63 Clinical manifestation of vitamin C deficiency is quite rare but has been reported in children with
severely restricted diets related to food faddism, psychiatric
or developmental problems, 64 or in children with end-stage
organ disease with severely compromised nutrition.65 It
also has been reported in young children who ingest only
well-cooked foods, few fruits and vegetables, and are supplemented with ultra heat-processed milk.66
The primary clinical manifestation of vitamin C deficiency is scurvy and reflects deterioration of elastic tissues.
Clinical features include follicular hyperkeratosis, petechiae, ecchymoses, coiled hairs, inflamed and bleeding
gums, perifollicular hemorrhages, and impaired wound
healing. Refusal to walk, with or without joint effusions and
arthralgia, dyspnoea, edema, weakness, fatigue, depression and Sjogren syndrome (dry eyes and mouth) also can
occur. In experimental subjects, gingival inflammation and
fatigue were among the most sensitive markers of deficiency
without being frankly scorbutic. Impaired bone growth,
disturbed ossification, and subperiosteal hemorrhage may
be present. Scurvy usually occurs at a plasma concentration
of < 0.2 mg/dL (11 mol/L) although leukocyte ascorbate concentration is considered a better measure of tissue
reserve than plasma ascorbate concentration and is the
preferred indicator of vitamin C status.67
Supplementation
Gastrointestinal disturbances such as nausea, abdominal
cramps, and diarrhea have been reported at vitamin C
intake of > 3 g/d probably as a result of the osmotic effect
of unabsorbed vitamin C.68 In vivo data do not clearly show
a causal relationship between excess vitamin C intake by
apparently healthy individuals and other adverse effects2
although increased urine oxalate excretion and development
of oxalate stones may be possible. However, individuals with
hemochromatosis, glucose-6-phosphate dehydrogenase
deficiency, and renal disorders may be more susceptible to
adverse effects of excess vitamin C intake, which include
excess iron absorption, pro-oxidant effects, and kidney
(oxalate) stone formation. There is also risk for reduced
vitamin B12 and copper levels, increased oxygen demand,
dental enamel erosion, allergic response,2 and blocking the
action of warfarin.6 Vitamin C intake of 250 mg/d or higher
has been associated with false negative result for stool and
gastric occult blood.69 High-dose vitamin C supplement
should be stopped before these laboratory tests.
69
pathway end products, CoA and acyl-CoA. CoA is hydrolyzed to pantothenic acid in a multiple-step reaction and is
excreted intact in the urine in proportion to dietary intake.
Deficiency State
Deficiency has been reported only with feeding semisynthetic diets70 or an antagonist to the vitamin, -methyl
pantothenic acid.71 Clinical manifestations may be nonspecific and include irritability, restlessness, fatigue, apathy,
malaise, sleep disturbances, nausea, vomiting and abdominal cramps, numbness, paresthesia, and staggering gait, and
increased sensitivity to insulin. Historically, pantothenic
acid was implicated in the burning feet syndrome that
affected prisoners of war in Asia during World War II.72
There are no data to support the use of whole blood,
plasma, or red cell concentrations as a marker of deficiency
state. Urinary excretion of pantothenic acid is strongly
dependent on dietary pantothenic acid.70
Supplementation
Source
Pantothenic acid is widely distributed in foods and found
in high concentrations in organ meats, yeast, egg yolk, fresh
vegetables, whole grains, and legumes. Intestinal microflora can synthesize pantothenic acid but its contribution in
humans has not been quantified.
Source
Biotin is widely distributed in natural food stuffs but its
concentration varies substantially with liver having the
highest content; fruits and most meats contain small
amounts. The contribution of biotin synthesized by intestinal microflora to the bodys needs has not been defined.
70
Deficiency State
Clinical deficiency of biotin is uncommon but has been
reported in individuals who consumed raw egg whites over
long periods75 or received total parenteral nutrition before
biotin supplementation.76 Infantile seizures either alone or
with other neurological or cutaneous findings, particularly
in the presence of ketolactic acidosis and organic aciduria,
are typical of biotinidase deficiency.77 Infants manifest
rash around the mouth, nose, and eyes as biotin deficiency
facies after several months of biotin-free total parenteral
nutrition. This rash may extend to ears and perineal orifices.
Alopecia totalis, hypotonia, lethargy, developmental delay,
and withdrawn behavior also may occur. Similar manifestations including ataxia and paresthesia may occur in older
children and adults. The list of inborn errors of metabolism
that result in biotin dependency and various degrees of
neurological and dermatologic abnormalities now extends
to holocarboxylase synthetase deficiency and a defect in
biotin transport.78
Biochemically, urine excretion of biotin is decreased
and 3-hydroxyisovaleric acid is increased.79 Plasma biotin
is not a sensitive indicator of inadequate biotin intake.
Reduced expression of SLC19A3, a potential biotin transporter, in leukocytes may prove to be a useful indicator of
marginal biotin deficiency.80
Biotinidase deficiency results in a relative biotin deficiency through lack of adequate digestion of protein-bound
biotin. It is treated with free (unbound) biotin at the estimated typical dietary intake of 50 to 150 mcg/d.81
Supplementation
Intakes of biotin at > 10 mg/d in a variety of subjects at
different life stages was not associated with documented
adverse effects.
Choline
Biochemistry and Physiology
Choline is the major source of methyl groups in the diet. It is
a precursor for acetylcholine, phospholipids, and the methyl
donor betaine. Functionally, it plays a critical role in the
structural integrity of cell membranes, methyl metabolism,
cholinergic neurotransmission, transmembrane signaling,
and lipid and cholesterol transport and metabolism.
Source
Choline in the diet is available as free choline or is bound as
water-soluble (phosphocholine, glycerophosphocholine) or
lipid soluble (sphingomyelin, phosphotidylcholine) esters.
It is widely distributed in foods with most in the form of
phosphatidylcholine in membranes. Foods especially rich
in choline compounds are milk, liver, eggs, and peanuts.
Lecithin, a phosphatidylcholine-rich fraction prepared
during commercial purification of phospholipids, is often
used interchangeably with phosphatidylcholine, and is
frequently added to food as an emulsifying agent and may
be a significant source of choline.
Endogenous synthesis of choline occurs via sequential
methylation of phosphatidylethanolamine catalyzed by the
enzyme phosphatidylethanolamine N-methyltransferase
and with S-adenosylmethionine as the methyl donor.
However, it is insufficient to compensate for the lack of
dietary choline.
All tissues accumulate choline by diffusion and mediated transport and a specific carrier mechanism transports
free choline across the blood-brain barrier at a rate proportional to the serum choline concentration. The liver and
kidney also readily take up choline where a large proportion
is oxidized to betaine. The methyl groups of betaine can be
scavenged and reused in single-carbon metabolism.
Deficiency State
Though many foods contain choline, there is at least a
twofold variation in dietary intake in humans. When
deprived of dietary choline, most men and postmenopausal
women developed signs of organ dysfunction (fatty liver
or muscle damage), while less than half of premenopausal
women developed such signs.82 Individuals receiving
total parenteral nutrition devoid of choline but adequate
for methionine and folate developed hepatic steatosis
and elevated alanine aminotransferase. These abnormalities resolved in some individuals when a source of dietary
choline was provided.83 In addition to the gender influence,
there also appears to be genetic polymorphism and epigenetics influence for susceptibility to choline deficiency. 82
Fasting plasma, erythrocyte, and tissue content of choline
and phosphatidylcholine can be used as markers of choline
status.
Supplementation
The critical adverse effect from high intake of choline is
hypotension, with corroborative evidence on cholinergic
side effects (eg, sweating and diarrhea, and fishy body
odor).1
71
References
72
73
66. Ratanachu-Ek S, Sukswai P, Jeerathanyasakun Y, Wongtapradit L. Scurvy in pediatric patients: a review of 28 cases. J
Med Assoc Thai. 2003;86:S734740.
67. Thurnham DI. Micronutrients and immune function: some
recent developments. J Clin Pathol. 1997;50:887891.
68. Wandzilak TR, DAndre SD, Davis PA, Williams HE. Effect
of high dose vitamin C on urinary oxalate levels. J Urol.
1994;151:834837.
69. Gogel HK, Tandberg D, Strickland RG. Substances
that interfere with guaiac card tests: implications for gastric aspirate
testing. Am J Emerg Med. 1989;7:474480.
70. Fry PC, Fox HM, Tao HG. Metabolic response to a pantothenic acid deficient diet in humans. J Nutr Sci Vitaminol (Tokyo).
1976;22:339346.
71. Hodges RE, Bean WB, Ohlson MA, Bleiler R. Human pantothenic acid deficiency produced by omega-methyl pantothenic
acid. J Clin Invest. 1959;38:14211425.
72. Glusman M. The syndrome of burning feet (nutritional
melagia) as a manifestation of nutritional deficiency. Am J
Med. 1947;3:211223.
73. Wolf B, Grier RE, Secor McVoy JR, Heard GS. Biotinidase
deficiency: a novel vitamin recycling defect. J Inherit Metab
Dis. 1985;8 (suppl 1):5358.
74. Mock DM, Heird GM. Urinary biotin analogs increase in
humans during chronic supplementation: the analogs are
biotin metabolites. Am J Physiol. 1997;272:E8385.
75. Baugh CM, Malone JH, Butterworth CE Jr. Human biotin
deficiency. A case history of biotin deficiency induced by
raw egg consumption in a cirrhotic patient. Am J Clin Nutr.
1968;21:173182.
76. Mock DM, Baswell DL, Baker H, Holman RT, Sweetman
L. Biotin deficiency complicating parenteral alimentation:
diagnosis, metabolic repercussions, and treatment. J Pediatr.
1985;106:762769.
77. Wolf B, Heard GS, Weissbecker KA, McVoy JR, Grier RE,
Leshner RT. Biotinidase deficiency: initial clinical features
and rapid diagnosis. Ann Neurol. 1985;18:614617.
78. Mardach R, Zempleni J, Wolf B, et al. Biotin dependency due to a
defect in biotin transport. J Clin Invest. 2002;109:16171623.
79. Mock NI, Malik MI, Stumbo PJ, Bishop WP, Mock DM.
Increased urinary excretion of 3-hydroxyisovaleric acid
and decreased urinary excretion of biotin are sensitive early
indicators of decreased biotin status in experimental biotin
deficiency. Am J Clin Nutr. 1997;65:951958.
80. Vlasova TI, Stratton SL, Wells AM, Mock NI, Mock DM.
Biotin deficiency reduces expression of SLC19A3, a potential
biotin transporter, in leukocytes from human blood. J Nutr.
2005;135:4247.
81. Wolf B, Heard GS, McVoy JR, Raetz HM. Biotinidase deficiency: the possible role of biotinidase in the processing of
dietary protein-bound biotin. J Inherit Metab Dis. 1984;7
(suppl 2):121122.
82. Zeisel SH. Gene response elements, genetic polymorphisms
and epigenetics influence the human dietary requirement for
choline. IUBMB Life. 2007;59:380387.
83. Buchman AL, Dubin MD, Moukarzel AA, et al. Choline
Fat-Soluble Vitamins
Winston Koo, MBBS, FACN, CNS, May Saba, PharmD, BCNSP, Mirjana Lulic-Botica, BSc, BCPS, and Judith Christie, RN, MSN
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 74
Vitamin A. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .76
Biochemistry and Physiology
Sources
Absorption and Metabolism
Deficiency
Adverse Effects
Vitamin D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
Biochemistry and Physiology
Sources
Absorption and Metabolism
Deficiency
Adverse Effects
Vitamin E. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
Biochemistry and Physiology
Sources
Absorption and Metabolism
Deficiency
Adverse Effects
Vitamin K . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Biochemistry and Physiology
Sources
Absorption and Metabolism
Deficiency
Adverse Effects
74
Learning Objectives
Introduction
FAT-SOLUBLE VITAMINS
Vitamin A
mcg RAE
(1 mcg = 3.3
IU)
VITAMIN D*
IU (1 mcg =
40 IU)
VITAMIN E
mg
VITAMIN K*
mcg
06 mo*
712 mo*
13 y
48 y
913 y
MALES
1418 y
FEMALES
1418 y
PREGNANCY
1418 y
LACTATION
1418 y
UL
400 (600)
500 (600)
300 (600)
400 (900)
600 (1700)
400 (1000)
400 (1000)
400 (2000)
400 (2000)
400 (2000)
4
5
6 (200)
7 (300)
11 (600)
2.0
2.5
30
55
60
900 (2800)
400 (2000)
15 (800)
75
700 (2800)
400 (2000)
15 (800)
75
750 (2800)
400 (2000)
15 (800)
75
19 (800)
75
NA
75
76
Vitamin A
Biochemistry and Physiology
Vitamin A, or retinoids, refers to retinol and its derivatives
including synthetic analogs that have the same -ionone
ring and similar biological activities. They also include
provitamin A carotenoids that are the dietary precursor of
retinol. The all-trans isomer is the most common and stable
form although many cis isomers also exist.
Retinoids are critical to the maintenance of proper
vision in the signal transmission of images to the visual
cortex. They are necessary for cell differentiation and integrity of epithelial cells throughout the body including normal
differentiation of the cornea and conjunctival membranes,
as well as for the photoreceptor rod and cone cells of the
retina. Other critical functions include maintenance of
immune function, expression of various genes that encode
for structural- and extracellular matrix-proteins, enzymes,
RBPs and receptors, and embryonic development of hindbrain, eyes, ears, heart, and limbs. Provitamin A carotenoids
must be converted to retinoids to exert vitamin A function.
In addition, carotenoids are excellent antioxidants and may
have other biological properties unrelated to their vitamin
A activity.
Sources
Vitamin A is present in the diet as retinyl esters derived
almost exclusively from animal sources (liver and fish
liver oils, dairy products, kidney, and eggs). Most of the
vitamin A in breast milk is in the form of retinyl palmitate
in milk fat and is in greatest concentration in colostrum
and transition milk. The vitamin A content of breast milk
is generally lower in vitamin A deficient populations but is
still sufficient to prevent subclinical deficiency in exclusively
breastfed infants during the first 6 months. 8 Cows milk has
relatively lower vitamin A content than human milk and
averages ~40 International Units (IU)/g fat.9 The U.S. Food
and Drug Administration encourages dairy producers to
fortify reduced fat milk to 2000 IU/L.9 Infant formulas are
fortified with vitamin A at 250 to 750 IU/100 kcal.10 Hightemperature sterilization of milk increases isomerization
of trans- to cis-retinol by as much as 34%. 3 Provitamin A
carotenoids (mainly -carotene) are distributed widely in
2010 A.S.P.E.N. www.nutritioncare.org
FAT-SOLUBLE VITAMINS
Deficiency
Vitamin A deficiency is an endemic nutrition problem
throughout much of the developing world and affects the
77
health and survival of infants, young children, and pregnant and lactating women. These life-stage groups represent
periods when both nutrition stress is high and the diet is
likely to be chronically deficient in vitamin A. There are
numerous clinical effects of vitamin A deficiency (Table
8-2) but the most specific manifestation is xerophthalmia.
In developed countries, risk factors for vitamin A deficiency
include fat malabsorption conditions, strict vegan diets,
fad diets, and severely restricted preformed- or provitamin
A. Ethanol decreases liver vitamin A stores by increasing
release of hepatic retinol independent of dietary intake
and decreasing conversion of -carotene to retinol, thus
promoting vitamin A deficiency. 3
Table 8-2 Clinical Effects of Vitamin A Deficiency3,19,20
Xerophthalmia*
XN: Impaired dark adaptation (from slowed regeneration of
rhodopsin) to night blindness
XCj: Conjunctival dryness (xerosis), XB: keratinization (Bitots spot)
XCo: Corneal xerosis, XU/K: Corneal ulceration and/or keratomalacia
of any part, XSc: Corneal scar in central part, XSp: Corneal scar
in peripheral part
XF: Xerophthalmia fundus
Generalized dysfunction of cellular and humoral immunity
withincreased morbidity and mortality especially from
measlesanddiarrhea
Impairs iron mobilization from stores
Defects in lung function
Follicular hyperkeratosis
* Classification according to reference 19.
78
Adverse Effects
There are substantial data on the adverse effects of high
vitamin A intake.18,23 Individuals with pre-existing liver
disease, hyperlipidemia, severe protein malnutrition, or
high alcohol intake, may be distinctly susceptible to the
adverse effects of preformed vitamin A intake. Short-term
large doses and even a single dose > 150,000 mcg in adults24
and proportionately lower in children can result in acute
toxicity characterized by nausea, vomiting, headache,
vertigo, blurred vision, muscular incoordination, and raised
intracranial pressure. In infants, there is also bulging of
fontanels.18,25 Animal studies demonstrated that a similarly
large single dose of vitamin A can be lethal.26
Chronic toxicity is usually associated with ingestion of
large doses > 30,000 mcg/day for months or years. Clinical
manifestations of chronic toxicity are varied and non-specific
and may include irritability, anorexia, skin desquamation,
and liver abnormalities including elevated liver enzymes,
fibrosis, and cirrhosis. In infants, additional non-hepatic
manifestations include bulging fontanel, craniotabes, and
laboratory findings of cortical hyperostosis, hypercalcemia,
and hyperphosphatemia, and metastatic calcifications may
occur at an intake between 5,500 to 6,750 mcg/d for 1 to
2010 A.S.P.E.N. www.nutritioncare.org
Vitamin D
Biochemistry and Physiology
Vitamin D (calciferol) refers to two fat-soluble seco-sterols:
cholecalciferol (vitamin D3) photosynthesized in the skin of
vertebrates, and ergocalciferol (vitamin D2) from the yeast
and plant sterol. The term vitamin D without the subscript
is frequently used generically to describe vitamins D2 and
D3 and, correspondingly, their metabolites. The parent
vitamin D compounds are biologically inert and require
two obligatory hydroxylation steps primarily in the liver
and kidney to 25 hydroxyvitamin D (25 OHD) and 1,25
dihydroxyvitamin D (1,25 (OH)2D), respectively. Quantitatively, 25 OHD is the major circulating metabolite. It is a
good marker of vitamin D status as it reflects the cumulative
FAT-SOLUBLE VITAMINS
Sources
Endogenous synthesis of vitamin D3 requires exposure
to sunlight or irradiation in the ultraviolet B range. It is
extremely effective and a 10- to 15-minute whole-body
exposure to peak summer sun will generate and release up
to 20,000 IU vitamin D3 into the circulation. 38,39 However,
prolonged sunlight exposure increases the conversion of
previtamin D3 to inactive metabolites. Cutaneous production of vitamin D3 is substantially diminished by decreased
exposure to sunlight from seasonal changes, or time of day,
clouds, aerosols, thick ozone, higher latitude, aging, clothing,
sunscreen use, and melanin pigmentation.40 In dark-skinned
79
80
25 OHD is 10 days to 3 weeks.46 Subsequent hydroxylation at the 1-carbon position to 1,25 (OH)2D in the kidney
is tightly regulated, principally through the action of parathyroid hormone in response to calcium and phosphorus
levels. The half-life of 1,25 (OH)2D is ~4 to 6 hours.47 It is
possible that activated macrophages, some lymphoma cells,
and cultured skin and bone cells also make 1,25 (OH)2D
and exert paracrine or autocrine actions. Excessive unregulated production of 1,25 (OH)2D by activated macrophages
and lymphoma cells is responsible for hypercalciuria and
hypercalcemia.
Enterohepatic circulation of vitamin D and its metabolites is present but probably has a limited role in their
conservation.48 Further hydroxylation at the 24-carbon of
25 OHD and 1,25 (OH)2D is the initial step in the metabolic degradation of these 2 vitamin D metabolites, with the
major metabolite (calcitroic acid) excreted in the urine.49
Deficiency
Clinical deficient states can result from lack of cutaneous
production of vitamin D3 with inadequate intake, impaired
absorption or metabolism of vitamin D to its active form
1,25 (OH)2D, or impaired recognition of 1,25 (OH)2D by
its receptor. Drugs such as glucocorticoids inhibit vitamin
D dependent calcium absorption. Phenobarbital and
phenytoin can alter the metabolism and circulating halflife of vitamin D metabolites. Chronic therapy with these
medications predisposes patients to the effects of vitamin
D deficiency.
Vitamin D deficiency results in inadequate mineralization, or demineralization, of the skeleton with occurrence
of rickets in the developing skeleton and osteomalacia in
adults. Rickets is characterized by widening at the ends of
long bones, rachitic rosary, deformations include bowed
legs and knock-knees, and frontal bossing of the skull.
In addition, secondary hyperparathyroidism occurs as a
homeostatic response to prevent a decrease in circulating
ionized calcium from vitamin D deficiency. Parathyroid
hormone mobilizes calcium from the skeleton resulting in
porotic bones, and conserves renal calcium but increases
phosphorus excretion as reflected by low or absent urine
calcium and increased urine phosphorus. Thus, vitamin D
deficiency is characterized by a low circulating concentration of 25 OHD, normal fasting circulating calcium with
low or low normal phosphorus, and elevated parathyroid
hormone, although hypocalcemia can occur in chronic
vitamin D deficiency. Increased bone turnover with
vitamin D deficiency is reflected by elevated bone alkaline
phosphatase, collagen, and non-collagenous bone proteins
2010 A.S.P.E.N. www.nutritioncare.org
Adverse Effects
Vitamin D toxicity from prolonged exposure to natural
sunlight has not been reported. Increased sunlight exposure
leads to a decrease in endogenous production of vitamin
D3 by conversion of previtamin D3 to inactive metabolites,
inhibition of 25-hydroxylase enzymes results in decreased
production of 25 OHD3, and stimulation of 24-hydroxylase enzyme results in increased metabolic degradation of
vitamin D metabolites. These processes contribute to the
prevention of hypervitaminosis D and vitamin D toxicity.
Excessive vitamin D intake from supplements can
result in hypervitaminosis D and is characterized by a
considerable increase in plasma 25 OHD in the range of
400 to 1250 mol/L (160500 ng/mL). 51,52 Other vitamin
D metabolites are also elevated although the increase in
1,25 (OH)2D is generally elevated to a much lower extent. 52
Clinical toxicity is manifested through hypercalcemia and
its multiple debilitating effects include polyuria, polydipsia,
depression, and metastatic calcification of soft tissues
including the kidney, blood vessels, heart, and lung. These
effects generally are seen with chronic supplementation in
excess of 20,000 IU daily53 although toxicity can occur at
lower doses. Toxicity from chronic intake of ergocalciferol
appears to require significantly higher doses. 54 It is possible
that this lower toxicity may be a result of its relatively low
bioactivity compared to cholecalciferol.
FAT-SOLUBLE VITAMINS
Vitamin E
Biochemistry and Physiology
There are 8 naturally occurring forms of vitamin E with
4 (-, -, -, and -) tocopherols, all in the RRR- form,
and 4 (-, -, -, and -) tocotrienols. The RRR- form of
-tocopherol is maintained in human plasma and occurs
naturally in foods. Eight different stereoisomers in equal
amounts are found in synthetic vitamin E (all rac-tocopherol): RRR-, RSR-, RRS-, RSS-, SRR-, SSR-, SRS-,
and SSS- forms with structural differences at the side chain
and at the ring/tail junction.
The various forms of vitamin E are not interconvertible in the human. The plasma concentration and biological
activity of different forms of vitamin E are dependent
primarily on the affinity of -tocopherol transfer protein
(-TTP) for them. RRR--tocopherol has the highest
affinity for -TTP. Other forms of vitamin E stereoisomers
have much lower binding affinity to -TTP.2 They may have
some biological functions55,56 and their intake are reported
as -tocopherol equivalent.2
Vitamin E functions primarily as a non-specific chainbreaking antioxidant acting as a peroxyl radical scavenger
that prevents the propagation of free radical reaction and
lipid peroxidation. 57 Vitamin E also may play important
roles in cell proliferation and differentiation, cell adhesion
and arachidonic acid cascade through inhibition of protein
kinase C activity, downregulating expression of intercellular cell adhesion molecule (ICAM-1) and vascular cell
adhesion molecule (VCAM-1), and upregulating expression of cytosolic phospholipase A 2 and cyclooxygenase-1.2
Current recommendations for vitamin E intake include
only the 2R-stereoisomeric forms of -tocopherol (RRR-,
RSR-, RRS-, RSS-) since they are maintained in human
plasma or tissue. Data on intakes from current surveys and
nutrient content of foods are presented as -tocopherol
equivalents (-TE) and include all 8 naturally occurring
forms of vitamin E, after adjustment for bioavailability
using previously determined equivalency. For example,
-tocopherol is estimated to have only 10% of the bioactivity of -tocopherol.
Based on the NHANES III (Third National Health and
Nutrition Examination Survey 1988 to 1994) data, ~80%
of the -TE from foods is contributed by foods containing
-tocopherol. Thus, the intake of -TE is usually greater
than the intake of -tocopherol alone.2 Milligrams of
-tocopherol in a meal including fortified food or multivitamins is based on the following conversion factors:
81
Sources
The main dietary sources of vitamin E are edible vegetable
oils. All of the -tocopherol present in natural foods is in the
RRR--tocopherol form. Oils from wheat germ, sunflower,
safflower, canola, olive, and cottonseed contain > 50% of the
tocopherol as -tocopherol. Soybean and corn oils contain
~10 times as much -tocopherol as -tocopherol. Palm and
rice bran oils contain high proportions of -tocopherol,
as well as various tocotrienols. Meat, poultry, fish, dairy
products, nuts, grains and fruit, especially the oils or fatty
portions, are other sources of -tocopherol. 2 Colostrum and
breast milk are rich sources of vitamin E. 58 Infant formulas
are fortified with a minimum of 0.7 IU/100 kcal.10 Infant
formulas fortified with the natural form of -tocopherol have
better bioavailability compared to the synthetic forms. 59
Estimation of dietary intake of vitamin E is difficult
because the source of oil is not always known with certainty
and most nutrient databases, as well as nutrition labels, do
not distinguish among the different tocopherols in food.
Food preparation, such as deep frying using vegetable oil,
leads to chemical structure modification and may result
in functional discrepancy. 56 Vitamin E esters usually as
acetate of either natural RRR- or the synthetic mixture (all
rac-) of -tocopherol are used in food fortification, supplements, and pharmacological agents. Vitamin E supplements
are available alone or as multivitamins in oral preparation.
Vitamin E is available in parenteral form only as part of a
multivitamin.11 A water-miscible formulation of vitamin E,
tocopheryl polyethylene glycol succinate 1000 (TPGS), is
absorbed, metabolized, and reverses signs of vitamin E deficiency in children60 with severe fat malabsorption.
82
Deficiency
Vitamin E requirements increase with increased intake of
polyunsaturated fatty acids, high-level physical activity, and
cigarette smoking. However, the extent of increased requirement and the compensatory effects of other antioxidants
are not defined. Intake of plant phenolic compounds and
2010 A.S.P.E.N. www.nutritioncare.org
Adverse Effects
All forms of vitamin E are absorbed and could contribute
to vitamin E toxicity although not all forms are maintained
in plasma. Normal adults appear to tolerate oral tocopherol
intake of 100 to 800 mg/d67 but adverse effects from the
FAT-SOLUBLE VITAMINS
83
Vitamin K
Sources
Vitamin K content in most foods is very low (< 10 mcg/
100 g), and the majority is obtained from a few leafy green
vegetables and 4 vegetable oils (soybean, cottonseed, canola,
and olive) that contain high amounts. Hydrogenation of
plant oils to form solid shortenings results in some conversion of phylloquinone to 2,3-dihydrophylloquinone but
it is uncertain whether these forms are metabolically and
functionally identical. These forms of vitamin K are most
Deficiency
Individuals at risk for vitamin K deficiency include those
with severe fat malabsorption conditions (eg, hepatobiliary
disorders) or after bariatric surgery, especially those with
severe diet restriction. Prolonged use of broad-spectrum
antibiotics may decrease vitamin K synthesis by intestinal
bacteria. Long-term differences in dietary vitamin K intake
modulate the response to coumarin anticoagulants, which
act by blocking the vitamin K recycling. Elevated intake of
vitamin E antagonizes vitamin K action probably through
its effect on vitamin K absorption and metabolism.2,74
The classic sign of vitamin K deficiency is an increase in
2010 A.S.P.E.N. www.nutritioncare.org
84
Adverse Effects
Vitamin K toxicity is rare. Parenteral administration of a
large amount of water-soluble synthetic vitamin K (vitamin
K 3) has been associated with hemolytic anemia, hyperbilirubinemia, and kernicterus4 and with liver damage. 3 No
adverse effects associated with other vitamin K supplements
or from food have been reported in healthy individuals
although high dietary intake or supplemental vitamin K can
inhibit the anticoagulation effect of vitamin K antagonists.
References
FAT-SOLUBLE VITAMINS
85
21. Wasantwisut E. Application of isotope dilution technique in vitamin A nutrition. Food Nutr Bull. 2002; 23(3
Suppl):103106.
22. World Health Organization, United Nations Childrens Fund,
VACG Task Force. Vitamin A Supplements: A Guide to Their
Use in the Treatment and Prevention of Vitamin A Deficiency and
Xerophthalmia. 2nd ed. Geneva: World Health Organization;
1997. http://www.who.int/nutrition/publications/micronutrients/vitamin_a_deficieny/9241545062/en/index.html.
Accessed July 25, 2009.
23. Penniston KL, Tanumihardjo SA. The acute and chronic toxic
effects of vitamin A. Am J Clin Nutr. 2006;83:191201.
24. Bendich A, Langseth L. Safety of vitamin A. Am J Clin Nutr.
1989;49:358371.
25. Persson B, Tunell R, Ekengren K. Chronic vitamin A intoxication during the first half year of life; description of 5 cases.
Acta Paediatr Scand. 1965;54:4960.
26. Macapinlac MP, Olson JA. A lethal hypervitaminosis A
syndrome in young monkeys (Macacus fascicularis) following
a single intramuscular dose of a water-miscible preparation containing vitamins A, D2 and E. Int J Vitam Nutr Res.
1981;51:331341.
27. Leo MA, Lieber CS. Alcohol, vitamin A, and beta-carotene:
adverse interactions, including hepatotoxicity and carcinogenicity. Clin Nutr. 1999;69:10711085.
28. Krasinski SD, Russell RM, Otradovec CL, et al. Relationship
of vitamin A and vitamin E intake to fasting plasma retinol,
retinol-binding protein, retinyl esters, carotene, alpha-tocopherol, and cholesterol among elderly people and young adults:
increased plasma retinyl esters among vitamin A-supplement
users. Am J Clin Nutr. 1989;49:112120.
29. World Health Organization, The Micronutrient Initiative. Safe Vitamin A Dosage During Pregnancy and Lactation.
Geneva: World Health Organization; 1998. http://www.who.
int/nutrition/publications/micronutrients/vitamin_a_deficieny/WHO_NUT_98.4/en/index.html. Accessed July 25,
2009.
30. Lascari AD. Carotenemia. A review. Clin Pediatr.
1981;20:2529.
31. Bendich A. The safety of beta-carotene. Nutr Cancer.
1988;11:207214.
32. Black HS. Reassessment of a free radical theory of cancer with
emphasis on ultraviolet carcinogenesis. Integr Cancer Ther.
2004;3:279293.
33. Haddad JG Jr, Hahn TJ. Natural and synthetic sources
of circulating 25-hydroxyvitamin D in man. Nature.
1973;244:515517.
34. Jurutka PW, Bartik L, Whitfield GK, et al. Vitamin D receptor:
key roles in bone mineral pathophysiology, molecular mechanism of action, and novel nutritional ligands. J Bone Miner Res.
2007;22 Suppl 2:v210.
35. Haussler MR, Haussler CA, Bartik L, et al. Vitamin D receptor:
molecular signaling and actions of nutritional ligands in
disease prevention. Nutr Rev. 2008;66 (10 Suppl 2):S98112.
36. Gordon CM, Williams AL, Feldman HA, et al. Treatment of
hypovitaminosis D in infants and toddlers. J Clin Endocrinol
Metab. 2008;93:27162721.
86
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Fluid Distribution. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Fluid Regulation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
Hypervolemia
Hypovolemia
Potassium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
Hyperkalemia
Hypokalemia
Magnesium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Hypermagnesemia
Hypomagnesemia
Calcium. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
Hypercalcemia
Hypocalcemia
Phosphorus. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
Hyperphosphatemia
Hypophosphatemia
Learning Objectives
Background
Water is the most abundant and probably the most important substance in the body. It is essential for digestion,
absorption, transport, and utilization of nutrients as well as
the excretion of waste products. The regulation of fluids and
electrolyte balance is maintained by an elaborate system
of interrelated regulatory systems to ensure proper cell
function. Unfortunately, fluid and electrolyte imbalances
are common, and when severe, these imbalances can have
a detrimental effect on major organ systems. This chapter
will focus on fluid and electrolyte homeostasis as well as
commonly encountered fluid and electrolyte abnormalities
and the treatment of these disorders.
Fluid Distribution
88
volume would increase as would the ECF and ICF osmolality. However, the ICF volume would decrease due to the
fluid shift caused by the hypertonic fluid. The volume of the
ECF increase would be determined by the tonicity and the
volume of the hypertonic solution given.7
The balance between the ECF and ICF is important in
fluid homeostasis, but these are not the only compartments
that must be maintained. The ECF components, the intravascular space and the interstitial space, are maintained
by Starling forces. Starling forces consist of plasma oncotic
pressure and hydrostatic pressure. When fluid moves from
the plasma to the interstitial space, edema occurs.9 As
vascular permeability increases, albumin leaks from the
plasma to the interstitial space. This capillary leak causes a
reduction in plasma oncotic pressure which in turn causes
fluid to move from the plasma to the interstitial space. If this
happens quickly, and the plasma volume is not replaced,
intravascular volume depletion can occur resulting in
hypotension and poor perfusion.
Fluid Regulation
Table 9-1 Distribution of Total Body Water as a Percent of Total Body Weight
Age/Life Stage
Intracellular
Extracellular
Transcellular
Premature infant
80%
52%
2.5%
3-month-old infant
70%
46%
6-month-old infant
60%
39%
Male 59%
Female 57%
50%
38%
36%
32%
26% Intravascular 6%
Interstitial 19%
22% Intravascular 5.5%
Interstitial 16.5%
19% Intravascular 5%
Interstitial 14%
18% Intravascular 4.5%
Interstitial 13.5%
16% Intravascular 4%
Interstitial 12%
Elderly patient
2%
1.8%
1.7%
1.5%
Hypervolemia
Hypervolemia, or increased TBW, causes a decreased
serum osmolarity resulting in dilute urine by suppressing
the levels of circulating antidiuretic hormone (ADH). The
precise mechanism by which plasma osmolarity suppresses
ADH is unclear, but it probably is related to specialized cells
that sense osmolarity changes and send messages to the
neuroendocrine cells located in the hypothalamus or the
organum vasculosum.17 In most cases, the thirst response
will be suppressed, ADH will be suppressed, and the excess
water will be excreted by the kidneys. However, in conditions where the low plasma osmolarity fails to inhibit
ADH secretion (such as in severe low-output congestive
heart failure), cell expansion, hypervolemia, and hyponatremia continue to progress.18 Symptoms include headache,
nausea, vomiting, muscle twitching, convulsion, and if
severe, death.19 Other conditions that may result in hypervolemia include kidney or liver failure with ascites, sepsis,
cardiac failure, and syndrome of inappropriate antidiuretic
hormone (SIADH).
Hypovolemia
Hypovolemia is a condition where TBW is decreased significantly enough to result in symptoms. The body can correct
the problem by stimulating the thirst response, increasing
ADH release, or both. When the plasma osmolarity is
increased or when the blood volume or pressure is reduced,
the thirst response is stimulated.17 In hypovolemia, the
serum osmolarity increases and the blood volume decreases,
which results in the release of ADH. The presence of ADH
89
Fluid Considerations/Requirements
inChildren
Fluid Requirements
10 kg
> 10 kg to
20 kg
> 20 kg
100 mL/kg
1000 mL + 50 mL/kg for
wt > 10 kg
1500 mL + 20 mL/kg for
wt > 20 kg
4 mL/kg/h
40 mL/h + 2 mL/kg/h
> 10 kg
60 mL/h + 1 mL/kg
> 20 kg
There are a number of conditions that require adjustments in fluid intake in order to provide optimal care. After
birth, a contraction of the ECF compartment takes place due
to the loss of interstitial fluid, which results in a 5% to 10%
weight loss in healthy neonates, possibly more in premature
infants.2631 Prematurity affects fluid balance. Premature
infants may require as much as 200 mL/kg/d to maintain
fluid balance due to their large insensible fluid losses. These
losses are due partially to the large skin-to-body surface area
ratio and partially to the immaturity of the skin which leads
to increased evaporative fluid loss. 32,33 In addition, phototherapy and radiant warmers increase water losses, often as
much as 20 to 40 mL/kg/d. 3437 Insensible fluid losses may
remain greater than 100 mL/kg/d for weeks in neonates
2010 A.S.P.E.N. www.nutritioncare.org
90
Electrolyte Assessment
Reference values for normal serum electrolyte concentrations based on age appear at the beginning of each
electrolyte section below. These values are included to
illustrate the differences in electrolyte concentrations for
various age groups. Laboratory reference values will vary
from institution to institution, and practitioners should use
the reference values listed at their individual institutions for
adjusting serum electrolyte values.
Sodium
Preterm:
Older Infants:
Children and Adolescents:
130140 mEq/L
133146 mEq/L
135145 mEq/L
91
Sodium
Potassium
Age
Magnesium
Age
Calcium
Phosphorus
00.5 y
0.51 y
12 y
25 y
69 y
10 y or older
120
200
225
300
400
500
500
700
1000
1400
1600
2000
00.5 y
0.51 y
13 y
48 y
913 y
> 13 y (M)
> 13 y (F)
30
75
80
130
240
400
360
00.5 y
0.51 y
13 y
48 y
8 y or older
210
270
500
800
1300
100
275
460
500
1250
92
of changes in hemodynamic parameters suggesting intravascular depletion like tachycardia, mild hypotension, and
orthostasis. Severe volume depletion is defined by the presence of more profound hemodynamic compromise, such
as moderate to severe hypotension, tachycardia, and poor
perfusion. Clinical symptoms of mild, moderate, and severe
dehydration generally correspond to a 5%, 10%, and 15%
weight loss in infants, respectively, which can be detected
if pre- and post-dehydration weights are available. In teenagers, mild, moderate, and severe dehydration correspond
to a 3%, 5%, and 7% loss in body weight, respectively.44 Fluid
therapy must include replacement of the deficit as well as
provision of maintenance fluids.
There are various methods used for correcting volume
depletion, but 3 principles always apply: (1) the fluid deficit
must be replaced with an appropriate fluid; (2) maintenance fluids must be provided on an ongoing basis; and (3)
if there are continued ongoing losses (eg, gastric drainage,
vomiting, diarrhea), these losses must be replaced on an
ongoing basis to prevent further fluid deficits. For example,
an infant weighing 5 kg has a 500 mL (10%) fluid deficit.
Given the degree of fluid deficit (10% or more), an isotonic
fluid bolus (20 mL/kg or 2% of the patients body weight
= 100 mL) would be indicated initially. After this bolus,
the remaining deficit (8% or 400 mL) would be replaced as
follows: 50% (200 mL) over the next 8 hours and 50% (200
mL) over the subsequent 16 hours. In addition to the deficit
Adapted from Matarese LE, Gottschlich MM, eds. Contemporary Nutrition Support Practice: A Clinical Guide. Whitmire SJ. Fluids and electrolytes, p. 130.
Copyright 1998 with permission from Elsevier.
2010 A.S.P.E.N. www.nutritioncare.org
93
below 125 mEq/L. Severe acute hyponatremia is life-threatening; fortunately the more common type of hyponatremia
seen in hospitalized patients is slow-developing and less
severe. The 2 most common causes of hyponatremia include
hypervolemic hyponatremia where the total body sodium is
normal to high and TBW is high, and hyponatremia due to
actual sodium and water losses. In kidney failure, liver failure
with ascites, hyperaldosteronism, and congestive heart
failure, the body accumulates sodium and fluid, resulting
in hypervolemic hyponatremia, sometimes referred to as
dilutional hyponatremia. Diarrhea and other GI losses such
as gastric suction, enterocutaneous fistulas, or necrotizing
Adapted from Matarese LE, Gottschlich MM, eds. Contemporary Nutrition Support Practice: A Clinical Guide. Whitmire SJ. Fluids and electrolytes, p. 129.
Copyright 1998 with permission from Elsevier.
2010 A.S.P.E.N. www.nutritioncare.org
94
Potassium
Newborn:
Infant:
Children and Adolescents:
3.75.9 mEq/L
4.15.3 mEq/L
3.44.7 mEq/L
Hypokalemia
Potassium-sparing diuretics
Nonsteroidal anti-inflammatory
drugs
Angiotensin converting enzyme
inhibitors
Angiotensin-II receptor blockers
Trimethoprim
Pentamidine
Cyclosporine
Tacrolimus
Heparin
Penicillin G potassium
-blockers
Succinylcholine
Loop diuretics
Thiazide diuretics
Fludrocortisones
High-dose glucocorticoids
High-dose penicillins
Phenolphthalein
Sodium polystyrene sulfonate
Sorbitol
2adrenergic agonists
Tocolytic agents
Theophylline
Caffeine
Insulin/dextrose
Hypermagnesemia
Hypomagnesemia
Tocolytic agents
Magnesium-containing antacids
Magnesium-containing enemas
Cisplatinum
Foscarnet
Amphotericin B
Aminoglycosides
Thiazide diuretics
Hyperkalemia
Hyperkalemia is defined as a serum potassium concentration greater than 4.7 to 5.9 mEq/L, depending on
the patients age. Signs and symptoms of hyperkalemia
include muscle twitching, cramping, weakness, ascending
paralysis, electrocardiogram (ECG) changes (eg, peaked
T-waves, prolonged PR interval), and dysrhythmias (eg,
bradyarrhythmias, ventricular fibrillation, and asystole).
Hyperkalemia is one of the most dangerous electrolyte
imbalances that develop in premature infants. The immaturity of the kidneys results in a reduced glomerular filtration
rate, urinary potassium excretion, acidosis, and immature
renal tubular response to aldosterone. 54 Hyperkalemia can
occur from excessive potassium intake in the presence of
altered kidney excretion or metabolic acidosis caused by
conditions such as diabetic ketoacidosis and renal tubular
acidosis. Metabolic acidosis causes hyperkalemia by causing
a shift of potassium from the ECF to the ICF in order to
balance the excess hydrogen ions that are moving into the
ICF. Clinically significant hyperkalemia can also develop as
a result of cell lysis (hemolysis) or tissue injury and death
Hypokalemia
Hypokalemia is defined as a serum potassium concentration
less than 3.4 mEq/L. It is a common electrolyte abnormality seen in clinical practice. Causes of hypokalemia
include medications, metabolic alkalosis, abnormal
GI losses, hyperaldosteronism, hypomagnesemia,
95
catecholamines, and inadequate intake. Signs and symptoms are non-specific but include dysrhythmias, paralysis,
muscle necrosis, and possibly death. The treatment of
hypokalemia depends on both the severity and the cause
of the hypokalemia. Oral potassium supplements are available as a variety of salts, and the choice of agent depends
on other concomitant electrolyte imbalances, cost, and
patient preference. In an asymptomatic patient with mild to
moderate hypokalemia, oral supplementation is preferred
because of safety reasons (eg, there is no risk for potassium extravasation). Oral supplementation also reduces the
risk of overcorrection causing hyperkalemia and too rapid
correction causing dysrhythmias. Oral potassium can be
irritating to the GI tract. For mild to moderate potassium
depletion, doses of 2 to 5 mEq/kg/d in divided doses, not
to exceed 1 to 2 mEq/kg as a single dose, taken with plenty
of water, are recommended. 55,56 Serum potassium should
be rechecked approximately 2 hours after the initial dose is
completed, and additional doses given, if needed.
The use of IV potassium supplementation should be
reserved for patients who are symptomatic, who have severe
hypokalemia, or when administration via the GI tract is
contraindicated. Dosages range from 0.5 to 1 mEq/kg
depending on the severity of the hypokalemia and kidney
function. Generally, infusion rates should not exceed
0.5 mEq/kg/h, unless continuous cardiac monitoring is
available. Potassium can be caustic to the vein, so to minimize
irritation, the concentration for administration through a
peripheral vein should not exceed 0.06mEq/mL. 56,57 When
administering IV potassium supplementation, co-administration with dextrose may worsen the hypokalemia by
stimulating insulin release, which promotes the intracellular shift of potassium. Concurrent hypomagnesemia may
result in refractory hypokalemia due to increased renal
potassium losses due to the kidneys attempt to conserve
magnesium and impairment of the Na+-K+-ATPase pump. 58
As such, it is important to correct a low magnesium level
while treating hypokalemia. Serum potassium should be
rechecked approximately 2 hours after the initial dose is
completed, and additional doses given, if needed.
Magnesium
All Age Groups:
1.62.3 mg/dL
96
Hypermagnesemia
Hypermagnesemia is defined as serum magnesium greater
than 2.4 mg/dL. Hypermagnesemia is usually well tolerated but can affect neurological, neuromuscular, and
cardiac function when magnesium concentrations exceed
3 mg/dL.62,63 Physical findings include nausea, vomiting,
diaphoresis, flushing, depressed mental function, drowsiness, muscular weakness, hypotension, and bradycardia. If
patients with severe hypermagnesemia are symptomatic,
IV calcium chloride should be administered immediately
to reduce the excitability of the cardiac muscle. (Note:
Calcium gluconate is generally not recommended in emergency situations.) Hemodialysis may be required to reduce
the magnesium concentration to a safer value. Treatments
for hypermagnesemia in an asymptomatic patient include
dietary magnesium restriction, administration of a loop
diuretic, and possibly hemodialysis if kidney dysfunction is
present.64, 65
2010 A.S.P.E.N. www.nutritioncare.org
Hypomagnesemia
Hypomagnesemia is defined as serum magnesium concentration less than 1.3 mg/dL. It is a common condition seen in
hospitalized patients. Signs and symptoms include apathy,
depression, psychosis, muscle weakness, vertigo, ataxia,
seizures, confusion, leg cramps, hyperactive tendon reflexes,
anorexia, nausea, vomiting, paresthesias, Chvosteks and
Trousseaus sign, spontaneous carpal-pedal spasm, and
cardiac complications including dysrhythmias.67 Hypomagnesemia can also cause other electrolyte abnormalities.
Hypomagnesemia may result in refractory hypokalemia
because of increased renal potassium losses due to the
kidneys attempt to conserve magnesium and impairment
of the Na+-K+-ATPase pump. 58 Hypomagnesemia may also
result in hypocalcemia. Magnesium deficiency can impair
parathyroid function, and hypomagnesemia accompanied
by hypoparathyroidism is a common cause of neonatal
hypocalcemia.68,69
Hypomagnesemia can be caused by decreased intake,
increased excretion, or intracellular shifts of magnesium.70
Excessive renal losses may occur in patients with acute
tubular necrosis, renal tubular acidosis, Bartter syndrome,
or hyperaldosteronism, or may be induced by medications
such as amphotericin, cisplatin, cyclosporine, aminoglycosides, and foscarnet.71 Intracellular shifts can be caused
by dextrose and/or insulin administration. Because only
about 1% to 2% of total body magnesium is found in the
ECF, serum concentrations are not a good reflection of total
body magnesium stores. Treatment of hypomagnesemia is
therefore empirical. The IV route is preferred in patients
with moderate to severe hypomagnesemia because of the
GI intolerance generally seen with large oral doses. Recommended doses are 0.2 to 0.4 mEq/kg (2550 mg/kg), up to
2 mEq (2 g) every 8 to 12 hours for 2 to 3 doses.72 There is
a renal magnesium threshold for magnesium reabsorption;
97
Age Group
Total Calcium
Age Group
Preterm
Full Term
6.211 mg/dL
7.610.4 mg/dL
Preterm
Full Term
< 36 h
Full Term
3684 h
> 84 h
1.752 mmol/L
1.051.37 mmol/L
1.11.42 mmol/L
Hypercalcemia
Calcium
10 d 2 y
212 y
> 12 y
911 mg/dL
8.810.8 mg/dL
8.610 mg/dL
1.21.38 mmol/L
Hypercalcemia is most often seen in patients with hyperparathyroidism or cancer with bone metastases. It can
also occur with toxic serum concentrations of vitamin A
or vitamin D, chronic ingestion of milk and/or calcium
carbonate-containing antacids in the setting of kidney
insufficiency, immobility, tuberculosis, and medications.
Clinical signs and symptoms include fatigue, nausea,
vomiting, constipation, anorexia, and confusion. In severe
cases, cardiac dysrhythmias may be present. Mild hypercalcemia typically responds to fluid and ambulation. In severe
hypercalcemia, immediate treatment should be started
to prevent acute kidney failure, obtundation, ventricular
dysrhythmias, coma, and death. If a loop diuretic and IV
fluid are used to treat hypercalcemia (eg, to increase calcium
excretion), IV hydration with 0.9% sodium chloride should
be started immediately to prevent dehydration. Hemodialysis may be necessary for patients with life-threatening
hypercalcemia or those with kidney failure. 80
Hypocalcemia
Measured hypocalcemia is commonly encountered in
patients with hypoalbuminemia but does not require treatment unless the corrected calcium or ionized calcium is
found to be low. Signs and symptoms of hypocalcemia
include hypotension, decreased myocardial contractility,
prolonged QT interval, paresthesias, Chvosteks and
Trousseaus signs, muscle cramps, tetany, and seizures.
Causes of hypocalcemia include vitamin D deficiency or
the inability to activate vitamin D, hyperphosphatemia,
2010 A.S.P.E.N. www.nutritioncare.org
98
Phosphorus
Newborn:
10 days2 years:
212 years:
> 12 years:
4.59 mg/dL
4.56.7 mg/dL
4.55.5 mg/dL
2.74.5 mg/dL
destruction and acidosis cause a shift to the ECF.77 Intravenous phosphorus requirements are 1 to 2.5 mmol/kg in
premature infants and 0.5 to 1 mmol/kg/d in term infants
and children up to 18 years of age. Oral phosphorus requirements vary with age (Table 9-4).
Hyperphosphatemia
Hyperphosphatemia is defined as a serum phosphate
concentration greater than 4.5 to 9 mg/dL, depending on
the patients age. Most patients are asymptomatic, but signs
and symptoms may include anorexia, nausea, vomiting,
dehydration, and neuromuscular irritability. The biggest
concern with hyperphosphatemia is metastatic calcification from elevated serum concentrations of calcium and
phosphate.91 Although various equations for predicting
metastatic calcification are used, the equations are not
accurate because of the variety of factors that determine
in vivo calcium/phosphate solubility. Ionized calcium is
much more reactive than phosphate, so hypercalcemia
with a mild hyperphosphatemia has a higher probability of
causing metastatic calcification than hyperphosphatemia
with slightly increased serum calcium. With the newer
phosphate-binding agents available, aluminum-containing
antacids are no longer recommended for the treatment or
prevention of hyperphosphatemia in patients with kidney
insufficiency because of the anemia, osteomalacia, and
central nervous system toxicity experienced with the use of
aluminum-containing agents in this patient population.92
Hypophosphatemia
Hypophosphatemia can be defined as a serum phosphorus
concentration below 2.7 mg/dL to 4.5 mg/dL, depending
on the patients age.47 Hypophosphatemia is common in
critical illness, malnutrition, alkalosis, and in patients
receiving phosphate binders (eg, aluminum- , magnesium- ,
and calcium-containing products, sevelamer, or sucralfate).
Signs and symptoms of hypophosphatemia include neurological, neuromuscular, cardiopulmonary, and hematologic
dysfunction.8890 Primary causes of hypophosphatemia
include inadequate intake of phosphate or the administration of large amounts of dextrose solutions in malnourished
patients who are at risk for developing refeeding syndrome
(Chapter 19).
Treatment of hypophosphatemia varies, depending
on the serum phosphorus concentration and the presence
of signs and symptoms. Mild asymptomatic hypophosphatemia can be treated with oral phosphate supplements,
assuming that the GI tract is functional. However, oral
supplements are not well absorbed and often cause diarrhea.
Patients with symptomatic or moderate to severe hypophosphatemia should be treated with IV phosphate. Two
salt forms are available for replacing phosphate: sodium
phosphate and potassium phosphate. Sodium phosphate
provides 4 mEq sodium for every 3 mmol phosphate, and
potassium phosphate provides 4.4 mEq potassium for
every 3 mmol phosphate. When ordering phosphate, the
dose should be ordered in millimoles of phosphate rather
than milliequivalent of the sodium or potassium component of the salt. Common recommendations for replacing
phosphate provide up to 0.32 mmol/kg of phosphate for
serum phosphorus levels < 1.5 mg/dL. This dose is often
inadequate and may require multiple boluses to reach
normal phosphorus levels.93,94 Higher doses have been
recommended for phosphorus replacement in adults, and
the higher supplemental doses have been used successfully
in pediatric patients (Table 9-6). Serum phosphate levels
should be checked 2 hours after the infusion is completed
and the patient should be redosed if needed. Phosphate
should be replaced no faster than 0.1 to 0.2 mmol/kg/h
to allow the phosphate time to move intracellularly and to
prevent possible hypocalcemia.95 If potassium phosphate
is used to replace the phosphate, the infusion rate should
be based on the potassium infusion rate. ECG monitoring
should accompany infusion of individual doses greater than
0.5 mEq/kg/h. Sodium phosphate is the preferred salt for
phosphate supplementation.
Table 9-6 Phosphate Replacement in Adults
Mild Depletion (2.33 mg/dL)
Moderate Depletion (1.62.2 mg/dL)
Severe Depletion (< 1.5 mg/dL)
0.16 mmol/kg
0.32 mmol/kg
0.64 mmol/kg
99
birth. On day 7, the patients serum sodium concentration is 137 mEq/L. The patient continues to receive
160 mL/kg/d of IV fluids that contain 3 mEq/kg/d
of sodium. Over the next 3 days, the patients serum
sodium concentration has decreased to 129 mEq/L.
The patient does not appear to be fluid overloaded,
septic, or suffering from necrotizing enterocolitis. The
patients weight has remained around 1250 g. What is
the most appropriate way to correct the serum sodium
concentration?
A. Increase the sodium concentration in the IV solution
because the current sodium intake is inadequate to
keep up with the renal and GI sodium losses.
B. Decrease the maintenance IV fluid rate by 25%
because the decrease in the serum sodium concentration is probably due to fluid overload.
C. Give sodium chloride orally to replace the sodium
deficit.
D. Decrease the maintenance IV fluid by 25% and
give sodium chloride orally because the decrease
in the serum sodium concentration is probably due
to fluid overload, but the serum sodium concentration is a critical value; thus treatment must begin
immediately.
3. An 8-year-old child who weighs 27 kg is admitted for
nausea, vomiting, and failure to thrive. The patient is
started on maintenance IV fluids containing D5W/0.2
NaCl with 20 mEq KCl per liter. The following morning,
the serum phosphorus concentration is 1.2 mg/dL and
the serum potassium concentration is 3.4 mEq/L.
What is the most appropriate way to correct the serum
phosphorus concentration?
A. Add sodium phosphate 1 mmol/kg to the maintenance IV fluid.
B. Add potassium phosphate 1 mmol/kg to the maintenance IV fluid.
C. Give sodium phosphate 27 mmol (36 mEq sodium)
intravenously over 8 hours.
D. Give potassium phosphate 27 mmol (40 mEq potassium) intravenously over 8 hours.
See p. 487 for answers.
References
100
20. Knepper MA, Rector CF. Urinary concentration and dilution. In: Brenner BM, Rector FC, eds. The Kidney. 4th ed. Vol
1. Philadelphia, PA: Saunders; 1991:445482.
21. Mange K, Matsuura D, Cizman B, et al. Language guiding
therapy: the case of dehydration versus volume depletion. Ann
Intern Med. 1997;127(9):848853.
22. Heitz U, Horne M. Pocket Guide to Fluid, Electrolytes, and AcidBase Balance. 5th ed. St Louis, MO: Mosby-Year Book; 2004.
23. Mirtallo J, Canada T, Johnson D, et al. Safe practices for parenteral nutrition. J Parenter Enteral Nutr. 2004;28(6):S39S70.
24. Holliday MA, Segar WE. The maintenance need for water in
parenteral fluid therapy. Pediatrics. 1957;19:823832.
25. Choong K, Bohn D. Maintenance parenteral fluids in the critically ill child. J Pediatr (Rio J). 2007;83(2 Suppl):S3S10.
26. Lorenz JM, Kleinman LI, Ahmed G, et al. Phases of fluid and
electrolyte homeostasis in the extremely low birth weight
infant. Pediatrics. 1995;96(3):484489.
27. Modi N. Sodium intake and preterm babies. Arch Dis Child.
1993;69:8791.
28. Bauer K, Bovermann G, Roithmaier A, Gotz M, Proiss A,
Versmold HT. Body composition, nutrition and fluid balance
during the first two weeks of life in preterm neonates weighing
less than 1500 grams. J Pediatr. 1991;118(4):615620.
29. Sankar MJ, Agarwal R, Mishra S, Deorari AK, Paul VK.
Feeding of low birth weight infants. Indian J Pediatr.
2008;75(5):459469.
30. Shaffer SG, Brandt SK, Hall RT. Postnatal changes in
total body water and extracellular volume in the preterm
infant with respiratory distress syndrome. J Pediatr.
1986;109(3):509514.
31. Tang W, Ridout D, Modi N. Influence of respiratory distress
syndrome on body composition after preterm birth. Arch Dis
Child Fetal Neonatal Ed. 1997;77(1):F28F31.
32. Rutter N, Hull D. Water loss from the skin of term and preterm
babies. Arch Dis Child. 1979;54(11):858868.
33. Costarino AT, Baumgart S. Modern fluid and electrolyte
management of the critically ill premature infant. Ped Clin
North Am. 1986;33(1):153178.
34. Kjartansson S, Hammarlund K, Sedin G. Insensible water
loss from the skin during phototherapy in term and preterm
infants. Acta Paediatr. 1992;81(10):764768.
35. Grunhagen DJ, de Boer MG, de Beaufort AJ, Walther FJ.
Transepidermal water loss during halogen spotlight therapy
in preterm infants. Pediatr Res. 2002;51(3):402405.
36. Maayan-Metzger A, Yosipovitch G, Hadad E, Sirota L. Trans
epidermal water loss and skin hydration in preterm infants
during phototherapy. Am J Perinatol. 2001;18(7):393396.
37. Hartnoll G. Basic principles and practical steps in the
management of fluid balance in the newborn. Semin Neonatal.
2003;8(4): 307313.
38. Davis ID, Avner ED. Neonatal-perinatal medicine: diseases of
the fetus and infant. In: Fanaraff AA, Martin RJ, eds. Fluid and
Electrolyte Management. 7th ed. St. Louis, MO: CV Mosby;
2002:619627.
39. Kumar V, Cotran RS, Robbins SL. Disorders of vascular flow
and shock. In: Kumar V, Cotran RS, Robbins SL, eds. Basic
Pathology. 5th ed. Philadelphia, PA: Saunders; 1992:6191.
40. Pincus MR, Preuss HG, Henry JB. Evaluation of renal function, water, electrolytes, acid-base balance and blood gasses.
In: Henry JB, ed. Clinical Diagnosis and Management by
Laboratory Methods. 19th ed. Philadelphia, PA: Saunders;
1996:139149.
41. Hwai-Ping S. Sodium, chloride, and potassium. In: Stipanuk
M, ed. Biochemical and Physiological Aspects of Human Nutrition. Philadelphia, PA: Saunders; 2000:686710.
42. Wilkins BH. Renal function in sick very low birthweight
infants: 3. Sodium, potassium, and water excretion. Arch Dis
Child. 1992; 67(10):11541161.
43. Hartnoll G, Betremieux P, Modi N. Randomised controlled
trial of postnatal sodium supplementation on body composition in 25 to 30 week gestational age infants. Arch Dis Child
Fetal Neonatal Ed. 2000;82(1):F24F28.
44. Roberts KB. Fluids and electrolytes: parenteral fluid therapy.
Pediatr Rev. 2001;22(11):380387.
45. Finberg L. Hypernatremic (hypertonic) dehydration in
infants. N Engl J Med. 1973;289(4):823832.
46. Holliday MA, Kalayci MN, Harrah J. Factors that limit brain
volume changes in response to acute and sustained hyper- and
hyponatremia. J Clin Invest. 1968;47(8):19161928.
47. Robertson J. Blood chemistries and body fluids. In: Robertson
J, Shilkofski N, eds. The Harriet Lane Handbook. 7th ed. Philadelphia, PA: Elsevier Mosby; 2006:661672.
48. Horisberger J, Lemas V, Kraehenbuhl J, Rossier BC. Structure-function relationship of Na,K-ATPase. Annu Rev Physiol.
1991;53:565584.
49. Rodriguez-Soriano J. Potassium homeostasis and its disturbance in children. Pediatr Nephrol. 1995;9(3):364374.
50. Ganguly A. Primary aldosteronism. N Engl J Med.
1998;339(25):18281834.
51. Kelvay LM, Bogden JD, Aladjem M, et al. Renal and gastrointestinal potassium excretion in humans: new insight based on
new data and review and analysis of published studies. J Am
Coll Nutr. 2007;26(2):103110.
52. Mathialahan T, Sandle GI. Dietary potassium and laxatives as
regulators of colonic potassium secretion in end-stage renal
disease. Nephrol Dial Transplant. 2003;18(2):341347.
53. Brown RS. Extrarenal potassium homeostasis. Kidney Int.
1986;30(1):116127.
54. Subramanian S, Agarwal R, Deorari AK, Paul VK,
Bagga A. Acute renal failure in neonates. Indian J Pediatr.
2008;75(4):385391.
55. Strom BL, Carson JL, Schinnar R, et al. Upper gastrointestinal
tract bleeding from oral potassium chloride. Comparative risk
from microencapsulated vs wax matrix formulations. Arch
Intern Med. 1987;147(5):954957.
56. Potassium chloride. In: Taketomo CK, Hodding JH, Kraus
DM, eds. Pediatric Dosage Handbook. 15th ed. Hudson, OH:
Lexi-Comp; 2008:14321435.
57. Potassium chloride. In: Phelps SJ, Hak EB, Crill CM, eds. The
Teddy Bear Book: Pediatric Injectable Drugs. 8th ed. Bethesda,
MD: American Society of Health-System Pharmacists;
2007:368369.
101
58. Whang R, Whang DD, Ryan MP. Refractory potassium repletion. A consequence of magnesium deficiency. Arch Intern
Med. 1992;152(1):4045.
59. Reinhart RA. Magnesium metabolism. A review with special
reference to the relationship between intracellular content
and serum levels. Arch Intern Med. 1988;148(11):24152420.
60. Zaloga GP, Roberts PR. Calcium, phosphorus, and magnesium disorders. In: Ayres SM, Grenvik NA, Holbrook PR,
Shoemaker WC, eds. Textbook of Critical Care. 4th ed. Philadelphia, PA: Saunders; 2000:905928.
61. Quamme GA. Laboratory evaluation of magnesium status.
Renal function and free intracellular magnesium concentration. Clin Lab Med. 1993;13(1):209223.
62. Teng RJ, Wu TJ, Sharma R, Garrison RD, Hudak ML. Early
neonatal hypotension in premature infants born to preeclamptic mothers. J Perinatol. 2006;26(8):471475.
63. Whang R, Ryder KW. Frequency of hypomagnesemia and
hypermagnesemia. Requested vs routine. JAMA. 1990;
263(22):30633064.
64. Reinhart RA. Magnesium metabolism. A review with special
reference to the relationship between intracellular content
and serum levels. Arch Intern Med. 1988;148:(11)24152420.
65. Van Hook JW. Endocrine crises. Hypermagnesemia. Crit Care
Clin. 1991;7(1):215223.
66. Ramsey PS, Rouse DJ. Magnesium sulfate as a Tocolytic
agent. Semin Perinatol. 2002;25(4):236247.
67. Weisinger JR, Bellorin-Font E. Magnesium and phosphorus.
Lancet. 1998;352(9125):391396.
68. Lee CT, Tsai WY, Tung YC, Tsau YK. Transient
pseudohypoparathyroidism as a cause of late-onset
hypocalcemia in neonates and infants. J Formos Med Assoc.
2008;107(10):806810.
69. Desai TK, Carlson RW, Geheb MA. Prevalence and
clinical implications of hypocalcemia in acutely ill
patients in a medical intensive care setting. Am J Med.
1988;84(2):209214.
70. Ryzen E, Wagers PW, Singer FR, Rude RK. Magnesium
deficiency in a medical ICU population. Crit Care Med.
1985;13(1):1921.
71. Dacey MJ. Hypomagnesemic disorders. Crit Care Clin.
2001;17(1):155173, viii.
72. Magnesium sulfate. In: Taketomo CK, Hodding JH, Kraus
DM, eds. Pediatric Dosage Handbook. 15th ed. Hudson, OH:
Lexi-Comp; 2008:10901092.
73. Topf MJ, Murray PT. Hypomagnesemia and hypermagnesemia. Rev Endocr Metab Disord. 2003;4(2):195206.
74. Oster JR, Epstein M. Management of magnesium depletion.
Am J Nephrol. 1988;8(5):349354.
75. Herbert P, Mehta N, Wang J, Hindmarsh T, Jones G, Cardinal
P. Functional magnesium deficiency in critically ill patients
identified using a magnesium-loading test. Crit Care Med.
1997;25(5):749755.
76. Chattopadhyay N, Mithal A, Brown EM. The calciumsensing receptor: a window into the physiology and
pathophysiology of mineral ion metabolism. Endocrinol Rev.
1996;17(5):289307.
102
PART II
10
Russell J. Merritt, MD, PhD, FAAP, Barbara Marriage, PhD, RD, and Ricardo Rueda, MD, PhD
Learning Objectives
CONTENTS
Nutrition Physiology of Pregnancy . . . . . . . . . . . . . . . . . . 105
Impact of Maternal Dietary Deficiencies onthe Fetus. . 107
Macronutrients
Calcium
Iron
Folic Acid and Vitamin B12
Vitamin E
Multiple Micronutrients
114
115
115
116
106
secretion. In intrauterine growth restriction (IUGR) pregnancies, placental weight is reduced more than fetal weight.
In contrast, the placenta may be increased in mass in gestational diabetes.1
Much of what we know about fetal substrate metabolism comes from extensive studies of the late gestation
ovine fetus initiated by Battaglia and many other investigators associated with the Perinatal Research Center at the
University of Colorado School of Medicine.28 The umbilical
vein transports oxygen and nutrients to the fetus toward the
fetal liver and ductus venosus, which variably shunts blood
around the liver to the fetal heart and brain. Until the time
of birth, when the lungs expand and pulmonary artery
resistance increases, the lungs are also partly bypassed. This
increases oxygenated blood flow to the fetal brain via the
ductus arteriosus that connects the pulmonary artery and
the aorta. Blood containing amino acids, metabolites, and
carbon dioxide (CO2) returns to the placenta by way of the
2 umbilical arteries.
The major energy source for the fetus is glucose, which
normally all comes from maternal transport to the fetus and
placenta.2 It also uses lactate produced in the placenta and
endogenously.9 Glucose uptake by placental and fetal tissues
and fetal growth are proportional to glucose delivery. 2 The
fetal pancreas secretes insulin by mid-gestation and responds
to variations in the glucose delivery rate. Normally the fetal
liver is not active in gluconeogenesis. Fetal tissue glucose
transporters and intracellular downstream metabolic regulators are modulated by glucose and insulin levels in the
fetus. Insulin-responsive fetal tissues include the heart,
liver, skeletal muscle, and adipose tissue. Placental glucose
uptake is not regulated by insulin. When fetal glucose supply
is limited, fetal glucose oxidation is maintained by virtue of
increased uterine artery/umbilical vein glucose gradient
and gluconeogenesis from amino acids in the fetus: the
fetus develops with mechanisms that tend to keep its energy
metabolism relatively constant, while growth is, at times of
deficient energy supply, expendable.3
Amino acids are the second most important macronutrient in the fetus with at least 14 complex amino acid
transporter systems on both of the syncytiotrophoblast
membranes.10 The large neutral and branched-chain amino
acids are transported most directly proportional to their
maternal concentration. 5 Because of shared transporters
and competition among amino acids for specific transporters, an increase in delivery of multiple amino acids
sharing the same transporter to the uterine artery may have
a different effect than an increase of a single amino acid on
the uptake and transport of a specific amino acid. All amino
2010 A.S.P.E.N. www.nutritioncare.org
107
Macronutrients
In terms of maternal macronutrient status, there is some
evidence that balanced protein/energy supplementation
may be beneficial for decreasing rates of low birth weight
(LBW) and small for gestational age (SGA) deliveries,
especially in populations where women have chronically
marginal nutrition status prior to pregnancy.15 However,
overall analysis of the available evidence suggests that
maternal supplementation with balanced or high-protein
diets had no beneficial effects on fetal growth. There is
limited evidence that protein supplementation adversely
affected fetal growth rate (as measured by mean birth
weight) and therefore potentially increased LBW deliveries.
The effect of energy/protein restriction has been also evaluated in women who were classified as obese pre-pregnancy
or had rapid early gestational weight gain. In women who
were obese before pregnancy, there have been no benefits to
fetal growth of restricting energy and protein during gestation, although evidence from controlled trials is limited. 23
It has also been reported that high intakes of protein and
fat during pregnancy may impair development of the
fetal pancreatic beta cells and lead to insulin deficiency in
theoffspring.16
2010 A.S.P.E.N. www.nutritioncare.org
108
On the other hand, very recent studies have demonstrated that a low-protein diet in utero had a deleterious
effect on bone development in the offspring that persisted
into adulthood.24 The offspring displayed significant differences in bone structure and density at various sites. These
differences are indicative of significantly altered bone
turnover.25
Table 10-1 Recommended Daily Nutrient Intakes During Pregnancy
Water
Energy
Carbohydrate
Total fiber
Linoleic acid
Linolenic acid
Protein
Vitamin A
Vitamin C
Vitamin D
Vitamin E
Vitamin K
Thiamin
Riboflavin
Niacin
Vitamin B6
Folate
Vitamin B12
Pantothenic acid
Biotin
Choline
Calcium
Chromium
Copper
Fluoride
Iodine
Iron
Magnesium
Manganese
Molybdenum
Phosphorus
Selenium
Zinc
Potassium
Sodium
Chloride
3L
Varies by age, pregnancy stage, BMI, activity
175 g
28 g
13 g
1.4 g
71 g
750770 mcg*
8085 mg/d*
5 mcg
15 mg
7590 mcg*
1.4 mg
1.4 mg
18 mg
1.9 mg
600 mcg
2.6 mcg
6 mg
30 mcg
450 mg
10001300 mg*
2930 mcg*
1000 mcg
3 mg
220 mcg
27 mg
350400 mg*
2 mg
50 mcg
7001250 mg*
60 mcg
1112 mg*
4.7 g
1.5 g
2.3 g
Calcium
In terms of specific micronutrients, maternal calcium
supplementation may have a beneficial effect on fetal
growth, particularly in women with low calcium status at
the outset of pregnancy or who were classified as being at
risk of gestational hypertension. Calcium supplementation during pregnancy can be linked directly to increased
bone density and bone length of neonates.26 The effects on
fetal growth appeared to come partly from a reduction in
preeclampsia and resultant lengthened gestation.23
Iron
Some studies have reported that iron deficiency anemia
early in pregnancy was associated with greater than a
twofold increase in the risks of LBW and preterm delivery.21
In addition, reduced iron availability for brain iron accretion is associated with persisting developmental and
behavioral changes. A number of conditions associated with
fetal growth retardation or macrosomia such as diabetes,
placental insufficiency, and smoking restrict iron availability
during gestation and predispose to later iron deficiency. 27
Vitamin E
The plasma concentration of -tocopherol, the most
common isomer of vitamin E, was positively related to fetal
growth (birth weight for gestation), reduced small-forgestation births, and increased risk of large-for-gestation
births. Concentrations of -tocopherol were positively
Multiple Micronutrients
There have been few studies published to examine whether
multiple micronutrient supplements might be more
beneficial than single micronutrients. There is evidence of
interactions of several micronutrients at the metabolic level.
Little is yet known about the significance of these interactions for pregnancy outcomes, especially in developing
countries where nutrient deficiencies rarely occur in isolation and multiple micronutrient deficiencies are common. 29
A meta-analysis of global multinutrient supplementation
studies found a small effect on birth weight between ironfolate supplementation or placebo. 30
109
110
Figure 10-1 Scheme of the role that programming of the fetal hypothalamic-pituitary-adrenal (HPA) axis during development plays in the link between
fetal growth and long-term disease in adulthood. Gc: Glucocorticoids; Dex: Dexamethasone.
111
112
Animal Models
113
114
Several animal studies have shown that prenatal glucocorticoid excess, either from endogenous overproduction from
maternal stress or through exogenous administration to the
mother or fetus, reduces birth weight and causes hypertension, hyperglycemia, and behavioral abnormality in the
offspring. These effects are transmitted across generations
without further exposure to glucocorticoids, an observation that supports an epigenetic mechanism.86
Rat offspring that have been exposed to excess prenatal
glucocorticoids undergo catch-up growth postnatally
and normalize body weight by weaning. Outcomes from
such offspring when they are adults are consistent with
the hypothesis that rapid postnatal catch-up growth is
deleterious to health.78 There are also accumulating data in
rodents to suggest that prenatal glucocorticoid overexposure programs an adverse adult cardiovascular, metabolic,
neuroendocrine, and behavioral phenotype. The phenotypic outcome is similar to that of the low-protein model.
Fetal glucocorticoid overexposure may be a common
mechanism for mediating fetal growth retardation and
metabolic programming. This suggestion is based on the
observation that dietary protein restriction during rat
pregnancy reduces 11-HSD2 activity. This enzyme, as
mentioned earlier, serves as a placental barrier to maternal
glucocorticoids by rapidly metabolizing maternal glucocorticoids to inert 11-keto forms to minimize fetal exposure to
glucocorticoids.78
HPA regulation can be programmed by nutrient restriction. In fetal rats nutrient restriction results in blunted
diurnal patterns of adrenocorticotropic hormone (ACTH)
at 4 weeks postnatal age, alterations in basal plasma corticosterone in adulthood, and altered basal HPA axis activity. 33
Normally, the presence of 11-HSD2 in the placental syncytiotrophoblasts protects the fetus from maternally derived
glucocorticoids. Maternal glucocorticoid levels are much
higher than fetal levels for most of pregnancy, so a relative
deficiency in placental 11-HSD2 would put the fetus at
great risk of increased glucocorticoid exposure. 87 In some
studies, a strong positive correlation has been reported
between placental 11-HSD2 activity and fetal weight at
term and birth weight in preterm infants. Maternal protein
restriction in rodents reduces the activity of 11-HSD2 in
the placenta. 11-HSD2 activity is influenced by multiple
maternal environmental factors and its modulation may
be a mechanism through which a variety of environmental
insults exert their programming effects. 86
Reduction in placental blood flow and consequent restriction of oxygen, nutrient transport, and fetal growth can
be produced in the rat by uterine artery ligation in late
gestation, uterine and umbilical artery embolism, or
carunclectomy.78,79 At 2 weeks of age, growth-retarded
offspring in this model have reduced nephron number. This
nephron deficit was associated with impaired renal function at 2 weeks of age despite compensatory hypertrophy
of remaining nephrons. Also, molecular analysis of skeletal
muscle from fetuses and 21-day-old offspring following
uterine artery ligation revealed that this mode of growth
restriction is associated with changes in both mitochondrial gene expression and function. In female offspring,
after uterine artery ligation, growth restriction was associated with increased fasting blood glucose levels and with
impaired glucose tolerance and lower insulin secretion
during a glucose tolerance test.78
115
116
gain than formula feeding and can be viewed as particularly beneficial in this population. An important medical
concern in such infants is to support brain development,
but most of the data on the importance of early nutrition for
brain development come from studies of premature infants.
It remains largely unknown if nutritional supplementation
and growth acceleration are beneficial in this regard for term
growth-retarded infants. For infants in underdeveloped
countries who develop extrauterine growth retardation
later in infancy and early childhood, short-term nutritional
supplementation has been found to reduce acute morbidity
and mortality (as well as progression of their malnutrition).
Premature infants, especially extremely low birth
weight (ELBW) infants, are at high risk of extrauterine
growth retardation. Their nutrition requirements exceed
those of term infants, given their immature development
and body composition. In these infants, developmental
achievement and reduced neurological complications are
associated with higher growth rates. Premature infants,
both in-hospital and following hospital discharge, are very
responsive to nutritional supplementation. Data from Lucas
and Singhal have been taken by some to demonstrate that
rapid growth should not be encouraged for this population,
based on higher blood pressure, leptin, blood lipids, and
split proinsulin values in premature infants when fed premature formula compared to term formula or human milk.
However, in their studies, these markers of cardiovascular
risk were not elevated above those for typical term infants.
These same investigators demonstrated developmental
disadvantages in these infants at least through childhood
from receiving standard term formulas versus more nutrientrich premature formula. Interestingly, while premature
infants fed premature formula grew faster than breastfed
infants, their neuro-developmental status was not better
than that of breastfed infants. Bone mineralization is
improved in premature infants given mineral-rich premature and post-discharge formulas compared to standard
term formulas. However, this may not persist into
adulthood.92
Human milk with human milk fortifier is the current
recommended approach for small premature infants as it can
preserve the beneficial nutrition and immunologic effects
of human milk while providing additional energy, protein,
minerals, and other micronutrients. For formula-fed premature infants, premature, not term, formula is recommended.
However, there is no evidence postdischarge formulas have
improved developmental outcomes versus infants fed standard term formulas after hospital discharge. However, in the
smallest infants, they have been observed to enhance head
2010 A.S.P.E.N. www.nutritioncare.org
growth. The recent U.S. data from Ehrenkranz on the positive developmental outcome of encouraging early growth in
premature infants dictates against any go-slow approach for
these infants to possibly reduce markers of future cardiovascular risk (to below values observed in healthy term
infants).93
References
1. Pardi G, Cetin I. Human fetal growth and organ development: 50 years of discoveries. Am J Obstet Gynecol.
2006;194(4):10881099.
2. Hay WW Jr. Placental-fetal glucose exchange and fetal
glucose metabolism. Trans Am Clin Climatol Assoc.
2006;117:321340.
3. Hay WW Jr. Recent observations on the regulation of fetal
metabolism by glucose. J Physiol. 2006;572(pt 1):1724.
4. Battaglia FC. Clinical studies linking fetal velocimetry, blood
flow and placental transport in pregnancies complicated
by intrauterine growth retardation (IUGR). Trans Am Clin
Climatol Assoc. 2003;114:305313.
5. Battaglia FC. In vivo characteristics of placental amino acid
transport and metabolism in ovine pregnancy--a review.
Placenta. 2002;23(Suppl A):S3S8.
6. Battaglia FC, Wilkening R, Meschia G. Unique organ specific
characteristics of amino acid metabolism in early development. Trans Am Clin Climatol Assoc. 1995;106:141149.
7. Barry JS, Anthony RV. The pregnant sheep as a model for
human pregnancy. Theriogenology 2008;69(1):5567.
8. Regnault TR, Friedman JE, Wilkening RB, Anthony RV, Hay
WW Jr. Fetoplacental transport and utilization of amino acids
in IUGR--a review. Placenta. 2005;26(Suppl A):S52S62.
9. Sparks JW, Hay WW Jr, Bonds D, Meschia G, Battaglia
FC. Simultaneous measurements of lactate turnover rate
and umbilical lactate uptake in the fetal lamb. J Clin Invest.
1982;70(1):179192.
117
118
119
82. Vickers MH, Breier BH, Cutfield WS, Hofman PL, Gluckman
PD. Fetal origins of hyperphagia, obesity, and hypertension
and postnatal amplification by hypercaloric nutrition. Am J
Physiol Endocrinol Metab. 2000;279(1):E83E87.
83. Woodall SM, Johnston BM, Breier BH, Gluckman PD.
Chronic maternal undernutrition in the rat leads to delayed
postnatal growth and elevated blood pressure of offspring.
Pediatr Res. 1996;40(3):438443.
84. Bouret SG, Simerly RB. Developmental programming of
hypothalamic feeding circuits. Clin Genet. 2006;70(4):295301.
85. Langley-Evans SC, Bellinger L, McMullen S. Animal models
of programming: early life influences on appetite and feeding
behaviour. Matern Child Nutr. 2005;1(3):142148.
86. Drake AJ, Tang JI, Nyirenda MJ. Mechanisms underlying the
role of glucocorticoids in the early life programming of adult
disease. Clin Sci. (Lond) 2007;113(5):219232.
87. Fowden AL, Forhead AJ. Endocrine mechanisms of intrauterine programming. Reproduction. 2004;127(5):515526.
88. Barker DJ, Shiell AW, Barker ME, Law CM. Growth in utero
and blood pressure levels in the next generation. J Hypertens.
2000;18(7):843846.
89. Stein AD, Lumey LH. The relationship between maternal
and offspring birth weights after maternal prenatal famine
exposure: the Dutch Famine Birth Cohort Study. Hum Biol.
2000;72(4):641654.
90. Pembrey ME, Bygren LO, Kaati G, et al. Sex-specific, maleline transgenerational responses in humans. Eur J Hum Genet.
2006;14(2):159166.
91. Heijmans BT, Tobi EW, Stein AD, et al. Persistent epigenetic differences associated with prenatal
exposure to famine in humans. Proc Natl Acad Sci. USA
2008;105(44):1704617049.
92. Fewtrell MS, Williams JE, Singhal A, Murgatroyd PR, Fuller
N, Lucas A. Early diet and peak bone mass: 20 year follow-up
of a randomized trial of early diet in infants born preterm.
Bone. 2009;45(1):142149.
93. Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage LA,
Poole WK. Growth in the neonatal intensive care unit influences neurodevelopmental and growth outcomes of extremely
low birth weight infants. Pediatrics. 2006;117(4):12531261.
94. Beardsall K, Diderholm BM, Dunger DB. Insulin and carbohydrate metabolism. Best Pract Res Clin Endocrinol Metab.
2008; 22(1):4155.
95. Alexe DM, Syridou G, Petridou ET. Determinants of early life
leptin levels and later life degenerative outcomes. Clin Med
Res. 2006;4(4):326335.
96. Palou A, Pico C. Leptin intake during lactation prevents
obesity and affects food intake and food preferences in later
life. Appetite. 2009;52(1):249252.
97. Stocker CJ, Cawthorne MA. The influence of leptin on
early life programming of obesity. Trends Biotechnol.
2008;26(10):545551.
98. Zeisel SH. Nutrigenomics and metabolomics will change
clinical nutrition and public health practice: insights from
studies on dietary requirements for choline. Am J Clin Nutr.
2007;86(3):542548.
11
Human Milk
Jacqueline J. Wessel, RD, CNSD
Learning Objectives
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Breastfeeding. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Preterm Breast Milk. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Fortification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Banked Human Milk. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Vitamin D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Growth of Breastfed Infants. . . . . . . . . . . . . . . . . . . . . . .
Breast Milk Safety and Administration . . . . . . . . . . . . . .
Breast Milk and Maternal Medications . . . . . . . . . . . . . .
120
120
121
122
122
123
123
123
124
125
Introduction
HUMAN MILK
2. Train all health care staff in skills necessary to implement this policy.
Society
Infant
121
Breastfeeding
122
Table 11-3 Nutrient Composition of Human Milk and Fortified Human Milk
Based on Intake of 150 mL/kg5459
Preterm Milk
Term Milk
Preterm milk with
Enfamil Human
Milk Fortifier with
iron
Preterm milk with
Similac Human
Milk Fortifier
Preterm/term
milk 1:1 with
Similac Special
Care 30 cal/oz
Prolact+4
H2MF
Recommended
Intake for
preterm infants
Calories
Protein
Calcium
Phosphorus
101 cal/kg
102 cal/kg
2.1 g/kg
1.5 g/kg
mixed to
120 cal/kg
3.75 g/kg
172.5 mg/
kg
94.5 mg/kg
120 cal/kg
3.6 g/kg
207 mg/kg
117 mg/kg
4.35 g/kg
preterm milk
125 cal/kg
174 mg/kg 95.6 mg/kg
3.75 g/kg
term milk
Fortifies up
Added
Added
120 cal/kg
minerals
to 3.45 g/kg minerals
120 cal/kg
Fortification
3-4 g/kg/d
120-230
mg/kg
60-140 mg/
kg
HUMAN MILK
123
Vitamin D
124
Storage Method
Temperature
Room temperature
Cooler with ice packs
Refrigerator,
thawedmilk
Refrigerator,
fortified milk
Refrigerator,
fresh milk
Freezer, home unit
Freezer
Freezer
25C, 75F
15C, 59F
4C, 39F
Length of Time
Recommended
<4h
24 h
24 h feeding when
possible
4C, 39F
24 hours
4C, 39F
48 h
20C, 4F
70C, 94F
3 mo
612 mo
> 12 mo
HUMAN MILK
125
References
1. American Academy of Pediatrics, Committee on Nutrition. Kleinman RE, ed. Pediatric Nutrition Handbook. 6th
ed. Elk Grove Village, IL: American Academy of Pediatrics;
2009:55.
2. Canadian Paediatric Society Nutrition Committee, American
Association of Pediatrics Committee on Nutrition. Breastfeeding; a commentary in celebration of the international year
of the child.1979. Pediatrics. 1978;65:591601.
3. International Pediatric Association: Recommendations for
action programmes to encourage breastfeeding. Acta Paediatr
Scand. 1976;65:275277.
4. WHO/UNICEF. Protecting, promoting, and supporting
breastfeeding; the special role of maternity services, a joint
WHO/UNICEF statement. Geneva Switzerland 1989, World
Health Organization.
5. US Department of Health and Human Services. Healthy
People 2010: Understanding and Improving Health and Objectives for Improving Health. Vols. 1, 2. 2nd ed. Washington, DC:
US Dept of Health and Human Services; 2000.
6. Centers for Disease Control and Prevention. Breastfeeding
Report Card, United States: Outcome Indicators. http://www.
cdc.gov/breastfeeding/data/report_card2.htm.Accessed
November 12, 2009.
7. Lawrence RA, Lawrence RM. Breastfeeding: A Guide for the
Medical Profession. 6th ed. St. Louis, MO: Mosby; 2005:187.
126
8. Fanaro S, Vigi V. Infant formulas supplemented with prebiotics: intestinal microbiota and immune responses. Minerva
Pediatr. 2008;60:327335.
9. Hatakka K, Savilahti E, Ponka A, et al. Effect of long-term
consumption of probiotic milk on infections in children
attending day care centres: double blind, randomized trial.
BMJ. 2001;322:1327.
10. May JT. Microbial contaminants and antimicrobial properties
of human milk. Microbiol Sci. 1988;5:4246.
11. Goldman AS, Garza C, Nichols BJ, et al. Immunologic factors
in human milk during the first year of lactation. J Pediatr.
1982;100:563.
12. Howie PW, Forsyth JS, Ogston SA, et al. Protective effect of
breastfeeding against infection. BMJ. 1990;336:92.
13. Duncan B, Ey J, Holberg CJ, Wright AJ, et al. Exclusive breastfeeding for at least 4 months protects against otitis media.
Pediatrics. 1993;91:867872.
14. Aniansson G, Alni B, Andersson A, et al. A prospective cohort
study on breastfeeding and otitis media in Swedish infants.
Pediatr Infectious Dis J. 1994;12:183188.
15. Paradise JL, Elster BA, Tan L. Evidence in infants with cleft
palate that breast milk protects against otitis media. Pediatrics.
1994;94:853860.
16. Pisacane A, Grazione L, Mazzarella G, et al. Breastfeeding and
urinary tract infections. J Pediatr. 1992;120:8789.
17. Vennemann MM, Bajanowski T, Brinkman B, et al. Does
breastfeeding reduce the risk of sudden infant death
syndrome? Pediatrics. 2009;123:e406e410.
18. Saarinen IJM, Kajosaari M, Backnman A, et al. Prolonged
breastfeeding as prophylaxis for atopic disease. Lancet.
1979;2:163166.
19. Kramer MS. Does breastfeeding help protect against atopic
disease? Biology, methodology and a golden jubilee of controversy. J Pediatr. 1988;112:181190.
20. Thygarejan A, Burks AW. American Academy of Pediatrics
recommendations on the effect of early nutritional interventions on the development of atopic disease. Curr Opin Pediatr.
2008;20:698702.
21. Greer FR, Sicherer SH, Burks W, et al. Effects of nutritional
interventions on the development of atopic disease in infants
and children; the role of maternal diet restriction, timing
of introduction of complementary foods, and hydrolyzed
formulas. Pediatrics. 2008;121:183190.
22. Hanson LA, Adlerberth I, Carlsson B, et al. Host defense of
the neonate and the intestinal flora. Acta Peadiatric Scand.
1989; 351(suppl):122125.
23. Koletzko S, Sherman P, Corey M, et al. Role of infant feeding
practices in development of Crohns disease in childhood.
BMJ. 1989;298:16171618.
24. Gerstein HC. Cows milk exposure and type I diabetes
mellitus. Diabetes Care. 1994;17:1319.
25. Savilahti E, Saarinen KM. Early infant feeding and type I
diabetes. Eur J Nutr. 2009;48(4):243249.
26. Davis MK, Savitz DA, Graubard BI. Infant feeding and childhood cancer. Lancet. 1988;2:365368.
27. Schanler RJ. The use of human milk for premature infants.
Pediatr Clin N Am. 2001;48:207219.
HUMAN MILK
127
128
12
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
Types of Infant Formulas. . . . . . . . . . . . . . . . . . . . . . . . . . 130
Standard Milk-Based Term Infant Formulas
Term Formulas Designed for Symptoms of Intolerance
Premature or Low Birth Weight Formulas
Premature Discharge Formulas
Specialized Infant Formulas
Learning Objectives
Introduction
130
Regulation
Product Line
Web Site
Abbott
Nutritionals
Mead Johnson
Nestle Gerber
Nutricia
Similac
www.abbottnutrition.com
Enfamil
GOOD START
Neocate and
other products
Bright Beginnings
Parents Choice
some modulars
www.meadjohnson.com
www.gerber.com
www.nutricia-na.com
PBM Nutritionals
Vitaflo
www.pbmproducts.com
www.vitaflousa.com
131
Table 12-2 Standard Milk Based Infant Formulas (per 100 calories) The standard dilution for these products is 0.67 kcal/mL (20 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
Bright Beginnings
Milk-based
(PBM Nutritionals)
2.2 (9%)
10.9 (43.6%)
nonfat milk, whey lactose
protein concentrate
Bright Beginnings
Organic
(PBM Nutritionals)
2.2 (9%)
organic reduced
minerals whey,
nonfat milk
10.6 (42%)
lactose
2.1 (8.5%)
(liquid): reduced
minerals whey,
nonfat milk
(powder): whey
protein concentrate,
nonfat milk
2.1 (8.5%)
whey, nonfat milk
10.9 (43.5%)
lactose
2.2 (9%)
whey protein
concentrate
(enzymatically
hydrolyzed, reduced
in minerals)
2.2 (9%)
whey protein
concentrate
(enzymatically
hydrolyzed, reduced
in minerals)
2.2 (9%)
whey protein
concentrate
(enzymatically
hydrolyzed, reduced
in minerals)
2.1 (8.4%)
nonfat milk, whey
protein concentrate
Enfamil PREMIUM
(Mead Johnson)
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
K
mg
Ca Phos Fe
mg mg mg
Osmolal- Comments
ity mOsm
5.3 (47.7%)
27 108 82
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunflower oil
5.3 (48%)
23 84 63
organic palm or
palm olien, high
oleic safflower
or sunflower oil,
coconut oil, soy oil
5.3 (48%)
27 108 78
palm olein, soy oil,
coconut oil, high
oleic sunflower oils
43 1.8
added nucleotides
42 1.8
43 1.8
300
11 (43.5%)
5.3 (48%)
27 108 78
lactose, galacto- palm olein, soy oil,
oligosaccharides coconut oil, high
oleic sunflower oils
43 1.8
300
11.2 (45%)
lactose, corn
maltodextrin
5.1 (46%)
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunflower oil
27 108 67
38 1.5
250
added nucleotides
28 mg/L, DHA 17 mg,
ARA 34 mg
PRSL 19.1/100 cals
added prebioticgalactooligosaccharide (GOS)
46 mg added
nucleotides, DHA 0.32%,
ARA 0.64% PRSL
19.5/100 cals
11.2 (45%)
lactose, corn
maltodextrin
5.1 (46%)
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunflower oil
27 108 67
38 1.5
250
5 mg added nucleotides
PRSL 19.5/100 cals
No DHA and ARA
11.2 (45%)
lactose, corn
maltodextrin
5.1 (46%)
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunflower oil
27 108 67
38 1.5
250
46 mg added
nucleotides, DHA 0.32%,
ARA 0.64%
PRSL 19.5/100 cals
10.9 (43.6%)
lactose
5.3 (47.7%)
27 108
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunflower oil
Parents Choice Organic 2.2 (9%)
10.6 (42%)
5.3 (48%)
23 84
(PBM Nutritionals)
organic reduced
organic lactose organic vegetable
mineral whey
oils (palm or
palm olein, high
oleic safflower or
sunflower, coconut,
soy)
Similac Advance
2.07 (8%)
11.2 (43%)
5.4 (49%)
24 105
EarlyShield
nonfat milk, whey lactose,
high oleic safflower
(Abbott Nutritionals)
protein concentrate galactose oligo- oil, soy oil, coconut
saccharides
oil
(GOS)
Similac Organic
2.07 (8%)
10.56 (42%)
5.49 (49%)
24 105
(Abbott Nutritionals)
organic nonfat dry organic corn
organic high oleic
milk
maltodextrin,
sunflower oil,
organic lactose, organic soy oil,
organic sugar
organic coconut oil
78
43 1.8
DHA, ARA
63
42 1.8
DHA, ARA
78
42 1.8
310
added nucleotides,
DHA 0.15%, ARA 0.40%
PRSL 18.7/100 cals
added prebiotics, GOS
78
42 1.8
225
132
Table 12-3 Infant Formulas for Symptoms of Intolerance (per 100 calories) Standard dilution is 0.67 kcal/mL (20 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
Bright Beginnings
Gentle
(PBM Nutritionals)
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
2.3 (9%)
10.8 (43%)
nonfat milk, whey corn syrup solids,
protein hydrolysate lactose
5.3 (48%)
palm olein, coconut
oil, soy oil, high
oleic safflower or
sunflower oil
Enfamil AR LIPIL
(Mead Johnson)
2.5 (10%)
nonfat milk
11 (44%)
lactose,
rice starch,
maltodextrin
Enfamil
Gentlease LIPIL
(Mead)
2.3 (9%)
partially
hydrolyzed
nonfat milk and
whey protein
concentrate
Phos
mg
Fe
mg
32 108 82
46
1.8
added nucleotides,
DHA, ARA
5.1 (46%)
palm olein, soy oil,
cocnut oil, high oleic
sunflower oil
40 108 78
53
DHA 17 mg,
ARA 34 mg
PRSL 22/100 cals
10.8 (43%)
corn syrup solid,
lactose
5.3 (48%)
palm olein, soy oil,
coconut oil, high
oleic sunflower oil
32 108 82
46
1.8 220
DHA 17 mg,
ARA 34 mg
PRSL 20/100 cals
Parents Choice
Gentle
(PBM Nutritionals)
2.3 (9%)
10.8 (43%)
nonfat milk, whey corn syrup solids
protein hydrolysate
5.3 (48%)
palm olein, cocnut
oil, soy oil, high
oleic safflower or
sunflower oil
32 108 82
46
1.8
DHA, ARA
Parents Choice
Lactose Free
with Lipids
(PBM Nutritionals)
2.3 (9%)
whey protein
concentrate, milk
protein isolate
10.8 (43%)
corn syrup solids
5.3 (48%)
palm olein, coconut
oil, soy oil, high
oleic safflower or
sunflower oil
32 108 82
46
1.8
DHA, ARA
Parents Choice
Sensitivity
(PBM Nutritionals)
2.6 (10%)
milk protein
isolate
10.5 (42%)
corn syrup solids,
sucrose
5.3 (48%)
palm olein, coconut
oil, soy oil, high
oleic safflower or
sunflower oil
DHA, ARA
5.46 (49%)
soy oil, cocnut oil
44 108 105 75
1.8 240
no DHA or ARA,
soy protein
Similac Sensitive
2.14 (9%)
(Abbott Nutritionals) milk protein
isolate
10.7 (43%)
corn maltodextrin,
sugar
5.4 (49%)
high oleic safflower
oil, soy oil, coconut
oil
30 107 84
56
1.8 200
DHA 0.15%,
ARA 0.40%
PRSL 19.9/100 cals
Similac
2.14 (9%)
Sensitive RS
milk protein
(Abbott Nutritionals) isolate
10.7 (43%)
corn syrup, rice
starch, sugar
5.4 (49%)
high oleic safflower
oil, soy oil, coconut
oil
30 107 84
56
1.8 180
DHA 0.15%,
ARA 0.40%
PRSL 19.9/100 cals
K
mg
Ca
mg
Osmolal- Comments
ity mOsm
133
Table 12-4 Soy-Based Infant Formulas (per 100 calories) Standard dilution for soy-based formulas is 0.67 kcal/mL (20 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
CHO g (% kcal)
Source
2.5 (10%)
enzymatically
hydrolyzed soy
protein isolate,
L-methionine
Similac Isomil
2.45 (10%)
Advance
soy protein isolate,
(Abbott Nutritionals) L-methionine
Bright Beginnings 2.5 (10%)
Soy
soy protein isolate,
L-methionine
(PBM Nutritionals)
Parents Choice
Soy DHA and ARA
(PBM Nutritionals)
2.5 (10%)
soy protein isolate
11 (44%)
corn maltodextrin,
sucrose
10.3 (41%)
corn syrup solids,
sugar
10.6 (42%)
corn syrup solids
10.6 (42%)
corn syrup solids
Fat g (% kcal)
Source
Na
mg
5.3 (48%)
palm olein, soy oil,
coconut oil, high oleic
sunflower oil
5.1 (46%)
palm olein, soy oil,
coconut oil, high
oleic safflower or
sunfloweroil
5.46 (49%)
high oleic safflower
oil, soy oil, cocnut oil
5.3 (48%)
palm olein, coconut
oil, soy oil, high oleic
safflower or sunflower
oil
5.3 (48%)
palm olein, coconut
oil, soy oil, high
oleic safflower or
sunfloweroil
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
36
120 105 69
40
116 105 63
1.8 180
DHA, ARA
PRSL 22.9/100 cals
44
108 105 75
1.8 200
36
120 105 83
1.8
DHA 0.15%,
ARA 0.40%
PRSL 22.8/100 cals
DHA 17 mg,
ARA 34 mg PRSL
36
120 105 69
1.8
DHA, ARA
PRSL 22.9/100 cals
infants specific needs.12 Soy milk protein is no less allergenic than cows milk protein. 3 Infants with documented
cows milk allergy should not be given a soy formula
because 10% to 14% of these babies will also have a sensitivity to soy protein.13,14 Infants with acute gastroenteritis
who were previously well can be managed with rehydration
and continued use of human milk or their usual cows milkbased formula at the standard dilution.12
134
acids and small peptides. The fat content is made up of longchain triglycerides (LCTs), varying amounts of MCTs, and
polyunsaturated vegetable oils to supply essential fatty acids
(EFAs). These formulas are lactose-free and because of the
hydrolyzed protein have a higher osmolarity. Protein hydrolysates are recommended in infants intolerant of cows milk
and soy proteins, and those with significant malabsorption
due to gastrointestinal or hepatobiliary disease (eg, cystic
fibrosis, short gut syndrome, biliary atresia, cholestasis,
protracted diarrhea). 3,17
Amino Acid Based
Amino acid-based infant formulas (Table 12-8) are indicated
in extreme protein hypersensitivity or when intolerance
symptoms persist on an extensively hydrolyzed formula.18
Approximately 2% to 10% of infants with cows milk protein
allergy develop persistent symptoms despite therapy with
partially hydrolyzed formula and thus require an amino
acid-based formula.19 There is no additional benefit to using
an amino acid-based formula if an extensively hydrolyzed
casein formula is effective. Other indications for the amino
Table 12-5 Premature Infant Formulas (per 100 calories) Standard dilution varies and can be 0.67 kcal/mL (20 kcal/oz) or 0.8 kcal/mL (24 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
Enfamil Premature
LIPIL
20 cal/oz (low iron or
iron fortified)
(Mead Johnson)
Enfamil Premature
LIPIL
24 cal/oz
(low iron or iron
fortified)
(Mead Johnson)
GOOD START
Premature
24 cal/oz
(Nestle/Gerber)
3 (12%)
11 (44%)
non-fat milk, whey corn syrup solids,
protein concentrate lactose
5.1 (44%)
MCT oil, soy oil, high
oleic vegetable oil
58
98
165 83
0.5 240
1.8
3 (12%)
11 (44%)
non-fat milk, whey corn syrup solids,
protein concentrate lactose
5.1 (44%)
MCT oil, soy oil, high
oleic vegetable oil
58
98
165 83
0.5 300
1.8
3 (12%)
enzymatically
hydrolyzed whey
protein isolate
(from cows milk)
3 (12%)
nonfat milk, whey
protein concentrate
10.5 (42%)
corn maltodextrin,
lactose
5.2 (46%)
MCT oil, soy oil, high
oleic safflower or
sunflower oil
55
120 164 85
1.8 275
Ca:Phos
ratio 1.9:1
10.3 (41%)
corn syrup solids,
lactose
5.43 (47%)
MCT, soy oil,
coconutoil
43
0.37 235
1.8
DHA 0.25%,
ARA0.40%, PRSL
27.8/100 cals
10.3 (41%)
3 (12%)
nonfat milk, whey corn syrup solids,
protein concentrate lactose
5.43 (47%)
MCT, soy oil,
coconutoil
43
0.37 280
1.8
DHA 0.25%,
ARA0.40%, PRSL
27.8/100 cals
3 (12%)
7.73 (31%)
nonfat milk, whey corn syrup solids,
protein concentrate lactose
6.61 (57%)
MCT, soy oil,
coconutoil
43
1.8 325
DHA 0.25%,
ARA0.40%, PRSL
27.8/100 cals
Similac Special
Care
20 cal/oz (low iron
oriron fortified)
(Abbott Nutritionals)
Similac Special
Care
24 cal/oz (low iron
oriron fortified)
(Abbott Nutritionals)
Similac Special
Care
30 cal/oz
(Abbott Nutritionals)
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
135
Table 12-6 Premature Discharge Infant Formulas (per 100 calories) Standard dilution for these formulas is 0.73 kcal/mL (22 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
Enfamil
EnfaCare LIPIL
(Mead Johnson)
2.8 (11%)
whey protein
concentrate,
nonfatmilk
CHO g (% kcal)
Source
10.4 (42%)
powder: lactose,
corn syrup solids
RTU: maltodextrin,
lactose
10.1 (40%)
Similac NeoSure 2.8 (11%)
(Abbott Nutritionals) nonfat milk, whey corn syrup solids,
protein concentrate lactose
Fat g (% kcal)
Source
Na
mg
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
5.3 (47%)
high oleic sunflower
or safflower oil, soy
oil, MCT, coconut oil
35
105 120 66
42 mg added
1.8 powder
nucleotides, DHA
260
liquid 250 17mg, ARA 34 mg,
PRSL 24/100 cals
5.5 (49%)
soy oil, coconut oil,
MCT
33
142 105 62
1.8 250
DHA 0.15%,
ARA0.40%, PRSL
25.2/100 cals
Table 12-7 Extensively Hydrolyzed Protein Infant Formulas (per 100 calories)
Product
(Manufacturer)
Protein g (% kcal)
Source
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Nutramigen LIPIL
(Mead Johnson)
2.8 (11%)
extensively
hydrolyzed casein,
L-cystine, L-tyrosine,
L-tryptophan
2.8 (11%)
extensively
hydrolyzed casein,
L-cystine, L-tyrosine,
L-tryptophan
2.8 (11%)
casein hydrolysate,
amino acids
10.3 (41%)
corn syrup solids,
modified corn
starch
5.2 (48%)
47
palm olein, soy oil,
coconut oil, high oleic
sunflower oil
10.3 (41%)
corn syrup solids,
modified corn
starch
5.3 (48%)
47
palm olein, soy oil,
coconut oil, high oleic
sunflower oil
10.2 (41%)
corn syrup solids,
dextrose, modified
corn starch
RTU-no dextrose
Similac Alimentum 2.75 (11%)
10.2 (41%)
Advance
casein hydrolysate, sugar, modified
(Abbott Nutritionals) L-cystine, L-tyrosine, tapioca starch
L-tryptophan
47
5.6 (48%)
MCT, soy oil, corn oil,
high oleic safflower or
sunflower oil
RTU-no corn oil
5.54 (48%)
44
high oleic safflower
oil, MCT, soy oil
Nutramigen with
Enflora LGG
(Mead Johnson)
Pregestimil LIPIL
(Mead Johnson)
Na
mg
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
52
118 105 75
1.8 370
DHA 0.15%,
ARA0.40%,
PRSL 25.3/100 cals
136
Table 12-8 Free Amino Acid Infant Formulas (per 100 calories)
Product
(Manufacturer)
Protein g (% kcal)
Source
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Similac EleCare
(Abbott Nutritional)
3.1 (15%)
free L-aa
10.7 (43%)
corn syrup solids
Neocate Infant
DHA & ARA
(Nutricia)
3.1 (12%)
free aa
11.7 (47%)
corn syrup solids
4.8 (42%)
45 150 116 84.2 1.5 350
high oleic safflower
oil, MCT oil, soy oil
4.5 (41%)
37.3 155.1 124 93.1 1.85 375
refined vegetable oil
(soy oil, coconut oil,
high oleic sunflower
oil)
5.3 (48%)
47 110 94 52
1.8 350
palm olein, soy oil,
coconut oil, high oleic
sunflower oil
10.3 (41%)
corn syrup solids,
modified tapioca
starch
Na
mg
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
DHA, ARA
PRSL 28/100 cals
available without
DHA & ARA also
DHA 17 mg,
ARA 34 mg,
PRSL 16.8/100 cals
Protein g (% kcal)
Source
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
RCF*
3.0 (12%)
10.1 (40%)
5.3 (48%)
44
(Abbott Nutritionals) soy protein isolate, selected by
high oleic safflower
L-methionine
physician or health oil, coconut oil, soy oil
care professional
3232A
2.8 (11%)
13.5 (54%)
4.2 (35%)
43
(Mead Johnson)
extensively
choice by physician, MCT, corn oil
hydrolyzed casein, modified tapioca
L-cystine, L-tyrosine, starch as stabilizer
L-tryptophan
*based on 100 calories when 52 gm of CHO are added to 13 oz of concentrate
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
108 105 75
1.8 168
no DHA/ARA
PRSL 25.8/100cals
109 94
1.88 type of
CHO will
influence
59 gm CHO added
to 1 quart prepared
product, PRSL
25/100 cals
63
137
manufacturers provide many different formulas in standard caloric dilution and some formulas at higher caloric
concentrations in convenient ready-to-feed nurser bottles.
Consumers can purchase ready-to-feed formula at standard dilution in quart-sized bottles or single-serving nurser
bottles. Ready-to-feed formula is the most expensive form
as the consumer is paying for the convenience.
Liquid concentrate is also considered commercially
sterile, but because it needs to be mixed with water to make
a standard dilution, it offers more opportunity for potential
contamination than ready-to-feed. Liquid concentrate is the
second choice in hospitals and can be used to make highercaloric concentrations. It is easy to mix for consumers and
offers some financial savings over ready-to-feed.
Powder is not sterile and must be mixed with water.
Because it is not sterile, it may contain pathogenic bacteria. 37
Powder formula has been associated with Enterobacter sakazakii contamination in immunocompromised neonates in
healthcare facilities.38 Because of the population that they
serve, hospitals only use powder when there is no other option
available.39 Reconstituted powder formulas have been safely
consumed by millions of infants worldwide over the past half
century, so parents of healthy newborns should continue
to feel comfortable using powder infant formulas. The
Protein g (% kcal)
Source
Enfamil Enfaport
LIPIL
(Mead Johnson)
3.5 (14%)
10.2 (41%)
calcium caseinate, corn syrup solids
sodium caseinate
5.4 (45%)
MCT (84%), soy oil
Monogen
(Nutricia)
2.7 (11%)
whey protein
concentrate,
L-amino acids
3 (12%)
2.8 (25%)
48
MCT (as fractionated
coconut oil), walnut
oil
4.6 (41%)
55
Lipistart
(vitaflo)
CHO g (% kcal)
Source
16 (64%)
corn syrup solids
12 (47%)
Fat g (% kcal)
Source
Na
mg
30
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
115 94
52
86
48
108 105 77
62
250-22
cal/oz
370-30
cal/oz
180-22
cal/oz
205-24
cal/oz
272-30
cal/oz
DHA, ARA
PRSL 29/100 cals
*comes in RTU, 30
cal/oz. Water can be
added to dilute to
lower concentrations
essential fatty acid
ratio n6:n3 of 4.6:1
*for > 1 y old
*for > 1 y old
Protein g (% kcal)
Source
Similac PM 60/40
2.2 (9%)
(Abbott Nutritionals) whey protein
concentrate,
sodium caseinate
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
10.2 (41%)
lactose
5.6 (50%)
24
high oleic safflower
oil, soy oil, coconut oil
K
mg
80
Ca
mg
56
Phos
mg
28
Fe
mg
Osmolal- Comments
ity mOsm
0.7 280
138
ESPGHAN (European Society for Pediatric Gastroenterology, Hepatology, and Nutrition) Committee on Nutrition
recommends that in the home powder infant formulas should
be freshly prepared for each feeding and any remaining milk
discarded to minimize potential risk of contamination.40
Consumers choose it because it is the least expensive form
and can be quickly mixed at any location when needed.
Water
Tap water or boiled and cooled water is adequate to use when
preparing formula for healthy infants with a normal immune
system and who are fed orally.41 Municipal tap water is more
regulated than bottled water. Municipal tap water is fluoridated whereas most bottled water is not. Bottled water or
infant fluoridated bottled water may be a good choice when
tap water is from a well because well water may contain high
levels of certain minerals. In hospitals only chilled, sterile
water is recommended in formula preparation.42
Infants consuming either human milk or infant formula
exclusively do not have a need for additional water in their diets.
Human milk or infant formula provides adequate free water for
hydration in hot or dry climates and if the infant is febrile.3
Standard Dilution
When mixing formulas, healthcare professions should
suggest that parents read the manufacturer instructions
on the can as instructions may vary by manufacturer and
by product. In powdered formulas, only use the scoop that
comes with that particular formula because scoop sizes are
different for each formula. Standard dilution for term infant
formulas is 20 kcal/oz, or 0.67 kcal/mL, and can be made
by mixing 1 scoop of powder for each 2 oz of water, or by
mixing a 13-oz can of concentrate with 13 oz of water.
Increasing Caloric Concentration
For special feeding situations, both powdered infant
formula or infant formula concentrate can be reconstituted
(Table 12-12) to provide formula with more concentrated
calories than standard dilution. 21 Concentrated liquids
from all manufacturers contain 40 kcal/oz, so the same
instructions for preparation and caloric concentration can
be used forall.
Amount of Powder/Liquid
Water
(oz)
20 kcal/oz
(0.67 kcal/mL)
Modular Macronutrients
Modular macronutrients can be used to increase caloric
concentration. Modulars are available as protein, carbohydrate, fat, and combinations of macronutrients. The fat
and/or carbohydrate modulars may not add as significantly
to the potential renal solute load (PRSL) and osmolality as
concentrating the formula with less water, and fat and/or
carbohydrate modulars should be considered when concentrating calories higher than 24 calories per ounce. Modular
macronutrients will not increase the concentration of
micronutrients like concentrating just the formula powder
or concentrate. This may be desirable in some situations
(renal insufficiency) and not desirable in other situations
(increased calcium and phosphorus needs in prematurity).
Modulars can be added to the formula when mixing
or they can be mixed with water and delivered as a bolus
through the tube separate from formula. To boost caloric
intake of formula, some common food products or ingredients may also be used (eg, table sugar, vegetable oil, or corn
starch). These ingredients may not be ideal, but they are
much less expensive than the manufactured modulars.
Increasing Concentration of Breast Milk
Infants born prematurely often have caloric and nutrient
deficits, even at discharge, and although breast milk is the
best choice for feeding these infants, it may not meet all of
the caloric and nutrient needs of the infant with significant comorbidities.43 Expert opinion and studies show
that preterm infants discharged from the hospital at suboptimal weight being fed breast milk should continue to be
supplemented to assure adequate nutrient intake.44,45 Some
healthcare professionals suggest adding premature infant
formula or infant formula powder to expressed breast milk to
increase the caloric and nutrient density. There is no evidence
for or against this practice, and there is a great potential for
error. Close medical monitoring is suggested.46
139
Toddler Formulas
Toddlers consuming adequate amounts of nutrients, especially iron, from solid foods do not need formula. Whole
cows milk is appropriate after 1 year of age. Toddler formulas
(Table 12-13) contain higher amounts of iron, vitamin C,
and vitamin E than cows milk and contain nutrients such
as zinc that cows milk does not contain. The calcium and
phosphorus levels of the toddler formulas are higher than
infant formulas to match the needs of the growing toddler.
Toddler formulas contain DHA and ARA.
Juice
After 6 months of age fruit juice can be introduced in a cup
and should be limited to 4 to 6 oz/d. 3 Only 100% fruit juice
should be offered. Fruit juice displaces the more nutrientdense human milk or infant formula and therefore should
be used in limited amounts. Infants should not be offered
juice in a bottle or in a cup that can be carried around and
should not consume juice just before bed because of the
increased risk for dental caries. Overconsumption of juice
can lead to osmotic diarrhea due to the high fructose and
sorbitol content. 3
Milk
Milk (whole, 2%, 1%, skim, goat), other than infant formula,
should be avoided during the first year of life. Milk is lower
in iron and has a higher renal solute load due to the higher
2010 A.S.P.E.N. www.nutritioncare.org
140
Table 12-13 Toddler Formulas (per 100 calories) Standard dilution for these products is 0.67 kcal/mL (20 kcal/oz).
Product
(Manufacturer)
Protein g (% kcal)
Source
CHO g (% kcal)
Source
Fat g (% kcal)
Source
Na
mg
K
mg
Ca
mg
Phos
mg
Fe
mg
Osmolal- Comments
ity mOsm
GOOD START
Gentle PLUS 2
(Nestle/Gerber)
2.2 (9%)
11.2 (45%)
whey protein
lactose, corn
concentrate
maltodextrin
(from cows milk,
enzymatically
hydrolyzed, reduced
in minerals)
5.1 (46%)
27
palm olein, soy oil,
coconut oil, high oleic
safflower or sunflower
oil
265
DHA 0.32%,
ARA 0.64%,
PRSL 146/L
GOOD START
Protect PLUS 2
(Nestle/Gerber)
2.2 (9%)
11.2 (45%)
whey protein
lactose, corn
concentrate
maltodextrin
(from cows milk,
enzymatically
hydrolyzed, reduced
in minerals)
5.1 (46%)
27
palm olein, soy oil,
coconut oil, high oleic
safflower or sunflower
oil
265
DHA 0.32%,
ARA 0.64%,
PRSL 146/litre
*added probiotics,
Bifidobacteria
lactis
Enfagrow
Premium
NextStep
(Mead Johnson)
2.6 (10%)
non-fat milk
10.5 (42%)
5.3 (48%)
36
lactose, corn syrup palm olein, soy oil,
solids
coconut oil, high oleic
sunflower oils
270
DHA 17 mg,
ARA 34 mg,
PRSL 26/100cals
300
DHA 0.15%,
ARA 0.40%,
PRSL 20/100 cals
5.49 (49%)
high oleic safflower
oil, soy and coconut
oils
24
105 150 81
10.5 (42%)
corn syrup solids,
lactose
5.3 (48%)
vegetable oils
36
DHA 17 mg,
ARA 34 mg,
PRSL 26/100cals
10.5 (42%)
corn syrup solids,
lactose
5.3 (48%)
vegetable oils
36
DHA 17 mg,
ARA 34 mg,
PRSL 26/100cals
* with added
prebiotic, FOS
Parents Choice
Stage 2 Formula
(PBM Nutritionals)
2.6 (10%)
non-fat milk
10.5 (42%)
corn syrup solids,
lactose
5.3 (48%)
36
palm olein, soy oil,
coconut oil, high oleic
safflower oil
DHA, ARA
2.8 (11%)
enzymatically
hydrolyzed soy
protein isolate
10.9 (44%)
corn maltodextrin,
sucrose
5.0 (45%)
40
palm olein, soy oil,
coconut oil, high oleic
safflower or sunflower
oil
180
DHA 0.32%,
ARA 0.64%,
PRSL 175/liter
5.46 (49%)
49
high oleic safflower
oil, soy oil, coconut oil
200
DHA 0.15%,
ARA 0.40%,
PRSL 23.7/100 cals
Enfagrow Soy
Next Step
(Mead Johnson)
4.4 (40%)
36
palm olein, soy oil,
coconut oil, high oleic
sunflower oils
230
DHA 17 mg,
ARA 34 mg,
PRSL 30/100 cals
3.3 (13%)
11.8 (47%)
soy protein isolate, corn syrup solids
L-methionine
References
141
142
13
Timothy Sentongo, MD
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143
Growth Assessment. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 144
Weight
Height (Length/Stature)
Head Circumference
Weight-for-Length
Failure to Thrive
Body Mass Index (BMI)
Ideal Body Weight (IBW)
Learning Objectives
Introduction
144
Growth Assessment
Weight
Infants should be undressed to the diaper and older children
measured in light clothing using an age-appropriate scale.
Measurements should be in metric units (kg) and rounded
off to 1 decimal point.
Age Group
Pre-term
infants
Birth
to 6 mo
Weight (kg)
636 mo
220 y
Pre-term
infants
Length (cm)
Birth to 6 mo
636 mo
Height (cm)
220 y
Birth to 6 mo
Head circumference
(cm)
636 mo
BMI (kg/m2)
220 y
Soft tissue measurements
Mid-arm circumference (mm)
Triceps skinfold (mm)
Short-Term
Long Term
Follow-Up
Daily
Weekly
12 wk
14 wk
28 wk
2 mo
6 mo
12 mo
4 wk
4 wk
2 mo
6 mo
612 mo
2 mo
6 mo
12 mo
36 mo
4 wk
4 wk
4 wk
312 mo
112 mo
The updated 2000 CDC age- and gender-based weightfor-age growth reference charts for children aged birth to
36months and 2 to 20 years should be used (see Chapter
33). Individualized weight-for-age percentiles and SD scores
(z scores) may be computed using the free access CDC Epi
Info nutrition calculator and statistics program. 6 Diseasespecific growth charts are available for Down syndrome,7
achrondroplasia, 8 and other genetic disorders. 3
Height (Length/Stature)
Linear growth status (cm) is influenced by hereditary
factors, nutrition, chronic disease, and genetic disorders.
Correct equipment and measuring technique are important
for obtaining reliable assessments. Accuracy is improved by
repeating the measurements and obtaining an average. Serially obtained growth measurements should be plotted on
age- and gender-appropriate CDC growth reference charts.
Length (supine) is measured to the nearest 0.1 cm in
children younger than 3 years or older children who cannot
stand. Length should be assessed using a length board
(stadiometer) or firm surface. It requires 2 measurers: one
to position the head and the other to stretch and straighten
the legs so that the knees are flat and feet at a 90-degree
angle with the footboard. 3
Standing height is measured to the nearest 0.1 cm. It is
obtained after age 2 years in children able to support their
weight evenly on both feet. Subjects should stand barefooted
with heels, buttocks, shoulders, and back of head against the
measuring device and eyes looking straight ahead. 3
The updated 2000 CDC age- and gender-based
Head Circumference
Brain growth occurs most rapidly from birth to age 36
months and thereafter slows down. Head circumference
(cm) must be routinely measured using a non-stretchable
measuring tape. Anteriorly the tape is placed just superior to the eyebrows and posteriorly it is placed so that the
maximum circumference is measured. 3
Impaired brain growth and size is a rare complication
of chronic malnutrition and is not characteristic of primary
skeletal disorders. Therefore, children with disproportionately sized heads should be evaluated for other disorders
impacting brain or skull size.
Weight-for-Length
Weight-for-length percentiles provide a means of assessing
a childs weight while taking into account his or her length.
They are used to screen overweight and underweight in
children aged < 36 months. See 2000 CDC weight-forlength growth reference charts for children aged birth to
36months.
Increased weight-for-length percentile > 90% indicates overweight.11 When associated with significantly
decreased length percentile (< 5%), increased weight-for-
145
Failure to Thrive
The term FTT is not a diagnosis or specific disease entity
but denotes weight gain or linear growth that is less than
expected for age. FTT may be referred to as wasting when
weight is disproportionately affected vs. stunting when
length/height is significantly impaired.
Wasting may occur following an acute illness. The
growth changes are: weight-for-length percentile or BMI
percentile < 5th percentile (~2 SD) or involuntary lack of
weight gain or loss resulting in dropping below any 2 major
percentile channels (95th, 90th, 75th, 50th, 25th, 10th,
and5th).
Stunting occurs following chronic inadequate caloric
intake, disease, or endocrinopathy. The diagnostic growth
findings are: length or height < 5th percentile (~2 SD) or
consecutive height measurements dropping below any
2major percentile channels.
Marasmus is moderate-to-severe FTT without edema.
Kwashiorkor is moderate-to-severe FTT in association with
hypoalbuminemia and edema.
The WHO classifies FTT as moderate or severe using
criteria of weight-for-length/height and length/height-for-age
SD scores and presence or absence of edema (Table 13-2).
Table 13-2 World Health Organization Classification of Malnutrition12
Classification
Edema
Moderate
Severe
No
Yes (Edematous
malnutrition)
Weight-for-length
3 to 2
< 3 (wasting)
z score*
Height-for-age z score*
3 to 2
< 3 (stunting)
*Growth channels of WHO international growth charts are along z scores
3; 2; 1; 0; 1; 2 and 3.
146
Corresponding z score
0.1
3rd
16th
50th
84th
97th
99.9th
3
2
1
0
1
2
3
147
3. The CDC recommends terminology of obesity in children with BMI > 95th percentile.
A. True
B. False
4. Diagnosis of FTT or wasting is based on each of the
following except:
A. Length < 3rd percentile
B. Weight-for-length or BMI < 3rd percentile
C. Change in weight percentile from 90th to 50th
D. Change in weight percentile from 50th to 25th
5. The 50th percentile weight, height, or BMI for age
corresponds with the following z score:
A. 2
B. 1
C. 0
D. 1
E. 2
6. Which of the following is incompatible with survival?
A. BMI < 3rd percentile
B. Change in weight percentile from 75th to 5th
percentile
C. Height z score 5
D. 60% of ideal body weight
7. Length measurements in an 18-month-old toddler are
obtained with the child standing upright.
A. True
B. False
See p. 487 for answers.
References
1. Tanner JM, Goldstein H, Whitehouse RH. Standards for childrens height at ages 2-9 years allowing for heights of parents.
Arch Dis Child. 1970;45(244):755762.
2. Ulijaszek SJ. Measurement error. In: Ulijaszek SJ, Johnston
FE, Preece MA, eds. The Cambridge Encyclopedia of Human
Growth and Development. Cambridge, UK: Cambridge
University Press; 1998:28.
3. Zemel BS, Riley EM, Stallings VA. Evaluation of methodology for nutritional assessment in children: anthropometry,
body composition, and energy expenditure. Annu Rev Nutr.
1997;17:211235.
4. Owen GM. The assessment and recording of measurements
of growth of children: report of a small conference. Pediatrics.
1973;51(3):461466.
5. Tanner JM. Normal growth and techniques of growth assessment. Clin Endocrinol Metab. 1986;15(3):411451.
6. Centers for Disease Control and Prevention. Epi Info Downloads. http://www.cdc.gov/epiinfo/downloads.htm. Accessed
November 12, 2009.
7. Myrelid A, Gustafsson J, Ollars B, Anneren G. Growth charts
for Downs syndrome from birth to 18 years of age. Arch Dis
Child. 2002;87(2):97103.
2010 A.S.P.E.N. www.nutritioncare.org
148
14
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149
What Is the Metabolic Syndrome in Adults? . . . . . . . . . . 150
Metabolic Syndrome Controversies AmongAdults . . . . . 150
Cut-points
Etiology
Synergistic Risk and Treatment
Learning Objectives
Introduction
Since the early 1900s evidence has suggested the coexistence of chronic disorders associated with diabetes mellitus
and coronary heart disease (CHD).1 It wasnt until the
years between 1980 and 1990 that scientists developed an
understanding of the mechanisms underlying the connection between obesity, hypertension, hyperlipidemia, and
type 2 diabetes mellitus and its relationship to increased
cardiovascular disease (CVD) risk. Metabolic syndrome
was a term first used to describe how environmental and
genetic factors work together to produce this constellation
of chronic metabolic disorders.1 However, it was the earlier
work of Gerald Reaven2 that set the precedent for how the
metabolic syndrome is applied within the clinical world
today. In 1988, Reavens Banting Lecture described the relationship between hyperinsulinemia, glucose intolerance,
hypertension, and free-fatty acid metabolism and their
association with CVD. Reaven hypothesized that the state
of insulin resistance was the driver for metabolic change and
represented the common pathophysiological link between
these otherwise unrelated metabolic events.2
Through the 1990s, distinct perspectives on the etiology
of metabolic syndrome began to emerge, coupled with
the inclusion of additional biomarkers such as measures
149
150
The early work of Hanefield and Reaven described the metabolic syndrome as a constellation of metabolic risk factors
including obesity, hypertension, dyslipidemia, and glucose
intolerance, all of which are associated with increased cardiovascular mortality. However, today the metabolic syndrome
is no longer viewed as a novel concept, but has surfaced as a
Table 14-1 Etiology and Diagnostic Criteria of the Metabolic Syndrome in Adults
National Cholesterol
Education Program, Adult
Treatment Panel III (NCEPATP III)6
World Health Organization
(WHO)7
International Diabetes
Federation (IDF)8
Diagnostic Criteria
Etiological Underpinnings
Recommendations
Cut-points
Evidence has failed to identify appropriate cut-point values
for any given metabolic component. Worldwide adoption
of Westernized eating and physical activity behaviors has
resulted in an increased prevalence of obesity, diabetes,
stroke, and heart disease in what were once healthy populations. Researchers have learned that the degree of metabolic
risk associated with developing CVD is distinct among
people with diverse ethnic backgrounds. One good example
is the differences of dyslipidemias among various ethnic
groups. In the African American population, the standard cutoff values for triglycerides and HDL-cholesterol
predicted insulin resistance is only 17%.10 Furthermore
insulin resistance and triglycerides were found to be
inverselycorrelated (ie, as insulin levels rose with insulin
resistance, triglycerides fell). In this way markers for dyslipidemia within the range of normal may prove insensitive
and fail to identify individuals, particularly African Americans, who are insulin resistant and at risk for cardiovascular
damage.11 Furthermore, ethnic differences in metabolic risk
have been reported for measures of waist circumference
and hypertension among African Americans, Hispanics,
Caucasians, Iranians, and Asians.9,1215 These findings
justify the need to assess the obese patient with a tailored
approach to capture global metabolic risks.
Etiology
Controversy around the metabolic syndrome has
surrounded the criteria utilized to make the diagnosis.
Because the etiological underpinnings of the metabolic
syndrome are substantially altered by ethnic variability
it is difficult to build a single, all-inclusive definition.1,4,9
Since Reaven, the basic connecting point uniting the
various components has been insulin resistance. Yet even
in this there is controversy. Some believe that the primary
pathway leading to the metabolic syndrome is the result of
glucose intolerance and diabetes9 whereas others suggest
that glucose has no direct role in the metabolic syndrome
or insulin resistance.16,17 These individuals promote the
surprising hypothesis that it is disordered fat metabolism,
not glucose metabolism, which is the etiologic prime mover
for development of insulin resistance and, as a result, the
metabolic syndrome. Although the current research has not
yet detailed the sequence of events leading to the insulinresistant state that precedes the metabolic syndrome,
mounting evidence supports the role of abnormal fat
metabolism.16,17
151
152
153
154
Acanthosis Nigricans
Insulin resistance and subsequent hyperinsulinemia initiate
a series of cascading metabolic events signaling total body
changes in metabolism. Ultimately the clustering of metabolic parameters, as seen in the metabolic syndrome, brings
attention to the serious health risks that are associated with
insulin resistance and obesity. Furthermore, such metabolic
changes identify individuals in most urgent need for lifestyle
intervention. Essentially, identifying certain physical signs
may alert clinicians early in the clinical course.
Risk associated with family history, socioeconomic
status, and nutrition and physical activity behaviors may be
effective screening tools for prevention activities. However,
as evidence suggests, a substantial number of children
and teens already have acquired the first signs of metabolic changes, which will with time place a serious health
burden on their cardiovascular system. Unlike family
history, ethnicity, lifestyle, behavior, and body mass index
(BMI) that predict future health burdens, the skin sign of
acanthosis nigricans represents a physical manifestation of
existing metabolic change.
Several studies indicate that acanthosis nigricans
is relatively common among children and adolescents,
particularly those who are obese. In a broad population
155
The focused review of systems and targeted physical examination of the child should be comprehensive in nature and
include, but not be limited to, the presence of anxiety, polyuria/dipsia, headaches, sleep problems, abdominal pain (a
focused review of systems); acanthosis nigricans, the presence of dysmorphic features, hirsutism and extreme acne,
tonsillar hypertrophy, abdominal tenderness, unexpected
rates of linear growth, and undescended testicles. A crucial
part of the childs physical exam should be the blood pressure, but it must be obtained using the correct cuff size at
rest. The results are then assessed using tables comparing
systolic and diastolic readings against normals for the
childs height percentile to ascertain at risk values over the
90th percentile and hypertensive values above the 95th
percentile.76
Due to the lack of a standard definition to diagnose the metabolic syndrome in the adult and pediatric populations, the
best course of treatment for individuals with the syndrome
remains to be determined. Experts disagree on whether the
metabolic syndrome should be treated differently from the
treatment prescribed for the individual components of the
syndrome. Some counsel a more comprehensive approach
to treatment for patients with the metabolic syndrome. Yet
what we do know is the chronic disease components that
derive the metabolic syndrome are a direct result of excess
adiposity and subsequent insulin resistance. Therefore,
national directives established by an expert committee
may be used as a guideline to assess overweight and obese
children and adolescents for health risks and mediate with
the appropriate lifestyle and treatment interventions. These
evidence-supported guidelines were published as a supplement to Pediatrics in December 2007, titled the Expert
Committee Recommendations Regarding the Prevention,
Assessment, and Treatment of Child and Adolescent Overweight and Obesity.56 There are 6 steps to consider when
assessing a childs health risk in a practice-based setting:
BMI Percentile
At least annually, but ideally at each well child visit, the
childs height and weight should be measured and the BMI
percentile value should be calculated and plotted on the
growth chart. The pediatrician should be looking for BMI
percentile trends that show an increasing weight-for-height
trajectory and classify the child as underweight, normal
weight, overweight, or obese. 56,75
Behaviors
A targeted history should capture information about nutrition and physical activity habits of the overweight or obese
child and the family. This information should serve as the
baseline and basis for prevention and intervention counseling directed both at the child and the family.
Laboratory Analysis
More invasive testing is required for overweight and obese
children to identify health risks that may be otherwise
hidden. It is important to note that Expert Committee56
guidelines on further laboratory testing mirror most of
the components that comprise the metabolic syndrome.
Table 14-2 lists what labs should be drawn at what degree of
obesity and at what age. The physician also should explore
concerns raised during the history and physical exam,
investigated fully along with the testing recommended for
all overweight children.
The above have been described by the Expert
Committee56 more explicitly and provide the pediatrician
with practice-ready guidelines to assess the global health
risk of the overweight or obese child. Furthermore, these
2010 A.S.P.E.N. www.nutritioncare.org
156
Table 14-2 Laboratory Testing Guidelines Based on Age, BMI Percentile, and Risk Factors Present
Laboratory Testing
Parameters
Age 29 years
BMI %
85th95th (with no additional risk factors*)
BMI %
> 85th95th (with additional risk factors*)
BMI %
> 95th99th (with or without additional risk factors*)
BMI %
> 99th (with or without additional risk factors*)
Age 1018 years
BMI %
85th95th (with no additional risk factors*)
BMI %
> 85th95th (with additional risk factors*)
BMI %
> 95th99th (with or without additional risk factors*)
BMI %
> 99th (with or without additional risk factors*)
Fasting Glucose
(Every 2 years)
X
X
X
X
X
X
*Risk factors refer to those risks found during the assessment of family history and physical examination. The laboratory guidelines recommended by the
Expert Committee are merely baseline recommendations. Any concerns found during the assessment of the family history and/or physical examination
should be evaluated and monitored.
recommendations emphasize the need for risk identification to occur sooner in the childs life, rather than later,
as identification of these health risks ultimately leads to
the establishment of an intervention program targeted at
reducing obesity-related comorbidities and controlling
body weight.
children in their first decade of life. The committee recommends screening children with risk factors beginning at the
age of 2 years with close follow-up, particularly where there
is a strong family history.85
157
158
Summary
References
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis
and classification of diabetes mellitus provisional report of a
WHO consultation. Diabet Med. 1998;15:539553.
International Diabetes Federation. The IDF consensus worldwide definition of the metabolic syndrome. 2006. www.idf.
org/webdata/docs/IDF_Meta_def_final.pdf.
Kahn JM, Beevers DG. Management of hypertension in ethnic
minorities. Heart. 2005;91:11051109.
Sumner AE, Finley KB, Genovese DJ, Criqui MH, Boston
RC. Fasting triglyceride and triglyceride-HDL cholesterol
ratio are not markers of insulin resistance in African Americans. Arch Intern Med. 2005:165:13951400.
Sumner AE, Cowie CC. Ethnic differences in the ability of
triglyceride levels to identify insulin resistance. Atherosclerosis. 2008;196:696703.
Zhu S, Heymsfield SB, Toyoshima H, Wang Z, Piertrobelli A,
Heshka S. Race-ethnicity-specific waist circumference cutoffs
for identifying cardiovascular disease risk factors. Am J Clin
Nutr. 2005;81:409415.
Esteghamati A, Ashraf H, Rashidi A, Meysamie A. Waist
circumference cut-off points for the diagnosis of metabolic syndrome in Iranian adults. Diabetes Res Clin Prac.
2008;82:104107.
Oka R, Kobayashi J, Yagi K, et al. Reassessment of the cutoff
values of waist circumference and visceral fat area for identifying Japanese subjects at risk for the metabolic syndrome.
Diabetes Res Clin Pract. 2008;79:474481.
Ferdinand KC, Saunders E. Hypertension related morbidity
and mortality in African Americanswhy we need to do
better. J Clin Hypertens. 2006;8: 2131.
Unger J. Reinventing Type 2 diabetes: pathogenesis, treatment, and prevention. JAMA. 2008;299:11851187.
Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat
storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr Rev. 2002;23:201229.
Iribarren C, Go AS, Husson G, et al. Metabolic syndrome and
early-onset coronary artery disease: is the whole greater than
its parts? J Am Coll Cardiol. 2006;48:18001807.
Alexander CM, Landsman PB, Teutsch SM, Haffner SM.
NCEP-defined metabolic syndrome, diabetes, and prevalence
of coronary heart disease among NHANES III participants
age 50 years and older. Diabetes. 2003;52:12101214.
McNeill AM, Rosamond WD, Girman CJ, et al. The metabolic syndrome and 11 year risk of incident cardiovascular
disease in the atherosclerosis risk in communities. Diabetes
Care. 2005;28: 385390.
Hoang KC, Ghanderhari H, Lopez VA, Barboza MG, Wong
ND. Global coronary heart disease assessment of individuals with the metabolic syndrome in the U.S. Diabetes Care.
2008;31:14051409.
Tong W, Lai H, Yang C, Ren S, Dai S, Lai S. Age, gender, and
metabolic syndrome related coronary heart disease in U.S.
adults. Int J Cardiol. 2005;104:288291.
Ford ES, Li C. Defining the metabolic syndrome in children
and adolescents: will the real definition please stand up? J
Pediatr. 2008;152:160164.
159
24. Weiss R, Dziura J, Burgert TS, et al. Obesity and the metabolic syndrome in children and adolescents. N Engl J Med.
2004;89:108113.
25. Cook S, Weitzman M, Auinger P, Nguyen M, Dietz WH.
Prevalence of metabolic syndrome phenotypes in adolescents: findings from the third national health and nutrition
examination survey, 1998-1994. Pediatr Adolesc Med.
2003;157:821827.
26. de Ferranti SD, Gauvreau K, Ludwig DS, Neufeld EJ,
Newburger JW, Rifai N. Prevalence of the metabolic
syndrome in American adolescents: findings from the third
national health and nutrition examination survey. Circulation.
2004;110:24942497.
27. Brambilla P, Lissau I, Flodmark CE, et al. Metabolic risk factor
clustering estimation in children: to draw a line across pediatric metabolic syndrome. Int J Obes. 2007;31:591600.
28. Steinberger J, Daniels SR, Eckel RH, et al. Progress and
Challenges in Metabolic Syndrome in Children and Adolescents: A Scientific Statement From the American Heart
Association Atherosclerosis, Hypertension, and Obesity in
the Young Committee of the Council on Cardiovascular
Disease in the Young; Council on Nursing; and Council on
Nutrition, Physical Activity, and Metabolism. Circulation.
2009;119:628647.
29. Huang TT. Finding thresholds of risk for components of the
pediatric metabolic syndrome. J Pediatr. 2008;152:158159.
30. Zimmet P, Alberti KG, Kaufman F, et al; IDF Consensus
Group. The metabolic syndrome in children and adolescents:
an IDF consensus report. Pediatr Diabetes. 2007; 8:299306.
31. Joliffe CJ, Janssen I. Development of age specific adolescent
metabolic syndrome criteria that are linked to the Adult Treatment Panel III and International Diabetes Federation criteria.
J Am Coll Cardiol. 2007;27:891898.
32. Moreno LA, Pineda I, Rodriquez G, Fleta J, Sarria A, Bueno M.
Waist circumference for the screening of metabolic syndrome
in children. Acta Pediatr. 2002;91:13071312.
33. Seidell JC, Perusse L, Despres JP, Bouchard C. Waist and
hip circumference have independent and opposite effects on
cardiovascular disease risk factors: the Quebec family study.
Am J Clin Nutr. 2001;74:315321.
34. Moreno LA, Joyanes M, Mesana MI, et al; AVENA Study
Group. Harmonization of anthropometric measurements for
a multicenter nutrition survey in Spanish adolescents. Nutrition. 2003;19:481486.
35. Moran A, Jacobs DR Jr, Steinberger J, et al. Association
between the insulin resistance of puberty and the insulin-like
growth factor/growth hormone axis. J Clin Endocrinol Metab.
2002;87:48174820.
36. Hannon TS, Janosky J, Arslanian SA. Longitudinal study
of physiological insulin resistance and metabolic changes of
puberty. Pediatr Res. 2006;60:759763.
37. Groner JA, Joshi M, Bauer JA. Pediatric precursors of adult
cardiovascular disease: noninvasive assessment of early
vascular changes in children and adolescents. Pediatrics.
2006;118(4):16831691.
38. Woo KS, Chook P, Yu CW, et al. Overweight in children is associated with arterial endothelial dysfunction and intima-media
thickness. Int J Obes Relat Metab Disord. 2004;28:852857.
2010 A.S.P.E.N. www.nutritioncare.org
160
39. Ferri C, Croce G, Confini V, et al. C-reactive protein: interaction with the vascular endothelium and possible role in human
atherosclerosis. Curr Pharm Des. 2007;13:16311645.
40. Valle M, Maros R, Gascon F, Canete R, Zafra MA, Morales R.
Low-grade systemic inflammation, hypoadiponectinemia and
a high concentration of leptin are present in very young obese
children, and correlate with metabolic syndrome. Diabetes
Metab. 2005; 3:5562.
41. Soriano-Guillen L, Hernandez-Garcia B, Pita J,
Dominguez-Garrido N, Del Rio-Camacho G, Rovira A.
High-sensitivity C-reactive protein is a good marker of
cardiovascular risk in obese children and adolescents. Eur J
Endocrinol. 2008;159:R1R4.
42. Poirier P, Giles TD, Bray GA, et al. Obesity and cardiovascular
disease: pathophysiology, evaluation and effect of weight loss.
Arterioscler Thromb Vasc Biol. 2006;26:968976.
43. Pai JK, Pischon T, Ma J, et al. Inflammatory markers and the
risk of coronary heart disease in men and women. N Engl J
Med. 2004;351:25992610.
44. Wozniak SE, Gee LL, Wachtel MS, Frezza EE. Adipose tissue:
the new endocrine organ? A review article. Dig Dis Sci. 2009
Sep;54(9):1847-56. Epub 2008 Dec 4.
45. Antuna-Puente B, Feve B, Fellahi S, Bastard JP. Adipokines:
the missing link between insulin resistance and obesity.
Diabetes Metab. 2008;34:211.
46. Fox CS, Massaro JM, Hoffmann U, et al. Abdominal visceral
and subcutaneous adipose tissue compartments: association
with metabolic risk factors in the Framingham Heart Study.
Circulation. 2007;116:3948.
47. Pou KM, Massaro JM, Hoffmann U, et al. Visceral and
subcutaneous adipose tissue volumes are cross-sectionally
related to markers of inflammation and oxidative stress: the
Framingham Heart Study. Circulation. 2007;116:12341241.
48. Lee S, Bacha F, Gungor N, Arslanian S. Comparison of
different definitions of pediatric metabolic syndrome: relation to abdominal adiposity, insulin resistance, adiponectin,
and inflammatory biomarkers. J Pediatr. 2008;152:177184.
49. Retnakaran R, Zinman B, Connelly PW, Harris SB, Hanley
AJG. Nontraditional risk factors in pediatric metabolic
syndrome. J Pediatr. 2006;148:176182.
50. Langenberg C, Bergstrom J, Scheidt-Nave C, Pfeilschifter J,
Barrett-Connor E. Cardiovascular death and the metabolic
syndrome: role of adiposity signaling hormones and inflammatory markers. Diabetes Care. 2006;29:13631369.
51. Kapiotis S, Holzer G, Schaller G, et al. A pro-inflammatory
state is detectable in obese children and is accompanied by
functional and morphological vascular changes. Arterioscler
Thromb Vasc Biol. 2006;26:25412546.
52. Schwimmer JB, Deutsch R, Kahen T, Lavine JE, Stanley C,
Behling C. Prevalence of fatty liver in children and adolescents. Pediatrics. 2006;118:13881393.
53. Manco M, Marcellini M, Devito R, Comparcola D, Sartorelli
MR, Nobili V. Metabolic syndrome and liver histology
in paediatric non-alcoholic steatohepatitis. Int J Obes.
2008;32:381387.
54. Schwimmer JB, Pardee PE, Lavine JE, Blumkin AK, Cook
S. Cardiovascular risk factors and the metabolic syndrome
in pediatric non-alcoholic fatty liver disease. Circulation.
2008;15:277283.
55. Barshop NJ, Sirlin CB, Schwimmer JB, Lavine JE. Review
article: epidemiology, pathogenesis and potential treatment of
paediatric non-alcoholic fatty liver disease. Aliment Pharmacol
Ther. 2008;28:1324.
56. Barlow SE; Expert Committee. Expert committee recommendations regarding the prevention, assessment, and treatment
of child and adolescent overweight and obesity: summary
report. Pediatrics. 2007;120:S164S192.
57. Dallo FJ, Weller SC. Effectiveness of diabetes mellitus
screening recommendations. Proc Natl Acad Sci.
2003;100:1057410579.
58. Lin HF, Boden-Albala B, Juo SH, Park N, Rundek T, Sacco
RL. Heritabilities of the metabolic syndrome and its components in the Northern Manhattan study. Diabetologia.
2005;48:2006-2012.
59. Butte NF, Comuzzie AG, Cole SA, et al. Quantitative genetic
analysis of the metabolic syndrome in Hispanic children.
Pediatr Res. 2005;58:12431248.
60. Goodman E. The role of socioeconomic status gradients in
explaining differences in US adolescents health. Am J Public
Health. 1999;89:15221528.
61. Chen E, Martin AD, Matthews KA. Trajectories of socioeconomic status across childrens lifetime predict health.
Pediatrics. 2007;120:e297e303. doi:10.1542/peds.20063098.
62. Wang Y. Cross national comparison of childhood obesity: The
epidemic and the relationship between obesity and socioeconomic status. Int J Epidemiol. 2001;30:11291136.
63. Singh SG, Kogan MD, Van Dyck PC, Siahpush M. Racial/
ethnic, socioeconomic, and behavioral determinants of
childhood and adolescent obesity in the United States:
analyzing independent and joint associations. Ann Epidemiol.
2008;18:682695.
64. American Dietetic Association. Factors associated with childhood overweight. http://www.adaevidencelibrary.com/topic.
cfm?cat=2792. Accessed December 3, 2008.
65. Marshall SJ, Gorely T, Biddle SJH. A descriptive epidemiology
of screen-based media use in youth: a review and critique. J
Adolesc. 2006;29:333349.
66. Kotz K, Story M. Food advertisements during childrens
Saturday morning television programming: are they consistent with dietary recommendations? J Am Diet Assoc.
1994;29:12961300.
67. Halford JC, Gillespie J, Brown V, Pontin EE, Dovey TM. Effect
of television advertisements of foods on food consumption in
children. Appetite. 2004;42:221225.
68. Halford JC, Boyland EJ, Hughes GM, Stacey L, McKean S,
Dovey TM. Beyond brand-effect of television food advertisements on food choice in children: the effects of weight status.
Public Health Nutr. 2008;11:897904.
69. Strong WB et al. Evidence based physical activity for schoolage youth. J Pediatr. 2005;146:732737.
70. Nguyen TT, Keil MF, Russell DL, et al. Relation of acanthosis
nigricans to hyperinsulinemia and insulin sensitivity in
overweight African American and white children. J Pediatr.
2001;138(4)474480.
71. Mukhtar Q , Cleverley G, Voorhees RE, McGrath JW.
Prevalence of acanthosis nigricans and its association with
hyperinsulinemia in New Mexico adolescents. J Adoles Health.
2001;28:372376.
72. Sinha S, Schwartz RA. Juvenile acanthosis nigricans. J Am
Acad of Dermatol. 2007;57:502508.
73. Ice CL, Murphy E, Minor VE, Neal WA. Metabolic syndrome
in fifth grade children with acanthosis nigricans: results from
the CARDIAC project. World J Pediatr. 2009;5:2330.
74. Guran T, Turan S, Akcay T, Bereket A. Significance of acanthosis nigricans in childhood obesity. J Paediatr and Child
Health. 2008;44:338341.
75. Murray R, Battista M. Managing the risk of childhood overweight and obesity in primary care practice. Curr Probl Pediatr
Adoles Health Care. 2009;39:145166.
76. National High Blood Pressure Education Program Working
Group on High Blood Pressure in Children and Adolescents.
The fourth report on the diagnosis, evaluation, and treatment
of high blood pressure in children and adolescents. Pediatrics.
2004;114(suppl):555576.
77. Freedman DS, Khan LK, Serdula MK, Dietz WH, Srinivasan
SR, Berenson GS. Racial differences in the tracking of childhood BMI to adulthood. Obes Res. 2005;13:928935.
78. Katzmarzyk PT, Prusse L, Malina RM, Bergeron J, Desprs
JP, Bouchard C. Stability indicators of the metabolic syndrome
from childhood and adolescence to young adulthood: the
Quebec family study. J Clin Epidemiol. 2001;54:190195.
79. Morrison JA, Fredman LA, Wang P, Glueck CJ. Metabolic
syndrome in childhood predicts adult metabolic syndrome
and type 2 diabetes mellitus 25-30 years later. J Pediatr.
2008;152:201206.
80. Whitaker RC, Wright JA, Pepe MS, Seidel KD, Dietz WH.
Predicting obesity in young adulthood from childhood and
parental obesity. N Engl J Med. 1997;337:869873.
81. Golley RK, Magarey AM, Steinveck KS, Baur LA, Daniels
LA. Comparison of metabolic syndrome prevalence using
six different definitions in overweight pre-pubertal children enrolled in a weight management study. Int J Obes.
2006;30:853860.
82. Perichart-Perera O, Balas-Nakash M, Schiffman-Selechnik
E, Barbato-Dosal A, Vadillo-Ortega F. Obesity increases
metabolic syndrome risk factors in school-aged children
from an urban school in Mexico City. J Am Diet Assoc.
2007;107:8191.
83. Dubois, K et al. Prevalence of the metabolic syndrome
in elementary school children. Acta Paediatrica.
2006;95:10051011.
84. Braunschweig CL, Gomez S, Liang H, et al. Obesity and risk
factors for the metabolic syndrome among low-income, urban,
African American schoolchildren: the rule rather than the
exception? Am J Clin Nutr. 2005;81:970975.
161
85. Daniels SR, Greer FR, and the Committee on Nutrition. Lipid
screening and cardiovascular health in childhood. Pediatrics.
2008;122:198208.
86. McBride PE, Einerson JA, Grant H, et al. Putting the diabetes
prevention program into practice: a program for weight
loss and cardiovascular disease reduction with metabolic
syndrome or type 2 diabetes mellitus. J Nutr Health Aging.
2008;12:745s749s.
87. Racette SB, Weiss EP, Hickner RC, Holloszy JO. Modest
weight loss improves insulin action in African Americans.
Metabolism. 2005;54:960965.
88. Moore LL, Singer MR, Bradlee ML, et al. Intake of fruits,
vegetables and dairy products in early childhood and subsequent blood pressure changes. Epidemiol. 2005;16:411.
89. Couch SC, Saelens BE, Levin L, Dart K, Falciglia G, Daniels
S. The efficacy of a clinic based behavioral nutrition intervention emphasizing a DASH-type diet for adolescents with
elevated blood pressure. J Pediatr. 2008;152:494502.
90. Thompson PD, Buchner D, Pina IL, et al. American Heart
Association Council on Clinical Cardiology Subcommittee
on Exercise, Rehabilitation, and Prevention; American
Heart Association Council on Nutrition, Physical Activity,
and Metabolism Subcommittee on Physical Activity. Exercise and Physical Activity in the Prevention and Treatment
of Atherosclerotic Cardiovascular Disease: A Statement
from the Council of Clinical Cardiology and the Counsel
on Nutrition, Physical Activity and Metabolism. Circulation.
2003;24:31093116.
91. Hopkins ND, Stratton G, Tinken TM, et al. Relationships
between measures of physical fitness, physical activity, body
composition, and vascular function in children. Atherosclerosis. 2009; 204:244249.
92. Meyer AA, Kundt G, Lenschow U, Schuff-Werner P, Kienast
W. Improvement of early vascular changes and cardiovascular risk factors in obese children after a six-month exercise
program. J Am Coll Cardiol. 2006;7:18651870.
93. Watts K, Beye P, Siafarikas A, et al. Exercise training normalizes vascular dysfunction and improves central adiposity in
obese adolescents. J Am Coll Cardiol. 2004;19:18231827.
94. McCrindle BW, Urbina EM, Dennison BA, et al. American
Heart Association Atherosclerosis, Hypertension, and
Obesity in Youth Committee; American Heart Association
Council of Cardiovascular Disease in the Young; American
Heart Association Council on Cardiovascular Nursing.
Drug therapy of high-risk lipid abnormalities in children and
adolescents: a scientific statement from the American Heart
Association Atherosclerosis, Hypertension, and Obesity in
Youth Committee, Council of Cardiovascular Disease in the
Young, with the Council on Cardiovascular Nursing. Circulation. 2007;115:19481967.
95. Spear BA, Barlow SE, Ervin C, et al. Recommendations for
treatment in children and adolescents with overweight or
obesity. Pediatrics. 2007;120:S254S288.
15
Lipid Disorders
Shirley Huang, MD, and Melanie Katrinak, RD, CSP, LDN
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 162
Types of Lipid Disorders. . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Genetic
Polygenic
Other
162
Learning Objectives
Background
LIPID DISORDERS
Polygenic
Of all types of lipid disorders, polygenic hypercholesterolemia (nonfamilial) is the most common. Lifestyle factors
such as diet, weight, and physical inactivity combined
with a genetic susceptibility are the cause of this form of
dyslipidemia. High triglycerides and low HDL levels are
often seen with obesity and/or a diet with food and drinks
high in simple carbohydrates. In the metabolic syndrome
(Chapter 14), a constellation of findings including high
163
Other
Other causes of lipid disorders include medications and
certain disease states. Medications such as progestins,
anabolic steroids, glucocorticoids, psychotherapeutic
drugs, and retinoic acid acne treatment can cause abnormal
lipid levels. In addition, diseases such as untreated hypothyroidism, renal disease, polycystic ovarian syndrome
(PCOS), or liver disease may also cause dyslipidemias.11
Who to screen?
Any patient > 2 years of age with any of the following CVD risk factors:
A parent, grandparent, aunt, or uncle with cardiovascular disease
< 55 years (male) or < 65 years (female). Cardiovascular disease
includes: myocardial infarction, sudden cardiac death, coronary
bypass surgery, balloon angioplasty, angina pectoris, coronary
atherosclerosis, peripheral vascular disease, or stroke or
A parent with a total cholesterol > 240 mg/dL or
A family history that is not available (adopted child) or
Other cardiovascular risk factors: obesity (BMI > 95th percentile),
sedentary lifestyle, smoking, hypertension, diabetes, congenital
heart disease, renal disease or
Treatment with retinoid acid, anticonvulsants, or oral contraceptives
Table 15-21
How to screen?
After a 9-12 hour fast, obtain a fasting lipid profile that includes:
Total cholesterol (TC)
High-density lipoprotein cholesterol (HDL)
Triglycerides (TG)
Low-density lipoprotein cholesterol (LDL), calculated*
If TG > 400 mg/dL, a Direct LDL needs to be ordered
* Calculated LDL = TC(HDL-TG/5) (if TG < 400 mg/dL)
164
Figure 15-1
*A more conservative HDL cut-point is chosen here. HDL > 40 mg/dL is a cut-point used in pediatric and adult
metabolic syndrome.1,6 The American Heart Assocation recommends HDL > 35 mg/dL in pediatrics.20
Adapted from The Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood
Cholesterol in Adults (Adult Treatment Panel III).1
Nutrition Management
The emphasis on a healthy diet and lifestyle is key in the
prevention of CVD and treatment of abnormal lipid levels.12
To prevent CVD in all children 2 years old, recommendations include a daily diet with total fat < 30% total calories,
saturated fat < 10% of total calories, trans fat < 1% of total
calories, and dietary cholesterol < 300 mg.1 Furthermore,
CVD prevention also includes increasing consumption of
fruits, vegetables, fish, whole grains, and reduced-fat dairy
products while reducing the intake of fruit juice, sugarsweetened beverages and foods, and salt.13 For children 2
years old at risk for CVD, the daily diet should be further
restricted to saturated fat < 7% of total calories and dietary
cholesterol to < 200 mg.1 For children between 12 months
and 2 years of age who are overweight, obese, or have a
family history of obesity, dyslipidemia, or CVD, reduced
fat milk is now considered safe and appropriate.1 Additional
dietary management for high-risk patients should always be
tailored based on individual lipid profile and involve counseling with a lipid specialist and registered dietitian or other
skilled clinician to ensure effective nutrition intervention
while maintaining appropriate growth and development.
2010 A.S.P.E.N. www.nutritioncare.org
Dietary Cholesterol
Dietary cholesterol is found in animal-based foods and
can be reduced by limiting foods such as butter, egg yolk,
high-fat meat, beef, poultry with skin, and whole milk dairy
products. Although limiting dietary cholesterol has less of a
serum lipid lowering effect and has a more variable response
among individuals than limiting saturated and trans fats, it
is still important because cholesterol and saturated fat are
found together in most foods.9,14 In general, LDL may be
decreased by 3% to 5% if dietary cholesterol is restricted
to < 200 mg daily. In addition, an increase of 100 mg/d of
dietary cholesterol increases total serum cholesterol by 2 to
3 mg/dL.
Dietary Fats
Saturated Fats
Limiting saturated fats can help to lower LDL levels.14 Saturated fat is found more in animal- than plant-based foods.
A major source of saturated fat is red meat, but dairy products are also commonly overlooked as a source of saturated
fat in childrens diets. Plant-based sources of saturated fat
LIPID DISORDERS
165
Trans Fats
Trans fats, or hydrogenated fats, are produced when fat is
hydrogenated to make it solid at room temperature. In
the process of hydrogenation, bonds in the cis position are
switched to the trans position, which has been shown to
increase LDL and decrease HDL cholesterol. Trans fats
are found mostly in stick margarine, high-fat baked goods,
shortening, commercial frying oils, and fried snack foods.
Examples of these foods are doughnuts, pastries, crackers,
cookies, potato and tortilla chips, french fries, and other
bakery and snack foods. Trans fats should be limited to
< 1% of total calories for all children 2 years old.1 Nutrition labels are allowed to list zero grams of fat per serving if
the product contains less than 0.5 g of trans fat per serving.
Therefore, to ensure a food is completely free of trans fat, it
is important to check the ingredients for hydrogenated and/
or partially hydrogenated oils.
Simple Carbohydrates
For the general population, current recommendations
encourage choosing mostly complex carbohydrates with
a limited intake of simple carbohydrates.13 Patients with
Fiber
Plant Sterols/Stanols
Plant sterols and stanols are essential components of cell
membranes in plants that are structurally similar to cholesterol. Plant stanols are saturated sterols and have no double
bonds. Their action is to inhibit absorption of dietary
cholesterol and to decrease re-absorption of cholesterol
from bile. Plant sterols or stanol esters have been incorporated into margarine/butter spreads, granola bars, yogurt
drinks, oatmeal, cereal, and some other foods and are also
available in caplets. A randomized control study in children
using 2 g of plant sterol in margarine per day decreased LDL
by 8%.21 Plant sterols are considered safe for children at
recommended doses of 2 g/d to lower LDL levels. However,
they may theoretically have the potential to decrease levels
of fat-soluble vitamins such as vitamin A (beta carotene)
2010 A.S.P.E.N. www.nutritioncare.org
166
benefits of soy remain controversial, soy can provide polyunsaturated fatty acids, fiber, vitamins, and minerals beneficial
for cardiovascular and overall health.17
Soy Protein
The effect of soy on lowering cholesterol remains controversial. While some studies show soy isoflavones can decrease
LDL and triglycerides and increase HDL, others show little
or no effect. Although a daily intake of 25 g of soy in adults
may decrease total and LDL cholesterol from 1.5% to 4.5%,
this may be related more to the use of soy as a substitute for
foods high in saturated fat.22,24,26 No recommendations have
been made for children. While the cholesterol-lowering
Garlic
Antioxidants
Antioxidants have been raised as a possible treatment for
high cholesterol because oxidized LDL is implicated in
plaque development. Although daily supplementation of
vitamins C and E may improve endothelial function, largescale clinical trials have not shown any benefit related to the
primary or secondary prevention of CVD. 24,27 Studies in children are limited, and antioxidant vitamin supplementation
is not currently recommended to manage dyslipidemia.27
Physical Activity
Physical activity is beneficial for children and adolescents
with dyslipidemia, due to its effects on raising HDL and
decreasing triglyceride levels. Improvement of LDL levels
and insulin resistance have also been documented.28,29 In
addition, physical activity plays a critical role in maintaining
an appropriate weight, which also affects cholesterol levels.
Physical activity should be encouraged in all patients with
dyslipidemia unless another medical condition contraindicates this. New physical activity guidelines recommend that
children have at least 60 minutes of moderate to vigorous
physical activity daily, including vigorous physical activity
at least 3 days per week. 30
Nutrition and physical activity recommendations for
specific lipid abnormalities are summarized in Table 15-3.
TABLE 15-3 Summary of Nutrition and Physical Activity Recommendations for Specific Lipid Abnormalities
Lipid Abnormality
Fiber
Simple
Carbohydrates
Dietary
Cholesterol
Trans Fat
Saturated Fat
Omega-3 Fats
Physical
Activity
High LDL
Increase
DGA
< 200 mg
DGA
Increase
High TG
DGA
Decrease
< 200 mg
Increase
Increase
Low HDL
DGA
DGA
< 200 mg
DGA
Increase
LIPID DISORDERS
Pharmacologic Intervention
Medications may be considered in conjunction with nutrition and physical activity interventions for patients 8 years
and older with an LDL 190 mg/dL (or 160 mg/dL with
a family history of early heart disease or 2 additional risk
factors present, or 130 mg/dL or > 100 mg/dL depending
if Type 1 diabetes mellitus or other high-risk conditions
exist).1,31 HMG CoA-reductase inhibitors, or statins, are the
recommended class of medications to lower LDL levels in
pediatrics. Statins have been shown to be safe and effective
in lowering cholesterol in a number of clinical trials, though
they have generally been short-term. 3236 Patients, however,
need to be monitored closely for liver and muscle side
effects. Niacin and bile acid-binding resins are other classes
of cholesterol-lowering medications but are not routinely
recommended in pediatrics due to limited effectiveness and
poor compliance. Cholesterol-absorption inhibitors are the
newest class that are often combined with other medications
such as statins, but have not yet been extensively studied in
children.
167
References
168
28. Maron BJ, Chaitman BR, Ackerman MJ, et al. Recommendations for physical activity and recreational sports participation
for young patients with genetic cardiovascular diseases. Circulation. 2004;109(22):28072816.
29. Strong WB, Malina RM, Blimkie CJ, et al. Evidencebased physical activity for school-age youth. J Pediatr.
2005;146(6):732737.
30. US Department of Health and Human Services. 2008 physical
activity guidelines for Americans. http://www.health.gov/
paguidelines. Accessed November 12, 2009.
31. Kavey R, Aladda V, et al. Cardiovascular Risk Reduction
in High-Risk Patients: A Scientific Statement From the
American Heart Association Expert Panel on Population
and Prevention Science; the Councils on Cardiovascular
Disease in the Young, Epidemiology and Prevention, Nutrition, Physical Activity and Metabolism, High Blood Pressure
Research, Cardiovascular Nursing, and the Kidney in Heart
Disease; and the Interdisciplinary Working Group on Quality
of Care and Outcomes Research: Endorsed by the American
Academy of Pediatrics. Circulation. 2006;114:27102738.
32. de Jongh S, Ose L, Szamosi T, et al. Efficacy and safety of
statin therapy in children with familial hypercholesterolemia:
a randomized double-blind, placebo-controlled trial with
simvastatin. Circulation. 2002;106(17):22312237.
33. Lambert M, Lupien PJ, Gagne C, et al. Treatment of familial
hypercholesterolemia in children and adults: effect of
lovastatin. Canadian Lovastatin in Children Study Group.
Pediatrics. 1996;97(5):619628.
34. McCrindle BW, Ose L, Marais AD. Efficacy and safety
of atorvastatin in children and adolescents with familial
hypercholesterolemia or severe hyperlipidemia: a multicenter, randomized, placebo-controlled trial. J Pediatr.
2003;143(1):7480.
35. de Jongh S, Lilien MR, opt Roodt J, et al. Early statin therapy
restores endothelial function in children with familial hyper
cholesterolemia. J Am Coll Cardiol. 2002;40(12):21172121.
36. Wiegman A, Hutten BA, de Groot E, et al. Efficacy and safety of
statin therapy in children with familial hypercholesterolemia:
a randomized controlled trial. JAMA. 2004;292(3):331337.
16
Catherine Christie, PhD, RD, Julia A. Watkins, PhD, MPH, and Judith C. Rodriguez, PhD, RD
Learning Objectives
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Clinical Approach to the Overweight/Obese Child or
Adolescent . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Use of Fad and Popular Diets . . . . . . . . . . . . . . . . . . . . . .
Lifelong Weight Management. . . . . . . . . . . . . . . . . . . . . .
Types of Fad and Popular Diets. . . . . . . . . . . . . . . . . . . . .
169
170
170
171
171
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
Background
170
25 years
BMI of 85th to
94th percentile
BMI of 95th
percentile
611 years
BMI of 85th to
94th percentile
BMI of 95th to
98th percentile
BMI of 99th
percentile
Target
1218 years
BMI of 85th to
94th percentile
171
172
173
174
Other Plans
Eat Right for Your Type28 was on the New York Times bestseller list and remains a popular diet plan today. The book
provides 4 diets based on the Blood Types O, A, B, and AB.
People with Type O blood are described as hunters, who
need a diet with high protein, lean, chemical-free meat,
poultry, and fish with limited grains, beans, and legumes.
Those with type A blood, the cultivators, need to eat predominantly vegetarian with fish and an emphasis on vegetables,
tofu, grains, beans, legumes, and fruit. Type B, the nomads,
should eat meat (no chicken), dairy, grains, beans, legumes,
vegetables, and fruit. Type AB, the enigma, can eat a mixed
diet in moderation including meat, seafood, dairy, tofu,
beans, legumes, grains, vegetables, and fruit. Each blood
type has a long list of forbidden foods including specific
meats and poultry, seafood, dairy, eggs, oils and fats, nuts
and seeds, beans, legumes, cereals, breads, muffins, grains,
pasta, vegetables, fruit, juices, fluids, spices, condiments,
herbal teas, and other beverages.
Each of the blood-type diets consists of whole, unprocessed foods and all recommend the consumption of
vegetables, which provide fiber, health-promoting nutrients,
and phytochemicals. Physical activity, stress management,
2010 A.S.P.E.N. www.nutritioncare.org
175
kids together about healthy eating for weight loss, separate meetings for adults and children with a mental health
professional to learn to change habits and behaviors and
receive group support, and meetings for adults to discuss
their specific questions while kids participate in 30 minutes
of aerobic exercise; during 2 weeks adults and kids exercise together. 35 Program effectiveness was evaluated in
a study of 1,022 families from 24 community-based and
hospital-based sites in Pennsylvania and California from
2004 through 2006. Food records, activity logs, program
questionnaires, and participant BMIs were obtained at the
beginning and the end of the program. Statistically significant improvements in BMI, eating, physical activity, and
self-esteem were reported. 36 A follow-up study looked at a
convenience sample of 86 children at 3 months, 88 children
at 6 months, 30 children at 12 months, and 15 children at 18
to 24 months after completion of the KidShape program.
Graduates of the program maintained a significant change
in BMI up to 24 months after the program and reported
continued improvement in eating and physical activity. 37
Conclusion
There are many diets that will result in weight loss or weight
maintenance in adults and children because they control
and lower caloric intake. The scientific evidence is strong
that a successful weight-loss or weight-maintenance plan
for children and adolescents should include reduced kilocalorie intake though consumption of 5 servings of fruits
and vegetables per day and minimization or elimination
of sugar-sweetened beverages (which are both present in
a balanced macronutrient reduced kilocalorie diet and the
Traffic Light Diet) as well as the behavioral components
(eg, eating breakfast daily, limiting meals outside the home
including at fast-food venues and other restaurants, eating
family meals at least 5 or 6 times per week, allowing the child
to self-regulate his or her meals and avoiding overly restrictive behaviors). Long-term weight management happens
when a plan is individualized to a familys needs and preferences. Kilocalorie- and portion-controlled diets such as the
balanced macronutrient low-calorie diets and the Traffic
Light Diet have been studied over time and may be more
conducive to long-term compliance in families than fat- and
carbohydrate-restricted diets.
Reviewing the various categories of popular and fad diets
indicates that all have strengths and weaknesses and have the
potential to result in weight loss in adults. However, for children and adolescents, these diets have not been studied due
to potential risks for growing children and adolescents from
elimination of key food groups, greater-than-recommended
2010 A.S.P.E.N. www.nutritioncare.org
176
References
18. Sharman MJ, Gomez AL, Kraemer WJ, Volek JS. Very
low-carbohydrate and low-fat diets affect fasting lipids and
postprandial lipemia differently in overweight men. J. Nutr.
2004;134:880885.
19. Brehm BJ, Seeley RJ, Daniels SR, et al. A randomized trial comparing a very low carbohydrate diet and a
kilocalorie-restricted low fat diet on body weight and cardiovascular risk factors in healthy women. J Clin Endocrinol
Metab. 2003;88(4):16171623.
20. Shai I, Schwarzfuchs D, Henkin Y, et al. Weight loss with a
low-carbohydrate, Mediterranean, or low-fat diet. N Engl J
Med. 2008;359:229241.
21. Pritikin R. The New Pritikin Program. New York, NY: Simon
& Schuster; 2000.
22. Ornish D. Eat More Weight Less. New York, NY: Harper
Collins; 1993.
23. Rolls B, Barnett RA. The Volumetrics Eating Plan. New York,
NY: Harper Collins; 2005.
24. Somers S. Eat Great, Lose Weight. New York, NY: Random
House; 1999.
25. WeightWatchers. www.weightwatchers.com. Accessed
December 10, 2008.
26. Barlow S, Dietz W. Obesity evaluation and treatment: expert
committee recommendations. Pediatrics. 1998;102(3).
27. Daniels SR, Arnett DK, Eckel RH, et al. Overweight in
children and adolescents: pathophysiology, consequences,
prevention and treatment. Circulation. 2005;111:19992012.
28. DAdamo P, Whitney C. Eat Right for Your Type. New York,
NY: GP Putnam & Sons; 1996.
29. American Dietetic Association. Pediatric Weight Management Evidence Based Nutrition Practice Guidelines. ADA
Evidence Library. www.adaevidenceanalysislibrary.com.
Accessed December 10, 2008.
177
17
Sports Nutrition
Jackie Buell, PhD, RD, LD, ATC, LAT and Diane L. Habash, PhD, RD, LD
Learning Objectives
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
Carbohydrates
Protein
Fat. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Hydration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Vitamins and Minerals. . . . . . . . . . . . . . . . . . . . . . . . . . . .
Timing to Sport. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Recovery Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Bedtime Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Nutrition Challenges . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Nutrition Resources. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
181
181
182
182
183
183
183
183
Introduction
SPORTS NUTRITION
179
4500 calories daily. Estimating energy needs and monitoring for desirable changes is necessary to confirm the
accuracy of the energy prescription.
Carbohydrates
The most obvious dietary need within the calories allotted
for the young athlete should be carbohydrate to fuel the
muscle. It has long been accepted that carbohydrate is
the primary fuel for human muscle as intensity of activity
increases, and this is well confirmed in adult athletes. It is
suggested that children may be able to use even more carbohydrate as substrate than adults5; thus, fueling the muscle
to at least the adult mass-dependent recommendation is
prudent. In adults, carbohydrate has been demonstrated
to be supportive before, during, and after endurance exercise when enough calories are consumed overall. 6 Ensuring
5 g of carbohydrate per kilogram body weight is the basic
starting point for all athletes.7,8 The need for carbohydrate
increases as the time and intensity of activity increases. The
more serious athlete who practices daily for 1 to 2 hours
might increase this fueling to 7 g/kg up through 11 to 12
g/kg for 3 to 4 hours of intense exercise per day.7,8 It is not
as simple as asking the frequency and length of practices;
to apply these guidelines it is important to gauge the actual
time spent in high-intensity activity. Most sport practice
sessions include periods of inactivity and rest. Including the
downtime of sessions would overestimate fueling needs.
Consuming too much simple carbohydrate may increase
dental cariesespecially when the choices include sticky
and fermentable sourcesand there is a high degree of
variability in how humans tolerate high carbohydrate. It is
prudent to consider the athletes preferences, tolerance, and
performance enhancement when fine-tuning the carbohydrate in the diet.
Awareness of the underlying health benefits of the
source of carbohydrate should also be emphasized in this
cohort as a strategy to encourage lifelong healthy habits.
Carbohydrate in the diet comes mostly from the grains,
fruit, and milk food groups with smaller contributions
from the vegetable group. Overall encouragement to use
whole grains over refined products will improve the fiber
content and nutrient density of the diet and may confer
long-term health benefits. The value of fruit in the diet is
uncontested, and the child or adolescent athlete needs 4
servings of milk/dairy daily for skeletal health. Even though
vegetables provide a minimal contribution, promoting
them for carbohydrate advantages may encourage better
consumption in this young population. Another potential
advantage of encouraging healthier carbohydrate sources
2010 A.S.P.E.N. www.nutritioncare.org
180
Carbohydrates Grains
(4 kcal/g)
Fruits
Milk
Vegetables
Protein
Meat
(4 kcal/g)
Milk
Beans
Peanut Butter
Fats
(9 kcal/g)
saturated
polyunsaturated
Protein
Protein is not typically viewed as energy for sport, but is
critical for growth and development. The amount of protein
needed in the athletes diet remains controversial in all
populations.10,11 The DRI for protein, without consideration
of athletic activity, ranges from the adult recommended
dietary allowance (RDA) of 0.8 g/kg/d to the child adolescent recommendation of 1.05 g/kg/d,4 and this current
protein DRI excludes additional protein possibly available
for muscle fueling or metabolism. It is well-documented
that adult athletes are able to utilize more than these DRIrecommended amounts to support the muscle protein
turnover and energy demands of varying activities.12 In
general, nutrition recommendations for adult athletes
encompass a range of about 1 to 1.7 g/kg/d.13 Bulk-seeking
strength athletes top the recommendation chart at 1.7 g/
kg/d as the extrapolated upper threshold that nitrogen
balance studies have demonstrated can physiologically be
used for lean mass accretion.14 It is intuitive that the upper
threshold for the child or adolescent athlete may be slightly
higher than these adult values due to compounded growth and
2010 A.S.P.E.N. www.nutritioncare.org
monounsaturated
Portion
Grams
Per
Portion
15
15
12
5
7
8
7
7
5
5
SPORTS NUTRITION
Fat
The ideal amount and type of fat in the diet remains as controversial for athletes as it is in general. The current acceptable
macronutrient distribution range from the DRIs calls for 20%
to 35% of the calories be taken as fat. Prudent heart health
recommendations encourage incorporation of mono- and
polyunsaturated fats to displace many of the typical saturated fats in the Western diet.21 When athletes are struggling
to maintain body mass during the competitive season, it is
imperative to encourage calorie-dense foods which typically
include a higher fat proportion. The hormonal milieu in the
body is undoubtedly supported by an adequate fat intake and
calorie balance. Recent literature has addressed fat loading
as a method of supporting fueling for sub-maximal endurance
exercise, but the studies thus far demonstrate a loss of high
gear or reduced ability to reach or sustain high intensity,22 and
there is no research on how these diets may influence body
composition or cardiovascular health (in child athletes).
181
Hydration
182
Timing to Sport
SPORTS NUTRITION
Recovery Nutrition
Recovery nutrition has become one of the hottest recommendation topics in recent years of sports nutrition.17,35
Consuming an appropriate recovery snack immediately
after exercise should become habit for athletes who do not
have at least 24 hours to recover the muscle nutritionally.
A recovery snack should be a priority for multiple-event
athletes or when the daily routine demands multiple practices close together. An appropriate recovery snack for
adult athletes suggests 40 to 60 g of carbohydrate and 15 to
20g of good-quality protein to provide the essential amino
acids. The carbohydrate for recovery has demonstrated a
more rapid period of glycogen restoration, and the additional calories in protein stimulate the muscle to set up an
anabolic environment.15,36 The current research supports
essential amino acids as the stimulus for this anabolic
change and leucine (a branched-chain essential amino acid)
is likely important to the response. 37
183
Nutrition Challenges
Nutrition Resources
Bedtime Nutrition
184
Table 17-3 Examples of Meals and Snacks Used With Timing Around Sport
Description
Pre-game
Target is high carbohydrate within mixed diet
(34 hr prior)
2 cups whole wheat pasta with 1 cup sauce and 3 oz lean meat, 1
Pasta meal
cup of salad, 1 piece garlic bread, and 1 cup 2% milk
Fast food
Wendys grilled chicken with baked potato with broccoli cheesesauce
Larger athletes might add a small Frosty to above to bring total to
Pre-event
Target is well-tolerated carbohydrates
(2060 min prior)
12 oz sweet tea with 1 oz animal crackers
20 oz sports drink
2 oz pretzels with water
During event
Sports drink only warranted if longer than 90 minutes of intense
(48 oz every
activity (otherwise water is great)
1520 min)
Per 8 oz
Gatorade G
Gatorade G2 series
Powerade
Powerade Zero
Accelerade (incl 4 g protein)
Recovery
(within 45 min)
kcal
g CHO
g pro
g fat
797
120
54
18
780
1080
117
143
44
59
16
31
kcal
g CHO
g pro
g fat
231
160
200
48
40
47
2
0
6
4
0
0
kcal
g CHO
Na (mg)
K (mg)
50
25
60
0
80
kcal
Target
200300
14
7
17
0
15
g CHO
Target
4060
110
110
55
55
120
g pro
Target
1015
30
30
30
33
15
g fat
Target low
11
1.5
20
10
18
11
2.5
10
10
Nutrition Goals
Whole grain or fruit pancakes with syrup, 8 oz milk, side of fresh fruit,
2oz lean meat, 16 oz water after meal will begin hydration on right path
12 oz sports drink, piece of fruit or pretzels; if urine is strong, morewater
48 oz of sports drink every 1520 minutes
SPORTS NUTRITION
185
Solutions
Limited nutrition
knowledge
related to sports
performance
Picky eater
Male athlete
with high growth
expectations
Stress that normal physical activity and adequate nutrition optimize growth
Educate that growth has genetic and environmental components
Consider that intense exercise may be associated with delayed growth
Check and reinforce appropriate athlete vs parent expectations of lean
massaccretion; use growth charts
Weight control
expectations
(typically females)
Vegetarianism
Encourage eating a diet with a wide variety of foods that will fuel the muscle
foroptimal performance
When in doubt, suggest evaluation by psychological professional and dietitian
Intervene early to increase chances for recovery
Use of
May be related to athletes desire to excel but
oftennot supported by evidence
performanceenhancing
Often used by athlete as a substitute for
supplements (legal adequatenutrition
and illegal)
Could be related to peer pressure and media
influences
There are no pediatric evidence-based clinical trials on the use or the adverse
effects of some supplements (herbals, ergogenic aids, etc.)
Recent review of supplement studies shows anabolic steroid use ranges from 1%
to 11.1% of adolescents surveyed and creatine use in reviewed studies ranged
from5.6% to 30%40
Supplements sought frequently for improving speed, decreasing weight, or
increasing size/bulk of athlete
Use of one multivitamin/mineral supplement daily has been suggested for all
individuals, not just athletes
Seek advice of certified sports dietitian (CSSD)
2010 A.S.P.E.N. www.nutritioncare.org
186
Resources
Supplement Information
SPORTS NUTRITION
References
187
188
40. Castillo EM, Comstock RD. Prevalence of use of performanceenhancing substances among United States adolescents.
Pediatr Clin N Am. 2007;54:663675.
41. American Dietetic Association; Dietitians of Canada; American College of Sports Medicine, Rodriguez NR, Di Marco
NM, Langley S. American College of Sports Medicine position stand. Nutrition and athletic performance. Med Sci Sports
Exerc. 2009;41(3):709731.
PART III
18
Developmental Delay
Kathleen J. Motil, MD, PhD
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Nutrition Problems. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Undernutrition, Growth Failure, and Overweight
Micronutrient Deficiencies
Osteopenia
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
Learning Objectives
Introduction
192
Nutrition Problems
Undernutrition, Growth Failure, and Overweight
Prevalence
The true prevalence of undernutrition, growth failure,
and overweight in children with developmental delay is
unknown. Estimates are limited to disorders such as cerebral
palsy, myelodysplasia, spina bifida, spinal cord injury, and
Rett syndrome.1120 Undernutrition, based on weight-forheight and triceps skinfold thickness, has been documented
in 29% to 46%, linear stunting in 23%, and overweight in
8% to 40% of individuals with these disorders. The prevalence of undernutrition increases with increasing age,
lower intelligence, and increased severity of neurological
impairment.17
Pathophysiology
Non-nutrition factors including the type and severity of
neurological disability, ambulatory status, and cognitive ability contribute to growth failure in children with
developmental delay.21 Children with seizures or spastic
quadriplegia and those who are non-ambulatory have lower
height z scores than children who lack these disabilities.18
Children with spastic hemiplegia have smaller measures
of breadth and length on the affected side, suggesting that
neurological impairment influences growth.21 Inherent
genetic factors may be associated with permanent linear
stunting.22 Height-for-age z scores may decrease with
advancing age independently of weight-for-age z scores in
individuals affected with scoliosis or contractures.18
Nutrition factors contribute to growth failure in
children with developmental delay based on correlations
between height and weight z score deficits.23 Nutrition
status explains 10% to 15% of the variability of linear
growth in children with cerebral palsy. 23 Nutrition status
has a stronger effect on linear growth in younger than in
older children, attesting to the irreversible effects of longterm undernutrition on growth. Inappropriate dietary
intake relative to nutrient needs, oral motor dysfunction,
increased nutrient losses, and altered energy expenditure
may account for the poor nutrition status of children with
developmental delay.
DEVELOPMENTAL DELAY
oral sensation, uncoordinated or involuntary tongue movements, or delayed development of age-appropriate oral motor
skills. Initiation of the swallowing reflex may be delayed,
resulting in food accumulation in the vallecula or pyriform
sinuses and subsequent aspiration. Neurologically impaired
children with these findings have lower height-, weight-, and
weight-for-height z scores, body fat, and arm muscle area
than unaffected children.16,34,35 Children with more severe
impairment who are unable to lift their heads or feed themselves have a higher risk of aspiration. 36 Early, persistent,
and severe feeding difficulties are markers of subsequent
poor health, nutrition status, and growth and identify children who may benefit from gastrostomy feedings. 35,37
Increased nutrient losses
193
Micronutrient Deficiencies
Vitamin, trace element, and essential fatty acid (EFA)
deficiencies have been documented in children with developmental delay who have reduced dietary intakes.4951
Iron, selenium, zinc, EFAs, and vitamins C, D, and E were
reported to be deficient in 15% to 50% of the children.4952
Some may develop nutrient deficiencies because enteral
formulas provide adequate amounts of micronutrients only
when volumes that meet their age-related DRI for energy are
consumed. 53 Because many children with developmental
delay require lower energy intakes, their micronutrient
2010 A.S.P.E.N. www.nutritioncare.org
194
Osteopenia
Osteopenia is prevalent in developmentally impaired children. 52,5759 Weight z score is the best correlate of bone
mineral density z score in children with developmental
delay. 58 Dietary calcium, vitamin D, and phosphorus intakes
are lower than the DRI in 50% to 80% of these children. 24,60
Non-ambulatory children have lower bone mineral content
than those who ambulate independently. 58 Limited ambulation, increased duration of anticonvulsant therapy, and
reduced sun exposure contribute to the pathogenesis of
osteopenia. 5760 Osteopenia is associated with an increased
fracture risk in developmentally impaired children. 5759
Supplemental calcium improves bone mineral density by
5% over 4 years in healthy children, but the effect of dietary
calcium in children with developmental delay is unknown.61
The use of bisphosphonates increased bone mineral density
by 89% over 18 months in children with cerebral palsy. 62
However, the relation between bisphosphonate use and
fracture risk or frequency in these children is unknown. The
use of bisphosphonates is limited because their indications
in childhood diseases are not well defined and their longterm effects on bone remodeling in children are unknown.
Nutrition Assessment
Medical History
The medical history includes information about the
etiology, duration, and severity of neurological impairment
and its expected course. These factors correlate with the
risk of undernutrition and may affect the type of nutritional
intervention required. Although the neurological condition
DEVELOPMENTAL DELAY
195
Physical Examination
Physical examination focuses on signs of undernutrition,
linear stunting, overweight, and specific nutrient deficiencies. Muscle tone, activity level, and the presence of athetoid
movement are relevant because they influence dietary
energy needs. Contractures and scoliosis are noteworthy
for positioning during meals. Abnormal breath sounds may
be suggestive of chronic respiratory problems associated
with aspiration. Abdominal distention in conjunction with
palpable masses suggests constipation. Examination of the
skin may reveal the presence of decubitus ulcers. Pallor,
skin rashes, smooth tongue, gingival bleeding, petechiae,
bone deformities, or pedal edema may suggest other micronutrient deficiencies.
2010 A.S.P.E.N. www.nutritioncare.org
196
Meal Observation
A 24-hour recall of habitual food intake or a 3-day record
of food consumption reflects the adequacy of dietary
energy and nutrient intake. 34 Meal observation, with
emphasis on the childs ability to feed independently and
the efficiency of the feeding process, may reveal a reason
for poor weight gain.12,16,29,30 An evaluation of oral motor
skills, including inadequate lip closure, drooling, a persistent extrusion reflex, gagging and delayed swallowing, or
coughing and choking during meals reflects poor feeding
capabilities. Limited texture tolerance may indicate poor
oral ability to manage food, resulting in self-restricted
eating patterns, reduced nutrient intake, and poor weight
gain.68,69 Consumption of inappropriate food textures may
result in aspiration. Fatigue and lethargy during meals may
suggest hypoxemia.70 Meal observation shows that children
with developmental delay may be offered less, consume less,
and spill more food than unaffected children. Classification systems based on measures of growth and patterns of
food consumption, such as eating efficiency and oral motor
feeding skills, may be helpful to assess the effectiveness of
oral feeding interventions.71,72
Diagnostic Studies
Although isolated nutrient deficiencies may be present in
children with developmental delay, extensive laboratory
evaluation is not necessary.4951 A complete blood count and
serum ferritin may document anemia or iron deficiency.
Serum electrolytes and blood urea nitrogen (BUN) reflect
hydration status; however, BUN may be low because of
poor protein intake and low muscle mass. Serum albumin
and prealbumin, factors that correlate strongly with the
risk of morbidity and mortality, are less reliable indicators
of nutrition status.73 Abnormal serum phosphorus, alkaline
phosphatase, and 25-hydroxyvitamin D levels may coincide
with poor bone mineral status. Bone densitometry may be
considered in children who have pathologic fractures. Bone
quantitative ultrasonography may be more easily performed
than bone densitometry; however, normative data for children are not yet available.74
Additional diagnostic studies may be helpful, depending
on the childs symptoms and the need for permanent enteral
access. A videofluoroscopic assessment of chewing and
swallowing function, using different food and beverage
textures, determines the degree of oral motor dysfunction
and risk of aspiration. Positioning the child during the
evaluation is important because some children may not
aspirate when placed upright, but do so in a reclined position. A swallowing function study may demonstrate silent
2010 A.S.P.E.N. www.nutritioncare.org
Nutrition Support
Nutrition support is provided enterally rather than parenterally, assuming competency of the gastrointestinal tract.
Enteral tube feedings are essential in children who cannot
meet their energy and nutrient needs orally.75 Evidence of
oral motor feeding difficulties, undernutrition (weight-forheight < 80% of expected, BMI < 5th percentile), growth
failure (height-for-age < 90% of expected), overweight
(BMI > 85th percentile), and individual nutrient deficiencies indicate the need for nutritional intervention.1
Nutrition Requirements
Energy requirements of children with developmental delay
vary with the severity of their neurological disability, their
mobility, the presence of feeding difficulties, altered body
composition, and the need for weight gain or loss and catchup growth. Dietary energy needs for the maintenance of
body weight may be estimated from either the DRI standards for basal energy expenditure (http://www.nal.usda.
gov/fnic/etext/000105.html), indirect calorimetry,76 or
height77 (Table 18-1). Dietary energy needs for catch-up
DEVELOPMENTAL DELAY
growth may approximate the DRI for basal energy expenditure 1.5. Monitoring the rate of weight gain or loss and
change in BMI is the best way to determine the adequacy of
diet in the child with developmental delay. Adequate provision of dietary protein, vitamins, and minerals is mandatory
when energy intakes are modified to obtain the desired rate
of weight change. In the absence of evidence-based nutrient
allowances for children with developmental delay, the
DRIs for protein, vitamins, and minerals in healthy children are recommended (http://www.nal.usda.gov/fnic/
etext/000105.html). Multivitamin and mineral supplements may be prudent for children with developmental
delay who rely primarily on table foods and beverages to
meet their nutrient needs, particularly in relation to the
need for improved vitamin D status.78
Table 18-1. Methods to Determine Dietary Energy Needs in Neurologically
Impaired Children
Dietary Reference Intake Standards for Basal Energy Expenditure
(http://www.nal.usda.gov/fnic/etext/000105.html)
Energy intake (kcal/d) = Basal Energy Expenditure (BEE) x 1.1
Age (y)
Basal Energy Expenditure (kcal/d)
Boys
Girls
38
1035
1004
913
1320
1186
1418
1729
1361
Indirect Calorimetry76
Energy intake (kcal/d)
= [basal energy expenditure (BMR) x muscle tone x activity] + growth
where:
BMR (kcal/d) = body surface area (m2) x metabolic rate (kcal/m2/h)
x 24 h
Muscle tone = 0.9 if decreased, 1 if normal, and 1.1 if increased
Activity = 1.1 if bedridden, 1.2 if wheelchair dependent or crawling,
and 1.3 if ambulatory
Growth = 5 kcal/g of desired weight gain (normal and catch-up
growth)
Height77
15 kcal/cm in children without motor dysfunction
14 kcal/cm in children with motor dysfunction who are ambulatory
11 kcal/cm in children who are non-ambulatory
197
Behavioral Modification
Behavioral therapy initiated by a skilled child psychologist
may improve the quantity of food consumed, the feeding
efficiency, and the range of textures accepted, as well as the
quality of feeding interactions between the caretaker and
the child.81
198
DEVELOPMENTAL DELAY
199
Feeding Intolerance
Acknowledgments
Ethical Considerations
Many families find the idea of a feeding gastrostomy difficult to accept.107 Caregivers believe that they have failed
to adequately care for the child when physicians insist on
gastrostomy placement. Starvation, quality of life, prolongation of life, and meaningful family interrelationships
constitute a framework for discussion. Although medical
opinions generally prevail, parental wishes should be
considered and respected.
Conclusion
References
200
3. Marchand V, Motil KJ, NASPGHAN Committee on Nutrition. Nutrition support for neurologically impaired children:
a clinical report of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition. J Pediatr
Gastroenterol Nutr. 2006;43(1):123135.
4. Samson-Fang L, Fung E, Stallings VA, et al. Relationship of
nutritional status to health and societal participation in children with cerebral palsy. J Pediatr. 2002;141(5):637643.
5. Rogers B. Feeding method and health outcomes of children
with cerebral palsy. J Pediatr. 2004;145(2 Suppl):S28S32.
6. Sleigh G, Brocklehurst P. Gastrostomy feeding in cerebral palsy: a systematic review. Arch Dis Child. 2004;89(6):534539.
7. Samson-Fang L, Butler C, ODonnell M. Effects of gastrostomy feeding in children with cerebral palsy: an AACPDM
evidence report. Dev Med Child Neurol. 2003;45(6):415426.
8. Sullivan PB, Juszczak E, Bachlet AM, et al. Gastrostomy tube
feeding in children with cerebral palsy: a prospective, longitudinal study. Dev Med Child Neurol. 2005; 47(2):7785.
9. Craig GM, Carr LJ, Cass H, et al. Medical, surgical, and health
outcomes of gastrostomy feeding. Dev Med Child Neurol.
2006;48(5):353360.
10. Strauss D, Kastner T, Ashwal S, White J. Tubefeeding and
mortality in children with severe disabilities and mental retardation. Pediatrics. 1997;99(3):358362.
11. Stallings VA, Charney EB, Davies JC, Cronk CE. Nutritional
status and growth of children with diplegic or hemiplegic
cerebral palsy. Dev Med Child Neurol. 1993;35(11):9971006.
12. Dahl M, Thommessen M, Rasmussen M, Selberg T. Feeding
and nutritional characteristics in children with moderate or
severe cerebral palsy. Acta Paediatr. 1996;85(6):697701.
13. Mita K, Akataki K, Ono Y, Ishida N, Oki T. Assessment of
obesity of children with spina bifida. Dev Med Child Neurol.
1993;35:305311.
14. Bandini LG, Schoeller DA, Fukagawa NK, Wykes LJ, Dietz
WH. Body composition and energy expenditure in adolescents with cerebral palsy or myelodysplasia. Pediatr Res.
1991;29(1):7071.
15. Schultz R, Glaze DG, Motil KJ, et al. The pattern of growth failure in Rett syndrome. Am J Dis Child. 1993;147(6):633637.
16. Troughton KE, Hill AE. Relation between objectively
measured feeding competence and nutrition in children with
cerebral palsy. Dev Med Child Neurol. 2001;43(3):187190.
17. Sanchez-Lastres J, Eiris-Punal J, Otero-Cepeda JL, PavonBelinchon P, Castro-Gago M. Nutritional status of mentally
retarded children in north-west Spain. I. Anthropometric
indicators. Acta Paediatr. 2003;92(6):747753.
18. Stevenson RD, Hayes RP, Cater LV, Blackman JA. Clinical
correlates of linear growth in children with cerebral palsy. Dev
Med Child Neurol. 1994;36(2):135142.
19. Nelson MD, Widman LM, Abresch RT, et al. Metabolic
syndrome in adolescents with spinal cord dysfunction. J Spinal
Cord Med. 2007;30(Suppl 1):S127S139.
20. Fiore P, Picco P, Castagnola E, et al. Nutritional survey of
children and adolescents with myelomeningocele (MMC):
overweight associated with reduced energy intake. Eur J
Pediatr Surg. 1998;8(Suppl 1):3436.
21. Stevenson RD, Roberts CD, Vogtle L. The effects of nonnutritional factors on growth in cerebral palsy. Dev Med Child
Neurol. 1995;37(2):124130.
2010 A.S.P.E.N. www.nutritioncare.org
22. Motil KJ, Morrissey M, Caeg E, Barrish JO, Glaze DG. Gastrostomy placement improves height and weight in girls with Rett
syndrome. J Pediatr Gastroenterol Nutr. 2009;49:237242.
23. Stallings VA, Charney EB, Davies JC, Cronk CE. Nutritionrelated growth failure of children with quadriplegic cerebral
palsy. Dev Med Child Neurol. 1993;35(2):126138.
24. Fried MD, Pencharz PB. Energy and nutrient intakes of children with spastic quadriplegia. J Pediatr.
1991;119(6):947949.
25. Reilly S, Skuse D. Characteristics and management of
feeding problems of young children with cerebral palsy. Dev
Med Child Neurol. 1992;34(5):379388.
26. Stallings VA, Zemel BS, Davies JC, Cronk CE, Charney
EB. Energy expenditure of children and adolescents with
severe disabilities: a cerebral palsy model. Am J Clin Nutr.
1996;64(4):627634.
27. Hillesund E, Skranes J, Trygg KU, Bohmer T. Micronutrient status in children with cerebral palsy. Acta Paediatr.
2007;96(8):11951198.
28. Mathisen BA, Shepherd K. Oral-motor dysfunction and
feeding problems in infants with myelodysplasia. Pediatr
Rehabil. 1997;1:117122.
29. Reilly S, Skuse D, Poblete X. Prevalence of feeding problems
and oral motor dysfunction in children with cerebral palsy: a
community survey. J Pediatr. 1996;129(6):877882.
30. Sullivan PB, Alder N, Allison ME, et al. Gastrostomy feeding
in cerebral palsy: too much of a good thing? Dev Med Child
Neurol. 2006;48(11):877882.
31. Gisel EG, Patrick J. Identification of children with cerebral
palsy unable to maintain a normal nutritional state. Lancet.
1988;1(8580):283286.
32. Sullivan PB, Lambert B, Rose M, Ford-Adams M, Johnson A,
Griffiths P. Prevalence and severity of feeding and nutritional
problems in children with neurological impairment: Oxford
Feeding Study. Dev Med Child Neurol. 2000;42(10):674680.
33. Thommessen M, Heiberg A, Kase BF, Llarson S, Riis G.
Feeding problems, height and weight in different groups of
disabled children. Acta Paediatr Scand. 1991;80(5):527533.
34. Sullivan PB, Juszczak E, Lambert BR, et al. Impact of feeding
problems on nutritional intake and growth: Oxford Feeding
Study II. Dev Med Child Neurol. 2002;44:461467.
35. Fung EB, Samson-Fang L, Stallings VA, Rose M, Ford-Adams
ME, Johnson A. Feeding dysfunction is associated with poor
growth and health status in children with cerebral palsy. J Am
Diet Assoc. 2002;102(3):361368.
36. Strauss D, Ashwal S, Shavelle R, Eyman RK. Prognosis for
survival and improvement in function in children with severe
developmental disabilities. J Pediatr. 1997;131(5):712717.
37. Motion S, Northstone K, Emond A, Stucke S, Golding J. Early
feeding problems in children with cerebral palsy: weight
and neurodevelopmental outcomes. Dev Med Child Neurol.
2002;44(1):4043.
38. Van Roon D, Steenbergen B. The use of ergonomic
spoons by people with cerebral palsy: effects of food
spilling and movement kinematics. Dev Med Child Neurol.
2006;48(11):888891.
39. Azcue MP, Zello GA, Levy LD, Pencharz PB. Energy
expenditure and body composition in children with spastic
quadriplegic cerebral palsy. J Pediatr. 1996;129(6):870876.
DEVELOPMENTAL DELAY
201
202
DEVELOPMENTAL DELAY
203
19
Eating Disorders
Christina Fitzgerald, MS, RD, LDN and Betsy Hjelmgren, MS, RD, LDN, CSP
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
Classification and Diagnosis of Eating Disorders. . . . . . 205
Physical Presentation
Etiology of Eating Disorders
204
Learning Objectives
Introduction
EATING DISORDERS
205
Physical Presentation
Physical presentation of a person with anorexia nervosa
includes lanugo-type hair, muscle wasting, dry skin,
cyanosis of extremities, bradycardia less than 60 beats/min,
and cachexia. When anorexia develops in childhood, the
first clinical sign may be failure to make weight gains while
continuing to grow in height as opposed to documented
weight loss. Growth charts should be evaluated for typical
growth patterns of the individual.9
Physical signs and symptoms of a person with bulimia
nervosa are more difficult to detect but may include parotid
gland enlargement, scarring of the hand used to stimulate
gag reflux (referred to as Russells sign), erosion of dental
enamel with increased dental caries, and sore red throat
Figure 19-2 American Psychiatric Association Diagnostic Criteria for
307.51 Bulimia Nervosa
206
Nutrition Requirements
Macronutrients
Energy Requirements
EATING DISORDERS
Protein Requirements
Recommended fat intake is 25% to 30% of total daily calories from fat, with appropriate sources of essential fatty
acids.4,15
Micronutrients
A number of micronutrient deficiencies occur in patients
with eating disorders. In both anorexia nervosa and bulimia
nervosa, zinc deficiency is common and documented as
resultant from suboptimal intake attributed to severe
caloric restriction, avoidance of red meat, and/or the adoption of an inadequate vegetarian lifestyle.16 Additionally,
riboflavin, thiamin, calcium, B-vitamin, and magnesium
deficiencies are well documented and are of concern in
both anorexia nervosa and bulimia nervosa.11 It is recommended to routinely screen and subsequently supplement
207
anorectic patients with thiamin and magnesium in addition to addressing any other found deficiencies.17 At onset
of intervention, provide a 100% RDA multivitamin with
minerals.15
Laboratory Assessment
A detailed laboratory assessment is recommended at time of
initial assessment. Although a complete blood count (CBC)
and chemistry profile is recommended, these traditional
tests are typically normal and may underestimate the physical damage and degree of malnutrition. More targeted and
sensitive tests are recommended, including zinc, iron, prealbumin, transferrin, ferritin, 25-OH vitamin D, thiamin,
and complement 3 (C3) level.18 Refer to Figure 19-5 for a
complete recommendation of laboratory tests.
Despite these normal CBC panels, elevated serum
cholesterol and abnormal lipoprotein profiles are often
found in an anorectic patient regardless of consumption
of extremely low-fat and low-cholesterol diets. Arden et al
postulates that mild hepatic dysfunction, decreased bile
acid secretion, and/or hypothalamic dysfunction may
contribute to these abnormalities.19
208
Parenteral Nutrition
Parenteral nutrition (PN) is only indicated in cases of digestive inability as it leads to a continued loss of hunger cues in
the eating-disordered individual. 20 When PN is initiated in
severely malnourished patients, caution needs to be taken
due to the possibility that refeeding syndrome might occur.
Refer to the Refeeding Syndrome section in this chapter for
definitions and guidelines.
EATING DISORDERS
Refeeding Syndrome
Definition and Incidence
Refeeding syndrome can be described as a cascade of
potentially fatal complications caused by shifts in fluid
and electrolytes as nutrition is reintroduced into the body,
taxing wasted and weakened tissues and demanding more
nutrients than are readily available.21,22 It is manifested in
an assemblage of symptoms that result from rapidly and
inappropriately refeeding (via oral, enteral, or parenteral
route) individuals who have been malnourished or starved
for a period of time, usually exceeding 7 to 10 days. 23
Other symptoms of refeeding syndrome include cardiac
dysfunction, edema, and neurological changes.24 Hypophosphatemia is the hallmark clinical sign of refeeding
syndrome, but hypomagnesemia and hypokalemia are also
common indicators. Glucose intolerance and thiamin deficiency are often present as well.25
The exact incidence of refeeding syndrome is unknown,
due in part to the lack of a universal definition 21 and also
poor recognition of the condition. It is known that 30% to
38% of previously unfed patients receiving PN containing
phosphorus experience hypophosphatemia, 26 and 100% of
these patients develop hypophosphatemia when no phosphorus has been added to the PN solution. It has also been
documented that when patients were vigorously refed,
80% experienced hypokalemia, hypomagnesemia, and/or
hypophosphatemia.27
209
210
70% ideal body weight are at the greatest risk.22 Categories of patients who may meet these criteria include those
with anorexia nervosa, alcoholism, cancer, uncontrolled
diabetes, marasmus, malabsorptive syndrome (eg, pancreatitis, cystic fibrosis, short bowel), prolonged fasting, morbid
obesity with profound weight loss, prolonged antacid use
(due to binding of phosphorus), and long-term diuretic use
(due to electrolyte losses), as well as postoperative patients,
the elderly, and patients allowed nothing by mouth for
greater than 5 to 7 days.21
If a patient meets the preceding high-risk criteria
for refeeding syndrome, there are several acceptable
approaches for preventing or treating refeeding (Table
19-1). Importantly, baseline electrolytes (including potassium, phosphorus, magnesium, and calcium) should be
obtained and corrected if low prior to the initiation of feeds.1
Electrolyte monitoring should continue 1 to 4 times per day
depending upon the severity of malnutrition, for the first 3
days.24 During this time, calories may be introduced at 50%
of goal, not to exceed 20 to 25 kcal/kg/d.1,21,22,24 Macronutrient distribution should limit carbohydrate intake to
2 to 3 g/kg/d based on actual body weight. No restriction
is necessary for protein or fat intake, and common recommendations for each are 1 to 1.5g/kg/d1,22,24 and 1 g/kg/d,
respectively.1 Fluid should be restricted to 800 to 1000
mL/d due to the potential risk of fluid overload and cardiac
decompensation.22,24
Days 47
Days 814
Calories
Carbohydrate
23 g/kg/d
Protein
Fat
Fluid
11.5 g/kg/d
1 g/kg/d
8001000 mL/d
Phosphorus
200300 mg daily
Multivitamin/mineral supplement
daily
Potassium
Magnesium
Thiamin
Other vitamins/minerals
EATING DISORDERS
During these first few days of renourishment, electrolytes, if low, should be corrected aggressively. Potassium
phosphate preferably, or sodium phosphate in the presence
of normal serum potassium, can be given intravenously for
moderate to severe hypophosphatemia.25 Different references recommend infusing 9 to 18 mmol over anywhere
from 2 to 12 hours.21,25,32 For orally fed patients with mild
to moderate hypophosphatemia, cows milk is an excellent
source of both phosphorus and potassium,25 and can be used
to treat mild electrolyte derangements. Oral sodium phosphate can also be used, at 500 mg 4 times per day until serum
phosphorus is stable, then decreased to 250 mg 3 times per
day for maintenance.22 Mild to moderate hypomagnesemia
can be treated with an initial dose of 0.5 mmol/kg over a
24-hour infusion, then maintained at 0.25 mmol/kg/d for
the next 5 days to maintain serum levels.21 For severe hypomagnesemia, infuse 24 mmol over 6 hours, then follow with
0.25 mmol/kg/d for the next 5 days as above.21
In addition to the attention paid to macronutrients and
electrolytes, patients at risk of refeeding should receive a
daily multivitamin/mineral supplement. Any signs or symptoms of thiamin deficiency can be treated with 200 to 300
mg of oral thiamin daily for 10 days to correct deficiency. 21
After electrolytes have stabilized and the patient has
received 72 hours of nutrition at 50% of goal, calories can
gradually be increased every 3 days by 200 to 300 kcal. 22,24,33
Continue to monitor and correct electrolytes as feedings
progress for the duration of the first 2 weeks of feeding. 21
With awareness and proper monitoring, refeeding
syndrome can be prevented or managed appropriately to
prevent serious complications and the potential of death.
Monitoring and correction of electrolytes, supplementation of nutrients, and conservative administration of
carbohydrate and fluid can save lives of those at highest risk
forrefeeding.
1. The following are common physical signs and symptoms of anorexia nervosa:
A. Lanugo-type hair, cyanosis of the extremities, and
erosion of the dental enamel
B. Cyanosis of the extremities, erosion of the dental
enamel, and Russells sign
C. Cachexia, cyanosis of the extremities, and muscle
wasting
D. Cachexia, Russells sign, and sore red throat
211
References
212
12. Kotler LA, Cohen P, Davies M, Pine DS, Walsh BT. Longitudinal relationships between childhood, adolescent, and
adult eating disorders. J Am Acad Child Adolesc Psychiatry.
2001;40(12):14341440.
13. Fairburn CG, Cowen PJ, Harrison PJ. Twin studies
and the etiology of eating disorders. Int J Eat Disord.
1999;26:349358.
14. de Zwaan M, Aslam Z, Mitchell JE. Research on energy
expenditure in individuals with eating disorders: a review. Int
J Eating Disord. 2002;32:127134.
15. The Royal College of Psychiatrists. Guidelines for the nutritional management of anorexia nervosa. http://www.rcpsych.
ac.uk/files/pdfversion/cr130.pdf. Accessed December 15,
2008.
16. Bakan R, Birmingman CL, Aeberhardt L, Goldner EM.
Dietary zinc intake of vegetarian and nonvegetarian patients
with anorexia nervosa. Int J Eating Disord. 1993;13:229233.
17. Winston AP, Jamieson CP, Madira W, et al. Prevalence of
thiamin deficiency in anorexia nervosa. Int J Eat Disord.
2000;28:451454.
18. Woosley M. Eating Disorders: A Clinical Guide to Counseling
and Treatment. Chicago, IL: American Dietetic Association;
2002.
19. Arden MR, Weiselberg EC, Nussbaum MP, et al. Effect of
weight restoration on the dyslipoproteinemia of anorexia
nervosa. J Adolesc Health. 1990;11:199202.
20. Setnick JS. The Eating Disorders Clinical Pocket Guide: Quick
Reference for Healthcare Professionals. Snack Time Press;
2005.
21. Mehanna HM, Moledina J, Travis J. Refeeding syndrome:
What it is, and how to prevent and treat it. BMJ.
2008;336:14951498.
20
Food Allergies
Mary Beth Feuling, MS, RD, CD, CNSD, Michael B. Levy, MD, and Praveen S. Goday, MBBS, CNSP
Learning Objectives
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Definitions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Pathophysiology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Major Food Allergens. . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Clinical Presentation. . . . . . . . . . . . . . . . . . . . . . . . . . . . .
213
213
214
215
215
215
IgE-Mediated Diseases
Mixed IgE and Non-IgE Mediated Diseases
Non-IgE Mediated Disease
Introduction
Definitions
Several terms may be used when defining adverse reactions to foods. An abnormal response to a food may include
allergy, hypersensitivity, or intolerance. Tolerance
usually refers to the ability to consume a food that may have
the potential for allergy or a food that previously caused
allergy and is now consumed without sequelae.
Adverse reactions to foods may occur within a spectrum of reactions ranging from immunoglobulin E (IgE)
to non-IgE mechanisms. Generally speaking, allergy or
hypersensitivity refers to IgE-mediated events and intolerance refers to non-IgE events. Other important definitions
that will be used in this chapter are outlined in Table 20-1.
213
214
Table 20-1 Definitions of Common Terms That Are Frequently Used in Association with Food Allergies
Term
Definition
For many clinicians, defining a food reaction as IgE-mediated or non-IgE mediated has great utility. IgE reactions
have been well understood and chemically described as a
cascade of events which involves a process that results from
mast cell or basophil degranulation at mucosal surfaces
or the skin. Because IgE can be quantitatively measured,
levels of food-specific IgE may aid in the diagnosis of IgEmediated food allergy and serial food-specific IgE levels
may be followed to help determine the development of
clinical tolerance.
Non-IgE food intolerance may include immunologic
and non-immunologic reactions due to the effects of other
components within food (eg, lactose, seafood toxins, or
naturally occurring pharmacologically active compounds
such as tyramine). These substances may cause an adverse
reaction, but are differentiated from true food allergy
because they do not involve the IgE cascade.
Epidemiology
Table 20-2 Clinical Food Allergy Syndromes Associated with IgE or Non-IgE Mechanisms
IgE-Mediated Syndromes
Non-IgE-Mediated Syndromes
Eosinophilic esophagitis
Eosinophilic gastritis
Eosinophilic gastroenteritis
Atopic dermatitis
Protein-induced enterocolitis
Protein-induced enteropathy
Food protein-induced enterocolitis syndrome (FPIES)
Dermatitis herpetiformis, gluten enteropathy
FOOD ALLERGIES
Pathophysiology
The production of IgE antibody may develop in the genetically predisposed individual through mechanisms that
involve multiple factors. Once allergen-specific IgE is
produced, binding to the high-affinity IgE receptor which
is present on mast cells and basophils occurs. Low-affinity
IgE receptors are present on eosinophils, monocytes, and
macrophages.6
There are multiple host, antigen, and allergen factors
that may be involved in the IgE-sensitization cascade which
may result in the subsequent development of clinical allergy.
These factors include the genetics of the host, immunologic
competence at the mucosal level, and allergen presentation by intact antigen-processing cells, as well as the route
of exposure to the allergen. Sensitization may occur via
215
Clinical Presentation
IgE-Mediated Diseases
The major IgE-mediated allergic diseases are oral allergy
syndrome, anaphylaxis, urticaria, and angioedema.
The pollen-associated oral allergy syndrome presents
with pruritus of the lips, palate, tongue, and oropharynx
following oral mucosal contact with fresh fruits and vegetables. The reaction usually does not occur following a
cooking process because the cross-reactive allergen is very
heat sensitive. These symptoms usually resolve without
treatment and generally do not progress to cause more
systemic involvement. Cross-reactivity between plant
pollens and fruits is responsible for the clinical syndrome.
Specifically, patients with ragweed sensitivity may have
these symptoms after ingesting watermelon, cantaloupe,
banana, or honeydew while patients sensitive to birch
pollen may notice symptoms with apple, pear, celery,
carrot, or peach.
Food-induced anaphylaxis is the most severe form of
2010 A.S.P.E.N. www.nutritioncare.org
216
Allergy Testing
Allergy skin prick testing is commonly used by the practicing allergist-immunologist to determine the presence
of IgE to specific foods. Clinical correlation of the patient
history to the testing results is important. The skin prick
technique is highly reproducible and extracts for these
tests are commercially available for hundreds of airborne
and food allergens. These tests are performed by applying
the extracts by a prick or puncture technique to the palmar
surface of the forearm or upper back. The allergy prick test
is actually a localized mediator-release phenomenon which
occurs following allergen presentation to skin mast cells.
The reaction is a nearly immediate wheal and flare reaction characteristic of IgE-mediated allergy. The test is read
within 20 minutes and correlates closely with the presence
of specific IgE to the suspected allergen. Positive tests indicate the presence of IgE but not clinical reactivity with an
estimated false positive rate of approximately 50%. A negative test has high negative predicted value of nearly 95%,
thus excluding the role of IgE.13
In-vitro radioallergosorbent tests (RAST) are blood
tests that are available for the determination of serumspecific IgE with close correlation to skin prick testing
results. The Pharmacia ImmunoCAP system has been
studied with food challenge results showing a greater than
95% predictive value for reactions to peanut, egg, and milk.
There are a small number of false negative ImmunoCAP
tests for peanuts. The established values can be utilized by
clinicians to determine when a food challenge may be safe
to perform in the patient with IgE-mediated food allergy.14
Management
FOOD ALLERGIES
217
Nutrition Assessment
Restriction of a childs diet due to the diagnosis of food allergies may have a severe impact on his or her nutrition intake.
This section provides a practical approach to identifying the
risk factors that can lead to nutrition deficiencies, undernutrition, and poor growth while providing guidelines for a
comprehensive nutrition assessment.
Because strict avoidance of the causative food is necessary, it is critical to clearly define the avoidance list. The
218
FOOD ALLERGIES
219
Diet 2
Diet 3
Breakfast
Lunch
Dinner
Breakfast
Lunch
Dinner
Breakfast
Lunch
Dinner
Whole milk
Whole milk
Whole milk
Whole milk
Whole milk
Whole milk
Cereal
Peanut
butter & jelly
sandwich
Cooked
carrots with
butter
Meatloaf
Cereal
Meatloaf
Peas
Banana
Peanut
butter & jelly
sandwich
Cooked
carrots
with butter
Enriched
soymilk
Cereal
Peas
Banana
Enriched
soy milk
Milk-free,
egg-free
meatloaf
Peas
Strawberries
Mashed
potatoes
Enriched
soymilk
Soynut butter
and jelly
sandwich
Cooked
carrots with
milk-free
margarine
Strawberries
Banana
Strawberries
Mashed
potatoes
Roll with
butter
Roll with
butter
Mashed
potatoes
made with
chicken broth
Milk-free roll
with milk-free
margarine
SNACK
SNACK
SNACK
SNACK
SNACK
SNACK
SNACK
SNACK
SNACK
Granola bar
Yogurt drink
Ice cream
Granola bar
Yogurt drink
Ice cream
Soy yogurt
Juice
Oatmeal
cookie
Juice
Oatmeal
cookie
Teddy
Grahams
Juice
FAAN Oatmeal
cookie*
Calories
Protein
Fat
1490 kcal
47 g
55 g
Calcium
Vitamin D
Iron
Zinc
1100 mg
203 IU
9.9 mg
8.9 mg
Nutrient
> 100
360
33% of
totalkcal
221
101
141
297
Calories
Protein
Fat
305 kcal
5g
2g
Calcium
Vitamin D
Iron
Zinc
98 mg
20 IU
4 mg
2.6 mg
Nutrient
25
41
6% of
totalkcal
20
10
59
87
Calories
Protein
Fat
1360 kcal
42 g
49 g
% Goal**
Calcium
Vitamin D
Iron
Zinc
754 mg
285 IU
10 mg
6 mg
> 100
321
32% of
totalkcal
151
285
147
201
Table 20-5 Key Micronutrients Provided by the Most Common Food Allergens and Alternative Food Sources That Can Serve as Food Substitutes for the
Allergenic Foods
Allergenic Foods
Micronutrients Provided
Milk
Wheat
Peanut/Tree nut
Fish/Shellfish
Egg
Soy
220
Nutrition Intervention
Education provides a family and patient the pathway for
success with an elimination diet. This includes education
regarding dietary avoidance and consideration of nutrition
deficiencies that may result. In addition, they must receive
education regarding the nutrition goals for the patient in order
to avoid nutrition consequences of food allergies. They must
also be educated about resources for obtaining additional
information regarding living with food allergies (eg, support
groups, local retail establishments that sell allergen-free
foods, cookbooks, and other helpful tips for the elimination
diet). A list of food allergy resources is provided in Table 20-6.
Each food-allergic child/family must be given a list of substitutions in order to be successful with strict avoidance of the
food allergens. In addition, a nutritionally complete formula
or beverage, if possible, should be encouraged. This type of
information assists the patient and family in living a normal
and well-nourished life despite having food allergies. Without
education, the recommendation of an elimination diet can be
overwhelming and unsuccessful as families struggle to find
accurate and useful information.
Protein
Examples
Cows Milk
Casein, whey
Infant formulas
PediaSure
Lactose-free
Infant formulas
Similac Sensitive
Formulas for older children
PediaSure
Soy
Soy
Infant formulas
Enfamil ProSobee
Similac Isomil Advance
Formulas for older children
Website
http://www.foodallergy.org
http://www.aaaai.org
http://www.eatright.org
http://www.aafa.org
http://www.apfed.org
http://www.acaai.org
Hydrolysate
Peptides,
amino acids
Infant formulas
Nutramigen
Similac Alimentum
Formulas for older children
Vital jr
Peptamen Jr
Elemental
Amino acids
Infant formulas
Neocate
EleCare
Nutramigen AA
older children (ELEMENTAL)
Neocate Jr
EleCare
EO28 Splash
FOOD ALLERGIES
221
Milk Substitutes
Milk substitutes must be used in combination with nutrition assessment and monitoring. There are many different
milk products in the marketplace that continue to
provide alternatives for the allergic patient. However, each
product should only be used with careful consideration of
the nutritional quality of the milk product. Many provide
adequate micronutrients such as vitamin D, calcium, and B
vitamins; however most provide minimal fat and protein.
Children under the age of 2 are at high risk for malnutrition
if one of the incomplete milk substitutes is used in place of
whole cows milk. See Table 20-8 for a list of the nutritional
constituents of various milk substitutes.
Micronutrient Supplementation
Table 20-8 Nutrition Comparisons of Various Milk Substitutes with Whole Milk
Nutrient per 8 oz.
Rice Milk,
Non-Enriched
Rice Milk,
Enriched,
Refrigerated
Soy Milk,
Enriched
Whole Milk
PediaSure
Almond
Milk
Hazelnut
Milk
Calories
120
120
130
150
237
70
110
7.1
2
2
Protein (g)
1
1
7
8
Carbohydrate (g)
25
25
17
11
26
11
18
11.8
2.5
3.5
Fat (g)
2
2
4
8
3.5
3
7.4
2.5
3.5
Unsaturated fat (g)
2
2
0.5
5
3.1
0
0
Saturated fat (g)
0
0
Calcium (mg)
20
300
300
294
230
300
300
1.8
0.1
3.3
0.36
0.36
Iron (mg)
NS
NS
Zinc (mg)
0.29
0.29
0.6
1
2.8
5.4
Selenium (mcg)
NS
NS
9
NS
0.12
0.15
0.107
0.64
NS
Thiamin-B1 (mg)
NS
0.07
0.447
0.5
0.5
0.5
Riboflavin-B2 (mg)
Niacin-B3 (mg)
0.8
0.8
0.8
0.261
4
NS
0.23
0.4
0.883
2.4
NS
60
12
88
NS
Folate (mcg)
1.5
1.5
3
1.07
1.4
NS
Oat Milk
Multigrain
Milk
130
4
24
2.5
2.5
0
300
0.36
0.5
500
100
160
5
30
2
2
0
300
1.08
0.5
500
100
222
Milk
Nuts
Soy
Wheat
Rice
Pasta
Bread/bread crumbs
Bread/bread crumbs
Cereals
Baby food
Bread/bread crumbs
Egg Beaters
Breakfast cereals
Frozen desserts
Breakfast cereals/
granola bars
Egg rolls
Cakes/cookies
Waffles
Crackers
Bread/bread crumbs
Cake/muffin mixes
Candy/chocolate
Candy/chocolate
Frozen desserts
Marshmallows
Canned tuna
Nut butters
Gluten-free products
Chicken hot dogs/
low-fat beef franks
Soy sauce
Waffles
Processed meats
Sauces/chili
Bouillon cubes
trace elements can be used for children with food allergies because the vast majority of these children are normal
except for their food allergies and atopic problems. Chapter
6 (Minerals), Chapter 7 (Water-Soluble Essential Micronutrients), and Chapter 8 (Fat-Soluble Vitamins) discuss these
topics. Recommendations for supplementations should be
made based on foods that need to be eliminated and the
patients nutrition status. There are several hypo-allergenic
multivitamin-multimineral supplements that are appropriate for children with food allergies (Table 20-10).
Table 20-10 Allergen-Free Multivitamins
All of these products are free of milk, soy, egg, wheat, peanut,
tree nut, fish, and shellfish.
Barbecue-flavored
potato chips
Modified food starch
Waffles
Soups
FOOD ALLERGIES
disease generally recurs. Three types of nutrition intervention have met with varying degrees of success in EE. First,
specific food elimination can be based on allergy testing
and clinical history.27 Even when allergy testing does not
reveal specific food allergens, elimination diets can be used.
Simply removing the 8 most common allergenic foods (milk,
soy, egg, wheat, peanut, tree nut, fish, and seafood) from
the diet has significant efficacy.28 Finally, a 100% amino
acid-based formula diet can be utilized, thus removing all
potential food allergens; this approach has been extremely
effective.29,30
Hence, medical nutrition therapy should be considered
as an effective treatment in all children diagnosed with EE.
When deciding on the use of a specific nutrition therapy,
the patients lifestyle and family resources also need to be
considered. This requires comprehensive education and
nutrition monitoring by a dietitian.
223
Enteral Nutrition
Enteral nutrition support of children presenting with food
allergies can be straightforward. Because most enteral
formulas contain cows milk protein, children with cows
milk protein allergies can be managed with soy-based,
protein hydrolysate, or elemental formula using the principles outlined earlier in this chapter.
Some of the formula intolerances that occur in young
children receiving nutrition support are probably secondary
to food allergies and are usually not recognized at the first
instance. Since one of the management strategies for formula
intolerances during nutrition support includes a transition to a protein hydrolysate/elemental formula, the acute
situation usually resolves. Often, food allergy is diagnosed
retrospectively when the child cannot be transitioned back
to a more standard formula.
Parenteral Nutrition
There are minimal data on PN support in children with
documented allergies to foods. Egg allergy can be a cause
for concern because these proteins can be found in intravenous lipid solutions. In patients with documented allergies
to eggs, 3 options could be consideredconsultation with
an allergist who may or may not do a skin prick test, lipidfree PN, or the use of Liposyn II. 32 There is a theoretical
risk with extremely soy-allergic patients needing PN. Most
of these patients probably tolerate intravenous lipid, but the
first 2 options outlined above should be considered.
A variety of allergies to PN have been described
through case reports in the literature. 3238 As with other
allergies, they appear to be more common in children. 3638
Skin rashes appear to be the most common manifestation.
However, they can present with dyspnea, cyanosis, nausea,
vomiting, headache, flushing, fever, and chest pain. Anaphylaxis can occur. 3739 All of these reactions can occur at the
first administration, after several days of administration, or
after reinstitution following a hiatus.
These reactions have been attributed to intravenous
lipid preparations, 32,34 crystalline amino acid solutions, 37
and multivitamin mixtures (either due to stabilizers and
emulsifiers in the M.V.I. Pediatric or due to vitamin
K). 3537,39
When these reactions occur, PN needs to be stopped and
appropriate drug treatment for the allergic reaction started.
If the reaction is severe and the patient is going to continue
to require PN, a multidisciplinary approach utilizing an
allergist, pharmacist, nutrition-support physician, and/
2010 A.S.P.E.N. www.nutritioncare.org
224
References
FOOD ALLERGIES
225
21
CONTENTS
Diabetes Mellitus. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
Definitions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
Type 1 Diabetes Mellitus
Type 2 Diabetes Mellitus
CF-Related Diabetes Mellitus
Consequences of Hyper/Hypoglycemia
in the CriticallyIll Child. . . . . . . . . . . . . . . . . . . . . . . . . . . 227
Glucose Control in Healthy Children with Diabetes. . . . .228
Short-Term Implications
Long-Term Complications
226
Learning Objectives
Diabetes Mellitus
Definitions
Type 1 Diabetes Mellitus
The most common form of diabetes in children remains
type 1 diabetes mellitus. This disorder is usually an autoimmune destruction of the beta cells of the pancreas, resulting
in an absolute deficiency of insulin.14 Incidence of type 1
diabetes mellitus in the United States and other western
countries has been increasing. In the United States, the
prevalence of type 1 diabetes mellitus at 18 years of age is 2
to 3 per 1000.1 The incidence of type 1 diabetes mellitus is
about 1.5 times greater in the American non-Hispanic white
population than in African Americans or Hispanic Americans. Children with type 1 diabetes mellitus are at risk for
developing ketoacidosis and require insulin to prevent
hyperglycemia.2 Because of a risk for developing low blood
glucose concentrations, current recommendations from the
American Diabetes Association (ADA) are to keep target
blood glucose goal ranges in children somewhat higher than
what is recommended for adults.2,4
Hyperglycemia appears to be common in pediatric intensive care units (ICUs) regardless of whether the child has
known diabetes mellitus.1619 In one retrospective study
involving 152 children in a pediatric ICU, blood glucose
concentrations greater than 125 mg/dL were observed in
over half of the patients within 24 hours of admission and
227
in almost 90% of the patients sometime during the admission.16 In a second retrospective study, almost 70% of 192
critically ill children had blood glucose concentrations
greater than 120 mg/dL within 24 hours of admission to a
pediatric ICU.17
Hyperglycemia has been linked to poor outcome. In
a study of 184 children less than 1 year of age who had
undergone cardiac surgery, hyperglycemia in the postoperative period was associated with increased mortality and
morbidity.18 However, in a fourth retrospective study of
1094 admissions to a pediatric intensive care unit, the risk of
having a blood glucose value over 200 mg/dL was less than
the risk of having a blood glucose value less than 65 mg/dL.18
Furthermore, the risk of dying was 6 times greater if the child
had a blood glucose less than 110 mg/dL than if all blood
glucose values were greater than 110 mg/dL.19
There are many reasons why hyperglycemia might affect
mortality and morbidity in critically ill patients. In vitro, high
glucose concentrations have been shown to cause abnormalities in several aspects of immune function, including
intracellular killing, complement function, granulocyte
adhesion, chemotaxis, phagocytosis, and respiratory burst
function.20 Glucose attaches itself to the third component of
complement, affecting this components ability to attach itself
to microbes and impairing opsonization of the microbe.21
On the other hand, it is not difficult to understand why low
blood sugars might affect mortality and morbidity in critically ill children. More than 60% of the basal metabolic rate
in an infant is estimated to be related to brain metabolism,
compared with less than 30% in an adult.22
Numerous studies in adult patients with diabetes have
shown increased morbidity and mortality associated with
hyperglycemia.23,24 These studies formed the backdrop for
a large retrospective study in surgical patients showing that
intensive insulin therapy reduced in-hospital mortality rates
by 34% and had a profound effect on a variety of morbidities.21
A similar retrospective study in non-surgical patients also
showed reductions in mortality and morbidity, although the
results were not as striking.25 These reports led many intensive care units to modify their management of hyperglycemia
in diabetic and non-diabetic patients. Since those initial
studies, there have been a plethora of additional reports, with
varying conclusions.26,27 Additional studies have focused on
the increased morbidity and morality associated with hypoglycemia in adult hospitalized patients,28,29 consistent with
the findings in children.
The optimal control of blood glucose in the adult hospitalized patient with or without diabetes remains controversial.
There are very limited data in children. While one recent
2010 A.S.P.E.N. www.nutritioncare.org
228
Long-Term Complications
In contrast to some older children with type 2 diabetes,
children with type 1 diabetes rarely have complications at
the time of diagnosis.38 A large prospective, randomized
study called the Diabetes Control and Complications Trial
(DCCT) established that the major risk factor for microvascular complications is glycemic control.3941 What appears to
be important is the exposure to elevated glucose concentrations over time.38 However, despite marked improvement in
treatment options, 12 years after diagnosis more than 50% of
patients with type 1 diabetes had developed complications or
comorbities.39,42 Persistently high blood glucoses over time
also appear to increase the risk of macrovascular complications, but these rarely occur in childhood.
are easiest to control if the PN is given as a continuous infusion, rather than being cycled. It should also be mentioned
that even if the IV infusion of dextrose is stopped, patients
with type 1 diabetes mellitus will continue to need some
insulin to inhibit hepatic gluconeogenesis and prevent the
child from developing ketoacidosis.
Administration of Insulin
Blood glucose concentrations in children with diabetes on
enteral nutrition (EN) support can usually be adequately
controlled by using subcutaneous injections of insulin.
Guidelines for adults have been published for the administration of insulin at the initiation of tube feedings, as the rate
of tube feedings increases, and for continuous intermittent
and nocturnal feeding schedules.44 By dosing the insulin on
a per kilogram body weight basis, these recommendations
can also be utilized in children. Of course, careful monitoring of blood glucose concentrations is required.
229
230
Panhypopituitarism
Another relatively uncommon endocrine condition that
might affect nutrition support is panhypopituitarism. Again,
no guidelines exist for how to manage nutrition support in
such patients. Children with panhypopituitarism are unable
to secrete a number of anterior pituitary hormones, including
growth hormone and corticotropin. This condition can be
the result of intracranial surgery, but can also be idiopathic.
Children with this condition often present with hypoglycemia and are at continuing risk for low blood sugars. The
hypoglycemia is due to an inability to counterregulate. 51 To
prevent hypoglycemia in a critically ill child with this condition, additional glucocorticoids are administered. In such
patients, a strong case can also be made for administering
nutrition support as a constant infusion, rather than giving
enteral feeds as boluses or cycled PN.
Future Research
References
16. Srinivasan V, Spinella PC, Drott HR, Roth CL, Helfaer MA,
Nadkarni V. Association of timing, duration, and intensity of
hyperglycemia with intensive care unit mortality in critically ill
children. Pediatr Crit Care Med. 2004;5(4):329336.
17. Faustino EV, Apkon M. Persistent hyperglycemia in critically ill
children. J Pediatr. 2005;146(1):57.
18. Yates AR, Dyke PC 2nd, Taeed R, et al. Hyperglycemia is a
marker for poor outcome in the postoperative pediatric cardiac
patient. Pediatr Crit Care Med. 2006;7(4):351355.
19. Wintergerst KA, Buckingham B, Gandrud L, Wong BJ, Kache S,
Wilson DM. Association of hypoglycemia, hyperglycemia, and
glucose variability with morbidity and death in the pediatric
intensive care unit. Pediatrics. 2006;118(1):173179.
20. Van den Berghe G, Wouters P, Weeker F, et al. Intensive
insulin therapy in the critically ill patients. N Engl J Med.
2001;345(19):13591367.
21. McMahon MM, Bistrian BR. Host defenses and susceptibility
to infection in patients with diabetes mellitus. Infect Dis Clin
North Am. 1995;9(1):19.
22. Haliday MA. Metabolic rate and organ size during growth from
infancy to maturity and during late gestation and early infancy.
Pediatrics. 1971;47(1):167179.
23. Capes SE, Hunt D, Malmberg K, Gerstein HC. Stress hyper
glycemia and increased risk of death after myocardial infarction
in patients with and without diabetes: a systematic overview.
Lancet. 2000:355(9206):773778.
24. Capes SE, Hunt D, Malmberg K, Pathak P. Stress
hyperglycemia and prognosis of stroke in nondiabetic
and diabetic patients: a systematic overview. Stroke.
2001;32(10):24262432.
25. Van den Berghe G, Wilmer A, Hermans C, et al. Intensive insulin therapy in the medical ICU. N Engl J Med.
2006;345(5):449461.
26. Inzucchi Se, Slegel MD. Glucose control in the ICUhow tight
is too tight? N Engl J Med. 2009;360(13):13461349.
27. The NICE-SUGAR Study Investigators. Intensive versus
conventional glucose control in critically ill patients. N Engl J
Med. 2009;360(13):12831297.
28. Turchin A, Matheny ME, Shubina M, et al. Hypoglycemia and
clinical outcomes in patients with diabetes hospitalized in the
general ward. Diabetes Care. 2009;32(7):11531157.
29. Arabi Y, Tamim HM, Rishu AH. Hypoglycemia with intensive insulin therapy in critically ill patients: Predisposing
factors and association with mortality. Crit Care Med.
2009;37(9):25362544.
30. Vlasselaers D, Milants I, Desmet L, et al. Intensive
insulin therapy for patients in paediatric intensive care:
a prospective, randomized controlled study. Lancet.
2009;373(9663):547556.
31. Agus MSD, Hirshberg EL. Pediatrics: Intensive insulin therapy
in critically ill children. Nature Rev Endo. 2009;5(7):360362.
32. Northam EA, Anderson PJ, Werther GA, Warne GL, Adler
RG, Andrewes D. Neuropsychological complications of
IDDM in children 2 years after disease onset. Diabetes Care.
1998;21(3):379384.
33. Rovet J, Alvarez M. Attentional functioning in children and
adolescents with IDDM. Diabetes Care. 1997;20(5):803810.
231
22
Bridget Reineking, MS, RD, CD and Sandy van Calcar, PhD, RD, CD
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
Phenylketonuria. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
Natural History
Maternal Phenylketonuria
Acute Management
Chronic Management
Galactosemia. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 241
Natural History
Acute Management
Chronic Management
232
Learning Objectives
Background
233
devoid of the amino acid(s) that cannot be fully metabolized, but will provide all other amino acids, carbohydrates,
fat sources, vitamins, and minerals. Medical foods often
provide a significant portion of a childs nutrition needs. 35
Figure 22-1 Basic Principles of Nutrition Management of Inborn Errors
ofMetabolism
234
Phenylketonuria
Natural History
PKU is an inborn error of phenylalanine (phe) metabolism
caused by a deficiency of the hepatic enzyme phenylalanine
hydroxylase (PAH), which catalyses the hydroxylation
of phe to tyrosine (Figure 22-2). PKU is an autosomal
recessive disorder with a carrier frequency of 1 in 50 and
incidence of approximately 1 in 10,000 to 15,000 births in
those of European ancestry. More than 500 mutations have
been identified in the PAH gene, but genotype/phenotype
correlations have not proven to predict outcome. 8
Maternal Phenylketonuria
Adapted from Acosta PB, Yannicelli S. The Ross Metabolic Formula System
Nutrition Support Protocols. 4th ed. Columbus, OH: Ross Products
Division/Abbott Laboratories; 2001. Copyright 2001 with permission
from Abbott Nutrition.
Acute Management
Initial Presentation
When an infant with PKU is identified by newborn
screening, treatment should be initiated as soon as possible.
Depending on the degree of elevation in plasma phe, dietary
phe is eliminated or greatly reduced in the diet until blood
phe levels decrease to the treatment range of 2 to 6 mg/dL.
This can be accomplished by feeding exclusively a phe-free,
but otherwise nutritionally complete, medical food. Once
plasma phe concentrations are reduced to < 6 mg/dL, a
limited quantity of an intact protein is added to the medical
food to meet minimum phe needs for growth and protein
maintenance (Table 22-2). 35 The intact protein source can
be provided by a standard infant formula or breast milk.
Phe concentrations may increase during times of illness
or severe injury. Prevention of catabolism can minimize
these elevations. During illness, individuals with PKU are
encouraged to reduce phe intake but continue to consume
the phe-free medical food. If gastrointestinal symptoms
develop, the medical food can be discontinued and carbohydrate-based beverages can be provided to increase caloric
intake to help slow catabolism. 3 Efforts should be made to
PHE
(mg/kg)
Nutrient
TYR
Protein
(mg/kg)
(g/kg)
Energy
(kcal/kg)
Infants
0 to < 3 mo
25 70
3 to < 6 mo
20 45
6 to < 9 mo
15 35
9 to < 12 mo
10 35
(mg/d)
1 to < 4 yr
200 400
4 to < 7 yr
210 450
7 to < 11 yr
220 500
120
(145 95)
120
300 350 3.50 3.00
(145 95)
110
250 300 3.00 2.50
(135 80)
105
250 300 3.00 2.50
(135 80)
(g/d)
(g/d)
(kcal/d)
1,300
1.72 3.00
30
(900 1800)
1,700
2.25 3.50
35
(1300 2300)
2,400
2.55 4.00
40
(1650 3300)
300 350
3.50 3.00
Women
11 to < 15 yr
250 750
3.45 5.00
50
15 to < 19 yr
230 700
3.45 5.00
55
19 yr
220 700
3.75 5.00
60
11 to < 15 yr
225 900
3.38 5.50
55
15 to < 19 yr
65
19 yr
70
2,200
(1500 3000)
2,100
(1200 3000)
2,100
(1400 2500)
Men
2,700
(2000 3700)
2,800
(2100 3900)
2,900
(2000 3300)
Adapted from Acosta PB, Yannicelli S. The Ross Metabolic Formula System
Nutrition Support Protocols. 4th ed. Columbus, OH: Ross Products
Division/Abbott Laboratories; 2001. Copyright 2001 with permission
from Abbott Nutrition.
Chronic Management
The goal of chronic management for PKU is restriction of
dietary phe to maintain blood phe concentrations within
the recommended treatment range of 2 to 6 mg/dL from
infancy to age 12 years and 2 to 10 mg/dL in adolescents.14
However, with the concern of maternal PKU, adolescent girls should be encouraged to maintain levels below
6 mg/dL. Additional treatment goals include maintenance
of adequate growth velocity and weight gain, prevention
of protein deficiency, and achieving adequate macro- and
micronutrient status. 35
235
= 49 g of Infant
Formula A Needed
= 5.3 g of protein
= 254 kcals
= 40 g of phe-free
medical food
= 192 kcal
Final Recipe
Product
Infant Formula A
Phe-free medical
food B
Total
Amount
Phe (mg)
Pro (g)
Calories
49 g
162 mg
5g
254 kcal
40 g
0 mg
6g
192 kcal
162 mg
11 g
446 kcal
236
Amount
Eaten
Phe
Protein
Calories
Medical Food
Froot Loops
Crackles
Low-Protein
Cereal
Blueberries,
fresh
Medical Food
French Fries,
Ore-Ida
Golden Fries
Broccoli,
cooked
Thousand
Island
Dressing
Peaches,
canned
Cantaloupe,
fresh
Medical Food
Sweet Potato,
with skin,
baked
Green beans,
canned
Pears, canned
Zoo Animal
Crackers,
Farleys
Sorbet,
strawberry
6 oz
6g
0 mg
17 mg
11 g
0.4 g
154 kcal
25 kcal
30 g
7 mg
0.2 g
120 kcal
32 g
8 mg
0.2 g
18 kcal
7 oz
0 mg
13 g
179 kcal
56 g
50 mg
1.4 g
80 kcal
28 g
14 mg
0.6 g
6.0 kcal
16 g
6 mg
0.1 g
59 kcal
57 g
8 mg
0.3 g
42 kcal
27 g
8 mg
0.2 g
9 kcal
7 oz
0 mg
13 g
179 kcal
22 g
25 mg
0.4 g
23 kcal
28 g
14 mg
0.4 g
6 kcal
90 g
8 mg
0.2 g
66 kcal
22 g
20 mg
1.6 g
94 kcal
122 g
15 mg
0.4 g
119 kcal
Total
200
mg
43.4 g
1179
kcal
% from
medical
food
0%
85.3%
43.4%
Meal
Breakfast
Lunch
Snack
Dinner
Snack
hamburgers, egg replacers, and cheese. Several lowprotein cookbooks for PKU are also available.28,29
In PKU, decreased PAH activity reduces the production
of tyrosine; thus, tyrosine becomes an essential amino acid
for this population (Figure 22-2). Medical foods for PKU
are supplemented with tyrosine and additional tyrosine
supplementation is indicated only if the combination of
intact protein and medical food does not meet tyrosine
requirements. Plasma tyrosine concentrations should be
routinely monitored in individuals with PKU. 3,4
Some micronutrients may be insufficient in the low-phe
diet, particularly in those consuming a suboptimal amount
of medical food. Inadequate intake of iron, folate, vitamin
B12 , calcium, and vitamin D has been reported. 3032 Dietary
intake should be analyzed for micronutrient content and
additional supplementation prescribed as needed. 33 In addition, intake of essential fatty acids (EFAs) may be low even
with sufficient intake of medical food and erythrocyte EFA
profiles should be routinely assessed. 3436 If low concentrations are found, vegetable oils such as canola or walnut oil or
a docosahexaenoic acid (DHA) supplement can be added to
the diet. Some medical formulas designed for PKU are now
supplemented with arachidonic acid (ARA) and DHA.
Large neutral amino acids (LNAAs): Phenylalanine
and other LNAAs (leucine, valine, isoleucine, methionine,
tyrosine, tryptophan, and threonine) share common transporters at the blood-brain barrier and intestinal mucosa. In
PKU, competitive inhibition from high concentrations of
phe reduce the transport of other LNAAs into the cerebral
cells, which may reduce synthesis of various neurotransmitters. 37 Supplementation with high doses of LNAAs
can reduce both blood and brain concentrations of phe;
improved executive function skills have been measured
in those with poor dietary control who were treated with
LNAA supplements. 38,39 Several LNAA formulations are
now commercially available.
Cofactor supplementation: A newer therapy for the treatment of PKU is supplementation with a synthetic form
of tetrahydrobiopterin, the cofactor for the PAH enzyme
(sapropterin dihydrochloride, Kuvan) (Figure 22-2).
For some individuals, administration of sapropterin can
improve PAH activity and, thus, lower blood phe levels.40,41
In a phase III randomized, placebo-controlled, double-blind
study, 44% of those taking sapropterin for 6 weeks showed
a reduced phe concentration of 30% or greater.40 Response
to sapropterin needs to be individually assessed as not all
individuals with PKU will respond to supplementation and
the degree of response to the drug varies. Supplementation
with sapropterin rarely allows for complete liberalization of
237
Methylmalonic Acidemia
Natural History
Methylmalonic acidemia (MMA) is an inborn error of
isoleucine (ile), methionine (met), threonine (thr), valine
(val), and odd-chain fatty acid metabolism caused by a
deficiency of the enzyme methylmalonyl-CoA mutase,
which converts methymalonyl CoA to succinyl-CoA with
eventual oxidation in the citric acid cycle (Figure 22-4). In
MMA, methylmalonyl CoA is not metabolized and leads
to accumulation of various methylmalonate metabolites.
The degree of deficiency in the mutase enzyme affects
the clinical outcome of this disorder.6,7,43 Those classified with mut-deficiency have some residual activity and
often a less severe clinical course than those with mut0
deficiency who have no remaining enzyme activity.43,44
Methylmalonyl-CoA mutase requires the cofactor 5-dehydroxyadenosylcobalamin, which is produced from vitamin
B12 . Defects in the production of the cobalamin cofactor can
also cause MMA. The estimated prevalence of MMA is 1 in
80,000 births.45,46
Infants with classic MMA caused by a severe deficiency
of the mutase enzyme often present in the first week of life
with overwhelming illness. Symptoms include poor feeding,
failure to thrive, hypotonia, vomiting, and dehydration with
ketosis, acidosis, hyperammonemia, and hypoglycemia.43,44
Acute episodes are often fatal without aggressive management. Screening for MMA is now included in the expanded
newborn screening panel, which is expected to improve
early diagnosis and clinical outcomes of this disorder.47
Individuals with a deficiency in the mutase enzyme
require medical nutrition therapy, described below. Despite
treatment, those with severe mutase deficiency often have
impaired developmental and medical outcomes.48,49 For
patients with MMA caused by a mild, late-onset mutase
deficiency or a defect in cofactor production, supplementation with high doses of vitamin B12 may improve metabolic
control and allow for more normal development. This is
particularly true for those with a cbl A defect.43 Other
cofactor deficiencies, such as cbl B and cbl C defect, can
have a more complicated clinical course and require both
medical nutrition therapy and vitamin B12 supplementation.43,50 Liver transplantation is now an option for treatment
of this disorder, particularly for those with severe enzyme
deficiency. 5153
2010 A.S.P.E.N. www.nutritioncare.org
238
Figure 22-4 Methylmalonic acidemia is caused by a defect in methylmalonyl CoA mutase. Two of 6 known cobalamin cofactor synthesis defects are
shown, which also cause methylmalonic acidemia.
Adapted from Nyhan WL, Barshop BA, Ozand PT. Atlas of Metabolic Diseases. 2nd ed. New York, NY: Oxford University Press Inc; 2005. Reproduced by
permission of Edward Arnold (Publishers) Ltd.
Acute Management
Initial Presentation
Infants with MMA presenting in an acute episode require
immediate medical attention to control acidosis, hyperammonemia, and hypoglycemia.43,54,55 Nutrition management
during the acute phase of illness concentrates on delivery of
nonprotein calories to help slow catabolism. Typically, intravenous (IV) dextrose at the maximum glucose infusion rate
with additional lipid is provided to achieve maximal caloric
intake. 55 IV carnitine may also be indicated.6
Medical foods for the treatment of MMA are available
which contain all amino acids except ile, met, thr, and val.56
If enteral feedings are poorly tolerated, specialized parenteral
solutions are available that lack the offending amino acids.
As metabolic control improves, a standard infant formula
or standard total parenteral nutrition solution is added to
provide a complete source of protein and meet the individuals
met, val, ile, and thr needs.3,5 Monitoring of ammonia, prealbumin, bicarbonate, and plasma amino acids is necessary to
adjust the dietary prescription to achieve optimal metabolic
control.
Illness
During illness and injury, an increase in the metabolic rate
leads to catabolism of protein. In MMA, the offending
amino acids cannot be utilized and a metabolic episode,
with symptoms similar to those observed during the initial
episode in infancy, can develop. Metabolic crisis associated
with illness or injury can occur at any age and can be life
threatening if not managed aggressively.48,55
2010 A.S.P.E.N. www.nutritioncare.org
Chronic Management
The long-term medical nutrition therapy for individuals
with mutase deficiency includes restriction of the amino
acids ile, met, thr, and val with adequate caloric intake to
prevent catabolism. 3,5,56 Often, a medical food restricted in
these 4 amino acids provides the primary source of calories, other amino acids, and micronutrients. Intact protein
from infant formula or expressed breast milk is added
to provide adequate intake of ile, val, thr, and met. This
allows for adequate growth and protein maintenance, but
prevents excessive intake of these amino acids, which can
lead to excessive production of methylmalonic acid and its
metabolites. 57 The amount of intact protein allowed in the
diet depends on the individuals tolerance for the offending
amino acids. Supplementation with individual amino acids,
particularly ile and/or val, may be required to meet needs of
these offending amino acids without increasing the concentration of other offending amino acids. 3,56 Depending on the
childs clinical status, foods such as fruits, vegetables, and
239
Adapted from Acosta PB, Yannicelli S. The Ross Metabolic Formula System
Nutrition Support Protocols. 4th ed. Columbus, OH: Ross Products
Division/Abbott Laboratories; 2001. Copyright 2001 with permission
from Abbott Nutrition.
Acute Management
Natural History
The most common inborn error of metabolism of the urea cycle
is deficiency of the enzyme ornithine transcarbamylase (OTC)
(Figure 22-5). OTC deficiency impairs urea cycle function,
resulting in hyperammonemia as conversion of ammonia to
nontoxic urea is impaired. Production of arginine is decreased,
and arginine becomes an essential amino acid in this disorder.6,7
OTC deficiency is an X-linked disorder; therefore, males are
often more severely affected than females. However, there is
a wide range of clinical presentations, including adult-onset
OTC deficiency in affected women.60
The severe form of the disease is characterized by
overwhelming hyperammonemia (> 400 mol/L) in the
newborn period causing recurrent vomiting, lethargy,
irritability, and seizures, which can quickly cause coma
and death if not treated aggressively. Mental retardation
is common in those surviving the initial episode.60 Milder
forms of the disorder may not present until later in life after
a severe illness and can include neurological complications,
such as psychosis.61 Long-term outcome with treatment
varies greatly and often depends on the ability to prevent
further episodes of hyperammonemia.
240
Chronic Management
Fatty acid oxidation requires entry of long-chain fatty acids (LCFAs) into the
mitochondria. This process requires L-carnitine. Carnitine cycle enzymes
include acyl-CoA synthetase (AS), carnitine palmitoyltransferase I and II
(CPT I and CPT II), and acylcarnitine/carnitine translocase (CT). Once in the
mitochondria, the -oxidation spiral sequentially oxidizes the fatty acyl-CoA
to the 2-carbon unit acetyl-CoA. Oxidation of LCFA requires very long-chain
acyl-CoA dehydrogenase (VLCAD) and a trifunction protein which includes
3 enzyme activities. In treatment of LCFA, supplementation with medium
chain triglycerides (MCTs) bypasses the long-chain fatty acid enzymes and
utilizes enzymes that oxidize medium and short-chain fatty acids including
medium-chain acyl-CoA dehydrogenase (MCAD) and short-chain acyl-CoA
dehydrogenase (SCAD) enzymes.
Adapted from Nyhan WL, Barshop BA, Ozand PT. Atlas of Metabolic
Diseases. 2nd ed. New York, NY: Oxford University Press Inc; 2005.
Reproduced by permission of Edward Arnold (Publishers) Ltd.
Acute Management
The primary goal of acute management of VLCADD is to
provide sufficient kilocalories to prevent or reduce catabolism of fat stores.6,7 A dextrose infusion at the upper threshold
of the glucose infusion rate is typically provided to reverse
catabolism.55,75 As enteral feeding becomes possible, sources
of glucose and medium-chain triglycerides (MCTs) can be
given to meet calorie demands. Intralipid is contraindicated in
VLCADD because it is a source of LCFAs.
Chronic Management
Long-term management of VLCADD includes prevention of fasting, restriction of long-chain fat intake, and
supplementation with MCTs. 5,74,76,77 Prevention of fasting,
especially in the newborn period when energy stores are
limited, is imperative to prevent breakdown of fat stores.
Fasting guidelines need to be individualized, but typically
fasting is limited to 4 hours for infants up to 4 months of
age.74,78 Longer periods of fasting can be allowed as the
infant ages, but often a feeding during the night is recommended during the first year.76
Nutrition therapy for VLCADD includes modification of fat sources to restrict intake of long-chain fat and
supplement with MCTs. Depending on the severity of the
disorder, long-chain fat intake may need to be reduced
below 20% of total kilocalories during infancy. 5,74,76,77 The
remaining fat kilocalories are supplied by MCTs, which
include fatty acids of 8 to 12 carbons in length and thus can
bypass the enzymatic block in LCFA oxidation. There are
several medical foods available for treatment of VLCADD
which contain limited amounts of long-chain fats and are
supplemented with MCTs. In mild forms of this disorder,
limited breastfeeding may be allowed.74,76,77
With the restriction in long-chain fat, it is important
to assess the intake of the EFAs linoleic acid (LA) and
alpha-linolenic acid (ALA). Supplementation with walnut,
flax, or safflower oil may be necessary to meet LA and ALA
needs.79 Some medical foods are supplemented with ARA
and DHA.
In older children with severe LCFA disorders, restriction of long-chain fat to approximately 10% of total calories
may be necessary to maintain metabolic control.77,80 All
fat from food should be considered long-chain fat since
medium- and short-chain fats are limited in natural
foodsources. Supplementation of low- or nonfat foods and
beverages with a commercial MCT source (available in
oil and powder) provides the remaining calories from fat.
With intense exercise, consuming MCT prior to activity
may be beneficial as an energy source during activity. 81
Addition of raw cornstarch to the nighttime feed may
be indicated in some children with VLCADD. Cornstarch
is a slowly digested source of glucose and is employed in
the treatment of glycogen storage disease to prevent hypoglycemia. 82 Cornstarch supplementation may be helpful
in fatty acid oxidation disorders to prevent low glucose
concentrations and reduce production of abnormal fat
metabolites during fasting.75 Because of poor digestion,
cornstarch is contraindicated before 9 months of age. 82
Routine monitoring to assess metabolic control
241
Galactosemia
Natural History
Classic galactosemia is an autosomal recessive disorder
caused by the enzymatic deficiency of galactose-1-phosphate uridyl transferase (GALT) resulting in elevations of
galactose-1-phosphate (Gal-1-P), galactitol, and galactonate (Figure 22-7).6,7 The incidence of classic galactosemia
is approximately 1 in 60,000 births. Various mutations
have been identified in the GALT gene. In the Caucasian
population, a common mutation is Q188R; homozygosity
of this mutation results in a severe phenotype, often with
0% enzyme.84 Another common mutation is S135L, which
results in a milder clinical course and is prevalent in the
African American population. Another form of galactosemia
is the Duarte variant, which results in a mild phenotype that
may not require dietary intervention. 85
Figure 22-7 Classic galactosemia is a caused by a deficiency of
galactose-1-phosphate uridyl transferase (GALT). Elevations in galactose1-phosphate, galactose, galactitol and galactonate are present in this
disorder.
Adapted from Acosta PB, Yannicelli S. The Ross Metabolic Formula System
Nutrition Support Protocols. 4th ed. Columbus, OH: Ross Products
Division/Abbott Laboratories; 2001. Copyright 2001 with permission
from Abbott Nutrition.
2010 A.S.P.E.N. www.nutritioncare.org
242
Acute Management
Infants identified with classic galactosemia should be
immediately placed on a soy-based infant formula. Soy
formulas, which contain soy protein isolate as the protein
source, have a very low galactose content compared to
cows milk-based formulas or breast milk. If an infant
does not tolerate enteral feeds, standard total parenteral
nutrition may be used. Efforts should be made to choose
medications that are free of lactose extenders. Unlike
some disorders of amino acid and fat metabolism, those
with galactosemia do not develop metabolic episodes
associated with illness.
Chronic Management
Long-term management of classic galactosemia requires
restriction of galactose in the diet. Galactose is primarily
derived from lactose.91 During infancy, powdered soy
formula is provided and breastfeeding is not allowed.
Powdered soy formula is recommended over ready-to-feed
or concentrated liquid soy formulas. Liquid soy formulas
contain a higher galactose content from the addition of
carageenan, although the digestive availability of galactose
from carageenan is unclear.92 Use of lactose-free elemental
formulas, which contain no galactose, have been used to
treat some infants with classic galactosemia with Gal-1-P
concentrations that have not decreased into the treatment
range by 4 to 6 months of age.93
When starting solids, all dairy products are contraindicated. Caregivers are instructed to check food labels
for lactose- and galactose-containing foods and ingredients (Table 22-4). Galactose is also found in organ meats
and some legumes. Fruits and vegetables contain varying
2010 A.S.P.E.N. www.nutritioncare.org
References
243
10. Bickel H, Gerrard J, Hickmans EM. The influence of phenylalanine intake on phenylketonuria. Lancet. 1953; 265:812813.
11. Berry HK, Wright S. Conference on treatment of phenylketonuria. J Pediatr. 1967;70:142147.
12. Koch R, Burton B, Hoganson G, et al. Phenylketonuria
in adulthood: a collaborative study. J Inherit Metab Dis.
2002;25:333346.
13. Azen CG, Koch R, Friedman EG, et al. Intellectual development in 12-year-old children treated for phenylketonuria. Am
J Dis Child. 1991;145:3539.
14. National Institutes of Health. Phenylketonuria (PKU):
Screening and Management. NIH Consensus Statement.
Washington DC; 2000.
15. Waisbren SE, Noel K, Fahrbach K, et al. Phenylalanine blood
levels and clinical outcomes in phenylketonuria: a systematic literature review and meta-analysis. Mol Genet Metab.
2007;92:6370.
16. Levy H, Ghavami M. Maternal phenylketonuria: A metabolic
teratogen. Teratology. 1996;53:176184.
17. Waisbern SF, Hanley W, Levy HL, et al. Outcomes at age
4 years in offspring of women with maternal phenylketonuria: the Maternal PKU Collaborative Study. JAMA.
2000;283:756762.
18. Waisbren SE, Azen C. Cognitive and behavioral development in maternal phenylketonuria offspring. Pediatrics.
2003;112:15441547.
19. Yagasaki M, Hashimoto S. Synthesis and application of
dipeptides; current status and perspectives. Appl Microbiol
Biotechnol. Epub. 2008;81:1322.
20. van Rijn M, Bekhof J, Dijkstra T, Smit PG, Moddermam P, van
Spronsen FJ. A different approach to breast-feeding of the infant
with phenylketonuria. Eur J Pediatr. 2003;162:323326.
21. Greve LC, Wheeler MD, Green-Burgeson DK, Zorn EM.
Breast-feeding in the management of the newborn with
phenylketonuria: a practical approach to dietary therapy. J Am
Diet Assoc. 1994;94:305309.
22. Lawrence RL. Breastfeeding: A Guide for the Medical Profession.
5th ed. St. Louis, MO: C.V. Mosby Co; 2005.
23. Leamons JA, Reyman D, Moye L. Amino acid composition
of preterm and term breast milk during early lactation. Early
Hum Dev. 1983;8:323329.
24. Gropper SS, Acosta PB. Effect of simultaneous ingestion of
L-amino acids and whole protein on plasma amino acid and
urea nitrogen concentrations in humans. J Parenteral Enteral
Nutr. 1991;15:4853.
25. Otten JJ, Helwig JP, Meyers LD. Dietary Reference Intakes: The
Essential Guide to Nutrient Requirements. Washington, DC: The
National Academies Press; 2006.
26. Singh R, Lesperance E, Crawford K. PKU Food List. 2nd ed.
Atlanta, GA: Emory University, Department of Human
Genetics, Division of Medical Genetics; 2006.
27. Schuett V. Low Protein Food List for PKU. 2nd ed. Seattle, WA:
National PKU News; 2002.
28. Schuett V. Low Protein Cookery for PKU. 4th ed. Madison, WI:
The University of Wisconsin Press; 1996.
29. Schuett V, Corry D. Apples to Zucchini: A Collection of Favorite
Low Protein Recipes. Seattle, WA: National PKU News; 2005.
244
30. Acosta PB, Yannicelli S, Singh RH, Elsas LJ, Mofidi S, Steiner
RD. Iron status of children with phenylketonuria undergoing
nutrition therapy assessed by transferrin receptors. Genet Med.
2004;6:96101.
31. Robinson M, White FJ, Cleary MA, et al. Increased risk of
vitamin B12 deficiency in patients with phenylketonuria on an
unrestricted or relaxed diet. J Pediatr. 2000;136:545547.
32. Acosta PB, Yannicelli S. Plasma micronutrient concentrations
in infants undergoing therapy for phenylketonuria. Biol Trace
Elem Res. 1999;67:7584.
33. Hvas AM, Nexo E, Nielsen JB. Vitamin B12 and vitamin B6
supplementation is needed among adults with phenylketonuria (PKU). J Inherit Metab Dis. 2006;29:4753.
34. Moseley K, Koch R, Moser AB. Lipid status and long-chain
polyunsaturated fatty acid concentrations in adults and adolescents with phenylketonuria on phenylalanine-restricted diet. J
Inherit Metab Dis. 2002;25:5664.
35. Koletzko B, Beblo S, Demmelmair H, Hanebutt FL. Omega-3
LC-PUFA supply and neurological outcomes in children
with phenylketonuria (PKU). J Pediatr Gastroenterol Nutr.
2009;48(suppl):S27.
36. Beblo S, Reinhardt H, Demmelmair H, Muntau AC, Koletzko
B. Effect of fish oil supplementation on fatty acid status, coordination, and fine motor skills in children with phenylketonuria.
J Pediatr. 2007;150:479484.
37. Puglisi-Allegra S, Cabib S, Pascucci T, et al. Dramatic brain
aminergic deficit in a genetic mouse model of phenylketonuria. Neuroreport. 2000;11:13611364.
38. Matalon R, Michals-Matalon K, Bhatia G, et al. Double blind
placebo controlled trial of large neutral amino acids in treatment of PKU: effect on blood phenylalanine. J Inherit Metab
Dis. 2007;30:153158.
39. Schindeler S, Ghosh-Jerath S, Thompson S, et al. The effects
of large neutral amino acid supplements in PKU: an MRS and
neuropsychological study. Mol Genet Metab. 2007;91:4854.
40. Levy HL, Milanowski A, Chakrapani A, et al. Efficacy of
sapropterin dihydrochloride (tetrahydrobiopterin, 6R-BH4)
for reduction of phenylalanine concentrations in patients with
phenylketonuria: a phase III randomized placebo-controlled
study. Lancet. 2007;370:504510.
41. Burton BK, Grange DK, Milanowski A, et al. The response
of patients with phenylketonuria and elevated serum phenylalanine to treatment with oral sapropterin dihydrochloride
(6R-tetrahydrobiopterin): a phase II, multicentre, open-label,
screening study. J Inherit Metab Dis. 2007;30:700707.
42. Singh R, Jurecki E, Rohr F. Recommendations for personalized dietary adjustments based on patient response to
tetrahydrobiopterin (BH4) in phenylketonuria. Top Clin Nutr.
2008;23:149157.
43. Merinero B, Perez C, Perez-Cerda A, et al. Methylmalonic
acidemia: examination of genotype and biochemical data in
32 patients belonging to mut, cbIA or cbIB complementation
group. J Inherit Metab Dis. 2008;31:5566.
44. Shevell M, Matiaszuk N, Ledley F, Rosenblatt D. Varying
neurological phenotypes among mut0 and mut patients
with methylmalonyl CoA mutase. Am J Med Genet.
1993;45:619624.
245
246
23
Cardiac Disease
Anupama Chawla, MD, CNSP, DCH (UK) Janice Antino, RD, MS, CNSD, CSP, and Mindy Freudenberg, RD, MS, CNSD
CONTENTS
Congenital Heart Disease . . . . . . . . . . . . . . . . . . . . . . . . . 247
Malnutrition and Growth
Nutrition Assessment
Nutrition Management
Complications After Congenital Heart Disease Surgery
Learning Objectives
1. Identify factors contributing to growth failure in children with congenital heart disease.
2. Summarize the components of nutrition assessment
and optimal methods of nutrient delivery in children
with congenital heart disease.
3. Recognize and manage postoperative complications
and prescribe appropriate nutrition therapy.
Cyanotic
248
CARDIAC DISEASE
249
Nutrition Assessment
A thorough and accurate nutrition assessment is the primary
step for early recognition of feeding difficulties and growth
delay in children with congenital heart disease. This will result
in early intervention to help prevent nutrition deficiencies and
optimize growth. A complete nutrition assessment includes a
combination of methods that should include an accurate feeding history, visual clinical assessment, anthropometric evaluation, and biochemical indices (Table 23-3). Anthropometric
data such as weight, length/height, weight-for-length, and head
circumference can be evaluated using published growth charts
from the Centers for Disease Control and Prevention (CDC)
or the World Health Organization (WHO) (see Chapter 33).
Measurements should be plotted and monitored over time
to determine growth velocity and degree of growth failure.
Congenital heart disease may be present in conjunction with
an underlying chromosomal abnormality. In these condi
tions anthropometric data can be evaluated using specialized
growthcharts as available for children with trisomy 21, trisomy 18, Turners syndrome, and for infants born preterm.
Table 23-3 Components of Nutrition Assessment
in Congenital Heart Disease
Medical history
Type of lesion (cyanotic vs. acyanotic),
current medications, other medical
conditions
Feeding history
Type of formula, concentration of formula,
preparation methods, and amount
consumed; duration of feeds
Physical exam
Fluid status/edema, cyanosis, respiratory
rate (tachypnea)
Biochemical indices
Serum electrolytes, albumin, prealbumin.
Total lymphocyte count and stool for alpha1-antitrypsin if suspicious of protein-losing
enteropathy
Anthropometric data
Weight, length/height, weight-for-length,
triceps skinfolds, mid-arm circumference
Evaluation of growth
Monitor weight gain and linear growth
overtime
Gastrointestinal function
Evaluation of bowel pattern (eg, frequency
and consistency), GI reflux
2010 A.S.P.E.N. www.nutritioncare.org
250
Nutrition Management
Desired Concentration
(cal/oz)
24
27*
Starting
Concentration
20 cal/oz
(breast milk,
ready-to-feed
formula)
24 cal/oz
Volume
Desired
Concentration
Term Infant
Formula
Powder
3 oz
24 cal/oz
1 teaspoon
4 oz
27* cal/oz
1 teaspoon
Modular
Volume of
Formula
20 cal/oz
To Prepare
2425 cal/oz
Formula
(amount of
modular)
To Prepare
2728* cal/oz
Formula
(amount of
modular)
3 oz
1 teaspoon
1 teaspoons
3 oz
1.5 mL
2.5 mL
3 oz
2 teaspoons
3 teaspoons
*2728 cal/oz may not supply enough water for some infants. Hydration
status and renal solute load should be carefully monitored.
Infants and children who are unable to meet their nutrition needs via the enteral route should be considered for
parental nutrition (PN). PN can be initiated pre- or postoperatively with a therapeutic goal of restoring or maintaining
nutrition status and inducing somatic growth. The optimal
timing for initiating PN is dependent on the childs baseline
nutrition status and disease acuity. In view of the relatively
CARDIAC DISEASE
251
252
Formula
Pregestemil (Mead Johnson)
Alimentum (Abbott)
Portagen (Mead Johnson)
Monogen (Nutricia)
EleCare (Abbott)
Enfaport (Enfamil)
MCT:LCT ratio
55:45
33:67
87:13
90:10
33:67
84:16
Formula
Vivonex Pediatric (Nestle Nutrition)
Peptamin Junior (Nestle Nutrition)
Vital jr (Abbott)
Neocate One + (Nutricia)
Neocate Junior (Nutricia)
Pepdite Junior (Nutricia)
MCT:LCT ratio
69:31
60:40
50:50
35:65
35:65
35:65
Chylothorax
Chylothorax, a known complication of pediatric cardiac
surgery, requires special nutrition support considerations.
Chylothorax is the accumulation of chyle within the pleural
space. The chyle leak can be the result of injury to the
thoracic duct, disruption of accessory lymphatics, or from
an increased systemic venous pressure exceeding that in the
thoracic duct.2124 Studies have suggested that the increase
in postoperative chylothorax complications from 1% or less
in the 1970s and 1980s to 2.5% to 4.7% currently may be
due to the increased complexity of the surgeries performed
and possibly to the earlier initiation of enteral feeds. 21 Chan
reported an incidence of 3.8% from 2000 to 2002 with a
higher percentage occurring after heart transplant and the
Fontan procedure.
The challenge in managing chylothorax is in maintaining fluids and electrolytes while minimizing the
lymphatic leak. Chylothorax can be corrected surgically
but the results are not always favorable and not always
feasible for children who are possibly already compromised
after having had congenital heart surgery. Adverse affects
of chylothorax include immunosuppression, need for longterm chest tubes and intravenous access, and prolonged
hospitalization. 22 Postoperative length of stay is reported
to be significantly longer with a median of 22 days versus
2010 A.S.P.E.N. www.nutritioncare.org
8 days if a chylous leak develops. 23 Conservative management is usually attempted prior to surgery for the resolution
of the leak.
Conservative management includes pleural space
evacuation, the use of low-fat diets with MCTs, or total
parenteral nutrition for complete enteric rest. The use
of a MCT-enriched diet is based on the understanding
that MCTs are readily absorbed by the enterocytes into
circulation, providing adequate calories and minimizing
lymphatic flow to allow for healing. 23 For formula feedings, a high-MCT low-LCT formula may be used (Table
23-5). To prevent EFA deficiency, 2% to 4% of total calories
should be in the form of linoleic acid with 0.25% to 0.5%
from linolenic acid. 24 If patients are on oral feedings and
adequate calories can be consumed, a low-fat diet may be
sufficient. In the study by EH Chan 34 of 48 patients (71%)
had resolution with changes to their enteral diet. 23
Octreotide, a long-acting synthetic analogue of somatostatin, has been used as a treatment for chylothorax
drainage that did not respond to dietary manipulations
alone. In a study conducted between 19812004, 83% of
patients receiving octreotide responded with complete
resolution of their chylothorax after approximately 15
days of treatment and no side effects from the octreotide
therapy were noted after 2 weeks of treatment. 20 In a study
by EH Chan et al the patients had variable results. There
was no decrease in drainage over the treatment period
in 4 of 5 patients and it was thought that octreotide has a
better outcome with a low-flow leak versus higher drainage
patterns noted in patients in this study.
The early diagnosis and treatment of chylothorax can
reduce the length of the chylous leak. At present, dietary
management is the mainstay of treatment when managing
these patients conservatively.
CARDIAC DISEASE
Acceptable
Borderline
High
< 170
170199
> 200
< 110
110129
> 200
253
254
Summary
1. The potential for growth and nutrition recovery in children with congenital heart disease seems to be most
affected by:
A. Degree of growth impairment
B. Feeding difficulties
C. Energy intake/expenditure
D. Age and timing of corrective surgery
2. Infants with congenital heart disease may require
caloric intake of
to thrive.
A. 100 kcal/kg/d
B. 120 kcal/kg/d
C. 90 kcal/kg/d
D. 140200 kcal/kg/d
References
CARDIAC DISEASE
255
24
Renal Disease
Christina L. Nelms, MS, RD, CSP, CNSC, LD, Marisa Juarez, MPH, RD, LD, and Bradley A. Warady, MD
CONTENTS
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
Kidney Development and Function . . . . . . . . . . . . . . . . . . 257
Chronic Kidney Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . 258
Hemodialysis
Peritoneal Dialysis
Growth and Development
Nutrition Assessment
Nutrition Requirements
Fluid and Electrolyte Balance
Cardiovascular Disease and Lipid Management
256
Learning Objectives
Background
RENAL DISEASE
257
Table 24-1 NKF KDOQI Classification of the Stages of Chronic Kidney Disease8
Stage
Description
1
2
3
4
5
> 90
6089
3059
1529
< 15 (or dialysis)
258
7037
100.0
1454
1220
613
594
278
193
158
141
114
111
104
99
90
82
75
75
66
47
43
37
32
30
26
25
14
11
7
6
1110
182
20.7
17.3
8.7
8.4
4.0
2.7
2.2
2.0
1.6
1.6
1.5
1.4
1.3
1.2
1.1
1.1
0.9
0.7
0.6
0.5
0.5
0.4
0.4
0.4
0.2
0.2
0.1
0.1
15.8
2.6
Primary Diagnosis
Obstructive uropathy
A/hypo/dysplastic kidney
Focal segmental glomerulosclerosis
Reflux nephropathy
Polycystic disease
Prune Belly
Renal infarct
Hemolytic uremic syndrome
SLE nephritis
Familial nephritis
Cystinosis
Pyelo/interstitial nephritis
Medullary cystic disease
Chronic glomerulonephritis
Congenital nephrotic syndrome
Membranoproliferative glomerulonephritis Type I
Bergers (IgA) nephritis
Idiopathic crescentic glomerulonephritis
Henoch-Schnlein nephritis
Membranous nephropathy
Wilms tumor
Membranoproliferative glomerulonephritis Type II
Other systemic immunologic disease
Wegeners granulomatosis
Sickle cell nephropathy
Diabetic glomerulonephritis
Oxalosis
Drash syndrome
Other
Unknown
Hemodialysis
Hemodialysis (HD) is the use of a machine to dialyze soluble
substances and water from the blood by diffusion through a
semipermeable membrane, using a catheter placed centrally
or a fistula. It is often done for 3 to 5 hours, 3 or more times
per week for patients in CKD5 who cannot live without
regular dialysis.
Malnutrition is a significant complication of CKD and
a strong predictor for morbidity and mortality for adults
receiving maintenance HD.1315 Protein-energy malnutrition (PEM) produces profound effects on growth and
development and may be associated with increased risk of
hospitalization and mortality in children on HD.13,16,17
In addition to dry weight, length/height, weight-forlength, body mass index (BMI)-for-age, head circumference,
dietary intake, and serum albumin, the 2009 NKF KDOQI
nutrition guidelines now include recommendations on
monitoring normalized protein catabolic rate (nPCR) for
children on HD. The primary biochemical marker of nutrition status has been albumin. However, recent studies
indicate nPCR is superior to albumin as a marker of nutrition status in children on maintenance HD.16,18,19 These
studies show serum albumin to be a poor indicator of nutrition status. Research also demonstrates that intradialytic
parenteral nutrition (IDPN) significantly improves weight
gain and nPCR in malnourished patients on HD.16,18 A
nPCR of < 1 g/kg/d is a strong predictor of weight loss in
adolescent patients.19
The protein catabolic rate (PCR) is a measure of protein
intake. The nPCR is the PCR normalized to a function of
body weight, measured in grams of protein per kilogram
per day. nPCR is determined by first calculating the urea
generation rate (G):
RENAL DISEASE
Peritoneal Dialysis
Peritoneal dialysis (PD) is typically recommended for
infants, toddlers, and approximately 50% of adolescents
needing dialysis treatment. It is usually a nightly process. PD
involves infusion of a glucose-based solution through a catheter surgically inserted into the peritoneal cavity. Diffusion
allows for waste products to cross the peritoneal membrane.
Fluid is then drained from the peritoneal cavity and fresh
fluid is infused. In most children, this process occurs over
10 to 12 hours while they sleep. A daytime dwell is often
left in the cavity during the day and drained before nightly
dialysis is resumed. The peritoneal membrane transport
capacity can be determined by conducting the Peritoneal
Equilibration Test (PET). Patients may be classified as
high, high-average, low-average, or low transporters
depending on how rapid solute (eg, creatinine, glucose)
moves across the peritoneal membrane during a 4-hour test
exchange. High transporters tend to have more porous peritoneal membranes and thus rapidly remove waste products
such as creatinine, but also tend to lose significant amounts
of potassium and protein across the peritoneum. The rapid
absorption of glucose from the dialysate decreases the
osmotic gradient and results in less fluid removal from these
same patients. In contrast, low transporters tend to remove
less kidney waste, but also lose less protein and potassium
and remove fluid well. The nutrition prescription for the
peritoneal dialysis patient is, in turn, often influenced by
what kind of transporter the PD patient is.
259
260
Reprinted with permission from: Mahan JD, Warady BA. Assessment and treatment of short stature in pediatric patients with chronic kidney disease: a
consensus statement. Pediatr Nephrol. 2006;21:917930.
2010 A.S.P.E.N. www.nutritioncare.org
RENAL DISEASE
261
Nutrition Assessment
Malnutrition is a serious complication of CKD, especially
in CKD5D. There is no single measurement to adequately
define nutrition status in CKD.25,16 It is challenging to assess
this population due to the metabolic and growth complexities that are present. Early nutritional intervention may be
2010 A.S.P.E.N. www.nutritioncare.org
262
RENAL DISEASE
Nutrition Requirements
Macronutrients
Energy
263
Micronutrients
Children with CKD are at risk for micronutrient deficiencies due to poor intake, poor absorption, abnormal renal
metabolism, medication interactions, and potential dialysis
losses. Adequate intake of fat-soluble and water-soluble
vitamins, zinc, and copper should be encouraged. There is
risk to growth and overall health if these micronutrients are
deficient. Supplementation of these vitamins and minerals
is necessary if dietary intake is low or if there is clinical
evidence of a deficiency and/or low blood levels. Because
excess losses of water-soluble vitamins are possible in all
dialysis patients, all children with stage 5D CKD should
take a water-soluble vitamin supplement.12
Fat-Soluble Vitamins
264
RENAL DISEASE
265
Minerals
Sodium
Sodium is often restricted to help control volume overload and blood pressure. According to the 2005 Dietary
Guidelines for Americans older than 2 years, all individuals with hypertension should limit sodium intake to
<1500 mg/d.62 This is complicated by the environmental
cues and peer pressures that promote high sodium intake,
especially where fast food is concerned. Stringent sodium
restrictions are challenging. A more reasonable sodium
restriction of 2000 to 3000 mg/d may be better accepted
and hence adhered to in older children or adolescents. The
amount of sodium restriction needed should be based on
individual patient parameters such as blood pressure, fluid
gains, and nutrition intake. Most sodium in the diet comes
from processed foods. Therefore, an increased intake of
fresh foods versus processed or canned foods will decrease
dietary sodium intake. Using natural herbs and spices
to season foods versus table salt is extremely helpful in
reducing sodium content in foods. It is not only important
to educate patients on low-sodium foods, but also on how
to read nutrition facts labels. According to the U.S. Department of Agriculture (USDA), foods with < 5 mg sodium per
serving are considered sodium- or salt-free. Foods with < 35
mg sodium per serving are considered very low sodium; and
foods with <140 mg sodium per serving are low sodium.62
The use of salt substitutes is often contraindicated in CKD
patients because potassium chloride is typically substituted
for sodium chloride. Potassium chloride can cause hyperkalemia in those at risk for the condition. Some PD patients
may lose large amounts of potassium across their peritoneal membrane and may actually benefit from additional
potassium.
Infants and children with CKD often have primary
disorders such as posterior urethral valves that cause
polyuria and salt wasting. These children must have supplemental sodium and free water to maintain proper balances.
Potassium
266
levels should be maintained within age-appropriate reference ranges for CKD stages 1 to 4; and between 4 to 6 mg/
dL for ages 1 to 2 years and 3.5 to 5.5 mg/dL for adolescents
for CKD stage 5 and 5D. Hypophosphatemia that arises
due to phosphate wasting disorders, overcorrection, or
other causes should be corrected by liberalization of diet or
medication changes. Hypophosphatemia is associated with
increased morbidity or mortality and poor growth.
The KDOQI pediatric bone guidelines also suggest
that when PTH levels are elevated for the given stage of
CKD, dietary phosphorus should be limited to the DRI for
age. When phosphorus values and PTH values exceed reference ranges for age and stage of CKD, phosphorus should
be limited to 80% of the DRI.12,66 However, this guideline
can equate to low intake in children younger than age 8. It
should be noted that less than 500 mg of phosphorus, even
in young children, may not allow for adequate caloric intake.
An exception are children who get a controlled amount of
phosphorus via a set amount of enteral formula by mouth or
feeding tube.
Limiting phosphorus in the pediatric diet may be a challenge, especially as fast food and convenience food increases
in the usual diet of children and adolescents. About 60% of
dietary phosphorus is absorbed from the typical naturalfood mixed diet. Assuming natural foods are consumed, the
average adult man consumes an average of 1550 mg of phosphorus per day, with over half consuming more than 1600
mg daily. The average woman consumes about 1000 mg
daily. Foods high in protein typically contribute the most
phosphorus in a natural diet with dairy and meats, including
fish, providing 20% to 30% each of the usual daily intake.
These numbers are increasing as more instant and restructured foods as well as colas, which have phosphate additives,
are on the market. Foods made with phosphate additives,
including many instant and restructured foods, have almost
100% absorption of phosphate content. Estimates are that
these foods could contribute to dietary phosphorus intake by
about 1 gm daily, even with unchanged protein and calcium
intakes.67 Additionally, Sullivan et al looked at chicken
products with phosphate-containing additives and found
that the phosphorus content of these products was higher
in every instance than the phosphorus content expected
from a nutrient database, averaging 84 mg/100 g of product
or greater. This study concludes that standard nutrition
databases do not currently account for the recent influx of
phosphate additives in foods. This, coupled with the great
variation of phosphorus content between products, makes
it difficult to estimate phosphorus content of foods and to
advise patients who need to limit dietary phosphorus. The
RENAL DISEASE
267
268
Renal Transplant
RENAL DISEASE
269
270
refer to the section on Kidney Development and Function.) Although kidney function is usually restored once
the etiology of AKI is eliminated or corrected, supportive
therapy is required in the interim. Therapy may or may not
include temporary dialysis. If dialysis is required, there is no
standard treatment modality. PD, HD, and continuous renal
replacement therapy (CRRT) are all used. CRRT is chosen
by an increasing number of pediatric centers because of the
safety and efficacy of the technique, even in those patients
who are experiencing hemodynamic instability.87,88
Nutrition assessment and planning for the patient with
AKI typically follow the same guidelines for CKD5D and
critical illness. There are no set standards for estimating
caloric and protein needs in the setting of AKI, either
with or without the use of dialysis. Needs are based on
the age-related needs of the patients, in addition to modifications based on comorbid medical conditions such as
sepsis. Hypercatabolism and alterations in metabolism are
common in AKI. Some of the alterations of metabolism
include decreased protein synthesis and inefficient use
of proteins by the cells, altered amino acid pools, hyper
glycemia secondary to insulin resistance, lipid alterations
caused by impaired lipolysis, acidosis, and electrolyte imbalances. The primary goal of nutrition therapy in patients with
AKI is to prevent catabolism as much as possible.
The patient with AKI and not on dialysis may need
more rigid electrolyte and fluid restrictions. If a nutritional supplement is required, use a renal supplement,
such as Suplena or Nepro, that is nutrient dense and has
a low renal solute load. For infants, Similac PM 60/40 is
usually the most appropriate choice. Once renal function is
restored, a regular diet and/or supplement is appropriate.
However, when dialysis is performed, depending on the
modality, restrictions may vary (see sections on Continuous
Renal Replacement Therapy, Hemodialysis, and Peritoneal
Dialysis). Nutrition guidelines in AKI when patients are
provided HD or PD are similar to those for CKD and HD
and PD. However, fluid and electrolyte concerns as well as
prevention of catabolism take priority to more long-term
concerns associated with CKD, such as CVD, growth, and
renal osteodystrophy.
RENAL DISEASE
Neonatal Issues
AKI is common in the neonatal intensive care unit and
may be of primary origin, such as congenital renal disease,
or secondary to conditions such as sepsis, drug toxicity,
obstruction, hypoxia, or respiratory distress. Twenty
percent of new dialysis cases are reported to be newborns.97
Mortality is high (46%) in neonates and low-birth-weight
infants with AKI.98 Dialysis, including PD, CRRT, or less
commonly HD, may be used to maintain fluid, acid-base,
and electrolyte balance as well as remove toxins in the short
or long term. It is important to remember that serum laboratory values, such as phosphorus and potassium, may have
higher normal limits for neonates than for older infants and
children. Fluid balance is important because patients may
have high urine output due to sodium and fluid-wasting
renal disorders, stomas, emesis, or suction. This may necessitate a high fluid intake, replacing losses and providing
maintenance needs. Poor urine output or additional
sources of fluids, such as medication drips, may lead to fluid
restrictions, and the need to concentrate formula with additives or to use parenteral nutrition (PN).99 Oliguric and
anuric infants typically should receive 25 to 30 mL/kg/d,
with infants < 26 weeks gestational age possibly needing
more.100
Controlling HTN and edema are often critical in this
population. Maintenance fluid needs are a good starting
point, with adjustment based on clinical conditions.101 Often
children with high fluid and sodium losses require sodium
supplementation of 1 to 3 mEq/kg/d.100 Correct acidosis
with supplementation of sodium bicarbonate.97 Sodium
bicarbonate supplementation of 1 to 2 mEq/kg/d may be
needed to prevent hyperkalemia. If serum potassium levels
are high, limit potassium to 1 to 2 mEq/kg/d.100
It is important to be aware of medications that may
affect nutrition. Pressors or narcotics may decrease gastric
motility and may affect tolerance to enteral feeding. Continuous jejunal or transpyloric feedings may be better tolerated
than nasogastric feedings. Anti-hypertensives can increase
271
272
occur include high blood pressure and proteinuria associated with a low GFR. Height and weight gains tend to be
impaired in these patients and mandate close monitoring/
supervision of their nutrition status. Growth goals are the
same as other neonates. Further discussion of the neonate is
found in the chapter on nutrition, growth, and development
(Chapter 13).
Enteral Nutrition
RENAL DISEASE
273
274
Parenteral Nutrition
Nephrotic Syndrome
Nephrolithiasis
RENAL DISEASE
275
Limit Acid-Based
Foods
Limit Meats
and Protein
Limit
Oxalate
Limit
Sodium
DRI Calcium
Intake*
X
X
X
X
Calcium-based stones
Oxalosis or hyperoxaluria
with calcium-based stones
Uric acid stones
X
X
X
X
X
X
X
X
Cystine stones
Struvite stones
Other kidney stones
X
X
X
DRI, especially
limit purines
X
*Although DRI calcium intake is appropriate general medical management for general pediatric health, including patients with a variety of kidney stone
disorders, avoidance of excessive or inadequate intakes of calcium are especially important in the types of kidney stones notated.
276
Oxalate
Primary hyperoxaluria is rare, and will be discussed further
below. However, secondary hyperoxaluria may result from
fat malabsorption or idiopathic increased absorption of
oxalate. Oxalate intake may need to be restricted in hyperoxaluric stone formers.119 Because hyperoxaluria is rare,
reported at only 6% of stone formers in one pediatric study,
it may be unnecessary to restrict oxalate to prevent stones in
hypercalciuria alone or in other types of kidney stones.117 In
fact, limiting oxalate in hypercalcuric patients is not shown
to reduce stone formation in a study of adult patients.120
Cystinuria
Cystinuria is an autosomal-recessive disorder and is the
cause of about 10% of kidney stones in children. The disorder
is related to impaired transport of the amino acids cystine,
ornithine, lysine, and arginine. Of these, cystine is insoluble
in the urine and thus it can cause stone formation. Recurrent
stone formation is common without medical management,
but even with medical management adherence may be poor
due to side effects and lack of treatment efficacy. Cystinuria
often leads to renal insufficiency, including ESRD, due to
recurrent stone formation and frequent intervention. Male
patients tend to be more severely affected. Medications
are often used in treatment; however, dietary interventions may be beneficial as well. As in other types of stones,
high fluid intake, low-sodium diet, and foods high in alkali,
such as fruits and vegetables, are recommended. Limiting
acid load by limiting excess protein intake is appropriate. A
2g sodium diet has been shown to reduce urinary cystine
concentration. A fluid intake of 3 L/d is recommended in
2010 A.S.P.E.N. www.nutritioncare.org
RENAL DISEASE
277
Bartters Syndrome
ARF
No dialysis
ARF
PD or HD
ARF
CRRT
CKD
(Stage 3-5)
CKD5
HD
CKD5
PD
Energy*
DRI
DRIdo not
supplement
to replace
urinary losses
Protein
DRI + 0.1**
g/kg IBW
DRI + 0.150.3** g/
kg IBW
(dependent on
age)
Sodium
13 mEq/kg
will vary
according to
edema or HTN
Will vary.
Consult with
renal/primary
team to
determine.
Will vary.
Consult with
renal/primary
team to
determine.
Typically no
restriction; may
need electrolyte
supplementation
Potassium
Restriction not
needed
Tightly limit
Limit
Stage 3:
100%140% x
DRI/kg IBW
Stage 4-5:
100%120% x
DRI/kg IBW
13 mEq/
kg will vary
according to
edema or HTN
unless sodium
wasting
Most will
tolerate > 3
mEq/kg/d
Phosphorus
Restriction not
needed
Will vary
according to
UOP. Consult
with renal
team to
determine.
DRI
Tightly limit
Limit
13 mEq/
kg will vary
according to
edema or HTN
unless sodium
wasting
Generally
unrestricted
unless low
transporter.
Will need to be
monitored.
Limit to 80%100% x DRI to keep serum levels WNL.
Will vary
according to
UOP. Consult
with renal
team to
determine.
Tightly limit
fat-soluble
vitamins
Will vary
according to
UOP. Consult
with renal
team to
determine.
Limit fatsoluble
vitamins
May need
additional
replacement
fluids.
Generally
unrestricted
Replace UOP,
insensible
losses, + UF
May need
supplementation
especially
selenium and
thiamin.
100% DRI.
Supplement
water soluble if
needed.
Fluids
Micronutrients
*DRI typically
appropriate unless
specific notation.
13 mEq/
kg will vary
according to
edema or HTN
unless sodium
wasting
13 mEq/kg
but will vary
according to
serum levels
and age
Replace UOP,
insensible
losses, + ~1
L/d
*Energy requirements may need to be adjusted for physical activity level and/or based on rate of weight gain or loss.
**Protein requirements may need to be adjusted according to dialytic protein and amino acid losses.
2010 A.S.P.E.N. www.nutritioncare.org
278
Other
There are many other renal disorders, including phosphate
metabolism disorders, cystinosis, Fanconis syndrome,
Liddle syndrome, Gordon syndrome, and others that are too
numerous to discuss in this context. Other disorders may
be primary renal disorders or renal disease secondary to a
systemic disease. In either case, the nutrition needs of these
patients should routinely be assessed because of the impact
that they have on the growth and development of these
2010 A.S.P.E.N. www.nutritioncare.org
References
RENAL DISEASE
279
24. Mendley SR, Majkowski NL. Urea and nitrogen excretion in pediatric peritoneal dialysis patients. Kidney Int.
2000;58:25642570.
25. Rees L, Shaw V. Nutrition in children with CRF and on dialysis. Pediatr Nephrol. 2007;22:16891702.
26. Mahan JD, Warady BA. Assessment and treatment of short
stature in pediatric patients with chronic kidney disease: a
consensus statement. Pediatr Nephrol. 2006;21:917930.
27. Tom A, McCauley L, Bell L, et al. Growth during maintenance
hemodialysis: impact of enhanced nutrition and clearance. J
Pediatr. 1999;134:464471.
28. Parekh RS, Flynn JT, Smoyer WE et al. Improved growth
in young children with severe chronic renal insufficiency
who use specified nutritional therapy. J Am Soc Nephrol.
2001;12:24182426.
29. Tonshoff B, Kiepe D, Ciarmatori S. Growth hormone/
insulin-like growth factor system in children with chronic
renal failure. Pediatr Nephrol. 2005;20:279289.
30. Vimalachandra D, Hodson EM, Willis NS, Craig JC, Cowell C,
Knight JF. Growth hormone for children with chronic kidney
disease. Cochrane Database Syst Rev. 2006;3:CD003264.
pub003262.
31. Greenbaum, LA, Hidalgo G, Chand D, et al. Obstacles to
the prescribing of growth hormone in children with chronic
kidney disease. Pediatr Nephrol. 2008;23:15311535.
32. Tom A, McCauley L, Bell L, et al. Growth during maintenance
hemodialysis: impact of enhanced nutrition and clearance. J
Pediatr. 1999;134:464471.
33. Fischbach M, Terzic J, Menouer S, et al. Intensified and daily
hemodialysis in children might improve statural growth.
Pediatr Nephrol. 2006;21:17461752.
34. Baumgartner RN, Roche AF, Himes JH. Incremental growth
tables: supplementary to previously published charts. Am J
Clin Nutr. 1986;43:711722.
35. Foster BJ, Leonard MB. Measuring nutritional status in
children with chronic kidney disease. Am J Clin Nutr.
2004;80:801814.
36. Wong CS, Gipson DS, Gillen DL, et al. Anthropometric
measures and risk of death in children with end-stage renal
disease. Am J Kidney Dis. 2000;36:811819.
37. Wong CS, Hingorani S, Gillen DL, et al. Hypoalbuminemia
and risk of death in pediatric patients with end-stage renal
disease. Kidney Int. 2001;61:630637.
38. Kavey RW, Allada V, Daniels SR, et al. Cardiovascular Risk
Reduction in High-Risk Pediatric Patients: A Scientific Statement from the American Heart Association Expert Panel on
Population and Prevention Science; the Councils on Cardiovascular Disease in the Young, Epidemiology and Prevention,
Nutrition, Physical Activity and Metabolism, High Blood
Pressure Research, Cardiovascular Nursing, and the Kidney
in Heart Outcomes Research: Endorsed by the American
Academy of Pediatrics. Circulation. 2006;114:27102738.
39. American Academy of Pediatrics Endorsed Policy Statement.
Cardiovascular risk reduction in high-risk pediatric populations. J Pediatr. 2007;119:618621.
280
RENAL DISEASE
281
282
Gastrointestinal Disease
25
CONTENTS
Learning Objectives
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283
Mechanisms of Nutrition Deficiency
Nutrition Assessment
Gastroesophageal Reflux. . . . . . . . . . . . . . . . . . . . . . . . . .
Celiac Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Inflammatory Bowel Disease. . . . . . . . . . . . . . . . . . . . . . .
Diarrhea . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Pancreatic Insufficiency . . . . . . . . . . . . . . . . . . . . . . . . . .
Pancreatitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Disorders of Chyle Loss. . . . . . . . . . . . . . . . . . . . . . . . . . .
Functional Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
286
287
289
291
292
293
294
295
Gastroparesis
Constipation
Cyclic Vomiting Syndrome
Irritable Bowel Syndrome
Introduction
284
Examples
Disordered Ingestion
Anorexia
Dental disease
Dysphagia of any cause
Esophagitis
Foreign body
Inadequate access to food
Pancreatic enzyme deficiency
Sucrase-isomaltase deficiency
Cholestasis with failure of bile salt secretion
Short bowel syndrome due to surgery
Celiac disease
Liver disease
Movement disorders
Fever
Failure of Digestion
Failure of Absorption
Increased Needs
Nutrition Assessment
Nutrition assessment is an essential component of the
evaluation of all children. Monitoring of growth should be
part of the routine well-child examination. Assessment of
both linear and ponderal growth of the child or adolescent
is central to evaluation of nutrition status. Alterations in
patterns of linear growth and weight gain (both inadequate
as well as excessive weight gain) may be the earliest manifestation of disease. Malnutrition is commonly diagnosed
in hospitalized patients and is also a common comorbidity
in patients with GI disease.13 Malnutrition during illness
may complicate the response to therapies or impair recovery
(Table 25-2). Thus, nutrition assessment is an integral part
of both the initial and ongoing evaluation of all children
with acute and/or chronic disease and is of crucial importance in the evaluation of the child with GI disease.
The initial nutrition evaluation includes both subjective and objective assessment of the patients current
nutrition status and projected nutrition requirements. The
subjective assessment includes the presence and duration
of GI symptoms, fever, frequent infections, fatigue, food
aversion, allergies to particular foods, or feeding intolerance. Specific attention is paid to previous growth, detailed
diet history, changes in body weight and dietary intake, GI
symptoms (eg, abdominal pain, diarrhea, and vomiting),
and anorexia. The objective assessment should include
data from clinical, anthropometric, and laboratory evaluations (Table 25-3). Clinical data include the diagnosis,
current medical or surgical problems, allergies, and medications that may affect nutrition support options. Objective
measures of nutrition status include growth indices (both
previous and current), current weight, and Tanner stage.
GASTROINTESTINAL DISEASE
Signs/Symptoms of
Deficiency
Iron
Fatigue
Microcytic anemia
Folate
Megaloblastic anemia
Glossitis
Diarrhea
Forgetfulness
Megaloblastic anemia
Ataxia
Diarrhea
Mental status changes
Paresthesias
Glossitis
Osteopenia
Osteoporosis
Tetany
Bone pain
Muscle weakness
Tetany
Osteomalacia
Rickets
Night blindness
Decreased appetite
Decreased immune function
Hyperkeratosis
Bleeding
Bruising
Hemolytic anemia
Truncal ataxia
Hypo or areflexia
Vitamin B12
Calcium
Vitamin D
Vitamin A
Vitamin K
Vitamin E
Zinc
Magnesium
Diarrhea
Dry skin
Skin sloughing on palms
Mental status changes
Hair loss
Growth stunting
Anorexia
Muscle cramps
Bone pain
Nausea
Seizures
Laboratory Markers
hgb
hct
MCV
RDW
% TIBC
ferritin
serum iron
serum folate
red blood cell folate
MCV
MCV
hgb
serum vitamin B12
plasma vitamin A
PTT
serum creatinine
creatinuria
serum vitamin E:total
serum lipid ratio
serum alk phos
serum zinc*
urinary zinc**
serum magnesium
* Zinc binds to serum proteins, which can make levels appear low if
protein levels are depleted.
** Associated with disease status.
285
286
Gastroesophageal Reflux
40%
30%
15%
12%
15%
Examples
Infant
Regurgitation
Vomiting
Feeding difficulties (acute)
Unexplained crying
Failure to thrive
Posturing
Vomiting
Cough
Abdominal pain
Sore throat
Respiratory difficulties
Hoarseness
Regurgitation
Chest pain
Heartburn
Epigastric pain
Dysphagia
Child
Adolescent
GASTROINTESTINAL DISEASE
Diarrhea
Constipation
Abdominal pain
Nausea/vomiting
Anorexia
Anemia
Weight gain/loss
Hepatotoxicity
Proton pump
inhibitor
H2 Histamine
receptor antagonist
X
X
X
X
X
X
X
X
X
X
X
287
Celiac Disease
288
GASTROINTESTINAL DISEASE
Gluten Free
Barley
Bulgur
Couscous
Dinkle (spelt)
Durum
Einkorn
Emmer
Farina
Fu
Graham flour
Kamut
Seitan
Semolina
Rye
Wheat
Triticale
Amaranth
Arrowroot
Buckwheat
Corn
Flaxseed
Rice
Millet
Milo
Potato flour
Quinoa
Sorghum
Soy
Tapioca
Tef
Taro flour
Urd
Removal of gluten from the diet usually results in clinical and histological recovery. Specialized oral feedings or
enteral feedings are rarely necessary in a patient with celiac
disease. Parenteral feedings are not used unless there is
some other indication for that therapy.
When the diagnosis of celiac disease is confirmed,
routine nutrition follow-up of the child is necessary to
monitor growth parameters, promote adherence to the
diet, and to assure proper nutrition, particularly because
many of the gluten-free products are not fortified. Ongoing
monitoring of vitamins (especially A, D, B12 , and folate), as
well as assessment of anemia and markers of bone health, is
suggested.40 Fortunately, with proper nutrition and adherence to the GF diet, bone density in children returns to
normal 1 year post-initiation of the diet.41
Clinical outcome is measured by resumption of normal
growth and weight gain and by monitoring of serologic
markers, especially tTG and IgA. Failure of tTG IgG and
IgA levels to return to normal values after adherence to a
gluten-free diet for 6 to 12 months warrants further inspection of the childs diet for inadvertent gluten consumption.
Inflammatory bowel disease (IBD) refers to chronic inflammation anywhere along the GI tract. The term includes
Crohns disease, ulcerative colitis, and indeterminate colitis
(ie, IBD with features that do not allow clear distinction
between Crohns disease and ulcerative colitis). These are
chronic conditions that affect children of any age although
they most commonly manifest in the second decade of life.
289
The most important risk factor for developing IBD is a positive family history.
The incidence of Crohns disease seems to be increasing
in childhood while the rates of ulcerative colitis are steady.
IBD may present at any age. Ulcerative colitis is more
common in younger children. Peak age of incidence of
Crohns disease is in the second decade of life.
Inflammation of the bowel leads to a number of
derangements that culminate in diarrhea, GI bleeding, and
abdominal pain. Other symptoms depend on the location
of inflammation within the GI tract. For example, vomiting
is more prominent in patients with gastric or small bowel
disease. The most common presentation is a patient with
abdominal pain and diarrhea. Stools may be bloody. Weight
loss is common, especially in patients with Crohns disease.
However, non-specific manifestations of the disease may
be the initial manifestations and failure to recognize their
importance may lead to delay in the diagnosis. Joint pain and
swelling, skin rashes, muscle pain, elevated liver enzymes,
or eye changes (uveitis, iritis, or episcleritis) may be present
prior to any specific GI manifestation. Oral ulcerations can
range from painless to severe pain with bleeding. It is well
recognized that deterioration of linear and ponderal growth
may precede the development of more specific symptoms
by months or in some cases even years.
The effect of IBD on nutrition status is multifaceted.
Growth, bone health, and macronutrient and micronutrient
stores are commonly affected and malnutrition is common.42
Weight loss is a common presenting symptom in all forms
of IBD. Linear growth failure can occur as well but is more
frequently associated with Crohns disease.
Nutrition assessment of children with IBD includes
measurements of weight, height, and calculation of body
mass index (BMI). These values should be plotted and
followed serially on appropriate Centers for Disease Control
and Prevention (CDC) growth charts.43 The importance
of tracking these measurements is emphasized by the fact
that upon diagnosis the majority of children with ulcerative colitis and especially Crohns disease will have growth
failure.44 In fact, reduced linear growth velocity may precede
GI manifestations of IBD by months or even years.4547
Criteria for defining growth failure include a height velocity
< 3rd percentile, a height < 3rd percentile, or a bone age less
than chronological age by 2 or more standard deviations.48
Males may be more susceptible to growth failure, but all
children with IBD are at risk.43,49 Factors affecting risk
include malnutrition, treatment modality, and intestinal
inflammation.
Malnutrition in IBD patients is multifactorial and
2010 A.S.P.E.N. www.nutritioncare.org
290
includes energy losses, increased requirements, malabsorption, and decreased energy intake due to diarrhea,
abdominal pain, and other disease-related side effects. 50
Further contributing to malnutrition is poor appetite
related cytokine activity in the inflammatory process. 51,52
Inflammatory cytokines have also been shown to reduce
insulin-like growth factor 1 (IGF-1). 5355 Children with
active Crohns disease often have lower levels of serum
IGF-1 levels than controls. In addition, chronic usage of
corticosteroids as treatment can suppress IGF-1 and also
decrease osteoblast activity.43,56 Limited data suggest that
nutrition therapy in comparison to corticosteroid usage
may be beneficial in the treatment of Crohns disease and
spare linear growth.48 More research is needed to substantiate these findings.48
Assessment of nutrition status includes sufficiency of
vitamin and mineral stores. Analyses of dietary records
and/or 24-hour food recall as well as laboratory values are
helpful in assessing micronutrient status. Commonly deficient nutrients include iron, folate, vitamin B12 , vitamin
D, calcium, zinc, and magnesium.43,57 Refer to Table 25-2
for information regarding deficiency signs/symptoms and
laboratory markers. Reduced iron stores are associated with
decreased intake, reduced absorption, and increased losses
(ie, blood loss). Often, iron supplementation is required
along with a diet rich in iron and vitamin C to correct
the deficiency. 57 Laboratory values to monitor iron status
include hemoglobin, reticulocyte count, mean corpuscular
volume red blood cell (RBC) distribution width, ferritin,
transferrin saturation, and iron. 50
Folate stores may be affected by insufficient intake,
because many good sources of folate (eg, leafy green
vegetables) are often not tolerated by the child with active
inflammation. In addition, medications used in disease
treatment such as methotrexate and sulphasalazine have
direct and deleterious effects on folate metabolism. Children on these medications need folate supplementation.43,57
Vitamin B12 deficiency may occur in children with
involvement of the stomach and terminal ileum and in
cases of bacterial overgrowth.44,57 Notably, deficiency may
be masked by supplementation of folate. Deficiency is
corrected by intramuscular injection, oral supplementation,
or nasalgel. 57
Maintenance of adequate calcium and vitamin D stores
is imperative in children with IBD, particularly in those
receiving steroid therapy due to the relationship of steroid
usage and decreased bone mineral density. Lactose intolerance often limits consumption of milk and dairy products,
rich in these nutrients. 57 Supplementation may be required
2010 A.S.P.E.N. www.nutritioncare.org
GASTROINTESTINAL DISEASE
Diarrhea
Osmotic diarrhea happens when a non-absorbed material, often carbohydrate, creates an osmotic load in the distal
291
292
Pancreatic Insufficiency
GASTROINTESTINAL DISEASE
293
Pancreatitis
294
a short period. Re-initiation of feedings is based on resolution of abdominal pain and ileus and vomiting. Most patients
with mild disease recover quickly without complications.
Patients with severe pancreatitis often show signs of
hemodynamic instability and are at risk for multiple complications including severe nutrition depletion. Nutrition
support is important in these patients and is considered an
active therapeutic intervention.95 Patients can be fed enterally by nasogastric (NG) or nasojejunal (NJ) tube or by the
intravenous route. Although there are a number of studies
in adults, data concerning nutrition support in pediatric
patients with pancreatitis are scarce. Adult studies suggest
a trend to fewer adverse outcomes in patients who receive
enteral as opposed to parenteral feedings, however the
effect on outcome is not clear.96 There does not seem to be a
difference in feedings of polymeric versus semi-elemental or
immune-enhancing formulas.97 A study in children with
severe acute pancreatitis found little difference between EN
and PN in length of stay, infection, mortality, or need for
surgery.98
Chronic pancreatitis is a condition where continued
inflammation of the pancreas produces irreversible changes
in the gland. In some cases it is related to continued recurrent injury from acute attacks. Often the etiology is not
clear even when risk factors are present. In adults, repeated
exposure to toxins (eg, alcohol) is often implicated. In children, hereditary factors such as mutation of the trypsinogen
molecule, or mutations of the cystic fibrosis transmembrane
regulator (CFTR) gene or trypsin inhibitor genes are often
sought. Autoimmune disorders, both isolated autoimmune
pancreatitis as well as systemic autoimmune diseases, are
seen. Patients with recurrent acute pancreatitis are at risk
for developing chronic pancreatitis.99
Treatment of chronic pancreatitis revolves around
several concerns: chronic pain, development of diabetes
due to islet cell destruction, and pancreatic insufficiency
due to acinar cell destruction. Pain impacts ability to ingest
nutrients, and the treatment of chronic pain is beyond the
scope of this chapter. In addition the treatment of diabetes
mellitus is discussed elsewhere (Chapter 21). Digestive
enzyme insufficiency is a late complication of chronic
pancreatitis. The treatment for pancreatic digestive enzyme
insufficiency is enzyme replacement. The goal is to provide
enzyme supplements enough to restore digestive function.
The dose of pancreatic enzyme is calculated according to
the lipase content. A usual dose of enzyme is 1000 to 2500
units of lipase per kilogram of body weight per meal. This is
often altered based on the estimated fat content of the meal.
This is discussed in more detail in the chapter on cystic
2010 A.S.P.E.N. www.nutritioncare.org
Chyle is a creamy fluid consisting of fat, protein, electrolytes, and lymphocytes and is an important aspect of the
metabolism of fat. Triglycerides are broken down in the
intestinal lumen to fatty acids and mono-acyl glycerols.
They are absorbed into the intestinal epithelial cells. This
process is aided by the action of bile salts mixing with the
fatty acids forming micelles that enhance the transport of
the molecules. Within the enterocytes, the absorbed fatty
acids are re-esterified to glycerol and the resultant lipid
is complexed with proteins (forming chylomicrons) and
transported through the lymphatic system of the GI tract
and abdomen ultimately into the thoracic duct and then
into the blood stream. Disorders of chyle loss are rare with
the most common disorders being chylothorax or chylous
ascites. Chylothorax is defined by pooling of lymphatic
fluid, chyle, in mediastinal or pleural cavities. Chylous
ascites occurs when there is lymphatic disruption in the
abdominal cavity with resultant pooling of chyle in the
abdomen. This condition is usually seen as a complication
of surgery100 although congenital defects of lymph flow are
also described. For more detailed information refer to the
chapter on cardiac disease (Chapter 23). A less common
disorder of chyle is that of intestinal lymphangiectasia. This
condition results from failure of lymph flow often related
to inflammation, causing enlargement of intestinal lymph
vessels, which causes breakage of the lacteals and spillage
of chyle into the intestinal lumen.101 As a result protein
losing enteropathy, hypoalbuminemia, and edema may
result. Hypogammaglobulinemia is often present. Edema
of the intestinal mucosa causes malabsorption. Steatorrhea is typically seen.102 Even more rare are the disorders
of chylomicron formation such as abetalipoproteinemia in
which there is a defect in the ability to complex the lipids
with carrier proteins and therefore the lipid is not carried
into the lymphatic system.
Regardless of the specific disorder of chyle processing,
the nutrition approach is similar. The initial treatment is
restriction of dietary fat. A low-fat, high-protein diet is
recommended because there is usually associated protein
loss in the GI tract. Because medium-chain length fatty
acids and triglycerides can be metabolized without entry
into the lymphatic system, dietary fats should be primarily
GASTROINTESTINAL DISEASE
Functional Disorders
295
Gastroparesis
Gastroparesis is the delay of emptying of the stomach in
the absence of a mechanical obstruction. There are multiple
causes (Table 25-12). Disorders of the intestinal musculature (myopathic) and the intestinal nervous system
(neuropathic), both congenital and acquired, are described.
Common causes are immaturity (especially in premature
infants), viral infections, systemic diseases, and drugs.
Gastroparesis often complicates the management of type 1
diabetes mellitus. Evaluation should include assessment of
gastric anatomy and function (eg, upper GI series x-ray or
gastric emptying study) but also the search for and treatment of any underlying condition and bacterial overgrowth.
Importantly, malnutrition may be both a cause as well as a
result of gastroparesis.
Table 25-12 Conditions Associated with Gastroparesis
Infection
Postviral illness (eg, rotovirus)
Neurological Disease
Mitochondrial disorders
Familial dysautonomia
Systemic Disease
Diabetes
Malnutrition
Connective tissue disorders
296
Constipation
Constipation is a common symptom among humans of all
ages and is often especially troubling in infants and young
children. It is most often both self-limited and short lived.
However, a substantial number of patients have symptoms
that persist for 6 months or more. Constipation may be
related to inadequate intake of fluid or fiber, side effects of
medication, inactivity, or disordered bowel motility. Most
cases are idiopathic and fulfill the definitions of functional
constipation. Although most recognize a role for diet in
both the etiology and the treatment of constipation, there
are little data to allow identification of specific foods as
either causal or beneficial.
The role of cows milk in constipation has been recognized for some time, although the mechanism is unclear.109
There is increasing evidence to suggest a role of cows milk
protein sensitivity in constipation.110,111 In addition, it is
known that the fat content of milk may also be associated
with harder or more difficult-to-pass stools.
Historically, a high-fiber diet has been recommended
for children with constipation. However, few studies document benefit.112,113 There is also no conclusive evidence that
fiber supplements or a lactose-free diet is beneficial in alleviating recurrent abdominal pain in children.114 However,
2010 A.S.P.E.N. www.nutritioncare.org
GASTROINTESTINAL DISEASE
PRESENTATION CHARACTERISTICS
Mucosal
Diarrhea
GI bleeding
Vomiting
Abdominal pain
Muscular
Vomiting
Colicky
Abdominal pain
Serosal
Ascites
Abdominal pain
The most frequently seen form of eosinophilic gastroenteritis is proctitis in infants. The disorder is characterized
by the bloody diarrhea in an infant less than 2 months of
age.
Cases of food-induced eosinophilic proctocolitis are
reported regardless of being breastfed or formula fed.
Infants presenting with this condition usually have normal
linear and ponderal growth. The infants will have diarrhea,
usually accompanied by mucous and/or blood, and often
with pain or straining at the time of bowel motion. Biopsy of
the rectal mucosa will show eosinophilic infiltration of the
mucosa. Frequently the diagnosis is made on clinical presentation alone. The dietary management of this condition
involves elimination of the offending protein until approximately 9 to 12 months of age. If an infant is breastfed then
the offending protein should be removed from the mothers
diet. Earlier introduction of the protein will usually result in
bleeding.123
In older children with eosinophilic conditions of the
gut symptoms include, but are not limited to, vomiting,
297
Summary
Because of the central role of the digestive system in maintaining normal nutrition, disease of the GI tract, liver, or
pancreas has a profound influence on growth and development of children. Provision of adequate nutrition support
can not only improve nutrition parameters and growth but
also in many cases can treat the underlying disease. Careful
evaluation of the child with assessment of nutrition needs is
the initial step in effective management. The clinical examination and judiciously applied laboratory investigation will
identify nutrition deficiencies. With understanding of the
underlying disease process, appropriate nutrition management can improve growth, quality of life, and outcomes of
these patients.
A 15 year old boy is admitted to the hospital because of diarrhea and abdominal pain. He has had unintentional weight
loss of 15 pounds in the last 3 months. Laboratory evaluation reveals:
Albumin 3.1
Total Protein 6.2
Hemoglobin/hematocrit 9.3/27
MCV 71 (nl >79)
WBC 11,500
Radiology studies demonstrated inflammation of the ileum
and cecum.
1. Diet therapy should include:
A. A polymeric defined formula diet
B. A semi-elemental defined formula diet
C. An elemental defined formula diet
D. Any of A, B, or C
E. A clear liquid diet
2. His medical therapy includes methotrexate and
sulphasalazine. Supplementation should include:
A. Folate, B12, and iron
B. Pyridoxine, thiamin, and magnesium
C. B12, vitamin C, and manganese
D. Folate, vitamin C, and copper
298
References
10. Nelson SP, Chen EH, Syniar GM, Christoffel KK. One year
follow up of symptoms of gastroesophageal reflux during
infancy. Pediatric Practice Research Group. Pediatrics.
1998;102:e67.
11. Vandenplas Y. Gastroesophageal reflux. In: Wyllie R, Hyams
JS, Kay M, eds. Pediatric Gastrointestinal and Liver Disease.
Pathophysiology, Diagnosis, Management. 3rd ed. London:
Saunders Elsevier; 2006:305325.
12. Pandolfino JE, Kwiatek MA, Kahrilas PJ.
The pathophysiologic basis for epidemiologic trends in gastroesophageal reflux
disease. Gastroenterol Clin North Am. 2008;37:827843.
13. Vandenplas Y, Hassall E. Mechanics of gastroesophageal
reflux and gastroesophageal reflux disease. J Pediatr Gastroenterol Nutr. 2002;35:119136.
14. Kawahara H, Dent J, Davidson G. Mechanics responsible
for gastroesophageal reflux in children. Gastroenterology.
1997;113:399408.
15. Vanderhoof JA, Moran JR, Harris CL, Merkel KL, Orenstein
SR. Efficacy of a pre-thickened infant formula: a multicenter,
double-blind, randomized, placebo-controlled parallel group
trial in 104 infants with symptomatic gastroesophageal reflux.
Clin Pediatr. 2003;42:483495.
16. Aggett PJ, Agostoni C, Axelsson I, et al. Anti-reflux or antiregurgitation milk products for infants and young children: a
commentary by the ESPGHAN Committee on Nutrition. J
Pediatr Gastroenterol Nutr. 2002;394:496498.
17. Vandenplas Y, De Wolf D, Sacre L. Influence of xanthines on
gastroesophageal reflux in infants at risk for sudden infant
death syndrome. Pediatrics. 1986;77:807810.
18. Wenzl TG, Schneider S, Scheele F, Silny J, Heimann G,
Skopnik H. Effects of thickened feeding on gastroesophageal
reflux in infants: a placebo-controlled crossover study using
intraluminal impedance. Pediatrics. 2003;111(4 Pt 1):e355e359.
19. Orenstein SR, Shalaby TM, Putnam PE. Thickened feedings
as a cause of increased coughing when used as therapy for
gastroesophageal reflux in infants. J Pediatr. 1992;121:913915.
20. Huang R-C, Forbes DA, Davies MW. Feed thickener
for newborn infants with gastro-oesophageal reflux.
Cochrane Database of Syst Rev. 2002;(3):CD003211.
doi:10.1002/14651858.CD003211.
21. Craig WR, Hanlon-Dearman A, Sinclair C, Taback SP,
Moffatt M. Metoclopramide, thickened feedings and positioning for gastro oesophageal reflux in children under two
years. Cochran Database Syst Rev. 2009;(3):CD003502.
doi:10.1002/14651858.CD003502 pub 2.
22. Bosscher D, Van Caillie-Bertrand M, Van Dyck K, Robberecht
H, Van Cauwenbergh R, Deelstra H. Thickening infant
formula with digestible and indigestible carbohydrate: availability of calcium, iron, and zinc in vitro. J Pediatr Gastroenterol
Nutr. 2000;30:373378.
23. Rudolph CD, Mazur LJ, Liptak GS, et al.; North American
Society for Pediatric Gastroenterology and Nutrition. Guidelines for evaluation and treatment of gastroesophageal reflux
in infants and children: recommendations of the North American Society for Pediatric Gastroenterology and Nutrition. J
Pediatr Gastroenterol Nutr. 2001;32:Supp 2:S131.
GASTROINTESTINAL DISEASE
24. Hills JM, Aaronson PI. The mechanism of action of peppermint oil on gastrointestinal smooth muscle. Gastroenterology.
1991;101:5565.
25. Wendl B, Pfeiffer A, Pehl C, Schmidt T, Kaess H. Effect of
decaffeination of coffee or tea on gastro-oesophageal reflux.
Aliment Pharmacol Ther. 1994;8:283287.
26. Pehl C, Pfeiffer A, Wendl B, Kaess H. The effect of decaffeination of coffee on gastroesophageal reflux in patients with reflux
disease. Aliment Pharmacol Ther. 1997;11:483486.
27. Bulat R, Fachnie E, Chauhan U, Chen Y, Tougas G. Lack of
effect of spearmint on lower esophageal sphincter function
and acid reflux in healthy volunteers. Aliment Pharmacol Ther.
1999;13:805812.
28. Penagini R, Mangano M, Bianchi PA. Effect of increasing the
fat content but not the energy load of a meal on gastro-oesophageal reflux and lower oesophageal sphincter motor function.
Gut. 1998;42:330333.
29. Lewis D, Khoshoo V, Pencharz PB, Golladay ES. Impact of
nutritional rehabilitation on gastroesophageal reflux in neurologically impaired children. J Pediatr Surg. 1994;29:167169.
30. Skidmore-Roth L. Mosbys Drug Guide for Nurses. 4th ed. St.
Louis, MO: Mosby; 2001.
31. Herzig SJ, Howell MD, Ngo LH, Marcantonio GR. Acidsuppressive medication use and the risk for hospital-acquired
pneumonia. JAMA. 2009;301(20):21202188.
32. Canani RB, Cirillo P, Roggero P, Romano C, Malamisura B,
et al. Therapy with gastric acid inhibitors increases the risk of
acute gastroenteritis and community-acquired pneumonia in
children. Pediatrics. 2006;117(5):e817e820.
33. Craig WR, Hanlon-Dearman A, Sinclair C, Taback SP,
Moffatt M. Metoclopramide, thickened feedings and positioning for gastro oesophageal reflux in children under two
years. Cochrane Database of Syst Rev. 2004;(3):CD003502.
doi:10.1002/14651858.CD003211.
34. Green PHR, Collier C. Celiac disease. N Engl J Med. 2007;
357:17311743.
35. DAmico MA, Holmes J, Stavropoulos SN, et al. Presenta
tion of celiac disease in the United States: prominent effect of
breast feeding. Clin Pediatr (Phila). 2005;44:249258.
36. Rampertab SD, Poorfan N, Baur P, Singh P, et al. Trends in
the presentation of celiac disease. Am J Med. 2006;119(4):
355.e9355.e14.
37. Haapalahti M, Kulmala P, Karttunen TJ, et al. Nutritional
status in adolescents and young adults with screen-detected
celiac disease. J Pediatr Gastroenterol Nutr. 2005;40:566570.
38. Fasano A, Catassi C. Current approaches to diagnosis and
treatment of celiac disease: an evolving spectrum. Gastroenterology. 2001;120:636651.
39. See J, Murray JA. Gluten-free diet: The medical and nutrition
management of celiac disease. Nutr Clin Pract. 2006;21:115.
40. Hallert C, Grant C, Grehn S, et al. Evidence of poor vitamin
status in celiac patients on a gluten-free diet for 10 Years.
Aliment Pharmacol Ther. 2002;16:13331339.
41. Mora S, Barera G, Beccio S, et al. A prospective, longitudinal
study of the long-term effect of treatment on bone density in
children with celiac disease. J Pediatr. 2001;139(4):473475.
42. Shamir R. Nutritional aspects in inflammatory bowel disease.
J Pediatr Gastroenterol Nutr. 2009;48:586588.
299
300
GASTROINTESTINAL DISEASE
95. March PC. What is the best way to feed patients with pancreatitis. Curr Opin Crit Care. 2009;15:131138.
96. Al-Omran M, Groof A, Wilke D. Enteral versus parenteral
nutrition for acute pancreatitis. Cochrane Database of Syst Rev.
2003;(1):CD002837. doi:10.1002/14651858.CD002837
97. Petrov MS, Loveday BP, Pylypchuk RD, McIlroy K, Phillips AR, Winsdor JA. Systematic review and meta-analysis of
enteral nutrition formulations in acute pancreatitis. 2009; Br
J Surg. 96:12431252.
98. Doley RP, Yadav TD, Wig JD, et al. Enteral nutrition in severe
acute pancreatitis. J Pancreas (Online). 2009;9:157162.
99. Werlin SL, Kugathasan S, Frautschy BC. Pancreatitis in children. J Pediatr Gastroenterol Nutr. 2003;12:4752.
100. Suddaby EC, Schiller S. Management of chylothorax in children. Pediatr Nurs. 2004;30:290295.
101. McDonald KQ , Bears CM. A preterm infant with intestinal
lymphangiectasia: a diagnostic dilemma. Neonatal Network.
2009;28:2936.
102. Bliss CM, Schroy PC. Primary intestinal lymphangiectasia.
Curr Treat Options Gastroenterol. 2004;7:36.
103. Hyams J, Colletti R, Faure C, et al. Functional gastrointestinal
disorders: Working Group Report of the First World Congress
of Pediatric Gastroenterology, Hepatology and Nutrition. J
Pediatr Gastroenterol Nutr. 2002;35:S110117.
104. Feigenbaum K. Treating gastroparesis. Diabetes Self Manag.
2006;23(5):24.26, 2830, 32.
105. Kim CH, Nelson DK. Venting percutaneous gastrostomy in
the treatment of refractory idiopathic gastroparesis. Gastrointest Endosc. 1998;47(1):6770.
106. Parkman HP, Hasler WL, Fisher RD. American Gastroenterological Association technical review on the
diagnosis and treatment of gastroparesis. Gastroenterology.
2004;127:15921622.
107. Parrish CR, Yoshida CM. Nutrition intervention for the
patient with gastroparesis: an update. Pract Gastroenterol.
2005;30:2966.
108. Gabay C, Kushner I. Acute-phase proteins and other
systemic responses to inflammation. New Engl J Med.
1999;320:448454.
109. Clein NW. Cows milk allergy in infants. Pediatr Clin North
Am. 1954;25:949962.
110. Iacono G, Cavataio F, Montalto G, et al. Intolerance of cows
milk and chronic constipation in children. N Engl J Med.
1998;339:11001104.
111. Dahr S, Tahan S, Sole D, et al. Cows milk protein intolerance
and chronic constipation in children. Pediatr Allergy Immunol.
2001;12:339342.
301
112. Guimres EV, Goulart EM, Penna FJ. Dietary fiber intake,
stool frequency and colonic transit time in chronic functional constipation in children. Braz J Med Biol Res.
2001;34:11471153.
113. Pijpers, MA, Tabbers MM, Benninga MA, et al. Currently
recommended treatments of childhood constipation are not
evidence based: a systematic literature review on the effect
of laxative treatment and dietary measures. Arch Dis Child.
2009;94:117131.
114. Huertas-Ceballos AA, Logan S, Bennett C, Macarthur C.
Dietary interventions for recurrent abdominal pain (RAP) and
irritable bowel syndrome (IBS) in childhood. Cochrane Database of Syst Rev. 2009;(1)CD003019. doi:10.1002/14651858.
CD003019.pub3.
115. Whitfield KL, Shulman RJ. Treatment options for functional
gastrointestinal disorders: from empiric to complementary
approaches. Pediatr Ann. 2009;38:288294.
116. Sood MR. Treatment approaches to irritable bowel syndrome.
Pediatr Ann. 2009;38:272276.
117. Goldstein R, Braverman D, Stankiewicz H. Carbohydrate
malabsorption and the effect of dietary restriction on symptoms of irritable bowel syndrome and functional bowel
complaints. Israel Med Assoc J. 2000;2:583587.
118. Fernndez-Baares F, Esteve-Pardo M, de Leon R, et al. Sugar
malabsorption in functional bowel disease: clinical implications. Am J Gastroenterol. 1993;88:20442050.
119. Shen YA, Nahas R. Complementary and alternative medicine
for treatment of irritable bowel syndrome. Can Fam Physician.
2009;55:143148.
120. Bijkerk CJ, Muris JWM, Knottnerus JA, Hoes AW, de Wit
NJ. Systematic review: the role of different types of fibre in
the treatment of irritable bowel syndrome. Aliment Pharmacol
Ther. 2004;19:245251.
121. Quartero AO, Meiniche-Schmidt V, Muris J, Rubin G, de
Wit N. Bulking agents, antispasmodic and antidepressant
medication for the treatment of irritable bowel syndrome.
Cochrane Database of Syst Rev. 2005;(2):CD003460.
doi:10.1002/14651858.CD003460.pub2.
122. Torii A, Toda G. Management of irritable bowel syndrome.
Intern Med. 2004;43:353358.
123. Talley NJ. Gut eosinophilia in food allergy and systemic
and autoimmune diseases. Gastroenterol Clin N Am.
2008;37:307332.
124. Salvatore S, Hauser B, Devreker T, Arrigo S, Vandenplas Y.
Chronic enteropathy and feeding in children: an update.
Nutrition. 2008;24:12051216.
26
Hepatic Disease
Samuel A. Kocoshis, MD and Renee A. Wieman, RD, CSP, LD, CNSD
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
Malnutrition in Liver Diseases . . . . . . . . . . . . . . . . . . . . . 303
Inadequate Intake
Iatrogenic Factors
Malabsorption
Hypermetabolism
302
Learning Objectives
1. Recognize the energy and protein requirements of children with chronic liver disease.
2. Describe nutrition support modalities for children with
chronic liver disease.
3. Understand the diagnosis and management of metabolic liver diseases such as glycogenoses and Wilsons
disease.
4. Comprehend the factors causing malabsorption in
pediatric liver disease.
Introduction
HEPATIC DISEASE
Malnutrition is common in pediatric liver disease and adversely affects survival prior to and following liver transplantation. Beyond the impaired intermediate metabol ism
of carbohydrate, protein, and fat, a variety of factors ac
counts for malnutrition in the context of primary hepatic
disease. Most studies exploring mechanisms of malnutrition in liver disease have been performed in adults, but it
is known that malnutrition can occur both prior to and
after liver transplantation in children. Even though those
mechanisms may not have been fully investigated in pediatric patients, it is quite likely that they contribute to pediatric as well as adult malnutrition among patients with
liverdisease.
Inadequate Intake
A foremost concern among adult and pediatric liver patients
is inadequate dietary intake. A simple reason that intake
might be inadequate is that these patients are frequently
offered low-protein, low-sodium diets that are unappetizing and unappealing. This practice may not only result
in suboptimal oral intake but also provide a diet of poor
caloric quality. Secondly, many forms of liver disease result
in anorexia. One potential mechanism is elevation of serum
and tissue leptin levels.4 Leptin, an appetite-suppressing
hormone, is probably cleared via the liver, and serum levels
are elevated in patients with hepatic fibrosis as well as
other forms of liver disease. The role of leptin in producing
anorexia among patients with liver disease remains controversial, specifically because leptin levels are consistently
normal in some forms of liver disease such as primary
biliary cirrhosis and consistently elevated in other forms
such as Laennecs cirrhosis.4 A third factor accounting for
303
Iatrogenic Factors
The effect of iatrogenic factors upon the nutrition state of
pediatric liver patients should not be minimized. Excessive
restriction of sodium in the absence of fluid overload will
result in a salt depletion syndrome, and may actually induce
a secondary hyperaldosteronism that would not have been
present otherwise. This factor can lead to both malnutrition and growth failure. Unwarranted protein restriction
can also result in deficiencies of both somatic and visceral
proteins with subsequent malnutrition. Therefore, clinicians caring for children with liver dysfunction should
refrain from restricting either salt or protein in the absence
of ascites or encephalopathy refractory to pharmacotherapy.
Known to be relatively safe, large-volume paracentesis has
been a popular therapy for adults with ascites, but it has
been relatively unpopular in the pediatric setting because
of concerns about the large, rapid fluid shifts that might be
induced.6 This potential complication notwithstanding,
the practice now has advocates within the pediatric hepatology community. If large-volume paracentesis is used
in pediatric patients, the loss of plasma proteins can be
prodigious, rendering the patient deficient in both visceral
and somatic proteins. Hence, for patients undergoing paracentesis, adequate protein intake should be maintained in
the absence of overt encephalopathy. Many of the medications administered to children with hepatic disease may
negatively impact upon their nutrition state. Diuretics, if
administered overzealously, may salt-deplete them. Broadspectrum systemic or enteral antibiotics may eliminate
vitamin K-synthesizing enteric flora, resulting in vitamin
K deficiency among cholestatic patients. Additionally,
2010 A.S.P.E.N. www.nutritioncare.org
304
neomycin, commonly administered for hepatic encephalopathy, is believed to produce villous atrophy, to reduce
intestinal surface area, and to result in malabsorption of
multiple nutrients. Administration of lactulose for encephalopathy may speed intestinal transit enough to result in
malabsorption as well. Finally, cholestyramine, an anion
exchange resin used to treat the pruritis of cholestasis by
binding bile acids, may be so efficient that the intraluminal
bile acid concentration may fall below the critical micellar
concentration, resulting in fat and fat-soluble vitamin
malabsorption.7 Because cholestyramine exchanges organic
anions for chloride, hyperchloremic metabolic acidosis
with resultant growth failure and malnutrition may occur
in children on this medication.
Malabsorption
Pediatric patients with liver disease are more likely to have
cholestatic disease than their adult counterparts. In cholestatic patients, bile acids are retained within the liver, and
excreted very poorly into bile. Therefore, intraduodenal
primary bile acid concentrations customarily fall below
the critical micellar concentration necessary for efficient
solubilization and transport of fat and fat-soluble vitamins across the unstirred water layer into the enterocyte.
Thus malabsorption is very common and requires enteral
administration of large quantities of fat-soluble vitamins
as well as an enteral cocktail of medium-chain triglycerides (MCTs) and long-chain triglycerides (LCTs) in food
or formula. Patients must receive enough long-chain fat
to prevent essential fatty acid deficiency (EFAD) and they
must receive enough medium-chain fat to optimize their
enteric fat balance. Linoleic and linolenic acids, which are
essential fatty acids (EFAs), are long-chain fats that can
only be derived via dietary LCTs. MCT formulas that were
designed during the late 1960s for cholestatic patients were
deficient in LCTs, commonly resulting in EFAD among this
population.8
Malabsorption may occur in select cases of pediatric
cholestasis from not only intraluminal bile acid deficiency
but also from exocrine pancreatic insufficiency. Both cystic
fibrosis and Shwachman-Diamond syndrome, systemic
disorders characterized by exocrine pancreatic insufficiency,
may present during the neonatal period with cholestasis.
Exocrine pancreatic insufficiency is a less-recognized
feature of two childhood disorders characterized by severe
cholestasis: progressive familial intrahepatic cholestasis,
type 1 (PFIC1) and Alagille syndrome. PFIC1, previously
known as Byler disease, is due to a mutation in the ATP8B1
gene, previously known as the FIC1 gene. ATP8B1 mutations
2010 A.S.P.E.N. www.nutritioncare.org
Hypermetabolism
The prevalence of hypermetabolism is unknown in children with liver disease, but at least 30% of cirrhotic adults
are hypermetabolic.2 Even though some cirrhotic adults
are normometabolic and a few are hypometabolic, those
who are hypermetabolic display measured resting energy
expenditure (REE) 20% or more above predicted energy
expenditure. Hypermetabolism in cirrhotic adults is closely
associated with suboptimal total body mass and total body
protein. One documented mechanism for this phenomenon
is increased adrenal tone presumably because of reduced
HEPATIC DISEASE
305
306
Wilsons Disease
Wilsons disease is due to a mutation in a p-type ATPase
responsible for transporting copper across membranes to
permit formation of metallothionein and to permit biliary
excretion of copper.19 Defective copper excretion results in
excessive hepatic copper stores leading to hepatic dysfunction. Excessive brain and renal copper stores are responsible
2010 A.S.P.E.N. www.nutritioncare.org
HEPATIC DISEASE
307
Energy
Protein
Goals for age are generally provided unless encephalopathy
with fulminant hepatic failure and an elevated ammonia
level are observed. Enough protein must be provided to
preserve lean body mass and prevent catabolism with the
breakdown of endogenous protein stores, but not contribute
to hyperammonemia and encephalopathy. Infants generally
require 3 to 4 g/kg/d.24 The use of branched-chain amino
acid (BCAA) formulas or supplements in infants and children, though improving nitrogen balance, 26 has fallen out of
favor because the cost/benefit ratio remains low.
308
Amount Given
Vitamin A (aqueous)
25-OH vitamin D
Vitamin E
Vitamin K
Fluids
Management of fluid homeostasis, ascites, and sodium
balance mandate maintaining a delicate balance for pediatric patients with liver disease. The decision to diurese a
patient with hyponatremia generally depends upon a global
assessment of fluid status. A patient with signs of dehydration should be given salt when hyponatremic whereas a
patient appearing overhydrated may actually be suffering
from dilutional hyponatremia and require diuresis with
fluid and salt restriction. Only non-nutritive fluids should
be limited when fluid restriction is initiated. 28 Salt restriction should not be excessive, because salt elimination is
much more benignly accomplished via loop diuretics.
Parenteral Nutrition
HEPATIC DISEASE
Conclusion
References
309
310
27
Intestinal Failure
Robert H. Squires, Jr., MD
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Definition of Intestinal Failure . . . . . . . . . . . . . . . . . . . . . 312
Scope of the Problem
Complications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316
Nutrition Deficiencies
Functional and Metabolic Complications
Liver Disease
Central Line Complications
Outcomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 318
Growth and Development
Quality of Life
Intestinal Transplant
Learning Objectives
Introduction
Intestinal failure (IF) in infants and children is a devastating condition that can be broadly defined as the inability
of the intestinal tract to sustain life without supplemental
parenteral nutrition (PN).1,2 Prior to PN, many infants died
as a consequence of insufficient bowel length or function.
Infants found to have an abdominal catastrophe at laparotomy for conditions such as necrotizing enterocolitis
(NEC), volvulus, or gastroschisis were simply closed and
made comfortable to await the natural course of their tragic
circumstance. However, with the development of safe PN,
central line placement and care, improved medical and
surgical management, including intestinal transplantation, these infants now have an opportunity to survive with
a satisfactory or even excellent quality of life. The clinical
course for the child can be challenging and frustrating
to the family as well as those who provide medical care
and support. Intestinal adaptation, if it occurs at all, can
require months or years before the intestine can adequately
311
312
INTESTINAL FAILURE
Intestinal Adaptation
Intestinal adaptation is a complex and incompletely understood process that ensues following significant bowel
resection to compensate for the loss of intestinal surface
area. It is characterized by both functional and morphologic
changes in the remnant bowel.12,13 Enteral nutrition (EN) is
necessary for adaptation. Clinical features associated with
patients who are eventually weaned from PN include the
absence of jaundice or cholestasis, a shorter duration of PN,
the presence of the ileal-cecal valve, an intact colon, small
bowel length greater than 15 cm, and placement of the small
intestine in continuity with the colon. 5 The underlying
physiologic mechanisms by which these clinical factors
might directly impact the adaptive process in the human
is incompletely understood. For instance, it is not clear
whether the ileal-cecal valve must be physically present or
whether its presence merely serves as a marker for a long
colon segment. Over time, the residual bowel lengthens and
dilates and is thought to be a positive sign of adaptation.
However, this anatomical change may be associated with
complications such as altered intestinal motility that can
result in stasis of luminal contents and bacterial overgrowth
which can negatively impact adaptation. Surgical techniques
aimed to improve intestinal function by increasing bowel
length and reducing bowel diameter include the Bianchi
procedure and Serial Transverse Enteroplasty (STEP).14,15
The adaptive process occurs over many months or
years. 5,16 Increased enteral intake, or hyperphagia, is associated with intestinal adaptation in adults.17 The impact of
luminal nutrients upon intestinal adaptation is complex,
but likely involves a direct trophic effect on intestinal
epithelium, as well as stimulation of trophic hormones
and pancreaticobiliary secretions.18 In animal models, it
appears that a more complex nutrient enhances adaptation better than one that is simple or more processed.
313
Management
Enteral Nutrition
Enteral feeding is essential if intestinal adaptation is to
occur. The 2 initial considerations are the type of formula
used and the manner in which it is provided. Breast milk
is preferred and its use has been associated with decreased
duration of PN. 3 The beneficial effects of breast milk are
attributable to its immunoprotective properties, effect on
postnatal development of intestinal flora, and its nutrient
composition that includes long-chain triglycerides (LCTs),
free amino acids, nucleotides, and growth factors as well
as complex protein and fat. 27 When breast milk is unavailable, however, formula choice is complicated by a variety of
options that reflect differences in the desired complexity of
protein, fat, and carbohydrate. Complex nutrients, such as
polysaccharides and whole protein, increase the functional
workload of the intestine, and as result, may stimulate adaptation.28 Unfortunately, studies to identify the ideal enteral
protein 2931 and lipid32 have been plagued by insufficient
2010 A.S.P.E.N. www.nutritioncare.org
314
Parenteral Nutrition
Parenteral nutrition (PN), first introduced in the late 1960s,
is now an established life-saving treatment for children with
IF/SBS. 3638 Components of PN include glucose, amino
acids, lipids, electrolytes, vitamins, minerals, trace elements,
and water. Glucose is the primary source of energy in PN,
but glucose oxidation varies depending upon age and diagnosis. 39,40 Glucose infusion rates that are in excess of the
patients oxidative capacity will promote fat deposition.41
While glucose infusion rates vary, an intake greater than
10 mg/kg/min may result in the conversion of glucose to
fat.42 An amino acid solution, ranging from > 2.5 g/kg/d for
preterm infants to 0.75 g/kg/d for adolescents, is administered to support protein metabolism.43 Amino acids are best
utilized when balanced with a proper proportion of nonnitrogen calories. The ideal ratio of nitrogen:non-nitrogen
calories is estimated to be between 1:150 to 1:400.44 The
lipid source currently available in the United States is soy
2010 A.S.P.E.N. www.nutritioncare.org
Fluid Management
In addition to PN, children may also require supplemental
fluid management. This is particularly important for children with an ileostomy or those who are in continuity, but
with a short length of colon. These children are at increased
risk of developing salt and water depletion and should be
monitored carefully.47 Unless the childs fluid and electrolyte requirements are very consistent day to day and week to
week, supplemental fluids should be calculated and administered separate from PN as this allows for more rapid,
and less expensive, adjustments in fluid and electrolyte
administration.
Medications
A variety of medications are used to manage symptoms and
complications associated with IF/SBS. It is important to
know that not one of them has been studied in a randomized fashion in an adequately powered study.
Intestinal dysmotility is common in children with IF/
SBS. Anti-motility agents (eg, loperamide) are used in an
INTESTINAL FAILURE
Non-Transplant Surgery
Half of children with SBS will have more than one abdominal operation.48 Following the initial surgical intervention,
subsequent surgeries often address complications such
as stricture, intestinal dilation, and placement of invasive
feeding devices which are all considered standard of care.49
Longitudinal intestinal lengthening and tailoring
(LILT) and STEP represent advanced techniques that
both taper dilated segments of bowel and increase intestinal length. Digestive function may improve not only
because aboral flow is facilitated by a normalized luminal
caliber, but also because subsequent adaptation may
lead to increased intestinal surface area. LILT was first
described in 1980 by Bianchi,15 and has since been used
at many centers.1 Criteria for using LILT vary, but generally require greater than 20 cm of symmetrically dilated
intestine in the context of residual intestine > 40 cm.1
Standardized indications, contraindications, and surgical
guidelines have not been developed, which may explain
inconsistent results reported in literature. 50 STEP involves
the application of a stapling device incompletely across
a dilated loop of intestine in serial fashion to create a
zig-zag pattern that results in a lumen both narrower and
longer. Since its description by Kim et al 51 in 2003, it has
315
General Principles
With the understanding that management of IF is highly
individualized and that evidence-based practice supported
by randomized, controlled trials is lacking, the following
outline might serve as a principled guide to the management of intestinal failure in children:
1. TPN
a. Glucose infusion rate: < 15 mg/kg/min for the duration
of infusion.
b. Protein: < 3 g/kg/d, closer to 3 for infants, lower for older
children.
c. Fat: In the first month, may need 12 g/kg/d; but after
that, with an eye on the degree of cholestasis and total
caloric needs, would consider different strategies.
i. 1 g/kg/d given every other day; or MWF or M/Th.
ii. Lipid reduction presumes that some enteral feeding
is possible.
iii. Follow essential fatty acids every 3 months.
d. Fluid: Based on needs, it is hard to provide sufficient calories with less than 125 cc/kg/d; may need additional salt
and water to support losses.
2. Enteral feeds
a. Use oral feeds if at all possible to maintain oral skills and
stimulate EGF, even if the child has a G-tube. 35 cc/feed
and then gradually increase as one can.
b. Breast milk is preferred and a casein hydrolysate/MCTcontaining formula is the authors fallback, assuming that
intestinal adaptation is favored by a more complex enteral
diet; use of an amino acid-containing formula can be
reserved for protein allergy.
c. J-tubes should be used rarely, as they further shorten the
length of useable bowel length.
3. Advancing feeds/reducing PN
a. The patient will tell you.
i. More concerned about the volume of stools than the
number (which can be confused with squirts); so,
for example, 10 stools per day may be okay if only
34 or so are big enough to leave the diaper; if the
bottom is not excoriated; if the family is comfortable; if
hydration is maintained.
2010 A.S.P.E.N. www.nutritioncare.org
316
ii. Aim for a rate of weight gain that follows the isopleth in
a box that surrounds the 5th percentile; adjust for SGA.
Not trying to achieve weight gain at the 50th percentile,
although if one can with minimal PN support, that is
fine. The goal is to find the minimal amount of PN to
support reasonable weight gain and growth.
b. Once on < 25% total calories from PN, and reasonable
stool output and satisfactory weight gain and growth
is established, one may consider a change from PN to
D10 LR observe for 23 months; then hold the fluids for
2months or so before pulling the line; unless the line is so
much trouble that keeping it in is too problematic.
4. Non-transplant surgery
a. When to consider in a child with short bowel syndrome
i. A sufficient length of small bowel is dilated and
1. Enteral feedings are not able to be advanced
2. The child is experiencing poor weight gain and/
or growth
b. What type of surgery?
i. May be surgeon specific; a learning curve is needed
for both the STEP and Bianchi, but more so for the
Bianchi
ii. You can STEP a STEP, and STEP a Bianchi, but you
cannot Bianchi a STEP
Complications
Nutrition Deficiencies
Deficiencies in vitamins, minerals, and trace elements
are associated with IF/SBS. Malabsorption of fat-soluble
vitamins A, D, E, and K may result from an insufficient
intraluminal concentration of bile acids secondary to
excess fecal loss. 53 Surgical resections of the duodenum or
ileum, which have unique absorptive functions, add to the
risk of developing nutrition deficiencies. The duodenum is
a primary site for iron and folate absorption and its resection can result in these micronutrient deficiencies. Absence
of the ileum, with its unique ability to absorb vitamin B12
and bile acids, places the patient at risk for fat-soluble
vitamin and vitamin B12 deficiency. Calcium and magnesium deficiencies can result from binding to intraluminal
long-chain fatty acid. 54,55 Deficiencies of other minerals and
trace elements such as zinc, riboflavin, thiamin, biotin, and
also selenium can occur. 56 In addition, nutrient deficiencies hinder intestinal adaptation further compounding the
clinical impact. 57
INTESTINAL FAILURE
Liver Disease
Liver disease is the most frequent complication of long-term
PN with consequences that include cirrhosis, end-stage liver
disease, and even death.80 The incidence of IF/SBS-associated liver disease (IFALD) is as high as 50% in infants who
receive PN for 2 months with end-stage liver disease developing in 90% of premature infants on PN for more than
3 months.81 Elevated serum transaminase and bilirubin
levels are commonly observed in infants on PN, but these
levels can normalize and jaundice resolve with intestinal
adaptation and PN withdrawal. The long-term outcome
of IFALD in patients who adapt is unknown. The population currently thought to be at greatest risk for IFALD are
premature infants with a birth weight < 1 kg and those with
IF/SBS resulting from surgical resection.8284 The etiology of
IFALD remains unknown but is likely multi-factorial with
prematurity, multiple abdominal surgeries, lack of enterally
stimulated bile flow, bacterial sepsis, and components or
deficiencies of PN infusates all potential contributors.
Treatment of PN-associated liver disease is empiric and
imperfect. Current strategies to avert liver disease associated with long-term PN include employing a choleretic such
as ursodeoxycholic acid, 85 bowel decontamination to treat
bacterial overgrowth, 83,85 vigilant daily catheter care,86 and
modifying PN formulations. Infusing PN calories over less
than 24 hours, casually referred to as cycling PN, is thought
to improve cholestasis.87 The underlying mechanism for this
action and effect is not clearly understood, but may involve
providing a metabolic rest from the continuous infusion
of calories, protein, and/or carbohydrate. Administration of
pediatric formulations of PN that include specific targeted
amino acids including taurine88 and limiting the infusion
of dextrose so as to potentially decrease steatosis have also
been utilized.89 Decreasing the aluminum and manganese
content in PN may decrease hepatotoxicity.90,91 Decreasing
the amount or altering the type of lipids administered may
improve serum transaminase levels, reverse PN-associated
liver disease, or reduce lipid peroxidation.46,92,93 Recently,
use of a fish oil-based intravenous lipid source was found to
be associated with improvement of serum bilirubin in two
children,46 although its long-term use in an animal model
was associated with increased fibrosis.94 Unfortunately,
most of these interventions to reduce or prevent IFALD
are employed empirically and lack significant clinical
confirmation in large pediatric trials. Once end-stage liver
317
318
Outcomes
Growth and Development
Infants and children must digest and absorb sufficient
calories, vitamins, minerals, and trace elements to gain
weight, grow, and develop. Children with IF/SBS are incapable of sustaining adequate growth and development
without supplemental PN. Successful intestinal adaptation
implies complete independence from PN while continuing
to demonstrate satisfactory growth and development.
However, there are no prospective studies available to
address long-term growth and development in children
with IF/SBS following PN withdrawal. A recent retrospective study of 87 children identified over a period of 16 years
provides some insight into the long-term problems that
children with IF/SBS may face.16 The authors found that in
those children who achieved enteral autonomy, maximum
weight gain and growth was achieved during the first 4 years
after weaning PN. However, between 4 and 8 years post-PN,
weight gain and growth, expressed as a z score, appeared
to decline with the weight z score 8 years post-PN almost
identical to the weight at the time of weaning and height z
score slightly lower than the score at the time of weaning.
Interestingly, 21 children were noted to enter puberty at an
age similar to their peers.
Quality of Life
Studies in adult patients have identified reduced quality of
life (QOL) scores in patients with SBS, which were further
reduced if the patient was on HPN. Interestingly, the presence of a stoma did not appear to influence their quality
of life.102 Similarly detailed studies that address QOL and
school performance have not been performed in children,
however, children on HPN appear to fare better than those
hospitalized for PN.103,104
Intestinal Transplant
Intestinal transplant is reserved as the final life-saving
procedure for patients with irreversible IF/SBS and lifethreatening complications of PN administration. In 1994,
very few intestinal transplants were performed in the pediatric population worldwide (approximately 25 according
to the Intestinal Transplant Registry), with unsatisfactory outcomes in these early years.1 With advances in
2010 A.S.P.E.N. www.nutritioncare.org
1. A 30-week infant with 35 cm of small bowel, an ilealcecal valve, and an intact colon has an advantage over a
full-term infant with the same anatomy because:
A. Etiologies for short bowel syndrome are different
between the two groups.
B. Premature infants who survive are constitutionally
stronger than full-term infants.
C. The small intestinal length doubles in the last
trimester.
D. Complications associated with PN are less frequent
in the premature infant.
2. A clinical feature associated with enteral autonomy is:
A. Being a female
B. Small intestine in continuity with the colon
C. Absent ileal-cecal valve
D. Having fewer than 10 stools per day
3. PN associated liver disease is caused by:
A. Intravenous lipid
B. Lack of enteral feeding
C. Recurrent infections
D. Multiple factors, including all of the above
4. The best method to provide enteral feeding for children
with intestinal failure is via:
A. Gastrostomy
B. Nasogastric tube
C. Jejunal feeding
D. Oral feeding
See p. 487 for answers.
INTESTINAL FAILURE
References
1. Goulet O, Ruemmele F, Lacaille F, Colomb V. Irreversible intestinal failure. J Pediatr Gastroenterol Nutr.
2004;38(3):250269.
2. Kocoshis SA, Beath SV, Booth IW, et al. Intestinal failure
and small bowel transplantation, including clinical nutrition: Working Group Report of the Second World Congress
of Pediatric Gastroenterology, Hepatology, and Nutrition. J
Pediatr Gastroenterol Nutr. 2004;39(Suppl 2):S655S661.
3. Andorsky DJ, Lund DP, Lillehei CW, et al. Nutritional and
other postoperative management of neonates with short
bowel syndrome correlates with clinical outcomes. J Pediatr.
2001;139(1):2733.
4. Goulet OJ, Revillon Y, Jan D, et al. Neonatal short bowel
syndrome. J Pediatr. 1991;119(1 ( Pt 1)):1823.
5. Quios-Teijeira RE, Ament ME, Reyen L, et al. Long-term
parenteral nutritional support and intestinal adaptation in
children with short bowel syndrome: a 25-year experience. J
Pediatr. 2004;145(2):157163.
6. Spencer AU, Neaga A, West B, et al. Pediatric short bowel
syndrome: redefining predictors of success. Ann Surg.
2005;242(3):403409; discussion 409412.
7. Touloukian RJ, Smith GJ. Normal intestinal length in preterm
infants. J Pediatr Surg. 1983;18(6):720723.
8. DeLegge M, Alsolaiman MM, Barbour E, et al. Short bowel
syndrome: parenteral nutrition versus intestinal transplantation. Where are we today? Dig Dis Sci. 2007;52(4):876892.
9. Wallander J, Ewald U, Lackgren G, et al. Extreme short bowel
syndrome in neonates: an indication for small bowel transplantation? Transplant Proc. 1992;24(3):12301235.
10. Schalamon J, Mayr JM, Hollwarth ME. Mortality and
economics in short bowel syndrome. Best Pract Res Clin
Gastroenterol. 2003;17(6):931942.
11. Colomb V. Economic aspects of paediatric home
parenteral nutrition. Curr Opin Clin Nutr Metab Care.
2000;3(3):237239.
12. Tavakkolizadeh A, Whang EE. Understanding and augmenting human intestinal adaptation: a call for more clinical
research. J Parenter Enteral Nutr. 2002;26(4):251255.
13. Drozdowski L, Thomson AB. Intestinal mucosal adaptation .
World J Gastroenterol. 2006; 2(29):46144627.
14. Javid P, Kim H, Duggan C, Jaksic T. Serial transverse enteroplasty is associated with successful short-term outcomes
in infants with short bowel syndrome. J Pediatr Surg.
2005;40(6):10191023; discussion 10231024.
15. Bianchi A. Intestinal loop lengthening--a technique
for increasing small intestinal length. J Pediatr Surg.
1980;15(2):145151.
16. Goulet O, Gobet B, Talbotec C, et al. Outcome and longterm growth after extensive small bowel resection in the
neonatal period: a survey of 87 children. Eur J Pediatr Surg.
2005;15(2):95101.
17. Crenn P, Morin MC, Joly F, Penven S, Thuillier F, Messing B.
Net digestive absorption and adaptive hyperphagia in adult
short bowel patients. Gut. 2004;53(9):12791286.
18. DiBaise JK, Young RJ, Vanderhoof JA. Intestinal rehabilitation and the short bowel syndrome: part 1. Am J Gastroenterol.
2004;99(7):13861395.
319
19. Weser E, Babbitt J, Hoban M, Vandeventer A. Intestinal adaptation. Different growth responses to disaccharides compared
with monosaccharides in rat small bowel. Gastroenterology.
1986;91(6):15211527.
20. Vanderhoof JA, Grandjean CJ, Burkley KT et al. Effect of
casein versus casein hydrolysate on mucosal adaptation
following massive bowel resection in infant rats. J Pediatr
Gastroenterol Nutr. 1984;3(2):262267.
21. Wasa M, Takagi Y, Sando K, et al. Intestinal adaptation in
pediatric patients with short-bowel syndrome. Eur J Pediatr
Surg. 1999;9(4):207209.
22. Gambarara M, Ferretti F, Bagolan P. Ultra-short-bowel
syndrome is not an absolute indication to smallbowel transplantation in childhood. Eur J Pediatr Surg.
1999;9(4):267270.
23. Jeppesen PB, Mortensen PB. The influence of a preserved
colon on the absorption of medium chain fat in patients with
small bowel resection. Gut. 1998;43(4):478483.
24. Nightingale JM, Lennard-Jones JE, Gertner DJ, et al.
Colonic preservation reduces need for parenteral therapy,
increases incidence of renal stones, but does not change high
prevalence of gall stones in patients with a short bowel. Gut.
1992;33(11):14931497.
25. Luo M, Fernndez-Estvariz C, Manatunga AK, et al. Are
plasma citrulline and glutamine biomarkers of intestinal
absorptive function in patients with short bowel syndrome? J
Parenter Enteral Nutr. 2007;31(1):17.
26. Bernal NP, Stehr W, Zhang Y, Profitt S, Erwin CR, Warner
BW. Evidence for active Wnt signaling during postresection
intestinal adaptation. J Pediatr Surg. 2005;40(6):10251029;
discussion 1029.
27. Levy J. Immunonutrition: the pediatric experience. Nutrition.
1998;14(78):641647.
28. Vanderhoof JA, Langnas AN. Short-bowel syndrome in children and adults. Gastroenterology. 1997;113(5):17671778.
29. Ksiazyk J, Piena M, Kierkus J, Lyszkowska M. Hydrolyzed
versus nonhydrolyzed protein diet in short bowel syndrome in
children. J Pediatr Gastroenterol Nutr. 2002;35(5):615618.
30. Bines J, Francis D, Hill D. Reducing parenteral requirement
in children with short bowel syndrome: impact of an amino
acid-based complete infant formula. J Pediatr Gastroenterol
Nutr. 1998;26(2):123128.
31. Taylor S, Sondheimer J, Sokol R, Silverman A, Wilson H.
Noninfectious colitis associated with short gut syndrome in
infants. J Pediatr. 1991;119(1 ( Pt 1)):2428.
32. Vanderhoof JA, Grandjean CJ, Kaufman SS, Burkely KT,
Antonson DL. Effect of high percentage medium-chain triglyceride diet on mucosal adaptation following massive bowel
resection in rats. J Parenter Enteral Nutr. 1984;8(6):685689.
33. Blackman JA, Nelson CL. Reinstituting oral feedings in
children fed by gastrostomy tube. Clin Pediatr. (Phila)
1985;24(8):434438.
34. Dsilna A, Christensson K, Alfredsson L, et al. Continuous
feeding promotes gastrointestinal tolerance and growth in
very low birth weight infants. J Pediatr. 2005;147(1):4349.
35. Shulman RJ, Redel CA, Stathos TH. Bolus versus continuous feedings stimulate small-intestinal growth and
development in the newborn pig. J Pediatr Gastroenterol Nutr.
1994;18(3):350354.
2010 A.S.P.E.N. www.nutritioncare.org
320
INTESTINAL FAILURE
321
90. Advenier E, Landry C, Colomb V, et al. Aluminum contamination of parenteral nutrition and aluminum loading in children
on long-term parenteral nutrition. J Pediatr Gastroenterol Nutr.
2003;36(4):448453.
91. Kafritsa Y, Fell J, Long S, Bynevelt M, Taylor W, Milla P.
Long-term outcome of brain manganese deposition in
patients on home parenteral nutrition. Arch Dis Child.
1998;79(3):263265.
92. Goulet O, de Potter S, Antbi H, et al. Long-term efficacy
and safety of a new olive oil-based intravenous fat emulsion
in pediatric patients: a double-blind randomized study. Am J
Clin Nutr. 1999;70(3):338345.
93. Cavicchi M, Crenn P, Beau P, Degott C, Boutron MC,
Messing B. Severe liver complications associated with longterm parenteral nutrition are dependent on lipid parenteral
input. Transplant Proc. 1998;30(6):2547.
94. Kohl M, Wedel T, Entenmann A, et al. Influence of
different intravenous lipid emulsions on hepatobiliary
dysfunction in a rabbit model. J Pediatr Gastroenterol Nutr.
2007;44(2):237244.
95. Rodrigues AF, Van Mourik IDM, Sharif K, et al. Management of end-stage central venous access in children referred
for possible small bowel transplantation. J Pediatr Gastroenterol Nutr. 2006;42(4):427433.
96. Lang EV, Reyes J, Faintuch S, Smith A, Abu-Elmagd K. Central
venous recanalization in patients with short gut syndrome:
restoration of candidacy for intestinal and multivisceral transplantation. J Vasc Interv Radiol. 2005;16(9):12031213.
97. Lichtman SN. Translocation of bacteria from gut lumen to
mesenteric lymph nodes--and beyond? J Pediatr Gastroenterol
Nutr. 1991;13(4):433434.
98. Piedra PA, Dryja DM, LaScolea LJ Jr. Incidence of catheterassociated gram-negative bacteremia in children with short
bowel syndrome. J Clin Microbiol. 1989;27(6):13171319.
99. Kurkchubasche AG, Smith SD, Rowe MI. Catheter sepsis in
short-bowel syndrome. Arch Surg. 1992;127(1):2124; discussion 2425.
100. Weber TR. Enteral feeding increases sepsis in infants with
short bowel syndrome. J Pediatr Surg. 1995;30(7):10861088;
discussion 10881089.
101. Moukarzel AA, Haddad I, Ament M, et al. 230 patient years of
experience with home long-term parenteral nutrition in childhood: natural history and life of central venous catheters. J
Pediatr Surg. 1994;29(10):13231327.
102. Carlsson E, Bosaeus I, Nordgren S. Quality of life and
concerns in patients with short bowel syndrome. Clin Nutr.
2003;22(5):445452.
103. Candusso M, Faraguna D, Sperli D, Dodaro N, et al. Outcome
and quality of life in paediatric home parenteral nutrition.
Curr Opin Clin Nutr Metab Care. 2002;5(3):309314.
104. Heine RG, Bines, JE. New approaches to parenteral nutrition in infants and children. J Paediatr Child Health.
2002;38(5):433437.
105. Botha JF, Grant WJ, Torres C, et al. Isolated liver transplantation in infants with end-stage liver disease due to short bowel
syndrome. Liver Transpl. 2006;12(7):10621066.
322
106. Diamond IR, Wales PW, Grant DR, Fecteau A. Isolated liver
transplantation in pediatric short bowel syndrome: is there a
role? J Pediatr Surg. 2006;41(5):955959.
107. Horslen SP, Sudan DL, Iyer KR et al. Isolated liver transplantation in infants with end-stage liver disease associated with
short bowel syndrome. Ann Surg. 2002;235(3):435439.
28
Pulmonary Disorders
Allison Mallowe, RD, LDN, Suzanne Michel, MPH, RD, LDN, and Maria Mascarenhas, MBBS
Learning Objectives
CONTENTS
Cystic Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323
Pathophysiology
Nutrition Support. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Pancreatic Enzymes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Eating Behaviors and CF. . . . . . . . . . . . . . . . . . . . . . . . . .
Nutrition-Related Cystic Fibrosis Complications. . . . . . .
329
330
330
331
Summary. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 334
Cystic Fibrosis
Pathophysiology
Cystic fibrosis (CF) is a common autosomal recessive
genetic disorder that is most frequently seen in people of
northern European descent. In the United States CF occurs
in approximately 1 in 2,500 live births and affects approximately 30,000 Americans. The CF gene was isolated in
19891 and since then more than 1,500 mutations have been
identified. The gene, which is localized on the short arm of
chromosome 7, is called the cystic fibrosis transmembrane
regulator (CFTR) and controls the flow of sodium and
chloride ions across the cell membrane. Mutations in the
gene result in abnormal epithelial ion transport in multiple
organs in the body. Clinical outcomes have been related to
the type of mutation. With the advent of almost universal
newborn screening programs in the United States, it is estimated that the average life expectancy will increase from the
current median predicted life expectancy of 37.4 years. 2
Diagnosis during the newborn period by newborn
screening results in early nutrition intervention and
improved nutrition status, which may lead to better quality
of life and prolonged survival. (See Table 28-1 for clinical
features and diagnosis of CF.) Historically an abnormal
323
324
Meconium ileus
Meconium peritonitis
Intestinal atresia
Recurrent obstructive respiratory
disease/infections
Rectal prolapse
Failure to thrive
Obstructive jaundice
Hyponatremic dehydration
Malabsorption
Salty taste in sweat (when
kissed)
Zinc deficiency
Fat-soluble vitamin deficiency
PULMONARY DISORDERS
Nutrition Assessment
325
Parameter
Anthropometrics
Occipital-frontal head
circumference
Body weight, height, length,
head circumference
MAMC, TSF
Nutritional Intake
24-hour recall
3-day dietary fat balance
coefficient of
fat absorption < 93% or 85%
in infants
fecal elastase used to define
steatorhhea
Biochemical
CBC with differential
Iron studies
Fat-soluble vitamins
Alpha-tocopherol
Serum plasma retinol
25-OH vitamin D
PIVKA II
Essential Fatty Acid Deficiency
Ratio of triene to tetraene
25-OH vitamin D
Diagnostic
Yearly
Nutrition Recommendations
Macronutrients
Energy
As previously described, individuals who have CF have
increased energy needs. Exact caloric prescriptions and
formulas to calculate caloric need are difficult to provide
due to individual patient variation11 but improved weight
status has been found at intakes ranging from 110% to 200%
of energy needs for the healthy population.12 Energy needs
of each patient should be assessed on an individual basis and
reflect the pattern of weight gain and fat stores.13 An estimation of individual caloric need is based upon nutrition status,
growth pattern, fat stores, current dietary intake, degree of
fat malabsorption, clinical status (including pulmonary
2010 A.S.P.E.N. www.nutritioncare.org
326
Micronutrients
All persons with CF, both PI and using PERT and PS,18
require supplementation with micronutrients. Deficiencies
of fat-soluble vitamins19 and zinc20 have been demonstrated
in infants diagnosed through newborn screening and do
not correct without appropriate supplementation and, if
indicated, PERT. Children and adolescents are at risk for
micronutrient deficiencies due to:14
Inadequate intake
Malabsorption possibly due to suboptimal PERT
Malabsorption due to residual or incomplete bile salt
absorption
Poor clinical status and poor lung function
Increased utilization and reduced bioavailability
Liver disease
Bowel resection
Late diagnosis of CF
Poor adherence to or inappropriate supplementation.
Fat-Soluble Vitamins
Multivitamins designed to meet the fat-soluble vitamin
needs of persons who have CF (CF-specific multivitamins)
are available in North America (Table 28-3). These vitamins
contain fat- and water-soluble vitamins and zinc. The content
reflects the recommendations provided in the U.S. Pediatric
Nutrition Consensus Report,13 the European Nutrition
Consensus Report,21 the U.S. Bone Consensus Report,22
and/or current research. Dosage and form of the CF-specific
multivitamin supplementation is dependent on results of
laboratory evaluation of vitamin levels and the patients age.
Patients may require additional single-nutrient supplements
(ie, vitamins A, E, D, K), if blood levels cannot be maintained on the CF-specific multivitamins. If the CF-specific
multivitamin is unavailable, single-vitamin supplements are
necessary to make up the difference in content of traditional
multivitamins compared to recommendations.
For additional information specific to vitamins and
CF the reader is referred to www.SourceCF.com for
newsletters.
PULMONARY DISORDERS
327
CFF Consensus
Report 2
012 months
13 years
1,500
5,000
48 years
5,00010,000
918 years
10,000
> 18 years
8000
012 months
13
4050
80150
48
100200
918 years
200400
> 18 years
400
012 months
13
4004
8004
48 years
800
918 years
800
> 18 years
800
012 months
13
300500
300500
48 years
300500
918 years
300500
> 18 years
1,000
012 months
13 years
48 years
9 yearsAdult
SourceCF
Drops, Chewables,
Softgels
ADEK Chewables
AquADEKs
Drops, Softgels
Vitamax
Drops, Chewables
Poly-Vi-Sol Drops
Centrum
Chewable, Tablet
5 (1 mL)
10 (2 mL)
200 / chewable
30 / chewable
400 / 2 chewables
60 / 2 tablets
400 / 2 chewables
60 / 2 tablets
400 (1 mL)
800 (2 mL)
400 (1 mL)
800 (2 mL)
400 / chewable
400 / chewable
800 / 2 chewables
800 / 2 tablets
800 / 2 chewables
800 / 2 tablets
300 (1 mL)
600 (2 mL)
0
0
200 / chewable
10 / chewable
400 / 2 chewables
50 / 2 tablets
400 / 2 chewables
50 / 2 tablets
7.5 (1 mL)
15 (2 mL)
0
0
7.5 / chewable
15 / chewable
15 / 2 chewables
22 / 2 tablets
1. The content of this table is as of December 2008. Products also contain a full range of water-soluble vitamins. Information not included in this table.
Fora copy of the full table, go to: www.SourceCF.com.
2. Cystic Fibrosis Foundation. Pediatric Nutrition for Patients with Cystic Fibrosis Consensus Conference Report March 2001 (Table 7).
3. Also contains mixed tocopherols.
4. Based on Guidelines to Bone Health and Disease in Cystic Fibrosis Consensus Conference Report June 2002 (Figure 2).
5. ADEK is registered trademark of Axcan Pharma Inc., AquADEKs is a registered trademark of Yasoo Health Inc., Vitamax is a registered trademark of
Shear/Kershman Labs. Inc., Poly-Vi-Sol Drops is a registered trademark of Mead Johnson and Company, Centrum is a registered trademark of Wyeth
Consumer Care.
Table reproduced with permission of SourceCF Inc., a subsidiary of Eurand Pharmaceuticals, Inc.
2010 A.S.P.E.N. www.nutritioncare.org
328
Vitamin A
Water-Soluble Vitamins
Deficiency of water-soluble vitamins in CF is rare, but
has been reported in patients not receiving multivitamins
and/or receiving medications interfering with B vitamins. 30 Over time, patients who have had a resection of the
terminal ileum will develop vitamin B12 deficiency requiring
supplementation.14
Minerals
Sodium Chloride
Persons who have CF lose excessive sodium in their sweat
and require supplementation throughout the year, especially
in the summer months. Inadequate salt intake can be life
threatening and/or result in poor appetite with subsequent
poor growth. All infants who have CF and an elevated sweat
test are to be supplemented with 12.6 mEq (1/8 teaspoon)
of salt daily from birth to 6 months of age at which time the
dose is increased to 25.2 mEq (1/4 teaspoon) daily.17 The
salt is added in small, frequent amounts to baby formula or
applesauce used to dose PERT until the daily dose is met.
Salt supplementation is continued until the child is eating
a diet rich in salt and added salt. Children and adolescents
are to be counseled to consume salt, over and above their
usual intake, when participating in physical activity. It may
be necessary to add salt to sports drinks to meet sodium
chloride needs (1/8 teaspoon to 12 oz). 31
Zinc
Pancreatic insufficient infants, prior to treatment with
PERT, lose excessive zinc in their stool. Acrodermatitis
enteropathica like rash, which resembles a severe rash in
the diaper area and the perioral area, is a clinical symptom
of zinc deficiency and is treated with PERT and zinc supplementation. More subtle symptoms of zinc deficiency which
can be seen at any age include lack of appetite, dysgeusia,
poor growth, and compromised immunity. Laboratory
studies to determine zinc status are generally uninformative.
When zinc deficiency is suspected the usual supplemental
dose is 1mg elemental zinc per kilogram daily for 6 months
in addition to that contained in the patients daily multivitamin. Over-the-counter infant vitamin drops do not contain
zinc. Over-the-counter childrens chewable vitamins and
adult formulations contain zinc as do all of the CF-specific
multivitamins. When choosing a zinc preparation, it is
important to note which zinc salt is in the chosen product so
that the correct dose can be determined. For example, zinc
sulfate is 23% elemental zinc but zinc gluconate is only 14%
elemental zinc.
PULMONARY DISORDERS
Iron
Iron status in the general population is affected by a
number of factors, including dietary intake, blood loss, and
medications. For persons with CF, all of these factors, plus
malabsorption, hemoptysis, short bowel syndrome, bacterial
overgrowth, liver and renal diseases, and chronic inflammation contribute to altered iron status. Patients can have iron
deficiency anemia, anemia of chronic disease or a combination of both. Prior to prescribing iron supplementation the
form of anemia must be defined. 32 Assessing iron status in
CF is complicated by chronic inflammation. Serum ferritin,
an acute-phase reactant, may be falsely elevated in CF, thus
masking iron deficiency anemia. Measurement of soluble
transferring receptor levels may be helpful in diagnosis.
Calcium
It is important to optimize the calcium intake of persons
who have CF. In lieu of specific calcium recommendations
for person who have CF, the RD should refer to those in the
DRI. The CFF Bone Consensus Report may be referred
to for more detail regarding bone health and CF. 22 At a
minimum, the adequate intake levels should be achieved in
all patients.
Magnesium
Patients receiving aminoglycosides may require supplemental magnesium. Blood levels should be monitored. 33
Fluoride
CF-specific vitamins do not contain fluoride; therefore the
patients primary care physician may need to prescribe a
supplement if local water is not fluoridated.
Assessment
Blood levels of fat-soluble vitamins should be measured at
diagnosis for patients diagnosed greater than 1 year of age
and annually thereafter.13 For newly diagnosed infants, it is
recommended that levels of vitamins E, A, and D be assessed
1 to 3 months after starting supplementation and annually
thereafter.17 Prothrombin time is insensitive to vitamin K
deficiency in CF.27 PIVKA II provides a better indicator of
vitamin K nutrition, but laboratory reference standards for
full-term infants are not available, and therefore checking
PIVKA II levels is not recommended in the newborn period.
Serum electrolytes and complete blood count is measured
at 2 to 3 months of age and annually or as indicated.17 For
children and adolescents assessment of fat-soluble vitamins, including PIVKA II and CBC with differential, is
done at diagnosis and annually, unless otherwise indicated.
329
Nutrition Support
There is a positive correlation between pulmonary function and weight.12 The CF Foundation recommends the
following nutrition goals:
Infants: weight for length at the 50th percentile by 2
years of age
Children 2 to 20 years of age: BMI percentile at or
above the 50th percentile
Adults: BMI of 22 for women and 23 for men12
Additionally, children are expected to meet their
genetic potential for growth. Therefore, nutrition interventions to reach and maintain weight and growth goals are
often necessary. Behavioral interventions for parents, caregivers, and patients may also help improve caloric intake,
thus weight gain. 34 Weight percentiles are believed to be at
their lowest during the first 2 years of life and early adolescence therefore making aggressive nutrition intervention
important during these time periods. 35 For toddlers and
older children, initially using high-calorie oral supplements or additives may be indicated. Infant formula can
be concentrated or high-calorie modulars added to provide
adequate calories. However in some children this may not
be sufficient to sustain a desired weight and growth status
and enteral feeding may be indicated. There are no specific
guidelines regarding when to initiate enteral feeding in the
pediatric CF population. The CF Foundation Clinical Practice Guidelines Subcommittee on Growth and Nutrition
has determined that for children with growth deficits, oral
or enteral supplements should be used to improve the rate
of weight gain.12 Enteral tube feeding can be via nasogastric
tube, gastrostomy tube, or jejunostomy tube. The nasogastric tube can be inserted and removed daily for short-term
use. Adherence to this intervention may prove difficult. A
gastrostomy tube may be more appropriate and allow for
flexibility of feeding. Feeding via the gastrostomy tube can
be intermittent, bolus, or continuous feeds. Continuous
feeds generally occur nocturnally. Nighttime feedings
allow for regular meal and snack pattern during the day.
Enteral tube feeding may be complicated by early morning
fullness and vomiting, loss of appetite, and poor body image
and self-esteem concerns. The reader is directed to papers
related to these concerns. 3537 It is important to strike a
balance between initiation of enteral feeding and severity
of lung disease to optimize the benefit of the intervention.
Feeding via a jejunostomy tube is infrequent in patients
2010 A.S.P.E.N. www.nutritioncare.org
330
Pancreatic Enzymes
PULMONARY DISORDERS
331
CF-Related Diabetes
CF-related diabetes (CFRD) shares features of both type
1 and type 2 diabetes, but it is a distinct clinical entity.43
Diabetes occurs when people either are insulin deficient or
insulin resistant. Individuals with CFRD are insulin deficient as a result of destruction by fibrosis or scarring of beta
cells in the pancreas that produce insulin.44 Glucose metabolism is strongly influenced by factors unique to CF, including
undernutrition, chronic and acute infection, elevated energy
expenditure, glucagon deficiency, malabsorption, abnormal
intestinal transit time, and liver dysfunction. These factors
are not static and may fluctuate over time in CF.43
Retrospective studies have shown that pulmonary
decline and weight loss begin 2 to 4 years before diagnosis of CFRD.43 Symptoms of CFRD include polydipsia,
polyuria, weight loss or inability to gain weight despite
aggressive nutrition intervention, poor growth, and poor
progression of puberty or unexplained chronic decline in
pulmonary function.43 Any patient with these symptoms
should be screened for CFRD using the CF Foundation
recommendation.43
In the routine management of CF, casual glucose levels
are measured annually. If the person has a random blood
glucose level of 126 mg/dL, further workup for CFRD
should be conducted. Tests include fasting blood glucose or
2-hour oral glucose tolerance test (OGTT). The CF Foundation assembled a consensus conference on CFRD and
issued recommendations for monitoring glucose intolerance; refer to these recommendations for further screening
of CFRD.43
When discrepancies exist between the nutrition
recommendations of the 2 diseases, the CFRD recommendations supersede those for type 1 and type 2 diabetes.
The CF Foundation Consensus Statement on Diagnosis,
Screening, and Management of Cystic Fibrosis Related
Diabetes Mellitus43 currently promotes these 2 principles
2010 A.S.P.E.N. www.nutritioncare.org
332
Lung Transplantation
Lung transplantation remains the most aggressive therapy
for end-stage lung disease.45 Persons with CF are candidates
for lung transplantation when they exhibit severely reduced
lung function and progressive deterioration in their health
and quality of life.16 For children and adults, the largest
obstacle to long-term survival remains chronic allograft
rejection secondary to the development of bronchiolitis
obliterans. Common weight criteria for adults are 80% to
130% IBW or BMI of 18.5 to 30 kg/m. There is no pediatric
weight criterion. Further research in the area of weight and
nutrition status is needed in the pediatric lung transplant
population.
Gastrointestinal
Gastrointestinal (GI) manifestations are frequently seen
in persons with CF and involve all parts of the GI tract.46
Gastroesophageal reflux (GER) occurs at all ages and the
incidence varies between 25% and 100% depending on the
study. Complications of GER include feeding disorders,
decreased caloric intake, failure to thrive, apnea in infants,
vomiting, esophagitis, worsening of lung disease, esophageal strictures, and Barretts esophagus. Reflux can be a
significant problem in lung transplant recipients and is associated with rejection in the posttransplant period. Often a
fundoplication is recommended in the pretransplant period.
Gastroparesis and constipation can be seen in CF and the
latter may be 3 times more frequent than DIOS. Bacterial
overgrowth occurs in up to 60% of patients.47 Predisposing
factors include frequent antibiotic use, intestinal dysmotility, history of previous GI surgery, and sticky intestinal
secretions that trap bacteria. DIOS is seen in all persons with
CF, not just in those with PI. Dehydration of the intestinal
secretions, coupled with electrolyte imbalances and sticky
mucus, and poor motility lead to the accumulation of stool
2010 A.S.P.E.N. www.nutritioncare.org
Bronchopulmonary Dysplasia
Bronchopulmonary dysplasia (BPD) is a form of chronic
lung disease that develops in preterm infants given positive
pressure ventilation and oxygen. The pathophysiology is
complex and due to small airway damage, abnormal alveolar
development, and decreased surface area for gas exchange.48
Additionally there is damage to small blood vessels in the
lungs and secondary damage to the heart and brain. BPD is
most commonly seen in preterm infants with a birth weight
of more than 1250 g and 30 weeks gestational age. Males
tend to be more affected. Surfactant therapy is used soon
after birth to prevent lung damage. Energy requirements
are increased and are in the range of 125 to 150 kcal/kg/d.49
Vitamin A supplementation is important and often used in
the neonatal intensive care unit to prevent BPD. Vitamin E
supplementation is not helpful. These patients also have large
insensible water losses and need extra fluid which results in
PULMONARY DISORDERS
Asthma
Asthma or RAD is the most common cause of hospital
admissions in children. It is a chronic pediatric lung
disease where there is chronic inflammation of the
airways involving eosinophils, mast cells, T lymphocytes,
macrophages, neutrophils, and epithelial cells. Additionally
there is variable air flow obstruction and increased bronchial responsiveness to a variety of environmental stimuli.
The presence of airway edema and mucus contributes to the
obstruction and bronchial reactivity that is seen. 50 Environmental triggers affect the normal development of the
respiratory and immune systems in genetically predisposed
individuals. 51 Patients often require chronic inhaled or
pulses of systemic corticosteroids. Excessive oral cortico
steroid use over time can result in growth failure, fluid and
sodium retention, a voracious appetite, excessive weight
gain, hypertension, glucose intolerance, and obesity. There
appears to be considerable variation in the side effects of
inhaled corticosteroids. Growth, puberty and bone health
can be affected depending on the duration and dose. 52
Early data suggest the use of antioxidants, Lactobacillus,
333
and omega-3 fatty acids has been associated with a reduction in symptoms and reduced development of asthma in
those with atopy. 52 In children a linear association between
obesity (increased BMI) and asthma has been noted with a
6% increase in prevalence per unit increase of BMI. 53
Technology Dependent
Children with chronic respiratory failure may require
chronic ventilatory support. At-risk children include those
with: neuromuscular dystrophy, spinal muscular atrophy,
Duschennes muscular dystrophy, spinal cord injuries,
BPD, congenital diaphragmatic hernia, severe lung
malformations, congenital hypoventilation syndrome,
and myelomeningiocoele. From the above diagnoses, of
patients who require ventilator support, patients with
BPD and neuromuscular dystrophy are the most frequent.
Respiratory support can be non-invasive (BIPAP) or invasive (tracheostomy and ventilator). The pathophysiology
includes respiratory insufficiency (seen in BPD and pulmonary hypoplasia), secondary damage to the lungs from
severe cardiac disease resulting in decreased surface area,
decreased oxygen absorption, increased carbon dioxide
retention, poor lung development, small-volume lungs,
and insufficient vascular bed. There may also be decreased
central drive for respiration as seen in persons with congenital central hypoventilation syndrome. Since these patients
have decreased work of breathing, their energy requirements are subsequently decreased. If careful attention is
not paid to their nutrition regimens, they can have excessive weight gain which can further impact their respiration
status negatively. Often, in an attempt to decrease calories,
overall nutrient intake, especially protein and mineral
intake, suffers. These patients may also be at risk for bone
disease due to decreased weight bearing and micronutrient
deficiency (vitamin D and calcium).
Nutrition Assessment
For children with BPD and asthma it is best to monitor
growth on a regular basis. Anthropometrics provide critical
information regarding the growth of the infant or child. In
the infant with BPD, factors that may increase caloric requirements include increased basal metabolic rate, increased
work of breathing, chronic illness or infections, and respiratory distress or metabolic complications.54 The infant may
struggle with fluid sensitivity, which may limit the intake of
calories and nutrients. Infants may have interrupted feeding
skill development, therefore may be poor oral feeders.54
334
Nutrition Recommendations
In critically ill infants with BPD, efforts are made to prolong
life. Caloric provision is often relegated to secondary
importance, compared to pulmonary edema, reduced
cardiac output, and electrolyte imbalance. 54 Please refer
to the section on nutrition in the neonatal intensive care
unit for more detailed information on caring for the critically ill infant. As the infants condition improves, concerns
regarding fluid overload and thermal losses remain.
Continued monitoring of intake, growth, and development
is essential. Individualizing macronutrient needs based on
the patients growth in relation to the goal is best. Specific
to the infant with BPD, calcium and phosphorous may be
further compromised by diuretic therapy, steroid therapy,
long-term use of parenteral nutrition, and feeding delays. 54
For further information on micronutrients, please refer to
the vitamin and mineral chapters (Chapters 6, 7, and 8).
A diet providing appropriate amounts of macro- and
micronutrients based on the U.S. Department of Agriculture dietary guidelines for age is recommended. Increased
appetite, abdominal fat accumulation, sodium and fluid
retention, and steroid-induced glucose intolerance are
common side effects of oral corticosteroid therapy. Monitoring weight and linear growth on a regular basis is
important. Nutrition support is discussed in Chapter 34.
Summary
References
PULMONARY DISORDERS
335
336
46. Mascarenhas MR. Treatment of gastrointestinal problems in cystic fibrosis. Curr Trea Options Gastroenterol.
2003;6(5):427441.
47. Fridge JL, Conrad C, Gerson L, Cox K. Risk factors for small
bowel bacterial overgrowth in cystic fibrosis. J Pediatr Gastroenterol Nutr. 2007;44(2):212218.
48. Rajiah P. Bronchopulmonary dysplasia [emedicine website].
June 9, 2006. Available at http://emedicine.medscape.com/
article/406564. Accessed December 2008.
49. Cox JH. Bronchopulmonary dysplasia. In: Groh-Wargo S,
Thompson M, Cox JH, eds. Nutritional Care for High-Risk
Newborns. Chicago, IL: Precept Press Inc; 2000.
50. Morris MJ. Asthma [emedicine website]. July 10, 2008. Available at http://emedicine.medscape.com/article/29630.
Accessed December 2008.
51. Sly PD. Asthma: Disease mechanisms and cell biology. In:
Taussig LM, Landau LI, eds. Pediatric Respiratory Medicine.
2nd ed. Mosby Elsevier; 2008;791804.
52. Landau LI, Martinez FD. Asthma: Treatment. In Pediatric Respiratory Medicine. 2nd ed. Mosby Elsevier;
2008:829844.
53. Sithole F, Douwes J, Burstyn I, et al. Body mass index in childhood: a linear association. Asthma. 2008;45(6):473477.
54. Queen Samour P, King K. Pulmonary diseases. In: Queen
Samour P, King K. Handbook of Pediatric Nutrition. 3rd ed.
Sudbury, MA: Jones and Bartlett Publication;2005:307349.
Organ Transplantation
29
Anita Nucci, PhD, RD, LD, Sharon Strohm, MBA, RD, LDN, Neelam Katyal, MS, RD, LDN, and Beth Lytle, RD, LDN
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 337
Indications for Organ Transplantation . . . . . . . . . . . . . . . 338
Nutrition Assessment Before Transplant . . . . . . . . . . . . . 339
Anthropometric Assessment
Physical Examination
Biochemical Tests
Macro- and Micronutrient Requirements
Feeding History
Learning Objectives
Introduction
338
Diagnoses
Intestine
Gastroschisis
Midgut volvulus
Necrotizing enterocolitis
Intestinal pseudoobstruction
Biliary atresia
Acute hepatic necrosis
Metabolic disease
Aplasia/hypoplasia/dysplasia
Obstructive uropathy
Focal segmental glomerular sclerosis
Congenital disease
Cardiomyopathies
Cystic fibrosis
Congenital heart disease (primarily those < 1 year of age)
Primary pulmonary hypertension
Liver
Kidney
Heart
Lung
ORGAN TRANSPLANTATION
339
Physical Examination
Along with anthropometric measurement, a child should
also be given a physical examination. This preliminary look
may then lead to more detailed evaluation, if necessary. Hair
that is sparse or easily breakable may indicate malnutrition.
Dry skin may be a sign of vitamin A or folic acid deficiency
and skeletal changes may point to problems with vitamin D
or calcium metabolism.
Biochemical Tests
Monitoring of laboratory tests helps the clinician to adjust
the provision of nutrients and electrolytes in the diet,
enteral feedings, intravenous replacement fluids, and/or PN
both pre- and posttransplant. Table 29-2 shows the basic
biochemical tests that are often monitored before transplant
by type of solid organ.913
Table 29-2 Biochemical Tests Monitored Before Transplant by Type of Solid Organ913
Biochemical Tests
Minerals
Other
Intestine
Liver
Kidney
X
X
X
X
X
X
X
X
Heart
Lung
X
X
X
X
X
X
X
X
X
X
X
X
X
X
A
E
D (25 and 1,25 OH)
B12
RBC folate
Zn
Cu
Se
Mn
Carnitine
X
X
X
X
X
X
X
X
A
E
D (25-OH) if no renal impairment
D (1,25 OH) if renal impairment
PTH
X
X
A
E
D (25-OH)
Glycosylated hemoglobin
340
Feeding History
A complete nutrition assessment should also include a
history of enteral feeding tolerance and current eating
habits. Children with chronic diseases have often tried
many different types of infant and enteral formulas before
choosing the most tolerated option. It is important to note
the type of formula and route and the percentage of calories
that are contributing to the childs total calorie intake when
providing both EN and PN.
Infants and children who receive long-term enteral
feedings are also at risk for oral aversion due to the absence
of oral feeding.9 Normal feeding and swallowing development may have been missed. For children who can swallow,
oral feeding of small amounts of varied food tastes and
textures is encouraged. Maintenance of oral stimulation
may help in the posttransplant period when the patient is
transitioning from enteral to oral feedings. Children with
oral aversion or other feeding issues should be referred to a
feeding clinic as soon as possible.17 A Video Feeding Study
may be needed to evaluate the safety of the child to take oral
fluids and solids.
ORGAN TRANSPLANTATION
341
Liver
Nutrition Requirements
Postoperative nutrition requirements for liver transplant
recipients should be designed to provide sufficient calories to minimize catabolism and prevent complications
from pretransplant nutrition issues. Biochemical parameters should be monitored and nutrition therapy adjusted
as needed.24 A postoperative liver transplant patient may
require PN if he or she is malnourished, has had complications, or if a lengthy recuperation is expected. During the
immediate postoperative period, PN should be infused
continuously.14 Guidelines for initiating postoperative PN
are shown in Table 29-3.18,24,25
A tube feeding may provide total enteral nutrition or be
used in conjunction with the oral route if intake is suboptimal. Breast milk is always preferred, if available. An intact
protein, age-appropriate formula should be used to start,
with a change to a hydrolyzed protein or a free amino-acid
hypoallergenic formula if an intolerance should occur. The
caloric density of formulas may need to be manipulated
depending upon fluid restrictions and caloric requirements. Initially the tube feeding should run continuously
until the oral diet is advanced. As the oral diet is advancing,
nocturnal enteral feedings or daytime bolus feedings may
need to be considered to support nutrition requirements. In
addition, a daily multivitamin supplement for age should be
provided.24 The posttransplant oral diet may be described
as a healthy diet for age with careful consideration of the
current Food Pyramid as well as the 2005 Dietary Guidelines
for Americans.26,27 The protein, fat, and carbohydrate content
of the diet should follow these guidelines with additional
consideration given for complications such as renal impairment, hypertension, hyperkalemia, and diabetes mellitus.
Calcium should be supplemented if intake is insufficient.
342
Table 29-3 Guidelines for Initiating Parenteral Nutrition after Liver Transplant18,24,25
Nutrient
Guidelines
Calories
Avoid overfeeding
Initially provide 120%130% of resting energy expenditure to meet postoperative transplant needs
Pre-existing nutrition needs, surgical complications, wound healing, sepsis, and rejection may further necessitate additional calories
Provide 15%20% of total calories as protein
Infants: 33.5 g/kg dry body weight
12 years: 2.53 g/kg dry body weight
313 years: 1.52.5 g/kg dry body weight
Adolescents: 11.5 g/kg dry body weight
Additional consideration should be given to renal function and wound healing
Trophamine (B. Braun, Irvine, CA) is the preferred amino acid source due to its branched chain amino acid content and lower pH
for maximum solubility of calcium and phosphorus (cysteine is added only for children < 1 year of age)
Provide 30% of total calories as fat
20% intravenous fat is preferred for its caloric density
Provide 50%55% of total calories as carbohydrate
Begin at same concentration as the initial intravenous fluid and advance glucose as tolerated to a maximum of:
Term Infants: 14 mg/kg/min
110 years: 11 mg/kg/min
1118 years: 8.5 mg/kg/min
Provide standard intravenous multivitamin for age
Provide parenteral trace mineral solution based on weight
Provide full amount of trace minerals in patients without liver dysfunction
Provide dose trace minerals + full amount of zinc with liver dysfunction secondary to cholestasis
Zinc should be supplemented at 1 times the DRI for age if wound healing is an issue
Based on weight, renal and cardiopulmonary function
Maintenance fluids:
110 kg dry body weight: 100 mL/kg/d
1020 kg dry body weight: 1000 mL + 50 mL/kg for each kg over 10
2030 kg dry body weight: 1500 mL + 20 mL for each kg over 20
Protein
Fat
Carbohydrate
Multivitamins
Trace Minerals
Fluid
Feeding Disorders
Advancement of the oral diet may be challenging in some
patients postoperatively. Oral aversion may occur in those
who required long-term PN, EN, and mechanical ventilation. These patients may require consultation from
an occupational and/or speech therapist. Patients who
required special diet restrictions before transplant (eg,
protein restriction) may continue to prefer the taste of their
restricted diet. Temporarily, these patients may continue
their pretransplant medical nutrition enteral formulas until
they readily incorporate a variety of foods in their diet.
Growth and Development
Long-term goals for patients after liver transplant include
nutrition and life-style factors. Optimization of linear
growth and physical development as well as socialization
and participation in daily activities are desired. Calorie
requirements will vary with age, activity, and growth rate.
Children who exhibit linear growth impairment will need
2010 A.S.P.E.N. www.nutritioncare.org
calorie goals based on the DRI for height age.9 Those without
growth delay may initially consume calories based on the
DRI for age. Growth delay is common after liver transplant
due to the nutrition impact of the original disease and use
of corticosteroids. Catch-up growth for weight occurs more
rapidly than height.1,28,29 Linear growth and weight should
be monitored at primary care physician visits, and transplant centers alerted for any change in growth velocity. 30
Kidney
Nutrition Requirements
The nutrition recommendations for kidney transplant
patients immediately and later after transplant are shown in
Table 29-4. An age-appropriate oral diet can be started once
bowel function has resumed. An enteral tube feeding is
rarely needed after kidney transplant. However, for patients
who were fed via gastrostomy tube prior to transplant, it
may be necessary to continue tube feedings and gradually
ORGAN TRANSPLANTATION
343
Immediately Posttransplant
Calories
Protein
Carbohydrates
Fat
Phosphorus
Calcium
Potassium
Sodium
Iron
Fluids
Vitamins
344
Heart
Nutrition Requirements
The nutrition goals for posttransplant pediatric heart transplant recipients include achieving and maintaining ideal
body weight, limiting sweets and foods high in concentrated sugar, reducing foods high in fat and cholesterol, and
limiting salt.12 Most pediatric heart transplant recipients
progress to a full oral diet within 4 days of transplant. 3942
The exception are those orally aversive infants and children
who were dependent on EN and/or total parenteral nutrition pretransplant. These patients may require additional
nutrition support while receiving intensive oral rehabilitation therapy.
Dietary Modification
Alterations in diet may be necessary due to side effects of
medications and immunosuppression therapy. Hypertension, hyperglycemia, and weight gain are common in
posttransplant pediatric heart recipients.12 Lipid abnormalities have been reported at 1 year posttransplant.43
Calcineurin inhibitors may create these elevated serum
lipid levels, and a heart-healthy, low-cholesterol, low-saturated fat diet is recommended. 39
Osteoporosis is universal among pediatric posttransplant recipients. 39 The combined effects of nutrition status
before transplant along with calcineurin inhibitors and
steroids result in decreased calcium absorption and bone
formation. Supplemental vitamin D (400800 International
Units) and calcium (12001500 mg) are recommended. 39
In addition, an annual dual-energy x-ray absorptiometry
(DEXA) scan may be warranted in some cases. There
are little data on the use of biphosphates, calcitonin, and
hormone replacement therapy in the pediatric posttransplant population.
Pretransplant growth parameters will impact the
amount of catch-up growth needed posttransplant.4445
Infants and children have demonstrated appropriate growth
velocity and weight gain posttransplant. Weight gain with
lack of linear catch-up growth is characteristic of the adolescent heart transplant population. 39
Lung
Nutrition Requirements
The post-lung transplant nutrition goals are to provide
energy to prevent weight loss, promote healing, and prevent
infection while minimizing gastrointestinal complications
and avoiding drug-nutrient interactions. Calorie needs are
increased due to the bodys effort to fight infection and
promote wound healing. In contrast, the body uses less
energy due to the decreased work of breathing and limited
activity level immediately posttransplant. Considering this
combined effect, calorie requirements are typically 100% to
120% of the DRI for age.14 Protein needs are elevated due
to the healing process after surgery and are two times the
DRI.16
Enteral/Oral Supplementation
After transplant, the patient should be advanced to a regular
diet as soon as tolerated. Oral nutrition supplements may
be encouraged to maximize energy intake. Initially the
patient may not be able to meet calorie needs from oral
intake because of side effects (ie, nausea and vomiting)
from immunosuppressive medications. If the patient loses
10% of his or her admission weight and is not able to meet
caloric needs orally then supplemental tube feeds should be
recommended. Tube feedings have been shown to be more
effective than oral intake for weight gain after transplant.46
The patient often eats orally throughout the day and receives
nocturnal tube feeds. For patients with CF and pancreatic
insufficiency, semi-elemental formula may be necessary. If
calorie and protein needs are not met by oral diet and/or EN
then PN should be initiated.
Pancreatic Enzyme Supplements/Vitamin-Mineral
Supplementation
Patients with CF and pancreatic insufficiency will continue
to have pancreatic disease posttransplant. Pancreatic
enzyme therapy should resume with all meals and snacks.
Previous vitamin and mineral supplementation should
continue. Patients with CF are at risk of developing high
vitamin A and vitamin E serum levels after transplant.
These levels should be monitored because adjustments are
frequently required.47 Magnesium levels tend to be low after
transplant and supplementation may be necessary.
Nutrition Management of Complications
Common complications after lung transplant that require
nutrition intervention include infection and rejection, diabetes, osteoporosis, renal complications, and
ORGAN TRANSPLANTATION
345
Food Safety
Because most transplant recipients are receiving immunosuppressive medications, they are particularly susceptible to
developing a foodborne illness caused by bacteria and other
pathogens that can contaminate food. The major pathogens
that can cause foodborne illness and the most common
sources of contamination associated with these pathogens
are shown in Table 29-5. 54 The United States Department
of Agriculture, Food Safety and Inspection Service recommends the following 4 basic steps to food safety: (1) clean
hands and surfaces often as bacteria can be spread from
surfaces to food; (2) separate meat, poultry, seafood, and
eggs from ready-to-eat foods to avoid cross-contamination;
(3) cook foods to proper temperatures; and (4) chill foods
promptly. In addition, it is important to adhere to the manufacturer Sell-by and Use-by dates when purchasing and
consuming perishable foods. When eating out, transplant
recipients should avoid any food that contains uncooked
ingredients and speak to the food preparer to ensure that
foods have been cooked to a proper minimum internal
temperature. In general, buffets should be avoided. When
traveling, gel packs should be used to keep food cold (40oF
or below) as well as insulated containers to keep cooked
food hot (140oF or above). 54
346
Bacteria
Campylobacter
Escherichia coli
Listeria monocytogenes
Salmonella
Vibrio vulnificus
Viruses
Norovirus
Protozoa
Cryptosporidium
Toxoplasma gondii
Sources
ORGAN TRANSPLANTATION
3. Which of the following supplements are not recommended for children with cystic fibrosis and pancreatic
insufficiency after lung transplantation?
A. Pancreatic enzymes
B. Standard enteral formula
C. Parenteral nutrition
D. Vitamins and minerals
See p. 487 for answers.
References
347
348
30
Nancy Sacks, MS, RD, LDN, Elizabeth Wallace, RD, CNSC, LDN, Seema Desai, MS, RD, LDN, CNSD, Vinod K. Prasad, MD, MRCP
(London), David Henry, MS, BCOP, Virginia Guzikowski, MSN, CRNP, Liesje Nieman Carney, RD, CNSD, LDN, Beth Bogucki Wright, MS,
RD, CSP, LDN, and Susan Rheingold, MD
CONTENTS
General Oncology Overview. . . . . . . . . . . . . . . . . . . . . . . .
Hematopoietic Transplant. . . . . . . . . . . . . . . . . . . . . . . . .
Late Effects of Treatment for
Survivors of Childhood Cancer . . . . . . . . . . . . . . . . . . . . .
Appendix 30-1: Algorithm For Nutritional
Intervention In The Pediatric Oncology Patient. . . . . . . .
Appendix 30-2: Algorithm for Nutritional Intervention
and Categories of Nutritional Status in the Pediatric
Oncology Patient References and Resources . . . . . . .
Appendix 30-3: Categories of Nutritional Status
for the Pediatric Oncology Patient. . . . . . . . . . . . . . . . . .
Appendix 30-4: Gastrointestinal Supportive
Care Medications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Learning Objectives
349
354
362
365
366
367
368
350
Radiation Therapy
Radiation therapy is a treatment used independently or in
conjunction with surgery and chemotherapy. Radiation
destroys the genetic material within the cell, thereby killing
the malignant cell.14 Complications arise from radiation
therapy because healthy cells in the radiation field are inevitably damaged during treatment. The area most susceptible
to nutrition effects of radiation is the GI tract.15 Mucosa
from the mouth to the anus can be damaged causing malabsorption of nutrients, diarrhea, and severe pain both with
and without oral intake. In adults, the effect on appetite is
dependent on the involved radiation field, total dose of radiation, and number of fractions received.16 Acute side effects
of radiation may begin as early as 1 week from the initiation of radiotherapy, and last several weeks after the final
fraction. Patients receiving cranial radiation can develop
postradiation somnolence syndrome 6 to 8 weeks after
completion of radiation, causing severe lethargy and flu-like
symptoms. Expected side effects based upon the location of
the radiation field are found in Table 30-1.
Table 30-1 Nutrition-Related Side Effects of Radiation Therapy
Location
Adverse Reaction
Nausea/Vomiting
Fatigue
Loss of Appetite
Alterations in Pituitary Functions
Growth Failure
Xerostomia
Sore Mouth/Throat
Dysphagia
Mucositis
Altered Taste and Smell
Fatigue
Loss of Appetite
Nausea/Vomiting
Diarrhea
Abdominal Cramping
Bloating
Gas
Enteritis and Colitis
Lactose Intolerance
Fatigue
Loss of Appetite
Chemotherapy
Classic cytotoxic chemotherapy works by inhibiting the
division of rapidly dividing cells.17 Although cancer cells
are the main target, rapidly dividing normal cells including
those of the GI tract, taste buds, hair, and bone marrow are
equally affected. The effect of chemotherapy on the patients
nutrition status is associated with the exact medication used,
dose, route of administration, and length of treatment.
351
Effect on nutrition
Interleukin-1 (IL-1)
Interleukin-6 (IL-6)
Interferon (INF)-g
Peptide YY (PYY)
Ghrelin
Physiological
The toxicity of chemotherapy can cause changes in organ
function resulting in altered nutrient metabolism and excretion. Cisplatin and cyclophosphamide can cause electrolyte
wasting by the kidneys that can last for weeks after the
agent is given. Mild to moderate transaminitis and hyperbilirubinemia are common side effects of chemotherapy,
demonstrating direct effects on hepatic function. Tumors
of the nasopharynx, neck, mediastinum, and GI tract and
organs can cause direct obstruction and lead to decreased
oral intake. Other side effects of therapy leading to suboptimal intake are described in detail above.
Psychological
Emotional well-being is essential to achieving and maintaining physical health, including an optimal nutrition
status. Depression and anxiety affects up to 20% of all
patients diagnosed with cancer and one study in children
with cancer found that 59% had mild psychological problems. 33 While anxiety appears to affect younger children,
and depression older, both can lead to inactivity, loss of
appetite, self-criticism, and hopelessness. Self-image and
self-esteem, including perception of weight status and physical appearance, are important to monitor in the pre-teen
and teenage population. Learned food aversions due to fear
from eating and vomiting seem to affect toddlers and young
children. A specialized feeding program may be necessary
to help overcome food aversions and resume normal oral
intake and eating patterns during or after completion of
treatment.
352
Outcomes Measures
Biochemical Evaluation
Laboratory tests should be closely monitored from initial
nutrition assessment through repletion. One should assess
basic electrolytes (sodium, potassium, chloride, bicarbonate), glucose, renal function (creatinine, blood urea
nitrogen), minerals (calcium, phosphorus, magnesium),
liver function tests (alkaline phosphatase, serum aminotransferases, gamma glutamyl transferase (GGT), total
bilirubin), triglycerides, and cholesterol. Serum albumin
and prealbumin values may reflect protein stores.
Table 30-3 Components of Nutrition Assessment
Medical History
Diagnosis stage and date
Past medical history
Medication history
Anticipated therapy protocol
Anthropometics*
Weight history
BMI/Weight-for-age
Height-for-age
Recent growth trends
% Ideal body weight
% Usual body weight
% Diagnosis weight
Mid-arm circumference
Triceps skinfold
Dietary Intake
Current intake (amounts, stage, feeding times)
Evaluation
Usual intake
Feeding behavior
Modified oral intake
Tube feeds
Parenteral nutrition
Vitamin/Mineral supplementation
Gastrointestinal
Nausea
Symptoms/Side
Vomiting
Effects
Constipation
Diarrhea
Dry mouth
Taste changes
Mouth sores
Difficulty swallowing
Early satiety
Laboratory
Electrolytes
Assessment
Blood glucose
Serum proteins
Absolute neutrophil count
Complete blood count
Liver function tests
Quality of Life
Activity level
Family support system
Depression/Anxiety
Pain
Treatment plan
Resources
* Refer to Appendix 30-1, Algorithm for Nutritional Intervention and Appendix
30-2, Categories of Nutritional Status in the Pediatric Oncology Patient.
Nausea/Vomiting
Anorexia
Diarrhea
Dysgeusia
Mucositis
Xerostomia
353
Oral Nutrition
Oral nutrition is important to continue to promote normalized and developmentally appropriate feeding for children
and should be encouraged as much as is medically reasonable. Patients and families should be educated on the
appropriate strategies to alter nutrient intake using modulars
and supplements. Unfortunately many pediatric patients
on oral diet alone have significant weight loss and muscle
wasting51,52 and need to be supported by other means.
Enteral Tube Feeding
Enteral tube feeding (TF) may significantly enhance a
childs nutrition status during cancer therapy. 5254 Selection
of the correct formula must be determined in order to maximize nutrient intake. Though no large clinical trials have
been conducted, research suggests that a calorie concentrated formula may be more beneficial to malnourished
patients receiving TF. 53 Preliminary evidence regarding
the use of TF as supplementation or full calories, prior to
developing weight loss, shows an overall improvement in
the patients nutrition status at the end of therapy. 55 Tube
feedings can be provided enterally via nasogastric (NG),
nasojejunal (NJ), or a percutaneous endoscopy gastrotomy
(PEG) tube. Benefits of a PEG tube include a one-time
placement in a location that is not apparent, especially in
an appearance-conscious adolescent. It also bypasses the
issue of tube displacement with emesis and traumatic tube
replacement during periods of mucositis. The placement of
2010 A.S.P.E.N. www.nutritioncare.org
354
Hematopoietic Transplant
Hematopoietic stem cell transplantation (HSCT) is performed to replace diseased and defective bone marrow and
restore hematopoietic and immunologic function. It is a
broad category and encompasses bone marrow transplantation (BMT), peripheral blood stem cell transplantation
(PBSCT), and umbilical cord blood transplantation
(UCBT) depending on the source of hematopoietic stem
2.
3.
4.
Transplantation Process
1. Conditioning. The purpose of the conditioning regimen
is to destroy the defective or diseased marrow, kill
cancerous cells, create space for donor cells, and prevent
rejection of donor cells by neutralizing the patients
immune system. Conditioning therapy (also known
as cytoreduction or preparative regimen) consists of a
combination of chemotherapy drugs with or without
radiation and is given over a number of days prior to
transplant. There are 2 types of conditioning regimen:
(1) myeloablative and (2) non-myeloablative. Myeloablative, also known as conventional conditioning
regimen, uses high-dose chemotherapy and/or total
body irradiation that destroys or myeloablates the
bone marrow. The high-dose chemotherapy and radiation causes acute and long-term toxicities including
severe mucositis, myelosuppression, nausea, vomiting,
liver function abnormalities, and cardiotoxicity. The
non-myeloablative conditioning uses a milder form of
5.
6.
355
356
Calories
Protein (g/kg/d)
012 mo
BMR* x 1.61.8
3
BMR x 1.61.8
2.53
16 y
710 y
BMR x 1.41.6
2.4
1114 y
BMR x 1.41.6
2
BMR x 1.51.6
1.8
1518 y
> 19 y
BEE**x 1.5
1.5
*For BMR equation, refer to Appendix 30-2.
**BEE equations:
Male: 66 + (13.7 x wt in kg) + (5 x ht) (6.8 x age)
Female: 665 + (9.6 x wt in kg) + (1.7 x ht) (4.7 x age)
Fluid Needs
In general, daily fluid needs can be assessed using the
Holliday-Segar method.70 However, one must take into
consideration the various factors that impact daily fluid
balance. For example, fluid need is increased with fever, GI
losses (eg, vomiting, diarrhea), mucositis, open skin wounds,
and other factors.67 Liver and kidney dysfunction may lead
to fluid retention and may require fluid restriction. In addition to the total parenteral nutrition, and oral or enteral
intake, patients receive additional fluid with medications
and blood products and these must be taken into account.
Therefore, close monitoring of fluid status is important.
Vitamin and Mineral Needs
Vitamin and mineral requirements for patients undergoing
HSCT have not been determined. The nutrition support
regimen should meet 100% of the dietary reference intake
(DRI) of vitamins and minerals. If oral intake or enteral
nutrition support does not meet the vitamin and mineral
needs, then an iron-free multivitamin and mineral supplement should be provided. Generally, iron supplementation
is not required in HSCT patients because they receive
frequent blood transfusions and supplemental iron can
result in iron overload. The risk of vitamin and mineral
deficiency is particularly higher if the patient has diarrhea,
vomiting, and malabsorption. Thiamin, vitamin K, vitamin
D, calcium, or zinc deficiencies have been seen in some
patients. Vitamin K deficiency as determined by PIVKAII level was seen in 31% of pediatric patients undergoing
HCST and was attributed to use of phenytoin, inadequate
intake, or malabsorption. 54 The investigators did not find a
direct correlation between prothrombin time and vitamin
K status. However, in clinical practice prothrombin time
is a good indicator of significant vitamin K deficiency
and can guide vitamin K therapy. Antibiotic use has been
shown to decrease the production of vitamin K in the body.
HSCT patients receive multiple antibiotic treatments posttransplant; therefore, vitamin K supplementation of 1 mg/
kg should be provided weekly.67,71 In patients with severe
chronic diarrhea, zinc losses may be significant and supplementation may be necessary.
Patients undergoing HSCT often receive multiple
agents that alter bone metabolism as part of their treatment.
These include methotrexate, steroids, cyclosporine, and
total body irradiation. For many patients, physical activity
and exposure to sun is limited and can lead to additional
nutrition problems like osteopenia, vitamin D deficiency,
and eventually poor bone health. Therefore, serum
vitamin D and calcium should be routinely monitored and
357
358
359
Table 30-6 Side Effects of Some of the Drugs Commonly Used During HSCT
Drug
Usage
Side effect
Steroids
Immunosuppressive
Methotrexate
Immunosuppressive
Cyclosporine
Immunosuppressive
Tacrolimus
Immunosuppressive
Sirolimus
Immunosuppressive
Cellcept
Immunosuppressive
Antithymoocyte
globulin (ATG)
Immunosuppressive
Acyclovir
Antiviral
Cefepime
Antibiotic
Amikacin
Antibiotic
Nephrotoxicity
Gentamicin
Antibiotic
Nephrotoxicity
Voriconazole
Antifungal
Amphotericin
Antifungal
Nephrotoxicity
Micafungin
Antifungal
Hepatotoxic
Source: Clinical Pharmacology [database online]. Tampa, FL: Gold Standard, Inc; 2009. http://www.clinicalpharmacology.com. Accessed December 2009.
Frequency
Phosphorus
Ionized calcium
LFTs, albumin, total bilirubin
Prothrombin time
Vitamin D (25 OH D2+D3)
Zinc
Manganese, copper, selenium
Triglycerides
Weights
Input/output
360
posttransplant.89 The same study showed that triceps skinfold (TSF) measurements and midarm circumference
also decreased. Therefore, TSF measurements should be
obtained and monitored. Fluid shifts will not affect skinfold measurement.89 Bone growth can be monitored by
obtaining a dual-energy x-ray absorptiometry (DEXA) scan
bi-annually.90
361
Skin
0
+ to ++
+ to +++
++ to +++
+ to ++++
Liver
0
0
+
++ to +++
++ to ++++
Gut
0
0
+
++ to +++
++ to ++++
Functional Impairment
0
0
+
++
+++
Reprinted with permission from Sullivan KM. Graft-vs-host disease. In: Blume KG, Forman SJ, Appelbaum F, eds. Thomas Hematopoietic Cell
Transplantation. 3rd ed. Oxford: Blackwell Publishing; 2004:635664.
affects the lower GI tract causing severe, high-output diarrhea associated with bleeding and cramping abdominal
pain.99 GVHD of the upper GI tract causes decreased appetite, nausea, and vomiting.79 Active GVHD leads to mucosal
degeneration, malabsorption, and protein loss. Liver GVHD
is characterized by cholestatic hyperbilirubinemia, however
it can be difficult to differentiate other causes of liver function impairment; the differential diagnosis includes SOS,
infection, sepsis, drug effects, iron overload, or PN-induced
cholestasis.98 Acute GVHD is graded based on the extent of
skin, liver, and gut involvement (see Table 30-8 for staging
and grading). Long-term survival (5 years) for grade III
is 25% and grade IV is 5%.100 Prevalence of acute GVHD
varies from 35% to 45% in full matched sibling donors to
60% to 80% in patients with 1 antigen HLA mismatched
transplant.101 With the same degree of HLA mismatch,
patients receiving umbilical cord blood transplant have
lower frequency of acute GVHD (35% to 65% compared to
60% to 80% in patients receiving unrelated donor graft).102
Chronic GVHD
Chronic GVHD involves skin, gut, liver, lungs, eyes, mouth,
and bone marrow. Risk factors for development of chronic
GVHD include the age of the patient and history of acute
GVHD. About 22% to 29% of pediatric patients undergoing
HSCT will develop chronic GVHD.103 In adult patients,
chronic GVHD ranges from 30% to 50% in HLA-matched
sibling transplant.104
A low-fiber, low-lactose, low-fat, bland diet may be
necessary during periods of acute GVHD, but use of this
dietary modification is not the norm.69 PN is warranted when
362
Hemorrhagic Cystitis
High-dose cyclophosphamide can lead to hemorrhagic
cystitis, a complication characterized by bloody urine.
Fluid intake may need to be twice the maintenance needs in
order to irrigate the blood clots in the bladder. Viral infections unique to the immuno-compromised patient, which
include adenovirus and BK virus, can also lead to hemorrhagic cystitis.67,87,108
Sinusoidal Obstruction Syndrome
SOS is usually seen between 1 to 4 weeks posttransplant
and reflects regimen-related toxicity.110 Injury to the
endothelial cells of the sinusoids, thickening of the hepatic
venules due to edema, and deposition of fibrin, factor VII,
and blood cell fragments leads to narrowing and increased
resistance to the blood flow through the venules. This causes
hepatic congestion and portal hypertension.111 Weight gain,
hyperbilirubinemia, ascites, right upper-quadrant pain,
and hepatomegaly are the clinical symptoms associated
with SOS. Severe SOS can lead to liver failure, hepatorenal
syndrome, portal hypertension, and multiorgan failure.
Pre-existing liver dysfunction, previous transplantation,
abdominal irradiation, conditioning regimen, adrenoleukodystrophy, and neuroblastoma are some of the risk factors
for SOS.112 The incidence of SOS in children after HSCT
ranges from 5% to 40%,112 and the mortality rate in severe
SOS has been reported to be as high as 47%.113
Conclusion
Nutrition assessment, support, and monitoring during
the transplant process are critical parts of patient management. These measures are even more important in pediatric
patients because of small size, need for continuing growth
and development, and the potential for rapid deterioration.
Comprehensive assessment may include information about
anthropometrics, food intake, appetite, presence of infection, organ dysfunction, wounds, nausea, vomiting, diarrhea,
and mucositis amongst others. Good clinical judgment is
critical and decision making should be individualized in an
effort to provide optimal nutrition management.
Bone Health115
Bone is a living growing tissue (206 bones in the body) made
up of calcium, phosphorus, magnesium, vitamin D, and
fluoride. All of these nutrients are important in the development and maintenance of bone and other calcified tissues. A
consequence of childhood cancer treatment is a decrease in
peak bone mass from normal levels and an increased loss of
calcium from the bones. Survivors are at increased risk for
osteoporosis, a result of poor bone formation or too much
bone loss; therefore fractures may occur as bones become
weaker. Osteoporosis is diagnosed by an X-ray technique,
known as DEXA, which measures bone density or bone
mass and takes less than 20 minutes to complete.
Risk factors for osteoporosis are as follows:
1. General risk factors include female, family history of
osteoporosis, Caucasian or Asian, older age, small/thin
frame, smoking, diet low in calcium, increased amounts
of alcohol, caffeine or soda, lack of weight-bearing exercise and diet that is high in salt.
2. Risk factors in survivors of childhood cancer include
anti-cancer treatment utilizing methotrexate or corticosteroids as well as radiation to weight-bearing bones.
Other medical treatments such as anticonvulsants
and medication used to treat early puberty and endometriosis (Lupron) can affect bones. Drugs including
aluminum-containing antacids (Maalox), cholesterol-
363
364
365
Appendix 30-1: Algorithm For Nutritional Intervention In The Pediatric Oncology Patient
Identify appropriate category: Age > 2 years choose either BMI (Body Mass Index) or IBW (Ideal Body Weight)
Age < 2 years choose WT/LT (Weight for Length) or IBW (Ideal Body Weight)
Normal
BMI
5-85th % ile
WT/LT
10-90th % ile
> 90-110 %
IBW
> 110-120%
> 120%
YES
NO
NO
NO
YES
Is patient a candidate
for tube feeding?
NO
YES
NO
NO
Monitor and intervene as needed
TPN
YES
YES
High risk of pulmonary aspiration or excessive emesis
YES
NO
NO
YES
NO
YES
Implement necessary changes
Post-pyloric feeds
Gastric feeds
NO
YES
Childrens Oncology Group, Cancer Control Nutrition Sub-Committee 10/04. Reprinted with permission.
366
Appendix 30-2: Algorithm for Nutritional Intervention and Categories of Nutritional Status in
the Pediatric Oncology Patient References and Resources
367
Childrens Oncology Group, Cancer Control Nutrition Sub-Committee 1/07. Reprinted with permission.
Appendix 30-3: Categories of Nutritional Status for the Pediatric Oncology Patient
Identify appropriate category: Age > 2 years choose either BMI1 (Body Mass Index) or IBW2 (Ideal Body Weight)
Age < 2 years choose WT/LT3 (Weight for Length) or IBW2 (Ideal Body Weight)
Weight loss/gain may or may not be present
Underweight
Normal
BMI
5-85th % ile
WT/LT
10-90th % ile
IBW
> 90-110 %
> 110-120%
> 120%
Childrens Oncology Group, Cancer Control Nutrition Sub-Committee 10/04. Reprinted with permission.
368
These agents have numerous uses, including to aid nasogastric intubations, and for gastroparesis, gastroesophageal
reflux, and intolerance to feedings.
Metoclopramide/Reglan
This drug blocks dopamine and at higher doses, 5 HT3
(serotonin) receptors. Its effects on motility may involve
increased release of acetylcholine in the gut. The effect on
the gut is increased forward peristalsis in the stomach and
duodenum. Metoclopramide has been used for the indications listed above as well as for chemotherapy-induced
nausea and vomiting. The latter use typically requires higher
doses. Data confirming the clinical utility vary with the indication. For chemotherapy-induced nausea and vomiting,
combinations of high-dose metoclopramide with dexamethasone and either diphenhydramine or lorazepam were
demonstrated to be effective in the 1980s, but serotonin
receptor antagonists such as ondansetron have been shown
to be superior. Metoclopramide is typically effective in
aiding intubations for patients with slow gastric motility and
for gastroparesis. Data proving utility in pediatric patients
with gastroesophageal reflux or feeding intolerance are
harder to find.120 Children are more sensitive to extrapyramidal side effects of dopamine blockers. Diphenhydramine
(Benadryl) is effective at preventing or treating dystonias,
while lorazepam is often more effective at reducing akathisias.121 While akathisias and dystonias are generally easily
reversed, there are a few reports of tardive dyskinesias
causing long-term movement disorders.120 In February 2009,
the Food and Drug Administration (FDA) announced it was
requiring a boxed warning in the labeling regarding the
risk of tardive dyskinesia with higher doses or longer-term
use of metoclopramide. Manufacturers must implement a
risk evaluation and mitigation strategy to ensure patients
receive a medication guide discussing the risk.122 The major
drug interactions involving metoclopramide relate to its
ability to speed transit through the gut. Drugs that are
designed for sustained release or that have slow dissolution
and absorption may have reduced absorption while a patient
2010 A.S.P.E.N. www.nutritioncare.org
Erythromycin
This drug is a macrolide antibiotic, but it also is a motilin
receptor agonist that increases proximal gut motility.120,124
Prokinetic effects are evident at doses as low as 4 to 12
mg/kg/d as compared to the 30 to 50 mg/kg/d used for
antimicrobial effects. Numerous studies suggest activity
of erythromycin for improving feeding tolerance in children.120,124 Typical problems with antimicrobial doses of
erythromycin include gastrointestinal upset, diarrhea, and
inhibition of P450 1A2 and 3A4 enzymes causing drug
interactions. These effects are less likely at the lower doses
used for motility. Although rare, there are some concerns
about adverse effects such as hepatotoxicity, ototoxicity,
cardiac arrhythmias, and pyloric stenosis.124 The latter has
been reported mostly in infants less than 2 weeks old. A
theoretical concern is the potential for low-dose antibiotics
to stimulate resistance among bacteria. This has not been
documented with the use of erythromycin for motility and
thus is an unknown. Caution should be used in patients with
hepatic impairment, largely due to the potential of erythromycin to cause hepatotoxicity.
Acid-Blocker Medications
Antiemetics
369
370
received therapy with anthracyclines (doxorubicin, daunorubicin, idarubicin, epirubicin, mitoxantrone, cisplatin, or
ifosfamide) are at the greatest risk for cardiovascular side
effects. The latter 2 can cause electrolyte abnormalities due
to renal tubular losses of calcium, magnesium, potassium,
phosphate, and bicarbonate. These losses may require supplementation, and oral supplementation generally tastes bad and
is poorly absorbed. Anthracyclines and mitoxantrone cause
damage to cardiac myofibrils that may result in heart failure
for some patients. This is most common when young children
receive the drug, and at lifetime doses above 300 mg/m2 for
younger children and 450 to 550 mg/m2 for older children
and adults. Ondansetron has a prolonged half-life in patients
with severe liver impairment and doses should be reduced.
Palonosetron, dolasetron, and granisetron do not need dose
adjustments in renal or hepatic dysfunction.134
Glucocorticoids (Dexamethasone/Decadron,
Methylprednisolone Sodium Succinate/Medrol)
These drugs have been used as antiemetics since the early
1980s, but it is still not clear how they work. They are used
mostly in combinations with a serotonin receptor antagonist
2010 A.S.P.E.N. www.nutritioncare.org
Phenothiazines (Prochlorperazine/Compazine,
Promethazine/ Phenergan, Chlorpromazine/
Thorazine) and Butyrophenones (Haloperidol/
Haldol, Droperidol/Inapsine)
These drugs block dopamine receptors in the vomiting
center and chemotherapy-receptor trigger-zone. These
agents are currently recommended mostly for acute nausea
and vomiting from mildly to moderately emetic chemotherapy regimens, or for breakthrough nausea and vomiting.
Prochlorperazine and promethazine have been used for
delayed nausea and vomiting with some success. Potential
side effects include sedation, anticholinergic effects such
as urinary retention or constipation, and alpha-adrenergic
blockade effects such as hypotension. However the major
adverse effects are extrapyramidal effects: akathisias
(restlessness) and dystonias (muscle contraction) such as
trismus and torticollis. These effects occur more frequently
in children and many pediatric practitioners limit the use
of dopamine blockers due to these side effects. Diphenhydramine (Benadryl) will generally reverse extrapyramidal
effects within 30 to 60 minutes, and has often been used
prophylactically to reduce the risk of extrapyramidal reactions. Also of note are specific issues with 2 of these drugs.
First, droperidol is either not used or dose-limited in many
institutions due to its ability to lengthen the QTc interval
and cause arrhythmias. Second, IV promethazine can be
very irritating on injection, and if injected into an artery
can cause significant tissue damage. Many institutions are
limiting its use to low doses or oral administration. Risk
may be minimal if central lines are available for administration, which is commonly the situation in children with
cancer. Additionally, promethazine has a boxed warning
in the package insert against using it in patients less than 2
years old, due to reports of fatal respiratory depression.
Appetite Stimulants
371
Cyproheptadine/Periactin
This drug is a serotonin antagonist and antihistamine that
has been studied as an appetite stimulant in cancer patients.
2010 A.S.P.E.N. www.nutritioncare.org
372
Magic Mouthwash
This usually contains viscous lidocaine, diphenhydramine,
and an antacid or nystatin. Lidocaine is a strong local anesthetic, diphenhydramine is a weak local anesthetic, and the
antacid may help neutralize acid in the stomach, although
the dose is probably too low to effectively raise gastric pH.
Nystatin is in combinations at some institutions because
mucositis may cause or be caused by Candida infections
(thrush). There are little if any published data on the efficacy of these combinations, but they relieve symptoms in
some patients. Combinations containing viscous lidocaine
have been reported to cause systemic lidocaine toxicity if
used in relatively high or frequent doses, especially in small
children or infants. Doses should be limited if the medication is to be swallowed. This is not a problem if it is swished
and spit out. Gelclair is a polymer-type combination of
polyvinylpyrrolidone, hyaluronic acid, and glycyrrhetinic
acid. It coats the mucosa of the mouth, protecting it from
air and irritants that elicit pain. It is suggested not to eat or
drink for an hour or more after using the Gelclair. Some
patients seem to benefit from this product. Opioids are used
when less potent treatments fail, and may allow the patient
2010 A.S.P.E.N. www.nutritioncare.org
Chlorhexidine/Peridex
Glutamine
Glutamine is an amino acid that is thought to be especially important to cells of the gut. Studies in the past have
suggested that it might help maintain the integrity of the gut
in patients receiving long-term total parenteral nutrition, or
may help prevent chemotherapy-induced mucositis. Studies
have suggested reductions in mucositis for autologous stem
cell transplant patients, but there also has been a suggestion
of increased relapses.141143 The most recent MASCC guidelines do not recommend the use of glutamine, but there is
enough interest that studies of specialized formulations of
glutamine are in clinical trials.
Palifermin/Kepivance
Palifermin/Kepivance is a recombinant human keratinocyte growth factor. It is approved and used for reducing
mucositis in patients with hematologic malignancies
receiving hematopoietic stem cell transplants. Use in solid
tumors is considered contraindicated by many practitioners due to the potential for stimulating epithelial cells (and
perhaps the cancers); however, studies in solid tumors in
patients receiving standard chemotherapy are in progress.
Palifermin is expensive and does cause some side effects
such as thick tongue, altered taste, and rashes. Palifermin
has been demonstrated to reduce the duration and severity
of oral mucositis as well as the use of opioids in patients
receiving autologous HSCT for hematologic malignancies.
There are less data for allogeneic transplants.
The MASCC guidelines mention numerous other
products (favorably or unfavorably), most of which are
373
References
374
375
376
99. Przepiorka D, Weisdorf D, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant.
1995;15(6):825828.
100. Cahn JY, Klein JP, et al. Prospective evaluation of 2 acute
graft-versus-host (GVHD) grading systems: a joint Societe
Francaise de Greffe de Moelle et Therapie Cellulaire (SFGMTC), Dana Farber Cancer Institute (DFCI), and International
Bone Marrow Transplant Registry (IBMTR) prospective
study. Blood. 2005;106(4):14951500.
101. Petersdorf EW, Longton GM, et al. The significance of
HLA-DRB1 matching on clinical outcome after HLA-A, B,
DR identical unrelated donor marrow transplantation. Blood.
1995;86(4):16061613.
102. Barker JN, Wagner JE. Umbilical-cord blood transplantation for the treatment of cancer. Nat Rev Cancer.
2003;3(7):526532.
103. Zecca M, Prete A, et al. Chronic graft-versus-host disease
in children: incidence, risk factors, and impact on outcome.
Blood. 2002;100(4):11921200.
104. Atkinson K. Chronic graft-versus-host disease. Bone Marrow
Transplant. 1990;5(2):6982.
105. Akpek G, Valladares JL, et al. Pancreatic insufficiency in
patients with chronic graft-versus-host disease. Bone Marrow
Transplant. 2001;27(2):163166.
106. Barker CC, Anderson RA, et al. GI complications in
pediatric patients post-BMT. Bone Marrow Transplant.
2005;36(1):5158.
107. Chakrabarti S, Collingham KE, et al. Isolation of viruses from
stools in stem cell transplant recipients: a prospective surveillance study. Bone Marrow Transplant. 2000;25(3):277282.
108. Wingard JR. Opportunistic infections after blood and marrow
transplantation. Transpl Infect Dis. 1999;1(1):320.
109. Burgunder MR, Dickson BJ. Hematopoietic stem cell transplantation. In: Kogut VJ, Luthringer SL. Nutritional Issues
in Cancer Care. Pittsburgh, PA: Oncology Nursing Society;
2005:253263.
110. Richardson PG, Elias AD, et al. Treatment of severe venoocclusive disease with defibrotide: compassionate use results
in response without significant toxicity in a high-risk population. Blood. 1998;92(3):737744.
111. Kumar S, DeLeve L, Kamath P, Tefferi A. Hepatic venoocclusive disease (sinusoidal obstruction syndrome) after
hematopoietic stem cell transplantation. Mayo Clin Proc.
2003;78:589598.
112. Cesaro S, Pillon M, et al. A prospective survey on incidence,
risk factors and therapy of hepatic veno-occlusive disease
in children after hematopoietic stem cell transplantation.
Haematologica. 2005;90(10):13961404.
113. Song JS, Seo JJ, et al. Prophylactic low-dose heparin or prostaglandin E1 may prevent severe veno-occlusive disease of the
liver after allogeneic hematopoietic stem cell transplantation
in Korean children. J Korean Med Sci. 2006;21(5):897903.
114. National Cancer Institute. Nutrition in Cancer Care (PDQ ).
http://www.cancer.gov/cancerinfo/pdq/supportivecare/
nutrition/healthprofessional. Accessed November 1, 2008.
115. Childrens Oncology Group. Long-term Follow-up Guidelines for Survivors of Childhood, Adolescent and Young
Adult Cancers. Version 3.0. Last updated March 2008. http://
www.survivorshipguidelines.org/pdf/LTFUGuidelines.pdf.
Accessed November 26, 2008.
116. Schwartz CL. Late effects of treatment in long-term survivors
of cancer. Cancer Treat Rev. 1995;21(4):355366.
117. Castellino S. GI Health - Gastrointestinal Health after Childhood Cancer. Health Link - Healthy living after treatment for
childhood cancer GI health, Version 3.0 - 10/08; 2008.
118. AICR. Nutrition and the Cancer Survivor, Special Population
Series. Washington, D.C.: American Institute for Cancer
Research; 2001.
119. Friedman D, Hudson MM, Landier W. Heart Health
Keeping Your Heart Healthy after Treatment for Childhood
Cancer. Health Link Healthy living after treatment for
childhood cancer Version 3.0 - 10/08; 2008.
120. Chicella MF, Batres LA, et al. Prokinetic drug therapy in
children: a review of current options. Ann Pharmacother.
2005;39(4):706711.
121. Kris MG, Hesketh PJ, et al. American Society of Clinical
Oncology guideline for antiemetics in oncology: update 2006.
J Clin Oncol. 2006;24(18):29322947.
122. Food and Drug Administration. U.S. Food and Drug Administration. 2009. http://www.fda.gov.
123. Food and Drug Administration. Metoclopramide boxed
warning announcement. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm.149533.htm.
Accessed September 1, 2009.
124. Curry JI, Lander TD, et al. Review article: erythromycin as
a prokinetic agent in infants and children. Aliment Pharmacol
Ther. 2001;15(5):595603.
125. Cuttica CE, Chicella MF, et al. Comparison of pantoprazole,
omeprazole and ranitidine in children requiring acid suppression: a prospective pilot study. J Pediatr Pharmacol Ther.
2004;9:198201.
126. Khan S, Shalaby TM, et al. The effects of increasing doses of
ranitidine on gastric pH in children. J Pediatr Pharmacol Ther.
2004;9(4):259264.
127. Wedlake LJ, Loader G. Cancer therapy-induced mucositis:
Where are we now? Clin Nutr Highlights. 2007;3:29.
128. Whitworth J, Christensen ML. Clinical management of
infants and children with gastroesophageal reflux disease. J
Pediatr Pharmacol Ther. 2004;9(4):243253.
129. Hatlebakk JG, Katz PO, et al. Proton pump inhibitors: better
acid suppression when taken before a meal than without a
meal. Aliment Pharmacol Ther. 2000;14(10):12671272.
130. Woods DJ, McClintock AD. Omeprazole administration. Ann
Pharmacother. 1993;27(5):651.
131. Olabisi A, Chen J, et al. Evaluation of different lansoprazole
formulations for nasogastric or orogastric administration.
Hosp Pharm. 2007;42(6):537543.
132. Schwartzberg LS. Chemotherapy-induced nausea and
vomiting: which antiemetic for which therapy? Oncology
(Williston Park). 2007;21(8):946953; discussion 954, 959,
962 passim.
377
31
CONTENTS
Nutrition Considerations in the
PediatricTrauma Patient . . . . . . . . . . . . . . . . . . . . . . . . . 378
General Background on Trauma in Childhood
Closed Head Injuries/Traumatic Brain Injury
Spinal Cord Trauma
Blunt Abdominal Trauma/Solid Visceral Injuries
Blunt Abdominal Trauma/Hollow Visceral Injuries
Thoracic Injuries
Summary on Pediatric Trauma
378
Learning Objectives
379
380
381
382
is a significant injury that demands prompt operative intervention to avoid morbidity and even mortality. Duodenal
hematomas become symptomatic when extraluminal blood
impacts on the luminal caliber, resulting in gastric outlet
obstruction. Passage of a transduodenal feeding tube is
almost always possible, either by radiographic guidance or
endoscopically. This allows the patient to receive enteral
nutrition despite the proximal obstruction, which may
require decompression with a concomitant nasogastric tube.
Operative intervention is rarely indicated in this injury.
Pancreatic injury may be concomitant so it is wise to check
for chemical pancreatitis before initiating feeds. Resolution
of the obstruction can occur within 10 to 14 days of injury
as evidenced by improved gastric emptying.
Duodenal perforation is a much more significant injury
which, depending on its extent, will require either primary
repair with drain placement or sometimes more complicated surgical solutions to protect the duodenal repair from
leakage (ie, pyloric exclusion with gastrojejunostomy).
Typically the pyloric channel will open 4 to 6 weeks later,
which prompts the gastrojejunostomy to close spontaneously. These patients will often require parenteral nutrition
support in the early postoperative phase until the gastrojejunal anastomosis becomes functional.
Small Intestine and Colon Injuries
Jejunoileal perforations or mesenteric injuries resulting in
devascularized segments of small intestine are frequently
associated with seatbelt injuries and other mechanisms
resulting in deceleration forces on the abdomen. These are
most commonly identified in the first 24 hours, however,
later strictures from ischemic injury without perforation
can cause late (> 4 weeks) symptoms of enteral intolerance.
Unless there is an extensive delay in diagnosis, most of these
injuries can be treated with resection and anastomosis.
When peritoneal contamination is extensive, the patients
hemodynamic status is not optimal, and the perfusion of
the involved segment is questionable, it is often prudent to
proceed with stomal diversion. These same principles apply
to colon perforation, which is rare and more commonly
encountered in penetrating mechanisms of injury. These
injuries require a brief period of non-enteral feeding, but
then permit rapid reinstitution of normal dietary habits.
Thoracic Injuries
Thoracic injuries in children may be remarkably unassociated with chest wall signs of trauma. The bony thoracic
case is sufficiently pliable in the young child to permit
complete transduction of the impact energy to the lung
2010 A.S.P.E.N. www.nutritioncare.org
Pediatric Burns
383
384
Clinical Example 1
Answer/Considerations
The energy assessment of the overweight and morbidly
obese teenager who requires critical care support is extraordinarily difficult. In this time of metabolic stress, the goal
should not be weight management, but preservation of
protein status. Estimates for caloric intake are best derived
from assessment of metabolically active tissues and these
are more closely related to ideal body weight than actual.
Not only will this patient need to heal her traumatic injuries
but she now has two additional wounds (thoracotomy and
laparotomy) to heal. Indirect calorimetry will likely provide
the best guidance in this scenario. Given the intra-abdominal injuries one would expect a period of ileus to follow
Clinical Example 2
Answer/Considerations
Pancreatic injuries, despite non-operative management, can
lead to significant metabolic stress as a consequence of the
inflammatory nature of the retroperitoneal injury (hematoma) which is further exacerbated by release of pancreatic
enzymes. While we can expect this patient to have been
active and healthy from a nutrition standpoint, we would
anticipate a return to GI function within perhaps 5 days.
The information related to the ERCP would suggest that
cautious introduction of enteral feeds would be acceptable
and that her response to enteral stimulation of pancreatic
secretions must be monitored. Should she develop recurrent pain and pancreatitis with oral intake, institution of
PN until such time as a jejunal feeding tube can be placed
would be appropriate.
Clinical Example 3
Answer/Considerations
This child has sustained a significant injury and is expected
to exhibit a stress response leading to catabolism. Nutrition
support will become essential to re-establishing a positive
nitrogen balance. Given the extent of injury this child will
likely have central venous access and provision of glucose
385
and protein can be started early via this route. At the time of
operation it would be ideal to have a nasoenteral tube placed
for subsequent enteral feeding access. Energy requirements
are notoriously difficult to predict and indirect calorimetry
remains the most accurate tool.
References
386
18. Schulman C, Ivascu F. Nutritional and metabolic consequences in the pediatric burn patient. J Craniofac Surg.
2008;19(4):891.
19. Palmieri T, Warner P, Mlcak R, et al. Inhalation injury in children: a 10 year experience at Shriners Hospitals for children. J
Burn Care Res. 2009;30(1):206.
20. Sheridan RL, Schnitzer JJ. Management of the high-risk pediatric burn patient. J Pediatr Surg. 2001 8;36(8):13081312.
21. Suman OE, Mlcak RP, Chinkes DL, Herndon DN. Resting
energy expenditure in severely burned children: analysis
of agreement between indirect calorimetry and prediction
equations using the Bland-Altman method. Burns. 2006
5;32(3):335342.
22. Chan MM, Chan GM. Nutritional therapy for burns in children and adults. Nutrition. 2009 3;25(3):261269.
23. Garrel DR, Razi M, Larivire F, et al. Improved clinical status
and length of care with low-fat nutrition support in burn
patients. J Parenter Enteral Nutr. 1995;19(6):482.
24. Saffle JR, Wiebke G, Jennings K, Morris SE, Barton RG.
Randomized trial of immune-enhancing enteral nutrition in
burn patients. J Trauma. 1997;42(5):793800.
25. Marin V, Rodriguez-Osiac L, Schlessinger L, Villegas J, Lopez
M, Castillo-Duran C. Controlled study of enteral arginine
supplementation in burned children: impact on immunologic
and metabolic status. Nutrition. 2006;22(7-8):705.
26. Cone L, Gilligan M, Kagan R, Mayes T, Gottschlich M.
Enhancing patient safety: the effect of process improvement
on bedside fluoroscopy time related to nasoduodenal feeding
tube placement in pediatric burn patients. J Burn Care Res.
2009;30(4):606.
27. Scaife CL, Saffle JR, Morris SE. Intestinal obstruction
secondary to enteral feedings in burn trauma patients. J
Trauma. 1999;47(5):859.
32
Surgery
Arlet G. Kurkchubasche, MD
CONTENTS
General Nutrition Considerations in
Pediatric Surgery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387
General Nutrition Considerations in Neonatal Surgery
Learning Objectives
388
SURGERY
389
390
SURGERY
391
Figure 32-1
392
The limits of tolerance are probably best defined by evaluating the volume and nature of the output. Because stool
output is not measured in terms of frequency of stools with
a stoma, the guideline to be used is that output should not
exceed one-third of total enteral intake. Other formulas
include the assessment of normal stoma output on the basis
of weight, with expected output estimated at 1 mL/kg/h
and tolerable amount to allow progression of enteral feeds
set at either 30 mL/kg/d or 2 mL/kg/h.15 Excess outputs
serve as indicators of inadequate absorptive capacity and
osmotic fluid losses and should be further evaluated with
the content of reducing substances and stool pH. Reducing
substances will shed light on carbohydrate malabsorption
with contents of 1% or greater in the presence of loose stools
constituting an indication for intervention, depending on
the type of enteral intake. Stool pH will reflect whether
there are organic acids generated by unabsorbed sugars, and
this can happen independent of the presence of reducing
substances.
Ideally a target caloric goal is established for each patient
and EN is pushed to the limit of tolerance with PN providing
the balance of calories necessary to achieve weight gain and
laboratory goals. While consistently pushing the limits
of the intestinal tract, it is important not to have frequent
setbacks which ultimately delay operation to restore continuity and thereby compromise the best case scenario for
complete adaptation. The use of distal refeeding provides
a valuable adjunct in these situations, at least contributing
to restoring a complete enterohepatic pathway for bile if the
distal ileum has been retained.16 The benefits are, however,
counterbalanced by the often arduous contraptions that
have to be created to collect and reinfuse the stoma output
and the risk of perforating the distal intestine with repeated
catheterization. The decisions involved in these complicated
cases require that all care providers reach consensus on the
short- and long-term plans for the patient. Only then can the
ideal nutrition support plan be instituted.
All surgical patients must be monitored carefully for
the onset of jaundice. Often the default reason for the onset
of direct hyperbilirubinemia is the use of PN; however,
surgical patients can develop intestinal obstruction whether
it be at the level of an anastomosis, at the level of a proximal
stoma, or at a random site due to peritoneal adhesions. These
conditions must remain in the foreground as correctable
causes of obstructive jaundice. Others conditions include
drug-related cholestasis (seen with cephalosporins), lowgrade sepsis, and even secondary disorders such as biliary
tract disease (gallstones and sludge) or pyloric stenosis.
SURGERY
393
394
SURGERY
395
396
Colon Atresia
Colon atresia is a rare condition, accounting for only 10%
of all intestinal atresia. Most common is a vascular defect
at the level of the middle colic artery resulting in an interruption of continuity at the mid-transverse colon. The right
colon becomes massively distended since a competent
ileocecal valve may not allow for distribution of the enteric
contents into the terminal ileum. Operatively there are
several solutions for this condition. An extended right hemicolectomy with anastomosis of the ileum to the transverse
colon results in permanent loss of a significant surface area
for nutrient and water absorption. Fortunately the left colon
can compensate for the water absorption with adaptation
and the patient should not have persistent watery stools.
More complicated solutions focus on preserving the right
colon with a proximal diverting stoma in the hopes that it
will decompress and regain its normal caliber and motility.
The nutrition considerations in these patients are generally
simple and are impacted by whether a second reconstructive operation is necessary.
Meconium Ileus and Variants
This is a condition usually associated with cystic fibrosis
(CF), resulting in distal ileal obstruction as a consequence
of the abnormal luminal contents which become inspissated.
Meconium ileus may be complicated further by either (1)
perforation of the obstructed bowel resulting in meconium
peritonitis or (2) volvulus of the distended bowel resulting
in ischemia and stricture formation which may appear identical to an atresia. While simple meconium ileus, in which
there is only luminal inspissation, can be decompressed with
sequential gastrograffin enemas, operation is sometimes
necessary. In the process of attempting to decompress the
obstructed segment with saline or N-acetylcysteine irrigation, it is possible to perforate or excessively traumatize the
intestine. Often the safest choice is placement of a T-tube
that provides access for continued postoperative irrigation
and gradual resolution of the obstruction. In the event of
complicated meconium ileus due to luminal discontinuity,
a resection and primary anastomosis may be possible, but
because of the risk of recurrent distal obstruction, creating
a vented anastomosis (Bishop Koop or Santulli stoma)
provides the greatest assurance of success. This allows
the enteric contents to evacuate per stoma should distal
obstruction re-occur and provides direct access to the
distal bowel to treat with 3% N-acetylcysteine solution. The
most difficult variant to deal with operatively is meconium
peritonitis, in which the site of perforation may be unable
to be identified or mobilized to create a stoma. A proximal
2010 A.S.P.E.N. www.nutritioncare.org
diverting stoma becomes necessary with staged exploration and restoration of continuity at a later date. If the distal
end of the bowel can be identified this is helpful in trying
to prepare it for the subsequent operation and for consideration of distal refeeding of proximal stoma output (see later
section in this chapter).
Nutrition management of these infants requires consideration of the likely underlying diagnosis of CF, which must
be verified by testing. While in the past it was taught that EN
with a protein hydrolysate formula or breast milk did not
require provision of pancreatic enzymes, common practice
now is to provide these enzymes nevertheless, to optimize
whatever enteral intake is available for absorption. This has
to be balanced with the past concerns for mucosal damage
from specific formulations of enzyme dosing. This infant
with a diverting stoma is a classic example of the nutrition
decision-making involved with a child who will require a
second, complicated operation, 6 weeks or more in the future
(see section on management of stoma output). Cholestatic
jaundice is based not only on total PN exposure, but also
on potentially underlying liver disease associated with CF.
Once intestinal continuity to a stoma or the rectum is established postoperatively the goal is to wean total PN support
expeditiously while recognizing that these infants tend to
have a higher than usual caloric requirement.
Hirschsprungs Disease
Hirschsprungs disease is a consequence of the absence of
the ganglion cells from the rectum and distal-most colon,
typically involving the rectosigmoid. The enteric ganglion
cells are essential to receptive relaxation of the intestine and
their absence results in a functional obstruction of the colon
with remarkable dilation of the normal colon proximal to
the transition zone beyond which these neural crest cells
failed to migrate during the embryonic period. The consequence is a functional obstruction at the level of the absence
of ganglion cells (aganglionosis). Hirschsprungs disease
most often affects term or near-term infants and is unusual
in preterm infants. The newborn who is diagnosed and
promptly undergoes reconstructive operationwhether it
be an open operation, laparoscopic assisted, or transanal
is likely to be able to start normal EN within 5 days of
operation. Therefore the provision of even peripheral total
PN, during the first few days of life, once the child has been
diagnosed, is probably sufficient. This scenario, however,
changes with a late diagnosis of Hirschsprungs disease,
which is often associated with significant failure to thrive,
enterocolitis, and long-segment disease. These situations are
independent predictors of a difficult postoperative course
SURGERY
397
Beckwith-Wiedemann syndrome is diagnosed when omphalocele is associated with macroglossia, hypoglycemia, and
hemihypertrophy. This is a transient metabolic condition
with no long-term implications for glycemic control, but
places the patient at risk for solid organ tumors. Associated
congenital heart disease may involve various intracardiac
defects or a constellation of defects involving the heart,
pericardium diaphragm, and sternum which are referred to
as the Pentalogy of Cantrell. Depending on the size of the
abdominal wall defect, the liver and intestine are typically
extra-abdominal, and reconstruction can be accomplished
in one vs. several stages.
A small omphalocele can be closed within days after
birth once associated anomalies have been excluded. The
child is typically kept NPO preoperatively, although this
may not strictly be necessary, but convenient for the preoperative evaluations. Once a small omphalocele is closed,
enteral alimentation can be commenced as soon as there
is evidence of GI motility/function. While there are no
predictable GI problems, extrinsic gastric compression from
the oversized liver and GER are not uncommon and may
require transient postpyloric feeding. In the more complicated giant omphaloceles, PN is commonly started to bridge
the childs nutrition needs while multiple operative procedures interfere with consistent EN. Ethical considerations
arise when an infant with omphalocele is diagnosed with
a lethal chromosomal disorder such as trisomy 18. Often
the surgical condition can be managed in a non-operative
manner and EN can be delivered in a relatively non-invasive
manner via a nasogastric feeding tube if the child will not
feed by mouth.
The outcomes of omphalocele should be excellent but
for the most complex disorders.
Gastroschisis
Gastroschisis is frequently the more complex defect from
the standpoint of operative interventions and providing
optimal nutrition support. This abdominal wall defect is
typically a relatively small full-thickness aperture to the
right of the umbilical cord insertion. Much of the GI tract
is extruded through this defect and is bathed in amniotic
fluid through pregnancy. The exposed intestine is at risk
for torsion, vascular impairment from a tight fascial defect
which remains fixed as the intestine and its mesentery
enlarge, and is subject to serosal irritation and trauma by
virtue of its extraperitoneal location. Immediate postnatal
coverage of the intestine is accomplished by either primary
abdominal closure or more commonly by placement of a
temporary silo. This sterile silastic tube accommodates
2010 A.S.P.E.N. www.nutritioncare.org
398
SURGERY
postnatal period there appears to be an intense inflammatory state as evidenced by elevated CRP which resolves as
the bowel recovers. The incidence of cholestatic jaundice is
likely multifactorial in this population.26
Congenital Diaphragmatic Hernia
Congenital diaphragmatic hernia (CDH) involves a heterotopic location for the viscera within either chest cavity
as well as associated developmental defects in lung and
pulmonary vascular development and maturation. While
the former is a surgically correctable defect, the second
component determines the childs clinical course and ultimate prognosis. Infants with CDH therefore fall into several
categories. There are those with a diaphragmatic defect,
usually first detected postnatally, in whom there is little
hemodynamic and pulmonary compromise; they tolerate
the operation well and can relatively quickly be transitioned to enteral feedings. Infants in whom the defect was
identified early in gestation tend to have a greater degree
of pulmonary hypoplasia and more difficulty with pulmonary hypertension. These infants may require cardiac and
pulmonary support via ECMO. Nutrition support in this
disorder therefore spans the whole spectrum and must be
individualized. The GI tract, although displaced initially, is
fundamentally normal in terms of its motility and absorption capacity. As such there are no specific recommendations
for the type of enteral alimentation. Consideration will
need to be given to the fact that total fluid provision will
likely be restricted in the postoperative patient as he or
she is weaned off ventilatory support. While it is enticing
to provide highly concentrated formulations, this has to be
balanced with the potential for mucosal damage leading to
enteric-derived sepsis in vulnerable infants who likely have
multiple vascular access points. Provision of adequate calories will likely be best accomplished by this means once the
infant has tolerated enteral feedings and becomes limited
in the amount of volume that can be delivered for reasons
of gastric capacity and emptying. All children with CDH
have some degree of foregut dysmotility, which renders
them at risk for GER.27,28 The risk of reflux with aspiration
is particularly pronounced in these patients who already
have a compromised pulmonary system, given the nature
of the congenital defect. This forms the basis for providing
continuous feeds initially to full enteral needs and then
transitioning them to bolus feeds. Again, the provision of
continuous enteral feedings should not be a disincentive to
allow for oral feeds. This can be structured to be the volume
of feeds targeted to be delivered over 1 hour or even more as
the child tolerates.
399
Hepatobiliary Disorders
Biliary Atresia
Infants presenting with direct hyperbilirubinemia are
promptly evaluated for surgically correctable biliary tract
disorders such as biliary atresia and choledochal cyst.
While other conditions such as neonatal hepatitis also lead
to similar laboratory presentations, they, as well as a host of
infectious disorders and enzymatic defects, require medical
support only. Biliary atresia most often becomes evident at 1
month of life and the infant may have experienced failure to
thrive of unknown etiology. Hallmarks of clinical diagnosis
include jaundice, acholic stools, and dark urine. Ultrasound,
HIDA scan, and percutaneous liver biopsy should confirm
the diagnosis. Once established, preoperative administration of vitamin K is one of the standard recommendations to
optimize coagulation parameters. The diagnosis is further
confirmed intraoperatively by demonstrating lack of the
2010 A.S.P.E.N. www.nutritioncare.org
400
SURGERY
401
402
Pyloric Stenosis
Infants with pyloric stenosis primarily require rehydration
and correction of electrolytes preoperatively but then can
be relied upon to resume a normal infant diet. How this is
initiated varies by institution. In general, no postoperative
feeding tubes are placed as these put the exposed pyloric
channel mucosa to the risk of perforation. The infants are
typically hungry and have good feeding skills. In this authors
institution, our feeding protocol was developed to provide a
consistent algorithm that allows the vast majority of infants
to be discharged to home within 24 hours of operation. We
hold oral feeds for 6 hours, and then test the stomach with 2
small-volume (15 mL) Pedialyte feeds 2 hours apart. If the
SURGERY
403
404
Insertion of Gastrostomy
When a gastrostomy tube is inserted for the sole purpose of
providing enteral access, enteral feedings can be commenced
2010 A.S.P.E.N. www.nutritioncare.org
SURGERY
405
4. Which clinical diagnosis is expected to have impairment of intestinal motility and may require prolonged
total PN support?
A. Gastroschisis
B. Omphalocele
C. Hirschsprungs disease
D. Malrotation without midgut volvulus
See p. 487 for answers.
References
406
PART IV
NUTRITION CARE OF
THE PEDIATRIC PATIENT
33
Learning Objectives
Contents
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
Nutrition Assessment of Premature Infants. . . . . . . . . . 409
Classification Parameters
Special Considerations
Energy Needs
Growth
Background
Classification Parameters
Premature infants are classified by the infants gestational
age, growth curve parameters, and maturational examination. The maturational examination, known as the
Ballard score, is a postnatal, indirect method of assessing a
neonates gestational age. It is based upon indicators of fetal
409
410
< 2500 g
< 1500 g
< 1000 g
< 750 g
Special Considerations
Specific medical conditions will alter the neonates nutrition needs and ability to feed enterally and by mouth. For
instance, gastrointestinal anomalies such as gastroschisis,
omphalocele, and necrotizing enterocolitis (NEC) will
mandate feeding the infant with parenteral nutrition (PN)
support for longer time periods until after surgical intervention. Furthermore, infants with NEC may suffer from
short bowel syndrome (SBS) postsurgery and may require
long-term PN support. The transition to enteral feeds is
often tenuous and requires slower advancement to full
2010 A.S.P.E.N. www.nutritioncare.org
Energy Needs
The premature infants weight may decrease 10% to 15%
in the first week of life due to decreased water content of
the extracellular volume. Until the weight is regained, the
infants birth weight should be used to estimate energy and
nutrient needs.
Growth
The premature infant should be weighed nude, at the same
time of day, on the same scale, on a daily basis to evaluate
nutrition status. The infants average daily weight gain is
compared (in grams per kilogram daily or grams per day)
to expected intrauterine weight velocity charts. Please refer
to Table 33-2.6 Extreme weight changes may be due to fluid
shifts or medical conditions, and these factors should be
considered before modifying an infants nutrition support
regimen.
Average Daily
Weight Gain
(g/d)
Mean Weight
(g)
Average Daily
Weight Gain
(g/kg/d)
2425
2526
2627
2728
2829
2930
3031
3132
3233
3334
3435
3536
3637
3738
Mean
11.4
15.7
18.6
21.4
22.6
23.1
24.3
25.7
27.1
30
31.4
34.3
35.7
31.4
25.2
904961
9611001
10011065
10651236
12361300
13001484
14841590
15901732
17321957
19572278
22782483
24832753
27532866
28663025
12.2
16
18
18.6
17.8
16.6
15.8
15.5
14.7
14.2
13.3
13.1
12.7
10.7
14.9
The purpose of the nutrition assessment of infants and children is to (1) evaluate growth, body size measurements, and
composition as compared to national standards; (2) determine an estimation of nutrient needs; and (3) evaluate the
adequacy of the nutrition regimen. Infants experience a more
rapid growth velocity as compared to children. The need
for medical care may impact the childs nutrition status by
altering metabolic requirements and nutrient intake, and can
result in weight loss and decreased height or length velocity.8
Refer to Table 33-3 for reference standards of growth velocity,
both weight and length, in healthy children.9
411
Weight (g/d)
Length (cm/mo)
< 3 mo
36 mo
612 mo
13 y
46 y
710 y
2535
1521
1013
410
58
512
2.63.5
1.62.5
1.21.7
0.71.1
0.50.8
0.40.6
412
kcal/kg
1989 RDAs
Protein g/kg
2002 DRIs
06 mo
712 mo
13 y
46 y
710 y
Males
1114 y
1518 y
108
98
102
90
70
1.52 (AI)
1.2
1.05
0.95 (48 y)
0.95 (913 y)
55
45
0.85 (1418 y)
47
40
0.85 (1418 y)
Females
1114 y
1518 y
Males
03
310
1018
1830
Females
03
310
1018
1830
kcal/d
60.9W 54
22.7W + 495
17.5W + 651
15.3W + 679
61W 51
22.5W + 499
12.2W + 746
14.7W + 496
100 mL/kg
1000 mL + 50 mL each kg
over 10 kg
1500 mL + 20 mL for each kg
over 20 kg
413
414
< 0.3
< 0.30.6
> 0.6
None
0.25 mg/d
0.5 mg/d
1 mg/d
None
None
0.25 mg/d
0.5 mg/d
None
None
None
None
Age
06 mo
6 mo3 y
36 y
616 y
*1 ppm = 1 mg/L31
Diet History
Anthropometric Measurements
Weight
Weights of children under the age of 2 should be measured
on a leveled scale that is frequently calibrated. The scale
should be calibrated regularly with appropriate standards.
The nude infant is placed on the scale, making sure the
weight is evenly distributed on either side of the center of
the scale. Weight is recorded to the nearest 10 g.12,32
For children older than 2 years of age, the weight should
be measured using a standing scale. The calibration of the
scale should also be done regularly with appropriate standards. Subjects are to stand still in the middle of the scale,
with their body weight evenly distributed between both
feet. Light indoor clothing should be worn. Weights should
be recorded to the nearest 100 g.12,32
Length/Height
With children under 3 years of age, length should be
obtained in the recumbent position using a length board.
This is a device consisting of a flat board and a moveable
footboard, which are perpendicular to the table surface. A
fixed measuring tape is present with the 0 end at the edge
of the headboard. The infants length should be recorded
as the distance between the headboard and the footboard.
An assistant should hold the infant/toddlers head with the
child looking upward and the crown of the childs head
against the headboard. The examiner holds the infants
legs straight with the feet against the board and the toes
facing up. The footboard is placed flat against the infant/
toddlers feet. The measurement is recorded to the nearest
0.1 cm.12,32
Children older than 3 years of age are to be measured,
without shoes, using a standiometer. A standiometer is a
metric tape affixed to a vertical surface and a moveable
415
Head Circumference
Occipital-frontal circumference (OFC) should be obtained
in infants and children until 3 years of age. Head circumference is measured with a narrow, non-stretchable measuring
tape. The tape should cross the forehead, just above the
supraorbital ridges, passing around the head at the same
level on both sides to the occiput. The tape should be moved
up and down until maximum circumference is obtained.
Sufficient tension should be placed on the tape to press the
hair against the skull. 37
Interpretation
Underweight
At risk for underweight
Adequate
Overweight
Obese
416
Growth Charts
Fenton for Premature Infants
The Babson and Benda 1976 fetal-infant growth graph
for premature infants was commonly used to assess growth
of premature infants in the neonatal intensive care unit
2010 A.S.P.E.N. www.nutritioncare.org
Biochemical Indices
Biochemical parameters are also an essential component of
a comprehensive nutrition assessment. Routine laboratory
monitoring with the pediatric population includes complete
blood counts and blood lead levels to rule out lead exposure. Abnormal hemoglobin (Hgb) and hematocrit (Hct)
levels can aid in the detection of iron, vitamin B12, and folate
deficiencies.
A complete blood count can identify the degree and
features of anemia. Red cells are microcytic and hypochromic
417
Serum albumin, transferrin, and prealbumin are indicators of visceral protein status. Depleted levels can be an
indication of visceral protein depletion and malnutrition.
However, other clinical conditions (eg, altered fluid status,
inflammation, acute infection) may falsely decrease levels.
Physical Exam
Physical examination of the pediatric patient is also a vital
component of the nutrition assessment. Physical examination can provide insight into conditions such as malnutrition,
obesity, edema, dehydration, and vitamin deficiencies and
excesses. Table 33-13 outlines some nutrition concerns
based on physical examination.2,33,39
Physical Examination
Skin Integrity
Pallor
Dermatitis
Spoon shape
Iron deficiency
Lackluster, dull
Protein deficiency
Vitamin A or C deficiency
Moon face
Protein-calorie malnutrition
Protein-calorie malnutrition
Neck
Enlarged thyroid
Iodine deficiency
Mouth
Bleeding gums
Vitamin C deficiency
Inflammed mucosa
Magenta color
Riboflavin deficiency
Vitamin A deficiency
Easily pluckable
Protein deficiency
Dentition
Abdomen
Rounded, distended
Temperature
Increased temperature
Respiration
Nails
Face
Tongue
Eyes
Hair
418
Description
Infants:
00.5 y:
108 x Wt (kg)
0.51 y: 98 x Wt
Children:
13 y: 102 x Wt
46 y: 90 x Wt
710 y: 70 x Wt
Males:
11-14 y: 55 x Wt
15-18 y:
45 x Wt
Females:
11-14 y: 47 x Wt
15-18 y: 40 x Wt
Estimated Energy
Replaces the 1989 Recommended
EER = TEE + energy deposition
Requirements
Dietary Allowances (RDA).
Ages 0-36 mo:
(EER) (new DRI/
0-3 mo: (89 x wt [kg] - 100) + 175
Energy needs were determined from
IOM equation) &
4-6 mo: (89 x wt [kg] - 100) + 56
children with normal growth, body
Physical Activity (PA) composition, and activity, and who are 7-12 mo: (89 x wt [kg] - 100) + 22
Co-Efficients
13-36 mo: (89 x wt [kg] -100) + 20
also metabolically normal.
Ages 3-8 yBoys:
EER = 88.5 - (61.9 x age [y]) + PA x (26.7 x wt [kg] + 903 x ht[m]) + 20 kcal
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.13 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.26 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.42 if PAL is estimated to be > 1.9 < 2.5 (very active)
Ages 3-8 yGirls:
EER = 135.3 - (30.8 x age [y]) + PA x (10 x wt [kg] + 934 x ht [m]) + 20 kcal
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.16 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.31 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.56 if PAL is estimated to be > 1.9 < 2.5 (very active)
Ages 9-18 yBoys:
EER = 88.5 - (61.9 x age [y]) + PA x ( 26.7 x wt [kg] + 903 x ht [m]) + 25 kcal
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.13 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.26 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.42 if PAL is estimated to be > 1.9 < 2.5 (very active)
Ages 9-18 yGirls:
EER = 135.3 - (30.8 x age [y]) + PA x (10 x wt [kg] + 934 x ht [m]) + 25 kcal
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.16 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.31 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.56 if PAL is estimated to be > 1.9 < 2.5 (very active)
EER (new DRI/IOM
Weight Maintenance TEE in Overweight Boys Ages 3-18 y:
equation) & obesity
TEE = 114 - (50.9 x age [y]) + PA x (19.5 x weight [kg] + 1161.4 x height [m])
co-efficients/factors
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.12 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.24 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.45 if PAL is estimated to be > 1.9 < 2.5 (very active)
Weight Maintenance TEE in Overweight Girls Ages 3-18 y:
TEE = 389 - (41.2 x age [y]) + PA x (15 x weight [kg] + 701.6 x height [m])
PA = 1 if PAL is estimated to be > 1 < 1.4 (sedentary)
PA = 1.18 if PAL is estimated to be > 1.4 < 1.6 (low active)
PA = 1.35 if PAL is estimated to be > 1.6 < 1.9 (active)
PA = 1.6 if PAL is estimated to be > 1.9 < 2.5 (very active)
Schofield
A predictive equation for calculating
Males:
basal metabolic rate (BMR) in healthy 0-3 y: (0.167 x wt [kg]) + (15.174 x ht [cm]) - 617.6
children that was developed by analysis 3-10 y: (19.59 x wt [kg]) + (1.303 x ht[cm]) + 414.9
of Fritz Talbot tables.
1018 y: (16.25 x wt [kg]) + (1.372 x ht[cm]) + 515.5
> 18 y: (15.057 x wt [kg]) + (1.0004 x ht[cm]) + 705.8
Females:
0-3 y: (16.252 x wt[kg]) + (10.232 x ht [cm]) - 413.5
3-10 y: (16.969 x wt [kg]) + (1.618 x ht [cm]) + 371.2
1018 y: (8.365 x wt [kg]) + (4.65 x ht [cm]) + 200
>18 y: (13.623 x wt [kg]) + (23.8 x ht [cm]) + 98.2)
FAO/WHO
The WHO equation was developed
Males:
for use in healthy children; however, it
03 y: (60.9 x wt [kg]) - 54
is commonly used to predict resting
310 y: (22.7 x wt [kg]) + 495
energy expenditure (REE) of acutely ill 1018 y: (17.5 xwt [kg]) + 651
patients in the hospital setting.
Females:
03 y: (61 x wt [kg]) - 51
310 y: (22.5 x wt [kg]) + 499
1018 y: (12.2 x wt [kg]) + 746
Based on the median energy intakes of
children followed in longitudinal growth
studies
It can overestimate needs in non-active
populations (eg, bedridden) and does
not provide a range of energy needs.
Though an outdated reference, still
widely used.
Applicable to Which
Patient Populations?
Source
Children with
normal growth, body
composition, and
activity, and who are also
metabolically normal.
Source: Ludlow V, Randall R, Burritt E, Rago D. Estimating Nutrient Needs, Pediatric Module. A.S.P.E.N. Enteral Nutrition Practitioner Tutorial Project, pending publication.
419
Description
White equation
Applicable to Which
Patient Populations?
Children with
developmental
disabilities
*This reference applies
to the specific ages
listed. Please refer to
another equation for
ages outside of the
referenced ages, and
apply an appropriate
activity/stress factor.
EE (kJ/day) = (17 age [mo]) + (48 weight [kg]) + (292 body temperature
[C]) - 9677
Starvation 0.700.85
Surgery 1.051.5
Sepsis 1.21.6
Closed head injury 1.3
Trauma 1.11.8
Growth failure 1.52
Burn 1.52.5
Pediatric hospitalized
population
02 y: 23 g/kg/d
Source
Children with
normal growth, body
composition, and
activity, and who are also
metabolically normal.
Children with
normal growth, body
composition, and
activity, and who are also
metabolically normal.
420
Reprinted with permission from Frisancho AR. New norms of upper limb fat and muscle areas for assessment of nutritional status.
Am J Clin Nutr. 1981;34:25402545.
421
Reprinted with permission from Frisancho AR. New norms of upper limb fat and muscle areas for assessment of nutritional status.
Am J Clin Nutr. 1981;34:25402545.
422
Fenton TR. Fetal-infant growth chart for preterm infants. BMC Pediatrics. 2003 Dec 16;3(1):13. Reproduced with permission from BioMed Central.
423
Appendix 33-5
424
425
426
427
428
429
430
References
431
9. Fomon SJ, Haschke F, Ziegler EE, Nelson SE. Body composition of reference children from birth to age 10 years. Am J Clin
Nutr. 1982;35:1169.
10. National Health and Nutrition Examination Survey. CDC
Growth Charts: United States. http://www.cdc.gov/nchs/
about/major/nhanes/growthcharts/background.htm.
Accessed October 18, 2008.
11. Waterlow JC, Buzina R, Keller W, Lane JM, Nichamon MZ. The
presentation and use of height and weight data for comparing
the nutritional status of children under the age of 10 years. Bull
World Health Organization. 1977;55(4):489498.
12. Frisancho AR. Anthropometric Standards for the Assessment of
Growth and Nutritional Status. Ann Arbor, MI: The University
of Michigan Press; 2004.
13. Food and Nutrition Board, Committee on Dietary Allowances. Recommended Dietary Allowances. 10th ed. Washington
DC: The National Academies Press; 1989.
14. World Health Organization. Energy and Protein Requirements. Report of a Joint FAO/WHO/UNU Expert
Consultation. Technical Report Series 724. World Health
Organization, Geneva; 1985.
15. Otten JJ, Pitzi Hellwig J, Meyers LD, eds. Dietary Reference
Intakes: The Essential Guide to Nutrient Requirements.
16. Holliday MA, Segar WE. The maintenance for water in parenteral fluid therapy. Pediatrics. 1957;19:823832.
17. Oh TH. Formulas for calculating fluid maintenance requirements. Anesthesiology. 1980; 53:351353.
18. American Academy of Pediatrics, Committee on Nutrition.
Adolescent nutrition. In: Kleinman RA, ed. Pediatric Nutrition Handbook. 5th ed. Elk Grove, IL: American Academy of
Pediatrics; 2004:149154.
19. Faulhaber D. Nutrition assessment of adolescents. In:
Nevin-Folino N, ed. Pediatric Manual of Clinical Dietetics.
2nd ed. Chicago, IL: Pediatric Nutrition Practice Group;
2003:163172.
20. American Academy of Pediatrics, Committee on Nutrition.
Calcium requirements of infants, children, and adolescents.
Pediatrics. 1999;104:11521157. http://aappublications.org/
cgi/content/full/pediatrics;104/5/1152. Accessed October
11, 2008.
21. Wyshak G, Frisch RE. Carbonated beverages, dietary calcium,
the dietary calcium/phosphorus ratio, and bone fractures in
girls and boys. J Adolesc Health. 1994;15:210215.
22. Borgna-Pignatti C, Marsella M. Iron deficiency in infancy and
childhood. Pediatric Annals. 2008;37(5):329337.
23. American Academy of Pediatrics, Committee on Nutrition. Iron fortification of infant formulas. Pediatrics.
1999;104:119123. http://aappublications.org/cgi/content/
full/pediatrics;104/1/119. Accessed October 12, 2008.
24. American Academy of Pediatrics, Committee on Nutrition. Iron supplementation for infants. Pediatrics.
1976;58:765768.
25. Siimes MA, Jarvenpa AL. Prevention of anemia and iron
deficiency in very-low-birth-weight infants. J Pediatr.
1982;101:277280.
26. Pupillo J. Bone up on new vitamin D recommendations: all
infants, children, adolescents should get at least 400 IU a day.
AAP News. 2008;29:1.
432
27. Wagner CL, Greer FR, and the Section on Breastfeeding and
Committee on Nutrition. Prevention of rickets and vitamin
D deficiency in infants, children, and adolescents. Pediatrics. 2008;122:11421152. http://aappublications.org/cgi/
content/full/pediatric;122/5/1142. Accessed November 13,
2008.
28. American Academy of Pediatrics, Section on Pediatric
Dentistry. Policy statement: Oral health risk assessment
timing and establishment of the dental home. Pediatrics.
2003;111:11131116.
29. US Department of Health and Human Services; National
Institute of Dental and Craniofacial Research. Oral health
in America: A report of the surgeon general. Rockville, MD:
National Institutes of Health; 2000.
30. American Academy of Pediatric Dentistry, Council on Clinical Affairs. Policy statement on the use of fluoride. Adopted
5/2000; revised: 5/2001. http://www.aapd.org/members/
referencemanual/pdfs/Fluoride.pdf. Accessed October 11,
2008.
31. US Department of Health and Human Services. Recommendations for using fluoride to prevent and control dental caries
in the United States. MMWR Recomm Rep. 2001;50(-14).
http://www.cdc.gov/mmwr/preview/mmwrhtml/rr5014a1.
htm. Accessed November 13, 2008.
34
CONTENTS
When to Use Enteral versus
ParenteralNutrition Support. . . . . . . . . . . . . . . . . . . . . . . 431
Determining the Best Enteral Approach. . . . . . . . . . . . . . 432
Functional Barriers to Oral Enteral Nutrition
Structural Barriers to Enteral Nutrition
Candidates for Enteral Nutrition
Inadequate Oral Intake and Prematurity
Inadequate Oral Intake and BronchopulmonaryDysplasia
Inadequate Oral Intake and Cystic Fibrosis
Inadequate Oral Intake and Congenital Heart Disease
Inadequate Oral Intake and Renal Disease
Inadequate Oral Intake in Burns, Sepsis/Critical Illness, and HIV
Enteral Nutrition: Routes of Administration
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 443
Learning Objectives
434
Patients with congenital anomalies such as tracheoesophageal fistula (TEF), esophageal atresia, cleft palate, and
Pierre Robin syndrome require a source of nutrition that
bypasses the oral route, at least until the condition is surgically repaired.4 EN support would also be indicated in cases
of obstruction of the oral cavity and surrounding areas such
as cancer of the head/neck, or mechanical ventilation. Injuries such as caustic ingestion, trauma, or burns to the head/
neck area may impede ability to consume oral intake as
well.4 Other types of severe oral injury that may warrant EN
support include mucositis and Stevens-Johnson syndrome
cases that involve oral lesions.
Functional barriers to safe oral intake may include neurological or neuromuscular diseases, genetic/metabolic
syndromes, and prematurity.4 For example, bulbar dysfunction is common in patients with severe spinal muscular
atrophy, which can lead to problems with eating and swallowing, as well as aspiration pneumonia. Reflux related to
delayed gastric emptying can also occur. 5 Chewing and
swallowing difficulties are common in children affected
by neuromuscular disorders, and can lead to aspiration.6
In fact, most children with cerebral palsy benefit from tube
feedings.6 Children with neurological impairments, such
as severe seizure disorders or anoxic brain injury, usually
require enteral feeding long-term. 3 Infants or children with
neurological impairments may also develop oral aversions
due to sensory integration disorders.
Structural barriers to oral intake include congenital anomalies, obstructions, injuries, and some types of surgery.
In premature infants, the inability to coordinate the suckswallow-breathe pattern may prevent safe oral feedings. 8
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
435
436
Increased
Nutrient Needs
Increased
GI Losses
Primary
Therapy
Oral Motor
Dysfunction
Structural or
Functional Abnormality
of GI Tract
Injury/Critical
Illness
Anorexia nervosa
Cystic fibrosis
Pancreatic
insufficiency
Metabolic
diseases
Prematurity
Congenital malformation
Burn
Anorexia related
to medical
condition
BPD
FTT (requires
catch-up growth)
SBS
Intolerance to
fasting
Neuromuscular
disorders
Intestinal pseudoobstruction
Trauma
Anorexia due
to treatment/
medications (eg,
chemotherapy)
Congenital heart
disease
Renal disease
Cholestatic liver
disease
Biliary atresia
Unpalatable diet
(eg, elemental
or metabolic
formula)
Neurological
impairment (eg,
cerebral palsy)
TEF
Proximal fistula with
highoutput
Surgery
Food aversion
Mucositis
Infection
Malabsorption
IBD
Proximal intestinal
obstruction
Oral intubation
Parenteral Nutrition
Gastrointestinal Conditions
Infants born with congenital anomalies of the GI tract
generally require PN. Children who develop GI disorders
that prohibit the use of enteral feedings rely on PN for their
nutrition, either temporarily or permanently. For instance,
SBS may result from major surgical resection of the GI
tract, or may develop in infants born with gastric anomalies.19 Intestinal atresia that requires extensive resection or
gastroschisis that causes necrotic bowel can lead to SBS.
Necrotizing enterocolitis (NEC), trauma, and volvulus are
other examples of conditions in which SBS can occur. PN
is always indicated for SBS in the immediate postoperative
period.19,20 Eventually, many individuals with SBS can adapt
to enteral feeds with the successful weaning of PN.19 NEC
in infants requires PN, as enteral feedings are withheld for a
period of time to support bowel healing and recovery. 3,21,22
Those with intestinal failure, as defined by inability of
the gut to digest or absorb an adequate amount, or extensive lesions and/or motor dysfunction, usually require
2010 A.S.P.E.N. www.nutritioncare.org
Prematurity
Premature and low birth weight infants need a source
of nutrition immediately after birth due to low nutrient
reserves, increased energy expenditure, immaturity of the
GI tract, as well as their increased propensity for acute and
chronic illness.8,21 Infants should receive PN if it is anticipated that they will be unable to receive enteral feedings
for more than 2 to 3 days. Infants have very limited fat and
glycogen stores, and when these have been depleted infants
begin to catabolize protein stores for energy. However,
EN is commonly withheld if the following conditions are
present: severe respiratory failure associated with hypoxia
and acidosis; hypotension treated with vasopressors; peri
natal asphyxia with signs of organ involvement; clinical
signs of NEC or intestinal obstruction; congenital heart
disease with decreased left-sided outflow; patent ductus
arteriosus with left to right shunt or requiring Indomethacin
or surgical ligation; or sepsis-associated paralytic ileus.
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
Anorexia Nervosa
Severely malnourished patients with anorexia nervosa who
require urgent nutrition rehabilitation may more readily
accept PN than EN support. However, this is a last resort
because PN is more expensive and carries increased risk for
complications.7
437
tubes.27 There were no documented cases of NEC or intestinal perforation. Although the parenteral group achieved
goal calories first (3.07 days 2.1 days for PN; 4.25 days
2.6 days for enteral), the benefits of EN far outweigh the
risks of PN. Pettignano et al concluded that vasoactive drug
infusions should not be considered a contraindication to
enteral feeding.27
Macronutrients
Parenterally, carbohydrates are administered in the form
of dextrose, which provides 3.4 kcal/g. Protein is administered as a crystalline amino acid solution, which provides
4 kcal/g. Specialized amino acid solutions are available for
infants and children; these include TrophAmine 6% and
10% (B.Braun), Premasol 6% and 10% (Baxter Healthcare),
and Aminosyn-PF 7% and 10% (Hospira). These specialized solutions contain high concentrations of essential
amino acids, including histadine and tyrosine; low concentrations of phenylalanine, methionine, and glycine; as well
as glutamic acid, aspartic acid, taurine, and N-acetyl-Ltyrosine. The amino acid solutions for infants are designed
to replicate plasma amino acids in breastfed infants. They
also have a lower pH, which allows higher levels of calcium
and phosphate to be added to the solution.
Using a more concentrated amino acid solution allows
for a smaller volume to be administered, which then allows
for a greater volume to be allotted for other solutions (ie,
dextrose), as well as electrolytes, minerals, trace elements,
vitamins, and intravenous fat emulsion (IVFE). Using a
less concentrated amino acid solution may sometimes be
necessary because of the difficulty in measuring extremely
small volumes of the more concentrated solutions. When an
infant or child is severely fluid restricted, it may be difficult
2010 A.S.P.E.N. www.nutritioncare.org
438
B. Braun (McGaw),
Bethlehem, PA
Hospira (Abbott),
Abbott Park, IL
Baxter,
Deerfield, IL
TrophAmine
July 20, 1984
Aminosyn PF
Sept. 6, 1985
PremaSol
June 23, 2003
0.820
1.400
0.820
1.200
0.340
0.480
0.420
0.200
0.780
<0.016
<0.024
0.480
0.240
0.044
0.240
0.760
1.200
0.677
-0.180
0.427
0.512
0.180
0.673
--0.312
0.044
---
0.820
1.400
0.820
-0.340
0.480
0.420
0.200
0.780
<0.016
-0.480
0.240
---
0.540
1.200
0.680
0.380
0.360
0.320
0.500
0.025
<0.050
0.698
1.227
0.812
0.495
0.385
0.527
0.820
0.070
0
0.540
1.200
0.680
0.380
0.360
0.320
0.500
0.025
0
5.5 (5.0-6.0)
875
100
15.5
97
5.5 (5.0-6.5)
788
100
15.2
100
5.5 (5.0-6.0)
865
100
15.5
--
5
97
<3
None
46
None
-94
<3
Reprinted with permission from: Nieman Carney L. Neonatal Nutrition Support (CEU Self-Study Course),
Nutrition Dimension, Ashland, OR. 2009.
2010 A.S.P.E.N. www.nutritioncare.org
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
439
Comparing ofofSelected
Comparison
SelectedIntravenous
IntravenousFat
Fat Emulsions
Emulsions
Product and
distributor
Intralipid
10% and 20%
(Clintec)
Liposyn II
10% and 20 %
(Abbott)
Liposyn III
10% and 20%
(Abbott)
Soybean
Soybean
50
26
10
9
3.5
65.8
17.7
8.8
4.2
3.4
54.5
22.4
10.5
8.3
4.2
1.1
2.0
1.2
2.25
1.1
2.0
1.2
2.5
1.1
2.0
1.2
2.5
260
260
276
258
284
292
Oil base
Fatty acid content (%)
Linoleic
Oleic
Palmitic
Linolenic
Stearic
Calories/ml
10%
20%
Egg yolk phospholipids (%)
Glycerin (%)
Osmolarity mOsm/L
10%
20%
Source:Drug
Drugfacts
facts
and
comparisons.1998
1998ed.
ed.St.St.Louis:
Louis:
Wolters
Kluwer
Co.
Source:
and
Comparisons.
Wolters
Kluwer
Health.
Reprinted with permission from: Nieman Carney L. Neonatal Nutrition Support (CEU Self-Study Course),
Nutrition Dimension, Ashland, OR. 2009.
440
Advance By
Infants (< 1 y)
Protein (g/kg/d)
CHO (mg/kg/min)
Preterm
1.53
57
Term
1.53
69
Fat (g/kg/d)
12
12
Children (110 y)
Protein (g/kg/d)
CHO (mg/kg/min)
Fat (g/kg/d)
Adolescents
Protein (g/kg/d)
CHO (mg/kg/min)
Fat (g/kg/d)
Goals
Preterm
1
12.5% dextrose
per day
0.51
Term
1
12 or 2.55%
dextrose per day
0.51
Preterm
34
812 (Max 1418)
Term
23
12 (Max 1418)
33.5 (Max
0.17g/kg/hr)
3 (Max 0.15g/
kg/hr)
12
10% dextrose
12
1
5% dextrose per day
0.51
1.53
810
23
0.81.5
3.5 or 10% dextrose
1
1
12 or 5% dextrose per day
1
0.82.5
56
12.5
Electrolytes
Close monitoring of electrolyte status is essential. In
newborns, the addition of electrolytes to PN may be
deferred until the second day of life in some cases. Potassium is generally added once normal kidney status and good
urine output are established, and sodium is often added once
diuresis begins.22 Daily adjustments to electrolyte intake
are often necessary. Electrolyte requirements of preterm
and full-term infants are generally similar, with the exception of calcium and phosphorus. Electrolytes are typically
prescribed per kilogram in infants and younger children,
and as units per day in larger children and adolescents.
Table 34-5 PN Electrolyte Dosing Guidelines*
Electrolyte
Preterm
Neonates
Infants/
Children
Adolescents &
Children > 50kg
Sodium
Potassium
Calcium
Phosphorus
Magnesium
Acetate
Chloride
2-5 mEq/k
2-5 mEq/k
1-2 mEq/kg
2-4 mEq/kg
2-4 mEq/kg
1-2 mEq/kg
0.5-4 mEq/k
10-20 mEq/d
2-4 mEq/kg
0.5-2 mmol/kg 10-40 mmol/d
1-2 mmol/kg
0.3-0.5 mEq/kg 0.3-0.5 mEq/kg 10-30 mEq/d
As needed to maintain acid base-balance
As needed to maintain acid base-balance
Multitrace-4
Neonatal
Multitrace-4
Pediatric
Multitrace-5
Concentrate
(Adolescent/
Adult)
1.5 mg
0.85 mcg
1 mg
1 mcg
5 mg
10 mcg
none
none
60 mcg
0.1 mg
25 mcg
0.1 mg
25 mcg
1 mg
0.5 mg
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
441
Preterm Neonates
< 3 kg (mcg/kg/d)
Children 10-40 kg
(mcg/kg/d)
Adolescents > 40 kg
(per day)
Zinc
Copper
Manganese
Chromium
Selenium
400
20
1
0.05-0.2
1.5-2
50-250
20
1
0.2
2
50-125
5.0-20
1
0.14-0.2
1.0-2
2-5 mg
200-500 mg
40-100 mcg
5-15 mcg
40-60 mcg
NAG-AMA Recommendations
Weight (kg)
<1
3-Jan
>3
Weight (kg)
< 2.5
> 2.5
Dose (mL)
1.5
3.25
5
Dose (mL)
2 mL/kg
5 mL
442
Complications of PN
Short-term potential adverse effects of PN include the
following: infection, hyperglycemia, electrolyte abnormalities, disturbance of acid-base balance, hypertriglyceridemia,
phlebitis, bacterial translocation, and compromised gut
integrity. With long-term PN support adverse effects
may include infection, PN-associated cholestasis, metabolic complications, disturbance of acid-base balance,
osteopenia, risk of vitamin/mineral deficiency or toxicity,
and continued risk of bacterial translocation. Nosocomial
infections appear to result either from improper care of the
catheter and/or frequent use of the catheter for purposes
other than delivery of nutrients (eg, blood draws, medication administration).1
Monitoring
Prior to initiation of PN support, it is recommended to
check the following biochemical indices: basic metabolic
panel (BMP), calcium, magnesium, phosphorus, liver function tests (ie, Alk Phos, ALT, AST, GGT), total bilirubin,
conjugated or direct bilirubin, prealbumin, albumin, and
triglyceride. Following the initiation of PN the patients
require close biochemical monitoring. The monitoring
is then slowly decreased in frequency depending on the
patients demonstration of stable parameters. Patients on
long-term PN, such as those with SBS, need regular monitoring of vitamin and mineral status.
Aluminum in PN
It has come to light that various products utilized during
PN compounding contain high levels of aluminum, which
can be especially dangerous for infants and children.
Preterm infants are extremely vulnerable to aluminum
toxicity due to immature renal function and the likelihood
for long-term PN. 36 As of July 2004, the Food and Drug
Administration (FDA) mandated that products used in
compounding PN should state the aluminum content on
the label. The FDA identified 5 mcg/kg/d as the maximum
amount of aluminum that can be safely tolerated and
amounts exceeding this limit may be associated with central
nervous system or bone toxicity. 36 It is essential for pharmacists, dietitians, physicians, and nurses to collaborate
to minimize the use of higher aluminum content products. However, it is difficult to achieve the recommended
aluminum intake level set by the FDA when patients are
receiving multiple medications and PN.
Providing PN is wrought with unique potential risks
and complications and calls for a diligent interdisciplinary
team approach. Initiating EN as soon as medically appropriate is the key to minimizing the potential adverse effects
of PN support.
Feeding Teams
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
443
444
Figure 34-1
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
445
Conclusion
Every child should be evaluated individually when determining the best way to provide optimal nutrition to promote
growth and development. What works for one child and
caregiver may not work for another. One must consider
many factors when initiating nutrition support including,
but not limited to, the patients baseline nutrition status, the
functionality of the GI tract, access options, administration
schedule, and modality. In pediatrics, one must consider the
effect that the nutrition support regimen will have on the
infant/child and the caregivers. It is essential to be flexible
and supportive when working with caregivers to develop a
nutrition plan of care that is best for their child and their
lives.
1. Which of the following are common behavioral strategies to address picky eating?
A. Offer foods on a divided plate
B. Offer 1 new or non-preferred food with 1 or 2
preferred foods
C. Be consistent
D. Encourage family mealtimes
E. All of the above
2. All of the following findings may justify nutrition
support in a toddler except:
A. Height at the 25th percentile on the growth curve
since infancy
B. No weight gain for 4 months
C. Decrease in 2 weight percentiles channels over the
past 6 months
D. Eats very slowly
3. A child with CF who has a very good appetite, yet is
moderately underweight, should receive:
A. PN in addition to oral feedings
B. Tube feedings only
C. No nutrition intervention
D. Oral nutrition supplements in between meals
See p. 487 for answers.
446
References
PARENTERAL AND ENTERAL NUTRITION SUPPORT: DETERMINING THE BEST WAY TO FEED
447
43. Tobin S, Cheng V, Schumacher C, et al. The role of an interdisciplinary feeding team in the assessment and treatment of
feeding problems. Building Block for Life. 2005;28(3):111.
44. Satter E. Child of Mine; Feeding with Love and Good Sense. Bull
Publishing; 2000.
Additional Readings
35
Beth Lyman, RN, MSN, Jennifer M. Colombo, MD, and Jodi L. Gamis, OTR
CONTENTS
Learning Objectives
Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 446
Parenteral Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
Type of Intravenous Catheter Needed
Site Care
Safety Issues
Nursing Care
Enteral Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Enteral Route Selection
Types of Tubes
Tube Insertion
Verification of Placement
Bolus vs. Continuous Feeds. . . . . . . . . . . . . . . . . . . . . . . .
Bolus Feedings
Continuous Feedings
Combination Feedings
Initiation and Advancement of Feedings . . . . . . . . . . . . .
Bolus Feedings
Continuous Feedings
Combination Feedings
Safety Issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Preparation
Hang Time for Formula
Feeding Pumps
Nursing Care. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Securing the Tube
Site Care
Checking Residual Volumes
Flushing the Tube
Use of Sterile vs. Non-Sterile Water
Reflux
Aspiration Precautions
Intake and Output
Weights
Assessment of Feeding Tolerance
Medication Administration
Oral Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
448
449
Background
452
452
453
454
456
Parenteral Nutrition
449
450
Table 35-1 Comparison of Techniques Used to Obtain Blood Samples from a VAD
Type of Technique
Description
Pros
Cons
Traditional
1. Stop infusion
2. Flush VAD with NS
3. Obtain a waste specimen of 35 mL
4. Obtain blood needed for labs ordered
5. Flush VAD with NS
6. Restart infusion
1. Fill a syringe with heparin and remove it
2. Stop infusion
3. Flush VAD with NS
4. Obtain a waste specimen of 35 mL
5. Have a nurse rotate the waste specimen
6. Obtain blood needed for labs ordered
7. Return the waste specimen to the patient
8. Flush VAD with NS
9. Restart infusion
1. Stop infusion
2. Aspirate 35 mL of blood into a syringe
3. Reinfuse that blood and repeat the aspiration/
reinfuse procedure a total of 3 times
4. Place a new syringe on the VAD and obtain blood
needed for ordered labs
Push-Pull Method
Site Care
Once the CVC is in place, routine skin disinfection and
dressing changes are necessary. In 2002, the Guidelines for
the Prevention of Intravascular Catheter Infections recommended the use of chlorhexidine gluconate (ChloraprepTM )
for skin antisepsis for all patients over the age of 7 days or 26
weeks gestation.15 Since that time, this product has become
the standard antiseptic used for routine VAD dressing
change skin antisepsis in this country. It is generally well
tolerated. Transparent dressings that are replaced every 7
days or when loose or soiled are the dressing of choice over
gauze and tape as the insertion site can easily be inspected.
A trial of chlorhexidine gluconate-impregnated dressings
(BiopatchTM) in neonates resulted in contact dermatitis
making it not recommended for use in this population.16
Another key aspect of the dressing is to place a stress
loop in the VAD if it is tunneled or in the extension tubing
and tape to provide another method to prevent accidental
dislodgement of the CVC.
Safety Issues
BSIs related to the CVC are known to be associated with an
increase in cholestasis in infants on PN.17 Therefore, many
measures must be in place to assure adequate infection
control processes to prevent CVC-related BSIs. Table 35-2
2010 A.S.P.E.N. www.nutritioncare.org
summarizes these recommendations. Aside from institution processes to safeguard the patient, individual patients
will require infection prevention measures as well. Attempts
must be made to prevent contamination of the catheter hub
by draining gastrostomy tubes, a leaking ostomy bag, or
other body fluids. Babies who are teething need to have a
pacifier to prevent them from sucking on their IV tubing.
Children who are suspected or known to be self-destructive
should be carefully monitored. Siblings need to be supervised when visiting their brother or sister.
For children who have a parent in the room at all times
monitoring of the parent and any activity with the CVC is
warranted. Parents should not be allowed to access or care
for a CVC unless specifically trained to do so and only with
the permission of the nurse.
One of many reports in the literature described the use
of a CVC by parents with Munchausen Syndrome by Proxy.18
This condition is now referred to as Pediatric Condition Falsification which is a form of child abuse where the caregiver is
the perpetrator. Because this condition can be life threatening for the child, any suspicion of this condition needs to
be addressed prior to placement of a CVC for PN. While this
condition is difficult to prove, certain typical behaviors seen
in the caregivers include doctor shopping to different health
care facilities, discrepancy between reported symptoms and
451
Action
Hand Hygiene
Use an antiseptic skin soap to wash hands before placing or accessing a VAD or
use an alcohol-based hand sanitizer.
Staff involved in CVC placement must be competent to do this procedure.
Use maximal sterile barriers including masks, gowns, drapes, sterile gloves, and a kit with all
needed supplies.
Allow nursing to call a time out for an observed break in sterile technique.
Avoid placement of a CVC in a febrile patient.
Avoid guidewire replacement of a CVC.
Vigorously clean the hub of the catheter with 70% alcohol or chlorhexidine wipes before entering the line.
Have a policy on routine injection cap changes.
Avoid stopcocks on CVCs.
Use sterile, prefilled syringes for CVC flushing.
Limit accessing for labs to 1 time per day if possible.
Use a kit with all supplies needed.
Use ChloraprepTM as preferred skin disinfectant.
Use a mask and gown for PICC dressings.
Use a transparent dressing preferentially over a gauze and tape dressing to allow for the site
to be assessed.
Restrain the patient as needed to prevent accidental dislodgement.
Place a stress loop in the VAD tubing or extension tubing.
Obtain a peripheral and central blood culture.
Staff obtaining blood cultures should be specially trained and competent to avoid contamination.
Draw a minimum of 1mL for the blood culture.
Use clinical and other labs to validate diagnosis.
Do not necessarily remove the CVC.
Lipidsevery 24 h
PNevery 72 h unless a 3-1 solution, then every 24 h or if cycled off a few hours/day
Evaluate all supplies used on CVCs for defects that could lead to infections.
When using outside pharmacies, surveillance of possible IV fluid related infections is needed.
Have a surveillance plan within the institution to trend infections and look for a root cause.
Catheter Insertion
Catheter Accessing
Diagnosis of BSI
IV Tubing Changes
Other
Nursing Care
In addition to prevention of VAD-related infections, the
nursing care of a child requiring PN is extensive. Table 35-3
summarizes the nursing activities required when taking care
of a child who requires PN. Most hospitals and other care
settings require competencies in the areas of CVC dressing
changes, obtaining lab specimens, and appropriate flushing,
etc. Meticulous nursing care for any patient receiving PN is
absolutely essential in the pediatric care setting.
Enteral Nutrition
Enteral Route Selection
Enteral nutrition should be considered in all children with
an intact and functional gastrointestinal (GI) tract who can
not take in enough calories orally to meet their needs for
growth and development. Ideally a multidisciplinary team
including a pediatric physician, a nurse, a dietitian, a speech
therapist, and/or an occupational therapist should assess
each patient and develop a feeding plan. Balancing caloric
needs with the childs ability to digest and absorb nutrients
is a dynamic process that may necessitate low rates of EN
via feeding tube.
Once a clearly identified indication for enteral tube
feeding has been established, several ways to administer EN
exist. Enteral access can be obtained through a nasogastric
2010 A.S.P.E.N. www.nutritioncare.org
452
Frequency
As needed
As needed
As needed but usually at least every 12 h
Every shift and as needed
At least every 4 h and more often if warranted
At least every 4 h and more often if warranted
After every interruption in TPN
As needed
Within 24 h of drawing blood and per hospital policy
Daily
At all times
Daily
Hourly
As needed
As needed
Per hospital policy
Ongoing
Types of Tubes
Nasogastric Tube
Nasogastric (NG) tubes are indicated for children who have
an intact and functional GI tract and will only require enteral
formula for a short period of time. These patients should have
minimal gastroesophageal reflux, normal gastric emptying,
and low risk of aspiration.20 NG tubes are soft flexible tubes
made of Silastic or polyurethane ranging in size from 5 Fr.
to 12 Fr. The tube size is chosen based on the patients age,
size, and type of formula needed. Small-diameter tubes are
more comfortable for the patient, but can become clogged,
particularly if the patient receives fiber-containing formula
or additives to the formula.
453
stomach because of delayed gastric emptying, gastroesophageal reflux, or risk of aspiration.20 These must be
administered as continuous drip feedings.
Jejunostomy Tube
Jejunostomy (J) tubes are surgically placed jejunal tubes.
These tubes can be inserted directly into the jejunum endoscopically or surgically. Typically, they are used in patients
who will require enteral nutrition access for the small bowel
for more than 6 months.20 The negative side to jejunostomy
tube use includes mandatory continuous feedings as well as
surgical emergencies (volvulus) around the tube.
Tube Insertion
Most NG tubes are placed at the bedside. An attempt is made
to make the child as comfortable as possible. An infant or
young child should be swaddled for comfort and to prevent
their arms from flailing about during tube insertion. The
cervical spine of the child should be slightly flexed and not
hyperextended.21 A small amount of lubricant is applied to
the tip of the tube or water may be used. The tip is inserted
into a nostril and then slid carefully down the nasopharynx,
oropharynx, esophagus, and into the stomach. It is then
secured into place at the nares. Parents are taught the technique of inserting NG tubes so the tubes can be replaced at
home when necessary.
Nasojejunal tubes are inserted in a similar manner;
however, they usually require the assistance of radiology or
fluoroscopy for accurate placement or confirmation. These
tubes cannot be replaced at home. If the tube migrates back
into the stomach, it must be replaced.
Gastrostomy tubes, gastrojejunostomy tubes, and
jejunostomy tubes must be placed endoscopically by a
pediatric gastroenterologist, radiologist, or by a pediatric
surgeon. If a tube is dislodged, families should be instructed
to insert a replacement tube or temporary tube in the tract
immediately to avoid premature closure of the tract until a
more permanent tube can be replaced.
Verification of Placement
Verifying correct placement of an enteral tube is imperative
prior to administering enteral formula. A tube accidentally
inserted into the respiratory tract will certainly have disastrous or even deadly consequences.
Radiology
A chest and abdominal x-ray which identifies the length of
the tube is the gold standard for confirming placement. The
tip of the feeding tube can be seen within the stomach or
2010 A.S.P.E.N. www.nutritioncare.org
454
Enteral formulas can be given through bolus feedings, continuous feedings, or a combination of both depending on a
number of factors. These factors include the childs condition
and nutrition status, anatomy, type of tube selected, indication for tube feedings, and family schedule. It is important to
consider family lifestyle when implementing a feeding plan.
The goal is to maximize the benefit of enteral feeding while
minimizing the complexity of nutrition support.
Bolus Feedings
Bolus feedings imitate regular mealtimes and, therefore,
are more physiologic compared to continuous feedings. A
feeding schedule is determined by a dietitian and pediatric
physician depending on the childs caloric and fluid requirements. Bolus feedings can only be administered into the
stomach and should never be administered past the pylorus.
Formula can be administered by gravity or over a pre-determined period of time by a feeding pump. Bolus feedings are
often more convenient for the patient and family because
the schedule is more flexible and the child is not tethered to
an infusion pump.
Continuous Feedings
Continuous feedings are infusions of enteral formula over
time. Patients, who cannot tolerate large volumes because
of delayed gastric emptying, GERD, or dumping syndrome,
are given slow continuous feeds. The continuous feedings
are generally smaller volumes infused slowly. A feeding
pump is required for these infusions which may limit the
childs ability to move. For ambulatory patients, there are
smaller feeding pumps that can be carried in a backpack
allowing for more flexibility and mobility.
Combination Feedings
Combination feedings are ideal for patients who need a
significant amount of calories but cannot tolerate large
volumes. Smaller bolus feedings can be given during the
day and the remainder can be administered continuously
overnight. This method also allows the child and parents to
sleep at night.21
Bolus Feedings
The total calculated volume of formula is based on the childs
caloric needs and requirements. This volume is divided into
6 to 8 boluses. It is recommended to give 25% of the childs
caloric needs on day 1 and advance by 25% each day until
the final caloric goal is achieved.21 The child is monitored
closely for tolerance to the increased volume. Signs of intolerance include vomiting, abdominal distention, or pain.
Bolus feedings are given over 15 to 20 minutes, however,
longer times may be required.
Once the child is doing well with the initial feeding
schedule and is meeting the caloric goal, boluses can be
compressed to 4 to 6 feedings. This allows for ease and
flexibility of administration by the family at home. Again,
the child is monitored for tolerance during this transition.
Boluses may need to be given more frequently in infants or
children who have metabolic disorders or increased caloric
needs (ie, burns, catch-up growth).
Continuous Feedings
It is recommended to start continuous feedings at 1 to 2
mL/kg/h for 24 hours and increase by 0.5 to 1 mL/kg/h
every 8 to 12 hours until the final caloric goal is met. 22
When the final goal is achieved, compressing the feeding
into a shorter time interval should be considered so the
child can have a few hours each day free from the feeding
tube. Generally 16 to 18 hours each day should enable a full
daily caloric requirement. The child is monitored closely for
tolerance to each increase in volume and during compression of feedings.
Combination Feedings
When a child cannot tolerate large volumes of bolus feedings, combination feedings should be considered. The total
daily caloric requirement is divided between 3 to 4 smaller
bolus feedings during the day followed by a continuous
infusion overnight. This can be accomplished starting with
continuous feedings and working up to the caloric goal.
455
Safety Issues
Preparation
Formula should be prepared by personnel and family
members who are trained to prepare formula in a clean
environment.21 Good hand washing is imperative prior
to preparation.21 The warm and nutrient-rich environment of enteral formulas promote the growth of bacteria.
Using appropriate handling, hygiene, and adherence to
guidelines for hang time will prevent most episodes of
contamination.26
Ready-to-feed and formulas from liquid concentrate
are and should remain sterile; however, bacteria and other
micro-organisms can populate rapidly if the right conditions
are met. Powdered, reconstituted formulas or those with
additives tend to grow more bacteria and therefore require
meticulous attention to principles of aseptic technique in
the preparation and administration of the formula. 21
Feeding Pumps
Feeding pumps are useful for delivering continuous feedings or slower infusions for bolus feedings. Some feeding
pumps are small enough to be worn in a small backpack
for ease of mobility and flexibility of schedules. The ideal
pediatric pump is not position sensitive, can be increased
in increments of 0.1 mL up to 5 mL/h, and is relatively
light weight. Providers and caregivers need to be aware
that enteral feeding pumps are not free of malfunctions.
2010 A.S.P.E.N. www.nutritioncare.org
456
Nursing Care
Securing the Tube
Nasoenteric tubes can easily be pulled out of the nose by an
infant or a child tugging on the tube, sneezing, or vomiting.
To prevent dislodgement, the tube is secured in place on the
childs face. It is recommended to fix the excess tubing to
the back of the infants or childs shirt (ie, with a tape tab
and safety pin). This helps to keep the tube out of sight and
prevents the child from grabbing it.
Transparent dressing is usually applied to the cheek on
the same side of the face as the nasoenteric tube. The enteral
tube is inserted as described previously. The portion of the
tube that remains outside the body is laid across the DuodermTM and then a transparent dressing is applied over the
tube and Duo-dermTM securing the tube to the face. This
method protects the childs skin from harsh adhesive tape
that may tear the skin.28
J tubes, G tubes, and low-profile skin-level G buttons
can also be pulled out by the child, although it is less likely.
Most types of permanent feeding devices have an internal
retaining device which may be a collapsible rubber stopper
or a water-filled balloon. Many permanent feeding devices
also have an external retaining device.
A similar technique can be used to secure a G or J tube
to the childs stomach. There are commercially available
products to help keep some tubes in place.
Site Care
Keeping the site clean and dry is the most important aspect
of caring for an enteral feeding tube. Washing the nose and
face with mild soap and water daily is enough to keep the
nasoenteric tubes clean.
Ostomy sites should be monitored for cleanliness
daily. Mild soap and water is enough to keep these sites
clean, gently scrubbing off any crusted areas. A 22 gauze
dressing that is cut half way through can be placed between
the device and the skin to keep the ostomy site dry if there is
some leakage. For the most part, a dressing is not required.
Antibiotic ointment may be needed if the site becomes
erythematous or tender. Caregivers are taught to apply the
ointment for up to 3 days and if the symptoms persist, a
physician should be contacted.
2010 A.S.P.E.N. www.nutritioncare.org
Reflux
Severe gastroesophageal reflux may be an indication for
enteral tube feeding when frequent emesis results in food
refusal and failure to thrive. Gastroesophageal reflux can
be frustrating and anxiety provoking for both the child
and caregivers. Frequent episodes of emesis are a source of
calorie loss in young children.
Reflux can worsen with enteral tube feedings. Children
are unable to self-limit their intake. Their stomach may not
be able to accommodate their goal volumes. Also, in older
children, the diet is changed from relatively thickened or
solid feeds to liquid formula, which can predispose them to
more reflux.20
Worsening reflux may be a sign of intolerance to a
feeding schedule or formula. Sepsis, respiratory embarrassment, or metabolic abnormalities have an impact on the
number of reflux episodes.
Aspiration Precautions
Infants and children lying flat are at increased of refluxing
and aspirating. It is important to position the infant or child
in a manner to prevent aspiration episodes with the head of
the child elevated at least 30 while receiving a feed.20,21 It
may also be necessary to feed the child at risk for aspiration
transpylorically.
457
Weights
Weights should be monitored daily during the initiation and
advancement phases of a feeding plan. The infant or child
should be weighed without clothing and on the same scale
every day at the same time. Ostomy bags should be emptied
just prior to the weight measurement. Individual weights
are less important than an overall weight trend.
Monitoring a childs weight at every follow-up visit
ensures that the child is receiving appropriate calories for
growth. If the child fails to gain weight or loses weight,
healthcare providers should investigate any potential
sources of calorie loss.
Medication Administration
Enteral feeding tubes can be utilized for medication administration. Some medications can be given via enteral tube
easily (eg, liquid suspension or solution, crushed solid
tablets, and capsules). More complex formulations including
extended or sustained released products are problematic.
2010 A.S.P.E.N. www.nutritioncare.org
458
Liquid forms of medications are preferred. Some solutions and suspensions may be too viscous or thick to safely
put through an enteral tube. These medications must be
diluted with sterile water. Suspensions must be shaken thoroughly prior to administration.21 Improper administration
can lead to inaccurate medication provision.
Medications in the solid form such as tablets or capsules
must be crushed or opened and mixed with purified water
prior to administering through an enteral feeding tube. It
is important to mix the medication well to prevent underdosing or overdosing of the prescribed medication.
After administering medications through an enteral
feeding tube, the tube must be flushed with purified water to
prevent the feeding tube from being clogged and to ensure
that the entire dose of medication is given to the child.
Caregivers should be aware of what medication is being
administered and where the medication is being administered. Certain medications including vitamins and minerals
require activation in the stomach or are predominantly
absorbed in the upper small intestine. Medications that alter
gastric pH may also affect the activity of concomitant medications. A consultation with a pharmacist is warranted when
deciding to give medications through an NJ tube or a GJ tube
as these tubes bypass the stomach and upper small intestine altogether. A pharmacist will also provide information
regarding medication compatibility if the decision is made to
add the medication directly to an enteral formula.21
Oral Nutrition
References
459
36
CONTENTS
Management of Pediatric Patients Receiving
Nutrition Support Therapy. . . . . . . . . . . . . . . . . . . . . . . . . 460
Nutrition Monitoring and Evaluation. . . . . . . . . . . . . . . . . 461
Enteral Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
Monitoring
Evaluation of Growth and Nutrition Adequacy
Evaluation of Formula Tolerance
Monitoring Laboratory Values
Oral Feeding
Revision of Enteral Plan
Parenteral Nutrition. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
Evaluate for Infusion-Related Incidents
Evaluate Tolerance of Volume
Evaluate Tolerance of Macronutrients
Monitoring the Integrity of PN Formulations
Comparison of 3-n-1 and 2-n-1 Formulations
Combination Enteral and Parenteral Nutrition
Reassessment of the Plan. . . . . . . . . . . . . . . . . . . . . . . . . 466
Learning Objectives
461
Enteral Nutrition
Monitoring
There are many parameters that can be used to monitor
patients receiving EN. During the initiation and advancement of EN, monitoring will be more frequent and become
less frequent as the child becomes more stable. A proposed
monitoring protocol for infants and children receiving EN
initially and then stable, both while in hospital and at home,
is provided in Table 36-1.9,10
462
Oral Feeding
Table 361 Suggested Parameters to Monitor for Infants and Children Receiving Enteral Nutrition9,10
Growth Parameters
Weight
NICU
Infants
Children
Length
NICU
Infants
Children
Height
(> 36 mo)
Head circumference
(< 36 mo)
Weight gain
Linear growth
Intake Parameters
Intake
Calories
Protein
Vitamins
Minerals
Fluid balance
GI Tolerance
Abdominal girth
Gastric residuals
Emesis
Stool
(volume, frequency,
consistency, color)
Ostomy
(volume, consistency)
Physical
Temperature
Tube placement
Tube site care
Initial Week
During Hospitalization
Outpatient EN-Only
Patient
Outpatient PN and/
or EN Patient
Daily
Daily
Daily
Daily
Daily
DailyTwice Weekly
WeeklyMonthly
WeeklyMonthly
Weekly to Every Clinic
Weekly
Weekly
Weekly
Baseline
Baseline
Baseline
Baseline
Weekly
Monthly
Monthly
Monthly
Monthly or at Clinic
Monthly or at Clinic
Baseline
WeeklyMonthly
Monthly or at clinic
Monthly or at clinic
DailyWeekly
Monthly
DailyWeekly
Monthly
WeeklyMonthly
Monthly
Weekly
Monthly
Daily
Weekly
Monthly
Weekly
As indicated
As ordered or reported
As reported
Daily as reported
As indicated
As ordered or reported
As reported
Daily as reported
As indicated
As ordered or reported
As reported
Report changes in stool
pattern
As indicated
As ordered or reported
As reported
Report changes in stool
pattern
Parenteral Nutrition
463
464
DehYDRATION
FLUID OVERLOAD
Rapid weight
Rapid weight
Intake: Intravenous
fluids, PN,
blood products,
medications, EN
Output: Urine,
gastric, stool, bile,
chest tube, wound,
skin
Laboratory
Decreased urine
output, dark urine
Increased urine
output in patients
with normal renal
and liver function
sodium, serum
osmolality, urine
specific gravity
albumin or Hgb
Vitals
Medications
Physical Exam
Blood urea
nitrogen, sodium,
serum osmolality,
urine specific
gravity, albumin
or Hgb
Heart rate,
losses with fever
Addition of diuretic
or change in
frequency
Thirst, dry lips, dry
mucous membranes,
dry skin, headache,
dizziness
Respiratory rate
Fluid retention with
steroids or excessive
sodium intake
Peripheral, facial,
and orbital edema,
abdominal girth,
shortness of breath
cycled 12 to 22 hours per day in an effort to ease daily activities and decrease hepatic complications related to PN. 23
Initially, blood glucose levels should be obtained during the
high rate of cyclic PN infusion and 1 to 2 hours after cycle
completion to evaluate for hyper- and hypoglycemia. Levels
> 150 mg/dL will require a decrease in dextrose amount or
lengthening of cyclic infusion to decrease the GIR. Levels
< 60 mg/dL or any symptoms of hypoglycemia will require
a longer taper period off of the cyclic PN or adjustment of
insulin if present in PN solution.24
Triglyceride levels are monitored daily as lipid intake is
increased and then decreased to weekly once dose is stable
and levels are adequate. Maximal lipid infusion for adults
is 0.125 g/kg/h.25 There is limited information regarding
maximal infusion rate for pediatrics but it is well-documented that longer infusion rates, 12 to 24 hours, improve
tolerance to lipid infusions.26 Decreasing the lipid dose or
infusing lipids every other day or 3 to 5 times a week instead
of daily in long-term PN patients will allow for further clearance of the lipid and possibly avoid triglyceride levels > 200
mg/dL. Limiting the dose and therefore decreasing phytosterol intake may also reduce the cholestasis commonly
noted in pediatric patients receiving long-term PN. 27 Calculating the lipid infusion rate in g/kg/h would be helpful if
hypertriglyceridemia is an issue.
465
466
Disadvantages
467
Compatibility of Medications
Physical Data
468
Trace Elements
(see also Chapter 6)
When a patient is receiving PN for several months, it is
important to assess the status of trace elements due to
the standardized way in which they are initially ordered
in PN patients and the known contamination of commercial products used to compound PN.70 After 3 consecutive
months on PN (unless clinically indicated earlier),71 a
patient should be assessed for zinc, selenium, manganese, copper, and chromium.45 Long-term use of standard
multi-trace products can result in both excessive and
deficient concentrations, especially in patients who have
abnormalities in the GI tract. Although measuring blood
concentrations of these trace elements is not necessarily
the most accurate due to the bodys ability to store these
elements, it is generally the easiest and least costly way to
monitor.72 It is also important to note that these elements
should not be checked during times of acute inflammatory
type conditions due to the bodys sequestration of some
elements during this time. Levels may be falsely decreased
in these types of situations.73
Assessment of these elements should occur every 3
to 12 months while receiving PN.45,72 If a patient is found
to have an elevated level of any standard element, the
multi-trace element of the PN can be omitted and separate elements may be added. Additionally, if any element
is found to be deficient, it can be added in addition to the
standard multi-trace. There are limits to concentrations of
trace elements in PN so this must be considered, and in that
situation, oral supplementation may be an option. After
altering the dosage of any element it should be rechecked
in approximately 1 to 3 months. If a patients condition
has completely stabilized, it is reasonable to monitor only
once per year.
Aluminum, considered an ultratrace element, is a
known contaminant of PN products. It is important to
monitor due to the patients continued exposure and risk
for toxicity. It is reasonable to check an aluminum level
each year during PN therapy. If the aluminum is elevated,
the PN pharmacist can look at the current PN components and make adjustments if possible since aluminum
contamination varies even between different PN product
manufacturers.74
Molybdenum deficiency is rare but can occur in a PN
patient, especially those with decreased intestinal absorption.75,76 If desired, a molybdenum level may be checked
yearly unless otherwise clinically indicated. If a deficiency
is detected, molybdenum may be supplemented via PN in
the form of ammonium molybdate.
469
Iodine is an element not supplied by standard multitrace products in PN. Therefore it is possible, although
unlikely, for a SNS patient to become deficient.77,78 This
can routinely be assessed with a yearly thyroid stimulating
hormone (TSH), triiodothyronine (T3) test, or urine
iodine concentration.
470
Vitamins
(see also Chapters 7 and 8)
Fat-soluble vitamins A, D, E, and K are standard components of most parenteral multivitamin products and EN
products. It is possible for a patient to accumulate excessive amounts due to the bodys storage of these vitamins,
but it is also possible for deficiency to occur with certain
disease states. Monitoring the levels of vitamins A, D, and
E once yearly is recommended unless otherwise clinically
indicated.45 Vitamin K status is more commonly assessed
by the PT as previously discussed.
The water-soluble vitamins including B1, B2 , B6, B12 ,
niacin, folic acid, pantothenic acid, biotin, and vitamin
C are also components of parenteral multivitamins and
EN. Routine monitoring of these vitamins is not indicated
(except for B12 and folic acid as previously discussed)
unless there is a clinical reason to believe that there is a
deficiency or toxicity.71
Infectious Complications
Table 36-7
471
Table 36-8
Parameter
Initial
Followup
Parameter
Initial
Followup
BMP
CMP
Magnesium
Phosphorous
Prealbumin
Triglycerides
PT/INR
CBC differential/
platelets
GGT
Daily
Weekly
Daily to Weekly
Daily to Weekly
Weekly
Daily to Weekly
Weekly
Weekly
Included in CMP
Every 1 to 4 weeks
Every 1 to 4 weeks
Every 1 to 4 weeks
Monthly
Monthly
Monthly
Every 1 to 4 weeks
Baseline if indicated
Monthly
Iron Studies
Zinc
Selenium
Manganese
Copper
Chromium
Vitamins A, D, and E
Vitamin B12 and Folate
Carnitine
TSH
3 mo
3 mo
3 mo
3 mo
3 mo
3 mo
6 mo
6 mo
3 mo
Baseline if indicated
Every 36 mo
Every 36 mo
Every 36 mo
Every 36 mo
Every 36 mo
Every 36 mo
Every 12 mo
Every 12 mo
Every 312 mo
Every 12 mo
Na
K
Ca
PO4
Mg
Cl
Ac
Zn
Cysteine
Vitamin K
Ranitidine
Famotidine
Adult
Pediatric
NICU
180
180
06
150
06
30
02
30
02
24
00.5
250
08
250
08
5
300
40
10
100
06
n/a
180
06
150
05
30
04.5
30
02
13
01
250
010
250
010
5
400
120
n/a
100
2
n/a
1000
500
50
2.5
10
1000
700
50
2.5
10
1000
700
50
Min 10 g/L or
DC Lipids
150
30
35
24
300
300
10
10
n/a
Folic Acid
Heparin
Vitamin C
Carnitine
Stability
mEq/L
mEq/kg
mEq/L
mEq/kg
mEq/L
mEq/kg
mmol/L
mmol/kg
mEq/L
mEq/kg
mEq/L
mEq/kg
mEq/L
mEq/kg
mg/L
Mcg/kg
mg/kg
***mg***
mg/L
mg/kg
mg/L
mg/kg
***mg***
mg/d
units/L
mg/d
mg/kg/d
Usual Dose
Comments
Osmolarity
X
X
X
X
X
X
X
X
X
X
X
X
X
X
Total zinc
or DC fat
mg/day
X
X
X
Not mg/L
X
X
X
X
Amino Acid
Min 20 g/L
Min 10 g/L or
DC Lipids
Dextrose
Min 50 g/L
Min 50 g/L
Min 50 g/L
Lipid
Min 5 g/L
Min 5 g/L
Min 5 g/L
Total dose
X
Need 20 g/L if
Dextrose
< 10% or DC
Lipids
DC Lipids if
< 50 g/L
DC Lipids if
< 5 g/L
472
Formula
Rate/Cycle
Route
MD:
Date:
wt_____ :____%ile
ht/lt_ __ :____%ile
wt/ht________%ile
dry wt__________
kcal/kg: ________________________________
g/kg: __________________________________
mL/kg:_ ________________________________
LABS
Date
Weight (kg)
Sodium / Potassium
Chloride / Carbon Dioxide
BUN / Creatine
Glucose / Calcium
Phosphorous / Magnesium
Total/conj. Bilirubin
Cholesterol / Triglyceride
AST / ALT
AlkPhos / GGT
Hgb / Hct
WBC / platelets
INR
Zinc
Albumin / Prealbumin
AA/Dex/lipid
Na (mEq/kg)
K (mEq/kg)
Cl (mEq/kg)
Anions (A,C)
Ca (mEq/kg)
P (mmol/kg)
Mg (mEq/kg)
Heparin
MVI:Peds/Adult
TMS:Peds/Adult
Zinc (mcg/kg)
Copper (mcg/kg)
Manganese (mcg/kg)
Chromium (mcg/kg)
Selenium (mcg/kg)
Carnitine (mg/kg)
Rate/Cycle
Total Volume/day
Total Calories/day
kcal/kg
GIR
NPC:N
Wt used for script
kcal/mL
Admit Date:
DX:
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
TPN SCRIPT
/
/
WA_______
HA________
BMI_______
IBW_______
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
/
ENTERAL
/
mL PN/EN
kcal / kcal/kg
% PN/EN
Protein (g)
Protein (g/kg)
UOP mL/kg/h
Emesis
Stool
Ostomy mL/d / mL/kg
Total mL
TOTALS
OUTPUT
473
D.O.B.
Physician:
Diagnosis:
Fax:
Phone:
Pager:
Enteral Formula:
Other Contact:
Family Contact:
Lab Location:
Date:
Every Month
Pre-Albumin
Triglyceride
GGT
MCV
Every 3 Months
Selenium
Manganese
Copper
Chromium
Zinc
Ferritin
Iron Serum
TIBC
Transferrin Saturation
Carnitine Total
Carnitine Free
Carnitine Ratio
Every 12 Months
Vitamin A
Vitamin D
Vitamin E
Vitamin B12
RBC Folate
TSH
Free T4
Aluminum
Recorded By:
Reviewed By:
Date Faxed to Physician:
474
References
475
476
65. Quigley EM, Marsh MN, Shaffer JL, Markin RS. Hepatobiliary complications of total parenteral nutrition.
Gastroenterology. 1993;104:286301.
66. Payne-James JJ, Silk DB. Hepatobiliary dysfunction associated with total parenteral nutrition. Dig Dis. 1991;9:106124.
67. McMillan NB, Mulroy C, MacKay MW, et al. Correlation of
cholestasis with serum copper and whole-blood manganese
levels in pediatric patients. Nutr Clin Pract. 2008;23:161165.
68. Duerksen DR, Papineau N. The prevalence of coagulation
abnormalities in hospitalized patients receiving lipid-based
parenteral nutrition. J Parenter Enteral Nutr. 2004;28:3033.
69. Deitcher SR. Interpretation of the international normalized
ratio in patients with liver disease. Lancet. 2002;359:4748.
70. Pluhator-Murton MM, Fedorak RN, Audette RJ, et al. Trace
element contamination of total parenteral nutrition. 1.
Contribution of component solutions. J Parenter Enteral Nutr.
1999;23:222227.
71. Jensen GL, Binkley J. Clinical manifestations of nutrient deficiency. J Parenter Enteral Nutr. 2002;26:S29S33.
72. Fuhrman MP. Micronutrient assessment in long-term
home parenteral nutrition patients. Nutr Clin Pract.
2006;21:566575.
73. Prelack K, Sheridan RL. Micronutrient supplementation
in the critically ill patient: strategies for clinical practice. J
Trauma. 2001;51:601620.
74. Poole RL, Hintz SR, Mackenzie NI, et al. Aluminum exposure
from pediatric parenteral nutrition: meeting the new FDA
regulation. J Parenter Enteral Nutr. 2008;32(3):242246.
75. Heimburger DC, McLaren DS, Shils M. Clinical manifestations of nutrient deficiencies and toxicities: a resume. In: Shils
ME, Shike M, Ross AC, Caballero B, Cousins RJ, eds. Modern
Nutrition in Health and Disease. 10th ed. Philadelphia, PA:
Lippincott Williams & Wilkins; 2006:595612.
76. Nielsen FH. Boron, manganese, molybdenum, and other
trace elements. In: Bowman BA, Russel RM, eds. Present
Knowledge in Nutrition. 8th ed. Washington, DC: ILSI Press;
2001:384400.
77. Ibrahim M, Morreale de Escobar GM, Visser TJ, et al.
Iodine deficiency associated with parenteral nutrition in
extreme preterm infants. Arch Dis Child Fetal Neonatal Ed.
2003;88:F5657.
78. Moukarzel AA, Buchman AL, Salas JS, et al. Iodine supplementation in children receiving long-term parenteral nutrition.
J Pediatr. 1992;121:252254.
79. Nagano T, Toyoda T, Tanabe H, et al. Clinical features of
hematological disorders caused by copper deficiency during
long-term enteral nutrition. Intern Med. 2005;44:554559.
37
CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 485
Clinical Care as a Team Approach . . . . . . . . . . . . . . . . . . 486
Approaching the Pediatric Patient: WhatShould
We Let Them Know?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 486
The Good Shepherds
The Liberators
The Educators
Learning Objectives
Introduction
478
The practice of medicine, even when dealing with decisions that raised fundamental ethical and social questions,
was historically the domain of an individual physician.
Physicians would decide if a critically ill baby had enough
potential to continue on the ventilator, whether an organ
should be given to a patient in heart failure, or if the elderly
womans pneumonia should be treated with antibiotics.
However, events such as abuses in human experimentation and the formation of a national commission to explore
medical ethics in the late 1960s and early 1970s began to
change the number of persons gathering around the bedside.
During the 1970s and 1980s, institutional review boards
(IRBs) examined research protocols to ensure subject
protection; orders to limit life-saving interventions began
to be documented and justified in the medical chart; and
ethics committees discussed situations that spurred moral
disagreement in the medical staff and/or the family. The
result has been a dramatic shift in how health care interacts
with the medical patient.1
Today, health care is not only affected by this history
but also the increasing role of the medical specialist. Any or
all of the following caregivers may be found participating in
the care of a sick child and her family: bedside nurse, charge
nurse, respiratory therapist, physician (including various
physician subspecialists and trainees), dietitian, physical
therapist, occupational therapist, speech pathologist, social
worker, case manager, psychologist, nurse practitioner,
physician assistant, chaplain, child life specialist, and music
therapist. The potential for benefit is high, as many professionals focus on specific aspects of that patients care, but
an obvious, and frequent, drawback is a lack of true teamwork. Most of the time, this problem originates from a lack
of defined roles mixed with poor communication; the result
may be conflicting messages presented to the family.
It is important to acknowledge that within the team lies
people with individual experiences and personal circumstances. Cultivating an environment where the team can
openly discuss concerns about patient care is a formidable,
but extremely valuable undertaking. As a participating
member of a health care team, it is each persons responsibility to form this team environment by communicating
effectively, understanding the underlying issues at hand,
and maintaining a complete picture of the goals and objectives of the clinical care of a patient.
The Liberators
Cassidy describes the liberators are those who fought to
counter the arguments of the Good Shepherds, especially
as researchers in the medical and social sciences made the
following important discoveries:
Withholding illness-related information from children
tends to increase their anxiety and fear. 3
Some sick children feel pressure to continue the faade
of not knowing about their illness in order to protect
their parents.4
Children are exceptionally observant and adept at
discovering the truth, regardless of the intent of parents
or health care providers to shield them from it. 5,6
Although a mature concept of death was thought to
be unattainable by a child, a majority of children reach
such an understanding by the age of 7 years.7
While this information argued for children to be given
information about their illness, the liberators erred in
assuming that the solution was to apply adult standards to
the pediatric population. The result was a standoff between
2 approaches that stood at opposing ends of the spectrum:
either tell children nothing in order to protect them from
a harsh reality, or tell children everything and potentially
bombard them with age-inappropriate information.
The Educators
A compromise is forged by approaching the patient in a
manner that makes use of a skill already rooted within the
practice of pediatrics: the assessment of a childs development. In this approach, caregivers interact with the pediatric
patient in a manner that is appropriate to her level of development; autonomy is not given fully and freely to the child,
but caregivers act in a manner that will inform and foster
the development of her increasing autonomy. This gradual
approach, especially in children with chronic, yet survivable
medical conditions (eg, diabetes) makes the transition to
adulthood more realistic than the sudden placement of full
responsibility upon a patient when she becomes an adult.
Used within this model is the concept of obtaining
assent from the pediatric patient. The American Academy
of Pediatrics (AAP) describes assent as having 4 elements:
1. Helping the patient achieve a developmentally appropriate awareness of the nature of his or her condition
2. Telling the patient what he or she can expect with tests
and treatment(s)
3. Making a clinical assessment of the patients understanding of the situation and the factors influencing
how he or she is responding (including whether there is
appropriate pressure to accept testing or therapy)
479
Another useful practice prior to discussing an ethical question, particularly one that focuses on the use or disuse of
a particular medical intervention, is to define the goals of
the act in question. For instance, when a child is placed on a
ventilator, certain goals are set for the usage of that intervention (although this process is rarely formalized). Often the
goal is the provision of oxygen and the removal of carbon
dioxide until appropriate lung healing, or maturation, can
take place. Once that goal is no longer served because the
2010 A.S.P.E.N. www.nutritioncare.org
480
a ventilator is difficult for all who care for the child, perhaps
the feeling that all natural caregiving responsibilities have
been fulfilled allows this to be more palatable limitation of
treatment than the withholding of nutrition.10
Another potential reason for the uniqueness of feeding
is the typical time course and physical appearance of a
person who dies from starvation or dehydration. This is not
to imply that starvation is not a natural part of the dying
process, in fact the bodys reaction to a deprivation of nutrition may actually serve to ease a persons death. However,
for families who are caring for a child who is not being
adequately fed, the visual appearance and the prolonged
time course prior to death may be too difficult to endure
regardless of any medical considerations. Additionally,
the supportive care programs that could provide palliative
care throughout the dying process for these children are
unavailable in many geographic locales.13,14 Therefore, it is
understandable why the medical provision of nutrition may
be seen as such an essential element of care.
481
be important in making certain choices about the appropriateness of medically delivered nutrition in the hospital or
at home. At the very least, open and transparent decisional
processes and communication with parents that allow for
their understanding of why, and how, and for how long a
certain method may be used, is required.
When surgically placed devices (eg, central venous
catheters, gastrostomy, or jejunostomy feeding tubes) are
considered, the risks and benefits require special attention
as the potential for pain, wound healing, aspiration, infection, thrombosis, or other complications must be weighed
against the patient and family goals. For most patients the
benefits are clear and the health care team makes reasonable decisions with the family to proceed with placement of
such devices. But on occasion, the initial placement, or their
replacement, become burdensome and cause all involved
to pause and reconsider their appropriateness. This may be
especially true in the terminal stages of many chronic and
debilitating conditions. In these cases, adequate time needs
to be devoted to discerning the best interests of the pediatric patientoften addressed differently by the patient,
parents, and clinicians. Clinicians sensitivity to parental or
patient goals and values that differ from their own, whether
in matters of culture and custom, faith and religious tradition, or social and family norms and expectations must be
present and allow for considerations beyond what may be
simply metabolically or physiologically apparent as a best
course of action. With long-term care and chronic debilitation, one may need to ask Does more always equate
to better?17
Perhaps the most heated ethical debates in nutrition
management of patients dependent upon medically delivered
nutrition and hydration are in the context of the withholding
or withdrawal (WH/WD) of such nutrition. For children,
especially those in whom a cognitive capacity is not appropriate for them to participate in decisions, parents normally
serve as surrogate decision makers as they are entrusted by
society with discerning and acting upon their childs best
interests. These interests, though not always knowable,
certainly include not being treated as mere passive objects,
and the same ethical and legal posture toward WH/WD
medical nutrition is due them that is due adult patients.10
Decisions that are made concerning medical nutrition
should be made like all other decisions about WH/WD
life-sustaining treatments, although as previously noted
these decisions may be fraught with greater psychological
and emotional duress among all partiesespecially when
the patient is not imminently dying.18, 19 As noted by Porta
and Frader, clinicians should neither demand nor reject
2010 A.S.P.E.N. www.nutritioncare.org
482
Palliative Care
The subject of palliative, and end-of-life, care has been of
increasing interest in pediatric medicine over the past decade.
Among resources that address what pediatric palliative care
should entail, the 2003 Institute of Medicine Report When
Children Die is among the most often cited.20 Among issues
noted therein, the attention to medically administered [artificial] nutrition and hydration (ANH) is noted as one that
warrants acknowledgement by clinicians of some residual
controversy and continued need for inquiry. The recent AAP
Clinical Report, Forgoing Medically Provided Nutrition and
Hydration in Children, helps by providing further guidance
in this sensitive area.21 Both of these resources, and numerous
articles that have been published in the peer-reviewed
medical literature, address aspects of ANH in the context
of life-limiting or threatening conditions, and specifically at
the end of life.22,23 Matters of values, open communication,
assigned meanings to feeding, and the overall goals of care
require attention in addressing this matter as much as any
other life-sustaining interventions.
The dietician may need to be a resource for parents,
families, other care team members, and for physicians in
particular as the issue of ANH is examined in the acute care
setting (hospital or clinic), home health, or home palliative and
hospice care. Attention to the normalcy of assisted feeding
devices, such as gastrostomy feeding tubes, is another sensitive
area that may require education as well as a values clarification. Petersen and colleagues reported a different perspective
on feeding through a gastrostomy tube by a majority of daily
caregivers of children with cerebral palsy.24
On occasion an ethical dilemma may arise in determining that the placement of a medical nutrition device
(feeding gastrostomy tube), is in the best interests of the
patient. The dietician as a key interdisciplinary health care
team member, or consultant, may bring clarity to decisions
to go forward with such a plan, or to reconsider it. Goals
clarification may be the greatest contribution to be made.
The dietician may also be called to advocate for correct
ethical action in beginning, sustaining, or advancing nutrition support for special needs children. She may find herself
in tension with primary or specialty clinicians who have
not prioritized nutrition for the NICU graduate, missed
faltering growth, or are uncertain about what to do next
for a child in whom a medical feeding device is not being
used and oral nutrition is inadequate to secure the goals of
health. Ethical action here requires professional competence, self-confidence, and a stance of advocacy for the child.
Effectively communicating with involved clinicians, and
parents, may include the provision of past and present goals
of nutrition, observed and documented failures (growth
curves, illness, and other outcomes), and recommendations
with evidence-based expectations for measurable outcomes
in an appropriate timetable.
Additionally, some NICU graduates have chronic
gastrointestinal illnesses such as cholestatic jaundice, liver
disease, or short bowel syndrome (SBS) following neonatal
surgery. The care of the child with SBS requires attentiveness to detail and elements of psychosocial support for the
child and family that complement the necessary medical,
nutrition, and pharmacy (home parenteral nutrition)
management of this condition. Those who await eventual
liver-bowel transplantation have lengthy waiting periods
and high morbidity and mortality.27
Other children with special nutrition needs include
those born with an inborn error of metabolism. Perhaps
diagnosed prenatally, or even after discharge home from the
hospital, many metabolic conditions result in acute illness
and nearly all have significant impact upon child growth
and development. The nutritionist must remind himself that
he is part of a team and work collaboratively in addressing
parent education, child health, and the provision of special
nutrition products. In extreme cases, such as certain urea
cycle disorders, liver transplantation may be the ultimate
goal and nutrition assessment and daily management brings
the nutritionist in close and frequent contact with the family.
Should such contact lead to shared feelings about the childs
illness, goals, and values, it is the nutritionists duty to raise
issues that might seem contrary to the goals of the current
care plan with the responsible physician or team leader.
Childhood Obesity
The recent increase in childhood obesity may be seen by
some as a mere medical, or strictly nutrition, condition.
This is far from the reality of complex social, behavioral,
educational, and even economic contributors to this
growing concern. Some have raised the issue of obesity
constituting medical neglect, and thereby warranting interventions that include social and legal actions akin to those
expected in physical or sexual child abuse, or gross parental
neglect of meeting a childs due needs (eg, food, clothing,
shelter, education, and health care). Recently, Varness and
colleagues have addressed the nature of these concerns and
how they are rarely borne out in such a manner that obesity
could legitimately be considered neglect. 30
The Adolescent
The period of adolescence is accompanied by major developmental and cognitive goals, not the least of which are
individuationthe process in which the young person
engages in developing his or her own self-identity and
strives to become an individual, distinct from parents and
siblingsand independence, whereby the young person
takes actions that allow for not only a separate identity
483
Summary
The timely, appropriate, and beneficial provision of pediatric nutrition requires attention to the pediatric patient and
his or her parents and family. Ethical challenges may best
be met with open dialogue, respectful listening, clarity of
communication, and sensitive provision of support directed
toward mutually derived nutrition goals. The dietician has
an integral role to play in these matters as a valued interdisciplinary team member across diverse diagnostic categories,
care environments, and the age continuum of pediatrics.
Case Studies
A Decision Not to Start Medical Nutrition Support?
A term infant is delivered by emergency cesarean section
for fetal bradycardia to a 25-year-old gravida I mother
with diabetes mellitus. He required extensive resuscitation
and had Apgar scores of 0, 0, and 3 at 1, 5, and 10 minutes
after birth, respectively. He is admitted to the NICU with
respiratory distress and hypoglycemia. His birth weight is
4.5 kg. He has poor cardiovascular function, hypotension,
and metabolic acidosis. His respiratory depression and
apnea require assisted ventilation. At 2 hours of age he has
a convulsion and after receiving an anticonvulsant he is
placed on a head-cooling protocol for severe birth asphyxia.
An MRI confirms diffuse and devastating ischemic brain
injury. On postnatal day 4 he fails a trial of extubation due
to apnea and inability to handle his own secretions. He has
2010 A.S.P.E.N. www.nutritioncare.org
484
no urine output for 7 days and after receiving initial intravenous dextrose the question of providing parenteral nutrition
or a trial of nasogastric tube feedings is raised on morning
ICU rounds. A discussion follows in which his prognosis
for recovery is described as bleakhis future survival is
contingent on the provision of a tracheostomy and placing
a feeding gastrostomybut his potential for future neurological function (motor, cognitive, and communicative) and
development is gone and he will remain infantile.
This infant presents a number of opportunities for
evaluating nutrition support: a large for gestational age
infant of a diabetic mother (complicated by hypoglycemia),
birth asphyxia (with circulatory shock, acidosis, and likely
impaired gastrointestinal perfusion, mucosal barrier function, and poor gut motility), neurological injury, and renal
failure. At a week of age he evidences multiple organ system
injury and failure of function. His prognosis for successful
oral feeding is nil, and in the face of renal failure the provision of parenteral nutrition must be questioned.
1. What is the ethical rationale for placing a central line
and providing parenteral nutrition?
2. What is the burden:benefit analysis of pursuing a
feeding gastrostomy and fundoplication?
3. What are the goals of nutrition support for this baby?
4. Can palliative care be provided without a specific
feeding regimen?
References
485
486
Chapter 4: Fats
Chapter 6: Minerals
1.
2.
3.
4.
488
4.
5.
6.
7.
489
490
milk is the most important therapy. A protein hydrolysate formula would be an effective substitute and if
not tolerated, an elemental formula.
491
492
493
494
Index
A
Abdominal injuries, 381382
Absorption
of carbohydrates, 20
of fats, 26
of minerals, 4647
of proteins, 3334
of vitamins, 57
Academy of Breastfeeding Medicine,
121
Acanthosis nigricans, 154
Acute kidney injury (AKI)
classification
postrenal, 257
prerenal, 257
renal, 257
defined, 269
electrolyte and fluid restrictions,
270
neonatal issues, 271272
nutrition assessment, 270
Adolescents
acne treatment, 78
calcium requirements, 413
dietary habits, 412
nutrition assessment, 412414
protein requirements, 40
Adrenal glands, injury to, 381
Alagille syndrome, 304, 305
metabolism
role of kidney, 39
role of liver, 3839
and nitrogen balance, 35
non-essential, 35t
placental concentrations, 106
urea cycle, 3738
Amylase
in pancreatic trauma, 381
Anaphylaxis, 214t, 215216
Anemia, 413
in transplant recipients, 343
Animal models
fetal origins of disease, 110
glucocorticoid exposure, 114
iron-deficient diets, 113
nutrient-restricted diets, 112113
of dietary manipulation, 112113
uterine artery ligation, 114
Anorexia nervosa, 205
diagnostic criteria, 205f
diet-induced thermogenesis, 206
energy requirements, 206
in childhood, 205
liver disease and, 303
physical presentation, 205
zinc deficiency, 207
495
496
INDEX
Anthropometric measurements
body mass index-for-age, 146, 415,
415t
head circumference, 144t, 145, 415
length/height, 144145, 414415
parental stature, 415
using knee height, 415
using length board, 414
using standiometer, 414415
using tibial length, 415
mid-arm circumference, 415416
triceps skinfold, 415416
percentiles, 421t
upper arm circumference
percentiles, 420t
weight, 414
method for obtaining, 144, 414
Appetite
control, 4
cultural aspect, 3
development during infancy, 4
hypothalamus role, 4
nutrient intake and, 34
phases
cephalic, 4
gastric, 4
intestinal, 4
programming of, 113
Arachidonic acid (ARA)
in infant formulas, 135136
Arginine
hyperammonemia and, 37
in fetal development, 106
Asthma, 333
B
Baby Friendly Hospital Initiative
(BFHI), 121
Ballard score, 409410, 410t
Bartters syndrome, 277
Bifidobacteria lactus, 137
Bifidobacterium bifidum, 120
Bile acid malabsorption, 316
Binge-eating disorder
diagnostic criteria, 206f
energy requirements, 206
Biotin
absorption, 6970
biochemistry, 69
deficiency state, 70
dietary reference intake, 58t
excretion, 6970
metabolism, 6970
physiology, 69
source, 69
supplementation, 70
Bloodstream infections, 317, 449,
470
Body mass index (BMI)
developmental delay and, 195
in growth assessment, 146
Body mass index-for-age, 415, 415t
Breast milk
adding infant formula to, 138
in chronic kidney disease, 273
cytomegalovirus in, 125
fortification, 122123, 122t
hang time, 124
hind milk, 122123
intestinal adaptation and, 313314
labeling, 124
maternal medications and, 125
preterm, 122, 122t
pumping, 122
safety, 124125
storage, 124, 124t
in tube feedings, 124
Breastfeeding
advocacy of, 120
appetite development
exposure to taste, 4
benefits of, 120121, 121t
dyad management, 121
effect on later obesity, 111
infant growth and, 123124
steps for successful, 121t
Bronchopulmonary dysplasia (BPD),
332333
Bulimia nervosa
diagnostic criteria, 205f
energy requirements, 206
physical presentation, 205206
Russells sign, 205
zinc deficiency, 207
Burn(s)
inhalation injury, 383
pediatric vs adult, 383
total body surface area, 383
Burn patient
energy requirements, 383384
hyperalbuminemia, 384
hyperglycemia in, 384
nutrition support, 384
pediatric, 383
postpyloric feeding, 384
protein requirements, 384
resting energy expenditure, 384
C
Calcium
absorption, 4849
body content, 45, 48
deficiency, 47, 49
deposits in kidney, 276
dietary reference intake, 46t
excess intake, adverse effects, 50
excretion, 49
homeostasis, 49
in chronic kidney disease, 267
medication interactions, 50
physiological functions, 48
requirements, 413
sources, 47t, 48
Campylobacter, 346t
Canadian Pediatric Society, 120
Cancer
Childrens Oncology Group
LongTerm Follow-Up
Guidelines for Survivors, 362
complementary and alternative
medicine, 354
disease outcome, 353
gastrointestinal supportive care
medications
acid-blockers, 368369
antiemetics, 369
antihistamines, 371
appetite stimulants, 371372
atypical antipsychotics, 371
cannabinoids, 371
glucocorticoids, 370
motility agents, 368
pheothiazines, 370
malnutrition in, 349350
mucositis medications, 372373
nutrition aspects
metabolic, 351
INDEX
physiological, 351
psychological, 351
nutrition assessment, 352353,
352t
nutrition intervention, 365f,
366t367t
overview, 349
survivors
gastrointestinal problems,
362363
heart health, 363364
osteoporosis, 363
treatment
complications, 354
tolerance, 353
chemotherapy, 350351
radiation therapy, 350, 350t
surgery, 350
Cancer cachexia, 351, 351t
Candida esophagitis, 315
Carbohydrate(s)
absorption, 20
as energy source, 17
classification, 17
complex, 17
digestion
lactose, 1719
starch, 19
malabsorption, 20
disorders, 20t
metabolism, 20
in sports nutrition, 179180, 180t
starches
oligosaccharides, 17
polysaccharides, 17
storage, 17
sugars
disaccharides, 17
monosaccharides, 17
Cardiovascular disease (CVD)
in chronic kidney disease, 268, 269
lipid management and, 268
in pediatric patients, 252254
risk factors, 162, 268
Celiac disease, 287289
at-risk groups, 288t
symptoms, 288t
497
498
INDEX
D
D-lactic acidosis,
in intestinal failure, 316
Dental caries, 414
Dentition, 45
by age of eruption, 5t
Depression, 205
Developmental delay
abnormal energy expenditure, 193
clinical features, 193
determining dietary energy needs,
197t
inappropriate dietary intake, 192
micronutrient deficiencies,
193194
nutrient loss, 193
nutrition assessment
diagnostic studies, 196
growth and anthropometric
measurements, 195
growth history, 194
meal observation, 196
medical history, 194
medications, 194
physical examination, 195
review of systems, 194
social history, 194195
nutrition problems
growth failure, 192
osteopenia, 194
overweight, 192
pathophysiology of, 192
prevalence of, 192
undernutrition, 192
nutrition support
behavioral modification, 197
enteral tube feeding, 197198
feeding intolerance, 199
formula administration, 198
nutrition requirements,
196197
positioning and oral feeding,
197
oral motor dysfunction, 192193
z scores, 192, 193
Developmental origins of health and
disease (DOHaD), 112, 115
animal models, 112114
fetal salvage hypothesis, 112
commercial meal/snack
replacements, 173174
food group guides/exchange
systems, 173
food timing/meals and snacks
combinations, 173
food-focused weight loss plans,
172
reduced macronutrient content
plans, 172173
WeightWatchers, 173174
Digestion
carbohydrates, 17
fats, 25
motor activity factors
caloric density, 13
consistency, 13
nutrient content,13
osmolarity, 13
physiology of, 12
proteins, 3233
small bowel motility, 12-13
Distal intestinal obstruction
syndrome, 324, 345
Docosahexaenoic acid (DHA),
135136
Drug-nutrient interactions, 466467
Duodenal hematoma, 381382
Duodenal injuries, 381382
Duodenal perforation, 381382
Dutch famine, 114115
Dyslipidemia, 268
Dysphagia, 8
E
Eating disorder(s)
anthropometric monitoring,
208209
classification of, 205
etiology, 206
laboratory tests for, 207t
meal-planning guidelines, 208
oral nutrition, 208
prevalence, 204
Eating disorder not otherwise
specified, 206f
Egg allergy, 233, 463
Electrolyte(s)
assessment, 90, 90t
INDEX
499
F
Failure to thrive, 145, 286
Familial hypercholesterolemia (FH),
163
Fat(s)
absorption, 26
adipose tissue, 24
classification, 23f
digestion, 25
in eating disorders, 207
fuel for preterm infant, 27
functions, 24f
in sports nutrition, 181
trans, 23
triglycerides, 22
Fatty acid(s)
essential, 2324
500
INDEX
long chain, 23
medium chain, 23
monounsaturated, 2223
oxidation, 2627
polyunsaturated, 23
Vitamin E intake, 23
saturated, 22, 23f
short chain, 23
synthesis, 26
Feeding difficulties
aspiration, 193
nutrient losses, 193
solid foods, 4
Feeding disorders
ethical considerations, 483
etiology of, 443
evaluation and treatment,
443444
nutrition intervention, 445t
posttransplant
liver, 342
treating, 443t
Feeding teams, 89, 442443, 444,
444f
Female Athlete Triad, 185t
Fenton fetal-infant growth chart,
422f
Fetal development
epigenetic changes, 107
glucose supply, 106
growth,
effect of calcium, 108
effect of iron, 108
effect of protein, 107108
effect of vitamin E, 108109
folic acid and, 108
vitamin B12, 108
hypothalamic-pituitary-adrenal
(HPA) axis, 107, 109110
insulin-responsive tissues, 106
intrauterine insults, 107
liver, 106
macronutrients in
amino acids, 106
glucose, 106
maternal nutrition and
calcium, 108
folic acid, 108
iron, 108
macronutrients, 107108
vitamin B12, 108
vitamin E, 108109
metabolic mediators
cortisol, 106
insulin, 106
insulin-like growth factors, 106
prenatal stress, impact of, 109110
substrate metabolism, 106
Fetal origins of adult disease,
110111
Fluid(s)
body distribution, 8788, 88t
deficit calculation, 89
imbalance, 90
maintenance, 412, 412t
regulation, 8889
requirements
in chronic kidney disease, 265
in hematopoietic stem cell
transplantation patient, 357
Holliday-Segar Formula, 89t
in intestinal failure/short bowel
syndrome, 314
in kidney transplant, 269,
in liver disease, 308
pediatric, 8990
Fluoride
drinking water levels, 414t
requirements, in adolescents, 414
supplementation, 414, 414t
topical, 414
Folate, 6566
absorption, 6566
biochemistry, 65
deficiency state, 66
dietary reference intake, 58t
excretion, 6566
metabolism, 6566
physiology, 65
source, 65
supplementation, 66
Folic acid, 108, 264
Food Allergen and Consumer
Protection Act (FALCPA), 221
Food allergens
alternative substitutes, 219t
cross-contamination, 221
hidden, 221, 222t
INDEX
G
Galactosemia
etiology, 241242
management
acute, 242
chronic, 242
diet, 242t
Gastroesophageal reflux (GER), 286,
332, 390
Gastrointestinal disease
disordered ingestion, 283-284
failure of absorption, 284
failure of digestion, 284
functional disorders, 295-296
increased needs, 284
nutrition assessment, 284-285,
285t
Gastrojejunostomy tube(s), 198, 404,
453
Gastroparesis, 295-296, 295t
Gastrostomy (G) tube
button device, 198t
in esophageal atresia, 393
insertion, 404, 453
PERT supplements, 330
safety issues, 450
Glucocorticoids
effect on fetus, 109110
as preterm delivery treatment, 109
Glucose
as fetal energy source, 106
in trauma response, 378379
Glycogen storage diseases (GSDs),
306
Graft versus host disease, 360361,
361t
Growth
average daily intrauterine weight
gain, 411t
breastfed infants, 123124
chronic kidney disease and,
259260
overview, 143144
of premature infant, 410
Growth assessment
body mass index, 146
failure to thrive (FTT), 145
head circumference, 144t, 145
H
Healthy People 2010, 120
Heart transplantation
biochemical tests, 339t
recipient
dietary modificiation, 344
nutrition requirements, 344
Hematopoietic stem cell
transplantation (HSCT), 354362
complications
bacterial overgrowth, 361
graft versus host disease,
360361, 361t
hemorrhagic cystitis, 362
infections, 361
mucositis, 360, 372373
pancreatic insufficiency, 361
sinusoidal obstruction
syndrome, 362
diet restrictions, 357358
medication side effects, 359t
nutrition support, 358-360
enteral nutrition, 358
oral, 357
procedure, 355
501
recipient
anthropometric assessment,
356
caloric requirements, 356, 356t
fluid requirements, 357
mineral requirements, 357
nutrition evaluation, 355356
protein requirements, 356, 356t
vitamin requirements, 357
Hemodialysis (HD)
in chronic kidney disease,
258259
in hypercalcemia, 97
for hypermagnesemia, 96
phosphorus clearance, 267
Holliday-Segar Formula, 49t
Honey, 139
Human milk, 120124
banked, 123
calcium content, 48
fortification, 122123, 122t
hind milk, 122123
iron content, 123
phosphorus content, 51
vitamin D content, 123
Human Milk Banking Association of
North America, 124
Hydration
guidance, 181t
in sports nutrition, 181182
Hydrochloric acid, 32
Hyperalbuminemia
in burn patients, 384
Hyperammonemia
treatment of, 37
Hypercalcemia, 50
Hypercalciuria
kidney stones and, 275
Hypergastinemia, 316
Hyperglycemia
in burn patient, 384
in critically ill patient, 227228
in refeeding syndrome, 209
in organ transplant recipient, 269,
341, 343, 344, 345
in trauma patient, 380
Hyperhomocysteinemia, 264
Hyperkalemia, 341
502
INDEX
I
Immunoglobulin E (IgE)
in food allergies, 214
mediated allergic diseases,
215216
reaction, 214
Immunosuppressive medications,
344, 345
kidney damage from, 269
side effects, 269
Immunosuppressive therapy
noncompliance with, 346
Inborn errors of metabolism (IEM),
232-242
defined, 232
newborn screening, 233
nutrition management, 233f
symptoms, 233t
Indirect calorimetry, 197t
Infant formula
as breast milk additive, 138
calcium salts, 48
concentrating, 250t
contamination, 124, 137
design of, 129130
forms
liquid concentrate, 137
powder, 137138
ready-to-feed, 137
functional ingredients
arachidonic acid (ARA),
135136
docosahexaenoic acid (DHA),
135136
nucleotides, 136
prebiotics, 136
probiotics, 136137
lactose in, 17
magnesium requirements, 52
manufacturers Web sites, 130t
medium-chain triglyceride, 252t
mixing
increasing caloric
concentration, 138, 138t
modular macronutrients, 138
standard dilution, 138
water, 138
phosphorus content, 51
Portagen, 124
regulation, 130
specialized
amino acid based, 134135,
136t, 220, 220t
carbohydrate free, 135, 136t
INDEX
longitudinal intestinal
lengthening and tailoring
(LILT), 315
Serial Transverse Enteroplasty
(STEP), 313, 315
Intestinal failure (IF)
causes, 312t
complications
bacterial overgrowth, 316
bile acid malabsorption, 316
D-lactic acidosis, 316
hypergastinemia, 316
nephrolithiasis, 316
rapid intestinal transit, 316
definition, 312
etiology of, 313
management
enteral nutrition, 313314
fluids, 314
general principles, 315316
medications, 314315
parenteral nutrition, 314
nutrition deficiencies, 316
prevalence of, 312
Intestine transplantation, 318, 339t,
340, 341
Intrauterine growth restriction
(IUGR), 106, 410
maternal dietary deficiencies, 107
placental transport insufficiency
and, 106
placental weight, 106
Intravenous fat emulsion (IVFE),
437, 439, 439t
Irritable bowel syndrome, 296
Iron
human milk fortifier, 123
deficiency, 113, 413
fetal growth and, 108
lactoferrin and, 123
requirements
adolescents, 413
premature infants, 413
Iron deficiency anemia, 413
J
Jejunostomy tube, 453
K
Kidney(s)
amino acid metabolism, 39
functions, 257
injury, 381
nephrogenesis, 257
phosphorus homeostatis and, 51
transplantation
biochemical tests
339t
calcium intake, 269
cardiovascular disease and, 269
complications, 343344
fluid intake, 269
recipient
nutrition requirements,
342343, 343t
trauma to, 381
Kidney disease. See also Acute
kidney injury
chronic
effect on growth, 259260
incidence, 256
nutrition assessment, 261268
primary diagnosis, 258t
stages of, 257t, 258
nephrotic syndrome, 274
stages, 257t, 258
Kidney stones. See Renal stones
L
Lactase, 17, 18, 18f
Lacto engineering, 122
Lactobacillus, 120
Lactobacillus GG, 137
Lactoferrin, 123
Lactose, 1719
Lanthanum carbonate, 267
Length board, 414
Leptin
appetite control, 4
food intake and adiposity, 113
liver disease and, 303
Leucine, 35
in fetal development, 106
Lipase
gastric, 25, 32
in pancreatic trauma, 381
503
504
INDEX
M
Magnesium
absorption, 52
body content, 45, 52
deficiency, 53
dietary reference intake, 46t
excess intake, adverse effects, 53
excretion, 52
in infant formulas, 52
metabolism, 5253
in chronic kidney disease, 268
physiological function of, 52
renal transport, 52
sources, 52
tissue distribution, 52
Malnutrition
in chronic kidney disease, 258
in liver disease, 303305
Waterlow criteria, 411, 411t
World Health Organization
classification, 145t
N
Nasogastric tube, 329, 452453
Nasojejunal tube, 453
National Cholesterol Education
Program, Adult Treatment Panel
III (NCEP-ATP III)
metabolic syndrome, 150t
recommendations, 155156,
157
risk factor profile, 153155
National Health and Nutrition
Examination Survey
food allergies, 215
National Kidney Foundation, 258
National Kidney Foundation Kidney
Disease Outcomes Quality
Initiative (NKF KDOQI), 257,
266
Necrotizing enterocolitis (NEC)
breast milk and, 121
in premature infants, 18
Neonatal hypermagnesemia, 53
Neonatal cholestasis, 305
INDEX
of premature infants
Ballard score, 409410, 410t
classification parameters,
409410
Lubchenco growth chart, 410
physical examination, 417, 417t
pretransplant, 339340
Waterlow criteria
stunting, 411, 411t
wasting, 411, 411t
Nutrition intervention
in cancer patients, 352, 353t
feeding disorders, 445t
Nutrition support
in burn patients, 384
closed head injuries, 380
for cystic fibrosis, 329330
developmental delay, 196199
during trauma, 379
for eating disorders, 208
pancreatic trauma, 381
traumatic brain injury, 380
O
Obesity
breastfeeding, 111
cardiovascular disease, 252254,
268
childhood, 169170
clinical approach, 170
ethical considerations, 483
postnatal weight gain and, 111
in trauma patients, 379, 384385
Oral allergy syndrome, 214t, 215
Oral motor dysfunction, 192193
Organ transplantation
biochemical tests pretransplant,
339t
complications, 343, 346
foodborne illnesses, 345
immunosuppressive therapy, 344,
346
indications for, 338339, 338t
physical examination, 339
quality of life, posttransplant, 346
recipient
anthropometric assessment,
338
dietary modification, 344
505
P
Pancreas
fetal, 106
injury to, 381, 385
Pancreatic enzyme replacement
therapy (PERT), 330
posttransplant, 344
Pancreatic insufficiency, 292293,
292t
Pancreatic pseudocyst, 381
Pancreatitis, 293294
Panhypopituitarism, 230
Pantothenic acid
absorption, 69
biochemistry, 69
deficiency state, 69
dietary reference intake, 58t
excretion, 69
metabolism, 69
physiology, 69
source, 69
supplementation, 69
Parenteral nutrition
access, 437
additives
carnitine, 441442
cysteine, 442
cysteine hydrochloride salt, 36
heparin, 441
aluminum in, 442
amino acid solutions, 37
506
INDEX
limits, 471t
in liver disease, 308, 317
macronutrients
amino acids, 437439, 438t
carbohydrates, 437
magnesium overdose, 53
medications
compatability of, 467
drug-nutrient interactions,
466467, 467t
nursing care, 451, 452t
peripheral, 433, 449
in premature infants, 410,
436437
safety issues
bloodstream infections, 317,
450, 470
prevention of, 451t
Munchausen Syndrome by Proxy,
450
site care, 450
chlorhexidine gluconate, 450
Guidelines for the Prevention
of Intravascular
Cather Infections
recommendations, 450
total nutrient admixtures
advantages and disadvantages,
466t
factors affecting stability, 465t
in trauma, 379
trace elements
chromium, 441, 441t
copper, 440, 441t
manganese, 441, 441t
selenium, 441, 441t
zinc, 440, 441t
in transplantation
intestine, 340341
liver, 341, 342t
vitamins, 441t
when to use, 433434
Pelvic injury, 382
Pepsinogen, 3233
Peptide, 32, 32t
Peripheral blood stem cell
transplantation (PBSCT).
See Hematopoietic stem cell
transplantation (HSCT)
INDEX
R
Refeeding syndrome
definition, 209
hyperglycemia, 209
hypokalemia, 209, 210
hypomagnesemia, 209, 210
hypophosphatemia, 209
incidence of, 209
preventing, 210211, 210t
treatment, 210211, 210t
Renal failure
creatinine clearance, 257
nutrition management, 277t
oxalosis, 278
Renal stones
2,8-dihydroxyadenine calculi, 276
calcium-based, 275276
cystine stones, 274
cystinuria, 276
infection stones, 276
nutrition management, 275t
in pediatric patients, 275
struvite calculi, 274, 276
uric acid, 274, 276
xanthine stones, 276
Renal tubular acidosis, 276277
Renal tubular disorders
Bartters syndrome, 277
nephrogenic diabetes insipidus,
277278
renal tubular acidosis, 276277
Resting energy expenditure (REE)
in burn patient, 384
in developmentally delayed child,
193
in liver disease, 304
in trauma patients, 379
World Health Organization
equation, 412, 412t
Rett syndrome, 192, 193
Riboflavin, 6263
absorption, 6263
biochemistry, 62
deficiency state, 63
dietary reference intake, 58t
excretion, 6263
metabolism, 6263
physiology, 62
source, 62
supplementation, 63
Rickets, 46, 413
S
Salmonella, 346t
Seatbelt injury, 381, 382
Serial transverse enteroplasty
(STEP), 313, 315
Serum albumin, 262
Sevelamer carbonate, 267
507
508
INDEX
Swallowing
esophageal phase, 5
oral phase, 5
pharyngeal phase, 5
Syndrome of inappropriate
antidiuretic hormone (SIADH),
89, 380
T
Tacrolimus, 53, 269, 341, 345
magnesium deficiency and, 53
Thiamin, 6162
absorption, 61
biochemistry, 61
deficiency state, 6162
dietary reference intake, 58t
excretion, 61
metabolism, 61
physiology, 61
source, 61
supplementation, 62
toxicity, 78
Thoracic injuries, 382
Toddler(s)
feeding problems, 78
feeding skills, 7
formula, 139, 140t
Total body water (TBW), 87, 88t
Total nutrient admixtures (TNAs),
464465, 465t, 466t
Trans-fatty acids, 23
Transsulfuration pathway, 36
Trauma
adrenal hemorrhage, 381
age-related injuries, 379
anatomical classification, 379
blunt abdominal, 380381
hollow visceral injuries,
381382
solid visceral injuries, 380381
brain injury, 380
child abuse, 381
childhood, 379
clinical examples, 384385
closed head injuries, 380
duodenal injuries, 381382
extremity injury, 382
facial, 382
hyperglycemia and, 380
U
U.S. Department of Agriculture,
Food Safety and Inspection
Service, 345
Umbilical cord blood transplantation
(UCBT). See Hematopoietic stem
cell transplantation (HSCT)
United Nations International
Childrens Fund (UNICEF), 121
Baby Friendly Hospital Initiative,
121
Urea cycle, 38f, 239f
Uric acid stones, 274, 276
Urinary extravasation, 381
V
Vascular access device (VAD), 449
blood sample techniques, 450t
Very long chain acyl-CoA
dehydrogenase deficiency
(VLCADD)
etiology, 240
INDEX
W
Wasting, 411, 411t
Wilsons disease, 306
509
Winkler, Marion, 3
World Health Organization
breastfeeding, 120, 129
growth charts, 143144
malnutrition classification, 145t
metabolic syndrome, 150t
resting energy expenditure
equation, 412, 412t
X
Xanthine stones, 276
Z
z scores
in developmental delay, 192, 193
percentile comparisons, 146t
Zinc
in chronic kidney disease, 265
in cystic fibrosis-specific
multivitamins, 327t, 328
in eating disorders, 207
in liver disease, 303, 308