Revision
Revision
Revision
1.4
Table 1.5
FIG. 1.3 New proposed biologic pathways that mediate effects of selected stressful or
adversarial poverty-associated risks to neurocognitive outcomes in children. Complex
interactions among key poverty-related risk factors, focusing on primary biologic
pathways related to malnutrition, infection and inflammation, and neuroendocrine
responses to stress. (From Jensen SKG, Berens AE, Nelson CA: Effects of poverty on
interacting biological systems underlying child development, Lancet 1:225–238, 2017,
Fig 1, p 228.)
Table 1.6
H e a t h C o n d i t i o n s i n C h i l d re n Wi t h S p e c i a l H e a l t h
C a r e N e e d s ( C S H C N )*
Attention-deficit/hyperactivity disorder
Depression
Anxiety
problems
Behavioral or conduct problems
Autism,
pervasive
developmental disorder,
autism spectrum disorder
Developmental delay
Intellectual
disability
Communication
disorder
Asthma
Diabetes
Epilepsy
or seizure disorder
Migraines
or frequent
headaches
Headinjury,traumatic
brain injury
Heartproblems,including
congenital
heart disease
Bloodproblems,including
anemia
or sickle cell
disease
Cystic fibrosis
Cerebral palsy
Muscular
dystrophy
Down syndrome
Arthritis
or joint problems
Allergies
Functional
Difficulties
in Children
With
Special H
e a l t h C a r e N e e d s ( C S H C N )*
Experiencing Difficulty With …
Breathing, or respiratory problem
Swallowing, digesting food, or metabolism
Blood circulation
Repeated or chronic
physical pain, including
headaches
Seeing, even
when
wearing glasses or contact
lenses
Hearing, even when using a hearing aid or other
devise
Taking care of self, such as eating, dressing, or bathing
Coordination or moving around
Using his/her hands
Learning, understanding, or paying attention
Speaking, communicating,
or being understood
Feeling anxious or depressed
Behavior problems
as acting out, fighting, bullying, or arguing
such
Making and keeping friends
Table 2.1
Child Health Disparities
FAMILY
HEALTH INDICATOR RACE/ETHNICITY RESIDENCE
INCOME
Child health status fair or poor Black & Hispanic > White & Asian Poor >
Not Poor
Children with special health care needs Black > White > Hispanic Poor >
(CSHCN) Not Poor
One or more chronic health conditions Black > White > Hispanic > Asian Poor >
Not Poor
Asthma Mainland Puerto Rican > Black > White & Poor > Urban > Rural
Mexican American Not Poor
Obesity Hispanic & Black > White and Asian Poor > Rural > Urban
Not Poor
Infant mortality Black > Hispanic > White Poor >
Not Poor
Low birthweight (<2,500 g.) Black > White, Hispanic, American Poor >
Indian/Native Alaskan, Asian/Pacific Not Poor
Islander
Mainland Puerto Rican > Mexican
American
Preterm birth (<37 wk) Black > American Indian/Native Poor >
Alaskan, Hispanic, White, Not Poor
Asian/Pacific Islander
Mainland Puerto Rican > Mexican
American
Seizure disorder, epilepsy Black > White, Hispanic Poor >
Not Poor
Bone, joint, or muscle problem White > Black, Hispanic Poor >
Not Poor
Ever breastfed White, Hispanic, Asian > Black Not Poor Urban > Rural
> Poor
No physical activity in the past week Hispanic > Black, Asian > White Poor >
Poor > Not Poor Not Poor
Hearing problem Poor >
Not Poor
Vision problem Poor >
Not Poor
Oral health problems (including caries Hispanic > Black > White, Asian Poor > Rural > Urban
and untreated caries) Not Poor
Attention-deficit/hyperactivity disorder White, Black > Hispanic Poor > Rural > Urban
(ADHD) Not Poor
Have ADHD but not taking medication Hispanic, Black > White
Anxiety problems White > Black, Hispanic Poor >
Not Poor
Depression Poor > Rural > Urban
Not Poor
Behavior or conduct problem (ODD, Black > White, Hispanic Poor >
conduct disorder) Not Poor
Autism Spectrum Disorder White > Black > Hispanic Poor >
Not Poor
Learning disability Black > White, Hispanic Poor > Rural > Urban
Not Poor
Developmental delay Black > White > Hispanic, Asian Poor >
Not Poor
Risk of developmental delay, by Hispanic > Black & White Poor >
parental concern Not Poor
Speech or language problems Poor >
Not Poor
Adolescent suicide attempts (consider, Girls: Hispanic > Black & White
attempt, needed medical attention for an Boys:Hispanic & Black > White
attempt)
Adolescent suicide rate Girls: American Indian > White,
Asian/Pacific Islander, Hispanic, Black
Boys: American Indian & White >
Hispanic, Black, Asian/Pacific Islander
Child maltreatment (reported) Black, American Indian/Alaskan Native, Poor >
Multiracial > White, Hispanic, Asian, Not Poor
Pacific islander
Table 2.2
Child Healthcare Disparities
FAMILY
HEALTHCARE INDICATOR RACE/ETHNICITY RESIDENCE
INCOME
Did not receive any type of medical care in past 12 mo Hispanic, Black, Asian Poor > Not Rural > Urban
> White Poor
No well-child checkup or preventive visit in past 12 mo Hispanic > White & Poor > Not Rural > Urban
Black Poor
Delay in medical care Hispanic > Black > Poor > Not
White Poor
Unmet need in healthcare due to cost Black > Hispanic > Poor > Not
White > Asian Poor
No coordinated, comprehensive, or ongoing care in a Hispanic > Black & Poor > Not Rural > Urban
medical home Asian > White Poor
Problem accessing specialist care when needed Hispanic & Black > Poor > Not
White Poor
No preventative dental care visit in past 12 mo Hispanic & Asian > Poor > Not Rural > Urban
Black > White Poor
No vision screening in past 2 yr Hispanic & Asian > Poor > Not
Black & White Poor
Did not receive needed mental health treatment or Black & Hispanic > Poor > Not
counseling in past 12 mo White Poor
Not receiving a physician recommendation for HPV Black & Hispanic >
vaccination among 13-17-yr- old girls White
Immunization rates: adolescent HPV vaccine Girls: White >
Black & Hispanic
Boys: Black &
Hispanic > White
HPV, Human papillomavirus.
Table 2.3
New Social and Health Inequities in the United States
BLACK NATIVE
WHITE HISPANIC NON- AMERICAN
TOTAL NON- ASIAN* OR HISPANIC OR
HISPANIC LATINO † ALASKA
NATIVE
Wealth: median household assets $68,828 $110,500 $89,339 $7,683 $6,314 NR
(2011)
Poverty: proportion living below 14.8%; 10.1%; 12.0%; 23.6%; 26.2%; 28.3%;
poverty level, all ages (2014); 21.0% 12.0% 12.0% 32.0% 38.0% 35.0%
children <18 yr (2014)
Unemployment rate (2014) 6.2% 5.3% 5.0% 7.4% 11.3% 11.3%
Incarceration: male inmates per 982 610 185 836 3,611 1,573
100,000 (2008)
Proportion with no health insurance, 13.3% 13.3% 10.8% 25.5% 13.7% 28.3%
age <65 yr (2014)
Infant mortality per 1000 live births 6.0 5.1 4.1 5.0 10.8 7.6
(2013)
Self-assessed health status (age- 8.9% 8.3% 7.3% 12.2% 13.6% 14.1%
adjusted): proportion with fair or
poor health (2014)
Potential life lost: person-years per 6621.1 6659.4 2954.4 4676.8 9490.6 6954.0
100,000 before age 75 yr (2014)
Proportion reporting serious 3.4% 3.4% 3.5% 1.9% 4.5% 5.4%
psychological distress ‡ in past 30
days, age ≥18 yr, age-adjusted
(2013–14)
Life expectancy at birth (2014), yr 78.8 79.0 NR 81.8 75.6 NR
Diabetes-related mortality: age- 20.9 19.3 15.0 25.1 37.3 31.3
adjusted mortality per 100,000
(2014)
Mortality related to heart disease: 167.0 165.9 86.1 116.0 206.3 119.1
age-adjusted mortality per 100,000
(2014)
HEALTH AND MULTISECTOR ACTIONS
• Ensuring food security for the family (or mother and child)
• Maternal education
• Safe drinking water and sanitation
• Handwashing with soap
• Reduced household air pollution
• Health education in schools
AGE-SPECIFIC ACTIONS
Prevention Treatment
ADOLESCENCE AND PRE-PREGNANCY
• Family planning
• Preconception care
PREGNANCY
• Appropriate care for normal and high-risk • Antenatal steroids for premature births
pregnancies (maternal tetanus vaccination) • Intermittent preventive treatment for malaria
CHILDBIRTH
• Maternal intrapartum care and monitoring
• Skilled delivery • Newborn resuscitation (e.g., Healthy Babies Breathe)
• Thermal care for all newborns • Premature: surfactant administration, continuous positive
• Clean cord and skin care airway pressure (CPAP), treatment of jaundice
• Early initiation and exclusive breastfeeding • Feeding support for small/preterm infants
within 1st hr
PRENATAL PERIOD
• Appropriate postnatal visits • Extra care for small and sick babies (kangaroo mother
care, treatment of infection, support for feeding,
management of respiratory complications)
• Antibiotics for newborns at risk and for treatment of
bacterial infections (PROM, sepsis, meningitis,
pneumonia)
INFANCY AND CHILDHOOD
• Exclusive breastfeeding for 6 mo and continued
breastfeeding up to at least 2 yr with appropriate
complementary feeding from 6 mo
• Monitoring and care for child growth and • Case management of severe acute malnutrition
development
• Routine immunization childhood diseases
• Micronutrient supplementation, including vitamin A
from 6 mo
• Prevention of childhood diseases • Management of childhood diseases
• Malaria (insecticide-treated bed nets) • Malaria (antimalarials)
• Pneumonia • Pneumonia (case management, antibiotics)
• Diarrhea (rotavirus immunization) • Diarrhea (ORS, zinc supplement, continued feeding)
• Meningitis (meningococcal/Hib/pneumococcal • Meningitis (case management, antibiotics)
vaccination) • Measles (vitamin A suppl)
• Measles (vaccination) • Comprehensive care of children exposed to or infected
• Prevention of mother-to-child HIV transmission with HIV (HAART)
HAART, Highly active antiretroviral therapy; ORS, oral rehydration solution (salts).
Adapted from Were W, Daelmans B, Bhutta ZA, et al. Children's health priorities and
interventions, BMJ 351:h4300, 2015.
FIG. 3.8 Coverage of interventions across the continuum of care based on the most
recent data since 2012 in Countdown countries. Bars show median national coverage of
interventions; dots show country-specific data. (From Countdown to 2030
Collaboration: Tracking progress towards universal coverage for reproductive,
maternal, newborn, and child health, Lancet 391:1538-1548, 2018. Fig 1.)
Vaccine-Preventable Diseases
In 2002 an estimated 1.5 million under-5 deaths were caused by vaccine-
preventable diseases. Top contributors were pneumococcus and rotavirus,
followed by Haemophilus influenzae B (Hib), measles, pertussis, and tetanus.
The World Health Organization (WHO) Expanded Program on Immunization
(EPI) has resulted in a dramatic reduction in deaths, illness, and disability from
many of these diseases, as well as the near-elimination of poliomyelitis.
Recommendations for routine immunizations have continued to grow with the
development of new vaccines that have demonstrated significant lifesaving
potential in industrialized countries (Table 3.2 ).
Table 3.2
Routine Immunizations Recommended by the World Health
Organization (2017)
INTERVAL BETWEEN
DOSES IN DOSES
VACCINE AGE AT 1st BOOSTER
PRIMARY ADOLESCENT CONSIDERATIO
(ANTIGEN) DOSE 3rd DOSE
SERIES 2nd to
1st to 2nd to
3rd
4th
BCG (bacille Birth 1
Calmette-Guérin)
CRS, Congenital rubella syndrome; IPV, inactivated polio vaccine; TB, tuberculosis.
Adapted from WHO Routine Guidelines for Immunization.
http://www.who.int/immunization/policy/Immunization_routine_table2.pdf?ua=1 .
promotion activities (Fig. 3.9 ).
FIG. 3.9 Health services delivery systems.
FIG. 3.10 Integrated Management of Childhood Illness (IMCI). (Adapted from WHO
2014. Revised WHO classification and treatment of pneumonia in children at health
facilities: evidence summaries; and WHO 2008. Integrated Management of Childhood
FIG. 3.11 Country categorization based on adolescent burden of disease.
Categorization into 3 groups according to adolescent burden of disease and reflecting
passage through epidemiologic transition. DALYs, Disability-adjusted life-years; HIV,
human immunodeficiency virus; NCD, noncommunicable diseases. (From Patton GC,
Sawyer SM, Santelli JS, et al: Our future: a Lancet commission on adolescent health
and wellbeing, Lancet 387:2423–2478, 2016, Fig 7.)
Valuable information can be obtained from PDSA cycles that are successful, and
those that are not, to help plan the next iteration of the PDSA cycle. The PDSA
cycle specifically requires that improvements be data driven. Many clinicians
attempt to make changes for improvement in their practice based on clinical
intuition rather than on interpretation of empirical data.
FIG. 4.1 A, The plan-do-study-act (PDSA) cycle. B, Use of PDSA cycles: a
ramp. (From Langley GJ: The improvement guide: a practical guide to
enhancing organizational performance, San Francisco, 1996, Jossey-Bass.
© 1996 by Gerald J. Langley, Kevin M. Nolan, Thomas W. Nolan, L.
Norman, and Lloyd P. Provost.)
FIG. 4.2 Improving quality by reducing variation.
FIG. 4.3 A and B, Lean methodology—waste reduction.
FIG. 4.4 Management sciences—discrete event stimulation. PICU, Pediatric intensive
care unit.
FIG. 4.5 Management sciences—cognitive mapping.
FIG. 4.6 Key driver diagram: theory of how to achieve an aim.
FIG. 4.7 Failure modes and effects analysis (FMEA).
FIG. 4.8 Pareto chart.
Table 4.1
FIG. 4.9 Development of a rigorous quality measure.
Table 4.2
Examples of National Pediatric Quality Measures
NQF NQF-ENDORSED
NQF-ENDORSED
PEDIATRIC INPATIENT NQF-ENDORSED INPATIENT
OUTPATIENT PEDIATRIC
QUALITY MEASURES PEDIATRIC CARE MEASURES
CARE MEASURES
INDICATORS AMONG PICUs
Neonatal PICU CAC-1 relievers for inpatient Appropriate testing for
bloodstream standardized asthma children with pharyngitis
infection rate mortality ratio CAC-2 systemic corticosteroids CAHPS clinician/group
Transfusion PICU severity- for inpatient asthma surveys (adult primary
reaction adjusted length Admit decision time to ED care, pediatric care, and
Gastroenteritis of stay departure time for admitted specialist care surveys)
admission rate PICU patients Child and adolescent
unplanned Follow-up after hospitalization for major depressive
readmission mental illness (FUH) disorder: diagnostic
rate NHSN Catheter-Associated evaluation
Urinary Tract Infection (CAUTI) Child and adolescent
outcome measure major depressive
NHSN Central Line-Associated disorder: suicide risk
Bloodstream Infection (CLABSI) assessment
outcome measure Follow-up after
Percent of residents or patients hospitalization for mental
assessed and appropriately given illness (FUH)
the pneumococcal vaccine (short Median time from ED
stay) arrival to ED departure
Restraint prevalence (vest and for discharged ED
limb) patients
Validated family-centered survey Pediatric symptom
questionnaire for parents' and checklist (PSC)
patients' experiences during
inpatient pediatric hospital stay
Nursing hours per patient day
Preventive care and screening:
screening for clinical depression
and follow-up plan
Skill mix (RN, LVN/LPN, UAP,
and contract)
FIG. 4.10 Success ingredients for large-scale quality improvement.
Table 4.3
Institute for QI organization for adult and pediatric care QI collaboratives, QI educational workshops
Healthcare and materials
Improvement (IHI)
National Initiative QI organization for pediatric care QI training, improvement networks
for Child Health
Quality (NICHQ)
The Joint Hospital accreditation organization Unannounced surveys to evaluate quality of
Commission care in hospitals
National QI organization Healthcare Effectiveness Data and
Committee for Information Set (HEDIS) and quality
Quality Assurance measures for improvement
(NCQA)
National Quality Multidisciplinary group including healthcare Endorsing national quality measures,
Forum (NQF) providers, purchases, consumers, and convening expert groups, and setting
accrediting bodies national priorities
improvements in process measures and outcomes measure in a hypothetical
CLA-BSI project. In this case, after improvement interventions targeted 2
process measures known to be important in CLA-BSI risk—the line insertion
and the line maintenance bundles—the QI team saw improvement in both
measures and coincident reduction in CLA-BSIs.
Table 5.1
Common Healthcare-Associated Conditions (HACs)
Targeted in Quality Improvement Efforts with Interventions
COST
POTENTIALLY EFFECTIVE
HAC DEFINITION PER
INTERVENTIONS
EVENT
Central line– Laboratory-confirmed bloodstream $55,646 Line insurance bundle (e.g.,
associated infection with central line in place at time handwashing, chlorhexidine scrub),
bloodstream of or 48 hr before onset of event (details at maintenance bundle (catheter care,
infections https://www.cdc.gov/nhsn ) change dressing, discuss daily if
catheter is needed)
Catheter- Urinary tract infection where an indwelling $7,200 Protocols for reviewing and removing
associated urinary urinary catheter was in place >2 days on catheters daily, clear indications for
tract infections day of event (details at inserting catheters, physician
https://www.cdc.gov/nhsn ) champions, audit and feedback of data
Adverse drug Harm associated with any dose of a drug $3,659 Pharmacist review of medication
events (details at http://www.nccmerp.org/types- order, computerized physician order
medication-errors ) entry, co-ordering of laxatives in
patients on opiates
Peripheral IV Moderate or serious harm (e.g., diminished — Hourly reviews of IV status,
infiltrates pulses, >30% swelling) associated with a limitations on use of desiccants
peripheral IV infiltrate (details at through peripheral IVs, remove IVs
http://www.solutionsforpatientsafety.org ) when no longer needed
Pressure injuries Localized damage to skin and/or underlying — Screening of high-risk patients (e.g.,
soft tissue usually over a bony prominence Braden Q Scale), regular turning of
or related to a device (details at low-mobility patients, regular
http://www.solutionsforpatientsafety.org ) inspection and skin care; specialized
device padding
Surgical site Infection of incision or deep tissue space — Surgical checklist, antimicrobial
infections after operative procedure (details at prophylaxis within 60 min before
https://www.cdc.gov/nhsn ) incision, preoperative baths,
postoperative antibiotic redosing
Venous Blood clot in deep vein, stratified as central $27,686 Screening for high-risk patients,
thromboembolism line–associated vs not (details at removal of central line catheters when
https://www.cdc.gov/nhsn ) no longer needed, targeted prophylaxis
IV, Intravenous line.
FIG. 5.2 Quality improvement interventions targeted process improvement
in the A, line insertion bundle, where performance improved from 46% to
95%), and B, line maintenance bundle, where performance improved from
13% to 66%. Coincident with the improved bundle performance, the rate of
processes, and easy access to lifts for larger children with limited mobility.
Violence and patient interaction injuries, often from children with psychiatric
disease or developmental disabilities, are a growing source of harm to clinicians.
Table 5.2
Table 5.3
Table 5.4
Table 5.5
Solutions to Avoid Diagnostic Errors
Cystic fibrosis
Severe immunodeficiency
High-risk solid-organ transplant candidates and/or recipients (e.g., lung,
multivisceral)
Chronic or severe respiratory failure
Muscular dystrophy
Complex multiple congenital malformation syndromes
Primary pulmonary hypertension
Severe chromosomal disorders (aneuploidy, deletions, duplications)
Adapted from The Together for Short Lives [formerly the Association for
Children's Palliative Care (ACT)] Life-limiting/Life-threatening Condition
Categories.
http://www.togetherforshortlives.org.uk/professionals/childrens_palliative_care_essentials/app
.
Although often mistakenly understood as equivalent to end-of-life care , the
scope and potential benefits of palliative care are applicable throughout the
illness trajectory . Palliative care emphasizes optimization of quality of life,
communication, and symptom control, goals that may be congruent with
maximal treatment aimed at sustaining or prolonging life.
The mandate of the pediatrician and other pediatric clinicians to attend to
children's physical, mental, and emotional health and development includes the
provision of palliative care for those who live with a significant possibility of
death before adulthood (Fig. 7.1 ). Such comprehensive physical, psychological,
social, and spiritual care requires an interdisciplinary approach.
FIG. 7.1 Typical illness trajectories for children with life-threatening illness. (From Field
M, Behrman R, editors: When children die: improving palliative and end-of-life care for
children and their families, Washington, DC, 2003, National Academies Press, p 74.)
Table 7.2
Developmental Questions, Thoughts, and Concepts of
Dying, With Responsive Strategies
TYPICAL
QUESTIONS
AND THOUGHTS THAT DEVELOPMENTAL STRATEGIES AND
STATEMENTS GUIDE BEHAVIOR UNDERSTANDING OF DEATH RESPONSES
ABOUT
DYING
MONTHS TO 3 YR OF AGE
“Mommy, Child has limited Child may have “sense” that Optimize comfort, and
don't cry.” understanding of events, something is wrong. Death is often consistency; familiar
“Daddy, future and past, and of the viewed as continuous with life persons, objects,
will you difference between living (analogous to being awake and being routines. Use soothing
still tickle and nonliving. asleep). songs, words, and
me when touch.
I'm dead?” “I will always love
you.”
“I will always take
care of you.”
“I will tickle you
forever.”
AGE 3-5 YR
“I did Concepts are simple and Child may see death as Assure child that
something reversible. Variations temporary and reversible and not illness not her fault.
bad and so I between reality and universal. Provide consistent
will die.” fantasy. Child may feel responsible for caregivers.
“Can I eat illness. Promote honest simple
anything I Death may be perceived as an language.
want in external force that can get you. Use books to explain
heaven?” the life cycle and
promote questions and
answers.
“You did not do
anything to cause
this.”
“You are so special to
us, and we will always
love you.”
“We know (God,
Jesus, Grandma,
Grandpa) are waiting
to see you.”
AGE 5-10 YR
“How will I Child begins to Child begins to understand death as Be honest and provide
die?” demonstrate organized, real and permanent. Death means that specific details if they
“Will it logical thought. Thinking your heart stops, your blood does not are requested. Help
hurt?” becomes less esoteric. circulate, and you do not breathe. It and support the child's
“Is dying Child begins to problem- may be viewed as a violent event. need for control.
scary?” solve concretely, reason Child may not accept death could Permit and encourage
logically, and organize happen to himself or to anyone he the child's
thoughts coherently. knows but starts to realize that people participation in
However, child has limited that he knows will die. decision making.
abstract reasoning. “We will work
together to help you
feel comfortable. It is
very important that
you let us know how
you are feeling and
what you need. We
will always be with
you, so you do not
need to be afraid.”
AGE 10-18 YR
“I'm afraid Abstract thoughts and Understand death as irreversible, Reinforce
if I die my logic possible. inevitable and universal. child/adolescent's self-
mom will Body image is Child needs reassurance of esteem, sense of
just break important. continued care and love. worth, and self-
down.” Child needs peer Search for meaning and purpose respect. Allow need
“I'm too relationships for of life. for privacy,
young to support and for independence, access
die. I want validation. to friends and peers.
to get Child expresses Tolerate expression of
married and altruistic values, such strong emotions and
have as staying alive for permit participation in
children.” family (parents, decision-making.
“Why is siblings) and donating “I can't imagine how
God letting organs/tissue. you must be feeling.
this Disbelief that she is Despite it all, you are
happen?” dying. doing an incredible
job. I wonder how I
can help?”
“What's most
important to you
now?”
“What are your hopes
… your worries?”
“You have taught me
so much; I will always
remember you.”
Adapted from Hurwitz C, Duncan J, Wolfe J: Caring for the child with cancer at the close of life,
JAMA 292:2141–2149, 2003.
overwhelmed to provide this at crucial times.
The Staff
Inadequate support for the staff providing palliative care can result in depression,
emotional withdrawal, and other symptoms. Offering educational opportunities
and emotional support for staff at various stages of caring for a child with life-
threatening illness can be helpful in bettering patient/family care and preventing
staff from experiencing compassion fatigue, burnout, and long-term
repercussions, including leaving the field.
Table 7.3
Resuscitation Status
Many parents do not understand the legal mandate requiring attempted
resuscitation for cardiorespiratory arrest unless a written DNR order is in place.
In broaching this topic, rather than asking parents if they want to forgo
cardiopulmonary resuscitation (CPR) for their child (and placing the full burden
of decision-making on them), it is preferable to discuss whether or not
resuscitative interventions are likely to benefit the child. It is important to make
recommendations based on overall goals of care and medical knowledge of
potential benefit and/or harm of these interventions. Once the goals of therapy
are agreed on, the physician is required to write a formal order. Out-of-hospital
DNR verification forms are available in many states, which if completed on
behalf of the child, affirm that rather than initiating resuscitative efforts,
emergency response teams are obligated to provide appropriate symptom
management with comfort and relief of suffering with appropriate interventions
when called to the scene.
Almost half of all states have implemented the physician orders for life
treatment (POLST) system . A POLST order is completed for children with
life-threatening illness, translating the expressed parent's and/or child's wishes
for interventions to do or not do into actionable orders (www.polst.org ). It is
usually helpful to frame discussions about POLST as ways for parents to
maintain some control, by communicating their goals and care preferences, so
that they may be honored, irrespective of the setting. It may also be beneficial to
write a letter that delineates decisions regarding resuscitation interventions and
supportive care measures to be undertaken for the child, particularly if POLST
are not available. The letter should be as detailed as possible, including
recommendations for comfort medications and contact information for
caregivers best known to the patient. Such a letter, given to the parents, with
copies to involved caregivers and institutions, can be a useful communication
aid, especially in times of crisis. In any case, if a child may die in the home
setting, and the parents opt to use on out-of-hospital DNR verification form or
POLST, plans to pronounce the child and provide support for the family must be
in place. If the child has been referred to hospice, the hospice usually fulfills
those responsibilities.
Conflicts in decision-making can occur within families, within healthcare
teams, between the child and family, and between the family and professional
caregivers (see Chapter 6 ). For children who are developmentally unable to
provide guidance in decision-making (neonates, very young children, or children
with cognitive impairment), parents and healthcare professionals may come to
different conclusions as to what is in the child's best interests. Decision-making
around the care of adolescents presents specific challenges, given the shifting
boundary that separates childhood from adulthood. In some families and
cultures, truth-telling and autonomy are secondary to maintaining the integrity of
the family (see Chapter 11 ). Although frequently encountered, differences in
opinion are often manageable for all involved when lines of communication are
kept open, team and family meetings are held, and the goals of care are clear.
Symptom Management
Intensive symptom management is another cornerstone of pediatric palliative
care. Alleviation of symptoms reduces suffering of the child and family and
allows them to focus on other concerns and participate in meaningful
experiences. Despite increasing attention to symptoms, and pharmacologic and
technical advances in medicine, children often suffer from multiple symptoms.
Table 7.4 provides key elements and general approaches to managing symptoms.
Table 7.4
Key Elements of Effective Symptom
Management
Setting the Stage
• Establish and periodically revisit goals of care and ensure that goals are
communicated to entire care team.
• Plan for symptoms (including unanticipated ones) before they occur.
Assessment
Treatment
Ongoing Care
Table 7.6
Pharmacologic Approach to Symptoms Commonly
Experienced by Children With Life-Threatening Illness
† Although the usual opioid starting dose is presented, dose may be titrated as needed. There is
no ceiling/maximum dose for opioids.
‡ Breakthrough dose is 10% of 24 hr dose. See Chapter 76 for information regarding titration of
opioids.
§ Side effects from opioids include constipation, respiratory depression, pruritus, nausea, urinary
Table 7.7
Nonpharmacologic Approach to Symptoms Commonly
Experienced by Children With Life-Threatening Illness
Dyspnea or air Suction oral secretions if present; positioning, comfortable loose clothing, fan to provide
hunger cool, blowing air.
Limit volume of intravenous fluids; consider diuretics if fluid overload/pulmonary edema
present.
Behavioral strategies, including breathing exercises, guided imagery, relaxation, music,
distraction.
Fatigue Sleep hygiene (establish a routine, promote habits for restorative sleep).
Regular, gentle exercise; prioritize or modify activities.
Address potentially contributing factors (e.g., anemia, depression, side effects of
medications).
Aromatherapy* : peppermint, rosemary, basil.
Nausea/vomiting Consider dietary modifications (bland, soft, adjust timing/volume of foods or feeds).
Aromatherapy* : ginger, peppermint, lavender, acupuncture/acupressure.
Constipation Increase fiber in diet, encourage fluids, ambulation (if possible).
Oral Oral hygiene and appropriate liquid, solid and oral medication formulation (texture, taste,
lesions/dysphagia fluidity). Treat infections, complications (mucositis, pharyngitis, dental abscess, esophagitis).
Oropharyngeal motility study and speech (feeding team) consultation.
Anorexia/cachexia Manage treatable lesions causing oral pain, dysphagia, or anorexia. Support caloric intake
during phase of illness when anorexia is reversible. Acknowledge that anorexia/cachexia
is intrinsic to the dying process and may not be reversible.
Prevent/treat coexisting constipation.
Pruritus Moisturize skin.
Trim child's nails to prevent excoriation.
Try specialized anti-itch lotions.
Apply cold packs.
Counterstimulation, distraction, relaxation.
Diarrhea Evaluate/treat if obstipation.
Assess and treat infection.
Dietary modification.
Depression Psychotherapy, behavioral techniques, setting attainable daily goals.
Aromatherapy* : bergamot, lavender.
Anxiety Psychotherapy (individual and family), behavioral techniques.
Aromatherapy* : clary sage, angelica, mandarin, lavender.
Agitation/terminal Evaluate for organic or drug causes.
restlessness Educate family.
Orient and reassure child; provide calm, nonstimulating environment, use familiar music,
verse, voice, touch.
Aromatherapy* : frankincense, ylang ylang.
* Best if aromatherapy is administered by a practitioner trained in aromatherapy use and safety
and if child has choice of essential oil aroma that stimulates positive response.
From Sourkes B, Frankel L, Brown M, et al: Food, toys, and love: pediatric palliative care, Curr
Probl Pediatr Adolesc Health Care 35:345–392, 2005.
GC/Chlamydia
Strongyloides spp.
Schistosoma spp.
Trypanosoma cruzi
ART, Automated reagin test; CXR, chest radiograph; ELISA, enzyme-linked
immunosorbent assay; FTA-ABS, fluorescent treponemal antibody absorption;
GC, gonococcus; HIV, human immunodeficiency virus; MHA-TP,
microhemagglutination test for Treponema pallidum ; PCR, polymerase chain
reaction; RPR, rapid plasma reagin; VDRL, Venereal Disease Research
Laboratories.
† See Chapter 10 .
Growth Delays
Physical growth delays are common in internationally adopted children and may
represent the combined result of many factors, such as unknown/untreated
medical conditions, malnutrition, and psychological deprivation. It is more
important to monitor growth over time, including preplacement measurements,
since trend data may provide a more objective assessment of the child's
nutritional and medical status. Children who present with low height-for-age
(growth stunting ) may have a history of inadequate nutrition as well as chronic
adversity. Although most children experience a significant catch-up in physical
growth following adoption, many remain shorter than their U.S. peers.
Developmental Delays
Many children adopted internationally exhibit delays in at least 1 area of
development, but most exhibit significant gains within the 1st 12 mo after
adoption. Children adopted at older ages are likely to have more variable
outcomes. In the immediate post-adoption period, it may be impossible to
determine with any certainty whether developmental delays will be transient or
long-lasting. Careful monitoring of development within the first years of
adoption can identify a developmental trend over time that may be more
predictive of long-term functioning than assessment at any specific point in time.
When in doubt, it is better to refer early for developmental intervention, rather
than wait to see if the children will catch up.
Language Development
inadequate health services before placement into foster care mean that children
enter foster care with a high prevalence of chronic medical, mental health,
developmental, dental, and educational problems (Table 9.1 ). Thus they are
defined as children with special health care needs (CSHCN). The greatest single
healthcare need of this population is for high-quality, evidence-based trauma-
informed mental health services to address the impacts of prior and ongoing
trauma, loss, and unpredictability. In addition, children in foster care have higher
rates of asthma, growth failure, obesity, vertically transmitted infections, and
neurologic conditions than the general pediatric population. Adolescents need
access to reproductive health and substance abuse services. Up to 60% of
children <5 yr old have a developmental delay in at least 1 domain and >40% of
school-age children qualify for special education services. Unfortunately,
educational difficulties persist despite improvements in school attendance and
performance after placement in foster care. Each placement change that is
accompanied by a change in school sets children back academically by about 4
mo. Federal legislation requires child welfare to maintain children in their school
of origin when possible, even if child welfare has to provide transportation to
ensure this.
Table 9.1
Health Issues of Children in Foster Care
Chronic Medical Problems
>70% of children have a history of abuse and neglect at entry into foster
care.
Monitor at all health visits for abuse or neglect or poor care in the home.
Developmental Problems
Dental Problems
Educational Problems
Although children in foster care are CSHCN, they often lack access to the
services they need. Most public and private child welfare agencies do not have
formal arrangements for accessing the needed array of health services and rely
on local physicians and health clinics funded by Medicaid. Health histories are
often sparse at admission because many have lacked regular care, or their
biological parents may not be available or forthcoming. Once children enter
foster care, there is often a diffusion of responsibility across caregivers and child
welfare. Foster parents usually receive little information about a child's
healthcare needs, but they are typically expected to decide when and where
children receive healthcare services. Child welfare caseworkers are responsible
for ensuring that a child's health needs are addressed but coordination across
multiple healthcare providers may be daunting. Uncertainty about who is legally
responsible for making healthcare treatment decisions and who may have access
to health information may delay or result in the denial of healthcare services.
Table 9.2
Bibliography
AAP District II Task Force on Health Care for Children in
Foster Care, District II Committee on Early Childhood,
Adoption, and Dependent Care. Fostering health: health care
for children and adolescents in foster care . AAP.: Elk Grove,
IL; 2005 [Available through the American Academy of
is limited tracking of refugees as they move to different cities or states. Thus,
many foreign-born children have had minimal pre- or postarrival screening for
infectious diseases or other health issues.
Immunization requirements and records also vary depending on entry status.
Internationally adopted children who are younger than 10 yr are exempt from
Immigration and Nationality Act regulations pertaining to immunization of
immigrants before arrival in the United States. Adoptive parents are required to
sign a waiver indicating their intention to comply with U.S.-recommended
immunizations, whereas older immigrants need only show evidence of up-to-
date, not necessarily complete, immunizations before application for permanent
resident (green card) status after arrival in the United States.
Infectious diseases are among the most common medical diagnoses identified
in immigrant children after arrival in the United States. Children may be
asymptomatic; therefore, diagnoses must be made by screening tests in addition
to history and physical examination. Because of inconsistent perinatal screening
for hepatitis B and hepatitis C viruses, syphilis, and HIV, and the high
prevalence of certain intestinal parasites and tuberculosis, all foreign-born
children should be screened for these infections on arrival in the United States.
Table 10.1 lists suggested screening tests for infectious diseases. Table 10.2 lists
incubation periods of common internationally acquired diseases. In addition to
these infections, other medical and developmental issues, including hearing,
vision, dental, and mental health assessments; evaluation of growth and
development; nutritional assessment; lead exposure risk; complete blood cell
count with red blood cell indices; microscopic urinalysis; newborn screening
(this could also be done in non-neonates) and/or measurement of thyroid-
stimulating hormone concentration; and examination for congenital anomalies
(including fetal alcohol syndrome) should be considered as part of the initial
evaluation of any immigrant child.*
Table 10.1
Screening Tests for Infectious Diseases in
International Adoptees and Foreign-Born
(Immigrant) Children
Recommended Tests
GC/Chlamydia
Chagas disease serology (endemic areas)
Malaria, thick and thin smears (endemic areas)
Filaria testing (endemic areas)
Urine for O&P for schistosomiasis, if hematuria present
Stool testing for enteric bacteria and viruses in children with diarrhea
† See text.
Table 10.2
Incubation Periods of Common Travel-Related Infections*
LONG
MEDIUM INCUBATION (10-21
SHORT INCUBATION (<10 DAYS) INCUBATION
DAYS)
(>21 DAYS)
Malaria Malaria Malaria
Arboviruses including dengue, yellow fever, Japanese Flaviviruses: tick-borne Schistosomiasis
encephalitis, Zika, chikungunya encephalitis and Japanese Tuberculosis
Hemorrhagic fevers: Lassa, Ebola, South American encephalitis Acute HIV
arenaviruses Hemorrhagic fevers: Lassa, Ebola, infection
Respiratory viruses including severe acute respiratory Crimean-Congo Viral hepatitis
syndrome Acute HIV infection Filariasis
Typhoid and paratyphoid Typhoid and paratyphoid Rickettsia: Q fever
Bacterial enteritis Giardia Secondary syphilis
Rickettsia : spotted fever group—Rocky Mountain Rickettsia : flea-borne, louse- Epstein-Barr virus
spotted fever, African tick typhus, Mediterranean borne, and scrub typhus, Q fever, including
spotted fever, scrub typhus, Q fever spotted fevers (rare) mononucleosis
Bacterial pneumonia including Legionella Cytomegalovirus Amoebic liver
Relapsing fever Toxoplasma disease
Amoebic dysentery Amoebic dysentery Leishmaniasis
Meningococcemia Histoplasmosis Brucella
Brucella (rarely) Brucella Bartonellosis
Leptospirosis Leptospirosis (chronic)
Fascioliasis Babesiosis Babesiosis
Rabies (rarely) Rabies Rabies
African trypanosomiasis (acute), East African (rarely) East African trypanosomiasis West African
(acute) trypanosomiasis
Hepatitis A (rarely) (chronic)
Measles Cytomegalovirus
* Diseases that commonly have variable incubation periods are shown more than once. However,
most diseases may rarely have an atypical incubation period, and this is not shown here.
HIV, Human immunodeficiency virus.
From Freedman DO: Infections in returning travelers. In Bennett JE, Dolin R, Blaser MJ, editors:
Mandell, Douglas, and Bennett's principles and practice of infectious diseases, ed 8, Philadelphia,
2015, Elsevier (Table 324-2).
Table 11.1
Culturally Informed Care
1. Respects the beliefs, values, and lifestyles of patients
2. Understands that health and illness are influenced by ethnic and cultural orientation, religious and spiritual
beliefs, and linguistic considerations
3. Has insight into one's own cultural biases; doesn't see “cultural issues” as something that only affects the
patient
4. Is sensitive to how differences in power and privilege affect the clinical encounter
5. Recognizes that the culturally constructed meaning of illness and health is as important a clinical issue as the
biomedical aspects of disease
6. Sensitive to within group variations in beliefs and practices; avoids stereotyping
The culturally informed physician (1) attempts to understand and respect the
beliefs, values, attitudes, and lifestyles of patients; (2) understands that health
and illness are influenced by ethnic and cultural orientation, religious and
spiritual beliefs, and linguistic considerations; (3) has insight into own cultural
biases and does not see cultural issues as something that only affects the patient;
(4) is sensitive to how differences in power and privilege may affect the quality
of the clinical encounter; (5) recognizes that in addition to the physiologic
aspects of disease, the culturally and psychologically constructed meaning of
illness and health is a central clinical issue; and (6) is sensitive to intragroup
variations in beliefs and practices and avoids stereotyping based on any group
affiliation. These core components of culturally-informed care are important for
interactions with all patients, regardless of race or ethnicity. Culturally sensitive
clinical care is essentially generally sensitive clinical care.
Becoming culturally informed is a developmental process. Fig. 11.1 displays a
framework that includes a continuum of perceptions and orientations to cultural
awareness. Individuals in the denial stage perceive their own cultural orientation
as the true one, with other cultures either undifferentiated or unnoticed. In the
defensive stage, other cultures are acknowledged but regarded as inferior to one's
own culture. The minimization stage is characterized by beliefs that fundamental
similarities among people outweigh any differences, and downplays the role of
culture as a source of human variation. The idea that one should be “color blind”
is an example of a common belief of individuals in the minimization stage.
FIG. 11.1 Development of intercultural sensitivity. (Adapted from Bennett MJ: A
developmental approach to training for intercultural sensitivity, Int J Intercultural
Relations 10(2):179–196, 1986.)
Adapted from Kleinman A, Eisenberg L, Good B: Culture, illness, and care: clinical lessons
from anthropologic and cross-cultural research, Ann Intern Med 88(2):251–258, 1978.
Once the patient's explanatory model is elicited and understood, the clinician
should be able to assess the congruity of this model and the biomedical model,
finding similarities. Then the process of negotiating can occur. Integrating
patient-held approaches to health with evidence-based biomedical standards of
care will help place care within the lifestyle and worldview of patients, leading
to increased adherence to medical care plans, better physician–patient
communication, enhanced long-term therapeutic relationship, and improved
patient (and physician) satisfaction.
11.1
Culture-Specific Beliefs
Robert M. Kliegman
Table 11.3
Cultural Values* Relevant to Health and Health-Seeking
Behavior
nation of origin, specific religious sect, degree of acculturation, age of patient, etc.
Table 11.4
Examples of Disease Beliefs and Health Practices in Select
Cultures
CULTURAL
BELIEF OR PRACTICE
GROUP
Latino Use of traditional medicines (nopales, or cooked prickly pear cactus, as a hypoglycemic agent)
along with allopathic medicine.
Recognition of disorders not recognized in Western allopathic medicine (empacho , in which food
adheres to the intestines or stomach), which are treated with folk remedies but also brought to the
pediatrician.
Cultural interpretation of disease (caida de mollera or “fallen fontanel”) as a cultural interpretation
of severe dehydration in infants.
Muslim Female genital mutilation: practiced in some Muslim countries; the majority do not practice it, and
it is not a direct teaching of the Koran.
Koranic faith healers: use verses from the Koran, holy water, and specific foods to bring about
recovery.
Native Traditional “interpreters” or “healers” interpret signs and answers to prayers. Their advice may be
American sought in addition or instead of allopathic medicine.
Dreams are believed to provide guidance; messages in the dream will be followed.
East and Concepts of “hot” and “cold,” whereby a combination of hot and cold foods and other
Southeast substances (e.g., coffee, alcohol) combine to cause illness. One important aspect is that
Asian Western medicines are considered “hot” by Vietnamese, and therefore nonadherence may
occur if it is perceived that too much of a medicine will make their child's body “hot.” Note:
Hot and cold do not refer to temperatures but are a typology of different foods; for example,
fish is hot and ginger is cold.
Foods, teas, and herbs are also important forms of medicine because they provide balance
between hot and cold.
Bibliography
Bennett MJ. A developmental approach to training for
intercultural sensitivity. Int J Intercult Rel . 1986;10(2):179–
196.
Betancourt JR. Cultural competence and medical education:
many names, many perspectives, one goal. Acad Med .
2006;81(6):499–501.
Dao DK, Goss AL, Hoekzema AS, et al. Integrating theory,
content, and method to foster critical consciousness in
medical students: a comprehensive model for cultural
competence training. Acad Med . 2017;92(3):335–344.
Fisher-Borne M, Cain JM, Martin SL. From mastery to
accountability: cultural humility as an alternative to cultural
competence. Soc Work Educ . 2015;34(2):165–181.
Foronda C, Baptiste DL, Reinholdt MM, Ousman K. Cultural
humility: a concept analysis. J Transcult Nurs .
2016;27(3):210–217.
Gregg J, Saha S. Losing culture on the way to competence: the
use and misuse of culture in medical education. Acad Med .
2006;81(6):542–547.
Hook JN, Davis DE, Owen J, et al. Cultural humility:
FIG. 12.1 Recommendations for preventive pediatric healthcare. (From Bright Futures
/American Academy of Pediatrics. Copyright 2017, American Academy of Pediatrics,
Elk Grove Village, IL. https://www.aap.org/en-us/documents/periodicity_schedule.pdf
.)
Comprehensive guides for care of well infants, children, and adolescents have
been developed based on the Periodicity Schedule to expand and further
recommend how practitioners might accomplish the tasks outlined. The current
guideline standard is The Bright Futures Guidelines for Health Supervision of
Infants, Children, and Adolescents , 4th edition
(https://brightfutures.aap.org/Pages/default.aspx ). These guidelines were
developed by AAP under the leadership of the Maternal Child Health Bureau of
the U.S. Department of Health and Human Services, in collaboration with the
National Association of Pediatric Nurse Practitioners, American Academy of
Family Physicians, American Medical Association, American Academy of
Pediatric Dentistry, Family Voices, and others.
Injury Control
Brian D. Johnston, Frederick P. Rivara
In all high-income countries of the world, and increasingly in many low- and
middle-income countries, injuries are the most common cause of death during
childhood and adolescence beyond the 1st few mo of life (Table 13.1 and Fig.
13.1 ). Injuries represent one of the most important causes of preventable
pediatric morbidity and mortality in the United States. Identification of risk
factors for injuries has led to the development of successful programs for
prevention and control. Strategies for injury prevention and control should be
pursued by the pediatrician in the office, emergency department (ED), hospital,
and community setting and should be done in a multidisciplinary, multifaceted
way.
Table 13.1
Injury Deaths in the United States, 2015* [N (Rate per
100,000)]
Statistics Query and Reporting System (WISQARS) (website). National Center for Injury
Prevention and Control, CDC (producer). https://www.cdc.gov/injury/wisqars/ .
All-cause data from CDC, National Center for Health Statistics: Compressed Mortality File 1999–
2015, Series 20, No 2U, 2016, as compiled from data provided by the 57 vital statistics
jurisdictions through the Vital Statistics Cooperative Program, CDC WONDER online database,
October 2018.
FIG. 13.1 Global injury deaths to children, adolescents, and young adults, 0-29 yr of
age, 2012. (From WHO: Injuries and Violence: The Facts 2012. Geneva: World Health
Organization, 2014.)
Injuries have identifiable risk and protective factors that can be used to define
prevention strategies. The term accidents implies a chance event occurring
without pattern or predictability. In fact, most injuries occur under fairly
predictable circumstances to high-risk children and families. Most injuries are
preventable.
Reduction of morbidity and mortality from injuries can be accomplished not
only through primary prevention (averting the event or injury), but also through
secondary and tertiary prevention. The latter 2 approaches include appropriate
stressed families, and to live in hazardous environments.
Efforts to control injuries include education or persuasion, changes in product
design, and modification of the social and physical environment. Efforts to
persuade individuals, particularly parents, to change their behaviors have
constituted the greater part of injury control efforts. Speaking with parents
specifically about using child car-seat restraints and bicycle helmets, installing
smoke detectors, and checking the tap water temperature is likely to be more
successful than offering well-meaning but too-general advice about supervising
the child closely, being careful, and childproofing the home. This information
should be geared to the developmental stage of the child and presented in
moderate doses in the form of anticipatory guidance at well-child visits. Table
13.2 lists important topics to discuss at each developmental stage. It is important
to acknowledge that there are many barriers to prevention adherence beyond
simple knowledge acquisition; pediatricians should be familiar with low-cost
sources for safety equipment such as bicycle helmets, smoke detectors, trigger
locks, and car seats in their community.
Table 13.2
Injury Prevention Topics for Anticipatory
Guidance by the Pediatrician
Newborn
Car seats
Tap water temperature
Smoke detectors
Sleep safe environments
Infant
Car seats
Tap water temperature
Bath safety
Choking prevention
Seatbelts
Safe storage of firearms in the home
Water skills training
Sports safety and concussion prevention
Seatbelts
Alcohol and drug use, especially while driving and swimming
Mobile phone use while driving
Safe storage of firearms
Sports safety and concussion prevention
Occupational injuries
The most successful injury prevention strategies generally are those involving
changes in product design . These passive interventions protect all individuals
in the population, regardless of cooperation or level of skill, and are likely to be
more successful than active measures that require repeated behavior change by
the parent or child. The most important and effective product changes have been
in motor vehicles, in which protection of the passenger compartment and use of
vehicle deaths among children and adolescents. The peak injury and death rate
for both males and females in the pediatric age-group occurs between 15 and 19
yr (see Table 13.1 ). Proper restraint use in vehicles is the single most effective
method for preventing serious or fatal injury. Table 13.3 shows the
recommended restraints at different ages. Fig. 13.5 provides examples of car
safety seats.
Table 13.3
Recommended Child-Restraint Methods
INFANTS TODDLERS (1-3) YOUNG CHILDREN
Recommended Birth to 1 yr or Older than 1 yr and weight Weight 40-80 lb and height under 4 ft 9 in;
age/weight below weight limit 20-40 lb. generally between 4 and 8 yr of age.
requirements of seat.
Type of seat Infant-only or rear-Convertible or forward- Belt-positioning booster seat.
facing convertible. facing harness seat.
Seat position Rear-facing only. Can be rear-facing until 30 Forward-facing. Place in back seat of vehicle.
Place in back seat lb if seat allows; generally
of vehicle. forward-facing. Place in
back seat of vehicle.
Notes Children should use Harness straps should be at Belt-positioning booster seats must be used
rear-facing seat or above shoulder level. with both lap and shoulder belts.
until at least 1 yr
and at least 20 lb.
Harness straps Most seats require top strap Make sure that lap belt fits low and tightly
should be at or for forward-facing use. across lap/upper thigh area, and that shoulder
below shoulder belt fits snugly, crossing chest and shoulder to
level. avoid abdominal injuries.
Data from http://www.safercar.gov/parents/CarSeats.htm .
FIG. 13.5 Car safety seats. A, Rear-facing infant seat. B, Forward-facing child harness
seat. C, Forward-facing convertible harness seat. D, Low-back booster seat. E, High-
back booster seat. (From Ebel BE, Grossman DC: Crash proof kids? An overview of
current motor vehicle child occupant safety strategies, Curr Probl Pediatr Adolesc
Health Care 33:33–64, 2003. Source: NHTSA.)
Much attention has been given to child occupants <8 yr old. Use of child-
restraint devices, infant car seats, and booster seats can be expected to reduce
fatalities by 71% and the risk of serious injuries by 67% in this age-group. All 50
depression and posttraumatic stress disorder (PTSD) as well as externalizing
problems, including delinquent behavior, aggression, and substance abuse.
The most ubiquitous source of witnessing violence for U.S. children is media
violence , sometimes referred to as virtual violence . This form of violence is
not experienced physically; rather it is experienced in realistic ways through
technology and ever more intense and realistic games. There is an ever-widening
array of screens that are part of children's everyday lives, including computers,
tablets, and cell phones, in addition to long-standing platforms, such as
televisions and movies. Recent tragic events, including mass shootings and acts
of terrorism, have increased the specter of fear among children as these events
are reenacted for them on the multiple screens they encounter. Although
exposure to media/virtual violence cannot be equated to exposure to real-life
violence, many studies confirm that media/virtual violence desensitizes children
to the meaning and impact of violent behavior. Violent video game exposure is
associated with: an increased composite aggression score; increased aggressive
behavior; increased aggressive cognitions; increased aggressive affect, increased
desensitization, and decreased empathy; and increased physiological arousal.
Violent video game use is a risk factor for adverse outcomes; however,
insufficient data exist to examine any potential link between violent video game
use and delinquency or criminal behavior. Table 14.1 lists interventions to reduce
exposure to media violence.
Table 14.1
Public Health Recommendations to Reduce
Effects of Media Violence on Children and
Adolescents
Parents should:
• Be made aware of the risks associated with children viewing
violent imagery, as it promotes aggressive attitudes, antisocial
behavior, fear, and desensitization.
• Review the nature, extent, and context of violence in media
available to their children before children view.
• Assist children's understanding of violent imagery appropriate to
their developmental level.
Professionals should:
• Offer support and advice to parents who allow their children
unsupervised access to extreme violent imagery, as this could
be seen as a form of emotional abuse and neglect.
• Educate all young people in critical film appraisal, in terms of
realism, justification, and consequences.
• Exercise greater control over access to inappropriate violent
media entertainment by young people in secure institutions.
• Use violent film material in anger management programs under
guidance.
Media producers should:
• Reduce violent content, and promote antiviolence themes and
publicity campaigns.
• Ensure that when violence is presented, it is in context and
associated with remorse, criticism, and penalty.
• Ensure that violent action is not justified, or its consequences
understated.
Policymakers should:
• Monitor the nature, extent, and context of violence in all forms
of media, and implement appropriate guidelines, standards, and
penalties.
• Ensure that education in media awareness is a priority and a part
of school curricula.
Impacts of Violence
All types of violence have a profound impact on health and development both
psychologically and behaviorally; it may influence how children view the world
and their place in it. Children can come to see the world as a dangerous and
unpredictable place. This fear may thwart their exploration of the environment,
which is essential to learning in childhood. Children may experience
overwhelming terror, helplessness, and fear, even if they are not immediately in
danger. Preschoolers are most vulnerable to threats that involve the safety (or
perceived safety) of their caretakers. High exposure to violence in older children
Refugee Agency, reported the astounding statistic of 65.6 million people forcibly
displaced worldwide.
Mortality and morbidity related to the long-term effects of war and civil strife
are often higher than that occurring during actual fighting. War and violence are
not listed as leading causes of childhood mortality, but the regions with the
highest levels of child mortality, especially among children <5 yr of age, are the
same locations involved in military conflicts. Nations experiencing conflict
devote substantial portions of their budgets to military expenditures at the
expense of the healthcare infrastructure; a substantial proportion of deaths
attributed to malnutrition, environmentally related infectious disease, or
inadequate immunization are related to the effects of war. Children experiencing
the trauma of wartime violence are at risk for long-term health sequelae, with
greater risk for obesity, hypertension, stroke, and cardiovascular disease.
During wartime, customary patterns of behavior are forced to change,
overcrowding is frequent, and essential resources, such as water and food
staples, may be polluted or contaminated. War is associated with plagues and
epidemics, and novel disease entities can develop. Reemergence of polio or
cholera and the increased virulence of tuberculosis have been associated with
conflict-affected regions and large population displacements.
The morbidity of children exposed to conflicts is significant (Table 14.2 ).
Many more children are physically harmed than killed. Children bear the
psychological scars of war resulting from exposure to violent events, loss of
primary caregivers, and forced removal from their homes. Impressment of
children into service as soldiers or agents is a form of exploitation associated
with long-term problems of adjustment, because child soldiers often lack the
appropriate education and socialization and thus their moral compass is often
misaligned. They are often incapable of understanding the sources of conflict or
why they have been targeted. Their thought processes are more concrete; it is
easier for them to dehumanize their adversaries. Children, who themselves are
exposed to violence and cruelty, frequently become the worst perpetrators of
atrocities.
Table 14.2
Impact of War on Children
Physical
Death
Rape
Abduction
Injuries
Amputations and fractures
Head trauma
Ballistic wounds
Blast injuries
Burns
Chemical and biologic induced
Malnutrition and starvation
Infectious disease
Displacement
Psychosocial
Exploitation
Conscription as soldiers
Coerced involvement in terrorist activities
Prostitution
Slavery
Forced adoption
Children 7-11 Yr
Decline in school performance
Truancy
Sleep disorders
Somatization
Depressive affect
Abnormally aggressive or violent behavior
Irrational fears
Regressive and childish behavior
Expressions of fearfulness, withdrawal, and worry
Adolescents 12-17 Yr
Human trafficking violates the fundamental human rights of child and adult
victims and impacts families, communities, and societies. Trafficked persons
originate from countries worldwide and may belong to any racial, ethnic,
religious, socioeconomic, or cultural group. They may be of any gender.
According to the United Nations Protocol to Prevent, Suppress and Punish
Trafficking in Persons , child trafficking refers to the “recruitment,
transportation, transfer, harboring or receipt of a person” under 18 yr old for
purposes of exploitation. Two major types of trafficking involve forced labor
and sexual exploitation (Table 15.1 ). While adult sex trafficking requires
demonstration of force, fraud, coercion, deception, or the abuse of power as a
means of exploitation, these are not required for persons younger than 18 yr.
Interpretation of the international protocol varies across the globe; U.S. law does
not require movement of a victim to qualify as human trafficking. In addition,
minors who “consent” to commercial sex in the absence of a third party
(trafficker) are victims of commercial sexual exploitation, because their age
precludes true informed consent.
Table 15.1
Types of Exploitation Included in Child
Trafficking
Sexual Exploitation
Prostitution of a child
Production of child sexual exploitation materials (child pornography)
Exploitation in context of travel and tourism
Engaging child in sex-oriented business
Child marriage or forced marriage
Live online sexual abuse
Labor Exploitation
Forced Begging
Forced Criminality
Forced Engagement in Armed Conflict
Illegal Adoption
The word victim is used in this chapter in the legal sense and refers to a person
who has been harmed as a result of a crime or other event. It is not intended to
imply any subjective interpretation of the person's feelings about his/her
situation or imply any judgment about that person's resilience.
Child trafficking may occur within the confines of the child's home country
(domestic trafficking) or may cross national borders (international , or
transnational , trafficking). Globally, victims tend to be trafficked within their
own country or to a country in the same region. In the United States, most
identified child sex trafficking victims are U.S. citizens or legal residents; few
statistical data exist on victims of child labor trafficking. Variations in definitions
of terms, problems with data collection, and underrecognition of victims
complicate estimates of the prevalence of human trafficking, but the
International Labour Organization estimates that 5.5 million of the world's
children are victims of forced labor (this includes human trafficking). In a study
of 55,000 officially identified trafficking victims, the United Nations Office on
Drugs and Crime estimated that approximately 17% were girls and 10% boys.
However, laws that define sexual exploitation in terms of girls and women, as
well as cultural views regarding gender roles, lead to underreporting of boys,
especially as victims of sex trafficking, so their numbers may be higher than
estimated.
Factors creating vulnerability to human trafficking exist at the individual,
family, community, and societal levels (Table 15.2 ). Age is an important risk
factor for adolescents since they are at a stage in their development at which they
have limited life experience, a desire to demonstrate their independence from
parental control, and a level of brain maturation that favors risk-taking and
impulsive behaviors over careful situational analysis and other executive
functions. They are also very interested in social media and are savvy at internet
use, which render them susceptible to online recruitment and solicitation.
Table 15.2
Vulnerability Factors for Child Trafficking
Individual
Family
Poverty
Violence, substance misuse, other dysfunction
Migration
Community
Societal
Recruitment of child victims for labor or sex trafficking often involves false
promises of romance, job opportunities, or a better life. Children may remain in
their exploitative situation for a number of reasons, including fear of violence to
themselves or their loved ones should they attempt escape; guilt and shame for
believing the fraudulent recruitment scheme or engaging in illegal and/or
socially condemned activities; humiliation and fear of criticism by authorities;
debt bondage (believing they owe the trafficker exorbitant amounts of money
and cannot leave until the debt is paid), and fear of arrest and/or deportation.
Many children do not recognize their victimization. Girls who believe their
trafficker is a boyfriend may view their commercial sexual activities as
demonstrations of their love; boys engaging in commercial sex to obtain shelter
or food while living on the street may feel they are exploiting buyers rather than
being victimized. Traffickers may use violence, economic manipulation, and
psychological manipulation to control their victims.
Clinical Presentation
Trafficked persons may seek medical care for any of the myriad physical and
emotional consequences of exploitation. They may present with traumatic
injuries inflicted by traffickers, buyers, or others or injuries related to unsafe
working conditions. They may present with a history of sexual assault, or
symptoms/signs of sexually transmitted infections (STIs) and infections related
to overcrowded, unsanitary conditions. They may request testing for HIV or
complain of signs/symptoms of HIV or infections endemic to the victim's home
country (e.g., malaria, schistosomiasis, tuberculosis). Other clinical presentations
may involve pregnancy and complications of pregnancy or abortion;
malnutrition and/or dehydration; exhaustion; conditions related to exposure to
toxins, chemicals, and dust; and signs and symptoms of posttraumatic stress
disorder (PTSD), major depression, suicidality, behavioral problems with
aggression, and somatization. Some children may have preexisting chronic
medical conditions that have been inadequately treated before or during the
exploitation (e.g., diabetes, seizure disorder, asthma). Trafficked persons may
also seek care for medical issues related to their children.
Many of the same factors that keep victims trapped in their exploitative
conditions also preclude them from disclosing their situation to others. Most
victims presenting for medical care at clinics, hospitals, and emergency
departments do not self-identify as trafficked persons. Consequently, it is
incumbent on the medical professional to be aware of risk factors so that
potential victims may be recognized and offered services. A trafficked child may
present to a medical facility alone, in the company of a parent/guardian (who
may or may not be aware of the trafficking situation), a friend or other person
not involved in the trafficking, a person working for the trafficker (who may
pose as a friend or relative), or the trafficker. Traffickers may be male or female,
adult or juvenile, and they may be family members, acquaintances, friends, or
strangers. On occasion, children are brought in by law enforcement or child
protective services, as known or suspected victims. Table 15.3 lists possible
indicators of labor or sex trafficking. In some cases, the best indicator is the
chief complaint , which may be a condition frequently associated with
trafficking (e.g., teen pregnancy, STI symptoms/signs (especially with history of
prior STI), preventable work-related injury). The practitioner may become
concerned about possible trafficking on recognizing the presence of 1 or more
risk factors (runaway status; recent migration and current work in sector known
for labor trafficking).
Table 15.3
Possible Indicators of Child Trafficking
Indicators at Presentation
Physical Findings
Child withdrawn and with flat affect; fearful; very anxious; intoxicated; or
with inappropriate affect
Motel key(s), multiple cell phones, large amounts of cash, or a few
expensive items (clothing, nails, etc.)
Tattoos (especially with street names or sexual innuendo)
Evidence of remote or acute inflicted injury (suspicious burns, bruising,
signs of strangulation, fractures, closed head injury, thoracoabdominal
trauma)
Malnutrition and/or poor hygiene
Poor dentition and/or dental trauma
Late presentation of illness/injury
Best interest of the child to be primary concern in all actions involving the
child
Protection from discrimination because of gender, race, ethnicity, culture,
socioeconomic status, disability, religion, language, country of origin, or
other status
Right to express views and be heard, appropriate to child's age and
development
Right to obtain information relevant to child, to be given in a way that
children understand
Right to privacy and confidentiality
Right to highest attainable standard of health and to access healthcare
services
Right to dignity, self-respect
Right to consideration of special needs (age, disability, etc.)
Right to respect of cultural and religious beliefs and practices
Trauma-Informed Care
* Appropriate therapy may differ with victims from varied cultures; there is a
United States
Abuse and neglect mostly occur behind closed doors and often are a well-kept
secret. Nevertheless, there were 4 million reports to CPS involving 7.2 million
children in the United States in 2015. Of the 683,000 children with substantiated
reports (9.2 per 1,000 children), 78.3% experienced neglect (including 1.9%
medical neglect), 17.2% physical abuse, 8.4% sexual abuse, and 6.2%
psychological maltreatment. While there had been a decline in rates beginning in
the early 1990s, rates increased in 2014 and 2015 from prior years. Likewise, the
rate of hospitalized children with serious physical abuse has not declined in
recent years. Medical personnel made 9.1% of all reports.
Other sources independent from the official CPS statistics cited above confirm
the prevalence of child maltreatment. In a community survey, 3% of parents
reported using very severe violence (e.g., hitting with fist, burning, using gun or
knife) against their child in the prior year. Considering a natural disinclination to
disclose socially undesirable information, such rates are both conservative and
alarming.
Etiology
Child maltreatment seldom has a single cause; rather, multiple and interacting
biopsychosocial risk factors at 4 levels usually exist. To illustrate, at the
individual level , a child's disability or a parent's depression or substance abuse
predispose a child to maltreatment. At the familial level , intimate partner (or
domestic) violence presents risks for children. Influential community factors
include stressors such as dangerous neighborhoods or a lack of recreational
facilities. Professional inaction may contribute to neglect, such as when the
treatment plan is not clearly communicated. Broad societal factors , such as
poverty and its associated burdens, also contribute to maltreatment. WHO
estimates the rate of homicide of children is approximately 2-fold higher in low-
income compared to high-income countries (2.58 vs 1.21 per 100,000
population), but clearly homicide occurs in high-income countries too. Children
in all social classes can be maltreated, and child healthcare professionals need to
guard against biases concerning low-income families.
In contrast, protective factors , such as family supports, or a mother's concern
for her child, may buffer risk factors and protect children from maltreatment.
Identifying and building on protective factors can be vital to intervening
effectively. One can say to a parent, “I can see how much you love [child's
name]. What can we do to keep her out of the hospital?” Child maltreatment
results from a complex interplay among risk and protective factors. A single
mother who has a colicky baby and who recently lost her job is at risk for
maltreatment, but a loving grandmother may be protective. A good
understanding of factors that contribute to maltreatment, as well as those that are
Table 16.1
Injury Patterns
METHOD OF
PATTERN OBSERVED
INJURY/IMPLEMENT
Grip/grab Relatively round marks that correspond to fingertips and/or thumb
Closed-fist punch Series of round bruises that correspond to knuckles of the hand
Slap Parallel, linear bruises (usually petechial) separated by areas of central sparing
Belt/electrical cord Loop marks or parallel lines of petechiae (the width of the belt/cord) with central
sparing; may see triangular marks from the end of the belt, small circular lesions
caused by the holes in the tongue of the belt, and/or a buckle pattern
Rope Areas of bruising interspersed with areas of abrasion
Other objects/household Injury in shape of object/implement (e.g., rods, switches, and wires cause linear
implements bruising)
Human bite Two arches forming a circular or oval shape, may cause bruising and/or abrasion
Strangulation Petechiae of the head and/or neck, including mucous membranes; may see
subconjunctival hemorrhages
Binding/ligature Marks around the wrists, ankles, or neck; sometimes accompanied by petechiae or
edema distal to the ligature mark
Marks adjacent to the mouth if the child has been gagged
Excessive hincar * Abrasions/burns, especially to knees
Hair pulling Traumatic alopecia; may see petechiae on underlying scalp, or swelling or tenderness
of the scalp (from subgaleal hematoma)
Tattooing or intentional Abusive cases have been described, but can also be a cultural phenomenon (e.g.,
scarring Maori body ornamentation)
*
Punishment by kneeling on salt or other rough substance.
FIG. 16.5 Immersion injury patterns. A, Sparing of the flexoral creases. B, Immersion
“stocking” burn. C, Immersion “glove” burn. D, Immersion buttocks burn. (From Jenny
C: Child abuse and neglect: diagnosis, treatment, and evidence, Philadelphia, 2011,
Saunders, p. 225, Fig 28-3)
Moderate Specificity
Adapted from Coley BD: Caffey's pediatric diagnostic imaging , ed 12, vol 2,
Philadelphia, 2013, Mosby/Elsevier, p 1588 (Box 144-1).
In corroborating the history and the injury, the age of a fracture can be crudely
estimated (Table 16.4 ). Soft tissue swelling subsides in 2-21 days. Subperiosteal
new bone is visible within 6-21 days. Loss of definition of the fracture line
occurs in 10-21 days. Soft callus can be visible after 9 days and hard callus at
14-90 days. These ranges are shorter in infancy and longer in children with poor
nutritional status or a chronic underlying disease. Fractures of flat bones such as
the skull do not form callus and cannot be aged, although soft tissue swelling
indicates approximate recency (within the prior week).
Table 16.4
Timetable of Radiologic Changes in Children's Fractures*
(in Days)
into childhood.
Adapted from Kleinman PK: Diagnostic imaging of child abuse , ed 3, Cambridge, UK, 2015,
Cambridge University Press, p 215.
Abusive head trauma (AHT) results in the most significant morbidity and
mortality. Abusive injury may be caused by direct impact, asphyxia, or shaking.
Subdural hematomas (Fig. 16.8 ), retinal hemorrhages, especially when
extensive and involving multiple layers, and diffuse axonal injury strongly
suggest AHT, especially when they occur together. The poor neck muscle tone
and relatively large heads of infants make them vulnerable to acceleration-
deceleration forces associated with shaking, leading to AHT. Children may lack
external signs of injury, even with serious intracranial trauma. Signs and
symptoms may be nonspecific, ranging from lethargy, vomiting (without
diarrhea), changing neurologic status or seizures, and coma. In all preverbal
children, an index of suspicion for AHT should exist when children present with
these signs and symptoms.
FIG. 16.8 CT scan indicating intracranial bleeding. A arrow, Older blood. B arrow, New
blood.
Acute intracranial trauma is best evaluated by initial and follow-up CT. MRI
is helpful in differentiating extra axial fluid, determining timing of injuries,
assessing parenchymal injury, and identifying vascular anomalies. MRI is best
obtained 5-7 days after an acute injury. Glutaric aciduria type 1 can present with
intracranial bleeding and should be considered. Other causes of subdural
hemorrhage in infants include arteriovenous malformations, coagulopathies,
birth trauma, tumor, and infections. When AHT is suspected, injuries elsewhere
—skeletal and abdominal—should be ruled out.
Retinal hemorrhages are an important marker of AHT (Fig. 16.9 ).
Whenever AHT is being considered, a dilated indirect eye examination by a
pediatric ophthalmologist should be performed. Although retinal hemorrhages
can be found in other conditions, hemorrhages that are multiple, involve >1 layer
of the retina, and extend to the periphery are very suspicious for abuse. The
mechanism is likely repeated acceleration-deceleration from shaking. Traumatic
retinoschisis points strongly to abuse.
With other causes of retinal hemorrhages, the pattern is usually different than
seen in child abuse. After birth, many newborns have them, but they disappear in
2-6 wk. Coagulopathies (particularly leukemia), retinal diseases, carbon
monoxide poisoning, or glutaric aciduria may be responsible. Severe,
noninflicted, direct crush injury to the head can rarely cause an extensive
hemorrhagic retinopathy. CPR rarely, if ever, causes retinal hemorrhage in
infants and children; if present, there a few hemorrhages in the posterior pole.
Trichomonas vaginalis is by culture (Diamond media or InPouch; Biomed
Diagnostics, White City, OR) or wet mount. Wet mount requires the presence of
vaginal secretions, viewing must be immediate for optimal results, and
sensitivity is only 44–68%; therefore false-negative tests are common. Experts
have determined that insufficient data exist to recommend commercially
available Trichomonas NAATs for prepubertal children. However, there is also
no reason to suspect that test performance in children would be different from
adults.
Table 16.5
Indications for STI Screening in Children
With Suspected Sexual Abuse
1. Child has experienced penetration or has evidence of recent or healed
penetrative injury to the genitals, anus, or oropharynx.
2. Child has been abused by a stranger.
3. Child has been abused by a perpetrator known to be infected with a
sexually transmitted infection (STI) or at high risk for STIs (e.g.,
intravenous drug abusers, men who have sex with men, persons with
multiple sexual partners, those with a history of STIs).
4. Child has a sibling, other relative, or another person in the household with
an STI.
5. Child lives in an area with a high rate of STI in the community.
6. Child has signs or symptoms of STIs (e.g., vaginal discharge or pain,
genital itching or odor, urinary symptoms, genital lesions or ulcers).
7. Child or parent requests STI testing.
From Centers for Disease Control and Prevention: Sexually transmitted diseases
treatment guidelines, 2015, MMWR 64(RR3):1–137, 2015.
Table 16.6
† Reports should be made to the agency in the community mandated to receive reports of
** Report if evidence exists to suspect abuse, including history, physical examination, or other
identified infections.
HIV, Human immunodeficiency virus; HSV, herpes simplex virus; SA, sexually associated; ST,
sexually transmitted.
From Centers for Disease Control and Prevention: Sexually transmitted diseases treatment
guidelines, 2015, MMWR 64(RR3):1-137, 2015 (Table 6).
problems that would necessitate a behavior change to improve outcomes are
used throughout the chapter.
FIG. 17.1 The 5 constructs that influence one's intent to perform a
behavior and the 4 influences that determine whether an intent will lead to
performing the behavior. Problem identification (box at upper right) is where
the process of thinking about health behavior changes begins. A clinician
can then help the patient decide on which behavior can help the patient to
meet the health goal. Once this is decided, to help with behavior change,
clinicians should think about intent, influences of intent, and the factors that
may facilitate or impede intent from leading to action.
Table 17.1
Table 17.2
Table 17.3
Table 17.5
Counseling for Obesity Using a Motivational Interviewing (MI) Approach
SPECIFIC
ACTION EXAMPLES
SKILLS
Engagement* O pen-ended “Now that we have finished the majority of the visit, I'd
questions like to talk about your weight. Is that okay? How do
you feel about your size?”
“Mrs. Smith, how do you feel about Jimmy's weight?”
A ffirmations “You definitely have shown how strong you are having
dealt with kids teasing you about your size.”
“Remember when you were having difficulty with
school? You were able to make a few changes, and now
you are doing well. I am confident we can do the same
with your weight.”
R eflective “You are feeling like your son is the same size as
listening everyone in your family, and you aren't concerned right
now.”
“Having your family watch TV before bed really works
for your family, Mrs. Smith.”
“You're not terribly excited about having to think of
ways to cook differently.”
S ummary “So far, we have discussed how challenging it would be to
statements lose weight and make changes for the whole family, but you
are willing to consider some simple changes.”
Focusing Set the agenda. “We could talk about increasing the amount of exercise
Jimmy has every week, reducing screen time, or
making a dietary change. What do you think would
work best?”
“Great, so we will talk about soda. What do you like
about it? How many times a week do you drink it?”
Evocation Reinforce any “Those are great reasons for thinking about cutting
change talk. back on soda.”
Change ruler. “On a scale of 1 to 10, how confident are you (or
important is it) that you can cut back on soda?”
“A 5. Why didn't you answer a 3?”
“What would it take to bring it to a 7?”
Planning Focus on how “Maybe completely eliminating soda is too difficult
to make the right now. Do you want to think of a couple of times
change, not during the week where you can reward yourself with a
“why” soda?”
anymore. “What will you drink after school instead of soda?”
Be concrete.
*
OARS is used to engage the patient and build rapport.
Adapted from Changing the conversation about childhood obesity, American Academy
of Pediatrics, Institute for Healthy Childhood Weight. https://www.aap.org/en-us/about-
the-aap/aap-press-room/pages/Changing-the-conversation-about-Childhood-
Obesity.aspx .
Shared Decision-Making
Shared decision-making has many similarities to the processes previously
described in that it emphasizes moving physicians away from a paternalistic
approach in dictating treatment to one where patients and clinicians collaborate
in making a medical decision, particularly when multiple evidence-based
treatments options exist. The pediatrician or clinician offers different treatment
options and describes the risks and benefits for each one. The patient or
caregiver expresses their values, preferences, and treatment goals, and a decision
is made together.
Shared decision-making is often facilitated by using evidence-based decision
aids such as pamphlets, videos, web-based tools, or educational workshops.
Condition-specific or more generic decision aids have been created and facilitate
the process of shared decision-making. Studies in adults show that such aids
improve knowledge and satisfaction, reduce decisional conflict, and increase the
alignment between patient preferences and treatment options. More study is
needed to assess behavioral and physiologic outcomes specifically when
temperament appears to result from genetic factors.
Table 18.1
much.
Based on data from Chess S, Thomas A: Temperament in clinical practice, New York, 1986,
Guilford.
The concept of temperament can help parents understand and accept the
characteristics of their children without feeling responsible for having caused
them. Children who have difficulty adjusting to change may have behavior
problems when a new baby arrives or at the time of school entry. In addition,
pointing out the child's temperament may allow for adjustment in parenting
styles. Behavioral and emotional problems may develop when the
temperamental characteristics of children and parents are in conflict. For
example, if parents who keep an irregular schedule have a child who is not
readily adaptable, behavioral difficulties are more likely than if the child has
parents who have predictable routines.
Table 18.2
Classic Developmental Stage Theories
SCHOOL
INFANCY TODDLERHOOD PRESCHOOL ADOLESCENCE
AGE
(0-1 YR) (2-3 YR) (3-6 YR) (12-20 YR)
(6-12 YR)
Freud: Oral Anal Phallic/oedipal Latency Genital
psychosexual
Erikson: Basic trust vs Autonomy vs shame Initiative vs guilt Industry vs Identity vs role
psychosocial mistrust and doubt inferiority diffusion
Piaget: Sensorimotor Sensorimotor Preoperational Concrete Formal operations
cognitive operations
Kohlberg: — Preconventional: avoid Conventional: Conventional: Postconventional:
moral punishment/obtain conformity (stage law and order moral principles
rewards (stages 1 and 2) 3) (stage 4)
Psychoanalytic Theories
At the core of Freudian theory is the idea of body-centered (or broadly,
“sexual”) drives; the emotional health of both the child and the adult depends on
adequate resolution of these conflicts. Although Freudian ideas have been
challenged, they opened the door to subsequent theories of development.
Erikson recast Freud's stages in terms of the emerging personality (see Table
18.2 ). The child's sense of basic trust develops through the successful
negotiation of infantile needs. As children progress through these psychosocial
stages, different issues become salient. It is predictable that a toddler will be
preoccupied with establishing a sense of autonomy, whereas a late adolescent
may be more focused on establishing meaningful relationships and an
occupational identity. Erikson recognized that these stages arise in the context of
Western European societal expectations; in other cultures, the salient issues may
be quite different.
Erikson's work calls attention to the intrapersonal challenges facing children at
different ages in a way that facilitates professional intervention. Knowing that
the salient issue for school-age children is industry vs inferiority, pediatricians
inquire about a child's experiences of mastery and failure and (if necessary)
suggest ways to ensure adequate successes.
Cognitive Theories
Cognitive development is best understood through the work of Piaget . A central
tenet of Piaget's work is that cognition changes in quality, not just quantity (see
Table 18.2 ). During the sensorimotor stage, an infant's thinking is tied to
immediate sensations and a child's ability to manipulate objects. The concept of
“in” is embodied in a child's act of putting a block into a cup. With the arrival of
language, the nature of thinking changes dramatically; symbols increasingly take
the place of objects and actions. Piaget described how children actively construct
knowledge for themselves through the linked processes of assimilation (taking
in new experiences according to existing schemata) and accommodation
Similar or Identical Elements Within 5 Theories of Health
Behavior
GENERAL
TENET OF
THE
THEORY THEORY
CONCEPT
HEALTH BELIEF OF OF SOCIAL COGNITIVE TRANSTHEORET
CONCEPT “ENGAGING
MODEL REASONED PLANNED THEORY MODEL
IN THE
ACTION BEHAVIOR
BEHAVIOR
IS LIKELY
IF …”
ATTITUDINAL BELIEFS
Appraisal of The positive Benefits, Behavioral Behavioral Outcome Pros, cons (decision
positive and aspects barriers/health beliefs and beliefs and expectations/expectancies balance)
negative outweigh the motive evaluation of evaluation of
aspects of the negative those beliefs those beliefs
behavior and aspects. (attitudes) (attitudes)
its expected
outcome
SELF-EFFICACY BELIEFS/BELIEFS ABOUT CONTROL OVER THE BEHAVIOR
Belief in One believes Self-efficacy — Perceived Self-efficacy Self-efficacy/tempta
one's ability in one's ability behavioral
to perform to perform the control
the behavior; behavior.
confidence
NORMATIVE AND NORM-RELATED BELIEFS AND ACTIVITIES
Belief that One believes Cues from media, Normative Normative Social support Helping relationship
others want that people friends (cues to beliefs and beliefs and (process of change)
one to important to action) motivation to motivation to
engage in the one want one comply comply
behavior to engage in (subjective (subjective
(and one's the behavior; norms) norms)
motivation to person has
comply); others'
may include support.
actual
support of
others
Belief that One believes — — — Social Social liberation (pr
others (e.g., that other environment/norms; of change)
peers) are people are modeling
engaging in engaging in
the behavior the behavior.
Responses to One receives Cues from media, — — Reinforcement Reinforcement
one's positive friends (cues to management/stimul
behavior that reinforcement action) control (processes o
increase or from others or change)
decrease the creates
likelihood positive
one will reinforcements
engage in the for oneself.
behavior;
may include
reminders
RISK-RELATED BELIEFS AND EMOTIONAL RESPONSES
Belief that One feels at Perceived — — Emotional coping Dramatic relief (pro
one is at risk risk with susceptibility/severity responses/expectancies change)
if one does regard to a (perceived threat) about environmental cues
not engage in negative
the behavior, outcome or
and that the disease.
consequences
may be
severe; may
include
actually
experiencing
negative
emotions or
symptoms
and coping
with them
INTENTION/COMMITMENT/PLANNING
Intending or One has — Behavioral Behavioral Self-control/self- Contemplation/prep
planning to formed strong intentions intentions regulation (stages of change); s
perform the behavioral liberation (process o
behavior; intentions to change)
setting goals engage in the
or making a behavior; one
commitment has set
to perform realistic goals
the behavior or made a firm
commitment
to engage in
the behavior.
From Noar SM, Zimmerman RS: Health behavior theory and cumulative knowledge regarding
health behaviors: are we moving in the right direction? Health Educ Res 20:275–290, 2005, Table
1.
Table 18.4
Relationship Between Standard Deviation (SD) and Normal
Range for Normally Distributed Quantities
OBSERVATIONS INCLUDED IN PROBABILITY OF A “NORMAL” MEASUREMENT DEVIATING
THE NORMAL RANGE FROM THE MEAN BY THIS AMOUNT
SD % SD %
±1 68.3 ≥1 16.0
±2 95.4 ≥2 2.3
±3 99.7 ≥3 0.13
FIG. 18.4 Relationship between percentile lines on the growth curve and frequency
distributions of height at different ages.
For any single measurement, its distance away from the mean can be
expressed in terms of the number of SDs (also called a z score ); one can then
consult a table of the normal distribution to find out what percentage of
measurements fall within that distance from the mean. Software to convert
anthropometric data into z scores for epidemiologic purposes is available. A
measurement that falls “outside the normal range”—arbitrarily defined as 2, or
sometimes 3, SDs on either side of the mean—is atypical, but not necessarily
indicative of illness. The further a measurement (height, weight, IQ) falls from
the mean, the greater is the probability that it represents not simply normal
variation, but rather a different, potentially pathologic condition.
Another way of relating an individual to a group uses percentiles. The
percentile is the percentage of individuals in the group who have achieved a
certain measured quantity (e.g., height of 95 cm) or a developmental milestone
(e.g., walking independently). For anthropometric data, the percentile cutoffs can
be calculated from the mean and SD. The 5th, 10th, and 25th percentiles
correspond to −1.65 SD, −1.3 SD, and −0.7 SD, respectively. Fig. 18.4
demonstrates how frequency distributions of a particular parameter (height) at
different ages relate to the percentile lines on the growth curve.
Bibliography
Chen Y, Baram TZ. Toward understanding how early-life stress
issues, and community violence. Evidence also suggests that some types of
challenging behavior apparent at a young age may persist. In one study, a high
percentage of preschoolers identified as having both internalizing and
externalizing behavior at age 3 yr continued to have similar difficulties at 6 yr.
Other risk factors for the development of challenging behavior include trauma
and developmental problems. Adverse childhood experiences (ACEs) , defined
as abuse and neglect, caregiver substance use, caregiver depression, and
domestic violence or criminality, are often present during childhood. In the
National Survey of Child and Adolescent Well-Being, 42% of children under 6
yr of age in the child welfare system had experienced 4 or more ACEs. Further,
there was a cumulative relationship between emotional and behavioral problems
and ACE exposure, with children exposed to 4 or more ACEs almost 5 times
more likely to have internalizing problems than children not exposed to ACEs. A
similar relationship was found for externalizing problems. Studies involving
children with developmental disabilities suggest emotional and behavioral
problems occur more frequently in this group than in typically developing
children. These children may have delays in self-regulation and communication
skills as well as increased family stress, which contribute to the increased
likelihood of behavioral challenges.
Table 19.1
Parenting as an Intervention
The influence of parenting practices on child behavior, development, and overall
adjustment has led to efforts to teach parenting as a method of primary
prevention. The Video Interaction Project (VIP) uses a coaching and education
model with recorded parent–child interactions to foster positive parenting
behavior. These parenting behaviors range from reading aloud to encouraging
interactive play. In an urban, low-income, primary care setting, parent and child
outcomes for the VIP group were compared to those from a lower-intensity
intervention (parent mailings encouraging positive parenting behaviors) and a
control group. VIP produced the most robust impacts on socioemotional
outcomes, including increased attention and decreased distress with separation,
hyperactivity, and externalizing behavior in toddlers.
Positive parenting as a public health intervention has resulted in decreased
rates of substantiated child maltreatment cases, out-of-home placements, and
child maltreatment injuries. Other effective public health approaches include
home-visiting programs, which have been deployed to at-risk families in an
effort to improve maternal and child outcomes. The Maternal, Infant and Early
Childhood Home Visiting Program, authorized as part of the Affordable Care
Act of 2010 and again in 2015, is part of the Medicare Access and Children's
Health Insurance Program (CHIP) Reauthorization Act. A key component of
home-visiting programs is the promotion of positive parenting behavior to foster
child developmental and school readiness. Group parenting programs have been
deployed as primary prevention to promote emotional and behavioral adjustment
in young children. There is moderate-quality evidence that group-based
parenting programs may improve parent–child interactions. These programs
typically employ praise, encouragement, and affection and have been associated
with improved self-esteem and social and academic competence.
Parenting behaviors have also been employed as an intervention to treat
emotional and behavioral problems in young children. Parenting interventions
such as Incredible Years, Triple P Positive Parenting Program, and New Forrest
Parenting Program are effective for at least short-term improvements in child
conduct problems, parental mental health, and parenting practices. Also called
parent training programs , most teach the importance of play, rewards, praise,
and consistent discipline and allow parents to practice new skills. This active-
learning component distinguishes parent training programs from educational
programs, which have been shown to be less effective.
Teaching emotional communication skills and positive parent–child
interaction skills are associated with parent training programs that demonstrate a
greater increase in parenting skills (Table 19.2 ). Several components are
associated with programs that show greater improvements in child externalizing
behavior: teaching parents to use time-out correctly, respond consistently, and
interact positively with their children. All successful programs require parents to
practice parenting skills during the program.
Table 19.2
Fetal Period
From the 9th wk on (fetal period), somatic changes consist of rapid body growth
as well as differentiation of tissues, organs, and organ systems. Fig. 20.1 depicts
changes in body proportion. By wk 10, the face is recognizably human. The
midgut returns to the abdomen from the umbilical cord, rotating
counterclockwise to bring the stomach, small intestine, and large intestine into
their normal positions. By wk 12, the gender of the external genitals becomes
clearly distinguishable. Lung development proceeds, with the budding of
bronchi, bronchioles, and successively smaller divisions. By wk 20-24, primitive
alveoli have formed and surfactant production has begun; before that time, the
absence of alveoli renders the lungs useless as organs of gas exchange.
CHAPTER 21
The Newborn
John M. Olsson
Prenatal Factors
Pregnancy is a period of psychologic preparation for the profound demands of
parenting. Women may experience ambivalence, particularly (but not
exclusively) if the pregnancy was unplanned. If financial concerns, physical
illness, prior miscarriages or stillbirths, or other crises interfere with psychologic
preparation, the neonate may not be welcomed. For adolescent mothers, the
demand that they relinquish their own developmental agenda, such as an active
social life, may be especially burdensome.
The early experience of being mothered may establish unconsciously held
expectations about nurturing relationships that permit mothers to “tune in” to
their infants. These expectations are linked with the quality of later infant–parent
interactions. Mothers whose early childhoods were marked by traumatic
separations, abuse, or neglect may find it especially difficult to provide
consistent, responsive care. Instead, they may reenact their childhood
experiences with their own infants, as if unable to conceive of the mother–child
relationship in any other way. Bonding may be adversely affected by several risk
factors during pregnancy and in the postpartum period that undermine the
mother–child relationship and may threaten the infant's cognitive and emotional
development (Table 21.1 ).
Table 21.1
Prenatal Risk Factors for Attachment
Recent death of a loved one
Previous loss of or serious illness in another child
Prior removal of a child
History of depression or serious mental illness
History of infertility or pregnancy loss
Troubled relationship with parents
Financial stress or job loss
Marital discord or poor relationship with the other parent
Recent move or no community ties
No friends or social network
Unwanted pregnancy
No good parenting model
Experience of poor parenting
Drug and/or alcohol abuse
Extreme immaturity
From Dixon SD, Stein MT: Encounters with children: pediatric behavior and
development, ed 4, Philadelphia, 2006, Mosby, p 131.
Social support during pregnancy, particularly support from the father and
close family members, is also important. Conversely, conflict with or
abandonment by the father during pregnancy may diminish the mother's ability
to become absorbed with her infant. Anticipation of an early return to work may
make some women reluctant to fall in love with their babies because of
anticipated separation. Returning to work should be delayed for at least 6 wk, by
which time feeding and basic behavioral adjustments have been established.
Many decisions have to be made by parents in anticipation of the birth of their
child. One important choice is that of how the infant will be nourished. Among
the important benefits of breastfeeding is its promotion of bonding. Providing
breastfeeding education for the parents at the prenatal visit by the pediatrician
and by the obstetrician during prenatal care can increase maternal confidence in
breastfeeding after delivery and reduce stress during the newborn period (see
Chapter 56 ).
1. The mother is asked to underline the response that comes closest to how
she has been feeling in the previous 7 days.
2. All 10 items must be completed.
3. Care should be taken to avoid the possibility of the mother discussing her
answers with others.
4. The mother should complete the scale herself, unless she has limited
English or has difficulty with reading.
5. The Edinburgh Postnatal Depression Scale may be used at 6-8 wk to
screen postnatal women. The child health clinic, a postnatal checkup, or a
home visit may provide a suitable opportunity for its completion.
Name:
Address:
Baby's age:
Because you have recently had a baby, we would like to know how you are
feeling. Please underline the answer that comes closest to how you have
felt in the past 7 days, not just how you feel today.
Here is an example, already completed.
I have felt happy:
Yes, all the time
Yes, most of the time
No, not very often
No, not at all
This would mean: “I have felt happy most of the time” during the past
week. Please complete the other questions in the same way.
In the past 7 days:
1. I have been able to laugh and see the funny side of things
As much as I always could
Not quite so much now
Definitely not so much now
Not at all
2. I have looked forward with enjoyment to things
As much as I ever did
Rather less than I used to
Definitely less than I used to
Hardly at all
*3. I have blamed myself unnecessarily when things went wrong
Yes, most of the time
Yes, some of the time
Not very often
No, never
4. I have been anxious or worried for no good reason
No, not at all
Hardly ever
Yes, sometimes
Yes, very often
*5. I have felt scared or panicky for no very good reason
Yes, quite a lot
Yes, sometimes
No, not much
No, not at all
*6. Things have been getting on top of me
Yes, most of the time I haven't been able to cope at all
Yes, sometimes I haven't been coping as well as usual
No, most of the time I have coped quite well
No, I have been coping as well as ever
*7. I have been so unhappy that I have had difficulty sleeping
Yes, most of the time
Yes, sometimes
Not very often
No, not at all
*8. I have felt sad or miserable
Yes, most of the time
Yes, quite often
Not very often
No, not at all
*9. I have been so unhappy that I have been crying
Yes, most of the time
Yes, quite often
Only occasionally
No, never
*10. The thought of harming myself has occurred to me
Yes, quite often
Sometimes
Hardly ever
Never
Physical Examination
Examination of the newborn should include an evaluation of growth (see
Chapter 20 ) and an observation of behavior . The average term newborn
interactions with caregivers or the wider environment may alter experience-
dependent processes that are critical to brain structure development and function
during infancy. Although for some processes, subsequent stimulation may allow
catch-up, as the periods of plasticity close during the rapid developmental
changes occurring in infancy, more permanent deficits may result.
The infant acquires new competences in all developmental domains. The
concept of developmental trajectories recognizes that complex skills build on
simpler ones; it is also important to realize how development in each domain
affects functioning in all the others. All growth parameters should be plotted
using the World Health Organization charts, which show how children from birth
through 72 mo “should” grow under optimal circumstances (see Chapter 23 ,
Figs. 23.1 and 23.2 ). Table 22.1 presents an overview of key milestones by
domain; Table 22.2 presents similar information arranged by age. Table 22.3
presents age at time of x-ray appearance of centers of ossification. Parents often
seek information about “normal development” during this period and should be
directed to reliable sources, including the American Academy of Pediatrics
website (healthychildren.org ).
Table 22.1
Developmental Milestones in 1st 2 Yr of Life
AVERAGE AGE OF
MILESTONE DEVELOPMENTAL IMPLICATIONS
ATTAINMENT (MO)
GROSS MOTOR
Holds head steady while sitting 2 Allows more visual interaction
Pulls to sit, with no head lag 3 Muscle tone
Brings hands together in midline 3 Self-discovery of hands
Asymmetric tonic neck reflex gone 4 Can inspect hands in midline
Sits without support 6 Increasing exploration
Rolls back to stomach 6.5 Truncal flexion, risk of falls
Walks alone 12 Exploration, control of proximity to parents
Runs 16 Supervision more difficult
FINE MOTOR
Grasps rattle 3.5 Object use
Reaches for objects 4 Visuomotor coordination
Palmar grasp gone 4 Voluntary release
Transfers object hand to hand 5.5 Comparison of objects
Thumb-finger grasp 8 Able to explore small objects
Turns pages of book 12 Increasing autonomy during book time
Scribbles 13 Visuomotor coordination
Builds tower of 2 cubes 15 Uses objects in combination
Builds tower of 6 cubes 22 Requires visual, gross, and fine motor
coordination
COMMUNICATION AND LANGUAGE
Smiles in response to face, voice 1.5 More active social participant
Monosyllabic babble 6 Experimentation with sound, tactile sense
Inhibits to “no” 7 Response to tone (nonverbal)
Follows 1-step command with 7 Nonverbal communication
gesture
Follows 1-step command without 10 Verbal receptive language (e.g., “Give it to
gesture me”)
Says “mama” or “dada” 10 Expressive language
Points to objects 10 Interactive communication
Speaks first real word 12 Beginning of labeling
Speaks 4-6 words 15 Acquisition of object and personal names
Speaks 10-15 words 18 Acquisition of object and personal names
Speaks 2-word sentences (e.g., 19 Beginning grammatization, corresponds with
“Mommy shoe”) 50-word vocabulary
COGNITIVE
Stares momentarily at spot where 2 Lack of object permanence (out of sight, out of
object disappeared mind; e.g., yarn ball dropped)
Stares at own hand 4 Self-discovery, cause and effect
Bangs 2 cubes 8 Active comparison of objects
Uncovers toy (after seeing it 8 Object permanence
hidden)
Egocentric symbolic play (e.g., 12 Beginning symbolic thought
pretends to drink from cup)
Uses stick to reach toy 17 Able to link actions to solve problems
Pretend play with doll (e.g., gives 17 Symbolic thought
doll bottle)
Table 22.2
Emerging Patterns of Behavior During the 1st Yr of Life*
NEONATAL PERIOD (1ST 4 WK)
Prone: Lies in flexed attitude; turns head from side to side; head sags on ventral suspension
Supine: Generally flexed and a little stiff
Visual: May fixate face on light in line of vision; doll's eye movement (oculocephalic reflex) of eyes on
turning of the body
Reflex: Moro response active; stepping and placing reflexes; grasp reflex active
Social: Visual preference for human face
AT 1 MO
Prone: Legs more extended; holds chin up; turns head; head lifted momentarily to plane of body on ventral
suspension
Supine: Tonic neck posture predominates; supple and relaxed; head lags when pulled to sitting position
Visual: Watches person; follows moving object
Social: Body movements in cadence with voice of other in social contact; beginning to smile
AT 2 MO
Prone: Raises head slightly farther; head sustained in plane of body on ventral suspension
Supine: Tonic neck posture predominates; head lags when pulled to sitting position
Visual: Follows moving object 180 degrees
Social: Smiles on social contact; listens to voice and coos
AT 3 MO
Prone: Lifts head and chest with arms extended; head above plane of body on ventral suspension
Supine: Tonic neck posture predominates; reaches toward and misses objects; waves at toy
Sitting: Head lag partially compensated when pulled to sitting position; early head control with bobbing
motion; back rounded
Reflex: Typical Moro response has not persisted; makes defensive movements or selective withdrawal
reactions
Social: Sustained social contact; listens to music; says “aah, ngah”
AT 4 MO
Prone: Lifts head and chest, with head in approximately vertical axis; legs extended
Supine: Symmetric posture predominates, hands in midline; reaches and grasps objects and brings them to
mouth
Sitting: No head lag when pulled to sitting position; head steady, tipped forward; enjoys sitting with full
truncal support
Standing: When held erect, pushes with feet
Adaptive: Sees raisin, but makes no move to reach for it
Social: Laughs out loud; may show displeasure if social contact is broken; excited at sight of food
AT 7 MO
Prone: Rolls over; pivots; crawls or creep-crawls (Knobloch)
Supine: Lifts head; rolls over; squirms
Sitting: Sits briefly, with support of pelvis; leans forward on hands; back rounded
Standing: May support most of weight; bounces actively
Adaptive: Reaches out for and grasps large object; transfers objects from hand to hand; grasp uses radial palm;
rakes at raisin
Language: Forms polysyllabic vowel sounds
Social: Prefers mother; babbles; enjoys mirror; responds to changes in emotional content of social contact
AT 10 MO
Sitting: Sits up alone and indefinitely without support, with back straight
Standing: Pulls to standing position; “cruises” or walks holding on to furniture
Motor: Creeps or crawls
Adaptive: Grasps objects with thumb and forefinger; pokes at things with forefinger; picks up pellet with
assisted pincer movement; uncovers hidden toy; attempts to retrieve dropped object; releases object
grasped by other person
Language: Repetitive consonant sounds (“mama,” “dada”)
Social: Responds to sound of name; plays peek-a-boo or pat-a-cake; waves bye-bye
AT 1 YR
Motor: Walks with one hand held; rises independently, takes several steps (Knobloch)
Adaptive: Picks up raisin with unassisted pincer movement of forefinger and thumb; releases object to other
person on request or gesture
Language: Says a few words besides “mama,” “dada”
Social: Plays simple ball game; makes postural adjustment to dressing
* Data are derived from those of Gesell (as revised by Knobloch), Shirley, Provence, Wolf, Bailey,
and others.
Data from Knobloch H, Stevens F, Malone AF: Manual of developmental diagnosis, Hagerstown,
MD, 1980, Harper & Row.
Table 22.3
Time of Radiographic Appearance of Centers of
Ossification in Infancy and Childhood
BOYS—AGE AT BONES AND EPIPHYSEAL GIRLS—AGE AT
APPEARANCE* CENTERS APPEARANCE*
HUMERUS, HEAD
3 wk 3 wk
CARPAL BONES
2 mo ± 2 mo Capitate 2 mo ± 2 mo
3 mo ± 2 mo Hamate 2 mo ± 2 mo
30 mo ± 16 mo Triangular † 21 mo ± 14 mo
42 mo ± 19 mo Lunate † 34 mo ± 13 mo
67 mo ± 19 mo Trapezium † 47 mo ± 14 mo
69 mo ± 15 mo Trapezoid † 49 mo ± 12 mo
66 mo ± 15 mo Scaphoid † 51 mo ± 12 mo
No standards available Pisiform † No standards available
METACARPAL BONES
18 mo ± 5 mo II 12 mo ± 3 mo
20 mo ± 5 mo III 13 mo ± 3 mo
23 mo ± 6 mo IV 15 mo ± 4 mo
26 mo ± 7 mo V 16 mo ± 5 mo
32 mo ± 9 mo I 18 mo ± 5 mo
FINGERS (EPIPHYSES)
16 mo ± 4 mo Proximal phalanx, 3rd finger 10 mo ± 3 mo
16 mo ± 4 mo Proximal phalanx, 2nd finger 11 mo ± 3 mo
17 mo ± 5 mo Proximal phalanx, 4th finger 11 mo ± 3 mo
19 mo ± 7 mo Distal phalanx, 1st finger 12 mo ± 4 mo
21 mo ± 5 mo Proximal phalanx, 5th finger 14 mo ± 4 mo
24 mo ± 6 mo Middle phalanx, 3rd finger 15 mo ± 5 mo
24 mo ± 6 mo Middle phalanx, 4th finger 15 mo ± 5 mo
26 mo ± 6 mo Middle phalanx, 2nd finger 16 mo ± 5 mo
28 mo ± 6 mo Distal phalanx, 3rd finger 18 mo ± 4 mo
28 mo ± 6 mo Distal phalanx, 4th finger 18 mo ± 5 mo
32 mo ± 7 mo Proximal phalanx, 1st finger 20 mo ± 5 mo
37 mo ± 9 mo Distal phalanx, 5th finger 23 mo ± 6 mo
37 mo ± 8 mo Distal phalanx, 2nd finger 23 mo ± 6 mo
39 mo ± 10 mo Middle phalanx, 5th finger 22 mo ± 7 mo
152 mo ± 18 mo Sesamoid (adductor pollicis) 121 mo ± 13 mo
HIP AND KNEE
Usually present at birth Femur, distal Usually present at birth
Usually present at birth Tibia, proximal Usually present at birth
4 mo ± 2 mo Femur, head 4 mo ± 2 mo
46 mo ± 11 mo Patella 29 mo ± 7 mo
FOOT AND ANKLE ‡
* To nearest month.
† Except for the capitate and hamate bones, the variability of carpal centers is too great to make
them very useful clinically.
‡ Standards for the foot are available, but normal variation is wide, including some familial
variants, so this area is of little clinical use.
Values represent mean ± standard deviation, when applicable.
The norms present a composite of published data from the Fels Research Institute, Yellow
B, Length for age and weight for age for girls, birth to 24 mo.
(Courtesy World Health Organization: WHO Child Growth
Standards, 2014.)
Cognitive Development
Exploration of the environment increases in parallel with improved dexterity
(reaching, grasping, releasing) and mobility. Learning follows the precepts of
Piaget's sensorimotor stage (see Chapter 18 ). Toddlers manipulate objects in
novel ways to create interesting effects, such as stacking blocks or filling and
dumping buckets. Playthings are also more likely to be used for their intended
purposes (combs for hair, cups for drinking). Imitation of parents and older
siblings or other children is an important mode of learning. Make-believe play
(symbolic play ) centers on the child's own body, such as pretending to drink
from an empty cup (Table 23.1 ; see also Table 22.1 ).
Table 23.1
Emerging Patterns of Behavior From 1-5 Yr of Age*
15 MO
Motor: Walks alone; crawls up stairs
Adaptive: Makes tower of 3 cubes; makes a line with crayon; inserts raisin in bottle
Language: Jargon; follows simple commands; may name a familiar object (e.g., ball); responds to his/her name
Social: Indicates some desires or needs by pointing; hugs parents
18 MO
Motor: Runs stiffly; sits on small chair; walks up stairs with 1 hand held; explores drawers and wastebaskets
Adaptive: Makes tower of 4 cubes; imitates scribbling; imitates vertical stroke; dumps raisin from bottle
Language: 10 words (average); names pictures; identifies 1 or more parts of body
Social: Feeds self; seeks help when in trouble; may complain when wet or soiled; kisses parent with pucker
24 MO
Motor: Runs well, walks up and down stairs, 1 step at a time; opens doors; climbs on furniture; jumps
Adaptive: Makes tower of 7 cubes (6 at 21 mo); scribbles in circular pattern; imitates horizontal stroke; folds
paper once imitatively
Language: Puts 3 words together (subject, verb, object)
Social: Handles spoon well; often tells about immediate experiences; helps to undress; listens to stories when
shown pictures
30 MO
Motor: Goes up stairs alternating feet
Adaptive: Makes tower of 9 cubes; makes vertical and horizontal strokes, but generally will not join them to
make cross; imitates circular stroke, forming closed figure
Language: Refers to self by pronoun “I”; knows full name
Social: Helps put things away; pretends in play
36 MO
Motor: Rides tricycle; stands momentarily on 1 foot
Adaptive: Makes tower of 10 cubes; imitates construction of “bridge” of 3 cubes; copies circle; imitates cross
Language: Knows age and sex; counts 3 objects correctly; repeats 3 numbers or a sentence of 6 syllables; most of
speech intelligible to strangers
Social: Plays simple games (in “parallel” with other children); helps in dressing (unbuttons clothing and puts
on shoes); washes hands
48 MO
Motor: Hops on 1 foot; throws ball overhand; uses scissors to cut out pictures; climbs well
Adaptive: Copies bridge from model; imitates construction of “gate” of 5 cubes; copies cross and square; draws
man with 2-4 parts besides head; identifies longer of 2 lines
Language: Counts 4 pennies accurately; tells story
Social: Plays with several children, with beginning of social interaction and role-playing; goes to toilet alone
60 MO
Motor: Skips
Adaptive: Draws triangle from copy; names heavier of 2 weights
Language: Names 4 colors; repeats sentence of 10 syllables; counts 10 pennies correctly
Social: Dresses and undresses; asks questions about meaning of words; engages in domestic role-playing
* Data derived from those of Gesell (as revised by Knobloch), Shirley, Provence, Wolf, Bailey, and
others. After 6 yr, the Wechsler Intelligence Scales for Children (WISC-IV) and other scales offer
the most precise estimates of cognitive development. To have their greatest value, they should be
administered only by an experienced and qualified person.
Emotional Development
Infants who are approaching the developmental milestone of taking their 1st
steps may be irritable. Once they start walking, their predominant mood changes
markedly. Toddlers are often elated with their new ability and with the power to
control the distance between themselves and their parents. Exploring toddlers
orbit around their parents, moving away and then returning for a reassuring
touch before moving away again. A child with secure attachment will use the
process is reciprocal between the child and the school. It is developmentally
based, recognizing the importance of early experiences for later development.
Rather than delaying school entry, high-quality early-education programs may be
the key to ultimate school success.
School makes increasing cognitive demands on the child. Mastery of the
elementary curriculum requires that many perceptual, cognitive, and language
processes work efficiently (Table 25.1 ), and children are expected to attend to
many inputs at once. The 1st 2-3 yr of elementary school are devoted to
acquiring the fundamentals: reading, writing, and basic mathematics skills. By
3rd grade, children need to be able to sustain attention through a 45 min period,
and the curriculum requires more complex tasks. The goal of reading a
paragraph is no longer to decode the words, but to understand the content; the
goal of writing is no longer spelling or penmanship, but composition. The
volume of work increases along with the complexity.
Table 25.1
Selected Perceptual, Cognitive, and Language Processes
Required for Elementary School Success
Table 27.1
Growth Velocity and Other Growth Characteristics by Age
Variation of normal
Constitutional tall stature
Familial tall stature
Endocrine conditions
Growth hormone excess
Precocious puberty
Congenital adrenal hyperplasia
Obesity
Genetic conditions
Marfan syndrome
Klinefelter syndrome
Sotos syndrome
Dental Development
Dental development includes mineralization, eruption, and exfoliation (Table
27.4 ). Initial mineralization begins as early as the 2nd trimester (mean age for
central incisors, 14 wk) and continues through 3 yr of age for the primary
(deciduous) teeth and 25 yr of age for the secondary (permanent) teeth.
Mineralization begins at the crown and progresses toward the root. Eruption
begins with the central incisors and progresses laterally. Exfoliation begins at
about 6 yr of age and continues through 12 yr. Eruption of the permanent teeth
may follow exfoliation immediately or may lag by 4-5 mo. The timing of dental
development is poorly correlated with other processes of growth and maturation.
Delayed eruption is usually considered when no teeth have erupted by
approximately 13 mo of age (mean + 3 SD). Common causes include congenital
or genetic disorders, endocrine disorders (e.g., hypothyroidism,
hypoparathyroidism), familial conditions, and (the most common) idiopathic
conditions. Individual teeth may fail to erupt because of mechanical blockage
(crowding, gum fibrosis). Causes of early exfoliation include hypophosphatasia,
histiocytosis X, cyclic neutropenia, leukemia, trauma, and idiopathic factors.
Nutritional and metabolic disturbances, prolonged illness, and certain
medications (tetracycline) frequently result in discoloration or malformations of
the dental enamel. A discrete line of pitting on the enamel suggests a time-
limited insult.
Table 27.4
Chronology of Human Dentition of Primary (Deciduous)
and Secondary (Permanent) Teeth
Bibliography
Centers for Disease Control and Prevention, National Center for
Health Statistics. CDC growth charts (website) .
http://www.cdc.gov/growthcharts/ .
De Onis M, Garza C, Onyango AW, et al. Comparison of the
WHO child growth standards and the CDC 2000 growth
charts. J Nutr . 2007;137:144–148.
Foote JM, Kirouac N, Lipman TH. PENS position statement on
linear growth measurement of children. J Pediatr Nurs .
2015;30(2):425–426.
Grummer-Strawn LM, Reinold C, Krebs NF, Centers for
Disease Control and Prevention. Use of World Health
Organization and CDC growth charts for children aged 0-59
months in the United States. MMWR Recomm Rep .
2010;59(RR–9):1–15.
Guo SS, Roche AF, Chumlea WC, et al. Growth in weight,
recumbent length and head circumference for preterm low-
birthweight infants during the first three years of life using
gestation-adjusted ages. Early Hum Dev . 1997;47:305–325.
incumbent on the pediatric clinician to conduct regular developmental
surveillance and periodic developmental screening at primary care health
supervision visits aimed at early identification and treatment.
Among the many types of developmental or behavioral conditions, the most
common include language problems, affecting at least 1 in 10 children (see
Chapter 52 ); behavior or emotional disorders, affecting up to 25% of children,
with 6% considered serious; attention-deficit/hyperactivity disorder, affecting 1
in 10 children (Chapter 49 ); and learning disabilities, affecting up to 10%
(Chapters 50 and 51 ). Less common and more disabling are the intellectual
disabilities (1–2%; Chapter 53 ); autism spectrum disorders (1 in 59 children;
Chapter 54 ); cerebral palsy and related motor impairments (0.3%, or 1 in 345
children; Chapter 616 ); hearing impairment, also referred to as deafness, hard-
of-hearing, or hearing loss (0.12%; Chapter 655 ); and nonrefractive vision
impairment (0.8%; Chapter 639 ).
Developmental Observation
Negative Indicators
Activities That the Child Cannot Do:
Sit unsupported by 12 mo
Walk by 18 mo (boys) or 2 yr (girls) (check creatine kinase urgently)
Walk other than on tiptoes
Run by 2.5 yr
Hold object placed in hand by 5 mo (corrected for gestation)
Reach for objects by 6 mo (corrected for gestation)
Point at objects to share interest with others by 2 yr
* Most children do not have “red flags” and thus require quality screening to
Adapted from Horridge KA. Assessment and investigation of the child with
disordered development. Arch Dis Child Educ Pract Ed 96:9–20, 2011.
Key historical elements include (1) eliciting and attending to the parents' or
caregivers' concerns around the child's development or behavior; (2) obtaining a
history of the child's developmental skills and behavior at home, with peers, in
school, and in the community; and (3) identifying the risks, strengths, and
protective factors for development and behavior in the child and family,
including the social determinants of health. During the office visit, the clinician
should make and document direct observations of the child's developmental
skills and behavioral interactions. Skills in all streams of development should be
considered along with observations of related neurologic functioning made on
physical examination.
With this history and observation, the clinician should create and maintain a
longitudinal record of the child's development and behavior for tracking the
child across visits. It is often helpful to obtain information from and share
information with other professionals involved with the child, including childcare
professionals, home visitors, teachers, after-school providers, and developmental
therapists. This provides a complete picture of the child's development and
behavior and allows collaborative tracking of the child's progress.
Table 28.3
Standardized Tools for General Developmental Screening
Table 28.4
Standardized Tools for General Behavioral Screening
Table 28.5
Standardized Tools for Language and Autism Screening
NUMBER
AGE ADMINISTRATION PURCHASE/ OBTAINMENT
SCREENING TEST OF REF*
RANGE TIME INFORMATION
ITEMS
LANGUAGE
Communication and 6-24 mo 24 5-10 min Paul H. Brookes Publishing 1
Symbolic Behavior Co
Scales: Developmental 800/638-3775
Profile (CSBS-DP): www.brookespublishing.com
Infant Toddler
Checklist
AUTISM
Modified Checklist for 16-48 20 (avg) 5-10 min www.m-chat.org/ 2
Autism in Toddlers, mo Follow up interview †
Revised with Follow-
up (M-CHAT-R/F)
Social Communication 4+ yr 40 (avg) 5-10 min Western Psychological 3, 4
Questionnaire (SCQ) Services
www.wpspublish.com
* Key reference sources:
Table 29.1
Conditions That Do and Do Not Require Exclusion From
Group Childcare Settings
The caregiver/teacher should determine if the illness (1) prevents the child
from participating comfortably in activities; (2) results in a need for care that is
greater than the staff can provide without compromising the health and safety of
other children; (3) poses a risk of spread of harmful diseases to others; or (4)
causes a fever and behavior change or other signs and symptoms (e.g., sore
throat, rash, vomiting, diarrhea). An unexplained temperature above 100°F
(37.8°C) (armpit) in a child <6 mo old should be medically evaluated. Any
infant <2 mo old with fever should receive immediate medical attention.
Most families need to arrange to keep sick children at home, such as staying
home from work or having backup plans with an alternative caregiver.
Alternative care arrangements outside the home for sick children are relatively
rare but may include either (1) care in the child's own center, if it offers special
provisions designed for the care of ill children (sometimes called the infirmary
model or sick daycare ), or (2) care in a center that serves only children with
pediatricians can help parents determine how to adjust childcare arrangements to
best meet their child's specific needs (e.g., allergies, eating and sleeping habits,
temperament and stress-regulation capacities). For most parents, finding
childcare that they can afford, access, manage, and accept as a good environment
for their child is a difficult and often distressing process. Many parents also
worry about how their child will fare in childcare (e.g., Will their child feel
distressed by group settings, suffer from separation from the parents, or even be
subjected to neglect or abuse?). These worries are especially likely among low-
income parents with fewer family and community resources. A few parents may
think of childcare only as “babysitting” and may not consider the consequences
for their child's cognitive, linguistic, and social development, focusing solely on
whether the child is safe and warm. These parents may be less likely to select a
high-quality childcare arrangement, which is especially problematic if the family
is facing socioeconomic challenges that already place them at risk of receiving
lower-quality care for their children. For these parents, it is vital to stress the
importance of quality and its implications for their child's cognitive, language,
and behavioral development and school readiness.
Table 29.2
Table 31.1
Normal Developmental Changes in Children's Sleep
SLEEP
AGE DURATION*
ADDITIONAL SLEEP ISSUES SLEEP DISORDERS
CATEGORY AND SLEEP
PATTERNS
Newborn (0-2 Total sleep: American Academy of Most sleep issues perceived as
mo) 10-19 hr per Pediatrics issued a revised problematic at this stage represent a
24 hr (average, recommendation in 2016 discrepancy between parental
13-14.5 hr), advocating against bed- expectations and developmentally
may be higher sharing in the 1st yr of life, appropriate sleep behaviors. Newborns
in premature instead encouraging who are extremely fussy and persistently
babies proximate but separate difficult to console, as noted by parents,
Bottle-fed sleeping surfaces for mother are more likely to have underlying
babies and infant for at least the 1st 6 medical issues such as colic,
generally sleep mo and preferably 1st yr of gastroesophageal reflux, and formula
for longer life. intolerance.
periods (2-5 hr Safe sleep practices for
bouts) than infants:
breastfed • Place baby on his or her back to
babies (1-3 sleep at night and during nap
hr). times.
Sleep periods • Place baby on a firm mattress
are separated with well-fitting sheet in safety-
by 1-2 hr approved crib.
awake. • Do not use pillows or comforters.
No established • Standards require crib bars to be
nocturnal-
no farther apart than in.
diurnal pattern
• Make sure baby's face and head
in 1st few wk;
stay uncovered and clear of
sleep is evenly
blankets and other coverings
distributed
during sleep.
throughout the
day and night,
averaging 8.5
hr at night and
5.75 hr during
day.
Infant (2-12 Recommended Sleep regulation or self- Behavioral insomnia of childhood;
mo) sleep duration (4- soothing involves the infant's sleep-onset association type
12 mo) is 12-16 hr ability to negotiate the sleep– Sleep-related rhythmic movements
(note that there is wake transition, both at sleep (head banging, body rocking)
great individual onset and following normal
variability in sleep awakenings throughout the
times during night. The capacity to self-
infancy). soothe begins to develop in
the 1st 12 wk of life and is a
reflection of both
neurodevelopmental
maturation and learning.
Sleep consolidation, or
“sleeping through the night,”
is usually defined by parents
as a continuous sleep episode
without the need for parental
intervention (e.g., feeding,
soothing) from the child's
bedtime through the early
morning. Infants develop the
ability to consolidate sleep
between 6 wk and 3 mo.
Toddler (1-2 Recommended Cognitive, motor, social, and Behavioral insomnia of childhood,
yr) sleep amount language developmental sleep-onset association type
is 11-14 hr issues impact sleep. Behavioral insomnia of childhood,
(including Nighttime fears develop; limit-setting type
naps). transitional objects and
Naps decrease bedtime routines are
from 2 to 1 important.
nap at average
age of 18 mo.
Preschool (3- Recommended Persistent cosleeping tends to be Behavioral insomnia of childhood, limit-
5 yr) sleep amount highly associated with sleep setting type
is 10-13 hr problems in this age-group.
(including Sleep problems may become Sleepwalking, sleep terrors, nighttime
naps). chronic. fears/nightmares, obstructive sleep
Overall, 26% apnea syndrome
of 4 yr olds
and just 15%
of 5 yr olds
nap.
Middle Recommended School and behavior problems Nightmares
childhood (6- sleep amount is 9- may be related to sleep problems.
12 yr) 12 hr. Media and electronics, such as Obstructive sleep apnea syndrome
television, computer, video Insufficient sleep
games, and the Internet,
increasingly compete for sleep
time.
Irregularity of sleep–wake
schedules reflects increasing
discrepancy between school
and non–school night
bedtimes and wake times.
Adolescence Recommended Puberty-mediated phase delay Insufficient sleep
(13-18 yr) sleep amount (later sleep onset and wake Delayed sleep–wake phase disorder
is 8-10 hr. times), relative to sleep-wake Narcolepsy
Later cycles in middle childhood Restless legs syndrome/periodic
bedtimes; Earlier required wake times limb movement disorder
increased Environmental competing
discrepancy priorities for sleep
between sleep
patterns on
weekdays and
weekends
* All recommended sleep amounts from Paruthi S, Brooks LJ, D'Ambrosio C, et al: Recommended
amount of sleep for pediatric populations: a consensus statement of the American Academy of
Sleep Medicine. J Clin Sleep Med 12:785–786, 2016.
FIG. 31.1 Mismatch between sleep needs/duration and time in bed,
resulting in insomnia.
Etiology
OSAS results from an anatomically or functionally narrowed upper airway; this
typically involves some combination of decreased upper airway patency (upper
airway obstruction and/or decreased upper airway diameter), increased upper
airway collapsibility (reduced pharyngeal muscle tone), and decreased drive to
breathe in the face of reduced upper airway patency (reduced central ventilatory
drive) (Table 31.4 ). Upper airway obstruction varies in degree and level (i.e.,
nose, nasopharynx/oropharynx, hypopharynx) and is most frequently caused by
adenotonsillar hypertrophy, although tonsillar size does not necessarily correlate
with degree of obstruction, especially in older children. Other causes of airway
obstruction include allergies associated with chronic rhinitis or nasal obstruction;
craniofacial abnormalities, including hypoplasia or displacement of the maxilla
and mandible; gastroesophageal reflux with resulting pharyngeal reactive edema
(see Chapter 349 ); nasal septal deviation (Chapter 404 ); and velopharyngeal
flap cleft palate repair. Reduced upper airway tone may result from
neuromuscular diseases, including hypotonic cerebral palsy and muscular
dystrophies (see Chapter 627 ), or hypothyroidism (Chapter 581 ). Reduced
central ventilatory drive may be present in some children with Arnold-Chiari
malformation (see Chapter 446 ); rapid-onset obesity with hypothalamic
dysfunction, hypoventilation, and autonomic dysregulation (Chapter 60.1 ); and
meningomyelocele (Chapter 609.4 ). In other situations the etiology is mixed;
individuals with Down syndrome (see Chapter 98.2 ), because of their facial
anatomy, hypotonia, macroglossia, and central adiposity, as well as the increased
incidence of hypothyroidism, are at particularly high risk for OSAS, with some
estimates of prevalence as high as 70%.
Table 31.4
Anatomic Factors That Predispose to
Obstructive Sleep Apnea Syndrome and
Hypoventilation in Children
Nose
Adenotonsillar hypertrophy
Macroglossia
Cystic hygroma
Velopharyngeal flap repair
Cleft palate repair
Pharyngeal mass lesion
Craniofacial
Micrognathia/retrognathia
Midface hypoplasia (e.g., trisomy 21, Crouzon disease, Apert syndrome)
Mandibular hypoplasia (Pierre Robin, Treacher Collins, Cornelia de Lange
syndromes)
Craniofacial trauma
Skeletal and storage diseases
Achondroplasia
Storage diseases (e.g., glycogen; Hunter, Hurler syndromes)
Clinicians should clinically reassess all patients with OSAS for persisting
signs and symptoms after therapy to determine whether further treatment
is required. (Evidence Quality: Grade B; Recommendation Strength:
Recommendation.)
Adapted from Marcus CL, Brooks LJ, Draper KA, et al: Diagnosis and
management of childhood obstructive sleep apnea syndrome. Pediatrics
130:576–584, 2012.
Parasomnias
Parasomnias are episodic nocturnal behaviors that often involve cognitive
disorientation and autonomic and skeletal muscle disturbance. Parasomnias may
be further characterized as occurring primarily during non-REM sleep (partial
arousal parasomnias) or in association with REM sleep, including nightmares,
hypnogogic hallucinations, and sleep paralysis; other common parasomnias
include sleep-talking and hypnic jerks or “sleep starts.”
Etiology
Partial arousal parasomnias represent a dissociated sleep–wake state, the
neurobiology of which remains unclear, although genetic factors and an intrinsic
oscillation of subcortical-cortical arousal with sleep have been proposed. These
episodic events, which include sleepwalking, sleep terrors, and confusional
arousals, are more common in preschool and school-age children because of the
relatively higher percentage of SWS in younger children. Partial arousal
parasomnias typically occur when SWS predominates, in the 1st third of the
night. In contrast, nightmares , which are much more common than partial
arousal parasomnias but are often confused with them, tend to be concentrated in
the last third of the night, when REM sleep is most prominent. Any factor
associated with an increase in the relative percentage of SWS (certain
medications, previous sleep restriction) may increase the frequency of events in
a predisposed child. There appears to be a genetic predisposition for both
sleepwalking and night terrors. Partial arousal parasomnias may also be difficult
to distinguish from nocturnal seizures. Table 31.6 summarizes similarities and
differences among these nocturnal arousal events.
Table 31.6
Key Similarities and Differentiating Features Between Non-
REM and REM Parasomnias as Well as Nocturnal Seizures
CONFUSIONAL SLEEP NOCTURNAL
SLEEPWALKING NIGHTMARES
AROUSALS TERRORS SEIZURES
Time Early Early Early-mid Late Any
Sleep stage SWA SWA SWA REM Any
EEG − − − − +
discharges
Scream − ++++ − ++ +
Autonomic + ++++ + + +
activation
Motor − + +++ + ++++
activity
Awakens − − − + +
Duration 0.5-10; more gradual 1-10; more 2-30; more gradual 3-20 5-15; abrupt
(min) offset gradual offset offset onset and offset
Postevent + + + − +
confusion
Age Child Child Child Child, young Adolescent,
adult young adult
Genetics + + + − ±
Organic CNS − − − − ++++
lesion
CNS, Central nervous system; EEG, electroencephalogram; REM, rapid eye movement; SWA,
slow-wave arousal.
From Avidan A, Kaplish N: The parasomnias: epidemiology, clinical features and diagnostic
approach. Clin Chest Med 31:353–370, 2010.
Epidemiology
Many children sleepwalk on at least one occasion; the lifetime prevalence by age
10 yr is 13%. Sleepwalking (somnambulism) may persist into adulthood, with
the prevalence in adults of approximately 4%. The prevalence is approximately
10 times greater in children with a family history of sleepwalking. The peak
prevalence of sleep terrors is 34% at age 1-5 yr, decreasing to 10% by age 7;
the age at onset is usually between 4 and 12 yr. Because of the common genetic
predisposition, the likelihood of developing sleepwalking after age 5 is almost 2-
fold higher in children with a history of sleep terrors. Although sleep terrors can
occur at any age from infancy through adulthood, most individuals outgrow
sleep terrors by adolescence. Confusional arousals (sleep drunkenness, sleep
inertia) usually occur with sleepwalking and sleep terrors; prevalence rates have
been estimated at >15% in children age 3-13 yr.
Clinical Manifestations
The partial arousal parasomnias have several features in common. Because they
typically occur at the transition out of “deep” sleep or SWS, partial arousal
neurologic disorder characterized by an almost irresistible urge to move the legs,
often accompanied by uncomfortable sensations in the lower extremities. Both
the urge to move and the sensations are usually worse at rest and in the evening
and are at least partially relieved by movement, including walking, stretching,
and rubbing, but only if the motion continues. RLS is a clinical diagnosis that is
based on the presence of these key symptoms (Table 31.7 ).
Table 31.7
Diagnostic Criteria for Restless Legs
Syndrome
A. An urge to move legs, usually accompanied by or in response to
uncomfortable and unpleasant sensations in the legs, characterized by the
following:
1. The urge to move the legs begins or worsens during periods of
rest or inactivity.
2. The urge to move the legs is partially or totally relieved by
movement.
3. The urge to move the legs is worse in the evening or at night than
during the day, or occurs only in the evening or at night.
B. The symptoms in Criterion A occur at least three times per week and have
persisted for at least 3 months.
C. The symptoms in Criterion A are accompanied by significant distress or
impairment in social, occupational, educational, academic, behavioral, or
other important areas of functioning.
D. The symptoms in Criterion A are not attributable to another mental
disorder or medical condition (e.g., arthritis, leg edema, peripheral
ischemia, leg cramps) and are not better explained by a behavioral
condition (e.g., positional discomfort, habitual foot tapping).
E. The symptoms are not attributable to the physiological effects of a drug or
abuse or medication (e.g., akathisia).
Specify whether:
Narcolepsy without cataplexy but with hypocretin deficiency: Criterion
B requirements of low CSF hypocretin-1 levels and positive
polysomnography/multiple sleep latency test are met, but no cataplexy is
present (Criterion B1 not met).
Narcolepsy with cataplexy but without hypocretin deficiency: In this
rare sub-type (less than 5% of narcolepsy cases), Criterion B
requirements of cataplexy and positive polysomnography/multiple sleep
latency test are met, but CSF hypocretin-1 levels are normal (Criterion
B2 not met).
Autosomal dominant cerebellar ataxia, deafness, and narcolepsy: This
sub-type is caused by exon 21 DNA (cytosine-5)-methyltransferase-1
mutations and is characterized by late-onset (age 30-40 years) narcolepsy
(with low or intermediate CSF hypocretin-1 levels), deafness, cerebellar
ataxia, and eventually dementia.
Autosomal dominant narcolepsy, obesity, and type 2 diabetes:
Narcolepsy, obesity, and type 2 diabetes are low CSF hypocretin-1 levels
have been described in rare cases and are associated with a mutation in
the myelin oligodendrocyte glycoprotein gene.
Narcolepsy without cataplexy but with hypocretin deficiency: This sub-
type is for narcolepsy that develops secondary to medical conditions that
cause infectious (e.g., Whipple's disease, sarcoidosis), traumatic, or
tumoral destruction of hypocretin neurons.
Severity:
Mild: Infrequent cataplexy (less than once per week), need for naps only
once or twice per day, and less disturbed nocturnal sleep.
Moderate: Cataplexy once daily or every few days, disturbed nocturnal
sleep and need for multiple naps daily.
Severe: Drug-resistant cataplexy with multiple attacks daily, nearly
constant sleepiness, and disturbed nocturnal sleep (i.e., movements,
insomnia, and vivid dreaming).
Table 31.9
BEARS Sleep Screening Algorithm
The BEARS instrument is divided into 5 major sleep domains, providing a comprehensive screen for the
major sleep disorders affecting children 2-18 yr old. Each sleep domain has a set of age-appropriate “trigger
questions” for use in the clinical interview.
B = Bedtime problems
E = Excessive daytime sleepiness
A = Awakenings during the night
R = Regularity and duration of sleep
S = Snoring
EXAMPLES OF DEVELOPMENTALLY APPROPRIATE TRIGGER QUESTIONS
Toddler/Preschool Child School-Age Child (6-12 yr) Adolescent (13-18 yr)
(2-5 yr)
1. Bedtime Does your child have any Does your child have any Do you have any problems
problems problems going to bed? problems at bedtime? (P) falling asleep at bedtime? (C)
Falling asleep? Do you any problems going to
bed? (C)
2. Excessive Does your child seem Does your child have difficulty Do you feel sleepy a lot during
daytime overtired or sleepy a lot waking in the morning, seem the day? In school? While
sleepiness during the day? Does your sleepy during the day, or take driving? (C)
child still take naps? naps? (P)
Do you feel tired a lot? (C)
3. Awakenings Does your child wake up a Does your child seem to wake Do you wake up a lot at night?
during the lot at night? up a lot at night? Any Do you have trouble getting
night sleepwalking or nightmares? (P) back to sleep? (C)
Do you wake up a lot at night?
Do you have trouble getting
back to sleep? (C)
4. Regularity Does your child have a What time does your child go to bed What time do you usually go
and regular bedtime and wake and get up on school days? to bed on school nights?
duration of time? What are they? Weekends? Do you think your child Weekends? How much sleep
sleep is getting enough sleep? (P) do you usually get? (C)
5. Snoring Does your child snore a Does your child have loud or nightly Does your teenager snore
lot or have difficulty snoring or any breathing difficulties loudly or nightly? (P)
breathing at night? at night? (P)
C, Child; P, parent.
From The Action Signs Project, Center for the Advancement of Children's
Mental Health at Columbia University.
Table 32.2
HEADSS* Screening Interview for Taking a
Rapid Psychosocial History
Parent Interview
Home
• How well does the family get along with each other?
Education
Activities
Drugs
Sexuality
• Are there any issues regarding sexuality or sexual activity that are of
concern to you?
Suicide/Depression
Adolescent Interview
Home
Education
Activities
Drugs
Sexuality
• Are there any issues regarding sexuality or sexual activity that are of
concern to you?
Suicide/Depression
• Everyone feels sad or angry some of the time. How about you?
• Did you ever feel so upset that you wished you were not alive or so angry
you wanted to hurt someone else badly?
Table 32.3
Select List of Mental Health Rating Scales in the Public
Domain
Psychopharmacology
David R. DeMaso, Heather J. Walter
Questions remain about the quality of the evidence supporting the use of many
psychotropic medications in children and adolescents. Therefore, cognitive,
emotional, and behavioral symptoms are targets for medication treatment when
(1) there is no or insufficient response to available evidence-based psychosocial
interventions, (2) the patient's symptoms convey significant risk of harm, or (3)
the patient is experiencing significant distress or functional impairment.
Common target symptoms include agitation, aggression, anxiety, depression,
hyperactivity, inattention, impulsivity, mania, obsessions, compulsions, and
psychosis (Table 33.2 ). All these can be quantitatively measured with
standardized symptom rating scales to establish baseline symptom severity and
facilitate “treating to target.”
Table 33.2
FDA SELECT
GENERIC (BRAND) DAILY
APPROVED DAILY MEDICAL
APPROXIMATE TARGET THERAPEUTIC
(Pediatric STARTING MONITORING
DURATION OF SYMPTOMS DOSAGE
age range in DOSE AND
ACTION RANGE*
years) PRECAUTIONS
STIMULANTS
Long Acting
OROS ADHD (6+) Inattention 18 mg Age 6-12: Personal and
methylphenidate Hyperactivity 18-54 mg family CV
(Concerta) Impulsivity Age >12: 18- history; personal
12 hr 72 mg seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse;
potential for GI
obstruction
Dexmethylphenidate ADHD (6+) Inattention 5 mg 5-30 mg Personal and
(Focalin XR) † Hyperactivity family CV
10-12 hr Impulsivity history; personal
seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Amphetamine ADHD (6+) Inattention 5 mg 5-30 mg Personal and
combination Hyperactivity family CV
(Adderall XR) † Impulsivity history; personal
12 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Lisdexamfetamine ADHD (6+) Inattention 20 mg 20-70 mg Personal and
(capsule † and Hyperactivity family CV
chewable) Impulsivity history; personal
(Vyvanse) seizure history;
12 hr Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Methylphenidate ADHD (6+) Inattention 10 mg 10-30 mg Personal and
transdermal Hyperactivity family CV
(Daytrana) Impulsivity history; personal
12 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse;
skin reactions
Methylphenidate ADHD (6+) Inattention 20 mg 20-60 mg Personal and
suspension Hyperactivity family CV
(Quillivant XR) Impulsivity history; personal
12 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Intermediate Acting
Methylphenidate ADHD (6+) Inattention 10 mg 10-60 mg Personal and
(Metadate CD, Hyperactivity family CV
Ritalin LA) Impulsivity history; personal
8 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Dextroamphetamine ADHD (6+) Inattention 5 mg 5-40 mg Personal and
(Dexedrine Hyperactivity family CV
Spansule) Impulsivity history; personal
8 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Methylphenidate ADHD (6+) Inattention 20 mg 20-60 mg Personal and
chewable Hyperactivity family CV
(Quillichew ER) Impulsivity history; personal
8 hr seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Short Acting
Dexmethylphenidate ADHD (6+) Inattention 5 mg 5-20 mg Personal and
(Focalin) Hyperactivity family CV
4-5 hr Impulsivity history; personal
seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Methylphenidate ADHD (6+) Inattention 5 mg 5-60 mg Personal and
(Ritalin, Methylin) Hyperactivity family CV
4 hr Impulsivity history; personal
seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Amphetamine ADHD (3+) Inattention Age 3-5: 5-40 mg Personal and
combination Hyperactivity 2.5 mg family CV
(Adderall) Impulsivity Age ≥6: 5 history; personal
4-5 hr mg seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
Dextroamphetamine ADHD (3+) Inattention Age 3-5: 5-40 mg Personal and
(Dexedrine) Hyperactivity 2.5 mg family CV
4 hr Impulsivity Age ≥6: 5 history; personal
mg seizure history;
Ht, Wt, BP, P;
bipolar or
psychotic
symptoms;
substance abuse
SELECTIVE NOREPINEPHRINE REUPTAKE INHIBITOR
Atomoxetine ADHD (6+) Inattention <70 kg: <70 kg: 0.5- Personal and
(Strattera) Hyperactivity 0.5 1.2 family CV
24 hours Impulsivity mg/kg/day mg/kg/day history; BP, P;
>70 kg: >70 kg: 40- liver injury;
40 mg 100 mg suicidal ideation;
bipolar or
psychotic
symptoms
ALPHA (α)-AGONISTS
Short Acting
Clonidine None Inattention 0.05 mg 27-40.5 kg: CV history; BP,
(Catapres) Hyperactivity 0.05-0.2 mg P; rebound
4 hr Impulsivity 40.5-45 kg: hypertension;
0.05-0.3 mg cardiac
>45 kg: 0.05- conduction
0.4 mg abnormalities
Guanfacine (Tenex) None Inattention 0.5 mg 27-40.5 kg: CV history; BP,
6 hr Hyperactivity 0.5-2 mg P; rebound
Impulsivity 40.5-45 kg: hypertension;
0.5-3 mg cardiac
>45 kg: 0.5-4 conduction
mg abnormalities
Long Acting
Clonidine (Kapvay) ADHD (6+) Inattention 0.1 mg 0.1-0.4 mg CV history; BP,
12 hr Hyperactivity P; rebound
Impulsivity hypertension;
cardiac
conduction
abnormalities
Guanfacine ADHD (6+) Inattention 1 mg Monotherapy CV history; BP,
(Intuniv) Hyperactivity : P; rebound
24 hr Impulsivity 25-33.9 kg: hypertension,
2-3 mg cardiac
34-41.4 kg: conduction
2-4 mg abnormalities
41.5-49.4 kg:
3-5 mg
49.5-58.4 kg:
3-6 mg
58.5-91 kg:
4-7 mg
>91 kg: 5-7
mg
Adjunctive
(with
stimulant):
0.05-0.12
mg/kg/day
* Doses shown in table may exceed maximum recommended dose for some children.
FDA, U.S. Food and Drug Administration; CV, cardiovascular; Ht, height; Wt, weight; BP, blood
pressure; P, pulse; GI, gastrointestinal.
Antidepressants
Antidepressant drugs act on pre- and postsynaptic receptors affecting the release
and reuptake of brain neurotransmitters, including norepinephrine, serotonin,
and dopamine (Table 33.4 ). There is strong evidence for the effectiveness of
antidepressant medications in the treatment of anxiety and obsessive-compulsive
disorders and weaker evidence for the treatment of depressive disorders. Suicidal
thoughts have been reported during treatment with all antidepressants. The
overall risk difference of suicidal ideation/attempts across all randomized
controlled trials (RCTs) of antidepressants and indications has been reported as
0.7%, corresponding to a number needed to harm of 143. All antidepressants
carry an FDA warning for suicidality; careful monitoring is recommended during
the initial stages of treatment and following dose adjustments.
Table 33.4
Select Medications for Depression and Anxiety in Children
and Adolescents
FDA SELECT
DAILY
APPROVED DAILY MEDICAL
GENERIC THERAPEUTIC
(Pediatric age TARGET SYMPTOMS STARTING MONITORING
(BRAND) DOSAGE
range in DOSE AND
RANGE*
years) PRECAUTIONS
SELECTIVE SEROTONIN REUPTAKE INHIBITORS
Citalopram None Depression 10 mg 10-40 mg Suicidal ideation;
(Celexa) Anxiety QT prolongation
Obsessions/compulsions at doses >40 mg;
abnormal
bleeding; mania;
SS, DS
Escitalopram Depression Depression 5 mg 5-20 mg Suicidal ideation;
(Lexapro) (12-17) Anxiety abnormal
Obsessions/compulsions bleeding; mania;
SS, DS
Fluoxetine Depression Depression Age 6- Depression: Suicidal ideation;
(Prozac) (8-17) Anxiety 12: 10 10-20 mg abnormal
OCD (7- Obsessions/compulsions mg Anxiety, bleeding; mania;
17) Age 13- OCD: 10-60 SS
17: 20 mg
mg
Sertraline OCD (6-17) Depression Age 6- 12.5-200 mg Suicidal ideation;
(Zoloft) Anxiety 12: abnormal
Obsessions/compulsions 12.5-25 bleeding; mania;
mg SS, DS
Age 13-
17: 25-
50 mg
ATYPICAL ANTIDEPRESSANTS
Bupropion None Depression 150 mg 150-300 mg Suicidal ideation;
(Wellbutrin neuropsychiatric
XL) reaction, seizures
(>300 mg/day),
BP; mania;
contraindicated in
patients with
seizure and eating
disorders
Duloxetine Anxiety (7-17) Depression 30 mg 30-60 mg Suicidal ideation;
(Cymbalta) Anxiety BP, P; liver
damage; severe
skin reactions;
abnormal
bleeding; mania;
SS, DS
Mirtazapine None Depression 7.5 mg 7.5-45 mg Suicidal ideation;
(Remeron) weight;
somnolence;
agranulocytosis;
QT prolongation;
mania; SS, DS
Venlafaxine None Depression 37.5 mg 37.5-225 mg Suicidal ideation;
(Effexor XR) Anxiety BP; abnormal
bleeding; mania;
SS, DS
TRICYCLIC ANTIDEPRESSANTS
Clomipramine OCD (10-17) Obsessions 25 mg 25-200 mg Suicidal ideation;
(Anafranil) Compulsions BP; P; ECG;
blood level;
mania; SS;
seizures; DS
ANXIOLYTIC AGENTS (SITUATIONAL USE)
Lorazepam None Anxiety 0.5 mg 0.5-2 mg Respiratory
(Ativan) depression;
sedation; physical
and psychological
dependence;
paradoxical
reactions
Clonazepam None Panic 0.5 mg 0.5-1 mg Respiratory
(Klonopin) depression;
sedation; physical
and psychological
dependence;
paradoxical
reactions; suicidal
ideation
Hydroxyzine Anxiety Anxiety 50 mg Age <6: 50 QT prolongation
(Atarax, mg
Vistaril) Age >6: 50-
100 mg
* Doses shown in table may exceed maximum recommended dose for some children.
OCD, Obsessive-compulsive disorder; BP, blood pressure; P, pulse; ECG, electrocardiogram; SS,
serotonin syndrome; DS, discontinuation syndrome.
Table 33.5
Select Medications for Psychosis, Mania, Irritability,
Agitation, Aggression, and Tourette Disorder in Children
and Adolescents
SELECT
FDA DAILY
DAILY MEDICAL
GENERIC APPROVED TARGET THERAPEUTIC
STARTING MONITORING
(BRAND) (Pediatric age SYMPTOMS DOSAGE
DOSE AND
range in years) RANGE*
PRECAUTIONS
SECOND-GENERATION ANTIPSYCHOTICS
Aripiprazole Bipolar (10- Mania Bipolar, Bipolar, BMI, BP, P,
(Abilify) 17) Psychosis schizophrenia: schizophrenia: fasting glucose
Available in Schizophrenia Irritability 2 mg 10-30 mg and lipids,
liquid (13-17) Aggression Autism: 2 mg Autism: 5-15 abnormal
preparation Irritability in Agitation Tourette: 2 mg movements;
autism (6-17) Vocal/motor mg Tourette: 5-20 compulsive
Tourette (6- tics mg behaviors;
17) neuroleptic
malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures
Olanzapine Bipolar (13- Mania 2.5 mg 2.5-20 mg BMI, BP, P,
(Zyprexa) 17) Psychosis fasting glucose
Available in Schizophrenia Agitation and lipids,
liquid, (13-17) abnormal
dissolvable, movements; skin
and IM rash (DRESS);
preparations neuroleptic
malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures
Quetiapine Bipolar (10- Mania 25 mg bid Bipolar: 400- BMI, BP, P,
(Seroquel) 17) Psychosis 600 mg fasting glucose
Schizophrenia Agitation Schizophrenia: and lipids,
(13-17) 400-800 mg abnormal
movements;
ophthalmologic
exam; neuroleptic
malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures; QT
prolongation
Risperidone Bipolar (10- Mania Bipolar, Bipolar, BMI, BP, P,
(Risperdal) 17) Psychosis schizophrenia: schizophrenia: fasting glucose
Available in Schizophrenia Irritability 0.5 mg 1-6 mg and lipids,
liquid and (13-17) Aggression Autism: Autism: 0.5-3 prolactin,
dissolvable Irritability in Agitation <20 kg: 0.25 mg abnormal
preparations autism (5-17) mg movements;
≥20 kg: 0.5 neuroleptic
mg malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures
Paliperidone Schizophrenia Psychosis 3 mg <51 kg: 3-6 BMI, BP, P,
(Invega) (12-17) mg fasting glucose
Available in ≥51 kg: 3-12 and lipids,
liquid and mg prolactin,
IM abnormal
preparations movements, QT
prolongation;
neuroleptic
malignant
syndrome;
potential for GI
obstruction;
leukopenia,
neutropenia,
agranulocytosis;
seizures
Lurasidone Schizophrenia Psychosis 40 mg 40-80 mg BMI, BP, P,
(Latuda) (13-17) fasting glucose
and lipids,
prolactin,
abnormal
movements;
neuroleptic
malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures
Asenapine Bipolar (10-17) Mania 2.5 mg twice daily 5-20 mg BMI, BP, P,
(Saphris) Psychosis fasting glucose
and lipids,
prolactin,
abnormal
movements; QT
prolongation;
neuroleptic
malignant
syndrome;
leukopenia,
neutropenia,
agranulocytosis;
seizures
FIRST-GENERATION ANTIPSYCHOTIC
Haloperidol Psychosis Mania 0.05 mg/kg/day 0.05-0.15 BP, P; abnormal
(Haldol) Tourette Psychosis mg/kg/day movements; QT
Available in disorder Irritability prolongation;
liquid and Severe Aggression neuroleptic
IM behavioral Agitation malignant
preparations disorders Vocal/motor syndrome;
Agitation (3- tics encephalopathy
17) when combined
with lithium;
leukopenia,
neutropenia,
agranulocytosis
MOOD STABILIZER
Lithium Bipolar (12-17) Mania Acute mania: Long-term Serum level,
carbonate 1800 mg/day control: 900- CBC/diff, thyroid
Available in Target level: 1200 mg/day function,
liquid 1.0-1.5 Target level: BUN/creatine,
preparation mEq/L 0.6-1.2 mEq/L UA, electrolytes,
FBS; ECG;
encephalopathy
when combined
with haloperidol
*
Doses shown in table may exceed maximum recommended dose for some children.
BMI, Body mass index; BP, blood pressure; P, pulse; IM, intramuscular; GI, gastrointestinal;
CBC/diff, complete blood count with differential; BUN, blood urea nitrogen; UA, urinalysis; FBS,
fasting blood sugar; ECG, electrocardiogram.
Table 33.6
Adverse Effects for Select Antipsychotic Medications
Table 33.7
Metabolic Monitoring Parameters Based on ADA/APA
Consensus Guidelines
Bibliography
American Diabetes Association, American Psychiatric
Association, American Association of Clinical
Endocrinologists, North American Association for the Study
of Obesity. Consensus development conference on
antipsychotic drugs and obesity and diabetes. Diabetes Care .
2004;27:596–601.
American Academy of Child Adolescent Psychiatry. Practice
parameter for the use of atypical antipsychotic medications in
may be less pronounced in practice than in theory. The quality of the therapist–
patient alliance is consistently an important predictor of treatment outcome. A
positive working relationship, expecting change to occur, facing problems
assertively, increasing mastery, and attributing change to the participation in the
therapy have all been connected to effective therapy.
Table 34.1
Effective Psychotherapies for Specific Behavioral Health
Disorders
Behavior Therapy
Behavior therapy is based on both classic (Pavlovian) and operant (Skinnerian)
conditioning. Both approaches do not concern themselves with the inner motives
of the individual, but instead address the antecedent stimuli and consequent
responses. The treatment begins with a behavioral assessment with interview,
observation, diary, and rating scale components, along with a functional analysis
of the setting context, immediately preceding external events, and real-world
consequences of the behavior. A treatment plan is developed to modify the
maladaptive functions of the behavior, using tools such as positive and negative
reinforcement, social and tangible rewards, shaping, modeling, and prompting to
increase positive behavior, and extinction, stimulus control, punishment,
response cost, overcorrection, differential reinforcement of incompatible
behavior, graded exposure/systematic desensitization, flooding, modeling, and
role-playing to decrease negative behavior.
Behavior therapy has shown applicability to anxiety disorders, obsessive-
compulsive and related disorders, behavior disorders, A DHD, and autism
spectrum disorder.
Cognitive-Behavioral Therapy
Cognitive-behavioral therapy (CBT) is based on social and cognitive learning
theories and extends behavior therapy to address the influence of cognitive
processes on behavior. CBT is a problem-oriented treatment centered on
correcting problematic patterns in thinking and behavior that lead to emotional
difficulties and functional impairments. The CBT therapist seeks to identify and
change cognitive distortions (e.g., learned helplessness, irrational fears), identify
and avoid distressing situations, and identify and practice distress-reducing
behavior. Self-monitoring (daily thought records), self-instruction (brief
sentences asserting thoughts that are comforting and adaptive), and self-
reinforcement (rewarding oneself) are key tools used to facilitate achievement of
the CBT goals. Table 34.2 outlines the key descriptive features of CBT that can
be used by PCPs when describing CBT to patients and their family members.
Table 34.2
Core Components and Characteristics of
Cognitive-Behavioral Therapy
• One 60- to 90-minute session each week, typically for 6-12 weeks
• Symptom measures typically are collected frequently.
• Treatment is goal-oriented and collaborative with patient as an active
participant.
• Treatment is focused on changing current problematic thoughts or
behaviors.
• Weekly homework typically is assigned.
From Coffey SF, Banducci AN, Vinci C: Common questions about cognitive
behavior therapy for psychiatric disorders. Am Fam Physician 92:807–812,
2015.
Table 34.3
Practice Elements in Interventions for 3 Common Child and
Adolescent Psychiatric Disorders
Psychoeducation is the education of the parent and child about the cause,
course, prognosis, and treatment of the disorder. Problem solving is techniques,
discussions, or activities designed to bring about solutions to targeted problems,
with the intention of imparting a skill for how to approach and solve future
problems in a similar manner. Relaxation is techniques designed to create and
maintain the physiologic relaxation response. Self-monitoring is the repeated
measurement of a target metric by the child. Cognitive/coping skills consist of
techniques designed to alter interpretations of events through examination of the
child's reported thoughts, accompanied by exercises designed to test the validity
of the reported thoughts. PCPs can incorporate some of these elements into their
anticipatory guidance work with pediatric patients.
Table 34.4
Pediatric psychosomatic medicine deals with the relation between physical and
psychological factors in the causation or maintenance of disease states. The
process whereby distress is experienced and expressed in physical symptoms is
referred to as somatization or psychosomatic illness . Even though present in
virtually every psychiatric disorder, physical symptoms are most prominent in
the various somatic symptom and related disorders.
In the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition
(DSM-5), illnesses previously referred to as “somatoform disorders” are
classified as somatic symptom and related disorders (SSRDs). In children and
adolescents, the SSRDs include somatic symptom disorder (Table 35.1 ),
conversion disorder (Table 35.2 ), factitious disorders (Table 35.3 ), illness
anxiety disorder (Table 35.4 ), and other specified/unspecified somatic
symptom disorders (Table 35.5 ), as well as psychological factors affecting
other medical conditions (Table 35.6 ).
Table 35.1
DSM-5 Diagnostic Criteria for Somatic
Symptom Disorder
A. One or more somatic symptoms that are distressing or result in significant
disruption of daily life.
B. Excessive thoughts, feelings, or behaviors related to the somatic symptoms
or associated health concerns, as manifested by at least one of the
following:
1. Disproportionate and persistent thoughts about the seriousness of
one's symptoms.
2. Persistent high level of anxiety about health and symptoms.
3. Excessive time and energy devoted to these symptoms or health
concerns.
C. Although any one somatic symptom may not be continuously present, the
state of being symptomatic is persistent (typically >6 mo).
Specify if:
With predominant pain (previously known as “pain disorder” in
DSM IV-TR): for individuals whose somatic symptoms
predominantly involve pain.
Persistent: A persistent course is characterized by severe
symptoms, marked impairment, and long duration (>6 mo).
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p 311.
Table 35.2
DSM-5 Diagnostic Criteria for Conversion
Disorder or Functional Neurologic Symptom
Disorder
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p 324.
Table 35.4
DSM-5 Diagnostic Criteria for Illness Anxiety
Disorder
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p 315.
Table 35.5
DSM-5 Diagnostic Criteria for Other
Specified/Unspecified Somatic Symptom and
Related Disorders
Other Specified
Unspecified
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p 327.
Table 35.6
DSM-5 Diagnostic Criteria for Psychological
Factors Affecting Other Medical Conditions
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p 322.
With the exception of illness anxiety disorder, in which there is a high level of
anxiety about health in the absence of significant somatic symptoms, and
psychological factors affecting other medical conditions, in which psychological
and/or behavioral factors adversely affect a medical condition, SSRDs are
classified on the basis of physical symptoms associated with significant distress
and impairment, with or without the presence of a diagnosed medical condition.
The symptoms form a continuum that can range from pain to disabling
neurologic symptoms and generally interfere with school/home life and peer
relationships.
Most patients with SSRDs are seen by primary care practitioners or by
pediatric subspecialists, who may make specialty-specific diagnoses such as
visceral hyperalgesia, chronic fatigue syndrome, psychogenic syncope, or
noncardiac chest pain. Even within psychiatry, SSRDs are variously referred to
as functional or psychosomatic disorders or as medically unexplained
symptoms . The nosologic heterogeneity across the pediatric subspecialties
contributes to the varying diagnostic labels. There is a significant overlap in the
symptoms and presentation of patients with somatic symptoms who have
received different diagnoses from different specialties. Moreover, SSRDs share
similarities in etiology, pathophysiology, neurobiology, psychological
mechanisms, patient characteristics, and management and treatment response,
which is indicative of a single spectrum of somatic disorders.
It is helpful for healthcare providers to avoid the dichotomy of approaching
illness using a medical model in which diseases are considered physically or
motor and vocal tics at some point in the illness (although not necessarily
concurrently). The tic disorders are hierarchical in order (i.e., TD followed by
PTD followed by provisional tic disorder), such that once a tic disorder at one
level of the hierarchy is diagnosed, a lower-hierarchy diagnosis cannot be made.
Other specified/unspecified tic disorders are presentations in which symptoms
characteristic of a tic disorder that cause significant distress or impairment
predominate but do not meet the full criteria for a tic or other
neurodevelopmental disorder.
Table 37.1
DSM-5 Diagnostic Criteria for Tic Disorders
Note: A tic is a sudden, rapid, recurrent, nonrhythmic motor movement or
vocalization.
Tourette Disorder
A. Both multiple motor and one or more vocal tics have been present at some
time during the illness, although not necessarily concurrently.
B. The tics may wax and wane in frequency but have persisted for >1 yr since
first tic onset.
C. Onset is before age 18 yr.
D. The disturbance is not attributable to the physiologic effects of a substance
(e.g., cocaine) or another medical condition (e.g., Huntington disease,
postviral encephalitis).
A. Single or multiple motor or vocal tics have been present during the illness,
but not both motor and vocal.
B. The tics may wax and wane in frequency but have persisted for >1 yr since
first tic onset.
C. Onset is before age 18 yr.
D. The disturbance is not attributable to the physiologic effects of a substance
(e.g., cocaine) or another medical condition (e.g., Huntington disease,
postviral encephalitis).
E. Criteria have never been met for Tourette disorder.
Specify if:
With motor tics only
With vocal tics only
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, p. 81.
Description
Tics are sudden, rapid, recurrent, nonrhythmic motor movements or
vocalizations. Simple motor tics (e.g., eye blinking, neck jerking, shoulder
shrugging, extension of the extremities) are fast, brief movements involving one
or a few muscle groups. Complex motor tics involve sequentially and/or
simultaneously produced, relatively coordinated movements that can seem
purposeful (e.g., brushing back one's hair bangs, tapping the foot, imitating
someone else's movement [echopraxia ], or making a sexual or obscene gesture
[copropraxia ]). Simple vocal tics (e.g., throat clearing, sniffing, coughing) are
solitary, meaningless sounds and noises. Complex vocal tics involve
recognizable word or utterances (e.g., partial words [syllables], words out of
context, coprolalia [obscenities or slurs], palilalia [repeating one's own sounds
or words], or echolalia [repeating the last heard word or phrase]).
Sensory phenomena (premonitory urges) that precede and trigger the urge to
neuroacanthocytosis, Huntington syndrome, various frontal-subcortical brain
lesions), but it is rare for tics to be the only manifestation of these disorders.
Table 37.2
Sustain Compliance
From Carey WB, Crocker AC, Coleman WL, et al, editors: Developmental-
behavioral pediatrics, ed 4, Philadelphia, 2009, Elsevier/Saunders, p 639.
FIG. 38.1 Normative fears throughout childhood and adolescence. (From Craske MG,
Stein MB: Anxiety. Lancet 388:3048–3058, 2016.)
Table 38.1
Differential Diagnosis of Anxiety Disorders
Shyness
Substance use
Substance use withdrawal
Hyperthyroidism
Arrhythmias
Pheochromocytoma
Mast cell disorders
Carcinoid syndrome
Anaphylaxis
Hereditary angioedema
Lupus
Autoimmune encephalitis
Body dysmorphic disorder
Autism spectrum disorder
Major depressive disorder
Delusional disorder
Oppositional defiant disorder
Embarrassing medical condition
Preschoolers typically have specific fears related to the dark, animals, and
imaginary situations, in addition to normative separation anxiety. Preoccupation
with orderliness and routines (just right phenomena) often takes on a quality of
anxiety for preschool children. Parents' reassurance is usually sufficient to help
the child through this period. Although most school-age children abandon the
imaginary fears of early childhood, some replace them with fears of bodily harm
or other worries (Table 38.2 ). In adolescence, general worrying about school
performance and worrying about social competence are common and remit as
the teen matures.
Table 38.2
DSM-5 Diagnostic Criteria for Specific
Phobia
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 197–198.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 202–203.
Children and adolescents with SP often maintain the desire for involvement
with family and familiar peers. When severe, the anxiety can manifest as a panic
attack. SP is associated with a decreased quality of life, with increased likelihood
of having failed at least 1 grade, and a 38% likelihood of not graduating from
high school. Its onset is typically during or before adolescence and is more
common in girls. A family history of SP or extreme shyness is common.
Approximately 70–80% of patients with SP have at least 1 comorbid psychiatric
disorder. Most shy patients do not have SP.
Social effectiveness therapy for children (SET-C), alone or with SSRIs, is
considered the treatment of choice for SP (see Table 33.4 ). SSRI and SET-C are
superior to placebo in reducing social distress and behavioral avoidance and
increasing general functioning. SET-C may be better than SSRI in reducing these
symptoms. SET-C, but not SSRI, may be superior to placebo in improving social
skills, decreasing anxiety in specific social interactions, and enhancing social
or discomfort in which patients experience abrupt onset of physical and
psychological symptoms called panic attacks (Table 38.4 ). Physical symptoms
can include palpitations, sweating, shaking, shortness of breath, dizziness, chest
pain, and nausea. Children can present with acute respiratory distress but without
fever, wheezing, or stridor, ruling out organic causes of the distress. The
associated psychological symptoms include fear of death, impending doom, loss
of control, persistent concerns about having future attacks, and avoidance of
settings where attacks have occurred (agoraphobia, Table 38.5 ).
Table 38.4
DSM-5 Diagnostic Criteria for Panic Disorder
A. Recurrent unexpected panic attacks. A panic attack is an abrupt surge of
intense fear or intense discomfort that reaches a peak within minutes, and
during which time 4 (or more) of the following symptoms occur:
Note: The abrupt surge can occur from a calm state or an anxious state.
1. Palpitations, pounding heart, or accelerated heart rate.
2. Sweating.
3. Trembling or shaking.
4. Sensations of shortness of breath or smothering.
5. Feelings of choking.
6. Chest pain or discomfort.
7. Nausea or abdominal distress.
8. Feeling dizzy, unsteady, light-headed, or faint.
9. Chills or heart sensations.
10. Paresthesias (numbness or tingling sensations).
11. Derealizations (feeling or unreality) or depersonalization (being
detached from one-self).
12. Fear of losing control or “going crazy.”
13. Fear of dying.
Note: Culture-specific symptoms (e.g., tinnitus, neck soreness, headache,
uncontrollable screaming or crying) may be seen. Such symptoms should
not count as 1 of the 4 required symptoms.
B. At least 1 of the attacks has been followed by 1 mo (or more) of 1 or both
of the following:
1. Persistent concern or worry about additional panic attacks or their
consequences (e.g., losing control, having a heart attack, “going
crazy”).
2. A significant maladaptive change in behavior related to the
attacks (e.g., behaviors designed to avoid having panic attacks,
such as avoidance of exercise or unfamiliar situations).
C. The disturbance is not attributable to the physiologic effects of a substance
(e.g., a drug of abuse, a medication) or another medical condition (e.g.,
hyperthyroidism, cardiopulmonary disorders).
D. The disturbance is not better explained by another mental disorder (e.g.,
the panic attacks do not occur only in response to feared social situations,
as in social anxiety disorder; in response to circumscribed phobic objects or
situations, as in specific phobia; in response to obsessions, as in obsessive-
compulsive disorder; or in response to reminders of traumatic events, as in
posttraumatic stress disorder; or in response to separation from attachment
figures, as in separation anxiety disorder).
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 208–209.
Table 38.5
DSM-5 Diagnostic Criteria for Agoraphobia
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 217–218.
PD is uncommon before adolescence, with the peak age of onset at 15-19 yr,
occurring more often in girls. The postadolescence prevalence of PD is 1–2%.
Early-onset PD and adult-onset PD do not differ in symptom severity or social
functioning. Early-onset PD is associated with greater comorbidity, which can
result from greater familial loading for anxiety disorders in the early-onset
subtype. Children of parents with PD are much more likely to develop PD. A
predisposition to react to autonomic arousal with anxiety may be a specific risk
factor leading to PD. Twin studies suggest that 30–40% of the variance is
attributed to genetics. The increasing rates of panic attack are also directly
related to earlier sexual maturity. Cued panic attacks can be present in other
anxiety disorders and differ from the uncued “out-of-the-blue” attacks in PD.
No randomized controlled trials (RCTs) have evaluated the effectiveness of
antidepressant medication in youth with PD. Open-label studies with SSRIs
appear to show effectiveness in the treatment of adolescents (see Table 33.4 ).
CBT may also be helpful. The recovery rate is approximately 70%.
Generalized anxiety disorder occurs in children who often experience
unrealistic worries about different events or activities for at least 6 mo with at
least 1 somatic complaint (Table 38.6 ). The diffuse nature of the anxiety
symptoms differentiates it from other anxiety disorders. Worries in children with
GAD usually center around concerns about competence and performance in
school and athletics. GAD often manifests with somatic symptoms, including
restlessness, fatigue, problems concentrating, irritability, muscle tension, and
sleep disturbance. Given the somatic symptoms characteristic of GAD, the
differential diagnosis must consider other medical causes. Excessive use of
caffeine or other stimulants in adolescence is common and should be determined
with a careful history. When the history or physical examination is suggestive,
the pediatrician should rule out hyperthyroidism, hypoglycemia, lupus,
pheochromocytoma, and other disorders (see Table 38.1 ; Fig. 38.2 ).
Table 38.6
DSM-5 Diagnostic Criteria for Generalized
Anxiety Disorder
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 222.
FIG. 38.2 Evaluation of worry, fear, and panic. PANDAS , Pediatric autoimmune
neuropsychiatric disorders associated with Streptococcus pyogenes . (From Kliegman
RM, Lye PS, Bordini B, et al, editors: Nelson pediatric symptom-based diagnosis,
Philadelphia, 2018, Elsevier, p 429).
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 237.
Note: The following criteria apply to adults, adolescents, and children older
than 6 yr. For children 6 yr and younger, see corresponding criteria below.
A. Exposure to actual or threatened death, serious injury, or sexual violence in
1 (or more) of the following ways:
1. Directly experiencing the traumatic event(s).
2. Witnessing, in person, the event(s) as it occurred to others.
3. Learning that the traumatic event(s) occurred to a close family
member or close friend. In cases of actual or threatened death of a
family member or friend, the event(s) must have been violent or
accidental.
4. Experiencing repeated or extreme exposure to aversive details of
the traumatic event(s) (e.g., 1st responders collecting human
remains; police officers repeatedly exposed to details of child
abuse).
Note: Criterion A4 does not apply to exposure through electronic media,
television, movies, or pictures, unless this exposure is work related.
B. Presence of 1 (or more) of the following intrusion symptoms associated
with the traumatic event(s), beginning after the traumatic event(s) occurred:
1. Recurrent, involuntary, and intrusive distressing memories of the
traumatic event(s).
Note: In children older than 6 yr, repetitive play may occur in which themes
or aspects of the traumatic event(s) are expressed.
2. Recurrent distressing dreams in which the content and/or effect of
the dream are related to the traumatic event(s).
Note: In children, there may be frightening dreams without recognizable
content.
3. Dissociative reactions (e.g., flashbacks) in which the individual
feels or acts as if the traumatic event(s) were recurring. (Such
reactions may occur on a continuum, with the more extreme
expression being a complete loss or awareness of present
surroundings.)
Note: In children, trauma-specific reenactment may occur in play.
4. Intense or prolonged psychological distress at exposure to internal
or external cues that symbolize or resemble an aspect of the
traumatic event(s).
5. Marked physiologic reactions to internal or external cues that
symbolize or resemble an aspect of the traumatic event(s).
C. Persistent avoidance of stimuli associated with the traumatic event(s),
beginning after the traumatic event(s) occurred, as evidenced by 1 or both
of the following:
1. Avoidance of or efforts to avoid distressing memories, thoughts,
or feelings about or closely associated with the traumatic
event(s).
2. Avoidance of or efforts to avoid external reminders (people,
places, conversations, activities, objects, situations) that arouse
distressing memories, thoughts, or feelings about or closely
associated with the traumatic event(s).
D. Negative alterations in cognitions and mood associated with the traumatic
event(s), beginning or worsening after the traumatic event(s) occurred, as
evidenced by 2 (or more) of the following:
1. Inability to remember an important aspect of the traumatic
event(s) (typically due to dissociative amnesia and not to other
factors such as head injury, alcohol, or drugs).
2. Persistent and exaggerated negative beliefs or expectations about
oneself, others, or the world (e.g., “I am bad,” “No one can be
trusted,” “The world is completely dangerous,” “My whole
nervous system is permanently ruined”).
3. Persistent, distorted cognitions about the cause or consequences
of the traumatic event(s) that lead the individual to blame
himself/herself or others.
4. Persistent negative emotional state (e.g., fear, horror, anger, guilt,
or shame).
5. Markedly diminished interest or participation in significant
activities.
6. Feelings of detachment or estrangement from others.
7. Persistent inability to experience positive emotions (e.g., inability
to experience happiness, satisfaction, or loving feelings).
E. Marked alterations in arousal and reactivity associated with the traumatic
event(s), beginning or worsening after the traumatic event(s) occurred, as
evidenced by 2 (or more) of the following:
1. Irritable behavior and angry outbursts (with little or no
provocation) typically expressed by verbal or physical aggression
toward people or objects.
2. Reckless or self-destructive behavior.
3. Hypervigilance.
4. Exaggerated startle response.
5. Problems with concentration.
6. Sleep disturbance (e.g., difficulty falling or staying asleep or
restless sleep).
F. Duration of the disturbance (Criteria B, C, D, and E) is more than 1 mo.
G. The disturbance causes clinically significant distress or impairment in
social, occupational, or other important areas of functioning.
H. The disturbance is not attributable to the physiologic effects of a substance
(e.g., medication, alcohol) or another medical condition.
Specify whether:
With dissociative symptoms: The individual's symptoms meet the criteria
for posttraumatic stress disorder, and in addition, in response to the stressor,
the individual experiences persistent or recurrent symptoms of either of the
following:
1. Depersonalization: Persistent or recurrent experiences of feeling
detached from, and as if one were an outside observer of, one's
mental processes or body (e.g., feeling as though one were in a
dream; feeling a sense of unreality of self or body or of time
moving slowly).
2. Derealization: Persistent or recurrent experiences of unreality of
surroundings (e.g., the world around the individual is experienced
as unreal, dreamlike, distant, or distorted).
Note: To use this subtype, the dissociative symptoms must not be attributable
to the physiologic effects of a substance (e.g., blackouts, behavior during
alcohol intoxication) or another medical condition (e.g., complex partial
seizures).
Specify if:
With delayed expression: If the full diagnostic criteria are not met until at
least 6 mo after the event (although the onset and expression of some
symptoms may be immediate).
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 271–274.
Events that pose actual or threatened physical injury, harm, or death to the
child, child's caregiver, or others close to the child, and that produce
considerable stress, fear, or helplessness, are required to make the diagnosis of
PTSD. Three clusters of symptoms are also essential for diagnosis:
reexperiencing, avoidance, and hyperarousal. Persistent reexperiencing of the
stressor through intrusive recollections, nightmares, and reenactment in play are
typical responses in children. Persistent avoidance of reminders and numbing of
emotional responsiveness, such as isolation, amnesia, and avoidance, constitute
the 2nd cluster of behaviors. Symptoms of hyperarousal, such as
hypervigilance, poor concentration, extreme startle responses, agitation, and
sleep problems, complete the symptom profile of PTSD. Occasionally, children
regress in some of their developmental milestones after a traumatic event.
Avoidance symptoms are usually observable in younger children, whereas older
children may better describe reexperiencing and hyperarousal symptoms.
Repetitive play involving the event, psychosomatic symptoms, and nightmares
may also be observed.
Initial interventions after a trauma should focus on reunification with a parent
and attending to the child's physical needs in a safe place. Aggressive treatment
of pain, and facilitating a return to comforting routines, including regular sleep,
is indicated. Long-term treatment may include individual, group, school-based,
or family therapy, as well as pharmacotherapy, in selected cases. Individual
treatment involves transforming the child's concept of himself or herself as
victim to that of survivor and can occur through play therapy, psychodynamic
therapy, or CBT. Group work is also helpful for identifying which children might
need more intensive assistance. Goals of family work include helping the child
establish a sense of security, validating the child's emotions, and anticipating
situations when the child will need more support from the family.
FIG. 39.1 Evaluation of mood disorders. (From Kliegman RM, Lye PS,
Bordini BJ, et al, editors: Nelson pediatric symptom-based diagnosis,
Philadelphia, 2018, Elsevier, p 426.)
Description
Major depressive disorder (MDD) is characterized by a distinct period of at
least 2 wk (an episode ) in which there is a depressed or irritable mood and/or
loss of interest or pleasure in almost all activities that is present for most of the
day, nearly every day (Table 39.1 ). Major depression is associated with
characteristic vegetative and cognitive symptoms, including disturbances in
appetite, sleep, energy, and activity level; impaired concentration; thoughts of
worthlessness or guilt; and suicidal thoughts or actions. Major depression is
considered mild if few or no symptoms in excess of those required to make the
diagnosis are present, and the symptoms are mildly distressing and manageable
and result in minor functional impairment. Major depression is considered severe
if symptoms substantially in excess of those required to make the diagnosis are
present, and the symptoms are highly distressing and unmanageable and
markedly impair function. Moderate major depression is intermediate in severity
between mild and severe.
Table 39.1
DSM-5 Diagnostic Criteria for Major
Depressive Episode
A. Five (or more) of the following symptoms have been present during the
same 2 wk period and represent a change from previous functioning; at
least 1 of the symptoms is either (1) depressed mood or (2) loss of interest
or pleasure.
1. Depressed most of the day, nearly every day, as indicated by
either subjective report (e.g., feels sad, empty, hopeless) or
observation made by others (e.g., appears tearful).
Note: In children and adolescents, can be irritable mood.
2. Markedly diminished interest or pleasure in all, or almost all,
activities most of the day, nearly every day (as indicated by either
subjective account or observation).
3. Significant weight loss when not dieting or weight gain (e.g., a
change of more than 5% of body weight in a month), or decrease
or increase in appetite nearly every day.
Note: In children, consider failure to make expected weight gain.
4. Insomnia or hypersomnia nearly every day.
5. Psychomotor agitation or retardation nearly every day
(observable by others, not merely subjective feelings of
restlessness or being slowed down).
6. Fatigue or loss of energy nearly every day.
7. Feelings of worthlessness or excessive or inappropriate guilt
(which may be delusional) nearly every day (not merely self-
reproach or guilt about being sick).
8. Diminished ability to think or concentrate, or indecisiveness,
nearly every day (either by subjective account or as observed by
others).
9. Recurrent thoughts of death (not just fear of dying), recurrent
suicidal ideation without a specific plan, or a suicide attempt or a
specific plan for committing suicide.
B. The symptoms cause clinically significant distress or impairment in social,
occupational, or other important areas of functioning.
C. The episode is not attributable to the physiologic effects of a substance or
to another medical condition.
Note: Criteria A-C represent a major depressive episode.
D. The occurrence of the major depressive episode is not better explained by
schizoaffective disorder, schizophrenia, schizophreniform disorder,
delusional disorder, or other specified and unspecified schizophrenia
spectrum and other psychotic disorders.
E. There has never been a manic episode or a hypomanic episode.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 125–126.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 168–169.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p. 156.
Epidemiology
The overall prevalence of parent-reported diagnosis of depressive disorder in the
United States (excluding DMDD) among 3-17 yr old children is approximately
2.1% (current) and 3.9% (ever); the prevalence rate increases to 12.8% (lifetime)
for 12-17 yr olds. The male:female ratio (excluding DMDD) is approximately 1 :
1 during childhood and beginning in early adolescence rises to 1 : 1.5-3.0 in
adulthood.
Based on rates of chronic and severe persistent irritability, which is the core
feature of DMDD, the overall 6 mo to 1 yr prevalence has been estimated in the
2–5% range. In 3 community samples, the 3 mo prevalence rate of DMDD
ranged from 0.8–3.3%, with the highest rates occurring in preschoolers
(although DSM-5 does not permit this diagnosis until age 6 yr). Approximately
pediatric practitioner can begin to assess the onset, duration, context, and
severity of the symptoms and associated dangerousness, distress, and functional
impairment. In the absence of acute dangerousness (e.g., suicidality, psychosis,
substance abuse) and significant distress or functional impairment, the pediatric
practitioner (or co-located behavioral health therapist) can schedule a follow-up
appointment within 1-2 wk to conduct a depression assessment. At this follow-
up visit, to assist with decision-making about appropriate level of care, a
depression-specific screening standardized rating scale can be administered to
assess symptom severity (Table 39.4 ), and additional risk factors can be
explored (see Etiology and Risk Factors earlier).
Table 39.4
Depression-Specific Rating Scales
NUMBER OF
NAME OF INSTRUMENT INFORMANT(S) AGE RANGE
ITEMS
Beck Depression Inventory Youth 13+ yr 21
Beck Depression Inventory for Youth Youth 7-14 yr 20
Center for Epidemiologic Studies-Depression- Youth 6-18 yr 20
Children
Children's Depression Rating Scale-Revised Youth, Parent, 6-18 yr 47
Clinician
Children's Depression Inventory, Second Edition Youth, Parent, 7-17 yr 28/17/12
Teacher
Depression Self-Rating Scale Youth 7-13 yr 18
Mood and Feelings Questionnaire Youth, Parent 7-18 yr 33-34
Patient Health Questionnaire-9 Youth 12/13+ yr 9
Preschool Feelings Checklist Parent 3-5.6 yr 20
PROMIS Emotional Distress-Depressive Youth, Parent Youth: 8-17 8/6
Symptoms yr
Parent: 5-17
yr
Reynolds Child Depression Scale Youth 8-13 yr 30
Reynolds Adolescent Depression Scale, Second Youth 11-20 yr 30
Edition
Description
The bipolar and related disorders include bipolar I, bipolar II, cyclothymic, and
other specified/unspecified bipolar and related disorders, as well as bipolar and
related disorder caused by another medical condition.
A manic episode is characterized by a distinct period of at least 1 wk in which
there is an abnormally and persistently elevated, expansive, or irritable mood
and abnormally and persistently increased goal-directed activity or energy that is
present for most of the day, nearly every day (or any duration if hospitalization is
necessary). The episode is associated with characteristic cognitive and
behavioral symptoms, including disturbances in self-regard, speech, attention,
thought, activity, impulsivity, and sleep (Table 39.5 ). To diagnose bipolar I
disorder , criteria must be met for at least 1 manic episode, and the episode must
not be better explained by a psychotic disorder. The manic episode may have
been preceded and may be followed by hypomanic or major depressive episodes.
Bipolar I disorder is rated as mild, moderate, or severe in the same way as the
depressive disorders (see Description section of Chapter 39.1 ).
Table 39.5
DSM-5 Diagnostic Criteria for a Manic
Episode
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 124.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 124.
1. Have you wished you were dead or wished you could go to sleep and not
wake up?
2. Have you actually had any thoughts about killing yourself?
If “Yes” to 2, answer questions 3, 4, 5, and 6.
If “No” to 2, go directly to question 6.
3. Have you thought about how you might do this?
4. Have you had any intention of acting on these thoughts of killing yourself,
as opposed to you having the thoughts but you definitely would not act
on them?
5. Have you started to work out or worked out the details of how to kill
yourself? Do you intend to carry out this plan?
6. Have you done anything, started to do anything, or prepared to do
anything to end your life?
Screening for suicide in schools is also fraught with problems related to low
specificity of the screening instrument and paucity of referral sites, as well as
poor acceptability among school administrators. Gatekeeper (e.g., student
support personnel) training appears effective in improving skills among school
personnel and is highly acceptable to administrators but has not been shown to
prevent suicide. School curricula (e.g., Signs of Suicide ) have shown some
preventive potential by teaching students to recognize the signs of depression
and suicide in themselves and others and providing them with specific action
steps necessary for responding to these signs. Peer helpers have not generally
been shown to be efficacious.
Bibliography
Barrocas AL, Hankin BL, Young JF, et al. Rates of nonsuicidal
self-injury in youth: age, sex, and behavioral methods in a
community sample. Pediatrics . 2012;130:39–45.
Björkenstam C, Kosidou K, Björkenstam E. Childhood
adversity and risk of suicide: cohort study of 548 721
adolescents and young adults in Sweden. BMJ .
2017;357:j1334.
Bohanna I, Wang X. Media guidelines for the responsible
CHAPTER 41
Eating Disorders
Richard E. Kreipe, Taylor B. Starr
Definitions
Anorexia nervosa (AN) involves significant overestimation of body size and
shape, with a relentless pursuit of thinness that, in the restrictive subtype,
typically combines excessive dieting and compulsive exercising. In the binge-
purge subtype, patients might intermittently overeat and then attempt to rid
themselves of calories by vomiting or taking laxatives, still with a strong drive
for thinness (Table 41.1 ).
Table 41.1
DSM-5 Diagnostic Criteria for Anorexia
Nervosa
Specify whether:
Restricting type (ICD-10-CM code F50.01): During the last 3
mo, the individual has not engaged in recurrent episodes of
binge eating or purging behavior (i.e., self-induced vomiting or
the misuse of laxatives, diuretics, or enemas). This subtype
describes presentations in which weight loss is accomplished
primarily through dieting, fasting, and/or excessive exercise.
Binge-eating/purging type (ICD-10-CM code F50.02): During
the last 3 mo, the individual has engaged in recurrent episodes
of binge eating or purging behavior (i.e., self-induced vomiting
or the misuse of laxatives, diuretics, or enemas).
Specify if:
In partial remission : After full criteria for anorexia nervosa
were previously met, Criterion A (low body weight) has not
been met for a sustained period, but either Criterion B (intense
fear of gaining weight or becoming fat or behavior that
interferes with weight gain) or Criterion C (disturbances in self-
perception of weight and shape) is still met.
In full remission : After full criteria for anorexia nervosa were
previously met, none of the criteria has been met for a sustained
period of time.
Specify current severity:
The minimum level of severity is based, for adults, on current
body mass index (BMI) (see below) or, for children and
adolescents, on BMI percentile. The ranges below are derived
from World Health Organization categories for thinness in
adults; for children and adolescents, corresponding BMI
percentiles should be used. The level of severity may be
increased to reflect clinical symptoms, the degree of functional
disability, and the need for supervision.
Mild : BMI ≥ 17 kg/m2
Moderate : BMI 16-16.99 kg/m2
Severe : BMI 15-15.99 kg/m2
Extreme : BMI < 15 kg/m2
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 338–339.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 345.
Children and adolescents with EDs may not fulfill criteria for AN or BN in the
Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5)
and may fall into a subcategory of atypical anorexia nervosa, or a more
appropriately defined category of avoidant/ restrictive food intake disorder
(ARFID). In these conditions, food intake is restricted or avoided because of
adverse feeding or eating experiences or the sensory qualities of food, resulting
in significant unintended weight loss or nutritional deficiencies and problems
with social interactions (Table 41.3 ).
Table 41.3
DSM-5 Diagnostic Criteria for
Avoidant/Restrictive Food Intake Disorder
A. An eating or feeding disturbance (e.g., apparent lack of interest in eating or
food; avoidance based on the sensory characteristics of food; concern about
aversive consequences of eating) as manifested by persistent failure to meet
appropriate nutritional and/or energy needs associated with one (or more)
of the following:
1. Significant weight loss (or failure to achieve expected weight
gain or faltering growth in children).
2. Significant nutritional deficiency.
3. Dependence on enteral feeding or oral nutritional supplements.
4. Marked interference with psychosocial functioning.
B. The disturbance is not better explained by lack of available food or by an
associated culturally sanctioned practice.
C. The eating disturbance does not occur exclusively during the course of
anorexia nervosa or bulimia nervosa, and there is no evidence of a
disturbance in the way in which one's body weight or shape is experienced.
D. The eating disturbance is not attributable to a concurrent medical condition
or not better explained by another mental disorder. When the eating
disturbance occurs in the context of another condition or disorder, the
severity of the eating disturbance exceeds that routinely associated with the
condition or disorder and warrants additional clinical attention.
Specify if:
In remission : After full criteria for avoidant/restrictive food
intake disorder were previously met, the criteria have not been
met for a sustained period of time.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 334.
Table 41.4
Eating and Weight Control Habits Commonly Found in
Children and Adolescents With an Eating Disorder (ED)
Table 41.5
Symptoms Commonly Reported by Patients With an Eating
Disorder (ED)
Abdomen Early fullness and discomfort Discomfort after Weight loss is associated
with eating a binge with reduced volume and
Constipation Cramps and tone of GI tract musculature,
Perceives contour as “fat,” often diarrhea with especially the stomach
preferring well-defined laxative abuse Laxatives may be used to
abdominal musculature relieve constipation or as a
cathartic
Symptom reduction with
healthy eating can take
weeks to occur
Extremities and Cold, blue hands and feet No Energy-conserving low body
musculoskeletal characteristic temperature with slow blood
symptoms flow most notable
Self-cutting or peripherally
burning on Quickly reversed with
wrists or arms healthy eating
Nervous system No characteristic symptom No characteristic Neurologic symptoms suggest
symptom diagnosis other than ED
Mental status Depression, anxiety, obsessive- Depression; PTSD; Underlying mood
compulsive symptoms, alone or in borderline disturbances can worsen with
combination personality disorder dysfunctional weight control
traits practices and can improve
with healthy eating
AN patients might report
emotional “numbness” with
starvation preferable to
emotionality associated with
healthy eating
AN, Anorexia nervosa; BN, bulimia nervosa; ED, eating disorder; GI, gastrointestinal; PTSD,
posttraumatic stress disorder.
Table 41.6
Signs Commonly Found in Patients With Eating Disorder
(ED) Relative to Prominent Feature of Weight Control
PROMINENT FEATURE
PHYSICAL CLINICAL COMMENTS
SIGN Binge RELATED TO ED SIGNS
Restrictive Intake
Eating/Purging
General Thin to cachectic, depending on Thin to overweight, Examine in hospital gown
appearance balance of intake and output depending on the Weight loss more rapid with
Might wear bulky clothing to balance of intake reduced intake and excessive
hide thinness and might resist and output through exercise
being examined various means Binge eating can result in
large weight gain, regardless
of purging behavior
Appearance depends on
balance of intake and output
and overall weight control
habits
Weight Low and falling (if previously Highly Weigh in hospital gown with
overweight, may be normal or high); variable, no underwear, after voiding
may be falsely elevated if patient depending on (measure urine SG)
drinks fluids or adds weights to body balance of Remain in gown until physical
before being weighed intake and exam completed to identify
output and state possible fluid loading (low
of hydration urine SG, palpable bladder) or
Falsification of adding weights to body
weight is
unusual
Metabolism Hypothermia: temp <35.5°C Variable, but Hypometabolism related to
(95.9°F), pulse <60 beats/min hypometabolic state disruption of hypothalamic
Slowed psychomotor response is less common than control mechanisms as a result
with very low core temperature in AN of weight loss
Signs of hypometabolism
(cold skin, slow capillary
refill, acrocyanosis) most
evident in hands and feet,
where energy conservation is
most active
Skin Dry Calluses over Carotenemia with large intake
Increased prominence of hair proximal knuckle of β-carotene foods
follicles joints of hand Russell's sign: maxillary
Orange or yellow hands (Russell's sign) incisors abrasion develops
into callus with chronic digital
pharyngeal stimulation,
usually on dominant hand
Hair Lanugo-type hair growth on face No characteristic Body hair growth conserves
and upper body sign energy
Scalp hair loss, especially Scalp hair loss “telogen
prominent in parietal region effluvium” can worsen weeks
after refeeding begins, as hair
in resting phase is replaced by
growing hair
Eyes No characteristic sign Subconjunctival Increased intrathoracic pressure
hemorrhage during vomiting
Teeth No characteristic sign Eroded dental Perimolysis, worse on lingual
enamel and surfaces of maxillary teeth, is
decayed, fractured, intensified by brushing teeth
missing teeth without preceding water rinse
Salivary glands No characteristic sign Enlargement, Parotid > submandibular
relatively nontender involvement with frequent and
chronic binge eating and induced
vomiting
Throat No characteristic sign Absent gag reflex Extinction of gag response with
repeated pharyngeal stimulation
Heart Bradycardia, hypotension, and Hypovolemia if Changes in AN resulting from
orthostatic pulse differential >25 dehydrated central hypothalamic and
beats/min intrinsic cardiac function
Orthostatic changes less
prominent if athletic, more
prominent if associated with
purging
Abdomen Scaphoid, organs may be palpable Increased bowel Presence of organomegaly
but not enlarged, stool-filled left sounds if recent requires investigation to
lower quadrant laxative use determine cause
Constipation prominent with
weight loss
Extremities and Cold, acrocyanosis, slow No characteristic Signs of hypometabolism
musculoskeletal capillary refill sign, but may have (cold) and cardiovascular
system Edema of feet rebound edema after dysfunction (slow capillary
Loss of muscle, subcutaneous, stopping chronic refill and acrocyanosis) in
and fat tissue laxative use hands and feet
Edema, caused by capillary
fragility more than
hypoproteinemia in AN, can
worsen in early phase of
refeeding
Nervous system No characteristic sign No characteristic Water loading before weigh-ins
sign can cause acute hyponatremia
Mental status Anxiety about body image, Depression, Mental status often improves with
irritability, depressed mood, evidence of PTSD, healthier eating and weight; SSRIs
oppositional to change more likely suicidal only shown to be effective for BN
than AN
AN, Anorexia nervosa; BN, bulimia nervosa; PTSD, posttraumatic stress disorder; SG, specific
gravity; SSRIs, selective serotonin reuptake inhibitors.
Differential Diagnosis
In addition to identifying symptoms and signs that deserve targeted intervention
for patients who have an ED, a comprehensive history and physical examination
are required to rule out other conditions in the differential diagnosis. Weight loss
can occur in any condition with increased catabolism (e.g., hyperthyroidism,
malignancy, occult chronic infection) or malabsorption (e.g., inflammatory
bowel disease, celiac disease) or in other disorders (Addison disease, type 1
diabetes mellitus, stimulant abuse), but these illnesses are generally associated
with other findings and are not usually associated with decreased caloric intake.
Patients with inflammatory bowel disease can reduce intake to minimize
abdominal cramping; eating can cause abdominal discomfort and early satiety in
AN because of gastric atony associated with significant weight loss, not
malabsorption. Likewise, signs of weight loss in AN might include hypothermia,
acrocyanosis with slow capillary refill, and neutropenia similar to some features
of sepsis, but the overall picture in EDs is one of relative cardiovascular stability
compared with sepsis. Endocrinopathies are also in the differential of EDs.
With BN, voracious appetite in the face of weight loss might suggest diabetes
mellitus, but blood glucose levels are normal or low in EDs. Adrenal
insufficiency mimics many physical symptoms and signs found in restrictive AN
but is associated with elevated potassium levels and hyperpigmentation. Thyroid
disorders may be considered, because of changes in weight, but the overall
presentation of AN includes symptoms of both underactive and overactive
limiting foods that might trigger a binge.
When initiating treatment of an ED in a primary care setting, the clinician
should be aware of common cognitive patterns. Patients with AN typically have
all-or-none thinking (related to perfectionism) with a tendency to overgeneralize
and jump to catastrophic conclusions, while assuming that their body is
governed by rules that do not apply to others. These tendencies lead to the
dichotomization of foods into good or bad categories, having a day ruined
because of one unexpected event, or choosing foods based on rigid self-imposed
restrictions. These thoughts may be related to neurocircuitry and
neurotransmitter abnormalities associated with executive function and rewards.
Weight loss in the absence of body shape, size, or weight concerns should raise
suspicion about ARFID, because the emotional distress associated with “forced”
eating is not associated with gaining weight, but with the neurosensory
experience of eating.
A standard nutritional balance of 15–20% calories from protein, 50–55% from
carbohydrate, and 25–30% from fat is appropriate. The fat content may need to
be lowered to 15–20% early in the treatment of AN because of continued fat
phobia. With the risk of low BMD in patients with AN, calcium and vitamin D
supplements are often needed to attain the recommended 1,300 mg/day intake of
calcium. Refeeding can be accomplished with frequent small meals and snacks
consisting of a variety of foods and beverages (with minimal diet or fat-free
products), rather than fewer high-volume high-calorie meals. Some patients find
it easier to take in part of the additional nutrition as canned supplements
(medicine) rather than food. Regardless of the source of energy intake, the risk
for refeeding syndrome (see Complications earlier) increases with the degree of
weight loss and the rapidity of caloric increases. Therefore, if the weight has
fallen below 80% of expected weight for height, refeeding should proceed
carefully (not necessarily slowly) and possibly in the hospital (Table 41.7 ).
Table 41.7
Potential Indications for Inpatient Medical
Hospitalization of Patients With Anorexia
Nervosa
Physical and Laboratory
Psychiatric
Miscellaneous
Patients with AN tend to have a highly structured day with restrictive intake,
in contrast to BN, which is characterized by a lack of structure, resulting in
chaotic eating patterns and binge-purge episodes. All patients with AN, BN, or
ED-NOS benefit from a daily structure for healthy eating that includes 3 meals
and at least 1 snack a day, distributed evenly over the day, based on balanced
meal planning. Breakfast deserves special emphasis because it is often the first
meal eliminated in AN and is often avoided the morning after a binge-purge
episode in BN. In addition to structuring meals and snacks, patients should plan
structure in their activities. Although overexercising is common in AN,
completely prohibiting exercise can lead to further restriction of intake or to
surreptitious exercise; inactivity should be limited to situations in which weight
loss is dramatic or there is physiologic instability. Also, healthy exercise (once a
CHAPTER 42
Description
Oppositional defiant disorder (ODD) is characterized by a pattern lasting at
least 6 mo of angry, irritable mood, argumentative/defiant behavior, or
vindictiveness exhibited during interaction with at least 1 individual who is not a
sibling (Table 42.1 ). For preschool children, the behavior must occur on most
days, whereas in school-age children, the behavior must occur at least once a
week. The severity of the disorder is considered mild if symptoms are confined
to only 1 setting (e.g., at home, at school, at work, with peers), moderate if
symptoms are present in at least 2 settings, and severe if symptoms are present in
≥4 settings.
Table 42.1
DSM-5 Diagnostic Criteria for Oppositional
Defiant Disorder
A. A pattern of angry/irritable mood, argumentative/defiant behavior, or
vindictiveness lasting at least 6 mo as evidenced by at least 4 symptoms
from any of the following categories, and exhibited during interaction with
at least 1 individual who is not a sibling:
Angry/Irritable Mood
1. Often loses temper.
2. Is often touchy or easily annoyed.
3. Is often angry and resentful.
Argumentative/Defiant Behavior
4. Often argues with authority figures or, for children
and adolescents, with adults.
5. Often actively defies or refuses to comply with
requests from authority figures or with rules.
6. Often deliberately annoys others.
7. Often blames others for his or her mistakes or
misbehavior.
Vindictiveness
8. Has been spiteful or vindictive at least twice within
the past 6 mo.
Note: The persistence and frequency of these
behaviors should be used to distinguish a behavior
that is within normal limits from a behavior that is
symptomatic. For children younger than 5 yr, the
behavior should occur on most days for a period of at
least 6 mo unless otherwise noted (Criterion A8). For
individuals 5 yr or older, the behavior should occur at
least once per week for at least 6 mo, unless
otherwise noted (Criterion A8). While these
frequency criteria provide guidance on a minimal
level of frequency to define symptoms, other factors
should be considered, such as whether the frequency
and intensity of the behaviors are outside a range that
is normative for the individual's developmental level,
gender, and culture.
B. The disturbance in behavior is associated with distress in the individual or
others in his or her immediate social context (e.g., family, peer group, work
colleagues), or it impacts negatively on social, educational, occupational, or
other important areas of functioning.
C. The behaviors do not occur exclusively during the course of a psychotic,
substance use, depressive, or bipolar disorder. Also, the criteria are not met
for disruptive mood dysregulation disorder.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 462–463.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 466.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 469–471.
Epidemiology
The prevalence of ODD is approximately 3%, and in preadolescents is more
common in males than females (1.4 : 1). One-year prevalence rates for IED and
CD approximate 3% and 5%, respectively. For CD, prevalence rates rise from
childhood to adolescence and are higher among males than females. The
prevalence of these disorders has been shown to be higher in lower
socioeconomic classes. This class of disorders constitutes the most frequent
referral problem for youth, accounting for one third to one half of all cases seen
in mental health clinics. Racial/ethnic minority youth with these disorders utilize
specialty mental health services at lower rates than their white peers.
Clinical Course
Oppositional behavior can occur in all children and adolescents at times,
particularly during the toddler and early teenage periods when establishing
autonomy and independence are normative developmental tasks. Oppositional
Table 42.4
Anger/Aggression-Specific Screening Instruments
For mild symptoms (manageable by the parent and not functionally impairing)
and in the absence of major risk factors (homicidality, assaultiveness, psychosis,
substance use, child maltreatment, parental psychopathology, or severe family
dysfunction), guided self-help (anticipatory guidance) with watchful waiting
and scheduled follow-up may suffice. Guided self-help can include provision of
educational materials (pamphlets, books, videos, workbooks, internet sites) that
provide information to the youth about dealing with anger-provoking situations,
and advice to parents about strengthening the parent–child relationship, effective
parenting strategies, and the effects of adverse environmental exposures on the
development of behavior problems. In a Cochrane review, media-based
parenting interventions had a moderate positive effect on child behavior
problems, either alone or as an adjunct to medication. An example of a self-help
program for parents is the Positive Parenting Program (Triple P;
www.triplep.net ), online version, in which parents can purchase 4 modules of
instruction addressing techniques for positive parenting and strategies for
encouraging good behavior, teaching new emotional and behavioral skills, and
managing misbehavior (see Chapter 19 ).
If the problematic behavior is occurring predominantly at school, the parent
can be advised about the role of a special education evaluation in the assessment
and management of the child's misbehavior, including the development of a
behavioral intervention plan to prevent disciplinary actions that is formalized in
an individualized educational plan (IEP) or 504 plan.
If a mental health clinician has been co-located or integrated into the primary
care setting, all parents of young children (universal prevention), as well as the
parents of youth with mild behavior problems (indicated prevention), can be
provided with a brief version of parent training . Programs targeted at toddlers
through 12 yr olds have been found to be effective in improving parenting skills,
parental mental health, and child emotional and behavior problems. For
example, Incredible Years (http://www.incredibleyears.com ) has a 6-8 session
Description
The schizophrenia spectrum and other psychotic disorders are primarily
characterized by the active (or positive) symptoms of psychosis, specifically
delusions, hallucinations, disorganized speech, or grossly disorganized or
catatonic behavior. Brief psychotic disorder is characterized by the duration of
1 or more of these symptoms for at least 1 day but <1 mo followed by complete
resolution. Emergence of symptoms may or may not be preceded by an
identifiable stressor (Table 47.1 ). Although brief, the level of impairment in this
disorder may be severe enough that supervision is required to ensure that basic
needs are met and the individual is protected from the consequences of poor
judgment and cognitive impairment.
Table 47.1
DSM-5 Diagnostic Criteria for Brief Psychotic
Disorder
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, p 94.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 96–97.
Table 47.3
DSM-5 Diagnostic Criteria for Schizophrenia
A. Two (or more) of the following, each present for a significant portion of
time during a 1 mo period (or less if successfully treated). At least 1 of
these must be (1), (2), or (3):
1. Delusions.
2. Hallucinations.
3. Disorganized speech (e.g., frequent derailment or incoherence).
4. Grossly disorganized or catatonic behavior.
5. Negative symptoms (i.e., diminished emotional expression or
avolition).
B. For a significant portion of the time since the onset of the disturbance,
level of functioning in 1 or more major areas, such as work, interpersonal
relations, or self-care, is markedly below the level achieved prior to the
onset (or when the onset is in childhood or adolescence, there is failure to
achieve expected level of interpersonal, academic, or occupational
functioning).
C. Continuous signs of the disturbance persist for at least 6 mo. This 6 mo
period must include at least 1 mo of symptoms (or less if successfully
treated) that meet Criterion A (i.e., active-phase symptoms) and may
include periods of prodromal or residual symptoms. During these
prodromal or residual periods, the signs of the disturbance may be
manifested by only negative symptoms or by 2 or more symptoms listed in
Criterion A present in an attenuated form (e.g., odd beliefs, unusual
perceptual experiences).
D. Schizoaffective disorder and depressive or bipolar disorder with psychotic
features have been ruled out because either (1) no major depressive or
manic episodes have occurred concurrently with the active-phase
symptoms; or (2) if mood episodes have occurred during active-phase
symptoms, they have been present for a minority of the total duration of the
active and residual periods of the illness.
E. The disturbance is not attributable to the physiologic effects of a substance
(e.g., a drug of abuse, a medication) or another medical condition.
F. If there is a history of autism spectrum disorder or a communication
disorder of childhood onset, the additional diagnosis of schizophrenia is
made only if prominent delusions or hallucinations, in addition to the other
required symptoms of schizophrenia, are also present for at least a month
(or less if successfully treated).
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 99–100.
Differential Diagnosis
The differential diagnosis for the psychotic disorders is broad and includes
reactions to substances/medications (dextromethorphan, LSD, hallucinogenic
mushrooms, psilocybin, peyote, cannabis, stimulants, inhalants; corticosteroids,
anesthetics, anticholinergics, antihistamines, amphetamines); medical conditions
causing psychotic-like symptoms (Table 47.4 ); and other psychiatric disorders
(depressive, bipolar, obsessive-compulsive, factitious, body dysmorphic,
posttraumatic stress, autism spectrum, communication, personality). The
differential diagnosis can be difficult because many conditions that can be
mistaken for psychosis also increase the risk for it.
Table 47.4
Select Neurologic and Systemic Causes of Depression
and/or Psychosis
CATEGORY DISORDERS
Head trauma Traumatic brain injury
Subdural hematoma
Infectious Lyme disease
Prion diseases
Neurosyphilis
Viral infections/encephalitides (HIV infection/encephalopathy, herpes encephalitis,
cytomegalovirus. Epstein-Barr virus)
Whipple disease
Cerebral malaria
Systemic infection
Inflammatory Autoimmune encephalitis
Celiac disease
Systemic lupus erythematosus
Sjögren syndrome
Temporal arteritis
Hashimoto encephalopathy
Sydenham chorea
Sarcoidosis
Neoplastic Primary or secondary cerebral neoplasm
Systemic neoplasm
Paraneoplastic encephalitis
Endocrine or acquired Hepatic encephalopathy
metabolic Uremic encephalopathy
Dialysis dementia
Hypo/hyperparathyroidism
Hypo/hyperthyroidism
Addison disease, Cushing disease
Postpartum
Vitamin deficiency: vitamin B12 , folate, niacin, vitamin C. thiamine
Gastric bypass–associated nutritional deficiencies
Hypoglycemia
Hyponatremia
Vascular Stroke
Cerebral autosomal dominant aneriopathy with subcortical infarcts and
leukoencephalopathy (CADASIL)
Degenerative Progressive supranuclear palsy
Huntington disease
Corticobasal ganglionic degeneration
Multisystem atrophy, striatonigral degeneration, olivopontocerebellar atrophy
Idiopathic basal ganglia calcifications, Fahr disease
Neuroacanthosis
Neurodegeneration with brain iron accumulation (NBIA)
Adrenoleukodystrophy
Metachromatic leukodystrophy
Demyelinating, Multiple sclerosis
dysmyelinating Acute disseminated encephalomyelitis
Adrenoleukodystrophy
Metachromatic leukodystrophy
Inherited metabolic Wilson disease
Posterior hom syndrome
Tay-Sachs disease
Neuronal ceroid lipofuscinosis
Niemann-Pick disease type C
Acute intermittent porphyria
Mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS)
Cerebrotendinous xanthomatosis
Homocystinuria
Ornithine transcarbamylase deficiency
Syndromes Williams
Prader-Willi
Fragile X
Deletion 22q11.2
ROHHAD
Epilepsy Ictal
Interictal
Postictal
Forced normalization
Postepilepsy surgery
Lafora progressive myoclonic epilepsy
Medications Analgesics
Androgens (anabolic steroids)
Antiarrhythmics
Anticonvulsants
Anticholinergics
Antibiotics
Antihypertensives
Antineoplastic agents
β-Blocking agents
Corticosteroids
Cyclosporin
Dopamine agonists
Oral contraceptives
Sedatives/hypnotics
Selective serotonin reuptake inhibitors (SSRIs) (serotonin syndrome)
Drugs of abuse Alcohol
Amphetamines
Cocaine
Hallucinogens
Marijuana and synthetic cannabinoids
Methylenedioxymethamphetamine (MDMA, Ecstasy)
Phencyclidine
Drug withdrawal Alcohol
syndromes Barbiturates
Benzodiazepines
Amphetamines
SSRIs
Toxins Heavy metals
Inhalants
Other Normal-pressure hydrocephalus
Ionizing radiation
Decompression sickness
ROHHAD, Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, autonomic
dysregulation.
Modified from Perez DL, Murray ED, Price BH: Depression and psychosis in neurological practice.
In Daroff RB, Jankovic J, Mazziotta JC, et al, editors: Bradley's neurology in clinical practice, 7th
ed, Philadelphia, 2015, Elsevier.
FIG. 47.1 Clinical characteristics of patients with anti–NMDA receptor encephalitis.
(Modified from Wandinger KP, Saschenbrecker S, Stoecker W, Dalmau J: Anti-NMDA-
receptor encephalitis: a severe multistage, treatable disorder presenting with
psychosis, J Neuroimmunol 231:86-91, 2011, Fig 2.)
Table 47.5
Special Problems in the Differential Diagnosis of Delirium*
CLINICAL
DELIRIUM DEMENTIAS SCHIZOPHRENIA DEPRESSION
FEATURE
Course Acute onset; hours, days, Insidious onset, Insidious onset, ≥6 Insidious onset,
or more months or years, mo, acute psychotic at least 2 wk,
progressive phases often months
Attention Markedly impaired Normal early; Normal to mild Mild
attention and arousal impairment later impairment impairment
Fluctuation Prominent in attention Prominent Absent Absent
arousal; disturbed fluctuations absent;
day/night cycle lesser disturbances in
day/night cycle
Perception Misperceptions; Perceptual Hallucinations, May have
hallucinations, usually abnormalities much auditory with mood-
visual, fleeting; less prominent; personal reference congruent
paramnesia paramnesia hallucinations
Speech and language Abnormal clarity, speed, Early anomia; empty Disorganized, with a Decreased
and coherence; disjointed speech; abnormal bizarre theme amount of
and dysarthric; comprehension speech
misnaming; characteristic
dysgraphia
Other cognition Disorientation to time, Disorientation to Disorientation to Mental
place; recent memory and time, place; multiple person; concrete slowing;
visuospatial abnormalities other higher interpretations indecisiveness;
cognitive deficits memory
retrieval
difficulty
Behavior Lethargy or delirium; Disinterested; Systematized Depressed
nonsystematized disengaged; delusions; paranoia; mood;
delusions; emotional disinhibited; bizarre behavior anhedonia; lack
lability delusions and other of energy; sleep
psychiatric and appetite
symptoms disturbances
Electroencephalogram Diffuse slowing; low- Normal early; mild Normal Normal
voltage fast activity; slowing later
specific patterns
* The characteristics listed are the usual ones and not exclusive.
From Mendez MF, Padilla CR: Delirium. In Daroff RB, Jankovic J, Mazziotta JC, et al, editors:
Bradley's neurology in clinical practice, 7th ed, Philadelphia, 2015, Elsevier.
Table 47.6
Features Suggesting Neurologic Disease in
Patients With Psychiatric Symptoms
Atypical Psychiatric Features
Comorbidity
In a review of 35 studies of youth with schizophrenia, rates of comorbidity
approximated 34% for posttraumatic stress disorder, 34% for attention-
deficit/hyperactivity and/or disruptive behavior disorders, and 32% for substance
abuse/dependence.
Sequelae
Follow-up studies of early-onset schizophrenia suggest moderate to severe
impairment across the life span. Poor outcome is predicted by low premorbid
functioning, insidious onset, higher rates of negative symptoms, childhood onset,
and low intellectual functioning. When followed into adulthood, youth with
schizophrenia demonstrated greater social deficits, lower levels of employment,
and were less likely to live independently, relative to those with other childhood
psychotic disorders.
Approximately 5–6% of individuals with schizophrenia die by suicide,
approximately 20% attempt suicide on one or more occasions, and many more
have suicidal ideation. Life expectancy is reduced in individuals with
schizophrenia because of associated medical conditions; a shared vulnerability
for psychosis and medical disorders may explain some of the medical
comorbidity of schizophrenia.
Psychosis Associated With Epilepsy
Joseph Gonzalez-Heydrich, Heather J. Walter, David R. DeMaso
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 115–116.
47.3
Catatonia in Children and
Adolescents
Joseph Gonzalez-Heydrich, Heather J. Walter, David R. DeMaso
Psychotic disorders
Paranoid schizophrenia, catatonic schizophrenia, psychosis,
autism, Prader-Willi syndrome, intellectual impairment
Mood disorders
Bipolar disorder: manic or mixed episodes
Major depressive disorder
Medical conditions
Endocrine abnormalities, infections, electrolyte imbalances,
mutations in SCN2A gene
Neurologic conditions
Epilepsy, strokes, traumatic brain injury, multiple sclerosis,
infectious and autoimmune encephalitis
Drugs
Withdrawal: benzodiazepines, L -dopa, gabapentin
Overdose: LSD, phencyclidine (PCP), cocaine, MDMA (Ecstasy),
disulfiram, levetiracetam
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 120–121.
Bibliography
Consoli A, Benmiloud M, Wachtel L, et al. Electroconvulsive
therapy in adolescents with the catatonia syndrome: efficacy
and ethics. J ECT . 2010;26(4):259–265.
Cornic F, Consoli A, Cohen D. Catatonic syndrome in children
to challenges with behavior, emotional, and academic functioning and social
interaction (Table 48.1 ).
Table 48.1
Table 48.2
often there is a combination of factors underlying weak academic performance. In addition to the
dysfunction in neurodevelopmental domains as listed in the table, the clinician must also consider
the possibility of limitations of intellectual and cognitive abilities or associated social and emotional
problems.
Academic Problems
Reading disorders (see Chapter 50 ) can stem from any number of
neurodevelopmental dysfunctions, as described earlier (see Table 48.2 ). Most
often, language and auditory processing weaknesses are present, as evidenced by
poor phonologic processing that results in deficiencies at the level of decoding
individual words and, consequently, a delay in automaticity (e.g., acquiring a
repertoire of words readers can identify instantly) that causes reading to be slow,
laborious, and frustrating. Deficits in other core neurodevelopmental domains
might also be present. Weak WM might make it difficult for a child to hold
sounds and symbols in mind while breaking down words into their component
than one EF simultaneously is encouraged as a means of scaffolding intervention
and building on previously mastered skills.
Table 48.3
Developmental Therapy
Speech-language pathologists offer intervention for children with various forms
of language disability. Occupational therapists focus on sensorimotor skills,
including the motor skills of students with writing problems, and physical
therapists address gross motor incoordination.
Curriculum Modifications
Many children with neurodevelopmental dysfunctions require alterations in the
school curriculum to succeed, especially as they progress through secondary
school. Students with memory weaknesses might need to have their courses
selected for them so that they do not have an inordinate cumulative memory load
in any single semester. The timing of foreign language learning, the selection of
a mathematics curriculum, and the choice of science courses are critical issues
for many of these struggling adolescents.
Strengthening of Strengths
Affected children need to have their affinities, potentials, and talents identified
clearly and exploited widely. It is as important to augment strengths as it is to
attempt to remedy deficiencies. Athletic skills, artistic inclinations, creative
talents, and mechanical abilities are among the potential assets of certain
students who are underachieving academically. Parents and school personnel
need to create opportunities for such students to build on these assets and to
achieve respect and praise for their efforts. These well-developed personal assets
can ultimately have implications for the transition into young adulthood,
including career or college selection.
Attention-Deficit/Hyperactivity
Disorder
David K. Urion
Table 49.2
Differences Between U.S. and European Criteria for ADHD
or HKD
Table 49.3
Differential Diagnosis of Attention-
Deficit/Hyperactivity Disorder (ADHD)
Psychosocial Factors
Fragile X syndrome
Fetal alcohol syndrome
Pervasive developmental disorders
Obsessive-compulsive disorder
Gilles de la Tourette syndrome
Attachment disorder with mixed emotions and conduct
Awareness that most public school systems have implemented some form of a
RtI to identify learning disabilities allows the primary care physician to
encourage parents to return to the school seeking an intervention to address their
child's concern. Receiving special education services in the form of an IEP may
be necessary for some children. However, the current approach to identifying
children with a learning disability allows school systems to intervene earlier,
when problems arise, and potentially avoid the need for an IEP. Pediatricians
with patients whose parents have received feedback from school with any of the
risk factors outlined in Table 51.2 should encourage the parents to discuss an
intervention plan with the child's teacher.
Table 51.2
Risk Factors for a Specific Learning
Disability Involving Mathematics
The child is at or below the 20th percentile in any math area, as reflected
by standardized testing or ongoing measures of progress monitoring.
The teacher expresses concerns about the child's ability to “take the next
step” in math.
There is a positive family history for math learning disability (this alone
will not initiate an intervention).
Parents think they have to “reteach” math concepts to their child.
Bibliography
Bartelet D, Ansari D, Vaessen A, Blomert L. Cognitive subtypes
of mathematics learning difficulties in primary education. Res
Dev Disabil . 2014;35(3):657–670.
Chodura S, Kuhn JT, Holling H. Interventions for children with
mathematical difficulties: a meta-analysis. Z Psychol .
2015;223(2):129–144.
Docherty SJ, Davis OSP, Kovas Y, et al. Genome-wide
association study identifies multiple loci associated with
mathematics ability and disability. Genes Brain Behav .
2010;9:234–247.
Geary DC. Mathematical cognition deficits in children with
learning disabilities and persistent low achievement: a five-
year prospective study. J Educ Psychol . 2012;104(1):206–
223.
Kaufman L, Mazzocco MM, Dowker A, et al. Dyscalculia from
a developmental and differential perspective. Front Psychol .
2013;4:1–5.
Kucian K. Developmental dyscalculia and the brain. Berch DB,
Geary DC, Koepke KM. Development of mathematical
cognition: neural substrates and genetic influences . Elsevier:
New York; 2016:165–193.
Mazzocco M. Mathematics awareness month: why should
pediatricians be aware of mathematics and numeracy? J Dev
Behav Pediatr . 2016;37:251–253.
Mazzocco MM, Quintero AI, Murphy MM, McCloskey M.
Genetic syndromes as model pathways to mathematical
learning difficulties: fragile X, Turner and 22q deletion
Oral language is a complex process that typically develops in the absence of
formal instruction. In contrast, written language requires instruction in
acquisition (word reading), understanding (reading comprehension), and
expression (spelling and composition). Unfortunately, despite reasonable
pedagogy, a subset of children struggle with development in one or several of
these areas. The disordered output of written language is currently referred to
within the Diagnostic and Statistical Manual for Mental Disorders, Fifth Edition
(DSM-5) as a specific learning disorder with impairment in written
expression (Table 51.3 ).
Table 51.3
DSM-5 Diagnostic Criteria for Specific
Learning Disability With Impairment in
Written Expression
A. Difficulties learning and using academic skills that have persisted for at
least 6 mo, despite the provision of interventions that target those
difficulties.
Difficulties with written expression (e.g., makes multiple
grammatical or punctuation errors within sentences; employs
poor paragraph organization; written expression of ideas lacks
clarity).
B. The affected academic skills are substantially and quantifiably below those
expected for the individual's chronological age, and cause significant
interference with academic or occupational performance, or with activities
of daily living, as confirmed by individually administered standardized
achievement measures and comprehensive clinical assessment. For
individuals age 17 yr and older, a documented history of impairing learning
difficulties may be substituted for the standardized assessment.
C. The learning difficulties begin during school-age years but may not
become fully manifest until the demands for those affected academic skills
exceed the individual's limited capacities (e.g., as in timed tests, reading or
writing lengthy complex reports for a tight deadline, excessively heavy
academic loads).
D. The learning difficulties are not better accounted for by intellectual
disabilities, uncorrected visual or auditory acuity, other mental or
neurologic disorders, psychosocial adversity, lack of proficiency in the
language of academic instruction, or inadequate educational instruction.
Adapted from the Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition, (Copyright 2013). American Psychiatric Association, pp 66–67.
Various terminology has been used when referring to individuals with writing
deficits; this subchapter uses the term impairment in written expression (IWE)
rather than “writing disorder” or “disorder of written expression.” Dysgraphia is
often used when referring to children with writing problems, sometimes
synonymously with IWE, although the two are related but distinct conditions.
Dysgraphia is primarily a deficit in motor output (paper/pencil skills), and IWE
is a conceptual weakness in developing, organizing, and elaborating on ideas in
writing.
Table 52.1
Normal Language Milestones: Birth to 5 Years
Classification
Each professional discipline has adopted a somewhat different classification
system, based on cluster patterns of symptoms. The American Psychiatric
Association (APA) Diagnostic and Statistical Manual of Mental Disorders, Fifth
Edition (DSM-5) organized communication disorders into: (1) language disorder
(which combines expressive and mixed receptive-expressive language
disorders), speech sound disorder (phonologic disorder), and childhood-onset
fluency disorder (stuttering); and (2) social (pragmatic) communication disorder,
which is characterized by persistent difficulties in the social uses of verbal and
nonverbal communication (Table 52.2 ). In clinical practice, childhood speech
and language disorders occur as a number of distinct entities.
Table 52.2
DSM-5 Diagnostic Criteria for
Communication Disorders
Language Disorder
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 42, 44, 47–48.
Table 52.3
Speech and Language Screening
REFER FOR SPEECH–LANGUAGE EVALUATION IF:
AT RECEPTIVE EXPRESSIVE
AGE
15 Does not look/point at 5-10 objects Is not using 3 words
mo
18 Does not follow simple directions (“get Is not using Mama, Dada, or other names
mo your shoes”)
24 Does not point to pictures or body parts Is not using 25 words
mo when they are named
30 Does not verbally respond or nod/shake Is not using unique 2-word phrases, including noun–verb
mo head to questions combinations
36 Does not understand prepositions or Has a vocabulary of <200 words; does not ask for things;
mo action words; does not follow 2-step echolalia to questions; language regression after attaining 2-
directions word phrases
Table 52.4
A. Disturbances in the normal fluency and time patterning of speech that are
inappropriate for the individual's age and language skills, persist over time,
and are characterized by frequent and marked occurrences of one (or more)
of the following:
1. Sound and syllable repetitions.
2. Sound prolongations of consonants as well as vowels.
3. Broken words (e.g., pauses within a word).
4. Audible or silent blocking (filled or unfilled pauses in speech).
5. Circumlocutions (word substitutions to avoid problematic words).
6. Words produced with an excess of physical tension.
7. Monosyllabic whole-word repetitions (e.g., “I-I-I-I see him”).
B. The disturbance causes anxiety about speaking or limitations in effective
communication, social participation, or academic or occupational
performance, individually or in any combination.
C. The onset of symptoms is in the early developmental period.
Note: Later-onset cases are diagnosed as 307.0 [F98.5] adult-onset fluency
disorder.
D. The disturbance is not attributable to a speech-motor or sensory deficit,
dysfluency associated with neurologic insult (e.g., stroke, tumor, trauma),
or another medical condition and is not better explained by another mental
disorder.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 45–46.)
Epidemiology
Although prevalence studies have produced a range of estimates for
developmental stuttering, it appears that 0.75–1% of the population is
experiencing this condition at any one time. Incidence rates are considerably
higher: Estimates to date suggest an incidence rate of approximately 5%, with
rates considerably higher among young children than older children or
adolescents. Seldom does a child begin stuttering before 2 yr of age or after 12
yr; in fact, the mean age of onset is 2-4 yr, and most children stop stuttering
abnormalities may be mendelian (autosomal dominant de novo, autosomal
recessive, X-linked) or nonmendelian (imprinting, methylation, mitochondrial
defects; see Chapter 97 ). De novo mutations may also cause other phenotypic
features such as seizures or autism; the presence of these features suggests more
pleotropic manifestations of genetic mutations. Consistent with the finding that
disorders altering early embryogenesis are the most common and severe, the
earlier the problem occurs in development, the more severe its consequences
tend to be.
Table 53.1
Clinical Manifestations
Early diagnosis of ID facilitates earlier intervention, identification of abilities,
realistic goal setting, easing of parental anxiety, and greater acceptance of the
child in the community. Most children with ID first come to the pediatrician's
attention in infancy because of dysmorphisms, associated developmental
disabilities, or failure to meet age-appropriate developmental milestones (Tables
53.2 and 53.3 ). There are no specific physical characteristics of ID, but
dysmorphisms may be the earliest signs that bring children to the attention of the
pediatrician. They might fall within a genetic syndrome such as Down syndrome
or might be isolated, as in microcephaly or failure to thrive. Associated
developmental disabilities include seizure disorders, cerebral palsy, and ASD.
Table 53.2
Physical Examination of a Child With Suspected
Developmental Disabilities
Table 53.3
Most children with ID do not keep up with their peers' developmental skills.
In early infancy, failure to meet age-appropriate expectations can include a lack
of visual or auditory responsiveness, unusual muscle tone (hypo- or hypertonia)
or posture, and feeding difficulties. Between 6 and 18 mo of age, gross motor
delay (lack of sitting, crawling, walking) is the most common complaint.
Language delay and behavior problems are common concerns after 18 mo (Table
53.4 ). For some children with mild ID, the diagnosis remains uncertain during
the early school years. It is only after the demands of the school setting increase
over the years, changing from “learning to read” to “reading to learn,” that the
child's limitations are clarified. Adolescents with mild ID are typically up to date
on current trends and are conversant as to “who,” “what,” and “where.” It is not
until the “why” and “how” questions are asked that their limitations become
apparent. If allowed to interact at a superficial level, their mild ID might not be
appreciated, even by professionals, who may be their special education teachers
or healthcare providers. Because of the stigma associated with ID, adolescents
may use euphemisms to avoid being thought of as “stupid” or “retarded” and
may refer to themselves as learning disabled, dyslexic, language disordered, or
slow learners. Some people with ID emulate their social milieu to be accepted.
They may be social chameleons and assume the morals of the group to whom
they are attached. Some would rather be thought “bad” than “incompetent.”
Table 53.4
Diagnostic Evaluation
Intellectual disability is one of the most frequent reasons for referral to pediatric
genetic providers, with separate but similar diagnostic evaluation guidelines put
forth by the American College of Medical Genetics, the American Academy of
Neurology, the American Academy of Pediatrics (AAP), and the American
Academy of Child and Adolescent Psychiatry. ID is a diagnosis of great clinical
heterogeneity, with only a subset of syndromic etiologies identifiable through
classic dysmorphology. If diagnosis is not made after conducting an appropriate
history and physical examination, chromosomal microarray is the recommended
first step in the diagnostic evaluation of ID. Next-generation sequencing
represents the new diagnostic frontier, with extensive gene panels (exome or
whole genome) that increase the diagnostic yield and usefulness of genetic
testing in ID. Other commonly used medical diagnostic testing for children with
ID includes neuroimaging, metabolic testing, and electroencephalography (Fig.
53.1 ).
FIG. 53.1 Algorithm for the evaluation of the child with unexplained global
developmental delay (GDD) or intellectual disability (ID). AA, amino acids; ASD, autistic
spectrum disorder; CK, creatine kinase; CSF, cerebrospinal fluid; FBC, full blood count;
GAA, guanidinoacetic acid; GAG, glycosaminoglycans; LFT, liver function test; OA,
organic acids; TFT, thyroid function tests; TSC, tuberous sclerosis complex; U&E, urea
and electrolytes; VLCFA, very long chain fatty acids; WES, whole exome sequencing;
WGS, whole genome sequencing; X-linked intellectual disability genes.
Table 53.5
Table 53.6
Treatable Intellectual Disability Endeavor
(TIDE) Diagnostic Protocol
Tier 1: Nontargeted Metabolic Screening to Identify 54 (60%) Treatable
IEM
Blood
Plasma amino acids
Plasma total homocysteine
Acylcarnitine profile
Copper, ceruloplasmin
Urine
Organic acids
Purines and pyrimidines
Creatine metabolites
Oligosaccharides
Glycosaminoglycans
Amino acids (when indicated)
Audiology
Ophthalmology
Cytogenetic testing (array CGH)
Thyroid studies
Complete blood count (CBC)
Lead
Metabolic testing
Brain MRI and 1H spectroscopy (where available)
Fragile X
Targeted gene sequencing/molecular panel
Other
Some children with subtle physical or neurologic findings can also have
determinable biologic causes of their ID (see Tables 53.2 and 53.3 ). How
intensively one investigates the cause of a child's ID is based on the following
factors:
Table 53.7
For persons with moderate ID, the goals of education are to enhance adaptive
abilities and “survival” academic and vocational skills so they are better able to
live and function in the adult world (Table 53.7 ). The concept of supported
employment has been very beneficial to these individuals; the person is trained
by a coach to do a specific job in the setting where the person is to work,
bypassing the need for a “sheltered workshop” experience and resulting in
successful work adaptation in the community. These persons generally live at
home or in a supervised setting in the community.
As adults, people with severe to profound ID usually require extensive to
pervasive supports (Table 53.7 ). These individuals may have associated
impairments, such as cerebral palsy, behavioral disorders, epilepsy, or sensory
impairments, that further limit their adaptive functioning. They can perform
simple tasks in supervised settings. Most people with this level of ID can live in
the community with appropriate supports.
The life expectancy of people with mild ID is similar to the general
population, with a mean age at death in the early 70s. However, persons with
severe and profound ID have a decreased life expectancy at all ages, presumably
from associated serious neurologic or medical disorders, with a mean age at
death in the mid-50s. Given that persons with ID are living longer and have high
rates of comorbid health conditions in adulthood (e.g., obesity, hypertension,
diabetes), ID is now one of the costliest ICD-10 diagnoses, with an average
lifetime cost of 1-2 million dollars per person. Thus the priorities for
pediatricians are to improve healthcare delivery systems during childhood,
facilitate the transition of care to adult providers, and ensure high-quality,
integrated community-based services for all persons with ID.
53.1
Intellectual Disability With
Regression
Bruce K. Shapiro, Meghan E. O'Neill
Alexander disease
Mitochondrial myopathy, encephalopathy, lactic acidosis, stroke
Progressive infantile poliodystrophy (Alpers disease)
Subacute necrotizing encephalomyelopathy (Leigh disease)
Trichopoliodystrophy (Menkes disease)
Neurocutaneous Syndromes
Chediak-Higashi syndrome
Neurofibromatosis*
Tuberous sclerosis*
Progressive Hydrocephalus *
Other Disorders of White Matter
Infectious Disease
Adrenoleukodystrophy
Alexander disease
Cerebrotendinous xanthomatosis
Progressive cavitating leukoencephalopathy
Other Diseases
Wilson disease
Friedreich ataxia
Pantothenate kinase neurodegeneration
Neurodegeneration with brain iron accumulation
treatment.
Bibliography
American Academy of Pediatrics, Committee on Children with
Disabilities. Pediatrician's role in the development and
CHAPTER 54
Definition
Autism spectrum disorder (ASD) is a neurobiologic disorder with onset in
early childhood. The key features are impairment in social communication and
social interaction accompanied by restricted and repetitive behaviors. The
presentation of ASD can vary significantly from one individual to another, as
well as over the course of development for a particular child. There is currently
no diagnostic biomarker for ASD. Accurate diagnosis therefore requires careful
review of the history and direct observation of the child's behavior.
From the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition,
(Copyright 2013). American Psychiatric Association, pp 50–51.
Table 54.2
Associated Features of Autism Not in DSM-5
Criteria
For version with full references, see Diagnostic and Statistical Manual of Mental
Disorders, Fifth Edition, Washington DC, 2013, American Psychiatric
Association. Adapted from Lai MC, Lombardo MV, Baron-Cohen S: Autism,
Lancet 383:896–910, 2014.
Language delay (in babbling or using words; e.g., using <10 words by age
2 yr).
Regression in, or loss of, use of speech.
Spoken language (if present) may include unusual features, such as
vocalizations that are not speech-like; odd or flat intonation; frequent
repetition of set words and phrases (echolalia); reference to self by name
or “you” or “she” or “he” beyond age 3 yr.
Reduced and/or infrequent use of language for communication; e.g., use of
single words, although able to speak in sentences.
Responding to Others
Adapted from Baird G, Douglas HR, Murphy MS: Recognizing and diagnosing
autism in children and young people: summary of NICE guidance. BMJ
343:d6360, 2011, Box 1, p 901.
Social-Emotional Reciprocity
Reduced social interactions in ASD may range from active avoidance or reduced
social response to having an interest in, but lacking ability to initiate or sustain,
an interaction with peers or adults. A young child with ASD may not respond
when his name is called, may exhibit limited showing and sharing behaviors, and
may prefer solitary play. In addition, the child may avoid attempts by others to
play and may not participate in activities that require taking turns, such as peek-
a-boo and ball play. An older child with ASD may have an interest in peers but
may not know how to initiate or join in play. The child may have trouble with
the rules of conversation and may either talk at length about an area of interest or
abruptly exit the interaction. Younger children often have limited capacity for
imaginative or pretend play skills. Older children may engage in play but lack
flexibility and may be highly directive to peers. Some children with ASD
interact well with adults but struggle to interact with same-age peers.
Table 54.4
Table 54.5
Common Co-occurring Conditions in Autism Spectrum
Disorder (ASD)
INDIVIDUALS
WITH
COMORBIDITY COMMENTS
AUTISM
AFFECTED
DEVELOPMENTAL DISORDERS
Intellectual ~45% Prevalence estimate is affected by the diagnostic boundary and
disability definition of intelligence (e.g., whether verbal ability is used as a
criterion).
In individuals, discrepant performance between subtests is common.
Language disorders Variable In DSM-IV, language delay was a defining feature of autism (autistic
disorder), but is no longer included in DSM-5.
An autism-specific language profile (separate from language
disorders) exists, but with substantial interindividual variability.
Attention- 28–44% In DSM-IV, not diagnosed when occurring in individuals with autism, but
deficit/hyperactivity no longer so in DSM-5.
disorder
Tic disorders 14–38% ~6⋅5% have Tourette syndrome.
Motor abnormality ≤79% See Table 54.2 .
GENERAL MEDICAL DISORDERS
Epilepsy 8–35% Increased frequency in individuals with intellectual disability or
genetic syndromes.
Two peaks of onset: early childhood and adolescence.
Increases risk of poor outcome.
Gastrointestinal 9–70% Common symptoms include chronic constipation, abdominal pain,
problems chronic diarrhea, and gastroesophageal reflux.
Associated disorders include gastritis, esophagitis, gastroesophageal
reflux disease, inflammatory bowel disease, celiac disease, Crohn
disease, and colitis.
Immune ≤38% Associated with allergic and autoimmune disorders.
dysregulation
Genetic disorders 10–20% Collectively called syndromic autism.
Examples include fragile X syndrome (21–50% of individuals
affected have autism), Rett syndrome (most have autistic features but
with profiles different from idiopathic autism), tuberous sclerosis
complex (24–60%), Down syndrome (5–39%), phenylketonuria (5–
20%), CHARGE syndrome* (15–50%), Angelman syndrome (50–
81%), Timothy syndrome (60–70%), and Joubert syndrome (~40%).
Sleep disorders 50–80% Insomnia is the most common.
PSYCHIATRIC DISORDERS
Anxiety ~40% Common across all age-groups.
Most common are social anxiety disorder (13–29% of individuals
with autism) and generalized anxiety disorder (13–22%).
High-functioning individuals are more susceptible (or symptoms are
more detectable).
Depression 12–70% Common in adults, less common in children.
High-functioning adults who are less socially impaired are more
susceptible (or symptoms are more detectable).
Obsessive- 7–24% Shares the repetitive behavior domain with autism that could cut
compulsive disorder across nosologic categories.
(OCD) Important to distinguish between repetitive behaviors that do not
involve intrusive, anxiety-causing thoughts or obsessions (part of
autism) and those that do (and are part of OCD).
Psychotic disorders 12–17% Mainly in adults.
Most commonly recurrent hallucinosis.
High frequency of autism-like features (even a diagnosis of ASD)
preceding adult-onset (52%) and childhood-onset schizophrenia (30–
50%).
Substance use ≤16% Potentially because individual is using substances as self-medication to
disorders relieve anxiety.
Oppositional 16–28% Oppositional behaviors could be a manifestation of anxiety, resistance to
defiant disorder change, stubborn belief in the correctness of own point of view, difficulty
seeing another's point of view, poor awareness of the effect of own
behavior on others, or no interest in social compliance.
Eating disorders 4–5% Could be a misdiagnosis of autism, particularly in females, because both
involve rigid behavior, inflexible cognition, self-focus, and focus on
details.
PERSONALITY DISORDERS †
Paranoid 0–19% Could be secondary to difficulty understanding others' intentions and
personality disorder negative interpersonal experiences.
Schizoid 21–26% Partly overlapping diagnostic criteria.
personality disorder
Schizotypal 2–13% Some overlapping criteria, especially those shared with schizoid
personality disorder personality disorder.
Borderline 0–9% Could have similarity in behaviors (e.g., difficulties in interpersonal
personality disorder relationships, misattributing hostile intentions, problems with affect
regulation), which requires careful differential diagnosis.
Could be a misdiagnosis of autism, particularly in females.
Obsessive- 19–32% Partly overlapping diagnostic criteria.
compulsive
personality disorder
Avoidant 13–25% Could be secondary to repeated failure in social experiences.
personality disorder
BEHAVIORAL DISORDERS
Aggressive ≤68% Often directed toward caregivers rather than noncaregivers.
behaviors Could be a result of empathy difficulties, anxiety, sensory overload,
disruption of routines, and difficulties with communication.
Self-injurious ≤50% Associated with impulsivity and hyperactivity, negative affect, and
behaviors lower levels of ability and speech.
Could signal frustration in individuals with reduced communication,
as well as anxiety, sensory overload, or disruption of routines.
Could also become a repetitive habit.
Could cause tissue damage and need for restraint.
Pica ~36% More likely in individuals with intellectual disability.
Could be a result of a lack of social conformity to cultural categories
of what is deemed edible, or sensory exploration, or both.
Suicidal ideation or 11–14% Risks increase with concurrent depression and behavioral problems, and
attempt after being teased or bullied.
* Coloboma of the eye; heart defects; atresia of the choanae; retardation of growth and
development, or both; genital and urinary abnormalities, or both; and ear abnormalities and
deafness.
† Particularly in high-functioning adults.
DSM-IV, Diagnostic and Statistical Manual of Mental Disorders, 4th edition ; DSM-5, Diagnostic
and Statistical Manual of Mental Disorders, Fifth edition.
Adapted from Lai MC, Lombardo MV, Baron-Cohen S: Autism, Lancet 383:896–910, 2014.
Epidemiology
The prevalence of ASD is estimated at 1 in 59 persons by the U.S. Centers for
Disease Control and Prevention (CDC). The prevalence increased significantly
over the past 25 years, primarily because of improved diagnosis and case finding
the child's age and the presenting concerns.
Assessment of ASD includes direct observation of the child to evaluate social
skills and behavior. Informal observation can be supplemented with structured
diagnostic tools such as the Autism Diagnostic Observation Schedule, Second
Edition (ADOS-2) and Autism Diagnostic Observation Schedule, Toddler
module (ADOS-T). These structured play-based assessments provide social
prompts and opportunities to evaluate the frequency and quality of a child's
social responsiveness to, initiation, and maintenance of social interactions; the
capacity for joint attention and shared enjoyment; the child's behavioral
flexibility; and presence of repetitive patterns of behavior. The ADOS-2 and
ADOS-T are not required for accurate diagnosis and do not stand alone, but
rather can be used to augment a careful history and observation. The Childhood
Autism Rating Scale, Second Edition (CARS-2) is a 15-item direct clinical
observation instrument that can assist clinicians in the diagnosis of ASD. The
Autism Diagnostic Interview-Revised (ADI-R) is a lengthy clinical interview tool
that is used primarily in research settings since it takes several hours to
administer. Other tools include standardized rating scales that parents and
teachers can complete to report on the child's social skills and behaviors.
Medical evaluation should include a thorough history and detailed physical
examination of the child, including direct behavioral observations of
communication and play. In addition, the examination should include
measurement of head circumference, careful evaluation for dysmorphic features,
and screening for tuberous sclerosis with Wood lamp exam. Children with ASD
should have genetic testing (described later), an audiology examination to rule
out hearing loss, and in children with pica, a lead test (Table 54.6 ).
Table 54.6
Medical and Genetic Evaluation of Children
With Autism Spectrum Disorder
Physical Examination
Table 54.8
Common Pharmacologic Treatments in Autism Spectrum
Disorder (ASD)
TARGET MEDICATION
EFFECTS SIDE EFFECTS MONITORING
SYMPTOM CLASS*
Hyperactivity Stimulants Decreased Activation, irritability, emotional Height, weight, BP,
and/or hyperactivity, lability, lethargy/social HR
Inattention impulsivity, improved withdrawal, stomach ache,
attention reduced appetite, insomnia,
increased stereotypy
α2 -Agonists Decreased Drowsiness, irritability, enuresis, Height, weight, BP,
hyperactivity, decreased appetite, dry mouth, HR
impulsivity, improved hypotension
attention
Selective Decreased Irritability, decreased appetite, Height, weight, BP,
norepinephrine hyperactivity, fatigue, stomach ache, nausea, HR
reuptake impulsivity, improved vomiting, racing heart rate
inhibitor attention
Anxiety Selective Decreased anxiety Activation, hyperactivity, Weight, BP, HR
serotonin inattention, sedation, change
reuptake in appetite, insomnia,
inhibitors stomach ache, diarrhea
Citalopram: prolonged QTc
interval
Irritability Atypical Decreased irritability, Somnolence, weight gain, Weight, BP, HR
antipsychotics aggression, self- extrapyramidal movements, Monitor CBC,
(risperidone, injurious behavior, drooling, tremor, dizziness, cholesterol,
aripiprazole) repetitive behavior, vomiting, gynecomastia ALT, AST,
hyperactivity prolactin,
glucose or
hemoglobin
A1c
Insomnia Melatonin Shortened sleep onset Nightmares, enuresis —
* Specific medications names are provided in parentheses when there is a FDA-approved
indication for the use of the medication to treat the symptom in children with ASD. Further
information about these medications is available in Chapter 33 .
BP, Blood pressure; HR, heart rate; CBC, complete blood count; ALT, alanine transaminase; AST,
aspartate transaminase.
inability to provide adequate energy substrate. Excess energy intakes can
increase the risk for obesity. Adequacy of energy intake in adults is associated
with maintenance of a healthy weight. The three components of energy
expenditure in adults are the basal metabolic rate (BMR), thermal effect of food
(e.g., energy required for digestion and absorption), and energy for physical
activity. In children, additional energy intake is required to support growth and
development.
Estimated energy requirement (EER) is the average dietary energy intake
predicted to maintain energy balance in a healthy individual and takes into
account age, gender, weight, stature, and level of physical activity (Table 55.1 ).
The 2015–2020 Dietary Guidelines for Americans refer to the 2008 Physical
Activity Guidelines for Americans. These guidelines recommend ≥60 min of
moderate- or vigorous-intensity aerobic physical daily for children and
adolescents. This activity should include vigorous intensity physical activity at
least 3 days per week. In addition, as part of their ≥60 min of daily physical
activity, children and adolescents are advised to incorporate muscle- and bone-
strengthening activity for ≥3 days a week, to maintain a healthy weight and to
prevent or delay progression of chronic noncommunicable diseases such as
obesity and CV disease.
Table 55.1
Equations to Estimate Energy Requirement
INFANTS AND YOUNG CHILDREN: EER (kcal/day) = TEE + ED
0-3 mo EER = (89 × weight [kg] − 100) + 175
4-6 mo EER = (89 × weight [kg] − 100) + 56
7-12 mo EER = (89 × weight [kg] − 100) + 22
13-36 mo EER = (89 × weight [kg] − 100) + 20
CHILDREN AND ADOLESCENTS 3-18 yr: EER (kcal/day) = TEE + ED
Boys
3-8 yr EER = 88.5 − (61.9 × age [yr] + PA × [(26.7 × weight [kg] + (903 × height [m])] + 20
9-18 yr EER = 88.5 − (61.9 × age [yr] + PA × [(26.7 × weight [kg] + (903 × height [m])] + 25
Girls
3-8 yr EER = 135.3 − (30.8 × age [yr] + PA [(10 × weight [kg] + (934 × height [m])] + 20
9-18 yr EER = 135.3 − (30.8 × age [yr] + PA [(10 × weight [kg] + (934 × height [m])] + 25
EER, Estimated energy requirement; TEE, total energy expenditure; ED, energy deposition
(energy required for growth /new tissue accretion).
PA indicates the physical activity coefficient:
For boys:
PA = 1.00 (sedentary, estimated physical activity level 1.0-1.4)
PA = 1.13 (low active, estimated physical activity level 1.4-1.6)
PA = 1.26 (active, estimated physical activity level 1.6-1.9)
PA = 1.42 (very active, estimated physical activity level 1.9-2.5)
For girls:
PA = 1.00 (sedentary, estimated physical activity level 1.0-1.4)
PA = 1.16 (low active, estimated physical activity level 1.4-1.6)
PA = 1.31 (active, estimated physical activity level 1.6-1.9)
PA = 1.56 (very active, estimated physical activity level 1.9-2.5)
Adapted from Kleinman RE, editor: Pediatric nutrition handbook, ed 7, Elk Grove Village, IL, 2013,
American Academy of Pediatrics.
Table 55.2
Acceptable Macronutrient Distribution Ranges
AMDA (% OF ENERGY)
Macronutrient Age 1-3 yr Age 4-18 yr
Fat 30-40 25-35
ω6 PUFAs (linoleic acid) 5-10 5-10
ω3 PUFAs (α-linolenic acid) 0.6-1.2 0.6-1.2
Carbohydrate 45-65 45-65
Protein 5-20 10-30
PUFAs, Polyunsaturated fatty acids.
Adapted from Otten JJ, Hellwig JP, Meyers LD, editors; Institute of Medicine: Dietary reference
intakes: the essential guide to nutrient requirements , Washington, DC, 2006, National Academies
Press.
Fat
Fat is the most calorically dense macronutrient, providing approximately 9
kcal/g. For infants, human milk and formula are the main dietary sources of fat,
whereas older children obtain fat from animal products, vegetable oils, and
margarine. The AMDR for fats is 30–40% of total energy intake for children 1-3
yr and 25–35% for children 4-18 yr of age. In addition to being energy dense,
fats provide essential fatty acids that have body structural and functional roles
(e.g., cholesterol moieties are precursors for cell membranes, hormones, and bile
acids). Fat intake facilitates absorption of fat-soluble vitamins (vitamins A, D, E,
and K). Both roles are relevant to neurologic and ocular development (Table
55.3 ).
Table 55.3
Dietary Reference Intakes: Macronutrients
LIFE
RDA OR AI* SELECTED FOOD ADVERSE EFFECTS OF
FUNCTION STAGE
(g/day) SOURCES EXCESSIVE CONSUMPTION
GROUP
TOTAL DIGESTIBLE CARBOHYDRATE
RDA based on its Infants Major types: No defined intake level for
role as the primary 0-6 mo 60* starches and potential adverse effects of
energy source for 7-12 mo 95* sugars, grains, total digestible carbohydrate is
the brain Children and vegetables identified, but the upper end of
AMDR based on >1 yr 130 (corn, pasta, rice, the AMDR was based on
its role as a source Pregnancy potatoes, and decreasing risk of chronic
of kcal to maintain ≤18 yr 175 breads) are disease and providing adequate
body weight 19-30 yr 175 sources of starch. intake of other nutrients.
Natural sugars are It is suggested that the maximal
found in fruits intake of added sugars be
and juices. limited to providing no more
Sources of added than 10% of energy.
sugars: soft
drinks, candy,
fruit drinks,
desserts, syrups,
and sweeteners †
TOTAL FIBER
Improves laxation, Infants Includes dietary fiber Dietary fiber can have variable
reduces risk of 0-6 mo ND naturally present in compositions; therefore it is
coronary artery (heart) 7-12 mo ND grains (e.g., oats, difficult to link a specific
disease, assists in Children wheat, unmilled rice) source of fiber with a particular
maintaining normal 1-3 yr 190* and functional fiber adverse effect, especially when
blood glucose levels 4-8 yr 25* synthesized or isolated phytate is also present in the
Males from plants or animals natural fiber source.
9-13 yr 31* and shown to be of As part of an overall healthy
benefit to health diet, a high intake of dietary
14-18 yr 38*
fiber will not produce
19-21 yr 38* deleterious effects in healthy
Females persons.
9-13 yr 26* Occasional adverse GI
14-18 yr 26* symptoms are observed when
19-21 yr 25* consuming some isolated or
Pregnancy synthetic fibers, but serious
≤18 yr 28* chronic adverse effects have
19-21 yr 28* not been observed because of
the bulky nature of fibers.
Excess consumption is likely to
be self-limiting; therefore, UL
was not set for individual
functional fibers.
TOTAL FAT
Energy source Infants Infants: Human UL is not set because there is
When found in 0-6 mo 31* milk or infant no defined intake of fat at
foods, is a source 7- 30* formula which adverse effects occur.
of ω3 and ω6 12 Insufficient Older children: High fat intake will lead to
PUFAs mo evidence to Butter, margarine, obesity. Upper end of AMDR
Facilitates 1- determine vegetable oils, is also based on reducing risk
absorption of fat- 18 AI or whole milk, of chronic disease and
soluble vitamins yr EAR; see visible fat on providing adequate intake of
AMDR, meat and poultry other nutrients. †
Table 55.2 products, invisible Low fat intake (with high
. fat in fish, carbohydrate) has been shown
shellfish, some to increase plasma
plant products triacylglycerol concentrations
such as seeds and and decrease HDL cholesterol.
nuts, bakery
products
ω6 POLYUNSATURATED FATTY ACIDS
Essential Infants Nuts, seeds; vegetable There is no defined intake of
component of 0-6 mo 4.4* oils such as soybean, ω6 level at which adverse
structural 7-12 mo 4.6* safflower, corn oil effects occur.
membrane lipids, Children Upper end of AMDR is based
involved with cell 1-3 yr 7* on the lack of evidence that
signaling 4-8 yr 10* demonstrates long-term safety
Precursor of Males and human in vitro studies that
eicosanoids show increased free radical
9-13 yr 12*
Required for formation and lipid
normal skin 14-18 yr 16* peroxidation with higher
function 19-21 yr 17* amounts of ω6 fatty acids.
Females Lipid peroxidation is thought to
9-13 yr 10* be a component of
14-18 yr 11* atherosclerotic plaques.
19-21 yr 12*
Pregnancy
≤18 yr 13*
19-21 yr 13*
Lactation
≤18 yr 13*
19-21 yr 13*
ω3 POLYUNSATURATED FATTY ACIDS
Involved with Infants Vegetable oils, e.g., No defined intake levels for
neurologic 0-6 mo 0.5* soybean, canola, flax potential adverse effects of ω3
development and 7-12 mo 0.5* seed oil; fish oils, fatty PUFAs are identified.
growth Children fish, walnuts; † smaller Upper end of AMDR is based
Precursor of 1-3 yr 0.7* amounts in meats and on maintaining appropriate
eicosanoids 4-8 yr 0.9* eggs balance with ω6 fatty acids and
Males the lack of evidence that
9-13 yr 1.2* demonstrates long-term safety,
along with human in vitro
14-18 yr 1.6*
studies that show increased free
19-21 yr 1.6*
radical formation and lipid
Females peroxidation with higher
9-13 yr 1.0* amounts of PUFAs.
14-18 yr 1.1* Because the longer-chain n -3
19-21 yr 1.1* fatty acids, eicosapentaenoic
Pregnancy acid (EPA) and
≤18 yr 1.4* docosahexaenoic acid (DHA),
19-21 yr 1.4* are biologically more potent
Lactation than their precursor, linolenic
≤18 yr 1.3* acid, much of the work on
19-21 yr 1.3* adverse effects of this group of
fatty acids has been on DHA
and EPA.
Lipid peroxidation is thought to
be a component in the
development of atherosclerotic
plaques.
SATURATED AND TRANS FATTY ACIDS
The body can No dietary Saturated fatty There is an incremental increase in
synthesize its needs for requirement acids are present plasma total and LDL cholesterol
saturated fatty acids in animal fats concentrations with increased intake
from other sources. (meat fats and of saturated or trans fatty acids;
butter fat), and therefore, saturated fat intake
coconut and palm should be limited to <10% with no
kernel oils. trans fat. † ‡
Trans fat: stick
margarines, foods
containing
hydrogenated or
partially
hydrogenated
vegetable
shortenings
CHOLESTEROL
No dietary Sources: liver, eggs,
requirement foods that contain
eggs, e.g., cheesecake,
custard pie
PROTEIN AND AMINO ACIDS ‡
Major structural Infants Proteins from No defined intake levels for
component of all 0-6 mo 9.1* animal sources, potential adverse effects of
cells in the body 7-12 mo 11.0 e.g., meat, protein are identified.
Functions as Children poultry, fish, Upper end of AMDR was
enzymes, in 1-3 yr 13 eggs, milk, based on complementing
membranes, as 4-8 yr 19 cheese, yogurt, AMDR for carbohydrate and
transport carriers, Males provide all 9 fat for the various age-groups.
and as some indispensable Lower end of AMDR is set at
9-13 yr 34
hormones amino acids in approximately the RDA.
14-18 yr 52
During digestion adequate amounts
≥19 yr 56
and absorption, and are
dietary protein is Females considered
broken down to 9-13 yr 34 “complete
amino acids, which ≥14 yr 46 protein.”
become the ≤18 yr Protein from
building blocks of 19-21 yr 71 plants, legumes,
these structural and grains, nuts,
functional seeds, and
compounds. vegetables tend to
Nine indispensable be deficient in ≥1
amino acids must of the
be provided in the indispensable
diet; the body can amino acids and
make the other are called
amino acids “incomplete
needed to protein.”
synthesize specific Vegan diets
structures from adequate in total
other amino acids. protein content
can be “complete”
by combining
sources of
incomplete
protein, which
lack different
indispensable
amino acids.
* Adequate intake.
† 2015–2020 Dietary Guidelines for Americans. US Department of Health and Human Services.
https://health.gov/dietaryguidelines/2015/ .
‡ Based on 1.5 g/kg/day for infants, 1.1 g/kg/day for 1-3 yr, 0.95 g/kg/day for 4-13 yr, 0.85
g/kg/day for 14-18 yr, 0.8 g/kg/day for adults, and 1.1 g/kg/day for pregnant (using pre-pregnancy
weight) and lactating women.
Note: Starred (*) numbers are AI; bold numbers are RDA. RDAs and AIs may both be used as
goals for individual intake. RDAs are set to meet the needs of 97-98% of members in a group. For
healthy breastfed infants, the AI is the mean intake. The AI for other life stage and gender groups
is believed to cover the needs of all members of the group, but lack of data prevents specifying
critical illnesses, burn injuries, compensated liver disease, and bariatric surgery
(e.g., laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass). Intake of
protein or specific amino acids needs to be limited in some health conditions,
such as renal disease and decompensated liver disease, and metabolic diseases
such as phenylketonuria and maple syrup urine disease, in which specific amino
acids can be toxic.
The amino acid content of dietary protein is also important. Certain amino
acids are indispensable , and humans depend on dietary sources to meet
adequacy and prevent deficiency. Certain amino acids are termed conditional
essential/indispensable , meaning they become essential in patients affected by
some diseases or during a certain life stage, such as with cysteine, tyrosine, and
arginine in newborns because of enzyme immaturity (Table 55.4 ). Human milk
contains both the indispensable and conditionally indispensable amino acids and
therefore meets the protein requirements for infants. Breast milk is considered
the optimal protein source for infants and is the reference amino acid
composition by which biologic quality is determined for infants. If a single
amino acid in a food protein source is low or absent but is required to support
normal metabolism, that specific amino acid becomes the limiting nutrient in
that food. For soy-based infant formula, supplementing the formula with the
limiting amino acid (methionine) is necessary. Certain amino acid–like
substances, such as creatinine, are used by some athletes and may enhance
performance. Such supplementation should be monitored for potential side
effects.
Table 55.4
Indispensable, Dispensable, and Conditionally
Indispensable Amino Acids in the Human Diet
Micronutrients
(See Chapters 61-67 .)
Vitamins and trace minerals, the dietary micronutrients , are essential for
growth and development and contribute to a host of physiologic functions. Many
U.S. children have suboptimal intake of iron, zinc, potassium, calcium, vitamin
D, and vitamin K, and excess intake of sodium. Dietary recommendations for
micronutrients were originally established to prevent deficiency and currently
also include the impact of micronutrients on long-term health outcomes (Table
55.5 ). Food fortification is an effective strategy to prevent some nutrient
deficiencies and has been successfully implemented to prevent iodine and folate
deficiency.
Table 55.5
Dietary Reference Intakes for Vitamins
ADVERSE
LIFE- RDA SELECTED
EFFECTS OF SPECIAL
NUTRIENT FUNCTION STAGE OR UL FOOD
EXCESSIVE CONSIDERATIONS
GROUP AI SOURCES
CONSUMPTION
Biotin (vitamin B7 Coenzyme in Infants (µg/day) Liver No adverse None
) synthesis of fat, 0-6 mo 5* ND Smaller effects of
glycogen, and 7-12 mo 6* ND amounts biotin in
amino acids Children (µg/day) in fruits humans or
1-3 yr 8* ND and animals have
4-8 yr 12* ND meats been found;
Males (µg/day) this does not
9-13 yr 20* ND mean there is
no potential
14-18 yr 25* ND
for adverse
19-21 yr 30* ND effects
Females (µg/day) resulting from
9-13 yr 20* ND high intakes.
14-18 yr 25* ND Because data
19-21 yr 30* ND on the adverse
Pregnancy (µg/day) effects of
≤18 yr 30* ND biotin are
19-21 yr 30* ND limited,
Lactation (µg/day) caution may
≤18 yr 35* ND be warranted.
19-21 yr 35* ND
Choline Precursor for Infants (mg/day) Milk, liver, Fishy body odor, Patients with
acetylcholine, eggs, sweating, trimethylaminuria,
phospholipids, 0-6 mo 125* ND peanuts salivation, renal disease, liver
and betaine 7-12 mo 150* ND hypotension, disease,
Children (mg/day) hepatotoxicity depression, and
1-3 yr 200* 1,000 Parkinson disease
4-8 yr 250* 1,000 may be at risk for
Males (mg/day) adverse effects
9-13 yr 375* 2,000 with choline
14-18 yr 550* 3,000 intakes at the UL.
AIs have been set
19-21 yr 550* 3,500
for choline, but
Females (mg/day)
there are little data
9-13 yr 375* 2,000 to assess whether
14-18 yr 400* 3,000 a dietary supply of
19-21 yr 425* 3,500 choline is needed
Pregnancy (mg/day) at all stages of the
≤18 yr 450* 3,000 life cycle, and the
19-21 yr 450* 3,500 choline
Lactation (mg/day) requirement might
≤18 yr 550* 3,000 be met by
19-21 yr 550* 3,500 endogenous
synthesis at some
of these stages.
Vitamin C Cofactor for Infants (mg/day) Citrus fruit, GI disturbances, Smokers require
(ascorbic acid, reactions 0-6 mo 40* ND tomatoes, kidney stones, additional 35
dehydroascorbic requiring reduced 7-12 mo 50* ND tomato juice, excess iron mg/day of vitamin
acid) copper or iron Children (mg/day) potatoes, absorption C over that
metalloenzyme 1-3 yr 15 400 Brussels needed by
and as a 4-8 yr 25 650 sprouts, nonsmokers.
protective Males (mg/day) cauliflower, Nonsmokers
antioxidant 9-13 yr 45 1,200 broccoli, regularly exposed
strawberries, to tobacco smoke
14-18 yr 75 1,800
cabbage, should ensure
19-21 yr 90 2,000
spinach they meet the
Females (mg/day) RDA for vitamin
9-13 yr 45 1,200 C.
14-18 yr 65 1,800
19-21 yr 75 2,000
Pregnancy (mg/day)
≤18 yr 80 1,800
19-21 yr 85 2,000
Lactation (mg/day)
≤18 yr 115 1,800
19-21 yr 120 2,000
Vitamin E (α- A metabolic Infants (mg/day) Vegetable No evidence Patients receiving
tocopherol function has oil, of adverse anticoagulant therapy
α-Tocopherol not yet been 0-6 mo 4* ND unprocessed effects from should be monitored
includes RRR- identified. 7-12 mo 5* ND cereal grains, consuming when taking vitamin E
α- tocopherol, Vitamin E's Children (mg/day) nuts, fruit, vitamin E supplements.
the only form major 1-3 yr 6 200 vegetables, naturally
of α- function 4-8 yr 7 300 meat occurring in
tocopherol that appears to be Males (mg/day) food
occurs as a 9-13 yr 11 600 Adverse
naturally in nonspecific 14-18 yr 15 800 effects from
foods, and the chain- 19-21 yr 15 1,000 vitamin E–
2R - breaking Females (mg/day) containing
stereoisomeric antioxidant. 9-13 yr 11 600 supplements
forms of α- 14-18 yr 15 800 may include
tocopherol 19-21 yr 15 1,000 hemorrhagic
(RRR- , RSR- , Pregnancy (mg/day) toxicity.
RRS-, and UL for
≤18 yr 15 800
RSS- α- vitamin E
19-21 yr 15 1,000
tocopherol) applies to any
that occur in Lactation (mg/day) form of α-
fortified foods ≤18 yr 19 800 tocopherol
and 19-21 yr 19 1,000 obtained from
supplements supplements,
It does not fortified
include the 2S foods, or a
- combination
stereoisomeric of these.
forms of α-
tocopherol
(SRR-, SSR-,
SRS-, and SSS-
α-tocopherol),
also found in
fortified foods
and
supplements
Vitamin K Coenzyme during Infants (µg/day) Green No adverse Patients receiving
synthesis of many 0-6 mo 2.0* ND vegetables effects anticoagulant therapy
proteins involved 7-12 mo 2.5* ND (collards, associated should monitor
in blood clotting Children (µg/day) spinach, with vitamin vitamin K intake.
and bone 1-3 yr 30* ND salad greens, K
metabolism 4-8 yr 55* ND broccoli), consumption
Males (µg/day) Brussels from food or
9-13 yr 60* ND sprouts, supplements
cabbage, have been
14-18 yr 75* ND
plant oil, reported in
19-21 yr 120* ND margarine humans or
Females (µg/day) animals; this
9-13 yr 60* ND does not mean
14-18 yr 75* ND there is no
19-21 yr 90* ND potential for
Pregnancy (µg/day) adverse
≤18 yr 75* ND effects
19-21 yr 90* ND resulting from
Lactation (µg/day) high intake.
≤18 yr 75* ND Because data
19-21 yr 90* ND on adverse
effects of
vitamin K are
limited,
caution may
be warranted.
determined to be low.
Table 55.6
Dietary Reference Intakes for Select Micronutrients and
Water
ADVERSE
LIFE- SELECTED
AI UL EFFECTS OF SPECIAL
NUTRIENT FUNCTION STAGE FOOD
(mg/day) (mg/day) EXCESSIVE CONSIDERATIONS
GROUP SOURCES
CONSUMPTION
Sodium Maintains fluid Infants Processed Hypertension AI is set based on
volume outside 0-6 mo 120 ND foods with Increased risk ability to obtain a
of cells and thus 7-12 mo 370 ND added of nutritionally
normal cell Children sodium cardiovascular adequate diet for
function 1-3 yr 1,000 1,500 chloride disease and other nutrients
4-8 yr 1,200 1,900 (salt), stroke and to meet the
Males benzoate, needs for sweat
9-13 yr 1,500 2,200 phosphate; losses for persons
salted engaged in
14-21 yr 1,500 2,300
meats, recommended
Females
bread, nuts, levels of physical
9-13 yr 1,500 2,200 cold cuts; activity.
13-21 yr 1,500 2,300 margarine; Persons engaged
Pregnancy and Lactation butter; salt in activity at
≥14 yr 1,500 2,300 added to higher levels or in
foods in humid climates
cooking or resulting in
at the table excessive sweat
Salt is about might need more
40% than the AI.
sodium by UL applies to
weight. apparently
healthy persons
without
hypertension; it
thus may be too
high for persons
who already have
hypertension or
who are under the
care of a health
professional.
Chloride With sodium, Infants Processed In concert with Chloride is lost,
maintains fluid foods with sodium, results in usually with
volume outside 0-6 mo 180 ND added hypertension sodium, in sweat,
of cells and thus 7-12 mo 570 ND sodium as well as in
normal cell Children chloride vomiting and
function 1-3 yr 1,500 2,300 (salt), diarrhea.
4-8 yr 1,900 2,900 benzoate, AI and UL are
Males phosphate; equimolar in
9-13 yr 2,300 3,400 salted amount to sodium
14-21 yr 2,300 3,600 meats, nuts, because most of
Females cold cuts; sodium in diet
9-13 yr 2,300 3,400 margarine; comes as sodium
13-21 yr 2,300 3,600 butter; salt chloride (salt).
Pregnancy and Lactation added to
≥14 yr 2,300 3,600 foods in
cooking or
at the table
Salt is about
60%
chloride by
weight.
Potassium Maintains fluid Infants Fruits and None documented Persons taking drugs
volume 0-6 mo 400 None set vegetables, dried from food alone, but for cardiovascular
inside/outside of 7-12 mo 700 peas, dairy potassium from disease such as ACE
cells and thus Children products, meats, supplements or salt inhibitors, ARBs, or
normal cell 1-3 yr 3,000 No UL nuts substitutes can potassium-sparing
function; acts to 4-8 yr 3,800 result in diuretics should be
blunt the rise of Males hyperkalemia and careful not to
blood pressure 9-13 yr 4,500 possibly sudden consume supplements
in response to death if excess is containing potassium
14-21 yr 4,700
excess sodium consumed by and might need to
Females
intake, and persons with consume less than the
decrease 9-13 yr 4,500 chronic renal AI.
markers of bone 13-21 yr 4,700 insufficiency
turnover and Pregnancy (kidney disease) or
recurrence of ≥14 yr 4,700 diabetes.
kidney stones Lactation
≥14 yr 5,100
Vitamin D Maintains serum Infants (µg/day) * Fish liver oils, Elevated plasma Patients receiving
(calciferol) calcium and 0-6 mo 10 25 flesh of fatty 25(OH)D glucocorticoid therapy
1 µg phosphorus 7-12 mo 10 38 fish, liver and concentration might require
calciferol concentrations Children (µg/day) * fat from seals causing additional vitamin D.
= 40 IU 1-3 yr 15 63 and polar bears, hypercalcemia
vitamin D 4-8 yr 15 75 eggs from hens
DRI Males (µg/day) * that have been
values are fed vitamin D,
9-21 yr 15 100
based on fortified milk
Females (µg/day) *
absence of products,
9-21 yr 15 100
adequate fortified cereals
exposure Pregnancy (µg/day) *
to ≤18 yr 15 100
sunlight. Lactation (µg/day)
≤18 yr 15 100
19-21 yr 15 100
Calcium Essential role in Infants Milk, cheese, Kidney stones, Amenorrheic women
blood clotting, 0-6 mo 200 1,000 yogurt, corn hypercalcemia, milk (exercise or anorexia
muscle 7-12 mo 260 1,500 tortillas, alkali syndrome, nervosa induced) have
contraction, Children calcium-set tofu, renal insufficiency reduced net calcium
nerve 1-3 yr 700 2,500 Chinese absorption.
transmission, 4-8 yr 1,000 2,500 cabbage, kale,
and bone and Males broccoli
tooth formation
9-18 yr 1,300 3,000
19-21 yr 1,000 2,500
Females
9-18 yr 1,300 3,000
19-21 yr
Pregnancy
≤18 yr 1,300 3,000
19-21 yr 1,000 2,500
Lactation
≤18 yr 1,300 3,000
19-21 yr 1,000 2,500
Iron Critical Infants Heme GI distress Persons with
component of 0-6 mo 0.27 40 sources: decreased gastric
enzymes, 7-12 mo 11 40 meat, acidity may be at
cytochromes, Children poultry, fish increased risk for
myoglobin, and 1-3 yr 7 40 Nonheme deficiency.
hemoglobin 4-8 yr 10 40 sources: Cow's milk is a
Males dairy, eggs, poor source of
9-13 yr 8 40 plant-based bioavailable iron
foods, and is not
14-18 yr 11 45
breads, recommended for
19-21 yr 8 45
cereals, children <1 yr
Females breakfast old.
9-13 yr 8 40 foods Neurocognitive
14-18 yr 15 45 deficits have been
19-21 yr 18 45 reported in
Pregnancy infants with iron
≤18 yr 27 45 deficiency.
19-21 yr 27 45 RDA for females
Lactation increases with
≤18 yr 10 45 menarche related
19-21 yr 9 45 to increased
losses during
menstruation.
Vegans and
vegetarians might
require iron
supplementation
or intake of iron-
fortified foods.
GI parasites can
increase iron
losses via GI
bleeds.
Iron supplements
can interfere with
zinc absorption,
and vice versa; if
supplements are
being used, the
doses should be
staggered.
Zinc Essential for Infants Meats, shellfish, Acutely, zinc Zinc supplements
proper growth legumes, supplements cause interfere with
and 0-6 mo 2 4 fortified cereals, GI irritation and iron absorption,
development; 7-12 mo 3 5 whole grains headache; chronic and vice versa;
important Children effects of zinc therefore, if
catalyst for 100 1-3 yr 3 7 supplementation supplements are
specific 4-8 yr 5 12 include impaired being used, the
enzymes Males immune function, doses should be
9-13 yr 8 23 changes in staggered.
14-18 yr 11 34 lipoprotein and Zinc deficiency
19-21 yr 11 40 cholesterol levels, can be associated
Females and reduced copper with stunting or
9-13 yr 8 23 status. impaired linear
14-18 yr 9 34 growth.
19-21 yr 8 40
Pregnancy
≤18 yr 12 34
19-21 yr 11 40
Lactation
≤18 yr 13 34
19-21 yr 12 40
Water Maintains Infants (L/day) All No UL because Recommended
homeostasis 0-6 mo 0.7 None set beverages, normally intakes for water
in the body 7-12 mo 0.8 including functioning are based on
Allows Children water kidneys can median intakes of
transport of 1-3 yr 1.3 Moisture in handle >0.7 L generally healthy
nutrients to 4-8 yr 1.7 foods (24 oz) of fluid persons who are
cells and Males (L/day) High- per hour adequately
removal 9-13 yr 2.4 moisture Symptoms of hydrated.
and foods water Persons can be
14-18 yr 3.3
excretion of include intoxication adequately
≥19 yr 3.7
waste watermelon, include hydrated at levels
products of Females (L/day) meats, and hyponatremia, above or below
metabolism 9-13 yr 2.1 soups which can the AIs provided;
14-18 yr 2.3 result in heart AIs provided are
≥19 yr 2.7 failure, and for total water in
Pregnancy (L/day) rhabdomyolysis temperate
≥14 yr 3.0 (skeletal climates.
Lactation (L/day) muscle tissue All sources can
≥14 yr 3.8 injury), which contribute to total
can lead to water needs:
kidney failure. beverages (tea,
coffee, juice,
soda, drinking
water) and
moisture found in
foods.
Moisture in food
accounts for
about 20% of
total water intake.
Thirst and
consumption of
beverages at
meals are
adequate to
maintain
hydration.
* Vitamin D RDA in IU/day: 40 if <1 yr, 600 if >1 yr of age or pregnant or lactating.
also describe growth of adequately nourished healthy children under best-care
practices.
In the clinical setting, the 2.3rd and 97.7th percentiles on the WHO growth
charts are used to identify insufficient and excessive growth from birth to 2 yr,
respectively. In contrast, the 5th and 95th percentiles are recommended for the
equivalent identification in the CDC growth charts from 2-20 yr (Table 55.7 ).
Note that length, weight, and weight-for-length are used in the WHO growth
charts from birth to 2 yr. Body mass index (BMI) can be calculated but is not
recommended for use in children <2 yr. Stature, weight, and BMI are used in the
CDC 2000 growth charts from 2-20 yr of age. These charts can be used to
categorize children 2-20 yr as underweight (<5th BMI percentile), normal
weight (5–85th), overweight (85–95th), and obese (≥95th BMI percentile).
Severe obesity is defined as BMI ≥120% of the 95th percentile, or BMI ≥35
kg/m2 (whichever is lower). This assessment corresponds to approximately the
99th percentile or a BMI z score ≥2.33. Severe obesity that exceeds the 99th
percentile is tracked on a specialized percentile curve for obesity. Furthermore,
adult classification is used for BMI ≥27 kg/m2 in adolescents over age 18 for
consideration of medication and bariatric surgery.
Table 55.7
Growth Chart Comparisons for Measuring Growth from
Birth to 20 Years
INSUFFICIENT EXCESSIVE
GROWTH AGE GROWTH BMI STATUS
GROWTH GROWTH
CHART RANGE METRICS PERCENTILE
PERCENTILE PERCENTILE
World Health Birth to Weight, length, <2.3rd >97.7th —
Organization, 2006 2 yr weight-for-length,
and head
circumference
US Centers for 2-20 yr Weight, height, body <5th >95th Under (<5th)
Disease Control and mass index (BMI) Normal (5–
Prevention, 2000 85th)
Over (85–95th)
Obese (>95th)
Severe Obesity
(≥120% of
95th, or ≥35
kg/m2 )
Table 56.1
Selected Beneficial Properties of Human Milk Compared
With Infant Formula
FACTOR ACTION
ANTIBACTERIAL FACTORS
Secretory IgA Specific antigen-targeted antiinfective action
Lactoferrin Immunomodulation, iron chelation, antimicrobial action, antiadhesive,
trophic for intestinal growth
κ-Casein Antiadhesive, bacterial flora
Oligosaccharides Prevention of bacterial attachment
Cytokines Antiinflammatory, epithelial barrier function
GROWTH FACTORS
Epidermal growth factor Luminal surveillance, repair of intestine
Transforming growth factor (TGF) Promotes epithelial cell growth (TGF-β)
Suppresses lymphocyte function (TGF-β)
Nerve growth factor Promotes neural growth
ENZYMES
Platelet-activating factor (PAF)– Blocks action of PAF
acetylhydrolase
Glutathione peroxidase Prevents lipid oxidation
Nucleotides Enhance antibody responses, bacterial flora
Adapted from Hamosh M: Bioactive factors in human milk, Pediatr Clin North Am 48:69–86, 2001.
Table 56.2
Table 56.3
Conditions for Which Human Milk May Have
a Protective Effect
Diarrhea
Otitis media
Urinary tract infection
Necrotizing enterocolitis
Septicemia
Infant botulism
Insulin-dependent diabetes mellitus
Celiac disease
Crohn disease
Childhood cancer
Lymphoma
Leukemia
Recurrent otitis media
Allergy
Hospitalizations
Infant mortality
Table 56.4
Ten Hospital Practices to Encourage and Support
Breastfeeding* ‡
1. Have a written breastfeeding policy that is routinely communicated to all health care staff.
2. Train all health care staff in the skills necessary to implement this policy.
3. Inform all pregnant women about the benefits and management of breastfeeding.
4. Help women initiate breastfeeding within 1 hour of birth.
5. Show women how to breastfeed and how to maintain lactation, even if they are separated from their newborns.
6. Give newborns no food or drink other than breast milk unless medically indicated.
7. Practice rooming-in; allow mothers and newborns to remain together 24 hours a day.
8. Encourage breastfeeding on demand.
9. Give no pacifiers or artificial nipples to breastfeeding infants. †
10. Foster the establishment of breastfeeding support groups and refer to them on discharge from the hospital or
birth center.
COMPONENTS OF SAFE POSITIONING FOR THE NEWBORN WHILE SKIN-TO-SKIN **
1. Infant's face can be seen.
2. Infant's head is in “sniffing” position.
3. Infant's nose and mouth are not covered.
4. Infant's head is turned to one side.
5. Infant's neck is straight, not bent.
6. Infant's shoulders and chest face mother.
7. Infant's legs are flexed.
8. Infant's back is covered with blankets.
9. Mother-infant dyad is monitored continuously by staff in the delivery environment and regularly on the
postpartum unit.
10. When mother wants to sleep, infant is placed in bassinet or with another support person who is awake and alert.
* The 1994 report of the Healthy Mothers, Health Babies National Coalition Expert Work Group
recommend that the UNICEF-WHO Baby Friendly Hospital Initiative be adapted for use in the
United States as the United States Breastfeeding Health Initiative, using the adapted ten hospital
practices above.
† The American Academy of Pediatrics endorsed the UNICEF-WHO Ten Steps to Successful
Breastfeeding, but does not support a categorical ban on pacifiers because of their role in
reducing the risk of sudden infant death syndrome and their analgesic benefit during painful
procedures when breastfeeding cannot provide the analgesia.
‡ Data from Baby-Friendly USA. Guidelines and evaluation criteria for facilities seeking baby-
friendly designation. Sandwich (MA): Baby Friendly USA, 2010. Available at
https://www.babyfriendlyusa.org/for-facilities/practice-guidelines/. Accessed 10 December 2018.
From ACOG Committee Opinion: Optimizing support for breastfeeding as part of obstetric
practice. Obstet Gynecol 132(4):e187-e195, 2018 (Box 1, p. e191 and Box 2, p. e192).
** Data from Ludington-Hoe SM, Morgan K. Infant assessment and reduction of sudden
unexpected postnatal collapse risk during skin-to-skin contact. Newborn Infant Nurs Rev
2014;14:28-33.
Table 56.5
Recommendations on Breastfeeding
Management for Healthy Term Infants
1. Exclusive breastfeeding for about 6 months
• Breastfeeding preferred; alternatively expressed mother's milk, or
donor breast milk
• To continue for at least the first year and beyond as long as
mutually desired by mother and child
• Complementary foods rich in iron and other micronutrients should
be introduced at about 6 mo of age
2. Peripartum policies and practices that optimize breastfeeding initiation
and maintenance should be compatible with the AAP and Academy of
Breastfeeding Medicine Model Hospital Policy and include the
following:
• Direct skin-to-skin contact with mothers immediately after delivery
until the first feeding is accomplished and encouraged throughout
the postpartum period
• Delay in routine procedures (weighing, measuring, bathing, blood
tests, vaccines, and eye prophylaxis) until after the first feeding is
completed
• Delay in administration of intramuscular vitamin K until after the
first feeding is completed but within 6 hr of birth
• Ensure 8-12 feedings at the breast every 24 hr
• Ensure formal evaluation and documentation of breastfeeding by
trained caregivers (including position, latch, milk transfer,
examination) at least once for each nursing shift
• Give no supplements (water, glucose water, commercial infant
formula, or other fluids) to breastfeeding newborn infants unless
medically indicated using standard evidence-based guidelines for
the management of hyperbilirubinemia and hypoglycemia
• Avoid routine pacifier use in the postpartum period
• Begin daily oral vitamin D drops (400 IU) at hospital discharge
3. All breastfeeding infants should be seen by a pediatrician within 48 to 72
hr after discharge from the hospital
• Evaluate hydration and elimination patterns
• Evaluate body weight gain (body weight loss no more than 7%
from birth and no further weight loss by day 5: assess feeding and
consider more frequent follow-up)
• Discuss maternal/infant issues
• Observe feeding
4. Mother and infant should sleep in proximity to each other to facilitate
breastfeeding
5. Pacifier should be offered, while placing infant in back-to-sleep-position,
no earlier than 3 to 4 weeks of age and after breastfeeding has been
established
Table 56.6
New mothers should be instructed about infant hunger cues, correct nipple
latch, positioning of the infant on the breast, and feeding frequency. It is also
suggested that someone trained in lactation observe a feeding to evaluate
positioning, latch, milk transfer, maternal responses, and infant satiety. Attention
to these issues during the birth hospitalization allows dialog with the mother and
family and can prevent problems that could occur with improper technique or
knowledge of breastfeeding. As part of the discharge teaching process, issues on
infant feeding, elimination patterns, breast engorgement, breast care, and
maternal nutrition should be discussed. A follow-up appointment is
recommended within 24-48 hr after hospital discharge.
Nipple Pain
Nipple pain is one of the most common complaints of breastfeeding mothers in
Important Principles for Weaning
Begin at 6 mo of age.
At the proper age, encourage a cup rather than a bottle.
Introduce 1 new food at a time.
Energy density should exceed that of breast milk.
Iron-containing foods (meat, iron-supplemented cereals) are required.
Zinc intake should be encouraged with foods such as meat, dairy products,
wheat, and rice.
Phytate intake should be low to enhance mineral absorption.
Breast milk should continue to 12 mo of age; formula or cow's milk is then
substituted.
Give no more than 24 oz/day of cow's milk.
Fluids other than breast milk, formula, and water should be discouraged.
Give no more than 4-6 oz/day of 100% fruit juice; no sugar-sweetened
beverages.
Juices should not be given before 12 mo of age. The volume of juices should
be limited to 4 oz/day in children 1-3 yr old, to 4-6 oz/day for 4-6 yr olds, and to
8 oz/day for 7-18 yr olds. Children taking medications metabolized by CYP3A4
must avoid grapefruit juices.
In the 2nd year of life, self-feeding becomes a norm and provides the
opportunity for the family to eat together with less stress. Self-feeding allows the
child to limit her intake. Child feeding is an interactive process. Children receive
cues regarding appropriate feeding behaviors from parents. Parents should praise
positive and ignore negative eating behaviors unless the behavior jeopardizes the
health and safety of the child. In addition, parents should eat with their toddlers
and not simply feed them, in order to model positive eating behaviors.
The 2 yr old child should progress from small pieces of soft food to prepared
setting. Another parenting challenge is to control the excess appetite of some
children and adolescents. Children should be supported to eat at a slower pace
and to chew their food properly. Conversation at the dinner table should be
encouraged to prolong eating to at least 15 min. Offering vegetables while
children are hungry at the beginning of the meal has been shown to increase
vegetable consumption. Useful strategies, when the child is still hungry after a
meal, include a 15-20 min pause (allow child to engage in another activity)
before a 2nd serving or offering foods that are insufficiently consumed, such as
vegetables, whole grains, or fruits.
Eating at School
The National School Lunch Program and the School Breakfast Program
provide low-cost meals to more than 5 billion children nationwide. Guidelines
for meals are taken from the Dietary Guidelines for Americans and the 2005
Dietary Reference. Recommendations include the use of age-grade portion sizes
and the amounts of vegetables and fruits, grains, and fats (Table 56.9 ). The
training and equipment for school food service staff, school community
engagement, parent education, and food industry involvement are among the
necessary components. The target year is 2020 for achieving recommendations
for sodium. In the meantime, while schools are working on implementing
changes, parents should be encouraged to examine the weekly menu with their
child and assist with their choices. If children bring their lunch from home,
recommendations for what constitutes a healthful lunch should be provided by
the pediatrician. Parents can be directed to www.choosemyplate.org for healthful
lunch ideas. In addition, parties within classrooms should be limited to once a
month.
Table 56.9
Revised National School Lunch Program and
School Breakfast Program Recommendations
Eating Out
The number of meals eaten outside the home or brought home from takeout
restaurants has increased in all age-groups of the U.S. population. The increased
convenience of this meal pattern is undermined by the generally lower
nutritional value of the meals, compared to home-cooked meals. Typically, meals
consumed or purchased in fast-food or casual restaurants are of large portion
size, are dense in calories, and contain large amounts of saturated fat, salt, and
sugar and low amounts of whole grains, fruits, and vegetables. Although still a
problem currently, trans fat is being phased out of most commercial restaurants
and prepared foods. Although an increasing number of restaurants offer healthier
alternatives, the vast majority of what is consumed at restaurants does not fit
MyPlate recommendations.
With increasing age, an increasing number of meals and snacks are also
consumed during peer social gatherings at friends' houses and parties. When a
large part of a child's or adolescent's diet is consumed on these occasions, the
diet quality can suffer, because food offerings are typically of low nutritional
value. Parents and pediatricians need to guide teens in navigating these
occasions while maintaining a healthful diet and enjoying meaningful social
interactions. These occasions often are also opportunities for teens to consume
alcohol; consequently, adult supervision is important.
developing regions except sub-Saharan Africa achieved the target to halve the
proportion of people living in extreme poverty, with the proportion falling from
47% in 1990 to 14% in 2015. Reductions in hunger were broadly consistent with
those of poverty reduction, and rates of undernourishment in developing regions
fell from 23% in 1990 to 13% in 2015. The prevalence of underweight children
(another MDG indicator of “hunger”) fell from 29% in 1990 to 15% in 2015 for
the developing regions combined. Rural children are almost twice as likely to be
underweight as urban children, and the poorest quintile is almost 3 times as
likely to be underweight as the richest quintile.
Eradicating poverty and hunger continue to be core targets of the Sustainable
Development Goals , as agreed by 193 countries of the United Nations General
Assembly in September 2015, and are to be achieved by 2030. In addition, in
2012 the World Health Assembly agreed to 6 global nutrition targets to be
reached by 2025, measured against a 2010 baseline, and the United Nations
Secretary-General launched the Zero Hunger Challenge with 5 objectives that
“would boost economic growth, reduce poverty and safeguard the environment”
and “would foster peace and stability” (Table 57.1 ).
Table 57.1
Global Food Security and Nutrition Targets
Measurement of Undernutrition
The term malnutrition encompasses both ends of the nutrition spectrum, from
undernutrition to overweight. Many poor nutritional outcomes begin in utero and
are manifest as low birthweight (LBW, <2,500 g). Preterm delivery and fetal
growth restriction are the 2 main causes of LBW, with prematurity relatively
more common in richer countries and fetal growth restriction relatively more
common in poorer countries.
Nutritional status is often assessed in terms of anthropometry (Table 57.2 ).
International standards of normal child growth under optimum conditions from
birth to 5 yr have been established by the World Health Organization (WHO). To
compile the standards, longitudinal data from birth to 24 mo of healthy,
breastfed, term infants were combined with cross-sectional measurements of
children ages 18-71 mo. The standards allow normalization of anthropometric
measures in terms of z scores (standard deviation [SD] scores). A z-score is the
child's height (weight) minus the median height (weight) for the child's age and
sex divided by the relevant SD. The standards are applicable to all children
everywhere, having been derived from a large, multicountry study reflecting
diverse ethnic backgrounds and cultural settings.
Table 57.2
Classification of Undernutrition
CLASSIFICATION INDEX GRADING
Gomez (underweight) 90–75% of median weight-for-age Grade 1 (mild)
75-60% Grade 2 (moderate)
<60% Grade 3 (severe)
Waterlow (wasting) 90–80% of median weight-for-height Mild
80–70% Moderate
<70% Severe
Waterlow (stunting) 95–90% of median height-for-age Mild
90–85% Moderate
<85% Severe
WHO (wasting) < −2 to > −3 SD weight-for-height Moderate
< −3 Severe
WHO (stunting) < −2 to > −3 SD height-for-age Moderate
<−3 Severe
WHO (wasting) (for age-group 6-59 mo) 115-125 mm mid-upper arm circumference Moderate
<115 mm Severe
SD, Standard deviation; WHO, World Health Organization.
FIG. 57.4 Measuring mid-upper arm circumference. (Courtesy of Nyani
Quarmyne/Panos Pictures.)
The risk of child death from infectious diseases increases even with mild
undernutrition, and as the severity of undernutrition increases, the risk increases
exponentially (Table 57.4 ). Undernutrition impairs immune function and other
host defenses; consequently, childhood infections are more severe and longer-
lasting in undernourished children and more likely to be fatal than the same
illnesses in well-nourished children. Infections can adversely affect nutritional
status, and young children can quickly enter a cycle of repeated infections and
ever-worsening malnutrition. Even for the survivors, physical and cognitive
damage as a result of undernutrition can impact their future health and economic
well-being. For girls, the cycle of undernutrition is passed on to the next
generation when undernourished women give birth to LBW babies.
Table 57.4
Hazard Ratios for All-Cause and Cause-Specific Deaths
Associated With Stunting, Wasting, and Underweight in
Children <5 yr
DEATHS
STANDARD DEVIATION (SD) SCORE
All Pneumonia Diarrhea Measles Other Infections
Height/length-for-age
< −3 5.5 6.4 6.3 6.0 3.0
−3 to < −2 2.3 2.2 2.4 2.8 1.9
−2 to < −1 1.5 1.6 1.7 1.3 0.9
≥ −1 1.0 1.0 1.0 1.0 1.0
Weight-for-length
< −3 11.6 9.7 12.3 9.6 11.2
−3 to < −2 3.4 4.7 3.4 2.6 2.7
−2 to < −1 1.6 1.9 1.6 1.0 1.7
≥ −1 1.0 1.0 1.0 1.0 1.0
Weight-for-age
< −3 9.4 10.1 11.6 7.7 8.3
−3 to < −2 2.6 3.1 2.9 3.1 1.6
−2 to < −1 1.5 1.9 1.7 1.0 1.5
≥ −1 1.0 1.0 1.0 1.0 1.0
From Black RE, Victora CG, Walker SP, et al: Maternal and child undernutrition and overweight in
low- and middle-income countries, Lancet 382:427–451, 2013.
Table 57.5
Examples of Nutrition-Specific and Nutrition-Sensitive
Interventions
Table 57.6
Table 57.7
Emergency Treatment in Severe Malnutrition
Stabilization
Table 57.8 summarizes the therapeutic directives for stabilization steps 1-7 (Fig.
57.7 ). Giving broad-spectrum antibiotics (Table 57.9 ) and feeding frequent
small amounts of F75 (a specially formulated low-lactose milk with 75 kcal and
0.9 g protein per 100 mL to which potassium, magnesium, and micronutrients
are added), will reestablish metabolic control, treat edema, and restore appetite.
The parenteral route should be avoided; children who lack appetite should be fed
by nasogastric tube, because nutrients delivered within the gut lumen help in its
repair. Table 57.10 provides recipes for preparing the special feeds and their
nutrient composition. Of the 2 recipes for F75, one requires no cooking, and the
other is cereal based and has a lower osmolality, which may benefit children
with persistent diarrhea. F75 is also available commercially; maltodextrins
replace some of the sugar, and potassium, magnesium, minerals, and vitamins
are already added.
Table 57.8
Therapeutic Directives for Stabilization of Malnourished
Children
STEP PREVENTION TREATMENT
1. Prevent/treat Avoid long gaps without food If conscious:
hypoglycemia and minimize need for 1. Give 10% glucose (50 mL), or a feed (see step 7), or
blood glucose <3 glucose: 1 tsp sugar under tongue, whichever is quickest.
mmol/L. 1. Feed immediately.
2. Feed every 2 hr for at least 1st day. Initially give
2. Feed every 3 hr day and
of feed every 30 min.
night (2 hr if ill).
3. Keep warm.
3. Feed on time.
4. Start broad-spectrum antibiotics.
4. Keep warm.
If unconscious:
5. Treat infections (they
1. Immediately give sterile 10% glucose (5 mL/kg)
compete for glucose).
rapidly by IV.
Note: Hypoglycemia and
hypothermia often coexist and 2. Feed every 2 hr for at least 1st day. Initially give
are signs of severe infection. of feed every 30 min. Use nasogastric (NG) tube if
unable to drink.
3. Keep warm.
4. Start broad-spectrum antibiotics.
2. Prevent/treat Keep warm and dry and feed Actively rewarm.
hypothermia frequently. 1. Feed.
axillary <35°C 1. Avoid exposure. 2. Skin-to-skin contact with caregiver (“kangaroo
(95°F); rectal 2. Dress warmly, including technique”) or dress in warmed clothes, cover
<35.5°C (95.9°F). head and cover with head, wrap in warmed blanket and provide indirect
blanket. heat (e.g., heater; transwarmer mattress;
3. Keep room hot; avoid incandescent lamp).
drafts. 3. Monitor temperature hourly (or every 30 min if
4. Change wet clothes and using heater).
bedding. 4. Stop rewarming when rectal temperature is 36.5°C
5. Do not bathe if very ill. (97.7°F).
6. Feed frequently day and
night.
7. Treat infections.
3. Prevent/treat Replace stool losses. Do not give IV fluids unless the child is in shock.
dehydration. 1. Give ReSoMal after each 1. Give ReSoMal 5 mL/kg every 30 min for 1st 2 hr
watery stool. ReSoMal orally or NG tube.
(37.5 mmol Na/L) is a low- 2. Then give 5-10 mL/kg in alternate hours for up to
sodium rehydration solution 10 hr. Amount depends on stool loss and eagerness
for malnutrition. to drink. Feed in the other alternate hour.
3. Monitor hourly and stop if signs of overload
develop (pulse rate increases by 25 beats/min and
respiratory rate by 5 breaths/min; increasing
edema; engorged jugular veins).
4. Stop when rehydrated (≥3 signs of hydration: less
thirsty, passing urine, skin pinch less slow, eyes
less sunken, moist mouth, tears, less lethargic,
improved pulse and respiratory rate).
4. Correct electrolyte 1. Give extra potassium (4 mmol/kg/day) and
imbalance—deficit magnesium (0.6 mmol/kg/day) for at least 2 wk (see
of potassium and Table 57.12 ).
magnesium, excess Note: Potassium and magnesium are already added
sodium. in Nutriset F75 and F100 packets.
5. Prevent/treat Minimize risk of cross-infection. Infections are often silent. Starting on 1st day, give
infections. 1. Avoid overcrowding. broad-spectrum antibiotics to all children.
2. Wash hands. 1. For antibiotic choices/schedule, see Table 57.9 .
3. Give measles vaccine to 2. Ensure all doses are given, and given on time.
unimmunized children age 3. Cover skin lesions so that they do not become
>6 mo. infected.
Note: Avoid steroids because they depress immune
function.
6. Correct Note: Folic acid, multivitamins, Do not give iron in the stabilization phase.
micronutrient zinc, copper, and other trace 1. Give vitamin A on day 1 (<6 mo 50,000 units; 6-
deficiencies. minerals are already added in 12 mo 100,000 units; >12 mo 200,000 units) if
Nutriset F75 and F100 packets. child has any eye signs of vitamin A deficiency or
has had recent measles. Repeat this dose on days 2
and 14.
2. Give folic acid, 1 mg (5 mg on day 1).
3. Give zinc (2 mg/kg/day) and copper (0.3
mg/kg/day). These are in the electrolyte/mineral
solution and Combined Mineral Vitamin mix
(CMV) and can be added to feeds and ReSoMal.
4. Give multivitamin syrup or CMV.
7. Start cautious 1. Give 8-12 small feeds of F75 to provide 130
feeding. mL/kg/day, 100 kcal/kg/day, and 1-1.5 g
protein/kg/day.
2. If gross edema, reduce volume to 100 mL/kg/day.
3. Keep a 24-hr intake chart. Measure feeds carefully.
Record leftovers.
4. If child has poor appetite, coax and encourage to
finish the feed. If unfinished, reoffer later. Use NG
tube if eating ≤80% of the amount offered.
5. If breastfed, encourage continued breastfeeding but
also give F75.
6. Transfer to F100 when appetite returns (usually
within 1 wk) and edema has been lost or is reduced.
7. Weigh daily and plot weight.
Table 57.9
Table 57.10
Recipes for Milk Formulas F75 and F100
F75 c
F75 b F100 d
(STARTER)
(STARTER) (CATCH-UP)
(CEREAL-BASED)
Dried skimmed milk (g) 25 25 80
Sugar (g) 100 70 50
Cereal flour (g) — 35 —
Vegetable oil (g) 30 30 60
Electrolyte/mineral solution (mL) a 20 20 20
Water: make up to (mL) 1000 1000 1000
Content/100 mL
Energy (kcal) 75 75 100
Protein (g) 0.9 1.1 2.9
Lactose (g) 1.3 1.3 4.2
Potassium (mmol) 4.0 4.2 6.3
Sodium (mmol) 0.6 0.6 1.9
Magnesium (mmol) 0.43 0.46 0.73
Zinc (mg) 2.0 2.0 2.3
Copper (mg) 0.25 0.25 0.25
% Energy from protein 5 6 12
% Energy from fat 32 32 53
Osmolality (mOsm/L) 413 334 419
a See Table 57.12 for recipe, or use commercially available therapeutic Combined Mineral Vitamin
mix (CMV).
b A comparable F75 can be made from 35 g dried whole milk, 100 g sugar, 20 g oil, 20 mL
electrolyte/mineral solution, and water to 1000 mL; or from 300 mL full-cream cow's milk, 100 g
sugar, 20 g oil, 20 mL electrolyte/mineral solution, and water to 1000 mL.
c This lower-osmolality formula may be helpful for children with dysentery or persistent diarrhea.
electrolyte/mineral solution, and water to 1000 mL; or from 880 mL full-cream cow's milk, 75 g
sugar, 20 g oil, 20 mL electrolyte/mineral solution, and water to 1000 mL.
Whisk at high speed to prevent oil from separating out.
Table 57.11
Table 57.12
Rehabilitation
The signals for entry to the rehabilitation phase are reduced or minimal edema
and return of appetite.
A controlled transition over 3 days is recommended to prevent refeeding
syndrome (see Chapter 58 ). After the transition, unlimited amounts should be
given of a high-energy, high-protein milk formula such as F100 (100 kcal and 3
g protein per 100 mL), or a ready-to-use therapeutic food (RUTF) , or family
foods modified to have comparable energy and protein contents.
To make the transition, for 2 days replace F75 with an equal volume of F100,
then increase each successive feed by 10 mL until some feed remains uneaten
(usually at about 200 mL/kg/day). After this transition, give 150-220 kcal/kg/day
and 4-6 g protein/kg/day, and continue to give potassium, magnesium, and
micronutrients. Add iron (3 mg/kg/day). If breastfed, encourage continued
breastfeeding. Children with severe malnutrition have developmental delays, so
loving care, structured play, and sensory stimulation during and after treatment
are essential to aid recovery of brain function.
Community-Based Treatment
Many children with severe acute malnutrition can be identified in their
communities before medical complications arise. If these children have a good
appetite and are clinically well, they can be rehabilitated at home through
community-based therapeutic care, which has the added benefit of reducing their
exposure to nosocomial infections and providing continuity of care after
recovery. It also reduces the time caregivers spend away from home and their
opportunity costs and can be cost-effective for health services.
Fig. 57.8 shows the criteria for inpatient and outpatient care. To maximize
coverage and compliance, community-based therapeutic care has 4 main
CHAPTER 58
Refeeding Syndrome
Robert M. Kliegman
The refeeding syndrome may occur if high-energy feeding is started too soon or
too vigorously, and it may lead to sudden death with signs of heart failure. Early
accounts of the syndrome were among starved survivors of wartime sieges and
concentration camps and among prisoners of war when given sudden access to
unlimited food. The refeeding syndrome occurs in malnourished individuals as a
result of untimely, overzealous oral, enteral, or parenteral (highest risk) feeding,
and the risk is not widely recognized. Refeeding syndrome has also been seen
among malnourished patients with anorexia nervosa with a body mass index
(BMI) <70% median values. Onset is usually 24-48 hr after the start of high-
energy feeding and is characterized by breathlessness, rapid pulse, increased
venous pressure, rapid enlargement of the liver, and watery diarrhea. Other
features are noted in Table 58.1 .
Table 58.1
Clinical Signs and Symptoms of Refeeding Syndrome
VITAMIN/THIAMINE SODIUM
HYPOPHOSPHATEMIA HYPOKALEMIA HYPOMAGNESEMIA
DEFICIENCY RETENTION
Cardiac Cardiac Cardiac Encephalopathy Fluid overload
Hypotension Arrhythmias Arrhythmias Lactic acidosis Pulmonary
Decreased stroke volume Respiratory Neurologic Death edema
Respiratory Failure Weakness Cardiac
Impaired diaphragm Neurologic Tremor compromise
contractility Weakness Tetany
Dyspnea Paralysis Seizures
Respiratory failure Gastrointestinal Altered mental status
Neurologic Nausea Coma
Paresthesia Vomiting Gastrointestinal
Weakness Constipation Nausea
Confusion Muscular Vomiting
Disorientation Rhabdomyolysis Diarrhea
Lethargy Muscle necrosis Other
Areflexic paralysis Other Refractory hypokalemia
Seizures Death and hypocalcemia
Coma Death
Hematologic
Leukocyte dysfunction
Hemolysis
Thrombocytopenia
Other
Death
Data from Kraft MD, Btaiche IF, Sacks GS: Review of RFS, Nutr Clin Pract 20:625–633, 2005.
From Fuentebella J, Kerner JA: Refeeding syndrome, Pediatr Clin North Am 56:1201–1210, 2009.
Bibliography
Friedli N, Stanga Z, Sobotka L, et al. Revisiting the refeeding
syndrome: results of a systematic review. Nutrition .
2017;31:151–160.
Pulani CD, Zettle S, Srinath A. Refeed syndrome. Pediatr Rev .
(WHO) charts for children up to 2 yr of age who are measured supine for length.
The CDC 2000 growth charts are recommended for children and adolescents
(age 2-20 yr) when measured with a standing height. The severity of
malnutrition (mild, moderate, or severe) may be determined by plotting the z
score (standard deviation [SD] from the mean) for each of these anthropometric
values (Table 59.1 ).
Table 59.1
Comprehensive Malnutrition Indicators
SEVERE MODERATE MILD
INDICATORS*
MALNUTRITION MALNUTRITION MALNUTRITION
Weight-for-length z score ≥ −3 z score or worse −2.0 to 2.99 z score −1.0 to −1.99 z score †
BMI-for-age z score ≥ −3 z score or worse −2.0 to 2.99 z score −1.0 to −1.99 z score †
Weight-for-length/height z score ≥ −3 z score or worse No data available No data available
Mid-upper arm circumference ≥ −3 z score or worse −2.0 to 2.99 z score −1.0 to −1.99 z score
(<5 yr of age)
Weight gain velocity (≤2 yr of ≤25% of norm 26–50% of norm 51–75% of the norm
age)
Weight loss (2-20 yr of age) >10% of UBW >7.5% UBW >5% UBW
Deceleration in weight-for- Deceleration across 3 z Deceleration across 2 z Deceleration across 1 z
length/height or BMI-for-age score lines score lines score line
Inadequate nutrient intake ≤25% of estimated 26–50% of estimated 51–75% of estimated
energy − protein need energy − protein need energy − protein need
* It is recommended that when a child meets more than one malnutrition acuity level, the provider
should document the severity of the malnutrition at the highest acuity level to ensure that an
appropriate treatment plan and appropriate intervention, monitoring, and evaluation are provided.
† Needs additional positive diagnostic criteria to make a malnutrition diagnosis.
Table 59.2
Table 59.3
Findings in Failure to Thrive in Infancy
SYSTEM-
APPROXIMATE
SYSTEM-SPECIFIC PHYSICAL SPECIFIC
CAUSE PERCENTAGE HISTORY
FINDINGS LABORATORY
OF ALL CASES
STUDIES
Psychosocial Up to 50% or Vague, None, may have soft neurologic signs None
more inconsistent
feeding history,
history of bottle
propping
CNS 13% Poor feeding, Grossly abnormal neurologic findings Frequent gross
gross abnormalities on
developmental EEG and MRI
delay, vomiting scan or grossly
abnormal tests of
neuromuscular
function
Gastrointestinal 10% Chronic Often negative, may have abdominal Abnormal
vomiting and/or distention barium, pH
diarrhea, probe, or
abnormal stools, endoscopic
crying with study; abnormal
feedings, stool findings
nocturnal (pH, reducing
cough/snoring substances, fat
stain, Wright
stain)
Cardiac 9% Slow feeding, Often cyanotic or have signs of Abnormal
dyspnea and congestive heart failure echocardiogram,
diaphoresis with ECG,
feeding, catheterization
restlessness and findings
diaphoresis
during sleep
Genetic 8% May have Often have facies typical of a May have typical
positive family syndrome, skeletal abnormalities, radiographic
history or a neurologic abnormalities, or findings,
history of visceromegaly chromosomal
developmental abnormalities,
delay abnormal
metabolic
screens
Pulmonary 3.5% Chronic or Grossly abnormal chest examination Abnormal chest
recurrent findings radiographs
dyspnea with
feedings,
tachypnea
Renal 3.5% May be Often negative, may have flank masses Abnormal
negative or may urinalysis,
have history of frequently
polyuria elevated BUN
and creatinine,
signs of renal
osteodystrophy
on radiographs
Endocrine 3.5% With With hypothyroidism, no wasting but Decreased T4 ,
hypothyroidism, mottling, umbilical hernia, often open increased TSH;
constipation and posterior fontanelle. With diabetes, glucosuria and
decreased often without specific abnormality, but hyperglycemia;
activity level; may have signs of dehydration, ketotic abnormal
with diabetes, breath, and hyperpnea. With pituitary function
polyuria, hypopituitarism and isolated growth study results
polydipsia hormone deficiency, growth normal
until 9 mo or later, then plateaus, but
normal weight for height; delayed tooth
eruption
BUN, Blood urea nitrogen; CNS, central nervous system; CT, computed tomography; ECG,
electrocardiogram; EEG, electroencephalogram; T4 , thyroxine; TSH, thyroid-stimulating hormone.
From Carrasco MM, Wolford JE: Child abuse and neglect. In Zitelli BJ, McIntire SC, Nowalk AJ,
editors: Atlas of pediatric physical diagnosis, ed 7, Philadelphia, 2018, Elsevier, Table 6.6.
Additional measurements that are useful for following the progress of the
acutely malnourished child are mid-upper arm circumference (MUAC) and
hand-grip strength. MUAC is a particularly useful anthropometric measure when
weight may be distorted by use of corticosteroids or fluid status (e.g., ascites,
edema).
For children 6 yr and older, hand-grip strength may be a more acute
proopiomelanocortin (POMC) deficiency, a prohormone precursor of
adrenocorticotropic hormone (ACTH) and melanocyte-stimulating hormone
(MSH), resulting in adrenal insufficiency, light skin, hyperphagia, and obesity.
Table 60.1
Endocrine and Genetic Causes of Obesity
Comorbidities
Complications of pediatric obesity occur during childhood and adolescence and
persist into adulthood. An important reason to prevent and treat pediatric obesity
is the increased risk for morbidity and mortality later in life. The Harvard
Growth Study found that boys who were overweight during adolescence were
twice as likely to die from CV disease as those who had normal weight. More
immediate comorbidities include type 2 diabetes, hypertension, hyperlipidemia,
and nonalcoholic fatty liver disease (NAFLD) (Table 60.2 ). Insulin resistance
increases with increasing adiposity and independently affects lipid metabolism
and CV health. The metabolic syndrome (central obesity, hypertension, glucose
intolerance, and hyperlipidemia) increases risk for CV morbidity and mortality.
NAFLD has been reported in 34% of patients treated in pediatric obesity clinic.
NAFLD is now the most common chronic liver disease in U.S. children and
adolescents. It can present with advanced fibrosis or nonalcoholic steatohepatitis
and may result in cirrhosis and hepatocellular carcinoma. Insulin resistance is
often associated. Furthermore, NAFLD is independently associated with
increased risk of CV disease.
Table 60.2
Obesity-Associated Comorbidities
Table 60.4
Intervention
Evidence shows that some interventions result in modest but significant and
sustained improvement in body mass. Based on behavior change theories,
treatment includes specifying target behaviors , self-monitoring , goal setting ,
stimulus control , and promotion of self-efficacy and self-management skills.
Behavior changes associated with improving BMI include drinking lower
quantities of sugar-sweetened beverages, consuming higher-quality diets,
increasing exercise, decreasing screen time, and self-weighing. Most successful
interventions have been family based and consider the child's developmental age.
“Parent-only” treatment can be as effective as “parent–child” treatment. Because
obesity is multifactorial, not all children and adolescents will respond to the
same approach. For example, loss-of-control eating, associated with weight gain
and obesity, predicts poor outcome in response to family-based treatment.
Furthermore, clinical treatment programs are expensive and not widely
available. Therefore, interest has grown in novel approaches such as internet-
based treatments and guided self-help.
It is important to begin with clear recommendations about appropriate
caloric intake for the obese child (Table 60.5 ). Working with a dietitian is
essential. Meals should be based on fruits, vegetables, whole grains, lean meat,
fish, and poultry. Prepared foods should be chosen for their nutritional value,
with attention to calories and fat. Foods that provide excessive calories and low
nutritional value should be reserved for infrequent treats.
Table 60.5
Recommended Caloric Intake Designated by Age and
Gender
Weight reduction diets in adults generally do not lead to sustained weight loss.
Therefore the focus should be on changes that can be maintained for life.
Attention to eating patterns is helpful. Families should be encouraged to plan
family meals, including breakfast. It is almost impossible for a child to make
changes in nutritional intake and eating patterns if other family members do not
make the same changes. Dietary needs also change developmentally; adolescents
require greatly increased calories during their growth spurts, and adults who lead
inactive lives need fewer calories than active, growing children.
Psychologic strategies are helpful. The “traffic light” diet groups foods into
those that can be consumed without any limitations (green), in moderation
(yellow), or reserved for infrequent treats (red) (Table 60.6 ). The concrete
categories are very helpful to children and families. This approach can be
adapted to any ethnic group or regional cuisine. Motivational interviewing
begins with assessing how ready the patient is to make important behavioral
changes. The professional then engages the patient in developing a strategy to
take the next step toward the ultimate goal of healthy nutritional intake. This
method allows the professional to take the role of a coach, helping the child and
family reach their goals. Other behavioral approaches include family rules about
where food may be consumed (e.g., “not in the bedroom”).
Table 60.6
“Traffic Light” Diet Plan
YELLOW LIGHT
FEATURE GREEN LIGHT FOODS RED LIGHT FOODS
FOODS
Quality Low-calorie, high-fiber, low- Nutrient-dense, but higher in High in calories, sugar, and fat
fat, nutrient-dense calories and fat
Types of Fruits, vegetables Lean meats, dairy, starches, Fatty meats, sugar, sugar-sweetened
food grains beverages, fried foods
Quantity Unlimited Limited Infrequent or avoided
Table 60.7
Medications for Weight Management With Mechanism of
Action, Availability, and Dosing
AVAILABLE MEAN
FOR PERCENTAGE
MECHANISM CHRONIC USE WEIGHT LOSS
MEDICATION ADVANTAGES DISADVANTAGES
OF ACTION
European
USA Placebo Drug
Union
Phentermine, 15-30 mg Sympathomimetic For No Not Not stated Inexpensive Side effect profile;
PO short- stated in in label no long-term data
term label
use
Orlistat, 120 mg PO tid Pancreatic lipase Yes Yes −2.6% † −6.1% † Not absorbed; Modest weight loss;
before meals inhibitor long-term data* side effect profile
Lorcaserin, 10 mg PO 5-HT2c serotonin Yes No −2.5% −5.8% Mild side Expensive; modest
bid agonist with little effects; long- weight loss
affinity for other term data*
serotonergic
receptors
Phentermine/topiramate Sympathomimetic Yes No −1.2% −7.8% Robust weight Expensive; teratogen
ER, 7.5 mg/46 mg or anticonvulsant (mid- loss; long-term
15 mg/92 mg PO (GABA receptor dose) data*
indicated as rescue modulation, −9.8%
(requires titration) carbonic (full
anhydrase dose)
inhibition,
glutamate
antagonism)
Naltrexone Opioid receptor Yes Yes −1.3% −5.4% Reduces food Moderately
SR/bupropion SR, 32 antagonist; craving; long- expensive; side
mg/360 mg PO dopamine and term data* effect profile
(requires titration) noradrenaline
reuptake inhibitor
Liraglutide, 3.0 mg GLP-1 receptor Yes Yes −3% −7.4% Side effect Expensive;
injection (requires agonist (full dose) profile; long- injectable
titration) term data*
* Data from randomized controlled trials lasting >52 wk.
†
Assuming the average patient in the orlistat and placebo groups weighed 100 kg at baseline.
Information is from U.S. product labels, except where noted. The data supporting these tables are
derived from the prescribing information labeling approved by the US Food and Drug
Administration.
ER, Extended release; SR, sustained release; PO, orally; bid, twice daily; tid, 3 times daily.
Adapted from Bray GA, Frühbeck G, Ryan DH, Wilding JPH: Management of obesity, Lancet
387:1947–1965, 2016, p 1950.
Breastfeeding: exclusive for 4-6 mo; continue with other foods for 12 mo.
Postpone introduction of baby foods to 4-6 mo and juices to 12 mo.
Families
Schools
Communities
Increase family-friendly exercise and safe play facilities for children of all
ages.
Develop more mixed residential-commercial developments for walkable
and bicyclable communities.
Discourage the use of elevators and moving walkways.
Provide information on how to shop and prepare healthier versions of
culture-specific foods.
Healthcare Providers
Industry
Adapted from Speiser PW, Rudolf MCJ, Anhalt H, et al: Consensus statement:
childhood obesity, J Clin Endocrinol Metab 90:1871–1887, 2005.
Table 60.9
Anticipatory Guidance: Establishing Healthy
Eating Habits in Children
Pediatric prevention efforts begin with careful monitoring of weight and BMI
percentiles at healthcare maintenance visits. Attention to changes in BMI
percentiles can alert the pediatric provider to increasing adiposity before the
child becomes overweight or obese. All families should be counseled about
healthy nutrition for their children, because the current prevalence of overweight
and obesity in adults is 65%. Therefore, approximately two thirds of all children
can be considered at risk for becoming overweight or obese at some time in their
lives. Those who have an obese parent are at increased risk. Prevention efforts
begin with promotion of exclusive breastfeeding for 6 mo and total breastfeeding
for 12 mo. Introduction of infant foods at 6 mo should focus on cereals, fruits,
and vegetables. Lean meats, poultry, and fish may be introduced later in the 1st
year of life. Parents should be specifically counseled to avoid introducing highly
sugared beverages and foods in the 1st year of life. Instead, they should expose
their infants and young children to a rich variety of fruits, vegetables, grains,
lean meats, poultry, and fish to facilitate acceptance of a diverse and healthy diet.
Parenting matters, and authoritative parents are more likely to have children
with a healthy weight than those who are authoritarian or permissive. Families
who eat regularly scheduled meals together are less likely to have overweight or
obese children. Child health professionals can address a child's nutritional status
and provide expertise in child growth and development.
physiologic processes are sensitive to a deficiency or excess of vitamin A or RA,
including reproduction, growth, bone development, and the functions of the
respiratory, gastrointestinal, hematopoietic, and immune systems. Vitamin A,
presumably by enhancing immune function and host defense, is particularly
important in developing countries; studies show that vitamin A supplementation
or therapy reduces morbidity and mortality from various infectious diseases,
including measles (see Chapter 273 ).
Vitamin A plays a critical role in vision, mediated by 11-cis retinal. The
human retina contains 2 distinct photoreceptor systems: the rods, in which
rhodopsin senses light of low-intensity, and the cones, in which iodopsins detect
different colors; 11-cis -retinal is the prosthetic group on both these visual
proteins. The mechanism of vitamin A action is similar for rods and cones, based
on photoisomerization of 11-cis to all-trans retinal (change shape when exposed
to light), which initiates signal transduction via the optic nerve to the brain,
resulting in visual sensation. After isomerization (also known as
photobleaching), a series of reactions serves to regenerate the 11-cis retinal for
resynthesis of rhodopsin and iodopsin; accessory cells, including retinal pigment
epithelium and Müller cells, are involved in this recycling process.
Vitamin A Deficiency
If the growing child has a well-balanced diet and obtains vitamin A from foods
rich in vitamin A or provitamin A (Table 61.1 ), the risk of vitamin A deficiency
is small. However, even subclinical vitamin A deficiency can have serious
consequences.
Table 61.1
Vitamin A Characteristics
UPPER LEVEL
RECOMMENDED DIETARY
AGE (UL) (µg retinol
ALLOWANCE (RDA) (µg COMMENTS
RANGE equivalents per
retinol equivalents per day)
day)
0-6 mo 400 600 The recommended intake for infants is an
7-12 mo 500 600 adequate intake, based on the amount of
vitamin A normally present in breast milk.
1-3 yr 300 600 The UL applies only to preformed vitamin A
4-8 yr 400 900 (retinol).
9-13 yr 600 1,700
14-18 yr 900, male; 700, female 2,800
Diagnosis
Most often, the diagnosis is based on the clinical features of angular cheilosis in
a malnourished child, who responds promptly to riboflavin supplementation. A
functional test of riboflavin status is done by measuring the activity of
erythrocyte glutathione reductase (EGR), with and without the addition of FAD.
An EGR activity coefficient (ratio of EGR activity with added FAD to EGR
activity without FAD) of >1.4 is used as an indicator of deficiency. Urinary
excretion of riboflavin <30 µg/24 hr also suggests low intakes.
Prevention
Table 62.1 lists the recommended daily allowance of riboflavin for infants,
children, and adolescents. Adequate consumption of milk, milk products, and
eggs prevents riboflavin deficiency. Fortification of cereal products is helpful for
those who follow vegan diets or who are consuming inadequate amounts of milk
products for other reasons.
Table 62.1
Water-Soluble Vitamins
NAMES TREATMENT
EFFECTS OF CAUSES OF DIETARY
AND BIOCHEMICAL ACTION OF
DEFICIENCY DEFICIENCY SOURCES
SYNONYMS DEFICIENCY
Thiamine Coenzyme in Neurologic 3-5 mg/day PO Polished rice– Meat,
(vitamin B1 ) carbohydrate metabolism (dry beriberi): thiamine for 6 based diets especially
Nucleic acid synthesis irritability, wk Malabsorptive pork; fish;
Neurotransmitter peripheral states liver
synthesis neuritis, Severe Rice
muscle malnutrition (unmilled),
tenderness, Malignancies wheat
ataxia Alcoholism germ;
Cardiac (wet enriched
beriberi): cereals;
tachycardia, legumes
edema,
cardiomegaly,
cardiac failure
Niacin Constituent of NAD and Pellagra 50-300 mg/day Predominantly Meat, fish,
(vitamin B3 ) NADP, important in manifesting as PO niacin maize-based poultry
respiratory chain, fatty acid diarrhea, diets Cereals,
synthesis, cell differentiation, symmetric scaly Anorexia legumes,
and DNA processing dermatitis in sun- nervosa green
exposed areas, and Carcinoid vegetables
neurologic syndrome
symptoms of
disorientation and
delirium
Biotin Cofactor for carboxylases, Scaly periorificial 1-10 mg/day Consumption Liver, organ
important in gluconeogenesis, dermatitis, PO biotin of raw eggs for meats, fruits
fatty acid and amino acid conjunctivitis, prolonged
metabolism alopecia, lethargy, periods
hypotonia, and Parenteral
withdrawn nutrition with
behavior infusates
lacking biotin
Valproate
therapy
Pantothenic Component of coenzyme A Experimentally Isolated deficiency Beef, organ
acid (vitamin and acyl carrier protein produced extremely rare in meats,
B5 ) involved in fatty acid deficiency in humans poultry,
metabolism humans: seafood,
irritability, fatigue, egg yolk
numbness, Yeast,
paresthesias soybeans,
(burning feet mushrooms
syndrome),
muscle cramps
Folic acid Coenzymes in amino acid and Megaloblastic 0.5-1 mg/day Malnutrition Enriched
nucleotide metabolism as an anemia PO folic acid Malabsorptive cereals, beans,
acceptor and donor of 1- Growth states leafy
carbon units retardation, Malignancies vegetables,
glossitis Hemolytic citrus fruits,
Neural tube anemias papaya
defects in Anticonvulsant
progeny therapy
*
For healthy breastfed infants, the values represent adequate intakes, that is, the mean intake of
apparently “normal” infants.
PO, Orally, IM, intramuscularly; IV, intravenously; INH, isoniazid; NAD, nicotinamide adenine
dinucleotide; NADP, nicotinamide adenine dinucleotide phosphate; OCP, oral contraceptive pill;
RDA, recommended dietary allowance.
From Dietary reference intakes (DRIs): Recommended dietary allowances and adequate intakes,
vitamins, Food and Nutrition Board, Institute of Medicine, National Academies.
http://www.nationalacademies.org/hmd/~/media/Files/Activity%20Files/Nutrition/DRI-
Tables/2_%20RDA%20and%20AI%20Values_Vitamin%20and%20Elements.pdf?la=en .
Treatment
Treatment includes oral administration of 3-10 mg/day of riboflavin, often as an
ingredient of a vitamin B–complex mix. The child should also be given a well-
balanced diet, including milk and milk products.
Riboflavin Toxicity
No adverse effects associated with riboflavin intakes from food or supplements
have been reported, and the upper safe limit for consumption has not been
established. Although the photosensitizing property of vitamin B2 suggests some
potential risks, limited absorption in high-intake situations precludes such
CHAPTER 64
Rickets
Bone consists of a protein matrix called osteoid and a mineral phase, principally
composed of calcium and phosphate, mostly in the form of hydroxyapatite .
Osteomalacia occurs with inadequate mineralization of bone osteoid in children
and adults. Rickets is a disease of growing bone caused by unmineralized matrix
at the growth plates in children only before fusion of the epiphyses. Because
growth plate cartilage and osteoid continue to expand but mineralization is
inadequate, the growth plate thickens. Circumference of the growth plate and
metaphysis is also greater, increasing bone width at the growth plates and
causing classic clinical manifestations, such as widening of the wrists and
ankles. The general softening of the bones causes them to bend easily when
subject to forces such as weight bearing or muscle pull. This softening leads to a
variety of bone deformities.
Rickets is principally caused by vitamin D deficiency and was rampant in
northern Europe and the United States during the early years of the 20th century.
Although largely corrected through public health measures that provided
children with adequate vitamin D, rickets remains a persistent problem in
developed countries, with many cases still secondary to preventable nutritional
vitamin D deficiency. It remains a significant problem in developing countries
and may be secondary to nutritional vitamin D deficiency and inadequate intake
of calcium (Table 64.1 ).
Table 64.1
Physical and Metabolic Properties and Food Sources of
Vitamins D, E, and K
NAMES AND BIOCHEMICAL EFFECTS OF EFFECTS OF
CHARACTERISTICS SOURCES
SYNONYMS ACTION DEFICIENCY EXCESS
VITAMIN D
Vitamin D3 (3- Fat-soluble, stable to Necessary for GI Rickets in Hypercalcemia, Exposure to
cholecalciferol), heat, acid, alkali, and absorption of growing which can sunlight
which is oxidation; bile calcium; also children; cause emesis, (UV light);
synthesized in the necessary for increases osteomalacia; anorexia, fish oils,
skin, and vitamin absorption; absorption of hypocalcemia pancreatitis, fatty fish,
D2 (from plants or hydroxylation in the phosphate; direct can cause hypertension, egg yolks,
yeast) are liver and kidney actions on bone, tetany and arrhythmias, and vitamin
biologically necessary for biologic including seizures CNS effects, D–fortified
equivalent; 1 µg = activity mediating polyuria, formula,
40 IU vitamin D. resorption nephrolithiasis, milk,
renal failure cereals,
bread
VITAMIN E
Group of related Fat-soluble; readily Antioxidant; Red cell Unknown Vegetable
compounds with oxidized by oxygen, protection of cell hemolysis in oils, seeds,
similar biologic iron, rancid fats; bile membranes from premature nuts, green
activities; α- acids necessary for lipid peroxidation infants; leafy
tocopherol is the absorption and formation of posterior vegetables,
most potent and free radicals column and margarine
most common form cerebellar
dysfunction;
pigmentary
retinopathy
VITAMIN K
Group of Natural compounds are Vitamin K– Hemorrhagic Not Green leafy
naphthoquinones fat-soluble; stable to dependent manifestations; established; vegetables,
with similar heat and reducing proteins include long-term bone analogs (no liver,
biologic activities; agents; labile to coagulation and vascular longer used) certain
K1 (phylloquinone) oxidizing agent, strong factors II, VII, IX, health caused legumes
from diet; K2 acids, alkali, light; bile and X; proteins C, hemolytic and plant
(menaquinones) salts necessary for S, Z; matrix Gla anemia, oils; widely
from intestinal intestinal absorption protein, jaundice, distributed
bacteria osteocalcin kernicterus,
death
CNS, Central nervous system; GI, gastrointestinal; UV, ultraviolet.
Etiology
There are many causes of rickets, including vitamin D disorders, calcium
deficiency, phosphorus deficiency, and distal renal tubular acidosis (Table 64.2 ).
Table 64.2
Causes of Rickets
Vitamin D Disorders
Calcium Deficiency
Low intake
Diet
Premature infants (rickets of prematurity)
Malabsorption
Primary disease
Dietary inhibitors of calcium absorption
Phosphorus Deficiency
Inadequate intake
Premature infants (rickets of prematurity)
Aluminum-containing antacids
Renal Losses
Clinical Manifestations
Most manifestations of rickets are a result of skeletal changes (Table 64.3 ).
Craniotabes is a softening of the cranial bones and can be detected by applying
pressure at the occiput or over the parietal bones. The sensation is similar to the
feel of pressing into a Ping-Pong ball and then releasing. Craniotabes may also
be secondary to osteogenesis imperfecta, hydrocephalus, and syphilis. It is a
normal finding in many newborns, especially near the suture lines, but typically
disappears within a few months of birth. Widening of the costochondral
junctions results in a rachitic “rosary ,” which feels like the beads of a rosary as
the examiner's fingers move along the costochondral junctions from rib to rib
(Fig. 64.1 ). Growth plate widening is also responsible for the enlargement at the
wrists and ankles (Fig. 64.2 ). The horizontal depression along the lower anterior
chest known as Harrison groove occurs from pulling of the softened ribs by the
diaphragm during inspiration. Softening of the ribs also impairs air movement
and predisposes patients to atelectasis and pneumonia. Valgus or varus
deformities of the legs are common; windswept deformity occurs when one leg
is in extreme valgus and the other is in extreme varus (Fig. 64.3 ).
Table 64.3
Clinical Features of Rickets
General
Head
Craniotabes
Frontal bossing
Delayed fontanel closure (usually closed by 2 yr)
Delayed dentition
No incisors by age 10 mo
No molars by age 18 mo
Caries
Craniosynostosis
Chest
Rachitic rosary
Harrison groove
Respiratory infections and atelectasis*
Back
Scoliosis
Kyphosis
Lordosis
Extremities
Hypocalcemic Symptoms †
Tetany
Seizures
Stridor caused by laryngeal spasm
* These features are most frequently associated with the vitamin D deficiency
disorders.
† These symptoms develop only in children with disorders that produce
FIG. 64.1 Rachitic “rosary” in a child with rickets. (Courtesy of Dr. Thomas
D. Thacher, Rochester, MN.)
FIG. 64.5 Radiographs of the knees in 7 yr old girl with distal renal tubular acidosis
and rickets. A, At initial presentation, there is widening of the growth plate and
metaphyseal fraying. B, Dramatic improvement after 4 mo of therapy with alkali.
Diagnosis
The diagnosis of rickets is based on the presence of classic radiographic
abnormalities. It is supported by physical examination findings, history, and
laboratory results consistent with a specific etiology (Table 64.4 ).
Table 64.4
Laboratory Findings in Various Disorders Causing Rickets
Clinical Evaluation
Because the majority of children with rickets have a nutritional deficiency, the
initial evaluation should focus on a dietary history, emphasizing intake of both
vitamin D and calcium. Most children in industrialized nations receive vitamin D
from formula, fortified milk, or vitamin supplements. Along with the amount,
the exact composition of the formula or milk is pertinent, because rickets has
occurred in children given products that are called “milk” (e.g., soy milk) but are
deficient in vitamin D and minerals.
Cutaneous synthesis mediated by sunlight exposure is an important source of
vitamin D. It is important to ask about time spent outside, sunscreen use, and
clothing, especially if there may be a cultural reason for increased covering of
the skin. Because winter sunlight is ineffective at stimulating cutaneous
synthesis of vitamin D, the season is an additional consideration. Children with
increased skin pigmentation are at increased risk for vitamin D deficiency
because of decreased cutaneous synthesis.
The presence of maternal risk factors for nutritional vitamin D deficiency,
including diet and sun exposure, is an important consideration when a neonate or
young infant has rachitic findings, especially if the infant is breastfed (Table 64.5
). Determining a child's intake of dairy products, the main dietary source of
calcium, provides a general sense of calcium intake. High dietary fiber can
interfere with calcium absorption.
Table 64.5
Risk Factors for Nutritional Rickets and
Osteomalacia and Their Prevention
Maternal Factors
Vitamin D deficiency
Dark skin pigmentation
Full body clothing cover
High latitude during winter/spring season
Other causes of restricted sun (UVB) exposure, e.g., predominant
indoor living, disability, pollution, cloud cover
Low–vitamin D diet
Low-calcium diet
Poverty, malnutrition, special diets
Infant/Childhood Factors
Preventive Measures
Sun exposure (UVB content of sunlight depends on latitude and season)
Vitamin D supplementation
Strategic fortification of the habitual food supply
Normal calcium intake
Table 67.1
Trace Elements
EFFECTS OF DIETARY
ELEMENT PHYSIOLOGY EFFECTS OF EXCESS
DEFICIENCY SOURCES
Chromium Potentiates the action of Impaired glucose Unknown Meat, grains,
insulin tolerance, peripheral fruits, and
neuropathy, and vegetables
encephalopathy
Copper Absorbed via Microcytic anemia, Acute: nausea, Vegetables,
specific intestinal osteoporosis, emesis, abdominal grains, nuts, liver,
transporter neutropenia, neurologic pain, coma, and margarine,
Circulates bound to symptoms, hepatic necrosis legumes, corn oil
ceruloplasmin depigmentation of hair Chronic toxicity
Enzyme cofactor and skin (liver and brain
(superoxide injury) occurs in
dismutase, Wilson disease (see
cytochrome oxidase, Chapter 384.2 ) and
and enzymes secondary to excess
involved in iron intake (see Chapter
metabolism and 384.3 ).
connective tissue
formation)
Fluoride Incorporated into bone Dental caries (see Chronic: dental fluorosis Toothpaste,
Chapter 338 ) (see Chapter 333 ) fluoridated water
Iodine Component of thyroid Hypothyroidism (see Hypothyroidism and Saltwater fish,
hormone (see Chapter Chapters 579 and 580 ) goiter (see Chapters 581 iodized salt
580 ) and 583 ); maternal
excess can cause
congenital
hypothyroidism and
goiter (see Chapter 584.1
).
Iron Component of Anemia (see Chapter Acute (see Chapter Meat,
hemoglobin, myoglobin, 482 ), decreased 77 ): nausea, fortified
cytochromes, and other alertness, impaired vomiting, diarrhea, foods
enzymes learning abdominal pain, and Deficiency
hypotension can also
Chronic excess result from
usually secondary to blood loss
hereditary disorders (hookworm
(see Chapter 489 ); infestation,
causes organ menorrhagia)
dysfunction.
Manganese Enzyme cofactor Hypercholesterolemia, Neurologic Nuts, meat,
weight loss, decreased manifestations, grains, tea
clotting proteins* cholestatic jaundice
Molybdenum Enzyme cofactor Tachycardia, tachypnea, Hyperuricemia and Legumes, grains,
(xanthine oxidase and night blindness, increased risk of gout liver
others) irritability, coma*
Selenium Enzyme cofactor Cardiomyopathy Nausea, diarrhea, Meat, seafood,
(prevents oxidative (Keshan disease ), neurologic whole grains,
damage) myopathy manifestations, nail and garlic
hair changes, garlic odor
Zinc Enzyme cofactor Decreased growth, Abdominal pain, Meat, shellfish,
Constituent of zinc- dermatitis of diarrhea, vomiting whole grains,
finger proteins, extremities and Can worsen copper legumes, cheese
which regulate gene around orifices, deficiency
transcription impaired immunity,
poor wound healing,
hypogonadism,
diarrhea
Supplements are
beneficial in
diarrhea and
improve
neurodevelopmental
outcomes.
* These deficiency states have been reported only in case reports associated with parenteral
nutrition or highly unusual diets.
angiotensin-aldosterone system and intrarenal mechanisms. In hyponatremia or
hypernatremia, the underlying pathophysiology determines the amount of
urinary Na+ , not the serum [Na+ ].
Hypernatremia
Hypernatremia is a [Na+ ] >145 mEq/L, although it is sometimes defined as
>150 mEq/L. Mild hypernatremia is fairly common in children, especially
among infants with gastroenteritis. Hypernatremia in hospitalized patients may
be iatrogenic—caused by inadequate water administration or, less often, by
excessive Na+ administration. Moderate or severe hypernatremia has significant
morbidity because of the underlying disease, the effects of hypernatremia on the
brain, and the risks of overly rapid correction.
Water Deficit
Nephrogenic Diabetes Insipidus
Acquired
X-linked (OMIM 304800)
Autosomal recessive (OMIM 222000)
Autosomal dominant (OMIM 125800)
Acquired
Autosomal recessive (OMIM 125700)
Autosomal dominant (OMIM 125700)
Wolfram syndrome (OMIM 222300/598500)
Premature infants
Radiant warmers
Phototherapy
Inadequate intake:
Ineffective breastfeeding
Child neglect or abuse
Adipsia (lack of thirst)
Diarrhea
Emesis/nasogastric suction
Osmotic cathartics (lactulose)
Cutaneous Losses
Burns
Excessive sweating
Renal Losses
The classic causes of hypernatremia from a water deficit are nephrogenic and
central diabetes insipidus (see Chapters 548 and 574 ). Hypernatremia
develops in diabetes insipidus only if the patient does not have access to water or
cannot drink adequately because of immaturity, neurologic impairment, emesis,
or anorexia. Infants are at high risk because of their inability to control their own
water intake. Central diabetes insipidus and the genetic forms of nephrogenic
diabetes insipidus typically cause massive urinary water losses and very dilute
urine. The water losses are less dramatic, and the urine often has the same
osmolality as plasma when nephrogenic diabetes insipidus is secondary to
intrinsic renal disease (obstructive uropathy, renal dysplasia, sickle cell disease).
The other causes of a water deficit are also secondary to an imbalance
between losses and intake. Newborns, especially if premature, have high
insensible water losses. Losses are further increased if the infant is placed under
a radiant warmer or with the use of phototherapy for hyperbilirubinemia. The
renal concentrating mechanisms are not optimal at birth, providing an additional
source of water loss. Ineffective breastfeeding, often in a primiparous mother,
can cause severe hypernatremic dehydration. Adipsia , the absence of thirst, is
usually secondary to damage to the hypothalamus, such as from trauma, tumor,
hydrocephalus, or histiocytosis. Primary adipsia is rare.
When hypernatremia occurs in conditions with deficits of sodium and water,
the water deficit exceeds the sodium deficit. This occurs only if the patient is
unable to ingest adequate water. Diarrhea results in depletion of both Na+ and
water. Because diarrhea is hypotonic—typical Na+ concentration of 35-65
mEq/L—water losses exceed Na+ losses, potentially leading to hypernatremia.
Most children with gastroenteritis do not have hypernatremia because they drink
through diarrhea, may need supplemental water and electrolytes (see Chapter 69
). Sodium intake is reduced if it contributed to the hypernatremia.
Hyponatremia
Hyponatremia, a very common electrolyte abnormality in hospitalized patients,
is a serum sodium level <135 mEq/L. Both total body sodium and TBW
determine the serum sodium concentration. Hyponatremia exists when the ratio
of water to Na+ is increased. This condition can occur with low, normal, or high
levels of body Na+ . Similarly, body water can be low, normal, or high.
Hyperlipidemia
Hyperproteinemia
HYPEROSMOLALITY
Hyperglycemia
Iatrogenic (mannitol, sucrose, glycine)
HYPOVOLEMIC HYPONATREMIA
EXTRARENAL LOSSES
RENAL LOSSES
EUVOLEMIC HYPONATREMIA
HYPERVOLEMIC HYPONATREMIA
Heart failure
Cirrhosis
Nephrotic syndrome
Acute, chronic kidney injury
Capillary leak caused by sepsis
Hypoalbuminemia caused by gastrointestinal disease (protein-losing
enteropathy)
* Most cases of proximal renal tubular acidosis are not caused by this primary
• Absence of:
Renal, adrenal, or thyroid insufficiency
Heart failure, nephrotic syndrome, or cirrhosis
Diuretic ingestion
Dehydration
• Urine osmolality >100 mOsm/kg (usually > plasma)
• Serum osmolality <280 mOsm/kg and serum sodium <135 mEq/L
• Urine sodium >30 mEq/L
• Reversal of “sodium wasting” and correction of hyponatremia with water
restriction
Hyperkalemia
Hyperkalemia—because of the potential for lethal arrhythmias—is one of the
most alarming electrolyte abnormalities.
Hemolysis
Tissue ischemia during blood drawing
Thrombocytosis
Leukocytosis
Familial pseudohyperkalemia (OMIM 609153/611184/612126)
Increased Intake
Intravenous or oral
Blood transfusions
Transcellular Shifts
Acidosis
Rhabdomyolysis
Tumor lysis syndrome
Tissue necrosis
Hemolysis/hematomas/gastrointestinal bleeding
Succinylcholine
Digitalis intoxication
Fluoride intoxication
β-Adrenergic blockers
Exercise
Hyperosmolality
Insulin deficiency
Malignant hyperthermia (OMIM 145600/601887)
Hyperkalemic periodic paralysis (OMIM 170500)
Decreased Excretion
Renal failure
Primary adrenal disease
Acquired Addison disease
21-Hydroxylase deficiency (OMIM 201910)
3β-Hydroxysteroid dehydrogenase deficiency (OMIM 201810)
Lipoid congenital adrenal hyperplasia (OMIM 201710)
Adrenal hypoplasia congenita (OMIM 300200)
Aldosterone synthase deficiency (OMIM 203400/610600)
Adrenoleukodystrophy (OMIM 300100)
Hyporeninemic hypoaldosteronism
Urinary tract obstruction
Sickle cell disease (OMIM 603903)
Kidney transplant
Lupus nephritis
Renal tubular disease
Pseudohypoaldosteronism type I (OMIM 264350/177735)
Pseudohypoaldosteronism type II (OMIM 145260)
Bartter syndrome, type 2 (OMIM 241200)
Urinary tract obstruction
Kidney transplant
Medications
Renin inhibitors
Angiotensin-converting enzyme inhibitors
Angiotensin II blockers
Potassium-sparing diuretics
Calcineurin inhibitors
Nonsteroidal antiinflammatory drugs
Trimethoprim
Heparin
Drospirenone (in some oral contraceptives)
Hypokalemia
Hypokalemia is common in children, with most cases related to gastroenteritis.
Alkalemia
Insulin
α-Adrenergic agonists
Drugs/toxins (theophylline, barium, toluene, cesium chloride,
hydroxychloroquine)
Hypokalemic periodic paralysis (OMIM 170400)
Thyrotoxic period paralysis
Refeeding syndrome
Decreased Intake
Anorexia nervosa
Extrarenal Losses
Diarrhea
Laxative abuse
Sweating
Sodium polystyrene sulfonate (Kayexalate) or clay ingestion
Renal Losses
With Metabolic Acidosis
* Most cases of proximal renal tubular acidosis are not caused by this primary
Because the intracellular [K+ ] is much higher than the plasma level, a
significant amount of K+ can move into cells without greatly changing the
intracellular [K+ ]. Alkalemia is one of the more common causes of a
transcellular shift. The effect is much greater with a metabolic alkalosis than
with a respiratory alkalosis. The impact of exogenous insulin on K+ movement
into the cells is substantial in patients with DKA. Endogenous insulin may be the
cause when a patient is given a bolus of glucose. Both endogenous (epinephrine
in stress) and exogenous (albuterol) β-adrenergic agonists stimulate cellular
uptake of K+ . Theophylline overdose, barium intoxication, administration of
cesium chloride (a homeopathic cancer remedy), and toluene intoxication from
paint or glue sniffing can cause a transcellular shift hypokalemia, often with
severe clinical manifestations. Children with hypokalemic periodic paralysis, a
rare autosomal dominant disorder, have acute cellular uptake of K+ (see Chapter
629 ). Thyrotoxic periodic paralysis, which is more common in Asians, is an
unusual initial manifestation of hyperthyroidism. Affected patients have
dramatic hypokalemia as a result of a transcellular shift of potassium.
Hypokalemia can occur during refeeding syndrome (see Chapters 58 and 364.8
).
Inadequate K+ intake occurs in anorexia nervosa ; accompanying bulimia and
laxative or diuretic abuse exacerbates the K+ deficiency. Sweat losses of K+ can
be significant during vigorous exercise in a hot climate. Associated volume
depletion and hyperaldosteronism increase renal losses of K+ (discussed later).
Diarrheal fluid has a high concentration of K+ , and hypokalemia as a result of
diarrhea is usually associated with metabolic acidosis resulting from stool losses
of bicarbonate. In contrast, normal acid-base balance or mild metabolic alkalosis
is seen with laxative abuse. Intake of SPS or ingestion of clay because of pica
increases stool losses of potassium.
Urinary potassium wasting may be accompanied by a metabolic acidosis
(proximal or distal RTA). In DKA, although it is often associated with normal
plasma [K+ ] from transcellular shifts, there is significant total body K+ depletion
from urinary losses because of the osmotic diuresis, and the K+ level may
decrease dramatically with insulin therapy (see Chapter 607 ). Both the polyuric
10 mg/dL × 0.25 = 2.5 mmol/L). Dividing the right-column unit by the
conversion factor converts to the units of the left-column unit.
Table 68.6
Conversion Factors for Calcium, Magnesium, and
Phosphorus
Magnesium Intake
Between 30% and 50% of dietary magnesium is absorbed. Good dietary sources
include green vegetables, cereals, nuts, meats, and hard water, although many
foods contain magnesium. Human milk contains approximately 35 mg/L of
magnesium; formula contains 40-70 mg/L. The small intestine is the major site
of magnesium absorption, but the regulation of magnesium absorption is poorly
understood. There is passive absorption, which permits high absorption in the
presence of excessive intake. It probably occurs by a paracellular mechanism.
Absorption is diminished in the presence of substances that complex with
magnesium (free fatty acids, fiber, phytate, phosphate, oxalate); increased
intestinal motility and calcium also decrease magnesium absorption. Vitamin D
and parathyroid hormone (PTH) may enhance absorption, although this effect is
limited. Intestinal absorption does increase when intake is decreased, possibly by
a saturable, active transport system. If there is no oral intake of magnesium,
obligatory secretory losses prevent the complete elimination of intestinal losses.
Magnesium Excretion
Renal excretion is the principal regulator of magnesium balance. There is no
defined hormonal regulatory system, although PTH may increase tubular
resorption. Approximately 15% of resorption occurs in the proximal tubule, and
70% in the thick ascending limb (TAL) of the loop of Henle. Proximal resorption
may be higher in neonates. High serum magnesium levels inhibit resorption in
the TAL, suggesting that active transport is involved. Approximately 5–10% of
filtered magnesium is resorbed in the distal tubule. Hypomagnesemia increases
absorption in the TAL and the distal tubule.
Hypomagnesemia
Hypomagnesemia is relatively common in hospitalized patients, although most
cases are asymptomatic. Detection requires a high index of suspicion because
magnesium is not measured in most basic metabolic panels.
Diarrhea
Nasogastric suction or emesis
Inflammatory bowel disease
Celiac disease
Cystic fibrosis
Intestinal lymphangiectasia
Small bowel resection or bypass
Pancreatitis
Protein-calorie malnutrition
Patiromer
Hypomagnesemia with secondary hypocalcemia (OMIM 602014)*
Renal Disorders
Medications
Amphotericin
Cisplatin
Cyclosporine, tacrolimus
Loop diuretics
Mannitol
Pentamidine
Proton pump inhibitors
Aminoglycosides
Thiazide diuretics
Epidermal growth factor receptor inhibitors (cetuximab)
Diabetes
Acute tubular necrosis (recovery phase)
Postobstructive nephropathy
Chronic kidney diseases
Interstitial nephritis
Glomerulonephritis
Post–renal transplantation
Hypercalcemia
Intravenous fluids
Primary aldosteronism
Genetic diseases
Gitelman syndrome (OMIM 263800)
Bartter syndrome (OMIM
241200/607364/602522/601678/300971/601198/613090)
Familial hypomagnesemia with hypercalciuria and
nephrocalcinosis (OMIM 248250)
Familial hypomagnesemia with hypercalciuria, nephrocalcinosis,
and severe ocular involvement (OMIM 248190)
Autosomal recessive renal magnesium wasting with
normocalciuria (OMIM 611718)
Renal cysts and diabetes syndrome (OMIM 137920)
Autosomal dominant hypomagnesemia (OMIM
160120/613882/154020)
EAST syndrome (OMIM 612780)
Autosomal dominant hypoparathyroidism (OMIM 146200)
Mitochondrial disorders (OMIM 500005)
Hypomagnesemia after transient neonatal hyperphenylalaninemia
Hypomagnesemia with impaired brain development
Hypomagnesemia with metabolic syndrome
Hyperuricemia, pulmonary hypertension, renal failure, alkalosis syndrome
(HUPRA)
HNF1B nephropathy
Miscellaneous Causes
Poor intake
Hungry bone syndrome
Insulin administration
Pancreatitis
Intrauterine growth restriction
Infants of diabetic mothers
Exchange transfusion
Hypophosphatemia
Because of the wide variation in normal plasma phosphorus levels, the definition
of hypophosphatemia is age dependent (see Table 68.8 ). The normal range
reported by a laboratory may be based on adult normal values and therefore may
be misleading in children. A serum phosphorus level of 3 mg/dL, a normal value
in an adult, indicates clinically significant hypophosphatemia in an infant.
The plasma phosphorus level does not always reflect the total body stores
because only 1% of phosphorus is extracellular. Thus, a child may have
significant phosphorus deficiency despite a normal plasma phosphorus
concentration when there is a shift of phosphorus from the ICS.
Decreased Intake
Nutritional
Premature infants
Low phosphorus formula
Antacids and other phosphate binders
Renal Losses
Hyperparathyroidism
Parathyroid hormone–related peptide
X-linked hypophosphatemic rickets (OMIM 307800)
Overproduction of fibroblast growth factor-23
Tumor-induced rickets
McCune-Albright syndrome (OMIM 174800)
Epidermal nevus syndrome
Neurofibromatosis
Autosomal dominant hypophosphatemic rickets (OMIM 193100)
Autosomal recessive hypophosphatemic rickets, types 1 and 2
(OMIM 241520/613312)
Fanconi syndrome
Dent disease (OMIM 300009/300555)
Hypophosphatemic rickets with hypercalciuria (OMIM 241530)
Volume expansion and intravenous fluids
Metabolic acidosis
Diuretics
Glycosuria
Glucocorticoids
Kidney transplantation
Multifactorial
Vitamin D deficiency
Vitamin D–dependent rickets type 1 (OMIM 264700)
Vitamin D–dependent rickets type 2 (OMIM 277440)
Alcoholism
Sepsis
Dialysis
Hyperphosphatemia
Etiology and Pathophysiology
Renal insufficiency is the most common cause of hyperphosphatemia, with the
severity proportional to the degree of kidney impairment (see Chapter 550 ).
This occurs because GI absorption of the large dietary intake of phosphorus is
unregulated, and the kidneys normally excrete this phosphorus. As renal function
deteriorates, increased excretion of phosphorus is able to compensate. When
kidney function is <30% of normal, hyperphosphatemia usually develops,
although this varies considerably depending on dietary intake. Many of the other
causes of hyperphosphatemia are more likely to develop in the setting of renal
insufficiency (Table 68.10 ).
Table 68.10
Causes of Hyperphosphatemia
Transcellular Shifts
Increased Intake
Decreased Excretion
Renal failure
Hypoparathyroidism or pseudohypoparathyroidism (OMIM
146200/603233/103580/241410/203330)
Acromegaly
Hyperthyroidism
Tumoral calcinosis with hyperphosphatemia (OMIM 211900)
Table 68.11
Appropriate Compensation During Simple Acid-Base
Disorders
Diagnosis
A systematic evaluation of an arterial blood gas (ABG) sample, combined with
the clinical history, can usually explain the patient's acid-base disturbance.
Assessment of an ABG sample requires knowledge of normal values (Table
68.12 ). In most cases, this is accomplished through a 3-step process (Fig. 68.8 ):
Table 68.12
A mixed acid-base disturbance is present if the respiratory compensation is
not appropriate. If the PCO 2 is greater than predicted, the patient has a concurrent
respiratory acidosis. A lower PCO 2 than predicted indicates a concurrent
respiratory alkalosis or, less frequently, an isolated respiratory alkalosis. Because
the appropriate respiratory compensation for a metabolic acidosis never
normalizes the patient's pH, the presence of a normal pH and a low [HCO3 − ]
occurs only if some degree of respiratory alkalosis is present. In this situation,
distinguishing an isolated chronic respiratory alkalosis from a mixed metabolic
acidosis and acute respiratory alkalosis may be possible only clinically. In
contrast, the combination of a low serum pH and a low [HCO3 − ] occurs only if a
metabolic acidosis is present.
Diarrhea
Renal tubular acidosis (RTA)
Distal (type I) RTA (OMIM 179800/602722/267300)*
Proximal (type II) RTA (OMIM 604278) †
Mixed (type III) RTA (OMIM 259730)
Hyperkalemic (type IV) RTA (OMIM
201910/264350/177735/145260) ‡
Urinary tract diversions
Posthypocapnia
Ammonium chloride intake
Tissue hypoxia
Shock
Hypoxemia
Severe anemia
Liver failure
Malignancy
Intestinal bacterial overgrowth
Inborn errors of metabolism
Medications
Nucleoside reverse transcriptase inhibitors
Metformin
Propofol
Linezolid
Ketoacidosis
Diabetic ketoacidosis
Starvation ketoacidosis
Alcoholic ketoacidosis
Kidney Failure
Poisoning
Ethylene glycol
Methanol
Salicylate
Toluene
Paraldehyde
* Along with these genetic disorders, distal RTA may be secondary to renal
disease or medications.
† Most cases of proximal RTA are not caused by this primary genetic disorder.
Gastric losses
Emesis
Nasogastric suction
Diuretics (loop or thiazide)
Chloride-losing diarrhea (OMIM 214700)
Low chloride formula
Cystic fibrosis (OMIM 219700)
Posthypercapnia
Encephalitis
Head trauma
Brain tumor
Central sleep apnea
Primary pulmonary hypoventilation (Ondine curse)
Stroke
Hypoxic brain damage
Obesity-hypoventilation (pickwickian) syndrome
Increased intracranial pressure
Medications
Narcotics
Barbiturates
Anesthesia
Benzodiazepines
Propofol
Alcohols
Diaphragmatic paralysis
Guillain-Barré syndrome
Poliomyelitis
Acute flaccid myelitis
Spinal muscular atrophies
Tick paralysis
Botulism
Myasthenia
Multiple sclerosis
Spinal cord injury
Medications
Vecuronium
Aminoglycosides
Organophosphates (pesticides)
Muscular dystrophy
Hypothyroidism
Malnutrition
Hypokalemia
Hypophosphatemia
Medications
Succinylcholine
Corticosteroids
Pulmonary Disease
Pneumonia
Pneumothorax
Asthma
Bronchiolitis
Pulmonary edema
Pulmonary hemorrhage
Acute respiratory distress syndrome
Neonatal respiratory distress syndrome
Cystic fibrosis
Bronchopulmonary dysplasia
Hypoplastic lungs
Meconium aspiration
Pulmonary thromboembolus
Interstitial fibrosis
Aspiration
Laryngospasm
Angioedema
Obstructive sleep apnea
Tonsillar hypertrophy
Vocal cord paralysis
Extrinsic tumor
Extrinsic or intrinsic hemangioma
Miscellaneous
Flail chest
Cardiac arrest
Kyphoscoliosis
Decreased diaphragmatic movement due to ascites or peritoneal dialysis
Pneumonia
Pulmonary edema
Cyanotic heart disease
Congestive heart failure
Asthma
Severe anemia
High altitude
Laryngospasm
Aspiration
Carbon monoxide poisoning
Pulmonary embolism
Interstitial lung disease
Hypotension
Pneumonia
Pulmonary edema
Asthma
Pulmonary embolism
Hemothorax
Pneumothorax
Respiratory distress syndrome (adult or infant)
Central Stimulation
Central nervous system disease
Subarachnoid hemorrhage
Encephalitis or meningitis
Trauma
Brain tumor
Stroke
Fever
Pain
Anxiety (panic attack)
Psychogenic hyperventilation or anxiety
Liver failure
Sepsis
Pregnancy
Mechanical ventilation
Hyperammonemia
Extracorporeal membrane oxygenation or hemodialysis
Medications
Salicylate intoxication
Theophylline
Progesterone
Exogenous catecholamines
Caffeine
Table 69.3
Hourly Maintenance Water Rate
Intravenous Solutions
The components of available solutions are shown in Table 69.4 . These solutions
are available with 5% dextrose (D5), 10% dextrose (D10), or without dextrose.
Except for Ringer lactate (lactated Ringer, LR), they are also available with
added potassium (10 or 20 mEq/L). A balanced IV fluid contains a base (lactate
or acetate), a more physiologic chloride concentration than NS, and additional
physiologic concentrations of electrolytes such as potassium, calcium, and
magnesium. Examples include LR and PlasmaLyte, and there is evidence
suggesting benefit versus NS in certain clinical situations. A hospital pharmacy
can also prepare custom-made solutions with different concentrations of sodium
or potassium. In addition, other electrolytes, such as calcium, magnesium,
phosphate, acetate, and bicarbonate, can be added to IV solutions. Custom-made
solutions take time to prepare and are much more expensive than commercial
solutions. The use of custom-made solutions is necessary only for patients who
have underlying disorders that cause significant electrolyte imbalances. The use
of commercial solutions saves time and expense.
Table 69.4
Composition of Intravenous Solutions*
Glucose
Maintenance fluids usually contain D5, which provides 17 calories/100 mL and
nearly 20% of the daily caloric needs. This level is enough to prevent ketone
production and helps minimize protein degradation, but the child will lose
weight on this regimen. The weight loss is the principal reason why a patient
needs to be started on TPN after a few days of maintenance fluids if enteral
feedings are still not possible. Maintenance fluids are also lacking in such crucial
nutrients as protein, fat, vitamins, and minerals.
magnitude of normal water losses. Table 69.5 lists the 3 sources of normal water
loss.
Table 69.5
Sources of Water Loss
• Urine: 60%
• Insensible losses: ≈35% (skin and lungs)
• Stool: 5%
Urine is the most important contributor to normal water loss. Insensible losses
represent approximately one third of total maintenance water (40% in infants;
25% in adolescents and adults). Insensible losses are composed of evaporative
losses from the skin and lungs that cannot be quantitated. The evaporative losses
from the skin do not include sweat, which would be considered an additional
(sensible) source of water loss. Stool normally represents a minor source of
water loss.
Maintenance water and electrolyte needs may be increased or decreased,
depending on the clinical situation. This may be obvious, as in the infant with
profuse diarrhea, or subtle, as in the patient who has decreased insensible losses
while receiving mechanical ventilation. It is helpful to consider the sources of
normal water and electrolyte losses and to determine whether any of these
sources is being modified in a specific patient. It is then necessary to adjust
maintenance water and electrolyte calculations.
Table 69.6 lists a variety of clinical situations that modify normal water and
electrolyte losses. The skin can be a source of very significant water loss,
particularly in neonates, especially premature infants, who are under radiant
warmers or are receiving phototherapy. Very-low-birthweight infants can have
insensible losses of 100-200 mL/kg/24 hr. Burns can result in massive losses of
water and electrolytes, and there are specific guidelines for fluid management in
children with burns (see Chapter 92 ). Sweat losses of water and electrolytes,
especially in a warm climate, can also be significant. Children with cystic
fibrosis and some children with pseudohypoaldosteronism have increased
sodium losses from the skin.
Table 69.6
Adjustments in Maintenance Water
Fever increases evaporative losses from the skin. These losses are somewhat
predictable, leading to a 10–15% increase in maintenance water needs for each
1°C (1.8°F) increase in temperature above 38°C (100.4°F). These guidelines are
for a patient with a persistent fever; a 1 hr fever spike does not cause an
appreciable increase in water needs.
Tachypnea or a tracheostomy increases evaporative losses from the lungs. A
humidified ventilator causes a decrease in insensible losses from the lungs and
can even lead to water absorption via the lungs; a ventilated patient has a
decrease in maintenance water requirements. It may be difficult to quantify the
changes that take place in the individual patient in these situations.
Replacement Fluids
The gastrointestinal (GI) tract is potentially a source of considerable water loss.
GI water losses are accompanied by electrolytes and thus may cause
disturbances in intravascular volume and electrolyte concentrations. GI losses
are often associated with loss of potassium, leading to hypokalemia. Because of
the high bicarbonate concentration in stool, children with diarrhea usually have a
metabolic acidosis , which may be accentuated if volume depletion causes
hypoperfusion and a concurrent lactic acidosis. Emesis or losses from an NG
tube can cause a metabolic alkalosis (see Chapter 68 ).
In the absence of vomiting, diarrhea, or NG drainage, GI losses of water and
electrolytes are usually quite small. All GI losses are considered excessive, and
the increase in the water requirement is equal to the volume of fluid losses.
Because GI water and electrolyte losses can be precisely measured, an
appropriate replacement solution can be used.
It is impossible to predict the losses for the next 24 hr; it is better to replace
excessive GI losses as they occur. The child should receive an appropriate
maintenance fluid that does not consider the GI losses. The losses should then be
replaced after they occur, with use of a solution with a similar electrolyte
concentration as the GI fluid. The losses are usually replaced every 1-6 hr,
depending on the rate of loss, with very rapid losses being replaced more
frequently.
Diarrhea is a common cause of fluid loss in children and can result in
dehydration and electrolyte disorders. In the unusual patient with significant
diarrhea and a limited ability to take oral fluid, it is important to have a plan for
replacing excessive stool losses. The volume of stool should be measured, and
an equal volume of replacement solution should be given. Data are available on
the average electrolyte composition of diarrhea in children (Table 69.4 ). With
this information an appropriate replacement solution can be designed. The
solution shown in Table 69.7 replaces stool losses of Na+ , K+ , Cl− , and
bicarbonate. Each 1 mL of stool should be replaced by 1 mL of this solution. The
average electrolyte composition of diarrhea is just an average, and there may be
considerable variation. It is therefore advisable to consider measuring the
electrolyte composition of a patient's diarrhea if the amount is especially
excessive or if the patient's serum electrolyte levels are problematic.
Table 69.7
Replacement Fluid for Diarrhea
Average Composition of Diarrhea
Sodium: 55 mEq/L
Potassium: 25 mEq/L
Bicarbonate: 15 mEq/L
Sodium: 60 mEq/L
Potassium: 10 mEq/L
Chloride: 90 mEq/L
Polyuria
Bibliography
Foster BA, Tom D, Hill V. Hypotonic versus isotonic fluids in
hospitalized children: a systematic review and meta-analysis.
J Pediatr . 2014;165:163–169 [e2].
CHAPTER 70
Deficit Therapy
Larry A. Greenbaum
Clinical Manifestations
The 1st step in caring for the child with dehydration is to assess the degree of
dehydration (Table 70.1 ), which dictates both the urgency of the situation and
the volume of fluid needed for rehydration. The infant with mild dehydration (3–
5% of body weight dehydrated) has few clinical signs or symptoms. The infant
may be thirsty; the alert parent may notice a decline in urine output. The history
is most helpful. The infant with moderate dehydration has clear physical signs
and symptoms. Intravascular space depletion is evident from an increased heart
rate and reduced urine output. This patient needs fairly prompt intervention. The
infant with severe dehydration is gravely ill. The decrease in blood pressure
indicates that vital organs may be receiving inadequate perfusion. Immediate and
aggressive intervention is necessary. If possible, the child with severe
dehydration should initially receive intravenous (IV) therapy. For older children
and adults, mild, moderate, or severe dehydration represents a lower percentage
of body weight lost. This difference occurs because water accounts for a higher
percentage of body weight in infants (see Chapter 68 ).
Table 70.1
Clinical Evaluation of Dehydration
Mild dehydration (<5% in an infant; <3% in an older child or adult):
Normal or increased pulse; decreased urine output; thirsty; normal
physical findings
Moderate dehydration (5–10% in an infant; 3–6% in an older child or
adult): Tachycardia; little or no urine output; irritable/lethargic; sunken
eyes and fontanel; decreased tears; dry mucous membranes; mild delay in
elasticity (skin turgor); delayed capillary refill (>1.5 sec); cool and pale
Severe dehydration (>10% in an infant; >6% in an older child or
adult): Peripheral pulses either rapid and weak or absent; decreased
blood pressure; no urine output; very sunken eyes and fontanel; no tears;
parched mucous membranes; delayed elasticity (poor skin turgor); very
delayed capillary refill (>3 sec); cold and mottled; limp, depressed
consciousness
Vital signs:
Pulse
Blood pressure
Intake and output:
Fluid balance
Urine output
Physical examination:
Weight
Clinical signs of depletion or overload
Electrolytes
The patient's intake and output are critically important in the dehydrated child.
The child who, after 8 hr of therapy, has more output than input because of
continuing diarrhea needs to be started on a replacement solution. See the
guidelines in Chapter 69 for selecting an appropriate replacement solution. Urine
output is useful for evaluating the success of therapy. Good urine output
indicates that rehydration has been successful.
often brought for medical attention with more profound dehydration.
Children with hypernatremic dehydration are often lethargic, and they may be
irritable when touched. Hypernatremia may cause fever, hypertonicity, and
hyperreflexia. More severe neurologic symptoms may develop if cerebral
bleeding or thrombosis occurs.
Overly rapid treatment of hypernatremic dehydration may cause significant
morbidity and mortality. Idiogenic osmoles are generated within the brain
during the development of hypernatremia; they increase the osmolality within
the cells of the brain, providing protection against brain cell shrinkage caused by
movement of water out of the cells and into the hypertonic ECF. Idiogenic
osmoles dissipate slowly during the correction of hypernatremia. With overly
rapid lowering of the extracellular osmolality during the correction of
hypernatremia, an osmotic gradient may be created that causes water movement
from the ECS into the cells of the brain, producing cerebral edema. Symptoms of
the resultant cerebral edema can range from seizures to brain herniation and
death.
To minimize the risk of cerebral edema during the correction of
hypernatremic dehydration, the serum sodium concentration should not decrease
by >10 mEq/L every 24 hr. The deficits in severe hypernatremic dehydration
may need to be corrected over 2-4 days (Table 70.4 ).
Table 70.4
Treatment of Hypernatremic Dehydration
Restore intravascular volume:
Normal saline: 20 mL/kg over 20 min (repeat until intravascular
volume restored)
Determine time for correction on basis of initial sodium
concentration:
• [Na] 145-157 mEq/L: 24 hr
• [Na] 158-170 mEq/L: 48 hr
• [Na] 171-183 mEq/L: 72 hr
• [Na] 184-196 mEq/L: 84 hr
Administer fluid at constant rate over time for correction:
Typical fluid: 5% dextrose + half-normal saline (with 20 mEq/L
KCl unless contraindicated)
Typical rate: 1.25-1.5 times maintenance
Follow serum sodium concentration
Adjust fluid on basis of clinical status and serum sodium concentration:
Signs of volume depletion: administer normal saline (20 mL/kg)
Sodium decreases too rapidly; either:
• Increase sodium concentration of IV fluid
• Decrease rate of IV fluid
Sodium decreases too slowly; either:
• Decrease sodium concentration of IV fluid
• Increase rate of IV fluid
Replace ongoing losses as they occur
Table 72.1
Examples of Effects of Gene Polymorphisms on Drug
Response
Table 72.2
Some Important Relationships Between Drugs and
Cytochrome P450 (CYP) Enzymes* and P-Glycoprotein
Transporter
Ciprofloxacin (Cipro
)
CYP2C9 Diclofenac (Voltaren † ), ibuprofen (Motrin † ), Fluconazole Rifampin (Rifadin †
piroxicam (Feldene † ), Losartan (Cozaar ), irbesartan (Diflucan ) )
(Avapro ), celecoxib (Celebrex ), tolbutamide (Orinase † Fluvastatin (Lescol )
Amiodarone
), warfarin (Coumadin † ), phenytoin (Dilantin )
(Cordarone )
Zafirlukast
(Accolate )
CYP2C19 Omeprazole, lansoprazole (Prevacid ), pantoprazole Cimetidine Rifampin
(Protonix ), (S)-mephenytoin, (S) -citalopram (Lexapro Fluvoxamine
); nelfinavir (Viracept ), diazepam (Valium † ),
voriconazole (Vfend )
CYP2D6 CNS-active agents: Atomoxetine (Strattera ), Fluoxetine
amitriptyline (Elavil † ), desipramine (Norpramin † ), (Prozac † )
imipramine (Tofranil † ), paroxetine (Paxil ), haloperidol Paroxetine
(Haldol ), risperidone (Risperdal ), thioridazine
† (Paxil )
(Mellaril † )
Antiarrhythmic agents: Mexiletine (Mexitil ), Amiodarone
propafenone (Rythmol ) (Cordarone † )
Quinidine
(Quinidex † )
β-Blockers: Propranolol (
Inderal † ), metoprolol Terbinafine
(Lopressor † ), timolol (Blocadren † )
Narcotics: Codeine, dextromethorphan, hydrocodone
(Vicodin † )
Others: Tamoxifen (Nolvadex ) Cimetidine
Ritonavir
CYP3A4 Calcium channel blockers: Diltiazem (Cardizem † ), Amiodarone Barbiturates
felodipine (Plendil ), nimodipine (Nimotop ), nifedipine Carbamazepine
(Adalat † ), nisoldipine (Sular ), nitrendipine, verapamil (Tegretol † )
(Calan † ) Phenytoin
(Dilantin † )
Immunosuppressive agents: Cyclosporine A Efavirenz (Sustiva )
(Sandimmune , Neoral † ), tacrolimus (Prograf )
Corticosteroids: Budesonide (Pulmicort ), cortisol, Fluconazole Nevirapine
17β-estradiol, progesterone, testosterone Ketoconazole (Viramune )
(Nizoral † )
Itraconazole
(Sporanox )
Macrolide antibiotics: Clarithromycin (Biaxin ), Clarithromycin
erythromycin (Erythrocin † ), troleandomycin (TAO ) Erythromycin
Troleandomycin
Anticancer agents: Cyclophosphamide (Cytoxan † ), Imatinib Rifampin
gefitinib (Iressa ), ifosfamide (Ifex ), tamoxifen, Ritonavir ‡
vincristine (Oncovin † ), vinblastine (Velban † ),
Benzodiazepines: Alprazolam (Xanax † ), midazolam St. John's wort
(Versed † ), triazolam (Halcion † )
Opioids: Alfentanil (Alfenta † ), fentanyl (Sublimaze † ),
sufentanil (Sufenta † )
HMG-CoA reductase inhibitors: Lovastatin (Mevacor
) † , simvastatin (Zocor ), atorvastatin (Lipitor )
HIV protease inhibitors: Indinavir (Crixivan ), Ritonavir ‡
nelfinavir, ritonavir (Norvir ), saquinavir (Invirase, Indinavir
Fortovase ), amprenavir (Agenerase )
Others: Quinidine (Quinidex † ), sildenafil (Viagra ), Grapefruit juice
eletriptan (Relpax ), ziprasidone (Geodon ) Nefazodone
(Serzone )
P- Aldosterone, amprenavir, atorvastatin, cyclosporine, Amiodarone Amprenavir
glycoprotein dexamethasone (Decadron † ), digoxin (Lanoxin † ), Carvedilol (Coreg ) Clotrimazole
diltiazem, domperidone (Motilium ), doxorubicin (Mycelex † )
(Adriamycin † ), erythromycin, etoposide (VePesid ), Clarithromycin Phenothiazine
fexofenadine (Allegra ), hydrocortisone, indinavir, Cyclosporine Rifampin
ivermectin (Stromectol ), lovastatin, loperamide Erythromycin Ritonavir ‡
(Imodium † ), nelfinavir, ondansetron (Zofran ), Itraconazole St. John's wort
paclitaxel (Taxol ), quinidine, saquinavir, simvastatin, Ketoconazole
verapamil, vinblastine, vincristine Quinidine
Ritonavir ‡
Tamoxifen
Verapamil
* www.drug-interactions.com .
Table 72.3
Internet Resources for Pharmacogenetics and
Pharmacogenomics*
INTRODUCTION TO PHARMACOGENOMICS
http://www.pharmgkb.org/
http://www.mayoclinic.org/healthy-lifestyle/consumer-health/in-depth/personalized-medicine/art-20044300
PHARMACOGENETICS: ALLELIC VARIANTS OF DRUG-METABOLIZING ENZYMES
CYP2C9 http://www.cypalleles.ki.se/cyp2c9.htm
CYP2C19 http://www.cypalleles.ki.se/cyp2c19.htm
CYP2D6 http://www.cypalleles.ki.se/cyp2d6.htm
CYP3A4 http://www.cypalleles.ki.se/cyp3a4.htm
CYP3A5 http://www.cypalleles.ki.se/cyp3a5.htm
UGTs https://www.pharmacogenomics.pha.ulaval.ca/ugt-alleles-nomenclature/
NAT1 and NAT2 http://nat.mbg.duth.gr/
PHARMACOGENETICS: SUBSTRATES OF DRUG-METABOLIZING ENZYMES
http://medicine.iupui.edu/clinpharm/ddis/clinical-table
http://www.mayomedicallaboratories.com/it-mmfiles/Pharmacogenomic_Associations_Tables.pdf
PHARMACOGENETICS-BASED DOSING GUIDELINES
Dosing guidelines incorporating pharmacogenetic data developed by the Clinical Pharmacogenetics
Implementation Consortium are available on the CPIC web page https://cpicpgx.org/ , which is mirrored at
PharmGKB: https://www.pharmgkb.org/page/cpic ,
or through the National Guidelines Clearinghouse website, a publically accessible resource for evidence-based
clinical guidelines sponsored by the Agency for Healthcare Research and Quality (AHRQ), U.S. Department of
Health Services, at https://www.guideline.gov/search?q=CPIC .
CYP2D6, CYP2C19, and antidepressants:
https://cpicpgx.org/guidelines/guideline-for-tricyclic-antidepressants-and-cyp2d6-and-cyp2c19/
CYP2D6 and codeine:
https://cpicpgx.org/guidelines/guideline-for-codeine-and-cyp2d6/
CYP2D6, CYP2C19, and SSRIs:
https://cpicpgx.org/guidelines/guideline-for-selective-serotonin-reuptake-inhibitors-and-cyp2d6-and-cyp2c19/
CYP3A5 and tacrolimus:
https://cpicpgx.org/guidelines/guideline-for-tacrolimus-and-cyp3a5/
HLA-B and abacavir and allopurinol:
https://cpicpgx.org/guidelines/guideline-for-abacavir-and-hla-b/
https://cpicpgx.org/guidelines/guideline-for-allopurinol-and-hla-b/
https://cpicpgx.org/guidelines/guideline-for-carbamazepine-and-hla-b/
SLCO1B1 and simvastatin:
https://cpicpgx.org/guidelines/guideline-for-simvastatin-and-slco1b1/
TPMT and thiopurines:
https://cpicpgx.org/guidelines/guideline-for-thiopurines-and-tpmt/
*
All sites were accessible on July 14, 2017.
The consequences of allelic variation in the UGT2B family are less certain.
The predominant routes of morphine elimination include biotransformation to
the pharmacologically active 6-glucuronide (M6G) and the inactive 3-
glucuronide (M3G). M6G formation is almost exclusively catalyzed by
UGT2B7, whereas several UGTs in the UGT1A subfamily and UGT2B7, both
contribute to M3G formation. Increased M6G:morphine ratios have been
reported in individuals homozygous for the SNPs constituting the UGT2B7*2
allele. Although individuals genotyped as UGT2B7*2/*2 may produce higher-
than-anticipated concentrations of pharmacologically active morphine and its
metabolites, prospective studies addressing phenotype-genotype correlations and
the consequences of morphine analgesia have had conflicting results.
Thiopurine S -Methyltransferase
Thiopurine S -methyltransferase (TPMT ) is a cytosolic enzyme that catalyses
the S -methylation of aromatic and heterocyclic sulfur-containing compounds,
Drug Absorption
Drug absorption mainly occurs through passive diffusion, but active transport or
facilitated diffusion may also be necessary for drug entry into cells. Several
physiologic factors affect this process, one or more of which may be altered in
certain disease states (e.g., inflammatory bowel disease, diarrhea), and thus
produce changes in drug bioavailability. The rate and extent of absorption can be
significantly affected by a child's normal growth and development.
Peroral Absorption
The most important factors that influence drug absorption from the
gastrointestinal (GI) tract are related to the physiology of the stomach, intestine,
and biliary tract (Fig. 73.3C and Table 73.1 ). The rate and extent of peroral
absorption of drugs depend primarily on the pH-dependent passive diffusion and
motility of the stomach and intestinal tract, because both these factors will
influence transit time of the drug. Gastric pH changes significantly throughout
development, with the highest (alkaline) values occurring during the neonatal
period. In the fully mature neonate the gastric pH ranges from 6-8 at birth and
drops to 2-3 within a few hours of birth. However, after the 1st 24 hr of life, the
gastric pH increases because of the immaturity of the parietal cells. As the
parietal cells mature, the gastric acid secretory capacity increases (pH decreases)
over the 1st few months of life, reaching adult levels by age 3-7 yr. As a result,
the peroral bioavailability of acid-labile drugs (e.g., penicillin, ampicillin) is
increased. In contrast, the absorption of weak organic acids (e.g., phenobarbital,
phenytoin) is relatively decreased, a condition that may necessitate
administration of larger doses in very young patients to achieve therapeutic
plasma levels.
Table 73.1
Developmental Alterations in Intestinal Drug Absorption
Table 73.2
Influence of Ontogeny on Drug Absorption
PHYSIOLOGIC ALTERATION NEONATES INFANTS CHILDREN
Oral absorption Erratic Increased Near adult
Intramuscular absorption Variable Increased Near adult
Percutaneous absorption Increased Increased Near adult
Rectal absorption Very efficient Efficient Near adult
Direction of alteration given relative to expected normal adult pattern.
Data from Morselli PL: Development of physiological variables important for drug kinetics. In
Morselli PL, Pippenger CE, Penry JK, editors: Antiepileptic drug therapy in pediatrics, New York,
1983, Raven Press.
Drug Distribution
Drug distribution is influenced by a variety of drug-specific physiochemical
factors, including the role of drug transporters, blood-tissue protein binding,
blood-tissue pH, and perfusion. However, age-related changes in drug
distribution are primarily related to developmental changes in body composition
and the quantity of plasma proteins capable of drug binding. Age-dependent
changes in the relative sizes of body water —total body water (TBW) and
extracellular water (ECW)—and fat compartments may alter the apparent
volume of distribution (VD) for a given drug. The absolute amounts and
distribution of body water and fat depend on a child's age and nutritional status.
Also, certain disease states (e.g., ascites, dehydration, burn injuries, skin
Table 73.3
Factors Influencing Drug Binding in Pediatric Patients
Table 73.4
Impact of Development on Drug Metabolism
Table 73.5
Impact of Development on Renal Drug Elimination
where glomerular filtration rate (GFR), active tubular secretion (ATS), and
active tubular reabsorption (ATR) of drugs can contribute to overall clearance.
As for hepatic drug metabolism, only free (unbound) drug and metabolite can be
filtered by a normal glomerulus and secreted or reabsorbed by a renal tubular
transport protein.
Renal clearance is limited in the newborn because of anatomic and functional
immaturity of the nephron unit. In both the term and the preterm neonate, GFR
ATS of β-lactam antibiotics by probenecid). Also, drug-drug interactions may
occur at the level of the receptor (through competitive antagonism); many of
which are intentional and produce therapeutic benefit in pediatric patients (e.g.,
antihistamine reversal of histamine effects, naloxone reversal of opiate adverse
effects).
Table 73.6
Mechanism of Drug Interactions 1
EXAMPLE DRUG
RESULT
COMBINATION
PHARMACODYNAMIC
Additive Use of multiple drugs with similar adverse effect profiles can lead to
additive effects:
Fentanyl + midazolam Increased sedation
Class 1A antiarrhythmic 2 Increased QT prolongation
+ erythromycin 3 Increased potential for nephrotoxicity
Vancomycin + an
aminoglycoside 4
Synergy Penicillin + an Improved bactericidal efficacy against some gram-positive
aminoglycoside 4 organisms; penicillin inhibits bacterial cell wall synthesis, which for
some gram-positive organisms can improve the intracellular
penetration of the aminoglycoside
Antagonism Opioid + naloxone Competitive receptor antagonism; decreased efficacy of the opioid,
reversal of sedation, respiratory depression, and hypotension
Donepezil + an Oppositional effects; acetylcholinesterase inhibitors such as
anticholinergic donepezil increase acetylcholine concentrations by slowing the
degradation of acetylcholine, and anticholinergic drugs antagonize
the effect of acetylcholine
PHARMACOKINETIC
Absorption Inhibition of P-gp 5 :
Amiodarone + digoxin Increased digoxin concentration; gut P-gp is an efflux transporter
that takes drugs from cell cytoplasm and transports them back into
the intestinal lumen for excretion, limiting bioavailability
Complex formation:
Oral quinolone and Decreased antibiotic concentrations due to binding in the gut
tetracycline antibiotics +
divalent/trivalent cations (eg,
Ca2+ , Mg2+ , Fe3+ , Al3+ )
Distribution Ceftriaxone + endogenous Displacement of bilirubin from albumin binding site, increased risk
bilirubin of kernicterus in neonates
Metabolism Induction of CYP isozymes 5 ,
6 :
3
Woosley RL, Romero KA: QT drugs list. Available at: www.crediblemeds.org
4 Gentamicin, tobramycin, amikacin, streptomycin, neomycin
5
Inhibitors and inducers of CYP enzymes and P-glycoprotein. Med Lett Drugs Ther 2017;
September 18 (epub). Available at: www.medicalletter.org/downloads/CYP_PGP_Tables.pdf
6 Cytochrome P450 (CYP) isozymes that can affect drug metabolism include CYP1A2, 2C8, 2C9,
2C19, 2D6, and 3A4.
7 Some protease inhibitors metabolized by CYP3A4 include atazanavir, darunavir, fosamprenavir,
indinavir, lopinavir/ritonavir, nelfinavir, and saquinavir.
8
Itraconazole, ketoconazole, posaconazole, and voriconazole are strong inhibitors of CYP3A4.
Fluconazole is a moderate CYP3A4 inhibitor.
This table is not an all-inclusive list of drug interactions. The prescriber is encouraged to assess
the possibility of drug interactions when prescribing medications. This table does not address the
chemical compatibility of drugs (eg, IV-line compatibility).
CYP = cytochrome P-450; P-gp = P-glycoprotein.
Modified from Rizack M, Hillman C: The Medical Letter Handbook of Adverse Drug Interactions.
New Rochelle, NY, The Medical Letter, 1989. IBM Micromedex DRUGDEX, Copyright IBM
Corporation 2018; Med Lett Drugs Ther 2018;60:e160.
Table 74.3
Specific Pediatric Diseases and Their Anesthetic
Implications
DISEASE IMPLICATIONS
RESPIRATORY SYSTEM
Asthma Intraoperative bronchospasm that may be life threatening
Pneumothorax or atelectasis
Optimal preoperative medical management is essential.
Difficult airway Special equipment and personnel may be required.
Should be anticipated with dysmorphic features or storage diseases
Patients with trisomy 21 may require atlantooccipital joint evaluation.
Increased risk with acute airway obstruction, epiglottitis, laryngotracheobronchitis, or airway
foreign body
Bronchopulmonary Barotrauma with positive pressure ventilation
dysplasia Oxygen toxicity, pneumothorax a risk
Cystic fibrosis Airway reactivity, bronchorrhea, increased intraoperative pulmonary shunt and hypoxia
Risk of pneumothorax, pulmonary hemorrhage
Atelectasis, risk of prolonged postoperative ventilation
Patient should be assessed for cor pulmonale.
Sleep apnea Pulmonary hypertension and cor pulmonale must be excluded.
Careful postoperative observation for obstruction required
CARDIAC
Bacterial endocarditis prophylaxis as indicated
Use of air filters; careful purging of air from the intravenous equipment
Physician must understand the effects of various anesthetics on the hemodynamics of specific
lesions.
Possible need for preoperative evaluation of myocardial function and pulmonary vascular
resistance
Provide information about pacemaker function and ventricular device function.
HEMATOLOGIC
Sickle cell disease Possible need for simple or exchange transfusion based on preoperative hemoglobin
concentration and percentage of hemoglobin S
Avoid hypoxemia, hypothermia, dehydration, and hyperviscosity states.
Oncology Pulmonary evaluation of patients who have received bleomycin, bis -chloroethyl-nitrosourea,
chloroethyl-cyclohexyl-nitrosourea, methotrexate, or radiation to the chest
Avoidance of high oxygen concentration
Cardiac evaluation of patients who have received anthracyclines; risk of severe myocardial
depression with volatile agents
Potential for coagulopathy
RHEUMATOLOGIC
Limited mobility of the temporomandibular joint, cervical spine, arytenoid cartilages
Careful preoperative evaluation required
Possible difficult airway
GASTROINTESTINAL
Esophageal, gastric Potential for reflux and aspiration
Liver Altered metabolism of many anesthetic drugs
Potential for coagulopathy and uncontrollable intraoperative bleeding
RENAL
Altered electrolyte and acid-base status
Altered clearance of many anesthetic drugs
Need for preoperative dialysis in selected cases
Succinylcholine to be used with extreme caution and only when the serum potassium level
has recently been shown to be normal
NEUROLOGIC
Seizure disorder Avoidance of anesthetics that may lower the seizure threshold
Optimal control ascertained preoperatively
Preoperative serum anticonvulsant measurements
Increased Avoidance of agents that increase cerebral blood flow
intracranial Maintain cerebral perfusion pressure.
pressure
Neuromuscular Avoidance of depolarizing relaxants; at risk for hyperkalemia
disease Patient may be at risk for malignant hyperthermia; avoid volatile anesthetics in myopathies.
Developmental Patient may be uncooperative during induction and emergence.
delay
Psychiatric Monoamine oxidase inhibitor (or cocaine) may interact with meperidine, resulting in
hyperthermia and seizures.
Selective serotonin reuptake inhibitors may induce or inhibit various hepatic enzymes that
may alter anesthetic drug clearance.
Illicit drugs may have adverse effects on cardiorespiratory homeostasis and may potentiate
the action of anesthetics.
ENDOCRINE
Diabetes Greatest risk is unrecognized intraoperative hypoglycemia; intraoperative blood glucose level
monitoring needed especially when insulin is administered.
SKIN
Burns Difficult airway
Fluid shifts
Bleeding
Risk of rhabdomyolysis and hyperkalemia from succinylcholine following burns for many
months
IMMUNOLOGIC
Retroviral drugs may inhibit benzodiazepine clearance.
Immunodeficiency requires careful infection control practices.
Cytomegalovirus-negative blood products, irradiation, or leukofiltration may be required.
METABOLIC
Careful assessment of glucose homeostasis in infants
Preanesthetic Evaluation
All children presenting for surgery should undergo a preanesthetic history and
multiorgan system assessment with assignment of American Society of
Anesthesiologists Physical Status (ASA-PS) (Table 74.4 ). Children of ASA-
PS I-II generally require a brief history, notation of medical allergies, and
physical examination focusing on the neurologic and cardiorespiratory systems,
with no additional testing. Patients with complex medical history of ASA-PS
≥III require a more comprehensive preanesthetic assessment often with ancillary
preoperative testing. Children should be screened for anesthetic risks, including
drug allergies, previous reactions to anesthetics, and family history of problems
with anesthesia (e.g., sudden perioperative death, hyperthermia after surgery),
which may indicate risk of malignant hyperthermia.
Table 74.4
American Society of Anesthesiology Physical
Status Classification
Respiratory System
Recent respiratory tract infections should be noted. Clear rhinorrhea without
fever is not associated with increased anesthetic risk. Respiratory illnesses
associated with fever, mucopurulent nasal discharge, productive cough, or lower
respiratory symptoms (wheezing, rales) are associated with increased airway
reactivity and anesthetic complications for up to 6 wk thereafter. There may also
be increased risk of perioperative laryngospasm and bronchospasm, reduced
mucociliary clearance, atelectasis, and hypoxemia. It is recommended that
elective procedures requiring general anesthesia be postponed 4-6 wk in this
setting.
Children with reactive airway disease require a thorough preanesthetic
assessment. Acute, potentially fatal bronchospasm can occur during induction of
anesthesia and endotracheal intubation for routine, minor surgery in children
with asthma. Children at increased risk for anesthetic complications have
experienced asthma exacerbations requiring (1) hospital admission within the
previous year; (2) emergency department (ED) care within the last 6 mo; (3)
previous intensive care unit (ICU) admission; or (4) previous parenteral systemic
corticosteroids. Ideally, children should be free of wheezing for least several
days before surgery, even if this necessitates increased controller medication
administration (β-adrenergic agonist and corticosteroids). Active wheezing is an
indication for delaying elective surgery. Chronic respiratory conditions such as
bronchopulmonary dysplasia and cystic fibrosis are also associated with
significant intraoperative risks. Every effort should be made to ensure that
children with such disorders achieve optimal respiratory status before surgery.
Airway Evaluation
Induction of general anesthesia is associated with reduced spontaneous
ventilation and airway reflexes. Prediction of difficult bag-mask ventilation
and/or intubation before anesthesia is critical. Congenital anomalies associated
with airway compromise include micrognathia, macroglossia, and thoracic
anomalies (Table 74.5 ). Conditions that impair mouth opening (e.g.,
temporomandibular joint disease) should also be noted. A history of wheezing or
stridor may indicate postoperative airway complications and difficult
intraoperative airway management. It is also essential to ask about a history of
sleep-disordered breathing using the STBUR (snoring, trouble breathing, un-
refreshed) index, which may be predictive of perioperative respiratory
complications.
Table 74.5
Common Difficult Airway Syndromes
Achondroplasia
Airway tumors, hemangiomas
Apert syndrome
Beckwith-Wiedemann syndrome
Choanal atresia
Cornelia de Lange syndrome
Cystic hygroma/teratoma
DiGeorge syndrome
Fractured mandible
Goldenhar syndrome
Juvenile rheumatoid arthritis
Mucopolysaccharidosis
Pierre Robin syndrome
Smith-Lemli-Opitz syndrome
Treacher-Collins syndrome
Trisomy 21
Turner syndrome
Cardiovascular System
Most anesthetic agents possess myocardial depressant properties. All patients
should be screened for the presence of heart disease. Important cardiovascular
considerations include history of congenital heart disease (CHD), cyanosis,
arrhythmias, or cardiomyopathy. Room-air pulse oximetry should be performed
as part of the preanesthetic evaluation. Accurate diagnosis of cardiac murmurs in
neonates is essential. A history of cardiac dysrhythmias should be investigated
because inhalational anesthetics may be arrhythmogenic. A pediatric cardiologist
should evaluate children with known CHD undergoing surgery. Preoperative
ancillary studies may include electrocardiogram (ECG), echocardiogram, or
cardiac catheterization. Lesions associated with increased anesthetic risk include
single-ventricle heart disease, fixed obstructive outflow tract lesions (aortic
valve and pulmonary valve stenosis), and cardiomyopathy. Children with these
conditions should be cared for by a cardiac anesthesia service . Antibiotic
prophylaxis for the prevention of bacterial endocarditis may also be indicated,
and the American Heart Association (AHA) guidelines should be followed.
Hematologic System
Evidence of coagulopathy should be sought. Easy bruising, familial bleeding
disorders, and anticoagulant (e.g., aspirin, heparin, warfarin) use should be
discussed. Preoperative adequacy of hemostatic function (e.g., platelet count,
fibrinogen, prothrombin time, partial thromboplastin time) and correction of
coagulopathic disorders may be indicated for complex procedures associated
with significant risk of perioperative hemorrhage. In neonates, assurance of
vitamin K prophylaxis and adequate coagulation status is critical before any
major surgery. Although anemia may be well tolerated in healthy children,
providing rapid-onset anxiolysis and amnesia.
Genetic Evaluation
Children with genetic conditions may have syndrome-specific anesthetic
considerations. For example, children with trisomy 21 may have cardiac
anomalies, macroglossia, upper airway obstruction, and hypothyroidism (see
Chapter 98.2 ). Atlantoaxial instability, common in trisomy 21, has been linked
to cervical dislocation and spinal cord trauma with neck extension during
intubation. Some anesthesiologists recommend extension and flexion lateral
neck films to detect instability before surgery. For children with other known
genetic disorders it is essential to review specific anesthetic considerations.
Preoperative Preparation
Preoperative Fasting
Preoperative fasting guidelines have been developed to reduce the incidence of
aspiration of gastric contents during anesthesia. Aspiration may lead to
laryngospasm, bronchospasm, and postoperative pneumonitis. Aspiration of
gastric contents may be a potentially lethal complication in children with chronic
lung disease or critical illness. Table 74.6 lists preoperative fasting guidelines
(e.g., nothing by mouth, or nil per os [NPO] status). Clear, sweet liquids (e.g.,
Pedialyte, 5% dextrose in water [D5W]) facilitate gastric emptying, prevent
hypoglycemia, and may be given up to 2 hr before anesthesia. Breast milk may
be given to infants up to 4 hr before surgery. Solids should be avoided for 6-8 hr
before surgery. Many conditions delay gastric emptying and may require
prolonged periods of fasting.
Table 74.6
General Anesthesia
Analgesia
Pediatric anesthesiologists are responsible for providing analgesia to children for
procedures within operating room (OR) and non-OR settings (Table 74.7 ).
Multimodal techniques exist to provide pain relief during operative procedures
for children of all ages, including critically ill infants. Effective analgesia is
essential to blunt physiologic responses to painful stimuli (surgery) and
modulate the deleterious physiologic and metabolic consequences. The response
to painful and stressful stimuli may provoke systemic inflammatory response
syndrome (SIRS) , which has been linked to increased catabolism, physiologic
instability, and mortality (see Chapter 88 ).
Table 74.7
Definitions of Anesthesia Care
Monitored Anesthesia Care
Deep Sedation
General Anesthesia
Regional Anesthesia
Local Anesthesia
No Anesthesiologist
Table 74.8
Selected Drugs Used in Anesthesia
Table 74.9
Intraoperative Pediatric Fluid Replacement
Third space interstitial fluid losses should be replaced with isotonic salt
solutions. For smaller operations, such as herniorrhaphy, pyloromyotomy, and
minor procedures, fluid replacement at 3-5 mL/kg/hr is indicated for insensible
losses. Complex abdominal or thoracic procedures with large insensible losses
may require an additional 8-10 mL/kg/hr of IV fluid replacement. Crystalloid
solution is indicated for blood loss as a 3 : 1 ratio. Allogenic blood products
should be replaced as a 1 : 1 ratio. Colloid (albumin) administration also
decreases the amount of crystalloid replacement needed for blood loss. During
large-volume transfusions, active fluid warming should be performed to prevent
hypothermia. With major surgery and resultant SIRS, capillary integrity is lost
even for the shortest-acting muscle relaxant, rocuronium, can take several
minutes. An intubating dose of rocuronium to rapidly induce paralysis in
emergency situations may not spontaneously reverse for 20 min or longer
(compared with about 3 min for succinylcholine). The effects of long-acting,
nondepolarizing NMBAs (vecuronium, pancuronium) are invariably reversed.
Residual NMB is common despite reversal with these agents. Sugammadex is
an alternative reversal agent that has a very low rate of residual NMB. Its
mechanism of action involves noncompetitive antagonism through encapsulation
of neuromuscular agents.
Table 74.10
Postanesthesia Recovery Scores
Postanesthetic Complications
Respiratory insufficiency following general anesthesia is common. Prolonged
emergence from anesthesia and respiratory depression may be caused by the
residual effects of opioids, hypnotic agents, or NMBAs. Pain may also cause
significant hypoventilation, especially after thoracic or abdominal surgery.
Delayed emergence from anesthesia may result from retention of inhaled
anesthetics worsened by hypoventilation. Hypothermia, especially in neonates,
delays metabolism and excretion of anesthetics and prolongs NMB.
Hypoventilation after surgery is associated with the development of atelectasis .
Microatelectasis may lead to postoperative infections. When airway obstruction
is present, maintenance of airway patency may necessitate oropharyngeal or
nasopharyngeal airway placement. In the setting of profound respiratory
depression, endotracheal intubation and mechanical ventilation may be
indicated.
Opioid reversal with naloxone may be indicated in rare instances when
excessive opioid effect is suspected. However, naloxone reverses both the
respiratory depressant and the analgesic effects of opioids. Following naloxone
reversal, a somnolent child with respiratory depression may experience increased
pain. Opioid reversal requires bedside attention by the physician to monitor the
and/or specially trained, experienced, credentialed, and qualified physicians.
Table 75.1
Systematic Approach to Sedation in Children
Comprehensive medical history and organ system assessment, anticipating
underlying medical problems that predispose the patient to anesthetic
complications
Careful physical examination focused on the cardiorespiratory system and
airway
Appropriate fasting
Informed consent
Pediatric drug dosing (mg/kg)
Appropriately sized equipment
Documentation of vital signs and condition on a time-based record
Rapid response (“code”) team to respond to emergencies with “crash cart”
Fully equipped and staffed recovery area
Discharge criteria documenting recovery from sedation
Table 75.2
Bibliography
Coté CJ, Wilson S. Guidelines for monitoring and management
of pediatric patients before, during, and after sedation for
diagnostic and therapeutic procedures: update 2016.
Pediatrics . 2016;138(1):e20161212.
Cramton REM, Gruchala NE. Managing procedural pain in
pediatric patients. Curr Opin Pediatr . 2012;24:530–538.
Doctor K, Roback MG, Teach SJ. An update on pediatric
Pain Categories and Characteristics
PAIN DEFINITION AND
CHARACTERISTICS
CATEGORY EXAMPLES
Somatic Pain resulting In skin and superficial structures: sharp, pulsatile, well localized
from injury to or In deep somatic structures: dull, aching, pulsatile, not well localized
inflammation of
tissues (e.g.,
skin, muscle,
tendons, bone,
joints, fascia,
vasculature)
Examples: burns,
lacerations,
fractures,
infections,
inflammatory
conditions
Visceral Pain resulting Aching and cramping; nonpulsatile; poorly localized (e.g., appendiceal
from injury to or pain perceived around umbilicus) or referred to distant locations (e.g.,
inflammation of angina perceived in shoulder)
viscera
Examples:
angina, hepatitic
distention, bowel
distention or
hypermobility,
pancreatitis
Neuropathic Pain resulting Spontaneous; burning; lancinating or shooting; dysesthesias (pins and
from injury to, needles, electrical sensations); hyperalgesia (amplification of noxious
inflammation of, stimuli); hyperpathia (widespread pain in response to a discrete noxious
or dysfunction of stimulus); allodynia (pain in response to nonpainful stimulation); pain may
the peripheral or be perceived distal or proximal to site of injury, usually corresponding to
central nervous innervation pathways (e.g., sciatica)
system
Examples:
complex regional
pain syndrome
(CRPS),
phantom limb
pain, Guillain-
Barré syndrome,
sciatica
Table 76.2
Pain Measurement Tools
AGE VALIDATION
NAME FEATURES ADVANTAGES LIMITATIONS
RANGE AND USES
Visual Analog Horizontal 10-cm 6-8 yr and Good Acute pain Cannot be used
Scale (VAS) line; subject marks a older psychometric Surgical in younger
spot on the line properties; pain children or in
between anchors of validated for Chronic those with
“no pain” (or neutral research pain cognitive
face) and “most pain purposes limitations
imaginable” (or sad Requires
face) language skills
and numerical
processing;
upper anchor of
“most pain”
requires an
experiential
reference point
that is lacking in
many children.
Likert Scale Integers from 0-10, 6-8 yr and Good Acute pain Same as for VAS.
inclusive, older psychometric Surgical
corresponding to a properties; pain
range from no pain to validated for Chronic
most pain research pain
purposes
Faces Scales Subjects rate their 4 yr and Can be used at Acute pain Choice of “no pain”
(e.g., FACES-R, pain by identifying older younger ages Surgical face affects responses
Wong-Baker, with line drawings of than VAS and pain (neutral vs smiling);
Oucher, Bieri, faces, or photos of Likert not culturally
McGrath scales) children universal.
Behavioral or Scoring of observed Some work May be used in FLACC, Nonspecific;
combined behaviors (e.g., facial for any ages; both infants and N-PASS: overrates pain in
behavioral- expression, limb some work nonverbal Acute pain toddlers and
physiologic movement) ± heart for specific children Surgical preschool children;
scales (e.g., rate and blood age-groups, pain underrates persistent
FLACC, N- pressure including pain; some measures
PASS, preterm are convenient, but
CHEOPS, OPS, infants others require
FACS, NIPS) videotaping and
complex processing;
vital sign changes
unrelated to pain can
occur and may affect
total score.
Autonomic Scores changes in All ages Can be used at Nonspecific; vital
measures (e.g., heart rate, blood all ages; useful sign changes
heart rate, blood pressure, or measures for patients unrelated to pain may
pressure, heart of heart rate receiving occur, and may
rate spectral variability (e.g., mechanical artifactually increase
analyses) “vagal tone”) ventilation or decrease score.
Hormonal- Plasma or salivary All ages Can be used at Nonspecific; changes
metabolic sampling of “stress” all ages unrelated to pain can
measures hormones (e.g., occur; inconvenient;
cortisol, epinephrine) cannot provide “real-
time” information;
standard normal
values not available
for every age bracket.
Behavioral Changes
Table 76.4
Table 76.6
Pediatric Dosage Guidelines for Opioid Analgesics
EQUI-
ANALGESIC PARENTERAL DOSING IV:PO DOSE ORAL DOSING
DRUG DOSES RATIO
IV Oral <50 kg >50 kg <50 kg >50 kg
Fentanyl 10 µg 100 0.5-1 0.5-1 Oral Oral Transdermal
µg µg/kg µg/kg transmucosal: transmucosal: patches
q1-2h q1-2h 1 : 10 10 µg/kg available;
0.5-1.5 0.5-1.5 Transdermal: Transdermal: patch reaches
µg/kg/hr µg/kg/hr 1 : 1 12.5-50 µg/hr steady state at
24 hr and
should be
changed q72h
Hydrocodone N/A 1.5 mg N/A N/A N/A 0.15 mg/kg 10 mg
Methadone 1 mg 2 mg 0.1 mg/kg 0.1 mg/kg 1 : 2 0.2 mg/kg q8-12h 2.5 mg TID
q8-24h q8-24h PO; available as
liquid or tablet
Morphine 1 mg 3 mg 0.05 Bolus: 5-8 1 : 3 Immediate Immediate
mg/kg mg q2-4h release: 0.3 release:
q2-4h mg/kg q3-4h 15-20 mg
0.01- Sustained q3-4h
0.03 release: Sustained
mg/kg/hr 20-35 kg: 10- release:
15 mg q8-12h 30-90 mg
35-50 kg: 15- q8-12h
30 mg q8-12h
Table 76.7
Management of Opioid-Induced Adverse Effects
Respiratory Naloxone: 0.01-0.02 mg/kg up to a full reversal dose of 0.1 mg/kg. May be given IV, IM,
depression SC, or via ET.
The full reversal dose should initially be used for apnea in opioid-naive patients. In opioid-
tolerant patients, a reduced dose should be given and titrated up slowly to treat symptoms
but prevent acute withdrawal.
Ventilation may need to be supported during this process.
Dose may be repeated every 2 min to a total of 10 mg.
Adult maximum dose is 2 mg/dose. Give with caution to patients who are receiving long-
term opioid therapy, as it may precipitate acute withdrawal.
Duration of effect is 1-4 hr; therefore close observation for re-narcotization is essential to
prevent re-narcotization.
Excessive sedation Methylphenidate * : 0.3 mg/kg per dose PO (typically 10-20 mg/dose to a teenager) before
without evidence of breakfast and lunch. Do not administer to patients receiving clonidine, because
respiratory dysrhythmias may develop.
depression Dextroamphetamine : 2.5-10 mg on awakening and at noon. Not for use in young children
or in patients with cardiovascular disease or hypertension.
Modafinil: Pediatric dose not established. May be useful in selected patients. Typical adult
dose: 50-200 mg/day.
Change opioid or decrease the dose.
Nausea and Metoclopramide † : 0.15 mg/kg IV up to 10 mg/dose q6-12h for 24 hr.
vomiting Trimethobenzamide: PO or PR if weight <15 kg, 100 mg q6h; if >15 kg, 200 mg q6h. (Note:
Suppository contains benzocaine 2%.) Not for use in newborn infants or premature
infants.
5-HT3 receptor blockers:
Ondansetron: 0.15 mg/kg up to 8 mg IV q6-8h not to exceed 32 mg/day (also available as a
sublingual tablet).
Granisetron: 10 to 20 µg/kg IV q12-24h.
Prochlorperazine * (Compazine): >2 yr or >20 kg, 0.1 mg/kg per dose q8h IM or PO up to
10 mg/dose.
Change opioid.
Pruritus Hydroxyzine : 0.5 mg/kg PO q6h.
Nalbuphine: 0.1 mg/kg IV q6h for pruritus caused by intraaxial opioids, especially fentanyl.
Administer slowly over 15-20 min. May cause acute reversal of systemic µ-receptor
effects and leave κ-agonism intact.
Naloxone: 0.003 to 0.1 mg/kg/hr IV infusion (titrate up to decrease pruritus and reduce
infusion if pain increases).
Ondansetron: 0.05 to 0.1 mg/kg IV or PO q8h.
Cyproheptadine † : 0.1-0.2 mg/kg PO q8-12h. Maximum dose 12 mg.
Change opioid.
Constipation Encourage water consumption, high-fiber diet, and vegetable fiber.
Bulk laxatives: Metamucil, Maltsupex.
Lubricants: Mineral oil 15-30 mL PO qd as needed (not for use in infants because of
aspiration risk).
Surfactants: Sodium docusate (Colace):
<3 yr: 10 mg PO q8h
3-6 yr: 15 mg PO q8h
6-12 yr: 50 mg PO q8h
>12 yr: 100 mg PO q8h
Stimulants:
Bisacodyl suppository (Dulcolax):
<2 yr: 5 mg PR qhs
>2 yr: 10 mg PR qhs
Senna syrup (218 mg/5 mL): >3 yr: 5 mL qhs.
Enema: Fleet hypertonic phosphate enema (older children; risk of hyperphosphatemia).
Electrolytic/osmotic: Milk of magnesia; for severe impaction: polyethylene glycol
(GoLYTELY, MiraLax).
Methylnaltrexone is an opioid antagonist that works in the colon and does not cross the
blood-brain barrier to reverse analgesia; given as subcutaneous injection every day or
every other day (0.15 mg/kg) and is effective in producing stool in 30-60 min in most
patients.
Urinary retention Straight catheterization, indwelling catheter.
*
Avoid in patients taking monoamine oxidase inhibitors.
† May be associated with extrapyramidal side effects, which may be more often seen in children
than in adults.
ET, Endotracheal tube; IV, intravenously; IM, intramuscularly; PO, orally; PR, rectally; SC,
subcutaneously.
Modified from Burg FD, Ingelfinger JR, Polin RA, et al, editors: Current pediatric therapy, ed 18,
Philadelphia, 2006, Saunders/Elsevier, p 16.
Table 76.8
Equianalgesic Doses and Half-Life (T1/2β ) of Some
Commonly Used Opioids
† Only part of its analgesic action results from action on µ-opioid receptors.
NOTES:
4. When starting opioid therapy for chronic pain, clinicians should prescribe
immediate-release opioids instead of extended-release/long-acting
(ER/LA) opioids.
5. When opioids are started, clinicians should prescribe the lowest effective
dosage. Clinicians should use caution when prescribing opioids at any
dosage, should carefully reassess evidence of individual benefits and
risks when increasing dosage to ≥50 morphine milligram equivalents
(MME)/day, and should avoid increasing dosage to ≥90 MME/day or
carefully justify a decision to titrate dosage to ≥90 MME/day.
6. Long-term opioid use often begins with treatment of acute pain. When
opioids are used for acute pain, clinicians should prescribe the lowest
effective dose of immediate-release opioids and should prescribe no
greater quantity than needed for the expected duration of pain severe
enough to require opioids. Three days or less will often be sufficient;
more than seven days will rarely be needed.
7. Clinicians should evaluate benefits and harms with patients within 1 to 4
weeks of starting opioid therapy for chronic pain or of dose escalation.
Clinicians should evaluate benefits and harms of continued therapy with
patients every 3 months or more frequently. If benefits do not outweigh
harms of continued opioid therapy, clinicians should optimize other
therapies and work with patients to taper opioids to lower dosages or to
taper and discontinue opioids.
From Dowell D, Haegerich TM, Chou R: CDC guideline for prescribing opioids
for chronic pain—United States, 2016, MMWR 65(1):1–49, 2016.
Because of the high risk of adverse side effects (respiratory depression), the
FDA has issued contraindications for the pediatric use of codeine and tramadol
(Table 76.10 ).
Table 76.10
Summary of FDA Recommendations
• Use of codeine to treat pain or cough in children <12 yr old is
contraindicated.
• Use of tramadol to treat pain in children <12 yr old is contraindicated.
• Use of tramadol for treatment of pain after tonsillectomy or adenoidectomy
in patients <18 yr old is contraindicated. (Codeine was already
contraindicated in such patients).
• Use of codeine or tramadol in children 12-18 yr old who are obese or who
have an increased risk of serious breathing problems, such as those with
obstructive sleep apnea or severe lung disease, is not recommended.
• Use of codeine or tramadol in breastfeeding women should be avoided.
From The Medical Letter: FDA warns against use of codeine and tramadol in
children and breastfeeding women, Med Lett 59(1521):86–88, 2017.
Local Anesthetics
Local anesthetics are widely used in children for topical application, cutaneous
infiltration, peripheral nerve block, neuraxial blocks (intrathecal or epidural
infusions), and IV infusions (Table 76.11 ) (see Chapter 74 ). Local anesthetics
can be used with excellent safety and effectiveness. Local anesthetics interfere
with neural transmission by blocking neuronal sodium channels. Excessive
systemic dosing can cause seizures, CNS depression, and (by cardiac and
arteriolar sodium channel blockade) hypotension, arrhythmias, cardiac
depression, and cardiovascular collapse. Local anesthetics therefore require a
strict maximum dosing schedule. Pediatricians should be aware of the need to
calculate these doses and adhere to guidelines.
Table 76.11
Topical Pharmacologic Management of Acute Pain in
Children
Lidocaine 70 mg and tetracaine Age ≥3 yr: apply patch 20–30 min needed to achieve maximum effect
70 mg (Synera patch)
Tetracaine 4% (Ametop) >1 mo and <5 yr: 30 min before venipuncture
apply 1 tube of gel (1 45 min before intravenous cannulation
g)
>5 yr: apply up to 5
tubes of gel (5 g)
WOUNDS
Lidocaine, epinephrine, Age ≥1 yr: apply to wound 20 min needed for maximum effect
tetracaine (LET) solution or gel*
* Also referred to as ALA on the basis of alternative names for the constituents: adrenaline,
lignocaine, amethocaine. These mixtures are locally made by hospital formularies, with a common
formula being lidocaine 4% plus epinephrine 0⋅1% plus tetracaine 0⋅5%. The cocaine-based
formulation was historically avoided on wounds of digits, ears, penis, nose, mucous membranes,
close to the eye, or deep wounds involving bone, cartilage, tendon, or vessels. The lidocaine-
based formulation can be used in such settings.
Adapted from Krauss BS, Calligaris L, Green SM, Barbi E: Current concepts in management of
pain in children in the emergency department, Lancet 387:83–92, 2016.
RECOMMENDED STAGE OF
MEDICATION CLASS/DRUG
TREATMENT
ANTIDEPRESSANTS
Tricyclics (e.g., amitriptyline, nortriptyline) 1st or 2nd
Serotonin and norepinephrine reuptake inhibitors (e.g., duloxetine, 1st or 2nd
venlafaxine)
ANTICONVULSANTS
Pregabalin 1st or 2nd
Gabapentin 1st or 2nd
Lamotrigine 2nd or 3rd (in pain after stroke)
Valproate 3rd
OPIOIDS *
Levorphanol
MISCELLANEOUS
Cannabinoids 2nd (in multiple sclerosis)
Mexiletine 3rd
* 2nd or 3rd treatment stage (no specification).
Adapted from Freynhagen R, Bennett MI: Diagnosis and management of neuropathic pain, BMJ
339:b3002, 2009.
Patients who present with mild to moderate pain should be treated with a
nonopioid.
Step 2
Patients who present with moderate to severe pain or for whom the step 1
regimen fails should be treated with an oral opioid for moderate pain
combined with a nonopioid analgesic.
Step 3
Patients who present with very severe pain or for whom the step 2 regimen
fails should be treated with an opioid used for severe pain, with or
without a nonopioid analgesic.
Oral medications are the first line of analgesic treatment. Because NSAIDs
affect platelet adhesiveness, they are typically not used. Opioid therapy is the
preferred approach for moderate or severe pain. Nonopioid analgesics are used
for mild pain, a weak opioid is added for moderate pain, and strong opioids are
administered for more severe pain. Adjuvant analgesics can be added, and side
effects and comorbid symptoms are actively managed. Determining the type and
sources of the pain will help develop an effective analgesic plan. Certain
treatments, such as the chemotherapeutic agent vincristine, are associated with
neuropathic pain. Such pain might require anticonvulsants or TCAs. Organ-
stretching pain from tumor growth within an organ might require strong opioids
and/or radiation therapy if the tumor is radiosensitive. Organ obstruction, such as
and aliphatic hydrocarbons were significant causes of mortality.
Table 77.1
Common Agents Potentially Toxic to Young Children (<6 yr) in Small Doses*
SUBSTANCE TOXICITY
Aliphatic hydrocarbons (e.g., gasoline, kerosene, lamp Acute lung injury
oil)
Antimalarials (chloroquine, quinine) Seizures, dysrhythmias
Benzocaine Methemoglobinemia
β-Adrenergic receptor blockers † Bradycardia, hypotension
Calcium channel blockers Bradycardia, hypotension, hyperglycemia
Camphor Seizures
Caustics (pH <2 or >12) Airway, esophageal and gastric burns
Clonidine Lethargy, bradycardia, hypotension
Diphenoxylate and atropine (Lomotil) CNS depression, respiratory depression
Hypoglycemics, oral (sulfonylureas and meglitinides) Hypoglycemia, seizures
Laundry detergent packets (pods) Airway issues, respiratory distress, altered mental status
Lindane Seizures
Monoamine oxidase inhibitors Hypertension followed by delayed cardiovascular
collapse
Methyl salicylate Tachypnea, metabolic acidosis, seizures
Opioids (especially methadone, buprenorphine) CNS depression, respiratory depression
Organophosphate pesticides Cholinergic crisis
Phenothiazines (especially chlorpromazine, Seizures, dysrhythmias
thioridazine)
Theophylline Seizures, dysrhythmias
Tricyclic antidepressants CNS depression, seizures, dysrhythmias, hypotension
* ”Small dose” typically implies 1 or 2 pills or 5 mL.
† Lipid-soluble β-blockers (e.g., propranolol) are more toxic than water-soluble β-blockers (e.g.,
atenolol).
CNS, Central nervous system.
Symptoms
Obtaining a description of symptoms experienced after ingestion, including their
timing of onset relative to the time of ingestion and their progression, can
generate a list of potential toxins and help anticipate the severity of the ingestion.
Coupled with physical exam findings, reported symptoms assist practitioners in
identifying toxidromes, or recognized poisoning syndromes, suggestive of
toxicity from specific substances or classes of substances (Tables 77.2 to 77.4 ).
Table 77.2
Selected Historical and Physical Findings in Poisoning
SIGN TOXIN
ODOR
Bitter almonds Cyanide
Acetone Isopropyl alcohol, methanol, paraldehyde, salicylates
Rotten eggs Hydrogen sulfide, sulfur dioxide, methyl mercaptans (additive to natural gas)
Wintergreen Methyl salicylate
Garlic Arsenic, thallium, organophosphates, selenium
OCULAR SIGNS
Miosis Opioids (except propoxyphene, meperidine, and pentazocine), organophosphates and other
cholinergics, clonidine, phenothiazines, sedative-hypnotics, olanzapine
Mydriasis Anticholinergics (e.g., antihistamines, TCAs, atropine), sympathomimetics (cocaine,
amphetamines, PCP), post–anoxic encephalopathy, opiate withdrawal, cathinones, MDMA
Nystagmus Anticonvulsants, sedative-hypnotics, alcohols, PCP, ketamine, dextromethorphan
Lacrimation Organophosphates, irritant gas or vapors
Retinal hyperemia Methanol
CUTANEOUS SIGNS
Diaphoresis Cholinergics (organophosphates), sympathomimetics, withdrawal syndromes
Alopecia Thallium, arsenic
Erythema Boric acid, elemental mercury, cyanide, carbon monoxide, disulfiram, scombroid,
anticholinergics, vancomycin
Cyanosis Methemoglobinemia (e.g., benzocaine, dapsone, nitrites, phenazopyridine), amiodarone,
(unresponsive to silver
oxygen)
ORAL SIGNS
Salivation Organophosphates, salicylates, corrosives, ketamine, PCP, strychnine
Oral burns Corrosives, oxalate-containing plants
Gum lines Lead, mercury, arsenic, bismuth
GASTROINTESTINAL SIGNS
Diarrhea Antimicrobials, arsenic, iron, boric acid, cholinergics, colchicine, opioid withdrawal
Hematemesis Arsenic, iron, caustics, NSAIDs, salicylates
Constipation Lead
CARDIAC SIGNS
Tachycardia Sympathomimetics, anticholinergics, antidepressants, antipsychotics, methylxanthines
(theophylline, caffeine), salicylates, cellular asphyxiants (cyanide, carbon monoxide,
hydrogen sulfide), withdrawal (ethanol, sedatives, clonidine, opioids), serotonin syndrome,
neuroleptic malignant syndrome, MDMA, cathinones
Bradycardia β-Blockers, calcium channel blockers, digoxin, clonidine, organophosphates, opioids,
sedative-hypnotics
Hypertension Sympathomimetics, anticholinergics, monoamine oxidase inhibitors, serotonin syndrome,
neuroleptic malignant syndrome, clonidine withdrawal
Hypotension β-Blockers, calcium channel blockers, cyclic antidepressants, iron, antipsychotics,
barbiturates, clonidine, opioids, arsenic, amatoxin mushrooms, cellular asphyxiants (cyanide,
carbon monoxide, hydrogen sulfide), snake envenomation
RESPIRATORY SIGNS
Depressed Opioids, sedative-hypnotics, alcohol, clonidine, barbiturates
respirations
Tachypnea Salicylates, sympathomimetics, caffeine, metabolic acidosis, carbon monoxide, hydrocarbon
aspiration
CENTRAL NERVOUS SYSTEM SIGNS
Ataxia Alcohols, anticonvulsants, sedative-hypnotics, lithium, dextromethorphan, carbon monoxide,
inhalants
Coma Opioids, sedative-hypnotics, anticonvulsants, antidepressants, antipsychotics, ethanol,
anticholinergics, clonidine, GHB, alcohols, salicylates, barbiturates
Seizures Sympathomimetics, anticholinergics, antidepressants (especially TCAs, bupropion,
venlafaxine), cholinergics (organophosphates), isoniazid, camphor, lindane, salicylates, lead,
nicotine, tramadol, water hemlock, withdrawal
Delirium/psychosis Sympathomimetics, anticholinergics, LSD, PCP, hallucinogens, lithium, dextromethorphan,
steroids, withdrawal, MDMA, cathinones
Peripheral Lead, arsenic, mercury, organophosphates, nicotine
neuropathy
GHB, γ-Hydroxybutyrate; LSD, lysergic acid diethylamide; MDMA, 3,4-
methylenedioxymethamphetamine (Ecstasy); NSAIDs, nonsteroidal antiinflammatory drugs; PCP,
phencyclidine; TCAs, tricyclic antidepressants.
Table 77.3
Recognizable Poison Syndromes (“Toxidromes”)
SIGNS
POSSIBLE
TOXIDROME Mental Bowel
Vital Signs Pupils Skin Other TOXINS
Status Sounds
Sympathomimetic Hypertension, Agitation, Dilated Diaphoretic Normal to Amphetamines,
tachycardia, psychosis, increased cocaine, PCP,
hyperthermia delirium, bath salts
violence (cathinones),
ADHD
medication
Anticholinergic Hypertension, Agitated, Dilated Dry, hot Diminished Ileus urinary Antihistamines,
tachycardia, delirium, retention TCAs, atropine,
hyperthermia coma, jimsonweed
seizures
Cholinergic Bradycardia, Confusion, Small Diaphoretic Hyperactive Diarrhea, Organophosphates
BP, and temp coma, urination, (insecticides,
typically fasciculations bronchorrhea, nerve agents),
normal bronchospasm, carbamates
emesis, (physostigmine,
lacrimation, neostigmine,
salivation pyridostigmine)
Alzheimer
medications,
myasthenia
treatments
Opioids Respiratory Depression, Pinpoint Normal Normal to Methadone,
depression coma, decreased buprenorphine,
bradycardia, euphoria morphine,
hypotension, oxycodone,
hypothermia heroin, etc.
Sedative- Respiratory Somnolence, Small or Normal Normal Barbiturates,
hypnotics depression, coma normal benzodiazepines,
HR normal to ethanol
decreased, BP
normal to
decreased,
temp normal
to decreased
Serotonin Hyperthermia, Agitation, Dilated Diaphoretic Increased Neuromuscular SSRIs, lithium,
syndrome (similar tachycardia, confusion, hyperexcitability: MAOIs, linezolid,
findings with hypertension coma clonus, tramadol,
neuroleptic or hyperreflexia meperidine,
malignant hypotension (lower > upper dextromethorphan
syndrome) (autonomic extremities)
instability)
Salicylates Tachypnea, Agitation, Normal Diaphoretic Normal Nausea, Aspirin and
hyperpnea, confusion, vomiting, aspirin-containing
tachycardia, coma tinnitus, ABGs products, methyl
hyperthermia with primary salicylate
respiratory
alkalosis and
primary
metabolic
acidosis; tinnitus
or difficulty
hearing
Withdrawal Tachycardia, Agitation, Dilated Diaphoretic Increased Lack of access to
(sedative- tachypnea, tremor, ethanol,
hypnotic) hyperthermia seizure, benzodiazepines,
hallucinosis, barbiturates,
delirium GHB, or
tremens excessive use of
flumazenil
Withdrawal Tachycardia Restlessness, Dilated diaphoretic Hyperactive Nausea, Lack of access to
(opioid) anxiety vomiting, opioids or
diarrhea excessive use of
naloxone
ABGs, Arterial blood gases; ADHD, attention-deficit/hyperactivity disorder; BP, blood pressure;
GHB, γ-hydroxybutyrate; HR, heart rate; MAOIs, monoamine oxidase inhibitors; PCP,
phencyclidine; SSRIs, selective serotonin reuptake inhibitors; temp, temperature; TCAs, tricyclic
antidepressants.
Table 77.4
Mini-Toxidromes
TOXIDROME SYMPTOMS AND SIGNS EXAMPLES
α1 -Adrenergic CNS depression, tachycardia, Chlorpromazine, quetiapine, clozapine, olanzapine,
receptor miosis risperidone
antagonists
α2 -Adrenergic CNS depression, bradycardia, Clonidine, oxymetazoline, tetrahydrozoline, tizanidine,
receptor agonist hypertension (early), dexmedetomidine
hypotension (late), miosis
Clonus/myoclonus CNS depression, myoclonic Carisoprodol, lithium, serotonergic agents, bismuth,
jerks, clonus, hyperreflexia organic lead, organic mercury, serotonin or neuroleptic
malignant syndrome
Sodium channel CNS toxicity, wide QRS Cyclic antidepressants and structurally related agents,
blockers propoxyphene, quinidine/quinine, amantadine,
antihistamines, bupropion, cocaine
Potassium channel CNS toxicity, long QT interval Antipsychotics, methadone, phenothiazines
blockers
Cathinones, Hyperthermia, tachycardia, See Chapter 140 .
synthetic delirium, agitation, mydriases
cannabinoids
CNS, Central nervous system.
From Ruha AM, Levine M: Central nervous system toxicity. Emerg Med Clin North Am 32(1):205–
221, 2014, p 208.
M ethanol, metformin
U remia
D iabetic ketoacidosis
P ropylene glycol
I soniazid, iron, massive ibuprofen
L actic acidosis
E thylene glycol
S alicylates
C ellular asphyxiants (cyanide, carbon monoxide, hydrogen sulfide)
A lcoholic ketoacidosis
T ylenol (clinical significance depends upon presence or absence of liver
injury)
Hyperglycemia
Salicylates (early)
Calcium channel blockers
Caffeine
Hypocalcemia
Ethylene glycol
Fluoride
Rhabdomyolysis
Digoxin
Lithium
QRS Prolongation
Tricyclic antidepressants
Diphenhydramine
Carbamazepine
Cardiac glycosides
Chloroquine, hydroxychloroquine
Cocaine
Lamotrigine
Quinidine, quinine, procainamide, disopyramide
Phenothiazines
Propoxyphene
Propranolol
Bupropion, venlafaxine (rare)
QTc Prolongation*
Amiodarone
Antipsychotics (typical and atypical)
Arsenic
Cisapride
Citalopram
Clarithromycin, erythromycin
Disopyramide, dofetilide, ibutilide
Fluconazole, ketoconazole, itraconazole
Methadone
Pentamidine
Phenothiazines
Sotalol
* This is a select list of important toxins, other medications are also associated
Principles of Management
The principles of management of the poisoned patient are supportive care,
decontamination, directed therapy (antidotes, ILE), and enhanced elimination.
Few patients meet criteria for all these interventions, although clinicians should
consider each option in every poisoned patient so as not to miss a potentially
lifesaving intervention. Antidotes are available for relatively few poisons (Tables
77.7 and 77.8 ), thus emphasizing the importance of meticulous supportive care
and close clinical monitoring.
Table 77.7
Common Antidotes for Poisoning
ADVERSE EFFECTS,
POISON ANTIDOTE DOSAGE ROUTE
WARNINGS, COMMENTS
Acetaminophen N -Acetylcysteine 140 mg/kg loading, PO Vomiting (patient-tailored
(Mucomyst) followed by 70 mg/kg regimens are the norm)
q4h
N -Acetylcysteine 150 mg/kg over 1 hr, IV Anaphylactoid reactions
(Acetadote) followed by 50 mg/kg (most commonly seen with
over 4 hr, followed by loading dose)
100 mg/kg over 16 hr (Higher doses of the infusion
are often recommended
depending on acetaminophen
level or degree of injury)
Anticholinergics Physostigmine 0.02 mg/kg over 5 min; IV/IM Bradycardia, seizures,
may repeat q5-10 min bronchospasm
to 2 mg max Note: Do not use if
conduction delays on ECG.
Benzodiazepines Flumazenil 0.2 mg over 30 sec; if IV Agitation, seizures from
response is inadequate, precipitated withdrawal
repeat q1min to 1 mg (doses over 1 mg)
max Do not use for unknown or
polypharmacy ingestions.
β-Blockers Glucagon 0.15 mg/kg bolus IV Vomiting, relative lack of
followed by infusion of efficacy
0.05-0.15 mg/kg/hr
Calcium channel Insulin 1 unit/kg bolus IV Hypoglycemia
blockers followed by infusion of Follow serum potassium and
0.5-1 unit/kg/hr glucose closely.
Calcium salts Dose depends on the IV
specific calcium salt
Carbon monoxide Oxygen 100% FIO 2 by non- Inhalation Some patients may benefit from
rebreather mask (or ET hyperbaric oxygen (see text).
if intubated)
Cyanide Hydroxocobalamin 70 mg/kg (adults: 5 g) IV Flushing/erythema, nausea, rash,
(Cyanokit) given over 15 min chromaturia, hypertension,
headache
Digitalis Digoxin-specific 1 vial binds 0.6 IV Allergic reactions (rare), return of
Fab antibodies mg of digitalis condition being treated with
(Digibind, DigiFab) glycoside; digitalis glycoside
#vials = digitalis
level × weight in
kg/100
Ethylene glycol, Fomepizole 15 mg/kg load; 10 IV Infuse slowly over 30 min.
methanol mg/kg q12h × 4 doses; If fomepizole is not
15 mg/kg q12h until available, can treat with oral
ethylene glycol level is ethanol (80 proof)
<20 mg/dL
Iron Deferoxamine Infusion of 5-15 IV Hypotension (minimized by
mg/kg/hr (max: 6 g/24 avoiding rapid infusion rates)
hr)
Isoniazid (INH) Pyridoxine Empirical dosing: IV May also be used for Gyromitra
70 mg/kg (max mushroom ingestions
dose = 5 g)
If ingested dose is
known: 1 g per
gram of INH
Lead and other BAL (dimercaprol) 3-5 mg/kg/dose q4h, Deep IM Local injection site pain and
heavy metals (e.g., for the 1st day; sterile abscess, vomiting,
arsenic, inorganic subsequent dosing fever, salivation,
mercury) depends on the toxin nephrotoxicity
Caution: prepared in peanut
oil; contraindicated in
patients with peanut allergy
Calcium disodium 35-50 mg/kg/day × 5 IV Vomiting, fever, hypertension,
EDTA days; may be given as arthralgias, allergic reactions,
a continuous infusion local inflammation,
or 2 divided doses/day nephrotoxicity (maintain
adequate hydration; follow UA
and renal function)
Dimercaptosuccinic 10 mg/kg/dose q8h × 5 PO Vomiting, hepatic transaminase
acid (succimer, days, then 10 mg/kg elevation, rash
DMSA, Chemet) q12h × 14 days
Methemoglobinemia Methylene blue, 0.1-0.2 mL/kg (1-2 IV Vomiting, headache, dizziness,
1% solution mg/kg) over 5-10 min; blue discoloration of urine
may be repeated q30-
60 min
Opioids Naloxone 1 mg if patient not IV, Acute withdrawal symptoms
likely to be intranasal, if given to addicted patients
addicted. IO, IM, May also be useful for
0.04-0.4 mg if nebulized clonidine ingestions
possibly addicted; (typically at higher doses)
repeated as
needed; may need
continuous
infusion
Organophosphates Atropine 0.05-0.1 mg/kg IV/ET Tachycardia, dry mouth, blurred
repeated q5-10 min as vision, urinary retention
needed
Pralidoxime (2- 25-50 mg/kg over 5-10 IV/IM Nausea, dizziness, headache,
PAM) min (max: 200 tachycardia, muscle rigidity,
mg/min); can be bronchospasm (rapid
repeated after 1-2 hr, administration)
then q10-12h as
needed
Salicylates Sodium Bolus 1-2 mEq/kg IV Follow potassium closely
bicarbonate followed by and replace as necessary.
continuous infusion Goal urine pH: 7.5-8.0
Sulfonylureas Octreotide and 1-2 µg/kg/dose (adults IV/SC
dextrose 50-100 µg) q6-8h
Tricyclic Sodium Bolus 1-2 mEq/kg; IV Indications: QRS widening (≥110
antidepressants bicarbonate repeated bolus dosing msec), hemodynamic instability;
as needed to keep QRS follow potassium.
<110 msec
BAL, British antilewisite; DMSA, dimercaptosuccinic acid; ECG, electrocardiogram; FIO 2 , fraction
of inspired oxygen; EDTA, ethylenediaminetetraacetic acid; ET, endotracheal tube; IO,
intraosseous; max, maximum; UA, urinalysis.
Table 77.8
Other Antidotes
ANTIDOTES TOXIN OR POISON
Latrodectus antivenin Black widow spider
Botulinum antitoxin Botulinum toxin
Diphenhydramine and/or Dystonic reactions
benztropine
Calcium salts Fluoride, calcium channel blockers
Protamine Heparin
Folinic acid Methotrexate, trimethoprim, pyrimethamine
Crotalidae-specific Fab antibodies Rattlesnake envenomation
Sodium bicarbonate Sodium channel blockade (tricyclic antidepressants, type 1
antiarrhythmics)
Supportive Care
Careful attention is paid first to the “ABCs” of airway, breathing, and
circulation; there should be a low threshold to aggressively manage the airway of
a poisoned patient because of the patient's propensity to quickly become
comatose. In fact, endotracheal intubation is often the only significant
intervention needed in many poisoned patients. An important caveat is the
tachypneic patient with a clear lung examination and normal oxygen saturation.
This should alert the clinician to the likelihood that the patient is compensating
for an acidemia. Paralyzing such a patient and underventilating might prove
fatal. If intubation is absolutely necessary for airway protection or a tiring
patient, a good rule of thumb is to match the ventilatory settings to the patient's
preintubation minute ventilation.
recommended.
Table 77.10
Treatment.
When considering the treatment of a patient poisoned or potentially poisoned
with APAP, and after assessment of the ABCs, it is helpful to place the patient
into one of the following four categories.
1 Prophylactic.
By definition, these patients have a normal aspartate transaminase (AST). If the
APAP level is known and the ingestion is within 24 hr of the level being drawn,
treatment decisions are based on where the level falls on the Rumack-Matthew
nomogram (Fig. 77.1 ). Any patient with a serum APAP level in the possible or
probable hepatotoxicity range per the nomogram should be treated with N -
acetylcysteine (NAC). This nomogram is only intended for use in patients who
present within 24 hr of a single acute APAP ingestion with a known time of
ingestion. If treatment is recommended, they should receive NAC as either oral
Pathophysiology.
SSRIs selectively block the reuptake of serotonin in the CNS. In contrast to
TCAs and atypical antidepressants, SSRIs do not directly interact with other
receptor types.
Table 77.11
Treatment.
Initial management includes a careful assessment for signs and symptoms of
serotonin syndrome and an ECG. Most patients simply require supportive care
and observation until their mental status improves and tachycardia, if present,
resolves. Management of serotonin syndrome is directed by the severity of
symptoms; possible therapeutic interventions include benzodiazepines in mild
cases and intubation, sedation, and paralysis in patients with severe
manifestations (e.g., significant hyperthermia). Because agonism at the 5-HT2A
serotonin receptor is thought to be primarily responsible for the development of
serotonin syndrome, use of the 5-HT2A receptor antagonist cyproheptadine may
also be helpful. Cyproheptadine is only available in an oral form.
Atypical Antidepressants.
The atypical antidepressant class includes agents such as venlafaxine and
duloxetine (SNRIs), bupropion (dopamine, norepinephrine, and some serotonin
reuptake blockade), and trazodone (serotonin reuptake blockade and peripheral
α-receptor antagonism). The variable receptor affinities of these agents lead to
some distinctions in their clinical manifestations and management.
Plants
Exposure to plants, both inside the home and outside in backyards and fields, is
one of the most common causes of unintentional poisoning in children.
Fortunately, the majority of ingestions of plant parts (leaves, seeds, flowers)
result in either no toxicity or mild, self-limiting effects. However, ingestion of
certain plants can lead to serious toxicity (Table 77.12 ).
Table 77.12
Dietary Supplements
Under the 1994 U.S. Dietary Supplement Health and Education Act, a dietary
supplement is a product taken by mouth that contains a dietary ingredient
intended to supplement the diet. These may include vitamins, minerals, herbs or
other botanicals, amino acids, and substances such as enzymes, organ tissues,
glands, and metabolites. Dietary supplements are the most frequently used
complementary therapies for children and adolescents (Table 78.1 ). Some uses
are common and recommended, such as vitamin D supplements for breastfed
infants and probiotics to prevent antibiotic-associated diarrhea, whereas other
uses are more controversial, such as using herbal products to treat otitis media.
Table 78.1
Commonly Used Dietary Supplements in Pediatrics
PRODUCT USES
VITAMINS
B2 (riboflavin) Migraine headache prophylaxis
B6 Pyridoxine-dependent epilepsy; neuropathy; nausea associated with pregnancy
(pyridoxine)
B9 (folate) Prevention of neural tube defects
D Prevention of rickets; treatment of vitamin D deficiencies
Multivitamins General health promotion
MINERALS
Iodine (salt) Prevent goiter and mental retardation
Iron Prevent and treat iron-deficiency anemia
Magnesium Constipation, asthma, migraine prevention
Zinc Diarrhea in nutrient-poor populations
HERBS
Aloe vera Mild burns
Chamomile Mild sedative, dyspepsia
Echinacea Prevention of upper respiratory infections
Ginger Nausea
Lavender Mild sedative
(aromatherapy)
Peppermint Irritable bowel syndrome
Tea tree oil Antibacterial (acne remedies), pediculicide (lice)
OTHER
Melatonin Insomnia
Omega-3 fatty ADHD, allergies, inflammation, anxiety and mood disorders
acids
Probiotics Antibiotic-associated diarrhea; Clostridium difficile –associated diarrhea; constipation; irritable
bowel syndrome; pouchitis; inflammatory bowel disorders
ADHD, Attention-deficit/hyperactivity disorder.
In the United States, dietary supplements do not undergo the same stringent
evaluation and postmarketing surveillance as prescription medications. Although
they may not claim to prevent or treat specific medical conditions, product labels
may make structure-function claims. For example, a label may claim that a
product “promotes a healthy immune system,” but it may not claim to cure the
common cold.
According to the 2012 National Health Interview Survey, 5% of U.S. children
used non-vitamin/mineral dietary supplements. (e.g., fish oil, melatonin,
prebiotics, probiotics) Use of dietary supplements is most common among
children whose families have higher income and education and whose parents
use supplements, among older children, and among those with chronic
conditions.
Despite this widespread use, many patients and their parents who use dietary
supplements do not talk with their physician about their use. Several guidelines
have called for more complete dietary supplement history taking by healthcare
professionals. The Joint Commission recommends that clinicians routinely ask
patients about their use of dietary supplements and include this information as
part of the medication reconciliation process.
Table 78.2
Clinical Toxicity of Selected Herbs
COMMON BOTANICAL
THERAPEUTIC USES POTENTIAL TOXICITY
NAME NAME
Aconite Aconitum spp. Sedative, analgesic, antihypertensive Cardiac arrhythmias
(monkshood,
wolfsbane)
Aloe Aloe spp. Burns, skin diseases Nephritis, GI upset
Betel nut Areca catechu Mood elevation Bronchoconstriction, oral cancers
Bloodroot Sanguinaria Emetic, cathartic, eczema GI upset, vertigo, visual
canadensis disturbances
Chaparral Larrea Aging, free radical scavenging Hepatitis
(greasewood) tridentata
Compound Q Trichosanthes Anthelmintic, cathartic Diarrhea, hypoglycemia, CNS
kirilowii toxicity
Dandelion Taraxacum Diuretic, heartburn remedy Anaphylaxis
officinale
Figwort (xuan Scrophularia Antiinflammatory, antibacterial Cardiac stimulation
shen) spp.
Ginseng Panax Antihypertensive, aphrodisiac, Ginseng abuse syndrome
quinquefolium stimulant, mood elevation, digestive aid
Goldenseal Hydrastis Digestive aid, mucolytic, anti-infective Uterine, cardiac stimulation; GI
canadensis upset, leukopenia
Hellebore Veratrum spp. Antihypertensive Vomiting, bradycardia, hypotension
Hyssop Hyssopus Asthma, mucolytic Seizures
officinalis
Juniper Juniperus Hallucinogen GI upset, seizures, renal injury,
communis hypotension, bradycardia
Kava kava Piper Sedative Inebriation
methysticum
Kombucha Stimulant Metabolic acidosis, hepatotoxicity,
death
Licorice Glycyrrhiza Indigestion Mineralocorticoid effects
spp.
Lily of the valley Convallaria Cardiotonic GI (nausea, vomiting), cardiac
spp. arrhythmias
Linn (willow) Salix caprea Purgative Hemolysis with glucose-6-
phosphate dehydrogenase
deficiency
Lobelia (Indian Lobelia spp. Stimulant Nicotine intoxication
tobacco)
Ma Huang Ephedra Stimulant Sympathetic crisis, especially with
sinica monamine oxidase inhibitors
Mandrake Mandragora Hallucinogen Anticholinergic syndrome
officinarum
Mormon tea Ephedra Stimulant, asthma, antipyretic Hypertension, sympathomimetic
nevadensis
Nutmeg Myristica Hallucinogen, abortifacient Hallucinations, GI upset
fragrans
Oleander Nerium Cardiac stimulant Cardiac arrhythmias
oleander
Passionflower Passiflora Hallucinogen Hallucinations, seizures,
caeruliea hypotension
Periwinkle Vinca spp. Antiinflammatory, diabetes Alopecia, seizures, hepatotoxicity
Pokeweed Phytolacca Arthritis, chronic pain GI upset, seizures, death
spp.
Sabah Sauropus Weight loss, vision Pulmonary injury
androgynus
Sage Salvia spp. CNS stimulant Seizures
Snakeroot Rauwolfia Sedative, antihypertensive Bradycardia, coma
serpentina
Squill Urginea Arthritis, cardiac stimulant Seizures, arrhythmias, death
maritima
Thorn apple Datura Hallucinations Anticholinergic
(jimsonweed) stramonium
Tonka bean Dipteryx Anticoagulant Bleeding diathesis
odorata
Valerian root Valeriana spp. Sedative Sedation, obtundation
Wild (squirting) Ecballium Constipation, antiinflammatory, Airway obstruction
cucumber elaterium rheumatic disease
Wormwood Artemisia spp. Stimulant, hallucinogen Hallucinations, seizures, uterine
(mugwort) stimulation
Yohimbine Corynanthe Aphrodisiac, stimulant Hypertension, sympathetic crisis
yohimbe
CNS, Central nervous system; GI, gastrointestinal.
From Kingston RL, Foley C: Herbal, traditional, and alternative medicines. In Haddad and
Winchester's clinical management of poisoning and drug overdose, ed 4, Philadelphia, 2007,
Saunders/Elsevier, p 1081.
Table 78.3
Common Herbal Dietary Supplement (HDS)–Drug
Interactions
Table 78.4
Acupuncture
Modern acupuncture incorporates treatment traditions from China, Japan, Korea,
France, and other countries. In the United States, acupuncturists are licensed to
practice in 45 states. Acupuncture can be delivered to pediatric patients in
hospital and clinic settings to treat a variety of ailments. Acupuncture is
particularly useful for children experiencing pain, and acupuncture services are
offered alongside conventional medicine and psychology by >50% of North
American academic pediatric chronic pain programs. The technique that has
undergone most scientific study involves penetrating the skin with thin, solid,
metallic needles manipulated by hand or by electrical stimulation. Variants
known to all office staff. Outdated medication, a laryngoscope with a failed light
source, or an empty oxygen tank represents a potential catastrophe in a
resuscitation scenario. Such an incident can be easily avoided if an equipment
checklist and regular maintenance schedule are implemented. A pediatric kit that
includes posters, laminated cards, or a color-coded length-based resuscitation
tape specifying emergency drug doses and equipment size are invaluable in
avoiding critical therapeutic errors during resuscitation.
Table 79.1
Recommended Drugs and Equipment for Pediatric Office
Emergencies
DRUGS/EQUIPMENT PRIORITY
DRUGS
Oxygen E
Albuterol for inhalation E
Epinephrine (1 : 1,000 [1 mg/mL]) E
Activated charcoal S
Antibiotics S
Anticonvulsants (diazepam/lorazepam) S
Corticosteroids (parenteral/oral) S
Dextrose (25%) S
Diphenhydramine (parenteral, 50 mg/mL) S
Epinephrine (1 : 10,000 [0.1 mg/mL]) S
Atropine sulfate (0.1 mg/mL) S
Naloxone (0.4 mg/mL) S
Sodium bicarbonate (4.2%) S
INTRAVENOUS FLUIDS
Normal saline (0.9 NS) or lactated Ringer solution (500 mL bags) S
5% dextrose, 0.45 NS (500 mL bags) S
EQUIPMENT FOR AIRWAY MANAGEMENT
Oxygen and delivery system E
Bag-valve-mask (450 mL and 1,000 mL) E
Clear oxygen masks, breather and non-rebreather, with reservoirs (infant, child, adult) E
Suction device, tonsil tip, bulb syringe E
Nebulizer (or metered-dose inhaler with spacer/mask) E
Oropharyngeal airways (sizes 00-5) E
Pulse oximeter E
Nasopharyngeal airways (sizes 12-30F) S
Magill forceps (pediatric, adult) S
Suction catheters (sizes 5-16F and Yankauer suction tip) S
Nasogastric tubes (sizes 6-14F) S
Laryngoscope handle (pediatric, adult) with extra batteries, bulbs S
Laryngoscope blades (straight 0-2; curved 2-3) S
Endotracheal tubes (uncuffed 2.5-5.5; cuffed 6.0-8.0) S
Stylets (pediatric, adult) S
Esophageal intubation detector or end-tidal carbon dioxide detector S
EQUIPMENT FOR VASCULAR ACCESS AND FLUID MANAGEMENT
Butterfly needles (19-25 gauge) S
Catheter-over-needle device (14-24 gauge) S
Arm boards, tape, tourniquet S
Intraosseous needles (16 and 18 gauge) S
Intravenous tubing, micro-drip S
MISCELLANEOUS EQUIPMENT AND SUPPLIES
Color-coded tape or preprinted drug doses E
Cardiac arrest board/backboard E
Sphygmomanometer (infant, child, adult, thigh cuffs) E
Splints, sterile dressings E
Automated external defibrillator with pediatric capabilities S
Spot glucose test S
Stiff neck collars (small/large) S
Heating source (overhead warmer/infrared lamp) S
E, Essential; S, strongly suggested.
From Frush K, American Academy of Pediatrics, Committee on Pediatric Emergency Medicine:
Policy statement-preparation for emergencies in the offices of pediatricians and pediatric primary
care providers, Pediatrics 120:200–212, 2007. Reaffirmed Pediatrics 128:e748, 2011.
Transport
Once the child has been stabilized, a decision must be made on how best to
transport a child to a facility capable of providing definitive care. If a child has
required airway or cardiovascular support, has altered mental state or unstable
vital signs, or has significant potential to deteriorate en route, it is not
appropriate to send the child via privately owned vehicle, regardless of
proximity to a hospital. Even when an ambulance is called, it is the PCP's
responsibility to initiate essential life support measures and to attempt to
stabilize the child before transport.
In metropolitan centers with numerous public and private ambulance agencies,
the PCP must be knowledgeable about the level of service provided by each. The
availability of BLS vs ALS services, the configuration of the transport team, and
pediatric expertise vary greatly among agencies and across jurisdictions. BLS
services provide basic support of airway, breathing, and circulation, whereas
https://emscimprovement.center/ .
Baseline readiness standards must be met by all EDs that care for children, to
ensure that children receive the best emergency care possible. Specific
recommendations on equipment, supplies, and medications for the ED are listed
and updates available on the AAP website. Table 79.2 lists sample policies,
procedures, and protocols specifically addressing the needs of children in the
ED.
Table 79.2
Guidelines for Pediatric-Specific Policies,
Procedures, and Protocols for the Emergency
Department (ED)
Illness and injury triage
Pediatric patient assessment and reassessment
Documentation of pediatric vital signs, abnormal vital signs, and actions to
be taken for abnormal vital signs
Immunization assessment and management of the underimmunized patient
Sedation and analgesia for procedures, including medical imaging
Consent (including situations in which a parent is not immediately
available)
Social and mental health issues
Physical or chemical restraint of patients
Child maltreatment (physical and sexual abuse, sexual assault, and neglect)
mandated reporting criteria, requirements, and processes
Death of the child in the ED
Do-not-resuscitate orders
Family-centered care, including:
1. Involving families in patient care decision-making and in
medication safety processes.
2. Family presence during all aspects of emergency care,
including resuscitation.
3. Education of the patient, family, and regular caregivers.
4. Discharge planning and instruction.
5. Bereavement counseling.
Communication with patient's medical home or primary healthcare
provider
Medical imaging policies that address age- or weight-appropriate dosing
for children receiving studies that impart ionizing radiation, consistent
with ALARA (as low as reasonably achievable) principles
All-hazard disaster preparedness plan that addresses the following pediatric
issues:
a. Availability of medications, vaccines, equipment, and
appropriately trained providers for children in disasters.
b. Pediatric surge capacity for both injured and noninjured
children.
c. Decontamination, isolation, and quarantine of families and
children of all ages.
d. A plan that minimizes parent-child separation and includes
system tracking of pediatric patients, allowing for the timely
reunification of separated children with their families.
e. Access to specific medical and mental health therapies, as well
as social services, for children in the event of a disaster.
f. Disaster drills, which should include a pediatric mass casualty
incident at least every 2 yr.
g. Care of children with special healthcare needs.
h. A plan that includes evacuation of pediatric units and pediatric
specialty units.
The way the family supports the child during a crisis, and consequently how
the family is supported in the ED when caring for the child, are critical to patient
recovery, family satisfaction, and mitigation of behavioral and mental health
impact. Commitment to patient- and family-centered care in the ED ensures that
the patient and family experience guides the practice of culturally sensitive care
and promotes patient dignity, comfort, and autonomy. In the ED setting,
particular issues, such as family presence, deserve specific attention. Surveys of
reflect physician behavior or [lack of] experience). Therefore, some other
assessment is needed that incorporates information on how sick the patient is, or
their specific diagnosis.
Table 79.3 provides a list of outcome measures for pediatric ED care
developed by Emergency Medical Services for Children Innovation and
Improvement Cente r supported by the Health Resources and Services
Administration of the U.S. Department of Health and Human Services.
Table 79.3
Stakeholder-Endorsed Outcome Measures for
Pediatric Emergency Care
• Overall patient satisfaction with ED visit—nurses
• Overall patient satisfaction with ED visit—physicians
• Parent/caregiver understanding of ED discharge instructions
• ED length of stay for patients <18 yr of age
• Percentage of patients <18 yr of age left without being seen (LWBS)
• Effective pediatric procedural sedation
• Acute fracture patients with documented reduction in pain within 90 min of
ED arrival
• Improvement in asthma severity score for patients with acute exacerbations
• ED revisit within 48 hr resulting in admission
• Medication error rates
• Global sentinel never events
• Unplanned return visit within 72 hr for the same/related asthma
exacerbation
• Failure to achieve seizure control within 30 min of ED arrival
• Return visits within 48 hr resulting in admission for all urgent and
emergency patients
Risk Adjustment
The purpose of measuring outcomes in the ED is to evaluate performance and
therefore to offer EDs and other components of the healthcare system the
ward care. A number of disease-specific acuity scoring systems are available for
use in the ED population, predominantly for those involved in trauma (e.g.,
Injury Severity Score, Trauma Score, Pediatric Trauma Score).
Bibliography
Alessandrini EA, Alpern ER, Chamberlain JM, et al.
Developing a diagnosis-based severity classification system
for use in emergency medical services for children. Acad
Emerg Med . 2012;19:70–78.
Bradman K, Borland M, Pascoe E. Predicting patient
disposition in a paediatric emergency department. J Paediatr
Child Health . 2014;50:E39–E44.
Chamberlain JM, Joseph JG, Pollack MM. Differences in
severity-adjusted pediatric hospitalization rates are associated
with race/ethnicity. Pediatrics . 2007;119:e1319–e1324.
Chamberlain JM, Patel KM, Pollack MM. The association of
emergency department care factors with admission and
discharge decisions for pediatric patients. J Pediatr .
2006;149:644–649.
Chamberlain JM, Patel KM, Pollack MM. The pediatric risk of
hospital admission score: a second-generation severity-of-
illness score for pediatric emergency patients. Pediatrics .
2005;115:388–395.
Defining quality performance measures for pediatric emergency
care. www.childrensnational.org/EMSC/PubRes/toolbox.aspx
.
Donabedian A. The quality of care: how can it be assessed?
the WHO manuals Prehospital Trauma Care Systems and Guidelines for
Essential Trauma Care both focus on guidelines for prehospital and trauma care
systems that are affordable and sustainable. The AAP course Pediatric Education
for Prehospital Professionals is a dynamic, modularized teaching tool designed
to provide specific pediatric prehospital education that can be adapted to any
EMS system. Table 79.6 describes additional prehospital resources.
Table 79.6
Pediatric Emergency Care Resources
Prehospital
Hospital Care
Humanitarian Emergencies
CHILDisaster Network
Registry for those with education and experience in humanitarian
emergencies to volunteer their time when needed in a disaster.
www.aap.org/disaster
The Sphere Project
Downloadable modules on disaster preparedness.
www.sphereproject.org
Management of Complex Humanitarian Emergencies: Focus on
Children and Families
Training course offered by the Children in Disasters Project, sponsored by
the Rainbow Center for Global Child Health (RCGCH) in Cleveland,
OH. Held in early June annually.
Manual for the Health Care of Children in Humanitarian Emergencies
WHO publication that provides comprehensive guidance on childcare in
emergencies; includes information on care of traumatic injuries and
mental health emergencies.
www.who.int/child_adolescent_health/documents/9789241596879/en/index.html
PEMdatabase.org
A website devoted to pediatric emergency medicine (PEM). Contains links
to conferences, evidence-based medicine reviews, research networks, and
professional organizations.
www.pemdatabase.org
HINARI Access to Research Initiative
Program established by WHO and others to enable developing countries to
gain access to one of the world's largest collections of biomedical and
health literature.
www.who.int/hinari/en
Involvement
Methods of Transport
ended questions that help distinguish between benign and potentially life-
threatening disease entities. The most common complaints leading to acute care
visits among children include fever, headache and altered mental status, trauma,
abdominal pain and vomiting, respiratory distress, and chest pain. Table 80.1
describes signs and symptoms that should prompt immediate transfer to an ED
or, if already in the ED, initiation of rapid intervention.
Table 80.1
History and Examination Findings That Should Prompt
Immediate Intervention and/or Transfer to Emergency
Department
Table 81.1
Normal Vital Signs According to Age
HEART RATE BLOOD PRESSURE (mm RESPIRATORY RATE
AGE
(beats/min) Hg) (breaths/min)
Premature 120-170* 55-75/35-45 † 40-70 ‡
0-3 mo 100-150* 65-85/45-55 35-55
3-6 mo 90-120 70-90/50-65 30-45
6-12 mo 80-120 80-100/55-65 25-40
1-3 yr 70-110 90-105/55-70 20-30
Table 81.3
Glasgow Coma Scale
EYE OPENING (TOTAL POSSIBLE POINTS 4)
Spontaneous 4
To voice 3
To pain 2
None 1
VERBAL RESPONSE (TOTAL POSSIBLE POINTS 5)
Older Children Infants and Young Children
Oriented 5 Appropriate words; smiles, fixes, and follows 5
Confused 4 Consolable crying 4
Inappropriate 3 Persistently irritable 3
Incomprehensible 2 Restless, agitated 2
None 1 None 1
MOTOR RESPONSE (TOTAL POSSIBLE POINTS 6)
Obeys 6
Localizes pain 5
Withdraws 4
Flexion 3
Extension 2
None 1
Adapted from Teasdale G, Jennett B: Assessment of coma and impaired consciousness: a
practical scale, Lancet 2:81–84, 1974.
Exposure
Exposure is the final component of the pediatric primary assessment. This
component of the exam is reached only after the child's airway, breathing, and
circulation have been assessed and determined to be stable or have been
stabilized through simple interventions. In this setting, exposure stands for the
dual responsibility of the provider to both expose the child to assess for
previously unidentified injures and consider prolonged exposure in a cold
environment as a possible cause of hypothermia and cardiopulmonary instability.
The provider should undress the child (as is feasible and reasonable) to perform
a focused physical exam, assessing for burns, bruising, bleeding, joint laxity, and
fractures. If possible, the provider should assess the child's temperature. All
maneuvers should be performed with careful maintenance of cervical spine
precautions.
Secondary Assessment
For healthcare providers in community or outpatient settings, transfer of care of
a child to emergency or hospital personnel may occur before a full secondary
assessment is possible. However, before the child is removed from the scene and
separated from witnesses or family, a brief history should be obtained for
medical providers at the accepting facility. The components of a secondary
assessment include a focused history and focused physical examination.
The history should be targeted to information that could explain
cardiorespiratory or neurologic dysfunction and should take the form of a
SAMPLE history : signs/symptoms, allergies, medications, past medical
history, timing of last meal, and events leading to this situation. Medical
personnel not engaged in resuscitative efforts can be dispatched to elicit history
from witnesses or relatives. The physical examination during the secondary
assessment is a thorough head-to-toe exam, although the severity of the child's
FIG. 81.12 A-E, Intubation technique. (From Fleisher G, Ludwig S:
Textbook of pediatric emergency medicine, Baltimore, 1983, Williams &
Wilkins, p. 1250.)
Table 81.4
Rapid Sequence Intubation
Tachyarrhythmias
Tachyarrhythmias represent a wide variety of rhythm disturbances of atrial and
ventricular origin (see Chapter 462 ). Sinus tachycardia is a normal physiologic
response to the body's need for increased cardiac output or oxygen delivery, as
occurs with fever, exercise, or stress. It can also occur in more pathologic states,
such as hypovolemia, anemia, pain, anxiety, and metabolic stress.
Tachyarrhythmias that do not originate in the sinus node are often categorized as
narrow complex rhythms (i.e., originating in the atrium, such as atrial flutter or
supraventricular tachycardia, SVT) and wide complex rhythms (i.e., rhythms of
ventricular origin, such as ventricular tachycardia).
The initial management of tachycardia includes confirmation that the child has
an adequate airway and life-sustaining breathing and circulation (Fig. 81.14 ).
For children with persistent symptoms, further treatment is based on whether the
QRS complex of the electrocardiogram (ECG) is narrow (≤0.09 sec) or wide
(>0.09 sec). For narrow complex tachycardia, providers must distinguish
between sinus tachycardia and SVT. In sinus tachycardia , (a) the history and
onset are consistent with a known cause of tachycardia, such as fever or
dehydration, and (b) P waves are consistently present, are of normal morphology,
FIG. 81.18 Pediatric advanced life support pulseless arrest algorithm. CPR,
Cardiopulmonary resuscitation; IO/IV, intraosseous/intravenous; PEA, pulseless
electrical activity; VF/VT, ventricular fibrillation/tachycardia. (From Kleinman ME,
Chameides L, Schexnayder SM, et al: 2010 American Heart Association guidelines for
cardiopulmonary resuscitation and emergency cardiovascular care. Part 14, Circulation
122[Suppl 3]:S876–S908, 2010, Fig 1, p S885.)
Table 81.6
Medications for Pediatric Resuscitation and Arrhythmias
Table 81.7
Table 82.1
Pediatric Trauma Score*
COMPONENT +2 +1 −1
Size ≥20 kg 10-20 kg <10 kg
Airway Normal Maintainable Unmaintainable
Systolic BP ≥90 mm Hg 50-90 mm Hg <50 mm Hg
CNS Awake Obtunded/LOC Coma/decerebrate
Open wound None Minor Major/penetrating
Skeletal None Closed fracture Open/multiple fractures
Sum total points
* Children with a Pediatric Trauma Score ≤6 are at increased risk of mortality as well as morbidity.
BP, Blood pressure; CNS, central nervous system; LOC, loss of consciousness.
From Tepas JJ 3rd, Mollitt DL, Talbert JL, et al: The Pediatric Trauma Score as a predictor of
injury severity in the injured child, J Pediatr Surg 22:14–18, 1987 (Table 1, p 15).
Primary Survey
During the primary survey, the physician quickly assesses and treats any life-
threatening injuries. The principal causes of death shortly after trauma are
airway obstruction, respiratory insufficiency, shock from hemorrhage, and
central nervous system (CNS) injury. The primary survey addresses the
ABCDEs : Airway, Breathing, Circulation, neurologic Deficit, and Exposure of
the patient and control of the Environment.
Airway/Cervical Spine
Optimizing oxygenation and ventilation, while protecting the cervical spine (C-
spine) from potential further injury, is of paramount importance. Initially, C-
spine injury should be suspected in any child sustaining multiple, blunt trauma.
Although C-spine injuries occur less often in children than adults, children are at
monitoring should also be used as an adjunct; however, it is less reliable in
patients with shock. In addition to looking visually for cyanosis, pulse oximetry
is standard. If ventilation is inadequate, bag-valve-mask ventilation with 100%
oxygen must be initiated immediately, followed by endotracheal intubation. End-
expiratory carbon dioxide (CO2 ) detectors or capnography help verify accurate
tube placement.
Head trauma is the most common cause of respiratory insufficiency. An
unconscious child with severe head injury may have a variety of breathing
abnormalities, including Cheyne-Stokes respiration, slow irregular breaths, and
apnea.
Although less common than a pulmonary contusion, tension pneumothorax
and massive hemothorax are immediately life threatening (Tables 82.2 and 82.3
). Tension pneumothorax occurs when air accumulates under pressure in the
pleural space. The adjacent lung is compacted, the mediastinum is pushed
toward the opposite hemithorax, and the heart, great vessels, and contralateral
lung are compressed or kinked (see Chapter 439 ). Both ventilation and cardiac
output are impaired. Characteristic findings include cyanosis, tachypnea,
retractions, asymmetric chest rise, contralateral tracheal deviation, diminished
breath sounds on the ipsilateral (more than contralateral) side, and signs of
shock. Needle thoracentesis, followed by thoracostomy tube insertion, is
diagnostic and lifesaving. Hemothorax results from injury to the intercostal
vessels, lungs, heart, or great vessels. When ventilation is adequate, fluid
resuscitation should begin before evacuation, because a large amount of blood
may drain through the chest tube, resulting in shock.
Table 82.2
Differential Diagnosis of Immediately Life-Threatening
Cardiopulmonary Injuries
MASSIVE CARDIAC
TENSION PNEUMOTHORAX
HEMOTHORAX TAMPONADE
Breath sounds Ipsilaterally decreased more than Ipsilaterally decreased Normal
contralaterally
Percussion Hyperresonant Dull Normal
note
Tracheal Contralaterally shifted Midline or shifted Midline
location
Neck veins Distended Flat Distended
Heart tones Normal Normal Muffled
Modified from Cooper A, Foltin GL: Thoracic trauma. In Barkin RM, editor: Pediatric emergency
medicine , ed 2, St Louis, 1997, Mosby, p 325.
Table 82.3
Life-Threatening Chest Injuries
Tension Pneumothorax
Massive Hemothorax
Cardiac Tamponade
Beck's triad:
1. Decreased or muffled heart sounds
2. Jugular venous distention
3. Hypotension (with narrow pulse pressure)
Must be drained
Modified from Krug SE: The acutely ill or injured child. In Behrman RE,
Kliegman RM, editors: Nelson essentials of pediatrics , ed 4, Philadelphia, 2002,
Saunders, p 97.
Circulation
Signs of shock include tachycardia; weak pulse; delayed capillary refill; cool,
mottled, pale skin; and altered mental state (see Chapter 88 ). The most common
type of shock in trauma is hypovolemic shock caused by hemorrhage. Cardiac
tamponade, which is a form of obstructive shock, may be suspected clinically or
diagnosed by focused assessment with sonography in trauma (FAST)
examination or echocardiography. Cardiac tamponade is best managed by
thoracotomy or pericardial window, although pericardiocentesis may be
necessary as a temporizing maneuver (see Table 82.3 ).
Early in shock, blood pressure remains normal because of compensatory
increases in heart rate and peripheral vascular resistance (Table 82.4 ). Some
individuals can lose up to 30% of blood volume before blood pressure declines.
It is important to note that 25% of blood volume equals 20 mL/kg, which is only
200 mL in a 10 kg child. Losses >40% of blood volume cause severe
hypotension that, if prolonged, may become irreversible. Direct pressure should
be applied to control external hemorrhage. When direct pressure does not control
hemorrhage, a tourniquet should be applied to a proximal pressure point. Blind
clamping of bleeding vessels, which risks damaging adjacent structures, is not
advisable.
Table 82.4
Systemic Responses to Blood Loss in Pediatric Patients
Neurologic Deficit
Neurologic status is briefly assessed by determining the level of consciousness
and evaluating pupil size and reactivity. The level of consciousness can be
the primary diagnostic tool, particularly in patients with abnormal GCS scores
and/or significant injury mechanisms, recognizing that CT is more sensitive in
detecting bony injury than plain radiographs. CT is also helpful if an odontoid
fracture is suspected, because young children typically do not cooperate enough
to obtain an open-mouth (odontoid) radiographic view. Use of cervical spine CT
scan must be balanced with the knowledge that CT exposes thyroid tissue to 90-
200 times the amount of radiation from plain films. MRI is indicated in a child
with suspected SCIWORA and in the evaluation of children who remain
obtunded.
Table 82.5
National Emergency X-Radiography
Utilization Study (NEXUS) to Rule Out
Cervical Spine Injury Following Blunt
Trauma
If none of the following is present, the patient is at very low risk for
clinically significant cervical spine injury:
Midline cervical tenderness
Evidence of intoxication
Altered level of alertness
Focal neurologic deficit
Distracting painful injury
Data from Hoffman JR, Mower WR, Wolfson AB, et al: Validity of a set of
clinical criteria to rule out injury to the cervical spine in patients with blunt
trauma, N Engl J Med 343:94–99, 2000; and Viccellio P, Simon H, Pressman
BD, et al: A prospective multicenter study of cervical spine injury in children,
Pediatrics 108:e20, 2001.
Thoracic Trauma
Pulmonary contusions occur frequently in young children with blunt chest
trauma. A child's chest wall is relatively pliable; therefore, less force is absorbed
by the rib cage, and more is transmitted to the lungs. Respiratory distress may be
noted initially or may develop during the 1st 24 hr after injury.
Rib fractures result from significant external force. They are noted in patients
with more severe injuries and are associated with a higher mortality rate. Flail
chest, which is caused by multiple rib fractures, is rare in children. Table 82.6
lists indications for operative management in thoracic trauma. (See Table 82.2
for the differential diagnosis of immediately life-threatening cardiopulmonary
injuries.)
Table 82.6
Indications for Operation in Thoracic Trauma
Thoracotomy Immediately or Shortly After Injury
Delayed Thoracotomy
Children <2 yr old are at very low risk of clinically important traumatic
brain injury if they have none of the following:
Severe mechanism of injury
History of LOC >5 sec
GCS ≤14 or other signs of altered mental status
Not acting normally per parent
Palpable skull fracture
Occipital/parietal/temporal scalp hematoma
Children 2-18 yr old are at very low risk of clinically important traumatic
brain injury if they have none of the following:
Severe mechanism of injury
History of LOC
History of vomiting
GCS ≤14 or other signs of altered mental status
Severe headache in the ED
Signs of basilar skull fracture
ED, Emergency department; GCS, Glasgow Coma Scale score; LOC, loss of
consciousness.
If none of the following is present, the patient is at very low risk for
clinically significant intraabdominal injury:
Glasgow Coma Scale score <14
Vomiting
Evidence of thoracic wall trauma
Decreased breath sounds
Evidence of abdominal wall trauma or seatbelt sign
Abdominal pain
Abdominal tenderness
Modified from Holmes JF, Lillis K, Monroe D, et al: Identifying children at very
low risk of clinically important blunt abdominal injuries, Ann Emerg Med
Commonly Used Coma Scores
POINTS DESCRIPTION
GLASGOW COMA SCALE (GCS) SCORE
Eye Opening
1 Does not open eyes
2 Opens eyes in response to noxious stimuli
3 Opens eyes in response to voice
4 Opens eyes spontaneously
Verbal Output
1 Makes no sounds
2 Makes incomprehensible sounds
3 Utters inappropriate words
4 Confused and disoriented
5 Speaks normally and oriented
Motor Response (Best)
1 Makes no movements
2 Extension to painful stimuli
3 Abnormal flexion to painful stimuli
4 Flexion/withdrawal to painful stimuli
5 Localized to painful stimuli
6 Obeys commands
FULL OUTLINE OF UNRESPONSIVENESS (FOUR) SCORE
Eye Response
4 Eyelids open or opened, tracking, or blinking to command
3 Eyelids open but not tracking
2 Eyelids closed but open to loud voice
1 Eyelids closed but open to pain
0 Eyelids remain closed with pain
Motor Response
4 Thumbs-up, fist, or “peace” sign
3 Localizing to pain
2 Flexion response to pain
1 Extension response to pain
0 No response to pain or generalized myoclonus status
Brainstem Reflexes
4 Pupil and corneal reflexes present
3 One pupil wide and fixed
2 Pupil or corneal reflexes absent
1 Pupil and corneal reflexes absent
0 Absent pupil, corneal, and cough reflex
Respiration
4 Not intubated, regular breathing pattern
3 Not intubated, Cheyne-Stokes breathing pattern
2 Not intubated, irregular breathing
1 Breathes above ventilatory rate
0 Breathes at ventilator rate or apnea
Adapted from Edlow JA, Rabinstein A, Traub SJ, Wijdicks EFM: Diagnosis of reversible causes of
coma, Lancet 384:2064-2076, 2014.
The most studied monitoring device in clinical practice is the ICP monitor .
such as electroencephalography (EEG) and cerebral blood flow (CBF) studies
are sometimes used to assist in making the diagnosis, repeated clinical
examination is the standard for diagnosis. The 3 components for determining
brain death are demonstration of coexisting irreversible coma with a known
cause , absence of brainstem reflexes , and apnea .
Before a determination of brain death may be made, it is of utmost importance
that the cause of the coma be determined using the history, any radiology, and
laboratory data, to rule out a reversible condition. Potentially reversible causes
of coma include metabolic disorders, toxins, sedative drugs, paralytic agents,
hypothermia, hypoxia, hypotension/shock, recent cardiopulmonary resuscitation
(CPR), hypo-/hyperglycemia, hypo-/hypernatremia, hypercalcemia,
hypermagnesemia, nonconvulsive status epilepticus, hypothyroidism,
hypocortisolism, hypercarbia, liver or renal failure, sepsis, meningitis,
encephalitis, subarachnoid hemorrhage, and surgically remediable brainstem
lesions. Confounding factors must be corrected before initiation of brain death
assessment.
Coma
The state of coma requires that the patient be unresponsive, even to noxious
stimuli. Any purposeful motor response, such as localization, does not constitute
coma. Likewise, any posturing (decerebrate or decorticate) is not consistent with
coma, and therefore not consistent with brain death. The presence of spinal cord
reflexes—even complex reflexes—does not preclude the diagnosis of brain
death.
Brainstem Reflexes
Brainstem reflexes must be absent. Table 86.1 lists the brainstem reflexes to be
tested, the brainstem location of each reflex, and the result of each test that is
consistent with a diagnosis of brain death.
Table 86.1
Brainstem Reflex Testing to Determine Brain Death
Apnea
Apnea is the absence of respiratory effort in response to an adequate stimulus.
An arterial partial pressure of carbon dioxide (PaCO 2 ) value ≥60 mm Hg and
>20 mm Hg above baseline is a sufficient stimulus. Apnea is clinically
confirmed through the apnea test. Because the apnea test has the potential to
destabilize the patient, it is performed only if the 1st 2 criteria for brain death
(irreversible coma and absence of brainstem reflexes) are already confirmed.
The apnea test assesses the function of the medulla in driving ventilation. It is
performed by first ensuring appropriate hemodynamics and temperature (>35°C)
and the absence of apnea-producing drug effects or significant metabolic
derangements. The patient is then preoxygenated with 100% oxygen for
approximately 10 min, and ventilation is adjusted to achieve a PaCO 2 of
approximately 40 mm Hg. A baseline arterial blood gas (ABG) result documents
CHAPTER 87
Syncope
Aarti S. Dalal, George F. Van Hare
Most patients with a vasovagal syncope episode will have prodromal features
followed by loss of motor tone. Once in a horizontal position, consciousness
returns rapidly, in 1-2 min; some patients may have 30 sec of tonic-clonic motor
activities, which should not be confused with a seizure (Table 87.2 ). Syncope
must also be distinguished from vertigo and ataxia (Table 87.3 ).
Table 87.2
Table 87.3
Syncope and Dizziness
Mechanisms
Syncope by whatever mechanism is caused by a lack of adequate cerebral blood
flow with loss of consciousness and inability to remain upright.
Primary cardiac causes of syncope (Table 87.4 ) include arrhythmias such as
long QT syndrome (LQTS), Wolff-Parkinson-White syndrome (particularly with
atrial fibrillation), ventricular tachycardia (VT), and occasionally
supraventricular tachycardia (see Chapter 462 ). VT may be associated with
hypertrophic cardiomyopathy (HCM), arrhythmogenic cardiomyopathy, repaired
congenital heart disease, or a genetic cause such as catecholaminergic
polymorphous ventricular tachycardia (CPVT). Other arrhythmias that may lead
to syncope are bradyarrhythmias such as sinus node dysfunction and high-grade
second- or third-degree atrioventricular (AV) block. Patients with congenital
complete AV block may present with syncope. Syncope may also be caused by
cardiac obstructive lesions, such as critical aortic stenosis, or coronary artery
anomalies, such as an aberrant left coronary artery arising from the right sinus of
Valsalva. Patients with primary pulmonary hypertension or Eisenmenger
syndrome may experience syncope. In all the obstructive forms of syncope,
exercise increases the likelihood of an episode because the obstruction interferes
with the ability of the heart to increased cardiac output in response to exercise.
Noncardiac causes of loss of consciousness include epilepsy, as well as
basilar artery migraine, hysterical syncope, and pseudoseizures (see Table 87.1 ).
Occasionally, patients with narcolepsy may present with syncope. Hypoglycemia
and hyperventilation may also present as syncope.
Evaluation
The most important goal in the evaluation of the new patient with syncope is to
diagnose life-threatening causes of syncope so that these causes can be managed.
Many patients presenting with sudden cardiac arrest caused by conditions such
as LQTS will have previously experienced an episode of syncope, so the
presentation with syncope is an opportunity to prevent sudden death.
The most important tool in evaluation is a careful history . The characteristics
of cardiac syncope differ significantly from the prodrome seen in
neurocardiogenic syncope (Table 87.5 ). Several red flags can be identified that
should lead the clinician to suspect that the mechanism is a life-threatening
cardiac cause rather than simple fainting (Table 87.6 ). The occurrence during
exercise suggests an arrhythmia or coronary obstruction. Injury because of an
episode of syncope indicates sudden occurrence with a lack of adequate
prodromal symptoms and suggests an arrhythmia. The occurrence of syncope
while recumbent would be quite unusual in a patient with neurocardiogenic
syncope and therefore suggests a cardiac or neurologic cause. Occasionally, a
patient with syncope caused by a tachyarrhythmia will report the sensation of a
racing heart before the event, but this is unusual.
Table 87.5
Differentiating Features for Causes of Syncope
NEUROCARDIOGENIC
Symptoms after prolonged motionless standing, sudden unexpected pain, fear, or unpleasant sight, sound, or
smell; pallor
Syncope in a well-trained athlete after exertion (without heart disease)
Situational syncope during or immediately after micturition, cough, swallowing, or defecation
Syncope with throat or facial pain (glossopharyngeal or trigeminal neuralgia)
ORGANIC HEART DISEASE (PRIMARY ARRHYTHMIA, OBSTRUCTIVE HYPERTROPHIC
CARDIOMYOPATHY, PULMONARY HYPERTENSION)
Brief sudden loss of consciousness, no prodrome, history of heart disease
Syncope while sitting or supine
Syncope with exertion
History of palpitations
Family history of sudden death
NEUROLOGIC
Seizures: preceding aura, post event symptoms lasting > 5 min (includes postictal state of decreased level of
consciousness, confusion, headache or paralysis)
Migraine: syncope associated with antecedent headaches with or without aura
OTHER VASCULAR
Carotid sinus: syncope with head rotation or pressure on the carotid sinus (as in tumors, shaving, tight collars)
Orthostatic hypotension: syncope immediately on standing especially after prolonged bed rest
DRUG INDUCED
Patient is taking a medication that may lead to long QT syndrome, orthostasis, or bradycardia
PSYCHIATRIC ILLNESS
Frequent syncope, somatic complaints, no heart disease
From Cohen G: Syncope and dizziness. In Kliegman RM, Lye PS, Bordini BJ, et al, editors:
Nelson pediatric symptom-based diagnosis. Philadelphia, 2018, Elsevier, Table 6.4.
Table 87.6
Red Flags in Evaluation of Patients With
Syncope
Syncope with activity or exercise or supine
Syncope not associated with prolonged standing
Syncope precipitated by loud noise or extreme emotion
Absence of presyncope or lightheadedness
Family history of syncope, drowning, sudden death, familial ventricular
arrhythmia syndromes,* cardiomyopathy
Syncope requiring CPR
Injury with syncope
Anemia
Other cardiac symptoms
Chest pain
Dyspnea
Palpitations
History of cardiac surgery
History of Kawasaki disease
Implanted pacemaker
Abnormal physical examination
Murmur
Gallop rhythm
Loud and single second heart sound
Systolic click
Increased apical impulse (tachycardia)
Irregular rhythm
Hypo- or hypertension
Clubbing
Cyanosis
Treatment
Therapy for vasovagal syncope includes avoiding triggering events (if possible),
fluid and salt supplementation, and if needed, midodrine (see Chapter 87.1 ,
Table 87.7 ). Immediately after the event, the patient should remain supine until
symptoms abate to avoid recurrence.
Table 87.7
First-Line Medications in Treatment of Postural
Tachycardia Syndrome (POTS)
TREATMENT
DRUG MECHANISM OF ACTION SIDE EFFECTS
GUIDELINES
Fludrocortisone Low dose: sensitizes α receptors Peripheral edema, headache, Monitor basic
Higher doses: mineralocorticoid effect irritability, hypokalemia, metabolic panel and
hypomagnesemia, acne magnesium.
Midodrine α1 -Agonist; produces vasoconstriction Scalp tingling, urinary Monitor supine
retention, goose bumps, blood pressure 30-
headache, supine 60 min after dose.
hypertension
Metoprolol β-Blocker Worsening of asthma, Use with
succinate/tartrate dizziness, fatigue caution in
asthma.
If fatigue is
severe, use at
bedtime.
Propranolol Nonselective β-blocker Bradycardia, gastrointestinal Use with caution in
symptoms, lightheadedness, diabetes and
sleepiness, hypotension, asthma.
syncope
Pyridostigmine Peripheral acetylcholinesterase inhibitor Symptoms of excessive Very useful if
that increases synaptic acetylcholine in cholinergic activity patient has
autonomic ganglia and at peripheral (diarrhea, urinary POTS and
muscarinic receptors incontinence, salivation) constipation.
Use with
caution in
asthma.
Contraindicated
in urinary or
bowel
obstruction.
Table 88.1
Types of Shock
Pathophysiology
An initial insult triggers shock, leading to inadequate oxygen delivery to organs
and tissues. Compensatory mechanisms attempt to maintain blood pressure (BP)
by increasing cardiac output and systemic vascular resistance (SVR). The body
also attempts to optimize oxygen delivery to the tissues by increasing oxygen
extraction and redistributing blood flow to the brain, heart, and kidneys at the
expense of the skin and gastrointestinal (GI) tract. These responses lead to an
initial state of compensated shock in which BP is maintained. If treatment is not
initiated or is inadequate during this period, decompensated shock develops,
with hypotension and tissue damage that may lead to multisystem organ
dysfunction and, ultimately, death (Tables 88.2 and 88.3 ).
Table 88.2
Criteria for Organ Dysfunction
ORGAN
CRITERIA FOR DYSFUNCTION
SYSTEM
Cardiovascular Despite administration of isotonic intravenous fluid bolus ≥60 mL/kg in 1 hr: decrease in BP
(hypotension) systolic BP <90 mm Hg, mean arterial pressure <70 mm Hg, <5th percentile for
age, or systolic BP <2 SD below normal for age
or
Need for vasoactive drug to maintain BP in normal range (dopamine >5 µg/kg/min or
dobutamine, epinephrine, or norepinephrine at any dose)
or
Two of the following:
Unexplained metabolic acidosis: base deficit >5.0 mEq/L
Increased arterial lactate: >1 mmol/L or >2× upper limit of normal
Oliguria: urine output <0.5 mL/kg/hr
Prolonged capillary refill: >5 sec
Core-to-peripheral temperature gap: >3°C (5.4°F)
Respiratory PaO 2 /FIO 2 ratio <300 in absence of cyanotic heart disease or preexisting lung disease
or
PaCO 2 >65 torr or 20 mm Hg over baseline PaCO 2
or
Need for >50% FIO 2 to maintain saturation ≥92%
or
Need for nonelective invasive or noninvasive mechanical ventilation
Neurologic GCS score ≤11
or
Acute change in mental status with decrease in GCS score ≥3 points from abnormal baseline
Hematologic Platelet count <100,000/mm3 or decline of 50% in platelet count from highest value recorded
over last 3 days (for patients with chronic hematologic or oncologic disorders)
or
INR >1.5
or
Activated prothrombin time >60 sec
Renal Serum creatinine >0.5 mg/dL, ≥2× upper limit of normal for age, or 2-fold increase in baseline
creatinine value
Hepatic Total bilirubin ≥4 mg/dL (not applicable for newborn)
Alanine transaminase level 2× upper limit of normal for age
BP, Blood pressure; FIO 2 , fraction of inspired oxygen; GCS, Glasgow Coma Scale; INR,
international normalized ratio; PaCO 2 , arterial partial pressure of carbon dioxide; PaO 2 , partial
pressure arterial oxygen; SD, standard deviations.
Table 88.3
Signs of Decreased Perfusion
Abnormal Vasodilation
Sepsis may change the capillary permeability in the absence of any change
in capillary hydrostatic pressure (endotoxins from sepsis, excess
histamine release in anaphylaxis).
Cardiac Dysfunction
Peripheral hypoperfusion may result from any condition that affects the
heart's ability to pump blood efficiently (ischemia, acidosis, drugs,
constrictive pericarditis, pancreatitis, sepsis).
FIG. 88.3 Mechanisms of vasodilatory shock. Septic shock and states of prolonged
shock causing tissue hypoxia with lactic acidosis increase nitric oxide synthesis,
activate the adenosine triphosphate (ATP)–sensitive and calcium-regulated potassium
channels (KATP and KCa , respectively) in vascular smooth muscle, and lead to depletion
of vasopressin. cGMP, Cyclic guanosine monophosphate. (From Landry DW, Oliver JA:
The pathogenesis of vasodilatory shock, N Engl J Med 345:588.595, 2001.)
Cardiopulmonary
Gastrointestinal
Hematologic
Neurologic
Other
† Wedge pressure, central venous pressure, and pulmonary artery diastolic pressures are equal.
Sepsis
Severe Sepsis
Septic Shock
Diagnosis
Shock
SYSTEM DISORDERS GOALS THERAPIES
Respiratory Acute respiratory Prevent/treat: hypoxia and Oxygen
distress respiratory acidosis Noninvasive ventilation
syndrome
Respiratory Prevent barotrauma Early endotracheal intubation and
muscle fatigue Decrease work of breathing mechanical ventilation
Central apnea Positive end-expiratory pressure
(PEEP)
Permissive hypercapnia
High-frequency ventilation
Extracorporeal membrane
oxygenation (ECMO)
Renal Prerenal failure Prevent/treat: hypovolemia, Judicious fluid resuscitation
Renal failure hypervolemia, hyperkalemia, Establishment of normal urine output
metabolic acidosis, and blood pressure for age
hypernatremia/hyponatremia, Furosemide (Lasix)
and hypertension Dialysis, ultrafiltration, hemofiltration
Monitor serum electrolytes
Hematologic Coagulopathy Prevent/treat: bleeding Vitamin K
(disseminated Fresh-frozen plasma
intravascular Platelets
coagulation)
Thrombosis Prevent/treat: abnormal clotting Heparinization
Gastrointestinal Stress ulcers Prevent/treat: gastric bleeding Histamine H2 -receptor–blocking
Ileus Avoid aspiration, abdominal agents or proton pump inhibitors
distention Nasogastric tube
Bacterial Avoid mucosal atrophy Early enteral feedings
translocation
Endocrine Adrenal Prevent/treat: adrenal crisis Stress-dose steroids in patients
insufficiency, previously given steroids
primary or Physiologic dose for presumed
secondary to primary insufficiency in sepsis
chronic steroid
therapy
Metabolic Metabolic acidosis Correct etiology Treatment of hypovolemia (fluids),
Normalize pH poor cardiac function (fluids,
inotropic agents)
Improvement of renal acid excretion
Low-dose (0.5-2.0 mEq/kg) sodium
bicarbonate if patient is not showing
response, pH <7.1, and ventilation
(CO2 elimination) is adequate
Table 88.9
Recommendations for Shock: Initial
Resuscitation and Infection Issues—Adults
Initial Resuscitation
1. Protocolized, quantitative resuscitation of patients with sepsis-induced
tissue hypoperfusion (defined as hypotension persisting after initial fluid
challenge or blood lactate concentration ≥4 mmol/L). Goals during the
1st 6 hr of resuscitation:
a. Central venous pressure 8-12 mm Hg
b. Mean arterial pressure (MAP) ≥65 mm Hg
c. Urine output ≥0.5 mL kg−1 hr
d. Central venous (superior vena cava) or mixed venous oxygen
saturation: 70% or 65%, respectively
2. In patients with elevated lactate levels, targeting resuscitation to normalize
lactate as rapidly as possible.
Diagnosis
Antimicrobial Therapy
Source Control
Infection Prevention
Adapted from Dellinger PR, Levy MM, Rhodes A, et al: Surviving sepsis
campaign: International guidelines for management of severe sepsis and septic
shock: 2012, Crit Care Med 41(2):580–637, 2013 (Table 5, p 589).
Table 88.10
Surviving Sepsis Campaign: Care Bundles
Adapted from Dellinger PR, Levy MM, Rhodes A, et al: Surviving Sepsis
campaign: international guidelines for management of severe sepsis and septic
shock: 2012. Crit Care Med 41(2):580–637, 2013 (Fig 1, p 591).
Table 88.11
Recommendations for Shock: Hemodynamic
Support and Adjunctive Therapy—Adults
Fluid Therapy of Severe Sepsis
Vasopressors
Inotropic Therapy
Corticosteroids
Adapted from Dellinger PR, Levy MM, Rhodes A, et al: Surviving Sepsis
campaign: international guidelines for management of severe sepsis and septic
shock: 2012, Crit Care Med 41(2):580–637, 2013 (Table 6, p 596).
Table 88.12
Recommendations for Shock: Special
Considerations in Pediatric Patients
Initial Resuscitation
1. For respiratory distress and hypoxemia, start with face mask oxygen or, if
needed and available, high-flow nasal cannula oxygen or nasopharyngeal
CPAP (NP CPAP). For improved circulation, peripheral intravenous
access or intraosseous access can be used for fluid resuscitation and
inotrope infusion when a central line is not available. If mechanical
ventilation is required, cardiovascular instability during intubation is less
likely after appropriate cardiovascular resuscitation.
2. Initial therapeutic end-points of resuscitation of septic shock: capillary
refill of ≤2 sec, normal blood pressure for age, normal pulses with no
differential between peripheral and central pulses, warm extremities,
urine output >1 mL kg−1 hr−1 , and normal mental status. ScvO 2
saturation ≥70% and cardiac index between 3.3 and 6.0 L/min/m2 should
be targeted thereafter.
3. Follow American College of Critical Care Medicine–Pediatric Advanced
Life Support (ACCM-PALS) guidelines for the management of septic
shock.
4. Evaluate for and reverse pneumothorax, pericardial tamponade, or
endocrine emergencies in patients with refractory shock.
Fluid Resuscitation
Corticosteroids
Mechanical Ventilation
Glycemic Control
1. Use diuretics to reverse fluid overload when shock has resolved, and if
unsuccessful, use continuous venovenous hemofiltration (CVVH) or
intermittent dialysis to prevent >10% total body weight fluid overload.
Nutrition
1. Enteral nutrition given to children who can be fed enterally, and parenteral
feeding in those who cannot (grade 2C).
Adapted from Dellinger PR, Levy MM, Rhodes A, et al: Surviving Sepsis
campaign: I nternational guidelines for management of severe sepsis and septic
shock: 2012, Crit Care Med 41(2):580–637, 2013 (Table 9, p 614).
Table 88.13
Cardiovascular Drug Treatment of Shock
DOSING
DRUG EFFECT(S) COMMENT(S)
RANGE
Dopamine ↑ Cardiac contractility 3-20 ↑ Risk of arrhythmias at high doses
Significant peripheral vasoconstriction µg/kg/min
at >10 µg/kg/min
Epinephrine ↑ Heart rate and ↑ cardiac contractility 0.05-3.0 May ↓ renal perfusion at high doses
Potent vasoconstrictor µg/kg/min ↑ Myocardial O2 consumption
Risk of arrhythmia at high doses
Dobutamine ↑ Cardiac contractility 1-10 —
Peripheral vasodilator µg/kg/min
Norepinephrine Potent vasoconstriction 0.05-1.5 ↑ Blood pressure secondary to ↑ systemic
µg/kg/min vascular resistance
No significant effect on cardiac ↑ Left ventricular afterload
contractility
Phenylephrine Potent vasoconstriction 0.5-2.0 Can cause sudden hypertension
µg/kg/min ↑ O2 consumption
Table 88.14
Vasodilators/Afterload Reducers in Treatment of Shock
DOSING
DRUG EFFECT(S) COMMENT(S)
RANGE
Nitroprusside Vasodilator (mainly arterial) 0.5-4.0 Rapid effect
µg/kg/min Risk of cyanide toxicity with
prolonged use (>96 hr)
Nitroglycerin Vasodilator (mainly venous) 1-20 Rapid effect
µg/kg/min Risk of increased intracranial
pressure
Prostaglandin Vasodilator 0.01-0.2 Can lead to hypotension
E1 Maintains an open ductus arteriosus in the µg/kg/min Risk of apnea
newborn with ductal-dependent congenital
heart disease
Milrinone Increased cardiac contractility Load 50 Phosphodiesterase inhibitor—slows
µg/kg cyclic adenosine monophosphate
over 15 breakdown
min
Improves cardiac diastolic function 0.5-1.0
Peripheral vasodilation µg/kg/min
Table 89.1
Typical Localizing Signs for Pulmonary Pathology
Table 89.2
Examples of Anatomic Sites of Lesions Causing
Respiratory Failure
Table 89.3
Nonpulmonary Causes of Respiratory Distress
Table 89.5
Typical Chronology of Heart Disease Presentation in
Children
AGE MECHANISM DISEASE
Newborn (1-10 ↑ Arteriovenous pressure difference Arteriovenous fistula (brain, liver)
days) Ductal closure Single ventricle lesions or severe ventricular outflow
obstruction
Independent pulmonary and systemic Transposition of the great arteries
blood flow
Pulmonary venous obstruction Total anomalous pulmonary venous return (TAPVR)
Young infant (1-6 ↓ Pulmonary vascular resistance Left-to-right shunt
mo) ↓ Pulmonary artery pressure Anomalous left coronary artery to the pulmonary
artery
Any age Rate disturbance Tachy- or bradyarrhythmias
Infection Myocarditis, pericarditis
Abnormal cardiac myocytes Cardiomyopathy
Excess afterload hypertension
Table 89.6
VT , Tidal volume.
Table 89.7
Pediatric Acute Respiratory Distress Syndrome (PARDS)
Definition
b If altitude is >1,000 m, the correction factor should be calculated as follows: PaO /FIO ×
2 2
Barometric pressure/760.
c This may be delivered noninvasively in the mild acute respiratory distress syndrome group.
d Use PAO -based metric when available. If PaO not available, wean FIO to maintain SpO
2 2 2 2
<97% to calculate OSI or SF ratio.
e For nonintubated patients treated with supplemental oxygen or nasal modes of noninvasive
ventilation, refer to reference below for “At Risk Criteria.”
of 1) between 30% and 65%. If more precise delivery of oxygen is desired, other
mask devices should be used.
A Venturi mask provides preset FIO 2 through a mask and reservoir system by
entraining precise flow rates of room air into the reservoir along with high-flow
oxygen. The adapter at the end of each mask reservoir determines the flow rate
of entrained room air and the subsequent FIO 2 . (Adapters provide FIO 2 of 0.30-
0.50.) Oxygen flow rates of 5-10 L/min are recommended to achieve the desired
FIO 2 and to prevent rebreathing. Partial rebreather and non-rebreather masks use
a reservoir bag attached to a mask to provide higher FIO 2 . Partial rebreather
masks have 2 open exhalation ports and contain a valveless oxygen reservoir
bag. Some exhaled gas can mix with reservoir gas, although most exhaled gas
exits the mask via the exhalation ports. Through these same ports, room air is
entrained, and the partial rebreather mask can provide FIO 2 up to 0.60, for as
long as oxygen flow is adequate to keep the bag from collapsing (typically 10-15
L/ min). As with nasal cannulas, smaller children with smaller tidal volumes
entrain less room air, and their Fio2 values will be higher. Non-rebreather
masks include 2 one-way valves, 1 between the oxygen reservoir bag and the
mask and the other on 1 of the 2 exhalation ports. This arrangement minimizes
mixing of exhaled and fresh gas and entrainment of room air during inspiration.
The 2nd exhalation port has no valve, a safeguard to allow some room air to
enter the mask in the event of disconnection from the oxygen source. A non-
rebreather mask can provide FIO 2 up to 0.95. The use of a non-rebreather mask
in conjunction with an oxygen blender allows delivery of FIO 2 between 0.50 and
0.95 (Table 89.8 ). When supplemental oxygen alone is inadequate to improve
oxygenation, or when ventilation problems coexist, additional therapies may be
necessary.
Table 89.8
Approximate Oxygen Delivery According to Device and Flow Rates in Infants and
Older Children*
DEVICE FLOW (L/min) FIO 2 DELIVERED
Nasal cannula 0.1-6 0.21-0.4
Simple face mask 5-10 0.4-0.6
Partial rebreather 6-15 0.55-0.7
Non-rebreather 6-15 0.7-0.95
Venturi mask 5-10 0.25-0.5
Hood/tent 7-12 0.21-1.0
High-flow systems 1-40 0.21-1.0
* Individual delivery varies and depends on the patient's size, respiratory rate, and volume moved
Airway Adjuncts
Maintenance of a patent airway is a critical step in maintaining adequate
oxygenation and ventilation. Artificial pharyngeal airways may be useful in
patients with oropharyngeal or nasopharyngeal airway obstruction and in those
with neuromuscular weakness in whom inherent extrathoracic airway resistance
contributes to respiratory compromise. An oropharyngeal airway is a stiff
plastic spacer with grooves along each side that can be placed in the mouth to
run from the teeth along the tongue to its base just above the vallecula. The
spacer prevents the tongue from opposing the posterior pharynx and occluding
the airway. Because the tip sits at the base of the tongue, it is usually not
tolerated by patients who are awake or whose gag reflex is strong. The
nasopharyngeal airway , or nasal trumpet , is a flexible tube that can be
inserted into the nose to run from the nasal opening along the top of the hard and
soft palate with the tip ending in the hypopharynx. It is useful in bypassing
obstruction from enlarged adenoids or from contact of the soft palate with the
posterior nasopharynx. Because it is inserted past the adenoids, a nasopharyngeal
airway should be used with caution in patients with bleeding tendencies.
Inhaled Gases
Helium-oxygen mixture (heliox) is useful in overcoming airway obstruction
and improving ventilation. Helium is much less dense and slightly more viscous
than nitrogen. When substituted for nitrogen, helium helps maintain laminar
flow across an obstructed airway, decreases airway resistance, and improves
ventilation. It is especially helpful in diseases of large airways obstruction in
which turbulent airflow is more common, such as acute
laryngotracheobronchitis, subglottic stenosis, and vascular ring. It is also used in
patients with severe status asthmaticus. To be effective, helium should be
administered in concentrations of at least 60%, so associated hypoxemia may
limit its use in patients requiring >40% oxygen.
Inhaled nitric oxide (iNO) is a powerful inhaled pulmonary vasodilator. Its
use may improve pulmonary blood flow and V̇/Q̇ mismatch in patients with
common pharmacologic regimen for facilitating intubation. In fact, sedation and
paralysis with neuromuscular blocking agents should be considered standard
unless contraindicated. The particular type and dose of each agent often depends
on the underlying disease and clinician preference. Table 89.10 lists commonly
used agents. Dexmedetomidine has been a standard sedating agent for
maintenance during mechanical ventilation. An alternative to this pharmacologic
approach is rapid sequence intubation , used when endotracheal intubation is
urgent, or the patient is suspected of having a full stomach and at increased risk
of aspiration (see Chapter 81 ).
Table 89.9
Average Size and Depth Dimensions for Tracheal Tubes
Table 89.10
Medications Commonly Used for Intubation
Assist-Control Mode.
In assist-control (AC) mode, every patient breath is triggered by pressure or flow
generated by patient inspiratory effort and “assisted” with either preselected
inspiratory pressure or volume. The rate of respirations is therefore determined
by the patient's inherent rate. A backup total (patient and ventilator) obligatory
rate is set to deliver a minimum number of breaths. On AC mode, with a backup
rate of 20 breaths/min and a patient's inherent rate of 15 breaths/min; the
ventilator will assist all the patient's breaths, and the patient will receive 5
additional breaths/min. On the other hand, a patient with an inherent rate of 25
breaths/min will receive all 25 breaths assisted. Although useful in some
patients, the AC mode cannot be used in the weaning process, which involves
gradual decrease in ventilator support.
Control Variable
Once initiated, either the VT or the pressure delivered by the machine can be
controlled. The machine-delivered breath is thus referred to as either volume
controlled or pressure controlled (Table 89.11 ). With volume-controlled
ventilation (VCV) , machine-delivered volume is the primary control, and the
inflation pressure generated depends on the respiratory system's compliance and
resistance. Changes in respiratory system compliance and resistance are
therefore easily detected from changes observed in inflation pressure. In
pressure-controlled ventilation (PCV) the pressure change above the baseline
is the primary control, and the Vt delivered to the lungs depends on the
respiratory system's C and R. Changes in respiratory system C and R do not
affect inflation pressure and may therefore go undetected unless the exhaled Vt
is monitored.
Table 89.11
VCV and PCV have their own advantages and disadvantages (Table 89.11 ).
Generally speaking, PCV is more efficient than VCV in terms of amount of VT
delivered for a given inflation pressure during ventilation of a lung that has
nonuniform TC, as in asthma. In VCV, relatively less-obstructed airways are
likely to receive more of the machine-delivered volume throughout inspiration
than relatively more-obstructed airways with longer TC (Fig. 89.11 A ). This
situation would result in uneven ventilation, higher peak inspiratory pressure
(PIP), and a decrease in CDYN . In PCV, because of a constant inflation pressure
that is held throughout inspiration, relatively less-obstructed lung units with
shorter TC would achieve pressure equilibration earlier during inspiration than
the relatively more-obstructed areas. Thus, units with shorter TCs would attain
their final volume earlier in inspiration, and those with longer TCs would
continue to receive additional volume later in inspiration (Fig. 89.11 B ). This
situation would result in more even distribution of inspired gas, delivery of more
VT for the same inflation pressure, and improved CDYN compared with VCV.
withdrawal manifestations, and prolonged neuromuscular blockade is associated
with neuromyopathy in critically ill patients. The benefits of sedation and
pharmacologic paralysis therefore should be carefully balanced with the risks,
and periodic assessments should be made to determine the need for their
continuation.
Cardiopulmonary Interactions
Mechanical ventilation can have salutary as well as adverse effects on cardiac
performance. By decreasing oxygen consumption necessary for work of
breathing, oxygen supply to vital organs is improved. Positive pressure breathing
decreases LV afterload, thus enhancing stroke volume and cardiac output in
patients with failing myocardium (e.g., myocarditis). On the other hand, the
decreased systemic venous return may further compromise stroke volume in
hypovolemic patients. Such patients will require intravascular fluid loading.
Also, an increase in pulmonary vascular resistance (PVR) caused by positive
intrathoracic pressure may result in further decompensation of a poorly
performing right ventricle. PVR is at its lowest value at an optimum FRC. When
FRC is too low or too high, PVR (and therefore the right ventricular afterload) is
increased. Both desirable and undesirable effects of cardiopulmonary
interactions may coexist and require ongoing assessment and necessary
interventions (Table 89.12 ).
Table 89.12
Table 90.1
Medications for Treatment of Altitude-Associated Illness in
Children (No Studies in Children for High-Altitude
Indications)
MEDICATION CLASSIFICATION INDICATION DOSE AND ROUTE ADVERSE EFFECTS
Acetazolamide Carbonic anhydrase AMS 2.5 mg/kg PO every 12 Collateral effects include
inhibitor prevention* hr; max 125 mg/dose paresthesias and taste
alteration
AMS treatment 2.5 mg/kg PO every 12
hr; max 250 mg/dose
Dexamethasone Steroid AMS Risk of adverse effects
prevention † precludes prophylactic use
AMS HACE 0.15 mg/kg PO/IM/IV Hypertension, gastrointestinal
treatment ‡ every 6 hr; max 4 hemorrhage, pancreatitis,
mg/dose growth inhibition
Nifedipine Calcium channel HAPE 0.5 mg/kg PO every 4-8 Hypotension
blocker treatment hr; max 20 mg/dose
(small
children) §
HAPE 30 mg SR PO every 12
treatment (>60 hr or 20 mg SR PO
kg) § every 8 hr
Reentry HAPE Same dose as HAPE
prevention treatment
Sildenafil Phosphodiesterase-5 HAPE ¶ 0.5 mg/kg/dose PO
inhibitor every 4-8 hr; max 50
mg/dose every 8 hr
*
AMS prophylaxis is not routinely recommended in children. It is indicated when rapid ascent
profile is unavoidable or with previous altitude illness in child about to undergo similar ascent
profile. Doses as low as 1.25 mg/kg every 12 hr have been successful in some children.
† Use not warranted due to risk of adverse effects. Use slow, graded ascent or acetazolamide.
‡ Oxygen and descent are the treatment of choice for severe AMS. If acetazolamide is not
tolerated, dexamethasone may be used. Oxygen, descent, and dexamethasone should be used in
HACE.
§ In emergency settings where oxygen and descent are not an option, nifedipine is indicated.
¶
In emergency settings where oxygen and descent are not an option, if nifedipine is not well
tolerated, sildenafil may provide an alternative.
AMS, Acute mountain sickness; HACE, high-altitude cerebral edema; HAPE, high-altitude
pulmonary edema, IM, intramuscularly; IV, intravenously; PO, orally; SR, sustained release.
Diagnosis
AMS is easily identified in older children and adolescents using the Self-Report
Lake Louise AMS Scoring System . The criteria require that the individual be
in the setting of a recent gain in altitude, be at the new altitude for at least several
hours, and report a headache plus at least 1 of the following symptoms:
gastrointestinal (GI) upset (anorexia, nausea, or vomiting), general weakness or
fatigue, dizziness or lightheadedness, or difficulty sleeping. Shortness of breath
minimal admixture with other populations, represent the extreme of adaptation
to high altitude and rarely experience HAPE. Other native populations residing
at high altitude, such as Andeans, do not appear to be protected from HAPE, and
certain populations may have genetic polymorphisms associated with pulmonary
edema.
A number of conditions may predispose a child to HAPE (Table 90.2 ).
Preexisting viral respiratory infections have been linked to HAPE, especially in
children. Cardiorespiratory conditions associated with pulmonary hypertension,
such as atrial and ventricular septal defects, pulmonary vein stenosis, congenital
absence of a pulmonary artery, and OSA, also predispose to HAPE. Down
syndrome is a risk factor for HAPE development, as are previously repaired
congenital heart defects and the presence of hypoplastic lungs. Undiagnosed
structural cardiopulmonary abnormalities may result in severe hypoxia and/or
altitude illness once ascent occurs.
Table 90.2
Conditions Associated With Increased Risk of
High-Altitude Pulmonary Edema (HAPE)
Environmental
Cardiac
Infectious
Pharmacologic
Systemic
Pathophysiology
Alveolar hypoxia results in vasoconstriction of pulmonary arterioles just
proximal to the alveolar capillary bed. Hypoxic pulmonary vasoconstriction is a
normal physiologic response to optimize ventilation/perfusion (V̇/Q̇) matching
preventive strategy for exposure to all water types and all ages is ensuring
appropriate supervision . Pediatricians should define for parents what
constitutes appropriate supervision at the various developmental levels of
childhood. Many parents either underestimate the importance of adequate
supervision or are simply unaware of the risks associated with water. Even
parents who say that constant supervision is necessary will often admit to brief
lapses while their child is alone near water. Parents also overestimate the
supervisory abilities of older siblings; many bathtub drownings occur when an
infant or toddler is left with a child <5 yr old.
Table 91.1
Approach to Prevention Strategies for Drowning
Prevent Injury
Winter :
◆ Glass front fireplaces/pellet stoves and radiators increase hand
burns.
◆ Treadmill injuries as more people exercise inside—child
imitates adults or young child touches belt.
Summer :
◆ Fireworks, sparkler—temperatures reach 537.8°C (1,000°F).
◆ Burn contact with hot grill; hand/feet burn from hot embers.
◆ Lawnmowers
Spring/Fall :
◆ Burning leaves
◆ Gasoline burns
◆ Tap water scalds are essentially preventable through a
combination of behavioral and environmental changes.
Pediatricians can play a major role in preventing the most common burns by
educating parents and healthcare providers. Simple, effective, efficient, and cost-
effective preventive measures include the use of appropriate clothing and smoke
detectors and the planning of routes for emergency exit from the home. The
National Fire Protection Association (NFPA) recommends replacing smoke
detector batteries annually and the smoke detector alarm every 10 yr (or earlier,
if indicated on the device). Child neglect and abuse must be seriously considered
when the history of the injury and the distribution of the burn do not match.
1. Extinguish flames by rolling the child on the ground; cover the child
with a blanket, coat, or carpet.
2. After determining that the airway is patent, remove smoldering clothing
or clothing saturated with hot liquid. Jewelry, particularly rings and
bracelets, should be removed or cut away to prevent constriction and
vascular compromise during the edema phase in the first 24-72 hr after
burn injury.
3. In cases of chemical injury , brush off any remaining chemical, if
powdered or solid; then use copious irrigation or wash the affected area
with water. Call the local poison control center for the neutralizing agent
to treat a chemical ingestion.
4. Cover the burned area with clean, dry sheeting and apply cold (not iced)
wet compresses to small injuries. Significant large-burn injury (>15% of
BSA) decreases body temperature control and contraindicates the use of
cold compresses.
5. If the burn is caused by hot tar , use mineral oil to remove the tar.
6. Administer analgesic medications.
Emergency Care
Supportive measures are as follows (Table 92.3 and Table 92.4 )
Table 92.3
Acute Treatment of Burns
Table 92.4
Table 92.5
Categories of Burn Depth
FIG. 92.2 Diagram of different burn depths. (From Hettiaratchy S, Papini R: Initial
outpatient basis unless family support is judged inadequate or there are issues of
child neglect or abuse. These outpatients do not require a tetanus booster (unless
not fully immunized) or prophylactic penicillin therapy. Blisters should be left
intact and dressed with bacitracin or silver sulfadiazine cream (Silvadene).
Dressings should be changed once daily, after the wound is washed with
lukewarm water to remove any cream left from the previous application. Very
small wounds, especially those on the face, may be treated with bacitracin
ointment and left open. Debridement of the devitalized skin is indicated when
the blisters rupture. A variety of wound dressings and wound membranes (e.g.,
AQUACEL Ag dressing [ConvaTec USA, Skillman, NJ] in soft felt-like material
impregnated with silver ion) may be applied to 2nd-degree burns and wrapped
with a dry sterile dressing. Similar wound membranes provide pain control,
prevent wound desiccation, and reduce wound colonization (Table 92.6 ). These
dressings are usually kept on for 7-10 days but are checked twice a week.
Table 92.6
Burns to the palm with large blisters usually heal beneath the blisters; they
should receive close follow-up on an outpatient basis. The great majority of
superficial burns heal in 10-20 days. Deep 2nd-degree burns take longer to heal
and may benefit from enzymatic debridement ointment (collagenase) applied
daily on the wound, which aids in the removal of the dead tissue. These
ointments should not be applied to the face, to avoid the risk of getting into the
eyes.
The depth of scald injuries is difficult to assess early; conservative treatment
is appropriate initially, with the depth of the area involved determined before
grafting is attempted (Fig. 92.4 ). This approach obviates the risk of anesthesia
large open wound, reduction of the patient's host resistance, and malnutrition.
These abnormalities set the stage for life-threatening bacterial infection
originating from the burn wound. Wound treatment and prevention of wound
infection also promote early healing and improve aesthetic and functional
outcomes. Topical treatment of the burn wound with 0.5% silver nitrate solution,
silver sulfadiazine cream, or mafenide acetate (Sulfamylon) cream or topical
solution at a concentration of 2.5–5% to be used for wounds with multidrug–
resistant bacteria aims at prevention of infection (Table 92.7 ). These 3 agents
have tissue-penetrating capacity. Regardless of the choice of topical
antimicrobial agent, it is essential that all 3rd-degree burn tissue be fully excised
before bacterial colonization occurs, and that the area is grafted as early as
possible to prevent deep wound sepsis. Children with a burn of >30% BSA
should be housed in a bacteria-controlled nursing unit to prevent cross-
contamination and to provide a temperature- and humidity-controlled
environment to minimize hypermetabolism.
Table 92.7
Topical Agents Used for Burns
Deep 3rd-degree burns of >10% BSA benefit from early excision and grafting.
To improve outcome, sequential excision and grafting of 3rd-degree and deep
2nd-degree burns is required in children with large burns. Prompt excision with
immediate wound closure is achieved with autografts , which are often meshed
to increase the efficiency of coverings. Alternatives for wound closure, such as
recovery. Table 92.8 lists the long-term disabilities and complications of burns.
Table 92.8
Common Long-Term Complications and
Disabilities in Patients With Burn Injuries
Complications Affecting the Skin and Soft Tissue
Hypertrophic scars
Susceptibility to minor trauma
Dry skin
Contractures
Itching and neuropathic pain
Alopecia
Chronic open wounds
Skin cancers
Orthopedic Disabilities
Amputations
Contractures
Heterotopic ossification
Temporary reduction in bone density
Metabolic Disabilities
Heat sensitivity
Obesity
Sleep disorders
Adjustment disorders
Posttraumatic stress disorder
Depression
Body image issues
Neuropathy and neuropathic pain
Long-term neurologic effects of carbon monoxide poisoning
Anoxic brain injury
Risk-taking behavior
Untreated or poorly treated psychiatric disorder
Modified from Sheridan RL, Schultz JT, Ryan CM, et al: Case records of the
Massachusetts General Hospital. Weekly clinicopathological exercises. Case 6-
2004: a 35-year-old woman with extensive, deep burns from a nightclub fire, N
Engl J Med 350:810–821, 2004.
Electrical Burns
There are 3 types of electrical burns: extension cord (minor), high-tension wire,
and lightning. Minor electrical burns usually occur as a result of biting on an
extension cord. These injuries produce localized burns to the mouth, which
usually involve the portions of the upper and lower lips that come in contact with
the extension cord. The injury may involve or spare the corners of the mouth.
Because these are nonconductive injuries (do not extend beyond the site of
injury), hospital admission is not necessary, and care is focused on the area of
the injury visible in the mouth, ensuring it is low voltage and does not cause
entry or exit wounds or cardiac issues. Treatment with topical antibiotic creams
Table 92.9
Electrical Injury: Clinical Considerations
Atropine
Digoxin
Fentanyl
Gentamicin
Lidocaine
Phenobarbital
Procaine
Propranolol
Sulfanilamide (AVC cream)
Succinylcholine
D -Tubocurarine
Adapted from Bope ET, Kellerman RD, editors: Conn's current therapy 2014 ,
Philadelphia, 2014, Elsevier/Saunders (Box 3, p 1135).
Hypothermia occurs when the body can no longer sustain normal core
temperature by physiologic mechanisms, such as vasoconstriction, shivering,
muscle contraction, and nonshivering thermogenesis. When shivering ceases, the
body is unable to maintain its core temperature; when the body core temperature
falls to <35°C (95°F), the syndrome of hypothermia occurs. Wind chill, wet or
inadequate clothing, and other factors increase local injury and may cause
dangerous hypothermia, even in the presence of an ambient temperature that is
not <17-20°C (50-60°F).
Clinical Manifestations
Frostnip
Frostnip results in the presence of firm, cold, white areas on the face, ears, or
Application of petrolatum (Vaseline) to the nose and ears helps protect against
frostbite.
Treatment at the scene aims at prevention of further heat loss and early
transport to adequate shelter (Table 93.2 ). Dry clothing should be provided as
soon as practical, and transport should be undertaken if the victim has a pulse. If
no pulse is detected at the initial review, cardiopulmonary resuscitation is
indicated (Fig. 93.1 ) (see Chapter 81 ). During transfer, jarring and sudden
motion should be avoided because of the risk of ventricular arrhythmia. It is
often difficult to attain a normal sinus rhythm during hypothermia.
Table 93.2
Management of Hypothermia
History and Physical Examination
Laboratory Tests
Passive Rewarming
Active Rewarming
<32°C (89.6°F), cardiovascular instability, patients at risk for developing
hypothermia
Close monitoring for core-temperature afterdrop
Acute: external and/or core rewarming
Chronic (<32°C [89.6°F] for >24 hr): core rewarming
Extracorporeal membrane oxygenation
Availability of rapid deployment
Adapted from Burg FD, Ingelfinger JR, Polin RA, Gershon AA, editors: Current
pediatric therapy, ed 18, Philadelphia, 2006, Saunders/Elsevier (Table 4, p 174).
than one disease gene contributes to a complex, or “blended,” phenotype. The
ability to sequence hundreds to thousands of genes at once has provided insight
into this added layer of complexity in disease pathogenesis.
Table 94.1
Approaches for Genetic Testing
TYPE OF
SAMPLE
MUTATION RESOLUTION ADVANTAGES DISADVANTAGES
REQUIREMENTS
TESTING
Linkage Depends on Possible when Can give only diagnostic Requires multiple
analysis location of specific disease- probability based on likelihood of family members with
polymorphic causing genetic genetic recombination between documented mendelian
markers near mutation is not presumed DNA mutation and pattern of inheritance
putative disease identifiable or polymorphic markers within family
gene found
Array Several hundred Able to detect Can miss small deletions or Single patient sample
comparative base pairs to small deletion or insertions depending on sufficient, but having
genomic several duplications within resolution of the array used sample from biological
hybridization hundreds of 1 or more genes parents can help with
(aCGH) kilobases interpretation
Direct DNA- Single–base-pair High specificity if Can miss deletion or duplication Single patient sample
based testing changes previously of a segment of gene sufficient, but having
(e.g., DNA described sample from biological
sequencing) deleterious parents can help with
mutation is found interpretation
Table 94.2
Table 95.1
Table 96.1
Main Classes, Groups, and Types of Sequence Variants
and Their Effects on Protein Products
From Turnpenny P, Ellard S (Editors): Emery's elements of medical genetics, ed 14, Philadelphia,
2012, Elsevier/Churchill Livingstone, p 23.
Table 97.1
Representative Examples of Disorders Caused by
Mutations in Mitochondrial DNA and Their Inheritance
The mitochondria are the cell's suppliers of energy, and it is not surprising that
the organs that are most affected by the presence of abnormal mitochondria are
those that have the greatest energy requirements, such as the brain, muscle,
heart, and liver (see Chapters 105.4 , 388 , 616.2 , and 629.4 ) (Fig. 97.16 ).
Common manifestations include developmental delay, seizures, cardiac
dysfunction, decreased muscle strength and tone, and hearing and vision
problems.
screening tests for mitochondrial disorders.
Table 97.2
Diseases Associated With Polynucleotide Repeat
Expansions
PARENT IN
WHOM LOCATION
REPEAT NORMAL ABNORMAL
DISEASE DESCRIPTION EXPANSION OF
SEQUENCE RANGE RANGE
USUALLY EXPANSION
OCCURS
CATEGORY 1
Huntington disease Loss of motor CAG 6-34 36-100 or More often Exon
control, more through father
dementia,
affective disorder
Spinal and bulbar Adult-onset CAG 11-34 40-62 More often Exon
muscular atrophy motor-neuron through father
disease
associated with
androgen
insensitivity
Spinocerebellar ataxia Progressive CAG 6-39 41-81 More often Exon
type 1 ataxia, through father
dysarthria,
dysmetria
Spinocerebellar ataxia Progressive CAG 15-29 35-59 — Exon
type 2 ataxia, dysarthria
Spinocerebellar ataxia Dystonia, distal CAG 13-36 68-79 More often Exon
type 3 (Machado- muscular through father
Joseph disease) atrophy, ataxia,
external
ophthalmoplegia
Spinocerebellar ataxia Progressive CAG 4-16 21-27 — Exon
type 6 ataxia,
dysarthria,
nystagmus
Spinocerebellar ataxia Progressive CAG 7-35 38-200 More often —
type 7 ataxia, through father
dysarthria,
retinal
degeneration
Spinocerebellar ataxia Progressive CAG 29-42 47-55 — Exon
type 17 ataxia, dementia,
bradykinesia,
dysmetria
Dentatorubral- Cerebellar CAG 7-25 49-88 More often Exon
pallidoluysian atrophy, ataxia, through father
atrophy/Haw River myoclonic
syndrome epilepsy,
choreoathetosis,
dementia
CATEGORY 2
Pseudoachondroplasia, Short stature, GAC 5 6-7 — Exon
multiple epiphyseal joint laxity,
dysplasia degenerative
joint disease
Oculopharyngeal Proximal limb GCG 6 7-13 — Exon
muscular dystrophy weakness,
dysphagia, ptosis
Cleidocranial Short stature, GCG, GCT, 17 27 (expansion — Exon
dysplasia open skull GCA observed in 1
sutures with family)
bulging calvaria,
clavicular
hypoplasia,
shortened
fingers, dental
anomalies
Synpolydactyly Polydactyly and GCG, GCT, 15 22-25 — Exon
syndactyly GCA
CATEGORY 3
Myotonic dystrophy Muscle loss, CTG 5-37 100 to several Either parent, 3′ untranslated
(DM1; chromosome cardiac thousand but expansion region
19) arrhythmia, to congenital
cataracts, frontal form through
balding mother
Myotonic dystrophy Muscle loss, CCTG <75 75-11,000 — 3′ untranslated
(DM2; chromosome 3) cardiac region
arrhythmia,
cataracts, frontal
balding
Friedreich ataxia Progressive limb GAA 7-2 200-900 or Autosomal Intron
ataxia, more recessive
dysarthria, inheritance, so
hypertrophic disease alleles
cardiomyopathy, are inherited
pyramidal from both
weakness in legs parents
Fragile X syndrome Intellectual CGG 6-52 200-2,000 or Exclusively 5′ untranslated
(FRAXA) impairment, more through region
large ears and mother
jaws,
macroorchidism
in males
Fragile site (FRAXE) Mild intellectual GCC 6-35 >200 More often 5′ untranslated
impairment through region
mother
Spinocerebellar ataxia Adult-onset CTG 16-37 107-127 More often 3′ untranslated
type 8 ataxia, through region
dysarthria, mother
nystagmus
Spinocerebellar ataxia Ataxia and ATTCT 12-16 800-4,500 More often Intron
type 10 seizures through father
Spinocerebellar ataxia Ataxia, eye CAG 7-28 66-78 — 5′ untranslated
type 12 movement region
disorders;
variable age at
onset
Progressive myoclonic Juvenile-onset 12-bp repeat 2-3 30-75 Autosomal 5′ untranslated
epilepsy type 1 convulsions, motif recessive region
myoclonus, inheritance, so
dementia transmitted by
both parents
From Jorde LB, Carey JC, Bamshad MJ, et al: Medical genetics , ed 3, St Louis, 2006, Mosby, p
82.
CHAPTER 98
Cytogenetics
Carlos A. Bacino, Brendan Lee
Table 98.1
Incidence of Chromosomal Abnormalities in Newborn
Surveys
98.1
Methods of Chromosome Analysis
Carlos A. Bacino, Brendan Lee
FIG. 98.3 Example of different chromosome types according to the position of the
centromere. On the left is a chromosome 1 pair with the centromere equidistant from
the short and long arm (also known as metacentric ). In the center is a chromosome 11
pair that is submetacentric. On the right is a chromosome 13 pair that is an example of
an acrocentric chromosome. Acrocentric chromosomes contain a very small short arm,
stalks, and satellite DNA. The black arrow indicates the position of the centromere. The
blue arrow shows the long arm of a chromosome. The red arrow shows the short arm
of a chromosome. The green arrow highlights the satellite region, which is made of
DNA repeats. The light area between the short arm and the satellite is known as the
stalk.
Table 98.2
Some Abbreviations Used for Description of
Chromosomes and Their Abnormalities
Molecular techniques (e.g., FISH, CMA, aCGH) have filled a significant void
for the diagnosing cryptic chromosomal abnormalities. These techniques identify
subtle abnormalities that are often below the resolution of standard cytogenetic
studies. Fluorescence in situ hybridization (FISH) is used to identify the
presence, absence, or rearrangement of specific DNA segments and is performed
with gene- or region-specific DNA probes. Several FISH probes are used in the
clinical setting: unique sequence or single-copy probes, repetitive-sequence
probes (alpha satellites in the pericentromeric regions), and multiple-copy probes
(chromosome specific or painting) (Fig. 98.4A and B ). FISH involves using a
unique, known DNA sequence or probe labeled with a fluorescent dye that is
complementary to the studied region of disease interest. The labeled probe is
exposed to the DNA on a microscope slide, typically metaphase or interphase
with a trisomy exhibit a consistent and specific phenotype depending on the
chromosome involved.
FISH is a technique that can be used for rapid diagnosis in the prenatal
detection of common fetal aneuploidies, including chromosomes 13, 18, and 21,
as well as sex chromosomes (see Fig. 98.4C and D ). Direct detection of fetal
cell-free DNA from maternal plasma for fetal trisomy is a safe and highly
effective screening test for fetal aneuploidy. The most common numerical
abnormalities in liveborn children include trisomy 21 (Down syndrome), trisomy
18 (Edwards syndrome), trisomy 13 (Patau syndrome), and sex chromosomal
aneuploidies: Turner syndrome (usually 45,X), Klinefelter syndrome (47,XXY),
47,XXX, and 47,XYY. By far the most common type of trisomy in liveborn
infants is trisomy 21 (47,XX,+21 or 47,XY,+21) (see Table 98.1 ). Trisomy 18
and trisomy 13 are relatively less common and are associated with a
characteristic set of congenital anomalies and severe intellectual disability (Table
98.3 ). The occurrence of trisomy 21 and other trisomies increases with
advanced maternal age (≥35 yr). Because of this increased risk, women who are
≥35 yr old at delivery should be offered genetic counseling and prenatal
diagnosis (including serum screening, ultrasonography, and cell-free fetal DNA
detection, amniocentesis, or chorionic villus sampling; see Chapter 115 ).
Table 98.3
Chromosomal Trisomies and Their Clinical Findings
Table 98.4
Clinical Features of Down Syndrome in the
Neonatal Period
Central Nervous System
Hypotonia*
Developmental delay
Poor Moro reflex*
Craniofacial
Cardiovascular
Musculoskeletal
Joint hyperflexibility*
Short neck, redundant skin*
Short metacarpals and phalanges
Short 5th digit with clinodactyly*
Single transverse palmar creases*
Wide gap between 1st and 2nd toes
Pelvic dysplasia*
Short sternum
Two sternal manubrium ossification centers
Gastrointestinal
Duodenal atresia
Annular pancreas
Tracheoesophageal fistula
Hirschsprung disease
Imperforate anus
Neonatal cholestasis
Cutaneous
Cutis marmorata
Table 98.5
Additional Features of Down Syndrome That
Can Develop or Become Symptomatic With
Time
Neuropsychiatric
Developmental delay
Seizures
Autism spectrum disorders
Behavioral disorders (disruptive)
Depression
Alzheimer disease
Sensory
Cardiopulmonary
Musculoskeletal
Atlantoaxial instability
Hip dysplasia
Slipped capital femoral epiphyses
Avascular hip necrosis
Recurrent joint dislocations (shoulder, knee, elbow, thumb)
Endocrine
Hematologic
Gastrointestinal
Celiac disease
Delayed tooth eruption
Respiratory
Obstructed sleep apnea
Frequent infections (sinusitis, nasopharyngitis, pneumonia)
Cutaneous
Hyperkeratosis
Seborrhea
Xerosis
Perigenital folliculitis
Developmental delay is universal (Tables 98.6 and 98.7 and Fig. 98.10 ).
Cognitive impairment does not uniformly affect all areas of development. Social
development is often relatively spared, but autism spectrum disorder can occur.
Children with Down syndrome have considerable difficulty using expressive
language. Understanding the individual developmental strengths and challenges
is necessary to maximize the educational process. Persons with Down syndrome
often benefit from programs aimed at cognitive training, stimulation,
development, and education. Children with Down syndrome also benefit from
anticipatory guidance, which establishes the protocol for screening, evaluation,
and care for patients with genetic syndromes and chronic disorders (Table 98.8 ).
Up to 15% of children with Down syndrome have misalignment of the 1st
cervical vertebra (C1), which places them at risk for spinal cord injury with neck
hyperextension or extreme flexion. Special Olympics recommends sports
participation and training but requires x-ray examination (full extension and
flexion views) of the neck before participation in sports that may result in
hyperextension or radical flexion or pressure on the neck or upper spine. Such
sports include diving starts in swimming, butterfly stroke, diving, pentathlon,
high jump, equestrian sports, gymnastics, football, soccer, alpine skinning, and
warm-up exercises placing stress on the head and neck. If atlantoaxial instability
is diagnosed, Special Olympics will permit participation if the parents or
guardians request so and only after obtaining written certification from a
physician and acknowledgment of the risks by the parent or guardian.
Table 98.6
Developmental Milestones
CHILDREN WITH DOWN SYNDROME UNAFFECTED CHILDREN
Milestone
Average (mo) Range (mo) Average (mo) Range (mo)
Smiling 2 1.5-3 1 1.5-3
Rolling over 6 2-12 5 2-10
Sitting 9 6-18 7 5-9
Crawling 11 7-21 8 6-11
Creeping 13 8-25 10 7-13
Standing 10 10-32 11 8-16
Walking 20 12-45 13 8-18
Talking, words 14 9-30 10 6-14
Talking, sentences 24 18-46 21 14-32
From Levine MD, Carey WB, Crocker AC, editors: Developmental-behavioral pediatrics, ed 2,
Philadelphia, 1992, Saunders.
Table 98.7
Self-Help Skills
Table 98.8
Health Supervision for Children With Down Syndrome
Compared with the general population, children with Down syndrome are at
increased risk for behavior problems; psychiatric comorbidity is an estimated
18–38% in this population. Common behavioral difficulties that occur in
children with Down syndrome include inattentiveness, stubbornness, and a need
for routine and sameness. Aggression and self-injurious behavior are less
common in this population than other children with similar degrees of
intellectual disability from other etiologies. All these behaviors can respond to
educational, behavioral, or pharmacologic interventions.
The life expectancy for children with Down syndrome is reduced and is
approximately 50-55 yr. Little prospective information about the secondary
medical problems of adults with Down syndrome is known. Retrospective
studies have shown premature aging and an increased risk of Alzheimer disease
in adults with Down syndrome. These studies have also shown unexpected
negative (protective) associations between Down syndrome and comorbidities.
Persons with Down syndrome have fewer-than-expected deaths caused by solid
tumors and ischemic heart disease. This same study reported increased risk of
adult deaths from congenital heart disease, seizures, and leukemia. In one large
study, leukemias accounted for 60% of all cancers in people with Down
syndrome and 97% of all cancers in children with Down syndrome. There was
decreased risk of solid tumors in all age-groups with Down syndrome, including
97% of the cases as a result of errors in meiosis. The majority of these occur in
maternal meiosis I (90%). Approximately 1% of persons with trisomy 21 are
mosaics, with some cells having 46 chromosomes, and another 4% have a
translocation that involves chromosome 21. The majority of translocations in
Down syndrome are fusions at the centromere between chromosomes 13, 14, 15,
21, and 22, known as robertsonian translocations. The translocations can be de
novo or inherited. Very rarely is Down syndrome diagnosed in a patient with
only a part of the long arm of chromosome 21 in triplicate (partial trisomy ).
Isochromosomes and ring chromosomes are other rarer causes of trisomy 21.
Down syndrome patients without a visible chromosome abnormality are the least
common. It is not possible to distinguish the phenotypes of persons with full
trisomy 21 and those with a translocation. Representative genes on chromosome
21 and their potential effects on development are noted in Table 98.9 . Patients
who are mosaic tend to have a milder phenotype.
Table 98.9
Genes Localized to Chromosome 21 That May Affect Brain
Development, Neuronal Loss, and Alzheimer-Type
Neuropathology
POSSIBLE
EFFECT IN
SYMBOL NAME FUNCTION
DOWN
SYNDROME
SIM2 Single-minded homolog 2 Brain development Required for synchronized cell division
and establishment of proper cell lineage
DYRK1A Dual-specificity tyrosine-(Y)- Brain development Expressed during neuroblast
phosphorylation regulated kinase proliferation
1A Believed important homolog in
regulating cell-cycle kinetics during
cell division
GART Phosphoribosylglycinamide Brain development Expressed during prenatal development
formyltransferase of the cerebellum
Phosphoribosylglycinamide
synthetase
Phosphoribosylaminoimidazole
synthetase
PCP4 Purkinje cell protein 4 Brain development Function unknown but found exclusively
in the brain and most abundantly in the
cerebellum
DSCAM Down syndrome cell adhesion Brain development Expressed in all molecule regions of the
molecule and possible brain and believed to have a role in
candidate gene for axonal outgrowth during development of
congenital heart the nervous system
disease
GRIK1 Glutamate receptor, ionotropic Neuronal loss Function unknown, found in the cortex
kainite1 in fetal and early postnatal life and in
adult primates, most concentrated in
pyramidal cells in the cortex
APP Amyloid beta (A4) precursor Alzheimer type Seems to be involved in plasticity,
protein (protease nexin-II, neuropathy neurite outgrowth, and neuroprotection
Alzheimer disease)
S100B S100 calcium binding protein β Alzheimer type Stimulates glial formation
(neural) neuropathy
SOD1 Superoxide dismutase 1, soluble Accelerated aging? Scavenges free superoxide molecules in
(amyotrophic lateral sclerosis, the cell and might accelerate aging by
adult) producing hydrogen peroxide and
oxygen
Table 98.10
Other Rare Aneuploidies and Partial Autosomal
Aneuploidies
Table 98.11
Findings That May Be Present in Trisomy 13 and Trisomy
18
TRISOMY 13 TRISOMY 18
HEAD AND FACE
Scalp defects (e.g., cutis aplasia) Small and premature appearance
Microphthalmia, corneal abnormalities Tight palpebral fissures
Cleft lip and palate in 60–80% of cases Narrow nose and hypoplastic nasal alae
Microcephaly Narrow bifrontal diameter
Microphthalmia Prominent occiput
Sloping forehead Micrognathia
Holoprosencephaly (arrhinencephaly) Cleft lip or palate
Capillary hemangiomas Microcephaly
Deafness
CHEST
Congenital heart disease (e.g., VSD, PDA, ASD) Congenital heart disease (e.g., VSD, PDA, ASD)
in 80% of cases Short sternum, small nipples
Thin posterior ribs (missing ribs)
EXTREMITIES
Overlapping of fingers and toes (clinodactyly) Limited hip abduction
Polydactyly Clinodactyly and overlapping fingers; index over
Hypoplastic nails, hyperconvex nails 3rd, 5th over 4th; closed fist
Rocker-bottom feet
Hypoplastic nails
GENERAL
Severe developmental delays and prenatal and Severe developmental delays and prenatal and
postnatal growth restriction postnatal growth restriction
Renal abnormalities Premature birth, polyhydramnios
Survival (see Table 98.3 ) Inguinal or abdominal hernias
Survival (see Table 98.3 )
ASD, Atrial septal defect; PDA, patent ductus arteriosus; VSD, ventricular septal defect.
From Behrman RE, Kliegman RM: Nelson essentials of pediatrics, ed 4, Philadelphia, 2002,
Saunders, p 142.
normal, but they are at increased risk for miscarriages, typically in paracentric
inversions, and chromosomally abnormal offspring in pericentric inversions.
Table 98.12
Table 98.13
9q22 Gorlin Multiple basal cell carcinomas, odontogenic keratocysts, palmoplantar pits,
calcification falx cerebri
9q34 9q34 deletion Distinct face with synophrys, anteverted nares, tented upper lip, protruding
tongue, midface hypoplasia, conotruncal heart defects, intellectual
disability
10p12-p13 DiGeorge type 2 Many of the DiGeorge type 1 and velocardiofacial type 1 features
(conotruncal defects, immunodeficiency, hypoparathyroidism, dysmorphic
features)
11p11.2 Potocki-Shaffer Multiple exostoses, parietal foramina, craniosynostosis, facial
dysmorphism, syndactyly, intellectual disability
11p13 WAGR Hypernephroma (W ilms tumor), a niridia, male g enital hypoplasia of
varying degrees, g onadoblastoma, long face, upward-slanting palpebral
fissures, ptosis, beaked nose, low-set poorly formed auricles, intellectual
disability (r etardation)
11q24.1- Jacobsen Growth restriction, intellectual disability, cardiac and digit anomalies,
11qter thrombocytopenia
15q11-q13 Prader-Willi Severe hypotonia and feeding difficulties at birth, voracious appetite and
(paternal) obesity in infancy, short stature (responsive to growth hormone), small
hands and feet, hypogonadism, intellectual disability
15q11-q13 Angelman Hypotonia, feeding difficulties, gastroesophageal reflux, fair hair and skin,
(maternal) midface hypoplasia, prognathism, seizures, tremors, ataxia, sleep
disturbances, inappropriate laughter, poor or absent speech, severe
intellectual disability
16p13.3 Rubinstein-Taybi Microcephaly, ptosis, beaked nose with low-lying philtrum, broad thumbs
and large toes, intellectual disability
17p11.2 Smith-Magenis Brachycephaly, midfacial hypoplasia, prognathism, myopia, cleft palate,
short stature, severe behavioral problems, intellectual disability
17p13.3 Miller-Dieker Microcephaly, lissencephaly, pachygyria, narrow forehead, hypoplastic
male external genitals, growth restriction, seizures, profound intellectual
disability
20p12 Alagille Bile duct paucity with cholestasis; heart defects, particularly pulmonary
artery stenosis; ocular abnormalities (posterior embryotoxon); skeletal
defects such as butterfly vertebrae; long nose
22q11.2 Velocardiofacial- Conotruncal cardiac anomalies, cleft palate, velopharyngeal incompetence,
DiGeorge hypoplasia or agenesis of thymus and parathyroid glands, hypocalcemia,
hypoplasia of auricle, learning disabilities, psychiatric disorders
22q13.3 Hypotonia, developmental delay, normal or accelerated growth, severe
deletion expressive language deficits, autistic behavior
Xp21.2- Duchenne muscular dystrophy, retinitis pigmentosa, adrenal hypoplasia,
p21.3 intellectual disability, glycerol kinase deficiency
Xp22.2- Ichthyosis, Kallmann syndrome, intellectual disability, chondrodysplasia
p22.3 punctata
Xp22.3 MLS M icrophthalmia, l inear s kin defects, poikiloderma, congenital heart
defects, seizures, intellectual disability
Table 98.14
Insertions
Insertions occur when a piece of a chromosome broken at 2 points is
incorporated into a break in another part of a chromosome. A total of 3
breakpoints are then required, and they can occur between 2 or within 1
chromosome. A form of nonreciprocal translocation, insertions are rare. Insertion
carriers are at risk of having offspring with deletions or duplications of the
inserted segment.
Isochromosomes
Isochromosomes consist of 2 copies of the same chromosome arm joined
through a single centromere and forming mirror images of one another. The most
commonly reported autosomal isochromosomes tend to involve chromosomes
with small arms. Some of the more common chromosome arms involved in this
formation include 5p, 8p, 9p, 12p, 18p, and 18q. There is also a common
isochromosome abnormality seen in long arm of the X chromosome and
associated with Turner syndrome. Individuals who have 1 isochromosome X
within 46 chromosomes are monosomic for genes in the lost short arm and
trisomic for the genes present in the long arm of the X chromosome.
Bibliography
Abnormalities of Chromosome Structure
Bassett AS, McDonald-McGinn DM, Devriendt K, et al.
Practical guidelines for managing patients with 22q11.2
deletion syndrome. J Pediatr . 2011;159:332–339.
Battaglia A, Hoyme HE, Dallapiccola B, et al. Further
delineation of deletion 1p36 syndrome in 60 patients: a
recognizable phenotype and common cause of developmental
delay and mental retardation. Pediatrics . 2008;121:404–410.
Calado RT, Young NS. Telomere diseases. N Engl J Med .
2009;361:2353–2365.
Fillion M, Deal C, Van Vliet G. Retrospective study of the
potential benefits and adverse events during growth hormone
treatment in children with Prader-Willi syndrome. J Pediatr .
2009;154:230–233.
Gicquel C, Rossignol S, Cabrol S, et al. Epimutation of the
telomeric imprinting center region on chromosome 11p15 in
Silver-Russell syndrome. Nat Genet . 2005;37:1003–1007.
Mefford H, Shapr A, Baker C, et al. Recurrent rearrangements
of chromosome 1q21.1 and variable pediatric phenotypes. N
Engl J Med . 2008;359:1685–1698.
Peters J. Prader-Willi and snoRNAs. Nat Genet . 2008;40:688–
689.
Rappold GA, Shanske A, Saenger P. All shook up by SHOX
deficiency. J Pediatr . 2005;147:422–424.
Robin NH, Shprintzen RJ. Defining the clinical spectrum of
deletion 22q11.2. J Pediatr . 2005;147:90–96.
Sahoo T, del Gaudio D, German JR, et al. Prader-Willi
phenotype caused by paternal deficiency for the HBII-85 C/D
box small nucleolar RNA cluster. Nat Genet . 2008;40:719–
721.
Sharp AJ, Mefford HC, Li K, et al. A recurrent 15q13.3
microdeletion syndrome associated with mental retardation
and seizures. Nat Genet . 2008;40:322–328.
Stafler P, Wallis C. Prader-Willi syndrome: who can have
growth hormone? Arch Dis Child . 2008;93:341–345.
Vissers LELM, van Ravenswaaji CMA, Admiraal R, et al.
Mutations in a new member of the chromodomain gene
family cause CHARGE syndrome. Nat Genet . 2004;36:955–
957.
Walter S, Sandig K, Hinkel GK, et al. Subtelomere FISH in 50
children with mental retardation and minor anomalies,
identified by a checklist, selects 10 rearrangements including
a de novo balanced translocation of chromosomes 17p13.3
and 20q13.33. Am J Med Genet . 2004;128A:364–373.
Youings S, Ellis K, Ennis S, et al. A study of reciprocal
translocations and inversions detected by light microscopy
with special reference to origin, segregation, and recurrent
abnormalities. Am J Med Genet . 2004;126A:46–60.
Yu S, Graf WD, Shprintzen RJ. Genomic disorders on
chromosome 22. Curr Opin Pediatr . 2012;24:665–671.
98.4
Sex Chromosome Aneuploidy
Carlos A. Bacino, Brendan Lee
About 1 in 400 males and 1 in 650 females have some form of sex chromosome
abnormality. Considered together, sex chromosome abnormalities are the most
common chromosome abnormalities seen in liveborn infants, children, and
adults. Sex chromosome abnormalities can be either structural or numerical and
can be present in all cells or in a mosaic form. Those affected with these
abnormalities might have few or no physical or developmental problems (Table
98.15 ).
Table 98.15
Sex Chromosome Abnormalities
APPROXIMATE
DISORDER KARYOTYPE
INCIDENCE
Klinefelter syndrome 47,XXY 1/580 males
48,XXXY 1/50,000-1/80,000 male births
Other (48,XXYY; 49,XXXYY;
mosaics)
XYY syndrome 47,XYY 1/800-1,000 males
Other X or Y chromosome 1/1,500 males
abnormalities
XX males 46,XX 1/20,000 males
Turner syndrome 45,X 1/2,500-1/5,000 females
Variants and mosaics
Trisomy X 47,XXX 1/1,000 females
48,XXXX and 49,XXXXX Rare
Other X chromosome abnormalities 1/3,000 females
XY females 46,XY 1/20,000 females
Turner Syndrome
Turner syndrome is a condition characterized by complete or partial monosomy
of the X chromosome and defined by a combination of phenotypic features
(Table 98.16 ). Half the patients with Turner syndrome have a 45,X chromosome
complement. The other half exhibit mosaicism and varied structural
abnormalities of the X or Y chromosome. Maternal age is not a predisposing
factor for children with 45,X. Turner syndrome occurs in approximately 1 in
5,000 female live births. In 75% of patients, the lost sex chromosome is of
paternal origin (whether an X or a Y). 45,X is one of the chromosome
abnormalities most often associated with spontaneous abortion. It has been
estimated that 95–99% of 45,X conceptions are miscarried.
Table 98.16
Signs Associated With Turner Syndrome
Short stature
Congenital lymphedema
Horseshoe kidneys
Patella dislocation
Increased carrying angle of elbow (cubitus valgus)
Madelung deformity (chondrodysplasia of distal radial epiphysis)
Congenital hip dislocation
Scoliosis
Widespread nipples
Shield chest
Redundant nuchal skin (in utero cystic hygroma)
Low posterior hairline
Coarctation of aorta
Bicuspid aortic valve
Cardiac conduction abnormalities
Hypoplastic left heart syndrome and other left-sided heart abnormalities
Gonadal dysgenesis (infertility, primary amenorrhea)
Gonadoblastoma (increased risk if Y chromosome material is present)
Learning disabilities (nonverbal perceptual motor and visuospatial skills)
(in 70%)
Developmental delay (in 10%)
Social awkwardness
Hypothyroidism (acquired in 15–30%)
Type 2 diabetes mellitus (insulin resistance)
Strabismus
Cataracts
Red-green color blindness (as in males)
Recurrent otitis media
Sensorineural hearing loss
Inflammatory bowel disease
Celiac disease (increased incidence)
Clinical findings in the newborns can include small size for gestational age,
webbing of the neck, protruding ears, and lymphedema of the hands and feet,
although many newborns are phenotypically normal (Fig. 98.17 ). Older children
and adults have short stature and exhibit variable dysmorphic features.
Congenital heart defects (40%) and structural renal anomalies (60%) are
common. The most common heart defects are bicuspid aortic valves, coarctation
of the aorta, aortic stenosis, and mitral valve prolapse. The gonads are generally
streaks of fibrous tissue (gonadal dysgenesis ). There is primary amenorrhea
and lack of secondary sex characteristics. These children should receive regular
endocrinologic testing (see Chapter 604 ). Most patients tend to be of normal
intelligence, but intellectual disability is seen in up to 6% of affected children.
They are also at increased risk for behavioral problems and deficiencies in
spatial and motor perception. Guidelines for health supervision for children with
Turner syndrome are published by the American Academy of Pediatrics (AAP)
and include pubertal induction, as well as treatment with growth hormone and
oxandrolone.
look for Y chromosome mosaicism in all 45,X patients. If Y chromosome
material is identified, laparoscopic gonadectomy is recommended.
Noonan syndrome shares many clinical features with Turner syndrome and
was formerly called pseudo-Turner syndrome , although it is an autosomal
dominant disorder resulting from mutations in several genes involved in the
RAS-MAPK (mitogen-activated protein kinase) pathway. The most common of
these is PTPN11 (50%), which encodes a protein-tyrosine phosphatase (SHP-2)
on chromosome 12q24.1. Other genes include SOS1 in 10–13%, RAF1 in 3–
17%, RITI in 5%, KRAS <5%, BRAF <2%, MAP2K <2%, and NRAS (only few
reported families). Overlapping phenotypes are seen in LEOPARD (lentigines,
electrocardiographic abnormalities, ocular hypertelorism, pulmonary stenosis,
abnormalities of genitalia, retardation of growth, deafness) syndrome,
cardiofaciocutaneous (CFC) syndrome, and Costello syndrome; these are
Noonan-related disorders. Features common to Noonan syndrome include short
stature, low posterior hairline, shield chest, congenital heart disease, and a short
or webbed neck (Table 98.17 ). In contrast to Turner syndrome, Noonan
syndrome affects both sexes and has a different pattern of congenital heart
disease, typically involving right-sided lesions.
Table 98.17
Signs Associated With Noonan Syndrome
Short stature
Failure to thrive (best to use Noonan growth curve)
Tall forehead
Epicanthal folds
Ptosis
Blue-green irises
Hypertelorism
Low nasal bridge, upturned nose
Downward-slanting palpebral fissures
Myopia
Nystagmus
Low-set and posteriorly rotated auricles
Dental malocclusion
Low posterior hairline
Short, webbed neck (excessive nuchal skin), cystic hygroma
Shield chest
Pectus carinatum superiorly
Scoliosis
Pigmented villonodular synovitis (polyarticular)
Cubitus valgus
Pulmonary valve stenosis (dysplastic valve)
Hypertrophic cardiomyopathy
Atrial septal defect, ventricular septal defect
Lymphedema
Nevi, lentigines, café au lait spots
Cryptorchidism
Small penis
Delayed puberty
Bleeding disorders, including thrombocytopenia and coagulation factor
deficiencies
Leukemia, myeloproliferative disorders, other malignancies
Cognitive delay (KRAS mutation)
Klinefelter Syndrome
Persons with Klinefelter syndrome are phenotypically male; this syndrome is the
most common cause of hypogonadism and infertility in males and the most
common sex chromosome aneuploidy in humans (see Chapter 601 ). Eighty
percent of children with Klinefelter syndrome have a male karyotype with an
extra chromosome X-47,XXY. The remaining 20% have multiple sex
chromosome aneuploidies (48,XXXY; 48,XXYY; 49,XXXXY), mosaicism
(46,XY/47,XXY), or structurally abnormal X chromosomes; the greater the
aneuploidy, the more severe the mental impairment and dysmorphism. Early
studies showed a birth prevalence of approximately 1 in 1,000 males, but more
recent studies suggest that the prevalence of 47,XXY appears has increased to
approximately 1 in 580 liveborn males; the reasons for this are still unknown but
hypothesized to be the result of environmental factors acting in spermatogenesis.
Errors in paternal nondisjunction in meiosis I account for half the cases.
Puberty commences at the normal age, but the testes remain small. Patients
develop secondary sex characters late, and 50% ultimately develop
gynecomastia. They have taller stature. Because many patients with Klinefelter
syndrome are phenotypically normal until puberty, the syndrome often goes
PHENOTYPE
DISORDER Clinical and Imaging ONSET PENETRANCE
Cognitive or Behavioral
Signs
FULL MUTATION (>200 repeats)
FXS Developmental delay: Hypothalamic Neonate M 100%
mean IQ = 42 in M; IQ is dysfunction:
higher if significant macroorchidism,
residual FMRP is 40%*
produced (e.g., females Facial features,
and mosaic males or 60%,* large
unmethylated full cupped ears,
mutations) elongated face,
Autism 20–30% high arched palate
ADHD 80% Connective tissue
Anxiety 70–100% abnormalities:
mitral valve
prolapse, scoliosis,
joint laxity, flat
feet
Others: seizures
(20%), recurrent
otitis media
(60%), strabismus
(8–30%)
PREMUTATION (55-200 repeats)
Female reproductive POF (<40 yr) Adult/childhood F 20% †
symptoms Early menopause (<45 F 30% †
yr)
FXTAS Cognitive decline, dementia, Gait ataxia, intention >50 yr M 33% ‡
apathy, disinhibition, tremor, parkinsonism, F unknown
irritability, depression neuropathy, autonomic
dysfunction
Neurodevelopmental ADHD, autism, or Mild features of FXS Childhood 8% (1/13)*
disorder developmental delay
* Frequency of those signs in prepubertal boys; one third of boys with FXS are without classic
Table 98.19
Therapy for FMR1 -Related Disorders
FUTURE
DISORDER SYMPTOM THERAPY AND INTERVENTIONS POTENTIAL
THERAPY
FULL MUTATION
FXS* ADHD Stimulants mGluR5 antagonists
Anxiety, hyperarousal, SSRIs, atypical antipsychotics, occupational mGluR5 antagonists
aggressive outbursts therapy, behavioral therapy, counseling
Seizures Carbamazepine, valproic acid mGluR5 antagonists
Cognitive deficit Occupational therapy, speech therapy, special mGluR5 antagonists
education support
PREMUTATION
POF Premature ovarian failure Reproductive counseling, egg donation Cryopreservation of
ovarian tissue
Hormone replacement therapy
FXTAS † Intention tremor β-Blockers
Parkinsonism Carbidopa/levodopa
Cognitive decline, dementia Acetylcholinesterase inhibitors
Anxiety, apathy, Venlafaxine, SSRIs
disinhibition, irritability,
depression
Neuropathic pain Gabapentin
*
These data are based on a survey in 2 large referral centers. Drugs for anxiety were more
frequently prescribed than those for neurologic signs.
† There have been no controlled studies to assess drugs for FXTAS. These data were collected
Bibliography
Fragile Chromosome Sites
Abrams L, Cronister A, Brown WT, et al. Newborn, carrier, and
early childhood screening recommendations for fragile X.
A classic example of imprinting disorder is seen in Prader-Willi syndrome and
Angelman syndrome, 2 very different clinical conditions. These syndromes are
usually associated with deletion of the same region in the proximal long arm of
chromosome 15. A deletion on the paternally derived chromosome causes
Prader-Willi syndrome, in which the maternally derived copy is still intact, but
some of the imprinted genes within this region normally remain silent. Prader-
Willi syndrome can be diagnosed clinically (Table 98.20 ) and confirmed with
genetic testing. Additional clinical features and issues of weight gain are noted
in Table 98.21 . The weight gain is difficult to control, but treatment with growth
hormone has resulted in improvements in height, lean body mass, decreased
adipose tissue, and improvement in cognitive function.
Table 98.20
Table 98.21
Table 98.22
Bibliography
Uniparental Disomy and Imprinting
Al-Saleh S, Al-Naimi A, Hamilton J, et al. Longitudinal
evaluation of sleep-disordered breathing in children with
Prader-Willi syndrome during 2 years of growth hormone
therapy. J Pediatr . 2013;162:263–268.
Butler MG, Sturich J, Lee J, et al. Growth standards of infants
with Prader-Willi syndrome. J Pediatr . 2011;127:687–695.
Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi
syndrome. Genet Med . 2012;14:10–26.
Eggermann T. Russell-Silver syndrome. Am J Med Genet C
somatic mutations that can target various mediators of transcriptional regulation,
numerous therapeutic avenues have emerged that show promise. An interesting
noncancer example is the genetic condition called Kabuki syndrome , which is
caused by mutations in either a histone methyltransferase (KMT2D ) or a histone
demethylase (KDM6A ) gene, each of which has a role in creating accessible
chromatin to allow genes to be expressed appropriately (Fig. 100.3 and Table
100.1 ). With the idea that increasing the amount of histone acetylation could
help to compensate for the inappropriate histone methylation, mice with Kabuki
syndrome were placed on a ketogenic diet, which increases the amount of β-
hydroxybutyrate, an endogenous inhibitor of histone deacetylases. Mice on this
diet showed improved neurogenesis and memory, suggesting that such an
intervention in children with Kabuki syndrome may also have beneficial effects.
FIG. 100.3 Kabuki syndrome in 18 mo old boy. A, Long palpebral fissures and eversion
of lateral portion of lower eyelids. B, Prominent fingertip pads. (From Jones, KL, Jones
MC, Del Campo, M, editors: Smith's recognizable patterns of human malformation, ed 7,
Philadelphia, 2013, Elsevier, p 158.)
Table 100.1
Clinical Manifestations of Kabuki Syndrome
Facial
Neurodevelopmental
Hypotonia
Developmental delay (IQ about 60; >80 in 10%)
Low birthweight
Postnatal growth deficiency
Microcephaly
Seizures
Autism
Extremity/Skeletal
Cardiovascular
Other
Nonimmune hydrops
Hypothyroidism
Precocious puberty
Delayed puberty
Lymphatic malformations
Feeding difficulties
Bibliography
Banka S, Lederer D, Benoit V, et al. Novel KDM6A (UTX)
mutations and a clinical and molecular review of the X-linked
Kabuki syndrome (KS2). Clin Genet . 2015;87:252–258.
Benjamin JS, Pilarowski GO, Carosso GA, et al. A ketogenic
diet rescues hippocampal memory defects in a mouse model
of Kabuki syndrome. Proc Natl Acad Sci USA .
2017;114(1):125–130.
Birney E, Smith GD, Greally JM. Epigenome-wide association
studies and the interpretation of disease–omics. PLoS Genet .
2016;12(6):e1006105.
Blewitt M, Whitelaw E. The use of mouse models to study
epigenetics. Cold Spring Harb Perspect Biol .
2013;5(11):a017939.
Chen F, Marquez H, Kim Y-K, et al. Prenatal retinoid deficiency
leads to airway hyperresponsiveness in adult mice. J Clin
Invest . 2014;124(2):801–811.
Dentici ML, Di Pede A, Lepri FR, et al. Kabuki syndrome:
clinical and molecular diagnosis in the first year of life. Arch
Dis Child . 2015;100:158–164.
Henikoff S, Greally JM. Epigenetics, cellular memory and gene
regulation. Curr Biol . 2016;26(14):R644–R648.
Janesick AS, Shioda T, Blumberg B. Transgenerational
inheritance of prenatal obesogen exposure. Mol Cell
Endocrinol . 2014;398(1–2):31–35.
Jirtle RL. The Agouti mouse: a biosensor for environmental
physical findings include dysmorphisms, organomegaly, neurologic impairment,
bone involvement, and dermatologic findings. Because many rare and novel
disorders are multisystemic, consultants play a critical role in every diagnostic
evaluation. Typical studies performed to address possible diagnoses are listed in
Table 101.1 ; neurodevelopmental or neurodegenerative phenotypes require even
more extensive studies (Table 101.2 ).
Table 101.1
Table 101.2
Diagnostic Evaluation of the Neurologically
Impaired Child
Consultations
Genetics/genetic counseling
Neurology
Ophthalmology
Endocrinology
Immunology
Rheumatology
Dermatology
Cardiology
Neuropsychology
Nutrition
Rehabilitative medicine
Physical therapy
Occupational therapy
Speech therapy
Procedures
Laboratory Evaluations
Electron microscopy of white blood cell buffy coat for inclusion bodies
Electron microscopy of skin biopsy for evidence of storage
Stool for ova and parasites, occult blood, fecal fat, or fecal calprotectin
Autoimmune antibodies
Vaccine response titers
C3/C4
Quantitative immunoglobulins
T-cell subsets
Conjunctival or salivary gland biopsy
Research Specimens
Cerebrospinal fluid
Serum
Plasma
Skin biopsy for fibroblasts and/or melanocytes
Isolated DNA/RNA
Urine
Studies Under Sedation
Isovaleric acidemia
Glutaric aciduria type I
3-Hydroxy-3-methylglutaric aciduria
Multiple carboxylase deficiency
Methylmalonic acidemia (methylmalonyl-CoA mutase deficiency)
Methylmalonic acidemia (cbl A and cbl B defects)
Propionic acidemia
3-Methylcrotonyl-CoA carboxylase deficiency
β-Ketothiolase deficiency
Phenylketonuria
Maple syrup urine disease
Homocystinuria
Citrullinemia type 1
Argininosuccinic acidemia
Tyrosinemia type I
Hemoglobinopathies
Other Disorders
Congenital hypothyroidism
Biotinidase deficiency
Congenital adrenal hyperplasia
Galactosemia
Hearing loss
Cystic fibrosis
Severe combined immunodeficiency (SCID) †
Critical congenital heart disease †
list of the disorders that are screened for by each state is available at http://genes-
r-us.uthscsa.edu/sites/genes-r-us/files/nbsdisorders.pdf .
† The inclusion of SCID and critical congenital heart disease received support of
Hemoglobinopathies
Others
Galactose epimerase deficiency
Galactokinase deficiency
Table 102.3
DETERIORATION IN CONSCIOUSNESS
Metabolic Acidosis
Organic acidemias
Disorders of pyruvate metabolism
Fatty acid oxidation defects
Fructose-1,6-bisphosphatase deficiency
Glycogen storage diseases
Mitochondrial respiratory chain defects
Disorders of ketone metabolism
Hypoglycemia *
Fatty acid oxidation defects
Disorders of gluconeogenesis
Disorders of fructose and galactose metabolism
Glycogen storage diseases
Disorders of ketogenesis
Organic acidemias
Hyperinsulinemic hypoglycemias
Mitochondrial respiratory chain defects
Neonatal intrahepatic cholestasis caused by citrin deficiency
Pyruvate carboxylase deficiency
Carbonic anhydrase VA deficiency
Hyperammonemia **
Urea cycle disorders
Organic acidemias
Fatty acid oxidation disorders
Disorders of pyruvate metabolism
GLUD1 -related hyperinsulinemic hypoglycemia
Carbonic anhydrase VA deficiency
SEIZURES AND HYPOTONIA
Antiquitin deficiency (pyridoxine-dependent epilepsy)
Pyridoxamine 5'-phosphate oxidase (PNPO) deficiency (pyridoxal phosphate-responsive epilepsy)
Folate metabolism disorders
Multiple carboxylase deficiency (holocarboxylase synthetase deficiency and biotinidase deficiency)
Urea cycle disorders
Organic acidemias
Fatty acid oxidation disorders
Disorders of creatine biosynthesis and transport
Disorders of neurotransmitter metabolism
Molybdenum cofactor deficiency and sulfite oxidase deficiency
Serine deficiency disorders
Glycine encephalopathy
Asparagine synthetase deficiency
Mitochondrial respiratory chain defects
Zellweger spectrum disorders
Congenital disorders of glycosylation
Purine and pyrimidine metabolism defects
NEONATAL APNEA
Glycine encephalopathy
Asparagine synthetase deficiency
Urea cycle disorders
Organic acidemias
Disorders of pyruvate metabolism
Fatty acid oxidation defects
Mitochondrial respiratory chain defects
*
Refer to Table 102.4 for more details on the metabolic disorders associated
with neonatal hypoglycemia.
**
Refer to Table 102.5 for more details on the differential diagnosis of neonatal
and infantile hyperammonemia.
Table 102.4
Table 102.6
Modified from Zinn AB: Inborn errors of metabolism. In Fanaroff & Martin's neonatal-perinatal
medicine: diseases of the fetus and infant, ed 10, vol 2, Philadelphia, 2015, Elsevier (Table 99.5, p
1579).
Table 102.7
Table 102.8
Table 102.9
Modified and adapted from El-Hattab AW: Inborn errors of metabolism, Clin
Perinatol 42:413-439, 2015 (Box 6, p 417).
Table 102.10
Table 102.11
Table 102.12
Hyperammonemia
Metabolic acidosis
Lactic acidosis
Ketosis
Hypoglycemia
Liver dysfunction
Pancytopenia
When blood ammonia, pH, and bicarbonate values are normal, other
aminoacidopathies (e.g. hyperglycinemia) or galactosemia should be considered.
Galactosemic infants may also manifest cataracts, hepatomegaly, ascites, and
jaundice.
Treatment
The majority of patients with genetic disorders of metabolism respond to one or
more of the following treatments:
Albinism
See also Chapters 640 and 672 .
Albinism is caused by deficiency of melanin , the main pigment of the skin
and eye (Table 103.1 ). Melanin is synthesized by melanocytes from tyrosine in a
membrane-bound intracellular organelle, the melanosome. Melanocytes
originate from the embryonic neural crest and migrate to the skin, eyes (choroid
and iris), hair follicles, and inner ear. The melanin in the eye is confined to the
iris stromal and retinal pigment epithelia, whereas in skin and hair follicles, it is
secreted into the epidermis and hair shaft. Albinism can be caused by
deficiencies of melanin synthesis, by some hereditary defects of melanosomes,
or by disorders of melanocyte migration. Neither the biosynthetic pathway of
melanin nor many facets of melanocyte cell biology are completely elucidated
(see Fig. 103.2 ). The end products are 2 pigments: pheomelanin, which is a
yellow-red pigment, and eumelanin, a brown-black pigment.
Table 103.1
Classification of Major Causes of Albinism
variants.
† Includes brown OCA.
‡ Tyrosinase-related protein 1.
3-Methylglutaconic Acidurias
The 3-methylglutaconic acidurias are a heterogeneous group of metabolic
disorders characterized by excessive excretion of 3-methylglutaconic acid in the
urine (Table 103.2 ). Other metabolites found in 3-methylglutaconic aciduria
patients may include 3-methylglutaric acid and 3-hydroxyisovaleric acid.
Current classification distinguishes primary and secondary forms. Primary 3-
methylglutaconic aciduria is caused by the deficiency of mitochondrial 3-
methylglutaconyl-CoA hydratase (see Fig. 103.4 ), formerly 3-methylglutaconic
aciduria type I . Secondary 3-methylglutaconic aciduria can be further classified
based on the underlying mechanism (e.g., defective phospholipid remodeling vs
dysfunction of mitochondrial membrane) or the known molecular cause. Known
secondary 3-methylglutaconic aciduria includes TAZ -related syndrome (Barth
syndrome ), OPA3 -related 3-methylglutaconic aciduria (Costeff syndrome ),
SERAC1 -related syndrome (MEGDEL syndrome ), TMEM70 -related
syndrome, and DNAJC19 -related syndrome (DCMA syndrome ).
Table 103.2
3-Methylglutaconic Acidurias
Table 103.3
Genetic Disorders of Neurotransmitters in Children
Clinical Manifestations of
Hyperammonemia
In the neonatal period , symptoms and signs are mostly related to brain
dysfunction and are similar regardless of the cause of the hyperammonemia. The
affected infant appears normal at birth but becomes symptomatic following the
introduction of dietary protein. Refusal to eat, vomiting, tachypnea, and lethargy
can quickly progress to a deep coma. Seizures are common. Physical
examination may reveal hepatomegaly in addition to obtundation.
Hyperammonemia can trigger increased intracranial pressure that may be
manifested by a bulging fontanelle and dilated pupils.
Treatment of Acute Hyperammonemia
Clinical outcome depends mainly on the severity and the duration of
hyperammonemia. Serious neurologic sequelae are likely in newborns with
severe elevations in blood ammonia (>300 µmol/L) for more than 12 hr. Thus,
acute hyperammonemia should be treated promptly and vigorously. The goal of
therapy is to lower the concentration of ammonia. This is accomplished by (1)
removal of ammonia from the body in a form other than urea and (2) minimizing
endogenous protein breakdown and favoring endogenous protein synthesis by
providing adequate calories and essential amino acids (Table 103.5 ). Fluid,
electrolytes, glucose (10–15%), and lipids (1-2 g/kg/24 hr) should be infused
intravenously, together with minimal amounts of protein (0.25 g/kg/24 hr),
preferably including essential amino acids. Oral feeding with a low-protein
formula (0.5-1.0 g/kg/24 hr) through a nasogastric tube should be started as soon
as sufficient improvement is seen.
Table 103.5
Treatment of Acute Hyperammonemia in an
Infant
requirement.
‡ The higher dose is recommended in the treatment of patients with citrullinemia
Because the kidneys clear ammonia poorly, its removal from the body must be
expedited by formation of compounds with a high renal clearance. An important
advance in the treatment of hyperammonemia has been the introduction of
acylation therapy by using an exogenous organic acid that is acylated
endogenously with nonessential amino acids to form a nontoxic compound with
high renal clearance. The main organic acids used for this purpose are sodium
salts of benzoic acid and phenylacetic acid. Benzoate forms hippurate with
endogenous glycine in the liver (see Fig. 103.12 ). Each mole of benzoate
removes 1 mole of ammonia as glycine. Phenylacetate conjugates with
glutamine to form phenylacetylglutamine, which is readily excreted in the urine.
One mole of phenylacetate removes 2 moles of ammonia as glutamine from the
body (see Fig. 103.12 ). Sodium phenylbutyrate, metabolized to phenylacetate, is
the primary oral formulation. For intravenous (IV) use, a combined formulation
of benzoate and phenylacetate (Ammonul) is commercially available.
Another valuable therapeutic adjunct is IV infusion of arginine , which is
effective in all patients (except those with arginase deficiency). Arginine
administration supplies the urea cycle with ornithine (see Fig. 103.12 ). In
patients with citrullinemia, 1 mole of arginine reacts with 1 mole of ammonia (as
carbamoyl phosphate) to form citrulline. In patients with argininosuccinic
acidemia, 2 moles of ammonia (as carbamoyl phosphate and aspartate) react
with arginine to form argininosuccinic acid. Citrulline and argininosuccinate are
less toxic than ammonia and more readily excreted by the kidneys. In patients
with CPS1 or OTC deficiencies arginine administration is indicated because this
amino acid is not produced in sufficient amounts to enable endogenous protein
synthesis. For enteral therapy, patients with OTC deficiency benefit from
during periods of reduced caloric intake caused by gastrointestinal illness or
increased energy expenditure during febrile illness. Under these conditions, the
body switches from using predominantly carbohydrate to predominantly fat as
its major fuel. Fatty acids are also important fuels for exercising skeletal muscle
and are the preferred substrate for normal cardiac metabolism. In these tissues,
fatty acids are completely oxidized to carbon dioxide and water. The end
products of hepatic fatty acid oxidation are the ketone bodies β-hydroxybutyrate
and acetoacetate. These cannot be oxidized by the liver but are exported to serve
as important fuels in peripheral tissues, particularly the brain, where ketone
bodies can partially substitute for glucose during periods of fasting.
Genetic defects have been identified in almost all the known steps in the fatty
acid oxidation pathway; all are recessively inherited (Table 104.1 ).
Table 104.1
Mitochondrial Fatty Acid Oxidation Disorders—Clinical and
Biochemical Features
ENZYME
GENE CLINICAL PHENOTYPE LABORATORY FINDINGS
DEFICIENCY
Carnitine OCTN2 Cardiomyopathy, skeletal myopathy, liver ↓ Total and free carnitine,
transporter SLC22A5 disease, sudden death, endocardial normal acylcarnitines,
fibroelastosis, prenatal and newborn acylglycine, and organic acids
screening diagnosis reported
Long-chain fatty FATP1-6 Rare, acute liver failure in childhood ↓ intracellular C14 -C18 fatty
acid transporter requiring liver transplantation acids, ↓ fatty acid oxidation
Carnitine CPT-IA Liver failure, renal tubulopathy, and sudden Normal or ↑ free carnitine,
palmitoyl death. Prenatal and newborn screening normal acylcarnitines,
transferase-I diagnosis reported, maternal preeclampsia, acylglycine, and organic acids
HELLP syndrome association described in a
few patients.
Carnitine CACT Chronic progressive liver failure, persistent ↑ Normal or ↓ free carnitine,
acylcarnitine SLC25A20 NH3 , hypertrophic cardiomyopathy. abnormal acylcarnitine profile
translocase Newborn screening diagnosis reported.
Carnitine CPT-II Early and late onset types. Liver failure, Normal or ↓ free carnitine,
palmitoyl encephalopathy, skeletal myopathy, abnormal acylcarnitine profile
transferase-II cardiomyopathy, renal cystic changes,
newborn screening diagnosis reported. Adult
form with acute rhabdomyolysis,
myoglobinuria.
Short-chain acyl- SCAD Clinical phenotype unclear. Many individuals Normal or ↓ free carnitine,
CoA ACADS appear to be normal. Others have a variety of elevated urine ethylmalonic
dehydrogenase inconsistent signs and symptoms. Subset may acid, inconsistently abnormal
have severe manifestations of unclear acylcarnitine profile
relationship to biochemical defects. Newborn
screening diagnosis reported; significance
being questioned.
Medium-chain MCAD Hypoglycemia, hepatic encephalopathy, Normal or ↓ free carnitine, ↑
acyl-CoA ACADM sudden death. Newborn screening diagnosis plasma acylglycine, plasma C6
dehydrogenase possible, maternal preeclampsia, HELLP -C10 free fatty acids, ↑ C8 -C10
syndrome association described rarely, acyl-carnitine
possible long Qt interval.
Very long-chain VLCAD Dilated cardiomyopathy, arrhythmias, Normal or ↓ free carnitine, ↑
acyl-CoA ACADVL hypoglycemia, and hepatic steatosis. Late- plasma C14:1 , C14
dehydrogenase onset, stress-induced rhabdomyolysis, acylcarnitine, ↑ plasma C10 -
episodic myopathy. Prenatal and newborn C16 free fatty acids
screening diagnosis possible.
ETF ETF-DH Nonketotic fasting hypoglycemia, congenital Normal or ↓ free carnitine,
dehydrogenase* anomalies, milder forms of liver disease, increased ratio of acyl:free
cardiomyopathy, and skeletal myopathy. carnitine, ↑ acylcarnitine,
Newborn screening diagnosis reported. urine organic acid and
acylglycines
ETF-α* α-ETF Nonketotic fasting hypoglycemia, congenital Normal or ↓ free carnitine,
anomalies, liver disease, cardiomyopathy, increased ratio of acyl:free
and skeletal myopathy also described. carnitine, ↑ acylcarnitine,
Newborn screening diagnosis reported. urine organic acid and
acylglycines
ETF-β* β-ETF Fasting hypoglycemia, congenital anomalies, Normal or ↓ free carnitine,
liver disease, cardiomyopathy, and skeletal increased ratio of acyl:free
myopathy also described. Newborn screening carnitine, ↑ acylcarnitine,
diagnosis reported. urine organic acid and
acylglycines
Short-chain L -3- SCHAD Hyperinsulinemic hypoglycemia, Normal or ↓ free carnitine,
hydroxyacyl- HAD1 cardiomyopathy, myopathy. Newborn elevated free fatty acids,
CoA screening diagnosis reported. inconsistently abnormal urine
dehydrogenase organic acid, ↑ 3-OH glutarate,
↑ plasma C4 -OH acylcarnitine
Long-chain L -3- LCHAD Newborn screening diagnosis reported, Normal or ↓ free carnitine,
hydroxyacyl- HADH-A maternal preeclampsia, HELLP syndrome, increased ratio of acyl:free
CoA and AFLP association described frequently. carnitine, ↑ free fatty acids, ↑
dehydrogenase See also MTP below for clinical C16 -OH and C18 -OH
manifestations. carnitines
MTP HADH-A, Severe cardiac and skeletal myopathy, Normal or ↓ free carnitine,
HADH-B hypoglycemia, acidosis, hyper NH3 , sudden increased ratio of acyl:free
death, elevated liver enzymes, retinopathy. carnitine, ↑ free fatty acids, ↑
Maternal preeclampsia, HELLP syndrome, C16 -OH and C18 -OH
and AFLP association described frequently. carnitines
Long-chain 3- LKAT Severe neonatal presentation, hypoglycemia, Normal or ↓ free carnitine,
ketoacyl-CoA HADH-B acidosis, ↑ creatine kinase, cardiomyopathy, increased ratio of acyl:free
thiolase neuropathy, and early death carnitine, ↑ free fatty acids, ↑
2-trans , 4-cis -
decadienoylcarnitine
Short-chain 2,3- ECHS1 Leigh disease, lactic acidosis, seizures, cystic Abnormal organic acids, 2-
enoyl-CoA degeneration of white matter, microcephaly, methacrylglycine, 2-methyl-
hydratase metabolic acidosis, extrapyramidal dystonia, 2,3 dihydroxybutyrate, also S-
dilated cardiomyopathy (2-carboxypropyl)cysteine, S-
(2-carboxyethyl) cysteamine.
Acylcarnitine shows ↑ C4OH
(inconsistently).
2,4-Dienoyl-CoA DECR1 Only 1 patient described, hypotonia in the Normal or ↓ free carnitine, ↑
reductase newborn, mainly severe skeletal myopathy acyl:free carnitine ratio,
and respiratory failure. Hypoglycemia rare. normal urine organic acids and
acylglycines
HMG CoA HMGCS2 Hypoketosis and hypoglycemia, rarely ↑ total plasma fatty acids,
synthetase myopathy enzyme studies in biopsied
liver may be diagnostic,
genetic testing preferred
HMG CoA lyase HMGCL Hypoketosis and hypoglycemia, rarely Normal free carnitine, ↑ C5 -
myopathy OH, and methylglutaryl-
carnitine, enzymes studies in
fibroblasts may be diagnostic
Monocarboxylate SLC16A1 Severe fasting induced ketoacidosis, rarely Profound ketoacidosis; no
transporter 1 hypoglycemia specific biomarkers yet
(MCT1) identified
*
Also known as glutaric acidemia type II or multiple acyl-CoA dehydrogenase defect (MADD).
AFLP, Acute fatty liver of pregnancy; CoA, coenzyme A; ETF, electron transport flavoprotein;
HELLP, hemolysis, elevated liver enzymes, low platelets; MTP, mitochondrial trifunctional protein;
NH3 , ammonia.
From Shekhawat PS, Matern D, Strauss AW: Fetal fatty oxidation disorders, their effect on
maternal health and neonatal outcome: impact of expanded newborn screening on their diagnosis
and management, Pediatr Res 57:78R–84R, 2005.
Table 104.2
Classification of Peroxisomal Disorders
Epidemiology
Except for X-linked adrenoleukodystrophy (ALD) , all the peroxisomal
disorders listed in Table 104.2 are autosomal recessive diseases . ALD is the
most common peroxisomal disorder, with an estimated incidence of 1 in 17,000
live births. The combined incidence of the other peroxisomal disorders is
estimated to be 1 in 50,000 live births, although with broader newborn screening
it is expected that the actual incidences of all of the disorders of very-long-chain
fatty acids will be more accurately established.
Pathology
Absence or reduction in the number of peroxisomes is pathognomonic for
disorders of peroxisome biogenesis. In most disorders, membranous sacs contain
peroxisomal integral membrane proteins, which lack the normal complement of
matrix proteins; these are peroxisome “ghosts.” Pathologic changes are observed
in most organs and include profound and characteristic defects in neuronal
migration, micronodular cirrhosis of the liver, renal cysts, chondrodysplasia
punctata, sensorineural hearing loss, retinopathy, congenital heart disease, and
dysmorphic features.
Pathogenesis
All pathologic changes likely are secondary to the peroxisome defect. Multiple
peroxisomal enzymes fail to function in the PBDs (Table 104.3 ). The enzymes
that are diminished or absent are synthesized but are degraded abnormally fast
because they may be unprotected outside the peroxisome. It is not clear how
defective peroxisome functions lead to the widespread pathologic
manifestations.
Table 104.3
Abnormal Laboratory Findings Common to
Zellweger Spectrum Disorders
Peroxisomes absent to reduced in number
Catalase in cytosol
Deficient synthesis and reduced tissue levels of plasmalogens
Defective oxidation and abnormal accumulation of very-long-chain fatty
acids
Deficient oxidation and age-dependent accumulation of phytanic acid
Defects in certain steps of bile acid formation and accumulation of bile
acid intermediates
Defects in oxidation and accumulation of L -pipecolic acid
Increased urinary excretion of dicarboxylic acids
Table 104.4
Main Clinical Abnormalities in Zellweger Syndrome
Patients with neonatal ALD show fewer, less prominent craniofacial features.
Neonatal seizures occur frequently. Some degree of psychomotor developmental
delay is present; function remains in the range of severe intellectual disability,
and development may regress after 3-5 yr of age, probably from a progressive
leukodystrophy. Hepatomegaly, impaired liver function, pigmentary
degeneration of the retina, and severely impaired hearing are invariably present.
Adrenocortical function is usually impaired and may require adrenal hormone
replacement. Chondrodysplasia punctata and renal cysts are absent.
Patients with infantile Refsum disease have survived to adulthood. They can
walk, although gait may be ataxic and broad based. Cognitive function is
generally impaired, but accurate assessment is limited, usually by the presence
of both vision and hearing impairment. Almost all have some degree of
sensorineural hearing loss and pigmentary degeneration of the retina. They have
moderately dysmorphic features that may include epicanthal folds, a flat nose
bridge, and low-set ears. Early hypotonia and hepatomegaly with impaired
function are common. Levels of plasma cholesterol and high-density and low-
density lipoprotein are often moderately reduced. Chondrodysplasia punctata
and renal cortical cysts are absent. Postmortem study in infantile Refsum disease
reveals micronodular liver cirrhosis and small, hypoplastic adrenals. The brain
shows no malformations, except for severe hypoplasia of the cerebellar granule
are normal, but plasma phytanic acid levels are increased and red blood cell
(RBC) plasmalogen levels are reduced. In other peroxisomal disorders, the
biochemical abnormalities are still more restricted. Therefore, a panel of tests is
recommended and includes plasma levels of VLCFAs and phytanic, pristanic,
and pipecolic acids and RBC levels of plasmalogens. Tandem mass spectrometry
techniques also permit convenient quantitation of bile acids in plasma and urine.
This panel of tests can be performed on very small amounts of venous blood and
permits detection of most peroxisomal disorders. Furthermore, normal results
make the presence of the typical peroxisomal disorder unlikely. Biochemical
findings combined with the clinical presentation are often sufficient to arrive at a
clinical diagnosis. Methods using dried blood spots of filter paper have been
developed and are being incorporated into newborn screening assays.
Table 104.5
Diagnostic Biochemical Abnormalities in Peroxisomal
Disorders
Table 104.6
Characteristics of the Major Lipoproteins
COMPOSITION
SIZE DENSITY
LIPOPROTEIN SOURCE Protein Lipid
(nm) (g/mL) Apolipoproteins
(%) (%)
Chylomicrons Intestine 80- <0.95 1-2 98-99 C-I, C-II, C-III, E, A-I, A-
1,200 II, A-IV, B-48
Chylomicron Chylomicrons 40-150 <1.0006 6-8 92-94 B-48, E
remnants
VLDL Liver, intestine 30-80 0.95-1.006 7-10 90-93 B-100, C-I, C-II, C-III
IDL VLDL 25-35 1.006-1.019 11 89 B-100, E
LDL VLDL 18-25 1.019-1.063 21 79 B-100
HDL Liver, intestine VLDL, 5-20 1.125-1.210 32-57 43-68 A-I, A-II, A-IV C-I, C-II,
chylomicrons C-III D, E
HDL, High-density lipoproteins; IDL, intermediate-density lipoproteins; LDL, low-density
lipoproteins; VLDL, very-low-density lipoproteins.
Hyperlipoproteinemias
Hypercholesterolemia
See Table 104.7 .
Table 104.7
Hyperlipoproteinemias
LIPOPROTEINS ESTIMATED
DISORDER CLINICAL FINDINGS GENETICS
ELEVATED INCIDENCE
Familial hypercholesterolemia LDL Tendon xanthomas, CHD AD 1 in 500
Familial defective ApoB-100 LDL Tendon xanthomas, CHD AD 1 in 1,000
Autosomal recessive LDL Tendon xanthomas, CHD AR <1 in
hypercholesterolemia 1,000,000
Sitosterolemia LDL Tendon xanthomas, CHD AR <1 in
1,000,000
Polygenic LDL CHD 1 in 30?
hypercholesterolemia
Familial combined LDL, TG CHD AD 1 in 200
hyperlipidemia
Familial LDL, TG Tuberoeruptive xanthomas, AD 1 in 10,000
dysbetalipoproteinemia peripheral vascular disease
Familial chylomicronemia TG ↑↑ Eruptive xanthomas, AR 1 in 1,000,000
(Frederickson type I) hepatosplenomegaly,
pancreatitis
Familial hypertriglyceridemia TG ↑ ±CHD AD 1 in 500
(Frederickson type IV)
Familial hypertriglyceridemia TG ↑↑ Xanthomas ± CHD AD —
(Frederickson type V)
Familial hepatic lipase VLDL CHD AR <1 in
deficiency 1,000,000
AD, Autosomal dominant; AR, autosomal recessive; CHD, coronary heart disease; LDL, low-
density lipoproteins, TG, triglycerides; VLDL, very-low-density lipoproteins.
Familial Hypercholesterolemia
Familial hypercholesterolemia (FH) is a monogenic autosomal co-dominant
disorder characterized by strikingly elevated LDL cholesterol (LDL-C),
premature cardiovascular disease (CVD), and tendon xanthomas. In the past, FH
referred to defects of LDL-R activity. The etiology of this lipoprotein
abnormality also includes defects in the genes for apoB (as well as PCSK-9). Of
the almost 1200 mutations described, some result in failure of synthesis of the
LDL-R (receptor negative), and others cause defective binding or release at the
lipoprotein-receptor interface. Receptor-negative mutations result in more severe
phenotypes than receptor-defective mutations.
Table 104.8
Percentage of Youths Younger than Age 18 Yr Expected to
Have Familial Hypercholesterolemia (FH) According to
Cholesterol Levels and Closest Relative With FH
PERCENTAGE WITH FH AT THAT LEVEL
TOTAL CHOL (mg/dL) LDL CHOL (mg/dL) Degree of Relative
General Population
First Second Third
180 122 7.2 2.4 0.9 0.01
190 130 13.5 5.0 2.2 0.03
200 138 26.4 10.7 4.9 0.07
210 147 48.1 23.6 11.7 0.19
220 155 73.1 47.5 27.9 0.54
230 164 90.0 75.0 56.2 1.8
240 172 97.1 93.7 82.8 6.3
250 181 99.3 97.6 95.3 22.2
260 190 99.9 99.5 99.0 57.6
270 200 100.0 99.9 99.8 88.0
280 210 100.0 100.0 100.0 97.8
290 220 100.0 100.0 100.0 99.6
300 230 100.0 100.0 100.0 99.9
310 240 100.0 100.0 100.0 100.0
Chol, Cholesterol; LDL, low-density lipoprotein.
From Williams RR, Hunt SC, Schumacher MC, et al: Diagnosing heterozygous familial
hypercholesterolemia using new practical criteria validated by molecular genetics, Am J Cardiol
72:171–176, 1993.
Table 104.9
Plasma Cholesterol and Triglyceride Levels in Childhood
and Adolescence: Means and Percentiles
Hypothyroidism
Nephrotic syndrome
Cholestasis
Anorexia nervosa
Drugs: progesterone, thiazides, carbamazepine (Tegretol), cyclosporine
Hypertriglyceridemia
Obesity
Type 2 diabetes
Alcohol
Renal failure
Sepsis
Stress
Cushing syndrome
Pregnancy
Hepatitis
AIDS, protease inhibitors
Drugs: anabolic steroids, β-blockers, estrogen, thiazides
Smoking
Obesity
Type 2 diabetes
Malnutrition
Drugs: β-blockers, anabolic steroids
Smith-Lemli-Opitz Syndrome
Patients with Smith-Lemli-Opitz syndrome (SLOS ) often have multiple
congenital anomalies and developmental delay caused by low plasma cholesterol
and accumulated precursors (Tables 104.11 and 104.12 ) (see Chapter 606.2 ).
Family pedigree analysis has revealed its autosomal recessive inheritance
pattern. Mutations in the DHCR7 (7-dehydrocholesterol-Δ7 reductase) gene
result in deficiency of the microsomal enzyme DHCR7, which is necessary to
complete the final step in cholesterol synthesis. It is not known why defects in
cholesterol synthesis result in congenital malformations, but since cholesterol is
a major component of myelin and a contributor to signal transduction in the
developing nervous system, neurodevelopment is severely impaired. The
incidence of SLOS is estimated to be 1 in 20,000-60,000 births among whites,
with a somewhat higher frequency in Hispanics and lower incidence in
individuals of African descent.
Table 104.11
Major Clinical Characteristics of Smith-
Lemli-Opitz Syndrome: Frequent Anomalies
(>50% of Patients)
Craniofacial
Microcephaly
Blepharoptosis
Anteverted nares
Retromicrognathia
Low-set, posteriorly rotated ears
Midline cleft palate
Broad maxillary alveolar ridges
Cataracts (<50%)
Skeletal Anomalies
Genital Anomalies
Hypospadias
Cryptorchidism
Sexual ambiguity (<50%)
Development
Cardiovascular
Atrioventricular canal
Secundum atrial septal defect
Patent ductus arteriosus
Membranous ventricular septal defect
Urinary Tract
Gastrointestinal
Hirschsprung disease
Pyloric stenosis
Refractory dysmotility
Cholestatic and noncholestatic progressive liver disease
Pulmonary
Pulmonary hypoplasia
Abnormal lobation
Endocrine
Adrenal insufficiency
Spontaneous abortion of SLOS fetuses may occur. Type II SLOS often leads
to death by the end of the neonatal period. Survival is unlikely when the plasma
cholesterol level is <20 mg/dL. Laboratory measurement should be performed
by gas chromatography, because standard techniques for lipoprotein assay
include measurement of cholesterol precursors, which may yield a false-positive
result. Milder cases may not present until late childhood. Phenotypic variance
ranges from microcephaly, cardiac and brain malformation, and multiorgan
system failure to only subtle dysmorphic features and mild developmental delay.
Treatment includes supplemental dietary cholesterol (egg yolk) and HMG-CoA
reductase inhibition to prevent the synthesis of toxic precursors proximal to the
enzymatic block.
Pharmacologic Therapy.
See Tables 104.13 and 104.14 .
Table 104.13
Drugs Used for the Treatment of Hyperlipidemia
Table 104.14
Adverse Effects of Cholesterol-Lowering
Drugs
Statins
Ezetimibe
Pcsk9 Inhibitors
Niacin
Skin flushing, pruritus, GI disturbances, blurred vision, fatigue, glucose
intolerance, hyperuricemia, hepatic toxicity, exacerbation of peptic ulcers
Adverse effects, especially flushing, occur more frequently with
immediate-release products.
Rare: Dry eyes, hyperpigmentation
Fish Oil
From The Medical Letter: Lipid-lowering drugs, Med Lett 58:133-140, 2016
(Table 2, p 136).
The lysosomal lipid storage diseases are diverse disorders, each caused by an
inherited deficiency of a specific lysosomal hydrolase leading to the
intralysosomal accumulation of the enzyme's particular substrate (Tables 104.15
and 104.16 ). Except for Wolman disease and cholesterol ester storage disease,
the lipid substrates share a common structure that includes a ceramide backbone
(2-N -acylsphingosine) from which the various sphingolipids are derived by
substitution of hexoses, phosphorylcholine, or 1 or more sialic acid residues on
the terminal hydroxyl group of the ceramide molecule. The pathway of
sphingolipid metabolism in nervous tissue (Fig. 104.15 ) and in visceral organs
(Fig. 104.16 ) is known; each catabolic step, with the exception of the catabolism
of lactosylceramide, has a genetically determined metabolic defect and a
resultant disease. Because sphingolipids are essential components of all cell
membranes, the inability to degrade these substances and their subsequent
accumulation results in the physiologic and morphologic alterations and
characteristic clinical manifestations of the lipid storage disorders (Table 104.15
). Progressive lysosomal accumulation of glycosphingolipids in the CNS leads to
neurodegeneration, whereas storage in visceral cells can lead to organomegaly,
skeletal abnormalities, pulmonary infiltration, and other manifestations. The
storage of a substrate in a specific tissue depends on its normal distribution in the
body.
Table 104.15
Clinical Findings in Lysosomal Storage Diseases
COARSE
FACIAL
HYDROPS
NOMENCLATURE ENZYME DEFECT FEATURES HEPATOSPLENOMEGALY
FETALIS
DYSOSTOSIS
MULTIPLEX
MUCOLIPIDOSES
Mucolipidoses II, I-cell N - (+) ++ +
disease Acetylglucosaminylphosphotransferase
Mucolipidosis III, N - − + (+)
pseudo-Hurler Acetylglucosaminylphosphotransferase
Mucolipidosis IV Unknown − − +
SPHINGOLIPIDOSES
Fabry disease α-Galactosidase − − −
Farber disease Acid ceramidase − − (+)
Galactosialidosis β-Galactosidase and sialidase (+) ++ ++
GM1 gangliosidosis β-Galactosidase (+) ++ +
GM2 gangliosidosis β-Hexosaminidases A and B − − (+)
(Tay-Sachs, Sandhoff
disease)
Gaucher type I Glucocerebrosidase − − ++
Gaucher type II Glucocerebrosidase (+) − ++
Gaucher type III Glucocerebrosidase (+) − +
Niemann-Pick type A Acid Sphingomyelinase (+) − ++
Niemann-Pick type B Acid Sphingomyelinase − − ++
Metachromatic Arylsulfatase A − − −
leukodystrophy
Krabbe disease β-Galactocerebrosidase − − −
LIPID STORAGE DISORDERS
Niemann-Pick type C Intracellular cholesterol transport − − (+)
Wolman disease Lysosomal acid lipase (+) − +
Ceroid lipofuscinosis, Palmitoyl-protein thioesterase (CLN1) − − −
infantile (Santavuori-
Haltia)
Ceroid lipofuscinosis, Pepstatin-insensitive peptidase − − −
late infantile (Jansky- (CLN2); variants in Finland (CLN5),
Bielschowsky) Turkey (CLN7), and Italy (CLN6)
Ceroid lipofuscinosis, CLN3, membrane protein − − −
juvenile (Spielmeyer-
Vogt)
Ceroid lipofuscinosis, CLN4, probably heterogeneous (+) − −
adult (Kufs, Parry)
OLIGOSACCHARIDOSES
Aspartylglucosaminuria Aspartylglucosylaminase − + (+)
Fucosidosis α-Fucosidase − ++ (+)
α-Mannosidosis α-Mannosidase − ++ +
β-Mannosidosis β-Mannosidase − + (+)
Schindler disease α-N -Acetylgalactosaminidase − − −
Sialidosis I Sialidase (+) − −
Sialidosis II Sialidase (+) ++ +
++, Prominent; +, often present; (+), inconstant or occurring later in the disease course; –, not
present.
Modified from Hoffmann GF, Nyhan WL, Zschoke J, et al: Storage disorders in inherited metabolic
diseases , Philadelphia, 2002, Lippincott Williams & Wilkins, pp 346–351.
Table 104.16
Lysosomal Storage Disorders in the Newborn Period:
Genetic and Clinical Characteristics of Neonatal
Presentation
Gaucher disease type 2 In utero– Normal Poor suck and Congenital – Glucocerebrosidase
6 months swallow, weak ichthyosis, deficiency
cry, squint, collodion skin
trismus,
strabismus,
opsoclonus,
hypertonic, later
flaccidity
Krabbe disease 3–6 Normal Irritability, tonic Increased CSF Optic Galactocerebrosidase
months spasms with light protein level atrophy deficiency
or noise
stimulation,
seizures,
hypertonia, later
flaccidity
GM1 gangliosidosis Birth Coarse Poor suck, weak Gingival Cherry-red β-Galactosidase
cry, lethargy, hypertrophy, spot (50%) deficiency
exaggerated edema, rashes
startle, blindness,
hypotonia, later
spasticity
Farber disease type I 2 weeks– Normal Progressive Joint swelling Grayish Lysosomal acid
4 months psychomotor with nodules, opacification ceramidase
impairment, hoarseness, lung surrounding
seizures, disease, retina in
decreased contractures, some
reflexes, fever, patients,
hypotonia granulomas, subtle
dysphagia, cherry-red
vomiting, spot
increased CSF
protein level
Table 105.1
Features of the Disorders of Carbohydrate Metabolism
105.1
Glycogen Storage Diseases
Priya S. Kishnani, Yuan-Tsong Chen
Keywords
glycogen
liver glycogen
hepatomegaly
myopathy
hypoglycemia
ketosis
enzyme replacement therapy
Table 105.2
Causes of Type B Lactic Acidosis
Type B1—Underlying Diseases
Renal failure
Hepatic failure
Diabetes mellitus
Malignancy
Systemic inflammatory response syndrome
Human immunodeficiency virus
Acetaminophen
Alcohols—ethanol, methanol, diethylene glycol, isopropanol, and
propylene glycol
Antiretroviral nucleoside analogs—zidovudine, didanosine, and lamivudine
β-Adrenergic agonists—epinephrine, ritodrine, and terbutaline
Biguanides—phenformin and metformin
Cocaine, methamphetamine
Cyanogenic compounds—cyanide, aliphatic nitriles, and nitroprusside
Diethyl ether
Fluorouracil
Halothane
Iron
Isoniazid
Linezolid
Nalidixic acid
Niacin
Propopol
Salicylates
Strychnine
Sugars and sugar alcohols—fructose, sorbitol, and xylitol
Sulfasalazine
Total parenteral nutrition
Valproic acid
Vitamin deficiencies—thiamine and biotin
Table 105.4
Mitochondrial Disease Criteria (Simplified Version for
Bedside Use)*
I. CLINICAL SIGNS AND SYMPTOMS, 1
POINT/SYMPTOM (max. 4 points)
A. Muscular B. CNS C. Multisystem
II. Metabolic/Imaging III. Morphology
Presentation (max. 2 Presentation (max. Disease (max. 3
Studies (max. 4 points) (max. 4 points)
points) 2 points) points)
Ophthalmoplegia † Developmental Hematology Elevated lactate † Ragged red/blue
delay fibers ‡
Facies myopathica Loss of skills GI tract Elevated L/P ratio COX-negative
fibers ‡
Exercise intolerance Stroke-like episode Endocrine/growth Elevated alanine † Reduced COX
staining ‡
Muscle weakness Migraine Heart Elevated CSF lactate † Reduced SDH
staining
Rhabdomyolysis Seizures Kidney Elevated CSF protein SDH positive
blood vessels †
Abnormal EMG Myoclonus Vision Elevated CSF alanine † Abnormal
mitochondria/EM
†
Cortical blindness Hearing Urinary TA excretion †
Pyramidal signs Neuropathy Ethylmalonic aciduria
Extrapyramidal Recurrent/familial Stroke-like picture/MRI
signs
Brainstem Leigh syndrome/MRI †
involvement
Elevated lactate/MRS
* Score 1: mitochondrial disorder unlikely; score 2 to 4: possible mitochondrial disorder; score 5 to
‡
This symptom in a higher percentage scores 4 points.
GI, gastrointestinal; L/P, lactate/pyruvate; COX, cytochrome C oxidase; SDH, succinate
dehydrogenase; EM, electron microscopy; EMG, electromyography; TA, tricarbon acid.
From Morava E, van den Heuvel L, Hol F, et al: Mitochondrial disease criteria – diagnostic
applications in children. Neurology 67:1823-1826, 2006, p 1824.
Table 105.5
Clues to the Diagnosis of Mitochondrial
Disease
Neurologic
Cardiovascular
Ophthalmologic
Gastroenterologic
Other
From Haas RH, Parikh S, Falk MJ, et al: Mitochondrial disease: a practical
approach for primary care physicians, Pediatrics 120:1326–1333, 2007 (Table 1,
p 1327).
Table 105.6
Clinical and Laboratory Features in Common Congenital
Disorders of Glycosylation (CDGs), with Clinically
Recognizable Phenotype and Abnormal Glycosylation,
Detectable by Serum Transferrin Isoform Analysis (TIEF)
OTHER
DEFECTIVE MOST FREQUENT SUGGESTIVE LABORATORY
BIOCHEMICAL
GENE CLINICAL FEATURES FEATURES ABNORMALITIES
ANOMALIES
PMM2 Strabismus, nystagmus, Inverted nipples Elevated serum Type 1 serum
smooth philtrum, large ears, and/or abnormal fat transaminases, TIEF, decreased
vomiting, diarrhea, FTT, axial pads, stroke-like hypoalbuminemia, PMM activity in
hypotonia, cerebellar vermis episodes decreased factor IX, leukocytes and
hypoplasia, ataxia, XI and AT activity, fibroblasts
psychomotor disability, low serum
seizures, spasticity, ceruloplasmin and
neuropathy, pigmentary TBG levels
retinitis
PMI Cholestasis, hepatomegaly, Hyperinsulinism, Elevated Type 1 serum
feeding difficulties, recurrent protein losing transaminases, TIEF, decreased
vomiting, chronic diarrhea, enteropathy hypoalbuminemia, PMI activity in
ascites, recurrent thrombosis, Normal hypoglycemia, leukocytes and
gastrointestinal bleeding intelligence and decreased factor IX, fibroblasts
absence of XI, and AT-III
neurologic activity
features
ALG6 Hypotonia, muscle weakness, (Distal limb Elevated serum Type 1 serum
seizures, ataxia, intellectual malformations) transaminases; TIEF, abnormal
disability, behavioral hypoalbuminemia; LLO results in
abnormalities decreased factor IX, fibroblasts
XI, and AT activity;
low serum IgG level
DPAGT1 Microcephaly, brain Congenital Decreased AT, Type 1 serum
malformations, hypotonia, myasthenia protein C, and protein TIEF
severe psychomotor disability, phenotype S activity; increased
seizures, spasticity, proximal In multisystem creatine kinase;
weakness, failure to thrive, phenotype: hypoalbuminemia;
joint contractures cataract normal creatine
kinase in myasthenia
SRD5A3 Developmental delay, Congenital cataract, Low anticoagulation Type 1 serum
hypotonia, ataxia, cerebellar retinal and iridic factors (AT, protein TIEF but reported
vermis hypoplasia, intellectual coloboma, glaucoma, C, and protein S false-negative
disability, speech delay, visual optic nerve dysplasia, activity), increased TIEF
loss ichthyosis serum transaminases
ATP6V0A2 Generalized cutis laxa, Cobblestone-like Mild coagulation Type 2 serum
hypotonia, strabismus, brain dysgenesis abnormalities, TIEF but
characteristic facial features, increased serum reported
joint laxity, seizures, motor transaminase levels false-negative
ATP6V1A and and language developmental Cardiovascular Mild coagulation TIEF
ATP6V1E1 delay, spontaneous anomalies abnormalities and Abnormal
improvement of cutis laxa by increased serum apoC-III IEF,
aging transaminase levels, characteristic
hypercholesterolemia MALDI TOF
profile
(Note
abnormal
skin
histology)
PGM1 Pierre Robin sequence, Cleft palate, Hypoglycemia, Mixed type 1/ 2
cholestasis, short stature, hyperinsulinism, increased serum serum TIEF,
dilated cardiomyopathy, normal intelligence transaminase levels, decreased
decreased AT fibroblast PGM1
activity
MAN1B1 Developmental delay, speech Obesity, autistic Increased serum Type 2 serum
delay, intellectual disability, features, inverted transaminase levels, TIEF, abnormal
muscle weakness nipples, characteristic low AT apoC-III IEF,
face diagnostic
MALDI TOF
profile
TMEM199 Cholestasis, hepatomegaly, Normal intelligence Decreased serum Type 2 serum
CCDC115 liver steatosis, liver fibrosis, Hepatomegaly ceruloplasmin, TIEF, abnormal
ATP6AP1 and liver failure, spontaneous Immune deficiency increased serum apoC-III IEF,
ATP6AP2 bleedings, motor transaminase levels, characteristic
developmental delay hypercholesterolemia, MALDI TOF
high AP profile
SLC39A8 Seizures, hypsarrhythmia, Dwarfism, Decreased serum Type 2 serum
hypotonia, developmental and craniosynostosis, manganese, high TIEF, abnormal
speech delay, FTT rhizomelia, Leigh serum transaminases, apoC-III,
disease abnormal coagulation characteristic
MALDI TOF
profile
AP, Alkaline phosphatase; AT, antithrombin; apoC-III: apolipoprotein C-III; FTT, failure to thrive;
LLO, lipid-linked oligosaccharides; MALDI-TOF, matrix-assisted laser desorption/ionization time of
flight; TBG, thyroxine-binding globulin; TIEF, transferrin isoelectric focusing.
Table 106.1
Major Molecular Categories of Mitochondrial Genes
CAUSAL DISEASE
COMPONENT GENE MUTATION EFFECTS
GENOME EXAMPLES
Electron Nuclear or Decreased functioning of electron transport chain complex Complex I
transport chain mtDNA deficiency
enzyme subunits Complex II
deficiency
Electron Nuclear Decreased assembly of electron transport chain enzyme Complex III
transport chain complex deficiency
assembly factors Complex IV
deficiency
Complex V
deficiency
Electron Nuclear Decreased functioning of electron transport chain Coenzyme Q10
transport chain deficiency
cofactors Iron sulfur
cluster defect
Lipoyltransferase
deficiency
mtDNA Nuclear or Decreased translation of protein-coding mitochondrial Combined oxidative
translation mtDNA DNA genes leading to decreased functioning of electron phosphorylation
transport chain enzymes complexes deficiency
mtDNA Nuclear Increased errors in mitochondrial DNA leading to increased Mitochondrial
maintenance presence of point mutations and deletions, resulting in DNA depletion
decreased translation of electron transport chain subunits syndromes
Mitochondrial
DNA multiple
deletion
disorders
Mitochondrial Nuclear Increased mtDNA point mutations and deletions; clumped OPA1 -related
membrane and fragmented mitochondria conditions
fission and MFN2 -related
fusion conditions
From McCormick EM, Muraresku CC, Falk MJ: Mitochondrial genomics: a complex field now
coming of age. Curr Genet Med Rep 6:52–61, 2018 (Table 1, p. 57).
Table 107.2
Mucopolysaccharidoses: Clinical, Molecular, and
Biochemical Aspects
MAIN
MPS GENE DEFECTIVE
EPONYM INHERITANCE CLINICAL ASSAY
TYPE CHROMOSOME ENZYME
FEATURES
I-H (Pfaundler-) AR IDUA Severe Hurler α-L -iduronidase L, F,
Hurler 4p16.3 phenotype, mental Ac, Cv
deficiency, corneal
clouding, death
usually before age
14 yr
I-S Scheie AR IDUA Stiff joints, α-L -iduronidase L, F,
4p16.4 corneal clouding, Ac, Cv
aortic valve
disease, normal
intelligence,
survive to
adulthood
I-HS Hurler-Scheie AR IDUA Phenotype α-L -iduronidase L, F,
4p16.4 intermediate Ac, Cv
between I-H and
I-S
II Hunter XLR IDS Severe Iduronate sulfate S, F,
Xq27.3-28 course: sulfatase Af, Ac,
similar to I-H Cv
but clear
corneas
Mild course:
less
pronounced
features, later
manifestation,
survival to
adulthood
with mild or
without
mental
deficiency
IIIA Sanfilippo A AR SGSH Behavioral Heparan-S- L, F,
17q25.3 problems, sulfamidase Ac, Cv
sleeping
IIIB Sanfilippo B AR NAGLU N -Acetyl-α-D - S, F,
disorder,
17q21 aggression, glucosaminidase Ac, Cv
IIIC Sanfilippo C AR HGSNAT progressive Acetyl-CoA- F, Ac
8p11.21 dementia, glucosaminide N -
mild acetyltransferase
IIID Sanfilippo D AR GNS dysmorphism, N - F, Ac
12q14 coarse hair, Acetylglucosamine–
clear corneas 6-sulfate sulfatase
Survival to
adulthood
possible
IVA Morquio A AR GALNS Short-trunk N - L, F,
16q24.3 dwarfism, fine Acetylgalactosamine- Ac
corneal opacities, 6-sulfate sulfatase
characteristic bone
dysplasia; final
height <125 cm
IVB Morquio B AR GLB1 Same as IVA, but β-Galactosidase L, F,
3p21.33 milder; adult Ac, Cv
height >120 cm
VI Maroteaux- AR ARSB Hurler phenotype N - L, F,
Lamy 5q11-q13 with marked Acetylgalactosamine- Ac
corneal clouding α-4-sulfate sulfatase
but normal (arylsulfatase B)
intelligence; mild,
moderate, and
severe expression
in different
families
VII Sly AR GUSB Varying from fetal β-Glucuronidase S, F,
7q21.11 hydrops to mild Ac, Cv
dysmorphism;
dense inclusions
in granulocytes
IX Hyaluronidase AR HYAL1 Periarticular Hyaluronidase 1 S
deficiency 3p21.3 masses, no Hurler
phenotype
MPSPS MPS plus AR VPS33A Mild Hurler No lysosomal
syndrome phenotype, enzyme deficiency
cognitive
deficiency,
organomegaly,
skeletal dysplasia,
pancytopenia,
renal
insufficiency,
optic atrophy,
early death
AR, Autosomal recessive; XLR, X-linked recessive; L, Leukocytes; S, serum; F, cultured
fibroblasts; Ac, cultured amniotic cells; Af, amniotic fluid; Cv, chorionic villus sampling; MIM,
Mendelian Inheritance in Man Catalogue.
Disease Entities
Mucopolysaccharidosis I
Mucopolysaccharidosis I (MPS-I) is caused by mutations of the IUA gene on
chromosome 4p16.3 encoding α-L -iduronidase. Mutation analysis has revealed 2
major alleles, W402X and Q70X, which account for more than half the MPS-I
alleles in the white population. The mutations that introduce stop codons with
ensuing absence of functional enzyme (null alleles), and in homozygosity or
compound heterozygosity, give rise to Hurler syndrome. Other mutations occur
in only one or a few individuals.
Deficiency of α-L -iduronidase results in a wide range of clinical involvement,
from severe Hurler syndrome to mild Scheie syndrome, which are ends of a
broad clinical spectrum. Homozygous nonsense mutations result in severe forms
of MPS-I, whereas missense mutations are more likely to preserve some residual
enzyme activity associated with a milder form of the syndrome.
Hurler Syndrome
The Hurler form of MPS-I (MPS I-H ) is a severe, progressive disorder with
involvement of multiple organs and tissues that results in premature death,
usually by 10 yr of age. An infant with Hurler syndrome appears normal at birth,
but inguinal hernias and failed neonatal hearing tests may be early signs.
Diagnosis is usually made at 6-24 mo, with evidence of hepatosplenomegaly,
coarse facial features, corneal clouding, large tongue, enlarged head
circumference, joint stiffness, short stature, and skeletal dysplasia. Acute
cardiomyopathy has been found in some infants <1 yr. Most patients have
recurrent upper respiratory tract and ear infections, noisy breathing, and
persistent copious nasal discharge. Valvular heart disease, notably with
incompetence of the mitral and aortic valves, regularly develops, and narrowing
of the coronary arteries occurs. Obstructive airway disease, especially during
sleep, may necessitate tracheotomy. Obstructive airway disease, respiratory
infection, and cardiac complications are the common causes of death (Table
107.3 ).
Table 107.3
Corneal clouding 90
Hepatomegaly 84
Upper airway obstruction → OSA 82
Kyphosis gibbus 75
Joint contractures 72
Hernia 70
Dysostosis multiplex 70
Cognitive impairment 60
Enlarged tongue 60
Splenomegaly 60
Eustachian tube obstruction → otitis media 55
Hip dysplasia 42
Genu valgum 38
Reactive airway disease 37
Scoliosis 35
Carpal tunnel syndrome 25
Pes cavus 18
Glaucoma 10
Heart failure 3
Cor pulmonale 2
OSA, Obstructive sleep apnea.
From Clarke LA, Atherton AM, Burton BK, et al: Mucopolysaccharidosis type I newborn screening:
best practices for diagnosis and management, J Pediatr 182:363–370, 2017 (Table 1, p 364).
Most children with Hurler syndrome acquire only limited language skills
because of intellectual disability, combined conductive and neurosensory hearing
loss, and an enlarged tongue. Progressive ventricular enlargement with increased
intracranial pressure caused by communicating hydrocephalus also occurs.
Corneal clouding, glaucoma, and retinal degeneration are common. Radiographs
show a characteristic skeletal dysplasia known as dysostosis multiplex (Figs.
107.3 and 107.4 ). The earliest radiographic signs are thick ribs and ovoid
vertebral bodies. Skeletal abnormalities (in addition to those shown in the
figures) include enlarged, coarsely trabeculated diaphyses of the long bones with
irregular metaphyses and epiphyses. With disease progression, macrocephaly
develops with thickened calvarium, premature closure of lambdoid and sagittal
sutures, shallow orbits, enlarged J -shaped sella, and abnormal spacing of teeth
with dentigerous cysts.
Treatment
Hematopoietic stem cell transplantation has resulted in significant clinical
improvement of somatic disease in patients with MPS I, II, and VI (Table 107.4
). Clinical effects are increased life expectancy with resolution or improvement
of growth, hepatosplenomegaly, joint stiffness, facial appearance, skin changes,
obstructive sleep apnea, heart disease, communicating hydrocephalus, and
hearing loss. Enzyme activity in serum and urinary GAG excretion normalize.
This is true for MPS I-H, II, and III. Patients with MPS-I who have undergone
transplantation before 9 mo of age may show normal cognitive development.
Transplantation before 24 mo and with a baseline mental development index >70
have improved long-term outcome. Transplantation does not significantly
improve the neuropsychological outcome of MPS patients with impaired
cognition at transplantation. Early transplantation in the MPS-II patient may
have the same effect. Transplantation in the MPS-VI patient stabilizes or
improves cardiac manifestations, posture, and joint mobility. Stem cell
transplantation does not correct skeletal or ocular anomalies.
Table 107.4
Therapies Aimed at Proximate Causes of
Mucopolysaccharidoses
HEMATOPOIETIC ENZYME
MPS
STEM CELL REPLACEMENT REMARKS
TYPE
TRANSPLANTATION THERAPY
I Yes Laronidase Developmental trajectory dependent on time of
(Aldurazyme) transplantation. Little effect on connective tissue
II Yes Idursulfase manifestations. Enzyme replacement immediately after
(Elaprase) diagnosis.
III No No Experimental intrathecal application of recombinant
heparin-N -sulfatase in MPS-IIIA.
IV Yes Elosulfase Improved daily activities. No effect on growth or skeletal
(Vimizim) dysplasia.
Table 107.5
Symptomatic Management of Mucopolysaccharidoses
PREDOMINANTLY
PROBLEM MANAGEMENT
IN
NEUROLOGIC
Hydrocephalus MPS I, II, VI, VII Funduscopy, CT scan
Chronic headaches All Ventriculoperitoneal shunting
Behavioral MPS-III Behavioral medication, sometimes
disturbance CT scan, ventriculoperitoneal shunting
Disturbed sleep– MPS-III Melatonin
wake cycle
Seizures MPS I, II, III EEG, anticonvulsants
Atlantoaxial MPS IV Cervical MRI, upper cervical fusion
instability
Spinal cord All Laminectomy, dural excision
compression
OPHTHALMOLOGIC
Corneal opacity MPS I, VI, VII Corneal transplant
Bibliography
Burton BK, Jego V, Mikl J, et al. Survival in idursulfase-treated
and untreated patients with mucopolysaccharidosis type II:
data from the Hunter Outcome survey (HOS). J Inherit Metab
Dis . 2017;40:867–874.
Clarke LA. Mucopolysaccharidosis type I. Pagon RA, Adam
MP, Ardinger HH, et al. Gene reviews (internet) . 2016
[Seattle].
Clarke LA, Atherton AM, Burton BK, et al.
Mucopolysaccharidosis type I newborn screening: best
practices for diagnosis and management. J Pediatr .
2017;182:363–370.
Dvorakova L, Vlaskova H, Sarjlija A, et al. Genotype-
phenotype correlation in 44 Czech, Slovak, Croatian and
Serbian patients with mucopolysaccharidosis type II. Clin
Genet . 2017;91:787–796.
FIG. 109.3 Unrelated 7 yr old female and 10 yr old male with progeria.
Appearance is remarkably similar between patients. (Photograph courtesy
of The Progeria Research Foundation)
Table 109.1
Features of Hutchinson-Gilford Progeria Syndrome and
Other Disorders With Overlapping Features
HUTCHINSON-
WIEDEMANN- ROTHMUND-
GILFORD WERNER COCKAYNE RESTRICTIVE
RAUTENSTRAUCH THOMPSON
PROGERIA SYNDROME SYNDROME DERMOPATHY
SYNDROME SYNDROME
SYNDROME
Causative LMNA Unknown WRN , LMNA CSA RECQL4 ZMPSTE24,
gene(s) (ERCC8) LMNA
CSB
(ERCC6)
Inheritance Autosomal Unknown, likely Recessive Recessive Recessive Recessive
Dominant recessive
Onset Infancy Newborn Young adult Newborn/infancy Infancy Newborn
Growth Postnatal Intrauterine Onset after Postnatal Postnatal Intrauterine
retardation puberty
Hair loss + Total + Scalp patchy + Scalp, − + Diffuse + Diffuse
sparse,
graying
Skin + + + + + +
abnormalities
Subcutaneous + + + + − −
fat loss
Skin + Rarely − + − − −
calcification
Short stature + + + + + +
Coxa valga + − − − − −
Acroosteolysis + + + − − −
Mandibular + + − − − +
dysplasia
Osteopenia + Mild + + − + +
Vasculopathy + − + + − −
Heart failure + − + − − −
Strokes + − − − − −
Insulin + − + Rarely − − −
Resistance
Diabetes − − + − − −
Hypogonadism + − + + + −
Dental + + + + + +
abnormality
Voice + − + − − −
abnormality
Hearing loss + − − + − −
Joint + − − − − +
contractures
Hyperkeratosis − − + − + −
Cataracts − − + + + −
Tumor − − + − + −
predisposition
Intellectual − + − + − −
disability
Neurologic − + + Mild + − −
disorder
Adapted from Hegele RA: Drawing the line in Progeria syndromes, Lancet 362;416–417, 2003.
Altered activity of each enzyme in the pathway has been associated with a
specific type of porphyria (Table 110.1 ). The first enzyme, ALA synthase
(ALAS), occurs in 2 forms. An erythroid specific form, ALAS2, is deficient in
X-linked sideroblastic anemia, as a result of mutations of the ALAS2 gene on
chromosome Xp11.2. Gain-of-function mutations of ALAS2 caused by deletions
in the last exon cause X-linked protoporphyria (XLP ), which is phenotypically
identical to erythropoietic protoporphyria.
Table 110.1
The Human Porphyrias: Mutations, Time of Presentation,
and Tissue- and Symptom-Based Classifications
Classification*
DISEASE ENZYME INHERITANCE PRESENTATION
H E A/N C
X-Linked δ-Aminolevulinate X-linked Childhood X X
protoporphyria (XLP) synthase 2 (ALAS2)
δ-Aminolevulinic acid δ-Aminolevulinic Autosomal recessive Mostly post X X* X
dehydratase porphyria acid dehydratase puberty
(ADP) (ALAD)
Acute intermittent Porphobilinogen Autosomal dominant Post puberty X X
porphyria (AIP) deaminase (PBGD)
Homozygous AIP Homozygous dominant Childhood X X X
Congenital Uroporphyrinogen Autosomal recessive In utero or infancy X X
erythropoietic III synthase (UROS)
porphyria (CEP)
Porphyria cutanea tarda Uroporphyrinogen Sporadic Adults X X
(PCT) type 1 decarboxylase
(UROD)
PCT type 2 † Autosomal dominant Adults X X
PCT type 3 Unknown Adults X X
Hepatoerythropoietic Homozygous dominant Childhood X X* X
porphyria (HEP)
Hereditary Coproporphyrinogen Autosomal dominant Post puberty X X X
coproporphyria (HCP) oxidase (CPOX)
Homozygous HCP Homozygous dominant Childhood X X X X
Variegate porphyria Protoporphyrinogen Autosomal dominant Post puberty X X X
(VP) oxidase (PPOX)
Homozygous VP Homozygous dominant Childhood X X X X
Erythropoietic Ferrochelatase Autosomal recessive (most Childhood X X
protoporphyria (EPP) (FECH) commonly heteroallelic
with hypomorphic allele)
Features
PRESENTING EXACERBATING MOST IMPORTANT
TREATMENT
SYMPTOMS FACTORS SCREENING TESTS
Acute Neurologic, adult Drugs (mostly P450 Urinary Hemin, glucose
intermittent onset inducers), progesterone, porphobilinogen
porphyria dietary restriction
Porphyria Skin blistering and Iron, alcohol, smoking, Plasma (or urine) Phlebotomy, low-
cutanea tarda fragility (chronic), estrogens, hepatitis C, porphyrins dose
adult onset HIV, halogenated hydroxychloroquine
hydrocarbons
Erythropoietic Phototoxic pain and Total erythrocyte Sun protection
protoporphyria swelling (mostly protoporphyrin with
acute), childhood metal-free and zinc
onset protoporphyrin
Table 110.3
Alcohol Tetracycline
ACEIs (especially enalapril) ‡
Spironolactone
CCBs (especially nifedipine) ‡
Ketoconazole
Ketamine*
*
Porphyria has been listed as a contraindication, warning, precaution, or adverse effect in U.S.
labeling for these drugs. Estrogens are also listed as harmful in porphyria but have been
implicated as harmful in acute porphyrias, mostly based only on experience with estrogen-
progestin combinations. Although estrogens can exacerbate porphyria cutanea tarda, there is little
evidence they are harmful in the acute porphyrias.
† Porphyria has been listed as a precaution in U.S. labeling for this drug. However, this drug is
Neurologic Mechanisms
The mechanism of neural damage in acute porphyrias is poorly understood. The
most favored hypothesis at present is that 1 or more heme precursors, or perhaps
a derivative, are neurotoxic. Increased ALA in AIP, HCP, VP, ADP, plumbism,
and hereditary tyrosinemia type 1, which have similar neurologic manifestations,
suggests that this substance or a derivative may be neuropathic. Porphyrins
derived from ALA after its uptake into cells may have toxic potential. ALA can
also interact with γ-aminobutyric acid (GABA) receptors. Severe AIP greatly
improves after allogeneic liver transplantation. This experience and the
demonstration that recipients of AIP livers develop porphyria support the
hypothesis that heme precursors from the liver cause the neurologic
manifestations.
Epidemiology
AIP occurs in all races and is the most common acute porphyria, with an
estimated prevalence in most countries of 5 in 100,000. In Sweden, prevalence
was estimated to be 7.7 in 100,000, including latent cases with normal porphyrin
precursors. A much higher prevalence of 60-100 in 100,000 in northern Sweden
is the result of a founder effect. The combined prevalence of AIP and VP in
Finland is approximately 3.4 in 100,000. A survey of chronic psychiatric patients
in the United States using an erythrocyte PBGD determination found a high
prevalence (210 in 100,000) of PBGD deficiency, but a study in Mexico found a
similar prevalence in psychiatric patients and controls. Population screening by
erythrocyte PBGD activity or DNA analysis revealed a prevalence of 200
heterozygotes per 100,000 people in Finland, and 1 in approximately 1,675 (60
in 100,000) in France. Studies using exomic/genomic databases show that the
estimated frequency of pathogenic mutations in the HMBS gene is 0.00056 (56
in 100,000) suggesting that the penetrance of this disorder may be as low as 1%,
and that carriers of PBGD mutations that can cause AIP are much more common
than previously believed.
Clinical Manifestations
Neurovisceral manifestations of acute porphyrias may appear any time after
puberty, but rarely before (Table 110.4 ). Symptomatic childhood cases have
been reported, but most were not adequately documented biochemically and
confirmed by genetic testing. Abdominal pain is the most common presenting
symptom in such cases, but seizures are common and may precede the diagnosis
of AIP. Other manifestations reported in children include peripheral neuropathy,
myalgias, hypertension, irritability, lethargy, and behavioral abnormalities. A
population-based study in Sweden indicated that symptoms suggestive of
porphyria may occur in heterozygotes during childhood, even, in contrast to
adults, when urinary porphyria precursors are not elevated. This study did not
compare the frequency of such nonspecific symptoms in a control group of
children. Very rare cases of homozygous AIP present differently, with severe
neurologic manifestations early in childhood.
Table 110.4
Common Presenting Symptoms and Signs of Acute
Porphyria
SYMPTOMS FREQUENCY
COMMENT
AND SIGNS (%)
GASTROINTESTINAL
Abdominal 85–95 Usually unremitting (for hours or longer) and poorly localized but can be
pain cramping.
Vomiting 43–88 Neurologic in origin and rarely accompanied by peritoneal signs, fever, or
leukocytosis.
Constipation 48–84 Nausea and vomiting often accompany abdominal pain. May be accompanied by
bladder paresis.
Diarrhea 5–12
NEUROLOGIC
Pain in 50–70 Pain may begin in the chest or back and move to the abdomen. Extremity pain
extremities, chest, neck, or head indicates involvement of sensory nerves; objective sensory
back loss reported in 10–40% of cases.
Paresis 42–68 May occur early or late during a severe attack. Muscle weakness usually begins
proximally rather than distally and more often in the upper than lower
extremities.
Respiratory 9–20 Preceded by progressive peripheral motor neuropathy and paresis.
paralysis
Mental 40–58 May range from minor behavioral changes to agitation, confusion,
symptoms hallucinations, and depression.
Convulsions 10–20 A central neurologic manifestation of porphyria or caused by hyponatremia,
which often results from syndrome of inappropriate antidiuretic hormone
secretion or sodium depletion.
CARDIOVASCULAR
Tachycardia 64–85 May warrant treatment to control rate, if symptomatic
Systemic 36–55 May require treatment during acute attacks, and sometimes becomes chronic.
arterial
hypertension
From Anderson KE, Bloomer JR, Bonkovsky HL, et al: Desnick recommendations for the
diagnosis and treatment of the acute porphyrias, Ann Intern Med 142(6):439–450, 2005.
Anxiety †
Perspiration †
Palpitation (tachycardia) †
Pallor ‡
Tremulousness ‡
Weakness
Hunger
Nausea
Emesis
Headache †
Mental confusion †
Visual disturbances (↓ acuity, diplopia) †
Organic personality changes †
Inability to concentrate †
Dysarthria
Staring
Paresthesias
Dizziness
Amnesia
Ataxia, incoordination
Refusal to feed ‡
Somnolence, lethargy ‡
Seizures ‡
Coma
Stroke, hemiplegia, aphasia
Decerebrate or decorticate posture
* Some of these features will be attenuated if the patient is receiving β-
FIG. 111.1 Incidence of hypoglycemia by birthweight, gestational age, and intrauterine
growth. (From Lubchenco LO, Bard H: Incidence of hypoglycemia in newborn infants
classified by birthweight and gestational age, Pediatrics 47:831–838, 1971.)
The onset of symptoms in neonates varies from a few hours to a week after
birth. In approximate order of frequency, symptoms include jitteriness or
tremors, apathy, episodes of cyanosis, seizures, intermittent apneic spells or
tachypnea, weak or high-pitched cry, limpness or lethargy, difficulty feeding
(latching on), and eye rolling. Episodes of sweating, sudden pallor, hypothermia,
and cardiac arrest and failure may also occur. Frequently, a clustering of episodic
symptoms may be noted. Because these clinical manifestations may result from
various causes, it is critical to measure serum glucose levels and determine
whether symptoms disappear with the administration of sufficient glucose to
raise the blood glucose to normal levels; if they do not, other diagnoses must be
considered.
Prematurity
Small for gestational age
Normal newborn
Panhypopituitarism
Isolated growth hormone deficiency
Adrenocorticotropic hormone deficiency
Addison disease (including congenital adrenal hypoplasia, adrenal
leukodystrophy, triple A syndrome, ACTH receptor deficiency, and
autoimmune disease complex)
Epinephrine deficiency
Glycogenolysis and Gluconeogenesis Disorders
Lipolysis Disorders
Fatty Acid Oxidation Disorders
Other Etiologies
Substrate-Limited Causes
Ketotic hypoglycemia
Poisoning—drugs
Salicylates
Alcohol
Oral hypoglycemic agents
Insulin
Propranolol
Pentamidine
Quinine
Disopyramide
Ackee fruit (unripe)—hypoglycin
Litchi – associated toxin (toxic hypoglycemic syndrome).
Vacor (rat poison)
Trimethoprim-sulfamethoxazole (with renal failure)
L -Asparaginase and other antileukemic drugs
Liver Disease
Reye syndrome
Hepatitis
Cirrhosis
Hepatoma
Systemic Disorders
Sepsis
Carcinoma/sarcoma (secreting—insulin-like growth factor II)
Heart failure
Malnutrition
Malabsorption
Antiinsulin receptor antibodies
Antiinsulin antibodies
Neonatal hyperviscosity
Renal failure
Diarrhea
Burns
Shock
Chiari malformation
Postsurgical complication
Pseudohypoglycemia (leukocytosis, polycythemia)
Excessive insulin therapy of insulin-dependent diabetes mellitus
Factitious disorder
Nissen fundoplication (dumping syndrome)
Falciparum malaria
Table 111.4
Correlation of Clinical Features With Molecular Defects in
Persistent Hyperinsulinemic Hypoglycemia in Infancy
FAMILY MOLECULAR
TYPE MACROSOMIA HYPOGLYCEMIA/HYPERINSULINEMIA
HISTORY DEFECTS
Sporadic Present at birth Moderate/severe in 1st days to weeks of life Negative ? SUR1 /KIR 6.2
mutations not always
identified in diffuse
hyperplasia
Autosomal Present at birth Severe in 1st days to weeks of life Positive SUR /KIR 6.2
recessive
Table 111.5
Analysis of Critical Blood Sample During
Hypoglycemia and 30 Min After Glucagon*
Substrates
Glucose
Free fatty acids
Ketones
Lactate
Uric acid
Ammonia
Hormones
Insulin
Cortisol
Growth hormone
Thyroxine, thyroid-stimulating hormone
Insulin-like growth factor binding protein-1 †
≥40 mg/dL after glucagon given at the time of hypoglycemia strongly suggests a
hyperinsulinemic state with adequate hepatic glycogen stores and intact
glycogenolytic enzymes. If ammonia is elevated to 100-200 µM, consider
activating mutation of glutamate dehydrogenase.
Table 111.6
Criteria for Diagnosing Hyperinsulinism
Based on “Critical” Samples (Drawn at a
Time of Fasting Hypoglycemia: Plasma
Glucose <50 mg/dL)
From Stanley CA, Thomson PS, Finegold DN, et al: Hypoglycemia in infants
and neonates. In Sperling MA, editor: Pediatric endocrinology , ed 2,
Philadelphia, 2002, Saunders, pp 135–159.
Table 111.7
Diagnosis of Acute Hypoglycemia in Infants
and Children
Acute Symptoms Present
1. Careful history for relation of symptoms to time and type of food intake,
considering age of patient. Exclude possibility of alcohol or drug
ingestion. Assess possibility of insulin injection, salt craving, growth
velocity, or intracranial pathology.
2. Careful examination for hepatomegaly (glycogen storage disease; defect
in gluconeogenesis); pigmentation (adrenal failure); stature and
neurologic status (pituitary disease).
3. Admit to hospital for provocative testing:
a. 24 hr fast under careful observation; when symptoms provoked,
proceed with steps 1-4 as when acute symptoms present.
b. Pituitary-adrenal function using arginine-insulin stimulation test if
indicated.
4. Consider molecular diagnostic test before liver biopsy for histologic and
enzyme determinations.
5. Oral glucose tolerance test (1.75 g/kg; max 75 g) if reactive hypoglycemia
suspected (e.g., dumping syndrome).
Table 113.1
Disorders Associated with a Large Anterior Fontanel
Hypothyroidism
Achondroplasia
Apert syndrome
Cleidocranial dysostosis
Congenital rubella syndrome
Hallermann-Streiff syndrome
Hydrocephaly
Hypophosphatasia
Intrauterine growth restriction
Kenny syndrome
Osteogenesis imperfecta
Prematurity
Pyknodysostosis
Russell-Silver syndrome
Trisomies 13, 18, and 21
Vitamin D deficiency rickets
Face
of discharge.
Table 113.4
Criteria for Discharge of Healthy Term Newborns*
GENERAL
Normal vital signs including respiratory rate <60 breaths/min; axillary temperature 36.5°C-37.4°C (97.7°-99.3°F)
in open crib
Physical examination reveals no abnormalities requiring continued hospitalization
Regular urination; stool × 1
At least 2 uneventful, successful feedings
No excessive bleeding 2 hr after circumcision
LABORATORY AND OTHER SCREENS
Maternal syphilis, hepatitis B surface antigen, and HIV status
Newborn hepatitis B vaccine administered or appointment for vaccination confirmed
Maternal tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis, adsorbed (Tdap) vaccination
Maternal influenza vaccination during flu season
Evaluation and monitoring for sepsis based on maternal risk factors including GBS colonization
Umbilical or newborn direct Coombs test and blood type if clinically indicated
Expanded newborn metabolic screening
Hearing screening
Screening for hypoglycemia based on infant risk factors
Pulse oximetry screening
Screening for hyperbilirubinemia, with management and follow-up as recommended based on level of jaundice
SOCIAL
Evidence of parental knowledge, ability, and confidence to care for the baby at home:
Feeding
Normal stool and urine output
Cord, skin, and genital care
Recognition of illness (jaundice, poor feeding, lethargy, fever, etc.)
Infant safety (car seat, supine sleep position, etc.)
Availability of family and physician support (physician follow-up)
Assessment of family, environmental, and social risk factors:
Substance abuse
History of child abuse
Domestic violence
Mental illness
Teen mother
Homelessness
Barriers to follow-up
Source of continuing medical care is identified.
* Refers to infants born between 37 and 42 wk of gestation after uncomplicated pregnancy, labor,
and delivery.
From American Academy of Pediatrics Committee on Fetus and Newborn: Hospital stay for
healthy term newborn infants, Pediatrics 135:948–953, 2015.
Bibliography
Additional practices that encourage successful breastfeeding include
antepartum education and encouragement, immediate postpartum mother–infant
contact with suckling, demand feeding, inclusion of maternal partners in
breastfeeding education, and support from experienced women. Nursing at first
for least 5 min at each breast is reasonable, allows a baby to obtain most of the
available breast contents, and provides effective stimulation for increasing the
milk supply. Nursing episodes should then be extended according to the comfort
and desire of the mother and infant. A confident and relaxed mother, supported
by an encouraging home and hospital environment, is likely to nurse well. The
Baby-Friendly Hospital Initiative , a global effort sponsored by WHO and the
UN Children's Fund to promote breastfeeding, recommends 10 steps to
successful breastfeeding (Table 113.5 ). When instituted together as a complete
bundle, these practices can improve multiple outcomes, including breastfeeding
initiation, duration of exclusive breastfeeding, and duration of overall
breastfeeding. In the United States, however, the vast majority of newborns are
still not delivered in Baby-Friendly hospitals that have implemented all 10 steps.
Educating mothers during pregnancy and showing mothers how to breastfeed are
the most widely implemented strategies, while establishment of written
breastfeeding policies, restriction of formula access, and establishment of
breastfeeding support groups after discharge are among the most challenging to
implement.
Table 113.5
Ten Steps to Successful Breastfeeding
Every facility providing maternity services and care for newborn infants should accomplish the following:
1. Have a written feeding policy that is routinely communicated to staff and patients, comply with WHO
restrictions on marketing of breast milk substitutes, and establish ongoing monitoring and data-management
systems.
2. Ensure that staff have sufficient knowledge, competence, and skills to support breastfeeding.
3. Discuss the importance and management of breastfeeding with pregnant women and their families.
4. Facilitate immediate and uninterrupted skin-to-skin contact and help initiate breastfeeding as soon as
possible after birth.
5. Support mothers to initiate and maintain breastfeeding and manage common difficulties.
6. Give newborn infants no food or drink other than breast milk unless medically indicated.
7. Practice rooming-in (allow mothers and infants to remain together) 24 hr a day.
8. Support mothers to recognize and respond to their infants' feeding cues.
9. Counsel mothers on the use and risks of feeding bottles, teats, and pacifiers.
10. Coordinate discharge to ensure timely access to ongoing support and care.
Adapted from Guideline: Protecting, promoting and supporting breastfeeding in facilities providing
maternity and newborn services. Geneva, 2017, World Health Organization.
Table 113.6
Drugs of Abuse and Adverse Infant Effects
CONTRAINDICATED
Amphetamines
Antineoplastic agents
Bromocriptine
Chloramphenicol
Clozapine
Cocaine
Cyclophosphamide
Doxorubicin
Ecstasy (MDMA)
Ergots
Gold salts
Heroin
Immunosuppressants
Methamphetamine
Phencyclidine (PCP)
Radiopharmaceuticals
Thiouracil
USE WITH CAUTION
Alcohol
Amiodarone
Anthraquinones (laxatives)
Aspirin (salicylates)
Atropine
β-Adrenergic blocking agents
Benzodiazepines
Birth control pills
Bromides
Cascara
Codeine
Dicumarol
Dihydrotachysterol
Domperidone
Estrogens
Hydrocodone
Lithium
Marijuana
Metoclopramide
Meperidine
Oxycodone
Phenobarbital*
Primidone
Reserpine
Salicylazosulfapyridine (sulfasalazine)
* Watch for sedation.
Contraindications to Breastfeeding
Medical contraindications to breastfeeding in the United States include infants
with galactosemia, maple syrup urine disease, and phenylketonuria. Maternal
conditions that contraindicate breastfeeding include infection with human T-cell
lymphotropic virus types 1 and 2, active tuberculosis (until appropriately treated
≥2 wk and not considered contagious), herpesvirus infection on breast, use of or
dependence on certain illicit drugs, and maternal treatment with some
radioactive compounds (Table 113.7 ). Because clean water and affordable
replacement feeding are available in the United States, it is recommended that
HIV-infected mothers not breastfeed their infants regardless of maternal viral
load and antiretroviral therapy. However, in resource-limited countries where
diarrhea and pneumonia are significant causes of infant and child mortality,
breastfeeding may not be contraindicated for HIV-positive mothers receiving
antiretroviral therapy. Donor human milk, particularly that purchased online,
may be contaminated with potential pathogens. Contamination is much less of a
concern with pasteurized human milk obtained from a milk bank.
Table 113.7
Summary of Infectious Agents Detected in Milk and
Newborn Disease
BREAST MILK
DETECTED MATERNAL INFECTION
REPORTED AS CAUSE
INFECTIOUS AGENT IN BREAST CONTRAINDICATION TO
OF NEWBORN
MILK? BREASTFEEDING?
DISEASE?
BACTERIA
Mastitis/Staphylococcus aureus Yes No No, unless breast abscess
present
Mycobacterium tuberculosis :
Active disease Yes No Yes, because of aerosol spread,
or tuberculosis mastitis
Purified protein derivative skin No No No
test result positive, chest
radiograph findings negative
Escherichia coli , other gram- Yes, stored Yes, stored —
negative rods
Group B streptococci Yes Yes No*
Listeria monocytogenes Yes Yes No*
Coxiella burnetii Yes Yes No*
Syphilis No No No †
VIRUSES
HIV Yes Yes Yes, developed countries
Cytomegalovirus:
Term infant Yes Yes No
Preterm infant Yes Yes Evaluate on an individual basis
Hepatitis B virus Yes, surface No No, developed countries ‡
antigen
Hepatitis C virus Yes No No §
Hepatitis E virus Yes No No
Human T-cell leukemia virus Yes Yes Yes, developed countries
(HTLV)-1
HTLV-2 Yes Uncertain Yes, developed countries
Herpes simplex virus Yes Yes No, unless breast vesicles
present
Rubella
Wild type Yes Yes, rare No
Vaccine Yes No No
Varicella-zoster virus Yes No No, cover active lesions ¶
Epstein-Barr virus Yes No No
Human herpesvirus (HHV)-6 No No No
HHV-7 Yes No No
West Nile virus Possible Possible Unknown
Zika virus Yes No No
PARASITES
Toxoplasma gondii Yes Yes, 1 case No
* Provided that the mother and child are taking appropriate antibiotics.
†
Treat mother and child if active disease.
‡ Immunize and immune globulin at birth.
§ Provided that the mother is HIV seronegative. Mothers should be counseled that breast milk
transmission of hepatitis C virus has not been documented, but is theoretically possible.
¶ Provide appropriate antivaricella therapy or prophylaxis to newborn.
Adapted from Jones CA: Maternal transmission of infectious pathogens in breast milk, J Paediatr
Child Health 37:576–582, 2001.
Bibliography
American Academy of Pediatrics, Committee on Pediatric
AIDS. Infant feeding and transmission of human
immunodeficiency virus in the United States. Pediatrics .
2013;131:391–396.
American Academy of Pediatrics, Section on Breastfeeding.
Breastfeeding and the use of human milk. Pediatrics .
2012;129:e827–e841.
Becker GE, Remmington S, Remmington T. Early additional
food and fluids for healthy breastfed full-term infants.
C H A P T E R 11 4
High-Risk Pregnancies
Kristen R. Suhrie, Sammy M. Tabbah
Table 114.1
Factors Associated With High-Risk Pregnancy
ECONOMIC
Poverty
Unemployment
Uninsured, underinsured
Poor access to prenatal care
CULTURAL/BEHAVIORAL
Low educational status
Poor healthcare attitudes
No care or inadequate prenatal care
Cigarette, alcohol, or illicit drug use
Age <20 or >40 yr
Unmarried
Short interpregnancy interval (<18 mo between pregnancies)
Lack of support group (husband, family, religion)
Stress (physical, psychologic)
Black race (preterm birth rates are 48% higher than for other women)
BIOLOGIC/GENETIC
Previous low-birthweight or preterm infant
Low weight for height
Poor weight gain during pregnancy
Short stature
Poor nutrition
Consanguinity
Intergenerational effects
Low maternal birthweight
Maternal obesity
Hereditary diseases (inborn error of metabolism)
REPRODUCTIVE
Previous cesarean birth
Previous infertility
Conception by reproductive technology
Prolonged gestation (>40 wk)
Prolonged labor
Previous infant with cerebral palsy, intellectual impairment, birth trauma, or congenital anomalies
Abnormal lie (breech)
Multiple gestations
Premature rupture of membranes
Infection (systemic, amniotic, extra-amniotic, cervical)
Preeclampsia or eclampsia
Uterine bleeding (abruptio placentae, placenta previa)
Parity (0 or >5 previous deliveries)
Uterine or cervical anomalies
Fetal disease
Abnormal fetal growth
Idiopathic premature labor
Iatrogenic prematurity
High or low levels of maternal serum α-fetoprotein
MEDICAL
Diabetes mellitus
Hypertension
Congenital heart disease
Autoimmune disease
Sickle cell anemia
Intercurrent surgery or trauma
Sexually transmitted infection
Maternal hypercoagulable states
Exposure to prescription medications
TORCH (toxoplasmosis, other agents, rubella, cytomegalovirus, herpes simplex) infection
Table 114.2
Maternal Conditions Affecting the Fetus or Neonate
Table 114.3
Maternal Infections Affecting the Fetus or Newborn
Table 115.1
Fetal Diagnosis and Assessment
The most common noninvasive tests are the nonstress test (NST ) and the
biophysical profile (BPP ). The NST monitors the presence of fetal heart rate
(FHR ) accelerations that follow fetal movement. A reactive (normal) NST
result demonstrates 2 FHR accelerations of at least 15 beats/min above the
baseline FHR lasting 15 sec during 20 min of monitoring. A nonreactive NST
result suggests possible fetal compromise and requires further assessment with a
BPP. Although the NST has a low false-negative rate, it does have a high false-
positive rate, which is often remedied by the BPP. The full BPP assesses fetal
breathing, body movement, tone, NST, and amniotic fluid volume. It effectively
combines acute and chronic indicators of fetal well-being, which improves the
predictive value of abnormal testing (Table 115.2 ). A score of 2 or 0 is given for
each observation. A total score of 8-10 is reassuring; a score of 6 is equivocal,
and retesting should be done in 12-24 hr; and a score of 4 or less warrants
immediate evaluation and possible delivery. The BPP has good negative
predictive value. The modified BPP consists of the combination of an US
estimate of amniotic fluid volume (the amniotic fluid index) and the NST. When
results of both are normal, fetal compromise is very unlikely. Signs of
progressive compromise seen on Doppler US include reduced, absent, or
reversed diastolic waveform velocity in the fetal aorta or umbilical artery (see
Fig. 115.5 and Table 115.1 ). The umbilical vein and ductus venosus waveforms
are also used to assess the degree of fetal compromise. Fetuses at highest risk of
stillbirth often have combinations of abnormalities, such as growth restriction,
oligohydramnios, reversed diastolic Doppler umbilical artery blood flow
velocity, and a low BPP.
Table 115.2
Table 115.3
Characteristics of Decelerations of Fetal Heart Rate (FHR)
LATE DECELERATION
Visually apparent, usually symmetric gradual decrease and return of the FHR associated with a uterine
contraction.
A gradual FHR decrease is defined as duration of ≥30 sec from the onset to the nadir of the FHR.
The decrease in FHR is calculated from the onset to the nadir of the deceleration.
The deceleration is delayed in timing, with the nadir of the deceleration occurring after the peak of the
contraction.
In most cases, the onset, nadir, and recovery of the deceleration occur after the beginning, peak, and ending of the
contraction, respectively.
EARLY DECELERATION
Visually apparent, usually symmetric gradual decrease and return of the FHR associated with a uterine
contraction.
A gradual FHR decrease is defined as duration of ≥30 sec from the onset to the FHR nadir.
The decrease in FHR is calculated from the onset to the nadir of the deceleration.
The nadir of the deceleration occurs at the same time as the peak of the contraction.
In most cases, the onset, nadir, and recovery of the deceleration are coincident with the beginning, peak, and
ending of the contraction, respectively.
VARIABLE DECELERATION
Visually apparent, abrupt decrease in FHR.
An abrupt FHR decrease is defined as duration <30 sec from the onset of the deceleration to the beginning of the
FHR nadir of the deceleration.
The decrease in FHR is calculated from the onset to the nadir of the deceleration.
The decrease in FHR is ≥15 beats/min, lasting ≥15 sec, and <2 min in duration.
When variable decelerations are associated with uterine contractions, their onset, depth, and duration commonly
vary with successive uterine contractions.
From Macones GA, Hankins GDV, Spong CY, et al: The 2008 National Institute of Child Health
and Human Development workshop report on electronic fetal monitoring: update on definitions,
interpretation, and research guidelines, Obstet Gynecol 112:661–666, 2008.
Late decelerations are associated with fetal hypoxemia and are characterized
by onset after a uterine contraction is well established and persists into the
interval following resolution of the contraction. The late deceleration pattern is
usually associated with maternal hypotension or excessive uterine activity, but it
may be a response to any maternal, placental, umbilical cord, or fetal factor that
limits effective oxygenation of the fetus. The significance of late decelerations
varies according to the underlying clinical context. They are most likely to be
associated with true fetal hypoxemia/acidemia when they are recurrent and occur
in conjunction with decreased or absent variability. Late decelerations represent
a compensatory, chemoreceptor-mediated response to fetal hypoxemia. The
transient decrease in FHR serves to increase ventricular preload during the peak
of hypoxemia (i.e., at the crest of a uterine contraction). If fetal acidemia
progresses, late decelerations may become less pronounced or absent, indicating
severe hypoxic depression of myocardial function. Prompt delivery is indicated
if late decelerations are unresponsive to oxygen supplementation, hydration,
discontinuation of labor stimulation, and position changes. Approximately 10–
15% of term fetuses have terminal (just before delivery) FHR decelerations that
are usually benign if they last <10 min before delivery.
A 3-tier system has been developed by a panel of experts for interpretation of
FHR tracings (Table 115.4 ). Category I tracings are normal and are strongly
predictive of normal fetal acid-base status at the time of the observation.
Category II tracings are not predictive of abnormal fetal status, but there is
insufficient evidence to categorize them as category I or III; therefore further
evaluation, surveillance, and reevaluation are indicated. Category III tracings
are abnormal and predictive of abnormal fetal acid-base status at the time of
observation. Category III tracings require prompt evaluation and efforts to
resolve expeditiously the abnormal FHR as previously discussed for late
decelerations.
Table 115.4
Three-Tier Fetal Heart Rate (FHR) Interpretation System
CATEGORY I
Category I FHR tracings include all the following:
• Baseline rate: 110-160 beats/min
• Baseline FHR variability: moderate
• Late or variable decelerations: absent
• Early decelerations: present or absent
• Accelerations: present or absent
CATEGORY II
Category II FHR tracings include all FHR tracings not categorized as category I or category III. Category II
tracings may represent an appreciable fraction of those encountered in clinical care. Examples of category II
FHR tracings include any of the following:
Baseline Rate
• Bradycardia not accompanied by absence of baseline variability
• Tachycardia
Baseline FHR Variability
• Minimal baseline variability
• Absence of baseline variability not accompanied by recurrent decelerations
• Marked baseline variability
Accelerations
• Absence of induced accelerations after fetal stimulation
Periodic or Episodic Decelerations
• Recurrent variable decelerations accompanied by minimal or moderate baseline variability
• Prolonged deceleration, ≥2 min but <10 min
• Recurrent late decelerations with moderate baseline variability
• Variable decelerations with other characteristics, such as slow return to baseline, “overshoots,” and “shoulders”
CATEGORY III
Category III FHR tracings include either:
• Absence of baseline FHR variability
or
Any of the following:
• Recurrent late decelerations
• Recurrent variable decelerations
• Bradycardia
• Sinusoidal pattern
Adapted from Macones GA, Hankins GDV, Spong CY, et al: The 2008 National Institute of Child
Health and Human Development workshop report on electronic fetal monitoring: update on
definitions, interpretation, and research guidelines, Obstet Gynecol 112:661–666, 2008.
Bibliography
for drugs that are contraindicated in pregnancy on the basis of animal and human
evidence and for which the risk exceeds the benefits.
The use of medications or herbal remedies during pregnancy is potentially
harmful to the fetus. Consumption of medications occurs during the majority of
pregnancies. The average mother has taken 4 drugs other than vitamins or iron
during pregnancy. Almost 40% of pregnant women receive a drug for which
human safety during pregnancy has not been established (category C pregnancy
risk). Moreover, many women are exposed to potential reproductive toxins, such
as occupational, environmental, or household chemicals, including solvents,
pesticides, and hair products. The effects of drugs taken by the mother vary
considerably, especially in relation to the time in pregnancy when they are taken
and the fetal genotype for drug-metabolizing enzymes.
Miscarriage or congenital malformations result from the maternal ingestion
of teratogenic drugs during the period of organogenesis. Maternal medications
taken later, particularly during the last few weeks of gestation or during labor,
tend to affect the function of specific organs or enzyme systems, and these
adversely affect the neonate rather than the fetus (Tables 115.5 and 115.6 ). The
effects of drugs may be evident immediately in the delivery room or later in the
neonatal period, or they may be delayed even longer. The administration of
diethylstilbestrol during pregnancy, for instance, increased the risk for vaginal
adenocarcinoma in female offspring in the 2nd or 3rd decade of life.
Table 115.5
Agents Acting on Pregnant Women That May Adversely Affect the Structure or
Function of the Fetus and Newborn
DRUG EFFECT ON FETUS
Accutane (isotretinoin) Facial-ear anomalies, heart disease, CNS anomalies
Alcohol Congenital cardiac, CNS, limb anomalies; IUGR; developmental delay;
attention deficits; autism
Aminopterin Abortion, malformations
Amphetamines Congenital heart disease, IUGR, withdrawal
ACE inhibitors, angiotensin Oligohydramnios, IUGR, renal failure, Potter-like syndrome
receptor antagonists
Azathioprine Abortion
Busulfan (Myleran) Stunted growth; corneal opacities; cleft palate; hypoplasia of ovaries, thyroid,
and parathyroids
Carbamazepine Spina bifida, possible neurodevelopmental delay
Carbimazole Scalp defects, choanal atresia, esophageal atresia, developmental delay
Carbon monoxide Cerebral atrophy, microcephaly, seizures
Chloroquine Deafness
Chorionic villus sampling Probably no effect, possibly limb reduction
Cigarette smoking LBW for gestational age
Cocaine/crack Microcephaly, LBW, IUGR, behavioral disturbances
Cyclophosphamide Multiple malformations
Danazol Virilization
17α-Ethinyl testosterone Masculinization of female fetus
(Progestoral)
Hyperthermia Spina bifida
Infliximab Possible increased risk of live vaccine associated disease in infant; neutropenia
Lithium Ebstein anomaly, macrosomia
Lopinavir-ritonavir Transient adrenal dysfunction
6-Mercaptopurine Abortion
Methyl mercury Minamata disease, microcephaly, deafness, blindness, mental retardation
Methyltestosterone Masculinization of female fetus
Misoprostol Arthrogryposis, cranial neuropathies (Möbius syndrome), equinovarus
Mycophenolate mofetil Craniofacial, limb, cardiovascular, CNS anomalies
Norethindrone Masculinization of female fetus
Penicillamine Cutis laxa syndrome
Phenytoin Congenital anomalies, IUGR, neuroblastoma, bleeding (vitamin K deficiency)
Polychlorinated biphenyls Skin discoloration—thickening, desquamation, LBW, acne, developmental
delay
Prednisone Oral clefts
Progesterone Masculinization of female fetus
Quinine Abortion, thrombocytopenia, deafness
Selective serotonin reuptake Small increased risk of congenital anomalies, persistent pulmonary hypertension
inhibitors of newborn
Statins IUGR, limb deficiencies, VACTERAL
Stilbestrol (diethylstilbestrol Vaginal adenocarcinoma in adolescence
[DES])
Streptomycin Deafness
Tetracycline Retarded skeletal growth, pigmentation of teeth, hypoplasia of enamel, cataract,
limb malformations
Thalidomide Phocomelia, deafness, other malformations
Toluene (solvent abuse) Craniofacial abnormalities, prematurity, withdrawal symptoms, hypertonia
Topiramate Cleft lip
Trimethadione and Abortion, multiple malformations, mental retardation
paramethadione
Valproate CNS (spina bifida), facial and cardiac anomalies, limb defects, impaired
neurologic function, autism spectrum disorder
Vitamin D Supravalvular aortic stenosis, hypercalcemia
Warfarin (Coumadin) Fetal bleeding and death, hypoplastic nasal structures
ACE, Angiotensin-converting enzyme; CNS, central nervous system; IUGR, intrauterine growth
restriction; LBW, low birthweight; VACTERAL, vertebral, anal, cardiac, tracheoesophageal fistula,
renal, arterial, limb.
Table 115.6
Agents Acting on Pregnant Women That May Adversely
Affect the Newborn Infant*
Acebutolol—IUGR, hypotension, bradycardia
Acetazolamide—metabolic acidosis
Amiodarone—bradycardia, hypothyroidism
Anesthetic agents (volatile)—CNS depression
Adrenal corticosteroids—adrenocortical failure (rare)
Ammonium chloride—acidosis (clinically inapparent)
Aspirin—neonatal bleeding, prolonged gestation
Atenolol—IUGR, hypoglycemia
Baclofen—withdrawal
Blue cohosh herbal tea—neonatal heart failure
Bromides—rash, CNS depression, IUGR
Captopril, enalapril—transient anuric renal failure, oligohydramnios
Caudal-paracervical anesthesia with mepivacaine (accidental introduction of anesthetic into scalp of baby)—
bradypnea, apnea, bradycardia, convulsions
Cholinergic agents (edrophonium, pyridostigmine)—transient muscle weakness
CNS depressants (narcotics, barbiturates, benzodiazepines) during labor—CNS depression, hypotonia
Cephalothin—positive direct Coombs test reaction
Dexamethasone—periventricular leukomalacia
Fluoxetine and other SSRIs—transient neonatal withdrawal, hypertonicity, minor anomalies, preterm birth,
prolonged QT interval
Haloperidol—withdrawal
Hexamethonium bromide—paralytic ileus
Ibuprofen—oligohydramnios, pulmonary hypertension
Imipramine—withdrawal
Indomethacin—oliguria, oligohydramnios, intestinal perforation, pulmonary hypertension
Intravenous fluids during labor (e.g., salt-free solutions)—electrolyte disturbances, hyponatremia, hypoglycemia
Iodide (radioactive)—goiter
Iodides—goiter
Lead—reduced intellectual function
Magnesium sulfate—respiratory depression, meconium plug, hypotonia
Methimazole—goiter, hypothyroidism
Morphine and its derivatives (addiction)—withdrawal symptoms (poor feeding, vomiting, diarrhea, restlessness,
yawning and stretching, dyspnea and cyanosis, fever and sweating, pallor, tremors, convulsions)
Naphthalene—hemolytic anemia (in G6PD-deficient infants)
Nitrofurantoin—hemolytic anemia (in G6PD-deficient infants)
Oxytocin—hyperbilirubinemia, hyponatremia
Phenobarbital—bleeding diathesis (vitamin K deficiency), possible long-term reduction in IQ, sedation
Primaquine—hemolytic anemia (in G6PD-deficient infants)
Propranolol—hypoglycemia, bradycardia, apnea
Propylthiouracil—goiter, hypothyroidism
Pyridoxine—seizures
Reserpine—drowsiness, nasal congestion, poor temperature stability
Sulfonamides—interfere with protein binding of bilirubin; kernicterus at low levels of serum bilirubin, hemolysis
with G6PD deficiency
Sulfonylurea agents—refractory hypoglycemia
Sympathomimetic (tocolytic β-agonist) agents—tachycardia
Thiazides—neonatal thrombocytopenia (rare)
Tumor necrosis factor blocking agents—neutropenia, possible increased risk of infection during 1st yr of life
Valproate—developmental delay
Zolpidem (Ambien)—low birthweight
* See also Table 115.5 .
Often the risk of controlling maternal disease must be balanced with the risk
FIG. 115.7 Assessment of fetal anatomy. A, Overall view of the uterus at 24 wk showing
a longitudinal section of the fetus and an anterior placenta. B, Transverse section at the
level of the lateral ventricle at 18 wk showing (on the right ) prominent anterior horns of
the lateral ventricles on either side of the midline echo of the falx. C, Cross section of
the umbilical cord showing that the lumen of the umbilical vein is much wider than that
of the 2 umbilical arteries. D, Four-chambered view of the heart at 18 wk with equal-
sized atria. E(i), Normal male genitals near term. E(ii), Hydrocele outlining a testicle
within the scrotum projecting into a normal-size pocket of amniotic fluid at 38 wk.
Approximately 2% of male infants after birth have clinical evidence of a hydrocele that
is often bilateral, not to be confused with subcutaneous edema occurring during
vaginal breech birth. F, Section of a thigh near term showing thick subcutaneous tissue
(4.6 mm between markers ) above the femur of a fetus with macrosomia. G, Fetal face
viewed from below, showing (from right to left ) the nose, alveolar margin, and chin at
20 wk. (From Special investigative procedures. In Beischer NA, Mackay EV, Colditz PB,
editors: Obstetrics and the newborn, ed 3, Philadelphia, 1997, Saunders.)
Table 115.7
Significance of Fetal Ultrasonographic Anatomic Findings
Table 115.8
Fetal Therapy
EXIT, Ex utero intrapartum treatment; IVIG, intravenous immune globulin; (?), possible but not
proved efficacy.
Numerous diagnoses have been evaluated for the possibility of fetal intervention
(Tables 116.1 and 116.2 ). Some have proved beneficial to the developing infant,
some have been abandoned, and some are still under investigation.
Table 116.1
Fetal Diagnoses Evaluated and Treated in Fetal Centers
Amniotic band syndrome (ABS)
Anomalies in monochorionic twins
Aortic stenosis
Arachnoid cyst
Bladder exstrophy
Bladder outlet obstruction
Bronchopulmonary sequestration (BPS)
Cervical teratoma
Cloaca
Cloaca exstrophy
Complete heart block
Congenital pulmonary airway malformation (CPAM)
Congenital diaphragmatic hernia (CDH)
Congenital high airway obstruction syndrome (CHAOS)
EXIT to airway procedure for CHAOS
Conjoined twins
Dandy-Walker malformation
Duodenal atresia
Encephalocele
Enteric duplicational atresia
Esophageal atresia
Gastroschisis
Hydrocephalus
Hydronephrosis
Hypoplastic left heart syndrome (HLHS)
Imperforate anus
Intraabdominal cyst
Lymphangioma
Mediastinal teratoma
Myelomeningocele, spina bifida
Neuroblastoma
Obstructive uropathy
Omphalocele
Pentalogy of Cantrell
Pericardial teratoma
Pleural effusions
Pulmonary agenesis
Pulmonary atresia with intact ventricular septum
Sacrococcygeal teratoma (SCT)
Twin reversed arterial perfusion (TRAP) sequence
Twin-twin transfusion syndrome (TTTS)
Vein of Galen aneurysm
EXIT, Ex utero intrapartum treatment.
Table 116.2
Indications and Rationales for in Utero Surgery on the
Fetus, Placenta, Cord, or Membranes
Fetal Centers
The value of fetal centers extends beyond fetal surgery. Often, families will
present to a fetal center with a newly discovered diagnosis and little
understanding of what the diagnosis means for their baby. Prenatal counseling
by the fetal team can provide comfort to the family by helping them understand
the diagnosis and treatment options and by developing a management plan that
may include fetal surgery. Some plans may call for enhanced monitoring of the
fetus and mother, followed by complex deliveries involving multidisciplinary
delivery teams and specialized equipment, as required for EXIT to ECMO, EXIT
to airway, EXIT to tumor resection, delivery to cardiac catheterization, and
procedures on placental support. Other plans may focus on postnatal therapy.
Not all severely affected fetuses have available therapies in utero or after
birth. In these lethal situations, fetal care planning will provide support for the
family and a plan for delivery room or nursery palliative care (Table 116.3 ) (see
Chapter 7 ).
Table 116.3
Selection of Patients for Fetal Repair
Bibliography
Adzick NS. Management of fetal lung lesions. Clin Perinatol .
2009;36(2):363–376.
Adzick NS, Thom EA, Spong CY, et al. A randomized trial of
prenatal versus postnatal repair of myelomeningocele. N Engl
J Med . 2011;364(11):993–1004.
Al-Maary J, Eastwood MP, Russo FM, et al. Fetal tracheal
occlusion for severe pulmonary hypoplasia in isolated
congenital diaphragmatic hernia: a systematic review and
meta-analysis of survival. Ann Surg . 2016;264(6):929–933.
Antiel RM, Flake AW. Responsible surgical innovation and
C H A P T E R 11 7
The term high-risk infant designates an infant at greater risk for neonatal
morbidity and mortality; many factors can contribute to an infant being high risk
(Table 117.1 ). High-risk infants are categorized into 4 main groups: the preterm
infant, infants with special health care needs or dependence on technology,
infants at risk because of family issues, and infants with anticipated early death.
Table 117.1
Factors in Considering Infants as High Risk for Morbidity
or Mortality in the Neonatal Period
MATERNAL DEMOGRAPHIC/SOCIAL FACTORS
Maternal age <16 yr or >40 yr
Illicit drug, alcohol, cigarette use
Poverty
Unmarried
Emotional or physical stress
MATERNAL MEDICAL HISTORY
Genetic disorders
Diabetes mellitus
Hypertension
Asymptomatic bacteriuria
Rheumatologic illness (systemic lupus erythematosus)
Immune-mediated diseases (IgG crossing placenta)
Long-term medication (see Chapters 115.4 and 115.5 )
PREVIOUS PREGNANCY
Intrauterine fetal demise
Neonatal death
Prematurity
Intrauterine growth restriction
Congenital malformation
Incompetent cervix
Blood group sensitization, neonatal jaundice
Neonatal thrombocytopenia
Hydrops
Inborn errors of metabolism
PRESENT PREGNANCY
Vaginal bleeding (abruptio placentae, placenta previa)
Sexually transmitted infections (colonization: herpes simplex, group B streptococcus, chlamydia, syphilis,
hepatitis B, HIV)
Multiple gestation
Preeclampsia
Premature rupture of membranes
Short interpregnancy time
Poly-/oligohydramnios
Acute medical or surgical illness
Inadequate prenatal care
Familial or acquired hypercoagulable states
Abnormal fetal ultrasonographic findings
Treatment of infertility
LABOR AND DELIVERY
Premature labor (<37 wk)
Postdates pregnancy (≥42 wk)
Fetal distress
Immature lecithin/sphingomyelin ratio; absence of phosphatidylglycerol
Breech presentation
Meconium-stained fluid
Nuchal cord
Cesarean delivery
Forceps delivery
Apgar score <4 at 5 min
NEONATE
Birthweight <2,500 g or >4,000 g
Birth <37 wk or ≥42 wk of gestation
Small or large for gestational age
Respiratory distress, cyanosis
Congenital malformation
Pallor, plethora, petechiae
Table 117.2
Characteristic Changes in Monochorionic Twins With
Uncompensated Placental Arteriovenous Shunts
TWIN ON:
Arterial Side—Donor Venous Side—Recipient
Prematurity Prematurity
Oligohydramnios Polyhydramnios
Small premature Hydrops
Malnourished Large premature
Pale Well nourished
Anemic Plethoric
Hypovolemic Polycythemic
Hypoglycemic Hypervolemic
Microcardia Cardiac hypertrophy
Glomeruli small or normal Myocardial dysfunction
Arterioles thin walled Tricuspid valve regurgitation
Right ventricular outflow obstruction
Glomeruli large
Arterioles thick walled
FIG. 117.5 Color-dye-stained twin-to-twin transfusion syndrome placenta that was
treated using the Solomon technique. Blue and green dye used to stain the arteries, and
pink and yellow dye used to stain the veins. After identification and coagulation of each
individual anastomosis, the complete vascular equator is coagulated from one
placental margin to the other. (From Slaghekke F, Lopriore E, Lewi L, et al: Fetoscopic
laser coagulation of the vascular equator versus selective coagulation for twin-to-twin
transfusion syndrome: an open-label randomized controlled trial, Lancet 383:2144–
2150, 2014, Fig 3.)
Diagnosis
A prenatal diagnosis of pregnancy with twins is suggested by a uterine size that
is greater than that expected for gestational age, auscultation of 2 fetal hearts,
and elevated maternal serum α-fetoprotein (AFP) or human chorionic
gonadotropin (hCG) levels. It is confirmed by ultrasonography. Physical
examination of twins is necessary but not sufficient to determine zygosity of
twins. In the event that congenital anomalies are present or there are transfusion
or transplantation considerations, genetic testing of zygosity should be
performed. While noninvasive prenatal testing (NIPT) is becoming more
common, the results should be interpreted with caution in multiple-gestation
pregnancies until more findings are better established.
or early preterm.
FIG. 117.6 Preterm birth rates in the United States in 2007, 2014, 2015, and 2016
(provisional). There was a recent rise in preterm birth rate from 2014 to 2016. Preterm
birth rate further divided into early preterm birth (birth <32 wk) and late preterm birth
(birth at wk). (From Hamilton BE, Martin JA, Osterman MJK, et al: Births:
provisional data for 2016, Vital Statistics Rapid Release 2017; 1–21, Fig 4.)
Etiology
Despite the frequency of preterm birth, it is often difficult to determine a specific
cause. The etiology of preterm birth is multifactorial and involves complex
interactions between fetal, placental, uterine, and maternal factors. In the setting
of maternal or fetal conditions that prompt early delivery, as well as placental
and uterine pathology, causes of preterm birth can sometimes be identified
(Table 117.3 ).
Table 117.3
Identifiable Risk Factors for Preterm Birth
FETAL
Fetal distress
Multiple gestation
Erythroblastosis
Nonimmune hydrops
PLACENTAL
Placental dysfunction
Placenta previa
Placental abruption
UTERINE
Bicornuate uterus
Incompetent cervix (premature dilation)
MATERNAL
Previous preterm birth
Preeclampsia
Black race
Chronic medical illness (cyanotic heart disease, renal disease, thyroid disease)
Short interpregnancy interval
Infection (Listeria monocytogenes, group B streptococcus, urinary tract infection, bacterial vaginosis,
chorioamnionitis)
Obesity
Drug abuse (cocaine)
Young or advanced maternal age
OTHER
Premature rupture of membranes
Polyhydramnios
Iatrogenic
Assisted reproductive technology
Trauma
Table 117.4
Many drugs apparently safe for adults on the basis of toxicity studies may be
harmful to newborns, especially premature infants. Oxygen and a number of
drugs have proved toxic to premature infants in amounts not harmful to term
infants. Thus, administering any drug, particularly in high doses, that has not
undergone pharmacologic testing in premature infants should be undertaken
carefully after risks have been weighed against benefits.
Table 117.5
Neonatal Morbidities Associated With Prematurity
RESPIRATORY
Respiratory distress syndrome (hyaline membrane disease)
Bronchopulmonary dysplasia*
Pneumothorax, pneumomediastinum; interstitial emphysema
Congenital pneumonia
Apnea
CARDIOVASCULAR
Patent ductus arteriosus
Hypotension
Bradycardia (with apnea)
HEMATOLOGIC
Anemia (early or late onset)
GASTROINTESTINAL
Poor gastrointestinal function—poor motility
Necrotizing enterocolitis*
Hyperbilirubinemia—direct and indirect
Spontaneous gastrointestinal isolated perforation
METABOLIC-ENDOCRINE
Hypocalcemia
Hypoglycemia
Hyperglycemia
Metabolic acidosis
Hypothermia
Euthyroid but low thyroxine status
Osteopenia
CENTRAL NERVOUS SYSTEM
Intraventricular hemorrhage*
Periventricular leukomalacia*
Seizures
Retinopathy of prematurity*
Deafness
Hypotonia
RENAL
Hyponatremia
Hypernatremia
Hyperkalemia
Renal tubular acidosis
Renal glycosuria
Edema
OTHER
Infections* (congenital, perinatal, nosocomial: bacterial, viral, fungal, protozoal)
* Major neonatal morbidities.
Another study found that morbidity and mortality among VLBW infants
decreased between 2000 and 2009. This study was limited to live-born infants
with birthweight of 500-1500 g. For infants born in 2009, this study found a
12.4% mortality rate; 28% of infants developed BPD, 7% severe ROP, 5% NEC,
15% late-onset sepsis, 6% grade III or IV IVH, and 3% PVL; 51% survived
without significant neonatal morbidity.
Outcomes may be slightly improving with time; survival among infants born
22-24 wk gestation increased from 30% in 2000–2003 to 36% in 2008–2011.
The percentage surviving without neurodevelopmental impairment increased
from 16% to 20% over this same period. However, extreme prematurity is still
associated with significant risk of both mortality and major neonatal morbidities.
For infants who survive to discharge, prematurity, as well as neonatal
morbidities, put them at increased risk for developmental delays and impairment
as they age (see Chapter 117.5 ).
Bibliography
Adams M, Bassler D, Bucher HU, et al. Variability of very low
IUGR is associated with medical conditions that interfere with the circulation
and efficiency of the placenta, with the development or growth of the fetus, or
with the general health and nutrition of the mother (Table 117.6 ). Many factors
are common to both prematurely born and LBW infants with IUGR. IUGR is
associated with decreased insulin production or insulin-like growth factor (IGF)
action at the receptor level. Infants with IGF-1 receptor defects, pancreatic
hypoplasia, or transient neonatal diabetes have IUGR. Genetic mutations
affecting the glucose-sensing mechanisms of the pancreatic islet cells result in
decreased insulin release (loss of function of the glucose-sensing glucokinase
gene) and give rise to IUGR.
Table 117.6
Factors Often Associated With Intrauterine Growth
Restriction
FETAL
Chromosomal disorders
Chronic fetal infections (cytomegalic inclusion disease, congenital rubella, syphilis)
Congenital anomalies—syndrome complexes
Irradiation
Multiple gestation
Pancreatic hypoplasia
Insulin deficiency (production or action of insulin)
Insulin-like growth factor type I deficiency
PLACENTAL
Decreased placental weight, cellularity, or both
Decrease in surface area
Villous placentitis (bacterial, viral, parasitic)
Infarction
Tumor (chorioangioma, hydatidiform mole)
Placental separation
Twin transfusion syndrome
MATERNAL/PATERNAL
Toxemia
Hypertension or renal disease, or both
Hypoxemia (high altitude, cyanotic cardiac or pulmonary disease)
Malnutrition (micronutrient or macronutrient deficiencies)
Chronic illness
Sickle cell anemia
Drugs (narcotics, alcohol, cigarettes, cocaine, antimetabolites)
IGF2 mutation (paternal)
Table 117.7
↓, Decreased; ↑, increased.
Large-for-Gestational-Age Infants
Infants with birthweight >90th percentile for gestational age are called large for
early intervention
Individuals with Disabilities Act
neonatal follow-up program
Table 117.8
Readiness for Discharge of High-Risk Infants Criteria
Resolution of acute life-threatening illnesses
Ongoing follow-up for chronic but stable problems:
Bronchopulmonary dysplasia
Intraventricular hemorrhage
Necrotizing enterocolitis after surgery or recovery
Ventricular septal defect, other cardiac lesions
Anemia
Retinopathy of prematurity
Hearing problems
Apnea
Cholestasis
Stable temperature regulation
Gain of weight with enteral feedings:
Breastfeeding
Bottle feeding
Gastric tube feeding
Free of significant apnea
Appropriate immunizations and planning for respiratory syncytial virus prophylaxis if indicated
Hearing screenings
Ophthalmologic examination if <30 wk of gestation or <1,500 g at birth
Parental knowledge, skill, and confidence documented in:
Administration of medications (diuretics, methylxanthines, aerosols, etc.)
Use of oxygen, apnea monitors, oximeters
Nutritional support:
Timing
Volume
Mixing concentrated formulas
Recognition of illness and deterioration
Basic cardiopulmonary resuscitation
Infant safety
Scheduling of referrals:
Primary care provider
Neonatal follow-up clinic
Occupational therapy/physical therapy
Imaging (head ultrasound)
Assessment of and solution to social risks
Data from American Academy of Pediatrics, American College of Obstetricians: Guidelines for
perinatal care, ed 7, Elk Grove Village, IL, 2013, American Academy of Pediatrics.
If all major medical problems have resolved and the home setting is adequate,
premature infants may then be discharged when their weight approaches 1,800-
2,000 g, they are >34-35 wk PMA, and all the above criteria are met. Parental
education, close follow-up, and healthcare provider accessibility are all essential
for early discharge protocols. Ideally, the primary caregivers for the infant have a
chance to provide infant care in the hospital with nursing supervision and help
before discharge home. All high-risk infants should follow-up with their primary
care provider within a few days of discharge.
Postdischarge Follow-Up
Medical Follow-Up
Even after discharge from the hospital, high-risk infants need very close medical
follow-up. They continued to be at increased risk for poor weight gain and
failure to thrive. In the setting of viral illness, premature infants are at increased
risk for significant respiratory distress. Infants who are sent home on oxygen
need very close medical follow-up with frequent visits and assessments, often
with pulmonology. Table 117.9 lists common sequelae of prematurity.
Table 117.9
Sequelae of Prematurity
IMMEDIATE LATE
Hypoxia, Intellectual disability, spastic diplegia, microcephaly, seizures, poor school performance
ischemia
Intraventricular Intellectual disability, spasticity, seizures, post hemorrhagic hydrocephalus
hemorrhage
Sensorineural Hearing and visual impairment, retinopathy of prematurity, strabismus, myopia
injury
Respiratory Bronchopulmonary dysplasia, pulmonary hypertension, bronchospasm, malnutrition, subglottic
failure stenosis
Necrotizing Short-bowel syndrome, malabsorption, malnutrition
enterocolitis
Cholestatic Cirrhosis, hepatic failure, malnutrition
liver disease
Nutrient Osteopenia, fractures, anemia, growth failure
deficiency
Social stress Child abuse or neglect, failure to thrive, divorce
Other sequelae Sudden infant death syndrome, infections, inguinal hernia, cutaneous scars (chest tube, patent
ductus arteriosus ligation, intravenous infiltration), gastroesophageal reflux, hypertension,
craniosynostosis, cholelithiasis, nephrocalcinosis, cutaneous hemangiomas
Developmental Follow-Up
It is well known that premature infants are at greater risk for developmental
delays than their term counterparts; the more preterm, the greater the risk of
delay. In addition, certain postnatal morbidities (severe BPD, grade III or IV
intraventricular hemorrhage, severe ROP) are associated with significantly
increased risk of developmental delays. It is very important that preterm infants
Table 119.1
Cyanosis
Central cyanosis generates a broad differential diagnosis encompassing
respiratory, cardiac, CNS, infectious, hematologic, and metabolic etiologies
(Table 119.2 ). Typically, 5 g/dL of deoxyhemoglobin must be present in the
blood for central cyanosis to be clinically apparent. If respiratory insufficiency is
caused by pulmonary conditions, respirations tend to be rapid with increased
work of breathing. If caused by CNS depression, respirations tend to be irregular
and weak and are often slow. Cyanosis unaccompanied by obvious signs of
respiratory difficulty suggests cyanotic congenital heart disease or
methemoglobinemia. Cyanosis resulting from congenital heart disease may,
however, be difficult to distinguish clinically from cyanosis caused by
respiratory disease. Episodes of cyanosis may also be the initial sign of
hypoglycemia, bacteremia, meningitis, shock, or pulmonary hypertension.
Peripheral acrocyanosis is common in neonates and thought to represent
peripheral venous congestion associated with immature control of peripheral
vascular tone. It does not usually warrant concern unless poor perfusion is
suspected.
Table 119.2
Differential Diagnosis of Cyanosis in the Newborn
CENTRAL OR PERIPHERAL NERVOUS SYSTEM HYPOVENTILATION
Birth asphyxia
Intracranial hypertension, hemorrhage
Oversedation (direct or through maternal route)
Diaphragm palsy
Neuromuscular diseases
Seizures
RESPIRATORY DISEASE
Airway
Choanal atresia/stenosis
Pierre Robin syndrome
Intrinsic airway obstruction (laryngeal/bronchial/tracheal stenosis)
Extrinsic airway obstruction (bronchogenic cyst, duplication cyst, vascular compression)
Lung
Respiratory distress syndrome
Transient tachypnea
Meconium aspiration
Pneumonia (sepsis)
Pneumothorax
Congenital diaphragmatic hernia
Pulmonary hypoplasia
CARDIAC RIGHT-TO-LEFT SHUNT
Abnormal Connections (Pulmonary Blood Flow Normal or Increased)
Transposition of great vessels
Total anomalous pulmonary venous return
Truncus arteriosus
Hypoplastic left heart syndrome
Single ventricle or tricuspid atresia with large ventricular septal defect but without pulmonic stenosis
Obstructed Pulmonary Blood Flow (Pulmonary Blood Flow Decreased)
Pulmonic atresia with intact ventricular septum
Tetralogy of Fallot
Critical pulmonic stenosis with patent foramen ovale or atrial septal defect
Tricuspid atresia
Single ventricle with pulmonic stenosis
Ebstein malformation of tricuspid valve
Persistent fetal circulation (persistent pulmonary hypertension of newborn)
METHEMOGLOBINEMIA
Congenital (hemoglobin M, methemoglobin reductase deficiency)
Acquired (nitrates, nitrites)
Inadequate ambient O2 or less O2 delivered than expected (rare)
Disconnection of O2 supply to nasal cannula, head hood
Connection of air, rather than O2 , to a mechanical ventilator
SPURIOUS/ARTIFACTUAL
Oximeter artifact (poor contact between probe and skin, poor pulse searching)
Arterial blood gas artifact (contamination with venous blood)
OTHER
Hypoglycemia
Adrenogenital syndrome
Polycythemia
Blood loss
hepatitis, congenital atresia of the bile ducts, inspissated bile syndrome after
erythroblastosis fetalis, syphilis, herpes simplex other congenital infections, or
other conditions (see Chapter 123.3 ).
Pain
Pain in neonates may be unrecognized and/or undertreated. The intensive care of
neonates may involve several painful procedures, including blood sampling
(heelstick, venous or arterial puncture), endotracheal intubation and suctioning,
mechanical ventilation, and insertion of chest tubes and intravascular catheters.
Pain in neonates results in obvious distress and acute physiologic stress
responses, which may have developmental implications for pain in later life.
Moreover, knowing that infants may experience pain contributes to parental
stress.
Pain and discomfort are potentially avoidable problems during the treatment
of sick infants. The most common causes of painful stimuli include circumcision
pain and that associated with phlebotomy for metabolic screening tests. Oral
sucrose solutions are well tolerated by most infants and have proven efficacy for
procedural pain. For NICU infants, the most frequently used drugs are
intermittent or continuous doses of opioids (morphine, fentanyl) and
benzodiazepines (midazolam, lorazepam). Although the long-term effects of
opioids and sedatives are not well established, the first concern should be the
treatment and/or prevention of acute pain. Continuous opiate infusions should be
used with caution. Some minor but painful procedures performed in well
neonates can be managed with oral sucrose solutions (Table 119.3 ).
Table 119.3
Pain in the Neonate: General Considerations
• Pain in newborns is often unrecognized and/or undertreated.
• If a procedure is painful in adults, it should be considered painful in newborns.
• Healthcare institutions should develop and implement patient care policies to assess, prevent, and manage pain
in neonates.
• Pharmacologic agents with known pharmacokinetic and pharmacodynamic properties and demonstrated efficacy
in neonates should be used. Agents known to compromise cardiorespiratory function should be administered
only by persons experienced in neonatal airway management and in settings with the capacity for continuous
monitoring.
• Educational programs to increase the skills of healthcare professionals in the assessment and management of
stress and pain in neonates should be provided.
• Further research is needed to develop and validate neonatal pain assessment tools that are useful in the clinical
setting; to determine optimal behavioral and pharmacologic interventions; and to study long-term effects of pain
and pain management.
Data from American Academy of Pediatrics Committee on Fetus and Newborn, Committee on
Drugs, Section on Anesthesiology, Section on Surgery; Canadian Paediatric Society, Fetus and
Newborn Committee: Prevention and management of pain and stress in the neonate, Pediatrics
105:454–461, 2000; and from Anand KJS; International Evidence-Based Group for Neonatal Pain:
Consensus statement for the prevention and management of pain in the newborn, Arch Pediatr
Adolesc Med 155:173–180, 2001.
119.1
Hyperthermia
Elizabeth Enlow, James M. Greenberg
Bibliography
Velasco R, Gomez B, Hernandez-Bou S, et al. Validation of a
predictive model for identifying febrile young infants with
cerebellar injury and may be more predictive of adverse long-term outcome.
Prognosis
The degree of IVH and presence of PVL are strongly linked to survival and
neurodevelopmental impairment (Tables 120.1 and 120.2 ). For infants with
birthweight <1,000 g, the incidence of severe neurologic impairment (defined as
Bayley Scales of Infant Development II mental developmental index <70,
psychomotor development index <70, cerebral palsy, blindness, or deafness)
after IVH is highest with grade IV hemorrhage and lower birthweight. PVL,
cystic PVL, and progressive hydrocephalus requiring shunt insertion are each
independently associated with a poorer prognosis (Table 120.3 ). Current data
suggest that outcomes for infants with grade III/IV intraventricular hemorrhage
may be improving, with rates of cerebral palsy and neurodevelopmental
impairment closer to 30–40% at age 2 yr.
Table 120.1
Short-Term Outcome of Germinal Matrix–Intraventricular
Hemorrhage as a Function of Severity of Hemorrhage and
Birthweight*
rates (early and late deaths) are approximately 50–100% greater for each grade of hemorrhage
and birthweight than those shown in the table for early deaths alone.
PHI, Periventricular hemorrhagic infarction; PVD, progressive ventricular dilation.
Data from Murphy BP, Inder TE, Rooks V, Taylor GA, et al. Posthemorrhagic ventricular dilatation
in the premature infant: natural history and predictors of outcome, Arch Dis Child Fetal Neonatal
Ed 87:F37–F41, 2002.
Adapted from Inder TE, Perlman JM, Volpe JJ: Preterm intraventricular
hemorrhage/posthemorrhagic hydrocephalus. In Volpe's neurology of the newborn, ed 6,
Philadelphia, 2018, Elsevier (Table 24-15).
Table 120.2
Table 120.3
Most infants with IVH and acute ventricular distention do not have PHH .
Only 10–15% of LBW neonates with IVH develop PHH, which may initially
present without clinical signs (enlarging head circumference, lethargy, a bulging
fontanel or widely split sutures, apnea, and bradycardia). In infants in whom
symptomatic hydrocephalus develops, clinical signs may be delayed 2-4 wk
despite progressive ventricular distention and compression and thinning of the
cerebral cortex. Many infants with PHH have spontaneous regression; only 3–
5% of VLBW infants with PHH ultimately require shunt insertion. Those infants
infants with severe fetal acidosis (pH <6.7) (90% death/impairment) and a base
deficit >25 mmol/L (72% mortality). Multiorgan failure and insult can occur
(Table 120.4 ).
Table 120.4
Etiology
Most neonatal encephalopathy and seizure, in the absence of major congenital
malformations or metabolic or genetic syndromes, appear to be caused by
perinatal events. Brain MRI or autopsy findings in full-term neonates with
encephalopathy demonstrate that 80% have acute injuries, <1% have prenatal
injuries, and 3% have non–hypoxic-ischemic diagnoses. Fetal hypoxia may be
caused by various disorders in the mother, including: (1) inadequate oxygenation
of maternal blood from hypoventilation during anesthesia, cyanotic heart
disease, respiratory failure, or carbon monoxide poisoning; (2) low maternal
blood pressure from acute blood loss, spinal anesthesia, or compression of the
vena cava and aorta by the gravid uterus; (3) inadequate relaxation of the uterus
to permit placental filling as a result of uterine tetany caused by the
administration of excessive oxytocin; (4) premature separation of the placenta;
(5) impedance to the circulation of blood through the umbilical cord as a result
of compression or knotting of the cord; and (6) placental insufficiency from
maternal infections, exposures, diabetes, toxemia or postmaturity.
Placental insufficiency often remains undetected on clinical assessment.
Intrauterine growth restriction may develop in chronically hypoxic fetuses
without the traditional signs of fetal distress. Doppler umbilical waveform
velocimetry (demonstrating increased fetal vascular resistance) and
cordocentesis (demonstrating fetal hypoxia and lactic acidosis) identify a
chronically hypoxic infant (see Chapter 115 ). Uterine contractions may further
reduce umbilical oxygenation, depressing the fetal cardiovascular system and
CNS and resulting in low Apgar scores and respiratory depression at birth.
After birth, hypoxia may be caused by (1) failure of oxygenation as a result of
severe forms of cyanotic congenital heart disease or severe pulmonary disease;
(2) severe anemia (severe hemorrhage, hemolytic disease); (3) shock severe
enough to interfere with the transport of oxygen to vital organs from
overwhelming sepsis, massive blood loss, and intracranial or adrenal
hemorrhage; or (4) failure to breathe after birth because of in utero CNS injury
or drug-induced suppression.
Table 120.5
Topography of Brain Injury in Term Infants With Hypoxic-
Ischemic Encephalopathy and Clinical Correlates
AREA OF LONG-TERM
LOCATION OF INJURY CLINICAL CORRELATES
INJURY SEQUELAE
Selective Entire neuraxis, deep cortical area, Stupor or coma Cognitive delay
neuronal brainstem and pontosubicular Seizures Cerebral palsy
necrosis Hypotonia Dystonia
Oculomotor abnormalities Seizure disorder
Suck/swallow abnormalities Ataxia
Bulbar and
pseudobulbar palsy
Parasagittal Cortex and subcortical white Proximal-limb weakness Spastic
injury matter Upper extremities affected > quadriparesis
Parasagittal regions, especially lower extremities Cognitive delay
posterior Visual and auditory
processing difficulty
Focal Cortex and subcortical white Unilateral findings Hemiparesis
ischemic matter Seizures common and Seizures
necrosis Vascular injury (usually middle typically focal Cognitive delays
cerebral artery distribution)
Periventricular Injury to motor tracts, especially Bilateral and symmetric Spastic diplegia
injury lower extremity weakness in lower
extremities
More common in preterm
infants
Adapted from Volpe JJ, editor: Neurology of the newborn , ed 4, Philadelphia, 2001, Saunders.
Clinical Manifestations
Intrauterine growth restriction with increased vascular resistance may be an
indication of chronic fetal hypoxia before the peripartum period. During labor,
the fetal heart rate slows and beat-to-beat variability declines. Continuous heart
rate recording may reveal a variable or late deceleration pattern (see Chapter 115
, Fig. 115.4 ). Particularly in infants near term, these signs should lead to the
administration of high concentrations of oxygen to the mother and consideration
of immediate delivery to avoid fetal death and CNS damage.
At delivery, the presence of meconium-stained amniotic fluid indicates that
fetal distress may have occurred. At birth, affected infants may have neurologic
impairment and may fail to breathe spontaneously. Pallor, cyanosis, apnea, a
slow heart rate, and unresponsiveness to stimulation are also nonspecific initial
signs of potential HIE. During the ensuing hours, infants may be hypotonic, may
change from a hypotonic to a hypertonic state, or their tone may appear normal
(Tables 120.6 and 120.7 ). Cerebral edema may develop during the next 24 hr
and result in profound brainstem depression. During this time, seizure activity
may occur; it may be severe and refractory to typical doses of anticonvulsants.
Although most often a result of the HIE, seizures in asphyxiated newborns may
also be caused by vascular events (hemorrhage, arterial ischemic stroke, or sinus
venous thrombosis), metabolic derangements (hypocalcemia, hypoglycemia),
CNS infection, and cerebral dysgenesis or genetic disorders (nonketotic
hyperglycinemia, vitamin-dependent epilepsies, channelopathies). Conditions
that result in neuromuscular weakness and poor respiratory effort may also result
secondarily in neonatal hypoxic brain injury and seizure. Such conditions might
include congenital myopathies, congenital myotonic dystrophy, or spinal
muscular atrophy.
Table 120.6
Poor Predictive Variables for Death/Disability After
Hypoxic-Ischemic Encephalopathy
• Low (0–3) 10 min Apgar score
• Need for CPR in the delivery room
• Delayed onset (≥20 min) of spontaneous breathing
• Severe neurologic signs (coma, hypotonia, hypertonia)
• Seizures onset ≤12 hr or difficult to treat
• Severe, prolonged (~7 days) EEG findings including burst suppression pattern
• Prominent MRI basal ganglia/thalamic lesions
• Oliguria/anuria >24 hr
• Abnormal neurologic exam ≥14 days
Table 120.7
Hypoxic-Ischemic Encephalopathy in Term Infants
Diagnosis
MRI is the most sensitive imaging modality for detecting hypoxic brain injury in
FIG. 120.8 MR images of focal ischemic cerebral injury. MRI was performed on the 3rd
postnatal day. A, Axial T2-weighted mage shows a lesion in the distribution of the main
branch of the left middle cerebral artery. B, Diffusion-weighted image demonstrates the
lesion more strikingly. (From Volpe JJ, editor: Neurology of the newborn , ed 5,
Philadelphia, 2008, Saunders/Elsevier, p 422.)
Table 120.8
Major Aspects of MRI in Diagnosis of Hypoxic-Ischemic
Encephalopathy in the Term Infant
MAJOR CONVENTIONAL MR FINDINGS IN 1ST WEEK
Cerebral cortical gray-white differentiation lost (on T1W or T2W)
Cerebral cortical high signal (T1W and FLAIR), especially in parasagittal perirolandic cortex
Basal ganglia/thalamus, high signal (T1W and FLAIR), usually associated with the cerebral cortical changes but
possibly alone with increased signal in brainstem tegmentum in cases of acute severe insult
Parasagittal cerebral cortex, subcortical white matter, high signal (T1W and FLAIR)
Periventricular white matter, decreased signal (T1W) or increased signal (T2W)
Posterior limb of internal capsule, decreased signal (T1W or FLAIR)
Cerebrum in a vascular distribution, decreased signal (T1W), but much better visualized as decreased diffusion
(increased signal) on diffusion-weighted MRI
Diffusion-weighted MRI more sensitive than conventional MRI, especially in 1st days after birth, when former
shows decreased diffusion (increased signal) in injured areas
FLAIR, Fluid-attenuated inversion recovery; MRI, magnetic resonance imaging; T1W and T2W,
T1- and T2-weighted images.
From Volpe JJ, editor: Neurology of the newborn, ed 5, Philadelphia, 2008, Elsevier (Table 9-16).
Table 120.9
Value of Electroencephalography in Assessment of
Asphyxiated Term Infants
Detection of severe abnormalities (i.e., CLV, FT, BSP) in 1st hours of life has a positive predictive value of an
unfavorable outcome of 80–90%.
Severe abnormalities may improve within 24 hr (~50% of BSP and 10% of CLV/FT).
Rapid recovery of severe abnormalities is associated with a favorable outcome in 60% of cases.
The combination of early neonatal neurologic examination and early aEEG enhances the positive predictive value
and specificity.
aEEG, Amplitude-integrated encephalography; BSP, burst-suppression pattern; CLV, continuous
low voltage; FT, flat trace.
From Inder TE, Volpe JJ: Hypoxic-ischemic injury in the term infant: clinical-neurological features,
diagnosis, imaging, prognosis, therapy. In Volpe's neurology of the newborn, ed 6, Philadelphia,
2018, Elsevier (Table 20-28).
Table 120.10
Electroencephalographic Patterns of Prognostic
Significance in Asphyxiated Term Infants*
ASSOCIATED WITH FAVORABLE OUTCOME
Mild depression (or less) on day 1
Normal background by day 7
ASSOCIATED WITH UNFAVORABLE OUTCOME
Predominant interburst interval >20 sec on any day
Burst-suppression pattern on any day
Isoelectric tracing on any day
Mild (or greater) depression after day 12
* Associations with favorable or unfavorable outcome are generally ≥90%, but the clinical context
must be considered.
collapse may follow the negative airway pressures generated during inspiration,
or it may result from incoordination of the tongue and other upper airway
muscles involved in maintaining airway patency. Central apnea, which is
caused by decreased central nervous system (CNS) stimuli to respiratory
muscles, results in both airflow and chest wall motion being absent. Gestational
age is the most important determinant of respiratory control, with the frequency
of central apnea being inversely related to gestational age. The immaturity of the
brainstem respiratory centers is manifest by an attenuated response to CO2 and a
paradoxical response to hypoxia that results in central apnea rather than
hyperventilation. Mixed apnea is most often observed in apnea of prematurity
(50–75% of cases), with obstructive apnea preceding central apnea. Short
episodes of apnea are usually central, whereas prolonged ones are often mixed.
Apnea depends on the sleep state; its frequency increases during active (rapid
eye movement) sleep.
Although apnea is usually observed in preterm infants as a result of immature
respiratory control or an associated illness, apnea in term infants is uncommon,
often associated with serious pathology, and demands prompt diagnostic
evaluation. Apnea accompanies many primary diseases that affect neonates
(Table 122.1 ). These disorders produce apnea by direct depression of CNS
control of respiration (hypoglycemia, meningitis, drugs, intracranial hemorrhage,
seizures), disturbances in oxygen delivery (shock, sepsis, anemia), or ventilation
defects (obstruction of the airway, pneumonia, muscle weakness).The term
neonate with apnea should receive continuous cardiorespiratory monitoring
while performing an assessment for bacterial or viral sepsis/meningitis,
intracranial hemorrhage, seizures, and airway instability. Supportive care and
close monitoring are essential while the underlying etiology is ascertained and
appropriately treated.
Table 122.1
Apnea of Prematurity
Apnea of prematurity results from immature respiratory control, most frequently
occurs in infants <34 wk of gestational age (GA), and occurs in the absence of
identifiable predisposing diseases. The incidence of idiopathic apnea of
prematurity varies inversely with GA. Apnea of prematurity is almost universal
in infants born at <28 wk GA, and the incidence rapidly decreases from 85% of
infants <30 wk GA to 20% of infants <34 wk GA. The onset of apnea of
prematurity can be during the initial days to weeks of age but is often delayed if
there is RDS or other causes of respiratory distress. In premature infants without
respiratory disease, apneic episodes can occur throughout the 1st 7 postnatal
days with equal frequency.
Apnea in preterm infants is defined as cessation of breathing for ≥20 sec or for
any duration if accompanied by cyanosis and bradycardia (<80-100 beats/min).
The incidence of associated bradycardia increases with the length of the
preceding apnea and correlates with the severity of hypoxia. Short apnea
episodes (10 sec) are rarely associated with bradycardia, whereas longer
episodes (>20 sec) have a higher incidence of bradycardia. Bradycardia follows
the apnea by 1-2 sec in >95% of cases and is most often sinus, but on occasion it
can be nodal. Vagal responses and rarely heart block are causes of bradycardia
without apnea. Short, self-resolving oxygen desaturation episodes noted with
continuous monitoring are normal in neonates, and treatment is not necessary.
Preterm infants born at <35 wk GA are at risk for apnea of prematurity and
therefore should receive cardiorespiratory monitoring. Apnea that occurs in the
absence of other clinical signs of illness in the 1st 2 wk in a preterm infant is
likely apnea of prematurity, and therefore additional evaluation for other
etiologies is often unwarranted. However, the onset of apnea in a previously well
preterm neonate after the 2nd wk of life (or, as previously, in a term infant at any
time) is a critical event that may be associated with serious underlying
pathology. Prompt investigation for medication side effects, metabolic
derangements, structural CNS anomalies, intracranial hemorrhage, seizures, or
sepsis/meningitis is warranted.
reliable and correlated with discharge home on oxygen, length of hospital stay,
and hospital readmissions in the 1st yr of life. The risk of neurodevelopmental
impairment and pulmonary morbidity and the severity of BPD are directly
correlated.
Table 122.2
Definition of Bronchopulmonary Dysplasia (BPD):
Diagnostic Criteria*
GESTATIONAL AGE
<32 Wk ≥32 Wk
Time point 36 wk PMA or discharge home, whichever >28 days but <56 days postnatal age or
of comes first discharge home, whichever comes first
assessment Treatment with >21% oxygen for at least Treatment with >21% oxygen for at least 28
28 days plus : days plus :
Mild BPD Breathing room air at 36 wk PMA or discharge Breathing room air by 56 days postnatal age or
home, whichever comes first discharge home, whichever comes first
Moderate Need † for <30% oxygen at 36 wk PMA or Need † for <30% oxygen at 56 days postnatal age
BPD discharge home, whichever comes first or discharge home, whichever comes first
Severe Need † for ≥30% oxygen and/or positive Need † for ≥30% oxygen and/or positive pressure
BPD pressure (PPV or NCPAP) at 36 wk PMA or (PPV or NCPAP) at 56 days postnatal age or
discharge home, whichever comes first discharge home, whichever comes first
* BPD usually develops in neonates being treated with oxygen and PPV for respiratory failure,
most frequently respiratory distress syndrome (RDS). Persistence of the clinical features of
respiratory disease (tachypnea, retractions, crackles) is considered common to the broad
description of BPD and has not been included in the diagnostic criteria describing the severity of
BPD. Infants treated with >21% oxygen and/or PPV for nonrespiratory disease (e.g., central
apnea or diaphragmatic paralysis) do not have BPD unless parenchymal lung disease also
develops and they have clinical features of respiratory distress. A day of treatment with >21%
oxygen means that the infant received >21% oxygen for >12 hr on that day. Treatment with >21%
oxygen and/or PPV at 36 wk PMA or at 56 days postnatal age or discharge should not reflect an
“acute” event, but should rather reflect the infant's usual daily therapy for several days preceding
and after 36 wk PMA, 56 days postnatal age, or discharge.
† A physiologic test confirming that the oxygen requirement at the assessment time point remains
Clinical Manifestations
Infants with NEC have a variety of signs and symptoms and may have an
insidious or sudden catastrophic onset (Table 123.1 ). The onset of NEC is
usually in the 2nd or 3rd week of life but can be as late as 3 mo in VLBW
infants. Age of onset is inversely related to gestational age. The first signs of
impending disease may be nonspecific, including lethargy and temperature
instability, or related to GI pathology, such as abdominal distention, feeding
intolerance, and bloody stools. Because of nonspecific signs, sepsis may be
suspected before NEC. The spectrum of illness is broad, ranging from mild
disease with only guaiac-positive stools to severe illness with bowel perforation,
peritonitis, systemic inflammatory response syndrome, shock, and death.
Laboratory derangements may include neutropenia, anemia, thrombocytopenia,
coagulopathy, and metabolic acidosis. Hypotension and respiratory failure are
common. Progression may be rapid, but it is unusual for the disease to progress
from mild to severe after 72 hr.
Table 123.1
Signs and Symptoms Associated With Necrotizing
Enterocolitis
GASTROINTESTINAL
Abdominal distention
Abdominal tenderness
Feeding intolerance
Delayed gastric emptying
Vomiting
Occult/gross blood in stool
Change in stool pattern/diarrhea
Abdominal mass
Erythema of abdominal wall
SYSTEMIC
Lethargy
Apnea/respiratory distress
Temperature instability
“Not right”
Acidosis (metabolic and/or respiratory)
Glucose instability
Poor perfusion/shock
Disseminated intravascular coagulopathy
Positive results of blood cultures
From Kanto WP Jr, Hunter JE, Stoll BJ: Recognition and medical management of necrotizing
enterocolitis, Clin Perinatol 21:335–346, 1994.
Diagnosis
A very high index of suspicion in treating preterm at-risk infants is crucial. Plain
abdominal radiographs are essential to make a diagnosis of NEC. The finding of
pneumatosis intestinalis (air in the bowel wall) confirms the clinical suspicion
of NEC and is diagnostic; 50–75% of patients have pneumatosis when treatment
is started (Fig. 123.5 ). Portal venous gas is a sign of severe disease, and
pneumoperitoneum indicates a perforation (Figs. 123.6 and 123.7 ). Ultrasound
with Doppler flow assessment may be useful to evaluate for free fluid, abscess,
and bowel wall thickness, peristalsis, and perfusion.
specific transcutaneous bilirubin measurement, progressing jaundice, or risk for
hemolysis or sepsis. Infants with severe hyperbilirubinemia may present with
lethargy and poor feeding and, without treatment, can progress to acute bilirubin
encephalopathy (kernicterus) (see Chapter 123.4 ).
Differential Diagnosis
The distinction between physiologic and pathologic jaundice relates to the
timing, rate of rise, and extent of hyperbilirubinemia, because some of the same
causes of physiologic jaundice (e.g., large RBC mass, decreased capacity for
bilirubin conjugation, increased enterohepatic circulation) can also result in
pathologic jaundice. Evaluation should be determined on the basis of risk
factors, clinical appearance, and severity of the hyperbilirubinemia (Tables 123.2
to 123.4 ). Jaundice that is present at birth or appears within the 1st 24 hr after
birth should be considered pathologic and requires immediate attention.
Potential diagnoses would include erythroblastosis fetalis, concealed
hemorrhage, sepsis, or congenital infections, including syphilis, cytomegalovirus
(CMV), rubella, and toxoplasmosis. Hemolysis is suggested by a rapid rise in
serum bilirubin concentration (>0.5 mg/dL/hr), anemia, pallor, reticulocytosis,
hepatosplenomegaly, and a positive family history. An unusually high proportion
of direct-reacting bilirubin may characterize jaundice in infants who have
received intrauterine transfusions for erythroblastosis fetalis. Jaundice that first
appears on the 2nd or 3rd day is usually physiologic but may represent a more
severe form. Familial nonhemolytic icterus (Crigler-Najjar syndrome ) and
early-onset breastfeeding jaundice are seen initially on the 2nd or 3rd day.
Jaundice appearing after the 3rd day and within the 1st week suggests bacterial
sepsis or urinary tract infection; it may also be caused by other infections,
notably syphilis, toxoplasmosis, CMV, and enterovirus. Jaundice secondary to
extensive ecchymosis or blood extravasation may occur during the 1st day or
later, especially in premature infants. Polycythemia may also lead to early
jaundice.
Table 123.2
Risk Factors for Development of Severe
Hyperbilirubinemia*
MAJOR RISK FACTORS
Predischarge TSB or TcB level in the high-risk zone (see Fig. 123.10 )
Jaundice observed in the 1st 24 hr
Blood group incompatibility with positive direct antiglobulin test, other known hemolytic disease (G6PD
deficiency), elevated end-tidal CO concentration
Gestational age 35-36 wk
Previous sibling received phototherapy
Cephalohematoma or significant bruising
Exclusive breastfeeding, particularly if nursing is not going well and weight loss is excessive
East Asian race †
MINOR RISK FACTORS
Predischarge TSB or TcB level in the high intermediate-risk zone
Gestational age 37-38 wk
Jaundice observed before discharge
Previous sibling with jaundice
Macrosomic infant of a diabetic mother
Maternal age ≥25 yr
Male gender
DECREASED RISK ‡
TSB or TcB level in the low-risk zone (see Fig. 123.10 )
Gestational age ≥41 wk
Exclusive bottle feeding
Black race
Discharge from hospital after 72 hr
* In infants ≥35 wk of gestation; factors in approximate order of importance.
‡ These factors are associated with decreased risk of significant jaundice, listed in order of
decreasing importance.
G6PD, Glucose-6-phosphate dehydrogenase; TcB, transcutaneous bilirubin; TSB, total serum
bilirubin.
Adapted from American Academy of Pediatrics Subcommittee on Hyperbilirubinemia:
Management of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation,
Pediatrics 114:297–316, 2004.
Table 123.3
CMV, Cytomegalovirus; CBC, complete blood count; HSV, herpes simplex virus; PCR,
polymerase chain reaction; INR, international normalized ratio; TORCH, toxoplasmosis, other,
rubella, CMV, herpes; * Hepatic failure syndromes: HSV, CMV, gestational alloimmune liver
disease, mitochondrial liver disease, familial hemophagocytic syndrome; RBC, red blood cell.
Table 123.4
Diagnostic Features of the Various Types of Neonatal
Jaundice
There is a long differential diagnosis for jaundice first recognized after the 1st
week of life, including breast milk jaundice, septicemia, congenital atresia or
paucity of the bile ducts, hepatitis, galactosemia, hypothyroidism, CF, and
congenital hemolytic anemia crises related to RBC morphology and enzyme
deficiencies (Fig. 123.9 ). The differential diagnosis for persistent jaundice
during the 1st mo of life includes hyperalimentation-associated cholestasis,
hepatitis, cytomegalic inclusion disease, syphilis, toxoplasmosis, familial
nonhemolytic icterus, congenital atresia of the bile ducts, galactosemia, and
inspissated bile syndrome following hemolytic disease of the newborn. Rarely,
physiologic jaundice may be prolonged for several weeks, as in infants with
hypothyroidism or pyloric stenosis.
metalloporphyrins
opisthions
phototherapy
Clinical Manifestations
Signs and symptoms of kernicterus usually appear 2-5 days after birth in term
infants and as late as the 7th day in preterm infants, but hyperbilirubinemia may
lead to encephalopathy at any time during the neonatal period. The early signs
may be subtle and indistinguishable from those of sepsis, asphyxia,
hypoglycemia, intracranial hemorrhage, and other acute systemic illnesses in a
neonate. Lethargy, poor feeding, and loss of the Moro reflex are common initial
signs. Subsequently, the infant may appear gravely ill and prostrate, with
diminished tendon reflexes and respiratory distress. Opisthotonos with a bulging
fontanel, twitching of the face or limbs, and a shrill, high-pitched cry may
follow. In advanced cases, convulsions and spasm occur, with affected infants
stiffly extending their arms in an inward rotation with the fists clenched (Table
123.5 ). Rigidity is rare at this late stage.
Table 123.5
Clinical Features of Kernicterus
ACUTE FORM
Phase 1 (1st 1-2 days): poor suck, stupor, hypotonia, seizures
Phase 2 (middle of 1st wk): hypertonia of extensor muscles, opisthotonos, retrocollis, fever
Phase 3 (after the 1st wk): hypertonia
CHRONIC FORM
1st yr: hypotonia, active deep tendon reflexes, obligatory tonic neck reflexes, delayed motor skills
After 1st yr: movement disorders (choreoathetosis, ballismus, tremor), upward gaze, sensorineural hearing loss
From Dennery PA, Seidman DS, Stevenson DK: Neonatal hyperbilirubinemia, N Engl J Med
344:581–590, 2001.
Many infants who progress to these severe neurologic signs die; the survivors
are usually seriously damaged but may appear to recover and for 2-3 mo show
few abnormalities. Later in the 1st yr, opisthotonos, muscle rigidity, irregular
movements, and convulsions tend to recur. In the 2nd yr the opisthotonos and
seizures abate, but irregular, involuntary movements, muscle rigidity, or, in some
infants, hypotonia increase steadily. By 3 yr of age, the complete neurologic
syndrome is often apparent: bilateral choreoathetosis with involuntary muscle
spasms, extrapyramidal signs, seizures, mental deficiency, dysarthric speech,
high-frequency hearing loss, squinting, and defective upward eye movements.
Pyramidal signs, hypotonia, and ataxia occur in a few infants. In mildly affected
infants, the syndrome may be characterized only by mild to moderate
neuromuscular incoordination, partial deafness, or “minimal brain dysfunction,”
occurring singly or in combination; these problems may be unapparent until the
child enters school (see Table 123.5 ).
Table 123.6
Table 123.7
Example of a Clinical Pathway for Management of the
Newborn Infant Readmitted for Phototherapy or Exchange
Transfusion
TREATMENT
Use intensive phototherapy and/or exchange transfusion as indicated in Figs. 123.13 and 123.14 .
LABORATORY TESTS
TSB and direct bilirubin levels
Blood type (ABO, Rh)
Direct antibody test (Coombs)
Serum albumin
Complete blood cell count with differential and smear for red cell morphology
Reticulocyte count
End-tidal CO concentration (if available)
Glucose-6-phosphate dehydrogenase if suggested by ethnic or geographic origin or if poor response to
phototherapy
Urine for reducing substances
If history and/or presentation suggest sepsis, perform blood culture, urine culture, and cerebrospinal fluid for
protein, glucose, cell count, and culture.
INTERVENTIONS
If TSB ≥25 mg/dL (428 µmol/L) or ≥20 mg/dL (342 µmol/L) in a sick infant or infant <38 wk gestation, obtain a
type and crossmatch, and request blood in case an exchange transfusion is necessary.
In infants with isoimmune hemolytic disease and TSB level rising in spite of intensive phototherapy or within 2-3
mg/dL (34-51 µmol/L) of exchange level (see Fig. 123.14 ), administer intravenous immune globulin 0.5-1 g/kg
over 2 hr and repeat in 12 hr if necessary.
If infant's weight loss from birth is >12% or there is clinical or biochemical evidence of dehydration, recommend
formula or expressed breast milk. If oral intake is in question, give intravenous fluids.
FOR INFANTS RECEIVING INTENSIVE PHOTOTHERAPY:
Breastfeed or bottle-feed (formula or expressed breast milk) every 2-3 hr.
If TSB ≥25 mg/dL (428 µmol/L), repeat TSB within 2-3 hr.
If TSB 20-25 mg/dL (342-428 µmol/L), repeat within 3-4 hr. If TSB <20 mg/dL (342 µmol/L), repeat in 4-6 hr. If
TSB continues to fall, repeat in 8-12 hr.
If TSB is not decreasing or is moving closer to level for exchange transfusion, or if the TSB/albumin ratio exceeds
levels shown in Fig. 123.14 , consider exchange transfusion (see Fig. 123.14 for exchange transfusion
recommendations).
When TSB is <13-14 mg/dL (239 µmol/L), discontinue phototherapy.
Depending on the cause of the hyperbilirubinemia, it is an option to measure TSB 24 hr after discharge to check
for rebound.
TSB, Total serum bilirubin.
From American Academy of Pediatrics Subcommittee on Hyperbilirubinemia: Management of
hyperbilirubinemia in the newborn infant 35 or more weeks of gestation, Pediatrics 114:297–316,
2004.
Phototherapy
Clinical jaundice and indirect hyperbilirubinemia are reduced by exposure to
high-intensity light in the visible spectrum. Bilirubin absorbs light maximally in
the blue range (420-470 nm). Broad-spectrum white, blue, and special narrow-
spectrum (super) blue lights have been effective in reducing bilirubin levels.
Bilirubin in the skin absorbs light energy, causing several photochemical
reactions. One major product from phototherapy is a result of a reversible
photoisomerization reaction converting the toxic native unconjugated 4Z,15Z-
bilirubin into an unconjugated configurational isomer, 4Z,15E-bilirubin, which
can then be excreted in bile without conjugation. The other major product from
phototherapy is lumirubin, which is an irreversible structural isomer converted
from native bilirubin that can be excreted by the kidneys in the unconjugated
state.
The therapeutic effect of phototherapy depends on the light energy emitted in
FIG. 124.2 Reference range for hematocrit and hemoglobin concentration during 1st
28 days of life. A and B, Late preterm and term infants (35-42 wk gestation). C and D,
Preterm infants (29-34 wk gestation). The reference ranges are shown for hematocrit (A
and C ) (41,957 patients) and blood hemoglobin (B and D ) (39,559 patients) during the
28 days after birth. Values were divided into 2 groups (A/B and C/D ) on the basis of
gestational age at delivery. Patients were excluded when their diagnosis included
abruption, placenta previa, or fetal anemia, or when a blood transfusion was given.
Analysis was not possible for patients <29 wk gestation because virtually all these had
repeated phlebotomy and erythrocyte transfusions. (From Jopling J, Henry E,
Wiedmeier SE, Christensin RD: Reference ranges for hematocrit and blood hemoglobin
concentration during the neonatal period: data from a multihospital health care system,
Pediatrics 123(2):e333–e337, 2009.)
Table 124.1
Differential Diagnosis of Neonatal Anemia
BLOOD LOSS ↑ RBC ↓ RBC PRODUCTION
Iatrogenic blood loss (phlebotomy) DESTRUCTION Physiologic anemia and anemia of prematurity
Placental hemorrhage Immune- Infection (rubella, CMV, parvovirus B19)
Placental previa Mediated Bone marrow suppression (acute stress in
Injury of umbilical or placental vessels Hemolysis perinatal period)
Fetomaternal transfusion Rh incompatibility Hemoglobinopathy (γ-globin mutation, unstable
Fetoplacental transfusion ABO incompatibility β-hemoglobinopathy, α-thalassemia major)
Twin-twin transfusion Minor antigen Bone marrow suppression (CMV, EBV)
Acute perinatal hemorrhage (e.g., incompatibility Diamond Blackfan anemia
cesarean birth, other obstetric trauma) RBC Membrane Schwachman-Diamond syndrome
Chronic in utero blood loss Disorders Congenital dyserythropoietic anemia
Hereditary Fanconi anemia
spherocytosis Pearson syndrome
Hereditary Congenital leukemia
elliptocytosis
Hereditary
pyropoikilocytosis
Hereditary
stomatocytosis
RBC Enzyme
Disorders
G6PD deficiency
Pyruvate kinase
deficiency
CMV, Cytomegalovirus, EBV, Epstein-Barr virus; G6PD, glucose-6-phosphate dehydrogenase;
RBC, red blood cell.
FIG. 124.3 Acid elution technique of Kleithauer (Kleihauer-Betke test). Fetal red blood
cells stain with eosin and appear dark . Adult RBCs do not stain and appear as
“ghosts.” (From Liley HG, Gardener G, Lopriore E, Smits-Wintjens V: Immune hemolytic
disease. In Orkin SH, Nathan DG, Ginsburg D, et al, editors: Nathan and Oski's
hematology and oncology of infancy and childhood, ed 8, Philadelphia, 2015, Elsevier,
Fig 3-2.)
FIG. 124.5 Evaluation of neonate with problematic jaundice of unclear cause. Not all
neonates who receive phototherapy for 2 days or more have hemolytic jaundice.
However, if hemolytic jaundice is suspected, this algorithm for stepwise evaluation of
the cause might be useful. CBC, Complete blood count; DAT, direct antiglobulin test;
EMA, eosin 5-maleimide; G6PD, glucose 6-phosphate dehydrogenase; HS, hereditary
spherocytosis; MCHC, mean corpuscular hemoglobin concentration; MCV, mean
corpuscular volume; RBC, red blood cell. (From Christensen RD: Neonatal erythrocyte
disorders. In Gleason CA, Juul SE, editors: Avery's diseases of the newborn, ed 10,
Philadelphia, 2018, Elsevier, Fig 81-15.)
Table 124.2
Table 124.3
Morphologic Abnormalities of Erythrocytes From Neonates
With Jaundice
Table 124.4
Suggested Transfusion Thresholds
When the decision to transfuse has been made, the appropriate blood product
should be selected and a safe volume of blood should be transfused at a safe rate.
It is important to transfuse packed erythrocytes (PRBCs) to all neonates in the
form of leukocyte-reduced or CMV-seronegative PRBCs, to reduce the risk of
CMV transmission. Irradiation of PRBCs removes the risk of transfusion-
associated graft-versus-host disease (GVHD) but does not eliminate the risk of
CMV transmission. The volume of transfusion should achieve the intended
therapeutic goal while limiting blood product exposure. Typical transfusion
protocols choose a transfusion volume ranging from 10-20 mL/kg. There are no
clear data to favor a specific amount, but lower volumes exposure infants to risks
unnecessarily while higher volumes may cause fluid overload. One logical goal
is to target a specific goal hemoglobin (Hb) concentration. The following
commonly used shorthand equation can provide a good estimate of required
blood volume, which usually results in a transfusion volume within the 10-20
mL/kg range:
Erythropoietin
Because of the low physiologic levels of erythropoietin in neonates, the role of
recombinant human erythropoietin (rhEPO ) has been investigated for the
treatment of anemia in neonates, particularly VLBW infants. A Cochrane review
documented that rhEPO is associated with a significant reduction in the number
of blood transfusions per infant, but also a significantly increased risk of
retinopathy of prematurity. There were no differences in mortality or other
neonatal morbidities among infants who did or did not receive rhEPO. Because
of these limited benefits and potential serious risks of early rhEPO therapy, there
is currently no strong indication for the routine use of rhEPO in infants with
anemia, although it should be considered in individual settings.
anemia is difficult to predict based on previous obstetric history, amniotic fluid
bilirubin determinants, or maternal antibody titer. Kell-alloimmunized infants
often have inappropriately low numbers of circulating reticulocytes caused by
erythroid suppression, and even low maternal titers of anti-Kell antibodies may
cause significant hypoproliferative anemia. Table 124.5 summarizes the clinical
characteristics of hemolytic disease caused by Rh, ABO, and Kell antigen
systems. There are no specific pharmacologic therapies available to prevent
sensitization caused by any blood group other than RhD. As with cases of Rh
and ABO incompatibility, exchange transfusion may be indicated for severe
hyperbilirubinemia or severe anemia in infants with HDFN caused by minor
antigen incompatibility.
Table 124.5
Hemolytic Disease of the Fetus and Newborn
Rh ABO KELL
BLOOD GROUPS
Mother Rh-negative O (occasionally B) K1-negative
Infant Rh-positive (D is most A (sometimes B) K1-positive
common)
CLINICAL FEATURES
Occurrence in 5% 40–50% Rare
firstborn
Severity in Predictable Difficult to predict Somewhat predictable
subsequent
pregnancies:
Stillbirth/hydrops Frequent (less with Rare 10%
Rh-immunoglobulin
use)
Severe anemia Frequent Rare Frequent
Jaundice Prominent, severe Mild-moderate Mild
LABORATORY TESTS
Direct Positive Positive or negative Positive or negative
antiglobulin test
(infant)
Reticulocyte Elevated Elevated Variable
count
Red blood cell Usually detectable May not be Usually detectable
(RBC) antibodies Antibody titers detectable Antibody titers may not correlate with fetal
(mother) may help predict Antibody titers disease; fetus can be affected at titers lower
severity of fetal may not than for Rh-mediated hemolysis.
disease. correlate with
fetal disease.
necessary for its full coagulation effects. In the absence of carboxylation, such
factors form PIVKA (proteins induced in vitamin K absence), which have
greatly reduced function; these can be measured and represent a sensitive marker
for vitamin K status. In contrast, factors V and VIII, fibrinogen, bleeding time,
clot retraction, and platelet count and function are normal for maturity.
Classically, vitamin K deficiency bleeding occurs early in the newborn period,
typically between day 2 and 7 of life, and most often in exclusively
breastfeeding infants who did not receive vitamin K prophylaxis at birth. Severe
vitamin K deficiency is also more common in premature infants. This
pathogenesis occurs from a lack of free vitamin K from the mother, coupled with
absence of bacterial intestinal flora normally responsible for the synthesis of
vitamin K. Breast milk is a poor source of vitamin K, which explains why
hemorrhagic complications are more frequent in exclusively breastfed than in
mixed-fed or formula-fed infants. This classic form of hemorrhagic disease of
the newborn, which is responsive to (and entirely prevented by) exogenous
vitamin K therapy, should be distinguished from rare congenital deficiencies of
clotting factors that are unresponsive to vitamin K, which can occur in otherwise
well-appearing infants (see Chapter 503 ).
Early-onset vitamin K deficiency bleeding (after birth but in 1st 24 hr) occurs
if the mother has been treated chronically with certain drugs (e.g., anticoagulant
warfarin, anticonvulsant phenytoin or phenobarbital, cholesterol-lowering
medication) that interfere with vitamin K absorption or function. These infants
can have severe bleeding, which is usually corrected promptly by vitamin K
administration, although some have a poor or delayed response. If a mother is
known to be receiving such drugs late in gestation, an infant PT should be
measured using cord blood, and the infant immediately given 1-2 mg of vitamin
K intravenously. If PT is greatly prolonged and fails to improve, or in the
presence of significant hemorrhage, 10-15 mL/kg of fresh-frozen plasma should
be administered. In contrast, late-onset vitamin K deficiency bleeding (after 2
wk of life) is usually associated with conditions that feature malabsorption of the
fat-soluble vitamin K, such as cystic fibrosis, neonatal hepatitis, or biliary
atresia, and bleeding can be severe (Table 124.6 ).
Table 124.6
Vitamin K Deficiency Bleeding (Hemorrhagic Disease of the
Newborn)
CLASSIC
EARLY-ONSET DISEASE LATE-ONSET DISEASE
DISEASE
Age 0-24 hr 2-7 days 1-6 mo
Potential sites Cephalohematoma Gastrointestinal Intracranial
of Subgaleal Ear-nose-throat- Gastrointestinal
hemorrhage mucosal
Intracranial Intracranial Cutaneous
Gastrointestinal Post-circumcision Ear-nose-throat-mucosal
Umbilicus Cutaneous Injection sites
Intraabdominal Injection sites Thoracic
Etiology/risks Maternal drugs (phenobarbital, phenytoin, Vitamin K Cholestasis: malabsorption of
warfarin, rifampin, isoniazid) that interfere deficiency vitamin K (biliary atresia, cystic
with vitamin K levels or absorption Exclusive fibrosis, hepatitis)
breastfeeding
Inherited coagulopathy Abetalipoprotein deficiency
Idiopathic in Asian breastfed
infants
Warfarin ingestion
Prevention Avoidance of high-risk medications Prevented by Prevented by parenteral and
Possibly antenatal vitamin K to parenteral high-dose oral vitamin K during
treatment of mother (20 mg) before vitamin K at periods of malabsorption or
birth and postnatal administration to birth cholestasis
infant soon after birth Oral vitamin
K regimens
require
repeated
dosing.
Incidence Very rare ~2% if infant not Dependent on primary disease
given vitamin K
soon after birth
Table 124.7
Conditions Associated With Nonimmune Hydrops
CARDIOVASCULAR HEMATOLOGIC
Malformation α-Thalassemia
Left heart hypoplasia Fetomaternal transfusion
Atrioventricular canal defect Parvovirus B19 infection
Right heart hypoplasia In utero hemorrhage
Closure of foramen ovale G6PD deficiency
Single ventricle Red cell enzyme deficiencies
Transposition of the great vessels THORACIC
Ventral septal defect Congenital cystic adenomatoid malformation of lung
Atrial septal defect Diaphragmatic hernia
Tetralogy of Fallot Intrathoracic mass
Ebstein anomaly Pulmonary sequestration
Premature closure of ductus Chylothorax
Truncus arteriosus Airway obstruction
Tachyarrhythmia Pulmonary lymphangiectasia
Atrial flutter Pulmonary neoplasia
Paroxysmal atrial tachycardia Bronchogenic cyst
Wolff-Parkinson-White syndrome INFECTIONS
Supraventricular tachycardia
Bradyarrhythmia Cytomegalovirus
Other arrhythmias Toxoplasmosis
High-output failure Parvovirus B19 (fifth disease)
Neuroblastoma Syphilis
Sacrococcygeal teratoma Herpes
Large fetal angioma Rubella
Placental chorioangioma MALFORMATION SEQUENCES
Umbilical cord hemangioma Noonan syndrome
Cardiac rhabdomyoma Arthrogryposis
Other cardiac neoplasia Multiple pterygia
Cardiomyopathy Neu-Laxova syndrome
CHROMOSOMAL Pena-Shokeir syndrome
45,X Myotonic dystrophy
Trisomy 21 Saldino-Noonan syndrome
Trisomy 18 METABOLIC
Trisomy 13 Gaucher disease
18q+ GM1 gangliosidosis
13q− Sialidosis
45,X/46,XX Mucopolysaccharide disorders
Triploidy
URINARY
Other
Urethral stenosis or atresia
CHONDRODYSPLASIAS
Posterior urethral valves
Thanatophoric dwarfism
Congenital nephrosis (Finnish)
Short rib polydactyly
Prune-belly syndrome
Hypophosphatasia
GASTROINTESTINAL
Osteogenesis imperfecta
Achondrogenesis Midgut volvulus
TWIN PREGNANCY Malrotation of the intestines
Twin-twin transfusion syndrome Duplication of the intestinal tract
Acardiac twin Meconium peritonitis
OTHER Hepatic fibrosis
Familial hemophagocytic lymphohistiocytosis Cholestasis
Fetal akinesia syndromes Biliary atresia
Congenital leukemia Hepatic vascular malformations
Infantile arterial calcification syndrome
Maternal diabetes
Lymphatic disorders
IPEX
Idiopathic
IPEX, Immune dysregulation polyendocrinopathy enteropathy, X-linked.
Adapted from Wilkins I: Nonimmune hydrops. In Creasy RK, Resnick R, Iams JD, et al, editors:
Creasy & Resnik's maternal-fetal medicine, ed 7, Philadelphia, 2014, Elsevier (Box 37-1).
FIG. 126.1 Neonatal abstinence score used for the assessment of infants displaying
neonatal abstinence syndrome. Evaluator should check sign or symptom observed at
various time intervals. Add scores for total at each evaluation. (Adapted from Finnegan
LP, Kaltenbach K. The assessment and management of neonatal abstinence syndrome.
In Hoekelman RA et al, editors: Primary pediatric care, ed 3, St Louis, 1992, Mosby, p
1367.)
Identifying which infants are at risk for NAS before discharge is important
because of the late onset of signs. Universal maternal screening for drug use is
recommended by the American College of Obstetricians and Gynecologists
(ACOG), and maternal consent should be obtained if drug testing is indicated.
Universal maternal drug testing has been shown to improve identification of
infants at risk for NAS but is more expensive and may not be helpful in states
with punitive legislation. Maternal testing is preferred over infant testing
because results are available promptly, typically by the time the infant is
delivered. Testing mothers on admission to the hospital can also exclude
iatrogenic exposure. Infant urine, meconium and umbilical cord testing are also
used to help identify infants at risk for withdrawal and identify more distant use
that can be given twice a day after loading doses, and a pharmacokinetic weight-
based weaning protocol is available. Sublingual buprenorphine has been
proposed as an alternate treatment. Buprenorphine and some methadone
formulations contain high ethanol levels, which may be deleterious to the infant.
Following a stringent NAS protocol with guidelines on initiation and weaning
has been shown to decrease both length of stay and number of opioid treatment
days and may be as important as which primary opioid is used for first-line
treatment.
Table 126.1
Medications Used in Pharmacologic Treatment of Neonatal
Abstinence Syndrome (NAS)
ADD
INITIAL DOSING
DRUG WEANING SCHEDULE ADJUVANT
DOSING INCREASES
THERAPY
Morphine 0.05 mg/kg/dose Increase dose 10– 10% of stabilizing dose q24h >1
q3h 20% mg/kg/day of
morphine
Unable to
wean for 2
days
Methadone 0.1 mg/kg/dose Increase to q4h if 0.7 mg/kg/dose q12h × 2 doses, Unable to wean
q6h for 4 doses unable to capture then 0.05 mg/kg/dose q12h × 2; for 2 days
0.04 mg/kg/dose q12h × 2;
0.03 mg/kg/dose q12h × 2;
0.02 mg/kg/dose q12h × 2;
0.01 mg/kg/dose q12h × 2;
0.01 mg/kg/dose q24h × 1
Buprenorphine 4 µg/kg q8h 2 µg/kg until 3 µg/kg/dose q8h × 3 doses; Unable to wean
maximum of 15 2 µg/kg/dose q8h × 3; for 2 days
µg/kg 2 µg/kg/dose q8h × 2;
2 µg/kg/dose q24h × 1
Phenobarbital 20 mg/kg — 5 mg/kg daily N/A
Clonidine 1.5 µg/kg/dose 25% dose 10% every day N/A
q3h escalation q24hr
N/A, Not available; q24h, every 24 hours.
Table 126.2
Diagnostic Features of Fetal Alcohol Spectrum Disorders
(FASDs)
DEFICIENT BRAIN
GROWTH,
FACIAL ABNORMAL NEUROBEHAVIORAL
TYPE OF FASD GROWTH
DYSMORPHOLOGY MORPHOGENESIS, FEATURES
OR ABNORMAL
NEUROPHYSIOLOGY
Fetal alcohol ≥2 of the Height OR Head circumference With cognitive
syndrome (FAS) following: and/or ≤10th centile impairment:
Short palpebral weight Structural brain Evidence of global
fissures (≤10th ≤10th anomalies impairment (general
centile) centile Recurrent nonfebrile conceptual ability
Thin vermilion seizures ≥1.5 SD below the
border of upper lip mean
Smooth philtrum or
Cognitive deficit in
at least 2
neurobehavioral
domains ≥1.5 SD
below the mean
With behavioral
impairment without
cognitive
impairment:
Evidence of
behavioral deficit in
at least 2 domains
≥1.5 SD below the
mean
Partial FAS (pFAS) ≥2 of the Height OR Head circumference With cognitive
following: and/or ≤10th centile impairment:
Short palpebral weight Structural brain Evidence of global
fissures (≤10th ≤10th anomalies impairment (general
centile) centile Recurrent nonfebrile conceptual ability
Thin vermilion seizures ≥1.5 SD below the
border of upper lip mean
Smooth philtrum or
Cognitive deficit in
at least 2
neurobehavioral
domains ≥1.5 SD
below the mean
With behavioral
impairment without
cognitive
impairment:
Evidence of
behavioral deficit in
at least 2 domains
≥1.5 SD below the
mean
Alcohol-related — — — With cognitive
neurodevelopmental impairment:
disorder (ARND) Evidence of global
impairment (general
conceptual ability
≥1.5 SD below the
mean
or
Cognitive deficit in
at least 2
neurobehavioral
domains ≥1.5 SD
below the mean
With behavioral
impairment without
cognitive
impairment:
Evidence of
behavioral deficit in
at least 2 domains
≥1.5 SD below the
mean
Alcohol-related — — One or more specific —
birth defect major malformations
(ARBD) demonstrated in animal
models and human
studies to be the result of
prenatal alcohol exposure
Neurobehavioral — — — Neurocognitive
disorder associated impairment (1)
with prenatal Global intellect
alcohol exposure Executive
(ND-PAE) functioning
Learning
Memory
Visual-spatial
reasoning
Impaired self-
regulation (1)
Mood or behavior
Attention
Impulse control
Impairments in
adaptive
functioning (2)
Language
Social
communication and
interaction
Daily living skills
Motor skills
Table 127.1
Morbidity in Infants of Diabetic Mothers
• Congenital anomalies
• Heart failure and septal hypertrophy of heart
• Surfactant deficiency, respiratory distress syndrome, transient tachypnea of the newborn, persistent pulmonary
hypertension
• Hyperbilirubinemia
• Hypoglycemia, hypocalcemia, hypomagnesemia
• Macrosomia, nerve injury related to birth trauma
• Renal vein thrombosis
• Small left colon
• Unexplained intrauterine demise
• Polycythemia
• Visceromegaly
• Predisposition to later-life obesity, insulin resistance, and diabetes
From Devaskar SU, Garg M: Disorders of carbohydrate metabolism in the neonate. In Martin RJ,
Fanaroff AA, Walsh MC, editors: Fanaroff & Martin's neonatal-perinatal medicine, ed 10,
Philadelphia, 2015, Elsevier (Box 95-3).
Table 128.1
Table 128.2
Examples of Malformations with Distinct Causes, Clinical
Features, and Pathogenesis
SELECTED
DISORDER CAUSE/INHERITANCE CLINICAL PATHOGENESIS
FEATURES
Spondylocostal Mendelian; autosomal Abnormal vertebral Deleterious sequence variants in DLL3
dysostosis recessive and rib and other genes
syndrome segmentation
Rubinstein-Taybi Autosomal dominant Intellectual Deleterious sequence variants in CBP and
syndrome disability EP300
Broad thumbs
and halluces;
valgus
deviation of
these digits
Hypoplastic
maxillae
Prominent nose
and columella
Congenital
heart disease
X-linked X-linked Male: severe Deleterious sequence variants in DCX
lissencephaly intellectual
disability,
seizures
Female:
variable
Aniridia Autosomal dominant Absent iris or Deleterious sequence variants in PAX6
iris/foveal
hypoplasia
Waardenburg Autosomal dominant Deafness Deleterious sequence variants in PAX3
syndrome, type I White forelock
Wide-spaced
eyes
Iris
heterochromia
and/or pale skin
pigmentation
Holoprosencephaly Loss of function or Microcephaly SHH, multiple other genes
heterozygosity for Cyclopia
multiple genes Single central
incisor
Velocardiofacial Microdeletion Congenital heart TBX1 haploinsufficiency/mutations;
syndrome 22q11.2 disease, including haploinsufficiency for other genes in the
conotruncal defects deleted interval also contributes to the
Cleft palate phenotype.
T-cell defects
Facial anomalies
Down syndrome Additional copy of Intellectual Increase in dosage of an estimated 250
chromosome 21 (trisomy disability genes on chromosome 21
21) Characteristic
dysmorphic features
Congenital heart
disease
Increased risk of
leukemia
Alzheimer disease
Neural tube defects Multifactorial Meningomyelocele Defects in folate sensitive enzymes or
folic acid uptake
Fetal alcohol Teratogenic Microcephaly Ethanol toxicity to developing brain
syndrome Developmental
delay
Facial abnormalities
Behavioral
abnormalities
Retinoic acid Teratogenic Microtia Isotretinoin effects on neural crest and
embryopathy Congenital heart branchial arch development
disease
Table 128.3
Causes of Congenital Malformations
MONOGENIC
X-linked hydrocephalus
Achondroplasia
Ectodermal dysplasia
Apert syndrome
Treacher Collins syndrome
CHROMOSOMAL ABERRATIONS and COPY NUMBER VARIANTS
Trisomy 21, 18, 13
XO, XXY
Deletions 4p−, 5p−, 7q−, 13q−, 18p−, 18q−, 22q−
Prader-Willi syndrome (70% of affected patients have deletion of chromosome 15 q11.2-q13)
MATERNAL INFECTION
Intrauterine infections (e.g., herpes simplex virus, cytomegalovirus, varicella-zoster virus, rubella virus, Zika
virus, toxoplasmosis)
MATERNAL ILLNESS
Diabetes mellitus
Phenylketonuria
Hyperthermia
UTERINE ENVIRONMENT
Deformation
Uterine pressure, oligohydramnios: clubfoot, torticollis, congenital hip dislocation, pulmonary hypoplasia, 7th
nerve palsy
Disruption
Amniotic bands, congenital amputations, gastroschisis, porencephaly, intestinal atresia
Twinning
ENVIRONMENTAL AGENTS
Polychlorinated biphenyls
Herbicides
Mercury
Alcohol
MEDICATIONS
Thalidomide
Diethylstilbestrol
Phenytoin
Warfarin
Cytotoxic drugs
Paroxetine
Angiotensin-converting enzyme inhibitors
Isotretinoin (vitamin A)
D -Penicillamine
Valproic acid
Mycophenolate mofetil
UNKNOWN ETIOLOGIES
Neural tube defects, such as anencephaly and spina bifida
Cleft lip/palate
Pyloric stenosis
SPORADIC SEQUENCE COMPLEXES
VATER/VACTERL sequence (vertebral defects, anal atresia, cardiac defects, tracheoesophageal fistula with
esophageal atresia, radial and renal anomalies)
Pierre Robin sequence
NUTRITIONAL
Neural tube defects due to low folic acid
From Behrman RE, Kliegman RM, editors: Nelson's essentials of pediatrics, ed 4, Philadelphia,
2002, Saunders.
Table 128.5
0.5% have 3. If 2 minor anomalies are present, the probability of an underlying syndrome or a
major anomaly (congenital heart disease, renal, central nervous system, limbic) is 5-fold that in
the general population. If 3 minor anomalies are present, there is a 20–30% probability of a major
anomaly.
From Kliegman RM, Greenbaum LA, Lye PS: Practical strategies in pediatric diagnosis and
therapy , ed 2, Philadelphia, 2004, Elsevier Saunders.
FIG. 128.10 Frequency of major malformations in relation to the number of
minor anomalies detected in a given newborn baby. (From Jones KJ:
Smith's recognizable patterns of human malformation, ed 6, Philadelphia,
2006, Saunders.)
Imaging Studies
Imaging studies can be critical in diagnosing an underlying genetic etiology. If
short stature or disproportionate stature (e.g., long trunk and short limbs) is
noted, a full skeletal survey with radiographs should be performed. The skeletal
survey can detect anomalies in bone number or structure that can be used to
narrow the differential diagnosis. When there are abnormal neurologic signs or
symptoms, such as microcephaly or hypotonia, brain imaging can be indicated.
Other studies, such as echocardiography and renal ultrasonography, can also be
useful to identify additional major or minor malformations that may serve as
diagnostic clues.
associated with birth defects and multiple congenital anomalies.
Table 128.7
Table 129.1
Nonbacterial Causes of Systemic Neonatal Infections
VIRUSES
Adenovirus
Cytomegalovirus (CMV)
Enteroviruses
Parechoviruses
Hepatitis B and C viruses
Herpes simplex virus (HSV)
Human immunodeficiency virus (HIV)
Parvovirus
Rubella virus
Varicella-zoster virus (VZV)
MYCOPLASMA
Mycoplasma hominis
Ureaplasma urealyticum
FUNGI
Candida spp.
Malassezia spp.
PROTOZOA
Plasmodia
Toxoplasma gondii
Trypanosoma cruzi
FIG. 129.2 Relative importance of neonatal viral infections related to the timing of
acquisition of infection. Viruses are listed in declining order of importance relative to
prenatal, perinatal (intrapartum), and postnatal timing of typical infection. Some
neonatal virus infections (e.g., cytomegalovirus) can be substantial causes of disease
whether acquired during gestation or acquired postpartum, whereas others (e.g.,
respiratory syncytial virus) are typically acquired in the postnatal period. EBV, Epstein-
Barr virus; HHV, human herpesvirus; HIV, human immunodeficiency virus; HSV, herpes
simplex virus; LCMV, lymphocytic choriomeningitis virus. (From Schleiss MR, Marsh
KJ: Viral infections of the fetus and newborn. In Gleason CA, Juul SE, editors: Avery's
diseases of the newborn, ed 10, Philadelphia, 2018, Elsevier, Fig 37-1.)
Table 129.2
Period of Transmission of Selected Viruses to the Fetus or
Newborn Infant
Table 129.3
Etiologic Agents of Neonatal Pneumonia According to
Timing of Acquisition
TRANSPLACENTAL
Cytomegalovirus (CMV)
Herpes simplex virus (HSV)
Mycobacterium tuberculosis
Rubella virus
Treponema pallidum
Varicella-zoster virus (VZV)
Listeria monocytogenes
PERINATAL
Anaerobic bacteria
Chlamydia
CMV
Enteric bacteria
Group B streptococci
Haemophilus influenzae
HSV
Listeria monocytogenes
Mycoplasma
POSTNATAL
Adenovirus
Candida spp.*
Coagulase-negative staphylococci
CMV
Enteric bacteria*
Enteroviruses
Influenza viruses A, B
Parainfluenza
Pseudomonas *
Respiratory syncytial virus (RSV)
Staphylococcus aureus
Mycobacterium tuberculosis
Legionella
* More likely with mechanical ventilation or indwelling catheters, or after abdominal surgery.
The most common bacterial causes of neonatal meningitis are GBS, E. coli,
and L. monocytogenes. S. pneumoniae, other streptococci, nontypable H.
influenzae, both coagulase-positive and coagulase-negative staphylococci,
Klebsiella, Enterobacter, Pseudomonas, Treponema pallidum, and
Mycobacterium tuberculosis infection involving the central nervous system
(CNS) may also result in meningitis.
Table 129.4
Rates of Early-Onset Sepsis Per 1,000 Live Births*
BIRTHWEIGHT (g)
401-1,500 1,501-2,500 >2,500 All
All 10.96 1.38 0.57 0.98
Group B streptococci 2.08 0.38 0.35 0.41
Escherichia coli 5.09 0.54 0.07 0.28
* NICHD Neonatal Research Network/CDC Surveillance Study of Early-Onset Sepsis.
Adapted from Stoll BJ, Hansen NI, Sanchez PJ, et al: Early onset neonatal sepsis: the burden of
group B streptococcal and E. coli disease continues, Pediatrics 127(5):817–826, 2011.
The incidence of meningitis is 0.2-0.4 per 1,000 live births in newborn infants
and is higher in preterm infants. Bacterial meningitis may be associated with
sepsis or may occur as a local meningeal infection. Up to one third of VLBW
infants with late-onset meningitis have negative blood culture results . The
discordance between results of blood and cerebrospinal fluid (CSF) cultures
suggests that meningitis may be underdiagnosed among VLBW infants and
emphasizes the need for culture of CSF in VLBW infants when late-onset sepsis
and obstetric risk factors (prematurity, prolonged ruptured membranes, maternal
chorioamnionitis). Signs and symptoms in the neonate are often subtle and
nonspecific. Temperature instability, tachypnea, lethargy, and poor feeding are
common initial signs and should raise suspicion for systemic or focal infection
(Table 129.5 ).
Table 129.5
Initial Signs and Symptoms of Infection in Newborn Infants
GENERAL
Fever, temperature instability
“Not doing well”
Poor feeding
Edema
GASTROINTESTINAL SYSTEM
Abdominal distention
Vomiting
Diarrhea
Hepatomegaly
RESPIRATORY SYSTEM
Apnea, dyspnea
Tachypnea, retractions
Flaring, grunting
Cyanosis
RENAL SYSTEM
Oliguria
CARDIOVASCULAR SYSTEM
Pallor; mottling; cold, clammy skin
Tachycardia
Hypotension
Bradycardia
CENTRAL NERVOUS SYSTEM
Irritability, lethargy
Tremors, seizures
Hyporeflexia, hypotonia
Abnormal Moro reflex
Irregular respirations
Full fontanel
High-pitched cry
HEMATOLOGIC SYSTEM
Jaundice
Splenomegaly
Pallor
Petechiae, purpura
Bleeding
Bacterial Sepsis
Neonates with bacterial sepsis may have either nonspecific manifestations or
focal signs of infection (Table 129.5 ), including temperature instability,
hypotension, poor perfusion with pallor and mottled skin, metabolic acidosis,
tachycardia or bradycardia, apnea, respiratory distress, grunting, cyanosis,
irritability, lethargy, seizures, feeding intolerance, abdominal distention,
jaundice, petechiae, purpura, and bleeding. Table 129.6 lists World Health
Organization international criteria for bacterial sepsis. The initial manifestation
may involve only limited symptomatology and only one system, such as apnea
alone or tachypnea with retractions, or tachycardia, or the infant may present
with an acute catastrophic manifestation with multiorgan dysfunction and shock.
Infants should be reevaluated over time to determine whether the symptoms
have progressed from mild to severe. Later complications of sepsis include
respiratory failure, pulmonary hypertension, cardiac failure, shock, renal failure,
liver dysfunction, cerebral edema or thrombosis, adrenal hemorrhage and/or
insufficiency, bone marrow dysfunction (neutropenia, thrombocytopenia,
anemia), and disseminated intravascular coagulopathy (DIC).
Table 129.6
Clinical Criteria for the Diagnosis of Sepsis in the
International Setting
IMCI AND WHO CRITERIA FOR SEVERE INFECTIONS IN CHILDREN
Neurologic: convulsions, drowsy or unconscious, decreased activity, bulging fontanel
Respiratory: respiratory rate >60 breaths/min, grunting, severe chest indrawing, central cyanosis
Cardiac: poor perfusion, rapid and weak pulse
Gastrointestinal: jaundice, poor feeding, abdominal distention
Dermatologic: skin pustules, periumbilical erythema or purulence
Musculoskeletal: edema or erythema overlying bones or joints
Other: temperature >37.7°C (99.9°F; or feels hot) or <35.5°C (95.9°F; or feels cold)
IMCI, Integrated Management of Childhood Illness; WHO, World Health Organization.
Adapted from WHO: Pocket book of hospital care for children: guidelines for the management of
common childhood illnesses, ed 2, Geneva, 2013, WHO, pp 45–69.
http://www.who.int/maternal_child_adolescent/documents/child_hospital_care/en/ .
Table 129.7
Serious Systemic Illness in Newborns: Differential
Diagnosis of Neonatal Sepsis
CARDIAC
Congenital: hypoplastic left heart syndrome, other structural disease, persistent pulmonary hypertension of the
newborn (PPHN)
Acquired: myocarditis, hypovolemic or cardiogenic shock, PPHN
GASTROINTESTINAL
Necrotizing enterocolitis
Spontaneous gastrointestinal perforation
Midgut volvulus
Hepatic failure (inborn errors of metabolism, neonatal iron storage disease)
HEMATOLOGIC
Neonatal purpura fulminans
Immune-mediated thrombocytopenia
Immune-mediated neutropenia
Severe anemia
Malignancies (congenital leukemia)
Langerhans cell histiocytosis
Hereditary clotting disorders
Familial hemophagocytosis syndrome
METABOLIC
Hypoglycemia
Adrenal disorders: adrenal hemorrhage, adrenal insufficiency, congenital adrenal hyperplasia
Inborn errors of metabolism: organic acidurias, lactic acidoses, urea cycle disorders, galactosemia
NEUROLOGIC
Intracranial hemorrhage: spontaneous, caused by child abuse
Hypoxic-ischemic encephalopathy
Neonatal seizures
Infant botulism
RESPIRATORY
Respiratory distress syndrome
Aspiration pneumonia: amniotic fluid, meconium, or gastric contents
Lung hypoplasia
Tracheoesophageal fistula
Transient tachypnea of the newborn
Table 129.8
Definitions of Systemic Inflammatory Respiratory
Response Syndrome (SIRS) and Sepsis in Pediatric
Patients
SIRS: the systemic inflammatory response to a variety of clinical insults, manifested by 2 or more of the
following conditions:
Temperature instability <35°C (95°F) or >38.5°C (101.3°F)
Respiratory dysfunction:
Tachypnea >2 SD above the mean for age
Hypoxemia (PaO 2 <70 mm Hg on room air)
Cardiac dysfunction:
Tachycardia >2 SD above the mean for age
Delayed capillary refill >3 sec
Hypotension >2 SD below the mean for age
Perfusion abnormalities:
Oliguria (urine output <0.5 mL/kg/hr)
Lactic acidosis (elevated plasma lactate and/or arterial pH <7.25)
Altered mental status
Sepsis: the systemic inflammatory response to an infectious process
From Adams-Chapman I, Stoll BJ: Systemic inflammatory response syndrome, Semin Pediatr
Infect Dis 12:5–16, 2001.
Temperature Instability
Laboratory Findings
Maternal history and infant signs should guide diagnostic evaluation (Table
129.9 ). Additionally, signs of systemic infection in newborn infants may be
unrevealing, so laboratory investigation plays a particularly important role in
diagnosis. Cultures and cell counts are obtained from blood and urine. CSF
should be sent for Gram stain, routine culture, cell count with differential, and
protein/glucose concentrations. Surface swabs, blood, and CSF are often
obtained for HSV testing. Except for culture and directed pathogen testing, no
single laboratory test is completely reliable for diagnosis of invasive infection in
the newborn. Complete blood count may demonstrate elevated or decreased
WBC count, often with a shift toward more immature forms. Thrombocytopenia
can be seen in systemic bacterial or viral infection. Hyponatremia, acidosis, and
other electrolyte abnormalities can be seen. Hyperbilirubinemia is nonspecific
but may be an indication of systemic infection. Elevated serum transaminases
may be a clue to systemic HSV or enterovirus infection.
Table 129.9
Evaluation of a Newborn for Infection or Sepsis
HISTORY (SPECIFIC RISK FACTORS)
Maternal infection during gestation or at parturition (type and duration of antimicrobial therapy):
Urinary tract infection
Chorioamnionitis
Maternal colonization with group B streptococci, Neisseria gonorrhoeae , herpes simplex
Low gestational age/birthweight
Multiple birth
Duration of membrane rupture
Complicated delivery
Fetal tachycardia (distress)
Age at onset (in utero, birth, early postnatal, late)
Location at onset (hospital, community)
Medical intervention:
Vascular access
Endotracheal intubation
Parenteral nutrition
Surgery
EVIDENCE OF OTHER DISEASES *
Congenital malformations (heart disease, neural tube defect)
Respiratory tract disease (respiratory distress syndrome, aspiration)
Necrotizing enterocolitis
Metabolic disease (e.g., galactosemia)
EVIDENCE OF FOCAL OR SYSTEMIC DISEASE
General appearance, neurologic status
Abnormal vital signs
Organ system disease
Feeding, stools, urine output, extremity movement
LABORATORY STUDIES
Evidence of Infection
Culture from a normally sterile site (blood, CSF, other)
Demonstration of a microorganism in tissue or fluid
Molecular detection (blood, urine, CSF) by specific PCR and/or 16S ribosomal DNA
Maternal or neonatal serology (syphilis, toxoplasmosis)
Evidence of Inflammation
Leukocytosis, increased immature/total neutrophil count ratio
Acute-phase reactants: C-reactive protein, erythrocyte sedimentation rate, procalcitonin
Cytokines: interleukin-6, interleukin-B, tumor necrosis factor
Pleocytosis in CSF or synovial or pleural fluid
Disseminated intravascular coagulation: fibrin degradation products, D-dimer
Evidence of Multiorgan System Disease
Metabolic acidosis: pH, PCO 2
Pulmonary function: PO 2 , PCO 2
Renal function: blood urea nitrogen, creatinine
Hepatic injury/function: bilirubin, alanine transaminase, aspartate transaminase, ammonia, prothrombin time,
partial thromboplastin time
Bone marrow function: neutropenia, anemia, thrombocytopenia
* Diseases that increase the risk of infection or may overlap with signs of sepsis.
Various serum biomarkers have been investigated for their ability to identify
infants with serious bacterial infection (SBI). An immature-to-total phagocyte
count (I/T ratio) (≥0.2) has the best sensitivity of the neutrophil indices for
predicting neonatal sepsis. After the newborn period, serum C-reactive protein
(CRP) and procalcitonin have demonstrated reasonable sensitivity and
specificity for SBI. CRP may be monitored in newborn infants to assess response
to therapy. Their value in the initial diagnosis of sepsis in the newborn period has
yet to be clarified, as does the value of these biomarkers in determining optimal
length of empirical therapy in infants with negative cultures. Cytokines (both
proinflammatory cytokines such as interleukin (IL)-6 and tumor necrosis factor-
α and antiinflammatory cytokines such as IL-4 and IL-10), chemokines, and
other biomarkers are increased in infected infants. Elevations of serum amyloid
A and the cell surface antigen CD64 also have high sensitivity for identifying
infants with sepsis. Chest radiography is generally not indicated in infants
without signs of respiratory infection.
Table 129.9 and 129.10 list clinical features and laboratory parameters that are
useful in the diagnosis of neonatal infection or sepsis.
Table 129.10
Culture-Based and Non–Culture-Based Diagnostics for
Neonatal Sepsis
Table 129.11
Management and Prevention of Neonatal Sepsis
Healthcare-Acquired Infections
David B. Haslam
Table 130.1
Definitions of Healthcare-Acquired Infections for Patients
<12 Mo Old*
NOSOCOMIAL BLOODSTREAM INFECTIONS
Laboratory-Confirmed Bloodstream Infection (LCBI)
Must meet 1 of the following definitions:
• Recognized pathogen in 1 or more blood specimens (culture-based or non–culture-based microbiologic
methods), performed for clinical diagnostic or therapeutic purposes and not related to infection at another site.
• Commensal organism (e.g., coagulase-negative staphylococci, diphtheroids, bacillus, viridans streptococci,
aerococcus, micrococcus, propionibacterium), identified from 2 or more blood specimens obtained on separate
instances (culture- or non–culture-based microbiologic methods), performed for clinical diagnostic or
therapeutic purposes and not related to infection at another site and at least 1 of the following signs: (1) fever
(temperature >38.0°C), (2) hypothermia (temperature <36.0°C), or (3) apnea or bradycardia.
Central Line–Associated Bloodstream Infection (CLABSI)
• LCBI (as defined above) and
• Central line or umbilical catheter in place for >2 days and
• Central line in place on day of or day before CLABSI diagnosis.
Pneumonia
• Two or more serial chest radiographs with new/progressive and persistent infiltrate, cavitation, consolidation, or
pneumatoceles for patients with underlying pulmonary or cardiac disease (respiratory distress syndrome,
bronchopulmonary dysplasia, pulmonary edema) or 1 chest radiograph with the aforementioned abnormalities
for patients without underlying pulmonary or cardiac disease and
• Worsening gas exchange and
• At least 3 of the following; (1) temperature instability; (2) white blood cell count <4,000/µL or >15,000/µL with
10% or more bands, (3) new-onset purulent sputum, change in character of sputum, increased respiratory
secretions, or increased suctioning requirements; (4) physical examination findings consistent with increased
work of breathing or apnea, wheezing, rales, or rhonchi; (5) cough; (6) bradycardia (<100 beats/min), and (7)
tachycardia (>170 beats/min).
Ventilator-Associated Pneumonia (VAP)
• Pneumonia (as defined above) and
• Patient on ventilator for >2 days and
• Ventilator in place on day of or day before VAP diagnosis
URINARY TRACT INFECTION
Symptomatic Urinary Tract Infection (SUTI)
• At least 1 of the following symptoms: (1) fever (temperature >38.0°C), (2) hypothermia (temperature <36.0°C),
(3) apnea, (4) bradycardia, (5) lethargy, (6) vomiting, or (7) suprapubic tenderness and
• Urine culture with no more than 2 species identified, at least 1 of which is present at >105 CFU/mL.
Asymptomatic Bacteremic Urinary Tract Infection (ABUTI)
• Urine culture with no more than 2 species identified, at least 1 of which is present at >105 CFU/mL and
• Bacteria identified in blood (culture-based or nonculture-based microbiologic method) that matches at least one
of the bacteria present at more than 105 CFU/mL in urine.
Catheter–Associated Urinary Tract Infection:
• Urinary tract infection (as defined above, either SUTI or ABUTI) and
• Indwelling urinary catheter for >2 days and
• Urinary catheter in place on day of or day before urinary tract infection diagnosis.
* Centers for Disease Control and Prevention/National Healthcare Safety Network.
Incidence
The most common HAIs in the neonatal intensive care unit (NICU) are
bloodstream infections, predominantly central line–associated bloodstream
infections. Ventilator-associated pneumonia (VAP) is the next most common,
followed by surgical site infection and catheter-associated urinary tract infection.
Approximately 11% of NICU patients develop nosocomial infection during
their hospitalization; up to 25% of VLBW infants will have blood culture–
proven sepsis during their hospitalization. Infection rates are highest among the
most premature infants. Ventilator-associated pneumonia accounts for
approximately 25% of HAIs.
Epidemiology
HAIs in the NICU are predominantly caused by gram-positive organisms. The
largest fraction of bloodstream infections (BSIs) in the NICU are caused by
coagulase-negative staphylococci (Table 130.2 ). Other agents that often cause
HAIs in the newborn include Staphylococcus aureus, enterococci, gram-negative
bacilli (Escherichia coli, Klebsiella pneumoniae, Enterobacter spp.,
Pseudomonas aeruginosa ), and Candida. Viruses contributing to HAIs in the
neonate include rotavirus, enteroviruses, hepatitis A virus (HAV), adenoviruses,
influenza, respiratory syncytial virus (RSV), rhinovirus, parainfluenza, and
herpes simplex virus (HSV).
Table 130.2
Distribution of Organisms Responsible for Late-Onset
Sepsis
VLBW INFANTS: NICHD NRN (%)
ORGANISM
1991–1993 1998–2000 2002–2008
Incidence of late-onset sepsis 25 21 25
GRAM POSITIVE
Staphylococcus, coagulase-negative 55 48 53
Staphylococcus aureus 9 8 11
Enterococcus /group D streptococcus 5 3 4
Group B streptococcus 2 2 2
Other 2 9 7
GRAM NEGATIVE
Enterobacter 4 3 3
Escherichia coli 4 5 5
Klebsiella 4 4 4
Pseudomonas 2 3 2
Other 4 1 2
Fungi
Candida albicans 5 6 5
Candida parapsilosis
Other 2 2 1
VLBW, Very-low-birthweight (≤1,500 g); NICHD NRN, National Institutes of Child Health and
Human Development Neonatal Research Network.
Data from (1) 1991–1993: Stoll BJ, Gordon T, Korones SB, et al: Late-onset sepsis in very low
birth weight neonates: a report from the NICHD NRN, J Pediatr 129:63–71, 1996; (2) 1998–2000:
Stoll BJ, Hansen N, Fanaroff AA, et al: Late-onset sepsis in very low birth weight neonates: the
experience of the NICHD NRN, Pediatrics 110:285–291, 2002; (3) 2002–2008: Boghossian NS,
Page GP, Bell EF, et al: Late-onset sepsis in very low birth weight infants from singleton and
multiple gestation births, J Pediatr 162:1120–1120, 2015.
Adapted from Ramasethu J. Prevention and treatment of neonatal nosocomial infections, Matern
Health Neonatol Perinatol 3(5), 2017.
Table 130.3
U.S. Centers for Disease Control and Prevention (CDC)
Guidelines for Hand Hygiene
• When hands are visibly dirty or contaminated with proteinaceous material or are visibly soiled with blood or
other body fluids, wash hands with either a nonantimicrobial soap and water or an antimicrobial soap and water
(categorization of recommendation: IA)
• If hands are not visibly soiled, use an alcohol-based hand rub for routinely decontaminating hands in all other
clinical situations described below (categorization of recommendation: IA). Alternatively, wash hands with an
antimicrobial soap and water in all clinical situations described below (categorization of recommendation: IB).
• Decontaminate hands before having direct contact with patients (categorization of recommendation: IB).
• Decontaminate hands before donning sterile gloves when inserting an intravascular catheter (categorization of
recommendation: IB).
• Decontaminate hands before inserting indwelling urinary catheters, peripheral vascular catheters, or other
invasive devices that do not require a surgical procedure (categorization of recommendation: IB).
• Decontaminate hands after contact with a patient's intact skin (categorization of recommendation: IB).
• Decontaminate hands after contact with body fluids or excretions, mucous membranes, nonintact skin, and
wound dressings if hands are not visibly soiled (categorization of recommendation: IB).
• Decontaminate hands if moving from a contaminated body site to a clean body site during patient care
(categorization of recommendation: II).
• Decontaminate hands after contact with inanimate objects (including medical equipment) in the immediate
vicinity of the patient (categorization of recommendation: II).
• Decontaminate hands after removing gloves (categorization of recommendation: IB).
• Before eating and after using a restroom, wash hands with a nonantimicrobial soap and water or with
antimicrobial soap and water (categorization of recommendation: IB).
• Antimicrobial-impregnated wipes may be considered as an alternative to washing hands with nonantimicrobial
soap and water. Because they are not as effective as alcohol-based hand rubs or washing hands with an
antimicrobial soap and water for reducing bacterial counts on the hands of healthcare workers, they are not a
substitute for hand antisepsis (categorization of recommendation: IB).
• Wash hands with nonantimicrobial soap and water or with antimicrobial soap and water if exposure to Bacillus
anthracis is suspected or proven (categorization of recommendation: II).
• No recommendation can be made regarding the routine use of non–alcohol-based hand rubs for hand hygiene in
healthcare settings. Unresolved issue.
CDC/Healthcare Infection Control Practices Advisory Committee System for Categorizing Recommendations
Category IA: Strongly recommended for implementation and strongly supported by well-designed experimental,
clinical, or epidemiologic studies.
Category IB: Strongly recommended for implementation and supported by certain experimental, clinical, or
epidemiologic studies and using theoretical rationale.
Category IC: Required for implementation, as mandated by federal or state regulation or standard.
Category II: Suggested for implementation and supported by suggestive clinical or epidemiologic studies or a
theoretic rationale.
No recommendation; Unresolved issue: Practices for which insufficient evidence or no consensus regarding
efficacy exists.
Adapted from Boyce JM, Pittet D; Healthcare Infection Control Practices Advisory Committee:
Guideline for hand hygiene in health-care settings: recommendations of the Healthcare Infection
Control Practices Advisory Committee and the HIPAC/SHEA/APIC/IDSA Hand Hygiene Task
Force, Am J Infect Control 30(8):S1–S46, 2002.
Table 130.4
Interventions to Prevent Catheter-Related Infections
• Perform effective hand hygiene before and after any interaction with the catheter.
• Use sterile gowns, gloves, drapes, cap, and mask during catheter insertion.
• Disinfect skin with appropriate agent (chlorhexidine is most often used in United States; other disinfectants may
be as effective).
• Use a transparent, semipermeable dressing to cover catheter site.
• Change the dressing when soiled or loose.
• Scrub access point with alcoholic chlorhexidine for at least 15 sec.
• Use aseptic nontouch technique to access the catheter.
• Change administration sets no more frequently than 96 hr unless required by the infused product.
• Avoid use of systemic prophylactic antibiotics for catheter insertion.
• Evaluate daily, and remove central venous catheter when no longer required.
• Ensure all healthcare professionals who interact with the patient are educated on central line management.
Adapted from Taylor JE, McDonald SJ, Tan K: Prevention of central venous catheter–related
infection in the neonatal unit: a literature review, J Matern Fetal Neonatal Med 28(10):1224–1230,
2015.
Ventilator-Associated Pneumonia
FIG. 131.1 Pathogenesis of hematogenous transplacental infections. (Adapted from
Klein JO, Remington JS: Current concepts of infections of the fetus and newborn
infant. In Remington JS, Klein JO, editors: Infectious diseases of the fetus and newborn
infant, ed 5, Philadelphia, 2002, Saunders.)
Table 131.1
Specific Agents in Effects of Transplacental Fetal Infection
on the Fetus and Newborn Infant
DISEASE
Intrauterine
ORGANISM Persistent
Growth Developmental Congenital
Prematurity Postnatal
Restriction/Low Anomalies Disease
Infection
Birthweight
Viruses CMV CMV CMV CMV CMV
HSV Rubella Rubella Rubella Rubella
Rubeola VZV* VZV VZV VZV
Smallpox HIV* Coxsackievirus HSV HSV
HBV B* Mumps* HBV
HIV* HIV* Rubeola HIV
Zika Vaccinia Zika
Smallpox
Coxsackievirus
B
Poliovirus
HBV
HIV
LCV
Parvovirus
Bacteria Treponema T. pallidum T. pallidum
pallidum M. tuberculosis M.
Mycobacterium L. tuberculosis
tuberculosis monocytogenes
Listeria C. fetus
monocytogenes S. typhi
Campylobacter Borrelia
fetus burgdorferi
Salmonella
typhi
Protozoa Toxoplasma T. gondii T. gondii T. gondii
gondii Plasmodium Plasmodium Plasmodium
Plasmodium * T. cruzi T. cruzi
Trypanosoma
cruzi
* Association of effect with infection has been suggested and is under consideration.
CMV , Cytomegalovirus; HBV , hepatitis B virus; HIV , human immunodeficiency virus; HSV ,
herpes simplex virus; LCV , lymphocytic choriomeningitis virus; VZV , varicella-zoster virus.
From Maldonado YA, Nizet V, Klein JO, et al: Current concepts of infections of the fetus and
newborn infant. In Wilson CB, Nizet V, Maldonado Y, et al, editors: Remington and Klein's
infectious diseases of the fetus and newborn, ed 8, Philadelphia, 2016, Elsevier (Table 1-5).
Clinical Manifestations
The clinical manifestations of intrauterine infections can range from
asymptomatic to severe multiorgan system complications. For some agents (e.g.,
CMV, T. pallidum ), ongoing injury after birth leads to late sequelae. The
specific clinical signs in the newborn period are usually not sufficient to make a
definitive diagnosis but are useful to guide more specific laboratory testing.
Symptomatic congenital infections often affect the central nervous system (CNS;
brain and eyes) and the reticuloendothelial system (RES; bone marrow, liver, and
spleen). Table 131.2 presents the clinical manifestations of some specific
congenital infections. Congenital Zika virus infection has features that are rarely
seen with other congenital infections (Table 131.3 ). No hematologic or hepatic
laboratory abnormalities have been documented in infants with congenital Zika
virus infection. Table 131.4 provides late sequelae of some congenital infections.
Table 131.2
Clinical Manifestations of Specific Neonatal Infections
Acquired in Utero or at Delivery
Rubella Virus Cytomegalovirus Toxoplasma gondii Herpes Simplex Virus Treponema pallidum
Hepatosplenomegaly Hepatosplenomegaly Hepatosplenomegaly Hepatosplenomegaly Hepatosplenomegaly
Jaundice Jaundice Jaundice Jaundice Jaundice
Pneumonitis Pneumonitis Pneumonitis Pneumonitis Pneumonitis
Petechiae or purpura Petechiae or purpura Petechiae or purpura Petechiae or purpura Petechiae
Meningoencephalitis Meningoencephalitis Meningoencephalitis Meningoencephalitis Meningoencephalitis
Hydrocephalus Hydrocephalus Hydrocephalus* Hydrocephalus Adenopathy
Adenopathy Microcephaly* Microcephaly Microcephaly Maculopapular
Hearing deficits Intracranial Maculopapular Maculopapular exanthems
Myocarditis calcifications* exanthems exanthems Bone lesions
Congenital defects* Hearing deficits Intracranial Vesicles* Glaucoma
Bone lesions* Chorioretinitis or calcifications* Myocarditis Chorioretinitis
Glaucoma* retinopathy Myocarditis Chorioretinitis or retinopathy
Chorioretinitis or Optic atrophy Bone lesions retinopathy Uveitis
retinopathy* Chorioretinitis or Cataracts
Cataracts* retinopathy* Conjunctivitis or
Microphthalmia Cataracts keratoconjunctivitis*
Optic atrophy
Microphthalmia
Uveitis
* Has special diagnostic significance for this infection
From Maldonado YA, Nizet V, Klein JO, et al: Current concepts of infections of the fetus and
newborn infant. In Wilson CB, Nizet V, Maldonado Y, et al, editors: Remington and Klein's
infectious diseases of the fetus and newborn, ed 8, Philadelphia, 2016, Elsevier (Table 1-6).
Table 131.3
Table 131.4
Late Sequelae of Intrauterine Infections.
INFECTION
CLINICAL SIGN
Cytomegalovirus Rubella Virus Toxoplasma gondii Treponema pallidum
Deafness + + + +
Dental/skeletal problems + + (−) +
Mental retardation + + + +
Seizures + + + +
+, Present; (−), rare or absent.
Diagnosis
During Pregnancy
The presence of IUGR or a physical abnormality on a prenatal fetal ultrasound
raises concern for a congenital infection. The well-known acronym TORCH —
T oxoplasma gondii , O ther (Treponema pallidum , human parvovirus B19, HIV,
Zika virus, others), R ubella, C ytomegalovirus, and H erpes simplex virus
(HSV)—is a useful mnemonic. However, the routine ordering of TORCH
serology panels is not recommended because the presence of a TORCH agent
IgG antibody in the mother indicates past infection but does not establish if the
infection occurred during pregnancy. Maternal IgM titers to specific pathogens
are only moderately sensitive, and a negative result cannot be used to exclude
infection.
In certain cases, a fetal blood sample with cordocentesis can be obtained and
tested for total and pathogen-specific IgM assays, polymerase chain reaction
The human birth canal is colonized with aerobic and anaerobic bacteria.
Ascending amniotic infection may occur with either apparently intact
membranes or relatively brief duration of membrane rupture. Infectious agents
can also be acquired as the newborn infant passes through the vaginal canal. This
acquisition may result in either colonization or disease. Factors influencing
which colonized infants will experience disease are not well understood but
include prematurity, underlying illness, invasive procedures, inoculum size,
virulence of the infecting organism, genetic predisposition, the innate immune
system, host response, and transplacental maternal antibodies.
Chorioamnionitis has been historically used to refer to microbial invasion of
the amniotic fluid, often as a result of prolonged rupture of the chorioamniotic
membrane for >18 hr. The term chorioamnionitis is confusing because it does
not convey the spectrum of inflammatory or infectious diseases, it leaves out
other intrauterine components that can be involved (e.g., decidua), and it results
in significant variability in clinical practice, with the potential for a significant
number of well newborns being exposed to antimicrobial agents. The term
intrauterine inflammation or infection at birth , abbreviated as Triple I , has
become more accepted because of the heterogeneous nature of conditions that
can affect the mother and neonate (Table 131.5 ). Regardless of the definition
used, prematurity (<37 wk) is associated with a greater risk of early-onset sepsis,
especially with group B streptococcus.
Table 131.5
Classification of Triple I and Isolated Maternal Fever
TERMINOLOGY FEATURES
Isolated maternal Maternal oral temperature ≥39°C is considered a “documented fever.”
fever If the oral temperature is ≥38°C but ≤39°C, repeat the measurement in 30 min. If the
repeat value is ≥38°C, it is considered a “documented fever.”
Suspected Triple I Fever without a clear source with any of the following:
1. Baseline fetal tachycardia (>160 beats/min for 10 min)
2. Maternal WBC >15,000/mm3
3. Purulent fluid from the cervical os
Confirmed Triple I All the above (from suspected Triple I) with any of the following:
1. Amniocentesis-proven infection through positive Gram stain
2. Low glucose of amniotic fluid or positive amniotic fluid culture
3. Placental pathology consistent with infection
Triple I, Intrauterine inflammation or infection at birth; WBC, white blood cell count.
Adapted from Higgins RD, Saade G; Chorioamnionitis Workshop participants: Evaluation and
management of women and newborns with a maternal diagnosis of chorioamnionitis: summary of
Fungi
Candida spp. (Chapter 261 )
Diagnosis
The maternal history provides important information about maternal exposures
to infectious diseases, bacterial colonization, immunity (natural and acquired),
and obstetric risk factors (prematurity, prolonged ruptured membranes,
chorioamnionitis). STIs acquired by a pregnant woman, including syphilis, N.
gonorrhoeae , and C. trachomatis, have the potential for perinatal transmission.
Neonates with perinatal infections often present with nonspecific symptoms
and signs; therefore the general diagnostic evaluation for the ill neonate as
discussed in Chapter 202 should be followed. Table 131.6 provides a summary
of laboratory tests that are useful to diagnose specific perinatal infections.
Table 131.6
Laboratory Tests in the Diagnosis of Specific Perinatal
Infections
ACCEPTABLE SPECIMEN(S)
INFECTIOUS AGENT FROM INFANT UNLESS LABORATORY TEST
OTHERWISE INDICATED
Chlamydia trachomatis Conjunctiva, nasopharyngeal swab, Culture using special transport
tracheal aspirate media
Nucleic acid amplification tests
(NAATs) are not FDA-approved
for specimens from neonates.*
Genital mycoplasmas Tracheal aspirate, blood, or cerebrospinal Culture using special transport
(Mycoplasma hominis , M. fluid (CSF) media
genitalium , Ureaplasma Real-time polymerase chain
urealyticum ) reactions (PCRs)
Neisseria gonorrhoeae Conjunctiva, blood, CSF, or synovial Finding gram-negative
fluid intracellular diplococci on Gram
stain is suggestive.
Culture on special media
establishes the diagnosis.
Syphilis (Treponema pallidum ) Serum (mother) Rapid plasma reagin (RPR) and if
reactive, a specific treponemal test †
Serum RPR
CSF Venereal Disease Research
Laboratories (VDRL)
Cytomegalovirus Urine, saliva, blood, or CSF PCR for detection of CMV DNA
Obtain within 2-4 wk of birth.
Enteroviruses Blood, nasopharyngeal swab, throat PCR
swab, conjunctival swab, tracheal Cell culture (sensitivity depends
aspirate, urine, stool, rectal swab, or CSF on serotype and cell lines used)
Hepatitis B Serum (mother) Hepatitis B surface antigen (HBsAg)
Serum If mother's HBsAg is positive, at age
9 mo, test the infant for HBsAg and
hepatitis B surface antibody.
Herpes simplex viruses 1 and 2 Conjunctiva, skin vesicle scraping, PCR or cell culture
whole blood, or mouth vesicles
CSF PCR
“Surface cultures” (mouth, nasopharynx, PCR or cell culture
conjunctiva, and anus)
Human immunodeficiency virus Serum (mother) Fourth-generation HIV
(HIV) antigen/antibody test
Whole blood HIV DNA PCR
Candida species Blood, skin biopsy, or CSF Culture
Zika virus Blood, urine, CSF NAT and serum IgM
NAT may be falsely negative
IgG antibodies may reflect
maternal exposure
Antibodies may cross react with
other flaviviruses
* Published evaluations of NAATs for these indications are limited, but sensitivity and specificity is
expected to be at least as high as those for culture. FDA, U.S. Food and Drug Administration.
† Treponemal tests include the T. pallidum particle agglutination (TP-PA) test, T. pallidum enzyme
immunoassay (TP-EIA), T. pallidum chemiluminescent assay (TP-CIA), and fluorescent
treponemal antibody absorption (FTA-ABS) test.
Bibliography
American Academy of Pediatrics. Syphilis. Kimberlin DW,
Brady MT, Jackson MA, Long SS. Red book: 2018-2021
report of the committee on infectious diseases . ed 31.
American Academy of Pediatrics: Itasca, IL; 2018:773–788.
Bilavsky E, Pardo J, Attias J, et al. Clinical implications for
children born with congenital cytomegalovirus infection
following a negative amniocentesis. Clin Infect Dis .
2016;63:33–38.
Boppana SB, Ross SA, Fowler KB. Congenital cytomegalovirus
infection: clinical outcome. Clin Infect Dis . 2013;57(Suppl
4):S178–S181.
CHAPTER 132
Table 132.1
Milestones in Early, Middle, and Late Adolescent
Development
LATE
VARIABLE EARLY ADOLESCENCE MIDDLE ADOLESCENCE
ADOLESCENCE
Approximate 10-13 yr 14-17 yr 18-21 yr
age range
Sexual maturity 1-2 3-5 5
rating*
Physical Females: secondary sex Females: peak growth velocity, Physical
characteristics (breast, menarche (if not already attained) maturation
pubic, axillary hair), start of Males: growth spurt, secondary sex slows
growth spurt characteristics, nocturnal Increased lean
Males: testicular emissions, facial and body hair, muscle mass in
enlargement, start of genital voice changes males
growth Change in body composition
Acne
Cognitive and Concrete operations Emergence of abstract thought Future-oriented
moral Egocentricity (formal operations) with sense of
Unable to perceive long- May perceive future implications, perspective
term outcome of current but may not apply in decision- Idealism
decisions making Able to think
Follow rules to avoid Strong emotions may drive things through
punishment decision-making independently
Sense of invulnerability Improved
Growing ability to see others' impulse control
perspectives Improved
assessment of
risk vs reward
Able to
distinguish law
from morality
Self- Preoccupied with changing Concern with attractiveness More stable
concept/identity body Increasing introspection body image
formation Self-consciousness about Attractiveness
appearance and may still be of
attractiveness concern
Consolidation
of identity
Family Increased need for privacy Conflicts over control and Emotional and
Exploration of boundaries independence physical
of dependence vs Struggle for greater autonomy separation from
independence Increased separation from parents family
Increased
autonomy
Reestablishment
of “adult”
relationship
with parents
Peers Same-sex peer affiliations Intense peer group involvement Peer group and
Preoccupation with peer culture values recede in
Conformity importance
Sexual Increased interest in sexual Testing ability to attract partner Consolidation
anatomy Initiation of relationships and of sexual
Anxieties and questions sexual activity identity
about pubertal changes Exploration of sexual identity Focus on
Limited capacity for intimacy and
intimacy formation of
stable
relationships
Planning for
future and
commitment
* See text and Figs. 132.1 and 132.2 .
FIG. 132.2 Sexual maturity ratings (1-5) of breast changes in adolescent
females. (Courtesy of J.M. Tanner, MD, Institute of Child Health,
Department for Growth and Development, University of London.)
Table 132.2
Table 132.3
Sexual Maturity Rating (SMR) Stages in Males
SMR
PUBIC HAIR PENIS TESTES
STAGE
1 None Preadolescent Preadolescent
2 Scant, long, slightly pigmented Minimal change/enlargement Enlarged scrotum, pink,
texture altered
3 Darker, starting to curl, small amount Lengthens Larger
4 Resembles adult type, but less Larger; glans and breadth Larger, scrotum dark
quantity; coarse, curly increase in size
5 Adult distribution, spread to medial Adult size Adult size
surface of thighs
From Tanner JM: Growth at adolescence, ed 2, Oxford, England, 1962, Blackwell Scientific.
FIG. 132.3 Sequence of pubertal events in males. Although the age of
activities, demanding privacy, and increasing argumentativeness, are normal;
extreme withdrawal or antagonism may be dysfunctional, signaling a mental
health or substance use concern. Bewilderment and dysphoria at the start of
middle school are normal; continued failure to adapt several months later
suggests a more serious problem. Although some degree of risk taking is normal,
progressive escalation of risk-taking behaviors is problematic. In general, when
the adolescent's behaviors cause significant dysfunction in the domains of home
life, academics, or peer relationships, they should be addressed by the parents
and healthcare provider, and referral to a mental health provider may be
considered. In most cases, parents can be reassured that although adolescence
can pose unique challenges, their adolescent, like most adolescents, will come
through it to become a successful and happy adult.
At national and international levels, adolescents are at risk for environmental,
health, behavioral, and societal challenges. Table 132.4 provides suggestions to
address these issues.
Table 132.4
Recommended Action Bundles* for Adolescent and Young
Adult Health Problems and Risks
COMMUNITY ELECTRONIC
PROBLEM/RISK SOCIAL INTERVENTIONS HEALTH,
STRUCTURAL SCHOOLS
AREA MARKETING INCLUDING MOBILE
FAMILY HEALTH
Sexual and Legislation Promote Cash transfer Target Quality
reproductive 18 years as the community programs, with knowledge, secondary
health, including minimum age of support for payments linked attitudes, and education
HIV marriage sexual and to staying in risk behaviors Comprehensive
Allow provision reproductive school sexuality
of contraception health, and HIV Positive youth education
to legal minors health access for development Safe schools
Legalize abortion adolescents Peer education with clean
toilets and
facilities for
menstrual care
School-based
health services
with condoms
and modern
contraceptives
Table 133.1
Table 136.1
Table 136.2
Leading Causes of Death Among 15-19 Yr Olds by Gender,
United States, 2014*
MALE FEMALE
LEADING
CAUSES OF Cause of Death Mortality Rate per Cause of Death Mortality Rate per
DEATH 100,000 Population 100,000 Population
#1 Accidents 24.9 Accidents 10.1
(unintentional (unintentional
injuries) injuries)
#2 Intentional self- 13.0 Intentional self- 4.2
harm (suicide) harm (suicide)
#3 Assault (homicide) 11.2 Malignant 2.5
neoplasms
* Based on data from Heron M: Deaths: leading causes for 2014, Natl Vital Stat Rep 65(5), 2016.
Table 136.3
Adolescent Health Outcomes by Race/Ethnicity, United
States, 2010–2012
AI/AN, American Indian or Alaska Native; API, Asian or Pacific Islander; HIV, human
immunodeficiency virus.
Access to Healthcare
Adolescents in the United States make fewer visits to physicians for ambulatory
office visits than any other age group; school-age children and adolescents are
more likely than younger children to have unmet health needs and delayed
medical care. Adolescents who actually receive preventive care may still not
have access to time alone with their provider to discuss confidential health
issues such as STIs, HIV, or pregnancy prevention. Less than half (40%) of
adolescents have time alone with their healthcare provider during a preventive
healthcare visit; sexually experienced teens report sexual health discussions
more often than nonsexually experienced teens, but the frequency is still low at
64% and 33.5% for sexually experienced females and males, respectively.
Young adults 18-24 yr are more likely to be insured with the 2010 Patient
Protection and Affordable Care Act (ACA ). Currently, the ACA permits
children to receive benefits from their parents' health plans through age 26 yr.
Healthy People provides science-based, 10-yr national objectives for measuring
and improving the health of all Americans by establishing benchmarks and
monitoring progress over time. The Healthy People 2020 agenda includes 11
adolescent-specific objectives with a goal of improving the healthy development,
health, safety, and well-being of adolescents and young adults over the next 10
yr (Table 136.4 ). This science-based initiative is centered around a framework
for public health prevention priorities and actions to improve the health status of
U.S. youth.
Table 136.4
Healthy People 2020 Adolescent Health (AH)
Objectives
Bibliography
Blum RW, Bastos FIPM, Kabiru CW, et al. Adolescent health in
the 21st century. Lancet . 2012;379:1567–1568.
Catalano RF, Fagan AA, Gavin LE, et al. Worldwide application
of prevention science in adolescent health. Lancet .
2012;379:1653–1662.
Centers for Disease Control and Prevention. Tables of summary
health statistics, National Health Interview Survey .
https://www.cdc.gov/nchs/nhis/shs/tables.htm ; 2015.
Centers for Disease Control and Prevention. HIV surveillance
report . [Vol 27, 2016]
http://www.cdc.gov/hiv/library/reports/hiv-surveillance.html ;
2015.
Centers for Disease Control and Prevention. Sexually
transmitted disease surveillance 2015 . [Atlanta; US
Department of Health and Human Services]
https://www.cdc.gov/std/stats15/default.htm ; 2016.
Centers for Disease Control and Prevention. 1991–2015 High
school youth risk behavior survey data .
http://nccd.cdc.gov/youthonline .
Hamilton BE, Martin JA, Osterman MJK, et al. Births: final
data for 2014. Natl Vital Stat Rep . 2015;64(12).
Heron M. Deaths: leading causes for 2014. Natl Vital Stat Rep .
2016;65(5)
dissemination of provider education about adolescent preventive health
guidelines have been demonstrated to improve the content of recommended care
(Table 137.1 ). The ease of recognition or expectation that an adolescent's needs
can be addressed in a setting relates to the visibility and flexibility of sites and
services. Staff at sites should be approachable, linguistically capable, and
culturally competent. Health services should be coordinated to respond to goals
for adolescent health at the local, state, and national levels. The coordination
should address service financing and delivery in a manner that reduces
disparities in care.
Table 137.1
Bright Futures/American Academy of Pediatrics
Recommendations for Preventive Healthcare for 11-21 Yr
Olds
http://www.cdc.gov/vaccines/schedules/hcp/index.html and
http://redbook.solutions.aap.org/SS/Immunization_Schedules.aspx .
† CDC recommends universal, voluntary HIV screening of all sexually active people, beginning at
age 13 yr. The American Academy of Pediatrics recommends offering routine HIV screening to all
adolescents at least once by 16-18 yr of age and to those younger if at risk. US Preventive
Services Task Force recommends offering routine HIV screening to all adolescents age 15 yr and
older at least once and to those younger if at risk. Patients who test positive for HIV should
receive prevention counseling and referral to care before leaving the testing site.
‡ Screening for cervical cancer, April 2012, US Preventive Services Task Force.
http://www.uspreventiveservicestaskforce.org/uspstf/uspscerv.htm .
§ Refer to specific guidance by age as listed in Bright Futures guidelines.
Table 137.2
Adolescent Screening Recommendations
18-21 YR OLD
11-14 YR OLD VISIT 15-17 YR OLD VISIT
VISIT
Universal
Action Action Action
Screening
Cervical N/A N/A Pap smear all
dysplasia* young women at
21 yr visit
Depression Adolescent depression screen Adolescent depression screen Adolescent
beginning at 12 yr visit depression screen
Dyslipidemia Lipid screen once at 9-11 yr Lipid screen once at 17-21 yr Lipid screen once
at 17-21 yr
Hearing Once at 11-14 yr Once at 15-17 yr Once at 18-
Audiometry, including 6,000 and Audiometry, including 6,000 and 21 yr
8,000 Hz high frequencies 8,000 Hz high frequencies Audiometry,
including
6,000 and
8,000 Hz
high
frequencies
HIV † Selective screening (see below) HIV test once at 15-18 yr HIV test once at
15-18 yr
Tobacco, Tobacco, alcohol, or drug use screen Tobacco, alcohol, or drug use screen Tobacco, alcohol,
alcohol, or or drug use screen
drug use
Vision At 12 yr visit At 15 yr visit N/A
Objective measure with age- Objective measure with age-
appropriate visual-acuity appropriate visual-acuity
measurement using HOTV or Lea measurement using HOTV or Lea
symbols, Sloan letters, or Snellen symbols, Sloan letters, or Snellen
letters letters
Table 137.3
Types of Minor Consent Statutes or Rules of Common Law
That Allow for Medical Treatment of a Minor Patient
Without Parental Consent
LEGAL
EXCEPTIONS
TO INFORMED MEDICAL CARE SETTING
CONSENT
REQUIREMENT
The “emergency” • The child is suffering from an emergent condition that places his or her life or health in
exception danger.
• The child's legal guardian is unavailable or unable to provide consent for treatment or
transport.
• Treatment or transport cannot be safely delayed until consent can be obtained.
• The professional administers only treatment for emergent conditions that pose an
immediate threat to the child.
The “emancipated • Married
minor” exception • Economically self-supporting and not living at home
• Active-duty status in the military
• In some states, a minor who is a parent or pregnant
• Some states might require a court to declare the emancipation of a minor.
The “mature Most states recognize a mature minor, in which a minor, usually ≥14 yr, displays sufficient
minor” exception maturity and intelligence to understand and appreciate the benefits, risks, and alternatives of
the proposed treatment and to make a voluntary and reasonable choice on the basis of that
information. States vary or whether a judicial determination is required.
Exceptions based Minor seeks:
on specific medical • Mental health services
condition (state • Pregnancy and contraceptive services
laws vary) • Testing or treatment for HIV infection or AIDS
• Sexually transmitted infection testing and treatment
• Drug and alcohol addiction treatment
Data from American Academy of Pediatrics: Consent for emergency medical services for children
and adolescents, Pediatrics 128:427–433, 2011.
Minors' right to consent for contraceptive services varies from state to state.
Almost 50% of states and the District of Columbia explicitly authorize all
minors to consent for their own contraceptive services; and 50% of states permit
minors to consent for their own contraceptive services under specific
circumstances, such as being married, a parent, currently or previously pregnant,
over a certain age, or a high school graduate, or per physician's discretion.
A minor's right to consent for mental healthcare and substance abuse
treatment services vary by state and age of minor, whether care is medical vs
nonmedical (e.g., counseling), and whether care is delivered as an inpatient vs
outpatient basis. Minor consent laws often contain provisions regarding
confidentiality and disclosure, even when general state consent laws do not have
such provisions.
The confidentiality of medical information and records of a minor who has
consented for his or her own reproductive healthcare is governed by numerous
federal and state laws. Laws in some states explicitly protect the confidentiality
of STI or contraceptive services for which minors have given their own consent
and do not allow disclosure of the information without the minor's consent. In
other states, laws grant physicians discretion to disclose information to parents.
The confidentiality of medical information and records of a minor who has
consented for his or her own healthcare is also governed by numerous federal
and state laws. Laws in some states explicitly protect the confidentiality of STI,
contraceptive, or mental health services for which minors have given their own
consent, and do not allow disclosure of the information without the minor's
consent. In other states, laws grant physicians discretion to disclose information
to parents. Title X and Medicaid both provide confidentiality protection for
family planning services provided to minors with funding from these programs.
Federal regulations issued under the Federal Health Insurance Portability and
Accountability Act of 1996, known as the HIPAA Privacy Rule , defer to state
and “other applicable laws” with respect to the question of whether parents have
can share even the most personal secret?), self-image (Is there anything you
would like to change about yourself?), depression (What do you see yourself
doing 5 yr from now?), school (How are your grades this year compared with
last year?), personal decisions (Are you feeling pressured to engage in any
behavior for which you do not feel you are ready?), and an eating disorder (Do
you ever feel that food controls you, rather than vice versa?). Bright Futures
materials provide questions and patient encounter forms to structure the
assessments. The HEADS/SF/FIRST mnemonic, basic or expanded, can be
useful in guiding the interview if encounter forms are not available (Table 137.4
). Based on the assessments, appropriate counseling or referrals are
recommended for more thorough probing or for in-depth interviewing.
Table 137.4
Adolescent Psychosocial Assessment:
HEADS/SF/FIRST Mnemonic
H ome. Space, privacy, frequent geographic moves, neighborhood
E ducation/School. Frequent school changes, repetition of a grade/in each
subject, teachers' reports, vocational goals, after-school educational clubs
(e.g., language, speech, math), learning disabilities
A buse. Physical, sexual, emotional, verbal abuse; parental discipline
D rugs. Tobacco, electronic cigarettes or vaping devices, alcohol,
marijuana, inhalants, “club drugs,” “rave” parties, others; drug of choice,
age at initiation, frequency, mode of intake, rituals, alone or with peers,
quit methods, number of attempts
S afety. Seat belts, helmets, sports safety measures, hazardous activities,
driving while intoxicated
S exuality/S exual Identity. Reproductive health (use of contraceptives,
presence of sexually transmitted infections, feelings, pregnancy)
F amily and F riends
Family: Family constellation; genogram;
single/married/separated/divorced/blended family; family
occupations and shifts; history of addiction in first- and second-
degree relatives; parental attitude toward alcohol and drugs;
parental rules; chronically ill, physically or mentally challenged
parent
Friends: Peer cliques and configuration (“preppies,” “jocks,”
“nerds,” “computer geeks,” cheerleaders), gang or cult
affiliation
I mage. Height and weight perceptions, body musculature and physique,
appearance (including dress, jewelry, tattoos, body piercing as fashion
trends or other statement)
R ecreation. Sleep, exercise, organized or unstructured sports, recreational
activities (television, video games, computer games, internet and chat
rooms, church or community youth group activities [e.g., Boy (BSA)/Girl
Scouts; Big Brother/Sister groups, campus groups]). How many hours per
day, days per week involved?
S pirituality and Connectedness. Use HOPE* or FICA † acronym;
adherence, rituals, occult practices, community service or involvement
T hreats and Violence. Self-harm or harm to others, running away, cruelty
to animals, guns, fights, arrests, stealing, fire setting, fights in school
From Dias PJ: Adolescent substance abuse: assessment in the office, Pediatr
Clin North Am 49:269–300, 2002.
Physical Examination
Vision Testing
The pubertal growth spurt may involve the optic globe, resulting in its
elongation and myopia in genetically predisposed individuals (see Chapter 636 ).
Vision testing should therefore be performed to detect this problem before it
affects school performance.
Audiometry
Highly amplified music of the kind enjoyed by many adolescents may result in
hearing loss or tinnitus (see Chapter 654 ). A hearing screening is recommended
CHAPTER 138
The process begins with the development of a transition policy and its
dissemination to all families of young adolescents, ensuring that families
understand that transition planning will be an element of health maintenance and
chronic care management visits throughout the adolescent years. By middle
adolescence, a transition plan should be developed with the youth and family
caregivers and updated at subsequent visits until the patient is ready for
implementation of the adult care model in early adulthood. Periodic readiness
assessments are key to plan and anticipate challenges. Critical to the transition
process is skills training for the adolescent in communication, self-advocacy,
and self-care. Some youth with SHCN depend on caregivers for navigating the
healthcare system on their behalf, and it is not realistic to expect increased
independence. For these youth, addressing guardianship, long-term care
planning, and advanced directives are important. Care coordination has been
found to facilitate navigation and engagement in an adult-oriented health system,
especially for adolescents with SHCN. The goal is to help all youth maximize
their potential as they become young adults.
specific psychiatric diagnoses whose definitions are associated with violent
behavior (Table 139.1 ). They occur with other disorders such as ADHD (see
Chapter 49 ) and increase an adolescent's vulnerability for juvenile delinquency,
substance use or abuse, sexual promiscuity, adult criminal behavior,
incarceration, and antisocial personality disorder. Other co-occurring risk factors
for youth violence include use of anabolic steroids, gang tattoos, belief in one's
premature death, preteen alcohol use, and placement in a juvenile detention
center.
Table 139.1
Oppositional Defiant Disorder, Conduct Disorder, and
Juvenile Delinquency
PSYCHIATRIC DISORDER LABELS
Oppositional Defiant Legal Label Juvenile Delinquency
Conduct Disorder
Disorder
Recurrent pattern of Repetitive and persistent pattern of Offenses that are illegal because of age;
negativistic, defiant, behavior that violates the basic rights illegal acts
disobedient, and hostile of others or major age-appropriate
behavior toward authority societal norms or rules
figures that has a significant
adverse effect on functioning
(e.g., social, academic,
occupational)
Examples: losing temper; Examples: physical fighting, Examples: single or multiple instances
arguing with adults; defying or deceitfulness, stealing, destruction of of being arrested or adjudicated for any
refusing to comply with request property, threatening or causing of the following: stealing, destruction of
or rules of adults; annoying physical harm to people or animals, property, threatening or causing
behavior; blaming others; being driving without a license, prostitution, physical harm to people or animals,
irritable, spiteful, resentful rape (even if not adjudicated in the driving without a license, prostitution,
legal system) rape
Diagnosed by a mental health Diagnosed by a mental health Adjudicated in the legal system
clinician practitioner
From Greydanus DE, Pratt HD, Patel DR, et al: The rebellious adolescent, Pediatr Clin North Am
44:1460, 1997.
Diagnosis
The assessment of an adolescent at risk or with a history of violent behavior or
victimization should be a part of the health maintenance visit of all adolescents.
The answers to questions about recent history of involvement in a physical fight,
carrying a weapon, or firearms in the household, as well as concerns that the
adolescent may have about personal safety, may suggest a problem requiring a
more in-depth evaluation. The FISTS mnemonic provides guidance for
structuring the assessment (Table 139.2 ). The additional factors of physical or
sexual abuse, serious problems at school, poor school performance and
attendance, multiple incidents of trauma, substance use, and symptoms
associated with mental disorders are indications for evaluation by a mental
health professional. In a situation of acute trauma, assault victims are not always
forthcoming about the circumstances of their injuries for fear of retaliation or
police involvement. Stabilization of the injury or the gathering of forensic
evidence in sexual assault is the treatment priority; however, once this is
achieved, addressing a more comprehensive set of issues surrounding the assault
is appropriate.
Table 139.2
FISTS Mnemonic to Assess an Adolescent's
Risk of Violence
F: Fighting (How many fights were you in last year? What was the last?)
I: Injuries (Have you ever been injured? Have you ever injured someone
else?)
S: Sex (Has your partner hit you? Have you hit your partner? Have you
ever been forced to have sex?)
T: Threats (Has someone with a weapon threatened you? What happened?
Has anything changed to make you feel safer?)
S: Self-defense (What do you do if someone tries to pick a fight? Have you
carried a weapon in self-defense?)
Treatment
In the patient with acute injury secondary to violent assault, the treatment plan
should follow standards established by the American Academy of Pediatrics
model protocol, which includes the stabilization of the injury, evaluation and
violent behaviors.
Table 139.3
WHO Youth Violence Prevention Strategies: Effectiveness
by Context
Table 140.1
Assessing the Seriousness of Adolescent Drug Abuse
VARIABLE 0 +1 +2
Age (yr) >15 <15
Sex Male Female
Family history of drug Yes
abuse
Setting of drug use In group Alone
Affect before drug use Happy Always poor Sad
School performance Good, improving Recently poor
Use before driving None Yes
History of accidents None Yes
Time of week Weekend Weekdays
Time of day After school Before or during school
Type of drug Marijuana, beer, Hallucinogens, Whiskey, opiates, cocaine,
wine amphetamines barbiturates
Total score: 0-3, less worrisome; 3-8, serious; 8-18, very serious.
Table 140.2
Stages of Adolescent Substance Abuse
STAGE DESCRIPTION
1 Potential for abuse
• Decreased impulse control
• Need for immediate gratification
• Available drugs, alcohol, inhalants
• Need for peer acceptance
2 Experimentation: learning the euphoria
• Use of inhalants, tobacco, marijuana, and alcohol with friends
• Few, if any, consequences
• Use may increase to weekends regularly
• Little change in behavior
3 Regular use: seeking the euphoria
• Use of other drugs, e.g., stimulants, LSD, sedatives
• Behavioral changes and some consequences
• Increased frequency of use; use alone
• Buying or stealing drugs
4 Regular use: preoccupation with the “high”
• Daily use of drugs
• Loss of control
• Multiple consequences and risk taking
• Estrangement from family and “straight” friends
5 Burnout: use of drugs to feel normal
• Polysubstance use/cross-addiction
• Guilt, withdrawal, shame, remorse, depression
• Physical and mental deterioration
• Increased risk taking, self-destructive, suicidal
Epidemiology
Alcohol, cigarettes, and marijuana are the most commonly reported substances
used among U.S. teens (Table 140.3 ). The prevalence of substance use and
associated risky behaviors vary by age, gender, race/ethnicity, and other
sociodemographic factors. Younger teenagers tend to report less use of drugs
than do older teenagers, except for inhalants (in 2016, 4.4% in 8th grade, 2.8%
in 10th grade, 1.0% in 12th grade). Males have higher rates of both licit and
illicit drug use than females, with greatest differences seen in their higher rates
of frequent use of smokeless tobacco, cigars, and anabolic steroids. For a
number of years, black 12th graders have reported lifetime, annual, 30-day, and
daily prevalence levels for nearly all drugs that were lower than those for white
or Hispanic 12th graders. That is less true today, with levels of drug use among
blacks more similar to the other groups.
Table 140.3
Trends in Annual Prevalence (%) of Use of Various Drugs
for Grades 8, 10, and 12 Combined
1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005
Any illicit drug c 20.2 19.7 23.2 27.6 31.0 33.6 34.1 32.2 31.9 31.4 31.8 30.2 28.4 27.6 27.1
Any illicit drug 12.0 12.0 13.6 14.6 16.4 17.0 16.8 15.8 15.6 15.3 16.3 14.6 13.7 13.5 13.1
other than ‡
marijuana c
Any illicit drug 23.5 23.2 26.7 31.1 34.1 36.6 36.7 35.0 34.6 34.1 34.3 32.3 30.8 30.1 30.1
including
inhalants c
Marijuana/hashish 15.0 14.3 17.7 22.5 26.1 29.0 30.1 28.2 27.9 27.2 27.5 26.1 24.6 23.8 23.4
Synthetic ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
marijuana
Inhalants 7.6 7.8 8.9 9.6 10.2 9.9 9.1 8.5 7.9 7.7 6.9 6.1 6.2 6.7 7.0
Hallucinogens 3.8 4.1 4.8 5.2 6.6 7.2 6.9 6.3 6.1 5.4 ‡ 6.0 4.5 4.1 4.0 3.9
LSD 3.4 3.8 4.3 4.7 5.9 6.3 6.0 5.3 5.3 4.5 4.1 2.4 1.6 1.6 1.5
Hallucinogens 1.3 1.4 1.7 2.2 2.7 3.2 3.2 3.1 2.9 2.8 ‡ 4.0 3.7 3.6 3.6 3.4
other than LSD
Ecstasy (MDMA), ― ― ― ― ― 3.1 3.4 2.9 3.7 5.3 6.0 4.9 3.1 2.6 2.4
d original
MDMA, revised ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Salvia ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Cocaine 2.2 2.1 2.3 2.8 3.3 4.0 4.3 4.5 4.5 3.9 3.5 3.7 3.3 3.5 3.5
Crack 1.0 1.1 1.2 1.5 1.8 2.0 2.1 2.4 2.2 2.1 1.8 2.0 1.8 1.7 1.6
Other cocaine 2.0 1.8 2.0 2.3 2.8 3.4 3.7 3.7 4.0 3.3 3.0 3.1 2.8 3.1 3.0
Heroin 0.5 0.6 0.6 0.9 1.2 1.3 1.3 1.2 1.3 1.3 0.9 1.0 0.8 0.9 0.8
With a needle ― ― ― ― 0.7 0.7 0.7 0.7 0.7 0.5 0.5 0.5 0.5 0.5 0.5
Without a needle ― ― ― ― 0.9 0.9 1.0 0.9 1.0 1.1 0.7 0.7 0.6 0.7 0.7
OxyContin ― ― ― ― ― ― ― ― ― ― ― 2.7 3.2 3.3 3.4
Vicodin ― ― ― ― ― ― ― ― ― ― ― 6.0 6.6 5.8 5.7
Amphetamines c 7.5 7.3 8.4 9.1 10.0 10.4 10.1 9.3 9.0 9.2 9.6 8.9 8.0 7.6 7.0
Ritalin ― ― ― ― ― ― ― ― ― ― 4.2 3.8 3.5 3.6 3.3
Adderall ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Methamphetamine ― ― ― ― ― ― ― ― 4.1 3.5 3.4 3.2 3.0 2.6 2.4
Bath salts ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
(synthetic
stimulants)
Tranquilizers 2.8 2.8 2.9 3.1 3.7 4.1 4.1 4.4 4.4 4.5 ‡ 5.5 5.3 4.8 4.8 4.7
OTC cough/cold ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
medicines
Rohypnol ― ― ― ― ― 1.1 1.1 1.1 0.8 0.7 0.9 ‡ 0.8 0.8 0.9 0.8
GHB b ― ― ― ― ― ― ― ― ― 1.4 1.2 1.2 1.2 1.1 0.8
Ketamine b ― ― ― ― ― ― ― ― ― 2.0 1.9 2.0 1.7 1.3 1.0
Alcohol 67.4 66.3 59.7 60.5 60.4 60.9 61.4 59.7 59.0 59.3 58.2 55.3 54.4 54.0 51.9
‡
Been drunk 35.8 34.3 34.3 35.0 35.9 36.7 36.9 35.5 36.0 35.9 35.0 32.1 31.2 32.5 30.8
Flavored alcoholic ― ― ― ― ― ― ― ― ― ― ― ― ― 44.5 43.9
beverages
Alcoholic ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
beverages
containing
caffeine
Any vaping ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Vaping nicotine ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Vaping marijuana ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Vaping just ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
flavoring
Dissolvable ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
tobacco products
Snus ― ― ― ― ― ― ― ― ― ― ― ― ― ― ―
Steroids 1.2 1.1 1.0 1.2 1.3 1.1 1.2 1.3 1.7 1.9 2.0 2.0 1.7 1.6 1.3
PEAK YEAR–2017
2016– CHANGE
2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2017
CHANGE Absolute
Change
Any illicit drug c 25.8 24.8 24.9 25.9 27.3 27.6 27.1 28.6 27.2 26.8 25.3 26.5 +1.2 -0.7
‡
Any illicit drug 12.7 12.4 11.9 11.6 11.8 11.3 10.8 11.4‡ 10.9 10.5 9.7 9.4 -0.3 -1.5 ss
other than
marijuana c
Any illicit drug 28.7 27.6 27.6 28.5 29.7 29.8 29.0 30.5 28.5 28.4 26.3 28.3 +2.0 ss -0.2
including ‡
inhalants c
Marijuana/hashish 22.0 21.4 21.5 22.9 24.5 25.0 24.7 25.8 24.2 23.7 22.6 23.9 +1.3 s -6.2 sss
Synthetic ― ― ― ― ― ― 8.0 6.4 4.8 4.2 3.1 2.8 -0.4 s -5.2 sss
marijuana
Inhalants 6.9 6.4 6.4 6.1 6.0 5.0 4.5 3.8 3.6 3.2 2.6 2.9 +0.2 -7.3 sss
Hallucinogens 3.6 3.8 3.8 3.5 3.8 3.7 3.2 3.1 2.8 2.8 2.8 2.7 0.0 -3.2 sss
LSD 1.4 1.7 1.9 1.6 1.8 1.8 1.6 1.6 1.7 1.9 2.0 2.1 +0.1 -4.3 sss
Ecstasy (MDMA), 2.7 3.0 2.9 3.0 3.8 3.7 2.5 2.8 2.2 ― ― ― ― ―
d original
MDMA, revised ― ― ― ― ― ― ― ― 3.4 2.4 1.8 1.7 -0.1 -1.6 sss
Salvia ― ― ― ― 3.5 3.6 2.7 2.3 1.4 1.2 1.2 0.9 -0.3 ss -2.7 sss
Cocaine 3.5 3.4 2.9 2.5 2.2 2.0 1.9 1.8 1.6 1.7 1.4 1.6 +0.2 -2.9 sss
Crack 1.5 1.5 1.3 1.2 1.1 1.0 0.9 0.8 0.7 0.8 0.6 0.7 +0.1 -1.7 sss
Other cocaine 3.1 2.9 2.6 2.1 1.9 1.7 1.7 1.5 1.5 1.5 1.2 1.3 +0.1 -2.7 sss
Heroin 0.8 0.8 0.8 0.8 0.8 0.7 0.6 0.6 0.5 0.4 0.3 0.3 0.0 -1.0 sss
With a needle 0.5 0.5 0.5 0.5 0.6 0.5 0.4 0.4 0.4 0.3 0.3 0.2 0.0 -0.5 sss
Without a needle 0.6 0.7 0.6 0.5 0.6 0.5 0.4 0.4 0.3 0.3 0.2 0.2 0.0 -0.9 sss
OxyContin 3.5 3.5 3.4 3.9 3.8 3.4 2.9 2.9 2.4 2.3 2.1 1.9 -0.2 -2.0 sss
Vicodin 6.3 6.2 6.1 6.5 5.9 5.1 4.3 3.7 3.0 2.5 1.8 1.3 -0.5 -5.2 sss
Amphetamines c 6.8 6.5 5.8 5.9 6.2 5.9 5.6 7.0 ‡ 6.6 6.2 5.4 5.0 -0.4 -1.6 sss
Ritalin 3.5 2.8 2.6 2.5 2.2 2.1 1.7 1.7 1.5 1.4 1.1 0.8 -0.2 -3.4 sss
Adderall ― ― ― 4.3 4.5 4.1 4.4 4.4 4.1 4.5 3.9 3.5 -0.3 -0.5 s
Methamphetamine 2.0 1.4 1.3 1.3 1.3 1.2 1.0 1.0 0.8 0.6 0.5 0.5 0.0 -3.6 sss
Bath salts ― ― ― ― ― ― 0.9 0.9 0.8 0.7 0.8 0.5 -0.3 s -0.4 s
(synthetic
stimulants)
Tranquilizers 4.6 4.5 4.3 4.5 4.4 3.9 3.7 3.3 3.4 3.4 3.5 3.6 +0.1 -1.9 sss
OTC cough/cold 5.4 5.0 4.7 5.2 4.8 4.4 4.4 4.0 3.2 3.1 3.2 3.0 -0.2 -2.4 sss
medicines
Rohypnol 0.7 0.8 0.7 0.6 0.8 0.9 0.7 0.6 0.5 0.5 0.7 0.5 -0.2 s -0.5 sss
GHB b 0.9 0.7 0.9 0.9 0.8 0.8 ― ― ― ― ― ― ― ―
Ketamine b 1.1 1.0 1.2 1.3 1.2 1.2 ― ― ― ― ― ― ― ―
Alcohol 50.7 50.2 48.7 48.4 47.4 45.3 44.3 42.8 40.7 39.9 36.7 36.7 0.0 -24.7 sss
Been drunk 30.7 29.7 28.1 28.7 27.1 25.9 26.4 25.4 23.6 22.5 20.7 20.4 -0.3 -16.5 sss
Flavored alcoholic 42.4 40.8 39.0 37.8 35.9 33.7 32.5 31.3 29.4 28.8 25.3 25.9 +0.5 -18.6 sss
beverages
Alcoholic ― ― ― ― ― 19.7 18.6 16.6 14.3 13.0 11.2 10.6 -0.6 -9.1 sss
beverages
containing
caffeine
Any vaping ― ― ― ― ― ― ― ― ― ― ― 21.5 ― ―
Vaping nicotine ― ― ― ― ― ― ― ― ― ― ― 13.9 ― ―
Vaping marijuana ― ― ― ― ― ― ― ― ― ― ― 6.8 ― ―
Vaping just ― ― ― ― ― ― ― ― ― ― ― 17.2 ― ―
flavoring
Dissolvable ― ― ― ― ― ― 1.4 1.4 1.2 1.1 0.9 0.9 0.0 -0.5
tobacco products
Snus ― ― ― ― ― ― 5.6 4.8 4.1 3.8 3.6 2.6 -1.0 sss -3.0 sss
Steroids 1.3 1.1 1.1 1.0 0.9 0.9 0.9 0.9 0.9 1.0 0.8 0.8 0.0 -1.2 sss
a The proportional change is the percent by which the most recent year deviates from the peak
year (or the low year) for the drug in question. Thus, if a drug was at 20% prevalence in the peak
year and declined to 10% prevalence in the most recent year, this would reflect a proportional
decline of 50%.
b Question was discontinued among 8th and 10th graders in 2012.
c In 2013, for the questions on the use of amphetamines, the text was changed on 2 of the
questionnaire forms for 8th and 10th graders and 4 of the questionnaire forms for 12th graders.
This change also impacted the any illicit drug indices. Data presented here include only the
changed forms beginning in 2013.
d In 2014, the text was changed on 1 of the questionnaire forms for 8th, 10th, and 12th graders to
include “Molly” in the description. The remaining forms were changed in 2015. Data for both
versions of the question are presented here.
considered in the differential diagnosis of a teen with an altered sensorium
(Table 140.5 ). An adolescent presenting to an emergency setting with an
impaired sensorium should be evaluated for substance use as a part of the
differential diagnosis (Table 140.6 ). Screening for substance use is
recommended for patients with psychiatric and behavioral diagnoses. Other
clinical manifestations of substance use are associated with the route of use;
intravenous drug use is associated with venous “tracks” and needle marks, and
nasal mucosal injuries are associated with nasal insufflation of drugs. Seizures
can be a direct effect of drugs such as cocaine, synthetic marijuana, and
amphetamines or an effect of drug withdrawal in the case of barbiturates or
tranquilizers.
Table 140.5
Common Names and Salient Features of Club Drugs Used
Recreationally
γ- γ- 1,4-
MDMA EPHEDRINE KETAMINE
HYDROXYBUTYRATE BUTYROLACTONE BUTANEDIOL
Common Ecstasy, Herbal Liquid Ecstasy, goop Blue nitro, longevity, Thunder nectar, K, special K,
name XTC, Ecstasy, herbal soap, Georgia homeboy, revivarant, GH serenity, pine vitamin K,
E, X, fuel, zest grievous bodily harm revitalizer, gamma G, needle extract, ket, kat
Adam, nitro, insom-X, zen, enliven,
hug remforce, firewater, revitalize plus,
drug, invigorate lemon drops
Molly
Duration of 4-6 hr 4-6 hr 1.5-3.5 hr 1.5-3.5 hr 1.5-3.5 hr 1-3 hr
action
Elimination 8-9 hr 5-7 hr 27 min ND ND 2 hr
half-life
Peak plasma 1-3 hr 2-3 hr 20-60 min* 15-45 min 15-45 min 20 min
concentration
Physical No No Yes Yes Yes No
dependence
Antidote No No No No No No
cross-reactions.
‡ Flunitrazepam can give positive results for benzodiazepines; ketamine can give positive results
for phencyclidine.
DEA, U.S. Drug Enforcement Agency, currently reviewing possibility of flunitrazepam being placed
into schedule of the U.S. Controlled Substance Act; GC/MS, gas chromatography–mass
spectroscopy. Duration, half-life, and peak plasma are probably different after high or sequential
doses because of nonlinear kinetics; ND, not determined in humans.
Modified from Ricaurte GA, McCann UD: Recognition and management of complications of new
recreational drug use. Lancet 365:2137–2145, 2005.
Table 140.6
Most Common Toxic Syndromes
ANTICHOLINERGIC SYNDROMES
Common Delirium with mumbling speech, tachycardia, dry, flushed skin, dilated pupils, myoclonus, slightly
signs elevated temperature, urinary retention, and decreased bowel sounds. Seizures and dysrhythmias may
occur in severe cases.
Common Antihistamines, antiparkinsonian medication, atropine, scopolamine, amantadine, antipsychotic agents,
causes antidepressant agents, antispasmodic agents, mydriatic agents, skeletal muscle relaxants, and many
plants (notably jimsonweed and Amanita muscaria ).
SYMPATHOMIMETIC SYNDROMES
Common Delusions, paranoia, tachycardia (or bradycardia if the drug is a pure α-adrenergic agonist),
signs hypertension, hyperpyrexia, diaphoresis, piloerection, mydriasis, and hyperreflexia. Seizures,
hypotension, and dysrhythmias may occur in severe cases.
Common Cocaine, amphetamine, methamphetamine (and its derivatives 3,4-methylenedioxyamphetamine, 3,4-
causes methylenedioxymethamphetamine, 3,4-methylenedioxyethamphetamine, and 2,5-dimethoxy-4-
bromoamphetamine), some synthetic marijuana, and OTC decongestants (phenylpropanolamine,
ephedrine, and pseudoephedrine). In caffeine and theophylline overdoses, similar findings, except for
the organic psychiatric signs, result from catecholamine release.
OPIATE, SEDATIVE, OR ETHANOL INTOXICATION
Common Coma, respiratory depression, miosis, hypotension, bradycardia, hypothermia, pulmonary edema,
signs decreased bowel sounds, hyporeflexia, and needle marks. Seizures may occur after overdoses of some
narcotics, notably propoxyphene.
Common Narcotics, barbiturates, benzodiazepines, ethchlorvynol, glutethimide, methyprylon, methaqualone,
causes meprobamate, ethanol, clonidine, and guanabenz.
CHOLINERGIC SYNDROMES
Common Confusion, central nervous system depression, weakness, salivation, lacrimation, urinary and fecal
signs incontinence, gastrointestinal cramping, emesis, diaphoresis, muscle fasciculations, pulmonary edema,
miosis, bradycardia or tachycardia, and seizures.
Common Organophosphate and carbamate insecticides, physostigmine, edrophonium, and some mushrooms.
causes
From Kulig K: Initial management of ingestions of toxic substances, N Engl J Med 326:1678,
1992. ©1992 Massachusetts Medical Society. All rights reserved.
Screening for Substance Abuse
Disorders
In a primary care setting the annual health maintenance examination provides an
opportunity for identifying adolescents with substance use or abuse issues. The
direct questions as well as the assessment of school performance, family
relationships, and peer activities may necessitate a more in-depth interview if
there are suggestions of difficulties in those areas. Several self-report screening
questionnaires also are available, with varying degrees of standardization,
length, and reliability. The CRAFFT mnemonic is specifically designed to
screen for adolescents' substance use in the primary setting (Table 140.7 ).
Privacy and confidentiality must be established when asking the teen about
specifics of their substance experimentation or use. Interviewing the parents can
provide additional perspective on early warning signs that go unnoticed or
disregarded by the teen. Examples of early warning signs of teen substance use
are change in mood, appetite, or sleep pattern; decreased interest in school or
school performance; loss of weight; secretive behavior about social plans; or
valuables such as money or jewelry missing from the home. The use of urine
drug screening is recommended when select circumstances are present: (1)
psychiatric symptoms to rule out comorbidity or dual diagnoses, (2) significant
changes in school performance or other daily behaviors, (3) frequently occurring
accidents, (4) frequently occurring episodes of respiratory problems, (5)
evaluation of serious motor vehicular or other injuries, and (6) as a monitoring
procedure for a recovery program. Table 140.8 shows common tests used for
detection by substance, along with the approximate retention time between use
and identification in the urine. Most initial screening uses an immunoassay
method, such as the enzyme-multiplied immunoassay technique, followed by a
confirmatory test using highly sensitive, highly specific gas chromatography–
mass spectrometry. The substances that can cause false-positive results should be
considered, especially when there is a discrepancy between the physical findings
and the urine drug screen result. In 2007 the American of Academy of Pediatrics
(AAP) released guidelines that strongly discourage routine home-based or
school-based testing.
Table 140.7
CRAFFT Mnemonic Tool
• Have you ever ridden in a C ar driven by someone (including yourself) who
was high or had been using alcohol or drugs?
• Do you ever use alcohol or drugs to R elax, feel better about yourself or fit
in?
• Do you ever use alcohol or drugs while you are by yourself (A lone)?
• Do you ever F orget things you did while using alcohol or drugs?
• Do your Family or F riends ever tell you that you should cut down on your
drinking or drug use?
• Have you ever gotten into T rouble while you were using alcohol or drugs?
From the Center for Adolescent Substance Abuse Research (CeASAR): The
CRAFFT screening interview. (Copyright John R. Knight, MD, Boston
Children's Hospital, 2015.)
Table 140.8
Urine Screening for Drugs Commonly Abused by
Adolescents
APPROXIMATE
MAJOR FIRST SECOND
DRUG INITIAL RETENTION
METABOLITE CONFIRMATION CONFIRMATION
TIME
Alcohol (blood) Acetaldehyde GC IA 7-10 hr
Alcohol (urine) Acetaldehyde GC IA 10-13 hr
Amphetamines TLC IA GC, GC/MS 48 hr
Barbiturates IA TLC GC, GC/MS Short-acting (24
hr); long-acting (2-
3 wk)
Benzodiazepines IA TLC GC, GC/MS 3 days
Cannabinoids Carboxy- and IA TLC GC/MS 3-10 days
hydroxymetabolites (occasional user);
1-2 mo (chronic
user)
Cocaine Benzoylecgonine IA TLC GC/MS 2-4 days
Methaqualone Hydroxylated TLC IA GC/MS 2 wk
metabolites
Opiates
Heroin Morphine IA TLC GC, GC/MS 2 days
Glucuronide
Morphine Morphine IA TLC GC, GC/MS 2 days
Glucuronide
Codeine Morphine IA TLC GC, GC/MS 2 days
Glucuronide
Phencyclidine TLC IA GC, GC/MS 8 days
GC, Gas chromatography; IA, immunoassay; MS, mass spectrometry; TLC, thin-layer
chromatography.
Modified from Drugs of abuse—urine screening [physician information sheet], Los Angeles,
Pacific Toxicology. From MacKenzie RG, Kipke MD: Substance use and abuse. In Friedman SB,
Fisher M, Schonberg SK, editors: Comprehensive adolescent health care, St Louis, 1998, Mosby.
Diagnosis
The Diagnostic and Statistical Manual of Mental Disorders (DSM-5) no longer
identifies substance use disorders as those of abuse or of dependence . A
substance use disorder is defined by a cluster of cognitive, behavioral, and
physiologic symptoms that indicate that an adolescent is using a substance even
though there is evidence that the substance is harming the adolescent. Even after
detoxification, a substance use disorder may leave persisting changes in brain
circuits with resulting behavioral changes. There are 11 criteria that describe a
pathologic pattern of behaviors related to use of the substance, falling into 4
categories: impaired control, social impairment, increased risk, and
pharmacologic criteria. The 1st category, impaired control , describes an
individual taking increasing amounts of the substance who expresses a persistent
desire to decrease use, with unsuccessful efforts. The individual may spend a
great deal of time obtaining the substance, using the substance, or recovering
from its effects and expresses an intense desire for the drug, usually in settings
where the drug had been available, such as a specific type of social situation.
The 2nd cluster of criteria (5-7) reflects social impairment, including the
inability to perform as expected in school, at home, or at a job; increasing social
problems; and withdrawing from the family. The 3rd cluster of 2 criteria
addresses increased risk associated with use of the substance, and the 4th
cluster includes 2 criteria addressing pharmacologic responses (tolerance
and/or withdrawal). The total number of criteria present is associated with a
determination of a mild, moderate, or severe disorder.
These criteria may have limitations with adolescents because of differing
patterns of use, developmental implications, and other age-related consequences.
Adolescents who meet diagnostic criteria should be referred to a program for
substance use disorder treatment unless the primary care physician has additional
training in addiction medicine.
assistance from a health professional qualified in substance abuse management
should be obtained.
Prevention
Preventing drug use among children and teens requires prevention efforts aimed
at the individual, family, school, and community levels. The National Institute on
Drug Abuse (NIDA) of the U.S. National Institutes of Health has identified
essential principles of successful prevention programs. Programs should enhance
protective factors (parent support) and reduce risk factors (poor self-control);
should address all forms of drug abuse (legal and illegal); should address the
specific type(s) of drug abuse within an identified community; and should be
culturally competent to improve effectiveness (Table 140.9 ). The highest-risk
periods for substance use in children and adolescents are during life transitions,
such as the move from elementary school to middle school, or from middle
school to high school. Prevention programs need to target these emotionally and
socially intense times for teens to adequately anticipate potential substance use
or abuse. Examples of effective research-based drug abuse prevention programs
featuring a variety of strategies are listed on the NIDA website
(www.drugabuse.gov ), and on the Center for Substance Abuse Prevention
website (www.prevention.samhsa.gov ).
Table 140.9
140.1
(50.8%); higher among 11th-grade female (72.1%) and 12th-grade female
(75.2%) than 9th-grade female (53.0%) and higher among 10th-grade male
(58.8%), 11th-grade male (68.7%), and 12th-grade male (71.5%) than 9th-grade
male (48.9%) students.
Multiple factors can affect a young teen's risk of developing a drinking
problem at an early age (Table 140.10 ). One third of high school seniors admit
to combining drinking behaviors with other risky behaviors, such as driving or
taking additional substances. Binge drinking remains especially problematic
among the older teens and young adults; 31% of high school seniors report
having 5 or more drinks in a row in the last 30 days. Higher use is seen in males
(23.8%) than in females (19.8%), and whites (24.0%) and Hispanics (24.2%)
than in blacks (12.4%). Teens with binge-drinking patterns are more likely to be
assaulted, engage in high-risk sexual behaviors, have academic problems, and be
injured than those teens without binge drinking patterns.
Table 140.10
Risk Factors for a Teen Developing a
Drinking Problem
Family Risk Factors
Diagnosis
Primary care settings provide the opportunity to screen teens for alcohol use or
problem behaviors. Brief alcohol screening instruments such as CRAFFT (see
Table 140.7 ) or AUDIT (Alcohol Use Disorders Identification Test, Table
140.11 ) perform well in a clinical setting as techniques to identify alcohol use
disorders. A score of ≥8 on the AUDIT questionnaire identifies people who drink
excessively and who would benefit from reducing or ceasing drinking. Teenagers
in the early phases of alcohol use exhibit few physical findings. Recent use of
alcohol may be reflected in elevated GGT and aspartate transaminase levels.
Table 140.11
Alcohol Use Disorders Identification Test (AUDIT)
SCORE (0-4)*
1. How often do you have a drink containing alcohol? Never (0) to more than
4 per wk (4)
2. How many drinks containing alcohol do you have on a typical day? One or 2 (0) to more
than 10 (4)
3. How often do you have 6 or more drinks on 1 occasion? Never (0) to daily or
almost daily (4)
4. How often during the last year have you found that you were not able to stop drinking Never (0) to daily or
once you had started? almost daily (4)
5. How often during the last year have you failed to do what was normally expected Never (0) to daily or
from you because of drinking? almost daily (4)
6. How often during the last year have you needed a first drink in the morning to get Never (0) to daily or
yourself going after a heavy drinking session? almost daily (4)
7. How often during the last year have you had a feeling of guilt or remorse after Never (0) to daily or
drinking? almost daily (4)
8. How often during the last year have you been unable to remember what happened the Never (0) to daily or
night before because you had been drinking? almost daily (4)
9. Have you or someone else been injured as a result of your drinking? No (0) to yes, during
the last year (4)
10. Has a relative, friend, doctor or other health worker been concerned about your No (0) to yes, during
drinking or suggested that you should cut down? the last year (4)
responsive to messages that emphasize the effects of tobacco use on appearance,
breath, and sports performance; lack of benefit for weight loss; monetary cost of
tobacco addiction; and deceptive marketing by the tobacco industry.
The approach to smoking cessation in adolescents includes the 5 A s (ask,
advise, assess, assist, and arrange) and use of nicotine replacement therapy
(NRT) in addicted teens who are motivated to quit. Consensus panels
recommend the 5 As, although evidence of efficacy in adolescents is limited.
Studies of the NRT patch in adolescents suggest a positive effect on reducing
withdrawal symptoms; pharmacotherapy should be combined with behavioral
therapy to increase cessation and lower relapse rates. In a limited number of
studies, cessation rates of 15% were reported at 3 and 6 mo. NRT is also
available as a gum, inhaler, nasal spray, lozenge, or microtab (Table 140.12 ).
Medications such as bupropion and varenicline improve smoking cessation rates
in adults but are not FDA approved for use in adolescents <18 yr old.
Preliminary studies in adolescents report cessation efficacy with 150 mg of
bupropion twice daily. In postmarketing surveillance, suicidal ideation and
suicide have been reported among patients taking bupropion and varenicline.
Table 140.12
Smoking Cessation Pharmacotherapy Available in the
United States
FDA-
THERAPY APPROVED STUDIED IN QUIT
NAME STRENGTHS ADULT AVAILABILITY*
BRAND ADOLESCENTS DATE
DOSING
NICOTINE REPLACEMENT THERAPY
Gum ‡ Nicorette 2 mg, 4 mg The 4-mg OTC* Yes
strength
should be
used by
patients
who smoke
≥25
cigarettes a
day;
otherwise,
2-mg
strength
should be
used.
Wk 1-6: 1
piece every
1-2 hr
Wk 7-9: 1
piece every
2-4 hr
Wk 10-12:
1 piece
every 4-8 hr
Inhaler Nicotrol 4 mg 6-16 cartridges a Rx No
Inhaler day for up to 12
wk
Lozenge Commit, 2 mg, 4 mg The 4-mg OTC No Prior to
Nicorette strength beginning
mini should be nicotine
used by replacement
patients therapy
who smoke
1st cigarette
within 30
min of
waking;
otherwise,
2-mg
strength
should be
used.
Wk 1-6: 1
lozenge
every 1-2 hr
Wk 7-9: 1
lozenge
every 2-4 hr
Wk 10-12:
1 lozenge
every 4-8 hr
Nasal Spray Nicotrol 0.5 mg/spray 1-2 sprays/hr up Rx Yes
NS to a maximum
of 80 sprays per
day
‡ Generics available.
†
None is FDA approved for use in patients younger than 18 yr.
From JP Karpinski et al: Smoking cessation treatment for adolescents, J Pediatr Pharmacol Ther
15(4):249–260, 2010.
Bibliography
report a higher use of cannabis “edibles.” It is important to recognize that as
perceived harm drops, marijuana use increases (Fig. 140.4 ).
FIG. 140.4 As the perceived harm of marijuana drops, use goes up. The
36.4% using in 2013 equates to about 11 students in the average class.
(From NIH National Institute on Drug Abuse.)
Clinical Manifestations
In addition to the “desired” effects of elation and euphoria, marijuana may cause
impairment of short-term memory, poor performance of tasks requiring divided
attention (e.g., those involved in driving), loss of critical judgment, decreased
coordination, and distortion of time perception (Table 140.13 ). Visual
hallucinations and perceived body distortions occur rarely, but “flashbacks” or
recall of frightening hallucinations experienced under marijuana's influence may
occur, usually during stress or with fever.
Table 140.13
Acute and Chronic Adverse Effects of
Cannabis Use
Acute Adverse Effects
Keywords
huffing
Clinical Manifestations
The major effects of inhalants are psychoactive (Table 140.15 ). The intoxication
lasts only a few minutes, so a typical user will huff repeatedly over an extended
period (hours) to maintain the high. The immediate effects of inhalants are
similar to alcohol: euphoria, slurred speech, decreased coordination, and
dizziness. Toluene , the main ingredient in model airplane glue and some rubber
cements, causes relaxation and pleasant hallucinations for up to 2 hr. Euphoria is
followed by violent excitement; coma may result from prolonged or rapid
inhalation. Volatile nitrites , such as amyl nitrite, butyl nitrite, and related
compounds marketed as room deodorizers, are used as euphoriants, enhancers of
musical appreciation, and sexual enhancements among older adolescents and
young adults. They may result in headaches, syncope, and lightheadedness;
profound hypotension and cutaneous flushing followed by vasoconstriction and
tachycardia; transiently inverted T waves and depressed ST segments on
electrocardiography; methemoglobinemia; increased bronchial irritation; and
increased intraocular pressure. There may be dermatologic findings, including
perianal/perioral dermatitis (“huffer rash”), frostbite, and contact dermatitis, as
well as epistaxis, nasal ulcers, and conjunctivitis.
Table 140.15
Stages in Symptom Development After Use of Inhalants
STAGE SYMPTOMS
1: Excitatory Euphoria, excitation, exhilaration, dizziness, hallucinations, sneezing, coughing, excess
salivation, intolerance to light, nausea and vomiting, flushed skin and bizarre behavior
2: Early CNS Confusion, disorientation, dullness, loss of self-control, ringing or buzzing in the head, blurred or
depression double vision, cramps, headache, insensitivity to pain, and pallor or paleness
3: Medium Drowsiness, muscular uncoordination, slurred speech, depressed reflexes, and nystagmus or rapid
CNS involuntary oscillation of the eyeballs
depression
4: Late CNS Unconsciousness that may be accompanied by bizarre dreams, epileptiform seizures, and EEG
depression changes
CNS, Central nervous system; EEG, electroencephalogram.
From Harris D: Volatile substance abuse, Arch Dis Child Educ Pract Ed 91:ep93-ep100, 2006.
Complications
Model airplane glue is responsible for a wide range of complications, related to
chemical toxicity, to the method of administration (in plastic bags, with resultant
suffocation), and to the dangerous setting in which the inhalation occurs (inner-
city roof tops). Common neuromuscular changes reported in chronic inhalant
abusers include difficulty coordinating movement, gait disorders, muscle
tremors, and spasticity, particularly in the legs (Table 140.16 ). Chronic use may
cause pulmonary hypertension, restrictive lung defects or reduced diffusion
capacity, peripheral neuropathy, hematuria, tubular acidosis, and possibly
cerebral and cerebellar atrophy. Chronic inhalant abuse has long been linked to
widespread brain damage and cognitive abnormalities that can range from mild
impairment (poor memory, decreased learning ability) to severe dementia. High-
frequency inhalant users were significantly more likely than moderate- and low-
frequency users to experience adverse consequences of inhalant intoxication,
such as behavioral, language, and memory problems. Certain risky behaviors
and consequences, such as engaging in unprotected sex or fighting while high on
inhalants, were dramatically more common among high-frequency than low-
frequency inhalant users. Death in the acute phase may result from cerebral or
pulmonary edema or myocardial involvement (Table 140.16 ).
Table 140.16
Diagnosis
Diagnosis of inhalant abuse is difficult because of the ubiquitous nature of the
products and decreased parental awareness of the dangers. In the primary care
setting, providers need to ask parents if they have witnessed any unusual
behaviors in their teen; noticed high-risk products in the teen's bedroom; seen
paint on the teen's hands, nose, or mouth; or found paint- or chemical-coated
rags. Complete blood count, coagulation studies, and hepatic and renal function
methamphetamine at least once, reflecting a steady decline in use.
Clinical Manifestations
Methamphetamine rapidly increases the release and blocks the reuptake of
dopamine, a powerful “feel good” neurotransmitter (Table 140.17 ). The effects
of amphetamines can be dose related. In small amounts, amphetamine effects
resemble other stimulants: increased physical activity, rapid and/or irregular
heart rate, increased blood pressure, and decreased appetite. High doses produce
slowing of cardiac conduction in the face of ventricular irritability. Hypertensive
and hyperpyrexic episodes can occur as seizures (see Table 140.6 ). Binge
effects result in the development of psychotic ideation with the potential for
sudden violence. Cerebrovascular damage, psychosis, severe receding of the
gums with tooth decay, and infection with HIV and hepatitides B and C can
result from long-term use. A withdrawal syndrome is associated with
amphetamine use, with early, intermediate, and late phases (Table 140.17 ). The
early phase is characterized as a “crash” phase with depression, agitation,
fatigue, and desire for more of the drug. Loss of physical and mental energy,
limited interest in the environment, and anhedonia mark the intermediate phase.
In the final phase, drug craving returns, often triggered by particular situations or
objects.
Table 140.17
Signs and Symptoms of Intoxication and Withdrawal
Treatment
Acute agitation and delusional behaviors can be treated with haloperidol or
droperidol. Phenothiazines are contraindicated and may cause a rapid drop in
blood pressure or seizure activity. Other supportive treatment consists of a
cooling blanket for hyperthermia and treatment of the hypertension and
arrhythmias, which may respond to sedation with lorazepam or diazepam. For
the chronic user, comprehensive CBT interventions have been demonstrated as
effective treatment options.
Bibliography
Setlik J, Bond R, Ho M. Adolescent prescription ADHA
medication abuse is rising along with prescriptions for these
medications. Pediatrics . 2009;124:875–880.
Spoth RL, Clair S, Shin C, et al. Long-term effects of universal
preventive interventions on methamphetamine use among
adolescents. Arch Pediatr Adolesc Med . 2006;160:876–882.
140.8
CHAPTER 142
Menstrual Problems
Krishna K. Upadhya, Gina S. Sucato
Table 142.1
Normal Menstruation
Data from many countries, including the United States, suggest that the average
age of menarche, or first menses, varies according to ethnic origin and
socioeconomic status. There is often a close concordance of the age at menarche
between mother and daughter, suggesting that genetic factors are determinants in
addition to individual factors such as weight, exercise level, and chronic medical
amenorrhea
polycystic ovary syndrome
PCOS
female athlete triad
FIG. 142.1 Initial diagnostic testing to evaluate amenorrhea. FSH, Follicle-stimulating
hormone; HCG, human chorionic gonadotropin; LH, luteinizing hormone; MRI,
magnetic resonance imaging; US, ultrasound.
Table 142.4
Laboratory Tests to Evaluate Patients With
Abnormal Uterine Bleeding
aggregation and von Willebrand factor multimers. Testing in the 1st 3 days of
menses and before any estrogen treatment is started minimizes the chances of
false-negative tests. Repeat testing can be warranted in patients for whom there
is a high pretest suspicion.
Table 142.5
Table 142.6
†
Adenomyosis is the presence of endometrial tissue within the uterine myometrium.
Table 142.7
Treatment for Dysmenorrhea
in patients with impaired renal or liver dysfunction, heart failure, or a history of GI bleeding or
ulcer. Full prescribing information can be found at:
http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020998s033 ,021156s003lbl.pdf.
DMPA, Depot medroxyprogesterone acetate; LARC, long-acting reversible contraceptive;
NSAIDs, nonsteroidal antiinflammatory drugs.
For those adolescents whose pain does not respond to optimally dosed
NSAIDs, or who also require contraception, the currently available forms of
hormonal contraception will improve dysmenorrhea. A number of trials have
investigated adjuvant treatments including heat, aromatherapy, acupressure,
acupuncture, transcutaneous nerve stimulation, herbal remedies, yoga, and
dietary supplements; however, the mainstay second-line treatment is hormones.
The mechanisms are not fully delineated but are presumed to include elimination
of progesterone production from the corpus luteum for those methods that
prevent ovulation, and decreased prostaglandin production from the diminished
endometrial lining. Up to 3 cycles may be required to appreciate the full benefit.
Methods and regimens that eliminate a placebo interval may provide better
relief. Females whose pain persists despite more than 3 mo of adequate
hormonal therapy require further evaluation and treatment.
Bibliography
Ryan SA. The treatment of dysmenorrhea. Pediatr Clin North
Am . 2017;64(2):331–342.
Youngster M, Laufer MR, Divasta AD. Endometriosis for the
primary care physician. Curr Opin Pediatr . 2013;25(4):454–
462.
142.4
Premenstrual Syndrome and
Premenstrual Dysphoric Disorder
Krishna K. Upadhya, Gina S. Sucato
Keywords
premenstrual syndrome
PMS
premenstrual dysphoric disorder
PMDD
Note: The symptoms in criteria A-C must have been met for most
menstrual cycles that occurred in the preceding year.
D. The symptoms are associated with clinically significant distress or
interference with work, school, usual social activities, or relationships with
others (e.g., avoidance of social activities; decreased productivity and
efficiency at work, school, or home).
E. The disturbance is not merely an exacerbation of the symptoms of another
disorder, such as major depressive disorder, panic disorder, persistent
depressive disorder (dysthymia), or a personality disorder (although it may
co-occur with any of these disorders).
F. Criterion A should be confirmed by prospective daily ratings during at least
2 symptomatic cycles. (Note: The diagnosis may be made provisionally
prior to this confirmation).
G. The symptoms are not attributable to the physiological effects of a
substance (e.g., a drug of abuse, a medication, other treatment) or another
medical condition (e.g., hyperthyroidism).
Bibliography
American Psychiatric Association. Diagnostic and statistical
manual of mental health disorders, ed 5 (DSM-5) . American
Psychiatric Publishing: Washington, DC; 2013.
Hofmeister S, Bodden S. Premenstrual syndrome and
premenstrual dysphoric disorder. Am Fam Physician .
2016;94(3):236–240.
Marjoribanks J, Brown J, O'Brien PM, et al. Selective serotonin
intrauterine devices (IUDs) and implants. Tier 2 methods have failure rates of 4-
7 pregnancies per 100 women in a year of typical use and include injectable
contraception, oral contraceptive pills, contraceptive patch, and vaginal ring.
Tier 3 methods have failure rates of >13 pregnancies per 100 women per year of
typical use and include the male and female condom, the diaphragm, withdrawal,
the sponge, fertility awareness–based methods, and spermicides.
FIG. 143.1 Effectiveness of contraceptive methods. (From Trussell J, Aiken ARA, Micks
E, Guthrie K. Contraceptive efficacy, safety, and personal considerations. In Hatcher
RA, Nelson AL, Trussell J, et al, eds. Contraceptive technology, ed 21, New York, 2018,
Ayer Company Publishers p. 102.)
Table 143.1
Efficacy of Contraceptives
FAILURE
RATE* SOME ADVERSE EFFECTS AND
METHOD SOME ADVANTAGES
Typical Perfect DISADVANTAGES
Use Use
Implant Convenience; long-term Irregular bleeding; removal complications
contraception; no patient
Nexplanon 0.1% 0.1% compliance required; rapid
return of fertility after
removal
Intrauterine Convenience; long-term Rare uterine perforation; risk of infection with
devices (lUDs) contraception; no patient insertion; anemia
compliance required; rapid
return of fertility after
removal
ParaGard 0.8% 0.6% Effective for 10 yr; Irregular/heavy bleeding and dysmenorrhea
T380A nonhormonal
Mirena 0.1% 0.1% Decreased menstrual Irregular bleeding in 1st 3-6 mo, followed by
bleeding and dysmenorrheal amenorrhea; ovarian cysts
Liletta 0.1% 0.1% Decreased menstrual Irregular bleeding in 1st 3-6 mo; ovarian cysts
bleeding and dysmenorrheal
Kyleena 0.2% 0.2% Smaller T-frame and Irregular bleeding in 1st 3-6 mo; ovarian cysts;
narrower insertion tube amenorrhea in 13% of users after 1 yr
Skyla 0.4% 0.3% Smaller T -frame and Irregular bleeding in 1st 3-6 mo; ovarian cysts;
narrower insertion tube amenorrhea in only 6% of users after 1 yr
Sterilization
Female 0.5% 0.5% Long-term contraception; no Potential for surgical complications; regret
patient compliance required among young women; reversal often not
possible and expensive
Male 0.15% 0.1% Long-term contraception; no Pain at surgical site, regret among young men;
patient compliance required reversal often not possible and expensive
Injectable Convenience; same as Delayed return to fertility, irregular bleeding
Depo-Provera 4% 0.2% progestin-only oral and amenorrhea; weight gain; may decrease
contraceptives bone mineral density
Combination 7% 0.3% Protection against ovarian Increased rate of thromboembolism, stroke, and
oral and endometrial cancer, PID, myocardial infarction in older smokers; nausea;
contraceptives and dysmenorrhea headache; contraindicated with breastfeeding
Progestin-only 7% 0.3% Protection against PID. iron- Irregular, unpredictable bleeding; must take at
oral deficiency anemia, and same time every day
contraceptives dysmenorrhea; safe in
breastfeeding women and
those with cardiovascular
risk
Transdermal Convenience of once-weekly Dysmenorrhea and breast discomfort may be
Evra 7% 0.3% application; same benefits as more frequent than with oral contraceptives;
combination oral application site reactions; detachment;
contraceptives increased estrogen exposure compared to oral
contraceptives
Vaginal Excellent cycle control; rapid Discomfort; vaginal discharge
NuvaRing 7% 0.3% return to fertility after
removal; convenience of
once-monthly insertion
Diaphragm 17% 16% Low cost; may reduce risk of High failure rate; cervical irritation; increased
with cervical cancer risk of urinary tract infection and toxic shock
spermicide syndrome; some require fitting by healthcare
professional; may be difficult to obtain;
available only by prescription
Condom
without
spermicide
Female 21% 5% Protection against STIs; High failure rate; difficult to insert; poor
covers external genitalia; acceptability
OTC
Male 13% 2% Protection against STIs, OTC High failure rate; allergic reactions; poor
acceptability; breakage possible
Withdrawal 20% 4% No drugs or devices High failure rate
Sponge 14-27% 9-20% OTC; low cost; no fitting High failure rate; contraindicated during
required; provides 24 hr of menses; increased risk of toxic shock syndrome
protection
Fertility 15% - Low cost; no drugs or High failure rate; may be difficult to learn;
awareness– devices requires relatively long periods of abstinence
based methods
Standard Days 12% 5%
method
TwoDay 14% 4%
method
Ovulation 23% 3%
method
Symptothermal 2% 0.4%
method
Spermicide 21% 16% OTC High failure rate; local irritation; must be
alone reapplied with repeat intercourse; increased
risk of HIV transmission
No method 85% 85% - –
* Risk of unintended pregnancy during first year of use; data from Trussel J, et al: In Hatcher RA
et al: Contraceptive technology. ed 21, New York, 2018, Ayer Company Publishers.
STIs, Sexually transmitted infections; PID, pelvic inflammatory disease; OTC, over the counter.
Adapted from The Medical Letter : Choice of contraceptives. Med Lett 57(1477):128, 2015.
143.1
Contraceptive Use
Tara C. Jatlaoui, Yokabed Ermias, Lauren B. Zapata
Keywords
sexual activity
sexual debut
Tara C. Jatlaoui, Yokabed Ermias, Lauren B. Zapata
Keywords
LARC
intrauterine devices
implants
Intrauterine Devices
IUDs are small, flexible, plastic objects introduced into the uterine cavity
through the cervix. They differ in size, shape, and the presence or absence of
pharmacologically active substances. In the United States, 5 IUDs are currently
approved by the Food and Drug Administration (FDA): the CuT380A (Paragard)
and 4 LNG IUDs (Liletta, Kyleena, Mirena, and Skyla). The effectiveness of the
Cu IUD is enhanced by the copper ions released into the uterine cavity, with
possible mechanisms including inhibition of sperm transport and prevention of
implantation; this IUD is effective for at least 10 yr.
The LNG IUDs also have various actions, from thickening of cervical mucus
and inhibiting sperm survival to suppressing the endometrium. LNG IUDs are
effective for at least 3 and 5 years. All IUDs have typical-use failure rates of
<1% (see Fig. 143.1 ).
Common misconceptions of IUDs among healthcare providers are that IUDs
cause infections, infertility, and generally are not safe for teens or nulliparous
women to use; these misconceptions are a barrier to teens accessing these highly
effective and acceptable methods. IUDs do not increase risk of infertility and
may be inserted safely in teens as well as nulliparous women (category 2; see
Table 143.2 ).
Although early studies suggested an increased risk for upper genital tract
infection, theoretically as a result of passing a foreign body through the cervix,
newer work has refuted these earlier concerns. Therefore, clinicians are
encouraged to consider use of IUDs in adolescents despite relatively high
prevalence rates of STIs in this population. Teens should be screened for
gonorrhea and chlamydia at or before IUD placement, although placement
should not be delayed if results have not returned and there are no signs of
current infection (e.g., purulent discharge, erythematous cervix). If STI testing is
positive with an IUD in place, the patient may be treated without removing the
IUD if she wants to continue the method. Evidence from 2 systematic reviews
did not find benefit in routinely administering misoprostol to women undergoing
routine IUD placement to decrease pain or improve provider ease of insertion. A
2007.
143.4
Other Progestin-Only Methods
Tara C. Jatlaoui, Yokabed Ermias, Lauren B. Zapata
Keywords
Depo-Provera
depot medroxyprogesterone acetate
DMPA
progestin-only pills
mini pills
Several progestin-only contraceptive methods are available and include the LNG
IUDs and implant (see Chapter 143.3 ), as well as an injectable and progestin-
only pills. These methods do not contain estrogen and may be useful for teens
with contraindications to estrogen (Table 143.3 ) and are considered generally
safe for use in teens (category 1 or 2; see Table 143.2 ). Progestins thicken
cervical mucus to block sperm entry into the uterine cavity as well as induce an
atrophic endometrium leading to either amenorrhea or less menstrual blood loss;
the implant and injectable additionally suppress ovulation. Teens should be
provided anticipatory counseling regarding bleeding irregularities that may
normally occur in the 1st 3-6 mo of hormonal contraception use.
Table 143.3
Conditions Classified as Category 3 and 4 for
Combined Hormonal Contraceptive Use
Category 4
Complicated valvular heart disease
Current breast cancer
Severe decompensated cirrhosis
Deep venous thrombosis/pulmonary embolism (acute; history, not on
anticoagulation or on established therapy for at least 3 mo with higher
risk recurrence; major surgery with prolonged immobilization)
Complicated diabetes with nephropathy, retinopathy, neuropathy, or other
vascular disease or duration of diabetes >20 yr
Migraine with aura
Hypertension (blood pressure >160/100 mm Hg) or hypertension with
vascular disease
Ischemic heart disease (history of or current)
Hepatocellular adenoma
Malignant liver tumor
Peripartum cardiomyopathy (diagnosed <6 mo prior or with moderately or
severely impaired cardiac function)
Postpartum <21 days
History of cerebrovascular accident
Systemic lupus erythematosus with positive antiphospholipid antibodies
Thrombogenic mutations
Viral hepatitis (acute or flare)
Category 3
Depo-Provera
An injectable progestin, depot medroxyprogesterone acetate (DMPA , Depo-
Provera) is a Tier 2 contraceptive method available as a deep intramuscular (IM)
injection (150 mg), or as a subcutaneous (SC) injection (104 mg) with typical-
use failure rates of 4% (see Table 143.1 ). Both preparations must be
readministered every 3 mo (13 wk) and act to inhibit ovulation. DMPA is
particularly attractive for adolescents who have difficulty with compliance, are
intellectually or physically impaired, and are chronically ill or have a condition
for which estrogen use is not recommended. Common concerns with DMPA
include bleeding changes, bone effects, and weight gain. After 1 yr of use, 50%
of DMPA users develop amenorrhea, which may be an added advantage for teens
with heavy menstrual bleeding, dysmenorrhea, anemias, or blood dyscrasias, or
for those with impairments that make hygiene difficult. Although studies have
demonstrated bone mineral density (BMD) loss in adolescents, potentially
increasing their risk for osteoporosis later in life, other studies have found that
BMD is recovered after discontinuation of this method, and it is thus considered
safe for use in this population. Healthcare providers may want to consider a
contraceptive containing estrogen in teens who are already at high risk for low
BMD, such as those receiving chronic corticosteroid therapy or those with eating
disorders (see Chapter 726 ). Although the FDA issued a black box warning in
2004, AAP and ACOG do not recommend limiting DMPA use to 2 yr for all
women and do not recommend routine BMD screening for females using
DMPA. Early weight gain may be predictive of progressive gain over time; thus
those teens gaining weight in the 1st 3-6 mo should consider another method.
Progestin-Only Pills
Progestin-only oral contraceptive pills (POPs ) are available for the adolescent
in whom the use of estrogen is potentially harmful, such those with active liver
disease, replaced cardiac valves, or hypercoagulable states (see Table 143.3 ).
POPs (mini pills ) are quickly effective after 2 days of initiation in thickening
cervical mucus, but are less reliable in inhibiting ovulation. Effects are short-
eliminated by interventions known collectively as emergency contraception (EC
) up to 120 hr after unprotected intercourse or contraceptive failure. Table 143.4
lists the indications for use of EC. EC methods include the Cu IUD and
emergency contraceptive pills, which include ulipristal acetate, levonorgestrel
(LNG), and COCs following the Yuzpe method. Although the mechanism of
action of the Cu IUD as EC is unclear, all emergency contraceptive pills work to
delay ovulation and are effective only for intercourse that occurs before
administration. Initiation of a regular contraceptive method is necessary to
prevent pregnancy for any intercourse that occurs for the remainder of the cycle
and for future cycles. If pregnancy has already occurred, emergency
contraceptive pills will not cause an abortion or have teratogenic effects on the
fetus.
Table 143.4
Laboratory Diagnosis
Physical Changes
Substance Use
See also Chapter 140 .
Teenagers who abuse drugs, alcohol, and tobacco have higher pregnancy rates
than their peers. Most substance-abusing mothers appear to decrease or stop their
substance use while pregnant. Use begins to increase again about 6 mo
postpartum, complicating the parenting process and the mother's return to
school.
Repeat Pregnancy
In the United States, approximately 20% of all births to adolescent mothers (age
15-19) are second order or higher. Prenatal care is begun even later with a 2nd
pregnancy, and the 2nd infant is at higher risk of poor outcome than the 1st birth.
Mothers at risk of early repeat pregnancy (<2 yr) include those who do not
initiate long-acting contraceptives after the index birth, those who do not return
to school within 6 mo of the index birth, those with mood disorders, those
receiving major childcare assistance from the adolescent's mother, those who are
married or living with the infant's father, those having peers who were
adolescent parents, and those who are no longer involved with the baby's father
and who meet a new boyfriend who wants to have a child. To reduce repeat
pregnancy rates in these teens, programs must be tailored for this population,
preferably offering comprehensive healthcare for both the young mother and her
child (Table 144.2 ). Healthcare providers should remember to provide positive
reinforcement for teen parenting successes (i.e., compliment teen parents when
they are doing a good job).
Table 144.2
Primarily Females
Primarily Males
Types of Rape
Rape and sexual assault can occur in a variety of circumstances (Tables 145.2
and 145.3 ). A victim can be sexually assaulted or raped by someone they know
or a by stranger, though more often the assailant is someone known to the victim.
Understanding those circumstances allows for a more trauma-sensitive approach
and may impact the medical management and response to the patient. The
circumstances and relationship of the assailant to the victim may impact if,
when, and how a patient discloses. The gender of the victim may also affect
disclosure; transgendered people and males are uniformly less likely to disclose
rape/sexual assault than females. The gender of the assailant may be the same or
different than the victim's, and there may be one or more than one perpetrator. In
any scenario the sexual assault/rape can be facilitated by threats or coercion,
physical force, or drugs.
Table 145.2
Types of Nonstranger Rape
Acquaintance Rape
Most common form of rape for adolescents age 16-24 yr.
Assailant may be a neighbor, classmate, or friend of the family.
Victims are more likely to delay seeking medical care, may never report the
crime (males > females), and are less likely to proceed with criminal
prosecution even after reporting the incident(s).
Date Rape
Sexual Abuse
All sexual contact or exposure between an adult and a minor, or when there
is a significant age or developmental difference between the youth.
The assailant may be a relative, close family friend, or someone of
authority.
Statutory Rape
Sexual activity between an adult and an adolescent under the age of legal
consent, as defined by individual state law.
Based on the premise that below a certain age or beyond a specific age
difference with the assailant, an individual is not legally capable of giving
consent to engage in sexual intercourse.
The intent of such laws is to protect youth from being victimized, but they
may inadvertently lead a teenager to withhold pertinent sexual
information from a clinician for fear that her sexual partner will be
reported to the law.
Male Rape
Gang Rape
Drug-Facilitated Rape
Gang Rape
Urine and blood for date rape drugs (GHB, Rohypnol, ketamine)
From Centers for Disease Control and Prevention: Sexually transmitted diseases:
treatment guidelines 2015, MMWR Recomm Rep 64(RR-3):1–140, 2015, and
Updated guidelines for antiretroviral postexposure prophylaxis after sexual,
injection drug use, or other nonoccupational exposure to HIV—United States,
2016.
Treatment
Treatment includes prophylactic antimicrobials for STIs (see Chapter 146 )
and emergency contraception (see Chapter 143 ). The Centers for Disease
Control and Prevention (CDC) reports that trichomoniasis, bacterial vaginosis,
gonorrhea, and chlamydial infection are the most frequently diagnosed infections
among women who have been sexually assaulted. Antimicrobial prophylaxis is
recommended for adolescent rape victims because of the risk of acquiring an STI
and the risk of pelvic inflammatory disease (Table 145.5 ). A two- or three-drug
antiretroviral regimen for HIV postexposure prophylaxis (PEP) must be
considered and an infectious disease specialist consulted if higher transmission
risk factors are identified (e.g., knowing that the perpetrator is HIV-positive,
significant mucosal injury of the victim) to prescribe a triple-antiretroviral
regimen (Fig. 145.1 ). Similar considerations should be made for possible
exposure to the hepatitis B virus in vaccinated/unvaccinated individuals.
Clinicians should review the importance for patient's compliance with medical
and psychological treatment and follow-up care.
Table 145.5
Postexposure Prophylaxis (PEP) for Acute
Sexual Assault Victims
Routine
Recommended Regimen for STI Prophylaxis
As Indicated
Preferred regimen:
Tenofovir 300 mg and fixed-dose combination emtricitabine, 200
mg (Truvada) once daily
plus
Raltegravir 400 mg twice daily or
Dolutegravir 50 mg daily ‡
Alternative regimens available (The National Clinicians Consultation
Center is a resource for providers prescribing PEP, reachable at 1-888-
448-4911.)
Data from Centers for Disease Control and Prevention: Sexually transmitted
diseases: treatment guidelines 2015, MMWR Recomm Rep 64(RR-3):1–140,
2015, and Updated guidelines for antiretroviral postexposure prophylaxis after
sexual, injection drug use, or other nonoccupational exposure to HIV—United
States, 2016.
FIG. 146.3 Cervical ectopy. (From Seattle STD/HIV Prevention Training Center,
University of Washington, Claire E. Stevens.)
Screening
Early detection and treatment are primary STI control strategies. Some of the
most common STIs in adolescents, including HPV, HSV, chlamydia, and
gonorrhea, are usually asymptomatic and if undetected can be spread
inadvertently by the infected host. Screening initiatives for chlamydial
infections have demonstrated reductions in PID cases by up to 40%. Although
federal and professional medical organizations recommend annual chlamydia
screening for sexually active females <25 yr old, according to the National
Center for Quality Assurance, in 2015 among sexually active 16-20 yr old
females, approximately 42% of commercial health maintenance organization
(HMO) members and 52% Medicaid HMO members were tested for chlamydia
during the previous year. The lack of a dialog about STIs or the provision of STI
services at annual preventive service visits to sexually experienced adolescents
are missed opportunities for screening and education. Comprehensive,
confidential, reproductive health services, including STI screening, should be
offered to all sexually experienced adolescents (Table 146.2 ).
Table 146.2
Routine Laboratory Screening
Recommendations for Sexually Transmitted
Infections in Sexually Active Adolescents and
Young Adults
Chlamydia Trachomatis and Neisseria Gonorrhoeae
Syphilis
• Screening adolescents for HCV who report risk factors, i.e., injection drug
use, receipt of an unregulated tattoo, received blood products or organ
donation before 1992, received clotting factor concentrates before 1987,
long-term hemodialysis.
• Given the high HCV prevalence among young injection drug users,
screening should be strongly considered.
MSM, Men who have sex with men; STD, sexually transmitted disease.
Urethritis
Urethritis is an STI syndrome characterized by inflammation of the urethra,
usually caused by an infectious etiology. Urethritis may present with urethral
discharge, dysuria, urethral irritation, or meatal pruritus. Urgency, frequency of
urination, erythema of the urethral meatus, and urethral pain or burning are less
common clinical presentations. Approximately 30–50% of males are
asymptomatic but may have signs of discharge on diagnosis. On examination,
associated with fever and malaise. The diagnosis may require Epstein-Barr virus
titers, or polymerase chain reaction (PCR) testing. Treatment is supportive care
including pain management.
Table 146.3
Signs, Symptoms, and Presumptive and Definitive
Diagnoses of Genital Ulcers
HERPES
SIGNS/SYMPTOMS SIMPLEX SYPHILIS (PRIMARY) CHANCROID
VIRUS
Ulcers Vesicles rupture Ulcer with well-demarcated indurated Unindurated and
to form shallow borders and a clean base (chancre) undermined borders and a
ulcers purulent base
Painful Painful Painless* Painful
Number of lesions Usually multiple Usually single Multiple
Inguinal First-time Usually mild and minimally tender Unilateral or bilateral
lymphadenopathy infections may painful adenopathy in
cause >50%
constitutional Inguinal bubo
symptoms and formation and rupture
lymphadenopathy. may occur.
Clinical suspicion Typical lesions; Early syphilis: typical chancre plus Exclusion of other causes
positive HSV-2 reactive nontreponemal test (RPR, of ulcers in the presence of
type-specific VDRL) and no history of syphilis, or 4- (a) typical ulcers and
serology test fold increase in quantitative lymphadenopathy, (b)
nontreponemal test in person with typical Gram stain, and (c)
history of syphilis; positive treponemal history of contact with
EIA with reactive nontreponemal test high-risk individual
(RPR, VDRL) and no prior history of (prostitute) or living in an
syphilis treatment endemic area
Definitive diagnosis Detection of HSV Identification of Treponema pallidum Detection of Haemophilus
by culture or PCR from a chancre or lymph node aspirate ducreyi by culture
from ulcer on dark-field microscopy
scraping or
aspiration of
vesicle fluid
* Primary syphilitic ulcers may be painful if they become co-infected with bacteria or 1 of the other
organisms responsible for genital ulcers.
DFA, Direct fluorescent antibody; EIA, enzyme immunoassay; HSV, herpes simplex virus; PCR,
polymerase chain reaction; RPR, rapid plasma reagin; VDRL, Venereal Disease Research
Laboratories.
Data from Centers for Disease Control and Prevention: Sexually transmitted diseases: treatment
guidelines, MMWR 64(RR-3), 2015. https://www.cdc.gov/std/tg2015/default.htm .
FIG. 146.8 Common normal and abnormal microscopic findings during
examination of vaginal fluid. KOH, Potassium hydroxide solution; PMN,
polymorphonuclear leukocyte; RBCs, red blood cells. (From Adolescent
medicine: state of the art reviews, vol 14, no 2, Philadelphia, 2003, Hanley
& Belfus, pp 350–351.)
Clinical laboratory–based vaginitis tests are also available. The Affirm VPIII
(Becton Dickenson, San Jose, CA) is a moderate-complexity nucleic acid probe
test that evaluates for T. vaginalis , G. vaginalis , and C. albicans and has a
sensitivity of 63% and specificity >99.9%, with results available in 45 min.
Some gonorrhea and chlamydia NAATs also offer an assay for T. vaginalis
testing of female specimens tested for N. gonorrhoeae and C. trachomatis ,
considered the gold standard for Trichomonas testing.
Objective signs of vulvar inflammation in the absence of vaginal pathogens,
along with a minimal amount of discharge, suggest the possibility of mechanical,
chemical, allergic, or other noninfectious irritation of the vulva (Table 146.4 ).
Table 146.4
Pathologic Vaginal Discharge
* If the cervical discharge appears normal and no WBCs are observed on the wet
prep of vaginal fluid, the diagnosis of PID is unlikely, and alternative causes of
pain should be investigated.
Table 146.6
Table 146.7
Table 146.8
Management Guidelines for Uncomplicated Genital Warts
and Genital Herpes in Adolescents and Adults
Intraanal WartsCryotherapy with liquid Management of intraanal warts should include consultation
nitrogen with a specialist.
or
Surgical removal
or
TCA or BCA 80-90% applied
to warts. A small amount
should be applied only to
warts and allowed to dry, at
which time a white “frosting”
develops. Can be repeated
weekly
HERPES SIMPLEX VIRUS (HSV; GENITAL HERPES)
First clinical Treat for 7-10 days with 1 Consider extending treatment if healing is incomplete after
episode of the following: 10 days of therapy
Acyclovir 400 mg orally 3
times daily
Acyclovir 200 mg orally 5
times daily
Valacyclovir 1 g orally twice
daily
Famciclovir 250 mg orally 3
times daily
Episodic therapy Treat with 1 of the Effective episodic treatment of recurrences requires
for recurrences following: initiation of therapy within 1 day of lesion onset or during
Acyclovir 400 mg orally 3 the prodrome that precedes some outbreaks. The patient
times daily for 5 days should be provided with a supply or a prescription for the
Acyclovir 800 mg orally medication with instructions to initiate treatment
twice daily for 5 days immediately when symptoms begin.
Acyclovir 800 mg orally 3
times daily for 2 days
Valacyclovir 500 mg orally
twice daily for 3 days
Valacyclovir 1,000 mg orally
once daily for 5 days
Famciclovir 125 mg orally
twice daily for 5 days
Famciclovir 1,000 mg orally
twice daily for 1 day
Famciclovir 500 mg orally
once, then 250 mg twice
daily for 2 days
Suppressive Treat with 1 of the All patients should be counseled regarding suppressive
therapy to following: therapy availability, regardless of number of outbreaks per
reduce frequency Acyclovir 400 mg orally year. Since the frequency of recurrent outbreaks diminishes
of recurrences twice daily over time in many patients, providers should periodically
Valacyclovir 500 mg orally discuss the need to continue therapy.
once daily* or 1 g orally once
daily
Famciclovir 250 mg orally
twice daily
* Valacyclovir 500 mg once daily might be less effective than other valacyclovir or acyclovir dosing
regimens in patients who have very frequent recurrences (i.e., ≥10 episodes per year).
Adapted from Centers for Disease Control and Prevention: Sexually transmitted diseases:
treatment guidelines, MMWR 64(RR-3), 2015. https://www.cdc.gov/std/tg2015/default.htm .
Table 147.1
Overview of Current Case Definitions for Systemic Exertion
Intolerance Disease (SEID) and Past Definitions of Chronic
Fatigue Syndrome or Myalgic Encephalomyelitis
The Institute of Medicine (IOM) 2015 recommendations apply to all ages and
include a special focus on pediatrics. The IOM suggested new diagnostic criteria
and a new name, systemic exertion intolerance disease (SEID) , to emphasize
the postexertion malaise criterion and better understand the illness (Table 147.2
). The most recent expert consensus report (June 2017) from the International
Writing Group for Pediatric ME/CFS provides a primer for diagnosis and
management.
Table 147.2
Criteria for Diagnosis of Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome
(ME/CFS)
Patient has each of the following 3 symptoms at least half the time, to
at least a moderately severe degree:
• A substantial reduction or impairment in the ability to engage in
preillness levels of occupational, educational, social, or
personal activities that persists for >6 mo and is accompanied
by fatigue, which is often profound, is of new or definite onset
(not lifelong), is not the result of ongoing excessive exertion,
and is not substantially alleviated by rest.
• Postexertional malaise*
• Unrefreshing sleep*
Plus at least 1 of the 2 following manifestations (chronic, severe):
• Cognitive impairment*
• Orthostatic intolerance
Epidemiology
Based on worldwide studies, 0.2–2.3% of adolescents or children have CFS.
Most epidemiology studies use the 1994 definition. CFS is more prevalent in
adolescents than in younger children. The variation in CFS prevalence estimates
may result from variations in case definition, study methodology and
application, study population composition (specialty vs general practice or
general population), and data collection (parent, self-reporting vs clinician
evaluation). Gender distribution in children differs from that in adults, with a
more equal distribution in children <15 yr old, while remaining 2-3–fold higher
in females 15-18 yr old. Few studies have reported the incidence of CFS among
children <10 yr old, leading to uncertainty in this group. In adolescents in The
Netherlands, the pediatrician-diagnosed incidence of CFS/ME was 0.01%, and in
CHAPTER 148
Evaluation of Suspected
Immunodeficiency
Kathleen E. Sullivan, Rebecca H. Buckley
Table 148.1
Predisposition to Specific Infections in Humans
AFFECTED
PATHOGEN PRESENTATION GENE/CHROMOSOMAL COMMENTS
REGION
BACTERIA
Streptococcus Invasive disease IRAK4, MyD88, C1QA, Also susceptible to other encapsulated
pneumoniae C1QB, C1QC, C4A+ C4B, bacteria
C2, C3
Neisseria Invasive disease C5, C6, C7, C8A, C8B, Recurrent disease common
C8G, C9, properdin
Burkholderia Invasive disease CYBB, CYBA, NCF1, NCF2 Also susceptible to staphylococcal and fungal
cepacia not pulmonary infections
colonization
Nocardia Invasive disease CYBB, CYBA, NCF1, NCF2 Also susceptible to staphylococcal and fungal
infections
Mycobacteria Usually IL12B, IL12RB1, IKBKG, Also susceptible to Salmonella typhi
nontuberculous IFNGR1, IFNGR2, STAT1 infections
mycobacteria (loss of function)
VIRUSES
Herpes Herpes simplex TRAF3, TRIF, TBK, Age of onset is typically outside the neonatal
simplex virus encephalitis UNC93B1, TLR3, STAT1 period.
Epstein-Barr Severe infectious SH2DIA, XIAP, ITK, CD27, Fulminant infectious mononucleosis,
virus mononucleosis, PRF1, STXBP2, UNC13D, malignant and nonmalignant
hemophagocytic LYST, RAB27A, STX11, lymphoproliferative disorders,
syndrome AP3B1 dysgammaglobulinemia, autoimmunity
Papillomavirus Warts RHOH, EVER1, EVER2, Warts are often progressive despite therapy.
CXCR4, DOCK8, GATA2,
STK4, SPINK5
Global Severe, progressive All types of severe Presentation depends on virus and infected
susceptibility viral infections combined immune organ
to viral deficiency, IFNAR2
infection
FUNGI
Candida Mucocutaneous AIRE, STAT1 (gain of AIRE deficiency is associated with
candida function), CARD9, STAT3, endocrinopathies, STAT1 (GOF) is associated
IL17F, IL17RC, IL17RA, with autoimmunity
ACT1
Dermatophytes Tissue invasion CARD9 Autosomal recessive
Aspergillus Deep infections CYBB, CYBA, NCF1, NCF2
Environmental Deep infections CYBB, CYBA, NCF1,
fungi NCF2, GATA2, STAT1 (gain
of function), CD40L
Table 148.2
Characteristic Clinical Patterns in Some Primary
Immunodeficiencies
FEATURES DIAGNOSIS
IN NEWBORNS AND YOUNG INFANTS (0-6 mo)
Hypocalcemia, unusual facies and ears, heart disease 22q11.2 deletion syndrome, DiGeorge
anomaly
Delayed umbilical cord detachment, leukocytosis, recurrent infections Leukocyte adhesion defect
Persistent thrush, failure to thrive, pneumonia, diarrhea Severe combined immunodeficiency
Bloody stools, draining ears, atopic eczema Wiskott-Aldrich syndrome
IN INFANTS AND YOUNG CHILDREN (6 mo to 5 yr)
Recurrent staphylococcal abscesses, staphylococcal pneumonia with Hyper-IgE syndrome, PGM3 deficiency
pneumatocele formation, coarse facial features, pruritic dermatitis
Persistent thrush, nail dystrophy, endocrinopathies Autoimmune polyendocrinopathy,
candidiasis, ectodermal dysplasia
Short stature, fine hair, severe varicella Cartilage hair hypoplasia with short-
limbed dwarfism
Oculocutaneous albinism, recurrent infection, hemophagocytic Chédiak-Higashi syndrome, Griscelli
syndrome syndrome, Hermansky-Pudlak syndrome
Table 148.3
Clinical Aids to the Diagnosis of
Immunodeficiency
Suggestive of B-Cell Defect (Humoral Immunodeficiency)
Systemic illness after vaccination with any live virus or bacille Calmette-
Guérin (BCG)
Unusual life-threatening complication after infection with benign viruses
(giant cell pneumonia with measles; varicella pneumonia)
Chronic oral candidiasis after age 6 mo
Chronic mucocutaneous candidiasis
Graft-versus-host disease after blood transfusion
Reduced lymphocyte counts for age
Low levels of immunoglobulins
Absence of lymph nodes and tonsils
Small thymus
Chronic diarrhea
Failure to thrive
Recurrent infections with opportunistic organisms
Suggestive of Asplenia
From Kliegman RM, Lye PS, Bordini BJ, ET AL, editors: Nelson pediatric
symptom-based diagnosis, Philadelphia, 2018, Elsevier, p 750.
Table 148.4
Characteristic Features of Primary Immunodeficiency
BCG, Bacille Calmette-Guérin; CMV, cytomegalovirus; EBV, Epstein-Barr virus; SLE, systemic
lupus erythematosus.
FIG. 148.1 Diagnostic testing algorithm for primary immunodeficiency diseases.
Common clinical scenarios are listed at the top. The 1st tier of testing is listed below
each category between the dark lines. The 2nd tier of testing is located below the 2nd
dark line. CBC, Complete blood count; DHR, dihydrorhodamine; MAI, Mycobacterium
avium-intracellulare infection.
Table 148.5
Special Physical Features Associated With
Immunodeficiency Disorders
CLINICAL FEATURES DISORDERS
DERMATOLOGIC
Eczema Wiskott-Aldrich syndrome, IPEX, hyper-IgE syndromes, hypereosinophilia
syndromes, IgA deficiency
Sparse and/or hypopigmented hair Cartilage-hair hypoplasia, Chédiak-Higashi syndrome, Griscelli syndrome
Ocular telangiectasia Ataxia-telangiectasia
Oculocutaneous albinism Chédiak-Higashi syndrome
Severe dermatitis Omenn syndrome
Erythroderma Omenn syndrome, SCID, graft-vs-host disease, Comel-Netherton syndrome
Recurrent abscesses with Hyper-IgE syndromes
pulmonary pneumatoceles
Recurrent organ granulomas or CGD
abscesses, lung, liver, and rectum
especially
Recurrent abscesses or cellulitis CGD, hyper-IgE syndrome, leukocyte adhesion defect
Cutaneous granulomas Ataxia telangiectasia, SCID, CVID, RAG deficiency
Oral ulcers CGD, SCID, congenital neutropenia
Periodontitis, gingivitis, stomatitis Neutrophil defects
Oral or nail candidiasis T-cell immune defects, combined defects (SCIDs); mucocutaneous
candidiasis; hyper-IgE syndromes; IL-12, -17, -23 deficiencies; CARD9
deficiency; STAT1 deficiency
Vitiligo B-cell defects, mucocutaneous candidiasis
Alopecia B-cell defects, mucocutaneous candidiasis
Chronic conjunctivitis B-cell defects
EXTREMITIES
Clubbing of nails Chronic lung disease caused by antibody defects
Arthritis Antibody defects, Wiskott-Aldrich syndrome, hyper-IgM syndrome
ENDOCRINOLOGIC
Hypoparathyroidism DiGeorge syndrome, mucocutaneous candidiasis
Endocrinopathies (autoimmune) Mucocutaneous candidiasis
Diabetes, hypothyroid IPEX and IPEX-like syndromes
Growth hormone deficiency X-linked agammaglobulinemia
Gonadal dysgenesis Mucocutaneous candidiasis
HEMATOLOGIC
Hemolytic anemia B- and T-cell immune defects, ALPS
Thrombocytopenia, small platelets Wiskott-Aldrich syndrome
Neutropenia Hyper-IgM syndrome, Wiskott-Aldrich variant, CGD
Immune thrombocytopenia B-cell immune defects, ALPS
SKELETAL
Short-limb dwarfism Short-limb dwarfism with T- and/or B-cell immune defects
Bony dysplasia ADA deficiency, cartilage-hair hypoplasia
ADA, Adenosine deaminase; ALPS, autoimmune lymphoproliferative syndrome; CGD, chronic
granulomatous disease; CVID, common variable immunodeficiency; IPEX, X-linked immune
dysfunction enteropathy polyendocrinopathy; SCID, severe combined immunodeficiency.
From Goldman L, Ausiello D: Cecil textbook of medicine, ed 22, Philadelphia, 2004, Saunders, p
1599.
Most immunologic defects can be excluded at minimal cost with the proper
choice of screening tests, which should be broadly informative, reliable, and
cost-effective (Table 148.6 and Figs. 148.2 and 148.3 ). A complete blood count
(CBC) with differential is the initial study if neutropenia is a consideration but is
less recognized as a screening test for T-cell defects. Lymphopenia is seen the
majority of T-cell defects. If an infant's neutrophil count is persistently elevated
in the absence of any signs of infection, a leukocyte adhesion deficiency should
be suspected. Normal lymphocyte counts are higher in infancy and early
childhood than later in life (Fig. 148.4 ). Knowledge of normal values for
absolute lymphocyte counts at various ages in infancy and childhood is crucial in
the detection of T-cell defects. Additional clues from the CBC include absence
of Howell-Jolly bodies, which argues against congenital asplenia. Normal
platelet size or count excludes Wiskott-Aldrich syndrome. When
immunodeficiency is suspected, obtaining IgG, IgA, IgM, and IgE levels can be
a useful strategy, since antibody defects are the most common type of
immunodeficiency. Immunoglobulin levels must be interpreted within the
context of age-specific normative data.
Table 148.6
Initial Screening Immunologic Testing of the
Child With Recurrent Infections
Complete Blood Count, Differential, and Erythrocyte
Sedimentation Rate
Ab, Antibody; ADA, adenosine deaminase; C, complement; CH, hemolytic complement; G6PD,
glucose-6-phosphate dehydrogenase; HLA, human leukocyte antigen; Ig, immunoglobulin; MPO,
myeloperoxidase; NADPH, nicotinamide adenine dinucleotide phosphate; PNP, purine nucleoside
phosphorylase; TRECs, T-cell receptor rearrangement excision circle; WBC, white blood cell; φX,
phage antigen.
One useful test for B-cell function is to determine the presence and titer of
isohemagglutinins , or natural antibodies to type A and B red blood cell
polysaccharide antigens. This test measures predominantly IgM antibodies.
Isohemagglutinins may be absent normally in the first 2 yr of life and are always
absent if the patient is blood type AB.
Because most infants and children are immunized with diphtheria-tetanus-
pertussis (dTP), conjugated Haemophilus influenzae type b, and pneumococcal
conjugate vaccine, it is often informative to test for specific antibodies to
diphtheria, tetanus, H. influenzae polyribose phosphate, and pneumococcal
antigens. If the titers are low, measurement of antibodies to diphtheria or tetanus
toxoids before and 2-8 wk after a pediatric dTP or dT booster is helpful in
assessing the capacity to form IgG antibodies to protein antigens. To evaluate a
CD Classification of Some Lymphocyte Surface Molecules
CD
TISSUE/LINEAGE FUNCTION
NUMBER
CD1 Cortical thymocytes; Lipid antigen presentation to TCRγδ cells
Langerhans cells
CD2 T and NK cells Binds LFA-3 (CD58); alternative pathway of T-cell activation
CD3 T cells TCR associated; transduces signals from TCR
CD4 T-helper cell subset Receptor for HLA class II antigens; associated with p56 Ick tyrosine
kinase
CD7 T and NK cells and their Mitogenic for T lymphocytes
precursors
CD8 Cytotoxic T-cell subset; Receptor for HLA class I antigens; associated with p56 Ick tyrosine
also on 30% of NK cells kinase
CD10 B-cell progenitors Peptide cleavage
CD11a T, B, and NK cells With CD18, ligand for ICAMs 1, 2, and 3
CD11b, c NK cells With CD18, receptors for C3bi
CD16 NK cells FcR for IgG
CD19 B cells Regulates B-cell activation
CD20 B cells Mediates B-cell activation
CD21 B cells C3d, also the receptor for EBV; CR2
CD25 T, B, and NK cells Mediates signaling by IL-2
CD34 Stem cells Binds to L -selectin
CD38 T, B, and NK cells and Associates with hyaluronic acid
monocytes
CD40 B cells and monocytes Initiates isotype switching in B cells when ligated
CD44 Bone marrow stromal and Matrix adhesion molecule
many other cells
CD45 All leukocytes Tyrosine phosphatase that regulates lymphocyte activation; CD45R0
isoform on memory T cells, CD45RA isoform on naïve T cells
CD56 NK cells Mediates NK homotypic adhesion
CD62L Marker for recent Cell adhesion molecule
thymic emigrants
Also found on other
leukocytes
CD69 T cells and NK cells Early activation marker
CD73 T and B cells Associates with AMP
CD80 B cells Co-stimulatory with CD28 on T cells to upregulate high-affinity IL-2
receptor
CD86 B cells Co-stimulatory with CD28 on T cells to upregulate high-affinity IL-2
receptor
CD117 Pro-B cells, double- Receptor for stem cell factor
negative thymocytes
CD127 T cells Mediates IL-7 signaling
CD132 T, B, and NK cells Mediates signaling by IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21
CD154 Activated CD4+ T cells Ligates CD40 on B cells and initiates isotype switching
CD278 T cells Interacts with B7-H2
AMP, Adenosine monophosphate; EBV, Epstein-Barr virus; ICAMs, intracellular adhesion
molecules; IL, interleukin; LFA, leukocyte function–activating antigen; NK, natural killer; TCR, T-
cell receptor.
Table 149.2
Common Cytokines
Table 150.1
X-Linked Agammaglobulinemia
Patients with X-linked agammaglobulinemia (XLA ), or Bruton
agammaglobulinemia , have a profound defect in B-lymphocyte development
resulting in severe hypogammaglobulinemia, an absence of circulating B cells,
small to absent tonsils, and no palpable lymph nodes.
Table 150.3
Table 150.4
Features of SAP (SH2D1A) and XIAP Deficiency
Clinical Manifestations
Affected males are usually healthy until they acquire EBV infection. The mean
age of presentation is <5 yr. There are 3 major clinical phenotypes: (1)
fulminant, often fatal, infectious mononucleosis (50% of cases); (2) lymphomas,
predominantly involving B-lineage cells (25%); and (3) acquired
hypogammaglobulinemia (25%). A less common manifestation is CNS
vasculitis. There is a marked impairment in production of antibodies to the EBV
transduction from the TCR to intracellular metabolic pathways (Fig. 151.1 ).
These patients have problems similar to those of other T-cell–deficient
individuals, and some with severe T-cell activation defects may clinically
resemble SCID patients (Table 151.1 ). In some cases, susceptibility to a single
pathogen or a limited number of pathogens dominates the clinical phenotype.
Susceptibility to Epstein-Barr virus, cytomegalovirus, and papillomavirus is
common in this set of T-cell defects. Most individuals with significant T-cell
activation defects will require a hematopoietic stem cell transplant. Although
each infection may be manageable early in life, the long-term prognosis is not
favorable in many of these conditions.
Table 151.1
Pathogenesis
SCID is caused by mutations in genes crucial for lymphoid cell development
(Table 152.1 and Fig. 152.1 ). All patients with SCID have very small thymuses
that contain no thymocytes and lack corticomedullary distinction or Hassall
corpuscles. The thymic epithelium appears histologically normal. Both the
follicular and the paracortical areas of the spleen are depleted of lymphocytes.
Lymph nodes, tonsils, adenoids, and Peyer patches are absent or extremely
underdeveloped.
Table 152.1
Genetic Basis of SCID and SCID Variants
ADDITIONAL
DISEASE INHERITANCE PRESUMED PATHOGENESIS TREATMENT
FEATURES
Reticular AR Impaired mitochondrial energy Severe neutropenia, GCSF, HSCT
dysgenesis metabolism and leukocyte deafness. Mutations in
differentiation adenylate kinase 2
Adenosine AR Accumulation of toxic purine Neurologic, hepatic, renal, HSCT, PEG-
deaminase nucleosides lung, and skeletal and bone ADA, gene
deficiency marrow abnormalities therapy
IL-2Rγ X-linked Abnormal signaling through by None HSCT
deficiency IL-2 receptor and other receptors
containing γc (IL-4, -7, -9, -15,
-21)
Jak3 AR Abnormal signaling downstream None HSCT
deficiency of γc
RAG1 and AR Defective V(D)J recombination None HSCT
RAG2
deficiency
Artemis AR Defective V(D)J recombination, DCLERE1C gene defects HSCT
deficiency radiation sensitivity
DNA-PK AR Defective V(D)J recombination None HSCT
deficiency
DNA ligase AR Defective V(D)J recombination, Growth delay, HSCT
IV radiation sensitivity microcephaly, bone
deficiency marrow abnormalities,
lymphoid malignancies
Cernunnos- AR Defective V(D)J recombination, Growth delay, HSCT
XLF radiation sensitivity microcephaly, birdlike
facies, bone defects
CD3δ AR Arrest of thymocytes Thymus size may be HSCT
deficiency differentiation at CD4− CD8− normal
stage
CD3ε AR Arrest of thymocytes γ/δ T cells absent HSCT
deficiency differentiation at CD4− CD8−
stage
CD3ζ AR Abnormal signaling None HSCT
deficiency
IL-7Rα AR Abnormal IL-7R signaling Thymus absent HSCT
deficiency
CD45 AR None HSCT
deficiency
Coronin-1A AR Abnormal T-cell egress from Normal thymus size. HSCT
deficiency thymus and lymph nodes Attention deficit disorder.
AR, Autosomal recessive; GCSF, granulocyte colony-stimulating factor; HSCT, hematopoietic
stem cell transplantation; IL, interleukin; Jak3, Janus kinase 3; PEG-ADA, polyethylene glycol-
modified adenosine deaminase; RAG1, RAG2, recombinase-activating genes 1 and 2; V(D)J,
variable, diversity, joining domains.
(Adapted from Roifman, CM. Grunebaum E: Primary T-cell immunodeficiencies. In Rich RR,
Fleisher TA, Shearer WT, et al, editors: Clinical immunology, ed 4. Philadelphia, 2013, Saunders,
pp 440–441).
FIG. 152.1 Relative frequencies of the different genetic types of severe combined
immunodeficiency (SCID). ADA, Adenosine deaminase; IL-7R, interleukin 7 receptor;
JAK, Janus kinase; RAG, recombinase-activating gene.
Wu U, Holland SM. Host susceptibility to non-tuberculous
mycobacterial infections. Lancet Infect Dis . 2015;15(8):968–
980.
152.4
Treatment of Cellular or Combined
Immunodeficiency
Kathleen E. Sullivan, Rebecca H. Buckley
Table 152.2
Infection in the Host Compromised by B- and T-Cell
Immunodeficiency Syndromes
APPROACH TO
IMMUNODEFICIENCY OPPORTUNISTIC ORGANISMS PREVENTION OF
TREATMENT OF
SYNDROME ISOLATED MOST FREQUENTLY INFECTIONS
INFECTIONS
B-cell immunodeficiencies Encapsulated bacteria (Streptococcus IVIG, 200-800 Maintenance
pneumoniae, Staphylococcus aureus, mg/kg IVIG for patients
Haemophilus influenzae , and Vigorous attempt with quantitative
Neisseria meningitidis ), to obtain and qualitative
Pseudomonas aeruginosa, specimens for defects in IgG
Campylobacter spp., enteroviruses, culture before metabolism (400-
rotaviruses, Giardia lamblia, antimicrobial 800 mg/kg every
Cryptosporidium spp., Pneumocystis therapy 3-5 wk)
jiroveci, Ureaplasma urealyticum , Incision and In chronic
and Mycoplasma pneumoniae drainage if recurrent
abscess present respiratory
Antibiotic disease, vigorous
selection on the attention to
basis of postural drainage
sensitivity data In selected cases
(recurrent or
chronic
pulmonary or
middle ear),
prophylactic
administration of
ampicillin,
penicillin, or
trimethoprim-
sulfamethoxazole
T-cell immunodeficiencies Encapsulated bacteria (S. Vigorous attempt Prophylactic
pneumoniae, H. influenzae, S. aureus to obtain administration of
), facultative intracellular bacteria specimens for trimethoprim-
(Mycobacterium tuberculosis , other culture before sulfamethoxazole
Mycobacterium spp., and Listeria antimicrobial for prevention of
monocytogenes ); Escherichia coli; P. therapy P. jiroveci
aeruginosa; Enterobacter spp.; Incision and pneumonia
Klebsiella spp.; Serratia marcescens; drainage if Oral
Salmonella spp.; Nocardia spp.; abscess present nonadsorbable
viruses (cytomegalovirus, herpes Antibiotic antimicrobial
simplex virus, varicella-zoster virus, selection on the agents to lower
Epstein-Barr virus, rotaviruses, basis of concentration of
adenoviruses, enteroviruses, sensitivity data gut flora
respiratory syncytial virus, measles Early antiviral No live virus
virus, vaccinia virus, and treatment for vaccines or
parainfluenza viruses); protozoa herpes simplex, bacille Calmette-
(Toxoplasma gondii and cytomegalovirus, Guérin vaccine
Cryptosporidium spp.); and fungi and varicella- Careful
(Candida spp., Cryptococcus zoster viral tuberculosis
neoformans, Histoplasma capsulatum infections screening
, and P. jiroveci ) Topical and
nonadsorbable
antimicrobial
agents frequently
are useful
IVIG, Intravenous immune globulin.
From Stiehm ER, Ochs HD, Winkelstein JA: Immunologic disorders in infants and children , ed 5,
Philadelphia, 2004, Saunders.
Autoimmune Lymphoproliferative
Syndrome
Autoimmune lymphoproliferative syndrome (ALPS ), also known as Canale-
Smith syndrome, is a disorder of abnormal lymphocyte apoptosis leading to
polyclonal populations of T cells (double-negative T cells), which express CD3
and α/β antigen receptors but do not have CD4 or CD8 co-receptors (CD3+ T-
cell receptor α/β+ , CD4− CD8− ). These T cells respond poorly to antigens or
mitogens and do not produce growth or survival factors (IL-2). The genetic
deficit in most patients is a germline or somatic mutation in the FAS gene, which
produces a cell surface receptor of the TNF receptor superfamily (TNFRSF6),
which, when stimulated by its ligand, will produce programmed cell death (Table
152.3 ). Persistent survival of these lymphocytes leads to immune dysregulation
and autoimmunity. ALPS is also caused by other genes in the Fas pathway
(FASLG and CASP10 ). In addition, ALPS-like disorders are associated with
other mutations: RAS-associated autoimmune lymphoproliferative disorder
(RALD), caspase-8 deficiency, Fas-associated protein with death domain
deficiency (FADD), and protein kinase C delta deficiency (PRKCD). These
disorders have varying degrees of immunodeficiency, autoimmunity, and
lymphoproliferation.
Table 152-3
Revised Diagnostic Criteria for Autoimmune
Lymphoproliferative Syndrome*
REQUIRED
1. Chronic (>6 months), nonmalignant, noninfectious lymphadenopathy, splenomegaly or both
2. Elevated CD3+TCRαβ+CD4-CD8- DNT cells (≥1.5% of total lymphocytes or 2.5% of CD3+ lymphocytes) in
the setting of normal or elevated lymphocyte counts
ACCESSORY
Primary
1. Defective lymphocyte apoptosis (in 2 separate assays)
2. Somatic or Germline pathogenic mutation in FAS , FASLG , or CASP10
Secondary
1. Elevated plasma sFasL levels (>200 pg/mL) OR elevated plasma interleukin-10 levels (>20 pg/mL) OR
elevated serum or plasma vitamin B 12 levels (>1500 ng/L) OR elevated plasma interleukin-18 levels >500
pg/mL
2. Typical immunohistological findings as reviewed by an experienced hematopathologist
3. Autoimmune cytopenias (hemolytic anemia, thrombocytopenia, or neutropenia) AND elevated
immunoglobulin G levels (polyclonal hypergammaglobulinemia)
4. Family history of a nonmalignant/noninfectious lymphoproliferation with or without autoimmunity
*
A definitive diagnosis is based on the presence of both required criteria plus one primary
accessory criterion. A probable diagnosis is based on the presence of both required criteria plus
one secondary accessory criterion.
From Petty RE, Laxer RM, Lindsley CB, Wedderburn LR, editors: Textbook of pediatric
rheumatology, ed 7, Philadelphia, 2016, Elsevier, Box 46-2.
Clinical Manifestations
ALPS is characterized by autoimmunity, chronic persistent or recurrent
lymphadenopathy , splenomegaly, hepatomegaly (in 50%), and
hypergammaglobulinemia (IgG, IgA). Many patients present in the 1st yr of life,
and most are symptomatic by age 5 yr. Lymphadenopathy can be striking (Fig.
152.4 ). Splenomegaly may produce hypersplenism. Autoimmunity also
produces anemia (Coombs-positive hemolytic anemia) or thrombocytopenia or a
mild neutropenia. The lymphoproliferative process (lymphadenopathy,
splenomegaly) may regress over time, but autoimmunity does not regress and is
characterized by frequent exacerbations and recurrences. Other autoimmune
features include urticaria, uveitis, glomerulonephritis, hepatitis, vasculitis,
panniculitis, arthritis, and CNS involvement (seizures, headaches,
encephalopathy).
Treatment
Rapamycin (sirolimus) will often control the adenopathy and autoimmune
cytopenias. Malignancies can be treated with the usual protocols used in patients
unaffected by ALPS. Stem cell transplantation is another possible option in
treating the autoimmune manifestations of ALPS.
Immune Dysregulation,
Polyendocrinopathy, Enteropathy, X-
Linked Syndrome
This immune dysregulation syndrome is characterized by onset within the 1st
few wk or mo of life with watery diarrhea (autoimmune enteropathy), an
eczematous rash (erythroderma in neonates), insulin-dependent diabetes
mellitus, hyperthyroidism or more often hypothyroidism, severe allergies, and
other autoimmune disorders (Coombs-positive hemolytic anemia,
thrombocytopenia, neutropenia). Psoriasiform or ichthyosiform rashes and
alopecia have also been reported.
Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX )
syndrome is caused by a mutation in the FOXP3 gene, which encodes a
forkhead-winged helix transcription factor (scurfin) involved in the function and
development of CD4+ CD25+ regulatory T cells (Tregs). The absence of Tregs
may predispose to abnormal activation of effector T cells. Dominant gain-of-
function mutations in STAT1 and other gene mutations (Table 152.4 ) produce an
IPEX-like syndrome, also associated with compromised Tregs.
Table 152.4
Clinical and Laboratory Features of IPEX and IPEX-Like
Disorders
Clinical Manifestations
Watery diarrhea with intestinal villous atrophy leads to failure to thrive in most
patients. Cutaneous lesions (usually eczema) and insulin-dependent diabetes
begin in infancy. Lymphadenopathy and splenomegaly are also present. Serious
bacterial infections (meningitis, sepsis, pneumonia, osteomyelitis) may be
related to neutropenia, malnutrition, or immune dysregulation. Laboratory
features reflect the associated autoimmune diseases, dehydration, and
malnutrition. In addition, serum IgE levels are elevated, with normal levels of
IgM, IgG, and IgA. The diagnosis is made clinically and by mutational analysis
of the FOXP3 gene.
Treatment
Inhibition of T-cell activation by cyclosporine, tacrolimus, or sirolimus with
corticosteroids is the treatment of choice, along with the specific care of the
endocrinopathy and other manifestations of autoimmunity. These agents are
transduction by type 1 and type 2 cytokine receptors within most hematopoietic
cells. Cytokines bind to their cognate receptor, triggering JAK-STAT pathways,
ultimately leading to the upregulation of genes involved in the immune response
against many pathogens. There are 4 JAK proteins (Jak1, Jak2, Jak3, Tyk2) and
6 STATs (1-6). Mutations in several JAKs and STATs cause immunodeficiency.
Table 152.5 includes diseases affecting STAT proteins characterized by immune
dysregulation. Chronic immunosuppression is necessary for control of STAT
defects. Ruxolitinib, a JAK-STAT inhibitor, has been used with some success.
With the advent of JAK-STAT immunomodulating therapies, more treatment
options will be available to patients.
Table 152.5
Defects of STAT Proteins Associated With
Immunodysregulation
AUTOIMMUNE OR
OTHER
PROTEIN LOF/GOF INFLAMMATORY IMMUNOPHENOTYPE
CHARACTERISTICS
COMPLICATIONS
STAT1 GOF IPEX-like enteropathy, Infections Variable lymphopenia,
enteropathy, endocrinopathy, CMC hypogammaglobulinemia,
dermatitis, cytopenias Viral infections abnormal T-cell function,
NTM reduced Th17 expression
Dimorphic mold
Respiratory
bacterial
STAT3 GOF Early onset enteropathy, Respiratory tract Increased DNT (CD3+
severe growth failure, infections CD4− CD8− )
lymphoproliferation, Herpes viral Hypogammaglobulinemia
autoimmune cytopenias, infections T-cell lymphopenia
inflammatory lung disease, T-cell LGL B-cell lymphopenia
type 1 diabetes, dermatitis, leukemia
arthritis NTM
STAT5B LOF Severe growth hormone Respiratory tract Lymphopenia
resistant growth failure, infections Reduced Treg cells
lymphocytic interstitial Viral infections Reduced γδ T cells
pneumonitis, atopic dermatitis Reduced NK cells
STAT, Signal transducer and activator of transcription; GOF, gain of function; LOF, loss of function;
IPEX, immunodysregulation, polyendocrinopathy, enteropathy, X-linked; CMC, chronic
mucocutaneous candidiasis; NTM, nontuberculous mycobacteria; DNT, double-negative T cell.
Table 152.6
Defects of Nuclear Factor-κB Pathways Associated With
Immune Dysregulation
AUTOIMMUNE OR
OTHER IMMUNOLOGIC
PROTEIN INHERITANCE INFLAMMATORY
MANIFESTATIONS PHENOTYPE
COMPLICATIONS
IKBKG XL Colitis Ectodermal Hypogammaglobulinemia
(NEMO) dysplasia Hyper IgM
Osteopetrosis Hyper IgA
Lymphedema Hyper IgD
Bacterial Poor antibody responses
infections Decreased NK cell
Opportunistic function
infections Decreased TLR responses
DNA viral
infections
NF-κB1 AD Pyoderma Atrophic gastritis Hypogammaglobulinemia
gangrenosum Squamous cell IgA deficiency
Lymphoproliferation carcinoma
Cytopenia Respiratory tract
Hypothyroidism infections
Alopecia areata Superficial skin
Enteritis infections
LIP Lung
NRH adenocarcinoma
Respiratory
insufficiency
Aortic stenosis
Non-Hodgkin
lymphoma
NF-κB2 AD Alopecia totalis Viral respiratory Early-onset
Trachyonychia infections hypogammaglobulinemia
Vitiligo Pneumonias Low vaccine responses
Autoantibodies: Sinusitis Variable B-cell counts
thyroid peroxidase, Otitis media Low switched memory B
glutamate Recurrent herpes cells (CD19+ CD27+ IgD
decarboxylase, Asthma − )
thyroglobulin Type 1 Chiari Low marginal zone B
Central adrenal malformation
cells (CD19+ CD27+
insufficiency Interstitial lung IgD+ )
disease
XL, X-linked; AD, autosomal dominant; LIP, lymphocytic interstitial pneumonitis; NRH,
nonregenerative hyperplasia.
Neutrophils
Thomas D. Coates
Table 153.1
Neutrophil and Monocyte Kinetics
NEUTROPHILS
Average time in mitosis (myeloblast to myelocyte) 7-9 days
Average time in postmitosis and storage (metamyelocyte to neutrophil) 3-7 days
Average half-life in the circulation 6 hr
Average total body pool 6.5 × 108 cells/kg
Average circulating pool 3.2 × 108 cells/kg
Average marginating pool 3.3 × 108 cells/kg
Average daily turnover rate 1.8 × 108 cells/kg
MONONUCLEAR PHAGOCYTES
Average time in mitosis 30-48 hr
Average half-life in the circulation 36-104 hr
Average circulating pool (monocytes) 1.8 × 107 cells/kg
Average daily turnover rate 1.8 × 109 cells/kg
Average survival in tissues (macrophages) Months
From Boxer LA: Function of neutrophils and mononuclear phagocytes. In Bennett JC, Plum F,
editors: Cecil textbook of internal medicine, ed 20, Philadelphia, 1996, Saunders.
Hematopoiesis
The hematopoietic progenitor system can be viewed as a continuum of
functional compartments, with the most primitive compartment composed of
very rare pluripotential stem cells , which have high self-renewal capacity and
give rise to more mature stem cells, including cells that are committed to either
lymphoid or myeloid development (Fig. 153.1 ). Common lymphoid progenitor
cells give rise to T- and B-cell precursors and their mature progeny (see Chapter
149 ). Common myeloid progenitor cells eventually give rise to committed
single-lineage progenitors of the recognizable precursors through a random
process of lineage restriction in a stepwise process (see Chapter 473 ). The
capacity of lineage-specific committed progenitors to proliferate and
differentiate in response to demand provides the hematopoietic system with a
remarkable range of response to changing requirements for mature blood cell
production.
CHAPTER 154
Development
Monocytes develop more rapidly during bone marrow hematopoiesis and
remain longer in the circulation than do neutrophils (see Table 153.1 ). The
monoblast is the first recognizable monocyte precursor, followed by the
promonocyte , with cytoplasmic granules and an indented nucleus, and finally
the fully developed monocyte with cytoplasmic granules filled with hydrolytic
enzymes. The transition from monoblast to mature circulating monocyte requires
about 6 days.
Three major subsets of human monocytes can be identified on the basis of
surface antigens: CD14++ CD16− classical monocytes that constitute the majority
of total monocytes in the resting state; the more mature CD14++ CD16+
proinflammatory (intermediate ) monocytes, which produce proinflammatory
hormone-like factors termed cytokines , such as tumor necrosis factor-α (TNF-
α), in response to microbial stimuli; and nonclassical (regulatory) monocytes
(CD14+ CD16++ ) that promote wound healing. Monocytes from these subsets
migrate into tissues in response to localized inflammation or injury and provide
proinflammatory host defense or antiinflammatory responses and wound
healing.
Tissue (organ)-specific macrophages arise from macrophage progenitors
that develop in the yolk sac and fetal liver before hematopoiesis occurs in the
bone marrow. These cells maintain their population through self-renewal. Tissue
macrophages can also be populated to some extent by circulating monocytes.
Monocytes or macrophages at sites of active inflammation mature into
proinflammatory (M1) macrophages or proresolving (M2) macrophages. In
ongoing tissue injury or inflammation, many (perhaps most) of the macrophages
will express a mix of the properties of the classic types.
Whether embryonic or blood derived, tissue macrophages are directed by
organ-specific factors to differentiate into macrophages characteristic of that
organ. Embryonic progenitors or monocytes in the liver become Kupffer cells
that bridge the sinusoids separating adjacent plates of hepatocytes. Those at the
Upregulated Functions in Macrophages
Activated in Response to Infection
Microbicidal and tumoricidal activity
Phagocytosis (of most particles) and pinocytosis
Phagocytosis-associated respiratory burst (O2 − , H2 O2 )
Generation of nitric oxide
Chemotaxis
Glucose transport and metabolism
Membrane expression of MHC, CD40, TNF receptor
Antigen presentation
Secretion:
Complement components
Lysozyme, acid hydrolases, and cytolytic proteinases
Collagenase
Plasminogen activator
Interleukins, including IL-1, IL-12, and IL-15
TNF-α
Interferons, including IFN-α and IFN-β
Antimicrobial peptides (cathelicidin, defensins)
Angiogenic factors
Allergic rhinitis
Asthma
Acute and chronic urticaria
Eczema
Angioedema
Hypersensitivity drug reactions (drug rash with eosinophilia and systemic
symptoms [DRESS])
Eosinophilic gastrointestinal disorders
Interstitial nephritis
Infectious Diseases
Tissue-Invasive Helminth Infections
Trichinosis
Toxocariasis
Strongyloidosis
Ascariasis
Filariasis
Schistosomiasis
Echinococcosis
Amebiasis
Malaria
Scabies
Toxoplasmosis
Other Infections
Pneumocystis jirovecii
Scarlet fever
Allergic bronchopulmonary aspergillosis (ABPA)
Coccidioidomycosis
Human immunodeficiency virus (HIV)
Malignant Disorders
Gastrointestinal Disorders
Rheumatologic Disease
Rheumatoid arthritis
Eosinophilic fasciitis
Scleroderma
Dermatomyositis
Systemic lupus erythematosus
IgG4-related disease
Eosinophilic granulomatosis with polyangiitis (Churg-Strauss vasculitis)
Miscellaneous
Allergic Diseases
Allergy is the most common cause of eosinophilia in children in the United
States. Patients with allergic asthma typically have eosinophils in the blood,
sputum, and/or lung tissue. Hypersensitivity drug reactions can elicit
eosinophilia, and when associated with organ dysfunction (e.g., DRESS [drug
rash with eosinophilia and systemic symptoms]), these reactions can be serious
(see Chapter 177 ). If a drug is suspected of triggering eosinophilia, biochemical
evidence of organ dysfunction should be sought, and if found, the drug should be
discontinued. Various skin diseases have also been associated with eosinophilia,
including atopic dermatitis/eczema, pemphigus, urticaria, and toxic epidermal
necrolysis.
Eosinophilic gastrointestinal diseases are important emerging allergic causes
of eosinophilia in tissue and, in some cases, peripheral blood (see Chapter 363 ).
In these conditions, eosinophils are recruited to esophagus, stomach, and/or
intestine, where they cause tissue inflammation and clinical symptoms such as
dysphagia, food aversion, abdominal pain, vomiting, and diarrhea. Treatment
CHAPTER 156
Neutrophils are the first line of defense against microbial invasion. They arrive
at the site of inflammation during the critical 2-4 hr after microbial invasion to
contain the infection and prevent hematogenous dissemination. This well-
orchestrated process is one of the most interesting stories in modern cell biology.
In fact, much of our knowledge about neutrophil function derives from studies
done in patients with genetic errors in neutrophil function. These critical
functions and their associated disorders are depicted in Fig. 153.2 . Children
with phagocytic dysfunction present at a young age with recurrent infections that
often involve unusual organisms and are poorly responsive to treatment.
Primary defects of phagocytic function comprise <20% of
immunodeficiencies, and there is significant overlap in the presenting signs and
symptoms between phagocytic disorders and lymphocyte and humeral disorders.
Children with phagocytic defects present with deep tissue infection, pneumonia,
adenitis, or osteomyelitis rather than bloodstream infections (Tables 156.1 and
156.2 and Fig. 156.1 ). A few clinical features point to phagocyte defects rather
than other immunodeficiencies, but correct diagnosis relies on highly specialized
laboratory tests.
Table 156.1
Infections and White Blood Cell Defects: Features That Can
Be Seen in Phagocyte Disorders
UNUSUALLY
SEVERE INFECTIONS RECURRENT INFECTIONS SPECIFIC INFECTIONS LOCATED
INFECTIONS
Diagnosis
Type of Diagnosis to Site of Diagnosis to Diagnosis to Site of
Microorganism to
Infection Consider Infection Consider Consider Infection
Consider
Cellulitis Neutropenia, Cutaneous Neutropenia, Staphylococcus Neutropenia, Umbilical LAD
LAD, CGD, CGD, LAD, epidermidis LAD cord
HIES HIES
Colitis Neutropenia, Gums LAD, Serratia CGD Liver CGD
CGD neutrophil marcescens, abscess
motility Nocardia,
disorders Burkholderia
cepacia
Osteomyelitis CGD, Upper and Neutropenia, Aspergillus Neutropenia, Gums LAD,
MSMD lower HIES, CGD, HIES neutrophil
pathway respiratory tract functional motility
defects neutrophil disorders
disorders
Gastrointestinal CGD, MSMD Nontuberculous MSMD
tract pathway mycobacteria, pathway
defects BCG defects,
(salmonella) SCID, CGD
Lymph nodes CGD, MSMD Candida Neutropenia,
pathway CGD, MPO
defects
(mycobacteria)
Osteomyelitis CGD, MSMD
BCG, Bacille Calmette-Guérin; CGD, chronic granulomatous disease; HIES, hyper-IgE syndrome;
LAD, leukocyte adhesion deficiency; MSMD, mendelian susceptibility to mycobacterial disease;
SCID, severe combined immunodeficiency.
From Leung DYM: Pediatric allergy principles and practice , ed 2, Philadelphia, 2010, Saunders, p
134.
Table 156.2
Clinical Disorders of Neutrophil Function
IMPAIRED CLINICAL
DISORDER ETIOLOGY
FUNCTION CONSEQUENCE
DEGRANULATION ABNORMALITIES
Chédiak-Higashi Autosomal recessive; disordered Decreased neutrophil Neutropenia; recurrent
syndrome (CHS) coalescence of lysosomal granules; chemotaxis, pyogenic infections;
responsible gene is CHSI/LYST , which degranulation, and propensity to develop
encodes a protein hypothesized to regulate bactericidal activity; marked
granule fusion platelet storage pool hepatosplenomegaly as a
defect; impaired NK manifestation of
function, failure to hemophagocytic
disperse melanosomes syndrome
Specific granule Autosomal recessive; functional loss of Impaired chemotaxis Recurrent deep-seated
deficiency myeloid transcription factor arising from a and bactericidal abscesses
mutation or arising from reduced activity; bilobed
expression of Gfi-1 or C/EBPε , which nuclei in neutrophils;
regulates specific granule formation defensins, gelatinase,
collagenase, vitamin
B12 –binding protein,
and lactoferrin
ADHESION ABNORMALITIES
Leukocyte Autosomal recessive; absence of Decreased binding of Neutrophilia; recurrent
adhesion CD11/CD18 surface adhesive iC3b to neutrophils bacterial infection
deficiency 1 glycoproteins (β2 -integrins) on leukocyte and impaired adhesion associated with a lack of
(LAD-1) membranes most commonly arising from to ICAM-1 and pus formation
failure to express CD18 messenger RNA ICAM-2
Leukocyte Autosomal recessive; loss of fucosylation Decreased adhesion to Neutrophilia; recurrent
adhesion of ligands for selectins and other glycol activated endothelium bacterial infection
deficiency 2 conjugates arising from mutations of expressing ELAM without pus
(LAD-2) GDP-fucose transporter
Leukocyte Autosomal recessive; impaired integrin Impaired neutrophil Neutrophilia, recurrent
adhesion function arising from mutations of adhesion and platelet infections, bleeding
deficiency 3 FERMT3 , which encodes kindlin-3 in activation tendency
(LAD-1 variant hematopoietic cells; kindlin-3 binds to β-
syndrome) integrin and thereby transmits integrin
activation
DISORDERS OF CELL MOTILITY
Enhanced motile Autosomal recessive gene responsible for Excessive Recurrent fever,
responses; FMF FMF on chromosome 16, which encodes accumulation of peritonitis, pleuritis,
for a protein called pyrin; pyrin regulates neutrophils at arthritis, amyloidosis
caspase-1 and thereby IL-1β secretion; inflamed sites,
mutated pyrin may lead to heightened possibly the result of
sensitivity to endotoxin, excessive IL-1β excessive IL-1β
production, and impaired monocyte production
apoptosis
DEPRESSED MOTILE RESPONSES
Defects in the IgG deficiencies; C3 and properdin Deficiency of serum Recurrent pyogenic
generation of deficiency can arise from genetic or chemotaxis and infections
chemotactic acquired abnormalities; mannose-binding opsonic activities
signals protein deficiency predominantly in
neonates
Intrinsic defects In the neonatal neutrophil there is Diminished Propensity to develop
of the neutrophil, diminished ability to express β2 -integrins, chemotaxis pyogenic infections
e.g., LAD, CHS, and there is a qualitative impairment in β2
specific granule -integrin function
deficiency,
neutrophil actin
dysfunction,
neonatal
neutrophils
Direct inhibition Ethanol, glucocorticoids, cyclic AMP Impaired locomotion Possible cause for
of neutrophil and ingestion; frequent infections;
mobility, e.g., impaired adherence neutrophilia seen with
drugs epinephrine arises from
cyclic AMP release from
endothelium
Immune Bind to Fc receptors on neutrophils in Impaired chemotaxis Recurrent pyogenic
complexes patients with rheumatoid arthritis, infections
systemic lupus erythematosus, and other
inflammatory states
Hyper-IgE Autosomal dominant; responsible gene is Impaired chemotaxis Recurrent skin and
syndrome STAT3 at times; impaired sinopulmonary infections,
regulation of cytokine eczema, mucocutaneous
production candidiasis, eosinophilia,
retained primary teeth,
minimal trauma fractures,
scoliosis, and
characteristic facies
Hyper-IgE Autosomal recessive; more than 1 gene High IgE levels, Recurrent pneumonia
syndrome–AR likely contributes to its etiology impaired lymphocyte without pneumatoceles
activation to sepsis, enzyme, boils,
staphylococcal mucocutaneous
antigens candidiasis, neurologic
symptoms, eosinophilia
MICROBICIDAL ACTIVITY
Chronic X-linked and autosomal recessive; Failure to activate Recurrent pyogenic
granulomatous failure to express functional gp91 phox neutrophil respiratory infections with catalase-
disease (CGD) in the phagocyte membrane in p22 burst, leading to positive microorganisms
phox (AR) failure to kill catalase-
Other AR forms of CGD arise from positive microbes
failure to express protein p47 phox or
p67 phox
G6PD deficiency <5% of normal activity of G6PD Failure to activate Infections with catalase-
NADPH-dependent positive microorganisms
oxidase; hemolytic
anemia
Myeloperoxidase Autosomal recessive; failure to process H2 O2 -dependent None
deficiency modified precursor protein arising from antimicrobial activity
missense mutation not potentiated by
myeloperoxidase
Rac2 deficiency Autosomal dominant; dominant negative Failure of membrane Neutrophilia, recurrent
inhibition by mutant protein of Rac2- receptor–mediated O2 bacterial infections
mediated functions − generation and
chemotaxis
Deficiencies of AR; failure to detoxify H2 O2 Excessive formation Minimal problems with
glutathione of H2 O2 recurrent pyogenic
reductase and infections
glutathione
synthetase
AMP, Adenosine monophosphate; AR, autosomal recessive; C, complement; CD, cluster of
differentiation; ELAM, endothelial-leukocyte adhesion molecule; FMF, familial Mediterranean
fever; G6PD, glucose-6-phosphate dehydrogenase; GDP, guanosine diphosphate; ICAM,
intracellular adhesion molecule; IL-1, interleukin-1; NADPH, nicotinamide adenine dinucleotide
phosphate; NK, natural killer.
Adapted from Curnutte JT, Boxer LA: Clinically significant phagocytic cell defects. In Remington
JS, Swartz MN, editors: Current clinical topics in infectious disease , ed 6, New York, 1985,
McGraw-Hill, p 144.
except in the most experienced hands. The assays must be done on freshly
obtained blood and are affected by many factors related to blood sampling itself.
It is best to assay other features of the suspected disorder, such as surface marker
expression, to establish a specific diagnosis.
Table 156.3
Leukocyte Adhesion Deficiency Syndromes
LEUKOCYTE
TYPE 1 TYPE 2 (LAD-2 TYPE 3 (LAD- E-SELECTIN Rac2
ADHESION
(LAD-1) or CDG-IIc) 3) DEFICIENCY DEFICIENCY
DEFICIENCY (LAD)
OMIM 116920 266265 612840 131210 602049
Inheritance pattern Autosomal Autosomal Autosomal Unknown Autosomal
recessive recessive recessive dominant
Affected protein(s) β2 -Integrin Fucosylated Kindlin 3 Endothelial E- Rac2
common proteins (e.g., selectin
chain sialyl-Lewisx , expression
(CD18) CD15s)
Neutrophil function Chemotaxis, Rolling, tethering Chemotaxis, Rolling, Chemotaxis,
affected tight adhesion, tethering superoxide
adherence superoxide production
production
Delayed umbilical cord Yes (severe Yes Yes Yes Yes
separation phenotype
only)
Leukocytosis/neutrophilia Yes Yes Yes No (mild Yes
neutropenia)
CDG-IIc, Congenital disorder of glycosylation IIc, OMIM, Online Mendelian Inheritance in Man.
From Leung DYM: Pediatric allergy principles and practice , ed 2, Philadelphia, 2010, Saunders, p
139.
Treatment
There is no specific therapy for MPO deficiency. Aggressive treatment with
antifungal agents should be provided for candidal infections. The prognosis is
usually excellent.
Table 156.4
Classification of Chronic Granulomatous Disease
NBT
COMPONENT FLAVOCYTOCHROME INCIDENCE
INHERITANCE SUBTYPE* SCORE (%
AFFECTED b SPECTRUM (% of Cases)
Positive)
gp91 phox X X910 0 0 60
X91− Low 80-100 5
(weak)
X91− Low 5-10 <1
X91+ 0 0 1
p22 phox A A220 0 0 4
A22+ N 0 <1
p47 phox A A470 N 0 † 25
p67 phox A A670 N 0 5
A67+ N 0 <1
p40 phox A A40− N 100 <1
CHAPTER 157
Leukopenia
Thomas F. Michniacki, Kelly J. Walkovich
Table 157.1
Diagnostic Approach for Patients With Leukopenia
Table 157.3
Table 157.4
Table 157.5
Forms of Drug-Induced Neutropenia
Acquired Neutropenia
Infection-Related Neutropenia
Transient neutropenia often accompanies or follows viral infections and is the
most frequent cause of neutropenia in childhood (Table 157.4 ). Viruses causing
mechanisms; select disorders are discussed next.
Table 157.6
Intrinsic Disorders of Myeloid Precursor Cells
Table 157.7
Causes of Lymphocytopenia
ACQUIRED
Infectious diseases AIDS, hepatitis, influenza, sepsis, tuberculosis, typhoid
Iatrogenic Corticosteroids, cytotoxic chemotherapy, high-dose PUVA, immunosuppressive
therapy, radiation, thoracic duct drainage
Systemic diseases Hodgkin disease, lupus erythematosus, myasthenia gravis, protein-losing enteropathy,
renal failure, sarcoidosis
Other Aplastic anemia, dietary deficiencies, thermal injury
INHERITED
Aplasia of lymphopoietic Cartilage-hair hypoplasia, ataxia-telangiectasia, SCID, thymoma, Wiskott-Aldrich
stem cells syndrome
PUVA, Psoralen and ultraviolet A irradiation; SCID, severe combined immunodeficiency.
Inherited Lymphopenia
Primary immunodeficiencies and bone marrow failure syndromes are the main
cause of inherited lymphopenia in children (see Table 157.7 ). Primary
immunodeficiency may result in a severe quantitative defect, as in XLA and
severe combined immunodeficiency (SCID), or a qualitative or progressive
defect, as in Wiskott-Aldrich syndrome and CVID. XLA is characterized by a
near-absence of mature B cells because of a mutation in BTK that results in a
dysfunctional tyrosine kinase. SCIDs are a genetically heterogeneous group of
disorders characterized by abnormalities of thymopoiesis and T-cell maturation.
Newborn screening for severe T-cell deficiency, by analysis of T-cell receptor
excision circles from dried blood spot Guthrie cards, aids in the rapid
identification and treatment of infants with SCID and other T-cell disorders.
Quantitative defects in lymphocytes can also be appreciated in select forms of
inherited bone marrow failure such as reticular dysgenesis, SCN secondary to
GFI1 mutation, and dyskeratosis congenita.
Neutrophilia
Neutrophilia is an increase in the total number of blood neutrophils that is 2 SD
above the mean count for age (see Chapter 748 ). Elevated absolute neutrophil
counts represent disturbances of the normal equilibrium involving bone marrow
neutrophil production, migration out of the marrow compartments into the
circulation, and neutrophil destruction. Neutrophilia may arise either alone or in
combination with enhanced mobilization into the circulating pool from either
the bone marrow storage compartment or the peripheral blood marginating pool
, by impaired neutrophil egress into tissues, or by expansion of the circulating
neutrophil pool secondary to increased granulocytopoiesis. Myelocytes are not
released to the blood except under extreme circumstances.
Table 158.1
Causes of Neutrophilia
Table 158.2
Causes of Monocytosis
CAUSE EXAMPLE
Infections
Bacterial Brucellosis, subacute bacterial endocarditis, syphilis, tuberculosis, typhoid
infections
Nonbacterial Fungal infections, kala-azar, malaria, Rocky Mountain spotted fever, typhus
infections
Hematologic Congenital and acquired neutropenias, hemolytic anemias
disorders
Malignant Acute myelogenous leukemia, chronic myelogenous leukemia, juvenile myelomonocytic
disorders leukemia, Hodgkin disease, non-Hodgkin lymphomas, preleukemia
Chronic Inflammatory bowel disease, polyarteritis nodosa, rheumatoid arthritis, sarcoidosis, systemic
inflammatory lupus erythematosus
diseases
Miscellaneous Cirrhosis, drug reaction, postsplenectomy, recovery from bone marrow suppression
Eosinophilia
Eosinophilia is defined as an absolute eosinophil count >1500 cells/µL. The
majority of eosinophilic conditions are reactive, including infections (especially
parasitic diseases), connective tissue disorders, allergic and hyperinflammatory
diseases, pulmonary disorders, and dermatologic conditions. Hypereosinophilic
syndrome and systemic mastocytosis are additional important causes of an
elevated eosinophil count. However, persistent eosinophilia can also herald a
malignancy such as leukemia, lymphoma, or carcinoma.
Basophilia
Basophilia is defined as an absolute basophil count >120 cells/µL. Basophilia is
a nonspecific sign of a wide variety of disorders and is usually of limited
diagnostic importance. Basophilia is most often present in hypersensitivity
reactions and frequently accompanies the leukocytosis of chronic myeloid
leukemia.
Lymphocytosis
The most common cause of lymphocytosis is an acute viral illness, as part of the
normal T-cell response to the infection. In infectious mononucleosis, the B cells
are infected with the Epstein-Barr virus, and the T cells react to the viral antigens
present in the B cells, resulting in atypical lymphocytes with characteristic
large, vacuolated morphology. Other viral infections classically associated with
lymphocytosis are cytomegalovirus and viral hepatitis. Chronic bacterial
infections such as tuberculosis and brucellosis may lead to a sustained
lymphocytosis. Pertussis is accompanied by marked lymphocytosis in
approximately 25% of infants infected before 6 mo of age. Thyrotoxicosis and
Addison disease are endocrine disorders associated with lymphocytosis.
CHAPTER 159
Membrane Receptors
Keywords
complement component deficiencies
C1q deficiency
C3 deficiency
MBL deficiency
complement deficiency and meningitis
Table 160.1
Complement Defects
GENETIC
ASSOCIATED
DISEASE DEFECT/PRESUMED INHERITANCE FUNCTIONAL DEFECT
FEATURES
PATHOGENESIS
C1q Mutation in C1QA , AR Absent CH50 hemolytic SLE, infections with
deficiency C1QB , C1QC: classical activity; defective activation of encapsulated
complement pathway the classical pathway, organisms
components diminished clearance of
apoptotic cells
C1r Mutation in C1R: AR Absent CH50 hemolytic SLE, infections with
deficiency classical complement activity; defective activation of encapsulated
pathway component the classical pathway organisms
C1s Mutation in C1S: AR Absent CH50 hemolytic SLE, infections with
deficiency classical complement activity; defective activation of encapsulated
pathway component the classical pathway organisms
C4 Mutation in C4A , C4B: AR Absent CH50 hemolytic SLE, infections with
deficiency classical complement activity; defective activation of encapsulated
pathway components the classical pathway, defective organisms
humoral immune response to
carbohydrate antigens in some
patients
C2 Mutation in C2: AR Absent CH50 hemolytic SLE, infections with
deficiency classical complement activity; defective activation of encapsulated
partway component the classical pathway organisms,
atherosclerosis
C3 Mutation in C3: central AR, gain-of- Absent CH50 and AH50 Infections;
deficiency complement component function AD hemolytic activity; defective glomerulonephritis,
opsonization, defective aHUS with gain-of-
humoral immune response function mutations
C5 Mutation in C5: AR Absent CH50 and AH50 Neisserial infections
deficiency terminal complement hemolytic activity; defective
component bactericidal activity
C6 Mutation in C6: AR Absent CH50 and AH50 Neisserial infections
deficiency terminal complement hemolytic activity; defective
component bactericidal activity
C7 Mutation in C7: AR Absent CH50 and AH50 Neisserial infections
deficiency terminal complement hemolytic activity; defective
component bactericidal activity
C8 α-γ Mutation in C8A, C8G: AR Absent CH50 and AH50 Neisserial infections
deficiency terminal complement hemolytic activity; defective
components bactericidal activity
C8b Mutation in C8B: AR Absent CH50 and AH50 Neisserial infections
deficiency terminal complement hemolytic activity. defective
component bactericidal activity
C9 Mutation in C9: AR Reduced CH50 and AH50 Mild susceptibility to
deficiency terminal complement hemolytic activity; deficient neisserial infections
component bactericidal activity
C1 Mutation in C1NH: AD Spontaneous activation of the Hereditary
inhibitor regulation of kinins and complement pathway with angioedema
deficiency complement activation consumption of C4/C2;
spontaneous activation of the
contact system with generation
of bradykinin from high-
molecular-weight kininogen
Factor B Mutation in CFB: AD Gain-of-function mutation with aHUS
activation of the increased spontaneous AH50
alternative pathway
Factor D Mutation in CFD: AR Absent AH50 hemolytic Neisserial infections
deficiency regulation of the activity
alternative complement
pathway
Properdin Mutation in CFP: XL Absent AH50 hemolytic Neisserial infections
deficiency regulation of the activity
alternative complement
pathway
Factor I Mutation in CFI: AR Spontaneous activation of the Infections, neisserial
deficiency regulation of the alternative complement infections, aHUS,
alternative complement pathway with consumption of preeclampsia,
pathway C3 membranoproliferative
glomerulonephritis
Factor H Mutation in CFH: AR Spontaneous activation of the Infections, neisserial
deficiency regulation of the alternative complement infections, aHUS,
alternative complement pathway with consumption of preeclampsia,
pathway C3 membranoproliferative
glomerulonephritis
MASP-1 Mutation in MASP1: AR Deficient activation of the Infections, 3MC
deficiency cleaves C2 and activates lectin activation pathway, cell syndrome
MASP-2 migration
AD, Autosomal dominant; aHUS, atypical hemolytic-uremic syndrome; AR, autosomal recessive;
SLE, systemic lupus erythematosus; XL, X-linked; 3MC, previously Carnevale, Mingarelli,
Malpuech, and Michels syndromes.
From Kliegman RM, Lye PS, Bordini BJ, et al, editors: Nelson pediatric symptom-based diagnosis,
Philadelphia, 2018, Elsevier, Table 41.11, p 765.
Anemias
Immunologic Disorders
Other Disorders
Patients with sensitive lymphomas and minimal tumor burden have favorable
outcomes after autologous HSCT, with disease-free survival rates of 50–60%,
whereas high-risk patients with bulky tumor or poorly responsive disease have a
poor outcome, with survival rates of 10–20%.
Autologous HSCT in patients with high-risk neuroblastoma is associated with
a better outcome than conventional chemotherapy. A Children's Oncology Group
(COG) study demonstrated further survival advantage by performing 2
sequential, or tandem , transplants that use different chemotherapeutic agents.
Because of these improved outcomes, tandem autologous transplants are now
considered the standard recommended treatment. In these patients,
posttransplantation infusion of a monoclonal antibody directed against a
molecule (GD2) expressed on the surface of neuroblastoma cells confers a
protection against the risk of tumor recurrence.
For children with brain tumors at high risk of relapse, or resistant to
conventional chemotherapy and irradiation, the dose-limiting toxicity for
intensifying therapy is myelosuppression, thus providing a role for stem cell
rescue. Several studies provide encouraging results for patients with different
histologic types of brain tumors treated with autologous HSCT.
Bibliography
Chen Y-B, Wang T, Hemmer MT, et al. GvHD after umbilical
FIG. 163.2 Acute graft-versus-host disease. Almost confluent eruption of erythematous
macules and papules in an immunodeficient neonate treated with extracorporeal
membrane oxygenation (ECMO) and transfusion of nonirradiated blood. (From Paller
AS, Mancini AJ, editors: Hurwitz clinical pediatric dermatology, ed 5, Philadelphia,
2016, Elsevier, p 577.)
Table 163.1
Clinical Staging and Grading* of Graft-Versus-Host Disease
(GVHD)
Table 163.2
Patients with chronic GVHD involving only the skin and liver have a
favorable course (Figs. 163.3 and 163.4 ). Extensive multiorgan disease may be
associated with a very poor quality of life, recurrent infections associated with
prolonged immunosuppressive regimens to control GVHD, and a high mortality
rate. Morbidity and mortality are highest in patients with a progressive onset of
chronic GVHD that directly follows acute GVHD, intermediate in those with a
quiescent onset after resolution of acute GVHD, and lowest in patients with de
novo onset in the absence of acute GVHD. Chronic GVHD can be classified as
mild, moderate, or severe depending on extent of involvement. Single-agent
prednisone is standard treatment at present, although other agents, including
extracorporeal photopheresis, MMF, anti-CD20 mAb, and pentostatin, have been
employed with variable success. Treatment with imatinib mesylate, which
inhibits the synthesis of collagen, has been effective in some patients with
chronic GVHD and sclerotic features. As a consequence of prolonged
immunosuppression, patients with chronic GVHD are particularly susceptible to
infections and should receive appropriate antibiotic prophylaxis, including
trimethoprim/sulfamethoxazole (TMP/SMX). Chronic GVHD resolves in most
pediatric patients but may require 1-3 yr of immunosuppressive therapy before
the drugs can be withdrawn without the disease recurring. Chronic GVHD
promotes the development of secondary neoplasms, in particular in patients with
Fanconi anemia, and has a significant impact on quality of life.
FIG. 163.3 Chronic graft-versus-host disease (GVHD), lichenoid. After bone marrow
transplantation, this boy had acute GVHD and subsequently developed cutaneous
scaling papules and plaques typical of lichen planus. (From Paller AS, Mancini AJ,
editors: Hurwitz clinical pediatric dermatology, ed 5, Philadelphia, 2016, Elsevier, p
577.)
underlying disease alone and require different types of monitoring.
Essentially, every organ system can be impacted by the long-term effects of
therapy, and each must be considered when undergoing late effects surveillance
(Table 165.1 ). As a result of growing evidence of the importance of lifelong
care for HSCT survivors, multiple groups have published recent consensus
guidelines to help in caring for this patient population. As the field of
survivorship continues to expand, we recommend the following reference for
real-time evidence-based recommendations from the Children's Oncology
Group : http://survivorshipguidelines.org .
Table 165.1
Summary of Late Effects After Hematopoietic Stem Cell
Transplantation (HSCT) in Childhood
† At given total dose, risks greater for single-fraction vs fractionated total body irradiation (TBI);
single-fraction myeloablative TBI (>500 cGy) now rarely used.
‡ Effects listed are those more likely to be associated with doses used in HSCT survivors (e.g.,
those given for leukemia treatment, <25 Gy); late effects are more likely if TBI also given.
** Include etoposide, teniposide.
urticaria/angioedema or atopic dermatitis. Xerosis , or dry skin, is the most
common skin abnormality of allergic children. Keratosis pilaris , often found on
facial cheeks and extensor surfaces of the upper arms and thighs, is a benign
condition characterized by skin-colored or slightly pink papules caused by
keratin plugs lodged in the openings of hair follicles. Examination of the skin of
the palms and soles may reveal thickened skin and exaggerated palmar and
plantar creases (hyperlinearity ) in children with moderate to severe atopic
dermatitis.
Diagnostic Testing
In Vitro Tests
Allergic diseases are often associated with increased numbers of eosinophils
circulating in the peripheral blood and invading the tissues and secretions of
target organs. Eosinophilia , defined as the presence of >500 eosinophils/µL in
peripheral blood, is the most common hematologic abnormality of allergic
patients. Seasonal increases in the number of circulating eosinophils may be
observed in sensitized patients after exposure to allergens such as tree, grass, and
weed pollens. The number of circulating eosinophils can be suppressed by
certain infections and systemic corticosteroids. In certain pathologic conditions,
such as drug reactions, eosinophilic pneumonias, and eosinophilic esophagitis,
significantly increased numbers of eosinophils may be present in the target organ
in the absence of peripheral blood eosinophilia. Increased numbers of
eosinophils are observed in a wide variety of disorders in addition to allergy;
eosinophil counts >1500 without an identifiable etiology should suggest 1 of the
2 hypereosinophilic syndromes (Table 167.1 ; see Chapter 155 ).
Table 167.1
Differential Diagnosis of Childhood
Eosinophilia
Physiologic
Prematurity
Infants receiving hyperalimentation
Hereditary
Infectious
Pulmonary
Dermatologic
Atopic dermatitis
Pemphigus
Dermatitis herpetiformis
Infantile eosinophilic pustular folliculitis
Eosinophilic fasciitis (Schulman syndrome)
Eosinophilic cellulitis (Wells syndrome)
Kimura disease (angiolymphoid hyperplasia with eosinophilia)
Hematologic/Oncologic
Immunologic
T-cell immunodeficiencies
Hyper-IgE (Job) syndrome
Wiskott-Aldrich syndrome
Graft-versus-host disease
Drug hypersensitivity
Postirradiation
Postsplenectomy
Endocrine
Addison disease
Hypopituitarism
Cardiovascular
Gastrointestinal
Benign proctocolitis
Inflammatory bowel disease
Eosinophilic gastrointestinal diseases (EGID)
Ascariasis
Capillariasis
Echinococcosis
Fascioliasis
Filariasis
Hookworm
Onchocerciasis
Malaria
Paragonimiasis
Schistosomiasis
Strongyloidiasis
Trichinosis
Visceral larva migrans
Infections
Immunodeficiency
Neoplastic Diseases
Hodgkin disease
IgE myeloma
Bronchial carcinoma
Other Diseases and Disorders
Alopecia areata
Bone marrow transplantation
Burns
Cystic fibrosis
Dermatitis, chronic acral
Erythema nodosum, streptococcal infection
Guillain-Barré syndrome
Kawasaki disease
Liver disease
Medication related
Nephritis, drug-induced interstitial
Nephrotic syndrome
Pemphigus, bullous
Polyarteritis nodosa, infantile
Primary pulmonary hemosiderosis
Juvenile idiopathic arthritis
Table 167.3
In Vivo Tests
Allergen skin testing is the primary in vivo procedure for the diagnosis of
allergic disease. Mast cells with sIgE antibodies attached to high-affinity
receptors on their surface reside in the skin of allergic patients. The introduction
of minute amounts of an allergen into the skin of the sensitized patient results in
cross-linking of IgE antibodies on the mast cell surface, thereby triggering local
mast cell activation. Once activated, these mast cells release a variety of
preformed and newly generated mediators that act on surrounding tissues.
Histamine is the mediator most responsible for the immediate wheal and flare
reactions observed in skin testing. Examination of the site of a positive skin test
result reveals a pruritic wheal surrounded by erythema. The time course of these
reactions is rapid in onset, reaching a peak within 10-20 min and usually
resolving over the next 30 min.
Evaluation of AR entails a thorough history, including details of the patient's
environment and diet and a family history of allergic conditions (e.g., eczema,
asthma, AR), physical examination, and laboratory evaluation. The history and
laboratory findings provide clues to the provoking factors. Symptoms such as
sneezing, rhinorrhea, nasal itching, and congestion and lab findings of elevated
IgE, sIgE antibodies, and positive allergy skin test results typify AR. Intermittent
AR differs from persistent AR by history and skin test results. Nonallergic
rhinitides give rise to sporadic symptoms; their causes are often unknown.
Nonallergic inflammatory rhinitis with eosinophils imitates AR in presentation
and response to treatment, but without elevated IgE antibodies. Vasomotor
rhinitis is characterized by excessive responsiveness of the nasal mucosa to
physical stimuli. Other nonallergic conditions, such as infectious rhinitis,
structural problems (e.g., nasal polyps, septal deviation), rhinitis
medicamentosa (caused by overuse of topical vasoconstrictors), hormonal
rhinitis associated with pregnancy or hypothyroidism, neoplasms, vasculitides,
and granulomatous disorders may mimic AR (Table 168.1 and Fig. 168.2 ).
Occupational risks for rhinitis include exposure to allergens (grain dust, insects,
latex, enzymes) and irritants (wood dust, paint, solvents, smoke, cold air).
Table 168.1
Causes of Rhinitis
ALLERGIC RHINITIS
Seasonal
Perennial
Perennial with seasonal exacerbations
NONALLERGIC RHINITIS
Structural/Mechanical Factors
Infectious
Acute infections
Chronic infections
Inflammatory/Immunologic
Physiologic
FIG. 168.2 Diagnostic algorithm for rhinitis. Nasal allergen challenge is a research
procedure and is not undertaken routinely. CNS, Central nervous system; NSAID,
nonsteroidal antiinflammatory drug. (From Greiner AN, Hellings PW, Rotiroti G,
Scadding GK: Allergic rhinitis, Lancet 378:2112–2120, 2011.)
exposure to mite allergen. Bed linen and blankets should be washed every week
in hot water (>54.4°C [130°F]). The only effective measure for avoiding animal
allergens in the home is the removal of the pet. Avoidance of pollen and outdoor
molds can be accomplished by staying in a controlled environment. Air
conditioning allows for keeping windows and doors closed, reducing the pollen
exposure. High-efficiency particulate air (HEPA) filters lower the counts of
airborne mold spores.
Oral antihistamines help reduce sneezing, rhinorrhea, and ocular symptoms.
Administered as needed, antihistamines provide acceptable treatment for mild-
intermittent disease. Antihistamines have been classified as first generation
(relatively sedating) or second generation (relatively nonsedating).
Antihistamines usually are administered by mouth but are also available for
topical ophthalmic and intranasal use. Both first- and second-generation
antihistamines are available as nonprescription drugs. Second-generation
antihistamines are preferred because they cause less sedation . Preparations
containing pseudoephedrine , typically in combination with other agents, are
used for relief of nasal and sinus congestion and pressure and other symptoms
such as rhinorrhea, sneezing, lacrimation, itching eyes, oronasopharyngeal
itching, and cough. Pseudoephedrine is available without prescription (generally
in fixed combination with other agents such as first-generation antihistamines:
brompheniramine, chlorpheniramine, triprolidine; second-generation
antihistamines: desloratadine, fexofenadine, loratadine; antipyretics:
acetaminophen, ibuprofen; antitussives: guaifenesin, dextromethorphan;
anticholinergic: methscopolamine). Pseudoephedrine is an oral vasoconstrictor
distrusted for causing irritability and insomnia and for its association with infant
mortality. Because younger children (2-3 yr) are at increased risk of overdosage
and toxicity, some manufacturers of oral nonprescription cough and cold
preparations have voluntarily revised their product labeling to warn against the
use of preparations containing pseudoephedrine for children <4 yr old.
Pseudoephedrine is misused as a starting material for the synthesis of
methamphetamine and methcathinone. Tables 168.2 , 168.3 , and 168.4 provide
examples of prescription, nonprescription, and combined oral agents,
respectively, for treatment of AR.
Table 168.2
Oral Allergic Rhinitis Treatments (Prescription, Examples)
GENERIC/BRAND STRENGTH FORMULATIONS DOSING
SECOND-GENERATION ANTIHISTAMINES
Desloratadine
Clarinex Reditabs* 2.5 mg, 5 mg Orally disintegrating Children 6-11 mo of age: 1 mg once daily
tablet
Clarinex Tablets 5 mg Tabs Children 12 mo-5 yr: 1.25 mg once daily
Clarinex Syrup 0.5 mg/mL Syrup Children 6-11 yr: 2.5 mg once daily
Adults and adolescents ≥12 yr: 5 mg once
daily
Levocetirizine
dihydrochloride
Xyzal Oral Solution 0.5 mg/mL Solution 6 mo-5 yr: max 1.25 mg once daily in the
PM
6-11 yr: max 2.5 mg once daily in the PM
LEUKOTRIENE ANTAGONIST
Montelukast
Singulair 10 mg Tablets 6 mo-5 yr: 4 mg daily
Singulair Chewables* 4 mg, 5 mg Chewable tablets 6-14 yr: 5 mg daily
Singulair Oral Granules 4 mg/packet Oral granules >14 yr: 10 mg daily
* Contains phenylalanine.
Dosing recommendations taken in part from Engorn B, Flerlage J, for the Johns Hopkins Hospital:
The Harriet Lane Handbook , ed 20, Philadelphia, 2015, Elsevier/Saunders.
Table 168.3
Oral Allergic Rhinitis Treatments (Nonprescription,
Examples)
Dosing recommendations taken in part from Engorn B, Flerlage J, for the Johns Hopkins Hospital:
The Harriet Lane Handbook , ed 20. Philadelphia, 2015 Elsevier/Saunders.
Table 168.4
Combined Antihistamine + Sympathomimetic (Examples)
Table 168.5
Miscellaneous Intranasal Sprays
Table 168.6
Intranasal Inhaled Corticosteroids
INDICATIONS (I),
MECHANISM(s) OF COMMENTS, CAUTIONS, ADVERSE EVENTS,
DRUG
ACTION (M), AND AND MONITORING
DOSING
Beclomethasone: I: AR Shake container before use; blow nose; occlude 1
OTC (over the M: Antiinflammatory, nostril, administer dose to the other nostril.
counter) immune modulator Adverse effects: Burning and irritation of nasal
Beconase AQ (42 6-12 yr: 1 spray in each mucosa, epistaxis
µg/spray) nostril bid; may increase Monitor growth.
Qnasl (80 µg/spray) if needed to 2 sprays in
OTC each nostril bid
>12 yr: 1 or 2 sprays in
each nostril bid
Flunisolide 6-14 yr: 1 spray each Shake container before use; blow nose; occlude 1
OTC nostril tid or 2 sprays in nostril, administer dose to the other nostril.
each nostril bid; not to Adverse effects: Burning and irritation of nasal
exceed 4 sprays/day in mucosa, epistaxis
each nostril Monitor growth.
≥15 yr: 2 sprays each
nostril bid (morning and
evening); may increase
to 2 sprays tid;
maximum dose: 8
sprays/day in each
nostril (400 µg/day)
Triamcinolone I: AR Shake container before use; blow nose; occlude 1
Nasacort AQ (55 M: Antiinflammatory, nostril, administer dose to the other nostril.
µg/spray) immune modulator Adverse effects: Burning and irritation of nasal
OTC 2-6 yr: 1 spray in each mucosa, epistaxis
Fluticasone nostril qd Monitor growth.
propionate (available 6-12 yr: 1-2 sprays in
as generic each nostril qd
preparation): ≥12 yr: 2 sprays in each
OTC nostril qd
I: AR Shake container before use; blow nose; occlude 1
M: Antiinflammatory, nostril, administer dose to the other nostril.
immune modulator Ritonavir significantly increases fluticasone serum
concentrations and may result in systemic
corticosteroid effects.
Use fluticasone with caution in patients receiving
ketoconazole or other potent cytochrome P450
3A4 isoenzyme inhibitor.
Adverse effects: Burning and irritation of nasal
mucosa, epistaxis
Monitor growth.
Flonase (50 µg/spray) ≥4 yr: 1-2 sprays in each
OTC nostril qd
Fluticasone furoate: 2-12 yr:
Veramyst (27.5 Initial dose: 1 spray
µg/spray) (27.5 µg/spray) per
nostril qd (55 µg/day)
Patients who do not
show adequate response
may use 2 sprays per
nostril qd (110 µg/day)
Once symptoms are
controlled, dosage may
be reduced to 55 µg qd
Total daily dosage
should not exceed 2
sprays in each nostril
(110 µg)/day
≥12 yr and adolescents:
Initial dose: 2 sprays
(27.5 µg/spray) per
nostril qd (110 µg/day)
Once symptoms are
controlled, dosage may
be reduced to 1 spray
per nostril qd (55
µg/day).
Total daily dosage
should not exceed 2
sprays in each nostril
(110 µg)/day.
Mometasone: I: AR Mometasone and its major metabolites are
M: Antiinflammatory, undetectable in plasma after nasal administration
immune modulator of recommended doses.
Nasonex (50 µg/spray) 2-12 yr: 1 spray in each Preventive treatment of seasonal AR should begin
nostril qd 2-4 wk prior to pollen season.
>12 yr: 2 sprays in each Shake container before use; blow nose; occlude 1
nostril qd nostril, administer dose to the other nostril.
Adverse effects: Burning and irritation of nasal
mucosa, epistaxis
Monitor growth.
Severity Classification*
Control Classification*
Management Patterns
* From National Asthma Education and Prevention Program Expert Panel Report
3 (EPR3): Guideline for the diagnosis and management of asthma, NIH Pub No
07-4051, Bethesda, MD, 2007, US Department of Health and Human Services;
National Institutes of Health; National Heart, Lung, and Blood Institute;
National Asthma Education and Prevention Program.
https://www.nhlbi.nih.gov/health-pro/guidelines/current/asthma-guidelines/full-
report .
Pathogenesis
Airflow obstruction in asthma is the result of numerous pathologic processes. In
the small airways, airflow is regulated by smooth muscle encircling the airway
lumen; bronchoconstriction of these bronchiolar muscular bands restricts or
blocks airflow. A cellular inflammatory infiltrate and exudates distinguished by
eosinophils, but also including other inflammatory cell types (neutrophils,
monocytes, lymphocytes, mast cells, basophils), can fill and obstruct the airways
and induce epithelial damage and desquamation into the airways lumen. Helper
T lymphocytes and other immune cells that produce proallergic,
proinflammatory cytokines (interleukin [IL]-4, IL-5, IL-13), and chemokines
(eotaxins) mediate this inflammatory process (see Fig. 169.2 ). Pathogenic
immune responses and inflammation may also result from a breach in normal
immune regulatory processes (e.g., regulatory T lymphocytes that produce IL-10
and transforming growth factor-β) that dampen effector immunity and
inflammation when they are no longer needed. Hypersensitivity or susceptibility
to a variety of provocative exposures or triggers (Table 169.3 ) can lead to
airways inflammation, AHR, edema, basement membrane thickening,
subepithelial collagen deposition, smooth muscle and mucous gland
hypertrophy, and mucus hypersecretion—all processes that contribute to airflow
obstruction.
Table 169.3
Asthma Triggers
COMMON VIRAL INFECTIONS OF RESPIRATORY TRACT
AEROALLERGENS IN SENSITIZED ASTHMATIC PATIENTS
Indoor Allergens
• Animal dander
• Dust mites
• Cockroaches
• Molds
Seasonal Aeroallergens
AIR POLLUTANTS
• Perfumes, hairsprays
• Cleaning agents
OCCUPATIONAL EXPOSURES
• Rhinitis
• Sinusitis
• Gastroesophageal reflux
DRUGS
Table 169.4
Formal Evaluation of Asthma Exacerbation Severity in the
Urgent or Emergency Care Setting*
SUBSET:
RESPIRATORY
MILD MODERATE SEVERE
ARREST
IMMINENT
SYMPTOMS
Breathlessness While walking While at rest (infant— While at rest (infant Extreme dyspnea
softer, shorter cry, —stops feeding) Anxiety
difficulty feeding)
Can lie down Prefers sitting Sits upright Upright, leaning
forward
Talks in… Sentences Phrases Words Unable to talk
Alertness May be agitated Usually agitated Usually agitated Drowsy or confused
SIGNS
Respiratory rate † Increased Increased Often >30
breaths/min
Use of accessory Usually not Commonly Usually Paradoxical
muscles; suprasternal thoracoabdominal
retractions movement
Wheeze Moderate; often Loud; throughout Usually loud; Absence of wheeze
only end- exhalation throughout
expiratory inhalation and
exhalation
Pulse rate (beats/min) <100 100-120 >120 Bradycardia
‡
Pulsus paradoxus Absent May be present Often present Absence suggests
<10 mm 10-25 mm Hg >25 mm Hg respiratory muscle
Hg (adult) fatigue
20-40 mm Hg
(child)
FUNCTIONAL ASSESSMENT
Peak expiratory flow ≥70% Approx. 40–69% or <40% <25% §
(value predicted or response lasts <2 hr
personal best)
PaO 2 (breathing air) Normal (test not ≥60 mm Hg (test not <60 mm Hg;
usually usually necessary) possible cyanosis
necessary)
and/or
PCO 2 <42 mm Hg <42 mm Hg (test not ≥42 mm Hg;
(test not usually usually necessary) possible respiratory
necessary) failure
SaO 2 (breathing air) at >95% (test not 90–95% (test not usually <90% Hypoxia despite
sea level usually necessary) oxygen therapy
necessary)
Hypercapnia (hypoventilation) develops more readily in young children than in adults
and adolescents.
* Notes:
† Normal breathing rates in awake children by age: <2 mo, <60 breaths/min; 2-12 mo, <50
<110 beats/min.
§ Peak expiratory flow testing may not be needed in very severe attacks.
• The presence of several parameters, but not necessarily all, indicates the general classification
of the exacerbation.
• Many of these parameters have not been systematically studied, especially as they correlate
with each other; thus they serve only as general guides.
• The emotional impact of asthma symptoms on the patient and family is variable but must be
recognized and addressed and can affect approaches to treatment and follow-up.
Adapted from National Asthma Education and Prevention Program Expert Panel Report 3
(EPR3): Guideline for the diagnosis and management of asthma, NIH Pub No 07-4051, Bethesda,
MD, 2007, US Department of Health and Human Services; National Institutes of Health; National
Heart, Lung, and Blood Institute; National Asthma Education and Prevention Program.
https://www.nhlbi.nih.gov/health-pro/guidelines/current/asthma-guidelines/full-report .
Differential Diagnosis
Many childhood respiratory conditions can present with symptoms and signs
similar to those of asthma (Table 169.5 ). Besides asthma, other common causes
of chronic, intermittent coughing include gastroesophageal reflux (GER) and
rhinosinusitis. Both GER and chronic sinusitis can be challeng spirometric lung
function testing ing to diagnose in children. Often, GER is clinically silent in
children, and children with chronic sinusitis do not report sinusitis-specific
symptoms, such as localized sinus pressure and tenderness. In addition, both
GER and rhinosinusitis are often comorbid with childhood asthma and, if not
specifically treated, may make asthma difficult to manage.
Table 169.5
Differential Diagnosis of Childhood Asthma
Upper Respiratory Tract Conditions
Allergic rhinitis*
Chronic rhinitis*
Sinusitis*
Adenoidal or tonsillar hypertrophy
Nasal foreign body
Laryngotracheobronchomalacia*
Laryngotracheobronchitis (e.g., pertussis)*
Laryngeal web, cyst, or stenosis
Exercise-induced laryngeal obstruction
Vocal cord dysfunction*
Vocal cord paralysis
Tracheoesophageal fistula
Vascular ring, sling, or external mass compressing on the airway (e.g.,
tumor)
Endobronchial tumor
Foreign body aspiration*
Chronic bronchitis from environmental tobacco smoke exposure*
Repaired tracheoesophageal fistula
Toxic inhalations
In early life, chronic coughing and wheezing can indicate recurrent aspiration,
tracheobronchomalacia (congenital anatomic abnormality of airways), foreign
body aspiration, cystic fibrosis, or bronchopulmonary dysplasia.
In older children and adolescents, vocal cord dysfunction (VCD) can
manifest as intermittent daytime wheezing. The vocal cords involuntarily close
inappropriately during inspiration and sometimes exhalation, producing
shortness of breath, coughing, throat tightness, and often audible laryngeal
wheezing and/or stridor. In most cases of VCD, spirometric lung function testing
reveals truncated and inconsistent inspiratory and expiratory flow-volume loops,
a pattern that differs from the reproducible pattern of airflow limitation in
asthma that improves with bronchodilators. VCD can coexist with asthma.
Hypercarbia and severe hypoxia are uncommon in VCD. Flexible
rhinolaryngoscopy in the patient with symptomatic VCD can reveal paradoxical
vocal cord movements with anatomically normal vocal cords. Prior to the
diagnosis, patients with VCD are often treated unsuccessfully with multiple
different classes of asthma medications. This condition can be well managed
with specialized speech therapy training in the relaxation and control of vocal
cord movement. Furthermore, treatment of underlying causes of vocal cord
irritability (e.g., high GER/aspiration, allergic rhinitis, rhinosinusitis, asthma)
FIG. 169.3 Spirometry. A, Spirometric flow-volume loops. Loop A is an expiratory flow-
volume loop of a nonasthmatic person without airflow limitation. B through E are
expiratory flow-volume loops in asthmatic patients with increasing degrees of airflow
limitation (B is mild; E is severe). Note the “scooped” or concave appearance of the
asthmatic expiratory flow-volume loops; with increasing obstruction, there is greater
“scooping.” B, Spirometric volume-time curves. Subject 1 is a nonasthmatic person;
subject 2 is an asthmatic patient. Note how the FEV1 and FVC lung volumes are
obtained. The FEV1 is the volume of air exhaled in the 1st sec of a forced expiratory
effort. The FVC is the total volume of air exhaled during a forced expiratory effort, or
forced vital capacity. Note that subject 2's FEV1 and FEV1 :FVC ratio are smaller than
subject 1's, demonstrating airflow limitation. Also, subject 2's FVC is very close to what
is expected.
FEV1 , forced expiratory volume in 1 sec; FVC, forced vital capacity; ICS,
inhaled corticosteroid; ppb, parts per billion.
† Of note, >50%of children with mild to moderate asthma will have a normal
Table 169.7
Assessing Asthma Severity and Initiating Treatment for
Patients Who Are Not Currently Taking Long-Term Control
Medications*
• Level of severity is determined by both impairment and risk. Assess impairment domain by
patient's/caregiver's recall of previous 2-4 wk. Symptom assessment for longer periods should
reflect a global assessment, such as inquiring whether a patient's asthma is better or worse since
the last visit. Assign severity to the most severe category in which any feature occurs.
• At present, there are inadequate data to correspond frequencies of exacerbations with different
levels of asthma severity. For treatment purposes, patients who had ≥2 exacerbations requiring
oral systemic corticosteroids in the past 6 mo, or ≥4 wheezing episodes in the past year, and who
have risk factors for persistent asthma, may be considered the same as patients who have
persistent asthma, even in the absence of impairment levels consistent with persistent asthma.
FEV1 , Forced expiratory volume in 1 sec; FVC, forced vital capacity; CS corticosteroid; ICS,
inhaled corticosteroid: EIB, exercise-induced bronchospasm.
Adapted from the National Asthma Education and Prevention Program Expert Panel Report 3
(EPR3): Guidelines for the diagnosis and management of asthma—summary report 2007, J
Allergy Clin Immunol 120(Suppl):S94–S138, 2007.
Table 169.8
Assessing Asthma Control and Adjusting Therapy in
Children*
†
Validated questionnaires for the impairment domain (the questionnaires do not assess lung
function or the risk domain) and definition of minimal important difference (MID) for each:
‡ ACQ values of 0.76-1.40 are indeterminate regarding well-controlled asthma.
§ Before step-up therapy: (a) review adherence to medications, inhaler technique, and
environmental control; (b) if alternative treatment option was used in a step, discontinue it and use
preferred treatment for that step.
• The stepwise approach is meant to assist, not replace, the clinical decision making required to
meet individual patient needs.
• The level of control is based on the most severe impairment or risk category. Assess impairment
domain by caregiver's recall of previous 2-4 wk. Symptom assessment for longer periods should
reflect a global assessment, such as inquiring whether the patient's asthma is better or worse
since the last visit.
• At present, there are inadequate data to correspond frequencies of exacerbations with different
levels of asthma control. In general, more frequent and intense exacerbations (e.g., requiring
urgent, unscheduled care, hospitalization, or intensive care unit admission) indicate poorer
disease control. For treatment purposes, patients who had ≥2 exacerbations requiring oral
systemic corticosteroids in the past year may be considered the same as patients who have not-
well-controlled asthma, even in the absence of impairment levels consistent with not-well-
controlled asthma.
• ATAQ, Asthma Therapy Assessment Questionnaire; MID = 1.0.
• ACQ, Asthma Control Questionnaire; MID = 0.5.
• ACT, Asthma Control Test; MID not determined.
FEV1 , Forced expiratory volume in 1 sec; FVC, forced vital capacity; EIB, exercise-induced
at home can be helpful in the assessment of asthmatic children with poor
symptom perception, moderate to severe asthma, or a history of severe asthma
exacerbations. PEF monitoring is feasible in children as young as 4 yr who are
able to master this skill. Use of a stoplight zone system tailored to each child's
“personal best” PEF values can optimize effectiveness and interest (see Fig.
169.4 ): The green zone (80–100% of personal best) indicates good control; the
yellow zone (50–80%) indicates less-than-optimal control and necessitates
increased awareness and treatment; and the red zone (<50%) indicates poor
control and greater likelihood of an exacerbation, requiring immediate
intervention. In actuality, these ranges are approximate and may need to be
adjusted for many asthmatic children by raising the ranges that indicate
inadequate control (e.g., yellow zone, 70–90% in children with poor perception
and those with lung hyperinflation). Once-daily PEF monitoring is preferable in
the morning when peak flows are typically lower. Adherence to PEF monitoring
is difficult, results may be variable and PEF monitoring alone is not more
effective than symptoms monitoring on influencing asthma outcomes. Therefore,
although PEF may be helpful in some circumstances to monitor those who are
poor perceivers of airway obstruction, PEF monitoring is no longer generally
recommended.
Adherence
Asthma is a chronic condition that is usually best managed with daily controller
medication. However, symptoms wax and wane, severe exacerbations are
infrequent, and when asthma is asymptomatic, a natural tendency is to reduce or
discontinue daily controller therapies. As such, adherence to a daily controller
regimen is frequently suboptimal; ICSs are underused 60% of the time. In one
study, children with asthma who required an oral corticosteroid course for an
asthma exacerbation had used their daily controller ICS 15% of the time.
Misconceptions about controller medication time to onset, efficacy, and safety
often underlie poor adherence and can be addressed by asking about such
concerns at each visit.
• Rhinitis
• Sinusitis
• Gastroesophageal reflux
Table 169.11
Stepwise Approach for Managing Asthma in Children*
THERAPY INTERMITTENT
AGE PERSISTENT ASTHMA: DAILY MEDICATION
† ASTHMA
Alternative Cromolyn
or
montelukast
5-11 Preferred SABA prn Low-dose Either low- Medium-dose High-dose ICS +
yr ICS dose ICS ± ICS + LABA LABA
LABA,
LTRA, or
theophylline
or
Medium-
dose ICS
Alternative Cromolyn, Medium- High-dose
LTRA, dose ICS + ICS + either
nedocromil, either LTRA
or LTRA or
theophylline or Theophylline
Theophylline
† Alphabetical order is used when more than 1 treatment option is listed within either preferred or
alternative therapy.
• The stepwise approach is meant to assist, not replace, the clinical decision making required to
meet individual patient needs.
• If alternative treatment is used and response is inadequate, discontinue it and use the preferred
treatment before stepping up.
• If clear benefit is not observed within 4-6 wk and patient/family medication technique and
adherence are satisfactory, consider adjusting therapy or alternative diagnosis.
• Studies on children age 0-4 yr are limited.
• Clinicians who administer immunotherapy or omalizumab should be prepared and equipped to
identify and treat anaphylaxis that may occur.
Long-Term Controller Medications
All levels of persistent asthma should be treated with ICS therapy to reduce
airway inflammation and improve long-term control (see Table 169.11 ). Other
long-term controller medications include LABAs, leukotriene modifiers,
cromolyn, sustained-release theophylline, and tiotropium in adolescents.
Omalizumab (Xolair) and mepolizumab (Nucala) are approved by the U.S. Food
and Drug Administration (FDA) for use as an add-on therapy in children ≥6 yr
and ≥12 yr who have severe allergic asthma or eosinophilic asthma, respectively,
that remains difficult to control. Corticosteroids are the most potent and most
effective medications used to treat both the acute (administered systemically)
and the chronic (administered by inhalation) manifestations of asthma. They are
available in inhaled, oral, and parenteral forms (Tables 169.12 and 169.13 ).
Table 169.12
Usual Dosages for Long-Term Control Medications
AGE
MEDICATION
0-4 yr 5-11 yr ≥12 yr
INHALED CORTICOSTEROIDS (see Table 169.13 )
Methylprednisolone: 0.25-2 mg/kg 0.25-2 mg/kg daily in 7.5-60 mg daily in a
2, 4, 8, 16, 32 mg daily in single single dose in AM or qod single dose in AM or
tablets dose in AM or as needed for control qod as needed for
Prednisolone: qod as needed Short-course “burst”: 1-2 control
5 mg tablets; 5 mg/5 for control mg/kg/day; maximum 60 Short-course “burst” to
mL, 15 mg/5 mL Short-course mg/day for 3-10 days achieve control: 40-60
Prednisone: “burst”: 1-2 mg/day as single or 2
1, 2.5, 5, 10, 20, 50 mg mg/kg/day; divided doses for 3-10
tablets; 5 mg/mL, 5 maximum 30 days
mg/5 mL mg/day for 3-10
days
Fluticasone/salmeterol N/A
(Advair):
DPI: 100, 250, or 500 µg/50 1 inhalation bid; dose depends 1 inhalation bid; dose
µg on level of severity or control depends on level of severity
(the 100/50 dosage is indicated or control
in children ≥4 yr)
HFA: 45 µg/21 µg, 115 2 inhalations bid; dose
µg/21 µg, 230 µg/21 µg depends on level of severity
or control
Budesonide/formoterol N/A 2 inhalations bid; dose
(Symbicort): depends on level of severity
HFA: 80 µg/4.5 µg, 160 or control
µg/4.5 µg
Mometasone/formoterol 2 inhalations bid; dose
(Dulera): depends on level of severity
HFA: 100 µg/5 µg, 200 or control
µg/5 µg
Leukotriene receptor
antagonists:
Montelukast 4 mg qhs (1-5 yr of 5 mg qhs (6-14 yr) 10 mg qhs (indicated in
(Singulair): age) children ≥15 yr)
4 or 5 mg chewable
tablet
4 mg granule packets
10 mg tablet
Zafirlukast (Accolate): N/A 10 mg bid (7-11 yr) 40 mg daily (20 mg tablet
10 or 20 mg tablet bid)
5-Lipoxygenase N/A N/A 1,200 mg bid (give 2 tablets
inhibitor: bid)
(Zileuton CR): 600 mg
tablet
Immunomodulators:
Omalizumab (anti-IgE; N/A N/A 150-375 mg SC q 2-4 wk,
Xolair): depending on body weight
SC injection, 150 and pretreatment serum IgE
mg/1.2 mL after level
reconstitution with 1.4
mL sterile water for
injection
Mepolizumab (anti–IL- N/A N/A 100 mg SC q 4 wk
5; Nucala):
SC injection, 100 mg
after reconstitution with
1.2 mL sterile water for
injection
bid, 2 times daily; DPI, dry powder inhaler; HFA, hydrofluoroalkane; MDI, metered-dose inhaler; q,
every; qhs, every night; qid, 4 times daily; qod, every other day; SC, subcutaneous(ly).
Table 169.13
Estimated Comparative Inhaled Corticosteroid Doses
Inhaled Corticosteroids
ICS therapy improves lung function; reduces asthma symptoms, AHR, and use
of “rescue” medications; improves quality of life; and most importantly reduces
the need for prednisone, urgent care visits, and hospitalizations by approximately
50%. Epidemiologic studies have also shown that ICS therapy substantially
lowers the risk of death attributable to asthma if used regularly. Because ICS
therapy can achieve all the goals of asthma management, it is viewed as first-line
treatment for persistent asthma.
Seven ICSs are FDA approved for use in children. The NIH and GINA
guidelines provide equivalence classifications (see Table 169.13 ), although
direct comparisons of efficacy and safety outcomes are lacking. ICSs are
available in metered-dose inhalers (MDIs) using hydrofluoroalkane (HFA) as
their propellant, in dry powder inhalers (DPIs), or in suspension for nebulization.
Fluticasone propionate, fluticasone furoate, mometasone furoate, ciclesonide,
and to a lesser extent budesonide are considered “second-generation” ICSs, in
that they have greater antiinflammatory potency and less systemic bioavailability
(and thus potential for systemic adverse effects) because of their extensive first-
pass hepatic metabolism. The selection of the initial ICS dose is based on the
determination of disease severity.
Even though ICSs can be very effective, there has been some reluctance to
treat children with ICSs due to parental and occasionally physician concerns
regarding their potential for adverse effects with chronic use. The adverse effects
that occur with long-term systemic corticosteroid therapy have not been seen or
have only rarely been reported in children receiving ICSs in recommended
doses. The risk of adverse effects from ICS therapy is related to the dose and
frequency of administration (Table 169.14 ). High doses (≥1,000 µg/day in
children) and frequent administration (4 times/day) are more likely to have both
local and systemic adverse effects. Children who receive maintenance therapy
with higher ICS doses are also likely to require frequent systemic corticosteroid
courses for asthma exacerbations, further increasing their risk of corticosteroid
adverse effects.
Table 169.14
Quick-Reliever Medications
Quick-reliever or “rescue” medications (SABAs, inhaled anticholinergics, and
short-course systemic corticosteroids) are used in the management of acute
asthma symptoms (Table 169.15 ).
Table 169.15
Management of Asthma Exacerbation (Status Asthmaticus)
RISK ASSESSMENT ON ADMISSION
Focused history Onset of current exacerbation
Frequency and severity of daytime and nighttime symptoms and activity limitation
Frequency of rescue bronchodilator use
Current medications and allergies
Potential triggers
History of systemic steroid courses, emergency department visits, hospitalization,
intubation, or life-threatening episodes
Clinical assessment Physical examination findings: vital signs, breathlessness, air movement, use of
accessory muscles, retractions, anxiety level, alteration in mental status
Pulse oximetry
Lung function (defer in patients with moderate to severe distress or history of labile
disease)
Risk factors for asthma See Table 169.16 .
morbidity and death
TREATMENT
Drug and Trade Name Mechanisms of Action and Dosing Cautions and Adverse Effects
Oxygen (mask or nasal Treats hypoxia Monitor pulse oximetry to
cannula) maintain O2 saturation >92%
Cardiorespiratory monitoring
Inhaled short-acting β- Bronchodilator During exacerbations, frequent
agonists: or continuous doses can cause
pulmonary vasodilation, V̇/Q̇
mismatch, and hypoxemia.
Adverse effects: palpitations,
tachycardia, arrhythmias,
tremor, hypoxemia
Albuterol nebulizer Nebulizer: 0.15 mg/kg (minimum 2.5 mg) as Nebulizer: when giving concentrated
solution (5 mg/mL often as every 20 min for 3 doses as needed, then forms, dilute with saline to 3 mL
concentrate; 2.5 mg/3 0.15-0.3 mg/kg up to 10 mg every 1-4 hr as total nebulized volume.
mL, 1.25 mg/3 mL, 0.63 needed, or up to 0.5 mg/kg/hr by continuous
mg/3 mL) nebulization
Albuterol MDI (90 2-8 puffs up to every 20 min for 3 doses as For MDI: use spacer/holding
µg/puff) needed, then every 1-4 hr as needed chamber
Levalbuterol (Xopenex) 0.075 mg/kg (minimum 1.25 mg) every 20 min Levalbuterol 0.63 mg is equivalent
nebulizer solution (1.25 for 3 doses, then 0.075-0.15 mg/kg up to 5 mg to 1.25 mg of standard albuterol for
mg/0.5 mL concentrate; every 1-4 hr as needed, or 0.25 mg/kg/hr by both efficacy and side effects.
0.31 mg/3 mL, 0.63 continuous nebulization
mg/3 mL, 1.25 mg/3
mL)
Systemic Antiinflammatory If patient has been exposed to
corticosteroids: chickenpox or measles,
consider passive
immunoglobulin prophylaxis;
also, risk of complications with
herpes simplex and
tuberculosis.
For daily dosing, 8 AM
administration minimizes
adrenal suppression.
Children may benefit from
dosage tapering if course
exceeds 7 days.
Adverse effects monitoring:
frequent therapy bursts risk
numerous corticosteroid
adverse effects (see Chapter 595
); see Table 169.14 for adverse
effects screening
recommendations.
Prednisone: 1, 2.5, 0.5-1 mg/kg every 6-12 hr for 48 hr, then 1-2
5, 10, 20, 50 mg mg/kg/day bid (maximum 60 mg/day)
tablets
Methylprednisolone
(Medrol): 2, 4, 8,
16, 24, 32 mg
tablets
Prednisolone: 5 mg
tablets; 5 mg/5 mL
and 15 mg/5 mL
solution
Depo-Medrol (IM); Short-course “burst” for exacerbation: 1-2
Solu-Medrol (IV) mg/kg/day qd or bid for 3-7 days
Dexamethasone See text See text
Anticholinergics: Mucolytic/bronchodilator Should not be used as first-line
therapy; added to β2 -agonist
therapy
Ipratropium:
Atrovent (nebulizer Nebulizer: 0.5 mg q6-8h (tid-qid) as needed
solution 0.5 mg/2.5 mL; MDI: 2 puffs qid
MDI 18 µg/inhalation)
Ipratropium with
albuterol:
DuoNeb nebulizer 1 vial by nebulizer qid Nebulizer: may mix ipratropium
solution (0.5 mg with albuterol
ipratropium + 2.5 mg
albuterol/3 mL vial)
Injectable Bronchodilator For extreme circumstances (e.g.,
sympathomimetic impending respiratory failure despite
epinephrine: high-dose inhaled SABA,
respiratory failure)
Adrenalin 1 mg/mL SC or IM: 0.01 mg/kg (max dose 0.5 mg); may
(1 : 1000) repeat after 15-30 min
EpiPen
autoinjection device
(0.3 mg; EpiPen Jr
0.15 mg)
Terbutaline: Terbutaline is β-agonist–
selective relative to
epinephrine.
Monitoring with continuous
infusion: cardiorespiratory
monitor, pulse oximetry, blood
pressure, serum potassium
Adverse effects: tremor,
tachycardia, palpitations,
arrhythmia, hypertension,
headaches, nervousness, nausea,
vomiting, hypoxemia
Brethine 1 mg/mL Continuous IV infusion (terbutaline only): 2-
10 µg/kg loading dose, followed by 0.1-0.4
µg/kg/min
Titrate in 0.1-0.2 µg/kg/min increments
every 30 min, depending on clinical
response.
RISK ASSESSMENT FOR DISCHARGE
Medical stability Discharge home if there has been sustained
improvement in symptoms and bronchodilator
treatments are at least 3 hr apart, physical
findings are normal, PEF >70% of predicted or
personal best, and oxygen saturation >92% when
breathing room air.
Home supervision Capability to administer intervention and to
observe and respond appropriately to clinical
deterioration
Asthma education See Table 169.8 .
(Courtesy of BJC Healthcare/Washington University School of Medicine,
Community Asthma Program, January 2000.)
Table 169.16
Risk Factors for Asthma Morbidity and
Mortality
Biologic
Environmental
Allergen exposure
Environmental tobacco smoke exposure
Air pollution exposure
Urban environment
Poverty
Crowding
Mother <20 yr old
Mother with less than high school education
Inadequate medical care:
Inaccessible
Unaffordable
No regular medical care (only emergency)
Lack of written Asthma Action Plan
No care sought for chronic asthma symptoms
Delay in care of asthma exacerbations
Inadequate hospital care for asthma exacerbation
Psychopathology in the parent or child
Poor perception of asthma symptoms or severity
Alcohol or substance abuse
Pruritus
Facial and extensor eczema in infants and children
Flexural eczema in adolescents
Chronic or relapsing dermatitis
Personal or family history of atopic disease
Associated Features
Xerosis
Cutaneous infections (Staphylococcus aureus, group A streptococcus,
herpes simplex, coxsackievirus, vaccinia, molluscum, warts)
Nonspecific dermatitis of the hands or feet
Ichthyosis, palmar hyperlinearity, keratosis pilaris
Nipple eczema
White dermatographism and delayed blanch response
Anterior subcapsular cataracts, keratoconus
Elevated serum IgE levels
Positive results of immediate-type allergy skin tests
Early age at onset
Dennie lines (Dennie-Morgan infraorbital folds)
Facial erythema or pallor
Course influenced by environmental and/or emotional factors
Table 170.2
Differential Diagnosis of Atopic Dermatitis (AD)
MAIN AGE
GROUP FREQUENCY* CHARACTERISTICS AND CLINICAL FEATURES
AFFECTED
OTHER TYPES OF DERMATITIS
Seborrheic Infants Common Salmon-red greasy scaly lesions, often on the scalp (cradle
dermatitis cap) and napkin area; generally presents in the 1st 6 wk of
life; typically clears within weeks
Seborrheic Adults Common Erythematous patches with yellow, white, or grayish scales
dermatitis in seborrheic areas, particularly the scalp, central face, and
anterior chest
Nummular Children and Common Coin-shaped scaly patches, mostly on legs and buttocks;
dermatitis adults usually no itch
Irritant contact Children and Common Acute to chronic eczematous lesions, mostly confined to the
dermatitis adults site of exposure; history of locally applied irritants is a risk
factor; might coexist with AD
Allergic contact Children and Common Eczematous rash with maximum expression at sites of direct
dermatitis adults exposure but might spread; history of locally applied
irritants is a risk factor; might coexist with AD
Lichen simplex Adults Uncommon One or more localized, circumscribed, lichenified plaques
chronicus that result from repetitive scratching or rubbing because of
intense itch
Asteatotic eczema Adults Common Scaly, fissured patches of dermatitis overlying dry skin,
most often on lower legs
INFECTIOUS SKIN DISEASES
Dermatophyte Children and Common One or more demarcated scaly plaques with central clearing
infection adults and slightly raised reddened edge; variable itch
Impetigo Children Common Demarcated erythematous patches with blisters or honey-
yellow crusting
Scabies Children Common † Itchy superficial burrows and pustules on palms and soles,
between fingers, and on genitalia; might produce secondary
eczematous changes
HIV Children and Uncommon Seborrhea-like rash
adults
CONGENITAL IMMUNODEFICIENCIES (see Table 170.3 )
Keratinization Disorders
Ichthyosis Infants and Uncommon Dry skin with fine scaling, particularly on the lower
vulgaris adults abdomen and extensor areas; perifollicular skin roughening;
palmar hyperlinearity; full form (i.e., 2 FLG mutations) is
uncommon; often coexists with AD
NUTRITIONAL DEFICIENCY–METABOLIC DISORDERS
Zinc deficiency Children Uncommon Erythematous scaly patches and plaques, most often around
(acrodermatitis the mouth and anus; rare congenital form accompanied by
enteropathica) diarrhea and alopecia
Biotin deficiency Infants Uncommon Scaly periorofacial dermatitis, alopecia, conjunctivitis,
(nutritional or lethargy, hypotonia
biotinidase
deficiency)
Pellagra (niacin All ages Uncommon Scaly crusted epidermis, desquamation, sun-exposed areas,
deficiency) diarrhea
Kwashiorkor Infants and Geographic Flaky scaly dermatitis, swollen limbs with cracked peeling
children dependent patches
Phenylketonuria Infants Uncommon Eczematous rash, hypopigmentation, blonde hair,
developmental delay
NEOPLASTIC DISEASE
Cutaneous T-cell Adults Uncommon Erythematous pink-brown macules and plaques with a fine
lymphoma scale; poorly responsive to topical corticosteroids; variable
itch (in early stages)
Langerhans cell Infants Uncommon Scaly and purpuric dermatosis, hepatosplenomegaly,
histiocytosis cytopenias
* Common = approximately 1 in 10 to 1 in 100; uncommon = 1 in 100 to 1 in 1000; rare = 1 in
Table 170.3
Features of Primary Immunodeficiencies Associated With
Eczematous Dermatitis
Treatment
The treatment of AD requires a systematic, multifaceted approach that
incorporates skin moisturization, topical antiinflammatory therapy, identification
and elimination of flare factors (Table 170.4 ), and, if necessary, systemic
therapy. Assessment of the severity also helps direct therapy (Table 170.5 ).
Table 170.4
Counseling and Aggravating Factors for
Patients With Atopic Dermatitis (AD)
Maintain cool temperature in bedroom, and avoid too many bed covers.
Increase emollient use with cold weather.
Avoid exposure to herpes sores; urgent visit if flare of unusual aspect.
Clothing: Avoid skin contact with irritating fibers (wool, large-fiber
textiles).
Do not use tight and too-warm clothing, to avoid excessive
sweating.
New, nonirritating clothing designed for AD children is being
evaluated.
Tobacco: Avoid exposure.
Vaccines: Normal schedule in noninvolved skin, including egg-allergic
patients (see text).
Sun exposure: No specific restriction.
Usually helpful because of improvement of epidermal barrier.
Encourage summer holidays in altitude or at beach resorts.
Physical exercise, sports: no restriction.
If sweating induces flares of AD, progressive adaptation to
exercise.
Shower and emollients after swimming pool.
Food allergens:
Maintain breastfeeding exclusively to 4-6 mo if possible.
Consider evaluation for early introduction of allergens (see
Chapter 176 ).
Otherwise normal diet, unless an allergy workup has proved the
need to exclude a specific food.
Indoor aeroallergens: House dust mites
Use adequate ventilation of housing; keep the rooms well aerated
even in winter.
Avoid wall-to-wall carpeting.
Remove dust with a wet sponge.
Vacuum floors and upholstery with an adequately filtered cleaner
once a week.
Avoid soft toys in bed (cradle), except washable ones.
Wash bedsheets at a temperature higher than 55°C (131°F) every
10 days.
Use bed and pillow encasings made of Gore-Tex or similar
material.
Furred pets: Advise to avoid. If allergy is demonstrated, be firm on
avoidance measures, such as pet removal.
Pollen: Close windows during peak pollen season on warm and dry
weather days, and restrict, if possible, time outdoors.
Windows may be open at night and early in the morning or during
rainy weather.
Avoid exposure to risk situations (lawn mowing).
Use pollen filters in motor vehicles.
Clothes and pets can vectorize aeroallergens, including pollen.
Cutaneous Hydration
ointments have a greater potential to occlude the epidermis, resulting in
enhanced systemic absorption.
Table 170.6
Selected Topical Corticosteroid Preparations*
Group 1
Group 2
Group 3
Group 4
Group 5
Group 6
Group 7
7 (least potent).
Table 171.1
Indications for Venom Immunotherapy (VIT) Against
Winged Hymenoptera
Table 172.1
Topical Ophthalmic Medications for Allergic Conjunctivitis
DRUG AND MECHANISM OF ACTION
CAUTIONS AND ADVERSE EVENTS
TRADE NAMES AND DOSING
Azelastine Antihistamine Not for treatment of contact lens–related irritation; the
hydrochloride Children ≥3 yr: 1 gtt bid preservative may be absorbed by soft contact lenses.
0.05% Wait at least 10 min after administration before
Optivar inserting soft contact lenses.
Emedastine Antihistamine Soft contact lenses should not be worn if the eye is
difumarate Children ≥3 yr: 1 gtt qid red. Wait at least 10 min after administration before
0.05% inserting soft contact lenses.
Emadine
Levocabastine Antihistamine Not for use in patients wearing soft contact lenses
hydrochloride Children ≥12 yr: 1 gtt bid-qid during treatment.
0.05% up to 2 wk
Livostin
Pheniramine maleate Antihistamine/vasoconstrictor Avoid prolonged use (>3-4 days) to avoid rebound
symptoms. Not for use with contact lenses.
0.3% Naphazoline Children >6 yr: 1-2 gtt qid
hydrochloride
0.025%
Naphcon-A, Opcon-
A
Cromolyn Mast cell stabilizer Can be used to treat giant papillary conjunctivitis and
sodium 4% Children >4 yr: 1-2 gtt q4-6h vernal keratitis. Not for use with contact lenses.
Crolom,
Opticrom
Lodoxamide Mast cell stabilizer Can be used to treat vernal keratoconjunctivitis. Not
tromethamine Children ≥2 yr: 1-2 gtt qid up for use in patients wearing soft contact lenses during
0.1% to 3 mo treatment.
Alomide
Nedocromil sodium Mast cell stabilizer Avoid wearing contact lenses while exhibiting the
2% Alocril Children ≥3 yr: 1-2 gtt bid signs and symptoms of allergic conjunctivitis.
Pemirolast Mast cell stabilizer Not for treatment of contact lens–related irritation; the
potassium 0.1% Children >3 yr: 1-2 gtt qid preservative may be absorbed by soft contact lenses.
Alamast Wait at least 10 min after administration before
inserting soft contact lenses.
Epinastine Antihistamine/mast cell Contact lenses should be removed before use. Wait at
hydrochloride stabilizer least 15 min after administration before inserting soft
0.05% Children ≥3 yr: 1 gtt bid contact lenses. Not for the treatment of contact lens
Elestat irritation.
Ketotifen Antihistamine/mast cell Not for treatment of contact lens–related irritation; the
fumarate 0.025% stabilizer preservative may be absorbed by soft contact lenses.
Zaditor Children ≥3 yr: 1 gtt bid q8- Wait at least 10 min after administration before
12h inserting soft contact lenses.
Olopatadine Antihistamine/mast cell Not for treatment of contact lens–related irritation; the
hydrochloride stabilizer preservative may be absorbed by soft contact lenses.
0.1%, 0.2%, Children ≥3 yr: 1 gtt bid (8 hr Wait at least 10 min after administration before
0.7% apart) inserting soft contact lenses.
Patanol Children≥2 yr: 1 gtt qd
Pataday
Pazeo
Alcaftadine, Antihistamine/mast cell Contact lenses should be removed before application;
0.25% stabilizer may be inserted after 10 min. Not for the treatment of
Lastacaft Children > 2 yr: 1 gtt bid q8- contact lens irritation.
12h
Bepotastine Antihistamine/mast cell Contact lenses should be removed before application,
besilate 1.5% stabilizer may be inserted after 10 min. Not for the treatment of
Bepreve Children >2 yr: 1 gtt bid q8- contact lens irritation.
12h
Ketorolac NSAID Avoid with aspirin or NSAID sensitivity. Use ocular
tromethamine Children ≥3 yr: 1 gtt qid product with caution in patients with complicated
0.5% ocular surgeries, corneal denervation or epithelial
Acular defects, ocular surface diseases (e.g., dry eye
syndrome), repeated ocular surgeries within a short
period, diabetes mellitus, or rheumatoid arthritis; these
patients may be at risk for corneal adverse events that
may be sight threatening. Do not use while wearing
contact lenses.
Fluorometholone Fluorinated corticosteroid If improvement does not occur after 2 days, patient
0.1%, 0.25% Children ≥2 yr, 1 gtt into should be reevaluated. Patient should remove soft
suspension conjunctival sac of affected contact lenses before administering (contains
(0.1%, 0.25%) eye(s) bid-qid. During initial benzalkonium chloride) and delay reinsertion of lenses
and ointment 24-48 hr, dosage may be for ≥15 min after administration. Close monitoring for
(0.1%) increased to 1 gtt q4h. development of glaucoma and cataracts.
FML, FML Ointment (~1.3 cm in length)
Forte, Flarex into conjunctival sac of
affected eye(s) 1-3 times
daily. May be applied q4h
during initial 24-48 hr of
therapy.
NSAID, Nonsteroidal antiinflammatory drug; bid, 2 times daily; gtt, drops; qid, 4 times daily; q4-6h;
every 4-6 hr; qd, every day.
Bibliography
Bielory BP, Shah SP, O'Brien TP, et al. Emerging therapeutics
for ocular surface disease. Curr Opin Allergy Clin Immunol .
2016;16(5):477–486.
Castillo M, Scott NW, Mustafa MZ, et al. Topical
antihistamines and mast cell stabilisers for treating seasonal
and perennial allergic conjunctivitis. Cochrane Database Syst
Rev . 2015;(6) [CD009566].
Chen JJ, Applebaum DS, Sun GS, et al. Atopic
keratoconjunctivitis: a review. J Am Acad Dermatol .
2014;70(3):569–575.
Esposito S, Fior G, Mori A, et al. An update on the therapeutic
CHAPTER 173
Urticaria and angioedema affect 20% of individuals at some point in their life.
Episodes of hives that last for <6 wk are considered acute, whereas those that
occur on most days of the week for >6 wk are designated chronic. The
distinction is important, because the causes and mechanisms of urticaria
formation and the therapeutic approaches are different in each instance.
Table 173.1
Table 173.2
Etiology of Chronic Urticaria
Idiopathic/autoimmune Approximately 30% of chronic urticaria cases are physical urticaria, and 60–70% are
idiopathic. Of the idiopathic cases approximately 35–40% have anti-IgE or anti-
FcεRI (high-affinity IgE receptor α chain) autoantibodies (autoimmune chronic
urticaria)
Physical Dermatographism
Cholinergic urticaria
Cold urticaria (see Table 173.5 )
Delayed pressure urticaria
Solar urticaria
Vibratory urticaria
Aquagenic urticaria
Autoimmune diseases Systemic lupus erythematosus
Juvenile idiopathic arthritis
Thyroid disease (Graves, Hashimoto)
Celiac disease
Inflammatory bowel disease
Leukocytoclastic vasculitis
Autoinflammatory/periodic See Tables 173.3 and 173.5 .
fever syndromes
Neoplastic Lymphoma
Mastocytosis
Leukemia
Angioedema Hereditary angioedema (autosomal dominant inherited deficiency of C1-esterase
inhibitor)
Acquired angioedema
Angiotensin-converting enzyme inhibitors
From Lasley MV, Kennedy MS, Altman LC: Urticaria and angioedema. In Altman LC, Becker JW,
Williams PV, editors: Allergy in primary care , Philadelphia, 2000, Saunders, p 234.
Table 173.3
Febrile Autoinflammatory Diseases Causing Urticaria in
Children
TIMING
GENE ATTACK CUTANEOUS EXTRACUTANEOUS
DISEASE INHERITANCE OF
(PROTEIN) LENGTH FEATURES CLINICAL FEATURES
ONSET
FCAS NLRP3 AD Brief; Neonatal Cold-induced Arthralgia
(cryopyrin) minutes to or urticaria Conjunctivitis
3 days infantile Headache
Muckle- NLRP3 AD 1-3 days Neonatal, Widespread urticaria Arthralgia/arthritis
Wells (cryopyrin) infantile, Sensorineural hearing
syndrome childhood loss
(can be Conjunctivitis/episcleritis
later) Headache
Amyloidosis
Chronic NLRP3 AD Continuous Neonatal Widespread urticaria Deforming
infantile (cryopyrin) flares or osteoarthropathy,
neurologic infantile epiphyseal overgrowth
cutaneous Sensorineural hearing
articular loss
syndrome; Dysmorphic facies
neonatal- Chronic aseptic
onset meningitis, headaches,
multisystem papilledema, seizures
inflammatory Conjunctivitis/uveitis,
disease optic atrophy
Growth retardation
Developmental delay
Amyloidosis
HIDS MVK AR 3-7 days Infancy Intermittent Arthralgia/arthritis
(mevalonate (<2 yr) morbilliform or Cervical
kinase) urticarial rash lymphadenopathy
Aphthous Severe abdominal pain
mucosal ulcers Diarrhea/vomiting
Erythema Headache
nodosum Elevated IgD and IgA
antibody levels
Elevated urine mevalonic
acid during attacks
Tumor TNFRSF1A AD >7 days Childhood Intermittent Migratory myalgia
necrosis (TNFR1) migratory Conjunctivitis
factor erythematous Serositis
receptor– macules and Amyloidosis
associated edematous
periodic plaques
syndrome overlying areas
of myalgia, often
on limbs
Periorbital
edema
Systemic- Polygenic Varies Daily Peak Nonfixed Polyarthritis
onset (quotidian) onset at 1- erythematous Myalgia
juvenile 6 yr rash; may be Hepatosplenomegaly
idiopathic urticarial Lymphadenopathy
arthritis With or without Serositis
(SoJIA) dermatographism
With or without
periorbital edema
PLAID PLCG2 AD N/A Infancy Urticaria induced Allergies
by evaporative Autoimmune disease
cooling Recurrent sinopulmonary
Ulcers in cold- infections
exposed areas Elevated IgE antibody
levels
Decreased IgA and IgM
antibody levels
Often elevated
antinuclear antibody
titers
AD, Autosomal dominant; AR, autosomal recessive; HIDS, hyperimmunoglobulinemia D
syndrome; FCAS, familial cold-induced autoinflammatory syndrome; N/A, not available; PLAID,
PLCγ2-associated antibody deficiency and immune dysregulation.
From Youseff MJ, Chiu YE: Eczema and urticaria as manifestations of undiagnosed and rare
diseases, Pediatr Clin North Am 64:39–56, 2017 (Table 2, pp 49–50).
Table 173.4
Physical Urticaria
Physically induced urticaria and angioedema share the common property of
being induced by an environmental stimulus, such as a change in temperature or
direct stimulation of the skin with pressure, stroking, vibration, or light (see
Table 173.2 ).
Cold-Dependent Disorders
Cold urticaria is characterized by the development of localized pruritus,
erythema, and urticaria/angioedema after exposure to a cold stimulus. Total body
exposure, as seen with swimming in cold water, can cause massive release of
vasoactive mediators, resulting in hypotension, loss of consciousness, and even
death if not promptly treated. The diagnosis is confirmed by challenge testing for
an isomorphic cold reaction by holding an ice cube in place on the patient's skin
for 5 min. In patients with cold urticaria, an urticarial lesion develops about 10
min after removal of the ice cube and on rewarming of the chilled skin. Cold
urticaria can be associated with the presence of cryoproteins such as cold
agglutinins, cryoglobulins, cryofibrinogen, and the Donath-Landsteiner antibody
seen in secondary syphilis (paroxysmal cold hemoglobinuria). In patients with
cryoglobulins the isolated proteins appear to transfer cold sensitivity and activate
the complement cascade on in vitro incubation with normal plasma. The term
idiopathic cold urticaria generally applies to patients without abnormal
circulating plasma proteins such as cryoglobulins. Cold urticaria has also been
reported after viral infections. Cold urticaria must be distinguished from the
familial cold autoinflammatory syndrome (see later, Diagnosis) (Table 173.5 ;
see also Table 173.3 and Chapter 188 ).
Table 173.5
Hereditary Diseases With Cold-Induced Urticaria
EPISODIC SYMPTOMS SUSTAINED/PROGRESSIVE SYMPTOMS
CAPS FCAS Urticarial rash, arthralgia, myalgia, chills, Renal amyloidosis
fever, swelling of extremities
MWS Urticarial rash, arthralgia, chills, fever Sensorineural deafness, renal amyloidosis
CINCA Fever Rash, arthritis, chronic meningitis, visual defect,
deafness, growth retardation, renal amyloidosis
NAPS12 Fever, arthralgia, myalgia, urticaria, Sensorineural deafness
(FCAS2) abdominal pain, aphthous ulcers,
lymphadenopathy
PLAID Urticaria induced by evaporative cooling, Serum low IgM and IgA levels; high IgE levels;
(FCAS3) sinopulmonary infections decreased B and NK cells; granulomata; antinuclear
antibodies
CAPS, Cryopyrin-associated periodic syndromes; FCAS, familial cold-induced autoinflammatory
syndrome; MWS, Muckle-Wells syndrome; CINCA, chronic infantile neurologic cutaneous articular
syndrome; NAPS, NLRP-12–associated periodic syndrome; PLAID, PLCG2-associated antibody
deficiency and immune dysregulation.
From Kanazawa N: Hereditary disorders presenting with urticaria, Immunol Allergy Clin NORTH
Am 34:169–179, 2014 (Table 4, p 176).
Cholinergic Urticaria
Cholinergic urticaria is characterized by the onset of small, punctate pruritic
wheals surrounded by a prominent erythematous flare and associated with
exercise, hot showers, and sweating. Once the patient cools down, the rash
usually subsides in 30-60 min. Occasionally, symptoms of more generalized
cholinergic stimulation, such as lacrimation, wheezing, salivation, and syncope,
are observed. These symptoms are mediated by cholinergic nerve fibers that
innervate the musculature via parasympathetic neurons and innervate the sweat
glands by cholinergic fibers that travel with the sympathetic nerves. Elevated
plasma histamine values parallel the onset of urticaria triggered by changes in
body temperature.
Dermatographism
The ability to write on skin, dermatographism (also called dermographism or
urticaria factitia ) may occur as an isolated disorder or may accompany chronic
urticaria or other physical urticaria. It can be diagnosed by observing the skin
after stroking it with a tongue depressor. In patients with dermatographism, a
linear response occurs secondary to reflex vasoconstriction, followed by
pruritus, erythema, and a linear flare caused by secondary dilation of the vessel
and extravasation of plasma.
complexes, malignancies, mixed connective tissue diseases, and cutaneous
blistering disorders (e.g., bullous pemphigoid; see Table 173.2 ). In general,
laboratory testing should be limited to a complete blood cell count with
differential, ESR determination, urinalysis, thyroid autoantibody testing, and
liver function tests. Further studies are warranted if the patient has fever,
arthralgias, or elevated ESR (Table 173.6 ; see also Table 173.4 ). Testing for
antibodies directed at the high-affinity IgE receptor may be warranted in patients
with intractable urticaria. Hereditary angioedema is potentially life threatening,
usually associated with deficient C1 inhibitor activity, and the most important
familial form of angioedema (see Chapter 160.3 ), but it is not associated with
typical urticaria. In patients with eosinophilia, stools should be obtained for ova
and parasite testing, because infection with helminthic parasites has been
associated with urticaria. A syndrome of episodic angioedema/urticaria and fever
with associated eosinophilia has been described in both adults and children. In
contrast to other hypereosinophilic syndromes, this entity has a benign course.
Table 173.6
Treatment
Acute urticaria is a self-limited illness requiring little treatment other than
antihistamines and avoidance of any identified trigger. Hydroxyzine and
diphenhydramine are sedating but are effective and frequently used for treatment
of urticaria. Loratadine, fexofenadine, and cetirizine are also effective and are
preferable because of reduced frequency of drowsiness and longer duration of
action (Table 173.7 ). Epinephrine 1 : 1,000, 0.01 mL/kg (maximum 0.3 mL)
intramuscularly, usually provides rapid relief of acute, severe
urticaria/angioedema but is seldom required. A short course of oral
corticosteroids should be given only for severe episodes of urticaria and
angioedema that are unresponsive to antihistamines.
Table 173.7
Treatment of Urticaria and Angioedema
† Monitor complete blood count and liver function tests at baseline, every 2 wk for 3 mo, and then
Hereditary Angioedema
Hereditary angioedema (HAE , types 1 and 2) is an inherited autosomal
dominant disease caused by low functional levels of the plasma protein C1
CHAPTER 174
Anaphylaxis
Hugh A. Sampson, Julie Wang, Scott H. Sicherer
Table 174.1
Symptoms and Signs of Anaphylaxis in Infants
ANAPHYLAXIS
SYMPTOMS ANAPHYLAXIS SIGNS THAT MAY BE DIFFICULT
ANAPHYLAXIS SIGNS IN
THAT INFANTS TO INTERPRET/UNHELPFUL IN INFANTS, AND
INFANTS
CANNOT WHY
DESCRIBE
GENERAL
Feeling of Nonspecific behavioral changes such as persistent crying,
warmth, fussing, irritability, fright, suddenly becoming quiet
weakness,
anxiety,
apprehension,
impending doom
SKIN/MUCOUS MEMBRANES
Itching of lips, Flushing (may also occur with fever, hyperthermia, or Rapid onset of hives (potentially
tongue, palate, crying spells) difficult to discern in infants with
uvula, ears, throat, acute atopic dermatitis; scratching
nose, eyes, etc.; and excoriations will be absent in
mouth-tingling or young infants); angioedema (face,
metallic taste tongue, oropharynx)
RESPIRATORY SYSTEM
Nasal congestion, Hoarseness, dysphonia (common after a crying spell); Rapid onset of coughing,
throat tightness; drooling or increased secretions (common in infants) choking, stridor, wheezing,
chest tightness; dyspnea, apnea, cyanosis
shortness of
breath
GASTROINTESTINAL SYSTEM
Dysphagia, Spitting up/regurgitation (common after feeds), loose Sudden, profuse vomiting
nausea, stools (normal in infants, especially if breastfed); colicky
abdominal abdominal pain
pain/cramping
CARDIOVASCULAR SYSTEM
Feeling faint, Hypotension (need appropriate-size blood pressure cuff; Weak pulse, arrhythmia,
presyncope, low systolic blood pressure for children is defined as <70 diaphoresis/sweating,
dizziness, mm Hg from 1 mo to 1 yr, and less than (70 mm Hg + [2 collapse/unconsciousness
confusion, blurred × age in yr]) from 1-10 yr; tachycardia, defined as >140
vision, difficulty beats/min from 3 mo to 2 yr, inclusive; loss of bowel and
in hearing bladder control (ubiquitous in infants)
CENTRAL NERVOUS SYSTEM
Headache Drowsiness, somnolence (common in infants after feeds) Rapid onset of unresponsiveness,
lethargy, or hypotonia; seizures
Adapted from Simons FER: Anaphylaxis in infants: can recognition and management be
improved? J Allergy Clin Immunol 120:537–540, 2007.
products. Patients with latex allergy may also experience food-allergic reactions
from homologous proteins in foods such as bananas, kiwi, avocado, chestnut,
and passion fruit. Anaphylaxis to galactose-α-1,3-galactose has been reported 3-
6 hr after eating red meat.
Table 174.2
Anaphylaxis Triggers in the Community*
Allergen Triggers (IgE-Dependent Immunologic Mechanism)*
Foods (e.g., peanut, tree nuts, shellfish, fish, milk, egg, wheat, soy, sesame,
meat [galactose-α-1,3-galactose])
Food additives (e.g., spices, colorants, vegetable gums, contaminants)
Stinging insects: Hymenoptera species (e.g., bees, yellow jackets, wasps,
hornets, fire ants)
Medications (e.g., β-lactam antibiotics, ibuprofen)
Biologic agents (e.g., monoclonal antibodies [infliximab, omalizumab] and
allergens [challenge tests, specific immunotherapy])
Natural rubber latex
Vaccines
Inhalants (rare) (e.g., horse or hamster dander, grass pollen)
Previously unrecognized allergens (foods, venoms, biting insect saliva,
medications, biologic agents)
cold water.
Adapted from Leung DYM, Sampson HA, Geha RS, et al: Pediatric allergy
principles and practice , Philadelphia, 2010, Elsevier, p 652.
Epidemiology
The overall annual incidence of anaphylaxis in the United States is estimated at
42 cases per 100,000 person-years, totaling >150,000 cases/yr. Food allergens
are the most common trigger in children, with an incidence rate of
approximately 20 per 100,000 person-years. An Australian parental survey
found that 0.59% of children 3-17 yr of age had experienced at least 1
anaphylactic event. Having asthma and the severity of asthma are important
anaphylaxis risk factors (Table 174.3 ). In addition, patients with systemic
mastocytosis or monoclonal mast cell–activating syndrome are at increased risk
for anaphylaxis, as are patients with an elevated baseline serum tryptase level.
Table 174.3
Patient Risk Factors for Anaphylaxis
Age-Related Factors
Drugs
β-Adrenergic blockers †
Angiotensin-converting enzyme (ACE) inhibitors †
Sedatives, antidepressants, narcotics, recreational drugs, and alcohol may
decrease the patient's ability to recognize triggers and symptoms.
Age
Asthma
Atopy
Drugs
Alcohol
Other cofactors such as exercise, infection, menses
* Atopic diseases are a risk factor for anaphylaxis triggered by food, latex, and
exercise, but not for anaphylaxis triggered by most drugs or by insect stings.
† Patients taking β-blockers or ACE inhibitors seem to be at increased risk for
Pathogenesis
Principal pathologic features in fatal anaphylaxis include acute bronchial
obstruction with pulmonary hyperinflation, pulmonary edema, intraalveolar
hemorrhaging, visceral congestion, laryngeal edema, and urticaria and
angioedema. Acute hypotension is attributed to vasomotor dilation and cardiac
dysrhythmias.
Most cases of anaphylaxis are believed to be the result of activation of mast
cells and basophils via cell-bound allergen-specific IgE molecules (see Fig.
174.1 ). Patients initially must be exposed to the responsible allergen to generate
allergen-specific antibodies. In many cases the child and the parent are unaware
of the initial exposure, which may be from passage of food proteins in maternal
breast milk or exposure to inflamed skin (e.g., eczematous lesions). When the
child is reexposed to the sensitizing allergen, mast cells and basophils, and
possibly other cells such as macrophages, release a variety of mediators
(histamine, tryptase) and cytokines that can produce allergic symptoms in any or
all target organs. Clinical anaphylaxis may also be caused by mechanisms other
than IgE-mediated reactions, including direct release of mediators from mast
cells by medications and physical factors (morphine, exercise, cold),
disturbances of leukotriene metabolism (aspirin and nonsteroidal
antiinflammatory drugs), immune aggregates and complement activation (blood
products), probable complement activation (radiocontrast dyes, dialysis
membranes), and IgG-mediated reactions (high-molecular-weight dextran,
chimeric or humanized monoclonal antibodies) (see Table 174.2 ).
Idiopathic anaphylaxis is a diagnosis of exclusion when no inciting agent is
identified and other disorders have been excluded (see Chapter 678.1 ).
Symptoms are similar to IgE mediated causes of anaphylaxis; episodes often
recur.
Clinical Manifestations
The onset of symptoms may vary depending on the cause of the reaction.
man syndrome (caused by vancomycin) should be considered.
Table 174.4
Diagnosis of Anaphylaxis
Anaphylaxis is highly likely when any 1 of the following 3 criteria is
fulfilled:
1. Acute onset of an illness (minutes to several hours) with
involvement of the skin and/or mucosal tissue (e.g., generalized
hives, pruritus or flushing, swollen lips/tongue/uvula)
AND at least 1 of the following :
a. Respiratory compromise (e.g., dyspnea,
wheeze/bronchospasm, stridor, reduced peak PEF,
hypoxemia)
b. Reduced BP or associated symptoms of end-organ
dysfunction (e.g., hypotonia [collapse], syncope,
incontinence)
2. Two or more of the following that occur rapidly after exposure
to a likely allergen for that patient (minutes to several hours):
a. Involvement of the skin/mucosal tissue (e.g.,
generalized hives, itch/flush, swollen
lips/tongue/uvula)
b. Respiratory compromise (e.g., dyspnea,
wheeze/bronchospasm, stridor, reduced PEF,
hypoxemia)
c. Reduced BP or associated symptoms (e.g., hypotonia
[collapse], syncope, incontinence)
d. Persistent gastrointestinal symptoms (e.g., crampy
abdominal pain, vomiting)
3. Reduced BP following exposure to known allergen for that
patient (minutes to several hours):
a. Infants and children: low systolic BP (age specific) or
>30% drop in systolic BP
b. Adults: systolic BP <90 mm Hg or >30% drop from
patient's baseline
Treatment
Anaphylaxis is a medical emergency requiring aggressive management with
intramuscular (IM, first line) or intravenous (IV) epinephrine, IM or IV H1 and
H2 antihistamine antagonists, oxygen, IV fluids, inhaled β-agonists, and
corticosteroids (Table 174.5 and Fig. 174.2 ). The initial assessment should
ensure an adequate airway with effective respiration, circulation, and perfusion.
Epinephrine is the most important medication, and there should be no delay in
its administration. Epinephrine should be given by the IM route to the lateral
thigh (1 : 1000 dilution, 0.01 mg/kg; maximum 0.5 mg). For children ≥12 yr,
many recommend the 0.5 mg IM dose. The IM dose can be repeated at intervals
of 5-15 min if symptoms persist or worsen. If there is no response to multiple
doses of epinephrine, IV epinephrine using the 1 : 10,000 dilution may be
needed. If IV access is not readily available, epinephrine can be administered via
the endotracheal or intraosseous routes.
Table 174.5
Management of a Patient With Anaphylaxis
MECHANISM(S)
TREATMENT DOSAGE(S) COMMENTS; ADVERSE REACTIONS
OF EFFECT
PATIENT EMERGENCY MANAGEMENT (dependent on severity of symptoms)
Epinephrine α1 -, β1 -, β2 - 0.01 mg/kg, up to Tachycardia, hypertension, nervousness,
(adrenaline) Adrenergic effects 0.5 mg IM in headache, nausea, irritability, tremor
lateral thigh
Adrenaclick,
Auvi-Q, EpiPen
Jr/EpiPen:
0.15 mg IM for 8-
25 kg
0.3 mg IM for 25
kg or more
Epinephrine
autoinjector:
0.1 mg for 7.5-15
kg
0.15 mg for 15 to
25 kg
0.3 mg for 25 kg
or more
Cetirizine (liquid) Antihistamine Cetirizine liquid: 5 Hypotension, tachycardia, somnolence
(competitive of H1 mg/5 mL
receptor) 0.25 mg/kg, up to
10 mg PO
Alternative: Antihistamine 1.25 mg/kg up to 50 Hypotension, tachycardia, somnolence,
Diphenhydramine (competitive of H1 mg PO or IM paradoxical excitement
receptor)
Transport to an emergency facility
EMERGENCY PERSONNEL MANAGEMENT (dependent on severity of symptoms)
Epinephrine α1 -, β1 -, β2 - 0.01 mg/kg, up to Tachycardia, hypertension, nervousness,
(adrenaline) Adrenergic effects 0.5 mg IM in headache, nausea, irritability, tremor
lateral thigh
Epinephrine
autoinjector:
0.1 mg for 7.5-15
kg
0.15 mg for 15 to
25 kg
0.3 mg for 25 kg
or more
0.01 mL/kg/dose
of 1 : 1,000 (vial)
solution, up to 0.5
mL IM
May repeat every
10-15 min
For severe
hypotension: 0.01
mL/kg/dose of 1 :
10,000 slow IV
push
Supplemental oxygen and airway management
Volume Expanders
Crystalloids 30 mL/kg in 1st hr Rate titrated against BP response
(normal saline or If tolerated, place patient supine with legs
Ringer lactate) raised.
Colloids 10 mL/kg rapidly Rate titrated against BP response
(hydroxyethyl followed by slow If tolerated, place patient supine with legs
starch) infusion raised.
Antihistamines
Cetirizine (liquid) Antihistamine Cetirizine liquid: 5 Hypotension, tachycardia, somnolence
(competitive of H1 mg/5 mL
receptor) 0.25 mg/kg, up to
10 mg PO
Alternative: Antihistamine 1.25 mg/kg, up to 50 Hypotension, tachycardia, somnolence,
Diphenhydramine (competitive of H1 mg PO, IM, or IV paradoxical excitement
receptor)
severity.
† Median times to respiratory or cardiac arrest are 5 min in iatrogenic
Adapted from Leung DYM, Szefler SJ, Bonilla FA Akdis CA, Sampson HA,
editors: Pediatric allergy principles and practice, Philadelphia, 2016, Elsevier, p
531.
Bibliography
Akin C. Mast cell activation syndromes presenting as
anaphylaxis. Immunol Allergy Clin North Am .
2015;35(2):277–285.
Alqurashi W, Stiell I, Chan K, et al. Epidemiology and clinical
predictors of biphasic reactions in children with anaphylaxis.
Ann Allergy Asthma Immunol . 2015;115(3):217–223.e2.
Boyce JA, Assa'ad A, Burks AW, et al. Guidelines for the
diagnosis and management of food allergy in the United
States: report of the NIAID-sponsored expert panel. J Allergy
Clin Immunol . 2010;126:S1–S58.
Brown JC, Tuuri RE, Akhter S, et al. Lacerations and embedded
needles caused by epinephrine autoinjector use in children.
Ann Emerg Med . 2017;67(3):307–315.
Dahdah L, Ceccarelli S, Amendola S, et al. IgE
immunoadsorption knocks down the risk of food-related
anaphylaxis. Pediatrics . 2015;136(6):e1617–e1620.
Dhami S, Panesar SS, Roberts G, et al. Management of
anaphylaxis: a systematic review. Allergy . 2014;69(2):168–
175.
Farbman KS, Michelson KA. Anaphylaxis in children. Curr
Opin Pediatr . 2016;28:294–297.
Fellinger C, Hemmer W, Wohrl S, et al. Clinical characteristics
and risk profile of patients with elevated baseline serum
CHAPTER 175
Serum Sickness
Anna Nowak-Węgrzyn, Scott H. Sicherer
Antibotulinum globulin
Antithymocyte globulin
Antitetanus toxoid
Antivenin (Crotalidae) polyvalent (horse serum based)
Crotalidae polyvalent immune Fab (ovine serum based)
Antirabies globulin
Infliximab
Rituximab
Etanercept
Anti-HIV antibodies ([PE]HRG214)
Hymenoptera stings
Streptokinase
H1N1 influenza vaccine
Drugs
Antibiotics
Cefaclor
Penicillins
Trimethoprim sulfate
Minocycline
Meropenem
Neurologic
Bupropion
Carbamazepine
Phenytoin
Sulfonamides
Barbiturates
* Based on review of most current literature. Other medications that are not
From Aceves SS: Serum sickness. In Burg FD, Ingelfinger JR, Polin RA,
Gershon AA, editors: Current pediatric therapy , ed 18, Philadelphia, 2006,
Elsevier, p 1138.
Etiology
Immune complexes involving heterologous (animal) serum proteins and
complement activation are important pathogenic mechanisms in serum sickness.
Antibody therapies derived from the horse, sheep, or rabbit are available for
treatment of envenomation by the black widow spider and a variety of snakes,
for treatment of botulism, and for immunosuppression (antithymocyte globulin
, ATG). The availability of alternative medical therapies, modified or
bioengineered antibodies, and biologics of human origin have supplanted the use
of nonhuman antisera, reducing the risk of serum sickness. However, rabbit-
generated ATGs, which target human T cells, continue to be widely used as
immunosuppressive agents during treatment of kidney allograft recipients; serum
CHAPTER 176
Food Allergy
IgE Mediated
Non–IgE Mediated
IgE, Immunoglobulin E.
Table 176.2
Differential Diagnosis of Adverse Food
Reactions
Gastrointestinal Disorders (with vomiting and/or diarrhea)
Food phobias
Genetics
Genetic factors play an important role in the development of food allergy.
Family and twin studies show that family history confers a 2-10–fold increased
risk, depending on the study setting, population, specific food, and diagnostic
test. Candidate gene studies suggest that genetic variants in the HLA-DQ locus
(HLA-DQB1*02 and DQB1*06:03P), filaggrin, interleukin-10, STAT6, and
FOXP3 genes are associated with food allergy, although the results are
inconsistent across different populations. In a genome-wide association study,
differential methylation at the HLA-DR and -DQ regions was associated with
food allergy. Epigenetic studies implicate DNA methylation effects on
interleukins 4, 5, and 10 and interferon (IFN)-γ genes and in the mitogen-
activated protein kinase (MAPK) pathway.
Pathogenesis
Food intolerances are the result of a variety of mechanisms, whereas food allergy
is predominantly caused by IgE-mediated and cell-mediated immune
mechanisms. In susceptible individuals exposed to certain allergens, food-
specific IgE antibodies are formed that bind to Fcε receptors on mast cells,
basophils, macrophages, and dendritic cells. When food allergens penetrate
mucosal barriers and reach cell-bound IgE antibodies, mediators are released
that induce vasodilation, smooth muscle contraction, and mucus secretion, which
result in symptoms of immediate hypersensitivity (allergy). Activated mast cells
and macrophages may release several cytokines that attract and activate other
cells, such as eosinophils and lymphocytes, leading to prolonged inflammation.
Symptoms elicited during acute IgE-mediated reactions can affect the skin
(urticaria, angioedema, flushing), gastrointestinal (GI) tract (oral pruritus,
angioedema, nausea, abdominal pain, vomiting, diarrhea), respiratory tract (nasal
congestion, rhinorrhea, nasal pruritus, sneezing, laryngeal edema, dyspnea,
wheezing), and cardiovascular system (dysrhythmias, hypotension, loss of
consciousness). In non-IgE food allergies, lymphocytes, primarily food allergen–
specific T cells, secrete excessive amounts of various cytokines that lead to a
“delayed,” more chronic inflammatory process affecting the skin (pruritus,
erythematous rash), GI tract (failure to thrive, early satiety, abdominal pain,
vomiting, diarrhea), and respiratory tract (food-induced pulmonary
hemosiderosis). Mixed IgE and cellular responses to food allergens can also lead
to chronic disorders, such as atopic dermatitis, asthma, eosinophilic esophagitis,
and gastroenteritis.
Children who develop IgE-mediated food allergies may be sensitized by food
allergens penetrating the GI barrier, referred to as class 1 food allergens , or by
food allergens that are partially homologous to plant pollens penetrating the
respiratory tract, referred to as class 2 food allergens . Any food may serve as a
class 1 food allergen, but egg, milk, peanuts, tree nuts, fish, soy, and wheat
account for 90% of food allergies during childhood. Many of the major
allergenic proteins of these foods have been characterized. There is variable but
significant cross-reactivity with other proteins within an individual food group.
Exposure and sensitization to these proteins often occur very early in life.
Virtually all milk allergies develop by 12 mo of age and all egg allergies by 18
mo, and the median age of 1st peanut allergic reactions is 14 mo. Class 2 food
allergens are typically vegetable, fruit, or nut proteins that are partially
homologous with pollen proteins (Table 176.3 ). With the development of
seasonal allergic rhinitis from birch, grass, or ragweed pollens, subsequent
ingestion of certain uncooked fruits or vegetables provokes the oral allergy
syndrome . Intermittent ingestion of allergenic foods may lead to acute
symptoms such as urticaria or anaphylaxis, whereas prolonged exposure may
lead to chronic disorders such as atopic dermatitis and asthma. Cell-mediated
sensitivity typically develops to class 1 allergens.
Table 176.3
Natural History of Food Allergy and Cross-Reactivity
Between Common Food Allergies
§ Allergy to fresh apples, carrots, and peaches (oral allergy syndrome ) is typically caused by
heat-labile proteins. Fresh fruit causes oral pruritus, but cooked fruit is tolerated. There is
generally no risk of anaphylaxis, although in rare cases, allergies to cross-reactive lipid transfer
protein can cause anaphylaxis after ingestion of fruits (e.g., peach) and vegetables.
Adapted from Lack G: Food allergy, N Engl J Med 359:1252–1260, 2008.
Clinical Manifestations
From a clinical and diagnostic standpoint, it is most useful to subdivide food
hypersensitivity disorders according to the predominant target organ (Table
176.4 ) and immune mechanism (see Table 176.1 ).
Table 176.4
Gastrointestinal Manifestations
GI food allergies are often the 1st form of allergy to affect infants and young
children and typically manifest as irritability, vomiting or “spitting-up,” diarrhea,
and poor weight gain. Cell-mediated hypersensitivities without IgE involvement
predominate, making standard allergy tests such as skin-prick tests and in vitro
tests for food-specific IgE antibodies of little diagnostic value.
Food protein–induced enterocolitis syndrome (FPIES) typically manifests
in the 1st several mo of life as irritability, intermittent vomiting, and protracted
diarrhea and may result in dehydration (Table 176.5 ). Vomiting generally occurs
1-4 hr after feeding, and continued exposure may result in abdominal distention,
bloody diarrhea, anemia, and failure to thrive. Symptoms are most often
provoked by cow's milk or soy protein–based formulas. A similar enterocolitis
syndrome occurs in older infants and children from rice, oat, wheat, egg, peanut,
nut, chicken, turkey, or fish. Hypotension occurs in approximately 15% of
patients after allergen ingestion and may initially be thought to be caused by
sepsis. FPIES usually resolves by age 3-5 yr.
Table 176.5
Food Protein–Induced Gastrointestinal Syndromes
EOSINOPHILIC
FPIES PROCTOCOLITIS ENTEROPATHY
GASTROENTEROPATHIES*
Age at onset 1 day–1 year 1 day–6 months Dependent of age Infant to adolescent
of exposure to
antigen, cow's
milk and soy up to
2 yr
Food proteins
implicated
Most common Cow's milk, Cow's milk, soy Cow's milk, soy Cow's milk, soy, egg white,
soy wheat, peanut
Less common Rice, Egg, corn, chocolate Wheat, egg Meats, corn, rice, fruits,
chicken, vegetables, fish
turkey, fish,
pea
Multiple food >50% both 40% both cow's milk Rare Common
hypersensitivities cow's milk and soy
and soy
Feeding at the time Formula >50% exclusive Formula Formula
of onset breastfeeding
Atopic background
Family history of 40–70% 25% Unknown ~50% (often history of
atopy eosinophilic esophagitis)
Personal history of 30% 22% 22% ~50%
atopy
Symptoms
Emesis Prominent No Intermittent Intermittent
Diarrhea Severe No Moderate Moderate
Bloody stools Severe Moderate Rare Moderate
Edema Acute, severe No Moderate Moderate
Shock 15% No No No
Failure to thrive Moderate No Moderate Moderate
Laboratory
findings
Anemia Moderate Mild Moderate Mild-moderate
Hypoalbuminemia Acute Rare Moderate Mild-severe
Methemoglobinemia May be No No No
present
Allergy evaluation
Food skin-prick test Negative † Negative Negative Positive in ~50%
Serum food allergen Negative † Negative Negative Positive in ~50%
IgE
Total IgE Normal Negative Normal Normal to elevated
Peripheral blood No Occasional No Present in <50%
eosinophilia
Biopsy findings
Colitis Prominent Focal No May be present
Lymph nodular No Common No Yes
hyperplasia
Eosinophils Prominent Prominent Few Prominent; also neutrophilic
infiltrates, papillary elongation,
and basal zone hyperplasia
Food challenge Vomiting in Rectal bleeding in 6- Vomiting, Vomiting and diarrhea in hours
1-4 hr; 72 hr diarrhea, or both to days
diarrhea in 5- in 40-72 hr
8 hr
Treatment Protein Protein elimination, Protein Protein elimination, good
elimination, symptoms clear in 3 elimination, response to casein hydrolysate,
80% respond days with casein symptoms clear in excellent response to elemental
to casein hydrolysate; 1-3 wk; diet; symptoms clear in 2-3 wk,
hydrolysate resume/continue rechallenge and excellent acute response to
and breastfeeding on biopsy in 1-2 yr steroids; rechallenge by
symptoms maternal antigen- introducing food at home and
clear in 3-10 restricted diet; biopsy in 1-2 yr
days; reintroduce at home
rechallenge after 9-12 mo of age
under
supervision
in 1.5-2 yr
Natural history Cow's Resolved by 9-12 mo Most cases resolve Typically a prolonged, relapsing
milk: in 2-3 yr course
60%
resolved
by 2 yr
Soy:
25%
resolved
by 2 yr
Reintroduction of Supervised At home, gradually Home, gradually Home, gradually advancing
the food food advancing from 1 oz advancing
challenge to full feedings over
2 wk
* Eosinophilic gastroenteropathies encompass esophagitis, gastritis, and gastroenterocolitis.
Table 176.6
There are no laboratory studies to help identify foods responsible for cell-
mediated reactions. Consequently, elimination diets followed by oral food
challenges are the only way to establish the diagnosis. Allergists experienced in
dealing with food allergic reactions and able to treat anaphylaxis should perform
food challenges. Before a food challenge is initiated, the suspected food should
be eliminated from the diet for 10-14 days for IgE-mediated food allergy and up
to 8 wk for some cell-mediated disorders, such as EoE. Some children with cell-
mediated reactions to cow's milk do not tolerate hydrolysate formulas and must
receive amino acid–derived formulas. If symptoms remain unchanged despite
appropriate elimination diets, it is unlikely that food allergy is responsible for the
child's disorder.
Treatment
Appropriate identification and elimination of foods responsible for food
hypersensitivity reactions are the only validated treatments for food allergies.
Complete elimination of common foods (milk, egg, soy, wheat, rice, chicken,
fish, peanut, nuts) is very difficult because of their widespread use in a variety of
processed foods. The lay organization Food Allergy Research and Education
(FARE , www.foodallergy.org ) provides excellent information to help parents
deal with both the practical and emotional issues surrounding these diets.
Validated educational materials are also available through the Consortium of
Food Allergy Research (www.cofargroup.org ).
Children with asthma and IgE-mediated food allergy, peanut or nut allergy, or
a history of a previous severe reaction should be given self-injectable
epinephrine and a written emergency plan in case of accidental ingestion (see
Chapter 174 ). Because many food allergies are outgrown, children should be
reevaluated periodically by an allergist to determine whether they have lost their
clinical reactivity. A number of clinical trials are evaluating the efficacy of oral,
sublingual, and epicutaneous (patch) immunotherapy for the treatment of IgE-
mediated food allergies (milk, egg, peanut). Combining oral immunotherapy
with anti-IgE treatment (omalizumab) may improve safety compared to oral
immunotherapy alone. Furthermore, extensively heated milk or egg in baked
products are tolerated by the majority of milk and egg–allergic children. Regular
ingestion of baked products with milk and egg appears to accelerate resolution of
milk and egg allergy. Table 176.7 provides vaccination recommendations for
egg-allergic children who require immunization.
Table 176.7
ACIP and AAP Recommendations for Administering Vaccines to Patients With Egg
Allergy
VACCINE ACIP, CDC, 2016 AAP, 2016
MMR/MMRV May be used May be used
Influenza Receive with no special precautions* Receive with no special precautions*
Rabies Use caution No specific recommendation
Yellow fever Contraindicated, but desensitization protocols Contraindicated, but desensitization protocols
may be followed to administer vaccine if may be followed to administer vaccine if
necessary (citing PI) necessary (citing PI)
*
In 2016, recommendations changed to suggest all children with any severity of egg allergy
receive the injectable influenza vaccine as appropriate for age in a medical setting without any
special testing and with the same precautions as those suggested for other vaccinations,
including a 15 minute observation period and being in a setting where personnel and equipment
are available to recognize and treat allergic reactions and anaphylaxis.
ACIP, Advisory Committee on Immunization Practices, Centers for Disease Control and
Prevention; AAP, American Academy of Pediatrics; PI, product insert.
From Boyce JA, Assa'ad A, Burks AW, et al: Guideline for the diagnosis and management of food
allergy in the United States: report of the NIAID-sponsored expert panel, J Allergy Clin Immunol
126(6):S1–S58, 2010 (Table V, p S31).
Prevention
It was once thought that avoidance of allergenic foods and delayed introduction
to the diet would prevent allergy, but the opposite is probably true; delayed
introduction of these foods may increase the risk of allergy, especially in children
with atopic dermatitis. A trial of early introduction of dietary peanut randomized
640 infants age 4-11 mo with severe eczema, egg allergy, or both to consume or
avoid peanut until age 60 mo. The early introduction of peanut dramatically
decreased the development of peanut allergy among children at high risk for this
allergy. A theory behind this approach is that early oral introduction of peanut
induces oral tolerance that precedes the potential sensitization to peanut via the
disrupted skin barrier. Infants with early-onset atopic disease (e.g., severe
eczema) or egg allergy in the 1st 4 to 6 mo of life might benefit from evaluation
by an allergist or physician trained in management of allergic diseases to
diagnose any food allergy and assist in implementing appropriate early peanut
introduction. The clinician can perform an observed peanut challenge for those
with evidence of a positive peanut skin test response or serum peanut-specific
IgE >0.35 kUA /L to determine whether they are clinically reactive before
initiating at-home introduction of infant-safe forms of peanut. Additional details
for the early introduction of peanut are available from the National Institute of
Allergy and Infectious Diseases (NIAID).*
There is no compelling evidence to support the practice of restricting the
maternal diet during pregnancy or while breastfeeding, or of delaying
introduction of various allergenic foods to infants from atopic families (Table
176.8 ). Exclusive breastfeeding for the 1st 4-6 mo of life may reduce allergic
disorders in the 1st few yr of life in infants at high risk for development of
allergic disease. Potentially allergenic foods (eggs, milk, wheat, soy, peanut/tree
nut products, fish) should be introduced after this period of exclusive
breastfeeding and may prevent the development of allergies later in life. Use of
hydrolyzed formulas may be beneficial if breastfeeding cannot be continued for
4-6 mo or after weaning, especially to prevent eczema in high-risk families, but
this approach remains controversial. Probiotic supplements may also reduce the
incidence and severity of eczema. Because some skin preparations contain
peanut oil, which may sensitize young infants, especially those with cutaneous
inflammation, such preparations should be avoided. Since inflamed/disrupted
skin barrier is a risk factor for food allergy, trials are underway to enhance the
skin barrier from birth, using emollients and decreasing bathing frequency, to
reduce the incidence of atopic dermatitis in high-risk neonates.
Table 176.8
Prevention of Food Allergy
Breastfeed exclusively for 4-6 mo.
Introduce solid (complementary) foods after 4-6 mo of exclusive
breastfeeding.
Introduce low-risk complementary foods 1 at a time.
Introduce potentially highly allergenic foods (fish, eggs, peanut, milk,
wheat) soon after the lower-risk foods (no need to avoid or delay).
Infants with early-onset atopic disease (e.g., severe eczema) or
egg allergy in the 1st 4-6 mo of life.
Do not avoid allergenic foods during pregnancy or nursing.
Soy-based formulas do not prevent allergic disease.
Bibliography
U.S. Food and Drug Administration (FDA) MedWatch program
(http://www.fda.gov/medwatch/index.html ) likely suffer from underreporting.
Cutaneous reactions are the most common form of ADRs, with ampicillin,
amoxicillin, penicillin, and trimethoprim/sulfamethoxazole (TMP/SMX) being
the most frequently implicated drugs (Tables 177.1 and 177.2 ). Although the
majority of ADRs do not appear to be allergic in nature, 6–10% can be attributed
to an allergic or immunologic mechanism. Importantly, given the high
probability of recurrence of allergic reactions, these reactions should be
preventable, and information technology–based interventions may be especially
useful to reduce risk of reexposure.
Table 177.1
Heterogeneity of Drug-Induced Allergic Reactions
ORGAN-
EXAMPLES OF CAUSATIVE
SPECIFIC CLINICAL FEATURES
AGENTS
REACTIONS
CUTANEOUS
Exanthems Diffuse fine macules and papules evolve over Allopurinol, aminopenicillins,
days after drug initiation cephalosporins, antiepileptic agents, and
Delayed-type hypersensitivity antibacterial sulfonamides
Urticaria, Onset within minutes of drug initiation IgE mediated: β-lactam antibiotics
angioedema Potential for anaphylaxis Bradykinin mediated: ACEI
Often IgE mediated
Fixed drug Hyperpigmented plaques Tetracycline, sulfonamides, NSAIDs, and
eruption Recur at same skin or mucosal site carbamazepine
Pustules Acneiform Acneiform: corticosteroids, sirolimus
Acute generalized exanthematous pustulosis AGEP: antibiotics, calcium-channel
(AGEP) blockers
Bullous Tense blisters Furosemide, vancomycin
Flaccid blisters Captopril, penicillamine
SJS Fever, erosive stomatitis, ocular involvement, Antibacterial sulfonamides,
purpuric macules on face and trunk with <10% anticonvulsants, oxicam NSAIDs, and
epidermal detachment allopurinol
TEN Similar features as SJS but >30% epidermal Same as SJS
detachment
Mortality as high as 50%
Cutaneous Erythematous/scaly plaques in photodistribution Hydrochlorothiazide, calcium channel
lupus blockers, ACEIs
Hematologic Hemolytic anemia, thrombocytopenia, Penicillin, quinine, sulfonamides
granulocytopenia
Hepatic Hepatitis, cholestatic jaundice Paraaminosalicylic acid, sulfonamides,
phenothiazines
Pulmonary Pneumonitis, fibrosis Nitrofurantoin, bleomycin, methotrexate
Renal Interstitial nephritis, membranous Penicillin, sulfonamides, gold,
glomerulonephritis penicillamine, allopurinol
MULTIORGAN REACTIONS
Anaphylaxis Urticaria/angioedema, bronchospasm, β-Lactam antibiotics, monoclonal
gastrointestinal symptoms, hypotension antibodies
IgE- and non–IgE-dependent reactions
DRESS Cutaneous eruption, fever, eosinophilia, hepatic Anticonvulsants, sulfonamides,
dysfunction, lymphadenopathy minocycline, allopurinol
Serum sickness Urticaria, arthralgias, fever Heterologous antibodies, infliximab
Systemic lupus Arthralgias, myalgias, fever, malaise Hydralazine, procainamide, isoniazid
erythematosus
Vasculitis Cutaneous or visceral vasculitis Hydralazine, penicillamine,
propylthiouracil
ACEI, Angiotensin-converting enzyme inhibitor; DRESS, drug rash with eosinophilia and systemic
symptoms; NSAID, nonsteroidal antiinflammatory drug; SJS, Stevens-Johnson syndrome; TEN,
toxic epidermal necrolysis.
From Khan DA, Solensky R: Drug allergy, J Allergy Clin Immunol 125:S126–S137, 2010 (Table 1,
p S127).
Table 177.2
MACULOPAPULAR EXANTHEMS—ANY DRUG CAN PRODUCE A RASH 7-10 DAYS AFTER THE
FIRST DOSE
Allopurinol
Antibiotics: penicillin, sulfonamides
Antiepileptics: phenytoin, phenobarbital
Antihypertensives: captopril, thiazide diuretics
Contrast dye: iodine
Gold salts
Hypoglycemic drugs
Meprobamate
Phenothiazines
Quinine
DRUG RASH WITH EOSINOPHILIA AND SYSTEMIC SYMPTOMS (DRESS)
Anticonvulsants: phenytoin, phenobarbital, valproate, lamotrigine
Antibiotics: sulfonamides, minocycline, dapsone, ampicillin, ethambutol, isoniazid, linezolid, metronidazole,
rifampin, streptomycin, vancomycin
Antihypertensives: amlodipine, captopril
Antidepressants: bupropion, fluoxetine
Allopurinol
Celecoxib
Ibuprofen
Phenothiazines
ERYTHEMA MULTIFORME/STEVENS-JOHNSON SYNDROME
Sulfonamides, phenytoin, barbiturates, carbamazepine, allopurinol, amikacin, phenothiazines
Toxic epidermal necrolysis: same as for erythema multiforme but also acetazolamide, gold, nitrofurantoin,
pentazocine, tetracycline, quinidine
ACUTE GENERALIZED EXANTHEMIC PUSTULOSIS
Antibiotics: penicillins, macrolides, cephalosporins, clindamycin, imipenem, fluoroquinolones, isoniazid,
vancomycin, minocycline, doxycycline, linezolid
Antimalarials: chloroquine, hydroxychloroquine
Antifungals: terbinafine, nystatin
Anticonvulsants: carbamazepine
Calcium-channel blockers
Furosemide
Systemic corticosteroids
Protease inhibitors
COLLAGEN VASCULAR OR LUPUS-LIKE REACTIONS
Procainamide, hydralazine, phenytoin, penicillamine, trimethadione, methyldopa, carbamazepine, griseofulvin,
nalidixic acid, oral contraceptives, propranolol
ERYTHEMA NODOSUM
Oral contraceptives, penicillin, sulfonamides, diuretics, gold, clonidine, propranolol, opiates
FIXED DRUG REACTIONS
Phenolphthalein, barbiturates, gold, sulfonamides, meprobamate, penicillin, tetracycline, analgesics
See Chapter 664 and Table 664.3 .
From Duvic M: Urticaria, drug hypersensitivity rashes, nodules and tumors, and
atrophic diseases. In Goldman L, Schafer AI, editors: Goldman-Cecil medicine,
ed 25, Philadelphia, 2016, Elsevier, Table 440.3.
Table 177.3
Groups of β-Lactam Antibiotics That Share Identical R1-
Group Side Chains*
Amoxicillin Ampicillin Ceftriaxone Cefoxitin Cefamandole Ceftazidime
Cefadroxil Cefaclor Cefotaxime Cephaloridine Cefonicid Aztreonam
Cefprozil Cephalexin Cefpodoxime Cephalothin
Cefatrizine Cephradine Cefditoren
Cephaloglycin Ceftizoxime
Loracarbef Cefmenoxime
* Each column represents a group with identical R1 side chains.
From Solensky R, Khan DA: Drug allergy: an updated practice parameter, Ann Allergy Asthma
Immunol 105:273e1–273e78, 2010 (Table 16, p 273e49).
Table 178.1
Table 178.2
Signs Suggestive of Rheumatic Disease
Mouth ulcers are part of the diagnostic criteria for SLE and Behçet disease
Laboratory Testing
There are no specific screening tests for rheumatologic disease. Once a
differential diagnosis is determined, appropriate testing can be performed (Tables
178.3 and 178.4 ). Initial studies are generally performed in standard local
laboratories. Screening for specific autoantibodies can be performed in
commercial laboratories, but confirmation of results in a tertiary care center
immunology laboratory is often necessary.
Table 178.3
Autoantibody Specificity and Disease Associations
PREVALENCE
ANTIBODY DISEASE SPECIFICITY
(%)
Antinuclear antibody SLE, juvenile — Associated with increased risk of uveitis
(ANA) rheumatoid arthritis, in JIA and psoriatic arthritis
dermatomyositis, Up to 30% of children testing positive for
scleroderma, psoriatic ANAs have no underlying rheumatic
arthritis, MCTD disease
Double-stranded SLE 60-70 High specificity for SLE; associated with
DNA (dsDNA) lupus nephritis
Smith (Sm) SLE 20-30 Highly specific for SLE; associated with lupus
nephritis
Smooth muscle (Sm) Autoimmune hepatitis — —
Pm-Scl Sclerodermatomyositis — —
(polymyositis-
scleroderma)
SSA (Ro) SLE, Sjögren 25-30 Associated with neonatal lupus syndrome,
syndrome subacute cutaneous lupus, thrombocytopenia
SSB (La) SLE, Sjögren 25-30 Usually coexists with anti-SSA antibody
syndrome
Ribonuclease protein MCTD, SLE 30-40 Suggestive of MCTD unless meets criteria for
(RNP) SLE
Histone Drug-induced lupus, — —
SLE
Centromere Limited cutaneous 70 Nonspecific for systemic sclerosis
systemic sclerosis
Topoisomerase I (Scl- Systemic sclerosis — Rare in childhood
70)
Antineutrophil Vasculitis — —
cytoplasmic
antibodies (ANCAs)
Cytoplasmic — cANCAs associated with granulomatosis with
(cANCAs)/PR3- polyangiitis (Wegener), cystic fibrosis
ANCA
Perinuclear — pANCAs associated with microscopic
(pANCAs)/MPO- polyangiitis, polyarteritis nodosa, SLE,
ANCA inflammatory bowel disease, cystic fibrosis,
primary sclerosing cholangitis, Henoch-
Schönlein purpura, Kawasaki disease, Churg-
Strauss syndrome
Anticitrullinated RF-positive JIA 50-90 Specific for JIA (RF+), may be positive before
protein (ACPA); also RF
called anti–cyclic
citrullinated protein
(anti-CCP)
MCTD, Mixed connective tissue disease; MPO-ANCA, antimyeloperoxidase; PR3-ANCA,
antiproteinase 3; RF, rheumatoid factor; SLE, systemic lupus erythematosus.
Adapted from Aggerwal A: Clinical application of tests used in rheumatology, Indian J Pediatr
69:889–892, 2002.
Table 178.4
Evaluation Based on Suspected Diagnosis of Rheumatic
Disease
SUSPECTED
FURTHER SUBSPECIALTY
RHEUMATIC INITIAL EVALUATION
EVALUATION EVALUATION
DISEASE(S)
Systemic CBC, ESR, ANA, ALT, AST, If ANA test result is Antiphospholipid Abs, lupus
lupus CPK, creatinine, albumin, total positive: anti-SSA (Ro), anticoagulant, anti–β2 -
erythematosus protein, urinalysis, BP, thyroid anti-SSB (La), anti-Smith, glycoprotein,
(SLE) profile and anti-RNP Abs; anti- echocardiogram; consider
Mixed dsDNA Ab, C3, C4, renal biopsy, PFTs,
connective Coombs, spot urine bronchoscopy with lavage,
tissue disease protein/creatinine ratio, HRCT of chest; consider
(MCTD) CXR lung biopsy
Juvenile CBC, CPK, ALT, AST, LDH, Consider MRI of muscle Consider electromyography
dermatomyositis aldolase, ANA; check gag reflex and possible muscle biopsy,
(JDM) PFTs, swallowing study,
serum neopterin
Juvenile idiopathic CBC, ESR, creatinine, ALT, Consider Ab titers to MRI
arthritis (JIA) AST, consider anti–streptolysin unusual infectious agents,
O/anti–DNAase B for purified protein derivative,
streptococcus-induced arthritis, RF, ANA, HLA-B27, anti-
Epstein-Barr virus titers, Lyme CCP
titer, parvovirus B19 titer, plain
radiograph of joints
Granulomatosis CBC, ANCA, AST, ALT, Spot urine Bronchoscopy with lavage,
with polyangiitis albumin, creatinine, ESR, protein/creatinine ratio, HRCT chest; consider lung
(Wegener urinalysis, CXR, BP anti–myeloperoxidase and and kidney biopsies
granulomatosis) anti–proteinase-3 Abs,
PFTs
Sarcoidosis CBC, electrolytes, AST, ALT, CXR, PFTs Consider testing for Blau
albumin, creatinine, calcium, syndrome in infants (see
phosphorous, ACE, BP Chapter 184 ); HRCT of
chest; consider renal and lung
biopsy
Localized Skin biopsy, CBC, ESR Serum IgG, ANA, RF,
scleroderma single-stranded DNA Ab,
antihistone Ab, CPK
Systemic ANA, CBC, ESR, BP, AST, Anti-Scl70, PFTs HRCT of chest,
scleroderma ALT, CPK, creatinine, CXR echocardiogram, upper GI
radiography series
Ab, Antibody; ACE, angiotensin-converting enzyme (normally elevated in childhood; interpret with
caution); ALT, alanine transaminase; ANA, antinuclear antibody; anti-dsDNA Ab, anti–double-
stranded DNA antibody; AST, aspartate transaminase; BP, blood pressure; CBCD, complete blood
count with differential; CCP, cyclic citrullinated protein; CPK, creatine phosphokinase; CXR, chest
radiograph; ESR, erythrocyte sedimentation rate; GI, gastrointestinal; HRCT, high-resolution CT;
LDH, lactate dehydrogenase; PFTs, pulmonary function tests; RF, rheumatoid factor; RNP,
ribonucleoprotein.
One essential laboratory test for rheumatic disease assessment is the complete
blood count (CBC), since it yields many diagnostic clues. Elevated WBC count
is compatible with malignancy, infection, systemic JIA, and vasculitis.
Leukopenia can be postinfectious, especially viral, or caused by SLE or
malignancy. Lymphopenia is more specific for SLE than is leukopenia. Platelets
are acute-phase reactants and are therefore elevated with inflammatory markers.
Exceptions are a bone marrow–occupying malignancy, such as leukemia or
neuroblastoma, SLE, and early Kawasaki disease. Anemia is nonspecific and
may be caused by any chronic illness, but hemolytic anemia (positive Coombs
test result) may point to SLE or MCTD. Rheumatoid factor (RF) is present in
<10% of children with JIA and thus has poor sensitivity as a diagnostic tool; RF
may be elevated by infections such as endocarditis, tuberculosis, syphilis, and
viruses (parvovirus B19, hepatitides B and C, mycoplasma), as well as primary
biliary cirrhosis and malignancies. In a child with chronic arthritis, RF serves as
a prognostic indicator.
Inflammatory markers (erythrocyte sedimentation rate, C-reactive protein) are
nonspecific and are elevated in infections and malignancies as well as rheumatic
diseases (Tables 178.5 and 178.6 ). Their levels may also be normal in rheumatic
diseases such as arthritis, scleroderma, and dermatomyositis. Inflammatory
marker measurements are more useful in rheumatic diseases for following
response to treatment than as diagnostic tests. Muscle enzymes include aspartate
transaminase (AST), alanine transaminase (ALT), creatinine phosphokinase
(CPK), aldolase, and lactate dehydrogenase (LDH), any of which may be
elevated in JDM as well as in other diseases causing muscle breakdown. Muscle-
building supplements, medications, and extreme physical activity may also cause
muscle breakdown and enzyme elevations. AST, ALT, and aldolase are also
elevated secondary to liver disease, and a γ-glutamyltransferase (GGT)
measurement may help differentiate whether the source is muscle or liver.
Table 178.5
Table 178.6
Conditions Associated With Elevated C-
Reactive Protein Levels
Normal Or Minor Elevation (<1 mg/dL)
1. Vigorous exercise
2. Common cold
3. Pregnancy
4. Gingivitis
5. Seizures
6. Depression
7. Insulin resistance and diabetes
8. Several genetic polymorphisms
9. Obesity
1. Myocardial infarction
2. Malignancies
3. Pancreatitis
4. Mucosal infection (bronchitis, cystitis)
5. Most systemic autoimmune diseases
6. Rheumatoid arthritis
From Firestein GS, Budd RC, Gabriel SE, et al, editors: Kelley & Firestein's
textbook of rheumatology, ed 10, Philadelphia, 2017, Elsevier (Table 57-4, p
849).
Hematopoietic Changes
Metabolic Changes
Hepatic Changes
Table 179.1
Multidisciplinary Treatment of Rheumatic Diseases in
Childhood
Accurate diagnosis and Pediatric rheumatologist
education of family Pediatrician
Nurse:
• Disease-related education
• Medication administration (injection teaching)
• Safety monitoring
Social worker:
• Facilitation of school services
• Resource identification (community, government, financial, advocacy groups,
vocational rehabilitation)
Physical medicine and Physical therapy:
rehabilitation • Addressing deficits in joint or muscle mobility, limb length discrepancies, gait
abnormalities, and weakness
Occupational therapy:
• Splinting to reduce joint contractures/deformities and lessen stress on joints;
adaptive devices for activities of daily living
Consultant team Ophthalmology:
• Eye screening for uveitis (see Table 180.4 )
• Screening for medication-related ocular toxicity (hydroxychloroquine,
glucocorticoids)
Nephrology
Orthopedics
Dermatology
Gastroenterology
Physical and psychosocial Nutrition:
growth and development • Addressing undernourishment from systemic illness and
obesity/overnourishment from glucocorticoids
School integration:
• Individualized educational plan (IEP) or 504 plan
Peer-group relationships
Individual and family counseling
Coordination of care Involvement of patient and family as active team members
Communication among healthcare providers
Involvement of school (school nurse) and community (social worker) resources
Therapeutics
A key principle of pharmacologic management of rheumatic diseases is that
early disease control, striving for induction of remission, leads to less tissue and
organ damage with improved short- and long-term outcomes. Medications are
chosen from broad therapeutic classes on the basis of diagnosis, disease severity,
anthropometrics, and adverse effect profile. Many drug therapies used do not
have U.S. Food and Drug Administration (FDA) indications for pediatric
rheumatic diseases given the relative rarity of these conditions. The evidence
base may be limited to case series, uncontrolled studies, or extrapolation from
use in adults. The exception is JIA, for which there is a growing body of
randomized controlled trial (RCT) evidence, particularly for newer therapeutics.
Therapeutic agents used for treatment of childhood rheumatic diseases have
various mechanisms of action, but all suppress inflammation (Table 179.2 ).
Both biologic and nonbiologic disease-modifying antirheumatic drugs
(DMARDs ) directly affect the immune system. DMARDs should be prescribed
by specialists. Live vaccines are contraindicated in patients taking
immunosuppressive glucocorticoids or DMARDs. A negative test result for
tuberculosis (purified protein derivative and/or QuantiFERON-TB Gold) should
be verified and the patient's immunization status updated, if possible, before
such treatment is initiated. Killed vaccines are not contraindicated, and annual
injectable influenza vaccine is recommended.
Table 179.2
Therapeutics for Childhood Rheumatic Diseases*
ADVERSE
CLASSIFICATION THERAPEUTIC † DOSE INDICATION † REACTIONS
Nonsteroidal Etodolac a PO once-daily JIA GI intolerance
antiinflammatory dose: Spondyloarthropathy (abdominal pain,
drugs (NSAIDs) ‡ 20-30 kg: 400 Pain nausea), gastritis,
mg Serositis hepatitis, tinnitus,
31-45 kg: 600 Cutaneous vasculitis anemia,
mg Uveitis pseudoporphyria,
46-60 kg: 800 aseptic meningitis,
mg headache, renal
>60 kg: 1,000 disease
mg
Ibuprofen a 40 mg/kg/day
PO in 3 divided
doses
Max: 2400
mg/day
Naproxen a 15 mg/kg/day
PO in 2 divided
doses
Max 1,000
mg/day
Celecoxib a 10-25 kg: 50
mg PO bid
>25 kg: 100 mg
PO bid
Meloxicam a 0.125 mg/kg
PO qd
Max 7.5 mg
Disease-modifying Methotrexate a 10-20 mg/m2 JIA GI intolerance
antirheumatic drugs /wk (0.35-0.65 Uveitis (nausea,
(DMARDs) mg/kg/wk) PO vomiting),
20-30 mg/m2 hepatitis,
/wk (0.65-1 myelosuppression,
mg/kg/wk) SC; mucositis,
higher doses teratogenesis,
better absorbed lymphoma,
by SC injection interstitial
pneumonitis
Leflunomide PO once daily: JIA Hepatitis, hepatic
10 to <20 kg: necrosis,
10 mg cytopenias,
20-40 kg: 15 mucositis,
mg teratogenesis,
>40 kg: 20 mg peripheral
neuropathy
Hydroxychloroquine 5 mg/kg PO qd; SLE Retinal toxicity,
do not exceed 5 JDMS GI intolerance,
mg/kg/daily Antiphospholipid rash, skin
Max 400 mg antibody syndrome discoloration,
daily anemia,
cytopenias,
myopathy, CNS
stimulation, death
(overdose)
Sulfasalazine a 30-50 Spondyloarthropathy, GI intolerance,
mg/kg/day in 2 JIA rash,
divided doses hypersensitivity
Adult max 3 reactions, Stevens-
g/day Johnson
syndrome,
cytopenias,
hepatitis,
headache
Tumor necrosis Adalimumab a SC once every JIA Injection site
factor (TNF)-α other wk: Spondyloarthropathy reaction, infection,
antagonists 10 to <15 kg: Psoriatic arthritis rash, cytopenias,
10 mg Uveitis lupus-like
15 to <30 kg: syndrome,
20 mg potential increased
≥30 kg: 40 mg malignancy risk
Etanercept a 0.8 mg/kg SC once JIA Injection site
weekly (max 50 reactions,
mg/dose) or 0.4 infections, rash,
mg/kg SC twice demyelinating
weekly (max 25 disorders,
mg/dose) cytopenias,
potential increased
malignancy risk
Infliximab 5-10 mg/kg IV JIA Infusion reactions,
every 4-8 wk Spondyloarthropathy hepatitis, potential
Uveitis increased
Sarcoidosis malignancy risk
Modulate T-cell Abatacept a IV every 2 wk JIA Infection,
activation ×3 doses, then headache,
monthly for ≥6 potential increased
yr of age: malignancy risk
<75 kg: 10
mg/kg
75-100 kg: 750
mg
>100 kg: 1,000
mg
SC once
weekly:
10 to <25 kg:
50 mg
≥25 to <50 kg:
87.5 mg
≥50 kg: 125 mg
Anti-CD20 (B-cell) Rituximab 575 mg/m2 , max SLE Infusion reactions,
antibody 1,000 mg, IV on lymphopenia,
days 1 and 15 reactivation
hepatitis B, rash,
serum sickness,
arthritis, PML
Anti-BLyS antibody Belimumab e 10 mg/kg IV every 2 SLE Infusion reactions,
wk ×3 doses, then infection,
every 4 wk depression
Interleukin (IL)-1 Anakinra 1-2 mg/kg/daily Systemic JIA Injection site
antagonist Adult max 100 CAPS reactions,
mg infection
Canakinumab b Given SC every CAPS Injection site
8 wk (CAPS) Systemic JIA reaction, infection,
every 4 wk diarrhea, nausea,
(systemic JIA): vertigo, headache
15-40 kg: 2
mg/kg (up to 3
mg/kg if
needed)
>40 kg: 150 mg
IV: Polyarticular JIA
<30 kg: 10
mg/kg/dose
every 4 wk
≥30 kg: 8
mg/kg/dose
every 4 wk;
maximum dose:
800 mg/dose
SC:
<30 kg: 162
mg/dose once
every 3 wk
≥30 kg: 162
mg/dose once
every 2 wk
IL-6 antagonist Tocilizumab a ≥2 yr and ≥30 Systemic JIA Infusion reactions,
kg: 8 elevated LFTs,
mg/kg/dose elevated lipids,
every 2 wk thrombocytopenia,
≥2 yr and ≤30 infections
kg: 12
mg/kg/dose
every 2 wk
Intravenous immune IVIG c 1,000-2,000 Kawasaki disease Infusion reaction,
globulin mg/kg IV JDMS aseptic meningitis,
infusion SLE renal failure
For JDMS, give
monthly
Cytotoxic Cyclophosphamide 0.5-1 g/m2 IV SLE Nausea, vomiting,
(max 1.5 g) Vasculitis myelosuppression,
monthly for 6 JDMS mucositis,
mo induction, Pulmonary hyponatremia,
then every 2-3 hemorrhage alopecia,
mo hemorrhagic
Oral regimen: cystitis, gonadal
1-2 failure,
mg/kg/daily; teratogenesis,
max 150 secondary
mg/daily malignancy
Immunosuppressive Mycophenolate Oral SLE GI intolerance
mofetil suspension: Uveitis (diarrhea, nausea,
max 1,200 vomiting), renal
mg/m2 /day PO impairment,
(up to 2 g/day) neutropenia,
divided bid teratogenesis,
Capsules: max secondary
1,500 mg/day malignancy, PML
PO for BSA
1.25-1.5 m2 , 2
g/day PO for
BSA >1.5 m2
divided bid
Glucocorticoids Prednisone a , d - f 0.05-2 SLE Cushing
mg/kg/day PO JDMS syndrome,
given in 1-4 Vasculitis osteoporosis,
divided doses; JIA increased appetite,
max varies by Uveitis weight gain,
individual (80 Sarcoidosis striae,
mg/daily) hypertension,
Adverse effects adrenal
are dose suppression,
dependent; hyperglycemia,
lowest effective infection,
dose should be avascular necrosis
used
Methylprednisolone 0.5-1.7 SLE
a , d - g mg/kg/day or 5- JDMS
2
25 mg/m /day Vasculitis
IM/IV in Sarcoidosis
divided doses Localized
every 6-12 hr scleroderma
For severe
manifestations:
30 mg/kg/dose
(max 1 g) daily
for 1-5 days
Intraarticular Dose varies by joint JIA Subcutaneous
and formulation atrophy, skin
hypopigmentation,
calcification,
infection
Prednisolone 1-2 drops into Uveitis Ocular
ophthalmic eye up to every hypertension,
suspension hr while awake glaucoma, nerve
Needs damage, cataract,
monitoring by infection
ophthalmologist
* Consult a clinical pharmacology reference for current dosing and monitoring guidelines, and
qd, Once daily; bid, twice daily; Blys, B-lymphocyte stimulator; BMP, basic metabolic panel; BSA,
body surface area; BUN, blood urea nitrogen; CAPS, cryopyrin-associated periodic syndrome;
CBC, complete blood count; CNS, central nervous system; dsDNA, double-stranded DNA; GI,
gastrointestinal; IM, intramuscular(ly); IV, intravenous(ly); IVIG, intravenous immune globulin;
JDMS, juvenile dermatomyositis; JIA, juvenile idiopathic arthritis; LFTs, liver function tests; PML,
progressive multifocal leukoencephalopathy; PO, by mouth; SC, subcutaneous(ly); SLE, systemic
lupus erythematosus; TB, tuberculosis.
Biologic Agents
Biologic agents are proteins that have been engineered to target and modulate
specific components of the immune system, with the goal of decreasing the
inflammatory response. Antibodies have been developed to target specific
cytokines such as IL-1 and IL-6 or to interfere with specific immune cell
function through depletion of B cells or suppression of T-cell activation (Table
179.3 ). The availability of biologic agents has dramatically increased the
therapeutic options for treating rheumatic disease recalcitrant to nonbiologic
therapies, and in some cases biologics are becoming first-line interventions. A
primary concern is the increased risk of malignancy when biologics are
combined with other immunosuppressants.
Table 179.3
Adapted from Cassidy JT, Levison JE, Bass JC, et al: A study of classification
criteria for a diagnosis of juvenile rheumatoid arthritis, Arthritis Rheum 29:174–
181, 1986.
Table 180.2
International League of Associations for Rheumatology
Classification of Juvenile Idiopathic Arthritis (JIA)
‡ Dactylitis is swelling of ≥1 digit(s), usually in an asymmetric distribution, that extends beyond the
joint margin.
§ A minimum of 2 pits on any 1 or more nails at any time.
ǁ
Enthesitis is defined as tenderness at the insertion of a tendon, ligament, joint capsule, or fascia
to bone.
¶ Inflammatory lumbosacral pain refers to lumbosacral pain at rest with morning stiffness that
improves on movement.
IBD, Inflammatory bowel disease; RF, rheumatoid factor.
From Firestein GS, Budd RC, Harris ED Jr, et al, editors: Kelley's textbook of rheumatology, ed 8,
Philadelphia, 2009, Saunders.
Table 180.3
Characteristics of ACR and ILAR Classifications of
Childhood Chronic Arthritis
Table 180.4
Overview of Main Features of Subtypes of Juvenile
Idiopathic Arthritis (JIA)
PEAK
% of
ILAR AGE at FEMALE:MALE ARTHRITIS EXTRAARTICULAR LABORATORY
ALL JIA
SUBTYPE ONSET RATIO PATTERN FEATURES INVESTIGATIONS
CASES
(yr)
Systemic 1-5 1 : 1 5-15 Polyarticular, often Daily fever; evanescent Anemia; WBC ↑↑;
arthritis affecting knees, rash; pericarditis; ESR ↑↑; CRP ↑↑;
wrists, and ankles; pleuritis ferritin ↑; platelets ↑↑
also fingers, neck, (normal or ↓ in
and hips MAS)
Oligoarthritis 2-4 3 : 1 40-50 Knees ++; ankles, Uveitis in 30% of cases ANA positive in
(but fingers + 60%; other test
ethnic results usually
variation) normal; may have
mildly ↑ ESR/CRP
Polyarthritis:
RF negative 2-4 and 3 : 1 and 10 : 1 20-35 Symmetric or Uveitis in 10% ANA positive in
10-14 asymmetric; small 40%; RF negative;
and large joints; ESR ↑ or ↑↑; CRP ↑
cervical spine; or normal; mild
temporomandibular anemia
joint
RF positive 9-12 9 : 1 <10 Aggressive Rheumatoid nodules in RF positive; ESR ↑↑;
symmetric 10%; low-grade fever CRP ↑/normal; mild
polyarthritis anemia
Epidemiology
The worldwide incidence of JIA ranges from 0.8-22.6 per 100,000 children per
year, with prevalence ranges from 7-401 per 100,000. These wide-ranging
numbers reflect population differences, particularly environmental exposure and
immunogenetic susceptibility, along with variations in diagnostic criteria,
difficulty in case ascertainment, and lack of population-based data. An estimated
300,000 U.S. children have arthritis, including 100,000 with a form of JIA.
Oligoarthritis is the most common subtype (40–50%), followed by
polyarthritis (25–30%) and systemic JIA (5–15%) (see Table 180.4 ). There is
no sex predominance in systemic JIA (sJIA ), but more girls than boys are
affected in both oligoarticular (3 : 1) and polyarticular (5 : 1) JIA. The peak age
at onset is 2-4 yr for oligoarticular disease. Age of onset has a bimodal
distribution in polyarthritis, with peaks at 2-4 yr and 10-14 yr. sJIA occurs
throughout childhood, with a peak at 1-5 yr.
FIG. 180.4 Oligoarticular juvenile idiopathic arthritis with swelling and
flexion contracture of the right knee.
Table 180.5
Frequency of Ophthalmologic Examination in Patients With
Juvenile Idiopathic Arthritis
Referral
• Patients should be referred at time of diagnosis, or suspicion, of JIA
Initial screening examination
• Should occur as soon as possible and no later than 6 wk from referral
• Symptomatic ocular patients should be seen within a week of referral
Ongoing screening
• Screening at two monthly intervals from onset of arthritis for 6 mo
• Followed by 3-4 monthly screening for time outlined below
Oligoarticular JIA, psoriatic arthritis, and enthesitis-related arthritis irrespective of ANA status, onset under 11 yr
AGE AT ONSET (YR) LENGTH OF SCREENING (YR)
<3 8
3-4 6
5-8 3
9-10 1
Polyarticular, ANA-positive JIA, onset <10 yr
AGE AT ONSET (YR) LENGTH OF SCREENING (YR)
<6 5
6-9 2
• Polyarticular, ANA-negative JIA, onset <7 yr
• 5-yr screening for all children
• Systemic JIA and rheumatoid factor–positive polyarticular JIA
Uveitis risk very low; however, diagnostic uncertainty in the early stages and overlap of symptoms may mean
initial screening is indicated
• All categories, onset >11 yr
1-yr screening for all children
• After stopping immunosuppression (eg, methotrexate)
Two monthly screening for 6 mo, then revert to previous screening frequency as above
• After discharge from screening
Patients should receive advice about regular self-monitoring by checking vision uniocularly once weekly and
when to seek medical advice
Screening may need to continue indefinitely in situations where a young person may be unable to detect a change
in vision or be unwilling to seek re-referral
Annual check by optometrist as a useful adjunct
From Clarke SLN, Sen ES, Ramanan AV: Juvenile idiopathic arthritis-associated uveitis. Pediatr
Rheumatol 14:27, 2016. p. 3.
1. Cytopenias
2. Abnormal liver function tests
3. Coagulopathy (hypofibrinogenemia)
4. Decreased erythrocyte sedimentation rate
5. Hypertriglyceridemia
6. Hyponatremia
7. Hypoalbuminemia
8. Hyperferritinemia
9. Elevated sCD25 and sCD163
Clinical Features*
1. Nonremitting fever
2. Hepatomegaly
3. Splenomegaly
4. Lymphadenopathy
5. Hemorrhages
6. Central nervous system dysfunction (headache, seizures, lethargy, coma,
disorientation)
Histopathologic Features*
Diagnosis
JIA is a clinical diagnosis without any diagnostic laboratory tests. The
meticulous clinical exclusion of other diseases and many mimics is therefore
essential. Laboratory studies, including tests for ANA and RF, are only
supportive or prognostic, and their results may be normal in patients with JIA
(see Tables 180.1 , 180.3 , and 180.4 ).
Differential Diagnosis
The differential diagnosis for arthritis is broad and a careful, thorough
investigation for other underlying etiology is imperative (Table 180.7 ). History,
physical examination, laboratory tests, and radiography may help exclude other
possible causes. Arthritis can be a presenting manifestation for any of the
multisystem rheumatic diseases of childhood, including systemic lupus
erythematosus (see Chapter 183 ), juvenile dermatomyositis (see Chapter 184 ),
sarcoidosis (see Chapter 190 ), and the vasculitic syndromes (see Chapter 192 ).
In scleroderma (see Chapter 185 ), limited ROM caused by sclerotic skin
overlying a joint may be confused with sequelae from chronic inflammatory
arthritis. Acute rheumatic fever is characterized by exquisite joint pain and
tenderness, remittent fever, and migratory polyarthritis. Autoimmune hepatitis
can also be associated with an acute arthritis.
Table 180.7
Conditions Causing Arthritis or Extremity
Pain
Rheumatic and Inflammatory Diseases
Seronegative Spondyloarthropathies
Infectious Illnesses
Reactive Arthritis
Immunodeficiencies
Hypogammaglobulinemia
Immunoglobulin A deficiency
Common variable immunodeficiency disease (CVID)
Human immunodeficiency virus (HIV)
Gout
Pseudogout
Mucopolysaccharidoses
Thyroid disease (hypothyroidism, hyperthyroidism)
Hyperparathyroidism
Vitamin C deficiency (scurvy)
Hereditary connective tissue disease (Marfan syndrome, Ehlers-Danlos
syndrome)
Fabry disease
Farber disease
Amyloidosis (familial Mediterranean fever)
Trauma
Patellofemoral syndrome
Hypermobility syndromes
Osteochondritis dissecans
Avascular necrosis (including Legg-Calvé-Perthes disease)
Hypertrophic osteoarthropathy
Slipped capital femoral epiphysis
Osteolysis
Benign bone tumors (including osteoid osteoma)
Langerhans cell histiocytosis
Rickets
Neuropathic Disorders
Peripheral neuropathies
Carpal tunnel syndrome
Charcot joints
Neoplastic Disorders
Leukemia
Neuroblastoma
Lymphoma
Bone tumors (osteosarcoma, Ewing sarcoma)
Histiocytic syndromes
Synovial tumors
Hematologic Disorders
Hemophilia
Hemoglobinopathies (including sickle cell disease)
Miscellaneous Disorders
Autoinflammatory diseases
Recurrent multifocal osteomyelitis
Pigmented villonodular synovitis
Plant-thorn synovitis (foreign body arthritis)
Myositis ossificans
Eosinophilic fasciitis
Tendinitis (overuse injury)
Raynaud phenomenon
Hemophagocytic syndromes
Pain Syndromes
Fibromyalgia
Growing pains
Depression (with somatization)
Complex regional pain syndrome
Many infections are associated with arthritis, and a recent history of infectious
symptoms may help make a distinction. Viruses, including parvovirus B19,
rubella, Epstein-Barr virus, hepatitis B virus, and HIV, can induce a transient
arthritis. Arthritis may follow enteric infections (see Chapter 182 ). Lyme
disease should be considered in children with oligoarthritis living in or visiting
endemic areas (see Chapter 249 ). Although a history of tick exposure, preceding
flulike illness, and subsequent rash should be sought, these are not always
present. Monoarticular arthritis unresponsive to antiinflammatory treatment may
be the result of chronic mycobacterial or other infection, such as Kingella kingae
, and the diagnosis is established by synovial fluid analysis (PCR) or biopsy.
Acute onset of fever and a painful, erythematous, hot joint suggests septic
arthritis (see Chapter 705 ). Isolated hip pain with limited ROM suggests
suppurative arthritis, osteomyelitis (see Chapter 704 ), toxic synovitis, Legg-
Calvé-Perthes disease, slipped capital femoral epiphysis, and chondrolysis of the
hip (see Chapter 698 ).
Lower-extremity arthritis and tenderness over insertion of ligaments and
tendons, especially in a boy, suggests ERA (see Chapter 181 ). Psoriatic
arthritis can manifest as limited joint involvement in an unusual distribution
(e.g., small joints of hand and ankle) years before onset of cutaneous disease.
Inflammatory bowel disease may manifest as oligoarthritis, usually affecting
joints in the lower extremities, as well as gastrointestinal symptoms, elevations
in ESR, and microcytic anemia.
Many conditions present solely with arthralgia (i.e., joint pain). Hypermobility
may cause joint pain, especially in the lower extremities. Growing pains should
be suspected in a child age 4-12 yr complaining of leg pain in the evening with
normal investigative studies and no morning symptoms. Nocturnal pain that
awakens the child also alerts to the possibility of a malignancy. An adolescent
modifying antirheumatic drugs (DMARDs), including methotrexate, and, if no
response, TNF inhibitors.
Table 180.8
Pharmacologic Treatment of Juvenile Idiopathic Arthritis
(JIA)
TYPICAL
TYPICAL DOSES JIA SUBTYPE SIDE EFFECT(S)
MEDICATIONS
NONSTEROIDAL ANTIINFLAMMATORY DRUGS
Naproxen 15 mg/kg/day PO divided bid Polyarthritis Gastritis, renal and hepatic toxicity,
(maximum dose 500 mg bid) Systemic pseudoporphyria
Oligoarthritis
Ibuprofen 40 mg/kg/day PO divided tid Same as above Same as above
(maximum dose 800 mg tid)
Meloxicam 0.125 mg/kg PO once daily Same as above Same as above
(maximum dose 15 mg daily)
DISEASE-MODIFYING ANTIRHEUMATIC DRUGS
Methotrexate 0.5-1 mg/kg PO or SC weekly Polyarthritis Nausea, vomiting, oral ulcerations,
(maximum dose 25 mg/wk) Systemic hepatic toxicity, blood count dyscrasias,
Persistent or immunosuppression, teratogenicity
extended
oligoarthritis
Sulfasalazine Initial 12.5 mg/kg PO Polyarthritis GI upset, allergic reaction, pancytopenia,
daily; increase by 10 renal and hepatic toxicity, Stevens-
mg/kg/day Johnson syndrome
Maintenance: 40-50 mg/kg
divided bid (maximum
dose 2 g/day)
Leflunomide* 10-20 mg PO daily Polyarthritis GI upset, hepatic toxicity, allergic rash,
alopecia (reversible), teratogenicity
(needs washout with cholestyramine)
BIOLOGIC AGENTS
Anti–Tumor Necrosis Factor-α
Etanercept 0.8 mg/kg SC weekly or 0.4 Polyarthritis Immunosuppressant, concern for
mg/kg SC twice weekly Systemic malignancy, demyelinating disease,
(maximum dose 50 mg/wk) Persistent or lupus-like reaction, injection site reaction
extended
oligoarthritis
Infliximab* 3-10 mg/kg IV q4-8 wk Same as above Same as above, infusion reaction
Adalimumab 10 to <15 kg: 10 mg SC Same as above Same as above
every other week
15 to <30 kg: 20 mg SC
every other week
>30 kg: 40 mg SC every
other week
Anticytotoxic T-Lymphocyte–Associated Antigen-4 Immunoglobulin
Abatacept <75 kg: 10 mg/kg/dose IV Polyarthritis Immunosuppressant, concern for
q4wk malignancy, infusion reaction
75-100 kg: 750 mg/dose
IV q4wk
>100 kg: 1,000 mg/dose
IV q4wk
SC once weekly:
10 to <25 kg: 50 mg
≥25 to <50 kg: 87.5 mg
≥50 kg: 125 mg
Anti-CD20
Rituximab* 750 mg/m2 IV 2 wk × 2 Polyarthritis Immunosuppressant, infusion reaction,
(maximum dose 1,000 mg) progressive multifocal encephalopathy
Interleukin-1 Inhibitors
Anakinra* 1-2 mg/kg SC daily (maximum Systemic Immunosuppressant, GI upset, injection
dose 100 mg/day) site reaction
Canakinumab 15-40 kg: 2 mg/kg/dose Systemic Immunosuppressant, headache, GI upset,
SC q8wk injection site reaction
>40 kg: 150 mg SC q8wk
Rilonacept* 2.2 mg/kg/dose SC weekly Systemic Immunosuppressant, allergic reaction,
(maximum dose 160 mg) dyslipidemia, injection site reaction
Interleukin-6 Receptor Antagonist
Tocilizumab IV q2 wk: Systemic Immunosuppressant, hepatic toxicity,
<30 kg: 12 mg/kg/dose Polyarthritis dyslipidemia, cytopenias, GI upset,
q2wk infusion reaction
>30 kg: 8 mg/kg/dose
q2wk (maximum dose 800
mg)
SC: Polyarthritis
<30 kg: 162 mg/dose
q3wk
≥30 kg: 162 mg/dose
q2wk
* Not indicated by the U.S. Food and Drug Administration for use in JIA as of 2018.
bid, Twice daily; GI, gastrointestinal; IV, intravenous; PO, oral; q, every; SC, subcutaneous; tid, 3
times daily.
Table 181.1
Overlapping Characteristics of the Spondyloarthritides*
JUVENILE JUVENILE
INFLAMMATORY REACTIVE
CHARACTERISTIC ANKYLOSING PSORIATIC
BOWEL DISEASE ARTHRITIS
SPONDYLITIS ARTHRITIS
Enthesitis +++ + + ++
Axial arthritis +++ ++ ++ +
Peripheral arthritis +++ +++ +++ +++
HLA-B27 positive +++ + ++ +++
Antinuclear antibody − ++ − −
positive
Rheumatoid factor − − − −
positive
SYSTEMIC DISEASE:
Eyes + + + +
Skin − +++ + +
Mucous membranes − − + +
Gastrointestinal tract − − ++++ +++
* Frequency of characteristics: −, absent; +, <25%; ++, 25–50%; +++, 50–75%; ++++, ≥75%.
From Cassidy JT, Petty RE: Textbook of pediatric rheumatology, ed 6, Philadelphia, 2011,
Elsevier/Saunders.
Table 181.2
Etiologic Microorganisms of Reactive
Arthritis
Probable
Chlamydia trachomatis
Shigella species
Salmonella enteritidis
Salmonella typhimurium
Yersinia enterocolitica
Yersinia pseudotuberculosis
Campylobacter jejuni and coli
Possible
Neisseria gonorrhoeae
Mycoplasma fermentans
Mycoplasma genitalium
Ureaplasma urealyticum
Escherichia coli
Cryptosporidium
Entamoeba histolytica
Giardia lamblia
Brucella abortus
Clostridium difficile
Streptococcus pyogenes
Chlamydia pneumoniae
Chlamydia psittaci
From Kim PS, Klausmeier TL, Orr DP: Reactive arthritis: a review, J Adolesc
Health 44:309–315, 2009 (Table 2, p 311).
Epidemiology
JIA is diagnosed in 90 per 100,000 U.S. children every year (see Chapter 180 ).
ERA accounts for 10–20% of JIA, and has a mean age at onset of 12 yr. In India,
ERA is the most common category of JIA, accounting for 35% of cases. Unlike
other JIA categories, males are affected more often than females, accounting for
60% of ERA cases. AS occurs in 0.2–0.5% of adults, with approximately 15% of
cases beginning in childhood. These disorders can be familial, largely as a result
of the influence of human leukocyte antigen (HLA )-B27 , which is found in
90% of JAS and 50% of ERA patients compared to 7% of healthy individuals.
Approximately 20% of children with ERA have a family history of HLA-B27–
associated disease, such as reactive arthritis, AS, or IBD with sacroiliitis.
Table 181.3
Assessment in Spondyloarthritis International Society
(ASAS) Classification Criteria for Spondyloarthritis (SpA)
Enthesitis-Related Arthritis
Children have ERA if they have either arthritis and enthesitis or arthritis or
enthesitis, with at least 2 of the following characteristics: (1) sacroiliac joint
tenderness or inflammatory lumbosacral pain, (2) presence of HLA-B27, (3)
onset of arthritis in a male older than 6 yr, (4) acute anterior uveitis, and (5) a
family history of an HLA-B27–associated disease (ERA, sacroiliitis with IBD,
reactive arthritis, or acute anterior uveitis) in a first-degree relative. Patients with
psoriasis (or a family history of psoriasis in a first-degree relative), a positive–
rheumatoid factor (RF) test result, or systemic arthritis are excluded from this
group. During the 1st 6 mo of disease the arthritis is typically asymmetric and
involves ≤4 joints. most frequently the knees, ankles, and hips. Inflammation of
the small joints of the foot, or tarsitis, is highly suggestive of ERA. Enthesitis is
typically symmetric and typically affects the lower limbs. Up to 40% of children
develop clinical or radiographic evidence of sacroiliac joint arthritis as part of
their disease; approximately 20% have evidence of sacroiliac joint arthritis at
diagnosis. When the sacroiliac or other axial joints are involved, children may
experience inflammatory back pain (Table 181.4 ), hip pain, and alternating
buttock pain. Patients may also experience pain with palpation of the lower back
or with pelvic compression. The risk of sacroiliac joint arthritis is highest in
children who are HLA-B27 positive and have an elevated C-reactive protein
(CRP). Untreated sacroiliitis may, but does not always, evolve into AS;
additional risk factors for progression are unclear.
Table 181.4
Symptoms Characteristic of Inflammatory
Back Pain
Psoriatic Arthritis
Psoriatic arthritis accounts for approximately 5% of JIA. Common clinical
features of psoriatic arthritis are nail pitting (Fig. 181.1 ), onycholysis, and
dactylitis (sausage-like swelling of fingers or toes).
1976. Image #6950.)
Rubella
Alphaviruses
Ross River
Chikungunya
O'nyong-nyong
Mayaro
Sindbis
Ockelbo
Pogosta
Orthopoxviruses
Adenoviruses
Adenovirus 7
Herpesviruses
Epstein-Barr
Cytomegalovirus
Varicella-zoster
Herpes simplex
Paramyxoviruses
Mumps
Flavivirus
Zika virus
Hepadnavirus
Hepatitis B
Enteroviruses
Echovirus
Coxsackievirus B
Table 183.2
† Had highest prevalence of central nervous system disease but did not describe incidence in 1st
yr.
‡ Headache reported in 95% of patients.
From Petty RE, Laxer RM, Lindsley CB, Wedderburn LR, editors: Textbook of pediatric
rheumatology, ed 7, Philadelphia, 2016, Elsevier (Table 23.5, p 291).
Renal disease in SLE is often asymptomatic, underscoring the need for careful
monitoring of blood pressure and urinalyses; in adolescents, SLE can present
with nephrotic syndrome and/or renal failure with the predominant symptoms
being edema, fatigue, changes in urine color, and nausea/vomiting. Because SLE
symptoms and findings may develop serially over several years, and not all may
be present simultaneously, the diagnosis may require longitudinal follow up.
SLE is often characterized by periods of flare and disease quiescence but may
follow a more smoldering disease course. The neuropsychiatric complications
of SLE may occur with or without apparently active SLE, posing a particularly
difficult diagnostic challenge in adolescents, who are already at high risk for
mood disorders (Fig. 183.2 ). Long-term complications of SLE and its therapy,
including accelerated atherosclerosis and osteoporosis, become clinically evident
in young to middle adulthood. SLE is a disease that evolves over time in each
affected individual, and new manifestations arise even many years after
diagnosis.
FIG. 183.2 Overlapping neuropsychiatric symptoms in pediatric SLE. Patients with
pediatric SLE typically have >1 neuropsychiatric symptom—in particular for seizures.
(From Silverman E, Eddy A: Systemic lupus erythematosus. In Cassidy JT, Petty RE,
Laxer RM, et al, editors, Textbook of pediatric rheumatology, ed 6, Philadelphia, 2011,
Saunders Elsevier, Fig 21-17, p 329.)
Diagnosis
The diagnosis of SLE requires a comprehensive clinical and laboratory
assessment revealing characteristic multisystem disease and excluding other
etiologies, including infection and malignancy. Presence of 4 of the 11
American College of Rheumatology (ACR) 1997 revised classification criteria
for SLE simultaneously or cumulatively over time establishes the diagnosis
(Table 183.3 ). Of note, although a positive antinuclear antibody (ANA) test
result is not required for the diagnosis of SLE, ANA-negative lupus is extremely
rare. ANA is very sensitive for SLE (95–99%), but it is not very specific (50%).
The ANA may be positive many years before a diagnosis of SLE is established.
However, most asymptomatic, ANA-positive patients do not have SLE or other
autoimmune disease.
Table 183.3
American College of Rheumatology (ACR)
1997 Revised Classification Criteria for
Systemic Lupus Erythematosus*
Malar rash
Discoid rash
Photosensitivity
Oral or nasal ulcers
Arthritis
Nonerosive, ≥2 joints
Serositis
Pleuritis, pericarditis, or peritonitis
Renal manifestations †
Consistent renal biopsy
Persistent proteinuria or renal casts
Seizure or psychosis
Hematologic manifestations †
Hemolytic anemia
Leukopenia (<4,000 leukocytes/mm3 )
Lymphopenia (<1,500 leukocytes/mm3 )
Thrombocytopenia (<100,000 thrombocytes/mm3 )
Immunologic abnormalities †
Positive anti–double-stranded DNA or anti-Smith antibody
False-positive rapid plasma reagin test result, positive lupus
anticoagulant test result, or elevated anticardiolipin IgG or IgM
antibody
Positive antinuclear antibody test result
were developed for classification in clinical trials and not for clinical diagnosis.
† Each of these criteria counts as a single criterion whether 1 or more definitions
are satisfied.
Antibodies against dsDNA and anti-Smith are specific for SLE (98%) but not
as sensitive (40–65%). Hypocomplementemia , although common in SLE, is
not one of the ACR classification criteria; however, hypocomplementemia has
been added to updated criteria validated by the Systemic Lupus International
Collaborating Clinics (SLICC) in 2012 (Table 183.4 ). Other differences in the
SLICC criteria include the addition of nonscarring alopecia, additional cutaneous
and neurologic manifestations of lupus, and a positive direct Coombs test in the
absence of hemolytic anemia. The SLICC criteria have been validated in
pediatric SLE and have been shown to have higher sensitivity (93% vs 77%) but
lower specificity (85% vs 99%) than the ACR criteria.
Table 183.4
Systemic Lupus International Collaborating
Clinics (SLICC) Classification Criteria for
Systemic Lupus Erythematosus*
Clinical Criteria
Immunologic Criteria
Differential Diagnosis
Multiorgan disease is the hallmark of SLE. Given its wide array of potential
clinical manifestations, SLE is in the differential diagnosis of many clinical
scenarios, including unexplained fevers, joint pain or arthritis, rash, cytopenias,
nephritis, nephrotic syndrome, pleural or pericardial effusions or other
cardiopulmonary abnormalities, and new-onset psychosis, movement disorders,
or seizures. For patients ultimately diagnosed with pediatric SLE, the initial
differential diagnosis often includes infections (sepsis, EBV, parvovirus B19,
endocarditis), malignancies (leukemia, lymphoma), poststreptococcal
glomerulonephritis, other rheumatologic conditions (juvenile idiopathic arthritis,
vasculitides), and drug-induced lupus.
Drug-induced lupus refers to the presence of SLE manifestations triggered
by exposure to specific medications, including hydralazine, minocycline, many
anticonvulsants, sulfonamides, and antiarrhythmic agents (Table 183.5 ). In
individuals prone to SLE, these agents may act as a trigger for true SLE, but
more often these agents provoke a reversible lupus-like syndrome. Unlike SLE,
drug-induced lupus affects males and females equally. A genetic predisposition
toward slow drug acetylation may increase the risk of drug-induced lupus.
Circulating antihistone antibodies are often present in drug-induced SLE; these
antibodies are only detected in up to 20% of individuals with SLE. Hepatitis,
which is rare in SLE, is more common in drug-induced lupus. Individuals with
drug-induced lupus are less likely to demonstrate antibodies to dsDNA,
hypocomplementemia, and significant renal or neurologic disease. In contrast to
SLE, manifestations of drug-induced lupus typically resolve after withdrawal of
the offending medication; however, complete recovery may take several months
to years, requiring treatment with hydroxychloroquine, NSAIDs, and/or
corticosteroids.
Table 183.5
Medications Associated With Drug-Induced
Lupus
Definite Association
Laboratory Findings
A positive ANA test is present in 95–99% of SLE patients. ANA has poor
specificity for SLE, however, because up to 20% of healthy individuals also have
a positive ANA test result, making the ANA a poor screen for SLE when used in
isolation. High titers are more suggestive of underlying autoimmune disease, but
ANA titers do not correlate with disease activity, so repeating ANA titers after
diagnosis is not helpful. Antibodies to dsDNA are specific for SLE, and in many
individuals, anti-dsDNA levels correlate with disease activity, particularly in
those with significant nephritis. Anti-Smith antibody, although found specifically
in patients with SLE, does not correlate with disease activity. Serum levels of
total hemolytic complement (CH50 ), C3, and C4 are typically decreased in
active disease and often improve with treatment. Table 183.6 lists autoantibodies
found in SLE along with their clinical associations. Hypergammaglobulinemia is
a common but nonspecific finding. Inflammatory markers, particularly
erythrocyte sedimentation rate, are often elevated in active disease. C-reactive
protein (CRP) correlates less well with disease activity; significantly elevated
CRP values often reflect infection, whereas chronic mild elevation may indicate
increased cardiovascular risk.
Table 183.6
Treatment
Treatment of SLE is tailored to the individual and is based on specific disease
manifestations and medication tolerability. For all patients, sunscreen and
avoidance of prolonged direct sun exposure and other UV light may help control
disease and should be reinforced at every visit with the patient.
Hydroxychloroquine is recommended for all individuals with SLE when
tolerated. In addition to treating mild SLE manifestations such as rash and mild
arthritis, hydroxychloroquine prevents SLE flares, improves lipid profiles, and
may improve mortality and renal outcomes. Potential toxicities include retinal
deposition and subsequent vision impairment; therefore, annual ophthalmology
exams are recommended for patients taking hydroxychloroquine, including
automated visual field testing as well as spectral-domain optical coherence
tomography (SD-OCT). Given that risk factors for ocular toxicity include
duration of use and dose, hydroxychloroquine in SLE should never be prescribed
at doses >6.5 mg/kg (maximum 400 mg daily), and newer ophthalmology
guidelines recommend limiting maintenance dosing to 4-5 mg/kg.
Corticosteroids are a treatment mainstay for significant manifestations of
Table 183.7
Prognosis
The severity of pediatric SLE is notably worse than the typical course for adult-
onset SLE. However, because of advances in the diagnosis and treatment of
SLE, survival has improved dramatically over the past 50 yr. Currently, the 5 yr
survival rate for pediatric SLE is approximately 95%, although the 10 yr survival
rate remains 80–90%. Given their long burden of disease, children and
adolescents with SLE face high risks of future morbidity and mortality from the
disease and its complications, as well as medication side effects (see Table 183.7
). Given the complex and chronic nature of SLE, it is optimal for children and
adolescents with SLE to be treated by pediatric rheumatologists in a
multidisciplinary clinic with access to a full complement of pediatric
subspecialists.
183.1
Neonatal Lupus
Deborah M. Friedman, Jill P. Buyon, Rebecca E. Sadun, Stacy P. Ardoin, Laura
E. Schanberg
cells (CD56), T-cell subsets (CD4, CD8, Th17), monocytes/macrophages
(CD14), and plasmacytoid DCs. Neopterin, IFN-inducible protein 10, monocyte
chemoattractant protein, myxovirus resistance protein, and von Willebrand factor
products, as well as other markers of vascular inflammation, may be elevated in
patients with JDM who have active inflammation.
Clinical Manifestations
Children with JDM present with either rash, insidious onset of weakness, or
both. Fevers, dysphagia or dysphonia, arthritis, muscle tenderness, and fatigue
are also commonly reported at diagnosis (Tables 184.1 and 184.2 ).
Table 184.1
Diagnostic Criteria for Juvenile Dermatomyositis
Classic rash Heliotrope rash of the eyelids
Gottron papules
Plus 3 of the following:
Weakness Symmetric
Proximal
Muscle enzyme elevation (≥1) Creatine kinase
Aspartate transaminase
Lactate dehydrogenase
Aldolase
Electromyographic changes Short, small polyphasic motor unit potentials
Fibrillations
Positive sharp waves
Insertional irritability
Bizarre, high-frequency repetitive discharges
Muscle biopsy Necrosis
Inflammation
Data from Bohan A, Peter JB: Polymyositis and dermatomyositis (2nd of 2 parts), N Engl J Med
292:403–407, 1975.
Table 184.2
Table 184.3
Frequency of Manifestations of Juvenile Dermatomyositis,
Juvenile Polymyositis, and Overlap Myositis
Laboratory Findings
Elevated serum levels of muscle-derived enzymes (creatine kinase [CK],
aldolase, aspartate transaminase, alanine transaminase [ALT], lactate
dehydrogenase) reflect muscle inflammation. Not all enzyme levels rise with
inflammation in a specific individual; ALT is usually elevated on initial
presentation, whereas CK level may be normal. The erythrocyte sedimentation
rate (ESR) is often normal, and the rheumatoid factor (RF) test result is typically
negative. There may be anemia consistent with chronic disease. Antinuclear
antibody (ANA) is present in >80% of children with JDM. Serologic testing
results are divided into 2 groups: myositis-associated antibodies (MAAs) and
myositis-specific antibodies (MSAs) . MAAs are associated with JDM, but are
not specific and can be seen in both overlap conditions and other rheumatic
diseases. MSAs are specific for myositis. Presence of MAAs such as SSA, SSB,
Sm, ribonucleoprotein (RNP), and double-stranded (ds) DNA may increase the
likelihood of overlap disease or connective tissue myositis. Antibodies to Pm/Scl
identify a small, distinct subgroup of myopathies with a protracted disease
course, often complicated by pulmonary interstitial fibrosis and cardiac
involvement. Similar to what is seen in adults, the presence of MSAs in JDM
such as anti–Jo-1, anti–Mi-2, anti-p155/140, anti-NXP2, and other myositis-
specific autoantibodies help define distinct clinical subsets and may predict the
development of complications, although differences remain in certain aspects
such as malignancy between adults and children. Anti-p155/140 antibodies also
fibers.
Autoimmunity is believed to be a key process in the pathogenesis of both
localized and systemic scleroderma, given the high percentage of affected
children with autoantibodies. Children with localized disease often have a
positive antinuclear antibody (ANA) test result (42%), and 47% of this subgroup
have antihistone antibodies. Children with JSSc have higher rates of ANA
positivity (80.7%) and may have anti–Scl-70 antibody (34%, antitopoisomerase
I). The relationship between specific autoantibodies and the various forms of
scleroderma is not well understood, and all antibody test results may be negative,
especially in JLS.
Classification
Localized scleroderma is distinct from systemic scleroderma and rarely
progresses to systemic disease. The category of JLS includes several subtypes
differentiated by both the distribution of the lesions and the depth of
involvement (Tables 185.1 and 185.2 ). Up to 15% of children have a
combination of 2 or more subtypes.
Table 185.1
Classification of Pediatric Scleroderma
(Morphea)
Localized Scleroderma
Plaque Morphea
Generalized Morphea
Bullous lesions that can occur with any of the subtypes of morphea
Linear Scleroderma
Linear lesions can extend through the dermis, subcutaneous tissue, and
muscle to underlying bone; more likely unilateral
Limbs/trunk:
One or more linear streaks of the extremities or trunk
Flexion contracture occurs when lesion extends over a joint; limb
length discrepancies
En coup de sabre:
Involves the scalp and/or face; lesions can extend into the central
nervous system, resulting in neurologic sequelae, most
commonly seizures and headaches
Parry-Romberg syndrome:
Hemifacial atrophy without a clearly definable en coup de sabre
lesion; can also have neurologic involvement
Deep Morphea
Systemic Sclerosis
Diffuse
Limited
Rare in childhood
Previously known as CREST (calcinosis cutis, Raynaud phenomenon,
esophageal dysfunction, sclerodactyly, and telangiectasia) syndrome
Table 185.2
Provisional Criteria for Classification of
Juvenile Systemic Sclerosis (JSSc)
Major Criterion (Required)*
Cutaneous: Sclerodactyly
Peripheral vascular: Raynaud phenomenon, nail fold capillary
abnormalities (telangiectasias), digital tip ulcers
Gastrointestinal: Dysphagia, gastroesophageal reflux
Cardiac: Arrhythmias, heart failure
Renal: Renal crisis, new-onset arterial hypertension
Respiratory: Pulmonary fibrosis (high-resolution CT/radiography),
decreased diffusing capacity for carbon monoxide, pulmonary arterial
hypertension
Neurologic: Neuropathy, carpal tunnel syndrome
Musculoskeletal: Tendon friction rubs, arthritis, myositis
Serologic: Antinuclear antibodies—SSc-selective autoantibodies
(anticentromere, antitopoisomerase I [Scl-70], antifibrillarin, anti-
PM/Scl, antifibrillin, or anti-RNA polymerase I or III
From Zulian F, Woo P, Athreya BH, et al: The Pediatric Rheumatology European
Society/American College of Rheumatology/European League against
Rheumatism provisional classification criteria for juvenile systemic sclerosis,
Arthritis Rheum 57:203–212, 2007.
Epidemiology
Juvenile scleroderma is rare, with an estimated prevalence of 1 in 100,000
children. LS is much more common than SSc in children, by a 10 : 1 ratio, with
linear scleroderma being the most common subtype. LS is predominantly a
pediatric condition, with 65% of patients diagnosed before age 18 yr. After age 8
yr the female/male ratio for both LS and SSc is approximately 3 : 1, whereas in
patients younger than 8 yr, there is no sex predilection.
Clinical Manifestations
Localized Scleroderma
The onset of scleroderma is generally insidious, and manifestations vary
according to disease subtype. The initial skin manifestations of localized disease
usually include erythema or a bluish hue seen around an area of waxy
induration; subtle erythema may be the only presenting sign (Fig. 185.1 ).
Edema and erythema are followed by indurated, hypopigmented or
hyperpigmented atrophic lesions (Fig. 185.2 ). LS varies in size from a few
centimeters to the entire length of the extremity, with varying depth. Patients
late in the course, are subtle, and include cough and dyspnea on exertion.
Pulmonary evaluation should include pulmonary function tests (PFTs) such as
diffusion capacity of carbon monoxide (DLCO ), bronchoalveolar lavage (BAL),
and high-resolution chest computed tomography (HRCT). PFTs reveal decreased
vital capacity and decreased DLCO , whereas neutrophilia or eosinophilia on
BAL suggest active alveolitis. Chest CT is much more sensitive than chest
radiographs, which are often normal, showing typical basilar ground-glass
abnormalities, reticular linear opacities, nodules, honeycombing, and mediastinal
adenopathy.
Gastrointestinal tract disease is seen in 25% of children with SSc. Common
manifestations include esophageal and intestinal dysmotility resulting in
dysphagia, reflux, dyspepsia, gastroparesis, bacterial overgrowth, dilated bowel
loops and pseudoobstruction, and dental caries, as well as malabsorption and
failure to thrive. Renal arterial disease can cause chronic or severe episodic
hypertension; unlike adult disease, renal crisis is rare. Cardiac fibrosis is
associated with arrhythmias, ventricular hypertrophy, and decreased cardiac
function. Mortality from JSSc is usually a result of cardiopulmonary disease. A
scoring system helps identify the severity of the multiorgan involvement (Table
185.3 ).
Table 185.3
Medsger Systemic Sclerosis Severity Scale*
ORGAN 2
0 (NORMAL) 1 (MILD) 3 (SEVERE) 4 (END STAGE)
SYSTEM (MODERATE)
General Wt loss <5% Wt loss 5–10% Wt loss 10– Wt loss 15–20% Wt loss 20%+
15%
Hct 37%+ Hct 33–37% Hct 29–33% Hct 25–29% Hct 25%
Hb 12.3+ g/dL Hb 11.0-12.2 g/dL Hb 9.7-10.9 Hb 8.3-9.6 g/dL Hb <8.3 g/dL
g/dL
Peripheral No RP; RP not RP requiring Digital pitting Digital tip Digital gangrene
vascular requiring vasodilators scars ulcerations
vasodilators
Skin TSS 0 TSS 1-14 TSS 15-29 TSS 30-39 TSS 40+
Joint/tendon FTP 0-0.9 cm FTP 1.0-1.9 cm FTP 2.0-3.9 cm FTP 4.0-4.9 cm FTP 5.0+ cm
Muscle Normal Proximal Proximal Proximal Ambulation aids
proximal weakness, mild weakness, weakness, severe required
muscle strength moderate
Gastrointestinal Normal Distal esophageal Antibiotics Malabsorption Hyperalimentation
tract esophagogram; hypoperistalsis; required for syndrome; required
normal small small bowel series bacterial episodes of
bowel series abnormal overgrowth pseudoobstruction
Lung DLCO DLCO 70– DLCO 50– DLCO <50% Oxygen required
80%+ 79% 69% FVC <50%
FVC 80%+ FVC 70–79% FVC 50– sPAP 65+
No fibrosis Basilar rales; 69% mm Hg
on fibrosis on sPAP 50-64
radiograph radiograph mm Hg
sPAP <35 sPAP 35-49
mm Hg mm Hg
Heart ECG normal ECG conduction ECG ECG arrhythmia CHF
defect arrhythmia requiring therapy
LVEF 50%+ LVEF 45–49% LVEF 40–44% LVEF 30–40% LVEF <30%
Kidney No history of History of SRC History of SRC History of SRC History of SRC with
SRC with with serum with serum with serum serum creatinine
serum creatinine <1.5 creatinine 1.5- creatinine 2.5-5.0 >5.0 mg/dL or
creatinine <1.3 mg/dL 2.4 mg/dL mg/dL dialysis required
mg/dL
* If 2 items are included for a severity grade, only 1 is required for the patient to be scored as
having disease of that severity level.
CHF, Congestive heart failure; DLCO , diffusing capacity for carbon monoxide, % predicted; ECG,
electrocardiogram; FTP, fingertip-to-palm distance in flexion; FVC, forced vital capacity, %
predicted; Hb, hemoglobin; Hct, hematocrit; LVEF, left ventricular ejection fraction; RP, Raynaud
phenomenon; sPAP, estimated pulmonary artery pressure by Doppler echo; SRC, scleroderma
renal crisis; TSS, total skin score; Wt, weight.
Modified from Medsger TA Jr, Bombardieri S, Czirjak L, et al: Assessment of disease severity and
prognosis, Clin Exp Rheumatol 21(3 Suppl 29):S51, 2003 (Table 1, p S-43).
Raynaud Phenomenon
Raynaud phenomenon (RP) is the most frequent initial symptom in pediatric
systemic sclerosis, present in 70% of affected children months to years before
other manifestations. RP refers to the classic triphasic sequence of blanching,
cyanosis, and erythema of the digits induced by cold exposure and/or emotional
stress. RP is typically independent of an underlying rheumatic disease (Raynaud
disease) but can result from rheumatic diseases such as scleroderma, systemic
lupus erythematosus (SLE), and mixed connective tissue disease (Fig. 185.6 ).
The color changes are brought about by (1) initial arterial vasoconstriction,
resulting in hypoperfusion and pallor (blanching), (2) venous stasis (cyanosis),
and (3) reflex vasodilation caused by the factors released from the ischemic
phase (erythema). The color change is classically reproduced by immersing the
hands in iced water and reversed by warming. During the blanching phase, there
is inadequate tissue perfusion in the affected area, associated with pain and
paresthesias and resulting in ischemic damage only when associated with a
rheumatic disease. The blanching usually affects the distal fingers but may also
involve thumbs, toes, ears, and tip of the nose. The affected area is usually well
among people along the Silk Road, evidence for genetic anticipation, and
genome-wide analysis. Genome wide analysis studies among Turkish and
Japanese BD patients confirm the marked association with HLA-B5101. Other
significant associations include interleukin (IL)-10 and IL-23R/IL-12Rβ2 genes.
Other possible susceptibility loci in a Turkish cohort demonstrate associations
with STAT4 (a transcription factor in a signaling pathway related to cytokines
such as IL-12, type I interferons, and IL-23) and ERAP1 (an endoplasmic
reticulum–expressed aminopeptidase that functions in processing of peptides
onto major histocompatibility complex class I).
The autoinflammatory nature of BD is suggested by its episodic nature, the
prominent innate immune system activation, the absence of identifiable
autoantibodies, and the co-association with the MEFV (Mediterranean fever)
gene. An infectious agent may be responsible for inducing the aberrant innate
immune system attacks in the genetically predisposed host. A number of
infectious agents have been implicated and include streptococci, herpes simplex
virus type 1, and parvovirus B19.
Table 186.1
The mean age of the first symptom is between 8 and 12 yr. The most frequent
initial symptom is a painful oral ulcer (Fig. 186.1 ). The oral ulcers are often
recurrent, may be single or multiple, range from 2-10 mm, and may be in any
Clinical Manifestations
International classification criteria have been developed for the diagnosis of
Sjögren syndrome in adult patients, but these criteria apply poorly to children.
Although diagnostic criteria in children have been proposed, they have not been
validated (Table 187.1 ). Recurrent parotid gland enlargement and parotitis are
the most common manifestations in children (>70%), whereas sicca syndrome
(dry mouth, painful mucosa, sensitivity to spicy foods, halitosis, widespread
dental caries) predominates in adults. In a cross-sectional study of children with
Sjögren syndrome, manifestations included recurrent parotitis (72%), sicca
symptoms (38%), polyarthritis (18%), vulvovaginitis (12%), hepatitis (10%),
Raynaud phenomenon (10%), fever (8%), renal tubular acidosis (9%),
lymphadenopathy (8%), and central nervous system (CNS) involvement (5%).
Table 187.1
Proposed Criteria for Pediatric Sjögren
Syndrome*
I. CLINICAL SYMPTOMS
1. Oral: recurrent parotitis or enlargement of parotid gland, dry
mouth (xerostomia)
2. Ocular: dry eyes (xerophthalmia) recurrent conjunctivitis without
obvious allergic or infectious etiology, keratoconjunctivitis sicca
3. Other mucosal: recurrent vaginitis
4. Systemic: fever, noninflammatory arthralgias, hypokalemic
paralysis, abdominal pain
II. IMMUNOLOGIC ABNORMALITIES
Presence of at least 1 of the following antibodies: anti-SSA, anti-
SSB, high-titer antinuclear antibody, rheumatoid factor
III. OTHER ABNORMALITIES OR INVESTIGATIONS
1. Biochemical: elevated serum amylase
2. Hematologic: leukopenia, high erythrocyte sedimentation rate
3. Immunologic: polyclonal hyperimmunoglobulinemia
4. Renal: renal tubular acidosis
5. Histologic proof of lymphocytic infiltration of salivary glands or
other organs (i.e., liver)
6. Objective documentation of ocular dryness (rose bengal staining
or Schirmer test)
7. Positive findings of parotid gland scintigraphy
IV. Exclusion of all other autoimmune diseases
Diagnosis
Clinical presentation of recurrent parotitis and or recurrent parotid gland
swelling in a child or adolescent is characteristic and should raise the suspicion
considered and evaluated. If the workup is reassuring, the inflammatory episodes
resolve, and the child is otherwise well without unusual physical findings,
observance is often warranted because these episodes are likely to resolve as the
child's immune system matures.
Table 188.1
Differential Diagnosis of Periodic Fever
Hereditary
See Table 188.2 .
Nonhereditary
A. Infectious
1. Hidden infectious focus (e.g., aortoenteric fistula, lung
sequestration)
2. Recurrent infection/reinfection (e.g., chronic meningococcemia,
immune deficiency)
3. Specific infection (e.g., Whipple disease, malaria)
B. Noninfectious inflammatory disorder:
1. Adult-onset Still disease
2. Systemic-onset juvenile idiopathic arthritis
3. Periodic fever, aphthous stomatitis, pharyngitis, and adenitis
4. Schnitzler syndrome
5. Behçet syndrome
6. Crohn disease
7. Sarcoidosis
C. Neoplastic
1. Lymphoma (e.g., Hodgkin disease, angioimmunoblastic
lymphoma)
2. Solid tumor (e.g., pheochromocytoma, myxoma, colon
carcinoma)
3. Histiocytic disorders
D. Vascular (e.g., recurrent pulmonary embolism)
E. Hypothalamic
F. Psychogenic periodic fever
G. Factitious or fraudulent
Adapted from Simon A, van der Meer JWM, Drenth JPH: Familial
autoinflammatory syndromes. In Firestein GS, Budd RC, Gabriel SE, et al,
editors: Kelley's textbook of rheumatology, ed 9, Philadelphia, 2012, Saunders
(Table 97-2).
Classification of Autoinflammatory
Disorders
Because of the rapidly expanding number of autoinflammatory disorders and
their varied clinical presentation, it can be difficult to group these disorders in a
meaningful manner. Some autoinflammatory disorders present with prominent
fevers and are known as hereditary periodic fever syndromes . These include 2
disorders with an autosomal recessive mode of inheritance, familial
Mediterranean fever (FMF ; MIM249100) and the hyperimmunoglobulinemia D
(hyper-IgD) with periodic fever syndrome (HIDS ; MIM260920). Hereditary
periodic fever syndromes with an autosomal dominant mode of inheritance
include the tumor necrosis factor (TNF) receptor–associated periodic syndrome
(TRAPS ; MIM191190) and a spectrum of disorders known as the cryopyrin-
associated periodic syndromes (CAPS ), or cryopyrinopathies. From mildest to
most severe, CAPS include the familial cold autoinflammatory syndrome
(FCAS1 ; MIM120100), Muckle-Wells syndrome (MWS ; MIM191100), and
neonatal-onset multisystem inflammatory disease (NOMID ; MIM607115) (also
known as chronic infantile neurologic cutaneous and articular syndrome,
CINCA ) (Table 188.2 ).
Table 188.2
Autoinflammatory Disorders
GENETIC
FUNCTIONAL ASSOCIATED
DISEASE DEFECT/PRESUMED INHERITANCE AFFECTED CELLS
DEFECTS FEATURES
PATHOGENESIS
Familial Mutations of MEFV AR Mature granulocytes, Decreased Recurrent
Mediterranean (lead to gain of pyrin cytokine-activated production of pyrin fever, serositis,
fever function, resulting in monocytes permits ASC- and
inappropriate IL-1β induced IL-1 inflammation
release) processing and responsive to
inflammation colchicine.
following Predisposes to
subclinical serosal vasculitis and
injury; macrophage inflammatory
apoptosis decreased bowel disease
Mevalonate Mutations of MVK (lead AR Affecting Periodic fever
kinase deficiencyto a block in the cholesterol and
(hyper IgD mevalonate pathway). synthesis; leukocytosis
syndrome) Interleukin-1β mediates pathogenesis of with high IgD
the inflammatory disease is unclear levels
phenotype
Muckle–Wells Mutations of NLRP3 AD PMNs, monocytes Defect in Urticaria,
syndrome (also called PYPAF1 or cryopyrin, involved SNHL,
NALP3 ) lead to in leukocyte amyloidosis
constitutive activation of apoptosis and NF-
the NLRP3 κB signaling and
inflammasome IL-1 processing
Familial cold Mutations of AD PMNs, monocytes Same as above Nonpruritic
autoinflammatory NLRP3 (see above) urticaria,
syndrome Mutations of arthritis, chills,
NLRP12 fever, and
leukocytosis
after cold
exposure
Neonatal-onset Mutations of NLRP3 PMNs, chondrocytes Same as above Neonatal-onset
multisystem (see above) rash, chronic
inflammatory meningitis, and
disease (NOMID) arthropathy
or chronic with fever and
infantile inflammation
neurologic
cutaneous and
articular
syndrome
(CINCA)
TNF receptor– Mutations of AD PMNs, monocytes Mutations of 55- Recurrent
associated TNFRSF1A (resulting in kDa TNF receptor fever, serositis,
periodic increased TNF leading to rash, and ocular
syndrome inflammatory signaling) intracellular or joint
(TRAPS) receptor retention inflammation
or diminished
soluble cytokine
receptor available
to bind TNF
Pyogenic sterile Mutations of PSTPIP1 AD Hematopoietic tissues, Disordered actin Destructive
arthritis, (also called C2BP1 ) upregulated in reorganization arthritis,
pyoderma (affects both pyrin and activated T cells leading to inflammatory
gangrenosum, protein tyrosine compromised skin rash,
acne (PAPA) phosphatase to regulate physiologic myositis
syndrome innate and adaptive signaling during
immune responses) inflammatory
response
Blau syndrome Mutations of NOD2 AD Monocytes Mutations in Uveitis,
(also called CARD15 ) nucleotide binding granulomatous
(involved in various site of CARD15, synovitis,
inflammatory processes) possibly disrupting campodactyly,
interactions with rash, and
lipopolysaccharides cranial
and NF-κB neuropathies,
signaling 30% develop
Crohn disease
Chronic recurrent Mutations of LPIN2 AR Neutrophils, bone Undefined Chronic
multifocal (increased expression of marrow cells recurrent
osteomyelitis and the proinflammatory multifocal
congenital genes) osteomyelitis,
dyserythropoietic transfusion-
anemia (Majeed dependent
syndrome) anemia,
cutaneous
inflammatory
disorders
Early-onset Mutations in IL-10 AR Monocyte/macrophage, IL-10 deficiency Enterocolitis,
inflammatory (results in increase of activated T cells leads to increase of enteric fistulas,
bowel disease many proinflammatory TNFγ and other perianal
cytokines) proinflammatory abscesses,
cytokines chronic
folliculitis
Early-onset Mutations in IL-10RA AR Monocyte/macrophage, Mutation in IL-10 Enterocolitis,
inflammatory (see above) activated T cells receptor alpha leads enteric fistulas,
bowel disease to increase of TNFγ perianal
and other abscesses,
proinflammatory chronic
cytokines folliculitis
Early-onset Mutations in IL-10RB AR Monocyte/macrophage, Mutation in IL-10 Enterocolitis,
inflammatory (see above) activated T cells receptor beta leads enteric fistulas,
bowel disease to increase of TNFγ perianal
and other abscesses,
proinflammatory chronic
cytokines folliculitis
AD, autosomal dominant; AR, autosomal recessive; Ig, immunoglobulin; IL, interleukin; NF-κB,
nuclear factor-κB; PMN, polymorphonuclear neutrophil; SNHL, sensorineural hearing loss; TNF,
tumor necrosis factor.
From Verbsky JW, Routes JR: Recurrent fever, infections, immune disorders, and
autoinflammatory diseases. In Kliegman RM, Lyse PS, Bordini BJ, et al, editors: Nelson pediatric
symptom-based diagnosis. Philadelphia, 2018, Elsevier, Table 41-5.
Table 188.3
Clinical Grouping of Autoinflammatory
Diseases by Skin Manifestations
Table 188.4
Autoinflammatory Bone Disorders
Table 188.5
Clues That May Assist in Diagnosis of Autoinflammatory
Syndromes
AGE OF ONSET
At birth NOMID, DIRA, MWS
Infancy and 1st yr of life HIDS, FCAS, NLRP12
Toddler PFAPA
Late childhood PAPA
Most common of autoinflammatory syndromes to have onset in TRAPS, DITRA
adulthood
Variable (mostly in childhood) All others
ETHNICITY AND GEOGRAPHY
Armenians, Turks, Italian, Sephardic Jews FMF
Arabs FMF, DITRA (Arab Tunisian)
Dutch, French, German, Western Europe HIDS, MWS, NLRP12
United States FCAS
Can occur in blacks (West Africa origin) TRAPS
Eastern Canada, Puerto Rico DIRA
Worldwide All others
TRIGGERS
Vaccines HIDS
Cold exposure FCAS, NLRP12
Stress, menses FMF, TRAPS, MWS, PAPA, DITRA
Minor trauma PAPA, MWS, TRAPS, HIDS
Exercise FMF, TRAPS
Pregnancy DITRA
Infections All, especially DITRA
ATTACK DURATION
<24 hr FCAS, FMF
1-3 days FMF, MWS, DITRA (fever)
3-7 days HIDS, PFAPA
>7 days TRAPS, PAPA
Almost always “in attack” NOMID, DIRA
INTERVAL BETWEEN ATTACKS
3-6 wk PFAPA, HIDS
>6 wk TRAPS
Mostly unpredictable All others
Truly periodic PFAPA, cyclic neutropenia
USEFUL LABORATORY TESTS
Acute-phase reactants must be normal between attacks PFAPA
Urine mevalonic acid in attack HIDS
IgD > 100 mg/dL HIDS
Proteinuria (amyloidosis) FMF, TRAPS, MWS, NOMID
RESPONSE TO THERAPY
Corticosteroid dramatic PFAPA
Corticosteroid partial TRAPS, FCAS, MWS, NOMID, PAPA*
Colchicine FMF, PFAPA (30% effective)
Cimetidine PFAPA (30% effective)
Etanercept TRAPS, FMF arthritis
Anti–IL-1 dramatic DIRA (anakinra), FCAS, MWS, NOMID,
PFAPA
Anti–IL-1 mostly TRAPS, FMF
Anti–IL-1 partial HIDS, PAPA
* For intraarticular corticosteroids.
DIRA, Deficiency of IL-1 receptor antagonist; DITRA, deficiency of IL-36 receptor antagonist
(generalized pustular psoriasis); FCAS, familial cold autoinflammatory syndrome; FMF, familial
weekly to 1-2 flares per year. Table 188.6 lists diagnostic criteria for FMF.
FIG. 188.2 Characteristic erysipeloid erythema associated with familial Mediterranean
fever. This rash appears during a flare and overlies the ankle or dorsum of the foot.
Table 188.6
Diagnostic Criteria for Familial
Mediterranean Fever (FMF)*
Major Criteria
Minor Criteria
† Typical attacks are defined as recurrent (≥3 of the same type), febrile (≥38°C),
Constantly Present
Specific Features
obligatory.
From Firestein GS, Budd RC, Gabriel SE, et al, editors: Kelly & Firestein's
textbook of rheumatology, ed 10, Philadelphia, 2016, Elsevier (Table 97-4, p
1674).
Standards for the treatment of HIDS are evolving. Very few patients respond
to colchicine, and milder disease courses may respond to nonsteroidal
FIG. 188.6 Cutaneous manifestations of tumor necrosis factor receptor–associated
periodic syndrome. A, Right flank of a patient with the T50M mutation. B, Serpiginous
rash involving the face, neck, torso, and upper extremities of a child with the C30S
mutation. C, Erythematous, macular patches with crusting on the flexor surface of the
right arm of a patient with the T50M mutation. (From Hull KM, Drewe, Aksentijevich I, et
al: The TNF receptor-associated periodic syndrome [TRAPS]: emerging concepts of an
autoinflammatory syndrome, Medicine (Baltimore) 81:349–368, 2002.)
Table 188.8
Diagnostic Indicators of Tumor Necrosis
Factor Receptor–Associated Periodic
Syndrome (TRAPS)
1. Recurrent episodes of inflammatory symptoms spanning >6 mo duration
(several symptoms generally occur simultaneously)
a. Fever
b. Abdominal pain
c. Myalgia (migratory)
d. Rash (erythematous macular rash occurs with myalgia)
e. Conjunctivitis or periorbital edema
f. Chest pain
g. Arthralgia or monoarticular synovitis
2. Episodes last >5 days on average (although variable)
3. Responsive to glucocorticosteroids but not colchicine
4. Affects family members in autosomal dominant pattern (although may not
always be present)
5. Any ethnicity may be affected
Table 188.9
Diagnostic Criteria for Familial Cold
Autoinflammatory Syndrome (FCAS)
From Hoffman HM, Wanderer AA, Broide DH: Familial cold autoinflammatory
syndrome: phenotype and genotype of an autosomal dominant periodic fever, J
Allergy Clin Immunol 108:615–620, 2001.
In contrast to FCAS, the febrile episodes of MWS are not cold induced but
are characterized by the same urticarial-like rash seen in FCAS (Fig. 188.8 ).
Many MWS patients also develop progressive sensorineural hearing loss, and
untreated, approximately 30% of MWS patients develop AA amyloidosis.
NOMID patients present in the neonatal period with a diffuse, urticarial rash,
daily fevers, and dysmorphic features (Fig. 188.9 ). Significant joint deformities,
particularly of the knees, may develop because of bony overgrowth of the
epiphyses of the long bones (Fig. 188.10 ). NOMID patients also develop
chronic aseptic meningitis, leading to increased intracranial pressure, optic disc
edema, visual impairment, progressive sensorineural hearing loss, and
intellectual disability (Fig. 188.11 ).
FIG. 188.8 Urticarial-like skin rash in a patient with Muckle-Wells syndrome. (Courtesy
Dr. D. L. Kastner, National Institutes of Health, Bethesda, Maryland; from Simon A, van
der Meer JWM, Drenth JPH: Familial autoinflammatory syndromes. In Firestein GS,
Budd RC, Gabriel SE, et al, editors: Kelley's textbook of rheumatology, ed 9,
Philadelphia, 2012, Saunders, Fig 97-14.)
disease occurs infrequently in children but typically manifests as renal
insufficiency, proteinuria, transient pyuria, or microscopic hematuria caused by
early monocellular infiltration or granuloma formation in kidney tissue. Only a
small fraction of children have hypercalcemia or hypercalciuria, which is
therefore an infrequent cause of kidney disease. Sarcoid granulomas can also
infiltrate the heart and lead to cardiac arrhythmias and, rarely, sudden death.
Other rare sites of disease involvement include blood vessels of any size, the
gastrointestinal tract, parotid gland, muscles, bones, and testes.
Table 190.1
Respiratory System
Peribronchial and interstitial infiltrates on chest radiograph
Pulmonary nodules
Musculoskeletal System
Gastrointestinal Tract
Extreme irritability
Aseptic meningitis (pleocytosis of cerebrospinal fluid)
Facial nerve palsy
Sensorineural hearing loss
Genitourinary System
Urethritis/meatitis, hydrocele
Other Findings
* Patients with fever at least 5 days and <4 principal criteria can be diagnosed
of the hands and feet are present, Kawasaki disease diagnosis can be made on
day 4 of illness. Experienced clinicians who have treated many patients with
Kawasaki disease may establish diagnosis before day 4 in rare cases.
‡ See differential diagnosis (Table 191.2 ).
coagulation.
Adenovirus
Enterovirus
Measles
Epstein-Barr virus
Cytomegalovirus
Bacterial Infections
Scarlet fever
Rocky Mountain spotted fever
Leptospirosis
Bacterial cervical lymphadenitis ± retropharyngeal phlegmon
Meningococcemia
Urinary tract infection
Rheumatologic Disease
* Detection of a virus does not exclude Kawasaki disease in the presence of the
Treatment
Patients with acute KD should be treated with 2 g/kg of IVIG as a single
infusion, usually administered over 10-12 hr within 10 days of disease onset, and
ideally as soon as possible after diagnosis (Table 191.3 ). In addition, moderate
(30-50 mg/kg/day divided every 6 hr) to high-dose aspirin (80-100 mg/kg/day
divided every 6 hr) should be administered until the patient is afebrile, then
lowered to antiplatelet doses. Other NSAIDs should not be given during therapy
with aspirin because they may block the action of aspirin. The mechanism of
action of IVIG in KD is unknown, but treatment results in defervescence and
resolution of clinical signs of illness in approximately 85% of patients. The
prevalence of coronary disease, in 20–25% in children treated with aspirin alone,
is <5% in those treated with IVIG and aspirin within the 1st 10 days of illness.
Strong consideration should be given to treating patients with persistent fever,
abnormal dimensions of the coronary arteries, or signs of systemic inflammation
who are diagnosed after the 10th day of fever. The dose of aspirin is usually
decreased from antiinflammatory to antithrombotic doses (3-5 mg/kg/day as a
single dose) after the patient has been afebrile for 48 hr. Aspirin is continued for
its antithrombotic effect until 6-8 wk after illness onset and is then discontinued
in patients who have had normal echocardiography findings throughout the
course of their illness. Patients with CAA continue with aspirin therapy and may
require anticoagulation, depending on the degree of coronary dilation (see later).
Table 191.3
Treatment of Kawasaki Disease
Acute Stage
Intravenous immune globulin 2 g/kg over 10-12 hr
and
Aspirin 30-50 mg/kg/day or 80-100 mg/kg/day divided every 6 hr orally
until patient is afebrile for at least 48 hr
Convalescent Stage
Aspirin 3-5 mg/kg once daily orally until 6-8 wk after illness onset if
normal coronary findings throughout course
Corticosteroids have been used as primary therapy with the 1st dose of IVIG
in hopes of improving coronary outcomes. A North American trial using a single
pulse dose of intravenous methylprednisolone (30 mg/kg) with IVIG as primary
therapy did not improve coronary outcomes. However, a trial in Japan utilizing
the Kobayashi score to identify high-risk children demonstrated improved
coronary outcomes with a regimen of prednisolone (2 mg/kg) plus IVIG as
primary therapy. Furthermore, a systematic review and meta-analysis of 16
comparative studies demonstrated that early treatment with corticosteroids
improved coronary artery outcomes in children with KD. Despite these
promising results, administration of corticosteroids as primary therapy to all
children with KD awaits the development of a risk score that identifies high-risk
children in a multiracial population.
IVIG-resistant KD occurs in approximately 15% of patients and is defined
by persistent or recrudescent fever 36 hr after completion of the initial IVIG
infusion. Patients with IVIG resistance are at increased risk for CAA.
Therapeutic options for the child with IVIG resistance include a 2nd dose of
IVIG (2 g/kg), a tapering course of corticosteroids, and/or infliximab (Table
191.4 ). For the most severely affected patients with enlarging coronary
aneurysms, additional therapies such as cyclosporine or cyclophosphamide may
be administered, with consultation from specialists in pediatric rheumatology
and cardiology.
Table 191.4
Treatment Options for IVIG-Resistant Patients With
Kawasaki Disease*
AGENT DESCRIPTION DOSE
MOST FREQUENTLY ADMINISTERED
IVIG: 2nd infusion Pooled polyclonal 2 g/kg IV
IG
IVIG + IVIG + IVIG: 2 g/kg IV + prednisolone 2 mg⋅kg−1 ⋅d−1 IV divided every 8 hr
prednisolone corticosteroid until afebrile, then prednisone orally until CRP normalized, then taper
over 2-3 wk
Infliximab Monoclonal Single infusion: 5 mg/kg IV given over 2 hr
antibody against
TNF-α
ALTERNATIVE TREATMENTS
Cyclosporine Inhibitor of IV: 3 mg⋅kg−1 ⋅d−1 divided every 12 hr
calcineurin-NFAT PO: 4-8 mg⋅kg−1 ⋅d−1 divided every 12 hr
pathway Adjust dose to achieve trough 50-150 ng/mL; 2 hr peak level 300-
600 ng/mL
Anakinra Recombinant IL- 2-6 mg⋅kg−1 ⋅d−1 given by subcutaneous injection
1β receptor
antagonist
Cyclophosphamide Alkylating agent 2 mg⋅kg−1 ⋅d−1 IV
blocks DNA
replication
Plasma exchange Replaces plasma Not applicable
with albumin
* IVIG resistance is defined as persistent or recrudescent fever at least 36 hours and <7 days after
completion of 1st IVIG infusion. The top 3 treatments have been most frequently used, although
no comparative effectiveness trial has been performed. Pulsed high-dose corticosteroid treatment
is not recommended. The alternative treatments have been used in a limited number of patients
with KD.
CRP, C-reactive protein; IG, immunoglobulin; IL, interleukin; IV, intravenous(ly); IVIG, intravenous
immune globulin; NFAT, nuclear factor of activated T cells; PO, oral; TNF, tumor necrosis factor.
FIG. 192.1 Distribution of vessel involvement in large, medium, and small vessel
vasculitis. There is substantial overlap with respect to arterial involvement, and all 3
major categories of vasculitis can affect any-size artery. Large vessel vasculitis affects
large arteries more often than other vasculitides. Medium vessel vasculitis
predominantly affects medium arteries. Small vessel vasculitis predominantly affects
small vessels, but medium arteries and veins may be affected, although immune
complex small vessel vasculitis rarely affects arteries. Not shown is variable vessel
vasculitis , which can affect any type of vessel, from aorta to veins. The diagram
depicts (from left to right ) aorta, large artery, medium artery, small artery/arteriole,
capillary, venule, and vein. ANCA, Antineutrophil cytoplasmic antibody; GBM,
glomerular basement membrane. (From Jennette JC, Falk RJ, Bacon PA, et al: 2012
Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides,
Arthritis Rheum 65(1):1–11, 2013, Fig 2, p 4.)
Table 192.1
Classification of Childhood Vasculitis
2012 Chapel Hill Consensus EUROPEAN LEAGUE AGAINST RHEUMATISM/PEDIATRIC
Conference Nomenclature of RHEUMATOLOGY EUROPEAN SOCIETY Classification of
Vasculitides Childhood Vasculitis
I. Large vessel vasculitis Predominantly large vessel vasculitis
Takayasu arteritis Takayasu arteritis
Giant cell arteritis Predominantly medium vessel vasculitis
II. Medium vessel vasculitis Childhood polyarteritis nodosa
Polyarteritis nodosa Cutaneous polyarteritis nodosa
Kawasaki disease Kawasaki disease
III. Small vessel vasculitis Predominantly small vessel vasculitis
Antineutrophil cytoplasmic Granulomatous:
antibody (ANCA)–associated • Granulomatosis with polyangiitis (Wegener granulomatosis)*
vasculitis • Eosinophilic granulomatosis with polyangiitis (Churg-Strauss
• Microscopic polyangiitis syndrome)*
• Granulomatosis with Nongranulomatous:
polyangiitis • Microscopic polyangiitis*
• Eosinophilic • Henoch-Schönlein purpura (IgA vasculitis)
granulomatosis with • Isolated cutaneous leukocytoclastic vasculitis
polyangiitis • Hypocomplementemic urticarial vasculitis
Immune complex small vessel Other vasculitides
vasculitis Behçet disease
• Anti–glomerular basement Vasculitis secondary to infection (including hepatitis B–associated
membrane (anti-GBM) polyarteritis nodosa), malignancies, and drugs (including
disease hypersensitivity vasculitis)
• IgA vasculitis (Henoch- Vasculitis associated with connective tissue disease
Schönlein purpura) Isolated vasculitis of central nervous system
• Hypocomplementemic Cogan syndrome
urticarial vasculitis Unclassified
IV. Variable vessel vasculitis
Behçet disease
Cogan syndrome
V. Single-organ vasculitis
Cutaneous leukocytoclastic
vasculitis
Cutaneous arteritis
Primary central nervous system
vasculitis
Isolated aortitis
Others
VI. Vasculitis associated with
systemic disease
Lupus vasculitis
Rheumatoid vasculitis
Sarcoid vasculitis
Others
VII. Vasculitis associated with
probable etiology
Hepatitis C virus–associated
cryoglobulinemic vasculitis
Hepatitis B virus–associated
vasculitis
Syphilis-associated aortitis
Drug-associated immune
complex vasculitis
Drug-associated ANCA-
associated vasculitis
Cancer-associated vasculitis
Others
Adapted from Jennette JC, Falk RJ, Bacon PA, et al: 2012 Revised International
Chapel Hill Consensus Conference nomenclature of vasculitides, Arthritis
Rheum 65:1–11; 2013; and Ozen S, Pistorio A, Iusan SM, et al:
EULAR/PRINTO/PRES criteria for Henoch-Schönlein purpura, childhood
polyarteritis nodosa, childhood Wegener granulomatosis and childhood Takayasu
arteritis: Ankara 2008. Part II. Final classification criteria, Ann Rheum Dis
69:798–806; 2010.
Table 192.2
Features That Suggest a Vasculitic Syndrome
Clinical Features
Laboratory Features
From Petty RE, Laxer RM, Lindsley CB, Wedderburn LR: Textbook of pediatric
rheumatology, ed 7, Philadelphia, 2016, Elsevier Saunders.
Table 192.3
Clinicopathologic Characteristics of Vasculitides in
Childhood
VESSELS
SYNDROME FREQUENCY CHARACTERISTIC PATHOLOGY
AFFECTED
Polyarteritis
Polyarteritis nodosa Rare Medium-size and Focal segmental (often near bifurcations);
small muscular fibrinoid necrosis; gastrointestinal, renal
arteries and microaneurysms; lesions at various stages of
sometimes arterioles evolution
Kawasaki disease Common Coronary and other Thrombosis, fibrosis, aneurysms, especially
muscular arteries of coronary vessels
Leukocytoclastic Vasculitis
Henoch-Schönlein purpura Common Arterioles and Leukocytoclasis; mixed cells, eosinophils,
(IgA vasculitis) venules, often small IgA deposits in affected vessels
arteries and veins
Hypersensitivity angitis Rare Arterioles and Leukocytoclastic or lymphocytic, varying
venules eosinophils, occasionally granulomatous;
widespread lesions at same stage of
evolution
Granulomatous Vasculitis
Granulomatosis with Rare Small arteries and Upper and lower respiratory tract,
polyangiitis (Wegener veins, occasionally necrotizing granulomata glomerulonephritis
granulomatosis) larger vessels
Eosinophilic Rare Small arteries and Necrotizing extravascular granulomata; lung
granulomatosis with veins, often involvement; eosinophilia
polyangiitis (Churg- arterioles and
Strauss syndrome) venules
Giant Cell Arteritis
Takayasu arteries Uncommon Large arteries Granulomatous inflammation, giant cells;
aneurysms, dissection
Temporal arteritis Rare Medium-size and Granulomatous inflammation, giant cell
large arteries arteries
Adapted from Cassidy JT, Petty RE: Textbook of pediatric rheumatology, ed 6, Philadelphia, 2011,
Elsevier Saunders.
Diagnosis
The diagnosis of HSP is clinical and often straightforward when the typical rash
is present. However, in at least 25% of cases, the rash appears after other
manifestations, making early diagnosis challenging. Table 192.4 summarizes the
EULAR/PRES classification criteria for HSP. Most patients are afebrile.
Table 192.4
Classification Criteria for Henoch-Schönlein
Purpura*
European League Against Rheumatism/Pediatric Rheumatology
European Society Criteria †
* Classification criteria are developed for use in research and not validated for
clinical diagnosis.
† Developed for use in pediatric populations only.
Diagnosis
Specific pediatric criteria for TA have been proposed (Table 192.5 ).
Radiographic demonstration of large vessel vasculitis is necessary . A thorough
physical examination is required to detect an aortic murmur, diminished or
asymmetric pulses, and vascular bruits. Four extremity blood pressures should
be measured; >10 mm Hg asymmetry in systolic pressure is indicative of
disease.
Table 192.5
Proposed Classification Criteria for Pediatric-
Onset Takayasu Arteritis
Angiographic abnormalities (conventional, CT, or magnetic resonance
angiography) of the aorta or its main branches and at least 1 of the
following criteria:
• Decreased peripheral artery pulse(s) and/or claudication of
extremities
• Blood pressure difference between arms or legs of >10 mm Hg
• Bruits over the aorta and/or its major branches
• Hypertension (defined by childhood normative data)
• Elevated acute-phase reactant (erythrocyte sedimentation rate or
C-reactive protein)
Differential Diagnosis
In the early phase of TA, when nonspecific symptoms predominate, the
differential diagnosis includes a wide array of systemic infections, autoimmune
Clinical Manifestations
The clinical presentation of PAN is variable but generally reflects the
distribution of inflamed vessels. Constitutional symptoms are present in most
children at disease onset. Weight loss and severe abdominal pain suggest
mesenteric arterial inflammation and ischemia. Renovascular arteritis can cause
hypertension, hematuria, or proteinuria, although glomerulonephritis is not
typical. Cutaneous manifestations include purpura, livedo reticularis, ulcerations,
digital ischemia, and painful nodules. Arteritis affecting the nervous system can
result in cerebrovascular accidents, transient ischemic attacks, psychosis, and
ischemic motor or sensory peripheral neuropathy (mononeuritis multiplex ).
Myocarditis or coronary arteritis can lead to heart failure and myocardial
ischemia; pericarditis and arrhythmias have also been reported. Arthralgias,
arthritis, or myalgias are frequently present. Less common symptoms include
testicular pain that mimics testicular torsion, bone pain, and vision loss as a
result of retinal arteritis. The pulmonary vasculature is usually spared in PAN.
Diagnosis
The diagnosis of PAN requires demonstration of vessel involvement on biopsy
or angiography (Table 192.6 ). Biopsy of cutaneous lesions shows small or
medium vessel vasculitis (see Fig. 192.6 ). Kidney biopsy in patients with renal
manifestations may show necrotizing arteritis. Electromyography in children
with peripheral neuropathy identifies affected nerves, and sural nerve biopsy
may reveal vasculitis. Conventional arteriography is the gold standard diagnostic
imaging study for PAN and reveals areas of aneurysmal dilation and segmental
stenosis, the classic “beads on a string” appearance (Fig. 192.7 ). MRA and
CTA, less invasive imaging alternatives, are gaining acceptance, but may not be
as effective in identifying small vessel disease or in younger children.
Table 192.6
FIG. 192.7 Child with polyarteritis nodosa. Abdominal aortogram shows bilateral renal
artery aneurysms (arrows) , superior mesenteric artery aneurysm (asterisk) , and left
common iliac artery occlusion (arrowhead) . (Courtesy of Dr. M. Hogan.)
Differential Diagnosis
Early skin lesions may resemble those of HSP, although the finding of nodular
affected tissues, thus the term pauci-immune vasculitis . ANCA-associated
vasculitis is categorized into 3 distinct forms: granulomatosis with polyangiitis
(GPA) , formerly Wegener granulomatosis; microscopic polyangiitis (MPA) ;
and eosinophilic granulomatosis with polyangiitis , formerly Churg-Strauss
syndrome (CSS ) (see Table 192.1 ).
Epidemiology
GPA is a necrotizing granulomatous small and medium vessel vasculitis that
occurs at all ages and targets the upper and lower respiratory tracts and the
kidneys. Although most cases of GPA occur in adults, the disease also occurs in
children with a mean age at diagnosis of 14 yr. There is a female predominance
of 3-4 : 1, and pediatric GPA is most prevalent in Caucasians.
MPA is a small vessel necrotizing vasculitis with clinical features similar to
those of GPA, but without granulomas and upper airway involvement. CSS is a
small vessel necrotizing granulomatous (allergic granulomatosis) vasculitis
associated with a history of refractory asthma and peripheral eosinophilia. MPA
and CSS are rare in children, and there does not appear to be a gender
predilection in either disease.
Pathology
Necrotizing vasculitis is the cardinal histologic feature in both GPA and MPA.
Kidney biopsies typically demonstrate crescentic glomerulonephritis with little
or no immune complex deposition (“pauci-immune”), in contrast to biopsies
from patients with SLE. Although granulomatous inflammation is common in
GPA and CSS, it is typically not present in MPA. Biopsies showing perivascular
eosinophilic infiltrates distinguish CSS syndrome from both MPA and GPA
(Table 192.7 ).
Table 192.7
Differential Diagnostic Features of Small Vessel Vasculitis
HENOCH- CHURG-
GRANULOMATOSIS MICROSCOPIC
FEATURE SCHÖNLEIN STRAUSS
WITH POLYANGIITIS POLYANGIITIS
PURPURA SYNDROME*
Signs and symptoms of + + + +
small vessel vasculitis †
IgA-dominant immune + − − −
deposits
Circulating − + (PR3) + (MPO > PR3) + (MPO)
antineutrophil
cytoplasmic antibodies
Necrotizing vasculitis − + + +
Granulomatous − + + −
inflammation
Asthma and eosinophilia − − + −
*
Eosinophilic granulomatosis with polyangiitis.
† Signs and symptoms of small vessel vasculitis include purpura, other rash, arthralgias, arthritis,
Pathogenesis
The etiology of ANCA-associated vasculitis remains unknown, although
neutrophils, monocytes, and endothelial cells are involved in disease
pathogenesis. Neutrophils and monocytes are activated by ANCAs, specifically
by the ANCA-associated antigens proteinase-3 (PR3) and myeloperoxidase
(MPO), and release proinflammatory cytokines such as TNF-α and IL-8.
Localization of these inflammatory cells to the endothelium results in vascular
damage characteristic of the ANCA vasculitides. Why the respiratory tract and
kidneys are preferential targets in GPA and MPA is unknown.
Clinical Manifestations
Early disease course is characterized by nonspecific constitutional symptoms,
including fever, malaise, weight loss, myalgias, and arthralgias. In GPA, upper
airway involvement can manifest as sinusitis, nasal ulceration, epistaxis, otitis
media, and hearing loss. Lower respiratory tract symptoms in GPA include
cough, wheezing, dyspnea, and hemoptysis. Pulmonary hemorrhage can cause
rapid respiratory failure. Compared with adults, childhood GPA is more
frequently complicated by subglottic stenosis (Fig. 192.8 ). Inflammation-
induced damage to the nasal cartilage can produce a saddle nose deformity (Fig.
192.8 ). Ophthalmic involvement includes conjunctivitis, scleritis, uveitis, optic
neuritis, and invasive orbital pseudotumor (causing proptosis). Perineural
vasculitis or direct compression on nerves by granulomatous lesions can cause
cranial and peripheral neuropathies. Hematuria, proteinuria, and hypertension in
GPA signal renal disease. Cutaneous lesions include palpable purpura and ulcers.
Venous thromboembolism is a rare but potentially fatal complication of GPA.
The frequencies of organ system involvement throughout the disease course in
GPA follow: respiratory tract, 74%; kidneys, 83%; joints, 65%; eyes, 43%; skin,
47%; sinuses, 70%; and nervous system, 20%. Table 192.8 lists the classification
criteria for pediatric-onset GPA.
FIG. 192.8 Adolescent girl with granulomatosis with polyangiitis. A and B, Anterior and
lateral views of saddle nose deformity. C, Segment of subglottic posterior tracheal
irregularity (between arrows ) on lateral neck radiograph.
Table 192.8
EULAR/PReS Classification Criteria for
Pediatric-Onset Granulomatosis with
Polyangiitis*
Diagnosis
GPA should be considered in children who have recalcitrant sinusitis, pulmonary
infiltrates, and evidence of nephritis. Chest radiography often fails to detect
pulmonary lesions, and chest CT may show nodules, ground-glass opacities,
mediastinal lymphadenopathy, and cavitary lesions (Fig. 192.9 ). The diagnosis
is confirmed by the presence of c-ANCA with anti-PR3 specificity (PR3-
ANCAs) and the finding of necrotizing granulomatous vasculitis on pulmonary,
sinus, or renal biopsy. The ANCA test result is positive in approximately 90% of
children with GPA, and the presence of anti-PR3 increases the specificity of the
test.
(small vessels with neutrophilic perivascular or extravascular neutrophilic
infiltration) (Table 192.9 ). Hypocomplementemic urticarial vasculitis
involves small vessels and manifests as recurrent urticaria that resolves over
several days but leaves residual hyperpigmentation. This condition is associated
with low levels of complement component C1q and systemic findings that
include fever, GI symptoms, arthritis, and glomerulonephritis. Some patients
with urticarial vasculitis have normal complement levels. Cryoglobulinemic
vasculitis can complicate mixed essential cryoglobulinemia and is a small vessel
vasculitis affecting skin, joints, kidneys, and lungs.
Table 192.9
these criteria are present. The presence of ≥3 criteria has a diagnostic sensitivity of 71.0% and
specificity of 83.9%. The age criterion is not applicable for children.
Adapted from Calabrese LH, Michel BA, Bloch DA, et al: The American College of Rheumatology
1990 criteria for the classification of hypersensitivity vasculitis, Arthritis Rheum 33:1108–1113,
1990 (Table 2, p 1110); and Textbook of pediatric rheumatology, ed 7, Philadelphia, 2016, Elsevier
(Table 38.2, p 511).
Other
• Drug-induced vasculitis
• Malignancy-associated vasculitis
• Rasmussen encephalitis
Other
Noninflammatory Vasculopathies
Modified from Gowdie P, Twilt M, Benseler SM: Primary and secondary central
nervous system vasculitis. J Child Neurol 27:1448–1459, 2012.
Table 192.11
Bibliography
Abril A. Churg-Strauss syndrome: an update. Curr Rheumatol
Rep . 2011;13:489–495.
Benseler SM. Central nervous system vasculitis in children.
Curr Rheumatol Rep . 2006;8:442–449.
Cassidy JT, Petty RE. Systemic vasculitis. Textbook of pediatric
rheumatology . ed 5. Elsevier Saunders: Philadelphia; 2005.
Dedeoglu F, Sundel RP. Vasculitis in children. Rheum Dis Clin
North Am . 2007;33:555–583.
Gray PEA, Bock A, Ziegler DS, et al. Neonatal Sweet
syndrome: a potential marker of serious systemic illness.
Pediatrics . 2012;129(5):e1333–e1359.
Gowdie P, Twitt M, Benseler SM. Primary and secondary
central nervous system vasculitis. J Child Neurol .
2012;27(11):1448–1459.
Van Mater H. Pediatric inflammatory brain diseases: a
diagnostic approach. Curr Opin Rheumatol . 2014;26(5):553–
561.
Bibliography
Barut K, Sahin S, Kasapcopur O. Pediatric vasculitis. Curr Opin
Rheumatol . 2016;28(1):29–38.
Eleftheriou D, Brogan PA. Therapeutic advances in the
Subsequent, repeated physical examinations of children with musculoskeletal
pain complaints may reveal eventual development and manifestations of
rheumatic or other diseases. The need for additional testing should be
individualized, depending on the specific symptoms and physical findings.
Laboratory screening and radiography should be pursued if there is suspicion of
certain underlying disease processes. Possible indicators of a serious, vs a
benign, cause of musculoskeletal pain include pain present at rest, pain that may
be relieved by activity, objective joint swelling on physical examination,
stiffness or limited range of motion in joints, bony tenderness, muscle weakness,
poor growth and/or weight loss, and constitutional symptoms (e.g., fever,
malaise) (Table 193.1 ). In the case of laboratory screenings, a complete blood
count (CBC) and erythrocyte sedimentation rate (ESR) are likely to be abnormal
in children whose pain is secondary to a bone or joint infection, SLE, or a
malignancy. Bone tumors, fractures, and other focal pathology resulting from
infection, malignancy, or trauma can often be identified through imaging studies,
including plain radiographs, MRI, and less often technetium-99m bone scans.
Table 193.1
CBC, Complete blood count; CRP, C-reactive protein level; ESR, erythrocyte sedimentation rate.
Adapted from Malleson PN, Beauchamp RD: Diagnosing musculoskeletal pain in children, CMAJ
165:183–188, 2001.
Table 193.2
Common Musculoskeletal Pain Syndromes in Children by
Anatomic Region
Treatment
following morning (Table 193.3 ). Pain often follows a day with exercise or
other physical activities. Physical findings are normal, and gait is not impaired.
Table 193.3
Inclusion and Exclusion Criteria for Growing Pains
Including Features of Restless Leg Syndromes (RLS)
FIG. 193.1 Fibromyalgia questionnaire. American College of Rheumatology criteria.
IBS, Irritable bowel syndrome. (Adapted from Wolfe F, Clauw DJ, Fitzcharles MA, et al.
The American College of Rheumatology preliminary diagnostic criteria for fibromyalgia
and measurement of symptom severity. Arthritis Care Res 62:600–610, 2010.)
Table 193.4
American College of Rheumatology
Fibromyalgia Diagnostic Criteria
The following 3 conditions must be met:
1. Widespread pain index (WPI) ≥7 and symptom severity (SS) scale score
≥5 or WPI 3-6 and SS scale score ≥9.
2. Symptoms have been present at a similar level for at least 3 mo.
3. The patient does not have a disorder that would otherwise explain the
pain.
Ascertainment of WPI
The WPI is the number of areas in which a patient has had pain over the last
week. The score will be between 0 and 19: left shoulder girdle left, right
shoulder girdle, left upper arm, right upper arm, left lower arm, right lower arm,
left hip (buttock, trochanter), right hip (buttock, trochanter), left upper leg, right
upper leg, left lower leg, right lower leg, left jaw, right jaw, chest, abdomen,
upper back, lower back, and neck.
Ascertainment of Ss Scale Score
The SS scale score is the sum of the severity of 3 symptoms (fatigue, waking
unrefreshed, and cognitive symptoms) plus the severity of somatic symptoms in
general. The final score is between 0 and 12.
• For each of the 3 symptoms, the level of severity over the past week is rated
using the following scale:
0 = No problem
1 = Slight or mild problems, generally mild or intermittent
2 = Moderate, considerable problems, often present and/or at a
moderate level
3 = Severe: pervasive, continuous, life-disturbing problems
• Considering somatic symptoms in general, the following scale is used to
indicated the number of symptoms:
0 = No symptoms
1 = Few symptoms
2 = Moderate number of symptoms
3 = Great deal of symptoms
• Somatic symptoms that can be considered include muscle pain, irritable
bowel syndrome, fatigue, thinking problems, muscle weakness, headache,
abdominal pain, numbness/tingling, dizziness, insomnia, depression,
constipation, pain in the upper abdomen, nausea, nervousness, chest pain,
blurred vision, fever, diarrhea, dry mouth, itching, wheezing, Raynaud
phenomenon, hives/welts, ringing in ears, vomiting, heartburn, oral ulcers,
loss of/change in taste, seizures, dry eyes, shortness of breath, loss of
appetite, rash, sun sensitivity, hearing difficulties, easy bruising, hair loss,
frequent urination, painful urination, and bladder spasms.
Adapted from Wolfe F, Clauw DJ, Fitzcharles MA, et al: The American College
of Rheumatology preliminary diagnostic criteria for fibromyalgia and
measurement of symptom severity, Arthritis Care Res 62: 600–610, 2010.
FIG. 193.2 Fibromyalgia tender points.
Although the majority of pediatric patients with CRPS present with a history
of minor trauma or repeated stress injury (e.g., caused by competitive sports), a
sizable proportion are unable to identify a precipitating event. Usual age of onset
is between 8 and 16 yr, and girls outnumber boys with the disease by as much as
6 : 1. Childhood CRPS differs from the adult form in that lower extremities,
rather than upper extremities, are most often affected. The incidence of CRPS in
children is unknown, largely because it is often undiagnosed or diagnosed late,
with the diagnosis frequently delayed by almost 1 yr. Left untreated, CRPS can
have severe consequences for children, including bone demineralization, muscle
wasting, and joint contractures.
An evidence-based approach to the treatment of CRPS continues to suggest a
multistage approach. Aggressive physical therapy (PT) should be initiated as
soon as the diagnosis is made and CBT added as needed. PT is recommended 3-
4 times/wk, and children may need analgesic premedication at the onset,
particularly before PT sessions. PT is initially limited to desensitization and then
moves to weight-bearing, range-of-motion, and other functional activities. CBT
used as an adjunctive therapy targets psychosocial obstacles to fully participating
in PT and provides pain-coping skills training. Sympathetic and epidural nerve
blocks should be attempted only under the auspices of a pediatric pain specialist.
The goal of both pharmacologic and adjunctive treatments for CRPS is to
provide sufficient pain relief to allow the child to participate in aggressive
physical rehabilitation. If CRPS is identified and treated early, the majority of
children and adolescents can be treated successfully with low-dose amitriptyline
(10-50 mg orally 30 min before bedtime), aggressive PT, and CBT interventions.
Opioids and anticonvulsants such as gabapentin can also be helpful. Notably,
multiple studies have shown that noninvasive treatments, particularly PT and
CBT, are at least as efficacious as nerve blocks in helping children with CRPS
achieve resolution of their symptoms.
There is growing evidence that some patients with CRPS I have a small fiber
polyneuropathy (see Chapter 193.2 ).
Bibliography
hydroxychloroquine, colchicine, cyclophosphamide, cyclosporine, and anti–
tumor necrosis factor [TNF] agents), as reported in small series and case reports.
Table 194.1
MAJOR
Typical inflammatory episodes of ear cartilage
Typical inflammatory episodes of nose cartilage
Typical inflammatory episodes of laryngotracheal cartilage
MINOR
Eye inflammation (conjunctivitis, keratitis, episcleritis, uveitis)
Hearing loss
Vestibular dysfunction
Seronegative inflammatory arthritis
*
The diagnosis is established by the presence of 2 major or 1 major and 2 minor
criteria. Histologic examination of affected cartilage is required when the
presentation is atypical.
Data from Michet CJ Jr, McKenna CH, Luthra HS, et al: Relapsing
polychondritis: survival and predictive role of early disease manifestations, Ann
Intern Med 104:74-78, 1986.
Mucha-Habermann Disease/Pityriasis
Lichenoides Et Varioliformis Acuta
Pityriasis lichenoides et varioliformis acuta (PLEVA ) is a benign, self-limited
cutaneous vasculitis characterized by episodes of macules, papules, and
papulovesicular lesions that can develop central ulceration, necrosis, and
crusting (Fig. 194.1 ). Different stages of development are usually seen at once.
PLEVA fulminans or febrile ulceronecrotic Mucha-Habermann disease
(FUMHD ) is the severe, life threatening form of PLEVA. Large, coalescing,
ulceronecrotic lesions are seen, accompanied by high fever and elevated
erythrocyte sedimentation rate (ESR). Systemic manifestations can include
interstitial pneumonitis, abdominal pain, malabsorption, arthritis, and neurologic
manifestations. PLEVA has a male predominance and occurs more frequently in
childhood. The diagnosis is confirmed by biopsy of skin lesions, which reveals
perivascular and intramural lymphocytic inflammation affecting capillaries and
venules in the upper dermis that may lead to keratinocyte necrosis. When disease
is severe, corticosteroids have been used with questionable effect, and
methotrexate has been reported to induce rapid remission in resistant cases.
Cyclosporine and anti-TNF agents have also been efficacious in case reports.
FIG. 194.1 Pityriasis lichenoides et varioliformis acuta (PLEVA). Symmetric, oval and
round, reddish brown macular, popular, necrotic, and crusted lesions on chest of 9 yr
old boy. (From Paller AS, Mancini AJ, editors: Hurwitz clinical pediatric dermatology, ed
5, Philadelphia, 2016, Elsevier, Fig 4-33, p 87.)
Sweet Syndrome
Sweet syndrome, or acute febrile neutrophilic dermatosis , is a rare entity in
children. It is characterized by fever, elevated neutrophil count, and raised,
tender erythematous plaques and nodules over the face, extremities, and trunk.
Skin biopsy reveals neutrophilic perivascular infiltration in the upper dermis.
Female predominance is seen in the adult population, whereas gender
distribution is equal in children. Established criteria are useful for diagnosis
(Table 194.2 ). Children can also have arthritis, sterile osteomyelitis, myositis,
and other extracutaneous manifestations. Sweet syndrome may be idiopathic or
secondary to malignancy (particularly acute myelogenous leukemia), drugs
(granulocyte colony-stimulating factor, tretinoin or trimethoprim-
sulfamethoxazole), or rheumatic diseases (Behçet disease, antiphospholipid
antibody syndrome, systemic lupus erythematosus). The condition usually
responds to treatment with corticosteroids, treatment of underlying disease, or
removal of associated medication.
Table 194.2
Diagnostic Criteria for Classic Sweet
Syndrome*
Major Criteria
Minor Criteria
Pyrexia >38°C
Association with underlying hematologic or visceral malignancy,
inflammatory disease or pregnancy, or preceded by an upper respiratory
or gastrointestinal infection or vaccination
Excellent response to systemic corticosteroids or potassium iodide
Abnormal laboratory values at presentation (3 of 4):
Erythrocyte sedimentation rate >20 mm/hr
Positive C-reactive protein test result
>8,000 leukocytes/mm3
>70% neutrophils/mm3
minor criteria.
Microscopy
The Gram stain is an extremely valuable diagnostic technique to provide rapid
and inexpensive information regarding the absence or presence of inflammatory
cells and organisms in clinical specimens. For some specimen types, the
presence of inflammatory and epithelial cells is used to judge the suitability of a
specimen for culture. For example, the presence of >10 epithelial cells per low-
power field in a sputum specimen is highly suggestive of a specimen
contaminated with oral secretions. In addition, a preliminary assessment of the
etiologic agent can be made based on the morphology (e.g., cocci vs rods) and
stain reaction (e.g., gram-positive isolates are purple; gram-negative isolates are
red) of the microorganisms. However, a negative Gram stain does not rule out
infection, since 104 to 105 microorganisms per milliliter (mL) in the specimen
are required for detection by this method.
In addition to the Gram stain, many other stains are used in microbiology, both
to detect organisms and to help infer their identity (Table 195.1 ).
Table 195.1
Stains Used for Microscopic Examination
TYPE OF
CLINICAL USE
STAIN
Gram stain Stains bacteria (with differentiation of gram-positive and gram-negative organisms), fungi,
leukocytes, and epithelial cells.
Potassium A 10% solution dissolves cellular and organic debris and facilitates detection of fungal elements
hydroxide in clinical specimens.
(KOH)
Calcofluor Nonspecific fluorochrome that binds to cellulose and chitin in fungal cell walls, can be combined
white stain with 10% KOH to dissolve cellular material.
Ziehl-Neelsen Acid-fast stains, using basic carbolfuchsin, followed by acid-alcohol decolorization and
and Kinyoun methylene blue counterstaining.
stains Acid-fast organisms (e.g., Mycobacterium ) resist decolorization and stain pink.
A weaker decolorizing agent is used for partially acid-fast organisms (e.g., Nocardia,
Cryptosporidium, Cyclospora, Isospora ).
Auramine- Acid-fast stain using fluorochromes that bind to mycolic acid in mycobacterial cell walls and
rhodamine resist acid-alcohol decolorization; usually performed directly on clinical specimens.
stain Acid-fast organisms stain orange-yellow against a black background.
Acridine Fluorescent dye that intercalates into DNA, used to aid in differentiation of organisms from
orange stain debris during direct specimen examination, and also for detection of organisms that are not
visible with Gram stain.
Bacteria and fungi stain orange, and background cellular material stains green.
Lugol iodine Added to wet preparations of fecal specimens for ova and parasites to enhance contrast of the
stain internal structures (nuclei, glycogen vacuoles).
Wright and Primarily for detecting blood parasites (Plasmodium, Babesia, and Leishmania ), detection of
Giemsa stains amoeba in preparations of cerebrospinal fluid, and fungi in tissues (yeasts, Histoplasma )
Trichrome Stains stool specimens for identification of protozoa.
stain
Direct Used for direct detection of a variety of organisms in clinical specimens by using specific
fluorescent- fluorescein-labeled antibodies (e.g., Pneumocystis jiroveci, many viruses).
antibody stain
Table 195.2
Multiplex Molecular Assays for Viral Diagnosis
AdV, Adenovirus; AH1, influenza A, hemagglutinin type 1; AH3, influenza A, hemagglutinin type 3;
CMV, cytomegalovirus; CoV, coronavirus; EV, enterovirus; flu A, influenza A; flu B, influenza B;
CHAPTER 197
Immunization Practices
Henry H. Bernstein, Alexandra Kilinsky, Walter A. Orenstein
Table 197.1a
Comparison of 20th Century Annual Morbidity and Current
Morbidity: Vaccine-Preventable Diseases
comparisons of morbidity and mortality for vaccine-preventable diseases in the United States,
JAMA 298(18):2155–2163, 2007.
†
Data from Centers for Disease Control and Prevention: Notifiable diseases and mortality tables,
MMWR 66(52):ND-924–ND-941, 2018.
‡
Hib <5 yr of age. An additional 237 cases of Haemophilus influenzae (<5 yr of age) have been
reported with unknown serotype.
Table 197.1b
Comparison of Pre–Vaccine Era Estimated Annual
Morbidity With Current Estimate: Vaccine-Preventable
Diseases
comparisons of morbidity and mortality for vaccine-preventable diseases in the United States,
JAMA 298(18):2155–2163, 2007.
† Data from Centers for Disease Control and Prevention: Viral hepatitis surveillance—United
States, 2016.
‡ Data from Centers for Disease Control and Prevention: Prevention of rotavirus gastroenteritis
among infants and children: recommendations of the Advisory Committee on Immunization
Practices, MMWR Recomm Rep 58(RR-2):1–25, 2009.
§ Data from Centers for Disease Controls and Prevention: Active bacterial core surveillance, 2016
(unpublished).
ǁ
Data from New Vaccine Surveillance Network 2017 data: U.S. rotavirus disease now has biennial
pattern (unpublished).
¶ Data from Centers for Disease Control and Prevention: Varicella Program, 2017 (unpublished).
The major indications for inducing passive immunity are immunodeficiencies
in children with B-lymphocyte defects who have difficulty making antibodies
(e.g., hypogammaglobulinemia, secondary immunodeficiencies), who have
exposure to infectious diseases or to imminent risk of exposure when there is
inadequate time for them to develop an active immune response to a vaccine
(e.g., newborn exposed to maternal hepatitis B), and who have infectious
diseases that require antibody administration as part of the specific therapy
(Table 197.2 ).
Table 197.2
Immunoglobulin and Animal Antisera Preparations
Active Immunization
Vaccines are defined as whole or parts of microorganisms administered to
prevent an infectious disease. Vaccines can consist of whole inactivated
microorganisms (e.g., polio, hepatitis A), parts of the organism (e.g., acellular
pertussis, HPV, hepatitis B), polysaccharide capsules (e.g., pneumococcal and
meningococcal polysaccharide vaccines), polysaccharide capsules conjugated to
protein carriers (e.g., Hib, pneumococcal, and meningococcal conjugate
vaccines), live-attenuated microorganisms (e.g., measles, mumps, rubella,
varicella, rotavirus, and live-attenuated influenza vaccines), and toxoids (e.g.,
tetanus, diphtheria) (Table 197.3 ). A toxoid is a bacterial toxin modified to be
nontoxic but still capable of inducing an active immune response against the
toxin.
Table 197.3
† There are various types of inactivated flu vaccines—IIV3, IIV4, RIV4, ccIIV4, aIIV3.
Data from US Food and Drug Administration: Vaccines licensed for use in the United States.
http://www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm093833.htm .
Table 197.4
Serum antibodies may be detected as soon as 7-10 days after initial injection
of antigen. Early antibodies are usually of the IgM class that can fix
complement. IgM antibodies tend to decline as IgG antibodies increase. The IgG
antibodies tend to peak approximately 1 mo after vaccination and with most
vaccines persist for some time after a primary vaccine course. Secondary or
booster responses occur more rapidly and result from rapid proliferation of
memory B and T lymphocytes.
Assessment of the immune response to most vaccines is performed by
measuring serum antibodies. Although detection of serum antibody at levels
considered protective after vaccination can indicate immunity, loss of detectable
antibody over time does not necessarily mean susceptibility to disease. Some
vaccines induce immunologic memory, leading to a booster or anamnestic
Vaccine Delivery
To ensure potency, vaccines should be stored at recommended temperatures
before and after reconstitution. A comprehensive resource for providers on
vaccine storage and handling recommendations and best practice strategies is
available (https://www.cdc.gov/vaccines/hcp/admin/storage/index.html ).
Expiration dates should be noted and expired vaccines discarded. Lyophilized
vaccines often have long shelf lives. However, the shelf life of reconstituted
vaccines generally is short, ranging from 30 min for varicella vaccine to 8 hr for
MMR vaccine.
All vaccines have a preferred route of administration, which is specified in
package inserts and in AAP and ACIP recommendations. Most inactivated
vaccines, including DTaP, hepatitis A, hepatitis B, Hib, inactivated influenza
vaccine (IIV ), HPV, PCV13, MCV4, and Tdap, are administered IM. In
contrast, MPSV4 and the more commonly used live-attenuated vaccines (MMR,
MMRV, and varicella) should be dispensed by the SC route. Rotavirus vaccine is
administered orally. IPV and PPS23 (pneumococcal polysaccharide vaccine) can
be given IM or SC. One influenza vaccine, LAIV, when recommended, is
administered intranasally, and another influenza vaccine is administered by the
intradermal route. For IM injections, the anterolateral thigh muscle is the
preferred site for infants and young children. The recommended needle length
varies depending on age and size: inch for newborn infants, 1 inch for infants
2-12 mo old, and inches for older children. For adolescents and adults, the
deltoid muscle of the arm is the preferred site for IM administration with needle
lengths of inches depending on patient size. Most IM injections can be
made with 23-25 gauge needles. For SC injections, needle lengths generally
range from inch with 23-25 gauge needles.
Additional aspects of immunization important for pediatricians and other
healthcare providers are detailed on the websites listed in Table 197.5 .
Table 197.5
Vaccine Websites and Resources
ORGANIZATION WEBSITE
HEALTH PROFESSIONAL ASSOCIATIONS
American Academy of Family Physicians (AAFP) http://www.familydoctor.org/online/famdocen/home.html
American Academy of Pediatrics (AAP) http://www.aap.org/
AAP Childhood Immunization Support Program http://www.aap.org/immunization/
American Association of Occupational Health http://www.aaohn.org/
Nurses (AAOHN)
Table 197.6
Combination Vaccines Licensed and Available in the
United States
Table 197.7
Table 197.8
Recommended Immunizations for International Travel*
LENGTH OF STAY
IMMUNIZATIONS Brief, Long-
<1 Term/Residential,
mo >1 mo
Review and complete age-appropriate childhood and adolescent schedule (see + +
text for details)
• DTaP, poliovirus, pneumococcal, and Haemophilus influenzae type b (Hib) vaccines
may be given at 4 wk intervals if necessary to complete recommended schedule
before departure.
• Influenza
• MMR: 2 additional doses given if <12 mo old at 1st dose
• Meningococcal disease (MenACWY) †
• Rotavirus
• Varicella
• Human papillomavirus (HPV)
• Hepatitis A: 2 additional doses given if <12 mo old at 1st dose ‡ §
• Hepatitis B §
• Tdap
Yellow fever ǁ + +
Typhoid fever ¶ ± +
Rabies** ± ±
Japanese encephalitis †† ± +
Cholera ‡‡ ± ±
* See disease-specific chapters in the Centers for Disease Control and Prevention's Yellow Book
** Indicated for people with high risk for animal exposure (especially to dogs) and for travelers to
countries with endemic infection.
†† For regions with endemic infection (see Health Information for International Travel). For high-
risk activities in areas experiencing outbreaks, vaccine is recommended, even for brief travel.
‡‡ Cholera vaccine (CVD 103-HgR, Vaxchora) is recommended for adult (18-64 yr old) travelers to
an area of active toxigenic V. cholerae O1 transmission.
+, Recommended; ±, consider; DTaP, diphtheria and tetanus toxoids and acellular pertussis.
Data from Centers for Disease Control and Prevention: Travelers' health.
https://wwwnc.cdc.gov/travel .
Table 197.9
Vaccination of Persons With Primary and Secondary
Immune Deficiencies
PRIMARY
CATEGORY SPECIFIC CONTRAINDICATED RISK-SPECIFIC EFFECTIVENESS AN
IMMUNODEFICIENCY VACCINES * RECOMMENDED COMMENTS
VACCINES *
B lymphocyte (humoral) Severe antibody OPV a Annual IIV is the only The effectiveness of
deficiencies (e.g., X-linked Smallpox b vaccine given to any vaccine will be
agammaglobulinemia and LAIV patients receiving IG uncertain if it depend
common variable BCG therapy; routine only on the humoral
immunodeficiency) Yellow fever virus inactivated vaccines response (e.g.,
(YFV) and live- can be given if not PPSV23).
receiving IGIV. IG therapy interferes
bacteria vaccines e with the immune
No data for
response to live
rotavirus vaccines
vaccines MMR and
VAR.
Less severe antibody OPV a Vaccines should All vaccines are
deficiencies (e.g., selective BCG be given as on the probably effective.
IgA deficiency and IgG YFV vaccine annual Immune response
subclass deficiency) Other live vaccines immunization may be attenuated.
d appear to be safe. schedule for
immunocompetent
people. e
PPSV23 should be
given beginning at
2 yr of age. f
T lymphocyte (cell- Complete defects (e.g., All live vaccines c , d , g The only vaccine that All inactivated vaccines
mediated and humoral) SCID, complete DiGeorge should be given if the are probably ineffective.
syndrome) patient is receiving IG
is annual IIV if there is
some residual antibody
protection.
Partial defects (e.g., most All live vaccines c , d , gRoutine Effectiveness of any
patients with DiGeorge inactivated vaccine depends on degr
syndrome, hyper-IgM vaccines should of immune suppression.
syndrome, Wiskott- be given. e
Aldrich syndrome, ataxia- PPSV23 should be
telangiectasia) given beginning at
2 yr of age. f
Interferon (IFN)-γ– All live vaccines for IL- None None
interleukin (IL)-12 axis 12/IL-12R deficiencies,
deficiencies IFN-γ, IFN-α, or STAT1
deficiencies
Complement Persistent complement, None PPSV23 should be All routine vaccines are
properdin, MBL, or factor given beginning at probably effective.
B deficiency; secondary 2 yr of age. f
deficiency because taking MCV series
eculizumab (Solaris) beginning in
infancy. h
MenB series
beginning at 10 yr
of age
Phagocytic function Chronic granulomatous Live-bacteria vaccines c None All inactivated
disease vaccines are safe and
probably effective.
Live-virus vaccines
are probably safe and
effective.
Phagocytic deficiencies MMR, MMRV, OPV, a PPSV23 should be All inactivated vaccines
that are undefined or smallpox, LAIV, YF, all given beginning at are safe and probably
accompanied by defect in bacteria vaccines 2 yr of age. f effective.
T-cell and NK-cell MCV series
dysfunction (e.g., beginning in
Chédiak-Higashi infancy. h
syndrome, leukocyte
adhesion defects,
myeloperoxidase
deficiency)
SECONDARY
SPECIFIC CONTRAINDICATED RISK-SPECIFIC EFFECTIVENESS AND
IMMUNODEFICIENCY VACCINES * RECOMMENDED VACCINES * COMMENTS
HIV/AIDS OPV a PPSV23 should be given Rotavirus vaccine is
Smallpox beginning at 2 yr of age. f recommended on standard
BCG MCV series beginning in schedule.
Combined MMRV infancy. h MMR and VAR are recommende
LAIV Consider Hib (if not for HIV-infected children who ar
Withhold MMR, asymptomatic or only low-level
administered in infancy). l
varicella, and zoster in immunocompromised.
severely All inactivated vaccines may be
immunocompromised effective.
persons.
YF vaccine may have
a contraindication or
precaution depending
on the indicators of
immune function. j
Generalized malignant Live-virus and live- PPSV23 should be given Effectiveness of any vaccine depends
neoplasm, transplantation, bacteria vaccines, beginning at 2 yr of age. f on degree of immune suppression;
autoimmune disease, depending on immune Annual IIV (unless receiving inactivated standard vaccines are
immunosuppressive or status. c , d , m intensive chemotherapy or anti- indicated if not highly
radiation therapy B cell antibodies). immunosuppressed, but doses should
Hib vaccine may be indicated. n be repeated after chemotherapy ends.
Asplenia (functional, LAIV PPSV23 should be given All routine vaccines are probably
congenital anatomic, beginning at 2 yr of age. f effective.
surgical) MCV series beginning in
infancy. h
MenB series beginning at 10 yr
of age.
Hib (if not administered in
infancy) o
Chronic renal disease LAIV PPSV23 should given beginning All routine vaccines are probably
at 2 yr of age. f effective.
HepB is indicated if not
previously immunized.
CNS anatomic barrier None PPSV23 should be given beginning All standard vaccines are indicated.
defect (cochlear implant, at 2 yr of age. f
congenital dysplasia of the
inner ear, persistent CSF
communication with naso-
/oropharynx)
* Other vaccines that are universally or routinely recommended should be given if not
contraindicated.
a
OPV is no longer available in the United States.
b This table refers to contraindications for nonemergency vaccination (i.e., ACIP
recommendations)
c Live-bacteria vaccines: BCG and oral Ty21a Salmonella typhi vaccine.
d Live-virus vaccines: MMR, MMRV, VAR, OPV, LAIV, YF, zoster, rotavirus, and vaccinia
(smallpox). Smallpox vaccine is not recommended for children or the general public.
e Children who are delayed or underimmunized should be immunized with routinely
recommended vaccines, according to age and catch-up schedule.
f PPSV23 is begun at 2 yr or older. If PCV13 is required, PCV13 doses should be administered
first, followed by PPSV23 at least 8 wk later; a 2nd dose of PPSV23 is given 5 yr after the 1st.
g Regarding T-lymphocyte immunodeficiency as a contraindication for rotavirus vaccine, data exist
only for SCID.
h Age and schedule of doses depend on the product; repeated doses are required.
Table 197.10
Standards for Child and Adolescent Immunization
Practices
AVAILABILITY OF VACCINES
Vaccination services are readily available.
Vaccinations are coordinated with other healthcare services and provided in a medical home when possible.
Barriers to vaccination are identified and minimized.
Patient costs are minimized.
ASSESSMENT OF VACCINATION STATUS
Healthcare professionals review the vaccination and health status of patients at every encounter to determine
which vaccines are indicated.
Healthcare professionals assess for and follow only medically accepted contraindications.
EFFECTIVE COMMUNICATION ABOUT VACCINE BENEFITS AND RISKS
Parents or guardians and patients are educated about the benefits and risks of vaccination in a culturally
appropriate manner and in easy-to-understand language.*
Healthcare professionals offer strong and consistent recommendations for all universally recommended vaccines
according to the current immunization schedule. They use presumptive language (e.g., these vaccines are
routine) and deliver this recommendation in the same manner for all vaccines.
Healthcare professionals answer parents' or guardians' and patients' questions thoroughly and emphasize an
unwavering commitment to the recommendation. If parents or guardians and patients are hesitant or refuse,
healthcare professionals persevere and offer the vaccine again at the next most appropriate time.
PROPER STORAGE AND ADMINISTRATION OF VACCINES AND DOCUMENTATION OF
VACCINATIONS
Healthcare professionals follow appropriate procedures for vaccine storage and handling.
Up-to-date, written vaccination protocols are accessible at all locations where vaccines are administered.
Persons who administer vaccines and staff who manage or support vaccine administration are knowledgeable and
receive ongoing education.
Healthcare professionals simultaneously administer as many indicated vaccine doses as possible.
Vaccination records for patients are accurate, complete, and easily accessible.
Healthcare professionals report adverse events following vaccination promptly and accurately to the Vaccine
Adverse Event Reporting System (VAERS) and are aware of a separate program, the National Vaccine Injury
Compensation Program (VICP).
Healthcare professionals and personnel review the immunization timeline with parents or guardians and patients
and schedule follow-up immunization visits before the family leaves the care setting.
All personnel who have contact with patients are appropriately vaccinated and communicate consistent messages
about vaccines.
IMPLEMENTATION OF STRATEGIES TO IMPROVE VACCINATION COVERAGE
Systems are used to remind parents or guardians, patients, and healthcare professionals when vaccinations are due
and to recall those who are overdue.
Office- or clinic-based patient record reviews and vaccination coverage assessments are performed annually.
Healthcare professionals practice community-based approaches.
Healthcare professionals understand cultural needs and disparities of different populations and use the most
effective strategies for these populations.
Most healthcare visits (including acute care or sick visits) are viewed as opportunities to review immunization
records, provide vaccines that are due, and catch up on missed vaccinations.
* Additional resources to help improve immunization rates include the following:
Bibliography
Ahmed F, Temte JL, Campos-Outcalt D, et al. Methods for
developing evidence-based recommendations by the
Standard Precautions
Standard precautions, formerly known as universal precautions , are intended to
protect healthcare workers from pathogens and should be used whenever there is
direct contact with patients. Infected patients are often contagious before
symptoms of disease develop. Asymptomatic infected patients are quite capable
of transmitting infectious agents. Standard precautions involve the use of
barriers—gloves, gowns, masks, goggles, and face shields—as needed, to
prevent transmission of microbes associated with contact with blood and body
fluids (Table 198.1 ).
Table 198.1
Recommendations for Application of Standard Precautions
for Care of All Patients in All Healthcare Settings
COMPONENT RECOMMENDATIONS
Hand hygiene Before and after each patient contact, regardless of whether
gloves are used.
After contact with blood, body fluids, secretions,
excretions, or contaminated items; immediately after
removing gloves; before and after entering patient rooms.
Alcohol-containing antiseptic hand rubs preferred except
when hands are visibly soiled with blood or other
proteinaceous material or if exposure to spores (e.g.,
Clostridium difficile , Bacillus anthracis ) or nonenveloped
viruses (norovirus) is likely to have occurred; in these
cases, soap and water is required.
PERSONAL PROTECTIVE EQUIPMENT (PPE)
Gloves For touching blood, body fluids, secretions, excretions, or
contaminated items; for touching mucous membranes and
nonintact skin.
Employ hand hygiene before and after glove use.
Gown During procedures and patient-care activities when contact of
clothing or exposed skin with blood, body fluids, secretions, or
excretions is anticipated.
Mask, eye protection (goggles), face shield During procedures and patient-care activities likely to
generate splashes or sprays of blood, body fluids, or
secretions, such as suctioning and endotracheal intubation,
to protect healthcare personnel.
For patient protection, use of a mask by the person inserting
an epidural anesthesia needle or performing myelograms
when prolonged exposure of the puncture site is likely to
occur.
Soiled patient-care equipment Handle in a manner that prevents transfer of
microorganisms to others and to the environment.
Wear gloves if equipment is visibly contaminated.
Perform hand hygiene.
ENVIRONMENT
Environmental control Develop procedures for routine care, cleaning, and disinfection
of environmental surfaces, especially frequently touched
surfaces in patient-care areas.
Textiles (linens) and laundry Handle in a manner that prevents transfer of microorganisms to
others and the environment.
PATIENT CARE
Injection practices (use of needles and other Do not recap, bend, break, or manipulate used needles; if
sharps) recapping is required, use a one-handed scoop technique
only.
Use needle-free safety devices when available, placing used
sharps in puncture-resistant container.
Use a sterile, single-use, disposable needle and syringe for
each injection.
Single-dose medication vials preferred when medications
may be administered to more than one patient.
Patient resuscitation Use mouthpiece, resuscitation bag, or other ventilation devices
to prevent contact with mouth and oral secretions.
Patient placement Prioritize for single-patient room if patient is at increased risk
for transmission, is likely to contaminate the environment, is
unable to maintain appropriate hygiene, or is at increased risk
for acquiring infection or developing adverse outcome following
infection.
Respiratory hygiene/cough etiquette (source Instruct symptomatic persons to cover nose/mouth when
containment of infectious respiratory secretions sneezing or coughing; use tissues with disposal in no-touch
in symptomatic patients) beginning at initial receptacles.
point of encounter, such as triage or reception Employ hand hygiene after soiling of hands with respiratory
areas in emergency department or physician secretions.
office Wear surgical mask if tolerated or maintain spatial
separation (>3 ft if possible).
Adapted from Kimberlin DW, Brady MT, Jackson MA, et al, editors: Red Book 2018–2021: Report
of the Committee on Infectious Diseases, ed 31, Elk Grove Village, IL, 2018, American Academy
of Pediatrics, pp 148–150.
Isolation
Isolation of patients infected with transmissible pathogens decreases the risk of
nosocomial transmission of organisms to staff and other patients. The specific
type of isolation depends on the infecting agent and potential route of
transmission. Transmission by contact is the most common mode of pathogen
transmission and involves direct contact with the patient or contact with a
contaminated intermediate object. Contact isolation requires the use of gown
and gloves when in contact with the patient or immediate surroundings.
Transmission by droplets involves the propulsion of infectious large particles
over a short distance (<3 ft), with deposition on another's mucous membranes or
skin. Droplet isolation requires the use of gloves and gowns, as well as masks
and eye guards when closer than 3 ft to the patient. Airborne transmission
occurs by dissemination of evaporated droplet nuclei (≤5 µm) or dust particles
carrying an infectious agent. Airborne infection isolation (AII ) requires the
use of masks and negative pressure air-handling systems to prevent spread of the
infectious agent. In the case of active pulmonary tuberculosis in older children
and adults, severe acute respiratory syndrome (SARS), or avian influenza, the
use of special high-density masks (N-95) or self-contained breathing systems
such as powered air-purifying respirators (PAPRs ) or controlled air-purifying
respirators (CAPRs ) are recommended. Positive pressure HEPA-filtered air-
handling systems are used in some institutions for housing seriously
immunocompromised patients and negative pressure systems for the care of
patients with highly contagious respiratory infections such as Ebola virus.
Standard precautions are indicated for all patients and are appropriate for use
in the clinic as well as the hospital. Additionally, for hospitalized patients,
further transmission-based precautions are indicated for certain infections
(Table 198.2 ). For contact and droplet isolation, single rooms are preferred but
not required. Cohorting children infected with the same pathogen is acceptable,
but the etiologic diagnosis should be confirmed by laboratory methods before
exposing infected children to one another. Transmission-based isolation
precautions should be continued for as long as a patient is considered
contagious.
Table 198.2
Clinical Syndromes and Conditions Warranting Empirical
Transmission-Based Precautions in Addition to Standard
Precautions Pending Confirmation of Diagnosis*
(e.g., neonates and adults with pertussis may not have paroxysmal or severe cough). The
clinician's index of suspicion should be guided by the prevalence of specific conditions in the
community as well as clinical judgment.
‡
The organisms listed are not intended to represent the complete, or even most likely, diagnoses,
but rather possible etiologic agents that require additional precautions beyond standard
precautions, until they can be ruled out.
§ These pathogens include enterohemorrhagic Escherichia coli O157:H7, Shigella spp., hepatitis
A virus, norovirus, rotavirus, Clostridium difficile .
AIIRs, Airborne infection isolation rooms; HIV, human immunodeficiency virus; MRSA, methicillin-
resistant S. aureus ; MSSA, methicillin-susceptible S. aureus ; VHF, viral hemorrhagic fever.
Adapted from Centers for Disease Control and
Preventionwebsite,http://www.cdc.gov/hicpac/2007ip/2007ip_table2.html .
The use of isolation techniques in outpatient settings has not been well
studied. Professional offices should establish procedures to ensure that proper
cleaning, disinfection, and sterilization methods are employed. Many practices
and clinics provide separate waiting areas for sick and well children. Triage of
patients is essential to ensure that contagious children or adults are not present in
waiting areas. Outbreaks of measles and varicella in patients within the waiting
area have been reported where the air exhaust from examination rooms enters
the waiting area. Cleaning the clinic environment is important, especially in
high-touch areas. Toys and items that are shared among patients should be
cleaned between uses; if feasible, disposable toys should be used. Toys
contaminated with blood or body fluids should be autoclaved or discarded.
Additional Measures
Other preventive measures include aseptic technique, catheter care, prudent use
of antibiotics through use of an effective antibiotic stewardship program ,
isolation of contagious patients, periodic cleansing of the environment,
disinfection and sterilization of medical equipment, reporting of infections, safe
handling of needles and other sharp instruments, and establishment of employee
health services. Sterile technique must be used for all invasive procedures,
including catheter placement and manipulation. The use of barrier techniques at
the time of IV catheter placement has reduced the rate of catheter-related
bloodstream infections by half. Appropriate catheter use also includes limiting
the duration and number of catheters employed, scrubbing catheter hubs with
every access, and removing catheters as soon as they become unnecessary.
Surgical Prophylaxis
Surgical antibiotic prophylaxis should be employed when there is a high risk of
postoperative infection or when the consequences of such infection would be
catastrophic. The choice of prophylactic antibiotic depends on the surgical site
and type of surgery. A useful classification of surgical procedures based on
infectious risk recognizes 4 preoperative wound categories: clean wounds, clean-
contaminated wounds, contaminated wounds, and dirty and infected wounds
(Table 198.3 ). The American College of Surgeons, Surgical Infection Society,
and American Academy of Pediatrics have made clinical recommendations
regarding antibiotic prophylaxis.
Table 198.3
Common Surgical Procedures for Which Perioperative
Prophylactic Antibiotics Are Recommended
NON β-
SURGICAL
LIKELY PATHOGENS RECOMMENDED DRUGS LACTAM
PROCEDURE
ALTERNATIVE
CLEAN WOUNDS
Cardiac surgery (e.g., Skin flora, enteric gram- Cefazolin or cefuroxime Clindamycin or
open heart surgery) negative bacilli vancomycin
Vascular surgery
Neurosurgery
Orthopedic surgery
(e.g., joint
replacement)
CLEAN-CONTAMINATED WOUNDS
Head and neck surgery Skin flora, oral Cefazolin + metronidazole, ampicillin- Clindamycin
involving oral cavity or anaerobes, oral sulbactam
pharynx streptococci
Gastrointestinal and Enteric gram-negative Cefazolin + metronidazole, Clindamycin
genitourinary surgery bacilli, anaerobes, gram- cefotetan or piperacillin-
positive cocci sulbactam
If colon is involved, consider
bacterial reduction with oral
neomycin and erythromycin.
CONTAMINATED WOUNDS
Traumatic wounds (e.g., Skin flora Cefazolin Clindamycin or
compound fracture) vancomycin
DIRTY WOUNDS
Appendectomy, penetrating Enteric gram-negative Cefazolin + metronidazole, cefoxitin, Clindamycin +
abdominal wounds, bacilli, anaerobes, gram- cefotetan or ampicillin-sulbactam aminoglycoside
colorectal surgery positive cocci
Adapted from Bratzler DW, Dellinger PD, Olsen KM, et al: Clinical practice guidelines from
antimicrobial prophylaxis in surgery, Am J Health Syst Pharm 70:195–283, 2013.
Table 199.1
Infectious Diseases in the Childcare Setting
INCREASED
DISEASE INCIDENCE WITH
CHILDCARE
Respiratory Tract Infections
Otitis media Yes
Sinusitis Probably
Pharyngitis Probably
Pneumonia Yes
Gastrointestinal Tract Infections
Diarrhea (rotavirus, calicivirus, astrovirus, enteric adenovirus, Giardia lamblia, Yes
Cryptosporidium, Shigella, Escherichia coli O157:H7, and Clostridium difficile )
Hepatitis A Yes
Skin Diseases
Impetigo Probably
Scabies Probably
Pediculosis Probably
Tinea (ringworm) Probably
Invasive Bacteria Infections
Haemophilus influenzae type b No*
Neisseria meningitidis Probably
Streptococcus pneumoniae Yes
Aseptic Meningitis
Enteroviruses Probably
Herpesvirus Infections
Cytomegalovirus Yes
Varicella-zoster virus Yes
Herpes simplex virus Probably
Bloodborne Infections
Hepatitis B Few case reports
HIV No cases reported
Hepatitis C No cases reported
Vaccine-Preventable Diseases
Measles, mumps, rubella, diphtheria, pertussis, tetanus Not established
Polio No
H. influenzae type b No*
Varicella Yes
Rotavirus Yes
* Not in the post–vaccine era; yes in the pre–vaccine era.
Epidemiology
Respiratory tract infections and gastroenteritis are the most common diseases
associated with childcare. These infections occur in children and their household
contacts, as well as childcare workers, and can spread into the community. The
severity of illness caused by a given respiratory and enteric pathogen depends on
the person's underlying health status, the inoculum, and prior exposures to the
pathogen, either by infection or immunization. Hepatitis B virus (HBV)
transmission has been reported rarely in a childcare setting. Transmission of
hepatitis C virus (HCV), hepatitis D virus (HDV), and HIV has not been
reported in a childcare setting. Some organisms, such as hepatitis A virus (HAV),
can cause subclinical disease in young children and produce overt and
sometimes serious disease in older children and adults. Other diseases, such as
otitis media and varicella, usually affect children rather than adults. Several
agents, such as cytomegalovirus and parvovirus B19, can have serious
consequences for the fetuses of pregnant women or for immunocompromised
persons. Because many childcare workers are women of childbearing age, they
should be encouraged to discuss possible risks with their physician if they
become pregnant. Both infections and infestations of the skin and hair may be
acquired through contact with contaminated linens or through close personal
contact, which is inevitable in childcare settings.
Table 200.1
Travel Vaccinations for Children
ROUTE
VACCINE FORMULATION AND SCHEDULE INDICATIONS COMMENTS
DOSE
Hepatitis A Pediatric: Havrix IM; 0.5 Primary series: 2 Children >6 Inactivated vaccine
(GlaxoSmithKline); mL doses, 6-18 mo apart months of age Lifelong protection is
720 EU Booster: currently not likely.
VAQTA (Merck); recommended
25 U
Adult: Havrix IM; 1.0 Primary series: 2 Adults ≥19 yr Inactivated vaccine
(GlaxoSmithKline); mL doses, 6-18 mo apart old Lifelong protection is
1440 EU Booster: currently not likely.
VAQTA (Merck); recommended
50 U
Hepatitis A and Twinrix IM; 1.0 Primary series: 3 Adults ≥18 yr Inactivated vaccine
B (GlaxoSmithKline) mL doses at 0, 1, and 6 old Lifelong protection is
mo likely.
Accelerated schedule: Accelerated schedule
0, 7, and 21 days; 4th is as effective.
dose 12 mo later
Boosters: not needed
Immunoglobulin, Injectable IM Travel up to 1 mo Infants <1 yr old Passive
human duration: 0.1 mL/kg immunizations
Travel up to 2 mo against hepatitis A.
duration: 0.2 mL/kg Its use would require
Travel 2 mo or more: delay of measles and
0.2 mL/kg (repeat varicella vaccinations
every 2 mo) (at least 3 mo).
Japanese Inactivated: Ixiaro IM Primary series: 2 Travel to high- Booster recommendation
encephalitis (Intercell USA) 2 doses at days 0 and 28 risk areas; is extrapolated from
virus (JEV) mo Booster: 1 dose 1 yr prolonged stays recommendation for
to later if exposure to individuals ≥17 yr old.
<3 JEV expected
yr
old:
0.25
mL
≥3
yr
old:
0.5
mL
Meningococcal, Quadrivalent: A, C, Y, SC; 0.5 Primary series: single ≥2 yr old Required for entry to
polysaccharide W135 mL dose Saudi Arabia during
Booster: 5 yr in the Hajj.
persons ≥4 yr old; 2–3 Recommended for
yr in children 2–4 yr travelers visiting
old “meningitis belt” in
sub-Saharan Africa
during dry months.
This vaccine is rarely
used in U.S. since
conjugate vaccines
are more
immunogenic.
Meningococcal, Quadrivalent: ACWY- IM: 0.5 Children 9-23 mo old: Routine Required for entry to
conjugate D: Menactra (Sanofi mL 2 doses, 3 mo apart vaccination in Saudi Arabia during
Pasteur) 2-55 yr old: 1 dose U.S. at ≥11-12 the Hajj.
yr old with Recommended for
recommended travelers visiting
booster 5 yr “meningitis belt” in
later sub-Saharan Africa
during dry months.
This vaccine should
not be used in infants
<9 mo old since it
may interfere with
antibody production
by pneumococcal
conjugate vaccine.
Quadrivalent: ACWY- IM; 0.5 Children initiating Routine Required for entry to
CRM: Menveo mL vaccination at 2 mo: vaccination in Saudi Arabia during
(Novartis) doses at 2, 4, 6, and U.S. at ≥11-12 the Hajj.
12 mo old yr old with Recommended for
Children starting recommended travelers visiting
vaccination at 7-23 booster 5 yr “meningitis belt” in
mo old: 2 doses, with later sub-Saharan Africa
2nd dose after 2 yr old during dry months.
and at least 3 mo after
1st dose
Rabies Inactivated IM; 1.0 Preexposure series: 3 Consider for young
mL doses at days 0, 7, and travelers planning
21 or 28 prolonged stays;
Booster: depends on especially away from
risk category and large urban centers with
serologic testing. adequate medical care
Postexposure: rabies systems and airport.
immune globulin; day
0; vaccines at days 0,
3, 7, and 14; 5th dose
at day 28 is
recommended if host
is
immunocompromised.
Typhoid fever Live-attenuated Ty21a1 Oral 1 capsule every other Persons ≥6 yr If series sequence not
day for 4 doses old completed, all 4
Boosters: every 5 yr doses need to be
repeated.
Contraindicated in
immunocompromised
hosts. Cannot be
taken with hot
beverage. Person
must not be taking
antibiotics.
Injectable IM; 0.5 Primary series: 1 dose Persons ≥2 yr
Polysaccharide Vi mL Booster: every 2 yr old
antigen
Yellow fever Live injectable SC; 0.5 Primary series: 1 dose ≥9 mo old Contraindicated in
mL Dose must be given at immunocompromised
least 10 days before hosts. Avoid in
arrival to risk area. pregnancy and in
Booster: no longer breastfeeding
required by WHO. mothers, unless high-
U.S. travelers: risk travel cannot be
recommended every avoided.
10 yr for high-risk Contraindicated in
travelers. infants <4 mo old.
Avoid in persons with
thymus disorders.
Infants 6-8 mo old:
consider vaccination
with caution if risk or
travel cannot be
avoided; consult
travel medicine
specialist.
Caution in persons
≥60 yr old (high risk
for vaccine-related
infection).
Requires official
certificate of
vaccination.
IM, Intramuscular; SC, subcutaneous; WHO, World Health Organization.
Cholera
Cholera is present in many low- and middle-income countries, but the risk for
infection among travelers to these countries is extremely low. At present, no
cholera vaccine is available for travelers in the United States, although an
effective vaccine is available in other countries. Travelers entering countries
reporting cholera outbreaks are at minimal risk of acquiring cholera if they take
adequate safe food and water precautions and practice frequent handwashing. No
country or territory currently requires cholera vaccination as a condition for
chloroquine , is widespread, and in most areas of the world other agents must be
used (Table 200.2 ). Factors that must be considered in choosing appropriate
chemoprophylaxis medications and dosing schedules include age of the child,
travel itinerary (including whether the child will be traveling to areas of risk
within a particular country and whether chloroquine-resistant P. falciparum is
present in the country), vaccinations being given, allergies or other known
adverse reactions to antimalarial agents, and the availability of medical care
during travel.
Table 200.2
Antimalarial Chemoprophylaxis for Children
ADULT PEDIATRIC
AREA DRUG ADVANTAGES DISADVANTAGES COMMENTS
DOSE DOSE
Chloroquine- Mefloquine* 250 mg Weight <10 Once-weekly Bitter taste Children
resistant area † salt kg: 5 mg salt dosing No pediatric going to
(228 (4.6 mg formulation malaria-
mg base)/kg/wk Side effects of endemic
base) sleep area for ≥4
tablets disturbance, wk
One vivid dreams Children
tablet unlikely to
weekly take daily
Weight 10-19 medication
kg:
tablet/wk
Weight
20-30 kg:
tablet/wk
Weight
31-45 kg:
tablet/wk
Weight >45
kg: 1
tablet/wk
Doxycycline 100 mg 2 mg/kg daily Known Cannot give to Children ≥8 yr
‡ tablet (max: 100 safety children <8 yr old going to
One mg) profile old area for <4 wk
tablet Readily Daily dosing who cannot
daily available in Must take with take or cannot
most food or causes obtain
pharmacies stomach upset atovaquone-
Photosensitivity proguanil
Yeast
superinfections
Atovaquone- 250/100 Pediatric Pediatric Daily dosing Children going
proguanil § adult tablet: 62.5 tablet Expensive to malaria-
(Malarone) tablet mg formulation Can cause endemic area
One atovaquone/25 available stomach upset for <4 wk
tablet mg proguanil Generally
daily well
Weight 5-8 tolerated
kg:
pediatric
tablet once
daily
Weight >8-10
kg:
pediatric
tablet once
daily
Weight >10-
20 kg: 1
pediatric
tablet once
daily
Weight >20-
30 kg: 2
pediatric
tablets once
daily
Weight >30-
40 kg: 3
pediatric
tablets once
daily
Weight >40
kg: 1 adult
tablet once
daily
Chloroquine- Chloroquine 500 mg 8.3 mg/kg salt Once- Bitter taste Best
susceptible phosphate salt (5 mg/kg weekly No pediatric medication for
area (300 base) weekly dosing formulation children
mg Generally traveling to
base) well areas with
One tolerated Plasmodium
tablet Safe in falciparum or
weekly pregnancy P. vivax that is
chloroquine
susceptible
* Chloroquine and mefloquine should be started 1-2 wk before departure and continued for 4 wk
after last exposure.
† Mefloquine resistance exists in western Cambodia and along the Thailand–Cambodia and
Thailand–Myanmar borders. Travelers to these areas should take doxycycline or atovaquone-
proguanil. See text for precautions about mefloquine use.
‡ Doxycycline should be started 1-2 days before departure and continued for 4 wk after last
should never be substituted for seeking appropriate medical care, but it can be
considered in special circumstances such as travel to remote areas, intolerance of
prophylaxis, or refusal of chemoprophylaxis by the traveler. Self-treatment
medication should be different than the prescribed chemoprophylaxis. The CDC
or a travel medicine specialist should be consulted if self-treatment medication is
being considered for a traveler.
Table 200.3
USUAL INCUBATION
DISEASE DISTRIBUTION
PERIOD (RANGE)
INCUBATION <14 DAYS
Chikungunya 2-4 days (1-14 days) Tropics, subtropics
Dengue 4-8 days (3-14 days) Topics, subtropics
Encephalitis, arboviral (Japanese 3-14 days (1-20 days) Specific agents vary by region
encephalitis, tick-borne encephalitis,
West Nile virus, other)
Enteric fever 7-18 days (3-60 days) Especially in Indian subcontinent
Acute HIV 10-28 days (10 days to 6 wk) Worldwide
Influenza 1-3 days Worldwide, can also be acquired while
traveling
Legionellosis 5-6 days (2-10 days) Widespread
Leptospirosis 7-12 days (2-26 days) Widespread, most common in tropical
areas
Malaria, Plasmodium falciparum 6-30 days (98% onset within 3 Tropics, subtropics
mo of travel)
Malaria, Plasmodium vivax 8 days to 12 mo (almost half Widespread in tropics and subtropics
have onset >30 days after
completion of travel)
Spotted-fever rickettsiae Few days to 2-3 wk Causative species vary by region
Zika virus infection 3-14 days Widespread in Latin America, endemic
through much of Africa, Southeast Asia,
and Pacific Islands
INCUBATION 14 DAYS TO 6 WK
Encephalitis, arboviral; enteric fever; See above incubation periods See above distribution for relevant
acute HIV; leptospirosis; malaria for relevant diseases. diseases.
Amebic liver abscess Weeks to months Most common in resource-poor countries
Hepatitis A 28-30 days (15-50 days) Most common in resource-poor countries
Hepatitis E 26-42 days (2-9 wk) Widespread
Acute schistosomiasis (Katayama 4-8 wk Most common in sub-Saharan Africa
syndrome)
INCUBATION >6 WK
Amebic liver abscess, hepatitis E, See above incubation periods See above distribution for relevant
malaria, acute schistosomiasis for relevant diseases. diseases.
Hepatitis B 90 days (60-150 days) Widespread
Leishmaniasis, visceral 2-10 mo (10 days to years) Asia, Africa, Latin America, southern
Europe, and the Middle East
Tuberculosis Primary, weeks; reactivation, Global distribution, rates, and levels of
years resistance vary widely.
From Wilson ME: Post-travel evaluation. In CDC Yellow Book , Chapter 5 (Table 5.3 ).
https://wwwnc.cdc.gov/travel/yellowbook/2018/post-travel-evaluation/fever-in-returned-travelers .
Among all persons returning from travel (children and adults), 3 major
patterns of illness have been noted (Table 200.5 ). The etiology of each of these
disease presentations in part depends on the country or geographic region visited
(see Table 200.3 ). Table 200.6 provides suggestive clues to a diagnosis.
Table 200.5
Patterns of Illness in Returning International Travelers
SYSTEMIC FEBRILE ILLNESS
Malaria
Dengue
Zika
Enteric fever (typhoid/paratyphoid)
Chikungunya virus
Spotted-fever rickettsiae
Hepatitis A
Acute HIV
Leptospirosis
Measles
Infectious mononucleosis
Respiratory causes (pneumonia, influenza)
Undetermined fever source
ACUTE DIARRHEA
Campylobacter
Shigella spp.
Salmonella spp.
Diarrheagenic Escherichia coli (enterotoxigenic E. coli , enteroadherent E. coli —not tested for by routine stool
culture methods)
Giardiasis (acute, persistent, or recurrent)
Entamoeba histolytica
Cryptosporidium spp.
Cyclospora cayetanensis
Presumed viral enteritis
DERMATOLOGIC MANIFESTATIONS
Rash with fever (dengue)
Arthropod-related dermatitis (insect bites)
Cutaneous larva migrans (Ancylostoma braziliense )
Bacterial skin infections—pyoderma, impetigo, ecthyma, erysipelas
Myiasis (tumbu and botfly)
Scabies
Tungiasis
Superficial mycosis
Animal bites
Leishmaniasis
Rickettsial diseases
Marine envenomation/dermatitis
Photoallergic dermatitis and phytophotodermatitis
Table 200.6
Table 201.1
Fevers Prone to Relapse
INFECTIOUS CAUSES
Relapsing fever (Borrelia recurrentis )
Q fever (Coxiella burnetii )
Typhoid fever (Salmonella typhi )
Syphilis (Treponema pallidum )
Tuberculosis
Histoplasmosis
Coccidioidomycosis
Blastomycosis
Melioidosis (Pseudomonas pseudomallei )
Lymphocytic choriomeningitis (LCM) infection
Dengue fever
Yellow fever
Chronic meningococcemia
Colorado tick fever
Leptospirosis
Brucellosis
Oroya fever (Bartonella bacilliformis )
Acute rheumatic fever
Rat-bite fever (Spirillum minus )
Visceral leishmaniasis
Lyme disease (Borrelia burgdorferi )
Malaria
Babesiosis
Noninfluenza respiratory viral infection
Epstein-Barr virus infection
NONINFECTIOUS CAUSES
Behçet disease
Crohn disease
Weber-Christian disease (panniculitis)
Leukoclastic angiitis syndromes
Sweet syndrome
Systemic lupus erythematosus and other autoimmune disorders
PERIODIC FEVER SYNDROMES (see Chapter 188 )
Familial Mediterranean fever
Cyclic neutropenia
Periodic fever, aphthous stomatitis, pharyngitis, and adenopathy (PFAPA)
Hyper–immunoglobulin D syndrome
Hibernian fever (tumor necrosis factor superfamily immunoglobulin A–associated syndrome [TRAPS])
Muckle-Wells syndrome
Others
The double quotidian fever (or fever that peaks twice in 24 hr) is classically
associated with inflammatory arthritis. In general, a single isolated fever spike is
not associated with an infectious disease. Such a spike can be attributed to the
infusion of blood products and some drugs, as well as some procedures, or to
manipulation of a catheter on a colonized or infected body surface. Similarly,
temperatures in excess of 41°C (105.8°F) are most often associated with a
noninfectious cause. Causes for very high temperatures (>41°C [105.8°F])
include central fever (resulting from central nervous system dysfunction
involving the hypothalamus or spinal cord injury), malignant hyperthermia,
malignant neuroleptic syndrome, drug fever, or heat stroke. Temperatures that
are lower than normal (<36°C [96.8°F]) can be associated with overwhelming
sepsis but are more often related to cold exposure, hypothyroidism, or overuse of
antipyretics.
Clinical Features
The clinical features of fever can range from no symptoms to extreme malaise.
Children might complain of feeling hot or cold, display facial flushing, and
experience shivering. Fatigue and irritability may be evident. Parents often
report that the child looks ill or pale and has a decreased appetite. The
underlying etiology also produces accompanying symptoms. Although the
underlying etiologies can manifest in varied ways clinically, there are some
predictable features. For example, fever with petechiae in an ill-appearing
patient indicates the high possibility of life-threatening conditions such as
meningococcemia, Rocky Mountain spotted fever, or acute bacterial
endocarditis.
Changes in heart rate, most frequently tachycardia, accompany fever.
Normally heart rate rises by 10 beats/min per 1°C (1.8°F) rise in temperature for
children >2 mo old. Relative tachycardia, when the pulse rate is elevated
disproportionately to the temperature, is usually caused by noninfectious
diseases or infectious diseases in which a toxin is responsible for the clinical
manifestations. Relative bradycardia (temperature-pulse dissociation), when
the pulse rate remains low in the presence of fever, can accompany typhoid
fever, brucellosis, leptospirosis, or drug fever. Bradycardia in the presence of
fever also may be a result of a conduction defect resulting from cardiac
involvement with acute rheumatic fever, Lyme disease, viral myocarditis, or
infective endocarditis.
Evaluation
Most acute febrile episodes in a normal host can be diagnosed by a careful
history and physical examination and require few, if any, laboratory tests.
Because infection is the most likely etiology of the acute fever, the evaluation
should initially be geared to discovering an underlying infectious cause (Table
201.2 ). The details of the history should include the onset and pattern of fever
and any accompanying signs and symptoms. The patient often displays signs or
symptoms that provide clues to the cause of the fever. Exposures to other ill
persons at home, daycare, and school should be noted, along with any recent
travel or medications. The past medical history should include information about
underlying immune deficiencies or other major illnesses and receipt of childhood
vaccines.
Table 201.2
Evaluation of Acute Fever
Thorough history: onset, other symptoms, exposures (daycare, school, family, pets, playmates), travel,
medications, other underlying disorders, immunizations
Physical examination: complete, with focus on localizing symptoms
Laboratory studies on a case-by-case basis:
• Rapid antigen testing
• Nasopharyngeal: respiratory viruses by polymerase chain reaction
• Throat: group A streptococcus
• Stool: NAAT for enteric pathogens, calprotectin
• Blood: complete blood count, blood culture, C-reactive protein, sedimentation rate, procalcitonin
• Urine: urinalysis, culture
• Cerebrospinal fluid: cell count, glucose, protein, Gram stain, culture
• Chest radiograph or other imaging studies on a case-by-case basis
NAAT, Nucleic acid amplification test.
Table 202.1
Bacterial Pathogens in Neonates and Young Infants With
Urinary Tract Infection, Bacteremia, or Meningitis
Many experts advocate that all neonates ≤28 days old undergo a complete
evaluation for serious infection, receive empirical antimicrobials, and be
hospitalized. Of the 3 widely used criteria, only the Rochester criteria allow
neonates ≤28 days to be designated as “low risk” and managed outside the
hospital without antimicrobials. In one study, <1% of low-risk infants ≤28 days
old had SBI; however, in another study applying the Boston and Philadelphia
criteria to neonates, 3–4% of those classified as low risk had SBI.
Young febrile infants ≥29 days old who appear ill (with signs of systemic
illness) require complete evaluation for SBI, including antimicrobials and
hospitalization; however, well-appearing infants can be managed safely as
outpatients using low-risk criteria as indicated in Table 202.2 . In each of these
approaches, infants must have a normal physical examination, must be able to
reliably obtain close follow-up, and must meet certain laboratory and/or
radiographic criteria. Based on these protocols, all infants following the Boston
or Philadelphia criteria would undergo lumbar puncture (LP), whereas low-risk
infants following the Rochester criteria would not. There is substantial variation
dipstick alone (99.2% vs 98.7%), but that dipstick alone had a higher positive
predictive value (PPV, 66.8% for dipstick alone vs 51.2% for traditional UA).
Enhanced UA includes hemocytometer cell count (to decrease variability of
urine cell counts) and Gram stain on uncentrifuged urine. The enhanced UA has
a higher sensitivity but comparable specificity to traditional UA. However, the
enhanced UA has not been studied in the most common protocols for evaluation
of the febrile infant, and many institutions/office practices do not perform this
test.
Cerebrospinal Fluid
CSF evaluation consists of culture and Gram stain, cell count, glucose and
protein. Polymerase chain reaction (PCR) testing may also be sent based on the
clinical scenario, usually for enterovirus or HSV. Normal CSF parameters vary
by age of the infant and should be interpreted in combination with other clinical
and historical risk factors, given that some infants with normal CSF parameters
may have CNS infections (Table 202.3 ). The CSF Gram stain can be a useful
adjunct to other CSF parameters given the high specificity of the test (99.3–
99.9%; i.e., relatively few false-positive results), although the range of reported
sensitivity is much broader (67–94.1%).
Table 202.3
infants who present to the emergency department with fever: establishing normal values by week
of age, Pediatr Infect Dis J 30(4):e63–e67, 2011. All infants were excluded if they had identified
viral or bacterial meningitis, positive blood or urine cultures, a ventriculoperitoneal shunt, recent
neurosurgery/antibiotics/seizure, or a traumatic LP.
‡ Data from Kestenbaum LA, Ebberson J, Zorc JJ, et al: Defining cerebrospinal fluid white blood
cell count reference values in neonates and young infants, Pediatrics 125(2):257–264, 2010.
Infants were excluded for traumatic LP, serious bacterial infection, congenital infection, seizure,
presence of ventricular shunt, or positive CSF testing for enterovirus.
§ Data from Shah SS, Ebberson J, Kestenbaum LA, et al: Age-specific reference values for
cerebrospinal fluid protein concentration in neonates and young infants, J Hosp Med 6(1):22–27,
2011. Infants were excluded for traumatic LP, serious bacterial infection, congenital infection,
seizure, presence of a ventricular shunt, positive CSF testing for enterovirus, or elevated serum
bilirubin.
Fever is the most common reason for a child to seek medical care. While most
infants and children have benign viral causes of fever, a small percentage will
have more serious infections. Unlike the situation in infants <2 mo of age, in
older children with fever, pediatricians can rely more readily on symptoms and
physical examination findings to establish a diagnosis. Diagnostic testing,
including laboratory testing and radiographic studies, is not routinely indicated
unless diagnostic uncertainty exists after examination or the patient appears
critically ill. Occult infections, such as urinary tract infection, may be present,
and screening for such infections should be guided by patient age, patient
gender, and degree of fever.
Diagnosis
The many potential causes of fever in older infants and children can be broadly
categorized into viral and bacterial infections, further organized by body region,
as well as the less common inflammatory, oncologic, endocrine, and medication-
induced causes (Table 203.1 ).
Table 203.1
Etiologies of Fever in Children >2 Mo of Age
INFECTIOUS
Central Nervous System
Bacterial meningitis
Viral meningitis
Viral encephalitis
Epidural abscess
Brain abscess
Ear, Nose, and Throat
Acute otitis media
Mastoiditis
Viral upper respiratory infection (i.e., common cold)
Acute bacterial sinusitis
Acute streptococcal pharyngitis
Acute viral pharyngitis
Retropharyngeal abscess
Ludwig angina
Peritonsillar abscess
Herpangina
Herpes simplex virus gingivostomatitis
Acute bacterial lymphadenitis
Viral laryngotracheobronchitis (i.e., croup)
Bacterial tracheitis
Epiglottitis
Lemierre syndrome
Face and Ocular
Parotitis (viral and bacterial)
Erysipelas
Preseptal cellulitis
Orbital cellulitis
Lower Respiratory Tract
Acute viral bronchiolitis
Pneumonia (viral and bacterial)
Complicated pneumonia (e.g., empyema, pleural effusion)
Tuberculosis
Cardiac
Pericarditis
Myocarditis
Endocarditis
Gastrointestinal
Gastroenteritis (viral and bacterial)
Mesenteric adenitis
Acute appendicitis
Hepatitis
Pancreatitis
Gallbladder disease (e.g., cholecystitis, cholangitis)
Intraabdominal abscess
Genitourinary
Urinary tract infection/pyelonephritis
Renal abscess
Epididymitis
Pelvic inflammatory disease
Tuboovarian abscess
Skin, Soft Tissue, and Muscle
Viral exanthemas (e.g., varicella, coxsackievirus, roseola, measles)
Scarlet fever
Syphilis
Cellulitis
Abscess
Necrotizing fasciitis
Myositis (viral and bacterial)
Bone and Joint
Osteomyelitis
Septic arthritis
Transient synovitis
Discitis
Toxin Mediated
Toxic shock syndrome
Staphylococcal scalded skin syndrome
Invasive Bacterial Infections
Occult bacteremia
Bacterial sepsis
Bacterial meningitis
Disseminated gonococcal infection
Vector-Borne (Tick, Mosquito)
Lyme disease
Rickettsiae (e.g., Rocky Mountain spotted fever, ehrlichiosis)
Arboviruses (e.g., West Nile virus)
Dengue fever
Inflammatory
Kawasaki disease
Acute rheumatic fever
Systemic lupus erythematosus
Inflammatory bowel disease
Juvenile idiopathic arthritis
Henoch-Schönlein purpura
Other rheumatologic diseases (e.g., dermatomyositis)
Periodic fever syndromes
Serum-like sickness syndrome
Oncologic
Leukemia
Lymphoma
Solid tumors (e.g., neuroblastoma)
Endocrine
Thyrotoxicosis/thyroid storm
Medication Induced
Serotonin syndrome
Anticholinergic toxidrome (e.g., antihistamines)
Sympathomimetic toxidrome (e.g., cocaine)
Salicylate toxicity
Other
Hemophagocytic lymphohistiocytosis
Macrophage activation syndrome
Ectodermal dysplasia
Dysautonomia
Viral Infections
Viral infections are the most common cause of fever, and the prevalence of
specific viral infections varies by season. In the summer and early fall,
enteroviruses (e.g., coxsackieviruses) predominate, usually presenting as hand-
foot-and-mouth disease, herpangina, aseptic meningitis, or a variety of other
manifestations. In the late fall and winter, viral upper and lower respiratory tract
groups together, as well as separately for B, previously healthy children ≥28 days old,
and C, neonates and children with comorbidities ≥28 days old. *Pseudomonas
aeruginosa, Klebsiella spp., Neisseria meningitidis, Haemophilus influenzae, other
gram-negative pathogens. † Enterococcus spp., viridans group streptococci, other
gram-positive pathogens. (From Agyeman PKA, Schlapbach LJ, Giannoni E, et al:
Epidemiology of blood culture-proven bacterial sepsis in children in Switzerland: a
population-based cohort study, Lancet Child Adolesc 1:124–133, 2017, Fig 4.)
Infants and children age 2-24 mo merit special consideration because they
have limited verbal skills, are at risk for occult bacterial infections, and may be
otherwise asymptomatic except for fever (see Chapter 202 ).
Table 203.2
Risk Factors for Urinary Tract Infection in Children 2-24 Mo
of Age
FEMALE
White race
Age <1 yr
Temperature ≥39°C (102.2°F)
Fever duration ≥2 days
No obvious source of infection
MALE
Uncircumcised boys at higher risk
Nonblack race
Temperature ≥39°C (102.2°F)
Fever duration >1 day
No obvious source of infection
Adapted from Subcommittee on Urinary Tract Infection et al. Urinary tract infection: clinical
practice guideline for the diagnosis and management of the initial UTI in febrile infants and
children 2 to 24 months, Pediatrics 128(3):595–610, 2011.
Occult Bacteremia
Occult bacteremia is defined as a positive blood culture for a pathogen in a well-
CHAPTER 204
Table 204.1
Summary of Definitions and Major Features of 4 Subtypes
of Fever of Unknown Origin (FUO)
HEALTHCARE-
IMMUNE-
FEATURE CLASSIC FUO ASSOCIATED HIV-RELATED FUO
DEFICIENT FUO
FUO
Definition >38°C (100.4°F), >3 wk, >2 ≥38°C (100.4°F), ≥38°C (100.4°F), ≥38°C (100.4°F), >3 wk for
visits or 1 wk in hospital >1 wk, not >1 wk, negative outpatients, >1 wk for
present or cultures after 48 hr inpatients, HIV infection
incubating on confirmed
admission
Patient Community, clinic, or Acute care Hospital or clinic Community, clinic, or
location hospital hospital hospital
Leading Cancer, infections, Healthcare- Majority caused by HIV itself, typical and
causes inflammatory conditions, associated infections, but atypical mycobacteria,
undiagnosed, habitual infections, cause documented CMV, lymphomas,
hyperthermia postoperative in only 40–60% toxoplasmosis,
complications, cryptococcosis, immune
drug fever reconstitution inflammatory
syndrome (IRIS)
History Travel, contacts, animal and Operations and Stage of Drugs, exposures, risk
emphasis insect exposure, procedures, chemotherapy, factors, travel, contacts,
medications, immunizations, devices, anatomic drugs administered, stage of HIV infection
family history, cardiac valve considerations, underlying
disorder drug treatment immunosuppressive
disorder
Examination Fundi, oropharynx, temporal Wounds, drains, Skin folds, IV sites, Mouth, sinuses, skin, lymph
emphasis artery, abdomen, lymph devices, sinuses, lungs, perianal area nodes, eyes, lungs, perianal
nodes, spleen, joints, skin, urine area
nails, genitalia, rectum or
prostate, lower-limb deep
veins
Investigation Imaging, biopsies, Imaging, bacterial CXR, bacterial Blood and lymphocyte
emphasis sedimentation rate, skin tests cultures cultures count; serologic tests; CXR;
stool examination; biopsies
of lung, bone marrow, and
liver for cultures and
cytologic tests; brain
imaging
Management Observation, outpatient Depends on Antimicrobial Antiviral and antimicrobial
temperature chart, situation treatment protocols protocols, vaccines,
investigations, avoidance of revision of treatment
empirical drug treatments regimens, good nutrition
Time course Months Weeks Days Weeks to months
of disease
Tempo of Weeks Days Hours Days to weeks
investigation
CMV, Cytomegalovirus; CXR, chest radiograph; HIV, human immunodeficiency virus; IV,
intravenous line.
Adapted from Mackowak PA, Durack DT: Fever of unknown origin. In Mandell GL, Bennett, JE,
Dolin R, editors: Mandell, Douglas, and Bennett's principles and practice of infectious diseases ,
ed 7, Philadelphia, 2010, Elsevier (Table 51-1).
Etiology
The many causes of FUO in children are infectious, rheumatologic (connective
tissue or autoimmune), autoinflammatory, oncologic, neurologic, genetic,
factitious, and iatrogenic processes (Table 204.2 ). Although oncologic disorders
should be seriously considered, most children with malignancies do not have
fever alone. The possibility of drug fever should be considered if the patient is
receiving any drug. Drug fever is usually sustained and not associated with other
symptoms. Discontinuation of the drug is associated with resolution of the fever,
generally within 72 hr, although certain drugs, such as iodides, are excreted for a
prolonged period, with fever that can persist for as long as 1 mo after drug
withdrawal.
Table 204.2
Diagnostic Considerations for Fever of Unknown Origin in
Children
ABSCESSES
Abdominal
Brain
Dental
Hepatic
Paraspinal
Pelvic
Perinephric
Rectal
Subphrenic
Psoas
BACTERIAL DISEASES
Actinomycosis
Bartonella henselae (cat-scratch disease)
Brucellosis
Campylobacter
Chlamydia
Francisella tularensis (tularemia)
Listeria monocytogenes (listeriosis)
Meningococcemia (chronic)
Mycoplasma pneumoniae
Rat-bite fever (Streptobacillus moniliformis ; streptobacillary form of rat-bite fever)
Salmonella
Tuberculosis
Whipple disease
Yersiniosis
LOCALIZED INFECTIONS
Cholangitis
Infective endocarditis
Lymphogranuloma venereum
Mastoiditis
Osteomyelitis
Pneumonia
Pyelonephritis
Psittacosis
Sinusitis
SPIROCHETES
Borrelia burgdorferi (Lyme disease)
Relapsing fever (Borrelia recurrentis)
Leptospirosis
Rat-bite fever (Spirillum minus ; spirillary form of rat-bite fever)
Syphilis
FUNGAL DISEASES
Blastomycosis (extrapulmonary)
Coccidioidomycosis (disseminated)
Histoplasmosis (disseminated)
RICKETTSIAE
African tick-bite fever
Ehrlichia canis
Q fever
Rocky Mountain spotted fever
Tick-borne typhus
VIRUSES
Cytomegalovirus
Hepatitis viruses
HIV
Epstein-Barr virus
PARASITIC DISEASES
Amebiasis
Babesiosis
Giardiasis
Malaria
Toxoplasmosis
Trichinosis
Trypanosomiasis
Visceral larva migrans (Toxocara )
RHEUMATOLOGIC DISEASES
Behçet disease
Juvenile dermatomyositis
Juvenile idiopathic arthritis
Rheumatic fever
Systemic lupus erythematosus
HYPERSENSITIVITY DISEASES
Drug fever
Hypersensitivity pneumonitis
Serum sickness
Weber-Christian disease
NEOPLASMS
Atrial myxoma
Cholesterol granuloma
Hodgkin disease
Inflammatory pseudotumor
Leukemia
Lymphoma
Pheochromocytoma
Neuroblastoma
Wilms tumor
GRANULOMATOUS DISEASES
Granulomatosis with polyangiitis
Crohn disease
Granulomatous hepatitis
Sarcoidosis
FAMILIAL AND HEREDITARY DISEASES
Anhidrotic ectodermal dysplasia
Autonomic neuropathies
Fabry disease
Familial dysautonomia
Familial Hibernian fever
Familial Mediterranean fever, many other autoinflammatory diseases (Chapter 188 )
Hypertriglyceridemia
Ichthyosis
Sickle cell crisis
Spinal cord/brain injury
MISCELLANEOUS
Addison disease
Castleman disease
Chronic active hepatitis
Cyclic neutropenia
Diabetes insipidus (central and nephrogenic)
Drug fever
Factitious fever
Hemophagocytic syndromes
Hypothalamic-central fever
Infantile cortical hyperostosis
Inflammatory bowel disease
Kawasaki disease
Kikuchi-Fujimoto disease
Metal fume fever
Pancreatitis
Periodic fever syndromes
Poisoning
Pulmonary embolism
Thrombophlebitis
Thyrotoxicosis, thyroiditis
Table 204.3
Subtle Physical Findings with Special Significance in
Patients with Fever of Unknown Origin
Malaria Recent travel to endemic areas in Asia, Africa, and Fever, headaches, nausea, emesis,
Central/South America diarrhea, hepatomegaly, splenomegaly,
anemia
Psittacosis Associated with contact with birds, especially Fever, pharyngitis,
(Chlamydia psittaci psittacine birds hepatosplenomegaly, pneumonia,
) blanching maculopapular eruptions;
erythema multiforme, marginatum, and
nodosum
Q fever (Coxiella Associated with farm, veterinary, or abattoir work; Atypical pneumonia, hepatitis,
burnetii ) consumption of unpasteurized milk; contact with hepatomegaly, relative bradycardia,
infected sheep, goats, or cattle splenomegaly
Rat-bite fever Recent bite or scratch by rat, mouse, or squirrel; Headaches, myalgias, polyarthritis, and
(Streptobacillus ingestion of food or water contaminated by rat maculopapular, morbilliform, petechial,
moniliformis ) excrement vesicular, or pustular rash over the
palms, soles, and extremities
Relapsing fever Associated with poverty, crowding, and poor High fever with rigors, headache,
(Borrelia sanitation (louse-borne), or with camping (tick- delirium, arthralgias, myalgias, and
recurrentis ) borne), particularly in the Grand Canyon hepatosplenomegaly
Rocky Mountain Associated with outdoor activity in the South Headache, petechial rash involving the
spotted fever Atlantic or southeastern United States and extremities, palms, and soles
exposure to Dermacentor tick bites
Tuberculosis Recent contact with tuberculosis; recent Night sweats, weight loss, atypical
immigration from endemic country; work or pneumonia, cavitary pulmonary lesions
residence in homeless shelters, correctional
facilities, or healthcare facilities
Tularemia Associated with bites by Amblyomma or Ulcerated skin lesions at a bite site,
Dermacentor ticks, deer flies, and mosquitoes or pneumonia, relative bradycardia,
direct contact with tissues of infected animals such lymphadenopathy, conjunctivitis
as rabbits, squirrels, deer, raccoons, cattle, sheep,
and swine
Whipple disease Potential association with exposure to sewage Chronic diarrhea, arthralgia, weight
(Tropheryma loss, malabsorption, malnutrition
whipplei )
Adapted from Wright WF, Mackowiak PA: Fever of unknown origin. In Bennett JF, Dolin R, Blaser
MJ, editors: Mandell, Douglas, and Bennett's principles and practice of infectious diseases, ed 8,
Philadelphia, 2015, Elsevier (Table 56-9).
Table 205.1
Most Common Causes of Infections in
Immunocompromised Children
BACTERIA, AEROBIC
Acinetobacter
Bacillus
Burkholderia cepacia
Citrobacter
Corynebacterium
Enterobacter spp.
Enterococcus faecalis
Enterococcus faecium
Escherichia coli
Klebsiella spp.
Listeria monocytogenes
Mycobacterium spp.
Neisseria meningitidis
Nocardia spp.
Pseudomonas aeruginosa
Staphylococcus aureus
Staphylococcus, coagulase-negative
Streptococcus pneumoniae
Streptococcus, viridans group
BACTERIA, ANAEROBIC
Bacillus
Clostridium
Fusobacterium
Peptococcus
Peptostreptococcus
Propionibacterium
Veillonella
FUNGI
Aspergillus
Candida albicans
Other Candida spp.
Cryptococcus neoformans
Fusarium spp.
Pneumocystis jiroveci
Zygomycoses (Mucor, Rhizopus, Rhizomucor)
VIRUSES
Adenoviruses
Cytomegalovirus
Epstein-Barr virus
Herpes simplex virus
Human herpesvirus 6
Polyomavirus (BK)
Respiratory and enteric community-acquired viruses
Varicella-zoster virus
PROTOZOA
Cryptosporidium parvum
Giardia lamblia
Toxoplasma gondii
205.1
Infections Occurring With Primary
Immunodeficiencies
Marian G. Michaels, Hey Jin Chong, Michael Green
Currently, more than 300 genes involving inborn errors of immunity have been
identified, accounting for a wide array of diseases presenting with susceptibility
to infection, allergy, autoimmunity, and autoinflammation, as well as
malignancy.
Table 205.2
Host Defense Defects and Common Pathogens by Time
After Bone Marrow or Hematopoietic Stem Cell
Transplantation
TIME COMMON
HOST DEFENSE DEFECTS CAUSES
PERIOD PATHOGENS
Pretransplant Neutropenia Underlying disease Aerobic gram-negative
Abnormal anatomic barriers Prior chemotherapy bacilli
Preengraftment Neutropenia Chemotherapy Aerobic gram-
Abnormal anatomic barriers Radiation positive cocci
Indwelling catheters Aerobic gram-
negative bacilli
Candida
Aspergillus
Herpes simplex
virus (in previously
infected patients)
Community-
acquired viral
pathogens
Postengraftment Abnormal cell-mediated immunity Chemotherapy Gram-positive cocci
Abnormal anatomic barriers Immunosuppressive Aerobic gram-
medications negative bacilli
Radiation Cytomegalovirus
Indwelling catheters Adenoviruses
Unrelated cord blood Community-
donor acquired viral
pathogens
Pneumocystis
jiroveci
Late Delayed recovery of immune function Time required to Varicella-zoster
posttransplant (cell-mediated, humoral, and abnormal develop donor-related virus
anatomic barriers) immune function Streptococcus
Graft-versus-host pneumoniae
disease
Solid-Organ Transplantation
Factors predisposing to infection after organ transplantation include those that
either existed before transplantation or are secondary to intraoperative events or
posttransplantation therapies (Table 205.3 ). Some of these additional risks
cannot be prevented, and some risks acquired during or after the operation
depend on decisions or actions of members of the transplant team. Organ
recipients are at risk for infection from potential exposure to pathogens in the
donor organ. Although some donor-derived infections can be anticipated through
donor screening, many pathogens are not routinely screened for, and strategies
defining when and how to screen for all but a small subset of potential pathogens
have not been identified or implemented. Similar to other children who have
undergone surgical procedures; surgical site infections are a frequent cause of
infection early after transplantation. Beyond this, the need for
immunosuppressive agents to prevent rejection is the major factor predisposing
to infection following transplantation. Despite efforts to optimize
immunosuppressive regimens to prevent or treat rejection with minimal
impairment of immunity, all current regimens interfere with the ability of the
immune system to prevent infection. The primary target of the majority of these
immunosuppressive agents in organ recipients is the cell-mediated immune
system, but regimens can and do impair many other aspects of the transplant
recipient's immune system as well.
Table 205.3
Risk Factors for Infections After Solid-Organ
Transplantation in Children
PRETRANSPLANTATION FACTORS
Age of patient
Underlying disease, malnutrition
Specific organ transplanted
Previous exposures to infectious agents
Previous immunizations
Presence of infection in the donor
INTRAOPERATIVE FACTORS
Duration of transplant surgery
Exposure to blood products
Technical problems
Organisms transmitted with donor organ
POSTTRANSPLANTATION FACTORS
Immunosuppression
Induction immunosuppression type
Maintenance immunosuppression
Augmented treatment for rejection
Indwelling catheters
Nosocomial exposures
Community exposures
Timing
The timing of specific types of infections is generally predictable, regardless of
which organ is transplanted. Infectious complications typically develop in 1 of 3
intervals: early (0-30 days after transplantation), intermediate (30-180 days), or
late (>180 days); most infections present in the 1st 180 days after
transplantation. Table 205.4 should be used as a general guideline to the types of
infections encountered but may be modified with the introduction of newer
immunosuppressive therapies and by the use of prophylaxis.
Table 205.4
Timing of Infectious Complications After Solid-Organ
Transplantation
EARLY PERIOD (0-30 DAYS)
Bacterial Infections
Gram-negative enteric bacilli
• Small bowel, liver, neonatal heart
Pseudomonas, Burkholderia, Stenotrophomonas, Alcaligenes
• Cystic fibrosis lung
Gram-positive organisms
• All transplant types
Fungal Infections
• All transplant types
Viral Infections
Herpes simplex virus
• All transplant types
Nosocomial respiratory viruses
• All transplant types
MIDDLE PERIOD (1-6 MO)
Viral Infections
Cytomegalovirus
• All transplant types
• Seronegative recipient of seropositive donor
Epstein-Barr virus
• All transplant types (small bowel the highest-risk group)
• Seronegative recipient
Varicella-zoster virus
• All transplant types
• Opportunistic infections
Pneumocystis jiroveci
• All transplant types
Toxoplasma gondii
• Seronegative recipient of cardiac transplant from a seropositive donor
Bacterial Infections
Pseudomonas, Burkholderia, Stenotrophomonas, Alcaligenes
• Cystic fibrosis lung
Gram-negative enteric bacilli
• Small bowel
LATE PERIOD (>6 MO)
Viral Infections
Epstein-Barr virus
• All transplant types, but less risk than middle period
Varicella-zoster virus
• All transplant types
Community-acquired viral infections
• All transplant types
Bacterial Infections
Pseudomonas, Burkholderia, Stenotrophomonas, Alcaligenes
• Cystic fibrosis lung
• Lung transplants with chronic rejection
Gram-negative bacillary bacteremia
• Small bowel
Fungal Infections
Aspergillus
• Lung transplants with chronic rejection
Adapted from Green M, Michaels MG: Infections in solid organ transplant recipients. In Long SS,
Prober C, Fisher M, editors: Principles and practice of pediatric infectious disease , ed 5,
Philadelphia, 2018, Elsevier (Table 95-1).
Table 207.2
Aminoglycoside-Modifying Enzymes*
acetylation, O-nucleotidylation, and O-phosphorylation. For each of these general reactions, there
are several different enzymes that attack a specific amino or hydroxyl group.
A, Amikacin; AAC, aminoglycoside acetyltransferase; ANT, aminoglycoside
nucleotidyltransferase; APH, aminoglycoside phosphotransferase; cr, ciprofloxacin resistance; Ar,
arbekacin, E, Enterobacteriaceae; Ent, enterococci, FQ, fluoroquinolone (acetylates the
piperazine ring in some fluoroquinolones), G, gentamicin; K, kanamycin; PR, Providencia-Proteus
; PS, pseudomonads; SA, staphylococci; SR, streptococci; T, tobramycin.
Adapted from Opal SM, Pop-Vicas A: Molecular mechanisms of antibiotic resistance in bacteria.
In Bennett JF, Dolin R, Blaser MJ, editors: Mandell, Douglas, and Bennett's principles and practice
of infectious diseases, ed 8, Philadelphia, 2015, Elsevier (Table 18-5).
Table 207.3
Penicillins
Although there has been ever-increasing emergence of resistance to penicillins,
these agents remain valuable and are commonly used for management of many
pediatric infectious diseases.
Penicillins remain the drugs of choice for pediatric infections caused by group
A and group B streptococcus, Treponema pallidum (syphilis), L. monocytogenes,
and N. meningitidis. The semisynthetic penicillins (nafcillin, cloxacillin,
dicloxacillin) are useful for management of susceptible (non-MRSA)
staphylococcal infections. The aminopenicillins (ampicillin, amoxicillin) were
developed to provide broad-spectrum activity against gram-negative organisms,
including E. coli and H. influenzae, but the emergence of resistance (typically
mediated by a β-lactamase) has limited their utility in many clinical settings. The
carboxypenicillins (ticarcillin) and ureidopenicillins (piperacillin, mezlocillin,
azlocillin) also have bactericidal activity against most strains of P. aeruginosa.
Resistance to penicillin is mediated by a variety of mechanisms (see Table
207.1 ). The production of β-lactamase is a common mechanism exhibited by
many organisms that may be overcome, with variable success, by including a β-
lactamase inhibitor in the therapeutic formulation with the penicillin. Such
combination products (ampicillin-sulbactam, amoxicillin-clavulanate, ticarcillin–
clavulanic acid [no longer available in the U.S.], piperacillin-tazobactam) are
potentially very useful for management of resistant isolates, but only if the
resistance is β-lactamase mediated. Notably, MRSA and S. pneumoniae mediate
resistance to penicillins through mechanisms other than β-lactamase production,
rendering these combination agents of little value for the management of these
infections.
Table 207.4 lists adverse reactions to penicillins.
Table 207.4
Adverse Reactions to Penicillins
Adapted from Doi Y, Chambers HF: Penicillins and β-lactamase inhibitors. In Bennett JF, Dolin R,
Blaser MJ, editors: Mandell, Douglas, and Bennett's principles and practice of infectious diseases,
ed 8, Philadelphia, 2015, Elsevier (Table 20-7).
pneumonia but is not indicated for MRSA pneumonia. Ceftaroline's activity is
attributed to its ability to bind to penicillin-binding protein 2a with higher
affinity than other β-lactams. Another fifth-generation cephalosporin with a
similar spectrum of activity, ceftobiprole , has been approved for use in Canada
and the European Union.
Table 207.5
Classification of Parenteral and Oral Cephalosporins
Adapted from Craig WA, Andes DR: Cephalosporins.IN Bennett JF, Dolin R, Blaser MJ, editors:
Mandell, Douglas, and Bennett's principles and practice of infectious diseases, ed 8, Philadelphia,
2015, Elsevier (Table 21-1).
Table 207.6
Potential Adverse Effects of Cephalosporins
Adapted from Craig WA, Andes DR: Cephalosporins. In Bennett JF, Dolin R, Blaser MJ, editors:
Mandell, Douglas, and Bennett's principles and practice of infectious diseases, ed 8, Philadelphia,
2015, Elsevier (Table 21-6).
Carbapenems
The carbapenems include imipenem (formulated in combination with cilastatin),
meropenem, ertapenem, and doripenem. The basic structure of these agents is
similar to that of β-lactam antibiotics, and these drugs have a similar mechanism
of action. The carbapenems provide the broadest spectrum of antibacterial
activity of any licensed class of antibiotics and are active against gram-positive,
gram-negative, and anaerobic organisms. Among the carbapenems, meropenem
is the only agent licensed for treatment of pediatric meningitis. At this time,
ertapenem and doripenem are not approved for pediatric use. Importantly,
infection; however, not all strains of MSSA or MRSA are susceptible to
clindamycin. Inducible clindamycin resistance in isolates initially reported as
susceptible must be ruled out by D-test or molecular methods. Clindamycin is
bacteriostatic and should not be used to treat endocarditis, persistent bacteremia,
or CNS infections caused by S. aureus. Given that the mechanism of action of
clindamycin involves inhibition or protein synthesis, many experts use
clindamycin to treat S. aureus toxin–mediated illnesses (e.g., TSS) to inhibit
toxin production.
Although the very-broad- spectrum carbapenems (meropenem, ertapenem,
and imipenem) have activity against MSSA, they have no activity against
MRSA. As a result, carbapenems are rarely used for empirical therapy of
possible staphylococcal infection and are too broad in most cases for use in
identified MSSA infections. Quinolone antibiotics have unpredictable activity
against MSSA and no activity against MRSA. Linezolid and daptomycin are
useful for serious S. aureus infections, particularly those caused by MRSA,
when treatment with vancomycin is ineffective or not tolerated.(Table 208.1 ). A
number of novel antistaphylococcal antibiotics have emerged for use in resistant
or refractory MSSA and MRSA infection in adults that may be required for
pediatric therapy in select patients under the guidance of a pediatric infectious
disease specialist. These include ceftaroline , a broad-spectrum
antistaphylococcal cephalosporin, and oritavancin and dalbavancin ,
lipoglycopeptides structurally related to vancomycin with very long half-lives
and broad activity against gram-positive organisms. Rifampin or gentamicin
may be added to a β-lactam or vancomycin for synergy in serious infections such
as endocarditis, particularly when prosthetic valve material is involved.
Table 208.1
Parenteral Antimicrobial Agent(s) for Treatment of Serious
Staphylococcus aureus Infections
Table 208.2
Diagnostic Criteria of Staphylococcal Toxic Shock
Syndrome
MAJOR CRITERIA (ALL REQUIRED)
Acute fever; temperature >38.8°C (101.8°F)
Hypotension (orthostatic, shock; blood pressure below age-appropriate norms)
Rash (erythroderma with convalescent desquamation)
MINOR CRITERIA (ANY 3 OR MORE)
Mucous membrane inflammation (vaginal, oropharyngeal or conjunctival hyperemia, strawberry tongue)
Vomiting, diarrhea
Liver abnormalities (bilirubin or transaminase greater than twice the upper limit of normal)
Renal abnormalities (blood urea nitrogen or creatinine greater than twice the upper limit of normal, or greater than
5 white blood cells per high-power field)
Muscle abnormalities (myalgia or creatinine phosphokinase greater than twice the upper limit of normal)
Central nervous system abnormalities (alteration in consciousness without focal neurologic signs)
Thrombocytopenia (≤100,000/mm3 )
EXCLUSIONARY CRITERIA
Absence of another explanation
Negative blood cultures (except occasionally for Staphylococcus aureus )
Data from Kimberlin DW, Brady MT, Jackson MA, Long SS, editors: Red book: 2015 report of the
Committee on Infectious Diseases , ed 30, Elk Grove Village, IL, 2015, American Academy of
Pediatrics.
Diagnosis
There is no specific laboratory test, and diagnosis depends on meeting certain
clinical and laboratory criteria in the absence of an alternate diagnosis (see Fig.
208.2 ). Appropriate tests reveal involvement of multiple organ systems,
including the hepatic, renal, muscular, gastrointestinal, cardiopulmonary, and
infections (IPIs) in children. By 2005, however, IPIs began to increase slightly
because of an increase in non-PCV7 serotypes, particularly serotype 19A.
Serotype replacement can result from expansion of existing nonvaccine
serotypes, as well as from vaccine-type pneumococci acquiring the
polysaccharide capsule of a nonvaccine serotype (serotype switching ). Since
the introduction of PCV13 in 2010 in the United States, there has been a decline
in IPIs caused by new vaccine serotypes, including 19A. Nonetheless, 19A
remains an important cause of meningitis. Indirect protection of unvaccinated
persons has occurred since PCV introduction, and this herd protection is likely a
result of decreases in nasopharyngeal carriage of virulent pneumococcal vaccine
serotypes.
S. pneumoniae is the most frequent cause of bacteremia, bacterial pneumonia,
otitis media, and bacterial meningitis in children. The decreased ability in
children <2 yr old to produce antibody against the T-cell–independent
polysaccharide antigens and the high prevalence of colonization may explain an
increased susceptibility to pneumococcal infection and the decreased
effectiveness of polysaccharide vaccines. Children at increased risk of
pneumococcal infections include those with sickle cell disease, asplenia,
deficiencies in humoral (B-cell) and complement-mediated immunity, HIV
infection, certain malignancies (e.g., leukemia, lymphoma), chronic heart, lung,
or renal disease (particularly nephrotic syndrome), cerebrospinal fluid (CSF)
leak, and cochlear implants. Table 209.1 lists other high-risk groups. Some
American Indian, Alaska Native, and African American children may also be at
increased risk. Children <5 yr old in out-of-home daycare are at increased risk
(approximately 2-fold higher) of experiencing IPIs than other children. Males
are more frequently affected than females. Because immunocompetent
vaccinated children have had fever episodes of IPI, the proportion of infected
children with immunologic risk factors has increased (estimated at 20%).
Table 209.1
Children at Increased Risk of Invasive Pneumococcal
Infection
Pathogenesis
Invasion of the host is affected by a number of factors. Nonspecific defense
mechanisms, including the presence of other bacteria in the nasopharynx, may
limit multiplication of pneumococci. Aspiration of secretions containing
pneumococci is hindered by the epiglottic reflex and by respiratory epithelial
cell cilia, which move infected mucus toward the pharynx. Similarly, normal
ciliary flow of fluid from the middle ear through the eustachian tube and sinuses
to the nasopharynx usually prevents infection with nasopharyngeal flora,
including pneumococci. Interference with these normal clearance mechanisms
by allergy, viral infection, or irritants (e.g., smoke) may allow colonization and
subsequent infection with these organisms in otherwise normally sterile sites.
20% fewer tympanostomy tube placements than unvaccinated children.
Following PCV13, a 64% reduction in IPIs caused by vaccine serotypes has
been seen, particularly in children <5 yr old. The number of pneumococcal
isolates and percentage of isolates with high-level penicillin resistance from
cultures taken from children with otitis media or mastoiditis for clinical
indications have decreased, largely related to decreases in serotype 19A. Rates of
hospitalization for pneumococcal pneumonia among U.S. children decreased
after PCV13 introduction. The number of cases of pneumococcal meningitis in
children remain unchanged, but the proportion of PCV13 serotypes have
decreased significantly. In addition, pneumococcal conjugate vaccines
significantly reduce nasopharyngeal carriage of vaccine serotypes. PCVs have
significantly decreased rates of invasive pneumococcal disease in children with
sickle cell disease, and studies suggest substantial protection for HIV-infected
children and splenectomized adults. Adverse events after the administration of
PCV have included local swelling and redness and slightly increased rates of
fever, when used in conjunction with other childhood vaccines.
Table 209.2
Table 209.3
minimum interval between doses is 4 wk. Minimum age for administration of 1st dose is 6 wk.
† Given at least 8 wk after the previous dose.
From Centers for Disease Control and Prevention. Licensure of a 13-valent pneumococcal
conjugate vaccine (PCV13) and recommendations for use among children: Advisory Committee
on Immunization Practices, MMWR 59(RR-11):1–18 (Table 8); 59:258–261, 2010 (Table 3).
Table 209.4
Medical Conditions or Other Indications for Administration
of PCV13,* and Indications for PPSV23 † Administration,
and Revaccination for Children Age 6–18 Yr ‡
UNDERLYING PCV13 PPSV23 REVACCINATION
RISK GROUP MEDICAL
RECOMMENDED RECOMMENDED 5 YR AFTER 1ST
CONDITION DOSE
Immunocompetent Chronic heart disease § ✓
persons Chronic lung disease || ✓
Diabetes mellitus ✓
Cerebrospinal fluid ✓ ✓
leaks
Cochlear implants ✓ ✓
Alcoholism ✓
Chronic liver disease ✓
Cigarette smoking ✓
Persons with Sickle cell disease, ✓ ✓ ✓
functional or anatomic other
asplenia hemoglobinopathies
Congenital or acquired ✓ ✓ ✓
asplenia
Immunocompromised Congenital or acquired ✓ ✓ ✓
persons immunodeficiencies ¶
HIV infection ✓ ✓ ✓
Chronic renal failure ✓ ✓ ✓
Nephrotic syndrome ✓ ✓ ✓
Leukemia ✓ ✓ ✓
Lymphoma ✓ ✓ ✓
Hodgkin disease ✓ ✓ ✓
Generalized ✓ ✓ ✓
malignancy
Iatrogenic ✓ ✓ ✓
immunosuppression**
Solid-organ transplant ✓ ✓ ✓
Multiple myeloma ✓ ✓ ✓
* 13-valent pneumococcal conjugate vaccine.
‡ Children age 2-5 yr with chronic conditions (e.g., heart disease, diabetes),
immunocompromising conditions (e.g., HIV), functional or anatomic asplenia (including sickle cell
disease), cerebrospinal fluid leaks, or cochlear implants, and who have not previously received
PCV13, have been recommended to receive PCV13 since 2010.
§ Including congestive heart failure and cardiomyopathies.
C3, and C4 deficiencies), and phagocytic disorders (excluding chronic granulomatous disease).
** Diseases requiring treatment with immunosuppressive drugs, including long-term systemic
Table 210.1
Definition of Streptococcal Toxic Shock Syndrome
CLINICAL CRITERIA
Hypotension plus 2 or more of the following:
Renal impairment
Coagulopathy
Hepatic involvement
Adult respiratory distress syndrome
Generalized erythematous macular rash
Soft tissue necrosis
DEFINITE CASE
Clinical criteria plus group A streptococcus from a normally sterile site
PROBABLE CASE
Clinical criteria plus group A streptococcus from a nonsterile site
Diagnosis
adherence to the Jones Criteria in 3 circumstances: (1) when chorea occurs as the
only major manifestation of acute RF, (2) when indolent carditis is the only
manifestation in patients who first come to medical attention only months after
the apparent onset of acute RF, and (3) in a limited number of patients with
recurrence of acute RF in particularly high-risk populations.
Table 210.2
Guidelines for the Diagnosis of Initial or Recurrent Attack
of Rheumatic Fever (Jones Criteria, Updated 2015)1-5
MAJOR MINOR SUPPORTING EVIDENCE OF ANTECEDENT GROUP A
MANIFESTATIONS MANIFESTATIONS STREPTOCOCCAL INFECTION
Carditis Clinical Positive throat culture or rapid streptococcal antigen test
Polyarthritis features: Elevated or increasing streptococcal antibody titer
Erythema Arthralgia
marginatum Fever
Subcutaneous Laboratory
nodules features:
Chorea Elevated acute-
phase reactants:
Erythrocyte
sedimentation rate
C-reactive protein
Prolonged P-R
interval
1. Initial attack : 2 major manifestations, or 1 major and 2 minor manifestations, plus evidence of
recent GAS infection. Recurrent attack : 2 major, or 1 major and 2 minor, or 3 minor
manifestations (the latter only in the Moderate/High-Risk population), plus evidence of recent GAS
infection (see text).
2. Low-Risk population is defined as acute rheumatic fever (ARF) incidence <2 per 100,000
school-age children per year, or all-age rheumatic heart disease (RHD) prevalence of <1 per
1,000 population. Moderate/High-Risk population is defined as ARF incidence >2 per 100,000
school-age children per year, or all-age RHD prevalence of >1 per 1,000 population.
3. Carditis is now defined as clinical and/or subclinical (echocardiographic valvulitis). See Table
210.3 .
4. Arthritis (major) refers only to polyarthritis in Low-Risk populations, but also to monoarthritis or
polyarthralgia in Moderate/High-Risk populations.
5. Minor criteria for Moderate/High-Risk populations only include monoarthralgia (polyarthralgia
for Low-Risk populations), fever of >38°C (>38.5°C in Low-Risk populations), ESR >30 mm/hr
(>60 mm/hr in Low-Risk populations).
From Gewitz MH, Baltimore RS, Tani LY, et al: Revision of the Jones Criteria for the diagnosis of
acute rheumatic fever in the era of Doppler echocardiography: a scientific statement from the
American Heart Association, Circulation 131(20):1806–1818, 2015.
Systemic lupus erythematosus (SLE) can usually be distinguished from acute
RF by antinuclear antibodies in SLE. Other causes of arthritis such as pyogenic
arthritis, malignancies, serum sickness, Lyme disease, sickle cell disease, and
reactive arthritis related to gastrointestinal infections (e.g., Shigella, Salmonella,
Yersinia ) should also be considered. Poststreptococcal reactive arthritis is
discussed earlier (see Chapter 210).
Table 210.3
When carditis is the sole major manifestation of suspected acute RF, viral
myocarditis, viral pericarditis, Kawasaki disease, and infective endocarditis
should also be considered. Patients with infective endocarditis may present with
both joint and cardiac manifestations. These patients can usually be
distinguished from patients with acute RF by blood cultures and the presence of
extracardiac findings (e.g., hematuria, splenomegaly, splinter hemorrhages).
When chorea is the sole major manifestation of suspected acute RF, Huntington
chorea, Wilson disease, SLE, and various encephalitides should also be
considered.
Treatment
All patients with acute rheumatic fever should be placed on bed rest and
monitored closely for evidence of carditis. They can be allowed to ambulate
when the signs of acute inflammation have improved. However, patients with
carditis require longer periods of bed rest.
Antibiotic Therapy
Appropriate antibiotic therapy instituted before the 9th day of symptoms of acute
GAS pharyngitis is highly effective in preventing first attacks of acute RF.
However, approximately 30% of patients with acute RF do not recall a preceding
episode of pharyngitis and did not seek therapy.
Secondary Prevention
Secondary prevention is directed at preventing acute GAS pharyngitis in patients
at substantial risk of recurrent acute RF. Secondary prevention requires
continuous antibiotic prophylaxis, which should begin as soon as the diagnosis
of acute RF has been made and immediately after a full course of antibiotic
therapy has been completed. Because patients who have had carditis with their
initial episode of acute RF are at higher risk for having carditis with recurrences
and for sustaining additional cardiac damage, they should receive long-term
antibiotic prophylaxis well into adulthood and perhaps for life (Tables 210.4 and
210.5 ).
Table 210.4
Chemoprophylaxis for Recurrences of Acute Rheumatic
Fever (Secondary Prophylaxis)
Adapted from Gerber MA, Baltimore RS, Eaton CB, et al: Prevention of rheumatic fever and
diagnosis and treatment of acute streptococcal pharyngitis: a scientific statement from the
American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of
the Council on Cardiovascular Disease in the Young, Circulation 119:1541–1551, 2009.
Table 210.5
Duration of Prophylaxis for People Who Have Had Acute Rheumatic Fever: AHA
Recommendations
CATEGORY DURATION
Rheumatic fever without carditis 5 yr or until 21 yr of age, whichever is longer
Rheumatic fever with carditis but without residual heart 10 yr or until 21 yr of age, whichever is longer
disease (no valvular disease* )
Rheumatic fever with carditis and residual heart disease 10 yr or until 40 yr of age, whichever is longer;
(persistent valvular disease* ) sometimes lifelong prophylaxis
* Clinical or echocardiographic evidence.
Adapted from Gerber MA, Baltimore RS, Eaton CB, et al: Prevention of rheumatic fever and
diagnosis and treatment of acute streptococcal pharyngitis: a scientific statement from the
American Heart Association (AHA) Rheumatic Fever, Endocarditis, and Kawasaki Disease
Committee of the Council on Cardiovascular Disease in the Young, Circulation 119:1541–1551,
2009.
Patients who did not have carditis with their initial episode of acute RF have a
relatively low risk for carditis with recurrences. Antibiotic prophylaxis should
continue in these patients until the patient reaches 21 yr of age or until 5 yr have
elapsed since the last rheumatic fever attack, whichever is longer. The decision
to discontinue prophylactic antibiotics should be made only after careful
consideration of potential risks and benefits and of epidemiologic factors such as
the risk for exposure to GAS infections.
The regimen of choice for secondary prevention is a single intramuscular
injection of benzathine penicillin G (600,000 IU for children weighing ≤60 lb
and 1.2 million IU for those >60 lb) every 4 wk (Table 210.4 ). In certain high-
risk patients, and in certain areas of the world where the incidence of rheumatic
fever is particularly high, use of benzathine penicillin G every 3 wk may be
necessary because serum concentrations of penicillin may decrease to marginally
effective levels after 3 wk. In the United States, administration of benzathine
penicillin G every 3 wk is recommended only for those who have recurrent acute
RF despite adherence to a 4 wk regimen. In compliant patients, continuous oral
antimicrobial prophylaxis can be used. Penicillin V (250 mg twice daily) and
sulfadiazine or sulfisoxazole (500 mg for those weighing ≤60 lb or 1,000 mg for
those >60 lb, once daily) are equally effective when used in such patients. For
the exceptional patient who is allergic to both penicillin and sulfonamides, a
macrolide (erythromycin or clarithromycin) or azalide (azithromycin) may be
used. Table 210.5 notes the duration of secondary prophylaxis.
Bibliography
birth. In utero infection may result in septic abortion or immediate distress after
birth. More than 80% of early-onset GBS disease presents as sepsis; pneumonia
and meningitis are other common manifestations. Asymptomatic bacteremia is
uncommon but can occur. In symptomatic patients, nonspecific signs such as
hypothermia or fever, irritability, lethargy, apnea, and bradycardia may be
present. Respiratory signs are prominent regardless of the presence of
pneumonia and include cyanosis, apnea, tachypnea, grunting, flaring, and
retractions. A fulminant course with hemodynamic abnormalities, including
tachycardia, acidosis, and shock, may ensue. Persistent fetal circulation may
develop. Clinically and radiographically, pneumonia associated with early-onset
GBS disease is difficult to distinguish from respiratory distress syndrome .
Patients with meningitis often present with nonspecific findings, as described for
sepsis or pneumonia, with more specific signs of CNS involvement initially
absent.
Table 211.1
Table 211.2
Non–Group A or B Streptococci
David B. Haslam
The genus Streptococcus is exceptionally diverse and includes the major human
pathogens Streptococcus pyogenes (group A streptococcus), Streptococcus
agalactiae (group B streptococcus), and Streptococcus pneumoniae . Other
important pathogens include large-colony species–bearing Lancefield groups C
and G antigens and numerous small-colony variants that may or may not express
Lancefield carbohydrate antigen among the viridians streptococci (Table 212.1 ).
This chapter focuses on Streptococcus dysgalactiae subspecies equisimilis,
commonly known as “group C and G streptococci”; Chapter 209 discusses S.
pneumoniae, and Chapter 213 discusses enterococci.
Table 212.1
Relationship of Streptococci Identified by Hemolysis and
Lancefield Grouping to Sites of Colonization and Disease
Group A Streptococcus Group B Other β-HEMOLYTIC Viridans
(S. pyogenes ) Streptococcus STREPTOCOCCI Streptococci
(S. agalactiae
)
Hemolysis β β β α
Lancefield A B C-H, K-V
group Especially C and G
Species or M types (>180) Serotypes (Ia, Ib, Streptococcus
strains II, III, IV, V, VI, bovis
VII, and VIII) Streptococcus
mitis
Streptococcus
mutans
Streptococcus
sanguis
Many others
Normal Pharynx, skin, anus Gastrointestinal Pharynx, skin, Pharynx,
flora and genitourinary gastrointestinal and nose, skin,
tract genitourinary tracts genitourinary
tract
Common Pharyngitis, tonsillitis, erysipelas, Puerperal sepsis Wound infections, Endocarditis,
human impetigo, septicemia, wound chorioamnionitis, puerperal sepsis, human bite
diseases infections, necrotizing fasciitis, endocarditis, cellulitis, sinusitis, infections
cellulitis, meningitis, pneumonia, neonatal sepsis, endocarditis, brain
scarlet fever, toxic shock–like meningitis, abscess, sepsis,
syndrome, rheumatic fever, acute osteomyelitis, nosocomial infections,
glomerulonephritis pneumonia opportunistic infections
α, Partial hemolysis; β, complete hemolysis; γ, no hemolysis (nonhemolytic).
Table 213.1
Table 213.2
Table 215.1
Types of Listeria monocytogenes Infections
Listeriosis in pregnancy
Neonatal listeriosis
Early onset
Late onset
Food-borne outbreaks/febrile gastroenteritis
Listeriosis in normal children and adults (rare)
Focal Listeria infections (e.g., meningitis, endocarditis, pneumonia, liver abscess, osteomyelitis, septic arthritis)
Listeriosis in immunocompromised persons
Lymphohematogenous malignancies
Collagen vascular diseases
Diabetes mellitus
HIV infection
Transplantation
Renal failure with peritoneal dialysis
Listeriosis in elderly persons
Pathology
Clinical Manifestations
The clinical presentation of listeriosis depends greatly on the age of the patient
and the circumstances of the infection.
Listeriosis in Pregnancy
Pregnant women have increased susceptibility to Listeria infectious
(approximately 20 times higher than nonpregnant women), probably because of
a relative impairment in cell-mediated immunity. L. monocytogenes has been
grown from placental and fetal cultures of pregnancies ending in spontaneous
abortion. The usual presentation in the 2nd and 3rd trimesters is a flulike illness
that may result in seeding of the uterine contents by bacteremia. Rarely is
maternal listeriosis severe, but meningitis in pregnancy has been reported.
Recognition and treatment at this stage are associated with normal pregnancy
outcomes, but the fetus may not be infected even if listeriosis in the mother is
not treated. In other instances, placental listeriosis develops with infection of the
fetus that may be associated with stillbirth or premature delivery. Delivery of an
infected premature fetus is associated with very high infant mortality.
Disseminated disease is apparent at birth, often with a diffuse pustular rash.
Infection in the mother usually resolves without specific therapy after delivery,
but postpartum fever and infected lochia may occur.
Neonatal Listeriosis
Two clinical presentations are recognized for neonatal listeriosis: early-onset
neonatal disease (<5 days, usually within 1-2 days of birth), which is a
predominantly septicemic form, and late onset neonatal disease (>5 days, mean
14 days of life), which is a predominantly meningitic form (Table 215.2 ). The
principal characteristics of the 2 presentations resemble the clinical syndromes
described for group B streptococcus (see Chapter 211 ).
Table 215.2
Characteristic Features of Early- and Late-Onset Neonatal
Listeriosis
FIG. 215.1 Listeria monocytogenes. The generalized maculopapular rash present at
birth disappeared within a few hours of life. (From Benitez-Segura I, Fiol-Jaume M,
Balliu PR, Tejedor M: Listeria monocytogenes : generalized maculopapular rash may be
the clue, Arch Dis Child Fetal Neonatal Ed 98(1):F64, 2013, Fig 1.)
Postneonatal Infections
Listeriosis beyond the newborn period may rarely occur in otherwise healthy
although maternal infection with sparing of the fetus has been reported. There is
no convincing evidence that L. monocytogenes is associated with repeated
spontaneous abortions in humans. The mortality rate is >50% for premature
infants infected in utero, 30% for early-onset neonatal sepsis, 15% for late-onset
neonatal meningitis, and <10% in older children with prompt institution of
appropriate antimicrobial therapy. Mental retardation, hydrocephalus, and other
CNS sequelae are reported in survivors of Listeria meningitis.
Prevention
Listeriosis can be prevented by pasteurization and thorough cooking of foods.
Irradiation of meat products may also be beneficial. Consumption of
unpasteurized or improperly processed dairy products should be avoided,
especially aged soft cheeses, uncooked and precooked meat products that have
been stored at 4°C (39.2°F) for extended periods, and unwashed vegetables
(Table 215.3 ). This avoidance is particularly important during pregnancy and for
immunocompromised persons. Infected domestic animals should be avoided
when possible. Education regarding risk reduction is aimed particularly at
pregnant women and people being treated for cancer.
Table 215.3
Prevention of Food-Borne Listeriosis
GENERAL RECOMMENDATIONS TO PREVENT LISTERIA INFECTION
FDA recommendations for washing and handling food:
• Rinse raw produce, such as fruits and vegetables, thoroughly under running tap water before eating, cutting, or
cooking. Even if the produce will be peeled, it should still be washed first.
• Scrub firm produce, such as melons and cucumbers, with a clean produce brush.
• Dry the produce with a clean cloth or paper towel.
• Separate uncooked meats and poultry from vegetables, cooked foods, and ready-to-eat foods.
Keep your kitchen and environment cleaner and safer.
• Wash hands, knives, countertops, and cutting boards after handling and preparing uncooked foods.
• Be aware that Listeria monocytogenes can grow in foods in the refrigerator. Use an appliance thermometer, such
as a refrigerator thermometer, to check the temperature inside your refrigerator. The refrigerator should be 4.5°C
(40°F) or lower and the freezer −17.8°C (0°F) or lower.
• Clean up all spills in your refrigerator promptly, especially juices from hot dog and lunch meat packages, raw
meat, and raw poultry.
• Clean the inside walls and shelves of your refrigerator with hot water and liquid soap, then rinse.
Cook meat and poultry thoroughly.
• Thoroughly cook raw food from animal sources, such as beef, pork, or poultry to a safe internal temperature. For
a list of recommended temperatures for meat and poultry, visit the safe minimum cooking temperatures chart at
http://www.FoodSafety.gov .
Store foods safely.
• Use precooked or ready-to-eat food as soon as you can. Do not store the product in the refrigerator beyond the
use-by date; follow USDA refrigerator storage time guidelines:
• Hot dogs: store opened package no longer than 1 wk and unopened package no longer than 2 wk in the
refrigerator.
• Luncheon and deli meat: store factory-sealed, unopened package no longer than 2 wk. Store opened packages
and meat sliced at a local deli no longer than 3-5 days in the refrigerator.
• Divide leftovers into shallow containers to promote rapid, even cooling. Cover with airtight lids or enclose in
plastic wrap or aluminum foil. Use leftovers within 3-4 days.
Choose safer foods.
• Do not drink raw (unpasteurized) milk, and do not eat foods that have unpasteurized milk in them.
RECOMMENDATIONS FOR PERSONS AT HIGHER RISK *
In addition to the recommendations listed above, include:
Meats
• Do not eat hot dogs, luncheon meats, cold cuts, other deli meats (e.g., bologna), or fermented or dry sausages
unless they are heated to an internal temperature of 73.9°C (165°F) or until steaming hot just before serving.
• Avoid getting fluid from hot dog and lunch meat packages on other foods, utensils, and food preparation
surfaces, and wash hands after handling hot dogs, luncheon meats, and deli meats.
• Pay attention to labels. Do not eat refrigerated pâté or meat spreads from a deli or meat counter or from the
refrigerated section of a store. Foods that do not need refrigeration, such as canned or shelf-stable pâté and meat
spreads, are safe to eat. Refrigerate after opening.
Cheeses
• Do not eat soft cheese such as feta, queso blanco, queso fresco, brie, Camembert, blue-veined, or panela (queso
panela) unless it is labeled as made with pasteurized milk. Make sure the label says “MADE WITH
PASTEURIZED MILK.”
Seafood
• Do not eat refrigerated smoked seafood, unless it is contained in a cooked dish, such as a casserole, or unless it
is a canned or shelf-stable product.
• Refrigerated smoked seafood, such as salmon, trout, whitefish, cod, tuna, and mackerel, is most often labeled as
“nova-style,” “lox,” “kippered,” “smoked,” or “jerky.”
• These fish are typically found in the refrigerator section or sold at seafood and deli counters of grocery stores
and delicatessens.
• Canned and shelf stable tuna, salmon, and other fish products are safe to eat.
Follow this general FDA advice for melon safety:
• Consumers and food preparers should wash their hands with warm water and soap for at least 20 sec before and
after handling any whole melon, such as cantaloupe, watermelon, or honeydew.
• Scrub the surface of melons, such as cantaloupes, with a clean produce brush under running water and dry them
with a clean cloth or paper towel before cutting. Be sure that your scrub brush is sanitized after each use, to
avoid transferring bacteria between melons.
• Promptly consume cut melon or refrigerate promptly. Keep your cut melon refrigerated ≤4.5°C (40°F) (0-1.1°C
[32-34°F] is best), for no more than 7 days.
• Discard cut melons left at room temperature for >4 hr.
* Including pregnant women, persons with weakened immune system, and older adults.
Table 218.1
Prevalence of Symptoms and Signs in Children and Young
People With Meningococcal Septicemia, Meningococcal
Disease, and Bacterial Meningitis
†
The age ranges in the 4 studies are 0-14 yr, 0-2 yr, 0-12 mo, and 0-13 wk.
‡ One study reported the number of children and young people with ear, nose, and throat
infections; the 4 other studies reported the number of ear infections only.
Classification of conditions presented in the table reflects the terminology used in the evidence.
N/A, Not applicable.
Adapted from National Collaborating Center for Women's and Children's Health (UK): Bacterial
meningitis and meningococcal septicaemia: management of bacterial meningitis and
meningococcal septicaemia in children and young people younger than 16 years in primary and
secondary care, NICE clinical guidelines, No 102, London, 2010, RCOG Press.
Antibiotics
Empirical antimicrobial therapy should be initiated immediately after the
diagnosis of invasive meningococcal infection is suspected and cultures are
obtained, using a third-generation cephalosporin to cover the most likely
bacterial pathogens until the diagnosis is confirmed. In regions with a high rate
of β-lactam–resistant S. pneumoniae , empirical addition of intravenous (IV)
vancomycin is recommended (see Chapter 621.1 ) while awaiting the outcome
of bacterial identification and sensitivity, but this is unnecessary in other settings
where cephalosporin resistance of pneumococci is very rare (in these settings a
risk assessment of each case should be made). Once the diagnosis of β-lactam–
sensitive meningococcal disease is confirmed in the laboratory, some authorities
recommend a switch to penicillin. Even with no evidence that survival outcomes
are different, however, limited evidence from one study indicates that, in
meningococcal purpura, necrotic skin lesions are less common among children
treated with ceftriaxone than with penicillin. Furthermore, it may be cost-
effective by using a once-daily dose of ceftriaxone for therapy in younger
children, and this is the recommended practice in the United Kingdom (Table
218.2 ). No adequate studies have investigated the optimal duration of therapy
for children, but the course is generally continued for 5-7 days.
Table 218.2
Treatment of Neisseria Meningitidis Invasive Infections
DOSING MAXIMUM
DRUG ROUTE DOSE INTERVAL DAILY NOTES
(hr) DOSE
Penicillin G IM or 300,000 4-6 12-24 Does not clear carriage, and
IV units/kg/day million units “prophylaxis” is required at the end of
treatment.
Ampicillin IM or 200-400 6 6-12 g Does not clear carriage, and
IV mg/kg/day “prophylaxis” is required at the end of
treatment.
Cefotaxime IM or 200-300 6-8 8-12 g Recommended in the neonate
IV mg/kg/day
Ceftriaxone IM or 100 12-24 2-4 g
Preferred treatment as only once or
IV mg/kg/day twice daily, and may reduce skin
complications.
ALTERNATIVE THERAPY IN THE FACE OF LIFE-THREATENING β-LACTAM ALLERGY
Chloramphenicol* IV 50-100 6 2-4 g
mg/kg/day
Meropenem † IV 60-120 8 1.5-6 g
mg/kg/day
Prevention
Secondary Prevention
Close contacts of patients with meningococcal disease are at increased risk of
infection because such individuals are likely to be colonized with the index
case's (hyperinvasive) strain. Antibiotic prophylaxis should be offered as soon as
possible to individuals who have been exposed directly to a patient's oral
secretions, for whom risk may be 1,000 times the background rate in the
population. This includes household, kissing, and close family contacts of cases,
as well as childcare and recent preschool contacts in the United States. Up to
30% of cases occur in the 1st wk, but risk persists for up to 1 yr after
presentation of the index case. Although prophylaxis is effective in preventing
secondary cases, co-primary cases may occur in the days after presentation of
the index case, and contacts should be carefully evaluated if they develop
symptoms. Advice on management of nonclose contacts, such as those in
daycare, nursery settings, or school and other institutions, varies in different
countries because the risk of a secondary case in this situation is low and opinion
on risk assessment varies. Ceftriaxone and ciprofloxacin are the most effective
agents for prophylaxis, with ciprofloxacin the drug of choice in some countries.
Rifampin is most widely used but fails to eradicate colonization in 15% of cases
(Table 218.3 ). Prophylaxis is not routinely recommended for medical personnel
except those with exposure to aerosols of respiratory secretions, such as through
mouth-to-mouth resuscitation, intubation, or suctioning before or in the 24 hr
after antibiotic therapy is initiated in the index case.
Table 218.3
Antibiotic Prophylaxis to Prevent Neisseria Meningitidis
Infection*
Table 218.4
Recommendations for Meningococcal Vaccination (United
States, 2017)
GENERAL POPULATION
<2 YR 2-10 YR 11-18 YR 19-55 YR
Not recommended Not A single dose of MenACWY-D or Not recommended
recommended MenACWY-CRM at age 11-12 yr with a
booster dose at age 16 yr
SPECIAL POPULATIONS AT INCREASED RISK OF MENINGOCOCCAL DISEASE
RISK FACTOR 2-18 7-23 MONTHS 2-55 YR
MONTHS
Persistent complement 4 doses of 2 doses of MenACWY-CRM, with 2nd dose 2 doses of
deficiencies, MenACWY- administered at age ≥12 mo and ≥3 mo after MenACWY-CRM or
functional or anatomic CRM at 2, 4, 1st dose, or 2 doses of MenACWY-D (not MenACWY-D 8-12
asplenia 6, and 12-15 indicated for functional or anatomic asplenia wk apart* §
mo* ‡ ) at age 9 and 12 mo* † and MenB vaccine (2
doses of 4CMenB or 3
doses of 2fHbp) †
At risk during a 4 doses of 2 doses of MenACWY-CRM, with 2nd dose 1 dose of MenACWY-
community outbreak MenACWY- administered at age ≥12 mo and ≥3 mo after CRM or MenACWY-
with a vaccine CRM at 2, 4, 1st dose, or 2 doses of MenACWY-D at age 9 D
capsular group 6, and 12-15 and 12 mo † MenB vaccine (2
covered by the mo doses of 4CMenB or 3
relevant vaccine doses of 2fHbp)
depending on capsular
group associated with
the outbreak †
Travel to or resident 4 doses of 2 doses of MenACWY-CRM, with 2nd dose 1 dose of MenACWY-
of countries where MenACWY- administered at age ≥12 mo and ≥3 mo after CRM or MenACWY-D* †
meningococcal CRM at 2, 4, 1st dose, or 2 doses of MenACWY-D at 9
disease is 6, and 12-15 and age 12 mo (could be reduced to 8 wk if
hyperendemic or mo* required for travel)* †
epidemic ǁ
Have HIV 4 doses of 2 doses of MenACWY-CRM or 2 doses of 2 doses of MenACWY-
MenACWY- MenACWY-D at age 9-23 mo, 12 wk apart* † CRM or MenACWY-D 8-
CRM at 2, 4, 12 wk apart* † ǁ
6, and 12-15
mo*
Other risk factors — — 1 dose MenACWY
* Booster every 5 yr if ongoing risk (after 3 yr if <7 yr old).
‡ Because of high risk for invasive pneumococcal disease, children with functional or anatomic
asplenia should not be immunized with MenACWY-D before age 2 yr, to avoid interference with
the immune response to the pneumococcal conjugate vaccine (PCV).
§ If MenACWY-D is used, it should be administered at least 4 wk. after completion of all PCV
doses.
ǁ For example, visitors to the “meningitis belt” of sub-Saharan Africa. Vaccination also is required
by the government of Saudi Arabia for all travelers to Mecca during the annual Hajj.
† Assuming not previously vaccinated.
Table 219.1
Recommended Treatment of Gonococcal Infections
LENGTH
INFECTION TREATMENT REGIMEN OF
THERAPY
Neonates Ophthalmia Ceftriaxone,* 25-50 mg/kg IV or IM (max 250 mg), plus Once
neonatorum lavage infected eye frequently until discharge eliminated
Disseminated Ceftriaxone,* 25-50 mg/kg IV or IM qd 7 days
infection or
Scalp abscess Cefotaxime, 25-50 mg/kg IV or IM q8–12h †
Septic
arthritis
Meningitis Ceftriaxone,* 25-50 mg/kg IV or IM qd 10-14 days
or
Cefotaxime, 25-50 mg/kg IV or IM q8-12h †
Endocarditis Ceftriaxone,* 25-50 mg/kg IV or IM qd Minimum 28
or days
Cefotaxime, 25-50 mg/kg IV or IM q8-12h †
Children ≤45 kg Pharyngeal Ceftriaxone, 25-50 mg/kg IV or IM (max 250 mg) Once
infection
Anorectal
infection
Urogenital
infection
Conjunctivitis Ceftriaxone, 50 mg/kg IM (max 1 g) ‡ Once
Disseminated Ceftriaxone, 50 mg/kg IV or IM qd (max 1 g daily) 7 days
infection
Septic
arthritis
Meningitis Ceftriaxone, 50 mg/kg IV or IM q12-24h (max 4 g daily) 10-14 days
Endocarditis Ceftriaxone, 50 mg/kg IV or IM q12-24h (max 4 g daily) Minimum 28
days
Adults, adolescents, Pharyngeal Ceftriaxone, 250 mg IM Once
and children >45 kg infection plus
Anorectal Azithromycin, 1 g PO
infection
Urogenital
infection
Conjunctivitis Ceftriaxone, 1 g IM Once
plus
Azithromycin, 1 g PO ‡
Disseminated Ceftriaxone, 1 g IV or IM qd § 7 days
infection plus
Septic Azithromycin, 1 g PO Once
arthritis
Meningitis Ceftriaxone, 1-2 g IV q12-24h 10-14 days
plus
Azithromycin, 1 g PO Once
Endocarditis Ceftriaxone, 1-2 g IV q12-24h Minimum 28
plus days
Azithromycin, 1 g PO Once
* When available, cefotaxime should be substituted for ceftriaxone in neonates with
hyperbilirubinemia (particularly those who are premature) and in those <28 days old if receiving
calcium-containing intravenous fluids. Consult neonatal dosing references.
† Dose and/or dosing frequency change after postnatal age >7 days. Consult neonatal dosing
references.
‡ Plus lavage of the infected eye with saline solution (once).
§ Ceftriaxone should be continued for 24-48 hr after clinical improvement begins, at which time
the switch may be made to an oral agent (e.g., cefixime or a quinolone) if antimicrobial
susceptibility is documented by culture. If no organism is isolated and the diagnosis is secure,
treatment with ceftriaxone should be continued for at least 7 days.
IM, Intramuscularly; IV, intravenously; PO, orally (by mouth); qd, every day; q8-12h, every 8 to 12
hours.
From Hsu KK, Wangu Z: Neisseria gonorrhoeae. In Long SS, Prober CG, Fischer M, editors:
Principles and practice of pediatric infectious diseases, ed 5, Philadelphia, 2018, Elsevier, Table
126.1.
Alternative regimens exist for adolescents and adults but are extremely
limited. For patients with cephalosporin allergy, the combination of gentamicin
(240 mg intramuscularly [IM]) plus azithromycin (2 g orally [PO]) cured 100%
mo to 3 yr old in at least some countries. With the exception of patients with
endocarditis, the presentation of invasive K. kingae infections is frequently mild,
and a body temperature <38°C (100.4°F), a normal C-reactive protein (CRP)
level, and a normal white blood cell (WBC) count are common, requiring a high
index of clinical suspicion.
Table 220.1
Septic Arthritis
Although K. kingae –driven arthritis especially affects the large, weight-bearing
joints, involvement of the small metacarpophalangeal, sternoclavicular, and
tarsal joints is not unusual (see Chapter 704 ). The disease has an acute
presentation, and children are brought to medical attention after a median of 3
days. The leukocyte count in the synovial fluid shows <50,000 WBCs/µL in
almost 25% of the patients, and the Gram stain of synovial fluid is positive in
in the immunologic response to thymus-independent type 2 antigens such as
unconjugated PRP, presumably explaining the high incidence of type b
infections in infants and young children in the prevaccine era.
The conjugate vaccines act as thymus-dependent antigens and elicit serum
antibody responses in infants and young children (Table 221.1 ). These vaccines
are believed to prime memory antibody responses on subsequent encounters with
PRP. The concentration of circulating anti-PRP antibody in a child primed by a
conjugate vaccine may not correlate precisely with protection, presumably
because a memory response may occur rapidly on exposure to PRP and provide
protection.
Table 221.1
Haemophilus influenzae Type b (Hib) Conjugate Vaccines
Available in the United States
Diagnosis
Presumptive identification of H. influenzae is established by direct examination
of the collected specimen after staining with Gram reagents. Because of its small
size, pleomorphism, and occasional poor uptake of stain, as well as the tendency
for proteinaceous fluids to have a red background, H. influenzae is sometimes
cardiac, pulmonary, muscular, or neurologic disorders have increased risk of
poor outcome beyond infancy. Table 224.1 lists caveats in assessment and care
of infants with pertussis. The specific, limited goals of hospitalization are to (1)
assess progression of disease and likelihood of life-threatening events at peak of
disease; (2) maximize nutrition; (3) prevent or treat complications; and (4)
educate parents in the natural history of the disease and in care that will be given
at home. Heart rate, respiratory rate, and pulse oximetry are monitored
continuously with alarm settings so that paroxysms can be witnessed and
recorded by healthcare personnel. Detailed cough records and documentation of
feeding, vomiting, and weight change provide data to assess severity. Typical
paroxysms that are not life threatening have the following features: duration <45
sec; red but not blue color change; tachycardia, bradycardia (not <60 beats/min
in infants), or oxygen desaturation that spontaneously resolves at the end of the
paroxysm; whooping or strength for brisk self-rescue at the end of the paroxysm;
self-expectorated mucus plug; and posttussive exhaustion but not
unresponsiveness. Assessing the need to provide oxygen, stimulation, or
suctioning requires skilled personnel who can watchfully observe an infant's
ability for self-rescue but who will intervene rapidly and expertly when
necessary. The benefit of a quiet, dimly lighted, undisturbed, comforting
environment cannot be overestimated or forfeited in a desire to monitor and
intervene. Feeding children with pertussis is challenging. The risk of
precipitating cough by nipple feeding does not warrant nasogastric, nasojejunal,
or parenteral alimentation in most infants. The composition or thickness of
formula does not affect the quality of secretions, cough, or retention. Large-
volume feedings are avoided.
Table 224.1
Caveats in Assessment and Care of Infants With Pertussis
• Infants with potentially fatal pertussis may appear well between episodes.
• A paroxysm must be witnessed before a decision is made between hospital and home care.
• Only analysis of carefully compiled cough record permits assessment of severity and progression of illness.
• Suctioning of nose, oropharynx, or trachea should not be performed on a “preventive” schedule.
• Feeding in the period following a paroxysm may be more successful than after napping.
• Family support begins at the time of hospitalization with empathy for the child's and family's experience to date,
transfer of the burden of responsibility for the child's safety to the healthcare team, and delineation of
assessments and treatments to be performed.
• Family education, recruitment as part of the team, and continued support after discharge are essential.
Within 48-72 hr, the direction and severity of disease are obvious from
analysis of recorded information. Hospital discharge is appropriate if, over 48 hr,
disease severity is unchanged or diminished, intervention is not required during
paroxysms, nutrition is adequate, no complication has occurred, and parents are
adequately prepared for care at home. Apnea and seizures occur in the
incremental phase of illness and in patients with complicated disease. Portable
oxygen, monitoring, or suction apparatus should not be needed at home.
Infants who have apnea, paroxysms that lead to life-threatening events, or
respiratory failure require escalating respiratory support and frequently require
intubation and pharmaceutically induced paralysis.
Antibiotics
An antimicrobial agent always is given when pertussis is suspected or confirmed
to decrease contagiousness and to afford possible clinical benefit. Azithromycin
is the drug of choice in all age-groups, for treatment or postexposure
prophylaxis (Table 224.2 ). Macrolide resistance has been reported rarely, and
recent isolates have retained susceptibility despite genetic strain adaptations.
Infantile hypertrophic pyloric stenosis (IHPS) is associated with macrolide
use in young infants, especially in those <14 days old, with highest risk in those
receiving erythromycin vs azithromycin. Benefits of postexposure prophylaxis
or treatment of infants far outweigh risk of IHPS. Young infants should be
managed expectantly if projectile vomiting occurs. The FDA also warns of risk
of fatal heart rhythms with use of azithromycin in patients already at risk for
cardiovascular events, especially those with prolongation of the QT interval.
Trimethoprim-sulfamethoxazole (TMP-SMX) is an alternative to azithromycin
for infants >2 mo old and children unable to receive azithromycin. Because of
limited effectiveness, treatment of B. parapertussis is based on clinical judgment
and is considered in high-risk populations. Agents are the same as for B.
pertussis . Treatment of infections caused by other Bordetella spp. should be
undertaken with consultation of a subspecialist.
Table 224.2
Recommended Antimicrobial Treatment and Postexposure
Prophylaxis for Pertussis
in patients ≥2 mo old who are allergic to macrolides, who cannot tolerate macrolides, or who are
infected with a rare macrolide-resistant strain of Bordetella pertussis .
Adapted from Centers for Disease Control and Prevention (CDC): Recommended antimicrobial
agents for treatment and postexposure prophylaxis of pertussis: 2005 CDC guidelines, MMWR
54:1–16, 2005.
Adjunct Therapies
No rigorous clinical trial has demonstrated a beneficial effect of β2 -adrenergic
stimulants such as salbutamol and albuterol. Fussing associated with aerosol
treatment triggers paroxysms. No randomized, blinded clinical trial of sufficient
size has been performed to evaluate the usefulness of corticosteroids in the
management of pertussis; their clinical use is not warranted. A randomized,
double-blind, placebo-controlled trial of pertussis immunoglobulin intravenous
(IGIV) was halted prematurely because of expiration/lack of additional supply of
study product; there was no indication of clinical benefit. Standard
immunoglobulin has not been studied and should not be used for treatment or
prophylaxis.
alteration in the expression of a number of host genes, including those encoding
proinflammatory mediators (inducible nitric oxide synthase, chemokines, IL-1β),
receptors or adhesion molecules (tumor necrosis factor [TNF]-α receptor, CD40,
intercellular adhesion molecule 1), and antiinflammatory mediators
(transforming growth factor-β1, TGF-β2). Other upregulated genes include those
involved in cell death or apoptosis (intestinal epithelial cell protease, TNF-R1,
Fas) and transcription factors (early growth response 1, interferon [IFN]
regulatory factor 1). S. Typhimurium can induce rapid macrophage death in
vitro, which depends on the host cell protein caspase-1 and is mediated by the
effector protein SipB (Salmonella invasion protein B). Intracellular S.
Typhimurium is found within specialized vacuoles that have diverged from the
normal endocytic pathway. This ability to survive within
monocytes/macrophages is essential for S. Typhimurium to establish a systemic
infection in the mouse. The mucosal proinflammatory response to S.
Typhimurium infection and the subsequent recruitment of phagocytic cells to the
site may also facilitate systemic spread of the bacteria.
Some virulence traits are shared by all salmonellae, but others are serotype
restricted. These virulence traits have been defined in tissue culture and murine
models, and it is likely that clinical features of human Salmonella infection will
eventually be related to specific DNA sequences. With most diarrhea-associated
nontyphoidal salmonelloses, the infection does not extend beyond the lamina
propria and the local lymphatics. Specific virulence genes are related to the
ability to cause bacteremia. These genes are found significantly more often in
strains of S. Typhimurium isolated from the blood than in strains recovered from
stool. Although both S. dublin and S. choleraesuis have a greater propensity to
rapidly invade the bloodstream with little or no intestinal involvement, the
development of disease after infection with Salmonella depends on the number
of infecting organisms, their virulence traits, and several host defense factors.
Various host factors may also affect the development of specific complications
or clinical syndromes (Table 225.1 ); of these factors, HIV infections are
assuming greater importance in Africa in all age-groups.
Table 225.1
Host Factors and Conditions Predisposing to Development
of Systemic Disease with Nontyphoidal Salmonella (NTS)
Strains
Neonates and young infants (≤3 mo old)
HIV/AIDS
Other immunodeficiencies and chronic granulomatous disease
Defects in interferon γ production or action
Immunosuppressive and corticosteroid therapies
Malignancies, especially leukemia and lymphoma
Hemolytic anemia, including sickle cell disease, malaria, and bartonellosis
Collagen vascular disease
Inflammatory bowel disease
Achlorhydria or use of antacid medications
Impaired intestinal motility
Schistosomiasis, malaria
Malnutrition
Clinical Manifestations
Acute Enteritis
The most common clinical presentation of salmonellosis is acute enteritis. After
an incubation period of 6-72 hr (mean: 24 hr), there is an abrupt onset of nausea,
vomiting, and crampy abdominal pain, located primarily in the periumbilical
area and right lower quadrant, followed by mild to severe watery diarrhea and
sometimes by diarrhea containing blood and mucus. A large proportion of
children with acute enteritis are febrile, although younger infants may exhibit a
normal or subnormal temperature. Symptoms usually subside within 2-7 days in
healthy children, and fatalities are rare. However, some children experience
severe disease with a septicemia-like picture (high fever, headache, drowsiness,
confusion, meningismus, seizures, abdominal distention). The stool typically
contains a moderate number of PMNs and occult blood. Mild leukocytosis may
necessary to perform susceptibility tests on all human isolates. Infections with
suspected drug-resistant Salmonella should be closely monitored and treated
with appropriate antimicrobial therapy.
Table 225.2
Treatment of Salmonella Gastroenteritis
ORGANISM AND INDICATION
Salmonella infections in infants <3 mo old or in immunocompromised persons (in addition to appropriate
treatment for underlying disorder)
DOSE AND DURATION OF TREATMENT
Cefotaxime, † 100-200 mg/kg/day every 6-8 hr for 5-14 days*
or
Ceftriaxone, 75 mg/kg/day once daily for 7 days*
or
Ampicillin, 100 mg/kg/day every 6-8 hr for 7 days*
or
Cefixime, 15 mg/kg/day for 7-10 days*
* A blood culture should be obtained prior to antibiotic therapy. In a well appearing
immunocompetent child without evidence of disseminated disease, a single dose of ceftriaxone
may be given followed by oral azithromycin; ampicillin, trimethoprim-sulfamethoxazole, or a
fluoroquinolone may be substituted once sensitivities are known.
† If available.
Prognosis
Most healthy children with Salmonella gastroenteritis recover fully. However,
malnourished children and children who do not receive optimal supportive
treatment are at risk for development of prolonged diarrhea and complications.
Young infants and immunocompromised patients often have systemic
involvement, a prolonged course, and extraintestinal foci. In particular, children
with HIV infection and Salmonella infections can have a florid course.
After infection, NTS are excreted in feces for a median of 5 wk. A prolonged
carrier state after nontyphoidal salmonellosis is rare but may be seen in children,
particularly those with biliary tract disease and cholelithiasis after chronic
hemolysis. During the period of Salmonella excretion, the individual may infect
others, directly by the fecal-oral route or indirectly by contaminating foods.
Prevention
Control of the transmission of Salmonella infections to humans requires control
of the infection in the animal reservoir, judicious use of antibiotics in dairy and
livestock farming, prevention of contamination of foodstuffs prepared from
animals, and use of appropriate standards in food processing in commercial and
private kitchens. Because large outbreaks are often related to mass food
production, it should be recognized that contamination of just one piece of
machinery used in food processing may cause an outbreak; meticulous cleaning
of equipment is essential. Clean water supply and education in handwashing and
food preparation and storage are critical to reducing person-to-person
transmission. Salmonella may remain viable when cooking practices prevent
food from reaching a temperature >65.5°C (150°F) for >12 min. Parents should
be advised of the risk of various pets(classically including reptiles and
amphibians but also rodents) and be given recommendations for preventing
transmission from these frequently infected hosts (Table 225.3 ).
Table 225.3
Recommendations for Preventing Transmission of
Salmonella from Reptiles and Amphibians to Humans
Pet store owners, healthcare providers, and veterinarians should provide information to owners and potential
purchasers of reptiles and amphibians about the risks for and prevention of salmonellosis from these pets.
Persons at increased risk for infection or serious complications from salmonellosis (e.g., children <5 yr old,
immunocompromised persons) should avoid contact with reptiles and amphibians and any items that have been
in contact with reptiles and amphibians.
Reptiles and amphibians should be kept out of households that include children <5 yr old or immunocompromised
persons. A family expecting a child should remove any pet reptile or amphibian from the home before the infant
arrives.
Reptiles and amphibians should not be allowed in childcare centers.
Persons should always wash their hands thoroughly with soap and water after handling reptiles and amphibians or
their cages.
Reptiles and amphibians should not be allowed to roam freely throughout a home or living area.
Pet reptiles and amphibians should be kept out of kitchens and other food preparation areas. Kitchen sinks should
not be used to bathe reptiles and amphibians or to wash their dishes, cages, or aquariums. If bathtubs are used
for these purposes, they should be cleaned thoroughly and disinfected with bleach.
Reptiles and amphibians in public settings (e.g., zoos, exhibits) should be kept from direct or indirect contact with
patrons except in designated “animal contact” areas equipped with adequate handwashing facilities. Food and
drink should not be allowed in animal contact areas.
From Centers for Disease Control and Prevention: Reptile-associated salmonellosis—selected
states, 1998–2002, MMWR 52:1206–1210, 2003.
Table 225.4
Complications
Although altered liver function is found in many patients with enteric fever,
clinically significant hepatitis, jaundice, and cholecystitis are relatively rare and
may be associated with higher rates of adverse outcome. Intestinal hemorrhage
(<1%) and perforation (0.5–1%) are infrequent among children. Intestinal
perforation may be preceded by a marked increase in abdominal pain (usually in
the right lower quadrant), tenderness, vomiting, and features of peritonitis.
Intestinal perforation and peritonitis may be accompanied by a sudden rise in
pulse rate, hypotension, marked abdominal tenderness and guarding, and
subsequent abdominal rigidity. A rising white blood cell count with a left shift
and free air on abdominal radiographs may be seen in such cases.
Rare complications include toxic myocarditis, which may manifest as
arrhythmias, sinoatrial block, or cardiogenic shock (Table 225.5 ). Neurologic
complications are also relatively uncommon among children; they include
delirium, psychosis, increased intracranial pressure, acute cerebellar ataxia,
chorea, deafness, and Guillain-Barré syndrome. Although case fatality rates may
be higher with neurologic manifestations, recovery usually occurs with no
sequelae. Other reported complications include fatal bone marrow necrosis, DIC,
hemolytic-uremic syndrome, pyelonephritis, nephrotic syndrome, meningitis,
endocarditis, parotitis, orchitis, and suppurative lymphadenitis.
Table 225.5
Extraintestinal Infectious Complications of Typhoid Fever
Caused by Salmonella enterica Serotype Typhi
ORGAN PREVALENCE
RISK FACTORS COMPLICATIONS
SYSTEM (%)
Central 3-35 Residence in endemic region, Encephalopathy, cerebral edema,
nervous malignancy, endocarditis, congenital subdural empyema, cerebral abscess,
system heart disease, paranasal sinus meningitis, ventriculitis, transient
infections, pulmonary infections, Parkinsonism, motor neuron disorders,
meningitis, trauma, surgery, ataxia, seizures, Guillain-Barré
osteomyelitis of skull syndrome, psychosis
Cardiovascular 1-5 Cardiac abnormalities—e.g., Endocarditis, myocarditis, pericarditis,
system existing valvular abnormalities, arteritis, congestive heart failure
rheumatic heart disease, congenital
heart defects
Pulmonary 1-6 Residence in endemic region, past Pneumonia, empyema, bronchopleural
system pulmonary infection, sickle cell fistula
anemia, alcohol abuse, diabetes,
HIV infection
Bone and joint <1 Sickle cell anemia, diabetes, Osteomyelitis, septic arthritis
systemic lupus erythematosus,
lymphoma, liver disease, previous
surgery or trauma, extremes of age,
corticosteroid use
Hepatobiliary 1-26 Residence in endemic region, Cholecystitis, hepatitis, hepatic
system pyogenic infections, intravenous abscesses, splenic abscess, peritonitis,
drug use, splenic trauma, HIV, paralytic ileus
hemoglobinopathy
Genitourinary <1 Urinary tract abnormalities, pelvic Urinary tract infection, renal abscess,
system pathology, systemic abnormalities pelvic infections, testicular abscess,
prostatitis, epididymitis
Soft tissue At least 17 cases Diabetes Psoas abscess, gluteal abscess,
infections reported in cutaneous vasculitis
English-
language
literature
Hematologic At least 5 cases Hemophagocytosis syndrome
reported in
English-
language
literature
From Huang DB, DuPont HL: Problem pathogens: extra-intestinal complications of Salmonella
enterica serotype Typhi infection, Lancet Infect Dis 5:341–348, 2005.
Diagnosis
The mainstay of the diagnosis of typhoid fever is a positive result of culture from
the blood or another anatomic site. Results of blood cultures are positive in 40–
60% of the patients seen early in the course of the disease, and serial blood
cultures may be required to identify Salmonella bacteremia. Stool and urine
viral infections such as Dengue fever, acute hepatitis, and infectious
mononucleosis.
Infection by Salmonella in general, and typhoid or paratyphoid fever in
particular, should be thoroughly considered in the differential diagnosis and
workup for fever in a returned traveler.
Treatment
An early diagnosis of typhoid fever and institution of appropriate treatment are
essential. The vast majority of children with typhoid fever can be managed at
home with oral antibiotics and close medical follow-up for complications or
failure of response to therapy. Patients with persistent vomiting, severe diarrhea,
and abdominal distention may require hospitalization and parenteral antibiotic
therapy.
There are general principles of typhoid fever management. Adequate rest,
hydration, and attention are important to correct fluid and electrolyte imbalance.
Antipyretic therapy (acetaminophen 10-15 mg/kg every 4-6 hr PO) should be
provided as required. A soft, easily digestible diet should be continued unless the
patient has abdominal distention or ileus. Antibiotic therapy is critical to
minimize complications (Table 225.6 ). It has been suggested that traditional
therapy with either chloramphenicol or amoxicillin is associated with relapse
rates of 5–15% and 4–8%, respectively, whereas use of the azithromycin,
quinolones and third-generation cephalosporins is associated with higher cure
rates. The antibiotic treatment of typhoid fever in children is also influenced by
the prevalence of antimicrobial resistance. Over the past 2 decades, emergence
of MDR strains of S. Typhi (i.e., isolates fully resistant to amoxicillin,
trimethoprim-sulfamethoxazole, and chloramphenicol) has necessitated
treatment with fluoroquinolones , which are the antimicrobial drug of choice for
treatment of salmonellosis in adults, with cephalosporins as an alternative. Some
regions are also reporting S. Typhi that produce extended-spectrum β-lactamases.
Fig. 225.7 shows known worldwide distribution patterns of antimicrobial
resistance among S. Typhi isolates.
Table 225.6
Treatment of Typhoid Fever in Children
OPTIMAL THERAPY ALTERNATIVE EFFECTIVE DRUGS
SUSCEPTIBILITY
Daily Dose Daily Dose
Antibiotic Days Antibiotic Days
(mg/kg/day) (mg/kg/day)
UNCOMPLICATED TYPHOID FEVER
Fully sensitive Chloramphenicol 50-75 14- Fluoroquinolone, e.g., 15 5-7*
21 ofloxacin or ciprofloxacin
Amoxicillin 75-100 14
Multidrug resistant Fluoroquinolone 15 5-7 Azithromycin 8-10 7
or
Cefixime 15-20 7-14 Cefixime 15-20 7-14
Quinolone resistant † Azithromycin 8-10 7 Cefixime 20 7-14
or
Ceftriaxone 75 10-
14
SEVERE TYPHOID FEVER
Fully sensitive Fluoroquinolone 15 10- Chloramphenicol 100 14-
(e.g., ofloxacin) 14 21
Amoxicillin 100
Multidrug resistant Fluoroquinolone 15 10- Ceftriaxone 60 10-
14 14
or
Cefotaxime ‡ 80 10-
14
Quinolone resistant Ceftriaxone 60 10- Azithromycin 10-20 7
14
Cefotaxime ‡ 80 10- Fluoroquinolone 20 7-14
14
* A 3-day course is also effective, particularly for epidemic containment.
†
The optimum treatment for quinolone-resistant typhoid fever has not been determined.
Azithromycin, third-generation cephalosporins, or high-dose fluoroquinolones for 10-14 days is
effective.
‡ If available.
Adapted from World Health Organization: Treatment of typhoid fever. In Background document:
the diagnosis, prevention and treatment of typhoid fever. Communicable disease surveillance and
response: vaccines and biologicals, Geneva, 2003, WHO, pp 19–23.
http://whqlibdoc.who.int/hq/2003/WHO_V&B_03.07.pdf .
reduced antigen-specific antibody-secreting cells with late and reduced mucosa
IgA production against Shigella . Less effective adaptive immunity may put
children at more risk for increased disease severity, mortality, and recurrences.
Table 226.1
Table 226.2
Clinical Complications of Shigellosis
INTESTINAL COMPLICATIONS
Rectal prolapse*
Toxic megacolon
Intestinal perforation
Intestinal obstruction
Appendicitis
Persistent diarrhea
EXTRAINTESTINAL COMPLICATIONS
Dehydration
Severe hyponatremia (serum sodium <126 mmol/L)*
Hypoglycemia
Focal infections (e.g., meningitis, osteomyelitis, arthritis, splenic abscesses, vaginitis)
Sepsis, usually in malnourished or immunocompromised persons
Seizure or encephalopathy
Leukemoid reaction (peripheral leukocytes >40 000/µL)*
POSTINFECTIOUS MANIFESTATIONS
Hemolytic-uremic syndrome (HUS)*
Reactive arthritis †
Irritable bowel syndrome (IBS) ‡
Malnutrition
* Significantly more common in episodes with Shigella dysenteriae type 1 than with all other
Shigella spp. among Bangladeshi children younger than 15 yr during the 1990s (rectal prolapse
[52% vs 15%], severe hyponatremia [58% vs 26%], leukemoid reaction [22% vs 2%], and HUS
[8% vs 1%]).
† Typical acute symptoms include asymmetric oligoarthritis (usually lower limb), enthesitis,
dactylitis, and back pain. Extraarticular manifestations include conjunctivitis and uveitis; urethritis
and other genitourinary tract manifestations; oral, skin, and nail lesions; and rarely, cardiac
abnormalities.
‡ IBS follows approximately 4% of Shigella episodes in studies from high-resource settings.
Adapted from Kotloff KL, Riddle MS, Platts-Mills JA, et al: Shigellosis, Lancet 391:801–810, 2018.
Table 227.1
Clinical Characteristics, Pathogenesis, and Diagnosis of
Diarrheagenic E. coli
Table 228.1
Recommended Antimicrobials for Cholera*
RECOMMENDING BODY ANTIBIOTIC OF CHOICE ALTERNATIVE
WHO † (antibiotics Adults Adults
recommended for cases with Doxycycline, 300 mg given as a Erythromycin, 250 mg 4 times a day
severe dehydration) single dose orally (PO) × 3 days PO
or
Tetracycline, 500 mg 4 times a
day × 3 days PO
Children Children
Tetracycline, 12.5 mg/kg/dose 4 Erythromycin, 12.5 mg/kg/dose 4
times a day × 3 days (up to 500 times a day × 3 days (up to 250 mg
mg/dose × 3 days) PO 4 times a day × 3 days) PO
PAHO ‡ (antibiotics Adults Adults
recommended for cases with Doxycycline, 300 mg PO given Ciprofloxacin, 1 g PO single dose
moderate to severe dehydration) as a single dose or
Azithromycin, 1 g PO single dose
(first line for pregnant women)
Children Children
Erythromycin, 12.5 mg/kg/dose Ciprofloxacin, 20 mg/kg PO as a
4 times a day × 3 days (up to single dose
500 mg/dose × 3 days) or
or Doxycycline, 2-4 mg/kg PO as a
Azithromycin, 20 mg/kg as a single dose
single dose (up to 1 g)
* Antibiotic selection must be based on sensitivity patterns of strains of Vibrio cholerae O1 or
O139 in the area.
† Adapted from World Health Organization: The treatment of diarrhea: a manual for physicians
and other senior health workers , 4th revision, Geneva, 2005, WHO.
‡ Adapted from Pan American Health Organization: Recommendations for clinical management of
Prevention
Improved personal hygiene, access to clean water, and sanitation are the
mainstays of cholera control. Appropriate case management substantially
decreases case fatalities to <1%. Travelers from developed countries often have
no prior exposure to cholera and are therefore at risk of developing the disease.
Children traveling to cholera-affected areas should avoid drinking potentially
contaminated water and eating high-risk foods such as raw or undercooked fish
and shellfish. No country or territory requires vaccination against cholera as a
condition for entry.
In 2016, a live oral cholera vaccine, CVD 103 Hg-R (Vaxchora, PaxVax), was
licensed in the United States for use in adults age 18-64 yr traveling to cholera-
affected areas.
Alarmed by the increasing prevalence of cholera, in 2011 the World Health
Assembly recommended the use of oral cholera vaccines to complement existing
water, sanitation, and hygiene initiatives for cholera control. Older-generation
parenteral cholera vaccines have not been recommended by World Health
Organization (WHO) because of the limited protection they confer and their high
reactogenicity. Oral cholera vaccines are safe, are protective for approximately
2-5 yr duration, and confer moderate herd protection. Three oral cholera
vaccines are currently available internationally and recognized by WHO (Table
228.2 ). An internationally licensed killed whole cell oral cholera vaccine with
recombinant B subunit (Dukoral, Crucell) has been available in >60 countries,
including the European Union, and provides protection against cholera in
endemic areas as well as cross-protection against certain strains of
enterotoxigenic E. coli . The 2 other vaccines (Shanchol, Shantha Biotech; and
Euvichol, Eubiologics) are variants of the 1st vaccine and contain the V. cholerae
O1 and O139 antigens but do not contain the B subunit. Without the B subunit,
these vaccines do not require buffer for administration, thereby reducing
administration costs and resources, making them easier to deploy.
Table 228.2
Oral cholera vaccines have been available for >2 decades, and with the WHO
declaration, countries are now using oral cholera vaccines in mass vaccination
campaigns where cholera remains a substantial problem. A cholera vaccine
stockpile, established by WHO, is now available and can be accessed by
countries at risk for cholera, supplementing efforts to lessen the impact of this
ongoing cholera scourge.
Bibliography
Ali M, Nelson AR, Lopez AL, Sack DA. Updated global burden
of cholera in endemic countries. PLoS Negl Trop Dis .
2015;9(6):e0003832.
Clemens JD, Nair GB, Ahmed T, et al. Cholera. Lancet .
2017;390:1539–1548.
Desai SN, Pezzoli L, Martin S, et al. A second affordable oral
cholera vaccine: implications for the global vaccine stockpile.
Lancet Glob Health . 2016;4(4):e223–e224.
Hall V, Medus C, Wahl G, et al. Vibrio cholerae serogroup O1,
serotype Inaba—Minnesota, August 2016. MMWR Morb
Mortal Wkly Rep . 2017;66(36):961–962.
Qadri F, Islam T, Clemons JD. Cholera in Yemen: an old foe
rearing its ugly head. N Engl J Med . 2017;377(21):2005–
2007.
Sauvageot D, Njanpop-Lafourcade B-M, Akilimali L, et al.
Cholera incidence and mortality in sub-Saharan African sites
during multi-country surveillance. PLoS Negl Trop Dis .
2016;10(5):e0004679.
UNICEF. Cholera toolkit . [New York, 20123, UNICEF]
https://www.unicef.org/cholera/index_71222.html .
Williams PCM, Berkley JA. Guidelines for the management of
paediatric cholera infection: a systematic review of the
CHAPTER 229
Campylobacter
Ericka V. Hayes
Etiology
Twenty-six species and 9 subspecies of Campylobacter are recognized (as of
December 2014). Most of these have been isolated from humans, and many are
considered pathogenic. The most significant of these are C. jejuni and C. coli,
which are believed to cause the majority of human enteritis. More than 100
serotypes of C. jejuni have been identified. C. jejuni has been subspeciated into
C. jejuni subsp. jejuni and C. jejuni subsp. doylei. Although C. jejuni subsp.
doylei has been isolated from humans, it is much less common, less hardy, and
more difficult to isolate. Other species, including Campylobacter fetus,
Campylobacter lari, and Campylobacter upsaliensis, have been isolated from
patients with diarrhea, although much less frequently (Table 229.1 ). Emerging
Campylobacter spp. have been implicated in acute gastroenteritis, inflammatory
bowel disease, and peritonitis, including C. concisus , and C. ureolyticus .
Additional Campylobacter spp. have been isolated from clinical specimens, but
their roles as pathogens have not been established.
Table 229.1
Epidemiology
Worldwide, Campylobacter enteritis is a leading cause of acute diarrhea. Efforts
to reduce Campylobacter contamination and use of safe handling practices have
led to decreased incidence. Campylobacter infections can be both food-borne
and water-borne and most frequently result from ingestion of contaminated
poultry (chicken, turkey) or raw milk . Less often, the bacteria come from
communities of bacteria encased in an extracellular matrix that protects the
organisms from the host immune response and the effects of antibiotics. Biofilm
formation requires pilus-mediated attachment to a surface, proliferation of the
organism, and production of exopolysaccharide as the main bacterial component
of the extracellular matrix. A mature biofilm can persist despite an intense host
immune response, is resistant to many antimicrobials, and is difficult to eradicate
with current therapies.
Clinical Manifestations
Most clinical patterns are related to opportunistic infections in
immunocompromised hosts (see Chapter 205 ) or are associated with shunts and
indwelling catheters (Chapter 206 ). P. aeruginosa may be introduced into a
minor wound of a healthy person as a secondary invader, and cellulitis and a
localized abscess that exudes green or blue pus may follow. The characteristic
skin lesions of P. aeruginosa, ecthyma gangrenosum , whether caused by direct
inoculation or a metastatic focus secondary to septicemia, begin as pink macules
and progress to hemorrhagic nodules and eventually to ulcers with ecchymotic
and gangrenous centers with eschar formation, surrounded by an intense red
areola (Table 232.1 and Fig. 232.1 ).
Table 232.1
FIG. 232.1 Round, nontender skin lesion on 2 yr old female's buttock.
Note the black ulcerated center of the lesion and its red margin. (From
Ghanaiem H, Engelhard D: A healthy 2-year-old child with a round black
skin lesion, J Pediatr 163:1225, 2013.)
Table 233.1
Table 233.2
ANTIMICROBIAL DURATION
AGE/CONDITIONS DOSE ROUTE*
AGENT †
≥8 yr Doxycycline 4.4 mg/kg/day divided twice daily; PO ≥6 wk
max 200 mg/day
plus
Rifampin 15-20 mg/kg/day in 1 or 2 divided PO ≥6 wk
doses; max 600-900 mg/day
Alternative:
Doxycycline 4.4 mg/kg/day divided twice daily; PO ≥6 wk
max 200 mg/day
plus
Streptomycin 20-40 mg/kg/day in 2-4 divided IM 2-3 wk
doses; max 1 g/day
or
Gentamicin 6-7.5 mg/kg/day in 3 divided IM/IV 1-2 wk
doses
<8 yr Trimethoprim- TMP (10 mg/kg/day; max 480 PO ≥6 wk
sulfamethoxazole mg/day) and SMX (50 mg/kg/day;
(TMP-SMX) max 2.4 g/day)
plus
Rifampin 15-20 mg/kg/day in 1 or 2 divided PO ≥6 wk
doses; max 600-900 mg/day
Meningitis, Doxycycline 4.4 mg/kg/day divided twice daily; PO ≥4-6 mo
osteomyelitis/spondylitis max 200 mg/day
endocarditis plus
Gentamicin 6-7.5 mg/kg/day in 3 divided IV 1-2 wk
doses
plus
Rifampin 15-20 mg/kg/day in 1 or 2 divided PO ≥4-6 mo
doses; max 600-900 mg/day
* PO, Oral (by mouth): IM, intramuscular; IV, intravenous.
† Longer courses of therapy may be needed for more severe or complicated cases.
Bartonella
Rachel C. Orscheln
The spectrum of disease resulting from human infection with Bartonella species
includes the association of bacillary angiomatosis and cat-scratch disease with
Bartonella henselae. There are more than 30 validated species of Bartonella ,
but 6 major species are responsible for most human disease: B. henselae,
B.quintana, B. bacilliformis, B. elizabethae, B. vinsonii, and B. clarridgeiae
(Table 236.1 ). The remaining Bartonella spp. have been found primarily in
animals, particularly rodents and moles. However, zoonotic infections from
animal-associated strains of Bartonella spp. have been reported. In 2013 a novel
Bartonella agent with the proposed name Candidatus Bartonella ancashi
(Bartonella ancashensis) was described as a cause of verruga peruana .
Table 236.1
Bartonella Species Causing Majority of Human Disease
236.1
Cat-Scratch Disease (Bartonella
henselae)
Rachel C. Orscheln
Keywords
Bartonella
B. henselae
CSD
encephalopathy
endocarditis
flea
granuloma
hepatosplenomegaly
inoculation papule
lymphadenitis
Parinaud oculoglandular syndrome
stellate macular retinopathy
Table 237.1
Diagnoses Considered in Subsequently Laboratory-
Confirmed Cases of Infant Botulism
Differential Diagnosis
Botulism is frequently misdiagnosed, most often as a polyradiculoneuropathy
(Guillain-Barré or Miller Fisher syndrome), myasthenia gravis, or a central
nervous system (CNS) disease (Table 237.2 ). In the United States, botulism is
more likely than Guillain-Barré syndrome , intoxication, or poliomyelitis to
cause a cluster of cases of acute flaccid paralysis. Botulism differs from other
flaccid paralyses in its initial and prominent cranial nerve palsies that are
disproportionate to milder weakness and hypotonia below the neck; in its
symmetry; and in its absence of sensory nerve damage. Spinal muscular atrophy
may closely mimic infant botulism at presentation.
Table 237.2
Conditions Considered in Differential Diagnosis of
Foodborne Botulism and Wound Botulism
Acute gastroenteritis
Myasthenia gravis
Guillain-Barré syndrome
Organophosphate poisoning
Meningitis
Encephalitis
Psychiatric illness
Cerebrovascular accident
Poliomyelitis
Hypothyroidism
Aminoglycoside-associated paralysis
Tick paralysis
Hypocalcemia
Hypermagnesemia
Carbon monoxide poisoning
Hyperemesis gravidarum
Laryngeal trauma
Diabetic complications
Inflammatory myopathy
Overexertion
Treatment
Human botulism immune globulin, given intravenously (BIG-IV, also referred to
as BabyBIG), is licensed for the treatment of infant botulism caused by type A or
B botulinum toxin. Treatment with BIG-IV consists of a single intravenous
infusion of 50-100 mg/kg (see package insert) that should be given as soon as
possible after infant botulism is suspected so as to immediately end the toxemia
that is the cause of the illness and arrest progression of paralysis. When the
Table 237.3
Complications of Infant Botulism
Acute respiratory distress syndrome
Aspiration
Clostridium difficile enterocolitis
Hypotension
Inappropriate antidiuretic hormone secretion
Long bone fractures
Misplaced or plugged endotracheal tube
Nosocomial anemia
Otitis media
Pneumonia
Pneumothorax
Recurrent atelectasis
Seizures secondary to hyponatremia
Sepsis
Subglottic stenosis
Tracheal granuloma
Tracheitis
Transfusion reaction
Urinary tract infection
Prognosis
When the regenerating nerve endings have induced formation of a new motor
end plate, neuromuscular transmission is restored. In the absence of
complications, particularly those related to hypoxia, the prognosis in infant
botulism is for full and complete recovery. Hospital stay in untreated infant
botulism averages 5.7 wk but differs significantly by toxin type, with patients
with untreated type B disease being hospitalized a mean of 4.2 wk and those
with untreated type A disease being hospitalized a mean of 6.7 wk.
In the United States, the case fatality ratio for hospitalized cases of infant
botulism is <1%. After recovery, patients with untreated infant botulism appear
to have an increased incidence of strabismus that requires timely screening and
treatment.
The case fatality ratio in foodborne and wound botulism varies by age, with
younger patients having the best prognosis. Some adults with botulism have
reported chronic weakness and fatigue for >1 yr as sequelae.
Prevention
associated with deposition of immune complexes and activation of complement,
are reported rarely after tetanus vaccination. Mass immunization campaigns in
developing countries have occasionally provoked a widespread hysterical
reaction.
Wound Management
Tetanus prevention measures after trauma consist of inducing active immunity to
tetanus toxin and of passively providing antitoxic antibody (Table 238.1 ).
Tetanus prophylaxis is an essential part of all wound management, but specific
measures depend on the nature of the injury and the immunization status of the
patient. Prevention of tetanus must be included in planning for the consequences
of bombings, natural disasters, and other possible civilian mass-casualty events.
Table 238.1
Tetanus Vaccination and Immune Globulin Use in Wound
Management
§ More frequent boosters are not needed and can accentuate adverse events.
DT, Diphtheria and tetanus toxoid vaccine; DTaP, combined diphtheria toxoid–tetanus toxoid–
acellular pertussis vaccine; Td, tetanus toxoid and reduced diphtheria toxoid vaccine; Tdap,
tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine; TIG, tetanus immune
globulin.
Data from Tetanus (lockjaw). In Kimberlin DW, Brady MT, Jackson MA, Long SS, editors: Red
book: 2015 report of the Committee on Infectious Diseases, ed 30, Elk Grove Village, IL, 2015,
American Academy of Pediatrics.
eliminating certain high carrier populations from testing (e.g., children under 1
yr of age) will increase the positive predictive value of laboratory testing.
Culture for organism isolation is a sensitive test but is labor intensive, taking
several days. Culture alone is not specific because it does not differentiate
between toxin-producing and non–toxin-producing strains.
Pseudomembranous nodules and characteristic plaques may be seen on
colonoscopy or sigmoidoscopy.
Treatment
Initial treatment of CDI involves discontinuation of any nonvital antibiotic
therapy and administration of fluid and electrolyte replacement. For mild cases,
this treatment may be curative. Persistent symptoms or moderate to severe
disease warrant antimicrobial therapy directed against C. difficile.
Oral metronidazole remains the first-line therapy for mild to moderate CDI in
children (Table 239.1 ). For more severe infection, oral vancomycin is approved
by the U.S. Food and Drug Administration (FDA) for CDI. Vancomycin exhibits
ideal pharmacologic properties for treatment of this enteric pathogen, since it is
not absorbed in the gut. Vancomycin is suggested as a first-line agent for severe
disease, as manifested by hypotension, peripheral leukocytosis, or severe
pseudomembranous colitis. Concerns about cost and the emergence of
vancomycin-resistant enterococci limit its use as first-line therapy in mild to
moderate disease. Fidaxomicin , a second-line agent not yet approved for
pediatric use, is a narrow-spectrum macrolide antibiotic with noninferior
efficacy to vancomycin but superior recurrence prevention. The cost of a course
of fidaxomicin can be twice that of vancomycin and 125-fold higher than
metronidazole. Reports have demonstrated high treatment efficacy for donor
(unaffected) fecal therapy (transplant).
Table 239.1
Recommendations for the Treatment of Clostridium difficile
Infection in Children
STRENGTH OF
CLINICAL RECOMMENDED PEDIATRIC MAXIMUM
RECOMMENDATION/QUALITY
DEFINITION TREATMENT DOSE DOSE
OF EVIDENCE
Initial episode, Metronidazole × 7.5 mg/kg/dose 500 mg tid or Weak/Low
nonsevere 10 days PO tid or qid qid
or
Vancomycin × 10 10 mg/kg/dose 125 mg qid Weak/Low
days PO qid
Initial episode, Vancomycin × 10 mg/kg/dose 500 mg qid Strong/Moderate
severe/fulminant 10 days PO or qid
PR
with or without
Metronidazole × 10 10 mg/kg/dose 500 mg tid Weak/Low
days IV* tid
First recurrence, Metronidazole × 7.5 mg/kg/dose 500 mg tid or Weak/Low
nonsevere 10 days PO tid or qid qid
or
Vancomycin × 10 10 mg/kg/dose 125 mg qid Weak/Low
days PO qid
Second or Vancomycin in a 10 mg/kg/dose 125 mg qid Weak/Low
subsequent tapered and qid
recurrence pulsed regimen †
or
Vancomycin × Vancomycin, 10 Vancomycin, Weak/Low
10 days, mg/kg/dose qid; 500 mg qid;
followed by rifaximin: no rifaximin, 400
rifaximin ‡ × 20 pediatric dosing mg tid
days
or
Fecal microbiota Weak/Very low
transplantation
* In cases of severe or fulminant Clostridium difficile infection associated with critical illness,
consider addition of intravenous metronidazole to oral vancomycin.
† Tapered and pulsed regimen: vancomycin, 10 mg/kg with max of 125 mg 4 times daily for 10-14
days, then 10 mg/kg with max of 125 mg twice daily for 1 wk, then 10 mg/kg with max of 125 mg
once daily for 1 wk, and then 10 mg/kg with max of 125 mg every 2 or 3 days for 2-8 wk.
‡ No pediatric dosing for rifaximin; not approved by the U.S. Food and Drug Administration for use
in children <12 yr old.
IV, Intravenously; PO, orally; PR, rectally; tid, 3 times daily; qid, 4 times daily.
Adapted from McDonald LC, Gerding DN, Johnson S, et al: Clinical practice guidelines for
Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases
Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA), Clin
Infect Dis 66(7):e1–e48, 2018 (Table 2).
Table 239.2
Recommendations for the Treatment of Clostridium difficile
Infection in Adults
STRENGTH OF
CLINICAL SUPPORTIVE RECOMMENDED
RECOMMENDATION/QUALITY
DEFINITION CLINICAL DATA TREATMENT*
OF EVIDENCE
Initial episode, Leukocytosis with VAN, 125 mg qid × 10 days Strong/High
nonsevere white blood cell or
count of ≤15,000 FDX, 200 mg bid × 10 days Strong/High
cells/mL and serum Alternate if above agents are Weak/High
creatinine level unavailable: metronidazole, 500 mg
<1.5 mg/dL tid PO × 10 days
Initial episode, Leukocytosis with VAN, 125 mg qid PO × 10 days Strong/High
severe † a white blood cell or
count of ≥15,000 FDX, 200 mg bid × 10 days Strong/High
cells/mL or a serum
creatinine level
>1.5 mg/dL
Initial episode, Hypotension or VAN, 500 mg qid PO or by Strong/Moderate (oral VAN)
fulminant shock, ileus, nasogastric tube. If ileus, consider Weak/Low (rectal VAN)
megacolon adding rectal instillation of VAN. Strong/Moderate (intravenous
Intravenous metronidazole (500 mg metronidazole)
every 8 hr) should be administered
with oral or rectal VAN, particularly
if ileus is present.
First VAN, 125 mg qid × 10 days, if Weak/Low
recurrence metronidazole was used for the
initial episode
or
Use prolonged tapered and Weak/Low
pulsed VAN regimen if standard
regimen was used for initial
episode (e.g., 125 mg qid for
10-14 days, bid for 1 wk, qd for
1 wk, and then every 2 or 3 days
for 2-8 wk)
or
FDX, 200 mg bid ×for 10 days if Weak/Moderate
VAN was used for the initial episode
Second or VAN in a tapered and pulsed Weak/Low
subsequent regimen
recurrence or
VAN, 125 mg qid PO × 10 days, Weak/Low
followed by rifaximin, 400 mg
tid × 20 days
or
FDX, 200 mg bid × 10 days Weak/Low
or
Fecal microbiota transplantation ‡ Strong/Moderate
* All randomized trials have compared 10-day treatment courses, but some patients (particularly
those treated with metronidazole) may have delayed response to treatment, and clinicians should
consider extending treatment duration to 14 days in those circumstances.
Clinical Manifestations
Anaerobic infections occur in a variety of sites throughout the body (Table 240.1
). Anaerobes often coexist synergistically with aerobes. Infections with
anaerobes are usually polymicrobial, including an aerobic component.
Table 240.1
Infections Associated With Anaerobic Bacteria
ANAEROBIC
SITE AND INFECTION MAJOR RISK FACTORS
BACTERIA*
CENTRAL NERVOUS SYSTEM
Cerebral abscess Cyanotic heart disease Polymicrobial
Cystic fibrosis Prevotella
Penetrating trauma Porphyromonas
Bacteroides
Fusobacterium
Peptostreptococcus
Epidural and subdural Direct extension from contiguous sinusitis, otitis Bacteroides fragilis †
empyemas, meningitis media, mastoiditis, or anatomic defect involving the Fusobacterium
dura Peptostreptococcus
Veillonella
UPPER RESPIRATORY TRACT
Dental abscess Poor periodontal hygiene Peptostreptococcus
Ludwig angina (cellulitis of Drugs producing gingival hypertrophy Fusobacterium
sublingual-submandibular
space)
Necrotizing gingivitis Prevotella
(Vincent stomatitis) melaninogenica
Fusobacterium
Chronic otitis-mastoiditis- Tympanic perforation Prevotella
sinusitis Tympanostomy tubes Bacteroides
Fusobacterium
Peptostreptococcus
Peritonsillar abscess Streptococcal pharyngitis Fusobacterium
Retropharyngeal abscess Penetrating injury Prevotella
Porphyromonas
Lemierre syndrome Preexisting viral or bacterial pharyngitis Fusobacterium
LOWER RESPIRATORY TRACT
Aspiration pneumonia Periodontal disease Polymicrobial
Prevotella
Porphyromonas
Fusobacterium
Peptostreptococcus
Necrotizing pneumonitis Bronchial obstruction P. melaninogenica
Lung abscess Altered gag or consciousness Bacteroides intermedius
Aspirated foreign body Fusobacterium
Sequestered lobe Peptostreptococcus
Vascular anomaly Eubacterium
B. fragilis
Veillonella
Septic pulmonary emboli Fusobacterium
INTRAABDOMINAL
Abscess Appendicitis Polymicrobial
B. fragilis
Bilophila wadsworthia
Peptostreptococcus
Clostridium spp.
Secondary peritonitis Penetrating trauma (especially of the colon) Bacteroides
Clostridium
Peptostreptococcus
Eubacterium
Fusobacterium
FEMALE GENITAL TRACT
Bartholin abscess Vaginosis B. fragilis
Tuboovarian abscess Intrauterine device Bacteroides bivius
Endometritis Peptostreptococcus
Pelvic thrombophlebitis Clostridium
Salpingitis Mobiluncus
Chorioamnionitis Actinomyces
Septic abortion Clostridium
SKIN AND SOFT TISSUE
Cellulitis Decubitus ulcers Varies with site and
contamination with oral or
enteric flora
Perirectal cellulitis Abdominal wounds Clostridium perfringens
(myonecrosis)
Myonecrosis (gas gangrene) Pilonidal sinus Bacteroides
Clostridium
Necrotizing fasciitis and Trauma Fusobacterium
synergistic gangrene Human and animal bites Clostridium tertium
Immunosuppressed or neutropenic patients Clostridium septicum
Varicella Anaerobic streptococci
BLOOD
Bacteremia Intraabdominal infection, abscesses, myonecrosis, B. fragilis
necrotizing fasciitis Clostridium
Peptostreptococcus
Fusobacterium
* Infections may also be from or may involve aerobic bacteria as the sole agent or as part of a
mixed infection; brain abscess may contain microaerophilic streptococci; intraabdominal infections
may contain gram-negative enteric organisms and enterococci; and salpingitis may contain
Neisseria gonorrhoeae and Chlamydia trachomatis .
† Bacteroides fragilis is usually isolated from infections below the diaphragm except for brain
abscesses.
Bacteremia
Anaerobes account for approximately 5% of bloodstream bacterial isolates in
adults, but this rate is lower in children. The most common blood isolates of
caused by Clostridium welchii type C (an organism not usually present in the
human intestine), the organism being transmitted by contaminated pig meat.
Anaerobic osteomyelitis , particularly of fingers and toes, can complicate any
process capable of producing hypoxic necrosis, including diabetes, neuropathies,
vasculopathies, and coagulopathies.
Diagnosis
The diagnosis of anaerobic infection requires a high index of suspicion and the
collection of appropriate and adequate specimens for anaerobic culture (Table
240.2 ). Culture specimens should be obtained in a manner that protects them
from contamination with mucosal bacteria and from exposure to ambient
oxygen. Swab samples from mucosal surfaces, nasal secretions, respiratory
specimens, and stool should not be sent for anaerobic culture because these sites
normally harbor anaerobic flora. Aspirates of infected sites, abscess material,
and biopsy specimens are appropriate for anaerobic culturing. Specimens should
be protected from atmospheric oxygen and transported to the laboratory
immediately. Anaerobic transport medium is used to increase the likelihood of
recovery of obligate anaerobes. Gram staining of abscess fluid from suspected
anaerobic infections is useful because even if the organisms do not grow in
culture, they can be seen on the smear.
Table 240.2
Clues to Presumptive Diagnosis of Anaerobic Infections*
Infection contiguous to or near a mucosal surface colonized with anaerobic bacteria (oropharynx, intestinal-
genitourinary tract)
Putrid odor
Severe tissue necrosis, abscesses, gangrene, or fasciitis
Gas formation in tissues (crepitus on exam or visible on plain radiograph)
Failure to recover organisms using conventional aerobic microbiologic methods, despite the presence of mixed
pleomorphic organisms on smears
Failure of organisms to grow after pretreatment with antibiotics effective against anaerobes
Failure of clinical response to antibiotic therapy poorly effective against anaerobic bacteria (e.g.,
aminoglycosides)
“Sulfur granules” in discharges caused by actinomycosis
Toxin-mediated syndromes: botulism, tetanus, gas gangrene, food poisoning, pseudomembranous colitis
Infections associated with anaerobic bacteria (see Table 240.1 )
Septic thrombophlebitis
Septicemic syndrome with jaundice or intravascular hemolysis
Typical appearance on Gram stain:
Bacteroides spp.—small, delicate, pleomorphic, pale, gram-negative bacilli
Fusobacterium nucleatum —thin, gram-negative bacilli with fusiform shape, pointed ends
Fusobacterium necrophorum —pleomorphic gram-negative bacilli with rounded ends
Peptostreptococcus —chained, gram-positive cocci similar to aerobic cocci
Clostridium perfringens —large, short, fat (boxcar-shaped) gram-positive bacilli
* Suspicion of anaerobic infection is critical before specimens are sampled for culture, to ensure
Treatment
Treatment of anaerobic infections usually requires adequate drainage and
appropriate antimicrobial therapy. Antibiotic therapy varies depending on the
suspected or proven anaerobe involved. Many oral anaerobic bacterial species
are susceptible to penicillins, although some strains may produce a β-lactamase.
Drugs that are active against such strains include metronidazole, penicillins
combined with β-lactamase inhibitors (ampicillin-sulbactam, ticarcillin-
clavulanate, piperacillin-tazobactam), carbapenems (imipenem, meropenem,
doripenem, ertapenem), clindamycin, tigecycline, linezolid, and cefoxitin.
Penicillin and vancomycin are active against the gram-positive anaerobes.
Increasing resistance to antimicrobials has been noted among anaerobes,
particularly with Bacteroides spp. Clindamycin is no longer recommended in the
empirical treatment of abdominal infections due to increasing resistance among
Bacteroides. Aerobes are usually present with the anaerobes, necessitating
broad-spectrum antibiotic combinations for empirical therapy. Specific therapy is
bovis. The pediatric dosage is 10-15 mg/kg/day orally (PO) in a single dose, not
to exceed 300 mg/day. The adult dosage is 5 mg/kg/day PO in a single dose, not
to exceed 300 mg/day. Alternative pediatric dosing is 20-30 mg/kg PO in a
single dose, not to exceed 900 mg/dose, given twice weekly under directly
observed therapy (DOT) , in which patients are observed to ingest each dose of
antituberculosis medication to maximize the likelihood of completing therapy.
The duration of treatment depends on the disease being treated (Table 241.1 ).
INH needs to be taken 1 hr before or 2 hr after meals because food decreases
absorption. It is available in liquid, tablet, intravenous (IV; not approved by the
FDA), and intramuscular (IM) preparations.
Table 241.1
Recommended Usual Treatment Regimens for Drug-
Susceptible Tuberculosis in Infants, Children, and
Adolescents
INFECTION/DISEASE
REGIMEN COMMENTS
CATEGORY
LATENT MYCOBACTERIUM TUBERCULOSIS INFECTION a
Isoniazid susceptible 12 weeks of isoniazid plus rifapentine,
once a week
or
4 mo of rifampin, once a day Continuous daily therapy is required.
or Intermittent therapy even by DOT is
not recommended.
9 mo of isoniazid, once a day If daily therapy is not possible, DOT
twice a week can be used for 9 mo.
Isoniazid resistant 4 mo of rifampin, once a day Continuous daily therapy is required.
Intermittent therapy even by DOT is
not recommended.
Isoniazid-rifampin Consult a tuberculosis specialist. Moxifloxacin or levofloxacin with or
resistant without ethambutol or pyrazinamide.
PULMONARY AND EXTRAPULMONARY INFECTION
Except meningitis b 2 mo of isoniazid, rifampin, pyrazinamide, Some experts recommend a 3-
and ethambutol daily or twice weekly, drug initial regimen (isoniazid,
followed by 4 mo of isoniazid and rifampin c rifampin, and pyrazinamide) if the
by DOT d for drug-susceptible M. tuberculosis risk of drug resistance is low.
DOT is highly desirable.
If hilar adenopathy only and the
risk of drug resistance is low, 6
mo course of isoniazid and
rifampin is sufficient.
Drugs can be given 2 or 3
times/wk under DOT.
9-12 mo of isoniazid and rifampin for drug-
susceptible Mycobacterium bovis
Meningitis 2 mo of isoniazid, rifampin, For patients who may have acquired
pyrazinamide, and an aminoglycoside e or tuberculosis in geographic areas where
ethionamide, once daily, followed by 7-10 resistance to streptomycin is common,
mo of isoniazid and rifampin, once daily kanamycin, amikacin, or capreomycin
or twice weekly (9-12 mo total) for drug- can be used instead of streptomycin.
susceptible M. tuberculosis
At least 12 mo of therapy without
pyrazinamide for drug-susceptible M.
bovis
a
Positive TST or IGRA result, no disease. See text for comments and additional
acceptable/alternative regimens.
b
Duration of therapy may be longer for human immunodeficiency virus (HIV)-infected people, and
additional drugs and dosing intervals may be indicated
c Medications should be administered daily for the 1st 2 wk to 2 mo of treatment and then can be
administered 2-3 times/wk by DOT. (Twice-weekly therapy is not recommended for HIV-infected
people.)
d If initial chest radiograph shows pulmonary cavities, and sputum culture after 2 mo of therapy
remains positive, the continuation phase is extended to 7 mo, for a total treatment duration of 9
mo.
e
Streptomycin, kanamycin, amikacin, or capreomycin.
DOT, Directly observed therapy; IGRA, interferon-γ release assay; TST, tuberculin skin test.
Adapted from American Academy of Pediatrics: Red book: 2018–2021 report of the Committee on
Infectious Diseases, ed 31, Elk Grove Village, IL, 2018, AAP (Table 3.85).
Table 241.2
Rifamycins
The rifamycins (rifampin, rifabutin, rifapentine) are a class of macrolide
antibiotics developed from Streptomyces mediterranei. Rifampin is a synthetic
derivative of rifamycin B, and rifabutin is a derivative of rifamycin S.
Rifapentine is a cyclopentyl derivative. The rifamycins inhibit the DNA-
dependent RNA polymerase of mycobacteria, resulting in decreased RNA
synthesis. These agents are generally bactericidal at treatment doses, but they
may be bacteriostatic at lower doses. Resistance is from a mutation in the DNA-
dependent RNA polymerase gene (rpoB ) that is often induced by previous
incomplete therapy. Cross-resistance between rifampin and rifabutin has been
demonstrated.
Rifampin is active against M. tuberculosis, M. leprae, M. kansasii, and M.
avium complex. Rifampin is an integral drug in standard combination treatment
of active M. tuberculosis disease and can be used as an alternative to INH in the
treatment of latent tuberculosis infection in children who cannot tolerate INH.
Rifabutin has a similar spectrum, with increased activity against M. avium
complex. Rifapentine is undergoing pediatric clinical trials and appears to have
activity similar to the activity of rifampin. The pediatric dosage of rifampin is
10-15 mg/ kg/day PO in a single dose, not to exceed 600 mg/day. The adult
dosage of rifampin is 5-10 mg/kg/day PO in a single dose, not to exceed 600
mg/day. Commonly used rifampin preparations include 150 and 300 mg capsules
and a suspension that is usually formulated at a concentration of 10 mg/mL. The
shelf life of rifampin suspension is short (approximately 4 wk), so it should not
Clofazimine
Clofazimine is a synthetic phendimetrazine tartrate derivative that acts by
binding to the mycobacterial DNA at guanine sites. It has a slow bactericidal
activity against M. leprae. Mechanisms of resistance are not well studied. No
cross-resistance between clofazimine and dapsone or rifampin has been shown.
Clofazimine is indicated as part of a combination therapy for the treatment of
M. leprae. It appears there may be some activity against other mycobacteria such
as M. avium complex, although treatment failures are common. Safety and
efficacy of clofazimine are poorly studied in children. The pediatric dosage is 1
mg/kg/day PO as a single dose, not to exceed 100 mg/day, in combination with
dapsone and rifampin, for 2 yr and then additionally as a single agent for >1 yr.
The adult dosage is 100 mg/day PO. Clofazimine should be taken with food to
increase absorption.
The most common adverse effect is a dosage-related, reversible, pink to tan-
brown discoloration of the skin and conjunctiva. Other adverse effects include a
dry, itchy skin rash, headache, dizziness, abdominal pain, diarrhea, vomiting,
peripheral neuropathy, and elevated hepatic transaminases.
Routine laboratory monitoring includes periodic liver function tests.
Table 241.3
Treatment of Nontuberculous Mycobacteria Infections in
Children
ORGANISM DISEASE INITIAL TREATMENT
SLOWLY GROWING SPECIES
Mycobacterium Lymphadenitis Complete excision of lymph nodes; if excision incomplete or disease recurs,
avium complex clarithromycin or azithromycin plus ethambutol and/or rifampin (or rifabutin).
(MAC); Pulmonary Clarithromycin or azithromycin plus ethambutol with rifampin or rifabutin
Mycobacterium infection (pulmonary resection in some patients who fail to respond to drug therapy).
haemophilum ; For severe disease, an initial course of amikacin or streptomycin often is
Mycobacterium included. Clinical data in adults with mild to moderate disease support that 3-
lentiflavum times-weekly therapy is as effective as daily therapy, with less toxicity. For
patients with advanced or cavitary disease, drugs should be given daily.
Mycobacterium Prosthetic Valve removal, prolonged antimicrobial therapy based on susceptibility
chimaera valve testing.
endocarditis
Disseminated See text.
Mycobacterium Pulmonary Rifampin plus ethambutol with isoniazid daily. If rifampin resistance is
kansasii infection detected, a 3-drug regimen based on drug susceptibility testing should be
used.
Osteomyelitis Surgical debridement and prolonged antimicrobial therapy using rifampin plus
ethambutol with isoniazid.
Mycobacterium Cutaneous None, if minor; rifampin, TMP-SMX, clarithromycin, or doxycycline* for
marinum infection moderate disease; extensive lesions may require surgical debridement.
Susceptibility testing not routinely required.
Mycobacterium Cutaneous and Daily intramuscular streptomycin and oral rifampin for 8 wk; excision to
ulcerans bone remove necrotic tissue, if present; potential response to thermotherapy.
infections
RAPIDLY GROWING SPECIES
Mycobacterium Cutaneous Initial therapy for serious disease is amikacin plus meropenem IV, followed
fortuitum group infection by clarithromycin, doxycycline,* TMP-SMX, or ciprofloxacin PO, on the
basis of in vitro susceptibility testing; may require surgical excision. Up to
50% of isolates are resistant to cefoxitin.
Catheter Catheter removal and amikacin plus meropenem IV; clarithromycin, TMP-
infection SMX, or ciprofloxacin, orally, on the basis of in vitro susceptibility testing.
Mycobacterium Otitis media; There is no reliable antimicrobial regimen because of variability in drug
abscessus cutaneous susceptibility. Clarithromycin plus initial course of amikacin plus cefoxitin or
infection imipenem/meropenem; may require surgical debridement on the basis of in
vitro susceptibility testing (50% are amikacin resistant).
Pulmonary Serious disease, clarithromycin, amikacin, and cefoxitin or
infection (in imipenem/meropenem on the basis of susceptibility testing; most isolates have
cystic fibrosis) very low MIC to tigecycline; may require surgical resection.
Mycobacterium Catheter Catheter removal; debridement, removal of foreign material; valve
chelonae infection, replacement; and tobramycin (initially) plus clarithromycin, meropenem, and
prosthetic linezolid.
valve
endocarditis
Disseminated Tobramycin and meropenem or linezolid (initially) plus clarithromycin.
cutaneous
infection
* Doxycycline can be used for short durations (i.e., ≤21 days) without regard to patient age, but for
longer treatment durations is not recommended for children <8 yr old. Only 50% of isolates of M.
can transmit M. tuberculosis to children, and children with HIV infection are at
increased risk for developing tuberculosis after infection. Specific groups are at
high risk for acquiring TBI and progressing to tuberculosis (Table 242.1 ).
Table 242.1
Groups at High Risk for Acquiring Tuberculosis Infection
and Developing Disease in Countries With Low Incidence
RISK FACTORS FOR TUBERCULOSIS INFECTION
Children exposed to high-risk adults
Foreign-born persons from high-prevalence countries
Homeless persons
Persons who inject drugs
Present and former residents or employees of correctional institutions, homeless shelters, and nursing homes
Healthcare workers caring for high-risk patients (if infection control is not adequate)
RISK FACTORS FOR PROGRESSION OF TUBERCULOSIS INFECTION TO TUBERCULOSIS
DISEASE
Infants and children ≤4 yr old, especially those <2 yr old
Adolescents and young adults
Persons co-infected with human immunodeficiency virus
Persons with skin test conversion in the past 1-2 yr
Persons who are immunocompromised, especially in cases of malignancy and solid-organ transplantation,
immunosuppressive medical treatments including anti–tumor necrosis factor therapies, diabetes mellitus,
chronic renal failure, silicosis, and malnutrition
RISK FACTORS FOR DRUG-RESISTANT TUBERCULOSIS
Personal or contact history of treatment for tuberculosis
Contacts of patients with drug-resistant tuberculosis
Birth or residence in a country with a high rate of drug resistance
Poor response to standard therapy
Positive sputum smears (acid-fast bacilli) or culture ≥2 mo after initiating appropriate therapy
Table 242.2
Tuberculin Skin Test (TST) or Interferon-γ Release Assay
(IGRA): Recommendations for Infants, Children, and
Adolescents*
‡ If the child is well and has no history of exposure, the TST or IGRA should be delayed up to 10
wk after return.
From American Academy of Pediatrics: Red book: 2018 report of the Committee on Infectious
Diseases , ed 30, Elk Grove Village, IL, 2015, AAP, p 831.
Table 242.3
Definitions of Positive Tuberculin Skin Test (TST) Results
in Infants, Children, and Adolescents*
INDURATION ≥5 mm
Children in close contact with known or suspected contagious people with tuberculosis disease
Children suspected to have tuberculosis disease:
• Findings on chest radiograph consistent with active or previously tuberculosis disease
• Clinical evidence of tuberculosis disease †
Children receiving immunosuppressive therapy ‡ or with immunosuppressive conditions, including HIV infection
INDURATION ≥10 mm
Children at increased risk of disseminated tuberculosis disease:
• Children <4 yr old
• Children with other medical conditions, including Hodgkin disease, lymphoma, diabetes mellitus, chronic
renal failure, or malnutrition (see Table 242.2 )
Children with increased exposure to tuberculosis disease:
• Children born in high-prevalence regions of the world
• Children often exposed to adults with HIV infection, homeless, users of illicit drugs, residents of nursing
homes, incarcerated or institutionalized, or migrant farm workers
• Children who travel to high-prevalence regions of the world
INDURATION ≥15 mm
Children ≥4 yr old without any risk factors
* These definitions apply regardless of previous BCG immunization; erythema at TST site does
not indicate a positive test result. Tests should be read at 48-72 hr after placement.
† Evidence by physical examination or laboratory assessment that would include tuberculosis in
Table 242.4
Recommendations for Use of Tuberculin Skin Test (TST)
and Interferon-γ Release Assay (IGRA) in Children
TST preferred, IGRA acceptable
• Children <2 yr of age*
IGRA preferred, TST acceptable
• Children >2 yr of age who have received BCG vaccine
• Children >2 yr of age who are unlikely to return for TST reading
TST and IGRA should be considered when:
The initial and repeat IGRAs are indeterminate or invalid.
The initial test (TST or IGRA) is negative, and:
• Clinical suspicion for TB disease is moderate to high. †
• The child has TB risk factor and is at high risk of progression and poor outcome (especially therapy with
immunomodulating biologic agent, e.g., TNF-α antagonist). †
The initial TST is positive and:
• >2 yr old and history of BCG vaccination.
• Additional evidence needed to increase adherence with therapy.
* Some experts do not use an IGRA for children younger than 2 yr because of a relative lack of
data for this age-group and the high risk of progression to disease.
† Positive result of either test is considered significant in these groups.
TWICE-
DAILY
DOSAGE WEEKLY MAXIMUM
DRUG DOSAGE ADVERSE REACTIONS
FORMS DOSAGE DOSE
(mg/kg)
(mg/kg/dose)
Ethambutol Tablets: 20 50 2.5 g Optic neuritis (usually reversible),
100 mg decreased red-green color
400 mg discrimination, gastrointestinal tract
disturbances, hypersensitivity
Isoniazid* Scored 10-15 † 20-30 Daily: Mild hepatic enzyme elevation,
tablets: 300 mg hepatitis, † peripheral neuritis,
100 mg Twice hypersensitivity
300 mg weekly:
Syrup: 10 900 mg
mg/mL
Pyrazinamide* Scored tablets: 30-40 50 2 g Hepatotoxic effects, hyperuricemia,
500 mg arthralgias, gastrointestinal tract upset
Rifampin* Capsules: 15-20 15-20 600 mg Orange discoloration of
150 mg secretions or urine, staining of
300 mg contact lenses, vomiting,
Syrup hepatitis, influenza-like reaction,
formulated thrombocytopenia, pruritus
from Oral contraceptives may be
capsules ineffective.
* Rifamate is a capsule containing 150 mg of isoniazid and 300 mg of rifampin. Two capsules
provide the usual adult (i.e., person weighing >50 kg) daily doses of each drug. Rifater, in the
United States, is a capsule containing 50 mg of isoniazid, 120 mg of rifampin, and 300 mg of
pyrazinamide. Isoniazid and rifampin also are available for parenteral administration. Many
experts recommend using a daily rifampin dose of 20–30 mg/kg/day for infants and toddlers and
for serious forms of tuberculosis such as meningitis and disseminated disease.
† When isoniazid in a dosage exceeding 10 mg/kg/day is used in combination with rifampin, the
incidence of hepatotoxic effects may be increased.
From American Academy of Pediatrics: Red book: 2018 report of the Committee on Infectious
Diseases , ed 30, Elk Grove Village, IL, 2015, AAP, p 842.
Table 242.6
Less Commonly Used Drugs for Treating Drug-Resistant
Tuberculosis in Infants, Children, and Adolescents*
DOSAGE, MAXIMUM
DRUGS DAILY DOSAGE (mg/kg) ADVERSE REACTIONS
FORMS DOSE
Vials: 500 15-30 (IV or IM 1 g Auditory and vestibular toxic
Amikacin † mg, 1 g administration) effects, nephrotoxic effects
Bedaquiline Tablets: 100 Adults and children ≥12 yr, QTc prolongation, reduced levels
mg >33 kg: 400 mg for 14 with efavirenz co-administration
days, then 200 mg 3 times
weekly for 22 wk
Capreomycin † Vials: 1 g 15-30 (IM administration) 1 g Auditory and vestibular toxicity
and nephrotoxic effects
Clofazimine Gelcaps: 2-3 mg/kg per day 100 mg QTc prolongation, reversible skin
50 mg pigmentation
100 mg
Cycloserine Capsules: 10-20, given in 2 divided 1 g Psychosis, personality changes,
250 mg doses seizures, rash
Delamanid Tablets: Adults and children QTc prolongation, adverse events
50 mg ≥13 yr, ≥35 kg: 100 with hypoalbuminemia, avoid if
100 mg mg twice daily metronidazole allergic
Children 6-12 yr, 20-
34 kg: 50 mg twice
daily
Ethionamide Tablets: 250 15-20, given in 2-3 divided 1 g GI tract disturbances, hepatotoxic
mg doses effects, hypersensitivity reactions,
hypothyroidism
Kanamycin Vials: 15-30 (IM or IV 1 g Auditory and vestibular toxic
75 mg/2 administration) effects, nephrotoxic effects
mL
500
mg/2
mL
1 g/3
mL
Levofloxacin Tablets: Adults: 750-1000 mg 1 g Theoretic effect on growing
250 mg (daily) cartilage, joint pain, GI tract
500 mg Children: 15-20 mg/kg disturbances, rash, headache,
750 mg daily restlessness, confusion
Oral
solution:
25/mL
Vials: 25
mg/mL
Linezolid Tablets: Children ≥12 yr: 10 600 mg Bone marrow suppression,
400 mg mg/kg daily peripheral neuropathy, lactic
600 mg Children <12 yr: 10 acidosis, potential overlapping
Syrup: mg/kg twice daily toxicity with nucleoside reverse
20 transcriptase inhibitors
mg/mL
Ofloxacin Tablets: Adults/adolescents: 800 mg Arthropathy, arthritis
200 mg 800 mg
300 mg Children 15-20 mg/kg
400 mg daily
Vials:
20
mg/mL
40
mg/mL
Moxifloxacin Tablets: Adults/adolescents: 400 mg Arthropathy, arthritis
400 mg 400 mg
IV Children: 7.5-10
solution: mg/kg daily
400
mg/250
mL in
0.8%
saline
Paraaminosalicylic Packets: 3 g 200-300 (2-4 times a day) 10 g GI tract disturbances,
acid (PAS) hypersensitivity, hepatotoxic
effects
Streptomycin † Vials: 20-40 (IM administration) 1 g Auditory and vestibular toxic
1 g effects, nephrotoxic effects, rash
4 g
* These drugs should be used in consultation with a specialist in tuberculosis.
Treatment
The primary goal of treatment is early antimicrobial therapy to prevent
permanent neuropathy. Leprosy is curable. Effective treatment requires
multidrug therapy (MDT) with dapsone , clofazimine , and rifampin .
Combination therapy is employed to prevent antimicrobial resistance. In the
United States, clinical providers considering a diagnosis and treatment of a
patient with HD should obtain consultation from the NHDP. The recommended
combination MDT can be obtained free of charge from the NHDP (Table 243.1 )
and in other countries through WHO (Table 243.2 ). Compared to WHO, the
NHDP advocates for a longer duration of treatment and daily rather than
monthly administration of rifampin, because shorter antimicrobial regimens have
been associated with greater risk of relapse. The recommended duration by the
WHO for tuberculoid disease is 6 mo and for lepromatous disease, 12 mo.
Table 243.1
NHDP-Recommended Multidrug Therapy Regimens for
Hansen Disease in the United States
TYPE OF LEPROSY PATIENT ANTIMICROBIAL DURATION OF
POPULATION THERAPY THERAPY
Multibacillary (LL, BL, Adult Dapsone, 100 mg/day, and 24 mo
BB) Rifampin, 600 mg/day, and
Clofazimine, 50 mg/day
Pediatric* Dapsone, 1 mg/kg/day, and
Rifampin, 10-20 mg/kg/day,
and
Clofazimine, 1 mg/kg/day †
Paucibacillary (TT, BT) Adult Dapsone, 100 mg/day, and 12 mo
Rifampin, 600 mg/day
Pediatric* Dapsone, 1 mg/kg/day, and
Rifampin, 10-20 mg/kg/day
*
Daily pediatric mg/kg dose should not exceed adult daily maximum.
† Clofazimine is only available through NHDP Investigational New Drug (IND) program; minimum
formulation is 50 mg, and capsules should not be cut. Alternative dosing includes clofazimine, 2
mg/kg every other day, or clarithromycin, 7.5 mg/kg/day.
NHDP multidrug therapy is daily and of longer duration than WHO-recommended regimen. All
drugs are administered orally.
NHDP, National Hansen's Disease Program; BB, Borderline; BL, borderline lepromatous; BT,
borderline tuberculoid; LL, lepromatous; TT, tuberculoid.
Table 243.2
WHO-Recommended Multidrug Therapy (MDT) Regimens
for Hansen Disease
rifampicin, 10 mg/kg once monthly; dapsone, 2 mg/kg/day; and clofazimine, 1 mg/kg on alternate
days.
WHO, World Health Organization; BB, Borderline; BL, borderline lepromatous; BT, borderline
tuberculoid; LL, lepromatous; TT, tuberculoid.
Observed differences in pathogenicity, clinical relevance, and spectrum of
clinical disease associated with the various NTM species emphasize the
importance of bacterial factors in the pathogenesis of NTM disease, although
exact virulence factors remain largely unknown.
Clinical Manifestations
Lymphadenitis of the superior anterior cervical or submandibular lymph nodes
is the most common manifestation of NTM infection in children (Table 244.1 ).
Preauricular, posterior cervical, axillary, and inguinal nodes are involved
occasionally. Lymphadenitis is most common in children 1-5 yr of age and has
been related to soil exposure (e.g., playing in sandboxes) and teething, although
exact predisposing conditions have not been found. Given the constant
environmental exposure to NTM, the occurrence of these infections might also
reflect an atypical immune response of a subset of the infected children during or
after their first contact with NTM. However, in healthy children with isolated
NTM lymphadenitis, immunodeficiency is very rare.
Table 244.1
Diagnosis
For infections of lymph nodes, skin, bone, and soft tissues, isolation of the
causative NTM bacteria by Mycobacterium culture, preferably with histologic
confirmation of granulomatous inflammation, normally suffices for diagnosis
(Table 244.2 ). The differential diagnosis of NTM lymphadenitis includes acute
bacterial lymphadenitis, tuberculosis, cat-scratch disease (Bartonella henselae) ,
mononucleosis, toxoplasmosis, brucellosis, tularemia, and malignancies,
especially lymphomas. Differentiation between NTM and M. tuberculosis may
be difficult, but children with NTM lymphadenitis usually have a Mantoux
tuberculin skin test reaction of <15 mm induration, unilateral anterior cervical
node involvement, a normal chest radiograph, and no history of exposure to
adult tuberculosis. Definitive diagnosis requires excision of the involved nodes
for culture and histology. Fine-needle aspiration for PCR and culture can enable
earlier diagnosis, before excisional biopsy.
Table 244.2
American Thoracic Society Diagnostic Criteria for
Nontuberculous Mycobacteria (NTM) Lung Disease
The minimum evaluation of a patient for NTM lung disease should include:
1. Chest radiograph or, when no cavitation is present, HRCT
2. At least 3 sputum or respiratory samples for AFB culture
3. Exclusion of other disease, such as tuberculosis
Clinical diagnosis of NTM is based on pulmonary symptoms, presence of nodules or cavities, as seen on chest
radiograph or an HRCT scan, with multifocal bronchiectasis with multiple small nodules, and exclusion of other
diagnoses.
Microbiologic diagnosis of NTM:
At least 2 expectorated sputa (or at least 1 bronchial wash or lavage) with positive cultures for NTM, or
transbronchial or other lung biopsy showing the presence of granulomatous inflammation or AFB, with 1 or
more sputum or bronchial washings that are culture positive for NTM.
AFB, Acid-fast bacilli; HRCT, high-resolution computed tomography.
Data from Griffith DE, Aksamit T, Brown-Elliott BA, et al: An official ATS/IDSA statement:
diagnosis, treatment and prevention of nontuberculous mycobacterial diseases, Am J Respir Crit
Care Med 175:367–416, 2007.
From Brown-Elliott BA, Wallace Jr. RJ: Infections caused by nontuberculous mycobacteria other
than Mycobacterium avium complex. In Bennett JF, Dolin R, Blaser MJ, editors: Mandell, Douglas,
and Bennett's principles and practice of infectious diseases, ed 8, Philadelphia, 2015, Elsevier
(Table 254-3).
Table 245.1
FIG. 245.6 Hutchinson teeth as a late manifestation of congenital syphilis.
Table 245.2
Table 245.3
is normal, and the CSF Venereal Disease Research Laboratory (VDRL) test result is negative,
some experts would treat with up to 3 weekly doses of penicillin G benzathine, 50,000 U/kg, IM.
Some experts also suggest giving these patients a single dose of penicillin G benzathine, 50,000
U/kg, IM, after the 10-day course of intravenous aqueous penicillin.
†
Early latent syphilis is defined as being acquired within the preceding year.
‡ Penicillin G benzathine and penicillin G procaine are approved for intramuscular administration
only.
§ Late latent syphilis is defined as syphilis beyond 1 yr duration.
¶ Some experts administer penicillin G benzathine, 2.4 million U, IM, once per week for up to 3 wk
after completion of these neurosyphilis treatment regimens.
IV , intravenously; IM , intramuscularly.
From American Academy of Pediatrics: Red book: 2015 report of the committee on infectious
diseases , ed 30, Elk Grove Village, IL, 2015, American Academy of Pediatrics, Table 3.74.
Acquired Syphilis
Primary, secondary, and early latent disease is treated with a single dose of
benzathine penicillin G (50,000 units/kg IM, maximum 2.4 million units).
Persons with late latent or tertiary disease require 3 doses at 1 wk intervals.
Nonpregnant penicillin-allergic patients without neurosyphilis may be treated
with either doxycycline (100 mg PO twice daily for 2 wk) or tetracycline (500
have an immunogenetic basis. Persons with certain HLA-DR allotypes may be
genetically predisposed to develop chronic Lyme arthritis. An autoinflammatory
response in the synovium can result in clinical symptoms long after the bacteria
have been killed by antibiotics.
Clinical Manifestations
The clinical manifestations of Lyme disease are divided into early and late stages
(Table 249.1 ). Early Lyme disease is further classified as early localized or early
disseminated disease. Untreated patients can progressively develop clinical
symptoms of each stage of the disease, or they can present with early
disseminated or with late disease without apparently having had any symptoms
of the earlier stages of Lyme disease.
Table 249.1
Treatment
Table 249.2 provides treatment recommendations. Most patients can be treated
with an oral regimen of antibiotic therapy. Young children are generally treated
with amoxicillin. Doxycycline has the advantages of good central nervous
system penetration and activity against A. phagocytophilum , which may be
transmitted at the same time as B. burgdorferi in certain geographic areas. In
general, children younger than 8 yr of age should not be treated with
doxycycline because of the risk of permanent staining of the teeth (although
courses of ≤2 wk are usually safe in this regard). Patients who are treated with
doxycycline should be alerted to the risk for developing photosensitivity in sun-
exposed areas while taking the medication; long sleeves, long pants, and hat are
recommended for activities in direct sunlight. The only oral cephalosporin
proved to be effective for the treatment of Lyme disease is cefuroxime axetil,
which is an alternative for persons who cannot take doxycycline or who are
allergic to penicillin. Macrolide antibiotics, including azithromycin, appear to
have limited activity.
Table 249.2
Recommended Treatment of Lyme Disease
Clinical Manifestations
The main syndromes associated with Ureaplasma spp., M. genitalium, and M.
hominis are displayed in Table 251.1 .
Table 251.1
Clinical Syndromes and Antibiotic Therapy for
Ureaplasmas and Mycoplasmas Infection
† CNS disease includes: meningitis, brain abscess, subdural empyema, and nonfunctioning CNS
shunts.
‡ The most commonly used quinolones are ciprofloxacin and moxifloxacin.
Bronchopulmonary Dysplasia
The role of these organisms in causing severe respiratory insufficiency, the need
for mechanical ventilation, the development of BPD, or death remains
controversial. Nevertheless, meta-analyses of published studies have identified
respiratory colonization with Ureaplasma spp. as an independent risk factor for
in humans where Amblyomma ticks frequently contain R. amblyommatis suggest
that the full range of agents that can cause spotted fever is still to be discerned.
Table 255.1
Summary of Rickettsial Diseases of Humans, Including
Rickettsia, Orientia, Ehrlichia, Anaplasma, Neorickettsia,
and Coxiella
PRESENTING
ARTHROPOD VECTOR, GEOGRAPHIC
GROUP OR DISEASE AGENT CLINICAL
TRANSMISSION HOSTS DISTRIBUTION
FEATURES*
SPOTTED FEVER GROUP
Rocky Mountain Rickettsia Tick bite: Dogs Western Fever, headache,
spotted fever rickettsii Dermacentor Rodents hemisphere rash,* emesis,
species (wood diarrhea, myalgias
tick, dog tick)
Rhipicephalus
sanguineus
(brown dog
tick)
Mediterranean Rickettsia Tick bite: R. Dogs Africa, Painless eschar
spotted fever conorii sanguineus (brown Rodents Mediterranean, (tache noir) with
(boutonneuse dog tick) India, Middle East regional
fever) lymphadenopathy,
fever, headache,
rash,* myalgias
African tick-bite Rickettsia Tick bite Cattle Sub-Saharan Fever, single or
fever africae Goats? Africa, Caribbean multiple eschars,
regional
lymphadenopathy,
rash* (can be
vesicular)
Tickborne Rickettsia Tick bite: ? Europe Eschar (scalp),
lymphadenopathy slovaca, Dermacentor painful
(TIBOLA); Rickettsia lymphadenopathy
Dermacentor- raoultii,
borne necrosis Rickettsia
and sibirica
lymphadenopathy mongolotimonae
(DEBONEL)
Rickettsia sp , “Rickettsia Dermacentor California Eschar, fever,
364D genotype philippi ” occidentalis headache,
(Pacific coast tick) lymphadenopathy,
malaise
Flea-borne Rickettsia felis Flea bite Opossums Western Fever, rash,*
spotted fever Cats hemisphere, headache
Dogs Europe
TRANSITIONAL GROUP
Rickettsialpox Rickettsia akari Mite bite Mice North America, Painless eschar,
Russia, Ukraine, ulcer or papule;
Adriatic, Korea, tender regional
South Africa lymphadenopathy,
fever, headache,
rash* (can be
vesicular)
Queensland tick Rickettsia Ixodes holocyclus, Bandicoots Australia, Fever, eschar,
typhus australis I. tasmani and Rodents Tasmania headache, myalgia,
lymphadenopathy
TYPHUS GROUP
Murine typhus Rickettsia typhi Flea feces Rats Worldwide Fever, headache,
Opossums rash,* myalgias,
emesis,
lymphadenopathy,
hepatosplenomegaly
Epidemic (louse- Rickettsia Louse feces Humans South America, Fever, headache,
borne) typhus prowazekii Central America, abdominal pain,
(recrudescent Mexico, Africa, rash,* CNS
form: Brill- Asia, Eastern involvement
Zinsser disease) Europe
Flying squirrel Rickettsia Louse feces? Flying Eastern United Same as above
(sylvatic) typhus prowazekii Flea feces or squirrels States (often milder)
bite?
SCRUB TYPHUS
Scrub typhus Orientia Chigger bite: Rodents? South Asia, Fever, rash,*
tsutsugamushi Leptotrombidium Japan, Indonesia, headache, painless
Korea, China, eschar,
Russia, Australia hepatosplenomegaly,
gastrointestinal
symptoms
ALT, alanine aminotransferase; ANC, absolute neutrophil count; AST, aspartate aminotransferase;
CNS, central nervous system; DFA, direct fluorescent antibody; IFA, indirect fluorescent antibody;
IgG, immunoglobulin G; IgM, immunoglobulin M; IH, immunohistochemistry; PCR, polymerase
chain reaction; WBC, white blood cell count.
Infections with other members of the spotted fever and transitional groups are
clinically similar to MSF, with fever, maculopapular rash, and eschar at the site
of the tick bite. Israeli spotted fever is generally associated with a more severe
course, including death, in children. African tick bite fever is relatively mild, can
include a vesicular rash, and often manifests with multiple eschars. New
potentially pathogenic rickettsial species have been identified, including R.
slovaca, the cause of tickborne lymphadenopathy or Dermacentor -borne
necrosis and lymphadenopathy. R. aeschlimannii, R. heilongjiangensis, R.
helvetica, R. massiliae, and R. raoultii are all reported to cause mild to moderate
illnesses in humans, although few cases have been described. Fortunately, the
vast majority of infections respond well to doxycycline treatment if instituted
early in illness; however, this is a significant challenge.
255.1
Rocky Mountain Spotted Fever
(Rickettsia rickettsii)
CHAPTER 260
Invasive fungal infections are a major cause of morbidity and mortality in the
growing number of immunocompromised children. Fortunately, the therapeutic
armamentarium for invasive fungal infections has markedly increased since the
turn of the century (Tables 260.1 and 260.2 ).
Table 260.1
Suggested Dosing of Antifungal Agents in Children and
Neonates
SUGGESTED
DRUG FORMULATIONS PEDIATRIC COMMENTS
DOSAGE
Amphotericin IV 1 mg/kg/day Generally less toxicity in children than adults; do not
B start with smaller test doses
deoxycholate
Lipid IV 5 mg/kg/day Generally, all lipid formulations are dosed the same;
amphotericin there is no clear indication of one formulation over
B another for clinical efficacy
formulations
Fluconazole IV, PO 12 mg/kg/day Loading dose (25 mg/kg) is recommended in neonates
based on pharmacokinetic simulations and likely
suggested in children, but insufficiently studied
Itraconazole PO 2.5 mg/kg/dose Divide dosage twice daily in children; follow trough
bid levels
Voriconazole IV, PO 8 mg/kg/dose Linear pharmacokinetics in children requires higher
bid IV dosing than in adults; 9 mg/kg/dose bid IV loading,
maintenance; 9 followed by maintenance dosing; follow trough levels
mg/kg/dose bid
oral maintenance
Posaconazole IV, PO Suspected to be Dosage unclear in children at present.
12-24 mg/kg/day In adults, max dosage for oral suspension is 800
divided tid (oral mg/day, and optimally divide this into 2 or 3 doses;
suspension) follow trough levels. Adult dosing for IV and
extended-release tablet is 300 mg twice on 1st day,
then 300 mg once daily.
Isavuconazole PO, IV No dosing in Adult dosing for IV and tablet is 200 mg 3 times on 1st
children day, then 200 mg once daily.
Micafungin IV 2-10 mg/kg/day Highest dosages in neonates (10 mg/kg/day), and lower
dosages in children; >8 yr of age, use adult dosage
Anidulafungin IV 1.5 mg/kg/day Loading dose of 3 mg/kg/day
Caspofungin IV 50 mg/m2 /day Load with 70 mg/m2 /day, then 50/mg/m2 /day as
maintenance dosage
Table 260.2
Suggested Antifungals for Specific More Common Fungal
Pathogens
AMPHOTERICIN
FUNGAL
B FLUCONAZOLE ITRACONAZOLE VORICONAZOLE POSACONAZOLE
SPECIES
FORMULATIONS
Aspergillus ++ − − − −
calidoustus
Aspergillus + − +/− ++ +
fumigatus
Aspergillus − − + ++ +
terreus
Blastomyces ++ + ++ + +
dermatitidis
Candida + ++ + + +
albicans
Candida + − +/− +/− +/−
glabrata
Candida + − − + +
krusei
Candida − ++ + + +
lusitaniae
Candida ++ ++ + + +
parapsilosis
Coccidioides ++ + ++ + ++
immitis
Cryptococcus ++ + + + +
spp.
Fusarium +/− − − ++ +
spp.
Histoplasma ++ + ++ + +
capsulatum
Mucor spp. ++ − +/− − +
Scedosporium − − +/− + +
apiospermum
Scedosporium − − +/− +/− +/−
prolificans
++ , preferred therapy(ies); +, usually active; +/−, variably active; −, usually not active.
C. krusei and some isolates of C. glabrata. Additionally, in centers where
fluconazole prophylaxis is used, another agent, such as amphotericin B
deoxycholate, should be used for treatment. The echinocandins have excellent
activity against most Candida species and have been used successfully in
patients with resistant organisms or in whom other therapies have failed. Several
studies have described the pharmacokinetics of antifungals in infants (Table
261.1 ).
Table 261.1
Prognosis
Mortality following invasive candidiasis in premature infants has been
consistently reported to be around 20% in large studies but can be as high as
50% in infants <1,500 g birthweight. Candidiasis is also associated with poor
neurodevelopmental outcomes, chronic lung disease, and severe retinopathy of
prematurity.
Bibliography
readily on routine blood culture media, with ≥90% of positive cultures identified
within 72 hr. CT may demonstrate findings consistent with invasive fungal
infection but also is limited by nonspecific findings and false negatives. The role
of screening by CT scan has not been well defined. In high-risk patients, serial
serum assays for (1,3)-β-D -glucan, a polysaccharide component of the fungal
cell wall, may contribute to the diagnosis of invasive Candida infection.
However, this test is not sensitive or specific enough to be used without a careful
assessment of the limitations of the assay.
Treatment
Echinocandins are favored as empirical therapy for moderately or severely ill
children and for those with neutropenia; fluconazole is acceptable for those who
are infected with a susceptible organism and are less critically ill; amphotericin
B products are also acceptable. Definitive antifungal selection should be made
based on susceptibility testing results. Fluconazole is not effective against C.
krusei and some isolates of C. glabrata. C. parapsilosis has occasional resistance
to the echinocandins, but the overall rate is still low. Amphotericin B
deoxycholate is inactive against approximately 20% of the strains of C.
lusitaniae, and therefore susceptibility testing should be performed for all strains
(Table 261.2 ). C. auris , a species first identified in 2009 that has caused
nosocomial infections worldwide, is resistant to most antifungals. An
echinocandin should be used until sensitivity results are available.
Table 261.2
Table 267.1
PRIMARY INFECTIONS
Severe, prolonged (≥6 wk), or progressive infection
RISK FACTORS FOR EXTRAPULMONARY DISSEMINATION
Primary or acquired cellular immune dysfunction (including patients receiving tumor necrosis factor inhibitors)
Neonates, infants, the elderly
Male sex (adult)
Filipino, African, Native American, or Latin American ethnicity
Late-stage pregnancy and early postpartum period
Standardized complement fixation antibody titer >1 : 16 or increasing titer with persisting symptoms
Blood group B
Human leukocyte antigen class II allele-DRBI*1301
Table 267.2
Patients should be followed closely because late relapse can occur, especially
in patients who are immunosuppressed or have severe manifestations. Treatment
is recommended for all HIV-infected patients with active coccidioidomycosis
and CD4 counts <250/µL. Following successful treatment, antifungals may be
stopped if the CD4 count exceeds 250/µL. Treatment should be continued if the
CD4 count remains less than 250/µL and should be given indefinitely in all HIV-
infected patients with coccidioidal meningitis.
First-line agents include oral and intravenous preparations of fluconazole (12
mg/kg/day IV or PO) and itraconazole (10 mg/kg/day). Serum levels of
itraconazole should be monitored.
Amphotericin B is preferred for initial treatment of severe infections.
Amphotericin B deoxycholate is less costly than lipid formulations and is often
well tolerated in children. Once a daily dose of amphotericin B deoxycholate of
CHAPTER 272
Table 272.1
Currently Licensed Antiviral Drugs
Table 272.2
Antiviral Therapies for Non-HIV Clinical Conditions*
ANTIVIRAL
VIRUS CLINICAL SYNDROME AGENT OF ALTERNATIVE ANTIVIRAL AGENTS
CHOICE
Influenza A Treatment Oseltamivir Zanamivir (>7 yr old)
and B (>2 wk old) Peramivir (>2 yr old)
Prophylaxis Oseltamivir Zanamivir (>5 yr old)
(>3 mo old)
Respiratory Bronchiolitis or pneumonia in Ribavirin
syncytial high-risk host aerosol
virus
Adenovirus In immunocompromised Cidofovir
patients:
Pneumonia
Viremia
Nephritis
Hemorrhagic cystitis
CMV Congenital CMV infection Ganciclovir Valganciclovir (if oral therapy appropriate; long-
(IV) term oral valganciclovir investigational but may
improve developmental and hearing outcomes)
Retinitis in AIDS patients Valganciclovir Ganciclovir
Cidofovir
Foscarnet
Ganciclovir ocular insert
Pneumonitis, colitis; Ganciclovir Foscarnet
esophagitis in (IV) Cidofovir
immunocompromised patients Valganciclovir
HSV Neonatal herpes Acyclovir
(IV)
Suppressive therapy following Acyclovir
neonatal herpes with central (PO)
nervous system involvement
HSV encephalitis Acyclovir
(IV)
HSV gingivostomatitis Acyclovir Acyclovir (IV)
(PO)
First episode genital infection Acyclovir Valacyclovir
(PO) Famciclovir
Acyclovir (IV) (severe disease)
Recurrent genital herpes Acyclovir Valacyclovir
(PO) Famciclovir
Suppression of genital herpes Acyclovir Valacyclovir
(PO) Famciclovir
Cutaneous HSV (whitlow, Acyclovir Penciclovir (topical)
herpes gladiatorum) (PO)
Eczema herpeticum Acyclovir Acyclovir (IV) (severe disease)
(PO)
Mucocutaneous infection in Acyclovir Acyclovir (PO) (if outpatient therapy acceptable)
immunocompromised host (IV)
(mild)
Mucocutaneous infection in Acyclovir
immunocompromised host (IV)
(moderate to severe)
Prophylaxis in bone marrow Acyclovir Valacyclovir
transplant recipients (IV)
Acyclovir-resistant HSV Foscarnet Cidofovir
Keratitis or keratoconjunctivitis Trifluridine Vidarabine
Varicella- Chickenpox, healthy child Supportive Acyclovir (PO)
zoster virus care
Chickenpox, Acyclovir
immunocompromised child (IV)
Zoster (not ophthalmic branch Supportive Acyclovir (PO)
of trigeminal nerve), healthy care
child
Zoster (ophthalmic branch of Acyclovir
trigeminal nerve), healthy child (IV)
Zoster, immunocompromised Acyclovir Valacyclovir
child (IV)
*
For antiviral agents for hepatitis B and hepatitis C, see Table 272.1 .
CMV, cytomegalovirus; HSV, herpes simplex virus.
Complications
Complications of measles are largely attributable to the pathogenic effects of the
virus on the respiratory tract and immune system (Table 273.1 , Fig. 273.2 ).
Several factors make complications more likely. Morbidity and mortality from
measles are greatest in individuals younger than 5 yr of age (especially <1 yr of
age) and older than 20 yr of age. In developing countries, higher case fatality
rates have been associated with crowding, possibly attributable to larger
inoculum doses after household exposure. Severe malnutrition in children results
in a suboptimal immune response and higher morbidity and mortality with
measles infection. Low serum retinol levels in children with measles are
associated with higher measles morbidity and mortality in developing countries
and in the United States. Measles infection lowers serum retinol concentrations,
so subclinical cases of hyporetinolemia may be made symptomatic during
measles. Measles infection in immunocompromised persons is associated with
increased morbidity and mortality. Among patients with malignancy in whom
measles develops, pneumonitis occurs in 58% and encephalitis occurs in 20%.
Table 273.1
Complications by Age for Reported Measles Cases, United
States, 1987–2000
Vaccine
Vaccination against measles is the most effective and safe prevention strategy.
Measles vaccine in the United States is available as a combined vaccine with
measles-mumps-rubella vaccine, the last of which is the recommended form in
most circumstances (Table 273.2 ). Following the measles resurgence of 1989-
1991, a 2nd dose of measles vaccine was added to the schedule. The current
recommendations include a 1st dose at 12-15 mo of age, followed by a 2nd dose
at 4-6 yr of age. However, the 2nd dose can be given any time after 30 days
following the 1st dose, and the current schedule is a convenience schedule.
Seroconversion is slightly lower in children who receive the 1st dose before or at
12 mo of age (87% at 9 mo, 95% at 12 mo, and 98% at 15 mo) because of
persisting maternal antibody; however, this is an evolving situation, with
children currently as young as 6 mo unprotected from maternal antibodies and
susceptible to measles infection. For children who have not received 2 doses by
11-12 yr of age, a 2nd dose should be provided. Infants who receive a dose
before 12 mo of age should be given 2 additional doses at 12-15 mo and 4-6 yr
of age. Children who are traveling should be offered either primary measles
immunization even as young as 6 mo or a 2nd dose even if <4 yr.
Table 273.2
Vaccine
Vaccination against measles is the most effective and safe prevention strategy.
Measles vaccine in the United States is available as a combined vaccine with
measles-mumps-rubella vaccine, the last of which is the recommended form in
most circumstances (Table 273.2 ). Following the measles resurgence of 1989-
1991, a 2nd dose of measles vaccine was added to the schedule. The current
recommendations include a 1st dose at 12-15 mo of age, followed by a 2nd dose
at 4-6 yr of age. However, the 2nd dose can be given any time after 30 days
following the 1st dose, and the current schedule is a convenience schedule.
Seroconversion is slightly lower in children who receive the 1st dose before or at
12 mo of age (87% at 9 mo, 95% at 12 mo, and 98% at 15 mo) because of
persisting maternal antibody; however, this is an evolving situation, with
children currently as young as 6 mo unprotected from maternal antibodies and
susceptible to measles infection. For children who have not received 2 doses by
11-12 yr of age, a 2nd dose should be provided. Infants who receive a dose
before 12 mo of age should be given 2 additional doses at 12-15 mo and 4-6 yr
of age. Children who are traveling should be offered either primary measles
immunization even as young as 6 mo or a 2nd dose even if <4 yr.
Table 273.2
Table 273.3
Suggested Intervals Between Immunoglobulin
Administration and Measles Immunization*
DOSE
INDICATION FOR IMMUNOGLOBULIN Units (U) or Interval
Route mg IgG/kg
Milliliters (mL) (mo) †
Tetanus (as tetanus Ig) IM 250 U 10 3
Hepatitis A prophylaxis (as Ig):
Contact prophylaxis IM 0.02 mL/kg 3.3 3
International travel IM 0.06 mL/kg 10 3
Hepatitis B prophylaxis (as hepatitis B Ig) IM 0.06 mL/kg 10 3
Rabies prophylaxis (as rabies Ig) IM 20 IU/kg 22 4
Varicella prophylaxis (as VariZIG) IM 125 U/10 kg 20-40 5
(maximum 625 U)
Measles prophylaxis (as Ig):
Standard IM 0.50 mL/kg 80 6
Immunocompromised host IV 400 mg/kg 8
Respiratory syncytial virus prophylaxis (palivizumab IM — 15 mg/kg None
monoclonal antibody) ‡ (monoclonal)
Cytomegalovirus immune globulin IV 3 mL/kg 150 6
Blood transfusion:
Washed RBCs IV 10 mL/kg Negligible 0
RBCs, adenine-saline added IV 10 mL/kg 10 3
Packed RBCs IV 10 mL/kg 20-60 6
Whole blood IV 10 mL/kg 80-100 6
Plasma or platelet products IV 10 mL/kg 160 7
Replacement (or therapy) of immune deficiencies (as IV — 300-400 8
IVIG)
ITP (as IVIG) IV — 400 8
ITP IV — 1,000 10
ITP or Kawasaki disease IV — 1,600-2,000 11
* Immunization in the form of measles-mumps-rubella (MMR), measles-mumps-rubella-varicella
(MMRV), or monovalent measles vaccine.
† These intervals should provide sufficient time for decreases in passive antibodies in all children
vigilance and maintenance of high levels of immunity in the United States are
necessary.
Pathology
Little information is available on the pathologic findings in rubella occurring
postnatally. The few reported studies of biopsy or autopsy material from cases of
rubella revealed only nonspecific findings of lymphoreticular inflammation and
mononuclear perivascular and meningeal infiltration. The pathologic findings for
CRS are often severe and may involve nearly every organ system (Table 274.1 ).
Table 274.1
Pathologic Findings in Congenital Rubella Syndrome
Differential Diagnosis
Poliomyelitis should be considered in the differential diagnosis of any case of
paralysis and is only 1 of many causes of acute flaccid paralysis in children and
adults. There are numerous other causes of acute flaccid paralysis (Table 276.1 ).
In most conditions, the clinical features are sufficient to differentiate between
these various causes, but in some cases nerve conduction studies and
electromyograms, in addition to muscle biopsies, may be required.
Table 276.1
Differential Diagnosis of Acute Flaccid Paralysis
REDUCTION
SITE, OR
SENSORY
CONDITION, CLINICAL ONSET OF PROGRESSION ABSENCE RESIDUAL
SIGNS AND
FACTOR, OR FINDINGS PARALYSIS OF PARALYSIS OF DEEP PARALYSIS
SYMPTOMS
AGENT TENDON
REFLEXES
Anterior Horn Cells of Spinal Cord
Poliomyelitis Paralysis Incubation 24-48 hr to onset No Yes Yes
(wild and period 7-14 of full paralysis;
vaccine- days (range: 4- proximal →
associated 35 days) distal,
paralytic asymmetric
poliomyelitis)
Nonpolio Hand-foot-and- As in As in No Yes Yes
enteroviruses mouth disease, poliomyelitis poliomyelitis
(including EV- aseptic
A71, EV D68) meningitis,
acute
hemorrhagic
conjunctivitis,
possibly
idiopathic
epidemic flaccid
paralysis
West Nile virus Meningitis As in As in No Yes Yes
encephalitis poliomyelitis poliomyelitis
Other Neurotropic Viruses
Rabies virus Mo–Yr Acute, Yes Yes No
symmetric,
ascending
Varicella-zoster Exanthematous Incubation Acute, Yes ± ±
virus vesicular period 10-21 symmetric,
eruptions days ascending
Japanese Incubation Acute, proximal, ± ± ±
encephalitis period 5-15 asymmetric
virus days
Guillain-Barré Syndrome
Acute Preceding Hr to 10 days Acute, Yes Yes ±
inflammatory infection, symmetric,
polyradiculo- bilateral facial ascending (days
neuropathy weakness to 4 wk)
Acute motor Fulminant, Hr to 10 days 1-6 days No Yes ±
axonal widespread
neuropathy paralysis,
bilateral facial
weakness,
tongue
involvement
Acute Traumatic Sciatic Neuritis
Intramuscular Acute, Hr to 4 days Complete, Yes Yes ±
gluteal injection asymmetric affected limb
Acute transverse Preceding Acute, Hr to days Yes Yes, early Yes
myelitis Mycoplasma symmetric
pneumoniae, hypotonia of
Schistosoma , lower limbs
other parasitic
or viral
infection
Epidural abscess Headache, back Complete Yes Yes ±
pain, local
spinal
tenderness,
meningismus
Spinal cord Complete Hr to days Yes Yes ±
compression;
trauma
Neuropathies
Exotoxin of In severe cases, Incubation Yes Yes
Corynebacterium palatal period 1-8 wk
diphtheriae paralysis, (paralysis 8-12
blurred vision wk after onset
of illness)
Toxin of Abdominal Incubation Rapid, ± No
Clostridium pain, diplopia, period 18-36 hr descending,
botulinum loss of symmetric
accommodation,
mydriasis
Tick bite Ocular Latency period Acute, No Yes
paralysis symptoms 5-10 days symmetric,
ascending
Diseases of the Neuromuscular Junction
Myasthenia Weakness, Multifocal No No No
gravis fatigability,
diplopia, ptosis,
dysarthria
Disorders of Muscle
Polymyositis Neoplasm, Subacute, Wk to mo No Yes
autoimmune proximal →
disease distal
Viral myositis Pseudoparalysis Hr to days No No
Metabolic Disorders
Hypokalemic Proximal limb, Sudden No Yes ±
periodic respiratory postprandial
paralysis muscles
Intensive Care Unit Weakness
Critical illness Flaccid limbs Acute, Hr to days ± Yes ±
polyneuropathy and respiratory following
weakness systemic
inflammatory
response
syndrome/sepsis
Modified from Marx A, Glass JD, Sutter RW: Differential diagnosis of acute flaccid paralysis and
its role in poliomyelitis surveillance, Epidemiol Rev 22:298–316, 2000.
Nonpolio Enteroviruses
Kevin Messacar, Mark J. Abzug
The genus Enterovirus contains a large number of viruses spread via the
gastrointestinal and respiratory routes that produce a broad range of illnesses in
patients of all ages. Many of the manifestations predominantly affect infants and
young children.
Etiology
Enteroviruses are nonenveloped, single-stranded, positive-sense viruses in the
Picornaviridae (“small RNA virus”) family, which also includes the rhinoviruses,
hepatitis A virus, and parechoviruses. The original human enterovirus subgroups
—polioviruses (see Chapter 276 ), coxsackieviruses, and echoviruses—were
differentiated by their replication patterns in tissue culture and animals (Table
277.1 ). Enteroviruses have been reclassified on the basis of genetic similarity
into 4 species, human enteroviruses A-D. Specific enterovirus types are
distinguished by antigenic and genetic sequence differences, with enteroviruses
discovered after 1970 classified by species and number (e.g., enterovirus D68
and A71). Although more than 100 types have been described, 10-15 account for
the majority of disease. No disease is uniquely associated with any specific
serotype, although certain manifestations are preferentially associated with
specific serotypes. The closely related human parechoviruses can cause clinical
presentations similar to those associated with enteroviruses.
Table 277.1
Classification of Human Enteroviruses
Family Picornaviridae
Genus Enterovirus
Subgroups* Poliovirus serotypes 1-3
Coxsackie A virus serotypes 1-22, 24 (23 reclassified as echovirus 9)
Coxsackie B virus serotypes 1-6
Echovirus serotypes 1-9, 11-27, 29-33 (echoviruses 10 and 28 reclassified as non-enteroviruses;
echovirus 34 reclassified as a variant of coxsackie A virus 24; echoviruses 22 and 23 reclassified
within the genus Parechovirus )
Numbered enterovirus serotypes (enterovirus 72 reclassified as hepatitis A virus)
* The human enteroviruses have been alternatively classified on the basis of nucleotide and
Epidemiology
Enterovirus infections are common, with a worldwide distribution. In temperate
climates, annual epidemic peaks occur in summer/fall, although some
transmission occurs year-round. Enteroviruses are responsible for 33–65% of
acute febrile illnesses and 55–65% of hospitalizations for suspected sepsis in
infants during the summer and fall in the United States. In tropical and
semitropical areas, enteroviruses typically circulate year-round. In general, only
a few serotypes circulate simultaneously. Infections by different serotypes can
occur within the same season. Factors associated with increased incidence and/or
severity include young age, male sex, exposure to children, poor hygiene,
overcrowding, and low socioeconomic status. More than 25% of symptomatic
infections occur in children younger than 1 yr of age. Breastfeeding reduces the
risk for infection, likely via enterovirus-specific antibodies.
Humans are the only known natural reservoir for human enteroviruses. Virus
is primarily spread person to person, by the fecal-oral and respiratory routes,
although types causing acute hemorrhagic conjunctivitis may be spread via
airborne transmission. Virus can be transmitted vertically prenatally or in the
peripartum period, or, possibly, via breastfeeding. Enteroviruses can survive on
environmental surfaces, permitting transmission via fomites. Enteroviruses also
can frequently be isolated from water sources, sewage, and wet soil. Although
contamination of drinking water, swimming pools and ponds, and hospital water
reservoirs may occasionally be responsible for transmission, such contamination
is often considered the result rather than the cause of human infection.
Transmission is common within families (≥50% risk of spread to nonimmune
household contacts), daycare centers, playgrounds, summer camps, orphanages,
and hospital nurseries; severe secondary infections may occur in nursery
outbreaks. Transmission risk is increased by diaper changing and decreased by
Except for epidemiologic studies or cases characteristic of specific serotypes
(e.g., enterovirus A71), serology is generally less useful than culture or nucleic
acid detection.
Differential Diagnosis
The differential diagnosis of enterovirus infections varies with the clinical
presentation (Table 277.2 ).
Table 277.2
Table 280.1
Evidence of Immunity to Varicella
Evidence of immunity to varicella consists of any of the following:
• Documentation of age-appropriate vaccination with a varicella vaccine:
• Preschool-age children (i.e., age ≥12 mo): 1 dose
• School-age children, adolescents, and adults: 2 doses*
• Laboratory evidence of immunity † or laboratory confirmation of disease
• Birth in the United States before 1980 ‡
• Diagnosis or verification of a history of varicella disease by a healthcare provider §
• Diagnosis or verification of a history of herpes zoster by a healthcare provider
*
For children who received their 1st dose at younger than age 13 yr and for whom the interval
between the 2 doses was 28 or more days, the 2nd dose is considered valid.
† Commercial assays can be used to assess disease-induced immunity, but they lack sensitivity to
always detect vaccine-induced immunity (i.e., they might yield false-negative results).
‡ For healthcare personnel, pregnant women, and immunocompromised persons, birth before
1980 should not be considered evidence of immunity.
§
Verification of history or diagnosis of typical disease can be provided by any healthcare provider
(e.g., school or occupational clinic nurse, nurse practitioner, physician assistant, or physician). For
persons reporting a history of, or reporting with, atypical or mild cases, assessment by a physician
or his/her designee is recommended, and one of the following should be sought: (1) an
epidemiologic link to a typical varicella case or to a laboratory-confirmed case or (2) evidence of
laboratory confirmation if it was performed at the time of acute disease. When such
documentation is lacking, persons should not be considered as having a valid history of disease,
because other diseases might mimic mild atypical varicella.
Vaccine
Varicella is a vaccine-preventable disease. Varicella vaccine contains live,
attenuated VZV (Oka strain) and is indicated for subcutaneous administration. In
the United States, varicella vaccine is recommended for routine administration
as a 2-dose regimen to healthy children at ages 12-15 mo and 4-6 yr.
Administration of the 2nd dose earlier than 4-6 yr of age is acceptable, but it
FIG. 281.3 Kinetics of antibody responses to Epstein-Barr virus (EBV) antigens in
infectious mononucleosis. EA, early antigen; EBNA, EBV-determined nuclear antigens;
IgG, immunoglobulin G; IgM, immunoglobulin M; VCA, viral capsid antigen.
Table 281.1
Correlation of Clinical Status and Antibody Responses to
Epstein-Barr Virus Infection
Anti-EA IgG antibodies are usually detectable for several months but may
persist or be detected intermittently at low levels for many years. Antibodies to
the diffuse-staining component of EA (EA-D) are found transiently in 80% of
patients during the acute phase of infectious mononucleosis. Antibodies to the
cytoplasmic-restricted component of EA (EA-R) emerge transiently in the
convalescence from infectious mononucleosis. High levels of antibodies to EA-
D or EA-R may be found also in immunocompromised patients with persistent
EBV infections and active EBV replication.
loss associated with congenital CMV infection will pass an initial hearing
screening exam but develop hearing loss in later infancy and early childhood.
Importantly, hearing loss can be progressive in infants with hearing loss
secondary to congenital CMV infections. Lastly, the diagnosis of congenital
CMV infection must be made within the first 2-3 wk of life, and congenital
CMV infection cannot be assumed to be the cause of hearing loss in older infants
without evidence of CMV infection in the newborn period.
Table 282.1
Findings in Infants With Symptomatic Congenital
Cytomegalovirus Infection
FINDINGS % OF INFANTS
CLINICAL FINDINGS
Prematurity (<37 wk) 24
Jaundice (direct bilirubin >2 mg/dL) 42
Petechiae 54
Hepatosplenomegaly 19
Purpura 3
Microcephaly 35
IUGR 28
1 clinical finding 41
2 clinical findings 59
LABORATORY FINDINGS
Elevated ALT (>80 IU/mL) 71
Thrombocytopenia (<100,000 k/mm3 ) 43
Direct hyperbilirubinemia (>2 mg/dL) 54
Head CT abnormalities 42
Findings in 70 infants with symptomatic congenital CMV infection identified during newborn
screening program for infants with congenital CMV infection at the University of Alabama
Hospitals over an approximate 20 yr interval.
CMV, cytomegalovirus; IUGR, in utero growth retardation; ALT, alanine aminotransferase.
Table 285.1
Subtypes of Novel Influenza A Viruses and Clinical
Syndromes in Human Infections
VARIANT
LPAI VIRUSES HPAI VIRUSES
VIRUSES*
Conjunctivitis H7N2, H7N3, H7N7, H10N7 H7N3, H7N7 H1N1v, H3N2v
Upper respiratory tract illness H6N1, H7N2, H7N3, H7N9, H5N1, H5N6, H1N1v, H1N2v,
H9N2, H10N7 H7N7 H3N2v
Lower respiratory tract disease, H7N2, H7N9, H9N2, H10N8 H5N1, H5N6, H1N1v, H3N2v
pneumonia H7N7, H7N9
Respiratory failure, acute respiratory H7N9, H10N8 H5N1, H5N6, H1N1v, H3N2v
distress syndrome H7N7, H7N9
Multiorgan failure H7N9, H10N8 H5N1, H5N6, —
H7N7, H7N9
Encephalopathy or encephalitis H7N9 H5N1 —
Fatal outcomes † H7N9, H9N2, H10N8 H5N1, H5N6, H1N1v, H3N2v
H7N7, H7N9
* Variant viruses of swine origin.
† High mortality in reported cases: about 40% for LPAI H7N9, about 50% for HPAI H5N1, and
Seasonal Influenza
An estimated 11,000-45,000 children younger than 18 yr of age are hospitalized
annually in the United States as a result of seasonal influenza-associated
complications, with approximately 6,000-26,000 hospitalizations in children
required for clinical decisions to prescribe antiviral medications, and prompt
suspicion or diagnosis of influenza may allow for early antiviral therapy to be
initiated and may reduce inappropriate use of antibiotics.
A number of diagnostic tests may be used for laboratory confirmation of
influenza (Table 285.2 ). Although rapid influenza diagnostic tests are often
employed because of their ease of use and fast results, they can have suboptimal
sensitivity to detect influenza virus infection, particularly for novel influenza
viruses. Sensitivities of rapid diagnostic tests are generally 50–70% compared to
viral culture or reverse-transcription polymerase chain reaction. Specificities are
higher, approximately 95–100%. Therefore false-negative results occur more
often than false-positive results, particularly when the prevalence of influenza is
high (i.e., during peak influenza activity in the community). The interpretation of
negative results should take into account the clinical characteristics and the
patient's risk for complications. If there is clinical suspicion for influenza in a
patient at high risk for complications (Table 285.3 ), early empiric treatment
should be given regardless of a negative rapid diagnostic test result, and another
type of test (e.g., reverse-transcription polymerase chain reaction or direct
fluorescent antibody testing) may be performed for confirmation.
Table 285.2
Influenza Virus Testing Methods
ACCEPTABLE TEST
METHOD COMMENTS
SPECIMENS TIME
Rapid Influenza Diagnostic Tests (antigen Nasopharyngeal (NP) <15 min Rapid turnaround;
detection) swab, aspirate or wash, suboptimal sensitivity
nasal swab, aspirate, or
wash, throat swab
Rapid Molecular Assay (influenza nucleic NP swab, nasal swab 15-30 min Rapid turnaround, high
acid amplification) sensitivity
Immunofluorescence, Direct (DFA) or NP swab or wash, 1-4 hr Relatively rapid
Indirect (IFA) Fluorescent Antibody bronchial wash, nasal or turnaround; requires
Staining (antigen detection) endotracheal aspirate laboratory expertise and
experience
RT-PCR (singleplex and multiplex; real- NP swab, throat swab, NP Varies by Excellent sensitivity,
time and other RNA-based) and other or bronchial wash, nasal or assay relatively rapid turnaround
molecular assays (influenza nucleic acid endotracheal aspirate, (generally 1- compared with
amplification) sputum 8 hr) conventional methods
Rapid cell culture (shell vials, cell NP swab, throat swab, NP 1-3 day Culture isolates important
mixtures; yields live virus) or bronchial wash, nasal or for strain information and
endotracheal aspirate, antiviral resistance
sputum monitoring
Viral tissue cell culture (conventional; NP swab, throat swab, NP 3-10 day Not recommended for
yields live virus) or bronchial wash, nasal or routine patient diagnosis
endotracheal aspirate,
sputum
Serologic tests (antibody detection) Paired (appropriately N/A (not Not recommended for
timed) acute and performed routine patient diagnosis,
convalescent serum during acute useful for research studies
specimens infection)
N/A, not applicable; NP, nucleoprotein; RT-PCR, reverse transcription-polymerase chain reaction.
Modified from Centers for Disease Control and Prevention (CDC): Influenza virus testing
methods. Available at https://www.cdc.gov/flu/professionals/diagnosis/table-testing-methods.htm
in Information for Health Professionals (https://www.cdc.gov/flu/professionals/index.htm ); and
from 2018 IDSA Clinical Practice Guidelines.
Table 285.3
Children and Adolescents Who Are at Higher Risk for
Influenza Complications for Whom Antiviral Treatment is
Recommended*
Table 285.4
Recommended Dosage and Schedule of Influenza Antiviral
Medications for Treatment and Chemoprophylaxis in
Children for the 2018-2019 Influenza Season: United States
TREATMENT
MEDICATION CHEMOPROPHYLAXIS DOSING**
DOSING**
ORAL OSELTAMIVIR *
Adults 75 mg twice daily 75 mg once daily
Children ≥12 mo
Body wt
≤15 kg (≤33 lb) 30 mg twice daily 30 mg once daily
>15-23 kg (33-51 lb) 45 mg twice daily 45 mg once daily
>23-40 kg (>51-88 60 mg twice daily 60 mg once daily
lb)
>40 kg (>88 lb) 75 mg twice daily 75 mg once daily
Infants 0-11 mo † 3 mg/kg per dose once 3 mg/kg per dose once daily
daily
Term infants ages 0-8 3 mg/kg per dose twice 3 mg/kg per dose once daily for infants 3-8 mo old; not
mo † daily recommended for infants <3 mo old unless situation judged
critical because of limited safety and efficacy data in this age
group
Preterm infants See details in footnote ‡ Not recommended
INHALED ZANAMIVIR §
Adults 10 mg (two 5 mg 10 mg (two 5 mg inhalations) once daily
inhalations) twice daily
Children (≥7 yr old 10 mg (two 5 mg 10 mg (two 5 mg inhalations) once daily
for treatment; ≥5 yr inhalations) twice daily
old for
chemoprophylaxis)
INTRAVENOUS PERAMIVIR
Adults 600 mg intravenous Not recommended
infusion once given over
15-30 min
Children (2-12 yr One 12 mg/kg dose, up to Not recommended
old) 600 mg maximum, via
intravenous infusion for 15-
30 min
Children (13-17 yr One 600 mg dose via Not recommended
old) intravenous infusion for 15-
30 min
ORAL BALOXAVIR ††
Adults
40 to <80 kg One 40 mg dose Not recommended
>80 kg One 80 mg dose Not recommended
Children
2-11 yr Not recommended Not recommended
12-17 yr, 40 to <80 One 40 mg dose Not recommended
kg
12-17 yr, >80 kg One 80 mg dose Not recommended
* Oseltamivir is administered orally without regard to meals, although administration with meals
data and limited safety data, oseltamivir can be used to treat influenza in both term and preterm
infants from birth because benefits of therapy are likely to outweigh possible risks of treatment.
CDC and US Food and Drug Administration (FDA)–approved dosing is 3 mg/kg per dose twice
daily for children aged 9-11 mo; the American Academy of Pediatrics recommends 3.5 mg/kg per
dose twice daily. The dose of 3 mg/kg provides oseltamivir exposure in children similar to that
achieved by the approved dose of 75 mg orally twice daily for adults, as shown in two studies of
oseltamivir pharmacokinetics in children. The AAP has recommended an oseltamivir treatment
dose of 3.5 mg/kg orally twice daily for infants 9-11 mo, on the basis of data that indicated that a
higher dose of 3.5 mg/kg was needed to achieve the protocol-defined targeted exposure for this
cohort as defined in the CASG 114 study. It is unknown whether this higher dose will improve
efficacy or prevent the development of antiviral resistance. However, there is no evidence that the
3.5 mg/kg dose is harmful or causes more adverse events to infants in this age group.
‡ Oseltamivir dosing for preterm infants. The wt-based dosing recommendation for preterm infants
is lower than for term infants. Preterm infants may have lower clearance of oseltamivir because of
immature renal function, and doses recommended for term infants may lead to high drug
concentrations in this age group. Limited data from the National Institute of Allergy and Infectious
Diseases Collaborative Antiviral Study Group provide the basis for dosing preterm infants by
using their postmenstrual age (gestational age plus chronological age): 1.0 mg/kg per dose orally
twice daily for those <38 wk postmenstrual age; 1.5 mg/kg per dose orally twice daily for those 38-
40 wk postmenstrual age; and 3.0 mg/kg per dose orally twice daily for those >40 wk
postmenstrual age. For extremely preterm infants (<28 wk), please consult a pediatric infectious
diseases physician.
§ Zanamivir is administered by inhalation by using a proprietary Diskhaler device distributed
Pathogenesis
Infection occurs via inoculation of the upper respiratory tract. Infection can
spread rapidly to the lower respiratory tract, but it is not clear whether the
dissemination is mediated by cell-to-cell spread or by aspiration of infected
materials from the upper tract. Severe lower respiratory tract illness, especially
wheezing, occurs mainly during the first year of life, at a time when the airways
are of a very small diameter and thus a high resistance. Maternal serum-
neutralizing antibodies that cross the placenta may afford a relative protection
against severe disease for several weeks or months after birth. Once infection is
established, it is likely that cytotoxic T cells recognize and eliminate virus-
infected cells, thus terminating the infection but also causing some
cytopathology. The virus appears to have specific mechanisms for inhibiting T-
cell responses during acute infection. Individuals with an underlying
predisposition for reactive airways disease (including adults) are susceptible to
severe wheezing during reinfection later in life, suggesting that HMPV may
cause smooth muscle hyperactivity, inflammation, or increased mucus
production in such individuals. Infection in otherwise healthy individuals
resolves without apparent long-term consequences in most cases. HMPV
infection is associated with exacerbations of asthma later in life.
Clinical Manifestations
HMPV is associated with the common cold (complicated by otitis media in
approximately 30% of cases) and with lower respiratory tract illnesses such as
bronchiolitis, pneumonia, croup, and exacerbation of reactive airways disease.
The profile of signs and symptoms caused by HMPV is very similar to that
caused by RSV (Table 288.1 ). Approximately 5–10% of outpatient lower
respiratory tract illnesses in otherwise healthy young children is associated with
HMPV infection, which is second in incidence only to RSV. Children with RSV
or HMPV infection require supplemental oxygen and medical intensive care at
similar frequencies.
Table 288.1
About half of the cases of HMPV lower respiratory tract illness in children
occur in the first 6 mo of life, suggesting that young age is a major risk factor for
severe disease. Both young adults and the elderly can have HMPV infection that
requires medical care including hospitalization, but severe disease occurs at
much lower frequencies in adults than in young children. Severe disease in
pediatric and older subjects is most common in immunocompromised patients or
those with complications of preterm birth, congenital heart disease, and
neuromuscular disease and can be fatal. A significant number of both adult and
pediatric patients with asthma exacerbations have HMPV infection; it is not clear
whether the virus causes long-term wheezing. RSV and HMPV coinfections
have been reported; coinfections may be more severe than infection with a single
virus, resulting in pediatric intensive care unit admissions. It is difficult to define
true coinfections because these viral RNA genomes can be detected by a reverse
transcriptase polymerase chain reaction (PCR) in respiratory secretions for at
least several weeks after illness, even when virus shedding has terminated.
Laboratory Findings
The virus can be visualized only with electron microscopy. The virus grows in
CHAPTER 289
Adenoviruses
Jason B. Weinberg, John V. Williams
Etiology
Adenoviruses are nonenveloped viruses with an icosahedral protein capsid. The
double-stranded DNA genome is contained within the particle complexed with
several viral proteins. Antigenic variability in surface proteins of the virion and
genomic sequencing define at least 70 serotypes grouped into seven species.
Species differ in their tissue tropism and target organs, causing distinct clinical
infections (Table 289.1 ). HAdVs can be shed from the gastrointestinal tract for
prolonged periods and can establish persistent infection of the tonsils and
adenoids.
Table 289.1
Epidemiology
HAdVs circulate worldwide and cause endemic infections year-round in
immunocompetent hosts. Asymptomatic infections are also common. Only about
one third of all known HAdV types are associated with clinically apparent
disease. The most prevalent types in recent surveillance studies are HAdV types
3, 2, 1, and 5. Epidemics of conjunctivitis (often severe), pharyngitis, and
respiratory disease occur, especially in schools and military settings. Outbreaks
of febrile respiratory illness caused by HAdV-4 and HAdV-7 are a major source
of morbidity in military barracks, with attack rates ranging from 25% to > 90%.
The spread of HAdV occurs by respiratory and fecal-oral routes. An important
factor in HAdV transmission, especially in epidemics, is the ability of the
nonenveloped particle to survive on inanimate objects in the environment.
Nosocomial outbreaks have been reported.
Pathogenesis
HAdVs bind to cell surface receptors and trigger internalization by endocytosis.
Acidification of the endosome induces conformational changes in the capsid,
leading to eventual translocation of the genome to the cell nucleus. Viral
messenger RNA transcription and genomic replication occur in the nucleus.
Progeny virion particles assemble in the nucleus. Lysis of the cell releases new
infectious particles and causes damage to epithelial mucosa, sloughing of cell
debris, and inflammation. Host responses to HAdV infection include the
recruitment of neutrophils, macrophages, and natural killer cells to the site of
infection and the elaboration by those cells of a number of cytokines and
chemokines. This host immune response is likely to contribute to the symptoms
of HAdV infection, but specific mechanisms of pathogenesis are poorly
understood. The strict species specificity of the adenoviruses has precluded the
development of an animal model for HAdVs, although recent work with HAdV
HPV DNA detected in older women reflects those HPV infections that became
established persistent infections. Persistence is now the known necessary
prerequisite for the development of significant precancerous lesions and cervical
cancer.
Approximately 15–20% of sexually active adolescents have detectable HPV at
any given time and have normal cytologic findings. The most common clinically
detected lesion in adolescent women is the cervical lesion termed low-grade
squamous intraepithelial lesion (LSIL) (Table 293.1 ). LSILs can be found in
25–30% of adolescents infected with HPV. External genital warts are much less
common, occurring in < 1% of adolescents, but approximately 10% of
individuals will develop genital warts in their lifetime. LSIL is a cytologic and
histologic term to reflect the benign changes caused by an active viral infection
and is likely present in most, if not all, women with HPV infection. The majority
of women, however, have very minute or subtle lesions not easily detected by
cytology. As with HPV DNA detection, most LSILs regress spontaneously in
young women and do not require any intervention or therapy. Less commonly,
HPV can induce more severe cellular changes, termed high-grade squamous
intraepithelial lesions (HSILs) (see Chapter 568 ).
Table 293.1
Terminology for Reporting Cervical Cytology and Histology
DESCRIPTIVE DIAGNOSIS OF
EQUIVALENT TERMINOLOGY
EPITHELIAL CELL ABNORMALITIES
SQUAMOUS CELL
Atypical squamous cells of undetermined Squamous atypia
significance (ASC-US)
Atypical squamous cells, cannot exclude
HSIL (ASC-H)
Low-grade squamous intraepithelial lesion Mild dysplasia, condylomatous atypia, HPV-related changes,
(LSIL) koilocytic atypia, cervical intraepithelial neoplasia (CIN) 1
High-grade squamous intraepithelial lesion Moderate dysplasia, CIN 2, severe dysplasia, CIN 3, carcinoma in
(HSIL) situ
GLANDULAR CELL
Endometrial cells, cytologically benign,
in a postmenopausal woman
Atypical
Endocervical cells, NOS
Endometrial cells, NOS
Glandular cells, NOS
Endocervical cells, favor neoplastic
Glandular cells, favor neoplastic
Pathogenesis
infection is termed the congenital Zika syndrome (CZS), which consists of
microcephaly, facial disproportion, hypertonia/spasticity, hyperreflexia,
irritability, seizures, arthrogryposis, ocular abnormalities, and sensorineural
hearing loss (Table 294.1 ). A comprehensive understanding of the precise
antecedents to CZS is not known. It appears that the earlier during pregnancy
that ZIKV infections occur, the greater the likelihood of and the more severe the
CZS. Vertical transmission appears to follow viremia with ZIKV, transiting the
uterus to infect the placenta and then the fetus. However, factors that affect the
occurrence or severity of CZS, such as age, ethnicity, or prior immune status of
the mother, are not known. In vitro studies have demonstrated that dengue
antibodies can enhance ZIKV infection in vitro, in Fc-receptor–bearing cells,
but, as yet, there is no evidence that a prior dengue infection alters the chance of
ZIKV crossing the placenta or increases the risk of CZS. Maternal-fetal
transmission of ZIKV can occur during labor and delivery. There are no reports
of ZIKV infection acquired by an infant at the time of delivery leading to
microcephaly. There are no data to contraindicate breastfeeding, although the
virus has been identified in breast milk. Maternal and newborn laboratory testing
is indicated during the first 2 wk of life if the mother had relevant epidemiologic
exposure within 2 wk of delivery and had clinical manifestations of ZIKV
infection (e.g., rash, conjunctivitis, arthralgia, or fever). Infants and children who
acquire ZIKV infection postnatally appear to have a mild course, similar to that
seen in adults.
Table 294.1
Surveillance Case Classification: Children, Neonate to 2
Years of Age, Born to Mothers With Any Evidence of Zika
Virus Infection During Pregnancy
ZIKA-ASSOCIATED BIRTH DEFECTS
Selected structural anomalies of the brain or eyes present at birth (congenital) and detected from birth to age
2 yr. Microcephaly at birth, with or without low birthweight, was included as a structural anomaly.
• Selected congenital brain anomalies: intracranial calcifications; cerebral atrophy; abnormal cortical formation
(e.g., polymicrogyria, lissencephaly, pachygyria, schizencephaly, gray matter heterotopia); corpus callosum
abnormalities; cerebellar abnormalities; porencephaly; hydranencephaly; ventriculomegaly/hydrocephaly.
• Selected congenital eye anomalies: microphthalmia or anophthalmia; coloboma; cataract; intraocular
calcifications; chorioretinal anomalies involving the macula (e.g., chorioretinal atrophy and scarring, macular
pallor, and gross pigmentary mottling), excluding retinopathy of prematurity; optic nerve atrophy, pallor, and
other optic nerve abnormalities.
• Microcephaly at birth: birth head circumference < 3rd percentile for infant sex and gestational age based on
INTERGROWTH-21st online percentile calculator (http://intergrowth21.ndog.ox.ac.uk/ ).
NEURODEVELOPMENTAL ABNORMALITIES POSSIBLY ASSOCIATED WITH CONGENITAL ZIKA
VIRUS INFECTION
Consequences of neurologic dysfunction detected from birth (congenital) to age 2 yr. Postnatal-onset
microcephaly was included as a neurodevelopmental abnormality.
• Hearing abnormalities: Hearing loss or deafness documented by testing, most frequently auditory brainstem
response (ABR). Includes sensorineural hearing loss, mixed hearing loss, and hearing loss not otherwise
specified. Failed newborn hearing screening is not sufficient for diagnosis.
• Congenital contractures: Multiple contractures (arthrogryposis) and isolated clubfoot documented at birth.
Brain anomalies must be documented for isolated clubfoot but not for arthrogryposis.
• Seizures: Documented by electroencephalogram or physician report. Includes epilepsy or seizures not otherwise
specified; excludes febrile seizures.
• Body tone abnormalities: Hypertonia or hypotonia documented at any age in conjunction with (1) a failed
screen or assessment for gross motor function; (2) suspicion or diagnosis of cerebral palsy from age 1 to 2 yr; or
(3) assessment by a physician or other medical professional, such as a physical therapist.
• Movement abnormalities: Dyskinesia or dystonia at any age; suspicion or diagnosis of cerebral palsy from age
1 to 2 yr.
• Swallowing abnormalities: Documented by instrumented or noninstrumented evaluation, presence of a
gastrostomy tube, or physician report.
• Possible developmental delay: Abnormal result from most recent developmental screening (i.e., failed screen
for gross motor domain or failed screen for two or more developmental domains at the same time point or age);
developmental evaluation; or assessment review by developmental pediatrician. Results from developmental
evaluation are considered the gold standard if available.
• Possible visual impairment: Includes strabismus (esotropia or exotropia), nystagmus, failure to fix and follow
at age < 1 yr; diagnosis of visual impairment at age ≥ 1 yr.
• Postnatal-onset microcephaly: Two most recent head circumference measurements reported from follow-up
care < 3rd percentile for child's sex and age based on World Health Organization child growth standards;
downward trajectory of head circumference percentiles with most recent measurement < 3rd percentile. Age at
measurement was adjusted for gestational age in infants born at < 40 wk of gestational age through age 24 mo
chronologic age.
From Rice ME, Galang RR, Roth NM, et al: Vital signs: Zika-associated birth defects and
neurodevelopmental abnormalities possibly associated with congenital Zika virus infection: US
territories and freely associated states, 2018. MMWR 67(31):858-866, 2018.
Clinical Features
Congenital Zika syndrome may be defined in a fetus with diagnostic evidence
of ZIKV infection, including (1) severe microcephaly (>3 SD below the mean),
partially collapsed skull, overlapping cranial sutures, prominent occipital bone,
redundant scalp skin, and neurologic impairment; (2) brain anomalies, including
cerebral cortex thinning, abnormal gyral patterns, increased fluid spaces,
subcortical calcifications, corpus callosum anomalies, reduced white matter, and
cerebellar vermis hypoplasia; (3) ocular findings, such as macular scarring, focal
pigmentary retinal mottling, structural anomalies (microphthalmia, coloboma,
cataracts, and posterior anomalies), chorioretinal atrophy, or optic nerve
hypoplasia/atrophy; (4) congenital contractures, including unilateral or bilateral
clubfoot and arthrogryposis multiplex congenita; and (5) neurologic impairment,
such as pronounced early hypertonia/spasticity with extrapyramidal symptoms,
motor disabilities, cognitive disabilities, hypotonia, irritability/excessive crying,
tremors, swallowing dysfunction, vision impairment, hearing impairment, and
epilepsy (see Table 294.1 ).
Acquired Zika virus infection may present with nonspecific viral syndrome–
like features. Nonetheless, patients are at increased risk of myelitis and Guillain-
Barré syndrome. In addition, the virus may remain present in the blood and body
fluids for months after resolution of clinical symptoms.
Management
For infants with confirmed Zika virus infection, close follow-up is necessary.
The appropriate follow-up evaluation depends upon whether or not the infant has
clinical signs and symptoms of congenital Zika syndrome. All infants should
have close monitoring of growth and development, repeat ophthalmologic
examinations, and auditory brainstem response testing (see Table 294.1 ).
Laboratory Diagnosis
Laboratory testing for Zika virus infection in the neonate includes the following:
serum and urine for Zika virus RNA via real-time reverse transcription
polymerase chain reaction (rRT-PCR) and serum Zika virus immunoglobulin M
(IgM) enzyme-linked immunosorbent assay (ELISA). If the IgM is positive, the
plaque reduction neutralization test (PRNT) is used to confirm the specificity of
the IgM antibodies against Zika virus and to exclude a false-positive IgM result.
If CSF is available, it should be tested for Zika virus RNA (via rRT-PCR), as
well as Zika virus IgM. CSF specimens need not be collected for the sole
purpose of Zika virus testing but may be reasonable for the evaluation of infants
with microcephaly or intracranial calcifications. A definitive diagnosis of
congenital Zika virus infection is confirmed by the presence of Zika virus RNA
in samples of serum, urine, or CSF collected within the first 2 days of life; IgM
antibodies may be positive or negative. A negative rRT-PCR result with a
positive Zika virus IgM test result indicates probable congenital Zika virus
infection.
Fetuses or infants born to mothers who test positive for ZIKV infection should
be studied sonographically or for clinical evidence of congenital Zika syndrome,
a comprehensive evaluation (including ophthalmologic examination, laboratory
tests, and specialist consultation) should be performed prior to hospital
discharge.
Prognosis
The prognosis of newborns with congenital Zika syndrome is unclear. Reported
acute mortality rates among live-born infants range from 4% to 6%. The
combination of Zika virus–related microcephaly and severe cerebral
abnormalities generally has a poor prognosis, but little is known about the
prognosis for congenitally infected infants with less severe or no apparent
abnormalities at birth.
Differential Diagnosis
The differential diagnosis for congenital Zika virus infection includes other
congenital infections and other causes of microcephaly.
Prevention
The prevention of the congenital Zika syndrome includes avoidance, if possible,
of travel to endemic regions; if travel to endemic regions cannot be avoided,
careful contraception (male and female) is essential, especially with the
knowledge that Zika virus can persist in semen for months after a primary
infection (Table 294.2 ).
Table 294.2
Bibliography
Adebanjo T, Godfred-Cato S, Viens L, et al. Update: interim
guidance for the diagnois, evaluation, and management of
infants with possible congenital Zika virus infection—United
States, October 2017. MMWR Morb Mortal Wkly Rep .
2017;66(41):1089–1098.
Baud D, Gubler DJ, Schaub B, et al. An update on Zika virus
infection. Lancet . 2017;390:2099–2109.
Brasil P, Sequeria PC, Freitas AD, et al. Guillain-Barre
syndrome associated with Zika virus infection. Lancet .
2016;387:1482.
Brooks RB, Carlos MP, Myers RA, et al. Likely sexual
transmission of Zika virus from a man with no symptoms of
infection—Maryland, 2016. MMWR Morb Mortal Wkly Rep .
2016;65(34):915–916.
CHAPTER 295
Etiology
There are at least four distinct antigenic types of dengue virus (dengue 1, 2, 3,
and 4), members of the family Flaviviridae. In addition, three other arthropod-
borne viruses (arboviruses) cause similar dengue fever syndromes with rash
(Table 295.1 ; see also Chapter 294 ).
Table 295.1
Vectors and Geographic Distribution of Dengue-Like
Diseases
GEOGRAPHIC GENUS AND
VIRUS VECTOR DISTRIBUTION
DISEASE
Togavirus Chikungunya Aedes aegypti Africa, India, Southeast Asia, Latin America,
Aedes United States
africanus
Aedes
albopictus
Togavirus O'nyong-nyong Anopheles East Africa
funestus
Flavivirus West Nile fever Culex molestus Europe, Africa, Middle East, India
Culex
univittatus
Epidemiology
Dengue viruses are transmitted by mosquitoes of the Stegomyia family. Aedes
aegypti, a daytime biting mosquito, is the principal vector, and all four virus
types have been recovered from it. Transmission occurs from viremic humans by
bite of the vector mosquito where virus multiplies during an extrinsic incubation
period and then by bite is passed on to a susceptible human in what is called the
urban transmission cycle. In most tropical areas, A. aegypti is highly urbanized,
breeding in water stored for drinking or bathing and in rainwater collected in any
container. Dengue viruses have also been recovered from Aedes albopictus, as in
the 2001 and 2015 Hawaiian epidemics, whereas outbreaks in the Pacific area
have been attributed to several other Aedes species. These species breed in water
trapped in vegetation. In Southeast Asia and West Africa, dengue virus may be
maintained in a cycle involving canopy-feeding jungle monkeys and Aedes
species, which feed on monkeys.
In the 19th and 20th centuries, epidemics were common in temperate areas of
the Americas, Europe, Australia, and Asia. Dengue fever and dengue-like
disease are now endemic in tropical Asia, the South Pacific Islands, northern
Australia, tropical Africa, the Arabian Peninsula, the Caribbean, and Central and
South America (Fig. 295.1 ). Dengue fever occurs frequently among travelers to
these areas. Locally acquired disease has been reported in Florida, Arizona, and
Texas, and imported cases in the United States occur in travelers to endemic
areas. More than 390 million dengue infections occur annually; approximately
96 million have clinical disease.
CHAPTER 297
Etiology
Six of the viral hemorrhagic fevers are caused by arthropod-borne viruses
(arboviruses) (Table 297.1 ). Four are caused by togaviruses of the family
Flaviviridae: Kyasanur Forest disease, Omsk hemorrhagic fever, dengue (see
Chapter 295 ), and yellow fever (see Chapter 296 ) viruses. Three are caused by
viruses of the family Bunyaviridae: Congo fever, Hantaan fever, and Rift Valley
fever (RVF) viruses. Four are caused by viruses of the family Arenaviridae:
Junin fever, Machupo fever, Guanarito fever, and Lassa fever. Two are caused by
viruses in the family Filoviridae : Ebola virus and Marburg virus, enveloped,
filamentous RNA viruses that are sometimes branched, unlike any other known
virus.
Table 297.1
Viral Hemorrhagic Fevers
† Chikungunya virus is associated infrequently with petechiae and epistaxis. Severe hemorrhagic
Epidemiology
With some exceptions, the viruses causing viral hemorrhagic fevers are
transmitted to humans via a nonhuman entity. The specific ecosystem required
for viral survival determines the geographic distribution of disease. Although it
is commonly thought that all viral hemorrhagic fevers are arthropod borne, seven
may be contracted from environmental contamination caused by animals or
animal cells or from infected humans (see Table 297.1 ). Laboratory and hospital
infections have occurred with many of these agents. Lassa fever and Argentine
and Bolivian hemorrhagic fevers are reportedly milder in children than in adults.
Table 297.2
Table 297.3
In 35–50% of patients, these diseases may become severe, with persistent high
temperature, increasing toxicity, swelling of the face or neck, microscopic
hematuria, and frank hemorrhages from the stomach, intestines, nose, gums, and
uterus. A syndrome of hypovolemic shock is accompanied by pleural effusion
and renal failure. Respiratory distress resulting from airway obstruction,
pleural effusion, or congestive heart failure may occur. A total of 10–20% of
patients experience late neurologic involvement, characterized by intention
tremor of the tongue and associated speech abnormalities. In severe cases, there
may be intention tremors of the extremities, seizures, and delirium. The
cerebrospinal fluid is normal. In Lassa fever, nerve deafness occurs in early
convalescence in 25% of cases. Prolonged convalescence is accompanied by
alopecia and, in Argentine and Bolivian hemorrhagic fevers, by signs of
autonomic nervous system lability, such as postural hypotension, spontaneous
flushing or blanching of the skin, and intermittent diaphoresis.
Laboratory studies reveal marked leukopenia, mild to moderate
thrombocytopenia, proteinuria, and, in Argentine hemorrhagic fever, moderate
abnormalities in blood clotting, decreased fibrinogen, increased fibrinogen split
Prevention
Primary prevention of rabies infection includes vaccination of domestic animals
and education to avoid wild animals, stray animals, and animals with unusual
behavior.
Postexposure Prophylaxis
The relevance of rabies for most pediatricians centers on evaluating whether an
animal exposure warrants PEP (Table 300.1 ). No case of rabies has been
documented in a person receiving the recommended schedule of PEP since
introduction of modern cellular vaccines in the 1970s.
Table 300.1
Rabies Postexposure Prophylaxis Guide
treatment of the exposed person with RIG (human) and vaccine should be initiated. The animal
should be euthanized immediately and tested.
† The animal should be euthanized and tested as soon as possible. Holding for observation is not
recommended. Immunization is discontinued if the immunofluorescent test result for the animal is
negative.
RIG, rabies immunoglobulin.
Given the incubation period for rabies, PEP is a medical urgency, not
emergency. Algorithms have been devised to aid practitioners in deciding when
to initiate rabies PEP (Fig. 300.1 ). The decision to proceed ultimately depends
on the local epidemiology of animal rabies as determined by active surveillance
programs, information that can be obtained from local and state health
departments. In general, bats, raccoons, skunks, coyotes, and foxes should be
considered rabid unless proven otherwise through euthanasia and testing of
brain tissue, whereas bites from small herbivorous animals (squirrels, hamsters,
gerbils, chipmunks, rats, mice, and rabbits) can be discounted. The response to
bites from a pet, particularly a dog, cat, or ferret, depends on local surveillance
statistics and on whether the animal is vaccinated and available for observation.
slower progression of disease.
The CDC Surveillance Case Definition for HIV infection is based on the age-
specific CD4+ T-lymphocyte count or the CD4+ T-lymphocyte percentage of
total lymphocytes (Table 302.1 ), except when a stage 3–defining opportunistic
illness (Table 302.2 ) supersedes the CD4 data. Age adjustment of the absolute
CD4 count is necessary because counts that are relatively high in normal infants
decline steadily until age 6 yr, when they reach adult norms. The CD4 count
takes precedence over the CD4 T-lymphocyte percentage, and the percentage is
considered only if the count is unavailable.
Table 302.1
HIV Infection Stage* Based on Age-Specific CD4+ T-
Lymphocyte Count or CD4+ T-Lymphocyte Percentage of
Total Lymphocytes
precedence over the CD4+ T-lymphocyte percentage, and the percentage is considered only if the
count is missing.
From Centers for Disease Control and Prevention: Revised surveillance case definition for HIV
infection—United States, 2014, MMWR 63(No RR-3):1-10, 2014.
Table 302.2
Stage 3–Defining Opportunistic Illnesses in HIV Infection
Bacterial infections, multiple or recurrent*
Candidiasis of bronchi, trachea, or lungs
Candidiasis of esophagus
Cervical cancer, invasive †
Coccidioidomycosis, disseminated or extrapulmonary
Cryptococcosis, extrapulmonary
Cryptosporidiosis, chronic intestinal (>1 mo duration)
Cytomegalovirus disease (other than liver, spleen, or nodes), onset at age > 1 mo
Cytomegalovirus retinitis (with loss of vision)
Encephalopathy attributed to HIV ‡
Herpes simplex: chronic ulcers (>1 mo duration) or bronchitis, pneumonitis, or esophagitis (onset at age > 1 mo)
Histoplasmosis, disseminated or extrapulmonary
Isosporiasis, chronic intestinal (>1 mo duration)
Kaposi sarcoma
Lymphoma, Burkitt (or equivalent term)
Lymphoma, immunoblastic (or equivalent term)
Lymphoma, primary, of brain
Mycobacterium avium complex or Mycobacterium kansasii, disseminated or extrapulmonary
Mycobacterium tuberculosis of any site, pulmonary, † disseminated, or extrapulmonary
Mycobacterium, other species or unidentified species, disseminated or extrapulmonary
Pneumocystis jiroveci (previously known as Pneumocystis carinii ) pneumonia
Pneumonia, recurrent †
Progressive multifocal leukoencephalopathy
Salmonella septicemia, recurrent
Toxoplasmosis of brain, onset at age > 1 mo
Wasting syndrome attributed to HIV ‡
* Only among children aged < 6 yr.
†
Only among adults, adolescents, and children aged ≥ 6 yr.
‡ Suggested diagnostic criteria for these illnesses, which might be particularly important for HIV
encephalopathy and HIV wasting syndrome, are described in the following references: Centers for
Disease Control and Prevention: 1994 Revised classification system for human immunodeficiency
virus infection in children less than 13 years of age, MMWR 43(No. RR-12), 1994; Centers for
Disease Control and Prevention: 1993 Revised classification system for HIV infection and
expanded surveillance case definition for AIDS among adolescents and adults, MMWR 41(No.
RR-17), 1992.
From Centers for Disease Control and Prevention: Revised surveillance case definition for HIV
infection—United States, 2014, MMWR 63(No RR-3):1-10, 2014.
Infections
Approximately 20% of AIDS-defining illnesses in children are recurrent
bacterial infections caused primarily by encapsulated organisms such as
Streptococcus pneumoniae and Salmonella as a result of disturbances in humoral
immunity. Other pathogens, including Staphylococcus, Enterococcus,
Pseudomonas aeruginosa, and Haemophilus influenzae, and other Gram-positive
and Gram-negative organisms may also be seen. The most common serious
infections in HIV-infected children are bacteremia, sepsis, and bacterial
pneumonia, accounting for more than 50% of infections in these patients.
Meningitis, urinary tract infections, deep-seated abscesses, and bone/joint
infections occur less frequently. Milder recurrent infections, such as otitis media,
sinusitis, and skin and soft tissue infections, are very common and may be
chronic with atypical presentations.
Opportunistic infections are generally seen in children with severe
2 wk of age. Plasma HIV RNA PCR assays, which detect viral replication, are as
sensitive as the DNA PCR for early diagnosis. Either the DNA or RNA PCR is
considered acceptable for infant testing. The commercially available HIV-1
assays are not designed for quantification of HIV-2 RNA and thus should not be
used to monitor patients with this infection.
Table 302.3
Viral diagnostic testing should be performed within the first 12-24 hr of life,
particularly for high-risk infants (i.e., those of mothers without sustained
virologic suppression, a late cART start, or a diagnosis with acute HIV during
the pregnancy); the tests can identify almost 40% of HIV-infected children. It
seems that many of these children have a more rapid progression of their disease
and deserve more aggressive therapy. Data suggest that if cART treatment starts
at this point, the outcome will be much better. In exposed children with negative
virologic testing at 1-2 days of life, additional testing should be done at 2-3 wk
of age, 4-8 wk of age, and 4-6 mo of age. For higher-risk infants, additional
virologic diagnostic testing should be considered at 2 to 4 wk after cessation of
ARV prophylaxis (i.e., at 8-10 wk of life) (Fig. 302.4 ). A positive virologic
assay (i.e., detection of HIV by PCR) suggests HIV infection and should be
confirmed by a repeat test on a second specimen as soon as possible because
false-positive tests can occur. A confirmed diagnosis of HIV infection can be
made with two positive virologic test results obtained from different blood
samples. HIV infection can be presumptively excluded in nonbreastfed infants
with two or more negative virologic tests (one at age ≥ 14 days and one at age ≥
4 wk) or one negative virologic test (i.e., negative NAT [RNA or DNA]) at age ≥
catalyzes the incorporation of the viral genome into the host's DNA.
Table 302.4
Summary of Antiretroviral Therapies Available in 2019
DRUG (TRADE
NAMES, DOSING SIDE EFFECTS COMMENTS
FORMULATIONS)
NUCLEOSIDE/NUCLEOTIDE REVERSE Class adverse effects:
TRANSCRIPTASE INHIBITORS Lactic acidosis with
hepatic steatosis,
particularly for older
members of the class
Abacavir Children: ≥3 mo Common: nausea, Can be given with food
(Ziagen, ABC): to 13 yr: 8 vomiting, anorexia, Genetic screening for
tablet: 300 mg; oral mg/kg/dose bid fever, headache, HLAB*5701 must be done prior
solution: 20 mg/mL (maximum dose: diarrhea, rash to initiation of ABC-containing
Trizivir: 300 mg bid) Less common: treatment. If test is positive, avoid
combination of >25 kg: 300 mg hypersensitivity, ABC. Do not restart ABC in
zidovudine (ZDV), bid which can be fatal, patients who had hypersensitivity-
lamivudine, ABC Children with Rare: lactic like symptoms (e.g., flu-like
(300, 150, 300 mg) stable CD4 acidosis with symptoms)
Epzicom: counts and hepatic steatosis,
combination of undetectable pancreatitis,
lamivudine, ABC viral load > 6 mo elevated
(300, 600 mg) while taking triglycerides,
Triumeq: ABC can myocardial
combination of transition to 16 infarction
ABC, lamivudine, mg/kg once daily
dolutegravir (600, (max: 600 mg)
300, 50 mg) Adolescents and
adults: 600 mg
once daily
Trizivir (>40 kg):
1 tablet bid
Epzicom (>25
kg): 1 tablet qd
Triumeq: 1 tablet
qd
Didanosine 2 wk to < 3 mo: Common: diarrhea, Food decreases bioavailability by
(Videx, ddl): 50 mg/m2 /dose abdominal pain, up to 50%. Take 30 min before or
powder for oral bid 3-8 mo: 100 nausea, vomiting 2 hr after meal. Tablets dissolved
solution (prepared mg/m2 /dose bid Less common: in water are stable for 1 hr (4 hr in
with solution >8 mo: 120 pancreatitis, buffered solution)
containing antacid): peripheral Drug interactions: antacids/gastric
10 mg/mL mg/m2 /dose neuropathy, acid antagonists may increase
(max: 200
electrolyte bioavailability; possible decreased
mg/dose) bid
abnormalities, absorption of fluoroquinolones,
Adolescents (>13 lactic acidosis with ganciclovir, ketoconazole,
yr) and adults < hepatic steatosis, itraconazole, dapsone, and some
60 kg: 250 mg hepatomegaly, protease inhibitors. Combination
once daily >60 retinal with d4T enhances toxicity; also
kg: 400 mg once depigmentation common if combined with
daily (to increase tenofovir.
adherence) Note: Due to increased side
effects compared with other
NRTIs, ddI is no longer
recommended for treatment of
HIV in children in the US.
Enteric-coated 20-25 kg: 200 Same as for ddl Same as for ddl
didanosine (Videx EC): mg once daily
capsule, delayed 25-60 kg: 250
release: 125, 200, 250, mg once daily
400 mg; generic: 200, ≥60 kg: 400 mg
250, 400 mg once daily
Emtricitabine Infants: 0-3 mo: Common: Patient should be tested for
(Emtriva, FTC): 3 mg/kg once headache, hepatitis B virus (HBV) because
capsule: 200 mg; daily insomnia, diarrhea, HBV exacerbation can occur
oral solution: 10 Children ≥ 3 mo nausea, skin when emtricitabine is
mg/mL to 17 yr, oral discoloration discontinued.
Truvada: solution: 6 Less common: Can be given without regard to
combination of mg/kg (max: 240 lactic acidosis with food. Oral solution should be
FTC, tenofovir mg) once daily hepatic steatosis, refrigerated if temperature above
disoproxil fumarate >33 kg, neutropenia 25°C (77°F)
(TDF) (200, 300 adolescents and COBI is a pharmacokinetc
mg) adults: 200 mg enhancer (boosting agent) used to
Truvada Low capsule or 240 optimize drug levels; it is not
Strength: mg solution once interchangeable with ritonavir. It
combinations of daily can alter renal tubular secretion of
FTC/TDF (100, Truvada, Cr, resulting in elevated Cr with
150 mg); (133, 200 Descovy, Atripla, normal GFR. Note oral solution is
mg); (167, 250 mg) Complera, less bioavailable and has a max
Atripla: Descovy, dose of 240 mg, while the max
combination of Stribild, dose for capsules is 200 mg.
FTC, TDF, Genvoya or
efavirenz (EFV) Biktarvy: adult
(200, 300, 600 mg) dose: 1 tablet
Descovy: once daily
combination of
FTC, tenofovir
disoproxil
alafenamide (TAF)
(200, 25 mg)
Complera:
combination of
FTC, TDF,
rilpivirine (RPV)
(200, 300, 25 mg)
Odefsey:
combination of
FTC, TAF, RPV
(25, 200, 25 mg)
Stribild:
combination of
FTC, TDF,
elvitegravir (EVG),
cobicistat (COBI)
(200, 300, 150, 150
mg)
Genvoya:
combination of
FTC, TAF, EVG,
COBI (200, 10,
150, 150 mg)
Biktarvy:
combination of
bictegravir (BIC),
FTC, TAF (50, 200,
25 mg)
Lamivudine Neonates (≥32 Common: No food restrictions
(Epivir, Epivir wk gestational headache, nausea Patient should be tested for
HBV, 3TC): tablet: age through 4 wk Less common: hepatitis B virus (HBV) because
150 (scored), 300 of age for term pancreatitis, HBV exacerbation can occur
mg (Epivir, infants): 2 peripheral when lamivudine is discontinued.
generic), 100 mg mg/kg/dose bid neuropathy, lactic M184V mutation for this drug
(Epivir HBV); ≥4 wk to <3 mo: acidosis with decreases viral fitness and can be
Solution: 5 mg/mL 4 mg/kg/dose bid hepatic steatosis, advantageous to maintain
(Epivir HBV), 10 ≥3 mo to <3 yr: 5 lipodystrophy including inducing AZT
mg/mL (Epivir) mg/kg/dose bid hypersusceptibility.
Combivir: (max 150 mg)
combination of ≥3 yr: 5
ZDV, lamivudine mg/kg/dose bid
(300, 150 mg) (max 150 mg) or
Trizivir, Epzicom, 10 mg/kg/dose
and Triumeq qd (max 300 mg)
combination (see For ≥14 kg with
abacavir) scored tablet
Symfi Lo (150 mg)
combination of 14 to <20 kg: 75
3TC, TDF, EFV mg bid or 150
(300, 300, 400 mg) mg qd (if >3 yr)
≥20 to <25 kg:
75 mg qAM and
150 mg qPM or
225 mg qd (if >3
yr)
≥25 kg: 150 mg
bid or 300 mg qd
Children should
be switched to
once-daily
dosing of
lamivudine (oral
solution or
tablets) from
twice-daily
dosing at ≥3 yr if
clinically stable
for 36 wk with
an undetectable
viral load and
stable CD4 T
lymphocyte
count
Adolescents and
adults: Combivir
(>30 kg), Trizivir
(>40 kg): 1 tablet
bid
Epzicom (>25
kg): 1 tablet qd
Triumeq (>40
kg): 1 tablet qd
Symfi Lo (>35
kg): 1 tablet qd
Stavudine ≥14 days and < Common: No food restrictions. Should not
(Zerit, d4T): 30 kg: 1 headache, nausea, be administered with ZDV
capsule: 15, 20, 30, mg/kg/dose bid hyperlipidemia, fat because of virologic antagonism.
40 mg; solution: 1 >30 kg: 30 mg maldistribution Higher incidence of lactic
mg/mL bid Less common: acidosis. Increased toxicity if
peripheral combined with ddl.
neuropathy, Note: Due to increased side
pancreatitis, lactic effects compared with other
acidosis, hepatic NRTIs, d4T is no longer
steatosis recommended for treatment of
HIV in children in the US.
Etravirine (ETR, Children <6 yr: Common: nausea, Always administer following a
Intelence): tablet: 25, consult with rash, diarrhea meal for absorption; taking on
100, 200 mg expert Less common: empty stomach decreases
16 to < 20 kg: hypersensitivity absorption by 50%. Tablets can be
100 mg bid reactions dispersed in water
20 to < 25 kg: Inducer of CYP3A4 enzymes and
125 mg bid inhibitor of CYP2C9 and
25 to < 30 kg: CYP2C19, causing multiple
150 mg bid interactions that should be
>30 kg, checked before initiating ETR.
adolescents and Should not be given in
adults: 200 mg combination with TPV, FPV,
bid ATV, or other nonnucleoside
reverse transcriptase inhibitors
Nevirapine High risk Common: skin No food restrictions
(Viramune, NVP): Prophylaxis: rash, headache, Drug interactions: induces hepatic
tablet: 200 mg; 3-dose series for fever, nausea, CYP450A enzymes (including
extended-release high-risk infants abnormal liver CYP3A and CYP2B6) activity
(XR) tablet: 100, >32 wk gestation function tests and decreases protease inhibitor
400 mg; at birth Less common: concentrations (e.g., IND, SQV,
suspension: 10 (including those hepatotoxicity LPV). Should not be given with
mg/mL born to mothers (rarely life- ATV. Reduces ketoconazole
not taking threatening), concentrations (fluconazole
HAART) hypersensitivity should be used as an alternative).
NOTE: DOSES reactions Rifampin decreases nevirapine
ARE A FLAT serum levels. Anticonvulsants and
DOSE, NOT psychotropic drugs using same
PER KG metabolic pathways as NVP
Dosing intervals: should be monitored. Oral
Within 48 hr of contraceptives may also be
birth, 48 hr after affected. XR formulation must be
first dose, 96 hr swallowed whole.
after second dose For children ≤2 yr, some experts
Birth weight 1.5- start with bid dosing without the
2 kg: 8 mg/dose 14 day lead-in of qd dosing.
PO Lead-in dosing decreases
Birth weight >2 occurrence of rash by allowing
kg: 12 mg/dose induction of cytochrome p450
PO metabolizing enzymes.
Treatment
(including
higher risk
prophylaxis
with empiric
therapy):
≥37 wk gestation
at birth:
Birth to age 4
wk: 6
mg/kg/dose bid
THEN
Age >4 wk: 200
mg/m2 /dose bid
34 to <37 wk
gestation at birth:
Birth to age 1
wk: 4
mg/kg/dose bid
Age 1 to 4 wk: 6
mg/kg/dose bid
Age >4 wk: 200
mg/m2 /dose bid
Note dose
adjustment is
optional at 4 wk
for empiric HIV
therapy for high
risk infants with
negative testing
≥1 mo to < 8 yr:
200 mg/m2 once
daily for 14 days;
then same dose
bid (max: 200
mg/dose)
≥8 yr: 120-150
mg/m2 once
daily for 14 days;
then bid (max:
200 mg/dose)
Adolescents and
adults: 200 mg
once daily for 14
days; then 200
mg bid
or
XR 400 mg qd
(after 14 day lead
in)
Rilpivirine Pediatric Headache, insomnia, Given with food only, 500 kcal
(Edurant, RPV): patients: consult rash, depression, mood meal. Do not use with proton
tablet: 25 mg with expert changes pump inhibitors; antacids have to
Complera: Adolescents (>12 be spaced from dose by 2 h before
combination of yr and 35 kg) or 4 h after.
RPV, FTC, TDF and adults: 25 Should not be used if viral load >
(25, 200, 300 mg) mg PO qd 100,000 copies/µL or drugs that
Odefsey: Complera or induce CYP3A or with proton
combination of Odefsey: 1 tablet pump inhibitors
FTC, TAF, RPV qd
(25, 200, 25 mg) Juluca (>18 yr):
Juluca: 1 tablet qd; only
combination of for use in adults
RPV, Dolutegravir with ≥6 mo
(DTG) (25, 50 mg) virologic
suppression with
no resistance to
replace current
regimen
PROTEASE INHIBITORS Class adverse effects: Gi side effects, hyperglycemia,
hyperlipidemia (except atazanavir and darunavir), lipodystrophy,
increased transaminases, increased bleeding disorders in
hemophiliacs. Can induce metabolism of ethinyl estradiol; use
alternate contraception (other than estrogen-containing oral
contraceptives). All of these drugs undergo hepatic metabolism,
mostly by CYP3A4, with many drug interactions. Treatment
note: except in rare instances, always administer with boosting
agent (ritonavir [RTV] or cobicistat [COBI]).
Atazanavir Infants and Common: elevation Administer with food to increase
(Reyataz, ATV): children ≥ 3 mo of indirect absorption. Review drug
powder packet: 50 and ≥ 5 kg: bilirubin; headache, interactions before initiating
mg/packet; capsule: 5 to < 15 kg: arthralgia, because ATV inhibits CYP3A4,
150, 200, 300 mg ATV 200 mg (4 depression, CYP1A2, CYP2C9, and UGT1A1
(Note: capsules and packets) + RTV insomnia, nausea, enzymes. Use with caution with
packets are not 80 mg qd vomiting, diarrhea, cardiac conduction disease or
interchangeable) 15 to < 25 kg: paresthesias liver impairment. Combination
Evotaz: ATV 250 mg (5 Less common: with EFV should not be used in
combination of packets) + RTV prolongation of PR treatment-experienced patients
ATV, COBI (300, 80 mg qd interval on because it decreases ATV levels.
150 mg) Note: Capsules electrocardiogram TDF, antacids, H2 -receptor
are not approved (ECG); rash, rarely antagonists, and proton-pump
for < 6 yr or < 15 Stevens-Johnson inhibitors decrease ATV
kg syndrome, diabetes concentrations. Patients taking
Children ≥6 yr mellitus, buffered ddl should take it at least
and ≥15 kg nephrolithiasis 2 hr before ATV
capsule dosing: COBI is a pharmacokinetc
15 to <35 kg: enhancer (boosting agent) used to
200 mg + RTV optimize drug levels; it is not
100 mg interchangeable with ritonavir. It
≥35 kg: 300 mg can alter renal tubular secretion of
+ RTV 100 mg Cr, resulting in elevated Cr with
Adolescents and normal GFR.
adults:
300 mg + RTV
100 mg
Adults (>18 yr):
Evotaz: 1 tablet
qd
Darunavir <3 yr or < 10 kg: Common: diarrhea, DRV should be given with food.
(Prezista, DRV): do not use nausea, vomiting, Contraindicated for concurrent
tablets: 75, 150, 3 to < 12 yr: abdominal pain, therapy with cisapride, ergot
600, 800 mg; 10 to < 11 kg: fatigue, headache alkaloids, benzodiazepines,
suspension: 100 DRV 200 mg + Less common: skin pimozide, or any major CYP3A4
mg/mL RTV 32 mg bid rashes (including substrates. Use with caution in
Prezcobix: 11 to < 12 kg: Stevens-Johnson patients taking strong CYP3A4
combination DRV, DRV 220 mg + syndrome), lipid inhibitors, or moderate/strong
COBI (800, 150 RTV 32 mg bid and liver enzyme CYP3A4 inducers. Adjust dose
mg) 12 to < 13 kg: elevations, with concurrent rifamycin
DRV 240 mg + hyperglycemia, fat therapy.
RTV 40 mg bid maldistribution Contains sulfa moiety: potential
13 to < 14 kg: for cross-sensitivity with
DRV 260 mg + sulfonamide class
RTV 40 mg bid
14 to < 15 kg:
DRV 280 mg +
RTV 48 mg bid
15 to < 30 kg:
DRB 375 mg +
RTV 48 mg bid
30 to < 40 kg:
DRV 450 mg +
RTV 100 mg bid
≥40 kg: DRV
600 mg + RTV
100 mg bid
Adolescents ≥ 40
kg and adults
with no DRV
mutations: DRV
800 mg + RTV
100 mg qd
Adults (>18 yr)
with no DRV
mutations:
Prezcobix: 1
tablet qd
Adolescents ≥ 40
kg and adults
with DRV
mutation(s):
DRV 600 mg +
RTV 100 mg bid
Fosamprenavir 6 mo to 18 yr: Common: nausea, Should be given with food. FPV is an
(Lexiva, FPV): <11 kg: FPV 45 vomiting, perioral inhibitor of the CYP450 system and
tablet: 700 mg; mg/kg/dose + paresthesias, an inducer, inhibitor, and substrate of
suspension: 50 RTV 7 headache, rash, CYP3A4, which can cause multiple
mg/mL mg/kg/dose bid lipid abnormalities drug interactions. Use with caution in
11 to < 15 kg: Less common: sulfa-allergic individuals.
FPV 30 Stevens-Johnson
mg/kg/dose + syndrome, fat
RTV 3 redistribution,
mg/kg/dose bid neutropenia,
15 to < 20 kg: elevated creatine
FPV 23 kinase,
mg/kg/dose + hyperglycemia,
RTV 3 diabetes mellitus,
mg/kg/dose bid elevated liver
>20 kg: FPV 18 enzymes,
mg/kg/dose angioedema,
(max: 700 mg) + nephrolithiasis
RTV 3
mg/kg/dose
(max: 100 mg)
bid
Adolescents > 18
yr and adults:
FPV 700 mg +
RTV 100 mg bid
or
FPV 1,400 mg +
RTV 200 mg qd
For protease
inhibitor (PI)–
experienced, the
once-daily dose
is not
recommended
Indinavir Not approved for Common: nausea, Reduce dose (600 mg IDV every 8 hr)
(Crixivan, IDV): use in infants or abdominal pain, with mild to moderate liver
capsule: 100, 200, children hyperbilirubinemia, dysfunction. Adequate hydration (at
400 mg Adolescents and headache, least 48 oz fluid/day in adults)
adults: IDV 800 dizziness, lipid necessary to minimize risk of
mg IDV + RTV abnormalities, nephrolithiasis. IDV is cytochrome
(100 mg to 200 nephrolithiasis, P450 3A4 inhibitor and substrate,
mg) bid metallic taste which can cause multiple drug
Less common: fat interactions: rifampin reduces levels;
redistribution, ketoconazole, ritonavir, and other
hyperglycemia, protease inhibitors increase IDV
diabetes mellitus, levels. Do not coadminister with EFV,
hepatitis, acute astemizole, cisapride, terfenadine
hemolytic anemia
Lopinavir/Ritonavir 14 days to 18 yr: Common: diarrhea, Do not administer before
(Kaletra, LPV/r): LPV 300 mg/m2 headache, nausea postmenstrual age of 42 wk and
tablet: 100/25 mg, /dose + RTV 75 and vomiting, lipid postnatal age of 14 days owing
200/50 mg; mg/m2 /dose bid elevation to potential severe toxicities.
solution: 80/20 mg In treatment Less common: fat No food restrictions but has better
per/mL (contains naive children >1 redistribution, GI tolerability when given with or
42% alcohol, 15% yr a dose of 230 hyperglycemia, after a meal.
propylene glycol) diabetes mellitus, Pills must be swallowed whole.
mg/m2 /dose bid pancreatitis, Oral solution should be given
can be used. hepatitis, PR with high-fat meal to increase
Adolescents (>18
interval absorption. Poor palatability of
yr) and adults:
prolongation oral solution is difficult to mask
LPV 400 mg + with flavorings or foods. Once-
RTV 100 mg bid
daily dosing is poorly tolerated in
or
most children, and plasma
800 mg LPV + concentration variability makes
200 mg RTV qd
qd dosing contraindicated in
If taken with children. Interacts with drugs
NVP, EFV, FPV, using CYP3A4, which can cause
or NFV:
multiple drug interactions
LPV 600 mg +
RTV 150 mg bid
Nelfinavir <2 yr: not Common: diarrhea, Administer with a meal to
(Viracept, NFV): recommended asthenia, optimize absorption; avoid acidic
tablet: 250, 625 mg Children 2-13 yr: abdominal pain, food or drink (e.g., orange juice).
45-55 skin rashes, lipid Tablet can be crushed or
mg/kg/dose bid abnormalities dissolved in water to administer
(max: 1,250 Less common: as a solution.
mg/dose) exacerbation of Nelfinavir inhibits CYP3A4
Adolescents and liver disease, fat activity, which may cause
adults: 1,250 mg redistribution, multiple drug interactions.
bid hyperglycemia, Rifampin, phenobarbital, and
diabetes mellitus, carbamazepine reduce levels.
elevation of liver Ketoconazole, ritonavir, indinavir,
enzymes and other protease inhibitors
increase levels. Do not
coadminister astemizole,
cisapride, terfenadine.
NFV is no longer recommended
for treatment for HIV due to
inferior potency compared to
newer agents and unpredictable
pharmacokinetics particularly in
adolescents.
Ritonavir Only use is to Common: nausea, Administration with food
(Norvir, RTV): enhance other PIs; headache, enhances bioavailability and
capsule: 100 mg; dose varies (see vomiting, reduces gastrointestinal
tablet: 100 mg; information for abdominal pain, symptoms. RTV solution should
solution: 80 mg/mL specific PI) diarrhea, taste not be refrigerated (store at 20-
(contains 43% aversion, lipid 25°C)
alcohol) abnormalities, RTV is potent inhibitor of
perioral CYP3A4 and CYP2D6 and
paresthesias inducer of CYP3A4 and CYP1A2
Less common: fat that leads to many drug
redistribution, interactions (e.g., protease
hyperglycemia, inhibitors, antiarrhythmics,
diabetes mellitus, antidepressants, cisapride). Use
pancreatitis, cautiously with inhaled steroids
hepatitis, PR (Cushing syndrome has been
interval reported)
prolongation,
allergic reactions
Saquinavir Infants and Common: diarrhea, Administer with a high-fat meal to
(Invirase, SQV): children < 16 yr: abdominal pain, enhance bioavailability. SQV is
capsule: 200 mg; not approved for headache, nausea, metabolized by CYP3A4, which may
tablet: 500 mg use skin rashes, lipid cause many drug interactions:
Adolescents and abnormalities rifampin, phenobarbital, and
adults: SQV Less common: carbamazepine decrease serum levels.
1,000 mg + RTV exacerbation of Saquinavir may decrease metabolism
100 mg bid chronic liver of calcium channel antagonists, azoles
disease, diabetes (e.g., ketoconazole), macrolides.
mellitus, Pretherapy EKG recommended and
pancreatitis, contraindicated in patients with
elevated liver prolonged QT interval.
transaminases, fat
maldistribution,
increase in both QT
and PR in ECG
Tipranavir <2 yr: not Common: diarrhea, No food restrictions. Better tolerated
(Aptivus, TPV): approved nausea, vomiting, with meal. Can inhibit human platelet
capsule: 250 mg; 2-18 yr fatigue, headache, aggregation: use with caution in
solution: 100 (treatment- skin rashes, patients at risk for increased bleeding
mg/mL (contains experienced elevated liver (trauma, surgery, etc.) or in patients
116 IU vitamin only): TPV 375 enzymes, lipid receiving concurrent medications that
E/mL) mg/m2 /dose + abnormalities may increase the risk of bleeding.
RTV 150 mg/m2 Less common: fat TPV is metabolized by CYP3A4,
(max: TPV 500 redistribution, which may cause many drug
mg + RTV 200 hepatitis, interactions. Contraindicated in
mg) bid hyperglycemia, patients with hepatic insufficiency or
or diabetes mellitus, in those receiving concurrent therapy
TPV 14 intracranial with amiodarone, cisapride, ergot
mg/kg/dose + hemorrhage alkaloids, benzodiazepines, pimozide.
RTV 6 TPV contains sulfonamide moiety, and
mg/kg/dose caution should be taken in patients
(max: same) bid with sulfonamide allergy
Adolescents (>18
yr) and adult:
TPV 500 mg +
RTV 200 mg bid
FUSION INHIBITORS
Enfuvirtide <6 yr: not Common: Local Must be given subcutaneously.
(Fuzeon, ENF): approved injection site Severity of reactions increased if
injection: Children ≥6 yr to reactions in 98% given intramuscularly. Apply ice after
lyophilized powder 16 yr: 2 (e.g., erythema, injection and massage the area to
of 108 mg mg/kg/dose SQ induration nodules, reduce local reactions. Injection sites
reconstituted in 1.1 (max: 90 mg) bid cysts, ecchymoses) should be rotated; recommended sites
mL of sterile water Adolescents (>16 Less common: are upper arm, anterior thigh, or
delivers 90 mg/mL yr) and adults: 90 increased incidence abdomen.
mg SQ bid of bacterial
pneumonia,
hypersensitivity,
fever, nausea,
vomiting, chills,
elevated liver
enzymes,
hypotension,
immune-mediated
reactions (e.g.,
glomerulonephritis,
Guillain-Barré
syndrome,
respiratory distress)
ENTRY INHIBITORS
Maraviroc Neonates/infants: Common: fever, Testing for CCR5-tropic virus
(Selzentry, MVC): not approved upper respiratory required; virus must not have
oral solution: 20 ≥2 yr and ≥ 10 infection–like mixed tropism (i.e.,
mg/mL; tablet: 25, kg: symptoms, rash, CCR5/CXC4) to have efficacy.
75, 150, 300 mg Given with abdominal pain, No food restrictions. MVC is a
CYP3A musculoskeletal CYP3A4 and P-glycoprotein
inhibitors (EVG, symptoms, (Pgp) substrate, which may cause
RTV, PIs except dizziness many drug interactions.
TPV/r): Less common: Tropism assay to exclude the
10 to < 20 kg: 50 cardiovascular presence of CXCR4 HIV is
mg bid abnormalities, required before using MVC.
20 to < 30 kg: 75 cholestatic Caution should be used when
mg bid jaundice, given to patients with hepatic
30 to < 40 kg: rhabdomyolysis, impairment or cardiac disease or
100 mg bid myositis, receiving CYP3A4 or P-
>40 kg: 150 mg osteonecrosis glycoprotein-modulating drugs.
bid
Given with
NRTIs, T-20,
TPV/r, NVP,
RAL, or other
drugs not
affecting
CYP3A:
10 to < 30 kg:
not
recommended
30 to < 40 kg:
300 mg bid
>40 kg: 300 mg
bid
Given with EFV,
ETR: not
recommended
Adolescents > 16
yr and adults:
150 mg bid if
given with potent
CYP3A inhibitor
(e.g., protease
inhibitor except
TPV)
300 mg bid if
given with not
potent CYP3A4
inhibitors (e.g.,
NRTI, TPV,
NVP, ENF, RAL)
600 mg bid if
given with potent
CYP3A4 inducer
(e.g., EFV, ETR,
rifampin,
phenobarbital)
Antiretroviral drugs often have significant drug–drug interactions, with each other and with other
classes of medicines, which should be reviewed before initiating any new medication.
The information in this table is not all-inclusive. Updated and additional information on dosages,
drug–drug interactions, and toxicities is available on the AIDSinfo website at
http://www.aidsinfo.nih.gov .
Modified from the Guidelines for use of antiretroviral agents in pediatric HIV infection.
http://aidsinfo.nih.gov/contentfiles/pediatricguidelines.pdf .
By targeting different points in the viral life cycle and stages of cell activation
and by delivering drug to all tissue sites, maximal viral suppression is feasible.
Combinations of three drugs consisting of a two-NRTI backbone of (1) a
thymidine analog NRTI (abacavir or zidovudine) or tenofovir and (2) a
nonthymidine analog NRTI (lamivudine or emtricitabine) to suppress replication
in both active and resting cells added to (3) a ritonavir-boosted PI
(lopinavir/ritonavir, atazanavir, or darunavir), an NNRTI (efavirenz or
nevirapine), or an INSTI (raltegravir or dolutegravir) can produce prolonged
suppression of the virus. The use of three drugs from three different classes
generally should be avoided but may be necessary in children with highly
resistant viruses; the drugs in these regimens should only be chosen by an HIV
specialist with pharmacist input. Combination treatment increases the rate of
toxicities (see Table 302.4 ), and complex drug–drug interactions occur among
many of the antiretroviral drugs. Many PIs are inducers or inhibitors of the
cytochrome P450 system and are therefore likely to have serious interactions
with multiple drug classes, including nonsedating antihistamines and
psychotropic, vasoconstrictor, antimycobacterial, cardiovascular, anesthetic,
analgesic, and gastrointestinal drugs (cisapride). Whenever new medications are
added to an antiretroviral treatment regimen, especially a protease inhibitor or
cobicistat containing regimen, a pharmacist and/or HIV specialist should be
consulted to address possible drug interactions. The inhibitory effect of ritonavir
(a PI) on the cytochrome P450 system has been exploited, and small doses of the
drug are added to several other protease inhibitors (e.g., lopinavir, atazanavir,
darunavir) to slow their metabolism by the P450 system and to improve their
pharmacokinetic profile. This strategy provides more effective drug levels with
Table 302.5
Recommendations for PJP Prophylaxis and CD4 Monitoring for HIV-Exposed Infants
and HIV-Infected Children, by Age and HIV Infection Status
AGE/HIV INFECTION PJP PROPHYLAXIS CD4 MONITORING
STATUS
Birth to 4-6 wk, HIV-exposed No prophylaxis None
HIV infection reasonably No prophylaxis None
excluded*
<1 yr, HIV-infected or HIV- Prophylaxis regardless of CD4 According to local practice for initiation or
indeterminate count or percentage follow-up of cART
1-5 yr, HIV-infected Prophylaxis if CD4 < 500 cells/µL According to local practice for initiation or
or < 15% † follow-up of cART
>6 yr, HIV-infected Prophylaxis if CD4 < 200 cells/µL According to local practice for initiation or
or < 15% † ‡ follow-up of cART
* See text.
† More frequent monitoring (e.g., monthly) is recommended for children whose CD4 counts or
percentages are approaching the threshold at which prophylaxis is recommended.
‡ Prophylaxis should be considered on a case-by-case basis for children who might otherwise be
at risk for PJP, such as children with rapidly declining CD4 counts or percentages or children with
category C conditions. Children who have had PJP should receive PJP prophylaxis until their CD4
count is ≥200 cells/mm3 for patients aged ≥6 yr, CD4 percentage is ≥15% or CD4 count is ≥500
cells/mm3 for patients aged 1 to <6 yr for >3 consecutive mo after receiving cART for ≥6 mo.
The National Perinatal HIV Hotline (1-888-448-8765) provides consultation on all aspects of
perinatal HIV care.
cART, combined antiretroviral therapy; PJP, Pneumocystis jiroveci pneumonia.
Table 302.6
Prophylaxis to Prevent First Episode of Opportunistic
Infections Among HIV-Exposed and HIV-Infected Infants
and Children, United States*
PREVENTIVE REGIMEN
PATHOGEN
INDICATION FIRST CHOICE ALTERNATIVE
STRONGLY RECOMMENDED AS STANDARD OF CARE
Pneumocystis HIV-infected or HIV-indeterminate TMP-SMX , 150/750 Dapsone: age ≥
pneumonia † infants aged 1-12 mo; HIV-infected mg/m2 body surface area 1 mo: 2 mg/kg
children aged 1-5 yr with CD4 count per day or 5-10 mg/kg/day (max: 100 mg)
of < 500 cells/µL or CD4 percentage (TMP)/25-50 mg/kg/day orally qd; or 4
of < 15%; HIV-infected children (SMX) (max: 320/1,600 mg/kg (max: 200
aged 6-12 yr with CD4 count of < mg) orally qd or bid 3 times mg) orally once
200 cells/µL or CD4 percentage of < weekly on consecutive days a week
15%; >13 yr with CD4 count <200 or Atovaquone:
<15% or age 1-3 mo and
qd or bid orally 3 times > 24 mo-12 yr:
weekly on alternate days 30-40 mg/kg
orally qd with
food; age 4-24
mo: 45 mg/kg
orally qd with
food; ≥ 13 yr
1500 mg orally
qd
Aerosolized
pentamidine:
age ≥ 5 yr: 300
mg once a
month by
Respirgard II
(Marquest,
Englewood, CO)
nebulizer
Malaria Living or traveling to area in which Same for HIV-infected and Doxycycline,
malaria is endemic HIV-uninfected children. 2.2 mg/kg body
Refer to weight
http://www.cdc.gov/malaria/ (maximum 100
for the most recent mg) orally qd for
recommendations. children >8 yr
Mefloquine , 5 mg/kg Chloroquine, 5
orally 1 time weekly (max: mg/kg base
250 mg) (equal 7.5 mg/kg
Atovaquone/proguanil chloroquine
(Malarone) qd phosphate)
11-20 kg: 62.5 mg/25 mg (1 orally up to 300
pediatric tablet) mg weekly (only
21-30 kg: 2 pediatric tablets for regions
31-40 kg: 3 pediatric tablets where the
>40 kg: 1 adult tablet parasite is
(250 mg/100 mg) sensitive)
Mycobacterium
tuberculosis
Isoniazid- TST reaction ≥ 5 mm Isoniazid, 10-15 mg/kg Rifampin, 10-20
sensitive or body weight (max: 300 mg) mg/kg body weight
Prior positive TST result without qd for 9 mo (max: 600 mg) orally
treatment or daily for 4-6 mo
or 20-30 mg/kg body weight
Close contact with any person (max: 900 mg) orally 2
who has contagious TB. TB times weekly for 9 mo;
disease must be excluded before DOT highly recommended
start of treatment
Isoniazid- Same as previous pathogen; Rifampin, 10-20 mg/kg body Consult TB expert
resistant increased probability of exposure to weight (max: 600 mg) orally
isoniazid-resistant TB daily for 4-6 mo
Multidrug- Same as previous pathogen; Choice of drugs requires
resistant increased probability of exposure to consultation with public health
(isoniazid and multidrug-resistant TB authorities and depends on
rifampin) susceptibility of isolate from
source patient
Mycobacterium For children age ≥ 6 yr with CD4 Clarithromycin, 7.5 mg/kg Azithromycin, 5
avium complex ‡ count of < 50 cells/µL; age 2-5 yr (max: 500 mg) orally bid mg/kg body
with CD4 count of < 75 cells/µL; age or weight (max:
1-2 yr with CD4 count of < 500 cells/ Azithromycin, 20 mg/kg 250 mg) orally
µL; age < 1 yr with CD4 count of < (max: 1,200 mg) orally once qd
750 cells/µL a week or
Children age ≥ 5
yr
Rifabutin, 300
mg orally qd
Varicella-zoster Exposure to varicella or shingles Varicella-zoster immunoglobulin If VariZIG is not
virus § with no history of varicella (VariZIG), 125 IU/10 kg (max: available and <
or 625 IU) IM, administered ideally 96 hr from
Zoster or seronegative status for within 96 hr after exposure; exposure,
VZV potential benefit up to 10 days acyclovir 20
or after exposure mg/kg (max: 800
Lack of evidence for age- mg) 4 times a
appropriate vaccination day for 5-7 days
or
IVIG, 400
mg/kg,
administered
once
Vaccine- Standard recommendations for HIV- Routine vaccinations (see Fig.
preventable exposed and HIV-infected children 302.5 )
pathogens
USUALLY RECOMMENDED
Toxoplasma Seropositive IgG to Toxoplasma and TMP-SMX, 150/750 mg/m2 Dapsone, age ≥
gondii ¶ severe immunosuppression: age < 6 orally qd or divided bid 1 mo: 2 mg/kg
yr with CD4 percentage < 15%; age or or 15 mg/m2
≥ 6 yr with CD4 count < 100 cells/µL Same dosage qd 3 times (max: 25 mg)
weekly on consecutive days orally qd
or plus
bid 3 times weekly on Pyrimethamine,
alternate days 1 mg/kg (max:
25 mg) orally qd
plus
Leucovorin, 5
mg orally every
3d
Invasive Hypogammaglobulinemia (i.e., IgG < IVIG 400 mg/kg body weight
bacterial 400 mg/dL) every 2-4 wk
infections
Cytomegalovirus CMV antibody positivity and Valganciclovir, 900 mg orally
severe immunosuppression (CD4 qd with food for older children
count < 50 cells/µL for >6 yr; who can receive adult dosing
CD4 percentage <5% for ≤6 yr)
For children aged 4 mo–16 yr,
valganciclovir oral solution 50
mg/mL at dose in milligrams = 7
× BSA × CrCl (up to maximum
CrCl of 150 mL/min/1.73 m2 )
orally qd with food (maximum
dose 900 mg/day)
* Information in these guidelines might not represent FDA approval or FDA-approved labeling for
products or indications. Specifically, the terms safe and effective might not be synonymous with
the FDA-defined legal standards for product approval.
† Daily trimethoprim-sulfamethoxazole (TMP-SMX) reduces the frequency of certain bacterial
infections. Compared with weekly dapsone, daily dapsone is associated with a lower incidence of
PCP but higher hematologic toxicity and mortality rates. Patients receiving therapy for
toxoplasmosis with sulfadiazine-pyrimethamine are protected against PCP and do not need TMP-
SMX. TMP-SMX, dapsone-pyrimethamine, and possibly atovaquone (with or without
pyrimethamine), protect against toxoplasmosis; however, data have not been prospectively
collected.
‡
Substantial drug interactions can occur between rifamycins (i.e., rifampin and rifabutin) and
protease inhibitors and nonnucleoside reverse transcriptase inhibitors. A specialist should be
consulted.
§ Children routinely being administered intravenous immunoglobulin (IVIG) should receive VariZIG
if the last dose of IVIG was administered more than 21 days before exposure.
¶ Protection against toxoplasmosis is provided by the preferred anti-Pneumocystis regimens and
possibly by atovaquone.
CMV, cytomegalovirus; FDA, U.S. Food and Drug Administration; HIV, human immunodeficiency
virus; IgG, immunoglobulin G; IM, intramuscularly; IVIG, intravenous immunoglobulin; PCP,
Pneumocystis pneumonia; TB, tuberculosis; TMP-SMX, trimethoprim-sulfamethoxazole; TST,
tuberculin skin test; VZV, varicella-zoster virus.
From Centers for Disease Control and Prevention (CDC): Guidelines for the prevention and
treatment of opportunistic infections among HIV-exposed and HIV-infected children, MMWR
Recomm Rep 58(RR-11):127-128, 2009, Table 1.
Transmissible Spongiform
Encephalopathies
David M. Asher
Table 304.1
Clinical and Epidemiologic Features of Human
Transmissible Spongiform Encephalopathies (Prion
Diseases)
Table 304.2
Iatrogenic Transmission of Creutzfeldt-Jakob Disease by
Products of Human Origin
INCUBATION TIME
PRODUCT NO. OF PATIENTS
Mean Range
Cornea 3 17 mo 16-18 mo
Dura mater allograft >100 7.4 yr 1.3-16 yr
Pituitary extract
Growth hormone >100* 12 yr 5-38.5 yr
Gonadotropin 4 13 yr 12-16 yr
Red blood cells 4 ? 6 yr 6.3-8.5 yr †
Plasma-derived coagulation factor VIII 1 ? > 11 yr ‡
* There have been 28 cases reported among approximately 8,000 recipients of human cadaveric
growth hormone in the United States; the remaining cases have been reported in other countries.
† The second transfusion-transmitted case of vCJD (Peden AH, Head MW, Ritchie DL, et al:
Preclinical vCJD after blood transfusion in a PRNP codon 129 heterozygous patient, Lancet
364:527-529, 2004) died of unrelated causes about 5 yr after transfusion but was found to have
accumulations of abnormal PrP in the spleen and cervical lymph node—a finding unique to vCJD
and interpreted as probable preclinical infection.
‡ The diagnosis of vCJD infection attributed to treatment with human plasma–derived coagulation
Clinical Manifestations
Kuru, no longer seen, is a progressive degenerative disease of the cerebellum
and brainstem with less obvious involvement of the cerebral cortex. The first
sign of kuru was usually cerebellar ataxia followed by progressive
incoordination. Coarse, shivering tremors were characteristic. Variable
abnormalities in cranial nerve function appeared, frequently with impairment in
conjugate gaze and swallowing. Patients died of inanition and pneumonia or of
burns from cooking fires, usually within 1 yr after onset. Although changes in
mentation were common, there was no frank dementia or progression to coma,
as in CJD. There were no signs of acute encephalitis, such as fever, headaches,
and convulsions.
CJD occurs throughout the world. Patients initially have either sensory
disturbances (most often visual) or confusion and inappropriate behavior,
progressing over weeks or months to frank dementia, akinetic mutism, and,
ultimately, coma. Some patients have cerebellar ataxia early in the disease, and
most patients experience myoclonic jerking movements. The mean survival time
of patients with sCJD has been < 1 yr from the earliest signs of illness, although
approximately 10% live for 2 yr. Variant CJD (Table 304.3 ) differs from the
more common sCJD; patients with vCJD are much younger at onset (as young as
12 yr) and more often present with complaints of dysesthesia and subtle
behavioral changes, often mistaken for psychiatric illness. Severe mental
deterioration occurs later in the course of vCJD. Patients with vCJD have
survived substantially longer than those with sCJD. Attempts have been made to
subclassify cases of CJD based on the electrophoretic differences in PrPTSE and
variation in its sensitivity to digestion with the proteolytic enzyme proteinase
(PK); the different variants are said to have somewhat different clinical features,
including the duration of illness, though all are ultimately fatal.
Table 304.3
Clinical and Histopathologic Features of Patients With Variant and Typical Sporadic
Creutzfeldt-Jakob Disease
SPORADIC CJD
FEATURE VARIANT CJD (FIRST 10 PATIENTS)
(185 PATIENTS)
Years of age at 29 (19-74) 65
death* (range)
Duration of 12 (8-23) 4
illness, mo
(range)
Presenting Abnormal behavior, dysesthesia Dementia
signs
Later signs Dementia, ataxia, myoclonus Ataxia, myoclonus
Periodic Rare Most
complexes on
EEG
PRNP 129 All tested (except one transfusion-transmitted case, one plasma- 83%
Met/Met derivative transmitted case; one possible clinical case in United
Kingdom where no tissue was available to confirm)
Histopathologic Vacuolation, neuronal loss, astrocytosis, plaques (100%) Vacuolation, neuronal
changes loss, astrocytosis,
plaques (≤15%)
Florid PrP 100% 0
plaques †
PrPTSE BSE-like ‡ Not BSE-like
glycosylation
pattern
* Median age and duration for variant CJD; averages for typical sporadic CJD.
nonglycosylated band glycoform of PrP-res (Collinge J, Sidle KC, Meads J, et al: Molecular
analysis of prion strain variation and the aetiology of “new variant” CJD, Nature 383:685-690,
1996).
BSE, bovine spongiform encephalopathy; CJD, Creutzfeldt-Jakob disease; EEG,
electroencephalogram; Met, codon 129 of one PRNP gene encoding for methionine; PRNP , prion
protein–encoding gene; PrP, prion protein.
Modified from Will RG, Ironside JW, Zeidler M, et al: A new variant of Creutzfeldt-Jakob disease in
the UK, Lancet 347:921-925, 1996.
GSS is a familial disease resembling CJD but with more prominent cerebellar
ataxia and amyloid plaques. Dementia may appear only late in the course, and
the average duration of illness is longer than typical sCJD. Progressively severe
insomnia and dysautonomia, as well as ataxia, myoclonus, and other signs
resembling those of CJD and GSS, characterize FFI and sporadic fatal insomnia.
A case of sporadic fatal insomnia has been described in a young adolescent.
GSS has not been diagnosed in children or adolescents.
A novel prion disease has been reported that is expressed in several
generations with an autosomal dominant pattern associated with a unique
mutation in the PRNP gene. The affected persons were middle-aged with a
history of chronic diarrhea for years plus autonomic neuropathy and modest
mental impairment but without full-blown dementia; PK-resistant PrP deposits
with amyloid properties occurred in the brain, lymphoid tissues, kidney, spleen,
and intestinal tract. The disease was not successfully transmitted to three lines of
CHAPTER 305
Parasites are divided into three main groups taxonomically: protozoans, which
are unicellular, and helminths and ectoparasites, which are multicellular.
Chemotherapeutic agents appropriate for one group may not be appropriate for
the others, and not all drugs are readily available (Table 305.1 ). Some drugs are
not available in the United States, and some are available only from the
manufacturer, specialized compounding pharmacies, or the Centers for Disease
Control and Prevention (CDC). Information on the availability of drugs and
expert guidance in management can be obtained by contacting the CDC Parasitic
Diseases Branch (1-404-718-4745; e-mail [email protected] (M-F, 8 AM -4 PM ,
Eastern time). For assistance in the management of malaria, healthcare providers
should call the CDC Malaria Hotline: 1-770-488-7788 or 1-855-856-4713 toll-
free (M-F, 9 AM -5 PM , Eastern time). For all emergency consultations after
hours, clinicians can contact the CDC Emergency Operations Center at 1-770-
488-7100 and request to speak with a CDC Malaria Branch clinician or on-call
parasitic diseases physician. Some antiparasitic drugs are not licensed for use in
the United States but can be obtained as investigational new drugs (INDs) from
the CDC; providers should call the CDC Drug Service, Division of Scientific
Resources and Division of Global Migration and Quarantine, at 1-404-639-3670.
Table 305.1
Drugs for Parasitic Infections
Parasitic infections are found throughout the world. With increasing travel, immigration, use of
immunosuppressive drugs, and the spread of HIV, physicians anywhere may see infections caused by previously
unfamiliar parasites. The table below lists first-choice and alternative drugs for most parasitic infections.
INFECTION DRUG ADULT DOSAGE PEDIATRIC DOSAGE
Acanthamoeba keratitis
Drug of choice: See footnote 1
Amebiasis (Entamoeba histolytica)
Asymptomatic infection
Drug of choice: Iodoquinol (Yodoxin) 2 650 mg PO tid × 20 days 30-40 mg/kg/day (max
1950 mg) in 3 doses PO ×
20 days
or Paromomycin 25-35 mg/kg/day PO in 3 doses × 25-35 mg/kg/day PO in 3
7 days doses × 7 days
Alternative: Diloxanide furoate 3 500 mg tid PO × 10 days 20 mg/kg/day PO in 3
doses × 10 days
Mild to moderate intestinal disease
Drug of choice: Metronidazole 500-750 mg tid PO × 7-10 days 35-50 mg/kg/day PO in 3
doses × 7-10 days
or Tinidazole 4 2 g PO once daily × 3 days 50 mg/kg/day PO (max 2 g)
in 1 dose × 3 days
Either followed by: Iodoquinol 2 650 mg PO tid × 20 days 30-40 mg/kg/day PO in 3
doses × 20 days (max 2 g)
or Paromomycin 25-35 mg/kg/day PO in 3 doses × 25-35 mg/kg/day PO in 3
7 days doses × 7 days
Alternative: Nitazoxanide 5 500 mg bid × 3 days 1-3 yr: 100 mg bid × 3 days
4-11 yr: 100 mg bid × 3
days
12+ yr: use adult dosing
Severe intestinal and extraintestinal disease
Drug of choice: Metronidazole 750 mg PO tid × 7-10 days 35-50 mg/kg/day PO in 3
doses × 7-10 days
or Tinidazole 4 2 g PO once daily × 5 days 50 mg/kg/day PO (max 2 g)
× 5 days
Either followed by: Iodoquinol 2 650 mg PO tid × 20 days 30-40 mg/kg/day PO in 3
doses × 20 days (max 2 g)
or Paromomycin 25-35 mg/kg/day PO in 3 doses × 25-35 mg/kg/day PO in 3
7 days doses × 7 days
Amebic meningoencephalitis, primary and granulomatous
Naegleria fowleri
Drug of choice: Amphotericin B 1.5 mg/kg/day IV in 2 divided 1.5 mg/kg/day IV in 2
deoxycholate 6 , 7 doses × 3 days, then 1 mg/kg divided doses × 3 days,
daily IV × 11 days then 1 mg/kg daily IV × 11
days
plus Amphotericin B 1.5 mg/kg intrathecally daily × 2 1.5 mg/kg intrathecally
deoxycholate 6 , 7 days, then 1 mg/kg intrathecally daily × 2 days, then 1
every other day × 8 days mg/kg intrathecally every
other day × 8 days
plus Rifampin 7 10 mg/kg (max 600 mg) IV or PO 10 mg/kg (max 600 mg) IV
daily × 28 days or PO daily × 28 days
plus Fluconazole 7 10 mg/kg (max 600 mg) IV or PO 10 mg/kg (max 600 mg) IV
daily × 28 days or PO daily × 28 days
plus Azithromycin 7 500 mg IV or PO daily × 28 days 10 mg/kg (max 500 mg) IV
or PO daily × 28 days
plus Miltefosine 6 , 7 , 8 50 mg PO tid × 28 days <45 kg: 50 mg bid (max 2.5
mg/kg) × 28 days
≥45 kg: use adult dosing
plus Dexamethasone 0.6 mg/kg/day IV in 4 divided 0.6 mg/kg/day IV in 4
doses × 4 days divided doses × 4 days
Acanthamoeba
Drug of choice: See footnotes 7 , 8
Balamuthia mandrillaris
Drug of choice: See footnotes 7 , 9a , 9b
Sappinia diploidea
Drug of choice: See footnote 10
Ancylostoma caninum (eosinophilic enterocolitis)
Drug of choice: Albendazole 7 400 mg PO once <10 kg/2 yr: 11
≥2 yr: see adult dosing
or Mebendazole 100 mg PO bid × 3 days 100 mg PO bid × 3 days 12
or Pyrantel pamoate (OTC) 7 11 mg/kg PO (max 1 g) × 3 days 11 mg/kg PO (max 1 g) × 3
days
or Endoscopic removal
Ancylostoma duodenale, see Hookworm
Angiostrongyliasis (Angiostrongylus cantonensis, Angiostrongylus costaricensis)
Drug of choice: See footnote 13
Anisakiasis (Anisakis spp.)
Treatment of Surgical or endoscopic removal
choice:
Alternative: Albendazole 7 , 14 400 mg PO bid × 6-21 days <10 kg/2 yr: 11
≥2 yr: see adult dosing
Ascariasis (Ascaris lumbricoides, roundworm)
Drug of choice: Albendazole 7 400 mg PO once <10 kg/2 yr: see adult
dosing 11
≥2 yr: see adult dosing
or Mebendazole 100 mg PO bid × 3 days or 500 100 mg PO bid × 3 days or
mg PO once 500 mg PO once 12
or Ivermectin 7 150-200 µg/kg PO once <15 kg: not indicated
≥15 kg: see adult dosing
Babesiosis (Babesia microti)
Drugs of choice: 15 Atovaquone 7 750 mg PO bid × 7-10 days 20 mg/kg (max 750 mg) PO
bid × 7-10 days
plus Azithromycin 7 500-1000 mg once, then 250 mg 10 mg/kg PO on day 1
daily × 7-10 days. Higher doses (max 500 mg/dose), then 5
(600-1000 mg) and/or prolonged mg/kg/day (max 250
therapy (6 wk or longer) may be mg/dose) PO on days 2-10
required for immunocompromised
patients.
or Clindamycin 7 300-600 mg IV qid or 600 mg tid 20-40 mg/kg/day IV or PO
PO × 7-10 days in 3 or 4 doses × 7-10 days
(max 600 mg/dose)
plus Quinine 7 648 mg tid PO × 7-10 days 10 mg/kg (max 648 mg) PO
tid × 7-10 days
Balamuthia mandrillaris, see Amebic meningoencephalitis, primary
Balantidiasis (Balantidium coli)
Drug of choice: Tetracycline 7 , 16 500 mg PO qid × 10 days <8 yr: not indicated
≥8 yr: 10 mg/kg (max 500
mg) PO qid × 10 days
Alternative: Metronidazole 7 750 mg PO tid × 5 days 35-50 mg/kg/day PO in 3
divided doses × 5 days
or Iodoquinol 2 , 7 650 mg PO tid × 20 days 30-40 mg/kg/day (max 2 g)
PO in 3 divided doses × 20
days
or 500 mg PO bid × 3 days 1-3 yr: 100 mg PO bid × 3
Nitazoxanide 4 , 7 days
biguanide (PHMB) and 0.02% chlorhexidine have been successfully used individually and in
combination in a large number of patients (Tabin G, et al: Cornea 20:757, 2001; Wysenbeek YS,
et al: Cornea 19:464, 2000). The expected treatment course is 6-12 mo. PHMB is no longer
available from Leiter's Park Avenue Pharmacy but is available from the O'Brien Pharmacy (1-800-
627-4360; distributes in many states) and the Greenpark Pharmacy (1-713-432-9855; Texas
only). Combinations with either 0.1% propamidine isethionate (Brolene) or hexamidine
(Desmodine) have been used (Seal DV: Eye 17:893, 2003) successfully, but these are not
available in the United States. Neomycin is no longer recommended due to high levels of
resistance (Acanthamoeba keratitis: Treatment guidelines from The Medical Letter 143, 8/1/2013).
In addition, the combination of chlorhexidine, natamycin (pimaricin), and debridement also has
been successful (Kitagawa K, et al: Jpn J Ophthalmol 47:616, 2003).
2 The drug is not available commercially but can be compounded by Expert Compounding
Pharmacy, 6744 Balboa Blvd, Lake Balboa, CA 91406 (1-800-247-9767 or 1-818-988-7979 or
[email protected] ).
3 The drug is not available commercially in the United States.
4 A nitroimidazole similar to metronidazole, tinidazole was approved by the FDA in 2004 and
appears to be as effective and better tolerated than metronidazole. It should be taken with food to
minimize GI adverse effects. For children and patients unable to take tablets, a pharmacist may
crush the tablets and mix them with cherry syrup (Humco, and others). The syrup suspension is
good for 7 days at room temperature and must be shaken before use. Ornidazole, a similar drug,
is also used outside the United States.
5 Nitazoxanide is FDA approved as a pediatric oral suspension for treatment of Cryptosporidium in
should be followed by treatment with a luminal agent, such as paromomycin
(which is preferred) or iodoquinol. Diloxanide furoate can also be used in
children > 2 yr of age, but it is no longer available in the United States.
Paromomycin should not be given concurrently with metronidazole or
tinidazole, because diarrhea is a common side effect of paromomycin and may
confuse the clinical picture. Asymptomatic intestinal infection with E. histolytica
should be treated, preferably with paromomycin or alternatively with either
iodoquinol or diloxanide furoate. For fulminant cases of amebic colitis, some
experts suggest adding dehydroemetine (1 mg/kg/ day subcutaneously or
intramuscularly, never intravenously), available only through the Centers for
Disease Control and Prevention. Patients should be hospitalized for monitoring
if dehydroemetine is administered. Dehydroemetine should be discontinued if
tachycardia, T-wave depression, arrhythmia, or proteinuria develops.
Table 307.1
Drug Treatment for Amebiasis
Table 308.1
Diagnosis
Giardiasis should be considered in children who have acute nondysenteric
diarrhea, persistent diarrhea, intermittent diarrhea and constipation,
malabsorption, chronic crampy abdominal pain and bloating, failure to thrive, or
weight loss. It should be particularly high in the differential diagnosis of children
in child care, children in contact with an index case, children with a history of
recent travel to an endemic area, and children with humoral immunodeficiencies.
Testing for giardiasis should be standard for internationally adopted children
Treatment
Children with acute diarrhea in whom Giardia organisms are identified should
receive therapy. In addition, children who manifest failure to thrive or exhibit
malabsorption or gastrointestinal tract symptoms such as chronic diarrhea should
be treated.
Asymptomatic excreters generally are not treated, except in specific instances
such as outbreak control, prevention of household transmission by toddlers to
pregnant women and patients with hypogammaglobulinemia or cystic fibrosis,
and situations requiring oral antibiotic treatment where Giardia may produce
malabsorption of the antibiotic.
The FDA has approved tinidazole and nitazoxanide for the treatment of
Giardia in the United States. Both medications have been used to treat Giardia
in thousands of patients in other countries and have excellent safety and efficacy
records against Giardia (Table 308.2 ). Tinidazole has the advantage of single-
dose treatment and very high efficacy (>90%), while nitazoxanide has the
advantage of a suspension form, high efficacy (80–90%), and very few adverse
effects. Metronidazole, though never approved by the FDA for treatment of
Giardia, is also highly effective (80–90% cure rate), and the generic form is
considerably less expensive than tinidazole or nitazoxanide. Frequent adverse
effects are seen with metronidazole therapy, and it requires 3-times-a-day dosing
for 5-7 days. Suspension forms of tinidazole and metronidazole must be
compounded by a pharmacy; neither drug is sold in suspension form.
Table 308.2
Drug Treatment for Giardiasis
Leishmaniasis (Leishmania)
Peter C. Melby
Table 311.1
Clinical and Epidemiologic Characteristics of Main
Leishmania spp.
MAIN
CLINICAL NATURAL MAIN
SUBGENUS CLINICAL RISK GROUPS
FORM PROGRESSION RESERVOIR
FEATURES
Leishmania Leishmania CL, DCL, Ulcerating Not well No well-defined risk Possums,
amazonensis DsCL lesions, single described groups rodents
† or multiple
Leishmania Viannia CL, MCL, Ulcerating Might self-heal in No well-defined risk Dogs,
braziliensis † DCL, LR lesions can 6 mo; 2.5% of groups humans,
progress to cases progress to rodents,
mucocutaneous MCL horses
form; local
lymph nodes
are palpable
before and
early on in the
onset of the
lesions
Leishmania Viannia CL, DsCL, Ulcerating Might self-heal No well-defined risk Possums,
guyanensis † MCL lesions, single within 6 mo groups sloths,
or multiple that anteaters
can progress to
mucocutaneous
form; palpable
lymph nodes
* Old World leishmaniasis.
‡ Estimates are of all New World leishmaniases, with Leishmania braziliensis comprising the vast
Etiology
Leishmania organisms are members of the Trypanosomatidae family and include
2 subgenera, Leishmania (Leishmania) and Leishmania (Viannia) . The parasite
is dimorphic, existing as a flagellate promastigote in the insect vector and as an
aflagellate amastigote that resides and replicates within mononuclear phagocytes
of the vertebrate host. Within the sandfly vector, the promastigote changes from
a noninfective procyclic form to an infective metacyclic stage (Fig. 311.1 ).
Fundamental to this transition are changes that take place in the terminal
polysaccharides of the surface lipophosphoglycan , which allow forward
migration of the infective parasites to be inoculated in the host skin during a
blood meal. Metacyclic lipophosphoglycan also plays an important role in the
entry and survival of Leishmania in the host cells. Once within the macrophage,
the promastigote transforms to an amastigote and resides and replicates within a
phagolysosome. The parasite is resistant to the acidic, hostile environment of the
macrophage and eventually ruptures the cell and goes on to infect other
macrophages. Infected macrophages have a diminished capacity to initiate and
respond to an inflammatory response, thus providing a safe haven for the
intracellular parasite.
Infection is divided into 2 main phases: acute and chronic (Table 313.1 ).
Acute infection is asymptomatic in up to 95% of infected individuals, but can
manifest as fever, lymphadenopathy, organomegaly, myocarditis, and
meningoencephalitis. Chronic infection in 60–70% of patients is indeterminate,
meaning the patient is asymptomatic but has a positive antibody titer.
Approximately 30% of infected persons proceed to chronic determinate or
symptomatic T. cruzi infection. The T. cruzi genome has been fully sequenced
and contains 12,000 genes, the most widely expanded among trypanosomatids,
and may reflect its ability to invade a wide variety of host tissues. Significant
variability has been also found, along with extensive epigenetic modification of
surface proteins, which may contribute to immune evasion. Six discrete typing
units (DTUs) are recognized, referred to as TcI to TcVI. A newly described 7th
type called Tcbat has recently been identified. DTUs may differ in geographic
distribution, predominant vector, and hosts and may also differ in disease
manifestations and response to treatment.
Table 313.1
Clinical Features and Diagnosis of Chagas Disease
GEOGRAPHIC
CLINICAL SIGNS/SYMPTOMS DIAGNOSIS
DISTRIBUTION
ACUTE FORMS *
Vectorial Endemic Incubation period: 1-2 wk Direct
countries Signs of portal of entry: indurated parasitological
cutaneous lesion (chagoma) or palpebral methods: patent
edema (Romaña sign) parasitemia up to
Most cases are mild disease (95–99%) and 90 days
unrecognized. Microscopic
Persistent fever, fatigue, lymphadenopathy, examination of
hepatomegaly, splenomegaly, morbilliform fresh blood,
rash, edema Giemsa-stained
In rare cases, myocarditis or thin and thick
meningoencephalitis blood films, or
Anemia, lymphocytosis, elevated buffy coat
AST/ALT concentrations Concentration
Risk of mortality: 0.2–0.5% methods:
microhematocrit
and Strout method
PCR techniques
Serology is not
useful.
Congenital Endemic and Incubation period: birth to several weeks Direct
nonendemic Most are asymptomatic or have mild parasitological
countries disease. methods
Prematurity, low birthweight, abortion, Concentration
neonatal death methods:
Fever, jaundice, edema, hepatomegaly, microhematocrit,
splenomegaly, respiratory distress Strout method
syndrome, myocarditis, Direct microscopy
meningoencephalitis also useful
Anemia and thrombocytopenia PCR: most
Risk of mortality: <2% sensitive technique
Serology: after 9
mo or later
Oral Restricted areas Incubation period: 3-22 days Same as for vectorial.
of endemic Fever, vomiting, periocular edema,
countries dyspnea, fever, myalgia, prostration, cough,
(Amazon basin) splenomegaly, hepatomegaly, chest pain,
and local abdominal pain, digestive hemorrhage
outbreaks Risk of mortality: 1–35%
Transfusion and Endemic and Incubation period: 8-160 days; persistent Same as for
transplant nonendemic fever vectorial.
countries Clinical characteristics similar to those of PCR techniques
vectorial cases (excluding portal of entry usually yield
signs) positive results
Risk of mortality is variable and depends days to weeks
on the severity of baseline disease. before
trypomastigotes
are detectable in
blood.
Tissue samples are
needed in some
circumstances.
Reactivation in Endemic and Behaves as other opportunistic infections Direct
HIV-infected nonendemic Reactivation with <200 CD4 cells per µL parasitological
patients countries (mostly with <100) methods, as in
Affects CNS (75–90%) as single or vectorial cases.
multiple space-occupying lesions or as Parasite can be
severe necrohemorrhagic found in CSF,
meningoencephalitis other body fluids,
Cardiac involvement (10–55%): and tissue samples.
myocarditis, pericardial effusion or PCR: not useful
worsening of previous cardiomyopathy for diagnosis of
Risk of mortality: 20% reactivation
Serology:
indicative of
chronic infection
and helpful in
cases of suspected
disease
Reactivation in Endemic and Reactivation after transplantation or in Direct
other nonendemic patients with hematologic malignancies parasitological
immunosuppressed countries Clinical characteristics similar to those of methods, as in
patients patients who undergo transfusion and those vectorial cases.
with panniculitis and other skin disorders Parasite can be
Risk of mortality is variable and depends found in tissue
on severity of baseline disease and prompt samples.
diagnosis. PCR: increasing
parasite load
detected with real-
time PCR in serial
specimens could
be indicative of a
high risk of
reactivation.
CHRONIC FORMS
Indeterminate Endemic and Asymptomatic Serology:
nonendemic Normal chest radiograph and 12-lead ECG. detection of IgG
countries PCR: low
sensitivity
Cardiac and Endemic and Cardiac manifestations: fatigue, syncope, Serology:
gastrointestinal nonendemic palpitations, dizziness, stroke; late detection of IgG
countries manifestations: chest pain (atypical), PCR: low
dyspnea, edema, left ventricular sensitivity
dysfunction, congestive heart failure;
alterations in 12-lead ECG,
echocardiography, or other heart function
tests
Gastrointestinal: dysphagia, regurgitation,
severe constipation (dilated esophagus or
colon); alterations in esophageal
manometry, barium swallow, or barium
enema
* Including reactivation in immunosuppressed patients.
ALT, Alanine transaminase; AST, aspartate transaminase; CNS, central nervous system; CSF,
cerebrospinal fluid; ECG, electrocardiogram; PCR, polymerase chain reaction.
From Pérez-Molina J, Molina I: Chagas disease, Lancet 391:82–92, 2018 (Table 2).
Table 314.1
World Health Organization Criteria for Severe Malaria, 2000
• Impaired consciousness
• Prostration
• Respiratory distress
• Multiple seizures
• Jaundice
• Hemoglobinuria
• Abnormal bleeding
• Severe anemia
• Circulatory collapse
• Pulmonary edema
P. vivax malaria has long been considered less severe than P. falciparum
malaria, but recent reports suggest that in some areas it is as frequent a cause of
severe disease and death as P. falciparum . Severe disease and death from P.
vivax are usually caused by severe anemia and sometimes splenic rupture. P.
ovale malaria is the least common type of malaria. It is similar to P. vivax
malaria and usually is found in conjunction with P. falciparum malaria. P.
malariae is the mildest and most chronic of all malaria infections. Nephrotic
syndrome is a rare complication of P. malariae infection that is not observed
with any other human malaria species. Nephrotic syndrome associated with P.
malariae infection is poorly responsive to corticosteroids. Low-level, undetected
P. malariae infection may be present for years and is sometimes unmasked by
immunosuppression or physiologic stress such as splenectomy or corticosteroid
treatment. P. knowlesi malaria is most often uncomplicated but can lead to severe
malaria and death if high-density parasitemia is present.
Recrudescence after a primary attack may occur from the survival of
erythrocyte forms in the bloodstream. Long-term relapse is caused by release of
merozoites from an exoerythrocytic source in the liver, which occurs with P.
vivax and P. ovale , or from persistence within the erythrocyte, which occurs
with P. malariae and rarely with P. falciparum . A history of typical symptoms in
a person >4 wk after return from an endemic area is therefore more likely to be
P. vivax, P. ovale, or P. malariae infection than P. falciparum infection. In the
most recent survey of malaria in the United States (2013) by the CDC, among
individuals in whom a malaria species was identified, 61% of cases were caused
by P. falciparum, 14% by P. vivax, 2% by P. malariae, 4% by P. ovale , and 2%
by mixed-species infection; 94% of P. falciparum infections were diagnosed
within 30 days of arrival in the United States, and 99% within 90 days of arrival.
In contrast, 54% of P. vivax cases occurred >30 days after arrival in the United
States.
for hypotension and cardiac monitoring for widening of the QRS complex or
lengthening of the QTc interval should be performed continuously, and blood
glucose monitoring for hypoglycemia should be performed periodically. Cardiac
adverse events may require temporary discontinuation of the drug or slowing of
the IV infusion. Parenteral therapy should be continued until the parasitemia is
<1%, which usually occurs within 48 hr, and the patient can tolerate oral
medication. Quinidine gluconate (United States) or quinine sulfate (other
countries) is administered for a total of 3 days for malaria acquired in Africa or
South America and for 7 days for malaria acquired in Southeast Asia.
Doxycycline, tetracycline, or clindamycin is then given orally to complete the
therapeutic course (see Tables 314.2 and 314.4 ). Although there are no data to
support the use of quinidine followed by atovaquone-proguanil or artemether-
lumefantrine , the difficulty of maintaining compliance with oral quinine has
led many clinicians to complete oral therapy after IV quinine with a complete
course of atovaquone-proguanil or artemether-lumefantrine.
Table 314.2
CDC Guidelines for Treatment of Malaria in the United
States (Based on Drugs Currently Available for Use in the
United States–Updated July 1, 2013)
(CDC Malaria Hotline: [770] 488-7788 or [855] 856-4713 toll-free Monday-Friday 9 AM to 5 PM EST; [770] 488-
7100 after hours, weekends, and holidays)
CLINICAL REGION RECOMMENDED DRUG AND RECOMMENDED
DIAGNOSIS/ INFECTION ADULT DOSE 1 DRUG AND
PLASMODIUM ACQUIRED PEDIATRIC DOSE 1 ;
spp. PEDIATRIC DOSE
SHOULD NEVER
EXCEED ADULT
DOSE
Uncomplicated Chloroquine- Atovaquone-proguanil (Malarone) 3 Atovaquone-
malaria/P. resistant or Adult tab = 250 mg atovaquone/100 proguanil
falciparum unknown mg proguanil (Malarone) 3
or resistance 2 4 adult tabs PO qd × 3 days Adult tab = 250 mg
Species not (All malarious atovaquone/100 mg
identified regions except proguanil
If “species not those Pediatric (ped) tab
identified” is specified as = 62.5 mg
subsequently “chloroquine- atovaquone/25 mg
diagnosed as P. sensitive,” proguanil
vivax or P. listed below) 5-8 kg: 2 ped tabs
ovale: see P. PO qd × 3 days
vivax and P. 9-10 kg: 3 ped tabs
ovale (below) PO qd × 3 days
regarding 11-20 kg: 1 adult tab
treatment with PO qd × 3 days
primaquine 21-30 kg: 2 adult
tabs PO qd × 3 days
31-40 kg: 3 adult
tabs PO qd × 3 days
>40 kg: 4 adult tabs
PO qd × 3 days
Artemether-lumefantrine (Coartem) 3
1 tablet = 20 mg artemether and 120 mg lumefantrine
A 3-day treatment schedule with a total of 6 oral doses is
recommended for both adult and pediatric patients based on
weight. The patient should receive the initial dose, followed by
2nd dose 8 hr later, then 1 dose PO bid for the following 2 days
5-<15 kg: 1 tablet per dose
15-<25 kg: 2 tablets per dose
25-<35 kg: 3 tablets per dose
≥35 kg: 4 tablets per dose
Quinine sulfate plus 1 of the Quinine sulfate 4
following: doxycycline, tetracycline, plus 1 of the
or clindamycin following:
Quinine sulfate: 542 mg base (=650 doxycycline, 6
4
mg salt) PO tid × 3 or 7 days 5
tetracycline, 6 or
Doxycycline: 100 mg PO bid × 7 days clindamycin
Tetracycline: 250 mg PO qid × 7 days Quinine sulfate: 8.3
Clindamycin: 20 mg base/kg/day PO mg base/kg (=10 mg
divided tid × 7 days salt/kg) PO tid × 3 or
7 days 5
Doxycycline: 2.2
mg/kg PO every 12
hr × 7 days
Tetracycline: 25
mg/kg/day PO
divided qid × 7 days
Clindamycin: 20 mg
base/kg/day PO
divided tid × 7 days
Mefloquine (Lariam and generics) 7 Mefloquine (Lariam
684 mg base (=750 mg salt) PO as and generics) 7
initial dose, followed by 456 mg base 13.7 mg base/kg
(=500 mg salt) PO given 6-12 hr after (=15 mg salt/kg) PO
initial dose as initial dose,
Total dose = 1,250 mg salt followed by 9.1 mg
base/kg (=10 mg
salt/kg) PO given 6-
12 hr after initial
dose. Total dose = 25
mg salt/kg
Uncomplicated Chloroquine- Chloroquine phosphate (Aralen and Chloroquine
malaria/P. sensitive generics) 8 phosphate (Aralen
falciparum (Central 600 mg base (=1,000 mg salt) PO and generics) 8
or America west immediately, followed by 300 mg 10 mg base/kg PO
Species not of Panama base (=500 mg salt) PO at 6, 24, and immediately,
identified Canal; Haiti; 48 hr followed by 5 mg
the Total dose: 1,500 mg base (=2,500 mg base/kg PO at 6, 24,
Dominican salt) and 48 hr
Republic; and or Total dose: 25 mg
most of the Hydroxychloroquine (Plaquenil and base/kg
Middle East) generics) or
620 mg base (=800 mg salt) PO Hydroxychloroquine
immediately, followed by 310 mg (Plaquenil and
base (=400 mg salt) PO at 6, 24, and generics)
48 hr 10 mg base/kg PO
Total dose: 1,550 mg base (=2,000 mg immediately,
salt) followed by 5 mg
base/kg PO at 6, 24,
and 48 hr
Total dose: 25 mg
base/kg
Uncomplicated All regions Chloroquine phosphate 8 : treatment Chloroquine
malaria/P. malariae as above phosphate: 8
or P. knowlesi or treatment as above
Hydroxychloroquine: treatment as or
above Hydroxychloroquine:
treatment as above
pregnant women, particularly in the 1st trimester because of a lack of sufficient safety data. For
pregnant women diagnosed with uncomplicated malaria caused by chloroquine-resistant P.
falciparum infection, atovaquone-proguanil or artemether-lumefantrine may be used if other
treatment options are not available or are not being tolerated, and if the potential benefit is judged
to outweigh the potential risks.
13 For P. vivax and P. ovale infections, primaquine phosphate for radical treatment of hypnozoites
should not be given during pregnancy. Pregnant patients with P. vivax and P. ovale infections
should be maintained on chloroquine prophylaxis for the duration of their pregnancy. The
chemoprophylactic dose of chloroquine phosphate is 300 mg base (=500 mg salt) orally once per
week. After delivery, pregnant patients who do not have G6PD deficiency should be treated with
primaquine.
14 Persons with a positive blood smear or history of recent possible exposure and no other
recognized pathology who have 1 or more of the following clinical criteria (impaired
consciousness/coma, severe normocytic anemia, renal failure, pulmonary edema, acute
respiratory distress syndrome, circulatory shock, disseminated intravascular coagulation,
spontaneous bleeding, acidosis, hemoglobinuria, jaundice, repeated generalized convulsions,
and/or parasitemia of >5%) are considered to have manifestations of more severe disease.
Severe malaria is most often caused by P. falciparum.
15 Patients diagnosed with severe malaria should be treated aggressively with parenteral
antimalarial therapy. Treatment with IV quinidine should be initiated as soon as possible after the
diagnosis has been made. Patients with severe malaria should be given an IV loading dose of
quinidine unless they have received >40 mg/kg of quinine in the preceding 48 hr or if they have
received mefloquine within the preceding 12 hr. Consultation with a cardiologist and a physician
with experience treating malaria is advised when treating malaria patients with quinidine. During
administration of quinidine, blood pressure monitoring (for hypotension) and cardiac monitoring
(for widening of the QRS complex and/or lengthening of the QTc interval) should be monitored
continuously and blood glucose (for hypoglycemia) should be monitored periodically. Cardiac
complications, if severe, may warrant temporary discontinuation of the drug or slowing of the
intravenous infusion.
16 Pregnant women diagnosed with severe malaria should be treated aggressively with parenteral
antimalarial therapy.
IV, Intravenous(ly); PO, orally (by mouth); qd, once daily; bid, twice daily; tid, 3 times daily; qid, 4
times daily.
From US Centers for Disease Control and Prevention.
http://www.cdc.gov/malaria/resources/pdf/treatmenttable.pdf .
Table 314.3
border.
‡ Any of the artemisinin combination treatments can be given except for artesunate-sulfadoxine-
pyrimethamine where P. vivax is resistant. Patients with P. vivax or P. ovale infections should also
be given a 14-day course of primaquine to eradicate hypnozoites (radical cure). However, severe
glucose-6-phosphate dehydrogenase deficiency is a contraindication because a 14-day course of
primaquine can cause severe hemolytic anemia in this group.
From White NJ, Pukrittayakamee S, Hien TT, et al: Malaria, Lancet 383:723–732, 2014.
Table 314.4
Treatment of Severe Malaria in Adults and Children in
Malaria-Endemic Areas
• Artesunate, 2.4 mg/kg by intravenous or intramuscular* injection, followed by 2.4 mg/kg at 12 hr and 24 hr;
continue injection once daily if necessary †
• Artemether, 3.2 mg/kg by immediate intramuscular* injection, followed by 1.6 mg/kg daily
• Quinine dihydrochloride, 20 mg salt per kg infused during 4 hr, followed by maintenance of 10 mg salt per kg
infused during 2-8 hr every 8 hr (can also be given by intramuscular injection* when diluted to 60-100 mg/mL)
Artesunate is the treatment of choice. Artemether should only be used if artesunate is unavailable. Quinine
dihydrochloride should be given only when artesunate and artemether are unavailable.
* Intramuscular injections should be given to the anterior thigh.
† Young children with severe malaria have lower exposure to artesunate and its main biologically
active metabolite dihydroartemisinin than do older children and adults. Revised dose regimens to
ensure similar drug exposures have been suggested.
From White NJ, Pukrittayakamee S, Hien TT, et al: Malaria, Lancet 383:723–732, 2014.
taste that usually requires that the cut tablet be disguised in another food, such as
chocolate syrup. Mefloquine should not be given to children if they have a
known hypersensitivity to mefloquine, are receiving cardiotropic drugs, have a
history of convulsive or certain psychiatric disorders, or travel to an area where
mefloquine resistance exists (the borders of Thailand with Myanmar and
Cambodia, western provinces of Cambodia, and eastern states of Myanmar).
Atovaquone-proguanil is started 1-2 days before travel, and mefloquine is started
2 wk before travel. It is important that these doses are given, both to allow
therapeutic levels of the drugs to be achieved and to be sure that the drugs are
tolerated. Doxycycline is an alternative for children >8 yr old. It must be given
daily and should be given with food. Side effects of doxycycline include
photosensitivity and vaginal yeast infections. Primaquine is a daily prophylaxis
option for children who cannot tolerate any of the other options, but it should be
provided in consultation with a travel medicine specialist if needed, and all
children should be checked for glucose-6-phosphate dehydrogenase deficiency
before prescribing this medication, because it is contraindicated in children with
G6PD deficiency. Provision of medication can be considered in individuals who
refuse to take prophylaxis or will be in very remote areas without accessible
medical care. Provision of medication for self-treatment of malaria should be
done in consultation with a travel medicine specialist, and the medication
provided should be different than that used for prophylaxis.
Table 314.5
Chemoprophylaxis of Malaria for Children
DOSAGE
AREA DRUG ADVANTAGES DISADVANTAGES BEST USE
(ORAL)
Chloroquine- Mefloquine* † <10 kg: 4.6 mg Once weekly Bitter taste Children
resistant area base (5 mg dosing No pediatric going to
salt)/kg/wk formulation malaria-
10-19 kg: Side effects of endemic
sleep area for ≥4
tab/wk
disturbance, wk
20-30 kg: vivid dreams Children
tab/wk unlikely to
31-45 kg: take daily
medication
tab/wk
>45 kg: 1
tab/wk (228 mg
base)
Doxycycline ‡ 2 mg/kg daily Inexpensive Cannot give to Children going
(max 100 mg) children <8 yr to area for <4
Daily dosing wk who cannot
Must take with take or cannot
food or causes obtain
stomach upset atovaquone-
Photosensitivity proguanil
Atovaquone/proguanil Pediatric Pediatric Daily dosing Children going
§ (Malarone) tabs: formulation Expensive to malaria-
62.5 mg Generally Can cause endemic area
atovaquone/ well stomach upset for <4 wk
25 mg tolerated
proguanil
Adult tabs:
250 mg
proguanil/
100 mg
proguanil
5-8 kg: pediatric
tab once daily
(off-label)
9-10 kg:
pediatric tab
once daily (off-
label)
11-20 kg: 1
pediatric tab
once daily
21-30 kg: 2
pediatric tabs
once daily
31-40 kg: 3
pediatric tabs
once daily
>40 kg: 1 adult
tab once daily
Chloroquine- Chloroquine 5 mg Once Bitter taste Best
susceptible phosphate base/kg/wk weekly No pediatric medication for
area (max: 300 mg dosing formulation children
base) Inexpensive traveling to
Generally areas with
well Plasmodium
tolerated falciparum or
Plasmodium
vivax that is
chloroquine
susceptible
Drugs used for
chloroquine-resistant
areas can also be used
in chloroquine-
susceptible areas
* Chloroquine and mefloquine should be started 1-2 wk prior to departure and continued for 4 wk
Table 316.1
Signs and Symptoms Occurring Before Diagnosis or
During the Course of Untreated Acute Congenital
Toxoplasmosis in 152 Infants (A) and in 101 of These Same
Children After They Had Been Followed ≥4 Yr (B)
† Patients with otherwise undiagnosed nonneurologic diseases during the 1st 2 mo of life.
Adapted from Eichenwald H: A study of congenital toxoplasmosis. In Slim JC, editor: Human
toxoplasmosis , Copenhagen, 1960, Munksgaard, pp. 41–49. Study performed in 1947. The most
severely involved institutionalized patients were not included in the later study of 101 children.
Systemic Signs
From 25% to >50% of infants with clinically apparent disease at birth are born
prematurely. Parasite clonal types other than type II are more often associated
with prematurity and more-severe disease. Intrauterine growth restriction, low
Apgar scores, and temperature instability are common. Other manifestations
may include lymphadenopathy, hepatosplenomegaly, myocarditis, pneumonitis,
nephrotic syndrome, vomiting, diarrhea, and feeding problems. Bands of
metaphyseal lucency and irregularity of the line of provisional calcification at
the epiphyseal plate may occur without periosteal reaction in the ribs, femurs,
and vertebrae. Congenital toxoplasmosis may be confused with erythroblastosis
fetalis resulting from isosensitization, although the Coombs test result is usually
negative with congenital T. gondii infection.
Skin
Cutaneous manifestations among newborn infants with congenital toxoplasmosis
include rashes and jaundice and/or petechiae secondary to thrombocytopenia, but
ecchymoses and large hemorrhages secondary to thrombocytopenia also occur.
Rashes may be fine punctate, diffuse maculopapular, lenticular, deep blue-red,
sharply defined macular, or diffuse blue and papular. Macular rashes involving
the entire body including the palms and soles, exfoliative dermatitis, and
cutaneous calcifications have been described. Jaundice with hepatic
involvement and/or hemolysis, cyanosis due to interstitial pneumonitis from
congenital infection, and edema secondary to myocarditis or nephrotic syndrome
may be present. Jaundice and conjugated hyperbilirubinemia may persist for
months.
diagnosis definitively and in immunocompromised patients for diagnosis and
monitoring treatment.
Table 316.2
Generalizations Concerning Clinical Presentations,
Toxoplasma -Specific Diagnostic Tests, and Treatment
Prevention
Although ascariasis is the most prevalent worm infection in the world, little
attention has been given to its control (Table 317.1 ). Anthelmintic
chemotherapy programs can be implemented in 1 of 3 ways: (1) offering
universal treatment to all individuals in an area of high endemicity; (2) offering
treatment targeted to groups with high frequency of infection, such as children
attending primary school; or (3) offering individual treatment based on intensity
of current or past infection. Improving education about and practices of sanitary
conditions and sewage facilities, discontinuing the practice of using human feces
as fertilizer, and education are the most effective long-term preventive measures.
Table 317.1
Bibliography
headache with elevated Toxocara antibody titers. Eosinophilia may be present in
50–75% of cases. The prevalence of positive Toxocara serology in the general
population supports that most children with T. canis infection are asymptomatic
and will not develop overt clinical sequelae over time. A correlation between
positive Toxocara serology and allergic asthma has also been described.
Table 324.1
Clinical Syndromes of Human Toxocariasis
Diagnosis
A presumptive diagnosis of toxocariasis can be established in a young child with
eosinophilia (>20%), leukocytosis, hepatomegaly, fevers, wheezing, and a
history of geophagia and exposure to puppies or unrestrained dogs. Supportive
laboratory findings include hypergammaglobulinemia and elevated
isohemagglutinin titers to A and B blood group antigens. Most patients with
VLM have an absolute eosinophil count >500/µL. Eosinophilia is less common
in patients with OLM. Biopsy confirms the diagnosis. When biopsies cannot be
obtained, an enzyme-linked immunosorbent assay using excretory-secretory
CHAPTER 328
Table 328.1
Key Features of Common Tapeworms in Children
PARASITE
GEOGRAPHY SOURCE SYMPTOMS TREATMENT
SPECIES
Taenia saginata Asia, Africa, Cysts in beef Abdominal discomfort, motile Praziquantel or
Latin America proglottid migration, passing niclosamide,
segments possibly
nitazoxanide
Taenia solium Asia, Africa, Cysticerci in Minimal, proglottids in stool Praziquantel or
Latin America pork niclosamide,
possibly
nitazoxanide
Taenia asiatica Asia Pigs Minimal Praziquantel or
niclosamide,
possibly
nitazoxanide
Diphyllobothrium Worldwide, Plerocercoid Usually minimal; with prolonged or Praziquantel or
spp. often northern cysts in heavy infection with D. latum , niclosamide
areas freshwater vitamin B12 deficiency
fish
Hymenolepis Worldwide, Infected Mild abdominal discomfort Praziquantel,
often northern humans, niclosamide, or
areas rodents nitazoxanide
Dipylidium Worldwide Domestic Proglottids in stool, anal pruritus Praziquantel or
caninum dogs and cats confused with pinworm niclosamide
Etiology
The beef tapeworm (Taenia saginata) , the pork tapeworm (T. solium), and the
Asian tapeworm (Taenia asiatica) are long worms (4-10 m) named for their
intermediate hosts (T. saginata , T. solium ) or geographic distribution (T.
asiatica ; larval host is the pig). The adult worms are found only in the human
intestine. As with the adult stage of all tapeworms, their body is a series of 100s
or 1000s of flattened segments (proglottids ) with an anterior attachment organ
(scolex) that anchors the parasite to the bowel wall. New segments arise from the
distal aspect of the scolex with progressively more mature segments attached
distally. The gravid terminal segments contain 50,000-100,000 eggs, and the
eggs or even detached intact proglottids pass out of the child through the anus
(with or separate from defecation). These tapeworms differ most significantly in
that the intermediate stage of the pork tapeworm (cysticercus ) can also infect
humans and cause significant morbidity (see Chapter 329 ), whereas the larval
stage of T. saginata does not cause human disease. T. asiatica is similar to and
often confused with the beef tapeworm.
Epidemiology
The pork and beef tapeworms are distributed worldwide, with the highest risk
for infection in Latin America, Africa, India, Southeast Asia, and China, where
the relevant intermediate host is raised domestically. The prevalence in adults
may not reflect the prevalence in young children, because cultural practices may
dictate how well meat is cooked and how much is served to children.
Pathogenesis
When children ingest raw or undercooked meat containing larval cysts, gastric
acid and bile facilitate release of immature scolices that attach to the lumen of
the small intestine. The parasite grows, adding new segments at the base of the
scolex. The terminal segments mature and after 2-3 mo produce eggs that are
CHAPTER 332
Disorders of organs outside the gastrointestinal (GI) tract can produce symptoms
and signs that mimic digestive tract disorders and should be considered in the
differential diagnosis (Table 332.1 ). In children with normal growth and
development, treatment may be initiated without a formal evaluation based on a
presumptive diagnosis after taking a history and performing a physical
examination. Poor weight gain or weight loss is often associated with a
significant pathologic process and usually necessitates a more formal evaluation.
Table 332.1
Some Nondigestive Tract Causes of Gastrointestinal
Symptoms in Children
ANOREXIA
Systemic disease: inflammatory, neoplastic
Cardiorespiratory compromise
Iatrogenic: drug therapy, unpalatable therapeutic diets
Depression
Anorexia nervosa
VOMITING
Inborn errors of metabolism
Medications: erythromycin, chemotherapy, nonsteroidal antiinflammatory drugs, marijuana
Increased intracranial pressure
Brain tumor
Infection of the urinary tract
Labyrinthitis
Adrenal insufficiency
Pregnancy
Psychogenic
Abdominal migraine
Poisoning/toxins
Renal disease
DIARRHEA
Infection: otitis media, urinary tract infection
Uremia
Medications: antibiotics, cisapride
Tumors: neuroblastoma
Pericarditis
Adrenal insufficiency
CONSTIPATION
Hypothyroidism
Spina bifida
Developmental delay
Dehydration: diabetes insipidus, renal tubular lesions
Medications: narcotics
Lead poisoning
Infant botulism
ABDOMINAL PAIN
Pyelonephritis, hydronephrosis, renal colic
Pneumonia (lower lobe)
Pelvic inflammatory disease
Porphyria
Fabry disease
Angioedema
Endocarditis
Abdominal migraine
Familial Mediterranean fever
Sexual or physical abuse
Systemic lupus erythematosus
School phobia
Sickle cell crisis
Vertebral disk inflammation
Psoas abscess
Pelvic osteomyelitis or myositis
Medications
ABDOMINAL DISTENTION OR MASS
Ascites: nephrotic syndrome, neoplasm, heart failure
Discrete mass: Wilms tumor, hydronephrosis, neuroblastoma, mesenteric cyst, hepatoblastoma, lymphoma
Pregnancy
JAUNDICE
Hemolytic disease
Urinary tract infection
Sepsis
Hypothyroidism
Panhypopituitarism
Dysphagia
Difficulty in swallowing is termed dysphagia. Painful swallowing is termed
odynophagia . Globus is the sensation of something stuck in the throat without
a clear etiology. Swallowing is a complex process that starts in the mouth with
mastication and lubrication of food that is formed into a bolus. The bolus is
pushed into the pharynx by the tongue. The pharyngeal phase of swallowing is
rapid and involves protective mechanisms to prevent food from entering the
airway. The epiglottis is lowered over the larynx while the soft palate is elevated
against the nasopharyngeal wall; respiration is temporarily arrested while the
upper esophageal sphincter opens to allow the bolus to enter the esophagus. In
the esophagus, peristaltic coordinated muscular contractions push the food bolus
toward the stomach. The lower esophageal sphincter relaxes shortly after the
upper esophageal sphincter, so liquids that rapidly clear the esophagus enter the
stomach without resistance.
Dysphagia is classified as oropharyngeal dysphagia and esophageal
dysphagia. Oropharyngeal dysphagia occurs when the transfer of the food
bolus from the mouth to the esophagus is impaired (also termed transfer
dysphagia ). The striated muscles of the mouth, pharynx, and upper esophageal
sphincter are affected in oropharyngeal dysphagia. Neurologic and muscular
disorders can give rise to oropharyngeal dysphagia (Table 332.2 ). Chiari
malformations, Russell-Silver syndrome, and cri du chat may present with upper
esophageal sphincter dysfunction, manifest by dysphagia with solids. The most
serious complication of oropharyngeal dysphagia is life-threatening aspiration.
Table 332.2
Causes of Oropharyngeal Dysphagia
NEUROMUSCULAR DISORDERS
Cerebral palsy
Brain tumors
Cerebrovascular disease/stroke
Chiari malformation
Polio and postpolio syndromes
Multiple sclerosis
Myositis
Dermatomyositis
Myasthenia gravis
Muscular dystrophies
Acquired or inherited dystonia syndrome
Dysautonomia
METABOLIC AND AUTOIMMUNE DISORDERS
Hyperthyroidism
Systemic lupus erythematosus
Sarcoidosis
Amyloidosis
INFECTIOUS DISEASE
Meningitis
Botulism
Diphtheria
Lyme disease
Neurosyphilis
Viral infection: polio, coxsackievirus, herpes, cytomegalovirus
STRUCTURAL LESIONS
Inflammatory: abscess, pharyngitis
Congenital web
Cricopharyngeal bar
Dental problems
Bullous skin lesions
Plummer-Vinson syndrome
Zenker diverticulum
Extrinsic compression: osteophytes, lymph nodes, thyroid swelling, aberrant
right subclavian artery (dysphagia lusoria)
OTHER
Corrosive injury
Side effects of medications
After surgery
After radiation therapy
Adapted from Gasiorowska A, Faas R: Current approach to dysphagia. Gastroenterol Hepatol
5(4):269–279, 2009.
Table 332.3
Causes of Esophageal Dysphagia
NEUROMUSCULAR
Eosinophilic esophagitis
Achalasia cardia
Diffuse esophageal spasm
Scleroderma
GERD
INTRINSIC LESIONS
Foreign bodies including pills
Esophagitis: GERD, eosinophilic esophagitis, infections
Stricture: corrosive injury, pill induced, peptic
Esophageal webs
Esophageal rings
Esophageal diverticula
Neoplasm
Chagas disease
EXTRINSIC LESIONS
Vascular compression
Mediastinal lesion
Cervical osteochondritis
Vertebral abnormalities
GERD , Gastroesophageal reflux disease.
Adapted from Gasiorowska A, Faas R: Current approach to dysphagia. Gastroenterol Hepatol
5(4):269–279, 2009.
Regurgitation
Regurgitation is the effortless movement of stomach contents into the esophagus
and mouth. It is not associated with distress, and infants with regurgitation are
often hungry immediately after an episode. The lower esophageal sphincter
prevents reflux of gastric contents into the esophagus. Regurgitation is a result of
gastroesophageal reflux through an incompetent or, in infants, immature lower
esophageal sphincter. This is often a developmental process, and regurgitation or
“spitting” resolves with maturity. Regurgitation should be differentiated from
vomiting, which denotes an active reflex process with an extensive differential
diagnosis (Table 332.4 ).
Table 332.4
Differential Diagnosis of Emesis During Childhood
hernias.
† Meningitis, sepsis.
Anorexia
Anorexia means prolonged lack of appetite. Hunger and satiety centers are
located in the hypothalamus; it seems likely that afferent nerves from the GI tract
to these brain centers are important determinants of the anorexia that
characterizes many diseases of the stomach and intestine (see Chapter 47 ).
Satiety is stimulated by distention of the stomach or upper small bowel, the
signal being transmitted by sensory afferents, which are especially dense in the
upper gut. Chemoreceptors in the intestine, influenced by the assimilation of
nutrients, also affect afferent flow to the appetite centers. Impulses reach the
hypothalamus from higher centers, possibly influenced by pain or the emotional
disturbance of an intestinal disease. Other regulatory factors include hormones,
ghrelin, leptin, and plasma glucose, which, in turn, reflect intestinal function (see
Chapter 47 ).
Vomiting
Vomiting is a highly coordinated reflex process that may be preceded by
increased salivation and begins with involuntary retching. Violent descent of the
diaphragm and constriction of the abdominal muscles with relaxation of the
gastric cardia actively force gastric contents back up the esophagus. This process
is coordinated in the medullary vomiting center, which is influenced directly by
afferent innervation and indirectly by the chemoreceptor trigger zone and higher
central nervous system (CNS) centers. Many acute or chronic processes can
cause vomiting (see Tables 332.1 and 332.4 ).
Vomiting caused by obstruction of the GI tract is probably mediated by
intestinal visceral afferent nerves stimulating the vomiting center (Table 332.5 ).
If obstruction occurs below the second part of the duodenum, vomitus is usually
bile stained. Emesis can also become bile stained with repeated vomiting in the
absence of obstruction when duodenal contents are refluxed into the stomach.
Nonobstructive lesions of the digestive tract can also cause vomiting; this
includes diseases of the upper bowel, pancreas, liver, or biliary tree. CNS or
metabolic derangements and cyclic vomiting syndrome (Table 332.6 ) can lead
to severe, persistent emesis. Marijuana use among teens has also led to cannabis
hyperemesis syndrome (see Chapter 140.3 ).
Table 332.5
Causes of Gastrointestinal Obstruction
ESOPHAGUS
Congenital
Esophageal atresia
Vascular rings
Schatzki ring
Tracheobronchial remnant
Acquired
Esophageal stricture
Foreign body
Achalasia
Chagas disease
Collagen vascular disease
STOMACH
Congenital
Antral webs
Pyloric stenosis
Acquired
Bezoar, foreign body
Pyloric stricture (ulcer)
Chronic granulomatous disease of childhood
Eosinophilic gastroenteritis
Crohn disease
Epidermolysis bullosa
SMALL INTESTINE
Congenital
Duodenal atresia
Annular pancreas
Malrotation/volvulus
Malrotation/Ladd bands
Ileal atresia
Meconium ileus
Meckel diverticulum with volvulus or intussusception
Inguinal hernia
Internal hernia
Intestinal duplication
Pseudoobstruction
Acquired
Postsurgical adhesions
Crohn disease
Intussusception
Distal ileal obstruction syndrome (cystic fibrosis)
Duodenal hematoma
Superior mesenteric artery syndrome
COLON
Congenital
Meconium plug
Hirschsprung disease
Colonic atresia, stenosis
Imperforate anus
Rectal stenosis
Pseudoobstruction
Volvulus
Colonic duplication
Acquired
Ulcerative colitis (toxic megacolon)
Chagas disease
Crohn disease
Fibrosing colonopathy (cystic fibrosis)
Table 332.6
Criteria for Cyclic Vomiting Syndrome
All of the criteria must be met for the consensus definition of cyclic vomiting syndrome:
• At least 5 attacks in any interval, or a minimum of 3 episodes during a 6-mo period
• Recurrent episodes of intense vomiting and nausea lasting 1 hr to 10 days and occurring at least 1 wk apart
• Stereotypical pattern and symptoms in the individual patient
• Vomiting during episodes occurs ≥4 times/hr for ≥1 hr
• Return to baseline health between episodes
• Not attributed to another disorder
From Li, B UK, Lefevre F, Chelimsky GG, et al: North American Society for Pediatric
Gastroenterology, Hepatology, and Nutrition consensus statement on the diagnosis and
management of cyclic vomiting syndrome. J Pediatr Gastroenterol Nutr 47:379–393, 2008.
Table 332.7
Complications of Vomiting
Table 332.8
Pharmacologic Therapies for Vomiting Episodes
DISORDER/THERAPEUTIC
DRUG DOSAGE
DRUG CLASS
REFLUX
Dopamine antagonist Metoclopramide 0.1-0.2 mg/kg PO or IV qid
(Reglan)
GASTROPARESIS
Dopamine antagonist Metoclopramide 0.1-0.2 mg/kg PO or IV qid
(Reglan)
Motilin agonist Erythromycin 3-5 mg/kg PO or IV tid-qid
INTESTINAL PSEUDOOBSTRUCTION
Stimulation of intestinal migratory Octreotide 1 µg/kg SC bid-tid
myoelectric complexes (Sandostatin)
CHEMOTHERAPY
Dopamine antagonist Metoclopramide 0.5-1.0 mg/kg IV qid, with antihistamine
prophylaxis of extrapyramidal side effects
Serotoninergic 5-HT3 antagonist Ondansetron (Zofran) 0.15-0.3 mg/kg IV or PO tid
Phenothiazines (extrapyramidal, Prochlorperazine ≈0.3 mg/kg PO bid-tid
hematologic side effects) (Compazine)
Chlorpromazine >6 mo of age: 0.5 mg/kg PO or IV tid-qid
(Thorazine)
Steroids Dexamethasone 0.1 mg/kg PO tid
(Decadron)
Cannabinoids Tetrahydrocannabinol 0.05-0.1 mg/kg PO bid-tid
(Nabilone)
POSTOPERATIVE
Ondansetron, See under chemotherapy
phenothiazines
MOTION SICKNESS, VESTIBULAR DISORDERS
Antihistamine Dimenhydrinate 1 mg/kg PO tid-qid
(Dramamine)
Anticholinergic Scopolamine Adults: 1 patch/3 days
(Transderm Scop)
ADRENAL CRISIS
Steroids Cortisol 2 mg/kg IV bolus followed by 0.2-0.4 mg/kg/hr IV
(±1 mg/kg IM)
ECG , Electrocardiogram; GI , gastrointestinal.
From Kliegman RM, Greenbaum LA, Lye PS, editors: Practical strategies in pediatric diagnosis
and therapy, ed 2, Philadelphia, 2004, Elsevier, p 317.
Table 332.9
Supportive and Nonpharmacologic Therapies for Vomiting
Episodes
DISEASE THERAPY
All Treat cause
• Obstruction: operate
• Allergy: change diet (±steroids)
• Metabolic error: Rx defect
• Acid peptic disease: H2RAs, PPIs, etc.
COMPLICATIONS
Dehydration IV fluids, electrolytes
Hematemesis Transfuse, correct coagulopathy
Esophagitis H2 RAs, PPIs
Malnutrition NG or NJ drip feeding useful for many chronic conditions
Meconium Gastrografin enema
ileus
DIOS Gastrografin enema; balanced colonic lavage solution (e.g., GoLYTELY)
Intussusception Barium enema; air reduction enema
Hematemesis Endoscopic: injection sclerotherapy or banding of esophageal varices; injection therapy, fibrin
sealant application, or heater probe electrocautery for selected upper GI tract lesions
Sigmoid Colonoscopic decompression
volvulus
Reflux Positioning; dietary measures (infants: rice cereal, 1 tbs/oz of formula)
Psychogenic Psychotherapy; tricyclic antidepressants; anxiolytics (e.g., diazepam: 0.1 mg/kg PO tid-qid)
components
DIOS , Distal intestinal obstruction syndrome; GI , gastrointestinal; H2 RA , H2 -receptor
antagonist; NG , nasogastric; NJ , nasojejunal; PPIs , proton pump inhibitors; tbs , tablespoon.
From Kliegman RM, Greenbaum LA, Lye PS, editors: Practical strategies in pediatric diagnosis
and therapy , ed 2, Philadelphia, 2004, Elsevier, p 319.
Diarrhea
Diarrhea is best defined as excessive loss of fluid and electrolyte in the stool.
Acute diarrhea is defined as sudden onset of excessively loose stools of >10
mL/kg/day in infants and >200 g/24 hr in older children, which lasts <14 days.
When the episode lasts longer than 14 days, it is called chronic or persistent
diarrhea.
Normally, a young infant has approximately 5 mL/kg/day of stool output; the
volume increases to 200 g/24 hr in an adult. The greatest volume of intestinal
water is absorbed in the small bowel; the colon concentrates intestinal contents
against a high osmotic gradient. The small intestine of an adult can absorb 10-11
L/day of a combination of ingested and secreted fluid, whereas the colon absorbs
approximately 0.5 L. Disorders that interfere with absorption in the small bowel
tend to produce voluminous diarrhea, whereas disorders compromising colonic
absorption produce lower-volume diarrhea. Dysentery (small-volume, frequent
bloody stools with mucus, tenesmus, and urgency) is the predominant symptom
of colitis.
The basis of all diarrheas is disturbed intestinal solute transport and water
absorption. Water movement across intestinal membranes is passive and is
determined by both active and passive fluxes of solutes, particularly sodium,
chloride, and glucose. The pathogenesis of most episodes of diarrhea can be
explained by secretory, osmotic, or motility abnormalities or a combination of
these (Table 332.10 ).
Table 332.10
Mechanisms of Diarrhea
PRIMARY STOOL
DEFECT EXAMPLES COMMENT
MECHANISM EXAMINATION
Secretory Decreased Watery, normal Cholera, toxigenic Escherichia Persists during fasting;
absorption, osmolality with ion coli ; carcinoid, VIP, bile salt malabsorption
increased gap <100 mOsm/kg neuroblastoma, congenital can also increase
secretion, chloride diarrhea, Clostridium intestinal water
electrolyte difficile , cryptosporidiosis secretion; no stool
transport (AIDS) leukocytes
Osmotic Maldigestion, Watery, acidic, and Lactase deficiency, glucose- Stops with fasting;
transport reducing galactose malabsorption, increased breath
defects substances; lactulose, laxative abuse hydrogen with
ingestion of increased carbohydrate
unabsorbable osmolality with ion malabsorption; no stool
substances gap >100 mOsm/kg leukocytes
Increased Decreased Loose to normal- Irritable bowel syndrome, Infection can also
motility transit time appearing stool, thyrotoxicosis, postvagotomy contribute to increased
stimulated by dumping syndrome motility
gastrocolic reflex
Decreased Defect in Loose to normal- Pseudoobstruction, blind loop Possible bacterial
motility neuromuscular appearing stool overgrowth
unit(s) stasis
(bacterial
overgrowth)
Decreased Decreased Watery Short bowel syndrome, celiac Might require elemental
surface area functional disease, rotavirus enteritis diet plus parenteral
(osmotic, capacity alimentation
motility)
Mucosal Inflammation, Blood and Salmonella, Shigella infection; Dysentery evident in
invasion decreased increased WBCs in amebiasis; Yersinia, blood, mucus, and
colonic stool Campylobacter infection WBCs
reabsorption,
increased
motility
VIP , Vasoactive intestinal peptide; WBC , white blood cell.
From Kliegman RM, Greenbaum LA, Lye PS, editors: Practical strategies in pediatric diagnosis
and therapy, ed 2, Philadelphia, 2004, Elsevier, p 274.
Secretory diarrhea occurs when the intestinal epithelial cell solute transport
system is in an active state of secretion. It is often caused by a secretagogue,
such as cholera toxin, binding to a receptor on the surface epithelium of the
bowel and thereby stimulating intracellular accumulation of cyclic adenosine
monophosphate or cyclic guanosine monophosphate. Some intraluminal fatty
acids and bile salts cause the colonic mucosa to secrete through this mechanism.
Diarrhea not associated with an exogenous secretagogue can also have a
secretory component (congenital microvillus inclusion disease). Secretory
diarrhea is usually of large volume and persists even with fasting. The stool
osmolality is predominantly indicated by the electrolytes and the ion gap is 100
mOsm/kg or less. The ion gap is calculated by subtracting the concentration of
electrolytes from total osmolality:
Table 332.11
Differential Diagnosis of Diarrhea
Constipation
Any definition of constipation is relative and depends on stool consistency, stool
frequency, and difficulty in passing the stool. A normal child might have a soft
stool only every second or third day without difficulty; this is not constipation. A
hard stool passed with difficulty every third day should be treated as
constipation. Constipation can arise from defects either in filling or emptying the
rectum (Table 332.12 ).
Table 332.12
Causes of Constipation
NONORGANIC (FUNCTIONAL)—RETENTIVE
Anatomic
Anal stenosis, atresia with fistula
Imperforate anus
Anteriorly displaced anus
Intestinal stricture (postnecrotizing enterocolitis)
Anal stricture
Abnormal Musculature
Prune-belly syndrome
Gastroschisis
Down syndrome
Muscular dystrophy
Intestinal Nerve or Muscle Abnormalities
Hirschsprung disease
Pseudoobstruction (visceral myopathy or neuropathy)
Intestinal neuronal dysplasia
Spinal cord lesions
Tethered cord
Autonomic neuropathy
Spinal cord trauma
Spina bifida
Chagas disease
Drugs
Anticholinergics
Narcotics
Methylphenidate
Phenytoin
Antidepressants
Chemotherapeutic agents (vincristine)
Pancreatic enzymes (fibrosing colonopathy)
Lead, arsenic, mercury
Vitamin D intoxication
Calcium channel blocking agents
Metabolic Disorders
Hypokalemia
Hypercalcemia
Hypothyroidism
Diabetes mellitus, diabetes insipidus
Porphyria
Intestinal Disorders
Celiac disease
Cow's milk protein intolerance
Cystic fibrosis (meconium ileus equivalent)
Inflammatory bowel disease (stricture)
Tumor
Connective tissue disorders
Systemic lupus erythematosus
Scleroderma
Psychiatric Diagnosis
Anorexia nervosa
A nursing infant might have very infrequent stools of normal consistency; this
is usually a normal pattern. True constipation in the neonatal period is most
likely secondary to Hirschsprung disease, intestinal pseudoobstruction, or
hypothyroidism.
Defective rectal filling occurs when colonic peristalsis is ineffective (in cases
of hypothyroidism or opiate use and when bowel obstruction is caused either by
a structural anomaly or by Hirschsprung disease). The resultant colonic stasis
leads to excessive drying of stool and a failure to initiate reflexes from the
rectum that normally trigger evacuation. Emptying the rectum by spontaneous
evacuation depends on a defecation reflex initiated by pressure receptors in the
rectal muscle. Therefore stool retention can also result from lesions involving
nerve endings probably account for the pain caused by ischemia. Perception of
these painful stimuli can be modulated by input from both cerebral and
peripheral sources. Psychologic factors are particularly important. Tables 332.13
and 332.14 list features of abdominal pain. Pain that suggests a potentially
serious organic etiology is associated with age younger than 5 yr; fever; weight
loss; bile- or blood-stained emesis; jaundice; hepatosplenomegaly; back or flank
pain or pain in a location other than the umbilicus; awakening from sleep in
pain; referred pain to shoulder, groin or back; elevated erythrocyte sedimentation
rate, white blood cell count, or C-reactive protein; anemia; edema;
hematochezia; or a strong family history of inflammatory bowel disease or celiac
disease.
Table 332.13
Chronic Abdominal Pain in Children
Table 332.14
Distinguishing Features of Acute Abdominal Pain in
Children
Gastrointestinal Hemorrhage
Bleeding can occur anywhere along the GI tract, and identification of the site
may be challenging (Table 332.15 ). Bleeding that originates in the esophagus,
stomach, or duodenum can cause hematemesis. When exposed to gastric or
intestinal juices, blood quickly darkens to resemble coffee grounds; massive
bleeding is likely to be red. Red or maroon blood in stools, hematochezia,
signifies either a distal bleeding site or massive hemorrhage above the distal
ileum. Moderate to mild bleeding from sites above the distal ileum tends to
cause blackened stools of tarry consistency (melena); major hemorrhages in the
duodenum or above can also cause melena.
Table 332.15
Differential Diagnosis of Gastrointestinal Bleeding in
Childhood
Erosive damage to the mucosa of the GI tract is the most common cause of
bleeding, although variceal bleeding secondary to portal hypertension occurs
often enough to require consideration. Prolapse gastropathy producing
subepithelial hemorrhage and Mallory-Weiss lesions secondary to mucosal tears
associated with emesis are causes of upper intestinal bleeds. Vascular
malformations are a rare cause in children; they are difficult to identify (Figs.
332.1 and 332.2 ). Upper intestinal bleeding is evaluated with
esophagogastroduodenoscopy. Evaluation of the small intestine is facilitated by
capsule endoscopy. The capsule-sized imaging device is swallowed in older
children or placed endoscopically in younger children. Lower GI bleeding is
investigated with a colonoscopy. In brisk intestinal bleeding of unknown
location, a tagged red blood cell scan is helpful in locating the site of the
bleeding, although CT angiography is usually diagnostic. Occult blood in stool is
usually detected by using commercially available fecal occult blood testing
cards, which are based on a chemical reaction between the chemical guaiac and
oxidizing action of a substrate (hemoglobin), giving a blue color. The guaiac test
is very sensitive, but random testing can miss chronic blood loss, which can lead
to iron-deficiency anemia. GI hemorrhage can produce hypotension and
tachycardia but rarely causes GI symptoms; brisk duodenal or gastric bleeding
can lead to nausea, vomiting, or diarrhea. The breakdown products of
intraluminal blood might tip patients into hepatic coma if liver function is
already compromised and can lead to elevation of serum bilirubin.
premolars. On the other hand, the permanent first, second, and third molars arise
from extension of the dental laminae distal to the second primary molars; buds
for these teeth develop at approximately 4 mo of gestation, 1 yr of age, and 4-5
yr of age, respectively.
Histodifferentiation–Morphodifferentiation
As the epithelial bud proliferates, the deeper surface invaginates and a mass of
mesenchyme becomes partially enclosed. The epithelial cells differentiate into
the ameloblasts that lay down an organic matrix that forms enamel; the
mesenchyme forms the dentin and dental pulp.
Calcification
After the organic matrix has been laid down, the deposition of the inorganic
mineral crystals takes place from several sites of calcification that later coalesce.
The characteristics of the inorganic portions of a tooth can be altered by
disturbances in formation of the matrix, decreased availability of minerals, or the
incorporation of foreign materials. Such disturbances can affect the color,
texture, or thickness of the tooth surface. Calcification of primary teeth begins at
3-4 mo in utero and concludes postnatally at approximately 12 mo, with
mineralization of the second primary molars (Table 333.1 ).
Table 333.1
Calcification, Crown Completion, and Eruption
Eruption
At the time of tooth bud formation, each tooth begins a continuous movement
toward the oral cavity. Table 333.1 lists the times of eruption of the primary and
permanent teeth.
Anomalies Associated With Eruption Pattern: Delayed eruption of the 20
primary teeth can be familial or indicate systemic or nutritional disturbances
such as hypopituitarism, hypothyroidism, cleidocranial dysplasia, trisomy 21,
and multiple other syndromes. Failure of eruption of single or small groups of
teeth can arise from local causes such as malpositioned teeth, supernumerary
teeth, cysts, or retained primary teeth. Premature loss of primary teeth is most
commonly caused by premature eruption of the permanent teeth. If the entire
dentition is advanced for age and sex, precocious puberty or hyperthyroidism
should be considered.
Natal teeth are observed in approximately 1 in 2,000 newborn infants, usually
in the position of the mandibular central incisors. Natal teeth are present at birth,
whereas neonatal teeth erupt in the first mo of life. Attachment of natal and
neonatal teeth is generally limited to the gingival margin, with little root
formation or bony support. They may be a supernumerary or a prematurely
erupted primary tooth. A radiograph can easily differentiate between the 2
CHAPTER 334
Table 334.1
Table 338.1
Red Flags Suggestive of a Spreading Dental Infection
• Pyrexia
• Tachycardia or tachypnea
• Trismus; may be relative due to pain or absolute due to a collection within the muscle causing muscle spasm in
cases of masticator space involvement
• Raised tongue and floor of mouth, drooling
• Periorbital cellulitis
• Difficulty with speaking, swallowing, and breathing
• Hypotension
• Increased white blood cell count
• Lymphadenopathy
• Dehydration
From Robertson DP, Keys W, Rautemaa-Richardson R, et al: Management of severe acute dental
mg fluoride per liter (ppm F) was introduced in the United States in the 1940s.
Because fluoride from water supplies is now one of several sources of fluoride,
the Department of Health and Human Services proposes to not have a fluoride
range, but instead to limit the recommendation to the lower limit of 0.7 ppm F.
The rationale is to balance the benefits of preventing dental caries with reducing
the chance of fluorosis. Children who reside in areas with fluoride-deficient
water supplies or who consume primarily bottled water, and are at risk for caries,
benefit from dietary fluoride supplements (Table 338.2 ). If the patient uses a
private water supply, it is necessary to get the water tested for fluoride levels
before prescribing fluoride supplements. To avoid potential overdoses, no
fluoride prescription should be written for more than a total of 120 mg of
fluoride. However, because of confusion regarding fluoride supplements among
practitioners and parents, association of supplements with fluorosis, and lack of
parent compliance with the daily administration, supplements may no longer be
the first-line approach for preventing caries in preschool-aged children.
Table 338.2
Supplemental Fluoride Dosage Schedule
Table 340.1
FIG. 341.1 Major aphthous in a child. (From Gürkan A, Özlü SG, Altiaylik-Özer P, et al:
Recurrent aphthous stomatitis in childhood and adolescence: a single-center
experience. Pediatr Dermatol 32(4):476–480, 2015. Fig. 1.)
Table 341.1
Differential Diagnosis of Oral Ulceration
CONDITION COMMENT
Common
Aphthous ulcers (canker sores) Painful circumscribed lesions; recurrences
Traumatic ulcers Accidents, chronic cheek biter, after dental local anesthesia
Hand, foot, and mouth disease Painful; lesions on tongue, anterior oral cavity, hands, and feet
Herpangina Painful; lesions confined to soft palate and oropharynx
Herpetic gingivostomatitis Vesicles on mucocutaneous borders; painful, febrile
Recurrent herpes labialis Vesicles on lips; painful
Chemical burns Alkali, acid, aspirin; painful
Heat burns Hot food, electrical
Uncommon
Neutrophil defects Agranulocytosis, leukemia, cyclic neutropenia; painful
Systemic lupus erythematosus Recurrent; may be painless
Behçet syndrome Resembles aphthous lesions; associated with genital ulcers, uveitis
Necrotizing ulcerative gingivostomatitis Vincent stomatitis; painful
Syphilis Chancre or gumma; painless
Oral Crohn disease Aphthous-like; painful
Histoplasmosis Lingual
Pemphigus May be isolated to the oral cavity
Stevens-Johnson syndrome May be isolated to or appear initially in the oral cavity
Herpetic Gingivostomatitis
After an initial incubation period of approximately 1 wk, the primary infection
with herpes simplex virus manifests as fever and malaise, usually in a child
younger than 5 yr (see Chapter 279 ). The oral cavity can show various
expressions, including the gingiva becoming erythematous, mucosal
hemorrhages, and clusters of small vesicles erupting throughout the mouth.
There is often involvement of the mucocutaneous margin and perioral skin (Fig.
341.2 ). The oral symptoms generally are accompanied by fever,
lymphadenopathy, and difficulty eating and drinking. The symptoms usually
regress within 2 wk without scarring. Fluids should be encouraged because the
child may become dehydrated. Analgesics and anesthetic rinses can make the
child more comfortable. Oral acyclovir, if taken within the first 3 days of
symptoms in immunocompetent patients, is beneficial in shortening the duration
of symptoms. Caution should be exercised to prevent autoinoculation, especially
of the eyes.
FIG. 344.1 A continuous tracing of esophageal motility showing 2
swallows, as indicated by the pharyngeal contraction associated with
relaxation of the upper esophageal sphincter (UES) and followed by
peristalsis in the body of the esophagus. The lower esophageal sphincter
(LES) also displays a transient relaxation (arrow) unassociated with a
swallow. There is an episode of gastroesophageal reflux (*) recorded by a
pH probe at the time of the transient LES relaxation. (Courtesy John Dent,
FRACP, PhD and Geoffrey Davidson, MD.)
Table 344.1
In addition to relaxing to move swallowed material past the GEJ into the
stomach, the LES normally relaxes to vent swallowed air or to allow retrograde
expulsion of material from the stomach. Perhaps as an extension of these
functions, the normal LES also permits physiologic reflux episodes, brief events
that occur approximately 5 times in the first postprandial hour, particularly in the
awake state, but are otherwise uncommon. Transient LES relaxation, not
associated with swallowing, is the major mechanism underlying pathologic
reflux (see Fig. 344.1 ).
The close linkage of the anatomy of the upper digestive and respiratory tracts
has mandated intricate functional protections of the respiratory tract during
retrograde movement of gastric contents as well as during swallowing. The
protective functions include the LES tone, the bolstering of the LES by the
surrounding diaphragmatic crura, and the backup protection of the UES tone.
Secondary peristalsis, akin to primary peristalsis but without an oral component,
originates in the upper esophagus, triggered mainly by GER, and thereby also
clears refluxed gastric contents from the esophagus. Another protective reflex is
the pharyngeal swallow (initiated above the esophagus, but without lingual
participation). Multiple levels of protection against aspiration include the
rhythmic coordination of swallowing and breathing and a series of protective
reflexes with esophagopharyngeal afferents and efferents that close the UES or
larynx. These reflexes include the esophago-UES contractile reflex, the
pharyngo-UES contractile reflex, the esophagoglottal closure reflex, and 2
pharyngoglottal adduction reflexes. The last 2 reflexes have chemoreceptors on
the laryngeal surface of the epiglottis and mechanoreceptors on the aryepiglottic
folds as their sites of stimulus. It is likely that interactions between the
esophagus and the respiratory tract, which cause extraesophageal manifestations
of gastroesophageal reflux disease (GERD), will be explained by subtle
abnormalities in these protective reflexes.
344.1
FIG. 345.3 H-type fistula (arrow) demonstrated in an infant after barium
swallow on frontal-oblique chest x-ray. The tracheal aspect of the fistula is
characteristically superior to the esophageal aspect. Barium is seen to
outline the tracheobronchial tree. (From Wyllie R, Hyams JS, editors:
Pediatric gastrointestinal and liver disease, ed 3, Philadelphia, 2006,
Saunders Elsevier, p. 299.)
Table 345.1
Clinical Aspects of Esophageal Developmental Anomalies
AGE AT PREDOMINANT
ANOMALY DIAGNOSIS TREATMENT
PRESENTATION SYMPTOMS
Isolated Newborns Regurgitation of Esophagogram* Surgery
atresia feedings Plain film: gasless
Aspiration abdomen
Atresia + Newborns Regurgitation of Esophagogram* Surgery
distal TEF feedings Plain film: gas-filled
Aspiration abdomen
H-type TEF Infants to adults Recurrent pneumonia Esophagogram* Surgery
Bronchiectasis Bronchoscopy †
Esophageal Infants to adults Dysphagia Esophagogram* Dilation ‡
stenosis Food impaction Endoscopy † Surgery §
Duplication Infants to adults Dyspnea, stridor, EUS* Surgery
cyst cough (infants) MRI/CT †
Dysphagia, chest pain
(adults)
Vascular Infants to adults Dyspnea, stridor, Esophagogram* Dietary
anomaly cough (infants) Angiography † modification ‡
Dysphagia (adults) MRI/CT/EUS Surgery §
Esophageal Children to adults Dysphagia Esophagogram* Dilation ‡
ring Endoscopy † Endoscopic
incision §
Esophageal Children to adults Dysphagia Esophagogram* Bougienage
web Endoscopy †
* Diagnostic test of choice.
†
Confirmatory test.
‡ Primary therapeutic approach.
Management
Initially, maintaining a patent airway, pre-operative proximal pouch
decompression to prevent aspiration of secretions, and use of antibiotics to
prevent consequent pneumonia are paramount. Prone positioning minimizes
movement of gastric secretions into a distal fistula, and esophageal suctioning
minimizes aspiration from a blind pouch. Endotracheal intubation with
mechanical ventilation is to be avoided if possible because it can worsen
distention of the stomach. Surgical ligation of the TEF and primary end-to-end
anastomosis of the esophagus via right-sided thoracotomy constitute the current
standard surgical approach. In the premature or otherwise complicated infant, a
primary closure may be delayed by temporizing with fistula ligation and
gastrostomy tube placement. If the gap between the atretic ends of the esophagus
is >3 to 4 cm (>3 vertebral bodies), primary repair cannot be done; options
include using gastric, jejunal, or colonic segments interposed as a neoesophagus.
Careful search must be undertaken for the common associated cardiac and other
locus is termed GERD1.
Table 349.1
Symptoms According to Age
Clinical Manifestations
Most of the common clinical manifestations of esophageal disease can signify
the presence of GERD and are generally thought to be mediated by the
pathogenesis involving acid GER (Table 349.2 ). Although less noxious for the
esophageal mucosa, nonacid reflux events are recognized to play an important
role in extraesophageal disease manifestations. Infantile reflux manifests more
often with regurgitation (especially postprandially), signs of esophagitis
(irritability, arching, choking, gagging, feeding aversion), and resulting failure to
thrive; symptoms resolve spontaneously in the majority of infants by 12-24 mo.
Older children can have regurgitation during the preschool years; this complaint
diminishes somewhat as children age, and complaints of abdominal and chest
pain supervene in later childhood and adolescence. Occasional children present
with food refusal or neck contortions (arching, turning of head) designated
Sandifer syndrome. The respiratory presentations are also age dependent:
GERD in infants may manifest as obstructive apnea or as stridor or lower airway
disease in which reflux complicates primary airway disease such as
laryngomalacia or bronchopulmonary dysplasia. Otitis media, sinusitis,
lymphoid hyperplasia, hoarseness, vocal cord nodules, and laryngeal edema have
all been associated with GERD. Airway manifestations in older children are
more commonly related to asthma or to otolaryngologic disease such as
laryngitis or sinusitis. Despite the high prevalence of GERD symptoms in
asthmatic children, data showing direction of causality are conflicting.
Table 349.2
Symptoms and Signs That May Be Associated With
Gastroesophageal Reflux
SYMPTOMS
Recurrent regurgitation with or without vomiting
Weight loss or poor weight gain
Irritability in infants
Ruminative behavior
Heartburn or chest pain
Hematemesis
Dysphagia, odynophagia
Wheezing
Stridor
Cough
Hoarseness
SIGNS
Esophagitis
Esophageal stricture
Barrett esophagus
Laryngeal/pharyngeal inflammation
Recurrent pneumonia
Anemia
Dental erosion
Feeding refusal
Dystonic neck posturing (Sandifer syndrome)
Apnea spells
Apparent life-threatening events
From Wyllie R, Hyams JS, Kay M, editors: Pediatric gastrointestinal and liver disease, ed 4,
Philadelphia, 2011, WB Saunders, Table 22.1, p. 235.
Diagnosis
For most of the typical GERD presentations, particularly in older children, a
thorough history and physical examination suffice initially to reach the
diagnosis. This initial evaluation aims to identify the pertinent positives in
support of GERD and its complications and the negatives that make other
diagnoses unlikely. The history may be facilitated and standardized by
questionnaires (e.g., the Infant Gastroesophageal Reflux Questionnaire, the I-
GERQ, and its derivative, the I-GERQ-R), which also permit quantitative scores
to be evaluated for their diagnostic discrimination and for evaluative assessment
of improvement or worsening of symptoms. The clinician should be alerted to
the possibility of other important diagnoses in the presence of any alarm or
warning signs : bilious emesis, frequent projectile emesis, gastrointestinal
bleeding, lethargy, organomegaly, abdominal distention, micro- or macrocephaly,
hepatosplenomegaly, failure to thrive, diarrhea, fever, bulging fontanelle, and
seizures. The important differential diagnoses to consider in the evaluation of an
infant or a child with chronic vomiting are milk and other food allergies,
eosinophilic esophagitis, pyloric stenosis, intestinal obstruction (especially
malrotation with intermittent volvulus), nonesophageal inflammatory diseases,
infections, inborn errors of metabolism, hydronephrosis, increased intracranial
pressure, rumination, and bulimia. Focused diagnostic testing, depending on the
presentation and the differential diagnosis, can then supplement the initial
examination.
Most of the esophageal tests are of some use in particular patients with
suspected GERD. Contrast (usually barium) radiographic study of the
esophagus and upper gastrointestinal tract is performed in children with
vomiting and dysphagia to evaluate for achalasia, esophageal strictures and
patients. It may also be an incidental finding in asymptomatic patients, notably
in those with EoE receiving topical swallowed corticosteroids. Esophageal viral
infections can also manifest in immunocompetent hosts as an acute febrile
illness. Infectious esophagitis, like other forms of esophageal inflammation,
occasionally progresses to esophageal stricture. Diagnosis of infectious
esophagitis is made by endoscopy, usually notable for white plaques in candida,
multiple superficial ulcers or volcano ulcers in herpes simplex virus, and single
deep ulcer in cytomegalovirus. Histopathologic examination solidifies the
diagnosis with the detection of yeast and pseudohyphae in Candida; tissue
invasion distinguishes esophagitis from mere colonization. Multinucleated giant
cells with intranuclear Cowdry type A (eosinophilic) and type B (ground glass
appearance) inclusions in HSV, and both intranuclear and intracytoplasmic
inclusions producing an owl's eye appearance in CMV are typically described.
Adding polymerase chain reaction, tissue-viral culture, and
immunocytochemistry enhances the diagnostic sensitivity and precision.
Treatment is with appropriate antimicrobial agents; azole therapy, particularly
oral fluconazole for Candida; oral acyclovir for HSV, and oral valganciclovir for
CMV, or alternatively intravenous ganciclovir in severe CMV disease.
Pill Esophagitis
This acute injury is produced by contact with a damaging agent. Medications
implicated in pill esophagitis include tetracycline, doxycycline, potassium
chloride, ferrous sulfate, nonsteroidal antiinflammatory medications, cloxacillin,
and alendronate (Table 350.1 ). Most often the offending tablet is ingested at
bedtime with inadequate water. This practice often produces acute discomfort
followed by progressive retrosternal pain, odynophagia, and dysphagia.
Endoscopy shows a focal lesion often localized to one of the anatomic narrowed
regions of the esophagus or to an unsuspected pathologic narrowing (Fig. 350.4
). Treatment is supportive; lacking much evidence, sucralfate, antacids, topical
anesthetics, and bland or liquid diets are often used. The offending pill may be
restarted after complete resolution of symptoms, if deemed necessary, though
with clear emphasis on ingestion with adequate volume of water, usually at least
4 oz.
Table 350.1
Medications Commonly Associated With Esophagitis or
Esophageal Injury
ANTIBIOTICS
Clindamycin
Doxycycline
Penicillin
Rifampin
Tetracycline
ANTIVIRAL AGENTS
Nelfinavir
Zalcitabine
Zidovudine
BISPHOSPHONATES
Alendronate
Etidronate
Pamidronate
CHEMOTHERAPEUTIC AGENTS
Bleomycin
Cytarabine
Dactinomycin
Daunorubicin
5-Fluorouracil
Methotrexate
Vincristine
NSAIDs
Aspirin
Ibuprofen
Naproxen
OTHER MEDICATIONS
Ascorbic acid
Ferrous sulfate
Lansoprazole
Multivitamins
Potassium chloride
Quinidine
Theophylline
From Katzka DA: Esophageal disorders caused by medications, trauma, and infection. In
Feldman M, Friedman LS, Brandt LJ, editors: Sleisenger and Fordtran's gastrointestinal and liver
disease , ed 10, 2016, Box 46.1.
coagulation necrosis and a somewhat protective thick eschar. They can produce
severe gastritis, and volatile acids can result in respiratory symptoms. Children
younger than 5 yr of age account for half of the cases of caustic ingestions, and
boys are far more often involved than girls.
Table 353.1
Ingestible Caustic Materials Around the House
OTHER
CATEGORY MOST DAMAGING AGENTS
AGENTS
Alkaline drain cleaners, milking machine Sodium or potassium hydroxide Ammonia
pipe cleaners Sodium
hypochlorite
Aluminum
particles
Acidic drain openers Hydrochloric acid
Sulfuric acid
Toilet cleaners Hydrochloric acid Ammonium
Sulfuric acid chloride
Phosphoric acid Sodium
Other acids hypochlorite
Oven and grill cleaners Sodium hydroxide
Perborate (borax)
Denture cleaners Persulfate (sulfur)
Hypochlorite (bleach)
Dishwasher detergent Sodium hydroxide
• Liquid Sodium hypochlorite
• Powdered Sodium carbonate
• Packaged
Bleach Sodium hypochlorite Ammonia salt
Swimming pool chemicals Acids, alkalis, chlorine
Battery acid (liquid) Sulfuric acid
Disk batteries Electric current Zinc or other
metal salts
Rust remover Hydrofluoric, phosphoric, oxalic, and other acids
Household delimers Phosphoric acid
Hydroxyacetic acid
Hydrochloric acid
Barbeque cleaners Sodium and potassium hydroxide
Glyphosate surfactant (RoundUp) acid Glyphosate herbicide Surfactants
Hair relaxer Sodium hydroxide
Weed killer Dichlorophenoxyacetate, ammonium phosphate,
propionic acid
From Wylie R, Hyams JS, Kay M, editors: Pediatric gastrointestinal and liver disease , ed 4,
Philadelphia, 2011, WB Saunders, Table 19.1, p. 198.
Source: National Library of Medicine: Health and safety information on household products
(website). http://householdproducts.nlm.nih.gov/
Caustic ingestions produce signs and symptoms such as vomiting, drooling,
refusal to drink, oral burns, dysphagia, dyspnea, abdominal pain, hematemesis,
and stridor. Twenty percent of patients develop esophageal strictures. Absence of
oropharyngeal lesions does not exclude the possibility of significant
esophagogastric injury, which can lead to perforation or stricture. The absence of
symptoms is usually associated with no or minimal lesions; hematemesis,
respiratory distress, or presence of at least 3 symptoms predicts severe lesions.
An upper endoscopy is recommended as the most efficient means of rapid
identification of tissue damage and must be undertaken in all symptomatic
children.
Dilution by water or milk is recommended as acute treatment, but
neutralization, induced emesis, and gastric lavage are contraindicated. Treatment
depends on the severity and extent of damage (Table 353.2 , Fig. 353.5 ).
Stricture risk is increased by circumferential ulcerations, white plaques, and
sloughing of the mucosa and is reported to occur in 70–100% of grade IIB and
grade III caustic esophagitis. Strictures can require treatment with dilation, and
in some severe cases, surgical resection and colon or small bowel interposition
are needed. Silicone stents (self-expanding) placed endoscopically after a
dilation procedure can be an alternative and conservative approach to the
management of strictures. Rare late cases of superimposed esophageal
carcinoma are reported. The role of corticosteroids is controversial; they are not
recommended in grade 1 burns, but they can reduce the risk of strictures in
more-advanced caustic esophagitis. Many centers also use proton pump
inhibitors as well as antibiotics in the initial treatment of caustic esophagitis on
the premise that reducing superinfection in the necrotic tissue bed will, in turn,
lower the risk of stricture formation. Studies examining the role of antibiotics in
caustic esophagitis have not reported a clinically significant benefit even in those
with grade 2 or greater severity of esophagitis.
Table 353.2
FIG. 353.5 Computed Tomography (CT) Grading of Corrosive Injuries of
the Esophagus and the Stomach. Grade 1, normal appearance; grade 2,
wall and soft tissue edema, increased wall enhancement (arrow) ; grade 3,
transmural necrosis with absent wall enhancement (arrow) . (From Chirica
M, Bonavina L, Kelly MD, et al: Caustic ingestion, Lancet 389:2041–2050,
2017, Fig. 1.)
Bibliography
Chirica M, Bonavina L, Kelly MD, et al. Caustic ingestion.
Lancet . 2017;389:2041–2050.
Contini S, Garatti M, Swarray-Deen A, et al. Corrosive
esophageal strictures in children: outcomes after timely or
pyloric canal is typical of congenital hypertrophic pyloric stenosis.
Differential Diagnosis
Gastric waves are occasionally visible in small, emaciated infants who do not
have pyloric stenosis. Infrequently, gastroesophageal reflux, with or without a
hiatal hernia, may be confused with pyloric stenosis. Gastroesophageal reflux
disease can be differentiated from pyloric stenosis by radiographic studies.
Adrenal insufficiency from the adrenogenital syndrome can simulate pyloric
stenosis, but the absence of a metabolic acidosis and elevated serum potassium
and urinary sodium concentrations of adrenal insufficiency aid in differentiation
(see Chapter 594 ). Inborn errors of metabolism can produce recurrent emesis
with alkalosis (urea cycle) or acidosis (organic acidemia) and lethargy, coma, or
seizures. Vomiting with diarrhea suggests gastroenteritis, but patients with
pyloric stenosis occasionally have diarrhea. Rarely, a pyloric membrane or
pyloric duplication results in projectile vomiting, visible peristalsis, and, in the
case of a duplication, a palpable mass (Table 355.1 ). Duodenal stenosis
proximal to the ampulla of Vater results in the clinical features of pyloric
stenosis but can be differentiated by the presence of a pyloric mass on physical
examination or ultrasonography.
Table 355.1
Anomalies of the Stomach
AGE AT SYMPTOMS
ANOMALY INCIDENCE TREATMENT
PRESENTATION AND SIGNS
Stomach
Gastric, antral, or 3/100,000, when combined Infancy Nonbilious emesis Gastroduodenostomy,
pyloric atresia with webs gastrojejunostomy
Pyloric or antral As above Any age Failure to thrive, Incision or excision,
membrane (web) emesis pyloroplasty
Microgastria Rare Infancy Emesis, Continuous-drip
malnutrition feedings or jejunal
reservoir pouch
Gastric Rare Any age Usually Usually unnecessary
diverticulum asymptomatic
Gastric duplication Rare; male:female, 1 : 2 Any age Abdominal mass, Excision or partial
emesis, gastrectomy
hematemesis;
peritonitis if
ruptured
Gastric teratoma Rare Any age Upper abdominal Resection
mass
Gastric volvulus Rare Any age Emesis, refusal to Reduction of
feed volvulus, anterior
gastropexy
Pyloric stenosis United States, 3/1,000 Infancy Non-bilious Pyloromyotomy
(infantile (range, 1-8/1,000 in various emesis
hypertrophic and regions); male:female, 4 : 1
adult forms)
Congenital absence Rare Childhood, Dyspepsia, if Usually unnecessary
of the pylorus adulthood symptomatic
Modified from Semrin MG, Russo MA: Anatomy, histology, and developmental anomalies of the
stomach and duodenum. In Feldman M, Friedman LS, Brandt LJ, editors: Sleisenger and
Fordtran's gastrointestinal and liver disease , ed 10, Philadelphia, Saunders, 2015, Table 48.1.
Treatment
The preoperative treatment is directed toward correcting the fluid, acid–base,
and electrolyte losses. Correction of the alkalosis is essential to prevent
postoperative apnea, which may be associated with anesthesia. Most infants can
be successfully rehydrated within 24 hr. Vomiting usually stops when the
stomach is empty, and only an occasional infant requires nasogastric suction.
The surgical procedure of choice is pyloromyotomy. The traditional Ramstedt
procedure is performed through a short transverse skin incision. The underlying
pyloric mass is cut longitudinally to the layer of the submucosa, and the incision
is closed. Laparoscopic technique is equally successful and in one study resulted
in a shorter time to full feedings and discharge from the hospital as well as
greater parental satisfaction. The success of laparoscopy depends on the skill of
the surgeon. Postoperative vomiting occurs in half the infants and is thought to
be secondary to edema of the pylorus at the incision site. In most infants,
feedings can be initiated within 12-24 hr after surgery and advanced to
maintenance oral feedings within 36-48 hr after surgery. Persistent vomiting
suggests an incomplete pyloromyotomy, gastritis, gastroesophageal reflux
disease, or another cause of the obstruction. The surgical treatment of pyloric
stenosis is curative, with an operative mortality of 0–0.5%. Endoscopic balloon
dilation has been successful in infants with persistent vomiting secondary to
incomplete pyloromyotomy.
Conservative management with nasoduodenal feedings is advisable in patients
who are not good surgical candidates. Oral and intravenous atropine sulfate
(pyloric muscle relaxant) has also been described when surgical expertise is not
CHAPTER 358
358.1
Chronic Intestinal Pseudoobstruction
Asim Maqbool, Kristin N. Fiorino, Chris A. Liacouras
Table 358.1
Causes of Secondary Chronic Intestinal
Pseudoobstruction in Children
Autoimmune
Autoimmune myositis
Autoimmune ganglionitis
Scleroderma
Endocrine
Diabetes mellitus
Hypoparathyroidism
Hypothyroidism
Gastrointestinal
Celiac disease
Eosinophilic gastroenteritis
Inflammatory bowel disease
Hematology/oncology
Multiple myeloma
Paraneoplastic syndromes
Pheochromocytoma
Sickle cell disease
Infection
Chagas disease
Cytomegalovirus
Epstein-Barr virus
Herpes zoster
JC virus
Kawasaki disease
Postviral neuropathy
Medications and toxins
Chemotherapy
Cyclopentolate and phenylephrine eye drops
Diltiazem and nifedipine
Fetal alcohol syndrome
Jellyfish envenomation
Opioid medications
Postanesthesia
Radiation injury
Mitochondrial disorders
Mitochondrial neurogastrointestinal encephalomyopathy
Musculoskeletal disorders
Ehlers-Danlos syndrome
Myotonic dystrophy
Duchenne muscular dystrophy
Rheumatology
Amyloidosis
Dermatomyositis
Polymyositis
Systemic lupus erythematous
From Bitton S, Markowitz JF: Ulcerative colitis in children and adolescents. In Wyllie R, Hyams
JS, Kay M, editors: Pediatric gastrointestinal and liver disease, 5th ed, Elsevier, 2016,
Philadelphia, Box. 44.3, p. 548.
Table 358.2
Table 358.3
Findings in Pseudoobstruction
GI
FINDINGS*
SEGMENT
Esophageal Abnormalities in approximately half of CIPO, although in some series up to 85% demonstrate
motility abnormalities
Decreased LES pressure
Failure of LES relaxation
Esophageal body: low-amplitude waves, poor propagation, tertiary waves, retrograde peristalsis,
occasionally aperistalsis
Gastric May be delayed
emptying
EGG Tachygastria or bradygastria may be seen
ADM Postprandial antral hypomotility is seen and correlates with delayed gastric emptying
Myopathic subtype: low-amplitude contractions, <10-20 mm Hg
Neuropathic subtype: contractions are uncoordinated, disorganized
Absence of fed response
Fasting MMC is absent, or MMC is abnormally propagated
Colonic Absence of gastrocolic reflex because there is no increased motility in response to a meal
ARM Normal rectoanal inhibitory reflex
* Findings can vary according to the segment(s) of the GI tract that are involved.
The initial focus is to rule out anatomic obstruction and to assess for bladder
involvement, because that is a frequent and significant extraintestinal
manifestation of concern. Manometric evidence of a normal migrating motor
complex and postprandial activity should redirect the diagnostic evaluation.
CIPO due to an intestinal myopathy may demonstrate manometry evidence of
low-amplitude contractions, whereas CIPO due to enteric neuropathy
demonstrates normal amplitude but poorly organized contractions (nonperistaltic
or tonic). Anorectal motility is normal and differentiates pseudoobstruction from
Hirschsprung disease. Full-thickness intestinal biopsy might show involvement
withholding of stool. An acute episode usually precedes the chronic course. This
acute event could include a social stressor such as initiation of toilet training,
birth of a sibling, starting daycare, or abuse. The acute episode may be a dietary
change from human milk to cow's milk, secondary to the change in the protein
and carbohydrate ratio or an allergy to cow's milk. Although iron has been
suspected of causing issues with cow's milk–related constipation, this has not
been consistently demonstrated or substantiated. The stool becomes firm,
smaller, and difficult to pass, resulting in anal irritation and often an anal fissure.
In toddlers, coercive or inappropriately early toilet training is a factor that can
initiate a pattern of stool retention. In older children, retentive constipation can
develop after entering a situation that makes stooling inconvenient such as
school. Because the passage of bowel movements is painful, voluntary
withholding of feces to avoid the painful stimulus develops.
Table 358.4
Rome IV Diagnostic Criteria for Defecatory Disorders in
Neonates and Toddlers
AGE
FGID CRITERIA REQUIREMENTS CRITERIA ELEMENTS
RANGE
Functional All pediatric Must include 1 month of ≥2 of the • 2 or fewer defections weekly
constipation age groups following in infants up to 4 months of • History of excessive stool retention
age: • History of hard/painful bowel
In toilet trained children, the following movements
additional criteria may be used: • History of large-diameter stools
• Presence of a large fecal mass in the
rectum
• At least 1 weekly episode of
incontinence after being toilet
trained
• History of large-diameter stools that
may clog the toilet
FGID, Functional gastrointestinal disorders.
Modified from Benninga MA, Faure C, Hyman PE, et al. Childhood functional gastrointestinal
disorders: neonate/toddler, Gastroenterology 150:1443–1455, 2016.
Table 358.5
Rome IV Diagnostic Criteria for Defecatory Disorders in
Children and Adolescents
AGE RANGE CRITERIA REQUIREMENTS CRITERIA ELEMENTS
Functional Developmental Must include ≥2 of the following ≥1/wk for • ≤ 2 defecations in the toilet per
Constipation age ≥4 yr 1 ≥1 mo with insufficient criteria to week
diagnose irritable bowel syndrome • ≥1 episode of fecal incontinence
per week
• History of retentive posturing or
excessive volitional stool retention
• History of painful or hard bowel
movements
• Presence of a large fecal mass in
the rectum
• History of large diameter stools that
can obstruct the toilet
• After appropriate evaluation,
symptoms cannot be fully
explained by another medical
condition
Nonretentive developmental ≥1-mo history of the following symptoms: • Defecation into places
Fecal age ≥4 yr inappropriate to the sociocultural
Incontinence context
• No evidence of fecal retention
• After appropriate evaluation,
symptoms cannot be fully
explained by another medical
condition
Modified from Hyams JS, Di Lorenzo C, Saps M, et al. Childhood functional gastrointestinal
disorders: child/adolescent, Gastroenterology 150:1456–1468, 2016.
Clinical Manifestations
When children have the urge to defecate, typical behaviors include contracting
the gluteal muscles by stiffening the legs while lying down, holding onto
furniture while standing, or squatting quietly in corners, waiting for the call to
stool to pass. The urge to defecate passes as the rectum accommodates to its
contents. A vicious cycle of retention develops, as increasingly larger volumes of
stool need to be expelled. Caregivers may misinterpret these activities as
straining, but it is withholding behavior. There is often a history of blood in the
stool noted with the passage of a large bowel movement. Findings suggestive of
underlying pathology include failure to thrive, weight loss, abdominal pain,
vomiting, or persistent anal fissure or fistula.
In functional constipation, daytime encopresis is common. Encopresis is
defined as voluntary or involuntary passage of feces into inappropriate places at
least once a mo for 3 consecutive months once a chronologic or developmental
age of 4 yr has been reached. Encopresis is not diagnosed when the behavior is
exclusively the result of the direct effects of a substance (e.g., laxatives) or a
reflexes. Spinal cord lesions can occur with overlying skin anomalies. Urinary
tract symptoms include recurrent urinary tract infection and enuresis. Children
with no evidence of abnormalities on physical examination rarely require
radiologic evaluation.
In refractory patients (intractable constipation), specialized testing should be
considered to rule out conditions such as hypothyroidism, hypocalcemia, lead
toxicity, celiac disease, and disorders of neuromuscular gastrointestinal
pathology (Table 358.6 ). Colonic transit studies using radiopaque markers or
scintigraphy techniques may be useful. Selected children can benefit from MRI
of the spine to identify an intraspinal process, motility studies to identify
underlying myopathic or neuropathic bowel abnormalities, or a contrast enema
to identify structural abnormalities. In patients with severe functional
constipation, water-soluble contrast enema reveals the presence of a mega
rectosigmoid (Fig. 358.3 ). Anorectal motility studies can demonstrate a pattern
of paradoxical contraction of the external anal sphincter during defecation,
which can be treated by behavior modification and biofeedback. Colonic
motility can guide therapy in refractory cases, demonstrating segmental
problems that might require surgical intervention.
Table 358.6
London Classification of Gastrointestinal Neuromuscular
Pathology
1. Neuropathies
1.1 Absent neurons
1.1.1 Aganglionosis*
1.2 Decreased numbers of neurons
1.2.1 Hypoganglionosis
1.3 Increased numbers of neurons
1.3.1 Ganglioneuromatosis †
1.3.2 IND, type B ‡
1.4 Degenerative neuropathy §
1.5 Inflammatory neuropathies
1.5.1 Lymphocytic ganglionitis ¶
1.5.2 Eosinophilic ganglionitis
1.6 Abnormal content in neurons
1.6.1 Intraneuronal nuclear inclusions
1.6.2 Megamitochondria
1.7 Abnormal neurochemical coding**
1.8 Relative immaturity of neurons
1.9 Abnormal enteric glia
1.9.1 Increased numbers of enteric glia
2. Myopathies
2.1 Muscularis propria malformations ††
2.2 Muscle cell degeneration
2.2.1 Degenerative leiomyopathy/ ‡‡
2.2.2 Inflammatory leiomyopathy
2.2.2.1 Lymphocytic leiomyositis
2.2.2.2 Eosinophilic leiomyositis
2.3 Muscle hyperplasia/hypertrophy
2.3.1 Muscularis mucosae hyperplasia
2.4 Abnormal content in myocytes
2.4.1 Filament protein abnormalities
2.4.1.1 Alpha-actin myopathy §§
2.4.1.2 Desmin myopathy
2.4.2 Inclusion bodies
2.4.2.1 Polyglucosan bodies
2.4.2.2 Amphophilic
2.4.2.3 Megamitochondria ¶¶
2.5 Abnormal supportive tissue
2.5.1 Atrophic desmosis***
3. ICC abnormalities (enteric mesenchymopathy)
3.1 Abnormal ICC networks †††
* Can include rare cases of non-Hirschsprung disease severe hypoplastic hypoganglionosis with
§ May occur with or without neuronal loss but is best regarded as a separate entity.
¶ May occur with neuronal degeneration and/or loss; lymphocytic epithelioganglionitis is a variant.
**
Includes neurotransmitter loss (e.g., reduced or absent expression) or loss of a neurochemically
defined functional subset of nerves (see text).
††
Includes absence, fusion, or additional muscle coats.
‡‡ Hollow visceral myopathy may be diagnosed in familial cases with other characteristic
phenotypic features; myopathy with autophagic activity and pink blush myopathy with nuclear
crowding are rare variants in which degenerative findings are less overt.
§§ Smooth muscle alpha-actin deficiency is best described, although deficiencies of other proteins
††† Generally reduced or absent ICC, although abnormal morphology also reported.
Table 358.7
Suggested Medications and Dosages for Disimpaction
Table 358.8
Suggested Medications and Dosages for Maintenance
Therapy of Constipation
Bibliography
Auth MKH, Vora R, Farrelly P, et al. Childhood constipation.
BMJ . 2012;345:38–43.
Bar-Maor JA, Eitan A. Determination of the normal position of
the anus (with reference to idiopathic constipation). J Pediatr
Gastroenterol Nutr . 1987;6:559–561.
Bekkali NLH, van den Berg MM, Dijkgraaf MGW, et al. Rectal
of stool. Some neonates pass meconium normally but subsequently present with
a history of chronic constipation. Failure to thrive with hypoproteinemia from
protein-losing enteropathy is a less common presentation because Hirschsprung
disease is usually recognized early in the course of the illness but has been
known to occur. Breastfed infants might not present as severely as formula-fed
infants.
Failure to pass stool leads to dilation of the proximal bowel and abdominal
distention. As the bowel dilates, intraluminal pressure increases, resulting in
decreased blood flow and deterioration of the mucosal barrier. Stasis allows
proliferation of bacteria, which can lead to enterocolitis (Clostridium difficile,
Staphylococcus aureus, anaerobes, coliforms) with associated diarrhea,
abdominal tenderness, sepsis, and signs of bowel obstruction. Red flags in the
neonatal period then include neonatal intestinal obstruction, bowel perforation,
delayed passage of meconium, abdominal distention relieved by digital rectal
stimulation or enemas, chronic severe constipation, and enterocolitis. Early
recognition of Hirschsprung disease before the onset of enterocolitis is essential
in reducing morbidity and mortality.
Hirschsprung disease in older patients must be distinguished from other
causes of abdominal distention and chronic constipation (see Tables 358.6 and
358.9 and Figs. 358.4 and 358.5 ). The history often reveals constipation starting
in infancy that has responded poorly to medical management. Failure to thrive is
not uncommon. Fecal incontinence, fecal urgency, and stool-withholding
behaviors are usually not present. Significant abdominal distention is unusual in
non-Hirschsprung related constipation, as is emesis. The abdomen is tympanitic
and distended, with a large fecal mass palpable in the left lower abdomen. Rectal
examination demonstrates a normally placed anus that easily allows entry of the
finger but feels snug. The rectum is usually empty of feces, and when the finger
is removed, there may be an explosive discharge of foul-smelling feces and gas.
The stools, when passed, can consist of small pellets, be ribbon-like, or have a
fluid consistency, unlike the large stools seen in patients with functional
constipation. Intermittent attacks of intestinal obstruction from retained feces
may be associated with pain and fever. Urinary retention with enlarged bladder
or hydronephrosis can occur secondary to urinary compression.
Table 358.9
Distinguishing Features of Hirschsprung Disease and
Functional Constipation
VARIABLE FUNCTIONAL HIRSCHSPRUNG DISEASE
HISTORY
Onset of After 2 yr of age At birth
constipation
Encopresis Common Very rare
Failure to thrive Uncommon Possible
Enterocolitis None Possible
Forced bowel Usual None
training
EXAMINATION
Abdominal Uncommon Common
distention
Poor weight gain Rare Common
Rectum Filled with stool Empty
Rectal examination Stool in rectum Explosive passage of stool
Malnutrition None Possible
INVESTIGATIONS
Anorectal Relaxation of internal anal sphincter Failure of internal anal sphincter relaxation
manometry
Rectal biopsy Normal No ganglion cells, increased acetylcholinesterase
staining
Barium enema Massive amounts of stool, no transition Transition zone, delayed evacuation (>24 hr)
zone
From Imseis E, Gariepy C: Hirschsprung disease. In Walker WA, Goulet OJ, Kleinman RE, et al,
editors: Pediatric gastrointestinal disease , ed 4, Hamilton, ON, 2004, BC Decker, p. 1035.
Table 361.1
Etiologic Classification of Peptic Ulcers
• Positive for Helicobacter pylori infection
• Drug (NSAID)-induced
• Helicobacter pylori and NSAID-positive
• H. pylori and NSAID-negative*
• Acid hypersecretory state (Zollinger-Ellison syndrome)
• Anastomosis ulcer after subtotal gastric resection
• Tumors (cancer, lymphoma)
• Rare specific causes
• Crohn disease of the stomach or duodenum
• Eosinophilic gastroduodenitis
• Systemic mastocytosis
• Radiation damage
• Viral infections (cytomegalovirus or herpes simplex infection, particularly in immunocompromised patients)
• Colonization of stomach with Helicobacter heilmannii
• Severe systemic disease
• Cameron ulcer (gastric ulcer where a hiatal hernia passes through the diaphragmatic hiatus)
• True idiopathic ulcer
* Requires search for other specific causes. NSAID, nonsteroidal antiinflammatory drug. (From
Vakil N, Megraud F: Eradication therapy for Helicobacter pylori, Gastroenterology 133:985–1001,
2007.)
Pathogenesis
Acid Secretion
By 3-4 yr of age, gastric acid secretion approximates adult values. Acid initially
secreted by the oxyntic cells of the stomach has a pH of approximately 0.8,
whereas the pH of the stomach contents is 1-2. Excessive acid secretion is
associated with a large parietal cell mass, hypersecretion by antral G cells, and
increased vagal tone, resulting in increased or sustained acid secretion in
response to meals and increased secretion during the night. The secretagogues
that promote gastric acid production include acetylcholine released by the vagus
nerve, histamine secreted by enterochromaffin cells, and gastrin released by the
G cells of the antrum. Mediators that decrease gastric acid secretion and enhance
protective mucin production include prostaglandins.
Mucosal Defense
A continuous layer of mucous gel that serves as a diffusion barrier to hydrogen
Table 361.2
Recommended Eradication Therapies for Helicobacter
pylori –Associated Disease in Children
Table 361.3
Antisecretory Therapy With Pediatric Dosages
FIG. 361.3 Rescue therapy for failed eradication of H. pylori . *Bismuth-
based therapy with tetracycline instead of amoxicillin if patients >8 yr.
Bismuth dose is 262 mg four times a day for patients 8–10 yr and 524 mg
four times a day for those >10 yr. (See Tables 361.2 and 361.3 .) In
adolescents, levofloxacin or tetracycline can be considered. High-dose
amoxicillin ranges from 750 mg twice a day for body weight 15–24 kg, 1000
mg twice a day for 25–34 kg, and 1500 mg twice a day for >35 kg. (Adapted
from Jones NL, Koletzko S, Goodman K, et al: Joint
ESPGHAN/NASPGHAN guidelines for the management of Helicobacter
pylori in children and adolescents, J Pediatr Gastroenterol Nutr 64:991–
1003, 2017.)
than dizygotic. The concordance rate in twins is higher in Crohn disease (36%)
than in ulcerative colitis (16%). Genetic disorders that have been associated with
IBD include Turner syndrome, the Hermansky-Pudlak syndrome, glycogen
storage disease type Ib, and various immunodeficiency disorders. In 2001, the
first IBD gene, NOD2, was identified through association mapping. A few
months later, the IBD 5 risk haplotype was identified. These early successes
were followed by a long period without notable risk factor discovery. Since
2006, the year of the first published genome-wide array study on IBD, there has
been an exponential growth in the set of validated genetic risk factors for IBD
(Table 362.1 ).
Table 362.1
Selection of Most Important Genes Associated With
Inflammatory Bowel Disease and the Most Commonly
Associated Physiological Functions and Pathways
Table 362.2
Table 362.3
Extraintestinal Complications of Inflammatory Bowel
Disease
MUSCULOSKELETAL
Peripheral arthritis
Granulomatous monoarthritis
Granulomatous synovitis
Rheumatoid arthritis
Sacroiliitis
Ankylosing spondylitis
Digital clubbing and hypertrophic osteoarthropathy
Periostitis
Osteoporosis, osteomalacia
Rhabdomyolysis
Pelvic osteomyelitis
Recurrent multifocal osteomyelitis
Relapsing polychondritis
SKIN AND MUCOUS MEMBRANES
Oral lesions
Cheilitis
Aphthous stomatitis, glossitis
Granulomatous oral Crohn disease
Inflammatory hyperplasia fissures and cobblestone mucosa
Peristomatitis vegetans
DERMATOLOGIC
Erythema nodosum
Pyoderma gangrenosum
Sweet syndrome
Metastatic Crohn disease
Psoriasis
Epidermolysis bullosa acquisita
Perianal skin tags
Polyarteritis nodosa
Melanoma and nonmelanoma skin cancers
OCULAR
Conjunctivitis
Uveitis, iritis
Episcleritis
Scleritis
Retrobulbar neuritis
Chorioretinitis with retinal detachment
Crohn keratopathy
Posterior segment abnormalities
Retinal vascular disease
BRONCHOPULMONARY
Chronic bronchitis with bronchiectasis
Chronic bronchitis with neutrophilic infiltrates
Fibrosing alveolitis
Pulmonary vasculitis
Small airway disease and bronchiolitis obliterans
Eosinophilic lung disease
Granulomatous lung disease
Tracheal obstruction
CARDIAC
Pleuropericarditis
Cardiomyopathy
Endocarditis
Myocarditis
MALNUTRITION
Decreased intake of food
• Inflammatory bowel disease
• Dietary restriction
Malabsorption
• Inflammatory bowel disease
• Bowel resection
• Bile salt depletion
• Bacterial overgrowth
Intestinal losses
• Electrolytes
• Minerals
• Nutrients
Increased caloric needs
• Inflammation
• Fever
HEMATOLOGIC/ONCOLOGIC
Anemia: iron deficiency (blood loss)
Vitamin B12 (ileal disease or resection, bacterial overgrowth, folate deficiency)
Anemia of chronic inflammation
Anaphylactoid purpura (Crohn disease)
Hyposplenism
Autoimmune hemolytic anemia
Coagulation abnormalities
Increased activation of coagulation factors
Activated fibrinolysis
Anticardiolipin antibody
Increased risk of arterial and venous thrombosis with cerebrovascular stroke, myocardial infarction, peripheral
arterial, and venous occlusions
Systemic lymphoma (nonenteric)
RENAL AND GENITOURINARY
Metabolic
• Urinary crystal formation (nephrolithiasis, uric acid, oxylate)
Hypokalemic nephropathy
Inflammation
• Retroperitoneal abscess
• Fibrosis with ureteral obstruction
• Fistula formation
Glomerulitis
Membrane nephritis
Renal amyloidosis, nephrotic syndrome
PANCREATITIS
Secondary to medications (sulfasalazine, 6-mercaptopurine, azathioprine, parenteral nutrition)
Ampullary Crohn disease
Granulomatous pancreatitis
Decreased pancreatic exocrine function
Sclerosing cholangitis with pancreatitis
HEPATOBILIARY
Primary sclerosing cholangitis
Small duct primary sclerosing cholangitis (pericholangitis)
Carcinoma of the bile ducts
Fatty infiltration of the liver
Cholelithiasis
Autoimmune hepatitis
ENDOCRINE AND METABOLIC
Growth failure, delayed sexual maturation
Thyroiditis
Osteoporosis, osteomalacia
NEUROLOGIC
Peripheral neuropathy
Meningitis
Vestibular dysfunction
Pseudotumor cerebri
Cerebral vasculitis
Migraine
Modified from Kugathasan S: Diarrhea. In Kliegman RM, Greenbaum LA, Lye PS, editors:
Practical strategies in pediatric diagnosis and therapy, ed 2, Philadelphia, 2004, WB Saunders, p
285.
362.1
Chronic Ulcerative Colitis
Clinical Manifestations
Blood, mucus, and pus in the stool as well as diarrhea are the typical
presentation of ulcerative colitis. Constipation may be observed in those with
proctitis. Symptoms such as tenesmus, urgency, cramping abdominal pain
(especially with bowel movements), and nocturnal bowel movements are
common. The mode of onset ranges from insidious with gradual progression of
symptoms to acute and fulminant (Table 362.4 and Figs. 362.1 and 362.2 ).
Fever, severe anemia, hypoalbuminemia, leukocytosis, and more than 5 bloody
stools per day for 5 days define fulminant colitis . Chronicity is an important
part of the diagnosis; it is difficult to know if a patient has a subacute, transient
infectious colitis or ulcerative colitis when a child has had 1-2 wk of symptoms.
Symptoms beyond this duration often prove to be secondary to IBD. Anorexia,
weight loss, and growth failure may be present, although these complications are
more typical of Crohn disease.
Table 362.4
Montreal Classification of Extent and Severity of Ulcerative
Colitis
• E1 (proctitis): inflammation limited to the rectum
• E2 (left-sided; distal): inflammation limited to the splenic flexure
• E3 (pancolitis): inflammation extends to the proximal splenic flexure
• S0 (remission): no symptoms
• S1 (mild): 4 or less stools per day (with or without blood), absence of systemic symptoms, normal inflammatory
markers
• S2 (moderate): 4 stools per day, minimum signs of systemic symptoms
• S3 (severe): 6 or more bloody stools per day, pulse rate of ≥90 beats/min, temperature ≥37.5°C (99.5°F),
hemoglobin concentration <105 g/L, erythrocyte sedimentation rate ≥30 mm/hr
E, extent; S, severity.
From Ordàs I, Eckmann L, Talamini M, et al: Ulcerative colitis, Lancet 380:1606–1616, 2012,
Panel 2, p 1610.
younger children. The gross appearance of the colitis or development of small
bowel disease eventually leads to the correct diagnosis; this can occur years after
the initial presentation.
Table 362.5
Infectious Agents Mimicking Inflammatory Bowel Disease
At the onset, the colitis of hemolytic uremic syndrome may be identical to that
of early ulcerative colitis. Ultimately, signs of microangiopathic hemolysis (the
presence of schistocytes on blood smear), thrombocytopenia, and subsequent
renal failure should confirm the diagnosis of hemolytic-uremic syndrome.
Although Henoch-Schönlein purpura can manifest as abdominal pain and bloody
stools, it is not usually associated with colitis. Behçet disease can be
distinguished by its typical features (see Chapter 186 ). Other considerations are
radiation proctitis, viral colitis in immunocompromised patients, and ischemic
colitis (Table 362.6 ). In infancy, dietary protein intolerance can be confused
with ulcerative colitis, although the former is a transient problem that resolves
on removal of the offending protein, and ulcerative colitis is extremely rare in
this age group. Hirschsprung disease can produce an enterocolitis before or
within months after surgical correction; this is unlikely to be confused with
ulcerative colitis.
Table 362.6
Chronic Inflammatory Bowel-Like Intestinal Disorders
Including Monogenetic Diseases
INFECTION (see Table 362.5 )
AIDS-Associated
Toxin
Immune–Inflammatory
Severe combined immunodeficiency diseases
Agammaglobulinemia
Chronic granulomatous disease
Wiskott-Aldrich syndrome
Common variable immunodeficiency diseases
Acquired immunodeficiency states
Dietary protein enterocolitis
Autoimmune polyendocrine syndrome type 1
Behçet disease
Lymphoid nodular hyperplasia
Eosinophilic gastroenteritis
Omenn syndrome
Graft-versus-host disease
IPEX (immune dysfunction, polyendocrinopathy, enteropathy, X-linked) syndromes
Interleukin-10 signaling defects
Autoimmune enteropathy*
Microscopic colitis
Hyperimmunoglobulin M syndrome
Hyperimmunoglobulin E syndromes
Mevalonate kinase deficiency
Familial Mediterranean fever
Phospholipase Cγ2 defects
IL10RA mutation
Familial hemophagocytic lymphohistiocytosis type 5
X-linked lymphoproliferative syndromes types 1, 2 (XIAP gene)
Congenital neutropenias
TRIM22 mutation
Leukocyte adhesion deficiency 1
VASCULAR–ISCHEMIC DISORDERS
Systemic vasculitis (systemic lupus erythematosus, dermatomyositis)
Henoch-Schönlein purpura
Hemolytic uremic syndrome
Granulomatosis with angiitis
OTHER
Glycogen storage disease type 1b
Dystrophic epidermolysis bullosa
X-linked ectodermal dysplasia and immunodeficiency
Dyskeratosis congenita
ADAM-17 deficiency
Prestenotic colitis
Diversion colitis
Kindler syndrome
Radiation colitis
Neonatal necrotizing enterocolitis
Typhlitis
Sarcoidosis
Hirschsprung colitis
Intestinal lymphoma
Laxative abuse
Endometriosis
Hermansky-Pudlak syndrome
Trichohepatoenteric syndrome
Phosphatase and tensin homolog (PTEN) hamartoma tumor syndrome
* May be the same as IPEX.
Diagnosis
The diagnosis of ulcerative colitis or ulcerative proctitis requires a typical
presentation in the absence of an identifiable specific cause (see Tables 362.5
and 362.6 ) and typical endoscopic and histologic findings (see Tables 362.2 and
362.4 ). One should be hesitant to make a diagnosis of ulcerative colitis in a
child who has experienced symptoms for <2-3 wk until infection has been
excluded. When the diagnosis is suspected in a child with subacute symptoms,
the physician should make a firm diagnosis only when there is evidence of
chronicity on colonic biopsy. Laboratory studies can demonstrate evidence of
ulcerative colitis and prevent recurrence. It is recommended that the medication
be continued even when the disorder is in remission. These medications might
also modestly decrease the lifetime risk of colon cancer.
Approximately 5% of patients have an allergic reaction to 5-ASA,
manifesting as rash, fever, and bloody diarrhea, which can be difficult to
distinguish from symptoms of a flare of ulcerative colitis. 5-ASA can also be
given in enema or suppository form and is especially useful for proctitis.
Hydrocortisone enemas are used to treat proctitis as well, but they are probably
not as effective. A combination of oral and rectal 5-ASA as well as monotherapy
with rectal preparation has been shown to be more effective than just oral 5-ASA
for distal colitis. Extended release budesonide may also induce remission in
patients with mild-to-moderate ulcerative colitis.
Probiotics are effective in adults for maintenance of remission for ulcerative
colitis, although they do not induce remission during an active flare. The most
promising role for probiotics has been to prevent pouchitis, a common
complication following colectomy and ileal–pouch anal anastomosis surgery.
Children with moderate to severe pancolitis or colitis that is unresponsive to
5-ASA therapy should be treated with corticosteroids, most commonly
prednisone. The usual starting dose of prednisone is 1-2 mg/kg/24 hr (40-60 mg
maximum dose). This medication can be given once daily. With severe colitis,
the dose can be divided twice daily and can be given intravenously. Steroids are
considered an effective medication for acute flares, but they are not appropriate
maintenance medications because of loss of effect and side effects, including
growth retardation, adrenal suppression, cataracts, osteopenia, aseptic necrosis of
the head of the femur, glucose intolerance, risk of infection, mood disturbance,
and cosmetic effects.
For a hospitalized patient with persistence of symptoms despite intravenous
steroid treatment for 3-5 days, escalation of therapy or surgical options should be
considered. The validated pediatric ulcerative colitis activity index can be used
to help determine current disease severity based on clinical factors and help
determine who is more likely to respond to steroids and those who will likely
require escalation of therapy (Table 362.7 ).
Table 362.7
Pediatric Ulcerative Colitis Activity Index
ITEM POINTS
(1) Abdominal Pain
No pain 0
Pain can be ignored 5
Pain cannot be ignored 10
(2) Rectal Bleeding
None 0
Small amount only, in <50% of stools 10
Small amount with most stools 20
Large amount (>50% of the stool content) 30
(3) Stool Consistency of Most Stools
Formed 0
Partially formed 5
Completely unformed 10
(4) Number of Stools Per 24 hr
0-2 0
3-5 5
6-8 10
>8 15
(5) Nocturnal Stools (Any Episode Causing Wakening)
No 0
Yes 10
(6) Activity Level
No limitation of activity 0
Occasional limitation of activity 5
Severe restricted activity 10
Sum of Index (0-85)
Table 362.8
Diagnosis
Crohn disease can manifest as a variety of symptom combinations (see Fig.
CHAPTER 364
Disorders of Malabsorption
Raanan Shamir
Table 364.1
Malabsorption Disorders and Chronic Diarrhea Associated
With Generalized Mucosal Defect
MUCOSAL DISORDERS
Gluten-sensitive enteropathy (celiac disease)
Cow's milk and other protein-sensitive enteropathies
Eosinophilic enteropathy
PROTEIN-LOSING ENTEROPATHY
Lymphangiectasia (congenital and acquired)
Disorders causing bowel mucosal inflammation, Crohn disease
CONGENITAL BOWEL MUCOSAL DEFECTS
Microvillous inclusion disease
Tufting enteropathy
Carbohydrate-deficient glycoprotein syndrome
Enterocyte heparan sulfate deficiency
Enteric anendocrinosis (NEUROG 3, PCSK1 mutations)
Tricho-hepatic-enteric syndrome
IMMUNODEFICIENCY DISORDERS
Congenital immunodeficiency disorders
Selective immunoglobulin A deficiency (can be associated with celiac disease)
Severe combined immunodeficiency
Agammaglobulinemia
X-linked hypogammaglobulinemia
Wiskott-Aldrich syndrome
Common variable immunodeficiency disease
Chronic granulomatous disease
ACQUIRED IMMUNE DEFICIENCY
HIV infection
Immunosuppressive therapy and post–bone marrow transplantation
AUTOIMMUNE ENTEROPATHY
IPEX (i mmune dysregulation, p olyendocrinopathy, e nteropathy, X -linked inheritance)
IPEX-like syndromes
Autoimmune polyglandular syndrome type 1
MISCELLANEOUS
Immunoproliferative small intestinal disease
Short bowel syndrome
Blind loop syndrome
Radiation enteritis
Protein–calorie malnutrition
Crohn disease
Pseudoobstruction
Table 364.2
Classification of Malabsorption Disorders and Chronic
Diarrhea Based on the Predominant Nutrient Malabsorbed
CARBOHYDRATE MALABSORPTION
Lactose malabsorption
Congenital lactase deficiency
Hypolactasia (adult type)
Secondary lactase deficiency
Congenital sucrase isomaltase deficiency
Glucose galactose malabsorption
FAT MALABSORPTION
Exocrine pancreatic insufficiency
Cystic fibrosis
Shwachman-Diamond syndrome
Johanson-Blizzard syndrome
Pearson syndrome
Secondary exocrine pancreatic insufficiency
Chronic pancreatitis
Protein–calorie malnutrition
Decreased pancreozymin/cholecystokinin secretion
Isolated enzyme deficiency
Enterokinase deficiency
Trypsinogen deficiency
Lipase/colipase deficiency
Disrupted enterohepatic circulation of bile salts
Cholestatic liver disease
Bile acid synthetic defects
Deconjugation of bile acids (bacterial overgrowth)
Bile acid malabsorption (terminal ileal disease)
Intestinal brush border disorders
Allergic enteropathy
Autoimmune enteropathy
Disorders in formation and transport of chylomicrons by enterocytes to the lymphatics
Abetalipoproteinemia
Homozygous hypobetalipoproteinemia
Chylomicron retention disease (Anderson disease)
Disorders of lymph flow
Lymphangiectasia primary/secondary
PROTEIN/AMINO ACID MALABSORPTION
Lysinuric protein intolerance (defect in dibasic amino acid transport)
Hartnup disease (defect in free neutral amino acids)
Blue diaper syndrome (isolated tryptophan malabsorption)
Oasthouse urine disease (defect in methionine absorption)
Lowe syndrome (lysine and arginine malabsorption)
Enterokinase deficiency
Protein-losing enteropathy
DGAT1 mutation
Congenital disorders of glycosylation
CD55 deficiency
MINERAL AND VITAMIN MALABSORPTION
Congenital chloride diarrhea
Congenital sodium absorption defect
Acrodermatitis enteropathica (zinc malabsorption)
Menkes disease (copper malabsorption)
Vitamin D–dependent rickets
Folate malabsorption
Congenital
Secondary to mucosal damage (celiac disease)
Vitamin B12 malabsorption
Autoimmune pernicious anemia
Decreased gastric acid (H2 blockers or proton pump inhibitors)
Terminal ileal disease (e.g., Crohn disease) or resection
Inborn errors of vitamin B12 transport and metabolism
Primary hypomagnesemia
DRUG INDUCED
Sulfasalazine: folic acid malabsorption
Cholestyramine: calcium and fat malabsorption
Anticonvulsant drugs such as phenytoin (causing vitamin D deficiency and folic acid and calcium malabsorption)
Gastric acid suppression: vitamin B12
Methotrexate: mucosal injury
Clinical Approach
The clinical features depend on the extent and type of the malabsorbed nutrient.
The common presenting features, especially in toddlers with malabsorption, are
diarrhea, abdominal distention, and failure to gain weight, with a fall in growth
chart percentiles. Physical findings include abdominal distention, muscle
wasting, and the disappearance of the subcutaneous fat, with subsequent loose
Severe PLE is often associated with malabsorption syndromes (CD, congenital
disorders of glycosylation, intestinal lymphangiectasia) and causes
hypoalbuminemia and edema. Other nutrient deficiencies include iron
malabsorption causing microcytic anemia and low reticulocyte count, low serum
folate levels in conditions associated with mucosal atrophy, especially in the
proximal part of the small intestinal tract and low serum vitamin A and vitamin
E concentrations in fat malabsorption.
The evaluation of a child with malabsorption should be proceed in a stepwise
manner. Clinical history alone might not be sufficient to make a specific
diagnosis, but it can direct the pediatrician toward a more structured and rational
investigative approach. Diarrhea is the main clinical expression of
malabsorption. The onset of diarrhea in early infancy suggests a congenital
defect (Table 364.3 ). In secretory diarrhea caused by disorders such as
congenital chloride diarrhea (CCD) and microvillus inclusion disease (MVID),
the stool is watery and voluminous and can be mistaken for urine (see Chapter
367 ). The onset of symptoms after the introduction of a particular food into a
child's diet can provide diagnostic clues, such as with sucrose in sucrase-
isomaltase deficiency. The nature of the diarrhea may be helpful: explosive
watery diarrhea suggests carbohydrate malabsorption; loose, bulky stools are
associated with CD; and pasty and yellowish offensive stools suggest an
exocrine pancreatic insufficiency. Stool color is usually not helpful; green stool
with undigested “peas and carrots” can suggest rapid intestinal transit in toddler's
diarrhea, which by itself is a self-limiting condition unassociated with failure to
thrive.
Table 364.3
364.1
Evaluation of Children With
Suspected Intestinal Malabsorption
Firas Rinawi, Raanan Shamir
Keywords
Steatorrhea
protein-losing enteropathy
carbohydrate malabsorption
Table 364.6
Clinical Spectrum of Celiac Disease
SYMPTOMATIC
Frank malabsorption symptoms and signs (e.g., chronic diarrhea, failure to thrive, weight loss)
Extraintestinal symptoms and signs (e.g., anemia, fatigue, hypertransaminasemia, neurologic disorders, short
stature, dental enamel defects, arthralgia, aphthous stomatitis)
SILENT
No apparent symptoms in spite of histologic evidence of villous atrophy
In most cases identified by serologic screening in at-risk groups (see Table 364.1 )
LATENT
Subjects who have a normal intestinal histology, but at some other time have shown a gluten-dependent
enteropathy
POTENTIAL
Subjects with positive celiac disease serology but without evidence of altered intestinal histology. Patients may or
may not have symptoms and signs of disease and may or may not develop a gluten-dependent enteropathy later
Diagnosis
The diagnosis of CD is based on a combination of symptoms, antibodies, HLA
status, and duodenal histology. The initial approach to symptomatic patients is to
test for anti-TG2 IgA antibodies and for total IgA in serum to exclude IgA
deficiency. If IgA anti-TG2 antibodies are negative, and serum total IgA is
normal for age, CD is unlikely to be the cause of the symptoms. If anti-TG2
antibody testing is positive the patients should be referred to a pediatric
gastroenterologist for further diagnostic workup, which depends on the serum
antibody levels.
IgA anti-TG2 decline if the patient is on a gluten free diet. In patients with
selective IgA deficiency, testing is recommended with IgG antibodies to TG2.
Patients with positive anti-TG2 antibody levels <10 times the upper limit of
normal should undergo upper endoscopy with multiple biopsies. In patients with
positive anti-TG2 antibody levels at or >10 times the upper limit of normal,
blood should be drawn for HLA and EMA testing. If the patient is positive for
EMA antibodies and positive for DQ2 or DQ8 HLA testing, the diagnosis of CD
is confirmed, a life-long gluten-free diet is started and the patient is followed for
the improvement of symptoms and the decline of antibodies. HLA testing is
almost always positive; thus, it is possible that HLA testing will not be necessary
in the future to establish diagnosis. In the rare case of negative results for HLA
and/or anti-EMA in a child with TG2 antibody titers >10 times the upper limits
of normal, the diagnostic workup should be extended, including repeated testing
and duodenal biopsies (Fig. 364.4 ). In asymptomatic persons belonging to high-
FIG. 364.5 Diagnostic algorithm for celiac disease (CD) in asymptomatic
children/adolescents belonging to at-risk groups, according to ESPGHAN.
EGD, Esophagogastroduodenoscopy; EMA , endomysial antibodies; Ig ,
immunoglobulin; HLA , human leukocyte antigen; TG2 , transglutaminase.
(Modified from Husby S, Koletzko S, Korponay-Szabò IR, et al: European
Society for Pediatric Gastroenterology, Hepatology and Nutrition Guidelines
for the diagnosis of celiac disease, J Pediatr Gastroenterol Nutr 54[1]:136–
160, 2012. Fig. 2.)
Table 364.7
Other Causes of Flat Mucosa
Autoimmune enteropathy
Tropical sprue
Giardiasis
HIV enteropathy
Bacterial overgrowth
Crohn disease
Eosinophilic gastroenteritis
Cow's milk enteropathy
Food allergy
Primary immunodeficiency
Graft-versus-host disease
Chemotherapy and radiation
Protein energy malnutrition
Treatment
The only treatment for CD is lifelong strict adherence to a gluten-free diet. This
requires a wheat-, barley-, and rye-free diet (Tables 364.8 and 364.9 ). Despite
evidence that oats are safe for most patients with CD, there is concern regarding
the possibility of contamination of oats with gluten during harvesting, milling,
and shipping. Nevertheless, it seems wise to add oats to the gluten-free diet only
when the latter is well established, so that possible adverse reactions can be
readily identified. There is a consensus that all CD patients should be treated
with a gluten-free diet regardless of the presence of symptoms. However,
whereas it is relatively easy to assess the health improvement after treatment of
CD in patients with clinical symptoms of the disease, it proves difficult in
persons with asymptomatic CD. The nutritional risks, particularly osteopenia
and increased risk for other autoimmune disorders, are those mainly feared for
subjects who have silent CD and continue on a gluten-containing diet. Little is
known about the health risks in untreated patients with potential CD.
Table 364.8
Principles of Initial Dietary Therapy for Patients With Celiac
Disease
Avoid all foods containing wheat, rye, and barley gluten (pure oats usually safe).
Avoid malt unless clearly labeled as derived from corn.
Use only rice, corn, maize, buckwheat, millet, amaranth, quinoa, sorghum, potato or potato starch, soybean,
tapioca, and teff, bean, and nut flours.
Wheat starch and products containing wheat starch should only be used if they contain <20 ppm gluten and are
marked “gluten free.”
Read all labels and study ingredients of processed foods.
Beware of gluten in medications, supplements, food additives, emulsifiers, or stabilizers.
Limit milk and milk products initially if there is evidence of lactose intolerance.
Avoid all beers, lagers, ales, and stouts (unless labeled gluten free).
Wine, most liqueurs, ciders, and spirits, including whiskey and brandy, are allowed.
Table 364.9
Some Potential Sources of Hidden Gluten
Beers, ales, other fermented beverages (distilled beverages acceptable)
Bouillon and soups
Candy
Communion wafers
Drink mixes
Gravy and sauces
Herbal tea
Imitation meat and seafood
Nutritional supplements
Play-Doh
Salad dressings and marinades
Self-basting turkeys
Soy sauce
From Kelly CP. Celiac disease. In Feldman M, Friedman LS, Brandt LJ, editors: Sleisenger and
Fordtran's gastrointestinal and liver disease , ed 10, Philadelphia, Elsevier, 2016. Box 107.3.
Short bowel (or short gut) syndrome results from congenital malformations or
resection of the small bowel (Table 364.11 ). Its incidence increases with low
birth weight and earlier gestational age and is estimated at 7/1,000 live births in
U.S. infants with birth weight <1,500 mg. Loss of >50% of the small bowel,
with or without a portion of the large intestine, can result in symptoms of
generalized malabsorptive disorder or in specific nutrient deficiencies,
depending on the region of the bowel resected. At birth, the length of small
bowel is 200-250 cm; by adulthood, it grows to 300-800 cm. Bowel resection in
an infant has a better prognosis than in an adult because of the potential for
intestinal growth and adaptation. An infant with as little as 15 cm of bowel with
an ileocecal valve, or 20 cm without, has the potential to survive and be
eventually weaned from parenteral nutrition.
Table 364.11
Causes of Short Bowel Syndrome
CONGENITAL
Congenital short bowel syndrome
Intestinal atresia
Gastroschisis
BOWEL RESECTION
Necrotizing enterocolitis
Volvulus with or without malrotation
Long segment Hirschsprung disease
Meconium peritonitis
Crohn disease
Trauma
In addition to the length of the bowel, the anatomic location of the resection is
also important. The proximal 100-200 cm of jejunum is the main site for
carbohydrate, protein, iron, and water-soluble vitamin absorption, whereas fat
absorption occurs over a longer length of the small bowel. Depending on the
The complications of intestinal failure include loss of venous access, life-
threatening infections, and TPN-induced cholestatic liver disease.
Table 365.1
Causes of Intestinal Failure in Children Requiring
Transplantation
SHORT BOWEL
• Congenital disorders
• Volvulus
• Gastroschisis
• Necrotizing enterocolitis
• Intestinal atresia
• Trauma
INTESTINAL DYSMOTILITY
• Intestinal pseudoobstruction
• Intestinal aganglionosis (Hirschsprung disease)
ENTEROCYTE DYSFUNCTION
• Microvillus inclusion disease
• Tufting enteropathy
• Autoimmune disorders
• Crohn disease
TUMORS
• Familial polyposis
• Inflammatory pseudotumor
Life-Threatening Infections
Life-threatening infections are usually catheter-related; the absence of significant
lengths of intestine may be associated with abnormal motility of the residual
bowel (producing both delayed or rapid emptying), with varying degrees of
bacterial overgrowth and possible bacterial or fungal translocation as a
consequence of loss of intestinal barrier function and/or loss of gut immunity.
CHAPTER 366
Table 366.1
Etiologies of Viral Gastroenteritis
COMMERCIALLY
ACUTE DURATION PRINCIPAL
INCUBATION RISK AVAILABLE
ETIOLOGY SIGNS AND OF VEHICLE AND
PERIOD FACTORS DIAGNOSTIC
SYMPTOMS ILLNESS TRANSMISSION
TEST
Caliciviruses 12-48 hr Nausea, 1-3 days Person-to-person Very No. Testing of
(including vomiting, (fecal-oral and contagious stool or vomitus
noroviruses abdominal aerosolized vomit), (chlorine using real time
and cramping, and food, water, and heat reverse
sapoviruses) diarrhea, and fomites resistant); transcriptase
fever, contaminated with produces (RT)-
myalgia, and human feces. large quantitative
some outbreaks in PCR is the
headache closed preferred
settings such method,
as cruise available in
ships, and public health
restaurants. laboratories.
Immunoassays
for norovirus
have poor
sensitivity.
FDA-cleared
multiplex PCR
assays are
available to
detect these
organisms.
Norovirus
genotyping (GI
and GII) is
performed by
CDC.
Rotavirus 2-4 days Often begins 3-8 days Person-to-person Nearly all Yes. Rotavirus:
(groups A- with (fecal-oral), infants and immunoassay
C), vomiting, fomites. Aerosol children (preferred), latex
astrovirus, followed by transmission of worldwide agglutination, and
and enteric watery rotavirus may be were immune-
adenovirus diarrhea, low- possible. infected by chromatography of
(serotypes 40 grade fever 2 yr of age stool. Enteric
and 41) before adenovirus:
vaccine immunoassay. FDA-
introduction. cleared multiplex
PCR assays are
available to detect
these organisms.
CDC, Centers for Disease Control and Prevention.
Modified from Centers for Disease Control and Prevention: Diagnosis and management of
foodborne illnesses, MMWR 53(RR-4):1–33, 2004.
Table 366.2
Etiologies of Bacterial Gastroenteritis
DURATION PRINCIPAL
INCUBATION ACUTE SIGNS
ETIOLOGY OF VEHICLE AND RISK FACTORS
PERIOD AND SYMPTOMS
ILLNESS TRANSMISSION
Bacillus cereus 1-6 hr Sudden onset of 24 hr Soil and water Improperly
(preformed emetic severe nausea and refrigerated cooked
toxin) vomiting; diarrhea or fried rice, meats
may be present
Bacillus cereus 8-16 hr Abdominal cramps, 1-2 days Soil and water Meats, stews,
(enterotoxins watery diarrhea; gravies, vanilla
formed in vivo) nausea and vomiting sauce
may be present
Campylobacter 1-5 days Diarrhea, (10–20% of 5-7 days Wild and domestic Raw and
jejuni episodes are (sometimes animals and animal undercooked
prolonged), cramps, >10 days) products, including poultry,
fever, and vomiting; usually self- pets unpasteurized milk,
bloody diarrhea, limiting untreated surface
bacteremia, water
extraintestinal
infections, severe
disease in
immunocompromised
Clostridium Unknown—can Mild to moderate Variable Person-person Immunosuppression,
difficile toxin appear weeks watery diarrhea that (fecal-oral), mostly intestinal disease or
after antibiotic can progress to within healthcare surgery, prolonged
cessation severe, facilities hospitalization,
pseudomembranous antibiotics
colitis with systemic
toxicity.
Clostridium 8-16 hr Watery diarrhea, 1-2 days Environment, Meats, poultry,
perfringens toxin nausea, abdominal human and animal gravy, dried or
cramps; fever is rare intestines precooked foods
with poor
temperature control
Enterohemorrhagic 1-9 days Watery diarrhea 4-7 days Food and water Undercooked beef
Escherichia coli (usually 3-4 that becomes contaminated with especially
(EHEC) including days) bloody in 1-4 feces from hamburger,
E. coli O157:H7 days in ~40% of ruminants; infected unpasteurized milk
and other Shiga infections; in people and animals and juice, raw fruits
toxin–producing contrast to (fecal-oral); and petting zoos,
E. coli (STEC) dysentery, bloody predominantly recreational
stools are large high-resource swimming, daycare.
volume and countries Antimotility agents
fever/toxicity are and antibiotics
minimal. increase risk of
More common in hemolytic uremic
children <4 yr syndrome
old.
Enterotoxigenic E. 1-5 days Watery diarrhea, 3-7 days Water or food Infants and young
coli (ETEC) abdominal cramps, contaminated with children in LMIC
some vomiting human feces and travelers
Salmonella , 1-5 days Diarrhea, (10–20% 5-7 days Domestic poultry, Ingestion of raw or
nontyphoidal prolonged), cramps, (sometimes cattle, reptiles, undercooked food,
fever, and vomiting; >10 days) amphibians, birds improper food
bloody diarrhea, usually self- handling, travelers,
bacteremia, limiting immunosuppression,
extraintestinal hemolytic anemia,
infections, severe achlorhydria,
disease in contact with
immunocompromised infected animal
Shigella spp. 1-5 days (up to Abdominal 5-7 days Infected people or Poor hygiene and
10 days for S. cramps, fever, fecally sanitation,
dysenteriae diarrhea contaminated crowding, travelers,
type 1) Begins with surfaces (fecal- daycare, MSM,
watery stools that oral) prisoners
can be the only
manifestation or
proceed to
dysentery.
Staphylococcus 1-6 hr Sudden onset of 1-3 days Birds, mammals, Unrefrigerated or
aureus (preformed severe nausea and dairy, and improperly
enterotoxin) vomiting environment refrigerated meats,
Abdominal potato and egg
cramps salads, cream
Diarrhea and pastries
fever may be
present
Vibrio cholerae O1 1-5 days Watery diarrhea and 3-7 days Food and water Contaminated water,
and O139 vomiting, that can be contaminated with fish, shellfish,
profuse and lead to human feces street-vended food
severe dehydration from endemic or
and death within epidemic settings;
hours. blood group O,
vitamin A
deficiency
Vibrio 2-48 hr Watery diarrhea, 2-5 days Estuaries and Undercooked or raw
parahaemolyticus abdominal cramps, marine seafood, such as
nausea, vomiting. environments; fish, shellfish
Bacteremia and currently
wound infections undergoing
occur uncommonly, pandemic spread
especially in high-risk
patients, e.g., with
liver disease and
diabetes.
Vibrio vulnificus 1-7 days Vomiting, diarrhea, 2-8 days Estuaries and Undercooked or raw
abdominal pain. marine shellfish, especially
Bacteremia and environments oysters, other
wound infections, contaminated
particularly in patients seafood, and open
with chronic liver wounds exposed to
disease (presents with seawater
septic shock and
hemorrhagic bullous
skin lesions)
Yersinia 1-5 days Diarrhea, (10–20% 5-7 days Swine products, Undercooked pork,
enterocolitica and prolonged), cramps, (sometimes occasionally improper food
Yersinia fever, and vomiting; >10 days) person-to-person handling,
pseudotuberculosis bloody diarrhea, usually self- and animal-to- unpasteurized milk,
bacteremia, limiting humans, water- tofu, contaminated
extraintestinal borne, bloodborne water, transfusion
infections, severe (can multiply from a bacteremic
disease in during person, cirrhosis,
immunocompromised; refrigeration) chelation therapy.
pseudoappendicitis
occurs primarily in
older children.
† FDA-cleared multiplex PCR assays are available but generally not recommended for diagnosis
Table 366.3
Etiologies of Parasitic Gastroenteritis
COMMERCIALLY
ACUTE SIGNS PRINCIPAL
INCUBATION DURATION RISK AVAILABLE
ETIOLOGY AND VEHICLE AND
PERIOD OF ILLNESS FACTORS DIAGNOSTIC
SYMPTOMS TRANSMISSION
TEST
Cryptosporidium 1-11 days Diarrhea 1-2 wk; may be Person-to-person Infants 6-18 mo Request specific
(usually watery), remitting and (fecal-oral), of age living in microscopic
bloating, relapsing over Contaminated food endemic examination of stool
flatulence, weeks to months and water settings in with special stains
cramps, (including LMIC, patients (direct fluorescent
malabsorption, municipal and with AIDS, antibody staining is
weight loss, and recreational water childcare preferable to
fatigue may wax contaminated with settings, modified acid fast)
and wane. human feces. drinking for Cryptosporidium.
Persons with unfiltered Immunoassays and
AIDS or surface water, PCR
malnutrition MSM, IgA sensitive than
have more deficiency microscopy.
severe disease.
Cyclospora 1-11 days Same as Same as Fresh produce Travelers,
cayetanensis Cryptosporidium Cryptosporidium (imported berries, consumption of
lettuce) fresh produce
imported from
the tropics.
IgA, Immunoglobulin A; LMID, low- and middle-income countries; MSM, men who have sex with
men; PCR, polymerase chain reaction.
Modified from Centers for Disease Control and Prevention: Diagnosis and management of
foodborne illnesses, MMWR 53(RR-4):1–33, 2004.
Table 366.4
Incidence of Bacterial and Parasitic Food-Borne Infections
in 2017 and Percentage Change Compared With 2014-2016
Average Annual Incidence by Pathogen FoodNet Sites,*
2014-2017 †
Table 366.5
Exposure or Condition Associated With Pathogens
Causing Diarrhea
EXPOSURE OR
PATHOGEN(S)
CONDITION
FoodBorne
Foodborne outbreaks in Norovirus, nontyphoidal Salmonella , Clostridium perfringens , Bacillus cereus ,
hotels, cruise ships, resorts, Staphylococcus aureus , Campylobacter spp., ETEC, STEC, Listeria , Shigella ,
restaurants, catered events Cyclospora cayetanensis , Cryptosporidium spp.
Consumption of Salmonella , Campylobacter , Yersinia enterocolitica , S. aureus toxin,
unpasteurized milk or dairy Cryptosporidium , and STEC. Listeria is infrequently associated with diarrhea,
products Brucella (goat milk cheese), Mycobacterium bovis , Coxiella burnetii
Consumption of raw or STEC (beef), C. perfringens (beef, poultry), Salmonella (poultry), Campylobacter
undercooked meat or poultry (poultry), Yersinia (pork, chitterlings), S. aureus (poultry), and Trichinella spp.
(pork, wild game meat)
Consumption of fruits or STEC, nontyphoidal Salmonella , Cyclospora , Cryptosporidium , norovirus,
unpasteurized fruit juices, hepatitis A, and Listeria monocytogenes
vegetables, leafy greens, and
sprouts
Consumption of Salmonella , Shigella (egg salad)
undercooked eggs
Consumption of raw Vibrio species, norovirus, hepatitis A, Plesiomonas
shellfish
Exposure or Contact
Swimming in or drinking Campylobacter , Cryptosporidium , Giardia , Shigella , Salmonella , STEC,
untreated fresh water Plesiomonas shigelloides
Swimming in recreational Cryptosporidium and other potentially waterborne pathogens when disinfectant
water facility with treated concentrations are inadequately maintained
water
Healthcare, long-term care, Norovirus, Clostridium difficile , Shigella , Cryptosporidium , Giardia , STEC,
prison exposure, or rotavirus
employment
Childcare center attendance Rotavirus, Cryptosporidium , Giardia , Shigella , STEC
or employment
Recent antimicrobial therapy C. difficile , multidrug-resistant Salmonella
Travel to resource- Escherichia coli (enteroaggregative, enterotoxigenic, enteroinvasive), Shigella ,
challenged countries typhi and nontyphoidal Salmonella , Campylobacter , Vibrio cholerae , Entamoeba
histolytica , Giardia , Blastocystis , Cyclospora , Cystoisospora , Cryptosporidium
Exposure to house pets with Campylobacter , Yersinia
diarrhea
Exposure to pig feces in Balantidium coli
certain parts of the world
Contact with young poultry Nontyphoidal Salmonella
or reptiles
Visiting a farm or petting STEC, Cryptosporidium , Campylobacter
zoo
Exposure or Condition
Age group Rotavirus (6-18 mo of age), nontyphoidal Salmonella (infants from birth to 3 mo of
age and adults >50 yr with a history of atherosclerosis), Shigella (1-7 yr of age),
Campylobacter (young adults)
Underlying Nontyphoidal Salmonella , Cryptosporidium , Campylobacter , Shigella , Yersinia
immunocompromising
condition
Hemochromatosis or Y. enterocolitica , Salmonella
hemoglobinopathy
AIDS, immunosuppressive Cryptosporidium , Cyclospora , Cystoisospora , microsporidia, Mycobacterium
therapies avium –intercellulare complex, cytomegalovirus
Anal-genital, oral-anal, or Shigella , Salmonella , Campylobacter , E. histolytica , Giardia lamblia ,
digital-anal contact Cryptosporidium
ETEC, enterotoxigenic Escherichia coli ; STEC, Shiga toxin–producing Escherichia coli .
From Shane AL, Mody RK, Crump JA, et al: 2017 Infectious Diseases Society for America clinical
practice guidelines for the diagnosis and management of infectious diarrhea, Clin Infect Dis
65(12):e45–80, 2017 (Table 2, p. e48).
Endemic Diarrhea. In the United States, rotavirus was the most common
cause of medically attended AGE among children younger than 5 yr until the
introduction of rotavirus vaccine for routine immunization of infants. Annual
epidemics swept across the country beginning in the southwest in November and
reaching the northeast by May, affecting nearly every child by the age of 2 yr.
Since vaccine introduction, healthcare utilization for AGE has decreased
markedly. Norovirus is the leading cause of AGE among children in the United
States seeking healthcare, followed by sapovirus, adenovirus 40 and 41, and
astrovirus (see Table 366.1 ).
Foodborne Transmission. The most comprehensive resource for describing
the burden of bacterial and protozoal diarrhea in the United States is the
Foodborne Diseases Active Surveillance Network (FoodNet) maintained by the
Centers for Disease Control and Prevention (CDC) (see Table 366.4 ). FoodNet
performs active laboratory-based surveillance of 9 bacterial and protozoal
enteric infections commonly transmitted by food. Among children 0-19 yr of age
in 2015, NTS was most common, followed by Campylobacter and Shigella ,
then STEC and Cryptosporidium . Vibrio, Yersinia, and Cyclospora were the
least common (see Table 366.5 ). Children younger than 5 yr have the highest
incidence of disease, and the elderly have the highest frequency of
hospitalization and death. Only 5% of these infections are associated with
recognized outbreaks.
Noninfectious agents may also cause foodborne gastrointestinal symptoms
due to a direct toxic effect of the food (mushrooms) or contamination (heavy
metals) (Table 366.6 ).
Table 366.6
Foodborne Noninfectious Illnesses
DURATION
INCUBATION SIGNS AND ASSOCIATED LABORATORY
ETIOLOGY OF TREATMENT
PERIOD SYMPTOMS FOODS TESTING
ILLNESS
Antimony 5 min-8 hr Vomiting, Usually self- Metallic Identification of Supportive care
usually <1 hr metallic taste limited container metal in
beverage or food
Arsenic Few hours Vomiting, colic, Several days Contaminated Urine Gastric lavage,
diarrhea food Can cause BAL
eosinophilia (dimercaprol)
Cadmium 5 min-8 hr Nausea, Usually self- Seafood, Identification of Supportive care
usually <1 hr vomiting, limited oysters, clams, metal in food
myalgia, lobster, grains,
increase in peanuts
salivation,
stomach pain
Ciguatera fish 2-6 hr GI: abdominal Days to A variety of Radioassay for Supportive
poisoning pain, nausea, weeks to large reef fish: toxin in fish or a care, IV
(ciguatera toxin) vomiting, months grouper, red consistent mannitol
diarrhea snapper, history Children
amberjack, and more
barracuda vulnerable
(most common)
3 hr Neurologic:
paresthesias,
reversal of hot
or cold, pain,
weakness
2-5 days Cardiovascular:
bradycardia,
hypotension,
increase in T-
wave
abnormalities
Copper 5 min-8 hr Nausea, Usually self- Metallic Identification of Supportive care
usually <1 hr vomiting, blue limited container metal in
or green beverage or food
vomitus
Mercury 1 wk or longer Numbness, May be Fish exposed to Analysis of Supportive care
weakness protracted organic blood, hair
of legs, mercury, grains
spastic treated with
paralysis, mercury
impaired fungicides
vision,
blindness,
coma
Pregnant
women and
the
developing
fetus are
especially
vulnerable
Mushroom toxins, <2 hr Vomiting, Self-limited Wild Typical Supportive care
short-acting diarrhea, mushrooms syndrome and
(muscimol, confusion, (cooking might mushroom
muscarine, visual not destroy identified or
psilocybin, disturbance, these toxins) demonstration of
Coprinus salivation, the toxin
atramentaria , diaphoresis,
ibotenic acid) hallucinations,
disulfiram-like
reaction,
confusion,
visual
disturbance
Mushroom toxins, 4-8 hr diarrhea; Diarrhea, Often fatal Mushrooms Typical Supportive
long-acting 24-48 hr liver abdominal syndrome and care, life-
(amanitin) failure cramps, leading mushroom threatening,
to hepatic and identified and/or may need life
renal failure demonstration of support
the toxin
Nitrite poisoning 1-2 hr Nausea, Usually self- Cured meats, Analysis of the Supportive
vomiting, limited any food, blood care, methylene
cyanosis, contaminated blue
headache, foods, spinach
dizziness, exposed to
weakness, loss excessive
of nitrification
consciousness,
chocolate-
brown blood
Pesticides Few minutes to Nausea, Usually self- Any Analysis of the Atropine; 2-
(organophosphates few hours vomiting, limited contaminated food, blood PAM
or carbamates) abdominal food (pralidoxime) is
cramps, used when
diarrhea, atropine is not
headache, able to control
nervousness, symptoms;
blurred vision, rarely
twitching, necessary in
convulsions, carbamate
salivation, poisoning
meiosis
Puffer fish <30 min Paresthesias, Death Puffer fish Detection of Life-
(tetrodotoxin) vomiting, usually in 4- tetrodotoxin in threatening,
diarrhea, 6 hr fish may need
abdominal pain, respiratory
ascending support
paralysis,
respiratory
failure
Scombroid 1 min-3 hr Flushing, rash, 3-6 hr Fish: bluefin, Demonstration Supportive
(histamine) burning tuna, skipjack, of histamine in care,
sensation of mackerel, food or clinical antihistamines
skin, mouth and marlin, escolar, diagnosis
throat, and mahi
dizziness,
urticaria,
paresthesias
Shellfish toxins Diarrheic Nausea, Hours to 2-3 A variety of Detection of the Supportive
(diarrheic, shellfish vomiting, days shellfish, toxin in shellfish; care, generally
neurotoxic, poisoning: 30 diarrhea, and primarily high-pressure self-limiting
amnesic) min-2 hr abdominal pain mussels, liquid
accompanied oysters, chromatography
by chills, scallops, and
headache, and shellfish from
fever the Florida
coast and the
Gulf of Mexico
Neurotoxic Tingling and
shellfish numbness of
poisoning: few lips, tongue,
minutes to and throat,
hours muscular aches,
dizziness,
reversal of the
sensations of
hot and cold,
diarrhea, and
vomiting
Amnesic Vomiting, Elderly are
shellfish diarrhea, especially
poisoning: 24- abdominal pain sensitive to
48 hr and neurologic amnesic
problems such shellfish
as confusion, poisoning
memory loss,
disorientation,
seizure, coma
Shellfish toxins 30 min-3 hr Diarrhea, Days Scallops, Detection of Life-
(paralytic shellfish nausea, mussels, clams, toxin in food or threatening,
poisoning) vomiting cockles water where fish may need
leading to are located; high- respiratory
paresthesias of pressure liquid support
mouth and lips, chromatography
weakness,
dysphasia,
dysphonia,
respiratory
paralysis
Sodium fluoride Few minutes to Salty or soapy Usually self- Dry foods (e.g., Testing of Supportive care
2 hr taste, numbness limited dry milk, flour, vomitus or
of mouth, baking powder, gastric
vomiting, cake mixes) washings
diarrhea, dilated contaminated Analysis of
pupils, spasms, with NaF- the food
pallor, shock, containing
collapse insecticides and
rodenticides
Thallium Few hours Nausea, Several days Contaminated Urine, hair Supportive care
vomiting, food
diarrhea,
painful
paresthesias,
motor
polyneuropathy,
hair loss
Tin 5 min-8 hr Nausea, Usually self- Metallic Analysis of the Supportive care
usually <1 hr vomiting, limited container food
diarrhea
Vomitoxin Few minutes to Nausea, Usually self- Grains such as Analysis of the Supportive care
3 hr headache, limited wheat, corn, food
abdominal pain, barley
vomiting
Zinc Few hours Stomach Usually self- Metallic Analysis of the Supportive care
cramps, nausea, limited container food, blood and
vomiting, feces, saliva or
diarrhea, urine
myalgias
BAL, bronchoalveolar lavage; GI, gastrointestinal.
From Centers for Disease Control and Prevention: Diagnosis and management of foodborne
illnesses, MMWR 53(RR-4):1–33, 2004.
Table 366.7
Proven Risk Factors With Direct Biologic Links to Diarrhea:
Relative Risks or Odds Ratios and 95% Confidence
Intervals
Table 366.8
Comparison of 3 General Pathogenic Mechanisms of
Enteric Infection
TYPE OF INFECTION
PARAMETER
I II III
Mechanism Noninflammatory Inflammatory, epithelial Penetrating
(enterotoxin or destruction (invasion,
adherence/superficial cytotoxin)
invasion)
Location Proximal small bowel Colon Distal small bowel
Illness Watery diarrhea Dysentery Enteric fever
Stool No fecal leukocytes Fecal Fecal mononuclear leukocytes
examination Mild or no ↑ lactoferrin polymorphonuclear
leukocytes
↑↑ Lactoferrin
Examples Vibrio cholerae Shigella Yersinia enterocolitica
ETEC EIEC Salmonella Typhi, S. Paratyhpi,
Clostridium perfringens STEC and occasionally NTS,
Bacillus cereus NTS Campylobacter, and Yersinia
Staphylococcus aureus Vibrio
Also † : parahaemolyticus
Giardia intestinalis Clostridium difficile
Rotavirus Campylobacter
Noroviruses jejuni
Cryptosporidium spp. Entamoeba
EPEC, EAEC histolytica *
Cyclospora
cayetanensis
* Although amebic dysentery involves tissue inflammation, the leukocytes are characteristically
Viral AGE causes a cytolytic infection of the small intestinal villus tips
resulting in decreased absorption of water, disaccharide malabsorption,
inflammation, and cytokine activation. The rotavirus protein NSP4 acts as a viral
enterotoxin that produces secretory diarrhea. In addition, rotavirus activates the
enteric nervous system causing decreased gastric emptying and increased
intestinal mobility. There is a genetic susceptibility to both rotavirus and
norovirus infection that is mediated by histo-blood group antigens on the
epithelial cell surface and in mucus secretions (Fig. 366.2 ).
FIG. 366.5 Pathogenesis of shigella infection and diarrhea. IL-8, interleukin-8. (Modified
from Opal SM, Keusch GT: Host responses to infection. In Cohen J, Powderly WG, Opal
SM, et al, editors: Infectious diseases, ed 2, London, 2004, Mosby, pp. 31–52.)
Table 366.9
Clinical Presentations Suggestive of Infectious Diarrhea Etiologies
FINDING LIKELY PATHOGENS
Persistent or chronic diarrhea Cryptosporidium spp., Giardia lamblia , Cyclospora cayetanensis , Entamoeba
histolytica , non-typhoidal Salmonella, Yersinia, and Campylobacter spp.
Visible blood in stool STEC, Shigella , Salmonella , Campylobacter , Entamoeba histolytica ,
noncholera Vibrio parahaemolyticus , Yersinia , Balantidium coli , and
Aeromonas
Fever Not highly discriminatory—viral, bacterial, and parasitic infections can cause
fever. In general, higher temperatures are suggestive of bacterial etiology or E.
histolytica . Patients infected with STEC usually are not febrile at time of
presentation
Abdominal pain STEC, Salmonella , Shigella , Campylobacter , Yersinia , noncholera Vibrio
species, Clostridium difficile
Severe abdominal pain, often STEC, Salmonella , Shigella , Campylobacter , and Yersinia enterocolitica
grossly bloody stools
(occasionally nonbloody), and
minimal or no fever
Persistent abdominal pain and Y. enterocolitica and Y. pseudotuberculosis ; may mimic appendicitis
fever
Nausea and vomiting lasting ≤24 Ingestion of Staphylococcus aureus enterotoxin or Bacillus cereus (short-
hr incubation emetic syndrome)
Diarrhea and abdominal Ingestion of Clostridium perfringens or Bacillus cereus (long-incubation emetic
cramping lasting 1-2 days syndrome)
Vomiting and nonbloody Norovirus (low-grade fever usually present during the first 24 hr in 40% of
diarrhea infections); diarrhea usually lasts 2-3 days or less; other viral diarrheas (e.g.,
rotavirus, enteric adenovirus, sapovirus, astrovirus) usually last 3-8 days.
Chronic watery diarrhea, often Brainerd diarrhea (epidemic secretory diarrhea, etiologic agent has not been
lasting a year or more identified); postinfectious irritable bowel syndrome
STEC, Shiga toxin–producing Escherichia coli .
From Shane AL, Mody RK, Crump JA, et al: 2017 Infectious Diseases Society for America clinical
practice guidelines for the diagnosis and management of infectious diarrhea, Clin Infect Dis
65(12):e45–80, 2017 (Table 3, p. e54).
In the past, many guidelines divided patients into subgroups for mild (3–5%),
moderate (6–9%), and severe (≥10%) dehydration. However, it is difficult to
distinguish between mild and moderate dehydration based on clinical signs
alone. Therefore most guidelines now combine mild and moderate dehydration
and simply use none, some, and severe dehydration. The individual signs that
best predict dehydration are prolonged capillary refill time >2 sec, abnormal skin
turgor, hyperpnea (deep, rapid breathing suggesting acidosis), dry mucous
membranes, absent tears, and general appearance (including activity level and
thirst). As the number of signs increases, so does the likelihood of dehydration.
Tachycardia, altered level of consciousness, and cold extremities with or without
hypotension suggest severe dehydration.
Viral Diarrhea. Symptoms of rotavirus AGE usually begin with vomiting
followed by frequent passage of watery nonbloody stools, associated with fever
A and B primarily affect preschool and school-age children in endemic
countries, other bacterial enteropathogens most often cause disease in
infants (particularly <3 mo), the immunocompromised, and children
with malnutrition. Additional risk factors include hemolytic anemia and
intravascular lesions for NTS, and iron overload, cirrhosis, and chelation
therapy for Yersinia sepsis. The distinct clones of NTS that have arisen
in sub-Saharan Africa described earlier are causing enteric fever–type
illnesses often in the absence of AGE. Shigella sepsis is rare and is seen
most often in malnourished and immunocompromised hosts.
4. Extraintestinal invasive infections can result from either local invasion or
bacteremic spread (Table 366.10 ). Examples of local invasion include
mesenteric adenitis, appendicitis, and rarely cholecystitis, mesenteric
venous thrombosis, pancreatitis, hepatic, or splenic abscess. Bacteremic
spread may result in pneumonia, osteomyelitis, meningitis (3 conditions
seen most commonly with NTS), abscesses, cellulitis, septic arthritis,
and endocarditis. Shigella can cause noninvasive contiguous infections
such as vaginitis and urinary tract infections.
Table 366.10
Intestinal and Extraintestinal Complications of Enteric Infections
ASSOCIATED ENTERIC
COMPLICATION
PATHOGEN(S)
Intestinal Complications
Persistent diarrhea All causes
Recurrent diarrhea (usually immunocompromised Salmonella, Shigella , Yersinia
persons) , Campylobacter, Clostridium
Toxic megacolon difficile , Entamoeba
Intestinal perforation histolytica , Cryptosporidium,
Rectal prolapse Giardia
Enteritis necroticans–jejunal hemorrhagic necrosis Shigella , C. difficile , E.
histolytica
Shigella , Yersinia, C. difficile
, E. histolytica
Shigella , STEC, C. difficile
Clostridium perfringens type
C beta toxin
Extraintestinal Complications
Dehydration, metabolic abnormalities, malnutrition, All causes
micronutrient deficiency Nontyphoidal Salmonella,
Bacteremia with systemic spread of bacterial Shigella, Yersinia,
pathogens, including endocarditis, osteomyelitis, Campylobacter
meningitis, pneumonia, hepatitis, peritonitis,
chorioamnionitis, soft tissue infection, and septic
thrombophlebitis
Local spread (e.g., vulvovaginitis and urinary tract Shigella
infection) Yersinia, Campylobacter
Pseudoappendicitis (occasionally)
Exudative pharyngitis, cervical adenopathy Yersinia
Rhabdomyolysis and hepatic necrosis Bacillus cereus emetic toxin
Postinfectious Complications
Reactive arthritis Salmonella, Shigella, Yersinia,
Campylobacter, Cryptosporidium,
C. difficile
Guillain-Barré syndrome Campylobacter
Hemolytic uremic syndrome STEC, Shigella dysenteriae 1
Glomerulonephritis, myocarditis, pericarditis Shigella, Campylobacter, Yersinia
Immunoglobulin A (IgA) nephropathy Campylobacter
Erythema nodosum Yersinia, Campylobacter,
Salmonella
Hemolytic anemia Campylobacter, Yersinia
Intestinal perforation Salmonella, Shigella,
Campylobacter, Yersinia,
Entamoeba histolytica
Osteomyelitis, meningitis, aortitis Salmonella, Yersinia, Listeria
STEC, Shiga toxin–producing Escherichia coli.
From Centers for Disease Control and Prevention: Managing acute gastroenteritis
among children, MMWR Recomm Rep 53:1–33, 2004.
Crampy abdominal pain and nonbloody diarrhea are the first symptoms of
STEC infection, sometimes with vomiting. Within several days, diarrhea
becomes bloody and abdominal pain worsens. Bloody diarrhea lasts between 1
and 22 days (median 4 days). In contrast to dysentery, the stools associated with
STEC hemorrhagic colitis are large volume and rarely accompanied by high
fever. ETEC produce a secretory watery diarrhea that affects infants and young
children in developing countries and is the major causative agents of travelers’
diarrhea, accounting for about half of all episodes in some studies. EPEC
remains a leading cause of persistent diarrhea associated with malnutrition
among infants from developing countries. EIEC, which are genetically,
vitamin deficiencies. Infection with Entamoeba can cause severe ulcerating
colitis, colonic dilation, and perforation. The parasite may spread systemically,
most commonly causing liver abscesses. In high-risk settings, it is critical to
exclude Entamoeba infection and tuberculosis before initiating corticosteroids
for presumed ulcerative colitis.
Differential Diagnosis
The physician should also consider noninfectious diseases that can present with
bright red blood per rectum or hematochezia (Table 366.11 ). In an infant or
young child without systemic symptoms, these may include anal fissures,
intermittent intussusception, juvenile polyps, and Meckel diverticulum.
Necrotizing enterocolitis can cause lower gastrointestinal bleeding in infants,
especially premature neonates. Inflammatory bowel disease should be
considered in older children. Examples of noninfectious causes of nonbloody
diarrhea include congenital secretory diarrheas, endocrine disorders
(hyperthyroidism), neoplasms, food intolerance, and medications (particularly
antibiotics). Noninfectious causes of chronic or relapsing diarrhea include cystic
fibrosis, celiac disease, milk protein intolerance, and congenital or acquired
disaccharidase deficiency. Significant abdominal pain should raise suspicion of
other infectious processes in the abdomen such as appendicitis and pelvic
inflammatory disease. Prominent vomiting with or without abdominal pain can
be a manifestation of pyloric stenosis, intestinal obstruction, pancreatitis,
appendicitis, and cholecystitis.
Table 366.11
Differential Diagnosis of Acute Dysentery and
Inflammatory Enterocolitis
Specific Infectious Processes
• Bacillary dysentery (Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Shigella boydii; invasive
Escherichia coli)
• Campylobacteriosis (Campylobacter jejuni)
• Amebic dysentery (Entamoeba histolytica)
• Ciliary dysentery (Balantidium coli)
• Bilharzial dysentery (Schistosoma japonicum, Schistosoma mansoni)
• Other parasitic infections (Trichinella spiralis)
• Vibriosis (Vibrio parahaemolyticus)
• Salmonellosis (Salmonella typhimurium)
• Typhoid fever (Salmonella typhi)
• Enteric fever (Salmonella choleraesuis, Salmonella paratyphi)
• Yersiniosis (Yersinia enterocolitica)
• Spirillar dysentery (Spirillum spp.)
Proctitis
• Gonococcal (Neisseria gonorrhoeae)
• Herpetic (herpes simplex virus)
• Chlamydial (Chlamydia trachomatis)
• Syphilitic (Treponema pallidum)
Other Syndromes
• Necrotizing enterocolitis of the newborn
• Enteritis necroticans
• Pseudomembranous enterocolitis (Clostridium difficile)
• Typhlitis
Chronic Inflammatory Processes
• Enteropathogenic and enteroaggregative E. coli
• Gastrointestinal tuberculosis
• Gastrointestinal mycosis
• Parasitic enteritis
Syndromes Without Known Infectious Cause
• Idiopathic ulcerative colitis
• Celiac disease
• Crohn disease
• Radiation enteritis
• Ischemic colitis
• Immune deficiency including HIV infection
• Allergic enteritis
From Mandell GL, Bennett JE, Dolin R, editors: Principles and practices of infectious diseases ,
ed 7, Philadelphia, 2010, Churchill Livingstone.
Important elements of the medical history include the duration of diarrhea and
a description of stools (frequency, amount, presence of blood or mucus), fever
(duration, magnitude), vomiting (onset, amount and frequency), and the amount
and type of solid and liquid oral intake. Clinical signs of dehydration should be
evaluated (Table 366.12 ): urine output (number of wet diapers per day and time
since the last urination), whether eyes appear sunken, whether the child is active,
whether the child drinks vigorously, and the date and value of the most recent
weight measurement. A documented weight loss can be used to calculate the
fluid deficit. The past medical history should identify comorbidities that might
increase the risk or severity of AGE.
Table 366.12
Clinical Signs Associated With Dehydration
MINIMAL OR NO
SYMPTOM SOME DEHYDRATION SEVERE DEHYDRATION
DEHYDRATION
Mental status Well; alert Normal, fatigued or Apathetic, lethargic, unconscious
restless, irritable
Thirst Drinks normally; might Thirsty; eager to drink Drinks poorly; unable to drink
refuse liquids
Heart rate Normal Normal to increased Tachycardia, with bradycardia in most
severe cases
Quality of Normal Normal to decreased Weak, thready, or impalpable
pulses
Breathing Normal Normal; fast Deep
Eyes Normal Slightly sunken Deeply sunken
Tears Present Decreased Absent
Mouth and Moist Dry Parched
tongue
Skinfold Instant recoil Recoil in <2 sec Recoil in >2 sec
Capillary Normal Prolonged Prolonged; minimal
refill
Extremities Warm Cool Cold; mottled; cyanotic
Urine output Normal to decreased Decreased Minimal
Modified from Duggan C, Santosham M, Glass RI: The management of acute diarrhea in children:
oral rehydration, maintenance, and nutritional therapy, MMWR Recomm Rep 41(RR-16):1–20,
1992; and World Health Organization: The treatment of diarrhoea: a manual for physicians and
other senior health workers, Geneva, 1995, World Health Organization; and Centers for Disease
Control and Prevention: Diagnosis and management of foodborne illnesses, MMWR 53(RR-4):1–
33, 2004.
Certain physical signs are best assessed before approaching the child directly,
so he/she remains calm, including general appearance (activity, response to
stimulation) and respiratory patterns. Skin turgor is assessed by pinching a small
skin fold on the lateral abdominal wall at the level of the umbilicus. If the fold
does not promptly return to normal after release, the recoil time is quantified as
delayed slightly or ≥2 sec. Excess subcutaneous tissue and hypernatremia may
produce a false negative test and malnutrition can prolong the recoil time. To
measure capillary refill time, the palmar surface of the child's distal fingertip is
pressed until blanching occurs, with the child's arm at heart level. The time
elapsed until restoration of normal color after release usually exceeds 2 sec in
the presence of dehydration. Mucous membrane moisture level, presence of
tears, and extremity temperature should be assessed.
Laboratory Diagnosis
Most cases of AGE do not require diagnostic laboratory testing. Stool specimens
could be examined for mucus, blood, neutrophils or fecal lactoferrin, a
neutrophil product. The finding of more than 5 leukocytes per high-power field
or a positive lactoferrin assay in an infant not breastfeeding suggests an infection
with a classical bacterial enteropathogen; patients infected with STEC and E.
histolytica usually have negative tests.
Laboratory diagnosis of viral AGE may be helpful when an outbreak is
suspected, cases are linked to a suspected outbreak, or when cohorting of
immunocompromised, or have hemolytic anemia or other risk factors. If diarrhea
persists with no cause identified, endoscopic evaluation may be indicated.
Biopsy specimens help in diagnosing inflammatory bowel disease or identifying
infecting agents that may mimic it. A sweat test is warranted if cystic fibrosis is
suspected.
Treatment
The broad principles of management of AGE in children include rehydration and
maintenance ORS plus replacement of continued losses in diarrheal stools and
vomitus after rehydration, continued breastfeeding, and refeeding with an age-
appropriate, unrestricted diet as soon as dehydration is corrected. Zinc
supplementation is recommended for children in developing countries.
Hydration
Children, especially infants, are more susceptible than adults to dehydration
because of the greater basal fluid and electrolyte requirements per kilogram and
because they are dependent on others to meet these demands (Table 366.13 ).
Dehydration must be evaluated rapidly and corrected in 4-6 hr according to the
degree of dehydration and estimated daily requirements. When there is emesis,
small volumes of ORS can be given initially by a dropper, teaspoon, or syringe,
beginning with as little as 5 mL at a time. The volume is increased as tolerated.
The low-osmolality World Health Organization (WHO) ORS containing 75 mEq
of sodium, 64 mEq of chloride, 20 mEq of potassium, and 75 mmol of glucose
per liter, with total osmolarity of 245 mOsm/L, is now the global standard of
care and more effective than home fluids. Soda beverages, fruit juices, tea, and
other home fluids are not suitable for rehydration or maintenance therapy
because they have inappropriately high glucose concentration and osmolalities
and low sodium concentrations. Tables 366.12 and 366.13 outline a clinical
evaluation plan and management strategy for children with moderate to severe
diarrhea. Replacement for emesis or stool losses is noted in Table 366.13 . Oral
rehydration can also be given by a nasogastric tube if needed; this is not the
usual route.
Table 366.13
Low-osmolarity ORS can be given to all age groups, with any cause of diarrhea. It is safe in the
presence of hypernatremia, as well as hyponatremia (except when edema is present). Some
commercially available formulations that can be used as ORS include Pedialyte Liters (Abbott
Nutrition), CeraLyte (Cero Products), and Enfalac Lytren (Mead Johnson). Popular beverages that
should not be used for rehydration include apple juice, Gatorade, and commercial soft drinks.
ORS, oral rehydration solution.
Modified from Centers for Disease Control and Prevention: Managing acute gastroenteritis among
children: oral rehydration, maintenance, and nutritional therapy. MMWR Recomm Rep 52(RR-
16):1–16, 2003; and World Health Organization. Pocket book of hospital care for children:
Guidelines for the management of common childhood illnesses, ed 2
(http://www.who.int/maternal_child_adolescent/documents/child_hospital_care/en/ ).
Antibiotic Therapy
Judicious antibiotic therapy for suspected or proven bacterial infections can
reduce the duration and severity of illness and prevent complications (Table
366.14 ). Several factors justify limited use. First, most episodes of AGE are
self-limited among otherwise healthy children. Second, the increasing
prevalence of antibiotic resistance has prompted restricted use of these drugs.
Third, antibiotics may worsen outcome, because some studies have shown that
antibiotic therapy of STEC infection increases the risk of HUS and prolongs
excretion of NTS without improving clinical outcome. Therefore antibiotics are
used primarily to treat severe infections, prevent complications in high-risk
hosts, or to limit the spread of infection. Microbiologic (culture) confirmation of
the etiology and susceptibility testing should be sought prior to treatment if
possible.
Table 366.14
Antibiotic Therapy for Infectious Diarrhea
artemether. These drugs can prolong the QT interval on the electrocardiogram and trigger
arrhythmias.
EIEC, enteroinvasive Escherichia coli ; EPEC, enteropathogenic E. coli ; ETEC, enterotoxigenic
E. coli ; GI, gastrointestinal; IV, intravenous; IM , intramuscular; max, maximum; SMX,
sulfamethoxazole; TMP, trimethoprim; WHO , World Health Organization.
Etiology
Table 367.1 summarizes the main etiologies of chronic diarrhea in infants and
children.
Table 367.1
Main Etiologies of Chronic Diarrhea in Children Older and Younger Than 2 Yr of Age
YOUNGER OLDER
ETIOLOGY
THAN 2 YR THAN 2 YR
Infections +++ +++
Postenteritis syndrome +++ +++
Immune deficiency ++ Rare
Celiac disease +++ (after gluten +++
introduction)
Food allergy +++ +
Inflammatory bowel disease + (rare) +++
Pancreatic insufficiency ++ ++
Cholestasis and insufficient bile acids ++ ++
Cystic fibrosis ++ +
Lactose intolerance ++ (mostly +++
postinfectious)
Intestinal lymphangiectasia + +
Motility disorders ++ Rare
Short bowel syndrome +++ +
Short bowel syndrome +++ +
Toddler's and functional diarrhea ++ ++
Excessive intake of fruit juices and fluids ++ ++
Congenital diarrheal disorders, including structural enterocyte defects and ++ Unlikely
enzymatic or transport malabsorption syndromes
Infectious
Enteric infections are by far the most frequent cause of persistent or chronic
diarrhea, both in developing and industrialized countries, however outcomes are
often very different. In the former, comorbid conditions, such as HIV/AIDS,
malaria, or tuberculosis, result in malnutrition that impairs the child's immune
response, thereby potentiating the likelihood of prolonging diarrhea or acquiring
another enteric infection. In children with HIV/AIDS, the viral infection itself
impairs immune function and may trigger a vicious circle with malnutrition.
Sequential infections with the same or different pathogens may also be
responsible for chronic diarrhea. In developing countries, enteroadherent
Escherichia coli and Giardia lamblia have been implicated in chronic diarrhea,
whereas, in developed countries, chronic infectious diarrhea usually runs a more
benign course and the etiology is often viral, with a major role of rotavirus and
norovirus (Table 367.2 ).
Table 367.2
A Comparative List of Prevalent Agents and Conditions in
Children With Persistent Infectious Diarrhea in
Industrialized and Developing Countries
AGENT/DISEASE
INDUSTRIALIZED COUNTRIES DEVELOPING COUNTRIES
Clostridium difficile Enteroaggregative E. coli
Enteroaggregative Escherichia coli Atypical E. coli
Shigella
Heat stable/heat labile enterotoxin-producing E. coli
Astrovirus Rotavirus*
Norovirus Cryptosporidium
Rotavirus* Giardia lamblia
Small intestinal bacterial overgrowth (SIBO) Tropical sprue
Postenteritis diarrhea syndrome
* More frequent in industrialized than in developing countries as agent of chronic diarrhea.
Table 367.3
Classification of Congenital Diarrheal Disorders Based on
Their Molecular Defect and Their Inheritance
Although CDDs are rare diseases, in most specific disorders the genetic defect
and transmission are known. The incidence of genetic disorders associated with
CDD can range from 1 in 2,500 for cystic fibrosis, 1 in 5,000 for sucrose-
isomaltase deficiency, 1 in 60,000 for congenital lactase deficiency, to 1 in
400,000 for trichohepatoenteric syndrome. For most CDDs, such as IPEX
syndrome or autoimmune polyglandular syndrome type 1, the clinical
application of exome sequencing is likely to increase identification of more
patients with these rare causes of chronic diarrhea. Selected CDDs are more
manifestations (arthritis, diabetes, thrombocytopenia, etc.) may suggest an
autoimmune disease. Specific skin lesions may be suggestive of acrodermatitis
enteropathica that might respond to zinc supplementation. Typical facial
abnormalities and woolly hair are associated with phenotypic diarrhea.
Investigations
Microbiologic investigation should include a thorough list of intestinal bacterial,
viral, and protozoan pathogens. Proximal intestinal bacterial overgrowth may be
determined using the lactulose hydrogen breath test, but false-positive tests are
common (see Chapter 364.4 ).
Initial investigations of a child with chronic diarrhea should always include an
assessment of intestinal inflammation using fecal calprotectin or lactoferrin, and
serology for celiac disease (see Chapter 364.2 ). The role of a mucosal biopsy is
determined by the noninvasive diagnostic evaluation in consultation with a
pediatric gastroenterologist.
Noninvasive assessment of digestive-absorptive function and of intestinal
inflammation plays a key role in the diagnostic workup (Table 367.4 ).
Abnormalities in the digestive-absorptive function tests suggest small bowel
involvement, whereas intestinal inflammation, as demonstrated by increased
fecal calprotectin or lactoferrin, supports colitis.
Table 367.4
Table 367.5
Stepwise Diagnostic Approach to Children and Infants With
Chronic Diarrhea
INITIAL EVALUATION
• Personal and family history: Prenatal sonography; feeding • Infectious workup: Stool cultures; parasites;
history; family history of protracted diarrhea; viruses
consanguinity • Allergic workup: Elimination diet trial
• Physical examination: Dysmorphism; skeletal
abnormalities; organomegaly; dermatitis
⇩
LABORATORY TESTS
• Stool analysis: Stool volume following fasting; stool • Blood and serum analysis: Serum electrolytes;
electrolytes and ion gap; pH and reducing substances; lipid profile; albumin and pre-albumin; amylase
steatocrit; fecal leukocytes and calprotectin; fecal and lipase; inflammatory markers; ammonia;
elastase; α 1 -antitrypsin celiac serology
⇩
IMAGING
• Abdominal ultrasound: Bowel wall thickening; liver and • X-ray and contrast studies: Congenital
bile disorders malformation; signs of motility disorders
⇩
ENDOSCOPIES AND INTESTINAL HISTOLOGY
Endoscopy and standard jejunal/colonic histology* ; morphometry; PAS staining; intestinal
immunohistochemistry; electron microscopy
⇩
GENETIC INVESTIGATION
• Specific molecular analysis • Whole-exome sequencing
⇩
OTHER SPECIAL INVESTIGATIONS
Sweat test; specific carbohydrates breath tests; 75 SeHCAT measurement; antienterocyte antibodies; metabolic
diseases workup; motility studies; neuroendocrine tumor markers
*
The decision to perform an upper or a lower endoscopy may be supported by noninvasive tests.
PAS , Periodic acid–Schiff; 75 SeHCAT , 75 Se-homocholic acid-taurine.
Treatment
Chronic diarrhea associated with impaired nutritional status should always be
considered a serious disease, and therapy should be started promptly. Treatment
includes general supportive measures, nutritional rehabilitation, elimination diet,
and medications. The latter include therapies for specific etiologies as well as
interventions aimed at counteracting fluid secretion and/or promoting restoration
of disrupted intestinal epithelium. Because death in most instances is caused by
dehydration, replacement of fluid and electrolyte losses is the most important
early intervention.
Nutritional rehabilitation is often essential and is based on clinical and
biochemical assessment. In moderate to severe malnutrition, caloric intake
should be carefully advanced to avoid the development of refeeding syndrome
and may be progressively increased to 50% or more above the recommended
dietary allowances. The intestinal absorptive capacity should be monitored by
digestive function tests. In children with steatorrhea, medium-chain triglycerides
may be the main source of lipids. A lactose-free diet should be started in all
Table 367.6
Antimicrobial Treatment for Persistent Diarrhea
Table 367.7
Diarrhea Caused by Neuroendocrine Tumors
SIGNS OF
TUMOR AND
SITE MARKERS HORMONE THERAPY
CELL TYPE
HYPERSECRETION
Carcinoid Intestinal Serotonin (5- Secretory diarrhea, Resection
argentaffin cells, HT), urine 5- crampy abdominal Somatostatin
typically midgut, HIAA * pain, flushing, analog,
also foregut and (diagnostic) wheezing (and cardiac (palliative)
hindgut, ectopic Also produce valve damage if Genetic MEN-1
bronchial tree substance P, foregut site)
neuropeptide K,
somatostatin,
VIP
Chromogranin
A
Gastrinoma, Pancreas, small Gastrin Multiple peptic ulcers, H2 -blockers,
Zollinger-Ellison bowel, liver, and secretory diarrhea PPI, tumor
syndrome spleen resection,
(gastrectomy)
Genetic MEN-1
Mastocytoma Cutaneous, Histamine , VIP Pruritus, flushing, H1 - and H2 -
intestine, liver, apnea blockers, steroids,
spleen If VIP, diarrhea resection if solitary
Medullary carcinoma Thyroid C-cells Calcitonin , VIP , Secretory diarrhea Radical
prostaglandins thyroidectomy ±
lymphadenectomy
(genetic MEN-
2A/B, familial
MTC)
Ganglioneuroma, Chromaffin cells; Metanephrines Hypertension, Perioperative α-
pheochromocytoma, abdominal > other and tachycardia, adrenergic (BP)
ganglioneuroblastoma, sites; extraadrenal catecholamines paroxysmal and β-
neuroblastoma or adrenal , VIP palpitations, sweating, adrenergic
VMA, HMA in anxiety, watery blockade with
neuroblastoma diarrhea † volume support
tumor resection
Genetic MEN-2
(RET gene),
VHL, NF-1,
SDH
Somatostatinoma Pancreas Somatostatin Secretory diarrhea, Resection
steatorrhea, Genetic MEN-1
cholelithiasis, diabetes
VIPoma Pancreas VIP , Secretory diarrhea, Somatostatin
prostaglandins achlorhydria, analogs,
hypokalemia resection
Genetic MEN-1
* Bold indicates major markers.
FIG. 368.1 Age distribution of functional gastrointestinal disorders in
infants, toddlers, children, and adolescents. *History may not be reliable
below this age. FAP-NOS , Functional abdominal pain—not otherwise
specified. (Modified from Benninga MA, Nurko S, Faure C, et al: Childhood
functional gastrointestinal disorders: neonate/toddler, Gastroenterology
150[6]:1443–1455.e2, 2016.)
Table 368.1
Diagnostic Criteria for Infant Regurgitation
Must include both of the following in otherwise healthy infants 3 wk to 12 mo of age:
1. Regurgitation 2 or more times per day for 3 or more wk
2. No retching, hematemesis, aspiration, apnea, failure to thrive, feeding or swallowing difficulties, or abnormal
posturing
From Benninga MA, Nurko S, Faure C, et al: Childhood functional gastrointestinal disorders:
neonate/toddler, Gastroenterology 150(6):1443–1455.e2, 2016.
Table 368.2
Diagnostic Criteria for Infant Rumination Syndrome
Must include all of the following for at least 2 mo:
1. Repetitive contractions of the abdominal muscles, diaphragm, and tongue
2. Effortless regurgitation of gastric contents, which are either expelled from the mouth or rechewed and
reswallowed
3. Three or more of the following:
a. Onset between 3 and 8 mo
b. Does not respond to management for gastroesophageal reflux disease and regurgitation
c. Unaccompanied by signs of distress
d. Does not occur during sleep and when the infant is interacting with individuals in the environment
From Benninga MA, Nurko S, Faure C, et al: Childhood functional gastrointestinal disorders:
neonate/toddler, Gastroenterology 150(6):1443–1455.e2, 2016.
Table 368.3
Diagnostic Criteria for Infant Colic
For clinical purposes, must include all of the following:
1. An infant who is < 5 mo of age when the symptoms start and stop
2. Recurrent and prolonged periods of infant crying, fussing, or irritability reported by caregivers that occur
without obvious cause and cannot be prevented or resolved by caregivers
3. No evidence of infant failure to thrive, fever, or illness
“Fussing” refers to intermittent distressed vocalization and has been defined as “[behavior] that is not quite crying
but not awake and content either.” Infants often fluctuate between crying and fussing, so that the 2 symptoms are
difficult to distinguish in practice.
For clinical research purposes, a diagnosis of infant colic must meet the preceding diagnostic criteria and also
include both of the following:
1. Caregiver reports infant has cried or fussed for 3 or more hr per day during 3 or more days in 7 days in a
telephone or face-to-face screening interview with a researcher or clinician
2. Total 24-hr crying plus fussing in the selected group of infants is confirmed to be 3 hr or more when measured
by at least 1 prospectively kept, 24-hr behavior diary
From Benninga MA, Nurko S, Faure C, et al: Childhood functional gastrointestinal disorders:
neonate/toddler, Gastroenterology 150(6):1443–1455.e2, 2016.
Table 368.4
Diagnostic Criteria for Functional Diarrhea
Must include all of the following:
1. Daily painless, recurrent passage of 4 or more large, unformed stools
2. Symptoms last more than 4 wk
3. Onset between 6 and 60 mo of age
4. No failure to thrive if caloric intake is adequate
From Benninga MA, Nurko S, Faure C, et al: Childhood functional gastrointestinal disorders:
neonate/toddler, Gastroenterology 150(6):1443–1455.e2, 2016.
Table 368.5
Diagnostic Criteria for Functional Constipation
Must include 1 mo of at least 2 of the following in infants up to 4 yr of age:
1. Two or fewer defecations per week
2. History of excessive stool retention
3. History of painful or hard bowel movements
4. History of large-diameter stools
5. Presence of a large fecal mass in the rectum
In toilet-trained children, the following additional criteria may be used:
6. At least 1 episode/wk of incontinence after the acquisition of toileting skills
7. History of large-diameter stools that may obstruct the toilet
From Benninga MA, Nurko S, Faure C, et al: Childhood functional gastrointestinal disorders:
neonate/toddler, Gastroenterology 150(6):1443–1455.e2, 2016.
Table 368.6
Potential Alarm Features in Constipation
Passage of meconium >48 hr in a term newborn
Constipation starting in the 1st mo of life
Family history of Hirschsprung disease
Ribbon stools
Blood in the stools in the absence of anal fissures
Failure to thrive
Bilious vomiting
Severe abdominal distension
Abnormal thyroid gland
Abnormal position of the anus
Absent anal or cremasteric reflex
Decreased lower extremity strength/tone/reflex
Sacral dimple
Tuft of hair on spine
Gluteal cleft deviation
Anal scars
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468.e2, 2016 (Table 3, p. 1465).
Functional Gastrointestinal Disorders in
Children and Adolescents
Functional nausea and functional vomiting may coexist or may occur
independently of one another (Table 368.7 ). These conditions occur without
coincident abdominal pain. The presentation may accompany autonomic
symptoms such as diaphoresis, pallor, tachycardia, and dizziness. The differential
diagnosis includes anatomical, inflammatory, infectious, and motility etiologies.
Anxiety and other behavioral conditions can be present with these FGIDs and
should be evaluated for and managed accordingly. Cyproheptadine may be
effective in the management of nausea.
Table 368.7
Diagnostic Criteria* for Functional Nausea and Functional
Vomiting
FUNCTIONAL NAUSEA
Must include all of the following fulfilled for the last 2 mo:
1. Bothersome nausea as the predominant symptom, occurring at least twice per week, and generally not related to
meals
2. Not consistently associated with vomiting
3. After appropriate evaluation, the nausea cannot be fully explained by another medical condition
FUNCTIONAL VOMITING
Must include all of the following:
1. On average, 1 or more episodes of vomiting per week
2. Absence of self-induced vomiting or criteria for an eating disorder or rumination
3. After appropriate evaluation, the vomiting cannot be fully explained by another medical condition
* Criteria fulfilled for at least 2 mo before diagnosis.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1457).
Table 368.8
Diagnostic Criteria* for Rumination Syndrome in Children
Must include all of the following:
1. Repeated regurgitation and rechewing or expulsion of food that:
a. Begins soon after ingestion of a meal
b. Does not occur during sleep
2. Not preceded by retching
3. After appropriate evaluation, the symptoms cannot be fully explained by another medical condition. An eating
disorder must be ruled out
* Criteria fulfilled for at least 2 mo before diagnosis.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1458).)
Table 368.9
Diagnostic Criteria* for Functional Dyspepsia
Must include 1 or more of the following bothersome symptoms at least 4 days/mo:
1. Postprandial fullness
2. Early satiation
3. Epigastric pain or burning not associated with defecation
4. After appropriate evaluation, the symptoms cannot be fully explained by another medical condition.
Within FD, the following subtypes are now adopted:
1. Postprandial distress syndrome includes bothersome postprandial fullness or early satiation that prevents
finishing a regular meal. Supportive features include upper abdominal bloating, postprandial nausea, or
excessive belching.
2. Epigastric pain syndrome, which includes all of the following: bothersome (severe enough to interfere with
normal activities) pain or burning localized to the epigastrium. The pain is not generalized or localized to other
abdominal or chest regions and is not relieved by defecation or passage of flatus. Supportive criteria can
include (a) burning quality of the pain but without a retrosternal component and (b) the pain commonly induced
or relieved by ingestion of a meal but may occur while fasting.
* Criteria fulfilled for at least 2 mo before diagnosis.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1460).
Table 368.10
Alarm Symptoms Usually Needing Further Investigations in
Children With Chronic Abdominal Pain
• Pain that wakes up the child from sleep
• Persistent right upper or right lower quadrant pain
• Significant vomiting (bilious vomiting, protracted vomiting, cyclical vomiting, or worrisome pattern to the
physician)
• Unexplained fever
• Genitourinary tract symptoms
• Dysphagia
• Odynophagia
• Chronic severe diarrhea or nocturnal diarrhea
• Gastrointestinal blood loss
• Involuntary weight loss
• Deceleration of linear growth
• Delayed puberty
• Family history of inflammatory bowel disease, celiac disease, and peptic ulcer disease
Table 368.11
Alarm Signs Usually Needing Further Investigations in
Children With Chronic Abdominal Pain
• Localized tenderness in the right upper quadrant
• Localized tenderness in the right lower quadrant
• Localized fullness or mass
• Hepatomegaly
• Splenomegaly
• Jaundice
• Costovertebral angle tenderness
• Arthritis
• Spinal tenderness
• Perianal disease
• Abnormal or unexplained physical findings
• Hematochezia
• Anemia
Pediatric IBS
Pediatric IBS can be classified into 4 groups: IBS with predominant
constipation (IBS-C), IBS with predominant diarrhea (IBS-D), IBS with
constipation and diarrhea, and unspecified IBS. IBS includes findings of
abdominal pain ≥ 4 days/mo associated with defecation and/or a change in
frequency of stool from baseline and/or a change in form/appearance of stool
(Table 368.12 ). It is noteworthy that pain does not resolve following resolution
of constipation; if it does, it is then reclassified as functional constipation. In
fact, IBS-C is often confused with functional constipation. IBS cannot be
explained by another or underlying medical condition. The pathophysiology of
IBS is thought to involve the brain-gut axis and includes a psychosocial stressor
component. Visceral hypersensitivity may be attenuated or amplified by
psychosocial stressors. Abdominal or rectal pain may occur. A postinfectious
IBS phenomenon is known to occur in children, adolescents, and adults and may
be driven by inflammatory cytokines. Perturbations in the intestinal microbiota
or by dysbiosis may be coincident, with causality or consequence not yet
established. The GI differential diagnosis includes anatomical, infectious,
inflammatory, and motility disorders as well as conditions associated with
malabsorption. Differentiation between those GI disorders and IBS is guided by
the history and physical, and markers of inflammation particularly in the stool
such as fecal calprotectin are clinically useful (see Tables 368.10 and 368.11 ).
Management of symptoms can include dietary modification to reduce or restrict
foods that may provoke symptoms or cause gas (see fiber section and
FODMAPS [fermentable oligo-di-monosaccharides and polyols] discussion
Chapter 57 ). Altering microbiota by use of probiotics has been effective; drug
therapy for IBS is noted in Table 368.13 . Peppermint oil has been shown to
reduce pain in children with IBS. Cognitive behavioral therapy is important to
identify possible psychosocial stressors and to help identify coping mechanisms.
Preliminary data suggests that transcutaneous neurostimulation may also be of
value.
Table 368.12
Diagnostic Criteria* for Irritable Bowel Syndrome
Must include all of the following:
1. Abdominal pain at least 4 days/mo associated with one or more of the following:
a. Related to defecation
b. A change in frequency of stool
c. A change in form (appearance) of stool
2. In children with constipation, the pain does not resolve with resolution of the constipation (children in whom
the pain resolves have functional constipation, not irritable bowel syndrome)
3. After appropriate evaluation, the symptoms cannot be fully explained by another medical condition
* Criteria fulfilled for at least 2 mo before diagnosis.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1461).
Table 368.13
Recommendations for Treatment of Irritable Bowel
Syndrome
RECOMMENDATIONS FOR TREATMENT OF IBS
• Mild symptoms often respond to dietary changes.
• Antispasmodics can be used as needed for abdominal pain or postprandial symptoms.
• Antidepressants can improve abdominal pain and global symptoms. They may be considered for patients with
moderate to severe symptoms.
IBS WITH CONSTIPATION (IBS-C)
• Fiber may relieve constipation in patients with mild symptoms.
• Polyethylene glycol can increase the frequency of bowel movements, but may not improve overall symptoms or
abdominal pain.
• Lubiprostone or linaclotide can be tried in patients whose symptoms have not responded to polyethylene glycol.
IBS WITH DIARRHEA (IBS-D)
• Taken as needed, loperamide can reduce postprandial urgency and stool frequency, but it does not improve
global symptoms.
• Rifaximin and eluxadoline have been modestly more effective than placebo in relieving symptoms.
• Alosetron should be reserved for women with severe, chronic IBS-D that is unresponsive to other drugs.
IBS , Irritable bowel syndrome.
From Drugs for irritable bowel syndrome, The Medical Letter 58(1504):121–126, 2016 (p. 121).
Abdominal Migraine
Abdominal migraine shares some features with cyclic vomiting syndrome.
Stereotypical patterns and symptoms afflict the patient, and are typically of acute
onset, intense, lasting for at least an hour, being either periumbilical or
generalized, and usually debilitating during a bout (Table 368.14 ). Episodes can
include anorexia, nausea, emesis, headaches, photophobia, and pallor. Episodes
are separated by weeks to months, with bouts occurring over at least a 6-mo
period. Between bouts, patients return to baseline functioning and are symptom
free. Triggers include sleep hygiene disruption, fatigue, travel, and are usually
alleviated by sleep. The differential diagnosis includes anatomical, infectious, or
inflammatory conditions, as well as hepatobiliary and pancreatic disorders,
neurological and metabolic conditions, and psychiatric disorders. Anatomical
obstruction of the GI or urological tract should be included in the differential
diagnosis. Preventing exposure to known triggers once identified is important.
Similar to prophylaxis for cyclic vomiting syndrome, cyproheptadine,
propranolol, and amitriptyline may be effective. Oral pizotifen (antiserotonin,
antihistamine) is an effective prophylactic agent. Anti-migraine therapies such as
triptans may be effective in aborting bouts. This disorder shares many features
with both cyclic vomiting syndrome and migraine headaches and may evolve
into migraine headaches in adulthood.
Table 368.14
Diagnostic Criteria* for Abdominal Migraine
Must include all of the following occurring at least twice:
1. Paroxysmal episodes of intense, acute periumbilical, midline, or diffuse abdominal pain lasting 1 hr or more
(should be the most severe and distressing symptom)
2. Episodes are separated by weeks to months.
3. The pain is incapacitating and interferes with normal activities
4. Stereotypical pattern and symptoms in the individual patient
5. The pain is associated with 2 or more of the following:
a. Anorexia
b. Nausea
c. Vomiting
d. Headache
e. Photophobia
f. Pallor
6. After appropriate evaluation, the symptoms cannot be fully explained by another medical condition.
* Criteria fulfilled for at least 6 mo before diagnosis.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1462).
Table 368.15
Diagnostic Criteria for Functional Constipation in Children
With Chronic Abdominal Pain
Must include 2 or more of the following occurring at least once per week for a minimum of 1 mo with insufficient
criteria for a diagnosis of irritable bowel syndrome:
1. Two or fewer defecations in the toilet per week in a child of a developmental age of at least 4 yr
2. At least 1 episode of fecal incontinence per week
3. History of retentive posturing or excessive volitional stool retention
4. History of painful or hard bowel movements
5. Presence of a large fecal mass in the rectum
6. History of large diameter stools that can obstruct the toilet
After appropriate evaluation, the symptoms cannot be fully explained by another medical condition.
From Hyams JS, Di Lorenzo C, Saps M, et al: Childhood functional gastrointestinal disorders:
child/adolescent, Gastroenterology 150(6):1456–1468, 2016 (p. 1464).
Table 369.1
Consensus Definition for Diagnostic Criteria and Red Flags
for Cyclic Vomiting Syndrome
DIAGNOSTIC CRITERIA
• Episodic (≥2 or more) attacks of intense nausea and paroxysmal vomiting lasting hours to days within a 6-mo
period
• Stereotypical pattern and symptoms in the individual patient
• Episodes are separated by weeks to months
• Return to baseline between episodes
• Not attributable to another disorder
RED FLAGS
• Bilious emesis, abdominal tenderness, and/or severe abdominal pain
• Attacks precipitated by an intercurrent illness, fasting, and/or a high protein meal
• Neurological abnormalities (mental status changes, ophthalmic abnormalities asymmetry/focal changes, ataxia)
• Atypical pattern or progression/deterioration from a typical presentation for the individual patient to a more
continuous or chronic pattern
Modified from the Rome IV Criteria: Li BU, Lefevre F, Chelimsky GG, et al: North American
Society for Pediatric Gastroenterology, Hepatology and Nutrition consensus statement on the
diagnosis and management of cyclic vomiting syndrome, J Pediatr Gastroenterol Nutr 47(3):379–
393, 2008.
Table 369.2
Supportive Care and Abortive Treatment Approaches in
Cyclic Vomiting Syndrome
SUPPORTIVE CARE
Fluid and electrolyte management Dextrose containing fluid (D10) and normal saline as a single infusion or as a
Y infusion
Nutrition • Resume enteral nutrition as soon as possible.
• If unable to tolerate enteral nutrition and meets criteria, start parenteral
nutrition after 3-4 days.
Medications Antiemetics Ondansetron
• 0.3-0.4 mg/kg/dose IV every 4-6 hr (maximum 16 mg/dose)
• Side effect: constipation, QTc prolongation
Alternative: Granisetron
Sedatives Diphenhydramine
• 1-1.25 mg/kg/dose IV every 6 hr
Lorazepam 0.05-0.1 mg/kg/dose IV every 6 hr
• Side effects: respiratory depression, hallucinations
Chlorpromazine 0.5-1 mg/kg/dose every 6-8 hr +
diphenhydramine IV
Analgesics Ketorolac 0.5 mg/kg/dose IV every 6 hr (maximum dose 30 mg)
Treatment of specific signs and Epigastric pain
symptoms • Acid reduction therapy with an H2 RA or a PPI
Diarrhea
• Antidiarrheals
Hypertension
• Short-acting ACE inhibitors such as captopril
Treatment of specific • Dehydration and electrolyte deficits: replace calculated deficits
complications • Metabolic acidosis: determine etiology and rectify
• SIADH: restrict free water intake
• Hyperemesis: IV acid reduction
• Weight loss: enteral or parenteral nutrition
ABORTIVE CARE
Antimigraine (triptans) Sumatriptan
• 20 mg intranasally at episode onset
• Side effects: neck pain/burning, coronary vasospasm
• Contraindications: basilar artery migraine
RECOVERY AND REFEEDING
• Feed ad libitum when the child declares that the episode is over
Medications listed above are for off-label use.
Modified from Li BU, Lefevre F, Chelimsky GG, et al: North American Society for Pediatric
Gastroenterology, Hepatology and Nutrition consensus statement on the diagnosis and
management of cyclic vomiting syndrome, J Pediatr Gastroenterol Nutr 47(3):379–393, 2008.
Table 369.3
Prophylactic Lifestyle Changes and Pharmacological
Options for Cyclic Vomiting Syndrome
LIFESTYLE MEASURES
Reassurance and • Episodes are not intentional.
anticipatory Guidance • The natural history of CVS is that it will resolve with time.
Avoidance of triggers • Identify dietary triggers (“vomit diary”) and avoid precipitating factors.
• Triggering foods may include chocolate, cheese, monosodium glutamate.
• Fasting a common trigger
• Excitement a potential trigger
• Excessive activity/exhaustion
• Avoid sleep deprivation and practice good sleep hygiene
Managing triggers • Provide supplemental energy as carbohydrates for fasting induced episodes.
• Provision of snacks between meals, before sleep and before exertion
Migraine headache • Aerobic exercise and avoidance of overexertion
type lifestyle • Regular mealtime schedule—avoid skipping meals
interventions • Avoid/moderate caffeine intake
PROPHYLACTIC PHARMACOLOGICAL APPROACHES
Age <5 yr Age ≥5 yr
Antihistamines: Tricyclic antidepressants:
• Cyproheptadine • Amitriptyline
• 0.25-0.5 mg/kg/day in 2 daily • Begin at 0.25-0.5 mg/kg qhs and increase weekly
divided doses or as a single by 5-10 mg until achieve 1-1.5 mg/kg
dose qhs • Monitor EKG for prolonged QTc interval at
• Side effects of increased baseline before initiation and 10 days after peak
appetite, weight gain, and dose achieved
sedation • Side effects: constipation, sedation, arrhythmias,
• Pizotifen behavioral changes
β-blockers: (2nd choice) Alternatives: nortriptyline
• Propranolol β-blockers: (2nd choice):
• 0.25-1 mg/kg/day, most often • Propranolol
10 mg 2-3×/day. Other agents:
• Side effects include lethargy Anticonvulsants:
and reduced exercise • Phenobarbital 2 mg/kg qhs
tolerance. • Side effects: sedation, cognitive impairment
• Contraindicated in asthma, Alternatives:
diabetes, heart disease, • Topiramate, valproic acid, gabapentin, levetiracetam
depression
• Taper over 1-2 wk to
discontinue
DIETARY SUPPLEMENTS
• L-Carnitine 50-100 mg/kg/day divided 2-3×/day, maximum dose of 2 g 2×/day
• Coenzyme Q10 200 mg 2×/day divided 2-3×/day, maximum dose 100 mg 3×/day
Medications listed above are for off-label use. CVS , cyclic vomiting syndrome.
Modified from Li BU, Lefevre F, Chelimsky GG, et al: North American Society for Pediatric
Gastroenterology, Hepatology and Nutrition consensus statement on the diagnosis and
management of cyclic vomiting syndrome, J Pediatr Gastroenterol Nutr 47(3):379–393, 2008.
Bibliography
Bhandari S, Venkatesan T. Clinical characteristics,
Comorbidities and hospital outcomes in hospitalizations with
cyclic vomiting syndrome: a nationwide analysis. Dig Dis Sci
. 2017;62(8):2035–2044.
Bhandari S, Venkatesan T. Novel treatments for cyclic vomiting
syndrome: beyond ondansetron and amitriptyline. Curr Treat
include that it is well-tolerated, non-invasive, and lacks ionizing radiation
exposure. CT is reserved for cases of nonvisualization of the appendix on
ultrasound, or when the ultrasound findings are inconclusive.
Table 370.1
Laboratory Findings
A variety of laboratory tests have been used in the evaluation of children with
suspected appendicitis. Individually, none are very sensitive or specific for
appendicitis, but collectively they can affect the clinician's level of suspicion and
decision-making to proceed with pediatric surgery consultation, discharge, or
imaging studies.
A complete blood count with differential and urinalysis are obtained. The
leukocyte count in early appendicitis may be normal, and typically is only mildly
elevated (11,000-16,000/mm3 ) with a left shift as the illness progresses in the
radial aspect of the upper extremity and are termed the VACTERL (v ertebral, a
nal, c ardiac, t racheal, e sophageal, r enal, l imb) anomalad.
Table 371.1
Associated Malformations
GENITOURINARY
• Vesicoureteric reflux
• Renal agenesis
• Renal dysplasia
• Ureteral duplication
• Cryptorchidism
• Hypospadias
• Bicornuate uterus
• Vaginal septa
VERTEBRAL
• Spinal dysraphism
• Tethered chord
• Presacral masses
• Meningocele
• Lipoma
• Dermoid
• Teratoma
CARDIOVASCULAR
• Tetralogy of Fallot
• Ventricular septal defect
• Transposition of the great vessels
• Hypoplastic left-heart syndrome
GASTROINTESTINAL
• Tracheoesophageal fistula
• Duodenal atresia
• Malrotation
• Hirschsprung disease
CENTRAL NERVOUS SYSTEM
• Spina bifida
• Tethered cord
Anorectal malformations, particularly anal stenosis and rectal atresia, can also
present as Currarino triad, which includes sacral agenesis, presacral mass, and
anorectal stenosis. These patients present with a funnel appearing anus, have
sacral bony defects on plain x-ray, and have a presacral mass (teratoma,
meningocele, dermoid cyst, enteric cyst) on exam or imaging. It is an autosomal
dominant disorder due in most patients to a mutation in the MNX1 gene.
A good correlation exists between the degree of sacral development and future
function. Patients with an absent sacrum usually have permanent fecal and
urinary incontinence. Spinal abnormalities and different degrees of dysraphism
are often associated with these defects. Tethered cord occurs in approximately
division of the fistula without injury to the urinary tract. The rectum is then
dissected proximally until enough length is gained to suture it to an appropriate
perineal position. The muscles of the sphincter complex are then sutured around
(and especially behind) the rectum.
Other operative approaches (such as an anterior approach) are used, but the
most popular procedure is by laparoscopy. This operation allows division of the
fistula under direct visualization and identification of the sphincter complex by
transillumination of perineum. Other imaging techniques in the management of
anorectal malformations include 3D endorectal ultrasound, intraoperative MRI,
and colonoscopy-assisted PSARPs, which may help perform a technically better
operation. None of these other procedures or innovations has demonstrated
improved outcomes.
A similar procedure can be done for female high anomalies with variations to
deal with separating the vagina and rectum from within the cloacal stem. When
the stem is longer than 3 cm, this is an especially difficult and complex
procedure.
Usually the colostomy can be closed 6 wk or more after the PSARP. Two
weeks after any anal procedure, twice-daily dilatations are performed by the
family. By doing frequent dilatations, each one is not so painful and there is less
tissue trauma, inflammation, and scarring.
Outcome
The ability to achieve rectal continence depends on both motor and sensory
elements. There must be adequate muscle in the sphincter complex and proper
positioning of the rectum within the complex. There must also be intact
innervation of the complex and of sensory elements, as well as the presence of
these sensory elements in the anorectum. Patients with low lesions are more
likely to achieve true continence. They are also, however, more prone to
constipation, which leads to overflow incontinence. It is very important that all
these patients are followed closely, and that the constipation and anal dilation are
well managed until toilet training is successful. Tables 371.2 and 371.3 outline
the results of continence and constipation in relation to the malformation
encountered.
Table 371.2
Types of Anorectal Malformation by Sex
MALE (PERCENTAGE CHANCE OF BOWEL CONTROL * )
• Rectoperineal fistula (100%)
• Rectourethral bulbar fistula (85%)
• Imperforate anus without fistula (90%)
• Rectourethral prostatic fistula (65%)
• Rectobladder neck fistula (15%)
FEMALE (PERCENTAGE CHANCE OF BOWEL CONTROL * )
• Rectoperineal fistula (100%)
• Rectovestibular fistula (95%)
• Imperforate anus without fistula (90%)
• Rectovaginal fistula (rare anomaly) †
• Cloaca (70%) ‡
* Provided patients have a normal sacrum, no tethered cord, and they receive a technically correct
Table 371.3
Tumors of the digestive tract in children are mostly polypoid. They are also
commonly syndromic tumors and tumors with known genetic identification
(Table 372.1 ). They usually manifest as painless rectal bleeding, but when large
they can cause obstruction or serve as lead points for intussusception. Most
intestinal tumors can be generally classified into 2 groups: hamartomatous or
adenomatous.
Table 372.1
General Features of the Inherited Colorectal Cancer
Syndromes
AGE RISK OF
POLYP GENETIC CLINICAL ASSOCIATED
SYNDROME OF COLON
DISTRIBUTION LESION MANIFESTATIONS LESIONS
ONSET CANCER
HAMARTOMATOUS POLYPS
Juvenile Large and small 1st ~10–50% PTEN, Possible rectal bleeding, Congenital
polyposis intestine, gastric decade SMAD4, abdominal pain, abnormalities in
polyps BMPR1A intussusception 20% of the
Autosomal nonfamilial
dominant type, clubbing,
AV
malformations
Peutz-Jeghers Small and large 1st Increased LKB1/STK11 Possible rectal bleeding, Orocutaneous
syndrome intestine decade Autosomal abdominal pain, melanin
dominant intussusception pigment spots
Cowden Colon 2nd Not PTEN gene Macrocephaly,
syndrome decade increased breast/thyroid/endometrial
cancers, developmental
delay
Bannayan- Colon 2nd Not PTEN gene Macrocephaly, speckled
Riley- decade increased penis, thyroid/breast
Ruvalcaba cancers, hemangiomas,
syndrome lipomas
ADENOMATOUS POLYPS
Familial Large intestine, 16 yr 100% 5q (APC gene), Rectal bleeding, Desmoids,
adenomatous often >100 (range: autosomal abdominal pain, bowel CHRPE, upper
polyposis 8-34 yr) dominant obstruction GI polyps,
(FAP) osteoma,
hepatoblastoma,
thyroid cancer
Attenuated Colon (fewer in >18 yr Increased APC gene Same as FAP Fewer
familial number) associated
adenomatous lesions
polyposis
(AFAP)
MYH- Colon >20 yr High risk MYH autosomal Same as FAP May be
associated recessive confused with
polyposis sporadic FAP or
AFAP; few
extraintestinal
findings
Gardner Large and small 16 yr 100% 5q (APC gene) Rectal bleeding, Desmoid
syndrome intestine (range: abdominal pain, bowel tumors,
8-34 yr) obstruction multiple
osteomas,
fibromas,
epidermoid
cysts
Hereditary Large intestine 40 yr 30% DNA Rectal bleeding, Other tumors
nonpolyposis mismatch abdominal pain, bowel (e.g., ovary,
colon cancer, repair genes obstruction ureter, pancreas,
(Lynch (MMR) stomach)
syndrome) Autosomal
dominant
APC, adenomatous polyposis coli; AV , arteriovenous; CHRPE , congenital hypertrophy of the
retinal pigment epithelium; GI, gastrointestinal; PTEN , phosphatase and tensin homolog.
Hamartomatous Tumors
Hamartomas are benign tumors composed of tissues that are normally found in
an organ but that are not organized normally. Juvenile, retention, or
inflammatory polyps are hamartomatous polyps, which represent the most
common intestinal tumors of childhood, occurring in 1–2% of children. Patients
generally present in the 1st decade, most often at ages 2-5 yr, and rarely at
younger than 1 yr. Polyps may be found anywhere in the gastrointestinal (GI)
tract, most commonly in the rectosigmoid colon; they are often solitary but may
be multiple.
Histologically, juvenile polyps are composed of hamartomatous collections of
mucus-filled glandular and stromal elements with inflammatory infiltrate,
confined to the inguinal region or pass down into the scrotum. Complete failure
of obliteration of the PV, mostly seen in infants, predisposes to a complete
inguinal hernia characterized by a protrusion of abdominal contents into the
inguinal canal and extending into the scrotum. Obliteration of the PV distally
(around the testis) with patency proximally results in the classic indirect inguinal
hernia with a bulge in the inguinal canal.
A hydrocele occurs when only fluid enters the patent PV; the swelling may
exist only in the scrotum (scrotal hydrocele), only along the spermatic cord in
the inguinal region (hydrocele of the spermatic cord), or extend from the
scrotum through the inguinal canal and even into the abdomen (abdominal–
scrotal hydrocele). A hydrocele is termed a communicating hydrocele if it
demonstrates fluctuation in size, often increasing in size after activity and, at
other times, being smaller when the fluid decompresses into the peritoneal cavity
often after laying recumbent. Occasionally, hydroceles develop in older children
following trauma, inflammation, torsion of the appendix testes, or in association
with tumors affecting the testis.
Although reasons for failure of closure of the PV are unknown, it is more
common in cases of testicular nondescent (cryptorchidism) and prematurity. In
addition, persistent patency of the PV is twice as common on the right side,
presumably related to later descent of the right testis and interference with
obliteration of the PV from the developing inferior vena cava and external iliac
vein. Table 373.1 lists the risk factors identified as contributing to failure of
closure of the PV and to the development of clinical inguinal hernia. The
incidence of inguinal hernia in patients with cystic fibrosis is approximately
15%, believed to be related to an altered embryogenesis of the wolffian duct
structures, which leads to an absent vas deferens and infertility in males with this
condition. There is also an increased incidence of inguinal hernia in patients with
testicular feminization syndrome and other disorders of sexual development.
The rate of recurrence after repair of an inguinal hernia in patients with a
connective tissue disorder approaches 50%, and often the diagnosis of
connective tissue disorders in children results from investigation following
development of a recurrent inguinal hernia.
Table 373.1
Predisposing Factors for Hernias
• Prematurity
• Urogenital
• Cryptorchidism
• Exstrophy of the bladder or cloaca
• Ambiguous genitalia
• Hypospadias/epispadias
• Increased peritoneal fluid
• Ascites
• Ventriculoperitoneal shunt
• Peritoneal dialysis catheter
• Increased intraabdominal pressure
• Repair of abdominal wall defects
• Severe ascites (chylous)
• Meconium peritonitis
• Chronic respiratory disease
• Cystic fibrosis
• Connective tissue disorders
• Ehlers-Danlos syndrome
• Hunter-Hurler syndrome
• Marfan syndrome
• Mucopolysaccharidosis
Table 377.1
FDA-Approved Pancreatic Enzyme Replacement Products
for Exocrine Pancreatic Insufficiency*
available formulations) for a 70-kg patient. WAC is wholesaler acquisition cost, or manufacturer's
published price to wholesalers; WAC represents published catalogue or list prices and may not
represent an actual transactional price.
‡ Viokace is only approved for use in adults.
§ Should be used in combination with a proton pump inhibitor to maximize absorption in the
duodenum.
|| FDA-approved only for treatment of adults with EPI due to chronic pancreatitis or
pancreatectomy.
¶ Should not be crushed or chewed.
** Capsules can be opened and contents sprinkled on soft acidic food (pH ≤ 4.5) such as
applesauce.
From The Medical Letter: Pancreatic enzyme replacement products, Med Lett 59(1531):170,
2017.
Source: AnalySource Monthly. September 5, 2017. Reprinted from First Databank, Inc. All rights
reserved. ©2017. www.fdbhealth.com/policies/drug-pricing-policy/ .
Table 377.2
Table 378.1
Etiology of Acute and Recurrent Pancreatitis in Children
DRUGS AND TOXINS
• Acetaminophen overdose
• Alcohol
• L -Asparaginase
• Azathioprine
• Cannabis
• Carbamazepine
• Cimetidine
• Corticosteroids
• Cytosine arabinoside
• Dapsone
• Didanosine
• Enalapril
• Erythromycin
• Estrogen
• Furosemide
• Glucagon-like peptide-1 agents
• Interferon α
• Isoniazid
• Lamivudine
• Lisinopril
• 6-Mercaptopurine
• Methyldopa
• Mesalamine
• Metronidazole
• Octreotide
• Organophosphate poisoning
• Pentamidine
• Procainamide
• Retrovirals: DDC (dideoxycytidine), DDI (dideoxyinosine), tenofovir
• Rifampin
• Sulfonamides: mesalamine, 5-aminosalicytates, sulfasalazine, trimethoprim-sulfamethoxazole
• Sulindac
• Tetracycline
• Thiazides
• Valproic acid
• Venom (spider, scorpion, Gila monster lizard)
• Vincristine
• Volatile hydrocarbons
GENETIC
• Cationic trypsinogen gene (PRSS1)
• Chymotrypsin C gene (CTRC)
• Cystic fibrosis gene (CFTR)
• Trypsin inhibitor gene (SPINK1)
INFECTIOUS
• Ascariasis
• Coxsackie B virus
• Echovirus
• Enterovirus
• Epstein-Barr virus
• Hepatitides A, B
• Herpes viruses
• Influenzae A, B
• Leptospirosis
• Malaria
• Measles
• Mumps
• Mycoplasma
• Rabies
• Rubella
• Reye syndrome: varicella, influenza B
• Septic shock
• Thyroid fever
OBSTRUCTIVE
• Ampullary disease
• Ascariasis
• Biliary tract malformations
• Choledochal cyst
• Choledochocele
• Cholelithiasis, microlithiasis, and choledocholithiasis (stones or sludge)
• Duplication cyst
• Endoscopic retrograde cholangiopancreatography (ERCP) complication
• Pancreas divisum
• Pancreatic ductal abnormalities
• Postoperative
• Sphincter of Oddi dysfunction
• Tumor
SYSTEMIC DISEASE
• Autoimmune pancreatitis (IgG4 -related systemic disease)
• Brain tumor
• Collagen vascular diseases
• Congenital partial lipodystrophy
• Crohn disease
• Diabetes mellitus (ketoacidosis)
• Head trauma
• Henoch-Schönlein purpura
• Hemochromatosis
• Hemolytic uremic syndrome
• Hyperlipidemia: types I, IV, V
• Hyperparathyroidism/hypercalcemia
• Kawasaki disease
• Malnutrition
• Organic acidemia
• Peptic ulcer
• Periarteritis nodosa
• Renal failure
• Scorpion venom
• Systemic lupus erythematosus
• Transplantation: bone marrow, heart, liver, kidney, pancreas
• Vasculitis
TRAUMATIC
• Blunt injury
• Burns
• Child abuse
• Hypothermia
• Surgical trauma
• Total-body cast
Clinical Manifestations
The severity of AP in children has been defined by a consensus committee.
Mild Acute Pancreatitis: AP that is not associated with organ failure, local or
systemic complications, and usually resolves within the 1st wk after
presentation. This is the most common form of pediatric AP.
The patient with mild AP has moderate to severe abdominal pain, persistent
greater than 7 times the upper limit of normal obtained within 24 hr of
presentation may predict a severe course. Serum lipase can be elevated in
nonpancreatic diseases. The serum amylase level is typically elevated for up to 4
days. A variety of other conditions can also cause hyperamylasemia without
pancreatitis (Table 378.2 ). Elevation of salivary amylase can mislead the
clinician to diagnose pancreatitis in a child with abdominal pain. The laboratory
can separate amylase isoenzymes into pancreatic and salivary fractions. Initially
serum amylase levels are normal in 10–15% of patients.
Table 378.2
Differential Diagnosis of Hyperamylasemia
PANCREATIC PATHOLOGY
• Acute or chronic pancreatitis
• Complications of pancreatitis (pseudocyst, ascites, abscess)
• Factitious pancreatitis
SALIVARY GLAND PATHOLOGY
• Parotitis (mumps, Staphylococcus aureus , cytomegalovirus, HIV, Epstein-Barr virus)
• Sialadenitis (calculus, radiation)
• Eating disorders (anorexia nervosa, bulimia)
INTRAABDOMINAL PATHOLOGY
• Biliary tract disease (cholelithiasis)
• Peptic ulcer perforation
• Peritonitis
• Intestinal obstruction
• Appendicitis
SYSTEMIC DISEASES
• Metabolic acidosis (diabetes mellitus, shock)
• Renal insufficiency, transplantation
• Burns
• Pregnancy
• Drugs (morphine)
• Head injury
• Cardiopulmonary bypass
Table 378.4
Complications of Acute Pancreatitis
LOCAL
Pseudocyst
Sterile necrosis
Infected necrosis
Abscess
GI bleeding
• Pancreatitis-related
• Splenic artery or splenic artery pseudoaneurysm rupture
• Splenic vein rupture
• Portal vein rupture
• Splenic vein thrombosis leading to gastroesophageal variceal bleeding
• Pseudocyst or abscess hemorrhage
• Postnecrosectomy bleeding
Nonpancreatitis-related
• Mallory-Weiss tear
• Alcoholic gastropathy
• Stress-related mucosal gastropathy
Splenic complications
• Infarction
• Rupture
• Hematoma
• Splenic vein thrombosis
Fistulization to or obstruction of the small intestine or colon
Hydronephrosis
SYSTEMIC
Respiratory failure
Renal failure
Shock
Hyperglycemia
Hypocalcemia
Disseminated intravascular coagulation
Fat necrosis (subcutaneous nodules)
Retinopathy
PSYCHOSIS
From Tenner S, Steinberg WM: Acute pancreatitis. In Feldman M, Friedman LS, Brandt LJ,
editors: Sleisenger and Fordtran's gastrointestinal and liver disease, ed 10, Philadelphia, 2016,
Elsevier (Box 58.7, p. 991).
acute recurrent pancreatitis
autoimmune pancreatitis
PRSS1 gene
CTRC gene
CFTR gene
SPINK1 gene
tropical pancreatitis
Table 378.5
From Schwarzenberg SJ, Bellin M, Husain SZ, et al: Pediatric chronic pancreatitis is associated
with genetic risk factors and substantial disease burden, J Pediatr 166:890–896, 2015 (Table II, p.
892).
Table 378.6
Table 378.7
Conditions Classification of Chronic Disorders Now
Recognized to Be Part of IgG4-Related Disease
Autoimmune pancreatitis (lymphoplasmacytic sclerosing pancreatitis)
Eosinophilic angiocentric fibrosis (affecting the orbits and upper respiratory tract)
Fibrosing mediastinitis
Hypertrophic pachymeningitis
Idiopathic hypocomplementaemic tubulointerstitial nephritis with extensive tubulointerstitial deposits
Inflammatory pseudotumor (affecting the orbits, lungs, kidneys, and other organs)
Küttner tumor (affecting the submandibular glands)
Mikulicz disease (affecting the salivary and lacrimal glands)
Multifocal fibrosclerosis (commonly affecting the orbits, thyroid gland, retroperitoneum, mediastinum, and other
tissues and organs)
Periaortitis and periarteritis
Inflammatory aortic aneurysm
Retroperitoneal fibrosis (Ormond disease)
Riedel thyroiditis
Sclerosing mesenteritis
Conditions once regarded as individual disorders now recognized to be part of IgG4-related disease
From Kamisawa T, Zen Y, Pillai S, Stone JH: IgG4-related disease, Lancet 385:1460–1471, 2015
(Panel 1, p. 1461).
Pancreatic Tumors
Meghen B. Browning, Steven L. Werlin, Michael Wilschanski
Table 380.1
Syndromes Associated With Pancreatic Neuroendocrine
Tumors (pNETs)*
The growth and development of the newly budded liver require interactions
with endothelial cells. Certain proteins are important for liver development in
animal models (Table 381.1 ). In addition to these proteins, microRNAs, which
consist of small noncoding, single-stranded RNAs, have a functional role in the
regulation of gene expression and hepatobiliary development in zebrafish and
mouse models.
Table 381.1
Selected Growth Factors, Receptors, Protein Kinases, and
Transcription Factors Required for Normal Liver
Development in Animal Models
INDUCTION OF HEPATOCYTE FATE THROUGH CARDIAC MESODERM
• Fibroblast growth factors (FGFs) 1, 2, 8
• FGF receptors 1, 4
INDUCTION OF HEPATOCYTE FATE THROUGH SEPTUM TRANSVERSUM
• Bone morphogenetic proteins 2, 4, 7
STIMULATION OF HEPATOBLAST GROWTH AND PROLIFERATION
• Hepatocyte growth factor (HGF)
• HGF receptor c-met
• “Pioneer” transcription factors Foxa1, Foxa2, and Gata4, Gata6
• Transcription factors Xbp1, Foxm1b, Hlx, Hex, Prox1
• Wnt signaling pathway, β-catenin
SPECIFICATION OF HEPATOCYTE LINEAGE
• HGF
• Transforming growth factor-β and its downstream effectors Smad 2, Smad 3
• Hepatocyte nuclear factors (HNFs) 1α, 4α, 6
SPECIFICATION OF CHOLANGIOCYTE LINEAGE
• Jagged 1 (Notch ligand) and Notch receptors 1, 2
• HNF6, HNF1β
• Wnt signaling pathway, β-catenin
• Vacuolar sorting protein Vps33b
FIG. 381.2 Hepatic morphogenesis. A, Ventral outgrowth of hepatic diverticulum from
foregut endoderm in the 3.5-wk embryo. B, Between the 2 vitelline veins, the enlarging
hepatic diverticulum buds off epithelial (liver) cords that become the liver parenchyma,
around which the endothelium of capillaries (sinusoids) align (4 wk embryo). C,
Hemisection of embryo at 7.5 wk. D, Three-dimensional representation of the hepatic
Table 381.2
Causes of Impaired Bile Acid Metabolism and
Enterohepatic Circulation
DEFECTIVE BILE ACID SYNTHESIS OR TRANSPORT
• Inborn errors of bile acid synthesis (reductase deficiency, isomerase deficiency)
• Progressive familial intrahepatic cholestasis (PFIC1, PFIC2, PFIC3)
• Intrahepatic cholestasis (neonatal hepatitis)
• Acquired defects in bile acid synthesis secondary to severe liver disease
ABNORMALITIES OF BILE ACID DELIVERY TO THE BOWEL
• Celiac disease (sluggish gallbladder contraction)
• Extrahepatic bile duct obstruction (e.g., biliary atresia, gallstones)
LOSS OF ENTEROHEPATIC CIRCULATION OF BILE ACIDS
• External bile fistula
• Cystic fibrosis
• Small bowel bacterial overgrowth syndrome (with bile acid precipitation, increased jejunal absorption, and
“short-circuiting”)
• Drug-induced entrapment of bile acids in intestinal lumen (e.g., cholestyramine)
BILE ACID MALABSORPTION
• Primary bile acid malabsorption (absent or inefficient ileal active transport)
• Secondary bile acid malabsorption
• Ileal disease or resection
• Cystic fibrosis
DEFECTIVE UPTAKE OR ALTERED INTRACELLULAR METABOLISM
• Parenchymal disease (acute hepatitis, cirrhosis)
• Regurgitation from cells
• Portosystemic shunting
• Cholestasis
Bibliography
Bates MD, Balistreri WF. The gastrointestinal tract:
development of the human digestive system. Fanaroff AA,
Martin RJ. Neonatal-perinatal medicine: diseases of the fetus
and infant . ed 7. Mosby: St. Louis; 2002:1255–1263.
Cardinale V, Wang Y, Carpino G, et al. The biliary tree-a
reservoir of multipotent stem cells. Nat Rev Gastroenterol
Hepatol . 2012;9:231–240.
Carpentier R, Suñer RE, van Hul N, et al. Embryonic ductal
plate cells give rise to cholangiocytes, periportal hepatocytes,
and adult liver progenitor cells. Gastroenterology .
2011;141:1432–1438.
nodules of various sizes (up to 5 cm) separated by broad septa, or micronodular ,
with nodules of uniform size (<1 cm) separated by fine septa; mixed forms
occur. The progressive scarring results in altered hepatic blood flow, with further
impairment of liver cell function. Increased intrahepatic resistance to portal
blood flow leads to portal hypertension.
The liver can be secondarily involved in neoplastic (metastatic) and non-
neoplastic (storage diseases, fat infiltration) processes, as well as a number of
systemic conditions and infectious processes. The liver can be affected by
chronic passive congestion (congestive heart failure) or acute hypoxia, with
hepatocellular damage.
Clinical Manifestations
Hepatomegaly
Enlargement of the liver can be caused by several mechanisms (Table 382.1 ).
Normal liver size estimations are based on age-related clinical indices, such as
the degree of extension of the liver edge below the costal margin, the span of
dullness to percussion, or the length of the vertical axis of the liver, as estimated
from imaging techniques. In children, the normal liver edge can be felt up to 2
cm below the right costal margin. In a newborn infant, extension of the liver
edge more than 3.5 cm below the costal margin in the right midclavicular line
suggests hepatic enlargement. Measurement of liver span is carried out by
percussing the upper margin of dullness and by palpating the lower edge in the
right midclavicular line. This may be more reliable than an extension of the liver
edge alone. The 2 measurements may correlate poorly.
Table 382.1
Mechanisms of Hepatomegaly
INCREASE IN THE NUMBER OR SIZE OF THE CELLS INTRINSIC TO THE LIVER
Storage
Fat: malnutrition, obesity, metabolic liver disease (diseases of fatty acid oxidation and Reye syndrome–like
illnesses), lipid infusion (total parenteral nutrition), cystic fibrosis, medication related, pregnancy
Specific lipid storage diseases: Gaucher, Niemann-Pick, Wolman disease
Glycogen: glycogen storage diseases (multiple enzyme defects); total parenteral nutrition; infant of diabetic
mother, Beckwith syndrome, poorly controlled type 1 diabetes mellitus (Mauriac syndrome)
Miscellaneous: α1 -antitrypsin deficiency, Wilson disease, hypervitaminosis A
Inflammation
Hepatocyte enlargement (hepatitis)
• Viral: acute and chronic
• Bacterial: sepsis, abscess, cholangitis
• Toxic: drugs
• Autoimmune
Kupffer cell enlargement
• Sarcoidosis
• Systemic lupus erythematosus
• Hemophagocytic lymphohistiocytosis
• Macrophage activating syndrome
INFILTRATION OF CELLS
Primary Liver Tumors: Benign
Hepatocellular
• Focal nodular hyperplasia
• Nodular regenerative hyperplasia
• Hepatocellular adenoma
Mesodermal
• Infantile hemangioendothelioma
• Mesenchymal hamartoma
Cystic masses
• Choledochal cyst
• Hepatic cyst
• Hematoma
• Parasitic cyst
• Pyogenic or amebic abscess
Primary Liver Tumors: Malignant
Hepatocellular
• Hepatoblastoma
• Hepatocellular carcinoma
Mesodermal
• Angiosarcoma
• Undifferentiated embryonal sarcoma
Secondary or metastatic processes
• Lymphoma
• Leukemia
• Lymphoproliferative disease
• Langerhans cell histiocytosis
• Neuroblastoma
• Wilms tumor
INCREASED SIZE OF VASCULAR SPACE
Intrahepatic obstruction to hepatic vein outflow
• Venoocclusive disease
• Hepatic vein thrombosis (Budd-Chiari syndrome)
• Hepatic vein web
Suprahepatic
• Congestive heart failure
Pericardial disease/tamponade/constrictive pericarditis
Post-Fontan procedure
Hematopoietic: sickle cell anemia, thalassemia
INCREASED SIZE OF BILIARY SPACE
Congenital hepatic fibrosis
Caroli disease
Extrahepatic obstruction
IDIOPATHIC
Various
• Riedel lobe
• Normal variant
• Downward displacement of diaphragm
The liver span increases linearly with body weight and age in both sexes,
ranging from approximately 4.5-5.0 cm at 1 wk of age to approximately 7-8 cm
in boys and 6.0-6.5 cm in girls by 12 yr of age. The lower edge of the right lobe
of the liver extends downward (Riedel lobe) and can normally be palpated as a
broad mass in some people. An enlarged left lobe of the liver is palpable in the
epigastrium of some patients with cirrhosis. Downward displacement of the liver
by the diaphragm (hyperinflation) or thoracic organs can create an erroneous
impression of hepatomegaly.
Examination of the liver should note the consistency, contour, tenderness, and
presence of any masses or bruits, as well as assessment of spleen size, along with
documentation of the presence of ascites and any stigmata of chronic liver
disease.
Ultrasound is useful in assessment of liver size and consistency, as well as
gallbladder size. Gallbladder length normally varies from 1.5-5.5 cm (average: 3
cm) in infants to 4-8 cm in adolescents; width ranges from 0.5-2.5 cm for all
ages. Gallbladder distention may be seen in infants with sepsis. The gallbladder
is often absent in infants with biliary atresia.
Jaundice (Icterus)
Yellow discoloration of the sclera, skin, and mucous membranes is a sign of
hyperbilirubinemia (see Chapter 123.3 ). Clinically apparent jaundice in children
and adults occurs when the serum concentration of bilirubin reaches 2-3 mg/dL
(34-51 µmol/L); the neonate might not appear jaundiced until the bilirubin level
is >5 mg/dL (>85 µmol/L). Jaundice may be the earliest and only sign of hepatic
dysfunction. Liver disease must be suspected in the infant who appears only
mildly jaundiced but has dark urine or acholic (light-colored) stools. Immediate
evaluation to establish the cause is required.
Measurement of the total serum bilirubin concentration allows quantitation of
jaundice. Bilirubin occurs in plasma in 4 forms: unconjugated bilirubin tightly
bound to albumin; free or unbound bilirubin (the form responsible for
kernicterus, because it can cross cell membranes); conjugated bilirubin (the only
fraction to appear in urine); and δ fraction (bilirubin covalently bound to
albumin), which appears in serum when hepatic excretion of conjugated
bilirubin is impaired in patients with hepatobiliary disease. The δ fraction
permits conjugated bilirubin to persist in the circulation and delays resolution of
jaundice. Although the terms direct and indirect bilirubin are used equivalently
with conjugated and unconjugated bilirubin, this is not quantitatively correct,
because the direct fraction includes both conjugated bilirubin and δ bilirubin.
Investigation of jaundice in an infant or older child must include
determination of the accumulation of both unconjugated and conjugated
bilirubin. Unconjugated hyperbilirubinemia might indicate increased production,
hemolysis, reduced hepatic removal, or altered metabolism of bilirubin (Table
382.2 ). Conjugated hyperbilirubinemia reflects decreased excretion by damaged
hepatic parenchymal cells or disease of the biliary tract, which may be a result of
obstruction, sepsis, toxins, inflammation, and genetic or metabolic disease (Table
382.3 ).
Table 382.2
Differential Diagnosis of Unconjugated Hyperbilirubinemia
INCREASED PRODUCTION OF UNCONJUGATED BILIRUBIN FROM HEME
Hemolytic Disease (Hereditary or Acquired)
Isoimmune hemolysis (neonatal; acute or delayed transfusion reaction; autoimmune)
• Rh incompatibility
• ABO incompatibility
• Other blood group incompatibilities
Congenital spherocytosis
Hereditary elliptocytosis
Infantile pyknocytosis
Erythrocyte enzyme defects
Hemoglobinopathy
• Sickle cell anemia
• Thalassemia
• Others
Sepsis
Microangiopathy
• Hemolytic-uremic syndrome
• Hemangioma
• Mechanical trauma (heart valve)
Ineffective erythropoiesis
Drugs
Infection
Enclosed hematoma
Polycythemia
• Diabetic mother
• Fetal transfusion (recipient)
• Delayed cord clamping
DECREASED DELIVERY OF UNCONJUGATED BILIRUBIN (IN PLASMA) TO HEPATOCYTE
Right-sided congestive heart failure
Portacaval shunt
DECREASED BILIRUBIN UPTAKE ACROSS HEPATOCYTE MEMBRANE
Presumed enzyme transporter deficiency
Competitive inhibition
• Breast milk jaundice
• Lucey-Driscoll syndrome
• Drug inhibition (radiocontrast material)
Miscellaneous
• Hypothyroidism
• Hypoxia
• Acidosis
DECREASED STORAGE OF UNCONJUGATED BILIRUBIN IN CYTOSOL (DECREASED Y AND Z
PROTEINS)
Competitive inhibition
Fever
DECREASED BIOTRANSFORMATION (CONJUGATION)
Neonatal jaundice (physiologic)
Inhibition (drugs)
Hereditary (Crigler-Najjar)
• Type I (complete enzyme deficiency)
• Type II (partial deficiency)
Gilbert disease
Hepatocellular dysfunction
ENTEROHEPATIC RECIRCULATION
Breast milk jaundice
Intestinal obstruction
• Ileal atresia
• Hirschsprung disease
• Cystic fibrosis
• Pyloric stenosis
Antibiotic administration
Table 382.3
Differential Diagnosis of Neonatal and Infantile Cholestasis
INFECTIOUS
Generalized bacterial sepsis
Viral hepatitis
• Hepatitides A, B, C, D, E
• Cytomegalovirus
• Rubella virus
• Herpesviruses: herpes simplex, human herpesvirus 6 and 7
• Varicella virus
• Coxsackievirus
• Echovirus
• Reovirus type 3
• Parvovirus B19
• HIV
• Adenovirus
Others
• Toxoplasmosis
• Syphilis
• Tuberculosis
• Listeriosis
• Urinary tract infection
TOXIC
Sepsis
Parenteral nutrition related
Drug, dietary supplement, herbal related
METABOLIC
Disorders of amino acid metabolism
• Tyrosinemia
Disorders of lipid metabolism
• Wolman disease
• Niemann-Pick disease (type C)
• Gaucher disease
Cholesterol ester storage disease
Disorders of carbohydrate metabolism
• Galactosemia
• Fructosemia
• Glycogenosis IV
Disorders of bile acid biosynthesis
Other metabolic defects
• α1 -Antitrypsin deficiency
• Cystic fibrosis
• Hypopituitarism
• Hypothyroidism
• Zellweger (cerebrohepatorenal) syndrome
• Wilson disease
• Gestational alloimmune liver disease (previously neonatal iron storage disease )
• Indian childhood cirrhosis/infantile copper overload
• Congenital disorders of glycosylation
• Mitochondrial hepatopathies
• Citrin deficiency
GENETIC OR CHROMOSOMAL
Trisomies 17, 18, 21
INTRAHEPATIC CHOLESTASIS SYNDROMES
“Idiopathic” neonatal hepatitis
Alagille syndrome
Intrahepatic cholestasis (progressive familial intrahepatic cholestasis [PFIC])
• FIC-1 deficiency
• Bile salt export pump (BSEP) deficiency
• MDR3 deficiency
• Tight junction protein 2 deficiency
• Farnesoid X receptor (FXR) mutations
Familial benign recurrent cholestasis associated with lymphedema (Aagenaes syndrome)
ARC (arthrogryposis, renal dysfunction, and cholestasis) syndrome
Caroli disease (cystic dilation of intrahepatic ducts)
EXTRAHEPATIC DISEASES
Biliary atresia
Sclerosing cholangitis
Bile duct stricture/stenosis
Choledochal–pancreaticoductal junction anomaly
Spontaneous perforation of the bile duct
Choledochal cyst
Mass (neoplasia, stone)
Bile/mucous plug (“inspissated bile”)
MISCELLANEOUS
Shock and hypoperfusion
Associated with enteritis
Associated with intestinal obstruction
Neonatal lupus erythematosus
Myeloproliferative disease (trisomy 21)
Hemophagocytic lymphohistiocytosis (HLH)
COACH syndrome (coloboma, oligophrenia, ataxia, cerebellar vermis hypoplasia, hepatic fibrosis)
Cholangiocyte cilia defects
Pruritus
Intense generalized itching can occur in patients with chronic liver disease often
in association with cholestasis (conjugated hyperbilirubinemia). Symptoms can
be generalized or localized (commonly to palms and soles), are usually worse at
night, are exacerbated with stress and heat, and are relieved by cool
temperatures. Pruritus is unrelated to the degree of hyperbilirubinemia; deeply
jaundiced patients can be asymptomatic.
The pathogenesis of pruritus remains unknown, however, multiple suspected
pruritogens have been reported including bile acids, histamine, serotonin,
progesterone metabolites, endogenous opioids, the potent neuronal activator
lysophosphatidic acid (LPA), and the LPA-forming enzyme, autotaxin (ATX).
Ultimately, a multifactorial process is suspected as evidenced by the
symptomatic relief of pruritus after administration of various therapeutic agents
including bile acid-binding agents (cholestyramine), choleretic agents
(ursodeoxycholic acid), opiate antagonists, antihistamines, serotonin reuptake
inhibitors (sertraline), and antibiotics. Plasmapheresis, molecular adsorbent
recirculating system therapy, and surgical diversion of bile (partial and total
biliary diversion) have been used in attempts to provide relief for medically
refractory pruritus.
Spider Angiomas
Vascular spiders (telangiectasias) , characterized by central pulsating arterioles
from which small, wiry venules radiate, may be seen in patients with chronic
liver disease. These are usually most prominent in the superior vena cava
distribution area (on the face and chest). Their size varies between 1 and 10 mm
and they exhibit central clearing with pressure. They presumably reflect altered
estrogen metabolism in the presence of hepatic dysfunction.
Biochemical Tests
Laboratory tests commonly used to screen for or to confirm a suspicion of liver
disease include measurements of serum aminotransferase (Table 382.4 ),
bilirubin (total and fractionated), alkaline phosphatase (AP) and gamma
glutamyl-transpeptidase (GGT) levels, as well as determinations of prothrombin
time (PT) or international normalized ratio (INR) and serum albumin level.
These tests are complementary, provide an estimation of synthetic and excretory
functions, and might suggest the nature of the disturbance (inflammation or
cholestasis).
Table 382.4
Causes of Elevated Serum Aminotransferase Levels*
CHRONIC, MILD ELEVATIONS, ALT > AST (<150 U/L OR 5 × NORMAL)
Hepatic Causes
α1 -Antitrypsin deficiency
Autoimmune hepatitis
Chronic viral hepatitis (B, C, and D)
Hemochromatosis
Medications and toxins
Steatosis and steatohepatitis
Wilson disease
Nonhepatic Causes
Celiac disease
Hyperthyroidism
SEVERE, ACUTE ELEVATIONS, ALT > AST (>1,000 U/L OR >20-25 × NORMAL)
Hepatic Causes
Acute bile duct obstruction
Acute Budd-Chiari syndrome
Acute viral hepatitis
Autoimmune hepatitis
Drugs and toxins
Hepatic artery ligation
Ischemic hepatitis
Wilson disease
SEVERE, ACUTE ELEVATIONS, AST > ALT (>1,000 U/L OR >20-25 × NORMAL)
Hepatic Cause
Medications or toxins in a patient with underlying alcoholic liver injury
Nonhepatic Cause
Acute rhabdomyolysis
CHRONIC, MILD ELEVATIONS, AST > ALT (<150 U/L, <5 × NORMAL)
Hepatic Causes
Alcohol-related liver injury (AST/ALT > 2 : 1, AST nearly always <300 U/L)
Cirrhosis
Nonhepatic Causes
Hypothyroidism
Macro-AST
Myopathy
Strenuous exercise
CHRONIC, MILD ELEVATIONS, ALT > AST (<150 U/L OR 5 × NORMAL)
* Virtually any liver disease can cause moderate aminotransferase elevations (5-15 × normal).
Table 383.1
Proposed Subtypes of Intrahepatic Cholestasis
A. Disorders of membrane transport and secretion
1. Disorders of canalicular secretion
a. Bile acid transport: BSEP deficiency
i. Persistent, progressive (PFIC type 2)
ii. Recurrent, benign (BRIC type 2)
b. Phospholipid transport: MDR3 deficiency (PFIC type 3)
c. Ion transport: cystic fibrosis (CFTR)
d. Tight junction defect (TJP2 deficiency)
2. Complex or multiorgan disorders
a. FIC1 deficiency
i. Persistent, progressive (PFIC type 1, Byler disease)
ii. Recurrent, benign (BRIC type 1)
b. Neonatal sclerosing cholangitis (CLDN1)
c. Arthrogryposis-renal dysfunction-cholestasis syndrome (VPS33B)
B. Disorders of bile acid biosynthesis, conjugation and regulation
1. Δ4 -3-Oxosteroid-5β- reductase deficiency
2. 3β- hydroxy-5-C27-steroid dehydrogenase/isomerase deficiency
3. Oxysterol 7α- hydroxylase deficiency
4. Bile acid-CoA Ligase deficiency
5. BAAT deficiency (familial hypercholanemia)
6. Farnesoid X receptor (FXR) deficiency.
C. Disorders of embryogenesis
1. Alagille syndrome (Jagged1 defect, syndromic bile duct paucity)
2. Ductal plate malformation (ARPKD, ADPLD, Caroli disease)
D. Unclassified (idiopathic “neonatal hepatitis”): mechanism unknown
Note: FIC1 deficiency, BSEP deficiency, and some of the disorders of bile acid biosynthesis are
characterized clinically by low levels of serum GGT despite the presence of cholestasis. In all
other disorders listed, the serum GGT level is elevated.
ADPLD, autosomal dominant polycystic liver disease (cysts in liver only); ARPKD, autosomal
recessive polycystic kidney disease (cysts in liver and kidney); BAAT, bile acid transporter; BRIC,
benign recurrent intrahepatic cholestasis; BSEP, bile salt export pump in; GGT, γ-glutamyl
transpeptidase; PFIC, progressive familial intrahepatic cholestasis.
From Balistreri WF, Bezerra JA, Jansen P, et al: Intrahepatic cholestasis: summary of an
American Association for the study of liver diseases single-topic conference, Hepatology
42(1):222–235, 2005.
Evaluation
Identification of cholestasis warrants a prompt effort to accurately diagnose the
cause (Table 383.2 ). Although cholestasis in the neonate may be the initial
manifestation of numerous and potentially serious disorders, the clinical
manifestations are usually similar and provide few clues about the etiology.
Affected infants have icterus, dark urine, light or acholic stools, and
hepatomegaly, all resulting from decreased bile flow as a result of either
hepatocyte injury or bile duct obstruction. Hepatic synthetic dysfunction can
lead to hypoprothrombinemia and bleeding. Administration of vitamin K should
be included in the initial treatment of cholestatic infants to prevent hemorrhage
(intracranial).
Table 383.2
Table 383.3
Phenotypic Classification of Primary Mitochondrial
Hepatopathies
RC (electron transport) defects (OXPHOS)
• Neonatal liver failure
• Complex I deficiency
• Complex IV deficiency (SCO1 mutations)
• Complex III deficiency (BCS1L mutations)
• Co-enzyme Q deficiency
• Multiple complex deficiencies (transfer and elongation factor mutations)
• mtDNA depletion syndrome (DUGOK , MPV17 , POLG , SUCLG1 , C10orf2/Twinkle mutations)
• Later-onset liver dysfunction or failure
• Alpers-Huttenlocher disease (POLG mutations)
• Pearson's marrow pancreas syndrome (mtDNA deletion)
• Mitochondrial neurogastrointestinal encephalopathy (TYMP mutations)
• NNH (MPV17 mutations)
Fatty acid oxidation defects
• Long-chain 3 hydroxyacyl-coenzyme A dehydrogenase
• Carnitine palmitoyltransferase I and II deficiencies
• Carnitine-acylcarnitinetranslocase deficiency
Urea cycle enzyme deficiencies
Electron transfer flavoprotein and electron transfer flavoprotein dehydrogenase deficiencies
Phosphoenol pyruvate carboxykinase (mitochondrial) deficiency; nonketotic hyperglycemia
Citrin deficiency; neonatal intrahepatic cholestasis caused by citrin deficiency (SLC25A13 mutations)
NN, Navajo neurohepatopathy; OXPHOS, oxidative phosphorylation; RC, respiratory chain.
From Lee WS, Sokol RJ: Mitochondrial hepatopathies: advances in genetics, therapeutic
approaches, and outcomes, J Pediatr 163[4]:942–948, 2013, Table 1, p. 943.
Intrahepatic Cholestasis
Neonatal Hepatitis
The term neonatal hepatitis implies intrahepatic cholestasis (see Fig. 383.1 ),
which has various forms (see Tables 383.1 and 383.4 ).
Table 383.4
Molecular Defects Causing Liver Disease
Table 383.5
Progressive Familial Intrahepatic Cholestasis
Table 383.6
FIG. 383.2 Posterior embryotoxon. (From Turnpenny PD, Ellard S: Alagille
syndrome: pathogenesis, diagnosis and management, Eur J Hum Genet
damage and cirrhosis. Growth failure is a major concern and is related in part to
malabsorption and malnutrition resulting from ineffective digestion and
absorption of dietary fat. Use of a medium-chain triglyceride-containing formula
can improve caloric balance. With chronic cholestasis and prolonged survival,
children with hepatobiliary disease can experience deficiencies of the fat-soluble
vitamins (A, D, E, and K). Metabolic bone disease is common. It is essential to
monitor the fat-soluble vitamin status in patients.
Table 383.7
Suggested Medical Management of Persistent Cholestasis
Table 384.1
Inborn Errors of Metabolism That Affect the Liver
DISORDERS OF CARBOHYDRATE METABOLISM
• Disorders of galactose metabolism
• Galactosemia (galactose-1-phosphate uridyltransferase deficiency)
• Disorders of fructose metabolism
• Hereditary fructose intolerance (aldolase deficiency)
• Fructose-1,6 diphosphatase deficiency
• Glycogen storage diseases
• Type I
• Von Gierke Ia (glucose-6-phosphatase deficiency)
• Type Ib (glucose-6-phosphatase transport defect)
• Type III Cori/Forbes (glycogen debrancher deficiency)
• Type IV Andersen (glycogen branching enzyme deficiency)
• Type VI Hers (liver phosphorylase deficiency)
• Congenital disorders of glycosylation (multiple subtypes)
DISORDERS OF AMINO ACID AND PROTEIN METABOLISM
• Disorders of tyrosine metabolism
• Hereditary tyrosinemia type I (fumarylacetoacetate deficiency)
• Tyrosinemia, type II (tyrosine aminotransferase deficiency)
• Inherited urea cycle enzyme defects
• CPS deficiency (carbamoyl phosphate synthetase I deficiency)
• OTC deficiency (ornithine transcarbamoylase deficiency)
• Citrullinemia type I (argininosuccinate synthetase deficiency)
• Argininosuccinic aciduria (argininosuccinate deficiency)
• Argininemia (arginase deficiency)
• N-AGS deficiency (N -acetylglutamate synthetase deficiency)
• Maple serum urine disease (multiple possible defects* )
DISORDERS OF LIPID METABOLISM
• Wolman disease (lysosomal acid lipase deficiency)
• Cholesteryl ester storage disease (lysosomal acid lipase deficiency)
• Homozygous familial hypercholesterolemia (low-density lipoprotein receptor deficiency)
• Gaucher disease type I (β-glucocerebrosidase deficiency)
• Niemann-Pick type C (NPC 1 and 2 mutations)
DISORDERS OF BILE ACID METABOLISM
• Defects in bile acid synthesis (several specific enzyme deficiencies)
• Zellweger syndrome—cerebrohepatorenal (multiple mutations in peroxisome biogenesis genes)
DISORDERS OF METAL METABOLISM
• Wilson disease (ATP7B mutations)
• Hepatic copper overload
• Indian childhood cirrhosis
• Neonatal iron storage disease
DISORDERS OF BILIRUBIN METABOLISM
• Crigler-Najjar (bilirubin-uridine diphosphoglucuronate glucuronosyltransferase mutations)
• Type I
• Type II
• Gilbert disease (bilirubin-uridine diphosphoglucuronate glucuronosyltransferase polymorphism)
• Dubin-Johnson syndrome (multiple drug-resistant protein 2 mutation)
• Rotor syndrome
MISCELLANEOUS
• α1 -Antitrypsin deficiency
• Citrullinemia type II (citrin deficiency)
• Cystic fibrosis (cystic fibrosis transmembrane conductance regulator mutations)
• Erythropoietic protoporphyria (ferrochelatase deficiency)
• Polycystic kidney disease
• Mitochondrial hepatopathies (see Table 383.3 and Chapter 388 )
* Maple syrup urine disease can be caused by mutations in branched-chain α-keto
dehydrogenase, keto acid decarboxylase, lipoamide dehydrogenase, or dihydrolipoamide
dehydrogenase.
Table 384.2
Clinical Manifestations That Suggest the Possibility of
Metabolic Disease
Recurrent vomiting, failure to thrive, short stature
Dysmorphic features
Jaundice, hepatomegaly (±splenomegaly), fulminant hepatic failure, edema/anasarca
Hypoglycemia, organic acidemia, lactic acidemia, hyperammonemia, bleeding (coagulopathy)
Developmental delay/psychomotor retardation, hypotonia, progressive neuromuscular deterioration, seizures,
myopathy, neuropathy
Cardiac dysfunction/failure
Unusual odors
Rickets
Cataracts
384.1
Inherited Deficient Conjugation of
Bilirubin (Familial Nonhemolytic
Unconjugated Hyperbilirubinemia)
CHAPTER 385
Viral Hepatitis
M. Kyle Jensen, William F. Balistreri
Table 385.1
Features of the Hepatotropic Viruses
VIROLOGY HAV RNA HBV DNA HCV RNA HDV RNA HEV RNA
Incubation (days) 15-19 60-180 14-160 21-42 21-63
Transmission
• Parenteral Rare Yes Yes Yes No
• Fecal–oral Yes No No No Yes
• Sexual No Yes Rare Yes No
• Perinatal No Yes Uncommon (5–15%) Yes No
Chronic infection No Yes Yes Yes No
Fulminant disease Rare Yes Rare Yes Yes
HAV, hepatitis A virus; HBV, hepatitis B virus; HCV, hepatitis C virus; HDV, hepatitis D virus; HEV,
hepatitis E virus.
Table 385.2
Causes and Differential Diagnosis of Hepatitis in Children
INFECTIOUS
Hepatotropic viruses
• Hepatitis A virus (HAV)
• Hepatitis B virus (HBV)
• Hepatitis C virus (HCV)
• Hepatitis D virus (HDV)
• Hepatitis E virus (HEV)
• Hepatitis non–A-E viruses
Systemic infection that can include hepatitis
• Adenovirus
• Arbovirus
• Coxsackievirus
• Cytomegalovirus
• Enterovirus
• Epstein-Barr virus
• “Exotic” viruses (e.g., yellow fever)
• Herpes simplex virus
• HIV
• Paramyxovirus
• Rubella
• Varicella zoster
Other
NONVIRAL LIVER INFECTIONS
Abscess
Amebiasis
Bacterial sepsis
Brucellosis
Fitz-Hugh-Curtis syndrome
Histoplasmosis
Leptospirosis
Tuberculosis
Other
AUTOIMMUNE
Autoimmune hepatitis
Sclerosing cholangitis
Other (e.g., systemic lupus erythematosus, juvenile inflammatory arthritis)
METABOLIC
α1 -Antitrypsin deficiency
Tyrosinemia
Wilson disease
Other
TOXIC
Iatrogenic or drug induced (e.g., acetaminophen)
Environmental (e.g., pesticides)
ANATOMIC
Choledochal cyst
Biliary atresia
Other
HEMODYNAMIC
Shock
Congestive heart failure
Budd-Chiari syndrome
Other
NONALCOHOLIC FATTY LIVER DISEASE
Idiopathic
Reye syndrome
Other
From Wyllie R, Hyams JS, editors: Pediatric gastrointestinal and liver disease , ed 3, Philadelphia,
2006, WB Saunders.
Other organ systems can be affected during acute HAV infection. Regional
lymph nodes and the spleen may be enlarged. The bone marrow may be
moderately hypoplastic, and aplastic anemia has been reported. Tissue in the
small intestine might show changes in villous structure, and ulceration of the
gastrointestinal tract can occur, especially in fatal cases. Acute pancreatitis and
myocarditis have been reported, though rarely, and nephritis, arthritis,
leukocytoclastic vasculitis, and cryoglobulinemia can result from circulating
immune complexes.
Diagnosis
Acute HAV infection is diagnosed by detecting antibodies to HAV, specifically,
anti-HAV (immunoglobulin [Ig] M) by radioimmunoassay or, rarely, by
identifying viral particles in stool. A viral polymerase chain reaction (PCR)
assay is available for research use (Table 385.3 ). Anti-HAV is detectable when
the symptoms are clinically apparent, and it remains positive for 4-6 mo after the
acute infection. A neutralizing anti-HAV (IgG) is usually detected within 8 wk of
symptom onset and is measured as part of a total anti-HAV in the serum. Anti-
HAV (IgG) confers long-term protection. Rises in serum levels of ALT, AST,
bilirubin, alkaline phosphatase, 5′-nucleotidase, and γ-glutamyl transpeptidase
are almost universally found and do not help to differentiate the cause of
hepatitis.
Table 385.3
Diagnostic Blood Tests: Serology and Viral Polymerase
Chain Reaction
HAV HBV HCV HDV HEV
ACUTE/ACTIVE INFECTION
Anti-HAV IgM (+) Anti-HBc IgM (+) Anti-HCV (+) Anti-HDV IgM (+) Anti-HEV IgM (+)
Blood PCR HBsAg (+) HCV RNA (+) Blood PCR Blood PCR positive*
positive* Anti-HBs (−) (PCR) positive
HBV DNA (+) HBsAg (+)
(PCR) Anti-HBs (−)
PAST INFECTION (RECOVERED)
Anti-HAV IgG(+) Anti-HBs (+) Anti-HCV (+) Anti-HDV IgG Anti-HEV IgG
Anti-HBc IgG (+) † Blood PCR (−) (+) (+)
Blood PCR (−) Blood PCR (−)
CHRONIC INFECTION
N/A Anti-HBc IgG (+) Anti-HCV (+) Anti-HDV IgG N/A
HBsAg (+) Blood PCR (+) (+)
Anti-HBs (−) Blood PCR (−)
PCR (+) or (−) HBsAg (+)
Anti-HBs (−)
VACCINE RESPONSE
Anti-HAV IgG (+) Anti-HBs (+) N/A N/A N/A
Anti-HBc (−)
* Research tool.
HAV, hepatitis A virus; HBs, hepatitis B surface; HBsAg, hepatitis B surface antigen; Ig,
immunoglobulin; PCR, polymerase chain reaction.
Complications
Although most patients achieve full recovery, distinct complications can occur.
ALF from HAV infection is an infrequent complication of HAV. Those at risk for
this complication are adolescents and adults, but also immunocompromised
patients or those with underlying liver disorders. The height of HAV viremia
may be linked to the severity of hepatitis. In the United States, HAV represents
<0.5% of pediatric-age ALF; HAV is responsible for up to 3% mortality in the
adult population with ALF. In endemic areas of the world, HAV constitutes up to
40% of all cases of pediatric ALF. HAV can also progress to a prolonged
cholestatic syndrome that waxes and wanes over several months. Pruritus and fat
malabsorption are problematic and require symptomatic support with antipruritic
medications and fat-soluble vitamin supplementation. This syndrome occurs in
the absence of any liver synthetic dysfunction and resolves without sequelae.
Treatment
There is no specific treatment for hepatitis A. Supportive treatment consists of
intravenous hydration as needed, and antipruritic agents and fat-soluble vitamins
for the prolonged cholestatic form of disease. Serial monitoring for signs of ALF
is prudent and, if ALF is diagnosed, a prompt referral to a transplantation center
can be lifesaving.
Prevention
Patients infected with HAV are contagious for 2 wk before and approximately 7
days after the onset of jaundice and should be excluded from school, childcare,
or work during this period. Careful hand-washing is necessary, particularly after
changing diapers and before preparing or serving food. In hospital settings,
contact and standard precautions are recommended for 1 wk after onset of
symptoms.
Immunoglobulin
Indications for intramuscular administration of Ig include preexposure and
postexposure prophylaxis (Table 385.4 ). Ig is recommended for preexposure
prophylaxis for susceptible travelers to countries where HAV is endemic, and it
provides effective protection for up to 2 mo. HAV vaccine given any time before
travel is preferred for preexposure prophylaxis in healthy persons, but Ig ensures
an appropriate prophylaxis in children younger than 12 mo old, patients allergic
to a vaccine component, or those who elect not to receive the vaccine. If travel is
planned in <2 wk, older patients, immunocompromised hosts, and those with
chronic liver disease or other medical conditions should receive both Ig and the
HAV vaccine.
Table 385.4
Indications and Updated Dosage Recommendations for
GamaSTAN S/D Human Immune Globulin for Preexposure
and Postexposure Prophylaxis Against Hepatitis A
Infection
Vaccine
The availability of 2 inactivated, highly immunogenic, and safe HAV vaccines
has had a major impact on the prevention of HAV infection. Both vaccines are
approved for children older than 12 mo. They are administered intramuscularly
in a 2-dose schedule, with the second dose given 6-12 mo after the first dose.
Seroconversion rates in children exceed 90% after an initial dose and approach
100% after the second dose; protective antibody titer persists for longer than 10
yr in most patients. The immune response in immunocompromised persons,
older patients, and those with chronic illnesses may be suboptimal; in those
patients, combining the vaccine with Ig for pre- and postexposure prophylaxis is
indicated. HAV vaccine may be administered simultaneously with other
vaccines. A combination HAV and HBV vaccine is approved in adults older than
age 18 yr. For healthy persons at least 12 mo old, vaccine is preferable to Ig for
preexposure and postexposure prophylaxis (see Table 385.3 ).
In the United States and some other countries, universal vaccination is now
recommended for all children older than 12 mo. Nevertheless, studies show
<50% of U.S. adolescents have received even a single dose of the vaccine, and
<30% have received the complete vaccine series. The vaccine is effective in
curbing outbreaks of HAV because of rapid seroconversion and the long
understood. To permit hepatocytes to continue to be infected, the core protein or
major histocompatibility class I protein might not be recognized, the cytotoxic
lymphocytes might not be activated, or some other, yet unknown mechanism
might interfere with destruction of hepatocytes. This tolerance phenomenon
predominates in the perinatally acquired cases, resulting in a high incidence of
persistent HBV infection in children with no or little inflammation in the liver,
normal liver enzymes, and markedly elevated HBV viral load. Although end-
stage liver disease rarely develops in those patients, the inherent HCC risk is
high, possibly related, in part, to uncontrolled viral replication cycles.
ALF has been seen in infants of chronic carrier mothers who have anti-HBe or
are infected with a precore-mutant strain. This fact led to the postulate that
HBeAg exposure in utero in infants of chronic carriers likely induces tolerance
to the virus once infection occurs postnatally. In the absence of this tolerance, the
liver is massively attacked by T cells and the patient presents with ALF.
Immune-mediated mechanisms are also involved in the extrahepatic
conditions that can be associated with HBV infections. Circulating immune
complexes containing HBsAg can result in polyarteritis nodosa, membranous or
membranoproliferative glomerulonephritis, polymyalgia rheumatica,
leukocytoclastic vasculitis, and Guillain-Barré syndrome.
Clinical Manifestations
Many acute cases of HBV infection in children are asymptomatic, as evidenced
by the high carriage rate of serum markers in persons who have no history of
acute hepatitis (Table 385.5 ). The usual acute symptomatic episode is similar to
that of HAV and HCV infections but may be more severe and is more likely to
include involvement of skin and joints (Fig. 385.2 ).
Table 385.5
Typical Interpretation of Test Results for Hepatitis B Virus
Infection
TOTAL IgM
ANTI- HBV
HBsAg ANTI- ANTI- INTERPRETATION
HBs DNA
HBc HBc
− − − − − Never infected
+ − − − + or Early acute infection; transient (up to 18 days) after vaccination
−
+ + + − + Acute infection
− + + + or − + or Acute resolving infection
−
− + − + − Recovered from past infection and immune
+ + − − + Chronic infection
− + − − + or False-positive (i.e., susceptible); past infection; “low-level” chronic
− infection; or passive transfer of anti-HBc to infant born to HBsAg-
positive mother
− − − + − Immune if anti-HBs concentration is ≥10 mIU/mL after vaccine
series completion; passive transfer after hepatitis B immune globulin
administration
−, negative; +, positive; anti-HBc, antibody to hepatitis B core antigen; anti-HBs, antibody to
hepatitis B surface antigen; HBsAg, hepatitis B surface antigen; HBV DNA, hepatitis B virus
deoxyribonucleic acid; IgM, immunoglobulin class M.
From Schillie S, Vellozzi C, Reingold A, et al: Prevention of hepatitis B virus infection in the United
States: recommendations of the advisory committee on immunization practices, MMWR 67(1):1–
29, 2018, Table 1, p. 7.
FIG. 385.2 The serologic course of acute hepatitis B. ALT, alanine aminotransferase;
HBc, hepatitis B core; HBeAg, hepatitis B e antigen; HBsAg, hepatitis B surface
antigen; HBV DNA, hepatitis B virus deoxyribonucleic acid; IgM, immunoglobulin class
M; PCR, polymerase chain reaction. (From Goldman L, Ausiello D: Cecil textbook of
medicine, ed 22, Philadelphia, 2004, WB Saunders, p 914.)
Table 385.6
Causes of Hepatitis Flares in Patients With Chronic
Hepatitis B
Diagnosis
The serologic profile of HBV infection is more complex than for HAV infection
and differs depending on whether the disease is acute or chronic (Fig. 385.3 , see
Table 385.5 ). Several antigens and antibodies are used to confirm the diagnosis
of acute HBV infection (see Table 385.3 ). Routine screening for HBV infection
requires assay of multiple serologic markers (HBsAg, anti-HBc, anti-HBs).
HBsAg is an early serologic marker of infection and is found in almost all
infected persons; its rise closely coincides with the onset of symptoms.
Persistence of HBsAg beyond 6 mo defines the chronic infection state. During
recovery from acute infection, because HBsAg levels fall before symptoms
wane, IgM antibody to HBcAg (anti-HBc IgM) might be the only marker of
acute infection. Anti-HBc IgM rises early after the infection and remains
positive for many months before being replaced by anti-HBc IgG, which then
persists for years. Anti-HBs marks serologic recovery and protection. Only anti-
HBs is present in persons immunized with hepatitis B vaccine, whereas both
anti-HBs and anti-HBc are detected in persons with resolved infection. HBeAg
is present in active acute or chronic infection and is a marker of infectivity. The
development of anti-HBe, termed seroconversion, marks improvement and is a
goal of therapy in chronically infected patients. HBV DNA can be detected in
the serum of acutely infected patients and chronic carriers. High DNA titers are
seen in patients with HBeAg, and they typically fall once anti-HBe develops.
hepatologist experienced in treating the disease.
The goal of treatment is to reduce viral replication defined by having
undetectable HBV DNA in the serum and development of anti-HBe, termed
seroconversion. The development of anti-HBe transforms the disease into an
inactive form, thereby decreasing infectivity, active liver injury and
inflammation, fibrosis progression, and the risk of HCC. Treatment is only
indicated for patients in the immune-active form of the disease, as evidenced by
elevated ALT and/or AST, who have fibrosis on liver biopsy, putting the child at
higher risk for cirrhosis during childhood.
Treatment Strategies
Interferon-α2b (IFN-α2b) has immunomodulatory and antiviral effects (Table
385.7 ). It has been used in children, with long-term viral response rates similar
to the 25% rate reported in adults. Interferon (IFN) use is limited by its
subcutaneous administration, treatment duration of 24 wk, and side effects (flu-
like symptoms, marrow suppression, depression, retinal changes, autoimmune
disorders). IFN is further contraindicated in decompensated cirrhosis. One
advantage of IFN, compared to other treatments, is that viral resistance does not
develop with its use.
Table 385.7
Positive and Negative Factors to Consider in the Decision
to Treat Hepatitis B With Peginterferon or a Nucleoside or
Nucleotide Analog
Table 385.8
Strategy to Eliminate Hepatitis B Virus Transmission in the
United States*
• Screening of all pregnant women for HBsAg
HBV DNA testing for HBsAg-positive pregnant women, with suggestion of maternal antiviral therapy to reduce
perinatal transmission when HBV DNA is >200,000 IU/mL
Prophylaxis (HepB vaccine and hepatitis B immunoglobulin) for infants born to HBsAg-positive † women
• Universal vaccination of all infants beginning at birth ‡ , § as a safeguard for infants born to HBV-infected
mothers not identified prenatally
• Routine vaccination of previously unvaccinated children aged <19 yr
• Vaccination of adults at risk for HBV infection, including those requesting protection from HBV without
acknowledgment of a specific risk factor
* Sources: Mast EE, Margolis HS, Fiore AE, et al: A comprehensive immunization strategy to
eliminate transmission of hepatitis B virus infection in the United States: recommendations of the
Advisory Committee on Immunization Practices (ACIP). Part 1: immunization of infants, children,
and adolescents, MMWR Recomm Rep 54(No. RR-16):1–31, 2005; Mast EE, Weinbaum CM,
Fiore AE, et al: A comprehensive immunization strategy to eliminate transmission of hepatitis B
virus infection in the United States: recommendations of the Advisory Committee on Immunization
Practices (ACIP). Part II: immunization of adults, MMWR Recomm Rep 55(No. RR-16):1–33,
2006.
† Refer to Table 385.9 for prophylaxis recommendations for infants born to women with unknown
HBsAg status.
‡ Within 24 hr of birth for medically stable infants weighing ≥2,000 g.
§ Refer to Table 385.9 for birth dose recommendations for infants weighing <2,000 g.
Table 385.9
Hepatitis B Vaccine Schedules for Infants, by Infant
Birthweight and Maternal Hepatitis B Surface Antigen
Status
SINGLE-ANTIGEN SINGLE-ANTIGEN +
MATERNAL HBsAg VACCINE
BIRTHWEIGHT COMBINATION VACCINE †
STATUS
DOSE AGE DOSE AGE
≥2,000 g Positive 1 Birth (≤12 hr) 1 Birth (≤12 hr)
HBIG Birth (≤12 hr) HBIG Birth (≤12 hr)
‡
2 1-2 mo 2 2 mo
3 6 mo § 3 4 mo
4 6 mo §
Unknown* 1 Birth (≤12 hr) 1 Birth (≤12 hr)
2 1-2 mo 2 2 mo
3 6 mo § 3 4 mo
4 6 mo §
Negative 1 Birth (≤24 hr) 1 Birth (≤24 hr)
2 1-2 mo 2 2 mo
3 6-18 mo § 3 4 mo
4 6 mo §
<2,000 g Positive 1 Birth (≤12 hr) 1 Birth (≤12 hr)
HBIG Birth (≤12 hr) HBIG Birth (≤12 hr)
2 1 mo 2 2 mo
3 2-3 mo 3 4 mo
4 6 mo § 4 6 mo §
Unknown 1 Birth (≤12 hr) 1 Birth (≤12 hr)
HBIG Birth (≤12 hr) HBIG Birth (≤12 hr)
2 1 mo 2 2 mo
3 2-3 mo 3 4 mo
4 6 mo § 4 6 mo §
Negative 1 Hospital discharge 1 Hospital discharge or age 1
or age 1 mo mo
2 2 mo 2 2 mo
3 6-18 mo § 3 4 mo
4 6 mo §
* Mothers should have blood drawn and tested for HBsAg as soon as possible after admission for
delivery; if the mother is found to be HBsAg positive, the infant should receive HBIG as soon as
possible but no later than age 7 days.
† Pediarix should not be administered before age 6 wk.
‡ HBIG should be administered at a separate anatomical site from vaccine.
§ The final dose in the vaccine series should not be administered before age 24 wk (164 days).
Table 385.10
Recommended Doses of Hepatitis B Vaccine by Group and
Vaccine Type
Table 385.11
Hepatitis B Vaccine Schedules for Children, Adolescents,
and Adults
Newer Treatments
Varying IFN-free regimens are now available for adults for all HCV genotypes
allowing even greater likelihood of achieving viral eradication, with completely
oral medication regimens, and without the use of IFN and its attendant side
effects. With the rapid development of new medications and regimens, frequent
review of up-to-date resources, such as www.hcvguidelines.org , will be vital to
provide optimal care (Table 385.12 ).
Table 385.12
Ongoing Studies With Direct Acting Antiviral Combinations
in Children With Chronic Hepatitis C Virus Infection
AGE
ESTIMATED
GENOTYPE RANGE IDENTIFIER COMPLETION
ENROLMENT
(YR)
Sofosbuvir + ledipasvir, with or 1,4,5,6 222 3–17 NCT02249182 July 2018
without ribavirin
Sofosbuvir + ribavirin 2,3 104 3–17 NCT02175758 April 2018
Ombitasvir + paritaprevir + 1,4 74 3–17 NCT02486406 September 2019
ritonavir, with or without
dasabuvir, with or without
ribavirin
Sofosbuvir + daclatasvir 4 40 8–17 NCT03080415 June 2018
Sofosbuvir + ledipasvir 1,4 40 12–17 NCT02868242 April 2019
Sofosbuvir + velpatasvir 1–6 200 3–17 NCT03022981 December 2019
Glecaprevir + pibrentasvir 1–6 110 3–17 NCT03067129 May 2022
Gratisovir + ribavirin 1–6 41 10–17 NCT02985281 June 2017
From Indolfi G, Serranti D, Resti M: Direct-acting antivirals for children and adolescents with
chronic hepatitis C. Lancet Child Adolesc 2:298-304, 2018 (Table 1, p. 299).
Prevention
No vaccine is yet available to prevent HCV, although ongoing research suggests
this will be possible in the future. Currently available Ig preparations are not
beneficial, likely because preparations produced in the United States do not
contain antibodies to HCV because blood and plasma donors are screened for
anti-HCV and excluded from the donor pool. Broad neutralizing antibodies to
HCV were found to be protective and might pave the road for vaccine
development.
Once HCV infection is identified, patients should be screened yearly with a
liver ultrasound and serum α-fetoprotein for HCC, as well as for any clinical
evidence of liver disease. Vaccinating the affected patient against HAV and HBV
will prevent superinfection with these viruses and the increased risk of
developing severe liver failure.
Prognosis
Viral titers should be checked yearly to document spontaneous remission. Most
patients develop chronic hepatitis. Progressive liver damage is higher in those
with additional comorbid factors such as alcohol consumption, viral genotypic
variations, obesity, and underlying genetic predispositions. Referral to a pediatric
hepatologist is strongly advised to take advantage of up-to-date monitoring
regimens and to optimize their enrollment in treatment protocols when available.
Hepatitis D
Etiology
CHAPTER 388
Mitochondrial Hepatopathies
Samar H. Ibrahim, William F. Balistreri
Table 388.1
Genotypic Classification of Primary Mitochondrial
Hepatopathies and Organ Involvement
RESPIRATORY OTHER
HEPATIC
GENE CHAIN ORGANS CLINICAL FEATURES
HISTOLOGY
COMPLEX INVOLVED
Deletion Multiple Steatosis, Kidney, heart, Sideroblastic anemia, variable
(Pearson) fibrosis CNS, muscle thrombocytopenia and neutropenia,
persistent diarrhea
MPV17 I, III, IV Steatosis CNS, muscle, Adult-onset multisystemic involvement:
gastrointestinal myopathy, ophthalmoplegia, severe
tract constipation, parkinsonism
DGUOK I, III, IV Steatosis, Kidneys, CNS, Nystagmus, hypotonia, renal Fanconi
fibrosis muscle syndrome, acidosis
MPV17 I, III, IV Steatosis, CNS, PNS Hypotonia
fibrosis
SUCLG1 I, III, IV Steatosis Kidneys, CNS, Myopathy, sensorineural hearing loss,
muscle respiratory failure
POLG1 I, III, IV Steatosis, CNS, muscle Liver failure preceded by neurologic
fibrosis symptoms, intractable seizures, ataxia,
psychomotor regression
C10orf2/Twinkle I, III, IV Steatosis CNS, muscle Infantile-onset spinocerebellar ataxia, loss
of skills
BCS1L III (GRACILE) CNS ±, muscle Fanconi-type renal tubulopathy
±, kidneys
SCO1 IV Steatosis, Muscle
fibrosis
TRMU I, III, IV Steatosis, Infantile liver failure with subsequent
fibrosis recovery
EFG1 I, III, IV Steatosis CNS Severe, rapidly progressive
encephalopathy
EFTu I, III, IV Unknown CNS Severe lactic acidosis, rapidly fatal
encephalopathy
CNS, central nervous system; GRACILE, growth restriction, aminoaciduria, cholestasis, iron
overload, lactic acidosis, and early death; PNS, peripheral nervous system.
From Lee WS, Sokol RJ: Mitochondrial hepatopathies: advances in genetics, therapeutic
approaches and outcomes, J Pediatr 163:942–948, 2013 (Table 2, p. 944).
Table 388.2
Hepatic Phenotypes of Mitochondrial Cytopathies
• Infantile liver failure
• Neonatal cholestasis
• Pearson syndrome
• Alpers disease
• Chronic liver disease
• Drug-induced mitochondrial toxicity
From Wyllie R, Hyams JS, Kay M, editors: Pediatric gastrointestinal and liver disease , ed 5,
Philadelphia, 2016, Elsevier (Box 71.2, p. 876).
Epidemiology
Mitochondrial respiratory chain disorders of all types affect 1 in 20,000 children
Table 388.3
Clinical Staging of Reye Syndrome and Reye-Like Diseases
Symptoms at the time of admission:
I. Usually quiet, lethargic and sleepy, vomiting, laboratory evidence of liver dysfunction
II. Deep lethargy, confusion, delirium, combativeness, hyperventilation, hyperreflexia
III. Obtunded, light coma ± seizures, decorticate rigidity, intact pupillary light reaction
IV. Seizures, deepening coma, decerebrate rigidity, loss of oculocephalic reflexes, fixed pupils
V. Coma, loss of deep tendon reflexes, respiratory arrest, fixed dilated pupils, flaccidity/decerebration
(intermittent); isoelectric electroencephalogram
Table 388.4
Diseases That Present a Clinical or Pathologic Picture
Resembling Reye Syndrome
• Metabolic disease
• Organic aciduria
• Disorders of oxidative phosphorylation
• Urea cycle defects (carbamoyl phosphate synthetase, ornithine transcarbamylase)
• Defects in fatty acid oxidation metabolism
• Acyl–coenzyme A dehydrogenase deficiencies
• Systemic carnitine deficiency
• Hepatic carnitine palmitoyltransferase deficiency
• 3-OH, 3-methylglutaryl-coenzyme A lyase deficiency
• Fructosemia
• Infantile liver failure syndrome 1. Caused by leucyl-tRNA synthetase (LARS) gene mutations
• Central nervous system infections or intoxications (meningitis), encephalitis, toxic encephalopathy
• Hemorrhagic shock with encephalopathy
• Drug or toxin ingestion (salicylate, valproate)
Diagnostic Evaluation
Screening tests include common biochemical tests (comprehensive metabolic
profile, INR, α-fetoprotein, CPK, phosphorus, complete blood cell count,
ammonia, lactate, pyruvate, serum ketone bodies: both quantitative 3-
hydroxybutyrate and quantitative acetoacetate, total free fatty acids, serum
acylcarnitine profile; serum-free and total carnitines, urine organic acids, and
serum amino acids) (Table 388.5 ). These results will guide subsequent
confirmatory testing to establish a molecular diagnosis. Genotyping, including
single gene or panel screening for common mitochondrial disease, is used in
clinical practice. Whole exome or genome sequencing is also helpful and is
replacing single gene or gene panel testing. However, the identification of
multiple gene variants of uncertain significance will require detailed clinical and
biochemical confirmation for interpretation. Tissue (liver biopsy, skin fibroblast,
and muscle biopsy) may be needed to make a specific biochemical diagnosis.
Table 388.5
TIER 1
Pre-/postprandial plasma lactate, glucose, FFA, and 3-OH
Plasma carnitine, acylcarnitines
Plasma amino acids, creatine kinase, thymidine
Urinary organic acids, amino acids, tubular resorption phosphate, albumin/creatinine ratio CSF lactate/protein (if
feasible)
Electrocardiography and echocardiography
Electroencephalography and visual-evoked potentials
Common mutations in POLG, DGUOK, MPV17, and TRMU
TIER 2
Tissue analysis
Liver biopsy : (if feasible). Tissue for light microscopy, electron microscopy, and Oil Red O stain
Frozen tissue for respiratory chain enzyme activity analysis and mtDNA copy number
Muscle biopsy : Tissue for light microscopy, electron microscopy, Oil Red O stain, and histochemistry for
respiratory chain complexes
Frozen tissue for respiratory chain enzyme activity analysis and mtDNA copy number
Skin biopsy: set up for fibroblast culture
TIER 3
Cranial MRI/MRS
TIER 4
Extended molecular screening. This will be guided by the clinical phenotype, results of the tissue analysis, and
local facilities
Currently suggested genes should include SUCLG1, BCS1L, SOC1, TFSM, TWINKLE, ACAD9, EARS2, GFM1,
RRM2B, TK2 , and SUCLA2
From Wyllie R, Hyams JS, Kay M, editors: Pediatric gastrointestinal and liver
disease , ed 5, Philadelphia, 2016, Elsevier (Box 71-3, p. 876).
Treatment of Mitochondrial
Hepatopathies
There is no effective therapy for most patients with mitochondrial hepatopathies;
neurologic involvement often precludes orthotopic liver transplantation. Patients
with mitochondrial disorders remain at risk for transplant-related worsening of
their underlying metabolic disease, especially patients with POLG -related
disease. Several therapeutic drug combinations―including antioxidants,
vitamins, cofactors, and electron acceptors―have been proposed, but no
randomized controlled trials have been completed to evaluate them. Treatment
strategies are supportive and include the infusion of sodium bicarbonate for
acute metabolic acidosis, transfusions for anemia and thrombocytopenia, and
exogenous pancreatic enzymes for pancreatic insufficiency. It is important to
discontinue or avoid medications that may exacerbate hepatopathy, including
sodium valproate, tetracycline, and macrolide antibiotics, azathioprine,
chloramphenicol, quinolones, and linezolid. Ringer lactate should be avoided
because patients with liver dysfunction may not be able to metabolize lactate.
Propofol should be avoided during anesthesia because of potential interference
with mitochondrial function. In patients with lactic acidosis, lactate levels should
be monitored during procedures. It is important to maintain anabolism using a
balanced intake of fat and carbohydrates while avoiding unbalanced intakes
(e.g., glucose only at a high intravenous rate) or fasting for >12 hr.
Drugs commonly used in children that can cause chronic liver injury, which can
mimic autoimmune hepatitis, include isoniazid, methyldopa, pemoline,
nitrofurantoin, dantrolene, minocycline, pemoline, and the sulfonamides.
Metabolic diseases can lead to chronic hepatitis, including α1 -antitrypsin
deficiency, inborn errors of bile acid biosynthesis, and Wilson disease.
Nonalcoholic steatohepatitis, usually associated with obesity and insulin
resistance, is another common cause of chronic hepatitis. It can progress to
cirrhosis but responds to weight reduction. In many cases the cause of chronic
hepatitis is unknown; in some, an autoimmune mechanism is suggested by the
finding of serum antinuclear and anti–SMAs and by multisystem involvement
(arthropathy, thyroiditis, rashes, Coombs-positive hemolytic anemia).
Table 389.1
Disorders Producing Chronic Hepatitis
• Chronic viral hepatitis
• Hepatitis B
• Hepatitis C
• Hepatitis D
• Autoimmune hepatitis
• Anti–actin antibody-positive
• Anti–liver-kidney microsomal antibody-positive
• Anti–soluble liver antigen antibody-positive
• Others (includes antibodies to liver-specific lipoproteins or asialoglycoprotein)
• Overlap syndrome with sclerosing cholangitis and autoantibodies
• Systemic lupus erythematosus
• Celiac disease
• Drug-induced hepatitis
• Metabolic disorders associated with chronic liver disease
• Wilson disease
• Nonalcoholic steatohepatitis
• α1 -Antitrypsin deficiency
• Tyrosinemia
• Niemann-Pick disease type 2
• Glycogen storage disease type IV
• Cystic fibrosis
• Galactosemia
• Bile acid biosynthetic abnormalities
Table 389.2
Classification of Autoimmune Hepatitis
TYPE 2 AUTOIMMUNE
VARIABLE TYPE 1 AUTOIMMUNE HEPATITIS
HEPATITIS
Characteristic autoantibodies Antinuclear antibody* Antibody against liver-kidney
microsome type 1*
Smooth-muscle antibody*
Antiactin antibody † Antibody against liver cytosol
type 1*
Autoantibodies against soluble liver antigen and Antibody against liver-kidney
liver-pancreas antigen ‡ microsomal type 3
Atypical perinuclear antineutrophil cytoplasmic
antibody
Geographic variation Worldwide Worldwide; rare in North
America
Age at presentation Any age Predominantly childhood and
young adulthood
Gender of patients Female in ~75% of cases Female in ~95% of cases
Association with other Common Common §
autoimmune diseases
Clinical severity Broad range, variable Generally severe
Histopathologic features at Broad range, mild disease to cirrhosis Generally advanced
presentation
Treatment failure Infrequent Frequent
Relapse after drug Variable Common
withdrawal
Need for long-term Variable ~100%
maintenance
* The conventional method of detection is immunofluorescence.
type 2 disease.
Modified from Krawitt EL: Autoimmune hepatitis, N Engl J Med 354:54–66, 2006.
Etiology
T Lymphocytes
hepatitis is being increasingly recognized with wider application of MR
cholangiography (Table 389.3 ). Patients with primary sclerosing cholangitis can
have elevated γ-globulin levels and autoantibodies; therefore liver biopsy
findings in these children may be especially important. Dilated or obliterated
veins on ultrasonography suggest the possibility of the Budd-Chiari syndrome.
Diagnosis of autoimmune liver disease in the setting of acute liver failure is
difficult and care should be taken in applying standardized approaches.
“Seronegative” autoimmune hepatitis has been described, so absence of classic
autoimmune markers does not exclude this diagnosis.
Table 389.3
Overlap Syndromes of Autoimmune Hepatitis
The liver is the main site of drug metabolism and is particularly susceptible to
structural and functional injury after the ingestion, parenteral administration, or
inhalation of chemical agents, drugs, plant derivatives (home remedies), herbal
or nutritional supplements, or environmental toxins. The possibility of drug use
or toxin exposure at home or in the parents’ workplace should be explored for
every child with liver dysfunction. The clinical spectrum of illness can vary from
asymptomatic biochemical abnormalities of liver function to fulminant failure.
Liver injury may be the only clinical feature of an adverse drug reaction or may
be accompanied by systemic manifestations and damage to other organs. In
hospitalized patients, clinical and laboratory findings may be confused with the
underlying illness. After acetaminophen, antimicrobials, supplements, and
central nervous system agents are the most commonly implicated drug classes
causing liver injury in children.
There is growing concern about environmental hepatotoxins that are insidious
in their effects. Many environmental toxins―including the plasticizers, biphenyl
A, and the phthalates―are ligands for nuclear receptors that transcriptionally
activate the promoters of many genes involved in xenobiotic and lipid
metabolism and may contribute to obesity and nonalcoholic fatty liver disease.
Some herbal, weight loss, and body building supplements have been associated
with hepatic injury or even liver failure (Table 390.1 ) related to their intrinsic
toxicity or because of contamination with fungal toxins, pesticides, or heavy
metals.
Table 390.1
Hepatotoxic Herbal Remedies, Dietary Supplements, and
Weight Loss Products
POPULAR HEPATOTOXIC TYPE OF LIVER
REMEDY SOURCE
USES COMPONENT INJURY
Ayurvedic herbal Multiple Multiple Uncertain (may Hepatitis
medicine contain heavy metal
contaminants)
Barakol Anxiolytic Cassia siamea Uncertain Reversible hepatitis or
cholestasis
Black cohosh Menopausal Cimicifuga racemosa Uncertain Hepatitis (causality
symptoms uncertain)
“Bush tea” Fever Senecio, Pyrrolizidine alkaloids SOS
Heliotropium,
Crotalaria spp.
Cascara Laxative Cascara sagrada Anthracene glycoside Cholestatic hepatitis
Chaparral leaf “Liver tonic,” Larrea tridenta Nordihydroguaiaretic Acute and chronic
(greasewood, burn salve, acid hepatitis, FHF
creosote bush) weight loss
Chaso/onshido Weight loss — N -nitro-fenfluramine Acute hepatitis, FHF
Chinese medicines
(traditional)
Jin bu huan Sleep aid, Lycopodium serratum Levo- Acute or chronic hepatitis
analgesic tetrahydropalmitine or cholestasis, steatosis
Ma huang Weight loss Ephedra spp. Ephedrine Severe hepatitis, FHF
Shou-wu-pian Anti-aging, Polygonum Anthraquinone Acute hepatitis or
neuroprotection, multiflorum Thunb cholestasis
laxative (fleeceflower root)
Syo-saiko-to Multiple Scutellaria root Diterpenoids Hepatocellular necrosis,
cholestasis, steatosis,
granulomas
Comfrey Herbal tea Symphytum spp. Pyrrolizidine alkaloid Acute SOS, cirrhosis
Germander Weight loss, Teucrium chamaedry, Diterpenoids, epoxides Acute and chronic
fever T. capitatum, T. hepatitis, FHF,
polium autoimmune injury
Greater celandine Gallstones, IBS Chelidonium majus Isoquinoline alkaloids Cholestatic hepatitis,
fibrosis
Green tea leaf Multiple Camellia sinensis Catechins Hepatitis (causality
extract questioned)
Herbalife Nutritional — Various; ephedra Severe hepatitis, FHF
supplement,
weight loss
Hydroxycut Weight loss Camellia sinensis , Uncertain Acute hepatitis, FHF
among other
constituents
Impila Multiple Callilepsis laureola Potassium atractylate Hepatic necrosis
Kava Anxiolytic Piper methysticum Kava lactone, Acute hepatitis,
pipermethystine cholestasis, FHF
Kombucha Weight loss Lichen alkaloid Usnic acid Acute hepatitis
Limbrel Osteoarthritis Plant bioflavonoids Baicalin, epicatechin Acute mixed
(Flavocoxid) hepatocellular-cholestatic
injury
Lipokinetix Weight loss Lichen alkaloid Usnic acid Acute hepatitis, jaundice,
FHF
Mistletoe Asthma, Viscus album Uncertain Hepatitis (in combination
infertility with skullcap)
Oil of cloves Dental pain Various foods, oils Eugenol Zonal necrosis
Pennyroyal Abortifacient Hedeoma pulegoides, Pulegone, Severe hepatocellular
(squawmint oil) Mentha pulegium monoterpenes necrosis
Prostata Prostatism Multiple Uncertain Chronic cholestasis
Sassafras Herbal tea Sassafras albidum Safrole HCC (in animals)
Senna Laxative Cassia angustifolia Sennoside alkaloids; Acute hepatitis
anthrone
Skullcap Anxiolytic Scutellaria Diterpenoids Hepatitis
Valerian Sedative Valeriana officinalis Uncertain Elevated liver enzymes
FHF, fulminant hepatic failure; HCC, hepatocellular carcinoma; SOS, sinusoidal obstruction
syndrome.
From Lewis JH: Liver disease caused by anesthetics, chemicals, toxins, and herbal preparations.
In Feldman M, Friedman LS, Brandt LJ, editors: Sleisenger and Fordtran's gastrointestinal and
liver disease , ed 10, Philadelphia, 2016, Elsevier, (Table 89.6).
Table 390.2
Patterns of Hepatic Drug Injury
DISEASE DRUG
Centrilobular necrosis Acetaminophen
Carbon tetrachloride
Cocaine
Ecstasy
Iron
Halothane
Microvesicular steatosis Valproic acid
Tetracycline
Toluene
Methotrexate
Acute hepatitis Isoniazid
Anti–tumor necrosis factor agents
Valproic acid
General hypersensitivity Sulfonamides
Phenytoin
Minocycline
Fibrosis Methotrexate
Cholestasis Chlorpromazine
Aniline
Erythromycin
Paraquat
Estrogens
Sertraline
Sinusoidal obstruction syndrome (venoocclusive disease) Irradiation plus busulfan
Arsenic
Cyclophosphamide
Portal and hepatic vein thrombosis Estrogens
Androgens
Biliary sludge Ceftriaxone
Hepatic adenoma or hepatocellular carcinoma Oral contraceptives
Anabolic steroids
Treatment
Treatment of drug- or toxin-related liver injury is mainly supportive. Contact
with the offending agent should be avoided. Corticosteroids might have a role in
immune-mediated disease. Treatment with n -acetylcysteine, by stimulating
glutathione synthesis, is effective in preventing or attenuating hepatotoxicity
when administered within 16 hr after an acute overdose of acetaminophen and
appears to improve survival in patients with severe liver injury even up to 36 hr
after ingestion (see Chapter 63 ). Intravenous L -carnitine may be of value in
Irritability, poor feeding, and a change in sleep rhythm may be the only findings
in infants; asterixis may be demonstrable in older children. Patients are often
somnolent, confused, or combative on arousal and can eventually become
responsive only to painful stimuli. Patients can rapidly progress to deeper stages
of coma in which extensor responses and decerebrate and decorticate posturing
appear. Respirations are usually increased early, but respiratory failure can occur
in stage IV coma (Table 391.1 ). The pathogenesis of hepatic encephalopathy is
likely related to increased serum levels of ammonia, false neurotransmitters,
amines, increased γ-aminobutyric acid receptor activity, or increased circulating
levels of endogenous benzodiazepine-like compounds. Decreased hepatic
clearance of these substances can produce marked central nervous system
dysfunction. The mechanisms responsible for cerebral edema and intracranial
hypertension in acute liver failure (ALF) suggest both cytotoxic and vasogenic
injury. There is increasing evidence for an inflammatory response (synthesis and
release of inflammatory factors from activated microglia and endothelial cells)
which acts in synergy with hyperammonemia to cause severe astrocyte
swelling/brain edema.
Table 391.1
Stages of Hepatic Encephalopathy
STAGES
I II III IV
Symptoms Periods of lethargy, Drowsiness, Stupor but Coma:
euphoria; reversal of inappropriate behavior, arousable; IVa responds to
day-night sleeping; agitation, wide mood confused, noxious stimuli;
may be alert swings, disorientation incoherent IVb no response
speech
Signs Trouble drawing Asterixis, fetor Asterixis, Areflexia, no
figures, performing hepaticus, incontinence hyperreflexia, asterixis, flaccidity
mental tasks extensor
reflexes,
rigidity
Electroencephalogram Normal Generalized slowing, q Markedly Markedly abnormal
waves abnormal bilateral slowing, d
triphasic waves waves, electrocortical
silence
Laboratory Findings
Serum direct and indirect bilirubin levels and serum aminotransferase activities
CHAPTER 392
Cystic lesions of liver may be initially recognized during infancy and childhood.
Hepatic fibrosis can also occur as part of an associated developmental defect
(Table 392.1 ). Cystic renal disease is usually associated and often determines
the clinical presentation and prognosis. Virtually all proteins encoded by genes
mutated in combined cystic diseases of the liver and kidney are at least partially
localized to primary cilia in renal tubular cells and cholangiocytes.
Table 392.1
A solitary, congenital liver cyst (nonparasitic) can occur in childhood and has
been identified in some cases on prenatal ultrasound. Abdominal distention and
pain may be present, and a poorly defined right-upper-quadrant mass may be
palpable. These benign lesions are best left undisturbed unless they compress
adjacent structures or a complication occurs, such as hemorrhage into the cyst.
Operative management is generally reserved for symptomatic patients and
enlarging cysts.
Choledochal Cysts
Choledochal cysts are congenital dilatations of the common bile duct that can
cause progressive biliary obstruction and biliary cirrhosis. Cylindrical (fusiform)
and spherical (saccular) cysts of the extrahepatic ducts are the most common
types (see Table 392.1 ). Choledochal cysts are classified according to the
Todani method (Fig. 392.1 ). Type 1 choledochal cysts, the most common
variant, involve a saccular or fusiform dilation of the common bile duct. Type II
cysts are congenital diverticula protruding from the common bile duct. Type III
cysts or choledochoceles involve a herniation of the intraduodenal segment of
the common bile duct into the duodenum. Type IVa cysts or Caroli disease
involve multiple intrahepatic and extrahepatic cysts. Type IVb cysts involve only
the extrahepatic duct. Solitary liver cysts (type V) are very rare.
CHAPTER 393
Anomalies
The gallbladder is congenitally absent in approximately 0.1% of the population.
Hypoplasia or absence of the gallbladder can be associated with extrahepatic
biliary atresia or cystic fibrosis. Duplication of the gallbladder occurs rarely.
Gallbladder ectopia may occur with a transverse, intrahepatic, left-sided, or
retroplaced location. Multiseptate gallbladder, characterized by the presence of
multiple septa dividing the gallbladder lumen, is another rare congenital
anomaly of the gallbladder.
Acute Hydrops
Table 393.1 lists the conditions associated with hydrops of the gallbladder.
Table 393.1
Conditions Associated With Hydrops of the Gallbladder
Cholelithiasis
Cholecystitis
Kawasaki disease
Streptococcal pharyngitis
Staphylococcal infection
Leptospirosis
Ascariasis
Threadworm
Sickle cell crisis
Typhoid fever
Thalassemia
Total parenteral nutrition
Prolonged fasting
Viral hepatitis
Sepsis
Henoch-Schönlein purpura
Mesenteric adenitis
Necrotizing enterocolitis
Table 393.2
Conditions Associated With Cholelithiasis
Chronic hemolytic disease (sickle cell anemia, spherocytosis, thalassemia, Gilbert disease)
Ileal resection or disease
Cystic fibrosis
Cirrhosis
Cholestasis
Crohn disease
Obesity
Insulin resistance
Prolonged parenteral nutrition
Prematurity with complicated medical or surgical course
Prolonged fasting or rapid weight reduction
Treatment of childhood cancer
Abdominal surgery
Pregnancy
Sepsis
Genetic (ABCB4, ABCG5/G8) progressive familial intrahepatic cholestasis
Cephalosporins
Etiology
Portal hypertension can result from obstruction to portal blood flow anywhere
along the course of the portal venous system (prehepatic, intrahepatic, or
posthepatic). Table 394.1 outlines the various disorders associated with portal
hypertension.
Table 394.1
Causes of Portal Hypertension
EXTRAHEPATIC PORTAL HYPERTENSION
Portal vein agenesis, atresia, stenosis
Portal vein thrombosis or cavernous transformation
Splenic vein thrombosis
Increased portal flow
Arteriovenous fistula
INTRAHEPATIC PORTAL HYPERTENSION
Hepatocellular disease
Acute and chronic viral hepatitis
Cirrhosis
Congenital hepatic fibrosis
Wilson disease
α1 -Antitrypsin deficiency
Glycogen storage disease type IV
Hepatotoxicity
Methotrexate
Parenteral nutrition
Biliary tract disease
Extrahepatic biliary atresia
Cystic fibrosis
Choledochal cyst
Sclerosing cholangitis
Intrahepatic bile duct paucity
Idiopathic portal hypertension
Postsinusoidal obstruction
Budd-Chiari syndrome
Venoocclusive disease
Ascites
Asim Maqbool, Jessica W. Wen, Chris A. Liacouras
Table 397.1
Causes of Fetal Ascites
Gastrointestinal disorders
Meconium peritonitis
Intestinal malrotation
Small intestinal or colonic atresia
Intussusception
Volvulus
Cystic fibrosis
Biliary atresia
Portal venous malformations
Infection
Parvovirus
Syphilis
Cytomegalovirus
Toxoplasmosis
Acute maternal hepatitis
Genitourinary disorders
Hydronephrosis
Polycystic kidney disease
Urinary obstruction
Ovarian cyst
Persistent cloaca
Chylous ascites
Cardiac disorders
Arrhythmia
Heart failure
Chromosomal abnormalities
Trisomy
Turner syndrome
Neoplasm
Hematologic
Hemolytic anemia
Neonatal hemochromatosis
Metabolic disease
Niemann-Pick type C
Congenital disorders of glycosylation
Wolman disease
Lysosomal storage disease
Other
Maternal/fetal abuse
Idiopathic
From Giefer MJ, Murray KF, Colletti RB: Pathophysiology, diagnosis, and management of
pediatric ascites, J Pediatr Gastroenterol Nutr 52(5):503–513, 2011 (Table 1).
Table 397.2
Causes of Neonatal Ascites
Hepatobiliary disorders
Cirrhosis
Alpha-1-antitrypsin deficiency
Congenital hepatic fibrosis
Viral hepatitis
Budd-Chiari syndrome
Biliary atresia
Bile duct perforation
Portal venous malformation
Ruptured mesenchymal hamartoma
Gastrointestinal disorders
Intestinal malrotation
Intestinal perforation
Acute appendicitis
Intestinal atresia
Pancreatitis
Chylous ascites
Intestinal lymphangiectasia
Lymphatic duct obstruction
Lymphatic duct trauma
Parenteral nutrition extravasation
Metabolic disease
Genitourinary disorders
Obstructive uropathy
Posterior urethral valves
Ureterocele
Lower ureteral stenosis
Ureteral atresia
Imperforate hymen
Bladder rupture
Bladder injury from umbilical artery catheterization
Nephrotic syndrome
Ruptured corpus luteum cyst
Cardiac
Arrhythmia
Heart failure
Hematologic
Neonatal hemochromatosis
Other
Cutis marmorata telangiectatica congenita
Intravenous vitamin E
Pseudo-ascites
Small bowel duplication
Abdominal trauma
Idiopathic
From Giefer MJ, Murray KF, Colletti RB: Pathophysiology, diagnosis, and management of
pediatric ascites, J Pediatr Gastroenterol Nutr 52(5):503–513, 2011 (Table 2).
Table 397.3
Causes of Ascites in Infants and Children
Hepatobiliary disorders Neoplasm Neoplasm
Cirrhosis Lymphoma
Congenital hepatic fibrosis Wilms tumor
Acute hepatitis Clear cell renal sarcoma
Budd-Chiari syndrome Glioma
Bile duct perforation Germ cell tumor
Liver transplantation Ovarian tumor
Gastrointestinal disorders Mesothelioma
Acute appendicitis Neuroblastoma
Intestinal atresia Metabolic disease
Pancreatitis Genitourinary disorders
Pyloric duplication Nephrotic syndrome
Serositis Peritoneal dialysis
Crohn disease Cardiac
Eosinophilic enteropathy Heart failure
Henoch-Schönlein purpura Pseudo-ascites
Chylous ascites Celiac disease
Intestinal lymphangiectasia Cystic mesothelioma
Lymphatic duct obstruction Omental cyst
Lymphatic duct trauma Ovarian cyst
Parenteral nutrition extravasation Other
Infectious Systemic lupus erythematosus
Tuberculosis Ventriculoperitoneal shunt
Abscess Vitamin A toxicity
Schistosomiasis Chronic granulomatous disease
Nonaccidental trauma
Protein losing enteropathy
Idiopathic
Modified from Giefer MJ, Murray KF, Colletti RB: Pathophysiology, diagnosis, and management of
pediatric ascites, J Pediatr Gastroenterol Nutr 52(5):503–513, 2011 (Table 3).
clubbing is a sign of chronic hypoxia and chronic lung disease (Fig. 400.3 ) but
may be a result of nonpulmonary etiologies (Table 400.2 ).
Table 400.1
Respiratory Sounds
BASIC
MECHANISMS ORIGIN ACOUSTICS RELEVANCE
SOUNDS
Lung Turbulent flow, Central (expiration), Low pass filtered noise (<100 to Regional
vortices, other lobar to segmental >1,000 Hz) ventilation,
airways (inspiration) airway caliber
Tracheal Turbulent flow, Pharynx, larynx, trachea, Noise with resonances (<100 to Upper airway
flow impinging on large airways >3,000 Hz) configuration
airway walls
ADVENTITIOUS SOUNDS
Wheezes Airway wall flutter, Central and lower Sinusoidal (<100 to >1,000 Hz, Airway
vortex shedding, airways duration typically >80 msec) obstruction, flow
other limitation
Rhonchi Rupture of fluid Larger airways Series of rapidly dampened Secretions,
films, airway wall sinusoids (typically <300 Hz and abnormal airway
vibration duration <100 msec) collapsibility
Crackles Airway wall stress- Central and lower Rapidly dampened wave Airway closure,
relaxation airways deflections (duration typically <20 secretions
msec)
Modified from Pasterkamp H, Kraman SS, Wodicka GR: Respiratory sounds. Advances beyond
the stethoscope, Am J Respir Crit Care Med 156[3]:974–987, 1997.
FIG. 400.3 A, Normal and clubbed finger viewed in profile. B, The normal
finger demonstrates a distal phalangeal finger depth (DPD)
/interphalangeal finger depth (IPD) ratio <1. The clubbed finger
demonstrates a DPD/IPD ratio >1. C, The normal finger on the left
demonstrates a normal profile (abc) with angle less than 180 degrees. The
clubbed finger demonstrates a profile angle >180 degrees. D, Schamroth
sign is demonstrated in the clubbed finger with the loss of diamond shape
window in between finger beds (arrow) that is demonstrated in the normal
finger. (From Wilmott RW, Bush A, Deterding RR, et al: Kendig's disorders
of the respiratory tract in children , ed 9, Philadelphia, 2019, Elsevier [Fig.
1.14, p. 20].)
Table 400.2
Nonpulmonary Diseases Associated With Clubbing
CARDIAC
Cyanotic congenital heart disease
Bacterial endocarditis
Chronic heart failure
HEMATOLOGIC
Thalassemia
Congenital methemoglobinemia (rare)
GASTROINTESTINAL
Crohn disease
Ulcerative colitis
Celiac disease
Chronic dysentery, sprue
Polyposis coli
Severe gastrointestinal hemorrhage
Small bowel lymphoma
Liver cirrhosis (including α1 -antitrypsin deficiency)
Chronic active hepatitis
OTHER
Thyroid deficiency (thyroid acropachy)
Thyrotoxicosis
Chronic pyelonephritis (rare)
Toxic (e.g., arsenic, mercury, beryllium)
Lymphomatoid granulomatosis
Fabry disease
Raynaud disease, scleroderma
Hodgkin disease
Familial
UNILATERAL CLUBBING
Vascular disorders (e.g., subclavian arterial aneurysm, brachial arteriovenous fistula)
Subluxation of shoulder
Median nerve injury
Local trauma
From Pasterkamp H: The history and physical examination. In Wilmott RW, Boat TF, Bush A, et al,
editors: Kendig and Chernick's disorders of the respiratory tract in children, ed 8, Philadelphia,
2012, Elsevier.
Table 400.3
Table 401.1
Indicators of Serious Chronic Lower Respiratory Tract
Disease in Children
Persistent fever
Ongoing limitation of activity
Failure to grow
Failure to gain weight appropriately
Clubbing of the digits
Persistent tachypnea and labored ventilation
Shortness of breath and exercise intolerance
Chronic purulent sputum
Persistent hyperinflation
Substantial and sustained hypoxemia
Refractory infiltrates on chest x-ray
Persistent pulmonary function abnormalities
Hemoptysis
Family history of heritable lung disease
Cyanosis and hypercarbia
Unusual (opportunistic) or recurrent nonpulmonary infections
Table 401.2
Differential Diagnosis of Recurrent and Persistent Cough in
Children
RECURRENT COUGH
Reactive airway disease (asthma)
Drainage from upper airways
Aspiration
Frequently recurring respiratory tract infections in immunocompetent or immunodeficient patients
Symptomatic Chiari malformation
Idiopathic pulmonary hemosiderosis
Hypersensitivity (allergic) pneumonitis
PERSISTENT COUGH
Hypersensitivity of cough receptors after infection
Reactive airway disease (asthma)
Chronic sinusitis
Chronic rhinitis (allergic or nonallergic)
Bronchitis or tracheitis caused by infection or smoke exposure
Bronchiectasis, including cystic fibrosis, primary ciliary dyskinesia, immunodeficiency
Habit cough
Foreign-body aspiration
Recurrent aspiration owing to pharyngeal incompetence, tracheolaryngoesophageal cleft, or tracheoesophageal
fistula
Gastroesophageal reflux, with or without aspiration
Pertussis
Extrinsic compression of the tracheobronchial tract (vascular ring, neoplasm, lymph node, lung cyst)
Tracheomalacia, bronchomalacia
Endobronchial or endotracheal tumors
Endobronchial tuberculosis
Hypersensitivity pneumonitis
Fungal infections
Inhaled irritants, including tobacco smoke
Irritation of external auditory canal
Angiotensin-converting enzyme inhibitors
Table 401.3
Modified from Chang AB, Landau LI, Van Asperen PP, et al: Cough in children: definitions and
clinical evaluation. Thoracic Society of Australia and New Zealand, Med J Aust 184(8):398–403,
2006, Table 2, p. 399.
Table 401.4
Table 401.5
Causes of Recurrent or Persistent Stridor in Children
RECURRENT
Allergic (spasmodic) croup
Respiratory infections in a child with otherwise asymptomatic anatomic narrowing of the large airways
Laryngomalacia
PERSISTENT
Laryngeal obstruction
• Laryngomalacia
• Papillomas, hemangiomas, other tumors
• Cysts and laryngoceles
• Laryngeal webs
• Bilateral abductor paralysis of the cords
• Foreign body
Tracheobronchial disease
• Tracheomalacia
• Subglottic tracheal webs
• Endobronchial, endotracheal tumors
• Subglottic tracheal stenosis, congenital or acquired
Extrinsic masses
• Mediastinal masses
• Vascular ring
• Lobar emphysema
• Bronchogenic cysts
• Thyroid enlargement
• Esophageal foreign body
Tracheoesophageal fistula
OTHER
Gastroesophageal reflux
Macroglossia, Pierre Robin syndrome
Cri-du-chat syndrome
Paradoxical vocal cord dysfunction
Hypocalcemia
Vocal cord paralysis
Chiari crisis
Severe neonatal episodic laryngospasm caused by SCN4A mutation
should suggest foreign-body aspiration.
Either wheezing or coughing when associated with tachypnea and hypoxemia
may be suggestive of interstitial lung disease (see Chapter 427.5 ). However,
many patients with interstitial lung disease demonstrate no symptoms other than
rapid breathing on initial physical examination. Although chest roentgenograms
may be normal in interstitial lung disease, diffuse abnormalities on chest X-ray
may support further evaluation in patients suspected to have interstitial lung
disease with characteristic findings described on high-resolution CT scan and
lung biopsy.
Repeated examination may be required to verify a history of wheezing in a
child with episodic symptoms and should be directed toward assessing air
movement, ventilatory adequacy, and evidence of chronic lung disease, such as
fixed overinflation of the chest, growth failure, and digital clubbing. Patients
should be assessed for oropharyngeal dysphagia in cases of suspected recurrent
aspiration. Clubbing suggests chronic lung infection and is rarely prominent in
uncomplicated asthma. Tracheal deviation from foreign body aspiration should
be sought. It is essential to rule out wheezing secondary to congestive heart
failure. Allergic rhinitis, urticaria, eczema, or evidence of ichthyosis vulgaris
suggests asthma or asthmatic bronchitis. The nose should be examined for
polyps, which can exist with allergic conditions or cystic fibrosis.
Sputum eosinophilia and elevated serum immunoglobulin E levels suggest
allergic reactions. A forced expiratory volume in 1 sec increase of 15% in
response to bronchodilators confirms reactive airways. Specific microbiologic
studies, special imaging studies of the airways and cardiovascular structures,
diagnostic studies for cystic fibrosis, and bronchoscopy should be considered if
the response is unsatisfactory.
Table 401.6
Diseases Associated With Recurrent, Persistent, or
Migrating Lung Infiltrates Beyond the Neonatal Period
Aspiration
Pharyngeal incompetence (e.g., cleft palate)
Laryngotracheoesophageal cleft
Tracheoesophageal fistula
Gastroesophageal reflux
Lipid aspiration
Neurologic dysphagia
Developmental dysphagia
Congenital anomalies
Lung cysts (cystic adenomatoid malformation)
Pulmonary sequestration
Bronchial stenosis or aberrant bronchus
Vascular ring
Congenital heart disease with large left-to-right shunt
Pulmonary lymphangiectasia
Genetic conditions
α1 -Antitrypsin deficiency
Cystic fibrosis
Primary ciliary dyskinesia (including Kartagener syndrome)
Sickle cell disease (acute chest syndrome)
Immunodeficiency, phagocytic deficiency
Humoral, cellular, combined immunodeficiency states
Chronic granulomatous disease and related phagocytic defects
Hyper immunoglobulin E syndromes
Complement deficiency states
Immunologic and autoimmune diseases
Asthma
Allergic bronchopulmonary aspergillosis
Hypersensitivity pneumonitis
Pulmonary hemosiderosis
Collagen-vascular diseases
Infection, congenital
Cytomegalovirus
Rubella
Syphilis
Infection, acquired
Cytomegalovirus
Tuberculosis
HIV
Other viruses
Chlamydia
Mycoplasma, Ureaplasma
Pertussis
Fungal organisms
Pneumocystis jiroveci
Visceral larva migrans
Inadequately treated bacterial infection
Interstitial pneumonitis and fibrosis
Usual interstitial pneumonitis
Lymphoid (AIDS)
Genetic disorders of surfactant synthesis, secretion
Desquamative
Acute (Hamman-Rich)
Alveolar proteinosis
Drug-induced, radiation-induced inflammation and fibrosis
Neoplasms and neoplastic-like conditions
Primary or metastatic pulmonary tumors
Leukemia
Histiocytosis
Eosinophilic pneumonias
Other etiologies
Bronchiectasis
Congenital
Postinfectious
Sarcoidosis
Symptoms associated with chronic lung infiltrates in the 1st several weeks of
life (but not related to neonatal respiratory distress syndrome) suggest infection
acquired in utero or during descent through the birth canal. Early appearance of
chronic infiltrates can also be associated with cystic fibrosis or congenital
anomalies that result in aspiration or airway obstruction. A history of recurrent
infiltrates such as in middle lobe syndrome (see Chapters 430 and 437 ),
wheezing, and cough may reflect asthma, even in the 1st yr of life.
A controversial association has been posed regarding recurrent lung infiltrates
in pulmonary hemosiderosis related to cow's milk hypersensitivity or unknown
causes appearing in the 1st yr of life. Children with a history of
bronchopulmonary dysplasia often have episodes of respiratory distress attended
by wheezing and new lung infiltrates. Recurrent pneumonia in a child with
frequent otitis media, nasopharyngitis, adenitis, or dermatologic manifestations
suggests an immunodeficiency state, complement deficiency, or phagocytic
defect (see Chapters 148 , 156 , and 160 ). Primary ciliary dyskinesia is also of
consideration in patients with frequent otitis media and suppurative
sinopulmonary disease, with or without accompanying heterotaxy, or history of
neonatal respiratory distress (see Chapter 433 ). Pulmonary sequestration may be
suspected in patients with recurrent findings on radiograph that occur in the
same location, both during illness and when well (see Chapter 423 ). Traction
bronchiectasis may also be suggested on radiography with persistent findings in
a given region of the film following a history of respiratory infection. Particular
attention must be directed to the possibility that the infiltrates represent
lymphocytic interstitial pneumonitis or opportunistic infection associated with
HIV infection (see Chapter 302 ). A history of paroxysmal coughing in an infant
suggests pertussis syndrome or cystic fibrosis. Persistent infiltrates, especially
with loss of volume, in a toddler may suggest foreign-body aspiration.
401.1
Extrapulmonary Diseases With
Pulmonary Manifestations
Susanna A. McColley
Table 401.7
Respiratory Signs and Symptoms Originating From
Outside the Respiratory Tract
SIGN OR NONRESPIRATORY
PATHOPHYSIOLOGY CLUES TO DIAGNOSIS
SYMPTOM CAUSE(S)
Chest pain Cardiac disease Inflammation (pericarditis), Precordial pain, friction rub on
ischemia (anomalous coronary examination; exertional pain,
artery, vascular disease) radiation to arm or neck
Chest pain Gastroesophageal Esophageal inflammation and/or Heartburn, abdominal pain
reflux disease spasm
Cyanosis Congenital heart Right-to-left shunt Neonatal onset, lack of response to
disease oxygen
Methemoglobinemia Increased levels of methemoglobin Drug or toxin exposure, lack of
interfere with delivery of oxygen to response to oxygen
tissues
Dyspnea Toxin exposure, drug Variable, but often metabolic Drug or toxin exposure confirmed
side effect, or overdose acidosis by history or toxicology screen,
normal oxygen saturation measured
by pulse oximetry
Anxiety, panic disorder Increased respiratory drive and Occurs during stressful situation,
increased perception of respiratory other symptoms of anxiety or
efforts depression
Exercise Anemia Inadequate oxygen delivery to Pallor, tachycardia, history of
intolerance tissues bleeding, history of inadequate diet
Exercise Deconditioning Self-explanatory History of inactivity, obesity
intolerance
Hemoptysis Nasal bleeding Posterior flow of bleeding causes History and physical findings
appearance of pulmonary origin suggest nasal source; normal chest
examination and chest radiography
Upper gastrointestinal Hematemesis mimics hemoptysis History and physical examination
tract bleeding suggest gastrointestinal source;
normal chest examination and chest
radiography
Wheezing, Congenital or acquired Pulmonary overcirculation Murmur
cough, cardiac disease (atrioseptal defect, ventriculoseptal Refractory to bronchodilators
dyspnea defect, patent ductus arteriosus), left Radiographic changes (prominent
ventricular dysfunction pulmonary vasculature, pulmonary
edema)
Wheezing, Gastroesophageal Laryngeal and bronchial response to Emesis, pain, heartburn
cough reflux disease stomach contents
Vagally mediated Refractory to bronchodilators
bronchoconstriction
Evaluation
In the evaluation of a child or adolescent with respiratory symptoms, it is
important to obtain a detailed past medical history, family history, and review of
systems to evaluate the possibility of extrapulmonary origin. A comprehensive
physical examination is also essential in obtaining clues as to extrapulmonary
disease.
Disorders of other organ systems, and many systemic diseases, can have
significant respiratory system involvement. Although it is most common to
encounter these complications in patients with known diagnoses, respiratory
system disease is sometimes the sole or most prominent symptom at the time of
presentation. Acute aspiration during feeding can be the presentation of
neuromuscular disease in an infant who initially appears to have normal muscle
tone and development. Complications can be life-threatening, particularly in
immunocompromised patients. The onset of respiratory findings may be
insidious; for example, pulmonary vascular involvement in patients with
systemic vasculitis may appear as an abnormality in diffusing capacity of the
lung for carbon monoxide before the onset of symptoms. Table 401.8 lists
disorders that commonly have respiratory complications.
Table 401.8
Bibliography
Gaude GS. Hemoptysis in children. Indian Pediatr .
2010;47(3):245–254.
Jindal A, Singhi S. Acute chest pain. Indian J Pediatr .
2011;78(10):1262–1267.
Loughlin GM. Chest pain. Loughlin GM, Eigen H. Respiratory
disease in children: diagnosis and management . Williams &
Wilkins: Baltimore; 1994:207–214.
CHAPTER 402
Table 402.1
Differential Diagnosis of Sudden Unexpected Infant Death
PRIMARY FREQUENCY
CAUSE OF POTENTIAL CONFOUNDING
DIAGNOSTIC DISTRIBUTION
DEATH DIAGNOSES
CRITERIA (%)
EXPLAINED AT AUTOPSY
Natural 18–20*
Infections History, autopsy, and If minimal findings: SIDS 35–46 †
cultures
Congenital anomaly History and autopsy If minimal findings: SIDS 14–24 †
Unintentional injury History, scene Traumatic child abuse 15*
investigation, autopsy
Traumatic child Autopsy and scene Unintentional injury 13–24*
abuse investigation
Other natural History and autopsy If minimal findings: SIDS, or intentional 12–17*
causes suffocation
UNEXPLAINED AT AUTOPSY
SIDS History, scene Intentional suffocation 80–82
investigation, absence of
explainable cause at
autopsy
Intentional Perpetrator confession, SIDS Unknown, but <5%
suffocation absence of explainable of all SUID
(filicide) cause at autopsy
Accidental History and scene Assigned to ICD-10 code (SIDS) for Varies with
suffocation or investigation, ideally US vital statistics database individual medical
strangulation in bed including doll re- Unexplained examiners and
(ASSB) enactment Undetermined coroners
Genetic mutations SCN5A , SCN1B-4B , May have negative family history Unknown, perhaps
SCN4A, long QT secondary to recessive mutations, de novo <10%
syndromes, plus Table mutation, or incomplete penetrance
402.4
* As a percentage of all sudden unexpected infant deaths explained at autopsy.
† As a percentage of all natural causes of sudden unexpected infant deaths explained at autopsy.
ICD-10, International Classification of Diseases, Version 10; SIDS, sudden infant death syndrome;
SUID, sudden unexpected infant death.
Modified from Hunt CE: Sudden infant death syndrome and other causes of infant mortality:
diagnosis, mechanisms and risk for recurrence in siblings, Am J Respir Crit Care Med
164(3):346–357, 2001.
Table 402.2
From Kliegman RM, Greenbaum LA, Lye PS: Practical strategies in pediatric
diagnosis and therapy , ed 2, Philadelphia, 2004, Elsevier Saunders, p. 98.
Table 402.3
IDIOPATHIC
Recurrent sudden infant death syndrome
CENTRAL NERVOUS SYSTEM
Congenital central hypoventilation
Neuromuscular disorders
Leigh syndrome
CARDIAC
Endocardial fibroelastosis
Wolff-Parkinson-White syndrome
Prolonged QT syndrome or other cardiac channelopathy
Congenital heart block
PULMONARY
Pulmonary hypertension
ENDOCRINE–METABOLIC
See Table 402.2
INFECTION
Disorders of immune host defense
CHILD ABUSE
Filicide or infanticide
Factitious syndrome (formerly Munchausen syndrome) by proxy
with both SIDS and other sleep-related SUID, risk reduction measures that will
be later described are applicable to all sleep-related SUID.
Pathology
Although there are no autopsy findings pathognomonic for SIDS and no findings
are required for the diagnosis, there are some that are commonly seen on
postmortem examination. Petechial hemorrhages are found in 68–95% of infants
who died of SIDS and are more extensive than in explained causes of infant
mortality. Pulmonary edema is often present and may be substantial. The reasons
for these findings are unknown. Infants who died of SIDS have higher levels of
vascular endothelial growth factor (VEGF) in the cerebrospinal fluid. These
increases may be related to VEGF polymorphisms (see “Genetic Risk Factors”
later and Table 402.4 ) or might indicate recent hypoxemic events because
VEGF is upregulated by hypoxia.
Table 402.4
CARDIAC CHANNELOPATHIES
Potassium ion channel genes (KCNE2, KCNH2, KCNQ1, KCNJ8)
Sodium ion channel gene (SCN5A) (long QT syndrome 3, Brugada syndrome)
GPD1-L-encoded connexin43 (Brugada syndrome)
SCN3B (Brugada syndrome)
CAV3 (long QT syndrome 9)
SCN4B (long QT syndrome 10)
SNTA-1 (long QT syndrome 11)
RyR2 (catecholaminergic polymorphic ventricular tachycardia)
SEROTONIN (5-HT)
5-HT transporter protein (5-HTT)
Intron 2 of SLC6A4 (variable number tandem repeat [VNTR] polymorphism)
5-HT fifth Ewing variant (FEV) gene
GENES PERTINENT TO DEVELOPMENT OF AUTONOMIC NERVOUS SYSTEM
Paired-like homeobox 2a (PHOX2A)
PHOX2B
Rearranged during transfection factor (RET)
Endothelin converting enzyme-1 (ECE1)
T-cell leukemia homeobox (TLX3)
Engrailed-1 (EN1)
Tyrosine hydroxylase (THO1)
Monamine oxidase A (MAOA)
Sodium/proton exchanger 3 (NHE3) (medullary respiratory control)
INFECTION AND INFLAMMATION
Complement C4A
Complement C4B
Interleukin-1RN (gene encoding IL-1 receptor antagonist [ra]; proinflammatory)
Interleukin-6 (IL-6; proinflammatory)
Interleukin-8 (IL-8; proinflammatory; associated with prone sleeping position)
Interleukin-10 (IL-10)
Vascular endothelial growth factor (VEGF) (proinflammatory)
Tumor necrosis factor (TNF)- α (proinflammatory)
OTHER
Mitochondrial DNA (mtDNA) polymorphisms (energy production)
Flavin-monooxygenase 3 (FMO3) (enzyme metabolizes nicotine; risk factor with smoking mothers)
Aquaporin-4 (T allele and CT/TT genotype associated with maternal smoking and with increased brain/body
weight ratio in SIDS infants)
SCN4A (nondystrophic myotonia, laryngospasm)
Modified from Hunt CE, Hauck FR: Sudden infant death syndrome: gene-
environment interactions. In Brugada R, Brugada J, Brugada P, editors: Clinical
care in inherited cardiac syndromes , Guildford, UK, 2009, Springer-Verlag
London.
SIDS infants have several identifiable changes in the lungs and other organs.
Nearly 65% of these infants have structural evidence of preexisting, chronic,
low-grade asphyxia, and other studies have identified biochemical markers of
asphyxia. Some studies have shown carotid body abnormalities, consistent with
a role for impaired peripheral arterial chemoreceptor function in SIDS.
Numerous studies have shown brain abnormalities that could cause or contribute
to an impaired autonomic response to an exogenous stressor, including in the
hippocampus and brainstem, the latter being the major area responsible for
respiratory and autonomic regulation. The affected brainstem nuclei include the
retrotrapezoid nucleus and the dorsal motor nucleus of the vagus, primary sites
upper airway reflexes. Decreases in 5-HT1A and 5-HT2A receptor
immunoreactivity have been observed in the dorsal nucleus of the vagus, solitary
nucleus, and ventrolateral medulla. There are extensive serotoninergic brainstem
abnormalities in SIDS infants, including increased 5-HT neuronal count, a lower
density of 5-HT1A receptor-binding sites in regions of the medulla involved in
homeostatic function, and a lower ratio of 5-HT transporter (5-HTT) binding
density to 5-HT neuronal count in the medulla. Male SIDS infants have lower
receptor-binding density than do female SIDS infants. Overall, these 5-HT–
related studies suggest that the synthesis and availability of 5-HT are decreased
within 5-HT pathways, and medullary tissue levels of 5-HT and its primary
biosynthetic enzyme (tryptophan hydroxylase) are lower in SIDS infants
compared with age-matched controls.
Table 402.5
Table 403.1
Symptom-Based Approach to BRUEs: Possible and Other
Conditions That Might Be Confused With BRUE
COMMON
SUGGESTIVE
AND/OR SUGGESTIVE
DIAGNOSTIC PHYSICAL TESTING TO
CONCERNING HISTORICAL
CATEGORIES EXAMINATION CONSIDER
CAUSES TO FINDINGS
FINDINGS
CONSIDER
Gastrointestinal GER Coughing, Gastric contents in Upper GI to
Intussusception vomiting, the nose and mouth assess for
Volvulus choking, gasping Choking, gagging, anatomic
Oropharyngeal temporally or oxygen anomalies
dysphagia related to feeds desaturation Clinical swallow
or regurgitation temporally related evaluation
of gastric to feeding or Abdominal
contents regurgitation of ultrasound
Feeding gastric contents pH probe
difficulties
Recent preceding
feed
Irritability
following feeds
Milk in
mouth/nose
Bilious emesis
Pulling legs to
chest
Bloody/mucousy
stool
Lethargy
following event
Infectious Upper and lower Preceding URI Fever/hypothermia NP swab for RSV,
respiratory tract symptoms Lethargy pertussis
infection (RSV, Multiple events Ill appearance Chest radiograph
pertussis, on the day of Coryza CBC and blood
pneumonia) presentation Cough culture
Bacteremia Sick exposures Wheeze Cerebrospinal
Meningitis Foul-smelling Tachypnea fluid analysis and
Urinary tract urine culture
infection Urinalysis and
culture
Neurologic Seizures Multiple events Papilledema EEG
Breath holding Loss of Abnormal Neuroimaging
spells consciousness muscular
Congenital Change in tone movements
central Abnormal Hypertonicity or
hypoventilation muscular flaccidity
syndrome movements Abnormal reflexes
Neuromuscular Eye deviation Micro- or
disorders Preceding macrocephaly
Congenital triggers Dysmorphic
malformations features
of the brain and Signs of trauma or
brainstem poisoning (see
Malignancy “Child
Intracranial maltreatment”
hemorrhage below)
Respiratory/ENT Apnea of Prematurity Wheezing Chest radiograph
prematurity Foreign body Stridor Neck radiograph
Apnea of Aspiration Crackles Laryngoscopy
infancy Noisy breathing Rhonchi Bronchoscopy
Periodic Tachypnea Esophagoscopy
breathing Polysomnography
Airway anomaly
Aspiration
Foreign body
Obstructive
sleep apnea
Child Nonaccidental Multiple events Bruising Skeletal survey
maltreatment head trauma Unexplained (especially in a Computed
Smothering vomiting or nonmobile child) tomography/MRI
Poisoning irritability Ear trauma, of the head
Factitious Recurrent hemotympanum Dilated
syndrome BRUEs Acute abdomen funduscopic
(formerly Historical Painful extremities examination if
Munchausen discrepancies Oral head imaging
syndrome) by Family history of bleeding/trauma concerning for
proxy unexplained Frenulum tears trauma
death, SIDS, or Unexplained Toxicology
BRUEs irritability screen
Single witness of Retinal Social work
event hemorrhages evaluation
Delay in seeking Depressed mental
care status
Cardiac Dysrhythmia Feeding Abnormal heart Four-extremity
(prolonged QT difficulties rate/rhythm blood pressure
syndrome, Growth Murmur Pre- and
Wolff- difficulties Decreased femoral postductal oxygen
Parkinson- Diaphoresis pulses saturation
White Prematurity measurements
syndrome) ECG
Cardiomyopathy Echocardiogram
Congenital heart Serum
disease electrolytes,
Myocarditis calcium,
magnesium
Metabolic/genetic Hypoglycemia Severe initial Dysmorphic Serum
Inborn errors of event features electrolytes;
metabolism Multiple events Microcephaly glucose, calcium,
Electrolyte Event associated Hepatomegaly and magnesium
abnormalities with period of levels
Genetic stress or fasting Lactate
syndromes Developmental Ammonia
including those delay Pyruvate
with craniofacial Associated Urine organic and
malformations anomalies serum amino
Growth acids
difficulties Newborn screen
Severe/frequent
illnesses
Family history of
BRUE,
consanguinity,
seizure disorder,
or SIDS
Initial History
An appropriate history and physical examination are key to evaluating an infant
who has experienced an acute event (Table 403.2 ). Attention should be given to
characterizing the event and interpreting the subjective experience of the
caregiver to provide an objective description. The following questions can guide
this process:
Table 403.2
Important Historical Features of a BRUE
PREEVENT
Condition of child Awake vs. asleep
Location of child Prone vs. supine, flat or upright, in crib/car seat, with pillows, blankets
Activity Feeding, crying, sleeping
EVENT
Respiratory effort None, shallow, gasping, increased
Duration of respiratory pauses
Color Pallor, red, cyanotic
Peripheral, whole body, circumoral, lighting of room
Tone/movement Rigid, tonic-clonic, decreased, floppy
Focal vs. diffuse
Ability to suppress movements
Level of consciousness Alert, interactive, sleepy, nonresponsive
Duration Time until normal breathing, normal tone, normal behavior
Detailed history of caregiver actions during event to aid in defining time course
Associated symptoms Vomiting, sputum production, blood in mouth/nose, eye rolling
POSTEVENT
Condition Back to baseline, sleepy, postictal, crying
If altered after event, duration of time until back to baseline
INTERVENTIONS
What was performed Gentle stimulation, blowing in face, mouth-to-mouth, cardiopulmonary
resuscitation
Who performed Medical professional vs. caregiver
intervention
Response to intervention Resolution of event vs. self-resolving
Duration of intervention How long was intervention performed
MEDICAL HISTORY
History of present illness Preceding illnesses, fever, rash, irritability, sick contacts
Past medical history Prematurity, prenatal exposures, gestational age, birth trauma
Noisy breathing since birth
Any medical problems, prior medical conditions, prior hospitalizations
Developmental delay
Medications
Feeding history Gagging, coughing with feeds, poor weight gain
Family history Neurologic problems
Cardiac arrhythmias
Sudden death, childhood deaths, BRUEs
Neonatal problems
Consanguinity
Social history Home situation
Caregivers
Smoke exposure
Medications in the home
BRUE , Brief resolved unexplained event.
From Kliegman RK, Lye PS, Bordini BJ, et al: Nelson pediatric symptom-based diagnosis ,
Philadelphia, 2018, Elsevier. Table 5.4.
What was the infant doing before, during, and after the event? An event
occurring during or after feeding will likely have a different explanation than
one occurring during sleep or after crying. The sequence of events can also be
diagnostic. A breath holding spell begins with crying, followed by a period of
apnea, perioral cyanosis, change of consciousness, and return to baseline.
Did the infant change color? It is often normal for infants to have blueish
discoloration (perioral cyanosis or acrocyanosis ) around the lips or hands
because of circulatory immaturity. Turning red or purple is also common when
infants cry or become upset. The clinician's goal is to distinguish less concerning
color change from central cyanosis , which is blue discoloration of the face,
trunk, gums, or tongue that can indicate hypoxemia.
Did the infant experience central or obstructive apnea, or just choking or
gagging? It is normal for infants to exhibit respiratory pauses of up to 20 sec
while awake and asleep. These can reflect periodic breathing of the newborn
or normal REM sleep. Much more concerning are periods of no air movement
that last longer than 20 sec. Obstructive apnea results in paradoxical movement
of the diaphragm and upper airway. In infants, this is most commonly caused by
upper and lower respiratory tract infections (e.g., bronchiolitis) and may precede
the recognition of symptoms typically seen in viral respiratory infections. Infants
also commonly gag or choke briefly during or shortly after feeds or with GER or
vomiting. The resulting reflexive pause in respiration to protect the airway is
management. Common causes of secondary nosebleeds from the anterior septum
include digital trauma, foreign bodies, dry air, and inflammation, including upper
respiratory tract infections, sinusitis, and allergic rhinitis (Table 405.1 ). There is
often a family history of childhood epistaxis. Nasal steroid sprays are commonly
used in children, and their chronic use may be associated with nasal mucosal
bleeding. Young infants with significant gastroesophageal reflux into the nose
rarely present with epistaxis secondary to mucosal inflammation. Susceptibility
is increased during respiratory infections and in the winter when dry air irritates
the nasal mucosa, resulting in formation of fissures and crusting. Severe
bleeding may be encountered with congenital vascular abnormalities, such as
hereditary hemorrhagic telangiectasia (see Chapter 459.3 ), varicosities,
hemangiomas, and, in children with thrombocytopenia, deficiency of clotting
factors, particularly von Willebrand disease (see Chapter 504 ), hypertension,
renal failure, or venous congestion. Recurrent epistaxis despite cauterization is
associated with mild coagulation disorders. The family history may be positive
for abnormal bleeding (epistaxis or other sites); specific testing for von
Willebrand disease is indicated because the prothrombin time or partial
thromboplastin time may be normal despite having a bleeding disorder. Nasal
polyps or other intranasal growths may be associated with epistaxis. Recurrent,
and often severe, unilateral nosebleeds may be the initial presenting symptom in
juvenile nasal angiofibroma, which occurs in adolescent males.
Table 405.1
Possible Causes of Epistaxis
Epistaxis digitorum (nose picking)
Rhinitis (allergic or viral)
Chronic sinusitis
Foreign bodies
Intranasal neoplasm or polyps
Irritants (e.g., cigarette smoke)
Septal deviation
Septal perforation
Trauma including child abuse
Vascular malformation or telangiectasia (hereditary hemorrhage telangiectasia)
Hemophilia
von Willebrand disease
Platelet dysfunction
Thrombocytopenia
Hypertension
Leukemia
Liver disease (e.g., cirrhosis)
Medications (e.g., aspirin, anticoagulants, nonsteroidal antiinflammatory drugs, topical corticosteroids)
Cocaine abuse
From Kucik CJ, Clenney T: Management of epistaxis, Am Fam Physician 71(2):305–311, 2005.
Clinical Manifestations
Epistaxis usually occurs without warning, with blood flowing slowly but freely
from 1 nostril or occasionally from both. In children with nasal lesions, bleeding
might follow physical exercise. When bleeding occurs at night, the blood may be
swallowed and become apparent only when the child vomits or passes blood in
the stools. Posterior epistaxis can manifest as anterior nasal bleeding, or, if
bleeding is copious, the patient might vomit blood as the initial symptom.
Treatment
Most nosebleeds stop spontaneously in a few minutes. The nares should be
compressed and the child kept as quiet as possible, in an upright position with
the head tilted forward to avoid blood trickling back into the throat. Cold
compresses applied to the nose can also help. If these measures do not stop the
bleeding, local application of a solution of oxymetazoline (Afrin) or
phenylephrine (Neo-Synephrine) (0.25–1%) may be useful. If bleeding persists,
an anterior nasal pack may need to be inserted; if bleeding originates in the
posterior nasal cavity, combined anterior and posterior packing is necessary.
After bleeding is under control, and if a bleeding site is identified, its obliteration
by cautery with silver nitrate may prevent further difficulties. Because the septal
cartilage derives its nutrition from the overlying mucoperichondrium, only one
side of the septum should be cauterized at a time to reduce the chance of a septal
perforation. During the winter, or in a dry environment, a room humidifier,
saline drops, and petrolatum (Vaseline) applied to the septum can help to prevent
epistaxis. Ointments prevent infection, increase moisture, decrease bleeding, and
are commonly used in clinical practice. Antiseptic cream (e.g., mupirocin) has
been used for epistaxis because it has been found that many patients with
idiopathic epistaxis have nasal bacterial colonization with subsequent
inflammation, new vessel formation, and irritation, likely leading to epistaxis.
However, studies showing the efficacy of antiseptics in epistaxis are equivocal.
Patients with severe epistaxis despite conservative medical measures should be
CHAPTER 407
The common cold is an acute viral infection of the upper respiratory tract in
which the symptoms of rhinorrhea and nasal obstruction are prominent.
Systemic symptoms and signs such as headache, myalgia, and fever are absent or
mild. The common cold is frequently referred to as infectious rhinitis but may
also include self-limited involvement of the sinus mucosa and is more correctly
termed rhinosinusitis.
Etiology
The most common pathogens associated with the common cold are the more
than 200 types of human rhinoviruses (see Chapter 290 ), but the syndrome can
be caused by many different virus families (Table 407.1 ). Rhinoviruses (HRV)
are associated with more than 50% of colds in adults and children. In young
children, other viral etiologies of the common cold include respiratory syncytial
virus (RSV; see Chapter 287 ), human metapneumovirus (MPV; see Chapter 288
), parainfluenza viruses (PIVs; see Chapter 286 ), and adenoviruses (see Chapter
289 ). Common cold symptoms may also be caused by influenza viruses,
nonpolio enteroviruses, and human coronaviruses. Many viruses that cause
rhinitis are also associated with other symptoms and signs such as cough,
wheezing, and fever.
Table 407.1
Pathogens Associated With the Common Cold
Epidemiology
Colds occur year-round, but the incidence is greatest from the early fall until the
late spring, reflecting the seasonal prevalence of the viral pathogens associated
with cold symptoms. In the northern hemisphere, the highest incidence of HRV
infection occurs in the early fall (August–October) and in the late spring (April–
May). The seasonal incidence for PIV usually peaks in the late fall and late
spring and is highest between December and April for RSV, influenza viruses,
MPV, and coronaviruses. Adenoviruses are detected at a low prevalence
throughout the cold season, and enteroviruses may also be detected during
summer months or throughout the year.
Young children have an average of 6-8 colds per yr, but 10–15% of children
have at least 12 infections per yr. The incidence of illness decreases with
increasing age, with 2-3 illnesses per yr by adulthood. The incidence of infection
is primarily a function of exposure to the virus. Children in out-of-home daycare
centers during the 1st yr of life have 50% more colds than children cared for
only at home. The difference in the incidence of illness between these groups of
children decreases as the length of time spent in daycare increases, although the
incidence of illness remains higher in the daycare group through at least the 1st 3
yr of life. When they begin primary school, children who attended daycare have
less frequent colds than those who did not. Mannose-binding lectin deficiency
course of a cold. Anterior cervical lymphadenopathy or conjunctival injection
may also be noted on exam.
Diagnosis
The most important task of the physician caring for a patient with a cold is to
exclude other conditions that are potentially more serious or treatable. The
differential diagnosis of the common cold includes noninfectious disorders and
other upper respiratory tract infections (Table 407.2 ).
Table 407.2
Conditions That Can Mimic the Common Cold
Laboratory Findings
Routine laboratory studies are not helpful for the diagnosis and management of
the common cold . A nasal smear for eosinophils (Hansel stain) may be useful if
allergic rhinitis is suspected (see Chapter 168 ). A predominance of
polymorphonuclear cells in the nasal secretions is characteristic of
uncomplicated colds and does not indicate bacterial superinfection. Self-limited
radiographic abnormalities of the paranasal sinuses are common during an
uncomplicated cold; imaging is not indicated for most children with simple
rhinitis.
The viral pathogens associated with the common cold can be detected by
sinuses are normally cleared readily, but during viral rhinosinusitis,
inflammation and edema can block sinus drainage and impair mucociliary
clearance of bacteria. The growth conditions are favorable, and high titers of
bacteria are produced.
Clinical Manifestations
Children and adolescents with sinusitis can present with nonspecific complaints,
including nasal congestion, purulent nasal discharge (unilateral or bilateral),
fever, and cough. Less-common symptoms include bad breath (halitosis), a
decreased sense of smell (hyposmia), and periorbital edema (Table 408.1 ).
Complaints of headache and facial pain are rare in children. Additional
symptoms include maxillary tooth discomfort and pain or pressure exacerbated
by bending forward. Physical examination might reveal erythema and swelling
of the nasal mucosa with purulent nasal discharge. Sinus tenderness may be
detectable in adolescents and adults. Transillumination reveals an opaque sinus
that transmits light poorly.
Table 408.1
Conventional Criteria for the Diagnosis of Sinusitis Based
on the Presence of at Least 2 Major or 1 Major and ≥2 Minor
Symptoms
Table 408.2
Antimicrobial Regimens for Acute Bacterial Rhinosinusitis
in Children
FIRST-LINE (DAILY
INDICATION SECOND-LINE (DAILY DOSE)
DOSE)
Initial empirical therapy Amoxicillin- • Amoxicillin-clavulanate (90 mg/kg/day PO bid)
clavulanate (45
mg/kg/day PO bid)
β-Lactam allergy
Type I hypersensitivity • Levofloxacin (10-20 mg/kg/day PO every 12-24 h)
Non-type I • Clindamycin* (30-40 mg/kg/day PO tid) plus cefixime (8
hypersensitivity mg/kg/day PO bid) or cefpodoxime (10 mg/kg/day PO bid)
Risk for antibiotic • Amoxicillin-clavulanate (90 mg/kg/day PO bid)
resistance or failed initial • Clindamycin* (30-40 mg/kg/day PO tid) plus cefixime (8
therapy mg/kg/day PO bid) or cefpodoxime (10 mg/kg/day PO bid)
• Levofloxacin (10-20 mg/kg/day PO every 12-24 h)
Severe infection requiring • Ampicillin/sulbactam (200-400 mg/kg/day IV every 6 h)
hospitalization • Ceftriaxone (50 mg/kg/day IV every 12 h)
• Cefotaxime (100-200 mg/kg/day IV every 6 h)
• Levofloxacin (10-20 mg/kg/day IV every 12-24 h)
*
Resistance to clindamycin (~31%) is found frequently among Streptococcus pneumoniae
serotype 19A isolates in different regions of the United States.
bid, 2 times daily; IV, intravenously; PO, orally; qd, daily; tid, 3 times a day.
From Chow AW, Benninger MS, Brook I, et al: IDSA clinical practice guideline for acute bacterial
rhinosinusitis in children and adults. CID 54:e72–e112, 2012, Table 9, p. e94.
Table 408.3
Indications for Referral to a Specialist
• Severe infection (high persistent fever with temperature >39°C [>102°F]; orbital edema; severe headache, visual
disturbance, altered mental status, meningeal signs)
• Recalcitrant infection with failure to respond to extended courses of antimicrobial therapy
• Immunocompromised host
• Multiple medical problems that might compromise response to treatment (e.g., hepatic or renal impairment,
hypersensitivity to antimicrobial agents, organ transplant)
• Unusual or resistant pathogens
• Fungal sinusitis or granulomatous disease
• Nosocomial infection
• Anatomic defects causing obstruction and requiring surgical intervention
• Multiple recurrent episodes of acute bacterial rhinosinusitis (3-4 episodes per year) suggesting chronic sinusitis
• Chronic rhinosinusitis (with or without polyps or asthma) with recurrent ABRS exacerbations
• Evaluation of immunotherapy for allergic rhinitis
From Chow AW, Benninger MS, Brook I, et al: IDSA clinical practice guideline for acute bacterial
rhinosinusitis in children and adults. CID 54:e72–e112, 2012, Table 14, p. e106.
Table 409.1
Infectious Agents That Cause Pharyngitis
VIRUSES BACTERIA
Adenovirus Streptococcus pyogenes (Group A streptococcus)
Coronavirus Arcanobacterium haemolyticum
Cytomegalovirus Fusobacterium necrophorum
Epstein-Barr virus Corynebacterium diphtheriae
Enteroviruses Neisseria gonorrhoeae
Herpes simplex virus (1 and 2) Group C streptococci
Human immunodeficiency virus Group G streptococci
Human metapneumovirus Francisella tularensis
Influenza viruses (A and B)
Yersinia pestis
Measles virus
Chlamydophila pneumoniae
Parainfluenza viruses
Chlamydia trachomatis
Respiratory syncytial virus
Rhinoviruses Mycoplasma pneumoniae
Mixed anaerobes (Vincent angina)
Table 409.2
Epidemiologic and Clinical Features Suggestive of Group A
Streptococcal and Viral Pharyngitis
FEATURE, BY SUSPECTED ETIOLOGIC AGENT
Group A Streptococcal
• Sudden onset of sore throat
• Age 5-15 yr
• Fever
• Headache
• Nausea, vomiting, abdominal pain
• Tonsillopharyngeal inflammation
• Patchy tonsillopharyngeal exudates
• Palatal petechiae
• Anterior cervical adenitis (tender nodes)
• Winter and early spring presentation
• History of exposure to strep pharyngitis
• Scarlatiniform rash
Viral
• Conjunctivitis
• Coryza
• Cough
• Diarrhea
• Hoarseness
• Discrete ulcerative stomatitis
• Viral exanthema
From Shulman ST, Bisno AL, Clegg HW, et al: Clinical practice guideline for the diagnosis and
management of group A streptococcal pharyngitis: 2012 update by the Infectious Diseases
Society of America. Clin Infect Dis 55(10):e86–e102, 2012, Table 4, p. e91.
Table 409.3
Positive Predictive Value of McIsaac Score in Children in
Clinical Studies*
Throat culture and rapid antigen-detection tests (RADTs) are the diagnostic
tests for GAS most available in routine clinical care. Throat culture plated on
blood agar remains the gold standard for diagnosing streptococcal pharyngitis.
There are both false-negative cultures as a consequence of sampling errors or
prior antibiotic treatment and false-positive cultures as a consequence of
misidentification of other bacteria as GAS. Streptococcal RADTs detect the
group A carbohydrate of GAS. They are used by the vast majority of office-
based pediatricians. All RADTs have very high specificity, generally ≥95%, so
when a RADT is positive it is assumed to be accurate and throat culture is
unnecessary. Because RADTs are generally much less sensitive than culture,
treatment should not be delayed for children with symptomatic pharyngitis and a
positive test for GAS. Presumptive antibiotic treatment can be started when there
is a clinical diagnosis of scarlet fever, a symptomatic child has a household
contact with documented streptococcal pharyngitis, or there is a history of ARF
in the patient or a family member, but a diagnostic test should be performed to
confirm the presence of GAS and antibiotics should be discontinued if GAS are
not identified.
A variety of antimicrobial agents are effective for GAS pharyngitis (Table
409.4 ). Group A streptococci are universally susceptible to penicillin and all
other β-lactam antibiotics. Penicillin is inexpensive, has a narrow spectrum of
activity, and has few adverse effects. Amoxicillin is often preferred for children
because of taste, availability as chewable tablets and liquid, and the convenience
of once-daily dosing. The duration of oral penicillin and amoxicillin therapy is
10 days. A single intramuscular dose of benzathine penicillin or a benzathine-
procaine penicillin G combination is effective and ensures compliance. Follow-
up testing for GAS is unnecessary after completion of therapy and is not
recommended unless symptoms recur.
Table 409.4
Recommended Treatment for Acute Streptococcal
Pharyngitis
MOST PATIENTS
WEIGHT <27 kg WEIGHT ≥27 kg ROUTE DURATION
Amoxicillin 50 mg/kg once daily (maximum 1,000 mg) Oral 10 days
Penicillin V 250 mg bid 500 mg bid Oral 10 days
Benzathine penicillin G 600,000 units 1.2 million units IM Once
Benzathine penicillin G + procaine 900,000 units + 300,000 900,000 units + IM Once
penicillin G units 300,000 units
PENICILLIN-ALLERGIC PATIENTS
ORAL DOSE FREQUENCY DURATION
Cephalosporins* Varies with agent chosen 10 days
Erythromycins 40 mg/kg/day up to 1,000 bid 10 days
Ethylsuccinate mg/day
Estolate 20-40 mg/kg/day up to bid 10 days
1,000 mg/day
Clarithromycin 15 mg/kg/day up to 500 bid 10 days
mg/day
Azithromycin † 12 mg/kg day 1; 6 mg/kg qd 5 days
days 2-5
Clindamycin 20 mg/kg/day up to 1.8 tid 10 days
g/day
Tonsillectomy alone is most commonly performed for recurrent or chronic
pharyngotonsillitis. Tonsillectomy has been shown to be effective in reducing the
number of infections and the symptoms of chronic tonsillitis such as halitosis,
persistent or recurrent sore throats, and recurrent cervical adenitis in severely
affected patients. In resistant cases of cryptic tonsillitis, tonsillectomy may be
curative. Rarely in children, tonsillectomy is indicated for biopsy of a
unilaterally enlarged tonsil to exclude a neoplasm or to treat recurrent
hemorrhage from superficial tonsillar blood vessels. Tonsillectomy has not been
shown to offer clinical benefit over conservative treatment in children with mild
symptoms or in those with severe infections 2 yr after surgery.
There are large variations in surgical rates among children across countries:
144 in 10,000 in Italy; 115 in 10,000 in the Netherlands; 65 in 10,000 in
England; and 50 in 10,000 in the United States. Rates are generally higher in
boys. With the issuance of practice guidelines, these variations may decrease.
The American Academy of Otolaryngology (AAO)–Head and Neck Surgery
Taskforce on Clinical Practice Guidelines: Tonsillectomy in Children issued
evidence-based guidelines in 2019 (Table 411.1 ). Table 411.2 illustrates the
differences and similarities between these guidelines with those of the other
major professional groups across the globe. The 2019 guidelines recommend
watchful waiting for recurrent throat infections if there has been <7 episodes in
the past year, <5 episodes/yr in the past 2 yr, or <3 episodes/yr in the past 3 yr.
Table 411.1
Paradise Criteria for Tonsillectomy
CRITERION DEFINITION
Minimum At least 7 episodes in the previous year, at least 5 episodes in each of the previous 2 yr, or at
frequency of sore least 3 episodes in each of the previous 3 yr
throat episodes
Clinical features • Sore throat plus at least 1 of the following features qualifies as a counting episode:
• Temperature of greater than 38.3°C (100.9°F)
• Cervical adenopathy (tender lymph nodes or lymph node size >2 cm)
• Tonsillar exudate
• Culture positive for group A β-hemolytic streptococcus
Treatment Antibiotics administered in the conventional dosage for proved or suspected streptococcal
episodes
Documentation Each episode of throat infection and its qualifying features substantiated by
contemporaneous notation in a medical record
If the episodes are not fully documented, subsequent observance by the physician of 2
episodes of throat infection with patterns of frequency and clinical features consistent with
the initial history*
* Allows for tonsillectomy in patients who meet all but the documentation criterion. A 12 mo
Table 411.2
Comparison of American, Italian, and Scottish Guidelines
for Tonsillectomy in Children and Adolescents
AAO-HNS
PARAMETER ITALIAN GUIDELINES SCOTTISH GUIDELINES
GUIDELINES
Audience Multidisciplinary Multidisciplinary Multidisciplinary
Target population Children and Children and adults Children 4-16 yr of age and
adolescents 1-18 yr of adults
age
Scope Treatment of children Appropriateness and safety of Management of sore throat and
who are candidates for tonsillectomy indications for tonsillectomy
tonsillectomy
Methods Based on a priori Systematic literature review, Italian Based on a priori protocol,
protocol, systematic National Program Guidelines scale systematic literature review,
literature review, of evidence quality Scottish Intercollegiate
American Academy of Guidelines Network scale of
Pediatrics scale of evidence quality
evidence quality
Recommendations
Recurrent Tonsillectomy is an Tonsillectomy is indicated in Tonsillectomy should be
infection option for children with patients with at least 1 yr of considered for recurrent,
recurrent throat recurrent tonsillitis (5 or more disabling sore throat caused by
infection that meets the episodes per year) that is disabling acute tonsillitis when the
Paradise criteria (see and impairs normal activities, but episodes are well documented,
Table 411.1 ) for only after an additional 6 mo of are adequately treated, and
frequency, severity, watchful waiting to assess the meet the Paradise criteria (see
treatment, and pattern of symptoms using a Table 411.1 ) for frequency of
documentation of clinical diary illness
illness
Pain control Recommendation to Recommendation for Recommendation for adequate
advocate for pain relief acetaminophen before and after dose of acetaminophen for pain
(e.g., provide surgery relief in children
information, prescribe)
and educate caregivers
about the importance of
managing and
reassessing pain
Antibiotic use Recommendation Recommendation for short-term NA
against perioperative perioperative antibiotics*
antibiotics
Steroid use Recommendation for a Recommendation for a single Recommendation for a single
single intraoperative intraoperative dose of intraoperative dose of
dose of dexamethasone dexamethasone dexamethasone
Sleep-disorderedRecommendation to Recommendation for diagnostic NA
breathing counsel caregivers testing in children with suspected
about tonsillectomy as a sleep respiratory disorders
means to improve
health in children with
sleep-disordered
breathing and comorbid
conditions
Polysomnography Recommendation to Recommendation for NA
counsel caregivers polysomnography when pulse
about tonsillectomy as a oximetry results are not conclusive
means to improve in agreement with Brouillette
health in children with criteria
abnormal
polysomnography
Surgical NA Recommendation for “cold” NA
technique technique
Hemorrhage Recommendation that NA NA
the surgeon document
primary and secondary
hemorrhage after
tonsillectomy at least
annually
Adjunctive NA NA Recommendation against
therapy Echinacea purpurea for
treatment of sore throat
Recommendation for
acupuncture in patients at risk
of postoperative nausea and
vomiting who cannot take
antiemetic drugs
* Statement made prior to most recent Cochrane review.
AAO-HNS, American Academy of Otolaryngology–Head and Neck Surgery; NA, not applicable.
Adapted with permission from Baugh RF, Archer SM, Mitchell RB, et al: American Academy of
Otolaryngology–Head and Neck Surgery Foundation. Clinical practice guideline: tonsillectomy in
children. Otolaryngol Head Neck Surg 144(1 Suppl):S23, 2011, Table 9.
Adenoidectomy
Adenoidectomy alone may be indicated for the treatment of chronic nasal
infection (chronic adenoiditis), chronic sinus infections that have failed medical
management, and recurrent bouts of acute otitis media, including those in
children with tympanostomy tubes who suffer from recurrent otorrhea.
Adenoidectomy may be helpful in children with chronic or recurrent otitis media
with effusion. Adenoidectomy alone may be curative in the management of
patients with nasal obstruction, chronic mouth breathing, and loud snoring
suggesting sleep-disordered breathing. Adenoidectomy may also be indicated for
Bleeding can occur in the immediate postoperative period or be delayed
(consider von Willebrand disease) after separation of the eschar. The Clinical
Guidelines for Tonsillectomy include a recommendation for a single intravenous
dose of intraoperative dexamethasone (0.5 mg/kg), which decreases
postoperative nausea and vomiting and reduces swelling. There is no evidence
that use of dexamethasone in postoperative tonsillectomy patients results in an
increased risk of postoperative bleeding. Routine use of antibiotics in the
postoperative period is ineffective and thus the AAO Clinical Practice
Guidelines advise against its use, although this recommendation is not consistent
among the major professional organizations who have issued guidelines (see
Table 411.2 ). Codeine is associated with excessive sedation and fatalities and is
not recommended.
Table 411.3
Risks and Potential Benefits of Tonsillectomy or
Adenoidectomy or Both
RISKS
• Cost*
• Risk of anesthetic accidents
• Malignant hyperthermia
• Cardiac arrhythmia
• Vocal cord trauma
• Aspiration with resulting bronchopulmonary obstruction or infection
• Risk of miscellaneous surgical or postoperative complications
• Hemorrhage
• Airway obstruction from edema of tongue, palate, or nasopharynx, or retropharyngeal hematoma
• Central apnea
• Prolonged muscular paralysis
• Dehydration
• Palatopharyngeal insufficiency
• Otitis media
• Nasopharyngeal stenosis
• Refractory torticollis
• Facial edema
• Emotional upset
• Unknown risks
POTENTIAL BENEFITS
• Reduction in frequency of ear, nose, throat illness, and thus in
• Discomfort
• Inconvenience
• School absence
• Parental anxiety
• Work missed by parents
• Costs of physician visits and drugs
• Reduction in nasal obstruction with improved
• Respiratory function
• Comfort
• Sleep
• Craniofacial growth and development
• Appearance
• Reduction in hearing impairment
• Improved growth and overall well-being
• Reduction in long-term parental anxiety
* Cost for tonsillectomy alone and adenoidectomy alone are somewhat lower.
Swelling of the tongue and soft palate can lead to acute airway obstruction in
the first few hours after surgery. Children with underlying hypotonia or
craniofacial anomalies are at greater risk for suffering this complication.
Dehydration from odynophagia is not uncommon in the first postoperative week.
Rare complications include velopharyngeal insufficiency, nasopharyngeal or
oropharyngeal stenosis, and psychologic problems.
Bibliography
Amoils M, Chang KW, Saynina O, et al. Postoperative
complications in pediatric tonsillectomy and adenoidectomy
in ambulatory vs inpatient settings. JAMA Otolaryngol Head
Neck Surg . 2016;142(4):344–350.
Baugh RF, Archer SM, Mitchell RB, et al. Clinical practice
guideline: tonsillectomy in children. Otolaryngol Head Neck
Surg . 2011;144(1 Suppl):S1–S30.
Bonuck KA, Freeman K, Henderson J. Growth and growth
biomarker changes after adenotonsillectomy: systematic
review and meta-analysis. Arch Dis Child . 2009;94:83–91.
Borgstrom A, Nerfeldt P, Friberg D. Adenotonsillotomy versus
adenotonsillectomy in pediatric obstructive sleep apnea: an
RCT. Pediatrics . 2017;139(4):e20163314.
Brigger MT, Cunningham MJ, Hartnick CJ. Dexamethasone
administration and postoperative bleeding risk in children
undergoing tonsillectomy. Arch Otolaryngol Head Neck Surg
. 2010;136(8):766–772; 10.1001/archoto.2010.133 [PMID]
CHAPTER 416
Table 416.1
Classification of Tracheomalacia
PRIMARY TRACHEOMALACIA
Congenital absence of tracheal-supporting cartilages
SECONDARY TRACHEOMALACIA
Esophageal atresia, tracheoesophageal fistula
Vascular rings (double aortic arch)
Tracheal compression from an aberrant innominate artery
Tracheal compression from mediastinal masses
Abnormally soft tracheal cartilages associated with connective tissue disorders
Prolonged mechanical ventilation, chronic lung disease
resistance. Marginal additional narrowing, such as that caused by inflammation
related to viral infection, is then more likely to result in wheezing.
Infant chest wall compliance is also quite high, thus the inward pressure
produced in normal expiration subjects the intrathoracic airways to collapse.
Differences in tracheal cartilage and airway smooth muscle tone increase the
collapsibility of the infant airways in comparison with older children. These
mechanisms combine to make the infant more susceptible to airway obstruction,
increased resistance, and subsequent wheezing. The mechanical portion of the
infant propensity to wheeze resolves with normal growth and muscular
development.
Although wheezing in infants most frequently results from inflammation due
to acute viral infections, there are many potential causes of wheezing (Table
418.1 ).
Table 418.1
Differential Diagnosis of Wheezing in Infancy
INFECTION
Viral
Respiratory syncytial virus
Human metapneumovirus
Parainfluenza
Adenovirus
Influenza
Rhinovirus
Bocavirus
Coronavirus
Enterovirus
Other
Chlamydia trachomatis
Tuberculosis
Histoplasmosis
Papillomatosis
ASTHMA
ANATOMIC ABNORMALITIES
Central Airway Abnormalities
Malacia of the larynx, trachea, and/or bronchi
Laryngeal or tracheal web
Tracheoesophageal fistula (specifically H-type fistula)
Laryngeal cleft (resulting in aspiration)
Extrinsic Airway Anomalies Resulting in Airway Compression
Vascular ring or sling
Mediastinal lymphadenopathy from infection or tumor
Mediastinal mass or tumor
Esophageal foreign body
Intrinsic Airway Anomalies
Airway hemangioma, other tumor
Congenital pulmonary airway malformation (cystic adenomatoid malformation)
Bronchial or lung cyst
Congenital lobar emphysema
Aberrant tracheal bronchus
Sequestration
Congenital heart disease with left-to-right shunt (increased pulmonary edema)
Foreign body
Immunodeficiency States
Immunoglobulin A deficiency
B-cell deficiencies
AIDS
Bronchiectasis
MUCOCILIARY CLEARANCE DISORDERS
Cystic fibrosis
Primary ciliary dyskinesia
Bronchiectasis
ASPIRATION SYNDROMES
Gastroesophageal reflux disease
Pharyngeal/swallow dysfunction
OTHER
Bronchopulmonary dysplasia
Eosinophilic granulomatosis with polyangiitis
Interstitial lung disease, including bronchiolitis obliterans
Heart failure
Anaphylaxis
Inhalation injury—burns
Acute Bronchiolitis
Acute bronchiolitis is a diagnostic term used to describe the clinical picture
produced by several different viral lower respiratory tract infections in infants
and very young children. The respiratory findings observed in bronchiolitis
include tachypnea, wheezing, crackles, and rhonchi which result from
inflammation of the small airways (Fig. 418.1 ). Despite its commonality, a
universal set of diagnostic criteria for bronchiolitis does not exist, with
significant disagreement about the upper age limit for appropriate use of the
diagnosis. Some clinicians restrict the term to children younger than 1 yr, and
others extend it to the age of 2 yr or beyond.
lower lung function at birth which improves with growth resulting in resolution
of wheezing by age 3; persistent wheezing, comprising about 14% of the cohort,
characterized by declining lung function and wheezing before and after age 3;
and late-onset wheezing, comprising 15% of the cohort, characterized by
relatively stable lung function and wheezing that does not begin until after age 3.
The remaining 50% of the population did not suffer a wheezing illness.
Following the cohort into adulthood revealed continued declines in the rates of
persistent symptoms. Similar patterns are also seen in birth cohort studies in
other countries.
Multiple studies attempting to predict which infants suffering from early
wheezing illnesses will go on to have asthma in later life have failed to achieve
discriminant validity. Interestingly, in both U.S. and U.K. prospective cohorts,
wheezing with an onset after the first 18-36 mo of life is one of the strongest
predictors of eventual asthma in both cohorts. Other proposed risk factors for
persistent wheezing include parental history of asthma and allergies, maternal
smoking, persistent rhinitis (apart from acute upper respiratory tract infections),
allergen sensitization, eczema, and peripheral eosinophilia, although no single
factor is strongly discriminative. Despite several randomized trials, there is no
evidence that early administration of inhaled corticosteroids to high-risk
populations can prevent the development of asthma.
Clinical Manifestations
History and Physical Examination
The initial history of a wheezing infant should describe the recent event
including onset, duration, and associated factors (Table 418.2 ). Birth history
includes weeks of gestation, neonatal complications including history of
intubation or oxygen requirement, maternal complications, and prenatal smoke
exposure. Past medical history includes any comorbid conditions. Family history
of cystic fibrosis, immunodeficiencies, asthma in a first-degree relative, or any
other recurrent respiratory conditions in children should be obtained. Social
history should include any second-hand tobacco or other smoke exposure,
daycare exposure, number of siblings, pets, and concerns regarding home
environment (e.g., dust mites, construction dust, heating and cooling techniques,
mold, cockroaches). The patient's growth chart should be reviewed for signs of
failure to thrive.
Table 418.2
Pertinent Medical History in the Wheezing Infant
Did the onset of symptoms begin at birth or thereafter?
Is the infant a noisy breather, and when is it most prominent?
Is the noisy breathing present on inspiration, expiration, or both?
Is there a history of cough apart from wheezing?
Was there an earlier lower respiratory tract infection?
Is there a history of recurrent upper or lower respiratory tract infections?
Have there been any emergency department visits, hospitalizations, or intensive care unit admissions for
respiratory distress?
Is there a history of eczema?
Does the infant cough after crying or cough at night?
How is the infant growing and developing?
Is there associated failure to thrive?
Is there a history of electrolyte abnormalities?
Are there signs of intestinal malabsorption including frequent, greasy, or oily stools?
Is there a maternal history of genital herpes simplex virus infection?
What was the gestational age at delivery?
Was the patient intubated as a neonate?
Does the infant bottle-feed in the bed or the crib, especially in a propped position?
Are there any feeding difficulties including choking, gagging, arching, or vomiting with feeds?
Is there any new food exposure?
Is there a toddler in the home or lapse in supervision in which foreign body aspiration could have occurred?
Change in caregivers or chance of nonaccidental trauma?
Differential Diagnosis
Persistent or recurrent symptoms should lead the clinician to consider entities
other than acute bronchitis. Many entities manifest with cough as a prominent
symptom (Table 418.3 ).
Table 418.3
Treatment
There is no specific therapy for acute bronchitis. The disease is self-limited, and
antibiotics, although often prescribed, do not hasten improvement. Frequent
shifts in position can facilitate pulmonary drainage in infants. Older children are
sometimes more comfortable with humidity, but this does not shorten the disease
course. Cough suppressants can relieve symptoms but can also increase the risk
of suppuration and inspissated secretions and therefore should be used
judiciously. Antihistamines dry secretions and are not helpful; expectorants are
likewise not indicated. Nonprescription cough and cold medicines should not be
used in children younger than 4 yr of age, and their use is cautioned in children
age 4-11 yr.
Chronic Bronchitis
Chronic bronchitis is well recognized in adults, formally defined as 3 mo or
longer of productive cough each year for 2 or more yr. The disease can develop
insidiously, with episodes of acute obstruction alternating with quiescent periods.
Some predisposing conditions can lead to progression of airflow obstruction or
chronic obstructive pulmonary disease, with smoking as the major factor (up to
80% of patients have a smoking history). Other conditions include air pollution,
CHAPTER 419
Plastic Bronchitis
Brett J. Bordini
Table 419.1
Conditions Associated With Plastic Bronchitis
PROVEN CONDITIONS
Congenital heart disease with Fontan physiology
Pulmonary lymphatic anomalies
Influenza A pulmonary infection
POSSIBLE CONDITIONS
Toxic inhalation
Sickle cell acute chest syndrome
Hypersecretory and near-fatal asthma (eosinophilic casts)
UNLIKELY AND UNPROVEN CONDITIONS
Cystic fibrosis
Chronic obstructive pulmonary disease
Bronchiectasis
Bacterial pneumonia
From Rubin BK: Plastic bronchitis, Clin Chest Med 37:405–408, 2016 (Box 1, p. 406).
FIG. 419.1 Tracheobronchial casts following bronchoscopic extraction.
Casts show branched architecture corresponding to the bronchial tree.
(From Corrin B, Nicholson AG: Pathology of the lungs , ed 3, London, 2011,
Churchill Livingstone. Fig 3.20.)
Epidemiology
Plastic bronchitis is rare, and its true prevalence in the pediatric population is not
known but is estimated to be 6.8 cases per 100,000 patients. Prevalence does
vary in relation to the underlying associated disease state, with rates as high as
4–14% estimated in patients who have undergone staged palliation of complex
congenital heart disease and much lower rates seen complicating asthma and
atopic disease. A slight male predominance exists for cast formation in the
setting of structural heart disease, whereas cast formation in the setting of
asthma and atopic disease demonstrates a female predominance. Children with
single-ventricle Fontan physiology are at high risk for developing plastic
bronchitis.
systemic lupus erythematosus [see Chapter 183 ], scleroderma [see Chapter 185
], Stevens-Johnson syndrome [see Chapter 177 ]), and inhalation of toxic fumes
or particulate exposure (NO2 , incinerator fly ash, NH3 , diacetyl flavorings from
microwave popcorn, papaverine, fiberglass) (Table 422.1 ). Postinfection
obliterans may be more common in the southern hemisphere and among persons
of Asian descent. BO is also commonly seen in post lung or bone marrow
transplant recipients.
Table 422.1
Etiology of Bronchiolitis Obliterans
POSTINFECTION
Adenovirus types 3, 7, and 21
Influenza
Parainfluenza
Measles
Respiratory syncytial virus
Varicella
Mycoplasma pneumoniae
POSTTRANSPLANTATION
Chronic rejection of lung or heart/lung transplantation
Graft versus host disease associated with bone marrow transplantation
CONNECTIVE TISSUE DISEASE
Juvenile idiopathic arthritis
Sjögren syndrome
Systemic lupus erythematosus
TOXIC FUME INHALATION
NO2
NH3
Diacetyl flavorings (microwave popcorn)
CHRONIC HYPERSENSITIVITY PNEUMONITIS
Avian antigens
Mold
ASPIRATION
Stomach contents: gastroesophageal reflux
Foreign bodies
DRUGS
Penicillamine
Cocaine
STEVENS-JOHNSON SYNDROME
Idiopathic
Drug induced
Infection related
From Moonnumakal SP, Fan LL: Bronchiolitis obliterans in children, Curr Opin Pediatr 20:272–
278, 2008.
Table 422.2
High-Resolution CT Patterns in Child With Interstitial Lung
Disease
GROUND-
STUDIES THICK MOSAIC
GLASS NODULES HONEYCOMBING
(N) SEPTA PATTERN
OPACITY
Bronchiolitis obliterans 4 — — — X —
Nonspecific interstitial 6 X — — — X
pneumonitis
Desquamative interstitial 4 X — — — X
pneumonitis
Follicular bronchitis or 4 X — — X —
neuroendocrine cell
hyperplasia of infancy
Lymphocytic interstitial 4 — — X — —
pneumonitis
Lymphangiomatosis 2 — X — — —
Lymphangiectasia 2 — X — — —
Pulmonary alveolar 2 X X — — —
proteinosis
From Long FR: Interstitial lung disease. In Slovis TL, editor: Caffey's pediatric diagnostic imaging ,
ed 11, Philadelphia, 2008, Mosby, Table 74.1; original data from Lynch DA, Hay T, Newell JD Jr, et
al: Pediatric diffuse lung disease: diagnosis and classification using high-resolution CT, AJR Am J
CHAPTER 424
Pulmonary Edema
Brandon T. Woods, Robert L. Mazor
Pathophysiology
Although pulmonary edema is traditionally separated into two categories
according to cause (cardiogenic and noncardiogenic ), the end result of both
processes is a net fluid accumulation within the interstitial and alveolar spaces.
Noncardiogenic pulmonary edema, in its most severe state, is also known as
acute respiratory distress syndrome (see Chapters 89 and 400 ).
The hydrostatic pressure and colloid osmotic (oncotic) pressure on either side
of a pulmonary vascular wall, along with vascular permeability, are the forces
and physical factors that determine fluid movement through the vessel wall.
Baseline conditions lead to a net filtration of fluid from the intravascular space
into the interstitium. This extra interstitial fluid is usually rapidly reabsorbed by
pulmonary lymphatics. Conditions that lead to altered vascular permeability,
increased pulmonary vascular pressure, and decreased intravascular oncotic
pressure increase the net flow of fluid out of the vessel (Table 424.1 ). Once the
capacity of the lymphatics for fluid removal is exceeded, water accumulates in
the lung.
Table 424.1
Etiology of Pulmonary Edema
INCREASED PULMONARY CAPILLARY PRESSURE
Cardiogenic, such as left ventricular failure
Noncardiogenic, as in pulmonary venoocclusive disease, pulmonary venous fibrosis, mediastinal tumors
INCREASED CAPILLARY PERMEABILITY
Bacterial and viral pneumonia
Acute respiratory distress syndrome
Inhaled toxic agents
Circulating toxins
Vasoactive substances such as histamine, leukotrienes, thromboxanes
Diffuse capillary leak syndrome, as in sepsis
Immunologic reactions, such as transfusion reactions
Smoke inhalation
Aspiration pneumonia/pneumonitis
Drowning and near drowning
Radiation pneumonia
Uremia
LYMPHATIC INSUFFICIENCY
Congenital and acquired
DECREASED ONCOTIC PRESSURE
Hypoalbuminemia, as in renal and hepatic diseases, protein-losing states, and malnutrition
INCREASED NEGATIVE INTERSTITIAL PRESSURE
Upper airway obstructive lesions, such as croup and epiglottitis
Reexpansion pulmonary edema
MIXED OR UNKNOWN CAUSES
Neurogenic pulmonary edema
High-altitude pulmonary edema
Eclampsia
Pancreatitis
Pulmonary embolism
Heroin (narcotic) pulmonary edema
Modified from Robin E, Carroll C, Zelis R: Pulmonary edema, N Engl J Med 288:239, 292, 1973;
and Desphande J, Wetzel R, Rogers M: In Rogers M, editor: Textbook of pediatric intensive care ,
ed 3, Baltimore, 1996, Williams & Wilkins, pp. 432–442.
Clinical Manifestations
The clinical features depend on the mechanism of edema formation. In general,
interstitial edema and alveolar edema prevent the inflation of alveoli, leading to
atelectasis and decreased surfactant production. This results in diminished
pulmonary compliance and tidal volume. The patient must increase respiratory
effort and/or the respiratory rate so as to maintain minute ventilation. The
earliest clinical signs of pulmonary edema include increased work of breathing,
tachypnea, and dyspnea. As fluid accumulates in the alveolar space, auscultation
reveals fine crackles and wheezing, especially in dependent lung fields. In
cardiogenic pulmonary edema, a gallop may be present, as well as peripheral
edema and jugular venous distention.
Chest radiographs can provide useful ancillary data, although findings of
initial radiographs may be normal. Early radiographic signs that represent
accumulation of interstitial edema include peribronchial and perivascular
cuffing. Diffuse streakiness reflects interlobular edema and distended pulmonary
lymphatics. Diffuse, patchy densities, the so-called butterfly pattern, represent
bilateral interstitial or alveolar infiltrates and are a late sign. Cardiomegaly is
often seen with cardiogenic causes of pulmonary edema. Heart size is usually
normal in noncardiogenic pulmonary edema (Table 424.2 ). Chest tomography
demonstrates edema accumulation in the dependent areas of the lung. As a
result, changing the patient's position can alter regional differences in lung
compliance, functional residual capacity, and alveolar ventilation.
Table 424.2
the point at which the left subclavian artery exits the aortic arch and measuring across to the point
at which the superior vena cava crosses the right mainstem bronchus. A vascular pedicle width
>70 mm on a portable digital anteroposterior radiograph of the chest obtained when the patient is
supine is optimal for differentiating high from normal-to-low intravascular volume.
From Ware LB, Matthay MA: Acute pulmonary edema, N Engl J Med 353:2788–2796, 2005.
Treatment
The treatment of a patient with noncardiogenic pulmonary edema is largely
supportive, with the primary goal to ensure adequate ventilation and
oxygenation. Additional therapy is directed toward the underlying cause.
Patients should receive supplemental oxygen to increase alveolar oxygen tension
and pulmonary vasodilation. Patients with pulmonary edema of cardiogenic
causes should be managed with diuretics, inotropic agents, and systemic
vasodilators to reduce left ventricular afterload (see Chapter 442 ). Diuretics are
also valuable in the treatment of pulmonary edema associated with total body
fluid overload (sepsis, renal insufficiency). Morphine is often helpful as a
vasodilator and a mild sedative.
Positive airway pressure improves gas exchange in patients with pulmonary
edema. In tracheally intubated patients, positive end-expiratory pressure can be
used to optimize pulmonary mechanics. Noninvasive forms of ventilation, such
as mask or nasal prong continuous positive airway pressure, are also effective.
The mechanism by which positive airway pressure improves pulmonary edema
CHAPTER 426
Etiology
Repeated aspiration of even small quantities of gastric, nasal, or oral contents
can lead to recurrent bronchitis or bronchiolitis, recurrent pneumonia, atelectasis,
wheezing, cough, apnea, and/or laryngospasm. Pathologic outcomes include
granulomatous inflammation, interstitial inflammation, fibrosis, lipoid
pneumonia, and bronchiolitis obliterans. Most cases clinically manifest as
airway inflammation and are rarely associated with significant morbidity. Table
426.1 lists underlying disorders that are frequently associated with recurrent
aspiration. Oropharyngeal incoordination is reportedly the most common
underlying problem associated with recurrent pneumonias in hospitalized
children. In 2 reports, between 26% and 48% of such children were found to
have dysphagia with aspiration as the underlying problem. Lipoid pneumonia
may occur after the use of home/folk remedies involving oral or nasal
administration of animal or vegetable oils to treat various childhood illnesses.
Lipoid pneumonia has been reported as a complication of these practices in the
Middle East, Asia, India, Brazil, and Mexico. The initial underlying disease,
language barriers, and a belief that these are not medications may delay the
diagnosis (see Chapter 11 ).
Table 426.1
Conditions Predisposing to Aspiration Lung Injury in
Children
Anatomical and Mechanical
Tracheoesophageal fistula
Laryngeal cleft
Vascular ring
Cleft palate
Micrognathia
Macroglossia
Cysts, tumors
Achalasia
Esophageal foreign body
Tracheostomy
Endotracheal tube
Nasal or oral feeding tube
Collagen vascular disease (scleroderma, dermatomyositis)
Gastroesophageal reflux disease
Obesity
Neuromuscular
Altered consciousness
Immaturity of swallowing/Prematurity
Dysautonomia
Increased intracranial pressure
Hydrocephalus
Vocal cord paralysis
Cerebral palsy
Muscular dystrophy
Hypotonia
Myasthenia gravis
Guillain-Barré syndrome
Spinal muscular atrophy
Ataxia-telangiectasia
Cerebral vascular accident
Miscellaneous
Poor oral hygiene
Gingivitis
Prolonged hospitalization
Gastric outlet or intestinal obstruction
Poor feeding techniques (bottle propping, overfeeding, inappropriate foods for toddlers)
Bronchopulmonary dysplasia
Viral infection/bronchiolitis
Table 426.2
Summary of Diagnostic Tests of Aspiration
Etiology
The most common sources of offending agents that cause HP include
agricultural aerosols, inhaled protein antigens from animals, antigens from
microorganisms of bacteria, fungi, or protozoan origin, as well as chemicals of
low and high molecular weight (Table 427.1 ). Many inciting agents are
associated with occupational diseases in which children do not regularly work.
However, the same diseases can occur in children due to exposures to similar
antigen sources in nonoccupational environments, or in occupational
environments with teenage workers. In addition to HP, the same antigens may
lead to allergic asthma or chronic bronchitis as seen with animal proteins,
contaminated metal working fluids, and other inhaled antigens.
Table 427.1
Antigen Sources Associated with Specific Causes of
Hypersensitivity Pneumonitis
More than 300 antigens have been associated with HP. In children, primary
sources of HP have been the result of exposure to pet birds (or feathers in
bedding and pillows) such as parakeets, canaries, cockatiels, or cockatoos.
Aerosol spread of bird droppings can also occur by clothes dryer vents or by
heating vents from a garage where the pet birds were housed. Humidifiers and
hot tubs are notorious for contamination with thermophilic organisms (bacteria
and mold) as well as Mycobacterium avium complex. Buildings with inadequate
ventilation and insufficient air turnover present an increased risk of mold
exposure from prior flooding or damp condensation. Despite exposures to the
same antigen sources, members of the same family may exhibit different
presentations of allergic disease. Some family members may have symptoms of
asthma or rhinitis, while another may have HP.
Pathogenesis
HP has been traditionally classified as acute, subacute, or chronic. During the
acute phase, the offending antigen triggers an inflammatory response promoting
the development of immune complexes. These immune complexes activate the
complement pathway, ultimately resulting in the accumulation of neutrophils in
the airway that release enzymes such as neutrophil elastase, that damage
surrounding lung tissue. Activated macrophages in the lung promote recruitment
of lymphocytes into the tissues. Pathology shows alveolitis with a mixed cellular
infiltration comprised of lymphocytes, macrophage, plasma cells, and
neutrophils. Continued exposure to the offending antigen will lead to subacute or
chronic HP. This chronic exposure results in the formation of loose,
noncaseating granulomas located near the respiratory or terminal bronchioles. It
is critical when a biopsy is being performed (transbronchial or surgical) that the
pathologist knows that HP is being considered as there are other interstitial lung
diseases (ILDs) that produce similar granulomas with subtle location differences
depending on their origin.
Table 427.2
Recurrent pneumonia
Pneumonia after repeat exposures (week, season, situation)
Cough, fever, and chest symptoms after making a job change or home change
Cough, fever, wheezing after return to school or only at school
Pet exposure (especially birds that shed dust such as pigeons, canaries, cockatiels, cockatoos)
Bird contaminant exposure (e.g., pigeon infestation)
Farm exposure to birds and hay
History of water damage despite typical cleaning
Use of hot tub, sauna, swimming pool
Other family members or workers with similar recurrent symptoms
Improvement after temporary environment change (e.g., vacation)
Table 427.3
Criteria Used in the Diagnosis of Hypersensitivity
Pneumonitis
1. Identified exposure to offending antigen(s) by:
• Medical history of exposure to suspected antigen in the patient's living environment
• Investigations of the environment confirm the presence of an inciting antigen
• Identification of specific immune responses (immunoglobulin G serum precipitin antibodies against the
identified antigen) are suggestive of the potential etiology but are insufficient in isolation to confirm a
diagnosis
2. Clinical, radiographic, or physiologic findings compatible with hypersensitivity pneumonitis:
• Respiratory and often constitutional signs and symptoms
• Crackles on auscultation of the chest
• Weight loss
• Cough
• Breathlessness
• Episodic fever
• Wheezing
• Fatigue
NOTE: These findings are especially suggestive of hypersensitivity pneumonitis when they appear or worsen
several hours after antigen exposure.
• A reticular, nodular, or ground-glass opacities on chest radiograph or high-resolution CT
• Abnormalities in the following pulmonary function tests
• Spirometry (restrictive, obstructive, or mixed patterns)
• Lung volumes (low or high)
• Reduced diffusion capacity by carbon monoxide
• Altered gas exchange either at rest or with exercise (reduced partial pressure of arterial oxygen by blood
gas or pulse oximeter testing)
3. Bronchoalveolar lavage with lymphocytosis:
• Usually with low CD4:CD8 ratio (i.e., CD8 is higher than normal)
• Lymphocyte stimulation by offending antigen results in proliferation and cytokine production
4. Abnormal response to inhalation challenge testing to the offending antigen:
• Reexposure to the environment
• Inhalation challenge to the suspected antigen (rarely done now because of the risk of exacerbating the
disease)
5. Histopathology showing compatible changes with hypersensitivity pneumonitis by 1 of these findings:
• Poorly formed, noncaseating granulomas (most often found closer to the respiratory epithelium where
deposition of the offending antigen occurs)
• Mononuclear cell infiltrate in the pulmonary interstitium
Laboratory
Most of the abnormal laboratory findings in HP are not specific and represent
evidence of activated inflammatory markers or lung injury. Nonspecific
elevation of immune globulins or the erythrocyte sedimentation rate and C-
reactive protein may also be found. Circulating immune complexes may be
detected. Lactate dehydrogenase may be elevated in the presence of lung
inflammation and normalizes with response to therapy.
Serum IgG precipitins to the offending agent are frequently positive and have
a poor positive predictive value for disease. Among asymptomatic pigeon
breeders, precipitating antibodies are nearly universal. False negatives can also
be seen due to fluctuating serum antibody levels over time, and lack of
standardized commercial antigens and reagents available for laboratory testing.
It is critical that laboratories familiar with the performance of these tests be
utilized. Those laboratories often recognize the value of processing antigens for
precipitation from the environmental source directly as the test substrate with
patient serum. Skin testing for IgE-mediated disease is not warranted unless
there is evidence of mixed lung pathology such as asthma and interstitial lung
opacities.
Radiology
Chest radiograph almost always precedes the use of high-resolution
computerized tomography (HRCT) of the chest in children because of the need
for sedation and concerns regarding risk of irradiation dose from HRCT. The
plain radiograph will often demonstrate a ground-glass appearance, interstitial
prominence, with a predominant location in the upper and middle lung fields. It
is common for a chest radiograph to be considered normal by a radiologist early
in the disease progression. Late in the disease, interstitial fibrosis may become
prominent in the presence of increasing dyspnea, hypoxemia on room air, and
clubbing of the fingers. Mediastinum widening from lymphadenopathy is not
reactive upper airway disease syndrome
high molecular weight
low molecular weight
Table 427.4
Table 427.5
High Molecular Weight Antigens Known to Induce
Occupational or Environmental Asthma
OCCUPATION
OR SOURCE
ENVIRONMENT
ANIMAL-DERIVED ANTIGENS
Agricultural Cow dander
worker
Bakery Lactalbumin
Butcher Cow bone dust, pig, goat dander
Cook Raw beef
Dairy industry Lactoserum, lactalbumin
Egg producer Egg protein
Farmer Deer dander, mink urine
Frog catcher Frog
Hairdresser Sericin
Ivory worker Ivory dust
Laboratory Bovine serum albumin, laboratory animal, monkey dander
technician
Nacre buttons Nacre dust
Pharmacist Endocrine glands
Pork producer Pig gut (vapor from soaking water)
Poultry worker Chicken
Tanner Casein (cow's milk)
Various Bat guano
Veterinarian Goat dander
Zookeeper Birds
CRUSTACEANS, SEAFOOD, FISH
Canning factory Octopus
Diet product Shark cartilage
Fish food factory Gammarus shrimp
Fish processor Clam, shrimp, crab, prawn, salmon, trout, lobster, turbot, various fishes
Fisherman Red soft coral, cuttlefish
Jewelry polisher Cuttlefish bone
Laboratory grinder Marine sponge
Oyster farm Hoya (oyster farm prawn or sea-squirt)
Restaurant seafood Scallop and shrimp
handler
Scallop plant King scallop and queen scallop
processor
Technician Shrimp meal (Artemia salina)
ARTHROPODS
Agronomist Bruchus lentis
Bottling Ground bug
Chicken breeder Herring worm (Anisakis simplex)
Engineer at electric Caddis flies (Phryganeidae)
power plant
Entomologist Lesser mealworm (Alphitobius diaperinus Panzer), moth, butterfly
Farmer Grain pests (Eurygaster and Pyrale )
Fish bait handler Insect larvae (Galleria mellonella) , mealworm larvae (Tenebrio molitor) , green bottle fly
larvae (Lucila caesar) , daphnia, fish-feed Echinodorus larva (Echinodorus plasmosus) ,
Chiromids midge (Chironomus thummi thummi)
Fish processing Herring worm (Anisakis simplex)
Flight crew Screw worm fly (Cochliomyia hominivorax)
Honey processors Honeybee
Laboratory worker Cricket, fruit fly, grasshopper (Locusta migratoria) , locust
Mechanic in a rye Confused flour beetle (Tribolium confusum)
plant
Museum curator Beetles (Coleoptera)
Seed house Mexican bean weevil (Zabrotes subfasciatus)
Sericulture Silkworm, larva of silkworm
Sewage plant Sewer fly (Psychoda alternata)
worker
Technician Arthropods (Chrysoperla carnea, Leptinotarsa decemlineata, Ostrinia nubilalis, and
Ephestia kuehniella), sheep blowfly (Lucilia cuprina)
Wool worker Dermestidae spp.
ACARIANS
Apple grower Fruit tree red spider mite (Panonychus ulmi)
Citrus farmer Citrus red mite (Panonychus citri)
Farmer Barn mite, two-spotted spider mite (Tetranychus urticae) , grain mite
Flour handler Mites and parasites
Grain-store worker Grain mite
Horticulturist Amblyseius cucumeris
Poultry worker Fowl mite
Vine grower McDaniel spider mite (Tetranychus mcdanieli)
MOLDS
Agriculture Plasmopara viticola
Baker Alternaria, Aspergillus (unspecified)
Beet sugar worker Aspergillus (unspecified)
Coal miner Rhizopus nigricans
Coffee maker Chrysonilia sitophila
Laborer Sooty molds (Ascomycetes , deuteromycetes)
Logging worker Chrysonilia sitophila
Plywood factory Neurospora
worker
Sausage processing Penicillium nalgiovense
Sawmill worker Trichoderma koningii
Stucco worker Mucor spp. (contaminating esparto fibers)
Technician Dictyostelium discoideum (mold), Aspergillus niger
MUSHROOMS
Agriculture Agaricus bisporus (white mushroom)
Baker Baker's yeast (Saccharomyces cerevisiae), Boletus edulis
Greenhouse worker Sweet pea (Lathyrus odoratus)
Hotel manager Boletus edulis
Mushroom Pleurotus cornucopiae
producer
Mushroom soup Mushroom unspecified
processor
Office worker Boletus edulis
Seller Pleurotus ostreatus (spores of white spongy rot)
ALGAE
Pharmacist Chlorella
Thalassotherapist Algae (species unspecified)
FLOURS
Animal fodder Marigold flour (Tagetes erecta)
Baker Wheat, rye, soya, and buckwheat flour; Konjac flour; white pea flour (Lathyrus sativus)
Food processing White Lupin flour (Lupinus albus)
POLLENS
Florist Cyclamen, rose
Gardener Canary island date palm (Phoenix canariensis) , Bell of Ireland (Moluccella laevis) , Bell
pepper, chrysanthemum, eggplant (Solanum melongena) , Brassica oleracea (cauliflower
and broccoli)
Laboratory worker Sunflower (Helianthus spp.), thale cress (Arabidopsis thaliana)
Olive farmer White mustard (Sinapis alba)
Processing worker Helianthus annuus
PLANTS
Brewery chemist Hops
Brush-maker Tampico fiber in agave leaves
Butcher Aromatic herb
Chemist Linseed oilcake, Voacanga africana seed dust
Cosmetics Dusts from seeds of Sacha Inchi (Plukenetia volubilis) , chamomile (unspecified)
Decorator Cacoon seed (Entage gigas)
Floral worker Decorative flower, safflower (Carthamus tinctorius) and yarrow (Achillea millefolium) ,
spathe flower, statice (Limonium tataricum) , baby's breath (Gypsophila paniculata) , ivy
(Hedera helix) , flower (various), sea lavender (Limonium sinuatum)
Food industry Aniseed, fenugreek, peach, garlic dust, asparagus, coffee bean, sesame seed, grain dust,
carrot (Daucus carota L.) , green bean (Phaseolus multiflorus) , lima bean (Phaseolus
lunatus), onion, potato, swiss chard (Beta vulgaris L.) , courgette, carob bean, spinach
powder, cauliflower, cabbage, chicory, fennel seed, onion seeds (Allium cepa , red onion),
rice, saffron (Crocus sativus), spices, grain dust
Gardener Copperleaf (Acalypha wilkesiana), grass juice, weeping fig (Ficus benjamina), umbrella tree
(Schefflera spp.), amaryllis (Hippeastrum spp.), Madagascar jasmine sap (Stephanotis
floribunda) , vetch (Vicia sativa)
Hairdresser Henna (unspecified)
Herbal tea Herbal tea, sarsaparilla root, sanyak (Dioscorea batatas), Korean ginseng (Panax ginseng),
processor tea plant dust (Camellia sinensis) , chamomile (unspecified)
Herbalist Licorice roots (Glycyrrhiza spp.), wonji (Polygala tenuifolia) , herb material
Horticulture Freesia (Freesia hybrida), paprika (Capsicum annuum) , Brazil ginseng (Pfaffia paniculata)
Laborer Citrus food handling (dl -limonene, l -citronellol, and dichlorophen)
Oil industry Castor bean, olive oilcake
Pharmaceutical Rose hip, passion flower (Passiflora alata) , cascara sagrada (Rhamnus purshiana)
Powder Lycopodium powder
Sewer Kapok
Sheller Almond shell dust
Stucco handler Esparto (Stipa tenacissima and Lygeum spartum )
Tobacco Tobacco leaf
manufacturer
PLANT-DERIVED NATURAL PRODUCTS
Baker Gluten, soybean lecithin
Candy maker Pectin
Glove Latex
manufacturer
Health professional Latex
Rose extraction Rose oil
BIOLOGIC ENZYMES
Baker Fungal amylase, fungal amyloglucosidase and hemicellulase
Cheese producer Various enzymes in rennet production (proteases, pepsine, chymosins)
Detergent industry Esterase, Bacillus subtilis
Factory worker Bacillus subtilis
Fruit processor Pectinase and glucanase
Hospital personnel Empynase (pronase B)
Laboratory worker Xylanase, phytase from Aspergillus niger
Pharmaceutical Bromelin, flaviastase, lactase, pancreatin, papain, pepsin, serratia peptidase, and lysozyme
chloride; egg lysozyme, trypsin
Plastic Trypsin
VEGETABLE GUMS
Carpet Guar
manufacturing
Dental hygienist Gutta-percha
Gum importer Tragacanth
Hairdresser Karaya
Printer Acacia
Table 427.6
Low Molecular Weight Chemicals Known to Induce
Occupational or Environmental Asthma
Table 427.7
Criteria for the Diagnosis of Reactive Airways Disease
Syndrome
• Absence of previous documented respiratory symptom
• Onset of symptoms most often occur after a single specific exposure
• Exposure is most often to a high concentration of gas, smoke, fume, or vapor with irritant qualities
• Symptoms occur within 24 hr of exposure and persist for 3 mo or longer
• Symptoms mimic asthma with cough, wheezing, shortness of breath, and/or dyspnea
• Pulmonary function tests may demonstrate airflow obstruction but not always
• Bronchial hyperresponsiveness is documented by methacholine challenge
• Alternative pulmonary diseases are not able to be found
Table 427.8
Key Elements in the Medical History and Physical Exam to
Raise Clinical Suspicion for Diagnostic Testing to Confirm
Eosinophilic Lung Disease
Medical history and examination
• Drug exposure (especially antibiotics, NSAIDs, antiepileptics, antileukotriene modifiers in EGPA) (see Table
478.10)
• Environmental inhalation exposures to dust or inhaled chemicals
• New onset of smoking cigarettes
• Travel or immigration status from areas endemic with various parasites or coccidioidomycosis
• Asthma (may be severe or poorly controlled with ABPA, CSS, or is relatively new in onset with IAEP)
• ABPA concurrent in 7–10% of patients with cystic fibrosis
• Extrapulmonary symptoms suggestive of vasculitis, neuropathy, heart failure, or neoplasm
• Rash (creeping eruption in visceral larval migrans disease or ulceration in EGPA)
Diagnostic imaging and testing
• Radiography helpful in AEP, CEP, and ABPA
• Radiography not diagnostic in EGPA or drug-induced eosinophilic disease of the lung
• Simple chest radiography findings
• Nonlobar infiltrate
• Classic description as mirror image of pulmonary edema with peripheral infiltrates
• Bilateral pleural effusion in AEP
• Central bronchiectasis in ABPA
• High-resolution computerized tomography of the chest
• Middle and upper lobe nonlobar infiltrates with areas of ground-glass appearance
• Mucous plugging in ABPA
• Central bronchiectasis in ABPA (confused with cystic fibrosis)
• Blood eosinophil count
• Elevated in many eosinophilic lung diseases
• Magnitude of eosinophil blood count does not distinguish different pulmonary diseases
• Usually not elevated in AEP (eosinophilic disease compartmentalized to lungs)
• May occasionally not be elevated in CEP or after use of corticosteroids
• Total serum IgE elevated in ABPA but not always in patients with cystic fibrosis with ABPA
• Serology for helminthic infections or parasites may be diagnostic but are usually not available acutely
• P-ANCA (MPO ANCA) is positive in 40–70% of EGPA (CSS)
• BAL eosinophil percentage
• ≥25% eosinophils diagnostic in AEP
• ≥40% eosinophils diagnostic in CEP or tropical pulmonary eosinophilia
• Eosinophil percentages below these criteria may require lung biopsy
• <25% eosinophils seen in connective tissue disease, sarcoid, drug-induced disease, histiocytosis X of
pulmonary Langerhans cells, and interstitial pulmonary fibrosis
• Lung biopsy
• Open lung biopsy or video-assisted thorascopic surgery when BAL nondiagnostic
• Transbronchial biopsy is usually insufficient with peripheral infiltrative disease
• Histology with alveolar and interstitial infiltrates of eosinophils, non-necrotizing non-granulomatous
vasculitis, multinucleated giant cells without granuloma
• EGPA shows eosinophil rich small to medium vessel, necrotizing, granulomatous vasculitis
ABPA , Allergic bronchopulmonary aspergillosis; AEP , acute eosinophilic pneumonia; BAL ,
bronchoalveolar lavage; CEP , chronic eosinophilic pneumonia; CSS , Churg-Strauss syndrome;
EGPA , eosinophilic granulomatosis with polyangiitis; IAEP , idiopathic acute eosinophilic
pneumonia; MPO ANCA , myeloperoxidase antineutrophil cytoplasmic antibody; NSAID ,
nonsteroidal antiinflammatory drug; P-ANCA , perinuclear antineutrophil cytoplasmic antibody.
Table 427.9
Classification of the Eosinophilic Pneumonias in Clinical
Practice
Eosinophilic pneumonias of unknown cause
Solitary idiopathic eosinophilic pneumonias
Idiopathic chronic eosinophilic pneumonia
Idiopathic acute eosinophilic pneumonia
Eosinophilic pneumonia in systemic syndromes
Eosinophilic granulomatosis with polyangiitis
Idiopathic hypereosinophilic syndromes (lymphocytic or myeloproliferative variant)
Eosinophilic pneumonias of known cause
Allergic bronchopulmonary aspergillosis and related syndromes (including bronchocentric granulomatosis)
Eosinophilic pneumonias of parasitic origin
Eosinophilic pneumonias of other infectious causes
Drug-induced eosinophilic pneumonias
Eosinophilic airways diseases
Eosinophilic asthma
Hypereosinophilic asthma
Idiopathic hypereosinophilic constrictive bronchitis
Other pulmonary syndromes with possible usually mild eosinophilia
Organizing pneumonia, asthma, idiopathic pulmonary fibrosis, Langerhans cell histiocytosis, malignancies, and so
forth
From Cottin V: Eosinophilic lung diseases, Clin Chest Med 37:535–556, 2016 (Box 1, p. 536).
Table 427.10
Antiinflammatory drugs and related drugs: acetylsalicylic acid, diclofenac, ibuprofen, naproxen, phenylbutazone,
piroxicam, sulindac, and tolfenamic acid
Antibiotics: ethambutol, fenbufen, minocycline, nitrofurantoin, penicillins, pyrimethamine, sulfamides,
sulfonamides, and trimethoprim-sulfamethoxazole
Other drugs: captopril, carbamazepine, and GM-CSF
A more extensive list of drugs reported to cause eosinophilic pneumonia may be found at www.pneumotox.com .
From Cottin V, Cordier JF: Eosinophilic lung diseases, Immunol Allergy Clin
North Am 32(4):557–586, 2012 (Box 6, p. 575).
Table 427.11
Diagnostic Criteria for Idiopathic Chronic Eosinophilic
Pneumonia and for Idiopathic Acute Eosinophilic
Pneumonia
IDIOPATHIC CHRONIC EOSINOPHILIC PNEUMONIA
1. Diffuse pulmonary alveolar consolidation with air bronchogram and/or ground-glass opacities at chest imaging,
especially with peripheral predominance.
2. Eosinophilia at bronchoalveolar lavage differential cell count ≥ 40% (or peripheral blood eosinophils ≥
1,000/mm3 ).
3. Respiratory symptoms present for at least 2 to 4 wk.
4. Absence of other known causes of eosinophilic lung disease (especially exposure to drug susceptible to induce
pulmonary eosinophilia).
IDIOPATHIC ACUTE EOSINOPHILIC PNEUMONIA
1. Acute onset with febrile respiratory manifestations (≤1 mo, and especially ≤ 7 days duration before medical
examination).
2. Bilateral diffuse infiltrates on imaging.
3. PaO 2 on room air ≤ 60 mm Hg (8 kPa), or PaO 2 /FIO 2 ≤ 300 mm Hg (40 kPa), or oxygen saturation on room air
< 90%.
4. Lung eosinophilia, with ≥ 25% eosinophils at BAL differential cell count (or eosinophilic pneumonia at lung
biopsy when done).
5. Absence of determined cause of acute eosinophilic pneumonia (including infection or exposure to drugs known
to induce pulmonary eosinophilia). Recent onset of tobacco smoking or exposure to inhaled dusts may be
present.
BAL , Bronchoalveolar lavage.
From Cottin V: Eosinophilic lung diseases, Clin Chest Med 37:535–556, 2016 (Box 2, p. 538).
Table 427.12
The pathogenesis of EGPA is still unknown but several factors are suspected
to contribute to the development of the disease. The possible link between
leukotriene-receptor antagonists (zafirlukast, montelukast, or pranlukast) is
controversial but still considered possible. It is suspected that use of this class of
adjunctive medications in severe asthma allows for the reduction in use of
corticosteroid leading to the full-blown (unmasking) manifestation of EGPA.
Isolated use of leukotriene-receptor antagonists may induce disease, lead to
remission with cessation of leukotriene-receptor antagonists, and cause
recurrence of EGPA upon reintroduction of this class of medications. Many
refrain from use of leukotriene-receptor antagonists when the EGPA syndrome
has been diagnosed.
Clinical and laboratory findings pinpoint the diagnosis with high specificity
(99.7%) and sensitivity (85%) when 4 of 6 criteria are met (asthma, eosinophilia
>10%, mononeuropathy or polyneuropathy, nonfixed pulmonary infiltrates,
paranasal sinus abnormalities, and biopsy findings of extravascular eosinophil
infiltrates). In contrast to GPA, the rhinitis is not destructive and nasal septal
perforation does not occur in EGPA.
Radiography of the chest by plain radiography or HRCT demonstrates the
migratory, peripheral predominant opacities with ground-glass appearance to full
consolidation. Bronchiectasis and bronchial wall thickening are reported. Pleural
effusion should raise suspicion for the presence of heart failure from
cardiomyopathy .
Laboratory findings include striking eosinophilia with values generally
between 5,000 and 20,000/mm3 at the time of diagnosis. These counts often
parallel vasculitis activity. The BAL shows striking eosinophilia with differential
counts of >60%. Other organ system levels reflect activity of eosinophils and are
not specific for the EGPA diagnosis.
ANCAs may be present in the EGPA syndrome. The perinuclear-ANCA
targeting myeloperoxidase is specifically found in EGPA in approximately 40%
Table 427.13
Criteria for the Diagnosis of Allergic Bronchopulmonary
Aspergillosis
Allergic bronchopulmonary aspergillosis–central bronchiectasis
• Medical history of asthma*
• Immediate skin prick test reaction to Aspergillus antigens*
• Precipitating (IgG) serum antibodies to Aspergillus fumigatus *
• Total IgE concentration > 417 IU/mL (>1,000 ng/mL)*
• Central bronchiectasis on chest CT*
• Peripheral blood eosinophilia >500/mm3
• Lung infiltrates on chest x-ray or chest HRCT
• Elevated specific serum IgE and IgG to A. fumigatus
Allergic bronchopulmonary aspergillosis seropositive †
• Medical history of asthma †
• Immediate skin prick test reaction to A. fumigatus antigens †
• Precipitating (IgG) serum antibodies to A. fumigatus †
• Total IgE concentration > 417 IU/mL (>1,000 ng/mL) †
* The criteria required for diagnosis of ABPA with central bronchiectasis.
Table 427.14
Staging of Allergic Bronchopulmonary Aspergillosis
Staging of allergic bronchopulmonary aspergillosis
Stage 1 Acute Upper and middle lobe infiltration High IgE
Stage 2 Remission No infiltrate off steroids >6 mo Normal to high IgE
Stage 3 Exacerbation Upper and middle lobe infiltration High IgE
Stage 4 CSD asthma Minimal infiltrate Normal to high IgE
Stage 5 End stage Fibrosis and/or bullae Normal
CSD , Corticosteroid dependent.
dose exceeds 200 mg, then the total dose should be divided equally and given
twice daily. Serum levels of itraconazole are necessary to ensure proper
absorption of the drug is occurring from the capsule form. The liquid form is
more readily absorbed and has achieved substantially higher levels. The use of
proton pump inhibitors and histamine 2 antagonists may reduce absorption by
blocking acid production. Voriconazole has been used as a substitute antifungal
medication. Proper dosing has been established for invasive Aspergillus disease,
but not for ABPA. Typical dosage regimen in children of 7 mg/kg/day may cause
hepatotoxicity so liver function must be monitored.
Omalizumab, an anti-IgE humanized monoclonal antibody, has been used in
case series of patients with cystic fibrosis and ABPA as well as a small cohort of
adults without cystic fibrosis but with ABPA. Both case series demonstrated
significant reductions in asthma exacerbations, ABPA exacerbations, and
glucocorticoid usage. The dose prescribed has been 300-375 mg every 2 wk by
subcutaneous injection.
Hypereosinophilic Syndrome
See Chapter 155 .
The HES is a descriptive name of a group of disorders that are characterized
by the persistent overproduction of eosinophils accompanied by eosinophil
infiltration in multiple organs with end-organ damage from mediator release.
The term HES should only be used when there is eosinophilia with end-organ
damage from the eosinophils and not from another cause. The discovery of
underlying genetic, biochemical, or neoplastic reasons for HES has led to the
classification of primary, secondary, and idiopathic HES (Table 427.15 ).
Specific syndromes such as EGPA have eosinophilia but the contribution of
eosinophils to the organ damage is incompletely understood.
Table 427.15
Table 427.16
The Pediatric Interstitial Lung Diseases in Children Under 2
Yr of Age
AGE-RELATED INTERSTITIAL LUNG DISEASES IN INFANCY AND EARLY CHILDHOOD
Diffuse developmental disorders
Acinar dysplasia
Congenital alveolar dysplasia
Alveolar capillary dysplasia with misalignment of pulmonary veins (some due to FOXF1 mutation)
Growth abnormalities reflecting deficient alveolarization
Pulmonary hypoplasia
Chronic neonatal lung disease
Chromosomal disorders
Congenital heart disease
Neuroendocrine cell hyperplasia of infancy
Pulmonary interstitial glycogenosis (infantile cellular interstitial pneumonia)
Surfactant dysfunction disorders (pulmonary alveolar proteinosis)
Surfactant protein–B mutation
Surfactant protein–C mutation
ABCA3 mutation
Granulocyte-macrophage colony-stimulating factor receptor (CSF2RA ) mutation
NKX2.1 (transcription factor for SP-B, SPC, ABCA3)
INTERSTITIAL LUNG DISEASE DISORDERS WITH KNOWN ASSOCIATIONS
Infectious/postinfectious processes
Adenovirus viruses
Influenza viruses
Chlamydia pneumoniae
Mycoplasma pneumoniae
Environmental agents
Hypersensitivity pneumonitis
Toxic inhalation
Aspiration syndromes
PULMONARY DISEASES ASSOCIATED WITH PRIMARY AND SECONDARY IMMUNE
DEFICIENCY
Opportunistic infections
Granulomatous lymphocytic interstitial lung disease associated with common variable immunodeficiency
syndrome
Lymphoid intestinal pneumonia (HIV infection)
Therapeutic interventions: chemotherapy, radiation, transplantation, and rejection
IDIOPATHIC INTERSTITIAL LUNG DISEASES
Usual interstitial pneumonitis
Desquamative interstitial pneumonitis
Lymphocytic interstitial pneumonitis and related disorders
Nonspecific interstitial pneumonitis (cellular/fibrotic)
Eosinophilic pneumonia
Bronchiolitis obliterans syndrome
Pulmonary hemosiderosis and acute idiopathic pulmonary hemorrhage of infancy
Pulmonary alveolar proteinosis
Pulmonary vascular disorders
Pulmonary lymphatic disorders
Pulmonary microlithiasis
Persistent tachypnea of infancy
Brain-thyroid-lung syndrome
SYSTEMIC DISORDERS WITH PULMONARY MANIFESTATIONS
Anti-GBM disease
Gaucher disease and other storage diseases
Malignant infiltrates
Hemophagocytic lymphohistiocytosis
Langerhans cell histiocytosis
Sarcoidosis
Systemic sclerosis
Polymyositis/dermatomyositis
Systemic lupus erythematosus
Rheumatoid arthritis
Lymphangioleiomyomatosis
Pulmonary hemangiomatosis
Neurocutaneous syndromes
Hermansky-Pudlak syndrome
Modified from Deutsch GH, Young LR, Deterding RR, et al: diffuse lung disease in young children:
application of a novel classification scheme, Am J Respir Crit Care Med 176:1120–1128, 2007.
Table 427.17
The Pediatric Interstitial Lung Diseases in Children Over 2
Yr of Age
DISORDERS OF THE IMMUNOCOMPETENT HOST
Disorders of Infancy
• Growth abnormalities
• NEHI
• Disorders of surfactant metabolism
Systemic Disease
• Immune mediated disorders
• Connective tissue disease related lung disease
• Pulmonary hemorrhage syndromes
• Storage diseases
• Sarcoidosis
DISORDERS OF THE IMMUNOCOMPROMISED HOST
• Opportunistic infections
• Related to treatment
• Chemotherapy
• Radiation
• Drug hypersensitivity
• Related to transplantation
• Rejection
• GVHD
• PTLD
• Lymphoid Infiltrates
GVHD , Graft-versus-host disease; NEHI , neuroendocrine cell hyperplasia of infancy; PTLD ,
post-transplant lymphoproliferative disease.
Modified from Fan LL, Dishop MK, Galambos C, et al: Diffuse lung disease in biopsied children 2
to 18 years of age. Application of the chILD Classification Scheme, Ann Am Thorac Soc
2015;12(10):1498–1505.
Table 427.18
Table 427.19
Diseases Associated With Pulmonary Fibrosis
• Idiopathic pulmonary fibrosis / nonspecific interstitial pneumonia
• Familial pulmonary fibrosis / familial interstitial pneumonia
• Hypersensitivity pneumonitis (many agents)
• Cryptogenic organizing pneumonia
• Adverse reaction to therapy (drugs, radiation)
• Pleuroparenchymal fibroelastosis
• Hermansky–Pudlak syndrome
• Sarcoidosis
• Eosinophilic pneumonia (primary or parasitic)
• Langerhans cell histiocytosis
• Dyskeratosis congenita
• Tuberous sclerosis
• Neurofibromatosis
• Erdheim–Chester disease
• Gaucher disease
• Niemann–Pick disease
• Familial hypocalciuric hypercalcemia
• Lysinuric protein intolerance
• IgG4 mediated immune disorder
• Myelodysplastic syndrome
• Progressive systemic sclerosis
• Other connective tissue diseases (SLE, dermatomyositis)
• Granulomatosis with polyangiitis
• Eosinophilic granulomatosis with polyangiitis
Table 427.20
Pediatric Fibrotic Lung Diseases
PATHOLOGY ADDITIONAL
DISEASES CT FINDINGS TREATMENT
FINDINGS EVALUATION
Surfactant Early: Diffuse ground Variable: fibrosis, Genetic testing Hydroxychloroquine,
dysfunction glass, septal honeycomb cysts at end azithromycin, high-
thickening (crazy stage, NSIP, CPI, few dose intravenous
paving) Chronic: See globules of pulmonary steroids. Genetic
NSIP alveolar proteinosis, foamy counseling
macrophages and
cholesterol clefts
(endogenous lipoid
pneumonia)
Aspiration Acute: Consolidation Airway-centered Video Stop aspiration
and centrilobular (tree lesions/bronchiolitis, food fluoroscopic through thickening
in bud) nodules with a particles with or without swallow feeds, gastric feeds,
dependent distribution. granulomas, foamy evaluation cleft repair
Chronic: possible UIP macrophages (endogenous
with honeycombing lipoid pneumonia),
organizing pneumonia
Radiation fibrosis
Architectural Pleural, septal, and
distortion, volume paraseptal fibrosis; reactive
loss, traction atypia of alveolar
bronchiectasis. Often epithelium and endothelium
with geometric
distribution related to
radiation field
Bronchopulmonary Hyperlucent regions, Alveolar simplification and Supportive care.
dysplasia architectural distortion enlargement. Patchy Consider inhaled
(linear and subpleural hyperinflation. Interstitial corticosteroids
triangular opacities) fibrosis, with or without
interlobular septal fibrosis.
Nonspecific Basilar predominant Interstitial lymphocytic
interstitial findings of ground- inflammation and fibrosis
pneumonia (NSIP) glass opacities (often with homogenous
with subpleural distribution
sparing), reticulation,
architectural
distortion, and traction
bronchiectasis
Hypersensitivity Patchy and often Airway-centered small non- Lymphocytosis Remove trigger,
pneumonitis parahilar reticulation, caseating granulomas, in intravenous steroids
(chronic) ground glass, multinucleated giant cells, bronchoalveolar
centrilobular nodules. lymphocytic bronchiolitis lavage,
Honeycombing (rare) and peribronchiolitis, precipitins to
airway fibrosis, organizing specific antigen
pneumonia
Autoimmune See NSIP. NSIP. Lymphoid Serologic Disease-specific
connective tissue Honeycombing (rare) hyperplasia. Fibrosis and studies immune modulation
disorders (collagen cystic change. Pleuritis and
vascular disease) pleural fibrosis (variable).
Chronic vasculopathy
(variable). Airway fibrosis
(variable).
Drug reactions Peripheral Variable: organizing Drug avoidance
predominant pneumonia, NSIP, UIP,
consolidation or DAD, pulmonary
ground glass. Reverse hemorrhage, eosinophilic
halo sign. See NSIP. pneumonia
Honeycombing (rare)
Infection Acute: Consolidation Acute: Neutrophilic Antimicrobials
and centrilobular (tree alveolitis (bacterial) or
in bud) nodules. lymphocytic bronchiolitis
Appearance and (viral). Chronic: Variable
distribution varies airway fibrosis
with type of infection. (constrictive/obliterative
Chronic: May bronchiolitis) and
progress to IPF/UIP interstitial fibrosis.
with honeycombing
Immunodeficiency Bronchiectasis, Follicular bronchiolitis or Immunologic Treat underlying
consolidation, diffuse lymphoid and genetic immunodeficiency
centrilobular nodules hyperplasia. NSIP. LIP. testing
GLILD
Table 427.21
† Rarer variant.
Adapted from Kaur A, Mathai SK, Schwartz DA: Genetics in idiopathic pulmonary fibrosis
pathogenesis, prognosis, and treatment. Frontiers Med 4:154, 2017. Tables 1 and 2.
Clinical Presentation
Patients with pulmonary fibrosis will typically present with nonspecific
respiratory symptoms such as cough, crackles, wheezes, prolonged expiratory
phase, exercise intolerance, and hypoxemia, especially at nighttime. Symptom
onset can be insidious or rapid.
Evaluation
Pulmonary function tests typically show restriction and reduced diffusion
capacity. Air trapping can also be seen. Patients may desaturate with exercise
challenges or 6-min walks.
There are a variety of findings on computed tomography scan that suggest
pulmonary fibrosis. These include reticular opacities, architectural distortion,
traction bronchiectasis, and honeycomb cysts. A common late finding in several
etiologies of fibrotic lung disease in children is nonspecific interstitial
pneumonia: subpleural sparing, ground-glass opacities, reticular change, and
bronchiectasis. Typical CT findings in pediatric patients with nonspecific
CHAPTER 428
Community-Acquired Pneumonia
Matthew S. Kelly, Thomas J. Sandora
Epidemiology
Pneumonia, defined as inflammation of the lung parenchyma, is the leading
infectious cause of death globally among children younger than 5 yr, accounting
for an estimated 920,000 deaths each year (Fig. 428.1 ). Pneumonia mortality is
closely linked to poverty. More than 99% of pneumonia deaths are in low- and
middle-income countries, with the highest pneumonia mortality rate occurring in
poorly developed countries in Africa and South Asia (Table 428.1 ).
FIG. 428.1 Pneumonia is the leading infectious killer of children worldwide,
as shown by this illustration of global distribution of cause-specific
infectious mortality among children younger than age 5 yr in 2015.
Pneumonia causes one-third of all under-5 deaths from infection. (From
World Health Organization and Maternal and Child Epidemiology
Estimation Group estimates, 2015.)
Table 428.1
Pneumonia Cases and Mortality Rate in Children Younger Than Age 5 Yr by UNICEF
Region, 2015
UNICEF PNEUMONIA CASES IN CHILDREN PNEUMONIA MORTALITY RATE (UNDER-
REGIONS YOUNGER THAN 5 YR OF AGE 5 DEATHS PER 1,000 LIVE BIRTHS)
West and Central 298,000 16.2
Africa
Sub-Saharan 490,000 13.7
Africa
Eastern and 177,000 10.9
Southern Africa
South Asia 282,000 7.9
Middle East and 46,000 4.1
North Africa
East Asia and the 81,000 2.7
Pacific
Latin America 23,000 2.1
and the
Caribbean
Least Developed 363,000 12.0
Countries
World 920,000 6.6
From United Nations Children's Fund: One is too many—ending child deaths from pneumonia and
diarrhoea. https://data.unicef.org/resources/one-many-ending-child-deaths-pneumonia-diarrhoea/
. Accessed January 21, 2017.
Etiology
Although most cases of pneumonia are caused by microorganisms, noninfectious
causes include aspiration (of food or gastric acid, foreign bodies, hydrocarbons,
and lipoid substances), hypersensitivity reactions, and drug- or radiation-induced
pneumonitis (see Chapter 427 ). The cause of pneumonia in an individual patient
is often difficult to determine because direct sampling of lung tissue is invasive
and rarely performed. Bacterial cultures of sputum or upper respiratory tract
samples from children typically do not accurately reflect the cause of lower
respiratory tract infection. Streptococcus pneumoniae (pneumococcus) is the
most common bacterial pathogen in children 3 wk to 4 yr of age, whereas
Mycoplasma pneumoniae and Chlamydophila pneumoniae are the most frequent
bacterial pathogens in children age 5 yr and older. In addition to pneumococcus,
other bacterial causes of pneumonia in previously healthy children in the United
States include group A streptococcus (Streptococcus pyogenes ; see Chapter 210
) and Staphylococcus aureus (see Chapter 208.1 ) (Tables 428.2 , 428.3 , and
428.4 ). S. aureus pneumonia often complicates an illness caused by influenza
viruses.
Table 428.2
Causes of Infectious Pneumonia
BACTERIAL
COMMON
Streptococcus pneumoniae Consolidation, empyema
Group B streptococci Neonates
Group A streptococci Empyema
Staphylococcus aureus Pneumatoceles, empyema; infants; nosocomial
pneumonia
Mycoplasma pneumoniae * Adolescents; summer–fall epidemics
Chlamydophila pneumoniae * Adolescents
Chlamydia trachomatis Infants
Mixed anaerobes Aspiration pneumonia
Gram-negative enterics Nosocomial pneumonia
UNCOMMON
Haemophilus influenzae type b Unimmunized
Moraxella catarrhalis
Neisseria meningitidis
Francisella tularensis Animal, tick, fly contact; bioterrorism
Nocardia species Immunocompromised patients
Chlamydophila psittaci * Bird contact (especially parakeets)
Yersinia pestis (plague) Rat contact; bioterrorism
Legionella species* Exposure to contaminated water; nosocomial
Coxiella burnetii * (Q fever) Animal (goat, sheep, cattle) exposure
VIRAL
COMMON
Respiratory syncytial virus Bronchiolitis
Parainfluenza types 1-4 Croup
Influenza A, B High fever; winter months
Adenovirus Can be severe; often occurs between January
and April
Human metapneumovirus Similar to respiratory syncytial virus
UNCOMMON
Rhinovirus Rhinorrhea
Enterovirus Neonates
Herpes simplex Neonates, immunocompromised persons
Cytomegalovirus Infants; immunocompromised persons
(particularly HIV-infected infants)
Measles Rash, coryza, conjunctivitis
Varicella Unimmunized; immunocompromised persons
Hantavirus Southwestern United States, rodents
Coronaviruses [severe acute respiratory syndrome (SARS), Asia, Arabian Peninsula
Middle East respiratory syndrome (MERS)]
FUNGAL
Histoplasma capsulatum Ohio/Mississippi River valley; bird, bat
contact
Blastomyces dermatitidis Ohio/Mississippi River valley
Coccidioides immitis Southwestern United States, Great Lakes states
Cryptococcus neoformans and C. gattii Bird contact; immunocompromised;
Northwestern United States (C. gattii)
Aspergillus species Immunocompromised persons; nodular lung
infection
Mucormycosis Immunocompromised persons
Pneumocystis jiroveci Immunocompromised persons (particularly
HIV-infected infants); steroids
RICKETTSIAL
Rickettsia rickettsiae Tick bite
MYCOBACTERIAL
Mycobacterium tuberculosis Travel to endemic region; exposure to high-
risk persons
Mycobacterium avium complex Immunocompromised (particularly HIV-
infected) persons
Other non-tuberculous mycobacteria Immunocompromised persons; cystic fibrosis
PARASITIC
Various parasites (e.g., Ascaris , Strongyloides species) Eosinophilic pneumonia
* Atypical pneumonia syndrome; may have extrapulmonary manifestations, low-grade fever,
Table 428.3
Adapted from Kliegman RM, Marcdante KJ, Jenson HJ, et al: Nelson essentials of pediatrics , ed
5, Philadelphia, 2006, Elsevier, p. 507.
Table 428.4
Pneumonia: Etiology Suggested by Exposure History
Table 428.5
Differential Diagnosis of Recurrent Pneumonia
HEREDITARY DISORDERS
Cystic fibrosis
Sickle cell disease
DISORDERS OF IMMUNITY
HIV/AIDS
Bruton agammaglobulinemia
Selective immunoglobulin G subclass deficiencies
Common variable immunodeficiency syndrome
Severe combined immunodeficiency syndrome
Chronic granulomatous disease
Hyperimmunoglobulin E syndromes
Leukocyte adhesion defect
DISORDERS OF CILIA
Primary ciliary dyskinesia
Kartagener syndrome
ANATOMIC DISORDERS
Pulmonary sequestration
Lobar emphysema
Congenital cystic adenomatoid malformation
Gastroesophageal reflux
Foreign body
Tracheoesophageal fistula (H type)
Bronchiectasis
Aspiration (oropharyngeal incoordination)
Aberrant bronchus
Adapted from Kliegman RM, Marcdante KJ, Jenson HJ, et al: Nelson essentials of pediatrics , ed
5, Philadelphia, 2006, Elsevier, p. 507.
Clinical Manifestations
Pneumonia is frequently preceded by several days of symptoms of an upper
respiratory tract infection, typically rhinitis and cough. In viral pneumonia, fever
is usually present but temperatures are generally lower than in bacterial
pneumonia. Tachypnea is the most consistent clinical manifestation of
pneumonia. Increased work of breathing accompanied by intercostal, subcostal,
and suprasternal retractions, nasal flaring, and use of accessory muscles is
common. Severe infection may be accompanied by cyanosis and lethargy,
especially in infants. Auscultation of the chest may reveal crackles and
wheezing, but it is often difficult to localize the source of these adventitious
sounds in very young children with hyperresonant chests. It is often not possible
to distinguish viral pneumonia (especially adenovirus) clinically from disease
caused by Mycoplasma and other bacterial pathogens.
Bacterial pneumonia in adults and older children typically begins suddenly
with high fever, cough, and chest pain. Other symptoms that may be seen include
drowsiness with intermittent periods of restlessness; rapid respirations; anxiety;
and, occasionally, delirium. In many children, splinting on the affected side to
minimize pleuritic pain and improve ventilation is noted; such children may lie
on one side with the knees drawn up to the chest.
Physical findings depend on the stage of pneumonia. Early in the course of
illness, diminished breath sounds, scattered crackles, and rhonchi are commonly
heard over the affected lung field. With the development of increasing
consolidation or complications of pneumonia such as pleural effusion or
empyema, dullness on percussion is noted and breath sounds may be diminished.
A lag in respiratory excursion often occurs on the affected side. Abdominal
distention may be prominent because of gastric dilation from swallowed air or
ileus. Abdominal pain is common in lower-lobe pneumonia. The liver may seem
enlarged because of downward displacement of the diaphragm secondary to
hyperinflation of the lungs or superimposed congestive heart failure.
Symptoms described in adults with pneumococcal pneumonia may be noted in
FIG. 428.3 Radiographic findings characteristic of pneumococcal pneumonia in a 14 yr
old boy with cough and fever. Posteroanterior (A) and lateral (B) chest radiographs
reveal consolidation in the right lower lobe, strongly suggesting bacterial pneumonia.
Table 428.6
Factors Suggesting Need for Hospitalization of Children
With Pneumonia
Age <6 mo
Immunocompromised state
Toxic appearance
Moderate to severe respiratory distress
Hypoxemia (oxygen saturation <90% breathing room air, sea level)
Complicated pneumonia*
Sickle cell anemia with acute chest syndrome
Vomiting or inability to tolerate oral fluids or medications
Severe dehydration
No response to appropriate oral antibiotic therapy
Social factors (e.g., inability of caregivers to administer medications at home or follow-up appropriately)
* Pleural effusion, empyema, abscess, bronchopleural fistula, necrotizing pneumonia, acute
Treatment
Treatment of suspected bacterial pneumonia is based on the presumptive cause
and the age and clinical appearance of the child. For mildly ill children who do
not require hospitalization, amoxicillin is recommended. With the emergence of
penicillin-resistant pneumococci, high doses of amoxicillin (90 mg/kg/day orally
divided twice daily) should be prescribed unless local data indicate a low
prevalence of resistance (Table 428.7 ). Therapeutic alternatives include
cefuroxime and amoxicillin/clavulanate. For school-aged children and
adolescents or when infection with M. pneumoniae or C. pneumoniae is
suspected, a macrolide antibiotic is an appropriate choice for outpatient
management. Azithromycin is generally preferred, while clarithromycin or
doxycycline (for children 8 yr or older) are alternatives. For adolescents, a
respiratory fluoroquinolone (levofloxacin, moxifloxacin) may also be considered
as an alternative if there are contraindications to other agents.
Table 428.7
Table 428.8
Cystic Fibrosis
Marie E. Egan, Michael S. Schechter, Judith A. Voynow
Table 432.1
Complications of Cystic Fibrosis
RESPIRATORY
Bronchiectasis, bronchitis, bronchiolitis, pneumonia
Atelectasis
Hemoptysis
Pneumothorax
Nasal polyps
Sinusitis
Reactive airway disease
Mucoid impaction of the bronchi
Allergic bronchopulmonary aspergillosis
Cor pulmonale
Respiratory failure
GASTROINTESTINAL
Meconium ileus, meconium plug (neonate)
Meconium peritonitis (neonate)
Distal intestinal obstruction syndrome (non-neonatal obstruction)
Rectal prolapse
Intussusception
Volvulus
Fibrosing colonopathy (strictures)
Appendicitis
Intestinal atresia
Pancreatitis
Biliary cirrhosis (portal hypertension: esophageal varices, hypersplenism)
Neonatal obstructive jaundice
Hepatic steatosis
Gastroesophageal reflux
Cholelithiasis
Inguinal hernia
Growth failure (malabsorption)
Vitamin deficiency states (vitamins A, K, E, D)
Insulin deficiency, symptomatic hyperglycemia, diabetes
Malignancy (rare)
OTHER
Infertility
Delayed puberty
Edema-hypoproteinemia
Dehydration–heat exhaustion
Hypertrophic osteoarthropathy-arthritis
Clubbing
Amyloidosis
Diabetes mellitus
Aquagenic palmoplantar keratoderma (skin wrinkling)
Adapted from Silverman FN, Kuhn JP: Essentials of Caffey's pediatric x-ray diagnosis, Chicago,
1990, Year Book, p. 649.
Genetics
CF occurs most frequently in white populations of northern Europe, North
America, and Australia/New Zealand. The prevalence in these populations varies
but approximates 1 in 3,500 live births (1 in 9,200 individuals of Hispanic
descent and 1 in 15,000 African Americans). Although less frequent in African,
Hispanic, Middle Eastern, South Asian, and eastern Asian populations, the
disorder does exist in these populations as well (Fig. 432.1 ).
CFTR is the deletion of a single phenylalanine residue at amino acid 508
(F508del). This mutation is responsible for the high incidence of CF in northern
European populations and is considerably less frequent in other populations,
such as those of southern Europe and Israel. Nearly 50% of individuals with CF
in the United States Cystic Fibrosis Foundation (CFF) Patient Registry are
homozygous for F508del, and approximately 87% carry at least 1 F508del gene.
Remaining patients have an extensive array of mutations, none of which has a
prevalence of more than several percentage points, except in certain populations;
for example, the W1282X mutation occurs in 60% of Ashkenazi Jews with CF.
Through the use of probes for 40 of the most common mutations, the genotype
of 80–90% of Americans with CF can be ascertained. Genotyping using a
discreet panel of mutation probes is quick and less costly than more
comprehensive sequencing and is the approach typically used in state newborn
screening programs. In remaining patients, sequencing the entire CFTR gene and
looking for deletions and duplications are necessary to establish the genotype.
As sequencing technologies evolve and costs decrease, sequencing the entire
CFTR gene may become mainstream for all patients.
Table 432.2
One Proposed Classification of Cystic Fibrosis
Transmembrane Conductance Regulator (CFTR) Mutations
FUNCTIONAL
CLASS EFFECT ON CFTR CFTR SAMPLE MUTATIONS
PRESENT?
I Lack of protein production No Stop codons (designation in X; e.g., Trp1282X,
Gly542X); splicing defects with no protein
production (e.g., 711+1G→T, 1717-1G→A)
II Defect in protein trafficking withNo/substantially Phe508del, Asn1303Lys, Gly85Gly,
ubiquitination and degradation in reduced leu1065Pro, Asp507, Ser549Arg
endoplasmic reticulum/Golgi body
III Defective regulation; CFTR not No (nonfunction Gly551Asp, Ser492Phe, Val520Phe,
activated by adenosine triphosphate CFTR present in Arg553Gly, Arg560Thr, Arg560Ser
or cyclic adenosine monophosphate apical
membrane)
IV Reduced chloride transport through Yes Ala455Glu, Arg117Cys, Asp1152His,
CFTR at the apical membrane Leu227Arg, Arg334Trp, Arg117His*
V Splicing defect with reduced Yes 3849+10kbC→T, 1811+16kbA→G, IVS8-5T,
production of CFTR 2789+5G→A
* Function of Arg117His depends on the length of the polythymidine track on the same
chromosome in intron 8 (IVS8): 5T, 7T, or 9T. There is more normal CFTR function with a longer
polythymidine track.
Excessive loss of salt in the sweat predisposes young children to salt depletion
episodes, especially during episodes of gastroenteritis and during warm weather.
These children may present with hypochloremic alkalosis. Hyponatremia is a
risk particularly in warm climates. Frequently, parents notice salt frosting of the
skin or a salty taste when they kiss the child. A few genotypes are associated
with normal sweat chloride values.
Table 432.3
Sweat Testing
The sweat test, which involves using pilocarpine iontophoresis to collect sweat
and performing chemical analysis of its chloride content, is the standard
approach to diagnosis of CF. The procedure requires care and accuracy. An
electric current is used to carry pilocarpine into the skin of the forearm and
locally stimulate the sweat glands. If an adequate amount of sweat is collected,
the specimens are analyzed for chloride concentration. Infants with a positive
newborn screen for CF should have the sweat chloride testing performed after
36-wk corrected gestational age and at a weight greater than 2 kg and at age
greater than 10 days to increase the likelihood of sufficient sweat collection for
an accurate study. Positive results should be confirmed; for a negative result, the
test should be repeated if suspicion of the diagnosis remains.
More than 60 mmol/L of chloride in sweat is diagnostic of CF when one or
more other criteria are present. In individuals with a positive newborn screen, a
sweat chloride level less than 30 mmol/L indicates that CF is unlikely.
Borderline (or intermediate) values of 30-59 mmol/L have been reported in
patients of all ages who have CF with atypical involvement and require further
testing. Table 432.4 lists the conditions associated with false-negative and false-
positive sweat test results.
Table 432.4
Conditions Associated With False-Positive and False-
Negative Sweat Test Results
WITH FALSE-POSITIVE RESULTS
Eczema (atopic dermatitis)
Ectodermal dysplasia
Malnutrition/failure to thrive/deprivation
Anorexia nervosa
Congenital adrenal hyperplasia
Adrenal insufficiency
Glucose-6-phosphatase deficiency
Mauriac syndrome
Fucosidosis
Familial hypoparathyroidism
Hypothyroidism
Nephrogenic diabetes insipidus
Pseudohypoaldosteronism
Klinefelter syndrome
Familial cholestasis syndrome
Autonomic dysfunction
Prostaglandin E infusions
Munchausen syndrome by proxy
WITH FALSE-NEGATIVE RESULTS
Dilution
Malnutrition
Edema
Insufficient sweat quantity
Hyponatremia
Cystic fibrosis transmembrane conductance regulator mutations with preserved sweat duct function
DNA Testing
Several commercial laboratories test for 30-96 of the most common CFTR
mutations. This testing identifies ≥90% of individuals who carry 2 CF mutations.
Some children with typical CF manifestations are found to have 1 or no
detectable mutations by this methodology. Some laboratories perform
comprehensive mutation analysis screening for all the >1,900 identified
mutations.
Pancreatic Function
The diagnosis of pancreatic malabsorption can be made by the quantification of
elastase-1 activity in a fresh stool sample by an enzyme-linked immunosorbent
assay specific for human elastase. The quantification of fat malabsorption with a
72-hr stool collection is rarely necessary in the clinical setting. CF-related
diabetes affects approximately 20% of adolescents and 40–50% of adults, and
clinical guidelines recommend yearly oral glucose tolerance testing (OGTT)
after age 10. OGTT may sometimes be clinically indicated at an earlier age. Spot
testing of blood and urine glucose levels and glycosylated hemoglobin levels are
not sufficiently sensitive.
that is not available, a lower pharyngeal swab taken during or after a forced
cough is obtained for culture and antibiotic susceptibility studies. Because
irreversible loss of pulmonary function from low-grade infection can occur
gradually and without acute symptoms, emphasis is placed on a thorough
pulmonary history and physical exam and routine pulmonary function testing.
Table 432.5 lists symptoms and signs that suggest the need for more intensive
antibiotic and physical therapy (PT). Protection against exposure to methicillin-
resistant S. aureus, P. aeruginosa, B. cepacia, and other resistant Gram-negative
organisms is essential, including contact isolation procedures and careful
attention to cleaning of inhalation therapy equipment. A nurse, physical
therapist, respiratory therapist, social worker, and dietitian, as members of the
multidisciplinary care team, should evaluate children regularly and contribute to
the development of a comprehensive daily care plan. Considerable education and
programs to empower families and older children to take responsibility for care
are likely to result in the best adherence to daily care programs. Screening
patients and caregivers for anxiety and depression annually is expected to
identify issues that can interfere with adherence to daily care. Standardization of
practice, on the part of both caregivers and families, as well as close monitoring
and early intervention for new or increasing symptoms appears to result in the
best long-term outcomes.
Table 432.5
Symptoms and Signs Associated With Exacerbation of
Pulmonary Infection in Patients With Cystic Fibrosis
SYMPTOMS
Increased frequency and duration of cough
Increased sputum production
Change in appearance of sputum
Increased shortness of breath
Decreased exercise tolerance
Decreased appetite
Feeling of increased congestion in the chest
SIGNS
Increased respiratory rate
Use of accessory muscles for breathing
Intercostal retractions
Change in results of auscultatory examination of chest
Decline in measures of pulmonary function consistent with the presence of obstructive airway disease
Fever and leukocytosis
Weight loss
New infiltrate on chest radiograph
aspects of the patient's history and examination, including anorexia, weight loss,
and diminished activity, must be used to guide the frequency and duration of
therapy. Antibiotic treatment varies from intermittent short courses of 1
antibiotic to nearly continuous treatment with 1 or more antibiotics. Dosages for
some antibiotics are often 2-3 times the amount recommended for minor
infections because patients with CF have proportionately more lean body mass
and higher clearance rates for many antibiotics than other individuals. In
addition, it is difficult to achieve effective drug levels of many antimicrobials in
respiratory tract secretions.
Table 432.6
Antimicrobial Agents for Cystic Fibrosis Lung Infection
† Quantity of ticarcillin.
‡
Quantity of piperacillin.
§ In mg per dose.
Table 432.7
Cystic Fibrosis Transmembrane Regulator Modulators for
Cystic Fibrosis
DRUG FDA-APPROVED INDICATION FORMULATIONS USUAL DOSAGE
Ivacaftor ≥ 12 mo with a responsive mutation 150 mg tabs; 50, 75 mg ≥ 6 years: 150 mg q12
1 granule packets 2 hr 3
Lumacaftor/ivacaftor ≥ 2 yr, F508del-homozygous 100/125, 200/125 mg tabs; 6-11 yr: 200/250
100/125, 150/188 mg mg q12 hr
granule packets 2 ≥ 12 yr: 400/250
mg q12 hr 4
Tezacaftor/ivacaftor ≥ 12 yr, F508del-homozygous or 100/150 mg tabs co- ≥ 12 yr: 100/150 mg
F508del-heterozygous with another packaged with ivacaftor tab qAM, then 150 mg
responsive mutation 1 150 mg tabs ivacaftor qPM
1 Responsive mutations are those in which chloride transport is expected to increase to at least
10% of untreated normal over baseline with drug therapy, based on clinical or in vitro data.
2 The granules should be mixed with 5 mL of room-temperature or cold soft food or liquid and
consumed within 1 hr.
3 In patients 12 mo to 6 yr old, the recommended dosage is 50 mg every 12 hr for those weighing
<14 kg, and 75 mg every 12 hr for those weighing ≥ 14 kg.
4 In patients 2-5 yr old, the recommended dosage is 100/125 mg every 12 hr for those weighing
Other Therapies
Attempts to clear recalcitrant atelectasis and airway plugging with
bronchopulmonary lavage and direct installation of various medications are
sometimes used in exceptional cases; there is no evidence for sustained benefit
from repeated procedures. Expectorants such as iodides and guaifenesin do not
effectively assist with the removal of secretions from the respiratory tract.
Inspiratory muscle training can enhance maximum oxygen consumption during
exercise, as well as FEV1 .
Table 433.1
Clinical Manifestations of Primary Ciliary Dyskinesia
RESPIRATORY
• Unexplained respiratory distress in term neonate
• Daily productive (wet) cough since early infancy
• Daily, nonseasonal rhinosinusitis since early infancy
• Chronic otitis media and persistent middle ear effusions
• Digital clubbing (rare in children)
• Atypical asthma unresponsive to therapy
• Recurrent pneumonias
• Bronchiectasis
LEFT-RIGHT LATERALITY DEFECTS
• Situs inversus totalis
• Heterotaxy with or without complex congenital heart disease
MISCELLANEOUS
• Male infertility, immotile sperm
• Female subfertility, ectopic pregnancy
• Hydrocephalus (rare)
Table 433.2
New Consensus-Based Primary Ciliary Dyskinesia (PCD)
Diagnostic Criteria by Age
NEWBORNS (0–1 MO OF AGE)
Situs inversus totalis and unexplained neonatal respiratory distress (NRD) at term birth, plus at least one of the
following:
• Diagnostic ciliary ultrastructure on electron micrographs or 2 mutations in PCD-associated gene
CHILDREN (1 MO TO 5 YR)
2 or more major PCD clinical criteria (NRD,* daily wet cough, persistent nasal congestion, laterality defect), plus
at least one of the following (nasal nitric oxide not included in this age group because it is not yet sufficiently
tested):
• Diagnostic ciliary ultrastructure on electron micrographs
• 2 mutations in 1 PCD-associated gene
• Persistent and diagnostic ciliary waveform abnormalities on high-speed videomicroscopy, on multiple occasions
CHILDREN (5–18 YR OF AGE) AND ADULTS
2 or more PCD clinical criteria (NRD,* daily productive cough or bronchiectasis, persistent nasal congestion,
laterality defect), plus at least one of the following:
• Nasal nitric oxide during plateau <77 nL/min on 2 occasions, >2 mo apart (with CF excluded)
• Diagnostic ciliary ultrastructure on electron micrographs
• Two mutations in 1 PCD-associated gene
• Persistent and diagnostic ciliary waveform abnormalities on high-speed videomicroscopy, on multiple occasions
* In term neonates.
Table 433.3
Electron Microscopic Findings in Primary Ciliary
Dyskinesia
DYNEIN ARM DEFECTS
• Total or partial absence of outer dynein arms
• Total or partial absence of inner and outer dynein arms
• Total or partial absence of inner dynein arms alone (rare)
RADIAL SPOKE DEFECTS
• Total absence of radial spokes
• Absence of radial spoke heads
MICROTUBULAR TRANSPOSITION DEFECTS
• Inner dynein arm defect with microtubular derangement (in some axonemes)
• Absent central pair of tubules with outer doublet transposition
OTHER
• Central microtubular agenesis
• Ciliary aplasia or reduced cilia numbers
• Normal ultrastructure
Table 434.1
Comparison of Surfactant Dysfunction Disorders
SP-B TTF-1
SP-C DISEASE ABCA3 DEFICIENCY
DEFICIENCY DISORDERS
Gene name SFTPB SFTPC ABCA3 NKX2-1
Age of onset Birth Birth–adulthood Birth–childhood; rarely adult Birth–childhood
Inheritance Recessive Dominant/sporadic Recessive Sporadic/dominant
Mechanism Loss of function Gain of toxic Loss of function Loss of
function or function
dominant negative Gain of
function
Natural Lethal Variable Generally lethal, may be chronic Variable
history
DIAGNOSIS
Biochemical Absence of SP-B None None None
(tracheal and presence of
aspirate) incompletely
processed proSP-C
Genetic Sequence SFTPB Sequence SFTPC Sequence ABCA3 Sequence NKX2-
(DNA) 1; deletion
analysis
Ultrastructural Disorganized Not specific; may Small dense lamellar bodies with Variable
(lung biopsy– lamellar bodies have dense eccentrically placed dense cores
electron aggregates
microscopy)
Treatment Lung Supportive care, Lung transplantation or Supportive care;
transplantation or lung compassionate care for infants with treat coexisting
compassionate transplantation if biallelic null mutations; lung conditions
care progressing transplantation for other mutations (hypothyroidism)
if progressing
SP , Surfactant protein.
Pathology
Histopathologically, these disorders share a unique constellation of features,
including AEC2 hyperplasia, alveolar macrophage accumulation, interstitial
thickening and inflammation, and alveolar proteinosis. A number of different
Wambach J, Wegner D, DePass K, et al. Single ABCA3
mutations and risk for neonatal respiratory distress syndrome.
Pediatrics . 2012;130:e1575–e1582.
Wang Y, Kuan PJ, Xing C, et al. Genetic defects in surfactant
protein A2 are associated with pulmonary fibrosis and lung
cancer. Am J Hum Genet . 2009;84:52–59.
434.2
Pulmonary Alveolar Proteinosis
Jennifer A. Wambach, Lawrence M. Nogee, F. Sessions Cole III, Aaron Hamvas
Table 434.2
Comparison of Pulmonary Alveolar Proteinosis Syndromes
GM-CSF LYSINURIC
MARS GATA2
AUTOIMMUNE RECEPTOR PROTEIN
DEFICIENCY DEFICIENCY
DEFICIENCY INTOLERANCE
Gene(s) CSFR2A, CSFR2B SLC7A7 MARS GATA2
Age of onset Adult > child Childhood to adult Childhood Childhood to adult Childhood to adult
Inheritance N.A. Recessive Recessive Recessive Sporadic/dominant
Mechanism Neutralizing Loss of function Loss of function Loss of function Loss of function;
antibodies to haploinsufficiency
GM-CSF
Other Emesis; failure to Liver disease; Immune deficiency;
manifestations thrive hypothyroidism myelodysplasia
DIAGNOSIS
Biochemical Detection of Elevated serum Increased cationic None None
serum GM-CSF GM-CSF levels amino acids in urine,
autoantibody especially lysine
Genetic (DNA N.A. Sequence CSFR2A, Sequence SLC7A7 Sequence MARS Sequence GATA2
) CSFR2B
Treatment Whole lung Whole lung lavage; Whole lung lavage, Whole lung lavage Whole lung lavage;
lavage; inhaled bone marrow dietary protein bone marrow
GM-CSF transplantation restriction, transplantation
administration of
citrulline and nitrogen
scavenging drugs
GM-CSF , Granulocyte-macrophage colony-stimulating factor.
Table 435.1
Diffuse Alveolar Hemorrhage Syndromes
CLASSIFICATION SYNDROME
DISORDERS WITH PULMONARY CAPILLARITIS
Idiopathic (isolated) pulmonary capillaritis (ANCA positive or negative)
Granulomatosis with polyangiitis (Wegener granulomatosis)
Microscopic polyangiitis
Systemic lupus erythematosus (SLE)
Scleroderma
Polymyositis
Goodpasture syndrome
Antiphospholipid antibody syndrome
Henoch-Schönlein purpura
Immunoglobulin A nephropathy
Behçet syndrome
Cryoglobulinemia
Drug-induced capillaritis (phenytoin, retinoic acid, propylthiouracil)
Idiopathic pulmonary-renal syndrome
Acute lung transplant rejection
Eosinophilic granulomatosis angiitis (Churg-Strauss syndrome)
Idiopathic pulmonary fibrosis
Disorders without pulmonary capillaritis
Noncardiovascular causes Idiopathic pulmonary hemosiderosis
Heiner syndrome
Acute idiopathic pulmonary hemorrhage of infancy
Bone marrow transplantation
Immunodeficiency
Coagulation disorders
Hemolytic uremic syndromes
Celiac disease (Lane-Hamilton syndrome)
SLE
Non-accidental trauma
Radiation therapy
Infection (HIV, cryptococcosis, Legionnaires disease)
Drugs—toxins
Cardiovascular causes Mitral stenosis
Pulmonary venoocclusive disease
Arteriovenous malformations
Pulmonary lymphangioleiomyomatosis
Pulmonary hypertension
Pulmonary capillary hemangiomatosis
Chronic heart failure
Vascular thrombosis with infarction
Endocarditis
Modified from Susarla SC, Fan LL: Diffuse alveolar hemorrhage syndromes in children, Curr Opin
Pediatr 19:314–320, 2007.
Epidemiology
Disorders that present as DAH are highly variable in their severity, as well as in
their associated symptomatology and identifiable abnormalities in laboratory
testing; the diagnosis may be significantly delayed, making frequency estimates
unreliable. Similarly, the prevalence of IPH is largely unknown. Of the children
and young adults diagnosed with IPH in the past, it has been postulated that the
etiology of the hemorrhage might have been discovered if they had been studied
with the newer and more advanced diagnostics available today; specific
serologic testing has vastly improved our ability to appreciate immune mediated
disease. Estimates of prevalence obtained from Swedish and Japanese
retrospective case analyses vary from 0.24 to 1.23 cases per million. Children
and adolescents account for 30% of cases. The ratio of affected males:females is
1 : 1 in the childhood diagnosis group, and men are only slightly more affected
in the group diagnosed as young adults.
Pathology
In pulmonary capillaritis, key histologic features include (1) fibrin thrombi,
which occlude capillaries, (2) fibrin clots adherent to interalveolar septae, (3)
source of morbidity and mortality and may only be recognized on postmortem
examination. A high level of clinical suspicion and appropriate identification of
at-risk individuals is therefore recommended.
Table 436.1
Risk Factors for Pulmonary Embolism
ENVIRONMENTAL
Long-haul air travel (>4 hr)
Obesity
Cigarette smoking
Hypertension
Immobility
WOMEN'S HEALTH
Oral contraceptives, including progesterone-only and, especially, third-generation pills
Pregnancy or puerperium
Hormone replacement therapy
Septic abortion
MEDICAL ILLNESS
Personal or family history of prior pulmonary embolism or deep venous thrombosis
Cancer
Heart failure
Chronic obstructive pulmonary disease
Diabetes mellitus
Inflammatory bowel disease
Antipsychotic drug use
Long-term indwelling central venous catheter
Permanent pacemaker
Internal cardiac defibrillator
Stroke with limb paresis
Spinal cord injury
Nursing home confinement or current or repeated hospital admission
SURGICAL
Trauma
Orthopedic surgery
General surgery
Neurosurgery, especially craniotomy for brain tumor
Vascular anomalies
May-Turner syndrome
THROMBOPHILIA
Factor V Leiden mutation
Prothrombin gene (20210G A) mutation
Hyperhomocysteinemia (including mutation in methylenetetrahydrofolate reductase)
Antiphospholipid antibody syndrome
Deficiency of antithrombin III, protein C, or protein S
High concentrations of factor VIII or XI
Increased lipoprotein (a)
NONTHROMBOTIC
Air
Foreign particles (e.g., hair, talc, as a consequence of intravenous drug misuse)
Amniotic fluid
Bone fragments, bone marrow
Fat
Tumors (Wilms tumor)
Modified from Goldhaber SZ: Pulmonary embolism, Lancet 363:1295–1305, 2004.
Etiology
A number of risk factors may be identified in children and adolescents; the
presence of immobility, malignancy, pregnancy, infection, indwelling central
venous catheters, and a number of inherited and acquired thrombophilic
conditions have all been identified as placing an individual at risk. In children, a
significantly greater percentage of VTEs are risk associated as compared with
their adult counterparts. Children with deep venous thrombosis (DVT) and
pulmonary embolism (PE) are much more likely to have one or more
identifiable conditions or circumstances placing them at risk. In contrast to
adults, idiopathic thrombosis is a rare occurrence in the pediatric population. In a
retrospective cohort of patients with VTE in U.S. children's hospitals from 2001
to 2007, the majority (63%) of affected children were found to have one or more
chronic medical comorbidities. In a large Canadian registry, 96% of pediatric
patients were found to have one risk factor and 90% had two or more risk
factors. In contrast, approximately 60% of adults with this disorder have an
identifiable risk factor (see Table 436.1 ). The most common identified risk
factors in children include infection, congenital heart disease, and the presence
of an indwelling central venous catheter.
Embolic disease in children is varied in its origin. An embolus can contain
thrombus, air, amniotic fluid, septic material, or metastatic neoplastic tissue.
Thromboemboli are the type most commonly encountered. A commonly
encountered risk factor for DVT and PE in the pediatric population is the
presence of a central venous catheter. More than 50% of DVTs in children and
more than 80% in newborns are found in patients with indwelling central venous
lines. The presence of a catheter in a vessel lumen, as well as instilled
medications, can induce endothelial damage and favor thrombus formation.
Children with malignancies are also at considerable risk. Although PE has
been described in children with leukemia, the risk of PE is more significant in
children with solid rather than hematologic malignancies. A child with
malignancy may have numerous risk factors related to the primary disease
process and the therapeutic interventions. Infection from chronic
nephrotic syndrome (see Chapter 545 ) and antiphospholipid antibody syndrome.
From one-quarter to one-half of children with systemic lupus erythematosus (see
Chapter 183 ) have thromboembolic disease. There is a significant association
with VTE onset in children for each inherited thrombophilic trait evaluated,
thereby illuminating the importance of screening for thrombophilic conditions
for those at risk for VTE. Septic emboli are rare in children but may be caused
by osteomyelitis, jugular vein or umbilical thrombophlebitis, cellulitis, urinary
tract infection, and right-sided endocarditis.
Other risk factors include infection, cardiac disease, recent surgery, and
trauma. Surgical risk is thought to be more significant when immobility will be a
prominent feature of the recovery. Use of oral contraceptives confers additional
risk, although the level of risk in patients taking these medications appears to be
decreasing, perhaps because of the lower amounts of estrogen in current
formulations. In a previously healthy adolescent patient, the risk factors are often
unknown or are similar to adults (see Tables 436.1 and 436.2 ).
Table 436.2
Clinical Decision Rules for Deep Vein Thrombosis and
Pulmonary Embolism
WELLS' SCORE FOR DEEP VEIN THROMBOSIS *
Active cancer +1 NA
Paralysis, paresis, or recent plaster cast on lower extremities +1 NA
Recent immobilization >3 days or major surgery within the past 4 wk +1 NA
Localized tenderness of deep venous system +1 NA
Swelling of entire leg +1 NA
Calf swelling >3 cm compared to asymptomatic side +1 NA
Unilateral pitting edema +1 NA
Collateral superficial veins +1 NA
Previously documented deep vein thrombosis +1 NA
Alternative diagnosis at least as likely as deep vein thrombosis −2 NA
WELLS' SCORE FOR PULMONARY EMBOLISM † , ‡
Alternative diagnosis less likely than pulmonary embolism +3 +1
Clinical signs and symptoms of deep vein thrombosis +3 +1
Heart rate >100 beats/min +1⋅5 +1
Previous deep vein thrombosis or pulmonary embolism +1⋅5 +1
Immobilization or surgery within the past 4 wk +1⋅5 +1
Active cancer +1 +1
Hemoptysis +1 +1
REVISED GENEVA SCORE FOR PULMONARY EMBOLISM § , ||
Heart rate ≥95 beats/min +5 +2
Heart rate 75–94 beats/min +3 +1
Pain on lower-limb deep venous palpation and unilateral edema +4 +1
Unilateral lower-limb pain +3 +1
Previous deep vein thrombosis or pulmonary embolism +3 +1
Active cancer +2 +1
Hemoptysis +2 +1
Surgery or fracture within the past 4 wk +2 +1
Age >65 yr +1 +1
*
Classification for original Wells' score for deep vein thrombosis: deep vein thrombosis unlikely if
score ≤2; deep vein thrombosis likely if score >2.
†
Classification for original Wells' score for pulmonary embolism: pulmonary embolism unlikely if
score ≤4; pulmonary embolism likely if score >4.
‡
Classification for simplified Wells' score for pulmonary embolism: pulmonary embolism unlikely if
score ≤1; pulmonary embolism likely if score >1.
§ Classification for original revised Geneva score for pulmonary embolism: non-high probability of
pulmonary embolism if score ≤10; high probability of pulmonary embolism if score >10.
|| Classification for simplified revised Geneva score for pulmonary embolism: non-high probability
of pulmonary embolism if score ≤4; high probability of pulmonary embolism if score >4.
From Di Nisio M, van Es N, Büller HR: Deep vein thrombosis and pulmonary embolism, Lancet
388:3060–3069, 2016 (Table 1, p. 3062).
Epidemiology
A retrospective cohort study was performed with patients younger than 18 yr of
age, discharged from 35 to 40 children's hospitals across the United States from
2001 to 2007. During this time, a dramatic increase was noted in the incidence of
VTE; the annual rate of VTE increased by 70% from 34 to 58 cases per 10,000
hospital admissions. Although this increased incidence was noted in all age
groups, a bimodal distribution of patient ages was found, consistent with prior
studies; infants younger than 1 yr of age and adolescents made up the majority of
admissions with VTE, but neonates continue to be at greatest risk. The peak
incidence for VTE in childhood appears to occur in the 1st mo of life. It is in this
neonatal period that thromboembolic events are more problematic, likely as a
result of an imbalance between procoagulant factors and fibrinolysis. The yearly
incidence of venous events was estimated at 5.3/10,000 hospital admissions in
children and 24/10,000 in the neonatal intensive care.
Pediatric autopsy reviews have estimated the incidence of thromboembolic
disease in children as between 1% and 4%, although not all were clinically
significant. Thromboembolic pulmonary disease is often unrecognized, and
antemortem studies may underestimate the true incidence. Pediatric deaths from
etc.) may become an alternate therapy for both acute PE and long-term treatment
(Table 436.3 ).
Table 436.3
Anticoagulant Therapies for Deep Vein Thrombosis and
Pulmonary Embolism
‡ Apixaban, edoxaban, and rivaroxaban are contraindicated in patients with a creatinine clearance
below 15 mL/min.
§ The recommended edoxaban dose is 30 mg once a day for patients with a creatinine clearance
of 30–50 mL/min, a bodyweight less than or equal to 60 kg, or for those on certain strong P-
glycoprotein inhibitors.
Medication dosing may vary with regard to primary diagnosis, age, comorbidities, and other
436.2
Pulmonary Hemorrhage and
Hemoptysis
Mary A. Nevin
Etiology
Table 436.4 and Table 435.1 (in Chapter 435 ) present conditions that can
manifest as pulmonary hemorrhage or hemoptysis in children. The chronic
(opposed to an acute) presence of a foreign body can lead to inflammation
and/or infection, thereby inducing hemorrhage. Bleeding is more likely to occur
in association with a chronically retained foreign body of vegetable origin.
Table 436.4
Etiology of Pulmonary Hemorrhage (Hemoptysis)
FOCAL HEMORRHAGE
Bronchitis and bronchiectasis (especially cystic fibrosis–related)
Infection (acute or chronic), pneumonia, abscess
Tuberculosis
Trauma
Pulmonary arteriovenous malformation (with or without hereditary hemorrhagic telangiectasia)
Foreign body (chronic)
Neoplasm including hemangioma
Pulmonary embolus with or without infarction
Bronchogenic cysts
DIFFUSE HEMORRHAGE
Idiopathic of infancy
Congenital heart disease (including pulmonary hypertension, venoocclusive disease, congestive heart failure)
Prematurity
Cow's milk hyperreactivity (Heiner syndrome)
Goodpasture syndrome
Collagen vascular diseases (systemic lupus erythematosus, rheumatoid arthritis)
Henoch-Schönlein purpura and vasculitic disorders
Granulomatous disease (granulomatosis with polyangiitis)
Celiac disease
Coagulopathy (congenital or acquired)
Malignancy
Immunodeficiency
Exogenous toxins, especially inhaled
Hyperammonemia
Pulmonary hypertension
Pulmonary alveolar proteinosis
Idiopathic pulmonary hemosiderosis
Tuberous sclerosis
Lymphangiomyomatosis or lymphangioleiomyomatosis
Physical injury or abuse
Catamenial
See also Table 435.1 .
Atelectasis
Ranna A. Rozenfeld
Pathophysiology
The causes of atelectasis can be divided into 5 groups (Table 437.1 ). Respiratory
syncytial virus (see Chapter 287 ) and other viral infections, including influenza
viruses in young children can cause multiple areas of atelectasis. Mucous plugs
are a common predisposing factor to atelectasis. Massive collapse of one or both
lungs is most often a postoperative complication but occasionally results from
other causes, such as trauma, asthma, pneumonia, tension pneumothorax (see
Chapter 439 ), aspiration of foreign material (see Chapters 414 and 425 ),
paralysis, or after extubation. Massive atelectasis is usually produced by a
combination of factors, including immobilization or decreased use of the
diaphragm and the respiratory muscles, obstruction of the bronchial tree, and
abolition of the cough reflex.
Table 437.1
Clinical Manifestations
Symptoms vary with the cause and extent of the atelectasis. A small area is
likely to be asymptomatic. When a large area of previously normal lung becomes
atelectatic, especially when it does so suddenly, dyspnea accompanied by rapid
shallow respirations, tachycardia, cough, and often cyanosis occurs. If the
obstruction is removed, the symptoms disappear rapidly. Although it was once
believed that atelectasis alone can cause fever, studies have shown no association
between atelectasis and fever. Physical findings include limitation of chest
excursion, decreased breath sound intensity, and coarse crackles. Breath sounds
are decreased or absent over extensive atelectatic areas.
Massive pulmonary atelectasis usually presents with dyspnea, cyanosis, and
tachycardia. An affected child is extremely anxious and, if old enough,
complains of chest pain. The chest appears flat on the affected side, where
decreased respiratory excursion, dullness to percussion, and feeble or absent
breath sounds are also noted. Postoperative atelectasis usually manifests within
24 hr of operation but may not occur for several days.
Acute lobar collapse is a frequent occurrence in patients receiving intensive
care. If undetected, it can lead to impaired gas exchange, secondary infection,
and subsequent pulmonary fibrosis. Initially, hypoxemia may result from
ventilation-perfusion mismatch. In contrast to atelectasis in adult patients, in
whom the lower lobes and, in particular, the left lower lobe are most often
involved, 90% of cases in children involve the upper lobes and 63% involve the
right upper lobe. There is also a high incidence of upper lobe atelectasis and,
especially, right upper lobe collapse in patients with atelectasis being treated in
neonatal intensive care units. This high incidence may be a result of movement
of the endotracheal tube into the right mainstem bronchus, where it obstructs or
causes inflammation of the bronchus to the right upper lobe.
bronchoscopic removal of a mucous plug from the right mainstem
bronchus.
Treatment
Treatment depends on the cause of the collapse (Table 437.2 ). If effusion or
pneumothorax is responsible, the external compression must first be removed.
Often vigorous efforts at cough, deep breathing, and percussion will facilitate
expansion. Aspiration with sterile tracheal catheters may facilitate removal of
mucous plugs. Continuous positive airway pressure may improve atelectasis.
Table 437.2
Pneumothorax
Glenna B. Winnie, Suraiya K. Haider, Aarthi P. Vemana, Steven V. Lossef
Table 439.1
Causes of Pneumothorax in Children
SPONTANEOUS
Primary Idiopathic (no underlying lung disease)
Spontaneous rupture of subpleural blebs
Secondary (underlying lung disease)
Congenital lung disease
• Congenital cystic adenomatoid malformation
• Bronchogenic cysts
• Pulmonary hypoplasia
• Birt-Hogg-Dube syndrome
Conditions associated with increased intrathoracic pressure
• Asthma
• Bronchiolitis
• Cystic fibrosis
• Airway foreign body
• Smoking (cigarettes, marijuana, crack cocaine)
Infection
• Tuberculosis
• Pneumocystis carinii (jirovecii)
• Echinococcosis
• Pneumatocele
• Lung abscess
• Bronchopleural fistula
Lung disease
• Langerhans cell histiocytosis
• Tuberous sclerosis
• Marfan syndrome
• Ehlers-Danlos syndrome
• Pulmonary fibrosis
• Sarcoidosis
• Rheumatoid arthritis, scleroderma, ankylosing spondylitis
• Metastatic neoplasm—usually osteosarcoma (rare)
• Pulmonary blastoma
TRAUMATIC
Non-iatrogenic
• Penetrating trauma
• Blunt trauma
Iatrogenic
• Thoracotomy
• Thoracoscopy, thoracentesis
• Tracheostomy
• Tube or needle puncture
• Mechanical ventilation
• High-flow therapy (moved from non-iatrogenic)
Adapted from Noppen M. Spontaneous pneumothorax:epidemiology, pathophysiology and cause.
Eur Respir Rev 19:117, 217–219, 2010 (Table 1, 2, p 218).
Bronchopulmonary Dysplasia
Sharon A. McGrath-Morrow, J. Michael Collaco
Bronchopulmonary Dysplasia
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of infancy and
childhood that occurs primarily in preterm infants born at less than 32 wk
gestation. BPD is characterized by alveolar hypoplasia, often with concomitant
small airway dysfunction and impaired pulmonary vascular growth. Contributing
factors to the development of BPD may include early gestational age, low birth
weight, lung barotrauma, exposure to hyperoxia, lung inflammation, and pre-
and postnatal infections, as well as potential modifier genes and epigenetic
factors. The currently accepted definition includes an oxygen requirement for 28
days postnatally, and the disorder is graded as mild, moderate, or severe on the
basis of supplemental oxygen and ventilation requirements at specific timepoints
(Table 444.1 ). For initial inpatient presentation and management, see Chapter
122 .
Table 444.1
Definitions of Bronchopulmonary Dysplasia
ADDITIONAL ADDITIONAL
FEATURES ADDITIONAL FEATURES OF
FEATURES OF MILD FEATURES OF
OF ALL BPD SEVERE BPD
BPD MODERATE BPD
<32 wk Breathing room air at 36 <30% supplemental oxygen >30% supplemental oxygen and/or
gestational wk PMA or at at 36 wk PMA or at positive pressure ventilation at 36 wk
age at birthdischarge, whichever discharge, whichever PMA or at discharge, whichever comes
Oxygen comes first comes first first
requirement
for at least
28 days
>32 wk Breathing room air at 56 <30% supplemental oxygen >30% supplemental oxygen and/or
gestational days of life or at at 56 days of life or at positive pressure ventilation at 56 days
age at birth discharge, whichever discharge, whichever of life or at discharge, whichever comes
Oxygen comes first comes first first
requirement
for at least
28 days
BPD , bronchopulmonary dysplasia; PMA , postmenstrual age.
Clinical Manifestations
Physical findings of the pulmonary exam vary with the severity of disease and
with respiratory illnesses. Although some patients may appear to be comfortably
breathing when well, they can experience significant deterioration when ill or
with periods of stress due to decreased pulmonary reserve secondary to alveolar
hypoplasia and small airway disease. Children with BPD may exhibit tachypnea,
head bobbing, and retractions when ill or at baseline depending on the severity
of disease. Although breath sounds may be clear, many patients have baseline
wheeze or coarse crackles. A persistent fixed wheeze or stridor suggests
subglottic stenosis (see Chapter 415 ) or large airway malacia. Fine crackles may
be present in patients prone to fluid overload. Chest radiographs may
demonstrate air trapping, focal atelectasis, interstitial changes, and/or
peribronchial thickening.
The most severely affected patients may require respiratory support to achieve
adequate gas exchange. Supplemental oxygen may be required to maintain
acceptable oxygen saturations and often is needed to minimize the work of
breathing. Chronic respiratory insufficiency may be evidenced as elevation of
serum bicarbonate, elevated carbon dioxide on blood gas analysis, hypoxemia,
or polycythemia; the most severe cases may require tracheostomy and
ventilation to achieve long-term respiratory stability. Patients must be monitored
for the development of pulmonary hypertension, especially if they require
supplemental oxygen and have chronic respiratory insufficiency.
Aspiration from dysphagia and/or gastroesophageal reflux (GERD) (see
Chapter 349 ) can compromise pulmonary status. The risk of aspiration may
increase during periods of illness due to worsening tachypnea and air trapping.
Other comorbidities resulting from premature birth that complicate the
management of BPD include fixed and functional upper airway obstruction,
CNS injuries leading to abnormal control of breathing, abnormal airway tone,
increased aspiration risk, gastrointestinal dysmotility, systemic hypertension, and
noninvasive long-term ventilation. The conditions leading to the need for home
ventilation are diverse. Most literature focuses on single-center experience, but
broad themes emerge. About two-thirds of children have a primary neurologic
indication, including neuromuscular weakness or abnormal ventilatory control,
and about one-third have chronic lung disease (Table 446.1 ).
Table 446.1
Indications for Long-Term Mechanical Ventilation
PULMONARY/ALVEOLAR
BRONCHOPULMONARY DYSPLASIA (BPD)
PARDS (Severe acquired lung disease, such as after pediatric acute respiratory distress syndrome)
Pulmonary fibrosis syndromes
AIRWAY
Severe tracheomalacia
Severe bronchomalacia
Obstructive sleep apnea
Storage diseases
CHEST (SEE CHAPTER 445 )
Kyphoscoliosis
Skeletal dysplasias
Obesity
NEUROMUSCULAR
Spinal muscular atrophy
Spinal cord injury
Diaphragmatic dysfunction
Mitochondrial diseases
CNS
Congenital central hypoventilation syndrome (CCHS)
Rapid-onset obesity with hypothalamic dysregulation, hypoventilation, and autonomic dysfunction (ROHHAD)
Severe ischemic brain injury
Myelomeningocele with Arnold-Chiari type II malformation
Acquired hypoventilation syndromes
Keywords
congenital central hypoventilation syndrome
Hirschsprung disease
neuroblastoma neural crest tumors
ganglioneuroblastoma
ganglioneuroma
diaphragm pacing
autonomic dysregulation
control of breathing
Table 446.2
Congenital Central Hypoventilation Syndrome-Related
Symptoms
THE SYMPTOMS EMERGE FROM DIFFERENT ORGAN SYSTEMS AND COULD BE
OVERLOOKED BY THE CLINICIANS
Respiratory symptoms Nocturnal hypoventilation and possible daytime hypoventilation
Ability to hold breath for a long period of time and absence of air hunger
afterwards
Cardiovascular Arrhythmias
symptoms Reduced heart rate variability
Vasovagal syncope
Syncope
Cold extremities
Postural hypotension
Neurologic symptoms Developmental delay
Seizures (primarily during infancy)
Motor and speech delay
Learning disabilities
Altered perception of pain
Gastrointestinal Hirschsprung disease-related symptoms: dysphagia, constipation, and gastroesophageal
symptoms reflux
Ophthalmologic Nonreactive/sluggish pupils
symptoms Altered lacrimation and near response
Anisocoria, miosis, and ptosis
Strabismus
Temperature instability Altered perspiring
Absence of fever with infections
Malignancies Tumors of neural crest origin
Psychological Decreased anxiety
From Lijubić K, Fister I Jr, Fister I: Congenital central hypoventilation syndrome: a comprehensive
review and future challenges, J Respir Med 856149:1–8, 2014 (Table 1, p. 3).
Genetics
The paired-like homeobox 2B (PHOX2B) gene is the disease-defining gene for
CCHS. PHOX2B encodes a highly conserved homeodomain transcription factor,
is essential to the embryologic development of the ANS from the neural crest,
and is expressed in key regions and systems that explain much of the CCHS
phenotype. Individuals with CCHS are heterozygous for either a polyalanine
repeat expansion mutation (PARM) in exon 3 of the PHOX2B gene (normal
number of alanines is 20 with normal genotype 20/20), such that individuals
with CCHS have 24-33 alanines on the affected allele (genotype range is 20/24-
20/33), or a non-polyalanine repeat expansion mutation (NPARM) resulting from
a missense, nonsense, frameshift, stop codon, or splice site mutation.
hypercarbia without respiratory distress during sleep will quickly lead the
clinician to consider the diagnosis of CCHS or LO-CCHS.
Table 446.3
Differential Diagnoses of Congenital Central
Hypoventilation Syndrome
METABOLIC
Mitochondrial defects, e.g., Leigh disease
Pyruvate dehydrogenase deficiency
Hypothyroidism
NEUROLOGIC
Structural central nervous system abnormalities, e.g., Arnold Chiari malformation, Moebius syndrome
Vascular injury, e.g., central nervous system (CNS) hemorrhage, infarct
Trauma
Tumor
PULMONARY
Primary lung disease
Respiratory muscle weakness, e.g., diaphragm paralysis, congenital myopathy
GENETIC
Prader Willi syndrome
Familial dysautonomia
SEDATIVE DRUGS
OTHER
Rapid-onset obesity, hypothalamic dysregulation hypoventilation, autonomic dysregulation (ROHHAD)
Modified from Healy F, Marcus CL: Congenital central hypoventilation syndrome in children,
Pediatr Respir Rev 12:253–263, 2011 (Table 1, p. 258).
Management
Supported Ventilation—Diaphragm Pacing
Depending on the severity of the hypoventilation, the individual with CCHS can
have various options for artificial ventilation: positive pressure ventilation
(noninvasive via mask or via tracheostomy) or negative pressure ventilation
(pneumosuit, chest cuirass, or diaphragm pacing). Chronic mechanical
ventilation is addressed in Chapters 446.1 and 446.4 . Diaphragm pacing offers
another mode of supported ventilation, involving bilateral surgical implantation
of electrodes beneath the phrenic nerves, with connecting wires to
subcutaneously implanted receivers. The external transmitter, which is much
smaller and lighter in weight than a ventilator, sends a signal to flat donut-shaped
antennae that are placed on the skin over the subcutaneously implanted
Takotsubo cardiomyopathy (primary or secondary)
Idiopathic (Common)
Anxiety, hyperventilation
Panic disorder
Table 449.2
Congenital Malformation Syndromes Associated With
Congenital Heart Disease
SYNDROME FEATURES
CHROMOSOMAL DISORDERS
Trisomy 21 (Down syndrome) Endocardial cushion defect, VSD, ASD
Trisomy 21p (cat-eye syndrome) Miscellaneous, total anomalous pulmonary venous return
Trisomy 18 VSD, ASD, PDA, TOF, coarctation of aorta, bicuspid aortic or
pulmonary valve
Trisomy 13 VSD, ASD, PDA, coarctation of aorta, bicuspid aortic or
pulmonary valve
Trisomy 9 Miscellaneous, VSD
XXXXY PDA, ASD
Penta X PDA, VSD
Triploidy VSD, ASD, PDA
XO (Turner syndrome) Bicuspid aortic valve, coarctation of aorta
Fragile X Mitral valve prolapse, aortic root dilatation
Duplication 3q2 Miscellaneous
Deletion 4p VSD, PDA, aortic stenosis
Deletion 9p Miscellaneous
Deletion 5p (cri du chat syndrome) VSD, PDA, ASD, TOF
Deletion 10q VSD, TOF, conotruncal lesions*
Deletion 13q VSD
Deletion 18q VSD
Deletion 1p36 ASD, VSD, PDA, TOF, cardiomyopathy
Deletion/duplication 1q21.1 ASD, VSD, PS
Deletion 17q11 (William syndrome) Supravalvar AS, branch PS
Deletion 11q 24-25 (Jacobsen syndrome) VSD, left sided lesions
SYNDROME COMPLEXES
CHARGE association (c oloboma, h eart, a VSD, ASD, PDA, TOF, endocardial cushion defect
tresia choanae, r etardation, g enital, and e ar
anomalies)
DiGeorge sequence, CATCH 22 (c ardiac Aortic arch anomalies, conotruncal anomalies
defects, a bnormal facies, t hymic aplasia, c left
palate, h ypocalcemia, and deletion 22q11)
Alagille syndrome (arteriohepatic dysplasia) Peripheral pulmonic stenosis, PS, TOF
VATER association (v ertebral, a nal, t racheoe VSD, TOF, ASD, PDA
sophageal, r adial, and r enal anomalies)
FAVS (f acioa uriculov ertebral s pectrum) TOF, VSD
CHILD (c ongenital h emidysplasia with i Miscellaneous
chthyosiform erythroderma, l imb d efects)
Mulibrey nanism (muscle, liver, brain, eye) Pericardial thickening, constrictive pericarditis
Asplenia syndrome Complex cyanotic heart lesions with decreased pulmonary blood
flow, transposition of great arteries, anomalous pulmonary
venous return, dextrocardia, single ventricle, single
atrioventricular valve
Polysplenia syndrome Acyanotic lesions with increased pulmonary blood flow, azygos
continuation of inferior vena cava, partial anomalous pulmonary
venous return, dextrocardia, single ventricle, common
atrioventricular valve
PHACE syndrome (p osterior brain fossa VSD, PDA, coarctation of aorta, arterial aneurysms
anomalies, facial h emangiomas, a rterial
anomalies, c ardiac anomalies and aortic
coarctation, e ye anomalies)
TERATOGENIC AGENTS
Congenital rubella PDA, peripheral pulmonic stenosis
Fetal hydantoin syndrome VSD, ASD, coarctation of aorta, PDA
Fetal alcohol syndrome ASD, VSD
Fetal valproate effects Coarctation of aorta, hypoplastic left side of heart, aortic
stenosis, pulmonary atresia, VSD
Maternal phenylketonuria VSD, ASD, PDA, coarctation of aorta
Retinoic acid embryopathy Conotruncal anomalies
OTHERS
Apert syndrome VSD
Autosomal dominant polycystic kidney disease Mitral valve prolapse
Carpenter syndrome PDA
Conradi syndrome VSD, PDA
Crouzon disease PDA, coarctation of aorta
Cutis laxa Pulmonary hypertension, pulmonic stenosis
De Lange syndrome VSD
Ellis–van Creveld syndrome Single atrium, VSD
Holt-Oram syndrome ASD, VSD, 1st-degree heart block
Infant of diabetic mother Hypertrophic cardiomyopathy, VSD, conotruncal anomalies
Kartagener syndrome Dextrocardia
Meckel-Gruber syndrome ASD, VSD
Noonan syndrome Pulmonic stenosis, ASD, cardiomyopathy
Pallister-Hall syndrome Endocardial cushion defect
Primary ciliary dyskinesia Heterotaxia disorders
Rubinstein-Taybi syndrome VSD
Scimitar syndrome Hypoplasia of right lung, anomalous pulmonary venous return to
inferior vena cava
Smith-Lemli-Opitz syndrome VSD, PDA
TAR syndrome (thrombocytopenia and absent ASD, TOF
radius)
Treacher Collins syndrome VSD, ASD, PDA
* Conotruncal includes TOF, pulmonary atresia, truncus arteriosus, and transposition of great
arteries.
ASD, Atrial septal defect; AV, aortic valve; PDA, patent ductus arteriosus; PS, pulmonary stenosis;
TOF, tetralogy of Fallot; VSD, ventricular septal defect.
Table 449.3
Cardiac Manifestations of Systemic Diseases
A careful family history may also reveal early (at age <50 yr) coronary artery
disease or stroke (suggestive of familial hypercholesterolemia or thrombophilia),
sudden death (suggestive of cardiomyopathy or familial arrhythmic disorder),
generalized muscle disease (suggestive of one of the muscular dystrophies,
dermatomyositis, or familial or metabolic cardiomyopathy), or first-degree
relatives with congenital heart disease.
Table 449.4
Pulse Rates at Rest
LOWER LIMITS OF NORMAL AVERAGE UPPER LIMITS OF NORMAL
AGE
(beats/min) (beats/min) (beats/min)
Newborn 70 125 190
1–11 mo 80 120 160
2 yr 80 110 130
4 yr 80 100 120
6 yr 75 100 115
8 yr 70 90 110
10 yr 70 90 110
GIRLS BOYS GIRLS BOYS GIRLS BOYS
12 yr 70 65 90 85 110 105
14 yr 65 60 85 80 105 100
16 yr 60 55 80 75 100 95
18 yr 55 50 75 70 95 90
Prevalence
Congenital heart disease (CHD) occurs in approximately 0.8% of live births. The
incidence is higher in stillborns (3–4%), spontaneous abortuses (10–25%), and
premature infants (approximately 2% excluding patent ductus arteriosus [PDA]).
This overall incidence does not include mitral valve prolapse, PDA of preterm
infants, and bicuspid aortic valves (present in 1–2% of adults). Congenital
cardiac defects have a wide spectrum of severity in infants: approximately 2-3 in
1,000 newborn infants will be symptomatic with heart disease in the 1st yr of
life. The diagnosis is established by 1 wk of age in 40–50% of patients with
CHD and by 1 mo of age in 50–60%. With advances in both corrective and
palliative surgery, the number of children with CHD surviving to adulthood has
increased dramatically. Despite these advances, CHD remains the leading cause
of death in children with congenital malformations. Table 451.1 summarizes the
relative frequency of the most common congenital cardiac lesions.
Table 451.1
Most congenital defects are well tolerated in the fetus because of the parallel
nature of the fetal circulation. Even the most severe cardiac defects, such as
hypoplastic left heart syndrome (HLHS) , can usually be well compensated
for by the fetal circulation. In HLHS the entire fetal cardiac output would be
ejected by the right ventricle via the ductus arteriosus into both the descending
and ascending aortae (the latter filling in a retrograde fashion), so that fetal organ
blood flow would be minimally perturbed. Because the placenta provides for gas
exchange and the normal fetal circulation has mixing between more highly and
more poorly oxygenated blood, fetal organ oxygen delivery is also not
dramatically affected. It is only after birth when the fetal pathways (ductus
arteriosus and foramen ovale) begin to close that the full hemodynamic impact
of an anatomic abnormality becomes apparent. One notable exception is the case
of severe regurgitant lesions, most frequently of the tricuspid valve. In these
lesions, such as Ebstein anomaly of the tricuspid valve or severe right
ventricular outflow obstruction (see Chapter 457.7 ), the parallel fetal circulation
cannot compensate for the volume load imposed on the right side of the heart. In
utero heart failure, often with fetal pleural and pericardial effusions, and
generalized ascites (nonimmune hydrops fetalis) may occur.
Although the most significant transitions in circulation occur in the immediate
perinatal period, the circulation continues to undergo changes after birth, and
these later changes may also have a hemodynamic impact on cardiac lesions and
their apparent incidence. As pulmonary vascular resistance (PVR) falls in the 1st
several wk of life, left-to-right shunting through intracardiac defects increases
and symptoms become more apparent. Thus, in patients with a ventricular
septal defect (VSD) , heart failure is often first noticed between 1 and 3 mo of
age (see Chapter 453.6 ). The severity of various defects can also change
dramatically with growth; some VSDs may become smaller and even close as
the child ages. Alternatively, stenosis of the aortic or pulmonary valve, which
syndrome or the Shprintzen (velocardiofacial) syndrome. The acronym
CATCH 22 has been used to summarize the major components of these
syndromes: cardiac defects, abnormal facies, thymic aplasia, cleft palate, and
hypocalcemia. The specific cardiac anomalies are conotruncal defects (tetralogy
of Fallot, truncus arteriosus, double-outlet right ventricle, subarterial VSD) and
branchial arch defects (coarctation of the aorta, interrupted aortic arch, right
aortic arch). Congenital airway anomalies such as tracheomalacia and
bronchomalacia are sometimes present. Although the risk of recurrence is
extremely low in the absence of a parental 22q11.2 deletion, it is 50% if 1 parent
carries the deletion. More than 90% of patients with the clinical features of
DiGeorge syndrome have a deletion at 22q11.2. A 2nd genetic locus on the short
arm of chromosome 10 (10p13p14) has also been identified, the deletion of
which shares some, but not all, phenotypic characteristics with the 22q11.2
deletion; patients with del(10p) have an increased incidence of sensorineural
hearing loss.
Other structural heart lesions associated with specific chromosomal
abnormalities include familial secundum atrial septal defect (ASD) associated
with heart block (the transcription factor Nkx2.5 on chromosome 5q35),
familial ASD without heart block (the transcription factor GATA4), Alagille
syndrome (Jagged1 on chromosome 20p12), and Williams syndrome (elastin
on chromosome 7q11). Of interest, patients with VSDs and atrioventricular
septal defects have been found to have multiple Nkx2.5 mutations in cells
isolated from diseased heart tissues, but not from normal heart tissues or from
circulating lymphocytes, indicating a potential role for somatic mutations
leading to mosaicism in the pathogenesis of congenital heart defects. Tables
451.2 and 451.3 are a compilation of known genetic causes of CHD.
Table 451.2
Genetics of Congenital Heart Disease: Defects Associated
With Syndromes
COMMON
CARDIOVASCULAR CHROMOSOMAL
GENE(S) IMPLICATED* CARDIAC
DISEASE LOCATION
DEFECTS
DiGeorge syndrome, 22q11.2, 11p13p14 TBX1 TOF, IAA, TA,
velocardiofacial syndrome VSD
Familial ASD with heart 5q35 NKX2.5 ASD, heart block
block
Familial ASD without 8p22-23 GATA4 ASD
heart block
Alagille syndrome (bile 20p12, 1p12 JAGGED1, NOTCH2 Peripheral
duct hypoplasia, right- pulmonary
sided cardiac lesions) hypoplasia, PS,
TOF
Holt-Oram syndrome 12q24 TBX5 ASD, VSD, PDA
(limb defects, ASD)
Trisomy 21 (Down 21q22 Not known AVSD
syndrome)
Isolated familial AV septal 1p31-p21, 3p25 CRELD1 AVSD
defect (without trisomy
21)
Familial TAPVR 4p13-q12 Not known TAPVR
Noonan syndrome (PS, 12q24, 12p1.21, 2p212, 3p25.2, PTPN11, KRAS, SOS1, SOS2, PS, ASD, VSD,
ASD, hypertrophic 7q34, 15q22.31, 11p15.5, 1p13.2, RAF1, BRAF, MEK1, HRAS, PDA,
cardiomyopathy) 10q25.2, 11q23.3,17q11.2 NRAS, SHOC2, CBL, NF1 cardiomyopathy
Ellis–van Creveld 4p16 EVC, EVC2 ASD, common
syndrome (polydactyly, atrium
ASD)
Char syndrome 6p12-21.1 TFAP2B PDA
(craniofacial, limb defects,
PDA)
Williams-Beuren 7q11.23 ELN (Elastin) Supravalvular
syndrome (supravalvular AS, peripheral
AS, branch PS, PS
hypercalcemia)
Marfan syndrome 15q21 Fibrillin Aortic aneurysm,
(connective tissue mitral valve
weakness, aortic root disease
dilation)
Familial laterality Xq24-2q7, 1q42, 9p13-21 ZIC3, DNAI1 Situs inversus,
abnormalities complex
congenital heart
disease
Turner syndrome X Not known Coarctation of
the aorta, aortic
stenosis
Trisomy 13 (Patau 13 Not known ASD, VSD,
syndrome) PDA, valve
abnormalities
Trisomy 18 (Edwards 18 Not known ASD, VSD,
syndrome) PDA, valve
abnormalities
Cri du chat syndrome 5p15.2 CTNND2 ASD, VSD,
PDA, TOF
Cat-eye syndrome 22q11 Not known TAPVR, TOF
Jacobsen 11q23 JAM3 HLHS
Costello 11p15.5 HRAS PS, hypertrophic
cardiomyopathy,
arrhythmias
CHARGE 8p12, 7q21.11 CHD7, SEMA3E ASD, VSD, TOF
Kabuki syndrome 12q13.12 MLL2 ASD, VSD,
TOF, coarctation,
TGA
Carney syndrome 2p16 PRKAR1A Atrial and
ventricular
myxomas
*
In many cases, mutation of a single gene has been closely linked to a specific cardiovascular
disease, for example, by finding a high incidence of mutations or deletions of that gene in a large
group of patients. These findings are often confirmed by studies in mice in which deletion or
alteration of the gene induces a similar cardiac phenotype to the human disease. In others,
mutation of a gene may increase the risk of cardiovascular disease, but with decreased
penetrance, suggesting that modifier genes or environmental factors play a role. Finally, in some
cases, gene mutations have only been identified in a small number of pedigrees, and confirmation
awaits screening of larger numbers of patients.
AS, aortic stenosis; ASD, atrial septal defect; AV, atrioventricular; AVSD, atrioventricular septal
defect; HLHS, hypoplastic left heart syndrome; IAA, interrupted aortic arch; PDA, patent ductus
arteriosus; PS, pulmonic stenosis; TA, truncus arteriosus; TAPVR, total anomalous pulmonary
venous return; TGA, transposition of great arteries; TOF, tetralogy of Fallot; VSD, ventricular
septal defect.
Table 451.3
Genetics of Isolated Congenital Heart Disease
(Nonsyndromic)
GENE
PROTEIN ENCODED CARDIAC DEFECTS
IMPLICATED*
GENES ENCODING TRANSCRIPTION FACTORS
ANKRD1 Ankyrin repeat domain TAPVR
CITED2 cAMP responsive element-binding ASD, VSD
protein
FOG2/ZFPM2 Friend of GATA TOF
GATA6 GATA6 transcription factor ASD, VSD, TOF, PS, AVSD, PDA
HAND2 Helix-loop-helix transcription factor TOF
IRX4 Iroquois homeobox 4 VSD
MED13L Mediator complex subunit 13-like TGA
NKX2-5/NKX2.5 Homeobox containing transcription factor ASD, VSD, TOF, HLHS, CoA, TGA, IAA
TBX20 T-Box 20 transcription factor ASD, VSD, mitral stenosis
ZIC3 Zinc finger transcription factor TGA, PS, TAPVR, HLHS, ASD, VSD
GENES ENCODING RECEPTORS AND SIGNALING MOLECULES
ACVR1/ALK2 BMP receptor AVSD
ACVR2B Activin receptor PS, DORV, TGA
ALDH1A2 Retinaldehyde dehydrogenase TOF
CFC1/CRYPTIC Cryptic protein TOF, TGA, AVSD, ASD, VSD, IAA,
DORV
CRELD1 Epidermal growth factor–related proteins ASD; AVSD
FOXH1 Forkhead activin signal transducer TOF, TGA
GDF1 Growth differentiation factor-1 TOF, TGA, DORV, heterotaxy
GJA1 Connexin 43 ASD, HLHS, TAPVR
LEFTY2 Left-right determination factor TGA, AVSD, IAA, CoA
NODAL Nodal homolog (TGF-β superfamily) TGA, PA, TOF, DORV, TAPVR, AVSD
NOTCH1 NOTCH1 (Ligand of JAG1) Bicuspid aortic valve, AS, CoA, HLHS
PDGFRA Platelet-derived growth factor receptor α TAPVR
SMAD6 MAD-related protein Bicuspid aortic valve, CoA, AS
TAB2 TGF-β–activated kinase Outflow tract defects
TDGF1 Teratocarcinoma-derived growth factor 1 TOF, VSD
VEGF Vascular endothelial growth factor CoA, outflow tract defects
GENES ENCODING STRUCTURAL PROTEINS
ACTC α Cardiac actin ASD
MYH11 Myosin heavy chain 11 PDA, aortic aneurysm
MYH6 α-Myosin heavy chain ASD, TA, AS, TGA
MYH7 β-Myosin heavy chain Ebstein anomaly, ASD
* In many cases, mutation of a single gene has been closely linked to a specific cardiovascular
disease, for example, by finding a high incidence of mutations or deletions of that gene in a large
group of patients. These findings are often confirmed by studies in mice in which deletion or
alteration of the gene induces a similar cardiac phenotype to the human disease. In others,
mutation of a gene may increase the risk of cardiovascular disease, but with decreased
penetrance, suggesting that modifier genes or environmental factors play a role. Finally, in some
cases, gene mutations have only been identified in a small number of pedigrees, and confirmation
awaits screening of larger numbers of patients.
AS, Aortic stenosis; ASD, atrial septal defect; AVSD, atrioventricular septal defect; cAMP, cyclic
adenosine monophosphate; CoA, coarctation of the aorta; DORV, double-outlet right ventricle;
HLHS, hypoplastic left heart syndrome; IAA, interrupted aortic arch; PA, pulmonary artery; PDA,
patent ductus arteriosus; PS, pulmonic stenosis; TA, truncus arteriosus; TAPVR, total anomalous
pulmonary venous return; TGA, transposition of the great arteries; TGF, transforming growth
factor; TOF, tetralogy of Fallot; VSD, ventricular septal defect.
Partially adapted from Fahed AC, Gelb BD, Seidman JG, Seidman CE: Genetics of congenital
heart disease: the glass half empty. Circ Res 112:707–720, 2013.
Table 451.4
Genetics of Cardiomyopathies
CHROMOSOMAL
CARDIOMYOPATHY GENE
LOCATION
Hypertrophic cardiomyopathy 14q1 β-Myosin heavy chain
15q2 α-Tropomyosin
1q31 Troponin T
19p13.2-19q13.2 Troponin I
11p13-q13 Myosin-binding protein C
12q23 Cardiac slow myosin regulatory light chain
13p21 Ventricular slow myosin essential light chain
2q31 Titin
3p25 Caveolin-3
Mitochondrial DNA tRNA-glycine
Mitochondrial DNA tRNA-isoleucine
Hypertrophic cardiomyopathy 7q36.1 AMP-activated protein kinase
with Wolff-Parkinson-White
syndrome
Other Genetic Diseases Causing Cardiac Hypertrophy
Familial amyloid disease 18q12.1 Transthyretin (TTR)
Noonan syndrome 12q24.1, 2p22.1, Protein tyrosine phosphatase 11 (PTPN11), son of
3p25, 12p12.1 sevenless homolog 1 (SOS1), RAF1 protooncogene,
GTPase KRAS
Fabry disease Xq22 α-Galactoside A (GLA)
Danon disease Xq24 Lysosomal-associated membrane protein 2 (LAMP2)
Hereditary hemochromatosis 6p21.3 Hereditary hemochromatosis protein (HFE)
Pompe disease 17q25 Acid α-glucosidase (GAA)
Dilated cardiomyopathy
X-linked Xp21 Dystrophin
Xp28 Tafazzin
Autosomal recessive 19p13.2-19q13.2 Troponin I
Autosomal dominant : Genes encoding multiple proteins have been identified, including cardiac
actin; desmin; δ-sarcoglycan; β-myosin heavy chain; cardiac troponin C and T; α-tropomyosin;
titin; metavinculin; myosin-binding protein C; muscle LIM protein; α-actinin-2; phospholamban;
Cypher/LIM binding domain 3; α-myosin heavy chain; SUR2A (regulatory subunit of KATP
channel); and lamin A/C.
Isolated noncompaction of the left ventricle : Autosomal dominant, autosomal recessive, X-
linked, and mitochondrial inheritance patterns have been reported. Genes that have been
implicated include those for α-dystrobrevin, Cypher/ZASP, lamin A/C, Tafazzin, MIB1, and LIM
domain-binding protein 3 (LDB3).
Partially adapted from Dunn KE, Caleshu C, Cirino AL, et al: A clinical approach to inherited
hypertrophy: the use of family history in diagnosis, risk assessment, and management, Circ
Cardiovasc Genet 6:118–131, 2013.
Progress has also been made in identifying the genetic basis of dilated
cardiomyopathy, which is familial in 20–50% of cases. Autosomal dominant
inheritance is most often encountered, and similar to hypertrophic
cardiomyopathy, multiple genes have been identified (see Table 451.2 ). X-
linked inheritance accounts for 5–10% of cases of familial dilated
cardiomyopathy. Mutations in the dystrophin gene (chromosome Xp21) are the
most common in this group, causing Duchene or Becker muscular dystrophy .
Mutations in the gene encoding tafazzin are associated with Barth syndrome
and some cases of isolated noncompaction of the left ventricle (LVNC).
Autosomal recessive inheritance is associated with a mutation in cardiac
troponin I. Mitochondrial myopathies may be caused by mutations of enzymes
of the electron transport chain encoded by nuclear DNA (in which inheritance
will follow mendelian genetic patterns) or enzymes of fatty acid oxidation
encoded by mitochondrial DNA (which is inherited solely from the mother).
Table 451.4 is a compilation of the most common genetic causes of
cardiomyopathy.
The genetic basis of heritable arrhythmias , most notably the long QT
syndromes , has been linked to mutations of genes coding for subunits of
cardiac potassium and sodium channels (see Table 451.2 ). Other heritable
arrhythmias include arrhythmogenic right ventricular dysplasia , familial
atrial fibrillation, familial complete heart block, and Brugada syndrome . Table
451.5 is a compilation of the most common genetic causes of arrhythmias.
Table 451.5
Genetics of Arrhythmias
CHROMOSOMAL
ARRHYTHMIA GENE(S) IMPLICATED
LOCATION
Complete heart block 19q13 Not known
Long QT syndrome
LQT1 (autosomal dominant) 11p15.5 KVLQT1 (K+ channel)
LQT2 (autosomal dominant) 7q35 HERG (K+ channel)
LQT3 (autosomal dominant) 3p21 SCN5A (Na+ channel)
LQT4 (autosomal dominant) 4q25-27 Not known
LQT5 (autosomal dominant) 21q22-q22 KCNE1 (K+ channel)
LQT6 21q22.1 KCNE2 (K+ channel)
Jervell and Lange-Nielsen syndrome (autosomal recessive, 11p15.5 KVLQT1 (K+ channel)
congenital deafness)
LQT8-13 Unknown Private mutations (rare)
Arrhythmogenic right ventricular dysplasia (ARVD): 11 genes are now associated with ARVD (ARVD1 through
ARVD11 ) usually with autosomal dominant inheritance, but with variable penetrance. These genes include TGF
β3 (transforming growth factor β), RYR2 (ryanodine receptor), LAMR1 (laminin receptor-1), PTPLA (protein
tyrosine phosphatase), DSP (desmoplakin), PKP2 (plakophilin-2), DSG2 (desmoglein), and DSC2 (desmocollin).
Familial atrial fibrillation (autosomal dominant) 10q22-q24, 6q14-16 Not known
11p15.5 KVLQT1 (K+ channel)
11p15.5 KCNQ1 (K+ channel)
21q22 KCNE2 (K+ channel)
17q23.1-q24.2 KCNJ2 (K+ channel)
7q35-q36 KCNH2 (K+ channel)
Brugada syndrome (right bundle branch block, ST segment 3p21-p24 SCN5A (Na+ channel)
elevation, unexpected sudden death) 3p22-p24 GPD-1L (glycerol-3-
phosphate dehydrogenase)
Catecholaminergic polymorphic ventricular tachycardia — RYR2 (autosomal dominant)
— CASQ2 (autosomal recessive)
Of all cases of congenital heart disease, 2–4% are associated with known
environmental or adverse maternal conditions and teratogenic influences,
including maternal diabetes mellitus, maternal phenylketonuria, or systemic
lupus erythematosus; congenital rubella syndrome; and maternal ingestion of
drugs (lithium, ethanol, warfarin, thalidomide, antimetabolites, vitamin A
derivatives, anticonvulsant agents) (see Table 449.2 ). Associated noncardiac
malformations noted in identifiable syndromes may be seen in as many as 25%
of patients with CHD.
Gender differences in the occurrence of specific cardiac lesions have been
identified. Transposition of the great arteries and left-sided obstructive lesions
are slightly more common in boys (65%), whereas ASD, VSD, PDA, and
pulmonic stenosis are more common in girls. No racial differences in the
occurrence of congenital heart lesions as a whole have been noted; for specific
lesions such as transposition of the great arteries, a higher occurrence is seen in
white infants.
ORGANIC FUNCTIONAL
Type I* /TYPE
Type I* Type II † Type IIIa ‡
IIIb ‡
Nonischemic Endocarditis Degenerative Rheumatic (chronic RF); Cardiomyopathy;
(perforation); (billowing/flail leaflets); iatrogenic (radiation/drug); myocarditis; left-
degenerative endocarditis (ruptured inflammatory ventricular
(annular chordae); traumatic (lupus/anticardiolipin), dysfunction (any
calcification); (ruptured chord/PM); eosinophilic (endocardial cause)
congenital (cleft rheumatic (acute RF) disease, endomyocardial
leaflet) fibrosis)
Ischemic — Ruptured PM — Functional
ischemic
* Mechanism involves normal leaflet movement.
†
Mechanism involves excessive valve movement.
‡ Restricted valve movement, IIIa in diastole, IIIb in systole.
In isolated mitral insufficiency, the mitral valve annulus is usually dilated, the
chordae tendineae are short and may insert anomalously, and the valve leaflets
are deformed. When mitral insufficiency is severe enough to cause clinical
symptoms, the left atrium enlarges as a result of the regurgitant flow, and the left
ventricle becomes hypertrophied and dilated. Pulmonary venous pressure is
increased, and the increased pressure ultimately results in pulmonary
hypertension and RV hypertrophy and dilation. Mild lesions produce no
symptoms; the only abnormal sign is the apical holosystolic murmur of mitral
regurgitation. Severe regurgitation results in symptoms that can appear at any
age, including poor physical development, frequent respiratory infections,
fatigue on exertion, and episodes of pulmonary edema or congestive heart
failure. Often, a diagnosis of reactive airways disease will have been made
because of the similarity in pulmonary symptoms, including wheezing, which
may be a dominant finding in infants and young children.
The typical murmur of mitral insufficiency is a moderately high-pitched,
apical blowing holosystolic murmur. If the insufficiency is moderate to severe, it
is usually associated with a low-pitched, apical mid-diastolic rumbling murmur
indicative of increased diastolic flow across the mitral valve. The pulmonic
direction of flow in the vertical vein that differentiates it from a left superior
vena cava. C, Total anomalous pulmonary venous drainage below the
diaphragm. The specimen shows the pulmonary veins as they enter the
confluence, whereas the echocardiogram demonstrates the descending
veins as they enter the liver. Note that the direction of flow is away from the
heart. AO, Aorta; CS, coronary sinus; DA, descending aorta; DV,
descending vein; LVV, left vertical vein; PA, pulmonary artery; PV,
pulmonary vein; PVC, pulmonary venous confluence; RA, right atrium.
(From Webb GD, Smallhorn, JF, Therrien J, Redington, AN: Congenital
heart disease in the adult and pediatric patient. In Braunwald's heart
disease: a textbook of cardiovascular medicine, ed 11, Philadelphia, 2018,
Elsevier, Fig 75-32, p 1553).
Table 458.1
Total Anomalous Pulmonary Venous Return
Clinical Manifestations
Two major clinical patterns of TAPVR are seen, depending on the presence or
FIG. 458.16 A, Coronal T1-weighted MR image of a patient with
heterotaxy syndrome (polysplenia) demonstrates a bilateral hyparterial
bronchial branching pattern (arrows) and left upper quadrant spleens. B,
More posterior coronal T1-weighted MR image shows left azygos-
hemiazygos continuation to the left superior vena cava and right thoracic
aorta. (From Applegate KE, Goske MJ, Pierce G, Murphy D: Situs revisited:
imaging of the heterotaxy syndrome, Radiographics 19:837–852, 1999, Fig
4.)
Table 458.2
FIG. 458.17 Pathway of left-right (LR) development in the mouse embryo, list of genes
associated with human LR asymmetry disorders, and corresponding phenotypes in
humans. LRD-containing monocilia generate leftward nodal flow, and polycystin 2–
containing cilia sense nodal flow and initiate an asymmetric calcium signal, which
induces Nodal expression around the node. Nodal signaling is involved in asymmetric
morphogenesis by inducing expression of the Nodal-responsive genes (NODAL,
appreciated by reviewing the embryology of the aortic arch (see Chapter 447 ,
Fig. 447.1 ). The most common anomalies include (1) double aortic arch (Fig.
459.1A ), (2) right aortic arch with a left ligamentum arteriosum, (3) anomalous
innominate artery arising farther to the left on the arch than usual, (4) anomalous
left carotid artery arising farther to the right than usual and passing anterior to
the trachea, and (5) anomalous left pulmonary artery (vascular sling ). In the
latter anomaly, the abnormal vessel arises from an elongated main pulmonary
artery or from the right pulmonary artery. It courses between and compresses the
trachea and the esophagus. Associated congenital heart disease may be present in
5–50% of patients, depending on the vascular anomaly.
Table 459.1
Vascular Rings
BARIUM
LESION SYMPTOMS PLAIN FILM BRONCHOSCOPY
SWALLOW
DOUBLE ARCH
Stridor AP—wider base of Bilateral Bilateral tracheal
Respiratory heart indentation compression—both
distress Lat.—narrowed of esophagus pulsatile
Swallowing trachea displaced
dysfunction forward at C3-C4
Reflex
apnea
ANOMALOUS INNOMINATE
Cough AP—normal Normal Pulsatile anterior
Stridor Lat.—anterior tracheal tracheal compression
Reflex compression
apnea
AP, Anteroposterior; L and l., left; Lat., lateral; MRI, magnetic resonance imaging; R and r., right.
From Kliegman RM, Greenbaum LA, Lye PS: Practical strategies in pediatric diagnosis and
therapy, ed 2, Philadelphia, 2004, Elsevier, p 88.
FIG. 459.1 Double aortic arch. A, Small anterior segment of the double aortic arch
(most common type). B, Operative procedure for release of the vascular ring. L., Left; a.
and art., artery; ant., anterior; innom., innominate; duct. arterios., ductus arteriosus;
pulm., pulmonary.
Clinical Manifestations
Fallot, transposition of the great arteries, congenitally corrected transposition of
the great arteries, single ventricle, tricuspid atresia, truncus arteriosus,
quadricuspid or bicuspid aortic valves, double-outlet ventricle). In addition,
acquired lesions of the coronary arteries caused by existing congenital heart
disease may develop as a consequence of hypertension or alterations in blood
flow; congenital heart lesions include coarctation of the aorta, supravalvular
aortic stenosis, aortic regurgitation, pulmonary atresia with intact ventricular
septum, hypoplastic left heart syndrome, and coronary ectasia secondary to
cyanotic heart disease.
Table 459.2
Congenital Anomalies of Coronary Arteries
Unassociated With Congenital Heart Disease
Anomalous Aortic Origin
Table 459.3
Table 461.1
Extracardiac Complications of Cyanotic Congenital Heart
Disease and Eisenmenger Physiology
PROBLEM ETIOLOGY THERAPY
Polycythemia Persistent hypoxia Phlebotomy if symptomatic
Relative anemia Nutritional deficiency Iron replacement
CNS abscess Right-to-left shunting Antibiotics, drainage
CNS thromboembolic stroke Right-to-left shunting or Anticoagulation, phlebotomy
polycythemia
Low-grade DIC, thrombocytopenia Polycythemia None for DIC unless
bleeding, then phlebotomy
Hemoptysis Pulmonary infarct, thrombosis, or Embolization
rupture of pulmonary artery
plexiform lesion
Plastic bronchitis Fontan procedure Bronchoscopy, vascular
coiling, lymphatic ablation
Gum disease Polycythemia, gingivitis, bleeding Dental hygiene
Gout Polycythemia, diuretic agent Allopurinol
Arthritis, clubbing Hypoxic osteoarthropathy None
Pregnancy complications: miscarriage, fetal Poor placental perfusion, poor Pregnancy prevention
growth retardation, prematurity increase, ability to increase cardiac output counseling, high-risk
maternal illness obstetric management
Infections Associated asplenia, DiGeorge Antibiotics
syndrome, endocarditis
Fatal RSV pneumonia with Ribavirin; RSV
pulmonary hypertension immunoglobulin (prevention)
Failure to thrive Increased oxygen consumption, Treat heart failure; correct
decreased nutrient intake defect early; increase caloric
intake
Protein-losing enteropathy s/p Fontan; high right-sided Oral budesonide or sildenafil
pressures
Chylothorax Injury to thoracic duct Medium-chain
triglyceride diet
Octreotide
Surgical ligation of
thoracic duct
Neurodevelopmental disabilities Chronic hypoxia, cardiac surgery, Early school-based
genetic evaluation and intervention
Psychosocial adjustment Limited activity, cyanotic Counseling
appearance, chronic disease,
multiple hospitalizations
CNS, Central nervous system; DIC, disseminated intravascular coagulation; RSV, respiratory
syncytial virus; s/p, status post (after).
It has become apparent that even the simplest congenital heart lesions can be
associated with long-term complications, including both cardiac and noncardiac
problems (Tables 461.4 and 461.5 and Fig. 461.2 ). Cardiac complications
include arrhythmias and conduction defects, ventricular dysfunction, residual
shunts, valvular lesions (regurgitation and stenosis), hypertension, and
aneurysms. Noncardiac sequelae (comorbidities ) include pulmonary, renal, and
hepatic dysfunction that is caused either directly or indirectly by the underlying
CHD. Abnormal pulmonary function most often presents as restrictive lung
physiology and likely results from prior sternotomy or thoracotomy, scoliosis,
diaphragmatic dysfunction, or parenchymal lung disease. Reduced pulmonary
function contributes to reduced exercise tolerance and is a risk factor for
mortality in adults with CHD. Renal dysfunction may result from chronic
cyanosis, multiple surgeries requiring CPB, or from other comorbid conditions,
such as hypertension and diabetes mellitus. Hepatic injury from chronic liver
congestion in patients with elevated central venous pressures, particularly
patients palliated with the Fontan procedure, can result in hepatic fibrosis,
cirrhosis, hepatic dysfunction, and rarely hepatocellular carcinoma. Adults with
CHD are at risk for developmental abnormalities such as intellectual
impairment, somatic abnormalities such as facial dysmorphism (cleft palate/lip),
CNS abnormalities such as seizure disorders from previous thromboembolic
events or cerebrovascular accidents, and impairments of hearing or vision loss.
Psychosocial problems involving employment, life and health insurance,
participation in sports, sexual activity, and contraception are common. As a
result of these long-term complications, the majority of adults with CHD need
lifelong follow-up. When adults with CHD are hospitalized, it is usually for
heart failure or an arrhythmia; others may require catheterization or another
cardiac surgical procedure.
Table 461.4
Risks in Adults Who Have Congenital Heart
Disease
Rhythm Disorders
Supraventricular tachycardia
Right bundle branch block
Heart block
Ventricular tachycardia
Sudden death
Coarctation of Aorta
Essential hypertension
Recoarctation
Aneurysm formation
Acquired Lesions
Table 461.5
Table 462.1
Antiarrhythmic Drugs Commonly Used in Pediatric
Patients, by Class
DRUG DRUG
DRUG INDICATIONS DOSING SIDE EFFECTS
INTERACTIONS LEVEL
CLASS IA: INHIBITS Na+ FAST CHANNEL, PROLONGS REPOLARIZATION
Quinidine SVT, atrial Oral: 30-60 Nausea, vomiting, Enhances digoxin, 2-6 µg/mL
fibrillation, atrial mg/kg/24 hr diarrhea, fever, may increase PTT
flutter, VT. In divided q6h cinchonism, QRS and when given with
atrial flutter, an (sulfate) or q8h QT prolongation, AV warfarin
AV node– (gluconate) nodal block, asystole
blocking drug In adults, 10 syncope,
(digoxin, mg/kg/day thrombocytopenia,
verapamil, divided q6h hemolytic anemia,
propranolol) must Max dose: 2.4 SLE, blurred vision,
be given first to g/24 hr convulsions, allergic
prevent 1 : 1 reactions, exacerbation
conduction of periodic paralysis
Procainamide SVT, atrial Oral: 15-50 PR, QRS, QT interval Toxicity increased 4-8
fibrillation, atrial mg/kg/24 hr prolongation, by amiodarone and µg/mL
flutter, VT divided q4h anorexia, nausea, cimetidine With
Max dose: 4 vomiting, rash, fever, NAPA
g/24 hr agranulocytosis, <40
IV: 10-15 thrombocytopenia, µg/mL
mg/kg over 30- Coombs-positive
45 min load hemolytic anemia,
followed by 20- SLE, hypotension,
80 µg/kg/min exacerbation of
Max dose: 2 periodic paralysis,
g/24 hr proarrhythmia
Disopyramide SVT, atrial Oral: <2 yr: 20- Anticholinergic 2-5 µg/ml
fibrillation, atrial 30 mg/kg/24 hr effects, urinary
flutter divided q6h or retention, blurred
q12h (long- vision, dry mouth, QT
acting form); 2- and QRS
10 yr: 9-24 prolongation, hepatic
mg/kg/24 hr toxicity, negative
divide q6h or inotropic effects,
q12h (long- agranulocytosis,
acting form); 11 psychosis,
yr: 5-13 hypoglycemia,
mg/kg/24 hr proarrhythmia
divided q6h or
q12h (long-
acting)
Max dose: 1.2
g/24 hr
CLASS IB: INHIBITS Na+ FAST CHANNEL, SHORTENS REPOLARIZATION
Lidocaine VT, VF IV: 1 mg/kg repeat qCNS effects, Propranolol, 1-5 µg/mL
5 min 2 times confusion, cimetidine,
followed by 20-50 convulsions, high- increases toxicity
µg/kg/min (max grade AV block,
dose: 3 mg/kg) asystole, coma,
paresthesias,
respiratory failure
Mexiletine VT Oral: 6-15 mg/kg/24 GI upset, skin rash, Cimetidine 0.8-2
hr divided q8h neurologic µg/mL
Phenytoin Digitalis Oral: 3-6 Rash, gingival Amiodarone, oral 10-20
intoxication mg/kg/24 hr hyperplasia, ataxia, anticoagulants, µg/mL
divided q12h lethargy, vertigo, cimetidine,
Max dose: 600 tremor, macrocytic nifedipine,
mg anemia, bradycardia disopyramide,
IV: 10-15 with rapid push increase toxicity
mg/kg over 1 hr
load
+
CLASS IC: INHIBITS Na CHANNEL
Flecainide SVT, atrial Oral: 6.7-9.5 Blurred vision, nausea, Amiodarone 0.2-1
tachycardia, VT mg/kg/24 hr decrease in increases toxicity µg/mL
divided q8h contractility,
In older proarrhythmia
children, 50-
200 mg/m2 /day
divided q12h
Propafenone SVT, atrial Oral: 150-300 Hypotension, Increases digoxin 0.2-1
tachycardia, atrial mg/m2 /24 hr decreased contractility, levels µg/mL
fibrillation, VT divided q6h hepatic toxicity,
paresthesia, headache,
proarrhythmia
CLASS II: β-BLOCKERS
Propranolol SVT, long QT Oral: 1-4 Bradycardia, loss of Co-administration
mg/kg/24 hr concentration, school with disopyramide,
divided q6h performance problems flecainide, or
Max dose 60 bronchospasm, verapamil may
mg/24 hr hypoglycemia, decrease
IV: 0.1-0.15 hypotension, heart ventricular
mg/kg over 5 block, CHF function.
min
Max IV dose:
10 mg
Atenolol SVT Oral: 0.5-1 Bradycardia, loss of Co-administration
mg/kg/24 hr once concentration, school with disopyramide,
daily or divided performance problems flecainide, or
q12h verapamil may
decrease
ventricular
function.
Nadolol SVT, long QT Oral: 1-2 mg/kg/24 Bradycardia, loss of Co-administration
hr given once daily concentration, school with disopyramide,
performance problems flecainide, or
bronchospasm, verapamil may
hypoglycemia, decrease
hypotension, heart ventricular
block, CHF function.
CLASS III: PROLONGS REPOLARIZATION
Amiodarone SVT, JET, VT Oral: 10 Hypothyroidism or Digoxin (increases 0.5-2.5
mg/kg/24 hr in hyperthyroidism, levels), flecainide, mg/L
1-2 divided elevated triglycerides, procainamide,
doses for 4-14 hepatic toxicity, quinidine,
days; reduce to pulmonary fibrosis warfarin,
5 mg/kg/24 hr phenytoin
for several
weeks; if no
recurrence,
reduce to 2.5
mg/kg/24 hr
IV: 2.5-5 mg/kg
over 30-60 min,
may repeat 3
times, then 2-10
mg/kg/24 hr
continuous
infusion
CLASS IV AND MISCELLANEOUS MEDICATIONS
Digoxin SVT (not WPW), Oral/load PAC, PVC, Quinidine 1-2 mg/mL
atrial flutter, atrial instructions: bradycardia, AV Amiodarone,
fibrillation Premature: 20 block, nausea, verapamil,
µg/kg vomiting, anorexia, increase
Newborn: 30 prolongs PR interval digoxin levels
µg/kg
>6 mo: 40
µg/kg
Give total dose
followed by q8-
12h × 2 doses
Maintenance:
10 µg/kg/24 hr
divide q12h
Max dose: 0.5
mg
IV: PO dose
Max dose: 0.5
mg
Verapamil SVT (not WPW) Oral: 2-7 Bradycardia, asystole, Use with β-blocker
mg/kg/24 hr high degree AV block, or disopyramide
divided q8h PR prolongation, exacerbates CHF,
Max dose: 480 hypotension, CHF increases digoxin
mg level and toxicity
IV: 0.1-0.2
mg/kg q 20 min
× 2 doses
Max dose: 5-10
mg
AV, Atrioventricular; CHF, congestive heart failure; CNS, central nervous systems; GI,
gastrointestinal; IV, intravenous; JET, junctional ectopic tachycardia; NAPA, N -acetyl
procainamide; PAC, premature atrial contraction; PTT, partial thromboplastin time; PVC,
premature ventricular contraction; SLE, systemic lupus erythematosus–like illness; SVT,
supraventricular tachycardia; VF, ventricular fibrillation; VT, ventricular tachycardia; WPW, Wolff-
Parkinson-White syndrome.
462.1
Principles of Antiarrhythmic
Therapy
Aarti S. Dalal, George F. Van Hare
FIG. 462.10 Ventricular arrhythmias. (From Park MY: Pediatric cardiology for
practitioners, ed 5, Philadelphia, 2008, Mosby Elsevier, Fig. 24-6, p 429.)
Table 462.2
Diagnosis of Tachyarrhythmias: Electrocardiographic
Findings
Table 462.3
Updated Summary of Heritable Arrhythmia Syndrome
Susceptibility Genes
GENE LOCUS PROTEIN
LONG QT SYNDROME (LQTS)
Major LQTS Genes
KCNQ1 (LQT1) 11p15.5 IKs potassium channel alpha subunit (KVLQT1, Kv 7.1)
KCNH2 (LQT2) 7q35-36 IKr potassium channel alpha subunit (HERG, Kv 11.1)
SCN5A (LQT3) 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
Minor LQTS Genes (Listed Alphabetically)
AKAP9 7q21-q22 Yotiao
CACNA1C 12p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
CALM1 14q32.11 Calmodulin 1
CALM2 2p21 Calmodulin 2
CALM3 19q13.2-q13.3 Calmodulin 3
CAV3 3p25 Caveolin-3
KCNE1 21q22.1 Potassium channel beta subunit (MinK)
KCNE2 21q22.1 Potassium channel beta subunit (MiRP1)
KCNJ5 11q24.3 Kir3.4 subunit of IKACH channel
SCN4B 11q23.3 Sodium channel beta4 subunit
SNTA1 20q11.2 Syntrophin-alpha1
TRIADIN KNOCKOUT (TKO) SYNDROME
TRDN 6q22.31 Cardiac triadin
ANDERSEN-TAWIL SYNDROME (ATS)
KCNJ2 (ATS1) 17q23 IK1 potassium channel (Kir2.1)
TIMOTHY SYNDROME (TS)
CACNA1C 12p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
Cardiac-Only TS (COTS)
CACNA1C 12p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
SHORT QT SYNDROME (SQTS)
KCNH2 (SQT1) 7q35-36 IKr potassium channel alpha subunit (HERG, Kv 11.1)
KCNQ1 (SQT2) 11p15.5 IKs potassium channel alpha subunit (KVLQT1, Kv 7.1)
KCNJ2 (SQT3) 17q23 IK1 potassium channel (Kir2.1)
CACNA1C (SQT4) 12p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
CACNB2 (SQT5) 10p12 Voltage-gated L-type calcium channel beta2 subunit
CACN2D1 (SQT6) 7q21-q22 Voltage-gated L-type calcium channel 2 delta1 subunit
CATECHOLAMINERGIC POLYMORPHIC VENTRICULAR TACHYCARDIA (CPVT)
RYR2 (CPVT1) 1q42.1-q43 Ryanodine receptor 2
CASQ2 (CPVT2) 1p13.3 Calsequestrin 2
KCNJ2 (CPVT3) 17q23 IK1 potassium channel (Kir2.1)
CALM1 14q32.11 Calmodulin 1
CALM3 19q13.2-q13.3 Calmodulin 3
TRDN 6q22.31 Cardiac triadin
BRUGADA SYNDROME (BrS)
SCN5A (BrS1) 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
Minor Brs Genes (Listed Alphabetically)
ABCC9 12p12.1 ATP-binding cassette, subfamily C member 9
CACNA1C 2p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
CACNA2D1 7q21-q22 Voltage-gated L-type calcium channel 2 delta1 subunit
CACNB2 10p12 Voltage-gated L-type calcium channel beta2 subunit
FGF12 3q28 Fibroblast growth factor 12
GPD1L 3p22.3 Glycerol-3-phosphate dehydrogenase 1–like
KCND3 1p13.2 Voltage-gated potassium channel (Ito ) subunit Kv 4.3
KCNE3 11q13.4 Potassium channel beta3 subunit (MiRP2)
KCNJ8 12p12.1 Inward rectifier K+ channel Kir6.1
HEY2 6q Hes-related family BHLH transcription factor with YRPW motif 2
PKP2 12p11 Plakophilin-2
RANGRF 17p13.1 RAN guanine nucleotide release factor 1
SCN1B 19q13 Sodium channel beta1
SCN2B 11q23 Sodium channel beta2
SCN3B 11q24.1 Sodium channel beta3
SCN10A 3p22.2 Sodium voltage-gated channel alpha10 subunit (Nav 1.8)
SLMAP 3p14.3 Sarcolemma-associated protein
EARLY REPOLARIZATION SYNDROME (ERS)
ABCC9 12p12.1 ATP-binding cassette, subfamily C member 9
CACNA1C 2p13.3 Voltage-gated L-type calcium channel (Cav 1.2)
CACNA2D1 7q21-q22 Voltage-gated L-type calcium channel 2 delta1 subunit
CACNB2 10p12 Voltage-gated L-type calcium channel beta2 subunit
KCNJ8 12p12.1 Inward rectifier K+ channel Kir6.1
SCN5A 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
SCN10A 3p22.2 Sodium voltage-gated channel alpha10 subunit (Nav 1.8)
IDIOPATHIC VENTRICULAR FIBRILLATION (IVF)
ANK2 4q25-q27 Ankyrin B
CALM1 14q32.11 Calmodulin 1
DPP6 7q36 Dipeptidyl-peptidase-6
KCNJ8 12p12.1 Inward rectifier K+ channel Kir6.1
RYR2 1q42.1-q43 Ryanodine receptor 2
SCN3B 11q23 Sodium channel beta3 subunit
SCN5A 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
PROGRESSIVE CARDIAC CONDUCTION DISEASE/DEFECT (PCCD)
SCN5A 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
TRPM4 19q13.33 Transient receptor potential cation channel, subfamily M, member 4
SICK SINUS SYNDROME (SSS)
ANK2 4q25-q27 Ankyrin B
HCN4 15q24-q25 Hyperpolarization-activated cyclic nucleotide–gated channel 4
MYH6 14q11.2 Myosin, heavy chain 6, cardiac muscle, alpha
SCN5A 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
“ANKYRIN-B SYNDROME”
ANK2 4q25-q27 Ankyrin B
FAMILIAL ATRIAL FIBRILLATION (FAF)
ANK2 4q25-q27 Ankyrin B
GATA4 8p23.1-p22 GATA-binding protein 4
GATA5 20q13.33 GATA-binding protein 5
GJA5 1q21 Connexin 40
KCNA5 12p13 IKur potassium channel (Kv 1.5)
KCNE2 21q22.1 Potassium channel beta subunit (MiRP1)
KCNH2 7q35-36 IKr potassium channel alpha subunit (HERG, Kv 11.1)
KCNJ2 17q23 IK1 potassium channel (Kir2.1)
KCNQ1 11p15.5 IKs potassium channel alpha subunit (KVLQT1, Kv 7.1)
NPPA 1p36 Atrial natriuretic peptide precursor A
NUP155 5p13 Nucleoporin 155 kD
SCN5A 3p21-p24 Cardiac sodium channel alpha subunit (Nav 1.5)
From Tester DJ, Ackerman MJ: Genetics of cardiac arrhythmias. In Braunwald's heart disease, ed
11, Philadelphia, 2018, Elsevier (Table 33.1, p 605.)
FIG. 462.11 Genotype-phenotype correlations in long QT syndrome (LQTS). About 75%
of clinically strong LQTS is caused by mutations in 3 genes (35% KCNQ1 , 30% KCNH2
, and 10% SCN5A ) encoding for ion channels that are critically responsible for the
orchestration of the cardiac action potential. Observed genotype-phenotype
correlations include swimming/exertion/emotion and LQT1, auditory
triggers/postpartum period and LQT2, and sleep/rest and LQT3. (From Tester DJ,
Ackerman MJ: Genetics of cardiac arrhythmias. In Braunwald's heart disease, ed 11,
Philadelphia, 2018, Elsevier, Fig 33-3, p 607.)
Genetic studies have identified mutations in cardiac potassium and sodium
channels (Table 462.3 ). Additional forms (up to 13 variants) of long QT
syndrome (LQTS) have been described, but these are much more uncommon.
Genotype may predict clinical manifestations; for example, LQTS type 1 (LQT1
) events are usually induced by stress or exertion, whereas events in LQT3 often
occur at rest, especially during sleep (Fig. 462.11 ). LQT2 events have an
intermediate pattern, often occurring in the postpartum period or with auditory
triggers. LQT3 has the highest probability for sudden death, followed by LQT2
and then LQT1. Drugs may prolong the QT interval directly but more often do
so when drugs such as erythromycin or ketoconazole inhibit their metabolism
(Table 462.4 ).
Table 462.4
Acquired Causes of QT Prolongation*
Drugs