You are on page 1of 30

Chapter 169   Cardiac Disorders

Alson S. Inaba and Timothy Horeczko

■  PERSPECTIVE into the fetal pulmonary artery via the right ventricle. Since
the fetal pulmonary vascular resistance (PVR) is higher than
Pediatric cardiac disease has typically been divided into con- the fetal systemic vascular resistance (SVR), this deoxygen-
genital and acquired disorders, with a further subdivision of ated blood bypasses the nonoxygenated fetal lungs via the
the congenital heart disorders into cyanotic and acyanotic patent ductus arteriosus (see Fig. 169-1). The poorly oxygen-
lesions. From a purely clinical standpoint, however, children ated blood enters the aorta via the patent ductus arteriosus and
with cardiac disorders present to the emergency department then mixes with the well-oxygenated blood in the descending
in one of two scenarios. In the first scenario, the child has signs aorta. The mixed blood in the descending aorta then returns
and symptoms that may represent an exacerbation or compli- to the placenta for oxygenation via the two umbilical
cation of an already known underlying cardiac disorder. In arteries.
cases with a known underlying cardiac disorder, early consulta- Once the infant is delivered and the umbilical cord is cut,
tion with the child’s cardiologist, along with comparisons of expansion and aeration of the lungs causes a decrease in PVR,
the child’s previous and most recent diagnostic cardiac studies with a concomitant increase in pulmonary blood flow. Increased
such as chest radiographs, electrocardiograms (ECGs), and oxygenation causes a physiologic closure of the umbilical arter-
echocardiograms, would also be extremely useful in the evalu- ies, umbilical vein, ductus venosus, and ductus arteriosus. An
ation and management phases. increase in the pulmonary blood flow to the infant’s left atrium
The second scenario represents more of a challenge to the also promotes closure of the foramen ovale. Complete ana-
emergency physician: the child with an undiagnosed congeni- tomic closure of the foramen ovale does not occur until about
tal or acquired cardiac disorder who presents to the emergency 3 months of age. Although the ductus arteriosus functionally
department with nonspecific or concerning signs and symp- closes at about 10 to 15 hours of life, complete anatomic closure
toms (Box 169-1). This chapter focuses on some of the more does not occur until about 2 to 3 weeks of life.
common and life-threatening cardiac disorders in infants and In the absence of any congenital cardiac defects, these tran-
children who present to the emergency department, with an sitional circulatory changes pose no physiologic problems to
emphasis on rapid evaluation, stabilization, and management the infant. However, closure of the ductus arteriosus can pose
of these disorders. life-threatening complications in neonates with specific con-
genital cardiac defects that are dependent on the patency of
the ductus arteriosus for survival.
■  PRINCIPLES OF DISEASE
Fetal and Neonatal Circulation
Pathophysiology of Cardiovascular
Some of the key features of the fetal circulation that differ Compensatory Responses
from that of a child are the presence of the ductus venosus,
ductus arteriosus, and a patent foramen ovale. During fetal There are two fundamental and clinically useful physiologic
development, oxygenation of the fetal circulation bypasses the formulas to keep in mind during the clinical assessment and
fetal lungs and is accomplished via the placenta. Blood that is management of cardiac disorders:
oxygenated by the placenta flows to the fetus via the umbilical
vein, bypasses the fetal liver via the ductus venosus, and Cardiac Output = (Stroke Volume) × (Heart Rate)
returns to the fetal heart via the inferior vena cava. Blood Blood Pressure = (Cardiac Output ) × (SVR )
returning from the inferior vena cava then enters the right
atrium and is preferentially shunted across to the left atrium The young myocardium is inefficient and unable to increase
via the patent foramen ovale (Fig. 169-1). The blood in the its contractility in response to demand.1 When more cardiac
left atrium is then pumped out of the aorta via the left ventri- output is needed, infants and children respond with an increase
cle. The oxygenated blood that is ejected through the ascend- in heart rate. Therefore, bradycardia in infants and young
ing aorta is preferentially directed to the fetal coronary and children is an ominous sign that connotes a severely compro-
cerebral circulations. mised cardiac output. Children develop the adult capacity to
Deoxygenated blood that returns to the right atrium via the increase their stroke volume to improve overall cardiac output
superior vena cava crosses the tricuspid valve and is pumped by 8 to 10 years of age.2

2138
2139
Common Presenting Signs and Symptoms of
BOX 169-1 Cardiac Disorders in Infants and Children

Chapter 169 / Cardiac Disorders


General SVC
Fussiness
AO
Lethargy RPA
Poor feeding (with or without associated diaphoresis) DA
Poor growth
LPA
Respiratory
Respiratory distress
Wheezing PA
Apnea FO LA
Cardiovascular
Tachycardia
RA
Shock
Paleness
Mottling
Cyanosis LV
Palpitations
Chest pain
Syncope
Various dysrhythmias RV

IVC

Based on the first physiologic formula, as stroke volume


decreases, a compensatory increase in the heart rate will be
necessary to preserve a normal cardiac output. A decrease in
stroke volume can be produced by a weak “pump,” decreased
volume in the circulation, or both. The most common cause Figure 169-1.  Normal intracardiac fetal circulation: Physiologic shunting
of decreased stroke volume in children is hypovolemia due to through the patent foramen ovale (FO) and the patent ductus arteriosus
(DA). Oxygenated blood from the placenta (red arrows) reaches the right
dehydration. Other causes of decreased stroke volume in chil-
atrium (RA) via the inferior vena cava (IVC). This well-oxygenated blood is
dren may be responsible (Box 169-2). Thus, tachycardia is the preferentially shunted from the RA across to the left atrium (LA) through
first compensatory cardiovascular response when the stroke the FO and is then ejected out the left ventricle (LV) to the ascending
volume is decreased. If tachycardia alone is not enough to aorta (AO). Deoxygenated blood (blue arrows) returning from the superior
maintain a normal cardiac output, the next compensatory vena cava (SVC) preferentially travels from the RA into the right ventricle
physiologic mechanism to preserve perfusion is an increase in (RV) and then out through the main pulmonary artery (PA). Because of
the high pulmonary vascular resistance in the fetal lungs, this
the SVR. This increase in SVR is exhibited as an increase in
deoxygenated blood bypasses lungs and enters the descending aorta via
the diastolic blood pressure, which in turn accounts for a the DA. Thus, the areas of the fetal body that are perfused by arteries
narrowed pulse pressure. The clinical examination of the proximal to the DA receive well-oxygenated blood, whereas those areas
extremities of a child with an increased SVR includes pallor, of the body that are perfused by arteries distal to the DA receive blood
mottling, cool skin, a delayed capillary refill time (>2 seconds), with a mixed oxygenation. LPA, left pulmonary artery; RPA, right
and weak or thready distal pulses. pulmonary artery.

Pathophysiology of Cyanosis Etiologies of Decreased Stroke Volume in


BOX 169-2 Infants and Children
Perspective
Hypovolemia (most commonly secondary to dehydration)
Cyanosis is a clinical sign caused by the presence of deoxygen-
Congestive heart failure (acquired or secondary to
ated blood in the capillary beds, most readily observed in the
underlying congenital cardiac defects)
mucous membranes, conjunctiva, nail beds, and skin. The
Myocarditis
presence of cyanosis usually means that there are at least 4 to
Hypertrophic cardiomyopathy with decreased diastolic
5 g/dL of deoxyhemoglobin in the blood, which correlates to
filling
an oxygen saturation of about 80 to 85%.3 Pathophysiologi-
Dilated cardiomyopathy with decreased systolic ejection
cally, central cyanosis results from a decrease in pulmonary
Pericarditis or pericardial effusion with cardiac tamponade
ventilation/oxygenation, a decrease in pulmonary perfusion,
Tachydysrhythmias with decreased diastolic filling times
shunting of deoxygenated blood directly into the systemic
circulation, or the presence of abnormal hemoglobin. Cyanosis
in the neonate can be due to a variety of cardiac, pulmonary, Clinical Features of Cyanosis
or hematologic causes. Cardiac causes of cyanosis include con-
genital lesions with right-to-left shunts as well as cardiac The region of the body that is cyanotic can provide important
lesions with decreased or increased pulmonary blood flow. clinical clues as to the cause of the cyanosis. Central cyanosis
Common pulmonary causes of cyanosis include bronchiolitis, involves the lips, tongue, and mucous membranes, whereas
pneumonia, and pulmonary edema. Methemoglobinemia can peripheral cyanosis (acrocyanosis) involves the hands and feet.
be one of the hematologic causes of cyanosis. Acrocyanosis is a common phenomenon in neonates caused by
2140
Clinical Clues to Help Distinguish between Key Elements to Elicit in the History of a Child
BOX 169-3 with a Known Cardiac Disorder
Table 169-1 Cardiac and Pulmonary Etiologies of  
PART V  ■  Special Populations / Section One • The Pediatric Patient

Central Cyanosis
Cardiac diagnosis
PULMONARY Congenital or acquired disorder?
CARDIAC ETIOLOGY ETIOLOGY Any episodes of previous decompensation (if so, are the
current signs and symptoms similar to or different
Respiratory status May be “comfortably Respiratory
blue” distress
from those previous episodes)?
Oxygen issues
Response to crying Worsening cyanosis Improved cyanosis Currently on home oxygen supplementation (continuous
Response to Minimal to no Improvement or only during feedings and sleep)?
oxygen improvement with oxygen
Baseline oxygen saturation (room air or while on home
Cyanosis due to severe pulmonary disease (e.g., severe pneumonia, tension oxygen)?
pneumothorax, acute chest syndrome of sickle cell disease) may not show Any recent need for increasing the amount of
significant improvement with supplemental oxygen, but these children will
also typically exhibit severe respiratory distress along with clinical cyanosis.
supplemental oxygen?
Medications
Names and dosages of all current medications (cardiac
and noncardiac medications)?
cold stress and peripheral vasoconstriction. Central cyanosis Were any of these cardiac medications stopped recently
reflects a pathologic origin and is an ominous sign. Infants with (by the cardiologist or parental noncompliance)?
cyanosis secondary to a congenital heart defect may not exhibit Any recent increases in the cardiac medications (reasons
as much respiratory distress as compared with the infant with for the increase, previous dosage versus the current
cyanosis due to a pulmonary cause. Thus, a cardiac cause for dosage, and the date this dosage was increased)?
central cyanosis may be more likely than a purely pulmonary Any new cardiac medications added recently and the
cause in the child who appears “comfortably blue.” Another reason for these additions?
important clinical clue to the cause of central cyanosis is that Recent digoxin level if the patient is on daily digoxin
cyanosis of cardiac origin usually worsens with crying, whereas therapy?
cyanosis due to a pulmonary cause may improve when the Results of most recent studies (chest radiograph,
infant cries.4 Cyanotic congenital heart defects with right-to- electrocardiogram, echocardiogram, and cardiac
left shunting will demonstrate a minimal improvement with catheterization)
supplemental oxygen, whereas cyanosis of a purely pulmonary When were the last studies performed, and what were
origin typically exhibits a significant improvement with sup- the results?
plemental oxygen (Table 169-1). Why were those studies performed (routine follow-up
studies or obtained because of decompensation from
baseline, or a planned evaluation for an upcoming
■  CLINICAL FEATURES AND DIAGNOSTIC surgical procedure)?
STRATEGIES: THE CARDIAC EVALUATION Surgical procedures
The key elements that should be elicited in the history of a Previous procedures and complications?
child with a known underlying cardiac disorder are listed in Any future planned procedures?
Box 169-3. Early consultation with the child’s cardiologist or
cardiac surgeon is extremely useful. The other important example, an infant who sweats during feeding may exhibit a
points to cover in the history of a child with a potential or splanchnic steal from anomalous coronary arteries, causing
suspected cardiac disorder are addressed in the subsequent transient ischemia, pain, color change, and diaphoresis that
sections of this chapter. resolve after eating.1 A child with an undiagnosed congenital
In addition to a well-focused history and physical examina- heart defect may take longer to feed, frequently pausing to
tion, a chest radiograph and an ECG should be obtained on catch his or her breath, with subsequent poor weight gain and
any child with a known or suspected cardiac disorder. Other gradually increased work of breathing due to developing CHF
ancillary studies that can also be useful include an arterial and pulmonary edema. Respiratory tract infections are common
blood gas analysis, hemoglobin/hematocrit levels, digoxin during childhood and may cause an acute deterioration in a
levels in those patients on daily digoxin, serum electrolytes in child with an underlying cardiac disorder. In turn, children
those patients on daily diuretic therapy, and the 100% oxygen with congenital heart disease (CHD) with large left-to-right
(hyperoxia) challenge. shunts and increased pulmonary blood flow tend to have a
higher incidence of lower respiratory tract infections. Acute
History respiratory distress in these patients may be from a combina-
tion of pulmonary and cardiac factors (e.g., CHF).
The presence of certain maternal medical conditions have
been associated with a higher incidence of cardiac disorders. Chest Pain
For example, congenital heart blocks have been associated
with maternal systemic lupus erythematosus and other colla- The majority of pediatric chest pain cases are noncardiac in
gen vascular disorders; infants of diabetic mothers have a origin and benign in nature. Common causes include muscu-
higher incidence of cardiomyopathy. loskeletal or chest wall pain, costochondritis, asthma exacerba-
Infants with an underlying congenital heart disorder may tions, pneumonia, pleurisy, gastritis, and gastroesophageal
exhibit diaphoresis during feeds and poor weight gain second- reflux. An unusual cause of acute, nonradiating, left-sided
ary to congestive heart failure (CHF). The cause of the infant’s chest pain in the adolescent is referred to as the precordial catch
hypoxia—cardiac or pulmonary—may be ascertained by the syndrome (also known as Texidor’s twinge). The pain, which
age at onset and the events surrounding a change in color. For is located in the left periapical area of the chest wall, occurs
2141
suddenly, often is exacerbated during inspiration, generally is
Table 169-2 Pediatric
not reproducible with chest wall compression, and usually Vital Signs and Pertinent Formulas
resolves within a few minutes. Patients may state the pain took for Estimating Blood Pressure

Chapter 169 / Cardiac Disorders


their breath away or they were afraid to move; usually it is of
Simplified pediatric vital signs5:
short duration and not associated with dysrhythmias or other
sequelae. The cause is unknown and findings on physical HEART RATE RESPIRATORY RATE
examination, chest radiograph, ECG, and echocardiogram are AGE GROUP (BEATS/MIN) (BREATHS/MIN)
all normal in these patients.
Chest pain or syncope on exertion may be due to an under- Newborn to 1 yr 140 40
lying cardiac condition and deserves a more thorough investi- 1–4 yr 120 30
gation, especially if there is a positive family history of sudden 4–12 yr 100 20
unexplained death in young adulthood. Myocardial involve- >12 yr 80 15
ment secondary to drug abuse (e.g., cocaine, amphetamines, Formulas to calculate the estimated normal blood pressures in
crystal methamphetamine) should always be considered as a children ≥1 year of age:
potential cause in any adolescent patient who presents with
Estimated systolic blood pressure (SBP): [age in years × 2] +
chest discomfort or pain. Pulmonary embolism is a possible 90 mm Hg
cause of chest pain, especially in pregnant adolescent females
Estimated diastolic blood pressure: 2/3 × [estimated SBP]
or those who are taking oral contraceptive agents. The rare
Minimum acceptable SPBs for age:
though life-threatening condition of aortic dissection must
always be considered as a cause of chest pain in a patient with Newborn to 1 mo 60 mm Hg
physical examination stigmata that are suggestive of a collagen 1 mo to 1 yr 70 mm Hg
vascular disorder, such as Marfan’s syndrome. Patients with a 1–10 yr [age in years × 2] + 70 mm Hg
known congenital heart defect or an acquired cardiac disorder >10 yr 90 mm Hg
(e.g., Kawasaki disease, acute rheumatic heart disease, myo-
carditis, pericarditis, cardiomyopathy) who present with chest
pain also deserve a more thorough diagnostic evaluation.
clinically the possibility of a coarctation of the aorta. Even with
an appropriately sized cuff, the blood pressures in the thighs
Physical Examination can be 10 to 20 mm Hg higher than the blood pressures in the
General Appearance and Pulses upper extremities due to the lack of well-designed blood pres-
sure cuffs for the legs. Therefore, if the measured blood pres-
All four extremities should be palpated for the presence and sure in the lower extremities is lower than the blood pressures
quality of pulses. The brachial and femoral pulses are the in the upper extremities, coarctation of the aorta should be
easiest to feel in infants. Bounding pulses are typically present suspected. Pulse oximetry readings that are lower in the legs
in infants with a patent ductus arteriosus. Coarctation of the than in the upper extremities are also suggestive of either a
aorta should be suspected in any child with strong pulses in coarctation of the aorta or a right-to-left-shunt across a patent
the upper extremities but weak pulses in the lower extremi- ductus arteriosus.7
ties. The pulse may be weak and thready in all extremities in
a child who presents with CHF and shock. Cardiac Auscultation

Vital Signs and Blood Pressures The intensity and degree of splitting of the S2 heart sound
(which reflects closure of the pulmonic and aortic valves) is
A mild resting tachypnea or tachycardia may be the only clini- extremely important in a pediatric cardiologic evaluation. In
cal clue to an underlying cardiovascular disorder. Age-related normal children, both components (aortic closure and pul-
variables in heart rates, respiratory rates, and blood pressures monic closure) of S2 should be heard along the left upper
often serve as a source of frustration and confusion to those sternal border (the pulmonic area). A widely split and fixed S2
clinicians who do not manage pediatric patients on a routine suggests a physiologic problem resulting from either a constant
basis. Although there are numerous tables of pediatric vital volume overload to the right side of the heart (e.g., atrial
signs with variations based on sleep or awake states, one can septal defect) or a pressure overload to the right side of
easily recall a rough estimate of the normal pediatric heart the heart (e.g., pulmonic stenosis). The classic congenital
rates and respiratory rates based on a simplified table of pedi- heart defect that is associated with a widely split and fixed S2
atric vital signs (Table 169-2).5 The methods to calculate the is the atrial septal defect. The intensity of the S2 component
normal expected blood pressures and hypotensive blood pres- may be louder than normal in the child with pulmonary
sures are also listed in Table 169-2.6 hypertension.
An accurate blood pressure reading is accomplished by using The third heart sound (S3), which is best heard along the
a cuff that covers two thirds of the upper arm or thigh. A cuff lower left sternal border or the apex, can be a normal finding
that is too narrow will overestimate the patient’s true blood in children and young adults. S3 is produced by a rapid filling
pressure and a cuff that is too large will underestimate the true of the ventricles and is heard during early diastole, just after
blood pressure. Any child with a suspected cardiac disorder the S2 sound. A loud S3, however, is always pathologic and is
should have blood pressures measured in both arms. If the due to dilated ventricles due to volume overload (e.g., CHF
blood pressure in the left arm is significantly lower than the and large ventricular septal defects). The fourth heart sound
blood pressure in the right arm, a coarctation of the aorta (S4) occurs late in diastole, just before the S1 sound. The
proximal to the origin of the left subclavian artery should be finding of an S4 is due to a decrease in compliance of a stiff,
suspected. Blood pressures must also be measured in the hypertrophic ventricle.
thighs in any child with a suspected aortic coarctation or docu- Cardiac murmurs are produced by turbulent blood flow
mented hypertensive blood pressures in the upper extremi- through the heart. The presence of a cardiac murmur may not
ties. The mere presence of femoral pulses does not rule out be associated with an underlying cardiac defect, however. The
2142
Persistence of a systolic murmur in the pulmonic area beyond
A Pathologic Etiology of a Heart Murmur this period should raise the possibility of a pathologic pulmo-
BOX 169-4 Should Be Suspected with Any of the nary arterial stenosis.
PART V  ■  Special Populations / Section One • The Pediatric Patient

Following Criteria Another common innocent murmur in children is Still’s


murmur, which is a systolic murmur that typically occurs in
Diastolic murmurs children between 2 and 6 years of age. This systolic murmur
Systolic murmurs that are louder than a grade 3/6, is best heard along the left midsternal border. The distinctive
continuous or associated with a thrill quality of this murmur has been described as being “vibra-
Murmurs that are associated with abnormal heart sounds tory,” “musical,” “zippy,” and “twanging” and results from
(clicks, rubs, or gallop rhythms) turbulent flow. The distinct quality of this murmur helps to
Presence of cyanosis or respiratory distress distinguish Still’s murmur from a ventricular septal defect
Bounding pulses or weak pulses murmur, which has a harsher quality. The intensity of Still’s
Abnormalities on the electrocardiogram murmur can be increased due to fever, excitement, exercise,
An abnormal cardiac silhouette, abnormal pulmonary or anemia.
vascularity or cardiomegaly on the chest radiograph
The Hyperoxia Test
Auscultation Locations of Common Systolic The hyperoxia test is an important bedside diagnostic tool to
BOX 169-5 Murmurs in Children help differentiate between cardiac and pulmonary causes of
central cyanosis. This test consists of assessing the rise in arte-
Left upper sternal border (pulmonic area) rial oxygenation with the administration of 100% oxygen. An
Pulmonic valvular stenosis arterial blood gas is measured on room air (if tolerated) and
Atrial septal defects (due to an increased pulmonic flow) repeated after several minutes of high-flow oxygen (100% O2)
Innocent pulmonic ejection murmur is administered; the two blood gas analyses are then compared.
Neonate pulmonic flow murmur When the child is breathing high-flow oxygen, an arterial
Patent ductus arteriosus (a continuous, “machinery” oxygen partial pressure (Pao2) of greater than 250 mm Hg vir-
sounding murmur) tually excludes hypoxia due to CHD—a “passed” hyperoxia
Left lower sternal border test.8 An arterial O2 reading of less than 100 mm Hg (in a child
Innocent vibratory Still’s murmur without obvious pulmonary disease) is consistent with a right-
Ventricular septal defects to-left shunt and is highly predictive of CHD—a “failed”
Endocardial cushion defects hyperoxia test.8 Values between 100 and 250 mm Hg may indi-
Tetralogy of Fallot cate lesions with intracardiac mixing. Pulse oximetry is not an
Hypertrophic cardiomyopathy appropriate substitute for an arterial blood gas; it is not sensi-
Apex tive enough to determine “pass or fail” of the test, since a child
Innocent vibratory Still’s murmur breathing high-flow O2 and registering 100% on pulse oxime-
Mitral regurgitation try may actually have a Pao2 of anywhere from 80 to
Aortic stenosis 680 mm Hg.1 Prolonged administration of 100% oxygen may
Hypertrophic cardiomyopathy cause some theoretical problems, such as closing the ductus
Right upper sternal border (aortic area) arteriosus in those infants with critical left heart obstructions
Aortic stenosis or by causing pulmonary vasodilation (which would potentially
Coarctation of the aorta worsen pulmonary vascular congestion).3 However, oxygen
should not be withheld initially in critically ill infants; continu-
ous reassessment is crucial in the evaluation and management
location, intensity, quality, timing, and radiation of the murmur of children with suspected CHD.1
determine whether the murmur is suggestive of an underlying
cardiac pathologic condition. Although systolic murmurs can Arterial Blood Gases
be present without any underlying anatomic abnormalities,
diastolic murmurs are always considered pathologic in nature. Patients with CHF exacerbation can exhibit respiratory acido-
Some of the other criteria that would suggest an underlying sis (low pH and Paco2) in addition to a low Pao2. In contrast,
anatomic cardiac abnormality are listed in Box 169-4. Murmurs children with compensated cyanotic congenital heart defects
may be very difficult to appreciate in the noisy emergency may have a normal pH despite a low Pao2. Patients with con-
department setting and given the degree of tachycardia that is genital heart defects who are not experiencing respiratory
often present even in normal infants. However, the location of failure are unlikely to exhibit elevation in Paco2. Any cardiac
the murmur may be a valuable clinical tool in determining the condition that results in inadequate tissue perfusion (i.e., any
underlying anatomic origin of the murmur (Box 169-5). of the acyanotic congenital heart defects that manifest in
Murmurs without any underlying anatomic abnormalities or CHF) will exhibit a metabolic acidosis with or without a
hemodynamic significance are termed innocent or functional respiratory compensation.
murmurs. All innocent murmurs are associated with normal
ECGs and normal chest radiographs. Two of the most common Hemoglobin/Hematocrit Levels and Serum Electrolytes
innocent murmurs encountered in the pediatric population are
the neonatal pulmonic flow murmur (also known as the periph- The hemoglobin and hematocrit levels may reveal a compen-
eral pulmonic stenosis murmur) and Still’s murmur. The pul- satory physiologic elevation (i.e., polycythemia) in children
monic flow murmur of the neonate is due to the relatively thin with cyanotic congenital heart defects. Any concurrent medical
walls and angulation of the right and left pulmonary arteries illness or blood loss that produces an acute anemia could
at birth. This systolic murmur is best heard at the left upper potentially precipitate an acute deterioration by compromising
sternal border with radiation throughout the entire chest, the oxygen carrying capacity in these children with an underly-
axilla, and back. It usually disappears by 3 to 6 months of age. ing congenital heart defect. A hemoglobin or hematocrit level
2143
Tr

Chapter 169 / Cardiac Disorders


Tr
Aorta
SVC
Aor
RPA RPA
PT LPA

RA LAA
RV LPA
RV LV
IVC LV
IVC

A Posteroanterior B Lateral
Figure 169-2.  Diagrammatic representations of the anatomy of the chest radiograph. A, Normal heart in a young man. Posteroanterior projection.
Aor, aorta; IVC, inferior vena cava; LAA, left atrial appendage; LPA, left pulmonary artery; LV, left ventricle; PT, pulmonary trunk; RA, right atrium;
RPA, right pulmonary artery; RV, right ventricle; SVC, superior vena cava; Tr, trachea. B, Right lateral projection of a normal heart in a young man.
IVC, inferior vena cava; LPA, left pulmonary artery; LV, left ventricle; RPA, right pulmonary artery; RV, right ventricle; Tr, trachea.

would also be helpful in evaluating whether a child’s pallor is


due to CHF or anemia. Serum electrolytes may be helpful
when evaluating children with acute dysrhythmias or sus-
pected metabolic acidosis and those children who are on
chronic diuretic therapy.

Chest Radiograph
Three features of the chest radiograph (Fig. 169-2) that deserve
special attention are (1) the cardiac size (cardiothoracic ratio),
(2) the cardiac shape (silhouette), and (3) the degree of pul-
monary vascular markings. The easiest method to determine
the heart size in children is to determine the cardiothoracic
ratio, which is obtained by comparing the largest transverse
diameter of the cardiac shadow on the posteroanterior view of
the chest radiograph to the widest internal diameter (measured
from the inside rib margin to the widest point above the cos-
tophrenic angle) of the chest. The normal cardiothoracic ratio
in children is approximately 50%. The cardiothoracic ratio is
not very accurate in newborns and small infants, in whom a
good inspiratory view is rarely obtained.9 A cardiac silhouette
that is larger than normal may be due to a shunt lesion, CHF,
or a pericardial effusion.9 An enlarged heart shadow on a chest
radiograph more reliably reflects a problem with volume over-
Figure 169-3.  Thymic shadow demonstrating the “sail sign” along the
load rather than pressure overload. Problems with pressure right cardiac border (dotted line).
overload are better represented on the ECG.
The cardiac size can be falsely increased in infants due to
the presence of the thymus, which can be seen in the medi- The degree of pulmonary vascular markings is one of the
astinum on the chest radiograph from birth until about 5 years key factors to consider when working through the differential
of age. The thymic borders are typically wavy in appearance diagnosis of congenital heart defects. An increase in pulmo-
and sometimes can be seen as the classic “sail sign” along nary vascularity is present when the pulmonary arteries appear
the superior right heart border (Fig. 169-3). The thymic enlarged and extend into the lateral third of the lung fields or
shadow may not be radiographically visible in infants during if there is an increased vascularity to the lung apices. Another
times of physiologic stress but should reappear when the infant criterion that suggests an increased pulmonary vascularity is
recovers. when the diameter of the right pulmonary artery in the right
The three classic cardiac silhouettes seen in patients with hilum on the posteroanterior view of the chest is wider than
congenital heart defects are (1) the “boot-shaped” heart the internal diameter of the trachea. The differential diagnosis
of tetralogy of Fallot (Fig. 169-4), (2) the “egg-on-a-string of a cyanotic infant with decreased vascular markings includes
silhouette” of transposition of the great vessels, and (3) the tetralogy of Fallot, pulmonary atresia, or tricuspid atresia. The
“snowman-shaped” or “figure-of-8 heart” of total anomalous cyanotic infant with increased vascular markings may have
pulmonary venous return. transposition of the great vessels, total anomalous pulmonary
2144

Table 169-3 Normal Electrocardiogram Values (PR, QRS,


QTc, and QRS axes) in Infants and Children6
PART V  ■  Special Populations / Section One • The Pediatric Patient

PR INTERVAL QRS DURATION


AGE AVERAGE (UPPER LIMIT) AVERAGE (UPPER LIMIT)

0–1 mo 0.10 (0.12) 0.05 (0.07)


1 mo to 1 yr 0.10 (0.14) 0.05 (0.07)
1–3 yr 0.11 (0.15) 0.06 (0.07)
3–8 yr 0.13 (0.17) 0.07 (0.08)
8–12 yr 0.15 (0.18) 0.07 (0.09)
12–16 yr 0.15 (0.19) 0.07 (0.10)
Adult 0.16 (0.21) 0.08 (0.10)
The QTc interval should not exceed:
0.45 seconds in infants <6 months old
0.44 seconds in children and adolescents
Normal QRS axes in infants and children:

AGE MEAN DEGREES (RANGE)

1 wk to 1 mo +110 (+30 to +180)


1–3 mo +70 (+10 to +125)
3 mo to 3 yr +60 (+10 to +110)
>3 yr +60 (+20 to +120)
Adults +50 (−30 to +105)
Normal T wave axis in infants and children: (+) = upright T
Figure 169-4.  The classic boot-shaped heart of tetralogy of Fallot. wave and (−) = inverted T wave

LEADS I,
AGE LEADS V1 AND V2 LEAD AVF V5, AND V6
venous return, or truncus arteriosus. Increased vascular mark-
Birth to 1 day +/− + +/−
ings in an acyanotic infant are suggestive of an endocardial
cushion defect, ventricular septal defect, atrial septal defect, 1–4 days +/− + +
or a patent ductus arteriosus. 4 days to − − +
In a normal left-sided aortic arch, the aorta descends to the adolescent
left of the midline and displaces the tracheal air shadow slightly Adolescent to + + +
toward the right of midline above the level of the carina. In adult
contrast to this, the tracheal air shadow may be midline or
deviated toward the left in the presence of a right-sided aortic
arch.3 This finding is important to note, since a right-sided
aortic arch is found in up to 25% of the children with tetralogy
of Fallot.9 Rib notching secondary to increased collateral blood
flow along the intercostal vessels can sometimes be appreci- with right ventricular hypertrophy and right bundle branch
ated between the fourth and eighth ribs in older children with block. A “superior” QRS axis (0 to −180 degrees with an S
undiagnosed coarctation of the aorta but is rarely visualized in wave in aVF greater than the R wave) may be suggestive of
children with coarctation of the aorta who are younger than 5 an endocardial cushion defect or tricuspid atresia.
years.9 Some of the more common indications for obtaining an
ECG in a pediatric patient include chest pain, dyspnea,
Electrocardiogram syncope, palpitations, and suspected dysrhythmias. Other
indications for obtaining an ECG are in those children with
The ECG findings in infants and children can sometimes be known cardiac disorders who present with signs and symptoms
problematic because various components of the ECGs change that could reflect an acute decompensation of their underlying
according to the child’s age (Table 169-3).6 At birth, the muscle disorder. A rare but potentially fatal congenital cardiac abnor-
mass of the right ventricle is greater than that of the left ven- mality that will demonstrate ECG abnormalities (i.e., ischemic
tricle; this is demonstrated by right axis deviation on the neo- changes) is the condition of the anomalous origin of the left
natal ECG. By the end of the first month of life, the left coronary artery. These infants have a history of poor feeding,
ventricle assumes dominance. By 6 months of age, the left irritability, and failure to thrive, then suddenly present with
ventricular to right ventricular mass ratio is 2 : 1, which then cardiogenic shock secondary to myocardial ischemia.
reaches the adult ratio of 2.5 : 1 by adolescence. The durations
of the PR interval, QRS complex, and QT intervals all increase Biochemical Markers
with age.
Left axis deviation is present when the QRS axis is less than The utility and clinical accuracy of cardiac biochemical markers
the lower limit of normal for the child’s age and occurs with such as creatinine phosphokinase MB and cardiac troponin-T
left ventricular hypertrophy and left bundle branch block. in the emergency department setting is currently limited in
Right axis deviation is present when the QRS axis is greater the pediatric population. Plasma homocysteine levels have
than the upper limit of normal for the child’s age and occurs been studied recently as a possible link to CHF in adults;
2145
however, there are currently no studies regarding plasma until about the 4th to 6th week of life when the left-to-right
homocysteine levels in pediatric cardiac disorders.9 Several shunt across the ventricular septal defect increases due to the
recent studies have evaluated the use of plasma B-type natri- decrease in the PVR. In general, the more severe the anatomic

Chapter 169 / Cardiac Disorders


uretic peptide levels in the assessment and management of defect is (i.e., lack of pulmonary blood flow or lack of systemic
CHF in adults.10 Studies of B-type natriuretic peptide levels blood flow), the earlier in life these conditions will manifest
in children have demonstrated a similar correlation of elevated with cyanosis and shock.
levels in children with CHF, and these also correlated to the Although CHD has been subdivided traditionally into cya-
clinical symptoms of heart failure and the ejection fraction as notic and acyanotic conditions, not all CHD lesions will fit
measured by echocardiography.11 The clinician is urged to neatly into a single category; some of the more complex defects
refer to the particular range of age-specific values from the have mixed pathophysiologic effects. Although the exact ana-
laboratory kit used at his institution. tomic diagnosis of a CHD is dependent on echocardiography
or cardiac catheterization, establishing the exact anatomic
■  SPECIFIC DISORDERS diagnosis is not entirely necessary in the emergency depart-
ment setting.
Congenital Heart Disease
Perspective Diagnostic Strategies
The incidence of CHD in the United States has remained The emergency physician must rely on several key elements
fairly constant at approximately 1%, or 8 to 10 cases per 1000 of the clinical examination in addition to chest radiograph and
live births. This equates to approximately 32,000 infants born ECG findings (Box 169-6) to narrow the diagnostic possibili-
each year with some form of CHD12 (Table 169-4). Although ties. For example, using the data in Box 169-6, the presence
a large percentage of CHD is now detected with prenatal of cyanosis, a grade 3/6 systolic-ejection murmur best heard at
ultrasonograms, one recent study also recommended routinely the mid-left sternal border, a boot-shaped heart, and decreased
measuring pulse oximeter readings in all newborns prior to pulmonary blood flow on the chest radiograph with evidence
discharge from the nursery as an additional inexpensive screen- of right ventricular hypertrophy on the ECG suggest tetralogy
ing tool for CHD.13 of Fallot. Only a brief discussion of some of the more common
CHDs is presented in this chapter.
Clinical Features
Management
The age, severity of symptoms, and time of presentation of a
child with CHD vary depending on the specific defect, com- The majority of children who present to the emergency depart-
plexity and severity of the defect, and timing of the normal ment with shock due to dehydration and hypovolemia are
physiologic changes that occur as the fetal circulation transi- typically given intravenous fluids in 20 mL/kg boluses.
tions to that of a neonate (Table 169-5). The more severe or However, if a child with a suspected CHD presents to the
complex CHD lesions may not be clinically apparent immedi- emergency department in possible cardiogenic shock, one
ately after birth. However, as the ductus arteriosus begins to could consider using smaller 10 mL/kg boluses to prevent an
close in the first several weeks of life, cardiac defects with iatrogenic complication of fluid overload. Frequent reassess-
obstructive lesions of the pulmonary or systemic circulations ment should be performed and the child’s response after each
will be unmasked, and these infants will present clinically with 10 mL/kg bolus monitored to determine whether additional
acute cyanosis, shock, or both. Even the harsh systolic murmur fluid boluses or inotropic agents are necessary.
of a large, isolated ventricular septal defect may not be heard

Table 169-5 Heart


Symptomatic Presentation of Congenital
Table 169-4 Incidence of Specific Congenital Heart Defects (CHDs) and Time of Presentation
Defects (CHDs)
DEFECT TIME OF PRESENTATION
DEFECT PERCENT OF CHD
CHDs That Present with Cyanosis
Acyanotic CHDs Transposition of the great arteries Birth to 2 wk
Ventricular septal defect 20–25%
Total anomalous pulmonary venous Birth to 2 wk
Atrial septal defect 5–10% return
Patent ductus arteriosus 5–10% Tricuspid atresia Birth to 2 wk
Coarctation of the aorta 8% Ebstein’s anomaly of the tricuspid Birth to 2 wk
Pulmonic stenosis 5–8% valve
Aortic stenosis 5% Truncus arteriosus Birth to 2 wk
Pulmonary atresia Birth to 2 wk
Cyanotic CHDs
Tetralogy of Fallot 10% Hypoplastic right heart syndrome Birth to 2 wk
Transposition of the great arteries 5% Hypoplastic left heart syndrome Birth to 2 wk
Tricuspid atresia 1–2% Tetralogy of Fallot Birth to 12 wk
Total anomalous pulmonary venous return 1% CHDs That Present with Shock
Truncus arteriosus <1% Coarctation of the aorta From 1st wk on
Pulmonary atresia <1% Aortic stenosis From 1st wk on
Ebstein’s anomaly <1% CHDs That Present with Congestive Heart Failure
Hypoplastic left heart syndrome <1% Ventricular septal defects From 4 wk on
Hypoplastic right heart syndrome <1% Patent ductus arteriosus From 4 wk on
2146
Clinical Clues to Aid in the Diagnosis of Ductal-Dependent Cardiac Lesions in
BOX 169-6 Congenital Heart Disease (CHD) BOX 169-7 the Neonate
PART V  ■  Special Populations / Section One • The Pediatric Patient

Presence or absence of central or peripheral cyanosis? Congenital heart diseases (CHDs) that require a patent
Central cyanosis with minimal respiratory distress ductus arteriosus to preserve blood flow from the aorta
(“comfortably blue”) is more suggestive of a CHD to the pulmonary circulation:
rather than a purely pulmonary etiology Tetralogy of Fallot
Abnormalities in cardiac auscultation? Tricuspid atresia
Murmurs: systolic versus diastolic, location, and radiation Pulmonary atresia
Quality of S1, S2, and the presence of any clicks or gallops Hypoplastic right heart syndrome
Change in the degree of central cyanosis with crying? Transposition of the great vessels
Worsening of cyanosis with crying suggests a cardiac CHDs that require a patent ductus arteriosus to preserve
rather than a purely pulmonary etiology blood flow from the main pulmonary artery to the
Response of the Pao2 to the hyperoxia challenge systemic circulation:
(administering 100% oxygen)? Severe coarctation of the aorta
Purely pulmonary causes of cyanosis: Severe aortic stenosis
Pao2 should rise to levels above 250 mm Hg Hypoplastic left heart syndrome
Cyanotic CHD associated with an increased pulmonary
blood flow:
Pao2 may occasionally reach as high as 150 mm Hg Prostaglandin E1 (PGE1) Infusion Preparation
Cyanotic CHD associated with a decreased pulmonary for Ductal-Dependent Congenital Heart
blood flow: BOX 169-8 Disease6
Pao2 will not rise over 100 mm Hg
Chest radiograph abnormalities? Infusion rate 0.05–0.1 µg/kg/min
Cardiac size and shape (one of the three classic cardiac Available PGE1 solution: 500 µg/mL (Alprostadil)—always
silhouettes)? follow the manufacturer’s directions.
Boot-shaped heart: tetralogy of Fallot (TOF) Add one ampule (500 µg) PGE1 to 250 mL of D5W. This
Egg-on-a-string silhouette: transposition of the great yields 2 µg/mL. To achieve 0.1 µg/kg/min, infuse at
vessels 0.05 mL/kg/min or 3 mL/kg/hr.
Snowman-shaped or figure-of-eight heart: total The above concentration of 2 µg/mL may be run as
anomalous pulmonary venous return (TAPVR) follows:
Degree of pulmonary blood flow?
Increased (acyanotic): atrial septal defect, Eisenmenger’s 2-kg infant 0.1 mL/min or 6 mL/hr to achieve
syndrome, ventricular septal defect, patent ductus 0.1 µg/kg/min of PGE1
arteriosus, endocardial cushion defects (ECDs) 2.5-kg infant 0.125 mL/min or 7.5 mL/hr to achieve
Increased (cyanotic): transposition of the great arteries 0.1 µg/kg/min of PGE1
(TGA), TAPVR, hypoplastic left heart syndrome, truncus 3-kg infant 0.15 mL/min or 9 mL/hr to achieve
arteriosus 0.1 µg/kg/min of PGE1
Decreased or normal (acyanotic): pulmonic stenosis (PS), 3.5-kg infant 0.175 mL/min or 10.5 mL/hr to achieve
aortic stenosis, coarctation of the aorta 0.1 µg/kg/min of PGE1
Decreased (cyanotic): TOF, severe PS, Ebstein’s anomaly, 4-kg infant 0.2 mL/min or 12 mL/hr to achieve
tricuspid atresia (TriA), pulmonary atresia, hypoplastic 0.1 µg/kg/min of PGE1
right heart syndrome 4.5-kg infant 0.225 mL/min or 13.5 mL/hour to achieve
Electrocardiographic abnormalities? 0.1 µg/kg/min of PGE1
Evidence of chamber enlargement: right ventricular 5-kg infant 0.25 mL/min or 15 mL/hour to achieve
hypertrophy, left ventricular hypertrophy, biventricular 0.1 µg/kg/min of PGE1
hypertrophy, right atrial hypertrophy, or left atrial From Siegfried BH, Henderson TO: Cardiology. In Gunn VL, Nechyba C
hypertrophy (eds): The Harriet Lane Handbook: A Manual for Pediatric House
An abnormal superior QRS axis is suggestive of ECD or Officers, 16th ed. Philadelphia, Mosby, 2002, p 123.
TriA

One unique pharmacologic intervention that can be life- adverse reaction of a PGE1 infusion is apnea; one
saving in infants involves the use of prostaglandin E1 (PGE1) should perform endotracheal intubation on these infants
to maintain the patency of the ductus arteriosus. CHD that prior to the initiation of the PGE1 infusion. Not only will
manifests within the first 2 to 3 weeks of life with a sudden intubation provide a secure airway, but controlled ventilation
onset of cyanosis or cardiovascular collapse is typically due to will also help decrease the infant’s work of breathing. Other
ductal-dependent cardiac lesions (Box 169-7).4 adverse reactions of a PGE1 infusion include fever, seizures,
Closure of the ductus arteriosus in patients with these spe- bradycardia, hypotension, flushing, and decreased platelet
cific cardiac lesions causes life-threatening situations due to aggregation.
either an interruption of blood flow to the lungs producing
cyanosis (i.e., tricuspid atresia) or a disruption of blood flow to Acyanotic Congenital Heart Defect
the systemic circulation producing shock (i.e., hypoplastic left
heart syndrome).4 Box 169-8 details one suggested method to Acyanotic CHD can be further subdivided (Fig. 169-5) into
prepare this potentially lifesaving PGE1 infusion. The PGE1 obstructive lesions (i.e., pulmonic stenosis, aortic stenosis, and
infusion is typically started at 0.05 to 0.1 µg/kg/min (the coarctation of the aorta) and lesions characterized by left-to-
method of preparation described in Box 169-8). A known right shunts with an associated increase in pulmonary blood
2147
Pulmonary blood flow Traditional closure of ventricular septal defects required
open heart surgery. Today, however, a transcatheter closure
technique that avoids the inherent risks and complications of

Chapter 169 / Cardiac Disorders


open heart surgery and cardiopulmonary bypass has supplanted
Increased Decreased or normal traditional methods.15–17
(left-to-right shunting) (obstructive lesions) Atrial Septal Defect
Perspective.  Atrial septal defects account for 5 to 10% of all
cases of CHD. The majority of infants and children with atrial
RVH LVH RVH LVH
septal defects remain clinically asymptomatic until adulthood.
Spontaneous closure has been reported in up to 40% of the
cases within the first 5 years of life.14
Clinical Features.  Large atrial septal defects, or those associated
ASD VSD PS AS with comorbid conditions such as bronchopulmonary dyspla-
ESM PDA CoA sia, can manifest with symptoms of CHF and pulmonary over-
ECD
circulation (e.g., dyspnea with feedings, poor weight gain, and
Figure 169-5.  Clinical clues to diagnosing acyanotic congenital heart frequent lower respiratory tract infections).3 The majority of
defects. AS, aortic stenosis; ASD, atrial septal defect; CoA, coarctation of atrial septal defects are discovered when a suspicious murmur
the aorta; ECD, endocardial cushion defect; ESM, Eisenmenger’s is detected on a routine physical examination. A widely split
syndrome; LVH, left ventricular hypertrophy; PDA, patent ductus
arteriosus; PS, pulmonic stenosis; RVH, right ventricular hypertrophy;
and fixed S2 is a characteristic finding of atrial septal defects.
VSD, ventricular septal defect. Diagnostic Strategies.  The chest radiograph of children with atrial
septal defects will reveal varying degrees of cardiomegaly,
right atrial and right ventricular enlargement, and a prominent
flow (i.e., ventricular septal defects, atrial septal defects, patent main pulmonary artery segment and increased pulmonary vas-
ductus arteriosus, and endocardial cushion defects). These cular markings. The ECG will reveal varying degrees of right
acyanotic lesions usually manifest within the first 6 months of axis deviation and right ventricular hypertrophy. All patients
life with symptoms of CHF; however, atrial septal defects can with unrepaired atrial septal defects will develop symptoms if
remain asymptomatic until adulthood. pulmonary hypertension ensues. Patients with large atrial
Ventricular Septal Defect septal defects that are not detected and repaired are at risk for
Perspective.  Ventricular septal defects are the most common development of Eisenmenger’s syndrome. Unlike ventricular
congenital cardiac defects and account for 20 to 25% of all septal defects, uncomplicated atrial septal defects are not asso-
cases of CHD. Spontaneous closure occurs in 30 to 40% of all ciated with high risk of bacterial endocarditis because of the
ventricular septal defects overall and in 50 to 70% of smaller lower turbulence and velocity of blood flow through the atrial
ventricular septal defects.14 septal defects.
Clinical Features.  The degree of symptoms is dependent on the Management.  Like ventricular septal defects, the traditional
size of the ventricular septal defects and the degree of PVR. closure of atrial septal defects required open heart surgery to
Most ventricular septal defects are clinically asymptomatic place a patch over the septal defect. Newer therapies involving
(minimal to no left-to-right shunting) immediately after birth closures with septal occluder devices placed via the transcath-
because of the high PVR. When the PVR decreases to the eter approach have been described.18,19 Antiplatelet therapy
normal levels at 6 to 8 weeks after birth, left-to-right shunting during the 6-month period after placement of the device is
can then occur and the typical systolic murmur of a ventricular typically given and is safe and effective in preventing throm-
septal defect will then be appreciated. Small ventricular septal bus formation on the surface of the septal occluder device.
defects may remain completely asymptomatic throughout Eisenmenger’s Syndrome.  Eisenmenger’s syndrome can occur in
childhood. Approximately 10% of the infants with large ven- any large left-to-right shunt defect that is not surgically cor-
tricular septal defects will eventually develop signs and symp- rected. When large left-to-right shunts are left untreated (i.e.,
toms of CHF (e.g., poor feeding and poor growth) by 2 to 3 large ventricular septal defects and atrial septal defects that
months of age because of the increased pulmonary blood flow. are not surgically corrected), irreversible changes can occur in
Older children with ventricular septal defects may exhibit the pulmonary arterioles, leading to pulmonary vascular
signs of decreased exercise tolerance and recurrent pulmonary obstruction and pulmonary hypertension. As the degree of
infections. If moderate to large ventricular septal defects are pulmonary hypertension increases, the PVR may then begin
not surgically corrected, irreversible changes in the pulmonary to exceed the SVR. This causes right-sided pressures to exceed
vasculature may begin to occur as early as 6 to 12 months of those on the left, causing right-to-left shunting. The reversal
age, which will result in an elevation of the PVR and pulmo- in the direction of shunt flow produces cyanosis. Clinical
nary hypertension. This in turn can lead to a reversal of the features of patients who have developed Eisenmenger’s
shunt direction across the ventricular septal defect to now syndrome include chest pain, dyspnea on exertion, and
become a right-to-left shunt, Eisenmenger’s syndrome, with hemoptysis.20
resultant cyanosis. Coarctation of the Aorta
Diagnostic Strategies.  The chest radiograph in children with small Perspective.  Coarctation of the aorta accounts for approximately
ventricular septal defects may be entirely normal, but cardio- 8% of all CHD, and up to 50% of patients with coarctation of
megaly with increased pulmonary vascular markings is usually the aorta also have an associated bicuspid aortic valve.21 The
present with untreated moderate to large ventricular septal area of coarctation can occur proximal to the insertion of the
defects. The ECG of moderate-sized ventricular septal defects ductus arteriosus (preductal type) or distal to the insertion of
typically reveals left ventricular hypertrophy, but biventricular the ductus arteriosus (postductal type). The majority of cases
hypertrophy may be present in ventricular septal defects with (89%) are of the postductal type.21
large left-to-right shunting. Clinical Features.  The severity of the symptoms and the age at
Management.  All ventricular septal defects, regardless of the time of presentation are dependent on the location of the
size of the defect, are at risk for bacterial endocarditis due to coarctation, the degree of narrowing, and the presence of any
the high velocity of turbulent blood flow through them. other associated cardiac defects. Infants with the preductal
2148
type of coarctation of the aorta may also exhibit differential Pulmonary blood flow
cyanosis if the ductus arteriosus remains open.22 The upper
half of the body is perfused with well-oxygenated blood sup-
PART V  ■  Special Populations / Section One • The Pediatric Patient

plied by the left ventricle and the ascending aorta. However,


the lower half of the body will appear cyanotic, as it is largely Increased Decreased
perfused via right-to-left shunting of deoxygenated blood from
the patent ductus arteriosus into the descending aorta.
Infants with the rarer preductal type of coarctation of the
aorta will present with signs of circulatory failure and shock RVH BVH RVH LVH
when the ductus arteriosus begins to close. Weaker pulses and
lower blood pressures in the lower extremities as compared
with the upper extremities are the classic physical examination TGA TruncA TOF TriA
findings in infants and children with coarctation of the aorta. TAPVR SV Severe PS PA
Most of the asymptomatic postductal cases of coarctation of HLHS EA HRHS
the aorta are diagnosed as a result of a cardiology referral for a Figure 169-6.  Clinical clues to diagnosing cyanotic congenital heart
systolic murmur or a hypertension workup, but infants with defects. BVH, biventricular hypertrophy; EA, Ebstein’s anomaly;
severe postductal coarctation of the aorta can also present HLHS, hypoplastic left heart syndrome; HRHS, hypoplastic right heart
during the first few weeks of life with signs of circulatory syndrome; LVH, left ventricular hypertrophy; PA, pulmonary atresia;
failure and shock. If a child is discovered to have hypertension PS, pulmonary stenosis; RVH, right ventricular hypertrophy; SV, single
on a routine physical examination, it is mandatory to obtain ventricle; TAPVR, total anomalous pulmonary venous return;
TGA, transposition of the great arteries; TOF, tetralogy of Fallot; TriA,
blood pressure measurements in the lower extremities to tricuspid atresia; TruncA, truncus arteriosus.
assess the possibility of coarctation of the aorta. A systolic
blood pressure in the right arm that is 15 to 20 mm Hg greater
than that in the legs is sufficient evidence to suspect coarcta-
tion of the aorta because the systolic blood pressures in the aortic arch (25% of patients), atrial septal defect (10% of
legs are normally higher than that in the arms.22 If the systolic patients), and anomalous origin of the left coronary artery.23
pressure in the right arm is higher than that in the left arm, Tetralogy of Fallot arises from a single embryologic defect in
the area of coarctation is probably preductal and located proxi- which the subpulmonic conus fails to expand, resulting in the
mal to the origin of the left subclavian artery. four abnormalities (Fig. 169-7): (1) right ventricular outflow
Diagnostic Strategies.  The chest radiograph will most often reveal tract obstruction; (2) a large, unrestrictive, malaligned ventric-
a normal-sized cardiac silhouette and normal pulmonary vas- ular septal defect; (3) an overriding aorta that receives blood
cular markings, but in children older than 5 years, it may flow from both ventricles; and (4) right ventricular hypertrophy
exhibit notching along the lower borders of the posterior fourth secondary to the high pressure load placed on the right ven-
to eighth ribs due to the pressure of the dilated collateral tricle by the right ventricular outflow tract obstruction. These
vessels. The absence of rib notching, however, does not rule anatomic defects collectively result in decreased pulmonary
out the possibility of coarctation of the aorta. The ECG typi- blood flow and varying degrees of right-to-left shunting of
cally reveals a left axis and left ventricular hypertrophy. deoxygenated blood across the ventricular septal defect.
Management.  Definitive surgical repair of coarctation of the Clinical Features.  The degree of cyanosis and the age of presen-
aorta involves resection of the narrowed section of the aorta tation are directly dependent on the degree of right ventricular
with an end-to-end anastomosis. Complications of undiag- outflow tract obstruction. Infants with tetralogy of Fallot typi-
nosed cases are related to the resultant hypertension and can cally have worsening of their cyanosis during crying and
include heart failure, hypertensive encephalopathy, and intra- feeding. Older children with tetralogy of Fallot may have cya-
cranial hemorrhages. notic exacerbations during periods of physical exertion. Infants
who have milder forms of right ventricular outflow tract
Cyanotic Congenital Heart Diseases obstruction may be acyanotic and are sometimes referred to as
having a “pink” tetralogy of Fallot. However, the majority of
Cyanotic CHDs are a result of either decreased pulmonary the cases of tetralogy of Fallot exhibit some degree of cyanosis.
blood flow to the lungs or right-to-left shunting of desaturated Infants with severe right ventricular outflow tract obstruction
blood directly into the systemic circulation. These cyanotic exhibit profound cyanosis within the first few days of life and
CHDs can be further subdivided into those conditions with may even require a PGE1 infusion to preserve pulmonary
an increased pulmonary blood flow and those lesions with blood flow via left-to-right shunting from the aorta into the
decreased pulmonary blood flow (Fig. 169-6). The classic cya- main pulmonary artery via the patent ductus arteriosus.
notic CHD can be remembered by the “five T’s”: truncus The physical examination can reveal varying degrees of
arteriosus, transposition of the great vessels, tricuspid atresia, cyanosis and a systolic ejection murmur along the left sternal
tetralogy of Fallot, and total anomalous pulmonary venous border. Chronic hypoxemia results in a compensatory polycy-
return. Other forms of cyanotic CHD include Ebstein’s themia and varying degrees of clubbing of the fingers and
anomaly, pulmonary atresia, severe pulmonary stenosis, hypo- toes.
plastic left heart syndrome, and hypoplastic right heart syn- Diagnostic Strategies.  The chest radiograph of a patient with cya-
drome. Because many of these cyanotic heart lesions are notic tetralogy of Fallot (see Fig. 169-4) reveals decreased
usually detected either on prenatal ultrasonographic examina- pulmonary vascular markings and a boot-shaped heart (sec-
tions or in the nursery, only tetralogy of Fallot is covered in ondary to a concave main pulmonary artery segment along the
this section. superior aspect of the left heart border). The heart size in
Tetralogy of Fallot tetralogy of Fallot is normal, and a right-sided aortic arch may
Perspective.  Tetralogy of Fallot accounts for approximately be seen in 25% of the cases. The ECG of cyanotic tetralogy
10% of all cases of CHD and is the most common cause of of Fallot reveals right ventricular hypertrophy and a right axis
cyanotic CHD beyond infancy. Tetralogy of Fallot is often deviation. Children with “pink” tetralogy of Fallot may not
associated with other cardiac defects, such as a right-sided initially exhibit any degree of right ventricular hypertrophy,
2149
Acute decrease in SVR
(e.g., crying)

Chapter 169 / Cardiac Disorders


Increased right-to-left shunting of
deoxygenated blood across the VSD

Decreased arterial PaO2


Increased arterial PCO2
Decreased arterial pH

Stimulation of hyperpnea
OAo (deep and rapid breathing)

OB
VSD
Increased negative intrathoracic
pressures with a resultant increase
of systemic venous return to the
right side of the heart

Figure 169-8.  Pathophysiologic mechanisms of a hypoxic (tet) spell.


SVR, systematic vascular resistance; VSD, ventricular septal defect.

systemic venous blood return to the right side of the heart.


This increased volume of blood in the right ventricle is then
Figure 169-7.  Diagrammatic representation of the right-to-left shunting shunted through the ventricular septal defect through the
that occurs in tetralogy of Fallot. Some of the deoxygenated blood (thick combination of the existing right ventricular tract outflow
blue arrow) in the right ventricle is shunted across the ventricular septal obstruction and the acute decrease in the SVR. This in turn
defect (VSD) into the left ventricle. This deoxygenated blood mixes with further decreases the arterial oxygen saturation, perpetuating
the well-oxygenated blood from the lungs (red arrow). The blood that is
ejected out through the overriding aorta (OAo) therefore contains blood
the hypoxic spell (Fig. 169-8).
of mixed oxygenation (purple arrows). The amount of deoxygenated Clinically these hypoxic spells are characterized by periods
blood that is shunted through the VSD (thick blue arrow) is dependent on of hyperpnea (rapid and deep respirations), uncontrollable
a combination of factors, including the severity of right ventricular crying, and worsening cyanosis. Limpness, seizures, cerebro-
outflow tract obstruction (OB), the size of the VSD, and the degree of vascular accidents, and even death have been reported with
systematic vascular resistance (SVR). When the SVR falls (as occurs during more severe tet spells. During a tet spell, the intensity of the
a tet spell), more deoxygenated blood from the right ventricle will be
shunted across the VSD into the systemic circulation, which results in
murmur decreases because of less blood flow through the right
hypoxia, metabolic acidosis, and worsening cyanosis. ventricular tract obstruction and more blood being shunted
from the right ventricle to the left ventricle through the ven-
tricular septal defect.
Management.  The overall treatment goals for tet spells (Box
but these acyanotic forms of tetralogy of Fallot gradually 169-9) are: (1) increasing the SVR, (2) abolishing the hyper-
develop the cyanotic form by 1 to 3 years of age. pnea, and (3) correcting the metabolic acidosis. Although
A potentially life-threatening complication of tetralogy of supplemental oxygen should be provided, this alone will not
Fallot that can be seen in a patient who presents to the emer- reverse a tet spell, since there is a decrease in the amount of
gency department is the so-called “tet spell,” which has also
been referred to as the “hypercyanotic spell” or the “hypoxic
spell.” Although these hypoxic spells can occur in children Management of Tetralogy of Fallot
with other forms of CHD, they are most commonly seen in BOX 169-9 Hypoxic Spells
infants and children with tetralogy of Fallot, and hence the
term tet spells. These episodes occur most commonly in infants, Place the infant in the knee-to-chest position to increase
with a peak incidence between 2 and 4 months of age.23 the SVR, which decreases the right-to-left shunt across
Any event that suddenly lowers the SVR, such as crying or the VSD.
defecation, will produce a large right-to-left shunt across the Provide supplemental oxygen (limited value by itself ).
ventricular septal defect, beginning the vicious cycle of a Morphine: 0.1–0.2 mg/kg intravenously (IV) or
hypoxic spell. Acute hypovolemia and tachycardia can also intramuscularly (IM)
precipitate tet spells. The large right-to-left shunt through the Fentanyl: 1 µg/kg/dose IV or IM as an alternative to
ventricular septal defect bypasses the lungs, which then causes morphine
a decrease in the Pao2, an increase in the Pco2, and a fall in Sodium bicarbonate: 1 mEq/kg IV
the arterial pH. These metabolic changes then stimulate the Consider ketamine: 1–2 mg/kg IV or IM
respiratory centers in the brain to produce hyperpnea (deep Consider propranolol6: 0.01–0.2 mg/kg IV
and rapid respirations), which increases the negative intratho- Consider phenylephrine6: 0.01–0.02 mg/kg IV
racic pressure during inspiration, causing an increase in the
2150
pulmonary blood flow and an increase in the amount of right- flow through a surgical conduit that was created to provide an
to-left shunting across the ventricular septal defect. The infant improvement in blood flow to the pulmonary system.25 Com-
should be picked up and placed in a knee-to-chest position. parison of the child’s other postoperative chest radiographs can
PART V  ■  Special Populations / Section One • The Pediatric Patient

Older children can be placed in the squatting position. Both help to determine whether there has been a change in the
maneuvers are believed to increase the SVR and decrease the heart size and pulmonary vascularity.
amount of systemic venous blood return to the right side of The postpericardiotomy syndrome is an inflammatory peri-
the heart. Morphine (0.1–0.2 mg/kg) has been traditionally carditis that can occur 1 to 6 weeks after any surgical procedure
given intramuscularly to suppress the respiratory center and that involved a pericardiotomy. This immunologic phenome-
thereby abolish the hyperpnea. A theoretical adverse effect of non is believed to occur as a sequela of blood in the pericardial
morphine, however, is that it can cause systemic vasodilation sac. This syndrome is characterized by fever, chest pain, and
(further decreasing the SVR) via endogenous histamine a pericardial effusion. A pericardial friction rub may be heard,
release.24 Although there are no current studies evaluating the depending on the amount of fluid that accumulates in the
use of other medications that may also suppress the respiratory pericardial sac. The chest radiograph may reveal an enlarged
centers, fentanyl and midazolam could be utilized for the same cardiac silhouette, and the echocardiogram will confirm the
effect without the potential risk of endogenous histamine diagnosis. Pericardiocentesis is rarely required but may be
release. Ketamine (1–2 mg/kg intravenously or intramuscu- necessary if the amount of pericardial effusion is significant
larly) has also been suggested for its sedative effect as well as enough to cause a pericardial tamponade. The majority of
for its effect on increasing the SVR.23 Sodium bicarbonate can cases will resolve within 2 to 3 weeks with bedrest and non-
be given to correct any metabolic acidosis and reduce the steroidal anti-inflammatory medication.
respiratory center–stimulating effects of acidosis. Most infants Many of the above-mentioned postoperative complications,
respond to these measures and exhibit an improvement in including the postpericardiotomy syndrome, can be avoided in
their oxygenation and a decrease in their degree of cyanosis. children who undergo closure of patent ductus arteriosus,
Those infants whose condition does not improve with the atrial septal defects, and ventricular septal defects via the
above measures may require a vasopressor such as phenyleph- transcatheter placement of various occluder devices.
rine to increase the SVR and thereby decrease the degree of
right-to-left shunting across the ventricular septal defect. An Respiratory Syncytial Virus Infections in Infants and Children
intravenous fluid bolus may also be considered to increase the with Congenital Heart Defects
volume of blood flow through the pulmonary artery. Proprano-
lol has also been used as an adjunct to break the cycle of a tet Respiratory syncytial virus (RSV) is the most common cause
spell. Although the exact pharmacophysiologic mechanisms by of lower respiratory tract infections in infants and children
which propranolol accomplishes this is uncertain, it is thought worldwide, with the majority of children being infected at
to increase the SVR and perhaps promote an increase in the least once by 2 years of age. Reinfection occurs commonly
pulmonary blood flow by reducing spasms of the right ven- throughout life.26 RSV lower respiratory tract infections account
tricular outflow tract obstruction. for more than 125,000 pediatric admissions annually in the
Palliative surgical procedures to increase the amount of United States, with a fatality rate of 6.3 deaths per 100,000
blood flow temporarily to the pulmonary arteries are performed patients up to 4 years of age (Box 169-10).27,28 Children with
in infants with severe cyanotic tetralogy of Fallot. The most CHD who develop RSV infections tend to have a higher rate
commonly performed procedure is the modified Blalock- of intensive care unit admissions and require mechanical ven-
Taussig shunt, in which an anastomosis is created between the tilation more frequently than those children who do not have
subclavian artery and the ipsilateral pulmonary artery. Defini- CHD. Children with CHD who require hospitalization for
tive surgical repair of tetralogy of Fallot consists of closing the RSV have a fatality rate that is two to six times greater than
ventricular septal defect and opening the right ventricular that of children without CHD.27,28 RSV is responsible for a
outflow tract obstruction by resection of the infundibular mortality rate of 40% in infants with CHD and up to 70% in
tissue. The mortality rate is 5 to 10% within the first 2 years infants with CHD and associated pulmonary hypertension.
after definitive surgical repair in uncomplicated tetralogy of Currently there are two products that can be used to prevent
Fallot cases. Complications that can occur after definitive sur- RSV infections. Both of these preparations require monthly
gical repair include complete heart block, ventricular dys- administration prior to the onset of the peak RSV season,
rhythmias, and right bundle branch block (secondary to the which typically runs from November through March in most
right ventriculotomy). Bacterial endocarditis prophylaxis is states. The two currently available preparations are (1) palivi-
still recommended after the definitive surgical repair of tetral- zumab (Synagis), which is a humanized monoclonal antibody
ogy of Fallot. administered intramuscularly at a dose of 15 mg/kg and (2)
RSV immunoglobulin (RespiGam), which requires intrave-
Postoperative Complications of Congenital Heart Defects nous administration at a dose of 750 mg/kg (which is equal to
15 mL/kg) and must be administered over a 4-hour period.26
A variety of postoperative complications can be seen in patients The use of palivizumab in the prevention of RSV infections
who present to the emergency department weeks to months in high-risk infants and children has largely replaced RSV
after cardiac surgery. The types of complications that could
occur in each case depend on the original underlying cardiac Conditions Associated with an Increased Risk
defect as well as on the surgical procedure that was used to BOX 169-10 of Severe or Fatal Respiratory Syncytial
correct that defect. Some of the complications that may be Virus Infections
seen in the emergency department include thrombosis of a
shunt-conduit with decreased flow, increased shunt-conduit Cyanotic or complex congenital heart defects
flow with resultant CHF, atrial and ventricular dysrhythmias, Pulmonary hypertension
heart blocks, myocardial ischemia, and endocarditis. The size Prematurity (especially those infants with
of the cardiac silhouette and the degree of pulmonary blood bronchopulmonary dysplasia or chronic lung disease)
flow visualized on the chest radiograph may provide valuable Immunodeficiency states
clues as to whether there is an increased or decreased blood
2151
cardiac toxins, and dysrhythmias that compromise cardiac
Recommendations for Palivizumab output.
BOX 169-11 Administration in Children with Congenital CHF can result from a derangement in one of the four

Chapter 169 / Cardiac Disorders


Heart Disease (CHD) primary determinants of normal cardiac function: (1) excessive
preload (e.g., large left-to-right shunts and severe chronic
CHD conditions that would benefit from palivizumab anemia); (2) decreased cardiac contractility (e.g., myocarditis);
administration (15 mg/kg intramuscularly every 30 days (3) excessive afterload (i.e., left-sided obstructive lesions); and
during the respiratory syncytial virus season only): (4) rhythm abnormalities that compromise cardiac output or
Cyanotic or complex CHD stroke volume (e.g., paroxysmal supraventricular tachycardia
Congenital heart failure requiring medications and severe forms of heart block). The treatment of CHF
Moderate to severe pulmonary hypertension depends on which of these four primary determinants of
Children with hemodynamically insignificant CHD who do normal cardiac function are compromised. For example, ino-
not require palivizumab administration: tropic agents and diuretics may be required in a child with
Secundum atrial septal defect volume overload and decreased cardiac contractility, whereas
Small ventricular septal defect vasodilating agents may be required in a child with CHF due
Pulmonic stenosis to an increased afterload.
Uncomplicated aortic stenosis
Mild coarctation of the aorta
Patent ductus arteriosus Clinical Features
Although the clinical manifestations of CHF depend on the
exact pathophysiologic cause of the CHF, common presenting
immunoglobulin because palivizumab can be given intramus- signs and symptoms include tachycardia, a gallop rhythm,
cularly, requires a 100-fold lesser volume, and has a 50-fold tachypnea with rales, hepatomegaly, peripheral edema, and
smaller protein load than RSV immunoglobulin.29 decreased peripheral perfusion of the extremities. Wheezing
In 1998, the Food and Drug Administration gave approval and a chronic cough may also be the presenting symptoms of
for the use of palivizumab in preventing RSV lower respiratory CHF.
tract infections in selected infants and children who were
deemed to have a higher risk of severe infections. During the Diagnostic Strategies
4-year period from 1998 to 2002, several multicenter studies
involving more than 24,000 subjects (including data from the The chest radiograph typically reveals an enlargement of the
Palivizumab Outcomes Registry) have demonstrated the effi- cardiac silhouette and varying degrees of pulmonary conges-
cacy of palivizumab in preventing hospitalizations of high-risk tion. An echocardiogram will be able to assess the ejection
infants and children.30 During this same 4-year period, another fraction as well as to identify underlying anatomic defects.
multicenter study involving six countries demonstrated a 45% Plasma B-type natriuretic peptide has been reported to be
relative reduction in hospitalization of 1287 high-risk children helpful in differentiating cardiac from pulmonary etiologies of
younger than 2 years with CHD who were given 15 mg/kg dyspnea in children.31 Other diagnostic studies to consider are
monthly intramuscular injections of palivizumab over a 5- case specific and depend on the suspected cause of the child’s
month period.29 This study also demonstrated a significant CHF.
reduction in the total number of hospital days, supplemental
oxygen requirement, total number of days in the intensive care Management
unit, and total number of days requiring intubation in the
group that received the monthly palivizumab intramuscular Acute stabilization of any child who presents with CHF
injections. Based on these results, the Food and Drug Admin- includes administration of supplemental oxygen and agents to
istration in September 2003 gave approval for the use of palivi- augment cardiac contractility and to improve cardiac output.
zumab in infants and children with hemodynamically significant Children who present in severe respiratory distress secondary
CHD (Box 169-11). The American Academy of Pediatrics to pulmonary edema may require intubation to support oxy-
emphasizes that prophylaxis with palivizumab should be initi- genation and ventilation. Children with respiratory distress
ated prior to the onset of the RSV season. Because RSV can and air hunger due to pulmonary congestion may also benefit
persist on environmental surfaces for several hours, the best from elevation of the head and upper torso in addition to
method to prevent the spread of RSV-contaminated secretions morphine sulfate (0.05–0.1 mg/kg/dose) administration. Con-
within the emergency department setting is meticulous hand tinuous positive airway pressure or biphasic positive airway
washing and wearing a mask when caring for children with a pressure ventilation may be useful initially to avert the need
documented or suspected RSV infection. for endotracheal intubation. Plasma B-type natriuretic peptide
levels have been shown to be elevated in children with CHF
and have also been used to monitor the response to treatment
Congestive Heart Failure regimens in patients with CHF.11
Perspective Diuretics and inotropic agents are the mainstay of treatment
for the majority of children with CHF. Furosemide (Lasix) in
Congestive heart failure is defined as a clinical syndrome in a dose of 1 mg/kg is the most common loop diuretic used to
which the cardiac output is unable to meet the hemodynamic increase renal perfusion and improve urine output. Digoxin
and metabolic demands of the body. Although there is a wide has remained the most widely used inotropic agent to treat
array of causes of CHF, the primary cause in infants and chil- CHF in children (Table 169-6). With the proper use of furo-
dren is CHD, which results in volume or pressure overload. semide and digoxin, most children with CHF will demonstrate
Other causes of CHF include the anomalous left coronary a favorable response. The narrow therapeutic index of digoxin
artery in infants, myocarditis, endocarditis, rheumatic heart requires that levels be monitored very closely to prevent iat-
disease, pericardial effusions, anemia, cardiomyopathies, sys- rogenic digoxin toxicity, which could cause a worsening of the
temic hypertension, hypothyroidism, electrolyte imbalances, preexisting CHF.
2152
direct effect on the enhancement of coronary blood flow. The
Table 169-6 Digoxin Dosing for Infants and Children with main toxicities of dobutamine are tachycardia, ventricular
Congestive Heart Failure6 ectopy, and hypotension.
PART V  ■  Special Populations / Section One • The Pediatric Patient

Epinephrine is a potent inotrope and chronotrope and also


TOTAL DIGITALIZING DAILY MAINTENANCE
AGE DOSE (ORAL)* DOSE (ORAL) increases the SVR. Epinephrine is useful in scenarios in which
poor cardiac output is also associated with diminished systemic
Premature 20 µg/kg/24 hr 5 µg/kg/day vascular tone. Toxicities associated with epinephrine include
Full term 30 µg/kg/24 hr 8–10 µg/kg/day tachydysrhythmias, severe hypertension, hyperglycemia, lactic
<2 yr 40–50 µg/kg/24 hr 10–12 µg/kg/day
acidosis, and hypokalemia.
Amrinone and milrinone are newer inotropic agents that also
2–10 yr 30–40 µg/kg/24 hr 8–10 µg/kg/day
have peripheral vasodilatory effects. These agents have been
>10 yr 0.75–1.25 mg/24 hr 0.125–0.25 mg/day
used in improving cardiac index in septic shock and in preven-
*The intravenous dose of digoxin is equal to 75% of the oral dose (except in tion of low cardiac output states for children with CHD. Side
children >10 years old, in whom the intravenous dose is the same as the oral effects of these medications include profound hypotension,
dose).
The total daily digitalizing dose (TDD) is given as follows: 12 of the TDD given
dysrhythmias, hypersensitivity reactions, fever, hepatotoxic-
initially, followed by 1 4 of the TDD given every 8–12 hours × 2 doses after ity, and thrombocytopenia.
the initial dose. Another vasodilating medication that has been used for arte-
The daily maintenance dose is divided into a twice-daily dosing regimen rial and venous dilatory effects is nitroprusside. Nitroprusside
(except in children >10 years of age, in whom the maintenance dose can be has potent vasodilatory effects on both the systemic and pul-
given as a single daily dosing regimen).
Intravenous digoxin is supplied as 100 µg/mL and 250 µg/mL solutions. monary circulatory systems. This medication has a prompt
Oral digoxin suspension is supplied as a 50 µg/mL elixir. onset of action and a short duration of action. Nitroprusside
Therapeutic range: 0.8–2.0 µg/L. must be used cautiously in patients with either hepatic and/or
A lower dosing regimen may be required in those patients with renal failure renal impairment to avoid cyanide toxicity (severe metabolic
since digoxin is excreted by the kidneys.
acidosis and coma) or thiocyanate toxicity (irritability, seizures,
abdominal pain, and vomiting).

Other inotropic agents that are used for the treatment of


CHF in infants and children are dopamine, dobutamine, and Pediatric Dysrhythmias
epinephrine (Table 169-7). Standardized drips have been Perspective
established and have essentially replaced the “rule of six.”32
Dopamine is an endogenous catecholamine with complex car- Dysrhythmias are not as common in children as they are in
diovascular effects. Dopamine can increase renal blood flow in adults. The most common cause of cardiopulmonary arrest in
low doses (2–5 µg/kg/min) and will increase the cardiac con- infants and children is the untreated progression of respiratory
tractility and heart rate in moderate doses (5–10 µg/kg/min). failure or shock rather than a primary cardiac dysrhythmia.33
Dopamine stimulates cardiac beta1-adrenergic receptors both Therefore, the most common arrest rhythm that will confront
directly and indirectly through the release of endogenous nor- the emergency physician will be asystole or bradycardia rather
epinephrine stored in the cardiac sympathetic nerves. Due to than ventricular fibrillation or ventricular tachycardia. When
this, some of the inotropic effects of dopamine may be reduced confronted with a child with a primary cardiac dysrhythmia,
in those patients with decreased endogenous myocardial nor- the physician must identify and treat the underlying cause
epinephrine stores (i.e., patients with chronic CHF and neo- quickly and systematically. Children who are at risk for devel-
nates). At higher doses (10–20 µg/kg/min), dopamine will also oping dysrhythmias are listed in Box 169-12. Various medica-
increase the SVR, but excessive vasoconstriction may compro- tions, drugs, and toxins can also precipitate dysrhythmias in
mise end-organ perfusion at infusion rates greater than children. Even those medications that are used to treat under-
20 µg/kg/min. If additional inotropic effects are required, lying cardiac problems such as digoxin, amiodarone, and pro-
the addition of either dobutamine or epinephrine infusions cainamide can themselves precipitate dysrhythmias. Drugs of
may be preferable to increasing the dopamine infusion greater abuse (e.g., cocaine and crystal methamphetamine) and over-
than 20 µg/kg/min. The major toxicities of dopamine are dose of prescription medications (e.g., cyclic antidepressants)
tachycardia, vasoconstriction, and ventricular ectopy. should always be considered when evaluating any previously
Dobutamine is a synthetic catecholamine with more selec- healthy adolescent patient who presents with an acute
tive cardiac inotropic effects and some beta2-adrenergic vaso- dysrhythmia.
dilatory effects. It is not a vasopressor per se, but could be a Pediatric dysrhythmias can be divided into three broad cat-
good adjunct in the treatment of low cardiac output states that egories of rhythms based on their effect on the child’s pulse:
are secondary to poor myocardial function. Dobutamine may slow (sinus bradycardia and heart blocks), fast (supraventricular
have an advantage over dopamine in that it has fewer arrhyth- tachycardia or ventricular tachycardia with a pulse), or absent
mogenic effects than dopamine and may also have a more (ventricular tachycardia without a pulse, ventricular fibrilla-

Table 169-7 Inotropic and Load-Altering Agents Used in the Treatment of Congestive Heart Failure
INOTROPE CHRONOTROPE VASCULAR EFFECTS INFUSION RANGES

Dobutamine + +/− Vasodilator 2–20 µg/kg/min


Dopamine + + Pressor 2–20 µg/kg/min
Epinephrine + + Pressor 0.1–1.0 µg/kg/min
Milrinone + No Vasodilator 0.5–2 mg/kg bolus over 5 minutes followed by 5–10 µg/kg/min
Nitroprusside No No Potent vasodilator 0.5–10 µg/kg/min
2153
dysrhythmias, including supraventricular tachycardia and
Conditions Associated with a High Risk of ventricular tachycardia.35,36
BOX 169-12 Developing Dysrhythmias

Chapter 169 / Cardiac Disorders


Congenital heart defects (uncorrected defects and Bradydysrhythmias
postoperative complications) Sinus Bradycardia.  Bradycardia is defined as a heart rate that is
Congenital complete heart blocks (e.g., maternal systemic slower than the lower limit of normal for a child’s age. Based
lupus erythematosus) on the current definition by the American Heart Association
Myocarditis (AHA) guidelines in Pediatric Advanced Life Support (PALS),
Rheumatic heart disease clinically significant bradycardia in children is defined as a
Kawasaki disease with involvement of the coronary arteries heart rate slower than 60 beats per minute that is associated
Cardiomyopathy with poor systemic perfusion.34 Bradycardia is poorly tolerated
Prolonged QT syndrome in infants and children because they are not physiologically
Aberrant atrioventricular conduction pathways (e.g., Wolff- capable of increasing their stroke volume to maintain an ade-
Parkinson-White syndrome) quate cardiac output in the face of significant bradycardia.
Electrolyte abnormalities (e.g., potassium, calcium, and The most common cause for symptomatic bradycardia in
magnesium disturbances) infants and children is hypoxia. Therefore, the first step in the
Commotio cordis management of symptomatic bradycardia in children is to
Profound hypothermia ensure adequate oxygenation and ventilation before automati-
Hypoxia cally reaching for medications or pacing. Epinephrine is the
first-line medication when treating symptomatic bradycardia in
children that is not responsive to appropriate oxygenation and
ventilation. This is in contrast to the treatment of bradycardia
tion, pulseless electrical activity, or asystole).34 The most in adults, in which atropine is considered to be the first-line
common dysrhythmia in children is supraventricular tachycar- medication. If additional doses of intravenous or intraosseous
dia, which occurs most commonly in infants and young chil- epinephrine are required when treating symptomatic bradycar-
dren. Although supraventricular tachycardia can spontaneously dia, the dose should remain at standard dosing (0.01 mg/kg)
occur in infants without any underlying structural cardiac and not be increased to the high dose (0.1 mg/kg) according to
defects, ventricular tachycardias typically are due to an under- AHA and PALS guidelines. Atropine will have no effect on the
lying myocardial abnormality. denervated heart. If vascular access is not available, both epi-
nephrine and atropine can be administered via the tracheal
Clinical Features tube, although the intravenous route is preferred.
Other causes of bradycardia include hypothermia, increased
Rhythm disturbances in infants can manifest with symptoms intracranial pressure, heart blocks (congenital and acquired), a
such as fussiness, lethargy, poor feeding, pallor, respiratory denervated heart status post–cardiac surgery, hypothyroidism,
distress, or cardiogenic shock. Older children present with sick sinus syndrome, and various medications and toxins (e.g.,
chest pain, palpitations, difficulty breathing, or syncope. The digoxin, beta-blockers, calcium channel blockers, and cholin-
type and degree of severity of the presenting signs and ergic agents). One should consider emergency pacing for
symptoms must be taken into account when considering the Mobitz type II second-degree atrioventricular block, complete
evaluation and management of the specific dysrhythmia in third-degree heart block, or sick sinus syndrome.
each case. Athletic adolescent patients may have resting baseline heart
rates slower than 60 beats per minute and do not require
Management emergency treatment if they are completely asymptomatic.

Not every child who exhibits a rhythm disturbance requires Tachydysrhythmias


emergency intervention. Emergency treatment and stabiliza-
tion of any dysrhythmia depend on two key clinical questions: Supraventricular Tachycardia
(1) Does the child have a pulse? and (2) If a pulse is present, Perspective.  Supraventricular tachycardia is the most common
is it slow or fast and is the child hemodynamically stable or symptomatic dysrhythmia in infants and children.34 In children
unstable? Children who have a pulse with good perfusion younger than 12 years, the most common cause of supraven-
parameters (i.e., an alert child with strong distal pulses, warm tricular tachycardia is a reentry mechanism due to an accessory
extremities, and brisk capillary refill times) do not require atrioventricular pathway.37 No cardiac abnormalities are found
emergency intervention unless they exhibit a rhythm that has in approximately half of the cases, and the Wolff-Parkinson-
the potential to rapidly degenerate into a more serious condi- White syndrome is present in only 10 to 20%.38 The QRS
tion. Children who exhibit ECG evidence of conduction complex is narrow (<0.08 seconds) in up to 90% of the cases
abnormalities (e.g., Mobitz type II second-degree heart blocks, of pediatric supraventricular tachycardia.39 The type of supra-
complete heart blocks, prolonged QT intervals, or aberrant ventricular tachycardia that occurs most commonly in infants
conduction such as the Wolff-Parkinson-White syndrome) and children involves a reentrant mechanism that utilizes an
may also warrant more emergency treatment. accessory pathway and the atrioventricular node. The ortho-
A summary of the treatment algorithms for pediatric dromic reentry phenomenon involves the normal antegrade
dysrhythmias and the most commonly used medications conduction from the atria to the ventricles down the atrioven-
in treating these dysrhythmias are listed in Box 169-13. tricular node with retrograde conduction back from the ven-
Although some medications can only be used to treat atrial tricles to the atria via the accessory pathway. Orthodromic
tachycardias (e.g., the use of adenosine for supraventricular conduction will produce a narrow QRS complex supraventric-
tachycardia) or ventricular tachycardias (e.g., the use of lido- ular tachycardia. The less common form of reentry mechanism
caine for ventricular tachycardia), amiodarone and procain- is the antidromic form in which conduction from the atria to
amide can be used for a wide array of both atrial and ventricular the ventricles first goes antegrade down the accessory pathway
2154
Summary of Pediatric Dysrhythmia Treatment Options and Defibrillation, Cardioversion, and
BOX 169-13 Medication in Resuscitation
PART V  ■  Special Populations / Section One • The Pediatric Patient

Simplified Summary of Dysrhythmia Treatment Options Defibrillation, Cardioversion, and Medications in


Asystole and pulseless electrical activity (PEA) Resuscitation
Cardiopulmonary resuscitation (CPR) and endotracheal Cardioversion
intubation Start at 0.5–1 joules/kg; then may double the dose up
Epinephrine to 2 joules/kg.
Consider and manage treatable causes (6 H’s and 5 T’s Defibrillation
mnemonic). Start at 2 joules/kg; then double the dose to 4 joules/kg
Ventricular fibrillation (VF) and pulseless ventricular for all subsequent attempts.
tachycardia (VT) Adenosine
Each defibrillation is followed immediately by 2 minutes Start at 0.1 mg/kg (maximum = 6 mg); then may repeat
of uninterrupted CPR (2 joules/kg for the first at 0.2 mg/kg × 2 doses (maximum = 12 mg/dose).
defibrillation followed by 4 joules/kg for all Epinephrine
subsequent defibrillations). Standard dose = 0.01 mg/kg (equals 0.1 mL/kg of the
Epinephrine is administered with the second 1 : 10,000 solution) intravenously (IV) or intraosseous
defibrillation at 4 joules/kg (repeat epinephrine every High dose = 0.1 mg/kg (equals 0.1 mL/kg of the 1 : 1,000
3–5 minutes followed by defibrillation). solution) IV, IO, or via the ETT
Antidysrhythmic agents are administered with the third TT (tracheal dose) = 0.1 mg/kg (equals 0.1 mL/kg of the
defibrillation at 4 joules/kg: amiodarone or lidocaine 1 : 1,000 solution)
(if amiodarone not available) or magnesium (for Neonatal doses: Always use the 1 : 10,000 solution as
suspected hypomagnesemia or torsades de 0.01 mg/kg IV, IO, or umbilical venous catheter or
pointes). 0.02–0.03 mg/kg via the TT
Simplified VF/pulseless VT algorithm: Atropine
Rhythm check (confirms VF or pulseless VT) → 0.02 mg/kg with minimum of 0.1 mg/dose to avoid
Defibrillation → paradoxical bradycardia. Maximum single dose is
2 minutes of uninterrupted CPR 0.5 mg in a child or 1 mg in an adolescent with an
(plus medications as indicated above) overall dosage total maximum of 1 mg in a child or
VT (with a pulse) 2 mg in an adolescent.
Unstable: Amiodarone
Immediate cardioversion (start at 0.5–1 joule/kg then 5 mg/kg intravenous bolus for ventricular fibrillation or
2 joules/kg) slowly over 20–60 minutes for stable ventricular
Stable: tachycardia to avoid the hypotensive effects of
Amiodarone or lidocaine or procainamide (note: amiodarone
avoid concurrent use of amiodarone and Can be used to treat a wide array of both atrial and
procainamide) ventricular dysrhythmias in children due to its ability
Supraventricular tachycardia to slow conduction both through the atrioventricular
Unstable: (AV) node and within the ventricles.
If intravenous access is immediately available, Avoid concurrent use of other medications (e.g.,
administer adenosine while preparing for cardio- procainamide) that can also prolong the QT interval.
version; otherwise perform immediate cardiover- Procainamide
sion if intravenous access is not available and/or if 15 mg/kg IV slowly over 30–60 minutes
the patient is hemodynamically very unstable. Can be used to treat a wide array of both atrial and
Stable: ventricular dysrhythmias in children due to its ability
Vagal maneuvers (ice water slurry to the face, to slow conduction both through the AV node and
Valsalva maneuver, blowing on an occluded straw within the ventricles.
or blowing on the tip of a syringe in an attempt to Avoid concurrent use of other medications (e.g.,
blow the plunger out) amiodarone) that can also prolong the QT interval.
Adenosine if vagal maneuvers fail Lidocaine
Bradycardia (hypoxia is the most common etiology) 1 mg/kg IV
Unstable: Does not cause QT prolongation like amiodarone and
Ensure adequate oxygenation and ventilation. procainamide.
Epinephrine Magnesium
Atropine (if suspicion of increased vagal tone or 25–50 mg/kg (maximum of 2 g) intravenous bolus for
cholinergic toxicity) VF or pulseless VT (if suspect hypomagnesemia or
Cardiac pacing torsades de pointes)
Stable: 6 H’s, 5 T’s: hypovolemia, hypoxia, hydrogen ion (acidosis),
No emergent treatment required. hypo-/hyperkalemia, hypoglycemia, and hypothermia;
toxins, tamponade, tension pneumothorax, thrombosis,
and trauma.
From Ralston M, et al: Recognition and management of cardiac arrest. In PALS Provider Manual. Dallas, American Heart Association, 2006, p 153.
2155

Chapter 169 / Cardiac Disorders


Figure 169-9.  An example of an electrocardiogram showing a wide-complex supraventricular tachycardia at a rate of approximately 270 beats/min in
an infant with Ebstein’s anomaly of the tricuspid valve. This infant was in supraventricular tachycardia for approximately 2 days and presented with an
acute exacerbation of her congestive heart failure, as evidenced by the cardiomegaly on the chest radiograph (see Fig. 169-10). Note that the
cardiothoracic ratio in this infant is approximately 70%.

then retrograde back to the atria via the atrioventricular node. Infants with supraventricular tachycardia typically present
Antidromic conduction will produce a wide QRS complex with very nonspecific symptoms, such as fussiness and diffi-
supraventricular tachycardia. Supraventricular tachycardia in a culty feeding. Although healthy infants can generally tolerate
child with a preexisting bundle branch block can also result in supraventricular tachycardia with heart rates approaching 300
a wide-complex supraventricular tachycardia. The ECG in beats per minute, if left untreated supraventricular tachycardia
Figure 169-9 reveals a case of a wide-complex supraventricular may begin to produce signs of CHF and shock. Older children
tachycardia in a child with Ebstein’s anomaly of the tricuspid with supraventricular tachycardia commonly present with pal-
valve who also presented with CHF (Fig. 169-10). pitations, difficulty breathing, and chest discomfort.
Clinical Features and Diagnostic Strategies.  The width of the QRS inter- Management.  Management of supraventricular tachycardia
val in patients with pediatric supraventricular tachycardia is depends on the hemodynamic stability of the child. If the
most commonly narrow-complex, with heart rates in infants child with supraventricular tachycardia is hemodynamically
usually greater than 220 beats per minute (Fig. 169-11). It is unstable and intravenous access in not available, immediate
sometimes difficult to distinguish between sinus tachycardia cardioversion starting at 0.5 to 1 joules/kg is the treatment of
and supraventricular tachycardia (Table 169-8). choice. If the child does not convert with this initial cardiover-
sion attempt, the energy dose can be doubled up to 2 joules/kg
on subsequent attempts. If the child is hemodynamically
stable, vagal maneuvers or adenosine, or both, can be attempted
initially before cardioversion, depending on the individual
case scenario. Regardless of the method selected to convert
the supraventricular tachycardia, a continuous rhythm strip
should always be run to document the response to each con-
version attempt. Vagal maneuvers (e.g., a bag containing a
slurry of crushed ice and water to the face, blowing on an
occluded straw, or blowing on the tip of a syringe) can be
attempted before adenosine administration only in the child
with hemodynamically stable supraventricular tachycardia.
Application of ice to the face has been demonstrated to be a
fairly effective method of converting supraventricular tachy-
cardia in infants and children.40,41 One method to perform this
maneuver is to fill a plastic bag or surgical glove with a slurry
of crushed ice and water, which is then placed over the infant’s
forehead, eyes, and bridge of the nose for 10 to 15 seconds.
Care must be taken not to occlude the nose or mouth with the
Figure 169-10.  Chest radiograph of same infant as in Figure 169-9. bag of ice water. External ocular pressure should be avoided,
as it can be dangerous in children because excessive pressure
PART V  ■  Special Populations / Section One • The Pediatric Patient 2156

Figure 169-11.  Three


electrocardiographic examples of
classic narrow-complex supraventricular
tachycardia in children. The heart rate is
approximately 240 beats/min in the
first two examples (A and B) and
approximately 270 beats/min in the
third example (C).

Table 169-8 Clinical and Electrocardiogram (ECG) Features to Differentiate Sinus Tachycardia from Supraventricular
Tachycardia in Children34

SINUS TACHYCARDIA SUPRAVENTRICULAR TACHYCARDIA

Precipitating event(s) Dehydration, fever, pain No precipitating event


P waves on ECG Present Absent
Heart rate varies with activity Yes No
Beat-to-beat variability Yes Constant R-R intervals
Heart rate in infants (beats/min) Usually <220 Usually >220
Heart rate in children (beats/min) Usually <180 Usually >180

can lead to a ruptured globe. Carotid massage is less effective sine fails to convert the supraventricular tachycardia, the dose
and is not recommended as a vagal maneuver in infants or is then doubled to 0.2 mg/kg with a maximum of 12 mg/dose.
children.34 This 0.2 mg/kg dose of adenosine can be attempted once more
The initial dose of adenosine in children is 0.1 mg/kg with during the third adenosine dose. Elective cardioversion or
a maximum initial dose of 6 mg. If this initial dose of adeno­ esophageal overdrive pacing under conscious sedation may be
2157
DELAY HR218 LEAD II AUTOGAIN

Adenosine 6 mg

Chapter 169 / Cardiac Disorders


LEAD II AUTOGAIN 17 FEB 1906:40 I

Figure 169-12.  An example of adenosine-induced wide-complex tachycardia. A dose of 6 mg of adenosine was administered to this previously healthy
15-year-old girl who presented with a 6-hour history of palpitations. She had no previous cardiac problems except for intermittent palpitations in the
past that always resolved spontaneously without any medical interventions. Once adenosine blocked the conduction through the atrioventricular node,
a wide-complex tachycardia appeared on the electrocardiogram (ECG), which was probably due to antegrade conduction through an accessory
pathway. During the 30 seconds of this wide-complex tachycardia, the patient remained alert with excellent perfusion parameters. This wide-complex
tachycardia then spontaneously converted to normal sinus rhythm. Although the patient’s postconversion ECG did not reveal an accessory pathway,
Holter monitoring 1 month later detected the classic ECG findings of Wolff-Parkinson-White syndrome.

required in children who fail to convert with adenosine. Com- beta-blockers, calcium-channel blockers, and digoxin), because
plications of adenosine administration include asystole and all of these medications only block conduction down the
various dysrhythmias, including adenosine-induced wide- atrioventricular node while leaving the accessory pathway
complex tachycardia (secondary to an occult accessory conduc- wide open to conduct the atrial tachycardia to the ventricles
tion pathway) (Fig. 169-12). The use of verapamil to convert at a potentially lethal rate.42 Under these circumstances,
supraventricular tachycardia in infants and younger children amiodarone, procainamide, or cardioversion would be the
should be avoided because of the high incidence of profound safer alternative. Consultation with the cardiologist and initia-
hypotension and cardiovascular collapse when this medication tion of anticoagulation should also be considered before con-
is administered in this age group.34 version of either of these two atrial dysrhythmias in the
Once the patient has been converted to sinus rhythm, a hemodynamically stable patient to prevent a thromboembolic
12-lead ECG should be obtained to assess for the possibility complication.
of Wolff-Parkinson-White syndrome or any other underlying Ventricular Tachycardia.  Ventricular tachycardia is not a common
conduction abnormalities that may have predisposed the child dysrhythmia in children. The majority of children with ven-
to developing the supraventricular tachycardia. tricular tachycardia have an underlying condition such as status
Atrial Flutter and Atrial Fibrillation.  Both atrial flutter and atrial post–cardiac surgery, myocarditis, prolonged QT syndrome,
fibrillation are rare in children and are usually associated with drug or toxin exposures (e.g., cyclic antidepressants), or elec-
underlying heart conditions (i.e., CHD, status post–open heart trolyte abnormalities. The treatment of ventricular tachycardia
surgical procedures that involved the atria, myocarditis, and will depend on whether a pulse is present and on the hemo-
digoxin toxicity). Hemodynamic stability of these two dys- dynamic status of the patient (Box 169-14). Torsades de
rhythmias depends on the rate of the ventricular response. pointes is a unique type of polymorphic ventricular tachycar-
Cardioversion is the treatment of choice for children who dia that is characterized by QRS complexes that change in
present with hemodynamically unstable atrial flutter or atrial polarity and amplitude. Prolonged QT syndrome, underlying
fibrillation. The initial treatment priority in patients with congenital cardiac defects, hypomagnesemia, and various
hemodynamically stable atrial flutter and atrial fibrillation is medications (e.g., cyclic antidepressants) have all been identi-
first to slow down the rate of the ventricular response with fied as known causes of torsades de pointes. The treatment of
medications such as digoxin, beta-blockers, or diltiazem. Once choice is intravenous magnesium. Class IA (i.e., procainamide)
the ventricular rate is controlled, the rhythm can then be con- and class III (i.e., amiodarone) antidysrhythmic agents are
verted and suppressed with amiodarone, procainamide, or both contraindicated in the treatment of torsades de pointes,
elective cardioversion. If the patient who presents with atrial because these two antidysrhythmic agents are capable of pro-
flutter and atrial fibrillation is known to have an underlying longing the QT interval, which could then precipitate the
Wolff-Parkinson-White syndrome, the four medications that degeneration of the torsades de pointes into a more lethal
should be avoided are the “A-B-C-D” medications (adenosine, rhythm.
2158
AHA recommendation for pediatric defibrillation with biphas­
Clinical Conditions in Which Bacterial ­ic defibrillators remains the same as for monophasic defibrilla-
BOX 169-14 Endocarditis Should Be Suspected in a Child tors (i.e., starting at 2 joules/kg followed by 4 joules/kg).
PART V  ■  Special Populations / Section One • The Pediatric Patient

with an Underlying Anatomic Cardiac Defect Asystole and Pulseless Electrical Activity.  Asystole is the most common
rhythm found in cases of out-of-hospital cardiac arrest in
Fever of unknown etiology children and is associated with a less than 1% chance of
A change in the quality of the preexisting heart murmur or survival.53–55 The use of high-dose epinephrine has been
the presence of a new heart murmur deemphasized in the 2005 PALS guidelines.40 The treatment
Development of a neurologic deficit (secondary to central algorithm and medication dosages for asystole and pulseless
nervous system emboli) electrical activity are listed in Box 169-13. Pulseless electrical
New-onset microscopic hematuria activity can either be a slow or fast rhythm and have a narrow
Splenomegaly or wide QRS complex.
Petechiae The key to survival from any pulseless electrical activity
Splinter hemorrhages involving the conjunctiva, nail beds, rhythm is to rapidly identify and correct the underlying cause.
palms, or soles The etiologies for pulseless electrical activity can be remem-
Myalgias bered by the “6 H’s and 5 T’s”: hypovolemia, hypoxemia,
hypothermia, hydrogen ion (acidosis) hypo-/hyperkalemia,
Pulseless Rhythms hypoglycemia, hypothermia, toxins, tamponade, tension pneu-
mothorax, thrombosis, and trauma.34,40 The most common
Ventricular Fibrillation and Pulseless Ventricular Tachycardia.  Ventricular cause of pulseless electrical activity in children is profound
fibrillation and pulseless ventricular tachycardia account for hypovolemia. Therefore, an intravenous fluid bolus should
approximately 10% of out-of-hospital cardiac arrest cases in always be considered as a therapeutic option during the treat-
which a terminal rhythm was recorded.43,44 The survival rate ment of pulseless electrical activity.
for out-of-hospital ventricular fibrillation and pulseless ven-
tricular tachycardia can be as high as 30%, whereas the survival
rate from asystolic cardiac arrest is less than 1%.31 In a recent Bacterial Endocarditis
study of in-hospital cardiac arrests, a shockable rhythm was Perspective
present during some point of the resuscitation in 25% of the
children.45 The survival rate of children who initially exhibited Bacterial endocarditis involves an infection of the endothelial
shockable rhythms were higher than those children who pre- surfaces of the heart with a propensity for the valves. The
sented with nonshockable rhythms. However the survival incidence of bacterial endocarditis in children may be increas-
rates in those children who later developed a shockable rhythm ing slightly due to the many advances in surgical technology
at some time during their resuscitation were not as good.45–47 that are now allowing many children with very complex con-
Ventricular fibrillation should also be suspected as the arrest genital heart lesions to survive. Children with indwelling intra-
rhythm in cases of commotio cordis or in cases of sudden venous lines with or without underlying CHD are also at risk
cardiac arrest. Current arrhythmia detection algorithms for for developing bacterial endocarditis. Although bacterial endo-
automated external defibrillators (AEDs) appear to have a high carditis most commonly occurs in children with an underlying
sensitivity and specificity for detecting shockable rhythms in CHD or an acquired cardiac lesion (e.g., acute rheumatic val-
children; in July 2003 the AHA gave their approval for the use vular heart disease), it can also occur in patients with no under-
of AEDs in children 1 to 8 years of age (class IIB recommenda- lying anatomic defects of the valves or endocardium. In a
tion). When an AED is to be used on a child younger than 8 series of 62 children with bacterial endocarditis, 19 (30%) of
years (or <25 kg) the use of a pediatric attenuator device is the children had normal cardiac anatomy.56
strongly recommended in order to deliver a more pediatric- Factors that predispose children with underlying anatomic
appropriate dose of defibrillation.48,49 The American Academy cardiac defects to bacterial endocarditis include dental proce-
of Pediatrics (AAP) also recently supported the use of AEDs dures and other surgical procedures involving the respiratory,
in children.50,51 There currently is still not enough clinical evi- gastrointestinal, or genitourinary tracts. Those cardiac lesions
dence to recommend for or against the use of AEDs in infants with a more turbulent blood flow or a higher flow velocity are
under 1 year of age. more prone to developing bacterial endocarditis secondary to
The treatment algorithm and medication dosages for ven- a greater risk of endothelial surface damage, which then
tricular fibrillation and pulseless ventricular tachycardia are increases the risk of platelet deposition and vegetation forma-
listed in Box 169-13. The 2005 PALS arrhythmia algorithms tion. Cardiac lesions that carry this higher risk include ven-
are basically the same as the 2000 treatment guidelines with tricular septal defects, aortic valvular stenosis, tetralogy of
one major change. Based on the 2005 PALS guidelines, ven- Fallot, single ventricle states, prosthetic valves, and postopera-
tricular fibrillation and pulseless ventricular tachycardia are tive systemic-to-pulmonary shunts. Isolated secundum atrial
now treated with single defibrillations followed immediately septal defects carry a much lower risk for bacterial endocarditis
by 2 minutes of uninterrupted cardiopulmonary resuscitation because the shunt flow through the atrial septal defect is typi-
(CPR).40 Although a single shock with a biphasic defibrillator cally of a much lower velocity.
has a high likelihood of terminating ventricular fibrillation, the
resulting rhythm is typically a nonperfusing rhythm, which Clinical Features and Diagnostic Strategies
therefore requires CPR in order to maintain perfusion to the
heart and brain until normal cardiac contractility can resume.40,52 The early clinical manifestations of bacterial endocarditis may
Epinephrine is given with the second defibrillation and antiar- be very nonspecific. The child may simply present with only
rhythmic agents are added to the treatment algorithm with the fever and tachycardia. However, bacterial endocarditis should
third defibrillation. Although biphasic defibrillators can convert be suspected in any child with an anatomic cardiac defect who
ventricular fibrillation at lower dosages than the previous presents with an unexplained fever. This diagnosis must
monophasic defibrillators in adults, until more data are gath- always be considered in any child with a known CHD or an
ered on biphasic defibrillation dosages in children, the current acquired cardiac lesion who presents with any of the condi-
2159
tions listed in Box 169-14. A new heart murmur is present in
fewer than 50% of the bacterial endocarditis cases.57 Procedures for Which Prophylaxis
BOX 169-16 Is Recommended
In addition to vigilance for the diagnosis of infective endo-

Chapter 169 / Cardiac Disorders


carditis—especially in children with CHD—the emergency
physician should be aware of the indications for prophylaxis. All dental procedures that involve manipulation of gingival
In 2007 the AHA in conjunction with the AAP and the Infec- tissue or the periapical region of teeth or perforation of
tious Diseases Society of America published revised guide- the oral mucosa*
lines for the prevention of infective endocarditis.58 The changes Consider prophylaxis for incisional procedures on the
simplify and greatly narrow the recommendations to provide respiratory tract, infected skin, or musculoskeletal tissue
prophylaxis for only the higher risk patients and procedures only for high-risk patients
(Box 169-15). For those children for whom prophylaxis is rec- *The following procedures do not need prophylaxis: routine anesthetic
ommended, the indications are (1) all dental procedures and injections through noninfected tissue, dental radiographs, placement
(2) any manipulation or perforation of the gingival or oral of removable prosthodontic or orthodontic appliances, adjustment of
mucosa.58 It should be noted that antibiotic prophylaxis for orthodontic appliances, placement of orthodontic brackets, shedding
of deciduous teeth, and bleeding from trauma to the lips or oral
infective endocarditis is no longer recommended for gastroin-
mucosa.
testinal and genitourinary procedures (Box 169-16). The com- From American Heart Association: Prevention of infective endocarditis.
mittee found that it is still reasonable to give prophylaxis for Guidelines from the American Heart Association. Circulation 116:1736,
procedures on the respiratory tract, infected skin, or musculo- 2007.
skeletal tissue only for high-risk patients (Table 169-9).58
Diagnostic studies to perform in a child with suspected
bacterial endocarditis include a complete blood cell count, C- reactive protein (CRP) assessment, measurement of erythro-
cyte sedimentation rate, three blood cultures, chest radiography,
and ECG. Cultures from scrapings of cutaneous emboli can
Cardiac Conditions for Which Endocarditis also aid in the diagnosis. Although the definitive diagnostic
BOX 169-15 Prophylaxis Is Reasonable study is the echocardiogram, it is only 80% sensitive in detect-
ing the nidus of infection on the endocardium or valves.59
Prosthetic cardiac valve or prosthetic material used for Streptococcus viridans and Staphylococcus aureus are the two most
cardiac valve repair common offending organisms recovered from the blood cul-
Previous infective endocarditis tures of children with bacterial endocarditis. Recent studies
Congenital heart disease (CHD)*: have shown that in children with CHD, 60% of the cases
Unrepaired cyanotic CHD, including palliative shunts and caused by staphylococcal species are methicillin resistant and
conduits were associated with increased risk of mortality.60
Completely repaired congenital heart defect with
prosthetic material or device during the first 6 months Management
after the procedure†
Repaired CHD with residual defects at the site or Antibiotics should be started immediately after blood culture
adjacent to the site of a prosthetic patch or device samples have been obtained. Although the choice of intrave-
(which inhibit endolethialization) nous antibiotics depends on the suspected source of seeding
Cardiac transplantation recipients who develop cardiac and the child’s immune status, a common recommended
valvulopathy regimen includes an aminoglycoside plus a penicillinase-
resistant penicillin such as oxacillin. If methicillin-resistant
*Except for those conditions above, antibiotic prophylaxis is no longer staphylococci are suspected, vancomycin should also be
recommended for any other form of CHD. included in the initial empirical antibiotic regimen.61

Prophylaxis is reasonable because endothelialization of prosthetic
Bacterial endocarditis in the preantibiotic era was nearly
material occurs within 6 months after the procedure.
From American Heart Association: Prevention of infective endocarditis. always a fatal disease. Although the survival rate in patients
Guidelines from the American Heart Association. Circulation 116:1736, with bacterial endocarditis has improved with the initiation of
2007. antibiotics, the current mortality rate is still 6 to 14%.62 Compli-

Table 169-9 Regimens for Prophylaxis of Infective Endocarditis


SINGLE DOSE 30–60 MINUTES BEFORE
PROCEDURE

SITUATION AGENT ADULTS CHILDREN

Oral Amoxicillin 2 g PO 50 mg/kg PO


Unable to take oral medications Cefazolin or ceftriaxone 1 g IM or IV 50 mg/kg IM or IV
Allergic to penicillins or Cephalexin 2 g PO 50 mg/kg PO
ampicillin—oral or
Clindamycin 600 mg PO 20 mg/kg PO
or
Azithromycin or clarithromycin 500 mg PO 15 mg/kg PO
Allergic to penicillins or Cefazolin or ceftriaxone 1 g IM or IV 50 mg/kg IM or IV
ampicillin—unable to take oral or
medications Clindamycin 600 mg IM or IV 20 mg/kg IM or IV
IM, intramuscularly; IV, intravenously; PO, orally.
Adapted from American Heart Association: Prevention of infective endocarditis. Guidelines from the American Heart Association. Circulation 116:1736, 2007.
2160
cations of bacterial endocarditis include systemic septic emboli, it is the definitive method to confirm the diagnosis and origin
pulmonary emboli, central nervous system emboli with resul- of myocarditis. If the endomyocardial biopsy is performed, it
tant neurologic deficits, dysrhythmias, CHF, myocarditis, myo- will typically demonstrate a myocardial inflammation with
PART V  ■  Special Populations / Section One • The Pediatric Patient

cardial abscesses, and valvular obstruction. Despite appropriate lymphocytic and monocytic infiltrates. The goal of treatment
antibiotic treatment, surgical intervention to remove septic is to maintain adequate cardiac output and to control any
vegetations or valve replacements is sometimes necessary. associated dysrhythmias. Children who present in CHF may
require inotropic support and diuretics. Digoxin and the
various pressors must be used very cautiously in children with
Myocarditis myocarditis because the inflamed myocardium is very sensi-
Perspective tive to the dysrhythmogenesis of these medications. The use
of beta-blockers is contraindicated,63 and the routine use of
Myocarditis is an inflammatory condition of the myocardium immunosuppressive agents remains controversial.64 Although
with various infectious and noninfectious origins. In the United the majority of children with acute viral myocarditis make a
States, the most common cause is viral, with coxsackievirus B full recovery, a few patients will progress to develop dilated
and enteroviruses accounting for the majority of cases. Other cardiomyopathy, which is characterized by dilated ventricles
viral causes include echoviruses, influenza A, influenza B, and impaired systolic contractility.
adenovirus, varicella-zoster virus, Epstein-Barr virus, cytomeg-
alovirus, and hepatitis B virus. Bacterial causes include
Corynebacterium diphtheriae, Streptococcus pyogenes, S. aureus, Pericarditis
Mycoplasma pneumoniae, Borrelia burgdorferi, and Meningococcus. Perspective
Noninfectious causes include Kawasaki disease, acute rheu-
matic fever (ARF), collagen vascular disorders (e.g., systemic Pericarditis is an inflammatory process within the pericardial
lupus erythematosus), toxins (e.g., cocaine and adriamycin), sac, which may not be associated with a pericardial effusion.
endocrine disorders (e.g., hyperthyroidism), and drug-induced The majority of cases of pericarditis in children are self-limited
hypersensitivity (e.g., penicillins, sulfonamides, phenytoin, and follow a benign clinical course. In children, there is nor-
carbamazepine). mally about 10 to 15 mL of fluid within the pericardial sac. A
sudden increase or a large amount of fluid within this pericar-
Clinical Features dial sac can cause a tamponade-induced decrease in stroke
volume, which will cause a diminished cardiac output and
Myocarditis usually has a gradual onset with preceding upper hypotension.
respiratory tract infection symptoms. The presenting signs and Although the most common causes of pericarditis include
symptoms of a child with myocarditis depend on the cause of bacterial and viral infections, other causes include ARF, sys-
the myocarditis, the age of the patient, and the degree of temic lupus erythematosus, uremia, postpericardiotomy syn-
myocardial inflammation. The key to diagnosing myocarditis drome, leukemia, lymphoma, and tuberculosis. Approximately
in infants and children is to suspect the diagnostic entity in 30% of pericarditis cases are due to bacteria such as Pneumococ-
the appropriate clinical setting. In mild cases, the only sign of cus, S. aureus, Meningococcus, and Haemophilus influenzae.2
myocarditis may be tachycardia. Tachycardia that is dispropor- Approximately 30% of the purulent bacterial pericarditis cases
tionate to the degree of fever should alert the clinician to the occur in children younger than 6 years.24 Although viral causes
possibility of myocarditis.63 Other presenting signs and symp- are common, a viral pathogen is recovered in only 20 to 30%
toms include fever, myalgias, fatigue, tachypnea, wheezing, of the cases.24 Common viral causes include coxsackieviruses,
abdominal pain, and chest pain. More severe cases of myocar- ECHO viruses, adenovirus, Epstein-Barr virus, and influenza
ditis can even present with signs and symptoms of acute CHF viruses.
and various dysrhythmias. The physical examination may
reveal a new murmur, a gallop rhythm, or a pericardial friction Clinical Features
rub with muffled heart tones (if the myocarditis is also accom-
panied by pericarditis). The presenting signs and symptoms of pericarditis depend on
the cause of the pericarditis as well as on the amount of fluid
Diagnostic Strategies and Management that has accumulated within the pericardial sac. Chest pain
that varies with position is a common complaint with pericar-
The evaluation and management of the child with myocarditis ditis. The chest pain that is classically associated with pericar-
depend on the suspected cause and the presenting signs and ditis is exacerbated with inspiration and the supine position
symptoms. Blood cultures and viral titers should be considered but relieved when the patient sits up or leans forward. Tachy-
in infectious and postinfectious cases. Appropriate antibiotics cardia is also a common finding in patients with pericarditis.
should be initiated immediately in cases with suspected bacte- Other physical examination findings that have been associated
rial origin. The chest radiograph in very mild cases may be with pericarditis include fatigue, tachypnea, neck vein disten-
normal, but in more advanced cases cardiomegaly will be tion, pulsus paradoxus, hepatomegaly, lower extremity edema,
evident. The ECG findings are usually nonspecific and can and thready distal pulses if heart failure is present. The cardiac
include low-voltage, nonspecific ST segment abnormalities, T auscultatory findings in a patient with pericarditis can include
wave inversions, atrioventricular blocks, and various other dys- a harsh-sounding friction rub or diminished or muffled heart
rhythmias. Creatine phosphokinase-MB, cTnT, CRP, and tones, if there is a significant amount of fluid within the peri-
erythrocyte sedimentation rate may be elevated. cardial sac. The pericardial friction rub, if present, is best heard
The echocardiogram is an essential component of the when the patient sits up or leans forward. This friction rub of
workup of any child with suspected myocarditis. The echocar- pericarditis can be distinguished from a pleural friction rub by
diogram will not only be useful to assess the degree of left having the patient hold his or her breath during auscultation.
ventricular function, but it will also be able to detect any The friction rub of pericarditis will remain present during
associated pericardial effusion that may accompany the myo- breath-holding while the pleural friction rub will no longer be
carditis. Although an endomyocardial biopsy is rarely required, heard while the patient holds the breath.
2161
Progression of EKG changes in pericarditis
7/11/90 7/18/90 7/23/90 12/19/90

Chapter 169 / Cardiac Disorders


I I I I

II II II II

Figure 169-13.  The classic electrocardiographic


(ECG) progression of a patient with pericarditis.
First phase: Diffuse ST segment elevation. aVF aVF aVF aVF
Second phase: ST segments back to isoelectric,
but decreased T wave amplitude. Third phase:
T wave inversion. Fourth phase: Complete
resolution. Notice that the first three phases of
the ECG abnormalities of pericarditis in this
patient are evident during the first 2 weeks of
his illness. A follow-up electrocardiogram V2 V2 V2 V2
obtained 5 months later reveals a complete
resolution of all the previous ECG
abnormalities.

V4 V4 V4 V4

V6 V6 V6 V6

The chest radiograph in a child with pericarditis may not accumulated within the pericardial space, the presence of an
reveal an enlarged cardiac silhouette, depending on the amount anterior and posterior fluid collection is suggestive of a large
of fluid that has accumulated within the pericardial sac. If collection.
there is a large collection of fluid within the pericardial sac, the
heart shadow on the chest radiograph will resemble a “water Management
bottle” silhouette. Approximately 50% of pericarditis cases
also have an associated pleural effusion.2 The management of a child with pericarditis depends on both
The classic ECG findings of pericarditis include diffuse ST the suspected cause and the amount of fluid that has accumu-
segment elevation and diffuse T wave inversions in all leads. lated within the pericardial space. An emergency pericardio-
The classic ECG changes associated with pericarditis evolve centesis will be required in those patients who develop signs
through four phases (Fig. 169-13). During the initial phase, of acute cardiac tamponade. Any fluid that is aspirated from
there is diffuse ST segment elevation in all leads secondary to the pericardial space should be sent for routine cell counts,
subepicardial inflammation; PR segment depression may also Gram’s stain, and cultures. Anti-inflammatory agents and
be seen. During the second phase, the previously elevated ST appropriate antibiotics should also be initiated based on the
segments begin to return to isoelectric baseline and the T suspected cause. Steroids are reserved for refractory cases that
wave amplitudes begin to decrease with flattening of the T are not responsive to the above agents and would be consid-
waves. During the third phase, although the ST segments are ered only after an infectious etiology is ruled out.65
now back to isoelectric baseline, the T waves are now inverted.
The fourth and final phase demonstrates complete resolution
of the ST segment and T wave abnormalities. Diminished Kawasaki Disease
ECG voltages in all leads can also occur if there is a significant Perspective
amount of fluid accumulated within the pericardial sac.
The diagnostic procedure of choice in any patient with sus- Kawasaki disease, originally described as mucocutaneous
pected pericarditis is the echocardiogram, because this study lymph node syndrome by Dr. Tomisaku Kawasaki in 1967, has
will confirm both the presence and the amount of accumulated emerged as a significant cause of acquired cardiac disease in
fluid within the pericardial sac. Although echocardiography children in the United States. An estimated 3000 to 5000 cases
cannot accurately quantify the exact amount of fluid that has of Kawasaki disease are diagnosed annually in the United
2162
inclusive criteria recommend that in a child who is febrile 5
BOX 169-17 Diagnostic Criteria for Kawasaki Disease (KD) days or greater, the presence of two or three criteria should
I. Fever ≥5 days prompt further testing. A CRP of 3 mg/dL or more and/or
PART V  ■  Special Populations / Section One • The Pediatric Patient

II. At least four of the five following physical examination an erythrocyte sedimentation rate (ESR) of 40 mm/hr or
findings: more necessitate further laboratory investigation, such as
1. Bilateral, nonexudative bulbar conjunctival injection those listed in Box 169-18. All of these children will also
(bilateral scleral injection with perilimbic sparing). need echocardiography to assess coronary aneurysm, and the
2. Oropharyngeal mucous membrane changes recommendation is to treat empirically those whose supple-
(pharyngeal erythema, red/cracked lips, and a mentary laboratory values (see Box 169-18) are positive for
strawberry tongue). systemic inflammation while awaiting echocardiographic
3. Cervical lymphadenopathy (with at least one node confirmation.67
>1.5 cm in diameter). Children with a CRP of less than 3 mg/dL and an ESR of
4. Peripheral extremity changes (diffuse erythema and less than 40 mm/hr may be followed daily and reassessed. It is
swelling of the hands and feet during the acute phase important to note that the committee also added a stipulation
or periungual desquamation during the convalescent that the infant 6 months or younger with fever of 7 or more
phase of the illness). This diffuse palmar erythema days should also undergo supplemental laboratory testing, and
seen in KD is in contrast to the discrete macular if any signs of systemic inflammation are found, should have
lesions of various viral illnesses (e.g., measles) that can an echocardiogram. This underscores the current limits of
sometimes be seen on the palms and soles. diagnosis of Kawasaki disease, as well as the obligation to
5. A polymorphous generalized rash (nonvesicular and prevent the disastrous sequelae of aneurysmal development.
nonbullous). There is no specific rash that is
pathognomonic for KD. Differential Considerations
In a child with ≥4 criteria, the diagnosis may be made on day 4 of the Measles can mimic Kawasaki disease but is rare in vaccinated
fever.67 children (i.e., a febrile illness with red eyes, a rash, and ery-
thema of the oropharynx). The classic location, distribution,
and progression of a measles rash starts on the head and face
States. Up to 20% of untreated children develop some degree and progresses caudally. The rash of Kawasaki disease typi-
of coronary artery abnormalities.66 This febrile, exanthema- cally begins on the trunk and then spreads to the face and
tous, multisystem vasculitis is seen most commonly in children extremities. In contrast, Kawasaki disease can exhibit a poly-
younger than 5 years of age with a male-to-female ratio of morphous rash that is nonbullous and nonvesicular.
1.5 : 1. The incidence is higher in Asian children and in certain The palmar lesions of measles are discrete macular lesions
geographic locations such as Hawaii. Although the exact (see Fig. 169-14F), whereas the palmar finding in children
cause of this vasculitis of small and medium-sized vessels with Kawasaki disease is diffuse erythema, which may later
remains unknown, early clinical recognition and initiation of lead to desquamation (see Fig. 169-14C).
high-dose aspirin and intravenous immunoglobulin therapies Streptococcal disease, including pharyngitis and scarlet
have improved the morbidity and mortality rates of Kawasaki fever, can be confused with Kawasaki disease, but conjuncti-
disease in children. vitis and swelling of the hands and feet are unusual for strep-
tococcal disease. Other infectious or autoimmune etiologies
Clinical Features that may mimic Kawasaki disease include Rocky Mountain
spotted fever and leptospirosis, or Stevens-Johnson syndrome
The key to prevention of the coronary artery complications of and juvenile rheumatoid arthritis.67
Kawasaki disease is first to recognize the clinical signs and As seen above, there are many imitators of Kawasaki
symptoms of this disease. In addition to fever, the physical disease. Conversely, this systemic vasculitis can affect any
examination of a child with Kawasaki disease may reveal the organ system, and thus mislead the clinician in diagnosis.
typical findings as listed in Box 169-17 and illustrated in Fig. For example, Kawasaki disease can present with nausea, vom-
169-14. The classic features of Kawasaki disease may manifest iting, and abdominal pain in a febrile child, which may be
simultaneously or in series over days; a careful history and mistaken for a surgical abdomen. In addition, a febrile, irritable
physical examination may elucidate the need for further child with Kawasaki disease may exhibit a cerebrospinal fluid
testing. In addition, very young children may not have a classic pleocytosis and be misdiagnosed with viral meningitis. For this
presentation and require further investigation. All children reason, Kawasaki disease should be included in the differential
with suspected Kawasaki disease, with either classic or incom- diagnosis of any child with several days of fever, rash, and
plete features, should undergo echocardiography to detect the nonpurulent conjunctivitis to avoid the pitfall of early diagnos-
presence and degree of coronary aneurysm.67 tic closure.

Incomplete Kawasaki Disease Clinical Course


The classic presentation of Kawasaki disease is a clinical diag- Kawasaki disease is postulated to be caused by an infectious
nosis of four or more of the five criteria in a child who is febrile agent that enters the respiratory tract and initiates an oligoclo-
five or more days. However, these strict criteria may miss a nal immunoglobulin A response, which activates lymphocytes,
substantial number of children who present with incomplete cytokines, and proteinases that weaken vessel walls and pre-
Kawasaki disease. Any child may have an incomplete presen- dispose the entire circulation to aneurysms.68 The main reason
tation, but this is mostly seen in infants less than 6 months to recognize this disease entity in children is to initiate prompt
old.67 therapies to prevent the cardiac complications of Kawasaki
The AHA’s Committee on Rheumatic Fever, Endocarditis, disease, which occur in two phases. Approximately 25% of
and Kawasaki Disease has published consensus guidelines on patients develop mild diffuse myocardial inflammation. This
the approach to incomplete Kawasaki disease.67 The more occurs during the acute febrile period and is characterized by
2163

Chapter 169 / Cardiac Disorders


A B
Figure 169-14.  Classic physical
examination findings of Kawasaki
disease. Note the bilateral nonexudative
scleral injections (A) with perilimbic
sparing (the thin margin of white sclera
around the cornea), red-cracked lips
with a strawberry tongue (B), diffuse
palmar erythema (C), red soles (D), and
the polymorphous exanthem (E). The
diffuse palmar erythema of Kawasaki
disease (C) is distinct from the palmar
findings seen in other viral illnesses such
as the discrete macular lesions on the
palms in this child with measles (F).

C D

Supplemental Laboratory Criteria for tachycardia, a gallop rhythm, or nonspecific ST-T wave
BOX 169-18 Kawasaki Disease changes. Up to 5% of the children also exhibit some degree of
CHF during this acute phase of their illness. This carditis
Albumin ≤3 g/dL usually resolves when the fever resolves. Pericardial effusions
Anemia for age also occur in up to 20 to 40% of cases. Mild mitral and aortic
Platelet count of ≥450,000/mm3 regurgitation is also seen in 1 to 2% of untreated cases on
White blood cell (WBC) count ≥15,000 mm3 echocardiographic examinations. This phase of the disease is
Elevation of alanine aminotransferase mild and self-resolving. Therapy during this phase of the
Sterile pyuria of ≥10 WBCs per high-power field illness is primarily supportive.
The second phase of the disease involves coronary artery
From Newburger JW, et al: Diagnosis, treatment, and long-term
management of Kawasaki disease: A statement for health professionals
dilation, which usually peaks 2 to 4 weeks from the onset of
from the Committee on Rheumatic Fever, Endocarditis, and Kawasaki the illness and is seen in 15 to 25% of the untreated patients
Disease, Council on Cardiovascular Disease in the Young, American with Kawasaki disease.69 Without the appropriate treatment,
Heart Association. Circulation 110:2747, 2004. 15 to 20% of children with Kawasaki disease go on to develop
2164
coronary aneurysms within 1 to 3 weeks from the onset of their include streptokinase, tissue-type plasminogen activator, and
illness.2 These coronary aneurysms can then lead to myocar- interventional cardiac catheterization.67
dial infarction, thrombosis, rupture, and a variety of ischemia- The follow-up of children with Kawasaki disease depends
PART V  ■  Special Populations / Section One • The Pediatric Patient

induced dysrhythmias. Significant risk factors for coronary on the degree and presence of carditis and coronary artery
aneurysmal formation include male gender, age younger than abnormalities detected on the initial echocardiogram. Other
1 year or greater than 8 years, a prolonged febrile period greater imaging modalities used to follow aneurysmal parameters
than 10 to 14 days, early myocarditis, anemia (Hgb < 10 g/dL), include electron-beam computed tomography, coronary mag-
white blood cell count greater than 30,000, an increased band netic resonance angiography, and multislice spiral computed
count, elevated ESR, elevated CRP level, low serum albumin tomography.67 Those children with more severe cardiac abnor-
levels, and aneurysms involving the renal, axillary, or iliac malities require very close follow-up by a cardiologist who is
arteries and giant coronary aneurysms (>8 mm in diameter). experienced in managing the cardiac complications of
Death from Kawasaki disease is primarily due to myocardial Kawasaki disease. Overall, prompt diagnosis and appropriate
infarction secondary to coronary artery occlusion. Giant coro- therapies can prevent coronary aneurysm formation in up to
nary artery aneurysmal rupture is rare. Although most of the 95% of the cases as well as rapid symptomatic improvement
fatalities that occur with Kawasaki disease occur within 6 in up to 90% of the cases.72
weeks from the onset of the illness, sudden death can also
occur many years after the illness. Prompt recognition and
treatment have decreased this mortality rate from 2% to less Acute Rheumatic Fever
than 0.01%.66 Perspective

Management Acute rheumatic fever is the result of a delayed immune reac-


tion of a group A streptococcal infection. ARF is one of the
The main goal of treatment during the acute febrile phase of most common causes of acquired heart disease in children. In
Kawasaki disease is to provide supportive care and to decrease the United States, ARF most commonly occurs in children 5
the inflammation of the myocardium and coronary arteries. to 15 years of age and has an attack rate of 0.3% in children
The two major components of treatment include intravenous with an untreated streptococcal infection. Although this disease
immunoglobulin (IVIG) infusion and high-dose aspirin affects multiple organ systems, carditis is the most serious
therapy, which together have an additive effect. This combi- complication.
nation of IVIG and high-dose aspirin, when initiated within 10
days from the onset of the illness, can substantially decrease Clinical Features and Diagnostic Considerations
the progression to coronary artery dilation and aneurysm for-
mation as compared with aspirin therapy alone and results in The diagnosis of ARF is based on the Jones criteria (Box 169-
a more rapid resolution of fever and the other indicators of 19). In addition to the Jones criteria, there must also be evi-
acute inflammation.66 However, despite prompt treatment dence of an antecedent streptococcal infection, which can be
with IVIG and high-dose aspirin, 2 to 4% of the children still documented with either a positive throat culture, a positive
develop coronary artery abnormalities.66 rapid streptococcal antigen test finding, or an elevated anti-
The current IVIG regimen involves an infusion of 2 g/kg streptolysin O titer. The streptozyme test is not as reliable and
over 10 to 12 hours. Side effects of IVIG include hypotension, therefore should not be used as a definitive test for evidence
nausea, vomiting, and seizures. Close cardiac monitoring of an antecedent group A streptococcal infection.73 The diag-
during the IVIG infusion is mandatory. The 5 to 10% of
children who receive IVIG who experience a persistent or
recurrent fever after the initial dose of IVIG may be given a Jones Criteria for the Diagnosis of Acute
second infusion at the same dose. Approximately two thirds of BOX 169-19 Rheumatic Fever (ARF)
those children who fail to respond to the initial dose of IVIG
will improve with the second infusion. The diagnosis of ARF is based on the documentation of an
Aspirin is initiated at 80 to 100 mg/kg/day orally divided into antecedent streptococcal infection and either two major
an every-6-hour dosing regimen until the child is afebrile for criteria or one major criteria + two minor criteria.
48 to 72 hours (or longer). This dose is then decreased to 3 Major criteria
to 5 mg/kg orally each day until the laboratory study results Carditis
return to normal, which typically occurs 6 to 8 weeks after the Migratory polyarthritis
onset of the disease. Aspirin therapy is continued beyond this Erythema marginatum
period only in those children in whom coronary artery abnor- Subcutaneous nodules
malities are present. Ibuprofen can antagonize the antiplatelet Chorea
effects of aspirin and should be avoided during treatment.67 Minor criteria
The use of corticosteroids as primary treatment has not been Clinical findings
well established. A recent multicenter, randomized, double- Fever
blind study to determine the efficacy of the addition of meth- Arthralgia
ylprednisolone to initial conventional therapy showed no Laboratory findings
difference in clinical outcome, including dimensions of coro- Elevated C-reactive protein or erythrocyte
nary artery aneurysms, length of hospital stay, and rate of re- sedimentation rate
treatment with IVIG.70 The current recommendations include Prolonged PR interval
corticosteroids for refractory cases of Kawasaki disease, in Supporting evidence of an antecedent group A
which children are continued on high-dose aspirin, a second streptococcal infection with either:
dose of IVIG is administered, and corticosteroids are added to A positive throat culture or a rapid streptococcal
decrease the risk of coronary artery aneurysm.67,71 Other com- antigen test
plications such as coronary aneurysmal thrombosis likewise An elevated antistreptolysin O titer
have limited data to guide management. Therapeutic options
2165
nosis of ARF is made in a patient with a documented antece­
dent streptococcal infection who exhibits either two major Cardiovascular Etiologies of Sudden Death in
BOX 169-20 Young Athletes
criteria or one major plus two minor criteria. The most common

Chapter 169 / Cardiac Disorders


presenting major criterion is the migratory polyarthritis, which
commonly involves the larger joints of the extremities as well Hypertrophic cardiomyopathy
as the smaller tarsal joints in the foot and the smaller carpal Various congenital coronary artery anomalies
joints in the hand. The carditis of ARF most commonly Prolonged QT interval syndrome
involves valvulitis of the mitral and aortic valves, which clini- Various preexcitation syndromes (e.g., Wolff-Parkinson-
cally manifests as occult mitral or aortic insufficiency. The White syndrome)
murmur of mitral insufficiency is characterized as a holosys- Commotio cordis
tolic murmur best heard over the apex with radiation to the Aortic rupture secondary to Marfan’s syndrome
axilla. The murmur of aortic insufficiency is characterized as a Idiopathic dilated cardiomyopathy
diastolic murmur that is best heard over the base of the heart. Myocarditis
Innocent murmurs that are normally exacerbated with fever Coronary artery disease secondary to Kawasaki disease
can be mistaken for the murmurs of mitral or aortic insuffi- Aortic stenosis
ciency. The other cardiac manifestations of ARF include CHF, Mitral valve prolapse
pericarditis, and various degrees of heart blocks. The two der-
matologic major criteria (erythema marginatum and subcuta-
neous nodules) and chorea occur less commonly than the The most common cardiovascular cause of sudden death in
migratory polyarthritis and carditis. Chorea may occur as the the athlete is hypertrophic cardiomyopathy, which accounts
only manifestation of ARF. If arthritis is used as a major com- for up to 36% of the cardiovascular-related cases.61
ponent, arthralgia cannot be used as a minor component to
make the diagnosis. Likewise, if carditis is used as a major Specific Disorders
component, then a prolonged PR interval cannot be used as a
minor component.73 Congenital Coronary Artery Anomalies.  An additional 24% of the cases
In addition to the ECG, CRP level, ESR, and documenta- of sudden death are due to various anomalies of the coronary
tion of an antecedent streptococcal infection, the diagnostic arteries.75 Congenital coronary artery anomalies are difficult to
workup of ARF should also include a chest radiograph as well detect from a clinical standpoint, but 37% of those individuals
as an echocardiogram to evaluate the degree of cardiac who died from congenital coronary artery anomalies did exhibit
involvement. previous symptoms of exercise-induced syncope or chest
pain.76 The exact pathophysiologic mechanism of sudden
Differential Considerations death in individuals with congenital coronary anomalies is
unknown. Although there are a variety of congenital coronary
The differential diagnosis of ARF includes myocarditis, bacte- artery anomalies, the most common potentially lethal lesion is
rial endocarditis, Lyme disease, systemic lupus erythemato- the anomalous left coronary artery in which the left main and
sus, juvenile rheumatoid arthritis, serum sickness, and septic right coronary arteries both arise from the right sinus of Val-
arthritis. salva. Individuals with this particular anomaly have a 46%
incidence of sudden death, with more than 85% of the known
Management cases of sudden death occurring during exercise.77 Congenital
coronary artery hypoplasia is another uncommon cause of
The acute management of ARF should first focus on stabiliz- exercise-induced sudden death. Any athlete with exertional
ing and treating any of the symptomatic cardiac manifestations syncope or chest pain should be evaluated by a cardiologist for
of the illness, such as CHF or tamponade due to a pericardial the possibility of congenital coronary artery anomalies. If an
effusion. Additional goals in ARF management include appro- anomaly is detected and surgically corrected, the athlete may
priate antibiotic therapy to eradicate the streptococcal infec- resume full activity and participation in competitive sports.
tion, bedrest, and anti-inflammatory agents for the arthritis. Marfan’s Syndrome.  Individuals with Marfan’s syndrome should
The use of steroids in the treatment of carditis should only be be evaluated for potential cardiac abnormalities before being
employed under direction of a cardiologist. Another important allowed to participate in competitive sports. Clinical manifes-
aspect in the management of ARF is the prevention of recur- tations of the disease include tall, slender habitus, striae
rent attacks through the prophylactic use of penicillin, which atrophicae of the skin (multiple stretch marks on skin), dispro-
can be given as monthly injections of 1.2 million units of ben- portionately long extremities as compared to the trunk, scolio-
zathine penicillin G. Alternative prophylactic regimens include sis, pectus excavatum or carinatum, and dislocated lenses of
oral penicillin administered twice daily, or for penicillin- the eye. Approximately 50% of patients with Marfan’s syn-
allergic patients, twice daily oral erythromycin. This prophy- drome have cardiac manifestations such as mitral valve pro-
laxis with penicillin or erythromycin is required until 18 years lapse or aortic dilation. The most serious cardiac complication
of age at the minimum but can be continued for life, depend- of Marfan’s syndrome is the progressive dilation of the aorta
ing on the degree of cardiac involvement and the risk for with the potential risk of aortic rupture, which most commonly
recurrence. involves the descending portion of the aorta. Therefore,
patients with Marfan’s syndrome should be prohibited from
participation in contact sports. Those individuals who are
Cardiac Causes of Sudden Death in   known to have aortic dilation should also be prohibited from
Young Athletes participation in any competitive sports regardless of the degree
Perspective of contact involved.75 All patients with Marfan’s syndrome
with or without cardiac involvement on their initial evaluation
The most common cause of sudden, unexplained death in should be followed by a cardiologist with serial imaging studies
athletes is various cardiac conditions, with only 15% of the of their aorta using echocardiography, magnetic resonance
cases stemming from noncardiovascular causes (Box 169-20).74 imaging, or computed tomography.
2166
Hypertrophic Cardiomyopathy use of digoxin is contraindicated in patients with hypertrophic
Perspective.  Although the nonobstructive form of hypertrophic cardiomyopathy because its positive inotropic effect may
cardiomyopathy is an uncommon cardiac malformation that worsen the left ventricular outflow obstruction. Sudden death
PART V  ■  Special Populations / Section One • The Pediatric Patient

occurs in only 0.2% of the general population, it is the single in these patients with hypertrophic cardiomyopathy is thought
most common cardiac cause of sudden death in the young to be due to exertion-induced ventricular fibrillation or pulse-
athlete.77–79 Hypertrophic cardiomyopathy is a familial disease less ventricular tachycardia. Therefore, the current recommen-
that is inherited in an autosomal dominant fashion with vari- dation is that all individuals who are diagnosed with
able penetrance. The hypertrophy of the left ventricle in this hypertrophic cardiomyopathy, as well as those individuals with
condition is idiopathic in nature and not due to chronic pres- an equivocal diagnosis of hypertrophic cardiomyopathy, should
sure overload conditions such as systemic hypertension or not participate in vigorous activities and competitive sports.
aortic stenosis. The systolic left ventricular contractile func- Prolonged QT Syndrome
tion is vigorous but the thickened muscle of the left ventricle Perspective.  In 1957, Jervell and Lange-Nielsen first described
is stiff, resulting in impaired ventricular relaxation and high the association of recurrent syncope, sudden death, and long
diastolic filling pressures.80 QT interval in a series of deaf patients. Later, in 1963, Romano
Sudden death in previously asymptomatic individuals with reported a similar association of symptoms with long QT inter-
hypertrophic cardiomyopathy occurs during moderate or severe vals in patients with normal hearing. Both the Jervell-Lange-
physical exertion. The proposed pathophysiologic mechanisms Nielsen and the Romano-Ward syndrome are inherited
of sudden death during exertion in these individuals is thought disorders with variable penetrance, characterized by a pro-
to be due to a transient decrease of blood flow out through the longed QT interval that has been associated with sudden
aorta or to dysrhythmias secondary to the hypertrophied ven- death. The corrected QT interval (QTc) in normal individuals
tricular myocardium. should not exceed 0.44 seconds in children or 0.42 seconds in
Clinical Features.  Some individuals with hypertrophic cardiomy- adolescents. Individuals with QTc intervals greater than 0.55
opathy have experienced previous “warning” episodes of chest seconds have a higher risk of sudden death. Prolongation of
pain, dyspnea, syncope, or palpitations during vigorous activi- the QT interval predisposes the individual to ventricular
ties. A family history of sudden unexplained death in young tachycardia, torsades de pointes, and ventricular fibrillation,
adults should also alert the clinician to the possibility of hyper- which is often initiated by a premature ventricular contraction
trophic cardiomyopathy. The majority of young athletes who occurring during the prolonged repolarization phase. In addi-
die from this condition have the nonobstructive form of hyper- tion to the inherited syndromes of prolonged QT intervals,
trophic cardiomyopathy, and the classic loud systolic ejection other causes of prolonged QT intervals include hypocalcemia,
murmur that is present with the obstructive form is not heard hypokalemia, hypomagnesemia, myocarditis, and medications
during the routine presports physical examinations.81 There- (e.g., procainamide, erythromycin, cyclic antidepressants, phe-
fore, the standard presports screening physical examination nothiazines, quinidine, organophosphates).
may fail to detect the presence of nonobstructive hypertrophic Clinical Features.  Symptoms in the young athlete that are sug-
cardiomyopathy in young athletes. If a systolic murmur along gestive of QT prolongation include exercise-induced palpita-
the lower left sternal border is heard on the routine screening tions, chest pains, syncope, dizziness, or atypical seizures. The
physical examination of a young athlete, a Valsalva maneuver young athlete who develops any of these symptoms should be
may help to differentiate the murmur of aortic stenosis from evaluated by a cardiologist, especially if the family history is
the systolic murmur associated with the obstructive form of positive for sudden unexplained death, cardiac problems,
hypertrophic cardiomyopathy. During the Valsalva maneuver, syncope, or deafness. Any young athlete who has been diag-
the venous blood return to the heart is decreased, which in nosed with a prolonged QT syndrome should be prohibited
turn transiently reduces the left ventricular size. The transient from participation in competitive sports and vigorous activi-
reduction in the size of the left ventricle will increase the ties. The growing popularity and presence of AEDs in public
degree of obstruction and thus cause an increase in the inten- places and at sporting events can potentially save the lives of
sity of the systolic murmur heard with the obstructive form of those athletes who suddenly collapse due to an underlying
hypertrophic cardiomyopathy. In contrast to this, the systolic prolonged QT syndrome–induced nonperfusing ventricular
murmur of aortic stenosis will decrease in intensity during a dysrhythmia.
Valsalva maneuver due to the transient reduction of blood flow Management.  Treatment of a prolonged QT interval depends
through the stenotic aortic valve. on the cause. Correction of any underlying metabolic disorder
Individuals who are suspected of having hypertrophic car- and discontinuing a medication that induced the prolongation
diomyopathy based on the above-mentioned exertional symp- of the QT interval are the easiest conditions to correct.
toms, a positive family history, or both should be referred to a Magnesium sulfate is the drug of choice in the treatment of
cardiologist for a more extensive workup. torsades de pointes. Antidysrhythmic agents that also can
Diagnostic Studies.  The ECG findings in individuals with hyper- prolong the QT interval, such as procainamide and amioda-
trophic cardiomyopathy typically reveal various degrees of left rone, should be avoided. Therefore, the safest medication to
ventricular hypertrophy and left atrial enlargement. Other use in a patient with prolonged QT interval-induced ventricu-
ECG findings include prominent Q waves in the inferolateral lar tachycardia or fibrillation is lidocaine. Beta-blockers have
leads or diffuse T wave inversions. The most accurate study been used to prevent sudden ventricular dysrhythmias in
to make the diagnosis of hypertrophic cardiomyopathy is the those patients with the familial forms of QT prolongation.
echocardiogram, which will demonstrate various degrees of Adjunctive treatment in these selected patients also includes
left ventricular hypertrophy most commonly involving the the insertion of pacemakers or internal defibrillators.
ventricular septum in up to 90% of the cases.80 Those patients Commotio Cordis.  The phenomenon of commotio cordis occurs
who have echocardiographic evidence of hypertrophic cardio- when an object such as a baseball strikes the chest and pro-
myopathy should be followed with serial echocardiographic duces sudden death. This phenomenon most commonly occurs
examinations to monitor the progression of their condition. in children between 5 and 15 years of age with no known pre-
Management.  Although beta-blockers have been used in disposing cardiac conditions.75,82 Although commotio cordis
patients with hypertrophic cardiomyopathy, they have not most commonly occurs in baseball, it has also been reported
been shown to prevent sudden death in these patients. The to occur in ice hockey, lacrosse, softball, and fist fights.69 In a
2167
few cases in which the cardiac rhythm was documented after immediately available and the patient is completely unrespon-
the blunt trauma to the chest, the most common rhythm docu- sive with no pulse after sustaining a direct blow to the chest,
mented was ventricular fibrillation.83 The majority of patients some practitioners have proposed that performing chest

Chapter 169 / Cardiac Disorders


who sustain commotio cordis do not survive, especially if they thumps during cardiopulmonary resuscitation may be of some
are not rapidly treated with defibrillation. If an AED is not benefit under this particular circumstance.

KEY CONCEPTS
■ The possibility of a congenital heart defect should be ■ Oxygen, diuretics (furosemide), and inotropic agents
considered in an infant who presents with central (digoxin) are the mainstay of treatment for infants and
cyanosis that does not respond to 100% supplemental children who present with CHF.
oxygen (hyperoxia challenge). ■ If vagal maneuvers fail to convert stable paroxysmal
■ Neonates with ductal-dependent cardiac lesions typically supraventricular tachycardia in children, rapid
present within the first 2 to 3 weeks of life with either adenosine administration (0.1 mg/kg for the first dose
acute cyanosis or shock. Initiation of a PGE1 infusion followed by 0.2 mg/kg on repeat doses) is the treatment
(0.05–0.1 µg/kg/min) will be lifesaving in these neonates. of choice. Verapamil should be avoided in children
■ Treatment of a hypoxic tet spell first includes placing the younger than 1 year due to its profound hypotensive
infant in the knee-to-chest position and providing effects.
supplemental oxygen. Sedative agents can be used to ■ One should consider the use of lidocaine instead of
decrease the infant’s hyperpnea. Various medications can amiodarone in cases of ventricular fibrillation or
be used as adjunctive treatment to increase the SVR and ventricular tachycardia due to medications (e.g.,
thereby decrease the degree of right-to-left shunting cyclic antidepressants) or toxins that prolong the QT
across the ventricular septal defect. interval.
■ Prompt recognition of the clinical findings and ■ Young athletes with a positive family history of sudden
symptoms of Kawasaki disease along with the rapid unexplained death or exertion-induced symptoms such
initiation of high-dose aspirin and IVIG infusion can as chest pain, dyspnea, palpitations, or syncope should
prevent the formation of coronary aneurysms. be evaluated by a cardiologist prior to their resumption
■ Acute bacterial endocarditis should always be considered of vigorous activity.
in a child with a known congenital heart defect or an ■ The increased presence of AEDs in public places and at
acquired cardiac defect who presents with a fever of sporting events can potentially save the lives of more
unknown origin, the development of acute neurologic young athletes who suddenly collapse secondary to
deficits, new-onset microscopic hematuria, myalgias, hypertrophic cardiomyopathy, prolonged QT
splenomegaly, petechiae, or other signs of systemic syndromes, and commotio cordis.
embolization.

The references for this chapter can be found online by accessing the
accompanying Expert Consult website.

You might also like