Passive Addiction and Teratogenic Effects




Abstract


Drugs can exert major effects on the developing central nervous system (CNS). In the broadest sense, drugs may disturb specific developmental events in the brain and, in turn, produce teratogenic effects. In addition, maternal ingestion of certain drugs can result in passive addiction of the fetus and postnatally lead to a neonatal withdrawal or abstinence syndrome. Although some effects may be transient, most are likely to persist in some form into childhood and adolescence with multiple systems affected spanning cognition, motor function, language, and behavior. This chapter provides a comprehensive overview of the neurological effects of a range of licit and illicit drugs on the infant and growing child. Drugs reviewed include fetal exposure to alcohol, antiepileptic drugs, stimulants, opioids, selective serotonin reuptake inhibitors (SSRIs), sedatives, anesthetics, and marijuana. Clinical considerations in the evaluation and care of the drug-exposed neonate are also discussed.


Drugs can exert major effects on the developing central nervous system (CNS). In the broadest sense, drugs may disturb specific developmental events in the brain and, in turn, produce teratogenic effects. In addition, maternal ingestion of certain drugs can result in passive addiction of the fetus, and postnatally lead to a neonatal withdrawal or abstinence syndrome. The capacity for teratogenicity was first recognized in the late 1950s and early 1960s with the recognition of the adverse effects of thalidomide. Believed to be safe, thalidomide was prescribed to large numbers of pregnant women as a treatment for morning sickness but was later found to result in a number of birth defects. The most notable of these was phocomelia, in which the bones of the arms and, in some cases, other limbs were extremely shortened or absent. With this medical tragedy came an increased awareness of the potential teratogenic risks of fetal exposure to prescribed and recreational drugs.


Evidence suggests that prenatal exposure to both licit and illicit drugs can have short-term and long-lasting effects on the structure and function of the developing CNS. These effects vary in severity, from profound effects on morphological structure to more subtle, but nonetheless clinically significant, neurological effects. Some of the latter may include a striking neonatal abstinence syndrome (NAS) or include a range of neonatal neurobehavioral difficulties ( Table 38.1 ). Further, few of these effects are transient, with most persisting in some form into childhood and adolescence. Multiple systems are often affected spanning cognition, motor function, language, and behavior. Fig. 38.1 provides a conceptual overview of the factors and processes involved in the effects of drug exposure in utero on child neurological development.



TABLE 38.1

Nature of Outcomes Related to Intrauterine Drug Exposure




















































































OUTCOME ALCOHOL AEDs STIMULANTS OPIOIDS SSRIs MARIJUANA
Congenital malformations X X
Neonatal abstinence X X X X X
Newborn neurobehavioral difficulties X X X X X
Global cognitive deficits/delay X X
Executive function X
Language problems X X X
Attention problems X X X
Externalizing problems X
Internalizing problems X

AEDs , Antiepileptic drugs; SSRIs , selective serotonin reuptake inhibitors.



Figure 38.1


Schematic model of factors involved in the effects of drug exposure in utero on child neurological developmental outcome.

CNS , Central nervous system.




Major Factors Involved in Neurologic Disturbances Associated With Intrauterine Drug Exposure


Almost all drugs unbound to protein move freely across the placenta, enter the fetal bloodstream, and can affect fetal brain development, either directly or indirectly ( Fig. 38.2 ). The direct effects of a drug on the developing brain will vary depending on the type of drug, the gestational timing of exposure, dose, the extent of drug distribution, and the number of drugs. The developmental stage of the fetus at the time of exposure as well as the sensitivity of different brain regions to different chemical agents also likely play a role. For prescribed drugs, information about timing and extent of exposure is typically known, whereas for alcohol and other illicit drugs, such information is harder to obtain accurately, thereby posing additional challenges for clinical assessment and diagnosis.




Figure 38.2


Pathogenesis of neurologic disturbances from intrauterine drug exposure.

CBZ , Carbamazepine.


In addition to these direct effects, drugs can have indirect effects on fetal brain development via their impact on other organ and physiological systems. Numerous drugs impact fetal blood flow and nutritional exposure. For example, cocaine impairs fetal oxygen and nutrient transfer via profound vasoconstriction of the umbilical vein. Acutely, these alterations may contribute to cerebral infarction and intracranial hemorrhage. Chronically, these perturbations may contribute to the documented impact of cocaine on cortical neuronal migration and differentiation.


It is also increasingly recognized that maternal and fetal genotypes may interact with a drug exposure to determine phenotypic effects ( Fig. 38.1 ). Functional polymorphisms in alcohol metabolism may best exemplify this phenomenon. The offspring of individuals with normal metabolism experience long-term impacts from in utero alcohol exposure, whereas the offspring of rapid metabolizers avoid these effects. Fetal genetic susceptibility has also been observed, with concordance in the manifestations of fetal alcohol spectrum disorder (FASD) between monozygotic twins but not dizygotic twins. Finally, there is also growing evidence to suggest epigenetic influences on outcome. These epigenetic modifications may even be transgenerational, placing subsequent generations at increased risk of drug dependence, even in the absence of direct exposure during gestation.


In addition to these drug- and infant-related factors, several other maternal and environmental factors may play a role in determining the clinical presentation of an infant and are therefore important to consider. Comorbid physical and mental health conditions in the mother, combined with the underlying disease state, may complicate the interpretation of prenatal drug effects and exacerbate risks for the infant. In addition, maternal life style factors, such as pregnancy, nutrition, and social disadvantage, which are correlated with substance use, may also contribute to later risks, including poor growth and neurodevelopmental impairment.




Developmental Consequences for the Infant


As a result of the various factors noted previously, considerable heterogeneity in the nature and severity of outcomes may be observed, including death, malformations, neurodevelopmental disability, and impaired neurobehavioral functioning (see Table 38.1 ). A distinction can also be made between drugs that are associated with congenital malformations in the newborn infant versus those that have more subtle but nonetheless clinically significant neurobehavioral effects, with or without withdrawal symptoms that may require medical management ( Table 38.2 ). In the following, we discuss fetal exposure to alcohol, antiepileptic drugs (AEDs), stimulants, opioids, selective serotonin reuptake inhibitors (SSRIs), sedatives, anesthetics, and marijuana.



TABLE 38.2

Major Drugs Associated With Teratogenic Effects, Congenital Malformations, and Neonatal Neurobehavioral Effects















Teratogenic effects
Alcohol
Antiepileptic drugs such as valproate, hydantoins, barbiturates, and carbamazepine
Stimulants (cocaine, amphetamines)
Neonatal neurobehavioral effects (including neonatal abstinence syndrome)



  • Stimulants (cocaine, amphetamines)



  • Opioids



  • Selective serotonin reuptake inhibitors



  • Benzodiazepines





Alcohol


FASD includes a range of possible diagnoses that may result from a woman drinking alcohol during pregnancy. These diagnoses, which vary in the severity of later abnormalities or impairment, include fetal alcohol syndrome (FAS), partial FAS, alcohol-related birth defects (ARBDs), alcohol-related neurodevelopmental disorder (ARND), and neurobehavioral disorder associated with prenatal alcohol exposure (ND-PAE). At the severe end of the spectrum is the clinical diagnosis of FAS, which was first described in 1968 by Lemoine and colleagues and then reported in further detail by Jones and colleagues. This disorder refers to a specific constellation of neural and extraneural anomalies that include abnormal facial features ( Fig. 38.3 ), poor body growth, and CNS abnormalities that are in turn associated with later cognitive, learning, and behavioral impairments. However, even in the absence of the full features of FAS, it is now also recognized that prenatal alcohol exposure can result in a range of less pronounced dysmorphic, cognitive, and behavioral effects of varying severity, termed fetal alcohol effects (with terminology noted previously).




Figure 38.3


Fetal alcohol syndrome: clinical appearance.

Two children: one (A) in the newborn period and (B) at the age of 6 months, and (C) the other at the age of 16 months. Note the short palpebral fissures, especially prominent in the newborn (A), and the low nasal bridge with short, upturned nose, epicanthal folds, long, hypoplastic philtrum, and convex upper lip with narrow vermilion border.

(A and B, From Jones KL, Smith DW. Recognition of the fetal alcohol syndrome in early infancy. Lancet . 1973;2:999–1001; C, from Hanson JW, Jones KL, Smith DW. Fetal alcohol syndrome: experience with 41 patients. JAMA . 1976;235:1458–1460.)


Prevalence


The prevalence of maternal alcohol use during pregnancy and FASD varies with the drinking patterns of a population. The accurate estimation of these rates is hindered by a number of methodological problems. These include (1) reliance on maternal recall and underreporting due to stigma and a fear of potential punitive consequences; (2) limitations of toxicological measures, such as urine and meconium; and (3) differences in the diagnostic criteria used to define FAS and FASD. Nonetheless, findings generally suggest that in the United States, approximately one-half of all women of childbearing age report having consumed alcohol in the past month, with 15% likely to have had a binge-drinking episode, defined as more than five standard alcoholic drinks on at least one occasion. Although most women reduce their alcohol intake during pregnancy, 8% to 11% continue to drink and 1% to 2% binge drink. Even higher rates of drinking during pregnancy are reported in the United Kingdom, Ireland, Australia, and New Zealand, with prevalence estimates ranging from 20% to 80%. Importantly, these rates were pervasive across all social groups. Finally, one of the most affected regions of the world is Africa, where binge drinking is reported by around one in four women, and in some cases half of all pregnant women.


The prevalence of FAS in the United States ranges from 6 to 9 cases per 1000 live births in the general population, with the risk increasing to 1% to 2% for infants from socioeconomically disadvantaged families and infants in foster care. Rates of FASD, which are more common but harder to detect because of their more subtle presentation, are even higher in the United States, ranging from 24 to 48 cases per 1000 live births or up to 5% of all live-born infants. A lower rate of FASD was found in Alberta, Canada (8.2 to 15.1 cases per 1000 live births), whereas a similar rate was reported for Lazio, Italy (20.3 to 40.5 cases per 1000 live births). Current rates in the United Kingdom are not available. It is important to note that these rates are widely regarded to underestimate the extent of this problem for the methodological reasons listed previously, as well as difficulties with both misdiagnosis and underdiagnosis.


Clinical Features


The diagnostic features of FAS are distinctive and include growth disturbance, characteristic facial anomalies, and neurological abnormalities. Growth disturbance is the hallmark of the disorder, with microcephaly present in nearly all cases. At birth, infants have a distinct pattern of growth restriction, with length often affected more than weight, which is a pattern different from that expected with intrauterine undernutrition. This poor growth persists postnatally, but weight gain is more disturbed than linear growth.


For a diagnosis of FAS, three criteria must be met: (1) prenatal and/or postnatal growth deficiency; (2) the presence of three key facial features—short palpebral fissures, hypoplastic philtrum, and a narrow vermilion lip border (see Fig. 38.3 ); and (3) evidence of structural or functional CNS impairment (discussed later). Confirmation of prenatal alcohol exposure through maternal report or infant toxicology strengthens, but is not required, for a FAS diagnosis. As with all recreational drugs, it is also important to clarify the extent of prenatal exposure to other drugs, such as tobacco, illicit substances, and any prescribed medications. The possibility of other genetic and environmental conditions that share similar dysmorphic features with FAS (e.g., Williams syndrome, fetal hydantoin syndrome, and trisomy 21) or present at older ages with a similar behavioral phenotype (e.g., attention-deficit/hyperactivity disorder [ADHD]) should also be considered.


In addition to the three previously noted defining criteria, a number of other clinical features may also aid FAS diagnosis ( Table 38.3 ). For example, a variety of limb anomalies can be observed in around one-half of these infants. These include abnormal palmar creases and minor joint abnormalities, such as an inability to completely extend the elbows, camptodactyly, and clinodactyly. Cardiac lesions occur in approximately one-half, but these lesions are usually not severe and mostly consist of septal defects, with atrial defects more common than ventricular defects. Minor ear anomalies occur in approximately one-fourth of the children, and hearing loss, primarily conductive and of a mild nature, occurs in 75% of infants. Optic nerve hypoplasia affects 75% of infants, but disturbances of visual acuity are not marked. Other less common anomalies include strabismus, ptosis, micrognathia, cleft palate, railroad track ears (prominent horizontal crus of the helix with prominent and parallel inferior crus of the antihelix), decreased elbow pronation/supination, joint contractures (most commonly incomplete extension of one or more digits), and palmar crease abnormalities.



TABLE 38.3

Clinical Features of the Fetal Alcohol Syndrome a












































































CLINICAL FEATURES APPROXIMATE FREQUENCY (%)
Growth
Prenatal growth deficiency 95
Postnatal deficiency 95%
Central nervous system
Microcephaly 95
Developmental delay 90
Facial
Short palpebral fissures 90
Epicanthal folds 50
Midfacial hypoplasia 65
Short, upturned nose 75
Hypoplastic long or smooth philtrum 90
Thin vermilion of upper lip 90
Limb
Abnormal palmar creases 55
Joint abnormalities 50
Cardiac
Cardiac defects 50
Other
Ear anomalies 25
Conductive hearing loss 75
Sensorineural hearing loss 10
Optic nerve hypoplasia 75
External genital anomalies 30
Cutaneous hemangioma 25

Numbers are rounded off to nearest 5%.

a See text for references.



The less severe FASD conditions are more difficult to diagnose because only some of the features are present. For example, ARND requires confirmation of prenatal alcohol exposure and evidence of structural or functional CNS impairment, such as learning and/or behavior problems, but not facial anomalies. When prenatal alcohol exposure is confirmed but all other criteria for FASD are not met, an infant can be described as exhibiting partial FAS or fetal alcohol effects. These diagnoses may be needed to ensure that a child receives ongoing developmental surveillance in view of high rates of neurodevelopmental impairment in children exposed to alcohol in utero.


Neurodevelopmental Consequences


The nature of a child’s later neurobehavioral problems and their manifestation varies depending on a number of factors. These include (1) the extent of prenatal alcohol exposure (dose, timing) and the severity of the condition (i.e., FAS or fetal alcohol effects); (2) the presence of other developmental risk factors in the child, mother, or family situation; and (3) the age of the child at the time of evaluation. Genetic factors also likely play a role. Nonetheless, reasonable consensus exists regarding the neurobehavioral profile of children with FASD. These problems span a number of developmental domains, including (1) cognition and executive impairment ; (2) language ; (3) behavioral and regulatory problems, especially ADHD ; and (4) motor and visuospatial deficits . The most disabling of these subsequent problems are the intellectual and behavioral impairments. A brief description of the neurobehavioral profile of children with FASD across each impairment domain is provided next.


Cognition and Executive Functioning


Children exposed prenatally to alcohol typically have intelligence quotient (IQ) scores in the low average to borderline range. Children with more dysmorphic features tend to have lower IQ scores, but cognitive problems are not limited to this group. Of those with FAS, around 25% to 50% will experience severe cognitive delay (IQ score <70), with the pooled prevalence of cognitive disability being 97 times higher in infants with FAS than the general population. In addition to global cognitive impairment, deficits in executive function and memory are also evident on neuropsychological testing. Executive deficits include problems with planning and organization, cognitive flexibility/set shifting, working memory, and behavioral inhibition, with parents reporting the greatest difficulty with inhibitory control and problem solving. In terms of memory deficits, problems encoding or memorizing information appear more prominent than problems with recall. Not surprisingly, given this constellation of cognitive impairments, learning problems are very common at school, even after controlling for IQ.


Language Development


Children with FASD are characterized by delays in the acquisition of language and understanding of spoken language. Common difficulties include word comprehension, naming ability, phonological processing, speech production, and articulation errors, resulting in poorer performance on tests of both receptive and expressive language development.


Behavior and Regulatory Problems


Virtually all infants with FASD exhibit serious attentional and behavioral problems, with 70% subsequently meeting clinical criteria for a diagnosis of ADHD. The next most common condition is oppositional defiant/conduct disorder, with children with FASD showing less guilt after misbehaving, less behavioral maturity for their age, and higher levels of antisocial behavior, including cruelty and stealing. Longer-term substance abuse and mood disorders, such as anxiety and depression, are also common. For example, relative to the general population, adults with FASD have more hospital admissions for alcohol abuse (9% vs. 2%) and psychiatric disorders (33% vs. 5%), and are also more likely to be prescribed psychotropic medications (57% vs. 27%).


Motor and Visual Function


Finally, deficits in both fine and gross motor development have been reported in children with FASD and include tremors, weak grasp, poor hand-eye coordination, and impaired postural balance. Visuospatial performance indicates poorer saccadic control, which results in the processing of visual stimuli in a disorganized way.


Neuropathology


The essential nature of the neural disturbance in FASD is an impairment of brain development ( Table 38.4 ). Several aspects of the developmental program appear to be involved, on the basis of neuropathological analysis of brain tissue from children with FAS who died in infancy. In keeping with the microcephaly, micrencephaly is common. The most striking additional abnormalities reported appear to involve neuronal and glial migration. Thus, in the series of infants studied by Clarren and colleagues, the most frequent abnormality was a leptomeningeal neuroglial heterotopia that took the form of a sheet of aberrant neuronal and glial cells covering portions of the cerebral, cerebellar, and brain stem surfaces ( Fig. 38.4 ). Aberrations of brain stem and cerebellar development, in large part related to faulty migration, also have been particularly frequent. Schizencephaly and polymicrogyria are other migrational disturbances observed. In addition, disordered midline prosencephalic formation (e.g., agenesis of the corpus callosum, septo-optic dysplasia, and incomplete holoprosencephaly) has been documented. Other developmental defects have included anencephaly, lumbar meningomyelocele, lumbosacral and sacral meningomyelocele, absent olfactory bulbs or arrhinencephaly , and disturbances of dendritic development.



TABLE 38.4

Major Neuropathological Features of Fetal Alcohol Syndrome a







In order of decreasing frequency:



  • Micrencephaly



  • Migrational abnormalities: neuronal > glial



  • Midline prosencephalic abnormalities: agenesis of the corpus callosum, septo-optic dysplasia, incomplete holoprosencephaly



  • Dendritic abnormalities



  • Disorders of neural tube formation


a See text for references.




Figure 38.4


Fetal alcohol syndrome: neuropathology.

Brain from a 6-week-old infant who was born after a 32-week gestation. (A) Right superolateral view of the cerebrum. Note that nearly all of the left hemisphere is covered by a massive sheet of tissue that crosses the midline and extends onto the superior and medial aspects of the right frontal lobe. (B) Section through the cerebral cortex covered by the aberrant tissue. Extending through a break in the pial surface (upper left of cortex) is the heterotopic sheet of tissue composed of neuronal, glial, and pial elements that have covered the true cortex. (A much smaller break in the pial surface is evident in the lower left of the figure.)

(From Jones KL, Smith DW. The fetal alcohol syndrome. Teratology . 1975;12:1–10.)


Thus it appears that multiple aspects of CNS development can be affected in severe cases. In chronological order, these include neurulation, canalization and retrogressive differentiation, prosencephalic development, neuronal proliferation, neuronal migration, and organizational events (see Chapter 1 , Chapter 2 , Chapter 3 , Chapter 4 , Chapter 5 , Chapter 6 , Chapter 7 ). The time periods of the most frequently reported occurrences (i.e., disorders of neuronal proliferation and migration and of midline prosencephalic development) range from the second to the fifth months of gestation, suggesting that teratogens could be acting either during these time periods or, of course, earlier.


Advanced magnetic resonance imaging (MRI) techniques have helped further define the neuropathology of FASD in the human infant ( Table 38.5 ). Key findings from studies using different imaging modalities are summarized next. However, it is important to note that almost all studies have been conducted in older children, thus limiting information about structural and functional brain abnormalities during infancy, or the way in which prenatal alcohol exposure affects the developing brain over time and age.



TABLE 38.5

Major Central Nervous System Alterations Defined by Magnetic Resonance Imaging in Fetal Alcohol Spectrum Disorders








  • Decreased brain size including lower white and gray matter volumes with enlarged ventricles



  • Disproportionately lower cortical volume of parietal, temporal and frontal lobes



  • Abnormal corpus callosum: partial or complete agenesis or hypoplasia, splenium most affected



  • Decreased volume of basal ganglia, especially caudate nucleus, and hippocampus



  • Decreased volume of cerebellum



  • Lower fractional anisotropy values in regions showing reduced volume suggesting altered microstructure



Volumetric MRI studies indicate both global and regional disturbances in cortical and subcortical development, alterations in cortical thickening, and specific regional vulnerability of the corpus callosum, cerebellum, and basal ganglia, especially the caudate nucleus. Findings confirm smaller brain size, with both white and gray matter volumes affected. In terms of regional abnormalities, a highly consistent finding across studies is the altered shape and area of the corpus callosum, further supporting the vulnerability of midline brain structures to the effects of alcohol. Abnormalities include partial or complete agenesis, underdevelopment, and corpus callosal thinning, particularly in the splenium which is involved in communication between the parietal and temporal lobes. Fig. 38.5 illustrates several examples of corpus callosum abnormalities. These imaging abnormalities have been related to motor function, attention, verbal learning, and executive function. Other subcortical structures affected by prenatal alcohol exposure include the cerebellum and caudate nucleus, which may help explain deficits seen in balance, bimanual coordination, memory, and attention among children with FASD.




Figure 38.5


Corpus callosum abnormalities in newborns with prenatal alcohol exposure.

Severe malformations, including partial agenesis (A and B) and hypoplasia (C) of the corpus callosum, observed in cases of very heavy prenatal alcohol exposure. More subtle callosal abnormalities are observed in most cases of fetal alcohol spectrum disorders.

(From Johnson VP, Swayze VW II, Sato Y, Andreasen NC. Fetal alcohol syndrome: craniofacial and central nervous system manifestations. Am J Med Genet . 1996;61:329–339.)


At the microstructural level, diffusion tensor imaging (DTI) studies indicate that white matter abnormalities also extend to other brain regions beyond the corpus callosum, including the anterior-posterior fiber bundles and the cerebellum. White matter abnormalities have also been seen in the frontal and temporal lobes, as well as several subcortical structures (i.e., globus pallidus, thalamus, and putamen), suggesting more widespread impacts on white matter integrity. While these abnormalities may contribute to the attention, executive function, and other neurobehavioral impairments of children with FASD, studies addressing these links are, as of yet, rare and insufficient to draw clear conclusions.


Taken together, these findings suggest prenatal alcohol exposure has global and regional effects on the development of the CNS. These abnormalities appear to reflect both the adverse effects of alcohol on different organizational events during fetal development, but also the cascading effects of early brain abnormalities on brain growth, myelination, and pruning, suggesting an altered trajectory of brain development in children with prenatal alcohol exposure.


Pathogenesis


The pathogenesis of these disturbances of CNS development has been studied in both clinical and experimental models—primarily the latter. Findings indicate that the adverse effects of alcohol and acetaldehyde, its major metabolite, likely result from some combination of (1) effects on fetal blood flow, (2) fetal malnutrition, (3) direct deleterious molecular effects, and (4) genetic/epigenetic alterations within the rapidly developing CNS. A brief review of each of these processes is given in the following sections.


Fetal Blood Flow


Maternal alcohol exposure has a variable impact on uterine blood flow, depending on the gestation of the fetus as well as the pattern of exposure. However, there is converging evidence to suggest that alcohol alters the development of new blood vessels and vascular remodeling, which are both essential to normal uteroplacental circulation during gestation. There is also evidence that alcohol may alter cerebral oxygen and glucose consumption in the fetus near term gestation, but these effects do not appear earlier in gestation. Further studies are needed to better understand how alcohol consumption affects uteroplacental hemodynamics during different maturational periods of pregnancy.


Fetal Malnutrition


The quality of maternal nutrition as well as the direct physiologic effects of alcohol on the fetus are also likely involved in the pathogenesis of FASD. Specifically, poor maternal nutritional status and vitamin deficiencies are common comorbidities of chronic alcoholism. Alcohol also interferes directly with the absorption, digestion, and utilization of nutrients. Retinol (vitamin A), folate, and zinc, three nutrients that are important for fetal brain development, have been shown to be directly affected by maternal alcohol use. First, retinoic acid, the oxidized form of retinol, plays a pivotal role in the development of the nervous system as well as limb morphogenesis. Alcohol competitively inhibits the oxidation of retinol in the liver. In mouse models, deficiencies in retinoic acid early in gestation alter the expression of the sonic hedgehog gene, resulting in the classic craniofacial and corpus callosum abnormalities characterizing severe FAS. Second, folic acid has numerous roles in the developing nervous system, especially neural tube closure (see Chapter 1 ). Alcohol interferes with folic acid absorption, inhibits its metabolism, and increases excretion. Clinical research has shown a significant reduction in fetal-to-maternal folate ratios associated with chronic maternal alcohol use. Third, zinc is necessary for neurogenesis, neuronal migration, and synaptogenesis. Alcohol induces the zinc binding protein metallothionein in the maternal liver, which sequesters zinc and results in fetal deficiency. Although other primary and secondary nutritional deficiencies may exist in the fetuses of mothers who consume alcohol during pregnancy, the relative importance of these perturbations in the overall pathogenesis of FASD requires additional investigation.


Molecular Effects


Major contributors to the cascade of developmental damage associated with fetal alcohol exposure include (1) excessive cell death (apoptosis), (2) deficient cell proliferation, (3) impaired cell migration, and (4) altered differentiation. In experimental models, neuronal and oligodendrocyte degeneration is a prominent feature of prenatal alcohol toxicity. In particular, alcohol induces two types of cell death in the developing fetal brain. These include apoptosis and necrosis. Necrosis appears to represent a minority of cell death associated with alcohol exposure, typically occurring following binge drinking or alcohol withdrawal. Even low levels of alcohol exposure early in gestation appear to promote apoptosis, via elevation of phospholipase C activity, promotion of intracellular calcium transit, and repression of the transcriptional effector, β-catenin. This is an abnormal process during this stage of development. Later in gestation, apoptosis naturally occurs in approximately one-third of postmitotic neurons. However, in experimental models, alcohol exposure may increase the extent of apoptosis, most likely through N -methyl- d -aspartate (NMDA) receptor blockade and hyperactivation of γ-aminobutyric acid type A (GABA A ) receptors.


Experimental models of fetal alcohol exposure during the equivalent of the human second trimester demonstrate a profound impact on neuronal proliferation, migration, and differentiation. Organization of neural circuitry relies on ongoing electrochemical activity, allowing firing neurons to locate and synapse with other cells as part of activity-dependent network formation. Neurosuppression via NMDA antagonism/GABA A agonism depresses electrochemical activity, suppressing neurogenesis and triggering apoptotic neuronal death in the developing brain. In addition, alcohol impairs the function of insulin-like growth factor receptors, potentially via inhibition of cyclins and cyclin-dependent kinases. This inhibition may help explain delays in progression through the neuronal cell cycle that have been observed in experimental models. Alcohol induces errors in neuronal migration, perhaps through disruption of L1 cell adhesion molecule and interference with glial fibers. Alcohol phosphorylates the cytoplasmic domain of L1 cell adhesion molecule, thus altering the conformation and function of the extracellular domain. These changes not only affect neuronal migration, but also promote aberrant dendritic morphology.


Genetic and Epigenetic Alterations


Maternal and fetal genetics play a prominent role in susceptibility to FASD. Alcohol is metabolized in the liver to acetaldehyde by alcohol dehydrogenase (ADH). Functional polymorphisms in the locus encoding the beta subunit of the Class I ADH (ADH1B) alter the rate of alcohol metabolism. Alcohol is more rapidly metabolized in individuals with the ADH1B*3 allele (15% to 20% of African Americans) compared with individuals with the more common ADH1B*1 allele. This finding appears to have functional significance because prenatal alcohol exposure has been associated with increased attention problems and externalizing behavior in adolescents born to mothers with two ADH1B*1 alleles. In contrast, these differences were not observed in adolescents whose mothers had at least one ADH1B*3 allele.


Fetal genetic differences may also play an important role in outcome . After prenatal alcohol exposure, monozygotic twins are more concordant in outcome than dizygotic twins. Neuronal nitric oxide synthase (nNOS) and oxyguanine glycosylase 1 (OGG1) protect the developing brain from injury induced by alcohol. Mice homozygous for null nNOS or OGG1 have more severe neuronal damage and functional deficiencies following alcohol exposure compared with wild-type mice. In contrast, homozygous mutation of the tissue plasminogen activator and Bax genes are protective against the effects of alcohol on fetal brain development. Further research is required to determine the array of genes that influence the spectrum of fetal alcohol disorders.


Changes in gene expression after prenatal alcohol exposure represent a relatively new field of study. However, there is evidence to suggest that alcohol alters the sequence of genes involved in methylation, chromatin remodeling, protein synthesis, and mRNA splicing. Alcohol also promotes epigenetic changes, including alterations in methylation and acetylation, that give rise to phenotypic changes in animal models. As noted earlier, there is also evidence that epigenetic modifications associated with prenatal alcohol exposure may be transgenerational, placing subsequent generations at increased risk of alcohol dependence. Further study of the mechanisms underlying these important transgenerational effects is needed.


Prevention


Consensus guidelines recommend that the safest choice for a woman is to not drink alcohol during pregnancy. Given that malformation risks are greatest when the fetus is exposed to alcohol during the first weeks and months of gestation, it is critical that women be advised about the risks to their infant as early as possible in the pregnancy. Indeed, advice should ideally be given before pregnancy because most women do not begin prenatal care until after the first important weeks of pregnancy have passed. As discussed previously, most women of childbearing age in the United States consume alcohol, and a small percentage continue during pregnancy. Reduction or cessation of alcohol consumption during any stage of pregnancy benefits the developing fetus. When this cessation is initiated very early in pregnancy, malformations as well as cognitive and motor impairment may be avoided or at least minimized. When it is carried out in midpregnancy, although malformations are not prevented, growth retardation is clearly diminished. Thus there is benefit to be gained, even if cessation of drinking is delayed.


Supplementation to correct the nutritional deficiencies of the fetus (discussed previously) has been investigated. Supplementation with retinoids reduces alcohol-induced ocular phenotypes in experimental models. Similarly, folic acid supplementation may prevent alcohol-induced cardiac defects. Supplementation with other vitamins having a vital role in normal human development also may hold promise, despite the unknown direct impact of alcohol on these molecules. Choline, a B-complex vitamin, has been considered, given that it is the methyl donor for DNA methylation and the precursor for acetylcholine and essential cell membrane constituents. Alcohol induces hypermethylation in the hippocampus and prefrontal cortex, resulting in long-term functional abnormalities; choline may be helpful in competitively reducing these effects. Vitamin E (α-tocopherol) encourages antioxidant activity, with deficiencies in vitamin E having an established role in developmental and behavioral deficits. Oxidative stress has a well-established role in the teratogenic effects of alcohol, promoting caspase-3 activity and subsequent cell death. In multiple experimental models, supplementation with vitamin E reduces cell loss within the brain following prenatal alcohol exposure. Importantly, however, none of the interventions described previously independently eliminate the full spectrum of fetal alcohol effects. In addition, the identification of at-risk women and the challenges of compliance in this population limit the feasibility of these interventions.


Treatment


Newborns with FASD represent a complex, high-risk population. Nutritional supplementation after fetal alcohol exposure has been proposed. As noted earlier, postnatal choline supplementation reduces cognitive deficits and behavioral outcomes in experimental models. Optimizing an infant’s postnatal experiences, educating parents, engaging social support, and mobilizing developmental and educational interventions may improve outcomes. These interventions demonstrate promise in mitigating some of the negative effects of fetal alcohol exposure.




Antiepileptic Drugs


AEDs are among the most common teratogenic drugs prescribed to women of childbearing age. Their primary use is in the treatment of epilepsy, but over half of AED prescriptions are for neuropathic pain, migraine headaches, and psychiatric disorders. A large number of drugs fall into this class, and prescription patterns have changed considerably in the last 2 decades as knowledge about the teratogenic effects of AEDs on the developing fetus and child have increased. Older drugs, such as valproate, phenytoin, phenobarbital, and carbamazepine, have declined in use among women of childbearing age and increasingly have been replaced with newer therapies, such as gabapentin, lamotrigine, levetiracetam, and topiramate. Because all of these drugs are still in use, and with more neonatal and outcome data available for such older drugs as valproate, we review these as exemplars. However, drugs such as trimethadione and paramethadione, which are no longer in clinical use because of their severe teratogenic effects, are not considered.


Prevalence


Approximately a third of individuals treated with AEDs are women of reproductive age. In the United States alone, recent estimates suggest that more than 4 million prescriptions for AEDs are written each year for women of childbearing age. Of these, approximately 17% were for valproate, and two-thirds were for conditions other than epilepsy—most notably psychiatric disorders. Consistent with these national trends, the prevalence of AED treatment for epilepsy and other indications during pregnancy is about 2%. Moreover, there is evidence to suggest that AED prescriptions during pregnancy are increasing. Analysis of data from the US Medication Exposure in Pregnancy Risk Evaluation Program found that in a cohort of almost 580,000 pregnant women, AED prescriptions more than tripled from 1996 to 2007. This change was driven primarily by a fivefold increase in the number of prescriptions for newer AEDs. In contrast, the use of older AEDs such as valproate and benzodiazepines was relatively stable, which was somewhat unexpected given contraindications for the use of these drugs in pregnancy. The most common indications were psychiatric disorders (48%), followed by epilepsy (21%) and pain disorders (22%), regardless of AED type (older vs. newer), year of conception, or gestational timing of exposure. Finally, 13% of deliveries involved AED combination therapy during pregnancy.


Clinical Features


Maternal-Fetal Effects


In addition to the risks of major congenital malformations (see later), a recent meta-analysis of 38 studies across both low and high income countries found that infants born to pregnant women who were treated with AEDs for epilepsy had increased odds of preterm (<37 weeks gestation) birth (OR = 1.16, 95% CI 1.01 to 1.34) and fetal growth restriction (OR = 1.26, 95% CI 1.20 to 1.33). Fetal and neonatal mortality were not increased. However, this finding is contrasted by recent observational data suggesting a higher incidence of intrauterine death associated with maternal AED polytherapy. Microcephaly has also been noted in about 12% of cases. For example, a prospective longitudinal study of 329 pregnant women with epilepsy reported an increased risk of microcephaly at birth and at 12 months of age (12%), but which normalized by 24 months. More research is needed to examine other potential neonatal neurobehavioral outcomes, as well as the extent to which these risks vary by AED, dose, epilepsy type, and therapy regimen (monodrug vs. polydrug exposure).


Major Congenital Malformations


AED exposure in pregnancy results in a constellation of fetal and developmental effects that range in severity from major congenital malformations to subtle variations in normal development. Early, large-scale retrospective studies in the 1970s and 1980s were the first to document the teratogenic effects of AEDs on the developing fetus. Findings suggested a more than twofold increased risk of major malformations in offspring of epileptic women on AEDs versus those not on medication or nonepileptic women. Subsequent studies in the 1990s supported this initial work. These studies focused largely on fetal valproate syndrome and fetal hydantoin syndrome ; the latter is a syndrome associated with phenytoin exposure or structurally similar agents, such as phenobarbital, carbamazepine, and oxcarbazepine. The clinical features of these syndromes are reviewed in Table 38.6 and Figs. 38.6 and 38.7 .



TABLE 38.6

Clinical Features of 63 Cases of the Fetal Hydantoin Syndrome


































































CLINICAL FEATURES PERCENTAGE AFFECTED a
Growth
Prenatal growth deficiency 19
Postnatal growth deficiency 26
Central nervous system
Microcephaly 29
Developmental delay or mental deficiency 38 b
Craniofacial
Large anterior and posterior fontanel 42
Metopic ridging 27
Medial epicanthal folds 46
Ocular hypertelorism 23
Broad and/or depressed nasal bridge 54
Cleft lip and/or palate 5
Limb
Nail and/or distal phalangeal hypoplasia 32
Fingerlike thumb 14
Other
Short neck with or without low hairline 18
Inguinal hernia 14
Bifid or shawl scrotum 33
Cardiac defect 8

Data from Hill RM, Verniaud WM, Horning MG, McCulley LB, Morgan NF. Infants exposed in utero to antiepileptic drugs. Am J Dis Child . 1974;127:645–653; and Hanson JW, Smith DW. The fetal hydantoin syndrome. J Pediatr . 1975;87:285–290.

a Data are expressed as percentage of those patients for whom information is available.


b Includes only those patients 4 years of age or older.




Figure 38.6


Fetal hydantoin syndrome: clinical appearance.

(A and B) Two different infants with the syndrome; note the similar facial appearance, especially the broad, depressed nasal bridge, the widely spaced eyes, the epicanthal folds, the low hairline at the forehead, and the short neck. (C) Hand of an affected newborn. Note hypoplasia of nails and distal phalanges. (D) Hand of an affected child at 1 year of age; some nail growth is evident.

(From Hill RM, Verniaud WM, Horning MG, McCulley LB, Morgan NF. Infants exposed in utero to antiepileptic drugs. Am J Dis Child . 1974;127:645–653.)



Figure 38.7


Fetal valproate syndrome, clinical appearance.

Infant (10 months old) whose gestation was complicated by administration of 2000 mg of valproate daily to the mother: note the microbrachycephaly, apparent hypertelorism, broad flat nasal bridge, small mouth with thin lips (A), and thin, tapering fingers (B).

(From Schorry EK, Oppenheimer SG, Saal HM. Valproate embryopathy: clinical and cognitive profile in 5 siblings. Am J Med Genet A . 2005;133:202–206.)


More recently, given the increased use of AEDs and a growing awareness of adverse effects on the cognition and behavior of children born to treated women, a number of new important data sources have been established. These include (1) large registries of pregnant epileptic women in North America, Scandinavia, the United Kingdom, India, and Australia ; (2) pharmaceutical company registries for specific drugs; (3) the European Surveillance of Congenital Anomalies (EUROCAT) database across 14 countries ; (4) the US National Birth Defects Prevention Study (NBDPS) ; and (5) prospective cohort studies of pregnant women treated with AEDs and their infants. Although not without methodological challenges, including limited information on newer AEDs, insufficient follow-up data, and variable methods used, these efforts have helped advance understanding of the neonatal and longer-term effects of AED exposure during pregnancy.


It is clear from this work that several factors have an influence on the teratogenic effects of AEDs ( Table 38.7 ). First, fetal effects differ for each AED . Valproate appears to be the most teratogenic, followed by phenobarbital and topiramate that carry moderate risks. Newer drugs, at least based on current evidence, appear to be generally less harmful, although not all are completely risk-free ( Table 38.8 ). Second, the dose of drug has been found to influence outcome. Higher maternal doses tend to carry more risk. Third, the timing of AED exposure during pregnancy is also important. This fact poses challenges, given that neural tube closure occurs between weeks 3 and 4 of gestation and systemic organogenesis between weeks 4 and 10. This early period of vulnerability is a time when women are often unaware of their pregnancy. Fourth, the type of epilepsy can further complicate outcomes for the fetus. Specifically, breakthrough seizures during pregnancy may result directly in fetal hypoxia, and consequent falls may increase the risk of premature labor and fetal death. Fifth, the presence of other comorbid conditions , such as psychiatric and endocrine disorders, and the potential for polypharmacy (e.g., antidepressants) may further increase the risk for the infant born to a woman with epilepsy. Sixth and relatedly, fetal effects can vary depending on whether one or multiple AEDs are prescribed . Delineation of the teratogenic potential of each drug or drug combination is very difficult based on current evidence, especially for newer drugs. However, clear evidence exists to show that polydrug therapy is associated with a higher risk of congenital malformations than monotherapy, especially if polydrug therapy includes valproate. Additional evidence is needed to characterize the impacts of different polytherapy combinations, to assist the clinical management of women (and their infants) when monotherapy is ineffective for seizure control. Finally, there is increasing evidence to suggest that maternal genetic factors relating to the cerebral disorder underlying the epilepsy or other heritable conditions in the family increase the risk of congenital malformations. For example, a parental history of major congenital malformations increases the fetal risk more than fourfold, and the birth of a previous child with malformations when on the same AED increases the risk 17-fold.



TABLE 38.7

Factors Influencing Teratogenic Effects of Antiepileptic Drugs








  • Mechanism of the specific drug



  • Dose



  • Timing of exposure during pregnancy



  • Type of epilepsy and level of seizure control



  • Monotherapy versus polytherapy



  • Presence of comorbid conditions and polytherapy



  • Maternal genetic factors



TABLE 38.8

Relative Risk of Major Malformations Associated With Different Antiepileptic Drugs Used During Pregnancy






































AGENT RELATIVE RISK OF MAJOR MALFORMATIONS COMPARED WITH UNEXPOSED
Valproate ( N = 323) 9.0 (3.4–23.3)
Hydantoins and agents with similar structure
Phenytoin ( N = 416) 2.6 (0.9–7.4)
Phenobarbital ( N = 199) 5.1 (1.8–14.9)
Carbamazepine ( N = 1033) 2.7 (1.0–7.0)
Oxcarbazepine ( N = 182) 2.0 (0.5–7.4)
Newer agents
Topiramate ( N = 359) 3.8 (1.4–10.6)
Lamotrigine ( N = 1562) 1.8 (0.7–4.6)
Levetiracetam ( N = 450) 2.2 (0.8–6.4)
Gabapentin ( N = 145) 0.6 (0.07–5.2)

Adapted from Hernandez-Diaz S, Smith CR, Shen A, et al. Comparative safety of antiepileptic drugs during pregnancy. Neurology . 2012;78:1692–1699.


With respect to individual AEDs, while the data are not perfectly consistent, several conclusions appear justified regarding the malformation and neurobehavioral risks associated with AED exposure during pregnancy. The most common major malformations reported in recent large series are neural tube defects, cardiac anomalies, oral clefts, hypospadias and other genitourinary anomalies, gastrointestinal anomalies, and skeletal deficits ( Table 38.9 ). The incidence of these malformations ranges from 2% to 10%, depending on the AED used, in contrast to a malformation rate of 1% to 3% in the general population.



TABLE 38.9

Types of Major Congenital Malformation by Antiepileptic Drugs






























































DRUG HYPOSPADIAS a NEURAL TUBE DEFECTS CARDIOVASCULAR ANOMALIES ORAL CLEFTS
No antiepileptic drugs ( N = 206,224) 0.04 0.12 0.33 0.11
Valproate ( N = 323) 3.1 1.2 2.5 1.2
Hydantoins and agents with similar structure
Phenytoin ( N = 416) 0.0 0.0 0.96 0.48
Phenobarbital ( N = 199) 0.97 0.0 2.5 2
Carbamazepine ( N = 1033) 0.19 0.29 0.29 0.48
Newer agents
Lamotrigine ( N = 1562) 0.0 0.13 0.19 0.45
Levetiracetam ( N = 450) 0.0 0.22 0.22 0.0
Topiramate ( N = 359) 1.1 0.0 0.28 1.4

Restricted for malformations identified before 5 days of age, including elective terminations. Confirmed by review of medical records. Some infants had more than one defect.

Adapted from Hernandez-Diaz S, Smith CR, Shen A, et al. Comparative safety of antiepileptic drugs during pregnancy. Neurology . 2012;78:1692–1699.

a Excludes mild glandular hypospadias. Restricted to male infants.



Neonatal Effects


In relation to the neonatal effects of AEDs, neonatal withdrawal has been described. This has been reported most extensively with phenobarbital. Onset generally occurs around 7 days of age, which is understandable in view of the slow elimination of phenobarbital in the immediate neonatal period due to its half-life of several days. The major features consist primarily of CNS phenomena, including jitteriness, overactivity, disturbed sleeping, excessive crying, hyperreflexia, and disturbed sucking. Overt gastrointestinal phenomena, such as diarrhea, may occur but usually are not prominent. The symptoms may appear after the infant is discharged from the hospital, and the infant’s irritability may be mistaken for colic or some other extraneural cause. Symptoms often worsen over several weeks and persist for several months .


Neonatal withdrawal from other AEDs has been less extensively described. Recent reports describe withdrawal symptoms after prolonged in utero exposure to gabapentin. Symptoms developed within 24 hours of life, including sneezing, irritability, jitteriness, and loose stools. Given the increasing utilization of other novel AEDs, withdrawal should remain in the differential diagnosis for exposed infants presenting with similar symptomatology.


An additional potential complication for the newborn infant associated with maternal AED treatment using a select group of AEDs is neonatal hemorrhage . The drugs incriminated are hepatic enzyme inducers and have included hydantoins , barbiturates , primidone (which is metabolized to phenobarbital in vivo), and carbamazepine . In one series of 111 infants born to epileptic women treated with phenytoin or phenobarbital, 8 exhibited severe bleeding. In this syndrome, the infant developed hemorrhage shortly after birth. This early onset is unlike hemorrhagic disease of the newborn secondary to vitamin K deficiency. Sites of hemorrhage are, in order of decreasing frequency, skin, liver, gastrointestinal tract, intracranial sites, and thorax. Intracranial hemorrhage has been reported in 20% to 25% of the cases with any bleeding. The course may be fulminating, and approximately 40% of reported infants have died. Clotting studies demonstrate diminution of the vitamin K–dependent clotting factors (factors 2, 7, 9, and 10), with prolongation of either the prothrombin time or partial thromboplastin time, or both. Vitamin K levels in cord blood are depressed, as evidenced by the presence of PIVKA-II (a protein, induced by vitamin K absence, of factor II), an incompletely carboxylated, functionally defective form of factor II (prothrombin). Indeed, in one study of 24 infants born to mothers on antiepileptic therapy during pregnancy, 13 (54%) had detectable levels of PIVKA-II, and of the four infants exposed to valproate (none of whom had detectable levels of PIVKA-II), the incidence was 65%. Moreover, vitamin K levels also were depressed in the infants. These findings suggested increased degradation of vitamin K with impaired action of vitamin K on hepatic production of prothrombin. The latter occurs because of insufficient carboxylation of the glutamic acid residues of prothrombin, a posttranslational event that requires vitamin K. The pathogenetic mechanism by which some AEDs lead to vitamin K deficiency may relate primarily to increased degradation of vitamin K by fetal hepatic microsomal mixed-function oxidase enzymes, known to be inducible by phenytoin, phenobarbital, primidone, and carbamazepine. The similarity in structure of these so-called enzyme-inducible AEDs is shown in Fig. 38.8 . Nevertheless, in recent years this disorder appears to have become rare. Three reports ( n totals = 105, 204, and 662) have shown no increased incidence of hemorrhagic complications among women treated with various combinations of phenobarbital, phenytoin, primidone, and carbamazepine during pregnancy. The women were not treated with vitamin K during pregnancy, although the infants did receive vitamin K at birth. Whether the decline in incidence in this disorder relates to the diminishing use of polytherapy or increasing use of carbamazepine or related factors is unclear.




Figure 38.8


Chemical structure of important anticonvulsant drugs.

Note the structural similarities of phenobarbital, phenytoin, and carbamazepine; each is composed of a basic five- or six-membered ring and a side chain substitution containing phenol groups. Valproic acid differs markedly from the other anticonvulsant drugs and is essentially a short-chain fatty acid.


Neurodevelopmental Consequences


Longitudinal follow-up data describing the longer-term outcomes of children exposed prenatally to AEDs is limited to a few studies. As illustrated in Table 38.10 , sample sizes tend to be small for individual AEDs and especially some newer AEDs. Outcomes are also incomplete, with an emphasis on early global cognitive development assessed using the Griffth and Bayley Scales, and/or general intelligence at later ages. While helpful in evaluating general function and risk, these measures provide no or limited information about other important aspects of neurodevelopmental functioning, such as executive function, memory, attention, and language, at least until recently. With this in mind, existing research relating to each outcome domain is summarized as follows.



TABLE 38.10

Impact of Antiepileptic Drugs on Early Childhood Development and Intelligence Quotient



























































































EARLY CHILDHOOD DEVELOPMENT SCHOOL AGE INTELLIGENCE QUOTIENT
N N N N
DRUG DRUG EXPOSED NOT DRUG EXPOSED MEAN DIFF (95% CF) DRUG EXPOSED NOT DRUG EXPOSED MEAN DIFF (95% CF)
Valproate 42 a 230 −8.00 (−12.79 to −3.21) 76 552 −8.94 (−11.96 to −5.92)
Hydantoins and agents with similar structure
Phenytoin 20 44 −0.12 (−7.54–7.30) 5 201 4.80 (−4.10–13.70)
Phenobarbital 14 201 −6.80 (−12.90 to −0.70)
Carbamazepine 50 a 79 −5.5 (0.34–10.83) 150 552 −0.03 (−3.08–3.01)
Newer agents
Levetiracetam 51 a 97 1.09 (−2.81–4.99)
Lamotrigine 34 a 230 −1.0 (−5.75–3.75) 29 210 −4.0 (−8.32–0.32)
Monotherapy 138 a 230 −4.00 (−6.86 to −1.14) 182 391 −1.30 (−4.12–1.51)
Polytherapy 30 a −6.00 (−13.27–1.27) 105 −8.57 (−11.77–5.38)

General development assessed with either Griffiths or Bayley depending on measure used most commonly across studies, with the largest N. Measure is Bayley unless otherwise marked ( ). Intelligence quotient was assessed with age appropriate standardized measures of intelligence.

Data from Bromley R, Weston J, Adab N, et al. Treatment for epilepsy in pregnancy: neurodevelopmental outcome in the child. Cochrane Database Syst Rev . 2014;CD010236.


Cognition and Executive Functioning


Prospective studies have consistently documented the negative effects of valproate on cognitive development, with an 8- to 9-point IQ decrement. Of note, significant effects appear to relate largely to high-dose valproate exposure (>800 mg daily). The NEAD study also found that higher doses of valproate exposure during pregnancy were associated with poorer memory and executive function in offspring at age 6. In addition, evidence consistently suggests that monotherapy may be less harmful than polytherapy, although historically polytherapy has generally included valproate (see Table 38.10 ). Findings for other AEDs or combinations are mixed and insufficient.


Language Development


There is some suggestion that, based on IQ, verbal abilities may be more impaired than nonverbal abilities in children born to mothers treated with valproate. Of note, lower doses of valproate appear to be associated with less severe effects, although performance is still negatively affected compared with controls. Carbamazepine appears to exert similar negative effects as lower doses of valproate. In contrast, studies suggest that language abilities are comparable to control children for other AEDs, including phenytoin, lamotrigine, topiramate, gabapentin, and levetiracetam. However, these conclusions are based on studies with small sample sizes and unstandardized language measures.


Behavior and Regulatory Problems


A small series of studies suggest that children exposed prenatally to valproate are at increased risk of behavior problems based on screening measures, such as the Vineland Adaptive Behavior Scales and Strengths and Difficulties Questionnaire. Virtually nothing is known about specific risks for mental health problems, such as ADHD and anxiety disorders. One exception is a population-based Danish study that has linked valproate to an increased risk of autism spectrum disorder (ASD).


Motor and Visual Function


There is little evidence to suggest that prenatal AED exposure is associated with later motor problems, at least on global measures of psychomotor development. The one exception is a small study of 56 AED-exposed and 77 nonexposed newborns which found that exposed infants had lower limb and axial tone, and were less irritable than nonexposed infants. However, little evidence exists to suggest that prenatal AED exposure is associated with later motor problems, at least on global measures of psychomotor development.


Neuropathology


The nature of the neural disturbance associated with intrauterine AED exposure in the human infant has yet to be well characterized. Experimental models have shown decreased brain weight after phenobarbital, phenytoin, valproic acid, or benzodiazepine exposure. In contrast, these morphologic effects have not been observed after exposure to clinically relevant doses of carbamazepine, lamotrigine, or levetiracetam. Neuroimaging studies with human infants or older children exposed prenatally to AED have not been reported. A recent study examining early cortical activity with electroencephalography (EEG) in a small series of AED exposed newborns ( N = 56; predominantly oxcarbazepine/carbamazepine monotherapy or polytherapies containing these agents) found both individual alpha oscillatory bouts and wider band spectra in these infants, as well as differences in interhemispheric synchrony, suggesting that functional brain networks may be altered as a result of prenatal AED exposure.


Pathogenesis


As with alcohol exposure, direct effects on fetal perfusion, fetal malnutrition, direct molecular effects of individual agents or their metabolite(s), and genetic variables are all likely contributors to the developmental anomalies associated with intrauterine exposure to AEDs ( Table 38.11 ). These impacts may be further modulated by genetic factors. While the mechanisms of adverse effects of older agents, such as phenobarbital and phenytoin, have been evaluated extensively, less insight exists regarding newer agents, such as levetiracetam and lamotrigine, which are now more widely used in the management of epilepsy during pregnancy.



TABLE 38.11

Pathogenesis of Anomalies Associated With Intrauterine Exposure to Antiepileptic Drugs















Valproate



  • Inhibition of folate utilization



  • Neuronal apoptosis



  • Generation of reactive oxygen species



  • Generation of epoxides

Hydantoins and agents with similar structure (phenytoin, phenobarbital, carbamazepine, oxcarbazepine)



  • Direct cardiotoxic effects



  • Increased degradation of vitamin K



  • Inhibition of folate uptake



  • Neuronal apoptosis



  • Generation of reactive oxygen species



  • Generation of epoxides

Newer antiepileptic drugs (lamotrigine, levetiracetam, topiramate, gabapentin)



  • Inhibition of folate utilization



  • Generation of epoxides



Fetal Blood Flow


AEDs have been found to have direct cardiotoxic effects in fetal experimental models. Phenytoin and phenobarbital, specifically, inhibit the potassium channel IKr at clinically relevant concentrations. Via this mechanism, phenytoin induces bradycardia and transient ventricular arrhythmias in experimental models. These fetal cardiac effects may result in episodic hypoxia, leading to hemorrhage and necrosis of developing tissues. Preliminary data suggest lamotrigine has similar potential. The relevance of this adverse effect for other AEDs requires further investigation.


Fetal Malnutrition


There is also a suggestion that nutritional deficiencies may potentially modulate the teratogenic effects of AEDs. As discussed previously, hepatic enzyme inducers, including phenytoin, phenobarbital, and carbamazepine, have been implicated as a risk factor for neonatal hemorrhage. This effect may relate to increased degradation of vitamin K by fetal hepatic microsomal mixed-function oxidases inducible by these agents.


Women with epilepsy and abnormal pregnancy outcomes also have significantly lower blood folate concentrations. Valproic acid functions as a noncompetitive inhibitor of cellular folate receptors and increases methylenetetrahydrofolate reductase activity, which is a crucial determinant of folate utilization in the methyl cycle. Phenobarbital and carbamazepine decrease the expression of reduced folate carrier (RFC), potentially reducing folate uptake and transfer to the fetus. Newer AEDs also alter folate uptake and transfer. Specifically levetiracetam and lamotrigine decrease placental expression of RCF and folate receptor α, respectively. In rodent models of fetal valproic acid or hydantoin exposure, treatment with folate reduces the risk of congenital malformation. Unfortunately, studies in women with epilepsy do not suggest any protective benefits of folic acid supplementation before conception. In this context, preconceptional folic acid supplementation is recommended in a similar fashion as it is for all women of childbearing age for the prevention of neural tube defects (see Chapter 1 ). The role of higher doses in women with epilepsy has yet to be determined.


Molecular Effects


Several AEDs induce widespread neuronal apoptosis in a manner similar to fetal alcohol exposure. As described in detail earlier, the mechanism of alcohol-induced apoptotic neurodegeneration is likely mediated through NMDA receptor blockade and hyperactivation of GABA A receptors . Similar neuronal apoptosis has been described in newborn rodents after exposure to phenytoin, phenobarbital, diazepam, and valproic acid, all of which directly activate GABA A receptors or increase levels of GABA through sodium channel blockade or inhibition of catabolism. Of concern, recent in vitro data suggest similar potential with oxcarbazepine. Interestingly, although carbamazepine appears to exert antiepileptic activity through a similar mechanism, apoptosis is only augmented in rat pups exposed at postnatal day 7 at concentrations that far exceed those used in clinical practice. Notably, AEDs with a less clear impact on GABA, including levetiracetam and lamotrigine, appear to induce little to no apoptosis in experimental models. However, with the exception of levetiracetam, these safer agents do potentiate cell death when used in combination with proapoptotic AEDs.


An additional mechanism of teratogenesis may be the generation of reactive oxygen species (ROS). Experimental models demonstrate the ability of fetal peroxidases to rapidly bioactivate hydantoin derivatives (phenytoin and carbamazepine) to free radical intermediates that initiate the formation of ROS. Valproic acid also increases the formation of ROS in embryos, although the mechanism has not been clearly described. In vitro models demonstrate ROS generation in astrocytes after exposure to gabapentin, oxcarbazepine, and topiramate, but minimal effects from levetiracetam and lamotrigine. ROS generation interferes with development by direct oxidative damage to cellular macromolecules, including DNA and RNA. In addition, ROS generation leads to dysregulation of signal transduction, thereby triggering apoptotic or necrotic cell death.


Later in gestation, the fetus develops an additional ability to generate toxic metabolites via the cytochrome P-450 enzyme system ( Fig. 38.9 ). Several AEDs, including phenytoin, phenobarbital, carbamazepine, lamotrigine, and valproic acid, undergo metabolism in multiple phases. The first phase is generally a two-step hydroxylation catalyzed by cytochrome P-450. The first step produces a highly reactive epoxide that can bind nucleic acids and impair developmental processes. Multiple systems exist to detoxify harmful epoxides, including epoxide hydrolase, resulting in a hydroxylated molecule. Glucuronidation of these molecules in a later phase produces a highly water-soluble metabolite suitable for urinary excretion. Increased exposure to epoxides in experimental models increases the risk of teratogenesis. For example, exogenous inhibition of microsomal epoxide hydrolase produces a higher incidence of fetal demise and anatomical abnormalities after phenytoin exposure in mice. Importantly, several AEDs (carbamazepine, oxcarbazepine, phenobarbital, phenytoin, valproic acid) induce the cytochrome P-450 system, increasing the generation of reactive epoxides, and valproic acid specifically inhibits microsomal epoxide hydrolase (see Fig. 38.9 ). Many investigators have proposed these interactions as a potential explanation for the increased incidence of teratogenesis observed with polytherapy compared with monotherapy. Of note, inhibitors of the cytochrome P-450 system decrease the incidence of teratogenesis in experimental models. Importantly, this modulatory effect of polytherapy does not occur when using agents with no impact on the cytochrome P-450 system.




Figure 38.9


Potential mechanism of teratogenic effect of major anticonvulsant drugs.

Generation of epoxide derivative is catalyzed by a mono-oxygenase that is induced by the drugs themselves (i.e., autoinduction). The potentially teratogenic epoxide derivative must be degraded by a hydrolase to the apparently harmless dihydrodiol derivative. Impaired activity of the hydrolase, because of either genetic variability or exogenous factors (e.g., valproic acid), could cause the epoxide derivative to accumulate and exert teratogenic effects.


However, recent evidence suggests apoptosis may not be the fundamental mechanism of neurodevelopmental alterations from AEDs. These experimental models in mice highlight the importance of neocortical dysgenesis through alteration of proliferation and differentiation of neural progenitor cells. Specifically, valproic acid inhibits histone deacetylase, alters the expression of G1-phase regulatory proteins, inhibits neural progenitor cells from exiting the cell cycle during neocortical histogenesis, and increases the production of projection neurons in superficial neocortical layers. These findings highlight our incomplete understanding of the fundamental mechanisms underpinning well-described syndromes associated with valproic acid and other AEDs.


Genetic Factors


Genetic factors appear to mediate the incidence and severity of anomalies after prenatal AED exposure. Syndromic features are dramatically more likely to recur in children born to mothers with a previously affected offspring compared with mothers whose previous offspring showed no observable effects. Maternal metabolic variability may explain this observation to some extent. Cytochrome P-450 polymorphisms are common and result in more rapid production of toxic epoxides in some individuals. In addition, epoxide hydrolase exhibits polymorphisms that result in large variability in the capacity to detoxify metabolic intermediates. Observational studies of pregnant females taking phenytoin have found that lower epoxide hydrolase activity correlates with fetal hydantoin syndrome in offspring ( Fig. 38.10 ). However, the fact that these reactive hydrophilic metabolites most likely do not cross the placenta raises questions regarding these associations. In this case, maternal genetics may represent a proxy for fetal genetics, which clearly play a role in the risk of adverse outcomes associated with prenatal AED exposure. Children with major congenital anomalies after phenytoin exposure are also characterized by abnormal metabolite detoxification in experimental tests conducted in vitro after birth. Additional fetal genetic factors may include a sensitivity to the cardiac effects of AED exposure.




Figure 38.10


Epoxide hydrolase activity in amniocyte samples from 19 prospectively monitored fetuses whose mothers were administered phenytoin during the pregnancy.

The samples from the four fetuses subsequently noted postnatally to exhibit the clinical features of fetal hydantoin syndrome are shown in the front row of bars, and the 15 samples from fetuses subsequently confirmed not to have the characteristic features of the syndrome are shown in the rear row of bars. Note the markedly lower epoxide hydrolase activities in the fetal samples from infants who exhibited the clinical features of fetal hydantoin syndrome.

(From Buehler BA, Delimont D, Van Waes M, Finnell RH. Prenatal prediction of risk of the fetal hydantoin syndrome. N Engl J Med . 1990;322:1567–1572.)


Prevention


As described, AED exposure during pregnancy increases the risk of congenital anomalies. However, maternal seizures put both the mother and fetus at risk for physical injury. Therefore preconception planning is ideal to allow careful consideration of the risks and benefits of transitioning the mother to a less teratogenic treatment plan. Expert recommendations suggest the avoidance of valproic acid, phenytoin, and phenobarbital. In addition, the avoidance of AED polytherapy is recommended if possible. However, experts emphasize that polytherapy may be preferable to monotherapy with valproic acid and may be necessary with newer AEDs to achieve seizure control. Based on available evidence, it appears levetiracetam and lamotrigine are the preferred AEDs in pregnancy, followed by carbamazepine and oxcarbazepine.


Treatment


Newborns with withdrawal from phenobarbital respond well to postnatal phenobarbital therapy, starting on a dose of 3 to 5 mg/kg per day and then tapering very slowly, over months, as symptoms are controlled. A single case report describing gabapentin withdrawal noted a positive response to postnatal gabapentin therapy, at a maximum dose of 5 mg/kg every 12 hours, followed by gradual weaning over 48 days. As discussed previously, the incidence as well as treatment of withdrawal from other AEDs is less clear. Similar to newborns with FASD, newborns exposed to any AED in utero may benefit from developmental care.


The management of newborn infants whose mothers have been on AEDs during pregnancy to prevent neonatal hemorrhage has previously been recommended as follows: (1) consideration of delivery by cesarean section if a difficult or traumatic delivery is anticipated, (2) administration of oral vitamin K (10 mg/day) to the mother before delivery during the last month of pregnancy (parenteral vitamin K should be administered as soon as possible after the onset of labor if oral vitamin K was not given), (3) administration of vitamin K to the infant intravenously immediately after birth, (4) administration of fresh frozen plasma to the infant if clotting studies are distinctly abnormal, and (5) consideration of exchange transfusion if hemorrhage ensues. Oral supplementation of vitamin K 1 to the mother for 2 to 4 weeks before delivery prevented neonatal coagulation defects in one study. However, in view of the recent studies showing no increase in the risk of hemorrhage in infants whose mothers did not receive vitamin K during pregnancy, its use has been questioned. A more selective approach may be preferable, such as in pregnancies where there is a higher likelihood of hemorrhage due to trauma, prematurity, or other related risk factors. At least based on current evidence, prenatal vitamin K appears to pose no risks to the fetus, but may have potential benefits.




Stimulants


Two classes of stimulants that are used recreationally and have been shown to affect fetal development are discussed here. These include cocaine and methamphetamines . The effect of cocaine on the fetus became an important issue during the crack cocaine epidemic of the 1980s. Cocaine is benzoylmethylecgonine, an alkaloid derived from the leaves of the Erythroxylon plant species. It is available in two forms: cocaine hydrochloride and highly purified cocaine alkaloid (free base). The latter is derived from the former principally by alkali extraction. Cocaine hydrochloride is heat labile but water soluble, and therefore is generally administered by nasal insufflation, orally, or intravenously. Cocaine alkaloid is heat stable but highly water insoluble and therefore is generally administered by inhalation (smoking). The cocaine alkaloid preparation is also called crack because of the popping sound made by the heated crystals.


In response to early concerns about the possibility of a “crack baby” syndrome, several large prospective longitudinal studies were initiated. These clinical studies, as well as experimental observations, have helped inform understanding of the neurological and developmental effects of prenatal cocaine exposure on the developing fetus and infant. Findings from these studies demonstrate that cocaine has the potential to cause, directly and indirectly, alterations in brain structure and associated behavioral functions in the child. However, in general, these effects were more subtle than first suggested. These longitudinal studies were also very important for the field in general because they helped advance methods and models of studying prenatal drug effects. Specifically, these studies helped (1) develop improved strategies for addressing the effects of confounding polydrug use and lifestyle factors; (2) establish more reliable and valid measures of drug exposure(s) and child outcomes assessments; (3) highlight the importance of postnatal environmental influences; and (4) emphasize the need for a life course perspective.


Because prenatal methamphetamine abuse is relatively recent, less is known about the outcomes of newborns after methamphetamine (Desoxyn in medical use; “meth” in the illicit setting) or amphetamine (Adderall, Dexedrine, Dextrostat, and Vyvanse in medical use; “speed” in the illicit setting) exposure. In contrast to cocaine, fewer studies have examined the effects of prenatal methamphetamine exposure on pregnancy, infant, and child outcomes. With the exception of a Swedish cohort ( n = 66 exposed infants) and the US–New Zealand Infant Development, Environment, and Lifestyle (IDEAL) study ( n = 204 exposed and 208 unexposed infants), most research in this area is cross-sectional and/or based on retrospective chart review. Thus, especially for longer-term outcomes, conclusions are heavily dependent on one, if not two, longitudinal studies (see later). The mechanism of action of methamphetamine is similar to cocaine, promoting dopamine release through inhibition of reuptake transporters. However, there are also several important differences between the two drugs. First, the half-life of methamphetamine is nearly four times longer than cocaine, resulting in a greater window of drug exposure for the fetus during pregnancy. Second, although both amphetamines and cocaine exert their effect by blocking the reuptake of dopamine, amphetamines also increase the release of dopamine . Despite these differences and based on available information, there appears to be significant overlap between the pathologic and developmental effects observed after exposure to these agents.


Prevalence


The exact prevalence of crack/cocaine use in pregnancy is not known, although estimates generally suggest that rates are declining relative to the rates for other drugs of abuse. Estimates vary widely depending on the method used to assess drug exposure, with hair and meconium analysis yielding higher rates than either self-report (which is susceptible to underreporting) or urine analysis (which captures only a short window of time during the pregnancy). For example, 11% of women self-reported illicit drug use in a large local cohort of 3000 urban women in Detroit; however, a 31% prevalence of cocaine was detected on the basis of infant meconium analysis shortly after delivery. These rates and discrepancies are not confined to urban areas ( Table 38.12 ). More recent studies suggest a declining rate nationally (~10% prevalence by meconium; ~7.5% by self-report), but demonstrate that cocaine remains a common drug of abuse, especially in some urban areas of the United States.



TABLE 38.12

Incidence of Cocaine Use in a Suburban US Hospital
















PRIVATE ( n = 366) CLINIC ( n = 134)
Cocaine use (meconium) 6.3% 26.9%
Cocaine use (maternal report) 0% 4.0%

Data from Schutzman DL, Frankenfield CM, Clatterbaugh HE, Singer J. Incidence of intrauterine cocaine exposure in a suburban setting. Pediatrics . 1991;88:825–827.


Although amphetamine-based stimulants have a long history of use for fatigue and weight loss, it was not until the 1990s and early 2000s that methamphetamine abuse became a public health problem. This largely occurred as a consequence of the advent of illicit home manufacturing of the drug. Commonly known as ice or crystal , this crystallized form is heated and inhaled as a vapor, intensifying the euphoric (and addictive) effects of the drug. These home-based manufacturing laboratories initially emerged on the West Coast and in deprived rural areas of the United States, but then quickly spread across the North American continent and to regions of Southeast and East Asia, Australasia, South Africa, and parts of Europe, such as the Czech and Slovak Republics.


Limited information exists regarding the prevalence of methamphetamine use during pregnancy. Estimates in the United States vary from 0.7% to 5.2%. Importantly, many of these women are also subject to high levels of social and psychological disadvantage. For example, a large study of United States ( n = 127) and New Zealand ( n = 97) women, who either reported using methamphetamine during pregnancy or whose infant’s meconium tested positive for the drug, found that social disadvantage, single motherhood, and delayed prenatal care were relatively common. A large proportion also had comorbid psychiatric (48% United States, 43% New Zealand) and other substance abuse disorders (71% in both United States and New Zealand women), highlighting the complex presentation of these women and infants.


Clinical Features


Maternal-Fetal Effects


Cocaine use during pregnancy has deleterious effects on both the mother and the fetus ( Table 38.13 ). The specific features and the magnitude of these effects are relatively consistent across studies. A recent meta-analysis of 31 studies found that cocaine use during pregnancy was associated with significantly higher odds of preterm birth (OR = 3.4), low birth weight (OR = 3.7), and being born small for gestational age (OR = 3.2). On average, cocaine-exposed infants were born 1.5 weeks earlier and were almost 500 g lighter than nonexposed infants. Findings are mixed as to whether these infants catch up in growth, with one study finding no differences in the height and weight of exposed children between the ages of 1 and 6 years, and another reporting that they were in fact heavier at age 13 months, potentially indicating some postnatal compensation for intrauterine growth deficiencies.



TABLE 38.13

Odds of Selected Neonatal Outcomes Associated With Intrauterine Cocaine and Methamphetamine Exposure










































COCAINE AMPHETAMINE
N INFANTS (EXPOSED AND NOT EXPOSED) OR (95% CI) N INFANTS
(EXPOSED AND NOT EXPOSED)
OR (95% CI)
Birth characteristics a
Preterm birth (<37 weeks’ gestation) 39,860 3.4 (2.7–4.2) 62,070 4.1 (3.1–5.6)
Low birth weight (<2500 grams) 38,796 3.7 (2.9–4.6) 26,132 4.0 (2.5–6.4)
Small for gestational age (<10th percentile for weight) 28,098 3.2 (2.4–4.3) 4,383 5.8 (1.4–24.1)
Neurobehavioral features b
123,101 Birth—3 days
No differences
3 weeks old
↑ Excitability
↓ State regulation
291,268 Birth—5 days
↑ Stress abstinence
↓ Quality of movement

OR , Unadjusted Odds Ratio (95% Confidence Interval).

Data from Tronick EZ, Frank DA, Cabral H, Mirochnick M, Zuckerman B. Late dose-response effects of prenatal cocaine exposure on newborn neurobehavioral performance. Pediatrics. 1996;98:76–83 and LaGasse LL, Wouldes T, Newman E, et al. Prenatal methamphetamine exposure and neonatal neurobehavioral outcome in the USA and New Zealand. Neurotoxicol Teratol . 2011;33:166–175.

a Data from Gouuin K, Murphy K, Shah PS. Effects of cocaine use during pregnancy on low birthweight and preterm birth: systematic review and metaanalyses. Am J Obstet Gynecol . 2011;204:340.e1–e12 and Ladhani NN, Shah PS, Murphy KE. Prenatal amphetamine exposure and birth outcomes: a systematic review and metaanalysis. Am J Obstet Gynecol . 2011;205:219.e211–e217.


b Based on NBAS (Tronick et al.) and NNNS (LaGasse et al.) exam. Findings reported are after adjustment for covariates.



Similarly, a recent meta-analysis of 10 studies found that methamphetamine exposure during pregnancy was associated with increased risks of preterm birth (OR = 4.1), low birth weight (OR = 4.0), and being born small for gestational age (OR = 5.8; see Table 38.13 ). Only three studies, two of which were from the same cohort, examined the extent to which these risks persisted after other confounding factors were taken into account. Findings from these studies showed that while risks were lower (OR range = 1.3 to 2.5), they remained statistically significant and consistent with unadjusted findings.


Neonatal Effects


The neonatal features of infants exposed to cocaine in utero are not characterized by a distinct constellation of craniofacial or other anomalies, but neurobehavioral features are common. The most consistent findings from both experimental and human infant studies indicate neurobehavioral and motor abnormalities. These consist principally of poorer state regulation, impaired arousal and altered sleep-wake states, poorer visual and auditory attentiveness, and increased periods of excitable and agitated behavior. Additional motor features include extensor and flexor hypertonicity, hyperreflexia, coarse tremor, and increased motor activity. Neither set of problems generally requires therapy, but rather appears to improve with time, especially the motor features. These adverse effects are even more evident when the fetus is exposed to other drugs of abuse, such as opioids, in addition to cocaine. The likelihood that these neurobehavioral characteristics are related to a direct effect of cocaine is supported by the demonstration of a dose-response relationship between the extent of maternal cocaine use during pregnancy and the severity of neonatal neurobehavioral and motor outcomes shortly after birth.


Methamphetamine exposed newborns are also characterized by a number of neurobehavioral features similar to those seen in cocaine-exposed infants. Neonatal intensive care unit (NICU) admission is often required. However, similar to cocaine, maternal methamphetamine use during pregnancy is generally not associated with a withdrawal syndrome that requires pharmacological treatment. Common features of methamphetamine withdrawal include a weaker and less coordinated suck, disorganized-state behavior, poorer quality of movement, and increased physiological/CNS stress. A dose-response relationship has been demonstrated for arousal and excitability. Severe symptoms requiring hospitalization resolve rapidly, with full resolution generally occurring by age 1 month.


An additional complication of prenatal cocaine exposure is sudden infant death syndrome (SIDS) , with this risk being approximately three- to sevenfold higher than for healthy unexposed infants. Both clinical and laboratory data suggest that the regulation of respiration and arousal are impaired in cocaine-exposed infants, and that such impairments could presage the occurrence of SIDS. Newborns exposed in utero to cocaine also have abnormalities of EEG and brain stem auditory evoked responses, which generally disappear after 1 to 6 months. However, more sophisticated EEG studies, based on quantified EEG, have shown persistence of abnormalities after 12 months. Limited experimental data suggest that the risk of SIDS may not be affected by methamphetamine exposure, although this conclusion requires further investigation.


Destructive Effects on the Central Nervous System


Cocaine appears to have both destructive effects (i.e., associated with the histopathological hallmarks of a destructive process, such as dissolution of tissue with a reactive cellular response) and development effects (i.e., teratogenic effects). It may be difficult to distinguish developmental from destructive effects in the fetus, because a destructive process that occurs in a rapidly developing tissue may not only have direct effects on already developed structure, but may also perturb the course of future developmental events. In addition, the reactive cellular response to tissue destruction may be less vigorous early in development, and thus evidence for a primarily destructive event may be absent or barely detectable morphologically. Indeed, distinction of the destructive effects from the teratogenic effects is difficult, in part because they may coexist in the same infant.


Cerebral Infarction and Intracranial Hemorrhage.


Cocaine and likely amphetamines are distinctive among the drugs that produce teratogenic effects on the CNS in their capacity to lead also to destructive neural effects . The first clearly documented example of a destructive lesion in the brain was the demonstration of infarction in the distribution of the middle cerebral artery in a newborn infant whose mother used cocaine by nasal insufflation in large doses during the 3 days before delivery. The presence of hemiparesis at birth and the computed tomography (CT) appearance of the lesion indicated that the infarct occurred shortly before birth, presumably during the period of cocaine exposure. Subsequently, numerous infants exposed to cocaine or methamphetamine in utero, with cerebral infarction in the distribution of major cerebral vessels, usually the middle cerebral artery, have been identified. a


a References .

In one series, 6% of cocaine-exposed infants exhibited cerebral infarction. Although all such lesions appear to have been prenatal in origin, the timing of the infarctions based on radiographical and clinical criteria has varied from hours to months before delivery. Thus, in the four affected newborns in the series of Dominguez and colleagues, three had well-established porencephaly, compatible with an event occurring weeks or months previously, and one had findings (edema) compatible with an acute event. A similar prenatal origin was apparent in an infant with hydranencephaly, related to destruction of the cerebral hemispheres in the distribution of the middle and anterior cerebral arteries, noted at birth after a pregnancy marked by cocaine exposure ( Fig. 38.11 ). However, it must be recognized that the reported cases were selected and were not identified in a prospective manner. In a prospective study of 717 cocaine-exposed infants, cranial ultrasonography, although not the optimal imaging modality, did not identify cerebral infarction. Smaller areas of apparent infarction have been described in basal ganglia, periventricular white matter, and brain stem by some investigators. However, it is unclear that such lesions are more common in cocaine-exposed infants versus appropriately chosen control infants.


Figure 38.11


Hydranencephaly in an infant exposed to cocaine in utero.

This computed tomography scan shows evidence of severe destruction of cerebral hemispheres in the distribution of the middle and anterior cerebral arteries. The appearance is consistent with severe ischemic insult in utero.

(Courtesy Dr. Elke Roland.)


Intracranial hemorrhage has been described in approximately 10% to 20% of primarily full-term cocaine or methamphetamine-exposed infants in one series, and in approximately 30% to 70% of very low-birth-weight, cocaine-exposed infants in three other series. Lesions appearing in more mature infants are not of major clinical importance. As previously noted, intracranial hemorrhage was not identified disproportionately in a prospective, controlled observational study of very-low-birth-weight infants. In summary, the available data derived from studies of cocaine or methamphetamine-exposed newborns by brain imaging lead to the tentative conclusion that both ischemic and hemorrhagic lesions may occur. However, available data indicate that the infarctions related directly to cocaine are likely exceedingly rare, and the hemorrhagic lesions are likely related largely to the degree of prematurity.


Teratogenic Effects on the Central Nervous System


The major abnormalities of CNS development reported after intrauterine cocaine exposure and the developmental event apparently affected (see Chapter 2 , Chapter 5 , Chapter 6 ) are summarized in Table 38.14 ).



TABLE 38.14

Major Developmental Disturbances Reported After Intrauterine Cocaine Exposure and Developmental Event Presumably Affected






















NEUROANATOMICAL FINDING DEVELOPMENTAL EVENT PRESUMABLY AFFECTED
Micrencephaly; less gray matter, predominantly in dorsal prefrontal and frontal brain regions with accompanying increases in CSF Neuronal proliferation
Agenesis of corpus callosum; agenesis of septum pellucidum; septo-optic dysplasia Prosencephalic development
Schizencephaly, lissencephaly, pachygyria, neuronal heterotopias Neuronal migration
Abnormal cortical neuronal differentiation Neuronal differentiation
Myelomeningocele, encephalocele Neural tube formation

Abnormalities are listed in order of decreasing frequency.

CSF, Cerebrospinal fluid.


Microcephaly.


An impairment of intrauterine brain growth , manifested as diminished head circumference at birth, is the most common brain abnormality in infants of cocaine-abusing mothers. a


a References .

Indeed, in one large study, 16% of newborns exposed to cocaine in utero (compared with 6% of control newborns) had microcephaly. The relationship between cocaine exposure and microcephaly remain after associated factors are taken into account, including maternal undernutrition, intrauterine infection, smoking, and the use of other illicit drugs during pregnancy. Moreover, in one study based on analysis of maternal hair, a clear dose-dependent relationship was shown between the level of cocaine exposure and risk for head size less than 10%. Consistent findings exist for methamphetamine-exposed infants.


Disturbances of Midline Prosencephalic Development and Neuronal Migration .


An earlier study of a selected group of seven infants with abnormal neurological or ocular findings suggested that disorders of midline prosencephalic development or neuronal migration may be associated with intrauterine cocaine exposure. Three of the seven infants had varying combinations of agenesis of the corpus callosum, absence of septum pellucidum, septo-optic dysplasia, schizencephaly, and neuronal heterotopias, and two of the three also had optic nerve hypoplasia and blindness. The remaining four infants had evidence of destructive cerebral lesions. Subsequent reports have confirmed these findings and have documented the occurrence of schizencephaly, lissencephaly, pachygyria, and neuronal heterotopias as manifestations of disorders of neuronal migration. Funduscopic examination of cocaine-exposed infants has revealed optic nerve dysgenesis, coloboma, hypoplasia, and atrophy. The possibility that both the disorders of midline prosencephalic development and neuronal migration and the disorders of optic nerve development are more common than is currently known is real because cranial ultrasonography and CT may underestimate these abnormalities, and careful funduscopic examinations are not often done in newborn infants. Nevertheless, it is noteworthy that in a recent careful study of 717 cocaine-exposed infants, cranial ultrasonography failed to detect any abnormalities of prosencephalic development or overt migrational disturbance. To date, no reports have identified these findings in amphetamine-exposed newborns. In contrast, preliminary studies of methamphetamine exposure found higher rates of facial dysmorphism, skeletal abnormalities, cardiac defects, and respiratory problems in infants. However, these abnormalities were not more common in the IDEAL study of 204 methamphetamine-exposed newborns, which included a matched comparison group. Thus further data are required to fully understand the impact of these agents on midline brain development, and how this may relate to the dose and timing of exposure, alongside other factors.


Neurodevelopmental Consequences


Neurodevelopmental outcomes of infants exposed to cocaine in utero have been addressed in numerous reports, with a number of useful reviews on this topic. Overall, findings are somewhat mixed, with some studies reporting negative impacts and others reporting small or no effects. However, the weight of evidence generally suggests the presence of a range of adverse impacts on child cognition, language, and behavior. While relatively subtle compared with the teratogenic effects seen with alcohol and AEDs, these impacts are nonetheless developmentally significant and persistent, at least into adolescence.


As discussed with AEDs and alcohol, the nature and severity of child developmental outcomes associated with prenatal cocaine exposure are influenced by a number of factors, including the amount and timing of maternal drug use, maternal and infant genetic factors, and other risk exposures during pregnancy (e.g., other drug use, maternal undernutrition) or at birth that are correlated with maternal cocaine use (e.g., preterm birth). For example, heavier use, although not always easy to interpret given the increased likelihood of polydrug use among these women, tends to be associated with poorer outcomes. The following is a brief overview of key findings across each key neurobehavioral domain ( Table 38.15 ).



TABLE 38.15

Neurodevelopmental Effects of Prenatal Cocaine Exposure in Infants, Children, and Adolescents





























DEVELOPMENTAL DOMAIN INFANCY CHILDHOOD ADOLESCENCE
Cognition Habituation
Recognition memory
Inhibitory control
Auditory working memory
Cognitive flexibility
Narrative memory
Inhibitory control
Visuospatial working memory
Problem solving
Learning
Educational achievement
Language Receptive and expressive language (incl. auditory comprehension)
Verbal reasoning
Syntax
Phonology
Educational achievement
Behavior Arousal, attention
Irritability, excitability
Attention and ADHD
Conduct disorder
Aggressive behavior
Internalizing problems, incl. anxiety and depression
Early-onset sexual behavior and risk-taking
Attention
ADHD
Delinquent and risk-taking behavior
Emotionally volatile
Unempathetic
Substance use/abuse
Motor Seizures, tremors
Respiratory distress/SIDS
Fine motor
Visuospatial abilities

ADHD, Attention-deficit/hyperactivity disorder; SIDS, sudden infant death syndrome.


Cognition and Executive Functioning


Most studies using measures of global cognitive functioning and IQ (e.g., Bayley Scales, Stanford Binet Test of Intelligence) have found that cocaine-exposed children generally perform in the low average score range . However, these cognitive effects appear to reflect other adverse exposures during pregnancy and/or in the postnatal environment, rather than the direct effects of cocaine. Similarly, children prenatally exposed to methamphetamine do not appear to be subject to cognitive delay or IQ deficits after covariate adjustment.


In contrast, when specific neuropsychological abilities have been examined in more recent studies, these suggest adverse effects on a range of abilities, including executive functioning, inhibitory control, working memory, and cognitive flexibility. a


a References .

Subtle effects on measures of executive functioning and attention, including poorer inhibitory control and sustained attention, are also apparent after methamphetamine exposure. Longer term, these challenges and related behavior problems (discussed later), combined with an often less than ideal family home situation, predispose these children to impaired problem solving, learning difficulties, educational underachievement, and in turn, reduced life course opportunities.


Language Development


Global language delays are more common among children and adolescents exposed in utero to cocaine, with problems spanning both comprehension and receptive abilities . Importantly, these effects persist after covariate adjustment. Specific problems with syntax and phonology have also been reported. In contrast, no significant between-group differences were found between exposed and nonexposed 3-year-old children on measures of receptive or expressive language development after methamphetamine exposure. However, these children are at increased risk of educational difficulties in mathematics and language at a later age, although the extent to which these learning difficulties reflect direct drug effects or associated family risk exposures remains unclear.


Behavior and Regulatory Problems


Consistent with findings showing impacts on limbic and hypothalamic systems, cocaine exposure is associated with persistent emotional and behavioral regulatory problems spanning from infancy through adolescence. These difficulties manifest in a number of ways, including attention problems, aggression, and an increased risk of externalizing and internalizing behavior problems, including ADHD, conduct problems, anxiety problems, and depression. During adolescence, risk-taking and delinquent behaviors are common, such as fighting, running away from home, purposefully breaking or damaging things, early sexual behavior, and substance abuse. These latter problems are independent of other perinatal exposures and have been shown to be exacerbated and, in some cases, mediated by adverse family circumstances, including parental ongoing drug use, family violence, poor parental supervision, and caregiver mental health problems.


Although limited, studies of amphetamine exposure suggest similar long-term challenges. Prenatal methamphetamine exposure places children at increased risk of anxious/depressive problems and emotional reactivity during the preschool period. By school age, externalizing behaviors (aggressive behavior, rule-breaking) and ADHD symptoms are also more common, suggesting the likelihood of higher rates of ADHD later in life. Follow-up studies of methamphetamine-exposed adolescents are needed.


Motor and Visual Function


Findings are mixed with respect to motor outcomes of children exposed to cocaine during pregnancy, with studies largely confined to infants and young children. Some studies find no enduring effects of cocaine, whereas others report subtle impairments in fine motor and visual perceptual domains, especially for boys. As with other outcomes, these developmental difficulties frequently occur in the context of family psychosocial disadvantage, which predicts later risk more strongly than prenatal cocaine exposure.


Beyond the neonatal period, little is known about the potential adverse motor effects of prenatal amphetamine exposure. Poor fine motor and, in particular, grasping skills were noted at age 3 in a cohort exposed to methamphetamine. To date, studies in early childhood and beyond have not emerged.


Neuropathology


As suggested by the hallmark microcephaly associated with in utero cocaine or amphetamine exposure, micrencephaly is prevalent in neuroimaging studies. Examination of regional brain volumetric differences between infants exposed in utero to cocaine, and those not exposed, suggest that cocaine exposure is associated with structural alterations in cortical gray and white matter development . These alterations were largely confined to prefrontal and frontal brain regions implicated in cognitive and behavioral control. Further analysis using functional connectivity MRI methods revealed both polydrug effects on amygdala-frontal, insular-frontal, and insular-sensorimotor circuits, but left amygdala-frontal functional connectivity alterations associated specifically with cocaine exposure. These results suggest that prenatal drug exposure perturbs functional connections between the amygdala and frontal cortex in a way that may disrupt top-down regulation of amygdala functions by the prefrontal cortex, potentially accounting for some of the difficulties these children experience regulating emotional arousal. These between-group differences remained after controlling for other confounding factors. However, unfortunately, the sample was too small to allow dose-response relations to be examined.


Volumetric studies of methamphetamine-exposed children similarly reveal both thinning of the cortex and reduced tissue volumes in dopamine-rich striatal areas, including the caudate nucleus and associated frontal and parietal areas. These findings are consistent with clinical and experimental research showing the neurotoxicity of methamphetamine to striatal neurons. Alterations in white matter microstructure within these areas, as well as the corpus callosum, have also been found in prenatal methamphetamine-exposed children at 3 to 4 years.


The possibility that cocaine or amphetamine exposure disturbs neuronal differentiation in cerebral, diencephalic, and brain stem structures is suggested by a series of neuroanatomical, neurobehavioral, neuropharmacological, neurochemical, and physiological studies, primarily in rats, but also in primates and most recently in human infants. Thus prenatal cocaine exposure in rats and monkeys results in persistent defects in learning and memory. Moreover, Dow-Edwards and colleagues, using radioactive 2-deoxyglucose autoradiography, found that adult rats exposed to cocaine prenatally had impaired glucose metabolism in the hippocampus, a structure crucial for memory and learning; the nigrostriatal pathway, important in the regulation of movement; the mesolimbic dopaminergic system, important for the reinforcing effects of drugs like cocaine; and the hypothalamus, important for reproductive function, growth regulation, and osmotic balance. Thus exposure to cocaine during the earliest phases of neuronal differentiation in the experimental animal can lead to profound and permanent effects on the function of many crucial neuronal systems. These effects should be considered teratogenic, because it appears that a permanent derangement of neuronal development has resulted. The correlate, if any, in humans remains to be defined, but there is one report of disturbances of neuronal differentiation demonstrated by immunocytochemical and histologic techniques in cerebral cortex of three infants exposed to cocaine in utero. Very similar findings have been observed in the cerebral cortex of the newborn rat exposed to cocaine in utero; importantly, these alterations in development persist into adulthood in animal models ( Fig. 38.12 ).




Figure 38.12


Impaired cerebral cortical neuronal organization (in the adult rat) after intrauterine cocaine exposure.

Animals were exposed to saline (control) or cocaine from intrauterine day E8 through E18 and sacrificed at 40 days after birth. Cresyl violet staining of the molecular layer (ML), supragranular layers (SGL), and infragranular layers (IGL) of cerebral cortex is shown. The severe disorganization of the cortical layering in the cocaine-exposed animals contrasts with the regular lamination in the control animals. In particular, note the inversion of nuclear staining intensity between the SGL and IGL and the focal heterogeneity of nuclear size within the SGL (arrow) .

(From Nassogne MC, Gressens P, Evrard P, Courtoy PJ. In contrast to cocaine, prenatal exposure to methadone does not produce detectable alterations in the developing mouse brain. Dev Brain Res . 1998;110:61–67.)


Pathogenesis


Cocaine and amphetamines produce both acute and chronic disturbances in fetal development. Acute deleterious effects most likely arise from increased levels of multiple monoamines exerting effects on cardiac, vascular, and uterine tissue. Chronic disturbances of CNS development result from a constellation of hemodynamic effects as well as nutritional, molecular, and genetic disturbances ( Fig. 38.13 ).




Figure 38.13


Scheme for the potentially deleterious effects of cocaine taken by pregnant women on their fetuses.

Effects on the maternal side of the placenta are shown in the upper portion of the figure and on the fetus in the lower portion. Effects that appear plausible on the basis of current information but that require more supporting information are indicated by interrupted lines.


Fetal Blood Flow


Cocaine and amphetamines act as stimulants by blocking the reuptake of dopamine, serotonin, and norepinephrine, leading to both euphoric and profound cardiovascular effects. Tachycardia and vasoconstriction in the mother, including direct vasoconstriction of the umbilical vein, impair placental blood flow , disrupting oxygen and nutrient transfer to the fetus. These acute alterations in fetal blood flow may contribute to the destructive effects of cocaine in the neonatal CNS. Increases in fetal cerebrovascular resistance have also been documented following cocaine administration. Although global cerebral blood flow is not disturbed, the state of development of cerebral vessels in the human fetus may explain the distribution of ischemic lesions observed in human studies. The extraparenchymal, leptomeningeal arteries begin to develop a distinct muscularis in the second trimester and have a well-developed muscularis in the third trimester of gestation, the time of occurrence of the cerebral infarcts reported in the cocaine-exposed infants. Thus the middle cerebral artery, the vessel affected in most cocaine-induced strokes, presumably does not develop the capacity to undergo spasm until the third trimester. Intracranial hemorrhage could also occur in the immediate neonatal period secondary to elevation in blood pressure and cerebral blood flow velocity that have been documented on the first and second postnatal day in infants exposed to cocaine in utero. Elevated levels of catecholamines in the presence of cocaine also increase uterine contractility, likely resulting in the increased rate of spontaneous abortion, preterm birth, and placental abruption associated with antepartum cocaine use.


Fetal Nutrition


Cocaine and amphetamines have important impacts on fetal nutrition , arising from both maternal malnutrition as well as direct effects. Cocaine and amphetamines are both associated with low body mass index in adults, potentially due in part to their anorexic effects. Cocaine also inhibits the placental transport of specific amino acids, including arginine, phenylalanine, and valine. Of interest, these effects appear to be exacerbated by concurrent exposures, including nicotine. In addition, catecholamines increase metabolism of nutrients, further contributing to fetal depletion.


Molecular Effects


Cocaine administration to neonatal rats results in the inhibition of DNA synthesis and alterations of membrane lipids in all brain regions, consistent with the direct effects shown in nonneural tissues. Deleterious effects on neuronal differentiation in rodent models have also been observed. The disturbances in neuronal number and differentiation have also been observed in primate models.


The mechanism of the effects of cocaine on neuronal differentiation and on development of crucial neuronal pathways in the cerebrum, diencephalon, and brain stem (subsequent to neuronal proliferation) could relate to the fundamental mechanisms of the effects of cocaine on neurotransmitters. The neuronal pathways involved either use as neurotransmitters several monoamines (e.g., norepinephrine, dopamine, serotonin) or are the targets of neurons that use monoamines as neurotransmitters. The derangements of homeostasis of these neurotransmitters by cocaine are, of course, the principal mechanisms of action of the drug in the adult brain. The specific nature of the derangements in the fetus requires further definition but appears similar. Although increased levels of these neurotransmitters might be expected, at least initially, chronic exposure to cocaine could lead to their depletion, as has been shown in experimental models and in adult humans. The findings of reduction in striatal dopamine in neonatal rabbits exposed to cocaine in utero and diminished cerebrospinal fluid levels of homovanillic acid, the principal metabolite of dopamine, in cocaine-exposed newborns supports this possibility. These neurotransmitters appear very early in brain development and play important regulatory roles in development of neuronal circuitry. Indeed, because these monoaminergic systems, which originate especially in the brain stem, have widely distributed contacts in the basal ganglia, cerebral cortex, hypothalamus, and elsewhere, disturbances in their function during development could have very far-reaching effects. For example, neurons containing norepinephrine in the locus ceruleus (a pigmented nucleus in the pons) give rise to ascending monosynaptic pathways that are distributed widely in the cerebral cortex and diencephalon. These pathways are considered to have primarily activating influences. A prominent dopaminergic system originates in the substantia nigra of the midbrain and terminates in the striatum. These connections are important in the regulation of movement and tone. Other dopaminergic systems terminate in limbic and cortical structures and are crucial not only for the reinforcing actions of drugs like cocaine but importantly also for motivational-attentional functions. Prominent serotoninergic systems originate in the raphe nuclei of the brain stem, project widely, and are crucial for regulation of sleep and level of alertness. However, because disturbances of the development of neuronal circuitry do not leave a readily identifiable, morphological stamp, and require highly sensitive immunochemical and neuropharmacological techniques for detection, identification of such neural abnormalities in humans is very difficult. However, impairment of subsequent cerebral cortical development by prenatal cocaine exposure during the period in which monoamines influence neural development has been shown in the primate as well as rodent cerebral cortex. Careful study of cerebral cortical development in the rat has shown in the adult cortex abnormalities of neuronal lamination and dendritic morphology after prenatal exposure to cocaine (see Fig. 38.12 ).


Genetic and Epigenetic Alterations


Whether the effects on neuronal proliferation and differentiation relate to perturbation of the action of critical genes , including immediate early genes, is an intriguing possibility. Cocaine exposure downregulates the placental norepinephrine transporter (NET), responsible for sodium-chloride-dependent reuptake of extracellular norepinephrine. Reduced placental NET expression may lead to increased circulating norepinephrine, downregulation of the steroid metabolic enzyme 11β-HSD-2, and fetal hypercortisolism, resulting in altered activity of the hypothalamic-pituitary-adrenal (HPA) axis. Altered function of the HPA axis has been implicated as a potential contributor to long-term behavioral and emotional disorders observed in children exposed prenatally to cocaine. These changes are associated with hypermethylation of genomic DNA, most profoundly in promoter regions. In adults, chronic cocaine exposure permanently alters brain-derived neurotrophic factor (BDNF) and Cdk5 promoters. Alteration of these and other promoters has the potential to produce profound alterations in development throughout life. Of interest, genetic modifications may not be confined to the maternal-fetal dyad, as evidenced by behavioral alterations in offspring after paternal cocaine exposure.


Prevention


Prevention of prenatal cocaine and illicit methamphetamine exposure is essential. Although human data regarding prescription amphetamine exposure are lacking, caution is warranted, and these agents should be avoided in pregnancy, if possible. Prenatal care among women exposed to cocaine improves birth weight in offspring and may be the first step in a comprehensive rehabilitation program that would ideally extend into longer-term parental drug treatment and family/parenting support.


Treatment


Abstinence from both cocaine and amphetamines occurs rapidly in the newborn and resolves relatively quickly. As discussed previously, nearly all exposed infants exhibit symptoms—most consistently hypertonia. However, severe symptoms resolve rapidly without pharmacological intervention. An enriched postnatal environment over the first several years appears crucial in modulating any effects of cocaine on the infant’s cognitive and behavioral development. Early intervention should be used in all high-risk infants, particularly those exposed to illicit stimulants prenatally.




Opioids


Opioids have been used for centuries to manage pain and have a long history of popularity as a recreational drug in view of their additional euphoric effects. Derived from opium, a powdered exudate from the fruit capsule of the poppy plant Papaver somniferum , opioids act by binding to opioid receptors throughout the body. The analgesic and euphoric effects of these drugs result through the activation of receptors in the central and peripheral nervous systems, whereas side effects such as respiratory depression, sedation, and reduced intestinal motility arise from peripheral receptors.


Until early this century, the most commonly used opioids were opium and heroin. For pregnant women with an addiction to heroin or other illicit opioids, the most effective mode of therapy has been maintenance with oral methadone since the early 1970s. Evidence shows that methadone maintenance is effective in stabilizing maternal blood opioid concentrations, facilitating engagement in treatment and prenatal care, and reducing lifestyle risks associated with risk-taking and illegal behaviors such as prostitution and criminal offending. These methadone programs have been the source of valuable information regarding the effects of opioids on the fetus and newborn. More recently, the efficacy of maintenance of pregnant women with another synthetic opioid, buprenorphine, has become a further focus of research and clinical interest, because of its longer half-life (>24 hours) and reduced risk of respiratory depression when overdosed. Early findings suggest some short-term neonatal benefits for the infant, but longer-term outcome research is still needed.


Of great public health significance has been the dramatic surge since the early 2000s in the use and abuse of prescription opioids, including hydrocodone (Vicodin) and oxycodone (Percocet). This increase has been especially noticeable in the United States, with the recognition of pain by the American Pain Society as a fifth vital sign and the active marketing of new opioid medications. This increased medical use has dramatically affected the numbers of pregnant women and infants affected by prenatal opioid use. It has also led to increases in the availability and appeal of prescription opioids (and in turn heroin) as a recreational drug of abuse across socioeconomically and demographically diverse groups.


Prevalence


As noted earlier, while there is evidence worldwide of increasing opioid use during pregnancy, this has been most prominent in the United States. Estimates suggest that on average between 2008 and 2012, 39% of Medicaid insured and 28% of privately insured women of reproductive age (15 to 44 years) filled an outpatient prescription for an opioid each year. These rates were highest in the Southern states, especially rural areas, and among non-Hispanic white women. The rates are somewhat lower during pregnancy, but still are high. A recent analysis of the prevalence and patterns of opioid use among pregnant women enrolled in commercial insurance plans (>500,000) in the United States between 2005 and 2011 found that 14.4% were dispensed an opioid at some time during pregnancy. No variation was found in rates across the three pregnancy trimesters, and the most commonly prescribed drugs were hydrocodone (6.8%), codeine (6.1%), and oxycodone (2%). A small proportion (2.2%) of women were dispensed opioids three or more times during their pregnancy. In general, similar rates and distributions have been reported among pregnant women on Medicaid in New York (9.5%) and in Tennessee (28%). The latter study also showed that women taking prescription opioids were more likely to be white (72.4% vs. 65.8%), have depression (5.3% vs. 2.7%) and anxiety disorders (4.3% vs. 1.6%), use tobacco (41.8% vs. 25.8%), and also be taking an SSRI (4.3% vs. 1.9%). These comorbid difficulties are lower, but similar in profile to those reported among pregnant women enrolled in methadone maintenance treatment programs.


Although the US accounts for more than 80% of the world’s consumption of opioid pain relievers, this issue is also of concern for other developed countries. For example, data from a population-based survey in Norway indicate that 6% of pregnant women filled at least one opioid prescription between 2004 and 2006.


The rising prevalence of opioid use in pregnancy has led to an increase in associated neonatal outcomes such as NAS ( Fig. 38.14 ). From 2000 to 2012, the number of newborns in the United States diagnosed with the syndrome increased nearly fivefold, reaching a rate of 5.8%. This equates on average to one newborn every 30 minutes in the United States. In contrast, estimates of rates of NAS in England and Australia have remained relatively stable, at around 2.7%. Findings from large-scale prevalence studies suggest that the risks of NAS are greatest for infants born to mothers characterized by long-term opioid use (relative risk = 2.1) and use later in pregnancy (relative risk =1.2). The presence of additional risk factors, such as maternal SSRI use (OR = 2.1) and cigarette smoking, was also associated with increased risk.




Figure 38.14


US incidence of neonatal abstinence syndrome (NAS) , 2000–2012.

This trend mirrors the rising prevalence of opioid use in the general population in the United States during this time period.

(From Pryor JR, Maalouf F, Krans EE, Schumacher RE, Cooper WO, Patrick SW. The opioid epidemic and neonatal abstinence syndrome in the USA: a review of the continuum of care. Arch Dis Child Fetal Neonatal Ed . 2017;102:F183–F187.)


Clinical Features


There has been an intermittent history of interest in the effects of opioid use during pregnancy on both the mother and infant, with studies primarily of heroin in the 1970s and 1980s, followed by methadone and, more recently, prescribed opioids, in line with population trends. The methodological quality of studies is variable and often limited by retrospective review of clinical databases rather than direct assessment of the infant, lack of an appropriate reference or control group, and examination of a limited range of outcomes. Nonetheless, converging evidence suggests that infants who are born to opioid-dependent mothers are characterized by two major features: poorer intrauterine growth and an increased risk of the withdrawal syndrome NAS .


Maternal-Fetal Effects


Among infants born to heroin-dependent women, the incidence of low birth weigh t (i.e., <2500 g) was approximately 40% to 50% among early studies. Relatedly, approximately one-third of exposed newborns will be small for gestational age (SGA), and approximately 40% will have a head circumference below the 10th percentile for gestational age ( Table 38.16 ). This occurrence of intrauterine growth restriction in heroin-exposed infants is reminiscent of experimental studies showing growth retardation in the progeny of female rats given morphine before but not after conception. However, it is likely that other factors, related to nutrition and infection, were present in the mothers before and during pregnancy. Unfortunately (and as highlighted earlier), many of the early heroin studies did not examine the relative contribution of these correlated lifestyle factors.



TABLE 38.16

Mean Birth Weight and Gestational Age of Infants With Different Drug Exposure During Gestation


































MATERNAL DRUG USE NO. OF INFANTS BIRTH WEIGHT: MEAN (g) GESTATIONAL AGE: MEAN (WEEKS)
Heroin 61 2490 38.0
Ex-addict 33 2616 38.6
Heroin and methadone 59 2535 38.3
Methadone 106 2961 39.4
Control 66 3176 40.0

Adapted from Kandall SR, Albin S, Lowinson J, Berle B, Eidelman AI, Gartner LM. Differential effects of maternal heroin and methadone use on birth weight, Pediatrics 58:681–685, 1976.


Infants born to methadone-maintained mothers have been shown to be at increased risk of being born early, and even when born close to term, to weigh less, be shorter, and have smaller head circumferences than infants born to non-drug-using mothers. Between 10% and 35% are low birth weight (i.e., <2500 g). Of these, two in five infants will be born SGA. Risks of SIDS, strabismus, hyaline membrane disease, vision abnormalities (particularly nystagmus), and congenital birth defects are also higher among infants born to mothers receiving methadone maintenance treatment compared with infants born to non-drug-dependent mothers. However, findings demonstrate that outcomes associated with maternal methadone use are generally better than for untreated heroin use during pregnancy. The reasons for the disturbances in intrauterine growth are not entirely clear, and the relative roles of intrauterine undernutrition, infection, and toxic effects of other drugs and exogenous materials remain to be elucidated. Although a direct effect of narcotic analgesics on cell number has been suggested by experimental studies (discussed in a later section), findings are mixed with respect to the analysis of human infants. Some studies are more supportive of an indirect effect related to impaired maternal nutrition and other factors, whereas other studies support the persistence of growth differences, even after covariate factors are taken into account.


Only a few studies have examined the neonatal effects of maternal buprenorphine treatment during pregnancy. Of these, some have found no differences in the risks of fetal death, preterm birth, low birth weight, and SGA/growth restriction, whereas others have reported a lower risk of preterm birth and higher birth weights for buprenorphine-exposed infants compared with methadone-exposed infants. A recent meta-analysis of 18 studies (three randomized controlled trials [RCTs] and 15 cohort) found that buprenorphine was associated with lower risk of preterm birth (RR = 0.40 to 0.67), greater birth weight (weighted mean difference = 265 to 277 g), and a larger head circumference (weighted mean difference = 0.68 to 0.90), with bigger differences observed in RCT studies. No differences were found for spontaneous fetal death, fetal congenital anomalies, and other growth measures.


Neonatal Abstinence Syndrome


Early studies suggested that the incidence of NAS in heroin-exposed infants was about 70%, varying from about 50% to 90%. a


a References .

In general, similar rates are reported among infants born to mothers enrolled in methadone maintenance treatment programs, with rates varying widely from 13% to 90%. This variability likely reflects both differences in clinical approaches with respect to maternal and infant management (in-hospital weaning of the infant vs. early discharge with home visiting care vs. in-hospital maternal detoxification during pregnancy and follow-up care) and other factors related to maternal drug use (i.e., maternal nicotine use, other drug exposure, methadone dose). b

b References .

There is some support that maternal maintenance with buprenorphine during pregnancy may be associated with a lower risk of NAS and in turn a shorter length of stay than with methadone.


Concerning the perinatal outcomes associated with maternal prescription opioid use, findings are now emerging but are exclusively confined to medical record review. For example, an analysis of more than 100,000 pregnant women who filled more than one prescription (28%) in Tennessee found that opioid-exposed infants with and without NAS (the latter implying high levels of exposure) were more likely to be born with low birth weight (no NAS 21.2%, NAS 11.8%) than nonexposed infants (9.9%).


The clinical presentation of NAS following opioid exposure consists of an array of signs and symptoms, including increased irritability, hypertonia, tremors, seizures, feeding intolerance, watery stools, emesis, and respiratory distress, with both male and female infants affected equally. The predominance of signs and symptoms relevant to the CNS and gastrointestinal tract relate to the particular concentration of opioid receptors in these regions (see later). These symptoms tend to emerge between 6 and 48 hours after birth as a result of the sudden discontinuation of exposure to the opioids being used or abused by the mother during pregnancy ( Fig. 38.15 ). The time of onset of the withdrawal syndrome in the newborn delivered to a heroin-addicted woman is usually quite early ( Table 38.17 ). Approximately 65% of infants present within the first 24 hours of life; an additional approximately 20% on the second day of life; and the remainder, or about 15%, on the third and fourth days. This contrasts with the time of onset of the withdrawal symptoms with methadone. Most infants exposed to methadone have the onset of overt symptoms on the second day of life ( Table 38.18 ), not the first day as with heroin. Median onset of therapy in a recent study was 35 hours. Ten percent to 15% of infants born to methadone-treated mothers have had onset of their neurological syndrome after day 3. This late onset is rare with infants passively addicted to heroin. The delay in onset correlates with the administration of the last dose of methadone before the time of delivery ( Fig. 38.16 ). Indeed, withdrawal symptoms in infants passively addicted to methadone may occur initially or may recur as late as 2 to 4 weeks or more after birth. In fact, an occasional infant has died before the nature of this later syndrome has been recognized.




Figure 38.15


Onset and duration of neonatal abstinence syndrome symptoms from heroin, methadone, and buprenorphine.

Note the delayed onset and prolonged duration of methadone withdrawal compared to heroin withdrawal. Interestingly, despite a long half-life, buprenorphine withdrawal symptoms appear early and resolve rapidly compared to methadone.

(Adapted from Binder T, Vavrinkova B. Prospective randomised comparative study of the effect of buprenorphine, methadone and heroin on the course of pregnancy, birthweight of newborns, early postpartum adaptation and course of the neonatal abstinence syndrome (NAS) in women followed up in the outpatient department. Neuro Endocrinol Lett . 2008;29:80–86.)


TABLE 38.17

Time of Onset of Withdrawal Syndrome in Infants Passively Addicted to Heroin
























TIME AFTER BIRTH (H) NO. OF INFANTS PERCENTAGE OF TOTAL a
0–12 76 29%
12–24 88 34%
24–48 56 21%
48–96 39 15%

Adapted from Zelson C, Rubio E, Wasserman E. Neonatal narcotic addiction: 10-year observation. Pediatrics . 1971;48:178–189.

a Total is all infants with withdrawal syndrome (i.e., 259 of 384, or 67.4% of complete series of infants born to heroin addicts).



TABLE 38.18

Time of Onset of Withdrawal Syndrome in Infants Passively Addicted to Methadone




























TIME AFTER BIRTH (H) NO. OF INFANTS PERCENTAGE OF TOTAL a
0–12 0
12–24 8 27%
24–48 16 53%
48–72 3 10%
>72 3 10%

Adapted from Stimmel B, Adamsons K. Narcotic dependency in pregnancy. JAMA . 1976;235:1121–1124.

a Total is all infants with withdrawal syndrome.




Figure 38.16


Passive addiction to methadone: relation of the time of the last maternal methadone dose (LMD) to the time of onset of neonatal withdrawal.

The interval between onset of withdrawal and time of last maternal dose did not vary markedly and approximated 48 hours.

(From Rosen TS, Pippenger CE. Pharmacologic observations on the neonatal withdrawal syndrome. J Pediatr . 1976;88:1044–1048.)


The reasons for the delayed onset and prolonged duration may relate to the pharmacokinetics of methadone elimination in the newborn. It is well known in adults that the withdrawal syndrome in methadone addiction develops more slowly (peak 6 days) and lasts longer than with heroin. Good evidence is available for avid tissue binding of methadone, with gradual and slow release on withdrawal. The critical point is that the clinician must be alert to this possibility of delayed onset in the infant of age 1 week or more who develops jitteriness, diarrhea, and other signs of withdrawal.


The likelihood of withdrawal symptoms in an infant exposed to heroin during pregnancy seems to relate primarily to five factors: (1) the amount of the maternal dose; (2) the length of the time from the last dose to birth; (3) the duration of maternal drug use/abuse; (4) the use of other drugs, particularly nicotine and SSRIs; and (5) the gestational age of the infant at birth. a


a References .

Withdrawal symptoms in the passively addicted newborn are more likely if the maternal dose has been high; if the last dose has been within 24 hours of the time of birth; if the mother has been a long-term user, smokes, or takes SSRIs; and if the infant is born at term.


Similarly, the gestational age of the infant at birth influences the likelihood and severity of neonatal withdrawal after methadone ( Fig. 38.17 ). In contrast to heroin, maternal methadone dose does not appear to be a critical determinant of the likelihood of withdrawal, although cord blood levels are inversely related to the occurrence of a neonatal withdrawal syndrome requiring therapy. In fact, the likelihood of neonatal withdrawal features directly correlates with more rapid rates of decline of neonatal methadone levels. Moreover, the severity of symptoms also relates particularly to the rate of elimination ( Fig. 38.18 ). This finding is entirely consistent with an observation in adult patients (i.e., a rapid decrease in the drug level in blood causes more frequent and more severe symptoms).




Figure 38.17


Relationship between the severity of the neonatal withdrawal syndrome after passive addiction to methadone and the gestational age of the infant.

Scores are expressed as median values, with ranges in parentheses. Note the increase in severity with advancing gestational age.

(From Doberczak TM, Kandall SR, Wilets I. Neonatal opiate abstinence syndrome in term and preterm infants. J Pediatr . 1991;118:933–937.)



Figure 38.18


Passive addiction to methadone: relation of occurrence and severity of signs of withdrawal to rate of elimination of methadone.

Data derived from study of 31 infants born to mothers receiving methadone during pregnancy. The fastest declines in plasma methadone (M) concentrations correlated with occurrence of the most severe signs.

(From Rosen TS, Pippenger CE. Pharmacologic observations on the neonatal withdrawal syndrome. J Pediatr . 1976;88:1044–1048.)


The initial and dominant symptoms and signs of the withdrawal syndrome relate primarily to disturbance of the CNS ( Table 38.19 ). A virtually invariable feature is jitteriness. As discussed in Chapter 12 , the movements of jitteriness are characterized primarily by tremulousness; are exquisitely stimulus-sensitive, rhythmic, and usually of equal rate and amplitude; and can be induced to cease by gentle passive flexion of the limb. Unlike seizure, jitteriness is accompanied neither by abnormalities of gaze or extraocular movement, nor by the clonic jerking of limbs. The tremulous movements in infants passively addicted to heroin are usually quite dramatic and have a coarse, flapping quality. The babies are very irritable and frequently are extremely active and hypertonic. Sleep periods are markedly diminished, and the cry is often high pitched and shrill. Sucking is excessive, and often frantic sucking of the fists or fingers has been noted. These frequent clinical features (75% to 100% of symptomatic cases) are very helpful in making the clinical distinction of the jitteriness of drug withdrawal from that resulting from other important causes (e.g., hypoglycemia and hypoxia). Hypoglycemia can cause jitteriness in the first 24 to 48 hours of life, but with hypoglycemia the infant is almost always stuporous as well, quite unlike the hyperalert, hyperactive infant with withdrawal symptoms. Similarly, hypoxic-ischemic encephalopathy characteristically causes jitteriness in the first 24 to 48 hours of life, but such infants almost always have had the characteristic history of a perinatal hypoxic-ischemic insult, are significantly stuporous, and very frequently exhibit seizures. The clinical picture may be complicated by concomitant passive addiction and hypoxic encephalopathy; in several series, 20% to 40% of addicted infants exhibited signs of fetal distress, Apgar scores of six or less at 1 or 5 minutes, or both.



TABLE 38.19

Signs of Withdrawal From Heroin









































RELATIVE FREQUENCY (PERCENTAGE OF TOTAL)
75%–100% 25%–75% <25% RARE
Jitteriness Poor feeding Fever Seizures
Irritability Vomiting
Hyperactivity-hypertonicity Diarrhea
Decreased sleeping Sneezing
Shrill cry Tachypnea
Excessive sucking Sweating


Next in frequency of occurrence are a series of symptoms relating especially to gastrointestinal disturbance (see Table 38.19 ). Poor feeding is the most prominent of these features (one that in fact may reflect more of a neurological than a gastrointestinal disturbance). Despite the initially excessive sucking described previously, the infant decreases its sucking rapidly with feeding. Poor coordination of sucking, swallowing, and respiration is very common. Regurgitation of feedings is also a relatively common feature. Diarrhea occurs in as many as 30% to 50% of the infants and can contribute to dehydration and electrolyte disturbances. These latter gastrointestinal phenomena tend to appear later (i.e., at days 4 to 6) than those related to the CNS.


Sneezing and tachypnea are less common disturbances (see Table 38.19 ). Still less common, but disturbing when they do occur, are fever and sweating . Fever must always raise the possibility of infection, and this possibility should be ruled out by appropriate diagnostic studies. Sweating, while uncommon, can be a helpful diagnostic sign because it is very unusual to see sweating in newborns, especially small newborns.


Seizures are a distinctly uncommon manifestation of neonatal withdrawal to opioids (see Table 38.19 ). The incidence has been approximately 1% to 2% of cases. It is in fact difficult to be convinced from the published data that any of the examples of seizure associated with withdrawal to heroin were not related to complicating factors, such as hypoxia-ischemia or metabolic disease, or were not examples of particularly marked jitteriness. This conclusion is compatible with the fact that seizures are not a feature of withdrawal to heroin in adult patients. Thus it should be emphasized that passive addiction to opioids should be low on the list of considerations when one is faced with a newborn with seizures, and even in the infant definitely passively addicted to heroin, seizure phenomena should raise the possibility of a serious complicating illness and provoke appropriate diagnostic studies.


Neurodevelopmental Consequences


Research examining the longer-term outcomes of children exposed to opioids during pregnancy is even more sparse than the short-term infant outcome studies reviewed earlier. In addition, much of what is known is confined to heroin/methadone exposure, and with a few exceptions, based on older cohorts of children where maternal methadone doses were typically lower than in current practice. Follow-up rates are also often poor and rarely extend into middle childhood when many cognitive functions are beginning to become apparent. As of yet no studies have examined the effects of maternal prescription opioid use on child development beyond the infancy period. Finally, it is important to note that, as with other drug exposure studies, most pregnant women who use opioids or are in methadone/buprenorphine treatment programs continue to use other drugs. They are also more likely to come from high-risk socioeconomic backgrounds and to raise their children in family circumstances characterized by ongoing drug use and high rates of interpartner violence, child maltreatment and parental dysfunction, poverty, and social risk, making it difficult without large samples to delineate the relative contributions of these factors from the direct effects of the target opioid drug.


Cognition and Executive Function


Studies examining the cognitive outcomes of children exposed to opioids during pregnancy report conflicting results. While some studies report no difference, others have found that opioid-exposed children perform less well on standardized cognitive measures. a


a References .

In general, cognitive function tends to be within the normal range, although sometimes somewhat lower in opioid-exposed infants than in control infants. Language is also often delayed. At least based on existing methodologically limited evidence, these variations appear to largely reflect the effects of family socioeconomic status and other factors correlated with prenatal opioid exposure. Beyond general cognition, little is known about the extent to which these children are also at risk of executive function and learning problems. The one exception is a study of 35 methadone/buprenorphine-exposed and 31 nonexposed comparison children followed to age 48 to 57 months, which found that exposed children performed less well on behavioral measures of inhibitory control and short-term memory.


Behavior and Regulatory Problems


There is fairly consistent evidence, at least based on parent and teacher report, to show that children exposed to opioids during pregnancy have higher levels of attention, hyperactivity, and impulsivity problems than non-opioid-exposed children. There is also some suggestion that these problems may become more problematic with age. However, the proportion of opioid-exposed children who meet either DSM or ICD criteria for a range of potential psychiatric disorders, including anxiety and conduct disorder, is not known.


Motor and Visual Function


There is increasing evidence that infants born to opioid-using mothers may be at increased risk for a range of visual abnormalities, including nystagmus, strabismus, reduced visual acuity, delayed visual maturation (DVM), impaired voluntary eye movements, and absent binocular vision. A recent study of 81 methadone-exposed and 26 nonexposed comparison infants at age 6 months (retention 79% and 52%, respectively) found that opioid-exposed infants had a fivefold increased risk of failing standardized ophthalmologic assessment, with 40% (32/81) failing and a further 11% (9/81) deemed borderline. Visual abnormalities included horizontal nystagmus ( n = 9/32), strabismus ( n = 20/32), and reduced visual acuity (>0.90 logMAR) ( n = 18/32). The latter was associated with other visual abnormalities in 11 of these infants. Further electrophysiological assessment of visual function using visual evoked potentials (VEP) found that 70% of opioid-exposed infants exhibited abnormal VEP parameters (slower peak times, smaller amplitudes), suggesting that prenatal drug exposure may have altered the functioning of visual pathways and/or cerebral sources of VEPs (see Chapter 10 ). These adverse clinical and electrophysiological outcomes were independent of other prenatal drug exposures.


Neuropathology


No detailed human neuropathological studies in the setting of fetal opioid exposure are available. However, experimental studies have identified a number of structural alterations that could potentially underlie the effects of maternal opioid use on infant and child outcomes. Although morphologic studies are not entirely uniform, the majority demonstrate decreases in cerebral and cerebellar weight. Zagon and McLaughlin also found reduced cerebellar area and decreased number and density of internal granular neurons in the cerebellum with perinatal methadone exposure.


Human imaging studies are confined to a small series of studies based on small samples of opioid-exposed infants ( n = 7 to 16), thus limiting interpretation and generalizability of results. Unfortunately, polydrug use was also common and, given sample size constraints, not able to be taken into account statistically. With these caveats in mind, several volumetric MRI studies suggest that prenatal opioid exposure may be associated with lower overall brain volume shortly after birth, with regions such as the basal ganglia more affected than others. Cortical thinning of cingulate and orbitofrontal cortices was also observed. Interestingly, these differences were not found in a cohort of infants born to opioid-dependent mothers who underwent detoxification during pregnancy. Finally, two studies suggest that maturation of white matter tracts may also be affected, with higher mean diffusivity (MD) values found for the superior longitudinal fasciculus. Extending this research with more controlled studies of larger samples will be important.


Pathogenesis


Antenatal opioid exposure has both acute and long-term consequences for the newborn. Acute effects occur because of a series of physiological disturbances after withdrawal of chronic opioid receptor stimulation. A cascade of molecular effects appears to be the most important source of long-term alterations in cognition and behavior.


Pathogenesis of Neonatal Abstinence


Opioids act through G protein–coupled receptors µ, κ, and δ; µ-opioid receptors appear to predominate in the developing brain. Neurons in the locus coeruleus of the pons have a high density of noradrenergic µ-opioid receptors, and this nucleus appears to be the principal site that triggers opioid withdrawal. µ-Opioid receptor stimulation suppresses production of cyclic adenosine monophosphate (cAMP), decreasing the release of norepinephrine from neurons in the locus coeruleus. The persistent presence of an opioid stimulus eventually leads to the compensatory release of norepinephrine, possibly through upregulation and/or supersensitization of adenylyl cyclase isoforms (described as development of alternative pathways and disuse hypersensitivity in the previous edition of this text). Upregulation/supersensitization appears to rely on phosphorylation of the opioid receptor after persistent stimulation, followed by uncoupling from the G protein and receptor colocalization with β-arrestins, protein kinases (including activated Src), and adenylyl cyclases. In the absence of depressive opioids, hyperactive neurons in the locus coeruleus release excessive amounts of norepinephrine, resulting in hyperthermia, hypertension, tremors, and tachycardia.


Other sites of altered enzymatic activity include the ventral tegmental area of the midbrain, the medial habenula of the thalamus, the hypothalamus, and the dorsal raphe nucleus of the brain stem. The ventral tegmental area releases less dopamine during opioid withdrawal, resulting in hyperirritability and anxiety. The midbrain and medial habenula release increased amounts of acetylcholine, contributing to diarrhea, vomiting, and diaphoresis. The hypothalamus releases increased amounts of corticotrophin, leading to increased stress and hyperphagia. The dorsal raphe nucleus releases decreased amounts of serotonin during opioid withdrawal, leading to sleep disturbances. Decreased levels of brain-derived neurotrophic factor and corticotrophin releasing-factor appear to underlie the latter; however, the mechanisms of all of these alterations remain incompletely elucidated.


Pathogenesis of Potential Long-Term Consequences


The molecular effects of opioid exposure include apoptosis as well as altered patterns of myelination. In vitro studies demonstrate the apoptotic effect of even a single dose of opioid on neural progenitor cells, increasing levels of caspase-3 in these proliferating cells. Opioids disrupt myelination through multiple mechanisms. Opioids accelerate maturation of preoligodendrocytes in the developing brain, a phenomenon that appears to disrupt the synchronized sequence of events contributing to normal connectivity. In addition, opioids suppress neuronal migration to the cortical plate, and alter dendritic growth and branching patterns. The underlying mechanisms of these effects are not completely understood, although they likely reflect, at least to some degree, suppression of activity-dependent network formation as well as phosphorylation and uncoupling of the opioid receptor after persistent stimulation. Receptor colocalization and binding with β-arrestin activates signal transduction, recruiting kinases. Formation of a receptor/β-arrestin/extracellular signal-regulated kinase (Erk) aggregate inhibits the growth-promoting effects of Erk. Alternatively, receptor/β-arrestin may bind to c-Jun N-terminal kinase and apoptosis signal-regulating kinase, increasing the activity of these mediators of cell death. In addition, chronic opioid exposure results in lower levels of BDNF in the hippocampus. Decreased levels of BDNF precipitate decreased activation of tropomyosin receptor kinase (Trk) receptors, expressed widely in this brain region. Trk receptors are responsible for the release of Akt serine/threonine kinase, an essential inhibitor of the apoptotic caspase cascade. Cumulatively these, and likely other perturbations, result in the phenotypic neurodevelopmental manifestations of fetal opioid exposure.


Prevention


As discussed earlier, complete abstinence represents the safest choice for optimal fetal development. In mothers with an opioid addiction, maintenance treatment offers an opportunity to minimize and stabilize the effects of opioid exposure on the fetus. Methadone is the historic mainstay of maintenance therapy. Benefits of methadone over continued use of illicit drugs include improved adherence to obstetric care, decreased risk of HIV infection, and increased fetal growth. Buprenorphine , a partial µ-opioid agonist, has emerged as a potential alternative maintenance therapy. Buprenorphine possesses greater affinity for the µ-opioid receptor, but causes less activation. Buprenorphine also has very low transplacental transfer. Randomized controlled trials (RCTs) have demonstrated shorter hospitalization and decreased dose and duration of morphine therapy for NAS with buprenorphine in comparison to methadone. Disadvantages of buprenorphine include the difficulty of initiating this therapy in pregnancy, with a higher early dropout rate observed in a recent randomized controlled trial. In addition, there is a lack of long-term data describing childhood outcomes after fetal buprenorphine exposure. Although such long-term data are lacking, preliminary data are hopeful. Experimental data suggest a lower impact on myelination in the developing brain. In clinical studies, neonatal head circumference is larger, and neonatal neurobehavior is improved with buprenorphine exposure compared with methadone. Four-month-old infants have a significantly shorter latency of peak response to pattern-reversal VEPs. These promising early findings suggest potential, but must be confirmed by long-term follow-up studies.


Pharmacological treatment of mothers aimed at modifying withdrawal in newborns has been explored recently. Ondansetron represents such a promising therapy. The Htr3a gene, which encodes the 5-HT3 receptor, has been identified as a modulator of the response to precipitated withdrawal in mice. In susceptible mice, administration of ondansetron, a 5-HT3 antagonist, reduces the physical symptoms of withdrawal. Ondansetron rapidly crosses the placenta in humans, raising the possibility of antenatal therapy to blunt NAS. This therapy requires further evaluation, including examination of the impact of antenatal intervention on developmental outcome. It is also unclear how long the modification of withdrawal symptoms will last after birth.


Treatment


The three important components in the clinical management of NAS secondary to opioids are recognition, supportive therapy, and drug therapy ( Table 38.20 ).


May 16, 2019 | Posted by in NEUROLOGY | Comments Off on Passive Addiction and Teratogenic Effects

Full access? Get Clinical Tree

Get Clinical Tree app for offline access