Drug Effects on the Fetus and Breast-Fed Infant



Drug Effects on the Fetus and Breast-Fed Infant

GERALD G. BRIGGS, BPharm

Women's Hospital, Long Beach Memorial Medical Center, Long Beach, California

CLINICAL OBSTETRICS AND GYNECOLOGY 2002;45:6-21

A major dilemma facing anyone who prescribes, dispenses, or administers drugs to a woman of childbearing age is that the patient may be or may become pregnant. This dilemma is heightened by the realization that drugs can and do adversely effect the embryo, fetus, and newborn. Fortunately, most medicinal agents produce no measurable effect on the developing offspring and, thus, are relatively safe during pregnancy. Drug-induced teratogenicity and toxicity are limited to a few therapeutic agents. Awareness of these agents can be an effective means of diminishing the anxiety of prescribing for a woman who is or may become pregnant. Communication of this information to the patient can also lessen the anxiety she may experience when she is taking the therapy.

A second important consideration is knowledge of the background incidence of birth defects, their various causes, and the percentage of the total that are believed to be drug-induced. The incidence of congenital malformations among live births is approximately 5–6% after long-term follow-up.

1–3 Approximately half of these are diagnosed shortly after birth. The causes of birth defects and the approximate percentage of each are single major mutant genes (15–20%), chromosomal aberrations (5%), multifactorial (polygene–environmental factor interactions) (20%), environmental factors (maternal conditions and diseases, deformations, radiation, hyperthermia, chemicals, drugs) (10%), and unknown (up to 65%).4–6 The combined incidence of birth defects resulting from radiation, hyperthermia, chemicals, and drugs is thought to be less than 1%6 but has been estimated in the past to be as high as 3%.4

Teratogen-induced congenital anomalies usually involve a combination of minor and major malformations.7 Underreporting of adverse outcomes is most likely common, therefore, because most drug-induced malformations are characterized by a pattern of minor anomalies rather than a single major defect.7 Part of the reason for this discrepancy is that the diagnosis of minor malformations is very difficult unless exposed offspring are studied in a systematic fashion with standardized physical examinations. Moreover, the long-term effects of drugs on neurobehavior are rarely studied. Henceforth, the terms “birth defects,” “anomalies,” or “congenital malformations” will be used to refer only to major structural anomalies unless otherwise stated.

The definitions used for major and minor malformations are as follows: major— “a defect that has either cosmetic or functional significance to the child”; minor— “a defect that occurs infrequently (in less than 4% of the population) but that has neither cosmetic nor functional significance to the child.”7 Deformations are defined as “abnormal form, shape or position of a part of the body caused by mechanical forces.”8

A second dilemma facing health professionals is the woman who requires pharmacologic therapy while breast feeding. In most cases, there is less concern about the possible toxicity from drugs in breast milk because the adverse consequences are usually transient, quickly observable, and far less severe than an analogous situation during gestation. However, there are some drugs (eg, antineoplastics, radiopharmaceuticals, drugs of abuse) that may injure the nursing infant without causing easily recognizable signs and symptoms. Thus, agents with this potential should be absolutely avoided during breast feeding.

This paper is designed to serve two objectives: to provide sufficient information to clinicians and other health professionals concerning the risks of drug-induced teratogenicity and toxicity in the embryo, fetus, or newborn; and to provide adequate information to health professionals concerning the adverse effects in the nursing infant from drugs excreted into breast milk. In both of these situations, the information should serve as a foundation to counsel pregnant and lactating women on the risks of toxicity to their offspring from therapeutic drugs. To have maximum effectiveness, however, a system of close communication between the patient and her health-care providers must be established and maintained.

Counseling Women About Drugs in Pregnancy

The counseling of women who are of childbearing age should be based on accurate information and is one of the primary objectives of this paper. This knowledge base should include agents known to cause malformations or toxicity, the critical periods for the adverse outcomes, an estimate of the magnitude of risk, and the types of malformations or toxicity (

Table 1). Typically, the critical period is the period of organogenesis (5–10 weeks after the last menstrual period, or 20–55 days after conception), but it can be at any time during gestation. The risk estimates apply only to populations, not to individual pregnancies, because many of the drug-induced adverse outcomes are genetically determined.

Table 1. Drugs That Cause Toxicity in the Fetus and/or Newborn

As previously stated, most medications pose little if any measurable risk to the embryo or fetus when consumed at therapeutic doses at any time in gestation. Moreover, drug-induced teratogenicity and toxicity are relatively rare and can usually be avoided altogether by avoiding the known critical periods for toxic drugs. However, many of the newer drugs have little or no reported human pregnancy experience, so the estimation of pregnancy risk is difficult at best. Three methods to reduce the risks arising from treating pregnant women with drugs are readily apparent: try to use only those agents considered relatively safe in pregnancy; if a drug known to be toxic must be used, avoid, if possible, the critical period associated with the its toxicity; and use new drugs with no human pregnancy experience only when necessary and only after careful review of the potential risks and benefits.

For each of the above methods, the risks and benefits should be carefully and thoroughly discussed with the patient so that she is fully informed. It is an important reality that many women will markedly overestimate the risk of congenital malformations from a specific drug.9 Therefore, health professionals need to invest adequate time and resources to overcome these misconceptions and misinformation.

NONPREGNANT WOMAN WHO REQUIRES THERAPY AND MAY BECOME PREGNANT

For the nonpregnant patient, the use of the safest drug, in terms of pregnancy risk, is the best course of action, assuming that the safest drug also has acceptable efficacy for the condition being treated. However, if therapy with a potentially harmful drug is required, the patient should be informed of the risks if she does conceive and plans made to modify her therapy if necessary. Establishment of a system of close communication with the patient, if her pregnancy status changes, would be ideal. Moreover, there are some toxic agents (eg, acitretin, etretinate) that have such prolonged elimination times (months or years) that they should not be prescribed to any woman who could conceive in the foreseeable future (within 3 years). Special care should be taken to exclude exposure to these agents in women who may become pregnant. For especially potent human teratogens (eg, isotretinoin, thalidomide), continued documentation that a sexually active woman is using effective contraceptive methods and that she is not pregnant is vital to prevent inadvertent exposures. One manufacturer of a potentially potent teratogenic agent with a prolonged elimination time (leflunomide) has even designed a drug elimination procedure so that systemic levels of the drug can be rapidly reduced in the event of an inadvertent pregnancy. The importance of the above steps is evident in view of the estimate that at least half of the pregnancies in North America are unplanned.3 Consequently, patients should be counseled to contact their health-care provider immediately if they miss a menstrual period or have other reason to believe that they may be pregnant. In many cases there is time to discontinue potentially toxic therapy because physiologic exchange (ie, substance transfer) is not established between the mother and the embryo until about the fourth week after conception.

PREGNANT WOMAN WHO REQUIRES DRUG THERAPY

Women who are pregnant and who need pharmacologic therapy require a more urgent approach. Because of the apparently common misconception of the risks of drug therapy during pregnancy, extra effort may be required to ensure the patient's compliance with the prescribed therapy. This is imperative in maternal conditions in which the disease itself is a greater risk factor for adverse pregnancy outcome than the drug therapy may be (eg, epilepsy, asthma, diabetes, severe hypertension, most infections). Counseling should include the risks (maternal and embryo/fetal) of the disease in the absence of therapy or in the presence of suboptimal treatment. Consideration should also be given to delaying the treatment, if possible, so that it avoids the critical period for toxicity or birth defects. Obviously, choosing a therapeutic agent that provides the lowest known risk is important. Frequently, however, the best therapy is also one that can produce defects or toxicity. In these cases, the benefits to the patient and her pregnancy in relation to the risks of therapy should be clearly defined and communicated to the patient. This involves discussing with her the reasons for choosing a particular agent, the critical period in pregnancy for adverse drug effects, the magnitude of the risk (actual percentages are important if known), and the type of malformations or toxicity. The discussion should also reinforce the risks of suboptimal therapy. If there is no human pregnancy experience, as is the case with most new drugs, then the fetal risk must be inferred from animal toxicity data and the experience, if any, with similar agents. Again, adequate time must be given so that she understands that the treatment plan designed for her is optimal. Consent of the patient is an important element of the treatment process and is a major determinant in ensuring her eventual compliance.

PREGNANT WOMAN ALREADY RECEIVING DRUG THERAPY

Women who present for pregnancy care or who have a pregnancy diagnosed and have been receiving drug therapy are common clinical situations. In some cases, the drug exposure may have occurred during a portion or all of the critical period. If it has not, then changing to a safer drug is indicated. A careful assessment of the fetal condition should be conducted (eg, ultrasound) followed by a discussion, in a calm and supportive manner, of the potential risks. Knowledge of the drugs that are potentially teratogenic or toxic, their critical periods of exposure, the magnitude of the risk, and the types of defects is crucial in this situation, as is knowledge of the background risk. Many women elect to terminate a wanted pregnancy out of fear of an adverse outcome, even if a nonteratogen was involved. Therefore, she must receive and understand an accurate assessment of the risk (with actual percentages if available) that has been imposed on her pregnancy by the specific drug exposure. Such information has been shown to significantly reduce the tendency for elective termination.9

Counseling Women About Drugs and Breast Feeding

As a general rule, any drug that can be given safely to a neonate is probably safe to use during breast feeding. The American Academy of Pediatrics periodically publishes a review of drugs and chemicals in human milk to help health-care professionals assess which agents can be safely used during lactation. The latest review was published in 2001.

10 As can been seen in Table 2A, Table 2B and Table 2C, only a small number of drugs are contraindicated during breast feeding or should be used cautiously because of potentially serious toxicity. Because almost all drugs are excreted into breast milk, those agents that are contraindicated are considered to pose an unacceptable risk to a nursing infant. Some drugs have caused rare toxicity and may require the measurement of blood concentrations in the infant. Most drugs, however, are considered compatible with breast feeding. Because of the importance of breast feeding in the long-term health of the infant, special care should be taken during the counseling process to allay the fears of the mother concerning exposing her infant to drugs via her breast milk.

Table 2A. Adverse Effects in the Nursing Infant From Drugs in Milk

Table 2B. Continued

Criteria for Establishing Teratogenicity

Various criteria have been proposed to determine whether an agent is a human teratogen. These criteria can be summarized as follows: (a) exposure to the drug at the critical time or times; (b) consistent findings by epidemiologic studies; (c) case reports, especially of a specific defect or syndrome; (d) a rare exposure associated with a rare defect; (e) evidence that the frequency of the specific outcomes is associated with the introduction or withdrawal of the agent; (f) teratogenicity in animals at doses equivalent to those in humans; and (g) biologic plausibility.

5,11 A drug does not have to meet all of the criteria to establish it as a human teratogen. One author, however, thought that at least three of the criteria (a, b, and c or a, c, and d) were essential for proof of human teratogenicity.5

Interpretation of Animal Reproduction Studies

New drugs released since 1966 have been required to undergo reproductive testing in animals.

6 Many of these drugs exhibit dose-related teratogenicity, variations, and toxicity in commonly used experimental animal species (mice, rats, and rabbits). Often, however, the doses that cause malformations are much higher, based on a body surface area or systemic exposure, than the human therapeutic dose. The relationship of these very high doses to human malformations is tenuous at best, and even less so when the teratogenic dose is also toxic to the mother. Additionally, some species have a greater genetic tendency for certain types of defects (eg, cleft lip and resorptions when some strains of mice are exposed to a drug) than other species, so these outcomes cannot be readily extrapolated to humans. Further, some agents are prodrugs that must undergo metabolism to the active component (eg, leflunomide, oxcarbazepine, sulindac). If metabolism of a prodrug is markedly different in an experimental species, then animal reproduction studies should be conducted with the active metabolite if they are to have any meaning in human reproduction.

For the newer agents, there are frequently few or no human pregnancy data, particularly regarding exposures during the period of organogenesis. Few of these drugs, however, at least at therapeutic doses, will prove to cause human birth defects. Importantly, all established human teratogens, except angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers, are also teratogenic in one or more animal species. Excluding these two drug classes, only lithium and tetracycline fail to demonstrate concordance between human and animal drug-induced malformations (ie, human birth defects mimic those observed in animals).6 Thus, knowledge of animal reproduction tests, combined with other criteria, can help determine the risk of human teratogenicity.

Placental Transfer of Drugs

Human placentas are hemochorial in that the fetal tissue is in direct contact with the maternal blood.

12 Examples of other mammals with hemochorial placentas are the mouse, rat, hamster, guinea pig, rabbit, and Rhesus monkey.6,12 A thin layer of trophoblast cells separates the maternal and fetal blood vessels and serves as the exchange surface between the two circulation systems. Almost all medications used for the treatment or prevention of human disease cross the placenta. The most important factor that determines the fetal drug concentration is the maternal drug concentration. Drug and placenta factors that determine the rate and degree of transfer, in addition to the maternal concentration, include the molecular weight, lipid solubility, protein binding, degree of ionization at physiologic pH, placental blood flow, and placental surface area (correlated with gestational age). Placental metabolism may also be important for some agents (eg, corticosteroids). Because transfer across the membrane is by passive diffusion and thus is concentration-dependent, drugs may also cross from the fetus to the mother. Drugs with molecular weights less than 1,000 cross readily, particularly those less than 500. Near term, when the placental surface area is at its maximum, the magnitude of drug transfer to the fetus is such that for most agents the maternal and fetal concentrations reach near unity.12,13

Factors Determining Excretion of Drugs in Breast Milk

Drugs are primarily excreted into breast milk by passive diffusion, the same mechanism that occurs during placental transfer. Factors that determine the amount of drug in milk are the drug concentration in the mother (dependent on dose and maternal clearance rate), molecular weight, lipid solubility, protein binding, and degree of ionization at physiologic pH.

14 Drugs that have a low molecular weight and are unionized and lipophilic pass into milk to the greatest extent. The distribution of a drug between milk and plasma can be characterized by the milk:plasma ratio. The ratio varies over time, so the ideal ratio would be determined based on the respective concentration–time curves (AUC), usually over 24 hours but sometimes less; however, ratios based on single points are often reported. Because milk is slightly acidic (pH about 7.2) compared with plasma (pH 7.4), the milk:plasma ratios are less than 1 for weak acids, approximately 1 for neutral compounds, and greater than 1 for weak bases. Weak bases (eg, some beta-blockers) may accumulate in breast milk from ion trapping. For drugs with data available, most have a milk:plasma ratio of 1 or less, about 25% have ratios greater than 1, and about 15% have ratios greater than 2.15

An estimation of the infant dose received via milk is important because this can help to determine the effects of the drug in the nursing infant. Based on a typical daily milk consumption of 150 mL/kg, and knowing the average amount of drug in milk over a 24-hour period, the infant's dose can be compared with the mother's dose or, if known, with the recommended dose for infants of similar age and weight. Infant doses for many drugs, however, are not known, so the dose consumed by the infant from milk is often expressed as a percentage of the mother's weight-adjusted dose.16 Because for most drugs there is no information about the no-effect dose in infants, one author arbitrarily assumed that levels of a drug in milk were safe if they were 10% or less of the infant therapeutic dose or, if not known, the adult dose standardized by weight.15 However, this assumption would not apply to drugs in which toxicity could be caused by very small amounts.15

Drugs That Are Teratogenic in Humans

Of the thousands of drugs available for human use, only a few, are thought to be teratogenic. Although of vital importance, defects and toxicity induced by drugs of abuse (eg, cocaine, alcohol, cigarette smoking, toluene inhalation) are not covered here because they are addressed in another article. Complete avoidance during pregnancy of all teratogenic and toxic agents, including drugs of abuse, is the best approach. However, some of these drugs are indicated for pathologies commonly observed in women of childbearing age. Epilepsy, asthma, severe hypertension, cancer, rheumatoid arthritis, and thyroid disease are just a few examples of conditions that frequently require treatment during pregnancy with drugs that are known to cause fetal harm. Pharmacologic treatment of these conditions often improves the outcome of the pregnancy in comparison to no treatment or suboptimal treatment. Thus, the patient must be informed of the risk:benefit ratio and she and her partner must understand and agree with the decision to treat.

In the section below, reference to gestational weeks implies dating from the first day of the last menstrual period (LMP). Three common abbreviations used are CNS (central nervous system), NTD (neural tube defects), and IUGR (intrauterine growth retardation). Three reference books have been used to assemble the information cited.

5,6,17

ANDROGENIC HORMONES

Critical Period: labial fusion (8th to 13th gestational week), clitoral hypertrophy (second and third trimesters)

Magnitude of Risk: unknown

Mechanism: hormonal

Defects and Toxicity: masculinization of female fetus (female pseudohermaphroditism) characterized by a normal XX karyotype and internal female reproductive organs but ambiguous external genitalia; no fetal effects if exposure is stopped before onset of androgen receptor sensitivity in eighth gestational week; no adverse effects reported in males

ANGIOTENSIN-CONVERTING ENZYME (ACE) INHIBITORS

Critical Period: second and third trimesters

Magnitude of Risk: unknown; dose-and duration-dependent

Mechanism: decreased fetal angiotensin II results in renal impairment and hypotension

Defects and Toxicity: fetus—renal impairment/anuria resulting in oligohydramnios; IUGR; renal tubular dysplasia; hypocalvaria (oligohydramnios allows the uterus to press on top of the fetal head; combined with fetal hypotension, the uterine pressure prevents blood flow to that portion of the fetal skull, resulting in the defect; classified as a deformation); persistent patent ductus arteriosus; stillbirths; newborn—anuria and hypotension that is resistant to both volume expansion and pressor agents; renal failure may require hemodialysis

Comment: Monitoring of amniotic fluids levels is recommended, but fetal renal impairment (as indicated by oligohydramnios) may not be reversible.

ANGIOTENSIN II RECEPTOR ANTAGONISTS

Critical Period: second and third trimesters

Magnitude of Risk: unknown; dose-and duration-dependent

Mechanism: blocks angiotensin II at receptor site; net effect is the same as ACE inhibitors

Defects and Toxicity: losartan, valsartan—renal toxicity (anhydramnios), hypoplastic lungs, IUGR, skull hypoplasia, stillbirth (defects and toxicity are identical to those observed with ACE inhibitors)

ANTICOAGULANTS (WARFARIN)

Critical Period: sixth to ninth week, fetal warfarin syndrome (FWS); throughout gestation, CNS defects

Magnitude of Risk: 5% to 10% (has been as high as 50% in some studies) for FWS (facial anomalies and epiphyseal stippling); 4% to 5% for CNS and other defects

Mechanism: vitamin K deficiency in embryo; microhemorrhage and scarring (CNS defects)

Defects and Toxicity: FWS—most common characteristics are nasal hypoplasia (depressed nose with flattened, upturned appearance, commonly results in neonatal respiratory distress secondary to upper airway obstruction) and stippled epiphyses; other defects are IUGR, eye defects (when exposure also occurs in second and third trimesters), hypoplasia of extremities, congenital heart disease, and CNS damage (mental retardation, spasticity, seizures, scoliosis, deafness and hearing loss, and death)

ANTICONVULSANTS

Critical Period: first trimester (malformations), third trimester (early hemorrhagic disease of newborn)

Magnitude of Risk: two-to threefold increase over risk from epilepsy alone; 12% to 15% for anticonvulsant drug therapy; 1% to 2% NTD (valproic acid); 1% NTD (carbamazepine)

Mechanism: probably multifactorial (polygene–drug interaction); folic acid deficiency may contribute; carbamazepine, phenytoin, and valproic acid produce toxic epoxide metabolites; if fetus is homozygous for low epoxide hydrolase activity, then high risk for embryopathy (multifactorial); early hemorrhagic disease of the newborn may result from depletion of vitamin K stores in fetus

Defects and Toxicity: carbamazepine—NTD (1%), minor craniofacial defects, fingernail hypoplasia, and developmental delay; mephobarbital—cardiac defects, cleft lip/palate; paramethadione (see trimethadione); phenobarbital—cardiac defects, cleft lip/palate, hypoplasia of midface and fingers, microcephaly, IUGR, impaired cognitive development, and early hemorrhagic disease of the newborn; phenytoin—fetal hydantoin syndrome, includes craniofacial defects and limb defects (eg, fingernail hypoplasia) and early hemorrhagic disease of the newborn; primidone—similar to phenobarbital; trimethadione—fetal trimethadione syndrome (includes some cases of paramethadione), cardiac septal, craniofacial, and genitourinary defects, IUGR, postnatal growth deficiency, and mental retardation; valproic acid/valproate—NTD (1%–2%) (exposure between the 17th and 30th day after fertilization), craniofacial, digit, and urogenital defects, IUGR, retarded psychomotor development

ANTIDEPRESSANTS (LITHIUM)

Critical Period: first trimester (cardiac defects), third trimester (newborn toxicity)

Magnitude of Risk: less than 1% to 8% (cardiac defects); less than 1% (Ebstein's anomaly)

Mechanism: Unknown

Defects and Toxicity: cardiac defects, including Ebstein's anomaly; transient toxicity in newborns has included cyanosis, hypotonia, thyroid suppression with goiter, bradycardia, atrial flutter, cardiomegaly, electrocardiogram abnormalities (T-wave inversion), hepatomegaly, gastrointestinal bleeding, diabetes insipidus (may be prolonged), polyhydramnios, shock, and seizures

ANTI-INFECTIVES

Aminoglycosides (Kanamycin and Streptomycin)

Critical Period: throughout pregnancy

Magnitude of Risk: low, probably less than 3%; dose-and duration-dependent

Mechanism: direct toxicity of eighth cranial nerve

Defects and Toxicity: hearing loss/deafness; combination therapy with other ototoxic agents increases risk

Comment: Although eighth cranial nerve damage and renal impairment may occur with all aminoglycosides, this toxicity has not been associated with other members of this class. This most likely reflects greater attention to maternal serum levels and shorter courses of therapy.

Fluconazole

Critical Period: first trimester

Magnitude of Risk: unknown, dose-related (400 mg/day or greater)

Mechanism: unknown

Defects and Toxicity: brachycephaly, craniosynostosis, proptosis, low nasal bridge, cleft palate, femoral bowing, thin ribs and long bones, joint contractures, cardiac malformations

Quinine (High-Dose)

Critical Period: first trimester

Magnitude of risk: observed only after use of toxic doses in unsuccessful abortion attempts

Mechanism: unknown

Defects and Toxicity: CNS anomalies (hydrocephalus, deafness, optic nerve damage) and limb defects most frequent; other defects involved the face, heart, and gastrointestinal and urogenital systems

Tetracycline

Critical Period: beyond fourth month

Magnitude of Risk: unknown, duration-dependent

Mechanism: chelation; drug forms a complex with calcium orthophosphate and is incorporated into bones and teeth undergoing calcification; complex is permanent in teeth because remodeling and calcium exchange do not occur after calcification is completed

Defects and Toxicity: intense yellow-gold fluorescence of mineralized skeletal structures and teeth; impairment of bone growth may occur but is not clinically significant

CORTICOSTEROIDS, SYSTEMIC (ALL MEMBERS OF CLASS)

Critical Period: before 10 weeks after LMP (oral clefts), IUGR (throughout gestation)

Magnitude of Risk: background risk for nonsyndromic cleft lip with or without cleft palate is 0.1–0.2%; may be increased two-to sevenfold;

unknown for IUGR, but prolonged therapy is required

Mechanism: unknown

Defects and Toxicity: nonsyndromic cleft lip with or without cleft palate; IUGR (about 300–400 g)

DIAGNOSTICS AND ANTIDOTES

Methylene Blue (Intra-amniotic Injection)

Critical Period: second and third trimesters

Magnitude of Risk: about 20% for jejunal–ileal atresias

Mechanism: vascular disruption due to arterial constriction, but cannot exclude methemoglobinemia-or hemolytic anemia-induced hypoxia shunting blood away from the intestine or a direct toxic effect of the dye when swallowed with amniotic fluid

Defects and Toxicity: jejunal and ileal atresias; fetal death has occurred; newborn toxicity includes hyperbilirubinemia, hemolytic anemia with or without Heinz body formation, methemoglobinemia, and deep blue staining of skin

Penicillamine

Critical Period: unknown

Magnitude of Risk: unknown, probably less than 5%

Mechanism: chelation of metals (eg, copper)

Defects and Toxicity: cutis laxa; limiting dose to 1 g/d or less during gestation may reduce risk; if cesarean section is planned, limit dose to 250 mg/day for 6 weeks before delivery and after surgery until wound healing is complete

SYNTHETIC ESTROGENS (DIETHYLSTILBESTROL)

Critical Period: 10 to 13 weeks of gestation (highest risk of vaginal adenocarcinoma but some risk up to 18 weeks); up to 20 weeks (genital malformations)

Magnitude of Risk: less than 0.1% (about 350–450 cases of vaginal cancer); unknown for genital malformations

Mechanism: possible inhibition of upward growth of vaginal plate or stimulation of müllerian epithelium so that it persists in the developing vagina

Defects and Toxicity: female: lower müllerian tract—vaginal adenosis, vaginal and cervical clear cell adenocarcinoma (more than 90% occur after 14 years of age), cervical and vaginal fornix defects, cockscomb (hood, transverse ridge of cervix), collar (rim, hood, transverse ridge of cervix), pseudopolyp, hypoplastic cervix, altered fornix of vagina; vaginal defects (exclusive of fornix)—incomplete transverse and/or longitudinal septum; upper Müllerian tract—uterine structural defects, fallopian tube defects; male: epididymal cysts, hypotrophic testis, microphallus, varicocele, capsular induration, altered semen (decreased count, concentration, motility, and morphology)

GASTROINTESTINAL AGENT (MISOPROSTOL)

Critical Period: highest risk occurs with exposure in weeks 6 to 8, but any time during first trimester conveys some risk

Magnitude of Risk: up to seven times background risk for Möbius syndrome and other malformations; almost all adverse outcomes have been related to unsuccessful abortion attempts with large, sometimes massive doses (orally, vaginally, or both); only a few cases have been reported when the drug was used for its approved indication

Mechanism: vascular disruption caused by uterine contractions; deformation

Defects and Toxicity: vaginal bleeding, abortion; if abortion does not occur, reported defects include Möbius syndrome (sixth and seventh nerve palsies), defect of cranium and overlying scalp, cleft lip/palate, ocular hypertelorism, arthrogryposis (multiple nonprogressive congenital joint contractures; may be a result of neurologic impaired leg movement; in addition, bilateral talipes equinovarus is a common feature), terminal transverse-limb defects (missing metacarpals and phalanges, hypoplasia of the thumbs and fingers), syndactyly, limb constriction ring or skin scars, and hydrocephalus

IMMUNOMODULATOR (THALIDOMIDE)

Critical Period: days 34 to 50 days after LMP (20–36 days after conception); critical maternal dose is at least 100 mg

Magnitude of Risk: 20–50%

Mechanism: one current theory is that thalidomide, or its metabolite, intercalates into DNA of specific promoter regions of genes that code for proteins involved in normal limb development, thereby inhibiting transcription of these genes and disrupting development of new blood vessels, resulting in truncation of limbs

Defects and Toxicity: limb defects—bilateral amelia or phocomelia of upper and lower limbs and various lesser degrees of reduction defects; osteochondritis of femoral head; laxity of cruciate ligaments in knee joints; other skeletal defects—malformations of spine, shoulder, hip/pelvis, and jaw; craniofacial—multiple defects of eyes, ears, face and skull, tongue, nose (including choanal atresia), teeth, midline hemangioma or nevus; CNS—facial nerve palsy, hydrocephalus, NTD, epilepsy, autism, Marcus Gunn phenomenon (jaw winking syndrome), crocodile-tear syndrome; major organs—multiple malformations of respiratory, cardiovascular, gastrointestinal, and genitourinary systems; miscellaneous—excessive sweating, inguinal hernias

ANTITHYROID AGENTS

Iodine

Critical Period: second and third trimesters

Magnitude of Risk: dose-and duration-dependent

Mechanism: inhibition of fetal thyroid gland with compensatory hypertrophy

Defects and Toxicity: hypothyroidism, goiter, may cause tracheal compression in newborn

Methimazole and Carbimazole

Critical Period: up to 9 weeks after LMP

Magnitude of Risk: unknown but probably low

Mechanism: possible phenotype (polygene–drug interaction)

Defects and Toxicity: a pattern of rare congenital malformations, possibly indicating a phenotype, that consists of some or all of the following: scalp (aplasia cutis congenita) or patchy hair defects, choanal atresia, esophageal atresia with tracheoesophageal fistula, minor facial anomalies (short up-slanting palpebral fissures, epicanthal folds, small nose and mouth, short philtrum), hypoplastic (hypothelia) or absent (athelia) nipples, psychomotor delay; goiter

VITAMIN A DERIVATIVES

Etretinate and Acitretin

Critical Period: day 15 after conception to end of first trimester

(same as isotretinoin)

Magnitude of Risk: unknown; may be same as isotretinoin

Mechanism: unknown; may be same as isotretinoin

Defects and Toxicity: upper/lower limb reduction defects, other skeletal anomalies (multiple synostoses, hip, cervical vertebrae), NTD and CNS defects (meningomyelocele, meningoencephalocele, peripheral facial nerve paresis), head (microcephaly, small mandible), facial dysmorphia (asymmetric nares, microtia or low-set, protruding ears with malformed antihelices, and enlarged, keyhole-shaped entrances to external ear canals), strabismus, cardiac malformations (including tetralogy of Fallot), syndactylies, and poor head control

Comment: Acitretin, an active metabolite of etretinate, undergoes interconversion to 13-cis-acitretin, also an active metabolite. In addition, when alcohol is consumed when acitretin is still in the system, acitretin may be converted back to etretinate, which has a very long elimination half-life (mean 120 days, but may be as long as 168 days). Therefore, current recommendations state that alcohol consumption should be avoided during therapy and for 2 months after cessation of therapy. Moreover, because of the prolonged elimination times of etretinate, acitretin, and 13-cis-acitretin, effective contraception should be used for at least 3 years after stopping acitretin and etretinate therapy.

Isotretinoin

Critical Period: day 15 after conception to end of first trimester

Magnitude of Risk: 30–50%; teratogenic dose range less than 0.2 to 1.5 mg/kg/day

Mechanism: disruption of initial differentiation and migration of cephalic neural crest cells

Defects and Toxicity: spontaneous abortions (may be most common adverse outcome); CNS—hydrocephalus, facial nerve (seventh) palsy, posterior fossa structure defects, cortical and cerebellar defects, cortical blindness, optic nerve hypoplasia, retinal defects, microphthalmia; craniofacial—facial dysmorphism, microtia or anotia, low-set ears, agenesis or marked stenosis of external ear canals, micrognathia, small mouth, microcephaly, depressed nasal bridge, triangular skull, cleft palate, hypertelorism; cardiovascular—transposition of great vessels, tetralogy of Fallot, double-outlet right ventricle, truncus arteriosus communis, ventricular septal defect, atrial septal defect, interrupted or hypoplastic aortic arch, retroesophageal right subclavian artery; thymic gland defects—ectopia, hypoplasia, or aplasia; miscellaneous defects (sporadic occurrence)—spina bifida, nystagmus, hepatic abnormality, hydroureter, decreased muscle tone, large scrotal sac, simian crease, limb reduction defects

Drugs That Cause Toxic Effects in the Fetus or Newborn

As shown in

Table 1, pharmacologic therapy during pregnancy can produce, in addition to birth defects, fetal and neonatal toxicity. This toxicity is an extension of a drug's pharmacologic effects and as a rule is identical to the toxicity observed in patients treated directly with the drug. As above, drugs of abuse are not discussed. Fortunately, most toxicity is transient or reversible when exposure to the offending drug is stopped. Toxicity in the fetus or newborn is frequently the result of prolonged, continuous use of the drug by the mother. However, there are notable exceptions to this rule. For example, abortions produced by mifepristone and misoprostol, bradycardia resulting from drug-induced maternal hypotension, and fetal/newborn toxicity associated with the use of narcotic analgesics close to delivery (eg, sinusoidal fetal heart rate patterns, newborn respiratory depression, and neonatal neurobehavioral dysfunction) may all occur with single doses or very brief therapeutic courses. Therefore, when a woman requires drug therapy during pregnancy, particularly continuous therapy, the lowest possible dose should be used, and close attention should be given to detect any signs and symptoms of toxicity.

Adverse Effects in the Infant From Drugs in Milk

As the physiologic process that governs the placental crossing of drugs to the embryo and fetus is similar to the excretion of drugs into breast milk, so the toxicity produced in the embryo and fetus is similar to the toxicity produced in the nursing infant. In other words, the toxic effects of a drug during lactation are specifically related to the pharmacologic characteristics of the drug. Six drugs of abuse (amphetamines, cocaine, ethanol, heroin, marijuana, and PCP) have been covered here because they are not covered elsewhere. With the exception of those agents contraindicated during nursing, the toxicity of most drugs is transient and reversible. Alert health professionals and mothers are the best defense against allowing this toxicity to become long-lasting or to cause permanent harm. Having said that, however, few studies have been conducted on the consequences of exposure in milk to neuroactive drugs on long-term neurobehavior. Psychotropic agents change the brain chemistry of the mother and may so in a nursing infant. That this may occur during a rapidly changing and very vulnerable period of infant neurodevelopment heightens the concern. The American Academy of Pediatrics has recognized this potential toxicity by placing antianxiety, antidepressant, and antipsychotic agents in a special category that exemplifies their concerns for long-term consequences. Fortunately, the few studies that have examined the effects on infant neurobehavior from exposure to psychoactive drugs in milk have not discovered long-term toxicity. This is reassuring, but more studies in this regard are warranted.

Conclusion

The best defense against drug-induced teratogenicity and toxicity during both pregnancy and lactation is knowledgeable and alert health-care professionals and patients. This combination can be very effective in preventing disasters in pregnancy and in nursing infants. There are a wide variety of resources available to professionals and the lay public that provide information on these subjects. Although the overall effectiveness of these resources is not entirely known, they do appear to be effective. The absence of pregnancy disasters similar in magnitude to the one that occurred with thalidomide 40 years ago appears to be evidence that the system is working. However, preventable congenital defects still occur. Although their actual numbers are small in relation to the total, a further reduction is certainly achievable, and so much remains to be accomplished.

To reiterate, steps can be taken to lessen the dilemma posed by prescribing, dispensing, and administering drugs to women who are or who may become pregnant or who are breast feeding. First, health-care professionals providing services to this population should be knowledgeable about the background risk and the risk that a drug represents to the embryo, fetus, or newborn. They should also know which drugs should be absolutely avoided during lactation and which, although compatible with breast feeding, may cause toxicity. Second, this information must be communicated to the patient in such a manner that she clearly understands the risks and benefits of therapy. Additionally, she needs to appreciate the risks of not properly treating her condition. Third, a system should be set up between the patient and her health care provider to assure close communication in the event her pregnancy status changes while she is receiving drug therapy. Although adherence to this type of system may be difficult, it should be a major factor in improving patient compliance and lessening the anxiety of medication use in this population. Moreover, it will assist in achieving a worthy and obtainable goal: the prevention of unnecessary poor outcomes in pregnancy and during breast feeding.

References

 

1. Oakley Jr. GP Frequency of human congenital malformations. Clin Perinatol 1986;13:545–54.  

2. Hook EB, Regal RR. Representative and misrepresentative associations of birth defects in livebirths. Am J Epidemiol 1993;137:660–75.  

3. Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Engl J Med 1998;338:1128–37.  

4. Kalter H, Warkany J. Congenital malformations. Etiologic factors and their role in prevention. N Engl J Med 1983;308:424–31.  

5. Shepard TH. Catalog of Teratogenic Agents. 10th ed. Baltimore: Johns Hopkins University Press; 2001.  

6. Schardein JL. Chemically Induced Birth Defects. 3d ed. New York: Marcel Dekker; 2000.  

7. Chambers CD, Braddock SR, Briggs GG, et al. Post-marketing surveillance for human teratogenicity: a model approach. Teratology 2001 (in press).  

8. Martinez-Frias ML, Bermejo E, Frias JL. Analysis of deformations in 26,810 consecutive infants with congenital defects. Am J Med Genet 1999;84:365–8.  

9. Koren G, Bologa M, Pastuszak A. How women perceive teratogenic risk and what they do about it. Ann NY Acad Sci 1993;678:317–24.  

10. Committee on Drugs, American Academy of Pediatrics. The transfer of drugs and other chemicals into human milk. Pediatrics 2001;108:776–789.  

11. Brent RL. Method of evaluating alleged human teratogens [editorial]. Teratology 1978;17:83.  

12. Sastry BVR. Techniques to study human placental transport. Adv Drug Deliver Rev 1999;38:17–39.  

13. Garland M. Pharmacology of drug transfer across the placenta. Obstet Gynecol Clin North Am 1998;25:21–42.  

14. Agatonovic-Kustrin S, Tucker IG, Zecevic M, et al. Prediction of drug transfer into human milk from theoretically derived descriptors. Anal Chim Acta 2000;418:181–95.  

15. Ito S. Drug therapy breast-feeding women. N Engl J Med 2000;343:118–26.  

16. Atkinson HC, Begg EJ, Darlow BA. Drugs in human milk. Clinical pharmacokinetic considerations. Clin Pharmacokinet 1988;14:217–40.  

17. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation. A Reference Guide to Fetal and Neonatal Risk. 6th ed. Philadelphia: Lippincott Williams & Wilkins; 2001.

Correspondence: Gerald G. Briggs, B Pharm, 9802 Saline Drive, Huntington Beach, CA 92646

Clin Obstet Gynecol 2002 March;45(1):6-21

Copyright © 2002 Lippincott Williams & Wilkins

All rights reserved

................
................

In order to avoid copyright disputes, this page is only a partial summary.

Google Online Preview   Download