throbber
The New England Journal of Medicine
`
`Review Article
`
`Drug Therapy
`
`A
`
`L A S T A I R
`
` J.J. W
`
`O O D
`
`, M.D.,
`
` Editor
`
`D
`
`RUGS
`
`
`
`IN
`
` P
`
`REGNANCY
`
`G
`
`IDEON
`
` P
`, M.D., A
` K
`ASTUSZAK
`NNE
`OREN
` S
` I
`, M.D.
`AND
`HINYA
`TO
`
`, M.S
`
`C
`
`.,
`
`B
`
`EFORE marketing a new drug, the manufac-
`turer almost never tests the product in preg-
`nant women to determine its effects on the fe-
`tus. Consequently, most drugs are not labeled for
`use during pregnancy. Typically, descriptions of drugs
`
`Physicians’ Desk Reference and
`that appear in the
`similar sources contain statements such as, “Use in
`pregnancy is not recommended unless the potential
`benefits justify the potential risks to the fetus.” Since
`the risk has been adequately established for only a
`few drugs, physicians caring for pregnant women
`have very little information to help them decide
`whether the potential benefits to the mother out-
`weigh the risks to the fetus. These typical disclaim-
`ers, although understandable from the medicolegal
`standpoint, put large numbers of women and their
`physicians in difficult situations for several reasons.
`One is that at least half the pregnancies in North
`America are unplanned,
` and every year, hundreds of
`1
`thousands of women therefore expose their fetuses
`to drugs before they know they are pregnant. Such
`women often interpret the statement that use during
`pregnancy is not recommended as meaning that the
`drug is not safe during pregnancy. There is evidence
`that this perception of fetal risk causes many women
`to consider or even seek termination of otherwise
` Another reason is that with
`wanted pregnancies.
`2,3
`the recent increase in the age at which women have
`children, conditions that necessitate long-term drug
`therapy are diagnosed in larger numbers of women
`
`From the Motherisk Program, Division of Clinical Pharmacology and
`Toxicology (G.K., A.P., S.I.), the Departments of Pediatrics and Population
`Health Sciences (G.K., S.I.), and the Research Institute (G.K., S.I.), Hos-
`pital for Sick Children; and the Departments of Pediatrics (G.K., S.I.),
`Pharmacology (G.K., S.I.), and Medicine (G.K.), University of Toronto —
`both in Toronto. Address reprint requests to Dr. Koren at the Division of
`Clinical Pharmacology, Hospital for Sick Children, 555 University Ave.,
`Toronto, ON M5G 1X8, Canada.
`©1998, Massachusetts Medical Society.
`
`1128
`
`ⴢ
`
`April 16, 1998
`
`before pregnancy. Furthermore, for pregnant wom-
`en with certain conditions once believed to be in-
`compatible with pregnancy, such as systemic lupus
`erythematosus and heart diseases, the outcome of
`pregnancy has improved dramatically in the past few
`decades.
`4
`In this article, we review current knowledge of the
`fetal and neonatal effects of prescription and over-
`the-counter drugs given to pregnant women, with
`an emphasis on the approaches used to determine
`safety and risk. In addition, we review approaches to
`communicating such information to pregnant wom-
`en and their families.
`
`HUMAN TERATOGENESIS
`Teratogenesis is defined as the dysgenesis of fetal
`organs as evidenced either structurally or function-
`ally (e.g., brain functions).
` The typical manifesta-
`5
`tions of teratogenesis are restricted growth or death
`of the fetus, carcinogenesis, and malformations,
` de-
`6
`fined as defects in organ structure or function. These
`abnormalities vary in severity (e.g., hypospadias that
`is mild and may be missed, or is severe, necessitating
`several corrective operations). Major malformations
`may be life-threatening and require major surgery or
`may have serious cosmetic or functional effects.
`
`A HISTORICAL PERSPECTIVE
`Several milestones highlight the problems of drug
`therapy facing pregnant women, their families, and
`health professionals.
`
`Thalidomide
`For decades it was believed that the placenta
`served as a barrier that protected the fetus from the
`adverse effects of drugs. The thalidomide disaster
`drastically changed this perception by demonstrat-
`ing that fetal exposure to the drug during critical pe-
`riods of development resulted in severe limb defects
`and other organ dysgenesis (e.g., kidney and heart
` Despite the high rates of malformations
`defects).
`6,7
`(20 to 30 percent) and their characteristic pattern,
`the teratogenicity of thalidomide was not suspected
`for years. The suffering it caused has prompted the
`belief that every drug has the potential to be a new
`thalidomide.
`2,3
`Most known human teratogens are associated
`with much lower rates of malformations, and the
`syndromes they cause are not always so pathogno-
`monic, making causation more difficult to confirm.
`Yet 35 years after the recognition of thalidomide-
`associated embryopathy, fewer than 30 drugs have
`been proved to be teratogenic in humans when used
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Page 1 of 10
`
`CELGENE EXHIBIT 2013
`Coalition for Affordable Drugs VI LLC (Petitioner) v. Celgene Corporation (Patent Owner)
`Case IPR2015-01103
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`DRUG THERAPY
`
`in clinically effective doses, and even fewer are cur-
`rently in clinical use (Table 1). Many other com-
`monly used drugs, including salicylates, glucocorti-
`coids, and spermicides, were once thought to be
`teratogenic but have been shown to be safe in sub-
`sequent studies that were larger and better con-
`trolled than the initial studies (Table 2).
`
`Bendectin
`One example of the gap between the perception
`of teratogenic risk and evidence-based proof of safety
`is the case of Bendectin. During the late 1950s and
`the 1960s, this drug, a combination of an antihista-
`mine (doxylamine) and pyridoxine, was the most
`widely used medication in the United States for nau-
`sea and vomiting associated with pregnancy. During
`the 1970s, many lawsuits claiming that Bendectin
`was teratogenic were filed against the manufacturer
`in American courts. Therefore, the drug was with-
`drawn from the market by its manufacturer in 1982,
`which left millions of pregnant women without a
`drug approved by the Food and Drug Administra-
`
`tion (FDA) for the treatment of nausea and vomit-
`ing. The rate of hospitalization for severe nausea and
`vomiting during pregnancy increased by a factor of
`2 in both the United States and Canada after Ben-
`dectin was withdrawn from the market (Fig. 1).
`The drug was withdrawn despite a substantial
`body of evidence that the rate of major malforma-
`tions among the children of women who had re-
`ceived Bendectin during pregnancy did not differ
`from the rate in the general population.
` With-
`24,25
`drawal of the drug from the American market did
`not decrease the rate of any specific category of mal-
`formation, as would be expected for a truly terato-
`genic drug estimated to have been used by up to 40
`percent of pregnant women at one time.
`26,27
`In Canada, the drug continues to be marketed un-
`der the trade name Diclectin. A review committee has
`advised the Canadian Minister of Health that the
` A recent study revealed that severe nau-
`drug is safe.
`27
`sea and vomiting of pregnancy often lead women to
`terminate or consider the termination of otherwise
`wanted pregnancies.
` Other formulations of dox-
`28
`
`T
` 1.
`ABLE
`
` D
`
`
`RUGS
`
`WITH
`
` P
` T
`ROVEN
`
` E
`
`ERATOGENIC
`FFECTS
`
` H
`.*
`IN
`UMANS
`
`D
`RUG
`
`T
`ERATOGENIC
`
` E
`FFECT
`
`Aminopterin†, methotrexate
`Angiotensin-converting–enzyme inhibitors
`
`Anticholinergic drugs
`Antithyroid drugs (propylthiouracil and meth-
`imazole)
`Carbamazepine
`Cyclophosphamide
`Danazol and other androgenic drugs
`Diethylstilbestrol†
`
`CNS and limb malformations
`Prolonged renal failure in neonates, decreased skull
`ossification, renal tubular dysgenesis
`Neonatal meconium ileus
`Fetal and neonatal goiter and hypothyroidism, apla-
`sia cutis (with methimazole)
`Neural-tube defects
`CNS malformations, secondary cancer
`Masculinization of female fetuses
`Vaginal carcinoma and other genitourinary defects
`in female and male offspring
`Neonatal hypoglycemia
`Ebstein’s anomaly
`Moebius sequence
`Constriction of the ductus arteriosus‡, necrotizing
`enterocolitis
`Facial and CNS defects
`Paramethadione†
`Growth retardation, CNS deficits
`Phenytoin
`Neonatal withdrawal syndrome when drug is taken
`Psychoactive drugs (e.g., barbiturates, opioids,
`in late pregnancy
`and benzodiazepines)
`Systemic retinoids (isotretinoin and etretinate) CNS, craniofacial, cardiovascular, and other defects
`Tetracycline
`Anomalies of teeth and bone
`Thalidomide
`Limb-shortening defects, internal-organ defects
`Trimethadione†
`Facial and CNS defects
`Valproic acid
`Neural-tube defects
`Warfarin
`Skeletal and CNS defects, Dandy–Walker syndrome
`
`Hypoglycemic drugs
`Lithium
`Misoprostol
`Nonsteroidal antiinflammatory drugs
`
`*Only drugs that are teratogenic when used at clinically recommended doses are listed. The list
`includes all drugs proved to affect neonatal morphology or brain development and some of the toxic
`manifestations predicted on the basis of the pharmacologic actions of the drugs. Data are from Briggs
`et al.
` CNS denotes central nervous system.
`8
`†The drug is not currently in clinical use.
`‡Sulindac probably does not have this effect.
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Volume 338 Number 16
`
`ⴢ
`
`1129
`
`Page 2 of 10
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`The New England Journal of Medicine
`
` 2.
`ABLE
`
` C
`
` D
` I
` T
`
`OMMON
`RUGS
`NITIALLY
`HOUGHT
`
`TO
`
` B
` T
`
`E
`ERATOGENIC
`
` S
` P
` S
`.
`BUT
`UBSEQUENTLY
`ROVED
`AFE
`
`T
`
`D
`RUG
`
`I
`NITIAL
`
` E
`VIDENCE
`
`
`
`OF
`
` R
`ISK
`
`S
`UBSEQUENT
`
` E
`VIDENCE
`
`
`
`OF
`
` S
`AFETY
`
`Diazepam*
`
`Oral clefts
`9
`
`Oral contraceptives
`
`Spermicides
`
`Salicylates
`
`Birth defects involving the vertebrae, anus,
`heart, trachea, esophagus, kidney, and
`limbs
`; masculinizing effects on female fe-
`13
`tuses resulting in pseudohermaphroditism
`14
`Limb defects, tumors, Down’s syndrome,
`and hypospadias
`
`17
`Cleft palate
` and congenital heart disease
`19
`
`Bendectin (doxylamine
`plus pyridoxine)
`
`Cardiac and limb defects
`22,23
`
`
`
`No increase in risk in large cohort and case–
`control studies
`
`10-12
`No association between first-trimester expo-
`sure to oral contraceptives and malforma-
`tions in general or external genital mal-
`
`formations in two meta-analyses
`15,16
`No increase in risk in a meta-analysis
`18
`
`No increase in risk in large cohort
`studies
`20,21
`No increase in risk in two meta-analyses
`
`24,25
`
`*Diazepam taken near term may cause the neonatal withdrawal syndrome or cardiorespiratory instability.
`
`this belief became evident after isotretinoin was in-
`troduced in North America in the early 1980s for
`the treatment of acne. For years before its clinical in-
`troduction, this drug had been known to cause mal-
`formations in animals.
` Despite explicit warning la-
`29
`bels, scores of children with retinoid embryopathy
`were born in the years after the drug was intro-
`duced.
` Such warnings are not sufficient, because
`30
`women taking isotretinoin may not plan their preg-
`nancies, or their birth-control methods may fail. In
`addition, some women and men are functionally il-
`literate, and they may not read or understand the
`content of a drug label.
`31
`The initial experience with isotretinoin led to the
`development of a more comprehensive program to
`prevent teratogenesis. The Retinoid Pregnancy Pre-
`vention Program includes explicit and detailed print-
`ed warnings as well as a line drawing of a malformed
`child,
` and as part of the program, women are asked
`32
`to sign a consent form indicating that they agree to
`use two effective methods of contraception before
`therapy is started. Since the program was implement-
`ed in 1989, a substantial number of fetuses have
`been exposed to the drug. As many as 30 percent of
`the women with exposed fetuses did not use any
`mode of contraception, even though they were cog-
`nizant of the high fetal risk.
` Many of these women
`32
`explained that they did not believe they were fertile,
`since they had not conceived during periods of
`months or years when they had not used contracep-
`tive methods.
`33
`
`CURRENT TRENDS IN PREVENTING FETAL
`EXPOSURE TO TERATOGENS
`The advent of effective injectable hormonal con-
`traceptives has made it possible to minimize the risk
`of an unplanned pregnancy during therapy with a
`known teratogen. This approach was first implement-
`ed in South America, where sexually active women
`with cutaneous leprosy were injected with medroxy-
`
`ylamine in combination with pyridoxine are available
`in other countries (e.g., South Africa, Spain, and
`Thailand).
`
`Isotretinoin
`The experience with thalidomide led drug regula-
`tors, drug manufacturers, and the medical commu-
`nity to believe that appropriate labeling of terato-
`genic drugs, with warnings not to take them around
`the time of conception, would be effective in pre-
`venting fetal exposure to the drugs. The naiveté of
`
`Prescriptions (⫻10⫺3)
`
`140
`
`120
`
`100
`
`80
`
`60
`
`40
`
`02
`
`0
`
`Hospitalization
`
`Bendectin
`
`Diclectin
`
`20
`
`15
`
`10
`
`5
`
`Hospitalizations (per 1000 live births)
`
`0
`1979
`
`1980
`
`1981
`
`1982
`
`1983
`
`1986
`
`1987
`
`1988
`
`1989
`
`1984
`1985
`Year
`
`Figure 1.
` Rates of Hospitalization among Pregnant Women
`with Severe Nausea and Vomiting and Numbers of Prescrip-
`tions for Bendectin and Diclectin in North America, 1979
`through 1989.
`Bendectin was withdrawn from the U.S. market in 1982, where-
`as Diclectin, the same drug, remained on the market in Cana-
`da. Adapted from Neutel and Johansen with the permission of
`the publisher.
`
`26
`
`1130
`
`ⴢ
`
`April 16, 1998
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Page 3 of 10
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`DRUG THERAPY
`
`progesterone before receiving a prescription for tha-
`lidomide.
` Yet numerous new cases of thalidomide-
`34
`associated embryopathy have been reported in the
`children of women who continued to take the drug
`after the period of contraceptive efficacy (three
`months) or who received the drug from their male
`partners.
`34
`Because any new drug may be teratogenic, it is
`important to develop more effective methods to pre-
`vent fetal exposure. One such method may be the
`use of implantable hormonal compounds (e.g.,
`levonorgestrel implants), which can provide long-
`term, reversible contraception for up to five years.
`Levonorgestrel implants have documented efficacy
`in young women in whom oral methods of contra-
`ception are likely to fail.
` Implants should be con-
`35
`sidered by sexually active women who are taking a
`teratogen medicinally (e.g., phenytoin or warfarin)
`or as part of a pattern of substance abuse (e.g., alco-
`hol or cocaine). Furthermore, women taking terato-
`genic drugs who are not sexually active should be in-
`formed of the availability of effective postcoital
`contraceptives.
`35,36
`
`THE PROCESS OF ESTABLISHING RISK
`OR SAFETY OF DRUGS IN PREGNANCY
`Every year, many new drugs are approved and
`marketed. By this stage, several thousand people
`have usually participated in studies of the drugs, but
`the majority have been men. Since there are scarcely
`any data on fetal effects at the time of marketing,
`data from studies in animals provide the initial
`guidelines.
`
`The Value of Studies in Animals
`Typically, studies of reproductive toxicology in an-
`imals compare the outcome of pregnancy in groups
`of animals receiving a range of doses of the drug in
`question during the period of organogenesis with
`the outcome in untreated (control) animals. The oc-
`currence of thalidomide-associated embryopathy led
`to the erroneous belief that human teratogenicity
`could not be predicted on the basis of studies in an-
`imals. However, every drug that has since been
`found to be teratogenic in humans has caused simi-
`lar teratogenic effects in animals (Table 3), except
`misoprostol, which causes a morphologic pattern
`known as the Moebius sequence in humans. In at
`least one case, that of isotretinoin, the studies in an-
`imals probably prevented a disaster similar to that of
`thalidomide.
`29
`However, there are drugs that have teratogenic ef-
`fects in animals when administered in high doses
`that are not teratogenic in humans given clinically
`relevant doses. For example, high doses of gluco-
` or benzodiazepines
` can cause
`corticoids
`19,70-75
`76,77
`oral clefts in animals, but clinically relevant doses in
`humans have no such effects.
` Similarly, sa-
`10-12,75
`
`licylates
` cause cardiac malformations in animals
`78-80
`but not in humans.
` Such discrepancies have led
`20,21
`to unwarranted anxiety on the part of women, their
`families, and physicians and may have contributed to
`unnecessary terminations of pregnancies.
` Although
`3
`studies in animals may identify teratogenic effects, it
`can be difficult to extrapolate these effects to humans.
`
`Epidemiologic Studies
`In addition to studies in animals, a variety of other
`approaches are used to identify possible drug terato-
`genicity and to assess the relation between drug ex-
`posure and fetal outcome. The first accounts of ad-
`verse fetal outcomes after exposure to a marketed
`drug are usually published in the form of case re-
`ports. These reports can be either very useful or use-
`less in establishing teratogenic risk on the basis of
`relatively simple statistical considerations. If the drug
`in question is taken by relatively small numbers of
`women (e.g., isotretinoin
`) or causes a rare malfor-
`30
`mation (e.g., ear agenesis
`), then a small number of
`81
`cases can establish a strong association. Warfarin,
`9
`diethylstilbestrol,
` and isotretinoin
` were originally
`82
`83
`identified as human teratogens on the basis of case
`reports. If, on the other hand, the drug is taken by
`many pregnant women (e.g., Bendectin), a small
`number of case reports of abnormalities may simply
`reflect the spontaneous occurrence of malformations
`in the general population, which ranges from 1 to
`5 percent, unless there is a characteristic pattern of
`malformations (as, for example, with alcohol or tha-
`lidomide). To date, prenatal exposure to many of the
`known human teratogens has been associated with
`characteristic patterns of malformations, and this has
`become an important tenet in establishing terato-
`genicity.
`Epidemiologic studies are typically designed to de-
`termine whether mothers who took a specific drug
`during pregnancy have a larger number of mal-
`formed children than mothers who did not (cohort
`studies) or whether mothers of children with a spe-
`cific malformation took the drug more often than
`mothers of children without the malformation (case–
`control studies).
`With the international development of teratology-
` a new source of data for pro-
`information services,
`84
`spective observational research has emerged. Preg-
`nant women taking prescription or over-the-counter
`drugs voluntarily call these centers for risk-assess-
`ment counseling, usually during the first trimester.
`Since the exposure data are recorded prospectively,
`the probability of recall bias is reduced, and follow-
`up of exposed pregnancies can extend well beyond
`parturition. Collaboration among these services can
`yield the large samples needed to study rare events
`more effectively.
`53,61,85
`Drug manufacturers may perform postmarketing
`cohort studies of prospectively reported exposures.
`
`Volume 338 Number 16
`
`ⴢ
`
`1131
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Page 4 of 10
`
`

`
`The New England Journal of Medicine
`
`T
`
` 3.
`ABLE
`
` T
` E
`ERATOGENIC
`FFECTS
`
`
`
` D
`
`OF
`RUGS
`
`IN
`
` ANIMALS AND HUMANS.*
`
`DRUG
`
`EFFECTS IN ANIMALS
`
`EFFECTS IN HUMANS
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Stillbirths and increased fetal loss in
`sheep and rabbits37
`
`Prolonged renal failure and hypotension in the newborn, decreased skull ossification, hypo-
`calvaria, and renal tubular dysgenesis38
`
`Neural-tube defects 40
`
`Growth retardation involving weight, length, and head circumference43; placental
`abruption44,45 and uterine rupture
`
`Fetal alcohol syndrome: prenatal and postnatal growth deficiency, CNS anomalies (micro-
`cephaly, behavioral abnormalities, and mental retardation), characteristic pattern of facial
`features (short palpebral fissures, hypoplastic philtrum, and flattened maxilla), and major
`organ-system malformations49; with age, facial features may become less distinctive, but
`short stature, microcephaly, and behavioral abnormalities persist50
`Retinoid embryopathy resulting in some or all of the following abnormalities30: CNS defects
`(hydrocephalus, optic-nerve blindness, retinal defects, microphthalmia, posterior fossa de-
`fects, and cortical and cerebellar defects); craniofacial defects (microtia or anotia, low-set
`ears, hypertelorism, depressed nasal bridge, microcephaly, micrognathia, and agenesis or
`stenosis of external ear canals); cardiovascular defects (transposition of great vessels, te-
`tralogy of Fallot, and ventricular or atrial septal defects); thymic defects (ectopia and hy-
`poplasia or aplasia); and miscellaneous defects (limb reduction, decreased muscle tone,
`spontaneous abortion, and behavioral abnormalities)
`Ebstein’s anomaly and other heart defects52,53
`Fetal Minamata disease: diffuse neuronal disintegration with gliosis, cerebral palsy, micro-
`cephaly, strabismus, blindness, speech disorders, motor impairment, abnormal reflexes,
`and mental retardation56
`
`Fetal hydantoin syndrome60: prenatal and postnatal growth deficiency, motor or mental de-
`ficiency, short nose with broad nasal bridge, microcephaly, hypertelorism, strabismus,
`epicanthus, wide fontanelles, low-set or abnormally formed ears, positional deformities
`of limbs, hypoplasia of nails and distal phalanges, hypospadias, hernia, webbed neck, low
`hairline, impaired neurodevelopment and low performance scores on tests of intelligence61
`Limb-shortening defects,63 loss of hearing, abducens paralysis, facial paralysis, anotia, micro-
`tia, renal malformations, congenital heart disease
`Neural-tube defects 66,67
`Fetal warfarin syndrome: skeletal defects (nasal hypoplasia and stippled epiphyses), limb hy-
`poplasia (particularly in distal digits), low birth weight (⬍10th percentile), hearing loss,
`and ophthalmic anomalies 69; CNS defects with exposure after first trimester; dorsal mid-
`line dysplasia (agenesis of corpus callosum and Dandy–Walker malformations) or ventral
`midline dysplasia (optic atrophy) 6
`
`Angiotensin-con-
`verting–enzyme
`inhibitors
`Carbamazepine
`
`Cocaine
`
`Ethanol
`
`Cleft palate, dilated cerebral ventricles,
`and growth retardation in mice 39
`Dose-dependent decrease in uterine
`blood flow, fetal hypoxemia, hyper-
`tension, and tachycardia in sheep41;
`reduced fetal weight, fetal edema, and
`increased resorption in rats and mice42
`Microcephaly, growth deficiency, and
`limb anomalies in dogs, chickens,
`and mice46-48
`
`Isotretinoin
`
`CNS, head, limb, and cardiovascular
`defects in rats and rabbits29
`
`Lithium
`Methyl mercury
`
`Phenytoin
`
`Heart defects in rats51
`CNS abnormalities in rats 54; growth re-
`tardation, motor disturbances, mi-
`croencephaly, and brain lesions in
`rhesus monkeys55
`Cleft palate, micromelia, renal defects,
`and hydrocephalus in rabbits, mice,
`and rats57-59
`
`Thalidomide†
`
`Valproic acid
`Warfarin†
`
`Limb-shortening defects in rabbits
`(most sensitive species)62
`Exencephaly in hamsters and mice 64,65
`Maxillonasal hypoplasia and skeletal
`anomalies in rats68
`
`*CNS denotes central nervous system.
`†Initial studies in animals failed to show teratogenicity; hence, documentation in humans preceded that in animals.
`
`Such studies were useful in establishing the safety
`and risk of Bendectin, isotretinoin, fluoxetine, and
`acyclovir.
`30,86
`Because most studies of teratogenic risk are limit-
`ed in size, meta-analyses of studies of similar design
`are becoming more frequent. A detailed, stepwise
`methodologic approach to meta-analysis of terato-
`logic studies has been described.
` The appropriate
`24
`use of this approach depends to a large extent on es-
`tablishing sound a priori criteria for methodologic
`quality and ensuring the inclusion of data from all
`available studies, in order to obviate any publication
`bias against negative results.
`Long-term studies are increasingly important, be-
`cause it is becoming clear that the long-term effects
`of teratogenic drugs on neurobehavioral develop-
`
`ment can have a more devastating effect on children
`and their families than structural anomalies. To date,
`several drugs have been shown to affect brain devel-
`opment, including carbamazepine, isotretinoin, phen-
`ytoin, valproic acid, and warfarin (Table 1). Carba-
`mazepine and valproic acid may cause cognitive
`brain dysfunction as part of the neural-tube defects
`they induce. Originally, isotretinoin was found to
`cause structural abnormalities that affected brain de-
`velopment, but recent studies have suggested that
`even phenotypically normal children may have ab-
`normal neurodevelopment.
` Warfarin was initially
`87
`associated with chondrodysplasia punctata and men-
`tal retardation and has subsequently been found to
`cause the Dandy–Walker brain malformation in an
`estimated 1 to 2 percent of exposed fetuses.
`6,69
`
`1132
`
`ⴢ
`
`April 16, 1998
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Page 5 of 10
`
`

`
`DRUG THERAPY
`
`Common Methodologic Issues
`No single approach can definitively establish the
`safety or risk of drugs, because of several underlying
`difficulties.
`
`Sample Size
`Most congenital malformations occur rarely, and
`many teratogens, even when known to be associated
`with an increased risk of a given malformation, do
`not affect the great majority of exposed fetuses. In
`fact, very few drugs increase the total malformation
`rate by a factor of more than two (isotretinoin and
`thalidomide are two such drugs). If, for example,
`the risk of major malformations in a given popu-
`lation is 3 percent, then at least 220 pregnancies
`with the specific exposure and a similar number of
`control pregnancies will be required to show a risk
`that is increased by a factor of 2.5, with a power of
`80 percent.
`
`Effect of Maternal Diseases
`Apart from drug therapy, many medical condi-
`tions themselves increase fetal risks. For example,
`pregnant women with hypertension or cancer are
`more likely to have infants with intrauterine growth
`retardation, and pregnant women with epilepsy or
`diabetes mellitus are more likely to have infants with
`malformations.88 Therefore, any attempt to establish
`the role of fetal exposure to drugs must also address
`the contributing and confounding risk of the under-
`lying maternal illness.
`
`Recall Bias in Retrospective Studies
`There is ample evidence of partial memory and
`bias in the way women recall the drugs they took
`during pregnancy. For example, women treated with
`a prescribed drug for a chronic illness tend to recall
`their treatment better than women who took an
`over-the-counter drug.89 Women who have given
`birth to malformed children may be more likely to
`remember the course of their pregnancies, in the ef-
`fort to understand what went wrong, than women
`who have given birth to healthy children, thus giv-
`ing rise to false positive associations. The initial sug-
`gestions that benzodiazepines, spermicides, and Ben-
`dectin, for example, were teratogenic were based on
`retrospective case–control studies subsequently re-
`futed by other, larger studies (Table 2).
`With improved epidemiologic methods, the reli-
`ability of the case–control design has improved. For
`example, recruiting mothers of infants with a differ-
`ent major malformation as controls may eliminate or
`at least reduce the problem of differential maternal
`recall. In a recent study, this approach was used to
`document the effect of the mothers’ knowledge of
`the study hypothesis (that folic acid deficiency caus-
`es spina bifida) on the information they reported.90
`
`Nonrandomized Observational Studies
`With prospective observational studies, the treat-
`ment decisions have not been made by the investi-
`gators collecting the data. As a result, the indica-
`tions for treatment and concurrent exposures are not
`standardized. Therefore, in comparisons of treated
`and untreated pregnant women or pregnant women
`who received two different drugs, preexisting con-
`founding factors are not randomly distributed be-
`tween the two groups. For example, in comparing
`the outcome of pregnancy in women who received
`carbamazepine and women who received phenytoin,
`one must address the issue of whether the two
`groups of women had the same type and severity of
`seizure disorder.
`Observational studies of neurobehavioral develop-
`ment require longer follow-up than observational
`studies of other abnormalities, and interpretation of
`the results is often complicated by numerous con-
`founding factors. Maternal and paternal IQ, socio-
`economic status, and educational levels all affect
`cognitive development in children.91 Any attempt to
`address the developmental effects of drugs without
`controlling for these factors is likely to be futile.
`
`Voluntary Reporting
`The information received by drug manufacturers is
`often a mix of prospective and retrospective case re-
`ports. The quality of the information about exposure
`is usually poor, and outcome data are sparse because
`of high rates of loss to follow-up. Most important,
`women and health professionals who contact manu-
`facturers are likely to report adverse fetal outcomes,
`not uneventful ones. For example, the pivotal study
`that described retinoid embryopathy contained two
`parts: prospectively collected data from a study co-
`hort, with a malformation rate of 36 percent, and data
`from voluntary retrospective reporting to the manu-
`facturer, with a malformation rate of 80 percent.30
`
`Meta-Analyses
`A common concern regarding the use of meta-
`analysis is the inevitable combination of data from
`studies that are not equivalent in terms of quality and
`methods. In addition, there is the concern that neg-
`ative studies (i.e., those that do not reject the null
`hypothesis) are less likely to be published than posi-
`tive studies and that an overall positive association
`may therefore merely reflect unbalanced reporting.
`
`COUNSELING WOMEN
`ABOUT TERATOGENIC RISKS
`In one study, women exposed to nonteratogenic
`drugs who sought counseling estimated, on average,
`that they had a 25 percent risk of major malforma-
`tions, which is in the range of the teratogenic risk as-
`sociated with thalidomide.2 After counseling, this es-
`
`Volume 338 Number 16
`
`ⴢ 1133
`
`Downloaded from www.nejm.org on March 4, 2007 . Copyright © 1998 Massachusetts Medical Society. All rights reserved.
`
`Page 6 of 10
`
`

`
`The New England Journal of Medicine
`
`timate was substantially reduced, thereby preventing
`numerous terminations of otherwise wanted preg-
`nancies.2,3 The same women correctly estimated the
`risk of major malformations in the general popula-
`tion (5 percent), indicating that the high risk they
`assigned to their own pregnancies was not due to a
`misunderstanding of the concept of base-line risk.
`What are the sources of this misperception? Nu-
`merous lay publications misinform women by as-
`signing risks to drugs not known to be teratogenic
`in humans.2,3 Women often report that their physi-
`cians have encouraged them to terminate otherwise
`wanted pregnancies just to be on the safe side, sug-
`gesting that many physicians are unfamiliar with the
`current literature on the safety of drugs taken during
`pregnancy.
`Physicians counseling women who are pregnant
`or are planning a pregnancy should make sure that
`they understand clearly the nature and magnitude of
`a risk associated with a drug. Women’s attitudes
`toward voluntary abortion differ. In addition, the
`same information about the nature and magnitude
`of a teratogenic risk may prompt different decisions
`by different women, according to the clinical situa-
`tion and specific circumstances. For example, wom-
`en with epilepsy that has been treated effectively
`with phenytoin since their childhood may be glad to
`hear that although the drug is teratogenic, the over-
`all risk of malformations is not high.61 In contrast,
`women who have been treated with phenytoin for a
`single grand mal seizure and who have normal chil-
`dren born before phenytoin was prescribed may find
`it unacceptable to continue an unplanned pregnancy
`after learning about the higher-than-normal risk of
`adverse effects.
`During counseling, it is important to ensure that
`a woman understands the concept of base-line

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket