CHAPTER 57

Psychopharmacology During Pregnancy and Lactation

Shona L. Ray-Griffith, M.D.

D. Jeffrey Newport, M.D.

Zachary N. Stowe, M.D.

The management of mental illness during pregnancy and lactation is a complicated clinical challenge encompassing two concomitant medical conditions (i.e., pregnancy and a psychiatric disorder) that requires consideration of the welfare of at least two patients (i.e., mother and child). Various nonpharmacological treatment options for maternal mental illnesses during the antepartum and postpartum periods are beyond the scope of this chapter. Viable treatment options must be available, accessible, affordable, and effective for the illness, and in many cases, this may involve pharmacotherapy. Unfortunately, definitive treatment guidelines for perinatal psychotropic medications remain unavailable. In the two decades since the initial iteration of this chapter, there has been an enormous expansion in the reproductive safety literature focusing on psychotropic medications.

In this chapter, we posit that minimizing infant exposure to the purported risks of both maternal mental illness and psychotropic medications is the preeminent clinical objective. Investigations and reviews focusing on reproductive safety of psychotropic medications typically provide only cursory information about the effect of illness, rely on maternal self-report to address concomitant fetal exposures, and use extensive statistical interrogation that may not have meaningful clinical import.

Space limitations preclude a comprehensive review of each class of psychotropic medications; rather, we review the available data with an emphasis on providing the clinician with the requisite components to interpret and apply medication reproductive safety data in the clinical decision. Also included are data regarding pharmacokinetic alterations during pregnancy and lactation and the potential clinical relevance of these data for psychotropic dosage management. The chapter concludes with a series of potential modifications of previously suggested treatment guidelines to optimize clinical decisions and a list of resources.

Minimizing Exposure of Offspring: Primary Therapeutic Objective

The clinical management of any medical condition during pregnancy and lactation must consider the reproductive safety of available therapies, the likelihood of illness recurrence without continued treatment, and the potential effect of untreated maternal illness. Minor ailments, such as headaches and nausea, are routinely treated during pregnancy with medications that often have limited reproductive safety data. Recent data indicate that 90% of pregnant women take one medication and that more than 50% take four or more medications (Ayad and Costantine 2015; Mitchell et al. 2011), confounding the ability to isolate the effect of an individual medication. Women with mental illness are frequently encouraged to discontinue psychotropic medication during pregnancy, and pregnancy is associated with high rates of both antidepressant discontinuation (Petersen et al. 2011) and nonadherence to prescribed psychiatric medications (Lupattelli et al. 2015). The desire to avoid offspring exposure to psychotropic medication is laudable; however, recommendations to discontinue medication are often made with limited knowledge of the potential risk of recurrent illness and of the effect of maternal illness on obstetrical and child outcomes.

An assessment of the risks of offspring exposure to maternal psychiatric illness must consider both the likelihood that an episode of illness will occur and the evidence that the illness may be harmful to the child. The clinician can estimate a patient’s likelihood of experiencing illness recurrence or exacerbation by carefully synthesizing prevalence data from epidemiological studies with evidence from the patient’s own history. Patients with frequent episodes of psychiatric illness, a declining course, or a history of perinatal illness are more likely to become ill in the current pregnancy or the postpartum period.

Investigations of the incidence and course of maternal mental illness have found no evidence that mental illness lies quiescent during pregnancy and considerable evidence that the postpartum period entails heightened vulnerability. The incidence of depression during the perinatal period is comparable to that in other populations (Buesching et al. 1986; Cutrona 1983; Kumar and Robson 1984; Manly et al. 1982; O’Hara et al. 1982; Watson et al. 1984). Two large studies, collectively comprising 122,400 women, found a 14%–20% incidence of prenatal major depressive disorder (MDD) (Marcus et al. 2003; Oberlander et al. 2006). In fact, 11% of women presenting for evaluation of postpartum depression report symptom onset before delivery (Stowe et al. 2005). Discontinuation of treatment during pregnancy in women with histories of MDD or bipolar disorder increases the risk of episodes during pregnancy (Cohen et al. 2006; Newport et al. 2008c; Viguera et al. 2007b). However, one group failed to identify an increase in depression with antidepressant discontinuation (Yonkers et al. 2011).

Psychotic disorders also may worsen during pregnancy (Glaze et al. 1991; McNeil et al. 1984a, 1984b). The course of obsessive-compulsive disorder (OCD) appears to be variable during pregnancy, with 14%–33% of patients experiencing exacerbation (Guglielmi et al. 2014; Jenike et al. 1990; Williams and Koran 1997). Women with OCD who continued pharmacotherapy throughout the perinatal period did not have a significant change in symptoms (House et al. 2016). Panic disorder during gestation is also variable, with 19% of patients experiencing more frequent panic attacks and 30% having less frequent attacks (Hertzberg and Wahlbeck 1999; Wisner et al. 1996a). Finally, obstetrical trauma and pregnancy loss can precipitate posttraumatic stress disorder (PTSD) (Allen 1998; Engelhard et al. 2001; Fones 1996), with PTSD symptoms following stillbirth persisting into the next pregnancy (Turton et al. 2001).

Postpartum mental illness has been documented for millennia and is substantiated by modern research. Early studies reported that psychiatric hospitalization rates increased during the first postpartum month (Kendler et al. 1993) and noted that up to 12.5% of all psychiatric admissions for women occurred during the first postpartum year (Duffy 1983). Postpartum depression affects 10%–22% of adult mothers and up to 26% of adolescent mothers (Stowe and Nemeroff 1995; Troutman and Cutrona 1990). Women with bipolar disorder also face considerable postpartum risks (Kendell et al. 1987; Targum et al. 1979). Postpartum OCD, often manifested by violence and contamination obsessions, may affect 9%–16% of new mothers (Miller et al. 2013; Zambaldi et al. 2009), with one-third experiencing new-onset OCD symptoms during the postpartum period (Miller et al. 2013). Nearly one-half of women with preexisting OCD report an exacerbation of symptoms during the postnatal period (Guglielmi et al. 2014). Thankfully, postpartum psychosis, the most severe postpartum syndrome, is a rare condition; however, its prevalence is at least 100-fold higher among women with bipolar disorder than among those with other affective or psychotic disorders (Brockington et al. 1982; Kendell et al. 1987).

Effect of Maternal Psychiatric Disorders

Not only the likelihood but also the potential effect of maternal mental illness on maternal and child well-being must be considered. Most studies focusing on obstetrical and developmental outcomes have focused on the effect of maternal depression, anxiety, and stress. Often these terms are used interchangeably, with most previous investigations focusing on maternal symptoms rather than diagnosis. Given the high rate of comorbidity of mood and anxiety disorders in women, the distinction between maternal depression and maternal anxiety may not have clinical import. The effect of maternal unipolar versus bipolar depression during gestation remains unexplored. Maternal depression during pregnancy has been associated with slower fetal growth (Hedegaard et al. 1996; Schell 1981; Uguz et al. 2013); increased risk of preterm delivery and other obstetrical complications (Korebrits et al. 1998; Liu et al. 2016; Oberlander et al. 2006; Orr and Miller 1995; Perkin et al. 1993; Steer et al. 1992; Uguz et al. 2013; Venkatesh et al. 2016); and long-standing cognitive, behavioral, and emotional changes in the offspring (Luoma et al. 2001, 2004; Meijer 1985; Nulman et al. 2002; O’Connor et al. 2003; Søndergaard et al. 2003; Stott 1973). Similarly, prenatal panic disorder is associated with higher rates of preterm birth and low birth weight (Uguz et al. 2013), and maternal anxiety is associated with higher rates of attentional problems (Van den Bergh and Marcoen 2004). Depressed pregnant women are less compliant with prenatal vitamins and obstetrical care; receive poorer nutrition; and have greater use of prescription opiates, hypnotics, alcohol, tobacco, and illicit substances (Newport et al. 2012; Zuckerman et al. 1989). Finally, depressed pregnant women, and those with PTSD, often experience suicidal thoughts (Newport et al. 2007b; Smith et al. 2006) and may engage in suicidal behavior.

Postpartum depression also carries deleterious consequences for infant development. As early as 3 months, infants of depressed mothers show less facial expression, less head orientation, less crying, and more fussiness compared with infants of nondepressed mothers (Martinez et al. 1996). As they age, the children of depressed mothers show ineffective emotional regulation (Downey and Coyne 1990), delayed motor development (Galler et al. 2000), poor interpersonal skills (Jameson et al. 1997), lower self-esteem (Downey and Coyne 1990), increased fear and anxiety (Lyons-Ruth et al. 2000), more aggression (Jameson et al. 1997), and more insecure and disorganized attachment behaviors (Martins and Gaffan 2000). Children of depressed mothers are ultimately more likely to experience emotional instability, to have behavior problems and suicidal behavior, and to require psychiatric treatment (Lyons-Ruth et al. 2000; Weissman et al. 1984).

Antepartum exacerbation of schizophrenia and other psychotic illnesses also warrants concern. Women with schizophrenia have a higher prevalence of substance abuse during pregnancy (Miller and Finnerty 1996; Taylor et al. 2015) and may have bizarre ideas about contraception, pregnancy, and child rearing that complicate their perinatal course (McEvoy et al. 1983; Riordan et al. 1999). Maternal schizophrenia has been associated with elevated rates of obstetrical complications (Bennedsen et al. 2001; Miller and Finnerty 1996) and fetal and neonatal death (Rieder et al. 1975).

The clinical data regarding the obstetrical and developmental consequences of maternal mental illness are supported by an extensive line of laboratory animal research. Preclinical studies across a variety of species indicate that stress during pregnancy and the early postpartum period adversely affects offspring growth, learning ability, and postnatal development, producing a range of biobehavioral aberrations that may persist into adulthood (for a review, see Newport et al. 2002).

Risks of Antepartum and Postpartum Exposure to Psychotropic Medication

Clinical decisions during pregnancy and the postpartum period must consider the risks of fetal and neonatal medication exposure. These risks may be broadly classified as acute or developmental adverse effects (Table 57–1). Acute effects are typically immediately evident and are not dependent on the developmental window of exposure. Examples include drug toxicity, drug withdrawal, and drug–drug interactions. Developmental effects are, by definition, dependent on the developmental window of exposure and are often not evident until later. These effects include somatic teratogenesis (i.e., major and minor malformations) and neurobehavioral teratogenesis (i.e., alterations in brain development that affect the child’s subsequent behavior, cognitive abilities, and emotional regulation). The window of vulnerability to somatic teratogenesis is limited to the embryonic phase of development, but because central nervous system (CNS) development continues long after delivery, the fetus and breast-feeding infant are equally vulnerable to the theoretical risks of neurobehavioral teratogenesis.

TABLE 57–1. Potential risks of medication exposure

Acute

Developmental

Pregnancy

Neonatal toxicity

Somatic teratogenesis

Neonatal withdrawal

Neurobehavioral teratogenesis

Drug–drug interactions

Lactation

Infant toxicity

Neurobehavioral teratogenesis

Drug–drug interactions

A decision to use psychotropic medication during pregnancy and/or lactation will carry complicated clinical, ethical, and potentially legal consequences. Although a rapidly expanding base of reproductive safety data has begun to address many of these concerns, review of the current literature reveals numerous methodological problems. The most glaring deficiencies are a frequent lack of appropriate control groups and an overreliance on retrospective data collection—deficiencies shown to introduce systematic biases that potentially lead to overestimation of the effect of psychotropic exposure (Newport et al. 2008a). In particular, most studies report outcomes of “depressed–treated women compared with nondepressed–untreated women,” eliminating any opportunity to disentangle treatment and illness effects on outcomes (McDonagh et al. 2014). Despite these limitations, ethical considerations preclude implementation of randomized controlled trials to evaluate psychotropic efficacy and safety during pregnancy and lactation. Therefore, the reproductive safety database for psychotropic medications is composed of a diverse conglomeration of case reports, case series by pharmaceutical companies and academic centers, birth registries, retrospective surveys, reports from teratology or poison control centers, clinical and preclinical pharmacokinetic investigations, review articles summarizing data from these sources, and the U.S. Food and Drug Administration (FDA) medication safety rating systems (Table 57–2).

TABLE 57–2. U.S. Food and Drug Administration use-in-pregnancy ratings (prior to 2015)

Category

Interpretation

A

Controlled studies show no risk: Adequate, well-controlled studies in pregnant women have failed to demonstrate risk to the fetus.

B

No evidence of risk in humans: Either animal findings show risk, but human findings do not; or, if no adequate human studies have been done, animal findings are negative.

C

Risk cannot be ruled out: Human studies are lacking, and animal studies are either positive for fetal risk or lacking as well. However, potential benefits may justify the potential risk.

D

Positive evidence of risk: Investigational or postmarketing data show risk to the fetus. Nevertheless, potential benefits may outweigh risks.

X

Contraindicated in pregnancy: Studies in animals or humans, or investigational or postmarketing reports, have shown fetal risk that clearly outweighs any possible benefit to the patient.

Source. Physicians’ Desk Reference 2007.

In addition to the available reproductive safety data, the patient’s clinical history is important in treatment selection. If the rationale for psychotropic therapy during pregnancy is to protect the mother and the child from the harmful sequelae of maternal mental illness, then a history of treatment response or nonresponse is critical. Regardless of the favorability of its reproductive safety profile, a medication that has been ineffective for or poorly tolerated by a particular patient is of little value during the reproductive period.

FDA Reproductive Safety Ratings

On June 30, 2015, the FDA’s new Pregnancy and Lactation Labeling Rule became effective, with a plan for staggered implementation over the following 3–5 years. The new labeling rule eliminates the pregnancy letter category previously used (see Table 57–2) and provides the following information for the use of medications in pregnancy and delivery when available: 1) pregnancy exposure registry, 2) narrative risk summary, 3) clinical considerations, and 4) data description of supporting evidence. Specifically, the clinical considerations are as follows: disease-associated maternal and embryo/fetal risk, dosage adjustment, maternal adverse reactions, fetal/neonatal adverse reactions, and labor or delivery. Two new sections also summarize evidence for lactation and evidence for males and females of reproductive potential. The labeling will be updated as evidence emerges but is required only for medications receiving FDA approval after June 30, 2001. Medications approved prior to this date are strongly encouraged to comply with the new recommendations but are not required to do so.

The result of these changes is two unintegrated systems for reproductive safety ratings that present a challenge for clinicians. Similarly, the rate of accrual for reproductive safety information in humans is variable and all obtained postmarketing. The new safety ratings will arguably have greater clinical import, because the older system often provided more favorable ratings to newer medications with very limited information and was often overvalued in clinical decisions. For example, a clinician may be tempted to use buspirone (Category B) over clonazepam (Category D) for panic disorder despite the very limited information underlying buspirone’s preferable pregnancy rating and its lack of efficacy for panic disorder. Similarly, lurasidone (Category B) may be used in lieu of lithium (Category D) for bipolar depression despite the virtual absence of human data on lurasidone’s safety during gestation and the lack of published clinical experience in the antepartum and postpartum periods.

Dosage Management: Perinatal Pharmacokinetics

Prescribing the minimal effective dosage is especially important during pregnancy and lactation. On learning that a patient is pregnant, clinicians and patients often instinctively lower psychotropic dosages in an effort to reduce fetal exposure, but indiscriminate dosage reduction may increase the vulnerability to relapse. If the therapeutic objective is to eliminate the child’s exposure to maternal illness while minimizing the child’s psychotropic exposure, then dosage management must be informed by an understanding of the factors governing the alterations in drug disposition across gestation, the placental passage, and excretion into breast milk.

Dosage adjustments may be required to maintain medication efficacy during pregnancy. For example, it may be necessary to increase the dosage of tricyclic antidepressants (TCAs) to approximately 1.6 times the preconception dosage to maintain therapeutic concentrations in late pregnancy (Altshuler et al. 1996; Wisner et al. 1993). Small studies examining selective serotonin reuptake inhibitors (SSRIs) have reported clearance changes over the course of pregnancy and the necessity for dosage adjustments in response to increased maternal symptoms (Freeman et al. 2008; Hostetter et al. 2000; Sit et al. 2008). However, the pattern of dosage adjustment during gestation is not uniform for all medications. For example, among anticonvulsant and mood-stabilizing medications, serum concentrations of lamotrigine (Ohman et al. 2000; Pennell et al. 2004; Polepally et al. 2014; Tran et al. 2002) and valproate (Otani 1985; Philbert et al. 1985) decline steadily across gestation, whereas carbamazepine concentrations (Bardy et al. 1982b; Battino et al. 1982; Bologa et al. 1991; Dam et al. 1979; Lander et al. 1981; Omtzigt et al. 1993; Otani 1985; Tomson et al. 1994; Yerby et al. 1985) undergo smaller changes that are primarily evident only in late pregnancy. These alterations in drug concentrations and dosing requirements likely result from effects of the physiological changes of pregnancy on the pharmacokinetics (Boobis and Lewis 1983; Frederiksen 2001; Little 1999; Wyska and Jusko 2001) and possibly the pharmacodynamics (Wyska and Jusko 2001) of psychotropic medications. Ultimately, an improved understanding of the factors governing perinatal pharmacokinetic and pharmacodynamic alterations may enable the development of personalized dosing strategies during gestation.

The level of fetal psychotropic exposure is another key consideration. All psychotropics studied to date cross the human placenta; yet there are significant differences in placental passage rates. Bidirectional placental transfer is mediated primarily by simple diffusion, and determinants of the transplacental diffusion rate include molecular weight, lipid solubility, degree of ionization, and protein-binding affinity (Audus 1999; W.M. Moore et al. 1966; Pacifici and Nottoli 1995). In addition, placental P-glycoprotein (P-gp), which actively transports substrates from the fetal to the maternal circulation, is likely a key determinant of fetal psychotropic exposure. Consequently, medications with greater affinity for P-gp should be associated with lower fetal-to-maternal medication ratios, which, in fact, has been confirmed in an investigation of antipsychotic placental passage (Newport et al. 2007a). Furthermore, clarification of the factors governing placental passage may ultimately contribute to the development of psychotropic agents with minimal rates of placental transfer (Wang et al. 2007).

Fetal plasma concentrations are not, however, the ultimate measure of functional psychotropic exposure and may even underestimate the more critical measure of fetal brain concentration. Certain physiological attributes of the human fetus, including high cardiac output, increased blood–brain barrier permeability, low plasma protein concentrations and plasma protein binding affinities, and low hepatic enzyme activity (Bertossi et al. 1999; Morgan 1997; Oesterheld 1998), may produce higher-than-anticipated fetal CNS concentrations of psychotropic medications than might be anticipated from circulating levels. Preclinical investigations from our group have found that transplacental passage of psychotropic drugs results in high levels in fetal CNS tissues and significant binding at neurotransmitter receptor and transporter sites (Capello et al. 2011).

Similar considerations apply when endeavoring to minimize the psychotropic exposure of nursing infants. Because the neonate has relatively low hepatic enzyme activity (Warner 1986) and low glomerular filtration and tubular secretion rates (Welch and Findlay 1981), psychotropic exposure may be higher than anticipated for breast-fed infants. A model to predict rates of breast-milk excretion from characteristics of the molecular structure of candidate medications has been proposed (Agatonovic-Kustrin et al. 2002). Such models may allow clinicians to estimate a nursing infant’s exposure without subjecting the infant to invasive procedures.

Antidepressants

The use of antidepressant medications in pregnancy has been extensively reviewed (McDonagh et al. 2014; Yonkers et al. 2014), and multiple meta-analyses have found no association between antidepressant use in pregnancy and congenital malformations and/or major malformations (Einarson and Einarson 2005; Grigoriadis et al. 2013). We include an overview of the individual categories of antidepressants in this section.

Selective Serotonin Reuptake Inhibitors

Reproductive safety data on the SSRIs have rapidly accrued over the past two decades. A recent meta-analysis investigating SSRIs found no overall association with birth defects (Reefhuis et al. 2015). Despite overall reassuring data, some concerns with specific SSRIs have emerged. Reefhuis et al. (2015) found an increased risk of anencephaly, atrial septal defects, right ventricular outflow tract obstruction, gastroschisis, and omphalocele with paroxetine and an increased risk of right ventricular outflow track obstruction defects and craniosynostosis with fluoxetine. Analysis of a managed care database identified a higher odds ratio (OR) for cardiovascular malformations with exposure to paroxetine compared with other antidepressants (GlaxoSmithKline 2005). Three large case–control studies of SSRI exposures (Alwan et al. 2007; Bérard et al. 2007; Louik et al. 2007) reported largely reassuring results, although with some concerns. The first (Alwan et al. 2007) reported small increases in the risk of three uncommon malformations—anencephaly (OR=2.4), craniosynostosis (OR=2.5), and omphalocele (OR=2.8) associated with use of fluoxetine, sertraline, or paroxetine. The second study (Louik et al. 2007) reported an increased risk of omphalocele (OR=5.7) and septal defects (OR=2.0) with sertraline exposure and of right ventricular outflow tract obstruction defects (OR=3.3) with paroxetine exposure. The third study (Bérard et al. 2007) found no evidence of increased risk of cardiovascular or other malformations with SSRI use, unless women were taking paroxetine at daily dosages of 25 mg or greater, in which case their infants were at an increased risk of cardiovascular (OR=3.1) and overall malformations (OR=2.2). In the Medicaid Analytic eXtract (MAX) database, Huybrechts et al. (2014a) found an overall adjusted risk of 1.06 (95% confidence interval [CI]=0.93–1.22) for any cardiac defect with use of an SSRI in the first trimester; a risk of 1.07 (95% CI=0.59–1.93) for a right ventricular outflow tract obstruction with use of paroxetine; and a risk of 1.04 (95% CI=0.76–4.41) for a ventricular septal defect with use of sertraline.

A handful of studies have systematically assessed child development after prenatal exposure to antidepressants. Two studies (Nulman et al. 1997a, 2002) assessed children (ages 15–86 months) who had been prenatally exposed to fluoxetine (n=90) or TCAs (n=126), collectively comparing them with nonexposed children (n=120). No differences were observed with respect to global cognitive, psychomotor, or language development. The same research group compared children exposed to SSRIs in utero with children of mothers without depression or SSRI use and found no difference in IQ or behavior (Nulman et al. 2012). A similar study that compared children (ages 6–40 months) with prenatal SSRI exposure (n=31) and children without antidepressant exposure (n=13) observed no differences in global cognition but reported lower psychomotor scores for the SSRI-exposed children (Casper et al. 2003).

Unfortunately, limitations of these studies render their implications speculative at best. Children were not age-matched in any of these studies, and the predictive validity of measured indices across child developmental stages has not been established (Black and Matula 2000). In addition, the Casper et al. (2003) study was confounded by the fact that 29% of the participants were enrolled after delivery, which could have resulted in an overrepresentation of children with developmental abnormalities.

A study of 69 (46 SSRI-exposed; 23 nonexposed) children that eliminated the age confound by examining the children at two fixed time points—ages 2 months and 8 months—reported no differences between the exposed and the nonexposed children in cognitive or motor development (Oberlander et al. 2004). Finally, an assessment of 4-year-old children found no evidence that prenatal SSRI exposure affected externalizing or attentional behaviors (Oberlander et al. 2007). It is noteworthy that several of these developmental studies involved a significant overlap in the cohort of children, thereby limiting further the cumulative sample size of extant follow-up studies.

Multiple studies have examined the association between maternal antidepressant use and autism spectrum disorders in offspring. The results were mixed, with four studies finding an association between antidepressant exposure and autism (Boukhris et al. 2016; Croen et al. 2011; El Marroun et al. 2014; Gidaya et al. 2014) and six finding no such association (Castro et al. 2016; Clements et al. 2015; Harrington et al. 2014; Hviid et al. 2013; Rai et al. 2013; Sørensen et al. 2013). In addition, close inspection reveals evidence of probable detection bias in at least some of these studies, with nonminority children of well-educated mothers overrepresented in the autism case subjects (Croen et al. 2011). These discordant and arguably flawed data preclude definitive conclusions regarding any autism risk attributable to prenatal antidepressant exposure.

Data regarding the effect of prenatal antidepressant exposure on rates of miscarriage, preterm delivery, and low birth weight are decidedly mixed. Some investigators have reported an association between antidepressant exposure and such outcomes (Chambers et al. 1996; Chun-Fai-Chan et al. 2005; Oberlander et al. 2006; Pastuszak et al. 1993; Ross et al. 2013; Simon et al. 2002), whereas others have not (Einarson et al. 2001, 2003; Kulin et al. 1998; Sivojelezova et al. 2005; Venkatesh et al. 2016). Multiple recent meta-analyses found an association between prenatal antidepressant exposure and preterm delivery (Huang et al. 2014; Huybrechts et al. 2014b; Ross et al. 2013) and low birth weight (Huang et al. 2014; Ross et al. 2013). Interpretation is complicated by other reports that link prenatal maternal stress and/or depression with prematurity and low birth weight (Orr et al. 2002; Steer et al. 1992). As a result, no definitive conclusions can be drawn as to whether antidepressant use during gestation conveys an adverse effect on fetal growth or the timing of parturition.

Finally, concerns have been expressed regarding neonatal SSRI syndromes, typically manifested by transient symptoms including respiratory difficulty and tremulousness (Moses-Kolko et al. 2005). Most controlled prospective studies suggest that SSRI exposure is associated with poor neonatal adaptation (Chambers et al. 1996; Costei et al. 2002; Källén 2004; Laine et al. 2003; Oberlander et al. 2004, 2006; Sivojelezova et al. 2005; Zeskind and Stephens 2004), although one study found no such association (Maschi et al. 2008). Closer scrutiny of these reports identified a cadre of methodological shortcomings. Limited effort was made to mask those evaluating the neonates as to exposure status, only one study (Oberlander et al. 2006) controlled for the effect of maternal mental illness, and key confounding factors such as gestational age at delivery and maternal use of other medications and/or habit-forming substances were either ignored altogether or controlled only in a rudimentary manner.

A case–control study comparing the exposures of neonates who “required observation” with those of “healthy” neonates (Misri et al. 2004) underscores the importance of controlling for confounding factors. In this study of antidepressant-exposed neonates (n=46) born to mothers with MDD, the mothers of infants who required observation had more severe symptoms of depression and anxiety, were more likely to have a comorbid anxiety disorder, and were exposed to higher dosages of clonazepam.

Debate has also focused on a more serious neonatal concern—a possible connection between late-pregnancy SSRI exposure and persistent pulmonary hypertension of the neonate (PPHN). A case–control study (Chambers et al. 2006) of 1,213 neonates (377 with PPHN) reported data supporting an association between SSRI exposure and PPHN (OR=6.1). However, a similar study of 1,104 SSRI-exposed neonates and 1,104 matched control subjects found no association (Andrade et al. 2009). A case–control study (Källén and Olausson 2008) of more than 831,000 neonates (of whom 506 were diagnosed with PPHN) from the Swedish Medical Birth Register observed a more modest association (OR=2.9). The overall rate of PPHN in this population-wide study was approximately 1 in 2,000, and the rate of PPHN with SSRI exposure was approximately 1 in 600. In the largest study to date, involving more than 3 million women, the adjusted odds of PPHN, after controlling for depression, was 1.10 (95% CI=0.77–1.35) for SSRI use and 1.14 (95% CI=0.74–1.74) for non-SSRI antidepressant use (Huybrechts et al. 2015). Finally, a study concluded that PPHN was associated not with SSRI exposure but rather with cesarean delivery prior to onset of labor (Wilson et al. 2011), underscoring the importance of controlling for all potential confounding factors. Overall, the SSRI-PPHN data are mixed, with a very low absolute rate of PPHN.

Pharmacokinetic data regarding the placental passage of SSRI antidepressants remain limited. However, existing studies have found that mean fetal-to-maternal ratios for numerous SSRIs and their active metabolites are uniformly less than 1.0, although considerable differences exist among agents (Hendrick et al. 2003; Rampono et al. 2009).

Published reports pertaining to SSRIs and lactation now encompass the largest data set for medications during breast feeding, including infant serum measures, breast milk concentrations, and pharmacokinetic studies of excretion. There are now published investigations for all of the SSRIs, including sertraline (Altshuler et al. 1995; Birnbaum et al. 1999; Dodd et al. 2000; Epperson et al. 1997, 2001; Hendrick et al. 2001a; Kristensen et al. 1998; Mammen et al. 1997; Stowe et al. 1997, 2003; Wisner et al. 1998), fluoxetine (Birnbaum et al. 1999; Burch and Wells 1992; Goldstein et al. 1997; Hendrick et al. 2001b; Kristensen et al. 1999; Lester et al. 1993; Suri et al. 2002; Taddio et al. 1996; Yoshida et al. 1998a), paroxetine (Birnbaum et al. 1999; Hendrick et al. 2001a; Ohman et al. 1999; Spigset et al. 1996; Stowe et al. 2000), escitalopram (Rampono et al. 2006), fluvoxamine (Hendrick et al. 2001a; Piontek et al. 2001; Wright et al. 1991), and citalopram (Heikkinen et al. 2002; Jensen et al. 1997; Schmidt et al. 2000; Spigset et al. 1997). Although infant follow-up data are limited, only a few isolated cases of adverse effects have been reported. Long-term neurobehavioral studies of infants exposed to SSRI antidepressants during lactation warrant continued examination. The pharmacokinetic profiles of breast milk excretion, including delineation of distribution gradients and time gradients, are best defined for sertraline (Stowe et al. 1997, 2003), paroxetine (Stowe et al. 2000), and fluoxetine (Suri et al. 2002). These studies indicate that quantitative infant SSRI exposure during lactation is considerably lower than transplacental exposure.

Other Antidepressants

Reproductive safety data are more limited for other classes of antidepressants, including bupropion, desvenlafaxine, duloxetine, mirtazapine, nefazodone, trazodone, venlafaxine, vilazodone, and vortioxetine. Prospective reports of first-trimester use of these agents have included 2,550 bupropion exposures producing 56 (2.2%) children with major malformations (Boshier et al. 2003; Briggs et al. 2005; Chun-Fai-Chan et al. 2005; Cole et al. 2007; GlaxoSmithKline 2005), 862 venlafaxine exposures with 91 (10.6%) major malformations (Einarson et al. 2001; GlaxoSmithKline 2005; Polen et al. 2013), 404 trazodone exposures with 10 (2.5%) major malformations (Briggs et al. 2005; GlaxoSmithKline 2005; McElhatton et al. 1996), 140 nefazodone exposures with 2 (1.4%) major malformations (GlaxoSmithKline 2005), 508 mirtazapine exposures with 14 (2.8%) major malformations (Djulus et al. 2006; GlaxoSmithKline 2005; Smit et al. 2015; Winterfeld et al. 2015), and a combined report of 121 nefazodone or trazodone exposures with 2 (1.7%) major malformations (Einarson et al. 2003). For duloxetine, there have been 165 exposures with 3 (1.8%) major malformations (Einarson et al. 2012). The only reported case of vilazodone use in pregnancy resulted in a full-term, healthy neonate (Morrison 2014). No published data are yet available for prenatal vortioxetine exposure.

A single study that followed an infant until 9 months after in utero exposure to duloxetine reported normal infant cognitive, language, motor, and psychomotor development (Bellantuono et al. 2013). Likewise, children exposed to venlafaxine in utero showed no differences in IQ or behavior compared with children of nondepressed mothers (Nulman et al. 2012).

A recent study found that prenatal exposure to serotonin-norepinephrine reuptake inhibitor antidepressants after the 20th week of gestation was associated with an increased risk of hypertensive disorders of pregnancy (OR=2.57; 95% CI=1.34–4.93), with an even higher (sixfold) increase among those receiving venlafaxine at dosages exceeding 187.5 mg/day (Newport et al. 2016).

Prenatal pharmacokinetic data are limited. High rates of placental transfer (i.e., umbilical cord concentrations in excess of maternal concentrations) have been reported for venlafaxine (Hendrick et al. 2003) and for its active metabolite desvenlafaxine (Hendrick et al. 2003; Rampono et al. 2009). Duloxetine has a single case report of a cord-to-maternal serum concentration ratio of 0.12, which suggests a low rate of placental transfer (Boyce et al. 2011).

Lactation data also remain limited. Best studied are venlafaxine/desvenlafaxine, for which three studies (Ilett et al. 1998; Newport et al. 2009; Rampono et al. 2011) encompassing 26 mother–infant nursing dyads have reported infant dosages that were 6.8%–8.1% of maternal dosages, which is within the notional 10% presumed safety level (Hendrick et al. 2003; Rampono et al. 2009). A single case report and a small case series of six women receiving duloxetine therapy during lactation described relative infant doses of less than 1% (Boyce et al. 2011; Lobo et al. 2008). The sole report of bupropion in nursing infants noted that the drug could not be detected in the plasma of a nursing infant (Briggs et al. 1993). Five studies totaling 55 neonates exposed to mirtazapine have reported no adverse outcomes and a low relative infant dose (Aichhorn et al. 2004; Klier et al. 2007; Kristensen et al. 2007; Smit et al. 2015; Tonn et al. 2009). No lactation data have been published for vilazodone or vortioxetine.

Tricyclic Antidepressants

Before the introduction of the SSRIs, TCAs were widely used during pregnancy and lactation. No clear association has been established between TCA exposure and congenital malformations. Early studies raised concerns about limb anomalies (Barson 1972; Elia et al. 1987; McBride 1972), but a meta-analysis by Altshuler et al. (1996) identified a congenital malformation incidence of only 3.14% (n=13) among 414 infants exposed to a TCA during the first trimester. A data review from the European Network of Teratology Information Services and the United Kingdom’s General Practice Research Database found similar rates of malformation after TCA exposure (McElhatton et al. 1996; Vasilakis-Scaramozza et al. 2013). Moreover, no adverse neurodevelopmental effects were reported in two studies involving 126 children with prenatal TCA exposure (Nulman et al. 1997a, 2002).

Few data exist regarding the acute effects of TCA exposure on fetal and neonatal well-being. There are case reports of fetal tachycardia and neonatal symptoms including tachypnea, tachycardia, cyanosis, irritability, hypertonia, clonus, and spasm (Eggermont 1973; ter Horst et al. 2012). A small (n=18) prospective study found no evidence of increased complications during labor and delivery but did report transient withdrawal symptoms among TCA-exposed neonates (Misri and Sivertz 1991).

TCAs have been widely used during lactation. The only adverse event reported to date is respiratory depression in a nursing infant exposed to doxepin, leading the authors to conclude that doxepin should be avoided but that most TCAs are safe for use during breast feeding (Matheson et al. 1985). This clinical finding is paralleled by pharmacokinetic data indicating that whereas all TCAs are excreted in breast milk, infant plasma concentrations are considerably higher for doxepin than for other TCAs (for a review, see Wisner et al. 1996b).

Monoamine Oxidase Inhibitors

Although the monoamine oxidase inhibitors (MAOIs) were introduced almost 50 years ago, reproductive safety data are sparse. The utility of MAOIs during pregnancy and lactation is severely limited by the potential for hypertensive crisis, which necessitates dietary constraints and avoidance of numerous medications that are commonly used during pregnancy (e.g., pseudoephedrine) or labor and delivery (e.g., meperidine).

Mood Stabilizers

Lithium

Early retrospective data suggested that lithium exposure was associated with a 400-fold increase in cardiac malformations—specifically, a defect of the tricuspid valve known as Ebstein’s anomaly (Nora et al. 1974; Weinstein and Goldfield 1975). However, a subsequent meta-analysis calculated the risk ratio for cardiac malformations as 1.2–7.7 and the risk ratio for overall congenital malformations as 1.5–3.0 (Cohen et al. 1994). Altshuler et al. (1996) estimated that the risk of Ebstein’s anomaly after prenatal lithium exposure rises from 1 in 20,000 to 1 in 1,000. A recent study indicated that lithium exposure in utero is associated with a higher rate of cardiovascular malformations but not of all major congenital malformations (Diav-Citrin et al. 2014). Additional studies (albeit small ones) also failed to confirm the early estimates regarding lithium’s teratogenic potential (Friedman and Polifka 2000; Jacobson et al. 1992; Källén and Tandberg 1983). Laboratory animal studies had indicated that neurobehavioral alterations also might be a concern for prenatal lithium exposure, but two studies of school-age children exposed to lithium during gestation found no evidence of adverse neurobehavioral sequelae (Schou 1976; van der Lugt et al. 2012).

The continued recommendation for prenatal assessment for fetal anomalies in women taking lithium includes a fetal echocardiogram between weeks 18 and 20 of gestation. In the event of unplanned conception during lithium therapy, the decision to continue or discontinue lithium should be informed by the severity and course of the patient’s illness and the time point in gestation when the exposure comes to attention. Discontinuing lithium therapy after cardiogenesis is complete, at approximately 9–11 weeks’ gestation, may be ill advised.

Lithium’s low therapeutic index raises concerns about acute perinatal toxicities. Lithium exposure later in gestation can result in fetal and neonatal cardiac arrhythmias (Wilson et al. 1983), hypoglycemia and nephrogenic diabetes insipidus (Mizrahi et al. 1979), thyroid dysfunction (Karlsson et al. 1975), polyhydramnios, premature delivery, and floppy infant syndrome (Llewellyn et al. 1998). Neonatal symptoms of lithium toxicity, including flaccidity, lethargy, and poor suck reflexes, may persist for more than 7 days (Woody et al. 1971).

In a pooled analysis of lithium placental passage and neonatal outcomes (Newport et al. 2005), we determined that 1) higher neonatal lithium concentrations were associated with significantly lower Apgar scores, longer hospital stays, and higher rates of CNS and neuromuscular complications; 2) umbilical cord (i.e., fetal) plasma concentrations were uniformly equivalent to maternal concentrations, suggesting that lithium rapidly equilibrates across the placenta; and 3) withholding lithium therapy for 24–48 hours prior to delivery resulted in a 0.28 mEq/L reduction in maternal (and presumably fetal) lithium concentrations, thereby likely improving neonatal outcomes.

The physiological alterations of pregnancy are of particular importance in the perinatal management of lithium. Changes in renal clearance over the course of pregnancy and the potential for abrupt volume changes during delivery as a result of copious diaphoresis and the loss of blood and amniotic fluid mandate careful monitoring of lithium levels during pregnancy and especially at delivery. Furthermore, nonsteroidal anti-inflammatory drugs, which inhibit renal clearance of lithium, should be avoided in mother and infant alike during the early postpartum period.

The existing database regarding lithium and lactation encompasses 25 mother–infant nursing dyads (Bogen et al. 2012; Fries 1970; Schou and Amdisen 1973; Skausig and Schou 1977; Sykes et al. 1976; Tunnessen and Hertz 1972; Viguera et al. 2007a; Weinstein and Goldfield 1969; Woody et al. 1971). Adverse events, including lethargy, hypotonia, hypothermia, cyanosis, electrocardiogram changes, poor feeding, slow growth, gross and fine motor delay, and elevated thyroid-stimulating hormone levels, were reported in five (16%) of these children (Bogen et al. 2012; Skausig and Schou 1977; Tunnessen and Hertz 1972; Viguera et al. 2007a; Woody et al. 1971), including one infant who developed frank lithium toxicity with a serum concentration of 1.4 mEq/L, which was double the maternal level (Skausig and Schou 1977). The American Academy of Pediatrics (American Academy of Pediatrics Committee on Drugs 2001) discourages the use of lithium during lactation. The largest pharmacokinetic study of lithium in lactation reported a milk-to-plasma ratio of 0.53 and an infant-to-maternal plasma ratio of 0.24 (Viguera et al. 2007a). In other studies, nursing infants have had lithium concentrations generally ranging from 5% to 65% of maternal levels (Bogen et al. 2012; Fries 1970; Kirksey and Groziak 1984; Schou and Amdisen 1973; Sykes et al. 1976; Tunnessen and Hertz 1972; Weinstein and Goldfield 1969), excluding the lone infant whose serum concentration was 200% of the maternal concentration (Skausig and Schou 1977). Because dehydration can increase vulnerability to lithium toxicity, the hydration status of nursing infants of mothers taking lithium should be carefully monitored (Llewellyn et al. 1998).

Valproate (Valproic Acid)

Prenatal exposure to valproate has been associated with numerous congenital malformations (rate of 6.7% for overall congenital malformations; Campbell et al. 2014), including neural tube defects (Bjerkedal et al. 1982; Centers for Disease Control 1992; Jäger-Roman et al. 1986; Lindhout and Schmidt 1986), craniofacial anomalies (Assencio-Ferreira et al. 2001; Lajeunie et al. 1998, 2001; Paulson and Paulson 1981), limb abnormalities (Rodríguez-Pinilla et al. 2000), cardiovascular anomalies (Dalens et al. 1980; Koch et al. 1983; Sodhi et al. 2001; Veiby et al. 2014), and hypospadias (Veiby et al. 2014). Valproate exposure prior to neural tube closure, during the fourth week of gestation, confers a 1%–2% risk of spina bifida, which is 10–20 times greater than the risk in the general population (Bjerkedal et al. 1982; Centers for Disease Control 1992; Rosa 1991). One meta-analysis placed the risk of neural tube defects even higher, at 3.8%, with particular vulnerability for the infants of women whose daily dosage exceeded 1,000 mg (Samrén et al. 1997). Other studies support this dose–response relationship (Canger et al. 1999; Kaneko et al. 1999; Omtzigt et al. 1992; Samrén et al. 1999; Tomson et al. 2011, 2015), leading one group to recommend that daily dosages not exceed 1,000 mg and that maternal serum concentrations not exceed 70 μg/mL to reduce the risk of malformations (Kaneko et al. 1999). In a case–control study examining the incidence of limb malformations in a cohort of more than 44,000 children, 67 of whom were exposed to valproate in the first trimester, Rodríguez-Pinilla et al. (2000) reported an OR of 6.17 for limb abnormalities among children exposed to valproate and estimated the risk of limb abnormalities from valproate exposure at 0.42%.

A fetal valproate syndrome was initially reported by DiLiberti et al. (1984) and subsequently confirmed by other investigators (Ardinger et al. 1988; Martínez-Frías 1990; Winter et al. 1987). The phenotypic attributes of fetal valproate syndrome include stereotypical facial features such as bifrontal narrowing, midface hypoplasia, a broad nasal bridge, a short nose with anteverted nares, epicanthal folds, micrognathia, a shallow philtrum, a thin upper lip, and a thick lower lip (McMahon and Braddock 2001; S.J. Moore et al. 2000). Many of the congenital malformations previously associated with valproate exposure have been recognized as attributes of fetal valproate syndrome (McMahon and Braddock 2001). Valproate’s antagonism of folate activity may underlie the risk of both spina bifida and fetal valproate syndrome. A case–control study comparing 57 children with fetal anticonvulsant syndromes, 46 of whose mothers were given valproate, with 152 control children found a significantly higher rate of homozygosity for a mutation in the gene for methylenetetrahydrofolate reductase (MTHFR), a key enzyme in folate metabolism, in the valproate-exposed children (Dean et al. 1999).

Neurodevelopmental outcomes associated with prenatal valproate exposure are equally of concern. A review found that developmental delay was evident in 20% and intellectual disability in 10% of children exposed to valproate monotherapy prenatally (Kozma 2001). An interim report from a prospective multicenter investigation of the neurodevelopmental effects of prenatal antiepileptic drug exposure noted that 24% of 2-year-olds with prenatal valproate exposure had mental developmental indexes of less than 70, more than double the rate associated with other antiepileptic drugs (Meador et al. 2006). More recent studies have consistently shown deleterious neurodevelopmental effects from prenatal valproate exposure (Baker et al. 2015; Cohen et al. 2013; Meador et al. 2009, 2013; Shallcross et al. 2011; Veiby et al. 2013), with particular consequences for verbal cognition (McVearry et al. 2009; Meador et al. 2011; Nadebaum et al. 2011a, 2011b). Retrospective reports indicate that varying degrees of cognitive impairment may be present in children manifesting the physical sequelae of fetal valproate syndrome (Adab et al. 2001; Gaily et al. 1990; Moore et al. 2000). Fetal valproate syndrome also has been associated with autism spectrum disorders (Bescoby-Chambers et al. 2001; Christensen et al. 2013; Moore et al. 2000; Williams and Hersh 1997; Williams et al. 2001; Wood et al. 2015).

Valproate exposure during gestation is also associated with risks for numerous fetal and neonatal toxicities, including hepatotoxicity (Kennedy and Koren 1998), coagulopathies (Mountain et al. 1970), and neonatal hypoglycemia (Ebbesen et al. 2000; Thisted and Ebbesen 1993). Ten of 13 infants who had neonatal hypoglycemia after prenatal valproate exposure developed withdrawal symptoms—including irritability, jitteriness, hypertonia, seizures, and vomiting—that began 12–24 hours after delivery and lasted up to 1 week (Ebbesen et al. 2000).

Pharmacokinetic studies in women with epilepsy have reported that maternal valproate concentrations steadily decline over the course of pregnancy, falling to levels as much as 50% lower than preconception concentrations (Yerby et al. 1990, 1992). Consistent findings from other studies indicate that valproate is more rapidly cleared during gestation, and especially during the final month of pregnancy (Nau et al. 1982b; Otani 1985; Philbert et al. 1985). Dosage increases during pregnancy therefore may be required to maintain therapeutic efficacy. Valproate readily crosses the human placenta, with umbilical cord concentrations at delivery equal to or slightly higher than maternal concentrations (Froescher et al. 1984b; Philbert et al. 1985; Yerby et al. 1990, 1992).

The database regarding valproate and lactation includes 41 mother–infant nursing dyads (Alexander 1979; Bardy et al. 1982a; Dickinson et al. 1979; Froescher et al. 1981; Nau et al. 1981; Piontek et al. 2000; Stahl et al. 1997; Tsuru et al. 1988; von Unruh et al. 1984; Wisner and Perel 1998). From these cases, only one adverse event, thrombocytopenia and anemia in an infant, has been reported (Stahl et al. 1997). The pharmacokinetic data indicate that valproate milk-to-plasma ratios are uniformly low and that serum concentrations of nursing infants are 2%–40% of maternal concentrations (Alexander 1979; Bardy et al. 1982a; Piontek et al. 2000; Stahl et al. 1997; von Unruh et al. 1984; Wisner et al. 1996b). In a study of the neurobehavioral effects of valproate exposure during lactation, Meador et al. (2014) found no adverse effects on cognitive functions.

In summary, prenatal valproate use raises grave safety concerns. In women of childbearing age, valproate should never be used except as a treatment of last resort. If valproate must be used during pregnancy, its risk may be reduced by being careful not to exceed 1,000 mg/day or a serum concentration of 70 μg/mL. Folate supplementation (4–5 mg/day) is also recommended, although no definitive evidence indicates that it reduces the risk of valproate-associated anomalies. All women of childbearing potential who are treated with valproate should receive concomitant folate supplementation, regardless of whether they plan to conceive. The preliminary evidence that aspects of fetal valproate syndrome other than neural tube defects may be associated with valproate’s antagonism of folate metabolism suggests that folate supplementation should be administered not only in the first trimester but also throughout gestation. Because of the potential for valproate-associated neonatal coagulopathies, oral vitamin K supplementation (10–20 mg/day) may be considered during the final month of gestation. Prenatal surveillance for congenital abnormalities should include maternal serum α-fetoprotein, fetal echocardiography, and a level 2 ultrasound at approximately 16–18 weeks’ gestation. Finally, genetic screening of women taking valproate for mutations in the MTHFR gene warrants future consideration.

In contrast to the marked risks of its use during pregnancy, valproate therapy during lactation appears to be well tolerated by nursing infants. Nevertheless, periodic assays of platelet counts and serum liver enzymes in nursing infants are recommended because the neurodevelopmental effect of nursing exposure to valproate is unclear.

Carbamazepine

Carbamazepine is associated with many of the same risks as valproate during gestation, although in many cases with less frequency or severity. For example, first-trimester carbamazepine exposure is associated with a 0.5%–1.0% risk of neural tube defects (Rosa 1991), which is approximately half that seen with valproate exposure (Lindhout and Schmidt 1986; Rosa 1991). A meta-analysis of five prospective studies encompassing 1,255 prenatal exposures indicated that carbamazepine exposure in utero is associated with an increased risk of neural tube defects, cleft palate, cardiovascular abnormalities, and urinary tract anomalies (Matalon et al. 2002). The European Surveillance of Congenital Anomalies (EUROCAT) study (Jentink et al. 2010), a large case–control investigation, reported that carbamazepine exposure was associated with an increased risk of spina bifida (OR=2.6) only. The Australian Register of Antiepileptic Drugs in Pregnancy showed that carbamazapine exposure in utero was associated with renal tract abnormalities (Vajda et al. 2013). An epidemiological study indicated that periconceptional folate supplementation was associated with a lower rate of neural tube defects among the children of women taking carbamazepine during pregnancy (Hernández-Díaz et al. 2001).

A fetal carbamazepine syndrome—manifested by a short nose, long philtrum, epicanthal folds, hypertelorism, upslanting palpebral fissures, and fingernail hypoplasia—has been described (Jones et al. 1989), and in one study, the phenotypic characteristics of this syndrome were observed in 6 of 47 children prenatally exposed (Ornoy and Cohen 1996). Other studies have confirmed the association with facial anomalies (Moore et al. 2000; Nulman et al. 1997b; Scolnik et al. 1994; Wide et al. 2000), but one of these studies found similar facial abnormalities among children born to women with epilepsy who were untreated during pregnancy (Nulman et al. 1997b). There has been an isolated case report of phocomelia (i.e., absence or underdevelopment of limbs) in an infant with in utero exposure (Dursun et al. 2012).

Data regarding the neurodevelopmental sequelae of prenatal carbamazepine exposure have been mixed. Some studies reported developmental delay in up to 20% of children with fetal carbamazepine syndrome (Jones et al. 1989; Meador et al. 2011; Moore et al. 2000; Ornoy and Cohen 1996; Veiby et al. 2013), but others did not find that association (Gaily et al. 1990; Scolnik et al. 1994; van der Pol et al. 1991; Wide et al. 2000). Recent systematic studies generally have found no evidence of developmental delay in children exposed to carbamazepine in utero (Baker et al. 2015; McVearry et al. 2009; Meador et al. 2006, 2009, 2011; Nadebaum et al. 2011b).

Potential fetal/neonatal toxicities associated with carbamazepine exposure include blood dyscrasias, coagulopathies, skin reactions, and hepatotoxicity. Most of these risks remain theoretical, although neonatal hepatotoxicity has been reported in a carbamazepine-exposed infant (Frey et al. 2002).

Pharmacokinetic studies of carbamazepine clearance during gestation have yielded mixed results. Some investigators have reported significant increases in carbamazepine clearance during the third trimester (Battino et al. 1982; Dam et al. 1979; Lander et al. 1981), but others have found no changes in clearance (Bardy et al. 1982b; Johnson et al. 2014; Otani 1985; Reisinger et al. 2013; Yerby et al. 1985). Placental pharmacokinetic studies indicate that the placental transfer of carbamazepine is lower than that of other anticonvulsants (Nau et al. 1982a; Yerby et al. 1990, 1992), with umbilical cord–to–maternal plasma ratios of 0.5–0.8 (Nau et al. 1982a).

The literature on carbamazepine and lactation encompasses 12 published reports and 144 mother–infant nursing pairs (Brent and Wisner 1998; Frey et al. 1990, 2002; Froescher et al. 1984a; Kaneko et al. 1982; Kok et al. 1982; Kuhnz et al. 1983; Merlob et al. 1992; Niebyl et al. 1979; Pynnönen and Sillanpää 1975; Pynnönen et al. 1977; Wisner and Perel 1998), representing the most extensive data set for any mood stabilizer in lactation. Included are 8 reports of adverse events: 1 drowsy, irritable infant with an undetectable serum concentration of carbamazepine (Kok et al. 1982); 2 “hyperexcitable” infants in whom carbamazepine levels were not reported (Kuhnz et al. 1983); 2 infants with cholestatic hepatitis in whom carbamazepine levels were not reported (Frey et al. 1990, 2002); 1 infant with poor nursing effort (Froescher et al. 1984a); 1 infant with an increased serum concentration of γ-glutamyl transpeptidase (GGT) but no overt clinical sequelae whose serum concentration was 33% of the maternal concentration (Merlob et al. 1992); and 1 infant with a seizurelike phenomenon whose carbamazepine level was 8% of the maternal level (Brent and Wisner 1998). Serum carbamazepine concentrations in the 8 nursing infants in whom these levels were assessed ranged from undetectable to 65% of the maternal level (Brent and Wisner 1998; Kok et al. 1982; Merlob et al. 1992; Pynnönen and Sillanpää 1975; Pynnönen et al. 1977; Wisner and Perel 1998).

Although the risks associated with prenatal carbamazepine exposure are marginally better than those for valproate exposure, the clinical recommendations are quite similar. Carbamazepine should be avoided in pregnancy, especially during the first trimester. Folate supplementation (4–5 mg/day) is also recommended, not only during gestation but also throughout the reproductive years because of the high prevalence of inadvertent conception in the United States. Women taking carbamazepine during gestation should receive prenatal surveillance for congenital abnormalities, including maternal serum α-fetoprotein, fetal echocardiography, and a level 2 ultrasound at approximately 16–18 weeks’ gestation. Carbamazepine has by far the most extensive database for mood stabilizers in lactation, but reports of hepatic dysfunction in nursing infants certainly raise concern. Thus, periodic assays of blood counts and serum liver enzymes of nursing infants are recommended.

Lamotrigine

Reproductive safety data for lamotrigine have rapidly accrued during the past decade and compare favorably with safety data for other mood stabilizers. The overall risk of major fetal malformations following first-trimester prenatal exposure to lamotrigine is 2.6% (207 per 7,951 exposures) (Campbell et al. 2014; Cunnington et al. 2011; Dominguez-Salgado et al. 2004; GlaxoSmithKline 2007; Holmes et al. 2006; Meador et al. 2006; Mølgaard-Nielsen and Hviid 2011; Morrow et al. 2006; Sabers et al. 2004; Vajda et al. 2003), a risk that is within the range associated with births not involving drug exposures. A report by the North American Pregnancy Registry (Holmes et al. 2006) noted a relatively high rate of midline facial clefts (0.89% of 564 exposures); however, the collective rate of orofacial clefts in the other registries was only 0.10% (3 per 2,956 exposures) (Dominguez-Salgado et al. 2004; GlaxoSmithKline 2007; Meador et al. 2006; Mølgaard-Nielsen and Hviid 2011; Morrow et al. 2006; Sabers et al. 2004; Vajda et al. 2003). Furthermore, the EUROCAT case–control study, encompassing 5,511 children with orofacial clefts and 80,052 children without clefts, reported an adjusted OR of 0.67 for clefts with lamotrigine exposure (Dolk et al. 2008). The U.K. Epilepsy and Pregnancy Register reported a higher risk of malformations at maternal dosages exceeding 200 mg/day (Morrow et al. 2006), although this finding was not confirmed in subsequent analyses (GlaxoSmithKline 2007; Mølgaard-Nielsen and Hviid 2011). Despite these reassuring findings, folate supplementation is recommended for all women of childbearing age taking any antiepileptic drug, including lamotrigine. Prospective neurodevelopmental data have been consistently favorable among children with prenatal lamotrigine exposure (Baker et al. 2015; McVearry et al. 2009; Meador et al. 2006, 2009, 2011; Nadebaum et al. 2011b).

Numerous pharmacokinetic studies have reported that lamotrigine clearance steadily increases across gestation (de Haan et al. 2004; Fotopoulou et al. 2009; Franco et al. 2008; Pennell et al. 2004, 2008; Petrenaite et al. 2005; Polepally et al. 2014; Reisinger et al. 2013; Tran et al. 2002). These studies are limited by polytherapy with other anticonvulsants, which are known to alter the metabolism of lamotrigine. It is unclear whether dosage changes would be necessary to maintain mood stability in patients with bipolar disorder and whether the adjunctive agents commonly used in the treatment of bipolar disorder would have similar effects on lamotrigine metabolism. Studies in patients taking lamotrigine also report that its rate of clearance abruptly declines after delivery (Ohman et al. 2000; Pennell et al. 2004; Polepally et al. 2014; Tran et al. 2002). Therefore, dosage reductions may be necessary after delivery to avoid maternal symptoms of lamotrigine toxicity, such as dizziness, nausea and vomiting, and diplopia (Tran et al. 2002). Published reports on the placental passage of lamotrigine indicate that lamotrigine concentrations in fetal circulation at delivery are equal to maternal concentrations (Myllynen et al. 2003; Ohman et al. 2000; Paulzen et al. 2015; Sathanandar et al. 2000; Tomson et al. 1997). There have been no reports of acute adverse events observed in neonates exposed to lamotrigine.

Reports regarding lamotrigine and lactation (Clark et al. 2013; GlaxoSmithKline 2007; Liporace et al. 2004; Newport et al. 2008c; Nordmo et al. 2009; Ohman et al. 2000; Page-Sharp et al. 2006; Rambeck et al. 1997; Tomson et al. 1997) collectively encompass 60 mother–infant nursing dyads. The only reported adverse event was an apneic episode in a nursing infant whose mother was taking lamotrigine 850 mg/day (Nordmo et al. 2009). Seven other infants were observed to have a benign thrombocytosis (Newport et al. 2008b). In the largest (n=30) of these studies (Newport et al. 2008b), the mean milk-to-plasma ratio was 41.3%, the relative infant dose equaled 9.2%, and the infant-to-maternal plasma ratio equaled 18.3%. Long-term neurobehavioral outcomes have not been studied in nursing infants exposed to lamotrigine.

Antipsychotics

Second-Generation Antipsychotics

Commonly prescribed antipsychotic medications, as well as agents used to treat the side effects of first-generation antipsychotics (FGAs), are covered in Chapters 2435. The second-generation antipsychotics (SGAs) have supplanted the FGAs as first-line medications for psychotic disorders and are also used for other psychiatric indications, including bipolar disorder, anxiety disorders, and treatment-resistant depression. Despite rapidly expanding use, the reproductive safety database regarding SGAs remains limited. A recent study that used a national pregnancy registry for SGAs reported an OR of 1.25 (95% CI=0.13–12.13) for major malformations in exposed infants compared with nonexposed infants (Cohen et al. 2015). A neurobehavioral outcome study identified a transient delay in cognitive, motor, social-emotional, and adaptive behavior in SGA-exposed infants that resolved by age 12 months (Peng et al. 2013).

Even though some data have been reassuring, the small volume of SGA data to date precludes definitive conclusions about their reproductive safety. Therefore, the routine use of SGAs during pregnancy and lactation cannot yet be recommended. Nonetheless, if a woman who is taking an SGA inadvertently conceives, a comprehensive risk–benefit assessment may indicate that continuing the SGA (to which the fetus has already been exposed) during gestation is preferable to switching to an FGA (to which the fetus has not yet been exposed).

Clozapine

Reproductive safety data for clozapine, the oldest of the SGAs, are limited to case reports (Barnas et al. 1994; Di Michele et al. 1996; Kornhuber and Weller 1991; Moreno-Bruna et al. 2012; Waldman and Safferman 1993), case series (McKenna et al. 2005; Stoner et al. 1997; Tenyi and Tixler 1998), and a retrospective review (Dev and Krupp 1995), collectively encompassing 80 children exposed to clozapine during pregnancy and/or lactation. Adverse sequelae associated with in utero clozapine therapy include maternal gestational diabetes (Dickson and Hogg 1998); several minor anomalies, including cephalohematoma, hyperpigmentation folds, and a coccygeal dimple, in an infant (Stoner et al. 1997); delayed peristalsis (Moreno-Bruna et al. 2012); transient low-grade fever in an infant also exposed to lithium (Stoner et al. 1997); and floppy infant syndrome in a newborn also exposed to lorazepam (Di Michele et al. 1996). In a review of 61 children exposed to clozapine prenatally, Dev and Krupp (1995) reported 5 cases of congenital malformations and 5 cases of neonatal syndromes; however, many of these mothers were taking other psychotropic medications during pregnancy. A recent study of neurodevelopmental consequences of prenatal exposure to SGAs reported higher rates of developmental delay at ages 2 months and 6 months among clozapine-exposed infants compared with infants exposed to other SGAs (Shao et al. 2015). The only reported case of clozapine use during lactation noted no adverse effects on the nursing infant (Kornhuber and Weller 1991).

The only investigation of the perinatal pharmacokinetics of clozapine found elevated concentrations in fetal serum and breast milk compared with concentrations in maternal serum and amniotic fluid (Barnas et al. 1994), leading the authors to conclude that clozapine accumulates in fetal circulation and breast milk. Although no cases of agranulocytosis have been reported in clozapine-exposed infants, this potential risk and the requisite laboratory monitoring limit the utility of clozapine during the peripartum.

Olanzapine

A published birth registry of 23 prospectively ascertained olanzapine-exposed pregnancies from the Lilly Worldwide Pharmacovigilance Safety Database reported no major malformations, 13% spontaneous abortions, 5% preterm deliveries, and 5% fetal deaths (Goldstein et al. 2000). In a subsequent examination of the same database, 610 prospectively identified pregnancies exposed to olanzapine resulted in 10% premature births, 9% spontaneous abortions, 8% perinatal conditions (i.e., an adverse event occurring within 7 days of birth), and 4% congenital anomalies (Brunner et al. 2013). A prospective study comparing outcomes among 151 pregnant women with SGA exposure (olanzapine, n=60; risperidone, n=49; quetiapine, n=36; clozapine, n=6) with outcomes among 151 pregnant control subjects reported no differences in rates of spontaneous abortion, stillbirth, major malformations, prematurity, or low birth weight (McKenna et al. 2005). In this study, one SGA-exposed child (olanzapine) was observed to have major malformations (a series of midline defects including an oral cleft, encephalocele, and aqueductal stenosis). A recent meta-analysis reported a malformation rate of 3.5% among 1,090 olanzapine-exposed infants, a rate that compares favorably to rates with other SGAs (Ennis and Damkier 2015).

In a study of antipsychotic placental passage rates and neonatal outcomes (Newport et al. 2007a), umbilical cord concentrations in olanzapine-exposed neonates (n=14) were 72.1% of maternal concentrations. In this study, there was a trend toward higher rates of low birth weight (30.8%; P<0.06) and neonatal intensive care unit admission (30.8%; P<0.09) among neonates exposed to olanzapine compared with those exposed to the other antipsychotics.

Case reports of 39 infants exposed to olanzapine during lactation with no evidence of infant toxicity currently appear in the literature (Croke et al. 2002; Friedman and Rosenthal 2003; Gardiner et al. 2003; Gilad et al. 2011; Goldstein et al. 2000; Kirchheiner et al. 2000; Whitworth et al. 2010). The Lilly Safety Database reported on 62 breast-feeding mother–infant dyads with a 15.6% rate of adverse events, identified as somnolence, irritability, tremors, and insomnia (Brunner et al. 2013). Pharmacokinetic studies of olanzapine exposure during lactation have reported that plasma concentrations were undetectable in infants during nursing (Gardiner et al. 2003; Kirchheiner et al. 2000; Stiegler et al. 2014) and that the median infant daily dosage via breast feeding was approximately 0.7%–1.8% of the maternal dosage (Ambresin et al. 2004; Aydin et al. 2015; Brunner et al. 2013; Croke et al. 2002; Gardiner et al. 2003).

Risperidone

Prospective data on pregnancy outcomes following first-trimester risperidone exposure include a collective study of outcomes for several SGAs (McKenna et al. 2005) reporting no major malformations among 49 risperidone-exposed infants and a recent meta-analysis reporting a 5.1% malformation rate among 432 risperidone-exposed infants (Ennis and Damkier 2015). An additional study in 68 women with first-trimester exposure and known outcomes reported 9 (13.2%) spontaneous abortions, 1 (1.5%) stillbirth, and 2 (2.9%) children with major malformations (Coppola et al. 2007). No data on the neurodevelopmental effects of risperidone exposure during pregnancy or lactation are available.

Pharmacokinetic studies have reported placental passage concentrations among neonates (n=6) of 49.2% (Newport et al. 2007a), milk-to-plasma ratios of less than 0.5 for risperidone and less than 0.88 for 9-hydroxyrisperidone (Hill et al. 2000; Ilett et al. 2004; Weggelaar et al. 2011), and infant plasma concentrations ranging from 2.3% to 4.7% of maternal levels (Ilett et al. 2004; Weggelaar et al. 2011).

Quetiapine

The reproductive safety literature for first-trimester quetiapine exposure is limited to a case series of two successive pregnancies (Grover and Madan 2012), which reported healthy, full-term deliveries, and the McKenna et al. (2005) study, which reported no major malformations among 36 infants exposed to quetiapine. In the Newport et al. (2007a) study of antipsychotic placental passage, quetiapine concentrations among neonates (n=21) were 23% of maternal concentrations. To our knowledge, this is the lowest placental passage rate ever reported for a psychotropic agent. No data are available on the neurodevelopmental effects of quetiapine exposure during pregnancy or lactation.

Three cases of quetiapine use during lactation following use during pregnancy estimated the nursing infant dosage at 0.09%–7.3% of the maternal daily dosage (Lee et al. 2004; Van Boekholt et al. 2015).

Aripiprazole

Reproductive safety data for aripiprazole include a case series of 86 mother–infant dyads that showed an increased risk of premature birth and fetal growth retardation but no increased risk of congenital malformations, miscarriages, preeclampsia, or gestational diabetes (Bellet et al. 2015). In addition, a recent meta-analysis reported a 5.0% malformation rate among 100 infants with first-trimester aripiprazole exposure (Ennis and Damkier 2015). A single case report indicated poor respiratory effort at birth (Watanabe et al. 2011), and two case reports and a case series showed no adverse obstetrical or neonatal outcomes (Gentile et al. 2011; Lutz et al. 2010; Windhager et al. 2014).

Of the three case reports of aripiprazole use during lactation, the milk excretion profiles were questionable in two (Lutz et al. 2010; Schlotterbeck et al. 2007), and the other reported a milk-to-plasma ratio of 0.051 (Watanabe et al. 2011). Placental transfer was 53.3%–54.7%, as shown in two reports of four mother–infant dyads (Watanabe et al. 2011; Windhager et al. 2014).

Other SGAs

Information about the use of other SGAs is scant. Use of ziprasidone, brexpiprazole, lurasidone, iloperidone, or asenapine during pregnancy has not been reported. A lone case report of use of paliperidone palmitate, the long-acting injectable formulation of paliperidone, during pregnancy (haloperidol was also used) indicated no adverse obstetrical outcomes (Özdemir et al. 2015).

Data concerning lactation were limited to a single case report of ziprasidone use during lactation that reported a milk-to-plasma ratio of 0.06 (Schlotterbeck et al. 2009).

First-Generation Antipsychotics

In contrast to the SGAs, the FGAs have an extensive reproductive safety database addressing both somatic and neurobehavioral teratogenicity. Furthermore, the historical use of phenothiazine antipsychotics to treat pregnancy-associated emesis aids in separating the effects of psychiatric illness and antipsychotic drugs on pregnancy outcome. Chlorpromazine, haloperidol, and perphenazine have received the greatest scrutiny, with no significant associations between these compounds and major malformations (Goldberg and DiMascio 1978; Hill and Stern 1979; Nurnberg and Prudic 1984).

In a study of 100 women taking haloperidol (mean dosage=1.2 mg/day) for hyperemesis gravidarum, no differences in gestational duration, fetal viability, or birth weight were noted (Van Waes and Van de Velde 1969). In a prospective study encompassing nearly 20,000 women receiving primarily phenothiazines for emesis, Milkovich and van den Berg (1976) found no significant association with neonatal survival rates or severe anomalies after controlling for maternal age, medication, and gestational age at exposure. Similar results have been obtained in several retrospective studies of women taking trifluoperazine for repeated abortions and emesis (Moriarty and Nance 1963; Rawlings et al. 1963). In contrast, Rumeau-Rouquette et al. (1977) reported a significant association of major anomalies with prenatal exposure to aliphatic phenothiazines but not to piperazine- or piperidine-class agents. Reanalysis of the data obtained by Milkovich and van den Berg (1976) did find a significant risk of malformations associated with phenothiazine exposure in weeks 4 through 10 of gestation (Edlund and Craig 1984).

Neurobehavioral outcome studies encompassing 203 children exposed to FGAs during gestation detected no significant differences in IQ scores at age 4 years (Kris 1965; Slone et al. 1977), although relatively low antipsychotic dosages were used by many women in these studies. Conversely, several laboratory animal studies (Hoffeld et al. 1968; Ordy et al. 1966; Robertson et al. 1980), although not all (Dallemagne and Weiss 1982), have identified persistent deficits in learning and memory among offspring prenatally exposed to FGA medications.

Beyond the teratogenic potential of the FGAs lies the possibility of fetal and infant toxicities such as neuroleptic malignant syndrome (James 1988) and extrapyramidal side effects (EPS) manifested by heightened muscle tone and increased rooting and tendon reflexes persisting for several months (Cleary 1977; Hill et al. 1966; O’Connor et al. 1981). Furthermore, prenatal exposure to FGAs has been associated with neonatal jaundice (Scokel and Jones 1962) and postnatal intestinal obstruction (Falterman and Richardson 1980).

In our study of antipsychotic placental passage, neonatal haloperidol (n=13) concentrations were 66% of maternal concentrations (Newport et al. 2007a).

In lactation, chlorpromazine is the most widely studied typical antipsychotic, with 7 infants exposed to chlorpromazine during nursing showing no developmental deficits at 16-month and 5-year follow-up evaluations (Kris and Carmichael 1957). However, 3 infants in another study whose mothers were prescribed both chlorpromazine and haloperidol showed evidence of developmental delay at 12–18 months (Yoshida et al. 1998b). Pharmacokinetic investigations of FGAs during lactation, including haloperidol (Stewart et al. 1980; Whalley et al. 1981; Yoshida et al. 1998b), trifluoperazine (Wilson et al. 1980; Yoshida et al. 1998b), perphenazine (Wilson et al. 1980), thioxanthenes (Matheson and Skjaeraasen 1988), and chlorpromazine (Yoshida et al. 1998b), have uniformly reported milk-to-plasma ratios of less than 1.0, although adequate control for distribution and time gradients was lacking in these studies. One group postulated that the physicochemical properties of perphenazine could lead it to become “trapped” in breast milk (Wilson et al. 1980); however, this speculation is unconfirmed.

Fetal and infant exposure to any of the various agents available for the management of EPS (e.g., diphenhydramine, benztropine, amantadine) also raises concern. Results of the Collaborative Perinatal Project indicated that first-trimester exposure to diphenhydramine, the best studied of these medications, was associated with major and minor congenital anomalies (Miller 1991; Wisner and Perel 1988). A case–control study found a significantly higher rate of prenatal diphenhydramine exposure among 599 infants with oral clefts than among 590 control infants (Saxén 1974). Despite these data, diphenhydramine is routinely used during pregnancy. Clinical studies of the teratogenic potential of benztropine and amantadine are lacking, although laboratory animal studies indicated that amantadine is associated with an elevated risk of congenital malformations (Hirsch and Swartz 1980). Perinatal toxicities, including neonatal intestinal obstruction after gestational exposure to benztropine (Falterman and Richardson 1980) and a possible neonatal diphenhydramine withdrawal syndrome manifested by tremulousness and diarrhea (Parkin 1974), also warrant concern.

In summary, FGAs have been widely used for more than 40 years, and the paucity of data linking these agents to either teratogenic or toxic effects suggests that their risks are minimal. In particular, piperazine phenothiazines (e.g., trifluoperazine, perphenazine) may have especially low teratogenic potential (Rumeau-Rouquette et al. 1977). Given the greater fetal risks associated with anticholinergic medications (e.g., diphenhydramine), their use should be avoided if possible. Consequently, FGAs used during the antepartum period should be kept at the lowest effective dosage to minimize the need for adjunctive medications to manage EPS.

Anxiolytics

Pharmacotherapy for anxiety disorders includes antidepressants, benzodiazepines, buspirone, and certain atypical antipsychotics.

Benzodiazepines

A retrospective analysis of more than 100,000 women found that at least 2% were prescribed a benzodiazepine during gestation (Bergman et al. 1992). The earliest benzodiazepine teratogenicity studies reported an increased risk of oral clefts after in utero diazepam exposure (Aarskog 1975; Saxén 1975; Saxén and Saxén 1975), but later studies failed to confirm this association (Ban et al. 2014; Entman and Vaughn 1984; Rosenberg et al. 1983; Shiono and Mills 1984). Prospective studies of first-trimester alprazolam exposure encompassing approximately 1,300 pregnancies indicated no excess of oral clefts or other birth defects (Barry and St Clair 1987; Schick-Boschetto and Zuber 1992; St Clair and Schirmer 1992), and a recent study of first-trimester exposure to diazepam and temazepam echoed this finding (Ban et al. 2014). A meta-analysis found that prenatal benzodiazepine exposure does confer an increased risk of oral clefts, although the absolute risk increased by only 0.01% (Altshuler et al. 1996). This conclusion is consistent with the findings of a later case–control study that found no difference in rates of prenatal benzodiazepine exposure between more than 38,000 infants with congenital anomalies and nearly 23,000 control children (Eros et al. 2002).

Although benzodiazepine teratogenicity data are somewhat mixed, benzodiazepine neonatal syndromes are well documented. Numerous groups have described a floppy infant syndrome characterized by hypothermia, lethargy, poor respiratory effort, and feeding difficulties following benzodiazepine exposure in late pregnancy (Erkkola et al. 1983; Fisher et al. 1985; Haram 1977; Källén and Reis 2012; Kriel and Cloyd 1982; McAuley et al. 1982; Sanchis et al. 1991; Speight 1977; Woods and Malan 1978). In a study of 53 infants with late-pregnancy lorazepam exposure, term infants whose mothers had taken oral lorazepam showed no evidence of toxicity other than a brief delay in establishing feeding, whereas preterm infants and term infants whose mothers had received larger intravenous doses of lorazepam had symptoms consistent with floppy infant syndrome (Whitelaw et al. 1981). Neonatal withdrawal syndromes, characterized by restlessness, hypertonia, hyperreflexia, tremulousness, apnea, diarrhea, and vomiting, have been described in infants whose mothers were taking alprazolam (Barry and St Clair 1987), chlordiazepoxide (Athinarayanan et al. 1976; Bitnun 1969; Stirrat et al. 1974), or diazepam (Backes and Cordero 1980; Mazzi 1977). Benzodiazepine neonatal syndromes have been reported to persist for as long as 3 months after delivery (for a review, see Miller 1991).

Pharmacokinetic studies during pregnancy indicate that benzodiazepines readily traverse the placenta and with prolonged administration may accumulate in the fetus (Mandelli et al. 1975; Shannon et al. 1972). For example, fetal concentrations of diazepam at delivery are typically higher than maternal concentrations (Erkkola et al. 1974). Because benzodiazepine metabolism is slower in the fetus than in the mother, it is understandable that an agent like diazepam (which has multiple active metabolites) might accumulate in the fetus. In addition, high concentrations of diazepam are sequestered in lipophilic fetal tissues, such as the brain, lungs, and heart (Mandelli et al. 1975). In contrast, fetal-to-maternal ratios of lorazepam (which has no active metabolites) are typically less than 1.0 (Whitelaw et al. 1981). Yet neonatal clearance of lorazepam is slow, with detectable levels evident 8 days after delivery (Whitelaw et al. 1981), and the clearance of chlordiazepoxide appears to be even slower (Athinarayanan et al. 1976).

Studies evaluating the neurobehavioral effects of prenatal benzodiazepine exposure are needed. A benzodiazepine exposure syndrome—consisting of growth retardation, dysmorphism, and mental and psychomotor retardation in infants (Laegreid et al. 1987)—has been reported, although some investigators have disputed this finding (Gerhardsson and Alfredsson 1987; Winter 1987). One group found no differences in the incidence of behavioral abnormalities at age 8 months or in IQ scores at age 4 years among children exposed to chlordiazepoxide during gestation (Hartz et al. 1975), and another group found no correlation between antenatal exposure and language competence at age 3 years (Odsbu et al. 2015). Nevertheless, a series of laboratory animal studies raised concerns that prenatal benzodiazepine exposure may produce deficits in memory and learning ability (Frieder et al. 1984; Hassmannova and Myslivecek 1994; Jaiswal and Bhattacharya 1993; Myslivecek et al. 1991).

Buist et al. (1990) concluded that benzodiazepines at low dosages present no contraindication to nursing. A recent retrospective cohort study found a sedation rate of 1.6% in infants exposed to benzodiazepines while breast feeding, but there was no association with maternal dosage (Kelly et al. 2012). Infants with impaired metabolic capacity may show sedation and poor feeding even with low maternal dosages (Wesson et al. 1985). Overall, benzodiazepines are associated with lower milk-to-plasma ratios than are other classes of psychotropics. For example, Wretlind (1987) found a milk-to-plasma ratio of 0.1–0.3 for oxazepam and calculated that the infant daily dosage via lactation is 1/1,000th of the maternal dosage. The percentage of the maternal dosage of lorazepam to which a nursing infant is exposed has been estimated to be 2.2% (Summerfield and Nielsen 1985).

In summary, benzodiazepines do not appear to carry a significant risk of somatic teratogenesis, but neurobehavioral sequelae remain obscure. Because benzodiazepines are associated with neonatal risks, they should be used judiciously and tapered before delivery if possible. Benzodiazepines should not be abruptly withdrawn during pregnancy. Because lorazepam and oxazepam are less dependent on hepatic metabolism, they theoretically have less potential for fetal accumulation during pregnancy. Finally, benzodiazepines can be safely administered, in judicious doses, during lactation. However, breast feeding should be discontinued if an infant experiences sedation or other signs of benzodiazepine toxicity.

Buspirone

Buspirone has garnered a unique, and arguably illogical, position within the psychotropic armamentarium. Although buspirone is labeled FDA Category B, published data are extremely limited. The only report of prenatal administration of buspirone identified 1 infant born with a major malformation among 14 infants with first-trimester buspirone exposure (Wilton et al. 1998). No published reports exist regarding buspirone’s safety during lactation.

Future Directions and General Recommendations

The development of prenatal and postnatal treatment guidelines is hampered by the haphazard accrual of clinical research data with inconsistent methodologies. Whereas clinical data have confirmed the teratogenic potential for only a few psychotropic agents, animal studies, which commonly use maternal concentrations exponentially higher than those seen in clinical care, show clear somatic and neurobehavioral teratogenicity (Elia et al. 1987). Such inconsistencies between clinical and preclinical data further confound efforts to construct reliable treatment recommendations. Similarly, advances in statistical methods and the accrual of data from a variety of sources have identified statistical findings that may have limited relevance to clinical decision making (Ranganathan et al. 2015). Because pregnant and nursing women are generally excluded from clinical trials of pharmacological agents, definitive clarification with proper control groups is unlikely to be forthcoming in the near future. Consequently, clinical treatment decisions are made on the basis of incomplete or uncertain information.

Informed Consent

Obtaining informed consent in the clinical decision cannot be overemphasized. Informed consent should include the following:

  • Agreement on the primary treatment objective, which typically is to minimize potential harm to the fetus (Acceptance of and adherence to this objective will help reduce the potential for subsequent maternal self-recrimination.)

  • Discussion of the availability, affordability, accessibility, and potential efficacy of nonpharmacological treatments

  • Consideration of the likelihood and severity of illness without continued pharmacotherapy

  • Review of the potential risks of untreated mental illness to the fetus and mother

  • Review of the potential risks of fetal psychotropic exposure

  • Acknowledgment that no risk-free alternative is available other than deciding not to conceive

  • Acknowledgment that the understanding of risks remains incomplete (Ultimately, it is impossible to provide an exhaustive list of all risks for any given psychotropic agent, but the evidence—or lack of evidence—for adverse effects should be reviewed.)

Nonpsychotropic Treatment Strategies to Reduce In Utero Exposures

Nonpsychotropic treatments should focus on maximizing maternal health during pregnancy, such as prenatal vitamins; adequate hydration; daily exercise; and avoidance of alcohol, tobacco, caffeine, and illicit substances. Good evidence indicates that concomitant exposures, such as to tobacco, increase both obstetrical and neonatal risk. Nonpsychotropic treatment strategies also include psychotherapy alternatives appropriate during pregnancy and the postpartum period.

Medication Selection

Medication selection requires several steps. Initial treatment planning for reproductive-age women with mental illness should include the potential pregnancy and future family planning. Notably, more than 45% of the pregnancies in the United States are unplanned, and knowledge of conception is often well into the organogenesis (Finer and Zolna 2016; Kost 2015). Choosing initial pharmacotherapy with this in mind supports “new and improved=limited data.”

Psychotropic therapy is administered after knowledge of conception only when the clinician and patient agree that the risks to the fetus of untreated mental illness likely exceed the risks of fetal psychotropic exposure. Consequently, both the safety and the efficacy of the psychotropic regimen are important considerations. A psychotropic agent is preferred if 1) it has previously been effective for the patient; 2) it has previously been well tolerated by the patient; 3) the fetus has already been exposed to it (i.e., the patient was taking the agent at knowledge of conception); and 4) it has a favorable reproductive safety profile. It is important to emphasize the potential hazards of switching agents during pregnancy or lactation. Changing medications generally should be avoided (unless well-defined risks are associated with the current regimen), because medication changes increase the number of medications to which the infant has been exposed, limit the clinician’s ability to apply available safety information (e.g., virtually no data exist on concomitant or tandem multiple medication exposures), and may heighten the mother’s vulnerability to illness.

Dosage Management

Maintaining maternal emotional well-being is the goal of psychotropic treatment in the antepartum and postpartum periods. Partial or subtherapeutic treatment heightens risk by continuing to expose the mother and infant to both illness and medications. The minimum effective dosage should be maintained throughout treatment, and the clinician should remain mindful that dosage requirements may change during pregnancy. Similarly, clinicians and patients should be aware that dosage adjustment may not significantly alter fetal exposure during pregnancy (i.e., the fetus is exposed to the maternal plasma concentration, not the maternal dosage). To minimize the potential for neonatal withdrawal and maternal toxicity after delivery, careful monitoring of side effects and serum concentrations may be indicated.

Monitoring of Nursing Infants

Because clinical laboratory assays typically lack the sensitivity to detect the serum concentrations of nursing infants, infant plasma monitoring for psychotropic medications is not routinely indicated. If there is an index of suspicion that a child is experiencing an adverse effect from nursing exposure to a psychotropic medication, breast feeding should be discontinued. However, routine laboratory monitoring (e.g., blood counts, electrolytes, hepatic profiles) is required when nursing mothers are taking medications (e.g., lithium, valproate, carbamazepine) with low therapeutic indices or known systemic toxicities.

Resources

For clinicians involved in the care of numerous women in their reproductive years, a valuable resource that includes an interactive e-book is Drugs in Pregnancy and Lactation by G.G. Briggs and R.K. Freeman.

Additional online resources include the following:

Centers for Disease Control and Prevention—www.cdc.gov/pregnancy/meds/treatingfortwo

InfantRisk Center—www.infantrisk.com

LactMed—https://toxnet.nlm.nih.gov/newtoxnet/lactmed.htm

Organization of Teratology Information Specialists: MotherToBaby—www.mothertobaby.org

Reprotox—www.reprotox.com

References

Aarskog D: Letter: Association between maternal intake of diazepam and oral clefts. Lancet 2(7941):921, 1975 53396

Adab N, Jacoby A, Smith D, et al: Additional educational needs in children born to mothers with epilepsy. J Neurol Neurosurg Psychiatry 70(1):15–21, 2001 11118242

Agatonovic-Kustrin S, Ling LH, Tham SY, et al: Molecular descriptors that influence the amount of drugs transfer into human breast milk. J Pharm Biomed Anal 29(1–2):103–119, 2002 12062670

Aichhorn W, Whitworth AB, Weiss U, et al: Mirtazapine and breast-feeding. Am J Psychiatry 161(12):2325, 2004 15569912

Alexander FW: Sodium valproate and pregnancy. Arch Dis Child 54(3):240, 1979 373647

Allen S: A quantitative analysis of the process, mediating variables, and impact of traumatic childbirth. Journal of Reproductive and Infant Psychology 16:107–131, 1998

Altshuler LL, Burt VK, McMullen M, et al: Breastfeeding and sertraline: a 24-hour analysis. J Clin Psychiatry 56(6):243–245, 1995 7775366

Altshuler LL, Cohen L, Szuba MP, et al: Pharmacologic management of psychiatric illness during pregnancy: dilemmas and guidelines. Am J Psychiatry 153(5):592–606, 1996 8615404

Alwan S, Reefhuis J, Rasmussen SA, et al; National Birth Defects Prevention Study: Use of selective serotonin-reuptake inhibitors in pregnancy and the risk of birth defects. N Engl J Med 356(26):2684–2692, 2007 17596602

Ambresin G, Berney P, Schulz P, et al: Olanzapine excretion into breast milk: a case report. J Clin Psychopharmacol 24(1):93–95, 2004 14709955

American Academy of Pediatrics Committee on Drugs: Transfer of drugs and other chemicals into human milk. Pediatrics 108(3):776–789, 2001 11533352

Andrade SE, McPhillips H, Loren D, et al: Antidepressant medication use and risk of persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf 18(3):246–252, 2009 19148882

Ardinger HH, Atkin JF, Blackston RD, et al: Verification of the fetal valproate syndrome phenotype. Am J Med Genet 29(1): 171–185, 1988 3125743

Assencio-Ferreira VJ, Abraham R, Veiga JC, et al: [Metopic suture craniosynostosis: sodium valproate teratogenic effect: case report] (Portuguese). Arq Neuropsiquiatr 59(2-B):417–420, 2001 11460190

Athinarayanan P, Pierog SH, Nigam SK, et al: Chlordiazepoxide withdrawal in the neonate. Am J Obstet Gynecol 124(2):212–213, 1976 1247060

Audus KL: Controlling drug delivery across the placenta. Eur J Pharm Sci 8(3):161–165, 1999 10379038

Ayad M, Costantine MM: Epidemiology of medications use in pregnancy. Semin Perinatol 39(7):508–511, 2015 26358804

Aydin B, Nayir T, Sahin S, et al: Olanzapine and quetiapine use during breastfeeding: excretion into breast milk and safe breastfeeding strategy. J Clin Psychopharmacol 35(2):206–208, 2015 25679127

Backes CR, Cordero L: Withdrawal symptoms in the neonate from presumptive intrauterine exposure to diazepam: report of case. J Am Osteopath Assoc 79(9):584–585, 1980 7380678

Baker GA, Bromley RL, Briggs M, et al; Liverpool and Manchester Neurodevelopment Group: IQ at 6 years after in utero exposure to antiepileptic drugs: a controlled cohort study. Neurology 84(4):382–390, 2015 25540307

Ban L, West J, Gibson JE, et al: First trimester exposure to anxiolytic and hypnotic drugs and the risks of major congenital anomalies: a United Kingdom population-based cohort study. PLoS One 9(6): e100996, 2014 24963627

Bardy AH, Granström ML, Hiilesmaa VK: Valproic acid and breast feeding, in Epilepsy, Pregnancy, and the Child. Edited by Janz D, Dam M, Richens A. New York, Raven, 1982a, pp 359–360

Bardy AH, Teramo K, Hiilesmaa VK: Apparent plasma clearances of phenytoin, phenobarbitone, primidone, and carbamazepine during pregnancy: results of the prospective Helsinki study, in Epilepsy, Pregnancy, and the Child. Edited by Janz D, Dam M, Richens A, et al. New York, Raven, 1982b, pp 141–145

Barnas C, Bergant A, Hummer M, et al: Clozapine concentrations in maternal and fetal plasma, amniotic fluid, and breast milk (abstract). Am J Psychiatry 151(6):945, 1994 8185013

Barry WS, St Clair SM: Exposure to benzodiazepines in utero. Lancet 1(8547):1436–1437, 1987 2884529

Barson AJ: Malformed infant (letter). BMJ 2(5804):45, 1972 5015974

Battino D, Avanzini G, Bossi L, et al: Monitoring of antiepileptic drug plasma levels during pregnancy and puerperium, in Epilepsy, Pregnancy, and the Child. Edited by Janz D, Dam M, Richens A, et al. New York, Raven, 1982, pp 147–154

Bellantuono C, Marini A, Lucarelli C: Infant health and neurodevelopmental outcomes following prenatal exposure to duloxetine. Clin Drug Investig 33(9):685–688, 2013 23873363

Bellet F, Beyens MN, Bernard N, et al: Exposure to aripiprazole during embryogenesis: a prospective multicenter cohort study. Pharmacoepidemiol Drug Saf 24(4):368–380, 2015 25683615

Bennedsen BE, Mortensen PB, Olesen AV, et al: Obstetric complications in women with schizophrenia. Schizophr Res 47(2–3):167–175, 2001 11278134

Bérard A, Ramos E, Rey E, et al: First trimester exposure to paroxetine and risk of cardiac malformations in infants: the importance of dosage. Birth Defects Res B Dev Reprod Toxicol 80(1):18–27, 2007 17187388

Bergman U, Rosa FW, Baum C, et al: Effects of exposure to benzodiazepine during fetal life. Lancet 340(8821):694–696, 1992 1355799

Bertossi M, Virgintino D, Errede M, et al: Immunohistochemical and ultrastructural characterization of cortical plate microvasculature in the human fetus telencephalon. Microvasc Res 58(1):49–61, 1999 10388603

Bescoby-Chambers N, Forster P, Bates G: Foetal valproate syndrome and autism: additional evidence of an association. Dev Med Child Neurol 43(12):847, 2001 11769274

Birnbaum CS, Cohen LS, Bailey JW, et al: Serum concentrations of antidepressants and benzodiazepines in nursing infants: a case series. Pediatrics 104(1):e11, 1999 10390297

Bitnun S: Possible effect of chlordiazepoxide on the fetus. Can Med Assoc J 100(7):351, 1969 PMC1945650

Bjerkedal T, Czeizel A, Goujard J, et al: Valproic acid and spina bifida. Lancet 2(8307):1096, 1982 6127554

Black MM, Matula K: Essentials of Bayley Scales of Infant Development, II: Assessment. New York, Wiley, 2000

Bogen DL, Sit D, Genovese A, et al: Three cases of lithium exposure and exclusive breastfeeding. Arch Women Ment Health 15(1):69–72, 2012 22277970

Bologa M, Tang B, Klein J, et al: Pregnancy-induced changes in drug metabolism in epileptic women. J Pharmacol Exp Ther 257(2):735–740, 1991 2033516

Boobis AR, Lewis PJ: Pharmacokinetics in pregnancy, in Clinical Pharmacology in Obstetrics. Edited by Lewis P. Boston, MA, Wright-PSG, 1983, pp 6–54

Boshier A, Wilton LV, Shakir SA: Evaluation of the safety of bupropion (Zyban) for smoking cessation from experience gained in general practice use in England in 2000. Eur J Clin Pharmacol 59(10):767–773, 2003 14615857

Boukhris T, Sheehy O, Mottron L, et al: Antidepressant use during pregnancy and the risk of autism spectrum disorder in children. JAMA Pediatr 170(2):117–124, 2016 26660917

Boyce PM, Hackett LP, Ilett KF: Duloxetine transfer across the placenta during pregnancy and into milk during lactation. Arch Women Ment Health 14(2):169–172, 2011 21359876

Brent NB, Wisner KL: Fluoxetine and carbamazepine concentrations in a nursing mother/infant pair. Clin Pediatr (Phila) 37(1):41–44, 1998 9475699

Briggs GG, Samson JH, Ambrose PJ, et al: Excretion of bupropion in breast milk. Ann Pharmacother 27(4):431–433, 1993 8477117

Briggs GG, Freeman RK, Yaffe SJ: Drugs in Pregnancy and Lactation, 7th Edition. Philadelphia, PA, Lippincott Williams & Wilkins, 2005

Brockington IF, Winokur G, Dean C: Puerperal psychosis, in Motherhood and Mental Illness. Edited by Brockington IF, Kumar R. London, Academic Press, 1982, pp 37–69

Brunner E, Falk DM, Jones M, et al: Olanzapine in pregnancy and breastfeeding: a review of data from global safety surveillance. BMC Pharmacol Toxicol 14:38, 2013 23902726

Buesching DP, Glasser ML, Frate DA: Progression of depression in the prenatal and postpartum periods. Women Health 11(2):61–78, 1986 3751082

Buist A, Norman TR, Dennerstein L: Breastfeeding and the use of psychotropic medication: a review. J Affect Disord 19(3): 197–206, 1990 2145340

Burch KJ, Wells BG: Fluoxetine/norfluoxetine concentrations in human milk. Pediatrics 89(4 pt 1):676–677, 1992 1557252

Campbell E, Kennedy F, Russell A, et al: Malformation risks of antiepileptic drug monotherapies in pregnancy: updated results from the UK and Ireland Epilepsy and Pregnancy Registers. J Neurol Neurosurg Psychiatry 85(9):1029–1034, 2014 24444855

Canger R, Battino D, Canevini MP, et al: Malformations in offspring of women with epilepsy: a prospective study. Epilepsia 40(9):1231–1236, 1999 10487185

Capello CF, Bourke CH, Ritchie JC, et al: Serotonin transporter occupancy in rats exposed to serotonin reuptake inhibitors in utero or via breast milk. J Pharmacol Exp Ther 339(1):275–285, 2011 21775476

Casper RC, Fleisher BE, Lee-Ancajas JC, et al: Follow-up of children of depressed mothers exposed or not exposed to antidepressant drugs during pregnancy. J Pediatr 142(4):402–408, 2003 12712058

Castro VM, Kong SW, Clements CC, et al: Absence of evidence for increase in risk for autism or attention-deficit hyperactivity disorder following antidepressant exposure during pregnancy: a replication study. Transl Psychiatry 6:e708, 2016 26731445

Centers for Disease Control (CDC): Spina bifida incidence at birth—United States, 1983–1990. MMWR Morb Mortal Wkly Rep 41(27):497–500, 1992 1614388

Chambers CD, Johnson KA, Dick LM, et al: Birth outcomes in pregnant women taking fluoxetine. N Engl J Med 335(14): 1010–1015, 1996 8793924

Chambers CD, Hernandez-Diaz S, Van Marter LJ, et al: Selective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newborn. N Engl J Med 354(6):579–587, 2006 16467545

Christensen J, Grønborg TK, Sørensen MJ, et al: Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA 309(16):1696–1703, 2013 23613074

Chun-Fai-Chan B, Koren G, Fayez I, et al: Pregnancy outcome of women exposed to bupropion during pregnancy: a prospective comparative study. Am J Obstet Gynecol 192(3):932–936, 2005 15746694

Clark CT, Klein AM, Perel JM, et al: Lamotrigine dosing for pregnant patients with bipolar disorder. Am J Psychiatry 170(11): 1240–1247, 2013 24185239

Cleary MF: Fluphenazine decanoate during pregnancy. Am J Psychiatry 134(7):815–816, 1977 869065

Clements CC, Castro VM, Blumenthal SR, et al: Prenatal antidepressant exposure is associated with risk for attention-deficit hyperactivity disorder but not autism spectrum disorder in a large health system. Mol Psychiatry 20(6):727–734, 2015 25155880

Cohen LS, Friedman JM, Jefferson JW, et al: A reevaluation of risk of in utero exposure to lithium. JAMA 271(2):146–150, 1994 8031346

Cohen LS, Altshuler LL, Harlow BL, et al: Relapse of major depression during pregnancy in women who maintain or discontinue antidepressant treatment. JAMA 295(5):499–507, 2006 16449615

Cohen LS, Viguera AC, McInerney KA, et al: Reproductive safety of second-generation antipsychotics: current data from the Massachusetts General Hospital National Pregnancy Registry for Atypical Antipsychotics. Am J Psychiatry 173(3):263–270, 2015 26441156

Cohen MJ, Meador KJ, Browning N, et al; NEAD study group: Fetal antiepileptic drug exposure: adaptive and emotional/behavioral functioning at age 6 years. Epilepsy Behav 29(2):308–315, 2013 24012508

Cole JA, Ephross SA, Cosmatos IS, et al: Paroxetine in the first trimester and the prevalence of congenital malformations. Pharmacoepidemiol Drug Saf 16(10):1075–1085, 2007 17729379

Coppola D, Russo LJ, Kwarta RF Jr, et al: Evaluating the postmarketing experience of risperidone use during pregnancy: pregnancy and neonatal outcomes. Drug Saf 30(3):247–264, 2007 17343431

Costei AM, Kozer E, Ho T, et al: Perinatal outcome following third trimester exposure to paroxetine. Arch Pediatr Adolesc Med 156(11):1129–1132, 2002 12413342

Croen LA, Grether JK, Yoshida CK, et al: Antidepressant use during pregnancy and childhood autism spectrum disorders. Arch Gen Psychiatry 68(11):1104–1112, 2011 21727247

Croke S, Buist A, Hackett LP, et al: Olanzapine excretion in human breast milk: estimation of infant exposure. Int J Neuropsychopharmacol 5(3):243–247, 2002 12366877

Cunnington MC, Weil JG, Messenheimer JA, et al: Final results from 18 years of the International Lamotrigine Pregnancy Registry. Neurology 76(21):1817–1823, 2011 21606453

Cutrona CE: Causal attributions and perinatal depression. J Abnorm Psychol 92(2):161–172, 1983 6863731

Dalens B, Raynaud EJ, Gaulme J: Teratogenicity of valproic acid. J Pediatr 97(2):332–333, 1980 6772753

Dallemagne G, Weiss B: Altered adult behavior of mice following postnatal treatment with haloperidol. Pharmacol Biochem Behav 16(5):761–767, 1982 7089034

Dam M, Christiansen J, Munck O, et al: Antiepileptic drugs: metabolism in pregnancy. Clin Pharmacokinet 4(1):53–62, 1979 421411

Dean JC, Moore SJ, Osborne A, et al: Fetal anticonvulsant syndrome and mutation in the maternal MTHFR gene. Clin Genet 56(3):216–220, 1999 10563481

de Haan GJ, Edelbroek P, Segers J, et al: Gestation-induced changes in lamotrigine pharmacokinetics: a monotherapy study. Neurology 63(3):571–573, 2004 15304599

Dev V, Krupp P: Adverse event profile and safety of clozapine. Reviews in Contemporary Pharmacotherapy 6(4):197–208, 1995

Diav-Citrin O, Shechtman S, Tahover E, et al: Pregnancy outcome following in utero exposure to lithium: a prospective, comparative, observational study. Am J Psychiatry 171(7):785–794, 2014 24781368

DiLiberti JH, Farndon PA, Dennis NR, et al: The fetal valproate syndrome. Am J Med Genet 19(3):473–481, 1984 6439041

Di Michele V, Ramenghi L, Sabatino G: Clozapine and lorazepam administration in pregnancy. Eur Psychiatry 11(4):214, 1996 19698455

Dickinson RG, Harland RC, Lynn RK, et al: Transmission of valproic acid (Depakene) across the placenta: half-life of the drug in mother and baby. J Pediatr 94(5):832–835, 1979 376803

Dickson RA, Hogg L: Pregnancy of a patient treated with clozapine. Psychiatr Serv 49(8):1081–1083, 1998 9712217

Djulus J, Koren G, Einarson TR, et al: Exposure to mirtazapine during pregnancy: a prospective, comparative study of birth outcomes. J Clin Psychiatry 67(8):1280–1284, 2006 16965209

Dodd S, Stocky A, Buist A, et al: Sertraline in paired blood plasma and breast-milk samples from nursing mothers. Hum Psychopharmacol 15(4):161–264, 2000 12404316

Dolk H, Jentink J, Loane M, et al; EUROCAT Antiepileptic Drug Working Group: Does lamotrigine use in pregnancy increase orofacial cleft risk relative to other malformations? Neurology 71(10):714–722, 2008 18650491

Dominguez-Salgado M, Morales A, Santiago Gomez R, et al: Gestational lamotrigine monotherapy: congenital malformations and psychomotor development (abstract). Epilepsia 45 (suppl 7):229–230, 2004

Downey G, Coyne JC: Children of depressed parents: an integrative review. Psychol Bull 108(1):50–76, 1990 2200073

Duffy CL: Postpartum depression: identifying women at risk. Genesis 11:21, 1983

Dursun A, Karadağ N, Karagöl B, et al: Carbamazepine use in pregnancy and coincidental thalidomide-like phocomelia in a newborn. J Obstet Gynaecol 32(5):488–489, 2012 22663328

Ebbesen F, Joergensen A, Hoseth E, et al: Neonatal hypoglycaemia and withdrawal symptoms after exposure in utero to valproate. Arch Dis Child Fetal Neonatal Ed 83(2):F124–F129, 2000 10952707

Edlund MJ, Craig TJ: Antipsychotic drug use and birth defects: an epidemiologic reassessment. Compr Psychiatry 25(1):32–37, 1984 6141893

Eggermont E: Withdrawal symptoms in neonates associated with maternal imipramine therapy. Lancet 2(7830):680, 1973 4125653

Einarson A, Fatoye B, Sarkar M, et al: Pregnancy outcome following gestational exposure to venlafaxine: a multicenter prospective controlled study. Am J Psychiatry 158(10):1728–1730, 2001 11579012

Einarson A, Bonari L, Voyer-Lavigne S, et al: A multicentre prospective controlled study to determine the safety of trazodone and nefazodone use during pregnancy. Can J Psychiatry 48(2):106–110, 2003 12655908

Einarson A, Smart K, Vial T, et al: Rates of major malformations in infants following exposure to duloxetine during pregnancy: a preliminary report. J Clin Psychiatry 73(11):1471, 2012 23218163

Einarson TR, Einarson A: Newer antidepressants in pregnancy and rates of major malformations: a meta-analysis of prospective comparative studies. Pharmacoepidemiol Drug Saf 14(12):823–827, 2005 15742359

Elia J, Katz IR, Simpson GM: Teratogenicity of psychotherapeutic medications. Psychopharmacol Bull 23(4):531–586, 1987 2893424

El Marroun H, White TJH, van der Knaap NJF, et al: Prenatal exposure to selective serotonin reuptake inhibitors and social responsiveness symptoms of autism: population-based study of young children. Br J Psychiatry 205(2):95–102, 2014 25252317

Engelhard IM, van den Hout MA, Arntz A: Posttraumatic stress disorder after pregnancy loss. Gen Hosp Psychiatry 23(2): 62–66, 2001 11313072

Ennis ZN, Damkier P: Pregnancy exposure to olanzapine, quetiapine, risperidone, aripiprazole and risk of congenital malformations: a systematic review. Basic Clin Pharmacol Toxicol 116(4):315–320, 2015 25536446

Entman SS, Vaughn WK: Lack of relation of oral clefts to diazepam use in pregnancy. N Engl J Med 310(17):1121–1122, 1984 6708998

Epperson CN, Anderson GM, McDougle CJ: Sertraline and breast-feeding. N Engl J Med 336(16):1189–1190, 1997 9102576

Epperson N, Czarkowski KA, Ward-O’Brien D, et al: Maternal sertraline treatment and serotonin transport in breast-feeding mother-infant pairs. Am J Psychiatry 158(10):1631–1637, 2001 11578995

Erkkola R, Kanto J, Sellman R: Diazepam in early human pregnancy. Acta Obstet Gynecol Scand 53(2):135–138, 1974 4822345

Erkkola R, Kero P, Kanto J, et al: Severe abuse of psychotropic drugs during pregnancy with good perinatal outcome. Ann Clin Res 15(2):88–91, 1983 6881907

Eros E, Czeizel AE, Rockenbauer M, et al: A population-based case-control teratologic study of nitrazepam, medazepam, tofisopam, alprazolum and clonazepam treatment during pregnancy. Eur J Obstet Gynecol Reprod Biol 101(2):147–154, 2002 11858890

Falterman CG, Richardson CJ: Small left colon syndrome associated with maternal ingestion of psychotropic drugs. J Pediatr 97(2):308–310, 1980 7400907

Finer LB, Zolna MR: Declines in unintended pregnancy in the United States, 2008–2011. N Engl J Med 374(9):843–852, 2016 26962904

Fisher JB, Edgren BE, Mammel MC, et al: Neonatal apnea associated with maternal clonazepam therapy: a case report. Obstet Gynecol 66 (3 suppl):34S–35S, 1985 4022513

Fones C: Posttraumatic stress disorder occurring after painful childbirth. J Nerv Ment Dis 184(3):195–196, 1996 8600227

Fotopoulou C, Kretz R, Bauer S, et al: Prospectively assessed changes in lamotrigine-concentration in women with epilepsy during pregnancy, lactation and the neonatal period. Epilepsy Res 85(1):60–64, 2009 19272754

Franco V, Mazzucchelli I, Gatti G, et al: Changes in lamotrigine pharmacokinetics during pregnancy and the puerperium. Ther Drug Monit 30(4):544–547, 2008 18641557

Frederiksen MC: Physiologic changes in pregnancy and their effect on drug disposition. Semin Perinatol 25(3):120–123, 2001 11453606

Freeman MP, Nolan PE Jr, Davis MF, et al: Pharmacokinetics of sertraline across pregnancy and postpartum. J Clin Psychopharmacol 28(6):646–653, 2008 19011433

Frey B, Schubiger G, Musy JP: Transient cholestatic hepatitis in a neonate associated with carbamazepine exposure during pregnancy and breast-feeding. Eur J Pediatr 150(2):136–138, 1990 2279511

Frey B, Braegger CP, Ghelfi D: Neonatal cholestatic hepatitis from carbamazepine exposure during pregnancy and breast feeding. Ann Pharmacother 36(4):644–647, 2002 11918515

Frieder B, Epstein S, Grimm VE: The effects of exposure to diazepam during various stages of gestation or during lactation on the development and behavior of rat pups. Psychopharmacology (Berl) 83(1): 51–55, 1984 6429700

Friedman JM, Polifka JE: Teratogenic Effects of Drugs: A Resource for Clinicians (TERIS), 2nd Edition. Baltimore, MD, Johns Hopkins University Press, 2000, pp ix–x

Friedman SH, Rosenthal MB: Treatment of perinatal delusional disorder: a case report. Int J Psychiatry Med 33(4):391–394, 2003 15152788

Fries H: Lithium in pregnancy. Lancet 1(7658):1233, 1970 4192416

Froescher W, Eichelbaum M, Nieson M, et al: Antiepileptic therapy with carbamazepine and valproic acid during pregnancy and the lactation period, in Advances in Epileptology: The 12th Epilepsy International Symposium. Edited by Dam M, Gram L, Penry JK. New York, Raven, 1981, pp 581–588

Froescher W, Eichelbaum M, Niesen M, et al: Carbamazepine levels in breast milk. Ther Drug Monit 6(3):266–271, 1984a 6390794

Froescher W, Gugler R, Niesen M, Hoffmann F: Protein binding of valproic acid in maternal and umbilical cord serum. Epilepsia 25(2):244–249, 1984b 6423379

Gaily E, Kantola-Sorsa E, Granström ML: Specific cognitive dysfunction in children with epileptic mothers. Dev Med Child Neurol 32(5):403–414, 1990 2113015

Galler JR, Harrison RH, Ramsey F, et al: Maternal depressive symptoms affect infant cognitive development in Barbados. J Child Psychol Psychiatry 41(6):747–757, 2000 11039687

Gardiner SJ, Kristensen JH, Begg EJ, et al: Transfer of olanzapine into breast milk, calculation of infant drug dose, and effect on breast-fed infants. Am J Psychiatry 160(8):1428–1431, 2003 12900304

Gentile S, Tofani S, Bellantuono C: Aripiprazole and pregnancy: a case report and literature review. J Clin Psychopharmacol 31(4):531–532, 2011 21720228

Gerhardsson M, Alfredsson L: In-utero exposure to benzodiazepines. Lancet 1(8533): 628, 1987 2881162

Gidaya NB, Lee BK, Burstyn I, et al: In utero exposure to selective serotonin reuptake inhibitors and risk for autism spectrum disorder. J Autism Dev Disord 44(10): 2558–2567, 2014 24803368

Gilad O, Merlob P, Stahl B, Klinger G: Outcome of infants exposed to olanzapine during breastfeeding. Breastfeed Med 6(2):55–58, 2011 21034242

GlaxoSmithKline: Epidemiology study: Updated preliminary report on bupropion and other antidepressants, including paroxetine in pregnancy and the occurrence of cardiovascular and major congenital malformations (study EPIP083). 2005. Available at: http://www.gsk-clinicalstudyregister.com/files2/3493.pdf. Accessed June 7, 2016.

GlaxoSmithKline: International Pregnancy Registry: Interim Report (1 September 1992 through 31 March 2007). Wilmington, NC, Inveresk, 2007

Glaze R, Chapman G, Murray D: Recurrence of puerperal psychosis during late pregnancy. Br J Psychiatry 159:567–569, 1991 1751870

Goldberg HL, DiMascio A: Psychotropic drugs in pregnancy, in Psychopharmacology: A Generation of Progress. Edited by Lipton HL, DiMascio A, Killam KF. New York, Raven, 1978, pp 1047–1055

Goldstein DJ, Sundell KL, Corbin LA: Birth outcomes in pregnant women taking fluoxetine. N Engl J Med 336(12):872–873, author reply 873, 1997 9072683

Goldstein DJ, Corbin LA, Fung MC: Olanzapine-exposed pregnancies and lactation: early experience. J Clin Psychopharmacol 20(4):399–403, 2000 10917399

Grigoriadis S, VonderPorten EH, Mamisashvili L, et al: Antidepressant exposure during pregnancy and congenital malformations: is there an association? A systematic review and meta-analysis of the best evidence. J Clin Psychiatry 74(4): e293–e308, 2013 23656855

Grover S, Madan R: Successful use of quetiapine in two successive pregnancies. J Neuropsychiatry Clin Neurosci 24(1):E38, 2012 22450643

Guglielmi V, Vulink NC, Denys D, et al: Obsessive-compulsive disorder and female reproductive cycle events: results from the OCD and reproduction collaborative study. Depress Anxiety 31(12):979–987, 2014 24421066

Haram K: “Floppy infant syndrome” and maternal diazepam. Lancet 2(8038):612–613, 1977 71430

Harrington RA, Lee LC, Crum RM, et al: Prenatal SSRI use and offspring with autism spectrum disorder or developmental delay. Pediatrics 133(5):e1241–e1248, 2014 24733881

Hartz SC, Heinonen OP, Shapiro S, et al: Antenatal exposure to meprobamate and chlordiazepoxide in relation to malformations, mental development, and childhood mortality. N Engl J Med 292(14): 726–728, 1975 1113782

Hassmannova J, Myslivecek J: Inhibitory and excitatory adult learning after prenatal diazepam application. Studia Psychologica (Bratislava) 36(5):323–326, 1994

Hedegaard M, Henriksen TB, Sabroe S, et al: The relationship between psychological distress during pregnancy and birth weight for gestational age. Acta Obstet Gynecol Scand 75(1):32–39, 1996 8560994

Heikkinen T, Ekblad U, Kero P, et al: Citalopram in pregnancy and lactation. Clin Pharmacol Ther 72(2):184–191, 2002 12189365

Hendrick V, Fukuchi A, Altshuler L, et al: Use of sertraline, paroxetine and fluvoxamine by nursing women. Br J Psychiatry 179:163–166, 2001a 11483479

Hendrick V, Stowe ZN, Altshuler LL, et al: Fluoxetine in nursing infants and breast milk. Biol Psychiatry 50:775–782, 2001b 11720696

Hendrick V, Stowe ZN, Altshuler LL, et al: Placental passage of antidepressant medications. Am J Psychiatry 160(5):993–996, 2003 12727706

Hernández-Díaz S, Werler MM, Walker AM, et al: Neural tube defects in relation to use of folic acid antagonists during pregnancy. Am J Epidemiol 153(10):961–968, 2001 11384952

Hertzberg T, Wahlbeck K: The impact of pregnancy and puerperium on panic disorder: a review. J Psychosom Obstet Gynaecol 20(2):59–64, 1999 10422037

Hill RC, McIvor RJ, Wojnar-Horton RE, et al: Risperidone distribution and excretion into human milk: case report and estimated infant exposure during breast-feeding. J Clin Psychopharmacol 20(2): 285–286, 2000 10770482

Hill RM, Stern L: Drugs in pregnancy: effects on the fetus and newborn. Drugs 17(3):182–197, 1979 88321

Hill RM, Desmond MM, Kay JL: Extrapyramidal dysfunction in an infant of a schizophrenic mother. J Pediatr 69(4):589–595, 1966 5921334

Hirsch MS, Swartz MN: Drug therapy: antiviral agents (first of two parts). N Engl J Med 302(16):903–907, 1980 6987519

Hoffeld DR, McNew J, Webster RL: Effect of tranquilizing drugs during pregnancy on activity of offspring. Nature 218(5139): 357–358, 1968 5649675

Holmes LB, Wyszynski DF, Baldwin EJ, et al: Increased risk for non-syndromic cleft palate among infants exposed to lamotrigine during pregnancy (abstract). Birth Defects Res A Clin Mol Teratol 76(5):318, 2006

Hostetter A, Stowe ZN, Strader JR Jr, et al: Dose of selective serotonin uptake inhibitors across pregnancy: clinical implications. Depress Anxiety 11(2):51–57, 2000 10812529

House SJ, Tripathi SP, Knight BT, et al: Obsessive-compulsive disorder in pregnancy and the postpartum period: course of illness and obstetrical outcome. Arch Women Ment Health 19(1):3–10, 2016 26173597

Huang H, Coleman S, Bridge JA, et al: A meta-analysis of the relationship between antidepressant use in pregnancy and the risk of preterm birth and low birth weight. Gen Hosp Psychiatry 36(1):13–18, 2014 24094568

Huybrechts KF, Palmsten K, Avorn J, et al: Antidepressant use in pregnancy and the risk of cardiac defects. N Engl J Med 370(25):2397–2407, 2014a 24941178

Huybrechts KF, Sanghani RS, Avorn J, Urato AC: Preterm birth and antidepressant medication use during pregnancy: a systematic review and meta-analysis. PLoS One 9(3):e92778, 2014b 24671232

Huybrechts KF, Bateman BT, Palmsten K, et al: Antidepressant use late in pregnancy and risk of persistent pulmonary hypertension of the newborn. JAMA 313(21):2142–2151, 2015 26034955

Hviid A, Melbye M, Pasternak B: Use of selective serotonin reuptake inhibitors during pregnancy and risk of autism. N Engl J Med 369(25):2406–2415, 2013 24350950

Ilett KF, Hackett LP, Dusci LJ, et al: Distribution and excretion of venlafaxine and O-desmethylvenlafaxine in human milk. Br J Clin Pharmacol 45(5):459–462, 1998 9643618

Ilett KF, Hackett LP, Kristensen JH, et al: Transfer of risperidone and 9-hydroxyrisperidone into human milk. Ann Pharmacother 38(2):273–276, 2004 14742766

Jacobson SJ, Jones K, Johnson K, et al: Prospective multicentre study of pregnancy outcome after lithium exposure during first trimester. Lancet 339(8792):530–533, 1992 1346886

Jäger-Roman E, Deichl A, Jakob S, et al: Fetal growth, major malformations, and minor anomalies in infants born to women receiving valproic acid. J Pediatr 108(6):997–1004, 1986 3086531

Jaiswal AK, Bhattacharya SK: Effects of gestational undernutrition, stress and diazepam treatment on spatial discrimination learning and retention in young rats. Indian J Exp Biol 31(4):353–359, 1993 8359834

James ME: Neuroleptic malignant syndrome in pregnancy. Psychosomatics 29(1):119–122, 1988 3340699

Jameson PB, Gelfand DM, Kulcsar E, et al: Mother-toddler interaction patterns associated with maternal depression. Dev Psychopathol 9(3):537–550, 1997 9327238

Jenike MA, Hyman S, Baer L, et al: A controlled trial of fluvoxamine in obsessive-compulsive disorder: implications for a serotonergic theory. Am J Psychiatry 147(9):1209–1215, 1990 2143637

Jensen PN, Olesen OV, Bertelsen A, et al: Citalopram and desmethylcitalopram concentrations in breast milk and in serum of mother and infant. Ther Drug Monit 19(2):236–239, 1997 9108657

Jentink J, Dolk H, Loane MA, et al; EUROCAT Antiepileptic Study Working Group: Intrauterine exposure to carbamazepine and specific congenital malformations: systematic review and case-control study. BMJ 341:c6581, 2010 21127116

Johnson EL, Stowe ZN, Ritchie JC, et al: Carbamazepine clearance and seizure stability during pregnancy. Epilepsy Behav 33:49–53, 2014 24632353

Jones KL, Lacro RV, Johnson KA, et al: Pattern of malformations in the children of women treated with carbamazepine during pregnancy. N Engl J Med 320(25): 1661–1666, 1989 2725616

Källén B: Neonate characteristics after maternal use of antidepressants in late pregnancy. Arch Pediatr Adolesc Med 158(4): 312–316, 2004 15066868

Källén B, Olausson PO: Maternal use of selective serotonin re-uptake inhibitors and persistent pulmonary hypertension of the newborn. Pharmacoepidemiol Drug Saf 17(8):801–806, 2008 18314924

Källén B, Reis M: Neonatal complications after maternal concomitant use of SSRI and other central nervous system active drugs during the second or third trimester of pregnancy. J Clin Psychopharmacol 32(5):608–614, 2012 22926593

Källén B, Tandberg A: Lithium and pregnancy: a cohort study on manic-depressive women. Acta Psychiatr Scand 68(2):134–139, 1983 6624510

Kaneko S, Suzuki K, Sato T, et al: The problems of antiepileptic medication in the neonatal period: is breast-feeding advisable? in Epilepsy, Pregnancy and the Child. Edited by Janz D, Dam M, Richens A. New York, Raven, 1982, pp 343–348

Kaneko S, Battino D, Andermann E, et al: Congenital malformations due to antiepileptic drugs. Epilepsy Res 33(2–3):145–158, 1999 10094426

Karlsson K, Lindstedt G, Lundberg PA, et al: Letter: Transplacental lithium poisoning: reversible inhibition of fetal thyroid. Lancet 1(7919):1295, 1975 48921

Kelly LE, Poon S, Madadi P, et al: Neonatal benzodiazepines exposure during breastfeeding. J Pediatr 161(3):448–451, 2012 22504099

Kendell RE, Chalmers JC, Platz C: Epidemiology of puerperal psychoses. Br J Psychiatry 150:662–673, 1987 3651704

Kendler KS, Kessler RC, Neale MC, et al: The prediction of major depression in women: toward an integrated etiologic model. Am J Psychiatry 150(8):1139–1148, 1993 8328557

Kennedy D, Koren G: Valproic acid use in psychiatry: issues in treating women of reproductive age. J Psychiatry Neurosci 23(4):223–228, 1998 9785701

Kirchheiner J, Berghöfer A, Bolk-Weischedel D: Healthy outcome under olanzapine treatment in a pregnant woman. Pharmacopsychiatry 33(2):78–80, 2000 10761825

Kirksey A, Groziak SM: Maternal drug use: evaluation of risks to breast-fed infants. World Rev Nutr Diet 43:60–79, 1984 6147939

Klier CM, Mossaheb N, Lee A, et al: Mirtazapine and breastfeeding: maternal and infant plasma levels. Am J Psychiatry 164(2):348–349, 2007 17267804

Koch S, Jäger-Roman E, Rating D, Helge H: Possible teratogenic effect of valproate during pregnancy. J Pediatr 103(6):1007–1008, 1983 6417292

Kok TH, Taitz LS, Bennett MJ, Holt DW: Drowsiness due to clemastine transmitted in breast milk. Lancet 1(8277):914–915, 1982 6122135

Korebrits C, Ramirez MM, Watson L, et al: Maternal corticotropin-releasing hormone is increased with impending preterm birth. J Clin Endocrinol Metab 83(5):1585–1591, 1998 9589660

Kornhuber J, Weller M: Postpartum psychosis and mastitis: a new indication for clozapine? Am J Psychiatry 148(12):1751–1752, 1991 1796996

Kost K: Unintended pregnancy rates at the state level: estimates for 2010 and trends since 2002. Guttmacher Institute, 2015. Available at: http://www.guttmacher.org/pubs/StateUP10.pdf. Accessed June 7, 2016.

Kozma C: Valproic acid embryopathy: report of two siblings with further expansion of the phenotypic abnormalities and a review of the literature. Am J Med Genet 98(2):168–175, 2001 11223853

Kriel RL, Cloyd J: Clonazepam and pregnancy. Ann Neurol 11(5):544, 1982 7103432

Kris EB: Children of mothers maintained on pharmacotherapy during pregnancy and postpartum. Curr Ther Res Clin Exp 7(12):785–789, 1965 4954450

Kris EB, Carmichael DM: Chlorpromazine maintenance therapy during pregnancy and confinement. Psychiatr Q 31(4):690–695, 1957 13518422

Kristensen JH, Ilett KF, Dusci LJ, et al: Distribution and excretion of sertraline and N-desmethylsertraline in human milk. Br J Clin Pharmacol 45(5):453–457, 1998 9643617

Kristensen JH, Ilett KF, Hackett LP, et al: Distribution and excretion of fluoxetine and norfluoxetine in human milk. Br J Clin Pharmacol 48(4):521–527, 1999 10583022

Kristensen JH, Ilett KF, Rampono J, et al: Transfer of the antidepressant mirtazapine into breast milk. Br J Clin Pharmacol 63(3):322–327, 2007 16970569

Kuhnz W, Jäger-Roman E, Rating D, et al: Carbamazepine and carbamazepine-10,11-epoxide during pregnancy and postnatal period in epileptic mother and their nursed infants: pharmacokinetics and clinical effects. Pediatr Pharmacol (New York) 3(3–4):199–208, 1983 6677873

Kulin NA, Pastuszak A, Sage SR, et al: Pregnancy outcome following maternal use of the new selective serotonin reuptake inhibitors: a prospective controlled multicenter study. JAMA 279(8):609–610, 1998 9486756

Kumar R, Robson KM: A prospective study of emotional disorders in childbearing women. Br J Psychiatry 144:35–47, 1984 6692075

Laegreid L, Olegård R, Wahlström J, et al: Abnormalities in children exposed to benzodiazepines in utero. Lancet 1(8524):108–109, 1987 2879165

Laine K, Heikkinen T, Ekblad U, et al: Effects of exposure to selective serotonin reuptake inhibitors during pregnancy on serotonergic symptoms in newborns and cord blood monoamine and prolactin concentrations. Arch Gen Psychiatry 60(7):720–726, 2003 12860776

Lajeunie E, Le Merrer M, Marchac D, et al: Syndromal and nonsyndromal primary trigonocephaly: analysis of a series of 237 patients. Am J Med Genet 75(2):211–215, 1998 9450889

Lajeunie E, Barcik U, Thorne JA, et al: Craniosynostosis and fetal exposure to sodium valproate. J Neurosurg 95(5):778–782, 2001 11702867

Lander CM, Livingstone I, Tyrer JH, et al: The clearance of anticonvulsant drugs in pregnancy. Clin Exp Neurol 17:71–78, 1981 7346203

Lee A, Giesbrecht E, Dunn E, et al: Excretion of quetiapine in breast milk. Am J Psychiatry 161(9):1715–1716, 2004 15337669

Lester BM, Cucca J, Andreozzi L, et al: Possible association between fluoxetine hydrochloride and colic in an infant. J Am Acad Child Adolesc Psychiatry 32(6):1253–1255, 1993 8282672

Lindhout D, Schmidt D: In-utero exposure to valproate and neural tube defects. Lancet 1(8494):1392–1393, 1986 2872511

Liporace J, Kao A, D’Abreu A: Concerns regarding lamotrigine and breast-feeding. Epilepsy Behav 5(1):102–105, 2004 14751214

Little BB: Pharmacokinetics during pregnancy: evidence-based maternal dose formulation. Obstet Gynecol 93(5 Pt 2):858–868, 1999 10912434

Liu C, Cnattingius S, Bergström M, et al: Prenatal parental depression and preterm birth: a national cohort study. BJOG 123(12):1973–1982, 2016 26786413

Llewellyn A, Stowe ZN, Strader JR Jr: The use of lithium and management of women with bipolar disorder during pregnancy and lactation. J Clin Psychiatry 59 (suppl 6):57–64, discussion 65, 1998 9674938

Lobo ED, Loghin C, Knadler MP, et al: Pharmacokinetics of duloxetine in breast milk and plasma of healthy postpartum women. Clin Pharmacokinet 47(2):103–109, 2008 18193916

Louik C, Lin AE, Werler MM, et al: First-trimester use of selective serotonin-reuptake inhibitors and the risk of birth defects. N Engl J Med 356(26):2675–2683, 2007 17596601

Luoma I, Tamminen T, Kaukonen P, et al: Longitudinal study of maternal depressive symptoms and child well-being. J Am Acad Child Adolesc Psychiatry 40(12):1367–1374, 2001 11765281

Luoma I, Kaukonen P, Mäntymaa M, et al: A longitudinal study of maternal depressive symptoms, negative expectations and perceptions of child problems. Child Psychiatry Hum Dev 35(1):37–53, 2004 15626324

Lupattelli A, Spigset O, Björnsdóttir I, et al: Patterns and factors associated with low adherence to psychotropic medications during pregnancy—a cross-sectional, multinational Web-based study. Depress Anxiety 32(6):426–436, 2015 25703355

Lutz UC, Hiemke C, Wiatr G, et al: Aripiprazole in pregnancy and lactation: a case report. J Clin Psychopharmacol 30(2):204–205, 2010 20520299

Lyons-Ruth K, Wolfe R, Lyubchik A: Depression and the parenting of young children: making the case for early preventive mental health services. Harv Rev Psychiatry 8(3):148–153, 2000 10973939

Mammen OK, Perel JM, Rudolph G, et al: Sertraline and norsertraline levels in three breastfed infants. J Clin Psychiatry 58(3):100–103, 1997 9108810

Mandelli M, Morselli PL, Nordio S, et al: Placental transfer to diazepam and its disposition in the newborn. Clin Pharmacol Ther 17(5):564–572, 1975 1126114

Manly PC, McMahon RJ, Bradley CF, et al: Depressive attributional style and depression following childbirth. J Abnorm Psychol 91(4):245–254, 1982 7130519

Marcus SM, Flynn HA, Blow FC, et al: Depressive symptoms among pregnant women screened in obstetrics settings. J Womens Health (Larchmt) 12(4):373–380, 2003 12804344

Martinez A, Malphurs J, Field T, et al: Depressed mothers’ and their infants’ interactions with nondepressed partners. Infant Mental Health Journal 17(1):74–80, 1996

Martínez-Frías ML: Clinical manifestation of prenatal exposure to valproic acid using case reports and epidemiologic information. Am J Med Genet 37(2):277–282, 1990 2248297

Martins C, Gaffan EA: Effects of early maternal depression on patterns of infant-mother attachment: a meta-analytic investigation. J Child Psychol Psychiatry 41(6):737–746, 2000 11039686

Maschi S, Clavenna A, Campi R, et al: Neonatal outcome following pregnancy exposure to antidepressants: a prospective controlled cohort study. BJOG 115(2):283–289, 2008 17903222

Matalon S, Schechtman S, Goldzweig G, et al: The teratogenic effect of carbamazepine: a meta-analysis of 1255 exposures. Reprod Toxicol 16(1):9–17, 2002 11934528

Matheson I, Skjaeraasen J: Milk concentrations of flupenthixol, nortriptyline and zuclopenthixol and between-breast differences in two patients. Eur J Clin Pharmacol 35(2):217–220, 1988 3191943

Matheson I, Pande H, Alertsen AR: Respiratory depression caused by N-desmethyldoxepin in breast milk. Lancet 2(8464): 1124, 1985 2865592

Mazzi E: Possible neonatal diazepam withdrawal: a case report. Am J Obstet Gynecol 129(5):586–587, 1977 910847

McAuley DM, O’Neill MP, Moore J, et al: Lorazepam premedication for labour. Br J Obstet Gynaecol 89(2):149–154, 1982 6121581

McBride WG: Limb deformities associated with iminodibenzyl hydrochloride. Med J Aust 1(10):492, 1972 5024423

McDonagh M, Matthews A, Phillipi C, et al: Antidepressant treatment of depression during pregnancy and the postpartum period. Evidence Report/Technology Assessment No 216 (AHRQ Publ No 14-E003-EF). 2014. Available at: www.effectivehealthcare.ahrq.gov/reports/final.cfm. Accessed June 7, 2016.

McElhatton PR, Garbis HM, Eléfant E, et al: The outcome of pregnancy in 689 women exposed to therapeutic doses of antidepressants. A collaborative study of the European Network of Teratology Information Services (ENTIS). Reprod Toxicol 10(4):285–294, 1996 8829251

McEvoy JP, Hatcher A, Appelbaum PS, et al: Chronic schizophrenic women’s attitudes toward sex, pregnancy, birth control, and childrearing. Hosp Community Psychiatry 34(6):536–539, 1983 6862399

McKenna K, Koren G, Tetelbaum M, et al: Pregnancy outcome of women using atypical antipsychotic drugs: a prospective comparative study. J Clin Psychiatry 66(4):444–449, quiz 546, 2005 15816786

McMahon CL, Braddock SR: Septo-optic dysplasia as a manifestation of valproic acid embryopathy. Teratology 64(2):83–86, 2001 11460259

McNeil TF, Kaij L, Malmquist-Larsson A: Women with nonorganic psychosis: mental disturbance during pregnancy. Acta Psychiatr Scand 70(2):127–139, 1984a 6485846

McNeil TF, Kaij L, Malmquist-Larsson A: Women with nonorganic psychosis: pregnancy’s effect on mental health during pregnancy. Acta Psychiatr Scand 70(2): 140–148, 1984b 6485847

McVearry KM, Gaillard WD, VanMeter J, et al: A prospective study of cognitive fluency and originality in children exposed in utero to carbamazepine, lamotrigine, or valproate monotherapy. Epilepsy Behav 16(4):609–616, 2009 19892603

Meador KJ, Baker GA, Finnell RH, et al; NEAD Study Group: In utero antiepileptic drug exposure: fetal death and malformations. Neurology 67(3):407–412, 2006 16894099

Meador KJ, Baker GA, Browning N, et al; NEAD Study Group: Cognitive function at 3 years of age after fetal exposure to antiepileptic drugs. N Engl J Med 360(16):1597–1605, 2009 19369666

Meador KJ, Baker GA, Browning N, et al; NEAD Study Group: Foetal antiepileptic drug exposure and verbal versus non-verbal abilities at three years of age. Brain 134(pt 2):396–404, 2011 21224309

Meador KJ, Baker GA, Browning N, et al; NEAD Study Group: Fetal antiepileptic drug exposure and cognitive outcomes at age 6 years (NEAD study): a prospective observational study. Lancet Neurol 12(3):244–252, 2013 23352199

Meador KJ, Baker GA, Browning N, et al; Neurodevelopmental Effects of Antiepileptic Drugs (NEAD) Study Group: Breastfeeding in children of women taking antiepileptic drugs: cognitive outcomes at age 6 years. JAMA Pediatr 168(8):729–736, 2014 24934501

Meijer A: Child psychiatric sequelae of maternal war stress. Acta Psychiatr Scand 72(6):505–511, 1985 2417452

Merlob P, Mor N, Litwin A: Transient hepatic dysfunction in an infant of an epileptic mother treated with carbamazepine during pregnancy and breastfeeding. Ann Pharmacother 26(12):1563–1565, 1992 1362364

Milkovich L, van den Berg BJ: An evaluation of the teratogenicity of certain antinauseant drugs. Am J Obstet Gynecol 125(2):244–248, 1976 773181

Miller ES, Chu C, Gollan J, et al: Obsessive-compulsive symptoms during the postpartum period: a prospective cohort. J Reprod Med 58(3–4):115–122, 2013 23539879

Miller LJ: Clinical strategies for the use of psychotropic drugs during pregnancy. Psychiatr Med 9(2):275–298, 1991 1677481

Miller LJ, Finnerty M: Sexuality, pregnancy, and childrearing among women with schizophrenia-spectrum disorders. Psychiatr Serv 47(5):502–506, 1996 8740491

Misri S, Sivertz K: Tricyclic drugs in pregnancy and lactation: a preliminary report. Int J Psychiatry Med 21(2):157–171, 1991 1894455

Misri S, Oberlander TF, Fairbrother N, et al: Relation between prenatal maternal mood and anxiety and neonatal health. Can J Psychiatry 49(10):684–689, 2004 15560315

Mitchell AA, Gilboa SM, Werler MM, et al; National Birth Defects Prevention Study: Medication use during pregnancy, with particular focus on prescription drugs: 1976–2008. Am J Obstet Gynecol 205(1): 51.e1–51.e8, 2011 21514558

Mizrahi EM, Hobbs JF, Goldsmith DI: Nephrogenic diabetes insipidus in transplacental lithium intoxication. J Pediatr 94(3):493–495, 1979 423043

Mølgaard-Nielsen D, Hviid A: Newer-generation antiepileptic drugs and the risk of major birth defects. JAMA 305(19):1996–2002, 2011 21586715

Moore SJ, Turnpenny P, Quinn A, et al: A clinical study of 57 children with fetal anticonvulsant syndromes. J Med Genet 37(7):489–497, 2000 10882750

Moore WM, Hellegers AE, Battaglia FC: In vitro permeability of different layers of the human placenta to carbohydrates and urea. Am J Obstet Gynecol 96(7):951–955, 1966 4959176

Moreno-Bruna MD, de Montgolfier I, Chaubaud M, et al: [Case report: Neonatal delayed peristalsis after in-utero exposure to clozapine] [Article in French]. Arch Pediatr 19:913–916, 2012 22884999

Morgan DJ: Drug disposition in mother and foetus. Clin Exp Pharmacol Physiol 24(11):869–873, 1997 9363372

Moriarty AJ, Nance MR: Trifluoperazine and Pregnancy. Can Med Assoc J 88(7):375–376, 1963 20327411

Morrison CM: A case report of the use of vilazodone in pregnancy. Prim Care Companion CNS Disord 16(2):pii:PCC. 13l01612, 2014 25133058

Morrow J, Russell A, Guthrie E, et al: Malformation risks of antiepileptic drugs in pregnancy: a prospective study from the UK Epilepsy and Pregnancy Register. J Neurol Neurosurg Psychiatry 77(2):193–198, 2006 16157661

Moses-Kolko EL, Bogen D, Perel J, et al: Neonatal signs after late in utero exposure to serotonin reuptake inhibitors: literature review and implications for clinical applications. JAMA 293(19):2372–2383, 2005 15900008

Mountain KR, Hirsh J, Gallus AS: Neonatal coagulation defect due to anticonvulsant drug treatment in pregnancy. Lancet 1(7641):265–268, 1970 4189292

Myllynen PK, Pienimäki PK, Vähäkangas KH: Transplacental passage of lamotrigine in a human placental perfusion system in vitro and in maternal and cord blood in vivo. Eur J Clin Pharmacol 58(10):677–682, 2003 12610744

Myslivecek J, Hassmannová J, Josífko M: Impact of prenatal low-dose diazepam or chlorpromazine on reflex and motor development and inhibitory-learning. Homeost Health Dis 33(1–2):77–88, 1991 1817694

Nadebaum C, Anderson V, Vajda F, et al: The Australian brain and cognition and antiepileptic drugs study: IQ in school-aged children exposed to sodium valproate and polytherapy. J Int Neuropsychol Soc 17(1):133–142, 2011a 21092354

Nadebaum C, Anderson VA, Vajda F, et al: Language skills of school-aged children prenatally exposed to antiepileptic drugs. Neurology 76(8):719–726, 2011b 21339499

Nau H, Rating D, Koch S, et al: Valproic acid and its metabolites: placental transfer, neonatal pharmacokinetics, transfer via mother’s milk and clinical status in neonates of epileptic mothers. J Pharmacol Exp Ther 219(3):768–777, 1981 6795343

Nau H, Kuhnz W, Egger HJ, et al: Anticonvulsants during pregnancy and lactation: transplacental, maternal and neonatal pharmacokinetics. Clin Pharmacokinet 7(6):508–543, 1982a 6819105

Nau H, Wittfoht W, Rating D, et al: Pharmacokinetics of valproic acid and its metabolites in a pregnant patient: stable isotope methodology, in Epilepsy, Pregnancy, and the Child. Edited by Janz D, Bossi L, Dam M, et al. New York, Raven, 1982b, pp 131–139

Newport DJ, Stowe ZN, Nemeroff CB: Parental depression: animal models of an adverse life event. Am J Psychiatry 159(8):1265–1283, 2002 12153816

Newport DJ, Viguera AC, Beach AJ, et al: Lithium placental passage and obstetrical outcome: implications for clinical management during late pregnancy. Am J Psychiatry 162(11):2162–2170, 2005 16263858

Newport DJ, Calamaras MR, DeVane CL, et al: Atypical antipsychotic administration during late pregnancy: placental passage and obstetrical outcomes. Am J Psychiatry 164(8):1214–1220, 2007a 17671284

Newport DJ, Levey LC, Pennell PB, et al: Suicidal ideation in pregnancy: assessment and clinical implications. Arch Women Ment Health 10(5):181–187, 2007b 17726640

Newport DJ, Brennan PA, Green P, et al: Maternal depression and medication exposure during pregnancy: comparison of maternal retrospective recall to prospective documentation. BJOG 115(6):681–688, 2008a 18410650

Newport DJ, Pennell PB, Calamaras MR, et al: Lamotrigine in breast milk and nursing infants: determination of exposure. Pediatrics 122(1):e223–e231, 2008b 18591203

Newport DJ, Stowe ZN, Viguera AC, et al: Lamotrigine in bipolar disorder: efficacy during pregnancy. Bipolar Disord 10(3): 432–436, 2008c 18402631

Newport DJ, Ritchie JC, Knight BT, et al: Venlafaxine in human breast milk and nursing infant plasma: determination of exposure. J Clin Psychiatry 70(9):1304–1310, 2009 19607765

Newport DJ, Ji S, Long Q, et al: Maternal depression and anxiety differentially impact fetal exposures during pregnancy. J Clin Psychiatry 73(2):247–251, 2012 22152400

Newport DJ, Hostetter AL, Juul SH, et al: Prenatal psychostimulant and antidepressant exposure and risk of hypertensive disorders of pregnancy. J Clin Psychiatry 77(11):1538–1545, 2016 28076672

Niebyl JR, Blake DA, Freeman JM, et al: Carbamazepine levels in pregnancy and lactation. Obstet Gynecol 53(1):139–140, 1979 760015

Nora JJ, Nora AH, Toews WH: Letter: Lithium, Ebstein’s anomaly, and other congenital heart defects. Lancet 2(7880):594–595, 1974 4140306

Nordmo E, Aronsen L, Wasland K, et al: Severe apnea in an infant exposed to lamotrigine in breast milk. Ann Pharmacother 43(11):1893–1897, 2009 19826099

Nulman I, Rovet J, Stewart DE, et al: Neurodevelopment of children exposed in utero to antidepressant drugs. N Engl J Med 336(4):258–262, 1997a 8995088

Nulman I, Scolnik D, Chitayat D, et al: Findings in children exposed in utero to phenytoin and carbamazepine monotherapy: independent effects of epilepsy and medications. Am J Med Genet 68(1):18–24, 1997b 8986270

Nulman I, Rovet J, Stewart DE, et al: Child development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: a prospective, controlled study. Am J Psychiatry 159(11):1889–1895, 2002 12411224

Nulman I, Koren G, Rovet J, et al: Neurodevelopment of children following prenatal exposure to venlafaxine, selective serotonin reuptake inhibitors, or untreated maternal depression. Am J Psychiatry 169(11): 1165–1174, 2012 23128923

Nurnberg HG, Prudic J: Guidelines for treatment of psychosis during pregnancy. Hosp Community Psychiatry 35(1):67–71, 1984 6141137

Oberlander TF, Misri S, Fitzgerald CE, et al: Pharmacologic factors associated with transient neonatal symptoms following prenatal psychotropic medication exposure. J Clin Psychiatry 65(2):230–237, 2004 15003078

Oberlander TF, Warburton W, Misri S, et al: Neonatal outcomes after prenatal exposure to selective serotonin reuptake inhibitor antidepressants and maternal depression using population-based linked health data. Arch Gen Psychiatry 63(8): 898–906, 2006 16894066

Oberlander TF, Reebye P, Misri S, et al: Externalizing and attentional behaviors in children of depressed mothers treated with a selective serotonin reuptake inhibitor antidepressant during pregnancy. Arch Pediatr Adolesc Med 161(1):22–29, 2007 17199063

O’Connor M, Johnson GH, James DI: Intrauterine effect of phenothiazines. Med J Aust 1(8):416–417, 1981 7254089

O’Connor TG, Heron J, Golding J, et al; ALSPAC Study Team: Maternal antenatal anxiety and behavioural/emotional problems in children: a test of a programming hypothesis. J Child Psychol Psychiatry 44(7):1025–1036, 2003 14531585

Odsbu I, Skurtveit S, Selmer R, et al: Prenatal exposure to anxiolytics and hypnotics and language competence at 3 years of age. Eur J Clin Pharmacol 71(3):283–291, 2015 25547568

Oesterheld JR: A review of developmental aspects of cytochrome P450. J Child Adolesc Psychopharmacol 8(3):161–174, 1998 9853690

O’Hara MW, Rehm LP, Campbell SB: Predicting depressive symptomatology: cognitive-behavioral models and postpartum depression. J Abnorm Psychol 91(6):457–461, 1982 7153419

Ohman I, Vitols S, Tomson T: Lamotrigine in pregnancy: pharmacokinetics during delivery, in the neonate, and during lactation. Epilepsia 41(6):709–713, 2000 10840403

Ohman R, Hägg S, Carleborg L, et al: Excretion of paroxetine into breast milk. J Clin Psychiatry 60(8):519–523, 1999 10485633

Omtzigt JG, Nau H, Los FJ, et al: The disposition of valproate and its metabolites in the late first trimester and early second trimester of pregnancy in maternal serum, urine, and amniotic fluid: effect of dose, co-medication, and the presence of spina bifida. Eur J Clin Pharmacol 43(4):381–388, 1992 1451717

Omtzigt JG, Los FJ, Meijer JW, et al: The 10,11-epoxide-10,11-diol pathway of carbamazepine in early pregnancy in maternal serum, urine, and amniotic fluid: effect of dose, comedication, and relation to outcome of pregnancy. Ther Drug Monit 15(1):1–10, 1993 8451773

Ordy JM, Samorajski T, Collins RL, et al: Prenatal chlorpromazine effects on liver, survival and behavior of mice offspring. J Pharmacol Exp Ther 151(1):110–125, 1966 5902168

Ornoy A, Cohen E: Outcome of children born to epileptic mothers treated with carbamazepine during pregnancy. Arch Dis Child 75(6):517–520, 1996 9014606

Orr ST, Miller CA: Maternal depressive symptoms and the risk of poor pregnancy outcome: review of the literature and preliminary findings. Epidemiol Rev 17(1):165–171, 1995 8521934

Orr ST, James SA, Blackmore Prince C: Maternal prenatal depressive symptoms and spontaneous preterm births among African-American women in Baltimore, Maryland. Am J Epidemiol 156(9):797–802, 2002 12396996

Otani K: Risk factors for the increased seizure frequency during pregnancy and puerperium. Folia Psychiatr Neurol Jpn 39(1):33–41, 1985 4054760

Özdemir AK, Pak SC, Canan F, et al: Paliperidone palmitate use in pregnancy in a woman with schizophrenia. Arch Women Ment Health 18(5):739–740, 2015 25599999

Pacifici GM, Nottoli R: Placental transfer of drugs administered to the mother. Clin Pharmacokinet 28(3):235–269, 1995 7758253

Page-Sharp M, Kristensen JH, Hackett LP, et al: Transfer of lamotrigine into breast milk. Ann Pharmacother 40(7–8):1470–1471, 2006 16868219

Parkin DE: Probable Benadryl withdrawal manifestations in a newborn infant. J Pediatr 85(4):580, 1974 4443870

Pastuszak A, Schick-Boschetto B, Zuber C, et al: Pregnancy outcome following first-trimester exposure to fluoxetine (Prozac). JAMA 269(17):2246–2248, 1993 8474204

Paulson GW, Paulson RB: Teratogenic effects of anticonvulsants. Arch Neurol 38(3): 140–143, 1981 6781455

Paulzen M, Lammertz SE, Veselinovic T, et al: Lamotrigine in pregnancy—therapeutic drug monitoring in maternal blood, amniotic fluid, and cord blood. Int Clin Psychopharmacol 30(5):249–254, 2015 26086711

Peng M, Gao K, Ding Y, et al: Effects of prenatal exposure to atypical antipsychotics on postnatal development and growth of infants: a case-controlled, prospective study. Psychopharmacology (Berl) 228(4):577–584, 2013 23559219

Pennell PB, Newport DJ, Stowe ZN, et al: The impact of pregnancy and childbirth on the metabolism of lamotrigine. Neurology 62(2):292–295, 2004 14745072

Pennell PB, Peng L, Newport DJ, et al: Lamotrigine in pregnancy: clearance, therapeutic drug monitoring, and seizure frequency. Neurology 70(22 pt 2):2130–2136, 2008 18046009

Perkin MR, Bland JM, Peacock JL, et al: The effect of anxiety and depression during pregnancy on obstetric complications. Br J Obstet Gynaecol 100(7):629–634, 1993 8369244

Petersen I, Gilbert RE, Evans SJ, et al: Pregnancy as a major determinant for discontinuation of antidepressants: an analysis of data from The Health Improvement Network. J Clin Psychiatry 72(7):979–985, 2011 21457681

Petrenaite V, Sabers A, Hansen-Schwartz J: Individual changes in lamotrigine plasma concentrations during pregnancy. Epilepsy Res 65(3):185–188, 2005 16084694

Philbert A, Pedersen B, Dam M: Concentration of valproate during pregnancy, in the newborn and in breast milk. Acta Neurol Scand 72(5):460–463, 1985 3936331

Physicians’ Desk Reference (PDR), 61st Edition. Montvale, NJ, Thomson Healthcare, 2007

Piontek CM, Baab S, Peindl KS, et al: Serum valproate levels in 6 breastfeeding mother-infant pairs. J Clin Psychiatry 61(3):170–172, 2000 10817100

Piontek CM, Wisner KL, Perel JM, et al: Serum fluvoxamine levels in breastfed infants. J Clin Psychiatry 62(2):111–113, 2001 11247095

Polen KND, Rasmussen SA, Riehle-Colarusso T, et al; National Birth Defects Prevention Study: Association between reported venlafaxine use in early pregnancy and birth defects, national birth defects prevention study, 1997–2007 (Part A). Birth Defects Res A Clin Mol Teratol 97(1):28–35, 2013 23281074

Polepally AR, Pennell PB, Brundage RC, et al: Model-based lamotrigine clearance changes during pregnancy: clinical implication. Ann Clin Transl Neurol 1(2):99–106, 2014 24883336

Pynnönen S, Sillanpää M: Letter: Carbamazepine and mother’s milk. Lancet 2(7934): 563, 1975 51396

Pynnönen S, Kanto J, Sillanpää M, et al: Carbamazepine: placental transport, tissue concentrations in foetus and newborn, and level in milk. Acta Pharmacol Toxicol (Copenh) 41(3):244–253, 1977 578653

Rai D, Lee BK, Dalman C, et al: Parental depression, maternal antidepressant use during pregnancy, and risk of autism spectrum disorders: population based case-control study. BMJ 346:f2059, 2013 23604083

Rambeck B, Kurlemann G, Stodieck SR, et al: Concentrations of lamotrigine in a mother on lamotrigine treatment and her newborn child. Eur J Clin Pharmacol 51(6):481–484, 1997 9112063

Rampono J, Hackett LP, Kristensen JH, et al: Transfer of escitalopram and its metabolite demethylescitalopram into breastmilk. Br J Clin Pharmacol 62(3):316–322, 2006 16934048

Rampono J, Simmer K, Ilett KF, et al: Placental transfer of SSRI and SNRI antidepressants and effects on the neonate. Pharmacopsychiatry 42(3):95–100, 2009 19452377

Rampono J, Teoh S, Hackett LP, et al: Estimation of desvenlafaxine transfer into milk and infant exposure during its use in lactating women with postnatal depression. Arch Women Ment Health 14(1):49–53, 2011 20960017

Ranganathan P, Pramesh CS, Buyse M: Common pitfalls in statistical analysis: clinical versus statistical significance. Perspect Clin Res 6(3):169–170, 2015 26229754

Rawlings WJ, Ferguson R, Maddison TG: Phenmetrazine and trifluoperazine. Med J Aust 1:370, 1963

Reefhuis J, Devine O, Friedman JM, et al; National Birth Defects Prevention Study: Specific SSRIs and birth defects: Bayesian analysis to interpret new data in the context of previous reports. BMJ 351:h3190, 2015 26156519

Reisinger TL, Newman M, Loring DW, et al: Antiepileptic drug clearance and seizure frequency during pregnancy in women with epilepsy. Epilepsy Behav 29(1):13–18, 2013 23911354

Rieder RO, Rosenthal D, Wender P, et al: The offspring of schizophrenics: fetal and neonatal deaths. Arch Gen Psychiatry 32(2):200–211, 1975 234727

Riordan D, Appleby L, Faragher B: Mother-infant interaction in post-partum women with schizophrenia and affective disorders. Psychol Med 29(4):991–995, 1999 10473327

Robertson RT, Majka JA, Peter CP, et al: Effects of prenatal exposure to chlorpromazine on postnatal development and behavior of rats. Toxicol Appl Pharmacol 53(3):541–549, 1980 7385249

Rodríguez-Pinilla E, Arroyo I, Fondevilla J, et al: Prenatal exposure to valproic acid during pregnancy and limb deficiencies: a case-control study. Am J Med Genet 90(5):376–381, 2000 10706358

Rosa FW: Spina bifida in infants of women treated with carbamazepine during pregnancy. N Engl J Med 324(10):674–677, 1991 1994251

Rosenberg L, Mitchell AA, Parsells JL, et al: Lack of relation of oral clefts to diazepam use during pregnancy. N Engl J Med 309(21):1282–1285, 1983 6633586

Ross LE, Grigoriadis S, Mamisashvili L, et al: Selected pregnancy and delivery outcomes after exposure to antidepressant medication: a systematic review and meta-analysis. JAMA Psychiatry 70(4): 436–443, 2013 23446732

Rumeau-Rouquette C, Goujard J, Huel G: Possible teratogenic effect of phenothiazines in human beings. Teratology 15(1):57–64, 1977 841482

Sabers A, Dam M, A-Rogvi-Hansen B, et al: Epilepsy and pregnancy: lamotrigine as main drug used. Acta Neurol Scand 109(1):9–13, 2004 14653845

Samrén EB, van Duijn CM, Koch S, et al: Maternal use of antiepileptic drugs and the risk of major congenital malformations: a joint European prospective study of human teratogenesis associated with maternal epilepsy. Epilepsia 38(9):981–990, 1997 9579936

Samrén EB, van Duijn CM, Christiaens GC, et al: Antiepileptic drug regimens and major congenital abnormalities in the offspring. Ann Neurol 46(5):739–746, 1999 10553991

Sanchis A, Rosique D, Catala J: Adverse effects of maternal lorazepam on neonates. DICP 25(10):1137–1138, 1991 1803810

Sathanandar S, Blesi K, Tran T, et al: Lamotrigine clearance increases markedly during pregnancy. Epilepsia 41:246, 2000

Saxén I: Letter: Cleft palate and maternal diphenhydramine intake. Lancet 1(7854): 407–408, 1974 4131054

Saxén I: Associations between oral clefts and drugs taken during pregnancy. Int J Epidemiol 4(1):37–44, 1975 1116890

Saxén I, Saxén L: Letter: Association between maternal intake of diazepam and oral clefts. Lancet 2(7933):498, 1975 51304

Schell LM: Environmental noise and human prenatal growth. Am J Phys Anthropol 56(1):63–70, 1981 7337145

Schick-Boschetto B, Zuber C: Alprazolam exposure during early human pregnancy. Teratology 45:460, 1992

Schlotterbeck P, Leube D, Kircher T, et al: Aripiprazole in human milk. Int J Neuropsychopharmacol 10(3):433, 2007 17291382

Schlotterbeck P, Saur R, Hiemke C, et al: Low concentration of ziprasidone in human milk: a case report. Int J Neuropsychopharmacol 12(3):437–438, 2009 19203410

Schmidt K, Olesen OV, Jensen PN: Citalopram and breast-feeding: serum concentration and side effects in the infant. Biol Psychiatry 47(2):164–165, 2000 10664835

Schou M: What happened later to the lithium babies? A follow-up study of children born without malformations. Acta Psychiatr Scand 54(3):193–197, 1976 970196

Schou M, Amdisen A: Lithium and pregnancy. 3. Lithium ingestion by children breast-fed by women on lithium treatment. BMJ 2(5859):138, 1973 4699592

Scokel PW3rd, Jones WN: Infant jaundice after phenothiazine drugs for labor: an enigma. Obstet Gynecol 20:124–127, 1962 13909880

Scolnik D, Nulman I, Rovet J, et al: Neurodevelopment of children exposed in utero to phenytoin and carbamazepine monotherapy. JAMA 271(10):767–770, 1994 7509419

Shallcross R, Bromley RL, Irwin B, et al; Liverpool Manchester Neurodevelopment Group; UK Epilepsy and Pregnancy Register: Child development following in utero exposure: levetiracetam vs sodium valproate. Neurology 76(4):383–389, 2011 21263139

Shannon RW, Fraser GP, Aitken RG, et al: Diazepam in preeclamptic toxaemia with special reference to its effect on the newborn infant. Br J Clin Pract 26(6):271–275, 1972 5044095

Shao P, Ou J, Peng M, et al: Effects of clozapine and other atypical antipsychotics on infants development who were exposed to as fetus: a post hoc analysis. PLoS One 10(4):e0123373, 2015 25909513

Shiono PH, Mills JL: Oral clefts and diazepam use during pregnancy. N Engl J Med 311(14):919–920, 1984 6472406

Simon GE, Cunningham ML, Davis RL: Outcomes of prenatal antidepressant exposure. Am J Psychiatry 159(12):2055–2061, 2002 12450956

Sit DK, Perel JM, Helsel JC, Wisner KL: Changes in antidepressant metabolism and dosing across pregnancy and early postpartum. J Clin Psychiatry 69(4):652–658, 2008 18426260

Sivojelezova A, Shuhaiber S, Sarkissian L, et al: Citalopram use in pregnancy: prospective comparative evaluation of pregnancy and fetal outcome. Am J Obstet Gynecol 193(6):2004–2009, 2005 16325604

Skausig OB, Schou M: [Breast feeding during lithium therapy] (Danish). Ugeskr Laeger 139(7):400–401, 1977 841726

Slone D, Siskind V, Heinonen OP, et al: Antenatal exposure to the phenothiazines in relation to congenital malformations, perinatal mortality rate, birth weight, and intelligence quotient score. Am J Obstet Gynecol 128(5):486–488, 1977 879206

Smit M, Wennink H, Heres M, et al: Mirtazapine in pregnancy and lactation: data from a case series. J Clin Psychopharmacol 35(2):163–167, 2015 25689290

Smith MV, Poschman K, Cavaleri MA, et al: Symptoms of posttraumatic stress disorder in a community sample of low-income pregnant women. Am J Psychiatry 163(5):881–884, 2006 16648330

Sodhi P, Poddar B, Parmar V: Fatal cardiac malformation in fetal valproate syndrome. Indian J Pediatr 68(10):989–990, 2001 11758141

Søndergaard C, Olsen J, Friis-Haschè E, et al: Psychosocial distress during pregnancy and the risk of infantile colic: a follow-up study. Acta Paediatr 92(7):811–816, 2003 12892160

Sørensen MJ, Grønborg TK, Christensen J, et al: Antidepressant exposure in pregnancy and risk of autism spectrum disorders. Clin Epidemiol 5:449–459, 2013 24255601

Speight AN: Floppy-infant syndrome and maternal diazepam and/or nitrazepam. Lancet 2(8043):878, 1977 72227

Spigset O, Carleborg L, Norström A, et al: Paroxetine level in breast milk. J Clin Psychiatry 57(1):39, 1996 8543546

Spigset O, Carieborg L, Ohman R, et al: Excretion of citalopram in breast milk. Br J Clin Pharmacol 44(3):295–298, 1997 9296327

St Clair SM, Schirmer RG: First-trimester exposure to alprazolam. Obstet Gynecol 80(5):843–846, 1992 1407925

Stahl MM, Neiderud J, Vinge E: Thrombocytopenic purpura and anemia in a breast-fed infant whose mother was treated with valproic acid. J Pediatr 130(6):1001–1003, 1997 9202628

Steer RA, Scholl TO, Hediger ML, et al: Self-reported depression and negative pregnancy outcomes. J Clin Epidemiol 45(10): 1093–1099, 1992 1474405

Stewart RB, Karas B, Springer PK: Haloperidol excretion in human milk. Am J Psychiatry 137(7):849–850, 1980 7386670

Stiegler A, Schaletzky R, Walter G, et al: Olanzapine treatment during pregnancy and breastfeeding: a chance for women with psychotic illness? Psychopharmacology (Berl) 231(15):3067–3069, 2014 24938920

Stirrat GM, Edington PT, Berry DJ: Letter: Transplacental passage of chlordiazepoxide. BMJ 2(5921):729, 1974 4859397

Stoner SC, Sommi RW Jr, Marken PA, et al: Clozapine use in two full-term pregnancies. J Clin Psychiatry 58(8):364–365, 1997 9515978

Stott DH: Follow-up study from birth of the effects of prenatal stresses. Dev Med Child Neurol 15(6):770–787, 1973 4129091

Stowe ZN, Nemeroff CB: Women at risk for postpartum-onset major depression. Am J Obstet Gynecol 173(2):639–645, 1995 7645646

Stowe ZN, Owens MJ, Landry JC, et al: Sertraline and desmethylsertraline in human breast milk and nursing infants. Am J Psychiatry 154(9):1255–1260, 1997 9286185

Stowe ZN, Cohen LS, Hostetter A, et al: Paroxetine in human breast milk and nursing infants. Am J Psychiatry 157(2):185–189, 2000 10671385

Stowe ZN, Hostetter AL, Owens MJ, et al: The pharmacokinetics of sertraline excretion into human breast milk: determinants of infant serum concentrations. J Clin Psychiatry 64(1):73–80, 2003 12590627

Stowe ZN, Hostetter AL, Newport DJ: The onset of postpartum depression: implications for clinical screening in obstetrical and primary care. Am J Obstet Gynecol 192(2):522–526, 2005 15695997

Summerfield RJ, Nielsen MS: Excretion of lorazepam into breast milk. Br J Anaesth 57(10):1042–1043, 1985 4041315

Suri R, Stowe ZN, Hendrick V, et al: Estimates of nursing infant daily dose of fluoxetine through breast milk. Biol Psychiatry 52(5):446–451, 2002 12242061

Sykes PA, Quarrie J, Alexander FW: Lithium carbonate and breast-feeding. BMJ 2(6047):1299, 1976 1000200

Taddio A, Ito S, Koren G: Excretion of fluoxetine and its metabolite, norfluoxetine, in human breast milk. J Clin Pharmacol 36(1):42–47, 1996 8932542

Targum SD, Davenport YB, Webster MJ: Postpartum mania in bipolar manic-depressive patients withdrawn from lithium carbonate. J Nerv Ment Dis 167(9):572–574, 1979 479871

Taylor CL, Stewart R, Ogden J, et al: The characteristics and health needs of pregnant women with schizophrenia compared with bipolar disorder and affective psychoses. BMC Psychiatry 15:88, 2015 25886140

Tenyi T, Tixler M: Clozapine in the treatment of pregnant schizophrenic women. Psychiatria Danubina 10:15–18, 1998

ter Horst PGJ, van der Linde S, Smit JP, et al: Clomipramine concentration and withdrawal symptoms in 10 neonates. Br J Clin Pharmacol 73(2):295–302, 2012 21801198

Thisted E, Ebbesen F: Malformations, withdrawal manifestations, and hypoglycaemia after exposure to valproate in utero. Arch Dis Child 69(3 Spec No):288–291, 1993 8215567

Tomson T, Lindbom U, Ekqvist B, et al: Disposition of carbamazepine and phenytoin in pregnancy. Epilepsia 35(1):131–135, 1994 8112235

Tomson T, Ohman I, Vitols S: Lamotrigine in pregnancy and lactation: a case report. Epilepsia 38(9):1039–1041, 1997 9579945

Tomson T, Battino D, Bonizzoni E, et al; EURAP study group: Dose-dependent risk of malformations with antiepileptic drugs: an analysis of data from the EURAP epilepsy and pregnancy registry. Lancet Neurol 10(7):609–617, 2011 21652013

Tomson T, Battino D, Bonizzoni E, et al; EURAP Study Group: Dose-dependent teratogenicity of valproate in mono- and polytherapy: an observational study. Neurology 85(10):866–872, 2015 26085607

Tonn P, Reuter SC, Hiemke C, et al: High mirtazapine plasma levels in infant after breast feeding: case report and review of the literature. J Clin Psychopharmacol 29(2):191–192, 2009 19512989

Tran TA, Leppik IE, Blesi K, et al: Lamotrigine clearance during pregnancy. Neurology 59(2):251–255, 2002 12136066

Troutman BR, Cutrona CE: Nonpsychotic postpartum depression among adolescent mothers. J Abnorm Psychol 99(1):69–78, 1990 2307769

Tsuru N, Maeda T, Tsuruoka M: Three cases of delivery under sodium valproate—placental transfer, milk transfer and probable teratogenicity of sodium valproate. Jpn J Psychiatry Neurol 42(1):89–96, 1988 3135429

Tunnessen WWJr, Hertz CG: Toxic effects of lithium in newborn infants: a commentary. J Pediatr 81(4):804–807, 1972 5074360

Turton P, Hughes P, Evans CD, et al: Incidence, correlates and predictors of post-traumatic stress disorder in the pregnancy after stillbirth. Br J Psychiatry 178: 556–560, 2001 11388974

Uguz F, Sahingoz M, Sonmez EO, et al: The effects of maternal major depression, generalized anxiety disorder, and panic disorder on birth weight and gestational age: a comparative study. J Psychosom Res 75(1):87–89, 2013 23751245

Vajda FJ, O’Brien TJ, Hitchcock A, et al: The Australian registry of anti-epileptic drugs in pregnancy: experience after 30 months. J Clin Neurosci 10(5):543–549, 2003 12948456

Vajda FJ, O’Brien TJ, Graham J, et al: Associations between particular types of fetal malformation and antiepileptic drug exposure in utero. Acta Neurol Scand 128(4):228–234, 2013 23461556

Van Boekholt AA, Hartong EGTM, Huntjens-Fleuren H, et al: Quetiapine concentrations during exclusive breastfeeding and maternal quetiapine use. Ann Pharmacother 49(6):743–744, 2015 25975996

Van den Bergh BR, Marcoen A: High antenatal maternal anxiety is related to ADHD symptoms, externalizing problems, and anxiety in 8- and 9-year-olds. Child Dev 75(4):1085–1097, 2004 15260866

van der Lugt NM, van de Maat JS, van Kamp IL, et al: Fetal, neonatal and developmental outcomes of lithium-exposed pregnancies. Early Hum Dev 88(6):375–378, 2012 22000820

van der Pol MC, Hadders-Algra M, Huisjes HJ, et al: Antiepileptic medication in pregnancy: late effects on the children’s central nervous system development. Am J Obstet Gynecol 164(1 pt 1):121–128, 1991 1986598

Van Waes A, Van de Velde EJ: Safety evaluation of haloperidol in the treatment of hyperemesis gravidarum. J Clin Pharmacol 9(4):224–227, 1969

Vasilakis-Scaramozza C, Aschengrau A, Cabral H, et al: Antidepressant use during early pregnancy and the risk of congenital anomalies. Pharmacotherapy 33(7):693–700, 2013 23744675

Veiby G, Daltveit AK, Schjølberg S, et al: Exposure to antiepileptic drugs in utero and child development: a prospective population-based study. Epilepsia 54(8):1462–1472, 2013 23865818

Veiby G, Daltveit AK, Engelsen BA, et al: Fetal growth restriction and birth defects with newer and older antiepileptic drugs during pregnancy. J Neurol 261(3):579–588, 2014 24449062

Venkatesh KK, Riley L, Castro VM, et al: Association of antenatal depression symptoms and antidepressant treatment with preterm birth. Obstet Gynecol 127(5):926–933, 2016 27054941

Viguera AC, Newport DJ, Ritchie J, et al: Lithium in breast milk and nursing infants: clinical implications. Am J Psychiatry 164(2):342–345, 2007a 17267800

Viguera AC, Whitfield T, Baldessarini RJ, et al: Risk of recurrence in women with bipolar disorder during pregnancy: prospective study of mood stabilizer discontinuation. Am J Psychiatry 164(12):1817–1824, quiz 1923, 2007b 18056236

von Unruh GE, Froescher W, Hoffmann F, et al: Valproic acid in breast milk: how much is really there? Ther Drug Monit 6(3):272–276, 1984 6438834

Waldman MD, Safferman AZ: Pregnancy and clozapine. Am J Psychiatry 150(1):168–169, 1993 8018113

Wang JS, Newport DJ, Stowe ZN, et al: The emerging importance of transporter proteins in the psychopharmacological treatment of the pregnant patient. Drug Metab Rev 39(4):723–746, 2007 18058331

Warner A: Drug use in the neonate: interrelationships of pharmacokinetics, toxicity, and biochemical maturity. Clin Chem 32(5):721–727, 1986 2421942

Watanabe N, Kasahara M, Sugibayashi R, et al: Perinatal use of aripiprazole: a case report. J Clin Psychopharmacol 31(3):377–379, 2011 21532364

Watson JP, Elliott SA, Rugg AJ, et al: Psychiatric disorder in pregnancy and the first postnatal year. Br J Psychiatry 144:453–462, 1984 6733369

Weggelaar NM, Keijer WJ, Janssen PKC: A case report of risperidone distribution and excretion into human milk: how to give good advice if you have not enough data available. J Clin Psychopharmacol 31(1):129–131, 2011 21192160

Weinstein MR, Goldfield M: Lithium carbonate treatment during pregnancy; report of a case. Dis Nerv Syst 30(12):828–832, 1969 5369134

Weinstein MR, Goldfield M: Cardiovascular malformations with lithium use during pregnancy. Am J Psychiatry 132(5):529–531, 1975 1119612

Weissman MM, Prusoff BA, Gammon GD, et al: Psychopathology in the children (ages 6–18) of depressed and normal parents. J Am Acad Child Psychiatry 23(1):78–84, 1984 6693680

Welch RM, Findlay JW: Excretion of drugs in human breast milk. Drug Metab Rev 12(2):261–277, 1981 7040016

Wesson DR, Camber S, Harkey M, et al: Diazepam and desmethyldiazepam in breast milk. J Psychoactive Drugs 17(1):55–56, 1985 3920372

Whalley LJ, Blain PG, Prime JK: Haloperidol secreted in breast milk. Br Med J (Clin Res Ed) 282(6278):1746–1747, 1981 6786603

Whitelaw AG, Cummings AJ, McFadyen IR: Effect of maternal lorazepam on the neonate. Br Med J (Clin Res Ed) 282(6270): 1106–1108, 1981 6113019

Whitworth A, Stuppaeck C, Yazdi K, et al: Olanzapine and breast-feeding: changes of plasma concentrations of olanzapine in a breast-fed infant over a period of 5 months. J Psychopharmacol 24(1):121–123, 2010 18801835

Wide K, Winbladh B, Tomson T, et al: Psychomotor development and minor anomalies in children exposed to antiepileptic drugs in utero: a prospective population-based study. Dev Med Child Neurol 42(2):87–92, 2000 10698324

Williams G, King J, Cunningham M, et al: Fetal valproate syndrome and autism: additional evidence of an association. Dev Med Child Neurol 43(3):202–206, 2001 11263692

Williams KE, Koran LM: Obsessive-compulsive disorder in pregnancy, the puerperium, and the premenstruum. J Clin Psychiatry 58(7):330–334, quiz 335–336, 1997 9269260

Williams PG, Hersh JH: A male with fetal valproate syndrome and autism. Dev Med Child Neurol 39(9):632–634, 1997 9344057

Wilson KL, Zelig CM, Harvey JP, et al: Persistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitors. Am J Perinatol 28(1):19–24, 2011 20607643

Wilson JT, Brown RD, Cherek DR, et al: Drug excretion in human breast milk: principles, pharmacokinetics and projected consequences. Clin Pharmacokinet 5(1): 1–66, 1980 6988135

Wilson N, Forfar JD, Godman MJ: Atrial flutter in the newborn resulting from maternal lithium ingestion. Arch Dis Child 58(7):538–539, 1983 6870336

Wilton LV, Pearce GL, Martin RM, et al: The outcomes of pregnancy in women exposed to newly marketed drugs in general practice in England. Br J Obstet Gynaecol 105(8):882–889, 1998 9746382

Windhager E, Kim SW, Saria A, et al: Perinatal use of aripiprazole: plasma levels, placental transfer, and child outcome in 3 new cases. J Clin Psychopharmacol 34(5):637–641, 2014 24949701

Winter RM: In-utero exposure to benzodiazepines. Lancet 1(8533):627–628, 1987 2881161

Winter RM, Donnai D, Burn J, et al: Fetal valproate syndrome: is there a recognisable phenotype? J Med Genet 24(11):692–695, 1987 3123693

Winterfeld U, Klinger G, Panchaud A, et al: Pregnancy outcomes following maternal exposure to mirtazapine: a multicenter, prospective study. J Clin Psychopharmacol 35(3):250–259, 2015 25830592

Wisner KL, Perel JM: Psychopharmacologic agents and electroconvulsive therapy during pregnancy and the puerperium, in Psychiatric Consultation in Childbirth Settings: Parent- and Child-Oriented Approaches. Edited by Cohen RL. New York, Plenum, 1988, pp 165–206

Wisner KL, Perel JM: Serum levels of valproate and carbamazepine in breastfeeding mother-infant pairs. J Clin Psychopharmacol 18(2):167–169, 1998 9555601

Wisner KL, Perel JM, Wheeler SB: Tricyclic dose requirements across pregnancy. Am J Psychiatry 150(10):1541–1542, 1993 8379562

Wisner KL, Peindl KS, Hanusa BH: Effects of childbearing on the natural history of panic disorder with comorbid mood disorder. J Affect Disord 41(3):173–180, 1996a 8988449

Wisner KL, Perel JM, Findling RL: Antidepressant treatment during breast-feeding. Am J Psychiatry 153(9):1132–1137, 1996b 8780414

Wisner KL, Perel JM, Blumer J: Serum sertraline and N-desmethylsertraline levels in breast-feeding mother-infant pairs. Am J Psychiatry 155(5):690–692, 1998 9585724

Wood AG, Nadebaum C, Anderson V, et al: Prospective assessment of autism traits in children exposed to antiepileptic drugs during pregnancy. Epilepsia 56(7):1047–1055, 2015 25963613

Woods DL, Malan AF: Side-effects of maternal diazepam on the newborn infant. S Afr Med J 54(16):636, 1978 741269

Woody JN, London WL, Wilbanks GD Jr: Lithium toxicity in a newborn. Pediatrics 47(1):94–96, 1971 5545409

Wretlind M: Excretion of oxazepam in breast milk. Eur J Clin Pharmacol 33(2):209–210, 1987 3691611

Wright S, Dawling S, Ashford JJ: Excretion of fluvoxamine in breast milk. Br J Clin Pharmacol 31(2):209, 1991 1904751

Wyska E, Jusko WJ: Approaches to pharmacokinetic/pharmacodynamic modeling during pregnancy. Semin Perinatol 25(3): 124–132, 2001 11453607

Yerby MS, Friel PN, Miller DQ: Carbamazepine protein binding and disposition in pregnancy. Ther Drug Monit 7(3):269–273, 1985 4049462

Yerby MS, Friel PN, McCormick K, et al: Pharmacokinetics of anticonvulsants in pregnancy: alterations in plasma protein binding. Epilepsy Res 5(3):223–228, 1990 2384078

Yerby MS, Friel PN, McCormick K: Antiepileptic drug disposition during pregnancy. Neurology 42 (4 suppl 5):12–16, 1992 1574166

Yonkers KA, Gotman N, Smith MV, et al: Does antidepressant use attenuate the risk of a major depressive episode in pregnancy? Epidemiology 22(6):848–854, 2011 21900825

Yonkers KA, Blackwell KA, Glover J, et al: Antidepressant use in pregnant and postpartum women. Annu Rev Clin Psychol 10:369–392, 2014 24313569

Yoshida K, Smith B, Craggs M, et al: Fluoxetine in breast-milk and developmental outcome of breast-fed infants. Br J Psychiatry 172:175–178, 1998a 9519072

Yoshida K, Smith B, Craggs M, et al: Neuroleptic drugs in breast-milk: a study of pharmacokinetics and of possible adverse effects in breast-fed infants. Psychol Med 28(1):81–91, 1998b 9483685

Zambaldi CF, Cantilino A, Montenegro AC, et al: Postpartum obsessive-compulsive disorder: prevalence and clinical characteristics. Compr Psychiatry 50(6):503–509, 2009 19840587

Zeskind PS, Stephens LE: Maternal selective serotonin reuptake inhibitor use during pregnancy and newborn neurobehavior. Pediatrics 113(2):368–375, 2004 14754951

Zuckerman B, Amaro H, Bauchner H, et al: Depressive symptoms during pregnancy: relationship to poor health behaviors. Am J Obstet Gynecol 160(5 pt 1):1107–1111, 1989 2729387