95
Views
15
CrossRef citations to date
0
Altmetric
Review

Treatment of nonpsychotic major depression during pregnancy: patient safety and challenges

, &
Pages 109-129 | Published online: 18 Sep 2014

Abstract

In pregnant women with major depression, the overarching goal of treatment is to achieve or maintain maternal euthymia, thus limiting both maternal and fetal exposure to the harmful effects of untreated or incompletely treated depression. However, the absence of uniformly effective therapies with guaranteed obstetric and fetal safety makes the treatment of major depression during pregnancy among the most formidable of clinical challenges. Clinicians and patients are still faced with conflicting data and expert opinion regarding the reproductive safety of antidepressants in pregnancy, as well as large gaps in our understanding of the effectiveness of most antidepressants and nonpharmacological alternatives for treating antenatal depression. In this paper, we provide a clinically focused review of the available information on potential maternal and fetal risks of untreated maternal depression during pregnancy, the effectiveness of interventions for maternal depression during pregnancy, and potential obstetric, fetal, and neonatal risks associated with antenatal antidepressant use.

Introduction

Major depression and other unipolar depressive syndromes are highly prevalent and disproportionately affect women.Citation1,Citation2 The peak incidence of major depression in women is during the reproductive years,Citation3 raising the possibility of depressive episode onset (or relapse in women already diagnosed with major depression) during pregnancy. Indeed, the prevalence of any depressive illness during pregnancy is estimated at 18.4% (7.3% for major depressive disorder during pregnancy).Citation4 Rates of mood-disorder onset in women are roughly equivalent between pregnant and nonpregnant samples,Citation5,Citation6 and the frequency of major depression during the second and third trimesters may even exceed that of the general population.Citation7

Although major depression and other depressive illnesses cannot be cured at present, their symptoms can be controlled in most patients with focused psychotherapy, appropriate pharmacotherapy, or the combination of the two.Citation8 In pregnant women with major depression, the overarching goal of treatment is to achieve or maintain maternal euthymia, thus limiting both maternal and fetal exposure to the harmful effects of untreated or incompletely treated depression. Ideally, this would be achieved using treatment modalities that have no possibility of harming the pregnancy or developing fetus. Effective nonpharmacological modalities may achieve these aims for many, but not all, women with depression. Indeed, a considerable number of women benefit from antidepressant treatments for achieving or maintaining euthymia during pregnancy. On the other hand, the use of antidepressants for treating maternal depression and other disorders during pregnancy has increased steadily in the last two decades,Citation9Citation13 which has raised concerns about the risks versus benefits of this practice.

The absence of uniformly effective therapeutics with guaranteed obstetric and fetal safety makes the treatment of major depression during pregnancy among the most formidable of clinical challenges.Citation14 Clinical practice guidelines can provide direction, but to follow these guidelines clinicians must translate estimates of treatment effectiveness and risk from rapidly evolving population-level data to individual patients, taking into account each patient’s tolerance of risk related to both the underlying illness and available interventions.Citation15,Citation16 Clinicians and patients are still faced with conflicting data and expert opinion regarding the reproductive safety of antidepressants in pregnancy, as well as gaps in our understanding of the effectiveness of most antidepressants and nonpharmacological alternatives for treating antenatal depression. This paper provides a clinically focused review of the available information on risks of untreated maternal depression during pregnancy, effectiveness of interventions for maternal depression during pregnancy, and potential harms of treatments for maternal depression during pregnancy, and presents recommendations for treating maternal depression during pregnancy.

Materials and methods

Relevant studies were identified via a Medline/PubMed search of the literature for reports and studies for the period beginning in 1996 and ending in 2013. Potential harms of interest included congenital malformations, adverse neonatal events, and obstetric complications. We used combinations of keywords that defined antidepressant exposures (antidepressants, selective serotonin-reuptake inhibitors [SSRIs], fluoxetine, fluvoxamine, paroxetine, sertraline, citalopram, escitalopram, venlafaxine, desvenlafaxine, duloxetine, bupropion, tricyclic antidepressants [TCAs], imipramine, desipramine, amitriptyline, nortriptyline, clomipramine, protriptyline, trimipramine, doxepin, monoamine oxidase inhibitors, phenelzine, tranylcypromine, isocarboxazid, mirtazapine, nefazodone, and vilazodone) with those that defined outcomes of interest (pregnancy outcome [congenital, fetal], birth outcome, malformations, congenital malformations, birth defects, cardiac/heart defects, persistent pulmonary hypertension of the newborn [PPHN], and neurobehavioral outcomes). Vortioxetine was not included because of its very recent regulatory approval. Milnacipran was not included because of its approval for treating fibromyalgia syndrome and limited clinical use for treating major depression. A similar approach was used to locate reports addressing benefits and harms of nonpharmacological treatments (psychotherapy, interpersonal therapy [IPT], cognitive behavioral therapy [CBT], electroconvulsive therapy [ECT], repetitive transcranial magnetic stimulation [rTMS], exercise, bright-light therapy [luminotherapy], therapeutic massage, and acupuncture). Systematic reviews, meta-analyses, randomized trials, and observational studies were reviewed. Additional papers and abstracts were retrieved from the bibliographic sections of review papers and individual reports. Throughout the paper, we generally refer to maternal depression during pregnancy as “antenatal depression” (as opposed to “antenatal major depression”), because several of the reviewed studies included persons with clinically significant depressive symptoms but unconfirmed major depressive disorder diagnoses and because of variability in diagnostic methods across studies restricted to major depression.

Results

Clinical impact of antenatal depression

Antenatal depression has been linked with worse general health status; poor maternal self-care during pregnancy; underutilization of prenatal care; increased maternal use of cigarettes, alcohol, and other abused substances; isolation or withdrawal from family or other supports; and increased risk of postpartum depression and maternal suicide,Citation17Citation23 the latter of which may account for up to 20% of postpartum deaths.Citation24 Other potential risks of untreated antenatal depression include poor nutrition intake during pregnancy, severe fatigue, and lack of motivation for self-care.Citation25 Women with depression or other psychiatric disorders for which antidepressants are often prescribed are less likely to get appropriate prenatal and pediatric care, which may increase the risk of increased postneonatal morbidity.Citation26,Citation27

Untreated or undertreated antenatal depression has also been associated with a variety of adverse obstetric and neonatal outcomes, including preeclampsia and eclampsia,Citation28,Citation29 miscarriage,Citation30 preterm birth or shorter gestational length,Citation6,Citation30Citation32 poor fetal and infant growth,Citation33Citation35 low infant birth weight/small for gestational age,Citation6,Citation30,Citation31,Citation36,Citation37 and low Apgar scores.Citation38 A recent meta-analysis of 30 heterogeneous studies showed a modest association between antenatal depression and increased risk of premature delivery (odds ratio [OR] 1.37, 95% confidence interval [CI] 1.04–1.81) and decreased likelihood of breastfeeding initiation (OR 0.68, 95% CI 0.61–0.76).Citation39 The meta-analysis did not demonstrate a statistically significant increased risk of most other perinatal outcomes in association with antenatal depression; however, antenatal depression was significantly but modestly associated with low birth weight (OR 1.39, 95% CI 1.00–1.94) and preeclampsia (OR 2.50, 95% CI 1.13–5.54) after restricting the analyses to adjusted data.

Maternal depression during pregnancy or the postpartum period has been associated with impaired mother–infant attachment and developmental difficulties in offspring.Citation40 Children exposed to untreated antenatal depression are at higher risk for developmental delay, impaired language development, and lower intelligence quotient (IQ) scores,Citation41Citation43 and poor socioemotional development, including worse emotional regulation and higher rates of depressive and anxiety symptoms.Citation44Citation46 Others have shown that approximately a third of school-age children experience depressive, anxiety, or disruptive disorders while their mother is depressed.Citation47

On the other hand, successful treatment of antenatal depression is associated with reduced psychiatric symptoms and diagnoses in their children. In a cohort of 151 mother– child pairs who participated in the STAR*D-Child (Sequenced Treatment Alternatives to Relieve Depression – Child) study, remission of maternal depression after 3 months of medication treatment occurred in 33% of subjectsCitation48 and was associated with an 11% decrease in the rates of an affective, anxiety, or disruptive behavior disorders in offspring (aged 7–17 years), as assessed by the Kiddie Schedule for Disorders and Schizophrenia. Continuing depression was associated with an 8% increase in the rates of these diagnoses. The relationship between improvement in maternal depression and lower rates of child psychopathology and functioning were still apparent after 1 year of follow-up.Citation49

Effectiveness of treatments for antenatal depression

Clinicians, patients, and their families have a wide range of treatments from which to choose. Common treatments include pharmacotherapy, psychotherapy, and other therapies.

Pharmacotherapy

The effectiveness of antidepressants for treating antenatal depression or preventing depressive relapse in women with depression during pregnancy has received limited investigation. In a nonrandomized, multicenter, prospective study of 201 antidepressant-treated patients (161 [81.1%] of whom were treated with SSRIs) recruited from specialty mental health treatment settings who were euthymic for at least 3 months prior to study entry, Cohen et al reported that discontinuation of antidepressants during pregnancy was associated with more frequent relapses (68% versus 26%) compared with women who continued their medication.Citation50 Women who stopped effective antidepressant treatment had five times the risk of relapse. Of those who stopped antidepressant treatment but restarted medication, the overall risk of relapse was also decreased, but to a smaller degree than patients who received continuous treatment throughout pregnancy. Lowering the dose of antidepressant did not reduce the risk of relapse. Relapses occurred more often in depressed patients with markers of more severe illness, including longer duration of illness, greater number of lifetime episodes, history of suicidal ideation, and family history of depressive illness.

A small prospective cohort study of 238 depressed pregnant women, 71 of whom were treated with SSRIs, reported that mean depressive symptom levels (as measured by the 29-item Structured Interview Guide for the Hamilton Depression Rating Scale with Atypical Depression Supplement and the 17-item version of the Hamilton Depression Rating Scale) in women with continuous depression during pregnancy and no SSRI treatment were significantly greater than exposure groups that consisted of depressed women who received SSRI treatment. The study authors suggest that the results were consistent with the “expected treatment effects of SSRIs”, presumably in comparison with nonpuerperal major depression.Citation51

It is currently unclear if antidepressant effectiveness is the same for treating puerperal and nonpuerperal major depression, and questions remain regarding the effectiveness of antidepressant treatment for preventing depressive relapses during pregnancy in women with established diagnoses of major depression.Citation15 In the Cohen et al study, women who chose to continue an effective antidepressant throughout pregnancy still had a 26% risk of relapse during follow-up.Citation50 Moreover, in a well-controlled study of 778 women with a history of any depressive disorder (as determined by lay interviewers using the Composite International Diagnostic Interview) who were recruited from obstetric rather than specialty mental health care settings, the risk of a major depressive episode during pregnancy did not differ statistically between women who took antidepressants and those who did not (hazard ratio 0.85, 95% CI 0.52–1.40).Citation52 The overall rate of major depressive episode onset during pregnancy was 16%, though differences by antidepressant exposure in rates of depressive episode occurrence were not reported, and the analyses were not stratified by depression severity or presence of comorbid illnesses, such as anxiety disorders.

Therefore, it seems clear that some pregnant women with antenatal depression will benefit from antidepressant treatment, but a substantial proportion of patients will not. This is broadly consistent with findings from studies of antidepressant effectiveness in the broader population of patients with nonpuerperal major depression. Comparative effectiveness studies of antidepressants for treating antenatal depression are lacking, and additional studies are needed to determine the effectiveness of antidepressants for treating antenatal depression across treatment settings. This would not only permit better estimates of antidepressant effectiveness overall and by individual agent, but would also have the potential to inform clinical practice by helping determine which patients are most likely to benefit from specific antidepressant medications during pregnancy.

Psychotherapy

There is evidence from controlled studies that systematically oriented psychotherapies, such as IPT and CBT, may be effective for treating antenatal depression or clinically significant depressive symptoms that occur during pregnancy.

Interpersonal therapy

A recent meta-analysis of 24 studies (18 of which focused on individual IPT and six of which focused on relational therapies) in pregnant or postpartum women with depression demonstrated significant positive effects on depressive symptoms.Citation53 Treatment-effect sizes were larger with individual IPT than relational therapies (such as marital and family therapies). Interventions that included an IPT component were also found to have greater effect sizes for reducing depressive symptom levels in another systematic review and meta-analysis of 27 heterogeneous studies that examined the effectiveness of pharmacologic and psychological interventions for unipolar depression during pregnancy or the postpartum.Citation54 Partner-assisted IPT, a form of IPT that was specifically developed for perinatal depression, showed promising results in a preliminary proof-of-concept trial in women with antenatal or postpartum depression.Citation55

Cognitive behavioral therapy

Although CBT is an established treatment for mild-to-moderate nonpuerperal major depression, far less is known about its effectiveness for treating antenatal depression. The same meta-analysis of 27 heterogeneous studies reviewed earlier found CBT to be beneficial for perinatal depression, albeit with small-to-medium effects.Citation54 In a randomized trial, CBT adapted for perinatal depression in outpatients was associated with greater improvement in depressive symptoms than standard care over 16 weeks of follow-up.Citation56 Most individual trials of CBT for perinatal depression have focused on the treatment of postpartum depression.Citation57Citation59 Therefore, additional investigations of the effectiveness of CBT for treating antenatal depression are needed.

Other psychotherapies

Difficulties with marital, family, and other important relationships are common complications of mood disorders in general, and are robust predictors of depression during pregnancy and the postpartum period.Citation60 It is therefore surprising that there has been so little investigation of the effectiveness of relational therapies (focused on marital and family problems) for treating antenatal depression. One meta-analysis reviewed earlier showed lower effect sizes with relational therapies than IPT in women with antenatal depression;Citation53 however, it seems reasonable that cases of maternal depression in which marital and family problems contribute heavily to depressive symptom burden may also stand to benefit most from these interventions as well as IPT. The effectiveness of psychodynamic psychotherapy and psychoanalytic approaches in this population is described only in case literature.Citation61

Other nonpharmacological interventions

Exercise

Physical exercise during pregnancy is associated with improved cardiorespiratory fitness and other health benefits, without evidence of harm to the newborn.Citation62 While not generally considered to be a stand-alone treatment for depression, exercise has been shown to help alleviate symptoms of major depression in nonpregnant samples.Citation63Citation65 One study showed that supervised aerobic exercise commenced at 16–20 weeks gestation and extended over 3 months resulted in reduced depressive symptoms in nulliparous women with no psychiatric history.Citation66 Results of two small studies suggest that significant decreases in depressive symptoms can occur in women who were previously inactive.Citation67,Citation68 Controlled studies of the benefits of exercise as either a primary or adjunctive intervention for treating antenatal depression are needed.

Luminotherapy

Luminotherapy, or bright-light therapy, is effective for seasonal and nonseasonal depression, though effect sizes for nonseasonal depression are smaller.Citation69,Citation70 In an open trial of 16 women with antenatal depression, early morning bright light (60 minutes at 10,000 lx) was associated with a 49% decrease in depressive symptoms over 3 weeks.Citation71 Further improvement was observed in seven patients who continued early morning bright-light therapy for 5 weeks. An initial randomized trial of ten patients with antenatal depression showed a higher positive response rate with luminotherapy delivered at 7,000 lx than a placebo condition consisting of subtherapeutic light exposure (60% versus 41%); however, these differences were not statistically significant.Citation72 In a larger randomized trial, significantly higher rates of positive treatment response were observed with luminotherapy (7,000 lx bright light) than placebo (70 lx dim red light) over 5 weeks (81.3% versus 45.5%).Citation73 Large, randomized trials are needed to establish the antidepressive effect of luminotherapy for treating antenatal depression.

Repetitive transcranial magnetic stimulation

One open-label study of ten women with antenatal depression who received 20 daily rTMS treatments, with or without concomitant antidepressants, showed a 70% positive treatment-response rate.Citation74 A larger study of 30 antenatally depressed patients who received rTMS over the left prefrontal cortex (6 days weekly for 3 weeks) showed significant reduction from baseline in Hamilton Depression Rating Scale scores. Rates of response and remission were 41.4% and 20.7%, respectively.Citation75 No adverse pregnancy or fetal outcomes were reported in either study. Randomized trials are needed to establish the antidepressive effect of rTMS for treating antenatal depression.

Electroconvulsive therapy

ECT is an established therapeutic modality for severe, catatonic, or psychotic depression, where rapid antidepressive effects are urgently required and lag time to therapeutic benefit associated with pharmacotherapy may be unacceptable. Extensive case-series data provide much of the rationale for the effectiveness and safety of ECT during pregnancy.Citation76,Citation77 Although ECT has been used safely during all three trimesters of pregnancy and no consistent patterns of fetal or birth complications have been reported,Citation76,Citation78 the anesthetic agents and neuromuscular blockers that are commonly employed as part of modern ECT techniques undergo transplacental passage, and case literature suggests that reduced fetal heart rate, uterine contractions, and premature labor may be adverse effects of ECT given in late pregnancy.Citation79 Randomized trials are lacking.

Complementary/alternative medicine treatments

There are very few high-quality studies of massage therapy, acupuncture, and hypnosis for treating antenatal depression.Citation80,Citation81 One randomized trial compared acupuncture (specific for depression), control acupuncture, and massage for 8 weeks in 150 women with antenatal depression. Acupuncture specific for depression was associated with significantly greater reduction in depressive symptoms than control acupuncture alone, or the two control conditions combined.Citation82 Rates of positive treatment response were also significantly higher with acupuncture specific for depression (63.0%) than the combined control conditions (44.3%).

Potential harms of treatments for maternal depression

Potential harms of treatments for maternal depression have been best studied with respect to pharmacotherapies in general and antidepressants in particular. Therefore, the current review focuses on antidepressant pharmacotherapy. presents a summary of the current US Food and Drug Administration (FDA) pregnancy-safety categories and listing of the safety category for commonly used antidepressants. Much less is known about the risks and benefits of psychotherapy or other therapies for antenatal depression.

Table 1 Summary of the current US Food and Drug Administration (FDA) pregnancy safety categories

Major congenital malformations

Major congenital malformations (MCMs) refer to structural defects present at birth that are associated with significant medical or cosmetic problems. In the US general population, the risk of MCMs is about one in every 33 infants (∼3%).Citation83 The majority of birth defects have no known etiology, while approximately a quarter are attributed to genetic, chromosomal, or cytogenetic disorders.Citation84 Only 2%–3% of birth defects are classified as teratogen-induced.Citation84 For most MCMs, the sensitive period during embryonic development occurs during the 3–8 weeks after conception, or 5–10 weeks after the last maternal menstrual period. Teratogenic drugs act by drug-specific mechanisms on developing cells and tissues, and will typically cause organ-specific defects in a consistent pattern, rather than “any” malformation.Citation85

Prior to 2000, small prospective studies showed no increased risk of MCMs in humans after in utero exposure to SSRIs,Citation86Citation90 the most common type of antidepressant used during pregnancy.Citation91 However, most MCMs are rare events, and prospective studies generally lack sufficient statistical power to detect subtle but potentially important increases in teratogenic risk. Very large observational studies using pregnancy registries or automated health outcome databases can provide useful information for estimating drug–MCM associations for antidepressants.

Most individual studies have considered fetal exposure to antidepressants as a single group or in broad classes (SSRIs, etc). The majority of individual studies have not demonstrated increased risk of MCMs associated with antidepressant exposure in the first trimester or at any time during pregnancy.Citation87,Citation89,Citation92Citation104 Meta-analyses of studies investigating the risk of antidepressants and “any” or “overall” MCMs have shown either small but statistically significant riskCitation105,Citation106 or absence of significant increase in risk ().Citation107Citation110

Table 2 Meta-analyses of observational studies of antenatal AD use and the risk of congenital malformations

Studies with larger cohorts have shown an association between first trimester antidepressant exposure and congenital heart defects, particularly septal defects.Citation93,Citation94,Citation98,Citation111Citation117 Although these results have not been replicated in several other studies,Citation51,Citation87,Citation90,Citation97,Citation100,Citation118Citation124 a small increase in the risk of any cardiac malformation or septal heart defect has been consistently reported across meta-analyses (), particularly for paroxetine,Citation105Citation107 and in one case fluoxetine.Citation108 An increased risk of heart defects when SSRIs were combined with benzodiazepines during pregnancy compared with no first trimester exposure to drugs in either class (adjusted rate difference =1.18%, 95% CI 0.18%–2.18%) has also been reported.Citation115 Neither drug class was associated with increased risk of heart defects when given individually.

Table 3 Meta-analyses of observational studies of antenatal AD use and the risk of cardiac malformations

Fewer studies of MCM risk with first trimester exposure to non-SSRI antidepressants exist. One analysis of birth registry data from Sweden examined the risk of congenital heart defects associated with maternal antidepressant use, including the use of TCAs, SSRIs, and serotonin–norepinephrine reuptake inhibitors (SNRIs), during early pregnancy, and reported an increased risk of ventricular or atrial septal defects with clomipramine (OR 2.22, 95% CI 1.29–3.82) and paroxetine (OR 2.29, 95% CI 1.28–4.09).Citation117 These results supported earlier findings by the same authors of an increased risk for any cardiovascular defect associated with early pregnancy exposure to TCAs, primarily clomipramine (OR 1.77, 95% CI 1.07–2.91).Citation125 A third study by the same group reported an increased risk of congenital heart disease in the offspring of 1,029 women who reported use of clomipramine during early pregnancy (OR 1.87, 95% CI 1.16–2.99).Citation98 Very little information on the teratogenic risk associated with other TCAs is available, though most studies have been negative.Citation126 A report from the Bupropion Pregnancy Registry maintained by the drug manufacturer documented birth outcomes among 1,213 infants with first trimester bupropion exposure, and showed no increase in the risk of any congenital malformation compared with other antidepressant exposures in the first trimester, or with bupropion exposure outside of the first trimester.Citation121 Thus far, studies of venlafaxine and mirtazapine have not shown increased risk of congenital malformations.Citation95,Citation127,Citation128

The potential for confounding by indication and detection bias in studies of antidepressant use during pregnancy and MCM risk has received intense focus. At least one very large population-based study suggested that the apparent increase in cardiac malformation risk associated with SSRIs also existed among depressed women who did not undergo drug treatment.Citation116 However, this point remains controversial, because of findings from other studies that suggest that SSRIs, not maternal depression, may account for complications.Citation6 One report has raised the possibility that higher risk of cardiac defects with paroxetine and other SSRIs may be due to detection bias. In that study, women who took SSRIs during pregnancy had 30% more prenatal ultrasound exams than women who did not take an antidepressant, while infants who had been exposed to SSRIs during pregnancy had twice as many echocardiograms in the first year of life than unexposed infants.Citation106

In summary, antidepressants in general and SSRIs more specifically have not been consistently associated with an increased risk of major congenital malformations overall. However, there appears to be a small but statistically significant risk for cardiovascular malformations (mainly septal defects) with some individual agents, such as paroxetine, and possibly also fluoxetine. Contradictory findings and additional factors make the overall literature difficult to translate to clinical practice. For example, the absolute risk with SSRIs for any cardiovascular malformation appears small, and may be susceptible to confounding (including confounding by indication) and detection bias.Citation99,Citation129 Even if the relationship between SSRIs and heart defects is causal, it is still not established if any specific agents are clearly safer than others, although paroxetine and fluoxetine have been most implicated as having higher risk than other SSRIs. Moreover, grouping together all cardiovascular or cardiac defects limits clinical translation because of the wide variety of defects under consideration that are expressed along a broad spectrum of severity and medical risk. It is unclear, for instance, if SSRIs are associated with the development of more severe forms of septal defects, including those requiring surgical management, or if in utero exposure to antidepressants affects rates of spontaneous septal defect closure. For non-SSRI antidepressants, MCM-risk estimates are even less clear. A potential relationship between clomipramine exposure and fetal cardiac defects requires further examination.

Minor congenital malformations

The term “minor congenital malformation” refers to congenital birth defects that have no major surgical or cosmetic importance. Very few studies to date have focused on the risk of minor congenital malformations associated with antidepressant use during pregnancy. Chambers et al reported no significant association between antenatal fluoxetine use and MCMs,Citation101 but also reported an increased number of exposed neonates that manifested three or more minor congenital anomalies. In a subsequent paper by Wisner et al, neither exposure to SSRIs nor maternal depression was associated with increased risk of minor physical anomalies.Citation51 A recent meta-analysis of heterogeneous studies (2,631 exposed pregnancies) showed no statistically significant increase in the risk of minor congenital malformations with SSRIs (OR 1.16, 95% CI 0.79–1.71).Citation105 The small overall sample size did not permit additional analyses of risk by individual drug.

Results of existing studies of antidepressants and the risk of minor congenital malformations can be interpreted as reassuring thus far. Although the medical significance of a given minor malformation may be limited, they are not medically inconsequential. Some minor malformations serve as risk markers for the presence of an occult MCM, particularly if three or more minor malformations are present.Citation130 As such, additional studies of minor congenital malformation risk associated with maternal use of antidepressants during pregnancy are needed.

Adverse neonatal events

Persistent pulmonary hypertension of the newborn

PPHN is caused by a failure of normal circulatory transition from a state of relatively high pressure in the pulmonary circulation before birth (to facilitate gas exchange at the placenta) to normal pulmonary artery pressure after delivery (to facilitate pulmonary gas exchange). If the normal decrease in pulmonary vascular tone does not occur, PPHN leading to hypoxemia results because of continued shunting of blood away from the lungs. Although PPHN is rare (one to two cases per 1,000 live births), severe cases are life-threatening.Citation131 Risk factors for PPHN include cesarean delivery, maternal African American or Asian race, high maternal pregravid body mass index (>27 kg/m2), maternal diabetes mellitus, maternal asthma, maternal smoking, neonatal sepsis, meconium aspiration, and gestational age 34–37 weeks and >41 weeks.Citation132

A potential association between late-pregnancy exposure to fluoxetine and PPHN was initially reported in a small prospective cohort study of 228 exposed pregnancies and 254 control pregnancies with no antenatal fluoxetine exposure.Citation86 An increased risk of PPHN was observed with late-versus first trimester fluoxetine exposure (2.7% versus 0%). A subsequent multicenter case-control study by Chambers et al involving 377 women whose infants had PPHN and 836 matched control mother–infant pairs found a sixfold-increased risk of PPHN with in utero SSRI exposure after 20 weeks’ gestation (adjusted OR 6.1, 95% CI 2.2–16.8).Citation101 Neither SSRI exposure prior to the 20th gestational week nor exposure to non-SSRI antidepressants at any point during pregnancy was associated with increased risk of PPHN. The first of two retrospective analyses of Swedish Medical Birth Register data found an association between maternal SSRI use during the early stages of pregnancy and PPHN (relative risk [RR] 2.4, 95% CI 1.2–4.3) after adjusting for maternal age, first parity, maternal body mass index, and smoking.Citation133 A more recent analysis of that data included antenatal exposures to SSRIs and non-SSRI antidepressants, and showed an increase in the risk of PPHN associated with antidepressant use in both early (RR 2.30, 95% CI 1.29–3.80) and later stages of pregnancy (RR 2.30, 95% CI 1.17–4.85).Citation94

The FDA responded to the initial positive report by issuing a public health advisory in 2006 about an increased risk of PPHN associated with maternal SSRI use after the 20th gestational week.Citation134 However, subsequent results of a retrospective cohort study of 1,104 infants exposed to antidepressants during the third trimesterCitation135 and a retrospective chart review of 24,214 deliveries (808 of which involved antepartum SSRI exposure)Citation96 did not find an association between antenatal SSRI exposure and PPHN. In a large case-control study of 11,923 births (20 with primary PPHN), cesarean delivery prior to the onset of labor, but not in utero SSRI exposure after 20 weeks’ gestation, was significantly associated with PPHN.Citation136 The largest study to date based on health registry data from five Scandinavian countries (1,618,255 singleton births) identified 33 PPHN cases among 11,014 late-pregnancy SSRI exposures, or approximately three per 1,000 infants, compared with the background PPHN rate of one to two per 1,000 births (adjusted OR 2.1, 95% CI 1.5–3.0).Citation137

In sum, it is unclear if late SSRI exposure increases the risk of PPHN, a rare but potentially serious adverse neonatal outcome. Results of positive studies have indicated an approximate two- to 2.5-fold increased risk of PPHN; however, the absolute risk appears to be very small, and the potential association between in utero antidepressants and PPHN remains controversial. If causal, the absolute risk has been estimated as three to 12 affected newborns per 1,000.Citation101,Citation137 Based in part on negative findings from more recent studies, the FDA has revised its original position, concluding that it is premature to reach definitive conclusions regarding the risk of PPHN with SSRI use in pregnancy.Citation138

Poor neonatal adaptation syndrome

The term “poor neonatal adaptation syndrome” (PNAS) refers to a constellation of behaviors and clinical signs observed in neonates (irritability, agitation, tremors, jitteriness, shivering, increased muscular tone, digestive disturbances or feeding difficulties, and respiratory distress) that has been linked with late-third trimester exposure to SSRIs and other antidepressants. PNAS is transient and generally mild in severity, requiring supportive care only. More severe cases may require that initial recovery take place in a neonatal intensive care unit.Citation139,Citation140 The pathophysiology of PNAS is thought to involve either a serotonergic rebound effect (analogous to discontinuation syndromes observed in adults who undergo abrupt withdrawal of serotonergic antidepressants) or direct serotonergic toxicity.Citation140,Citation141

An early description of PNAS was provided by Chambers et al, who observed increased rates of neonatal respiratory problems, cyanosis with feeding, jitteriness, and special care nursery admission associated with third trimester exposure to fluoxetine.Citation86 Oberlander et al reported on the incidence of transient neonatal complications associated with second and third trimester exposure to SSRIs, with or without concomitant antenatal exposure to benzodiazepine or clonazepam, among 46 exposed and 23 unexposed newborns.Citation142 Transient irritability, jitteriness, hypothermia, sleep disruption, increased muscle tone, hypothermia, and initial lack of crying were observed in nearly 30% of exposed neonates. Others have provided evidence of PNAS associated with antenatal use of TCAs and SNRIs.Citation97,Citation133,Citation143Citation147 Overall, the risk of PNAS appears to be similar between late pregnancy exposure to TCA, SNRIs, and SSRIs.Citation148,Citation149

A meta-analysisCitation150 of prospective studies that examined adverse fetal and neonatal effects associated with antenatal antidepressant exposure (totaling 1,066 mother–infant pairs) showed a trend-level association in the primary analysis between third trimester SSRI exposure and PNAS that became significant after including results of a large studyCitation151 that included infants with in utero SSRI exposure during the first and third trimesters (OR 1.99, 95% CI 1.43–2.77). A more recent meta-analysis of 12 studies (3,780 exposed infants) showed significantly increased risk of PNAS (OR 5.07, 95% CI 3.25–7.90), infant respiratory distress (OR 2.20, 95% CI 1.81–2.66), and tremors (OR 7.89, 95% CI 3.33–18.73).Citation152

Although not all studies are in agreement,Citation153,Citation154 the published literature appears to be more consistent with regard to a statistically and clinically significant association between antenatal antidepressant exposure during late pregnancy and the risk of PNAS, which may occur in 10%–30% of neonates with third trimester exposure to SSRIs or SNRIs.Citation155,Citation156 To limit the risk of PNAS occurrence, the FDA issued a recommendation in 2004 that health care providers consider tapering antidepressants used during the third trimester.Citation157 However, the effectiveness of this practice for reducing the risk of PNAS has not been established in controlled studies. One population-based study of 119,547 birth records that compared the risk of adverse neonatal outcomes according to fetal antidepressant-exposure status during the last 14 days of pregnancy showed no difference in the risk of developing clinical signs of PNAS between infants in the exposed group and propensity score-matched controls.Citation158 Further research is needed in order to determine whether suspension of effective antidepressant treatment during the late-third trimester reduces the risk of PNAS without increasing the risk of maternal depressive relapse, particularly during the postpartum period.

Adverse obstetric outcomes

Premature delivery

Premature delivery refers in general terms to the birth of an infant at <37 weeks gestational age, although prematurity can also refer to the birth of an infant before vital organs have developed sufficiently to allow postnatal survival. It is often used synonymously with a related term, preterm delivery (which refers only to delivery prior to 37 weeks’ gestation), and occasionally with early preterm delivery (eg, delivery prior to 34 weeks’ gestation). Regardless, decreased gestational age at delivery has been associated with increased neonatal morbidity and mortality compared with term-born children.Citation159,Citation160 One meta-analysis of 15 studies estimated the effects on antenatal antidepressant use on gestational age at delivery, and found a small but statistically significant difference between antidepressant-exposed and -unexposed babies (standardized mean difference [SMD] −0.23, 95% CI −0.34 to −0.12).Citation161 As referenced by the authors, this equates clinically with a difference in gestational age of 2–3 days.

With regard to premature delivery as a discrete outcome, observational studies have documented an increased risk associated with antenatal antidepressant exposure.Citation87,Citation162Citation165 However, not all studies are in agreement,Citation89,Citation90,Citation92,Citation100,Citation166,Citation167 and one study even documented a reduced risk of spontaneous preterm birth associated with medication treatment of depressive symptoms during pregnancy, although the main exposure group included both sedative medications and antidepressants.Citation32 Most studies to date have focused on antenatal SSRI use; however, an increased risk of preterm delivery has also been reported for TCAs, venlafaxine, and mirtazapine.Citation97,Citation127,Citation128

Whether or not an association between maternal antidepressant exposure and the risk of premature delivery is causal or confounded by underlying maternal depression has also been investigated, and is an ongoing focus of clinical debate.Citation168,Citation169 A prospective cohort study documented a higher risk of preterm delivery with continuous exposure to SSRIs or maternal depression across gestation compared with partial or no exposure.Citation51 Another prospective study found significantly higher rates of premature delivery and special care nursery admissions in infants born to women with antenatal depression treated with antidepressants (predominantly SSRIs) compared with antenatally depressed women with no antidepressant or only limited antidepressant use and nondepressed controls.Citation170 An inverse relationship between antidepressant dose and gestational age at delivery was observed. A very recent cohort study of 2,793 pregnant women documented an increased risk of preterm birth associated with SSRIs with or without a major depressive episode, while no increase in risk was observed for major depressive episode without SSRI use.Citation171 Moreover, a recent meta-analysis of 14 studies found an increased risk of preterm delivery associated with maternal antidepressant use during pregnancy (pooled OR 1.55, 95% CI 1.38–1.74).Citation161 Comparisons of depressed mothers with and without antidepressant exposure (five studies) showed a trend-level association with similar effect size as that of the main analysis (pooled OR 1.58, 95% CI 0.97–2.56), suggesting that the effect of antenatal antidepressant exposure on the risk of premature delivery may be independent of maternal depression. Others, however, have documented significantly higher rates of preterm delivery with antenatal SSRI exposure that attenuated after controlling for factors that may serve as proxy indicators of maternal depression severity and psychiatric comorbidity.Citation6,Citation171

In summary, there is evidence of an increased incidence of preterm delivery associated with maternal antidepressant use in some studies and a greater risk of preterm delivery in the most recent meta-analysis addressing this topic, but it is still unclear whether maternal antidepressant use increases the risk of premature delivery independently of other factors. Maternal depression severity and psychiatric comorbidity may serve as important sources of confounding in existing studies.

Spontaneous abortion

Spontaneous abortion refers to the loss of a fetus prior to the 20th gestational week in the absence of an elective termination of pregnancy or other outside intervention. Spontaneous abortion is thought to occur in 12%–15% of diagnosed pregnancies,Citation172 with higher rates (approaching 40%) in pregnant women over the age of 40 years.Citation172 Chromosomal abnormalities are the leading cause of spontaneous abortions, while thrombotic events leading to inadequate fetal blood supply and adverse maternal health factors are less common causes.Citation172,Citation173 Spontaneous abortion has been linked with both maternal depressionCitation174 and maternal use of various antidepressants during pregnancy, including SSRIs, venlafaxine, and other agents.Citation90,Citation103,Citation120,Citation127,Citation174Citation178 Other findings from mainly small prospective cohort studies have been negative.Citation86,Citation89,Citation179,Citation180

Three meta-analyses document small increases in the risk of spontaneous abortion associated with maternal antidepressant use during pregnancy. Hemels et al conducted a meta-analysis of six homogeneous cohort studies (1,534 exposed pregnancies) and reported a pooled RR of 1.45 (95% CI 1.19–1.77) and an increase in risk of 3.9% (95% CI 1.9%–6.0%) among antidepressant-exposed pregnancies versus matched control pregnancies.Citation174 A subsequent meta-analysis of nine nonheterogeneous studies (five of which reported on rates of spontaneous abortion in 1,900 exposed pregnancies) by Rahimi et al reported a summary OR of 1.7 (95% CI 1.28–2.24).Citation109 The majority of reviewed studies did not adequately address important confounding factors, including history of miscarriage, rates of induced abortion, maternal age, and severity of maternal depression.Citation126,Citation181 The most recent meta-analysis by Ross et al included three studies that met prespecified quality-assessment criteria (1,503 exposed pregnancies), and generally supported the conclusions of earlier meta-analyses by reporting a pooled OR of 1.47 (95% CI 0.99–2.17).Citation161 Comparison with a depressed but antidepressant-unexposed control group was not possible, owing to a lack of data.

Not included in the meta-analyses was a very recent population-based retrospective cohort study using data from all identified pregnancies in Denmark (1997–2008), 22,061 of which involved evidence of antenatal antidepressant use and 1,843 of which included a diagnosis of depression but no evidence of antenatal antidepressant use.Citation182 A total of 2,637 (12.0%) cases in the antidepressant-exposed group and 1,843 (11.4%) in the depressed but nonexposed group ended in spontaneous abortion (RR 1.14, 95% CI 1.10–1.18). However, after restricting the analysis to women with a registry-based diagnosis of depression, the adjusted RR for spontaneous abortion with antenatal antidepressant exposure was 1.00 (95% CI 0.80–1.24). No individual SSRIs were associated with spontaneous abortions; however, venlafaxine (RR 1.80, 95% CI 1.19–2.72), duloxetine (RR 3.12, 95% CI 1.55–6.31) and mirtazapine (RR 2.23, 95% CI 1.34–3.70) were associated with an increased risk of spontaneous abortion.

In sum, available studies suggest that antenatal antidepressant use (including SSRIs) may be associated with a small but statistically significant increase in the risk of spontaneous abortion. However, conclusions from available studies are contradictory, and the validity of the small effect sizes documented in meta-analyses and most individual studies are threatened by possible confounding. Many studies did not adequately control for important risk factors for spontaneous abortion, including history of miscarriage, maternal age, maternal smoking status, severity of maternal depression, concomitant psychiatric disorders (including substance-use disorders), other diagnoses for which antidepressants may be prescribed, and maternal use of other medications that may also elevate the risk of spontaneous abortion.Citation126 The larger risk estimates documented for some non-SSRI antidepressantsCitation176,Citation178,Citation182 may reflect greater severity of underlying depression, since these agents are not generally considered immediate first-line therapeutic options. Moreover, many studies did not clearly distinguish between spontaneous and induced abortions,Citation161,Citation181 which could have resulted in biased estimates of spontaneous abortion risk with antidepressant use, given that maternal depression itself may increase the risk of elective termination of pregnancy.Citation183,Citation184

Poor infant growth and low birth weight

Several observational studies have documented small increases in the risk of low birth weight for gestational age with maternal antidepressant use during pregnancy.Citation6,Citation86,Citation100,Citation151,Citation162,Citation164,Citation185 Others have documented linear decreases in gestational age associated with higher antidepressant doses used during pregnancy,Citation170 or increased risk of low birth weight associated with high doses of fluoxetine taken throughout pregnancy.Citation102 However, the majority of individual published studies have been negative,Citation51,Citation90,Citation100,Citation127,Citation153,Citation175,Citation179,Citation186Citation192 and one study documented a possible increase in the risk of large birth weight with the use of TCAs early in pregnancy.Citation87

Underlying maternal depression and anxiety disorders have also been associated with poor infant growth and low birth weight,Citation6,Citation30,Citation31,Citation33Citation37 raising the possibility that the relationship between maternal antidepressant use during pregnancy and the risk of low birth weight or poor fetal growth may be confounded by antidepressant indication. However, few studies accounted for maternal depression diagnosis or depression severity, or risk factors for low birth weight that may correlate significantly with maternal depression, such as smoking, alcohol use, and suboptimal maternal weight gain during pregnancy.Citation17,Citation126,Citation193Citation195 Even for studies that did account, at least partially, for these factors, results are mixed. For example, in one prospective cohort study of 174 pregnant women (46 of whom took an SSRI during pregnancy, 31 of whom were diagnosed with major depression but did not take an SSRI during pregnancy, and 97 of whom were exposed to neither depression nor an SSRI during pregnancy), no significant associations between antenatal SSRI exposure or antenatal depression and infant weight, head circumference, or length were observed compared with no antenatal exposure to SSRIs or depression.Citation196 By contrast, a small case-control study of 27 pregnant women who took antidepressant medications for diagnosed major depression and 27 matched controls with depression severity assessed using the Beck Depression Inventory (second edition)Citation197 showed an increased risk of low birth weight (OR 8.33, 95% CI 1.11–62.67) and significantly lower postpartum body weight, length, and head circumference measured at 1 month postdelivery with antenatal antidepressant exposure.Citation165 There was no significant association between depressive symptoms, smoking, or alcohol use and risk of low birth weight or subsequent infant-growth measures, suggesting an independent effect of medication exposure.

A recent meta-analysis of 20 studies (over 5,000 exposed pregnancies) with moderate heterogeneity reported a small SMD in birth weight between infants with in utero antidepressant exposure and those with no such exposure (SMD −0.10, 95% CI −0.16 to −0.03), corresponding to a difference of −74 g (95% CI −117 to −31 g).Citation161 After restricting the control group to infants born to depressed mothers with no antidepressant exposure, the SMD in birth weight was nearly zero and not statistically significant (SMD −0.02, 95% CI −0.10 to 0.06).

In sum, maternal antidepressant use during pregnancy may be associated with small decreases in birth weight, but maternal depression may mediate this relationship. Furthermore, data from the most recent meta-analysis suggest that the average difference in birth weight between antidepressant-exposed and -unexposed babies is approximately 74 g, or ∼0.16 lb.Citation161 The clinical significance of such a small difference in birth weight may be questionable, especially for birth weights that still fall within the normal range.

Preeclampsia

Gestational hypertension (blood pressure ≥140/90 mmHg on at least two separate readings spaced ≥6 hours apart after 20 weeks’ gestation occurring in women who were normotensive before pregnancy) and preeclampsia (gestational hypertension accompanied by proteinuria) collectively refer to a spectrum of hypertensive conditions that develop during pregnancy, with or without accompanying organ dysfunction.Citation198 Approximately half of gestational hypertension cases that develop prior to 30 weeks’ gestation will progress to preeclampsia.Citation198 Maternal and perinatal outcomes related to mild gestational hypertension and preeclampsia are similar to those of normotensive pregnancies.Citation199 Severe gestational hypertension, on the other hand, is associated with higher rates of cesarean delivery, placental abruption, and preterm delivery, while severe preeclampsia is associated with greater risk of perinatal mortality, placental abruption, and severe maternal morbidity and mortality compared with unaffected pregnancies.Citation200,Citation201 Risk factors for gestational hypertension and preeclampsia include advanced maternal age, nulliparity, history of hypertension or renal disease, obesity, family or personal history of preeclampsia, multifetal gestation, and history of diabetes.Citation198 More recently, maternal depression and anxiety disordersCitation202,Citation203 and greater depression severityCitation204 have been shown to increase the risk of developing preeclampsia, independently of the effects of maternal age, race, and pregravid body mass index.

A potential link between maternal antidepressant use during pregnancy and gestational hypertension, preeclampsia, or the two combined has been suggested. A cohort study by Qiu et al of pregnant women with a mood or anxiety disorder diagnosis showed a modest but not statistically significant increase in the risk of gestational hypertension associated with maternal use of SSRIs (OR 1.81, 95% CI 0.69–4.75) and SSRIs with adjunctive non-SSRI psychotropic medications (mainly TCAs and benzodiazepines, OR 2.88, 95% CI 0.40–20.98).Citation202 However, this study had low statistical power, and was not designed to investigate the effect of antidepressants on the risk of preeclampsia. Retrospective studies using much-larger cohorts have reported significant associations between maternal antidepressant use during pregnancy and the risk of gestational hypertension and/or preeclampsia.Citation94,Citation205 One nested case-control study that focused on the risk of gestational hypertension associated with use of antidepressants during pregnancy documented a significant increase in risk associated with general use of antidepressants (OR 1.52, 95% CI 1.10–2.09) and with SSRIs (OR 1.53, 95% CI 1.01–2.33) and paroxetine (OR 1.81, 95% CI 1.02–3.23) in stratified analyses.Citation206 A large retrospective cohort study of 69,448 pregnancies in women with insurance claim-based diagnoses of depression found increased risk of preeclampsia associated with SSRI (RR 1.22, 95% CI 0.97–1.54), SNRI (RR 1.95, 95% CI 1.25–3.03), and TCA monotherapy (RR 3.23, 95% CI 1.87–5.59) compared with no antidepressant use.Citation207 Similar findings were reported in comparisons between cohort members with evidence of continuous antidepressant use during pregnancy and those with evidence of antidepressant discontinuation during gestational weeks 10–24 or beyond.

In summary, available evidence suggests that pregnant women treated with antidepressants are at higher risk for gestational hypertension and preeclampsia. This includes evidence of a possible effect independent of underlying maternal depression or anxiety disorder diagnoses; however, existing studies do not address confounding by depression severity. On the one hand, findings of increasing risk of preeclampsia with SNRIs and TCAs may not be that surprising, since the choice to use these medications over first-line treatments in pregnancy may reflect greater symptom burden and treatment resistance. These findings are noteworthy, given the risk of blood pressure increases that can occur during treatment with SNRIs and selected TCAs.Citation208Citation210 Importantly, it is still unclear if antenatal antidepressant exposure is associated with severe (rather than mild) gestational hypertension or preeclampsia specifically.

Neurodevelopmental and behavioral outcomes

Several studies have examined the risk of impaired long-term neurobehavioral and cognitive development in children exposed in utero to antidepressants. Most studies have focused on the potential effects of SSRIs and SNRIs. In general, the majority of reports are from small prospective cohort studies that have shown no significant effect of maternal antenatal antidepressant use on global IQ, temperament, or language, motor, or behavioral development in offspring.Citation43,Citation186,Citation189,Citation211,Citation212 Two prospective studies showed that poor neonatal adaptation associated with antidepressant exposure in utero was not associated with subsequent developmental impairments.Citation44,Citation142 In two of the reviewed studies, maternal depression, maternal depression severity during pregnancy, and the number of depressive episodes after delivery were identified as risk factors for poorer behavioral, cognitive, and language development in offspring.Citation43,Citation44 Follow-up studies of TCA-exposed babies have also shown normal motor, behavioral, and cognitive development.Citation43,Citation144,Citation189

On the other hand, Casper et al reported a significantly higher prevalence of subtle problems with motor development and control and lower Apgar scores in offspring (up to 40 months of age) of depressed mothers who received SSRI treatment during pregnancy (31 exposed pregnancies) compared with babies of mothers diagnosed with major depression not treated with medications during pregnancy.Citation153 Alcohol consumption was reported more frequently among the exposed pregnancies. An analysis of data from the Danish National Birth Cohort that included 415 pregnancies with second or third trimester antidepressant use and 489 pregnancies with maternal depression but no antidepressant use showed delayed motor development in offspring from exposed pregnancies as assessed by telephone interview, although time required to sit and walk still fell within the normal range of development.Citation213 Children of antidepressant-treated mothers were at significantly higher risk of not being able to sit without support at 6 months of age (OR 2.1, 95% CI 1.23–3.60) or occupy themselves at 19 months of age (OR 2.1, 95% CI 1.09–4.02). Neither study was able to evaluate the effects of individual medications. Finally, a meta-analysis of observational studies reporting the effects of maternal antidepressant use on 1-minute (ten studies) and 5-minute Apgar scores (14 studies) found small but statistically significant differences between antidepressant-exposed and -unexposed babies at 1 minute (SMD −0.19, 95% CI −0.30 to −0.08) and 5 minutes (SMD −0.33, 95% CI −0.47 to −0.20).Citation161 As pointed out by the authors, this equates clinically with a difference in Apgar score of about 0.5 point at each time point.

In summary, relatively few studies have been conducted that focus on the effects of in utero antidepressant exposure on neurobehavioral, cognitive, and motor development. Most studies focused on such outcomes shortly after delivery or during early childhood. Few data are available regarding whether or not these outcomes related to cognitive or behavioral functioning persist later in life.Citation214 Many studies did not adequately control for the potential effects of maternal IQ, socioeconomic status, fetal exposure to alcohol and nicotine, duration and severity of maternal depression, and other concomitant fetal medication exposures that can also impact brain development. Data on the effects of individual antidepressant medications are also lacking. Further studies are needed to assess the independent effects of individual antidepressant medications on both early and later childhood neurobehavioral, cognitive, and motor development.

Summary and practical implications

Clinicians who treat women with antenatal depression are fortunate to have a large number of pharmacological and nonpharmacological therapeutic tools at their disposal. In spite of the wide range of treatment options and increasing recognition of the adverse effects of untreated or undertreated antenatal depression, rates of psychiatric treatment among pregnant and postpartum women with mood disorders, including major depression, are very low.Citation215 Even when women are appropriately screened, research suggests that more than 80% of women with antenatal depression do not receive treatment.Citation5 This is particularly troubling in view of the accumulating evidence suggesting that successful treatment of maternal depression leads to better outcomes for both mothers and their children, particularly if remission of depressive symptoms can be achieved.Citation48,Citation49,Citation216Citation219

It is important to recognize that several pragmatic challenges make treating antenatal depression very difficult. First, there is little information about the comparative effectiveness of many of the most commonly employed therapeutic modalities for treating major depression in pregnant women, and many patients who receive treatment with psychotherapy or antidepressant medications – whether pregnant or not – do not benefit, or experience only modest improvement in depressive symptoms and functioning. The pregnancy and developing fetus are therefore still exposed to the potentially harmful effects of residual depressive symptoms in many cases, and we are not yet able to predict with acceptable certainty which pregnant women are more likely to benefit from a given set of treatments.

Second, particularly with respect to the potential harms associated with treating antenatal depression with antidepressants, a great deal of experience and skill is required to interpret a conflicting and in some cases controversial medical literature made up almost entirely of observational studies with increasingly complex designs and statistical approaches. The complex designs and statistical approaches are needed to reduce, or at least limit to the greatest extent possible, the effects of confounding and bias inherent in nonrandomized studies. However, they also make studies more difficult to interpret, and most clinicians lack the advanced methodological training to do so. Because observational studies cannot firmly establish causal links between antidepressant use during pregnancy and adverse obstetric or fetal outcomes, no single study can be regarded as definitive. This does not mean that observational studies cannot yield data of sufficient quality to inform clinical practice. Arguments in favor of causality can be made with consistent replication of findings across numerous studies from different populations. However, this does mean that clinicians must invest considerable effort to familiarize themselves with the large volume of existing literature and stay up to date with the newest published data. The increasing time demands and rigors of daily practice erode and work against many clinicians’ ability to maintain this capability.

In spite of these challenges, we believe existing literature and clinical practice guidelines do provide useful guidance.Citation16 First, it is helpful to remember that although this review is focused on major depression, not all antenatal depression is major depressive disorder. Depressive symptoms characterize a large number of conditions that can arise during pregnancy, ranging from nonspecific reactions to stressors to major affective syndromes, including major depression and bipolar (type I or II) depression. This is not to say that other forms of maternal depression are unimportant; indeed, other depressive states and even high levels of maternal stress during pregnancy can adversely affect fetal and birth outcomes,Citation220 and many physical symptoms overlap between major depression and pregnancy, such as fatigue, sleep disruption, weight change, and concentration difficulties.Citation221 Although treatment for nonspecific or poorly characterized depressive states may be required to protect against their potential adverse effects, there is currently little or no high-quality evidence supporting the use of antidepressants or other therapies for nonspecific or poorly characterized depressive states. On the other hand, the use of antidepressants and/or structured psychotherapies for patients with a firm diagnosis of major depression in general and severe major depression in particular is clearly supported.

In addition to establishing the diagnosis of major depression, factors that bear on treatment selection and likelihood of antepartum depression relapses (for those with existing diagnoses of major depression) must also be assessed. Assessment of the former includes, at minimum, evaluation of current depressive symptom severity and impact on daily functioning, history of treatment(s) and treatment response, availability of treatment resources, medical and obstetric history, and patient preference.Citation220 Additional factors that drive treatment selection include duration, recurrence, and severity of depressive symptoms during past depressive episodes; history of self-harm tendencies; comorbid psychiatric and substance-use disorders (including alcohol, drugs of abuse, and tobacco: currently, during periods of active depression treatment, and when not receiving treatment); quality and availability of social supports; family history of major depression and treatment response; family situation; and evidence of domestic abuse or violence. Risk factors for depressive relapse during pregnancy include longer illness duration, history of recurrent illness, history of suicidality, family history of depressive illness, lack of social support, comorbid anxiety disorders, existing or impending single parenthood, low socioeconomic status, unintended pregnancy, exposure to domestic violence, and ongoing stressful life events.Citation50,Citation221

Because maternal major depression can respond positively to pharmacotherapy, psychotherapy, and other interventions, and because no single intervention is uniformly successful or devoid of risk, it is perhaps ideal for clinicians to avoid the “simple” choice of whether or not to treat antenatal depression with antidepressants in favor of beginning with a broad list of reasonable therapeutic options. Such a list can be generated by considering what treatments would have the highest chance of achieving remission in an individual patient, given a thorough assessment of the factors listed earlier. This may include nonpharmacological treatments only, antidepressant medication only, or a combination of these. As such, the initial list may be quite large, particularly for patients with limited treatment histories. Each option can be discussed with the patient and support system, mindful that there are no guarantees of positive outcome. The initial list of options can then be narrowed based on relative obstetric and fetal safety of each available service, clinical factors, and individual patient values and concerns. Fully informed decision making requires that the risks of both untreated maternal depression and the risks associated with each proposed intervention be reviewed, and that all reasonable treatment options be discussed.

Disclosure

The authors report no conflicts of interest in this work. In the past, Dr Bobo has received grants/research support from Cephalon and has served on speaker panels for Janssen Pharmaceutica and Pfizer.

References

  • KesslerRCBerglundPDemlerOThe epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R)JAMA20032893095310512813115
  • BurtVKEpidemiology of depression throughout the female life cycleJ Clin Psychiatry20026391511995779
  • BurkeKCBurkeJDRaeDSRegirerDAComparing age at onset of major depression and other psychiatric disorders by birth cohorts in five US community populationsArch Gen Psychiatry1991487897951929768
  • GavinNIGaynesBNLohrKNMeltzer-BrodySGartlehnerGSwinsonTPerinatal depression: a systematic review of prevalence and incidenceObstet Gynecol20051061071108316260528
  • MarcusSMFlynnHABlowFCBarryKLDepressive symptoms among pregnant women screened in obstetrics settingsJ Womens Health (Larchmt)20031237338012804344
  • OberlanderTFWarburtonWMisriSAghajanianJHertzmanCNeonatal outcomes after prenatal exposure to selective serotonin reuptake inhibitor antidepressants and maternal depression using population-based linked health dataArch Gen Psychiatry20066389890616894066
  • BennettHAEinarsonAKorenGEinarsonTRPrevalence of depression during pregnancy: systematic reviewObstet Gynecol200410369870915051562
  • NuttDJDavidsonJRGelenbergAJInternational consensus statement on major depressive disorderJ Clin Psychiatry201071Suppl E1e0820371035
  • CooperWOWillyMEPontSJRayWAIncreasing use of antidepressants in pregnancyAm J Obstet Gynecol200719654454517547888
  • AndradeSERaebelMABrownJUse of antidepressant medications during pregnancy: a multisite studyAm J Obstet Gynecol200819819419517905176
  • RamosEOraichiDReyEBlaisLBérardAPrevalence and predictors of antidepressant use in a cohort of pregnant womenBJOG20071141055106417565615
  • VerversTKaasenbroodHVisserGSchobbenFde Jong-van den BergLEgbertsTPrevalence and patterns of antidepressant drug use during pregnancyEur J Clin Pharmacol20066286387016896784
  • BakkerMKKöllingPvan den BergPBde WalleHEde Jong van den BergLTIncrease in use of selective serotonin reuptake inhibitors in pregnancy during the last decade, a population-based cohort study from The NetherlandsBr J Clin Pharmacol20086560060617953715
  • KuehnBMNo easy answers for physicians caring for pregnant women with depressionJAMA200930224132414242019996392
  • WisnerKLAppelbaumPSUhlKGoldkindSFPharmacotherapy for depressed pregnant women: overcoming obstacles to gathering essential dataClin Pharmacol Ther20098636236519763116
  • YonkersKAWisnerKLStewartDEThe management of depression during pregnancy: a report from the American Psychiatric Association and the American College of Obstetricians and GynecologistsObstet Gynecol200911470371319701065
  • ZuckermanBAmaroHBauchnerHCabralHDepressive symptoms during pregnancy: relationship to poor health behaviorsAm J Obstet Gynecol1989160110711112729387
  • PerkinMRBlandJMPeacockJLAndersonHRThe effect of anxiety and depression during pregnancy on obstetric complicationsBr J Obstet Gynaecol19931006296348369244
  • PritchardCWDepression and smoking in pregnancy in ScotlandJ Epidemiol Community Health1994483773827964337
  • IkardFFTomkinsSThe experience of affect as a determinant of smoking behavior: a series of validity studiesJ Abnorm Psychol1973811721814697798
  • OrrSTBlazerDGJamesSAReiterJPDepressive symptoms and indicators of maternal health status during pregnancyJ Womens Health (Larchmt)20071653554217521257
  • StoweZNHostetterALNewportDJThe onset of postpartum depression: implications for clinical screening in obstetrical and primary careAm J Obstet Gynecol200519252252615695997
  • NulmanIKorenGRovetJNeurodevelopment of children following prenatal exposure to venlafaxine, selective serotonin reuptake inhibitors, or untreated maternal depressionAm J Psychiatry20121691165117423128923
  • LindahlVPearsonJLColpeLPrevalence of suicidality during pregnancy and the postpartumArch Womens Ment Health20058778715883651
  • PearlsteinTPerinatal depression: treatment options and dilemmasJ Psychiatry Neurosci20083330231818592032
  • VintzileosAAnanthCVSmulianJCScorzaWEKnuppelRAThe impact of prenatal care on postneonatal deaths in the presence and absence of antenatal high-risk conditionsAm J Obstet Gynecol20021871258126212439516
  • KellyRHDanielsenBHGoldingJMAndersTFGilbertWMZatzickDFAdequacy of prenatal care among women with psychiatric diagnoses giving birth in California in 1994 and 1995Psychiatr Serv1999501584159010577877
  • KimDRSockolLESammelMDKellyCMoseleyMEppersonCNElevated risk of adverse obstetric outcomes in pregnant women with depressionArch Womens Ment Health20131647548223934018
  • MulderEJRobles de MedinaPGHuizinkACVan den BerghBRBuitelaarJKVisserGHPrenatal maternal stress: effects on pregnancy and the (unborn) childEarly Hum Dev20027031412441200
  • GroteNKBridgeJAGavinARMelvilleJLIyengarSKatonWJA meta-analysis of depression during pregnancy and the risk of preterm birth, low birth weight, and intrauterine growth restrictionArch Gen Psychiatry2010671012102420921117
  • SteerRASchollTOHedigerMLFischerRLSelf-reported depression and negative pregnancy outcomesJ Clin Epidemiol199245109310991474405
  • DayanJCreveuilCMarksMNPrenatal depression, prenatal anxiety, and spontaneous preterm birth: a prospective cohort study among women with early and regular carePsychosom Med20066893894617079701
  • DiegoMAFieldTHernandez-ReifMSchanbergSKuhnCGonzalez-QuinteroVHPrenatal depression restricts fetal growthEarly Hum Dev200985657018723301
  • RahmanAIqbalZBunnJLovelHHarringtonRImpact of maternal depression on infant nutritional status and illness: a cohort studyArch Gen Psychiatry20046194695215351773
  • UguzFGezgincKYaziciFAre major depression and generalized anxiety disorder associated with intrauterine growth restriction in pregnant women? A case-control studyGen Hosp Psychiatry201133640. e7e921749842
  • PaarlbergKMVingerhoetsAJPasschierJDekkerGAHeinenAGvan GeijnHPPsychosocial predictors of low birthweight: a prospective studyBr J Obstet Gynaecol199910683484110453835
  • EvansJHeronJPatelRRWilesNDepressive symptoms during pregnancy and low birth weight at termBr J Psychiatry2007191848517602131
  • MarcusSMDepression during pregnancy: rates, risks and consequences – Motherisk Update 2008Can J Clin Pharmacol200916e15e2219164843
  • GrigoriadisSVonderPortenEHMamisashviliLThe impact of maternal depression during pregnancy on perinatal outcomes: a systematic review and meta-analysisJ Clin Psychiatry201374e321e34123656857
  • McFarlandJSalisburyALBattleCLHawesKHalloranKLesterBMMajor depressive disorder during pregnancy and emotional attachment to the fetusArch Womens Ment Health20111442543421938509
  • DeaveTHeronJEvansJEmondAThe impact of maternal depression in pregnancy on early child developmentBJOG20081151043105118651886
  • PaulsonJFKeefeHALeifermanJAEarly parental depression and child language developmentJ Child Psychol Psychiatry20095025426219175819
  • NulmanIRovetJStewartDEChild development following exposure to tricyclic antidepressants or fluoxetine throughout fetal life: a prospective, controlled studyAm J Psychiatry20021591889189512411224
  • MisriSReebyePKendrickKInternalizing behaviors in 4-year-old children exposed in utero to psychotropic medicationsAm J Psychiatry20061631026103216741203
  • MurrayLSinclairDCooperPDucournauPTurnerPSteinAThe socioemotional development of 5-year-old children of postnatally depressed mothersJ Child Psychol Psychiatry1999401259127110604404
  • DowneyGCoyneJCChildren of depressed parents: an integrative reviewPsychol Bull199010850762200073
  • PilowskyDJWickramaratnePJRushAJChildren of currently depressed mothers: a STAR*D ancillary studyJ Clin Psychiatry20066712613616426099
  • WeissmanMMPilowskyDJWickramaratnePJRemissions in maternal depression and child psychopathology: a STAR*D-child reportJAMA20062951389139816551710
  • PilowskyDJWickramaratnePTalatiAChildren of depressed mothers 1 year after the initiation of maternal treatment: findings from the STAR*D-Child StudyAm J Psychiatry20081651136114718558646
  • CohenLSAltshulerLLHarlowBLRelapse of major depression during pregnancy in women who maintain or discontinue antidepressant treatmentJAMA200629549950716449615
  • WisnerKLSitDKHanusaBHMajor depression and antidepressant treatment: impact on pregnancy and neonatal outcomesAm J Psychiatry200916655756619289451
  • YonkersKAGotmanNSmithMVDoes antidepressant use attenuate the risk of a major depressive episode in pregnancy?Epidemiology20112284885421900825
  • ClaridgeAMEfficacy of systemically oriented psychotherapies in the treatment of perinatal depression: a meta-analysisArch Womens Ment Health20141731524240636
  • SockolLEEppersonCNBarberJPA meta-analysis of treatments for perinatal depressionClin Psychol Rev20113183984921545782
  • BrandonARCeccottiNHynanLSShivakumarGJohnsonNJarrettRBProof of concept: partner-assisted interpersonal psychotherapy for perinatal depressionArch Womens Ment Health20121546948023053218
  • O’MahenHHimleJAFedockGHenshawEFlynnHA pilot randomized controlled trial of cognitive behavioral therapy for perinatal depression adapted for women with low incomesDepress Anxiety20133067968723319454
  • MeagerIMilgromJGroup treatment for postpartum depression: a pilot studyAust N Z J Psychiatry1996308528609034477
  • ApplebyLWarnerRWhittonAFaragherBA controlled study of fluoxetine and cognitive-behavioural counselling in the treatment of postnatal depressionBMJ19973149329369099116
  • ChabrolHTeissedreFSaint-JeanMTeisseyreNRogeBMulletEPrevention and treatment of post-partum depression: a controlled randomized study on women at riskPsychol Med2002321039104712214785
  • GlazierRHElgarFJGoelVHolzapfelSStress, social support, and emotional distress in a community sample of pregnant womenJ Psychosom Obstet Gynaecol20042524725515715023
  • St-AndréMPsychotherapy during pregnancy: opportunities and challengesAm J Psychother1993475725908285302
  • NascimentoSLSuritaFGCecattiJGPhysical exercise during pregnancy: a systematic reviewCurr Opin Obstet Gynecol20122438739423014142
  • BlumenthalJABabyakMAMooreKAEffects of exercise training on older patients with major depressionArch Intern Med19991592349235610547175
  • BlumenthalJABabyakMADoraiswamyPMExercise and pharmacotherapy in the treatment of major depressive disorderPsychosom Med20076958759617846259
  • BabyakMBlumenthalJAHermanSExercise treatment for major depression: maintenance of therapeutic benefit at 10 monthsPsychosom Med20006263363811020092
  • Robledo-ColoniaAFSandoval-RestrepoNMosquera-ValderramaYFEscobar-HurtadoCRamírez-VélezRAerobic exercise training during pregnancy reduces depressive symptoms in nulliparous women: a randomised trialJ Physiother20125891522341377
  • GastonAPrapavessisHTired, moody and pregnant? Exercise may be the answerPsychol Health2013281353136923837826
  • SongøygardKMStafneSNEvensenKASalvesenKAVikTMørkvedSDoes exercise during pregnancy prevent postnatal depression? A randomized controlled trialActa Obstet Gynecol Scand201291626721880023
  • GoldenRNGaynesBNEkstromRDThe efficacy of light therapy in the treatment of mood disorders: a review and meta-analysis of the evidenceAm J Psychiatry200516265666215800134
  • EvenCSchroderCMFriedmanSRouillonFEfficacy of light therapy in nonseasonal depression: a systematic reviewJ Affect Disord2008108112317950467
  • OrenDAWisnerKLSpinelliMAn open trial of morning light therapy for treatment of antepartum depressionAm J Psychiatry200215966666911925310
  • EppersonCNTermanMTermanJSRandomized clinical trial of bright light therapy for antepartum depression: preliminary findingsJ Clin Psychiatry20046542142515096083
  • Wirz-JusticeABaderAFrischUA randomized, double-blind, placebo-controlled study of light therapy for antepartum depressionJ Clin Psychiatry20117298699321535997
  • KimDREppersonNPareEAn open label pilot study of transcranial magnetic stimulation for pregnant women with major depressive disorderJ Womens Health (Larchmt)20112025526121314450
  • Hızlı SayarGOztenETufanETranscranial magnetic stimulation during pregnancyArch Womens Ment Health20141731131524248413
  • MillerLJUse of electroconvulsive therapy during pregnancyHosp Community Psychiatry1994454444508045538
  • O’ReardonJPCristanchoMAvon AndreaeCVCristanchoPWeissDAcute and maintenance electroconvulsive therapy for treatment of severe major depression during the second and third trimesters of pregnancy with infant follow-up to 18 months: case report and review of the literatureJ ECT201127e23e2620562638
  • RabheruKThe use of electroconvulsive therapy in special patient populationsCan J Psychiatry20014671071911692973
  • LeiknesKACookeMJJarosch-von SchwederLHarboeIHoieBElectroconvulsive therapy during pregnancy: a systematic review of case studiesArch Womens Ment Health Epub11242013
  • DennisCLAllenKInterventions (other than pharmacological, psychosocial or psychological) for treating antenatal depressionCochrane Database Syst Rev2008CD00679518843730
  • SadoMOtaEStickleyAMoriRHypnosis during pregnancy, childbirth, and the postnatal period for preventing postnatal depressionCochrane Database Syst Rev20126CD00906222696381
  • ManberRSchnyerRNLyellDAcupuncture for depression during pregnancy: a randomized controlled trialObstet Gynecol201011551152020177281
  • RynnLCragonJCorreaAUpdate of overall prevalence of major birth defects – Atlanta, Georgia, 1978–2005MMWR Morb Mortal Wkly Rep2008571518185492
  • FinnellRHTeratology: general considerations and principlesJ Allergy Clin Immunol1999103S337S3429949334
  • MalmHPrenatal exposure to selective serotonin reuptake inhibitors and infant outcomeTher Drug Monit20123460761423042258
  • ChambersCDJohnsonKADickLMFelixRJJonesKLBirth outcomes in pregnant women taking fluoxetineN Engl J Med1996335101010158793924
  • EricsonAKällénBWiholmBDelivery outcome after the use of antidepressants in early pregnancyEur J Clin Pharmacol19995550350810501819
  • GoldsteinDJCorbinLASundellKLEffects of first-trimester fluoxetine exposure on the newbornObstet Gynecol1997897137189166307
  • KulinNAPastuszakASageSRPregnancy outcome following maternal use of the new selective serotonin reuptake inhibitors: a prospective controlled multicenter studyJAMA19982796096109486756
  • PastuszakASchick-BoschettoBZuberCPregnancy outcome following first-trimester exposure to fluoxetine (Prozac)JAMA1993269224622488474204
  • BoboWVEpsteinRAJrHayesRMThe effect of regulatory advisories on maternal antidepressant prescribing, 1995–2007: an interrupted time series study of 228,876 pregnanciesArch Womens Ment Health201417172624196827
  • NordengHvan GelderMMSpigsetOKorenGEinarsonAEberhard-GranMPregnancy outcome after exposure to antidepressants and the role of maternal depression: results from the Norwegian Mother and Child Cohort StudyJ Clin Psychopharmacol20123218619422367660
  • MalmHArtamaMGisslerMRitvanenASelective serotonin reuptake inhibitors and risk for major congenital anomaliesObstet Gynecol201111811112021646927
  • ReisMKällénBDelivery outcome after maternal use of antidepressant drugs in pregnancy: an update using Swedish dataPsychol Med2010401723173320047705
  • EinarsonAChoiJEinarsonTRKorenGIncidence of major malformations in infants following antidepressant exposure in pregnancy: results of a large prospective cohort studyCan J Psychiatry20095424224619321030
  • WichmanCLMooreKMLangTRSt SauverJLHeiseRHJrWatsonWJCongenital heart disease associated with selective serotonin reuptake inhibitor use during pregnancyMayo Clin Proc200984232719121250
  • DavisRLRubanowiceDMcPhillipsHRisks of congenital malformations and perinatal events among infants exposed to antidepressant medications during pregnancyPharmacoepidemiol Drug Saf2007161086109417729378
  • KällénBAOtterbladOPMaternal use of selective serotonin re-uptake inhibitors in early pregnancy and infant congenital malformationsBirth Defects Res A Clin Mol Teratol20077930130817216624
  • LouikCLinAEWerlerMMHernández-DíazSMitchellAAFirst-trimester use of selective serotonin-reuptake inhibitors and the risk of birth defectsN Engl J Med20073562675268317596601
  • MalmHKlaukkaTNeuvonenPJRisks associated with selective serotonin reuptake inhibitors in pregnancyObstet Gynecol20051061289129616319254
  • ChambersCDHernandez-DiazSVan MarterLJSelective serotonin-reuptake inhibitors and risk of persistent pulmonary hypertension of the newbornN Engl J Med200635457958716467545
  • HendrickVSmithLMSuriRHwangSHaynesDAltshulerLBirth outcomes after prenatal exposure to antidepressant medicationAm J Obstet Gynecol200318881281512634662
  • Klieger-GrossmannCWeitznerBPanchaudAPregnancy outcomes following use of escitalopram: a prospective comparative cohort studyJ Clin Pharmacol20125276677022075232
  • ManakovaEHubickovaLAntidepressant drug exposure during pregnancy. CZTIS small prospective studyNeuro Endocrinol Lett201132Suppl 1535622167208
  • MylesNNewallHWardHLargeMSystematic meta-analysis of individual selective serotonin reuptake inhibitor medications and congenital malformationsAust N Z J Psychiatry2013471002101223761574
  • Bar-OzBEinarsonTEinarsonAParoxetine and congenital malformations: meta-analysis and consideration of potential confounding factorsClin Ther20072991892617697910
  • GrigoriadisSVonderPortenEHMamisashviliLAntidepressant exposure during pregnancy and congenital malformations: is there an association? A systematic review and meta-analysis of the best evidenceJ Clin Psychiatry201374e293e30823656855
  • RigginLFrankelZMorettiMPupcoAKorenGThe fetal safety of fluoxetine: a systematic review and meta-analysisJ Obstet Gynaecol Can20133536236923660045
  • RahimiRNikfarSAbdollahiMPregnancy outcomes following exposure to serotonin reuptake inhibitors: a meta-analysis of clinical trialsReprod Toxicol20062257157516720091
  • AddisAKorenGSafety of fluoxetine during the first trimester of pregnancy: a meta-analytical review of epidemiological studiesPsychol Med200030899410722179
  • BakkerMKKerstjens-FrederikseWSBuysCHde WalleHEde Jong-van den BergLTFirst-trimester use of paroxetine and congenital heart defects: a population-based case-control studyBirth Defects Res A Clin Mol Teratol2010889410019937603
  • BérardARamosEReyEBlaisLSt-AndréMOraichiDFirst trimester exposure to paroxetine and risk of cardiac malformations in infants: the importance of dosageBirth Defects Res B Dev Reprod Toxicol200780182717187388
  • MerlobPBirkESirotaLAre selective serotonin reuptake inhibitors cardiac teratogens? Echocardiographic screening of newborns with persistent heart murmurBirth Defects Res A Clin Mol Teratol20098583784119691085
  • PedersenLHHenriksenTBVestergaardMOlsenJBechBHSelective serotonin reuptake inhibitors in pregnancy and congenital malformations: population based cohort studyBMJ2009339b356919776103
  • OberlanderTFWarburtonWMisriSRiggsWAghajanianJHertzmanCMajor congenital malformations following prenatal exposure to serotonin reuptake inhibitors and benzodiazepines using population-based health dataBirth Defects Res B Dev Reprod Toxicol200883687618293409
  • Jimenez-SolemEAndersenJTPetersenMExposure to selective serotonin reuptake inhibitors and the risk of congenital malformations: a nationwide cohort studyBMJ Open20122e001148
  • KällénBOtterbladOPAntidepressant drugs during pregnancy and infant congenital heart defectReprod Toxicol20062122122216406480
  • EinarsonAPistelliADeSantisMEvaluation of the risk of congenital cardiovascular defects associated with use of paroxetine during pregnancyAm J Psychiatry200816574975218381907
  • ReisMKällénBCombined use of selective serotonin reuptake inhibitors and sedatives/hypnotics during pregnancy: risk of relatively severe congenital malformations or cardiac defects. A register studyBMJ Open20133e002166
  • Diav-CitrinOShechtmanSWeinbaumDParoxetine and fluoxetine in pregnancy: a prospective, multicentre, controlled, observational studyBr J Clin Pharmacol20086669570518754846
  • ColeJAModellJGHaightBRCosmatosISStolerJMWalkerAMBupropion in pregnancy and the prevalence of congenital malformationsPharmacoepidemiol Drug Saf20071647448416897811
  • RamosESt-AndréMReyEOraichiDBerardADuration of antidepressant use during pregnancy and risk of major congenital malformationsBr J Psychiatry200819234435018450657
  • NulmanIKorenGThe safety of fluoxetine during pregnancy and lactationTeratology1996533043088879088
  • WogeliusPNørgaardMGislumMMaternal use of selective serotonin reuptake inhibitors and risk of congenital malformationsEpidemiology20061770170417028507
  • KällénBAOlaussonPOMaternal drug use in early pregnancy and infant cardiovascular defectReprod Toxicol20031725526112759093
  • UdechukuANguyenTHillRSzegoKAntidepressants in pregnancy: a systematic reviewAust N Z J Psychiatry20104497899621034181
  • DjulusJKorenGEinarsonTRExposure to mirtazapine during pregnancy: a prospective, comparative study of birth outcomesJ Clin Psychiatry2006671280128416965209
  • LennestalRKällénBDelivery outcome in relation to maternal use of some recently introduced antidepressantsJ Clin Psychopharmacol20072760761318004128
  • SackettJCWellerRAWellerEBSelective serotonin reuptake inhibitor use during pregnancy and possible neonatal complicationsCurr Psychiatry Rep20091125325719470288
  • AdamsMHudginsLThe importance of minor anomalies in the evaluation of the newbornNeoreviews20034e99e104
  • Walsh-SukysMCTysonJEWrightLLPersistent pulmonary hypertension of the newborn in the era before nitric oxide: practice variation and outcomesPediatrics2000105142010617698
  • Hernández-DíazSVan MarterLJWerlerMMLouikCMitchellAARisk factors for persistent pulmonary hypertension of the newbornPediatrics2007120e272e28217671038
  • KällénBOlaussonPOMaternal use of selective serotonin re-uptake inhibitors and persistent pulmonary hypertension of the newbornPharmacoepidemiol Drug Saf20081780180618314924
  • US Food and Drug AdministrationPublic health advisory: treatment challenges of depression in pregnancy and the possibility of persistent pulmonary hypertension in newborns2013 Available from: http://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/drugsafetyinformationforheathcareprofessionals/publichealthadvisories/ucm124348.htmAccessed August 6, 2014
  • AndradeSEMcPhillipsHLorenDAntidepressant medication use and risk of persistent pulmonary hypertension of the newbornPharmacoepidemiol Drug Saf20091824625219148882
  • WilsonKLZeligCMHarveyJPCunninghamBSDolinskyBMNapolitanoPGPersistent pulmonary hypertension of the newborn is associated with mode of delivery and not with maternal use of selective serotonin reuptake inhibitorsAm J Perinatol201128192420607643
  • KielerHArtamaMEngelandASelective serotonin reuptake inhibitors during pregnancy and risk of persistent pulmonary hypertension in the newborn: population based cohort study from the five Nordic countriesBMJ2012344d801222240235
  • US Food and Drug AdministrationFDA drug safety communication: selective serotonin reuptake inhibitor (SSRI) antidepressant use during pregnancy and reports of a rare heart and lung condition in newborn babies2011 Available from: http://www.fda.gov/drugs/drugsafety/ucm283375.htmAccessed August 6, 2014
  • KalraSEinarsonAKorenGTaking antidepressants during late pregnancy. How should we advise women?Can Fam Physician2005511077107816121827
  • Moses-KolkoELBogenDPerelJNeonatal signs after late in utero exposure to serotonin reuptake inhibitors: literature review and implications for clinical applicationsJAMA20052932372238315900008
  • IsbisterGKDawsonAWhyteIMPriorFHClancyCSmithAJNeonatal paroxetine withdrawal syndrome or actually serotonin syndrome?Arch Dis Child Fetal Neonatal Ed200185F147F14811561552
  • OberlanderTFMisriSFitzgeraldCEKostarasXRurakDRiggsWPharmacologic factors associated with transient neonatal symptoms following prenatal psychotropic medication exposureJ Clin Psychiatry20046523023715003078
  • WayCMSafety of newer antidepressants in pregnancyPharmacotherapy20072754655217381382
  • MisriSSivertzKTricyclic drugs in pregnancy and lactation: a preliminary reportInt J Psychiatry Med1991211571711894455
  • SchimmellMSKatzEZShaagYPastuszakAKorenGToxic neonatal effects following maternal clomipramine therapyJ Toxicol Clin Toxicol1991294794841749054
  • CoweLLloydDJDawlingSNeonatal convulsions caused by withdrawal from maternal clomipramineBr Med J (Clin Res Ed)198228418371838
  • FerreiraECarcellerAMAgogueCEffects of selective serotonin reuptake inhibitors and venlafaxine during pregnancy in term and preterm neonatesPediatrics2007119525917200271
  • Ter HorstPGJansmanFGvan LingenRASmitJPde Jong-van den BergLTBrouwersJRPharmacological aspects of neonatal antidepressant withdrawalObstet Gynecol Surv20086326727918348740
  • BoucherNKorenGBeaulac-BaillargeonLMaternal use of venlafaxine near term: correlation between neonatal effects and plasma concentrationsTher Drug Monit20093140440919455083
  • LattimoreKADonnSMKacirotiNKemperARNealCRJrVazquezDMSelective serotonin reuptake inhibitor (SSRI) use during pregnancy and effects on the fetus and newborn: a meta-analysisJ Perinatol20052559560416015372
  • KällénBNeonate characteristics after maternal use of antidepressants in late pregnancyArch Pediatr Adolesc Med200415831231615066868
  • GrigoriadisSVonderPortenEHMamisashviliLThe effect of prenatal antidepressant exposure on neonatal adaptation: a systematic review and meta-analysisJ Clin Psychiatry201374e309e32023656856
  • CasperRCFleisherBELee-AncajasJCFollow-up of children of depressed mothers exposed or not exposed to antidepressant drugs during pregnancyJ Pediatr200314240240812712058
  • MaschiSClavennaACampiRSchiavettiBBernatMBonatiMNeonatal outcome following pregnancy exposure to antidepressants: a prospective controlled cohort studyBJOG200811528328917903222
  • GrigoriadisSMamisashviliLRossLEAchieving the balance: treating depressed pregnant women with antidepressantsJ Clin Psychiatry20137437537623656844
  • KorenGNordengHAntidepressant use during pregnancy: the benefit-risk ratioAm J Obstet Gynecol201220715716322425404
  • US Food and Drug AdministrationFDA MedWatch drug alert on Effexor and SSRIs2004 Available from: http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm154975.htmAccessed August 6, 2014
  • WarburtonWHertzmanCOberlanderTFA register study of the impact of stopping third trimester selective serotonin reuptake inhibitor exposure on neonatal healthActa Psychiatr Scand201012147147919878137
  • EngleWAMorbidity and mortality in late preterm and early term newborns: a continuumClin Perinatol20113849351621890021
  • Shapiro-MendozaCKLackritzEMEpidemiology of late and moderate preterm birthSemin Fetal Neonatal Med20121712012522264582
  • RossLEGrigoriadisSMamisashviliLSelected pregnancy and delivery outcomes after exposure to antidepressant medication: a systematic review and meta-analysisJAMA Psychiatry20137043644323446732
  • SimonGECunninghamMLDavisRLOutcomes of prenatal antidepressant exposureAm J Psychiatry20021592055206112450956
  • LundNPedersenLHenriksenTSSRI exposure in utero and pregnancy outcomeJ Matern Fetal Neonatal Med20061945
  • WenSWYangQGarnerPSelective serotonin reuptake inhibitors and adverse pregnancy outcomesAm J Obstet Gynecol200619496196616580283
  • LewisAJGalballyMOpieGBuistANeonatal growth outcomes at birth and one month postpartum following in utero exposure to antidepressant medicationAust N Z J Psychiatry20104448248720397792
  • PearsonKHNonacsRMVigueraACBirth outcomes following prenatal exposure to antidepressantsJ Clin Psychiatry2007681284128917854255
  • EinarsonAChoiJEinarsonTRKorenGAdverse effects of antidepressant use in pregnancy: an evaluation of fetal growth and preterm birthDepress Anxiety201027353819691030
  • PalmstenKHernández-DíazSCan nonrandomized studies on the safety of antidepressants during pregnancy convincingly beat confounding, chance, and prior beliefs?Epidemiology20122368668822872113
  • DavalosDBYadonCATregellasHCUntreated prenatal maternal depression and the potential risks to offspring: a reviewArch Womens Ment Health20121511422215285
  • SuriRAltshulerLHellemannGBurtVKAquinoAMintzJEffects of antenatal depression and antidepressant treatment on gestational age at birth and risk of preterm birthAm J Psychiatry20071641206121317671283
  • YonkersKANorwitzERSmithMVDepression and serotonin reuptake inhibitor treatment as risk factors for preterm birthEpidemiology20122367768522627901
  • García-EnguídanosACalleMEValeroJLunaSDomínguez-RojasVRisk factors in miscarriage: a reviewEur J Obstet Gynecol Reprod Biol200210211111911950476
  • GoddijnMLeschotNJGenetic aspects of miscarriageBaillieres Best Pract Res Clin Obstet Gynaecol20001485586511023805
  • HemelsMEEinarsonAKorenGLanctotKLEinarsonTRAntidepressant use during pregnancy and the rates of spontaneous abortions: a meta-analysisAnn Pharmacother20053980380915784808
  • EinarsonAFatoyeBSarkarMPregnancy outcome following gestational exposure to venlafaxine: a multicenter prospective controlled studyAm J Psychiatry20011581728173011579012
  • Chun-Fai-ChanBKorenGFayezIPregnancy outcome of women exposed to bupropion during pregnancy: a prospective comparative studyAm J Obstet Gynecol200519293293615746694
  • EinarsonAChoiJEinarsonTRKorenGRates of spontaneous and therapeutic abortions following use of antidepressants in pregnancy: results from a large prospective databaseJ Obstet Gynaecol Can20093145245619604427
  • Nakhai-PourHRBroyPBérardAUse of antidepressants during pregnancy and the risk of spontaneous abortionCMAJ20101821031103720513781
  • EinarsonABonariLVoyer-LavigneSA multicentre prospective controlled study to determine the safety of trazodone and nefazodone use during pregnancyCan J Psychiatry20034810611012655908
  • McElhattonPRGarbisHMElefantEThe 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 Toxicol1996102852948829251
  • TuccoriMTestiAAntonioliLSafety concerns associated with the use of serotonin reuptake inhibitors and other serotonergic/noradrenergic antidepressants during pregnancy: a reviewClin Ther200931Pt 11426145319698902
  • KjaersgaardMIParnerETVestergaardMPrenatal antidepressant exposure and risk of spontaneous abortion – a population-based studyPLoS One20138e7209524015208
  • WalfischASermerCMatokIEinarsonAKorenGPerception of teratogenic risk and the rated likelihood of pregnancy termination: association with maternal depressionCan J Psychiatry20115676176722152645
  • BonariLPintoNAhnEEinarsonASteinerMKorenGPerinatal risks of untreated depression during pregnancyCan J Psychiatry20044972673515633850
  • OberlanderTFWarburtonWMisriSAghajanianJHertzmanCEffects of timing and duration of gestational exposure to serotonin reuptake inhibitor antidepressants: population-based studyBr J Psychiatry200819233834318450656
  • HeikkinenTEkbladUKeroPEkbladSLaineKCitalopram in pregnancy and lactationClin Pharmacol Ther20027218419112189365
  • SivojelezovaAShuhaiberSSarkissianLEinarsonAKorenGCitalopram use in pregnancy: prospective comparative evaluation of pregnancy and fetal outcomeAm J Obstet Gynecol20051932004200916325604
  • CosteiAMKozerEHoTItoSKorenGPerinatal outcome following third trimester exposure to paroxetineArch Pediatr Adolesc Med20021561129113212413342
  • NulmanIRovetJStewartDENeurodevelopment of children exposed in utero to antidepressant drugsN Engl J Med19973362582628995088
  • CohenLSHellerVLBaileyJWGrushLAblonJSBouffardSMBirth outcomes following prenatal exposure to fluoxetineBiol Psychiatry200048996100011082474
  • SuriRAltshulerLHendrickVRasgonNLeeEMintzJThe impact of depression and fluoxetine treatment on obstetrical outcomeArch Womens Ment Health2004719320015241665
  • LaineKHeikkinenTEkbladUKeroPEffects of exposure to selective serotonin reuptake inhibitors during pregnancy on serotonergic symptoms in newborns and cord blood monoamine and prolactin concentrationsArch Gen Psychiatry20036072072612860776
  • WeinbergerAHMazureCMMorlettAMcKeeSATwo decades of smoking cessation treatment research on smokers with depression: 1990–2010Nicotine Tob Res2013151014103123100459
  • DavidsonKMRitsonEBThe relationship between alcohol dependence and depressionAlcohol Alcohol1993281471558517885
  • BodnarLMWisnerKLMoses-KolkoESitDKHanusaBHPrepregnancy body mass index, gestational weight gain, and the likelihood of major depressive disorder during pregnancyJ Clin Psychiatry2009701290129619607761
  • WisnerKLBogenDLSitDDoes fetal exposure to SSRIs or maternal depression impact infant growth?Am J Psychiatry201317048549323511234
  • BeckATSteerRABrownGKBeck Depression Inventory2nd edSan AntonioPsychological Corporation1996
  • SibaiBHDiagnosis and management of gestational hypertension and preeclampsiaObstet Gynecol200310218119212850627
  • PodymowTAugustPHypertension in pregnancyAdv Chronic Kidney Dis20071417819017395120
  • GroftonENCapewellVNataleRGrattonRJObstetrical intervention rates and maternal and neonatal outcomes of women with gestational hypertensionAm J Obstet Gynecol200118579880311641654
  • SteegersEAvon DadelszenPDuvekotJJPijneborgRPre-eclampsiaLancet201037663164420598363
  • QiuCWilliamsMACalderon-MargalitRCripeSMSorensenTKPreeclampsia risk in relation to maternal mood and anxiety disorders diagnosed before or during early pregnancyAm J Hypertens20092239740219197246
  • KurkiTHiilesmaaVRaitasaloRMattilaHYlikorkalaODepression and anxiety in early pregnancy and risk for preeclampsiaObstet Gynecol20009548749010725477
  • QiuCSanchezSELamNGarciaPWilliamsMAAssociations of depression and depressive symptoms with preeclampsia: results from a Peruvian case-control studyBMC Womens Health200771517900360
  • TohSMitchellAALouikCWerlerMMChambersCDHernández-DíazSSelective serotonin reuptake inhibitor use and risk of gestational hypertensionAm J Psychiatry200916632032819122006
  • De VeraMABérardAAntidepressant use during pregnancy and the risk of pregnancy-induced hypertensionBr J Clin Pharmacol20127436236922435711
  • PalmstenKSetoguchiSMargulisAVPatrickARHernández-DíazSElevated risk of preeclampsia in pregnant women with depression: depression or antidepressants?Am J Epidemiol201217598899722442287
  • ThaseMEffects of venlafaxine on blood pressure: a meta-analysis of original data from 3,744 depressed patientsJ Clin Psychiatry1998595025099818630
  • WalshBTHadiganCMWongLMIncreased pulse and blood pressure associated with desipramine treatment of bulimia nervosaJ Clin Psychopharmacol19921231631681629381
  • GoldbergJFErnstCLHypertensionGoldergJFErnstCLManaging the Side Effects of Psychotropic MedicationsWashingtonAmerican Psychiatric2012137138
  • ReebyePNMorisonSJPanikkarHMisriSGrunauREAffect expression in prenatally psychotropic exposed and nonexposed mother-infant dyadsInfant Ment Health J200223403416
  • OberlanderTFReebyePMisriSPapsdorfMKimJGrunauREExternalizing and attentional behaviors in children of depressed mothers treated with a selective serotonin reuptake inhibitor antidepressant during pregnancyArch Pediatr Adolesc Med2007161222917199063
  • PedersenLHHenriksenTBOlsenJFetal exposure to antidepressants and normal milestone development at 6 and 19 months of agePediatrics2010125e600e60820176667
  • SieSDWenninkJMvan DrielJJMaternal use of SSRIs, SNRIs and NaSSAs: practical recommendations during pregnancy and lactationArch Dis Child Fetal Neonatal Ed201297F472F47623080479
  • Vesga-LópezOBlancoCKeyesKOlfsonMGrantBFHasinDSPsychiatric disorders in pregnant and postpartum women in the United StatesArch Gen Psychiatry20086580581518606953
  • WickramaratnePGameroffMJPilowskyDJChildren of depressed mothers 1 year after remission of maternal depression: findings from the STAR*D-Child studyAm J Psychiatry201116859360221406462
  • FosterCEWebsterMCWeissmanMMRemission of maternal depression: relations to family functioning and youth internalizing and externalizing symptomsJ Clin Child Adolesc Psychol20083771472418991123
  • CoiroMJRIleyABroitmanMMirandaJEffects on children of treating their mothers’ depression: results of a 12-month follow-upPsychiatr Serv20126335736322388476
  • GunlicksMLWeissmanMMChange in child psychopathology with improvement in parental depression: a systematic reviewJ Am Acad Child Adolesc Psychiatry20084737938918388766
  • WisnerKLZarinDAHolmboeESRisk-benefit decision making for treatment of depression during pregnancyAm J Psychiatry20001571933194011097953
  • ChaudronLHComplex challenges in treating depression during pregnancyAm J Psychiatry2013170122023288385