Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jul 21.
Published in final edited form as: J Clin Psychopharmacol. 2014 Apr;34(2):244–255. doi: 10.1097/JCP.0000000000000087

Pharmacotherapy for Mood Disorders in Pregnancy

A Review of Pharmacokinetic Changes and Clinical Recommendations for Therapeutic Drug Monitoring

Kristina M Deligiannidis *,†,, Nancy Byatt ‡,§, Marlene P Freeman
PMCID: PMC4105343  NIHMSID: NIHMS600935  PMID: 24525634

Abstract

Objective

Pharmacotherapy for mood disorders during pregnancy is often complicated by pregnancy-related pharmacokinetic changes and the need for dose adjustments. The objectives of this review are to summarize the evidence for change in perinatal pharmacokinetics of commonly used pharmacotherapies for mood disorders, discuss the implications for clinical and therapeutic drug monitoring (TDM), and make clinical recommendations.

Methods

The English-language literature indexed on MEDLINE/PubMed was searched for original observational studies (controlled and uncontrolled, prospective and retrospective), case reports, and case series that evaluated or described pharmacokinetic changes or TDM during pregnancy or the postpartum period.

Results

Pregnancy-associated changes in absorption, distribution, metabolism, and elimination may result in lowered psychotropic drug levels and possible treatment effects, particularly in late pregnancy. Mechanisms include changes in both phase 1 hepatic cytochrome P450 and phase 2 uridine diphosphate glucuronosyltransferase enzyme activities, changes in hepatic and renal blood flow, and glomerular filtration rate. Therapeutic drug monitoring, in combination with clinical monitoring, is indicated for tricyclic antidepressants and mood stabilizers during the perinatal period.

Conclusions

Substantial pharmacokinetic changes can occur during pregnancy in a number of commonly used antidepressants and mood stabilizers. Dose increases may be indicated for antidepressants including citalopram, clomipramine, imipramine, fluoxetine, fluvoxamine, nortriptyline, paroxetine, and sertraline, especially late in pregnancy. Antenatal dose increases may also be needed for lithium, lamotrigine, and valproic acid because of perinatal changes in metabolism. Close clinical monitoring of perinatal mood disorders and TDM of tricyclic antidepressants and mood stabilizers are recommended.

Keywords: pharmacokinetics, pregnancy, antidepressants, mood stabilizers


Untreated mood disorders during pregnancy are associated with health risks to both mother and fetus, making the goal of euthymia paramount.14 Guidelines for the management of major depressive disorder and bipolar disorder during pregnancy stress the importance of preconception treatment planning and close clinical monitoring.2,5 Most women who discontinue maintenance antidepressants or mood-stabilizer pharmacotherapy for a pregnancy will experience a relapse during the pregnancy. Therefore, immediate and prophylactic treatment of many women with mood disorders will include pharmacologic treatment.6,7 Treatment during pregnancy is complicated by pharmacokinetic changes, which can result in lowered psychotropic drug levels and/or treatment efficacy.

Therapeutic drug monitoring (TDM) is an integral aspect of the standard of care for medications with established therapeutic ranges, a narrow therapeutic index or significant pharmacokinetic variability, which includes some mood stabilizers and tricyclic antidepressants (TCAs). Given the evidence for changes in perinatal physiology and pharmacokinetics, TDM has the potential to optimize dosing by avoiding supratherapeutic doses that would increase drug exposure to the mother and fetus or subtherapeutic dosing that would expose the dyad to the consequences of undertreated psychiatric illness. A comprehensive discussion that includes the potential risks of undertreated mental illness for the mother and fetus, benefits of nonpharmacologic and potential risks, and benefits to psychopharmacologic treatment816 is essential to quality patient care. In cases of polypharmacy, TDM can be valuable in avoiding adverse effects due to drug-to-drug interactions. For medications without standardized therapeutic ranges, an individual woman’s drug level at a time when she is clinically asymptomatic that can be used as a target serum drug level during pregnancy may be useful.17,18

The objectives of this review are to summarize the evidence for changes in perinatal pharmacokinetics of psychotropic medications commonly used for mood disorders during pregnancy, consider the implications for clinical and TDM, and make clinical recommendations.

MATERIALS AND METHODS

The English-language literature indexed on MEDLINE/PubMed was searched for the period between 1966 and 2013 using the following key terms: antidepressant, fluoxetine, sertraline, paroxetine, citalopram, escitalopram, fluvoxamine, venlafaxine, desvenlafaxine, duloxetine, bupropion, mirtazapine, trazodone, vilazodone, amitriptyline, clomipramine, desipramine, doxepin, imipramine, nortriptyline, protriptyline, trimipramine, amoxapine, maprotiline, selective serotonin reuptake inhibitor (SSRI), selective norepinephrine reuptake inhibitor (SNRI), tricyclic, tetracyclic, mood stabilizer, antiepileptic, lithium, lamotrigine (LTG), carbamazepine (CBZ), valproic acid (VPA), pregnancy, prenatal, delivery, post-partum, female, gender, pharmacokinetics, dosing, therapeutic drug monitoring, and drug level. Resultant articles were cross-referenced for other relevant articles not identified in the initial search. Original observational studies, case reports, and case series that evaluated or described pharmacokinetic changes or TDM during pregnancy or the postpartum were included.

RESULTS

Pharmacokinetics During the Perinatal Period

The drug dose–effect relationship is complex, including not only interperson pharmacokinetic and pharmacodynamic variability but also genetic variability controlling the translation of proteins involved in metabolizing enzymes, drug transporters, and drug targets. Small-to-moderate sex differences, which exist in drug absorption, distribution, metabolism, and elimination,1921 may be amplified during pregnancy. For example, women have a slower gastric emptying22 and small bowel and colonic transit time when compared with men, which are further slowed during pregnancy.23,24 The increased plasma volume, change in protein binding, and lower ratio of lean muscle to adipose tissue in pregnant women may result in a greater volume of drug distribution for lipophilic drugs. Together, these changes likely contribute to a small effect in the peak plasma concentrations.19 Hepatic clearance of psychotropic medications is also altered, most notably due to pregnancy-associated changes in metabolic enzymes. Of the multiple families of metabolic enzymes,25 the phase 1 metabolism cytochrome P450 (CYP) family has been best studied in pregnancy. Individual drugs often undergo metabolism by several CYP and/or non-CYP pathways. Other families of enzymes include phase 2 metabolism enzymes such as uridine diphosphate glucuronosyltransferase (UGT) and N-acetyltransferase. Increased sex steroids associated with pregnancy may modulate several of the CYP450 and UGT isoforms in a clinically relevant manner. Synthetic analogs of sexsteroids associated with pregnancy, such as those used in hormone replacement therapy or oral contraceptives, modulate several CYP450 isoforms via inhibition (CYP1A2,26 CYP2C19,27 CYP2B6,28 and CYP3A429), induction (CYP2A630), or increasing glucuronidation by UGT1A4 and possibly UGT2B7.31,32 Cytochrome P450 and UGT changes may also have effects on adverse effect burden for both mother and fetus/infant because drug exposure partially depends on metabolism. Changes in maternal drug pharmacokinetics in conjunction with placental transfer and fetal drug metabolism affect fetal psychotropic drug exposure.3341

Cytochrome P1A2 is less active in women compared with men42 and is inhibited by sex steroids.43,44 The activity of CYP1A2 is reduced by 65% to 70% at the end of pregnancy compared with the postpartum period32 and may affect psychotropics metabolized by CYP1A2 including fluvoxamine, duloxetine, amitriptyline, clomipramine, desmethylimipramine, imipraminem, and doxepin.

Greater CYP3A4 activity in women (via progesterone effects)42,45 has the potential to accelerate metabolism and reduce plasma levels of some commonly used psychotropics partially metabolized via CYP3A4 including citalopram, escitalopram, fluoxetine, paroxetine, trazodone, venlafaxine, desvenlafaxine, bupropion, mirtazapine, amitriptyline, clomipramine, imipramine, trimipramine, doxepin, and CBZ.

During pregnancy, CYP2C19 activity is reduced by almost 50%.46 Pharmacokinetic changes associated with CYP2C19 during pregnancy may affect the metabolism of citalopram, escitalopram, sertraline, fluoxetine, vilazodone, venlafaxine, amitriptyline, clomipramine, trimipramine, imipramine, and desmethylimipramine.47

Cytochrome P2D6 is generally induced during pregnancy,32,48 and CYP2D6-associated pharmacokinetic changes may affect the metabolism of numerous psychotropics from the SSRIs, SNRIs, and TCA drug classes. Maternal CYP metabolic phenotype (eg, poor, intermediate, extensive, and ultrarapid) is an important determinate of metabolism. Different allelic forms of CYP2D6 may result in differing therapeutic dosing requirements. In nonpregnant patients, dose adjustments may be required based in part on CYP2D6 phenotype.49 In pregnancy, phenotype influences metabolic ratios of medications biotransformed by CYP2D6.48

Increased renal blood flow and the associated increase in glomerular filtration rate (GFR) may increase drug (eg, lithium) elimination during pregnancy.50 Although the studies are inconsistent,51,52 increased hepatic blood flow may account for increased clearance and decreased concentration of high extraction ratio drugs during pregnancy.

The postpartum period is characterized by a rapid decline in sex steroid levels, contraction of plasma volume, reestablishment of hepatic enzyme activity after a period of metabolic refractory activity,5356 and a return of the GFR to prepregnancy levels. As a consequence, increased drug blood levels may result and manifest as adverse effects of toxicity, especially when an increased dose used during pregnancy is continued into the postpartum.57

In addition to changes in maternal pharmacokinetics, patient-specific psychologic factors and physiologic effects of pregnancy on vulnerability and treatment responsiveness may also influence treatment outcomes. Pregnancy brings about a psychosocial context, which differs among women, aspects of which can affect the risk of mood disorder recurrence during pregnancy or treatment efficacy.58 Physiologic changes associated with pregnancy may interact with underlying sexual dimorphisms in the localization and concentration of endogenous neurotransmitters and their degradative enzymes and transporters, having the potential to clinically affect antidepressant pharmacodynamics (eg, drug-receptor interactions), particularly if they are sex steroid hormone responsive.5962

Evidence for Change in Perinatal Phase 1 Metabolism: SSRIs

Some women will experience increased metabolism of SSRIs in late pregnancy and may require higher dosing, especially during the third trimester to maintain clinical benefits. Fluoxetine is predominantly metabolized by CYP2C9 to its active metabolite norfluoxetine, which is also pharmacologically active63,64; CYP3A4, CYP2D6, and CYP2C19 additionally contribute to fluoxetine’s metabolism.6367 Mean fluoxetine-metabolite ratio levels decrease between 20 to 26 weeks and 30 to 36 weeks’ gestation, suggesting increased clearance during pregnancy, whereas postpartum fluoxetine-metabolite ratio levels increase, suggesting reduced clearance after delivery.56 Similarly, higher maternal serum concentrations of fluoxetine and norfluoxetine in the postpartum period, compared with the third trimester, have been reported in women taking the same fluoxetine dose during both periods.68 Decreased albumin levels in pregnancy may result in low plasma trough concentrations of highly protein-bound fluoxetine and norfluoxetine. Pregnancy-associated induced demethylation of fluoxetine by CYP2D6 may contribute to lower trough levels in pregnancy compared with the postpartum period.69

Sertraline and its main weakly active metabolite N-desmethylsertraline are substrates of several P450 enzymes, including CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. Sertraline dose increases are often required early in the third trimester to treat emergent depressive symptoms or maintain euthymia70 with some women experiencing increased drug metabolism from second to third trimester.71

Paroxetine has no known pharmacologically active metabolites and is predominantly metabolized via CYP2D6 and to a lesser extent via CYP3A4.72 Steadily decreasing plasma levels of paroxetine and increasing depressive symptoms can occur in pregnancy in women with the CYP2D6-extensive or ultrarapid metabolizer genotype. Cytochrome P2D6 intermediate and poor metabolizers may have increasing plasma levels of paroxetine throughout pregnancy.73 Antidepressant accumulation in poor and intermediate metabolizers could potentially have adverse effects on the fetus.74

Citalopram, a racemic mixture of R- and S-citalopram, and the more pharmacologically active S-enantiomer, escitalopram, are metabolized via demethylation at CYP2C19, CYP2D6, and CYP3A4.75,76 Main metabolites include desmethylcitalopram and didesmethylcitalopram. Doses of citalopram result in low trough plasma concentrations and metabolites during pregnancy but a significantly higher mean desmethylcitalopram-citalopram metabolic ratio compared with 2 months postpartum, suggesting induced metabolism during pregnancy.53 Decreased level/dose ratios are associated with lowered drug efficacy and increased dose requirements in the second half of gestation.55 Enhanced demethylation of citalopram by CYP2D6 may in part contribute to lower trough levels in pregnancy as compared with the postpartum period.53,77

Fluvoxamine is an achiral drug without clinically significant active metabolites. It undergoes metabolism via CYP2D6 and CYP1A2, and its main metabolite is fluvoxamine acid.78 Pharmacokinetic studies during pregnancy were not found for fluvoxamine, but CYP2D6 activity has been shown to be increased during the third trimester and associated with decreased plasma drug concentrations and possibly diminished efficacy. This effect may be offset by the decreased CYP1A2 activity seen throughout pregnancy.32

Vilazodone is metabolized through CYP, mainly CYP3A4 with lesser contributions from CYP2C19 and CYP2D6, and non-CYP pathways.78 Pharmacokinetic studies during pregnancy were not found for vilazodone, but the 2 main CYP isoenzymes responsible for its metabolism demonstrate different overall changes in activity during pregnancy (ie, CYPC19 is reduced, and CYP2D6 is increased). The net effect of these changes on vilazodone during pregnancy is unknown.

Evidence for Change in Perinatal Phase 1 Metabolism: SNRIs

Venlafaxine is renally excreted and predominantly metabolized by CYP2D6 into O-desmethylvenlafaxine (ie, desvenlafaxine) and CYP3A4 to N-desmethylvenlafaxine. Although dose adjustments for sex are not generally indicated, women have higher dose-corrected concentrations than men.79,80 An analysis that compared venlafaxine and its metabolic products in the first and third trimester to the postpartum period did not find concentration differences.77 In contrast, 1 case report of a pregnant adolescent patient with bipolar II disorder treated with venlafaxine extended release and concomitant psychotropics reported 2-fold higher venlafaxine plasma levels in the postpartum than during pregnancy for the same dose.57

Unlike venlafaxine, desvenlafaxine is metabolized by CYP3A4 and is biotransformed to N,O-didesmethylvenlafaxine and a hydroxylated metabolite.81 No pharmacokinetic studies during pregnancy were located for desvenlafaxine, but based on studies of CYP3A4 with other antidepressants, its metabolism has the potential to be reduced during pregnancy.

Duloxetine undergoes oxidative metabolism to 4-, 5-, and 6-hydroxy duloxetine by CYP2D6 and CYP1A2 and subsequent sulfate and glucuronide conjugation.82 Nonpregnant women have a 64% higher average steady-state concentration than men, in part because of lower CYP1A2 activity in women.83 No pharmacokinetic studies during pregnancy were located for duloxetine; however, 1 study reported duloxetine plasma levels in healthy lactating postpartum women comparable with those observed in healthy adults.84

Evidence for Change in Perinatal Phase 1 Metabolism: Other Antidepressants

Bupropion is extensively metabolized in the liver primarily by CYP2B6 to its main active metabolite, hydroxybupropion, and to threo- and erythrohydrobupropion via carbonyl reduction. During pregnancy, the placenta is a site of bupropion biotrans-formation, although carbonyl reduction is favored over oxidative biotransformation at this tissue site.38,85,86 To a lesser extent, the CYP1A2, 2A6, 2C9, 2D6, 2E1, and 3A4 isoforms are also involved in bupropion metabolism.8789 Sex hormones inhibit up to 50% of bupropion hydroxylation via CYP2B6,28 but the isoform shows high interindividual variability.90 No systematic studies have been published that inform the pharmacokinetics or TDM for bupropion in pregnancy.

Mirtazapine is a tetracyclic antidepressant that is rapidly and completely absorbed. Major pathways of biotransformation include demethylation and oxidation followed by conjugation. Mirtazapine undergoes N-demethylation via CYP3A4 into the pharmacologically active desmethylmirtazapine and hydroxylation by CYP2D6 and CYP1A2. Because several metabolic pathways contribute to mirtazapine’s biotransformation, genetic polymorphisms affecting activity in any one of the pathways are not hypothesized to result in clinically significant changes in mirtazapine plasma concentrations9193; however, CYP2D6 genotype may influence the concentrations of the enantiomers of mirtazapine and its metabolites.94 Nonpregnant women exhibit longer elimination half-lives and a 15% lower drug disposition as compared with men.95 Women show higher plasma concentrations compared with men,94 but it is unknown if this results in differences in clinical efficacy. No pharmacokinetic studies during pregnancy were located for mirtazapine.

Trazodone is highly lipophilic, well absorbed, and extensively metabolized by the liver via hydroxylation, N-oxidation and N-dealkylation. Cytochrome P3A4 mediates the N-dealkylation that results in the formation of the active metabolite 1-m-chlorophenylpiperazine, which then undergoes hydroxylation by CYP2D6.96 Therapeutic response has been associated with plasma trazodone concentrations of approximately 700 ng/mL but not with 1-m-chlorophenylpiperazine97 plasma levels. Cytochrome P1A2 may also be involved in trazodone metabolism. Higher drug concentrations have been found in females98 as compared with men. Pharmacokinetic data are limited to 1 case report of a pregnant patient treated with trazodone extended release of 150 mg/d in conjunction with other psychotropics. Plasma trazodone drug levels and the elimination half-life remained stable throughout pregnancy.57

Evidence for Change in Perinatal Metabolism: TCA

Tricyclic antidepressants metabolism is complex, and the pharmacokinetic changes observed in pregnancy likely reflect a summation of multiple metabolic effects including phase 1 and 2 metabolism. Generally, tertiary TCAs are demethylated by varying CYP450 enzymes into secondary TCAs and then undergo hydroxylation by CYP2D6 and glucuronidation by UGT. No pharmacokinetic or TDM studies during pregnancy were located for amitriptyline, desipramine, doxepin, protriptyline, trimipramine, amoxapine, or maprotiline.

A landmark study by Wisner et al99 (1993) evaluated nortriptyline, clomipramine, and imipramine levels during pregnancy compared with nonpregnancy doses and levels. The final dose required during pregnancy to maintain therapeutic drug levels was an average of 1.3 to 2.0 times the dose required when not pregnant. Doses increased over the second half of pregnancy, especially in the third trimester. The third trimester was associated with rapid acceleration of the required dose increase,99 and patients responded at plasma levels similar to those before pregnancy. Subsequent case reports of decreased plasma levels of nortriptyline and imipramine during pregnancy associated with recurrence of depressive symptoms confirm these findings.100 Nortriptyline level/dose ratios rise during the 2 to 6 postpartum weeks and then decline and stabilize around week 11 and beyond (Table 1).54

TABLE 1.

Change in Perinatal Antidepressant Phase 1 Metabolism and Indications for Perinatal Dose Adjustments

Antidepressant Main Isoenzymes Involved
in Metabolism101
Evidence of Change in Metabolism in Pregnancy
Dose increase may be indicated after 20 weeks gestation
Citalopram CYPC19, CYP2D6, CYP3A4 Increased drug metabolism between
20 weeks and delivery53,55
Clomipramine CYP1A2, CYP2C19, CYP2D6, CYP3A4 Decreased plasma levels99,100
Imipramine CYP1A2, CYP2C19, CYP2D6, CYP3A4 Decreased plasma levels99
Dose increase may be indicated in third trimester
Fluoxetine CYP2C9, CYP2C19, CYP2D6, CYP3A4 Higher maternal serum concentrations in
the postpartum period compared with
the third trimester68,69; mean ratios of
fluoxetine to metabolite levels
decreased during pregnancy.56
Fluvoxamine CYP1A2, CYP2D6 Increased CYP2D6 activity in third
trimester32
Nortriptyline CYP2D6 Decreased plasma levels99,100
Paroxetine CYP2D6, CYP3A4 Plasma levels may increase or decrease,
varies with metabolizer genotype.73,74
Sertraline CYP2B6, CYP2C9, CYP2C19, CYP2D6,CYP3A4 Marked heterogeneity in range of
pharmacokinetic changes70,71
Dose adjustments generally not indicated
Venlafaxine CYP2D6, CYP2C19, CYP3A4 Metabolism consistent across pregnancy
and the postpartum77; report of 2-fold
increase plasma level in postpartum57
Insufficient data on need for dose adjustment
Amitriptyline CYP1A2, CYP2C19, CYP2D6, CYP3A4 [Insufficient data for change in metabolism in pregnancy.]
Bupropion CYP2B6, CYP2D6, CYP1A2, CYP2A6, CYP2C9, CYP2E1, CYP3A4
Desipramine CYP2D6
Desvenlafaxine CYP3A4
Doxepin CYP1A2, CYP2D6, CYP2C19, CYP3A4
Duloxetine CYP1A2, CYP2D6
Mirtazapine CYP1A2, CYP2D6, CYP3A4
Trazodone CYP3A4
Trimipramine CYP2C19, CYP2D6, CYP3A4
Vilazodone CYP3A4, CYP2C19, CYP2D6

Evidence for Change in Perinatal Metabolism of Mood Stabilizers: Lithium, LTG, CBZ, and VPA

Lithium

Although effects of pregnancy on the pharmacokinetics of antidepressant metabolism are variable, pregnancy more clearly affects the pharmacokinetics of lithium and some antiepileptic drugs (AEDs). Lithium is rapidly absorbed through the upper gastrointestinal tract and is almost exclusively renally eliminated without undergoing biotransformation. It is filtered through the glomeruli as a free ion, and 80% is reabsorbed by the proximal tubule.102,103 The pharmacodynamics of lithium is influenced by weight, renal function, age, coadministered medications, pregnancy, and lactation.102,104 If the excretion of lithium is impaired, plasma ion concentrations can increase dramatically and precipitate toxicity.103 Lithium has an established therapeutic drug level and a narrow therapeutic index of 0.6 to 1.2 mEq/L. Lithium clearance is 20% to 30% of the GFR and thus varies with GFR. During pregnancy, lithium increases by 30% to 50% because of increased renal blood flow and GFR, particularly in the last months of gestation,50,104,105 causing plasma levels to decrease substantially, increasing the risk of maternal relapse.106 At delivery, vascular volume rapidly decreases, and lithium clearance precipitously decreases to prepregnancy levels.102,107

Lamotrigine

Lamotrigine is the most widely prescribed AED for epilepsy in women of reproductive age108 and is often used for maintenance treatment of bipolar disorder. Lamotrigine almost entirely undergoes hepatic glucuronic acid conjugation to its inactive metabolite LTG 2-N-glucuronide via UGT1A4. The sex steroid changes that occur in pregnancy increase phase 2 glucuronidation, which leads to increased LTG clearance.109,110 2-N-glucuronide/LTG ratios are reported to be 175% higher in the third trimester than at baseline. Less significant pregnancy-induced changes can also affect LTG levels, such as the degree of plasma protein binding, absorption, or transplacental transfer.109,111

Although there is substantial variability in the pharmacokinetics of LTG among women,112114 plasma concentrations consistently decline in pregnancy.17,113117 Lamotrigine clearance increases substantially at pregnancy onset118 and continues to increase progressively through the third trimester.17,18,113 Lamotrigine clearance potentially increases greater than 330% between preconception and the third trimester. An average dose increase of 250% is required to sustain therapeutic drug levels across pregnancy in women with epilepsy.113 Beginning within days of delivery and continuing during the first weeks post-partum, LTG elimination rate drops rapidly and plasma concentrations increase dramatically.109,113,115,116,119

Carbamazepine

Carbamazepine is metabolized hepatically mainly via conversion to an active metabolite, 10,11-epoxide. The 10,11-epoxide is then metabolized to inactive compounds via glucuronidation, conjugation, and hydroxylation. Carbamazepine induces the P450 system, which can increase the metabolism of medications administered concurrently.120 Carbamazepine clearance seems to increase in pregnancy,121 with some studies reporting declining total concentrations of CBZ during the second and third trimester,122127 whereas others have not found a significant change in CBZ plasma clearance.128 A study of 22 pregnancies reported a 42% decline in total plasma level and a 22% decline in free concentration.129 The measurement of total plasma concentrations of CBZ may be misleading, given that studies suggest that although the total concentration of CBZ decreases significantly in pregnancy, free-CBZ levels may not change when compared with baseline123,129 until delivery.129 The decrease in protein binding is likely the cause of the decrease in total CBZ concentration because the free concentration is less affected.129

Valproic Acid

Of all the mood stabilizers, VPA has the greatest risk of teratogenicity including neural tube defect and neurocognitive impairment15,16 and should not be considered a first-line mood stabilizer in women of reproductive potential. However, VPA is often prescribed to women of childbearing age.130 If the decision is made that VPA is the appropriate medication for perinatal mood stabilization, despite the risks, a number of measures can be taken to minimize fetal risk.

Highly protein bound, VPA pharmacologic activity is due to the free or unbound drug that crosses the blood-brain barrier. Valproic acid metabolism is complex because it undergoes a variety of different metabolic processes including mitochondrial beta-oxidation, CYP450-dependent processes including CYP2C9, CYP2C19, and CYP2A6, and glucuronidation. The half-life of VPA can be altered dramatically when administered with other medications that affect these pathways.120

An increase in VPA clearance resulting in a decrease in serum VPA levels has been observed particularly at the end of the third trimester.128,131,133 The plasma concentration of VPA decreases by as much as 50% in the last few weeks of pregnancy; however, no significant changes have been found in unbound concentrations.131,134 Concentration-dependent, VPA plasma protein binding decreases during pregnancy, and the free fraction increases as serum concentrations rise.134 Although VPA is highly protein bound, several authors133136 have noted that while the total VPA levels decline, free levels do not and in fact remain unchanged or even increased. This suggests that both free and total plasma levels of VPA should be measured during pregnancy.134,135 Valproic acid concentration decreases sharply in the immediate postpartum period (Table 2).131

TABLE 2.

Change in Perinatal Metabolism of Mood Stabilizers and Recommendations for TDM or Dose Adjustments

Mood
Stabilizer
Main Mechanisms
of Metabolism
Evidence of Change in
Metabolism in Pregnancy
Recommendations for Clinical
Monitoring and TDM
Lithium GI absorption
Excreted via urine
Increased renal blood flow and GFR50,105
Increased renal lithium clearance137
Lower serum concentrations137,138
TDM up to monthly during pregnancy and up
to weekly or biweekly in the last month
of pregnancy106,138 for women who are
required for reinitiation of lithium and have
unstable mood symptoms or concurrent
medical conditions affecting lithium serum
concentrations
TDM each trimester for euthymic women on
stable doses of lithium
Maintain lowest effective level.139
Check maternal lithium level before delivery.
Check therapeutic drug levels immediately after
delivery and for any clinical worsening or
change in clinical status related to obstetrical,
medical, and psychiatric condition.138
Once medically stable, restart preconception dose.
LTG Hepatic glucuronic acid
conjugation via UGT1A4
Increased phase 2 glucuronidation109
LTG clearance increases and plasma
concentration decreases across
pregnancy.109,111,112,114116,118
Postpartum LTG elimination rate drops
rapidly.109,113,115,116
Mean dose increase of 250% required to sustain
therapeutic drug levels across pregnancy in
women with epilepsy113
Use option preconception drug level as a guide
for adjusting dosage.140
Use lowest effective dose15
Empirically taper to preconception dose in the
postpartum period111,113,116
CBZ CYP3A4, CYP2B6, CYP2C8,
CYP2E1, CYP2C9,
CYP1A2, UGT
Excreted via urine (72%) and
feces (28%)
Data are conflicting; CBZ clearance
may increase in pregnancy, whereas
total CBZ concentration decreases
significantly in pregnancy.122127
Free CBZ levels do not change when
compared with baseline123,129 until
delivery.129
Both free and plasma concentrations should
be monitored.122124,129
Lowest effective dose should be used.15
After delivery, the dose should be tapered
rapidly to avoid toxicity buildup and
maintain prepregnancy CBZ levels.
VPA Highly protein bound; complex
hepatic metabolism via
mitochondrial beta-oxidation,
CYP450 (eg, CYP2C9,
CYP2C19, and CYP2A6),
and glucuronidation
Plasma concentration decreases.128,131133
No significant changes in free or unbound
concentrations in pregnancy131,134
Sharp decrease in VPA concentration in
the immediate postpartum period131
Both free and total plasma levels of VPA
should be measured during pregnancy.134,135
Baseline VPA levels should be obtained before
conception to identify the optimal serum
concentration for mood stabilization.
Use optimal drug level to dose adjustments
during pregnancy.
Levels should be checked at least monthly to
maintain the preconception serum
concentration.141
After delivery, the dose should be tapered
rapidly to avoid toxicity buildup and to
maintain preconception VPA levels.

GI indicates gastrointestinal.

DISCUSSION AND CLINICAL RECOMMENDATIONS

Antidepressants

The relationships between drug dose, TDM, and therapeutic effects are complex. Interindividual differences result from pharmacokinetic and pharmacodynamic variability, as well as genetic variants influencing the translation of proteins involved in metabolizing enzymes, drug transporters, and drug targets. At the level of the individual, perinatal changes in pharmacokinetics may or may not lead to changes in drug or metabolite levels. In addition, changes in drug or metabolite levels do not necessarily lead to alternations in clinical status.

Although SSRI concentrations fluctuate in pregnancy, the relationship between SSRI blood levels and clinical response is not well established.142144 Despite the lack of clarity in the relationship between SSRI blood levels and clinical response, data suggest that many pregnant women on antidepressant monotherapy may require dose increases, especially after 20 weeks gestation,70 to treat depressive symptoms or maintain euthymia. Studies to date suggest broad interindividual variability in pharmacokinetic changes, with some but not all women experiencing faster SSRI metabolism in late pregnancy. To maintain euthymia for some women in late pregnancy, the SSRI dose may need to be increased almost 2-fold the dose required earlier in pregnancy.70 Based on population data, at this time, there is insufficient evidence to support routine therapeutic blood level monitoring of antidepressants other than TCAs during pregnancy or the postpartum. Therapeutic drug monitoring is especially helpful with the TCA class, not only to monitor efficacy but also toxicity. Clinical presentation, ideally in combination with the use of measurement-based care using standardized assessments145147 during the perinatal period, should primarily inform treatment decisions.

Although not studied with most non-TCA antidepressants, a potential patient-specific option is to use the individual woman’s antidepressant level at a time when she is clinically asymptomatic as a target serum level during pregnancy. Although these antidepressants do not have an established therapeutic range, the individual woman’s antidepressant level can be used as a guide for adjusting dosage during pregnancy and in the postpartum. However, studies are necessary to determine if this strategy would improve clinical care and whether it would be cost-effective. In 2004, the Food and Drug Administration suggested that physicians may consider tapering antidepressants in the third trimester to lower the risk of complications associated with late gestational exposure to SSRIs;148 however, research does not support that discontinuing antidepressants in the third trimester diminishes the risk for neonatal symptoms.149 Postpartum pharmacokinetic changes can result in the emergence of antidepressant adverse effects soon after delivery for women who are required for a dose increase during pregnancy. We recommend close clinical monitoring; if adverse effects emerge, the dosage can be lowered. We do not recommend immediately decreasing the antidepressant to the preconception dose in the absence of adverse effects.

Standards have been established for TDM for TCAs outside the perinatal period, and these serve as a basis for the measurement and interpretation of perinatal drug levels. During pregnancy, we recommend monthly monitoring of trough drug levels. Monitoring is especially important in the third trimester when dose increases may be necessary. In the postpartum period, we recommend careful monitoring for adverse effects and tapering to the preconception dose to mitigate adverse effects that may be associated with rising serum levels 2 to 6 weeks after delivery. We recommend checking a blood level whenever adverse effects emerge in the early postpartum or at least at week 6 when drug levels have been reported to peak before eventual stabilization around week 11.54 Dose decreases may be indicated around week 6 to ameliorate adverse effects.

Mood Stabilizers

Women with bipolar disorder are at a heightened risk of relapse in the postpartum,150 and therefore, clinical monitoring and optimization of treatment is imperative in late pregnancy and early postpartum. Pregnancy markedly affects the pharmacokinetics of both LTG and lithium, with plasma concentrations declining throughout pregnancy.17,113116 Therapeutic drug monitoring of AEDs is common in the treatment of epilepsy during pregnancy and may also be useful in the treatment of bipolar disorder during the perinatal period.

Therapeutic drug monitoring for lithium is well established, and in pregnancy, renal lithium clearance almost doubles, lowering serum concentrations and increasing the potential for relapse.137 Several authors106,138 recommend frequent TDM of lithium in pregnancy, up to monthly during pregnancy and up to weekly or biweekly in the last month of pregnancy. This frequency of monitoring may be indicated in women who have discontinued lithium and require reinitiation of the drug or in women with medical comorbidities affecting lithium absorption or clearance, such as hyperemesis gravidarum or dehydration. For euthymic women on stable doses of lithium, we recommend checking a lithium level every trimester. Although 0.8 to 1.0 mEq/L is widely cited as a target effective level,151 during pregnancy, women should be maintained at the lowest effective level.139 Therapeutic drug monitoring can also be used to prevent women from lithium toxicity that can occur during immediate changes in physiology from acute illnesses such as gastroenteritis or other causes of volume depletion including hyperemesis gravidarum.14 Although the evidence is equivocal, some authors recommend a sustained-release preparation during pregnancy152 because they are often thought to produce more stable lithium levels than immediate release preparations.104

A maternal lithium level should be checked when women present for delivery, and adequate hydration should be ensured; nephrotoxins and nonsteroidal antiinflammatory drugs should be avoided.153 Although some authors have recommended holding lithium at labor onset137,139 or 24 to 48 hours before a scheduled cesarean delivery or induction,104,106,107,139 this has not been found to mitigate adverse perinatal outcomes and infant complications. Others have recommended increasing hydration around delivery to avoid lithium toxicity.104 Therapeutic drug levels should be checked 24 hours after delivery and then after each dose adjustment.138

At delivery, the risk of some lithium-induced perinatal complications may also be related to neonatal serum levels, especially for levels greater than 0.64 mEq/L.139 Thus, using the lowest effective dose that is still within therapeutic range is recommended.14 If neonatal lithium toxicity is suspected, the neonate’s serum lithium level should be monitored. Lithium containing sampling devices should be avoided because they can yield inaccurate lithium levels.154,155 Maternal GFR returns to pregravid levels immediately after delivery, and lithium levels return to preconception levels.139 Vascular volume rapidly decreases by approximately 40% and renal lithium clearance decreases to prepregnancy levels, leading to increased serum concentrations and risk of maternal lithium toxicity;106,107,156 thus, preconception doses should be restarted immediately after delivery.14

The American Academy of Neurology states that evidence supports active monitoring of LTG levels during pregnancy.114 In view of the interpatient variability, some authors recommend the establishment of a baseline LTG concentration in any woman of childbearing age with epilepsy116,118 and at least monthly monitoring during pregnancy and weekly during puerperium.113,118 In contrast to the use of LTG in the treatment of epilepsy, LTG dosing in bipolar disorder is typically guided by clinical response. Blood levels are not routinely obtained, and target therapeutic blood levels are not defined because they are for other mood stabilizers such as lithium and VPA. However, an individual’s preconception level could potentially be used as a guide for prophylactically increasing dosage during pregnancy18,140,157 because LTG levels decline and potentially reduce risk of relapse and postpartum psychosis.140,158,160

If a preconception LTG level is not available for reference, increases in LTG dosing in late pregnancy should be driven by clinical response with a low threshold for increased dosage. Post-partum LTG toxicity is common and can occur in more than 25% of women, whereas LTG’s elimination rate drops rapidly and plasma concentrations increase dramatically immediately after delivery until 2 to 3 weeks postpartum.109,113,115,116 Empirically tapering to the preconception dose in the postpartum period can reduce toxicity.111,113,116

For women receiving CBZ, both free and plasma concentrations should be monitored122124,129 in combination with close clinical monitoring; both should guide dose adjustments.129 Similar to the other mood stabilizers, the lowest effective dose should be used in pregnancy. After delivery, the dose should be tapered rapidly to avoid toxicity and maintain prepregnancy CBZ levels.

For women who plan to conceive while using VPA for mood stabilization, baseline VPA levels should be obtained before conception to identify the optimal serum concentration for mood stabilization and guide the dose adjustments during pregnancy. Free and plasma levels should be checked at least monthly to maintain the preconception serum concentration.141 Valproic acid easily transfers across the placenta, and congenital malformations correlate with maternal VPA concentrations and daily dose.131,161,163 The lowest effective dosage of VPA, preferably less than 1000 mg/d, should be used in pregnancy.135 To decrease the incidence of neural tube defects, all women of childbearing age taking most anticonvulsants should be treated with up to 4 to 5 mg/d of folic acid. After delivery, the dose should be tapered rapidly to avoid toxicity buildup and to maintain preconception VPA levels.

Therapeutic drug monitoring can also be used to mitigate some of the risks associated with drug interactions, which lithium, LTG, VPA, and CBZ are all very sensitive to. Therapeutic drug monitoring is imperative to monitor for changes in lithium levels that can occur secondary to drug-to-drug interactions. Lamotrigine, VPA, and CBZ are AEDs that can induce or inhibit other drugs and thereby diminish or enhance the pharmacologic effect of other drugs. Carbamazepine induces enzyme activity and thus decreases drug concentrations and the effect of other drugs. In contrast, VPA inhibits enzyme activity and increases drug concentrations, which carries the risk of causing drug toxicity. Therapeutic drug monitoring can be used to monitor drug concentrations and optimize treatment.164

Further Recommendations for Clinical Care

The use of antidepressant or mood-stabilizing medications during pregnancy involves complex clinical decisions based on the risks and benefits of medications as well as clinical status, history, and treatment preferences of each woman.4,5,106,113,139,165 Data-driven clinical recommendations for frequency of clinical monitoring and TDM across pregnancy and the postpartum are urgently needed, whereas therapeutic ranges have typically been derived in nonpregnant populations and have not yet been validated in pregnancy.166 The available data support frequent clinical monitoring of symptoms throughout pregnancy and postpartum; therefore, this is strongly recommended.

Further research is critically needed to examine how TDM of antidepressants and mood stabilizers in pregnancy will inform the efficacy and safety of psychotropic use for women and their infants. The literature reflects great interindividual variability regarding the effects of pregnancy on metabolism of medications where such data are available. Research is required to identify biomarkers that would specifically inform which women are at risk for clinically significant pharmacokinetic changes in their medications across pregnancy and postpartum.

Acknowledgments

AUTHOR DISCLOSURE INFORMATION

Dr Deligiannidis has received research support from UMass Medical School (NIHUL1RR031982), NIH (NIMH 1K23MH097794), Worcester Foundation for Biomedical Research, and Forest Research Institute and royalties from an employee invention (National Institutes of Health/National Institute of Child Health and Human Development). Dr Byatt has also received grant/funding support from the Massachusetts Department of Mental Health for the Massachusetts Child Psychiatry Access Program for Moms, Meyers Primary Care Institute and the Rosalie Wolf Interdisciplinary Geriatric Healthcare Research Center Small Grants Initiative. Dr Byatt’s work was on this project supported by the National Center for Research Resources and the National Center for Advancing Translational Sciences of the National Institutes of Health through grant number KL2TR000160. Dr Freeman has received research support from GlaxoSmithKline, Lilly, and Forest, has served on advisory boards or consulted for Johnson & Johnson, Genentech, Takeda and Lundbeck, Otsuka, and Pamab, and has received a stipend for medical editing from DSM Nutritionals.

REFERENCES

  • 1.Chung TK, Lau TK, Yip AS, et al. Antepartum depressive symptomatology is associated with adverse obstetric and neonatal outcomes. Psychosom Med. 2001;63(5):830–834. doi: 10.1097/00006842-200109000-00017. [DOI] [PubMed] [Google Scholar]
  • 2.Yonkers KA, Wisner KL, Stewart DE, et al. The management of depression during pregnancy: a report from the American Psychiatric Association and the American College of Obstetricians and Gynecologists. Gen Hosp Psychiatry. 2009;31(5):403–413. doi: 10.1016/j.genhosppsych.2009.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Grote NK, Bridge JA, Gavin AR, et al. A meta-analysis of depression during pregnancy and the risk of preterm birth, low birth weight, and intrauterine growth restriction. Arch Gen Psychiatry. 2010;67(10):1012–1024. doi: 10.1001/archgenpsychiatry.2010.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wisner KL, Sit DK, Hanusa BH, et al. Major depression and antidepressant treatment: impact on pregnancy and neonatal outcomes. Am J Psychiatry. 2009;166(5):557–566. doi: 10.1176/appi.ajp.2008.08081170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yonkers KA, Wisner KL, Stowe Z, et al. Management of bipolar disorder during pregnancy and the postpartum period. Am J Psychiatry. 2004;161(4):608–620. doi: 10.1176/appi.ajp.161.4.608. [DOI] [PubMed] [Google Scholar]
  • 6.Cohen LS, Altshuler LL, Harlow BL, et al. Relapse of major depression during pregnancy in women who maintain or discontinue antidepressant treatment. JAMA. 2006;295(5):499–507. doi: 10.1001/jama.295.5.499. [DOI] [PubMed] [Google Scholar]
  • 7.Viguera AC, Whitfield T, Baldessarini RJ, et al. Risk of recurrence in women with bipolar disorder during pregnancy: prospective study of mood stabilizer discontinuation. Am J Psychiatry. 2007;164(12):1817–1824. doi: 10.1176/appi.ajp.2007.06101639. quiz 1923. [DOI] [PubMed] [Google Scholar]
  • 8.Byatt N, Deligiannidis KM, Freeman MP. Antidepressant use in pregnancy: a critical review focused on risks and controversies. Acta Psychiatr Scand. 2013;127(2):94–114. doi: 10.1111/acps.12042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.‘t Jong GW, Einarson T, Koren G, et al. Antidepressant use in pregnancy and persistent pulmonary hypertension of the newborn (PPHN): a systematic review. Reprod Toxicol. 2012;34(3):293–297. doi: 10.1016/j.reprotox.2012.04.015. [DOI] [PubMed] [Google Scholar]
  • 10.Yonkers KA, Norwitz ER, Smith MV, et al. Depression and serotonin reuptake inhibitor treatment as risk factors for preterm birth. Epidemiology. 2012;23(5):677–685. doi: 10.1097/EDE.0b013e31825838e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hayes RM, Wu P, Shelton RC, et al. Maternal antidepressant use and adverse outcomes: a cohort study of 228,876 pregnancies. Am J Obstet Gynecol. 2012;207(1):49. doi: 10.1016/j.ajog.2012.04.028. e1–49.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bodén R, Lundgren M, Brandt L, et al. Risks of adverse pregnancy and birth outcomes in women treated or not treated with mood stabilisers for bipolar disorder: population based cohort study. BMJ. 2012;345:e7085. doi: 10.1136/bmj.e7085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Galbally M, Roberts M, Buist A. Mood stabilizers in pregnancy: a systematic review. Aust N Z J Psychiatry. 2010;44(11):967–977. doi: 10.3109/00048674.2010.506637. [DOI] [PubMed] [Google Scholar]
  • 14.Gentile S. Lithium in pregnancy: the need to treat, the duty to ensure safety. Expert Opin Drug Saf. 2012;11(3):425–437. doi: 10.1517/14740338.2012.670419. [DOI] [PubMed] [Google Scholar]
  • 15.Tomson T, Battino D, Bonizzoni E, et al. Dose-dependent risk of malformations with antiepileptic drugs: an analysis of data from the EURAP epilepsy and pregnancy registry. Lancet Neurol. 2011;10(7):609–617. doi: 10.1016/S1474-4422(11)70107-7. [DOI] [PubMed] [Google Scholar]
  • 16.Werler MM, Ahrens KA, Bosco JL, et al. Use of antiepileptic medications in pregnancy in relation to risks of birth defects. Ann Epidemiol. 2011;21(11):842–850. doi: 10.1016/j.annepidem.2011.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Pennell PB, Newport DJ, Stowe ZN, et al. The impact of pregnancy and childbirth on the metabolism of lamotrigine. Neurology. 2004;62(2):292–295. doi: 10.1212/01.wnl.0000103286.47129.f8. [DOI] [PubMed] [Google Scholar]
  • 18.Pennell PB, Peng L, Newport DJ, et al. Lamotrigine in pregnancy: clearance, therapeutic drug monitoring, and seizure frequency. Neurology. 2008;70(22 Pt 2):2130–2136. doi: 10.1212/01.wnl.0000289511.20864.2a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yonkers KA, Kando JC, Cole JO, et al. Gender differences in pharmacokinetics and pharmacodynamics of psychotropic medication. Am J Psychiatry. 1992;149(5):587–595. doi: 10.1176/ajp.149.5.587. [DOI] [PubMed] [Google Scholar]
  • 20.Bies RR, Bigos KL, Pollock BG. Gender differences in the pharmacokinetics and pharmacodynamics of antidepressants. J Gend Specif Med. 2003;6(3):12–20. [PubMed] [Google Scholar]
  • 21.Kando JC, Yonkers KA, Cole JO. Gender as a risk factor for adverse events to medications. Drugs. 1995;50(1):1–6. doi: 10.2165/00003495-199550010-00001. [DOI] [PubMed] [Google Scholar]
  • 22.Hutson WR, Roehrkasse RL, Wald A. Influence of gender and menopause on gastric emptying and motility. Gastroenterology. 1989;96(1):11–17. doi: 10.1016/0016-5085(89)90758-0. [DOI] [PubMed] [Google Scholar]
  • 23.Sadik R, Abrahamsson H, Stotzer PO. Gender differences in gut transit shown with a newly developed radiological procedure. Scand J Gastroenterol. 2003;38(1):36–42. doi: 10.1080/00365520310000410. [DOI] [PubMed] [Google Scholar]
  • 24.Lorena SL, Tinois E, Hirata ES, et al. Scintigraphic study of gastric emptying and intragastric distribution of a solid meal: gender differences. Arq Gastroenterol. 2000;37(2):102–106. doi: 10.1590/s0004-28032000000200006. [DOI] [PubMed] [Google Scholar]
  • 25.Flockhart DA. [Accessed March 13, 2013];Drug Interactions: Cytochrome P450 Drug Interaction Table. 2007 2009 [cited 2011 April 17]; Available at: http://medicine.iupui.edu/clinpharm/ddis/table.asp. [Google Scholar]
  • 26.Balogh A, Klinger G, Henschel L, et al. Influence of ethinylestradiol-containing combination oral contraceptives with gestodene or levonorgestrel on caffeine elimination. Eur J Clin Pharmacol. 1995;48(2):161–166. doi: 10.1007/BF00192743. [DOI] [PubMed] [Google Scholar]
  • 27.Palovaara S, Tybring G, Laine K. The effect of ethinyloestradiol and levonorgestrel on the CYP2C19-mediated metabolism of omeprazole in healthy female subjects. Br J Clin Pharmacol. 2003;56(2):232–237. doi: 10.1046/j.1365-2125.2003.01868.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Palovaara S, Pelkonen O, Uusitalo J, et al. Inhibition of cytochrome P450 2B6 activity by hormone replacement therapy and oral contraceptive as measured by bupropion hydroxylation. Clin Pharmacol Ther. 2003;74(4):326–333. doi: 10.1016/S0009-9236(03)00202-9. [DOI] [PubMed] [Google Scholar]
  • 29.Palovaara S, Kivistö KT, Tapanainen P, et al. Effect of an oral contraceptive preparation containing ethinylestradiol and gestodene on CYP3A4 activity as measured by midazolam 1’-hydroxylation. Br J Clin Pharmacol. 2000;50(4):333–337. doi: 10.1046/j.1365-2125.2000.00271.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chang SY, Chen C, Yang Z, et al. Further assessment of 17alpha-ethinyl estradiol as an inhibitor of different human cytochrome P450 forms in vitro. Drug Metab Dispos. 2009;37(8):1667–1675. doi: 10.1124/dmd.109.026997. [DOI] [PubMed] [Google Scholar]
  • 31.Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens, reduces lamotrigine serum concentrations. Epilepsia. 2005;46(9):1414–1417. doi: 10.1111/j.1528-1167.2005.10105.x. [DOI] [PubMed] [Google Scholar]
  • 32.Anderson GD. Pregnancy-induced changes in pharmacokinetics: a mechanistic-based approach. Clin Pharmacokinet. 2005;44(10):989–1008. doi: 10.2165/00003088-200544100-00001. [DOI] [PubMed] [Google Scholar]
  • 33.DeVane CL, Stowe ZN, Donovan JL, et al. Therapeutic drug monitoring of psychoactive drugs during pregnancy in the genomic era: challenges and opportunities. J Psychopharmacol. 2006;20(suppl 4):54–59. doi: 10.1177/1359786806066054. [DOI] [PubMed] [Google Scholar]
  • 34.Syme MR, Paxton JW, Keelan JA. Drug transfer and metabolism by the human placenta. Clin Pharmacokinet. 2004;43(8):487–514. doi: 10.2165/00003088-200443080-00001. [DOI] [PubMed] [Google Scholar]
  • 35.Wang JS, Newport DJ, Stowe ZN, et al. The emerging importance of transporter proteins in the psychopharmacological treatment of the pregnant patient. Drug Metab Rev. 2007;39(4):723–746. doi: 10.1080/03602530701690390. [DOI] [PubMed] [Google Scholar]
  • 36.Hakkola J, Raunio H, Purkunen R, et al. Detection of cytochrome P450 gene expression in human placenta in first trimester of pregnancy. Biochem Pharmacol. 1996;52(2):379–383. doi: 10.1016/0006-2952(96)00216-x. [DOI] [PubMed] [Google Scholar]
  • 37.Ter Horst PG, Jansman FG, van Lingen RA, et al. Pharmacological aspects of neonatal antidepressant withdrawal. Obstet Gynecol Surv. 2008;63(4):267–279. doi: 10.1097/OGX.0b013e3181676be8. [DOI] [PubMed] [Google Scholar]
  • 38.Earhart AD, Patrikeeva S, Wang X, et al. Transplacental transfer and metabolism of bupropion. J Matern Fetal Neonatal Med. 2010;23(5):409–416. doi: 10.1080/14767050903168424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Heikkine T, Ekblad U, Laine K. Transplacental transfer of citalopram, fluoxetine and their primary demethylated metabolites in isolated perfused human placenta. BJOG. 2002;109(9):1003–1008. doi: 10.1111/j.1471-0528.2002.01467.x. [DOI] [PubMed] [Google Scholar]
  • 40.Hendrick V, Stowe ZN, Altshuler LL, et al. Placental passage of antidepressant medications. Am J Psychiatry. 2003;160(5):993–996. doi: 10.1176/appi.ajp.160.5.993. [DOI] [PubMed] [Google Scholar]
  • 41.Rampono J, Simmer K, Ilett KF, et al. Placental transfer of SSRI and SNRI antidepressants and effects on the neonate. Pharmacopsychiatry. 2009;42(3):95–100. doi: 10.1055/s-0028-1103296. [DOI] [PubMed] [Google Scholar]
  • 42.Anderson GD. Sex and racial differences in pharmacological response: where is the evidence? Pharmacogenetics, pharmacokinetics, and pharmacodynamics. J Womens Health (Larchmt) 2005;14(1):19–29. doi: 10.1089/jwh.2005.14.19. [DOI] [PubMed] [Google Scholar]
  • 43.Pollock BG, Wylie M, Stack JA, et al. Inhibition of caffeine metabolism by estrogen replacement therapy in postmenopausal women. J Clin Pharmacol. 1999;39(9):936–940. doi: 10.1177/00912709922008560. [DOI] [PubMed] [Google Scholar]
  • 44.Lane JD, Steege JF, Rupp SL, et al. Menstrual cycle effects on caffeine elimination in the human female. Eur J Clin Pharmacol. 1992;43(5):543–546. doi: 10.1007/BF02285099. [DOI] [PubMed] [Google Scholar]
  • 45.Tracy TS, Venkataramanan R, Glover DD, et al. Temporal changes in drug metabolism (CYP1A2, CYP2D6 and CYP3A activity) during pregnancy. Am J Obstet Gynecol. 2005;192(2):633–639. doi: 10.1016/j.ajog.2004.08.030. [DOI] [PubMed] [Google Scholar]
  • 46.McGready R, Stepniewska K, Seaton E, et al. Pregnancy and use of oral contraceptives reduces the biotransformation of proguanil to cycloguanil. Eur J Clin Pharmacol. 2003;59(7):553–557. doi: 10.1007/s00228-003-0651-x. [DOI] [PubMed] [Google Scholar]
  • 47.Rudorfer MV, Potter WZ. Metabolism of tricyclic antidepressants. Cell Mol Neurobiol. 1999;19(3):373–409. doi: 10.1023/a:1006949816036. [DOI] [PubMed] [Google Scholar]
  • 48.Wadelius M, Darj E, Frenne G, et al. Induction of CYP2D6 in pregnancy. Clin Pharmacol Ther. 1997;62(4):400–407. doi: 10.1016/S0009-9236(97)90118-1. [DOI] [PubMed] [Google Scholar]
  • 49.Kirchheiner J, Nickchen K, Bauer M, et al. Pharmacogenetics of antidepressants and antipsychotics: the contribution of allelic variations to the phenotype of drug response. Mol Psychiatry. 2004;9(5):442–473. doi: 10.1038/sj.mp.4001494. [DOI] [PubMed] [Google Scholar]
  • 50.Dunlop W. Serial changes in renal haemodynamics during normal human pregnancy. Br J Obstet Gynaecol. 1981;88(1):1–9. doi: 10.1111/j.1471-0528.1981.tb00929.x. [DOI] [PubMed] [Google Scholar]
  • 51.Robson SC, Mutch E, Boys RJ, et al. Apparent liver blood flow during pregnancy: a serial study using indocyanine green clearance. Br J Obstet Gynaecol. 1990;97(8):720–724. doi: 10.1111/j.1471-0528.1990.tb16246.x. [DOI] [PubMed] [Google Scholar]
  • 52.Nakai A, Sekiya I, Oya A, et al. Assessment of the hepatic arterial and portal venous blood flows during pregnancy with Doppler ultrasonography. Arch Gynecol Obstet. 2002;266(1):25–29. doi: 10.1007/pl00007495. [DOI] [PubMed] [Google Scholar]
  • 53.Heikkinen T, Ekblad U, Kero P, et al. Citalopram in pregnancy and lactation. Clin Pharmacol Ther. 2002;72(2):184–191. doi: 10.1067/mcp.2002.126181. [DOI] [PubMed] [Google Scholar]
  • 54.Wisner KL, Perel JM, Peindl KS, et al. Effects of the postpartum period on nortriptyline pharmacokinetics. Psychopharmacol Bull. 1997;33(2):243–248. [PubMed] [Google Scholar]
  • 55.Sit DK, Perel JM, Helsel JC, et al. Changes in antidepressant metabolism and dosing across pregnancy and early postpartum. J Clin Psychiatry. 2008;69(4):652–658. doi: 10.4088/jcp.v69n0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sit D, Perel JM, Luther JF, et al. Disposition of chiral and racemic fluoxetine and norfluoxetine across childbearing. J Clin Psychopharmacol. 2010;30(4):381–386. doi: 10.1097/JCP.0b013e3181e7be23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Klier CM, Mossaheb N, Saria A, et al. Pharmacokinetics and elimination of quetiapine, venlafaxine, and trazodone during pregnancy and postpartum. J Clin Psychopharmacol. 2007;27(6):720–722. doi: 10.1097/JCP.0b013e31815a57d8. [DOI] [PubMed] [Google Scholar]
  • 58.Grote NK, Frank E. Difficult-to-treat depression: the role of contexts and comorbidities. Biol Psychiatry. 2003;53(8):660–670. doi: 10.1016/s0006-3223(03)00006-4. [DOI] [PubMed] [Google Scholar]
  • 59.Jiang H, Xie T, Ramsden DB, et al. Human catechol-O-methyltransferase down-regulation by estradiol. Neuropharmacology. 2003;45(7):1011–1018. doi: 10.1016/s0028-3908(03)00286-7. [DOI] [PubMed] [Google Scholar]
  • 60.Xie T, Ho SL, Ramsden D. Characterization and implications of estrogenic down-regulation of human catechol-O-methyltransferase gene transcription. Mol Pharmacol. 1999;56(1):31–38. doi: 10.1124/mol.56.1.31. [DOI] [PubMed] [Google Scholar]
  • 61.Worda C, Sator MO, Schneeberger C, et al. Influence of the catechol-O-methyltransferase (COMT) codon 158 polymorphism on estrogen levels in women. Hum Reprod. 2003;18(2):262–266. doi: 10.1093/humrep/deg059. [DOI] [PubMed] [Google Scholar]
  • 62.Sacher J, Wilson AA, Houle S, et al. Elevated brain monoamine oxidase A binding in the early postpartum period. Arch Gen Psychiatry. 2010;67(5):468–474. doi: 10.1001/archgenpsychiatry.2010.32. [DOI] [PubMed] [Google Scholar]
  • 63.von Moltke LL, Greenblatt DJ, Duan SX, et al. Human cytochromes mediating N-demethylation of fluoxetine in vitro. Psychopharmacology (Berl) 1997;132(4):402–407. doi: 10.1007/s002130050362. [DOI] [PubMed] [Google Scholar]
  • 64.Liu ZQ, Shu Y, Huang SL, et al. Effects of CYP2C19 genotype and CYP2C9 on fluoxetine N-demethylation in human liver microsomes. Acta Pharmacol Sin. 2001;22(1):85–90. [PubMed] [Google Scholar]
  • 65.Eap CB, Bondolfi G, Zullino D, et al. Concentrations of the enantiomers of fluoxetine and norfluoxetine after multiple doses of fluoxetine in cytochrome P4502D6 poor and extensive metabolizers. J Clin Psychopharmacol. 2001;21(3):330–334. doi: 10.1097/00004714-200106000-00013. [DOI] [PubMed] [Google Scholar]
  • 66.Margolis JM, O’Donnell JP, Mankowski DC, et al. (R)-, (S)-, and racemic fluoxetine N-demethylation by human cytochrome P450 enzymes. Drug Metab Dispos. 2000;28(10):1187–1191. [PubMed] [Google Scholar]
  • 67.Scordo MG, Spina E, Dahl ML, et al. Influence of CYP2C9, 2C19 and 2D6 genetic polymorphisms on the steady-state plasma concentrations of the enantiomers of fluoxetine and norfluoxetine. Basic Clin Pharmacol Toxicol. 2005;97(5):296–301. doi: 10.1111/j.1742-7843.2005.pto_194.x. [DOI] [PubMed] [Google Scholar]
  • 68.Kim J, Riggs KW, Misri S, et al. Stereoselective disposition of fluoxetine and norfluoxetine during pregnancy and breast-feeding. Br J Clin Pharmacol. 2006;61(2):155–163. doi: 10.1111/j.1365-2125.2005.02538.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Heikkinen T, Ekblad U, Palo P, et al. Pharmacokinetics of fluoxetine and norfluoxetine in pregnancy and lactation. Clin Pharmacol Ther. 2003;73(4):330–337. doi: 10.1016/s0009-9236(02)17634-x. [DOI] [PubMed] [Google Scholar]
  • 70.Hostetter A, Stowe ZN, Strader JR, Jr., et al. Dose of selective serotonin uptake inhibitors across pregnancy: clinical implications. Depress Anxiety. 2000;11(2):51–57. [PubMed] [Google Scholar]
  • 71.Freeman MP, Nolan PE, Jr., Davis MF, et al. Pharmacokinetics of sertraline across pregnancy and postpartum. J Clin Psychopharmacol. 2008;28(6):646–653. doi: 10.1097/JCP.0b013e31818d2048. [DOI] [PubMed] [Google Scholar]
  • 72.Jornil J, Jensen KG, Larsen F, et al. Identification of cytochrome P450 isoforms involved in the metabolism of paroxetine and estimation of their importance for human paroxetine metabolism using a population-based simulator. Drug Metab Dispos. 2010;38(3):376–385. doi: 10.1124/dmd.109.030551. [DOI] [PubMed] [Google Scholar]
  • 73.Ververs FF, Voorbij HA, Zwarts P, et al. Effect of cytochrome P450 2D6 genotype on maternal paroxetine plasma concentrations during pregnancy. Clin Pharmacokinet. 2009;48(10):677–683. doi: 10.2165/11318050-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 74.Laine K, Kytölä J, Bertilsson L. Severe adverse effects in a newborn with two defective CYP2D6 alleles after exposure to paroxetine during late pregnancy. Ther Drug Monit. 2004;26(6):685–687. doi: 10.1097/00007691-200412000-00016. [DOI] [PubMed] [Google Scholar]
  • 75.Greenblatt DJ, von Moltke LL, Harmatz JS, et al. Human cytochromes and some newer antidepressants: kinetics, metabolism, and drug interactions. J Clin Psychopharmacol. 1999;19(5 suppl 1):23S–35S. doi: 10.1097/00004714-199910001-00003. [DOI] [PubMed] [Google Scholar]
  • 76.van Harten J. Clinical pharmacokinetics of selective serotonin reuptake inhibitors. Clin Pharmacokinet. 1993;24(3):203–220. doi: 10.2165/00003088-199324030-00003. [DOI] [PubMed] [Google Scholar]
  • 77.O’Brien L, Baumer C, Thieme D, et al. Changes in antidepressant metabolism in pregnancy evidenced by metabolic ratios in hair: a novel approach. Forensic Sci Int. 2010;196(1–3):93–96. doi: 10.1016/j.forsciint.2009.12.034. [DOI] [PubMed] [Google Scholar]
  • 78.Mandrioli R, Mercolini L, Saracino MA, et al. Selective serotonin reuptake inhibitors (SSRIs): therapeutic drug monitoring and pharmacological interactions. Curr Med Chem. 2012;19(12):1846–1863. doi: 10.2174/092986712800099749. [DOI] [PubMed] [Google Scholar]
  • 79.Reis M, Lundmark J, Björk H, et al. Therapeutic drug monitoring of racemic venlafaxine and its main metabolites in an everyday clinical setting. Ther Drug Monit. 2002;24(4):545–553. doi: 10.1097/00007691-200208000-00014. [DOI] [PubMed] [Google Scholar]
  • 80.Klamerus KJ, Parker VD, Rudolph RL, et al. Effects of age and gender on venlafaxine and O-desmethylvenlafaxine pharmacokinetics. Pharmacotherapy. 1996;16(5):915–923. [PubMed] [Google Scholar]
  • 81.Preskorn S, Patroneva A, Silman H, et al. Comparison of the pharmacokinetics of venlafaxine extended release and desvenlafaxine in extensive and poor cytochrome P450 2D6 metabolizers. J Clin Psychopharmacol. 2009;29(1):39–43. doi: 10.1097/JCP.0b013e318192e4c1. [DOI] [PubMed] [Google Scholar]
  • 82.Lantz RJ, Gillespie TA, Rash TJ, et al. Metabolism, excretion, and pharmacokinetics of duloxetine in healthy human subjects. Drug Metab Dispos. 2003;31(9):1142–1150. doi: 10.1124/dmd.31.9.1142. [DOI] [PubMed] [Google Scholar]
  • 83.Lobo ED, Quinlan T, O’Brien L, et al. Population pharmacokinetics of orally administered duloxetine in patients: implications for dosing recommendation. Clin Pharmacokinet. 2009;48(3):189–197. doi: 10.2165/00003088-200948030-00005. [DOI] [PubMed] [Google Scholar]
  • 84.Lobo ED, Loghin C, Knadler MP, et al. Pharmacokinetics of duloxetine in breast milk and plasma of healthy postpartum women. Clin Pharmacokinet. 2008;47(2):103–109. doi: 10.2165/00003088-200847020-00003. [DOI] [PubMed] [Google Scholar]
  • 85.Wang X, Abdelrahman DR, Zharikova OL, et al. Bupropion metabolism by human placenta. Biochem Pharmacol. 2010;79(11):1684–1690. doi: 10.1016/j.bcp.2010.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hemauer SJ, Patrikeeva SL, Wang X, et al. Role of transporter-mediated efflux in the placental biodisposition of bupropion and its metabolite, OH-bupropion. Biochem Pharmacol. 2010;80(7):1080–1086. doi: 10.1016/j.bcp.2010.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Faucette SR, Hawke RL, Shord SS, et al. Evaluation of the contribution of cytochrome P450 3A4 to human liver microsomal bupropion hydroxylation. Drug Metab Dispos. 2001;29(8):1123–1129. [PubMed] [Google Scholar]
  • 88.Hesse LM, Venkatakrishnan K, Court MH, et al. CYP2B6 mediates the in vitro hydroxylation of bupropion: potential drug interactions with other antidepressants. Drug Metab Dispos. 2000;28(10):1176–1183. [PubMed] [Google Scholar]
  • 89.Kirchheiner J, Klein C, Meineke I, et al. Bupropion and 4-OH-bupropion pharmacokinetics in relation to genetic polymorphisms in CYP2B6. Pharmacogenetics. 2003;13(10):619–626. doi: 10.1097/00008571-200310000-00005. [DOI] [PubMed] [Google Scholar]
  • 90.Hesse LM, He P, Krishnaswamy S, et al. Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by cytochrome P450 2B6 in human liver microsomes. Pharmacogenetics. 2004;14(4):225–238. doi: 10.1097/00008571-200404000-00002. [DOI] [PubMed] [Google Scholar]
  • 91.Störmer E, von Moltke LL, Shader RI, et al. Metabolism of the antidepressant mirtazapine in vitro: contribution of cytochromes P-450 1A2, 2D6, and 3A4. Drug Metab Dispos. 2000;28(10):1168–1175. [PubMed] [Google Scholar]
  • 92.Delbressine LP, Moonen ME, Kaspersen FM, et al. Pharmacokinetics and biotransformation of mirtazapine in human volunteers. Clin Drug Investig. 1998;15(1):45–55. doi: 10.2165/00044011-199815010-00006. [DOI] [PubMed] [Google Scholar]
  • 93.Timmer CJ, Sitsen JM, Delbressine LP. Clinical pharmacokinetics of mirtazapine. Clin Pharmacokinet. 2000;38(6):461–474. doi: 10.2165/00003088-200038060-00001. [DOI] [PubMed] [Google Scholar]
  • 94.Jaquenoud Sirot E, Harenberg S, Vandel P, et al. Multicenter study on the clinical effectiveness, pharmacokinetics, and pharmacogenetics of mirtazapine in depression. J Clin Psychopharmacol. 2012;32(5):622–629. doi: 10.1097/JCP.0b013e3182664d98. [DOI] [PubMed] [Google Scholar]
  • 95.Borobia AM, Novalbos J, Guerra-López P, et al. Influence of sex and CYP2D6 genotype on mirtazapine disposition, evaluated in Spanish healthy volunteers. Pharmacol Res. 2009;59(6):393–398. doi: 10.1016/j.phrs.2009.02.006. [DOI] [PubMed] [Google Scholar]
  • 96.Rotzinger S, Bourin M, Akimoto Y, et al. Metabolism of some “second”-and “fourth”-generation antidepressants: iprindole, viloxazine, bupropion, mianserin, maprotiline, trazodone, nefazodone, and venlafaxine. Cell Mol Neurobiol. 1999;19(4):427–442. doi: 10.1023/a:1006953923305. [DOI] [PubMed] [Google Scholar]
  • 97.Mihara K, Yasui-Furukori N, Kondo T, et al. Relationship between plasma concentrations of trazodone and its active metabolite, m-chlorophenylpiperazine, and its clinical effect in depressed patients. Ther Drug Monit. 2002;24(4):563–566. doi: 10.1097/00007691-200208000-00016. [DOI] [PubMed] [Google Scholar]
  • 98.Prapotnik M, Waschgler R, König P, et al. Therapeutic drug monitoring of trazodone: are there pharmacokinetic interactions involving citalopram and fluoxetine? Int J Clin Pharmacol Ther. 2004;42(2):120–124. doi: 10.5414/cpp42120. [DOI] [PubMed] [Google Scholar]
  • 99.Wisner KL, Perel JM, Wheeler SB. Tricyclic dose requirements across pregnancy. Am J Psychiatry. 1993;150(10):1541–1542. doi: 10.1176/ajp.150.10.1541. [DOI] [PubMed] [Google Scholar]
  • 100.Altshuler LL, Hendrick VC. Pregnancy and psychotropic medication: changes in blood levels. J Clin Psychopharmacol. 1996;16(1):78–80. doi: 10.1097/00004714-199602000-00015. [DOI] [PubMed] [Google Scholar]
  • 101.Wynn GH, Sandson N, Muniz J. Clinical Manual of Drug Interaction: Principles for Medical Practice. Washington, DC: American Psychiatric Publishing, Inc.; 2009. [Google Scholar]
  • 102.Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach. Part II: Clinical pharmacology and therapeutic monitoring. CNS Drugs. 2009;23(4):331–349. doi: 10.2165/00023210-200923040-00005. [DOI] [PubMed] [Google Scholar]
  • 103.Malhi GS, Tanious M. Optimal frequency of lithium administration in the treatment of bipolar disorder: clinical and dosing considerations. CNS Drugs. 2011;25(4):289–298. doi: 10.2165/11586970-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 104.Malhi GS, Tanious M, Das P, et al. The science and practice of lithium therapy. Aust N Z J Psychiatry. 2012;46(3):192–211. doi: 10.1177/0004867412437346. [DOI] [PubMed] [Google Scholar]
  • 105.Thomsen K, Schou M. Avoidance of lithium intoxication: advice based on knowledge about the renal lithium clearance under various circumstances. Pharmacopsychiatry. 1999;32(3):83–86. doi: 10.1055/s-2007-979199. [DOI] [PubMed] [Google Scholar]
  • 106.Llewellyn A, Stowe ZN, Strader JR., Jr. The use of lithium and management of women with bipolar disorder during pregnancy and lactation. J Clin Psychiatry. 1998;59(suppl 6):57–64. discussion 65. [PubMed] [Google Scholar]
  • 107.Grandjean EM, Aubry JM. Lithium: updated human knowledge using an evidence-based approach: part III: clinical safety. CNS Drugs. 2009;23(5):397–418. doi: 10.2165/00023210-200923050-00004. [DOI] [PubMed] [Google Scholar]
  • 108.Sabers A, Dam M, A-Rogvi-Hansen B, et al. Epilepsy and pregnancy: lamotrigine as main drug used. Acta Neurol Scand. 2004;109(1):9–13. doi: 10.1034/j.1600-0404.2003.00200.x. [DOI] [PubMed] [Google Scholar]
  • 109.Ohman I, Beck O, Vitols S, et al. Plasma concentrations of lamotrigine and its 2-N-glucuronide metabolite during pregnancy in women with epilepsy. Epilepsia. 2008;49(6):1075–1080. doi: 10.1111/j.1528-1167.2007.01471.x. [DOI] [PubMed] [Google Scholar]
  • 110.Ohman I, Luef G, Tomson T. Effects of pregnancy and contraception on lamotrigine disposition: new insights through analysis of lamotrigine metabolites. Seizure. 2008;17(2):199–202. doi: 10.1016/j.seizure.2007.11.017. [DOI] [PubMed] [Google Scholar]
  • 111.Tran TA, Leppik IE, Blesi K, et al. Lamotrigine clearance during pregnancy. Neurology. 2002;59(2):251–255. doi: 10.1212/wnl.59.2.251. [DOI] [PubMed] [Google Scholar]
  • 112.Petrenaite V, Sabers A, Hansen-Schwartz J. Individual changes in lamotrigine plasma concentrations during pregnancy. Epilepsy Res. 2005;65(3):185–188. doi: 10.1016/j.eplepsyres.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • 113.Fotopoulou C, Kretz R, Bauer S, et al. Prospectively assessed changes in lamotrigine-concentration in women with epilepsy during pregnancy, lactation and the neonatal period. Epilepsy Res. 2009;85(1):60–64. doi: 10.1016/j.eplepsyres.2009.02.011. [DOI] [PubMed] [Google Scholar]
  • 114.Harden CL, Pennell PB, Koppel BS, et al. Practice parameter update: management issues for women with epilepsyVfocus on pregnancy (an evidence-based review): vitamin K, folic acid, blood levels, and breastfeeding: report of the Quality Standards Subcommittee and Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and American Epilepsy Society. Neurology. 2009;73(2):142–149. doi: 10.1212/WNL.0b013e3181a6b325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Pennell PB, Hovinga CA. Antiepileptic drug therapy in pregnancy I: gestation-induced effects on AED pharmacokinetics. Int Rev Neurobiol. 2008;83:227–240. doi: 10.1016/S0074-7742(08)00013-5. [DOI] [PubMed] [Google Scholar]
  • 116.Tomson T, Battino D. Pharmacokinetics and therapeutic drug monitoring of newer antiepileptic drugs during pregnancy and the puerperium. Clin Pharmacokinet. 2007;46(3):209–219. doi: 10.2165/00003088-200746030-00002. [DOI] [PubMed] [Google Scholar]
  • 117.de Haan GJ, Edelbroek P, Segers J, et al. Gestation-induced changes in lamotrigine pharmacokinetics: a monotherapy study. Neurology. 2004;63(3):571–573. doi: 10.1212/01.wnl.0000133213.10244.fd. [DOI] [PubMed] [Google Scholar]
  • 118.Franco V, Mazzucchelli I, Gatti G, et al. Changes in lamotrigine pharmacokinetics during pregnancy and the puerperium. Ther Drug Monit. 2008;30(4):544–547. doi: 10.1097/FTD.0b013e318178e2a9. [DOI] [PubMed] [Google Scholar]
  • 119.Ohman I, Vitols S, Tomson T. Lamotrigine in pregnancy: pharmacokinetics during delivery, in the neonate, and during lactation. Epilepsia. 2000;41(6):709–713. doi: 10.1111/j.1528-1157.2000.tb00232.x. [DOI] [PubMed] [Google Scholar]
  • 120.Keck PE, Jr., McElroy SL. Clinical pharmacodynamics and pharmacokinetics of antimanic and mood-stabilizing medications. J Clin Psychiatry. 2002;63(suppl 4):3–11. [PubMed] [Google Scholar]
  • 121.De Santis M, De Luca C, Mappa I, et al. Antiepileptic drugs during pregnancy: pharmacokinetics and transplacental transfer. Curr Pharm Biotechnol. 2011;12(5):781–788. doi: 10.2174/138920111795470958. [DOI] [PubMed] [Google Scholar]
  • 122.Bernus I, Hooper WD, Dickinson RG, et al. Metabolism of carbamazepine and co-administered anticonvulsants during pregnancy. Epilepsy Res. 1995;21(1):65–75. doi: 10.1016/0920-1211(95)00012-y. [DOI] [PubMed] [Google Scholar]
  • 123.Tomson T, Lindbom U, Ekqvist B, et al. Epilepsy and pregnancy: a prospective study of seizure control in relation to free and total plasma concentrations of carbamazepine and phenytoin. Epilepsia. 1994;35(1):122–130. doi: 10.1111/j.1528-1157.1994.tb02921.x. [DOI] [PubMed] [Google Scholar]
  • 124.Battino D, Binelli S, Bossi L, et al. Plasma concentrations of carbamazepine and carbamazepine 10,11-epoxide during pregnancy and after delivery. Clin Pharmacokinet. 1985;10(3):279–284. doi: 10.2165/00003088-198510030-00007. [DOI] [PubMed] [Google Scholar]
  • 125.Dam M, Christiansen J, Munck O, et al. Antiepileptic drugs: metabolism in pregnancy. Clin Pharmacokinet. 1979;4(1):53–62. doi: 10.2165/00003088-197904010-00005. [DOI] [PubMed] [Google Scholar]
  • 126.Lander CM, Eadie MJ. Plasma antiepileptic drug concentrations during pregnancy. Epilepsia. 1991;32(2):257–266. doi: 10.1111/j.1528-1157.1991.tb05253.x. [DOI] [PubMed] [Google Scholar]
  • 127.Gjerde IO, Strandjord RE, Ulstein M. The course of epilepsy during pregnancy: a study of 78 cases. Acta Neurol Scand. 1988;78(3):198–205. doi: 10.1111/j.1600-0404.1988.tb03646.x. [DOI] [PubMed] [Google Scholar]
  • 128.Otani K. Risk factors for the increased seizure frequency during pregnancy and puerperium. Folia Psychiatr Neurol Jpn. 1985;39(1):33–41. doi: 10.1111/j.1440-1819.1985.tb01942.x. [DOI] [PubMed] [Google Scholar]
  • 129.Yerby MS, Friel PN, McCormick K, et al. Pharmacokinetics of anticonvulsants in pregnancy: alterations in plasma protein binding. Epilepsy Res. 1990;5(3):223–228. doi: 10.1016/0920-1211(90)90042-t. [DOI] [PubMed] [Google Scholar]
  • 130.Wisner KL, Leckman-Westin E, Finnerty M, et al. Valproate prescription prevalence among women of childbearing age. Psychiatr Serv. 2011;62(2):218–220. doi: 10.1176/ps.62.2.pss6202_0218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Philbert A, Pedersen B, Dam M. Concentration of valproate during pregnancy, in the newborn and in breast milk. Acta Neurol Scand. 1985;72(5):460–463. doi: 10.1111/j.1600-0404.1985.tb00901.x. [DOI] [PubMed] [Google Scholar]
  • 132.Levy RH, Yerby MS. Effects of pregnancy on antiepileptic drug utilization. Epilepsia. 1985;26(suppl 1):S52–S57. doi: 10.1111/j.1528-1157.1985.tb05724.x. [DOI] [PubMed] [Google Scholar]
  • 133.Koerner M, Yerby M, Friel P, et al. Valproic acid disposition and protein binding in pregnancy. Ther Drug Monit. 1989;11(3):228–230. doi: 10.1097/00007691-198905000-00002. [DOI] [PubMed] [Google Scholar]
  • 134.Yerby MS, Friel PN, McCormick K. Antiepileptic drug disposition during pregnancy. Neurology. 1992;42(4 suppl 5):12–16. [PubMed] [Google Scholar]
  • 135.Johannessen SI. Pharmacokinetics of valproate in pregnancy: mother-foetus-newborn. Pharm Weekbl Sci. 1992;14(3A):114–117. doi: 10.1007/BF01962699. [DOI] [PubMed] [Google Scholar]
  • 136.Riva R, Albani F, Contin M, et al. Mechanism of altered drug binding to serum proteins in pregnant women: studies with valproic acid. Ther Drug Monit. 1984;6(1):25–30. doi: 10.1097/00007691-198403000-00006. [DOI] [PubMed] [Google Scholar]
  • 137.Linden S, Rich CL. The use of lithium during pregnancy and lactation. J Clin Psychiatry. 1983;44(10):358–361. [PubMed] [Google Scholar]
  • 138.Ward S, Wisner KL. Collaborative management of women with bipolar disorder during pregnancy and postpartum: pharmacologic considerations. J Midwifery Womens Health. 2007;52(1):3–13. doi: 10.1016/j.jmwh.2006.09.002. [DOI] [PubMed] [Google Scholar]
  • 139.Newport DJ, Viguera AC, Beach AJ, et al. Lithium placental passage and obstetrical outcome: implications for clinical management during late pregnancy. Am J Psychiatry. 2005;162(11):2162–2170. doi: 10.1176/appi.ajp.162.11.2162. [DOI] [PubMed] [Google Scholar]
  • 140.Sabers A. Algorithm for lamotrigine dose adjustment before, during, and after pregnancy. Acta Neurol Scand. 2012;126(1):e1–e4. doi: 10.1111/j.1600-0404.2011.01627.x. [DOI] [PubMed] [Google Scholar]
  • 141.Krishnamurthy KB. Managing epilepsy during pregnancy: assessing risk and optimizing care. Curr Treat Options Neurol. 2012;14(4):348–355. doi: 10.1007/s11940-012-0184-7. [DOI] [PubMed] [Google Scholar]
  • 142.DeVane CL, Liston HL, Markowitz JS. Clinical pharmacokinetics of sertraline. Clin Pharmacokinet. 2002;41(15):1247–1266. doi: 10.2165/00003088-200241150-00002. [DOI] [PubMed] [Google Scholar]
  • 143.Lundmark J, Reis M, Bengtsson F. Therapeutic drug monitoring of sertraline: variability factors as displayed in a clinical setting. Ther Drug Monit. 2000;22(4):446–454. doi: 10.1097/00007691-200008000-00014. [DOI] [PubMed] [Google Scholar]
  • 144.Mauri MC, Laini V, Cerveri G, et al. Clinical outcome and tolerability of sertraline in major depression: a study with plasma levels. Prog Neuropsychopharmacol Biol Psychiatry. 2002;26(3):597–601. doi: 10.1016/s0278-5846(01)00314-1. [DOI] [PubMed] [Google Scholar]
  • 145.Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression. Development of the 10-item Edinburgh Postnatal Depression Scale. Br J Psychiatry. 1987;150:782–786. doi: 10.1192/bjp.150.6.782. [DOI] [PubMed] [Google Scholar]
  • 146.Bergink V, Kooistra L, Lambregtse-van den Berg MP, et al. Validation of the Edinburgh Depression Scale during pregnancy. J Psychosom Res. 2011;70(4):385–389. doi: 10.1016/j.jpsychores.2010.07.008. [DOI] [PubMed] [Google Scholar]
  • 147.Sachs GS, Guille C, McMurrich SL. A clinical monitoring form for mood disorders. Bipolar Disord. 2002;4(5):323–327. doi: 10.1034/j.1399-5618.2002.01195.x. [DOI] [PubMed] [Google Scholar]
  • 148.Oberlander TF, Warburton W, Misri S, et al. Effects of timing and duration of gestational exposure to serotonin reuptake inhibitor antidepressants: population-based study. Br J Psychiatry. 2008;192(5):338–343. doi: 10.1192/bjp.bp.107.037101. [DOI] [PubMed] [Google Scholar]
  • 149.Warburton W, Hertzman C, Oberlander TF. A register study of the impact of stopping third trimester selective serotonin reuptake inhibitor exposure on neonatal health. Acta Psychiatr Scand. 2010;121(6):471–479. doi: 10.1111/j.1600-0447.2009.01490.x. [DOI] [PubMed] [Google Scholar]
  • 150.Viguera AC, Nonacs R, Cohen LS, et al. Risk of recurrence of bipolar disorder in pregnant and nonpregnant women after discontinuing lithium maintenance. Am J Psychiatry. 2000;157(2):179–184. doi: 10.1176/appi.ajp.157.2.179. [DOI] [PubMed] [Google Scholar]
  • 151.Sproule B. Lithium in bipolar disorder: can drug concentrations predict therapeutic effect? Clin Pharmacokinet. 2002;41(9):639–660. doi: 10.2165/00003088-200241090-00002. [DOI] [PubMed] [Google Scholar]
  • 152.Schou M. Lithium treatment during pregnancy, delivery, and lactation: an update. J Clin Psychiatry. 1990;51(10):410–413. [PubMed] [Google Scholar]
  • 153.Galbally M, Snellen M, Walker S, et al. Management of antipsychotic and mood stabilizer medication in pregnancy: recommendations for antenatal care. Aust N Z J Psychiatry. 2010;44(2):99–108. doi: 10.3109/00048670903487217. [DOI] [PubMed] [Google Scholar]
  • 154.Malzacher A, Engler H, Drack G, et al. Lethargy in a newborn: lithium toxicity or lab error? J Perinat Med. 2003;31(4):340–342. doi: 10.1515/JPM.2003.049. [DOI] [PubMed] [Google Scholar]
  • 155.Tanaka T, Moretti ME, Verjee ZH, et al. A pitfall of measuring lithium levels in neonates. Ther Drug Monit. 2008;30(6):752–754. doi: 10.1097/FTD.0b013e3181898978. [DOI] [PubMed] [Google Scholar]
  • 156.Schou M, Amdisen A, Steenstrup OR. Lithium and pregnancy. II. Hazards to women given lithium during pregnancy and delivery. Br Med J. 1973;2(5859):137–138. doi: 10.1136/bmj.2.5859.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Perucca E. Is there a role for therapeutic drug monitoring of new anticonvulsants? Clin Pharmacokinet. 2000;38(3):191–204. doi: 10.2165/00003088-200038030-00001. [DOI] [PubMed] [Google Scholar]
  • 158.Harlow BL, Vitonis AF, Sparen P, et al. Incidence of hospitalization for postpartum psychotic and bipolar episodes in women with and without prior prepregnancy or prenatal psychiatric hospitalizations. Arch Gen Psychiatry. 2007;64(1):42–48. doi: 10.1001/archpsyc.64.1.42. [DOI] [PubMed] [Google Scholar]
  • 159.Heron J, Robertson Blackmore E, McGuinness M, et al. No ‘latent period’ in the onset of bipolar affective puerperal psychosis. Arch Womens Ment Health. 2007;10(2):79–81. doi: 10.1007/s00737-007-0174-z. [DOI] [PubMed] [Google Scholar]
  • 160.Sit D, Rothschild AJ, Wisner KL. A review of postpartum psychosis. J Womens Health (Larchmt) 2006;15(4):352–368. doi: 10.1089/jwh.2006.15.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Mawhinney E, Campbell J, Craig J, et al. Valproate and the risk for congenital malformations: is formulation and dosage regime important? Seizure. 2012;21(3):215–218. doi: 10.1016/j.seizure.2012.01.005. [DOI] [PubMed] [Google Scholar]
  • 162.Semczuk-Sikora A, Czuczwar S, Semczuk A, et al. Valproic acid transfer across human placental cotyledon during dual perfusion in vitro. Ann Agric Environ Med. 2010;17(1):153–157. [PubMed] [Google Scholar]
  • 163.Tsuru N, Maeda T, Tsuruoka M. Three cases of delivery under sodium valproateVplacental transfer, milk transfer and probable teratogenicity of sodium valproate. Jpn J Psychiatry Neurol. 1988;42(1):89–96. doi: 10.1111/j.1440-1819.1988.tb01960.x. [DOI] [PubMed] [Google Scholar]
  • 164.Sirmagul B, Atli O, Ilgin S. The effect of combination therapy on the plasma concentrations of traditional antiepileptics: a retrospective study. Hum Exp Toxicol. 2012;31(10):971–980. doi: 10.1177/0960327112446516. [DOI] [PubMed] [Google Scholar]
  • 165.Tuccori M, Testi A, Antonioli L, et al. Safety concerns associated with the use of serotonin reuptake inhibitors and other serotonergic/noradrenergic antidepressants during pregnancy: a review. Clin Ther. 2009;31(Pt 1):1426–1453. doi: 10.1016/j.clinthera.2009.07.009. [DOI] [PubMed] [Google Scholar]
  • 166.Matsui DM. Therapeutic drug monitoring in pregnancy. Ther Drug Monit. 2012;34(5):507–511. doi: 10.1097/FTD.0b013e318261c372. [DOI] [PubMed] [Google Scholar]

RESOURCES