The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
ReviewsFull Access

Complex Combination Pharmacotherapy for Bipolar Disorder: Knowing When Less Is More or More Is Better

Abstract

Combination pharmacotherapy for bipolar disorder is commonplace and often reflects the severity and complexity of the illness and the comorbid conditions frequently associated with it. Across treatment settings, about one-fifth of patients with bipolar disorder appear to receive four or more psychotropic medications. Practice patterns often outpace the evidence-based literature, insofar as few systematic studies have examined the efficacy and safety of two or more medications for any given phase of illness. Most randomized trials of combination pharmacotherapy focus on the utility of pairing a mood stabilizer with a second-generation antipsychotic for prevention of either acute mania or relapse. In real-world practice, patients with bipolar disorder often take more elaborate combinations of mood stabilizers, antipsychotics, antidepressants, anxiolytics, stimulants, and other psychotropics for indefinite periods that do not necessarily arise purposefully and logically. In this article, I identify clinical factors associated with complex combination pharmacotherapy for patients with bipolar disorder; describe approaches to ensuring that each component of a treatment regimen has a defined role; discuss the elimination of unnecessary, ineffective, or redundant drugs in a regimen; and address complementary, safe, rationale-based drug combinations that target specific domains of psychopathology for which monotherapies often provide inadequate benefit.

Combination pharmacotherapy—sometimes derisively called polypharmacy to connote drugs that are deemed unnecessary or inappropriate in a regimen—has long been a cornerstone of treatment for complex medical conditions ranging from hypertension to infectious disease to oncology. Psychiatry has lagged behind other areas of medicine by fostering the idea that psychiatric disorders somehow ought to be treatable with only one medication, no matter how complex the problem may be, and that the deliberate juxtaposition of two or more drugs likely reflects shoddy prescribing rather than the pursuit of pharmacodynamic synergy. In the case of bipolar disorder, in which comorbid conditions are more common than rare (1) and symptom targets are often heterogeneous, the idea that one medication will reliably and effectively resolve all features of a complex clinical presentation is often unrealistic and naïve. In part, such fanciful expectations reflect a dubious assumption that a single underlying neurobiological process accounts for the entirety and diversity of psychopathology features shown by people with bipolar disorder. Seldom does one all-encompassing intervention ameliorate all signs of psychopathology as comprehensively as an antibiotic for pneumonia treats cough, fever, weakness, and dyspnea or a nitrate for angina eliminates chest pain, nausea, diaphoresis, and weakness. Accordingly, in this article, I provide an overview of the varied symptoms of psychopathology for which patients with bipolar disorder are prescribed medications so that practitioners can devise regimens that are complementary, nonredundant, purposeful, and evidence based.

Targets of Pharmacotherapy

Rather than speak of medications in a narrow fashion as simply treating mania or depression, it is often more useful to identify their utility for specific targets of pharmacotherapy according to dimensions of psychopathology, as illustrated in Figure 1.

FIGURE 1.

FIGURE 1. Domains of psychopathology

Ideally, a pharmacotherapy regimen for bipolar disorder includes as many components as necessary to successfully address all the relevant aspects of psychopathology described in Figure 1. Elegant combination pharmacotherapy is economical (making use of one drug with multiple effects when feasible), purposeful (with every component serving a definable function), and efficient (with dosing and the number of drugs no higher than necessary to address the intended targets). Less elegant are assemblages of medications that accrue over time without regard to intended effects, lack of efficacy, mechanistic redundancies, pharmacokinetic interactions, or ongoing relevance.

Prevalence of Prescription of Complex Combination Pharmacotherapy

Cross-sectional descriptive studies have suggested that about one-fifth (2) to one-third (3) of patients with bipolar disorder are taking four or more psychotropic medications at the time of hospitalization. Among 4,035 mostly outpatient entrants to the National Institute of Mental Health (NIMH) Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD), 18% were taking four or more psychotropic drugs, and 40% took three or more (4). One European study found that nearly 80% of outpatients with bipolar disorder took a mean of 3.8 medications, most often a mood stabilizer (92%), followed by an antidepressant (59%), benzodiazepines (43%), antipsychotics (39%), and thyroid hormone (21%; 5). Another report from the United Kingdom found that, in 2009, nearly half of patients with bipolar disorder were prescribed two or more psychotropic drugs on an ongoing basis; nearly half of that group chronically took lithium plus an antipsychotic drug (6).

Separate from merely counting how many medications a patient takes is determining whether the components of a pharmacotherapy regimen are appropriate and complementary (as opposed to inappropriate and pharmacologically conflictual). Considerations of drug appropriateness include the following:

  • Is the intended use of a drug aligned with its known pharmacodynamic effects? For example, lamotrigine would not be considered relevant to treat acute mania; chronic sleep aids may be unnecessary and counterproductive in patients with hypersomnia.

  • Are the known pharmacodynamic effects of a drug contrary to current symptoms? Antidepressants generally have no value during acute mania; and psychostimulants are likely counterproductive when treating acute psychosis.

  • Do the effects of one drug contradict or conflict with those of another? An example is simultaneous use of an anticholinergic drug (such as benztropine) and a procholinergic drug (such as donepezil).

  • Are current medications consistent with the patient’s present clinical state? Antidepressants would be illogical during acute mania, as would be the absence of one or more antimanic drugs.

  • Are adverse effects of one drug causing or mimicking psychiatric symptoms? Akathisia can be mistaken for agitation; anticholinergics impair cognitive function; and sleeplessness or loss of libido can be caused by a monoaminergic antidepressant or by depression. Adverse effects may also exacerbate a medical problem (e.g., beta blockers worsening asthma; noradrenergic drugs driving hypertension).

  • Are medications being retained if they have been deemed ineffective (and have all medications in a regimen received an adequate trial and been deemed effective or ineffective)?

  • Are medications being used and retained even when they have an abundance of negative controlled trial data for an intended purpose, such as topiramate or gabapentin in mania or paroxetine in bipolar depression, or an illogical rationale, such as use of modafinil or amphetamine for agitation or anxiety?

  • Have clinically important drug interactions gone unrecognized? For example, carbamazepine and primidone are potent inducers of cytochrome P450 enzymes and may effectively reduce serum levels and pharmacodynamic efficacy of drugs that undergo Phase I oxidative metabolism.

Do Guidelines Offer Guidance?

Practice guidelines convey only limited insight into the utility of complex combination therapy. Beyond specifying episode features such as with versus without psychosis or mixed versus pure affective polarity, they tend to say little about contexts and comorbid conditions that typically influence treatment decisions. Examples might include mania in someone with or without metabolic syndrome, in someone with or without substance misuse, or in a sexually active woman of child-bearing age or a person with bipolar depression with versus without prominent anxiety, with comorbid ADHD, or with suicidal features. For a patient with severe, acute mania, some guidelines advocate a combination of two drugs (usually lithium or divalproex plus an antipsychotic) as a first- (79) or second- (10) line intervention. Some make no specific mention of combining three or more agents in any phase of bipolar disorder so much as replacing an ineffective medication with an alternative (e.g., the 2018 Canadian Network for Mood and Anxiety Treatments Guidelines; 8). Others identify triple (or greater) therapy regimens as worthy of consideration only after the failure of multiple single- or dual-drug therapy efforts for manic or mixed episodes (e.g., 9, 10), and these regimens usually consist of two mood stabilizers plus a first-generation antipsychotic or a second-generation antipsychotic (SGA).

For acute bipolar depression, current guidelines have not suggested a role for more than two drugs in combination (generally a mood stabilizer or SGA plus an antidepressant). Guidelines usually list novel agents (such as pramipexole [11], inositol [12], anti-inflammatory or antioxidant drugs [13], or thyroid hormone [14]) collectively as last-step options, but without regard to the total net number of medications in a regimen. Most also seldom offer explicit recommendations about when and how to discontinue a drug once it has been introduced into a regimen, apart from often blanket discouragement of long-term use of antidepressants for people with bipolar disorder (despite a lack of consensus among experts and a limited evidence base from which to inform decisions about deprescribing antidepressants when they are acutely effective; 15).

No systematic studies have looked at combining two (or more) monoaminergic antidepressants that have complementary mechanisms as a strategy to treat bipolar depression, unlike for major depressive disorder. The relatively small handful of randomized trials that have involved the use of traditional antidepressants to treat bipolar depression have generally not demonstrated robust efficacy—not unlike for treatment-resistant unipolar depression. However, with no studies of the use of combination antidepressants in treating bipolar depression, the potential utility of novel or multiple antidepressant approaches remains unknown. Clinical characteristics associated with the use of combination therapy for bipolar disorder are summarized in Table 1 (24, 1618).

TABLE 1. Factors associated with complex combination pharmacotherapy for patients with bipolar disorder

CharacteristicFindings
SexFemale (2)
Race or ethnicityWhite (2)
Age (years)>50 (16)
PsychosisPresent (2)
Drug dosageTypically low (16)
Depressive illness burdenHigh (4)
Comorbid personality disordersBorderline personality disorder (2)
Comorbid psychiatric diagnosesPosttraumatic stress disorder (2)
Anxiety disorders (2, 3)
History of 1 or more suicide attemptsPresent (3, 4)
Personality featuresLow ratings on openness, extraversion, and conscientiousness (17, 18)

TABLE 1. Factors associated with complex combination pharmacotherapy for patients with bipolar disorder

Enlarge table

Polypharmacy has been identified as one of the biggest contributors to guideline-discordant treatment of bipolar disorder (18), yet herein lies a major dilemma for practitioners: Guideline recommendations are based on traditional efficacy trials for specific phases of an illness rather than on effectiveness studies with real-world populations. Snippets of guideline recommendations can be judged for concordance with prescribed medications for specific illness phases of bipolar disorder (e.g., in the STEP-BD, guideline-concordant treatments were prescribed for more than 80% of manic-hypomanic, mixed, or depressive episodes per se; 19), but most patients with bipolar disorder outside of clinical trials require treatment for more than simply a current affective phase of illness. Clinician surveys that examine guideline nonadherence in treating bipolar disorder have pointed to the failure of guidelines to address particular features of unique clinical populations (20). Indeed, it would be difficult to craft an effective guideline-driven drug regimen for a bipolar II depressed patient with prominent anxiety, attentional complaints, substance use comorbidity, chronic suicidal ideation, metabolic syndrome, obstructive sleep apnea, and chronic kidney disease.

Guidelines become less useful when diagnoses and illness phases do not conform neatly to DSM-5 criteria, patient priorities drive adherence, and prominent comorbid conditions and complex features (e.g., trauma histories) must be addressed. In addition, the clinical trials literature does not indicate whether and when mono- or dual pharmacotherapies yield better outcomes than more extensive but thoughtfully devised combination regimens is largely unknown. The shortcomings of current practice guidelines for bipolar disorder are summarized in Box 1.

Box 1. Limitations of current practice guidelines for bipolar disorder

  • Largely ignore the presence of psychiatric and medical comorbid conditions.

  • Do not account for patients whose bipolar spectrum symptoms may not fulfill traditional DSM-5 diagnostic criteria.

  • Often lump together recommendations for patients with bipolar I vs. bipolar II disorder, those with vs. without rapid cycling, or those with chronic vs. nonchronic presentations.

  • Do not account for risk-benefit situations (e.g., metabolic risk vs. gravity of psychiatric disability when considering clozapine).

  • Minimally consider incorporating specific medications for narrow intended purposes (e.g., lithium to reduce suicide risk; topiramate solely for weight loss).

  • Do not account for multipurpose drugs with greater value in unique situations (e.g., divalproex for patients with bipolar disorder who experience migraines; bupropion for patients with bipolar II depression and obesity, cigarette smoking, and concerns about iatrogenic sexual dysfunction).

  • May blur opinion-based with evidence-based recommendations.

  • Provide little if any guidance on when and how to deprescribe.

Rationales for Pharmacotherapy Combinations

From one perspective, combining diverse medications makes sense with a disease entity in which known drug mechanisms of action are complementary and related to the pathophysiology of the underlying disease process. In the case of bipolar disorder, such exactitude is unfortunately largely unknown and may at best be speculative. Although a systematic review of current theories about the pathogenesis of bipolar disorder is beyond the scope of this article, theories that have been reviewed elsewhere (e.g., Manji et al. [21]), ranging from the level of molecular-intracellular function to neuronal networks, include the following: monoaminergic dysregulation; elevated intracellular calcium and abnormal calcium signaling (e.g., calcium channel blockade); circadian dysrhythmias (e.g., light entrainment of the circadian pacemaker; sleep-wake cycle perturbations); dysfunction of purine metabolism (hyperuricemia in mania); endocrinopathies (e.g., glucocorticoid receptor dysfunction, hypothalamic-pituitary-adrenocortical dysregulation, hypothalamic-pituitary-thyroid axis dysfunction); abnormal signal transduction (e.g., overactive inositol phosphate signaling, elevated intracellular inositol or cyclic adenosine monophosphate); immuno-inflammatory mechanisms, impaired neuronal plasticity, regulation of oxidative stress, and neuroprotection against neuronal loss; and disrupted connectivity of neural circuits.

Table 2 summarizes presumed neuronal and molecular-cellular mechanisms of action for psychotropic drugs commonly used to target mania, depressive symptoms, or both in patients with bipolar disorder (2255). The list of potential mechanisms is far from complete due to the limited knowledge about bipolar disorder’s basic pathophysiology, along with emerging novel hypotheses that do not yet clearly translate to any specific biological interventions (e.g., mitochondrial dysfunction, shortened telomere length).

TABLE 2. Putative mechanisms of action of antimanic and antidepressant drugsa

MechanismLithiumDVPXCBZLTGSGAsMADs
PKC inhibition√ (22)√ (23)− (24)− (25)??
BDNF upregulation√ (26)√ (23)√ (27)√ (28, 29)√ (30)√ (31)
bcl–2 upregulation√ (32)√ (33)√ (27)√ (34)? b? c
GSK3β inhibition√ (37)√ (38)? d? e
MARCKS downregulation√ (23)√ (23)
GABA upregulation√ (41)√ (42)√ (43)?
Glutamate downregulation√ (44)√ (42)√ (45)√ (46)?? f
Na+ channel blockade√ (48)√ (49)√ (50)√ (51)?g
Ca++ channel blockade
5-HT2A antagonismhh
5-HT1A partial agonism?ii
5-HT1B antagonism, partial agonism or inverse agonism? jk? k
D2 antagonismll√ lk
D3 partial agonismm
5-HT7 antagonismnn

a√, predominantly robust positive published associations; −, predominantly or solely negative published findings; ?, inadequate or inconclusive published findings from which to draw associations; BDNF, brain-derived neurotrophic factor; CBZ, carbamazepine; DVPX, divalproex; GABA, gamma-aminobutyric acid; LTG, lamotrigine; MARCKS, myristoylated alanine-rich C kinase substrate; MADs, monoaminergic antidepressants; PKC, protein kinase C.

bLimited preclinical data suggest that olanzapine and clozapine may upregulate regional b-cell lymphoma 2 (bcl–2 mRNA) and protein expression in rat frontal cortex and hippocampus (35).

cLimited preclinical data show increased bcl–2 mRNA production in frontal and subcortical brain regions of rats stressed and then treated with fluoxetine, reboxetine, tranylcypromine, or electroconvulsive seizures (36).

dPreclinical data show that olanzapine or aripiprazole, but not haloperidol, may attenuate stress-induced downregulation of hippocampal glycogen synthase kinase-3 (GSK3β) in rats subjected to immobilization stress (39).

eA small preclinical database suggests that some antidepressants (notably, fluoxetine) may regulate neurogenesis through GSK3β signaling (40).

fPreclinical (in vitro) studies suggest that selective serotonin reuptake inhibitors (SSRIs) may downregulate release of glutamate from microglia (47).

gTricyclics block sodium channels; effects of SSRIs or other monoaminergic antidepressants are less well established (52).

h5-HT2A antagonism associated with all atypical antipsychotics, mirtazapine, nefzodone, and trazodone.

i5HT1A partial agonism is associated with aripiprazole, asenapine, brexpiprazole, cariprazine, clozapine, lurasidone, olanzapine, quetiapine, ziprasidone, nefazodone, vilazodone; it is associated with full agonism with vortioxetine. Over time, SSRIs downregulate 5-HTA1 somatodendritic autoreceptors.

jPreliminary studies have suggested the possibility that 5-HT1B receptors may be a target of lithium (53).

k5-HT1B antagonism is associated with asenapine, ziprasidone, and aripiprazole; partial agonism is associated with vortioxetine.

lLithium, divalproex and carbamazepine may each indirectly downregulate D2 receptor signaling via G-protein–coupled effects (22, 54, 55)

mD3 partial agonists include aripiprazole, brexpiprazole, and cariprazine.

n5-HT7 antagonism associated with several second-generation antipsychotics (SGAs; asenapine, clozapine, lurasidone, olanzapine, and ziprasidone; aripiprazole is a weak partial agonist) and antidepressants (amitriptyline, clomipramine, imipramine, and vortioxetine).

TABLE 2. Putative mechanisms of action of antimanic and antidepressant drugsa

Enlarge table

In addition, as noted in the table and its footnotes, in many instances only preliminary animal or in vitro data exist regarding the effects of specific psychotropic drugs on possible receptor targets; these effects may not be so straightforward or easily translated. For example, neither lithium nor anticonvulsant mood stabilizers exert known direct effects on specific serotonin receptors, but they might exert indirect modulating effects; possible in vitro effects of monoaminergic antidepressants on ion channels may not translate to pharmacodynamic effects in humans. Preclinical or in vitro receptor binding studies may not necessarily provide information about regional anatomical differences in drug effects. Existing treatments, for better or worse, are geared more practically toward altering observable psychopathology rather than modifying an identified neurobiological process.

Some proposed mechanisms are likely more specific to the treatment of depression than to that of mania (e.g., serotonin reuptake inhibition) or vice versa (e.g., protein kinase C inhibition or inositol depletion), and even identified neuronal or molecular mechanisms do not necessarily provide information about pharmacodynamic class effects (e.g., mania may respond to some drugs that inhibit protein kinase C inhibitors [e.g. tamoxifen; 56], but not all [e.g., omega-3 fatty acids; 57]). Existing knowledge about drug mechanisms of action also does not specifically address pharmacodynamic effects relevant to particular illness subcomponents (e.g., use of a drug for narrow purposes such as putative antisuicide effects, such as lithium [58]), procognitive effects (e.g., withania somnifera [59] or lurasidone [60]), anticraving effects (e.g., naltrexone), or targeting of impulsive aggression (e.g., divalproex; 61). It is also worth noting that nuanced differences exist within the broad mechanisms included in Table 2 whose depth is beyond the scope of this article. For example, some anticonvulsant drugs (divalproex, carbamazepine) are believed to exert antiglutamatergic effects only through N-methyl-D-aspartate receptor blockade, whereas others (lamotrigine) appear to also affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. Calcium channel blocking effects of mood-stabilizing anticonvulsants may occur through L-type calcium channels (carbamazepine); L- and T-type calcium channels (divalproex); or L-, P/Q-, R-, and T-type calcium channels (lamotrigine). Among SGAs, individual agents vary in their specific receptor profiles and affinities, as well as in their unique targets (e.g., 5-HT7 antagonism or D3 partial agonism are not universally shared targets across SGAs, as noted in Table 2).

Complex Combination Pharmacotherapy for Bipolar Disorder and Clinical Outcomes

Almost no systematic studies have provided information about whether and when a more extensive pharmacotherapy regimen leads to a better or worse clinical outcome than a simpler regimen. Such questions may never be answerable because of the inherent complexities of controlled study designs and the matching of drug regimens to individual patient characteristics that in themselves influence medication choice. It is obviously also an oversimplification to count the sheer number of medications someone takes without regard to the appropriateness, responsivity, redundancies, and purposefulness of each drug on a treatment roster, as though counting instruments in an orchestra without distinguishing woodwinds from brass or strings or the composition of the overall ensemble. Specific medication choices in a given regimen, and their response likelihoods, are also often made not just on the basis of an overall Food and Drug Administration indication (e.g., acute mania) but more precisely according to other characteristics associated with more specific symptom domains (e.g., divalproex vs. lithium in mixed episodes; lamotrigine for maintenance therapy in patients more prone to depressive rather than manic recurrences) (62).

Existing randomized combination pharmacotherapy trials for bipolar disorder focus mainly on the potential utility of two medications versus one during either acute manic or depressive phases of illness or during maintenance and relapse prevention phases. This literature is summarized in Tables 3-5, respectively, (6388) for treatment of acute bipolar manic or mixed episodes, acute bipolar depression, or bipolar maintenance and relapse prevention, respectively. For acute bipolar depression, Table 4 focuses on combinations of mood stabilizers or adjunctive SGAs with mood stabilizers; I did not include studies of traditional antidepressant or novel compounds (e.g., pramipexole, modafinil, psychostimulants) added to a mood stabilizer because such published studies generally do not account for variability in inherent potential antidepressant properties among mood stabilizers.

TABLE 3. Randomized trials of mono- vs. dual pharmacotherapies in acute bipolar manic or mixed episodes

Trial result and interventionDurationOutcomeAdverse effects
Positive trials
 Allupurinol (N=218) or placebo (N=215) plus mood stabilizers (meta-analysis of five randomized trials; 63)4–8 weeksAdjunctive allopurinol (dosed at 300–600 mg/day) was more effective than placebo in reducing baseline mania symptoms; greater effect in pure than in mixed maniaNo significant differences from placebo
 Aripiprazole (N=253) or placebo (N=131) plus lithium or divalproex (64)6 weeksAdjunctive aripiprazole (mean dose=19 mg/day) was more effective than placebo; significantly greater reduction in mood elevation, hypersexuality, irritability, speech, aggression, insight); no significantly greater reduction in depression severity scoresMore extrapyramidal symptoms with aripiprazole than with placebo, but no significant differences in weight changes or metabolic laboratory parameters
 Asenapine (N=158) or placebo (N=166) plus lithium or divalproex (65)12 weeks (primary efficacy at week 3)Adjunctive asenapine (mean dose=11.8 mg/day) was more effective than placebo in reducing mania symptoms; no between-groups differences in response or remission rates or changes from baseline in depression symptom severityMore sedation and somnolence, treatment-emergent depression, hypoesthesia, and weight gain with asenapine than with placebo
 Celecoxib (N=23) or placebo (N=23) plus divalproex (66)6 weeksAdjunctive celecoxib (400 mg/day) was more effective than placebo in reducing severity of mania symptomsNo significant differences from placebo
 Olanzapine (N=51) or placebo (N=48) plus lithium or divalproex (67)6 weeksAdjunctive olanzapine (mean modal dose=8.6 mg/day) was more effective than placebo in reducing both manic and depressive symptomsGreater weight gain with combination than with monotherapy
 Olanzapine (N=100) or placebo (N=101) plus lithium or divalproex (68)6 weeksAdjunctive olanzapine was more effective than placebo in reducing both mania and depressive symptomsMore weight gain and higher fasting blood sugar with olanzapine than with placebo
 Risperidone (N=52), haloperidol (N=53), or placebo (N=51) plus lithium or divalproex (69)3 weeksAdjunctive risperidone (mean dose=3.8 mg/day) or haloperidol (mean dose 6.2 mg/day) was more effective than placebo in reduction in mania symptom severityGreater weight gain with adjunctive risperidone than placebo
 Quetiapine (N=91) or placebo (N=100) plus lithium or divalproex (70)3 weeksAdjunctive quetiapine (mean dose=504 mg/day) was more effective than placebo in reduction in mania symptom severity reduction (greatest improvements in irritability, aggression, sleep, and motor activity)Greater somnolence and dry mouth with adjunctive quetiapine than with placebo
 Lithium (N=173) or placebo (N=182) plus quetiapine XR (71)6 weeksAdjunctive lithium (mean modal dose=1,085.5 mg/day, mean serum [Li+]=0.72 mEq/L) was more effective than placebo in reduction in mania symptom severity, psychosis, agitation, aggression; no between-group differences in reduction in depressive symptom severity (mean quetiapine dose=623.1 mg/day)More tremor, somnolence, dizziness, diarrhea, and vomiting with lithium than with placebo
 Tamoxifen (N=20) or placebo (N=20) plus lithium (72)6 weeksAdjunctive tamoxifen (fixed dose 80 mg twice daily) was more effective than placeboMore fatigue with tamoxifen than with placebo
Negative trials
 Olanzapine (N=58) or placebo (N=60) plus carbamazepine (73)6 weeksNo significant between-groups differences in mania symptom severity or response rates with adjunctive olanzapine plus carbamazepine (mean dose=617.5 mg/day) vs. carbamazepine monotherapy (mean dose=717.3 mg/day)Greater weight gain and higher triglyceride levels with olanzapine than with placebo; carbamazepine is known to reduce serum olanzapine levels by approximately 40%–50%
 Paliperidone ER (N=150) or placebo (N=150) plus lithium or divalproex (74)6 weeksAdjunctive paliperidone (mean dose=8.1 mg/day) no different from placeboGreater somnolence, akathisia, weight gain, and increased appetite with paliperidone ER than with placebo
 Topiramate (N=143) or placebo (N=144) plus divalproex or lithium (75)8 weeksAdjunctive topiramate (mean dose 254.7 mg/day) no different from placeboParesthesias, diarrhea, and anorexia more common with topiramate than with placebo
 Ziprasidone (N=439) or placebo (N=217) plus lithium or divalproex (76)3 weeksAdjunctive ziprasidone (mean modal dose=60.1 mg/day in low-dose arm, 133.8 mg/day in high dose arm) no different from placebo in changes from baseline mania or levels of depression symptom severityMore sedation and somnolence with ziprasidone than with placebo

TABLE 3. Randomized trials of mono- vs. dual pharmacotherapies in acute bipolar manic or mixed episodes

Enlarge table

TABLE 4. Randomized Trials of mono- versus dual pharmacotherapies for acute bipolar depression

InterventionDurationOutcomeAdverse effects
Lamotrigine (N=64) or placebo (N=60) plus lithium (77)8 weeksAdjunctive lamotrigine (200 mg/day) was more effective than placeboNone greater with adjunctive lamotrigine than with lithium monotherapy
Lamotrigine (N=101) or placebo (N=101) plus quetiapine (78)12 weeksAdjunctive lamotrigine (200 mg/day) > placeboNone greater with adjunctive lamotrigine than with placebo
Lurasidone (N=183) or placebo (N=165) plus lithium or divalproex (79)6 weeksAdjunctive lurasidone (mean dose 66.3 mg/day) was more effective than placeboGreater nausea, somnolence, tremor, akathisia, and insomnia with adjunctive lurasidone than with placebo
Ziprasidone (N=147) or placebo (N=147) plus lithium or divalproex or lamotrigine (80)6 weeksAdjunctive ziprasidone (mean dose=89.8 mg/day) no different from placeboMore nausea, fatigue, dizziness, sedation, somnolence with adjunctive ziprasidone than with placebo

TABLE 4. Randomized Trials of mono- versus dual pharmacotherapies for acute bipolar depression

Enlarge table

TABLE 5. Randomized trials of mono- versus dual pharmacotherapies for bipolar maintenance and relapse prevention

Trial result and interventionDurationOutcomeAdverse effects
Positive trials
 Aripiprazole (N=168) or placebo (N=169) plus lithium or divalproex (81)12-week acute stabilization followed by 52-week maintenanceLower 52-week relapse rate (any mood episode) with aripiprazole (mean dose 15.8–16.9 mg/day; 17%) than with placebo (29%); aripiprazole was more effective than placebo for mania but not depressive relapseComparable weight changes or metabolic parameters with aripiprazole or placebo
 Lithium plus divalproex combination vs. either monotherapy (BALANCE; 82)24 monthsLithium plus divalproex (N=110) was more effective than divalproex monotherapy (N=110) but not lithium monotherapy (N=110); lithium was more effective than divalproex for depressive relapsesNo significant differences in serious adverse events among the treatment arms
 Lithium or divalproex plus aripiprazole of placebo (83)12-week acute stabilization followed by 52-week maintenanceAdjunctive aripiprazole (mean dose=16.1 mg/day if index episode manic, 16.8 mg/day if mixed) was more effective than placebo for prevention of manic but not mixed episodesMore tremor with aripiprazole than with placebo
 Olanzapine (N=51) or placebo (N=48) plus lithium or divalproex (84)18 monthsAdjunctive olanzapine (mean dose=8.6 mg/day) was more effective than placebo for symptomatic but not syndromal relapse for mania or depression (but no significant differences for time to mania relapse or time to depressive relapse)More weight gain and less insomnia with adjunctive olanzapine than with placebo
 Risperidone long-acting injectable (N=65) or placebo (N=59) plus treatment as usual (85)16-week open stabilization, then 52 weeksAdjunctive risperidone long-acting injectable (modal dose 25 mg/day) was more effective than placebo for time to any mood episodeTremor, insomnia, muscle rigidity, and mania more likely with risperidone long-acting injectable than with placebo
 Ziprasidone (N=127) or placebo (N=113) plus lithium or divalproex (86)8-week open stabilization, then 16 weeksAdjunctive ziprasidone (mean modal doses of 80, 119, and 160 mg/day across three strata) was more effective than placebo in time to any mood episode and, separately, time until a manic or until a depressive relapseIncidence of tremor greater with ziprasidone than placebo
Negative trials
 Lurasidone (N=246) or placebo (N=250) plus lithium or divalproex (87)28 weeksNo advantage for adjunctive lurasidone over placebo in time until recurrence of a depressive, manic, or mixed episode; if index episode polarity was depressed, lurasidone was more effective than placebo for time until recurrence of any mood episodeNo clinically meaningful differences with lurasidone versus placebo
 Oxcarbazepine (N=26) or placebo (N=29) plus lithium (88)52 weeksNo advantage for adjunctive oxcarbazepine (mean modal dose 1,200 mg/day) over placebo in time until recurrence of any mood episodeDrowsiness, diarrhea, and tremor more common with oxcarbazepine than placebo

TABLE 5. Randomized trials of mono- versus dual pharmacotherapies for bipolar maintenance and relapse prevention

Enlarge table

Some naturalistic studies have suggested that patients with bipolar disorder who take fewer medications (e.g., one) manifest more symptom stability (remain relapse-free) over time than those on two, three, or more medications (5, 89); still others have suggested that when medication regimens include certain core pharmacotherapies such as lithium, there may be less need to add additional medications (90). Open (nonrandomized) data suggest that inpatients with mania who begin pharmacotherapy with a combination of lithium and divalproex at the outset may incur a lesser dosage burden of antipsychotic cotherapy than those who begin on lithium without divalproex (91). The NIMH Lithium Treatment Moderate Dose Use Study (LiTMUS) trial found that cotherapy with subtherapeutic doses of lithium has not shown greater symptomatic benefit than placebo (although a post hoc analysis suggested that low-dose adjunctive lithium may be associated with lower dosages of concomitant SGAs; 92). By contrast, use of other medications (notably, chronic benzodiazepine use) in a regimen for treatment of bipolar disorder may be associated with higher recurrence rates and more severe overall illness (93)—not necessarily in a causal fashion, but more likely because of the stigmata of pathology.

The literature on extensive polypharmacotherapy of psychotropic drugs is largely more naturalistic and observational than randomized, which makes it hard to draw causal inferences about treatment outcomes. In such studies, medications are usually chosen on the basis of patients’ individual conditions, leading to the problem of confounding by indication. For example, some observational studies have attributed lower suicide attempts or completions to the inclusion of lithium in a treatment regimen over other mood stabilizers (94), but these studies have not accounted for the possibility that clinicians might avoid prescribing lithium to patients at a higher risk of suicide (e.g., in the aftermath of an overdose or other attempt), relegating prescription of lithium to a potential marker or artifact of a better prognosis at baseline. Similarly, some observational studies of rapid cycling have concluded that prescription of antidepressants may lead to more frequent episodes (95), without considering the reverse possibility that frequent depressive episodes may cause more prescription of antidepressants. In any nonrandomized pharmacotherapy study (but especially in those involving multidrug regimens), possible confounding by indication poses obstacles to inferring causal drug effects as a result of unrecognized and unaccounted-for moderators and mediators of outcome (62).

A further dilemma when trying to assess extensive combination pharmacotherapy regimens involves uncertainties about within-class differences versus generalizabilities among medications. For example, the relative magnitude of antimanic efficacy across individual SGAs appears comparable (96), although individual agents may vary in their antidepressant, anxiolytic, or tolerability profiles. Similarly, monoaminergic antidepressants are often lumped together as homogeneous entities, although the absence of placebo-controlled studies for most newer agents (vortioxetine, vilazodone, desvenlafaxine, levomilnacipran, mirtazapine) makes it difficult to generalize about within-class efficacy or the potential for synergy from particular antidepressant combinations.

What About Antidepressant Adjuncts to Mood Stabilizers for Bipolar Disorder?

Practice guidelines (7, 8, 10) and expert consensus statements (15) emphasize the importance of avoiding antidepressant monotherapies in bipolar I depression (with possibly greater leniency in bipolar II depression). Such recommendations mainly reflect concerns about the potential for antidepressants to destabilize mood via a treatment-emergent affective switch (TEAS). Consequently, guidelines have advised using antidepressants only in tandem with mood stabilizers in bipolar depression, mainly on the basis of assumptions that mood stabilizers help safeguard against TEAS outcomes rather than the perception that most mood stabilizers exert meaningful intrinsic antidepressant effects that synergize with antidepressant cotherapies. One often cited example, the NIMH STEP-BD study, found only a modest antidepressant response rate with mood stabilizer monotherapy (about 24%) and no added benefit with antidepressant cotherapy (97). Said differently, the current evidence base has suggested that neither mood stabilizers nor monoaminergic antidepressants, nor their combination, exert reliable and robust antidepressant effects on bipolar depression. Yet, contemporary observational studies have found that about one-third of patients with bipolar disorder take antidepressants on a long-term basis (>90 days) as part of their overall pharmacotherapy regimen (98). Such prescribing habits notwithstanding, the Florida Medicaid Guidelines (9) noted that “there is inadequate information (including negative trials) to recommend adjunctive antidepressants . . . for bipolar depression.”

It is also worth noting that clinicians in real-world practice settings may sometimes use selective serotonin reuptake inhibitors (SSRIs) or other antidepressants for their putative anxiolytic properties (in addition to, or possible entirely apart from, depression as an intended treatment target). However, apart from one secondary analysis from a negative randomized controlled trial of paroxetine for bipolar depression (99), no prospective randomized trials have demonstrated anxiolytic efficacy for monoaminergic antidepressants in bipolar disorder. Although this absence of evidence is not evidence of absence and does not negate the possibility of benefit, prescribers must recognize that such assumptions have not been empirically tested.

Antipsychotic Combination Pharmacotherapy

The sustained use of two or more antipsychotic drugs, although more often seen with patients with schizophrenia or schizoaffective disorder, has been observed in 15%−20% of patients with bipolar disorder, often at prescribed daily doses higher than those seen during monotherapy (100). On entry into the STEP-BD, about 10% of patients with bipolar disorder had been taking two or more antipsychotics (101). Psychosocial functioning and symptom status were no different from those who were taking only one antipsychotic, but side effect burden and service utilization were both significantly more extensive. Practice guidelines, such as the Texas Implementation of Medication Algorithms or the Florida Medicaid Guidelines, explicitly advise against the use of two (or more) SGAs, mirroring the generally minimal, if any, benefit seen with the use of antipsychotic combination pharmacotherapy with people with schizophrenia.

Triple Therapy

Very few randomized trials have examined the use of three (or more) psychotropic medications in any phase of bipolar disorder. The existing literature is limited by methodological limitations such as small sample sizes, high dropout, and poor tolerability. For example, among rapid cyclers whose symptoms had not stabilized after 16 weeks of lithium plus divalproex, Kemp et al. (102) found no advantage of the addition of lamotrigine over placebo but suggested that the failure to demonstrate an advantage may have been a Type II error because of the small number of randomized participants. Other short-term open trials with small sample sizes (i.e., N<10) or case series have shown improvement among outpatients with bipolar disorder whose inadequate responses to dual-agent mood stabilizers (lithium plus divalproex) improved with the addition of a third (e.g., carbamazepine), but the lack of larger trials or randomized study designs limits the ability to distinguish multidrug interventions from possible outcomes with alternative strategies, including longer trial durations on existing regimens.

Deprescribing and Maintenance of Pharmacological Hygiene

Surprisingly few randomized pharmacotherapy discontinuation trials exist in research on bipolar disorder. Although such studies provide the most rigorous data about the optimal treatment duration for any medication, that information is especially important during combination therapies, in which unnecessarily prolonged inclusion of a drug jeopardizes overall treatment adherence and imposes an additive side effect burden, cost, and risk for drug-drug interactions. To the extent that randomized clinical trials nowadays fall mainly under the auspices of pharmaceutical industry sponsorship, there is little economic incentive for commercial manufacturers to drive the deprescribing of their products.

Give the scarcity of empirical information about when a particular cotherapy may no longer be beneficial or necessary, the components of a pharmacology regimen can potentially accrue in a manner sometimes akin to hoarding. Because pharmacotherapy per se is for most patients with bipolar disorder an indefinite, open-ended undertaking, it can be difficult to fathom specified end points for stopping certain medications within a regimen. One conspicuous exception is the frequent admonition against long-term antidepressant use espoused in some practice guidelines, based on theoretical concerns about accelerated cycling frequency. In fact, however, randomized discontinuation trial data would suggest that, contrary to popular belief, cessation of an antidepressant after an initial robust response may incur a greater risk for depression relapse (103, 104), except in patients with past-year rapid cycling, for whom long-term antidepressant use has been associated with more depressive episodes (104).

Rapid cycling may represent a particular instance in which monotherapy may rarely produce improvement. A notable example is a randomized trial specifically with patients with rapid-cycling bipolar disorder (105) in which open-label dual therapy with lithium plus divalproex was then converted to monotherapy with either agent. Only one-quarter of 254 rapid-cycling patients initially stabilized during combination therapy with lithium plus divalproex (meaning that most rapid-cycling patients either poorly tolerated or did not benefit from this combination); after subsequent randomization to either agent as monotherapy, only about half of patients remained relapse free over a 20-month period. Unfortunately, this study did not examine whether the continued combination of lithium plus divalproex might have prolonged the time until relapse more than occurred with either pharmacotherapy. It also did not examine whether the total number of relapses per year among rapid-cycling patients was less after randomization to either monotherapy than before.

In the case of adjunctive SGAs, Yatham and colleagues (106) studied patients with manic or mixed bipolar episodes who were stabilized on lithium or divalproex plus an SGA (olanzapine or risperidone) and then compared time until relapse among those randomized to continued combination therapy versus mood stabilizer monotherapy for one year. Relapse rates were significantly lower for those in the combination condition than in the monotherapy condition (hazard ratio=0.53, 95% confidence interval=0.33–0.86) up to 24 weeks, but beyond that time frame the combination provided no continued advantage in preventing relapse over monotherapy. However, weight gain was significantly greater with a continued adjunctive SGA beyond this time (mean 3.2 kg gain vs. ≤0.1 kg gain, respectively).

A basic principle of psychopharmacological hygiene involves discontinuing a psychotropic drug that exerts no benefit after an adequate trial has elapsed. More ambiguous can be knowing whether and when to stop ancillary pharmacotherapies for bipolar disorder, such as benzodiazepines or sleep aids, nonbenzodiazepine anxiolytics (buspirone, hydroxyzine, anxiolytic anticonvulsants), psychostimulants (particularly if used off label to counteract sedation, obesity, or slowed attentional processing), beta blockers for social anxiety or tremor, thyroid hormone in the absence of intrinsic thyroid disease (or safety concerns related to cardiac arrhythmias or osteoporosis), and vitamins or nutritional supplements.

Other tenets of basic pharmacological hygiene include avoiding mechanistic redundancies, contradictory mechanisms, and irrational strategies to avoid future events (e.g., maintaining antidepressants during an acute manic episode on the basis of the notion that doing so may ward off a subsequent depression); optimizing dosing of a first drug before adding additional agents aimed at the same intended symptom target; not maintaining sleep aids when hypersomnia is present; minimizing the number of new or additive drug exposures during pregnancy, to the extent feasible; and knowing why a prescribed drug was originally introduced to a regimen and addressing its utility and ongoing relevance for a given patient.

As a practical matter, it can be helpful (as well as rapport building) for patients and clinicians to conduct periodic “performance evaluations” of a pharmacotherapy regimen and review and mutually decide what value and cost each drug brings to the patient’s treatment. Medications whose relevance is uncertain can then be further assessed one by one (thus changing only one variable at a time), either by tracking intended target symptoms more closely and formally or by dosage reduction or cautious elimination to determine whether a unique benefit indeed exists.

Another useful practical rule of thumb involves the concept of making no changes to a drug regimen for at least four to six months once unequivocal wellness has been established. This approach may also sometimes include prolonging or suspending a cross-taper in progress. Although “stalled” cross-tapers can leave patients on potentially unnecessary or duplicative medications, and as such are less than elegant, greater gains are sometimes accomplished by changing nothing once wellness is achieved and letting patients accrue lengthier periods of euthymia before embarking on efforts to prune a drug regimen.

The four- to six-month time frame corresponds to the conventional time period that defines recovery, as noted by nomenclature task forces of both the American College of Neuropsychopharmacology (107) and the MacArthur Foundation (108). Symptoms that arise within this time frame after remission from an acute episode are generally recognized as signs of relapse (i.e., the index episode itself reignites), and symptoms that emerge beyond four to six months of wellness are considered signs of a recurrence (i.e., a new episode). Statistically, survivorship of four to six months of wellness after an index episode categorically places someone at a lesser risk for clinical deterioration and consequently represents a logical and potentially safer time to ask whether a particular component of a regimen is or is not actually necessary to sustain wellness.

Future Directions

Several important unmet needs exist for which further studies and guidelines can advance knowledge and clinical practice about complex combination pharmacotherapy for patients with bipolar disorder. These needs include examining sequential pharmacotherapies and specific augmentation-versus-replacement strategies among patients whose responses to core mood stabilizer monotherapies are inadequate; undertaking randomized discontinuation trials to determine whether and when a particular medication has a finite optimal duration of benefit (and at what point diminishing returns might be expected); and conducting formal studies designed specifically to treat common conditions that are comorbid with bipolar disorder, including anxiety disorders; alcohol and substance use disorders; and adult ADD, ADHD, and cognitive dysfunction. Such comorbid conditions contribute to combination pharmacotherapy, but the dearth of dedicated studies targeting such presentations severely limits the evidence base for differential therapeutics.

Other needs are identification of factors and clinical circumstances in which augmentation strategies versus switching from one drug to another produces better outcomes and testing hierarchical treatment approaches as a means to ultimately simplify pharmacotherapy regimens in complex or comorbid presentations. For example, in the case of bipolar disorder with comorbid obsessive-compulsive disorder, some authors have argued that optimization of mood stabilizers can adequately treat both conditions, obviating the necessity of SSRI cotherapy (109); in the case of mood disorder with concurrent alcohol use disorder, further studies are needed to determine whether and when pharmacotherapy of mood symptoms alone may simultaneously reduce excessive drinking (110).

Conclusions

Complex pharmacotherapy regimens can be elegant amalgams of complementary and synergistic elements in which the pharmacodynamic whole becomes greater than the sum of its parts, or they can represent more haphazard collections of medications united by more arbitrariness than purposeful intent. Good clinical practice involves following a logical rationale and combining appropriate, nonredundant drugs that serve identifiable functions and avoid pharmacokinetic or pharmacodynamic hazards. With diligent outcome tracking and a systematic approach to medication changes, logical and strategic medication combinations can be devised to target key symptom domains and minimize unnecessary exposure to redundant, ineffective, or otherwise superfluous psychotropic agents.

Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City.
Send correspondence to Dr. Goldberg ().

Dr. Goldberg is a consultant with Neurocrine, Lundbeck, Otsuka, Sunovion, and WebMD. He is with the speakers bureaus of Allergan, Neurocrine, Otsuka, Sunovion, and Takeda-Lundbeck and receives royalties from American Psychiatric Association Publishing.

References

1 McElroy SL, Altshuler LL, Suppes T, et al.: Axis I psychiatric comorbidity and its relationship to historical illness variables in 288 patients with bipolar disorder. Am J Psychiatry 2001; 158:420–426 CrossrefGoogle Scholar

2 Golden JC, Goethe JW, Woolley SB: Complex psychotropic polypharmacy in bipolar disorder across varying mood polarities: a prospective cohort study of 2712 inpatients. J Affect Disord 2017; 221:6–10 CrossrefGoogle Scholar

3 Weinstock LM, Gaudiano BA, Epstein-Lubow G, et al.: Medication burden in bipolar disorder: a chart review of patients at psychiatric hospital admission. Psychiatry Res 2014; 216:24–30 CrossrefGoogle Scholar

4 Goldberg JF, Brooks JO 3rd, Kurita K, et al.: Depressive illness burden associated with complex polypharmacy in patients with bipolar disorder: findings from the STEP-BD. J Clin Psychiatry 2009; 70:155–162 CrossrefGoogle Scholar

5 Adli M, Whybrow PC, Grof P, et al.: Use of polypharmacy and self-reported mood in outpatients with bipolar disorder. Int J Psychiatry Clin Pract 2005; 9:251–256 CrossrefGoogle Scholar

6 Hayes J, Prah P, Nazareth I, et al.: Prescribing trends in bipolar disorder: cohort study in the United Kingdom THIN primary care database 1995–2009. PLoS One 2011; 6:e28725CrossrefGoogle Scholar

7 American Psychiatric Association: Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry 2002; 159(Suppl):1–50 Google Scholar

8 Yatham LN, Kennedy SH, Parikh SV, et al.: Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder. Bipolar Disord 2018; 20:97–170CrossrefGoogle Scholar

9 2015 Florida Best Practice Psychotherapeutic Medication Guidelines for Adults. Tampa, University of South Florida, Florida Medicaid Drug Therapy Management Program for Behavioral Health, 2015.Google Scholar

10 Suppes T, Dennehy EB, Hirschfeld RM, et al.: The Texas implementation of medication algorithms: update to the algorithms for treatment of bipolar I disorder. J Clin Psychiatry 2005; 66:870–886 CrossrefGoogle Scholar

11 Dell’Osso B, Ketter TA: Assessing efficacy/effectiveness and safety/tolerability profiles of adjunctive pramipexole in bipolar depression: acute versus long-term data. Int Clin Psychopharmacol 2013; 28:297–304 CrossrefGoogle Scholar

12 Nierenberg AA, Ostacher MJ, Calabrese JR, et al.: Treatment-resistant bipolar depression: a STEP-BD equipoise randomized effectiveness trial of antidepressant augmentation with lamotrigine, inositol, or risperidone. Am J Psychiatry 2006; 163:210–216 CrossrefGoogle Scholar

13 Bauer IE, Green C, Colpo GD, et al.: A double-blind, randomized, placebo-controlled study of aspirin and N-acetylcysteine as adjunctive treatments for bipolar depression. J Clin Psychiatry 2018; 80:18m12200. doi: 10.4088/JCP.18m12200CrossrefGoogle Scholar

14 Parmentier T, Sienaert P: The use of triiodothyronine (T3) in the treatment of bipolar depression: a review of the literature. J Affect Disord 2018; 229:410–414 CrossrefGoogle Scholar

15 Pacchiarotti I, Bond DJ, Baldessarini RJ, et al.: The International Society for Bipolar Disorders (ISBD) task force report on antidepressant use in bipolar disorders. Am J Psychiatry 2013; 170:1249–1262 CrossrefGoogle Scholar

16 Hung GC, Yang SY, Chen Y, et al.: Psychotropic polypharmacy for the treatment of bipolar disorder in Taiwan. Psychiatr Serv 2014; 65:125–128LinkGoogle Scholar

17 Sachs GS, Peters AT, Sylvia L, et al.: Polypharmacy and bipolar disorder: what’s personality got to do with it? Int J Neuropsychopharmacol 2014; 17:1053–1061 CrossrefGoogle Scholar

18 Baek JH, Ha K, Yatham LN, et al.: Pattern of pharmacotherapy by episode types for patients with bipolar disorders and its concordance with treatment guidelines. J Clin Psychopharmacol 2014; 34:577–587 CrossrefGoogle Scholar

19 Dennehy EB, Bauer MS, Perlis RH, et al.: Concordance with treatment guidelines for bipolar disorder: data from the Systematic Treatment Enhancement Program for Bipolar Disorder. Psychopharmacol Bull 2007; 40:72–84 Google Scholar

20 Perlis RH: Use of treatment guidelines in clinical decision making in bipolar disorder: a pilot survey of clinicians. Curr Med Res Opin 2007; 23:467–475 CrossrefGoogle Scholar

21 Manji HK, Quiroz JA, Payne JL, et al.: The underlying neurobiology of bipolar disorder. World Psychiatry 2003; 2:136–146 Google Scholar

22 Malhi GS, Tanious M, Das P, et al.: Potential mechanisms of action of lithium in bipolar disorder: current understanding. CNS Drugs 2013; 27:135–153 CrossrefGoogle Scholar

23 Lenox RH, McNamara RK, Watterson JM, et al.: Myristoylated alanine-rich C kinase substrate (MARCKS): a molecular target for the therapeutic action of mood stabilizers in the brain? J Clin Psychiatry 1996; 57(Suppl 13):23–33 Google Scholar

24 Morishita S, Watanabe S: The direct effect of lithium and carbamazepine on protein kinase C in rat brain. Jpn J Psychiatry Neurol 1994; 48:123–126 Google Scholar

25 Hahn CG, Gyulai L, Baldassano CF, et al.: The current understanding of lamotrigine as a mood stabilizer. J Clin Psychiatry 2004; 65:791–804 CrossrefGoogle Scholar

26 Tramontina JF, Andreazza AC, Kauer-Sant’anna M, et al.: Brain-derived neurotrophic factor serum levels before and after treatment for acute mania. Neurosci Lett 2009; 452:111–113CrossrefGoogle Scholar

27 Chang YC, Rapoport SI, Rao JS: Chronic administration of mood stabilizers upregulates BDNF and bcl-2 expression levels in rat frontal cortex. Neurochem Res 2009; 34:536–541 CrossrefGoogle Scholar

28 Li N, He X, Zhang Y, et al.: Brain-derived neurotrophic factor signalling mediates antidepressant effects of lamotrigine. Int J Neuropsychopharmacol 2011; 14:1091–1098CrossrefGoogle Scholar

29 Abelaira HM, Réus GZ, Ribeiro KF, et al.: Effects of acute and chronic treatment elicited by lamotrigine on behavior, energy metabolism, neurotrophins and signaling cascades in rats. Neurochem Int 2011; 59:1163–1174 CrossrefGoogle Scholar

30 Kusumi I, Boku S, Takahashi Y: Psychopharmacology of atypical antipsychotic drugs: from the receptor binding profile to neuroprotection and neurogenesis. Psychiatry Clin Neurosci 2015; 69:243–258 CrossrefGoogle Scholar

31 Zhou C, Zhong J, Zou B, et al.: Meta-analyses of comparative efficacy of antidepressant medications on peripheral BDNF concentration in patients with depression. PLoS One 2017; 12:e0172270. doi: 10.1371/journal.pone.0172270 CrossrefGoogle Scholar

32 Manji HK, Moore GJ, Chen G: Lithium up-regulates the cytoprotective protein Bcl-2 in the CNS in vivo: a role for neurotrophic and neuroprotective effects in manic depressive illness. J Clin Psychiatry 2000; 61(Suppl 9):82–96 Google Scholar

33 Gray NA, Zhou R, Du J, et al.: The use of mood stabilizers as plasticity enhancers in the treatment of neuropsychiatric disorders. J Clin Psychiatry 2003; 64(Suppl 5):3–17 Google Scholar

34 Leng Y, Fessler EB, Chuang DM: Neuroprotective effects of the mood stabilizer lamotrigine against glutamate excitotoxicity: roles of chromatin remodelling and Bcl-2 induction. Int J Neuropsychopharmacol 2013; 16:607–620 CrossrefGoogle Scholar

35 Bai O, Zhang H, Li XM: Antipsychotic drugs clozapine and olanzapine upregulate bcl-2 mRNA and protein in rat frontal cortex and hippocampus. Brain Res 2004; 1010:81–86CrossrefGoogle Scholar

36 Kosten TA, Galloway MP, Duman RS, et al.: Repeated unpredictable stress and antidepressants differentially regulate expression of the bcl-2 family of apoptotic genes in rat cortical, hippocampal, and limbic brain structures. Neuropsychopharmacology 2008; 33:1545–1558 CrossrefGoogle Scholar

37 Klein PS, Melton DA: A molecular mechanism for the effect of lithium on development. Proc Natl Acad Sci USA 1996; 93:8455–8459 CrossrefGoogle Scholar

38 Harwood AJ, Agam G: Search for a common mechanism of mood stabilizers. Biochem Pharmacol 2003; 66:179–189 CrossrefGoogle Scholar

39 Park SW, Phuong VT, Lee CH, et al.: Effects of antipsychotic drugs on BDNF, GSK-3β, and β-catenin expression in rats subjected to immobilization stress. Neurosci Res 2011; 71:335–340 CrossrefGoogle Scholar

40 Hui J, Zhang J, Kim H, et al.: Fluoxetine regulates neurogenesis in vitro through modulation of GSK-3β/β-catenin signaling. Int J Neuropsychopharmacol 2014; 18:pyu099. doi: 10.1093/ijnp/pyu099 CrossrefGoogle Scholar

41 Ahluwalia P, Grewaal DS, Singhal RL: Brain gabaergic and dopaminergic systems following lithium treatment and withdrawal. Prog Neuropsychopharmacol 1981; 5:527–530CrossrefGoogle Scholar

42 Löscher W: Valproate: a reappraisal of its pharmacodynamic properties and mechanisms of action. Prog Neurobiol 1999; 58:31–59 CrossrefGoogle Scholar

43 Battistin L, Varotto M, Berlese G, et al.: Effects of some anticonvulsant drugs on brain GABA level and GAD and GABA-T activities. Neurochem Res 1984; 9:225–231 CrossrefGoogle Scholar

44 Basselin M, Villacreses NE, Chen M, et al.: Chronic carbamazepine administration reduces N-methyl-D-aspartate receptor-initiated signaling via arachidonic acid in rat brain. Biol Psychiatry 2007; 62:934–943 CrossrefGoogle Scholar

45 Ghasemi M, Dehpour AR: The NMDA receptor/nitric oxide pathway: a target for the therapeutic and toxic effects of lithium. Trends Pharmacol Sci 2011; 32:420–434 CrossrefGoogle Scholar

46 Lee CY, Fu WM, Chen CC, et al.: Lamotrigine inhibits postsynaptic AMPA receptor and glutamate release in the dentate gyrus. Epilepsia 2008; 49:888–897 CrossrefGoogle Scholar

47 Dhami KS, Churchward MA, Baker GB, et al.: Fluoxetine and citalopram decrease microglial release of glutamate and D-serine to promote cortical neuronal viability following ischemic insult. Mol Cell Neurosci 2013; 56:365–374 CrossrefGoogle Scholar

48 Yanagita T, Maruta T, Uezono Y, et al.: Lithium inhibits function of voltage-dependent sodium channels and catecholamine secretion independent of glycogen synthase kinase-3 in adrenal chromaffin cells. Neuropharmacology 2007; 53:881–889 CrossrefGoogle Scholar

49 Van den Berg RJ, Kok P, Voskuyl RA: Valproate and sodium currents in cultured hippocampal neurons. Exp Brain Res 1993; 93:279–287CrossrefGoogle Scholar

50 Ambrósio AF, Silva AP, Malva JO, et al.: Carbamazepine inhibits L-type Ca2+ channels in cultured rat hippocampal neurons stimulated with glutamate receptor agonists. Neuropharmacology 1999; 38:1349–1359 CrossrefGoogle Scholar

51 Zona C, Avoli M: Lamotrigine reduces voltage-gated sodium currents in rat central neurons in culture. Epilepsia 1997; 38:522–525 CrossrefGoogle Scholar

52 Lenkey N, Karoly R, Kiss JP, et al.: The mechanism of activity-dependent sodium channel inhibition by the antidepressants fluoxetine and desipramine. Mol Pharmacol 2006; 70:2052–2063 CrossrefGoogle Scholar

53 Massot O, Rousselle JC, Fillion MP, et al.: 5-HT1B receptors: a novel target for lithium. Possible involvement in mood disorders. Neuropsychopharmacology 1999; 21:530–541CrossrefGoogle Scholar

54 Ramadan E, Basselin M, Taha AY, et al.: Chronic valproate treatment blocks D2-like receptor-mediated brain signaling via arachidonic acid in rats. Neuropharmacology 2011; 61:1256–1264 CrossrefGoogle Scholar

55 Basselin M, Chang L, Chen M, et al.: Chronic carbamazepine administration attenuates dopamine D2-like receptor-initiated signaling via arachidonic acid in rat brain. Neurochem Res 2008; 33:1373–1383 CrossrefGoogle Scholar

56 Zarate CA Jr, Singh JB, Carlson PJ, et al.: Efficacy of a protein kinase C inhibitor (tamoxifen) in the treatment of acute mania: a pilot study. Bipolar Disord 2007; 9:561–570CrossrefGoogle Scholar

57 Sarris J, Mischoulon D, Schweitzer I: Omega-3 for bipolar disorder: meta-analyses of use in mania and bipolar depression. J Clin Psychiatry 2012; 73:81–86 CrossrefGoogle Scholar

58 Cipriani A, Hawton K, Stockton S, et al.: Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ 2013; 346:f3646. doi: 10.1136/bmj.f3646 CrossrefGoogle Scholar

59 Chengappa KN, Bowie CR, Schlicht PJ, et al.: Randomized placebo-controlled adjunctive study of an extract of Withania somnifera for cognitive dysfunction in bipolar disorder. J Clin Psychiatry 2013; 74:1076–1083 CrossrefGoogle Scholar

60 Yatham LN, Mackala S, Basivireddy J, et al.: Lurasidone versus treatment as usual for cognitive impairment in euthymic patients with bipolar I disorder: a randomised, open-label, pilot study. Lancet Psychiatry 2017; 4:208–217 CrossrefGoogle Scholar

61 Swann AC: Treatment of aggression in patients with bipolar disorder. J Clin Psychiatry 1999; 60(Suppl 15):25–28 Google Scholar

62 Goldberg JF: Personalized pharmacotherapy for bipolar disorder: how to tailor findings from randomized trials to individual patient-level outcomes. Focus 2019; 17:206–217Google Scholar

63 Bartoli F, Crocamo C, Clerici M, et al.: Allopurinol as add-on treatment for mania symptoms in bipolar disorder: systematic review and meta-analysis of randomised controlled trials. Br J Psychiatry 2017; 210:10–15 CrossrefGoogle Scholar

64 Vieta E, T’joen C, McQuade RD, et al.: Efficacy of adjunctive aripiprazole to either valproate or lithium in bipolar mania patients partially nonresponsive to valproate/lithium monotherapy: a placebo-controlled study. Am J Psychiatry 2008; 165:1316–1325 CrossrefGoogle Scholar

65 Szegedi A, Calabrese JR, Stet L, et al.: Asenapine as adjunctive treatment for acute mania associated with bipolar disorder: results of a 12-week core study and 40-week extension. J Clin Psychopharmacol 2012; 32:46–55 CrossrefGoogle Scholar

66 Arabzadeh S, Ameli N, Zeinoddini A, et al.: Celecoxib adjunctive therapy for acute bipolar mania: a randomized, double-blind, placebo-controlled trial. Bipolar Disord 2015; 17:606–614 CrossrefGoogle Scholar

67 Tohen M, Chengappa KN, Suppes T, et al.: Efficacy of olanzapine in combination with valproate or lithium in the treatment of mania in patients partially nonresponsive to valproate or lithium monotherapy. Arch Gen Psychiatry 2002; 59:62–69 CrossrefGoogle Scholar

68 Houston JP, Tohen M, Degenhardt EK, et al.: Olanzapine-divalproex combination versus divalproex monotherapy in the treatment of bipolar mixed episodes: a double-blind, placebo-controlled study. J Clin Psychiatry 2009; 70:1540–1547 CrossrefGoogle Scholar

69 Sachs GS, Grossman F, Ghaemi SN, et al.: Combination of a mood stabilizer with risperidone or haloperidol for treatment of acute mania: a double-blind, placebo-controlled comparison of efficacy and safety. Am J Psychiatry 2002; 159:1146–1154 CrossrefGoogle Scholar

70 Sachs G, Chengappa KN, Suppes T, et al.: Quetiapine with lithium or divalproex for the treatment of bipolar mania: a randomized, double-blind, placebo-controlled study. Bipolar Disord 2004; 6:213–223 CrossrefGoogle Scholar

71 Bourin MS, Severus E, Schronen JP, et al.: Lithium as add-on to quetiapine XR in adult patients with acute mania: a 6-week, multicenter, double-blind, randomized, placebo-controlled study. Int J Bipolar Disord 2014; 2:14. doi: 10.1186/s40345-014-0014-9 CrossrefGoogle Scholar

72 Amrollahi Z, Rezaei F, Salehi B, et al.: Double-blind, randomized, placebo-controlled 6-week study on the efficacy and safety of the tamoxifen adjunctive to lithium in acute bipolar mania. J Affect Disord 2011; 129:327–331 CrossrefGoogle Scholar

73 Tohen M, Bowden CL, Smulevich AB, et al.: Olanzapine plus carbamazepine v carbamazepine alone in treating manic episodes. Br J Psychiatry 2008; 192:135–143CrossrefGoogle Scholar

74 Berwaerts J, Lane R, Nuamah IF, et al.: Paliperidone extended-release as adjunctive therapy to lithium or valproate in the treatment of acute mania: a randomized, placebo-controlled study. J Affect Disord 2011; 129:252–260 CrossrefGoogle Scholar

75 Roy Chengappa KN, Schwarzman LK, Hulihan JF, et al.: Adjunctive topiramate therapy in patients receiving a mood stabilizer for bipolar I disorder: a randomized, placebo-controlled trial. J Clin Psychiatry 2006; 67:1698–1706 CrossrefGoogle Scholar

76 Sachs GS, Vanderburg DG, Karayal ON, et al.: Adjunctive oral ziprasidone in patients with acute mania treated with lithium or divalproex, part 1: results of a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry 2012; 73:1412–1419 CrossrefGoogle Scholar

77 van der Loos ML, Mulder PG, Hartong EG, et al.: Efficacy and safety of lamotrigine as add-on treatment to lithium in bipolar depression: a multicenter, double-blind, placebo-controlled trial. J Clin Psychiatry 2009; 70:223–231 CrossrefGoogle Scholar

78 Geddes JR, Gardiner A, Rendell J, et al.: Comparative evaluation of quetiapine plus lamotrigine combination versus quetiapine monotherapy (and folic acid versus placebo) in bipolar depression (CEQUEL): a 2 × 2 factorial randomised trial. Lancet Psychiatry 2016; 3:31–39 CrossrefGoogle Scholar

79 Loebel A, Cucchiaro J, Silva R, et al.: Lurasidone as adjunctive therapy with lithium or valproate for the treatment of bipolar I depression: a randomized, double-blind, placebo-controlled study. Am J Psychiatry 2014; 171:169–177 CrossrefGoogle Scholar

80 Sachs GS, Ice KS, Chappell PB, et al.: Efficacy and safety of adjunctive oral ziprasidone for acute treatment of depression in patients with bipolar I disorder: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry 2011; 72:1413–1422 CrossrefGoogle Scholar

81 Marcus R, Khan A, Rollin L, et al.: Efficacy of aripiprazole adjunctive to lithium or valproate in the long-term treatment of patients with bipolar I disorder with an inadequate response to lithium or valproate monotherapy: a multicenter, double-blind, randomized study. Bipolar Disord 2011; 13:133–144 CrossrefGoogle Scholar

82 Geddes JR, Goodwin GM, Rendell J, et al.: Lithium plus valproate combination therapy versus monotherapy for relapse prevention in bipolar I disorder (BALANCE): a randomised open-label trial. Lancet 2010; 375:385–395 CrossrefGoogle Scholar

83 Yatham LN, Fountoulakis KN, Rahman Z, et al.: Efficacy of aripiprazole versus placebo as adjuncts to lithium or valproate in relapse prevention of manic or mixed episodes in bipolar I patients stratified by index manic or mixed episode. J Affect Disord 2013; 147:365–372CrossrefGoogle Scholar

84 Tohen M, Chengappa KN, Suppes T, et al.: Relapse prevention in bipolar I disorder: 18-month comparison of olanzapine plus mood stabiliser v mood stabiliser alone. Br J Psychiatry 2004; 184:337–345 CrossrefGoogle Scholar

85 Macfadden W, Alphs L, Haskins JT, et al.: A randomized, double-blind, placebo-controlled study of maintenance treatment with adjunctive risperidone long-acting therapy in patients with bipolar I disorder who relapse frequently. Bipolar Disord 2009; 11:827–839CrossrefGoogle Scholar

86 Bowden CL, Vieta E, Ice KS, et al.: Ziprasidone plus a mood stabilizer in subjects with bipolar I disorder: a 6-month, randomized, placebo-controlled, double-blind trial. J Clin Psychiatry 2010; 71:130–137 CrossrefGoogle Scholar

87 Calabrese JR, Pikalov A, Streicher C, et al.: Lurasidone in combination with lithium or valproate for the maintenance treatment of bipolar I disorder. Eur Neuropsychopharmacol 2017; 27:865–876 CrossrefGoogle Scholar

88 Vieta E, Cruz N, García-Campayo J, et al.: A double-blind, randomized, placebo-controlled prophylaxis trial of oxcarbazepine as adjunctive treatment to lithium in the long-term treatment of bipolar I and II disorder. Int J Neuropsychopharmacol 2008; 11:445–452CrossrefGoogle Scholar

89 Peselow ED, Naghdechi L, Pizano D, et al.: Polypharmacy in maintenance of bipolar disorder. Clin Neuropharmacol 2016; 39:132–134 CrossrefGoogle Scholar

90 Fornaro M, De Berardis D, Koshy AS, et al.: Prevalence and clinical features associated with bipolar disorder polypharmacy: a systematic review. Neuropsychiatr Dis Treat 2016; 12:719–735 CrossrefGoogle Scholar

91 Reischies FM, Hartikainen J, Berghöfer AM: Initial triple therapy of acute mania, adding lithium and valproate to neuroleptics. Pharmacopsychiatry 2002; 35:244–246CrossrefGoogle Scholar

92 Nierenberg AA, Friedman ES, Bowden CL, et al.: Lithium treatment moderate-dose use study (LiTMUS) for bipolar disorder: a randomized comparative effectiveness trial of optimized personalized treatment with and without lithium. Am J Psychiatry 2013; 170:102–110CrossrefGoogle Scholar

93 Perlis RH, Ostacher MJ, Miklowitz DJ, et al.: Benzodiazepine use and risk of recurrence in bipolar disorder: a STEP-BD report. J Clin Psychiatry 2010; 71:194–200 CrossrefGoogle Scholar

94 Goodwin FK, Fireman B, Simon GE, et al.: Suicide risk in bipolar disorder during treatment with lithium and divalproex. JAMA 2003; 290:1467–1473 CrossrefGoogle Scholar

95 Schneck CD, Miklowitz DJ, Miyahara S, et al.: The prospective course of rapid-cycling bipolar disorder: findings from the STEP-BD. Am J Psychiatry 2008; 165:370–377, quiz 410CrossrefGoogle Scholar

96 Perlis RH, Welge JA, Vornik LA, et al.: Atypical antipsychotics in the treatment of mania: a meta-analysis of randomized, placebo-controlled trials. J Clin Psychiatry 2006; 67:509–516 CrossrefGoogle Scholar

97 Sachs GS, Nierenberg AA, Calabrese JR, et al.: Effectiveness of adjunctive antidepressant treatment for bipolar depression. N Engl J Med 2007; 356:1711–1722CrossrefGoogle Scholar

98 Yoon W, Shon S-H, Hong Y, et al.: Antidepressant prescription patterns in bipolar disorder: a nationwide, register-based study in Korea. J Korean Med Sci 2018; 33:e290CrossrefGoogle Scholar

99 McElroy SL, Weisler RH, Chang W, et al.: A double-blind, placebo-controlled study of quetiapine and paroxetine as monotherapy in adults with bipolar depression (EMBOLDEN II). J Clin Psychiatry 2010; 71:163–174 CrossrefGoogle Scholar

100 Procyshyn RM, Honer WG, Wu TK, et al.: Persistent antipsychotic polypharmacy and excessive dosing in the community psychiatric treatment setting: a review of medication profiles in 435 Canadian outpatients. J Clin Psychiatry 2010; 71:566–573 CrossrefGoogle Scholar

101 Brooks JO 3rd, Goldberg JF, Ketter TA, et al.: Safety and tolerability associated with second-generation antipsychotic polytherapy in bipolar disorder: findings from the Systematic Treatment Enhancement Program for Bipolar Disorder. J Clin Psychiatry 2011; 72:240–247CrossrefGoogle Scholar

102 Kemp DE, Gao K, Fein EB, et al.: Lamotrigine as add-on treatment to lithium and divalproex: lessons learned from a double-blind, placebo-controlled trial in rapid-cycling bipolar disorder. Bipolar Disord 2012; 14:780–789CrossrefGoogle Scholar

103 Altshuler LL, Post RM, Hellemann G, et al.: Impact of antidepressant continuation after acute positive or partial treatment response for bipolar depression: a blinded, randomized study. J Clin Psychiatry 2009; 70:450–457 CrossrefGoogle Scholar

104 Ghaemi SN, Ostacher MM, El-Mallakh RS, et al.: Antidepressant discontinuation in bipolar depression: a Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD) randomized clinical trial of long-term effectiveness and safety. J Clin Psychiatry 2010; 71:372–380 CrossrefGoogle Scholar

105 Calabrese JR, Shelton MD, Rapport DJ, et al.: A 20-month, double-blind, maintenance trial of lithium versus divalproex in rapid-cycling bipolar disorder. Am J Psychiatry 2005; 162:2152–2161 CrossrefGoogle Scholar

106 Yatham LN, Beaulieu S, Schaffer A, et al.: Optimal duration of risperidone or olanzapine adjunctive therapy to mood stabilizer following remission of a manic episode: a CANMAT randomized double-blind trial. Mol Psychiatry 2016; 21:1050–1056 CrossrefGoogle Scholar

107 Rush AJ, Kraemer HC, Sackeim HA, et al.: Report by the ACNP Task Force on response and remission in major depressive disorder. Neuropsychopharmacology 2006; 31:1841–1853 CrossrefGoogle Scholar

108 Frank E, Prien RF, Jarrett RB, et al.: Conceptualization and rationale for consensus definitions of terms in major depressive disorder: remission, recovery, relapse, and recurrence. Arch Gen Psychiatry 1991; 48:851–855 CrossrefGoogle Scholar

109 Amerio A, Odone A, Marchesi C, et al.: Treatment of comorbid bipolar disorder and obsessive-compulsive disorder: a systematic review. J Affect Disord 2014; 166:258–263CrossrefGoogle Scholar

110 Pettinati HM, O’Brien CP, Dundon WD: Current status of co-occurring mood and substance use disorders: a new therapeutic target. Am J Psychiatry 2013; 170:23–30CrossrefGoogle Scholar