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.

×
Clinical SynthesisFull Access

Developments in Psychopharmacology for Major Depressive Disorder

Abstract

Abstract:

After years of emphasizing the same monoamine-based neurotransmitter mechanisms for treating depression, the antidepressant medication pipeline is broadening its reach. While serotonin, norepinephrine, and dopamine remain important treatment targets, researchers are working on compounds targeting their receptors in novel ways, as well as different combinations of monoaminergic actions that they hope will lead to improved efficacy and/or decreased side effects. At the same time, other researchers are focusing on completely different avenues of drug development, such as medications that target glutamate or acetylcholine neurotransmission, opioid receptors, or hormonal systems such as vasopressin and melatonin. Advances in pharmacogenetics also offer the possibility of targeting medications more specifically to individuals depending on their likelihood of response or side effects. This article outlines some promising directions for antidepressant drug development and discusses examples for each that have been undergoing testing.

Introduction

The last major development in the pharmacotherapy of major depressive disorder (MDD) was indeed revolutionary. The approval in the 1980s of selective serotonin reuptake inhibitors (SSRIs) for depression, which appeared to match the effectiveness of older antidepressants without as much of a side effect burden, may have been one of the main reasons that the use of antidepressants tripled in the following years, with SSRIs accounting for more than half of all antidepressant medication prescriptions by 2006 (1).

In the two decades since, much of the action in antidepressant development has revolved around expanding upon the currently known antidepressant medication classes by identifying other selective reuptake inhibitors, developing other medications that mimic the action of already-known antidepressants, or testing medications approved for other indications as depression augmentation or monotherapy agents. Still, these new medications have not yielded a substantial advance in the rate of treatment response for depression, with only half of patients in the STAR*D trial achieving symptomatic remission within the first two treatment stages (2, 3).

More recently, however, advances in knowledge about the pathophysiology of depression have opened up additional potential directions for drug development. For example, discoveries about the role of other neurotransmitters in MDD have led to the development of potential antidepressants tapping the glutamatergic and nicotinic neurotransmitter systems (Table 1). Other avenues for drug development have targeted hormonal systems such as the hypothalamic-pituitary-adrenal axis, known to be involved in the body’s response to stress, and the circadian system, which might affect depression via its effects on sleep. At the same time, the increasing ease of gene sequencing and the growing body of research on genetic polymorphisms associated with antidepressant response means that it may soon be possible not only to profile an individual patient’s likelihood of experiencing side effects with a given medication but also to choose the most promising pharmacological treatment. This article will outline some of the mechanisms for antidepressant action that are attracting the most attention from drug developers and that could represent the next wave of antidepressants.

Table 1. Antidepressant Medications in the Pipeline
Medication Class and Examples in DevelopmentMechanismEvidenceTesting Status
Monoamine-based
 Vortioxetine (Lu AA21004)
5HT3a receptor antagonist, 5HT7 receptor antagonist, 5HT1B partial agonist, 5HT1A agonist, and 5HT transporter inhibitor
Reduced symptoms of depression in Phase III trials; may have fewer cognitive side effects; was effective at preventing relapse in an open-label trial; did not separate from placebo in a “failed” trial
Manufacturer filed for FDA approval in late 2012
 Levomilnacipran
Serotonin/norepinephrine reuptake inhibitor
Approved in Europe for depression; showed efficacy for depression in Phase III trials
Recently finished Phase III testing; manufacturer filed for FDA approval in late 2012
 Edivoxetine (LY 2216684)
Selective norepinephrine reuptake inhibitor
Study showed greater symptomatic improvement and better response/remission rates than placebo
In Phase III testing as adjunctive treatment for partial responders and as relapse prevention
 Brexpiprazole (OPC-34712)
D2 partial agonist, 5HT1a partial agonist, 5HT2 antagonist
Showed positive results as augmentation treatment in Phase II trials
In Phase III testing as adjunctive treatment
 Lisdexamfetamine
Prodrug of dextroamphetamine, which blocks reuptake of norepinephrine and dopamine and increases their release
Showed positive results as augmentation treatment for depression in full or partial remission with residual executive dysfunction
In Phase III testing for nonresponders to depression treatment and for those with residual depressive symptoms
 Amitifadine (EB-1010)
Triple reuptake inhibitor
Showed positive results on several depression outcome measures in Phase II trial
In Phase IIb/IIIa testing
 TriRima (CX 157)
Reversible MAO-A inhibitor
Completed safety and tolerability (Phase I) testing
Phase II trials were recently completed
Glutamatergic



 Ketamine
NMDA receptor antagonist
Small studies have shown rapid antidepressant effects after IV administration
Multiple Phase II through IV trials in progress
 Riluzole
NMDA receptor antagonist
Has demonstrated antidepressant-like effects in several small studies
In Phase II testing for MDD and BPAD
 Traxoprodil (CP 101 606)
NMDA receptor antagonist
Has shown antidepressant effects in nonresponders
Unknown
 Amantadine
NMDA receptor antagonist
Showed positive effects in study of augmentation in imipramine nonresponders
Unknown
 Dextromethorphan
NMDA receptor antagonist; serotonin transporter inhibitor; mu opioid receptor potentiation

In testing in Asia as an adjunctive treatment for BPAD
 Memantine
NMDA receptor antagonist
Positive results in small open-label trial and study of memantine plus escitalopram for depression with alcohol dependence. No effect of monotherapy in a double-blind, randomized controlled trial
Multiple Phase 4 trials recently completed
 Farampator (CX-691/Org 24448)
AMPA receptor modulator

Trials terminated due to side effect concerns
Nicotinic



 Mecamylamine (TC 5214)
Nicotinic receptor antagonist
Did not meet primary endpoints in Phase III trials
Pulled from development
 Varenicline
alpha4beta2 partial agonist, alpha7 full agonist
Positive effect on mood in depressed smokers in open-label study
Unknown
Opioid-based



 Buprenorphine
mu opioid receptor agonist, kappa opioid receptor antagonist
Antidepressant effects found as long as three decades ago
In Phase II and III testing
 Buprenorphine + samidorphan (ALKS 33) (ALKS 54651)
Buprenorphine + mu opioid receptor antagonist
Evidence of decreased symptoms in treatment-resistant patients after 1 week
In Phase II testing
HPA axis: SSR149415
Vasopressin V1b receptor antagonist
Mixed results on efficacy for depression in Phase IIb trials
Recently completed some Phase IIb trials
Melatonergic: agomelatinemelatonergic receptor agonist (MT1/MT2); 5HT2c antagonistApproved in Europe; poor results from Phase III trials in U.S.Pulled from pharmaceutical development in the U.S.
Table 1. Antidepressant Medications in the Pipeline
Enlarge table

Monoamine-Based Antidepressants

All of the antidepressant medications prescribed today capitalize on some aspect of monoamine neurotransmission, whether serotonin, norepinephrine, or dopamine. The most common are the serotonergic agents, which includes selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs). These agents all inhibit the serotonin transporter, with varying degrees of agonism or antagonism at specific receptor subtypes; some also inhibit the norepinephrine transporter.

One of the newer variations on this class is vilazodone, which was approved by the FDA for depression in 2011. This medication both inhibits the serotonin transporter and acts as a partial agonist at 5HT1A receptors; thus, it is sometimes also called a SPARI (serotonin partial agonist reuptake inhibitor) (4, 5). Whether this additional action of 5HT1A partial agonism boosts the efficacy of serotonin inhibition or mitigates the side effects of 5HT1A partial agonism remains unknown, but this combination of mechanisms may result in lower rates of sexual dysfunction and weight gain. (4)

Another multimodal serotonergic medication in development is vortioxetine, a 5HT3a receptor antagonist, 5HT7 receptor antagonist, 5HT1B partial agonist, 5HT1A agonist, and 5HT transporter inhibitor (6). Vortioxetine has been found to reduce symptoms of depression in several Phase III trials (79) and has been associated with a more favorable cognitive effect profile (9); it has also been found to be effective in preventing relapse of depression in an open-label trial (10). In a “failed study,” while vortioxetine did not separate from placebo, neither did the other active treatment, duloxetine (11).

Among medications that combine noradrenergic and serotonergic activity is the serotonin-norepinephrine reuptake inhibitor levomilnacipran. This compound, which is an enantiomer of the fibromyalgia medication milnacipran, has received approval as an antidepressant in Europe. The manufacturer recently presented evidence of greater improvement in depressive symptoms with levomilnacipran compared with placebo from two Phase III trials (12). Another possibility is the selective norepinephrine reuptake inhibitor edivoxetine. While it primarily has been studied for attention deficit hyperactivity disorder, it is also being considered as a potential treatment for major depression; a recent study showed greater symptomatic improvement and higher response and remission rates with edivoxetine compared with placebo in patients with major depression (13).

In addition, there is interest in developing antidepressants with dopaminergic activity, perhaps in combination with serotonergic and noradrenergic actions. The idea is that targeting dopaminergic neurotransmission will enhance overall efficacy; reduce anhedonia, apathy, and cognitive impairment; and minimize residual fatigue and sleepiness (as suggested by the dopamine reuptake inhibitor modafinil augmentation studies of SSRIs (14)). In addition, given that dopaminergic medications have been used to treat SSRI-induced sexual dysfunction (15, 16), there is hope that dopaminergic antidepressants may cause less sexual dysfunction than SSRIs and SNRIs.

One dopaminergic compound currently under study is the dopamine (D2) and 5HT1a partial agonist/5HT2 antagonist brexpiprazole, which is structurally related to the atypical antipsychotic (and depression augmentation agent) aripiprazole. Brexpiprazole showed promise in a Phase II trial as an augmentation treatment for MDD (17). Another potential antidepressant with dopaminergic activity is lisdexamfetamine. This ADHD medication is a prodrug of the stimulant dextroamphetamine, which blocks presynaptic reuptake of norepinephrine and dopamine and increases their release. Shire has reported positive Phase II results from a trial using lisdexamfetamine as an augmentation treatment for major depression in full or partial remission but with continued executive dysfunction (18, 19).

Because of the role of dopamine in the brain’s reward circuit and addictive behaviors, there has been some concern about the risk of abuse with dopaminergic medications, perhaps limiting enthusiasm for studying dopaminergic antidepressants. But there have been animal studies that have found no evidence of abuse liability with certain dopaminergic medications. For example, in a study of rats given a medication with dopaminergic as well as serotonergic and noradrenergic activity, the rats did not exhibit self-administration of the medication (a marker of abuse liability) (20); another study found that administration of a similar medication was associated with decreased ethanol consumption in alcohol-preferring rats (21).

Triple uptake inhibitors (TUIs) are attracting particular interest because of the promise of modulating three monoamine neurotransmitter systems at once. The hope is that these “all-in-one” compounds would have the synergistic effects of triple inhibition and lead to more robust antidepressant effects without requiring high occupancy of the serotonin transporter, thus minimizing many of the side effects seen with SSRIs. The TUI amitifadine has demonstrated positive results on several depression outcome measures, including an anhedonia measure, without significant weight gain or sexual dysfunction in a small phase 2 trial (22).

Finally, in a parallel line of research, pharmaceutical companies are pursuing variations on monoamine oxidase inhibitors, which decrease the breakdown of serotonin, norepinephrine, and dopamine and thus increase their levels. One of the major limitations of the early monoamine oxidase inhibitors was that they affected both the MAO-A and MAO-B forms of the enzyme and are generally irreversible, making it particularly dangerous to ingest dietary tyramine. Without either form of the enzyme available to break down tyramine for 2 weeks (the length of time it takes to regenerate the enzyme), patients were at high risk for hypertensive crisis after consuming foods containing tyramine. This was the impetus behind the development of reversible MAOIs, such as moclobemide and the newer CX 157 (TriRima). TriRima is thought to be more potent than moclobemide and also to be a powerful inhibitor of MAO-A (23), which is primarily responsible for breaking down serotonin, norepinephrine, melatonin and epinephrine (dopamine is equally metabolized by MAO-A and MAO-B).

Other Neurotransmitter-Based Treatments

Breaking away from the traditional focus on serotonin, norepinephrine, and dopamine, a number of researchers are investigating drugs that work on other neurotransmitters that have been implicated in depression, including glutamatergic medications and compounds that target nicotinic receptors.

Glutamatergic drugs

Glutamate is one of the main excitatory neurotransmitters in the CNS, exerting its effects primarily via one of several receptor subtypes: N-methyl-d-aspartate (NMDA) receptors, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors, kainate receptors, and type I, II, III metabotropic glutamate receptors. Because of evidence of glutamate dysfunction in major depression and of neuroprotective properties of NMDA receptor antagonists (24), there is speculation that they may be effective antidepressants.

Of the NMDA antagonists currently under study for depression, the anesthetic ketamine may have the most buzz. A number of small studies have found rapid and in some cases sustained antidepressant effects after ketamine injections, for a total sample size of 163 patients (25). A number of trials are attempting to confirm these effects in larger, more rigorous trials and to examine how the effects can be sustained. The main drawback of ketamine at this point is that it must be administered intravenously in a hospital setting.

Other NMDA antagonists receiving attention for depression include riluzole, amantadine, traxoprodil, and dextromethorphan. Riluzole, a medication that noncompetitively inhibits NMDA receptors as well as the release of glutamic acid and that was originally approved for amyotrophic lateral sclerosis, has demonstrated antidepressant-like effects in several small studies (26, 27). Amantadine, which possesses dopaminergic activity in addition to antagonizing NMDA receptors, showed positive effects in a double-blind augmentation study for depressed imipramine nonresponders (28). Another NMDA receptor antagonist, traxoprodil, has demonstrated antidepressant effects in patients who have not responded to SSRI treatment (29). Finally, the cold remedy dextromethorphan is attracting attention as well because it has some actions similar to ketamine, such as NMDA antagonism, serotonin transporter inhibition, and mu (opiate) receptor potentiation. As Nuedexta (dextromethorphan/quinidine), it has already received FDA approval for the treatment of pseudobulbar affect, and some researchers speculate that it could find utility as a conventional antidepressant, rapid-acting antidepressant, or treatment for treatment-refractory depression (30).

However, results with memantine have been somewhat disappointing. Despite promising results from a small open-label study with depressed patients (31) and a double-blind randomized controlled trial of memantine plus escitalopram in major depression comorbid with alcohol dependence (32), Zarate et al. found no effect in a double-blind, randomized, controlled study of memantine for depression monotherapy (33).

Additionally, several concerns about glutamatergic medications have reduced enthusiasm for them, most notably questions about psychedelic effects. Some of these agents possess hallucinogenic properties and may induce psychosis-like symptoms in subjects who have no previous history of psychosis (34, 35).

The potential role of glutamatergic agents that act on AMPA, kainate, or metabotropic glutamate receptors in treating psychiatric disorders is not as well studied, although there is considerable interest in these compounds as well. Given the beneficial effects of glutamatergic agents such as AMPA receptor modulators on cognition, there was hope that agents could be effective in the treatment of cognitive dysfunction in depression, or in the treatment of MDD presenting with prominent cognitive dysfunction (3637). Unfortunately, one of the first trials of an AMPA receptor modulator, farampator (CX-691/Org 24448), was terminated early due to concerns about adverse effects observed in other studies (38).

Nicotinic Medications

Several lines of evidence have led to interest in nicotinic medications for depression. Several antidepressants, such as the tricyclic antidepressant imipramine, have antagonist activity at the nicotinic receptor, and there is evidence of nicotinic receptor dysfunction in depression (39). The link between nicotine dependence and depression is well established, with higher rates of smoking in patients with major depressive disorder (40) and vice versa (41). In addition, the nicotinic receptor may be involved in memory, cognition and behavioral reinforcement/addiction. For example, the alpha4beta2 subtype of nicotinic receptors has been reported to play a role in acetylcholine-mediated dopamine release in areas of the brain that are involved in behavioral reinforcement and addiction—specifically, the striatum, ventral tegmental area, and nucleus accumbens (4244). The alpha7 receptors have been linked to learning and memory in preclinical studies (45).

An early open-label study of the alpha4beta2 partial agonist and alpha7 full agonist varenicline, already approved for smoking cessation, found an improvement in mood when depressed smokers started taking the medication to augment their existing psychotropic treatment (46). However, despite promising Phase II studies, the noncompetitive nicotinic receptor antagonist mecamylamine did not meet primary endpoints in Phase III trials and has been pulled from development (47).

Opioid-Based Treatments

With evidence accumulating for the role of the endogenous opioid system in regulating mood, attention is turning to opioid-based medications for their potential role in depression treatment. There are three types of opioid receptors—delta, kappa, and mu—and all three have been linked variously to monoaminergic activity (4850), behaviors in stressful situations such as the forced swim test (51), and antidepressant response (52, 53). In humans, depressed individuals and healthy comparison subjects exhibited different patterns of mu opioid receptor availability in emotionally neutral and sad states (54), supporting a role for opioids in mood. In addition, polymorphisms in the mu opioid receptor gene were associated with citalopram response in STAR*D (55).

While much remains to be understood about the relationship between the opioid system and mood disorders—it may play a role in schizophrenia and impulse control disorders as well as mood disorders and addiction (56, 57)—researchers are already proceeding with studies of opioid-based medications for depression. As early as 1982, some studies were finding antidepressant effects of buprenorphine, a mu opioid receptor agonist and kappa opioid receptor antagonist (58), and it is currently being tested as monotherapy or augmentation for depression. Another drug in Phase II testing is ALKS 5461, which combines buprenorphine and the mu antagonist samidorphan; it showed improvements in depression symptoms in treatment-resistant patients after one week in Phase II trials (59).

Hormone-Based Treatments

Unsurprisingly given their powerful and far-ranging effects on the body, a number of hormones can influence psychiatric symptoms. Hypothyroidism, for example, often causes depressive symptoms, while corticosteroids can induce mood lability, mood disturbance, or even psychosis. Thus a number of antidepressant development approaches are focusing on hormonal pathways.

Treatments That Act on the Hypothalamic-Pituitary-Adrenal Axis

Disturbances in the hypothalamic-pituitary-adrenal (HPA) axis are a well-established feature of depression. In particular, basic and clinical studies have found evidence of increased secretion of the hypothalamic neuropeptides vasopressin and corticotrophin-releasing factor (CRF) in depression and anxiety, leading to interest in treatments addressing these mechanisms. Vasopressin is released from the pituitary during stress and potentiates the body’s release of adrenocorticotropin in response to CRF. Animal studies in rats and birds first suggested that vasopressin may be essential to the body’s ability to adapt to stress (6062). Because the V1b receptor is responsible for the pituitary response to vasopressin, it has become a target for drug development in depression and anxiety. The nonpeptide V1b receptor antagonist SSR149415 showed mixed results on efficacy for depression in several phase IIb studies for efficacy and tolerability, though the studies did not yield significant results for efficacy as treatment for generalized anxiety disorder. The authors concluded that “the antidepressant potential of SSR149415 needs to be further evaluated” (62).

Melatonergic Medications

The circadian system plays an important role in mood disorders. Sleep disturbances are a core feature of both major depression and bipolar disorder; seasonal affective disorder is tied to the length of daylight in different seasons; and there is even some evidence that sleep deprivation can cause mood symptoms. Thus, the hormone melatonin, which is a serotonin precursor that maintains the body’s internal clock and may also play a neuroprotective role, has drawn significant attention for its potential in depression treatment.

One melatonergic drug under study for depression as well as anxiety and obsessive-compulsive disorder is agomelatine, which is a melatonergic receptor agonist (MT1/MT2) as well as a 5HT2c antagonist. While early research concentrated on its utility for sleep disorders, the first dose-finding trials for its use in major depression were published in 2002 (63), and it gained approval for treating depression in Europe in 2009. Because it has minimal serotonergic action other than 5HT2c antagonism, agomelatine is thought to be less likely to cause gastrointestinal, sexual, and metabolic side effects; its main side effect appears to be dizziness (64). Discontinuation syndrome may also be less frequent with this medication (65). However, in some studies, approximately 1%–4.5% of patients taking the medication developed a transient and reversible elevation in hepatic transaminases, sometimes only at higher doses (6668). Due to these concerns as well as poor Phase III trial results in the U.S (69), Novartis discontinued development of agomelatine in the U.S. in October 2011, although clinical trials by other groups are continuing.

Pharmacogenetics

The hot word in medicine is pharmacogenetics, and the field of antidepressant research is no exception. The idea is that by knowing a person’s genetic profile with regard to variants that affect response to medication treatment, it would be possible to personalize his or her treatment to be more effective while reducing side effects.

The targets attracting the most interest in depression research are genes coding for neurotransmitter or drug transporters or receptors and genes coding for enzymes that break down those neurotransmitters or drugs. Some of the earliest studies focused on polymorphisms in the promoter region of the serotonin transporter gene, such as 5-HTTLPR, which was linked to affective disorders more than 15 years ago (70) and has been implicated in response to antidepressants and antidepressant-induced mania (71). More recently, it has become possible to screen the genome with hundreds or thousands of single-nucleotide polymorphisms (SNPs) in genome-wide association studies (GWAS) to identify other potential targets. For example, STAR*D collected DNA from approximately 2,000 study participants with nonpsychotic major depressive disorder; this data were used to identify new genes associated with citalopram response and resistance, evaluate the role of genes previously thought to be associated with some aspect of depression treatment, and examine markers associated with side effects such as suicidal ideation (72). Another GWAS, the Genome-based Therapeutic Drugs for Depression (GENDEP) study of 760 adults with moderate-to-severe depression, focused on serotonergic, noradrenergic, neurotrophic, and glutamatergic mechanisms (73).

Perhaps not unexpectedly given the limited sample sizes and the large number of polymorphisms being evaluated, these large studies have sometimes produced contradictory results. For example, the STAR*D data did not confirm earlier suggestions of a significant relationship between treatment response and polymorphisms in the BDNF gene (74) and provided a complicated picture about the role of the serotonin transporter gene and its promoter region (reported variously to have no relationship to citalopram response, a relationship to tolerability but not treatment response, and a relationship to citalopram response in non-Hispanic whites in STAR*D) (72). The Munich Antidepressant Response Signature (MARS) study of 387 adults with depressive disorders (including about 10% with bipolar depression) failed to replicate the STAR*D findings, although a predictive model including effects of and interactions between three genes involved in serotonergic, glutamatergic, and HPA activity accounted for 13% of the variance in remission after 5 weeks, consistent with the STAR*D findings (75). Part of the difficulty is that the magnitude of the relationship between individual variants and treatment response has generally been modest (76). Another problem is that multiple genetic variations may influence response to an individual drug, making it even more difficult to predict response with the knowledge currently available.

Another major question with pharmacogenetics and personalized medicine is cost effectiveness. Perlis et al. calculated that testing 40-year-old men with major depression for response to SSRIs and using bupropion in likely nonresponders would cost $93,520 per additional quality-adjusted life-year (QALY) compared with treating all patients with an SSRI first and switching in the case of nonresponse (77); by contrast, the average cost per QALY for dialysis compared with the next cheaper options is $129,090 (78). An analysis of the economics of using 5-HTTLPR genotyping in Europe concluded that testing would be cost-effective in middle-income countries if it cost $100 or less (79).

Ethical and legal questions pose another area of concern. Among the potential risks cited in a survey of 75 psychiatrists at “early adopter” institutions offering pharmacogenetic testing clinically, respondents expressed the most concern about the risks of getting secondary information about disease risks and endorsed the importance of confidentiality and informed consent; however, there was a lack of consensus overall about risks and the safeguards needed to protect patients (80).

Nevertheless, pharmacogenetic testing is already available for several hundred dollars a test (81) at a number of institutions. In the survey by Hoop et al. cited above, 64% of respondents had ordered at least one pharmacogenetic test in the last year. Genetic testing appears to be ordered most often for cases of treatment-resistant depression or medication intolerance (80, 82). Thus researchers have recommended developing guidelines for testing that take into account clinical severity and indicators of treatment resistance (82).

Conclusion

Despite mixed or disappointing results from some of these drug trials and difficult times for psychiatric drug development in general, there are nevertheless promising signs in antidepressant medication research. Our increasing ability to target individual neurotransmitter receptors with specific agonist or antagonist activity could lead to more effective and more tolerable medications. After a long drought in the development of truly novel pathways for antidepressant action, the fact that mechanisms other than monoamine neurotransmission are being targeted is encouraging. Furthermore, in addition to allowing for the personalization of medication treatment, the study of psychiatric genetics and biomarkers of antidepressant response could expand knowledge about the pathophysiology of depression and unearth new treatment targets for the future.

Address correspondence to Trina Chang, M.D., M.P.H., Depression Clinical and Research Program, Massachusetts General Hospital, 1 Bowdoin Square, 6th Floor, Boston, MA 02114; e-mail:

Author Information and CME Disclosure

Trina E. Chang, M.D., M.P.H., Depression Clinical and Research Program, Massachusetts General Hospital, Boston, MA.

Stephen M. Stahl, M.D., Ph.D., Department of Psychiatry, University of California-San Diego, San Diego, CA; Department of Psychiatry, University of Cambridge, Cambridge, U.K.

Dr. Chang has received research support for work with Alkermes, AstraZeneca, CeNeRx, Euthymics, Forest, GlaxoSmithKline, Johnson &Johnson, Neuralstem, and Pfizer and has received travel reimbursement from Bristol-Myers Squibb.

In the past 12 months, Dr. Stahl has served as a consultant for Acadia, AstraZeneca, Avanir, Biomarin, Bristol-Myers Squibb, CeNeRx, Dey, Eli Lilly, Forest, GenoMind, GlaxoSmithKline, Johnson & Johnson, Jazz, Lundbeck, Merck, Neuronetics, Novartis, Noven, ONO, Orexigen, Otsuka, PamLabs, Pfizer, RCT Logic, Rexahn, Roche, Servier, Shire, Solvay, Sunovion, Trius, and Valeant. He has served on speakers’ bureaus for Arbor Scientia, AstraZeneca, Eli Lilly, Forest, J&J, Merck, Neuroscience Education Institute, Pfizer, Servier, and Sunovion. He has received research and/or grant support from AstraZeneca, CeNeRx, Eli Lilly, Forest, GenOmind, Merck, Neuronetics, PamLabs, Pfizer, Roche, Schering Plough, Sepracor, Servier, Shire, Sunovion, Torrent, and Trovis.

References

1 http://www.cdc.gov/nchs/data/nhsr/nhsr008.pdfGoogle Scholar

2 Rush AJ, Trivedi MH, Wisniewski SR, Stewart JW, Nierenberg AA, Thase ME, Ritz L, Biggs MM, Warden D, Luther JF, Shores-Wilson K, Niederehe G, Fava MSTAR*D Study Team: Bupropion-SR, sertraline, or venlafaxine-XR after failure of SSRIs for depression. N Engl J Med 2006; 354:1231–1242CrossrefGoogle Scholar

3 Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, Ritz L, Nierenberg AA, Lebowitz BD, Biggs MM, Luther JF, Shores-Wilson K, Rush AJSTAR*D Study Team: Medication augmentation after the failure of SSRIs for depression. N Engl J Med 2006; 354:1243–1252CrossrefGoogle Scholar

4 Schwartz TL, Siddiqui UA, Stahl SM: Vilazodone: a brief pharmacologic and clinical review of the novel serotonin partial agonist and reuptake inhibitor. Therapeutic Advances in Psychopharmacology 2011; 1:81–87CrossrefGoogle Scholar

5 Stahl SM: Stahl’s Essential Psychopharmacology, 4th edition, chapter 7. New York, Cambridge University Press, in press.Google Scholar

6 Mørk A, Pehrson A, Brennum LT, Nielsen SM, Zhong H, Lassen AB, Miller S, Westrich L, Boyle NJ, Sánchez C, Fischer CW, Liebenberg N, Wegener G, Bundgaard C, Hogg S, Bang-Andersen B, Stensbøl TB: Pharmacological effects of Lu AA21004: a novel multimodal compound for the treatment of major depressive disorder. J Pharmacol Exp Ther 2012; 340:666–675CrossrefGoogle Scholar

7 Alvarez E, Perez V, Dragheim M, Loft H, Artigas F: A double-blind, randomized, placebo-controlled, active reference study of Lu AA21004 in patients with major depressive disorder. Int J Neuropsychopharmacol 2012; 15:589–600CrossrefGoogle Scholar

8 Henigsberg N, Mahableshwarkar AR, Jacobsen P, Chen Y, Thase ME: A randomized, double-blind, placebo-controlled 8-week trial of the efficacy and tolerability of multiple doses of lu aa21004 in adults with major depressive disorder. J Clin Psychiatry 2012; 73:953–959CrossrefGoogle Scholar

9 Katona C, Hansen T, Olsen CK: A randomized, double-blind, placebo-controlled, duloxetine-referenced, fixed-dose study comparing the efficacy and safety of Lu AA21004 in elderly patients with major depressive disorder. Int Clin Psychopharmacol 2012; 27:215:223Google Scholar

10 Boulenger JP, Loft H, Florea I: A randomized clinical study of Lu AA21004 in the prevention of relapse in patients with major depressive disorder. J Psychopharmacol 2012; [Epub ahead of print]CrossrefGoogle Scholar

11 Baldwin DS, Loft H, Dragheim M: A randomised, double-blind, placebo controlled, duloxetine-referenced, fixed-dose study of three dosages of Lu AA21004 in acute treatment of major depressive disorder (MDD). Eur Neuropsychopharmacol 2012; 22:482–491CrossrefGoogle Scholar

12 Bose A, Gommoll C, Li H, Rut A: Levomilnacipran in the treatment of major depressive disorder: an analysis of efficacy data from 2 phase III studies. Poster presented at American Psychiatric Association annual meeting, May 8, 2012, Philadelphia, PA.Google Scholar

13 Pangallo B, Dellva MA, D’Souza DN, Essink B, Russell J, Goldberger C: A randomized, double-blind study comparing LY2216684 and placebo in the treatment of major depressive disorder. J Psychiatr Res 2011; 45:748–755CrossrefGoogle Scholar

14 Fava M, Thase ME, DeBattista C, Doghramji K, Arora S, Hughes RJ: Modafinil augmentation of selective serotonin reuptake inhibitor therapy in MDD partial responders with persistent fatigue and sleepiness. Ann Clin Psychiatry 2007; 19:153–159CrossrefGoogle Scholar

15 Damsa C, Bumb A, Bianchi-Demicheli F, Vidailhet P, Sterck R, Andreoli A, Beyenburg S: “Dopamine-dependent” side effects of selective serotonin reuptake inhibitors: a clinical review. J Clin Psychiatry 2004; 65:1064–1068CrossrefGoogle Scholar

16 Clayton AH, Warnock JK, Kornstein SG, Pinkerton R, Sheldon-Keller A, McGarvey EL: A placebo-controlled trial of bupropion SR as an antidote for selective serotonin reuptake inhibitor-induced sexual dysfunction. J Clin Psychiatry 2004; 65:62–67CrossrefGoogle Scholar

17 Thase M, Fava M, Hobart M, Skuban A, Zhang P, McQuade RD, Carson WH, Sanchez R, Forbes RA: Efficacy and safety of adjunctive OPC-34712 in major depressive disorder: a phase II, randomized, placebo-contolled study. Poster presented at the American Psychiatric Association annual meeting, Honolulu, HI, May 16, 2011.Google Scholar

18 Harvey P, Roth RM, Bilder RM, Richards C, Lasser R, Geibel B, Gao J, Scheckner B, Trivedi M: Assessment of executive dysfunction in adults with major depressive disorder receiving lisdexamfetamine dismesylate augmentation of escitalopram. Poster presented at the American Psychiatric Association annual meeting, May 6, 2012, Philadelphia, PA.Google Scholar

19 Keefe R, Boyd A, Madhoo M, Roth RM, Sambunaris A, Wu J, Trivedi M, Anderson CS, Lasser R: Lisdexamfetamine dimesylate in the treatment of cognitive dysfunction in patients with partially or fully remitted major depressive disorder. Poster presented at the American Psychiatric Association annual meeting, May 8, 2012, Philadelphia, PA.Google Scholar

20 Liang Y, Shaw AM, Boules M, Briody S, Robinson J, Oliveros A, Blazar E, Williams K, Zhang Y, Carlier PR, Richelson E: Antidepressant-like pharmacological profile of a novel triple reuptake inhibitor, (1S,2S)-3-(methylamino)-2-(naphthalen-2-yl)-1-phenylpropan-1-ol (PRC200-SS). J Pharmacol Exp Ther 2008; 327:573–583CrossrefGoogle Scholar

21 McMillen BA, Shank JE, Jordan KB, Williams HL, Basile AS: Effect of DOV 102,677 on the volitional consumption of ethanol by Myers’ high ethanol-preferring rat. Alcohol Clin Exp Res 2007; 31:1866–1871CrossrefGoogle Scholar

22 Tran P, Skolnick P, Czobor P, Huang NY, Bradshaw M, McKinney A, Bymaster F, Fava M: Efficacy and safety of the novel triple reuptake inhibitor EB-1010 in the treatment of patients with major depressive disorder. Poster presented at the American College of Neuropsychopharmacology annual meeting, December 8, 2010, Miami Beach, Fla. http://s358120677.onlinehome.us/wp-content/uploads/2011/03/clinical_r3_press_quality.pdfGoogle Scholar

23 Fowler JS, Logan J, Azzaro AJ, Fielding RM, Zhu W, Poshusta AK, Burch D, Brand B, Free J, Asgharnejad M, Wang GJ, Telang F, Hubbard B, Jayne M, King P, Carter P, Carter S, Xu Y, Shea C, Muench L, Alexoff D, Shumay E, Schueller M, Warner D, Apelskog-Torres K: Reversible inhibitors of monoamine oxidase-A (RIMAs): robust, reversible inhibition of human brain MAO-A by CX157. Neuropsychopharmacology 2010; 35:623–631CrossrefGoogle Scholar

24 Sanacora G: Do glutamatergic agents represent a new class of antidepressant drugs? Part 1. J Clin Psychiatry 2009; 70:1473–1475CrossrefGoogle Scholar

25 Aan Het Rot M, Zarate CA, Charney DS, Mathew SJ: Ketamine for depression: where do we go from here? Biol Psychiatry 2012; 72:537–547CrossrefGoogle Scholar

26 Sanacora G, Kendell SF, Levin Y, Simen AA, Fenton LR, Coric V, Krystal JH: Preliminary evidence of riluzole efficacy in antidepressant-treated patients with residual depressive symptoms. Biol Psychiatry 2007; 61:822–825CrossrefGoogle Scholar

27 Zarate CA, Payne JL, Quiroz J, Sporn J, Denicoff KK, Luckenbaugh D, Charney DS, Manji HK: An open-label trial of riluzole in patients with treatment-resistant major depression. Am J Psychiatry 2004; 161:171–174CrossrefGoogle Scholar

28 Rogóz Z, Skuza G, Daniel WA, Wójcikowski J, Dudek D, Wróbel A: Amantadine as an additive treatment in patients suffering from drug-resistant unipolar depression. Pharmacol Rep 2007; 59:778–784Google Scholar

29 Preskorn SH, Baker B, Kolluri S, Menniti FS, Krams M, Landen JW: An innovative design to establish proof of concept of the antidepressant effects of the NR2B subunit selective N-methyl-D-aspartate antagonist, CP-101,606, in patients with treatment-refractory major depressive disorder. J Clin Psychopharmacol 2008; 28:631–637CrossrefGoogle Scholar

30 Lauterbach EC: An extension of hypotheses regarding rapid-acting, treatment-refractory, and conventional antidepressant activity of dextromethorphan and dextrorphan. Med Hypotheses 2012; 78:693–702CrossrefGoogle Scholar

31 Ferguson JM, Shingleton RN: An open-label, flexible-dose study of memantine in major depressive disorder. Clin Neuropharmacol 2007; 30:136–144CrossrefGoogle Scholar

32 Muhonen LH, Lönnqvist J, Juva K, Alho H: Double-blind, randomized comparison of memantine and escitalopram for the treatment of major depressive disorder comorbid with alcohol dependence. J Clin Psychiatry 2008; 69:392–399CrossrefGoogle Scholar

33 Zarate CA, Singh JB, Quiroz JA, De Jesus G, Denicoff KK, Luckenbaugh DA, Manji HK, Charney DS: A double-blind, placebo-controlled study of memantine in the treatment of major depression. Am J Psychiatry 2006; 163:153–155CrossrefGoogle Scholar

34 Canan F, Ataoglu A: Memantine-related psychotic symptoms in a patient with bipolar disorder. J Clin Psychiatry 2010; 71:957CrossrefGoogle Scholar

35 Smith EJ: Amantadine-induced psychosis in a young healthy patient. Am J Psychiatry 2008; 165:1613CrossrefGoogle Scholar

36 Mathew SJ, Keegan K, Smith L: Glutamate modulators as novel interventions for mood disorders. Rev Bras Psiquiatr 2005; 27:243–248CrossrefGoogle Scholar

37 Sanacora G: Do glutamatergic agents represent a new class of antidepressant drugs? Part 2. J Clin Psychiatry 2009; 70:1604–1605CrossrefGoogle Scholar

38 http://www.clinicaltrials.gov/ct2/show/study/NCT00113022?term=NCT00113022&rank=1Google Scholar

39 Saricicek A, Esterlis I, Maloney KH, Mineur YS, Ruf BM, Muralidharan A, Chen JI, Cosgrove KP, Kerestes R, Ghose S, Tamminga CA, Pittman B, Bois F, Tamagnan G, Seibyl J, Picciotto MR, Staley JK, Bhagwagar Z: Persistent β(2)*-nicotinic acetylcholinergic receptor dysfunction in major depressive disorder. Am J Psychiatry 2012; 169:851–859CrossrefGoogle Scholar

40 Zimmerman M, Chelminski I, McDermut W: Major depressive disorder and axis I diagnostic comorbidity. J Clin Psychiatry 2002; 63:187–193CrossrefGoogle Scholar

41 Grant BF, Hasin DS, Chou SP, Stinson FS, Dawson DA: Nicotine dependence and psychiatric disorders in the United States: results from the national epidemiologic survey on alcohol and related conditions. Arch Gen Psychiatry 2004; 61:1107–1115CrossrefGoogle Scholar

42 Chen Y, Sharples TJ, Phillips KG, Benedetti G, Broad LM, Zwart R, Sher E: The nicotinic alpha 4 beta 2 receptor selective agonist, TC-2559, increases dopamine neuronal activity in the ventral tegmental area of rat midbrain slices. Neuropharmacology 2003; 45:334–344CrossrefGoogle Scholar

43 Grilli M, Patti L, Robino F, Zappettini S, Raiteri M, Marchi M: Release-enhancing pre-synaptic muscarinic and nicotinic receptors co-exist and interact on dopaminergic nerve endings of rat nucleus accumbens. J Neurochem 2008; 105:2205–2213CrossrefGoogle Scholar

44 Sharples CG, Kaiser S, Soliakov L, Marks MJ, Collins AC, Washburn M, Wright E, Spencer JA, Gallagher T, Whiteaker P, Wonnacott S: UB-165: a novel nicotinic agonist with subtype selectivity implicates the alpha4beta2* subtype in the modulation of dopamine release from rat striatal synaptosomes. J Neurosci 2000; 20:2783–2791Google Scholar

45 Levin ED: α7-Nicotinic receptors and cognition. Curr Drug Targets 2012; 13:602–606CrossrefGoogle Scholar

46 Philip NS, Carpenter LL, Tyrka AR, Whiteley LB, Price LH: Varenicline augmentation in depressed smokers: an 8-week, open-label study. J Clin Psychiatry 2009; 70:1026–1031CrossrefGoogle Scholar

47 http://www.astrazeneca.com/Media/Press-releases/Article/20032012tc5214-failed-phase-iii-endpointGoogle Scholar

48 Margolis EB, Hjelmstad GO, Bonci A, Fields HL: Kappa-opioid agonists directly inhibit midbrain dopaminergic neurons. J Neurosci 2003; 23:9981–9986CrossrefGoogle Scholar

49 Margolis EB, Hjelmstad GO, Bonci A, Fields HL: Both kappa and mu opioid agonists inhibit glutamatergic input to ventral tegmental area neurons. J Neurophysiol 2005; 93:3086–3093CrossrefGoogle Scholar

50 Land BB, Bruchas MR, Schattauer S, Giardino WJ, Aita M, Messinger D, Hnasko TS, Palmiter RD, Chavkin C: Activation of the kappa opioid receptor in the dorsal raphe nucleus mediates the aversive effects of stress and reinstates drug seeking. Proc Natl Acad Sci USA 2009; 106:19168–19173CrossrefGoogle Scholar

51 Ide S, Sora I, Ikeda K, Minami M, Uhl GR, Ishihara K: Reduced emotional and corticosterone responses to stress in mu-opioid receptor knockout mice. Neuropharmacology 2010; 58:241–247CrossrefGoogle Scholar

52 Berrocoso E, Rojas-Corrales MO, Micó JA: Non-selective opioid receptor antagonism of the antidepressant-like effect of venlafaxine in the forced swimming test in mice. Neurosci Lett 2004; 363:25–28CrossrefGoogle Scholar

53 Ide S, Fujiwara S, Fujiwara M, Sora I, Ikeda K, Minami M, Uhl GR, Ishihara K: Antidepressant-like effect of venlafaxine is abolished in μ-opioid receptor-knockout mice. J Pharmacol Sci 2010; 114:107–110CrossrefGoogle Scholar

54 Kennedy SE, Koeppe RA, Young EA, Zubieta JK: Dysregulation of endogenous opioid emotion regulation circuitry in major depression in women. Arch Gen Psychiatry 2006; 63:1199–1208CrossrefGoogle Scholar

55 Garriock HA, Tanowitz M, Kraft JB, Dang VC, Peters EJ, Jenkins GD, Reinalda MS, McGrath PJ, von Zastrow M, Slager SL, Hamilton SP: Association of mu-opioid receptor variants and response to citalopram treatment in major depressive disorder. Am J Psychiatry 2010; 167:565–573CrossrefGoogle Scholar

56 Tejeda HA, Shippenberg TS, Henriksson R: The dynorphin/κ-opioid receptor system and its role in psychiatric disorders. Cell Mol Life Sci 2012; 69:857–896CrossrefGoogle Scholar

57 Carlezon WA, Béguin C, Knoll AT, Cohen BM: Kappa-opioid ligands in the study and treatment of mood disorders. Pharmacol Ther 2009; 123:334–343CrossrefGoogle Scholar

58 Emrich HM, Vogt P, Herz A: Possible antidepressive effects of opioids: action of buprenorphine. Ann N Y Acad Sci 1982; 398:108–112CrossrefGoogle Scholar

59 Fava M, Bodkin JA, Thase M, Trivedi M, Leigh-Pemberton R, Du Y, Ehrich E: A pilot study of ALKS 5461 (buprenorphine combined with ALKS 33) in treatment-resistant depression. Poster presented at NCDEU annual meeting, May 31, 2012, Phoenix, AZ.Google Scholar

60 Ebner K, Wotjak CT, Landgraf R, Engelmann M: Forced swimming triggers vasopressin release within the amygdala to modulate stress-coping strategies in rats. Eur J Neurosci 2002; 15:384–388CrossrefGoogle Scholar

61 Goodson JL, Evans AK: Neural responses to territorial challenge and nonsocial stress in male song sparrows: segregation, integration, and modulation by a vasopressin V1 antagonist. Horm Behav 2004; 46:371–381CrossrefGoogle Scholar

62 Griebel G, Beeske S, Stahl SM: The V1b receptor antagonist SSRI49415 in the treatment of major depressive and generalized anxiety disorders: results from four double-blind, placebo-controlled studies. J Clin Psychiatry (in press)Google Scholar

63 Lôo H, Hale A, D’haenen H: Determination of the dose of agomelatine, a melatoninergic agonist and selective 5-HT(2C) antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study. Int Clin Psychopharmacol 2002; 17:239–247CrossrefGoogle Scholar

64 Demyttenaere K: Agomelatine: a narrative review. Eur Neuropsychopharmacol 2011; 21(Suppl 4):S703–S709CrossrefGoogle Scholar

65 Montgomery SA, Kennedy SH, Burrows GD, Lejoyeux M, Hindmarch I: Absence of discontinuation symptoms with agomelatine and occurrence of discontinuation symptoms with paroxetine: a randomized, double-blind, placebo-controlled discontinuation study. Int Clin Psychopharmacol 2004; 19:271–280CrossrefGoogle Scholar

66 Kennedy SH, Rizvi SJ: Agomelatine in the treatment of major depressive disorder: potential for clinical effectiveness. CNS Drugs 2010; 24:479–499CrossrefGoogle Scholar

67 Stahl SM, Fava M, Trivedi MH, Caputo A, Shah A, Post A: Agomelatine in the treatment of major depressive disorder: an 8-week, multicenter, randomized, placebo-controlled trial. J Clin Psychiatry 2010; 71:616–626CrossrefGoogle Scholar

68 Zajecka J, Schatzberg A, Stahl S, Shah A, Caputo A, Post A: Efficacy and safety of agomelatine in the treatment of major depressive disorder: a multicenter, randomized, double-blind, placebo-controlled trial. J Clin Psychopharmacol 2010; 30:135–144CrossrefGoogle Scholar

69 http://psychnews.psychiatryonline.org/newsarticle.aspx?articleid=1032188&RelatedNewsArticles=trueGoogle Scholar

70 Collier DA, Stöber G, Li T, Heils A, Catalano M, Di Bella D, Arranz MJ, Murray RM, Vallada HP, Bengel D, Müller CR, Roberts GW, Smeraldi E, Kirov G, Sham P, Lesch KP: A novel functional polymorphism within the promoter of the serotonin transporter gene: possible role in susceptibility to affective disorders. Mol Psychiatry 1996; 1:453–460Google Scholar

71 Mundo E, Walker M, Cate T, Macciardi F, Kennedy JL: The role of serotonin transporter protein gene in antidepressant-induced mania in bipolar disorder: preliminary findings. Arch Gen Psychiatry 2001; 58:539–544CrossrefGoogle Scholar

72 Laje G, Perlis RH, Rush AJ, McMahon FJ: Pharmacogenetics studies in STAR*D: strengths, limitations, and results. Psychiatr Serv 2009; 60:1446–1457CrossrefGoogle Scholar

73 Uher R, Huezo-Diaz P, Perroud N, Smith R, Rietschel M, Mors O, Hauser J, Maier W, Kozel D, Henigsberg N, Barreto M, Placentino A, Dernovsek MZ, Schulze TG, Kalember P, Zobel A, Czerski PM, Larsen ER, Souery D, Giovannini C, Gray JM, Lewis CM, Farmer A, Aitchison KJ, McGuffin P, Craig I: Genetic predictors of response to antidepressants in the GENDEP project. Pharmacogenomics J 2009; 9:225–233CrossrefGoogle Scholar

74 Domschke K, Lawford B, Laje G, Berger K, Young R, Morris P, Deckert J, Arolt V, McMahon FJ, Baune BT: Brain-derived neurotrophic factor (BDNF) gene: no major impact on antidepressant treatment response. Int J Neuropsychopharmacol 2010; 13:93–101CrossrefGoogle Scholar

75 Horstmann S, Lucae S, Menke A, Hennings JM, Ising M, Roeske D, Müller-Myhsok B, Holsboer F, Binder EB: Polymorphisms in GRIK4, HTR2A, and FKBP5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology 2010; 35:727–740CrossrefGoogle Scholar

76 Laje G, McMahon FJ: The pharmacogenetics of major depression: past, present, and future. Biol Psychiatry 2007; 62:1205–1207CrossrefGoogle Scholar

77 Perlis RH, Patrick A, Smoller JW, Wang PS: When is pharmacogenetic testing for antidepressant response ready for the clinic? A cost-effectiveness analysis based on data from the STAR*D study. Neuropsychopharmacology 2009; 34:2227–2236CrossrefGoogle Scholar

78 Lee CP, Chertow GM, Zenios SA: An empiric estimate of the value of life: updating the renal dialysis cost-effectiveness standard. Value Health 2009; 12:80–87CrossrefGoogle Scholar

79 Olgiati P, Bajo E, Bigelli M, De Ronchi D, Serretti A: Should pharmacogenetics be incorporated in major depression treatment? Economic evaluation in high- and middle-income European countries. Prog Neuropsychopharmacol Biol Psychiatry 2012; 36:147–154CrossrefGoogle Scholar

80 Hoop JG, Lapid MI, Paulson RM, Roberts LW: Clinical and ethical considerations in pharmacogenetic testing: views of physicians in 3 “early adopting” departments of psychiatry. J Clin Psychiatry 2010; 71:745–753CrossrefGoogle Scholar

81 http://www.nigms.nih.gov/Research/FeaturedPrograms/PGRN/Background/pgrn_faq.htmGoogle Scholar

82 Rundell JR, Staab JP, Shinozaki G, Saad-Pendergrass D, Moore K, McAlpine D, Mrazek D: Pharmacogenomic testing in a tertiary care outpatient psychosomatic medicine practice. Psychosomatics 2011; 52:141–146CrossrefGoogle Scholar