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.

×
Published Online:https://doi.org/10.1176/foc.3.1.3

Clinical context

Epidemiology update

The statistics for the epidemiology and course of major depressive disorder (MDD) remain impressive. As of 2000, MDD ranked as the fourth leading cause of disability for women and seventh for men, and it is projected to be second only to cardiovascular diseases by the year 2020 (1, 2). In the United States, 16.2% of the population have had MDD in their lifetime and 6.6% have experienced an episode of major depression within the past year (3).

Of those with MDD, about 90% are classified as at least moderately depressed. The odds ratios for women compared with men are 1.7 for lifetime MDD and 1.4 for 12-month prevalence. Fifty percent of persons who have had one episode of major depression will have at least one more episode, as will 70% of those who have had two episodes and 90% of those who have had three episodes (4). About 20%–25% of persons who have major depression will develop chronic depression, with the episode lasting at least 2 years (5). It has been estimated that as many as 30% of patients who present for primary care have MDD (6, 7).

These epidemiological data indicate that major depressive disorder continues to be widespread, is frequently chronic and recurrent, and presents physicians of all disciplines with a substantial clinical challenge.

Diagnosis

According to DSM-IV-TR (8) criteria, a diagnosis of MDD requires that at least five of nine symptoms be present (Table 1), although many other symptoms have been associated with the disorder (Table 2). However, in a key genetic epidemiological study of twins, Kendler and Gardner found that the presence of as few as four symptoms of MDD with less than severe impairment and with symptoms lasting less than 14 days in one twin was associated with a substantial risk of MDD in the co-twin (9). Dysfunction can occur with two of the nine symptoms, and functioning is unequivocally worse for patients with severe depression, defined as meeting threshold criteria for three or more of the nine symptoms (1012). With as few as two of the nine symptoms of depression associated with dysfunction, cause and effect cannot be determined (do the symptoms cause the dysfunction or does the dysfunction cause the symptoms?), leaving the threshold for treatment unclear. A full spectrum of depressive disorders has been proposed (8) (Table 3).

Minor and subsyndromal depression may precede an episode of MDD as prodromal symptoms, occur after an episode of MDD as residual symptoms, or persist without ever fully meeting the diagnostic criteria for MDD (1316). Whether or not minor and subsyndromal depression should be treated with antidepressants is still an open question, with some studies finding no difference between active drug and placebo. For example, in a placebo-controlled study comparing paroxetine and problem-solving psychotherapy for elderly primary care patients with minor depression (17), these treatments were modestly better than placebo, but functioning improved only among participants whose functioning was the worst at baseline. Moreover, remission rates were similar for all three treatment groups: paroxetine, 53.1%; psychotherapy, 44.0%; and placebo, 49.1%. A similar study of younger primary care patients with minor depression found no differences among treatment groups receiving paroxetine, psychotherapy, or placebo (18). However, Judd and colleagues (19) found active drug to be superior to placebo in a study of fluoxetine, which was significantly more effective than placebo in improving symptoms of minor depression and improving psychosocial functioning.

When minor or subsyndromal depression is the outcome of an episode of major depression, in contrast to an outcome of full remission, patients are at higher risk of relapse and recurrence (13, 20). Remission, therefore, has become the goal of treatment (21, 22). Unfortunately, only a minority of patients achieve remission with available treatments, and multimodal interventions or thoughtful polypharmacy may be necessary (6, 12). Residual symptoms have increasingly attracted the attention of researchers, and targeted interventions are under development for the management of persistent fatigue, insomnia, and amotivational states (2123).

Although minor and subsyndromal depression are associated with distress and dysfunction, it is not clear when subsyndromal and minor depression should be treated: What is the threshold for treatment? In major depressive disorder, residual symptoms after a course of treatment are also associated with distress and dysfunction. In both cases, if distress and dysfunction are present, clinicians should treat their patients to remission whenever possible. One problem in practice, however, is that it can be difficult to determine when and whether remission occurs if depressive symptoms are not measured periodically. Patient rating forms provide a simple means of measuring symptoms. The Quick Inventory of Depressive Symptomatology–Self-Report version (QIDS-SR) (24) or the Beck Depression Inventory II (BDI-II) (25) can easily be incorporated into clinical practice. Patients can complete such instruments in a few minutes and help clinicians track progress (or lack thereof). Reviewing the self-report with the patient allows the clinician to have a frank and focused discussion with the patient about residual symptoms, negotiate the next steps, and evaluate the effectiveness of any interventions.

Comorbidity

Nearly three-quarters of persons with MDD have at least one other comorbid psychiatric disorder (3). Most common are anxiety disorders (59%), followed by impulse control disorders (30%) and substance use disorders (24%). Those with comorbid disorders seek help more often and, as would be expected, do not function as well as those without any comorbid disorders (3). One of the great paradoxes of modern clinical trials of treatments for MDD is that many studies exclude most patients with comorbid disorders, which limits the generalizability of results. Zimmerman and colleagues found that among patients treated for depression in a large outpatient clinic, only 14% would meet criteria for inclusion in clinical trials (26). Clinicians should actively probe for comorbid anxiety, impulse, and substance use disorders and treat them accordingly.

Medical comorbidity and MDD are reciprocal: Patients with medical comorbidity have higher rates of depression, and those with MDD have higher rates of some medical disorders, such as cardiovascular disease (2729). Cardiovascular outcomes have been shown to be adversely affected by MDD. For patients who have depression after myocardial infarction (MI), the risk of death is three to four times higher than for those without depression, even when the analysis is covaried by left ventricular function (3032). Research suggests that while social support may not directly affect survival, high levels of social support may act as a buffer against the high risk of death in patients with post-MI depression (33).

Major depression, but not minor depression, has been found to be an independent risk factor for the development of MI—and it is as strong a risk factor as impaired left ventricular function (34). The SADHART trial, although not designed to address the question of whether antidepressant treatment improves cardiovascular outcomes for patients with post-MI depression, suggested that sertraline is a safe treatment for depression in these patients and that it might improve cardiovascular outcomes (35). Ongoing studies are addressing this question. When consulting with medical colleagues, it is important to avoid the mind-set that some depression is normal and temporary after MI and therefore that it should not be treated. Whenever depressive symptoms are present, they should be assessed (using a patient-rated measure), treatment should be initiated, and progress should be tracked.

Pathophysiology

Stress has long been implicated in the precipitation of episodes of major depression (36), but the pathophysiology of the relationship between stress and depression is quite complex. Stress has been shown in animal models to decrease gene expression of BcL-2 and BDNF (37), proteins necessary to maintain robust and healthy neurons (38). Antidepressants have been found to increase gene expression of BDNF and block the decrease precipitated by stress (39). A longitudinal study of more than 1,000 persons between ages 11 and 26 years found a gene-environment interaction that suggests that individuals with the short arm allele of the serotonin transporter gene develop depression under conditions of stressful life events or traumatic parenting, whereas those with the homozygous long arm of the gene appear protected from developing major depression (40). While stress may be a causal factor, alone it may not be sufficient to precipitate major depressive episodes.

The gene-environment relationship becomes more complex when theories of neurogenesis come into play. BDNF is necessary for the generation of new neurons in the hippocampus and dentate gyrus in adults (39, 41). Because of animal models showing a decrease in BDNF with stress and an increase with antidepressants, some researchers have postulated that in human beings impaired neurogenesis may be at the heart of the pathophysiology of depression (39). Although early studies did not find a significant difference in hippocampal volume between depressed individuals and healthy control subjects (42, 43), a decrease in BDNF is consistent with more recent neuroimaging data that show decreased hippocampal volume in patients with chronic or recurrent MDD (44, 45). No actual evidence for decreased neurogenesis in adult humans has been produced, however, nor any fundamental data on the function of new neurons and the clinical correlates of decreased neurogenesis (46). The pros and cons of these two arguments are elegantly discussed by Sapolsky (47).

Treatment strategies and evidence

Antidepressants

The large pharmaceutical companies all advertise their antidepressants as first-line agents; but “first-line” in this context is a marketing tool, not an evidence-based concept. Instead of conceptualizing one antidepressant or another as first-line, it makes sense to tailor the treatment choices to the individual patient and, as much as clinically appropriate, involve the patient in the decision of which medication to try and which side effects to risk. In this model of shared decision making, the physician and the patient reach concordance about the best course of action (48, 49). When concordance is reached, patients are more apt to take their medication regularly than when they are merely told what to do by their physician. This model of concordance and shared decision making is new to psychiatry and still evolving in other areas of medicine (48, 50). Those interested in more detail on these issues might want to read Jay Katz’s classic, The Silent World of Doctor and Patient (51), and Carl Schneider’s counterargument to Katz, The Practice of Autonomy (52). Those interested in better communication of risk and negotiating around treatment choices might want to read Gerd Gigerenzer’s excellent book Calculated Risks (53) and Barry Schwartz’s The Paradox of Choice (54) as well as Ury and Fisher’s classic on negotiation, Getting to Yes (55).

In the past few years, the hypothesis that dual-action antidepressants—agents that inhibit reuptake of both serotonin and norepinephrine—are better than single-action antidepressants has been at the center of the efficacy wars. A fundamental part of the terrain of this battleground is the emphasis on remission (becoming completely free of depression) as opposed to response (showing 50% improvement) (22). In a widely cited analysis of pooled data, Thase and colleagues found a remission rate of 45% among patients who received venlafaxine, a reuptake inhibitor of both norepinephrine and serotonin, compared with 35% for various selective serotonin reuptake inhibitors (SSRIs) and 25% for placebo (56). In contrast, Montgomery and colleagues reported that escitalopram was superior to venlafaxine (57). Why the discrepancy? A careful look at the Thase study reveals that the SSRI in six of the eight studies analyzed used fluoxetine, an agent that has a long half-life and possibly a slower onset of action than other SSRIs, so the results of the meta-analysis may not be generalizable to other SSRIs. Also, one of the studies lasted only 6 weeks, and some of the studies may have included SSRI-resistant patients.

It is essential that the treatment choices be tailored to the individual patient, typically on the basis of estimates of tolerability, possible symptom clusters, and prior treatment, and for the clinician and the patient to negotiate about a limited number of antidepressants and to share decision making.

Psychotherapies

The past two decades have seen the emergence of evidence-based psychotherapies, notably cognitive behavior therapy (CBT) and its variants (58) and interpersonal therapy (IPT) for depression (59). While studies with adult patients have generally found that the efficacy of these therapies can equal that of antidepressants, it is important to note that the subjects who agree to participate in clinical trials are those who are willing and able to participate in psychotherapy. Those who are too depressed to do the assignments associated with the psychotherapy obviously would not participate in the trials. Behavioral activation therapies can sometimes be helpful in these instances. Converging evidence supports the practice of combined therapy with antidepressants and targeted psychotherapies. In one key study, the combination of nefazodone and the cognitive behavioral analysis system of psychotherapy (CBASP) was superior to either treatment alone (60). Similarly, the combination of CBT and fluoxetine was better than either one alone for depression in adolescents (61). Results of a meta-analysis further support the combination of medication and targeted psychotherapy (62).

Long-term treatment

Since the seminal studies by Prien et al. (63), Keller et al. (64), and the Pittsburgh group (65, 66), clinical practice has shifted from time-limited treatment for each depressive episode to long-term maintenance treatment for many patients. Guidelines suggest that all patients receive continuation treatment for 4 to 6 months after response to prevent a relapse. Patients who have had three or more lifetime episodes, those whose index episode lasted 2 years or longer, those with double depression, and those whose first onset of depression occurred at 50 years of age or later should receive indefinite maintenance treatment to prevent a recurrence of depression.

Although indefinite treatment is beneficial for many patients, most stop taking their antidepressants within 6 months (67, 68). To address this reality, several sequential studies of cognitive behavior therapy have been developed that would help maintain euthymia after discontinuation of antidepressants. Paykel and colleagues found that CBT was effective for residual symptoms and prevented relapse (69). Giovanni Fava found that well-being therapy, a modification of CBT, also prevented recurrence (70). Segal and Teasdale developed a group CBT called mindfulness-based cognitive therapy (MBCT) that shows promise in this area and is being assessed for efficacy in a randomized clinical trial (71, 72).

Questions and controversies

Risk and benefit of antidepressants Among pediatric patients

The Food and Drug Administration (FDA) held extensive hearings in September 2004 on the risk-benefit ratio of antidepressants for children and adolescents (73). An FDA advisory panel examined data from randomized clinical trials with some 5,000 pediatric subjects and found little evidence of efficacy for antidepressants in this age group and some evidence of increased suicidal thoughts. In response to the FDA hearings, the American Psychiatric Association issued a statement that access to antidepressants should be maintained and that close monitoring is needed for adolescents and children taking antidepressants (74). The American College of Neuropsychopharmacology formed its own task force to assess pediatric antidepressant use. After reviewing clinical trials, epidemiological studies, and autopsy studies, the task force concluded that SSRIs and other newer antidepressant drugs “do not increase the risk of suicidal thinking or suicide attempts” in youths (75). There are other possible explanations for increased suicidal thoughts in youths taking antidepressants, such as initial activation of psychomotorically retarded depressives (giving them the energy to act on latent impulses), precipitation of mania or panic, and induction of akathisia. In examining this controversial issue, both efficacy and the side effect of emergent suicidal ideation and behavior need to be addressed.

First, randomized clinical trials of antidepressants for children and adolescents have a high placebo response rate, making it difficult to detect any therapeutic signal of the antidepressants. If one assumes that the antidepressants are ineffective for these patients, then the high placebo response rate confirms that the antidepressants do not work. On the other hand, if one assumes the opposite, that the antidepressants are effective, then the criticism is leveled at the methods used in the trials, which inflate the placebo rate. Generally, however, symptoms improve for most youths in clinical trials whether they are taking the active antidepressant or placebo. Much has been written about the placebo effect in antidepressant trials (7682). The issue is inherently complex, and it may be further complicated in the case of pediatric trials. Rater bias and competency are among the most difficult factors to assess. Baseline depression scale scores may be inflated at the start of the trial to meet inclusion criteria, and scores may be deflated as the trial progresses with the expectation of improvement; as a result, both participants taking antidepressants and those taking placebo show improvement. Furthermore, because of safety concerns and because of volunteer bias, youths who participate in trials are moderately depressed at most, not severely depressed. With these factors combined, the results of randomized clinical trials show a minimal level of effectiveness for antidepressants that may not be generalizable to the population that will be treated (83). Parker has noted the discrepancy between patients in clinical care and subjects in clinical trials and questioned the relevance of modern clinical trials for actual practice (84). Parker’s discussion refers to trials in general; the problem is compounded in pediatric studies, because parents decide whether to enroll their children in clinical trials—and parents of severely depressed youths will very likely be reluctant to have their children participate in research.

Second, no suicides occurred during any of the trials, and although the relative risk ratio for active drugs compared with placebo approached 2 (indicating that about twice as many taking active drug had suicidal thoughts or behaviors), the absolute difference was about 1.5% (73). Suicidal thoughts and suicide attempts were among the events counted, but given inconsistencies in the rules and definitions, it is unclear what was actually assessed. Another consideration is that the randomized clinical trials specifically excluded subjects considered to be at risk of suicide (those who had recently attempted suicide or currently had suicidal ideation). Thus, the trials are not designed to show the efficacy of the antidepressants in reducing suicidal ideation or behavior in those who have these problems at baseline.

It is likely that the analytical challenges of a high placebo response and the lack of any completed suicides in the studies led the FDA advisory panel to recommend that antidepressants carry a warning label indicating that pediatric patients be followed carefully, especially in the early stages of treatment, rather than an outright ban on prescribing antidepressants to youths.

Thus, when using antidepressants to treat a depressed child or adolescent, monitor carefully for the emergence or exacerbation of suicidal thoughts (perhaps with additional measures such as the QIDS-SR or another patient-rated instrument appropriate for youths), see the patient frequently enough to detect any exacerbation, and evaluate carefully for symptoms of a bipolar disorder.

An update on treatment-resistant depression

Because only a minority of patients achieve remission with antidepressant monotherapy, many clinicians turn to either augmentation or combination treatments for patients who have a partial response or no response to antidepressants (85). The sections below provide a brief review of what is new with these strategies.

Augmentation

Technically, augmentation in this context is the addition of a non–antidepressant medication to an antidepressant medication regimen. The oldest and most studied of such agents is lithium (8688). In a meta-analysis, Bauer and colleagues found that the average response rate with the addition of lithium was 45%, compared with 18% with placebo (86). Several other reports, however, found no significant difference between lithium and placebo augmentation (88). The only study published to date that has addressed the issue of duration of treatment with lithium augmentation found that patients who responded to and continued taking lithium in addition to their antidepressant did substantially better than those who were given placebo in addition to the antidepressant (89). Few studies have been published that examine the efficacy of lithium augmentation for SSRIs and other more recently introduced antidepressants.

Thyroid hormone augmentation, specifically with T3, is the second most widely studied augmentation, but this agent, too, has not been adequately studied for augmentation of the newer antidepressants. The largest of recent studies compared lithium and thyroid augmentation of tricyclic antidepressants and found them about equally effective and better than placebo (90).

One practice favored by many clinicians and slowly gaining supporting evidence from randomized controlled trials is augmentation of antidepressants with atypical antipsychotic medications for patients with nonpsychotic, treatment-resistant unipolar depression. Olanzapine (91), risperidone (92, 93), ziprasidone (94, 95), aripiprazole (96), and quetiapine (97) have been studied and found to be superior to placebo as augmentation of antidepressants for nonpsychotic depression. The long-term risks and benefits of using these medications will become clearer over time. One concern is that atypical antipsychotics have been associated with the metabolic syndrome (98, 99).

Another agent favored in clinical practice that has garnered some supportive data is the addition of the antinarcolepsy agent modafinil. An initial case series supported the use of modafinil, and subsequent reports have confirmed that it might be useful for patients with residual fatigue and sleepiness (100102). Some data support the use of la-motrigine (103106), pramipexole (107), folate (108, 109), and s-adenosylmethionine (110113), but more studies are needed to establish efficacy for these agents.

Combination

Combination treatment refers to the use of two different antidepressants together. Although combination treatment is commonly used, few studies have been published. Initial case series have suggested that the combination of SSRIs and bupropion has potential (114, 115). A controlled trial of the combination of SSRIs and mirtazapine found that mirtazapine was substantially better than placebo (116).

Switching

Switching within the same antidepressant class may be useful because mechanisms of action may differ across agents in the same class (117). Overall, the probability of response to a second SSRI after nonresponse to a first SSRI is about 50% (118). Reported response rates include 76% for patients who were switched from fluoxetine to citalopram (119), 63% for patients switched from sertraline to fluoxetine (120), and 50% for patients switched from one SSRI to another (121).

Switching classes of antidepressant is an alternative strategy (122124). In a study of chronically depressed patients treated with imipramine or sertraline in which nonresponders were switched from one agent to the other, Thase and colleagues reported response rates of 44%–60% after the switch (124). Switching from SSRIs to venlafaxine yielded response rates from 30% to 69% (123, 125127). Switching to mirtazapine, bupropion, tricyclics, and monoamine oxidase inhibitors has also been studied (128135).

STAR*D

None of the augmentation, combination, or switching options cited above, with the exception of lithium and thyroid hormone, have been evaluated in published comparison studies. To address this gap in the literature, the National Institute of Mental Health sponsored the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study (136, 137), the largest clinical trial ever conducted for depression (www.star-d.org). Patients with MDD who present in either psychiatric or primary care settings are invited to participate and are treated with citalopram (open-label) at maximally tolerated doses for up to 12 weeks. Those who do not remit are randomly assigned to a second treatment through equipoise-stratified randomization (138). Equipoise-stratified randomization essentially allows the patient and the clinician to decide which group of options makes clinical sense. For example, patients who find that they are unable to tolerate citalopram would not want to opt to be randomized to an augmentation strategy, whereas those who are doing reasonably well with citalopram but fall short of remission would not want to stop citalopram treatment and start something else. Some patients may choose all options or may opt out of having cognitive behavior therapy as either an augmentation or a switch strategy.

The augmentation/combination options if citalopram fails to bring a patient to remission include buspirone, bupropion, or CBT; the switch options include sertraline, bupropion, venlafaxine, or CBT. If these options fail to bring about remission, subsequent levels include augmentation with either lithium or thyroid hormone and switching to either mirtazapine or nortriptyline. Finally, if these options fail, patients are randomized to either tranylcypromine or the combination of venlafaxine and mirtazapine. Publication of results is expected to begin this year.

Conclusions

Depressive disorders, including major, minor, and subsyndromal depression, are widespread. Our understanding of the pathophysiology of these disorders is growing but far from complete. A stress-diathesis model has empirical support, but the story of the relationship between stress, genes, and neurogenesis is just beginning. Clinicians use widely different thresholds for prescribing an antidepressant, but when one is prescribed, the patient needs to be monitored carefully and systematically. Clinicians should consider using depression rating scales such as the QIDS-SR or the BDI-II. Taking the medication regularly is more important than which medication the patient takes. Comorbid conditions, especially anxiety disorders, need to be diagnosed and treated. New paradigms of shared decision making and negotiation can lead to greater concordance and improved outcomes. New psychotherapies are gaining ground, and combined treatment has increasing empirical support. Finally, treating depressed youths has become more complicated because of the controversy surrounding SSRIs and suicidality, and treatment must be delivered with great care.

Table 1. DSM-IV-TR Symptoms of Major Depressive Episode
1. Depressed mood
2. Markedly diminished interest or pleasure
3. Significant weight loss or weight gain, or decrease or increase in appetite
4. Insomnia or hypersomnia
5. Psychomotor agitation or retardation
6. Fatigue or loss of energy
7. Feelings of worthlessness or excessive or inappropriate guilt
8. Diminished ability to think or concentrate, or indecisiveness
9. Recurrent thoughts of death, suicidal ideation, or a suicide attempt
Table 1. DSM-IV-TR Symptoms of Major Depressive Episode
Enlarge table
Table 2. Other Common Symptoms in Unipolar Depression
Psychological
    Irritability
    Anxiety or nervousness
    Reduced libido
    Hypersensitivity to rejection or criticism
    Perfectionism or obsessiveness
    Indecisiveness
    Preoccupation with oneself
Behavioral
    Crying spells
    Interpersonal friction or confrontation
    Anger attacks or outbursts
    Avoidance of anxiety-provoking situations
    Reduced productivity
    Social withdrawal
    Avoidance of emotional and sexual intimacy
    Reduced leisure-time activities
    Development of rituals or compulsions
    Workaholic behaviors
    Substance use or abuse
    Self-sacrifice or victimization
    Self-cutting/mutilation
    Violent or assaultive behaviors
Physical
    Sexual arousal difficulties
    Erectile dysfunction
    Delayed orgasm or inability to achieve orgasm
    Pains and aches
    Headaches
    Muscle tension
    Gastrointestinal upset
    Heart palpitations
    Burning or tingling sensations
Table 2. Other Common Symptoms in Unipolar Depression
Enlarge table
Table 3. DSM-IV-TR Spectrum of Depressive Disorders
Major depressive disorder
Dysthymic disorder
Minor depressive disorder
Recurrent brief depressive disorder
Mixed anxiety-depressive disorder
Table 3. DSM-IV-TR Spectrum of Depressive Disorders
Enlarge table

From the Bipolar Clinic and Research Program at Massachusetts General Hospital, Boston.

CME Disclosure Statement Andrew A. Nierenberg, M.D., Associate Director, Depression and Clinical Research Program, Medical Director, Bipolar Programs, Massachusetts General Hospital.

Consultant: Lilly, Shire, Glaxo, Innapharma, Genaissance. Grant support: Lilly, Wyeth, Glaxo, Bristol-Myers Squibb, Cyberonics, Lichtwer, Pfizer, Cederroth, Forest, Janssen. Honoraria: Lilly, Wyeth, Glaxo

Polina Eidelman, B.A., Yelena Wu, B.A., Megan Joseph, B.A., Bipolar Clinic and Research Program, Massachusetts General Hospital. No disclosure of financial interests or affiliations to report.

Disclosure of Unapproved, Off-Label, or Investigational Use of a Product

APA policy requires disclosure by CME authors of unapproved or investigational use of products discusssed in CME programs. Off-label use of medications by individual physicians is permitted and common. Decisions about off-label use can be guided by the scientific literature and clinical experience.

Send reprint requests to Andrew A. Nierenberg, M.D., Massachusetts General Hospital, 15 Parkman Street, WAC 812, Boston, MA 02114; e-mail, .
References

1 Murray CJL, Lopez A: Global Health Statistics: A Compendium of Incidence, Prevalence, and Mortality Estimates for Over 2000 Conditions. Cambridge, Mass, Harvard School of Public HealthGoogle Scholar

2 Üstün TB, Ayuso-Mateos JL, Chatterji S, Mathers C, Murry CJL: Global burden of depressive disorders in the year 2000. Br J Psychiatry 2004; 184:386–392CrossrefGoogle Scholar

3 Kessler RC, Berglund P, Demler O, Jin R, Koretz D, Merikangas KR, Rush AJ, Walters EE, Wang PS: The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R). JAMA 2003; 289:3095–3105CrossrefGoogle Scholar

4 Kupfer DJ: Long-term treatment of depression. J Clin Psychiatry 1991; 52(suppl):28–34Google Scholar

5 Judd LL, Akiskal HS, Maser JD, Zeller PJ, Endicott J, Coryell W, Paulus MP, Kunovac JL, Leon AC, Mueller TI, Rice JA, Keller MB: A prospective 12-year study of subsyndromal and syndromal depressive symptoms in unipolar major depressive disorders. Arch Gen Psychiatry 1998; 55:694–700CrossrefGoogle Scholar

6 McQuaid JR, Stein MB, Laffaye C, McCahill ME: Depression in a primary care clinic: the prevalence and impact of an unrecognized disorder. J Affect Disord 1999; 55:1–10CrossrefGoogle Scholar

7 Cassano P, Fava M: Depression and public health: an overview. J Psychosom Res 2002; 53:849–857CrossrefGoogle Scholar

8 American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision. Washington, DC, American Psychiatric Association, 2000Google Scholar

9 Kendler KS, Gardner CO: Boundaries of major depression: an evaluation of DSM-IV criteria. Am J Psychiatry 1998; 155:172–177Google Scholar

10 Maier W, Gansicke M, Weiffenbach O: The relationship between major and subthreshold variants of unipolar depression. J Affect Disord 1997; 45:41–51CrossrefGoogle Scholar

11 Miller IW, Keitner GI, Schatzberg AF, Klein DN, Thase ME, Rush AJ, Markowitz JC, Schlager DS, Kornstein SG, Davis SM, Harrison WM, Keller MB: The treatment of chronic depression, part 3: psychosocial functioning before and after treatment with sertraline or imipramine. J Clin Psychiatry 1998; 59:608–619CrossrefGoogle Scholar

12 Rapaport MH, Judd LL: Minor depressive disorder and subsyndromal depressive symptoms: functional impairment and response to treatment. J Affect Disord 1998; 48:227–232CrossrefGoogle Scholar

13 Judd LL, Schettler PJ, Akiskal HS: The prevalence, clinical relevance, and public health significance of subthreshold depressions. Psychiatr Clin North Am 2002; 25:685–698CrossrefGoogle Scholar

14 Rapaport MH, Judd LL, Schettler PJ, Yonkers KA, Thase ME, Kupfer DJ, Frank E, Plewes JM, Tollefson GD, Rush AJ: A descriptive analysis of minor depression. Am J Psychiatry 2002; 159:637–643CrossrefGoogle Scholar

15 Sadek N, Bona J: Subsyndromal symptomatic depression: a new concept. Depress Anxiety 2000; 12:30–39CrossrefGoogle Scholar

16 Sullivan PF, Prescott CA, Kendler KS: The subtypes of major depression in a twin registry. J Affect Disord 2002; 68:273–284CrossrefGoogle Scholar

17 Williams JW Jr, Barrett J, Oxman T, Frank E, Katon W, Sullivan M, Cornell J, Sengupta A: Treatment of dysthymia and minor depression in primary care: a randomized controlled trial in older adults. JAMA 2000; 284:1519–1526CrossrefGoogle Scholar

18 Barrett JE, Williams JW Jr, Oxman TE, Frank E, Katon W, Sullivan M, Hegel MT, Cornell JE, Sengupta AS: Treatment of dysthymia and minor depression in primary care: a randomized trial in patients aged 18 to 59 years. J Fam Pract 2001; 50:405–412Google Scholar

19 Judd LL, Rapaport MH, Yonkers KA, Rush AJ, Frank E, Thase ME, Kupfer DJ, Plewes JM, Schettler PJ, Tollefson G: Randomized, placebo-controlled trial of fluoxetine for acute treatment of minor depressive disorder. Am J Psychiatry 2004; 161:1864–1871CrossrefGoogle Scholar

20 Paykel ES, Ramana R, Cooper Z, Hayhurst H, Kerr J, Barocka A: Residual symptoms after partial remission: an important outcome in depression. Psychol Med 1995; 25:1171–1180CrossrefGoogle Scholar

21 Keller MB: Remission versus response: the new gold standard of antidepressant care. J Clin Psychiatry 2004; 65(suppl 4):53–59Google Scholar

22 Nierenberg AA, Wright EC: Evolution of remission as the new standard in the treatment of depression. J Clin Psychiatry 1999; 60(suppl 22):7–11CrossrefGoogle Scholar

23 Fava GA, Fabbri S, Sonino N: Residual symptoms in depression: an emerging therapeutic target. Prog Neuropsychopharmacol Biol Psychiatry 2002; 26:1019–1027CrossrefGoogle Scholar

24 Rush AJ, Trivedi MH, Ibrahim HM, Carmody TJ, Arnow B, Klein DN, Markowitz JC, Ninan PT, Kornstein S, Manber R, Thase ME, Kocsis JH, Keller MB: The 16-Item Quick Inventory of Depressive Symptomatology (QIDS), clinician rating (QIDS-C), and self-report (QIDS-SR): a psychometric evaluation in patients with chronic major depression. Biol Psychiatry 2003; 54:573–583CrossrefGoogle Scholar

25 Beck AT, Steer RA, Brown GK: Manual for the Beck Depression Inventory–II. San Antonio, Tex, Psychological Corporation, 1996Google Scholar

26 Zimmerman M, Mattia JI, Posternak MA: Are subjects in pharmacological treatment trials of depression representative of patients in routine clinical practice? Am J Psychiatry 2002; 159:469–473CrossrefGoogle Scholar

27 Katon WJ: Clinical and health services relationships between major depression, depressive symptoms, and general medical illness. Biol Psychiatry 2003; 54:216–226CrossrefGoogle Scholar

28 Patten SB: Long-term medical conditions and major depression in the Canadian population. Can J Psychiatry 1999; 44:151–157CrossrefGoogle Scholar

29 Wells KB, Golding JM, Burnam MA: Psychiatric disorder in a sample of the general population with and without chronic medical conditions. Am J Psychiatry 1988; 145:976–981CrossrefGoogle Scholar

30 Frasure-Smith N, Lespérance F: Depression and other psychological risks following myocardial infarction. Arch Gen Psychiatry 2003; 60:627–636CrossrefGoogle Scholar

31 Frasure-Smith N, Lespérance F, Talajic M: Depression following myocardial infarction: impact on 6-month survival. JAMA 1993; 270:1819–1825CrossrefGoogle Scholar

32 Frasure-Smith N, Lespérance F, Talajic M: Depression and 18-month prognosis after myocardial infarction. Circulation 1995; 91:999–1005CrossrefGoogle Scholar

33 Frasure-Smith N, Lespérance F, Gravel G, Masson A, Juneau M, Talajic M, Bourassa MG: Social support, depression, and mortality during the first year after myocardial infarction. Circulation 2000; 101:1919–1924CrossrefGoogle Scholar

34 Frasure-Smith N, Lespérance F, Talajic M: The impact of negative emotions on prognosis following myocardial infarction: is it more than depression? Health Psychol 1995; 14:388–398CrossrefGoogle Scholar

35 Shapiro PA, Lesperance F, Frasure-Smith N, O’Connor CM, Baker B, Jiang JW, Dorian P, Harrison W, Glassman AH: An open-label preliminary trial of sertraline for treatment of major depression after acute myocardial infarction (the SADHAT Trial). Sertraline Anti-Depressant Heart Attack Trial. Am Heart J 1999; 137:1100–1106CrossrefGoogle Scholar

36 Post RM: Transduction of psychosocial stress into the neurobiology of recurrent affective disorder. Am J Psychiatry 1992; 149:999–1010CrossrefGoogle Scholar

37 Luo C, Xu H, Li XM: Post-stress changes in BDNF and Bcl-2 immunoreactivities in hippocampal neurons: effect of chronic administration of olanzapine. Brain Res 2004; 1025:194–202CrossrefGoogle Scholar

38 Hasler G, Wayne CD, Husseini KM, Dennis SC: Discovering endophenotypes for major depression. Neuropsychopharmacology 2004; 29:1765–1781CrossrefGoogle Scholar

39 Duman RS: Depression: a case of neuronal life and death? Biol Psychiatry 2004; 56:140–145CrossrefGoogle Scholar

40 Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, McClay J, Mill J, Martin J, Braithwaite A, Poulton R: Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003; 301(5631):386–389CrossrefGoogle Scholar

41 Hashimoto K, Shimizu E, Iyo M: Critical role of brain-derived neurotrophic factor in mood disorders. Brain Res Brain Res Rev 2004; 45:104–114CrossrefGoogle Scholar

42 Coffey CE, Wilkinson WE, Weiner RD, Parashos IA, Djang WT, Webb MC, Figiel GS, Spritzer CE: Quantitative cerebral anatomy in depression: a controlled magnetic resonance imaging study. Arch Gen Psych 1993; 50:7–16CrossrefGoogle Scholar

43 Axelson DA, Doraiswamy PM, McDonald WM, Boyko OB, Tupler LA, Patterson LJ, Nemeroff CB, Ellinwood EH Jr, Krishnan KR: Hypercortisolemia and hippocampal changes in depression. Psychiatry Res 1993; 47:163–173CrossrefGoogle Scholar

44 Sheline YI: 3D MRI studies of neuroanatomic changes in unipolar major depression: the role of stress and medical comorbidity. Biol Psychiatry 2000; 48:791–800CrossrefGoogle Scholar

45 Henn FA, Vollmayr B: Neurogenesis and depression: etiology or epiphenomenon? Biol Psychiatry 2004; 56:146–150CrossrefGoogle Scholar

46 Mervaala E, Fohr J, Kononen M, Valkonen-Korhonen M, Vainio P, Partanen K, Partanen J, Tiihonen J, Viinamaki H, Karjalainen AK, Lehtonen J: Quantitative MRI of the hippocampus and amygdala in severe depression. Psychol Med 2000; 30:117–125CrossrefGoogle Scholar

47 Sapolsky RM: Is impaired neurogenesis relevant to the affective symptoms of depression? Biol Psychiatry 2004; 56:137–139CrossrefGoogle Scholar

48 Bissell P, May CR, Noyce PR: From compliance to concordance: barriers to accomplishing a re-framed model of health care interactions. Soc Sci Med 2004; 58:851–862CrossrefGoogle Scholar

49 Higgins N, Livingston G, Katona C: Concordance therapy: an intervention to help older people take antidepressants. J Affect Disord 2004; 81:287–291CrossrefGoogle Scholar

50 Charles C, Gafni A, Whelan T: Shared decision-making in the medical encounter: what does it mean? (Or it takes at least two to tango). Soc Sci Med 1997; 44:681–692CrossrefGoogle Scholar

51 Katz J: The Silent World of Doctor and Patient. New York, Free Press, 1994Google Scholar

52 Schneider CE: The Practice of Autonomy: Patients, Doctors, and Medical Decisions. New York, Oxford University Press, 1998Google Scholar

53 Gigerenzer G: Calculated Risks: How to Know When Numbers Deceive You. New York, Simon & Schuster, 2003Google Scholar

54 Schwartz B: The Paradox of Choice: Why More Is Less. New York, Harper Collins, 2004Google Scholar

55 Fisher R, Ury W: Getting to Yes: Negotiating Agreement Without Giving In. New York, Penguin, 1991Google Scholar

56 Thase ME, Entsuah AR, Rudolph RL: Remission rates during treatment with venlafaxine or selective serotonin reuptake inhibitors. Br J Psychiatry 2001; 178:234–241CrossrefGoogle Scholar

57 Montgomery SA, Huusom AK, Bothmer J: A randomized study comparing escitalopram with venlafaxine XR in primary care patients with major depressive disorder. Neuropsychobiology 2004; 50:57–64CrossrefGoogle Scholar

58 Beck AT, Rush AJ, Shaw BF, Emery G: Cognitive Therapy of Depression. New York, Guilford, 1987Google Scholar

59 Weissman MN, Markowitz JC: Comprehensive Guide to Interpersonal Psychotherapy. New York, Basic Books, 2000Google Scholar

60 Kocsis JH, Rush AJ, Markowitz JC, Borian FE, Dunner DL, Koran LM, Klein DN, Trivedi MH, Arnow B, Keitner G, Kornstein SG, Keller MB: Continuation treatment of chronic depression: a comparison of nefazodone, cognitive behavioral analysis system of psychotherapy, and their combination. Psychopharmacol Bull 2003; 37(4):73–87Google Scholar

61 March J, Silva S, Petrycki S, Curry J, Wells K, Fairbank J, Burns B, Domino M, McNulty S, Vitiello B, Severe J; Treatment for Adolescents With Depression Study (TADS) Team: Fluoxetine, cognitive-behavioral therapy, and their combination for adolescents with depression: Treatment for Adolescents With Depression Study (TADS) randomized controlled trial. JAMA 2004; 292:807–820CrossrefGoogle Scholar

62 Thase ME, Greenhouse JB, Frank E, Reynolds CF 3rd, Pilkonis PA, Hurley K, Grochocinski V, Kupfer DJ: Treatment of major depression with psychotherapy or psychotherapy-pharmacotherapy combinations. Arch Gen Psychiatry 1997; 54:1009–1015CrossrefGoogle Scholar

63 Prien RF, Kupfer DJ, Mansky PA, Small JG, Tuason VB, Voss CB, Johnson WE: Drug therapy in the prevention of recurrences in unipolar and bipolar affective disorders: report of the NIMH Collaborative Study Group comparing lithium carbonate, imipramine, and a lithium carbonate-imipramine combination. Arch Gen Psychiatry 1984; 41:1096–1104CrossrefGoogle Scholar

64 Keller MB, Klerman GL, Lavori PW, Coryell W, Endicott J, Taylor J: Long-term outcome of episodes of major depression: clinical and public health significance. JAMA 1984; 252:788–792CrossrefGoogle Scholar

65 Frank E, Kupfer DJ, Perel JM, Cornes C, Jarrett DB, Mallinger AG, Thase ME, McEachran AB, Grochocinski VJ: Three-year outcomes for maintenance therapies in recurrent depression. Arch Gen Psychiatry 1990; 47:1093–1099CrossrefGoogle Scholar

66 Kupfer DJ, Frank E, Perel JM, Cornes C, Mallinger AG, Thase ME, McEachran AB, Grochocinski VJ: Five-year outcome for maintenance therapies in recurrent depression. Arch Gen Psychiatry 1992; 49:769–773CrossrefGoogle Scholar

67 Lin EH, Von Korff M, Katon W, Bush T, Simon GE, Walker E, Robinson P: The role of the primary care physician in patients’ adherence to antidepressant therapy. Med Care 1995; 33:67–74CrossrefGoogle Scholar

68 McManus P, Mant A, Mitchell P, Dudley J: Length of therapy with selective serotonin reuptake inhibitors and tricyclic antidepressants in Australia. Aust N Z J Psychiatry 2004; 38:450–454CrossrefGoogle Scholar

69 Paykel ES, Scott J, Teasdale JD, Johnson AL, Garland A, Moore R, Jenaway A, Cornwall PL, Hayhurst H, Abbott R, Pope M: Prevention of relapse in residual depression by cognitive therapy: a controlled trial. Arch Gen Psychiatry 1999; 56:829–835CrossrefGoogle Scholar

70 Fava GA, Rafanelli C, Grandi S, Conti S, Belluardo P: Prevention of recurrent depression with cognitive behavioral therapy: preliminary findings. Arch Gen Psychiatry 1998; 55:816–820CrossrefGoogle Scholar

71 Segal ZV, Mark J, Williams G, Teasdale JD: Mindfulness-Based Cognitive Therapy for Depression: A New Approach to Preventing Relapse. New York, Guilford, 2001Google Scholar

72 Teasdale JD, Segal ZV, Williams JM, Ridgeway VA, Soulsby JM, Lau MA: Prevention of relapse/recurrence in major depression by mindfulness-based cognitive therapy. J Consult Clin Psychol 2000; 68:615–623CrossrefGoogle Scholar

73 Transcript of the Psychopharmacologic Drugs Advisory Committee joint meeting with the Pediatric Advisory Committee, September 13–14, 2004. Available at http://www.fda.gov/ohrms/dockets/ac/04/transcripts/2004-4065T2.htmGoogle Scholar

74 American Psychiatric Association news release, October 15, 2004. Available at http://www.psych.org/news_room/press_releases/04-55apaonfdablackboxwarning.pdfGoogle Scholar

75 American College of Neuropsychopharmacology Executive Summary of the Preliminary Report of the Task Force on SSRIs and Suicidal Behavior in Youth, January 21, 2004. Available at http://www.acnp.org/exec_summary.pdfGoogle Scholar

76 Kahn A, Warner HA, Brown WA: Symptom reduction and suicide risk in patients treated with placebo in antidepressant clinical trials: an analysis of the Food and Drug Administration database. Arch Gen Psychiatry 2000; 57:311–317CrossrefGoogle Scholar

77 Leber P: Placebo controls: no news is good news. Arch Gen Psychiatry 2000; 57:319–320CrossrefGoogle Scholar

78 Hirschfeld, RM: Suicide and antidepressant treatment. Arch Gen Psychiatry 2000; 57:325–326CrossrefGoogle Scholar

79 Leon, AC: Placebo protects subjects from nonresponse: a paradox of power. Arch Gen Psychiatry 2000; 57:329–330CrossrefGoogle Scholar

80 Lee S, Walker JR, Jakul L, Sexton K: Does elimination of placebo responders in a placebo run-in increase the treatment effect in randomized clinical trials? A meta-analytic evaluation. Depress Anxiety 2004; 19:10–19CrossrefGoogle Scholar

81 Trivedi MH, Rush J: Does a placebo run-in or a placebo treatment cell affect the efficacy of antidepressant medications? Neuropsychopharmacology 1994; 11:33–43CrossrefGoogle Scholar

82 Faries DE, Heiligenstein JH, Tollefson GD, Potter WZ: The double-blind variable placebo lead-in period: results from two antidepressant clinical trials. J Clin Psychopharmacol 2001; 21:561–568CrossrefGoogle Scholar

83 Zimmerman M, Chelminski I, Posternak MA: Exclusion criteria used in antidepressant efficacy trials: consistency across studies and representativeness of samples included. J Nerv Ment Dis 2004; 192:87–94CrossrefGoogle Scholar

84 Parker G: Evaluating treatments for the mood disorders: time for the evidence to get real. Aust N Z J Psychiatry 2004; 38:415–418Google Scholar

85 Fredman SJ, Fava M, White CN, Nierenberg AA, Rosenbaum JF: Partial response, non-response, and relapse on SSRIs in major depression: a survey of current “next-step” practices. J Clin Psychiatry 2000; 61:403–408CrossrefGoogle Scholar

86 Bauer M, Adli M, Baethge C, Berghofer A, Sasse J, Heinz A, Bschor T: Lithium augmentation therapy in refractory depression: clinical evidence and neurobiological mechanisms. Can J Psychiatry 2003; 48:440–448CrossrefGoogle Scholar

87 Bschor T, Lewitzka U, Sasse J, Adli M, Koberle U, Bauer M: Lithium augmentation in treatment-resistant depression: clinical evidence, serotonergic and endocrine mechanisms. Pharmacopsychiatry 2003; 36(suppl 3): S230–S234CrossrefGoogle Scholar

88 Nierenberg AA, Papakostas GI, Petersen T, Montoya HD, Worthington JJ, Tedlow J, Alpert JE, Fava M: Lithium augmentation of nortriptyline for subjects resistant to multiple antidepressants. J Clin Psychopharmacol 2003; 23:92–95CrossrefGoogle Scholar

89 Bauer M, Bschor T, Kunz D, Berghofer A, Andreas S, Muller-Oerlinghausen B: Double-blind, placebo-controlled trial of the use of lithium to augment antidepressant medication in continuation treatment of unipolar major depression. Am J Psychiatry 2000; 157:1429–1435CrossrefGoogle Scholar

90 Joffe RT, Singer W, Levitt AJ, McDonald: A placebo-controlled comparison of lithium and triiodothyronine augmentation of tricyclic antidepressants in unipolar refractory depression. Arch Gen Psychiatry 1993; 50:387–393CrossrefGoogle Scholar

91 Shelton RC, Tollefson GD, Tohen M, Stahl S, Gannon KS, Jacobs TG, Buras WR, Bymaster FP, Zhang W, Spencer KA, Feldman PD, Meltzer HY: A novel augmentation strategy for treating resistant major depression. Am J Psychiatry 2001; 158:131–134CrossrefGoogle Scholar

92 Ostroff RB, Nelson JC: Risperidone augmentation of selective serotonin reuptake inhibitors in major depression. J Clin Psychiatry 1999; 60:256–259CrossrefGoogle Scholar

93 Rapaport M, Gharabawi G, Canuso C, Lasser R, Loescher A: Preliminary results from the ARISe-RD (Risperidone Augmentation in Resistant Depression) trial. Proceedings of the 43rd Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); May 27–30, 2003; Boca Raton, Fla, p 122Google Scholar

94 Dunner DL, Amsterdam JD, Shelton RC, Hassman H, Rosenthal M, Romano SJ: Adjunctive ziprasidone in treatment-resistant depression: a pilot study. Proceedings of the 43rd Annual Meeting of the New Clinical Drug Evaluation Unit (NCDEU); May 27–30, 2003; Boca Raton, Fla, p 123Google Scholar

95 Papakosta GI, Peterson T, Nierenberg AA, Murakami JL, Alpert JE, Rosenbaum JF, Fava M: Ziprasidone augmentation in selective serotonin reuptake inhibitors for SSRI resistant major depressive disorder. J Clin Psychiatry 2004; 65:217–221CrossrefGoogle Scholar

96 Worthington JJ, Kinrys G, Wygant LE, Pollack MH: Aripiprazole as an augmentor of selective serotonin reuptake inhibitor in depression and anxiety disorder patients. Int Clin Psychopharmacol 2005; 20:9–11CrossrefGoogle Scholar

97 Adson DE, Kushner MG, Eiben KM, Schulz SC: Preliminary experience with adjunctive quetiapine in patients receiving selective serotonin reuptake inhibitors. Depress Anxiety 2004; 19:121–126CrossrefGoogle Scholar

98 Toalson P, Ahmed S, Hardy T, Kabinoff G: The metabolic syndrome in patients with severe mental illnesses. Prim Care Companion J Clin Psychiatry 2004; 6:152–158CrossrefGoogle Scholar

99 Newcomer JW, Nasrallah HA, Loebel AD: The Atypical Antipsychotic Therapy and Metabolic Issues National Survey: practice patterns and knowledge of psychiatrists. J Clin Psychopharmacol 2004; 24(5 suppl 1):S1–S6CrossrefGoogle Scholar

100 Menza MA, Kaufman KR, Castellanos A: Modafinil augmentation of antidepressant treatment in depression. J Clin Psychiatry 2000; 61:378–381CrossrefGoogle Scholar

101 DeBattista C, Doghramji K, Menza MA, Rosenthal MH, Fieve RR; Modafinil in Depression Study Group: Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: a preliminary double-blind, placebo-controlled study. J Clin Psychiatry 2003; 64:1057–1064CrossrefGoogle Scholar

102 DeBattista C, Lembke A, Solvason HB, Ghebremichael R, Poirier J: A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol 2004; 24:87–90CrossrefGoogle Scholar

103 Barbosa L, Berk M, Vorster M: A double-blind, randomized, placebo-controlled trial of augmentation with lamotrigine or placebo in patients concomitantly treated with fluoxetine for resistant major depressive episodes. J Clin Psychiatry 2003; 64:403–407CrossrefGoogle Scholar

104 Rocha FL, Hara C: Lamotrigine augmentation in unipolar depression. Int Clin Psychopharmacol 2003; 18:97–99CrossrefGoogle Scholar

105 Barbee JG, Jamhour NJ: Lamotrigine as an augmentation agent in treatment-resistant depression. J Clin Psychiatry 2002; 63:737–741CrossrefGoogle Scholar

106 Fatemi SH, Rapport DJ, Calabrese JR, Thuras P: Lamotrigine in rapid-cycling bipolar disorder. J Clin Psychiatry 1997; 58:522–527CrossrefGoogle Scholar

107 Sporn J, Ghaemi SN, Sambur MR, Rankin MA, Recht J, Sachs GS, Rosenbaum JF, Fava M: Pramipexole augmentation in the treatment of unipolar and bipolar depression: a retrospective chart review. Ann Clin Psychiatry 2000; 12:137–140CrossrefGoogle Scholar

108 Coppen A, Bailey J: Enhancement of the antidepressant action of fluoxetine by folic acid: a randomised, placebo controlled trial. J Affect Disord 2000; 60:121–130CrossrefGoogle Scholar

109 Taylor MJ, Carney SM, Goodwin GM, Geddes JR: Folate for depressive disorders: systematic review and meta-analysis of randomized controlled trials. J Psychopharmacol 2004; 18:251–256CrossrefGoogle Scholar

110 Rosenbaum JF, Fava M, Falk WE, Pollack MH, Cohen LS, Cohen BM, Zubenko GS: The antidepressant potential of oral S-adenosyl-l-methionine. Acta Psychiatr Scand 1990; 81:432–436CrossrefGoogle Scholar

111 Bressa GM: S-adenosyl-l-methionine (SAMe) as antidepressant: meta-analysis of clinical studies. Acta Neurol Scand Suppl 1994; 154:7–14CrossrefGoogle Scholar

112 Mischoulon D, Fava M: Role of S-adenosyl-L-methionine in the treatment of depression: a review of the evidence. Am J Clin Nutr 2002; 76: 1158S–1161SCrossrefGoogle Scholar

113 Nguyen M, Gregan A: S-adenosylmethionine and depression. Aust Fam Physician 2002; 31:339–343Google Scholar

114 Bodkin JA, Lasser RA, Wines JD Jr, Gardner DM, Baldessarini RJ: Combining serotonin reuptake inhibitors and bupropion in partial responders to antidepressant monotherapy. J Clin Psychiatry 1997; 58:137–145CrossrefGoogle Scholar

115 Landen M, Bjorling G, Agren H, Fahlen T: A randomized, double-blind, placebo-controlled trial of buspirone in combination with an SSRI in patients with treatment-refractory depression. J Clin Psychiatry 1998; 59:664–668CrossrefGoogle Scholar

116 Carpenter LL, Jocic Z, Hall JM, Rasmussen SA, Price LH: Mirtazapine augmentation in the treatment of refractory depression. J Clin Psychiatry 1999; 60:45–49CrossrefGoogle Scholar

117 Fava M: New approaches to the treatment of refractory depression. J Clin Psychiatry 2000; 61(suppl 1):26–32CrossrefGoogle Scholar

118 Thase ME, Ferguson JM, Lydiard RB, Wilcox CS: Citalopram treatment of paroxetine-intolerant depressed patients. Depress Anxiety 2002; 16: 128–133CrossrefGoogle Scholar

119 Thase ME, Feighner JP, Lydiard RB: Citalopram treatment of fluoxetine nonresponders. J Clin Psychiatry 2001; 62:683–687CrossrefGoogle Scholar

120 Thase ME, Blomgren SL, Birkett MA, Apter JT, Tepner RG: Fluoxetine treatment of patients with major depressive disorder who failed initial treatment with sertraline. J Clin Psychiatry 1997; 58:16–21CrossrefGoogle Scholar

121 Joffe RT, Levitt AJ, Sokolov ST, Young LT: Response to an open trial of a second SSRI in major depression. J Clin Psychiatry 1996; 57:114–115Google Scholar

122 Kornbluh R, Papakostas GI, Petersen T, Neault NB, Nierenberg AA, Rosenbaum JF, Fava M: A survey of prescribing preferences in the treatment of refractory depression: recent trends. Psychopharmacol Bull 2001; 35(3):150–156Google Scholar

123 Nierenberg AA, Feighner JP, Rudolph R, Cole JO, Sullivan J: Venlafaxine for treatment-resistant unipolar depression. J Clin Psychopharmacol 1994; 14:419–423CrossrefGoogle Scholar

124 Thase ME, Rush AJ, Howland RH, Kornstein SG, Kocsis JH, Gelenberg AJ, Schatzberg AF, Koran LM, Keller MB, Russell JM, Hirschfeld RM, LaVange LM, Klein DN, Fawcett J, Harrison W: Double-blind switch study of imipramine or sertraline treatment of antidepressant-resistant chronic depression. Arch Gen Psychiatry 2002; 59:233–239CrossrefGoogle Scholar

125 de Montigny C, Silverstone PH, Debonnel G, Blier P, Bakish D: Venlafaxine in treatment-resistant major depression: a Canadian multicenter, open-label trial. J Clin Psychopharmacol 1999; 19:401–406CrossrefGoogle Scholar

126 Mitchell PB, Schweitzer I, Burrows G, Johnson G, Polonowita A: Efficacy of venlafaxine and predictors of response in a prospective open-label study of patients with treatment-resistant major depression. J Clin Psychopharmacol 2000; 20:483–487CrossrefGoogle Scholar

127 Saiz-Ruiz J, Ibanez A, Diaz-Marsa M, Arias F, Padin J, Martin-Carrasco M, Montes JM, Ferrando L, Carrasco JL, Martin-Ballesteros E, Jorda L, Chamorro L: Efficacy of venlafaxine in major depression resistant to selective serotonin reuptake inhibitors. Prog Neuropsychopharmacol Biol Psychiatry 2002; 26:1129–1134CrossrefGoogle Scholar

128 Fava M, Dunner DL, Greist JH, Preskorn SH, Trivedi MH, Zajecka J, Cohen M: Efficacy and safety of mirtazapine in major depressive disorder patients after SSRI treatment failure: an open-label trial. J Clin Psychiatry 2001; 62:413–420CrossrefGoogle Scholar

129 Stern WC, Harto-Truax N, Bauer N: Efficacy of bupropion in tricyclic-resistant or intolerant patients. J Clin Psychiatry 1983; 44(5 Pt 2):148–152Google Scholar

130 Weintraub D: Nortriptyline in geriatric depression resistant to serotonin reuptake inhibitors: case series. J Geriatr Psychiatry Neurol 2001; 14:28–32CrossrefGoogle Scholar

131 Nierenberg AA, Papakostas GI, Petersen T, Kelly KE, Iacoviello BM, Worthington JJ, Tedlow J, Alpert JE, Fava M: Nortriptyline for treatment-resistant depression. J Clin Psychiatry 2003; 64:35–39CrossrefGoogle Scholar

132 Thase ME, Rush AJ, Howland RH, Kornstein SG, Kocsis JH, Gelenberg AJ, Schatzberg AF, Koran LM, Keller MB, Russell JM, Hirschfeld RM, LaVange LM, Klein DN, Fawcett J, Harrison W: Double-blind switch study of imipramine or sertraline treatment of antidepressant-resistant chronic depression. Arch Gen Psychiatry 2002; 59:233–239CrossrefGoogle Scholar

133 McGrath PJ, Stewart JW, Harrison W, Quitkin FM: Treatment of tricyclic refractory depression with a monoamine oxidase inhibitor antidepressant. Psychopharmacol Bull 1987; 23(1):169–172Google Scholar

134 Thase ME, Mallinger AG, McKnight D, Himmelhoch JM: Treatment of imipramine-resistant recurrent depression, IV: a double-blind crossover study of tranylcypromine for anergic bipolar depression. Am J Psychiatry 1992; 149:195–198CrossrefGoogle Scholar

135 Fava M, McGrath PJ, Sheu WP; Reboxetine Study Group: Switching to reboxetine: an efficacy and safety study in patients with major depressive disorder unresponsive to fluoxetine. J Clin Psychopharmacol 2003; 23:365–369CrossrefGoogle Scholar

136 Fava M, Rush AJ, Trivedi MH, Nierenberg AA, Thase ME, Sackeim HA, Quitkin FM, Wisniewski S, Lavori PW, Rosenbaum JF, Kupfer DJ: Background and rationale for the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study. Psychiatr Clin North Am 2003; 26:457–494CrossrefGoogle Scholar

137 Rush AJ, Fava M, Wisniewski SR, Lavori PW, Trivedi MH, Sackeim HA, Thase ME, Nierenberg AA, Quitkin FM, Kashner TM, Kupfer DJ, Rosenbaum JF, Alpert J., Stewart JW, McGrath PJ, Biggs MM, Shores-Wilson K, Lebowitz BD, Ritz L, Niederehe G; STAR*D Investigators Group: Sequenced Treatment Alternatives to Relieve Depression (STAR*D): rationale and design. Controlled Clin Trials 2004; 25:119–142CrossrefGoogle Scholar

138 Lavori PW, Rush JA, Wisniewski S, Fava M, Kupfer DJ, Nierenberg AA, Quitkin F, Thase ME, Trivedi MH: Strengthening clinical effectiveness trials: equipoise-stratified randomization. Biol Psychiatry 2001; 50:792–801CrossrefGoogle Scholar