120

  • July 2020
  • PDF

This document was uploaded by user and they confirmed that they have the permission to share it. If you are author or own the copyright of this book, please report to us by using this DMCA report form. Report DMCA


Overview

Download & View 120 as PDF for free.

More details

  • Words: 4,583
  • Pages: 6
Use of Atypical Antipsychotics for Treatment-Resistant Major Depressive Disorder George I. Papakostas, MD, and Richard C. Shelton, MD

Corresponding author George I. Papakostas, MD Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WACC #812, Boston, MA 02114, USA. E-mail: [email protected] Current Psychiatry Reports 2008, 10:481–486 Current Medicine Group LLC ISSN 1523-3812 Copyright © 2008 by Current Medicine Group LLC

Despite the progressive increase in the number of pharmacologic agents with potential antidepressant activity, many patients suffering from major depressive disorder (MDD) continue to be symptomatic. Clearly, an urgent need exists to develop safer, better tolerated, and more effective treatments for MDD. Use of atypical antipsychotic agents as adjunctive treatment for treatment-resistant MDD (TRD) represents one such effort toward novel pharmacotherapy development. Atypical antipsychotic agents have been hypothesized to be beneficial in treating mood disorders, including TRD, as a result of their complex mechanisms of action. After an initial series of positive case reports, series, and small clinical trials, subsequent larger-scale projects have yielded conflicting results. However, more recently, larger-scale clinical trials have supported the effectiveness of at least some of these medications. This review summarizes the existing data regarding the effectiveness of these medications in treating TRD, including biochemical rationale and clinical data.

Introduction Despite the progressive increase in the number of available antidepressants, many patients suffering from depression continue to be symptomatic. For example, in the fi rst level of the Sequenced Treatment Alternatives to Relieve Depression (STAR*D) study, only about 30% of patients had achieved remission of their depressive episode after up to 12 weeks of therapy with citalopram [1]. Moreover, it was recently reported that as many as half of all patients enrolled in two academic-based depression specialty clin-

ics did not achieve remission despite receiving a series of adequate trials of antidepressant therapies [2]. To complicate matters further, residual symptoms among remitters are common and associated with poorer psychosocial functioning [3] and increased relapse rates [4]. Yet little consensus exists among psychiatrists regarding optimizing treatment for patients with incomplete response. Generally, management of treatment-resistant major depressive disorder (TRD) can involve pharmacologic and nonpharmacologic interventions, such as the use of cognitive-behavioral psychotherapy [5••], electroconvulsive therapy [6], vagus nerve stimulation [7], and transcranial magnetic stimulation [8]. Pharmacologic interventions can involve increasing the dose of an antidepressant [9], switching to a second antidepressant [10–13], or using adjunctive pharmacotherapeutic strategies. When a second antidepressant is added to an existing antidepressant, this is termed combination pharmacotherapy. When a nonantidepressant agent is added, this is termed augmentation treatment. Combination strategies can involve adding bupropion [14], mirtazapine [13,15], and tricyclic antidepressants [9,16] to an antidepressant treatment regimen. Augmentation strategies can involve adding lithium [9,17,18], triiodothyronine [18], buspirone [14], pindolol [19], omega-3 fatty acids [20], dopaminergic agents [21,22], folates, or S-adenosyl methionine [23,24] to an antidepressant treatment regimen. Nevertheless, as described previously, despite the broad armamentarium available to clinicians for managing TRD, many patients remain symptomatic despite several adequate pharmacologic and nonpharmacologic trials. Clearly, there is an urgent need to develop safer, better tolerated, and more effective treatments for major depressive disorder. Using atypical antipsychotic agents as adjunctive treatment for TRD represents one such effort toward novel pharmacotherapy development.

Mechanisms of Action Atypical antipsychotics are a heterogeneous group, each with a distinct and complex set of receptor affi nities involving dopaminergic and serotoninergic receptors and

482

Mood Disorders

various effects on noradrenergic, histaminergic, and cholinergic systems. As a result, the receptor-binding profi le of the atypical neuroleptics differs substantially from that of other typical agents. This difference may provide clinical advantages to the atypical antipsychotic agents in therapeutic areas other than in schizophrenia. Specifically, similar to the typical antipsychotic agents, atypical antipsychotics act as dopamine-2 (D2)–receptor antagonists [25]. However, unlike typical antipsychotic agents, atypical antipsychotics, including olanzapine; quetiapine; risperidone; ziprasidone [25]; and, to a much lesser extent, aripiprazole [26], act as serotonin-2 (5-HT2)–receptor antagonists. Ziprasidone [25] and aripiprazole [26] also act as serotonin-1A (5-HT1A)–receptor agonists, whereas ziprasidone and risperidone [25] act as serotonin-1D (5-HT1D) agonists. Antagonism of 5-HT1D, a presynaptic autoreceptor inhibiting serotonin release, indicates potential efficacy in major depression [27], whereas selective 5-HT1A–receptor partial agonists, such as gepirone [28], have shown antidepressant effects in clinical trials. Similarly, 5-HT2 –receptor antagonists, such as trazodone [29] and nefazodone [30], also have shown antidepressant effects. Aripiprazole and ziprasidone have other unique neurochemical properties that distinguish them from the other atypicals. Aripiprazole is a potent dopamine D2-receptor partial agonist, blocking D2 receptors under hyperdopaminergic conditions while acting as a D2 agonist under hypodopaminergic conditions [31–35]. Ziprasidone has been shown to inhibit the neuronal uptake of 5-HT and norepinephrine in a way comparable to the antidepressant imipramine [36].

Clinical Effects of Atypical Antipsychotics in Treatment-Resistant Depression The neurochemical effects described previously led to the testing, initially, of atypical antipsychotics for cognitive and mood symptoms associated with schizophrenia. This subsequently led to the evaluation of atypicals for mood symptoms in TRD and bipolar depression. This section reviews the data supporting effects of these drugs individually and focuses primarily on controlled clinical trials.

Olanzapine The fi rst controlled test of the effects of an atypical in TRD was a small study with olanzapine that compared the effects of olanzapine alone (ie, olanzapine + placebo), olanzapine plus fluoxetine, and fluoxetine alone in treating TRD [37]. All participants had previously failed adequate trials of a selective serotonin reuptake inhibitor (SSRI) and a non-SSRI antidepressant, as well as a prospective treatment period with fluoxetine alone (up to 60 mg/d). Nonresponders (n = 28) to this run-in treatment then were randomly assigned to one of the three condi-

tions in a double-blind fashion. The average maximum doses in the double-blind period were as follows: 12.5 mg/d for olanzapine alone; 52 mg/d for fluoxetine alone; and 13.5 and 52 mg/d, respectively, for the olanzapine and fluoxetine combination. The continuation of fluoxetine for this period yielded essentially no further improvement. Olanzapine administered alone achieved a modest benefit over fluoxetine, whereas the olanzapine plus fluoxetine combination resulted in robust and significantly greater improvement in depressive symptoms than either monotherapy. Remission, defi ned as a 17-item Hamilton Depression Rating Scale (HAM-D) score of 7 or less, was achieved in 60% of patients receiving the combination, 25% receiving olanzapine alone, and 20% receiving fluoxetine alone. Two large-scale clinical trials testing the effects of the olanzapine plus fluoxetine combination were undertaken next. In the fi rst, 500 patients were prospectively treated with nortriptyline; responders were excluded, leading into the double-blind phase [38]. Patients then were randomly assigned to one of four treatment groups for 8 weeks: olanzapine plus placebo, fluoxetine plus placebo, a combination of olanzapine plus fluoxetine, or a continuation of nortriptyline. Olanzapine was dosed in groups at 6 to 12 mg/d and fluoxetine at 25 to 50 mg/d. The combination of olanzapine and fluoxetine produced a rapid effect relative to the other groups. However, the groups did not differ at end point. Several design and execution factors may have affected these results. Patients had failed only one adequate trial of an SSRI before entry. The doses of olanzapine and fluoxetine were lower than in the previous study (mean maximal dose of olanzapine, 8.5 mg/d; mean maximal dose for fluoxetine, 36.5 mg/d). Furthermore, the use of nortriptyline may have been problematic. Although the mean dose in the initial phase was respectable (104.6 mg/d), many patients achieve their maximum dose relatively late in phase 1. Given the delay until maximum response to antidepressant treatment, the response to the dose achieved before phase 2 may have been experienced after the start of this period. A second study [39] using venlafaxine as a comparator suffered from many of the same design problems and is not reviewed in detail here. Despite these discouraging results, two subsequent large-scale trials undertaken [40] were of similar design to the original study by Shelton et al. [37]. In each, patients with a single drug failure were treated prospectively with fluoxetine. Those who did not respond were randomly assigned to continuation fluoxetine (+ placebo), olanzapine (+ placebo), or the olanzapine and fluoxetine combination. In one (n = 638), combined olanzapine/fluoxetine produced no greater effect than either monotherapy. However, the fluoxetine monotherapy condition produced a robust effect by study’s end, indicating another failed trial. However, the second project (n = 675) confi rmed the more robust effect of combined olanzapine/fluoxetine in treatment-resistant unipolar major depression compared

Atypical Antipsychotics for Treatment-Resistant Major Depressive Disorder Papakostas and Shelton

with olanzapine or fluoxetine monotherapy. The pooled analysis of both studies also showed a significantly superior effect of the combination. Meanwhile, a successful trial of the combination was tested in bipolar I depression (non–treatment-resistant), leading to US Food and Drug Administration (FDA) approval for this indication [41].

Risperidone The fi rst published report of the effectiveness of an atypical antipsychotic in TRD was with risperidone [42]. In this case series, eight patients who failed to respond to an adequate trial of an SSRI were given risperidone in an open fashion, and all experienced a rapid and robust effect. However, there were no controlled clinical trial data with risperidone in TRD until recently. In one study [43], 489 patients with one to three prior documented failures were treated prospectively with citalopram, 20 to 60 mg/d. Those who experienced greater than 50% improvement were excluded. The remainder (n = 390) were given 4 to 6 weeks of open-label risperidone, 0.25 to 2.0 mg/d, in combination with citalopram. Patients with 17-item HAM-D scores of 7 or less or a Clinical Global Impression Severity Scale score of 2 or less (ie, “much improved” or “very much improved”) were randomized to risperidone or placebo augmentation (n = 243 remitters, 241 randomized). The primary end point, time to relapse, did not differ between groups. The median time to relapse was 102 days for risperidone plus citalopram and 85 days for placebo plus citalopram. Relapse rates were 53.3% with the combination and 54.6% with continuation of citalopram alone. These data indicate that continuation of risperidone plus citalopram was no more effective for preventing relapse than citalopram alone. In a second controlled trial [44], 463 depressed patients were treated with an optimized trial of an antidepressant. The 274 patients who did not respond were randomized to receive risperidone, 1 to 2 mg/d, or placebo combined with the initial antidepressant for 6 weeks. Mean HAM-D scores dropped from 24.2 to 15.2 in the risperidone group and from 24.6 to 17.5 in the placebo group, a statistically significant difference. In a third controlled trial, Keitner et al. [45] studied 97 outpatients who met the DSM-IV criteria for a unipolar, nonpsychotic major depressive episode and failed to respond or only partially responded to at least a 5-week trial of an adequate dose of antidepressant monotherapy. Patients were randomized to receive adjunctive risperidone or placebo for a total of 4 weeks of treatment. The odds of remitting were significantly better for patients treated with adjunctive risperidone than placebo (52% vs 24%, respectively).

Quetiapine Quetiapine has received FDA approval for treating bipolar depression as monotherapy. However, there are now

483

three controlled trials in unipolar TRD. One study (n = 40) evaluated the effects of quetiapine for SSRI/(SNRI) partial response [46]. In this 8-week, double-blind, placebo-controlled study, patients were randomized (2:1 ratio) to receive quetiapine, 200 to 400 mg/d, or placebo along with their previous SSRI/serotonin–norepinephrine reuptake inhibitor (SNRI). Twenty-one of 26 quetiapinetreated patients (80.8%) completed the study, compared with 11 of 14 (78.6%) in the placebo group. At the end of 8 weeks, the quetiapine group had significantly lower 17-item HAM-D scores than the placebo group (8.3 vs 14.7) plus higher response (67% vs 27%) and remission rates (43% vs 15%). In a separate study, McIntyre et al. [47] studied 58 patients who did not experience sufficient symptom improvement after adequate treatment with an SSRI or venlafaxine. Patients were randomized in double-blind fashion to receive adjunctive treatment with quetiapine or placebo for 8 weeks. Eighteen of 29 quetiapine-treated and 16 of 29 placebo-treated patients completed the study. A greater resolution of depressive symptoms was reported among patients treated with adjunctive quetiapine than placebo (mean difference in the reduction in HAM-D scores during treatment was 5.7 points in favor of quetiapine, P < 0.01). Finally, in a third trial, Khullar et al. [48] studied 15 outpatients with TRD (to SSRIs or SNRIs) who underwent augmentation with quetiapine or placebo for 8 weeks. Quetiapine was more effective in reducing depressive symptoms than placebo, as 37% of patients remitted after adjunctive treatment with quetiapine, whereas none remitted after treatment with placebo. Additional largerscale studies of quetiapine therapy for TRD recently have been completed by the manufacturer, although results are not yet available in the published literature.

Ziprasidone An initial open trial supported the effects of ziprasidone in TRD. In this study, 20 patients who had an incomplete response to a trial of an SSRI were treated with ziprasidone in an open fashion [49]. Thirteen (65%) completed the trial; of this group, eight (61.5%) were responders, as defi ned by a 50% improvement in 17-item HAM-D score. Of all the patients randomized, 10 were responders (50%), and 5 remitted (25%). In a second randomized, open trial, adult outpatients who had not responded to a prior trial with an antidepressant were treated prospectively with sertraline, 100 to 200 mg/d, for 6 weeks [50]. Those who failed to achieve a 30% or greater change in Montgomery-Asberg Depression Rating Scale score were randomly assigned to continuation with sertraline; sertraline plus ziprasidone, 80 mg/d (mean dose, 78 mg/d); or sertraline plus ziprasidone, 160 mg/d (mean dose, 129.9 mg/d) for 8 weeks. The mean change from baseline was greater in the two ziprasidone

484

Mood Disorders

aripiprazole, 1.7% for placebo; P > 0.05). The results of a separate study of identical design also demonstrated greater remission rates for adjunctive aripiprazole than placebo-treated patients [53••]. The results of these two trials led to FDA approval of a new indication for aripiprazole as adjunctive treatment to antidepressants for antidepressant-resistant major depressive disorder (Fig. 1). This was the fi rst approval for an adjunctive treatment in major depressive disorder and the fi rst FDA approval for the use of any medication for TRD.

Conclusions

Figure 1. Aripiprazole augmentation for antidepressant-resistant major depressive disorder: results of two identical randomized, double-blind, phase 3 trials (P < 0.05, aripiprazole augmentation vs antidepressant monotherapy for both trials).

augmentation groups (80 mg/d: -5.98 points; 160 mg/d: -8.27 points) than in the sertraline monotherapy group (-4.45 points), although this did not achieve statistical significance. Respective response rates were 19%, 32%, and 10%, and remission rates were 5%, 21%, and 5%. Although these results suggest possible benefit, they did not conclusively support ziprasidone augmentation.

Aripiprazole Until recently, aripiprazole had received relatively little attention in this area. In one open-label trial, 12 patients who had failed to respond sufficiently to an adequate trial of an SSRI were treated with adjunctive aripiprazole in an open fashion for 8 weeks [51]. Nine patients (75%) patients completed the trial; five (55.6%) achieved response (mean change in HAM-D score > 50%), whereas seven of the total group (58.3%) responded. More recently, results of two positive, double-blind, placebo-controlled trials investigating the use of adjunctive aripiprazole in major depressive disorder were published. In the fi rst such study, Berman and colleagues [52••] focused on the use of aripiprazole augmentation for patients resistant to up to three “retrospective” (historical) antidepressant trials. To “confi rm” treatment resistance, those patients underwent an 8-week, openlabel trial with an SSRI (fluoxetine, sertraline, paroxetine, or escitalopram) or an SNRI (venlafaxine). The patients who made insufficient symptom improvement had aripiprazole or placebo added to their SSRI or SNRI regimen under double-blind conditions for a total of 6 weeks. A statistically significant difference in remission rates was also observed, with 26% remission for aripiprazole, compared with 15.7% remission for placebo (P < 0.05). This study also reported relatively low rates of discontinuation due to intolerance in the two treatment groups (2% for

TRD is a common clinical occurrence, and its management poses a formidable clinical challenge for clinicians and patients alike. Despite several pharmacologic and nonpharmacologic treatment options for TRD, many patients remain symptomatic. Thus, there is an urgent need to develop novel treatments for TRD. From the evidence available to date, it appears that augmenting antidepressants with atypical antipsychotics (specifically aripiprazole, olanzapine, quetiapine, and risperidone) can be effective in some patients, at least during the acute phase of treatment. However, the long-term efficacy, tolerability, and safety of this treatment are not yet understood. In particular, the risk of metabolic (weight gain and hyperlipidemia or dyslipidemia), endocrine (hyperprolactinemia), cardiac (QTc prolongation and arrhythmogenesis), and central nervous system (akathisia, parkinsonism, tardive dyskinesia, neuroleptic malignant syndrome) adverse events during treatment with these agents in major depressive disorder needs to be better quantified. Further research also is required into how this compares with other augmentation strategies and other strategies for addressing TRD.

Acknowledgments This work was supported in part by National Institute of Mental Health grant K24 MH01741 (to Dr. Shelton).

Disclosures Dr. Papakostas has received research support from Bristol-Myers Squibb, Forest Pharmaceuticals, the National Institute of Mental Health, Pamlab, Pfi zer, and Precision Human BioLaboratories; has served as a consultant for Bristol-Myers Squibb, Eli Lilly and Company, Evotec, GlaxoSmithKline, Inflabloc Pharmaceuticals, Jazz Pharmaceuticals, Pamlab, Pfi zer, Pierre Fabre Medicament, Shire Pharmaceuticals, and Wyeth; has received honoraria from Bristol-Myers Squibb, Eli Lilly and Company, Evotec, GlaxoSmithKline, Inflabloc Pharmaceuticals, Jazz Pharmaceuticals, H. Lundbeck A/S, Pamlab, Pfi zer, Pierre Fabre Medicament, Shire Pharmaceuticals, Titan Pharmaceuticals, and Wyeth; and has served on the speakers’ bureau for Bristol-Myers Squibb and Pfi zer.

Atypical Antipsychotics for Treatment-Resistant Major Depressive Disorder Papakostas and Shelton

Dr. Shelton has received grant/research support from Eli Lilly and Company, GlaxoSmithKline, Janssen Pharmaceutica, Pfi zer, Sanofi-Aventis, Wyeth-Ayerst Laboratories, AstraZeneca Pharmaceuticals, and Abbott Laboratories; has served as a paid consultant for Pfi zer, Janssen Pharmaceutica, and Sierra Neuropharmaceuticals; and has served on the speakers’ bureau for Bristol-Myers Squibb, Eli Lilly and Company, Janssen Pharmaceutica, Pfi zer, GlaxoSmithKline, Wyeth-Ayerst Laboratories, and Abbott Laboratories.

13.

14.

15.

16.

17.

References and Recommended Reading Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

18.

19. 1.

Trivedi MH, Rush AJ, Wisniewski SR, et al.: Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry 2006, 163:28–40. 2. Petersen T, Papakostas GI, Posternak MA, et al.: Empirical testing of two models for staging antidepressant treatment resistance. J Clin Psychopharmacol 2005, 25:336–341. 3. Papakostas GI, Petersen T, Denninger JW, et al.: Psychosocial functioning during the treatment of major depressive disorder with fluoxetine. J Clin Psychopharmacol 2004, 24:507–511. 4. Paykel ES, Ramana R, Cooper Z, et al.: Residual symptoms after partial remission: an important outcome in depression. Psychol Med 1995, 25:1171–1180. 5.•• Thase ME, Friedman ES, Biggs MM, et al.: Cognitive therapy versus medication in augmentation and switch strategies as second-step treatments: a STAR*D report. Am J Psychiatry 2007, 164:739–752. Large, randomized, double-blind, placebo-controlled trials confi rming the fi ndings of Shelton et al. [37]. 6. UK ECT Review Group: Efficacy and safety of electroconvulsive therapy in depressive disorders: a systematic review and meta-analysis. Lancet 2003, 361:799–808. 7. Rush AJ, Marangell LB, Sackeim HA, et al.: Vagus nerve stimulation for treatment-resistant depression: a randomized, controlled acute phase trial. Biol Psychiatry 2005, 58:347–354. 8. O’Reardon JP, Solvason HB, Janicak PG, et al.: Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry 2007, 62:1208–1216. 9. Fava M, Alpert J, Nierenberg A, et al.: Double-blind study of high-dose fluoxetine versus lithium or desipramine augmentation of fluoxetine in partial responders and nonresponders to fluoxetine. J Clin Psychopharmacol 2002, 22:379–387. 10. Papakostas GI, Fava M, Thase ME: Treatment of SSRI-resistant depression: a meta-analysis comparing within- versus across-class switches. Biol Psychiatry 2008, 63:699–704. 11. Rush AJ, Trivedi MH, Wisniewski SR, et al.: BupropionSR, sertraline, or venlafaxine-XR after failure of SSRIs for depression. N Engl J Med 2006, 354:1231–1242. 12. Fava M, Rush AJ, Wisniewski SR, et al.: A comparison of mirtazapine and nortriptyline following two consecutive failed medication treatments for depressed outpatients: a STAR*D report. Am J Psychiatry 2006, 163:1161–1172.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

485

McGrath PJ, Stewart JW, Fava M, et al.: Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: a STAR*D report. Am J Psychiatry 2006, 163:1531–1541. Trivedi MH, Fava M, Wisniewski SR, et al.: Medication augmentation after the failure of SSRIs for depression. N Engl J Med 2006, 354:1243–1252. Carpenter LL, Yasmin S, Price LH: A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biol Psychiatry 2002, 51:183–188. Nelson JC, Mazure CM, Jatlow PI, et al.: Combining norepinephrine and serotonin reuptake inhibition mechanisms for treatment of depression: a double-blind, randomized study. Biol Psychiatry 2004, 55:296–300. Nierenberg AA, Papakostas GI, Petersen T, et al.: Lithium augmentation of nortriptyline for subjects resistant to multiple antidepressants. J Clin Psychopharmacol 2003, 23:92–95. Nierenberg AA, Fava M, Trivedi MH, et al.: A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: a STAR*D report. Am J Psychiatry 2006, 163:1519–1530. Ballesteros J, Callado LF: Effectiveness of pindolol plus serotonin uptake inhibitors in depression: a meta-analysis of early and late outcomes from randomised controlled trials. J Affect Disord 2004, 79:137–147. Peet M, Horrobin DF: A dose-ranging study of the effects of ethyl-eicosapentaenoate in patients with ongoing depression despite apparently adequate treatment with standard drugs. Arch Gen Psychiatry 2002, 59:913–919. Patkar AA, Masand PS, Pae CU, et al.: A randomized, double-blind, placebo-controlled trial of augmentation with an extended release formulation of methylphenidate in outpatients with treatment-resistant depression. J Clin Psychopharmacol 2006, 26:653–656. Ravindran AV, Kennedy SH, O’Donovan MC, et al.: Osmotic-release oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: results of a double-blind, randomized, placebocontrolled trial. J Clin Psychiatry 2008, 69:87–94. Alpert JE, Mischoulon D, Rubenstein GE, et al.: Folinic acid (Leucovorin) as an adjunctive treatment for SSRIrefractory depression. Ann Clin Psychiatry 2002, 14:33–38. Alpert JE, Papakostas G, Mischoulon D, et al.: S-adenosylL-methionine (SAMe) as an adjunct for resistant major depressive disorder: an open trial following partial or nonresponse to selective serotonin reuptake inhibitors or venlafaxine. J Clin Psychopharmacol 2004, 24:661–664. Richelson E, Souder T: Binding of antipsychotic drugs to human brain receptors focus on newer generation compounds. Life Sci 2000, 68:29–39. Shapiro DA, Renock S, Arrington E, et al.: Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 2003, 28:1400–1411. Stahl SM, Shayegan DK: The psychopharmacology of ziprasidone: receptor-binding properties and real-world psychiatric practice. J Clin Psychiatry 2003, 64(Suppl 19):6–12. Feiger AD, Heiser JF, Shrivastava RK, et al.: Gepirone extended-release: new evidence for efficacy in the treatment of major depressive disorder. J Clin Psychiatry 2003, 64:243–249. Cunningham LA, Borison RL, Carman JS, et al.: A comparison of venlafaxine, trazodone, and placebo in major depression. J Clin Psychopharmacol 1994, 14:99–106. Feighner J, Targum SD, Bennett ME, et al.: A double-blind, placebo-controlled trial of nefazodone in the treatment of patients hospitalized for major depression. J Clin Psychiatry 1998, 59:246–253.

486

Mood Disorders

31.

Kikuchi T, Tottori K, Uwahodo Y, et al.: 7-(4-[4-[2,3-Dichlorophenyl)-1-piperazinyl]butyloxy)-3,4dihydro-2(1H)-quinolinone (OPC-14957), a new putative antipsychotic drug with both presynaptic dopamine autoreceptor agonistic activity and postsynaptic D2 receptor antagonistic activity. J Pharmacol Exp Ther 1995, 274:329–336. Inoue T, Domae M, Yamada K, Furukawa T: Effects of the novel antipsychotic agent 7-(4-[4-(2,3-dichlorophenyl)1-piperazinyl] butyloxy)-3,4-dihydro-2(1H)-quinolinone (OPC-14597) on prolactin release from the rat anterior pituitary gland. J Pharmacol Exp Ther 1996, 277:137–143. Matsubayashi H, Amano T, Sasa M: Inhibition by aripiprazole of dopaminergic inputs to striatal neurons from substantia nigra. Psychopharmacology (Berl) 1999, 146:139–143. Lawler CP, Prioleau C, Lewis MM, et al.: Interactions of the novel antipsychotic aripiprazole (OPC-14597) with dopamine and serotonin receptor subtypes. Neuropsychopharmacology 1999, 20:612–627. Burris KD, Molski TF, Xu C, et al.: Aripiprazole, a novel antipsychotic, is a high-affi nity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther 2002, 302:381–389. Schmidt AW, Lebel A, Howard HR Jr, Zom SH: Ziprasidone: a novel antipsychotic agent with a unique human receptor binding profile. Eur J Pharmacol 2001, 425:197–201. Shelton RC, Tollefson GD, Tohen M, et al.: A novel augmentation strategy for treating resistant major depression. Am J Psychiatry 2001, 158:131–134. Shelton RC, Williamson DJ, Corya SA, et al.: Olanzapine/ fluoxetine combination for treatment-resistant depression: a controlled study of SSRI and nortriptyline resistance. J Clin Psychiatry 2005, 66:1289–1297. Corya SA, Williamson D, Sanger TM, et al.: A randomized, double-blind comparison of olanzapine/fluoxetine combination, olanzapine, fluoxetine, and venlafaxine in treatment-resistant depression. Depress Anxiety 2006, 23:364–372. Thase ME, Corya SA, Osuntokun O, et al.: A randomized, double-blind comparison of olanzapine/fluoxetine combination, olanzapine, and fluoxetine in treatmentresistant major depressive disorder. J Clin Psychiatry 2007, 68:224–236. Tohen M, Vieta E, Calabrese J, et al.: Efficacy of olanzapine and olanzapine-fluoxetine combination in the treatment of bipolar I depression. Arch Gen Psychiatry 2003, 60:1079–1088. Ostroff RB, Nelson JC: Risperidone augmentation of selective serotonin reuptake inhibitors in major depression. J Clin Psychiatry 1999, 60:256–259. Rapaport MH, Gharabawi GM, Canuso CM, et al.: Effects of risperidone augmentation in patients with treatmentresistant depression: results of open-label treatment followed by double-blind continuation. Neuropsychopharmacology 2006, 31:2505–2513.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

Mahmoud RA, Pandina GJ, Turkoz I, et al.: Risperidone for treatment-refractory major depressive disorder: a randomized trial. Ann Intern Med 2007, 147:593–602. 45. Keitner GI, Garlow SJ, Ryan CE: Risperidone augmentation for patients with difficult-to-treat major depression [poster]. Presented at the 159th Annual Meeting of the American Psychiatric Association. Toronto, Canada; May 20–26, 2006. 46. Mattingly G, Ilivicky H, Canale J, Anderson R: Quetiapine augmentation for treatment-resistant depression [poster]. Presented at the 159th Annual Meeting of the American Psychiatric Association. Toronto, Canada; May 20–26, 2006. 47. McIntyre A, Gendron A, McIntyre A: Quetiapine adjunct to selective serotonin reuptake inhibitors or venlafaxine in patients with major depression, comorbid anxiety, and residual depressive symptoms: a randomized, placebo-controlled pilot study. Depress Anxiety 2007, 24:487–494. 48. Khullar A, Chokka P, Fullerton D, et al.: Quetiapine as treatment of non-psychotic unipolar depression with residual symptoms: double blind, randomized, placebo controlled study [poster]. Presented at the 159th Annual Meeting of the American Psychiatric Association. Toronto, Canada; May 20–26, 2006. 49. Papakostas GI, Petersen TJ, Nierenberg AA, et al.: Ziprasidone augmentation of selective serotonin reuptake inhibitors (SSRIs) for SSRI-resistant major depressive disorder. J Clin Psychiatry 2004, 65:217–221. 50. Dunner DL, Amsterdam JD, Shelton RC, et al.: Efficacy and tolerability of adjunctive ziprasidone in treatment-resistant depression: a randomized, open-label, pilot study. J Clin Psychiatry 2007, 68:1071–1077. 51. Papakostas GI, Petersen TJ, Kinrys G, et al.: Aripiprazole augmentation of selective serotonin reuptake inhibitors for treatment-resistant major depressive disorder. J Clin Psychiatry 2005, 66:1326–1330. 52.•• Berman RM, Marcus RN, Swanink R, et al.: The efficacy and safety of aripiprazole as adjunctive therapy in major depressive disorder: a multicenter, randomized, doubleblind, placebo-controlled study. J Clin Psychiatry 2007, 68:843–853. One of two identical but separate randomized, double-blind, placebo-controlled trials of aripiprazole in TRD. FDA approval of aripiprazole as adjunctive treatment for TRD was based on these two trials. 53.•• Marcus RN, McQuade RD, Carson WH, et al.: The efficacy and safety of aripiprazole as adjunctive therapy in major depressive disorder: a second multicenter, randomized, double-blind, placebo-controlled study. J Clin Psychopharmacol 2008, 28:156–165. The other of two identical but separate randomized, double-blind, placebo-controlled trials of aripiprazole in TRD. FDA approval of aripiprazole as adjunctive treatment for TRD was based on these two trials.

Related Documents

120
November 2019 43
120
November 2019 38
120
November 2019 36
120
July 2020 23
120
June 2020 21
120
November 2019 39