Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Oct 6.
Published in final edited form as: J Clin Psychiatry. 2010 Sep 21;72(1):75–80. doi: 10.4088/JCP.09m05258gre

Impact of Second-Generation Antipsychotics and Perphenazine on Depressive Symptoms in a Randomized Trial of Treatment for Chronic Schizophrenia

Donald E Addington 1, Somaia Mohamed 1, Robert A Rosenheck 1, Sonia M Davis 1, Thomas Scott Stroup 1, Joseph P McEvoy 1, Marvin S Swartz 1, Jeffrey A Lieberman 1
PMCID: PMC5052810  NIHMSID: NIHMS816830  PMID: 20868641

Abstract

Background

According to the American Psychiatric Association Clinical Practice Guidelines for schizophrenia, second-generation antipsychotics may be specifically indicated for the treatment of depression in schizophrenia. We examined the impact of these medications on symptoms of depression using the data from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE), conducted between January 2001 and December 2004.

Method

Patients with DSM-IV–defined schizophrenia (N = 1,460) were assigned to treatment with a first-generation antipsychotic (perphenazine) or one of 4 second-generation drugs (olanzapine, quetiapine, risperidone, or ziprasidone) and followed for up to 18 months (phase 1). Patients with tardive dyskinesia were excluded from the randomization that included perphenazine. Depression was assessed with the Calgary Depression Scale for Schizophrenia (CDSS). Mixed models were used to evaluate group differences during treatment with the initially assigned drug. An interaction analysis evaluated differences in drug response by whether patients had a baseline score on the CDSS of ≥ 6, indicative of a current major depressive episode (MDE).

Results

There were no significant differences between treatment groups on phase 1 analysis, although there was a significant improvement in depression across all treatments. A significant interaction was found between treatment and experiencing an MDE at baseline (P = .05), and further paired comparisons suggested that quetiapine was superior to risperidone among patients who were in an MDE at baseline (P = .0056).

Conclusions

We found no differences between any second-generation antipsychotic and the first-generation antipsychotic perphenazine and no support for clinical practice recommendations, but we did detect a signal indicating a small potential difference favoring quetiapine over risperidone only in patients with an MDE at baseline.


Since their introduction in the 1990s, second-generation antipsychotics (SGAs) have become the drugs of choice in the treatment of schizophrenia, despite a lack of conclusive evidence of superior efficacy as assessed by measures of general psychopathology.15 One meta-analysis has, however, suggested that not all SGAs are equivalent.6

Depression is a common symptom over the course of schizophrenia.7,8 It is a predictor of attempted suicide and suicide9,10 and is an important determinant of quality of life.11 When depressive symptoms meet the syndromal criteria for major depressive disorder, antidepressants have been suggested as adjunctive treatment to antipsychotics.12

Early suggestions that first-generation antipsychotics (FGAs) might have antidepressant properties notwithstanding,13 depression has been identified as a potential treatment target for which SGAs were suggested to have a differential effect in comparison to FGAs.1417 The American Psychiatric Association Clinical Practice Guidelines assigned a level II evidence for a recommendation on the use of SGAs for the treatment of depression in schizophrenia not associated with relapse.12 While some studies suggest a specific antidepressant effect for SGAs, and for olanzapine in particular, mediated through 1 or more non-D2 pathways,15 others have suggested that the difference detected in some studies may have reflected akinesia due to a lack of prophylactic anticholinergic medication in the FGA arm of the studies involving moderate to high doses of the high-potency drug haloperidol.18 It has also been suggested that in drug-naive patients, depression in schizophrenia is related to low presynaptic dopamine function.19 In contrast to trials that have not consistently demonstrated clinical superiority of SGAs over FGAs, clozapine has more consistently been more effective than other antipsychotics including SGAs in the treatment of refractory schizophrenia.20,21 More specifically, in the treatment of symptoms of depression, clozapine has been shown to be more effective than risperidone in reducing symptoms of depression in a study of people with treatment-resistant schizophrenia.22 Thus, the only antipsychotic that has consistently shown evidence of superior efficacy in terms of the positive symptoms of schizophrenia has also shown increased effectiveness against symptoms of depression.

The published reports on the treatment of depression in schizophrenia with antipsychotics usually examine depression as a secondary outcome measure. A recent meta-analysis of SGAs for people with both schizophrenia and depression included only 3 methodologically rigorous studies for which depression was the primary outcome.23 The conclusion of this review was that there were insufficient data to guide patients, prescribers, caregivers, or policy makers and that further studies were warranted.

In 1999, the National Institute of Mental Health initiated the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE), which used an experimental study design to compare the effectiveness of 1 FGA (perphenazine) and all 4 SGAs (olanzapine, risperidone, quetiapine, and ziprasidone) other than clozapine, that were available in the United States in January 2002 for the treatment of chronic schizophrenia. A report on the primary clinical outcomes from CATIE, considering only treatment on the initial randomly assigned drug (phase 1), found that patients treated with olanzapine remained on treatment with their medicine longer than those treated with quetiapine or risperidone and were less likely than all of those receiving other drugs to switch drugs for lack of efficacy.5 None of the second-generation drugs showed statistically significantly greater efficacy or tolerability than the first-generation drug, perphenazine, nor were there any significant differences on measures of neurologic side effects. Weight gain with olanzapine was substantial, averaging 2 lb per month, with concomitant increases in hemoglobin A1c, cholesterol, and triglycerides.

The clinical outcome assessments used in the CATIE study were selected to represent all symptoms and outcomes of relevance to clinical practice, including depression.24 In this study, we examine the differential impact of 4 second- generation drugs and perphenazine on symptoms of depression in the overall CATIE study sample and evaluate whether there are differences in drug effects in the subsample who met criteria for a major depressive episode (MDE) on the primary measure of depressive symptoms (Calgary Depression Scale for Schizophrenia [CDSS]).

METHOD

Study Setting and Design

CATIE was conducted between January 2001 and December 2004 at 57 US sites and included an algorithmically determined series of treatment phases. Patients were initially assigned to olanzapine, perphenazine, quetiapine, risperidone, or ziprasidone under double-blind conditions. However, patients with tardive dyskinesia (TD) (15% of the sample) were not considered in the randomization that included perphenazine and thus were not available for comparisons involving that drug. Although not reported here, patients who discontinued their first treatment were invited to further random assignment to other SGAs, including clozapine, if they so desired. Open treatment was also offered to patients who refused a second blind assignment or whose treatment failed after a second assignment (phase 3), when a small number chose FGAs.

Participants

The study was approved by an institutional review board at each site. Patients 18 to 65 years of age with a diagnosis of schizophrenia25 who were able to take oral antipsychotic medication were eligible. Patients or their guardians provided written informed consent. Patients were excluded if they had a diagnosis of schizoaffective disorder or mental retardation or other cognitive disorders, an unstable serious medical condition, past adverse reactions to a proposed treatment, or treatment-resistant schizophrenia or if they were in their first episode of schizophrenia, pregnant, or breastfeeding.

Interventions

Identical capsules contained olanzapine (7.5 mg), quetiapine (200 mg), risperidone (1.5 mg), perphenazine (8 mg), or ziprasidone (40 mg). Ziprasidone was approved for use by the US Food and Drug Administration during the trial and was added in January 2002, after 40% of the sample had been recruited. Medications were flexibly dosed with 1 to 4 capsules daily, as judged by the study doctor. Concomitant medications were permitted, except for additional antipsychotic agents. Further details about blinding, later phases of treatment, and modal dosing have been presented elsewhere.5,26

Measures

A full description of the measures used in this study is reported elsewhere.24 The outcome of primary interest for this analysis is depression, which was assessed with the CDSS.27 The CDSS is a measure of depression specifically designed to assess depression in schizophrenia separate from negative symptoms.28 It has been validated in independent studies29,30 and recommended as the gold standard for assessing depression in schizophrenia for clinical trials.31

For the purpose of this analysis, patients with a baseline score of ≥ 6 on the CDSS were identified as meeting CDSS criteria for an MDE and thus most likely to benefit from treatment. This level of depression has been previously identified as an appropriate cutoff for the prediction of a major depressive disorder, with a specificity of 77% and sensitivity of 92%.32

Statistical Methods

For consistency and comparability, the statistical methods used in the analysis of continuous measures in this study were the same as those used in the original publication from CATIE.5 The main analyses are limited to the period of treatment with the initially assigned drug (phase 1). The central analysis was a paired comparison between treatment groups of average CDSS scores from all timepoints using a mixed model including terms representing treatment group, the baseline value of the CDSS, time (treated as a classification variable for months 1, 3, 6, 9, 12, 15, and 18), site, a history of recent clinical exacerbation, and baseline-by-time interactions. The baseline-by-time term adjusts for baseline differences in characteristics of patients who dropped out early and thus are less well represented at later timepoints. Treatment-by-time interactions to evaluate differences in time trends between groups were also tested. A random subject effect and a first-order autoregressive covariance structure were used to adjust standard errors for the correlation of observations from the same individual.

Two hundred thirty-one patients with TD were ex cluded from assignment to perphenazine, and ziprasidone was added to the trial after 40% of the patients had been enrolled. Randomization occurred under 4 separate regimens: including and excluding patients with TD, and including and excluding ziprasidone. Analyses were thus con ducted on 4 different datasets with overlapping membership. Each dataset included only patients with an equal chance of being randomly assigned to the treatments under comparison. Perphenazine-treated patients, in particular, were compared only to equivalent patients who did not have TD at baseline.

The primary comparison between the 4 treatments available at the beginning of the trial was an overall 3 degree of freedom test. This test was performed on analytic dataset 1, excluding both patients with TD and those randomly assigned to ziprasidone. If the overall test was significant at P < .05, the 3 second-generation drugs were compared with perphenazine with a Hochberg adjustment for multiple comparisons33 in which the smallest P value was compared to .05/3 = .017 and the largest to P = .05.

Next, with the use of dataset 2, which excludes perphenazine and includes TD patients, the 3 second-generation drugs were compared to each other via step-down testing. If the overall 2 degree of freedom test was significant at P < .05, an α of P < .05 was applied for all comparisons.

Datasets 3 and 4 were used to compare ziprasidone to the other 4 drugs among patients randomized after ziprasidone became available, but with TD patients excluded from the perphenazine comparison. Hochberg adjustment for 4 pairwise comparisons was used to compare ziprasidone and perphenazine in dataset 3 and ziprasidone to the other 3 drugs in dataset 4. The smallest P value was considered significant if P = .05/4 = .013.

Because the impact of these medications on depressive symptoms may have been different among patients who met criteria for an MDE at the time of study entry than among patients who did not, a set of interaction analyses was conducted within each of the 4 strata. Within each stratum, an interaction term was modeled representing the interaction of treatment group by a dichotomous variable indicating whether the patient had met criteria for depression using 2 criteria: a categorical criterion of a major depressive disorder using the CDSS cutoff score of ≤ 6 or greater at baseline and a continuous criterion level of depression assessed on the CDSS. These analyses allowed us to determine whether there were differences between treatment effects among patients who met these a priori criteria for depression and patients who did not. If the interaction term was significant, paired comparisons between treatments were conducted among patients who met the criterion for depression and among patients who did not. Because these analyses were descriptive in nature, an α level of P < .05 was used to test paired comparisons.

RESULTS

Characteristics and Disposition

The baseline demographic and clinical characteristics of the patients have been described in a previous publication and showed no significant differences between treatment groups on baseline measures.5 Although 1,493 patients were enrolled in the study, all data from 1 site (33 patients) were excluded prior to analysis due to concerns about data integrity, and 17 patients never took study drug. A total of 448 (30.69%) patients had a CDSS score of ≤ 6 at baseline and were considered to be in a current MDE. Patients with an MDE were more often white (P = .04), female (P = .006), and younger (P = .02) and had fewer years of treatment (P = .02).

The total CDSS score improved over time in all groups (Figure 1). The mixed models, however, revealed no overall significant differences between treatments within any of the 4 strata (Table 1). There were also no significant treatment-by-time interactions indicating differences in rates of change in depressive symptoms.

Figure 1.

Figure 1

CDSS Least-Squares Means for Each Treatment Group, Corrected for Baseline Levels of Depression

Table 1.

Mixed-Model Analyses of CDSS Least Squares Means Across Treatment Groups and Interaction Between Treatment and Being in a Major Depressive Episode (MDE) at Baselinea

Main Effect of Treatment Group
Interaction of Treatment Group by MDE
Paired Comparisonb
Olanzapine (O) Perphenazine (P) Quetiapine (Q) Risperidone (R) Ziprasidone (Z) F P F P F P
Total n 328 256 326 332 182
Dataset 1 (df= 3): P vs O, Q, and R (excluding patients with tardive dyskinesia and those taking Z)
n 263 256 261 269
Mean CDSS
    Total sample 3.80 3.80 3.67 3.96 0.79 .50 3.32 .02
    No MDE 2.28 2.44 2.35 2.33
    MDE 8.95 8.71 8.51 9.11 NS
Dataset 2: O vs Q vs R (including patients with tardive dyskinesia but excluding those taking Z or P)
n 328 326 332
Mean CDSS
    Total sample 3.73 3.73 3.97 1.43 .24 2.90 .05
    No MDE 2.28 2.12 2.42
    MDE 8.78 8.52 9.06 2.9; Q < R .0056*
Dataset 3: Z vs P (excluding patients with tardive dyskinesia but including those taking Z)
n 146 150
Mean CDSS
    Total sample 3.23 3.73 3.84 3.84 4.23 1.30 .27 1.86 .11
    No MDE 2.45 2.53 2.27 2.47 2.48
    MDE 9.07 8.63 8.55 8.70 9.34 NS
Dataset 4: Z vs O, Q, and R (including patients with tardive dyskinesia and those taking Z)
n 177 181 174 178
Mean CDSS
    Total sample 3.86 3.68 3.77 3.89 0.37 .77 0.75 .15
    No MDE 2.41 2.28 2.41 2.32
    MDE 8.88 8.61 8.91 9.29 NS
a

Least squares means of CDSS scores from months 1, 3, 6, 9, 12, 15 (4,816 patient month observations for data set 1; 4,480 for data set 2; 1,285 for data set 3; and 3,802 for data set 4).

b

All pairwise P values < .05 are presented.

*

Statistically significant using criteria for multiple comparisons.

Abbreviations: CDSS = Calgary Depression Scale for Schizophrenia, NS = nonsignificant.

Interaction analyses of treatment group by MDE at baseline showed interactions between the presence of major depressive disorder at baseline and treatment group in 2 of the analytic strata. The first interaction was observed in dataset 1, the stratum that included patients without TD assigned perphenazine, olanzapine, quetiapine, or risperidone (P < .02), but examination of paired comparisons showed no significant differences involving perphenazine.

An interaction was also observed in dataset 2, the stratum that included all patients randomly assigned to olanzapine, risperidone, or quetiapine (P = .05). Further paired comparisons of CDSS scores among patients meeting criteria for MDE showed a small but statistically significant difference between quetiapine and risperidone (mean = 8.52 for quetiapine vs 9.06 for risperidone, P = .0056), indicating that patients receiving quetiapine had lower depression scores than those receiving risperidone, specifically among patients who met criteria for MDE (Figure 2). Further examination of paired differences between these drugs at specific time-points showed that lower depression scores with quetiapine were observed at only 4 of 7 timepoints: months 3, 6, 9, and 18 (Figure 2).

Figure 2.

Figure 2

CDSS Least-Squares Means for Quetiapine and Risperidone Patient Groups Who Did and Did Not Meet Criteria for an MDE, Corrected for Baseline Levels of Depression

DISCUSSION

The main finding of this study is that we found no evidence of a class benefit of the use of SGAs compared with FGAs in the treatment of symptoms of depression, even in the subset that was above the baseline threshold for MDD. Depression was not the primary outcome measure for this study, and the sample size was not powered for this outcome. Thus, these analyses should be considered descriptive.

Despite this, our post hoc assessment of the results suggests that the clinical importance of the results would not be different if the sample had been larger. The standard deviation for depression scores was 5.0, and the few differences favoring SGAs are all less than 0.1 (0.026–0.04), resulting in effect sizes of less than 0.01. An effect size of 0.2 is considered small, and anything less than 0.2 is not likely to be of clinical importance.

However, in a subsample of schizophrenia patients identified as meeting criteria for MDE, those assigned to quetiapine had lower scores than those assigned to risperidone, but, again, no FGA-SGA differences were seen. These results are in contrast to studies that have reported a difference in change in depression between SGAs and haloperidol.1517 Furthermore, the findings do not lend empirical support to the recommendation for SGAs in schizophrenia with depression of the American Psychiatric Association Clinical Practice Guidelines, a level II recommendation meaning “Recommended with moderate clinical confidence.”12 It has been suggested that the findings of reduced changes in depression with haloperidol as compared to SGAs may have been due to akinetic extrapyramidal side effects (EPS) in the absence of prophylactic anticholinergics.18 The CATIE results presented here, in contrast to earlier studies, used an intermediate-potency FGA, perphenazine, and found no significant differences among groups in the incidence of extrapyramidal side effects, akathisia, or movement disorders or in the prescription of concomitant anticholinergic or antidepressant medications.5 Although there were no overall differences in frequency of antidepressant prescription between antipsychotics in the original CATIE report,5 the rate of prescription was highest in the risperidone group, 16%, and lowest in the quetiapine group, 8%, with perphenazine between these 2 SGAs, at 11%. Although the exact timing and duration of antidepressant treatment are not known, the results favoring quetiapine over risperidone are not likely to be an artifact of greater use of concomitant antidepressant treatment in the quetiapine group.

Another possible explanation of the differences in findings between this study and earlier studies is that both of the earlier studies found differences in change in positive and negative symptoms of schizophrenia or global psychopathology, as assessed by the Positive and Negative Syndrome Scale (PANSS), between SGAs and haloperidol. In contrast to those other studies, there were no differences in change in global psychopathology in the CATIE study. In studies of treatment response to antipsychotic medications, there is evidence that depressive symptoms in acute schizophrenia improve in conjunction with changes in global psychopathology.34 Although the CATIE study was not an acute treatment study, there were statistically significant changes in global psychopathology over time. If the primary driver of reductions in depression is improvement in global psychopathology, then one would expect that treatments of equivalent efficacy in the treatment of general psychopathology would have equivalent effects on depression in schizophrenia. Such a general principle would explain the finding in studies comparing clozapine to other antipsychotics in treatment-resistant schizophrenia in which differential improvements in depression coincided with differential improvements in general psychopathology favoring clozapine.22

Despite the finding of no general effect for SGAs, this study did find evidence of a statistically significant difference between quetiapine and risperidone. While the size of the difference is clinically small, these results are congruent with a study comparing quetiapine with haloperidol in partially responsive schizophrenia35 that showed a differential effect of quetiapine on the PANSS depression factor compared with haloperidol despite no difference in change in global psychopathology between treatments. Positron emission tomography studies in humans suggest that risperidone and quetiapine are at opposite ends of the range of dopamine affinity.36,37 The low and transient D2 occupancy of quetiapine appears to account for its low potential for EPS. This would fit the theory that depression may be exacerbated either by EPS or by the high dopamine blockade that underlies EPS. In contrast to quetiapine, risperidone has the highest D2 receptor affinity of the drugs used in this study, comparable to that of haloperidol.38 It would also fit with a theory that the mesolimbic dopamine reward circuit plays a part in depression39 and that higher occupancy of dopamine D2 receptors may be associated with increased feelings of dysphoria.40,41 The same theory might also explain the earlier findings of reduced depression in studies comparing SGAs versus relatively high doses of haloperidol, but these pharmacologic conceptualizations remain speculative. An alternative explanation might be that a metabolite of quetia-pine, N-desalkylquetiapine, has antidepressant properties.42

Strengths of the study were its large sample size, long duration of follow-up, and recruitment of patients from diverse representative sites with minimal exclusion criteria—all of which increase the generalizability of the results. The investigators also selected a depression scale that was specifically designed for the assessment of depression in schizophrenia and that avoids the confounds of negative symptoms, extrapyramidal symptoms, and depression.

Limitations of this study include the use of secondary outcome data and data loss from attrition. While patients treated with olanzapine stayed significantly longer on treatment than risperidone or quetiapine, there were no differences in duration of treatment between patients treated with quetiapine and risperidone.

In contrast to some previous research and a level II APA guideline, this study of the impact of antipsychotics on depressive symptoms in patients with schizophrenia found no differences between any SGA, including olanzapine, and the FGA perphenazine, but we did detect a signal indicating a small difference favoring quetiapine over risperidone that was limited to patients with an MDE at baseline.

Acknowledgment

This analysis was conducted with the assistance of staff of the VA Northeast Program Evaluation Center, who provided analytic support.

Funding/support: This article was based on results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) project, supported by the National Institute of Mental Health (N01 MH90001). Medication was provided by AstraZeneca Pharmaceuticals LP; Bristol-Myers Squibb Company; Forest Pharmaceuticals, Inc; Janssen Pharmaceutica Products, LP; Eli Lilly and Company; Otsuka Pharmaceutical Co, Ltd; Pfizer Inc; and Zenith Goldline Pharmaceuticals, Inc. The Foundation of Hope of Raleigh, North Carolina, also supported this work.

Footnotes

Trial Registration: clinicaltrials.gov Identifier: NCT00014001

Drug names: clozapine (Clozaril, FazaClo, and others), haloperidol (Haldol and others), olanzapine (Zyprexa), quetiapine (Seroquel), risperidone (Risperdal and others), ziprasidone (Geodon).

Potential conflicts of interest: Dr Davis is an employee of Quintiles.

Dr Stroup has been a consultant for Janssen and Eli Lilly. Dr McEvoy has been a consultant for Pfizer and Indevus, has received grant/research support from Sanofi and Pfizer, and has received honoraria from Eli Lilly. Dr Swartz has received grant/research support and honoraria from and been a speakers/advisory board member for Eli Lilly. Dr Lieberman receives grant/research support from Allon, Forest, Merck, and Pfizer; is an advisory board member for Bioline and Eli Lilly; and holds a patent for Repligen. In 2007–2008, he received grant/research support from AstraZeneca, Bristol-Myers Squibb, Forest, GlaxoSmithKline, Janssen, and Wyeth; was a consultant for Cephalon, Eli Lilly, and Pfizer; was an advisory board member for AstraZeneca, Eli Lilly, Forest, GlaxoSmithKline, Janssen, Otsuka, Pfizer, and Wyeth; held a patent with Repligen; and was a Data and Safety Monitoring Board member for Solvay. Drs Addington, Mohamed, and Rosenheck report no financial or other relationship relevant to the subject of this article.

REFERENCES

  • 1.Geddes J, Freemantle N, Harrison P, et al. Atypical antipsychotics in the treatment of schizophrenia: systematic overview and meta-regression analysis. BMJ. 2000;321(7273):1371–1376. doi: 10.1136/bmj.321.7273.1371. doi:10.1136/bmj.321.7273.1371 PubMed. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jones PB, Barnes TR, Davies L, et al. Randomized controlled trial of the effect on quality of life of second- vs first-generation antipsychotic drugs in schizophrenia: Cost Utility of the Latest Antipsychotic Drugs in Schizophrenia Study (CUtLASS 1). Arch Gen Psychiatry. 2006;63(10):1079–1087. doi: 10.1001/archpsyc.63.10.1079. doi:10.1001/archpsyc.63.10.1079 PubMed. [DOI] [PubMed] [Google Scholar]
  • 3.Leucht SA, Pitschel-Walz GA, Abraham DA, et al. Efficacy and extra-pyramidal side-effects of the new antipsychotics olanzapine, quetiapine, risperidone, and sertindole compared to conventional antipsychotics and placebo: a meta-analysis of randomized controlled trials. Schizophr Res. 1999;35(1):51–68. doi: 10.1016/s0920-9964(98)00105-4. doi:10.1016/S0920-9964(98)00105-4 PubMed. [DOI] [PubMed] [Google Scholar]
  • 4.Leucht S, Wahlbeck K, Hamann J, et al. New generation antipsychotics versus low-potency conventional antipsychotics: a systematic review and meta-analysis. Lancet. 2003;361(9369):1581–1589. doi: 10.1016/S0140-6736(03)13306-5. doi:10.1016/S0140-6736(03)13306-5 PubMed. [DOI] [PubMed] [Google Scholar]
  • 5.Lieberman JA, Stroup TS, McEvoy JP, et al. Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353(12):1209–1223. doi: 10.1056/NEJMoa051688. doi:10.1056/NEJMoa051688 PubMed. [DOI] [PubMed] [Google Scholar]
  • 6.Davis JM, Chen N, Glick ID. A meta-analysis of the efficacy of second-generation antipsychotics. Arch Gen Psychiatry. 2003;60(6):553–564. doi: 10.1001/archpsyc.60.6.553. doi:10.1001/archpsyc.60.6.553 PubMed. [DOI] [PubMed] [Google Scholar]
  • 7.Zisook S, McAdams LA, Kuck J, et al. Depressive symptoms in schizophrenia. Am J Psychiatry. 1999;156(11):1736–1743. doi: 10.1176/ajp.156.11.1736. PubMed. [DOI] [PubMed] [Google Scholar]
  • 8.Martin RL, Cloninger CR, Guze SB, et al. Frequency and differential diagnosis of depressive syndromes in schizophrenia. J Clin Psychiatry. 1985;46(11, Pt 2):9–13. PubMed. [PubMed] [Google Scholar]
  • 9.Haw C, Hawton K, Sutton L, et al. Schizophrenia and deliberate self-harm: a systematic review of risk factors. Suicide Life Threat Behav. 2005;35(1):50–62. doi: 10.1521/suli.35.1.50.59260. doi:10.1521/suli.35.1.50.59260 PubMed. [DOI] [PubMed] [Google Scholar]
  • 10.Hawton K, Sutton L, Haw C, et al. Schizophrenia and suicide: systematic review of risk factors. Br J Psychiatry. 2005;187(1):9–20. doi: 10.1192/bjp.187.1.9. doi:10.1192/bjp.187.1.9 PubMed. [DOI] [PubMed] [Google Scholar]
  • 11.Lehman AF. A quality of life interview for the chronically mentally ill. Eval Program Plann. 1988;11(1):51–62. doi:10.1016/0149-7189(88)90033-X. [Google Scholar]
  • 12.American Psychiatric Association Practice Guideline for the Treatment of Patients With Schizophrenia. Am J Psychiatry. (Second Edition.) 2004;161(suppl):1–55. [PubMed] [Google Scholar]
  • 13.Klein DF. Importance of psychiatric diagnosis in prediction of clinical drug effects. Arch Gen Psychiatry. 1967;16(1):118–126. doi: 10.1001/archpsyc.1967.01730190120016. PubMed. [DOI] [PubMed] [Google Scholar]
  • 14.Levinson DF, Umapathy C, Musthaq M. Treatment of schizoaffective disorder and schizophrenia with mood symptoms. Am J Psychiatry. 1999;156(8):1138–1148. doi: 10.1176/ajp.156.8.1138. PubMed. [DOI] [PubMed] [Google Scholar]
  • 15.Tollefson GD, Sanger TM, Lu YTME, et al. Depressive signs and symptoms in schizophrenia: a prospective blinded trial of olanzapine and haloperidol. Arch Gen Psychiatry. 1998;55(3):250–258. doi: 10.1001/archpsyc.55.3.250. doi:10.1001/archpsyc.55.3.250 PubMed. [DOI] [PubMed] [Google Scholar]
  • 16.Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. J Clin Psychiatry. 1997;58(12):538–546. doi: 10.4088/jcp.v58n1205. PubMed. [DOI] [PubMed] [Google Scholar]
  • 17.Csernansky JG, Mahmoud R, Brenner R; Risperidone-USA-79 Study Group A comparison of risperidone and haloperidol for the prevention of relapse in patients with schizophrenia. N Engl J Med. 2002;346(1):16–22. doi: 10.1056/NEJMoa002028. doi:10.1056/NEJMoa002028 PubMed. [DOI] [PubMed] [Google Scholar]
  • 18.Rosenheck RA. Open forum: effectiveness versus efficacy of second-generation antipsychotics: haloperidol without anticholinergics as a comparator. Psychiatr Serv. 2005;56(1):85–92. doi: 10.1176/appi.ps.56.1.85. doi:10.1176/appi.ps.56.1.85 PubMed. [DOI] [PubMed] [Google Scholar]
  • 19.Hietala J, Syvälahti E, Vilkman H, et al. Depressive symptoms and presynaptic dopamine function in neuroleptic-naive schizophrenia. Schizophr Res. 1999;35(1):41–50. doi: 10.1016/s0920-9964(98)00113-3. doi:10.1016/S0920-9964(98)00113-3 PubMed. [DOI] [PubMed] [Google Scholar]
  • 20.Lewis SW, Barnes TR, Davies L, et al. Randomized controlled trial of effect of prescription of clozapine versus other second-generation anti-psychotic drugs in resistant schizophrenia. Schizophr Bull. 2006;32(4):715–723. doi: 10.1093/schbul/sbj067. doi:10.1093/schbul/sbj067 PubMed. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.McEvoy JP, Lieberman JA, Stroup TS, et al. CATIE Investigators. Effectiveness of clozapine versus olanzapine, quetiapine, and risperidone in patients with chronic schizophrenia who did not respond to prior atypical antipsychotic treatment. Am J Psychiatry. 2006;163(4):600–610. doi: 10.1176/ajp.2006.163.4.600. doi:10.1176/appi.ajp.163.4.600 PubMed. [DOI] [PubMed] [Google Scholar]
  • 22.Azorin JM, Spiegel R, Remington G, et al. A double-blind comparative study of clozapine and risperidone in the management of severe chronic schizophrenia. Am J Psychiatry. 2001;158(8):1305–1313. doi: 10.1176/appi.ajp.158.8.1305. doi:10.1176/appi.ajp.158.8.1305 PubMed. [DOI] [PubMed] [Google Scholar]
  • 23.Furtado VA, Srihari V. Atypical antipsychotics for people with both schizophrenia and depression. Cochrane Database Syst Rev. 2008;(1):CD005377. doi: 10.1002/14651858.CD005377.pub2. PubMed. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Swartz MS, Perkins DO, Stroup TS, et al. Assessing clinical and functional outcomes in the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial. Schizophr Bull. 2003;29(1):33–43. doi: 10.1093/oxfordjournals.schbul.a006989. PubMed. [DOI] [PubMed] [Google Scholar]
  • 25.First MB, Spitzer RL, Gibbon MB, et al. Structured Clinical Interview for DSM IV Axis I and II Disorders–Patient Edition (SCID-I/P) Biometric Research Institute, New York State Psychiatric Institute; New York, NY: 1996. pp. 1–136. [Google Scholar]
  • 26.Stroup TS, McEvoy JP, Swartz MS, et al. The National Institute of Mental Health Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) project: schizophrenia trial design and protocol development. Schizophr Bull. 2003;29(1):15–31. doi: 10.1093/oxfordjournals.schbul.a006986. PubMed. [DOI] [PubMed] [Google Scholar]
  • 27.Addington D, Addington J, Maticka-Tyndale E, et al. Reliability and validity of a depression rating scale for schizophrenics. Schizophr Res. 1992;6(3):201–208. doi: 10.1016/0920-9964(92)90003-n. doi:10.1016/0920-9964(92)90003-N PubMed. [DOI] [PubMed] [Google Scholar]
  • 28.Addington D, Addington J, Maticka-Tyndale E. Specificity of the Calgary Depression Scale for schizophrenics. Schizophr Res. 1994;11(3):239–244. doi: 10.1016/0920-9964(94)90017-5. doi:10.1016/0920-9964(94)90017-5 PubMed. [DOI] [PubMed] [Google Scholar]
  • 29.Collins AA, Remington G, Coulter K, et al. Depression in schizophrenia: a comparison of three measures. Schizophr Res. 1996;20(1-2):205–209. doi: 10.1016/0920-9964(95)00107-7. doi:10.1016/0920-9964(95)00107-7 PubMed. [DOI] [PubMed] [Google Scholar]
  • 30.Kontaxakis VP, Havaki-Kontaxaki BJ, Stamouli SS, et al. Comparison of four scales measuring depression in schizophrenic inpatients. Eur Psychiatry. 2000;15(4):274–277. doi: 10.1016/s0924-9338(00)00232-7. doi:10.1016/S0924-9338(00)00232-7 PubMed. [DOI] [PubMed] [Google Scholar]
  • 31.Collaborative Working Group on Clinical Trial Evaluations Atypical antipsychotics for treatment of depression in schizophrenia and affective disorders. J Clin Psychiatry. 1998;59(suppl 12):41–45. [PubMed] [Google Scholar]
  • 32.Addington D, Addington J, Maticka-Tyndale E. Assessing depression in schizophrenia: the Calgary Depression Scale. Br J Psychiatry Suppl. 1993;163(22):39–44. [PubMed] [Google Scholar]
  • 33.Hochberg Y. A sharper Bonferroni procedure for multiple tests of significance. Biometrika. 1988;75(4):800–802. doi:10.1093/biomet/75.4.800. [Google Scholar]
  • 34.Jäger M, Riedel M, Schmauss M, et al. Depression during an acute episode of schizophrenia or schizophreniform disorder and its impact on treatment response. Psychiatry Res. 2008;158(3):297–305. doi: 10.1016/j.psychres.2007.01.002. doi:10.1016/j.psychres.2007.01.002 PubMed. [DOI] [PubMed] [Google Scholar]
  • 35.Emsley RA, Buckley P, Jones AM, et al. Differential effect of quetiapine on depressive symptoms in patients with partially responsive schizophrenia. J Psychopharmacol. 2003;17(2):210–215. doi: 10.1177/0269881103017002010. doi:10.1177/0269881103017002010 PubMed. [DOI] [PubMed] [Google Scholar]
  • 36.Kapur S, Zipursky R, Jones C, et al. A positron emission study of quetiapine in schizophrenia: a preliminary finding of an antipsychotic effect with only transiently high dopamine D2 receptor occupancy. Arch Gen Psychiatry. 2000;57(6):533–539. doi: 10.1001/archpsyc.57.6.553. doi:10.1001/archpsyc.57.6.553 PubMed. [DOI] [PubMed] [Google Scholar]
  • 37.Kapur S, Remington G, Zipursky RB, et al. The D2 dopamine receptor occupancy of risperidone and its relationship to extrapyramidal symptoms: a PET study. Life Sci. 1995;57(10):PL103–PL107. doi: 10.1016/0024-3205(95)02037-j. doi:10.1016/0024-3205(95)02037-J PubMed. [DOI] [PubMed] [Google Scholar]
  • 38.Strange PG. Antipsychotic drugs: importance of dopamine receptors for mechanisms of therapeutic actions and side effects. Pharmacol Rev. 2001;53(1):119–133. PubMed. [PubMed] [Google Scholar]
  • 39.Nestler EJ, Carlezon WA., Jr The mesolimbic dopamine reward circuit in depression. Biol Psychiatry. 2006;59(12):1151–1159. doi: 10.1016/j.biopsych.2005.09.018. doi:10.1016/j.biopsych.2005.09.018 PubMed. [DOI] [PubMed] [Google Scholar]
  • 40.de Haan L, Lavalaye J, van Bruggen M, et al. Subjective experience and dopamine D2 receptor occupancy in patients treated with antipsychotics: clinical implications. Can J Psychiatry. 2004;49(5):290–296. doi: 10.1177/070674370404900503. PubMed. [DOI] [PubMed] [Google Scholar]
  • 41.Voruganti LN, Awad AG. Subjective and behavioural consequences of striatal dopamine depletion in schizophrenia—findings from an in vivo SPECT study. Schizophr Res. 2006;88(1–3):179–186. doi: 10.1016/j.schres.2006.07.012. doi:10.1016/j.schres.2006.07.012 PubMed. [DOI] [PubMed] [Google Scholar]
  • 42.Jensen NH, Rodriguiz RM, Caron MG, et al. N-desalkylquetiapine, a potent norepinephrine reuptake inhibitor and partial 5-HT1A agonist, as a putative mediator of quetiapine's antidepressant activity. Neuropsychopharmacology. 2008;33(10):2303–2312. doi: 10.1038/sj.npp.1301646. doi:10.1038/sj.npp.1301646 PubMed. [DOI] [PubMed] [Google Scholar]

RESOURCES