Scolaris Content Display Scolaris Content Display

Tratamiento de mantenimiento con fármacos antipsicóticos para la esquizofrenia

This is not the most recent version

Collapse all Expand all

References

References to studies included in this review

Andrews 1976 {published data only}

Andrews P, Hall JN, Snaith RP. A controlled trial of phenothiazine withdrawal in chronic schizophrenic patients. British Journal of Psychiatry 1976;128:451‐5.

Arato 2002 {published data only}

Arato M, O'Connor R, Meltzer HY. A 1‐year, double‐blind, placebo‐controlled trial of ziprasidone 40, 80 and 160 mg/day in chronic schizophrenia: the Ziprasidone Extended Use in Schizophrenia (ZEUS) study. International Clinical Psychopharmacology 2002;17:207‐15.

Baro 1970 {published data only}

Baro F, Brugmans J, Dom R, Van Lommel R. Maintenance therapy of chronic psychotic patients with a weekly oral dose of R 16341. A controlled double‐blind study. Journal of Clinical Pharmacology 1970;10:330‐41.

Beasley 2003 {published data only}

Beasley CM, Sutton VK, Hamilton SH, Walker DJ, Dossenbach M, Taylor CC, et al. A double‐blind, randomised, placebo‐controlled trial of olanzapine in the prevention of psychotic relapse. Journal of Clinical Psychopharmacology 2003;23:582‐94.
Beasley CM, Sutton VK, Taylor CC, Sethuraman G, Dossenbach M, Naber D. Is quality of life among minimally symptomatic patients with schizophrenia better following withdrawal or continuation of antipsychotic treatment?. Journal of Clinical Psychopharmacology 2006;26:40‐4.
Beasley CM, Walker DJ, Sutton VK, Alaka K, Naber D, Namjoshi M. The effectiveness of olanzapine versus placebo in maintaining the quality of life in stabilized patients with schizophrenia. European Neuropsychopharmacology 2003;13:S342.

Blackburn 1981 {published data only}

Blackburn HL, Allen JL. Behavioral effects of interrupting and resuming tranquilizing medication among schizophrenics. Journal of Nervous and Mental Disease 1981;133:303‐8.

Boonstra 2011 {published data only}

Boonstra G. Schizophrenia termination of pharmacotherapy‐STOP‐trial. Current Controlled Trials2005. [ISRCTN: 6332944]
Boonstra G, Burger H, Grobbe DE, Kahn RS. Antipsychotic prophylaxis is needed after remission from a first psychotic episode in schizophrenia patients: results from an aborted randomised trial. International Journal of Psychiatry in Clinical Practice 2011;15(2):128‐34.

Caffey 1964 {published data only}

Caffey EM, Diamond LS, Frank TV, Grasberger JC, Herman L, Klett CJ, et al. Discontinuation or reduction of chemotherapy in chronic schizophrenics. Journal of Chronic Diseases 1964;17:347‐58.
Caffey EM, Forrest IS, Frank TV, Klett CJ. Phenothiazine excretion in chronic schizophrenics. American Journal of Psychiatry 1963;120:578‐82.

Channabasavanna 1987 {published data only}

Channabasavanna SM, Michael A. Penfluridol maintenance therapy in schizophrenia: a controlled study. Indian Journal of Psychiatry 1987;29:333‐6.

Chen 2010 {published data only}

Chen EYH, Hui CLM, Lam M, Law CW, Chiu CPY, Chung DWS, et al. A double‐blind randomised placebo‐controlled study of relapse prevention in remitted first‐episode psychosis patients following one year of maintenance therapy. Schizophrenia Research 2008;98:11‐2.
Chen EYH, Hui CLM, Lam MML, Chiu CPY, Law CW, Chung DWS, et al. Maintenance treatment with quetiapine versus discontinuation after one year of treatment in patients with remitted first episode psychosis: randomised controlled trial. British Medical Journal 2010;341:c4024.

Cheung 1981 {published data only}

Cheung HK. Schizophrenics fully remitted on neuroleptics for 3‐5 years ‐ to stop or continue drugs?. British Journal of Psychiatry 1981;138:490‐4.

Clark 1975 {published data only}

Clark ML, Huber WK, Hill D, Wood F, Costiloe JP. Pimozide in chronic schizophrenic outpatients. Diseases of the Nervous System 1975;36:137‐41.

Cooper 2000 {published data only}

Cooper SJ, Butler A, Tweed J, Raniwalla J, Welch C. Zotepine in the prevention of relapse. Proceedings of the 6th World Congress of Biological Psychiatry; 1997 June 22‐27; Nice, France. 1997.
Cooper SJ, Butler A, Tweed J, Welch C, Raniwalla J. Zotepine in the prevention of recurrence: a randomised, double‐blind, placebo‐controlled study for chronic schizophrenia. Psychopharmacology 2000;150:237‐43.
Cooper SJ, Butler A, Tweed JA, Welch CP, Wighton AJ, Appleby P, et al. Zotepine is effective in preventing recurrence in patients with chronic schizophrenia. Proceedings of the 11th European College of Neuropsychopharmacology Congress; 1998 Oct 31 ‐ Nov 4; Paris, France. 1998.
Cooper SJ, Butler A, Tweed JA, Welch CP, Wighton AJ, Appleby P, et al. Zotepine is effective in preventing recurrence in patients with chronic schizophrenia. Schizophrenia Research 2000;41:207‐8.

Crow 1986 {published data only}

Crow TJ, MacMillan JF, Johnson AL, Johnstone EC. A randomised controlled trial of prophylactic neuroleptic treatment. British Journal of Psychiatry 1986;148:120‐7.

Denijs 1973 {published data only}

Denijs EL, Vereeken JL. Pimozide (OrapR, R 6238) in residual schizophrenia. A clinical evaluation with long‐term double‐blind follow‐up. Psychiatria, Neurologia, Neurochirurgia 1973;76:47‐59.

Doddi 1979 {published data only}

Dotti A, Bersani G, Rubino IA, Eliseo C. Double‐blind trial of fluphenazine decanoate against placebo in ambulant maintenance treatment of chronic schizophrenics [Studio in doppio cieco della flufenazina decanoato versus placebo nella terapia ambulatoriale di mantenimento di pazienti schizofrenici cronici]. Rivista di Psichiatria 1979;14:374‐83.

Eklund 1991 {published data only}

Eklund K. Low dose of haloperidol decanoate is effective against relapses in schizophrenia patients. A double‐blind placebo controlled study. Proceedings of the 17th Collegium Internationale Neuro‐Psychopharmacologicum Congress; 1990 Sep 10‐14; Kyoto, Japan. 1990:287.
Eklund K, Forsman A. Minimal effective dose and relapse ‐ double‐blind trial: haloperidol decanoate vs. placebo. Clinical Neuropharmacology 1991;14:S7‐S15.

Elie 1975 {published data only}

Elie R, Gagnon MA, Gauthier R, Jequier JC. Effects of neuroleptic withdrawal on the drug‐induced extrapyramidal syndrome of chronic schizophrenia [Effets d'un sevrage neuroleptique sur le syndrome extrapyramidal medicamenteux de la schizophrenie chronique]. Union Medicale du Canada 1975;104:909‐14.

Freeman 1962 {published data only}

Freeman LS, Alson E. Prolonged withdrawal of chlorpromazine in chronic patients. Diseases of the Nervous System 1962;23:522‐5.

Gallant 1974 {published data only}

Gallant DM, Mielke DH, Spirtes MA, Swanson WC, Bost R. Penfluridol: an efficacious long‐acting oral antipsychotic compound. American Journal of Psychiatry 1974;131:699‐702.

Gardos 1984 {published data only}

Gardos G, Cole JO, Rapkin RM, LaBrie RA, Baquelod E, Moore P, et al. Anticholinergic challenge and neuroleptic withdrawal. Changes in dyskinesia and symptom measures. Archives of General Psychiatry 1984;41:1030‐5.

Garfield 1966 {published data only}

Garfield SL, Gershon S, Sletten I, Neubauer H, Ferrel E. Withdrawal of ataractic medication in schizophrenic patients. Diseases of the Nervous System 1966;27:321‐5.

Gitlin 1988 {published data only}

Gitlin MJ, Midha KK, Fogelson D, Nuechterlein KH. Persistence of fluphenazine in plasma after decanoate withdrawal. Journal of Clinical Psychopharmacology 1988;8:53‐6.

Goldberg 1981 {published data only}

Goldberg SC, Shenoy RS, Sadler A, Hamer R, Ross B. The effects of a drug holiday on relapse and tardive dyskinesia in chronic schizophrenics. Psychopharmacology Bulletin 1981;17:116‐7.
Shenoy RS, Sadler AG, Goldberg SC, Hamer RM, Ross B. Effects of a six‐week drug holiday on symptom status, relapse, and tardive dyskinesia in chronic schizophrenics. Journal of Clinical Psychopharmacology 1981;1:141‐5.

Gross 1960 {published data only}

Gross M, Hitchman IL, Reeves WP, Lawrence J, Newell PC. Discontinuation of treatment with ataractic drugs. A preliminary report. American Journal of Psychiatry 1960;116:931‐2.

Gross 1974 {published data only}

Gross HS. A double‐blind comparison of once‐a‐day pimozide, trifluoperazine, and placebo in the maintenance care of chronic schizophrenic outpatients. Current Therapeutic Research 1974;16:696‐705.

Hershon 1972 {published data only}

Hershon HI, Kennedy PF, McGuire RJ. Persistence of extra‐pyramidal disorders and psychiatric relapse after withdrawal of long‐term phenothiazine therapy. British Journal of Psychiatry 1972;120(554):41‐50.
Stevens BC. Role of fluphenazine decanoate in lessening the burden of chronic schizophrenics on the community. Psychological Medicine 1973;3:141‐58.

Hirsch 1973 {published data only}

Hankoff LD, Engelhardt DM, Freedman N, Mann D, Maroolis R. Denial of illness in schizophrenic outpatients: effects of psychopharmacological treatment. Archives of General Psychiatry 1960;3:657‐66.
Hirsch SR, Gaind R, Rohde PD, Stevens BC, Wing JK. Outpatient maintenance of chronic schizophrenic patients with long‐acting fluphenazine: double‐blind placebo trial. Report to the Medical Research Council Committee on Clinical Trials in Psychiatry. British Medical Journal 1973;1:633‐7.

Hirsch 1996 {published data only}

Hirsch SR, Bowen JT, Emami J. The effect of life events and medication in the aetiology of schizophrenic relapse. Schizophrenia Research 1993;9:266.
Hirsch SR, Bowen JT, Emami J, Cramer P, Jolley A, Haw C, et al. A one year prospective study of the effect of life events and medication in the aetiology of schizophrenic relapse. British Journal of Psychiatry 1996;168:49‐56.

Hogarty 1973 {published data only}

Goldberg SC, Schooler NR, Hogarty GE, Roper M. Prediction of relapse in schizophrenic outpatients treated by drug and sociotherapy. Archives of General Psychiatry 1977;34:171‐84.
Hogarty GE, Goldberg SC. Drug and sociotherapy in the aftercare of schizophrenic patients. One‐year relapse rates. Archives of General Psychiatry 1973;28:54‐64.
Hogarty GE, Goldberg SC, Schooler NR. Drug and sociotherapy in the aftercare of schizophrenic patients. III. Adjustment of nonrelapsed patients. Archives of General Psychiatry 1974;31:609‐18.
Hogarty GE, Goldberg SC, Schooler NR, Ulrich RF. Drug and sociotherapy in the aftercare of schizophrenic patients. II. Two‐year relapse rates. Archives of General Psychiatry 1974;31:603‐8.
Hogarty GE, Munetz MR. Pharmacogenic depression among outpatient schizophrenic patients: a failure to substantiate. Journal of Clinical Psychopharmacology 1984;4:17‐24.
Hogarty GE, Schooler NR, Ulrich R, Mussare F, Ferro P, Herron E. Fluphenazine and social therapy in the aftercare of schizophrenic patients. Relapse analyses of a two‐year controlled study of fluphenazine decanoate and fluphenazine hydrochloride. Archives of General Psychiatry 1979;36:1283‐94.
Hogarty GE, Ulrich RF. Temporal effects of drug and placebo in delaying relapse in schizophrenic outpatients. Archives of General Psychiatry 1977;34:297‐301.

Hough 2010 {published data only}

Hough D, Gopal S, Vijapukar U, Lim P, Morozova M, Eerdekens M. Paliperidone palmitate in prevention of symptom recurrence in patients with schizophrenia: a randomised, double‐blind, placebo‐controlled study. Proceedings of the 161st Annual Meeting of the American Psychiatric Association; 2008 May 3‐8, Washington DC, USA. 2008.
Hough D, Gopal S, Vjapukar U, Lim P, Morozowa M, Eerdekens M. Paliperidone palmitate maintenance treatment in delaying the time to relapse in patients with schizophrenia: a randomised, double‐blind, placebo‐controlled study. Schizophrenia Research 2010;116:107‐17.

Kane 1979 {published data only}

Kane JM, Rifkin A, Quitkin F, Nayak D, Saraf K, Ramos Lorenzi JR, et al. Low dose fluphenazine decanoate in maintenance treatment of schizophrenia. Psychiatry Research 1979;1:341‐8.

Kane 1982 {published data only}

Kane JM, Rifkin A, Quitkin F, Nayak D, Ramos‐Lorenzi J. Fluphenazine vs. placebo in patients with remitted, acute first‐episode schizophrenia. Archives of General Psychiatry 1982;39:70‐3.
Kane JM, Rifkin A, Woerner M, Reardon G. Low‐dose neuroleptics in outpatient schizophrenics. Psychopharmacology Bulletin 1982;18:20‐1.

Keskiner 1968 {published data only}

Keskiner A, Holden JMC, Itil TM. Maintenance treatment of schizophrenic outpatients with a depot phenothiazine. Psychosomatics 1968;9:166‐71.
Keskiner A, Simeon J, Fink M, Itil TM. Long‐acting phenothiazine (fluphenazine decanoate) treatment of psychosis. Archives of General Psychiatry 1968;18:477‐81.
King DJ, Devaney N, Cooper SJ, Blomqvist M, Mitchel MJ. Pharmacokinetics and antipsychotic effect of remoxipride in chronic schizophrenic patients. Journal of Psychopharmacology 1990;4:83‐9.
Kinross‐Wright J, Charalampous KD. A controlled study of a very long‐acting phenothiazine preparation. International Journal of Neuropsychiatry 1965;1:66‐70.

Kramer 2007 {published data only}

Kramer M, Simpson G, Maciulis V, Kushner S, Vijapurkar U, Lim P, et al. Paliperidone extended‐release tablets for prevention of symptom recurrence in patients with schizophrenia: a randomised, double‐blind, placebo‐controlled study. Journal of Clinical Psychopharmacology 2007;27:6‐14.

Kurland 1975 {published data only}

Kurland AA, Ota KY. Penfluridol: once‐a‐week oral maintenance neuroleptic in the management of chronic schizophrenics. Psychopharmacology Bulletin 1975;11:12‐4.
Kurland AA, Ota KY, Slotnick VB. Penfluridol: a long‐acting oral neuroleptic. A controlled study. Journal of Clinical Pharmacology 1975;15:611‐21.

Leff 1971 {published data only}

Leff JP, Hirsch SR, Gaind R, Rohde PD, Stevens BC. Life events and maintenance therapy in schizophrenic relapse. British Journal of Psychiatry 1973;123:659‐60.
Leff JP, Wing JK. Trial of maintenance therapy in schizophrenia. British Medical Journal 1971;3:599‐604.

Levine 1980 {published data only}

Gelenberg AJ, Doller JC, Schooler NR, Mieske M, Severe J, Mandel MR. Acute extrapyramidal reactions with fluphenazine hydrochloride and fluphenazine decanoate. American Journal of Psychiatry 1979;136:217‐9.
Levine J, Schooler NR, Severe J, Escobar J, Gelenberg A, Mandel M, et al. Discontinuation of oral and depot fluphenazine in schizophrenic patients after one year of continuous medication: a controlled study. Advances in Biochemical Psychopharmacology 1980;24:483‐93.
Mandel MR, Severe JB, Schooler NR, Gelenberg AJ, Mieske M. Development and prediction of postpsychotic depression in neuroleptic‐treated schizophrenics. Archives of General Psychiatry 1982;39:197‐203.
Schooler NR, Levine J. Dosage and side effect comparisons between oral and depot fluphenazine. Psychopharmacology Bulletin 1977;13:29‐31.
Schooler NR, Levine J, NIMH‐PRB Collaborative Fluphenazine Study Group. The initiation of long‐term pharmacotherapy in schizophrenia: dosage and side effect comparison between oral and depot fluphenazine. Pharmacopsychiatry 9;1976:159‐69.
Schooler NR, Levine J, NIMH‐PRB Collaborative Fluphenazine Study Group. The initiation of long‐term pharmacotherapy in schizophrenia: dosage and side effect comparisons between oral and depot fluphenazine. Pharmacopsychiatry 1976;9:159‐69.
Schooler NR, Levine J, Severe JB. Depot fluphenazine in the prevention of relapse in schizophrenia: evaluation of a treatment regimen. Psychopharmacology Bulletin 1979;15:44‐7.
Schooler NR, Levine J, Severe JB, Brauzer B, DiMascio A, Klerman GL, et al. Prevention of relapse in schizophrenia. An evaluation of fluphenazine decanoate. Archives of General Psychiatry 1980;37:16‐24.

Marjerrison 1964 {published data only}

Marjerrison G, Irvine D, Stewart CN, Williams R, Matheu H, Demay M. Withdrawal of long‐term phenothiazines from chronically hospitalized psychiatric patients. Canadian Psychiatric Association Journal 1964;9(4):290‐8.

McCreadie 1989 {published data only}

McCreadie RG, Wiles D, Grant S, Crockett GT, Mahmood Z, Livingston MG, et al. The Scottish first episode schizophrenia study. Acta Psychiatrica Scandinavica 1989;80:597‐602.

Melnyk 1966 {published data only}

Melnyk WT, Worthington AG, Laverty SG. Abrupt withdrawal of chlorpromazine and thioridazine from schizophrenic in‐patients. Canadian Psychiatric Association Journal 1966;11:410‐3.

Morton 1968 {published data only}

Morton MR. A study of the withdrawal of chlorpromazine or trifluoperazine in chronic schizophrenia. American Journal of Psychiatry 1968;124:1585‐8.

Nishikawa 1982 {published data only}

Nishikawa T, Tsuda A, Tanaka M, Koga I, Uchida Y. Prophylactic effect of neuroleptics in symptom‐free schizophrenia. Psychopharmacology 1982;77:301‐4.

Nishikawa 1984 {published data only}

Nishikawa T, Tsuda A, Tanaka M, Hoaki Y, Koga I, Uchida Y. Prophylactic effect of neuroleptics in symptom‐free schizophrenics: a comparative dose response study of haloperidol and propericiazine. Psychopharmacology 1984;82:153‐6.

Odejide 1982 {published data only}

Odejide OA, Aderounmu AF. Double‐blind placebo substitution: withdrawal of fluphenazine decanoate in schizophrenic patients. Journal of Clinical Psychiatry 1982;43:195‐6.

Olson 1962 {published data only}

Olson GW, Peterson DB. Intermittent chemotherapy for chronic psychiatric inpatients. Journal of Nervous and Mental Disease 1962;134:1459.
Olson GW, Peterson DB. Sudden removal of tranquilizing drugs from chronic psychiatric patients. Journal of Mental and Nervous Disease 1960;131:252‐5.

Ota 1973 {published data only}

Ota KY, Kurland AA. A double‐blind comparison of haloperidol oral concentrate, haloperidol solutabs and placebo in the treatment of chronic schizophrenia. Journal of Clinical Pharmacology 1973;13:99‐110.

Peuskens 2007 {published data only}

Peuskens J, Trivedi J, Malyarov S, Brecher M, Svensson O, Miller F, et al. Prevention of schizophrenia relapse with extended release quetiapine fumarate dosed once daily: a randomised, placebo‐controlled trial in clinically stable patients. Psychiatry 2007;4(11):34‐50.

Pfizer 2000 {published data only}

Pfizer 2000. Multicentre double‐blind study of ziprasidone versus placebo in relapse prevention for hospitalised patients with chronic or subchronic schizophrenia. Study report. Unpublished.

Pietzcker 1993 {published data only}

Gaebel W, Frick U, Kopcke W, Linden M, Mueller P, Mueller Spahn F, et al. Early neuroleptic intervention in schizophrenia: are prodromal symptoms valid predictors of relapse?. British Journal of Psychiatry Supplementum 1993;163:8‐12.
Gaebel W, Janner M, Frommann N, Pietzcker A, Kopcke W, Linden M, et al. First vs multiple episode schizophrenia: two‐year outcome of intermittent and maintenance medication strategies. Schizophrenia Research 2002;53(1‐2):145‐59.
Gaebel W, Moeller HJ. Treatment strategies in first episode schizophrenia. Proceedings of the 12th World Congress of Psychiatry; 2002 Aug 24‐29; Yokohama, Japan. 2002.
Pietzcker A, Gaebel W, Koepcke W, Linden M, Mueller P, Mueller‐Spahn F, et al. Intermittent versus maintenance neuroleptic long‐term treatment in schizophrenia ‐ 2‐year results of a German multicenter study. Journal of Psychiatric Research 1993;27:321‐39.
Pietzcker A, Gaebel W, Kopcke W, Linden M, Muller P, Muller‐Spahn, et al. A German multicenter study on the neuroleptic long‐term therapy of schizophrenic patients. Preliminary report. Pharmacopsychiatry 1986;19:161‐6.

Pigott 2003 {published data only}

Pigott TA, Carson WH, Saha AR, Torbeyns AF, Stock EG, Ingenito GG. Aripiprazole for the prevention of relapse in stabilized patients with chronic schizophrenia: a placebo‐controlled 26‐week study. Journal of Clinical Psychiatry 2003;64:1048‐56.

Prien 1968 {published data only}

Gardos G, Cole JO, LaBrie RA. A 12‐year follow‐up study of chronic schizophrenics. Psychopharmacology 1982;33:983‐4.
Prien RF, Cole JO. High dose chlorpromazine therapy in chronic schizophrenia. Report of National Institute of Mental Health ‐ psychopharmacology research branch collaborative study group. Archives of General Psychiatry 1968;18:482‐95.
Prien RF, Cole JO, Belkin NF. Relapse in chronic schizophrenics following abrupt withdrawal of tranquillizing medication. British Journal of Psychiatry 1968;115:679‐86.
Prien RF, DeLong SL, Cole JO, Levine J. Ocular changes occurring with prolonged high dose chlorpromazine therapy. Results from a collaborative study. Archives of General Psychiatry 1970;23:464‐8.
Prien RF, Levine J, Cole JO. Indications for high dose chlorpromazine therapy in chronic schizophrenia. Diseases of the Nervous System 1970;31:739‐45.

Prien 1969 {published data only}

Prien RF, Levine J, Cole JO. High dose trifluoperazine therapy in chronic schizophrenia. American Journal of Psychiatry 1969;126:305‐13.

Rifkin 1979 {published data only}

Rifkin A, Quitkin F, Kane J, Klein DF. Fluphenazine decanoate, oral fluphenazine, and placebo in the treatment of remitted schizophrenics. II. Rating scale data. Psychopharmacology Bulletin 1977;13:40‐50.
Rifkin A, Quitkin F, Kane J, Klein DF, Ross D. The effect of fluphenazine upon social and vocational functioning in remitted schizophrenics. Biological Psychiatry 1979;14:499‐508.
Rifkin A, Quitkin F, Klein DF. Fluphenazine decanoate, oral fluphenazine, and placebo in treatment of remitted schizophrenics. II. Rating scale data. Archives of General Psychiatry 1977;34:15‐9.
Rifkin A, Quitkin F, Rabiner CJ, Klein DF. Comparison of fluphenazine decanoate, oral fluphenazine, and placebo in remitted outpatient schizophrenics. Psychopharmacology Bulletin 1976;12:24‐6.
Rifkin A, Quitkin F, Rabiner CJ, Klein DF. Fluphenazine decanoate, fluphenazine hydrochloride given orally, and placebo in remitted schizophrenics. I. Relapse rates after one year. Archives of General Psychiatry 1977;34:43‐7.

Roelofs 1974 {published data only}

Roelofs GA. Penfluridol (R 16341) as a maintenance therapy in chronic psychotic patients: a double‐blind clinical evaluation. Acta Psychiatrica Scandinavica 1974;50:219‐24.

Ruskin 1991 {published data only}

Ruskin PE, Nyman G. Discontinuation of neuroleptic medication in older, outpatient schizophrenics. A placebo‐controlled, double‐blind trial. Journal of Nervous and Mental Disease 1991;179:212‐4.

Sampath 1992 {published data only}

Sampath G, Shah A, Krska J, Soni SD. Neuroleptic discontinuation in the very stable schizophrenic patient ‐ relapse rates and serum neuroleptic levels. Human Psychopharmacology 1992;7:255‐64.
Soni SD, Mallik A, Schiff AA. Sulpiride in negative schizophrenia: a placebo‐controlled double‐blind assessment. Human Psychopharmacology 1990;5:233‐8.

Schering Plough 2010 {published data only}

Kane JM, Mackle M, Snow‐Adami L, Zhao J, Szegedi A, Panagides J. A randomised placebo‐controlled trial of asenapine for the prevention of relapse of schizophrenia after long‐term treatment. Journal of Clinical Psychiatry 2011;72(3):349‐55.

Schiele 1961 {published data only}

Schiele BC, Vestre ND, Stein KE. A comparison of thioridazine, trifluoperazine, chlorpromazine, and placebo: a double‐blind controlled study on the treatment of chronic, hospitalized, schizophrenic patients. Journal of Clinical and Experimental Psychopathology and Quarterly Review of Psychiatry and Neurology 1961;22:151‐62.

Shawver 1959 {published data only}

Shawver JR, Gorham DR, Leskin LW, Good WW, Kabnick DE. Comparison of chlorpromazine and reserpine in maintenance drug therapy. Diseases of the Nervous System 1959;20:452‐7.

Spohn 1986 {published data only}

Spohn HE, Coyne L, Larson J, Mittleman F, Spray J, Hayes K. Episodic and residual thought pathology in chronic schizophrenics: effect of neuroleptics. Schizophrenia Bulletin 1986;12:394‐407.

Troshinsky 1962 {published data only}

Troshinsky C, Aaronson HG, Stone RK. Maintenance phenothiazines in aftercare of schizophrenic patients. Pennsylvania Psychiatric Bulletin 1962;2:11‐5.

Vandecasteele 1974 {published data only}

Vandecasteele AJ, Vereecken JL. A double‐blind clinical evaluation of penfluridol (R 16 341) as a maintenance therapy in schizophrenia. Acta Psychiatrica Scandinavica 1974;50:346‐53.

Whittaker 1963 {published data only}

Whittaker CB, Hoy RM. Withdrawal of perphenazine in chronic schizophrenia. British Journal of Psychiatry 1963;109:422‐7.

Wistedt 1981 {published data only}

Wistedt B. A depot neuroleptic withdrawal study. A controlled study of the clinical effects of the withdrawal of depot fluphenazine decanoate and depot flupenthixol decanoate in chronic schizophrenic patients. Acta Psychiatrica Scandinavica 1981;64:65‐84.
Wistedt B. Neuroleptics and depression. Archives of General Psychiatry 1982;39:745.
Wistedt B, Jorgensen A, Wiles D. A depot neuroleptic withdrawal study. Plasma concentration of fluphenazine and flupenthixol and relapse frequency. Psychopharmacology 1982;78:301‐4.
Wistedt B, Palmstierna T. Depressive symptoms in chronic schizophrenic patients after withdrawal of long‐acting neuroleptics. Journal of Clinical Psychiatry 1983;44:369‐71.
Wistedt B, Ranta J. Comparative double‐blind study of flupenthixol decanoate and fluphenazine decanoate in the treatment of patients relapsing in a schizophrenic symptomatology. Acta Psychiatrica Scandinavica 1983;67:378‐88.
Wistedt B, Wiles D, Jorgensen A. A depot neuroleptic withdrawal study neurological effects. Psychopharmacology 1983;80:101‐5.

Zissis 1982 {published data only}

Zissis NP, Psaras M, Lyketsos G. Haloperidol decanoate, a new long‐acting antipsychotic, in chronic schizophrenics: double‐blind comparison with placebo. Current Therapeutic Research 1982;31:650‐5.

References to studies excluded from this review

Allen 1997 {published data only}

Allen DN, Gilbertson MW, Barry E, Kammen DP, Gurklis JA. Haloperidol improves memory in schizophrenia. Proceedings of the 150th Annual Meeting of the American Psychiatric Association; 1997 May 17‐22; San Diego, California, USA. 1997.

Bourin 2008 {published data only}

Bourin M, Debelle M, Heisterberg J, Josiassen MK, Ostergaard JB, Sands E. Efficacy of bifeprunox in patients in the post‐acute, maintenance phase of schizophrenia: findings from a 6‐month study. Schizophrenia Research 2008;98:158.

Branchey 1981 {published data only}

Branchey MH, Branchey LB, Richardson MA. Effects of gradual decrease and discontinuation of neuroleptics on clinical condition and tardive dyskinesia. Psychopharmacology Bulletin 1981;17:118‐20.
Branchey MH, Branchey LB, Richardson MA. Effects of neuroleptic adjustment on clinical condition and tardive dyskinesia in schizophrenic patients. American Journal of Psychiatry 1981;138:608‐12.

Breier 1987 {published data only}

Breier A, Wolkowitz OM, Doran AR, Roy A, Boronow J, Hommer DW, et al. Neuroleptic responsivity of negative and positive symptoms in schizophrenia. American Journal of Psychiatry 1987;144:1549‐55.

Chouinard 1980 {published data only}

Chouinard G, Annable L, Jones BD, Collu R. Lack of tolerance to long‐term neuroleptic treatment in dopamine tuberoinfundibular system. Acta Psychiatrica Scandinavica 1980;64:353‐62.

Claghorn 1974 {published data only}

Claghorn JL, Johnstone EE, Cook TH, Itschner L. Group therapy and maintenance treatment of schizophrenics. Archives of General Psychiatry 1974;31:361‐5.

Collins 1967 {published data only}

Collins AD, Dundas J. A double‐blind trial of amitriptyline‐perphenazine, perphenazine and placebo in chronic withdrawn inert schizophrenics. British Journal of Psychiatry 1967;113:1425‐9.

Condray 1995 {published data only}

Condray R, Van Kammen DP, Steinhauer SR, Kasparek A, Yao JK. Language comprehension in schizophrenia: trait or state indicator?. Biological Psychiatry 1995;38:287‐96.

Curson 1985 {published data only}

Curson DA, Barnes TR, Bamber RW, Platt SD, Hirsch SR, Duffy JC. Long term depot maintenance of chronic schizophrenic out patients: the seven year follow up of the Medical Research Council fluphenazine/placebo trial. II. The incidence of compliance problems, side effects, neurotic symptoms and depression. British Journal of Psychiatry 1985;146:469‐74.
Curson DA, Barnes TR, Bamber RW, Platt SD, Hirsch SR, Duffy JC. Long‐term depot maintenance of chronic schizophrenic out‐patients: the seven year follow‐up of the Medical Research Council fluphenazine‐ placebo trial I. Course of illness, stability of diagnosis, and the role of a special maintenance clinic. British Journal of Psychiatry 1985;146:464‐9.
Curson DA, Barnes TR, Bamber RW, Platt SD, Hirsch SR, Duffy JC. Long‐term depot maintenance of chronic schizophrenic out‐patients: the seven year follow‐up of the Medical Research Council fluphenazine/placebo trial. III. Relapse postponement or relapse prevention? The implications for long‐term outcome. British Journal of Psychiatry 1985;146:474‐80.

Degkwitz 1970 {published data only}

Degkwitz R, Bauer MP, Gruber M, Hampel G, Luxenburger O, Richartz M, et al. Time relationship between the appearance of persisting extrapyramidal hyperkineses and psychotic recurrences following sudden interruption of prolonged neuroleptic therapy of chronic schizophrenic patients. Arzneimittelforschung 1970;20(7):890‐3.

Diamond 1960 {published data only}

Diamond LS, Marks JB. Discontinuance of tranquilizers among chronic schizophrenic patients receiving maintenance dosage. Journal of Nervous and Mental Disease 1960;131:247‐51.

Double 1993 {published data only}

Double DB, Warren GC, Evans M, Rowlands RP. Efficacy of maintenance use of anticholinergic agents. Acta Psychiatrica Scandinavica 1993;88:381‐4.
Elie R, Morin L, Tetreault L. Effects of ethopropazine and trihexyphenidyl on several parameters of the neuroleptic syndrome [Effets de l'ethopropazine et du trihexyphenidyle sur quelques parametres du syndrome neuroleptique]. Encephale 1972;61:32‐52.

Engelhardt 1967 {published data only}

Engelhardt DM, Rosen B, Freedman N, Margolis R. Phenothiazines in prevention of psychiatric hospitalisation. Archives of General Psychiatry 1967;16:98‐101.

Gleeson 2004 {published data only}

Gleeson J, Wade D, Mcgorry P, Albiston D, Castle D, Gilbert M, et al. Episode ii: prevention of relapse following early psychosis. Schizophrenia Research 2004;70:61‐2.

Goldberg 1967 {published data only}

Goldberg SC, Schooler NR, Mattsson N. Paranoid and withdrawal symptoms in schizophrenia: differential symptom reduction over time. Journal of Nervous and Mental Disease 1967;145:158‐62.
Mefferd RBJr, Labrosse EH, Gawienowski AM, Williams RJ. Influence of chlorpromazine on certain biochemical variables of chronic male schizophrenics. Journal of Nervous and Mental Disease 1958;127:167‐79.

Good 1958 {published data only}

Good WW, Sterling M, Holtzman WH. Termination of chlorpromazine with schizophrenic patients. American Journal of Psychiatry 1958;115:443‐8.

Greenberg 1966 {published data only}

Greenberg LM, Roth S. Differential effects of abrupt versus gradual withdrawal of chlorpromazine in hospitalized chronic schizophrenic patients. American Journal of Psychiatry 1966;123:221‐6.

Hine 1958 {published data only}

Hine FR. Chlorpromazine in schizophrenic withdrawal and in the withdrawn schizophrenic. Journal of Nervous and Mental Disease 1958;127:220‐7.

Hunt 1967 {published data only}

Hunt PV. A comparison of the effects of oxypertine and trifluoperazine in withdrawn schizophrenics. British Journal of Psychiatry 1967;113:1419‐24.

Ionescu 1983 {published data only}

Ionescu R, Tiberiu C, Miklos R, Angelescu C, Persiceanu AM. Penfluridol in the maintenance therapy of schizophrenia. Neurologie et Psychiatrie 1983;21:33‐41.

Janecek 1963 {published data only}

Janecek J, Schiele BC, Bellville T, Anderson R. The effects of withdrawal of trifluoperazine on patients maintained on the combination of tranylcypromine and trifluoperazine: a double‐blind study. Current Therapeutic Research, Clinical and Experimental 1963;85:608‐15.

Johnstone 1988 {published data only}

Johnstone EC, Crow TJ, Frith CD, Owens DG. The Northwick Park "functional" psychosis study: diagnosis and treatment response. Lancet 1988;2:119‐25.
Johnstone EC, Crow TJ, Owens DGC, Frith CD. The Northwick Park 'functional' psychosis study. Phase 2 ‐ maintenance treatment. Journal of Psychopharmacology 1991;5:388‐95.
Johnstone EC, Owens DGC. Does early treatment have an effect on outcome?. Proceedings of the 10th European College of Neuropsychopharmacology Congress; 1997 Sep 13‐17; Vienna, Austria. 1997.

Kellam 1971 {published data only}

Kellam AM, Jones KS. A double‐blind controlled trial of thiothixene and perphenazine in chronic schizophrenics shown to require maintenance therapy. Acta Psychiatrica Scandinavica 1971;47:174‐85.

Lauriello 2005 {published data only}

Lauriello J, McEvoy JP, Rodriguez S, Bossie CA, Lasser RA. Long‐acting risperidone vs. placebo in the treatment of hospital inpatients with schizophrenia. Schizophrenia Research 2005;72:249‐58.

Lecrubier 1997 {published data only}

Lecrubier Y. Amisulpride in deficit schizophrenia. Proceedings of the 6th World Congress Biological Psychiatry, 1997 Jun 22‐27; Nice, France. 1997.

Loo 1997 {published data only}

Loo H, Poirier‐Littre MF, Theron M, Rein W, Fleurot O. Amisulpride versus placebo in the medium‐term treatment of the negative symptoms of schizophrenia. British Journal of Psychiatry 1997;170:18‐22.

Mefferd 1958 {published data only}

Mefferd RB, Labrosse EH, Gawienowski AM, Williams RJ. Influence of chlorpromazine on certain biochemical variables of chronic male schizophrenics. Journal of Nervous and Mental Disease 1958;127(2):167‐79.

Mosher 1975 {published data only}

Bola JR, Mosher LR. Treatment of acute psychosis without neuroleptics: two‐year outcomes from the Soteria project. Journal of Nervous and Mental Disease 2003;191:219‐29.
Matthews SM, Roper MT, Mosher LR, Menn AZ. A non neuroleptic treatment for schizophrenia: analysis of the two year postdischarge risk of relapse. Schizophrenia Bulletin 1979;5:322‐33.
Mosher LR, Menn A, Matthew SM. Soteria: evaluation of a home‐based treatment for schizophrenia. American Journal of Orthopsychiatry 1975;45:455‐67.
Mosher LR, Menn AZ. Community residential treatment for schizophrenia: two year follow up. Hospital and Community Psychiatry 1978;29:715‐23.

Müller 1982 {published data only}

Müller P, Hartmann W, Jung F, Kind J, Lohrengel S, Steuber H. To prevent relapse of schizophrenic psychoses [Zur Rezidivprophylaxe schizophrener Psychosen]. Ferdinand Enke Verlag Stuttgart 1982;14:1‐144.

Paul 1972 {published data only}

Paul GL, Tobias LL, Holly BL. Maintenance psychotropic drugs in the presence of active treatment programs. A "triple‐blind" withdrawal study with long‐term mental patients. Archives of General Psychiatry 1972;27:106‐15.

Peet 1981 {published data only}

Peet M, Middlemiss DN, Yates RA. Propranolol in schizophrenia II. Clinical and biochemical aspects of combining propranolol with chlorpromazine. British Journal of Psychiatry 1981;138:112‐7.

Pickar 1986 {published data only}

Pickar D, Labarca R, Doran AR, Wolkowitz OM, Roy A, Breier A, et al. Longitudinal measurement of plasma homovanillic acid levels in schizophrenic patients. Correlation with psychosis and response to neuroleptic treatment. Archives of General Psychiatry 1986;43:669‐76.

Pickar 2003 {published data only}

Pickar D, Bartko JJ. Effect size of symptom status in withdrawal of typical antipsychotics and subsequent clozapine treatment in patients with treatment‐resistant schizophrenia. American Journal of Psychiatry 2003;160:1133‐8.

Pigache 1993 {published data only}

Pigache RM. Effects of placebo, orphenadrine, and rising doses of chlorpromazine, on PAT performance in chronic schizophrenia. A two year longitudinal study. Schizophrenia Research 1993;10:51‐9.
Pigache RM. The clinical relevance of an auditory attention task (PAT) in a longitudinal study of chronic schizophrenia with placebo substitution for chlorpromazine. Schizophrenia Research 1993;10:39‐50.
Pigache RM, Norris HN. Measurement of drug action in schizophrenia. Clinical Science 1973;44:28P.
Pigache RM, Norris HN. Selective attention as an index of the anti‐psychotic action of chlorpromazine in schizophrenia. British Psychological Society Bulletin 1973;20:160.

Rassidakis 1970 {published data only}

Rassidakis NC, Kondakis X, Papanastassiou A, Michalakeas A. Withdrawal of antipsychotic drugs from chronic psychiatric patients. Bulletin of the Menninger Clinic 1970;34(4):216‐22.

Ravaris 1965 {published data only}

Ravaris CL, Weaver LA, Brooks GW. A controlled study of fluphenazine enanthate in chronic schizophrenic patients. Diseases of the Nervous System 1965;25:33‐9.
Ravaris CL, Weaver LA, Brooks GW. Further studies with fluphenazine enanthate: II. Relapse rate in patients deprived of medication. American Journal of Psychiatry 1967;124:248‐9.

Schlossberg 1978 {published data only}

Schlossberg A, Shadmi M. A comparative controlled study of two long‐acting phenothiazines: pipotiazine palmitate and fluphenazine decanoate. Current Therapeutic Research 1978;23(5):642‐54.

Singh 1990 {published data only}

Singh H, Hunt JI, Vitiello B, Simpson GM. Neuroleptic withdrawal in patients meeting criteria for supersensitivity psychosis. Journal of Clinical Psychiatry 1990;51:319‐21.

Smelson 2006 {published data only}

Smelson DA, Tunis SL, Nyhuis AW, Faries DE, Kinon BJ, Ascher‐Svanum H. Antipsychotic treatment discontinuation among individuals with schizophrenia and co‐occurring substance use. Journal of Clinical Psychopharmacology 2006;26:666‐7.

Soni 1990 {published data only}

Soni SD, Mallik A, Schiff AA. Sulpiride in negative schizophrenia: a placebo‐controlled double‐blind assessment. Human Psychopharmacology 1990;5:233‐8.

Tollefson 1999 {published data only}

Dellva MA, Tollefson GD, Mattler CA, Kinon BJ, Grundy SL. A controlled, double‐blind investigation of the clozapine discontinuation syndrome with conversion to either olanzapine or placebo. Schizophrenia Research 1999;36:277‐8.
Tollefson GD, Dellva MA, Mattler CA, Kane JM, Wirshing DA, Kinon BJ, et al. Controlled, double‐blind investigation of the clozapine discontinuation symptoms with conversion to either olanzapine or placebo. Journal of Clinical Psychopharmacology 1999;19:435‐43.
Tollefson GD, Dellva MA, Mattler CA, Kinon BJ, Grundy SL. A controlled, double‐blind investigation of the clozapine discontinuation syndrome with conversion to either olanzapine or placebo. Proceedings of the 11th European College of Neuropsychopharmacology Congress; 1998 Oct 31 ‐ Nov 4; Paris, France. 1998.

Vaddadi 1986 {published data only}

Vaddadi KS, Gilleard CJ, Mindham RH, Butler R. A controlled trial of prostaglandin E1 precursor in chronic neuroleptic resistant schizophrenic patients. Psychopharmacology 1986;88:362‐7.

Van Kammen 1982 {published data only}

Van Kammen DP, Bunney WE, Docherty JP, Marder SR, Ebert MH, Rosenblatt JE, et al. D‐Amphetamine‐induced heterogeneous changes in psychotic behavior in schizophrenia. American Journal of Psychiatry 1982;139:991‐7.
Van Kammen DP, Bunney WE, Docherty JP, Jimerson DC, Post RM, Siris P, et al. Amphetamine‐induced catecholamine activation in schizophrenia and depression: behavioral and physiological effects. Advances in Biochemical Psychopharmacology 1977;16:655‐9.
Van Kammen DP, Docherty JP, Marder SR, Schulz SC, Bunney WE. Lack of behavioral supersensitivity to d‐amphetamine after pimozide withdrawal. A trial with schizophrenic patients. Archives of General Psychiatry 1980;37:287‐90.
Van Kammen DP, Docherty JP, Marder SR, Siris SG, Bunney WE. D amphetamine raises serum prolactin in man: evaluations after chronic placebo, lithium and pimozide treatment. Life Sciences 1978;23:1487‐92.

Van Praag 1973 {published data only}

Van Praak HM, Dols LCW. Fluphenazine enanthate and fluphenazine decanoate: a comparison of their duration of action and motor side effects. American Journal of Psychiatry 1973;130:801‐4.

Wiedemann 2001 {published data only}

Wiedemann G, Hahlweg K, Muller U, Feinstein E, Hank G, Dose M. Effectiveness of targeted intervention and maintenance pharmacotherapy in conjunction with family intervention in schizophrenia. European Archives of Psychiatry and Clinical Neuroscience 2001;251:72‐84.

Wright 1964 {published data only}

Wright RLD, Lynes PG. Value of continuous drug administration for chronic long‐term mental hospital patients. Canadian Psychiatric Association Journal 1964;60:352‐7.

Wunderink 2006 {published data only}

Wunderink A, Nienhuis, Sytema S, Wiersma D. Guided discontinuation versus maintenance treatment in remitted first episode psychosis: relapse rates and functional outcome. Schizophrenia Research 2006;86:S51.

Zeller 1956 {published data only}

Zeller WW, Graffagnino PN, Cullen CF, Rietman HJ. Use of chlorpromazine and reserpine in the treatment of emotional disorders. Journal of the American Medical Association 1956;160:179‐84.

Zwanikken 1973 {published data only}

Zwanikken 1973. Penfluridol (R 16341). A long‐acting oral neuroleptic, as maintenance therapy for schizophrenic and mentally retarded patients. Psychiatria, Neurologia, Neurochirurgia 1973;76:83‐92.

Eerdekens 2010 {published data only}

Eerdekens. Evaluate the efficacy in the prevention of recurrence of the symptoms of schizophrenia. Sponsor: Johnson & Johnson Pharmaceutical Research & Development, LL.C2010. [CTG: NCT00111189]

Almerie 2007

Almerie MQ, Alkhateeb H, Essali A, Matar HE, Rezk E. Cessation of medication for people with schizophrenia already stable on chlorpromazine. Cochrane Database of Systematic Reviews 2007, Issue 1. [DOI: 10.1002/14651858.CD006329]

Altman 1996

Altman DG, Bland JM. Detecting skewness from summary information. British Medical Journal 1996;313:1200.

Baldessarini 1985

Baldessarini RJ. Chemotherapy in psychiatry: principles and practice. Cambridge, MA: Harvard University Press, 1985.

Barnes 1989

Barnes TR. A rating scale for drug‐induced akathisia. British Journal of Psychiatry 1989;154:672‐6.

Berger 2003

Berger M. Psychische Erkrankungen. Klinik und Therapie. 2nd Edition. München: Urban & Fischer, 2003.

Bland 1997

Bland JM, Kerry SM. Statistics notes. Trials randomised in clusters. British Medical Journal 1997;315:600.

Boissel 1999

Boissel JP, Cucherat M, Li W, Chatellier G, Gueyffier F, Buyse M, et al. The problem of therapeutic efficacy indices. 3. Comparison of the indices and their use. Therapie 1999;54(4):405‐11.

Borenstein 2006 [Computer program]

Borenstein M, Hedges LV, Higgins JPT, Rothstein H. Comprehensive Meta‐analysis Version 2. Biostat, 2006.

Carpenter 1994

Carpenter WT, Buchanan RW. Schizophrenia. New England Journal of Medicine 1994;330:681‐90.

Davis 1975

Davis JM. Overview: maintenance therapy in psychiatry: I. Schizophrenia. American Journal of Psychiatry 1975;132(12):1237‐45.

Davis 1989

Davis JM, Barter JT, Kane JM. Antipsychotic drugs. In: Kaplan HI, Sadock BJ editor(s). Comprehensive Textbook of Psychiatry. 5th Edition. Baltimore: Williams & Wilkins, 1989:1591‐1626.

Deeks 2000

Deeks J. Issues in the selection for meta‐analyses of binary data. Proceedings of the 8th International Cochrane Colloquium; 2000 Oct 25‐28; Cape Town, South Africa. 2000.

Der‐Simonian 1986

Der‐Simonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7:177‐88.

Divine 1992

Divine GW, Brown JT, Frazer LM. The unit of analysis error in studies about physicians' patient care behavior. Journal of General Internal Medicine 1992;7:623‐9.

Duggan 2005

Duggan L, Fenton M, Rathbone J, Dardennes R, El‐Dosoky A, Indran S. Olanzapine for schizophrenia. Cochrane Database of Systematic Reviews 2005, Issue 2. [DOI: 10.1002/14651858.CD001359.pub2]

Duval 2000

Duval S, Tweedie R. Trim and fill: a simple funnel‐plot‐based method of testing and adjusting for publication bias in meta‐analysis. Biometrics 2000;56:455‐63.

Egger 1997

Egger M, Davey‐Smith G, Schneider M, Minder CSO. Bias in meta‐analysis detected by a simple, graphical test. British Medical Journal 1997;13:629‐34.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JP, Curtin F, Worthington HV, Vail A. Meta‐analyses involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31:140‐9.

Endicott 1976

Endicott J, Spitzer RL, Fleiss JL, Cohen J. The global assessment scale. A procedure for measuring overall severity of psychiatric disturbance. Archives of General Psychiatry 1976;33(6):766‐71.

Gilbert 1995

Gilbert P, Harris MJ, McAdams LA, Bowers MB, Charney DS, Glazer WM. Neuroleptic withdrawal in schizophrenic patients. A review of the literature. Archives of General Psychiatry 1995;52:173‐88.

Gulliford 1999

Gulliford MC, Ukoumunne OC, Chinn S. Components of variance and intraclass correlations for the design of community‐based surveys and intervention studies: data from the Health Survey for England 1994. American Journal of Epidemiology 1999;149:876‐83.

Guy 1976

Guy W. ECDEU assessment manual for psychopharmacology‐revised 1976. Washington, DC: DHEW. National Institute of Mental Health, 1976.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.2 [updated September 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Kane 1988

Kane JM, Honigfeld G, Singer J, Meltzer H, and the Clozaril Collaborative study group. Clozapine for the treatment‐resistant schizophrenic. A double‐blind comparison with chlorpromazine. Archives of General Psychiatry 1988;45:789‐96.

Kay 1986

Kay SR, Opler LA, Fiszbein A. Positive And Negative Syndrome Scale (PANSS) Manual. North Tonawanda, NY: Multi‐Health Systems, 1986.

Kellner 1973

Kellner R, Sheffield BF. A self‐rating scale of distress. Psychological Medicine 1973;3:88‐100.

Kishimoto 2012

Kishimoto T, Robenzadeh A, Leucht C, Leucht S, Watanabe K, Mimura M, et al. New results alter balance of evidence of long‐acting injectable vs. oral antipsychotics regarding relapse prevention in schizophrenia: a systematic review and meta‐analysis. in press.

Klein 1969

Klein DF, Davis JM. Diagnosis and Drug Treatment of Psychiatric Disorders. Baltimore: Williams & Wilkins, 1969.

Leon 2006

Leon AC, Mallinckrodt CH, Chuang‐Stein C, Archibald DG, Archer GE, Chartier K. Attrition in randomised controlled clinical trials: methodological issues in psychopharmacology. Biological Psychiatry 2006;59:1001‐5.

Leucht 2003

Leucht S, Barnes TR, Kissling W, Engel RR, Correll C, Kane JM. Relapse prevention in schizophrenia with new‐generation antipsychotics: a systematic review and exploratory meta‐analysis of randomised, controlled trials. American Journal of Psychiatry 2003;160:1209‐22.

Leucht 2005a

Leucht S, Kane JM, Kissling W, Hamann J, Etschel E, Engel R. Clinical implications of Brief Psychiatric Rating Scale scores. British Journal of Psychiatry 2005;187:366‐71.

Leucht 2005b

Leucht S, Kane JM, Kissling W, Hamann J, Etschel E, Engel RR. What does the PANSS mean?. Schizophrenia Research 2005;79(2‐3):231‐8. [PUBMED: 15982856]

Leucht 2009

Leucht S, Corves C, Arbter D, Engel R, Li C, Davis JM. A meta‐analysis comparing second‐generation and first‐generation antipsychotics for schizophrenia. Lancet 2009;373:31‐41.

Leucht 2009a

Leucht S, Komossa K, Rummel‐Kluge C, Corves C, Hunger H, Schmid F, et al. A meta‐analysis of head to head comparisons of second generation antipsychotics in the treatment of schizophrenia. American Journal of Psychiatry 2009;166:152‐63.

Leucht 2009b

Leucht S, Arbter D, Engel RR, Kissling W, Davis JM. How effective are second‐generation antipsychotic drugs? A meta‐analysis of placebo‐controlled trials. Molecular Psychiatry 2009;14:429‐47.

Leucht 2011

Leucht C, Heres S, Kane JM, Kissling W, Davis JM, Leucht S. Oral versus depot antipsychotic drugs for schizophrenia ‐ a critical systematic review and meta‐analysis of randomised long‐term trials. Schizophrenia Research 2011;127:83‐92.

Marshall 2000

Marshall M, Lockwood A, Bradley C, Adams C, Joy C, Fenton M. Unpublished rating scales: a major source of bias in randomised controlled trials of treatments for schizophrenia. British Journal of Psychiatry 2000;176:249‐52.

Marvaha 2004

Marwaha S, Johnson S. Schizophrenia and employment? A review. Social Psychiatry and Psychiatric Epidemiology 2004;39:337‐49.

Moher 2001

Moher D, Schulz KF, Altman D. The CONSORT statement: revised recommendations for improving the quality of reports of parallel‐group randomised trials. Journal of the American Medical Association 2001;285:1987‐1.

Moncrieff 2006

Moncrieff J. Does antipsychotic withdrawal provoke psychosis? Review of the literature on rapid onset psychosis (supersensitivity psychosis) and withdrawal‐related relapse. Acta Psychiatrica Scandinavica 2006;114(1):3‐13.

Overall 1962

Overall JE, Gorham DR. The Brief Psychiatric Rating Scale. Psychological Reports 1962;10:799‐12.

Papanikolaou 2004

Papanikolaou PN, Churchill R, Wahlbeck K, Ioannidis JP. Safety reporting in randomised trials of mental health interventions. American Journal of Psychiatry 2004;161:1692‐7.

Peters 2008

Peters JL, Sutton AJ, Jones DR, Abrams KR, Rushton L. Contour‐enhanced meta‐analysis funnel plots help distinguish publication bias from other causes of asymmetry. Journal of Clinical Epidemiology 2008;61:991‐6.

Pope 2010

Pope A, Adams C, Paton C, Weaver T, Barnes TR. Assessment of adverse effects in clinical studies of antipsychotic medication: survey of methods used. British Journal of Psychiatry 2010;197:67‐72.

Ray 2009

Ray WA, Chung CP, Murray KT, Hall K, Stein CM. Atypical antipsychotic drugs and the risk of sudden cardiac death. New England Journal of Medicine 2009;360:225‐35.

Robinson 1999

Robinson D, Woerner MG, Alvir JM, Bilder R, Goldman R, Geisler S, et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Archives of General Psychiatry 1999;56:241‐7.

Schulz 1995

Schulz KF, Chalmers I, Hayes RJ, Altman DG. Empirical evidence of bias. Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. Journal of the American Medical Association 1995;273:408‐12.

Simpson 1970

Simpson EN, Angus JWF. A rating scale for extrapyramidal side‐effects. Acta Psychiatrica Scandinavica Supplementum 1970;212:11‐9.

Srisurapanont 2004

Srisurapanont M, Maneeton B, Maneeton N. Quetiapine for schizophrenia. Cochrane Database of Systematic Reviews 2004, Issue 2. [DOI: 10.1002/14651858.CD000967.pub2]

Stata 2002 [Computer program]

Stata Corporation. Intercooled Stata 7.0 for Windows. Stata Corporation, 2002.

Tiihonen 2009

Tiihonen J, Lönnqvist J, Wahlbeck K, Klaukka T, Niskanen L, Tanskanen A, et al. 11‐year follow‐up of mortality in patients with schizophrenia: a population‐based cohort study (FIN11 study). Lancet 2009;374:620‐7.

Tsuang 1978

Tsuang MT. Suicide in schizophrenics, manics, depressives, and surgical controls: a comparison with general population suicide mortality. Archives of General Psychiatry 1978;35:153‐55.

Viguera 1997

Viguera AC, Baldessarini RJ, Hegarty JD, van Kammen DP, Tohen M. Clinical risk following abrupt and gradual withdrawal of maintenance neuroleptic treatment. Archives of General Psychiatry 1997;54:49‐55.

Wahlbeck 1999

Wahlbeck K, Cheine M, Essali MA, Rezk E. Clozapine vs typical neuroleptic medication for schizophrenia. Cochrane Database of Systematic Reviews 1999, Issue 2. [DOI: 10.1002/14651858.CD000059]

Walsh 2002

Walsh E, Buchanan A, Fahy T. Violence and schizophrenia: examining the evidence. British Journal of Psychiatry 2002;180:490‐5.

Weinmann 2009

Weinmann S, Read J, Aderhold V. Influence of antipsychotics on mortality in schizophrenia: systematic review. Schizophrenia Research 2009;113(1):1‐11.

Wilkinson 2000

Wilkinson G, Hesdon B, Wild D, Cookson R, Farina C, Sharma V, et al. Self‐report quality of life measure for people with schizophrenia: the SQLS. British Journal of Psychiatry 2000;177:42‐6.

Xia 2009

Xia J, Adams CE, Bhagat N, Bhagat V, Bhoopathi P, El‐Sayeh H, et al. Loss to outcomes stakeholder survey: the LOSS study. Psychiatric Bulletin 2009;33(7):254‐7.

References to other published versions of this review

Leucht 2012

Leucht S, Tardy M, Komossa K, Heres S, Kissling W, Salanti G, et al. Antipsychotic drugs versus placebo for relapse prevention in schizophrenia: systematic review and meta‐analysis. Lancet in press.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Andrews 1976

Methods

Randomisation: randomised, no further details.
Allocation: pharmacists held the key.
Blinding: double, identical capsules.
Duration: 42 weeks.
Design: parallel.
Location: single‐centre.
Setting: hospital.

Participants

Diagnosis: schizophrenia (clinical diagnosis), continuously in hospital for at least 6 years (mean 28 years).
N=32.
Gender: 32 men.
Age: mean 58 years.
History: duration stable‐8 weeks, duration ill NI‐ mean duration of hospitalisation 28 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean Wing Behaviour Scale Withdrawal Score 2.14, baseline antipsychotic dose‐216mg/day CPZ equivalent

Interventions

1. Drug: Chlorpromazine ‐ mean dose: 216mg/day. N=15.

Allowed dose range: the participants were kept on their initial dose.

2. Placebo: Duration of taper 0 days. N=17.

Rescue medication: benzodiazepines, anticholinergics.

Outcomes

Examined:

Relapse (need of antipsychotic medication).

Leaving the study early.

Unable to use / Not included:

Behaviour: Ward Behaviour Rating Scale of Wing (no SD / no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Low risk

Pharmacists held the key.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed the trial.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No obvious other bias.

Arato 2002

Methods

Randomisation: random, computer‐generated randomisation code.
Allocation: procedure not described.
Blinding: double.
Duration: 12 months.
Design: parallel.
Location: multi‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic, stable schizophrenia (DSM‐III‐R), less than markedly ill on Clinical Global Impression Scale.

N=278.
Gender: 203 men, 75 women.
Age: mean 49.7 years.
History: duration stable‐ n.i., duration ill‐ mean 21.8 years, number of previous hospitalisations‐ mean 10.1, age at onset‐ mean 27.9 years, severity of illness‐ mean PANSS 85.8, mean CGI severity 4.02, baseline antipsychotic dose n.i..  

Interventions

1. Drug: ziprasidone ‐ Fixed doses of 40, 80 or 160 mg/day.** N=207.

2. Placebo: Duration of taper <3 days. N=71.

Rescue medication: anticholinergics, lorazepam, temazepam, no additional antipsychotic medication.

Outcomes

Examined:

Relapse: (Clinical Global Impressionof much worse or more, PANSS items hostility or uncooperativeness > 6, or in need for additional treatment for exacerbation of symptoms).

Leaving the study early.

Adverse events: binary outcome for generel, specific (movement disorders) ‐ interviews.

Unable to use / Not included:

Mental state: PANSS total score and subscores (no predefined outcome of interest).

Global state: much worse or more ‐ Clinical Global Impression Severity Scale (no prespecified outcome of interest).

Functioning: Global Assessment of Functioning Scale (no prespecified outcome of interest).

Adverse effects: extrapyramidal symptoms (Simpson Angus Scale, Barnes Akathisia Scale, Abnormal Involuntary Movements Scale ‐ all no SD / continous side‐effect results were not among the prespecified outcomes of interest).

Physiological measures: ECG, vital signs, weight, ophthalmological assessment, lab tests (all no SD, no data / not prespecified outcomes of interest).

Notes

** The results of the three dose groups were pooled. 16 participants from one centre were excluded due to protocol violations. Intention‐to‐treat were only those participants who had received at least one dose. How many did not receive one dose is unclear.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised, computer‐generated randomised code.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

64% of the participants left the study early, most due to relapse. The rate was higher in the placebo group (86%) than in the medication group (˜57%). This was probably not a problem for the primary outcome relapse, but for secondary outcomes for which the last‐observation‐carried‐forward method was used. Appropriate survival curve analysis was used for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

No selective reporting.

Other bias

Low risk

No obvious other bias.

Baro 1970

Methods

Randomisation: matched pairs were formed and then randomised, no further details.
Allocation: procedure not described.
Blinding: double, indistinguishable placebo.
Duration: 10 weeks.
Design: parallel.
Location: single‐centre.
Setting: hospital, sponsored.

Participants

Diagnosis: chronic psychotic hospitalised patients mainly with schizophrenic and paranoid behaviour patterns, suspected of relapsing after withdrawal of medication (clinical diagnosis).
N=26.
Gender: 26 men.
Age: n.i.
History: duration stable‐ 8 months pre‐treatment with penfluridol to find optimum dose, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., but hospitalised, baseline antipsychotic dose‐ 23.4mg/day.

Interventions

1. Drug: penfluridol once weekly ‐ Fixed dose, mean dose: n.i., range 10‐40mg/weekly. N=13.

2. Placebo: Duration of taper: 0 days. N=13.

Rescue medication: sedative neuroleptics allowed for 2 weeks, dexbenzitide.

Outcomes

Examined:

Relapse: need of medication as decided by two psychiatrists.

Unable to use / Not included:

Mental state: Psychiatric Evaluation Scale (no predefined outcome of interest).

Adverse effects: movement disorders (Factor Construct Outcome Scale, no data for randomised phase / continous side‐effect results were not among the prespecified outcomes of interest), neurologic effects (graphometric and tapping test, no data for randomised phase / no prespecified outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Matched pairs were formed and then randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, indistinguishable placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, indistinguishable placebo.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Apart from those participants who relapsed no participant left the study early and relapse was the only outcome.

Selective reporting (reporting bias)

High risk

Adverse events were not reported for the double‐blind phase.

Other bias

Low risk

No obvious other bias.

Beasley 2003

Methods

Randomisation: randomised, 2:1 ratio, by an interactive voice response system.
Allocation: interactive voice response system.
Blinding: double, no further details.
Duration: one year, but the study was terminated early. Maximum length was 30 weeks.
Design: parallel.
Location: multi‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (n=266) or schizoaffective disorder (n=60, DSM‐IV). BPRS total score <36, positive symptoms at most mild, Global Assessment of Functioning at least 40, currently on maintenance antipsychotic medication.

N=326.
Gender: 173 men, 153 women.
Age: mean 35.9 years.
History: duration stable‐ 8 weeks, duration ill‐ mean 11.1 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 24.7 years, severity of illness‐ mean PANSS total score at baseline 43, baseline antipsychotic dose‐ mean 13.4 mg olanzapine/day.

Interventions

Participants were first converted to olanzapine and then stabilized for 8 weeks before randomisation.

1. Drug: olanzapine ‐ Fixed dose of either 10, 15 or 20 mg/day. Mean dose 13.4 mg/day. N=224.

2. Placebo: Duration of taper: 0 days. N=102.

Rescue medication: a one time increase of the same medication (olanzapine or placebo) was allowed. Furthermore, antiparkinson medication and benzodiazepines were allowed.

Outcomes

Examined:

Relapse: any BPRS positive item > 4, absolute increase of a positive item or of the positive subscore, hospitalisation due to positive symptoms, suicide or suicide attempt.

Leaving the study early.

Adverse effects: binary outcomes for general, specific (movement disorders) ‐ open interviews.

Quality of life: Heinrich Carpenter Quality of Life Scale.

Unable to use / Not included:

Mental state: PANSS (no prespecified outcome of interest).

Adverse effects: adverse effects with an incidence < 10% (no data), laboratory, EPS‐scales (in part no data / no prespecified outcome of interest), EPS‐scales (no SD / continous side‐effect results were not among the prespecified outcomes).

Physiological measures: vital signs (no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised, 2:1 ratio, by an interactive voice response system.

Allocation concealment (selection bias)

Low risk

Interactive voice response system.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition of 26% was acceptable, but many more participants in the placebo group than in the olanzapine group left the study early. Kaplan‐Meier survival analysis was used for the analysis of relapse, ANOVA based on last‐observation‐carried‐forward was used for continuous outcomes.

Selective reporting (reporting bias)

High risk

Only those adverse events with a frequency of at least 10% were reported. Use of antiparkinson medication has not been reported.

Other bias

High risk

The study was terminated early when there was a sufficient difference, but this was preplanned.

Blackburn 1981

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, thiamine chloride used as placebo, participants and nurses were told that a new medication was given, but nurses soon new that this was a placebo.
Duration: 16 weeks.
Design: parallel.
Location: single‐centre.
Setting: in hospital.

Participants

Diagnosis: schizophrenia (clinical diagnosis).

N=45.
Gender: 45 men.
Age: 20‐40 years.
History: duration stable‐ n.i., but clinically tranquilised and making a satisfactory adjustment on phenothiazine medication, duration ill‐ n.i., but mean length of current hospitalisation 45 months (range 3‐129), number of previous hospitalisations‐ all more than one, age at onset‐ n.i., severity of illness‐ n.i., but all in open hospital ward, baseline antipsychotic dose‐ prochlorpromazine 15‐150mg/day, perphenazine 12‐24mg/day, chlorpromazine 50‐800mg/day, promazine 200‐400mg/day, trifluoperazine 6mg/day.

Interventions

1. Drug: prochlorpromazine, perphenazine, chlorpromazine, promazine or trifluoperazine. Fixed doses continued with the same drug and dose taken before the study. Mean dose: n.i. N=30.

2. Placebo: Duration of taper: 0 days. N=15*.

Rescue medication: not allowed.

Outcomes

Examined:

Relapse (need of medication or deterioriation of state or transfer to closed ward)

Unable to use / Not included:

Behaviour: Patient Adjustment Report (no prespecified outcome of interest).

Mental state: Taylor Manifest Anxiety Scale (no prespecified outcome of interest).

Notes

* Another 15 participants were treated only for 8 weeks with placebo and then switched back to their initial antipsychotic drug.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

High risk

Double, thiamine chloride used as placebo, participants and nurses were told that a new medication was given, but nurses soon new that this was a placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, thiamine chloride used as placebo, participants and nurses were told that a new medication was given, but nurses soon new that this was a placebo.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only completers were included in the statistical analysis, but because the drop‐out rate was only 13% we did not consider this a source of bias.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other risk of bias.

Boonstra 2011

Methods

Randomisation: An independent rater created randomisation lists stratified for gender with randomly permuted blocks of 4 allocation groups.

Allocation: procedure not described.
Blinding: open.
Duration: 24 months.
Design: parallel.
Location: multi‐center.
Setting: outpatient.

Participants

Diagnosis: first episode schizophrenia (DSM‐IV).

N=20.
Gender: 17 men, 3 women.
Age: mean 29.8 years.
History: duration stable‐ 1 year, duration ill‐ 2.6 years, number of previous hospitalisations‐ 0, age at onset‐ 27.3 years, severity of illness‐ PANSS total score 49, baseline antipsychotic dose‐ 3mg/day haloperidol equivalents (olanzapine, risperidone, quetiapine, zuclopenthixol).

Interventions

1. Drug: olanzapine, risperidone, quetiapine, zuclopenthixol. Flexible doses. Mean dose: n.i. N=9.

2. No treatment: Duration of taper: 6‐12 weeks. N=11.

Rescue medication: not indicated.

Outcomes

Examined:

Relapse: clinicial judgement.

Leaving the study early.

Rehospitalisation.

Notes

Sponsor: The Netherlands Organisation for Health Research and Development and EliLilly.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An independent rater created randomisation lists stratified for gender with randomly permuted blocks of 4 allocation groups.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

High risk

Open study.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Open study.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

5 out of 20 participants left the study early (25%). Probably an acceptable rate, there was no big difference between drug and placebo group. Kaplan‐Meier survival curves were used for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Premature termination after interim analysis.

Caffey 1964

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double, identical tablets. However, placebo dose reduction group received medication only every other day. Therefore, blinding was not fully maintained.
Duration: 16 weeks.
Design: parallel.
Location: multi‐centre.
Setting: in hospital.

Participants

Diagnosis: schizophrenia (clinical diagnosis), one third paranoid subtype, without central nervous system disease, without lobotomy.

N=259.
Gender: all men.
Age: mean 40 years.
History: duration stable‐ stable doses for at least 3 months before the study, duration ill‐ n.i., but currently hospitalized for a mean of 10 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ chlorpromazine mean 400mg/day, thioridazine mean 350mg/day.

Interventions

1. Drug: chlorpromazine or thioridazine.* Fixed dose, continuation of the dose given in the stabilization phase. Mean dose: chlorpromazine mean 400mg/day, thioridazine mean 350mg/day. N=88.

2. Placebo: Duration of taper: 1 ‐ 8 days. N=171.

Rescue medication: not indicated.

Outcomes

Examined:

Relapse: definitive worsening of the condition and medication again necessary, usually joint decision of treatment team.

Unable to use / Not included:

Mental state: Inpatient Multidimensional Psychiatric Scale (no prespecified outcome of interest).

Behaviour: Psychotic Reaction Profile Scale (no prespecified outcome of interest).

Notes

* There was another group which received half the original dose. It was not considered in this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

High risk

Double, identical tablets. However, placebo dose reduction group received medication only every other day. Therefore, blinding was not fully maintained.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical tablets. However, placebo dose reduction group received medication only every other day. Therefore, blinding was not fully maintained.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear whether there were dropouts or whether the authors analysed only study completers.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Unclear risk

22 participants who had relapsed in the first 8 weeks were entered in the study again. As the number is small, it is unclear whether they affected the results.

Channabasavanna 1987

Methods

Randomisation: n.i., but double‐blind study.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 12 weeks.
Design: parallel.
Location: single‐centre.
Setting: unclear.

Participants

Diagnosis: chronic schizophrenia (DSM‐III), all on maintenance medication for control of continuous symptoms, all stable for at least 6 months.

N=30.
Gender: 16 men, 12 women.
Age: mean 36.0 years.
History: duration stable‐ at least 6 months, duration ill‐ mean 11.1 years, number of previous hospitalisations‐ n.i., age at onset‐ 24.9 years, severity of illness‐ n.i., baseline antipsychotic dose‐ mean 297.5 mg/day chlorpromazine equivalent.

Interventions

1. Drug: penfluridol. Fixed dose of 55mg/week. N=15.

2. Placebo: Duration of taper: 0 days. N=15.

Rescue medication: antiparkinson medication and haloperidol, but this was considered to be a relapse.

Outcomes

Examined:

Relapse (need of additional haloperidol medication).

Unable to use / Not included:

Mental state (Scale for the Assessment of Positive Symptoms and Negative Symptoms ‐ no data /no predefined outcome of interest).

Adverse effects: extrapyramidal side‐effects (Simpson Angus Scale ‐ no data / continuous side‐effect results were not among the prespecified outcomes).

Physiological measures: mean body weight, pulse rate, blood pressure, laboratory (all no data / no prespecified outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

N.i., but double‐blind study.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only study completers were used in the final analysis, but as there were only two dropouts (one in each group) this was not necessarily a problem.

Selective reporting (reporting bias)

Low risk

Rating scale results were not reported, but these were not of interest for the review.

Other bias

Low risk

No clear evidence for other bias.

Chen 2010

Methods

Randomisation: sequence by computer, fixed block size of four without stratification.
Allocation: AstraZeneca prepared individually numbered sets of study drugs, packed them according to the randomisation sequence and then shipped them to the study team in numbered but apparently identical sets.
Blinding: identical capsules, "investigators, patients and all research staff were blind to the study drugs and the block size".
Duration: 1 year.
Design: parallel.
Location: single‐center (all in Early Assessment Service for Young People with Psychosis (EASY) in Hong Kong).
Setting: outpatient.

Participants

Diagnosis: schizophrenia and related  psychoses (DSM‐IV), all first episode, all well remitted, all had remained well on maintenance medication for 1 year.

N=178.
Gender: 24.2
Age: 80 men, 98 women.
History: duration stable‐ 1 year, duration ill‐ 2.3 years, number of previous hospitalisations‐ 0 (first episode), age at onset‐ 21.9 years, severity of illness‐ mean PANSS 36, baseline antipsychotic dose‐ 153mg/day chlorpromazine equivalents.

Interventions

1. Drug: quetiapine. Fixed dose of 400mg/day. N=89.

2. Placebo: Duration of taper (days): 35. N=89.

Rescue medication: antipsychotics not allowed.

Outcomes

Examined:

Relapse: (i)  an increase in at least one of the following Positive and Negative Syndrome Scale psychotic symptom items to a threshold  score (delusion, hallucinatory behaviour, conceptual disorganisation, unusual thought content, suspiciousness; (ii) Clinical Global  Impression Severity of Illness 3 or above and (iii) CGI  change 5 or above).

Leaving the study early.

Rehospitalisation.

Suicide attempts.

Adverse effects: akathisia, tardive dyskinesia, tremor, sedation, weight gain.

Open employment status.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sequence by computer, fixed block size of four without stratification.

Allocation concealment (selection bias)

Low risk

AstraZeneca prepared individually numbered sets of study drugs, packed them according to the randomisation sequence and then shipped them to the study team in numbered but apparently identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Identical capsules, "investigators, patients and all research staff were blind to the study drugs and the block size".

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Identical capsules, "investigators, patients and all research staff were blind to the study drugs and the block size".

Incomplete outcome data (attrition bias)
All outcomes

High risk

72% of the participants left the study early. As most participants dropped out after relapse this outcome was not affected, but it is a source of bias for other outcomes. Survival analysis for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Cheung 1981

Methods

Randomisation: randomly in group of 15 each, no further details.
Allocation: procedure not indicated.
Blinding: double, no further details.
Duration: 18 months.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (mainly Schneiderian first‐rank symptoms), last relapse 30‐60 months ago, fully remitted since and maintained on antipsychotic drugs.

N=30.
Gender: 12 men, 18 women.
Age: 39.9 years.
History: duration stable‐ mean 44 months, duration ill‐ mean 10.2 years, number of previous hospitalisations‐ mean 1.6, age at onset‐ mean 29.7 years, severity of illness‐ n.i., baseline antipsychotic dose‐ 151 mg/day chlorpromazine equivalents.

Interventions

1. Drug: switched to various antipsychotic drugs with similar profile as the previous one. Fixed/flexible dose: probably flexible. Allowed dose range: n.i.. Mean dose: n.i.. N=15.

2. Placebo: benzodiazepine (‘active placebo’). Duration of taper 0 days. N=15.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: recurrence of symptoms definitely of schizophrenic type, or symptoms not diagnostic of schizophrenia (e.g. sleep problems) which could not be controlled with other measures than antipsychotic drugs or ECT.

Leaving the study early.

Quality of life: subjective distress (Symptom Questionnaire of Kellner and Sheffield).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly in group of 15 each, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

12 participants left the study early (40%), among those 10 from the placebo group and 8 for relapse. Outcomes other than relapse and leaving early are clearly prone to bias due to this difference in leaving the study early.

Selective reporting (reporting bias)

Low risk

Use of benzodiazepines was not indicated, but this was not an outcome of interest in our review.

Other bias

Low risk

No evidence for other bias.

Clark 1975

Methods

Randomisation: random, in blocks of eight, stratified for age, duration ill and time since last admission.
Allocation: procedure not described.
Blinding: double, identical capsules, each participant had an individual stock bottle.
Duration: 24 weeks.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), 22 undifferentiated, 7 paranoid, 1 schizoaffective, no severe other psychiatric or somatic illnesses, no severely ill participants.

N=40.
Gender: 40 women.
Age: mean 42.8 years (range 24‐60).
History: duration stable‐ maintained on medication in an outpatient status for at least 3 months, "relatively stable state of health”, duration ill‐ mean 11.6 years, number of previous hospitalisations‐ mean 6.1, age at onset ‐ NI , severity of illness‐ mean CGI severity score 2.94, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: pimozide.* Flexible dose. Allowed dose range: 2‐20mg/day. Mean dose: 5.3 mg/day. N=15.

2. Drug: thioridazine.* Flexible dose. Allowed dose range: 75‐750mg/day. Mean dose: 189mg/day. N=15.

3. Placebo: Duration of taper: 0 days. N=10.

Rescue medication: antiparkinson medication, bedside sedation.

Outcomes

Examined:

Relapse (worsening of global state).

Leaving early.

Global state: number of participants improved according to Clinical Global Impressions Scale.

Adverse effects: binary outcomes ‐ open interview.

Unable to use / Not included:

Mental state: BPRS (no SD / no prespecified outcome of interest).

Functioning: Katz Lyerly Scale of Social Adjustment, Patient Rating Form, Family Rating Form (all no SD / no prespecified outcomes of interest).

Physiological measures: biological parameters (temperature, mean weight, pulse, blood pressure, all no data / all no prespecified outcomes of interest), laboratory (blood count, urine analysis, liver enzymes, blood sugar, protein bound iodine, all no prespecified outcomes of interest).

Notes

* The results of pimozide and thioridazine were combined in the analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random, in blocks of eight, stratified for age, duration ill and time since last admission.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules, each participant had an individual stock bottle.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules, each participant had an individual stock bottle.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall 36% left the study early. The specific reasons why the participants dropped out were not indicated by group.

Selective reporting (reporting bias)

Low risk

No clear source for selective reporting.

Other bias

Low risk

No clear other sources of bias.

Cooper 2000

Methods

Randomisation: computer‐generated randomisation list.
Allocation: allocation to treatment was on a double‐blind basis, codes were not broken until the time of analysis.
Blinding: double‐blind, no further details.
Duration: 26 weeks.
Design: parallel.
Location: multi‐centre, multi‐national.
Setting: inpatient (n=33) and outpatient (n=86), sponsored.

Participants

Diagnosis: chronic schizophrenia (DSM‐III‐R), at least mildly ill according to CGI, had a history of recurrence in last 18 months, currently maintained on antipsychotic medication.

N=121.
Gender: 82 men, 37 women (intent‐to‐treat dataset).
Age: 42.3 years.
History: duration stable‐ n.i., duration ill‐ mean 13.6 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 28.7 years, severity of illness‐ mean BPRS 49.1, mean CGI 4.2, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: zotepine. Fixed dose of 300mg/day which could be reduced once to 150mg/day. Mean dose: n.i.. N =63.

2. Placebo: Duration of taper: 0 days. N =58.

Rescue medication: antipsychotic drugs not allowed, but benzodiazepines.

Outcomes

Examined:

Relapse: (i) a moderate clinical deterioration from baseline (an increase in CGI severity score of at least 2 points plus an increase of 2 points in at least two positive symptom items on the BPRS persisting for two assessments over 3 days, but not requiring hospitalisation; (ii) deterioration requiring hospitalisation accompanied, on one assessment, by an increase in CGI severity score of at least 2 points plus an increase of 2 points in at least two positive symptom items on the BPRS; and (iii) severe clinical deterioration (an increase in CGI severity score to ‘severely ill’ for 24 hours, or, if in hospital, requiring special observation for suicidal or aggressive behaviour).

Global state: number of participants improved according to CGI.

Adverse effects: binary outcomes ‐ open interview.

Unable to use / Not included:

Mental state: BPRS, SANS (no prespecified outcomes of interest).

Adverse effects: extrapyramidal side‐effects (SAS, AIMS, no SD / continous side‐effect results were not among the prespecified outcomes).

Physiological measures: laboratory, vital signs, ECG (all no data / no prespecified outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation list.

Allocation concealment (selection bias)

Low risk

Allocation to treatment was on a double‐blind basis, codes were not broken until the time of analysis.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall rate of participants leaving the study early was very high (76%) and many more participants in the placebo group than in the drug group dropped out due to relapse. Kaplan‐Meier survival analysis was used for primary outcome relapse. No full ITT analysis, only those participants with at least one post‐baseline assessment were included, but only two participants were excluded on this basis.

Selective reporting (reporting bias)

High risk

Only those adverse events that were reported on at least four occasions and serious adverse events were reported.

Other bias

Low risk

No clear other bias.

Crow 1986

Methods

Randomisation: random, no further details.
Allocation: allocation lists prepared by pharmacy for five antipsychotic drugs mentioned below, concealment is unclear.
Blinding: double, no further details.
Duration: 104 weeks.
Design: parallel.
Location: multi‐centre.
Setting: outpatient.

Participants

Diagnosis: first episode of schizophrenia (Present State Examination).

N=120.
Gender: 74 men, 46 women.
Age: mean 26.3 years (range 16‐59 years).
History: duration stable‐ 30 days after discharge all on active medication, duration ill‐ 2.8 months (between illness onset and admission to hospital), number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ most participants were ‘well’ at the beginning of the study (91 well, 13 psychotic features, 10 defect state, 6 unspecific symptoms), baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: flupenthixol i.m., chlorpromazine, haloperidol, pimozide, trifluoperazine Flexible dose. Allowed dose range: no upper limit, but lower limit was flupenthixol i.m. 40mg/month, chlorpromazine 200mg/day, haloperidol 3mg/day, pimozide 4mg/day, trifluoperazine 5mg/day. Mean dose: flupenthixol 84mg/month (n=31), chlorpromazine 366mg/day (n=3), haloperidol 11.8mg/day (n=3), pimozide 7.8mg/day (n=5), trifluoperazine 11.5mg/day (n=12). N=54.

2. Placebo: Duration of taper (days): 30 days on drug, then received half dose for 30 days before they were put on placebo. N=66.

Rescue medication: antiparkinson medication, antidepressants, anxiolyties.

Outcomes

Examined:

Relapse: rehospitalisation or rehospitalisation thought necessary although not possible or need of medication.

Unable to use / Not included:

Hallucinations, delusions (no data / no predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Allocation lists prepared by pharmacy for five antipsychotic drugs mentioned below, concealment is unclear.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No clear bias. overall rate of leaving early of 11% is acceptable. Survival curve analysis was used for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Blind was broken when a participant relapsed.

Denijs 1973

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 26 weeks.
Design: parallel.
Location: single‐centre.
Setting: in hospital.

Participants

Diagnosis: residual schizophrenia (DSM‐II), chronic, currently treated with antipsychotic drugs.

N=40.
Gender: 40 women.
Age: mean 58.5 years.
History: duration stable‐ all participants were switched to two months treatment with pimozide and only those who were treated effectively (=markedly improved) were randomised, duration ill‐ mean 30.5 years, duration of current hospitalisation mean 24.5 years (range 1‐43), number of previous hospitalisations‐ n.i., age at onset‐ mean 28 years, severity of illness‐ n.i., baseline antipsychotic dose‐ pimozide mean 7.72mg/day.

Interventions

1. Drug: pimozide. Flexible dose. Allowed dose range: n.i.. Mean dose: n.i.. N=20.

2. Placebo: Duration of taper: 0 days. N=20.

Rescue medication: not allowed, only dose increase of pimozide or placebo‐pimozide was possible. Additional use of haloperidol meant relapse.

Outcomes

Examined:

Relapse: need of additional haloperido)

Adverse effects: number of participants with at least one movement disorder, rigor and tremor.

Unable to use / Not included:

Mental state: Overall Factor Construct Scale (no mean, no SD / no prespecified outcome of interest)

Behaviour: ‘Psychiatric Evaluation Scale’ (no mean, no SD / no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 (5%) of the participants left the study early which is an acceptable rate. Both participants were included in the endpoint analysis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Doddi 1979

Methods

Randomisation: no details (just reported as a "randomised study”).
Allocation: procedure not described.
Blinding: "double‐blind” ("patients and authors were not aware of the allocated treatment”).
Duration: 9 months.
Design: randomised, parallel (enriched design: patients, who responded to fluphenazine long‐acting treatment (25 or 50 mg/month) for at least six to 12 months before study entry, were randomised to continue that treatment or to placebo). Ten out of 20 patients had been previously recruited in a study comparing fluphenazine with trifluorazine.
Location: no clear details.
Setting: outpatients.

Participants

Diagnosis: chronic schizophrenia with an acute episode within 6 to 12 months before study entry (no details about diagnostic criteria).

N=20.
Gender: all men.
Age: 19 to 32 years.
History: duration stable at least six months, duration ill‐ some were first episode patients, some were patients with recurrence, number of previous hospitalisations‐ no data, age at onset‐ no data, severity of illness‐ fluphenazine group had a mean BPRS baseline score of 24.56 (SD 3.56); placebo group had a mean BPRS baseline score of 21.71, baseline antipsychotic dose (25 or 50 mg/month).

Interventions

1. Drug: fluphenazine depot. Fixed dose: 25 or 50 mg/month (long‐acting formulation). Mean dose: n.i.. N=10 randomised (but data available only for 9 patients who completed the study).

2. Placebo: Duration of taper (days): n.i.. N=10 randomised (but data available only for 7 patients who completed the study).

Rescue medication: antiparkinson medication at study entry (and then progressively tapered off, without a prespecified schedule).

Outcomes

Examined:

Relapse: defined as worsening of clinical status needing an adjunctive new antipsychotic treatment.

Unable to use / Not included:

Mental state: BPRS (no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No details (just reported as a "randomised study”).

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind ("patients and authors were not aware of the allocated treatment”).

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind ("patients and authors were not aware of the allocated treatment”).

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

25% of the participants dropped out, all due to relapse. This may still be acceptable.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Eklund 1991

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, placebo injections, no further details.
Duration: 48 weeks.
Design: parallel.
Location: single‐centre.
Setting: in‐ and outpatients.

Participants

Diagnosis: schizophrenia (Research Diagnostic Criteria), requiring neuroleptic maintenance treatment to prevent relapse.

N=43.
Gender: n.i..
Age: mean 51.7 (range 25‐65) years.
History: duration stable‐ remained in the study after 15 weeks of haloperidol decanoate, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ 60mg haloperidol decanoate per month (˜3.5mg/day haloperidol).

Interventions

1. Drug: haloperidol decanoate 60mg/4 weeks. Fixed dose. N=20.

2. Placebo: Duration of taper: 0 days, but all on depot medication before study. N=23.

Rescue medication: anticholinergics and sedation.

Outcomes

Examined:

Relapse: clinical judgement.

Leaving the study early.

Unable to use / Not included:

Mental state: Comprehensive Psychopathological Rating Scale (no mean, no SD / no prespecified outcome of interest).

Adverse effects: extrapyramidal side‐effects, tardive dyskinesia (no mean, no SD / continuous side‐effect results were not among the prespecified outcomes).

Physiological measures: laboratory (prolactin and haloperidol levels, no mean/SD / no prespecified outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo injections, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo injections, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

A considerable number of participants (42%) left the study early. The number was clearly higher in the placebo group and the reasons differed. Data were analysed on an intent‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

No selective reporting.

Other bias

Low risk

No clear other bias.

Elie 1975

Methods

Randomisation: random number table.
Allocation: all personnel except for the treating psychiatrist remained unaware of the code until the end of the study.
Blinding: double (patients, scientists, nurses, only the treating psychiatrist knew the treatment).
Duration: 12 days.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis).

N=14.
Gender: 14 women.
Age: n.i..
History: duration stable‐ n.i., duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: chlorpromazine ‐ Fixed dose. Allowed dose range n.i.. Mean dose n.i.. N=7.

2. Placebo: Duration of taper: 0 days. N=7.

Rescue medication: benztropine.

Outcomes

Examined:

Relapse: worsening of psychotic symptoms.

Leaving the study early.

Unable to use / Not included:

Behaviour: NOSIE (no data / no prespecified outcome of interest).

Neurophysiological tests (no SDs / no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table.

Allocation concealment (selection bias)

Low risk

All personnel except for the treating psychiatrist remained unaware of the code until the end of the study.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double (patients, scientists, nurses, only the treating psychiatrist knew the treatment).

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double (patients, scientists, nurses, only the treating psychiatrist knew the treatment).

Incomplete outcome data (attrition bias)
All outcomes

Low risk

One participant in the placebo group left the study prematurely which is an acceptable rate.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Freeman 1962

Methods

Randomisation: participants were ranked for morbidity, then matched, then randomised.
Allocation: procedure not described.
Blinding: double, identical capsules, each participant was provided medication in individual container. Staff guessed on which medication the participants were but could not guess adequately.
Duration: 26 weeks.
Design: parallel.
Location: single‐centre.
Setting: in hospital.

Participants

Diagnosis: chronic, long term hospitalised male psychotics (clinical diagnosis), 86 schizophrenia, 6 chronic brain syndrome, 2 personality disorders, 2 n.i..

N=96.
Gender: 96 men.
Age: 43.6 years.
History: duration stable‐ treated with chlorpromazine for at least 2 months, not ready for discharge, not assaultive, duration ill‐ n.i. but duration of current hospitalisation 12.3 years, number of previous hospitalisations NI‐ , age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ 224 mg chlorpromazine per day.

Interventions

1. Drug: chlorpromazine ‐ Flexible dose. Allowed dose range: n.i.. Mean dose: n.i.. N=48.

2. Placebo: Duration of taper: 0 days. N=48.

Rescue medication: occasional use of sedatives, antipsychotics were not allowed.

Outcomes

Examined:

Relapse: condition worsened to such a point that ordinarily a complete change in treatment would be considered.

Leaving early due to inefficacy.

Unable to use / Not included:

Behaviour: Lyon’s Behaviour Scale (no SD / no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were ranked for morbidity, then matched, then randomised.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double, identical capsules, each participant was provided medication in individual container. Staff guessed on which medication the participants were but could not guess adequately.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules, each participant was provided medication in individual container. Staff guessed on which medication the participants were but could not guess adequately.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It can be that there were participants leaving the study early but this was not clearly reported.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Blind was broken once a participant relapsed.

Gallant 1974

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 12 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: severely ill, chronically hospitalized people with schizophrenia (clinical diagnosis).

N=50.
Gender: 25 men, 25 women.
Age: medium 41.5 years.
History: duration stable‐ 12 weeks stabilisation phase., but how long the participants were stable is unclear, duration ill‐ n.i., number of previous hospitalisations‐ n.i., but median duration of current hospitalisation 15.5 years, age at onset‐ n.i., severity of illness‐ all severely ill (Clinical Global Impression Score=6), baseline antipsychotic dose‐ 100‐160 mg/week penfluridol.

Interventions

1. Drug: penfluridol once weekly. Fixed dose. Allowed dose range: 40‐160 mg/week. Mean dose: n.i.. N=25.

2. Placebo: Duration of taper: 0 days. N=25.

Rescue medication:  antiparkinson medication.

Outcomes

Examined:

Relapse: worsening of global state.

Leaving the study early.

Global state: number of participants according to the Clinical Global Impression Scale.

Adverse effects: extrapyramidal side‐effects.

Unable to use / Not included:

Mental state: Brief Psychiatric Rating Scale (no mean, no SD / no prespecified outcome of interest).

Behaviour: Nurses´ Observation Scale for Inpatient Evaluation (no mean, no SD / no prespecified outcome of interest).

Physiological measures: laboratory, ECG, photosensitivity tests, ophthalmologic examinations, vital signs (no clear data / no prespecified outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

It is not entirely clear, whether there were dropouts in addition to 18 participants (7 drug, 11 placebo, 36%) who left the study early due to relapse. However, the 36% drop out rate can be a problem for other outcomes.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Gardos 1984

Methods

Randomisation: randomised, 3:1 ratio.
Allocation: procedure not described.
Blinding: double, ‘matching placebos’ and sesame oil for fluphenazine decanoate treated participants.
Duration: 10 weeks.
Design: parallel.
Location: two centres.
Setting: outpatient.

Participants

Diagnosis: chronic psychotic outpatients (DSM‐III), schizophrenia (n=26), mental retardation with psychosis (n=9), organic brain syndrome (n=1).

N=36.
Gender: 17 men, 19 women.
Age: mean 45.8 years.
History: duration stable‐ n.i., but all receiving maintenance neuroleptic therapy, all for at least 5 years, duration ill‐ n.i., but mean duration of neuroleptic treatment 13.4 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ mean 365 mg/day chlorpromazine equivalents.

Interventions

1. Drug: various antipsychotic drugs. Fixed dose: keeping the dose of the antipsychotic the participant was on at the beginning of the study. Mean dose: 365mg/day chlorpromazine equivalents. N=9.

2. Placebo: Duration of taper: 28 days. N=27.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: major clinical deterioration.

Leaving the study early.

Unable to use / Not included:

Global state: Clinical Global Impression (no data for each group separately/no prespecified outcome of interest).

Mental state: Brief Psychiatric Rating Scale, Profile of Mood Symptoms (no data for each group separately/no prespecified outcome of interest).

Adverse effects: extrapyramidal side‐effects (Abnormal Involuntary Movement Scale, Dyskinesia Rating Scale, no data for each group separately / continuous side‐effect results were not among the prespecified outcomes), other adverse effects (Treatment Emergent Symptoms Scale, no data for each group separately / continuous side‐effect results were not among the prespecified outcomes).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, 3:1 ratio (information obtained from author).

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, ‘matching placebos’ and sesame oil for fluphenazine decanoate treated participants.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, ‘matching placebos’ and sesame oil for fluphenazine decanoate treated participants.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

The differential dropout rate (placebo group 8/27, 0/9 maintenance group, all due to relapse) can have biased other outcomes than relapse and leaving the study early. But data on such other outcomes were not available anyways.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Garfield 1966

Methods

Randomisation: matched in three groups according to age and hospitalisation, then randomised using a table of random numbers.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 22 weeks (experimental phase).
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: schizophrenia (clinical diagnosis), undifferentiated type (n=10), hebephrenic (n=6), catatonic (5), paranoid (5), acute undifferentiated (n=1).

N=27.
Gender: 27 women.
Age: mean 42.4 years.
History: duration stable‐ on continuous phenothiazine medication at sufficient dose for at least 6 months, then stabilised another 2 months on the ward, total 8 months, duration ill NI‐ duration of current hospitalisation mean 11.42 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ chlorpromazine mean 610mg/day (n=17), thioridazine mean 480mg/day (n=5), trifluoperazine mean 25mg/day (n=3), perphenazine 24mg/day (n=1), prochlorperazine 60mg/day (n=1).

Interventions

1. Drug: remained on previous antipsychotic medication (chlorpromazine, thioridazine, trifluoperazine, perphenazine, prochlorperazine). Fixed/flexible dose: not clear, but probably fixed. Allowed dose range: n.i.. Mean dose: n.i., because it is unclear which patients were allocated to which group. N=9.

2. Placebo: Duration of taper: 7 days. N=9**.

Rescue medication: tranquilizer (=benzodiazepine).

Outcomes

Examined:

Relapse: worsening by three points on the factor scores of the IMPS or withdrawn due to being worse.

Leaving the study early.

Global state: improvement by three points on the factor scores of the IMPS or withdrawn due to being ready for discharge.

Unable to use / Not included:

Mental state: Inpatient Multidimensional Psychiatric Scale (no data / no prespecified outcome of interest).

Behaviour: Psychotic Reaction Profile (no data / no prespecified outcome of interest).

Notes

** a second placebo group that was referred to a specialised ward was not used in our calculations (n=9).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Matched in three groups according to age and hospitalisation, then randomised using a table of random numbers.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

There was a considerable number of participants leaving the study early (28%). The approach how missing data were handled is not specified.

Selective reporting (reporting bias)

Low risk

Only two factors of the IMPS were presented, but this was no outcome of interest.

Other bias

Low risk

No clear other bias.

Gitlin 1988

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 24 weeks (but we used only the first 12 weeks of this cross‐over study).
Design: cross‐over.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: schizophrenia (n=10), schizoaffective disorder (n=2) according to Research Diagnostic Criteria, first episode no more than 2 years ago.

N=12.
Gender: 8 men, 4 women.
Age: mean 25 years.
History: duration stable‐ all stabilised on 12.5mg/two weeks fluphenazine depot for one year, duration ill‐ < 3years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i.,  baseline antipsychotic dose‐ 12.5 mg/two weeks fluphenazine i.m..

Interventions

1. Drug: fluphenazine i.m. Fixed dose 12.5mg/two weeks fluphenazine i.m.. Mean dose: 12.5mg/two weeks fluphenazine i.m.. N=n.i..

2. Placebo: Duration of taper 0 days, but depot study. N=n.i..

Rescue medication: n.i..

Outcomes

Unable to use / Not included:

Relapse: no data for first cross‐over phase.

Prolactin levels (no data for first cross‐over phase / no prespecified outcome of interest).

Notes

Depot study, at six weeks the full plasma level could still be measured, even at the end of 12 weeks 33% still had substantial fluphenazine plasma levels. 

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear, because not indicated.

Selective reporting (reporting bias)

High risk

Data not presented for both groups separately.

Other bias

Unclear risk

Unclear ‐ baseline imbalance can not be addressed.

Goldberg 1981

Methods

Randomisation: randomly assigned.
Allocation: procedure not described.
Blinding: double, placebo injection.
Duration: 6 weeks.
Design: parallel.
Location:  single‐center.
Setting: outpatient.

Participants

Diagnosis: chronic schizophrenic outpatients (DSM‐III).

N=31.
Gender: n.i..
Age: 37 years.
History: duration stable‐ 2 years on fluphenazine decanoate 3 weekly, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ 24 years, severity of illness‐ mean GAS (Global Assessment Scale Endicott 1976by Spitzer & Endicott 1976), baseline antipsychotic dose‐ 39.3mg/ 3 weekly fluphenazine decanoate.

Interventions

1. Drug: fluphenazine decanoate‐ Fixed doses. Allowed dose range: n.i. ‐ same dose as before. Mean dose: n.i.. N=14.
2. Placebo: Duration of taper: 0 days, but all on depot. N=17.
Rescue medication: n.i..

Outcomes

Examined:

Relapse: clinical judgement.

Leaving the study early.

Adverse effects: tardive dyskinesia (AIMS).

Unable to use / Not included:

Social Adjustment Scale.

Depression: SADS (no mean, no SD / no prespecified outcome of interest).

Functioning: GAS (no mean, no SD / no prespecified outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further deatils.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo injection.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo injection.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

3 out of 30 participants (10%) left the study early which is an acceptable rate, irrespective of the statistical analysis (completer analysis).

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Gross 1960

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, unidentifiable capsules.
Duration: 6 months.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: chronic psychotic patients (mainly schizophrenia, clinical diagnosis).

N=144.
Gender: n.i..
Age: n.i..
History: duration stable‐ "observed on the same drugs for 4.5 months”, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: continuation of antipsychotic taken before the study ‐ Fixed/flexible dose: unclear. Allowed dose range: unclear. Mean dose: n.i.. N=46.

2. Placebo: Duration of taper: "4 weeks to five months, usually 2 months”. N=98.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: clinical diagnosis.

Unable to use / Not included:

Social adjustment: (not reported for the randomised participants / no predefined outcome of interest).

Rehospitalisation (unclear numbers).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, unidentifiable capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, unidentifiable capsules.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Whether participants left the study early is unclear.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

In case of relapse the blind was broken.

Gross 1974

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 16 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis) with positive or negative symptoms, responsive to treatment with antipsychotic drugs, all so ill that they required continuous treatment with antipsychotic medication for at least 3 months.

N=61.
Gender: 37 men, 24 women.
Age: mean 45.7 years.
History: duration stable‐ all participants had received a neuroleptic for at least 4 weeks, then stabilized on a fixed dose for 2 weeks, the last 2 weeks of which they were stabilized on a fixed dose, duration ill‐ at least 2 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i. , severity of illness‐ n.i., baseline antipsychotic dose‐ chlorpromazine maximum dose 500mg/day, thioridazine 500mg/day, fluphenazine 30mg/day, trifluoperazine 30mg/day, other equipotent antipsychotics or combinations not exceeding the maximum doses.

Interventions

1. Drug: pimozide ‐ Flexible dose. Allowed dose range: 2‐12 mg/day. Mean dose: 6.3 mg/day. N=21.

2. Drug: trifluoperazine. Flexible dose. Allowed dose range: 5‐30 mg/day. Mean dose: 17.5 mg/day. N=20.

3. Placebo: Duration of taper: 21 days. N=20.

Rescue medication: chloralhydrate, antiparkinson medication.

Outcomes

Examined:

Relapse: at least minimally worse on CGI.

Leaving the study early.

Unable to use / Not included:

Mental state: BPRS (no predefined outcome of interest).

Global state: CGI (no predefined outcome of interest).

Social activity: Family Rating Form (no SD / no predefined outcome of interest).

Social adjustment: Harbor View House Residents Rating Report (no SD / no predefined outcome of interest).

Adverse effects: open interview (no data).

Physiological measures: vital signs, laboratory (both no data / no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall number of participants leaving the study early (41%) was considerable, with a higher drop‐out rate in the placebo group.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Hershon 1972

Methods

Randomisation: randomised, no further details.
Allocation: capsules dispensed by the hospital pharmacist who was the only person who knew what the capsules were and to whom they were given.
Blinding: double, placebo capsules, no further details.
Duration: range 13‐22 weeks, mean 16 weeks.
Design: parallel.
Location: 2 centres.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), >70% of them with extrapyramidal side effects after long treatment with phenothiazines.

N=63.
Gender: 32 men, 31 women.
Age: mean 57 years.
History: duration stable‐ n.i., duration ill‐ n.i., but currently hospitalised for at least 4 years and treated with phenothiazines for a mean duration of 9.4 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ mean 17mg/day trifluoperazine (86% of the participants).

Interventions

1. Drug: trifluoperazine ‐ Fixed dose (maintaining the initial dose, necessity of dose increase was considered to be a relapse). Mean dose: 17 mg/day. N=31.

2. Placebo: Duration of taper: 0 days. N=32.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: deterioriation of participant’s condition to such a degree that additional antipsychotic medication was necessary.

Unable to use / Not included:

Adverse effects: movement disorders (no randomised data).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Low risk

Capsules dispensed by the hospital pharmacist who was the only person who knew what the capsules were and to whom they were given.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo capsules, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo capsules, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

16% left the study early, all but one due to relapse. This appears acceptable. relapse and death were the only outcomes.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Participants with a relapse were probably removed from the study and the blind broken. Study was probably terminated early.

Hirsch 1973

Methods

Randomisation: randomly allocated by research assistant.
Allocation: a part from the research assistant no one knew who was on drug or placebo until the data were analysed.
Blinding: double, sesame oil injections, unmarked ampoules. Blinding was tested at the end of the trial and it worked.
Duration: 9 months.
Design: parallel.
Location: two centres.
Setting: outpatient.

Participants

Diagnosis: chronic schizophrenia (Present State Examination), chronicity defined by at least 2 admissions or 1 admission lasting longer than 6 months, 71 schizophrenic psychosis with delusions or auditory hallucinations, six non affective delusional psychoses, three catatonic schizophrenia.

N=81.
Gender: 52 men, 29 women.
Age: mean 43.4 years.
History: duration stable‐ at least 8 weeks, duration ill‐ n.i., number of previous hospitalisations‐ 24 had ≤3 and 57 had ≥4), age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ 86% fluphenazine depot 25mg/month, no additional antipsychotic medication.

Interventions

1. Drug ‐ Fixed/flexible dose: Allowed dose range: 25mg/month ‐ no upper limit. Mean dose: 26.4mg/month. N=41.

2. Placebo: Duration of taper: n.i.. N=40.

Rescue medication: antidepressants, antiparkinson medication

Outcomes

Examined:

Relapse: deterioration of condition to a degree that participant had to be taken out of the trial to ensure that active medication was prescribed, prescription of oral phenothiazines.

Adverse effects: use of antiparkinson medication.

Unable to use / Not included:

Mental state: Present State Examination (no data / no predefined outcome of interest).

Social functioning: Social Performance Schedule, Events Schedule of Bron and Birley (both no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly allocated by research assistant.

Allocation concealment (selection bias)

Low risk

Apart from the research assistant no one knew who was on drug or placebo until the data were analysed.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double, sesame oil injections, unmarked ampoules. Blinding was tested at the end of the trial and it worked.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, sesame oil injections, unmarked ampoules. Blinding was tested at the end of the trial and it worked.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 43% of the participants left the study early (no complete ITT for some outcomes).

Selective reporting (reporting bias)

Low risk

No evidence for selected reporting.

Other bias

Low risk

No evidence of other bias

Hirsch 1996

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: one year.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (DSM‐III‐R).

N=21.
Gender: data on randomised subsample are not available.
Age: data on randomised subsample are not available.
History: duration stable‐ for at least 6 months and all on maintenance treatment, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: fluphenazine depot. Fixed dose of 25mg/2 weeks. N=11.

2. Placebo: Duration of taper: 0 days. N=10.

Rescue medication:
Haloperidol was given to participants who developed prodromal symptoms and was continued for 2 weeks unless relapse occurred.

Outcomes

Unable to use / Not included:

No data could be used because they have not been presented for the randomised subset.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Uncear, whether there were any drop‐outs.

Selective reporting (reporting bias)

High risk

No useable data because data of the randomised subsample have not been presented.

Other bias

Low risk

No clear evidence for other bias.

Hogarty 1973

Methods

Randomisation: randomly assigned, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules, no further details.
Duration: 2‐3 years (data available up to 2 years).
Design: parallel.
Location: three centres.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (DSM‐II, undifferentiated type 46.3%, paranoid 39%, acute differentiated 8%, schizoid affective 2.7%, other 3.8%), currently hospitalised for less than 2 years.

N=374.
Gender: 159 men, 215 women.
Age: mean 34.4 years.
History: duration stable‐ 2 months transition phase, those who relapsed during this time were replaced, duration ill‐ n.i., number of previous hospitalisations‐ mean 2.6, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ mean 265mg chlorpromazine per day.

Interventions

Previous medication was gradually shifted to chlorpromazine for two months.
1. Drug: chlorpromazine ‐ Flexible dose. Allowed dose range: 100mg/day. Mean dose: ˜ 260mg/day. N=192.

2. Placebo: Duration of taper: 0 days. N=182.

Rescue medication: not indicated, but probably not allowed.

Outcomes

Examined:

Relapse: clinical deterioration of such magnitude that hospitalisation appeared imminent.

Unable to use / Not included:

Leaving the study early (numbers not specified for each group separately).

Mental state: Brief Psychiatric Rating Scale, Inpatient Multidimensional Psychiatric Scale, Springfield Symptom Index, Hopkin’s Symptom Distress Check List (all no SDs and data only given for subgroups / no predefined outcome of interest).

Social behaviour and adjustment: Katz Adjustment Scale, Major Role Adjustment Inventory (both no SDs and data presented only for subgroups / no predefined outcome of interest).

Notes

Half of the participants randomly received major role therapy in addition to chlorpromazine or placebo. For the purpose of this review the four resulting groups were pooled as described above.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Relatively few participants left the study early due to reasons other than relapse which was the only outcome (n=31). Although it is unclear in which group they occurred the small percentage does not represent an important risk of bias.

Selective reporting (reporting bias)

Low risk

No clear evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Hough 2010

Methods

Randomisation: patients were randomised in a 1 to 1 ratio (via a sponsor prepared, computer generated randomisation scheme, assigned by an interactive voice system).
Allocation: interactive voice system.
Blinding: double, no further details.
Duration: variable (the trial was terminated early after an interim analysis).
Design: parallel.
Location: multi‐centre.
Setting: n.i..

Participants

Diagnosis: schizophrenia (DSM‐IV‐TR).

N=410.
Gender: 220 men, 88 women.
Age: mean 39 years.
History: duration stable‐ 12 weeks prospectively stable on fixed dose paliperidone, duration ill‐ mean 12 years, number of previous hospitalisations‐ median 2.6, age at onset‐ mean 27.3 years, severity of illness‐ PANSS total mean 53 points, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: paliperidone palmitate depot ‐ Fixed dose: originally 25, 50 or 100mg/4 weeks; this dose was maintained. Mean dose: n.i.. N=206.

2. Placebo: Duration of taper: 0 days. N=204.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: psychiatric rehospitalisation, deliberate self‐injury or violent behaviour, suicidal or homicidal ideation, certain predefined PANSS score.

Leaving the study early.

Rehospitalisation.

Death natural causes and suicide.

Unable to use / Not included:

Mental state: Positive and Negative Syndrome Scale (no predefined outcome of interest).

Adverse effects: open interviews (only a few adverse events were indicated and these were not of interest for the review).

Prolactin levels (no predefined outcome of interest).

Notes

The study was stopped early after a significant interim analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Patients were randomised in a 1 to 1 ratio (via a sponsor prepared, computer generated randomisation scheme, assigned by an interactive voice system).

Allocation concealment (selection bias)

Low risk

Interactive voice system.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall high drop‐out rate (45%). Clearly more participants in the placebo group (95) than in the drug group (31) left the study early due to relapse. This imbalance may have biased the results of other outcomes such as adverse events. Kaplan‐Meier survival curve analysis was used for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

Those adverse events that occurred in at least 2% of the participants and severe adverse events were presented. We feel that's acceptable.

Other bias

High risk

Study was stopped early after an interim analysis.

Kane 1979

Methods

Randomisation: matched then each pair randomised, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 6 months.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: probable or definite schizophrenia, any subtype (Research Diagnostic Criteria), in remission for at least 4 weeks or at stable clinical plateau despite vigorous chemotherapy.

N=16.
Gender: 14 men, 2 women.
Age: 26.7 years.
History: duration stable‐ mean 22.9 months in remission (minimum 6 months), duration ill‐ mean 6.1 years, number of previous hospitalisations‐ n.i., but a mean of 2.4 previous episodes, age at onset‐ mean 20.6 years, severity of illness‐ n.i., baseline antipsychotic dose‐ 3.8mg fluphenazine biweekly.

Interventions

1. Drug: fluphenazine decanoate ‐ Flexible dose. Allowed dose range: 1.25‐5.0mg biweekly. Mean dose: n.i.. N=8.

2. Placebo: Duration of taper: 0 days, but previously treated with depot medication. N=8.

Rescue medication: minor tranquilisers, additional antipsychotic drugs were not allowed.

Outcomes

Examined:

Relapse: increase in or re‐emergence of significant symptoms suggesting imminent psychotic relapse.

Unable to use / Not included:

Leaving study early (no data).

Adverse effects (no data).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Matched, then each pair randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Two participant in the drug group (1 relapse, 1 unclear) left the study early, and 7/8 participants in the placebo group dropped out due to relapse. As relapse and dropout were the only outcomes, this did not lead to bias.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Kane 1982

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, all participants received both pills and injections (active or placebo) to maintain double‐blind conditions.
Duration: 1 year.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: first episode schizophrenia (clinical diagnosis), no evidence of drug abuse or important medical illnesses. When diagnoses were reassessed by Research Diagnostic Criteria, 19 had schizophrenia, 3 had unspecific schizophrenic psychoses, 4 had other psychiatric disorders, one mania with schizotypal features and one depression with schizotypal features.

N=28.
Gender: 14 men, 14 women.
Age: mean 21.9 years.
History: duration stable‐ stable remission of at least 4 weeks, mean 16.9 weeks, duration ill‐ mean 17.6 weeks, number of previous hospitalisations‐ 0, age at onset‐ mean 21.5 years, severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: oral fluphenazine ‐ Flexible dose. Allowed dose range: 5‐20mg/day. Mean dose: n.i.. N=n.i..

2. Drug: depot fluphenazine ‐ Flexible dose. Allowed dose range: 12.5‐50/mg biweekly. Mean dose: n.i.. N=n.i..

2. Placebo: Duration of taper: 0 days. N=17.

Rescue medication: not indicated.

Outcomes

Examined:

Relapse: a substantial clinical deterioration with a potential for marked social impairment. Patients were considered dropouts only if they showed no signs of clinical deterioration at the time they left the study.

Leaving the study early.

Unable to use / Not included:

Social aspects of premorbid personality: Premorbid Asocial Adjustment Scale (data on placebo group only / no predefined outcome of interest).

Notes

The design was changed during the study in that only non‐compliant patients were randomised to depot fluphenazine or depot placebo, and the randomisation was changed to 2‐1‐1 (placebo, oral fluphenazine, depot fluphenazine).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, all participants received both pills and injections (active or placebo) to maintain double‐blind conditions.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, all participants received both pills and injections (active or placebo) to maintain double‐blind conditions.

Incomplete outcome data (attrition bias)
All outcomes

High risk

20 out of 28 participants  left the study early, 10 for other reasons than relapse, which was the only outcome apart from leaving the study early. This may present a bias.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Unclear risk

The design was changed during the study in that only non‐compliant patients were randomised to depot fluphenazine or depot placebo, and the randomisation was changed to 2‐1‐1 (placebo, oral fluphenazine, depot fluphenazine). It is unclear whether this biased the results.

Keskiner 1968

Methods

Randomisation: randomly assigned, no further details.
Allocation: procedure not described.
Blinding: double, placebo treated participants received injections of sesame oil in a similar amount.
Duration: 12 weeks.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), 12 paranoid, 3 hebephrenic, 2 catatonic, 1 simple, 6 chronic undifferentiated, on antipsychotic medication for a mean duration of 2 years.

N=24.
Gender: 4 men, 20 women.
Age: mean 36 years.
History: duration stable‐ minimum six weeks stable on oral fluphenazine, duration ill‐ mean 12.4 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 23.6 years , severity of illness‐ n.i., baseline antipsychotic dose‐ mean 28.5 mg fluphenazine decanoate biweekly.

Interventions

1. Drug: fluphenazine decanoate ‐ Flexible doses. Allowed dose range: 12.5‐75/mg biweekly. Mean dose: n.i.. N=13

2. Placebo: sesame oil injections. Duration of taper: 0 days. N=11.

Rescue medication: antiparkinson medication, additional fluphenazine decanoate ‐ but this was considered to be a relapse.

Outcomes

Examined:

Relapse: clinical deterioration requiring additional antipsychotic drug treatment.

Leaving study early.

Unable to use / Not included:

Global state: 7 point scale of severity (no data / no predefined outcome of interest).

Mental state: scale published by the authors (no SD / no predefined outcome of interest).

Adverse effects: scale published by the authors (no numbers).

Physiological measures: ECG, EEG, laboratory (all no data / no predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo treated participants received injections of sesame oil in a similar amount.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo treated participants received injections of sesame oil in a similar amount.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant left the study early.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

In case of deterioration the participants received additional antipsychotic drugs. This is a problem for the analysis of side‐effects.

Kramer 2007

Methods

Randomisation: randomised, computerized randomisation and stratification scheme.
Allocation: interactive voice‐response system.
Blinding: double, no further details.
Duration: variable.
Design: parallel.
Location:  multi‐centre.
Setting: outpatient, sponsored.

Participants

Diagnosis: schizophrenia (DSM‐IV), 80% paranoid subtype, 14% undifferentiated subtype, initially with acute exacerbation, then 8 weeks run in and 6 weeks stabilisation phase.

N=207.
Gender: 121 men, 86 women.
Age: 38.3 years.
History: duration stable‐ at least 8 weeks, duration ill‐ mean 12.1 years, number of previous hospitalisations‐ median 3, age at onset‐ 26.2 years, severity of illness‐ mean PANSS total score 52.2, mean CGI severity 2.6, baseline antipsychotic dose‐ 10.8mg/day paliperidone.

Interventions

1. Drug: paliperidone‐ Flexible doses. Allowed dose range: 3 ‐ 15mg/day Mean dose: 10.8 mg/day. N=105.

2. Placebo: Duration of taper: 0 days. N=102.

Rescue medication: benzodiazepines, antiparkinson medication, propanolol, antidepressants when the dose was stable for at least 3 months before the study.

Outcomes

Examined:

Relapse: (a) psychiatric hospitalisation (involuntary or voluntary admission); b) increase in Positive and Negative Syndrome Scale (PANSS) total score by 25% for 2 consecutive days for patients who scored more than 40 at randomisation or a 10‐point increase for patients who scored 40 or below at randomisation; c) increase in the Clinical Global Impression‐Severity (CGI‐S) score to at least 4, for patients who scored 3 or below at randomisation, or to at least 5, for patients whose CGI‐S scores were 4 at randomisation, for 2 consecutive days; d) deliberate self‐injury or aggressive behavior, or suicidal or homicidal ideation and aggressive behavior that was clinically significant; e) increase in prespecified individual PANSS item scores to at least 5, for patients whose scores were 3 or below at randomisation, or to at least 6, for patients whose scores were 4 at randomisation, for 2 consecutive days).

Quality of life: Schizophrenia Quality‐of‐Life Scale.

Unable to use / Not included:

Mental state: PANSS (no predefined outcome of interest).

Behaviour: suicide, aggression (only mean scores which were no predefined outcomes of interest).

Functioning: Personal and Social Performance Scale (no predefined outcome of interest).

Global state: CGI‐severity (only mean score which was no predefined outcome of interest).

Adverse effects: World Health Organization Adverse Reaction Terminology dictionary (no data / no predefined outcome of interest), movement disorders (Simpson Angus Scale, Barnes Akathisia Rating Scale, and Abnormal Involuntary Movement Scale (all no data / continuous side‐effect results were not among the predefined outcomes of interest).

Physiological measures: laboratory (except for metabolic problems no data), vital signs, ECG, prolactin (all no data / no predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomised, computerized randomisation and stratification scheme.

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only 28 out of 207 participants left the study prematurely for another reason than relapse. Therefore, missing outcomes may not pose a problem for the primary outcome which was assessed with the Kaplan‐Meier method. Nevertheless, high discontinuations due to relapse (75/207) which were much more frequent in the placebo group than in the drug group pose a major problem for secondary outcomes. No full ITT (participants had to receive at least one dose post‐baseline) but only two participants were excluded on this basis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Study was terminated after an interim analysis showed a clear advantage of paliperidone.

Kurland 1975

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 12 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis).

N=35.
Gender: 19 men, 16 women.
Age: mean 43.9 years.
History: duration stable‐ maintained on neuroleptic for at least 3 months, prospective 12 week stabilization phase during which participants were switched to penfluridol, duration ill‐ n.i., number of previous hospitalisations‐ mean 1.34, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ mean 64.1mg/week penfluridol.

Interventions

1. Drug: penfluridol ‐ Flexible dose. Allowed dose range: 20‐120mg/week. Mean dose:n.i.. N=18.

2. Placebo: Duration of taper: 0 days. N=17.

Rescue medication: antiparkinson medication, it seems that haloperidol was not allowed in the double‐blind phase.

Outcomes

Examined:

Relapse: psychiatric decompensation that could not be controlled by dose increase.

Leaving the study early.

Unable to use / Not included:

Global state: Clinical Global Impression Scale (no numbers / no predefined outcomes of interest).

Mental state: Brief Psychiatric Rating Scale (no numbers / no predefined outcomes of interest).

Behaviour: Nurses Observation Scale for Inpatient Behaviour (no numbers / no predefined outcomes of interest).

Physiological measures: vital signs (weight, pulse, blood pressure, respiratory frequency, temperature ‐ no numbers / no predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

12 of 35 participants left the study early (34%), 11 of them were in the placebo group. As all participants in the placebo group discontinued due to relapse, the primary outcome is not affected. But the results of all other outcomes are biased by this effect.

Selective reporting (reporting bias)

Low risk

Results on rating scales have not been reported, but these were not outcomes of interest in our review.

Other bias

Low risk

No clear other bias

Leff 1971

Methods

Randomisation: random, no further details.
Allocation: trial medication was held by the unit secretary and dispensed to Julian Leff who gave it to the treating consultant. Only the unit secretary knew which pills were active drug and which were placebo.
Blinding: double, no further details. But side‐effects were not troublesome in any patient and therefore doctors concerned probably received no clues about whether a patient was on active drug or not.
Duration: one year.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (Present State Examination), recently recovered from an acute episode, 32 florid schizophrenia, 3 delusional psychosis.

N=35.
Gender: n.i..
Age: 16‐55 years.
History: duration stable‐ n.i., but stabilised at the pre‐admission level during a 6‐12 weeks outpatient period and recently recovered from an acute episode, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: trifluoperazine or chlorpromazine (depending on the previous medication so that so far as the patient was concerned there was no apparent change in medication). Flexible dose. Allowed dose range: trifluoperazine 5‐25mg/day, chlorpromazine 100‐500mg/day. Mean dose: chlorpromazine 157.1 mg/day, trifluoperazine 12.3mg/day. N=20.

2. Placebo: Duration of taper: not indicated, probably 0 days. N=15.

Rescue medication: antiparkinson medication, antidepressants, no antipsychotics (doctors received a letter asking them not to prescribe other medication).

Outcomes

Examined:

Relapse: physician was sufficiently concerned about the patient’s status to want to be certain that he was on active drug.

Leaving the study early.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Low risk

Trial medication was held by the unit secretary and dispensed to Julian Leff who gave it to the treating consultant. Only the unit secretary knew which pills were active drug and which were placebo.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double, no further details. But side‐effects were not troublesome in any patient and therefore doctors concerned probably received no clues about whether a patient was on active drug or not.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details. But side‐effects were not troublesome in any patient and therefore doctors concerned probably received no clues about whether a patient was on active drug or not.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall drop‐out rate was 60%, almost all due to relapse which occured much more frequently in the placebo group. This poses a problem for other outcomes than relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Levine 1980

Methods

Randomisation: random 2:1, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 15 weeks.
Design: parallel.
Location: four hospitals.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (DSM‐II).

N=67.
Gender: 34 men, 33 women.
Age: mean 31.7 years.
History: duration stable‐ continuously and successfully treated for one year, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ oral fluphenazine mean 24.4mg/day, depot fluphenazine 30.9mg/3 weeks.

Interventions

1. Drug: oral fluphenazine (n=6) or depot fluphenazine (n=11). Fixed/flexible dose: unclear. Allowed dose range: unclear. Mean dose: unclear. N=17.

2. Placebo: Duration of taper: 0 days. N=50.

Rescue medication: n.i., but antipsychotics were probably not allowed.

Outcomes

Examined:

Relapse: rehospitalisation or deterioration in clinical condition which could not be managed within protocol limits (e.g., increased psychological support or adjustment of dosage).

Adverse effects: tardive dyskinesia (AIMS).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random 2:1, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear whether there were participants who left the study early.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Marjerrison 1964

Methods

Randomisation: randomly assigned.
Allocation: procedure not described.
Blinding: double ‐ (apart from previous antipsychotic group) ‐ three different colours which were again changed. Double‐blind condition maintained for patients, ward nurses and psychiatrists.
Duration: 7 months.
Design: parallel.
Location:  single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic psychotic patients, treatment resistive in closed wards. No seizures, no antidepressants, no candidates for discharge.

N=88.
Gender: 38 men, 40 women.
Age: 47 years.
History: duration stable‐ 1 year on medication, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ mean 28.1 years, severity of illness‐ mean 11.6 on modified Psychotic Reaction Profile (PRP), baseline antipsychotic dose‐ 39.3mg/ 3 weekly fluphenazine decanoate.

Interventions

1. Drug: trifluoperazine (10‐90 mg/day), chlorprothixene (50‐450 mg/day), same medication (various drugs). Flexible doses. Allowed dose range: n.i.. Mean dose: n.i.. N=54.
2. Placebo: Duration of taper: 0 days. N=34.
Rescue medication: antiparkinson, barbiturate sedation.

Outcomes

Examined:

Relapse: clinical judgement.

Unable to use / Not included:

Ward behaviour: unpublished rating scale (no predefined outcome of interest).

Urinary excretion (no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, different colours.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, different colours.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Drop‐outs 10 out of 88 is acceptable (11%), although only completers were analysed.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

McCreadie 1989

Methods

Randomisation: assumed, because study was double‐blind and because the first study phase was randomised (no further details).
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 12 months.
Design: parallel.
Location:  single‐center.
Setting: outpatient.

Participants

Diagnosis: first episode schizophrenia (Present State Examination, Feighner criteria and Research Diagnostic Criteria).

N=15.
Gender: n.i.
Age: n.i.
History: duration stable‐ 1 year, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: pimozide once weekly or i.m. flupenthixol. Flexible doses. Allowed dose range: n.i.. Mean dose: n.i.. N=8.
2. Placebo: Duration of taper: 0 days N=7.
Rescue medication: antiparkinson medication.

Outcomes

Examined:

Relapse: re‐admission.

Unable to use / Not included:

Leaving early (no data).

Cognition (no data for withdrawal study / no predefined outcome of interest).

Adverse effects: parkinsonism, tardive dyskinesia (no data for withdrawal study).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation assumed.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear whether there were missing data.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Unclear risk

Not entirely clear.

Melnyk 1966

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, "the staff, patients and investigators were not aware of which patients were to receive placebo instead of their medication”.
Duration: 6 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: schizophrenia (clinical diagnosis), paranoid schizophrenia (n=19), undifferentiated schizophrenia (n=8), catatonic schizophrenia (n=8), hebephrenic schizophrenia (n=4), acute schizophrenic reaction (n=1).

N=40.
Gender: 20 men, 20 women.
Age: n.i..
History: duration stable‐ not indicated, but mean 4.6 months on current medication, duration ill‐ mean 12.18 years, number of previous hospitalisations‐ n.i. but mean duration of current hospitalisation 18 months, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: chlorpromazine (n=6) or thioridazine (n=14). Flexible dose. Allowed dose range: 100‐600mg/day. Mean dose: n.i.. N=20.

2. Placebo: Duration of taper (days): 0 days. N=20.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: symptoms similar to those which had characterized the patient’s illness prior to successful treatment by phenothiazines.

Unable to use / Not included:

Withdrawal symptoms (no numbers for each group separately / no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, "the staff, patients and investigators were not aware of which patients were to receive placebo instead of their medication”.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, "the staff, patients and investigators were not aware of which patients were to receive placebo instead of their medication”.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It was not reported whether participants left the study early, but it is well possible that there weren’t any, because it was a relatively short inpatient study.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Morton 1968

Methods

Randomisation: random, no further details.
Allocation: the hospital pharmacist was responsible for supplying placebo and active drugs to the ward, no one concerned with the care of patients knew which patients were started on placebo.
Blinding: double, identical tablets, but in most cases nurses made correct forecasts on who was on drug and who was on placebo. Blind was broken when a participant relapsed.
Duration: 6 months.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis by two psychiatrists).

N=40.
Gender: 40 men.
Age: 25‐55 years.
History: duration stable‐ maintenance doses of tranquilisers had been administered for at least 18 months, in six participants who had to change treatment no change in symptoms was noted during 6 weeks, duration ill‐ n.i., number of previous hospitalisations‐ n.i., but duration of current hospitalisation > 2 years, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ all but six participants were on chlorpromazine or trifluoperazine, dose n.i..

Interventions

1. Drug: chlorpromazine or trifluoperazine. Fixed/flexible dose: n.i.. Allowed dose range: n.i.. Mean dose: n.i.. N=20.

2. Placebo: Duration of taper: 0 days. N=20.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: worsening of global state.

Unable to use / Not included:

Mental state: Wing Scale (no SD / no predefined outcome of interest).

Global state: clinical impression of severity (no predefined outcome of interest).

Behaviour: Wing Scale (no SD / no predefined outcome of interest).

Leaving the study early (no data).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Low risk

The hospital pharmacist was responsible for supplying placebo and active drugs to the ward, no one concerned with the care of patients knew which patients were started on placebo.

Blinding (performance bias and detection bias)
Subjective outcomes

High risk

Double, identical tablets, but in most cases nurses made correct forecasts on who was on drug and who was on placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical tablets, but in most cases nurses made correct forecasts on who was on drug and who was on placebo.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear whether there were dropouts.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Blind was broken when a participant relapsed.

Nishikawa 1982

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, drug appearance was made identical with respect to taste, colour and volume by adding a kind of "stomatics”.
Duration: three years.
Design: cross‐over
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (clinical diagnosis) in remission.

N=30.
Gender: 21 men, 9 women.
Age: mean 33.2 years.
History: duration stable‐ "in remission”, but details were not reported, duration ill‐ mean 7.3 years, number of previous hospitalisations‐ mean 2.4, age at onset‐ 25.9 years, severity of illness‐ "in remission”, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: chlorpromazine. Fixed dose of 75mg/day. N=10.

2. Drug: haloperidol. Fixed dose of 3mg/day. N=10.

3. Placebo: Duration of taper (days): 0 days. N=10.

Rescue medication: only nitrazepam for sleep and biperiden for extrapyramidal side‐effects, no additional antipsychotic drugs.

Outcomes

Examined:

Relapse: clinical judgement.

Unable to use / Not included:

Number of symptom free days (no SD’s / no predefined outcome of interest).

Notes

There were also a diazepam and an imipramine group which were not of interest for the current review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, drug appearance was made identical with respect to taste, colour and volume by adding a kind of "stomatics”.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, drug appearance was made identical with respect to taste, colour and volume by adding a kind of "stomatics”.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant left the study early due to other reasons than relapse in the first phase of the study, the only outcome apart from leaving the study early.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

The doses used were very low for Western standards. The study was initially planned as a cross‐over trial, but due to high dropout rates after the first phase only the first treatment phase was analysed. Nevertheless, this did not interfere with the aims of our review.

Nishikawa 1984

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, drug appearance was made identical with respect to powder, color, tast and volume by adding a gastric acid.
Duration: one year.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (DSM‐III), in remission or residual state.

N=87.
Gender: 53 men, 34 women.
Age: mean 41 years.
History: duration stable‐ n.i., but in remission, duration ill‐ mean 8.2 years, number of previous hospitalisations‐ mean 3.4, age at onset‐ mean 32.8 years, severity of illness‐ in remission or residual symptoms, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: haloperidol combined with biperidine and nitrazepam. Fixed dose: 1, 3 or 6 mg/day.* N=37.

2. Drug: propericiazine combined with biperidine and nitrazepam. Fixed dose: 10, 30 or 60 mg/day.* N=37.

3. Placebo combined with biperidine and nitrazepam. Duration of taper: 0 days. N=13.

Rescue medication: not indicated, probably no additional antipsychotic medication allowed.

Outcomes

Examined:

Relapse: clinical judgement.

Leaving the study early.

Unable to use / Not included:

Prolactin levels (no predefined outcome of interest).

Notes

* only the highest doses were analysed for the purpose of this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, drug appearance was made identical with respect to powder, color, tast and volume by adding a gastric acid.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, drug appearance was made identical with respect to powder, color, tast and volume by adding a gastric acid.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

While in the placebo group and in the haloperidol group the rates of participants leaving early due to other reasons were low, 9 out of 12 participants in the propericiazine group discontinued due to overdose. It is questionable whether relapse rates could be accurately measured, because most participants did not reach the endpoint.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Odejide 1982

Methods

Randomisation: participants were matched for age, sex, duration of illness, and severity of symptoms in the preceding episode and then assigned based on a randomised schedule.
Allocation: procedure not described.
Blinding: double, evaluating psychiatrist and participants were unaware of the contents of their injections. It seems that the treating psychiatrist was aware of the treatment.
Duration: 12 months.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (ICD‐9 and Present State Examination), with two or more episodes and several first rank symptoms in previous episode, free of psychopathology for at least 12 months, on fluphenazine decanoate for at least 2 years.

N=70.
Gender: n.i..
Age: n.i..
History: duration stable‐ at least 12 months free of psychopathology, duration ill‐ n.i., number of previous hospitalisations‐ n.i., but at least two previous episodes, age at onset‐ n.i., severity of illness‐ BPRS < 10 in all participants, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: fluphenazine decanoate. Fixed dose of 50mg i.m. four/eight weekly. N=35.

2. Placebo: vitamin B complex i.m.. Duration of taper: 0 days. N=35.

Rescue medication: nitrazepam for sleep and benzhexol for extrapyramidal side‐effects; additional antipsychotic drugs were not allowed.

Outcomes

Examined:

Relapse (re‐emergence of definite schizophrenic psychopathology necessitating hospital admission or other major treatment change).

Adverse effects: tardive dyskinesia (Aquired Involuntary Movements Scale).

Unable to use / Not included:

Mental state: Brief Psychiatric Rating Scale (no mean, no SD / no predefined outcome of interest).

Adverse effects: extrapyramidal symptoms ‐ use of antiparkinson medication (combined with nitrazepam), use of additional nitrazepam for sleep (combined with use of antiparkinson medication).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were matched for age, sex, duration of illness, and severity of symptoms in the preceding episode and then assigned based on a randomised schedule.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, evaluating psychiatrist and participants were unaware of the contents of their injections. It seems that treating psychiatrist was aware of the treatment.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, evaluating psychiatrist and participants were unaware of the contents of their injections. It seems that treating psychiatrist was aware of the treatment.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall drop‐out rate drug 40%, placebo 66%, most due to relapse. This poses a risk for bias for other outcomes. Completer analysis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other sources of bias.

Olson 1962

Methods

Randomisation: randomly selected and then assigned.
Allocation: procedure not described.
Blinding: identical pink capsules. Nurses, raters and patients were blind to the procedure. Treating physician was led to believe that half of the patients were on placebo, the other half on drug.
Duration: 30 days.
Design: parallel.
Location: single‐center.
Setting: inpatients.

Participants

Diagnosis: chronically mentally ill, 67%‐83% schizophrenia (clinical diagnosis), in hospital and apparently treated with antipsychotic drugs for the last 18 months.

N=60.
Gender: n.i..
Age: mean 51 years.
History: duration stable‐ at least 60 days plus 30 days prospectively, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i.

Interventions

1. Drug: chlorpromazine or thioridazine. Fixed/flexible dose: n.i.. Allowed dose range: n.i.. Mean dose: n.i.. N=30.

2. Placebo: Duration of taper: 0 days. N=30.

Rescue medication: not indicated.

Outcomes

Examined:

Relapse: attrition because of behavioural upset.

Unable to use / Not included:

Behaviour: various scales (no data reported / no predefined outcome of interest).

Notes

There were several study phases (alternation between drug and placebo). Only the first month was of interest for the review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly selected and then assigned.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Identical pink capsules. Nurses, raters and patients were blind to the procedure. Treating physician was led to believe that half of the patients were on placebo, the other half on drug.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Identical pink capsules. Nurses, raters and patients were blind to the procedure. Treating physician was led to believe that half of the patients were on placebo, the other half on drug.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

It is unclear how many participants left the study during the first month.

Selective reporting (reporting bias)

High risk

Data on behaviour scales were not reported, including aggressive behaviour which was an outcome in our review.

Other bias

Low risk

No evidence for other bias.

Ota 1973

Methods

Randomisation: unclear, randomisation assumed due to double‐blinding.
Allocation: procedure not described.
Blinding: double, all participants received both (placebo) tablets and (placebo) liquid, no further details.
Duration: 90 days.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), all had previously responded to haloperidol and were adequately maintained on it.

N=49.
Gender: 24 men, 20 women.
Age: mean 42.5 years.
History: duration stable‐ all stabilised for 30 days on haloperidol concentrate, duration ill‐ n.i., but mean duration of hospitalisation 13.7 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean BPRS 46.6 (16 items scale, rating system unclear), mean Clinical Global Impression of severity 4.9, baseline antipsychotic dose‐ mean 9.3 mg haloperidol/day.

Interventions

1. Drug: haloperidol tablets.* Flexible dose. Allowed dose range: n.i.. Mean dose: mean 8.8mg/day. N=17.

2. Drug: haloperidol liquid.* Flexible dose. Allowed dose range: n.i.. Mean dose: 10.4 mg/day. N=16.

3. Placebo: Duration of taper: 0 days. N=16.

Rescue medication: antiparkinson medication was allowed.

Outcomes

Examined:

Relapse: deterioration of global state.

Unable to use / Not included:

Mental state: Brief Psychiatric Rating Scale (no SD / no predefined outcome of interest).

Global state: Clinical Global Impression of Severity (no SD / no predefined outcome of interest).

Behaviour: Nurses Observation Scale for Inpatient Evalutation (no SD / no predefined outcome of interest).

Adverse effects: laboratory (insufficient data / no predefined outcome of interest), vital signs (insufficient data / no predefined outcome of interest).

Notes

*Groups 1 and 2 were pooled for the purpose of this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Unclear, randomisation assumed due to double‐blinding.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, all participants received both (placebo) tablets and (placebo) liquid, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, all participants received both (placebo) tablets and (placebo) liquid, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Acceptable dropout rate (10%), which should not affect other outcomes (completer analysis).

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Peuskens 2007

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 52 weeks, however terminated early after a mean duration of 120 days.
Design: parallel.
Location: multi‐centre.
Setting: probably mainly outpatients.

Participants

Diagnosis: schizophrenia (DSM‐IV), duration ill at least 2 years, Positive and Negative Syndrome Scale total score <60 before randomised phase, Clinical Global Impression Severity Scale not more than moderately ill.

N=197.
Gender: 103 men, 69 women.
Age: mean 35 years.
History: duration stable‐ at least 20 weeks, retrospectively at least one month (no change of overall severity and medication), prospectively 16 weeks stabilisation phase during which all participants were switched to quetiapine, duration ill‐ mean 8.7 years, number of previous hospitalisations‐ n.i., but mean number of episodes 4.3, age at onset‐ mean 26.5 years, severity of illness‐ mean Clinical Global Impression of severity 2.7, mean Positive and Negative Syndrome Scale total score 48.2, baseline antipsychotic dose‐ quetiapine 649 mg/day.

Interventions

1. Drug: quetiapine XR. Flexible dose 400‐800mg/day. Mean dose: 669mg/day. N=94.

2. Placebo: Duration of taper: 4 days.N=103.

Rescue medication: anticholinergic medication, sleep medication, lorazepam, no additional antipsychotic drugs.

Outcomes

Examined:

Relapse: increase of Positive and Negative Syndrome Scale by at least 30 percent from baseline, Clinical Global Impression Scale much or very much worse, need for additional antipsychotic medication.

Leaving the study early.

Adverse events: open interviews.

Global state: number of participants improved according to Clinical Global Impression Scale.

Extrapyramidal side‐effects: use of antiparkinson medication, Barnes Akathisia Scale, Simpson Angus Scale, Aquired Involuntary Movements Scale.

Unable to use / Not included:

Mental state: Positive and Negative Syndrome Scale / no predefined outcome of interest.

Laboratory: haematology, chemistry, glucose, Hba1c, insulin, lipids, urine analysis, thyroid function), ECG, vital signs, mean weight gain (all no predefined outcomes of interest).

Compliance (pill count / no predefined outcome of interest).

Notes

No participant terminated the preplanned study duration of one year. The authors reported that data after 6 months are not reliable because only a few patients were left. Therefore, relapse data after 6 months were not extracted.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Correct randomisation assumed, because recent study from industry.

Allocation concealment (selection bias)

Low risk

Correct allocation concealment assumed, because recent study from industry.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall drop‐out rate was 41%, most of them due to relapse (76%), which occured much more frequently in the placebo group. This difference in attrition may have biased the results of other outcomes than relapse. Kaplan‐Meier survival curve analysis was used for the primary outcome relapse.

Selective reporting (reporting bias)

High risk

Only adverse events with a frequency of at least 5% were reported.

Other bias

High risk

The study was terminated early after an interim analysis showed a clear superiority of quetiapine; there were certain baseline discrepancies in terms of mean age, duration ill and number of previous episodes.

Pfizer 2000

Methods

Randomisation: randomised, computer‐generated randomised code.
Allocation: Treatment cards numbered for each subject entering double‐blind phase, investigator and pharmacist was to allocate numbers to subjects in strict sequence of entry to study.
Blinding: double, identical capsules in blisters.
Duration: 52 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic or subchronic schizophrenia DSM‐III‐R.

N=146.
Gender: 39 women, 107 men.
Age: mean 50 years.
History: duration stable‐ n.i., duration ill‐ mean 21.5 years, number of previous hospitalisations‐ mean 10.7, age at onset‐ mean 27.7 years, severity of illness‐ PANSS 87.1, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: ziprasidone. Fixed dose. Allowed dose range: 160 mg/day. Mean dose: 160mg/day. N=71.

2. Placebo: Duration of taper: 0 days. N=75.

Rescue medication: other antipsychotics not allowed, concomitant medication for movement disorders, hypnotics, sedatives, anxiolytics.

Outcomes

Examined:

Relapse: as defined by CGI‐Improvement scale of 6 or more and/or score of 6 or more on PANSS items P7,G8 on two successive days.

Adverse effects: number of participants with at least one adverse event, akathisia, dyskinesia, dystonia, tremor, use of antiparkinson medication, weight gain.

Unable to use / Not included:

Global state: mean Clinical Global Impression Severity Scale (no means, no SDs / no predefined outcome of interest).

Mental state: Brief Psychiatric Rating Scale, AMDP system, Paranoid Depression Scale (all no means, no SDs / no predefined outcomes of interest).

Functioning: Global Assessment Scale (no mean, no SD / no predefined outcome of interest).

Subjective well‐being (own scale ‐ no mean, no SD).

Adverse effects: extrapyramidal side‐effects (Aquired Involuntary Movement Scale ‐ no SD, Simpson Angus Scale, Dosage Record and Treatment Emergent Symptoms Scale ‐ all no means, no SDs / continuous side‐effect results were not among the prespecified outcome).

Concept of illness (concept of illness scale ‐ no mean, no SD / no predefined outcome of interest).

Physiological measures: routine laboratory, ECG, EEG physical exams and vital signs (all no data / no predefined outcome of interest).

Pharmacokinetics (no predefined outcome of interest).

Compliance: doctors’ assessment (no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomised code.

Allocation concealment (selection bias)

Low risk

Treatment cards numbered for each subject entering double‐blind phase, investigator and pharmacist was to allocate numbers to subjects in strict sequence of entry to study.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

68% overall dropout, most due to relapse, which occured much more frequently in the placebo group, thus not a problem for this outcome and for drop‐out but for other outcomes.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Pietzcker 1993

Methods

Randomisation: centrally randomised by a specialised unit using an "adaptive randomisation method”.
Allocation: procedure not described.
Blinding: open, only key rating scales were additionally rated by a second blind assessor.
Duration: 2 years.
Design: parallel.
Location: multi‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia or schizoaffective disorder (ICD‐9 and Research Diagnostic Criteria).

N=237.
Gender: 124 women, 113 men.
Age: mean 34.6 years.
History: duration stable‐ at least 3 months in addition titrated to minimally effective dose which was maintained for at least 4 weeks, duration ill‐ mean 7.3 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 27.3 years, severity of illness‐ mean CGI 3.8; mean BPRS total score 28.5, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: various antipsychotic drugs. Flexible dose, minimum 100mg/day chlorpromazine equivalent. Allowed dose range: 100 ‐ unlimited chlorpromazine equivalents/day. Mean dose: 201 mg/day. N=122.

2. No treatment (=crisis management, medication was only given in case of a full relapse). Duration of taper: 50% every two weeks, thus after 6 weeks only 12.5% of initial dose left, thus 42 days. Note that participants were not withdrawn after they had received crisis intervention. N=115.

Rescue medication: in the no treatment group additional antipsychotic medication could only be given in case of relapse.

Outcomes

Examined:

Relapse: Brief Psychiatric Rating Scale total score ‐ >10 increase, Global Assessement Scale <20 reduction, deterioration Clinical Global Impression Scale CGI >7.

Unable to use / Not included:

Global state: Clinical Global Impression (no means, no SDs / no predefined outcome of interest).

Mental state: Brief Psychiatric Rating Scale, AMDP system, Paranoid Depression Scale (all no means, no SDs / no predefined outcome of interest).

Functioning: Global Assessment Scale (no mean, no SD / no predefined outcome of interest).

Subjective well‐being (own scale ‐ no mean, no SD / no predefined outcome of interest).

Adverse effects: extrapyramidal side‐effects (Aquired Involuntary Movement Scale ‐ no SD, Simpson Angus Scale, Dosage Record and Treatment Emergent Symptoms Scale ‐ all no means, no SDs / continuous side‐effect results were not among the predefined outcomes of interest).

Concept of illness (concept of illness scale ‐ no mean, no SD).

Compliance: doctors’ assessment (no predefined outcome of interest).

Physiological measures: routine laboratory, ECG, EEG (no data / no predefined outcome of interest).

Notes

There was a third group using intermittent treatment which was not of interest for this review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centrally randomised by a specialised unit using an "adaptive randomisation method”.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Open, only key rating scales were additionally rated by a second blind assessor.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Open, only key rating scales were additionally rated by a second blind assessor.

Incomplete outcome data (attrition bias)
All outcomes

High risk

High two year discontinuation rate of 43.7%. Analysis was intention‐to‐treat based on Kaplan‐Meier survival curve analysis, completer analyses were presented in addition if different. A risk of bias can not be excluded given the high discontinuation rate.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Pigott 2003

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, no further details.
Duration: 26 weeks.
Design: parallel.
Location: multi‐centre.
Setting: in‐ and outpatient, sponsored.

Participants

Diagnosis: chronic schizophrenia (DSM‐IV), at least two years of continuous antipsychotic medication.

N=310.
Gender: 174 men, 136 women.
Age: mean 42 years.
History: duration stable‐ no significant improvement or worsening of symptoms for at least 3 months, but all participants with significant symptoms (PANSS total score of at least 60, but CGI‐severity score no more than moderately ill), duration ill‐ at least 2 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean PANSS total score at baseline 81.1, mean CGI severity score at baseline 3.52, approximately 50% were in hospital, 20% were in partially supervised facilities, the rest were outpatients, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: aripiprazole. Fixed dose of 15mg/day. N=155.

2. Placebo: Duration of taper (days): n.i. (pre‐trial medication was tapered, when appropriate, before stopping treatment). N=155.

Rescue medication: additional antipsychotic drugs were not allowed.

Outcomes

Examined:

Relapse: CGI at least minimally worse, a PANSS score of ‐ 5 (moderately severe) on the subscore items of hostility or uncooperativeness on 2 successive days; or a ‐ 20% increase in PANSS total score.

Leaving the study early.

Body weight (mean change and number of participants with increase of body weight).

Unable to use / Not included:

Global state: CGI (no SD; no dichotomous data / no predefined outcome of interest).

Mental state: PANSS, BPRS (no SD / no predefined outcome of interest).

Adverse effects: extrapyramidal side‐effects ‐ Simpson‐Angus Scale, Abnormal Involuntary Movement Scale, Barnes Akathisia Scale (all no SD / No predefined outcome of interest).

Physiological measures: vital signs (pulse rate, systolic and diastolic blood pressure, no data / no predefined outcome of interest), laboratory (haematology, no data; serum chemistries, no data a apart from creatinine phosphate / no predefined outcome of interest) urine tests, ECG (both no data / no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Correct randomisation assumed, because recent study from industry..

Allocation concealment (selection bias)

Low risk

Correct allocation concealment assumed, because recent study from industry.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

A high number of participants (62.2%) left the study early, mostly because of relapse (61%), which was more frequent in the placebo group. For other outcomes this could be a problem. For the primary outcome survival analysis was used which was not a full ITT (one post‐baseline/dose) but only few participants were excluded.

Selective reporting (reporting bias)

High risk

Only those adverse events that occurred in at least 5% of the participants in either group were reported.

Other bias

Low risk

No clear other bias.

Prien 1968

Methods

Randomisation: "randomly assigned”, no further details.
Allocation: procedure not described.
Blinding: double, liquid form. no further details.
Duration: 24 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatient.

Participants

Diagnosis: schizophrenia (clinical diagnosis), continuously hospitalized for at least two years.

N=420.
Gender: n.i..
Age: mean 41.6 years.
History: duration stable‐ patients were observed on their normal hospital medicatin for eight weeks, duration ill‐ mean 17.4 years, mean age at first hospitalisation 24.2 years, mean duration of current hospitalisation‐ mean 13.1 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 24.2 years, severity of illness‐ on the average markedly ill, participants were required to show positive or negative symptoms, baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: chlorpromazine. Fixed dose of 300mg/day. N=208.

*2. Drug: chlorpromazine. Fixed dose of 2000mg/day (titrated within 45 days, dose reduction to 1500mg/day was possible). N=208.

3. Placebo: Duration of taper: 0 days. N=212.

*4. Routine treatment (any antipsychotic medication, any dose). N=210.

Rescue medication: n.i., but probably not allowed.

Outcomes

Examined:

Relapse: apatient was considered relapsed if he regressed and had to be returned to known medication before the end of the 24 week period.

Adverse effects: based on clinical interview.

Unable to use / Not included:

Mental state: Inpatient Multidimensional Psychiatric Scale, Brief Psychiatric Rating Scale (both only p‐values / no predefined outcome of interest).

Global state: CGI severity (no predefined outcome of interest).

Behaviour: Nurses’ Observation Scale for Inpatient Evaluation (only p‐values / no predefined outcome of interest).

Readiness for discharge: Discharge‐Readiness Inventory (only p‐values / no predefined outcome of interest).

Ophthalmologic examination (no predefined outcome of interest).

Notes

*We only analysed the low dose group, because the high dose was excessively high (2000mg chlorpromazine per day) and because the conventional treatment group was not double‐blind.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, liquid formulation.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, liquid formulation.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall 26% dropped out (of which 87% due to relapse). 15% of the participants in the drug group compared to 38% of the participants in the placebo group left the study early. This difference in attrition is a problem for the analysis of other outcomes than relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Prien 1969

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules. Whether blinding was successful was not assessed, although in one group high doses associated with a lot of side‐effects were administered.
Duration: 24 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), hospitalised for at least 2 years.
N=341.

Gender: n.i..
Age: mean 41.8 years.
History: duration stable‐ not clearly indicated, all were observed on their normal hospital medication for 4 weeks, quote "we may assume that the patients were well stabilised”, duration ill‐ n.i., number of previous hospitalisations‐ n.i., but mean length of current hospitalisation 15 years, age at onset‐ n.i. , severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: high dose trifluoperazine. Fixed dose of 80mg/day (reached within 35 days). N=117.

2. Drug: low dose trifluoperazine. Fixed dose of 15mg/day. N=113.

3. Placebo: Duration of taper: 0 days. N=111.

Rescue medication: not indicated, but probably not allowed.

Outcomes

Examined:

Relapse: worsening of global state.

Adverse effects: clinical interview based on 40 items checklist.

Unable to use / Not included:

Mental state: Inpatient Multidimensional Psychiatric Scale, Brief Psychiatric Rating Scale (both only p‐values / no predefined outcome of interest).

Global state: CGI (no predefined outcome of interest).

Behaviour: Nurses’ Observation Scale for Inpatient Evaluation (only p‐values / no predefined outcome of interest).

Readiness for discharge: Discharge‐Readiness Inventory (only p‐values / no predefined outcome of interest).

Ophthalmologic examination (no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules. Whether blinding was successful was not assessed, although in one group high doses associated with a lot of side‐effects were administered.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules. Whether blinding was successful was not assessed, although in one group high doses associated with a lot of side‐effects were administered.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition was considerable (33%) and clearly more participants discontinued the study early in the placebo group (53%) than in the two drug groups (23%), mainly due to inefficacy, which can be a problem for other outcomes than relapse. Not all participants were included in the final analysis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

The high‐dose group used too high doses (80mg/day) for current standards, even the low‐dose would nowadays be considered to be quite high /15mg/day).

Rifkin 1979

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐dummy technique, procyclidine was added to fluphenazine to avoid unmasking by extrapyramidal side‐effects.
Duration: one year.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (hospital diagnosis, there was an additional evaluation based on research criteria (Kraepelinian), but the results of all participants are presented here), in remission (no positive symptoms, but other symptoms could be present). Patients who were uncooperative in the stabilisation phase were not included in the study.

N=73.
Gender: 50 men, 23 women.
Age: mean 23.3 years.
History: duration stable‐ at least four weeks stable on fluphenazine before randomisation, duration ill‐ n.i., number of previous hospitalisations‐ mean 1.72 previous episodes, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: fluphenazine decanoate combined with procyclidine Flexible dose of 0.5‐2.0ml biweekly. Mean dose: n.i.. N=23.

2. Drug: oral fluphenazine combined with procyclidine. Flexible dose of 5‐20mg/day. Mean dose: n.i.. N=28.

3. Placebo: Duration of taper: 0 days. N=22.

Rescue medication: not clearly indicated, but probably not allowed.

Outcomes

Examined:

Relapse: substantial deterioration with a potential of marked social impairment.

Leaving the study early.

Adverse effects: dropout due to specific adverse events.

Unable to use / Not included:

Global state (CGI ‐ no SD, data for relapsed subgroup only).

mental state (Brief Psychiatric Rating Scale ‐ no SD, data for relapsed subgroup only).

Social adjustment: Katz Adjustment Scale ‐ no SD, only data for relapsed subgroup and a matched but not randomised subsample.

Akinesia: Periodic Evaluation Record (no SD, data for relapsed subgroup only).

Death.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double‐dummy technique, procyclidine was added to fluphenazine to avoid unmasking by extrapyramidal side‐effects.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐dummy technique, procyclidine was added to fluphenazine to avoid unmasking by extrapyramidal side‐effects.

Incomplete outcome data (attrition bias)
All outcomes

High risk

67% of the participants discontinued the study due to relapse (41%) or other reasons. More participants in the drug group discontinued due to adverse events, while more participants in the placebo group discontinued due to relapse. This differential attrition can cause bias.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Roelofs 1974

Methods

Randomisation: divided into two comparable groups by an unbiased statistician.
Allocation: procedure not explained.
Blinding: double, identical capsules.
Duration: 6 months.
Design: parallel.
Location: single‐centre.
Setting: inpatient, sponsored.

Participants

Diagnosis: chronic psychotic inpatients (clinical diagnosis), 13 schizophrenia, 1 dementia, 1 paranoia.

N=15.
Gender: 6 men, 9 women.
Age: median 54 years.
History: duration stable‐ n.i., duration ill‐ mean 17.7 years, number of previous hospitalisations‐ n.i., but mean duration of hospitalisation 11.3 years, age at onset‐ mean 36.3 years, severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: penfluridol. Fixed/flexible dose: unclear, but different doses according to pretrial medication. Allowed dose range: unclear, but all participants received 40mg/week. Mean dose: 40mg/week. N=7.

2. Placebo: Duration of taper: 0 days. N=8.

Rescue medication:
Dexetimide was given prophylactically to prevent extrapyramidal side‐effects.

Outcomes

Examined:

Relapse: need of additional antipsychotic medication.

Leaving the study early.

Unable to use / Not included:

Mental state: Zwanikken Scale (no mean, no SD / no predefined outcome of interest).

Behaviour: Zwanikken Scale (no mean, no SD / no predefined outcome of interest).

Adverse effects: interview (no data).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Divided into two comparable groups by an unbiased statistician.

Allocation concealment (selection bias)

Unclear risk

Procedure not explained.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant left the study early.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear other bias.

Ruskin 1991

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, "participants and investigators were blind to treatment”, no further details.
Duration: 6 months.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (DSM‐III), not dangerous to themselves, no hospitalisation in the last year.

N=23.
Gender: 23 men.
Age: > 50 years, mean 60.1 years.
History: duration stable‐ at least 1 month, last hospitalisation an average of 12.8 years ago, duration ill‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean BPRS psychosis subscale 6.2, baseline antipsychotic dose‐ 325 chlorpromazine equivalents (according to Davis’s equivalents).

Interventions

Before randomisation all participants were put on haloperidol for one month or until they were considered stable.

1. Drug: haloperidol. Fixed dose (dose before randomisation was maintained). Mean dose: n.i.. N=11.

2. Placebo: Duration of taper: 14 days. N=12.

Rescue medication: n.i., but probably not allowed.

Outcomes

Examined:

Relapse: significant clinical design defined by either reoccurrence of symptoms or worsening of existing symptoms or prodromals signs such as sleep problems or anxiety.

Unable to use / Not included:

Mental state: BPRS (no data for each group / no predefined outcome of interest).

Quality of life: Heinrich Scale (no data for each group).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, "participants and investigators were blind to treatment”, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, "participants and investigators were blind to treatment”, no further details.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

47% of the participants left the study early, most of them due to a relapse (55%). This attrition can be a source of bias for other outcomes than relapse.

Selective reporting (reporting bias)

High risk

Data on quality of life were not reported.

Other bias

Low risk

No clear evidence for other bias.

Sampath 1992

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, placebo was sesame oil of identical volume and identical in physical appearance.
Duration: 12 months.
Design: parallel.
Location: single‐centre.
Setting: inpatient, sponsored.

Participants

Diagnosis: chronic schizophrenia (Research Diagnostic Criteria), stable for at least 5 years (absence of clinical deterioration and/or an increase of neuroleptic medication, retrospectively and in addition prospectively for at least 12 months), all on fluphenazine decanoate.

N=24.
Gender: n.i..
Age: mean 57.3 years.
History: duration stable‐ retrospectively at least 5 years, prospectively for 12 months, mean 7 years, duration ill‐ mean 33.1 years, number of previous hospitalisations‐ n.i., but mean duration of hospitalisation 24.9 years (unclear whether current or life‐time total), age at onset‐ mean 24.3 years, severity of illness‐ mean BPRS total score 24.9, baseline antipsychotic dose‐ mean 41.9 mg fluphenazine / 4 weeks.

Interventions

1. Drug: fluphenazine decanoate.Fixed dose: mean 50.4mg/4 weeks. N=12.

2. Placebo: Duration of taper: 0 days, but all participants were on depot medication before the study. N=12.

Rescue medication: n.i., but probably not allowed.

Outcomes

Examined:

Relapse: at least 25% increase of Brief Psychiatric Rating Scale total score and judgement of by nurse according to Psychotic Inpatient Profile.

Unable to use / Not included:

Mental state: Brief Psychiatric Rating Scale total, Psychotic Inpatient Profile (for both scales means for subgroups only / no predefined outcome of interest).

Physiological measures: prolactin levels (no SD’s / no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo was sesame oil of identical volume and identical in physical appearance.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo was sesame oil of identical volume and identical in physical appearance.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There is no statement on participants leaving the study early.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

There was a baseline imbalance in terms of gender and in terms of baseline fluphenazine dose.

Schering Plough 2010

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules in taste.
Duration: 6 months.
Design: parallel.
Location: multi‐centre.
Setting: unclear.

Participants

Diagnosis: schizophrenia (DSM‐IV).

N=386.
Gender: 221 men, 165 women.
Age: mean 38.9 years.
History: duration stable‐ 30 weeks, duration ill‐ mean 12.7 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 26.7 years, severity of illness‐ n.i., baseline antipsychotic dose‐ all on asenapine 10 or 20mg/day.

Interventions

1. Drug: asenapine. Fixed dose (same dose as at end of stabilisation phase): mean 17.6 mg/day. N=194.

2. Placebo: Duration of taper: 0 days. N=192.

Rescue medication: benzodiazepines, anticholinergics, antidepressants.

Outcomes

Examined:

Relapse: CGI‐severity >=4, moderately ill for one week was accompanied by: PANSS total score increase >=20% (a 10 point increase if PANSS was lower than 50), a PANSS item score >=5 on hostility of uncooperativeness or a PANSS item score >=5 and two items of unusual thought content, conceptual disorganisation or hallucinatory behaviour. Relapse was also judged to appear if in the investigator's opinion schizophrenia, risk of violence to self or others, or suicide risk increased so >=1 of the following was required: an additional >=2mg/day lorazepam, compared with the highest open label dose for 1 week, addition of antipsychotic, addition or dosage increase of an antidepressant or mood‐stabiliser, increased psychiatric care, arrest or imprisonment, electroconvulsive therapy, or other relevant measures.

Death: suicidal ideation, suicide attempts.

Adverse effects: at least one adverse event, at least one movement disorder, akathisia, sedation, weight gain.

Unable to use / Not included:

Mental state: PANSS (no predefined outcome of interest).

Global state: CGI (no predefined outcome of interest).

Leaving the study early (data are unclear).

Hospitalisation (no data).

Electrocardiogram (no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules in taste.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules in taste.

Incomplete outcome data (attrition bias)
All outcomes

High risk

High dropout rate, but exact number of drop‐outs could not be calculated. Drop‐outs were not clearly enough reported. Survival curve analysis was used for the primary outcome relapse.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for selective reporting.

Schiele 1961

Methods

Randomisation: random, no further details.
Allocation: only the hospital pharmacist had the code on what medication the patient was on.
Blinding: double, identical capsules, each participant had his own container.
Duration: 16 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: schizophrenia (clinical diagnosis), all withdrawn of subject to periodic disturbances, all needed supervision or management.

N=80.
Gender: 80 men.
Age: younger than 55 years, mean 40.6 years.
History: duration stable‐ n.i. ("participants had attained and maintained some degree of improvement”), duration ill‐ n.i., but mean duration of current hospitalisation 10 years, number of previous hospitalisations‐ mean 1.6, age at onset‐ n.i., severity of illness‐ n.i., but "most required closed ward care”, median baseline antipsychotic dose‐ chlorpromazine 475mg/day (n=30), mepazine 200 mg/day (n=35), trifluoperazine 30 mg/day (n=6), prochlorpromazine (n=2, dose not indicated), combinations of drugs (n=7, doses not indicated)

Interventions

1. Drug: chlorpromazine; flexible dose; allowed dose range 200 to 1000 mg/day; mean dose: 894 mg/day (here mean maximum dose); N=20

2. Drug: trifluoperazine; flexible dose; allowed dose range 10 to 50 mg/day; mean dose: 29 mg/day (here mean maximum dose); N=20

3. Drug: thioridazine; flexible dose; allowed dose range 200 to 1000 mg/day; mean dose: 958 mg/day (here mean maximum dose); N=20

4. Placebo: duration of taper: 0 days; N=20

Rescue medication: phenobarbital and bentropine methansulfonate, no additional antipsychotic drugs

Outcomes

Examined:

Relapse: worsening of global state

Adverse effects: clinical interview, number of participants receiving antiparkinson medication

Unable to use / Not included:

Global state: CGI‐Severity Scale (no predefined outcome of interest)

Behaviour: Manifest Behaviour Scale (no SD / no predefined outcome of interest)

Personality: Minnesota Multiphasic Personality Inventory (no SD / no predefined outcome of interest)

Notes

The results of all drug groups were pooled

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details

Allocation concealment (selection bias)

Low risk

Only the hospital pharmacist had the code on what medication the patient was on

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules, each participant had his own container.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules, each participant had his own container.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 3 out of 80 participants left the study early and the reasons were well described.

Selective reporting (reporting bias)

Low risk

No selective reporting.

Other bias

Low risk

No evidence for other bias.

Shawver 1959

Methods

Randomisation: matched and then randomised by a research assistant.
Allocation: by a research assistant who carefully guarded the identity of patients and the assigned treatment regimen. Furthermore, medication was assigned by the director of professional services who kept the names for use in case a patient had to be withdrawn from the study.
Blinding: double, identical capsules.
Duration: 26 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), less than 50 years, on chlorpromazine for at least six months, had reached a stable improved state.

N=80.
Gender: n.i..
Age: all <50 years.
History: duration stable‐ n.i., duration ill‐ n.i., number of previous hospitalisations‐ n.i., but median duration of current hospitalisation eight years, age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: chlorpromazine. Fixed dose of 200mg/day. N=40.

2. Drug: reserpine*. Fixed dose of 2 mg/day. N=40.

3. Placebo: Duration of taper 0 days. N=40.

Rescue medication: not indicated, probably not allowed.

Outcomes

Examined:

Leaving the study early.

Unable to use / Not included:

Mental state: Lorr Multidimensional Scale for Rating Psychiatric Patients (no SD / no predefined outcome of interest).

Behaviour: Psychiatric Behaviour Rating Scales (no SD / no predefined outcome of interest).

Notes

*this group was not used in the analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Matched and then randomised by a research assistant.

Allocation concealment (selection bias)

Low risk

By a research assistant who carefully guarded the identity of patients and the assigned treatment regimen. Furthermore, medication was assigned by the director of professional services who kept the names for use in case a patient had to be withdrawn from the study.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall 11% dropped out, most of them due to relapse (88%) in the placebo group. As relapse, drop‐out and suicide were the only outcomes, this did not produce a risk of bias.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence of other bias.

Spohn 1986

Methods

Randomisation: random, no further details.
Allocation: procedure not explained.
Blinding: double, placebo matching in kind and dose the previous medication.
Duration: 10 weeks.
Design: parallel.
Location: three hospitals.
Setting: probably inpatients.

Participants

Diagnosis: schizophrenia (Schedule for Affective Disorder and Schizophrenia, and Research Diagnostic Criteria), all had previously responded to antipsychotic drugs.

N=100.
Gender:  73 men, 27 women.
Age: mean 32.6 years.
History: duration stable‐ prospectively participants had remained for 10 weeks on the same medication before the study, duration ill‐ mean 9.7 years, number of previous hospitalisations‐ n.i., average cumulative hospitalisation 6.5 years, age at onset‐ mean 22.9 years, severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: various antipsychotic drugs. Fixed/flexible dose: probably flexible. Allowed dose range: n.i.. Mean dose: n.i.. N=36.

2. Placebo: Duration of taper 0 days. N=64.

Rescue medication: n.i..

Outcomes

Examined:

Relapse: first signs of symptoms according to ward staff and project nurse, full deterioration was not waited for.

Unable to use / Not included:

Performance tests: Rohrschach test, Wechsler Adult Intelligence Scale (all no clear mean’s, n´s, no SD’s / no predefined outcomes of interest).

Mental state: Brief Psychiatric Rating Scale (no clear mean, no number of participatns, no SD / no predefined outcome of interest).

Thought disorder: Thought Disorder Index, Psychological Rating Scale (all no clear mean’s, no SD’s / no predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not explained.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo matching in kind and dose the previous medication.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo matching in kind and dose the previous medication.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear, because these have not been indicated.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Unclear risk

Unclear, baseline data have not been presented for both groups separately.

Troshinsky 1962

Methods

Randomisation: randomised, no further details.
Allocation: psychiatrist without contact to the participants held the key and filled the medication containers.
Blinding: double, exact placebo replicas.
Duration: ˜ 43 weeks.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia without positive symptoms (clinical diagnosis).

N=43.
Gender: 16 men, 27 women.
Age: typically 40‐50 years.
History: duration stable‐ out of hospital for at least a year (typically 2‐4 years), duration ill‐ n.i., number of previous hospitalisations‐ typically 2‐3, age at onset n.i., severity of illness n.i., but no positive symptoms at baseline, baseline antipsychotic dose‐ maximum 300mg chlorpromazine per day.

Interventions

1. Drug: various phenothiazines, mainly chlorpromazine. Fixed/flexible dose: flexible. Allowed dose range: not limited, but complete discontinuation was not allowed. Mean dose: 150‐200mg/day chlorpromazine. N=24.

2. Placebo: Duration of taper: 0 days. N=19.

Rescue medication: not allowed.

Outcomes

Examined:

Relapse: clinical judgement.

Service use: number of participants rehospitalised.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Low risk

Psychiatrist without contact to the participants held the key and filled the medication containers.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, exact placebo replicas.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, exact placebo replicas.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear ‐ whether participants discontinued the study prematurely was not reported.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Some placebo participants continued to take medication, study terminated early.

Vandecasteele 1974

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double, identical capsules.
Duration: 6 months
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (DSM‐II), catatonic type (n=2), residual type (n=15), hebephrenic type (n=1), simple type (n=2), paranoid type (n=1).

N=21.
Gender: 21 women.
Age: mean 58 years.
History: duration stable‐ successfully maintained on penfluridol for at least 6 months, duration ill‐ mean 28.5 years, median duration of current hospitalisation 21 years, number of previous hospitalisations‐ n.i., age at onset‐ 29.5 years, severity of illness‐ n.i., baseline antipsychotic dose‐ mean 43 mg/week penfluridol.

Interventions

1. Drug: penfluridol. Fixed dose, mean 43mg/week. N=10.

2. Placebo: Duration of taper: 0 days. N=11.

Rescue medication: antiparkinson medication, haloperidol, but this was considered to be a sign of relapse.

Outcomes

Examined:

Relapse: use of additional haloperidol.

Leaving the study early.

Unable to use / Not included:

Mental state: Zwanikken scale (no data / no predefined outcome of interest).

Adverse effects: Zwanikken scale (no data / continuous side‐effect results were not among the predefined outcomes of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No participant left the study prematurely.

Selective reporting (reporting bias)

High risk

Data on side‐effects and the mental state were not reported.

Other bias

Low risk

No evidence for other bias.

Whittaker 1963

Methods

Randomisation: arbitrarily allocated.
Allocation: procedure not described.
Blinding: double (only the pharmacist knew which bottles were active. Participants were asked whether they were aware of the medication, but only one realised a change in taste. Nurses were also asked, but did not guess the right medication better than by chance alone.
Duration: 10 weeks.
Design: parallel.
Location: two centres.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis by at least two psychiatrists), all with paranoid condition, two additionally catatonic tendencies and one hebephrenic features, six were leucotomised.

N=26.
Gender: 26 men.
Age: mean 50.7 years.
History: duration stable‐ n.i., but all had been receiving maintenance doses of perphenazine for a mean of 16 months, duration ill‐ n.i., but mean duration of current hospitalisation 16.5 years, number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ n.i., baseline antipsychotic dose‐ two 12mg tid, one 20mg tid, all other 8mg tid and most less.

Interventions

1. Drug: perphenazine liquid. Fixed dose (same dose as before the start of the study) two 12mg tid, one 20mg tid, all other 8mg tid and most less. Mean dose: see above. N=13.

2. Placebo. Duration of taper: 0 days. N=13.

3. No medication*. Duration of taper: 0 days. N=13.

Rescue medication: not allowed apart from antiparkinson medication.

Outcomes

Examined:

Relapse: "major relapse" = replaced on active medication.

Unable to use / Not included:

Mental state: self‐developed psychiatric rating scale ‐ unpublished scale (no predefined outcome of interest).

Behaviour: Fergus Falls Behaviour Rating Scale (no mean, no SD / no predefined outcome of interest).

Notes

*This group was not used for the review.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Arbitrarily allocated.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double ‐ only the pharmacist knew which bottles were active. Participants were asked whether they were aware of the medication, but only one realised a change in taste. Nurses were also asked, but did not guess the correct medication better than by chance alone.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double ‐ only the pharmacist knew which bottles were active. Participants were asked whether they were aware of the medication, but only one realised a change in taste. Nurses were also asked, but did not guess the correct medication better than by chance alone.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts were not reported. It is not clear, whether there really no dropouts.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No clear evidence for other bias.

Wistedt 1981

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double, placebo sesame oil.
Duration: 26 weeks.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (according to Bleuler’s concept, with three primary symptoms), duration ill at least 2 years.

N=41.
Gender: 15 men, 23 women.
Age: mean 43.1 years.
History: duration stable‐ outpatient and continuous antipsychotic treatment for at least one year, on flupenthixol depot or fluphenazine depot for at least three months, prospective stabilisation phase of 6 months, duration ill‐ mean 13.3 years, number of previous hospitalisations‐ n.i., age at onset‐ mean 29.8 years, severity of illness‐ mean Comprehensive Psychopathological Rating Scale schizophrenia score 2.3, baseline antipsychotic dose‐ mean 21.42mg fluphenazine/3 weeks or 27.5mg flupenthixol/three weeks.

Interventions

1. Drug: fluphenazine depot (most around 12.5 ‐ 25 mg/3 weeks, mean 21.42mg/3 weeks) or flupenthixol depot (most around 20‐40mg/3 weeks, mean 27.5/3 weeks) ‐ Fixed dose. N=24.

2. Placebo: Duration of taper: 0 days. N=17.

Rescue medication: chloral hydrate, antiparkinson medication, additional antipsychotic drugs were not allowed.

Outcomes

Examined:

Relapse: psychotic behaviour or increase in six subscales of the Comprehensive Psychopathological Rating Scale.

Unable to use / Not included:

Mental state: Comprehensive Psychopathological Rating Scale (no SD / no predefined outcome of interest).

Behaviour: Nurses Observation Scale of Inpatient Evaluation (no SD / no predefined outcome of interest).

Adverse effects: extrapyramidal side‐effects (Simpson Angus Scale, Acquired Involuntary Movements Scale, Akathisia Rating Scale (all no SD, akathisia scale was not published / continuous side‐effect results were not among the predefined outcomes of interest).

Physiological measures: various laboratory tests (no data / no predefined outcome of interest).

Life events (Life Event Scale / no predefined outcome of interest).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Random, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, placebo sesame oil.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, placebo sesame oil.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall 3 (7%) out of 41 participants left the study early. Although only completers were analysed, due to the low rate this is not a problem.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No obvious risk for other bias.

Zissis 1982

Methods

Randomisation: randomly assigned according to pre‐established randomisation code.
Allocation: randomisation code was unknown to the evaluating investigators.
Blinding: double, administered by a particular nurse.
Duration: 16 weeks.
Design: parallel.
Location: single‐centre.
Setting: inpatient.

Participants

Diagnosis: chronic schizophrenia (Feighner’s criteria), treated with antipsychotic drugs for at least 2 years and currently under control.

N=32.
Gender: 9 men, 23 women.
Age: mean 46.5 years.
History: duration stable‐ n.i., but treated with antipsychotic drugs for at least 2 years and currently under control, duration ill‐ mean 24.4 years, number of previous hospitalisations‐ n.i., but mean duration of current hospitalisation 9.6 years, age at onset‐ mean 22.1 years, severity of illness‐ n.i., baseline antipsychotic dose‐ n.i..

Interventions

1. Drug: haloperidol decanoate. Flexible dose. Allowed dose range: starting dose 1.5 ml (=150mg) four‐weekly, maximum 3ml (=300mg) four‐weekly. Median dose 1.5 ml four‐weekly. N=16.

2. Placebo: Duration of taper: 0 days. N=16.

Rescue medication: antiparkinson medication, oral haloperidol, but this was considered to be a relapse.

Outcomes

Examined:

Relapse: addition of oral haloperidol.

Leaving the study early.

Adverse effects: open interviews.

Unable to use / Not included:

Mental state: Brief Psychiatric Rating Scale (no SD / no predefined outcome of interest).

Global state: Clinical Global Impression Severity Scale (no predefined outcome of interest).

Behaviour: Nurses Observation Scale for Inpatient Evaluation (no SD / no predefined outcome of interest).

Adverse effects: use of antiparkinson medication (only indicated for haloperidol group), at least one movement disorder and sedation (patients received haloperidol).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly assigned according to pre‐established randomisation code.

Allocation concealment (selection bias)

Low risk

Randomisation code was unknown to the evaluating investigators.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double, administered by a particular nurse.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double, administered by a particular nurse.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

12 out of 16 participants in the placebo group compared to 0 out of 16 in the haloperidol group were withdrawn from the trial due to inefficacy of treatment. As the only outcomes were relapse, number of participants improved and leaving the study early this should not have been a problem.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No other bias.

General abbreviations

CNS: central nervous system

CPZ: chlorpromazine

DSM: Diagnostic and Statistical Manual of Mental Disorders

ECG: electrocardiography

ECT: electroconvulsive therapy

EASY: Early Assessment Service for Young People with Psychosis

EEG: electroencephalography

EPS: extrapyramidal symptoms

HbA1c: glycated haemoglobin

ICD: International Statistical Classification of Diseases and Related Health Problems

IM: intramuscular injection

ITT: intention to treat

LOCF: last observation carried forward

NI: not indicated

SD: standard deviation

tid: ter in die (3 times a day)

Rating scales

AIMS: Abnormal Involuntary Movement Scale

AMDP: Arbeitsgemeinschaft für Methodik und Dokumentation in der Psychiatrie

BAS: Barnes Akathisia Scale

CGI: Clinical Global Impression

BPRS: Brief Psychiatric Rating Scale

GAS: Global Assessment Scale

IMPS: Inpatient Multidimensional Psychiatric Rating Scale

MMPI: Minnesota Multiphasic Personality Inventory

NOSIE: Nurses Observation Scale for Inpatient Evaluation

PANSS: Positive And Negative Syndrome Scale

PRP: Psychotic Reaction Profile

PRS: Psychiatric Rating Scale

PSE: Present State Examination

RDC: Research Diagnostic Criteria

SADS: Schedule for Affective Disorders

SANS: Scale for the Assessment of Negative Symptoms

SAS: Simpson‐Angus Scale

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Allen 1997

Allocation: controlled clinical trial, not randomised.

Bourin 2008

Allocation: randomised.

Participants: not stabilised on antipsychotic drugs.

Branchey 1981

Allocation: not randomised, matched groups.

Breier 1987

Allocation: not randomised.

Chouinard 1980

Allocation: not randomised.

Claghorn 1974

Allocation: randomised.

Participants: schizophrenia.

Intervention: thiothixene alone versus thiothixene plus group therapy versus chlorpromazine alone versus chlorpromazine plus group therapy.

Collins 1967

Allocation: not randomised.

Condray 1995

Allocation: not randomised.

Curson 1985

Allocation: not randomised.

Degkwitz 1970

Allocation: not randomised.

Diamond 1960

Allocation: not randomised.

Double 1993

Allocation: randomised.

Participants: schizophrenia.

Intervention: all participants were on neuroleptics and antiparkinson medication at baseline. They were then randomised to neuroleptics plus continuation of antiparkinson medication versus neuroleptics alone.

Engelhardt 1967

Allocation: randomised.

Participants: chronic schizophrenic outpatients, not truly stabilised on antipsychotic drugs.

Gleeson 2004

Allocation: randomised.

Participants: first‐episode psychosis.

Intervention: treatment as usual (including antipsychotics) versus multimodal relapse prevention therapy (including antipsychotics and cognitive behavioral therapy/family intervention).

Goldberg 1967

Allocation: not randomised.

Good 1958

Allocation: randomised.

Participants: schizophrenia.

Interventions: chlorpromazine versus placebo.

Outcomes: no usable outcome.

Greenberg 1966

Allocation: randomised.

Participants: chronic schizophrenic patients.

Intervention: abrupt versus gradual withdrawal of chlorpromazine, but chlorpromazine was withdrawn from both groups. Thus not appropriate control group.

Hine 1958

Allocation: not randomised.

Hunt 1967

Allocation: not randomised.

Ionescu 1983

Allocation: not randomised.

Janecek 1963

Allocation: randomised.

Participants: 50% not diagnosed as with schizophrenia.

Johnstone 1988

Allocation: not randomised.

Kellam 1971

Allocation: not randomised.

Lauriello 2005

Allocation: randomised.

Participants: participants were acutely ill, not stable.

Lecrubier 1997

Allocation: randomised.

Participants: not stable, not all on antipsychotics before the study.

Loo 1997

Allocation: randomised.

Participants: participants were not stable, most not on antipsychotics before the study.

Mefferd 1958

Allocation: randomised.

Participants: men with schizophrenia.

Intervention: chlorpromazine versus placebo.

Outcome: no usable outcome.

Mosher 1975

Allocation: not randomised.

Müller 1982

Allocation: part of the participants was matched, not randomised.

Paul 1972

Allocation: not randomised.

Peet 1981

Allocation: randomised.

Participants: schizophrenia.

Intervention: chlorpromazine versus chlorpromazine plus propranolol.

Pickar 1986

Allocation: not randomised.

Pickar 2003

Allocation: not randomised.

Pigache 1993

Allocation: randomised.

Participants: chronic schizophrenia.

Intervention: chlorpromazine, placebo, orphenadrine.

Outcome: no relevant outcome, only auditory attention task.

Rassidakis 1970

Allocation: not randomised.

Ravaris 1965

Allocation: randomised.

Participants: chronic schizophrenia.

Intervention: fluphenazine elixir plus placebo injection versus fluphenazine enanthate injection plus oral placebo.

Schlossberg 1978

Allocation: randomised.

Participants: not stable.

Singh 1990

Allocation: not randomised.

Smelson 2006

Allocation: not randomised.

Soni 1990

Allocation: randomised.

Participants: schizophrenia, not stabilised on antipsychotic drugs, because all had been withdrawn from antipsychotic drugs for 8‐20 months before study start.

Tollefson 1999

Allocation: randomised, but switch study with very short duration (3‐5 days).

Vaddadi 1986

Allocation: randomised.

Participants: schizophrenia.

Intervention: depot antipsychotics (fluphenazine depot, flupenthixol depot or clopenthixol depot) plus oral dihomo gammalinolenic acid (DHLA) versus oral DHLA plus placebo injections versus DHLA placebo capsules and placebo injections. What is lacking is a depot antipsychotic only group.

Van Kammen 1982

Allocation: not randomised.

Van Praag 1973

Allocation: randomised.

Participants: psychotic participants.

Intervention: fluphenazine enanthate versus fluphenazine decanoate.

Wiedemann 2001

Allocation: randomised.

Participants: schizophrenia.

Intervention: continuation of current antipsychotic versus gradual withdrawal. However, antipsychotic was given again when early warning signs appeared, i.e. intermittent treatment,  a design that was excluded a prior by our protocol.

Wright 1964

Allocation: not randomised.

Wunderink 2006

Allocation: randomised.

Participants: schizophrenia and related psychotic disorder.

Intervention: continuation of current antipsychotic versus gradual withdrawal. However, antipsychotic was given again when early warning signs appeared, i.e. intermittent treatment,  a design that was excluded by the protocol. Approximately 50% of participants were never withdrawn.

Zeller 1956

Allocation: all participants were in hospital. 95 were allocated to placebo (not randomly). Then 81 participants were "selected at random to match” the intervention group. We feel that this is no appropriate randomisation method.

Zwanikken 1973

Allocation: randomised.

Participants: more than 50% had mental retardation, not schizophrenia.

Characteristics of ongoing studies [ordered by study ID]

Eerdekens 2010

Trial name or title

A randomised double‐blind placebo‐controlled parallel group study evaluating paliperidone palmitate in the prevention of recurrence in patients with schizophrenia.

Methods

Allocation: randomised, no further details.

Blinding: double‐blind, no further details.

Location: multicentre study.

Duration: the study consists of 5 periods: an up to 7‐day screening/washout/tolerability period, a 9‐week open‐label transition period, a 24‐week open‐label maintenance period, a randomised, variable‐length double‐blind, placebo‐controlled recurrence prevention period, and an up to 52‐week open‐label extension period.

Participants

Diagnosis: stable and symptomatic schizophrenia according to DSM‐IV‐TM.

Interventions

1. Paliperidone palmitate.

2. Placebo.

Outcomes

The primary outcome is the time from randomisation to the first recurrence.

Mental state: PANSS.

Global state: CGI‐S.

Personal and Social Performance Scale, adverse events, labs and ECG‐tests.

Starting date

March 2005.

Contact information

ClinicalTrials.gov identifier: NCT00111189.

Notes

CGI: Clinical Global Impression

DSM: Diagnostic and Statistical Manual of Mental Disorders

ECG: electrocardiography

PANSS: Positive And Negative Syndrome Scale

Data and analyses

Open in table viewer
Comparison 1. Maintenance treatment with antipsychotic drugs versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse: up to 3 months Show forest plot

34

3942

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.24, 0.38]

Analysis 1.1

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1 Relapse: up to 3 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1 Relapse: up to 3 months.

2 Relapse: 4 to 6 months Show forest plot

40

5285

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.30, 0.42]

Analysis 1.2

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2 Relapse: 4 to 6 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2 Relapse: 4 to 6 months.

3 Relapse: 7 to 12 months Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 1.3

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3 Relapse: 7 to 12 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3 Relapse: 7 to 12 months.

4 Relapse: > 12 months Show forest plot

6

811

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.42, 0.82]

Analysis 1.4

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4 Relapse: > 12 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4 Relapse: > 12 months.

5 Relapse: independent of duration Show forest plot

62

6392

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.29, 0.41]

Analysis 1.5

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5 Relapse: independent of duration.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5 Relapse: independent of duration.

6 Leaving the study early: due to any reason Show forest plot

47

4718

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.46, 0.61]

Analysis 1.6

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6 Leaving the study early: due to any reason.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6 Leaving the study early: due to any reason.

6.1 up to 3 months

8

245

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.07, 0.72]

6.2 4 to 6 months

17

1646

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.35, 0.66]

6.3 7 to 12 months

18

2420

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.46, 0.66]

6.4 > 12 months

4

407

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.36, 1.26]

7 Leaving the study early: due to adverse events Show forest plot

43

4333

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.70, 1.91]

Analysis 1.7

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7 Leaving the study early: due to adverse events.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7 Leaving the study early: due to adverse events.

7.1 up to 3 months

8

245

Risk Ratio (M‐H, Random, 95% CI)

2.84 [0.12, 65.34]

7.2 4 to 6 months

14

1549

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.57, 1.74]

7.3 7 to 12 months

17

2339

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.59, 2.60]

7.4 > 12 months

4

200

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8 Leaving the study early: due to inefficacy Show forest plot

46

4546

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

Analysis 1.8

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8 Leaving the study early: due to inefficacy.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8 Leaving the study early: due to inefficacy.

8.1 up to 3 months

9

295

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.07, 0.79]

8.2 4 to 6 months

16

1661

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.31, 0.54]

8.3 7 to 12 months

18

2420

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.28, 0.45]

8.4 > 12 months

3

170

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.08, 0.95]

9 Global state: number of participants improved Show forest plot

14

1524

Risk Ratio (M‐H, Random, 95% CI)

2.34 [1.68, 3.26]

Analysis 1.9

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9 Global state: number of participants improved.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9 Global state: number of participants improved.

9.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

4.61 [1.22, 17.40]

9.2 4 to 6 months

8

1037

Risk Ratio (M‐H, Random, 95% CI)

2.33 [1.69, 3.21]

9.3 7 to 12 months

5

438

Risk Ratio (M‐H, Random, 95% CI)

1.95 [0.91, 4.18]

10 Service use: number of participants hospitalised Show forest plot

16

2090

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.27, 0.55]

Analysis 1.10

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10 Service use: number of participants hospitalised.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10 Service use: number of participants hospitalised.

10.1 up to 3 months

2

55

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.04, 4.06]

10.2 4 to 6 months

3

109

Risk Ratio (M‐H, Random, 95% CI)

0.08 [0.01, 0.42]

10.3 7 to 12 months

8

1295

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.18, 0.57]

10.4 > 12 months

3

631

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.44, 0.70]

11 Service use: number of participants discharged Show forest plot

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

Analysis 1.11

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11 Service use: number of participants discharged.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11 Service use: number of participants discharged.

11.1 4 to 6 months

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

12 Death: any Show forest plot

14

2356

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.28, 2.11]

Analysis 1.12

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12 Death: any.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12 Death: any.

12.1 up to 3 months

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

12.2 4 to 6 months

5

856

Risk Ratio (M‐H, Random, 95% CI)

2.18 [0.48, 9.81]

12.3 7 to 12 months

8

1464

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.08, 1.27]

13 Death: due to natural causes Show forest plot

14

2401

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.39, 3.97]

Analysis 1.13

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13 Death: due to natural causes.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13 Death: due to natural causes.

13.1 4 to 6 months

5

856

Risk Ratio (M‐H, Random, 95% CI)

2.18 [0.48, 9.81]

13.2 7 to 12 months

9

1545

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.09, 3.36]

14 Suicide Show forest plot

8

1941

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.04, 3.28]

Analysis 1.14

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14 Suicide.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14 Suicide.

14.1 up to 3 months

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.2 4 to 6 months

2

730

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.3 7 to 12 months

5

1175

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.04, 3.28]

15 Suicide attempts Show forest plot

5

1177

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.10, 2.33]

Analysis 1.15

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15 Suicide attempts.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15 Suicide attempts.

15.1 4 to 6 months

2

466

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 71.51]

15.2 7 to 12 months

3

711

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.04, 1.61]

16 Suicide ideation Show forest plot

3

556

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.04, 10.56]

Analysis 1.16

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16 Suicide ideation.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16 Suicide ideation.

16.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 3.88]

16.2 4 to 6 months

1

386

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

16.3 7 to 12 months

1

121

Risk Ratio (M‐H, Random, 95% CI)

2.77 [0.11, 66.57]

17 Violent/aggressive behaviour Show forest plot

5

680

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.15, 0.52]

Analysis 1.17

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17 Violent/aggressive behaviour.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17 Violent/aggressive behaviour.

17.1 up to 3 months

1

26

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.50]

17.2 4 to 6 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.2 [0.01, 3.92]

17.3 7 to 12 months

3

614

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.14, 0.53]

18 Adverse effects: at least one adverse event Show forest plot

10

2184

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.87, 1.18]

Analysis 1.18

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18 Adverse effects: at least one adverse event.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18 Adverse effects: at least one adverse event.

18.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.30, 0.93]

18.2 4 to 6 months

3

776

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.15]

18.3 7 to 12 months

6

1359

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.88, 1.38]

19 Adverse effects: movement disorders: at least one movement disorder Show forest plot

22

3411

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.25, 1.93]

Analysis 1.19

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19 Adverse effects: movement disorders: at least one movement disorder.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19 Adverse effects: movement disorders: at least one movement disorder.

19.1 up to 3 months

4

158

Risk Ratio (M‐H, Random, 95% CI)

2.42 [0.70, 8.33]

19.2 4 to 6 months

8

1658

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.06, 1.99]

19.3 7 to 12 months

10

1595

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.11, 2.07]

20 Adverse effects: movement disorders: akathisia Show forest plot

12

2026

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.87, 3.51]

Analysis 1.20

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20 Adverse effects: movement disorders: akathisia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20 Adverse effects: movement disorders: akathisia.

20.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.94 [0.24, 15.97]

20.2 4 to 6 months

6

1009

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.41, 6.80]

20.3 7 to 12 months

5

968

Risk Ratio (M‐H, Random, 95% CI)

1.74 [0.88, 3.45]

21 Adverse effects: movement disorders: akinesia Show forest plot

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

Analysis 1.21

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21 Adverse effects: movement disorders: akinesia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21 Adverse effects: movement disorders: akinesia.

21.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

22 Adverse effects: movement disorders: dyskinesia Show forest plot

13

1820

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.28, 0.97]

Analysis 1.22

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22 Adverse effects: movement disorders: dyskinesia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22 Adverse effects: movement disorders: dyskinesia.

22.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.5 [0.06, 34.91]

22.2 4 to 6 months

3

418

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.84]

22.3 7 to 12 months

9

1353

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.30, 1.58]

23 Adverse effects: movement disorders: dystonia Show forest plot

6

824

Risk Ratio (M‐H, Random, 95% CI)

1.89 [1.05, 3.41]

Analysis 1.23

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23 Adverse effects: movement disorders: dystonia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23 Adverse effects: movement disorders: dystonia.

23.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

2.5 [0.13, 49.22]

23.2 4 to 6 months

2

382

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.29]

23.3 7 to 12 months

3

393

Risk Ratio (M‐H, Random, 95% CI)

3.97 [0.44, 35.54]

24 Adverse effects: movement disorders: rigor Show forest plot

5

249

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.54, 2.88]

Analysis 1.24

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24 Adverse effects: movement disorders: rigor.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24 Adverse effects: movement disorders: rigor.

24.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.24, 2.22]

24.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

1.98 [0.67, 5.85]

24.3 7 to 12 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25 Adverse effects: movement disorders: tremor Show forest plot

10

1468

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.81, 1.93]

Analysis 1.25

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25 Adverse effects: movement disorders: tremor.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25 Adverse effects: movement disorders: tremor.

25.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.40, 3.01]

25.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.33, 2.61]

25.3 7 to 12 months

6

1259

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.82, 2.43]

26 Adverse effects: movement disorders: use of antiparkinson medication Show forest plot

7

1317

Risk Ratio (M‐H, Random, 95% CI)

1.40 [1.03, 1.89]

Analysis 1.26

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26 Adverse effects: movement disorders: use of antiparkinson medication.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26 Adverse effects: movement disorders: use of antiparkinson medication.

26.1 4 to 6 months

3

841

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.90, 2.61]

26.2 7 to 12 months

4

476

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.86, 2.05]

27 Adverse effects: sedation Show forest plot

10

2146

Risk Ratio (M‐H, Random, 95% CI)

1.50 [1.22, 1.84]

Analysis 1.27

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27 Adverse effects: sedation.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27 Adverse effects: sedation.

27.1 4 to 6 months

6

1577

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.86, 2.07]

27.2 7 to 12 months

4

569

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.90, 3.31]

28 Adverse effects: weight gain Show forest plot

10

2321

Risk Ratio (M‐H, Random, 95% CI)

2.07 [1.31, 3.25]

Analysis 1.28

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28 Adverse effects: weight gain.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28 Adverse effects: weight gain.

28.1 4 to 6 months

3

736

Risk Ratio (M‐H, Random, 95% CI)

1.76 [0.92, 3.37]

28.2 7 to 12 months

7

1585

Risk Ratio (M‐H, Random, 95% CI)

2.57 [1.30, 5.07]

29 Quality of life Show forest plot

3

527

Std. Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.15, ‐0.09]

Analysis 1.29

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29 Quality of life.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29 Quality of life.

29.1 (7 to 12) months

2

509

Std. Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.26, 0.01]

29.2 (> 12 ) months

1

18

Std. Mean Difference (IV, Random, 95% CI)

‐0.61 [‐1.66, 0.45]

30 Number of participants employed: 7 to 12 months Show forest plot

2

259

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.75, 1.23]

Analysis 1.30

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30 Number of participants employed: 7 to 12 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30 Number of participants employed: 7 to 12 months.

Open in table viewer
Comparison 2. Subgroup analysis (relapse at 12 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Subgroup analysis: participants with a first episode Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

Analysis 2.1

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 1 Subgroup analysis: participants with a first episode.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 1 Subgroup analysis: participants with a first episode.

1.1 first episode

8

528

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.38, 0.58]

1.2 not first episode

19

2141

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.31, 0.49]

2 Subgroup analysis: participants in remission Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 2.2

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 2 Subgroup analysis: participants in remission.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 2 Subgroup analysis: participants in remission.

2.1 in remission

8

516

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.24, 0.61]

2.2 not in remission

16

2153

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

3 Subgroup analysis: various durations of stability before entering the study Show forest plot

18

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.3

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 3 Subgroup analysis: various durations of stability before entering the study.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 3 Subgroup analysis: various durations of stability before entering the study.

3.1 stable at least 1 month

5

428

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.15, 0.46]

3.2 stable at least 3 months

5

806

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.21, 0.44]

3.3 stable at least 6 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.69]

3.4 stable at least 12 months

5

326

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.57]

3.5 stable at least 3 to 6 years

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.18, 0.78]

4 Subgroup analysis: abrupt withdrawal versus tapering Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 4 Subgroup analysis: abrupt withdrawal versus tapering.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 4 Subgroup analysis: abrupt withdrawal versus tapering.

4.1 Abrupt withdrawal

16

1946

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.34, 0.54]

4.2 Taper

8

723

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.23, 0.50]

5 Subgroup analysis: single antipsychotic drugs Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 5 Subgroup analysis: single antipsychotic drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 5 Subgroup analysis: single antipsychotic drugs.

5.1 Chlorpromazine

2

406

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.36, 0.55]

5.2 Fluphenazine depot

6

296

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.14, 0.39]

5.3 Haloperidol depot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.04, 0.55]

5.4 Quetiapine

1

178

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.34, 0.69]

5.5 Paliperidone

2

617

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.30, 0.45]

5.6 Various, mixed groups of antipsychotic drugs

10

705

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.27, 0.65]

5.7 Ziprasidone

2

424

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.43, 0.64]

6 Subgroup analysis: depot versus oral drugs Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 6 Subgroup analysis: depot versus oral drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 6 Subgroup analysis: depot versus oral drugs.

6.1 depot

7

663

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.23, 0.41]

6.2 oral

14

1785

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.37, 0.57]

7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.7

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs.

7.1 First‐generation antipsychotic drugs

18

1430

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

7.2 Second‐generation antipsychotic drugs

6

1239

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.53]

8 Subgroup analysis: appropriate versus unclear allocation concealment Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 2.8

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 8 Subgroup analysis: appropriate versus unclear allocation concealment.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 8 Subgroup analysis: appropriate versus unclear allocation concealment.

8.1 appropriate allocation concealment

9

1410

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.33, 0.52]

8.2 unclear allocation concealment

15

1259

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.28, 0.53]

9 Subgroup analysis: blinded versus open trials Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 2.9

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 9 Subgroup analysis: blinded versus open trials.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 9 Subgroup analysis: blinded versus open trials.

9.1 blinded trials

22

2412

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.35, 0.51]

9.2 unblinded trials

2

257

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.17, 0.39]

Open in table viewer
Comparison 3. Sensitivity analysis (relapse at 12 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exclusion of studies that were not explicitly described as randomised Show forest plot

23

2654

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 3.1

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 1 Exclusion of studies that were not explicitly described as randomised.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 1 Exclusion of studies that were not explicitly described as randomised.

2 Exclusion of non‐double‐blind studies Show forest plot

22

2412

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.35, 0.51]

Analysis 3.2

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 2 Exclusion of non‐double‐blind studies.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 2 Exclusion of non‐double‐blind studies.

3 Fixed‐effects model Show forest plot

24

2669

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.36, 0.44]

Analysis 3.3

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 3 Fixed‐effects model.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 3 Fixed‐effects model.

4 Original authors' assumptions on dropouts Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

Analysis 3.4

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 4 Original authors' assumptions on dropouts.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 4 Original authors' assumptions on dropouts.

5 Inclusion of only large studies (> 200 participants) Show forest plot

5

1506

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.48]

Analysis 3.5

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 5 Inclusion of only large studies (> 200 participants).

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 5 Inclusion of only large studies (> 200 participants).

6 Exclusion of studies with clinical diagnosis Show forest plot

16

2325

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.36, 0.49]

Analysis 3.6

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 6 Exclusion of studies with clinical diagnosis.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 6 Exclusion of studies with clinical diagnosis.

7 Three months stable Show forest plot

20

2942

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.30, 0.55]

Analysis 3.7

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 7 Three months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 7 Three months stable.

8 Six months stable Show forest plot

13

1382

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.26, 0.61]

Analysis 3.8

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 8 Six months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 8 Six months stable.

9 Nine months stable Show forest plot

10

831

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.29, 0.73]

Analysis 3.9

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 9 Nine months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 9 Nine months stable.

10 Exclusion of studies with unclear randomisation method Show forest plot

8

1546

Risk Ratio (IV, Random, 95% CI)

0.41 [0.34, 0.50]

Analysis 3.10

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 10 Exclusion of studies with unclear randomisation method.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 10 Exclusion of studies with unclear randomisation method.

11 Exclusion of studies with unclear allocation concealment method Show forest plot

9

1410

Risk Ratio (IV, Random, 95% CI)

0.41 [0.33, 0.52]

Analysis 3.11

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 11 Exclusion of studies with unclear allocation concealment method.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 11 Exclusion of studies with unclear allocation concealment method.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

original image
Figures and Tables -
Figure 2

original image
Figures and Tables -
Figure 3

Summary of pooled results
Figures and Tables -
Figure 4

Summary of pooled results

Funnel plot of comparison: 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, outcome: 1.1 Relapse.
Figures and Tables -
Figure 5

Funnel plot of comparison: 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, outcome: 1.1 Relapse.

Summary of subgroup analysis
Figures and Tables -
Figure 6

Summary of subgroup analysis

Meta‐regression study duration
Figures and Tables -
Figure 7

Meta‐regression study duration

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1 Relapse: up to 3 months.
Figures and Tables -
Analysis 1.1

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1 Relapse: up to 3 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2 Relapse: 4 to 6 months.
Figures and Tables -
Analysis 1.2

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2 Relapse: 4 to 6 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3 Relapse: 7 to 12 months.
Figures and Tables -
Analysis 1.3

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3 Relapse: 7 to 12 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4 Relapse: > 12 months.
Figures and Tables -
Analysis 1.4

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4 Relapse: > 12 months.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5 Relapse: independent of duration.
Figures and Tables -
Analysis 1.5

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5 Relapse: independent of duration.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6 Leaving the study early: due to any reason.
Figures and Tables -
Analysis 1.6

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6 Leaving the study early: due to any reason.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7 Leaving the study early: due to adverse events.
Figures and Tables -
Analysis 1.7

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7 Leaving the study early: due to adverse events.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8 Leaving the study early: due to inefficacy.
Figures and Tables -
Analysis 1.8

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8 Leaving the study early: due to inefficacy.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9 Global state: number of participants improved.
Figures and Tables -
Analysis 1.9

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9 Global state: number of participants improved.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10 Service use: number of participants hospitalised.
Figures and Tables -
Analysis 1.10

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10 Service use: number of participants hospitalised.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11 Service use: number of participants discharged.
Figures and Tables -
Analysis 1.11

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11 Service use: number of participants discharged.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12 Death: any.
Figures and Tables -
Analysis 1.12

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12 Death: any.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13 Death: due to natural causes.
Figures and Tables -
Analysis 1.13

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13 Death: due to natural causes.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14 Suicide.
Figures and Tables -
Analysis 1.14

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14 Suicide.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15 Suicide attempts.
Figures and Tables -
Analysis 1.15

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15 Suicide attempts.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16 Suicide ideation.
Figures and Tables -
Analysis 1.16

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16 Suicide ideation.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17 Violent/aggressive behaviour.
Figures and Tables -
Analysis 1.17

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17 Violent/aggressive behaviour.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18 Adverse effects: at least one adverse event.
Figures and Tables -
Analysis 1.18

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18 Adverse effects: at least one adverse event.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19 Adverse effects: movement disorders: at least one movement disorder.
Figures and Tables -
Analysis 1.19

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19 Adverse effects: movement disorders: at least one movement disorder.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20 Adverse effects: movement disorders: akathisia.
Figures and Tables -
Analysis 1.20

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20 Adverse effects: movement disorders: akathisia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21 Adverse effects: movement disorders: akinesia.
Figures and Tables -
Analysis 1.21

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21 Adverse effects: movement disorders: akinesia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22 Adverse effects: movement disorders: dyskinesia.
Figures and Tables -
Analysis 1.22

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22 Adverse effects: movement disorders: dyskinesia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23 Adverse effects: movement disorders: dystonia.
Figures and Tables -
Analysis 1.23

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23 Adverse effects: movement disorders: dystonia.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24 Adverse effects: movement disorders: rigor.
Figures and Tables -
Analysis 1.24

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24 Adverse effects: movement disorders: rigor.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25 Adverse effects: movement disorders: tremor.
Figures and Tables -
Analysis 1.25

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25 Adverse effects: movement disorders: tremor.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26 Adverse effects: movement disorders: use of antiparkinson medication.
Figures and Tables -
Analysis 1.26

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26 Adverse effects: movement disorders: use of antiparkinson medication.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27 Adverse effects: sedation.
Figures and Tables -
Analysis 1.27

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27 Adverse effects: sedation.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28 Adverse effects: weight gain.
Figures and Tables -
Analysis 1.28

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28 Adverse effects: weight gain.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29 Quality of life.
Figures and Tables -
Analysis 1.29

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29 Quality of life.

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30 Number of participants employed: 7 to 12 months.
Figures and Tables -
Analysis 1.30

Comparison 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30 Number of participants employed: 7 to 12 months.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 1 Subgroup analysis: participants with a first episode.
Figures and Tables -
Analysis 2.1

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 1 Subgroup analysis: participants with a first episode.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 2 Subgroup analysis: participants in remission.
Figures and Tables -
Analysis 2.2

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 2 Subgroup analysis: participants in remission.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 3 Subgroup analysis: various durations of stability before entering the study.
Figures and Tables -
Analysis 2.3

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 3 Subgroup analysis: various durations of stability before entering the study.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 4 Subgroup analysis: abrupt withdrawal versus tapering.
Figures and Tables -
Analysis 2.4

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 4 Subgroup analysis: abrupt withdrawal versus tapering.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 5 Subgroup analysis: single antipsychotic drugs.
Figures and Tables -
Analysis 2.5

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 5 Subgroup analysis: single antipsychotic drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 6 Subgroup analysis: depot versus oral drugs.
Figures and Tables -
Analysis 2.6

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 6 Subgroup analysis: depot versus oral drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs.
Figures and Tables -
Analysis 2.7

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 8 Subgroup analysis: appropriate versus unclear allocation concealment.
Figures and Tables -
Analysis 2.8

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 8 Subgroup analysis: appropriate versus unclear allocation concealment.

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 9 Subgroup analysis: blinded versus open trials.
Figures and Tables -
Analysis 2.9

Comparison 2 Subgroup analysis (relapse at 12 months), Outcome 9 Subgroup analysis: blinded versus open trials.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 1 Exclusion of studies that were not explicitly described as randomised.
Figures and Tables -
Analysis 3.1

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 1 Exclusion of studies that were not explicitly described as randomised.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 2 Exclusion of non‐double‐blind studies.
Figures and Tables -
Analysis 3.2

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 2 Exclusion of non‐double‐blind studies.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 3 Fixed‐effects model.
Figures and Tables -
Analysis 3.3

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 3 Fixed‐effects model.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 4 Original authors' assumptions on dropouts.
Figures and Tables -
Analysis 3.4

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 4 Original authors' assumptions on dropouts.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 5 Inclusion of only large studies (> 200 participants).
Figures and Tables -
Analysis 3.5

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 5 Inclusion of only large studies (> 200 participants).

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 6 Exclusion of studies with clinical diagnosis.
Figures and Tables -
Analysis 3.6

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 6 Exclusion of studies with clinical diagnosis.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 7 Three months stable.
Figures and Tables -
Analysis 3.7

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 7 Three months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 8 Six months stable.
Figures and Tables -
Analysis 3.8

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 8 Six months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 9 Nine months stable.
Figures and Tables -
Analysis 3.9

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 9 Nine months stable.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 10 Exclusion of studies with unclear randomisation method.
Figures and Tables -
Analysis 3.10

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 10 Exclusion of studies with unclear randomisation method.

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 11 Exclusion of studies with unclear allocation concealment method.
Figures and Tables -
Analysis 3.11

Comparison 3 Sensitivity analysis (relapse at 12 months), Outcome 11 Exclusion of studies with unclear allocation concealment method.

Table 1. Design of a future study

Methods

Allocation: randomised ‐ clearly described generation of sequence and concealment of allocation.
Blinding: double ‐ described and tested.
Duration: 3 years.

Participants

People with schizophrenia or schizophrenia like disorder in remission for at least one month.
N=500.
Age: any.
Sex: both.
History: any.

Interventions

1. Any antipsychotic drug (flexible dose within appropriate range).

2. Placebo (after gradual withdrawal of the previous antipsychotic drug).

Outcomes

Relapse (primary outcome)

Rehospitalisation for psychosis

Time ill

Global state (number of participants improved)

Leaving the study early (including specific causes)

Death (natural and unnatural causes)

Violence

Quality of life

Satisfaction with care

Side‐effects

Employment and other measures of functioning

Figures and Tables -
Table 1. Design of a future study
Summary of findings for the main comparison. Maintenance treatment with antipsychotic drugs versus placebo/no treatment for schizophrenia

Maintenance treatment with antipsychotic drugs versus placebo/no treatment for schizophrenia

Patient or population: patients with schizophrenia
Settings: Inpatients and Outpatients
Intervention: Maintenance treatment with antipsychotic drugs versus placebo/no treatment

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

Maintenance treatment with antipsychotic drugs versus placebo/no treatment

Relapse: 7 to 12 months
Follow‐up: 7‐12 months

642 per 1000

263 per 1000
(218 to 315)

RR 0.41
(0.34 to 0.49)

2669
(24 studies)

⊕⊕⊕⊕
high1,2,3,4

Leaving the study early: due to any reason
Follow‐up: 1‐36 months

544 per 1000

288 per 1000
(250 to 332)

RR 0.53
(0.46 to 0.61)

4718
(47 studies)

⊕⊕⊕⊕
high1,2,5

Service use: Number of participants hospitalised
Follow‐up: 1‐24 months

256 per 1000

97 per 1000
(69 to 141)

RR 0.38
(0.27 to 0.55)

2090
(16 studies)

⊕⊕⊕⊕
high1,5,6

Suicide
Follow‐up: 3‐12 months

2 per 1000

1 per 1000
(0 to 7)

RR 0.34
(0.04 to 3.28)

1941
(8 studies)

⊕⊕⊝⊝
low5,7,8

Quality of life
Follow‐up: 7‐18 months

The mean quality of life in the intervention groups was
0.62 standard deviations lower
(1.15 to 0.09 lower)

527
(3 studies)

⊕⊕⊝⊝
low2,5,9,10,11

SMD ‐0.62 (‐1.15 to ‐0.09)

Number of participants employed: 7 to 12 months

504 per 1000

484 per 1000
(378 to 620)

RR 0.96
(0.75 to 1.23)

259
(2 studies)

⊕⊕⊝⊝
low5,12,13,14

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: Confidence interval; RR: Risk ratio;

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1 Risk of bias: rated 'no' ‐ many studies did not report the methods for sequence generation and/or allocation concealment. However, the ones that did, showed a similar effect size in subgroup and sensitivity analyses.
2 Inconsistency: rated 'no' ‐ although the p value for heterogeneity was statistically significant and the I‐square higher than 50%, the direction of the effect of almost all studies was the same. Therefore, this inconsistency does not challenge the overall results.
3 Publication bias: rated 'undetected' ‐ although the funnel plot was asymmetrical, the trim and fill test did not change the point estimate and the point estimate was also similar when only large studies were included.
4 Large effect: statistically significant RR that was lower than 0.5 (the actual value was 0.41).
5 Publication bias: The possibility of publication bias was only examined concerning the primary outcome (relapse at 12 months).
6 Large effect: statistically significant RR that was lower than 0.50 (the actual value was 0.38).
7 Almost all studies contributing to this outcome used adequate randomisation and allocation methods.
8 Imprecision: rated 'very serious' ‐ only few studies contribute data to this rare event an the CI was quite wide.
9 Risk of bias: rated 'no' ‐ the two large studies (out of three) that drove the effect used appropriate randomisation and allocation methods.
10 Indirectness: rated 'serious' ‐ the rating scales in the studies have been criticized for eventually not measuring what people understand by quality of life.
11 Imprecise data ‐ only a few studies contributed to this rare event and the confidence interval was large.
12 Risk of bias: rated 'no' ‐ the two studies used appropriate randomisation and allocation methods.
13 Indirectness: rated 'serious' ‐ the only two studies included mixed groups of employed and non‐employed participants at baseline, and it is unclear whether employment was supported or competitive employment.
14 Imprecision: rated 'serious' ‐ only two studies contributed to this event which is difficult to measure because it depends on various factors (e.g. the existence of supported employment, rural versus service economy etc).

Figures and Tables -
Summary of findings for the main comparison. Maintenance treatment with antipsychotic drugs versus placebo/no treatment for schizophrenia
Comparison 1. Maintenance treatment with antipsychotic drugs versus placebo/no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse: up to 3 months Show forest plot

34

3942

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.24, 0.38]

2 Relapse: 4 to 6 months Show forest plot

40

5285

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.30, 0.42]

3 Relapse: 7 to 12 months Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

4 Relapse: > 12 months Show forest plot

6

811

Risk Ratio (M‐H, Random, 95% CI)

0.59 [0.42, 0.82]

5 Relapse: independent of duration Show forest plot

62

6392

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.29, 0.41]

6 Leaving the study early: due to any reason Show forest plot

47

4718

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.46, 0.61]

6.1 up to 3 months

8

245

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.07, 0.72]

6.2 4 to 6 months

17

1646

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.35, 0.66]

6.3 7 to 12 months

18

2420

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.46, 0.66]

6.4 > 12 months

4

407

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.36, 1.26]

7 Leaving the study early: due to adverse events Show forest plot

43

4333

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.70, 1.91]

7.1 up to 3 months

8

245

Risk Ratio (M‐H, Random, 95% CI)

2.84 [0.12, 65.34]

7.2 4 to 6 months

14

1549

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.57, 1.74]

7.3 7 to 12 months

17

2339

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.59, 2.60]

7.4 > 12 months

4

200

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

8 Leaving the study early: due to inefficacy Show forest plot

46

4546

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

8.1 up to 3 months

9

295

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.07, 0.79]

8.2 4 to 6 months

16

1661

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.31, 0.54]

8.3 7 to 12 months

18

2420

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.28, 0.45]

8.4 > 12 months

3

170

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.08, 0.95]

9 Global state: number of participants improved Show forest plot

14

1524

Risk Ratio (M‐H, Random, 95% CI)

2.34 [1.68, 3.26]

9.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

4.61 [1.22, 17.40]

9.2 4 to 6 months

8

1037

Risk Ratio (M‐H, Random, 95% CI)

2.33 [1.69, 3.21]

9.3 7 to 12 months

5

438

Risk Ratio (M‐H, Random, 95% CI)

1.95 [0.91, 4.18]

10 Service use: number of participants hospitalised Show forest plot

16

2090

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.27, 0.55]

10.1 up to 3 months

2

55

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.04, 4.06]

10.2 4 to 6 months

3

109

Risk Ratio (M‐H, Random, 95% CI)

0.08 [0.01, 0.42]

10.3 7 to 12 months

8

1295

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.18, 0.57]

10.4 > 12 months

3

631

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.44, 0.70]

11 Service use: number of participants discharged Show forest plot

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

11.1 4 to 6 months

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

12 Death: any Show forest plot

14

2356

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.28, 2.11]

12.1 up to 3 months

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

12.2 4 to 6 months

5

856

Risk Ratio (M‐H, Random, 95% CI)

2.18 [0.48, 9.81]

12.3 7 to 12 months

8

1464

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.08, 1.27]

13 Death: due to natural causes Show forest plot

14

2401

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.39, 3.97]

13.1 4 to 6 months

5

856

Risk Ratio (M‐H, Random, 95% CI)

2.18 [0.48, 9.81]

13.2 7 to 12 months

9

1545

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.09, 3.36]

14 Suicide Show forest plot

8

1941

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.04, 3.28]

14.1 up to 3 months

1

36

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.2 4 to 6 months

2

730

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14.3 7 to 12 months

5

1175

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.04, 3.28]

15 Suicide attempts Show forest plot

5

1177

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.10, 2.33]

15.1 4 to 6 months

2

466

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 71.51]

15.2 7 to 12 months

3

711

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.04, 1.61]

16 Suicide ideation Show forest plot

3

556

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.04, 10.56]

16.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 3.88]

16.2 4 to 6 months

1

386

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

16.3 7 to 12 months

1

121

Risk Ratio (M‐H, Random, 95% CI)

2.77 [0.11, 66.57]

17 Violent/aggressive behaviour Show forest plot

5

680

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.15, 0.52]

17.1 up to 3 months

1

26

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.50]

17.2 4 to 6 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.2 [0.01, 3.92]

17.3 7 to 12 months

3

614

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.14, 0.53]

18 Adverse effects: at least one adverse event Show forest plot

10

2184

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.87, 1.18]

18.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.30, 0.93]

18.2 4 to 6 months

3

776

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.15]

18.3 7 to 12 months

6

1359

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.88, 1.38]

19 Adverse effects: movement disorders: at least one movement disorder Show forest plot

22

3411

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.25, 1.93]

19.1 up to 3 months

4

158

Risk Ratio (M‐H, Random, 95% CI)

2.42 [0.70, 8.33]

19.2 4 to 6 months

8

1658

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.06, 1.99]

19.3 7 to 12 months

10

1595

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.11, 2.07]

20 Adverse effects: movement disorders: akathisia Show forest plot

12

2026

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.87, 3.51]

20.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.94 [0.24, 15.97]

20.2 4 to 6 months

6

1009

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.41, 6.80]

20.3 7 to 12 months

5

968

Risk Ratio (M‐H, Random, 95% CI)

1.74 [0.88, 3.45]

21 Adverse effects: movement disorders: akinesia Show forest plot

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

21.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

22 Adverse effects: movement disorders: dyskinesia Show forest plot

13

1820

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.28, 0.97]

22.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.5 [0.06, 34.91]

22.2 4 to 6 months

3

418

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.84]

22.3 7 to 12 months

9

1353

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.30, 1.58]

23 Adverse effects: movement disorders: dystonia Show forest plot

6

824

Risk Ratio (M‐H, Random, 95% CI)

1.89 [1.05, 3.41]

23.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

2.5 [0.13, 49.22]

23.2 4 to 6 months

2

382

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.29]

23.3 7 to 12 months

3

393

Risk Ratio (M‐H, Random, 95% CI)

3.97 [0.44, 35.54]

24 Adverse effects: movement disorders: rigor Show forest plot

5

249

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.54, 2.88]

24.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.24, 2.22]

24.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

1.98 [0.67, 5.85]

24.3 7 to 12 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25 Adverse effects: movement disorders: tremor Show forest plot

10

1468

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.81, 1.93]

25.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.40, 3.01]

25.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.33, 2.61]

25.3 7 to 12 months

6

1259

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.82, 2.43]

26 Adverse effects: movement disorders: use of antiparkinson medication Show forest plot

7

1317

Risk Ratio (M‐H, Random, 95% CI)

1.40 [1.03, 1.89]

26.1 4 to 6 months

3

841

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.90, 2.61]

26.2 7 to 12 months

4

476

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.86, 2.05]

27 Adverse effects: sedation Show forest plot

10

2146

Risk Ratio (M‐H, Random, 95% CI)

1.50 [1.22, 1.84]

27.1 4 to 6 months

6

1577

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.86, 2.07]

27.2 7 to 12 months

4

569

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.90, 3.31]

28 Adverse effects: weight gain Show forest plot

10

2321

Risk Ratio (M‐H, Random, 95% CI)

2.07 [1.31, 3.25]

28.1 4 to 6 months

3

736

Risk Ratio (M‐H, Random, 95% CI)

1.76 [0.92, 3.37]

28.2 7 to 12 months

7

1585

Risk Ratio (M‐H, Random, 95% CI)

2.57 [1.30, 5.07]

29 Quality of life Show forest plot

3

527

Std. Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.15, ‐0.09]

29.1 (7 to 12) months

2

509

Std. Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.26, 0.01]

29.2 (> 12 ) months

1

18

Std. Mean Difference (IV, Random, 95% CI)

‐0.61 [‐1.66, 0.45]

30 Number of participants employed: 7 to 12 months Show forest plot

2

259

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.75, 1.23]

Figures and Tables -
Comparison 1. Maintenance treatment with antipsychotic drugs versus placebo/no treatment
Comparison 2. Subgroup analysis (relapse at 12 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Subgroup analysis: participants with a first episode Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

1.1 first episode

8

528

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.38, 0.58]

1.2 not first episode

19

2141

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.31, 0.49]

2 Subgroup analysis: participants in remission Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

2.1 in remission

8

516

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.24, 0.61]

2.2 not in remission

16

2153

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

3 Subgroup analysis: various durations of stability before entering the study Show forest plot

18

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 stable at least 1 month

5

428

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.15, 0.46]

3.2 stable at least 3 months

5

806

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.21, 0.44]

3.3 stable at least 6 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.69]

3.4 stable at least 12 months

5

326

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.57]

3.5 stable at least 3 to 6 years

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.18, 0.78]

4 Subgroup analysis: abrupt withdrawal versus tapering Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Abrupt withdrawal

16

1946

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.34, 0.54]

4.2 Taper

8

723

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.23, 0.50]

5 Subgroup analysis: single antipsychotic drugs Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Chlorpromazine

2

406

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.36, 0.55]

5.2 Fluphenazine depot

6

296

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.14, 0.39]

5.3 Haloperidol depot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.04, 0.55]

5.4 Quetiapine

1

178

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.34, 0.69]

5.5 Paliperidone

2

617

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.30, 0.45]

5.6 Various, mixed groups of antipsychotic drugs

10

705

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.27, 0.65]

5.7 Ziprasidone

2

424

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.43, 0.64]

6 Subgroup analysis: depot versus oral drugs Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 depot

7

663

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.23, 0.41]

6.2 oral

14

1785

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.37, 0.57]

7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 First‐generation antipsychotic drugs

18

1430

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

7.2 Second‐generation antipsychotic drugs

6

1239

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.37, 0.53]

8 Subgroup analysis: appropriate versus unclear allocation concealment Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

8.1 appropriate allocation concealment

9

1410

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.33, 0.52]

8.2 unclear allocation concealment

15

1259

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.28, 0.53]

9 Subgroup analysis: blinded versus open trials Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

9.1 blinded trials

22

2412

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.35, 0.51]

9.2 unblinded trials

2

257

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.17, 0.39]

Figures and Tables -
Comparison 2. Subgroup analysis (relapse at 12 months)
Comparison 3. Sensitivity analysis (relapse at 12 months)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Exclusion of studies that were not explicitly described as randomised Show forest plot

23

2654

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

2 Exclusion of non‐double‐blind studies Show forest plot

22

2412

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.35, 0.51]

3 Fixed‐effects model Show forest plot

24

2669

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.36, 0.44]

4 Original authors' assumptions on dropouts Show forest plot

24

2669

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.49]

5 Inclusion of only large studies (> 200 participants) Show forest plot

5

1506

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.48]

6 Exclusion of studies with clinical diagnosis Show forest plot

16

2325

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.36, 0.49]

7 Three months stable Show forest plot

20

2942

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.30, 0.55]

8 Six months stable Show forest plot

13

1382

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.26, 0.61]

9 Nine months stable Show forest plot

10

831

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.29, 0.73]

10 Exclusion of studies with unclear randomisation method Show forest plot

8

1546

Risk Ratio (IV, Random, 95% CI)

0.41 [0.34, 0.50]

11 Exclusion of studies with unclear allocation concealment method Show forest plot

9

1410

Risk Ratio (IV, Random, 95% CI)

0.41 [0.33, 0.52]

Figures and Tables -
Comparison 3. Sensitivity analysis (relapse at 12 months)