Scolaris Content Display Scolaris Content Display

Maintenance treatment with antipsychotic drugs for schizophrenia

Collapse all Expand all

References

Aripiprazole 2003 {published data only}6164

Baker RA, Meyer JM, Kaplita S, Forbes A, Grossman F, Pikalov A, Marcus RN. Long-term effects of aripiprazole on the lipid profiles of patients with schizophrenia in a 26-week placebo-controlled trial. Schizophrenia Bulletin 2007;33(2):S419. CENTRAL [CSZG: 15132]
Carson W, McQuade R, Saha A, Torbeyns A, Stock E. Aripiprazole versus placebo for relapse prevention in patients with chronic schizophrenia. European Neuropsychopharmacology 2002;12(Suppl 3):S288. CENTRAL [CSZG: 9067]
Carson W, Pigott T, Saha A, Ali M, McQuade RD, Torbeyns AF, et al. Aripiprazole vs placebo in the treatment of stable, chronic schizophrenia. In: Proceedings of the 156th Annual Meeting of the American Psychiatric Association; 2003 May 17-22; San Francisco, California, USA. 2003. CENTRAL [CSZG: 9986]
Carson W, Weiden P, Waldeck R, Tafesse E, Cislo P, L'Italien G, Iwamoto T. Aripiprazole is not associated with increased diabetes risk: a long-term model. Schizophrenia Research 2004;67(Suppl 1):181. CENTRAL [CSZG: 13175]
Carson WH, Ingenito GG, McQuade RD, Stock EG, Iwamoto T. Schizophrenia: safety/tolerability of aripiprazole. In: Proceedings of the 12th World Congress of Psychiatry; 2002 Aug 24-29; Yokohama, Japan. 2002. CENTRAL [CSZG: 19426]
Carson WH, Pigott TA, Saha AR, Ali MW, McQuade RD, Torbeyns AF, et al. Aripiprazole vs placebo in the treatment of chronic schizophrenia. International Journal of Neuropsychopharmacology 2002;5(Suppl 1):S187. CENTRAL [CSZG: 8326]
Casey D, Saha A, Marcus R, Carson WH, McQuade RD, Torbeyns AF, et al. Aripiprazole versus placebo for relapse prevention in patients with chronic schizophrenia. Schizophrenia Research 2003;60:276. CENTRAL [CSZG: 9689]
Casey D, Stock E, Jody D, Kaplita S, Saha A, Liston H, et al. Long-term treatment with aripiprazole: effects on plasma lipis levels and glycaemic control. Int J Neuropsychopharmacol 2004 Jun;7(Suppl 2):S417. CENTRAL [CSZG: 13177]
Dubitsky GM, Harris R, Laughren T, Hardeman S. Abilify (aripiprazole) tablets; medical review part 1. http://www.fda.gov/cder/foi/nda/2002/21-436_Abilify.htm2002:1-50. CENTRAL [CSZG: 9980]
Dubitsky GM, Harris R, Laughren T, Hardeman S. Abilify (aripiprazole) tablets; medical review part 2. http://www.fda.gov/cder/foi/nda/2002/21-436_Abilify.htm2002:50-110. CENTRAL [CSZG: 9981]
Dubitsky GM, Harris R, Laughren T, Hardeman S. Abilify (aripiprazole) tablets; medical review part 3. http://www.fda.gov/cder/foi/nda/2002/21-436_Abilify.htm2002:111-75. CENTRAL [CSZG: 9982]
Dubitsky GM, Harris R, Laughren T, Hardeman S. Abilify (aripiprazole) tablets; medical review part 4. http://www.fda.gov/cder/foi/nda/2002/21-436_Abilify.htm2002:176-232. CENTRAL [CSZG: 9983]
Marder SR, Kaplita S, Saha AR, Carson WH Jr, Torbeyns A, Stock EG. Glycemic control and plasma lipids in long-term aripiprazole treatment. In: Proceedings of the 156th Annual Meeting of the American Psychiatric Association; 2003 May 17-22; San Francisco, California, USA. 2003. CENTRAL [CSZG: 9799]
McQuade R, Kostic D, Marcus R, Carson W, Torbeyns A, Kerselaers W, Yocca F. Aripiprazole versus olanzapine in a 52-week, open-label extension study. Schizophrenia Bulletin 2005;31:S496-7. CENTRAL [CSZG: 11624]
Mortimer AM. Aripiprazole is effective for relapse prevention in people with chronic stable schizophrenia. Evidence-Based Mental Health 2004;7(2):41. CENTRAL [CSZG: 13962]
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. CENTRAL [CSZG: 9896]
Pigott TA, Saha AR, Ali MW, McQuade RD, Torbeyns AF, William HC, et al. Aripiprazole versus placebo in the treatment of chronic schizophrenia. In: Proceedings of the 155th Annual Meeting of the American Psychiatric Association; 2002 May 18-23; Philadelphia, Pennsylvania, USA. 2002. CENTRAL [CSZG: 9018]
Stock E, Marder SR, Jody D, Kaplita S, Saha A, Carson W, et al. Plasma lipids levels and glycemic control in long-term treatment with aripiprazole. European Neuropsychopharmacology 2003;13(4):S327. CENTRAL [CSZG: 9854]
Torbeyns A, Marder SR, Carson W, Jody D, Kaplita S, Saba A, et al. Glycemic control and plasma lipids in long-term treatment with aripiprazole. Schizophrenia Research 2004;67(1):192-3. CENTRAL [CSZG: 11339]

Aripiprazole 2017 {published data only}16575

Correll C, Kohegyi E, Zhao C, Baker RA, McQuade R, Salzman P, et al. Oral aripiprazole is an effective maintenance treatment in adolescents with schizophrenia. Neuropsychopharmacology2014:S349-S350. CENTRAL [CSZG: 29288]
Correll CU, Kohegyi E, Zhao C, Baker RA, McQuade R, Salzman PM, et al. Oral aripiprazole as maintenance treatment in adolescent schizophrenia: results from a 52-week, randomized, placebo-controlled withdrawal study. Journal of the American Academy of Child and Adolescent Psychiatry 2017;56(9):784-92. CENTRAL [CSZG: 36321]
NCT01149655. A long-term multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy, safety, and tolerability of aripiprazole (opc 14597) as maintenance treatment in adolescent patients with schizophrenia. http://ClinicalTrials.gov/show/NCT011496552010. CENTRAL [CSZG: 21085]

Aripiprazole depot 2012 {published data only}12553

Baker R, Fleischhacker W, Sanchez R, Perry P, Jin N, Johnson B, et al. Long-term safety and tolerability of once-monthly aripiprazole intramuscular depot for maintenance treatment in schizophrenia. International Journal of Neuropsychopharmacology 2012;15:111. CENTRAL [CSZG: 28035]
Baker RA, Carson WH, Perry PP, Sanchez R, Zhao J, McQuade RD, et al. Effects of aripiprazole once-monthly vs placebo on domains of personal and social performance in younger and older patients with schizophrenia. CNS Spectrums 2013;18(6):376-7. CENTRAL [CSZG: 28526]
Baker RA, Eramo A, Carson WH, Perry P, Sanchez R, Zhao J, et al. Effects of aripiprazole once-monthly vs. placebo on domains of personal and social performance in younger and older patients with schizophrenia. Schizophrenia Bulletin 2013;39(Suppl 1):S321-S322. CENTRAL [CSZG: 28131]
Baker RA, Eramo A, Nylander AG, Tsai LF, Peters-Strickland TS, Kostic D, et al. The effect of previous dose or oral aripiprazole (10 or 30 mg/day) on the efficacy and tolerability of aripiprazole once-monthly: Results from post-hoc analyses from two double-blind, randomized, controlled trials. Schizophrenia Research 2014;153:S293. CENTRAL [CSZG: 29386]
Carson WH, Perry PP, Sanchez R, Jin N, Forbes RA, McQuade RD, et al. Effects of ARI-IM-depot on secondary efficacy outcomes in maintenance treatment of schizophrenia. International Journal of Neuropsychopharmacology 2012;15:112-3. CENTRAL [CSZG: 28030]
Eramo A, Fleischhacker WW, Sanchez R, Perry P, Jin N, Johnson B, et al. A placebo-controlled study of efficacy and safety of aripiprazole oncemonthly for long-term maintenance treatment in schizophrenia. European Psychiatry 2013;28(1):1. CENTRAL [CSZG: 28187]
Eramo A, Fleischhacker WW, Sanchez R, Tsai LF, Peters-Strickland TS, Baker RA, et al. All-cause discontinuation and safety of aripiprazole once-monthly for the treatment of schizophrenia: A pooled analysis of two double-blind, randomized, controlled trials. Schizophrenia Research 2014;153:S174. CENTRAL [CSZG: 29437]
Fleischhacker WW, Baker RA, Eramo A, Sanchez R, Tsai LF, Peters-Strickland T, et al. Effects of aripiprazole once-monthly on domains of personal and social performance: results from 2 multicenter, randomized, double-blind studies. Schizophrenia Research 2014;159(2-3):415-20. CENTRAL [CSZG: 32163]
Fleischhacker WW, Perry P, Sanchez R, Jin N, Peters-Strickland T, Johnson B, et al. Functional outcomes with aripiprazole once-monthly in two double-blind, placebo- and active-controlled studies (aspire us 246 and aspire eu 247) for the treatment of schizophrenia. Value in Health 2013;16(7):A550. CENTRAL [CSZG: 28480]
Fleischhacker WW, Sanchez R, Johnson B, Jin N, Forbes RA, McQuade R, et al. Long-term safety and tolerability of aripiprazole once-monthly in maintenance treatment of patients with schizophrenia. International Clinical Psychopharmacology 2013;28:171-6. CENTRAL [CSZG: 28391]
Fleischhacker WW, Sanchez R, Lan-Feng T, Peters-Strickland T, Baker RA, Eramo A, et al. Safety and Effectiveness of Aripiprazole Oncemonthly for the Treatment of Schizophrenia: A Pooled Analysis of Two Double-blind, Randomized, Controlled Trials. Neuropsychopharmacology 2013;38:S394-S395. CENTRAL [CSZG: 28459]
Kane JM, Sanchez R, Baker RA, Eramo A, Peters-Strickland T, Perry PP, et al. Patient-centered outcomes with aripiprazole once-monthly for maintenance treatment in patients with schizophrenia: Results from two multicenter, randomized, double-blind studies. Clinical Schizophrenia and Related Psychoses 2015;9(2):79-87. CENTRAL [CSZG: 32183]
Kane JM, Sanchez R, Perry PP, Jin N, Johnson B, Forbes RA, et al. Efficacy of aripiprazole-IM-depot for long-term maintenance treatment of schizophrenia. International Journal of Neuropsychopharmacology 2012;15:S119. CENTRAL [CSZG: 28033]
Kane JM, Sanchez R, Perry PP, Jin N, Johnson BR, Forbes RA, et al. Aripiprazole intramuscular depot as maintenance treatment in patients with schizophrenia: A 52-week, multicenter, randomized, double-blind, placebo-controlled study. Journal of Clinical Psychiatry 2012;73(5):617-24. CENTRAL [CSZG: 24369]
Loze J, Carson WH, Perry PP, Sanchez R, Jin N, Forbes RA, et al. Psychosocial and overall effectiveness of aripiprazole intramuscular depot vs. placebo for maintenance treatment in schizophrenia. European Neuropsychopharmacology 2012;22:S328. CENTRAL [CSZG: 24886]
NCT00705783. Aripiprazole (OPC-14597) as maintenance treatment in patients with schizophrenia. https://www.ClinicalTrials.gov/ct/show/2008. CENTRAL [CSZG: 16546]
Sanchez R, Johnson B, Jin N, Forbes RA, Carson WH, McQuade RD, et al. Patient-reported outcomes (PROs) with aripiprazole intramuscular depot (ARI-IMD) for maintenance treatment in schizophrenia. International Journal of Neuropsychopharmacology 2012;15:133. CENTRAL [CSZG: 28040]
Sanchez R, Perry PP, Jin N, Johnson, RA Forbes, RD McQuade, WH, et al. A placebo-controlled study of efficacy/ safety of aripiprazole intramuscular depot for long-term maintenance treatment of schizophrenia. European Neuropsychopharmacology 2012;(Suppl 2):S327. CENTRAL [CSZG: 24893]

Asenapine 2011 {published data only}10424

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. CENTRAL [CSZG: 22873]
Kane JM, Mackle M, Snow-Adami L, Zhao J, Szegedi A, Panagides J. Double-blind, placebo-controlled trial of asenapine in prevention of relapse after long-term treatment of schizophrenia. International Journal of Neuropsychopharmacology 2010;13:223. CENTRAL [CSZG: 21634]
Mackle M, Snow-Adami L, Zhao J, Szegedi A, Panagides J. Double-blind, placebo-controlled trial of asenapine in prevention of relapse after long-term treatment of schizophrenia. European Neuropsychopharmacology 2009;19(Suppl 3):S543. CENTRAL [CSZG: 19377]
NCT00150176. A randomized, placebo-controlled, double-blind trial of asenapine in the prevention of relapse after long-term treatment of schizophrenia. https://www.ClinicalTrials.gov/ct/show/2005. CENTRAL [CSZG: 14708]
The NHSC . Asenapine (Saphris) for schizophrenia. Report2010. CENTRAL [CSZG: 21910]

Brexpiprazole 2017 {published data only}26950

Baker R, Hobart M, Forbes A, Ouyang J, Weiller E. Effect of brexpiprazole on long-term functioning in adults with schizophrenia. Australian and New Zealand Journal of Psychiatry 2017;51(Suppl 1):148. CENTRAL [CSZG: 35955]
Correll CU, Shi L, Weiss C, Hobart M, Eramo A, Duffy RA, et al. Successful switching of patients with acute schizophrenia from another antipsychotic to brexpiprazole: comparison of clinicians' choice of cross-titration schedules in a post-hoc analysis of a randomized double-blind maintenance treatment study. CNS Spectrums 2019;24(5):507-17. CENTRAL
Fleischhacker WW, Hobart M, Ouyang J, Forbes A, Pfister S, McQuade R, et al. Brexpiprazole (OPC-34712) efficacy and safety as maintenance therapy in adults with schizophrenia: Randomised, double-blind, placebo-controlled study. European Neuropsychopharmacology 2015;25:S527. CENTRAL [CSZG: 33322]
Fleischhacker WW, Hobart M, Ouyang J, Forbes A, Pfister S, McQuade RD, et al. Efficacy and sfety of Brexpiprazole (OPC-34712) as maintenance treatment in adults with schizophrenia: a randomized, double-blind, placebo-controlled study. International Journal of Neuropsychopharmacology 2017;20(1):11-21. CENTRAL [CSZG: 35475]
NCT01668797. Efficacy, Safety, and tolerability of Brexpiprazole (OPC-34712) as maintenance treatment in adults with schizophrenia. https://ClinicalTrials.gov/show/NCT016687972012. CENTRAL [CSZG: 35608]
Therrien F, Weiss C, Jin N, Baker RA, MacKenzie E, Meehan SR. Effect of brexpiprazole on patient functioning in patients with schizophrenia: results from a long-term, randomised, double-blind, placebo-controlled, maintenance study. Neuropsychopharmacology 2017;43((Suppl.1)):S244-S245. CENTRAL [CSZG: 37935]
Weiller E, Hobart M, Pfster S, Forbes A, Ouyang J, Weiss C. Effect of brexpiprazole on long-term remission in adults with schizophrenia: Results from a randomized, double-blind, placebo-controlled, maintenance study. Schizophrenia Bulletin 2017;43:S202-3. CENTRAL [CSZG: 36050]
Weiss C, Forbes A, Hobart M, Pfster S, Ouyang J, Weiller E. Short-term and long-term efficacy of brexpiprazole in adults with schizophrenia: Effect across marder factors. Schizophrenia Bulletin 2017;43:S155. CENTRAL [CSZG: 36051]
Weiss C, Ouyang J, Eramo A, Duffy RA, Weiller E, Baker RA. Switching patients with acute schizophrenia to brexpiprazole: post-hoc analysis of a double-blind randomized maintenance treatment study. Neuropsychopharmacology 2015;40:S227-8. CENTRAL [CSZG: 33450]
Weiss C, Weiller E, Hobart M, Ouyang J. Effect of brexpiprazole on weight and metabolic parameters: analysis of a maintenance trial in schizophrenia. European Neuropsychopharmacology 2016;26:S556. CENTRAL [CSZG: 35746]

Cariprazine 2016 {published data only}18421

Correll CU, Potkin SG, Durgam S, Chang CT, Szatmari B, Laszlovszky I, et al. Sustained remission with cariprazine treatment: post-hoc analysis of a randomised, double-blind, schizophrenia relapse prevention trial. Europan Neuropsychopharmacology 2017;27((Suppl. 4)):S935-S936. CENTRAL [CSZG: 37757]
Correll CU, Potkin SG, Zhong Y, Harsányi J, Szatmári B, Earley W. Long-term remission with cariprazine treatment in patients with schizophrenia: a post-hoc analysis of a randomized, double-blind, placebo.controlled relapse prevention trial. Journal of Clinical Psychiatry 2019;80(2):18m12495. CENTRAL
Durgam S, Earley W, Li R, Li D, Lu K, Laszlovszky I, et al. Corrigendum to "Long-term cariprazine treatment for the prevention of relapse in patients with schizophrenia: A randomized, double-blind, placebo-controlled trial" [Schizophr. Res. 176 (2016) 264-71]. Schizophrenia Research 2017;InPress:InPress. CENTRAL [CSZG: 35204]
Durgam S, Earley W, Li R, Li D, Lu K, Laszlovszky I, et al. Long-term cariprazine treatment for the prevention of relapse in patients with schizophrenia: A double-blind, placebo-controlled trial. European Neuropsychopharmacology 2015;25:S512-3. CENTRAL [CSZG: 33317]
Durgam S, Earley W, Li R, Li D, Lu K, Laszlovszky I, et al. Long-term cariprazine treatment for the prevention of relapse in patients with schizophrenia: A randomized, double-blind, placebo-controlled trial. Schizophrenia Research 2016;176(2-3):264-71. CENTRAL [CSZG: 35204]
Durgam S, Earley W, Li R, Li D, Lu K, Laszlovszky I, et al. Long-term cariprazine treatment for the prevention of relapse in patients with schizophrenia: additional analysis from a randomized, double-blind, placebo-controlled trial. Neuropsychopharmacology 2015;40:S380-1. CENTRAL [CSZG: 33316]
Earley W, Guo H, Luchini R. Modified cariprazine relapse prevention clinical trial results. Schizophrenia Research 2018;199:452-3. CENTRAL
Kunovac J, Kane J, Durgam S, Chang CT, Nemeth G, Laszlovszky I, et al. Characteristics associated with relapse in patients with schizophrenia: Post hoc analysis of a randomized, double-blind, placebo-controlled, cariprazine relapse prevention trial. Schizophrenia Bulletin 2017;43:S166-7. CENTRAL [CSZG: 36002]
NCT01412060. Cariprazine relative to placebo in the prevention of relapse of symptoms in patients with schizophrenia. http://ClinicalTrials.gov/show/NCT014120602011. CENTRAL [CSZG: 23106]
Potkin S, Correll C, Chang CT, Szatmari B, Laszlovszky I, Earley W. Long-term remission with cariprazine treatment in patients with schizophrenia: a post hoc analysis of a randomized, double-blind, placebo-controlled, relapse prevention trial. Schizophrenia Bulletin 2017;43:S118. CENTRAL [CSZG: 36026]

Chlorpromazine 1959 {published data only}2474

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. CENTRAL [CSZG: 2560]

Chlorpromazine 1962 {published data only}759

Freeman LS, Alson E. Prolonged withdrawal of chlorpromazine in chronic patients. Diseases of the Nervous System 1962;23:522-5. CENTRAL [CSZG: 809]

Chlorpromazine 1968 {published data only}3766

Crane GE. Tardive dyskinesia in schizophrenic patients treated with psychotropic drugs. Agressologie 1968;9(2):209-16. CENTRAL [CSZG: 3170]
De Long SL. Incidence and significance of chlorpromazine-induced eye changes. Diseases of the Nervous System 1968;29:19-22. CENTRAL [CSZG: 18229]
Gardos G, Cole JO, LaBrie RA. A 12-year follow-up study of chronic schizophrenics. Psychopharmacology 1982;33:983-4. CENTRAL [CSZG: 853]
Prien RF, Cole JO, Belkin NF. Relapse in chronic schizophrenics following abrupt withdrawal of tranquillizing medication. British Journal of Psychiatry 1968;115:679-86. CENTRAL [CSZG: 3951]
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. CENTRAL [CSZG: 3950]
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. CENTRAL [CSZG: 3952]
Prien RF, Levine J, Cole JO. Indications for high dose chlorpromazine therapy in chronic schizophrenia. Diseases of the Nervous System 1970;31:739-45. CENTRAL [CSZG: 2345]
Prien RF, Levine J, Switalski RW. Discontinuation of chemotherapy for chronic schizophrenics. Hospital and Community Psychiatry 1971;22(1):4-7. CENTRAL [CSZG: 2346]

Chlorpromazine 1973 {published data only}1800

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. CENTRAL [CSZG: 1873]
Hogarty G, Goldberg SC. Drug and social therapy in schizophrenic outpatients. Unknown Source1994:18. CENTRAL [CSZG: 5049]
Hogarty GE, Goldberg S, Schooler N. Proceedings: drug and sociotherapy in the aftercare of schizophrenic patients. Psychopharmacology Bulletin 1974;10(4):47. CENTRAL [CSZG: 4745]
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. CENTRAL [CSZG: 1876]
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. CENTRAL [CSZG: 1875]
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. CENTRAL [CSZG: 1874]
Hogarty GE, Munetz MR. Pharmacogenic depression among outpatient schizophrenic patients: a failure to substantiate. Journal of Clinical Psychopharmacology 1984;4:17-24. CENTRAL [CSZG: 1877]
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. CENTRAL [CSZG: 1277]
Hogarty GE, Ulrich RF. Temporal effects of drug and placebo in delaying relapse in schizophrenic outpatients. Archives of General Psychiatry 1977;34:297-301. CENTRAL [CSZG: 1878]
Hogarty GE. NIHM-PRB collaborative outpatient schizophrenic study. Psychopharmacology Bulletin 1974;10(2):54-5. CENTRAL [CSZG: 1276]

Chlorpromazine 1975 {published data only}1827

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. CENTRAL [CSZG: 1911]

Chlorpromazine 1976 {published data only}208

Andrews P, Hall JN, Snaith RP. A controlled trial of phenothiazine withdrawal in chronic schizophrenic patients. British Journal of Psychiatry 1976;128:451-5. CENTRAL [CSZG: 226]

Fluphenazine 1979 {published data only}2340

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. CENTRAL [CSZG: 2428]
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. CENTRAL [CSZG: 2424]
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. CENTRAL [CSZG: 3959]
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. CENTRAL [CSZG: 2426]
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. CENTRAL [CSZG: 3960]

Fluphenazine 1980 {published data only}3772

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. CENTRAL [CSZG: 3961]
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. CENTRAL [CSZG: 1683]
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. CENTRAL [CSZG: 3962]
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. CENTRAL [CSZG: 3964]
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. CENTRAL [CSZG: 3965]
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. CENTRAL [CSZG: 2521]
Schooler NR, Levine J. Dosage and side effect comparisons between oral and depot fluphenazine. Psychopharmacology Bulletin 1977;13:29-31. CENTRAL

Fluphenazine 1982 {published data only}1251

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. CENTRAL [CSZG: 1304]
Kane JM, Rifkin A, Woerner M, Reardon G. Low-dose neuroleptics in outpatient schizophrenics. Psychopharmacology Bulletin 1982;18:20-1. CENTRAL [CSZG: 1602]

Fluphenazine depot 1968 {published data only}1259

Keskiner A, Holden JMC, Itil TM. Maintenance treatment of schizophrenic outpatients with a depot phenothiazine. Psychosomatics 1968;9:166-71. CENTRAL [CSZG: 1312]
Keskiner A, Simeon J, Fink M, Itil TM. Long-acting phenothiazine (fluphenazine decanoate) treatment of psychosis. Archives of General Psychiatry 1968;18:477-81. CENTRAL
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. CENTRAL [CSZG: 1323]
Kinross-Wright J, Charalampous KD. A controlled study of a very long-acting phenothiazine preparation. International Journal of Neuropsychiatry 1965;1:66-70. CENTRAL [CSZG: 1335]

Fluphenazine depot 1973 {published data only}3723

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. CENTRAL [CSZG: 959]
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. CENTRAL [CSZG: 3799]

Fluphenazine depot 1979a {published data only}1444

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. CENTRAL [CSZG: 1514]

Fluphenazine depot 1979b {published data only}1233

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. CENTRAL [CSZG: 1286]
Kane JM. Low dose medication strategies in the maintenance treatment of schizophrenia. Schizophrenia Bulletin 1983;9(4):528-32. CENTRAL

Fluphenazine depot 1981 {published data only}4324

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. CENTRAL [CSZG: 5007]
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. CENTRAL [CSZG: 2577]

Fluphenazine depot 1982 {published data only}2098

Odejide OA, Aderounmu AF. Double-blind placebo substitution: withdrawal of fluphenazine decanoate in schizophrenic patients. Journal of Clinical Psychiatry 1982;43:195-6. CENTRAL [CSZG: 2184]

Fluphenazine depot 1992 {published data only}2407

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. CENTRAL [CSZG: 2493]
Soni SD, Mallik A, Schiff AA. Sulpiride in negative schizophrenia: a placebo-controlled double-blind assessment. Human Psychopharmacology 1990;5:233-8. CENTRAL [CSZG: 2697]

Haloperidol 1973 {published data only}2232

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. CENTRAL [CSZG: 2318]

Haloperidol 1991 {published data only}2385

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. CENTRAL [CSZG: 2471]

Haloperidol depot 1982 {published data only}3162

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. CENTRAL [CSZG: 3256]

Haloperidol depot 1991 {published data only}9291

Eklund K, Forsman A. Minimal effective dose and relapse - double-blind trial: haloperidol decanoate vs. placebo. Clinical Neuropharmacology 1991;14:S7-S15. CENTRAL [CSZG: 747]
Eklund K. Low dose of haloperidol decanoate is effective against relapses in schizophrenia patients. A double-blind placebo controlled study. In: Proceedings of the 17th Collegium Internationale Neuro-Psychopharmacologicum Congress; 1990 Sep 10-14; Kyoto, Japan. 1990:287. CENTRAL [CSZG: 12924]

Iloperidone 2016 {published data only}17965

CTRI/2010/091/001409. A clinical trial to study prevention of relapse in patients with schizophrenia reveiving either flexible dose iloperidone or placebo in long-term use.. http://ctri.nic.in/Clinicaltrials/login.php2010. CENTRAL
NCT01291511. Efficacy in prevention of relapse of schizophrenia in subjects taking either placebo or iloperidone. http://ClinicalTrials.gov/show/NCT012915112011. CENTRAL [CSZG: 22671]
Weiden PJ, Manning R, Wolfgang CD, Ryan JM, Mancione L, Han G, et al. A randomized trial of iloperidone for prevention of relapse in schizophrenia: the REPRIEVE study. CNS drugs 2016;30(8):735-47. CENTRAL [CSZG: 35243]

Lurasidone 2016 {published data only}18736

NCT01435928. PEARL Schizophrenia Maintenance. http://ClinicalTrials.gov/show/NCT014359282011. CENTRAL [CSZG: 23439]
Tandon R, Cucchiaro J, Phillips D, Hernandez D, Mao Y, Pikalov A, et al. A double-blind, placebo-controlled, randomized withdrawal study of lurasidone for the maintenance of efficacy in patients with schizophrenia. Journal of Psychopharmacology 2016;30(1):69-77. CENTRAL [CSZG: 33434]
Tandon R, Loebel A, Philips D, Pikalov A, Hernandez D Mao Y, et al. Lurasidone for maintenance of efficacy in patients with schizophrenia. European Neuropsychopharmacology 2014;24(Suppl 2):S559-560. CENTRAL [CSZG: 27840]
Tandon R, Loebel A, Philips D, Pikalov A, Hernandez D, Mao Y, Cucchiaro J. A double-blind, placebo-controlled, randomized withdrawal study of lurasidone for the maintenance of efficacy in patients with schizophrenia. Schizophrenia Research 2014;153(1):S372. CENTRAL [CSZG: 29681]
Tandon R, Loebel A, Philips D, Pikalov A, Hernandez D, Mao Y, et al. A double-blind, placebo-controlled, randomized withdrawal study of lurasidone for the maintenance of efficacy in patients with schizophrenia. Int J Neuropsychopharmacol 2014;17:S153-154. CENTRAL [CSZG: 29954]

Olanzapine 1999 {published data only}4622

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. CENTRAL [CSZG: 5651]
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. CENTRAL [CSZG: 5305]
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. In: Proceedings of the 11th European College of Neuropsychopharmacology Congress; 1998 Oct 31 - Nov 4; Paris, France. 1998. CENTRAL [CSZG: 4859]

Olanzapine 2003 {published data only}14624

Beasley C Jr, Hamilton S, Dossenbach M. Relapse prevention with olanzapine. European Neuropsychopharmacology 2000;10(Suppl 3):S304. CENTRAL [CSZG: 4620]
Beasley CM, Hamilton SH, Dossenbach M. Relapse prevention with olanzapine. Schizophrenia Research 2000;41(1):196-7. CENTRAL [CSZG: 4874]
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. CENTRAL [CSZG: 10010]
Beasley CM, Sutton VK, Taylor CC, Sethuraman G, Dossenbach M, Naber D, Pickar D. Does maintenance treatment with atypical antipsychotics worsen quality of life among stable patients with schizophrenia? Neuropsychopharmacology 2005;30(S1):S189. CENTRAL [CSZG: 13107]
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. CENTRAL [CSZG: 14454]
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. CENTRAL [CSZG: 9849]
EliLilly Company. Olanzapine relapse prevention versus placebo in the treatment of schizophrenia. http://www.Clinicalstudyresults.org/.2005. CENTRAL [CSZG: 18733]

Paliperidone 2007 {published data only}15435

Eerdekens. Evaluate the efficacy in the prevention of recurrence of the symptoms of schizophrenia. Sponsor: Johnson & Johnson Pharmaceutical Research & Development, LL.C2010. CENTRAL [CTG: NCT00111189]
Kramer M, Kushner S, Sherr J, Vijapurkar U, Lim P, Eerdekens M. Delaying symptom recurrence in patients with schizophrenia with paliperidone extended-release tablets: an international, randomized, double-blind, placebo-controlled study. Schizophrenia Bulletin 2007;33(2):438. CENTRAL [CSZG: 15185]
Kramer M, Kushner S, Vijapurkar U, Lim P, Eerdekens M. Delaying symptom recurrence in patients with schizophrenia treated with paliperidone extended-release tablets. European Neuropsychopharmacology 2006;16(Suppl 4):S386. CENTRAL [CSZG: 13313]
Kramer M, Kushner S, Vijapurkar U, Lim P, Eerdekens M. Delaying symptom recurrence in patients with schizophrenia with paliperidone extended-release tablets: an international, randomized, double-blind, placebo-controlled study. International Journal of Neuropsychopharmacology 2006;9(Suppl 1):S274. CENTRAL [CSZG: 19545]
Kramer M, Simpson G, Maciulis V, Kushner S, Liu Y, Lim P, et al. One-year open-label safety and efficacy study of paliperidone extended-release. CNS Spectrums 2010;15(8):506-14. CENTRAL [CSZG: 20885]
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. CENTRAL [CSZG: 13907]
Markowitz M, Fu DJ, Levitan B, Gopal S, Turkoz I, Alphs L. Long-acting injectable paliperidone palmitate versus oral paliperidone extended release. Annals of General Psychiatry 2013;12:22. CENTRAL [CSZG: 28633]
Wang D, Gopal S, Baker S, Narayan VA. Trajectories and changes in individual items of positive and negative syndrome scale among schizophrenia patients prior to impending relapse. NPJ Schizophrenia 2018;4(1):10. CENTRAL

Paliperidone 2014 {published data only}22628

Rui Q, Wang Y, Liang S, Liu Y, Wu Y, Wu Q, et al. Relapse prevention study of paliperidone extended-release tablets in Chinese patients with schizophrenia. Progress in Neuro-Psychopharmacology and Biological Psychiatry 2014;53:45-53. CENTRAL [CSZG: 29320]
Zhang H, Li H, Liu Y, Wu C, Wu Q, Nuamah I. Safety and efficacy of paliperidone extended-release in Chinese patients with schizophrenia. Neuropsychiatric Disease and Treatment 2016;12:69-77. CENTRAL [CSZG: 33455]

Paliperidone depot1M 2010 {published data only}11886

Alphs L, Bossie C, Kern-Sliwa J, Ma Y-W, Haskins JT. Paliperidone palmitate: clinical response in subjects with schizophrenia with recent vs. longer-term duration of illness. In: Proceedings of the 162nd Annual Meeting of the American Psychiatric Association. San Francisco, CA., 2009 May 16-21. CENTRAL [CSZG: 19067]
Emsley R, Nuamah I, Gopal S, Hough D, Fleischhacker WW. Relapse after antipsychotic discontinuation in schizophrenia as a withdrawal phenomenon vs illness recurrence: a post hoc analysis of a randomized placebo-controlled study. Journal of Clinical Psychiatry 2018 Jun;79(4):17m11874. CENTRAL [CSZG: 38184]
Emsley R, Nuamah I, Hough D, Gopal S. Treatment response after relapse in a placebo-controlled maintenance trial in schizophrenia. Schizophrenia Research 2012;138(1):29-34. CENTRAL [CSZG: 24214]
Gopal S, Vijapurkar U, Lim P, Morozova M, Eerdekens M, Hough D. A 52-week open-label study of the safety and tolerability of paliperidone palmitate in patients with schizophrenia. Journal of Psychopharmacology 2011;25(5):685-97. CENTRAL [CSZG: 22903]
Gopal S, Vijapurkar U, Lim P, Morozova M, Eerdekens M. Long-term efficacy, safety and tolerability of paliperidone palmitate in patients with schizophrenia. In: Proceedings of the 162nd Annual Meeting of the American Psychiatric Association. San Francisco, CA, 2009 May 16-21. CENTRAL [CSZG: 19076]
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. In: Proceedings of the 161st Annual Meeting of the American Psychiatric Association; 2008 May 3-8, Washington DC, USA. 2008. CENTRAL [CSZG: 16486]
Hough D, Gopal S, Vijapurkar U, Lim P, Morozova M, Eerdekens M. Paliperidone palmitate, an injectable antipsychotic, in prevention of symptom recurrence in patients with schizophrenia: A randomized, double-blind, placebo-controlled study. In: Proceedings of the 63rd Annual Convention of the Society of Biological Psychiatry. Washington, DC, USA: Elsevier Science Inc, 2008:S285-6. CENTRAL [CSZG: 20281]
Hough D, Gopal S, Vijapurkar U, Lim P, Morozova M, Eerdekens M. Paliperidone palmitate, an injectable antipsychotic, in prevention of symptom recurrence in patients with schizophrenia: a randomized, double-blind, placebo-controlled study. Biological Psychiatry 2008;63(7 Suppl):285S-6S. CENTRAL [CSZG: 19530]
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. CENTRAL [CSZG: 19704]
Hulihan J, Bossie CA, Fu DJ, Kern Sliwa J, Ma Y-W, Alphs LD. Remission with continued paliperidone palmitate treatment in stable subjects with schizophrenia. Schizophrenia Research 2012;136:S252. CENTRAL [CSZG: 29467]
Kozma CM, Slaton T, Dirani R, Fastenau J, Gopal S, Hough D. Changes in schizophrenia-related hospitalization and ER use among patients receiving paliperidone palmitate. Current Medical Resarch and Opinion 2011;27(8):1603-11. CENTRAL
Sliwa JK, Bossie CA, Fu DJ, Turkoz I, Alphs L. Long-term tolerability of once-monthly injectable paliperidone palmitate in subjects with recently diagnosed schizophrenia. Neuropsychiatric Disease and Treatment 2012;8:375-85. CENTRAL [CSZG: 28007]
Sliwa JK, Fu DJ, Bossie CA, et al. Body mass index and metabolic parameters in patients with schizophrenia during long-term treatment with paliperidone palmitate. BMC Psychiatry 2014;14:S52. CENTRAL [CSZG: 29148]
Wang D, Gopal S, Baker S, Narayan VA. Trajectories and changes in individual items of positive and negative syndrome scale among schizophrenia patients prior to impending relapse. NPJ Schizophrenia 2018;4(1):10. CENTRAL

Paliperidone depot1M 2015 {published data only}16601

Bossie C, Turkoz I, Alphs L, Fu DJ. Monotherapy with once monthly paliperidone palmitate for psychotic, depressive, and manic symptoms in schizoaffective disorder. Schizophrenia Bulletin 2015;41:S303. CENTRAL [CSZG: 29400]
Bossie CA, Turkoz I, Alphs L, Mahalchick L, Fu DJ. Paliperidone palmitate once-monthly treatment in recent onset and chronic illness patients with schizoaffective disorder. Journal of Nervous and Mental Ddisease 2017;205(4):324-8. CENTRAL [CSZG: 35417]
Fu DJ, Alphs L, Lindenmayer JP, Schooler N, Simonson RB, Turkoz I, et al. Paliperidone palmitate long-acting injectable delays psychotic and mood symptom relapse in schizoaffective disorder. European Neuropsychopharmacology 2014;24:S534-5. CENTRAL [CSZG: 27785]
Fu DJ, Bossie C, Turkoz I, Mahalchick L, Alphs L. Paliperidone palmitate treatment response in early and chronic illness schizoaffective disorder patients. Schizophrenia Bulletin 2015;41:S312. CENTRAL [CSZG: 29446]
Fu DJ, Turkoz I, Simonson RB, Walling D, Schooler N, Lindenmayer JP, et al. Effect of paliperidone palmitate once-monthly in improving and maintaining functioning in subjects with schizoaffective disorder using the domains of the personal and social performance scale. Neuropsychopharmacology 2014;39:S365. CENTRAL [CSZG: 29278]
Fu DJ, Turkoz I, Simonson RB, Walling D, Schooler N, Lindenmayer JP, et al. Paliperidone palmitate delays relapse in patients with schizoaffective disorder. Biological Psychiatry 2014;1:263S. CENTRAL [CSZG: 28974]
Fu DJ, Turkoz I, Simonson RB, Walling D, Schooler N, Lindenmayer JP, et al. Paliperidone palmitate long-acting injectable in acute exacerbation of schizoaffective disorder. Schizophrenia Bulletin 2013;39:S331. CENTRAL [CSZG: 28167]
Fu DJ, Turkoz I, Simonson RB, Walling D, Schooler N, Lindenmayer JP, et al. Paliperidone palmitate once-monthly injectable treatment for acute exacerbations of schizoaffective disorder. Journal of Clinical Psychopharmacology 2016;36(4):372-6. CENTRAL [CSZG: 35803]
Fu DJ, Turkoz I, Simonson RB, Walling DP, Schooler NR, Lindenmayer JP, et al. Paliperidone palmitate once-monthly reduces risk of relapse of psychotic, depressive, and manic symptoms and maintains functioning in a double-blind, randomized study of schizoaffective disorder. Journal of Clinical Psychiatry 2015;76(3):253-62. CENTRAL [CSZG: 29752]
Fu DJ, Turkoz I, Walling D, Lindenmayer JP, Schooler NR, Alphs L. Paliperidone palmitate once-monthly maintains improvement in functioning domains of the Personal and Social Performance scale compared with placebo in subjects with schizoaffective disorder. Schizophrenia Research 2018;192:185-93. CENTRAL [CSZG: 35804]
Joshi K, Lin J, Lingohr-Smith M, Fu D. Medical cost reductions for paliperidone palmitate versus placebo in randomized double-blind trial of patients with schizoaffective disorder. Journal of Managed Care and Specialty Pharmacy 2015;21:536-7. CENTRAL
Joshi K, Lin J, Lingohr-Smith M, Fu DJ. Estimated medical cost reductions for paliperidone palmitate vs placebo in a randomized, double-blind relapse-prevention trial of patients with schizoaffective disorder. Journal of Medical Economics 2015;18:629-36. CENTRAL [CSZG: 33677]
NCT01193153. A study to evaluate the efficacy of paliperidone palmitate in the prevention of relapse of the symptoms of schizoaffective disorder. http://ClinicalTrials.gov/show/NCT011931532010. CENTRAL [CSZG: 21094]
Turkoz I, Fu DJ, Walling D, Schooler NR, Lindenmayer JP, Simonson RB, et al. Effect of paliperidone palmitate once-monthly in improving and maintaining functioning in subjects with schizoaffective disorder using the domains of the personal and social performance scale. Schizophrenia Bulletin 2015;41:S193-4. CENTRAL [CSZG: 29687]

Paliperidone depot3M 2015 {published data only}18982

Bell LKS, Turkoz I, Kim E. Paliperidone palmitate once-every-3-months in adults with early illness schizophrenia. Early Intervention in Psychiatry 2019;13(3):667-72. CENTRAL
Berwaerts J, Liu Y, Gopal S, Nuamah I, Xu H, Savitz A, et al. Efficacy and safety of paliperidone palmitate 3 month formulation: a randomized, double-blind, placebo-controlled study. Schizophrenia Bulletin 2015;41:S302. CENTRAL [CSZG: 29393]
Berwaerts J, Liu Y, Gopal S, Nuamah I, Xu H, Savitz A, et al. Efficacy and safety of paliperidone palmitate 3-month formulation in schizophrenia: a randomized, double-blind, placebo-controlled study. European Psychiatry 2015;30:272. CENTRAL [CSZG: 32149]
Berwaerts J, Liu Y, Gopal S, Nuamah I, Xu H, Savitz A, et al. Efficacy and safety of the 3-month formulation of paliperidone palmitate vs placebo for relapse prevention of schizophrenia: a randomized clinical trial. JAMA Psychiatry 2015;72(8):830-9. CENTRAL [CSZG: 33027]
Chirila C, Zheng Q, Sawyerr G, Nuamah I. Healthcare resource use of paliperidone palmitate 3-month injection vs placebo. In: 168th Annual Meeting of American Psychiatric Association; Toronto, Canada. May 16-20, 2015. CENTRAL [CSZG: 33035]
Gopal S, Furey M, Hibar D, Kolb H, Saad Z, Savitz A, et al. Potential biomarkers of impending schizophrenia relapse. Biological Psychiatry 2017;(10 Suppl):S126-S127. CENTRAL [CSZG: 36107]
Gopal S, Xu H, McQuarrie K, Savitz A, Nuamah I, Woodruff K, et al. Caregiver burden in schizophrenia following paliperidone palmitate long acting injectables treatment: pooled analysis of two double-blind randomized phase three studies. NPJ Schizophrenia 2017;3(1):23. CENTRAL [CSZG: 36325]
Mathews M, Nuamah I, Savitz A, Gopal S. Efficacy and safety of paliperidone palmitate 3-month formulation vs placebo for relapse prevention of Schizophrenia: Treatment effect using a number needed to treat analysis. Biological Psychiatry 2017;81(10 Suppl 1):S350-1. CENTRAL [CSZG: 36437]
NCT01529515. A study of paliperidone palmitate 3 month formulation for the treatment of patients with schizophrenia. http://ClinicalTrials.gov/show/NCT015295152012. CENTRAL [CSZG: 23705]
Wang D, Gopal S, Baker S, Narayan VA. Trajectories and changes in individual items of positive and negative syndrome scale among schizophrenia patients prior to impending relapse. NPJ Schizophrenia 2018;4(1):10. CENTRAL

Penfluridol 1970 {published data only}279

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. CENTRAL [CSZG: 297]

Penfluridol 1974a {published data only}799

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. CENTRAL [CSZG: 849]

Penfluridol 1974b {published data only}2354

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. CENTRAL [CSZG: 2440]

Penfluridol 1974c {published data only}2833

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. CENTRAL [CSZG: 2919]

Penfluridol 1975 {published data only}1515

Kurland AA, Ota KY, Slotnick VB. Penfluridol: a long-acting oral neuroleptic. A controlled study. Journal of Clinical Pharmacology 1975;15:611-21. CENTRAL [CSZG: 1586]
Kurland AA, Ota KY. Penfluridol: once-a-week oral maintenance neuroleptic in the management of chronic schizophrenics. Psychopharmacology Bulletin 1975;11:12-4. CENTRAL [CSZG: 1585]

Penfluridol 1987 {published data only}4221

Channabasavanna SM, Michael A. Penfluridol maintenance therapy in schizophrenia: a controlled study. Indian Journal of Psychiatry 1987;29(4):333-6. CENTRAL [CSZG: 1094]

Perphenazine 1963 {published data only}

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

Pimozide 1971 {published data only}1548

Huber W, Serafetinides EA, Colmore JP, Clark M. Pimozide in chronic schizophrenic patients. Journal of Clinical Pharmacology 1971;11(4):304-9. CENTRAL [CSZG: 1618]

Pimozide 1973 {published data only}1356

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. CENTRAL [CSZG: 1409]

Quetiapine 2007 {published data only}10847

D1441C00131. A 1-year, international, multicenter, randomized, double-blind, parallel-group, placebo-controlled phase IV study to evaluate prevention of relapse in patients in stable condition with chronic schizophrenia receiving either quetiapine fumarate IR (SEROQUEL) or placebo. https://astrazenecagrouptrials.pharmacm.com/ST/Submission/View?id=3852003. CENTRAL
D1444C00004. A 1-year, international, multicenter, randomized, double-blind, parallelgroup, placebo-controlled phase iii study to evaluate prevention of relapse in patients in stable condition with chronic schizophrenia receiving either sustained-release quetiapine fumarate (SEROQUEL) or placebo. https://astrazenecagrouptrials.pharmacm.com/ST/Submission/View?id=14182006. CENTRAL
NCT00228462. A 1-year, multicenter, randomized, double-blind, parallel group, placebo-controlled phase 3 study to evaluate prevention of relapse in patients in stable chronic schizophrenia receiving either sustained-release quetiapine fumarate (SEROQUEL) or placebo (abbreviated). https://www.ClinicalTrials.gov/ct/show/2005. CENTRAL [CSZG: 14874]
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. CENTRAL [CSZG: 16025]
Peuskens J, Trivedi JK, Brecher M, Miller F, Study I . Long-term symptomatic remission of schizophrenia with once-daily extended release quetiapine fumarate: Post-hoc analysis of data from a randomized withdrawal, placebo-controlled study. International Clinical Psychopharmacology 2010;25(3):183-7. CENTRAL [CSZG: 20670]
Peuskens J, Trivedi JK, Brecher M, Svensson O, Miller F, Meulien D. Long-term symptomatic remission of schizophrenia with once-daily extended release quetiapine fumarate. In: Proceedings of the 161st Annual Meeting of the American Psychiatric Association; 2008 May 3-8; Washington DC, USA. 2008. CENTRAL [CSZG: 16500]
Peuskens J, Trivedi JK, Brecher M, Svensson O, Miller F, Meulien D. Long-term symptomatic remission of schizophrenia with once-daily extended release quetiapine fumarate. Schizophrenia Research 2008;98:14-5. CENTRAL [CSZG: 16344]
Peuskens J, Trivedi JK, Malyarov S, Brecher M, Svensson O, Miller F, et al. A randomized, placebo-controlled, relapse-prevention study with once-daily quetiapine sustained release in patients with schizophrenia. Proceedings of the 11th International Congress on Schizophrenia Research; 2007 Mar 28-Apr 1; Colorado Springs, Colorado, USA2007. CENTRAL [CSZG: 15271]
Peuskens JC, Trivedi JK, Malyarov S, Brecher M, Svensson O, Miller F, et al. A randomized, placebo-controlled, relapse-prevention study with once daily quetiapine sustained release in patients with schizophrenia. Schizophrenia Bulletin 2007;33(2):453. CENTRAL [CSZG: 15225]

Quetiapine 2009a {published and unpublished data}12006NCT00658645

NCT00658645. A multinational, randomised, double-blind, fixed-dose, bifeprunox study combining a 12-week placebo-controlled, quetiapine-referenced phase with a 12-month quetiapine-controlled phase in patients with schizophrenia. https://www.clinicaltrials.gov/ct2/show/NCT00658645?term=NCT00658645&rank=1. CENTRAL [CSZG: 16241]

Quetiapine 2009b {published and unpublished data}12579NCT00704509

NCT00704509. A multinational, randomised, double-blind, fixed-dose, bifeprunox study combining a 12-week placebo-controlled, quetiapine-referenced phase with a 12-month quetiapine-controlled phase in patients with schizophrenia. https://www.clinicaltrials.gov/ct2/show/NCT00704509?term=NCT00704509&rank=1. CENTRAL [CSZG: 16566]

Quetiapine 2010 {published data only}10486

Chen E, Hui C, Chang WC, Chan S, Lee E, Honer W. The effect of early medication discontinuation on long-term clinical outcome in first episode psychosis. Schizophrenia Bulletin 2018;44(Suppl 1):S97. CENTRAL
Chen E, Hui C, Lam M, Law C, Chiu C, Chung D, et al. Assessing the efficacy of relapse prevention in single episode psychosis patients with stable maintenance treatment for at least 1 year: a double-blind randomized placebo-controlled. Early Intervention in Psychiatry 2008;2(Suppl 1):A5. CENTRAL [CSZG: 19113]
Chen EY, Hui CL, Lam M, Law CW, Chiu CP, Chung DW, 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. CENTRAL [CSZG: 16325]
Chen EY, Hui CL, Lam M, Law CW, Chiu CP, Chung DW, et al. A double-blind randomized placebo-controlled relapse prevention study in remitted first-episode psychosis patients following one year of maintenance treatment. Early Intervention in Psychiatry 2010;4(Suppl 1):13. CENTRAL [CSZG: 23475]
Chen EY, Hui CL, Lam MM, Chiu CP, Law CW, Chung DW, et al. Maintenance treatment with quetiapine versus discontinuation after one year of treatment in patients with remitted first episode psychosis: randomised controlled trial. BMJ 2010;341:c4024. CENTRAL [CSZG: 20857]
Chen YH, Hui LM, Lam M, Law CW, Chiu PY, Chung WS, et al. A double-blind randomized placebo-controlled relapse prevention study in remitted first-episode psychosis patients following one year of maintanence therapy. European Psychiatry 2008;23:S107-S8. CENTRAL [CSZG: 20468]
Hui C L, Wong GH, Tang JY, Chang WC, Chan SK, Lee EH, et al. Predicting 1-year risk for relapse in patients who have discontinued or continued quetiapine after remission from first-episode psychosis. Schizophrenia Research 2013;150(1):297-302. CENTRAL [CSZG: 28601]
Hui C, Chen E, Lam M, Law C, Chiu C, Chung D, et al. Can we predict relapse in single episode psychosis patients with stable maintenance treatment for at least one year? Early Intervention in Psychiatry 2008;2(Suppl 1):A124. CENTRAL [CSZG: 19123]
Hui CL, Chen EY, Lam M, Law CW, Chiu CP, Chung DW, et al. Predictors for relapse in a double-blind randomized placebo-controlled discontinuation study of remitted first-episode psychosis patients. Schizophrenia Research 2008;98:13. CENTRAL [CSZG: 16333]
Hui CL, Honer W, Lee E, Chan S, Chang WC, Chen E. The 10-year clinical outcome of early medication discontinuation in remitted first episode psychosis. Early Intervention in Psychiatry 2018;12(1):46. CENTRAL
Hui CL, Honer WG, Lee EH, Chang WC, Chan SK, Chen ES, et al. Long-term effects of discontinuation from antipsychotic maintenance following first-episode schizophrenia and related disorders: a 10 year follow-up of a randomised, double-blind trial. Lancet Psychiatry 2018 May;5(5):432-42. CENTRAL [CSZG: 38003]
Hui CL, Honer WG, Lee EH, Chang WC, Chan SK, Chen ES, et al. Predicting first-episode psychosis patients who will never relapse over 10 years. Psychological Medicine 2019;49(13):2206-14. CENTRAL
Hui CL, Honer WG, Lee EH, Chang WC, Chan SK, Chen EY. Factors associated with successful medication discontinuation after a randomized clinical trial of relapse prevention in first-episode psychosis: a 10-year follow-up. JAMA Psychiatry 2019;76(2):217-9. CENTRAL
Hui CL, Li YK, Li AW, Lee EH, Chang WC, Chan SK, et al. Visual working memory deterioration preceding relapse in psychosis. Psychological Medicine 2016;46:2435-44. CENTRAL [CSZG: 33663]
Hui LM, Chen YH, Lam M, Law CW, Chiu PY, Chung WS, et al. A double-blind randomized placebo-controlled study of relapse predictors in remitted first-episode psychosis patients. European Psychiatry 2008;23:S120-S1. CENTRAL [CSZG: 20469]
NCT00334035. Duration of maintenance anti-psychotic therapy after first –episode schizophrenia: a double-blind randomized placebo-control relapse prevention study. https://www.ClinicalTrials.gov/ct/show/2006. CENTRAL [CSZG: 14607]

Trifluoperazine 1969 {published data only}2258

Prien RF, Levine J, Cole JO. High dose trifluoperazine therapy in chronic schizophrenia. American Journal of Psychiatry 1969;126:305-13. CENTRAL [CSZG: 2344]
Prien RF, Levine J, Switalski RW. Discontinuation of chemotherapy for chronic schizophrenics. Hospital and Community Psychiatry 1971;22(1):4-7. CENTRAL [CSZG: 2346]

Trifluoperazine 1972 {published data only}951

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. CENTRAL [CSZG: 1001]
Stevens BC. Role of fluphenazine decanoate in lessening the burden of chronic schizophrenics on the community. Psychological Medicine 1973;3:141-58. CENTRAL [CSZG: 2739]

Various drugs 1960 {published data only}877

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. CENTRAL [CSZG: 927]

Various drugs 1961 {published data only}6740

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. CENTRAL [CSZG: 9157]

Various drugs 1962a {published data only}31690

Olson GW, Peterson DB. Intermittent chemotherapy for chronic psychiatric inpatients. Journal of Nervous and Mental Disease 1962;134:1459. CENTRAL [CSZG: 38115]
Olson GW, Peterson DB. Sudden removal of tranquilizing drugs from chronic psychiatric patients. Journal of Mental and Nervous Disease 1960;131:252-5. CENTRAL

Various drugs 1962b {published data only}2769

Troshinsky C, Aaronson HG, Stone RK. Maintenance phenothiazines in aftercare of schizophrenic patients. Pennsylvania Psychiatric Bulletin 1962;2:11-5. CENTRAL [CSZG: 2855]

Various drugs 1964a {published data only}3067

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. CENTRAL [CSZG: 3153]
Caffey EM, Forrest IS, Frank TV, Klett CJ. Phenothiazine excretion in chronic schizophrenics. American Journal of Psychiatry 1963;120:578-82. CENTRAL [CSZG: 420]

Various drugs 1964b {published data only}4149

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. CENTRAL [CSZG: 4833]

Various drugs 1966a {published data only}3195

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. CENTRAL [CSZG: 3289]

Various drugs 1966b {published data only}3495

Melnyk WT, Worthington AG, Laverty SG. Abrupt withdrawal of chlorpromazine and thioridazine from schizophrenic in-patients. Canadian Psychiatric Association Journal 1966;11:410-3. CENTRAL [CSZG: 3590]

Various drugs 1968 {published data only}2006

Morton MR. A study of the withdrawal of chlorpromazine or trifluoperazine in chronic schizophrenia. American Journal of Psychiatry 1968;124:1585-8. CENTRAL [CSZG: 2092]

Various drugs 1971 {published data only}3723

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(854):633-7. CENTRAL [CSZG: 3799]
Hirsch SR, Leff JP, Wing JK. Outpatient maintenance of chronic schizophrenics with long-acting. British Medical Journal 1973;2(5868):715-6. CENTRAL [CSZG: 3839]
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. CENTRAL
Leff JP, Wing JK. Trial of maintenance therapy in schizophrenia. British Medical Journal 1971;3:599-604. CENTRAL [CSZG: 3840]
Leff JP. Influence of selection of patients on results of clinical trials. British Medical Journal 1973;4(5885):156-8. CENTRAL [CSZG: 7717]
Stevens B. The social value of fluphenazine decanoate. Acta Psychiatrica Belgica 1976;76(5):792-804. CENTRAL [CSZG: 2745]
Stevens BC. Role of fluphenazine decanoate in lessening the burden of chronic schizophrenics on the community. Psychological Medicine 1973;3(2):141-58. CENTRAL [CSZG: 2739]

Various drugs 1974 {published data only}3237

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. CENTRAL [CSZG: 3331]

Various drugs 1975 {published data only}3868

Clark ML, Huber WK, Hill D, Wood F, Costiloe JP. Pimozide in chronic schizophrenic outpatients. Diseases of the Nervous System 1975;36(3):137-41. CENTRAL [CSZG: 1235]
Clark. Pimozide vs thioridazine vs placebo. Bulletin 1973;9:59-63. CENTRAL [CSZG: 4106]

Various drugs 1981a {published data only}6229

Blackburn HL, Allen JL. Behavioral effects of interrupting and resuming tranquilizing medication among schizophrenics. Journal of Nervous and Mental Disease 1981;133:303-8. CENTRAL [CSZG: 8369]

Various drugs 1981b {published data only}442

Cheung HK. Schizophrenics fully remitted on neuroleptics for 3-5 years - to stop or continue drugs? British Journal of Psychiatry 1981;138:490-4. CENTRAL [CSZG: 460]

Various drugs 1981c {published data only}3827

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. CENTRAL [CSZG: 27894]
Wistedt B, Palmstierna T. Depressive symptoms in chronic schizophrenic patients after withdrawal of long-acting neuroleptics. Journal of Clinical Psychiatry 1983;44:369-71. CENTRAL [CSZG: 3068]
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. CENTRAL [CSZG: 3069]
Wistedt B, Wiles D, Jorgensen A. A depot neuroleptic withdrawal study neurological effects. Psychopharmacology 1983;80:101-5. CENTRAL [CSZG: 3070]
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. CENTRAL [CSZG: 27892]
Wistedt B. Neuroleptics and depression. Archives of General Psychiatry 1982;39:745. CENTRAL [CSZG: 4059]

Various drugs 1982 {published data only}2081

Nishikawa T, Tsuda A, Tanaka M, Koga I, Uchida Y. Prophylactic effect of neuroleptics in symptom-free schizophrenia. Psychopharmacology 1982;77:301-4. CENTRAL [CSZG: 2167]

Various drugs 1984a {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. CENTRAL

Various drugs 1984b {published data only}2082

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. CENTRAL [CSZG: 2168]

Various drugs 1986a {published data only}599

Crow TJ, MacMillan JF, Johnson AL, Johnstone EC. A randomised controlled trial of prophylactic neuroleptic treatment. British Journal of Psychiatry 1986;148:120-7. CENTRAL [CSZG: 648]

Various drugs 1986b {published data only}2626

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. CENTRAL [CSZG: 2712]

Various drugs 1989 {published data only}3775

McCreadie RG, Wiles D, Grant S, Crockett GT, Mahmood Z, Livingston MG, et al. The Scottish first episode schizophrenia study. VII two-year follow-up. Acta Psychiatrica Scandinavica 1989;80:597-602. CENTRAL [CSZG: 3968]
Scottish Schizophrenia Research Group. The Scottish First Episode Schizophrenia Study II. Treatment. British Journal of Psychiatry 1987;150:334-8. CENTRAL [CSZG: 3969]
Scottish Schizophrenia Research Group. The Scottish First Episode Schizophrenia Study V. One-year follow-up. British Journal of Psychiatry 1988;152:470-476. CENTRAL [CSZG: 3970]
Scottish Schizophrenia Research Group. The Scottish First Episode Schizophrenia Study. III. Cognitive performance. British Journal of Psychiatry 1987;150:338-340. CENTRAL [CSZG: 16867]
Scottish Schizophrenia Research Group. The Scottish first episode schizophrenia study. VIII. Five-year follow-up. British Journal of Psychiatry 1992;161:496-500. CENTRAL [CSZG: 2542]

Various drugs 1993 {published data only}2205

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. CENTRAL [CSZG: 831]
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. CENTRAL [CSZG: 8197]
Gaebel W, Moeller HJ. Treatment strategies in first episode schizophrenia. In: Proceedings of the 12th World Congress of Psychiatry; 2002 Aug 24-29; Yokohama, Japan. 2002. CENTRAL [CSZG: 8597]
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. CENTRAL [CSZG: 3944]
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. CENTRAL [CSZG: 3943]

Various drugs 2011 {published data only}10307

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. CENTRAL [CSZG: 22940]
Boonstra G. Schizophrenia termination of pharmacotherapy-STOP-trial. Current Controlled Trials2005. CENTRAL [CSZG: 14487] [ISRCTN: 6332944]

Ziprasidone 2002 {published data only}227

Arato M, O'Connor R, Bradbury JE, Meltzer H. Long-term ziprasidone in schizophrenia. In: Proceedings of the 151st Annual Meeting of the American Psychiatric Association; 1998 May 30 - Jun 4; Toronto, Ontario, Canada. 1998. CENTRAL [CSZG: 3189]
Arato M, O'Connor R, Bradbury JE, Meltzer H. The efficacy of ziprasidone in the long-term treatment of negative symptoms and prevention of exacerbation of schizophrenia. In: Proceedings of the 21st Collegium Internationale Neuro-Psychopharmacologicum Congress; 1998 Jul 12-16; Glasgow, UK. Amsterdam, Netherlands: Cambridge University Press, 1998. CENTRAL [CSZG: 3188]
Arato M, O'Connor R, Bradbury JE, Meltzer H. Ziprasidone in the long-term treatment of negative symptoms and prevention of exacerbation of schizophrenia. In: Proceedings of the 9th Congress of the Association of European Psychiatrists; 1998 Sep 20-24; Copenhagen, Denmark. 1998. CENTRAL [CSZG: 3190]
Arato M, O'Connor R, Bradbury JE, Meltzer H. Ziprasidone in the long-term treatment of negative symptoms and prevention of exacerbation of schizophrenia. Schizophrenia Research 1999;36(1-3):270. CENTRAL [CSZG: 5626]
Arato M, O'Connor R, Meltzer H, Bradbury J. Ziprasidone: efficacy in the prevention of relapse and in the long-term treatment of negative symptoms of chronic schizophrenia. In: Proceedings of the 10th European College of Neuropsychopharmacology Congress; 1997 Sep 13-17; Vienna, Austria. 1997. CENTRAL [CSZG: 245]
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. CENTRAL [CSZG: 18486]
Bernardo M, Aranza JR, Rubio-Terres C, Rejas J. Cost-effectiveness analysis of schizophrenia relapse prevention. Clinical Drug Investigation. New Zealand 2006;26(8):447-57. CENTRAL [CSZG: 13898]
Bernardo M, Aranza JR, Rubio-Terres C, Rejas J. Cost-effectiveness analysis of the prevention of relapse in schizophrenia in the ZEUS longitudinal study Ziprasidone Extended Use in Schizophrenia (ZEUS). Acta Espanolas de Psiquiatria 2007;35(4):259-62. CENTRAL [CSZG: 15396]
Meltzer HY, O´Connor R. Long-term efficacy of ziprasidone in schizophrenia. Schizophrenia research 2001;(1-2):S. 239. CENTRAL [CSZG: 6050]
Meltzer HY, O´Connor R. Path analysis of the ZEUS study provides evidence of a direct effect of ziprasidone on primary negative symptoms in chronic, stable schizophrenia. Conference Poster1997. CENTRAL [CSZG: 2033]
Meltzer HY, O'Connor R. Long-term efficacy of ziprasidone in schizophrenia: results of two controlled trials. In: Proceedings of the 39th Annual Meeting of the American College of Neuropsychopharmacology; 2000 Dec 10-14; San Juan, Puerto Rico. 2000. CENTRAL [CSZG: 7609]
O'Connor R, Schooler NR. Penultimate observation carried forward (POCF): a new approach to analysis of long-term symptom change in chronic relapsing conditions [letter]. Schizophrenia Research 2003;60:319-20. CENTRAL [CSZG: 9365]
Pfizer 2000. Multicentre double-blind study of ziprasidone versus placebo in relapse prevention for hospitalised patients with chronic or subchronic schizophrenia. Study report Unpublished. CENTRAL
Schooler N. A basis for optimism: the implications of long term treatment. In: Proceedings of the 9th Congress of the Association of European Psychiatrists; 1998 Sep 20-24; Copenhagen, Denmark. 1998. CENTRAL [CSZG: 5864]

Zotepine 2000 {published data only}544

Cooper SJ, Butler A, Tweed J, Raniwalla J, Welch C. Zotepine in the prevention of relapse. Biological Psychiatry 1997;42:41s. CENTRAL [CSZG: 17007]
Cooper SJ, Butler A, Tweed J, Raniwalla J, Welch C. Zotepine in the prevention of relapse. In: Proceedings of the 6th World Congress of Biological Psychiatry; 1997 June 22-27; Nice, France. 1997. CENTRAL [CSZG: 569]
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. CENTRAL [CSZG: 5732]
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. In: Proceedings of the 11th European College of Neuropsychopharmacology Congress; 1998 Oct 31 - Nov 4; Paris, France. 1998. CENTRAL [CSZG: 5784]
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. CENTRAL [CSZG: 4925]

Allen 1997 {published data only}

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

Bai 2003 {published data only}

Bai YM, Yu SC, Chen JY, Lin CY, Chou P, Lin CC. Risperidone for pre-existing severe tardive dyskinesia: a 48-week prospective follow-up study. International Clinical Psychopharmacology 2005;20:79-85. CENTRAL
Bai YM, Yu SC, Lin CC. Risperidone for severe tardive dyskinesia: a 12 week randomised, double-blind, placebo-controlled study. Journal of Clinical Psychiatry 2003;64:1342-8. CENTRAL

Bechdolf 2016 {published data only}

Bechdolf A, Mueller H, Gaebel W, Hasan A, Maier W, Juckel G, et al. PREVENT: a randomized controlled trial comparing cognitive behavioural therapy, clinical management and aripiprazole and clinical management and placebo for the prevention of first episode psychosis. Early Intervention in Psychiatry 2016;10(Suppl 1):40. CENTRAL

Bo 2017 {published data only}

Bo Q, Li F, Li X, Wang Z, Dong F, He F, et al. Symptomatic remission in schizophrenia: results from a risperidone maintenance treatment study. Psychiatry Research 2017;258:289-94. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL

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. CENTRAL

Brown 2018 {published data only}

Brown D, Daniels K, Pichereau S, Sand M. A phase IC study evaluating the safety, tolerability, pharmacokinetics and cognitive outcomes of BI409306 in patients with mild to moderate schizophrenia. Neurology and Therapy 2018;7(1):129-39. CENTRAL
Sand M, Nakagome K, Cordes J, Brenner R, Gründer G, Keefe R, et al. Effects of BI409306 on positive and negative syndrome scale in schizophrenia: a randomized double-blind placebo-controlled phase II trial. Biological Psychiatry 2018;83(9):S207-8. CENTRAL

Cather 2018 {published data only}

Cather C, Brunette MF, Mueser KT, Babbin SF, Rosenheck R, Correll CU, et al. Impact of comprehensive treatment for first episode psychosis and substance use outcomes: a randomized controlled trial. Psychiatry Research 2018;268:303-11. CENTRAL
Fulford D, Piskulic D, Addington J, Kane JM, Schooler N, Mueser KT. Prospective relationships between motivation and functioning in recovery after a first episode of schizophrenia. Schizophrenia Bulletin 2018;44(2):369-77. CENTRAL
Mueser KT, Meyer-Kalos PS, Glynn SM, Lynde DW, Robinson DG, Gingerich S, et al. Implementation and fidelity assessment of the NAVIGATE treatment program for first episode psychosis in a multi-site study. Schizophrenia Research 2019;204:271-81. CENTRAL
Schooler N, Mueser K, Estroff S, Correll C, Robinson D, Marcy P, et al. Elements of recovery in first episode psychosis: developing measurable criteria. Early Interventions in Psychiatry 2016;10(Suppl 1):149. CENTRAL

Cheng 2019 {published data only}

Cheng Z, Yuan Y, Han X, Yang L, Zeng X, Yang F, et al. Rates and predictors of one year antipsychotic treatment discontinuation in first episode schizophrenia: results from ab open-label randomized "real-world" clinical trial. Psychiatry Research 2019;273:631-40. CENTRAL

Chopra 2019 {published data only}

Chopra S, Fornito A, Francey S, O’Donoghue B, Nelson B, Graham J, et al. Gray matter changes in medicated and unmedicated first episode psychosis: a randomized placebo-controlled trial. Schizophrenia Bulletin 2019;45:S172-3. CENTRAL

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. CENTRAL

Chouinard 1993 {published data only}

Andris J. Clinical trials van risperidone [Clinical trails van risperidone]. Unknown Source1997:14. CENTRAL
Chouinard G, Albright PS. Economic and health state utility determinations for schizophrenic patients treated with risperidone or haloperidol. Journal of Clinical Psychopharmacology 1997;17(4):298-307. CENTRAL
Chouinard G, Arnott W. The effect of risperidone on extrapyramidal symptoms in chronic schizophrenic patients. Biological Psychiatry 1992;31:158. CENTRAL
Chouinard G, Jones B, Remington G, Bloom D, Addington D, MacEwan GW, et al. A Canadian multicenter placebo-controlled study of fixed doses of risperidone and haloperidol in the treatment of chronic schizophrenic patients. Journal of Clinical Psychopharmacology 1993;13(1):25-40. CENTRAL
Chouinard G. Effects of risperidone in tardive dyskinesia: an analysis of the Canadian multicenter risperidone study. Journal of Clinical Psychopharmacology1995;15(Suppl 1):S36-44. CENTRAL
Czobor P, Volavka J, Meibach RC. Effect of risperidone on hostility in schizophrenia. Journal of Clinical Psychopharmacology 1995;15(4):243-9. CENTRAL
Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. Journal of Clinical Psychological Medicine 1997;58(12):538-46. CENTRAL
McEvoy JP. Efficacy of risperidone on positive features of schizophrenia. Journal of Clinical Psychiatry 1994;55(5 Suppl):18-21. CENTRAL
Moller HJ, Muller H, Borison RL, Schooler NR, Chouinard G. A path-analytical approach to differentiate between direct and indirect drug effects on negative symptoms in schizophrenic patients. A re- evaluation of the North American risperidone study. European Archives of Psychiatry and Clinical Neuroscience 1995;245(1):45-9. CENTRAL
Musser WS, Kirisci L. Critique of the Canadian multicenter placebo-controlled study of risperidone and haloperidol. Journal of Clinical Psychopharmacology 1995;15(3):226-8. CENTRAL
NCT00249132. Risperidone versus haloperidol versus placebo in the treatment of chronic schizophrenia. https://www.ClinicalTrials.gov/ct/show/2005. CENTRAL
Simpson GM, Lindenmayer JP. Extrapyramidal symptoms in patients treated with risperidone. Journal of Clinical Psychopharmacology 1997;17(3):194-201. CENTRAL

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. CENTRAL

Clark 1967 {published data only}

Clark ML, Ray TS, Paredes A, Ragland RE, Costiloe JP, Smith CW, et al. Chlorpromazine in women with chronic schizophrenia: the effect on cholesterol levels and cholesterol-behavior relationships. Psychosomatic Medicine 1967;29(6):634-42. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL
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. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL

Durgam 2016 {published data only}

Barabassy A, Szatmàri B, Laszlovszky I, Harsanyi J, Acsai K, Sebe B, et al. Efficacy of cariprazine in the treatment of negative symptoms of schizophrenia: post hoc analysis versus aripiprazole. European Neuropsychopharmacology 2019;29(Suppl 1):S417-8. CENTRAL
Cutler AJ, Durgam S, Lu K, Laszlovszky I, Szalai E, Edwards J, Earley W. Efficacy of cariprazine in negative, cognitive, social function symptoms in schizophrenia: post hoc analysis of a randomized controlled trial. European Neuropsychopharmacology 2016;26(2):S551-2. CENTRAL
Durgam S, Earley W, Lu K, Németh G, Laszlovszky I, Szatmari B, Patel M, Nasrallah H. Cariprazine for negative symptoms of schizophrenia: a pooled post-hoc analysis of 2 randomized double-blind placebo and active controlled trials. In: 29th European College of Neuropsychopharmacology Congress. Vienna, Austria, 2016, Sep 17-20. CENTRAL
Marder S, Fleischhacker WW, Earley W, Kaifeng L, Zhong Y, Németh G. Efficacy of cariprazine across symptom domains in patients with acute exacerbation of schizophrenia: pooled analysis from 3 phase II/III studies. European Neuropsychopharmacology 2019;29(1):127-36. CENTRAL

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. CENTRAL

Fleischhacker 2014 {published data only}

Fleischhacker WW, Sanchez R, Perry PP, Jin N, Peters-Strickland T, Johnson BR, et al. Aripiprazole once-monthly for treatment of schizophrenia: double-blind, randomised, non-inferiority study. British Journal of Psychiatriy 2014;205(2):135-44. CENTRAL

Francey 2018 {published data only}

Francey S, Nelson B, Jessica G, Lara B, Yuen HP, O'Donoghue B, et al. Antipsychotic medication in first episode psychosis: an RCT to assess the risk benefit ratio. Early Intervention in Psychiatry 2018;12(Suppl 1):70. CENTRAL
Francey S. A randomized placebo-controlled trial of intensive psychosocial treatment plus or minus antipsychotic medication for first episode psychosis with low risk of self harm of aggression. The STAGES study: staged treatment and acceptability guidelines in early psychosis. Early Interventions in Psychiatry 2016;10(Suppl 1):9. CENTRAL
O'Donoghue B, Francey SM, Nelson B, Ratheesh A, Allott K, Grahan J, et al. Staged treatment and acceptability guidelines in early psychosis study (STAGES): a randomized placebo-controlled trial of intensive psychosocial treatment plusof minus antipsychotic medication for first episode with low risk of self harm or aggression: study protocol and baseline characteristics of patients. Early Interventions in Psychiatry 2019;13(4):953-60. CENTRAL

Freedman 1982 {published data only}

Freedman R, Kirch D, Bell J, Adler LE, Pecevich M, Pachtman E, et al. Clonidine treatment of schizophrenia: double blind comparison to placebo and neuroleptic drugs. Acta Psychiatrica Scandinavica 1982;65:35-45. CENTRAL

Gallant 1964 {published data only}

Gallant DM, Edwards CG, Bishop MP, Galbraith GC. Withdrawal symptoms after abrupt cessation of antipsychotic compounds: clinical confirmation in chronic schizophrenics. American Journal of Psychiatry 1964;121:491-3. CENTRAL

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. CENTRAL

Gitlin 2001 {published data only}

Gitlin M, Nuechterlein K, Subotnik KL, Ventura J, Mintz J, Fogelson DL, et al. Clinical outcome following neuroleptic discontinuation in patients with remitted recent-onset schizophrenia. American Journal of Psychiatry 2001;158:1835-42. CENTRAL

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. CENTRAL

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. CENTRAL
Mefferd RB Jr, 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. CENTRAL

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. CENTRAL
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;127:167-79. CENTRAL

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. CENTRAL

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. CENTRAL

Hirsch 1989 {published data only}

Hirsch SR, Jolley AG, Barnes TR, Liddle PF, Curson DA, Patel A, et al. Dysphoric and depressive symptoms in chronic schizophrenia.. Schizophrenia Research 1989;2(3):259-64. CENTRAL
Hirsch SR, Jolley AG. The dysphoric syndrome in schizophrenia and its implications for relapse [Das dysphorische Syndrom in der Schizophrenie und seine Implikationen fuer den Rueckfall]. In: Boeker W//Brenner HD//Waldvogel DM, editors(s). Schizophrenie Als Systemische Stoerung. Die Bedeutung Intermediaerer Prozesse Fuer Theorie Und Therapie. Bern, Switzerland: Verlag Hans Huber, 1989:94-103. CENTRAL
Hirsch SR, Jolley AG. The dysphoric syndrome in schizophrenia and its implications for relapse. British Journal of Psychiatry. Supplements 1989;155(Suppl 5):46-50. CENTRAL

Hirsch 1996 {published data only}

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. CENTRAL
Hirsch SR, Bowen JT, Emami J. The effect of life events and medication in the aetiology of schizophrenic relapse. Schizophrenia Research 1993;9:266. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL
Johnstone EC, Owens DGC. Does early treatment have an effect on outcome? In: Proceedings of the 10th European College of Neuropsychopharmacology Congress; 1997 Sep 13-17; Vienna, Austria. 1997. CENTRAL

Keefe 2018 {published data only}

Keefe RS, Harvey PD, Khan A, Saoud JB, Staner C, Davidson M, et al. Cognitive effects of MIN-101 in patients with schizophrenia and negative symptoms: results from a randomized controlled trial. Journal of Clinical Psychiatry 2018;79(3):17m11753. CENTRAL

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. CENTRAL

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. CENTRAL

Lecrubier 1997 {published data only}

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

Liu 2018 {published data only}

Liu CC. A proposed alternative between discontinuation and maintenance of antipsychotics: a guided dose reduction trial for patients with remitted psychosis. Schizophrenia Bulletin 2018;44(Suppl 1):S414. CENTRAL

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. CENTRAL

Mahal 1975 {published data only}

Mahal AS, Janakiramaiah N. A double blind placebo controlled trial of pimozide (R6238) on 49 hospitalized chronic schizophrenics. Indian Journal of Psychiatry 1975;17(1):45-55. CENTRAL

Marder 1994 {published data only}

Andris J. Clinical trials van risperidone [Clinical trails van risperidone]. Unknown Source1997:14. CENTRAL
Czobor P, Volavka J, Meibach RC. Effect of risperidone on hostility in schizophrenia. Journal of Clinical Psychopharmacology 1995;15(4):243-9. CENTRAL
Czobor P, Volavka J. Quantitative electroencephalogram examination of effects of risperidone in schizophrenic patients. Journal of Clinical Psychopharmacology 1993;13(5):332-42. CENTRAL
Lindenmayer JP, =The RSG. Incidence of EPS with risperidone compared with haloperidol and placebo in patients with chronic schizophrenia. In: Proceedings of the 146th Annual Meeting of the American Psychiatric Association; 1993 May 22-27; San Francisco, California, USA. 1993. CENTRAL
Marder S. Risperidone versus haloperidol versus placebo in the treatment of chronic schizophrenia. Clinical Research Report (RIS-INT-3)1991. CENTRAL
Marder SR, Chouinard G, Davis JM. The clinical actions of risperidone. In: Proceedings of the 10th Congress of the European College of Neuropsychopahrmacology; 1997 Sep 13-17, Vienna, Austria. 1997. CENTRAL
Marder SR, Chouinard G, Davis JM. The clinical actions of risperidone. In: Proceedings of the 35th Annual Meeting of the American College of Neuropsychopharmacology; 1996 Dec 9-13; San Juan, Puerto Rico. 1996. CENTRAL
Marder SR, Chouinard G, Davis JM. The clinical actions of risperidone. In: Proceedings of the 6th World Congress of Biological Psychiatry; 1997 Jun 22-27; Nice, France. 1997. CENTRAL
Marder SR, Davis JM, Chouinard G. The effects of risperidone on the five dimensions of schizophrenia derived by factor analysis: combined results of the North American trials. Journal of Clinical Psychological Medicine 1997;58(12):538-46. CENTRAL
Marder SR, Meibach RC. Risperidone in the treatment of schizophrenia. American Journal of Psychiatry 1994;151(6):825-35. CENTRAL
McEvoy JP. Efficacy of risperidone on positive features of schizophrenia. Journal of Clinical Psychiatry 1994;55(5 Suppl):18-21. CENTRAL
Moller HJ, Muller H, Borison RL, Schooler NR, Chouinard G. A path-analytical approach to differentiate between direct and indirect drug effects on negative symptoms in schizophrenic patients. A re- evaluation of the North American risperidone study. European Archives of Psychiatry and Clinical Neuroscience 1995;245(1):45-9. CENTRAL
NCT00249132. Risperidone versus haloperidol versus placebo in the treatment of chronic schizophrenia. https://www.ClinicalTrials.gov/ct/show/2005. CENTRAL
Simpson GM, Lindenmayer JP. Extrapyramidal symptoms in patients treated with risperidone. Journal of Clinical Psychopharmacology 1997;17(3):194-201. CENTRAL

Mathur 1981 {published data only}

Mathur S, Hall JN. Phenothiazine withdrawal in schizophrenics in a hostel. British Journal of Psychiatry 1981;138:271-2. CENTRAL

Meehan 2019 {published data only}

Meehan SR, Zhang P, Hobart M, Hefting N, Baker RA, Weiss C. Short term efficacy of brexpiprazole in patients with schizophrenia with clinically relevant levels of negative symptoms. European Neuropsychopharmacology 2019;29(Suppl 1):S424. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL
Mosher LR, Menn A, Matthew SM. Soteria: evaluation of a home-based treatment for schizophrenia. American Journal of Orthopsychiatry 1975;45:455-67. CENTRAL
Mosher LR, Menn AZ. Community residential treatment for schizophrenia: two year follow up. Hospital and Community Psychiatry 1978;29:715-23. CENTRAL

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. CENTRAL

NCT03559426 {published data only}

NCT03559426. Research into antipsychotic discontinuation and reduction (RADAR): a randomized controlled trial. https://clinicaltrials.gov/. CENTRAL

Nishikawa 1989 {published data only}

Nishikawa T, Tanaka M, Tsuda A, Koga I, Uchida Y. Prophylactic effects of neuroleptics in symptom-free schizophrenics: a comparative dose-response study of timiperone and sulpiride. Biological Psychiatry 1989;25(7):861-6. CENTRAL
Nishikawa T. Personal Communication. Personal Communication2009. CENTRAL

Oosthuizen 2003 {published data only}

Oosthuizen PP. Treatment of first episode schizophrenia with low-dose haloperidol: a study in the Western Cape province of South Africa. Dissertation presented for the Degree of Doctor of Philosophy at the University of Stellenbosch 2003 March. CENTRAL

Pasamanick 1967 {published data only}

Davis AE, Dinitz S, Pasamanick B. The prevention of hospitalization in schizophrenia: five years after an experimental program. American Journal of Orthopsychiatry 1972;42(3):375-88. CENTRAL
Pasamanick B, Scarpitti FR, Dinitz S. Schizophrenic in theCcommunity: an Experimental Study in the Prevention of Hospitalisation. New York: Meredith Publishing Company, 1967. CENTRAL
Pasamanick B, Scarpitti FR, Lefton M, Dinitz F, Wernert JJ, McPheeters H. Home vs hospital care for schizophrenics. JAMA 1964;187(3):177-81. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL

Pigache 1993 {published data only}

Pigache RM, Norris HN. Measurement of drug action in schizophrenia. Clinical Science 1973;44:28P. CENTRAL
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. CENTRAL
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. CENTRAL
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. CENTRAL

Ran 2002 {published data only}

Ran MS, Xiang MZ, Chan CL, Leff J, Simpson P, Huang MS, et al. Effectiveness of psychoeducational intervention for rural Chinese families experiencing schizophrenia. Social Psychiatry and Psychiatric Epidemiology 2003;38:69-75. CENTRAL
Ran MS, Xiang MZ, Huang MS. A control study of psychoeducational family intervention for relatives of schizophrenics in a Chinese rural community. Chinese Journal of Psychiatry 2001;34(2):98-101. CENTRAL
Ran MS. Community mental health in China - A randomized controlled trial of psychoeducational family intervention for carers of persons with schizophrenia in a rural area in Chengdu. http://hdl.handle.net/10722/35402 submitted at the University of Hong Kong in March 2002. CENTRAL

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. CENTRAL

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. CENTRAL
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. CENTRAL

Ruiz 1975 {published data only}

Ruiz Ruiz M, Miro Quintana L, Sentis Vilalta J. A clinical study with pimozide in chronic schizophrenics [Estudio clinico con pimocide en las esquizophrenias de evolucion cronica]. Revista de Psiquiatria Y Psicologia Medica de Europa Y America Latinas 1975;12(4):247-56. CENTRAL

Ruiz Veguilla 2013 {published data only}

NCT01765829. Clinical trial to evaluate the efficacy of treatment vs discontinuation in a first episode of non-affective psychosis (NONSTOP). https://www.clinicaltrials.gov/ct2/show/NCT01765829?term=NCT01765829&rank=12013. CENTRAL

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. CENTRAL

Singer 1971 {published data only}

Singer K, Cheng MN. Thiopropazate hydrochloride in persistent dyskinesia. British Medical Journal 1971;4:22-25. CENTRAL

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. CENTRAL

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. CENTRAL

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. CENTRAL

Stuerup 2017 {published data only}

Dolmer S, Nielsen M, Birk M, Mors O, Sturup AE, Albert N, et al. Tailor tapered discontinuation versus maintenance therapy of antipsychotic medication in patients with newly diagnosed schizophrenia spectrum disorders in remission of psychotic symptoms. Schizophrenia Bulletin 2018;44(Suppl 1):S134-S135. CENTRAL
EudraCT 2016-000565-23. TAILOR - a randomized clinical trial: Tapered discontinuation versus maintenance therapy of antipsychotic medication in patients with newly diagnosed schizophrenia or schizophreniform psychosis in remission of psychotic symptoms. https://www.clinicaltrialsregister.eu/ctr-search/search?query=2016-000565-232017. CENTRAL
Stuerup AE, Jensen H, Dolmer S, Albert N, Birk M, Hjorthoj C, et al. Discontinuation versus maintenance therapy with antipsychotic medication in schizophrenia. TAILOR: a randomized controlled trial. Early Interventions in Psychiatry 2018;12:141. CENTRAL
Stuerup AE, Jensen HD, Dolmer S, Birk M, Albert N, Nielsen M, et al. TAILOR - tapered discontinuation versus maintenance therapy of antipsychotic medication in patients with newly diagnosed schizophrenia or persistent delusional disorder in remission of psychotic symptoms: study protocol for a randomized clinical trial. Trials 2017;18(1):445. CENTRAL

Sumitomo 2008 {published data only}

Sumitomo Dainippon Pharma. Randomized, placebo-controlled, double-blind, parallel-group, confirmatory study of SM-13496 (lurasidone HCl) in patients with schizophrenia: a phase III study. Summary of study results. Sumitomo Dainippon Pharma Reports2008. CENTRAL

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. CENTRAL

Van Kammen 1982 {published data only}

Van Kammen DP, Bunney WE Jr, 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. CENTRAL
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. CENTRAL
Van Kammen DP, Docherty JP, Marder SR, Schulz SC, Bunney WE Jr. Lack of behavioral supersensitivity to d-amphetamine after pimozide withdrawal. A trial with schizophrenic patients. Archives of General Psychiatry 1980;37:287-90. CENTRAL
Van Kammen DP, Docherty JP, Marder SR, Siris SG, Bunney WE Jr. D amphetamine raises serum prolactin in man: evaluations after chronic placebo, lithium and pimozide treatment. Life Sciences 1978;23:1487-92. CENTRAL

Vanover 2018 {published data only}

Vanover K, Dmitrienko A, Glass S, Kozauer S, Saillard J, Weingart M, et al. Lumateperone (ITI-007) for the treatment of schizophrenia: placebo-controlled clinical trials and an open-label safety switching study. Schizophrenia Bulletin 2018;44(Suppl 1):S341. CENTRAL

Van Praag 1973 {published data only}

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

Weller 2018 {published data only}

Weller A, Killackey E, Gleeson J, Allott K, Alvarez-Jimenez M, Bendall S, et al. Less is more? An Australian RCT comparing dose reduction of anti-psychotic medication to maintenance treatment. Early Interventions in Psychiatry 2018;12(Suppl 1):207. CENTRAL
Weller A, et al. Can antipsychotic dose reduction lead to better functional recovery in first episode psychosis? A randomized controlled trial of antipsychotic dose reduction: the REDUCE trial: study protocol. Early Intervention in Psychiatry 2019;13(6):1345-1356. CENTRAL

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. CENTRAL

Wright 1964 {published data only}

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

Wunderink 2006 {published data only}

Faber G, Smid HG, Van Gool AR, Wiersma D, Van Den Bosch RJ. The effect of guided discontinuation of antipsychotics on neurocognition in first onset psychosis. European Psychiatry 2012;27:275-80. CENTRAL
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. CENTRAL

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. CENTRAL

Zou 2018 {published data only}

Zou X, Zhu Y, Jackson JW, Bellavia A, Fitxmaurice GM, Centorrino F, et al. The role of PANSS symptoms and adverse events in explaining the effects of paliperidone on social functioning: a causal mediation analysis approach. NPJ Schizophrenia 2018;4(1):13. CENTRAL

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. CENTRAL

Ascher‐Svanum 2011 {published data only}

Ascher-Svanum H, Nyhuis A, Faries D, Novick D, Kinon B. Differences between schizophrenia patients who switch vs discontinue antipsychotic therapy. Schizophrenia Bulletin 2011 March;37(Suppl 1, Abstracts for the 13th International Congress on Schizophrenia Research (ICOSR)):96. CENTRAL

Decot 2011 {published data only}

Decot H, Zhang F, Weinberger DR, Apud J. Effect of placebo and reintroduction of antipsychotics in patients with schizophrenia based on COMT Val108/158Met polymorphism. In: International Congress on Schizophrenia Research 84, downloaded from: schizophreniabulletin.oxfordjournals.org,. 2011. CENTRAL

Eisenberg 2016 {published data only}

Eisenberg D, Yankowitz L, Kohn P, Hegarty C, Ianni A, Rubinstein D, Gregory M, Dickinson D, Apud J, Berman K. Neuroleptic-induced striatal blood flow changes and neurocognitive functioning in schizophrenia. Neuropsychopharmacology 2016;41:S223-4. CENTRAL

EUCTR2005‐005499‐34 {published and unpublished data}

EUCTR2005-005499-34. A double-blind, randomised, placebo-controlled, quetiapine-referenced, multicentre study of the long-term bifeprunox efficacy, safety and tolerability in patients with stable schizophrenia. www.clinicaltrialsregister.eu. CENTRAL

Zhang 2006 {published data only}

Zhang Q, He FR, Tian YL, Zhao S, Sun ZH. Double blind study of the contact between antipsychotic and the negative symptoms of the deteriorated schizophrenia. Medical Journal of Chinese People Health 2006;3:182-3. CENTRAL

NCT03503318 {published data only}

NCT03503318. A multicenter, randomized, double-blind, placebo-controlled study to evaluate the efficacy, safety, and tolerability of risperidone extended-release injectable suspension (TV-46000) for subcutaneous use as maintenance treatment in adult and adolescent patients with schizophrenia. https://clinicaltrials.gov. CENTRAL

NCT03593213 {published data only}

NCT03593213. Clinical trial evaluating the efficacy, safety and tolerability of cariprazine in a dose-reduction paradigm in the prevention of relapse in patients with schizophrenia. https://clinicaltrials.gov. CENTRAL

NCT03893825 {published data only}

NCT03893825. A study to test if TV-46000 is safe for maintenance treatment of schizophrenia. https://clinicaltrials.gov. CENTRAL

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. Art. No: CD006329. [DOI: 10.1002/14651858.CD006329]

Altman 1996

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

American Psychiatric Association 1987

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 3rd rev edition. Washington DC: American Psychiatric Association, 1987.

Andreasen 2005

Andreasen NC, Carpenter WT Jr, Kane JM, Lasser RA, Marder SR, Weinberger DR. Remission in schizophrenia: proposed criteria and rationale for consensus. American Journal of Psychiatry 2005;162(3):441-9.

Auquier 2003

Auquier P, Simeoni MC, Sapin C, Reine G, Aghababian V, Cramer J, et al. Development and validation of a patient-based health-related quality of life questionnaire in schizophrenia: the S-QoL. Schizophrenia Research 2003;63(1-2):137-49.

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. BMJ 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]

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

Boyer 2010

Boyer L, Simeoni MC, Loundou A, D'Amato T, Reine G, Lancon C, et al. The development of the S-QoL 18: a shortened quality of life questionnaire for patients with schizophrenia. Schizophrenia Research 2010;121:241-50.

Burns 2007

Burns T, Patrick D. Social functioning as an outcome measure in schizophrenia studies. Acta Psychiatrica Scandinavica 2007;116:403-18.

Carpenter 1994

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

Davis 1974

Davis JM. Dose equivalence of the antipsychotic drugs. Journal of Psychiatric Research 1974;11:65-9.

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, editors(s). Comprehensive Textbook of Psychiatry. 5th edition. Baltimore: Williams & Wilkins, 1989:1591-626.

Deeks 2000

Deeks J. Issues in the selection for meta-analyses of binary data. In: 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. Art. No: CD001359. [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 CS. Bias in meta-analysis detected by a simple, graphical test. BMJ 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.

EuroQol 1990

The EuroQol Group. EuroQol - a new facility for the measurement of health-related quality of life. Health Policy 1990;16(3):199-208.

Gardner 2010

Gardner DM, Murphy AL, O'Donnell H, Centorrino F, Baldessarini RJ. International consensus study of antipsychotic dosing. American Journal of Psychiatry 2010;167(6):686-93.

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.

Gopal 2010

Gopal S, Gassman-Mayer C, Palumbo J, Samtani MN, Shiwach R, Alphs L. Practical guidance or dosing and switching paliperidone palmitate treatment in patients with schizophrenia. Current Medical Research and Opinion 2010;26(2):377-87.

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 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327(7414):557-60.

Higgins 2011

Higgins JP, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated September 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane-handbook.org.

Kalali 1999

Kalali A. Patient satisfaction with, and acceptability of, atypical antipsychotics. Current Medical Research and Opinion 1999;15:135-7.

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.

Kishi 2019

Kishi T, Ikuta T, Matsui Y, Inada K, Matsuda Y, Mishima K, et al. Effect of discontinuation vs maintenance of antipsychotic medication on relapse rates in patients with remitted/stable first-episode psychosis: a meta-analysis. Psychological Medicine 2019;49(5):772-9.

Kishimoto 2014

Kishimoto T, Robenzadeh A, Leucht C, Leucht S, Watanabe K, Mimura M, et al. Long-acting injectable vs oral antipsychotics for relapse prevention in schizophrenia: a meta-analysis of randomized trials. Schizophrenia Bulletin2014;40(1):192-213.

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. [PMID: 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.

Leucht 2013

Leucht S, Cipriani A, Spineli L, Mavridis M, Orey D, Richter F, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet 2013;382(9896):951962.

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.

McGrath 2008

McGrath J, Saha S, Chant D, Welham J. Schizophrenia: a concise overview of incidence, prevalence, and mortality.. Epidemiologic Reviews 2008;30:67-76.

Moher 2001

Moher D, Schulz KF, Altman D. The CONSORT statement: revised recommendations for improving the quality of reports of parallel-group randomised trials. JAMA 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.

Morosini 2000

Morosini PL, Magliano L, Brambilla L, Ugolini S, Pioli R. Development, reliability and acceptability of a new version of the DSM-IV Social and Occupational Functioning Assessment Scale (SOFAS) to assess routine social functioning. Acta Psychiatrica Scandinavica 2000;101:323-9.

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.

R Core Team 2018 [Computer program]

Foundation for Statistical ComputingR: A language and environment for statistical computing. R Core Team, Version 3.6.2. Vienna: Foundation for Statistical Computing, 2018.

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.

Schneider 1983

Schneider LC, Struening EL. SLOF: a behavioural rating scale for assessing the mentally ill. Social Work Research & Abstracts 1983 Fall;19:9-21.

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. JAMA 1995;273:408-12.

Schwarzer 2007

Schwarzer G. Meta: an R package for meta-analyses. R News 2007;7(3):40-5.

Shaffer 1983

Shaffer D, Gould MS, Brasic J, Ambrosini P, Fisher P, Bird H, et al. A Children's Global Assessment Scale (CGAS). JAMA Psychiatry 1983;40:1228-31.

Sheenan 1983

Sheenan DV. The Sheenan Disability Scale. In: The Anxiety Disease and How to Overcome it. New York: Charles Scribner and Sons, 1983:151.

Shokraneh 2017

Shokraneh F, Adams CE. Study-based registers of randomized controlled trials: Starting a systematic review with data extraction or meta-analysis. BioImpacts 2017;4:209-17.

Simpson 1970

Simpson EN, Angus JW. 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. Art. No: CD000967. [DOI: 10.1002/14651858.CD000967.pub2]

Stata 2002 [Computer program]

Intercooled Stata 7.0 for Windows. Stata Corporation. 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;35:153-5.

Vernon 2010

Vernon MK, Revicki DA, Awad G, Dirani R, Panish J, Canuso CM, et al. Psychometric evaluation of the Medication Satisfaction Questionnaire (MSQ) to assess satisfaction with antipsychotic medication among schizophrenia patients. Schizophrenia Research 2010;118(1-3):271-8.

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.

Vita 2018

Vita A, Barlati S. Recovery from schizophrenia: is it possible? Curr Opin Psychiatry 2018;31(00):1-10.

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. Art. No: CD000059. [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.

Zhao 2016

Zhao YJ, Lin L, Teng M, Khoo AL, Soh LB, Furukawa TA, et al. Long-term antipsychotic treatment in schizophrenia: systematic review and network meta-analysis of randomised controlled trials. BJPsych Open 2016;2(1):59-66.

References to other published versions of this review

Bergman 2016

Bergman H, Davies S, Dawson S, Kakourou A, Spineli L. Targeted update of Maintenance treatment with antipsychotic drugs for schizophrenia. The Cochrane Collaboration, Targeted Updates Project2016. [https://community.cochrane.org/news/targeted-updates-project-case-study-b]

Komossa 2009

Komossa K, Depping AM, Heres S, Kissling W, Davis JM, Leucht S. Maintenance treatment with antipsychotic drugs for schizophrenia. Cochrane Database of Systematic Reviews 2009, Issue 4. Art. No: CD008016. [DOI: 10.1002/14651858.CD008016]

Leucht 2012a

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. Lancet2012;379(9831):2063-71.

Leucht 2012b

Leucht S, Tardy M, Komossa K, Heres S, Kissling W, Davis JM. Maintenance treatment with antipsychotic drugs for schizophrenia. Cochrane Database of Systematic Reviews 2012, Issue 5. Art. No: CD008016. [DOI: 10.1002/14651858.CD008016.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Aripiprazole 2003

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 15 mg/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.

Service use ‐ number of participants hospitalised (including non psychiatric reasons).

Death.

Suicide attempts.

Adverse effects.

Violent/aggressive behaviour.

Unable to use/Not included

Global state: CGI (no usable data ).

Mental state: PANSS, BPRS (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Aripiprazole 2017

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, no further details.
Duration: terminated early after 37 events (pre‐planned), mean duration of treatment was 164,5 days.
Design: parallel.
Location: multi‐centre.
Setting: outpatients.

Participants

Diagnosis: adolescent patients (13 to 17 years old) with schizophrenia (DSM‐IV‐TR) stabilised on aripiprazole for 7 to 21 weeks before entering the double‐blind phase.

N = 146.
Gender: 96 men, 50 women.
Age: mean 15,4 years (range 13 to 17 years).
History: duration stable‐ at least 7 weeks (clinical judgment and rating scale defined), remission at baseline‐ n.i., duration ill‐ 2.1 years, number of previous hospitalisations‐ n.i., age at onset‐ 13.3 years, severity of illness‐ mean PANSS total score 64,6, mean CGI‐S total score 3,1, baseline antipsychotic dose‐n.i.

Interventions

1. Drug: aripiprazole. Flexible dose. Allowed dose range: 10 mg/day to 30 mg/day. Mean dose: 19,2 mg/day. N = 98.

2. Placebo: inert placebo. Duration of taper: n.i. N = 48.

Rescue medication: not allowed.

Outcomes

Examined

Relapse: rating scale based and/or need for hospitalisation and/or clinical judgment.

Leaving the study early (any reason, adverse events, inefficacy).

Global state‐ number of patients in symptomatic remission (Andreasen criteria, LOCF endpoint cross‐sectional criteria)

Global state‐ number of patients in sustained remission (Andreasen criteria, maintained for 6 months)

Social functioning: Children´s Global Assessment Scale

Adverse events

Death.

Suicidal ideation: Columbia Suicide Severity Rating Scale.

Unable to use/Not included

Global state ‐ number of participants improved (no usable data).

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

Quality of life: Pediatric Quality of Life Enjoyment and Satisfaction Questionnaire (no usable data).

Notes

Sponsored by Otzuka Pharmaceutical Development and Commercialization.

Adolescent patients subgroup.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomised (2:1 ratio), no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

Low risk

The overall attrition rate of 33% could still be acceptable, though higher than 25%, but more participants in the placebo group left the study early, mostly due to relapse. This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were both analysed on an ITT basis (LOCF method) and provided for completers at several time points.

Selective reporting (reporting bias)

Low risk

The primary outcome relapse was reported as prespecified; Efficacy and Safety: Quality of Life score not reported completely, but it is not a primary outcome.

Other bias

High risk

Terminated early (after 37 relapse events), but it was pre‐planned.

Aripiprazole depot 2012

Study characteristics

Methods

Randomisation: sponsor‐prepared computer‐generated randomisation code (2:1 ratio), stratified by region and by last stabilisation dose of study drug.
Allocation: interactive voice/web response system.
Blinding: IM trial medications had different appearance, but were prepared and administered by an unblinded Trial Drug Manager.
Duration: terminated early after the first pre‐planned interim analysis (64 events); pre‐planned duration: 52 weeks.
Design: parallel.
Location: multi‐centre (108 centres).
Setting: outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), diagnosed at least 3 years before.
N = 403.
Gender: 241 men, 162 women.
Age: 40.6 years.
History: duration stable‐ at least 12 weeks, duration ill‐ 14.6 years, number of previous hospitalisations‐ n.i., age at onset‐ 26 years, severity of illness‐ mean PANSS total score 54.5, mean CGI‐S total score 2.9, baseline antipsychotic dose‐ 391.6 mg/4 weeks, remission at baseline‐ not in remission (at least moderately ill at CGI‐S).

Interventions

1. Drug: aripiprazole IM, 1‐month formulation. N = 269

Flexible dose. Mean dose: 396.3 mg/4 weeks. Allowed dose range: either 300 mg or 400 mg/4 weeks.

2. Placebo: duration of taper: depending on the aripiprazole depot half‐life (between 29.9 and 46.5 days). N = 134.

Rescue medication: benzodiazepines (maximum 6 mg/day) and anticholinergics (antiparkinson) were permitted, although not within 8 to 12 hours (respectively) of rating scale assessments.

Outcomes

Examined

Relapse (rating scale defined and/or need for hospitalisation and/or emergent violent behaviour).

Leaving the study early (any cause, adverse events, inefficacy).

Global state ‐ Number of participants in sustained remission (Andreasen criteria).

Service use ‐ Number of participants hospitalised.

Participants´satisfaction with care: Patient Satisfaction with Medication Questionnaire.

Social functioning: Personal and Social Performance scale.

Adverse effects.

Death.

Suicide ideation and behaviour: CGI‐SS, Columbia Suicide Severity Rating Scale and Columbia Classification Algorythm of Suicide Assessment.

Violent/aggressive behaviour.

Unable to use/Not included

Global state ‐ number of participants improved (no usable data).

Mental state: Positive and Negative Symptoms Scale total scores and subscales (no predefined outcome of interest).

Neurocognitive function: Trail Making Test (A), Tower of London, Letter‐Number Span (no predefined outcome of interest).

Compliance to treatment: Drug Attitude Inventory Score, Medication Adherence Questionnaire (no predefined outcome of interest).

Carer´s satisfaction with care: Investigator´s Assessment Questionnaire (no usable data).

Notes

Sponsored by Otsuka.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sponsor‐prepared computer‐generated randomisation code (2:1 ratio), stratified by region and by last stabilisation dose of study drug

Allocation concealment (selection bias)

Low risk

Interactive voice/web response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

IM trial medications had different appearance (aripiprazole: milky white suspension; placebo: clear solution); they were prepared and administered by an unblinded Trial Drug Manager.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

IM trial medications had different appearance (aripiprazole: milky white suspension; placebo: clear solution); they were prepared and administered by an unblinded Trial Drug Manager. Two participants were unblinded at the site level (one incident with depot dose log, one incidental access to the drug storage cabinet given by the monitor), both withdrawn from the study.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

IM trial medications had different appearance (aripiprazole: milky white suspension; placebo: clear solution); they were prepared and administered by an unblinded Trial Drug Manager. Two participants were unblinded at the site level (one incident with depot dose log, one incidental access to the drug storage cabinet given by the monitor), both withdrawn from the study.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The total attrition rate (35%) was higher than 25% but could still be acceptable. However more participants in the placebo group dropped out, mostly due to relapse (reasons are unbalanced between groups). The primary outcome (relapse) was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were analysed on an ITT basis (LOCF).

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Study terminated early after an interim analysis (64 relapse events), but it was pre‐planned.

Asenapine 2011

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 mg/day or 20 mg/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.

Suicidal ideation and behaviour.

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 usable data ).

Leaving the study early (data are unclear).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules in taste.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules in taste.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical capsules in taste.

Incomplete outcome data (attrition bias)
All outcomes

High risk

High dropout rate, but exact number of dropouts could not be calculated. Dropouts 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 clear evidence for other bias.

Brexpiprazole 2017

Study characteristics

Methods

Randomisation: sponsor‐prepared computer‐generated randomisation code (1:1 ratio), assignment of blocks of randomisation numbers to trial centres and individual numbers to patients.
Allocation: interactive voice/web response system.
Blinding: double‐blind, identical capsules.
Duration: terminated early after the first pre‐planned interim analysis (64 events); pre‐planned duration: 52 weeks.
Design: parallel.
Location: multi‐centre (49 sites across 7 countries).
Setting: inpatients and outpatients

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), diagnosed at least 3 years before; stabilised on study drug after resolution of an acute exacerbation.
N = 202.
Gender: 123 men, 79 women.
Age: 40.3 years.
History: duration stable‐ at least 12 weeks, duration ill‐ 13 years, number of previous hospitalisations‐ n.i., age at onset‐ 27.2 years, severity of illness‐ mean PANSS total score 57.3, mean CGI‐S total score 3.1, baseline antipsychotic dose‐ n.i., remission at baseline‐ n.i. (probably not in remission).

Interventions

1. Drug: Brexpiprazole. N = 97

Fixed dose. Mean dose: 3.6 mg/day. Allowed dose range: 1 mg/day to 4 mg/day.

2. Placebo: duration of taper: no. N = 105.

Rescue medication: n.i.

Outcomes

Examined

Relapse (rating scale defined and/or need for hospitalisation and/or emergent violent/suicidal behaviour).

Leaving the study early (any cause, adverse events, inefficacy).

Social functioning: Personal and Social Performance scale.

Adverse effects

Suicidal ideation and behavior: Columbia Suicide Severity Scale

Unable to use/Not included

Global state ‐ number of participants improved: CGI‐I defined (no usable data).

Global state ‐ number of participants in remission: CGI‐S defined (no usable data).

Mental state: Positive and Negative Symptoms Scale total score and subscales (no predefined outcome of interest)

Affective symptoms: PANSS excited component score, PANSS Marder Anxiety/Depression score (no predefined outcome of interest).

Neurocognitive function: Cogstate computerized cognitive test battery (no predefined outcome of interest)

Social functioning: Global Assessment of functioning (already used data regarding Personal and Social Performance scale).

Notes

Sponsored by Otsuka.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sponsor‐prepared computer‐generated randomisation code, assignment of blocks of randomisation numbers to trial centres and individual numbers to patients.

Allocation concealment (selection bias)

Low risk

Interactive voice/web response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition rate of 46% was high, and more participants in the placebo group left the study early, mostly due to relapse. This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were analysed on an ITT basis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Terminated early after an interim analysis, but it was pre‐planned.

Cariprazine 2016

Study characteristics

Methods

Randomisation: randomised (1:1 ratio), no further details.
Allocation: interactive web response system.
Blinding: double‐blind, identical appearing capsules.
Duration: open‐ended, duration varied from 26 to 72 weeks (mean exposure duration: 232 days in the double‐blind phase).
Design: parallel.
Location: multi‐centre.
Setting: outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), stabilized on treatment for at least 12 weeks before double‐blind phase.
N = 200.
Gender: 132 men, 68 women.
Age: 38.5 years.
History: duration stable‐ at least 12 weeks (duration of treatment), duration ill‐ 11.2 years, mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ mean 4.6., age at onset‐ n.i, severity of illness‐ mean PANSS total score 50.9, mean CGI‐S total score 2.7, baseline antipsychotic dose‐n.i. (for the cariprazine arm: 7.1 mg/day), remission at baseline‐ 85% of the participants met symptomatic remission criteria (Andreasen) at double‐blind baseline.

Interventions

1. Drug: cariprazine. N = 101

Fixed dose. Mean dose: 7.07 mg/day. Allowed dose range: 3 mg/day to 9 mg/day.

2. Placebo: duration of taper: n.i. N = 99.

Rescue medication: anticholinergics (antiparkinson), beta‐blocker (akatihsia), lorazepam or oxazepam (anxiety/agitation).

Outcomes

Examined

Relapse (clinical judgement and/or need of hospitalisation).

Leaving the study early (any reason, inefficacy, adverse events)

Global state ‐ number of participants in sustained remission (Andreasen criteria)

Social functioning: Personal and Social Performance Scale

Adverse events

Death

Suicide attempts

Suicidal ideation: Columbia‐Suicide Severity Rating Scale

Unable to use/Not included

Mental state: PANSS total score and subscores (no predefined outcome of interest), negative symptoms assessed with NSA‐16 (no predefined outcome of interest).

Global state ‐ number of improved participants: CGI‐S and CGI‐I scores (no usable data).

Notes

Sponsored by Forest, Actavis.

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

Investigators and patients were blinded to the double‐blind treatment assignment through an interactive web‐response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical appearing capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical appearing capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical appearing capsules.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition rate was high (68%); it was slightly different between the study arms, and reasons were different (more participants in the placebo group left the study early due to relapse). This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. No full ITT analysis for other outcomes (only patients that had at least one evaluation were included).

Selective reporting (reporting bias)

Low risk

No clear evidence of selective reporting.

Other bias

High risk

Terminated early, but it was pre‐planned.

Chlorpromazine 1959

Study characteristics

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‐blind, 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 200 mg/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.

Suicide attempts.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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, dropout 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 for other bias.

Chlorpromazine 1962

Study characteristics

Methods

Randomisation: participants were ranked for morbidity, then matched, then randomised.
Allocation: procedure not described.
Blinding: double‐blind, 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 of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, 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)
Subjective outcomes

Low risk

Double‐blind, 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‐blind, 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 could 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

Blinding was broken once a participant relapsed.

Chlorpromazine 1968

Study characteristics

Methods

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

Participants

Diagnosis: schizophrenia (clinical diagnosis), continuously hospitalised 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 medication 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 300 mg/day. N = 208.

*2. Drug: chlorpromazine. Fixed dose of 2000 mg/day (titrated within 45 days, dose reduction to 1500 mg/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: a patient was considered relapsed if he regressed and had to be returned to known medication before the end of the 24‐week period.

Leaving the study early.

Global state: number of participants improved.

Death.

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: number of participants in remission (no usable data, only reported for a subgroup of patients evaluated by the same rater during the trial).

Behaviour: Nurses’ Observation Scale for Inpatient Evaluation (no predefined outcome of interest).

Readiness for discharge: Discharge‐Readiness Inventory (no predefined outcome of interest).

Ophthalmologic examination (no predefined outcome of interest).

Notes

Quote: *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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, liquid formulation.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, liquid formulation.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Chlorpromazine 1973

Study characteristics

Methods

Randomisation: randomly assigned, no further details.
Allocation: procedure not described.
Blinding: double‐blind, identical capsules, no further details.
Duration: 2 to 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: 100 mg/day. Mean dose: ~ 260 mg/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.

Service use: number of participants hospitalised.

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 (no usable data).

Number of participants employed (no usable data).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules, no further details.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Chlorpromazine 1975

Study characteristics

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‐blind (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).

Extrapyramidal symptoms: clinical and electrophysiological evaluation (no usable data).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind (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.

Chlorpromazine 1976

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: pharmacists held the key.
Blinding: double‐blind, 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‐216 mg/day CPZ equivalent

Interventions

1. Drug: Chlorpromazine ‐ mean dose: 216 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Fluphenazine 1979

Study characteristics

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 mL to 2.0 mL biweekly. Mean dose: n.i.. N = 23.

2. Drug: oral fluphenazine combined with procyclidine. Flexible dose of 5 mg/day to 20 mg/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. Prophylactic antiparkinson medication.

Outcomes

Examined

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

Leaving the study early.

Adverse effects: dropout due to specific adverse events.

Death.

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).

Employment status (no usable data, unclearly reported).

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).

Suicide attempts (unclearly reported, probably not for the global sample).

Notes

* 11 out of 73 patients were then diagnosed as non schizophrenic by the research staff.

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 of participants and personnel (performance bias)
All outcomes

Low risk

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

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.

Fluphenazine 1980

Study characteristics

Methods

Randomisation: random 2:1, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 24.4 mg/day, depot fluphenazine 30.9 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Fluphenazine 1982

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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: 5‐20 mg/day. Mean dose: n.i.. N = n.i..

2. Drug: depot fluphenazine ‐ Flexible dose. Allowed dose range: 12.5 mg to 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, 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‐blind, 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.

Fluphenazine depot 1968

Study characteristics

Methods

Randomisation: randomly assigned, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 to 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.

Service use: number of participants hospitalised.

Adverse effects (at least one movement disorder).

Unable to use / Not included:

Global state: 7‐point scale of severity (no usable data for the two study arms ).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Fluphenazine depot 1973

Study characteristics

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‐blind, 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 25 mg/month, no additional antipsychotic medication.

Interventions

1. Drug ‐ Fixed/flexible dose: allowed dose range: 25 mg/month ‐ no upper limit. Mean dose: 26.4 mg/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.

Leaving the study early.

Service use: number of participants hospitalised.

Number of participants employed.

Death.

Violent/aggressive behaviour.

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 (no usable data)

Suicidal ideation (no usable data, only reported as referred by the patients´ informants to the study rater).

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 of participants and personnel (performance bias)
All outcomes

Low risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double‐blind, 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‐blind, 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

Fluphenazine depot 1979a

Study characteristics

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 mg 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 mg or 50 mg/month).

Interventions

1. Drug: fluphenazine depot. Fixed dose: 25 mg 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.

Leaving the study early.

Global state: number of participants improved.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

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.

Fluphenazine depot 1979b

Study characteristics

Methods

Randomisation: matched then each pair randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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‐ 3.8 mg fluphenazine biweekly, remission at baseline: yes..

Interventions

1. Drug: fluphenazine decanoate ‐ Flexible dose. Allowed dose range: 1.25 mg to 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 tranquillisers, additional antipsychotic drugs were not allowed.

Outcomes

Examined

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

Leaving the study early.

Unable to use/Not included

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

Low risk

Two participants 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.

Fluphenazine depot 1981

Study characteristics

Methods

Randomisation: randomly assigned.
Allocation: procedure not described.
Blinding: double‐blind, placebo injection.
Duration: 6 weeks.
Design: parallel.
Location:  single‐centre.
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 1976 by Spitzer & Endicott 1976), baseline antipsychotic dose‐ 39.3 mg/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.

Service use: number of participants hospitalised.

Social functioning: Global Assessment Scale (GAS).

Unable to use/Not included

Community adjustment: Weissman Social Adjustment scale (no usable data)

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

Adverse effects: tardive diskinesia (no usable 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)

Unclear risk

Procedure not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo injection.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo injection.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Fluphenazine depot 1982

Study characteristics

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‐blind, 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 50 mg IM four/eight weekly. N = 35.

2. Placebo: vitamin B complex IM. 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).

Leaving the study early.

Death.

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

Unable to use/Not included

Mental state: BPRS (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 of participants and personnel (performance bias)
All outcomes

High risk

Double‐blind, 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)
Subjective outcomes

Unclear risk

Double‐blind, 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‐blind, 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 dropout rate drug 40% versus 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.

Fluphenazine depot 1992

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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, remission at baseline: yes (study defined).

Interventions

1. Drug: fluphenazine decanoate.Fixed dose: mean 50.4 mg/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 BPRS total score and judgement of by nurse according to Psychotic Inpatient Profile.

Unable to us /Not included

Mental state: BPRS 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo was sesame oil of identical volume and identical in physical appearance,

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo was sesame oil of identical volume and identical in physical appearance.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Haloperidol 1973

Study characteristics

Methods

Randomisation: unclear, randomisation assumed due to double‐blinding.
Allocation: procedure not described.
Blinding: double‐blind, 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.

Leaving the study early.

Global state ‐ number of participants improved.

Adverse effects (movement disorders).

Suicide ideation.

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 (NOSIE) (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Haloperidol 1991

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, "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.

Leaving the study early.

Service use: number of participants hospitalised.

Death.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, "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.

Haloperidol depot 1982

Study characteristics

Methods

Randomisation: randomly assigned according to pre‐established randomisation code.
Allocation: randomisation code was unknown to the evaluating investigators.
Blinding: double‐blind, 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 (= 150 mg) four‐weekly, maximum 3 mL (= 300 mg) 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.

Global state: number of participants improved.

Unable to use/Not included

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

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

Adverse effects (no usable data).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, administered by a particular nurse.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, administered by a particular nurse.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Haloperidol depot 1991

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 to 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‐ 60 mg haloperidol decanoate per month (~3.5 mg/day haloperidol).

Interventions

1. Drug: haloperidol decanoate 60 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo injections, no further details.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo injections, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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 ITT basis.

Selective reporting (reporting bias)

Low risk

No selective reporting.

Other bias

Low risk

No clear other bias.

Iloperidone 2016

Study characteristics

Methods

Randomisation: computer‐generated random sequence (1:1 ratio).
Allocation: centralised, interactive voice response system.
Blinding: double‐blind, identical appearing capsules.
Duration: terminated early after 68 relapse events, patients were followed up for up to 26 weeks.
Design: parallel.
Location: multi‐centre.
Setting: outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV), not hospitalised ar the time of screening, then stabilised on iloperidone for at least 12 weeks before the relapse‐prevention phase.
N = 303.
Gender: 178 men, 125 women.
Age: 38.3 years.
History: duration stable‐ clinically stable for at least 12 weeks, no change in treatment for at least 4 weeks, duration ill‐ 12.4 years, mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ 25.9 years, severity of illness‐ mean PANSS total score 55,4, mean CGI‐S total score 3,3, baseline antipsychotic dose‐ n.i., remission at baseline‐ n.i..

Interventions

1. Drug: Iloperidone. N = 153

Flexible dose. Mean dose: 15 mg/day. Allowed dose range: 8 mg/day to 24 mg/day.

2. Placebo: duration of taper: n.i. N = 150.

Rescue medication: anticholinergics (antiparkinson medication), lorazepam (agitation, severe restlessness, insomnia), zolpidem (insomnia).

Outcomes

Examined

Relapse (rating scale based and/or clinical judgement and/or need for hospitalisation or increase in the level of psychiatric care).

Leaving the study early ‐ due to adverse events.

Social functioning: Sheehan Disability Scale.

Adverse effects

Death

Unable to use/Not included

Leaving the study early ‐ due to any cause/inefficacy (no usable data, no crude numbers for relapse are available, subjects withdrawn due to early termination counted as dropouts).

Global state‐ Number of participants improved (no usable data)

Global state ‐ Number of participants in remission (no usable data, CGI‐I not reported).

Mental state: PANSS change in total score, BPRS change in total score (no predefined outcomes of interest)

Notes

Sponsored by Vanda Pharma.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence.

Allocation concealment (selection bias)

Low risk

Centralised, interactive voice response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical appearing capsules

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical appearing capsules

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical appearing capsules

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition rate (57%) was high, and more participants in the placebo group left the study early due to relapse. This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were analysed on an ITT basis (not full ITT, because only participants who received at least one dose of study medication were included) .

Selective reporting (reporting bias)

Unclear risk

Some secondary efficacy outcomes are reported incompletely so that they cannot be entered in the meta‐analysis (e.g. CGI‐I scores).

Other bias

High risk

Terminated early, but it was pre‐planned.

Lurasidone 2016

Study characteristics

Methods

Randomisation: computer‐generated randomisation scheme (1:1 ratio).
Allocation: interactive voice/web response system.
Blinding: double‐blind, identically‐matched placebo.
Duration: 28 weeks.
Design: parallel.
Location: multi‐centre.
Setting: n.i.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), stabilised on study drug (after experiencing an acute exacerbation) for at least 12 weeks before randomisation. 87% paranoid type, 8% undifferentiated type, 4.9% disorganised type.
N = 285.
Gender: 178 men, 107 women.
Age: 42,7 years.
History: duration stable‐ at least 12 weeks, duration ill‐ 17,1 years, number of previous hospitalisations‐ 74% of the participants had at least 1 prior hospitalisation, 50% had four or more prior hospitalisations for schizophrenia, number of psychotic exacerbations in the previous 2 years‐ 1.8, age at onset‐ 23.7 years, severity of illness‐ mean PANSS total score 54.4, mean CGI‐S total score 2.72, baseline antipsychotic dose‐, remission at baseline‐ n.i..

Interventions

1. Drug: Lurasidone. N = 144

Flexible dose. Mean dose: 78.9 mg/day. Allowed dose range: 40 mg/day to 80 mg/day.

2. Placebo: duration of taper: no taper. N = 141.

Rescue medication: anticholinergics (antiparkinson), benzodiazepines (insomnia, anxiety/agitation ‐ with restrictions). Limited use of psychotropic medications (including antipsychotics other than lurasidone) immediately prior to study discontinuation was permitted.

Outcomes

Examined

Relapse (rating scale based and/or clinical judgement and/or need for hospitalisation or increase in the level of psychiatric care).

Leaving the study early (any cause,adverse events, inefficacy).

Service use ‐ Number of participants hospitalised.

Participants´satisfaction with care: Health Economics Exit Questionnaire.

Quality of life: EuroQol 5 Dimensions, Visual Analog Scale (EQ‐VAS).

Social functioning: Specific Levels of Functioning, modified.

Adverse effects.

Death.

Suicidal ideation and behaviour: Columbia Suicide Severity Rating Scale.

Unable to use/Not included

Global state: CGI‐S change scores (no usable data).

Mental state: Positive and Negative Syndrome Scale total score and subscores (no predefined outcomes of interest).

Depressive symptoms: Montgomery Asberg Depression Rating Scale (no predefined outcome of interest).

Compliance to treatment: Brief Adherence Rating Scale (no predefined outcome of interest).

Service use: Health Services Utilization Questionnaire (no usable data).

Quality of life: Short Form‐12v2 Health Survey (SF‐12) (no usable data, available only for the Physical Component score, subscore of the scale. Chose the EuroQol data over these data)

Smoking attitude (no predefined outcome of interest).

Notes

Sponsored by Sunovion, Takeda.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random sequence generation,

Allocation concealment (selection bias)

Low risk

Interactive Voice/Web Response System.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identically‐matched placebo.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identically‐matched placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identically‐matched placebo.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The overall attrition rate (54%) was high, substantially similar between the two groups. Only slightly more participants in the placebo group left the study early due to inefficacy/relapse. The primary efficacy outcome (relapse) was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were all analysed on an ITT basis (MMRM).

Selective reporting (reporting bias)

Unclear risk

Many secondary efficacy outcomes (but not the primary outcome) are reported incompletely so that they could not be entered in a meta‐analysis.

Other bias

Unclear risk

Following the end of the subject’s participation in the study, the PI or an authorized delegate had to report SAEs.

Olanzapine 1999

Study characteristics

Methods

Randomisation: randomly assigned (1:1), no further details.
Allocation: procedure not described.
Blinding: double‐blind, no further details.
Duration: 3 to 5 days.
Design: parallel (optional crossover treatment for relapsed patients).
Location: multi‐centre.
Setting: inpatients and outpatients (numbers not available).

Participants

Diagnosis: schizophrenia (DSM‐IV), received clozapine for a minimum of 4 weeks before entering the study, had to undergo an elective discontinuation of clozapine (49% due to patient inconvenience, 37% due to adverse events, 13% due to partial response).

N = 106.
Gender: 75 men, 31 women.
Age: mean 38.8 years
History: duration stable‐ received clozapine for at least 4 weeks before study entrance, duration ill‐ n.i., mean duration of antipsychotic treatment‐ range from 4 weeks to >1 year (4 weeks to 6 months: N = 39, 6months‐1year: N = 18, >1year: N = 59), number of previous hospitalizations‐ n.i., age at onset‐ n.i., severity of illness‐ mean PANSS total score 64.5 points, baseline antipsychotic dose‐ clozapine 324 mg/day (gradual tapering from baseline dose to 300 mg/day in 2 to 12 days).

Interventions

1. Drug: olanzapine. Fixed dose, 10 mg/day. N = 53.

2. Placebo: inert placebo. duration of taper 2 to 12 days. N = 53.

Rescue medication: only benzodiazepines (for agitation) and anticholinergic (for evident EPS). Patients who relapsed could be removed from the double‐blind treatment and enter and optional open‐label cross‐over treatment (clozapine+olanzapine).

Outcomes

Examined

Relapse: worsening on at least one of the following COSTART events: schizophrenic reaction, hallucinations, delusions, thinking abnormal.

Leaving the study early (any reason, inefficacy)

Unable to use/Not included

Global state: CGI‐S mean change (no usable data).

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

Depressive symptoms: Montgomery Asberg Depression Rating Scale (no predefined outcome of interest).

Performance tests: MiniMental State Examination (no predefined outcome of interest).

Adverse events: no usable data.

Suicidality: no usable data.

Notes

Not explicitly stated, probably sponsored (EliLilly)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomly assigned (1:1), no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

Low risk

The overall attrition rate of 10% was acceptable, but more participants in the placebo group than in the olanzapine group left the study early due to inefficacy. This may be a source of bias for outcomes other than relapse and leaving the study early, but data on such other outcomes are not available. Analyses were all done on an ITT basis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Unclear risk

Very short follow‐up (3 to 5 days, difficult to discriminate between withdrawal symptoms and illness recurrence); 13% of the randomised patients had partial response to clozapine.

Olanzapine 2003

Study characteristics

Methods

Randomisation: randomised, 2:1 ratio, by an interactive voice response system.
Allocation: interactive voice response system.
Blinding: double‐blind, 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 stabilised for 8 weeks before randomisation.

1. Drug: olanzapine ‐ Fixed dose of either 10, 15 mg/day 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.

Death, Suicide attempts.

Violent/aggressive behaviour

Quality of life: Heinrich Carpenter Quality of Life Scale (QLS).

Service use ‐ number of participants hospitalised.

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/continuous 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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 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 LOCF 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.

Paliperidone 2007

Study characteristics

Methods

Randomisation: randomised, computerised randomisation and stratification scheme.
Allocation: interactive voice‐response system.
Blinding: double‐blind, 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‐ 10.8 mg/day paliperidone.

Interventions

1. Drug: paliperidone‐ Flexible doses. Allowed dose range: 3 mg/day to 15 mg/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 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).

Leaving the study early.

Global state ‐ number of participants improved.

Global state ‐ number of participants in remission (CGI‐based).

Service use: number of participants hospitalised.

Quality of life: Schizophrenia Quality‐of‐Life Scale (SQLS)

Social functioning: Personal and Social Performance scale.

Adverse effects.

Violent/aggressive behaviour.

Death; Suicide attempts.

Unable to use/Not included

Mental state: PANSS (no predefined outcome 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, computerised randomisation and stratification scheme.

Allocation concealment (selection bias)

Low risk

Interactive voice‐response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Paliperidone 2014

Study characteristics

Methods

Randomisation: computer‐based randomisation (1:1 ratio).
Allocation: interactive voice/web response system (online IWRS/IVRS)
Blinding: double‐blind, matching placebo.
Duration: terminated early after an interim analysis. Mean duration of exposure in the double‐blind phase: 10 weeks; the longest patient remained in the study for 54 weeks.
Design: parallel.
Location: multi‐centre
Setting: outpatients.

Participants

Diagnosis: schizophrenia (DSM‐IV‐TR), stabilised on paliperidone before double‐blind phase.
N = 136.
Gender: 55 men, 80 women.
Age: 31,7 years.
History: duration stable‐ at least 6 weeks, duration ill‐ at least 1 year, mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ 48% were previously hospitalised, age at onset‐ n.i., severity of illness‐ mean PANSS total score 52,4, mean CGI‐S total score 2,9, baseline antipsychotic dose‐ n.i., remission at baseline‐ 76,5% were in remission at baseline (CGI‐S based).

Interventions

.1. Drug: Paliperidone ER N = 65

Fixed dose. Mean dose: 9.5 mg/day. Allowed dose range: 3 mg/day to 12 mg/day.

2. Placebo: duration of taper: n.i. N = 71.

Rescue medication: benzodiazepines (anxiety, agitation), antiparkinson medication (anticholinergics).

Outcomes

Examined

Relapse (rating scale based and/or clinical judgement and/or need for hospitalisation or increase in the level of psychiatric care)

Leaving the study early (any reason, adverse events, inefficacy).

Global state ‐ number of participants in remission (CGI‐S defined).

Service use ‐ number of patients hospitalised

Social functioning: Personal and Social Performance scale.

Adverse effects

Death

Suicidal ideation and behavior: Columbia Suicide Severity Rating Scale.

Violent/aggressive behaviour

Unable to use/Not included

Global state ‐ number of participants improved (no usable data).

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

Depressive symptoms: PANSS Marder Anxiety/Depression (no predefined outcome of interest).

Subjective sleep measures: Sleep Visual Analog scale (no predefined outcome of interest)

Notes

Sponsored by Janssen Research & Development, LLC.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐based randomisation.

Allocation concealment (selection bias)

Low risk

Interactive web/voice response system.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, matching placebo.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, matching placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, matching placebo.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition rate of 67% was high, and more participants in the placebo group left the study early, mostly due to relapse. This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were analysed on an ITT basis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

Terminated early after an interim analysis, but it was pre‐planned.

Paliperidone depot1M 2010

Study characteristics

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‐blind, 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., remission at baseline: 36% of the patients met remission criteria.

Interventions

1. Drug: paliperidone palmitate depot ‐ Fixed dose: originally 25 mg, 50 mg or 100 mg/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.

Global state ‐ number of participants in sustained remission (Andreasen criteria).

Quality of life: Schizophrenia Quality of Life Scale (SQLS)

Social functioning: Personal and Social Performance scale (PSP).

Suicidal ideation and attempts.

Violent/aggressive behaviour.

Death.

Adverse effects.

Unable to use/Not included

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

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

Overall high dropout 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 is acceptable.

Other bias

High risk

Study was stopped early after an interim analysis.

Paliperidone depot1M 2015

Study characteristics

Methods

Randomisation: sponsor‐prepared computer‐generated randomisation scheme (stratified by absence or presence of mood stabilisers or antidepressants and study centre).
Allocation: central, interactive voice/web response system.
Blinding: double‐blind, matching placebo injections, administered by a person distinct from other study personnel.
Duration: 65 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatients and outpatients (only outpatients during double‐blind phase).

Participants

Diagnosis: schizoaffective disorder (Structured Clinical Interview for DSM‐IV Axis I Disorders).
N = 334.
Gender: 169 men, 165 women.
Age: 38.6 years.
History: duration stable‐ 12 weeks (scale defined, duration of the stabilisation phase, with paliperidone palmitate as monotherapy or adjunctive therapy), duration ill‐ 12.2 years, mean duration of hospitalisation‐ n.i, number of previous hospitalisations‐ mean 3.9, age at onset‐ 30,9 years, severity of illness‐ mean PANSS total score 51.5, mean CGI‐S total score 2.4, baseline antipsychotic dose‐ n.i, remission at baseline‐ yes (97% according to the CGI‐S‐SCA rating system)

Interventions

1. Drug: Paliperidone palmitate depot (1‐month formulation). N = 164

Fixed dose. Mean dose: 114.3 mg eq/month. Allowed dose range: 50 mg to 150 mg eq/month.

2. Placebo: duration of taper: depending on the long elimination half‐life of paliperidone depot. N = 170

Rescue medication: anticholinergics (antiparkinson medication); benzodiazepines not allowed within 8 hours prior to efficacy assessments, further antipsychotics or initiation of antidepressants or mood stabilizers were not allowed.

Outcomes

Examined

Relapse (rating scale based and/or clinical judgement and/or need for hospitalisation or increase in the level of psychiatric care)

Leaving the study early (any reason, inefficacy, adverse events).

Global state ‐ number of participants improved (CGI‐S‐SCA defined, as reported by the authors)

Global state ‐ number of participants in sustained remission (at least 'mildly ill' at CGI‐S‐SCA evaluation).

Patient´s satisfaction with care: Medication Satisfaction Questionnaire.

Service use ‐ number of participants hospitalised: Resource Utilization Questionnaire

Social functioning: Personal and Social Performance Scale.

Number of participants employed

Adverse effects (clinical judgement and rating scale based)

Suicidal ideation: Columbia‐ Suicide Severity Rating Scale.

Violent/aggressive behaviour

Death

Unable to use/Not included

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

Affective symptoms: Hamilton Depression Rating Scale, Young Mania Rating Scale (no predefined outcomes of interest).

Notes

Sponsored by Janssen Research & Development, LLC.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sponsor‐prepared computer‐generated stratified randomisation scheme

Allocation concealment (selection bias)

Low risk

Central, interactive voice/web response system.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, matching placebo injections, administered by a person distinct from other study personnel

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double‐blind, matching placebo injections, administered by a person distinct from other study personnel

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, matching placebo injections, administered by a person distinct from other study personnel

Incomplete outcome data (attrition bias)
All outcomes

High risk

The total attrition rate was high (51%), and differed substantially among study arms (drug arm: 39% versus placebo arm: 61%); almost double participants in the placebo group left the study early due to relapse. This may be a source of bias for outcomes other than dropouts and relapse, which was assessed using the Kaplan Mayer survival´s curve analysis. Data for secondary outcomes were analysed on an ITT basis (LOCF method).

Selective reporting (reporting bias)

Low risk

No clear evidence for selective reporting.

Other bias

Low risk

No clear evidence for bias.

Paliperidone depot3M 2015

Study characteristics

Methods

Randomisation: sponsor‐prepared computer‐generated randomisation scheme, balanced using permuted blocks across the treatment groups and stratified by study centre to ensure balance of treatment allocation within a centre.
Allocation: interactive voice/web response system.
Blinding: double‐blind, identical appearing capsules, administered by a person distinct from other study personnel.
Duration: open‐ended after 66 weeks (longest patient), variable duration of the double‐blind phase (median: 158 days).
Design: parallel.
Location: multi‐centre (64 centres in 8 countries).
Setting: outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), symptomatically stable when enrolled, then stabilised on paliperidone LAI for at least 12 weeks before randomisation.
N = 305.
Gender: 228 men, 77 women.
Age: 37.8 years.
History: duration stable‐ at least 12 weeks (duration of the open‐label maintenance phase), duration ill‐ 10.8 years, mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ 26.9 years, severity of illness‐ mean PANSS total score 54.5, mean CGI‐S total score 2.7, baseline antipsychotic dose‐ paliperidone palmitate 3 months formulation 210 mg eq/3 months, remission at baseline‐ 53% of the participants were remitters at baseline.

Interventions

1. Drug: Paliperidone palmitate depot (3‐month formulation). N = 160

Fixed dose. Mean dose: 402 mg eq/3 months. Allowed dose range: 175 mg to 525 mg eq/3 months.

2. Placebo: duration of taper: depending on the elimination half‐life of paliperidone depot (between 84 and 139 days) N = 145.

Rescue medication: anticholinergics (antiparkinson), beta‐blocker (akathisia), lorazepam (anxiety/agitation), zolpidem (sleep disturbances).

Outcomes

Examined

Relapse (rating scale based and/or clinical judgement and/or need for hospitalisation or increase in the level of psychiatric care)

Leaving the study early (any reason, inefficacy, adverse events).

Social functioning: Personal and Social Performance Scale.

Adverse effects (clinical judgement and rating scale based).

Violent/aggressive behaviour.

Death.

Suicide ideation: Columbia Suicide Severity Rating scale based.

Unable to use/Not included

Global state ‐ number of participants improved (no usable data).

Global state ‐ number of participants in remission (no usable data, only available for a subgroup of patients in remission at baseline, reported as change in remitter status, PANSS defined).

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

Depressive symptoms: PANSS Marder factors (no predefined outcome of interest).

Notes

Sponsored by Janssen Research & Development, LLC.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Sponsor‐prepared computer‐generated randomisation scheme, balanced using permuted blocks across the treatment groups and stratified by study centre to ensure balance of treatment allocation within a centre.

Allocation concealment (selection bias)

Low risk

Interactive voice/web response system.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, identical appearing capsules, administered by a person distinct from other study personnel.

Blinding (performance bias and detection bias)
Subjective outcomes

Low risk

Double‐blind, identical appearing capsules, administered by a person distinct from other study personnel.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical appearing capsules, administered by a person distinct from other study personnel.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The overall attrition rate of 30% could still be acceptable, though higher than 25%, but three times as many participants in the placebo group left the study early due to relapse. This difference may have biased the results of outcomes other than leaving the study early and relapse, which was assessed using the Kaplan‐Meier survival curve analysis. Data for secondary outcomes were analysed on an ITT basis.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting. All the outcomes have been reported in the protocol‐specified way.

Other bias

High risk

Study terminated early after an interim analysis, but this was pre‐planned.

Penfluridol 1970

Study characteristics

Methods

Randomisation: matched pairs were formed and then randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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‐ 23.4 mg/day.

Interventions

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

Leaving the study early

Unable to use/Not included

Global state ‐ number of participants improved (no usable data).

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

Adverse effects: movement disorders (Factor Construct Outcome Scale, no data for randomised phase/continuous 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, indistinguishable placebo.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, indistinguishable placebo.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Penfluridol 1974a

Study characteristics

Methods

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

Participants

Diagnosis: severely ill, chronically hospitalised 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 mg to 160 mg/week penfluridol.

Interventions

1. Drug: penfluridol once weekly. Fixed dose. Allowed dose range: 40 mg to 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 improved (CGI based).

Adverse effects: extrapyramidal side effects.

Unable to use/Not included

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

Behaviour: NOSIE (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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% dropout 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.

Penfluridol 1974b

Study characteristics

Methods

Randomisation: divided into two comparable groups by an unbiased statistician.
Allocation: procedure not explained.
Blinding: double‐blind, 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 40 mg/week. Mean dose: 40 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Penfluridol 1974c

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 43 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Penfluridol 1975

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 stabilisation 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 64.1 mg/week penfluridol.

Interventions

1. Drug: penfluridol ‐ Flexible dose. Allowed dose range: 20 mg to 120 mg/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.

Adverse effects (at least one movement disorder).

Unable to use/Not included

Global state: Clinical Global Impression Scale (no usable data for remission).

Mental state: BPRS (no numbers/no predefined outcomes of interest).

Behaviour: NNOSIE (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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

Penfluridol 1987

Study characteristics

Methods

Randomisation: n.i., but double‐blind study.
Allocation: procedure not described.
Blinding: double‐blind, 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 55 mg/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).

Leaving the study early.

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 (SAS) ‐ 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double, identical capsules.

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.

Perphenazine 1963

Study characteristics

Methods

Randomisation: arbitrarily allocated.
Allocation: procedure not described.
Blinding: double‐blind (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 12 mg three times, one 20 mg three times per day, all other 8 mg three times per day and most less.

Interventions

1. Drug: perphenazine liquid. Fixed dose (same dose as before the start of the study) two 12 mg three times, one 20 mg three times, all other 8mg three times 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.

Violent/aggressive behaviour.

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 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 of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind ‐ 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)
Subjective outcomes

Low risk

Double‐blind ‐ 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‐blind ‐ 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.

Pimozide 1971

Study characteristics

Methods

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

Participants

Diagnosis: chronic schizophrenia (clinical diagnosis), receiving maintenance treatment.
N = 20.
Gender: only male participants.
Age: 42.6 years.
History: duration stable‐n.i., duration ill‐ n.i., mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean BPRS total score 34.6, mean CGI‐S total score 3.73, baseline antipsychotic dose‐ n.i., remission at baseline‐ 40% were in remission at baseline (CGI‐S defined).

Interventions

1. Drug: Pimozide. N = 10

Flexible dose. Mean dose: 40 mg/day.

2. Placebo: duration of taper: abrupt withdrawal. N = 10.

Rescue medication: n.i.

Outcomes

Examined

Leaving the study early.

Global state ‐ number of participants improved (CGI‐I defined).

Global state ‐ number of participants in remission (CGI‐S defined).

Adverse events.

Unable to use/Not included

Mental state: BPRS (no predefined outcome of interest)

Behavior: NOSIE (no predefined outcome of interest).

Notes

Sponsored by McNeil Laboratories.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

Low risk

Only study completers were used in the final analysis, but as there was only one dropout (in the drug arm, before receiving the first dose of medication) this was not necessarily a problem.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

High risk

The pimozide doses (40 mg/day) were very high for current standards.

Pimozide 1973

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 haloperidol)

Leaving the study early.

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

Death.

Violent/aggressive behaviour.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Quetiapine 2007

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 PANSS total score 48.2, baseline antipsychotic dose‐ quetiapine 646 mg/day, remission at baseline ‐ yes (92% of the randomised patients met criteria for symptomatic remission).

Interventions

1. Drug: quetiapine XR. Flexible dose 400 mg to 800 mg/day. Mean dose: 669 mg/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 PANSS 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.

Global state: number of participants in symptomatic remission (Andreasen criteria).

Global state: number of participants in sustained remission (Andreasen criteria, maintained for at least 6 months).

Adverse events: open interviews.

Death.

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

Unable to use/Not included

Global state: number of participants improved (no usable data).

Service use ‐ number of participants hospitalised (unclearly reported).

Mental state: PANSS/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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 occurred 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.

Quetiapine 2009a

Study characteristics

Methods

Randomisation: randomised (1:1:1 ratio) to bifeprunox, quetiapine or placebo, no further detail.
Allocation: procedure not described.
Blinding: double‐blind, encapsulated tablets.
Duration: 12 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatients and outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), in the maintenance phase (no acute exacerbation and medication unchanged for at least 4 weeks).
N = 144.
Gender: 78 men, 66 women.
Age: 39 years.
History: duration stable‐ at least 4 weeks, duration ill‐ n.i., mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean PANSS total score 80.2, mean CGI‐S total score 4,2, baseline antipsychotic dose‐ n.i., remission at baseline‐ not in remission (still experiencing clinically significant symptoms, not sufficiently controlled with medication, at least moderately ill at CGI‐S at baseline.

Interventions

1. Drug: Quetiapine. N = 76

Fixed dose. Mean dose: 600 mg/day.

2. Placebo: duration of taper: 21 days, N = 68.

Rescue medication: n.i.

Outcomes

Examined

Quality of life: Schizophrenia Quality of Life (S‐QoL) scale.

Social functioning: Personal and Social Performance scale.

Death

Unable to use/Not included

Relapse (no usable data)

Leaving the study early (no usable data, available only for the whole study duration)

Global state ‐ number of participants in remission/improved: CGI‐S, CGI‐I (no usable data)

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

Depressive symptoms: Calgary Depression Scale for Schizophrenia (no predefined outcome of interest)

Service use ‐ number of participants hospitalised (no usable data)

Social functioning: Global Assessment of Functioning (PSP data used)

Adverse effects (no usable data)

Notes

Sponsored by Lundbeck.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Only stated randomised, no further detail.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, encapsulated tablets.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, encapsulated tablets.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, encapsulated tablets.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Global and per‐arm attrition rate are not reported for the randomised placebo‐controlled period, but only for the total duration of the study. Efficacy and safety were assessed on a ITT base.

Selective reporting (reporting bias)

Unclear risk

Relapse was not a pre‐specified outcome. several efficacy outcomes cited as assessed but not reported throughout the text.

Other bias

High risk

Terminated early for negative efficacy results after a pooled interim analysis.

Quetiapine 2009b

Study characteristics

Methods

Randomisation: randomised (1:1:1 ratio) to bifeprunox, quetiapine or placebo, no further detail.
Allocation: procedure not described.
Blinding: double‐blind, encapsulated tablets.
Duration: 12 weeks.
Design: parallel.
Location: multi‐centre.
Setting: inpatients and outpatients.

Participants

Diagnosis: Schizophrenia (DSM‐IV‐TR), in the maintenance phase (no acute exacerbation and medication unchanged for at least 4 weeks).
N = 235.
Gender: 127 men, 108 women.
Age: 38 years.
History: duration stable‐ at least 4 weeks, duration ill‐ n.i., mean duration of hospitalisation‐ n.i., number of previous hospitalisations‐ n.i., age at onset‐ n.i., severity of illness‐ mean PANSS total score 79,5, mean CGI‐S total score 4,2, baseline antipsychotic dose‐ n.i., remission at baseline‐ not in remission (still experiencing clinically significant symptoms, not sufficiently controlled with medication, at least moderately ill at CGI‐S at baseline.

Interventions

1. Drug: Quetiapine. N = 116.

Fixed dose. Mean dose: 600 mg/day.

2. Placebo: Duration of taper: 21 days, N = 119.

Rescue medication: n.i.

Outcomes

Examined

Quality of life: Schizophrenia Quality of Life (S‐QoL) scale.

Social functioning: Personal and Social Performance scale.

Death

Unable to use/Not included

Relapse (no usable data)

Leaving the study early (no usable data, available only for the whole study duration)

Global state ‐ number of participants in remission/improved: CGI‐S, CGI‐I (no usable data, available data on "at least much improved")

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

Depressive symptoms: Calgary Depression Scale for Schizophrenia (no predefined outcome of interest)

Service use ‐ number of participants hospitalised (no usable data)

Social functioning: Global Assessment of Functioning (PSP data used)

Adverse effects (no usable data)

Suicide attempts (no usable data).

Notes

Sponsored by Lundbeck.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Only stated randomised, no further details.

Allocation concealment (selection bias)

Unclear risk

Procedure not described.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, encapsulated tablets.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, encapsulated tablets.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, encapsulated tablets.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Global and per‐arm attrition rate are not reported for the randomised placebo‐controlled period, but only for the total duration of the study. Efficacy and safety were assessed on an ITT base.

Selective reporting (reporting bias)

Unclear risk

Relapse was not a pre‐specified outcome. several efficacy outcomes cited as assessed but not reported throughout the text.

Other bias

High risk

Terminated early for negative outcome data after a pooled interim analysis. 8 patients in the quetiapine arm and 11 patients in the placebo arm either continued taking or started concomitant new antipsychotic treatment during the study.

Quetiapine 2010

Study characteristics

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‐centre (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: 80 men, 98 women.
Age: 24.2 years.
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‐ 153 mg/day chlorpromazine equivalents.

Interventions

1. Drug: quetiapine. Fixed dose of 400 mg/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 PANSS 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

Supported by investigator initiated trial award from AstraZeneca.

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 of participants and personnel (performance bias)
All 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)
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.

Trifluoperazine 1969

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 80 mg/day (reached within 35 days). N = 117.

2. Drug: low‐dose trifluoperazine. Fixed dose of 15 mg/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.

Leaving the study early.

Global state: number of participants improved.

Service use; number of participants discharged.

Adverse effects: clinical interview based on 40 items checklist.

Unable to use/Not included

Mental state: Inpatient Multidimensional Psychiatric Scale, BPRS (both only P values/no predefined outcome of interest).

Behaviour: NOSIE (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, 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)
Subjective outcomes

Unclear risk

Double‐blind 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‐blind, 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 (80 mg/day) for current standards, even the low‐dose would nowadays be considered to be quite high (15 mg/day).

Trifluoperazine 1972

Study characteristics

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‐blind, 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 17 mg/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: deterioration of participant’s condition to such a degree that additional antipsychotic medication was necessary.

Leaving the study early.

Death.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo capsules, no further details.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo capsules, no further details.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1960

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, unidentifiable capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, unidentifiable capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1961

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: only the hospital pharmacist had the code on what medication the patient was on.
Blinding: double‐blind, 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 475 mg/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 mg/day 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 mg/day 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

Leaving the study early.

Global state ‐ number of participants improved (clinical judgement, categories comparable to CGI).

Adverse effects: clinical interview, number of participants receiving antiparkinson medication

Unable to use/Not included

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

Personality traits: Minnesota Multiphasic Personality Inventory (MMPI) (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules, each participant had his own container.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules, each participant had his own container.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1962a

Study characteristics

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‐centre.
Setting: inpatients.

Participants

Diagnosis: chronically mentally ill, 67% to 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 phase 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 of participants and personnel (performance bias)
All 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)
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.

Various drugs 1962b

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: psychiatrist without contact to the participants held the key and filled the medication containers.
Blinding: double‐blind, 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 to 50 years.
History: duration stable‐ out of hospital for at least a year (typically 2 to 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 300 mg 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 mg/day to 200 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, exact placebo replicas.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, exact placebo replicas.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1964a

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 hospitalised 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 400 mg/day, thioridazine mean 350 mg/day.

Interventions

1. Drug: chlorpromazine or thioridazine.* Fixed dose, continuation of the dose given in the stabilisation phase. Mean dose: chlorpromazine mean 400 mg/day, thioridazine mean 350 mg/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 (IMPS) (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 of participants and personnel (performance bias)
All outcomes

High risk

Double‐blind, 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)
Subjective outcomes

High risk

Double,‐blind 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‐blind, 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.

Various drugs 1964b

Study characteristics

Methods

Randomisation: randomly assigned.
Allocation: procedure not described.
Blinding: double‐blind ‐ (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 mg/day to 90 mg/day), chlorprothixene (50 mg/day to 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.

Leaving the study early.

Adverse effects.

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, different colours.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, different colours.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, different colours.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts 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.

Various drugs 1966a

Study characteristics

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‐blind, 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 610 mg/day (N = 17), thioridazine mean 480 mg/day (N = 5), trifluoperazine mean 25 mg/day (N = 3), perphenazine 24 mg/day (N = 1), prochlorperazine 60 mg/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: tranquilliser (= benzodiazepine).

Outcomes

Examined

Relapse: worsening by three points on the factor scores of the Inpatient Multidimensional Psychiatric Scale (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.

Service use: number of participants discharged.

Unable to use/Not included

Mental state: IMPS (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Various drugs 1966b

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, "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 mg/day to 600 mg/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, "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)
Subjective outcomes

Unclear risk

Double‐blind, "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‐blind, "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 were not 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.

Various drugs 1968

Study characteristics

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‐blind, 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 to 55 years.
History: duration stable‐ maintenance doses of tranquiliser 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.

Global state: number of participants improved.

Adverse effects.

Unable to use/Not included

Mental state: Wing Scale (no SD/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 of participants and personnel (performance bias)
All outcomes

High risk

Double‐blind, 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)
Subjective outcomes

High risk

Double‐blind, 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‐blind, 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

Blinding was broken when a participant relapsed.

Various drugs 1971

Study characteristics

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‐blind, 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 (PSE)), recently recovered from an acute episode, 32 florid schizophrenia, 3 delusional psychosis.

N = 35.
Gender: n.i..
Age: 16 to 55 years.
History: duration stable‐ n.i., but stabilised at the pre‐admission level during a 6 to 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 mg/day to 25 mg/day, chlorpromazine 100 mg/day to 500 mg/day. Mean dose: chlorpromazine 157.1 mg/day, trifluoperazine 12.3 mg/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.

Service use: number of participants hospitalised.

Adverse effects.

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)

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 of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind, 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)
Subjective outcomes

Low risk

Double‐blind, 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‐blind, 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 dropout rate was 60%, almost all due to relapse which occurred 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.

Various drugs 1974

Study characteristics

Methods

Randomisation: randomised, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 500 mg/day, thioridazine 500 mg/day, fluphenazine 30 mg/day, trifluoperazine 30 mg/day, other equipotent antipsychotics or combinations not exceeding the maximum doses.

Interventions

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

2. Drug: trifluoperazine. Flexible dose. Allowed dose range: 5 mg/day to 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.

Global state: number of participants improved (CGI defined).

Unable to use/Not included

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

Global state: number of participants in remission (no usable data).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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 dropout 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.

Various drugs 1975

Study characteristics

Methods

Randomisation: random, in blocks of eight, stratified for age, duration ill and time since last admission.
Allocation: procedure not described.
Blinding: double‐blind, 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 to 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 mg/day to 20 mg/day. Mean dose: 5.3 mg/day. N = 15.

2. Drug: thioridazine.* Flexible dose. Allowed dose range: 75 mg/day to 750 mg/day. Mean dose: 189 mg/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 the study early.

Global state: number of participants improved (CGI based).

Global state ‐ number of participants in remission (CGI based)

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 (no data available )

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1981a

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 to 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 to 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 mg/day to 150mg/day, perphenazine 12 mg/day to 24 mg/day, chlorpromazine 50 mg/day to 800 mg/day, promazine 200 mg/day to 400 mg/day, trifluoperazine 6 mg/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 deterioration of state or transfer to closed ward)

Leaving the study early.

Global state ‐ number of participants improved

Service use ‐ number of participants hospitalised/discharged.

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 of participants and personnel (performance bias)
All outcomes

High risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

High risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

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

Various drugs 1981b

Study characteristics

Methods

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

Participants

Diagnosis: schizophrenia (mainly Schneiderian first‐rank symptoms), last relapse 30 to 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, SQKS).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Various drugs 1981c

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, placebo sesame oil.
Duration: 26 weeks.
Design: parallel.
Location: single‐centre.
Setting: outpatient.

Participants

Diagnosis: schizophrenia (according to Bleuler’s concept, with at least three primary symptoms ‐ e.g. autism, disturbance of affects, association and volition ‐ and at least two secondary symptoms ‐ hallucinations, persecution‐), 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 21.42 mg fluphenazine/3 weeks or 27.5 mg flupenthixol/three weeks.

Interventions

1. Drug: fluphenazine depot (most around 12.5 mg to 25 mg/3 weeks, mean 21.42 mg/3 weeks) or flupenthixol depot (most around 20 mg to 40 mg/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.

Leaving the study early.

Service use: number of participants hospitalised.

Adverse effects.

Unable to use/Not included

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

Behaviour: NOSIE (no SD/no predefined outcome 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, placebo sesame oil.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo sesame oil.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1982

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, 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 75 mg/day. N = 10.

2. Drug: haloperidol. Fixed dose of 3 mg/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.

Leaving the study early (due to adverse events).

Global state ‐ number of participants in sustained remission (study defined).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, 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‐blind, 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.

Various drugs 1984a

Study characteristics

Methods

Randomisation: randomised, 3:1 ratio.
Allocation: procedure not described.
Blinding: double‐blind, ‘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: 365 mg/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.

Death.

Unable to use/Not included

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

Mental state:BPRS, Profile of Mood Symptoms (PRS) (no data for each group separately/no prespecified outcome of interest).

Adverse effects: extrapyramidal side‐effects (Abnormal Involuntary Movement Scale (AIMS), 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

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

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, ‘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 anyway.

Selective reporting (reporting bias)

Low risk

No evidence for selective reporting.

Other bias

Low risk

No evidence for other bias.

Various drugs 1984b

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not described.
Blinding: double‐blind, drug appearance was made identical with respect to powder, colour, taste 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 mg, 3 mg or 6 mg/day.* N = 37.

2. Drug: propericiazine combined with biperidine and nitrazepam. Fixed dose: 10, 30 mg/day 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.

Global state ‐ number of participants in sustained remission (study defined).

Unable to use/Not included

Prolactin levels (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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, drug appearance was made identical with respect to powder, colour, taste and volume by adding a gastric acid.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, drug appearance was made identical with respect to powder, colour, taste and volume by adding a gastric acid.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, drug appearance was made identical with respect to powder, colour, taste 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.

Various drugs 1986a

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: allocation lists prepared by pharmacy for five antipsychotic drugs mentioned below, concealment is unclear.
Blinding: double‐blind, 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 to 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 IM, chlorpromazine, haloperidol, pimozide, trifluoperazine Flexible dose. Allowed dose range: no upper limit, but lower limit was flupenthixol IM 40 mg/month, chlorpromazine 200 mg/day, haloperidol 3 mg/day, pimozide 4 mg/day, trifluoperazine 5 mg/day. Mean dose: flupenthixol 84 mg/month (N = 31), chlorpromazine 366 mg/day (N = 3), haloperidol 11.8 mg/day (N = 3), pimozide 7.8 mg/day (N = 5), trifluoperazine 11.5 mg/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, anxiolytics.

Outcomes

Examined

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

Leaving the study early.

Unable to use/Not included

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

Global state: clinical judgment of patients global state at endpoint (not usable data/no predefined outcome of interest)

Death, Suicide attempts (no usable data, only reported for the total sample).

Disturbed behaviour/non‐cooperation (no usable data, only reported for the total sample).

Use of antiparkinson medication (unclearly reported, probably referred to the baseline intake).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Various drugs 1986b

Study characteristics

Methods

Randomisation: random, no further details.
Allocation: procedure not explained.
Blinding: double‐blind, 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: BPRS (no clear mean, no number of participants, no SD/no predefined outcome of interest).

Thought disorder: Thought Disorder Index, PRS (all no clear mean’s, no SDs/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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double, placebo matching in kind and dose the previous medication.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, placebo matching in kind and dose the previous medication.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, 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.

Various drugs 1989

Study characteristics

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‐blind, no further details.
Duration: 12 months.
Design: parallel.
Location:  single‐centre.
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 IM 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.

Rehospitalisation.

Unable to use/Not included

Leaving the study early (no data).

Global state ‐ number of participants in remission (no data for withdrawal study).

Social adjustment (no data for withdrawal study).

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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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

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.

Various drugs 1993

Study characteristics

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 100 mg/day chlorpromazine equivalent. Allowed dose range: 100 mg ‐ 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: BPRS total score ‐ >10 increase, GAS < 20 reduction, deterioration Clinical Global Impression Scale CGI >7.

Leaving the study early.

Service use: number of participants hospitalised.

Unable to use/Not included

Global state: CGI (no usable data ).

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

Functioning: Strauss and Carpenter scale, another scale validated by the study group (no usable data)

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

Adverse effects: extrapyramidal side‐effects (AIMS ‐ no SD, SAS, 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 of participants and personnel (performance bias)
All outcomes

High risk

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

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 ITT 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.

Various drugs 2011

Study characteristics

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‐centre.
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‐ 3 mg/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 to 12 weeks. N = 11.

Rescue medication: not indicated.

Outcomes

Examined

Relapse: scale defined or need of hospitalisation for any psychiatric indication.

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 of participants and personnel (performance bias)
All outcomes

High risk

Open study.

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.

Ziprasidone 2002

Study characteristics

Methods

Randomisation: random, computer‐generated randomisation code.
Allocation: drug treatment cards numbered for each subject entering the double‐blind phase; investigator and pharmacist allocated numbers to subjects in strict sequence of entry into the study.
Blinding: double‐blind, identical capsules.
Duration: 12 months.
Design: parallel.
Location: multi‐centre (26 European centres).
Setting: inpatient.

Participants

Diagnosis: chronic, stable schizophrenia (DSM‐III‐R), less than markedly ill on Clinical Global Impression Scale. 56% of the participants had predominantly negative symptoms at baseline.

N = 278 (originally 294 were randomised, but 16 were then excluded from all the analyses due to protocol deviations in one centre).
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, duration of current hospitalisation‐ 68 months, 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 mg/day, 80 mg/day or 160 mg/day.** N = 207 (originally 219 randomised).

2. Placebo: duration of taper < 3 days. N = 71 (originally 75 randomised).

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

Outcomes

Examined

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

Leaving the study early.

Social functioning: Global Assessment of Functioning scale.

Adverse events

Violent/aggressive behaviour.

Death.

Suicidal ideation.

Unable to use/Not included

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

Global state: Clinical Global Impression Severity Scale (no usable data for remission).

Service use ‐ number of participants hospitalised (no usable data, unclearly reported).

Subjective well‐being: own scale (no usable data).

Concept of illness: Concept of illness scale (no predefined outcome of interest).

Adverse effects: extrapyramidal symptoms (SAS, BAS, AIMS ‐ all no SD/continuous 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.

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)

Low risk

Drug treatment cards numbered for each participant entering the double‐blind phase; investigator and pharmacist allocated numbers to participants in strict sequence of entry into the study.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Subjective outcomes

Unclear risk

Double‐blind, identical capsules.

Blinding (performance bias and detection bias)
Objective outcomes

Low risk

Double‐blind, identical capsules.

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 LOCF 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.

Zotepine 2000

Study characteristics

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 300 mg/day which could be reduced once to 150 mg/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).

Leaving the study early.

Global state: number of participants improved (CGI based).

Global state: number of participants in remission (CGI based).

Adverse effects: binary outcomes ‐ open interview.

Suicide ideation

Unable to use/Not included

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

Adverse effects: extrapyramidal side‐effects (SAS, AIMS, no SD/continuous 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 of participants and personnel (performance bias)
All outcomes

Unclear risk

Double‐blind, no further details.

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.

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
LAI: long‐acting injectable
LOCF: last observation carried forward
n.i.: not indicated
SD: standard deviation

Rating scales

AIMS: Abnormal Involuntary Movement Scale
AMDP: Arbeitsgemeinschaft für Methodik und Dokumentation in der Psychiatrie
BAS: Barnes Akathisia Scale
BPRS: Brief Psychiatric Rating Scale
CGI: Clinical Global Impression ‐S: severity, ‐I: improvement
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.

Bai 2003

Allocation: randomised.

Participants: not stabilised on antipsychotic drugs.

Bechdolf 2016

Allocation: randomised.

Participants: including only those at clinical high risk for psychosis (CHR).

Bo 2017

Allocation: randomised.

Participants: clinically stable for at least 4 weeks, treated with risperidone monotherapy at an optimal dose.

Intervention: risperidone (baseline dose), risperidone (gradual dose reduction by 50%), no placebo arm.

Bourin 2008

Allocation: randomised.

Participants: not stabilised on antipsychotic drugs.

Branchey 1981

Allocation: not randomised, matched groups.

Breier 1987

Allocation: not randomised.

Brown 2018

Allocation: randomised.

Participants: mild‐to‐moderate schizophrenia, unclear clinical stability.

Intervention: BI 409306 (inhibitor of phosphodiesterase 9A), not currently approved for schizophrenia, versus placebo.

Cather 2018

Allocation: cluster randomised.

Participants: first episode of non‐affective psychosis,

Intervention: community care compared to NAVIGATE program, which included individual resilience therapy, family education, supported employment and personalised medication management. The specific effect of maintenance therapy with antipsychotic treatment cannot be assumed from this design.

Cheng 2019

Allocation: randomised.

Participants: first episode schizophrenia, stabilised on antipsychotic medication.

Intervention: risperidone versus olanzapine versus aripiprazole; participants failing the initially‐assigned antipsychotic were switched to one of the other two. No real placebo or discontinuation arm.

Chopra 2019

Allocation: randomised,no hint for real maintenance study design.

Participants: first episode schizophrenia; inclusion criteria allow acute patients.

Chouinard 1980

Allocation: not randomised.

Chouinard 1993

Allocation: randomised.

Participants: not clinically stable.

Claghorn 1974

Allocation: randomised.

Participants: schizophrenia.

Intervention: thiothixene alone versus thiothixene plus group therapy versus chlorpromazine alone versus chlorpromazine plus group therapy.

Clark 1967

Allocation: randomised.

Participants: not stabilised on antipsychotic drugs (discontinued medication for at least 6 months before study entry).

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.

Durgam 2016

Allocation: randomised.

Participants: acute exacerbation of schizophrenia.

Engelhardt 1967

Allocation: randomised.

Participants: outpatients with chronic schizophrenia not truly stabilised on antipsychotic drugs.

Fleischhacker 2014

Allocation: randomised.

Participants: schizophrenia, receiving maintenance treatment, stabilised on study drug for at least 8 weeks.

Intervention: aripiprazole LAI (400 mg/4 weeks), aripiprazole oral (10 mg/day to 30 mg/day), aripiprazole LAI suboptimal dose (50 mg/4 weeks), no real placebo arm.

Francey 2018

Allocation: randomised.

Participants: first episode of psychosis, not stable on antipsychotic medication.

Freedman 1982

Allocation: randomised.

Participants: discontinued antipsychotic medication for some weeks, then kept in the study only if showing signs of psychotic exacerbation.

Gallant 1964

Allocation: randomised.

Participants: unclear baseline clinical status and unclear whether they were stabilised on antipsychotic medication.

Intervention: I. butaperazine, trifluoperazine, inert placebo; II. trifluperidol, trifluoperazine, phenobarbital.

Outcome: no predefined outcome of interest.

Gitlin 1988

Allocation: randomised (no further details).

Participants: schizophrenia or schizoaffective disorder, stabilised on the same depot medication for 1 year.

Intervention: fluphenazine decanoate, placebo (cross‐over design).

Outcome: no usable data for relevant outcomes (data up to the point of first cross‐over are not available).

Gitlin 2001

Allocation: randomised.

Participants: schizophrenia or schizoaffective disorder, clinically stable and with stabilised maintenance antipsychotic therapy.

Intervention: fluphenazine decanoate, placebo; cross‐over design.

Outcome: no usable data (data up to the point of first cross‐over are not available).

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 outcomes.

Greenberg 1966

Allocation: randomised.

Participants: patients with chronic schizophrenia.

Intervention: abrupt versus gradual withdrawal of chlorpromazine, but chlorpromazine was withdrawn from both groups. Thus not appropriate control group.

Hine 1958

Allocation: not randomised.

Hirsch 1989

Allocation: randomised.

Participants: schizophrenia, clinically stable for at least 6 months, no florid psychotic symptoms.

Intervention: fluphenazine decanoate, placebo.

Outcome: no usable data for relevant outcomes.

Hirsch 1996

Allocation: randomised (no further details).

Participants: schizophrenia (DSM‐III‐R), clinically stable and receiving maintenance treatment.

Intervention: fluphenazine depot, placebo.

Outcome: no usable data for relevant outcomes (not presented for the randomised subset).

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.

Keefe 2018

Allocation: randomised.

Participants: schizophrenia with relevant negative symptoms, unclear clinical stability.

Intervention: MIN‐101 (roluperidone), not currently approved, versus placebo.

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.

Liu 2018

Allocation: randomised.

Participants: schizophrenia‐related psychotic disorders, under remitted states.

Intervention: maintenance therapy with antipsychotics versus guided dose reduction; no real discontinuation or placebo arm.

Loo 1997

Allocation: randomised.

Participants: participants were not stable, most not on antipsychotics before the study.

Mahal 1975

Allocation: randomised.

Participants: schizophrenia, on maintenance phenotiazine medication

Intervention: pimozide, placebo; cross‐over design.

Outcome: no usable data for relevant outcomes.

Marder 1994

Allocation: randomised.

Participants: not clinically stable.

Mathur 1981

Allocation: randomised.

Participants: chronic schizophrenia, stabilised on antipsychotic treatment for at least 6 months before study entry.

Intervention: chlorpromazine, placebo; cross‐over design.

Outcome: no usable data for relevant outcomes (data up to the point of first cross‐over are not available).

Meehan 2019

Allocation: randomised.

Participants: schizophrenia, not adequately stable.

Mefferd 1958

Allocation: randomised.

Participants: men with schizophrenia.

Intervention: chlorpromazine versus placebo.

Outcome: no usable outcome.

Mosher 1975

Allocation: not randomised.

Müller 1982

Allocation: some of the participants were matched, not randomised.

NCT03559426

Allocation: randomised.

Participants: schizophrenia spectrum disorders, currently on antipsychotic medication.

Intervention: maintenance treatment with antipsychotics versus dose reduction programme; no real discontinuation or placebo arm.

Nishikawa 1989

Allocation: randomised.

Participants: outpatients diagnosed with schizophrenia, in remission at baseline.

Intervention: timiperone, sulpiride, placebo. The placebo arm was only retrospective, derived from data from previous studies.

Oosthuizen 2003

Allocation: randomised.

Participants: first episode of psychosis, not clinically stable.

Pasamanick 1967

Allocation: randomised.

Participants: 152 state hospital patients with schizophrenia (severely impaired at time of enrollment), 29 ambulatory schizophrenia patients (not acutely ill but recruited only if severe enough to warrant hospitalisation).

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.

Ran 2002

Allocation: randomised.

Participants: chronic schizophrenia, unclear clinical status, 30% uncovered

Intervention: antipsychotic therapy + family psychoeducational intervention, antipsychotic treatment alone, control group (in which quote: "medication was not encouraged nor discouraged").

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.

Ruiz 1975

Allocation: randomised.

Participants: chronic schizophrenia, same antipsychotic treatment for at least one month before study entry.

Intervention: pimozide, placebo.

Outcome: no usable data for relevant outcomes (data for the double‐blind phase are not avaiable).

Ruiz Veguilla 2013

Allocation: randomised.

Participants: diagnosed with non‐affective psychosis (first episode), receiving antipsychotic treatment for 12 months since clinical stabilisation, at the same dose for at least 4 months.

Intervention: continual antipsychotic treatment, treatment discontinuation.

Outcome: study not performed (stopped after recruitment of 16 patients), no data available.

Schlossberg 1978

Allocation: randomised.

Participants: not stable.

Singer 1971

Allocation: randomised.

Participants: unclear if clinically stable, probabily not taking antipsychotics before study entry.

Intervention: thiopropazate, placebo; cross‐over design.

Outcome: no usable data for relevant outcomes (data up to the point of first cross‐over are not available).

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 to 20 months before study start.

Stuerup 2017

Allocation: randomised.

Participants: participants with newly diagnosed schizophrenia spectrum disorder, from the outpatient early intervention program (OPUS), meeting remission criteria for at least 3 months before study entry.

Intervention: maintenance treatment with antipsychotics versus tapering/discontinuation; the control arm does not necessarily imply complete discontinuation of antipsychotic medication in all cases.

Sumitomo 2008

Allocation: randomised.

Participants: schizophrenia, not described as clinically stable in inclusion criteria.

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.

Vanover 2018

Allocation: randomised.

Participants: acute exacerbation of schizophrenia.

Weller 2018

Allocation: randomised.

Participants: young people with a first episode of affective/non‐affective psychosis (unclear proportion), meeting remission criteria for at least 3 months.

Intervention: maintenance treatment with antipsychotic drugs versus dose reduction strategy; no real discontinuation or placebo arm.

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 quote: "selected at random to match” the intervention group. We feel that this is not an appropriate method of randomisation.

Zou 2018

Allocation: randomised.

Participants: people with schizophrenia, experiencing an acute episode.

Zwanikken 1973

Allocation: randomised.

Participants: more than 50% had mental retardation, not schizophrenia.

DSM‐III‐R: Diagnostic and Statistical Manual of Mental Disorders, Third Edition‐Revised
LAI: LAI: long‐acting injectable

Characteristics of studies awaiting classification [ordered by study ID]

Ascher‐Svanum 2011

Methods

Allocation: post‐hoc analysis of a 1‐year randomised open‐label trial.

Participants

Diagnosis: schizophrenia.

Interventions

1. Switching of antipsychotic medication

2. Discontinuation of antipsychotic therapy

Outcomes

Change scores on standard efficacy and tolerability measures (no usable data reported).

Notes

The conference abstract did not report any usable data for outcomes of interest in this review. We tried to contact the trials Authors to ask for further data but did not receive any reply.

Decot 2011

Methods

Allocation: the study is described as double‐blind; no details about random sequence generation and allocation concealment procedure.

Cross‐over design.

Participants

Diagnosis: schizophrenia. No details about the baseline clinical status.

Interventions

1. Standard antipsychotics

2. Placebo

Outcomes

Efficacy, analysis based on the COMT Val108/158Met polymorphism (no usable data reported).

Notes

The conference abstract did not report any usable data for outcomes of interest in this review. We tried to contact the trial Authors to ask for further data but did not receive any reply.

Eisenberg 2016

Methods

Allocation: the study is described as blinded; no details about random sequence generation and allocation concealment.

Participants

Diagnosis: schizophrenia or schizoaffective disorder. No details about the baseline clinical status.

Interventions

1. Atypical antipsychotic monotherapy

2. Placebo

Outcomes

Cognitive function, positron emission tomography scanning (no predefined outcomes of interest).

Notes

The conference abstract did not report any usable data for outcomes of interest in this review. We tried to contact the trial authors to ask for further data but did not receive an informative reply.

EUCTR2005‐005499‐34

Methods

Allocation: randomised.

Location: multi‐centre.

Participants

Diagnosis: stable schizophrenia.

Interventions

1, Bifeprunox (not marketed drug)

2. Quetiapine

3. Placebo

Outcomes

Standard efficacy, safety and tolerability measures.

Notes

Prematurely ended, no data available. We were not able to find further information.

Zhang 2006

Methods

Allocation: randomised.

Setting: inpatients.

Participants

Diagnosis: "deteriorated" schizophrenia, on antipsychotic treatment before study entry (no details).

Interventions

1. Standard antipsychotic treatment

2. Discontinuation of antipsychotic treatment

Outcomes

Efficacy on negative symptoms (no predefined outcome of interest).

Notes

The paper did not report any usable data for outcomes of interest in this review. We tried to contact the trial authors to ask for further data but did not receive an informative reply.

Characteristics of ongoing studies [ordered by study ID]

NCT03503318

Study name

A multicenter, randomized, double‐blind, placebo‐controlled study to evaluate the efficacy, safety, and tolerability of risperidone extended‐release injectable suspension (TV‐46000) for subcutaneous use as maintenance treatment in adult and adolescent patients with schizophrenia.

Methods

Randomised controlled trial.

Participants

Individuals diagnosed with schizophrenia, clinically stable and eligible for risperidone treatment.

Interventions

Risperidone ER injectable suspension, subcutaneous injections (two different dose regimens) versus placebo.

Outcomes

Efficacy, safety and tolerability outcomes (time to impending relapse, number maintaining stability, number acheiving remission, number with adverse events).

Starting date

April 2018

Contact information

[email protected]

Notes

NCT03593213

Study name

Clinical trial evaluating the efficacy, safety and tolerability of cariprazine in a dose‐reduction paradigm in the prevention of relapse in patients with schizophrenia.

Methods

Randomised controlled trial.

Participants

Schizophrenia, in maintenance/relapse prevention phase.

Interventions

Cariprazine (two different dose regimens) versus placebo.

Outcomes

Time to impending relapse.

Starting date

July 2018

Contact information

IR‐[email protected]

Notes

NCT03893825

Study name

A study to test if TV‐46000 is safe for maintenance treatment of schizophrenia

Methods

Randomised controlled trial.

Participants

Schizophrenia, in maintenance/relapse prevention phase.

Interventions

TV‐46000 (risperidone extended release injectable suspension) versus matching placebo.

Outcomes

Number of adverse events, dropouts due to adverse events.

Starting date

April 2019

Contact information

[email protected]

Notes

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

1.1 Relapse: 1. Within pre‐specified time periods Show forest plot

71

19996

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.33, 0.40]

Analysis 1.1

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1: Relapse: 1. Within pre‐specified time periods

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1: Relapse: 1. Within pre‐specified time periods

1.1.1 up to 3 months

44

6362

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.28, 0.40]

1.1.2 4‐6 months

49

7599

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.31, 0.42]

1.1.3 7‐12 months

30

4249

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.32, 0.45]

1.1.4 > 12 months

10

1786

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.33, 0.64]

1.2 Relapse: 2. Independent of duration Show forest plot

71

8666

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.30, 0.40]

Analysis 1.2

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2: Relapse: 2. Independent of duration

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2: Relapse: 2. Independent of duration

1.3 Leaving the study early: 1. Due to any reason (acceptability of treatment) Show forest plot

56

7001

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.49, 0.61]

Analysis 1.3

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3: Leaving the study early: 1. Due to any reason (acceptability of treatment)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3: Leaving the study early: 1. Due to any reason (acceptability of treatment)

1.3.1 up to 3 months

11

517

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.17, 0.67]

1.3.2 4 to 6 months

18

1792

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.37, 0.65]

1.3.3 7 to 12 months

24

3951

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.48, 0.65]

1.3.4 > 12 months

5

741

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.51, 0.82]

1.4 Leaving the study early: 2. Due to adverse events (overall tolerability) Show forest plot

53

6627

Risk Ratio (M‐H, Random, 95% CI)

1.27 [0.85, 1.89]

Analysis 1.4

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4: Leaving the study early: 2. Due to adverse events (overall tolerability)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4: Leaving the study early: 2. Due to adverse events (overall tolerability)

1.4.1 up to 3 months

10

371

Risk Ratio (M‐H, Random, 95% CI)

2.84 [0.12, 65.34]

1.4.2 4 to 6 months

15

1852

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.63, 2.28]

1.4.3 7 to 12 months

23

3870

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.69, 1.97]

1.4.4 > 12 months

5

534

Risk Ratio (M‐H, Random, 95% CI)

5.70 [1.28, 25.33]

1.5 Leaving the study early: 3. Due to inefficacy Show forest plot

55

6537

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.32, 0.43]

Analysis 1.5

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5: Leaving the study early: 3. Due to inefficacy

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5: Leaving the study early: 3. Due to inefficacy

1.5.1 up to 3 months

11

421

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.07, 0.64]

1.5.2 4 to 6 months

16

1661

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.31, 0.54]

1.5.3 7 to 12 months

24

3951

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

1.5.4 > 12 months

4

504

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.29, 0.64]

1.6 Global state: number of participants improved (at least minimally) Show forest plot

16

1878

Risk Ratio (M‐H, Random, 95% CI)

2.12 [1.58, 2.85]

Analysis 1.6

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6: Global state: number of participants improved (at least minimally)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6: Global state: number of participants improved (at least minimally)

1.6.1 up to 3 months

3

119

Risk Ratio (M‐H, Random, 95% CI)

4.76 [1.65, 13.68]

1.6.2 4 to 6 months

8

1037

Risk Ratio (M‐H, Random, 95% CI)

2.33 [1.69, 3.21]

1.6.3 7 to 12 months

4

388

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.89, 3.13]

1.6.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.36 [0.88, 2.09]

1.7 Global state: number of participants in symptomatic remission Show forest plot

7

867

Risk Ratio (M‐H, Random, 95% CI)

1.73 [1.20, 2.48]

Analysis 1.7

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7: Global state: number of participants in symptomatic remission

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7: Global state: number of participants in symptomatic remission

1.7.1 up to 3 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

2.50 [0.63, 10.00]

1.7.2 4 to 6 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.79, 3.87]

1.7.3 7 to 12 months

5

807

Risk Ratio (M‐H, Random, 95% CI)

1.70 [1.11, 2.59]

1.8 Global state: number of participants in sustained remission Show forest plot

8

1807

Risk Ratio (M‐H, Random, 95% CI)

1.67 [1.28, 2.19]

Analysis 1.8

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8: Global state: number of participants in sustained remission

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8: Global state: number of participants in sustained remission

1.8.1 7 to 12 months

6

1443

Risk Ratio (M‐H, Random, 95% CI)

1.83 [1.49, 2.25]

1.8.2 >12 months

2

364

Risk Ratio (M‐H, Random, 95% CI)

1.29 [1.13, 1.47]

1.9 Service use: number of participants hospitalised Show forest plot

21

3558

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.32, 0.57]

Analysis 1.9

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9: Service use: number of participants hospitalised

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9: Service use: number of participants hospitalised

1.9.1 up to 3 months

2

55

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.04, 4.06]

1.9.2 4 to 6 months

4

419

Risk Ratio (M‐H, Random, 95% CI)

0.19 [0.03, 1.32]

1.9.3 7 to 12 months

11

2119

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.23, 0.56]

1.9.4 > 12 months

4

965

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.44, 0.69]

1.10 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 1.10

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10: Service use: number of participants discharged

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10: Service use: number of participants discharged

1.10.1 4 to 6 months

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

1.11 Death: due to any reason Show forest plot

25

5181

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.39, 2.11]

Analysis 1.11

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11: Death: due to any reason

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11: Death: due to any reason

1.11.1 up to 3 months

3

415

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.11.2 4 to 6 months

6

1159

Risk Ratio (M‐H, Random, 95% CI)

2.30 [0.59, 8.98]

1.11.3 7 to 12 months

15

3273

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.11, 1.12]

1.11.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

5.18 [0.25, 107.12]

1.12 Death: due to natural causes Show forest plot

25

5226

Risk Ratio (M‐H, Random, 95% CI)

1.35 [0.50, 3.60]

Analysis 1.12

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12: Death: due to natural causes

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12: Death: due to natural causes

1.12.1 up to 3 months

2

379

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.12.2 4 to 6 months

6

1159

Risk Ratio (M‐H, Random, 95% CI)

2.30 [0.59, 8.98]

1.12.3 7 to 12 months

16

3354

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.11, 2.58]

1.12.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

3.11 [0.13, 75.78]

1.13 Death: due to suicide Show forest plot

19

4634

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.12, 2.97]

Analysis 1.13

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13: Death: due to suicide

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13: Death: due to suicide

1.13.1 up to 3 months

3

415

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.13.2 4 to 6 months

3

1033

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.13.3 7 to 12 months

12

2852

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.06, 2.21]

1.13.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

3.11 [0.13, 75.78]

1.14 Number with suicide attempts Show forest plot

12

3123

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.19, 1.99]

Analysis 1.14

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14: Number with suicide attempts

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14: Number with suicide attempts

1.14.1 4 to 6 months

3

776

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.13, 71.51]

1.14.2 7 to 12 months

9

2347

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.13, 1.69]

1.15 Number with suicide ideation Show forest plot

13

3255

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.33, 1.16]

Analysis 1.15

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15: Number with suicide ideation

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15: Number with suicide ideation

1.15.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 3.88]

1.15.2 4 to 6 months

1

386

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.15.3 7 to 12 months

10

2486

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.24, 1.09]

1.15.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.35, 4.74]

1.16 Violent/aggressive behaviour Show forest plot

12

2856

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.24, 0.59]

Analysis 1.16

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16: Violent/aggressive behaviour

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16: Violent/aggressive behaviour

1.16.1 up to 3 months

1

26

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.50]

1.16.2 4 to 6 months

2

350

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.20, 1.08]

1.16.3 7 to 12 months

8

2146

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.19, 0.66]

1.16.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.01, 4.28]

1.17 Adverse effects: at least one adverse event Show forest plot

18

4352

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.98, 1.25]

Analysis 1.17

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17: Adverse effects: at least one adverse event

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17: Adverse effects: at least one adverse event

1.17.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.30, 0.93]

1.17.2 4 to 6 months

4

1079

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.85, 1.12]

1.17.3 7 to 12 months

12

2890

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.99, 1.33]

1.17.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.75 [1.24, 2.45]

1.18 Adverse effects: movement disorders: at least one movement disorder Show forest plot

29

5276

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.25, 1.85]

Analysis 1.18

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18: Adverse effects: movement disorders: at least one movement disorder

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18: Adverse effects: movement disorders: at least one movement disorder

1.18.1 up to 3 months

4

158

Risk Ratio (M‐H, Random, 95% CI)

2.42 [0.70, 8.33]

1.18.2 4 to 6 months

8

1658

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.06, 1.99]

1.18.3 7 to 12 months

16

3126

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.17, 2.05]

1.18.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.58, 2.54]

1.19 Adverse effects: movement disorders: akathisia Show forest plot

21

4214

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.93, 2.38]

Analysis 1.19

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19: Adverse effects: movement disorders: akathisia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19: Adverse effects: movement disorders: akathisia

1.19.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

2.68 [0.49, 14.82]

1.19.2 4 to 6 months

6

1191

Risk Ratio (M‐H, Random, 95% CI)

2.14 [0.50, 9.11]

1.19.3 7 to 12 months

12

2620

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.71, 1.61]

1.19.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.73 [0.42, 7.11]

1.20 Adverse effects: movement disorders: akinesia Show forest plot

3

397

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.08, 3.42]

Analysis 1.20

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20: Adverse effects: movement disorders: akinesia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20: Adverse effects: movement disorders: akinesia

1.20.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

1.20.2 7 to 12 months

2

348

Risk Ratio (M‐H, Random, 95% CI)

0.16 [0.01, 3.98]

1.21 Adverse effects: movement disorders: dyskinesia Show forest plot

18

3200

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.33, 0.91]

Analysis 1.21

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21: Adverse effects: movement disorders: dyskinesia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21: Adverse effects: movement disorders: dyskinesia

1.21.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.50 [0.06, 34.91]

1.21.2 4 to 6 months

3

418

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.84]

1.21.3 7 to 12 months

13

2399

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.37, 1.27]

1.21.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.04, 3.29]

1.22 Adverse effects: movement disorders: dystonia Show forest plot

13

2767

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.99, 2.70]

Analysis 1.22

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22: Adverse effects: movement disorders: dystonia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22: Adverse effects: movement disorders: dystonia

1.22.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

2.50 [0.13, 49.22]

1.22.2 4 to 6 months

2

382

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.29]

1.22.3 7 to 12 months

9

2002

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.65, 4.09]

1.22.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.01, 4.28]

1.23 Adverse effects: movement disorders: rigor Show forest plot

9

922

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.70, 2.79]

Analysis 1.23

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23: Adverse effects: movement disorders: rigor

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23: Adverse effects: movement disorders: rigor

1.23.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.22, 6.62]

1.23.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

1.98 [0.67, 5.85]

1.23.3 7 to 12 months

4

693

Risk Ratio (M‐H, Random, 95% CI)

1.80 [0.29, 11.24]

1.24 Adverse effects: movement disorders: tremor Show forest plot

18

3353

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.95, 1.98]

Analysis 1.24

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24: Adverse effects: movement disorders: tremor

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24: Adverse effects: movement disorders: tremor

1.24.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.46, 3.16]

1.24.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.33, 2.61]

1.24.3 7 to 12 months

12

2790

Risk Ratio (M‐H, Random, 95% CI)

1.62 [1.04, 2.54]

1.24.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.10, 2.79]

1.25 Adverse effects: movement disorders: use of antiparkinson medication Show forest plot

13

2908

Risk Ratio (M‐H, Random, 95% CI)

1.35 [1.10, 1.65]

Analysis 1.25

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25: Adverse effects: movement disorders: use of antiparkinson medication

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25: Adverse effects: movement disorders: use of antiparkinson medication

1.25.1 4 to 6 months

3

841

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.90, 2.61]

1.25.2 7 to 12 months

9

1733

Risk Ratio (M‐H, Random, 95% CI)

1.37 [1.06, 1.78]

1.25.3 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.64, 1.57]

1.26 Adverse effects: sedation Show forest plot

18

4078

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.24, 1.86]

Analysis 1.26

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26: Adverse effects: sedation

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26: Adverse effects: sedation

1.26.1 up to 3 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 3.70]

1.26.2 4 to 6 months

7

1880

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.89, 2.12]

1.26.3 7 to 12 months

9

1844

Risk Ratio (M‐H, Random, 95% CI)

1.78 [1.25, 2.53]

1.26.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.15, 7.27]

1.27 Adverse effects: weight gain Show forest plot

19

4767

Risk Ratio (M‐H, Random, 95% CI)

1.69 [1.21, 2.35]

Analysis 1.27

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27: Adverse effects: weight gain

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27: Adverse effects: weight gain

1.27.1 4 to 6 months

4

1039

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.81, 2.73]

1.27.2 7 to 12 months

14

3394

Risk Ratio (M‐H, Random, 95% CI)

1.80 [1.17, 2.77]

1.27.3 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

2.18 [1.06, 4.48]

1.28 Participant´s satisfaction with care Show forest plot

2

737

Risk Ratio (M‐H, Random, 95% CI)

1.21 [1.10, 1.33]

Analysis 1.28

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28: Participant´s satisfaction with care

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28: Participant´s satisfaction with care

1.28.1 7 to 12 months

1

403

Risk Ratio (M‐H, Random, 95% CI)

1.19 [1.02, 1.38]

1.28.2 > 12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.22 [1.08, 1.38]

1.29 Quality of life (various scales, different timepoints) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.29

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29: Quality of life (various scales, different timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29: Quality of life (various scales, different timepoints)

1.29.1 up to 3 months ‐ Schizophrenia Quality of Life at endpoint (low score=better)

2

379

Mean Difference (IV, Random, 95% CI)

‐2.00 [‐5.80, 1.80]

1.29.2 7 to 12 months ‐ Self‐report Quality of Life Scale change from baseline to endpoint (low score=better)

2

595

Mean Difference (IV, Random, 95% CI)

‐4.10 [‐6.32, ‐1.88]

1.29.3 7 to 12 months ‐ Heinrichs Carpenter Quality of Life Scale change from baseline to endpoint (low score=better)

1

304

Mean Difference (IV, Random, 95% CI)

‐11.36 [‐14.67, ‐8.05]

1.29.4 7 to 12 months ‐ European Quality of Life Visual Analog Scale at endpoint (low score=better)

1

277

Mean Difference (IV, Random, 95% CI)

‐6.30 [‐23.41, 10.81]

1.29.5 > 12 months ‐ Symptom Questionnaire of Kellner and Sheffield at endpoint (low score=better)

1

18

Mean Difference (IV, Random, 95% CI)

‐4.90 [‐14.33, 4.53]

1.30 Quality of life (across all scales and timepoints) Show forest plot

7

1573

Std. Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.57, ‐0.07]

Analysis 1.30

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30: Quality of life (across all scales and timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30: Quality of life (across all scales and timepoints)

1.31 Number of participants in employment Show forest plot

3

593

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.82, 1.41]

Analysis 1.31

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 31: Number of participants in employment

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 31: Number of participants in employment

1.31.1 7 to 12 months

2

259

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.75, 1.23]

1.31.2 > 12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.97, 2.00]

1.32 Social Functioning (various scales, different timepoints) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.32

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 32: Social Functioning (various scales, different timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 32: Social Functioning (various scales, different timepoints)

1.32.1 up to 3 months ‐ Personal and Social Performance at endpoint (low score=better)

2

379

Mean Difference (IV, Random, 95% CI)

‐5.66 [‐11.50, 0.18]

1.32.2 up to 3 months ‐ Global Assessment Scale at endpoint (low score=better)

1

120

Mean Difference (IV, Random, 95% CI)

‐3.61 [‐4.66, ‐2.56]

1.32.3 4 to 6 months ‐ Sheehan Disability Schedule change from baseline to endpoint (low score=better)

1

270

Mean Difference (IV, Random, 95% CI)

‐2.00 [‐3.60, ‐0.40]

1.32.4 7 to 12 months ‐ Personal and Social Performance change from baseline to endpoint (low score=better)

7

1823

Mean Difference (IV, Random, 95% CI)

‐4.92 [‐5.96, ‐3.89]

1.32.5 7 to 12 months ‐ Global Assessment of Functioning at endpoint (low score=better)

1

275

Mean Difference (IV, Random, 95% CI)

‐8.80 [‐13.22, ‐4.38]

1.32.6 7 to 12 months ‐ Specific Levels of Functioning change from baseline to endpoint (low score=better)

1

246

Mean Difference (IV, Random, 95% CI)

‐2.40 [‐4.85, 0.05]

1.32.7 7 to 12 months ‐ Children Global Assessment Scale change from baseline to endpoint (low score=better)

1

146

Mean Difference (IV, Random, 95% CI)

‐4.60 [‐9.84, 0.64]

1.32.8 > 12 months ‐ Personal and Social Performance change from baseline to endpoint (low score=better)

1

329

Mean Difference (IV, Random, 95% CI)

‐3.60 [‐6.76, ‐0.44]

1.33 Social Functioning (across all scales and timepoints) Show forest plot

15

3588

Std. Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.53, ‐0.34]

Analysis 1.33

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 33: Social Functioning (across all scales and timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 33: Social Functioning (across all scales and timepoints)

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

2.1 Subgroup analysis: participants with a first episode Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

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

2.1.1 first episode

8

528

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.38, 0.58]

2.1.2 not first episode

24

3585

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.31, 0.46]

2.2 Subgroup analysis: participants in remission at baseline Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

Analysis 2.2

Comparison 2: Subgroup analysis (relapse at 12 months), Outcome 2: Subgroup analysis: participants in remission at baseline

Comparison 2: Subgroup analysis (relapse at 12 months), Outcome 2: Subgroup analysis: participants in remission at baseline

2.2.1 in remission

10

1050

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.33, 0.60]

2.2.2 not in remission

19

3063

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.30, 0.44]

2.3 Subgroup analysis: various durations of stability before entering the study Show forest plot

24

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

2.3.1 stable at least 1 month

6

574

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.20, 0.50]

2.3.2 stable at least 3 months

10

2250

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.26, 0.43]

2.3.3 stable at least 6 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.69]

2.3.4 stable at least 12 months

5

326

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.57]

2.3.5 stable at least 3 to 6 years

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.18, 0.78]

2.4 Subgroup analysis: abrupt withdrawal versus tapering Show forest plot

29

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

2.4.1 Abrupt withdrawal

18

2348

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.35, 0.53]

2.4.2 Taper

11

1765

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.24, 0.44]

2.5 Subgroup analysis: single antipsychotic drugs Show forest plot

29

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

2.5.1 Chlorpromazine

2

406

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.36, 0.55]

2.5.2 Fluphenazine depot

6

296

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.14, 0.39]

2.5.3 Haloperidol depot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.04, 0.55]

2.5.4 Various, mixed groups of antipsychotic drugs

10

705

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.27, 0.65]

2.5.5 Quetiapine

1

178

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.34, 0.69]

2.5.6 Paliperidone

4

1256

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

2.5.7 Aripiprazole

2

549

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.14, 0.86]

2.5.8 Brexpiprazole

1

202

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.23, 0.56]

2.5.9 Ziprasidone

1

278

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.39, 0.64]

2.5.10 Cariprazine

1

200

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.38, 0.77]

2.6 Subgroup analysis: depot versus oral drugs Show forest plot

26

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

2.6.1 depot

10

1705

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.23, 0.39]

2.6.2 oral

16

2187

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.38, 0.55]

2.7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs Show forest plot

29

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

2.7.1 First‐generation antipsychotic drugs

18

1430

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

2.7.2 Second‐generation antipsychotic drugs

11

2683

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.48]

2.8 Subgroup analysis: appropriate versus unclear allocation concealment Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

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

2.8.1 appropriate allocation concealment

13

2708

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.30, 0.45]

2.8.2 unclear allocation concealment

16

1405

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.30, 0.54]

2.9 Subgroup analysis: blinded versus open trials Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

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

2.9.1 blinded trials

27

3856

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

2.9.2 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

3.1 Exclusion of studies that were not explicitly described as randomised Show forest plot

28

4098

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

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

3.2 Exclusion of non‐double‐blind studies Show forest plot

27

3856

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

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

3.3 Fixed‐effects model Show forest plot

29

4113

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.35, 0.41]

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

3.4 Original authors' assumptions on dropouts Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

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

3.5 Inclusion of only large studies (> 200 participants) Show forest plot

10

2950

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.45]

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)

3.6 Exclusion of studies with clinical diagnosis Show forest plot

22

4054

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.48]

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

3.7 Three months stable Show forest plot

29

4622

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.24, 0.42]

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

3.8 Six months stable Show forest plot

20

2549

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.20, 0.45]

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

3.9 Nine months stable Show forest plot

15

1806

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.19, 0.52]

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

3.10 Exclusion of studies with unclear randomisation method Show forest plot

11

2644

Risk Ratio (IV, Random, 95% CI)

0.36 [0.29, 0.43]

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

3.11 Exclusion of studies with unclear allocation concealment method Show forest plot

13

2708

Risk Ratio (IV, Random, 95% CI)

0.37 [0.30, 0.45]

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 (results of the original search)For the review update in 2018: 3 reports describing the 2 studies originally excluded from quantitative synthesis were moved to excluded studies (no usable data for outcomes of interest); 3 reports on 1 study, originally excluded (short duration of follow‐up), were moved to included studies; one report originally included as independent study was moved as secondary publication of another included study.

Figures and Tables -
Figure 1

Study flow diagram (results of the original search)

For the review update in 2018: 3 reports describing the 2 studies originally excluded from quantitative synthesis were moved to excluded studies (no usable data for outcomes of interest); 3 reports on 1 study, originally excluded (short duration of follow‐up), were moved to included studies; one report originally included as independent study was moved as secondary publication of another included study.

Study flow diagram (results of the 2017/2018/2019 update search and combined results of the original search and the update search)

Figures and Tables -
Figure 2

Study flow diagram (results of the 2017/2018/2019 update search and combined results of the original search and the update search)

Size of trial over time
Figures and Tables -
Figure 3

Size of trial over time

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Figures and Tables -
Figure 4

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Figures and Tables -
Figure 5

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Funnel plot of comparison: 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, outcome: Relapse

Figures and Tables -
Figure 6

Funnel plot of comparison: 1 Maintenance treatment with antipsychotic drugs versus placebo/no treatment, outcome: Relapse

Meta‐regression on duration of clinical stability before study start (relapse at 12 months)The size of the bubbles is proportional to the inverse variance of the treatment effect.

Figures and Tables -
Figure 7

Meta‐regression on duration of clinical stability before study start (relapse at 12 months)

The size of the bubbles is proportional to the inverse variance of the treatment effect.

Meta‐regression on duration of taper in the placebo group (relapse at 12 months)The size of the bubbles is proportional to the inverse variance of treatment effect.

Figures and Tables -
Figure 8

Meta‐regression on duration of taper in the placebo group (relapse at 12 months)

The size of the bubbles is proportional to the inverse variance of treatment effect.

Meta‐regression on mean dose in chlorpromazine equivalents (relapse at 12 months)The size of the bubbles is proportional to the inverse variance of treatment effect.

Figures and Tables -
Figure 9

Meta‐regression on mean dose in chlorpromazine equivalents (relapse at 12 months)

The size of the bubbles is proportional to the inverse variance of treatment effect.

Meta‐regression on study duration (relapse, all studies included).The size of the bubbles is proportional to the inverse variance of treatment effect.

Figures and Tables -
Figure 10

Meta‐regression on study duration (relapse, all studies included).

The size of the bubbles is proportional to the inverse variance of treatment effect.

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1: Relapse: 1. Within pre‐specified time periods

Figures and Tables -
Analysis 1.1

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 1: Relapse: 1. Within pre‐specified time periods

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2: Relapse: 2. Independent of duration

Figures and Tables -
Analysis 1.2

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 2: Relapse: 2. Independent of duration

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3: Leaving the study early: 1. Due to any reason (acceptability of treatment)

Figures and Tables -
Analysis 1.3

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 3: Leaving the study early: 1. Due to any reason (acceptability of treatment)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4: Leaving the study early: 2. Due to adverse events (overall tolerability)

Figures and Tables -
Analysis 1.4

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 4: Leaving the study early: 2. Due to adverse events (overall tolerability)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5: Leaving the study early: 3. Due to inefficacy

Figures and Tables -
Analysis 1.5

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 5: Leaving the study early: 3. Due to inefficacy

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6: Global state: number of participants improved (at least minimally)

Figures and Tables -
Analysis 1.6

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 6: Global state: number of participants improved (at least minimally)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7: Global state: number of participants in symptomatic remission

Figures and Tables -
Analysis 1.7

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 7: Global state: number of participants in symptomatic remission

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8: Global state: number of participants in sustained remission

Figures and Tables -
Analysis 1.8

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 8: Global state: number of participants in sustained remission

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9: Service use: number of participants hospitalised

Figures and Tables -
Analysis 1.9

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 9: Service use: number of participants hospitalised

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10: Service use: number of participants discharged

Figures and Tables -
Analysis 1.10

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 10: Service use: number of participants discharged

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11: Death: due to any reason

Figures and Tables -
Analysis 1.11

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 11: Death: due to any reason

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12: Death: due to natural causes

Figures and Tables -
Analysis 1.12

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 12: Death: due to natural causes

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13: Death: due to suicide

Figures and Tables -
Analysis 1.13

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 13: Death: due to suicide

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14: Number with suicide attempts

Figures and Tables -
Analysis 1.14

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 14: Number with suicide attempts

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15: Number with suicide ideation

Figures and Tables -
Analysis 1.15

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 15: Number with suicide ideation

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16: Violent/aggressive behaviour

Figures and Tables -
Analysis 1.16

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 16: Violent/aggressive behaviour

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17: Adverse effects: at least one adverse event

Figures and Tables -
Analysis 1.17

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 17: Adverse effects: at least one adverse event

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18: Adverse effects: movement disorders: at least one movement disorder

Figures and Tables -
Analysis 1.18

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 18: 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: akathisia

Figures and Tables -
Analysis 1.19

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 19: Adverse effects: movement disorders: akathisia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20: Adverse effects: movement disorders: akinesia

Figures and Tables -
Analysis 1.20

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 20: Adverse effects: movement disorders: akinesia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21: Adverse effects: movement disorders: dyskinesia

Figures and Tables -
Analysis 1.21

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 21: Adverse effects: movement disorders: dyskinesia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22: Adverse effects: movement disorders: dystonia

Figures and Tables -
Analysis 1.22

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 22: Adverse effects: movement disorders: dystonia

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23: Adverse effects: movement disorders: rigor

Figures and Tables -
Analysis 1.23

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 23: Adverse effects: movement disorders: rigor

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24: Adverse effects: movement disorders: tremor

Figures and Tables -
Analysis 1.24

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 24: Adverse effects: movement disorders: tremor

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25: Adverse effects: movement disorders: use of antiparkinson medication

Figures and Tables -
Analysis 1.25

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 25: Adverse effects: movement disorders: use of antiparkinson medication

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26: Adverse effects: sedation

Figures and Tables -
Analysis 1.26

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 26: Adverse effects: sedation

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27: Adverse effects: weight gain

Figures and Tables -
Analysis 1.27

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 27: Adverse effects: weight gain

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28: Participant´s satisfaction with care

Figures and Tables -
Analysis 1.28

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 28: Participant´s satisfaction with care

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29: Quality of life (various scales, different timepoints)

Figures and Tables -
Analysis 1.29

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 29: Quality of life (various scales, different timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30: Quality of life (across all scales and timepoints)

Figures and Tables -
Analysis 1.30

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 30: Quality of life (across all scales and timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 31: Number of participants in employment

Figures and Tables -
Analysis 1.31

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 31: Number of participants in employment

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 32: Social Functioning (various scales, different timepoints)

Figures and Tables -
Analysis 1.32

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 32: Social Functioning (various scales, different timepoints)

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 33: Social Functioning (across all scales and timepoints)

Figures and Tables -
Analysis 1.33

Comparison 1: Maintenance treatment with antipsychotic drugs versus placebo/no treatment, Outcome 33: Social Functioning (across all scales and timepoints)

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 at baseline

Figures and Tables -
Analysis 2.2

Comparison 2: Subgroup analysis (relapse at 12 months), Outcome 2: Subgroup analysis: participants in remission at baseline

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 (specify duration of illness)

Interventions

1. Any antipsychotic drug (flexible dose within appropriate range)

2. Placebo (after gradual ‐ rather than abrupt ‐ withdrawal of the previous antipsychotic drug)

Outcomes

Relapse (primary outcome)

Rehospitalisation for psychosis

Global state (number of participants improved, in symptomatic and sustained remission)

Global state (number of participants in recovery)

Leaving the study early (including specific causes)

Death (natural and unnatural causes)

Violent behaviour

Quality of life

Satisfaction with care and other measures of subjective well‐being/recovery

Side‐effects (well reported)

Social functioning, employment and other measures of functioning

Figures and Tables -
Table 1. Design of a future study
Summary of findings 1. 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: schizophrenia
Setting: inpatients and outpatients
Intervention: maintenance treatment with antipsychotic drugs
Comparison: placebo/no treatment

Outcomes

Illustrative comparative risks (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty 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

606 per 1.000

230 per 1.000
(194 to 273)

RR 0.38
(0.32 to 0.45)

4249
(30 RCTs)

⊕⊕⊕⊕
HIGH1 2 3 4

Leaving the study early: due to any reason (acceptability of treatment)

Follow‐up: 1‐24 months

541 per 1.000

292 per 1.000
(265 to 330)

RR 0.54
(0.49 to 0.61)

7001
(56 RCTs)

⊕⊕⊕⊕
HIGH5 6

Service use: number of participants hospitalised

Follow‐up: 1‐36 months

177 per 1.000

76 per 1.000
(57 to 101)

RR 0.43
(0.32 to 0.57)

3558
(21 RCTs)

⊕⊕⊕⊕
HIGH6 7

Death: due to suicide

Follow‐up: 1‐15 months

1 per 1.000

1 per 1.000
(0 to 4)

RR 0.60
(0.12 to 2.97)

4634
(19 RCTs)

⊕⊕⊝⊝
LOW6 8

Quality of life (various scales; low score=better)

Follow‐up: 3‐18 months

The mean quality of life in the intervention group was 0.32 standard deviations lower (from 0.57 to 0.07 standard deviations lower), with lower scores reflecting a better condition.

1573
(7 RCTs)

⊕⊕⊝⊝
LOW5 6 9 10 11

SMD ‐0.32 (‐0.57 to ‐0.07)

Number of participants in employment

Follow‐up: 9‐15 months

344 per 1.000

372 per 1.000
(282 to 486)

RR 1.08
(0.82 to 1.41)

593
(3 RCTs)

⊕⊕⊝⊝
LOW6 12 13

Social functioning (various scales; low score=better)

Follow‐up: 1‐15 months

The mean social functioning in the intervention group was 0.43 standard deviations lower (from 0.53 to 0.34 standard deviations lower), with lower scores reflecting a better condition.

3588
(15 RCTs)

⊕⊕⊕⊝
MODERATE6 14 15

SMD ‐0.43 (‐0.53 to ‐0.34)

*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: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

1 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 (Analysis 3.5).

2 Risk of bias: rated 'no' ‐ many studies did not report the methods for sequence generation and/or allocation concealment. However, in subgroup analysis (Analysis 2.8) studies reporting high standards of methods showed a similar effect size as compared to studies with unclear methods. Also, in a sensitivity analysis excluding studies with unclear methods (Analysis 3.10 and Analysis 3.11), the effect sizes did not change substantially. Early terminated studies were not judged to contribute substantial weight to this outcome.

3 Inconsistency: rated 'no' ‐ the P value for heterogeneity was statistically significant and the I2 higher than 50%. However, results of individual studies differed rather in magnitude of effect (which could be partly explained by subgroup analyses) rather than in direction of effect. Therefore, this inconsistency does not challenge the overall results.

4 No indirectness was found in terms of study population nor of interventions. In terms of outcome, we followed the original authors definitions of relapse. These definitions used different criteria, but all addressed symptomatic deterioration related to relapse. Therefore, this was not judged to lead to indirectness.

5 Inconsistency: rated 'no' ‐ the P value for heterogeneity was statistically significant and the I‐square higher than 50%. However, results of individual studies differed rather in magnitude of effect than in direction of effect, which was the same in almost all the studies. Therefore, this inconsistency does not challenge the overall results.

6 Publication bias: it is unlikely that a study was unpublished because of unfavourable data in a secondary outcome. As a possible publication bias had no effect on the results for the primary outcome (relapse at 7 to 12 months), we deem that there was no relevant publication bias for this secondary outcome.

7 Indirectness: hospitalisation due to relapse was our primary interest, but in some studies reasons for hospitalisation were unclearly reported. Overall, we do not deem that this uncertainty was an important source of indirectness.

8 Imprecision: rated 'very serious' ‐ only few studies with few events contributed data to this outcome. The CI was wide, ranging from substantial harm to substantial benefit.

9 Risk of bias: rated 'serious' ‐ five out of seven studies were terminated early after interim analyses, possibly leading to overestimation of effect.

10 Indirectness: some rating scales used in the studies have been criticised for eventually not measuring what people understand by quality of life. However, it was decided not to further lower the quality of evidence for this outcome after downgrading for other factors, despite some uncertainty.

11 Imprecise data ‐ only a few studies provided data for this outcome and the confidence interval was large.

12 Indirectness: rated 'serious' ‐ the only three studies included mixed groups of employed and non‐employed participants at baseline, and it is unclear whether employment was supported or competitive employment.

13 Imprecision: rated 'serious' ‐ only three studies contributed to this event which depends on various factors (e.g. the existence of supported employment, rural versus service economy etc).

14 Risk of bias: rated 'serious' ‐ eleven out of fifteen studies were terminated early after interim analyses, possibly leading to overestimation of effects.

15 Indirectness: rated 'no' ‐ different rating scales were used in the studies, but this was not judged to challenge the results.

Figures and Tables -
Summary of findings 1. 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

1.1 Relapse: 1. Within pre‐specified time periods Show forest plot

71

19996

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.33, 0.40]

1.1.1 up to 3 months

44

6362

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.28, 0.40]

1.1.2 4‐6 months

49

7599

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.31, 0.42]

1.1.3 7‐12 months

30

4249

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.32, 0.45]

1.1.4 > 12 months

10

1786

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.33, 0.64]

1.2 Relapse: 2. Independent of duration Show forest plot

71

8666

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.30, 0.40]

1.3 Leaving the study early: 1. Due to any reason (acceptability of treatment) Show forest plot

56

7001

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.49, 0.61]

1.3.1 up to 3 months

11

517

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.17, 0.67]

1.3.2 4 to 6 months

18

1792

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.37, 0.65]

1.3.3 7 to 12 months

24

3951

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.48, 0.65]

1.3.4 > 12 months

5

741

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.51, 0.82]

1.4 Leaving the study early: 2. Due to adverse events (overall tolerability) Show forest plot

53

6627

Risk Ratio (M‐H, Random, 95% CI)

1.27 [0.85, 1.89]

1.4.1 up to 3 months

10

371

Risk Ratio (M‐H, Random, 95% CI)

2.84 [0.12, 65.34]

1.4.2 4 to 6 months

15

1852

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.63, 2.28]

1.4.3 7 to 12 months

23

3870

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.69, 1.97]

1.4.4 > 12 months

5

534

Risk Ratio (M‐H, Random, 95% CI)

5.70 [1.28, 25.33]

1.5 Leaving the study early: 3. Due to inefficacy Show forest plot

55

6537

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.32, 0.43]

1.5.1 up to 3 months

11

421

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.07, 0.64]

1.5.2 4 to 6 months

16

1661

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.31, 0.54]

1.5.3 7 to 12 months

24

3951

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

1.5.4 > 12 months

4

504

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.29, 0.64]

1.6 Global state: number of participants improved (at least minimally) Show forest plot

16

1878

Risk Ratio (M‐H, Random, 95% CI)

2.12 [1.58, 2.85]

1.6.1 up to 3 months

3

119

Risk Ratio (M‐H, Random, 95% CI)

4.76 [1.65, 13.68]

1.6.2 4 to 6 months

8

1037

Risk Ratio (M‐H, Random, 95% CI)

2.33 [1.69, 3.21]

1.6.3 7 to 12 months

4

388

Risk Ratio (M‐H, Random, 95% CI)

1.67 [0.89, 3.13]

1.6.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.36 [0.88, 2.09]

1.7 Global state: number of participants in symptomatic remission Show forest plot

7

867

Risk Ratio (M‐H, Random, 95% CI)

1.73 [1.20, 2.48]

1.7.1 up to 3 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

2.50 [0.63, 10.00]

1.7.2 4 to 6 months

1

40

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.79, 3.87]

1.7.3 7 to 12 months

5

807

Risk Ratio (M‐H, Random, 95% CI)

1.70 [1.11, 2.59]

1.8 Global state: number of participants in sustained remission Show forest plot

8

1807

Risk Ratio (M‐H, Random, 95% CI)

1.67 [1.28, 2.19]

1.8.1 7 to 12 months

6

1443

Risk Ratio (M‐H, Random, 95% CI)

1.83 [1.49, 2.25]

1.8.2 >12 months

2

364

Risk Ratio (M‐H, Random, 95% CI)

1.29 [1.13, 1.47]

1.9 Service use: number of participants hospitalised Show forest plot

21

3558

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.32, 0.57]

1.9.1 up to 3 months

2

55

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.04, 4.06]

1.9.2 4 to 6 months

4

419

Risk Ratio (M‐H, Random, 95% CI)

0.19 [0.03, 1.32]

1.9.3 7 to 12 months

11

2119

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.23, 0.56]

1.9.4 > 12 months

4

965

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.44, 0.69]

1.10 Service use: number of participants discharged Show forest plot

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

1.10.1 4 to 6 months

3

404

Risk Ratio (M‐H, Random, 95% CI)

2.76 [0.69, 11.06]

1.11 Death: due to any reason Show forest plot

25

5181

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.39, 2.11]

1.11.1 up to 3 months

3

415

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.11.2 4 to 6 months

6

1159

Risk Ratio (M‐H, Random, 95% CI)

2.30 [0.59, 8.98]

1.11.3 7 to 12 months

15

3273

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.11, 1.12]

1.11.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

5.18 [0.25, 107.12]

1.12 Death: due to natural causes Show forest plot

25

5226

Risk Ratio (M‐H, Random, 95% CI)

1.35 [0.50, 3.60]

1.12.1 up to 3 months

2

379

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.12.2 4 to 6 months

6

1159

Risk Ratio (M‐H, Random, 95% CI)

2.30 [0.59, 8.98]

1.12.3 7 to 12 months

16

3354

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.11, 2.58]

1.12.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

3.11 [0.13, 75.78]

1.13 Death: due to suicide Show forest plot

19

4634

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.12, 2.97]

1.13.1 up to 3 months

3

415

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.13.2 4 to 6 months

3

1033

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.13.3 7 to 12 months

12

2852

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.06, 2.21]

1.13.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

3.11 [0.13, 75.78]

1.14 Number with suicide attempts Show forest plot

12

3123

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.19, 1.99]

1.14.1 4 to 6 months

3

776

Risk Ratio (M‐H, Random, 95% CI)

3.00 [0.13, 71.51]

1.14.2 7 to 12 months

9

2347

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.13, 1.69]

1.15 Number with suicide ideation Show forest plot

13

3255

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.33, 1.16]

1.15.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 3.88]

1.15.2 4 to 6 months

1

386

Risk Ratio (M‐H, Random, 95% CI)

Not estimable

1.15.3 7 to 12 months

10

2486

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.24, 1.09]

1.15.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.35, 4.74]

1.16 Violent/aggressive behaviour Show forest plot

12

2856

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.24, 0.59]

1.16.1 up to 3 months

1

26

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.50]

1.16.2 4 to 6 months

2

350

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.20, 1.08]

1.16.3 7 to 12 months

8

2146

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.19, 0.66]

1.16.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.01, 4.28]

1.17 Adverse effects: at least one adverse event Show forest plot

18

4352

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.98, 1.25]

1.17.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.30, 0.93]

1.17.2 4 to 6 months

4

1079

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.85, 1.12]

1.17.3 7 to 12 months

12

2890

Risk Ratio (M‐H, Random, 95% CI)

1.15 [0.99, 1.33]

1.17.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.75 [1.24, 2.45]

1.18 Adverse effects: movement disorders: at least one movement disorder Show forest plot

29

5276

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.25, 1.85]

1.18.1 up to 3 months

4

158

Risk Ratio (M‐H, Random, 95% CI)

2.42 [0.70, 8.33]

1.18.2 4 to 6 months

8

1658

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.06, 1.99]

1.18.3 7 to 12 months

16

3126

Risk Ratio (M‐H, Random, 95% CI)

1.55 [1.17, 2.05]

1.18.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.58, 2.54]

1.19 Adverse effects: movement disorders: akathisia Show forest plot

21

4214

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.93, 2.38]

1.19.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

2.68 [0.49, 14.82]

1.19.2 4 to 6 months

6

1191

Risk Ratio (M‐H, Random, 95% CI)

2.14 [0.50, 9.11]

1.19.3 7 to 12 months

12

2620

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.71, 1.61]

1.19.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.73 [0.42, 7.11]

1.20 Adverse effects: movement disorders: akinesia Show forest plot

3

397

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.08, 3.42]

1.20.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.09, 9.92]

1.20.2 7 to 12 months

2

348

Risk Ratio (M‐H, Random, 95% CI)

0.16 [0.01, 3.98]

1.21 Adverse effects: movement disorders: dyskinesia Show forest plot

18

3200

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.33, 0.91]

1.21.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

1.50 [0.06, 34.91]

1.21.2 4 to 6 months

3

418

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.11, 0.84]

1.21.3 7 to 12 months

13

2399

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.37, 1.27]

1.21.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.04, 3.29]

1.22 Adverse effects: movement disorders: dystonia Show forest plot

13

2767

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.99, 2.70]

1.22.1 up to 3 months

1

49

Risk Ratio (M‐H, Random, 95% CI)

2.50 [0.13, 49.22]

1.22.2 4 to 6 months

2

382

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.29]

1.22.3 7 to 12 months

9

2002

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.65, 4.09]

1.22.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.01, 4.28]

1.23 Adverse effects: movement disorders: rigor Show forest plot

9

922

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.70, 2.79]

1.23.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.22, 6.62]

1.23.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

1.98 [0.67, 5.85]

1.23.3 7 to 12 months

4

693

Risk Ratio (M‐H, Random, 95% CI)

1.80 [0.29, 11.24]

1.24 Adverse effects: movement disorders: tremor Show forest plot

18

3353

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.95, 1.98]

1.24.1 up to 3 months

2

69

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.46, 3.16]

1.24.2 4 to 6 months

3

160

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.33, 2.61]

1.24.3 7 to 12 months

12

2790

Risk Ratio (M‐H, Random, 95% CI)

1.62 [1.04, 2.54]

1.24.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.10, 2.79]

1.25 Adverse effects: movement disorders: use of antiparkinson medication Show forest plot

13

2908

Risk Ratio (M‐H, Random, 95% CI)

1.35 [1.10, 1.65]

1.25.1 4 to 6 months

3

841

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.90, 2.61]

1.25.2 7 to 12 months

9

1733

Risk Ratio (M‐H, Random, 95% CI)

1.37 [1.06, 1.78]

1.25.3 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.64, 1.57]

1.26 Adverse effects: sedation Show forest plot

18

4078

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.24, 1.86]

1.26.1 up to 3 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 3.70]

1.26.2 4 to 6 months

7

1880

Risk Ratio (M‐H, Random, 95% CI)

1.37 [0.89, 2.12]

1.26.3 7 to 12 months

9

1844

Risk Ratio (M‐H, Random, 95% CI)

1.78 [1.25, 2.53]

1.26.4 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.15, 7.27]

1.27 Adverse effects: weight gain Show forest plot

19

4767

Risk Ratio (M‐H, Random, 95% CI)

1.69 [1.21, 2.35]

1.27.1 4 to 6 months

4

1039

Risk Ratio (M‐H, Random, 95% CI)

1.49 [0.81, 2.73]

1.27.2 7 to 12 months

14

3394

Risk Ratio (M‐H, Random, 95% CI)

1.80 [1.17, 2.77]

1.27.3 >12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

2.18 [1.06, 4.48]

1.28 Participant´s satisfaction with care Show forest plot

2

737

Risk Ratio (M‐H, Random, 95% CI)

1.21 [1.10, 1.33]

1.28.1 7 to 12 months

1

403

Risk Ratio (M‐H, Random, 95% CI)

1.19 [1.02, 1.38]

1.28.2 > 12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.22 [1.08, 1.38]

1.29 Quality of life (various scales, different timepoints) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.29.1 up to 3 months ‐ Schizophrenia Quality of Life at endpoint (low score=better)

2

379

Mean Difference (IV, Random, 95% CI)

‐2.00 [‐5.80, 1.80]

1.29.2 7 to 12 months ‐ Self‐report Quality of Life Scale change from baseline to endpoint (low score=better)

2

595

Mean Difference (IV, Random, 95% CI)

‐4.10 [‐6.32, ‐1.88]

1.29.3 7 to 12 months ‐ Heinrichs Carpenter Quality of Life Scale change from baseline to endpoint (low score=better)

1

304

Mean Difference (IV, Random, 95% CI)

‐11.36 [‐14.67, ‐8.05]

1.29.4 7 to 12 months ‐ European Quality of Life Visual Analog Scale at endpoint (low score=better)

1

277

Mean Difference (IV, Random, 95% CI)

‐6.30 [‐23.41, 10.81]

1.29.5 > 12 months ‐ Symptom Questionnaire of Kellner and Sheffield at endpoint (low score=better)

1

18

Mean Difference (IV, Random, 95% CI)

‐4.90 [‐14.33, 4.53]

1.30 Quality of life (across all scales and timepoints) Show forest plot

7

1573

Std. Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.57, ‐0.07]

1.31 Number of participants in employment Show forest plot

3

593

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.82, 1.41]

1.31.1 7 to 12 months

2

259

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.75, 1.23]

1.31.2 > 12 months

1

334

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.97, 2.00]

1.32 Social Functioning (various scales, different timepoints) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

1.32.1 up to 3 months ‐ Personal and Social Performance at endpoint (low score=better)

2

379

Mean Difference (IV, Random, 95% CI)

‐5.66 [‐11.50, 0.18]

1.32.2 up to 3 months ‐ Global Assessment Scale at endpoint (low score=better)

1

120

Mean Difference (IV, Random, 95% CI)

‐3.61 [‐4.66, ‐2.56]

1.32.3 4 to 6 months ‐ Sheehan Disability Schedule change from baseline to endpoint (low score=better)

1

270

Mean Difference (IV, Random, 95% CI)

‐2.00 [‐3.60, ‐0.40]

1.32.4 7 to 12 months ‐ Personal and Social Performance change from baseline to endpoint (low score=better)

7

1823

Mean Difference (IV, Random, 95% CI)

‐4.92 [‐5.96, ‐3.89]

1.32.5 7 to 12 months ‐ Global Assessment of Functioning at endpoint (low score=better)

1

275

Mean Difference (IV, Random, 95% CI)

‐8.80 [‐13.22, ‐4.38]

1.32.6 7 to 12 months ‐ Specific Levels of Functioning change from baseline to endpoint (low score=better)

1

246

Mean Difference (IV, Random, 95% CI)

‐2.40 [‐4.85, 0.05]

1.32.7 7 to 12 months ‐ Children Global Assessment Scale change from baseline to endpoint (low score=better)

1

146

Mean Difference (IV, Random, 95% CI)

‐4.60 [‐9.84, 0.64]

1.32.8 > 12 months ‐ Personal and Social Performance change from baseline to endpoint (low score=better)

1

329

Mean Difference (IV, Random, 95% CI)

‐3.60 [‐6.76, ‐0.44]

1.33 Social Functioning (across all scales and timepoints) Show forest plot

15

3588

Std. Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.53, ‐0.34]

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

2.1 Subgroup analysis: participants with a first episode Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

2.1.1 first episode

8

528

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.38, 0.58]

2.1.2 not first episode

24

3585

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.31, 0.46]

2.2 Subgroup analysis: participants in remission at baseline Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

2.2.1 in remission

10

1050

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.33, 0.60]

2.2.2 not in remission

19

3063

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.30, 0.44]

2.3 Subgroup analysis: various durations of stability before entering the study Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.3.1 stable at least 1 month

6

574

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.20, 0.50]

2.3.2 stable at least 3 months

10

2250

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.26, 0.43]

2.3.3 stable at least 6 months

1

20

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.04, 2.69]

2.3.4 stable at least 12 months

5

326

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.17, 0.57]

2.3.5 stable at least 3 to 6 years

2

54

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.18, 0.78]

2.4 Subgroup analysis: abrupt withdrawal versus tapering Show forest plot

29

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.4.1 Abrupt withdrawal

18

2348

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.35, 0.53]

2.4.2 Taper

11

1765

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.24, 0.44]

2.5 Subgroup analysis: single antipsychotic drugs Show forest plot

29

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.5.1 Chlorpromazine

2

406

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.36, 0.55]

2.5.2 Fluphenazine depot

6

296

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.14, 0.39]

2.5.3 Haloperidol depot

1

43

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.04, 0.55]

2.5.4 Various, mixed groups of antipsychotic drugs

10

705

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.27, 0.65]

2.5.5 Quetiapine

1

178

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.34, 0.69]

2.5.6 Paliperidone

4

1256

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.44]

2.5.7 Aripiprazole

2

549

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.14, 0.86]

2.5.8 Brexpiprazole

1

202

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.23, 0.56]

2.5.9 Ziprasidone

1

278

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.39, 0.64]

2.5.10 Cariprazine

1

200

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.38, 0.77]

2.6 Subgroup analysis: depot versus oral drugs Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.6.1 depot

10

1705

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.23, 0.39]

2.6.2 oral

16

2187

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.38, 0.55]

2.7 Subgroup analysis: first‐ versus second‐generation antipsychotic drugs Show forest plot

29

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.7.1 First‐generation antipsychotic drugs

18

1430

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.25, 0.48]

2.7.2 Second‐generation antipsychotic drugs

11

2683

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.48]

2.8 Subgroup analysis: appropriate versus unclear allocation concealment Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

2.8.1 appropriate allocation concealment

13

2708

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.30, 0.45]

2.8.2 unclear allocation concealment

16

1405

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.30, 0.54]

2.9 Subgroup analysis: blinded versus open trials Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

2.9.1 blinded trials

27

3856

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

2.9.2 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

3.1 Exclusion of studies that were not explicitly described as randomised Show forest plot

28

4098

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.33, 0.46]

3.2 Exclusion of non‐double‐blind studies Show forest plot

27

3856

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.33, 0.48]

3.3 Fixed‐effects model Show forest plot

29

4113

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.35, 0.41]

3.4 Original authors' assumptions on dropouts Show forest plot

29

4113

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.32, 0.46]

3.5 Inclusion of only large studies (> 200 participants) Show forest plot

10

2950

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.31, 0.45]

3.6 Exclusion of studies with clinical diagnosis Show forest plot

22

4054

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.34, 0.48]

3.7 Three months stable Show forest plot

29

4622

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.24, 0.42]

3.8 Six months stable Show forest plot

20

2549

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.20, 0.45]

3.9 Nine months stable Show forest plot

15

1806

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.19, 0.52]

3.10 Exclusion of studies with unclear randomisation method Show forest plot

11

2644

Risk Ratio (IV, Random, 95% CI)

0.36 [0.29, 0.43]

3.11 Exclusion of studies with unclear allocation concealment method Show forest plot

13

2708

Risk Ratio (IV, Random, 95% CI)

0.37 [0.30, 0.45]

Figures and Tables -
Comparison 3. Sensitivity analysis (relapse at 12 months)