Scolaris Content Display Scolaris Content Display

افزایش دوز داروی آنتی‌‌سایکوتیک برای عدم پاسخ به درمان در بیماری اسکیزوفرنی

Contraer todo Desplegar todo

Referencias

Bjørndal 1980 {published data only}

Bjørndal N, Bjerre M, Gerlach J, Kristjansen P, Magelund G, Oestrich IH, et al. High dosage haloperidol therapy in chronic schizophrenic patients: a double‐blind study of clinical response, side effects, serum haloperidol, and serum prolactin. Psychopharmacology 1980;67(1):17‐23. CENTRAL

Goff 2013 {published data only}

Goff DC, McEvoy JC, Citrome L, Mech AW, Bustillo JR, Gil R, et al. High‐dose oral ziprasidone versus conventional dosing in schizophrenia patiens with residual symptoms: The ZEBRAS study. Journal of Clinical Psychopharmacology 2013;33(4):485‐90. CENTRAL
NCT00403546. High dose oral ziprasidone versus conventional dosing in schizophrenia patients with residual symptoms. clinicaltrials.gov/show/NCT00403546 (accessed 5 December 2015). CENTRAL

Honer 2011 {published data only}

AstraZeneca. A Canadian, multicentre, double‐blind, randomized, parallel‐group study of the safety, tolerability, and efficacy of treatment with higher doses of quetiapine fumarate (Seroquel) greater than 800mg/day in schizophrenic or schizoaffective subjects. Clinical Study Report CNS.000‐105‐629 30 November 2006. CENTRAL
Honer WG, MacEwan GW, Gendron A, Stip E, Labelle A, Williams R, et al. A randomized, double‐blind, placebo‐controlled study of the safety and tolerability of high‐dose quetiapine in patients with persistent symptoms of schizophrenia or schizoaffective disorder. Journal of Clinical Psychiatry 2012;73(1):13‐20. CENTRAL
Honer WG, MacEwan W, Gendron A, Stip E, Labelle A, Williams R, et al. A double‐blind, placebo‐controlled study of the safety and tolerability of quetiapine 1200mg/d versus 800mg/d in patients with persistent symptoms of schizophrenia or schizoaffective disorder. Schizophrenia Research 2010;117(2‐3):375. CENTRAL
NCT00328978. A Canadian, multicenter, double‐blind, randomized, parallel‐group studyof the safety, tolerability, and efficacy of treatment with higher doses of quetiapine fumarate (Seroquel) greater than 800mg/day in schizophrenic or schizoafective subjects. clinicaltrials.gov/show/NCT00328978 (accessed 5 December 2015). CENTRAL

Huang 1987 {published data only}

Huang CC, Gerhardstein RP, Kim DY, Hollister L. Treatment‐resistant schizophrenia ‐ controlled study of moderate and high dose thiothixene. International Clinical Psychopharmacology 1987;2(1):69‐75. CENTRAL

Kinon 1993 {published data only}

Johns CA, Mayerhoff DI, Lieberman JA, Kane JM. Schizophrenia: alternative neuroleptic strategies. In: Angrist B, Schulz SC editor(s). The Neuroleptic‐Nonresponsive Patient: Characterization and Treatment. Washington, DC, USA: American Psychiatric Press, Inc, 1990:53‐66. CENTRAL
Kinon BJ, Kane JM, Johns C, Perovic R, Ismi M, Koreen A, et al. Treatment of neuroleptic‐resistant schizophrenic relapse. Psychopharmacology Bulletin 1993;29(2):309‐14. CENTRAL

Lindenmayer 2011 {published data only}

Lindenmayer J‐P, Citrome L, Khan A, Kaushik S, Kaushik S. A randomized, double‐blind, parallel‐group, fixed‐dose, clinical trial of quetiapine at 600 versus 1200mg/d for patients with treatment‐resistant schizophrenia or schizoaffective disorder. Journal of Clinical Psychopharmacology 2011;31(April):160‐8. CENTRAL
NCT00297947. A randomized, double‐blind, parallel‐group, fixed dose, clinical trial of quetiapine 600mg/day vs 1200mg/day for patients with treatment‐resistant schizophrenia or schizoaffective disorder. clinicaltrials.gov/show/NCT00297947 2006 (accessed 6 December 2015). CENTRAL

Loebel 2014 {published data only}

Loebel A, Silva R, Goldman R, Watabe K, Cucchiaro J, Kane J. Optimizing Treatment with Lurasidone in Patients with Schizophrenia: Results of a Randomized, Double‐blind, Placebo‐controlled Trial (OPTIMIZE Trial). Neuropsychopharmacology 2014;39(Suppl 1):S473–S647. CENTRAL
Loebel A, Silva R, Goldman R, Watabe K, Pikalov A, Cucchiaro J, et al. Optimizing treatment with lurasidone in patients with schizophrenia: Results of a randomized, double‐blind, placebo‐controlled trial. European Psychiatry 2015;30(Suppl 1):1722. CENTRAL

McEvoy 1991 {published data only}

McEvoy JP, Hogarty GE, Steingard S. Optimal dose of neuroleptic in acute schizophrenia. A controlled study of the neuroleptic threshold and higher haloperidol dose. Archives of General Psychiatry 1991;48(8):739‐45. CENTRAL

McGorry 2011 {published data only}

McGorry PD, Cocks J, Power P, Burnett P, Harrigan S, Lambert T. Very low‐dose risperidone in first‐episode psychosis: a safe and effective way to initiate treatment. Schizophrenia Research and Treatment2011, issue 2011:631690. [DOI: 10.1155/2011/631690]CENTRAL

Sakurai 2016 {published data only}

Sakurai H, Suzuki T, Bies RR, Pollock BG, Mimura M, Kapur S, et al. Increasing Versus Maintaining the Dose of Olanzapine or Risperidone in Schizophrenia Patients Who Did Not Respond to a Modest Dosage: A Double‐Blind Randomized Controlled Trial. Journal of Clinical Psychiatry 2016;77(10):1381‐90. CENTRAL

Agid 2013 {published data only}

Agid O, Schulze L, Arenovich T, Sajeev G, McDonald K, Foussias G, et al. Antipsychotic response in first‐episode schizophrenia: efficacy of high doses and switching. European Neuropsychopharmacology 2013;23(9):1017‐22. CENTRAL

Badgett 1996 {published data only}

Badgett S, Hitzemann RJ, Burr G, Piscani K, Frecska E, Hirschowitz J. Dose reduction in schizophrenia. Proceedings of the 149th Annual Meeting of the American Psychiatric Association. New York, USA, 1996 May 4‐9. CENTRAL

Bai 2002 {published data only}

Bai YM, Lin CC, Yu SC. Risperidone for severe tardive dyskinesia: one year follow up study. International Journal of Neuropsychopharmacology 2002;5(Suppl 1):S165. CENTRAL

Baker 2003 {published data only}

Baker RW, Kinon BJ, Maguire GA, Liu H, Hill AL. Effectiveness of rapid initial dose escalation of up to forty milligrams per day of oral olanzapine in acute agitation. Journal of Clinical Psychopharmacology 2003;23(4):342‐8. CENTRAL

Bastecky 1982 {published data only}

Bastecky J, Kalvach Z, Halkova E. Longterm application of high and medium doses of clorotepin to chronic schizophrenics: an open comparative study. Activitas Nervosa Superior 1982;24(4):230‐2. CENTRAL

Bitter 1989 {published data only}

Bitter I, Volavka J, Cooper J, Scheurer, Camus L, Bakall R. Haloperidol blood levels and clinical effects in schizophrenia. Psychiatry Today: Accomplishments and Promises. Excerpta Medica International Congress Series 1989;899:350. CENTRAL

Bondolfi 1995 {published data only}

Bondolfi G, Baumann P, Patris M, May J, Billeter U, Dufour H, et al. A randomized double‐bind trial of risperidone versus clozapine for treatment‐resistant chronic schizophrenia. European Neuropsychopharmacology 1995;5(3):349. CENTRAL

Branchey 1981 {published data only}

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(5):608‐12. CENTRAL
Branchey MH, Branehey LB, Richardson MA. Effects of gradual decrease and discontinuation of neuroleptics on clinical condition and tardive dyskinesia. Psychopharmacology Bulletin 1981;17(1):118‐20. CENTRAL

Canuso 2010 {published data only}

Canuso CM, Lindenmayer JP, Kosik‐Gonzalez C, Turkoz I, Carothers J, Bossie CA, et al. A randomized, double‐blind, placebo‐controlled study of 2 dose ranges of paliperidone extended‐release in the treatment of subjects with schizoaffective disorder. Journal of Clinical Psychiatry 2010;71(5):587‐98. CENTRAL

Chen 1998 {published data only}

Chen YG, Zhao JP, Xie G. A study on serum concentration and clinical response of clozapine with different dose administration for treatment of schizophrenia. Chinese Journal of Psychiatry 1998;31(2):104‐7. CENTRAL

Clerc 1989 {published data only}

Clerc G. Double‐blind study of amisulpride at different dosages in negative schizophrenic patients. In: Borenstein P, Boyer P, Braconnier A, Carnoy P, Clerc G, Costa e Silva JA, et al. editor(s). Amisulpride. Paris: Expansion Scientifique Francaise, 1989:105‐10. CENTRAL

CN138032 {published data only}

Bristol‐Myers Squibb. A multicenter, randomized, double‐blind study of flexible doses of aripiprazole versus perphenazine in the treatment of patients with treatment‐resistant schizophrenia [Clinical Study Report CN138032]. ctr.bms.com/pdf//CN138‐032%20ST.pdf (accessed 7 December 2015). CENTRAL

Cookson 1987 {published data only}

Cookson IB. The effects of a 50% reduction of cis(z)‐flupenthixol decanoate in chronic schizophrenic patients maintained on a high dose regime. International Clinical Psychopharmacology 1987;2(2):141‐9. CENTRAL

Coryell 1998 {published data only}

Coryell W, Miller DD, Perry PJ. Haloperidol plasma levels and dose optimization. American Journal of Psychiatry 1998;155(1):48‐53. CENTRAL

Daniel 1997 {published data only}

Daniel D, Reeves K, Harrigan EP. The efficacy and safety of ziprasidone 80 mg‐day and 160 mg‐day in schizophrenia and schizoaffective disorder. Schizophrenia Research 1997;24(1‐2):204. CENTRAL

DeBuck 1972 {published data only}

DeBuck RP. Relative safety and efficacy of high and low dose administration of fluphenazine‐hcl to psychotic patients. Proceedings of the 8th International Congress of the C.I.N.P. Copenhagen, Denmark, 1972:265‐72. CENTRAL

de Leon 2007 {published data only}

de Leon J, Diaz FJ, Josiassen RC, Cooper TB, Simpson GM. Weight gain during a double‐blind multidosage clozapine study. Journal of Clinical Psychopharmacology 2007;27(1):22‐7. CENTRAL

Dencker 1978 {published data only}

Dencker SJ, Johansson R, Lundin L, Malm U. High doses of fluphenazine enanthate in schizophrenia. A controlled study. Acta Psychiatrica Scandinavica 1978;57(5):405‐14. CENTRAL

Ericksen 1978 {published data only}

Ericksen SE, Hurt SW, Chang S. Haloperidol dose, plasma levels, and clinical response: a double‐blind study. Psychopharmacology Bulletin 1978;14(2):15‐6. CENTRAL

Gardos 1971 {published data only}

Gardos G, Cole JO, Orzack MH, Weinberger E, McCraith D. Differential effects of high and low doses of thiothixene in schizophrenics. Proceedings of the 5th World Congress of Psychiatry. Ciudad de Mexico, Mexico, 1971:306‐7. CENTRAL

Gulliver 2010 {published data only}

Gulliver A, Sun B, Karagianis JL, Zhao F, Watson SB, McDonnell DP. Dose‐associated changes in safety and efficacy parameters observed in a 24‐week maintenance trial of olanzapine long‐acting injection in patients with schizophrenia. Proceedings of the 163rd Annual Meeting of the American Psychiatric Association. New Orleans, LA, 2010. CENTRAL

Harris 1997 {published data only}

Harris MJ, Paulsen JS, Lacro JoP, Rockwell E, Jeste DV. Neuroleptic maintenance treatment in the elderly. Proceedings of the 150th Annual Meeting of the American Psychiatric Association; 1997 May 17‐22; San Diego, California, USA. 1997. CENTRAL

Hirschowitz 1995 {published data only}

Hirschowitz J, Hitzemann R, Curtis C, Piscani K. Dose reduction in schizophrenia. Proceedings of the 34th Annual Meeting of the American College of Neuropsychopharmacology; 1995 Dec 11‐15; San Juan, Puerto Rico. 1995. CENTRAL

Hirschowitz 1997 {published data only}

Hirschowitz J, Hitzemann R, Piscani K, Burr G, Frecska E, Culliton D, et al. The Dose Reduction in Schizophrenia (DORIS) study: a final report. Schizophrenia Research 1997;23(1):31‐43. CENTRAL

Itil 1970 {published data only}

Itil T. Fluphenazine ‐ Hi versus Lo dose. Psychopharmacology Bulletin 1970;7(2):52‐5. CENTRAL
Itil T, Keskiner A, Heinemann L, Han T, Gannon P, Hsu W. Treatment of resistant schizophrenics with extreme high dosage fluphenazine hydrochloride. Psychosomatics 1970;11(5):456‐63. CENTRAL

Janicak 1997 {published data only}

Janicak PG, Javaid JI, Sharma RP, Leach A, Dowd S, Davis JM. A two‐phase, double‐blind randomized study of three haloperidol plasma levels for acute psychosis with reassignment of initial non‐responders. Acta Psychiatrica Scandinavica 1997;95(4):343‐50. CENTRAL

Kane 1985 {published data only}

Kane JM, Rifkin A, Woerner M, Reardon G, Kreisman D, Blumenthal R, et al. High‐dose versus low‐dose strategies in the treatment of schizophrenia. Psychopharmacology Bulletin 1985;21(3):533‐7. CENTRAL

Lehmann 1980 {published data only}

Lehmann E, Quadbeck H, Tegeler J, Fararuni M, Heinrich K. Drug‐response differences of high and standard dosage of fluphenazine‐decanoate in relation to schizophrenic symptoms. Pharmakopsychiatrie und Neuropsychopharmakologie 1980;13(3):117‐29. CENTRAL

McCreadie 1979 {published data only}

McCreadie RG, Flanagan WL, McKnight J, Jorgensen A. High dose flupenthixol decanoate in chronic schizophrenia. British Journal of Psychiatry 1979;135:175‐9. CENTRAL

Mitchell 2004 {published data only}

Mitchell M, Earley W, Bari M, Riesenberg R, Marquez E, Kurtz D, et al. Preliminary pharmacokinetic and tolerability profiles of olanzapine 20, 30, and 40 mg/day. Schizophrenia Research 2004;67(1):164. CENTRAL
Mitchell M, Riesenberg R, Bari MA, Marquez E, Kurtz D, Falk D, et al. A double‐blind, randomized trial to evaluate the pharmacokinetics and tolerability of 30 or 40 mg/d oral olanzapine relative to 20 mg/d oral olanzapine in stable psychiatric subjects. Clinical Therapeutics 2006;28(6):881‐92. CENTRAL

NCT00539071 {published data only}

NCT00539071. High dose risperidone consta for patients with schizophrenia with unsatisfactory response to standard dose risperidone or long‐acting injectable. clinicaltrials.gov/ct2/show/NCT00539071 (accessed 6 December 2015). CENTRAL

NCT00862992 {published data only}

NCT00862992. Safety, pharmacokinetics and efficacy study of MP‐214 in patients with schizophrenia. clinicaltrials.gov/ct2/show/NCT00862992 (accessed 8 December 2015). CENTRAL

NCT01457339 {published data only}

NCT01457339. Assess the safety and pharmacokinetics of ascending, multiple oral doses of spd489 in adults with schizophrenia. clinicaltrials.gov/ct2/show/NCT01457339 (accessed 8 December 2015). CENTRAL

NCT01569659 {published data only}

NCT01569659. High dose lurasidone for patients with treatment resistant schizophrenia. clinicaltrials.gov/ct2/show/NCT01569659 (accessed 8 December 2015). CENTRAL

Simpson 1999 {published data only}

Simpson GM, Josiassen RC, Stanilla JK, de Leon J, Nair C, Abraham G, et al. Double‐blind study of clozapine dose response in chronic schizophrenia. American Journal of Psychiatry 1999;156(11):1744‐50. CENTRAL

Suzuki 1992 {published data only}

Suzuki E, Kanba S, Nibuya M, Koshikawa H, Nakaki T, Yagi G. Plasma homovanillic acid, plasma anti‐D1 and ‐D2 dopamine‐receptor activity, and negative symptoms in chronically mediated schizophrenia. Biological Psychiatry 1992;31(4):357‐64. CENTRAL

Volavka 1996 {published data only}

Volavka J, Cooper TB, Czobor P, Meisner M. Effect of varying haloperidol plasma levels on negative symptoms in schizophrenia and schizoaffective disorder. Psychopharmacology Bulletin 1996;32(1):75‐9. CENTRAL

Andreasen 1982

Andreasen NC, Olsen S. Negative v positive schizophrenia: Definition and validation. Archives of General Psychiatry 1982;39(7):789‐94.

Baldessarini 1988

Baldessarini RJ, Cohen BM, Teicher MH. Significance of neuroleptic dose and plasma level in the treatment of psychoses. Archives of General Psychiatry 1988;45(1):79‐91.

Barnes 1989

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

Barnes 2003

Barnes TR, Buckley P, Schulz, SC. Treatment‐resistant schizophrenia. In: Hirsch SR, Weinberger D editor(s). Schizophrenia. 2nd Edition. Oxford: Blackwell Science Ltd, 2003:489–516.

Bland 1997

Bland JM. Statistics notes. Trials randomised in clusters. BMJ 1997;315(7108):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 [Apercu sur la problematique des indices d'efficacite therapeutique, 3: comparaison des indices et utilisation. Groupe d'Etude des Indices D'efficacite]. Therapie 1999;54(4):405‐11. [PUBMED: 10667106]

Carpenter 1994

Carpenter WT, Buchanan RW. Schizophrenia. New England Journal of Medicine 1994;330(10):681‐90.

Clark 2011

Clark SL, Adkins DE, van den Oord EJ. Analysis of efficacy and side effects in CATIE demonstrates drug response subgroups and potential for personalized medicine. Schizophrenia Research 2011;132(2‐3):114‐20.

Correll 2011

Correll CU, Cañas F, Larmo I, Levy P, Montes JM, Fagiolini A, et al. Individualizing antipsychotic treatment selection in schizophrenia: characteristics of empirically derived patient subgroups. European Psychiatry 2011;26(Suppl 1):3‐16.

Davey Smith 1998

Davey Smith G, Egger M. Meta‐analysis: Unresolved issues and future developments. BMJ 1998;316(7126):221‐5.

Davis 2004

Davis JM, Chen N. Dose‐response and dose equivalence of antipsychotics. Journal of Clinical Psychopharmacology 2004;24(2):192‐208.

Deeks 2000

Deeks J. Issues in the selection for meta‐analyses of binary data. Proceedings of the 8th International Cochrane Colloquium. Cape Town: The Cochrane Collaboration, 2000.

Deeks 2011

Deeks JJ, Higgins JP, Altman DG. Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JP, Green S, editor (s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐88.

Divine 1992

Divine GW, Brown JT, Frazier LM. The unit of analysis error in studies about physicians' patient care behavior. Journal of General Internal Medicine 1992;7(6):623‐9.

Dold 2015

Dold M, Fugger G, Aigner M, Lanzenberger R, Kasper S. Dose escalation of antipsychotic drugs in schizophrenia: a meta‐analysis of randomized controlled trials. Schizophrenia Research 2015;166(1‐3):187‐93.

Donner 2002

Donner A, Klar N. Issues in the meta‐analysis of cluster randomized trials. Statistics in Medicine 2002;21(19):2971‐80.

DSM‐IV‐TR 1994

American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. Fourth. Washington DC: American Psychiatric Association, 1994:34.

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ 1997;315(7109):629‐34.

Elbourne 2002

Elbourne D, Altman DG, Higgins JP, Curtina F, Worthingtond HV, Vaile A. Meta‐analyses involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140‐9.

Eum 2016

Eum S, Lee AM, Bishop JR. Pharmacogenetic tests for antipsychotic medications: clinical implications and considerations. Dialogues in Clinical Neuroscience 2016;18(3):323‐37.

Friede 2017

Friede T, Röver C, Wandel S, Neuenschwander B. Meta‐analysis of few small studies in orphan diseases. Research Synthesis Methods 2017;8(1):79‐91.

Furukawa 2006

Furukawa TA, Barbui C, Cipriani A, Brambilla P, Watanabe N. Imputing missing standard deviations in meta‐analyses can provide accurate results. Journal of Clinical Epidemiology 2006;59(7):7‐10.

Furukawa 2011

Furukawa TA, Akechi T, Wagenpfeil S, Leucht S. Relative indices of treatment effect may be constant across different definitions of response in schizophrenia trials. Schizophrenia Research 2011;126(1‐3):212‐9.

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.

Gulliford 1999

Gulliford MC. 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(9):876‐83.

Guy 1976

Guy U. ECDEU assessment manual for psychopharmacology. Revised. Rockville, Maryland, USA: National Institute of Mental Health, 1976.

Haase 1961

Haase H. Extrapyramidal modification of fine movements. A “conditio sine qua non” of the fundamental therapeutic action of neuroleptic drugs. Revue Canadienne de Biologie 1961;20:425‐49.

Harvey 1997

Harvey PD, Davidson M, Mueser KT, Parrella M, White L, Powchik P. Social‐Adaptive Functioning Evaluation (SAFE): a rating scale for geriatric psychiatric patients. Schizophrenia Bulletin 1997;23(1):131‐45.

Hasan 2012

Hasan A, Falkai P, Wobrock T, Lieberman J, Glenthoj B, Gattaz WF, et al. World federation of societies of biological psychiatry (WFSBP) guidelines for the biological treatment of schizophrenia, part 1: update 2012 on the acute treatment of schizophrenia and the management of treatment resistance. World Journal of Biological Psychiatry 2012;13(5):318‐78.

Heinrichs 1984

Heinrichs DW, Hanlon TE, Carpenter WT. The Quality of life scale: an instrument for rating the schizophrenic deficit sndrome. Schizophrenia Bulletin 1984;10(3):388‐98.

Hiemke 2011

Hiemke C, Baumann P, Bergemann N, Conca A, Dietmaier O, Egberts K, et al. AGNP consensus guidelines for therapeutic drug monitoring in psychiatry: update 2011. Pharmacopsychiatry 2011;44(6):195‐235.

Higgins 2003

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

Higgins 2011a

Higgins JP, Green S. Chapter 7: Selecting studies and collecting data. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011b

Higgins JP, Altman DG, Sterne JA. Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Honigfeld 1965

Honigfeld G, Klett CJ. The nurses' observation scale for inpatient evaluation: a new scale for measuring improvement in chronic schizophrenia. Journal of Clinical Psychology 1965;21:65‐71.

Honigfeld 1973

Honigfeld G. NOSIE‐30: History and current status of its use in pharmacopsychiatric research. In: P. Pichot, R. Olivier‐Martin editor(s). Modem Problems in Pharmacopsychiatry: Psychological Measurement. Basel: Karger, 1973.

Howes 2017

Howes OD, McCutcheon R, Agid O, de Bartolomeis A, van Beveren NJ, Birnbaum ML, et al. Treatment‐Resistant Schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. American Journal of Psychiatry 2017;174(3):216‐29.

Hutton 2009

Hutton JL. Number needed to treat and number needed to harm are not the best way to report and assess the results of randomised clinical trials. British Journal of Haematology 2009;146(1):27‐30.

IntHout 2015

IntHout J, Ioannidis JP, Borm GF, Goeman JJ. Small studies are more heterogeneous than large ones: a meta‐meta‐analysis. Journal of Clinical Epidemiology 2015;68(8):860‐9.

Jäger 2007

Jäger M, Riedel M, Messer T, Laux G, Pfeiffer H, Naber D, et al. Psychopathological characteristics and treatment response of first episode compared with multiple episode schizophrenic disorders. European Archives of Psychiatry and Clinical Neuroscience 2007;257(1):47‐53.

Kane 1988

Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the treatment‐resistant schizophrenic. A double‐blind comparison with chlorpromazine. Archives of General Psychiatry 1988;45(9):789‐96.

Kane 1996

Kane JM. Pharmacologic treatment of schizophrenia. Biological Psychiatry 1999;46(10):1396–1408.

Kay 1986

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

Kinon 2004

Kinon BJ, Ahl J, Stauffer VL, Hill AL, Buckley PF. Dose Response and Atypical Antipsychotics in Schizophrenia. CNS Drugs 2004;18(9):597‐616.

Lehmann 2004

Lehman AF, Lieberman JA, Dixon LB, McGlashan TH, Miller AL, Perkins DO, et al. Practice guideline for the treatment of patients with schizophrenia, second edition. American Journal of Psychiatry 2004;161(2 Suppl):1‐56.

Leon 2006

Leon AC, Mallinckrodt CH, Chuang‐Stein C, Archibald DG, Archer GE, Chartier K. Attrition in randomized controlled clinical trials: methodological issues in psychopharmacology. Biological Psychiatry 2006;59(11):1001‐5.

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. [PUBMED: 16199797]

Leucht 2005b

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

Leucht 2007

Leucht S, Engel RR, Bauml J, Davis JM. Is the superior efficacy of new generation antipsychotics an artifact of LOCF?. Schizophrenia Bulletin 2007;33(1):183‐91. [PUBMED: 16905632]

Leucht 2009

Leucht S, Corves C, Arbter D, Engel R, Li C, Davis JM. Second‐generation versus first‐generation antipsychotic drugs for schizophrenia: a meta‐analysis. Lancet 2009;373(9657):31‐41.

Leucht 2013

Leucht S, Cipriani A, Spineli L, Mavridis D, 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):951‐62.

Leucht 2014

Leucht S. Measurements of response, remission, and recovery in schizophrenia and examples for their clinical application. Journal of Clinical Psychiatry 2014;75(Suppl 1):8‐14.

Lieberman 2005

Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. New England Journal of Medicine 2005;353(12):1209–23.

Loebel 2013

Loebel A, Cucchiaro J, Sarma K, Xu L, Hsu C, Kalali AH, et al. Efficacy and safety of lurasidone 80 mg/day and 160 mg/day in the treatment of schizophrenia: a randomized, double‐blind, placebo‐ and active‐controlled trial. Schizophrenia Research 2013;145(1‐3):101‐9.

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(5):337‐49.

Miller 2009

Miller R. Mechanisms of Action of Antipsychotic Drugs of Different Classes, Refractoriness to Therapeutic Effects of Classical Neuroleptics, and Individual Variation in Sensitivity to their Actions: PART I. Current Neuropharmacology 2009;7(4):302‐14.

Moher 2001

Moher D, Schulz KF, Altman D. The CONSORT statement: revised recommendations for improving the quality of reports of parallel‐group randomised trials. Journal of the American Medical Association 2001;285(15):1987–91.

Nakajima 2016

Nakajima S, Uchida H, Bies RR, Caravaggio F, Suzuki T, Plitman E, et al. Dopamine D2/3 Receptor Occupancy Following Dose Reduction Is Predictable With Minimal Plasma Antipsychotic Concentrations: An Open‐Label Clinical Trial. Schizophrenia Bulletin 2016;42(1):212‐9.

Nebert 2000

Nebert DW, Dieter MZ. The evolution of drug metabolism. Pharmacology 2000;61(3):124‐35.

Neborsky 1981

Neborsky R, Janowsky D, Munson E, Depry D. Rapid treatment of acute psychotic symptoms with high‐ and low‐dose haloperidol. Behavioral considerations. Archives of General Psychiatry 1981;38(2):195‐9.

NIMH 1970

National Institute of Mental Health. Clinical global impression. In: Guy W, Bonato RR editor(s). Manual for the ECDEU Assessment Battery. Washington DC: NIMH, 1970.

Overall 1962

Overall JE, Gorham DR. The brief psychiatric rating scale. Psychological Reports 1962;10:799‐812.

Palmer 2005

Palmer BA, Pankratz VS, Bostwick JM. The lifetime risk of suicide in schizophrenia: a reexamination. Archives of General Psychiatry 2005;62(3):247‐53.

Samara 2015a

Samara MT, Leucht C, Leeflang MM, Anghelescu IG, Chung YC, Crespo‐Facorro B, et al. Early Improvement As a Predictor of Later Response to Antipsychotics in Schizophrenia: A Diagnostic Test Review. American Journal of Psychiatry 2015;172(7):617‐29.

Samara 2015b

Samara MT, Helfer B, Rothe PB, Leucht S. Increasing antipsychotic dose versus switching antipsychotic for non response in schizophrenia. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD011884]

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JP, Deeks JJ, Glasziou P, et al. Chapter 12: Interpreting results and drawing conclusions. In Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Simpson 1970

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

Sterne 2011

Sterne JA, Egger M, Moher D. Chapter 10: Addressing reporting biases. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Intervention. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Swift 2002

Swift RH, Harrigan EP, Cappelleri JC, Kramer D, Chandler LP. Validation of the behavioural activity rating scale (BARS): a novel measure of activity in agitated patients. Journal of Psychiatric Research 2002;36(2):87‐95.

Ukoumunne 1999

Ukoumunne OC, Gulliford MC, Chinn S, Sterne JA, Burney PG. Methods for evaluating area‐wide and organistation‐based intervention in health and health care: a systematic review. Health Technology Assessment 1999;3(5):1‐75.

Vijayan 2012

Vijayan NN, Mathew A, Balan S, Natarajan C, Nair CM, Allencherry PM, et al. Antipsychotic drug dosage and therapeutic response in schizophrenia is influenced by ABCB1 genotypes: a study from a south Indian perspective. Pharmacogenomics 2012;13(10):1119‐27.

von Hippel 2015

von Hippel PT. The heterogeneity statistic I2 can be biased in small meta‐analyses. BMC Medical Research Methodology 2015;15(1):35.

Vos 2012

Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990‐2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012;380(9859):2163‐96.

Xia 2010

Xia J, Adams CE, Bhagat N, Bhagat V, Bhoopathi P, El‐Sayeh H, et al. Loss to outcomes stakeholder survey: the LOSS study. Schizophrenia Research 2010;117(2‐3):278.

Helfer 2015

Helfer B, Leucht S, Rothe PH, Samara MT. Increasing antipsychotic dose for non response in schizophrenia. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD011883]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Bjørndal 1980

Methods

Allocation: randomised, no further details
Blinding: double, no further details
Duration: 16 weeks; 4 weeks non‐randomised open‐label run‐in phase and 12 weeks randomised double‐blind phase
Design: parallel
Location: not indicated, probably single centre, Denmark
Setting: inpatients

Participants

Diagnosis: chronic schizophrenia, severity of illness was 4 to 6 according to the disability scale used (0 indicated no symptoms, while a score of 7 indicated extremely severe degree of illness), no further details.
N = 29, data only on 23 completers
Gender: 29 men, 0 women
Age: mean 34 years (range 19 to 60 years) for 23 completers
History: duration of illness mean 9 years (range 2 to 20), length of stay in hospital mean 5 years (range 0.5 to 17), duration of antipsychotic treatment mean 8 years (range 1 to 18) for 23 completers; no further details.

Interventions

All participants firstly received open‐label 2 mg haloperidol tablets for 4 weeks (maximum dosage being 12 mg/day). Participants were then randomised to either:

1. dose increase: 20 mg haloperidol tablets; mean 103 mg/day at endpoint. N = 12; or

2. dose maintenance: 2 mg haloperidol tablets; mean 15 mg/day at endpoint. N = 11.

The starting dose corresponded to the number of tablets reached during the open pretest period (mean 10 mg/day in both groups). During the first 6 weeks, the dose was adjusted according to effect and side effects, while during the following 6 weeks the dose was maintained constant as far as possible.

Rescue medication: orphenadrine and chloralodol; no further details.

Outcomes

Clinically relevant response (defined as > 0% BPRS total score reduction)

Global state: improved, possible improved, unchanged, or deteriorated (Global Assessment Scale)

Adverse effects

Unable to use:

Leaving the study early (no separate numbers for the two groups)

Mental state: general (BPRS total score, no mean, no SD), Nurses Observation Scale for Inpatient Evaluation (NOSIE total score, no SD)

Plasma levels of haloperidol (no mean, no SD) and prolactin (no SD), and not protocol outcomes

Other laboratory investigations (no mean, no SD), and not protocol outcomes

Notes

In this study, there was a 4‐week, open, pretest period during which participants received 2 mg haloperidol tablets (mean 10 mg/day). It is not clearly indicated whether all participants entering the double‐blind phase were non‐responders during the run‐in phase. Nevertheless, based on participant description and BPRS total scores at baseline, we assumed this is the case.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...patients were randomized..." (pg. 18); no further details

Allocation concealment (selection bias)

Unclear risk

No details were presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...12‐week double‐blind phase..." (pg. 18); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...12‐week double‐blind phase..." (pg. 18); no further details

Incomplete outcome data (attrition bias)
All outcomes

High risk

Quote: "Twenty‐nine male, chronic schizophrenic patients ... consent to participate in the study. Six of the patients dropped out during the study, independent of the drug administered. The 23 patients completing the study ..." (pg. 18); no further details. Only completers were analysed.

Selective reporting (reporting bias)

High risk

BPRS total score, NOSIE, plasma levels and other laboratory values were assessed but no usable data were reported in the results.

Other bias

Low risk

No obvious risk for other bias

Goff 2013

Methods

Allocation: randomised, no further details
Blinding: double, no further details
Duration: 11 weeks; 3 weeks non‐randomised open‐label run‐in phase and 8 weeks randomised double‐blind phase.
Design: parallel
Location: multi‐centre (USA)
Setting: inpatients (N = 30) and outpatients (N = 45)

Participants

Diagnosis: schizophrenia or schizoaffective disorder (DSM‐IV)
N = 75
Gender: 52 men, 23 women
Age: mean 40 years (SD = 11.9), range 16 to 65 years
History: age at onset mean 25 years (SD = 9.1), no further details

Interventions

All participants firstly received open‐label ziprasidone treatment, titrated up to 160 mg/day, for a minimum of 3 weeks. Participants with persistent psychotic symptoms defined by a score of 4 (moderate) or greater on any item of PANSS despite ziprasidone treatment were then randomised to either:

1. dose increase: ziprasidone 320 mg/day; dose could be decreased to 240 mg/day. N = 38; or

2. dose maintenance: ziprasidone 160 mg/day. N = 37.

Rescue medication: benztropine, propranolol, lorazepam, zolpidem; no further details.

Outcomes

Global state: clinically relevant response (defined as ≥20% PANSS total score reduction)

Mental state: positive symptoms (PANSS positive subscore)

Adverse effects: cardiac — QTc prolongation (number of participants with QTc longer than 500 msec)

Unable to use:

Leaving the study early (numbers not presented)

Overall mental state (PANSS total score, no mean, no SD)

Global state (change in CGI‐I, no SD; and CGI‐S, no mean, no SD)

Negative symptoms (PANSS negative subscore, no mean, no SD)

Depression (Calgary Depression Rating Scale (CDRS), no mean, no SD)

Functioning (Global Assessment of Functioning, GAF, no mean, no SD)

Adverse effects (EPS ‐ SAS (no mean, no SD), Akathisia ‐ BAS (no mean, no SD), tardive dyskinesia ‐ AIMS (no mean, no SD), Side Effect Checklist (no numbers), rate of adverse effects (no SD))

Vital signs (no numbers)

Plasma levels (no SD)

Cognition (Schizophrenia Cognition Rating Scale, no mean, no SD)

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...eligible patients were randomly assigned to ziprasidone 40 mg capsules or matching placebo in a 1:1 ratio stratified according to the duration of prior ziprasidone treatment..." (pg. 486); no further details.

Allocation concealment (selection bias)

Unclear risk

No details were presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: " For the 8‐week double‐blind, placebo‐controlled trial..." (pg. 486); no further details.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: " For the 8‐week double‐blind, placebo‐controlled trial..." (pg. 486); no further details.

Incomplete outcome data (attrition bias)
All outcomes

High risk

44% of the participants left the study early. Numbers per group as well as reasons for leaving the study early were not specified. For the primary outcome of clinically relevant response, only completers were analysed.

Selective reporting (reporting bias)

High risk

PANSS total score, PANSS negative sbscore, CGI, CDRS, SCoRS, SAS, BAS, AIMS, Side Effect Checklist, vital signs and plasma levels were assessed but no usable data were reported in the results.

Other bias

Low risk

No obvious risk for other bias

Honer 2011

Methods

Allocation: randomised, 2:1 with a computerised schedule
Blinding: double, no further details
Duration: 12 weeks; 4 weeks non‐randomised open‐label run in phase and 8 weeks randomised double‐blind phase.
Design: parallel
Location: multi‐centre (Canada)
Setting: inpatients (N = 27) and outpatients (N = 104)

Participants

Diagnosis: schizophrenia or schizoaffective disorder (DSM IV)
N = 131
Gender: 90 men, 41 women
Age: mean 39.7 years (SD = 12.1), range 18 to 65 years
History: not stated

Interventions

All participants firstly received open‐label quetiapine treatment 800 mg/day for 4 weeks. Participants with persistent positive and negative symptoms and CGI≥4 despite quetiapine treatment were then randomised to either:

1. dose increase: quetiapine 1200 mg/day; dose could be decreased. N = 88; or

2. dose maintenance: quetiapine 800 mg/day. N = 43.

Rescue medication: flurazepam, zaleplon, lorazepam, anticholinergic medication.
Antidepressant, mood‐stabilizing, or hypnotic medications were continued, if subjects were taking stable doses for a 30‐day period prior to trial entry.

Outcomes

Global state: clinically relevant response (defined as ≥ 20% PANSS total score reduction)

Leaving the study early (due to side effects, any reason, inefficacy)

Mental state: general mental state (PANSS total score), positive symptoms (PANSS positive subscore), negative symptoms (PANSS negative subscore)

Global state (CGI‐Severity)

Adverse effects: (at least one adverse effect, SAS (improved, no change, worsened), BAS (improved, no change, worsened), AIMS (improved, no change, worsened), death/suicide, BMI & weight increase, increase in heart rate, QTcF prolongation (≥450 ms), dizziness, headache, fatigue, somnolence, anxiety, dyskinesia, tremor).

Functioning (SOFAS)

Unable to use:

Plasma levels, not a protocol outcome

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...randomised (2:1, with a computerized schedule) to supplementation with quetiapine or with placebo. The person who generated the randomization schedule was not involved in determining subject eligibility, administering treatment, or determining outcome." (pg. 14).

Allocation concealment (selection bias)

Low risk

Quote: "The person who generated the randomization schedule was not involved in determining subject eligibility, administering treatment, or determining outcome." (pg. 14).

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...double‐blind, placebo‐controlled trial..." (pg. 13); no further details.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...double‐blind, placebo‐controlled trial..." (pg. 13); no further details.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

21.3% of the participants left the study early. Reasons for leaving the study early were described. An ITT approach was used.

Selective reporting (reporting bias)

Low risk

Free from selective reporting

Other bias

Low risk

No obvious risk for other bias

Huang 1987

Methods

Randomisation: randomised, no further details
Blinding: double, no further details
Duration: 11 weeks; 2 weeks non‐randomised single‐blind run in phase and 9 weeks randomised double‐blind phase.
Design: parallel
Location: single‐centre (USA)
Setting: inpatients

Participants

Diagnosis: schizophrenia (DSM III); treatment resistant.
N = 50
Gender: not indicated
Age: 21 to 55 years
History: mentally ill for 2 years or more; treated with 2 or more antipsychotics at usual therapeutic doses (equivalent to thiothixene 60 mg/day) for 6 months or more without appreciable remission.

Interventions

All participants firstly received single‐blind thiothixene 60 mg/day for 2 weeks to confirm treatment resistance e.g. not showing moderate improvement during the first 3 weeks of treatment. Participants were then randomised to either:

1. dose increase: thiothixene up to 400 mg/day. N = 25; or

2. dose maintenance: thiothixene 60 mg/day. N = 25.

Rescue medication: benztropine; no further details.

Outcomes

Global state: clinically relevant response (defined as moderate improvement in Roerig Global Scale (RGS) (Guy 1976))

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

Mental state (BPRS total score, BPRS factors such as anxiety‐depression, anergia, thought disturbance, activity, hostility‐suspicion), Nurses Observation Scale for Inpatient Evaluation (NOSIE total score, NOSIE factors such as social competence, social interest, personal neatness, irritability, manifest psychosis, retardation, depression).

Adverse effects (side effect check list, dystonia, dry mouth, blurred vision, drowsiness, orthostatic hypotension, tremor, dizziness, drooling, constipation, ataxia, akathisia, palpitations, headache, premature ventricular contractions).

Unable to use:

Vital signs (CBC, urinalysis, SMA‐12, ECG, blood pressure, pulse rate); no data were presented and not protocol outcomes.

Notes

50 participants were randomised. Eight of the 50 participants (4 in each group) showing moderate improvement on the RGS in the first 21 days were eliminated from the study as they were not considered to be treatment‐resistant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...randomly assigned..." (pg. 70); no further details

Allocation concealment (selection bias)

Unclear risk

No details were presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...under double‐blind control to..." (pg. 70); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...under double‐blind control to..." (pg. 70); no further details

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Unclear whether all those who left early were reported. It may be that 26% (13/50) left the study early.

Selective reporting (reporting bias)

High risk

No usable data on vital signs were reported in the results

Other bias

Low risk

No obvious risk for other bias

Kinon 1993

Methods

Randomisation: randomised, stratified based on week 3 serum fluphenazine levels
Blinding: double, no further details
Duration: 8 weeks; 4 weeks non‐randomised open‐label run in phase and 4 weeks randomised double‐blind phase.
Design: parallel
Location: single‐centre (USA)
Setting: inpatients

Participants

Diagnosis: schizophrenia, schizoaffective disorder or schizophreniform disorder (DSM III‐R)
N = 58
Gender: 64.1% males (of all participants entering the open label run in phase) (N = 156). Not indicated for randomised participants.
Age: mean 29.4 years (SD = 7.0), range 18 to 50 years for all participants entering the open label run in phase (N = 156), not indicated for randomised participants.
History: age at first hospitalisation ‒ mean 23.0 years (SD = 6.5); number of previous hospitalisations ‒ mean 2.6 (SD = 2.2); data for all participants entering the open label run in phase (N = 156), not indicated for randomised participants alone.

Interventions

All participants firstly received open label fluphenazine 20 mg/day for 4 weeks (N = 156). Participants who had a rating of worse than mild on each of the four BPRS psychotic items and a rating of less than much improved on the CGI‐I were considered non‐responders (N = 58) and were then randomised to:

1. dose increase: Fluphenazine 80 mg/day. N = 16; or

2. dose maintenance: Fluphenazine 20 mg/day. N = 18; or

3. additional intervention: haloperidol 20 mg/day. N = 13.

Rescue medication: benztropine, no further details.

Outcomes

Global state: clinically relevant response (defined as CGI‐I ≤ 2, at least much improved)

Mental state: general (BRPS total score), negative symptoms (modified SANS)

Unable to use:

Extrapyramidal symptoms (modified SAS, no mean)

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Non responders were then randomly assigned to double‐blind treatment for...", "Subjects were stratified
based on Week 3 serum fluphenazine levels..." (pg. 310); no further details.

Allocation concealment (selection bias)

Unclear risk

No details were presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...double‐blind treatment..." (pg.310); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...double‐blind treatment..." (pg.310); no further details

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Outcome not addressed. Data were presented for 81% (47/58) of all randomised participants

Selective reporting (reporting bias)

High risk

SAS was used but scores were available only for two items, not total

Other bias

Low risk

No obvious risk for other bias

Lindenmayer 2011

Methods

Randomisation: randomised, no further details
Blinding: double, no further details
Duration: 12 weeks; 4 weeks non‐randomised open‐label run in phase and 8 weeks randomised double‐blind phase.
Design: parallel
Location: multi‐centre (USA)
Setting: inpatients

Participants

Diagnosis: schizophrenia or schizoaffective disorder (DSM‐IV‐R); suboptimal past treatment response
N = 60
Gender: 55 men, 5 women
Age: mean 40.15 years (SD = 10.2)
History: age at onset ‒ mean 20.27 years (SD = 5.88); number of previous hospitalisations ‒ mean = 6.7 (SD = 5.88); suboptimal past treatment response, defined by (i) presence of persistent positive symptoms after at least 6 contiguous weeks of treatment with one or more typical or atypical antipsychotics at dosages ≥ 600 mg/d of chlorpromazine equivalents and (ii) poor level of functioning for the past 2 years defined as lack of competitive employment or enrolment in an academic or vocational program, and not having age‐expected interpersonal relations with someone outside the biological family of origin.

Interventions

All participants firstly received open label quetiapine 600 mg/day for 4 weeks. Participants who did not demonstrate an initial response to quetiapine treatment defined as ≤ 15% PANSS total score reduction were then randomised to either:

1. dose increase: quetiapine 1200 mg/day. N = 29; or

2. dose maintenance: quetiapine 600 mg/day. N = 31.

Concomitant mood stabilisers on a stable dose for the past 2 months before trial initiation were allowed to continue.

Outcomes

Global state: clinically relevant response (defined as ≥ 20% PANSS total score reduction)

Leaving the study early (due to side effects, any reason, inefficacy)

Mental state: general (PANSS total score), positive symptoms (PANSS positive subscore), negative symptoms (PANSS negative subscore)

Adverse effects: (SAS, AIMS, BAS, BMI/weight change, orthostatic hypotension, somnolence, agitation, constipation, weight gain)

Functional changes (Social‐Adaptive Functioning Evaluation ‐ SAFE)

Aggressive behaviour (BARS total score at endpoint)

Unable to use:

Global state (CGI no numbers presented)

Vital signs and ECG; no data presented and not protocol outcomes

Laboratory values (ALT, AST, GGT, glukose, lactate dehydrogenase, cholesterol, triglycerides), not protocol outcomes

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...patients were randomly assigned to...", "Stratification was done at the time of randomization to avoid imbalances secondary to the presence of concomitant mood stabilizer medications or the use of benztropine (or other antiparkinsonian agents) at baseline." (pg. 161); no further details.

Allocation concealment (selection bias)

Unclear risk

No details were presented

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "This was a prospective, randomised, double‐blind, parallel group, 8‐week trial of...", "All clinical and research stuff (subject, caregiver, investigator, outcomes assessor), except for the research pharmacists, were blinded to the randomization status." (pg. 161).

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "This was a prospective, randomised, double‐blind, parallel group, 8‐week trial of...", "All clinical and research stuff (subject, caregiver, investigator, outcomes assessor), except for the research pharmacists, were blinded to the randomization status." (pg. 161).

Incomplete outcome data (attrition bias)
All outcomes

High risk

53.3% of the participants left the study early. Numbers per group as well as reasons for leaving the study early were specified. Last observation carried forward (LOCF) methods were used.

Selective reporting (reporting bias)

High risk

Data on CGI, vital signs and ECG were not reported in the results.

Other bias

Low risk

No obvious risk for other bias

Loebel 2014

Methods

Randomisation: randomised, no further details
Blinding: double, no further details
Duration: 6 weeks; 2 weeks double‐blind run in phase and 4 weeks randomised double‐blind phase
Design: parallel
Location: multi‐centre
Setting: inpatients

Participants

Diagnosis: acute schizophrenia, no further details

N = 95
Gender: 60.1% males (of initially randomised participants to lurasidone 80 mg/day) (N = 198)
Age: mean 40.5 years; range 18 to 75 years for all initially randomised participants to lurasidone 80 mg/day

History: no details

Interventions

Participants were firstly randomised to double‐blind treatment with lurasidone 20 mg/day, lurasidone 80 mg/day, or placebo. After 2 weeks, only participants who were randomised to lurasidone 80 mg/day (N = 198) and showed < 20% PANSS total score reduction, were re‐randomised to either:

1. dose increase: lurasidone 160 mg/day. N = 43; or

2. dose maintenance: lurasidone 80 mg/day. N = 52.

Rescue medication: no details

Outcomes

Leaving the study early: due to side effects

Mental state: general (PANSS total score)

Global state (CGI‐Severity change)

Unable to use:

Weight gain (not separately presented for the two groups)

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "Eligible patients were randomised to..." (pg. 476); no further details

Allocation concealment (selection bias)

Unclear risk

No details are presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...to double‐blind treatment with..." (pg. 476); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...to double‐blind treatment with..." (pg. 476); no further details

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Outcome not addressed

Selective reporting (reporting bias)

Unclear risk

Insufficient information to permit judgement

Other bias

Low risk

No obvious risk for other bias

McEvoy 1991

Methods

Randomisation: randomised, no further details
Blinding: double, no further details
Duration: 6 weeks; 4 weeks non‐randomised open‐label run‐in phase and 2 weeks randomised double‐blind phase.
Design: parallel
Location: single‐centre (USA)
Setting: inpatients

Participants

Diagnosis: schizophrenia or schizoaffective disorder (RDC).

N = 48
Gender: 53.8% males (of all participants entering the open‐label run in phase) (N = 95). Not indicated for randomised participants.
Age: mean 31.5 years (SD = 9.5) for all participants entering the open‐label run in phase, not indicated for randomised participants.

History: no details

Interventions

All participants firstly received open label haloperidol at antipsychotic threshold (NT)* for at least 2 weeks. Non‐responding participants (not ready for discharge) were then randomised to either:

1. dose increase: haloperidol at a dosage two to ten times higher than NT dosage; mean 11.6 mg/day (SD = 4.7). N = 25; or

2. dose maintenance: haloperidol at NT dosage; mean 3.4 mg/day (SD = 2.3). N = 23.

Rescue medication: biperiden, lorazepam, diphenhydramine, no further details

Outcomes

Global state: clinically relevant response (defined as BPRS total score ≤ 32 (16 items), with all psychosis items (conceptual disorganisation, hallucinatory behavior, hostility, suspiciousness. and unusual thought content) rated "mild" or less; their CGl global severity item was rated "mild" or less; and their CGI global change item was rated at least "moderately improved").

Leaving the study early: due to adverse events

Mental sate: general (BRPS total score)

Global state (CGI‐S)

Unable to use:

Negative symptoms: Wing negative symptoms scale (no mean, no SD)

Mental state; self‐report of perceived medication effects: Medication Response Questionnaire (no mean, no SD)

Adverse events: Extrapyramidal Side Effects Scale (no mean, no SD) and anticholonergic side effects (no numbers presented)

Notes

* Antipsychotic threshold (NT): the lowest antipsychotic dosage at which individual patients develop slight increase in rigidity is hypothesised to correspond to the lowest dosages at which these patients attain maximum antipsychotic benefit (Haase 1961).

** Response criterion: BPRS total score ≤ 32 (16 items), with all psychosis items (conceptual disorganisation, hallucinatory behaviour, hostility, suspiciousness. and unusual thought content) rated "mild" or less; their CGl global severity item was rated "mild" or less; and their CGI global change item was rated at least "moderately improved".

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...patients were randomly assigned..." (pg. 740); no further details

Allocation concealment (selection bias)

Unclear risk

No details are presented

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Quote: "...on a double‐blind basis..." (pg. 740); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...on a double‐blind basis..." (pg. 740); no further details

Incomplete outcome data (attrition bias)
All outcomes

Low risk

An ITT approach was used. 8.3% (4/48) of participants left the study early due to severe extrapyramidal side effects. This is probably the overall attrition rate.

Selective reporting (reporting bias)

High risk

Wing negative symptom scale, Medication Response Questionnaire, Extrapyramidal Side Effects Scale and anticholinergic side effects were not presented separately for the group of non‐responders

Other bias

Low risk

No obvious risk for other bias

McGorry 2011

Methods

Randomisation: randomised, no further details
Blinding: single
Duration: 8 weeks; 4 weeks non‐randomised open‐label run in phase and 4 weeks randomised single‐blind phase
Design: parallel
Location: single‐centre (Australia)
Setting: inpatients

Participants

Diagnosis: schizophrenia, schizophreniform, schizoaffective, delusional disorder, psychotic disorder not otherwise specified, or brief psychosis (DSM‐IV); first psychotic episode
N = 26
Gender: 18 men, 8 women
Age: mean 21.62 years (SD = 3.98)
History: age at onset ‒ mean 21 years (SD = 3.8); duration of untreated psychosis ‒ mean 267.1 days (SD = 416.5); a first episode of psychosis

Interventions

All participants firstly received open label risperidone 2 mg/day for 4 weeks. Participants who were considered 'slow responders' defined as a score of > 3 on each of the BPRS psychosis subscale items (i.e. mild), a CGI‐S > 3, and a CGI‐I > 3 were then randomised to:

1. dose increase: risperidone 3 or 4 mg/day (if required). N = 9; or

2. dose maintenance: risperidone 2 mg/day. N = 8; or

3. additional intervention: risperidone 2 mg/day with addition of lithium, titrated up to therapeutic levels (0.6 to 1.2 mmol). N = 9.

Outcomes

Global state: clinically relevant response (defined as ≥ 20% BPRS total score reduction)
Leaving the study early (due to adverse events and any reason)
Mental state: general (BPRS total score), positive symptoms (BPRS psychosis subscale), negative symptoms (BPRS negative subscale)
Global state (CGI‐S)
Quality of life: number of participants with at least 50% improvement inHQLS and mean endpoint score in HQLS
Unable to use:

Global state (CGI‐I, no numbers)

Extrapyramidal adverse events (not separately presented for the group of 'slow responders') and weight gain (no SD).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote: "...slow responders were randomised..." (pg. 3); no further details

Allocation concealment (selection bias)

Unclear risk

No details are presented

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "...randomised single blind to one of three open treatment groups" (pg. 3); no further details

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Quote: "...randomised single blind to one of three open treatment groups" (pg. 3); no further details

Incomplete outcome data (attrition bias)
All outcomes

Low risk

19.2% (5/26) participants left the study early. A LOCF approach was adopted for analysis of Phase II data

Selective reporting (reporting bias)

High risk

Original study author sent us data on most outcomes but data on CGI‐I, extrapyramidal side effects and weight gain were not reported.

Other bias

Low risk

No obvious risk for other bias

Sakurai 2016

Methods

Randomisation: randomised to 1 of the 2 treatment groups in a 1:1 ratio by simple randomisation stratified by their antipsychotic type and treatment setting; computer‐generated randomisation list.
Blinding: double; identical powder form in amount and color; participants blinded to their allocated intervention; assessors blinded to the allocation.
Duration: 4 weeks
Design: parallel
Location: single‐centre (Japan)
Setting: inpatients and outpatients

Participants

Diagnosis: schizophrenia, schizoaffective or persistent delusional disorder (ICD‐10)

N = 103
Gender: 38 men, 65 women
Age: mean 50.7 years (SD = 15.81)

History: duration of illness ‒ mean 16.05 years (SD = 14.4); total duration of antipsychotic treatment‐ mean 10.9 years (SD = 14).

Interventions

All participants had been receiving olanzapine 10 mg/day or risperidone 3 mg/day for at least 4 weeks. Participants who had a total score ≥ 60 on the PANSS, ≥ 3 on the CGI‐S, and ≤ 70 on the GAF were considered non‐responders and were randomised to either:

1. dose increase: olanzapine 20 mg/day or risperidone 6 mg/day (double antipsychotic dose). N = 52; or

2. dose maintenance: olanzapine 10 mg/day or risperidone 3 mg/day. N = 51.

Outcomes

Global state: clinically relevant response (defined as ≥ 25% PANSS total score reduction)
Leaving the study early (due to adverse events, any reason and inefficacy)

Mental state: general (PANSS total score), positive symptoms (PANSS positive subscore), negative symptoms (PANSS negative subscore)

Global state (CGI‐I)

Functioning: overall (GAF)

Adverse effects (EPS ‐ SAS, Akathisia ‐ BAS, tardive dyskinesia ‐ AIMS)

Unable to use:

Global state: CGI‐S (no mean, no SD)

Plasma concentrations, not a protocol outcome

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "...randomly allocated to 1 of the 2 treatment groups in a 1:1 ratio by simple randomization stratified by their antipsychotic type (ie, olanzapine or risperidone) and treatment setting (ie, inpatient or outpatient)....according to a computer‐generated randomization list..." (pg. 1382).

Allocation concealment (selection bias)

Low risk

Quote: "The person who was independent of this study in the central office prepared a piece of paper on which 1 of the assigned groups was designated according to a computer‐generated randomization list, inserted it into an envelope on which a participant ID number was written, and sealed it. Upon registration of each participant, 1 of the investigators opened the envelope that corresponded to the participant's ID, and the person who prepared the envelopes confirmed that the envelopes were appropriately opened." (pg. 1382).

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "During the 4‐week observation, all antipsychotic drugs were provided in identical powder form in amount and color with lactose added... Thus, the participants were blinded to their allocated intervention." (pg. 1382).

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "The following assessments were performed by assessors who were blinded to the allocation..." (pg. 1383).

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Significantly more participants in the dose increase group (30.8%) than in the dose continuation group (13.7%) left the study early due to side effects. Reasons for leaving the study early were described. An ITT approach was used. Results between ITT analysis and only competers were similar.

Selective reporting (reporting bias)

High risk

CGI‐S was used but scores at endpoint were not available

Other bias

Low risk

No obvious risk for other bias

Scales

AIMS: Abnormal Involuntary Movement Score BAS: Barnes Akathisia Scale

BARS: Behavioral Activity Rating Scale

BAS: Barnes Akathisia Rating Scale

BPRS: Brief Psychiatric Rating Scale

CDRS: Calgary Depression Rating Scale

CGI‐I: Clinical Global impression‐Improvement

CGI‐S: Clinical Global impression‐Severity

CPRS: Comprehensive Psychopathological Rating Scale

GAF: Global Assessment of Functioning

HQLS: Heinrichs‐Carpenter‐Hanlon Quality of Life Scale

NOSIE: Nurse's Observation Scale for Inpatient Evaluation

PANSS: Positive and Negative Syndrome Scale for Schizophrenia

RGS: Roering Global Scale

SAFE: Social‐Adaptive Functioning Evaluation

SANS: Scale for the Assessment of Negative Symptoms

SAS: Simpson Angus Scale

SCoRS: Schizophrenia Cognition Rating Scale

SOFAS: Social and Occupational Functioning Scale

Diagnostic Tools

DSM: Diagnostic and Statistical Manual of Mental Disorders

ICD: International Classification of Diseases

RDC: Research Diagnostic Criteria for schizophrenia or schizoaffective disorders

Others

BMI: Body‐mass‐index

ECG: Electrocardiogram

EPS: Extrapyramidal Symptoms

ITT: Intention‐to‐treat

LOCF: Last observation carried forward

mg: Milligram

msec: Millisecond

N: Number

n.i.: Not indicated

SD: Standard deviation

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Agid 2013

Allocation: not randomised

Badgett 1996

Allocation: randomised
Participants: schizophrenia (DSM‐III‐R) based on the CASH‐interview; at baseline, participants were stabilised on 20 mg/day of haloperidol, not non‐responders

Bai 2002

Allocation: not randomised

Baker 2003

Allocation: randomised
Participants: schizophrenia, schizoaffective disorder, schizophreniform disorder, or bipolar I disorder, manic or mixed episode (DSM‐IV); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: rapid dose escalation or usual clinical practice

Bastecky 1982

Allocation: not indicated
Participants: schizophrenia (chronic schizophrenics); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment

Bitter 1989

Allocation: randomised
Participants: schizophrenia or schizoaffective disorder (acutely exacerbating)
Interventions: initially 3 plasma levels of haloperidol; if no improvement after 6 weeks, randomly re‐assignment to one of the 3 plasma levels of haloperidol for another 6 weeks.
Outcomes: no usable data.

Bondolfi 1995

Allocation: randomised
Participants: schizophrenia (chronic according to DSM‐III), treatment‐resistant (defined by unresponsiveness or intolerance to appropriate doses of two different classes of conventional antipsychotics for at least 4 weeks each); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: no antipsychotic dose increase versus maintenance Comparison; either risperidone or clozapine; drug dosages could be changed after day 14 depending on each participant's response.

Branchey 1981

Allocation: randomised
Participants: schizophrenia, chronic type (according to Research Diagnostic Criteria); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: loxapine continuation versus loxapine decrease; no dose increase group

Canuso 2010

Allocation: randomised
Participants: acute exacerbation of schizoaffective disorder (DSM‐IV); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.

Chen 1998

Allocation: not indicated
Participants: no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment

Clerc 1989

Allocation: distributed by drawing of lots
Participants: hospitalised participants all with negative symptoms (ICD‐9 and DSM‐III); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: 2 ranges of amisulpride doses (20 mg to 120 mg and 100 mg to 600 mg); no dose continuation group

CN138032

Allocation: randomised
Participants: treatment‐resistant schizophrenia
Interventions: 1. open‐label phase olanzapine or risperidone; 2. single‐blind placebo washout period; 3. double‐blind phase perphenazine vs. aripiprazole; 4. open‐label aripiprazole extended phase.

Cookson 1987

Allocation: randomised
Participants: schizophrenia (hebephrenic or paranoid, ICD‐9); treatment‐resistant to low dosages of antipsychotics; improved with higher dosages.
Interventions: 50% reduction of dosage or dosage continuation; no dose increase group

Coryell 1998

Allocation: randomised
Participants: schizophrenia, acute exacerbation (DSM‐III)
Interventions: 1st phase: low‐dose (< 18ng/ml), intermediate or high‐dose (> 25 ng/ml) range of haloperidol plasma levels for 3 weeks; 2nd phase: if no improvement, antipsychotic dose could randomly remain the same, be reduced or increased.

Daniel 1997

Allocation: not indicated
Participants: schizophrenia or schizoaffective disorder; no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: ziprasidone 80 mg vs. ziprasidone 160 mg; no dose maintenance group

de Leon 2007

Allocation: randomised
Participants: schizophrenia or schizoaffective disorder (DSM‐III); Kane et al criteria (Kane 1988) for defining treatment resistance.
Interventions: 1st phase: 16 weeks in one of the following dose‐groups: 100, 300, 600 mg/day clozapine; if no improvement, randomization to one of the two other dosage groups; no dose maintenance group.

DeBuck 1972

Allocation: not randomised for non‐responders
Participants: any psychoses with the exception of chronic organic psychosis with superimposed non‐psychotic symptomatology, and endogenous depression with retardation and apathy, and "acute organic psychosis".

Dencker 1978

Allocation: randomised
Participants: schizophrenia; all participants firstly received open‐label 12.5 mg fluphenazine enanthate per week for 2 weeks and those who were proven "refractory to ordinary doses of neuroleptics" were then randomised.

Interventions: 250 mg fluphenazine enanthate per week versus 12.5 mg fluphenazine enanthate per week.
Outcomes: no usable data due to the cross‐over design of the trial.

Ericksen 1978

Allocation: randomised
Participants: schizophrenia (acutely decompensated); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: haloperidol standard dose versus haloperidol high loading dose; no dose maintenance group.

Gardos 1971

Allocation: not indicated
Participants: schizophrenia (treatment resistant, decompensated); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: thiothixene high dose versus thiothixene low dose
Outcomes: no usable data

Gulliver 2010

Allocation: randomised
Participants: schizophrenia, participants who maintained stability on open‐label oral olanzapine
Interventions: randomised to low, medium or high doses of olanzapine long‐acting injection for 24 weeks

Harris 1997

Allocation: not indicated
Participants: people with schizophrenia and related psychotic disorders over the age of 45 who met DSM‐IV criteria for "in remission''; not non‐responders.

Hirschowitz 1995

Allocation: not indicated
Participants: participants with schizophrenia, stabilised on 20 mg/day; not non‐responders.

Hirschowitz 1997

Allocation: randomised
Participants: schizophrenia (DSM‐III‐R) based on the CASH‐interview; participants free of medication at baseline or stabilised on 20 mg/day.
Interventions: no dose continuation or increase group

Itil 1970

Allocation: not indicated.
Participants: schizophrenia (chronic); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: fluphenazine 30 mg versus fluphenazine 800 mg; no dose maintenance or increase group.

Janicak 1997

Allocation: randomised.
Participants: schizophrenia or schizoaffective disorder; acutely psychotic.
Interventions: low, middle or high plasma level range of haloperidol; after 2 weeks, 50% of the non‐responders were randomly reassigned to the middle plasma level group.

Kane 1985

Allocation: not indicated.
Participants: schizophrenia (remitted or stable); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: low‐dose fluphenazine decanoate versus standard‐dose fluphenazine decanoate; no dose increase group.

Lehmann 1980

Allocation: randomised.

Participants: schizophrenia; no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: haloperidol 10 mg/day, 20 mg/day or individual dose of haloperidol; no dose increase or maintenance group.

McCreadie 1979

Allocation: randomised
Participants: schizophrenia (female, drug‐resistant, chronic); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: flupentixol decanoate high‐dose (200 mg/2 weeks) versus standard dose (40 mg/2 weeks); no dose increase or maintenance group.

Mitchell 2004

Allocation: randomised
Participants: schizophrenia, schizoaffective disorder, bipolar I disorder (DSM‐IV‐R); stable inpatients; the run‐in phase was not used to identify participants that have not responded to their current antipsychotic treatment.
Interventions: run‐in phase of olanzapine 20 mg/day for 10 days; then, randomisation to olanzapine 20 mg/day, 30 mg/day or 40 mg/day for 10 days; finally, for an additional 10 days, 30 mg/day participants received olanzapine 40 mg/day; all other participants remained on the same dose.
Outcomes: no usable data

NCT00539071

Allocation: randomised
Participants: schizophrenia or schizoaffective disorder; eligible participants would be receiving or had received treatment with risperidone oral or long‐acting, or a combination that did not exceed 50 mg/2 weeks of long‐acting or oral risperidone 8 mg/day for at least 6 weeks within seven years of study entry without satisfactory response.
Interventions: long‐acting risperidone 50 mg/week vs 75 mg to 100 mg/2 weeks.
Outcomes: no usable data

NCT00862992

Allocation: randomised
Participants: schizophrenia (DSM‐IV‐TR); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: low‐dose versus medium‐dose versus high‐dose of cariprazine; no dose increase or maintenance group.

NCT01457339

Allocation: randomised
Participants: clinically stable participants with schizophrenia (on a stable dose of an antipsychotic drug); no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: SPD489 multiple doses versus placebo; no dose increase or maintenance group.

NCT01569659

Allocation: randomised
Participants: schizophrenia or schizoaffective disorder (DSM‐IV); Kane et al criteria (Kane 1988) for treatment‐resistance; no run‐in phase to confirm that participants have not responded to their current antipsychotic treatment.
Interventions: low dosage of lurasidone (80 mg/day) versus high dose of lurasidone (up to 240 mg/day); no dose increase or maintenance group.

Simpson 1999

Allocation: randomised
Participants: any psychoses with the exception of chronic organic psychosis with superimposed non‐psychotic symptomatology, endogenous depression and acute organic psychosis.
Interventions: clozapine 100, 300 or 600 mg/d; if no improvement was observed after 16 weeks, participants were randomised to one of the other two dosages, no maintenance group.

Suzuki 1992

Allocation: randomised
Participants: schizophrenia (DSM‐III); not non‐responders

Volavka 1996

Allocation: not randomised for the group of non‐responders

Data and analyses

Open in table viewer
Comparison 1. Antipsychotic dose increase versus antipsychotic dose maintenance

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Global state: 1a. Clinically relevant response as defined by trials Show forest plot

9

533

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.86, 1.40]

Analysis 1.1

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 1 Global state: 1a. Clinically relevant response as defined by trials.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 1 Global state: 1a. Clinically relevant response as defined by trials.

2 Global state: 1b. Any change (improvement in Global Assesment scale) Show forest plot

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.76 [0.32, 1.80]

Analysis 1.2

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 2 Global state: 1b. Any change (improvement in Global Assesment scale).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 2 Global state: 1b. Any change (improvement in Global Assesment scale).

3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor) Show forest plot

3

196

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.40, 0.19]

Analysis 1.3

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor).

4 Global state: 2b. Average change score ( CGI‐Severity, high = poor) Show forest plot

1

95

Mean Difference (IV, Random, 95% CI)

‐0.4 [‐0.80, ‐0.00]

Analysis 1.4

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 4 Global state: 2b. Average change score ( CGI‐Severity, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 4 Global state: 2b. Average change score ( CGI‐Severity, high = poor).

5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor) Show forest plot

1

103

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.35, 0.35]

Analysis 1.5

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor).

6 Leaving the study early: 1. Tolerability ‐ due to adverse events Show forest plot

7

496

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.52, 5.07]

Analysis 1.6

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 6 Leaving the study early: 1. Tolerability ‐ due to adverse events.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 6 Leaving the study early: 1. Tolerability ‐ due to adverse events.

7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason Show forest plot

5

353

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.89, 1.90]

Analysis 1.7

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason.

8 Leaving the study early: 3. Efficacy ‐ due to inefficacy Show forest plot

4

336

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.30, 2.28]

Analysis 1.8

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 8 Leaving the study early: 3. Efficacy ‐ due to inefficacy.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 8 Leaving the study early: 3. Efficacy ‐ due to inefficacy.

9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

‐1.81 [‐7.31, 3.69]

Analysis 1.9

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor).

10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor) Show forest plot

3

258

Mean Difference (IV, Random, 95% CI)

‐1.44 [‐6.85, 3.97]

Analysis 1.10

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor).

11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor) Show forest plot

4

389

Mean Difference (IV, Random, 95% CI)

‐2.13 [‐6.16, 1.90]

Analysis 1.11

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor).

12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor) Show forest plot

3

99

Mean Difference (IV, Random, 95% CI)

‐1.25 [‐4.60, 2.11]

Analysis 1.12

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor).

13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor) Show forest plot

1

42

Mean Difference (IV, Random, 95% CI)

‐2.38 [‐6.15, 1.39]

Analysis 1.13

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor).

14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor) Show forest plot

4

141

Mean Difference (IV, Random, 95% CI)

‐1.75 [‐4.25, 0.76]

Analysis 1.14

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor).

15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor) Show forest plot

1

42

Mean Difference (IV, Random, 95% CI)

3.70 [‐5.38, 12.78]

Analysis 1.15

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor).

16 Mental state: 2a. Positive symptoms ‐ Clinically important change Show forest plot

1

17

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.58, 3.07]

Analysis 1.16

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 16 Mental state: 2a. Positive symptoms ‐ Clinically important change.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 16 Mental state: 2a. Positive symptoms ‐ Clinically important change.

17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

‐0.94 [‐2.79, 0.90]

Analysis 1.17

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor).

18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor) Show forest plot

3

238

Mean Difference (IV, Random, 95% CI)

0.04 [‐1.31, 1.40]

Analysis 1.18

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor).

19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

0.40 [‐2.94, 3.74]

Analysis 1.19

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor).

20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

0.32 [‐1.48, 2.11]

Analysis 1.20

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor).

21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

‐0.15 [‐0.96, 0.67]

Analysis 1.21

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor).

22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐1.97, 1.17]

Analysis 1.22

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor).

23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor) Show forest plot

1

34

Mean Difference (IV, Random, 95% CI)

1.5 [‐14.33, 17.33]

Analysis 1.23

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor).

24 Adverse effects ‐ At least one adverse effect Show forest plot

2

191

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.55, 1.50]

Analysis 1.24

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 24 Adverse effects ‐ At least one adverse effect.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 24 Adverse effects ‐ At least one adverse effect.

25 Adverse effects ‐ Cardiac: QTc prolongation Show forest plot

2

206

Risk Ratio (M‐H, Random, 95% CI)

2.47 [0.12, 50.39]

Analysis 1.25

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 25 Adverse effects ‐ Cardiac: QTc prolongation.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 25 Adverse effects ‐ Cardiac: QTc prolongation.

26 Adverse effects ‐ Cardiac: Orthostatic hypotension Show forest plot

2

102

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.25, 4.82]

Analysis 1.26

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 26 Adverse effects ‐ Cardiac: Orthostatic hypotension.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 26 Adverse effects ‐ Cardiac: Orthostatic hypotension.

27 Adverse effects ‐ Cardiac: Palpitations Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.70]

Analysis 1.27

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 27 Adverse effects ‐ Cardiac: Palpitations.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 27 Adverse effects ‐ Cardiac: Palpitations.

28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.70]

Analysis 1.28

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions.

29 Adverse effects ‐ Constipation Show forest plot

2

102

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.44, 5.38]

Analysis 1.29

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 29 Adverse effects ‐ Constipation.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 29 Adverse effects ‐ Constipation.

30 Adverse effects ‐ Dizziness Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.41, 1.44]

Analysis 1.30

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 30 Adverse effects ‐ Dizziness.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 30 Adverse effects ‐ Dizziness.

31 Adverse effects ‐ Drooling Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.20, 20.41]

Analysis 1.31

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 31 Adverse effects ‐ Drooling.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 31 Adverse effects ‐ Drooling.

32 Adverse effects ‐ Death (suicide or naturalistic cause) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.32

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 32 Adverse effects ‐ Death (suicide or naturalistic cause).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 32 Adverse effects ‐ Death (suicide or naturalistic cause).

33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.38 [0.59, 3.26]

Analysis 1.33

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor).

34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.7 [‐0.87, 2.27]

Analysis 1.34

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor).

35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

0.41 [‐1.15, 1.96]

Analysis 1.35

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor).

36 Adverse effects ‐ Extrapyramidal: akathisia Show forest plot

2

65

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.04, 14.02]

Analysis 1.36

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 36 Adverse effects ‐ Extrapyramidal: akathisia.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 36 Adverse effects ‐ Extrapyramidal: akathisia.

37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.47 [0.42, 5.14]

Analysis 1.37

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor).

38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐0.2 [‐0.74, 0.34]

Analysis 1.38

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor).

39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.59, 0.37]

Analysis 1.39

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor).

40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia Show forest plot

3

196

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.09, 2.73]

Analysis 1.40

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia.

41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.51, 2.93]

Analysis 1.41

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS.

42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

0.93 [‐1.04, 2.91]

Analysis 1.42

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor).

43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

0.21 [‐0.83, 1.26]

Analysis 1.43

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor).

44 Adverse effects ‐ Extrapyramidal: tremor Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

1.59 [0.59, 4.26]

Analysis 1.44

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 44 Adverse effects ‐ Extrapyramidal: tremor.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 44 Adverse effects ‐ Extrapyramidal: tremor.

45 Adverse effects ‐ Headache Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

1.46 [0.52, 4.08]

Analysis 1.45

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 45 Adverse effects ‐ Headache.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 45 Adverse effects ‐ Headache.

46 Adverse effects ‐ Somnolence and/or drowsiness Show forest plot

4

256

Risk Ratio (M‐H, Random, 95% CI)

1.76 [0.81, 3.81]

Analysis 1.46

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 46 Adverse effects ‐ Somnolence and/or drowsiness.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 46 Adverse effects ‐ Somnolence and/or drowsiness.

47 Adverse effects ‐ Weight at endpoint (high = poor) Show forest plot

2

165

Mean Difference (IV, Random, 95% CI)

‐1.85 [‐7.09, 3.39]

Analysis 1.47

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 47 Adverse effects ‐ Weight at endpoint (high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 47 Adverse effects ‐ Weight at endpoint (high = poor).

48 Behaviour: average endpoint score (BARS, high = good) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.50, 0.30]

Analysis 1.48

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 48 Behaviour: average endpoint score (BARS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 48 Behaviour: average endpoint score (BARS, high = good).

49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good) Show forest plot

1

103

Mean Difference (IV, Random, 95% CI)

‐0.60 [‐1.00, 1.80]

Analysis 1.49

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good).

50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.16 [‐0.47, 0.79]

Analysis 1.50

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor).

51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good) Show forest plot

1

131

Mean Difference (IV, Random, 95% CI)

0.5 [‐3.80, 4.80]

Analysis 1.51

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good).

52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good) Show forest plot

1

17

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.52

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good).

53 Quality of life: Average endpoint score (HQLS, high = good) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

5.5 [‐13.66, 24.66]

Analysis 1.53

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 53 Quality of life: Average endpoint score (HQLS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 53 Quality of life: Average endpoint score (HQLS, high = good).

Open in table viewer
Comparison 2. Subgroup analysis and investigation of heterogeneity

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Single antipsychotic drugs Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 1 Single antipsychotic drugs.

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 1 Single antipsychotic drugs.

1.1 Fluphenazine

1

34

Risk Ratio (M‐H, Random, 95% CI)

2.25 [0.22, 22.53]

1.2 Haloperidol

2

71

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.67, 1.80]

1.3 Quetiapine

2

191

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.15, 2.86]

1.4 Risperidone

1

17

Risk Ratio (M‐H, Random, 95% CI)

9.9 [0.63, 155.08]

1.5 Thiothixene

1

42

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.26]

1.6 Ziprasidone

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.45, 1.41]

2 Clinical state, stage or problem Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 2 Clinical state, stage or problem.

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 2 Clinical state, stage or problem.

2.1 First episode patients

1

17

Risk Ratio (M‐H, Random, 95% CI)

9.9 [0.63, 155.08]

2.2 Not first episode patients

8

516

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.86, 1.34]

Open in table viewer
Comparison 3. Sensitivity analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Excluding studies with unclear randomisation methods Show forest plot

2

234

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.77, 1.44]

Analysis 3.1

Comparison 3 Sensitivity analysis, Outcome 1 Excluding studies with unclear randomisation methods.

Comparison 3 Sensitivity analysis, Outcome 1 Excluding studies with unclear randomisation methods.

2 Excluding studies with unclear allocation concealment methods Show forest plot

2

234

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.77, 1.44]

Analysis 3.2

Comparison 3 Sensitivity analysis, Outcome 2 Excluding studies with unclear allocation concealment methods.

Comparison 3 Sensitivity analysis, Outcome 2 Excluding studies with unclear allocation concealment methods.

3 Exclusion of studies with high risk of bias regarding blinding Show forest plot

8

516

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.86, 1.34]

Analysis 3.3

Comparison 3 Sensitivity analysis, Outcome 3 Exclusion of studies with high risk of bias regarding blinding.

Comparison 3 Sensitivity analysis, Outcome 3 Exclusion of studies with high risk of bias regarding blinding.

4 Exclusion of studies with high risk of bias regarding incomplete outcome data Show forest plot

6

375

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.92, 1.54]

Analysis 3.4

Comparison 3 Sensitivity analysis, Outcome 4 Exclusion of studies with high risk of bias regarding incomplete outcome data.

Comparison 3 Sensitivity analysis, Outcome 4 Exclusion of studies with high risk of bias regarding incomplete outcome data.

5 Exclusion of studies with imputed values Show forest plot

8

458

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.90, 1.50]

Analysis 3.5

Comparison 3 Sensitivity analysis, Outcome 5 Exclusion of studies with imputed values.

Comparison 3 Sensitivity analysis, Outcome 5 Exclusion of studies with imputed values.

6 Fixed effects Show forest plot

9

533

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.88, 1.38]

Analysis 3.6

Comparison 3 Sensitivity analysis, Outcome 6 Fixed effects.

Comparison 3 Sensitivity analysis, Outcome 6 Fixed effects.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

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

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 1 Global state: 1a. Clinically relevant response as defined by trials.
Figuras y tablas -
Analysis 1.1

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 1 Global state: 1a. Clinically relevant response as defined by trials.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 2 Global state: 1b. Any change (improvement in Global Assesment scale).
Figuras y tablas -
Analysis 1.2

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 2 Global state: 1b. Any change (improvement in Global Assesment scale).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor).
Figuras y tablas -
Analysis 1.3

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 4 Global state: 2b. Average change score ( CGI‐Severity, high = poor).
Figuras y tablas -
Analysis 1.4

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 4 Global state: 2b. Average change score ( CGI‐Severity, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor).
Figuras y tablas -
Analysis 1.5

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 6 Leaving the study early: 1. Tolerability ‐ due to adverse events.
Figuras y tablas -
Analysis 1.6

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 6 Leaving the study early: 1. Tolerability ‐ due to adverse events.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason.
Figuras y tablas -
Analysis 1.7

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 8 Leaving the study early: 3. Efficacy ‐ due to inefficacy.
Figuras y tablas -
Analysis 1.8

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 8 Leaving the study early: 3. Efficacy ‐ due to inefficacy.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor).
Figuras y tablas -
Analysis 1.9

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor).
Figuras y tablas -
Analysis 1.10

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor).
Figuras y tablas -
Analysis 1.11

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor).
Figuras y tablas -
Analysis 1.12

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor).
Figuras y tablas -
Analysis 1.13

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor).
Figuras y tablas -
Analysis 1.14

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor).
Figuras y tablas -
Analysis 1.15

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 16 Mental state: 2a. Positive symptoms ‐ Clinically important change.
Figuras y tablas -
Analysis 1.16

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 16 Mental state: 2a. Positive symptoms ‐ Clinically important change.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor).
Figuras y tablas -
Analysis 1.17

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor).
Figuras y tablas -
Analysis 1.18

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor).
Figuras y tablas -
Analysis 1.19

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor).
Figuras y tablas -
Analysis 1.20

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor).
Figuras y tablas -
Analysis 1.21

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor).
Figuras y tablas -
Analysis 1.22

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor).
Figuras y tablas -
Analysis 1.23

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 24 Adverse effects ‐ At least one adverse effect.
Figuras y tablas -
Analysis 1.24

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 24 Adverse effects ‐ At least one adverse effect.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 25 Adverse effects ‐ Cardiac: QTc prolongation.
Figuras y tablas -
Analysis 1.25

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 25 Adverse effects ‐ Cardiac: QTc prolongation.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 26 Adverse effects ‐ Cardiac: Orthostatic hypotension.
Figuras y tablas -
Analysis 1.26

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 26 Adverse effects ‐ Cardiac: Orthostatic hypotension.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 27 Adverse effects ‐ Cardiac: Palpitations.
Figuras y tablas -
Analysis 1.27

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 27 Adverse effects ‐ Cardiac: Palpitations.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions.
Figuras y tablas -
Analysis 1.28

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 29 Adverse effects ‐ Constipation.
Figuras y tablas -
Analysis 1.29

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 29 Adverse effects ‐ Constipation.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 30 Adverse effects ‐ Dizziness.
Figuras y tablas -
Analysis 1.30

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 30 Adverse effects ‐ Dizziness.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 31 Adverse effects ‐ Drooling.
Figuras y tablas -
Analysis 1.31

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 31 Adverse effects ‐ Drooling.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 32 Adverse effects ‐ Death (suicide or naturalistic cause).
Figuras y tablas -
Analysis 1.32

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 32 Adverse effects ‐ Death (suicide or naturalistic cause).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor).
Figuras y tablas -
Analysis 1.33

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor).
Figuras y tablas -
Analysis 1.34

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor).
Figuras y tablas -
Analysis 1.35

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 36 Adverse effects ‐ Extrapyramidal: akathisia.
Figuras y tablas -
Analysis 1.36

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 36 Adverse effects ‐ Extrapyramidal: akathisia.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor).
Figuras y tablas -
Analysis 1.37

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor).
Figuras y tablas -
Analysis 1.38

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor).
Figuras y tablas -
Analysis 1.39

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia.
Figuras y tablas -
Analysis 1.40

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS.
Figuras y tablas -
Analysis 1.41

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor).
Figuras y tablas -
Analysis 1.42

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor).
Figuras y tablas -
Analysis 1.43

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 44 Adverse effects ‐ Extrapyramidal: tremor.
Figuras y tablas -
Analysis 1.44

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 44 Adverse effects ‐ Extrapyramidal: tremor.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 45 Adverse effects ‐ Headache.
Figuras y tablas -
Analysis 1.45

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 45 Adverse effects ‐ Headache.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 46 Adverse effects ‐ Somnolence and/or drowsiness.
Figuras y tablas -
Analysis 1.46

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 46 Adverse effects ‐ Somnolence and/or drowsiness.

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 47 Adverse effects ‐ Weight at endpoint (high = poor).
Figuras y tablas -
Analysis 1.47

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 47 Adverse effects ‐ Weight at endpoint (high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 48 Behaviour: average endpoint score (BARS, high = good).
Figuras y tablas -
Analysis 1.48

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 48 Behaviour: average endpoint score (BARS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good).
Figuras y tablas -
Analysis 1.49

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor).
Figuras y tablas -
Analysis 1.50

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good).
Figuras y tablas -
Analysis 1.51

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good).
Figuras y tablas -
Analysis 1.52

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good).

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 53 Quality of life: Average endpoint score (HQLS, high = good).
Figuras y tablas -
Analysis 1.53

Comparison 1 Antipsychotic dose increase versus antipsychotic dose maintenance, Outcome 53 Quality of life: Average endpoint score (HQLS, high = good).

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 1 Single antipsychotic drugs.
Figuras y tablas -
Analysis 2.1

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 1 Single antipsychotic drugs.

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 2 Clinical state, stage or problem.
Figuras y tablas -
Analysis 2.2

Comparison 2 Subgroup analysis and investigation of heterogeneity, Outcome 2 Clinical state, stage or problem.

Comparison 3 Sensitivity analysis, Outcome 1 Excluding studies with unclear randomisation methods.
Figuras y tablas -
Analysis 3.1

Comparison 3 Sensitivity analysis, Outcome 1 Excluding studies with unclear randomisation methods.

Comparison 3 Sensitivity analysis, Outcome 2 Excluding studies with unclear allocation concealment methods.
Figuras y tablas -
Analysis 3.2

Comparison 3 Sensitivity analysis, Outcome 2 Excluding studies with unclear allocation concealment methods.

Comparison 3 Sensitivity analysis, Outcome 3 Exclusion of studies with high risk of bias regarding blinding.
Figuras y tablas -
Analysis 3.3

Comparison 3 Sensitivity analysis, Outcome 3 Exclusion of studies with high risk of bias regarding blinding.

Comparison 3 Sensitivity analysis, Outcome 4 Exclusion of studies with high risk of bias regarding incomplete outcome data.
Figuras y tablas -
Analysis 3.4

Comparison 3 Sensitivity analysis, Outcome 4 Exclusion of studies with high risk of bias regarding incomplete outcome data.

Comparison 3 Sensitivity analysis, Outcome 5 Exclusion of studies with imputed values.
Figuras y tablas -
Analysis 3.5

Comparison 3 Sensitivity analysis, Outcome 5 Exclusion of studies with imputed values.

Comparison 3 Sensitivity analysis, Outcome 6 Fixed effects.
Figuras y tablas -
Analysis 3.6

Comparison 3 Sensitivity analysis, Outcome 6 Fixed effects.

Table 1. Suggested design for future study

Methods

Randomisation: random.

Allocation: concealed.
Blinding: double blind.
Duration: at least 2 weeks run‐in phase to confirm non response to initial treatment and at least 4 weeks randomised double‐blind phase.

Setting: in‐ or out‐patients.

Participants

Diagnosis: patients with schizophrenia, schizoaffective disorder or schizophreniform disorder.

N > 300
Gender: male and female patients.
Age: mean 30 years (SD = 7.0), range 18 to 65 years.

Interventions

All patients firstly receive treatment with one antipsychotic drug for at least 2 weeks. Those patients who do not at least minimally improve after 2 weeks of treatment, are considered non‐responders and are randomised to:

1. Increasing the dose of the antipsychotic drug above the officially recommended dose range.

2. Continuing treatment with the antipsychotic drug at the same, initial dose (within the officially recommended dose range).

Outcomes

Response (e.g. defined as PANSS or BPRS decrease ≥ 50%).*

Relapse.

Leaving the study early due to any reason.

Leaving the study early due to side effects.

General mental state‐average change in general mental state scores.

Adverse effects‐ at least one adverse effect; clinically important general adverse effects; sudden and unexpected death.

Service use‐ time in hospital.

Quality of life.

All outcomes by time ‐ short term (up to 12 weeks), medium term (13 to 26 weeks) and long term (over 26 weeks).

Notes

*Primary outcome of interest.

Figuras y tablas -
Table 1. Suggested design for future study
Summary of findings for the main comparison. Antipsychotic dose increase compared to antipsychotic dose continuation for non response in schizophrenia

Antipsychotic dose increase compared to antipsychotic dose continuation for non response in schizophrenia

Patient or population: non response in schizophrenia
Setting: inpatients and outpatients
Intervention: antipsychotic dose increase
Comparison: antipsychotic dose continuation

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with antipsychotic dose continuation

Risk with antipsychotic dose increase

Global state: clinically relevant response: Assessed with response ratio
follow‐up: range 2 weeks to 12 weeks

Study population

RR 1.09
(0.86 to 1.40)

533
(9 RCTs)

⊕⊕⊝⊝
LOW 1

309 per 1000

336 per 1000
(265 to 432)

Leaving the study early: tolerability ‐ leaving early due to adverse effects.
Assessed with risk ratio follow‐up: range 2 weeks to 9 weeks

Study population

RR 1.63
(0.52 to 5.07)

496
(7 RCTs)

⊕⊝⊝⊝
VERY LOW 1 2

74 per 1000

121 per 1000
(39 to 376)

Leaving the study early: acceptability ‐ leaving early due to any reason.
Assessed with: Risk ratio
follow‐up: range 2 weeks to 9 weeks

Study population

RR 1.30
(0.89 to 1.90)

353
(5 RCTs)

⊕⊕⊝⊝
LOW 1

23 per 100

30 per 100
(20 to 43)

General mental state : PANSS total score change*
assessed with: Weighted mean difference
follow‐up: range 2 weeks to 9 weeks

The mean general mental state ‐ PANSS total score change ranged from −8.9 to 0.03 points

MD 1.44 points lower
(6.85 lower to 3.97 higher)

258
(3 RCTs)

⊕⊝⊝⊝
VERY LOW 3 4 5

One other trial used the BPRS total score change and showed no clear difference between the two groups.

Pre‐defined outcome: Clinically important change in general mental state not reported.

Adverse effects ‐ at least one adverse effect
assessed with: Risk ratio
follow‐up: range 2 weeks to 9 weeks

Study population

RR 0.91
(0.55 to 1.50)

191
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 1 6

716 per 1000

652 per 1000
(394 to 1000)

Service use: time in hospital

see comment

(0 studies)

No studies reported this outcome.

Quality of life ‐ clinically important change in quality of life (defined as at least 50% improvement in HQLS)

Study population

not estimable

17
(1 RCT)

⊕⊕⊝⊝
LOW 1

0 per 1000

0 per 1000
(0 to 0)

*The risk in the intervention group (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; OR: Odds ratio;

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 Imprecision for dichotomous outcomes: a) sample size should be >800 and/or total number of events>300; in our review, both numbers are much smaller. b) the pooled estimate of effect includes both no effect and an appreciable benefit or appreciable harm.

2 Inconsistency: Heterogeneity: Tau² = 1.03; Chi² = 11.24, df = 5 (P = 0.05); I² = 56%

3 Inconsistency: Heterogeneity: Tau² = 14.71; Chi² = 5.70, df = 2 (P = 0.06); I² = 65%

4 Imprecision for continuous outcomes: a) sample size is lower than 400, b) confidence interval includes no effect and the upper or lower confidence limit crosses the minimal important difference (MID), either for benefit of harm

5 Indirectness: The pre‐specified outcome (clinical important change in mental state) was not reported.

6 Inconsistency: Heterogeneity: Tau² = 0.10; Chi² = 3.62, df = 1 (P = 0.06); I² = 72%

Figuras y tablas -
Summary of findings for the main comparison. Antipsychotic dose increase compared to antipsychotic dose continuation for non response in schizophrenia
Comparison 1. Antipsychotic dose increase versus antipsychotic dose maintenance

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Global state: 1a. Clinically relevant response as defined by trials Show forest plot

9

533

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.86, 1.40]

2 Global state: 1b. Any change (improvement in Global Assesment scale) Show forest plot

1

23

Risk Ratio (M‐H, Random, 95% CI)

0.76 [0.32, 1.80]

3 Global state: 2a. Average endpoint score (CGI‐Severity , high = poor) Show forest plot

3

196

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.40, 0.19]

4 Global state: 2b. Average change score ( CGI‐Severity, high = poor) Show forest plot

1

95

Mean Difference (IV, Random, 95% CI)

‐0.4 [‐0.80, ‐0.00]

5 Global state: 2c. Average endpoint score (CGI‐Improvement, high = poor) Show forest plot

1

103

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.35, 0.35]

6 Leaving the study early: 1. Tolerability ‐ due to adverse events Show forest plot

7

496

Risk Ratio (M‐H, Random, 95% CI)

1.63 [0.52, 5.07]

7 Leaving the study early: 2. Acceptability of treatment ‐ due to any reason Show forest plot

5

353

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.89, 1.90]

8 Leaving the study early: 3. Efficacy ‐ due to inefficacy Show forest plot

4

336

Risk Ratio (M‐H, Random, 95% CI)

0.82 [0.30, 2.28]

9 Mental state: 1a. General ‐ average endpoint score (PANSS total, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

‐1.81 [‐7.31, 3.69]

10 Mental state: 1b. General ‐ average change score (PANSS total, high = poor) Show forest plot

3

258

Mean Difference (IV, Random, 95% CI)

‐1.44 [‐6.85, 3.97]

11 Mental state: 1c. General ‐ average endpoint and/or change score (PANSS total, high = poor) Show forest plot

4

389

Mean Difference (IV, Random, 95% CI)

‐2.13 [‐6.16, 1.90]

12 Mental state: 1d. General ‐ average endpoint score (BPRS total, high = poor) Show forest plot

3

99

Mean Difference (IV, Random, 95% CI)

‐1.25 [‐4.60, 2.11]

13 Mental state: 1e. General ‐ average change score (BPRS total, high = poor) Show forest plot

1

42

Mean Difference (IV, Random, 95% CI)

‐2.38 [‐6.15, 1.39]

14 Mental state: 1f. General ‐ average endpoint and/or change score (BPRS total, high = poor) Show forest plot

4

141

Mean Difference (IV, Random, 95% CI)

‐1.75 [‐4.25, 0.76]

15 Mental state: 1g. General ‐ average change score (NOSIE total, high = poor) Show forest plot

1

42

Mean Difference (IV, Random, 95% CI)

3.70 [‐5.38, 12.78]

16 Mental state: 2a. Positive symptoms ‐ Clinically important change Show forest plot

1

17

Risk Ratio (M‐H, Random, 95% CI)

1.33 [0.58, 3.07]

17 Mental state: 2b. Positive symptoms ‐ average endpoint subscore (PANSS positive, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

‐0.94 [‐2.79, 0.90]

18 Mental state: 2c. Positive symptoms ‐ average change subscore (PANSS positive, high = poor) Show forest plot

3

238

Mean Difference (IV, Random, 95% CI)

0.04 [‐1.31, 1.40]

19 Mental state: 2d. Positive symptoms ‐ average endpoint subscore (BPRS positive,high = poor) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

0.40 [‐2.94, 3.74]

20 Mental state: 3a. Negative symptoms ‐ average endpoint subscore (PANSS negative, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

0.32 [‐1.48, 2.11]

21 Mental state: 3b. Negative symptoms ‐ average change subscore (PANSS negative, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

‐0.15 [‐0.96, 0.67]

22 Mental state: 3c. Negative symptoms ‐ average endpoint subscore (BPRS negative, high = poor) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐1.97, 1.17]

23 Mental state: 3d. Negative symptoms ‐ average endpoint score (SANS, high = poor) Show forest plot

1

34

Mean Difference (IV, Random, 95% CI)

1.5 [‐14.33, 17.33]

24 Adverse effects ‐ At least one adverse effect Show forest plot

2

191

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.55, 1.50]

25 Adverse effects ‐ Cardiac: QTc prolongation Show forest plot

2

206

Risk Ratio (M‐H, Random, 95% CI)

2.47 [0.12, 50.39]

26 Adverse effects ‐ Cardiac: Orthostatic hypotension Show forest plot

2

102

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.25, 4.82]

27 Adverse effects ‐ Cardiac: Palpitations Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.70]

28 Adverse effects ‐ Cardiac: Premature Ventricular Contractions Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.13, 69.70]

29 Adverse effects ‐ Constipation Show forest plot

2

102

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.44, 5.38]

30 Adverse effects ‐ Dizziness Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.41, 1.44]

31 Adverse effects ‐ Drooling Show forest plot

1

42

Risk Ratio (M‐H, Random, 95% CI)

2.0 [0.20, 20.41]

32 Adverse effects ‐ Death (suicide or naturalistic cause) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

33 Adverse effects ‐ Extrapyramidal: Categorical deterioration (AIMS, high = poor) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.38 [0.59, 3.26]

34 Adverse effects ‐ Extrapyramidal: average endpoint score (AIMS, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.7 [‐0.87, 2.27]

35 Adverse effects ‐ Extrapyramidal: average change score (AIMS, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

0.41 [‐1.15, 1.96]

36 Adverse effects ‐ Extrapyramidal: akathisia Show forest plot

2

65

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.04, 14.02]

37 Adverse effects ‐ Extrapyramidal: categorical deterioration (BAS, high = poor) Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.47 [0.42, 5.14]

38 Adverse effects ‐ Extrapyramidal: average endpoint score (BAS, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐0.2 [‐0.74, 0.34]

39 Adverse effects ‐ Extrapyramidal: average change score (BAS, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.59, 0.37]

40 Adverse effects ‐ Extrapyramidal: dystonia and/or dyskinesia Show forest plot

3

196

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.09, 2.73]

41 Adverse effects ‐ Extrapyramidal: categorical deterioration in SAS Show forest plot

1

131

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.51, 2.93]

42 Adverse effects ‐ Extrapyramidal: average endpoint score (SAS, high = poor) Show forest plot

2

191

Mean Difference (IV, Random, 95% CI)

0.93 [‐1.04, 2.91]

43 Adverse effects ‐ Extrapyramidal: average change score (SA, high = poor) Show forest plot

2

163

Mean Difference (IV, Random, 95% CI)

0.21 [‐0.83, 1.26]

44 Adverse effects ‐ Extrapyramidal: tremor Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

1.59 [0.59, 4.26]

45 Adverse effects ‐ Headache Show forest plot

2

173

Risk Ratio (M‐H, Random, 95% CI)

1.46 [0.52, 4.08]

46 Adverse effects ‐ Somnolence and/or drowsiness Show forest plot

4

256

Risk Ratio (M‐H, Random, 95% CI)

1.76 [0.81, 3.81]

47 Adverse effects ‐ Weight at endpoint (high = poor) Show forest plot

2

165

Mean Difference (IV, Random, 95% CI)

‐1.85 [‐7.09, 3.39]

48 Behaviour: average endpoint score (BARS, high = good) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

‐0.10 [‐0.50, 0.30]

49 Functioning ‐ Global Assessment of Functioning: average change score (GAF, high = good) Show forest plot

1

103

Mean Difference (IV, Random, 95% CI)

‐0.60 [‐1.00, 1.80]

50 Functioning ‐ Social ‐ Adaptive Functioning Evaluation: average endpoint score (SAFE, high = poor) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

0.16 [‐0.47, 0.79]

51 Functioning ‐ Social and Occupational Functioning Assessment: average endpoint score (SOFAS, high = good) Show forest plot

1

131

Mean Difference (IV, Random, 95% CI)

0.5 [‐3.80, 4.80]

52 Quality of life: Clinically important change (at least 50% improvement HQLS, high = good) Show forest plot

1

17

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

53 Quality of life: Average endpoint score (HQLS, high = good) Show forest plot

1

17

Mean Difference (IV, Random, 95% CI)

5.5 [‐13.66, 24.66]

Figuras y tablas -
Comparison 1. Antipsychotic dose increase versus antipsychotic dose maintenance
Comparison 2. Subgroup analysis and investigation of heterogeneity

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Single antipsychotic drugs Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Fluphenazine

1

34

Risk Ratio (M‐H, Random, 95% CI)

2.25 [0.22, 22.53]

1.2 Haloperidol

2

71

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.67, 1.80]

1.3 Quetiapine

2

191

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.15, 2.86]

1.4 Risperidone

1

17

Risk Ratio (M‐H, Random, 95% CI)

9.9 [0.63, 155.08]

1.5 Thiothixene

1

42

Risk Ratio (M‐H, Random, 95% CI)

1.75 [0.94, 3.26]

1.6 Ziprasidone

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.45, 1.41]

2 Clinical state, stage or problem Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 First episode patients

1

17

Risk Ratio (M‐H, Random, 95% CI)

9.9 [0.63, 155.08]

2.2 Not first episode patients

8

516

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.86, 1.34]

Figuras y tablas -
Comparison 2. Subgroup analysis and investigation of heterogeneity
Comparison 3. Sensitivity analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Excluding studies with unclear randomisation methods Show forest plot

2

234

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.77, 1.44]

2 Excluding studies with unclear allocation concealment methods Show forest plot

2

234

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.77, 1.44]

3 Exclusion of studies with high risk of bias regarding blinding Show forest plot

8

516

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.86, 1.34]

4 Exclusion of studies with high risk of bias regarding incomplete outcome data Show forest plot

6

375

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.92, 1.54]

5 Exclusion of studies with imputed values Show forest plot

8

458

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.90, 1.50]

6 Fixed effects Show forest plot

9

533

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.88, 1.38]

Figuras y tablas -
Comparison 3. Sensitivity analysis