Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis

Andrea Cipriani, Toshi A Furukawa, Georgia Salanti, Anna Chaimani, Lauren Z Atkinson, Yusuke Ogawa, Stefan Leucht, Henricus G Ruhe, Erick H Turner, Julian P T Higgins, Matthias Egger, Nozomi Takeshima, Yu Hayasaka, Hissei Imai, Kiyomi Shinohara, Aran Tajika, John P A Ioannidis, John R Geddes, Andrea Cipriani, Toshi A Furukawa, Georgia Salanti, Anna Chaimani, Lauren Z Atkinson, Yusuke Ogawa, Stefan Leucht, Henricus G Ruhe, Erick H Turner, Julian P T Higgins, Matthias Egger, Nozomi Takeshima, Yu Hayasaka, Hissei Imai, Kiyomi Shinohara, Aran Tajika, John P A Ioannidis, John R Geddes

Abstract

Background: Major depressive disorder is one of the most common, burdensome, and costly psychiatric disorders worldwide in adults. Pharmacological and non-pharmacological treatments are available; however, because of inadequate resources, antidepressants are used more frequently than psychological interventions. Prescription of these agents should be informed by the best available evidence. Therefore, we aimed to update and expand our previous work to compare and rank antidepressants for the acute treatment of adults with unipolar major depressive disorder.

Methods: We did a systematic review and network meta-analysis. We searched Cochrane Central Register of Controlled Trials, CINAHL, Embase, LILACS database, MEDLINE, MEDLINE In-Process, PsycINFO, the websites of regulatory agencies, and international registers for published and unpublished, double-blind, randomised controlled trials from their inception to Jan 8, 2016. We included placebo-controlled and head-to-head trials of 21 antidepressants used for the acute treatment of adults (≥18 years old and of both sexes) with major depressive disorder diagnosed according to standard operationalised criteria. We excluded quasi-randomised trials and trials that were incomplete or included 20% or more of participants with bipolar disorder, psychotic depression, or treatment-resistant depression; or patients with a serious concomitant medical illness. We extracted data following a predefined hierarchy. In network meta-analysis, we used group-level data. We assessed the studies' risk of bias in accordance to the Cochrane Handbook for Systematic Reviews of Interventions, and certainty of evidence using the Grading of Recommendations Assessment, Development and Evaluation framework. Primary outcomes were efficacy (response rate) and acceptability (treatment discontinuations due to any cause). We estimated summary odds ratios (ORs) using pairwise and network meta-analysis with random effects. This study is registered with PROSPERO, number CRD42012002291.

Findings: We identified 28 552 citations and of these included 522 trials comprising 116 477 participants. In terms of efficacy, all antidepressants were more effective than placebo, with ORs ranging between 2·13 (95% credible interval [CrI] 1·89-2·41) for amitriptyline and 1·37 (1·16-1·63) for reboxetine. For acceptability, only agomelatine (OR 0·84, 95% CrI 0·72-0·97) and fluoxetine (0·88, 0·80-0·96) were associated with fewer dropouts than placebo, whereas clomipramine was worse than placebo (1·30, 1·01-1·68). When all trials were considered, differences in ORs between antidepressants ranged from 1·15 to 1·55 for efficacy and from 0·64 to 0·83 for acceptability, with wide CrIs on most of the comparative analyses. In head-to-head studies, agomelatine, amitriptyline, escitalopram, mirtazapine, paroxetine, venlafaxine, and vortioxetine were more effective than other antidepressants (range of ORs 1·19-1·96), whereas fluoxetine, fluvoxamine, reboxetine, and trazodone were the least efficacious drugs (0·51-0·84). For acceptability, agomelatine, citalopram, escitalopram, fluoxetine, sertraline, and vortioxetine were more tolerable than other antidepressants (range of ORs 0·43-0·77), whereas amitriptyline, clomipramine, duloxetine, fluvoxamine, reboxetine, trazodone, and venlafaxine had the highest dropout rates (1·30-2·32). 46 (9%) of 522 trials were rated as high risk of bias, 380 (73%) trials as moderate, and 96 (18%) as low; and the certainty of evidence was moderate to very low.

Interpretation: All antidepressants were more efficacious than placebo in adults with major depressive disorder. Smaller differences between active drugs were found when placebo-controlled trials were included in the analysis, whereas there was more variability in efficacy and acceptability in head-to-head trials. These results should serve evidence-based practice and inform patients, physicians, guideline developers, and policy makers on the relative merits of the different antidepressants.

Funding: National Institute for Health Research Oxford Health Biomedical Research Centre and the Japan Society for the Promotion of Science.

Copyright © 2018 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study selection process RCTs=randomised controlled trials. *Industry websites, contact with authors, and trial registries. The total number of unpublished records is the total number of results for each drug and on each unpublished database source. †522 RCTs corresponded to 814 treatment groups.
Figure 2
Figure 2
Network meta-analysis of eligible comparisons for efficacy (A) and acceptability (B) Width of the lines is proportional to the number of trials comparing every pair of treatments. Size of every circle is proportional to the number of randomly assigned participants (ie, sample size).
Figure 3
Figure 3
Forest plots of network meta-analysis of all trials for efficacy (A) and acceptability (B) Antidepressants were compared with placebo, which was the reference compound. OR=odds ratio. CrI=credible interval.
Figure 4
Figure 4
Head-to-head comparisons for efficacy and acceptability of the 21 antidepressants Drugs are reported in alphabetical order. Data are ORs (95% CrI) in the column-defining treatment compared with the row-defining treatment. For efficacy, ORs higher than 1 favour the column-defining treatment (ie, the first in alphabetical order). For acceptability, ORs lower than 1 favour the first drug in alphabetical order. To obtain ORs for comparisons in the opposite direction, reciprocals should be taken. Significant results are in bold and underscored. The certainty of the evidence (according to GRADE) was incorporated in this figure (appendix pp 231–65). OR=odds ratio. CrI=credible interval. Agom=agomelatine. Amit=amitriptyline. Bupr=bupropion. Cita=citalopram. Clom=clomipramine. Dulo=duloxetine. Esci=escitalopram. Fluo=fluoxetine. Fluv=fluvoxamine. Miln=milnacipran. Mirt=mirtazapine. Nefa=nefazodone. Paro=paroxetine. Rebo=reboxetine. Sert=sertraline. Traz=trazodone. Venl=venlafaxine. Vort=vortioxetine. *Moderate quality of evidence. †Low quality of evidence. ‡Very low quality of evidence.
Figure 5
Figure 5
Two-dimensional graphs about efficacy and acceptability in all studies (A) and head-to-head (B) studies only Data are reported as ORs in comparison with reboxetine, which is the reference drug. Error bars are 95% CrIs. Individual drugs are represented by different coloured nodes. Desvenlafaxine, levomilnacipran, and vilazodone were not included in the head-to-head analysis because these three antidepressants had only placebo-controlled trials. ORs=odds ratios. 1=agomelatine. 2=amitriptyline. 3=bupropion. 4=citalopram. 5=clomipramine. 6=desvenlafaxine. 7=duloxetine. 8=escitalopram. 9=fluoxetine. 10=fluvoxamine. 11=levomilnacipran. 12=milnacipran. 13=mirtazapine. 14=nefazodone. 15=paroxetine. 16=reboxetine. 17=sertraline. 18=trazodone. 19=venlafaxine. 20=vilazodone. 21=vortioxetine. 22=placebo.

References

    1. GBD 2013 DALYs and HALE Collaborators. Murray CJ, Barber RM. Global, regional, and national disability-adjusted life years (DALYs) for 306 diseases and injuries and healthy life expectancy (HALE) for 188 countries, 1990–2013: quantifying the epidemiological transition. Lancet. 2015;386:2145–2191.
    1. GBD 2015 DALYs and HALE Collaborators Global, regional, and national disability-adjusted life-years (DALYs) for 315 diseases and injuries and healthy life expectancy (HALE), 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388:1603–1658.
    1. WHO . World Health Organisation; Geneva: 2017. Depression: fact sheet. (accessed Sept 21, 2017).
    1. Ioannidis JP. Effectiveness of antidepressants: an evidence myth constructed from a thousand randomized trials? Philos Ethics Humanit Med. 2008;3:14.
    1. Harmer CJ, Duman RS, Cowen PJ. How do antidepressants work? New perspectives for refining future treatment approaches. Lancet Psychiatry. 2017;4:409–418.
    1. Higgins JP, Welton NJ. Network meta-analysis: a norm for comparative effectiveness? Lancet. 2015;386:628–630.
    1. Hróbjartsson A, Thomsen AS, Emanuelsson F. Observer bias in randomized clinical trials with measurement scale outcomes: a systematic review of trials with both blinded and nonblinded assessors. CMAJ. 2013;185:E201–E211.
    1. Furukawa TA, Salanti G, Atkinson LZ. Comparative efficacy and acceptability of first-generation and second-generation antidepressants in the acute treatment of major depression: protocol for a network meta-analysis. BMJ Open. 2016;6:e010919.
    1. Cipriani A, Furukawa TA, Salanti G. Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatment meta-analysis. Lancet. 2009;373:746–758.
    1. Furukawa TA, Cipriani A, Barbui C, Brambilla P, Watanabe N. Imputing response rates from means and standard deviations in meta-analyses. Int Clin Psychopharmacol. 2005;20:49–52.
    1. Rhodes KM, Turner RM, Higgins JP. Predictive distributions were developed for the extent of heterogeneity in meta-analyses of continuous outcome data. J Clin Epidemiol. 2015;68:52–60.
    1. Salanti G, Ades AE, Ioannidis JP. Graphical methods and numerical summaries for presenting results from multiple-treatment meta-analysis: an overview and tutorial. J Clin Epidemiol. 2011;64:163–171.
    1. Higgins JP, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta-analysis: concepts and models for multi-arm studies. Res Synth Methods. 2012;3:98–110.
    1. Dias S, Welton NJ, Caldwell DM, Ades AE. Checking consistency in mixed treatment comparison meta-analysis. Stat Med. 2010;29:932–944.
    1. Salanti G, Del Giovane C, Chaimani A, Caldwell DM, Higgins JP. Evaluating the quality of evidence from a network meta-analysis. PLoS One. 2014;9:e99682.
    1. Salanti G, Dias S, Welton NJ. Evaluating novel agent effects in multiple-treatments meta-regression. Stat Med. 2010;29:2369–2383.
    1. Chaimani A, Higgins JP, Mavridis D, Spyridonos P, Salanti G. Graphical tools for network meta-analysis in STATA. PLoS One. 2013;8:e76654.
    1. Lunn D, Spiegelhalter D, Thomas A. The BUGS project: evolution, critique and future directions. Stat Med. 2009;28:3049–3067.
    1. Chaimani A, Salanti G. Visualizing assumptions and results in network meta-analysis: the network graphs package. Stata J. 2015;15:905–950.
    1. Schwarzer G. Network meta-analysis. In: Schwarzer G, Carpenter JR, Rücker G, editors. Meta-analysis with R. Springer; Berlin: 2015. pp. 187–216.
    1. Cipriani A, Zhou X, Del Giovane C. Comparative efficacy and tolerability of antidepressants for major depressive disorder in children and adolescents: a network meta-analysis. Lancet. 2016;388:881–890.
    1. Thapar A, Collishaw S, Pine DS, Thapar AK. Depression in adolescence. Lancet. 2012;379:1056–1067.
    1. Walkup JT. Antidepressant efficacy for depression in children and adolescents: industry- and NIMH-funded studies. Am J Psychiatry. 2017;174:430–437.
    1. Rutherford BR, Roose SP. A model of placebo response in antidepressant clinical trials. Am J Psychiatry. 2013;170:723–733.
    1. Cook RJ, Zeng L, Yi GY. Marginal analysis of incomplete longitudinal binary data: a cautionary note on LOCF imputation. Biometrics. 2004;60:820–828.
    1. Furukawa TA, Cipriani A, Atkinson LZ. Placebo response rates in antidepressant trials: a systematic review of published and unpublished double-blind randomised controlled studies. Lancet Psychiatry. 2016;3:1059–1066.
    1. Perlis RH, Perlis CS, Wu Y, Hwang C, Joseph M, Nierenberg AA. Industry sponsorship and financial conflict of interest in the reporting of clinical trials in psychiatry. Am J Psychiatry. 2005;162:1957–1960.
    1. Turner EH, Matthews AM, Linardatos E, Tell RA, Rosenthal R. Selective publication of antidepressant trials and its influence on apparent efficacy. N Engl J Med. 2008;358:252–260.
    1. Furukawa TA, Akechi T, Shimodera S. Strategic use of new generation antidepressants for depression: SUN(^_^)D study protocol. Trials. 2011;12:116.
    1. Furukawa TA, Miura T, Chaimani A. Using the contribution matrix to evaluate complex study limitations in a network meta-analysis: a case study of bipolar maintenance pharmacotherapy review. BMC Res Notes. 2016;9:218.
    1. Wager E. The need for trial identifiers. Curr Med Res Opin. 2004;20:203–206.
    1. Ioannidis JP. Meta-analyses can be credible and useful: a new standard. JAMA Psychiatry. 2017;74:311–312.

Source: PubMed

3
Subskrybuj