Scolaris Content Display Scolaris Content Display

Imunomodulatori i imunosupresivi za relapsno‐remitirajuću multiplu sklerozu: mrežna meta‐analiza

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Achiron 1998 {published data only}

Achiron A, Gabbay U, Gilad R, Hassin B, Barak Y, Gornish M, et al. Intravenous immunoglobulin treatment in multiple sclerosis. Effect on relapses. Neurology 1998;50(2):398‐402.

ADVANCE 2014 {published data only}

Calabresi PA, Kieseier BC, Arnold DL, Balcer LJ, Boyko A, Pelletier J, et al. Pegylated interferon beta‐1a for relapsing‐remitting multiple sclerosis (ADVANCE): a randomised, phase 3, double‐blind study. Lancet Neurology 2014;13:657‐65.

AFFIRM 2006 {published data only}

Polman CH, O'Connor PW, Havrdova E, Hutchinson M, Kappos L, Miller DH, et al. A randomized, placebo‐controlled trial of natalizumab for relapsing multiple sclerosis. New England Journal of Medicine 2006;354(9):899‐910.

ALLEGRO 2012 {published data only}

Comi G, Jeffery D, Kappos L, Montalban X, Boyko A, Rocca MA, et al. Placebo‐controlled trial of oral laquinimod for multiple sclerosis. New England Journal of Medicine 2012;366(11):1000‐9.

BECOME 2009 {published data only}

Cadavid D, Wolansky LJ, Skurnick J, Lincoln J, Cheriyan J, Szczepanowski K, et al. Efficacy of treatment of MS with IFNbeta‐1b or glatiramer acetate by monthly brain MRI in the BECOME study. Neurology 2009;72(23):1976‐83.

BEYOND 2009 {published data only}

O'Connor P, Filippi M, Arnason B, Comi G, Cook S, Goodin D, et al. 250 microg or 500 microg interferon beta‐1b versus 20 mg glatiramer acetate in relapsing‐remitting multiple sclerosis: a prospective, randomised, multicentre study. Lancet Neurology 2009;8(10):889‐97.

Bornstein 1987 {published data only}

Bornstein MB, Miller A, Slagle S, Weitzman M, Crystal, Drexler E, et al. A pilot trial of Cop 1 in exacerbating‐remitting multiple sclerosis. New England Journal of Medicine 1987;317(7):408‐14.

BRAVO 2014 {published data only}

Vollmer TL, Sorensen PS, Selmaj K, Zipp F, Havrdova E, Cohen JA, et al. A randomized placebo‐controlled phase III trial of oral laquinimod for multiple sclerosis. Journal of Neurology 2014;261(4):773‐83.

CAMMS223 2008 {published data only}

CAMMS223 Trial Investigators, Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, et al. Alemtuzumab vs. interferon beta‐1a in early multiple sclerosis. New England Journal of Medicine 2008;359(17):1786‐801.

CARE‐MS I 2012 {published data only}

Cohen JA, Coles AJ, Arnold DL, Confavreux C, Fox EJ, Hartung HP, et al. Alemtuzumab versus interferon beta 1a as first‐line treatment for patients with relapsing‐remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet 2012;380(9856):1819‐28.

CARE‐MS II 2012 {published data only}

Coles AJ, Twyman CL, Arnold DL, Cohen JA, Confavreux C, Fox EJ, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease‐modifying therapy: a randomised controlled phase 3 trial. Lancet 2012;380(9856):1829‐39.

CombiRx 2013 {published data only}

Lublin FD, Cofield SS, Cutter GR, Conwit R, Narayana PA, Nelson F, et al. Randomized study combining interferon and glatiramer acetate in multiple sclerosis. Annals of Neurology 2013;73(3):327‐40.

Comi 2001 {published data only}

Comi G, Filippi M, Wolinsky JS. European/Canadian multicenter, double‐blind, randomized, placebo‐controlled study of the effects of glatiramer acetate on magnetic resonance imaging‐‐measured disease activity and burden in patients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate Study Group. Annals of Neurology 2001;49(3):290‐7. [PUBMED: 11261502]

CONFIRM 2012 {published data only}

Fox RJ, Miller DH, Phillips JT, Hutchinson M, Havrdova E, Kita M, et al. Placebo‐controlled phase 3 study of oral BG‐12 or glatiramer in multiple sclerosis. New England Journal of Medicine 2012;367(12):1087‐97.

DEFINE 2012 {published data only}

Gold R, Kappos L, Arnold DL, Bar‐Or A, Giovannoni G, Selmaj K, et al. Placebo‐controlled phase 3 study of oral BG‐12 for relapsing multiple sclerosis. New England Journal of Medicine 2012;367(12):1098‐107.

Etemadifar 2007 {published data only}

Etemadifar M, Janghorbani M, Shaygannejad V. Comparison of interferon beta products and azathioprine in the treatment of relapsing‐remitting multiple sclerosis. Journal of Neurology 2007;254(12):1723‐8.

EVIDENCE 2007 {published data only}

Schwid S, Panitch H. Full results of the Evidence of Interferon Dose‐Response‐European North American Comparative Efficacy (EVIDENCE) study: a multicenter, randomized, assessor‐blinded comparison of low‐dose weekly versus high‐dose, high‐frequency interferon beta‐1a for relapsing multiple sclerosis. Clinical Therapeutics 2007;29(9):2031‐48.

Fazekas 1997 {published data only}

Fazekas F, Deisenhammer F, Strasser‐Fuchs S, Nahler G, Mamoli B. Randomised placebo‐controlled trial of monthly intravenous immunoglobulin therapy in relapsing‐remitting multiple sclerosis. Lancet 1997;349(9052):589‐93.

Fazekas 2008 {published data only}

Fazekas F, Lublin F, Li D, Freedman M, Hartung H, Rieckmann P, et al. Intravenous immunoglobulin in relapsing‐remitting multiple sclerosis: a dose‐finding trial. Neurology 2008;71(4):265‐71.

FREEDOMS 2010 {published data only}

Kappos L, Radue EW, O'Connor P, Polman C, Hohlfeld R, Calabresi P, et al. A placebo‐controlled trial of oral fingolimod in relapsing multiple sclerosis. New England Journal of Medicine 2010;362(5):387‐401.

FREEDOMS II 2014 {published data only}

Calabresi PA, Radue EW, Goodin D, Jeffery D, Rammohan KW, Reder AT, et al. Safety and efficacy of fingolimod in patients with relapsing‐remitting multiple sclerosis (FREEDOMS II): a double‐blind, randomised, placebo‐controlled, phase 3 trial. Lancet Neurology 2014;13(6):545‐56.

GALA 2013 {published data only}

Khan O, Rieckmann P, Boyko A, Selmaj K, Zivadinov R, GALA Study Group. Three times weekly glatiramer acetate in relapsing‐remitting multiple sclerosis. Annals of Neurology 2013;73(6):705‐13.

Goodkin 1991 {published data only}

Goodkin D, Bailly R, Teetzen M, Hertsgaard D, Beatty W. The efficacy of azathioprine in relapsing‐remitting multiple sclerosis. Neurology 1991;41:20‐5.

IFNB MS Group 1993 {published data only}

IFNB MSG. Interferon beta‐1b is effective in relapsing‐remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double‐blind, placebo‐controlled trial. The IFNB Multiple Sclerosis Study Group. Neurology 1993;43(4):655‐61.

INCOMIN 2002 {published data only}

Durelli L, Verdun E, Barbero P, Bergui M, Versino E, Ghezzi A, et al. Every‐other‐day interferon beta‐1b versus once‐weekly interferon beta‐1a for multiple sclerosis: results of a 2‐year prospective randomised multicentre study (INCOMIN). Lancet 2002;359:1453‐60.

Johnson 1995 {published data only}

Johnson K, Brooks B, Cohen J, Ford C, Goldstein J, Lisak R, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing‐remitting multiple sclerosis: results of a phase III multicenter, double‐blind placebo‐controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology 1995;45(7):1268‐76.

Koch‐Henriksen 2006 {published data only}

Koch‐Henriksen N, Sørensen P, Christensen T, Frederiksen J, Ravnborg M, Jensen K, et al. A randomised study of two interferon‐beta treatments in relapsing‐remitting multiple sclerosis. Neurology 2006;66(7):1056‐60.

Lewanska 2002 {published data only}

Lewanska M, Siger‐Zajdel M, Selmaj K. No difference in efficacy of two different doses of intravenous immunoglobulins in MS: clinical and MRI assessment. European Journal of Neurology 2002;9(6):565‐72.

MAIN TRIAL {published data only}

Massacesi L, Tramacere I, Amoroso S, Battaglia MA, Benedetti MD, Filippini G, et al. Azathioprine versus beta interferons for relapsing‐remitting multiple sclerosis: a multicentre randomized non‐inferiority trial. PLoS One 2014;9(11):e113371.

Millefiorini 1997 {published data only}

Millefiorini E, Gasperini C, Pozzilli C, D'Andrea F, Bastianello S, Trojano M, et al. Randomized placebo‐controlled trial of mitoxantrone in relapsing‐remitting multiple sclerosis: 24‐month clinical and MRI outcome. Journal of Neurology 1997;244(3):153‐9.

MSCRG 1996 {published data only}

Jacobs L, Cookfair D, Rudick R, Herndon R, Richert J, Salazar A, et al. Intramuscular interferon beta‐1a for disease progression in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Annals of Neurology 1996;39:285‐94.

OWIMS 1999 {published data only}

OWIMS. Evidence of interferon beta‐1a dose response in relapsing‐remitting MS: the OWIMS Study. The Once Weekly Interferon for MS Study Group. Neurology 1999;53(4):679‐86.

PRISMS 1998 {published data only}

PRISMS. Randomised double‐blind placebo‐controlled study of interferon beta‐1a in relapsing/remitting multiple sclerosis. PRISMS (Prevention of Relapses and Disability by Interferon beta‐1a Subcutaneously in Multiple Sclerosis) Study Group. Lancet 1998;352(9139):1498‐504.

REGARD 2008 {published data only}

Mikol D, Barkhof F, Chang P, Coyle P, Jeffery D, Schwid S, et al. Comparison of subcutaneous interferon beta‐1a with glatiramer acetate in patients with relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in Relapsing MS Disease [REGARD] study): a multicentre, randomised, parallel, open‐label trial. Lancet Neurology 2008;7(10):903‐14.

SELECT 2013 {published data only}

Gold R, Giovannoni G, Selmaj K, Havrdova E, Montalban X, Radue EW, et al. Daclizumab high‐yield process in relapsing‐remitting multiple sclerosis (SELECT): a randomised, double‐blind, placebo‐controlled trial. Lancet 2013;381(9884):2167‐75.

TEMSO 2011 {published data only}

O'Connor P, Wolinsky JS, Confavreux C, Comi G, Kappos L, Olsson TP, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. New England Journal of Medicine 2011;365(14):1293‐303.

TENERE 2014 {published data only}

Vermersch P, Czlonkowska A, Grimaldi LM, Confavreux C, Comi G, Kappos L, et al. Teriflunomide versus subcutaneous interferon beta‐1a in patients with relapsing multiple sclerosis: a randomised, controlled phase 3 trial. Multiple Sclerosis (Houndmills, Basingstoke, England) 2014;20(6):705‐16.

TOWER 2014 {published data only}

Confavreux C, O'Connor P, Comi G, Freedman MS, Miller AE, Olsson TP, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Neurology 2014;13(3):247‐56.

TRASFORMS 2010 {published data only}

Cohen JA, Barkhof F, Comi G, Hartung HP, Khatri BO, Montalban X, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. New England Journal of Medicine 2010;362(5):402‐15.

References to studies excluded from this review

ACT 2009 {published data only}

Cohen JA, Imrey PB, Calabresi PA, Edwards KR, Eickenhorst T, Felton WL, et al. Results of the Avonex Combination Trial (ACT) in relapsing‐remitting MS. Neurology 2009;72(6):535‐41.

Ashtari 2011 {published data only}

Ashtari F, Savoj MR. Effects of low dose methotrexate on relapsing‐remitting multiple sclerosis in comparison to Interferon β‐1α: a randomized controlled trial. Journal of Research in Medical Sciences 2011;16(4):457‐62.

ATAMS 2014 {published data only}

Kappos L, Hartung HP, Freedman MS, Boyko A, Radü EW, Mikol DD, et al. Atacicept in multiple sclerosis (ATAMS): a randomised, placebo‐controlled, double‐blind, phase 2 trial. Lancet Neurology 2014;13(4):353‐63.

Calabrese 2012 {published data only}

Calabrese M, Bernardi V, Atzori M, Mattisi I, Favaretto A, Rinaldi F, et al. Effect of disease‐modifying drugs on cortical lesions and atrophy in relapsing‐remitting multiple sclerosis. Multiple Sclerosis (Houndmills, Basingstoke, England) 2012;18(4):418‐24.

CHOICE 2010 {published data only}

Wynn D, Kaufman M, Montalban X, Vollmer T, Simon J, Elkins J, et al. Daclizumab in active relapsing multiple sclerosis (CHOICE study): a phase 2, randomised, double‐blind, placebo‐controlled, add‐on trial with interferon beta. Lancet Neurology 2010;9(4):381‐90.

Etemadifar 2006 {published data only}

Etemadifar M, Janghorbani M, Shaygannejad V. Comparison of Betaferon, Avonex, and Rebif in treatment of relapsing‐remitting multiple sclerosis. Acta Neurologica Scandinavica 2006;113(5):283‐7.

FORTE 2011 {published data only}

Comi G, Cohen JA, Arnold DL, Wynn D, Filippi M, FORTE Study Group. Phase III dose‐comparison study of glatiramer acetate for multiple sclerosis. Annals of Neurology 2011;69(1):75‐82.

Freedman 2012 {published data only}

Freedman MS, Wolinsky JS, Wamil B, Confavreux C, Comi G, Kappos L, et al. Teriflunomide added to interferon‐β in relapsing multiple sclerosis: a randomized phase II trial. Neurology 2012;78(23):1877‐85.

Havrdova 2009 {published data only}

Havrdova E, Zivadinov R, Krasensky J, Dwyer MG, Novakova I, Dolezal O, et al. Randomized study of interferon beta‐1a, low‐dose azathioprine, and low‐dose corticosteroids in multiple sclerosis. Multiple Sclerosis (Houndmills, Basingstoke, England) 2009;15(8):965‐76.

Kappos 2006 {published data only}

Kappos L, Antel J, Comi G, Montalban X, O'Connor P, Polman CH, et al. Oral fingolimod (FTY720) for relapsing multiple sclerosis. New Egyptian Journal of Medicine 2006;355(11):1124‐40.

Kappos 2008 {published data only}

Kappos L, Gold R, Miller DH, Macmanus DG, Havrdova E, Limmroth V, et al. Efficacy and safety of oral fumarate in patients with relapsing‐remitting multiple sclerosis: a multicentre, randomised, double‐blind, placebo‐controlled phase IIb study. Lancet 2008;372(9648):1463‐72.

Kappos 2011 {published data only}

Kappos L, Li D, Calabresi PA, O'Connor P, Bar‐Or A, Barkhof F, et al. Ocrelizumab in relapsing‐remitting multiple sclerosis: a phase 2, randomised, placebo‐controlled, multicentre trial. Lancet 2011;378(9805):1779‐87.

Khoury 2010 {published data only}

Khoury SJ, Healy BC, Kivisäkk P, Viglietta V, Egorova S, Guttmann CR, et al. A randomized controlled double‐masked trial of albuterol add‐on therapy in patients with multiple sclerosis. Archives of Neurology 2010;67(9):1055‐61.

Knobler 1993 {published data only}

Knobler RL, Greenstein JI, Johnson KP, Lublin FD, Panitch HS, Conway K, et al. Systemic recombinant human interferon‐beta treatment of relapsing‐remitting multiple sclerosis: pilot study analysis and six‐year follow‐up. Journal of Interferon Research 1993;13(5):333‐40.

Saida 2012 {published data only}

Saida T, Kikuchi S, Itoyama Y, Hao Q, Kurosawa T, Nagato K, et al. A randomized, controlled trial of fingolimod (FTY720) in Japanese patients with multiple sclerosis. Multiple Sclerosis (Houndmills, Basingstoke, England) 2012;18(9):1269‐77.

SENTINEL 2006 {published data only}

Rudick RA, Stuart WH, Calabresi PA, Confavreux C, Galetta SL, Radue EW, et al. Natalizumab plus interferon beta‐1a for relapsing multiple sclerosis. New England Journal of Medicine 2006;354(9):911‐23.

Sorensen 2014 {published data only}

Sorensen PS, Lisby S, Grove R, Derosier F, Shackelford S, Havrdova E, et al. Safety and efficacy of ofatumumab in relapsing‐remitting multiple sclerosis: a phase 2 study. Neurology 2014;82(7):573‐81.

DECIDE {published data only}

Multicenter, double‐blind, randomized, parallel‐group, monotherapy, active‐control study to determine the efficacy and safety of daclizumab high yield process (DAC HYP) versus Avonex® (interferon β 1a) in patients with relapsing‐remitting multiple sclerosis. https://clinicaltrials.gov/ct2/show/study/NCT01064401 (accessed 30 September 2014).

NCT01247324 {published data only}

A randomized, double‐blind, double‐dummy, parallel‐group study to evaluate the efficacy and safety of Ocrelizumab in comparison to interferon beta‐1a (Rebif®) in patients with relapsing multiple sclerosis. https://clinicaltrials.gov/ct2/show/study/NCT01247324 (accessed 30 September 2014).

NCT01412333 {published data only}

A randomized, double‐blind, double‐dummy, parallel‐group study to evaluate the efficacy and safety of Ocrelizumab in comparison to Interferon Beta‐1a (Rebif®) in patients with relapsing multiple sclerosis. https://clinicaltrials.gov/ct2/show/study/NCT01412333 (accessed 30 September 2014).

Aharoni 2014

Aharoni R. Immunomodulation neuroprotection and remyelination. The fundamental therapeutic effects of glatiramer acetate: a critical review. Journal of Autoimmunity 2014;54:81‐92. [DOI: 10.1016/j.jaut.2014.05.005]

Association of British Neurologists 2005

Association of British Neurologists 2005. Guidelines for the use of intravenous immunoglobulin in neurological diseases. www.theabn.org/documents/IVIg‐Guidelines(accessed October 2013).

Bennett 2009

Bennett J, Stüve O. Update on inflammation, neurodegeneration, and immunoregulation in multiple sclerosis: therapeutic implications. Clinical Neuropharmacology 2009;32(3):121‐32.

Birnbaum 2008

Birnbaum G, Cree B, Altafullah I, Zinser M, Reder A. Combining beta interferon and atorvastatin may increase disease activity in multiple sclerosis. Neurology 2008;71(18):1390–5.

Caldwell 2005

Caldwell D, Ades A, Higgins J. Simultaneous comparison of multiple treatments: combining direct and indirect evidence as well. BMJ 2005;331(7521):897‐900.

Carroll 2014

Carroll CA, Fairman KA, Lage MJ. Updated cost‐of‐care estimates for commercially insured patients with multiple sclerosis: retrospective observational analysis of medical and pharmacy claims data. BMC Health Services Research 2014;14:286. [http://www.biomedcentral.com/1472‐6963/14/286]

Chaimani 2012

Chaimani A, Salanti G. Using network meta‐analysis to evaluate the existence of small‐study effects in a network of interventions. Research Synthesis Methods 2012;3(2):161‐76.

Chaimani 2013

Chaimani A, Higgins JP, Mavridis D, Spyridonos P, Salanti G. Graphical tools for network meta‐analysis in Stata. PLoS One 2013;8(10):e76654.

Claussen 2012

Claussen MC, Korn T. Immune mechanisms of new therapeutic strategies in MS: teriflunomide. Clinical Immunology 2012;142:49‐56.

Compston 2008

Compston A, Coles A. Multiple sclerosis. Lancet 2008;372:1502‐17.

DerSimonian 1986

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

Ebers 2008

Ebers G, Heigenhauser L, Daumer M, Lederer C, Noseworthy J. Disability as an outcome in MS clinical trials. Neurology 2008;71:624–31.

Elovaara 2008

Elovaara I, Apostolski S, van Doorn P, Gilhus N, Hietaharju A, Honkaniemi J, et al. EFNS guidelines for the use of intravenous immunoglobulin in treatment of neurological diseases: EFNS task force on the use of intravenous immunoglobulin in treatment of neurological diseases. European Journal of Neurology 2008;15(9):893‐908.

EMA 2006

European Agency for the Evaluation of Medicinal Products. Committee for proprietary medicinal products European public assessment report: Tysabri. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000603/wapp/Post‐authorisation/human_wapp_000166.jsp&mid=WC0b01ac058001d128(accessed June 2014).

EMA 2011

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Gilenya. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002202/human_med_001433.jsp&mid=WC0b01ac058001d124(accessed June 2014).

EMA 2013a

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Aubagio. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002514/human_med_001645.jsp&mid=WC0b01ac058001d124(accessed June 2014).

EMA 2013b

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Lemtrada. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Product_Information/human/003718/WC500150521.pdf(accessed June 2014).

EMA 2014a

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Tecfidera. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Product_Information/human/002601/WC500162069.pdf(accessed June 2014).

EMA 2014b

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Plegridy. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Summary_for_the_public/human/002827/WC500170305.pdf(accessed June 2015).

EMA 2014c

European Medicines Agency. Refusal of the marketing authorisation for Nerventra (laquinimod). http://www.ema.europa.eu/docs/en_GB/document_library/Summary_of_opinion_‐_Initial_authorisation/human/002546/WC500160120.pdf(accessed June 2014).

EMEA 1997

European Agency for the Evaluation of Medicinal Products. Committee for proprietary medicinal products European public assessment report: Avonex. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Summary_for_the_public/human/000102/WC500029423.pdf(accessed June 2015).

EMEA 1998

European Agency for the Evaluation of Medicinal Products. Committee for proprietary medicinal products European public assessment report: Rebif. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Assessment_Report_‐_Variation/human/000136/WC500122183.pdf(accessed June 2014).

EMEA 2002

European Agency for the Evaluation of Medicinal Products. Committee for proprietary medicinal products European public assessment report: Betaferon. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000081/human_med_000673.jsp&mid=WC0b01ac058001d124(accessed June 2014).

FDA 1993

U.S. Food, Drug Administration. Betaseron interferon beta‐1b subcutaneous. Drug Approval Package ‐ Licensing Action 1993. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/103471s5063s5067s5079s5088s5120s5124s5136s5138lbl.pdf(accessed June 2014).

FDA 1996

U.S. Food, Drug Administration. Glatiramer acetate (Capoxane) Product Approval Information ‐ Licensing Action 1996. http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Label_ApprovalHistory#apphist(accessed June 2014).

FDA 2000

U.S. Food, Drug Administration. Mitoxantrone (Novantrone) Product Approval Information ‐ Licensing Action 2000. http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Overview&DrugName=NOVANTRONE&CFID=17505855&CFTOKEN=fca8add70180e2b1‐F1C49694‐F504‐892A‐3B263F68EAFF59DC(accessed June 2014).

FDA 2002

U.S. Food, Drug Administration. Interferon beta‐1a (Rebif) Product Approval Information ‐ Licensing Action 2002. http://www.fda.gov/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/ucm080737.htm(accessed June 2014).

FDA 2003

U.S. Food, Drug Administration. Interferon beta‐1a (Avonex) Product Approval Information ‐ Licensing Action 2003. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2003/103628s5021TOC.cfm(accessed June 2014).

FDA 2004

U.S. Food, Drug Administration. Tysabri (Natalizumab) Product Approval Information 2004. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/125104s000_Natalizumab.cfm(accessed June 2014).

FDA 2006

U.S. Food, Drug Administration. FDA Approves Resumed Marketing of Tysabri Under a Special Distribution Program. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2006/ucm108662.htm(accessed June 2014).

FDA 2010

U.S. Food, Drug Administration. Gilenya (Fingolimod) Product Approval Information 2010. http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Set_Current_Drug&ApplNo=022527&DrugName=GILENYA&ActiveIngred=FINGOLIMOD&SponsorApplicant=NOVARTIS&ProductMktStatus=1&goto=Search.DrugDetails(accessed June 2014).

FDA 2012

U.S. Food, Drug Administration. Aubagio (Teriflunomide) Product Approval Information 2012. http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Set_Current_Drug&ApplNo=202992&DrugName=AUBAGIO&ActiveIngred=TERIFLUNOMIDE&SponsorApplicant=SANOFI%20AVENTIS%20US&ProductMktStatus=1&goto=Search.DrugDetails(accessed June 2014).

FDA 2013

U.S. Food, Drug Administration. Tecfidera (Dimethyl fumarate) Product Approval Information 2013. http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.Set_Current_Drug&ApplNo=204063&DrugName=TECFIDERA&ActiveIngred=DIMETHYL%20FUMARATE&SponsorApplicant=BIOGEN%20IDEC%20INC&ProductMktStatus=1&goto=Search.DrugDetails(accessed June 2014).

FDA 2014a

U.S. Food, Drug Administration. Alemtuzumab (Lemtrada) Product Approval Information. Licensing Action 2014. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2014/103948Orig1s5139ltr.pdf(accessed Sept 2014).

FDA 2014b

U.S. Food, Drug Administration. Peginterferon beta‐1a (Plegridy) Product Approval Information. Licensing Action 2014. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/biologics/2004/125083ltr.pdf(accessed September 2014).

FDA 2014c

U.S. Food, Drug Administration. Imuran Product Information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/016324s037,017391s016lbl.pdf(accessed June 2015).

Filippini 2013

Filippini G, Del Giovane C, Vacchi L, D’Amico R, Di Pietrantonj C, Beecher D, et al. Immunomodulators and immunosuppressants for multiple sclerosis: a network meta‐analysis. Cochrane Database of Systematic Reviews 2013, Issue 6. [DOI: 10.1002/14651858.CD008933]

Fox 2004

Fox E. Mechanism of action of mitoxantrone. Neurology 2004;12:15‐8.

Glenny 2005

Glenny A, Altman D, Song F, Sakarovitch C, Deeks J, D'Amico R, et al. Indirect comparisons of competing interventions. Health Technology Assessment 2005;9:1‐34.

Goodin 2002

Goodin DS, Frohman EM, Garmany GP, Halper J, Likosky WH, Lublin FD, et al. Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and the MS Council for Clinical Practice Guidelines. Disease modifying therapies in multiple sclerosis: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and the MS Council for Clinical Practice Guidelines. Neurology 2002;58(2):169‐78.

Goodman 2009

Goodman A, Rossman H, Bar‐Or A, Miller A, Miller D, Schmierer K, et al. GLANCE: results of a phase 2, randomized, double‐blind, placebo‐controlled study. Neurology 2009;72(9):806–12.

GRADE Working Group 2004

GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328:1490‐4.

GRADEpro 2008 [Computer program]

Brozek J, Oxman A, Schünemann H. GRADEpro. Version 3.2 for Windows. Brozek J, Oxman A, Schünemann H, 2008.

Hadjigeorgiou 2013

Hadjigeorgiou GM, Doxani C, Miligkos M, Ziakas P, Bakalos G, Papadimitriou D, et al. A network meta‐analysis of randomized controlled trials for comparing the effectiveness and safety profile of treatments with marketing authorization for relapsing multiple sclerosis. Journal of Clinical Pharmacy and Therapeutics 2013;38(6):433‐9.

Higgins 2011

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

Higgins 2012

Higgins JPT, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta‐analysis: concepts and models for multi‐arm studies. Research Synthesis Methods 2012;3:98‐110.

Hill‐Cawthorne 2012

Hill‐Cawthorne GA, Button T, Tuohy O, Jones JL, May K, Somerfield J, et al. Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis. Journal of Neurology, Neurosurgery, and Psychiatry 2012;83:298‐304.

Hu 2012

Hu X, Miller L, Richman S, Hitchman S, Glick G, Liu S, et al. A novel PEGylated interferon beta‐1a for multiple sclerosis: safety, pharmacology, and biology. Journal of Clinical Pharmacology 2012;52:798–808.

Inusah 2010

Inusah S, Sormani MP, Cofield SS, Aban IB, Musani SK, Srinivasasainagendra V, et al. Assessing changes in relapse rates in multiple sclerosis. Multiple Sclerosis 2010;16:1414–21.

Jackson 2014

Jackson D, Barrett JK, Rice S, White IR, Higgins JP. A design‐by‐treatment interaction model for network meta‐analysis with random inconsistency effects. Statistics in Medicine 2014;33(21):3639‐54.

Kieseier 2011

Kieseier BC. The mechanism of action of interferon‐β in relapsing multiple sclerosis. CNS Drugs 2011;25:491‐502.

Kremenchutzky 2006

Kremenchutzky M, Rice G, Baskerville J, Wingerchuk D, Ebers G. The natural history of multiple sclerosis: a geographically based study 9: observations on the progressive phase of the disease. Brain 2006;129:584‐94.

Kurtzke 1983

Kurtzke J. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 1983;33:1444‐52.

Lublin 1996

Lublin F, Reingold S. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology 1996;46:907‐11.

Mandala 2002

Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, et al. Alteration of lymphocyte trafficking by sphingosine‐1‐phosphate receptor agonists. Science 2002;296:346‐9.

McDonald 2001

McDonald W, Compston A, Edan G, Goodkin D, Hartung H, Lublin F. Recommended diagnostic criteria for multiple sclerosis: guidelines from the international panel on the diagnosis of multiple sclerosis. Annals of Neurology 2001;50:121‐7.

Miladinovic 2014

Miladinovic B, Hozo I, Chaimani A, Djulbegovic B. Indirect treatment comparison. Stata Journal 2014;14(1):76‐86.

Nicholas 2012

Nicholas R, Straube S, Schmidli H, Pfeiffer S, Friede T. Time‐patterns of annualized relapse rates in randomized placebo‐controlled clinical trials in relapsing multiple sclerosis: a systematic review and meta‐analysis. Multiple Sclerosis 2012;18:1290‐6.

Oh 2013

Oh J, Calabresi PA. Emerging injectable therapies for multiple sclerosis. Lancet Neurology 2013;12:1115–26.

Peters 2008

Peters J, Sutton A, Jones D, Abrams K, Rushton L. Contour‐enhanced meta‐analysis funnel plots help distinguish publication bias from other causes of asymmetry. Journal of Clinical Epidemiology 2008;61(10):991‐6.

Polman 2005

Polman C, Reingold S, Edan G, Filippi M, Hartung H, Kappos L, et al. Diagnostic criteria for multiple sclerosis: 2005 revisions to the 'McDonald Criteria'. Annals of Neurology 2005;58:840‐6.

Polman 2011

Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Annals of Neurology 2011;69:292–302.

Poser 1983

Poser C, Paty D, Scheinberg L, McDonald W, Davis F, Ebers G, et al. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Annals of Neurology 1983;13:227‐31.

Ravnborg 2010

Ravnborg M, Sørensen P, Andersson M, Celius E, Jongen P, Elovaara I, et al. Methylprednisolone in combination with interferon beta‐1a for relapsing‐remitting multiple sclerosis (MECOMBIN study): a multicentre, double blind, randomised, placebo‐controlled, parallel‐group trial. Lancet Neurology 2010;9:672‐80.

Salanti 2011

Salanti G, Ades A, Ioannidis J. Graphical methods and numerical summaries for presenting results from multiple‐treatment meta‐analysis: an overview and tutorial. Journal of Clinical Epidemiology 2011;64:163‐71.

Salanti 2012

Salanti G. Indirect and mixed‐treatment comparison, network, or multiple‐treatments meta‐analysis: many names, many benefits, many concerns for the next generation evidence synthesis tool. Research Synthesis Methods 2012;3(2):80‐97.

Salanti 2014

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.

Scalfari 2014

Scalfari A, Neuhaus A, Daumer M, Muraro PA, Ebers GC. Onset of secondary progressive phase and long‐term evolution of multiple sclerosis. Journal of Neurology, Neurosurgery & Psychiatry 2014;85:67‐75.

Schmied 2003

Schmied M, Duda PW, Krieger JI, Trollmo C, Hafler DA. In vitro evidence that subcutaneous administration of glatiramer acetate induces hyporesponsive T cells in patients with multiple sclerosis. Clinical Immunology 2003;106:163‐74.

Schünemann 2011

Schünemann H, Oxman A, Higgins J, Vist G, Glasziou P, Guyatt G. Chapter 11: Presenting results and ‘Summary of findings’ tables. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Stangel 1999

Stangel M, Toyka K, Gold R. Mechanisms of high‐dose intravenous immunoglobulins in demyelinating diseases. Archives of Neurology 1999;56(6):661‐3.

Steinvorth 2013

Steinvorth SM, Röver C, Schneider S, Nicholas R, Straube S, Friede T. Explaining temporal trends in annualised relapse rates in placebo groups of randomised controlled trials in relapsing multiple sclerosis: systematic review and meta‐regression. Multiple Sclerosis 2013;19:1580‐6.

Stellmann 2012

Stellmann JP, Neuhaus A, Herich L, Schippling S, Roeckel M, Daumer M, et al. Placebo cohorts in phase‐3 MS treatment trials, predictors for on‐trial disease activity 1990‐2010 based on a meta‐analysis and individual case data. PLoS One 2012;7(11):e50347. [DOI: 10.1371/journal.pone.0050347]

Tiede 2003

Tiede I, Fritz G, Strand S, Poppe D, Dvorsky R, Strand D, et al. CD28‐dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. Journal of Clinical Investigation 2003;111:1133‐45. [PUBMED: 12697733]

Varrin‐Doyer 2014

Varrin‐Doyer M, Zamvil SS, Schulze‐Topphoff U. Laquinimod, an up‐and‐coming immunomodulatory agent for treatment of multiple sclerosis. Experimental Neurology 2014;262:66‐71.

Veroniki 2013

Veroniki AA, Vasiliadis HS, Higgins JP, Salanti G. Evaluation of inconsistency in networks of interventions. Internal Journal Epidemiology 2013;42(1):332‐45.

Weiner 1993

Weiner H, Mackin G, Matsui M, Orav E, Khoury S, Dawson D, et al. Double‐blind pilot trial of oral tolerization with myelin antigens in multiple sclerosis. Science 1993;259(5099):1321–4.

White 2011

White IR. Multivariate random‐effects meta‐regression: updates to mvmeta. Stata Journal 2011;11:255‐70.

White 2012

White IR, Barrett JK, Jackson D, Higgins JPT. Consistency and inconsistency in network meta‐analysis: model estimation using multivariate meta‐regression. Research Synthesis Methods 2012;3(2):111‐25.

Wilms 2010

Wilms H, Sievers J, Rickert U, Rostami‐Yazdi M, Mrowietz U, Lucius R. Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL‐1beta, TNF‐alpha and IL‐6 in an in‐vitro model of brain inflammation. Journal of Neuroinflammation 2010;7:30.

Wuest 2011

Wuest SC, Edwan JH, Martin JF, Han S, Perry JS, Cartagena CM, et al. A role for interleukin‐2 trans‐presentation in dendritic cell‐mediated T cell activation in humans, as revealed by daclizumab therapy. Nature Medicine 2011;17:604–9.

Yednock 1992

Yednock TA, Cannon C, Fritz LC, Sanchez‐Madrid F, Steinman L, Karin N. Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature 1992;356:63‐6.

Yousry 2006

Yousry T, Major E, Ryschkewitsch C, Fahle G, Fischer S, Hou J, et al. Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy. New England Journal of Medicine 2006;354(9):924‐33. [PUBMED: 16510746]

Zintzaras 2012

Zintzaras E, Doxani C, Mprotsis T, Schmid CH, Hadjigeorgiou G. Network analysis of randomized controlled trials in multiple sclerosis. Clinical Therapeutics 2012;34(4):857–69.

References to other published versions of this review

Tramacere 2014

Tramacere I, Del Giovane C, Salanti G, D'Amico R, Pacchetti I, Filippini G. Immunomodulators and immunosuppressants for relapsing‐remitting multiple sclerosis: a network meta‐analysis. Cochrane Database of Systematic Reviews 2014, Issue 11. [DOI: 10.1002/14651858.CD011381]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Achiron 1998

Methods

RCT

Participants

Age: 19 to 60 years; definite RRMS; mean disease duration 4 years; mean EDSS 3.0; prior use of DMT not reported

Interventions

Loading dose of immunoglobulins 0.4 g/kg body weight intravenously daily for 5 consecutive days followed by additional booster doses of immunoglobulins 0.4 g/kg body weight intravenously daily every 2 months for 24 months (n = 20)

Placebo consisting of 0.9% saline administered with the same schedule as the active treatment (n = 20)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Miles Inc. Cutter Biological, Bayer and Promedico

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were assigned to receive immunoglobulin or placebo by a block‐stratified randomisation procedure, designed to ensure groups balanced for YER, age, and disease duration" (Page 399)

Allocation concealment (selection bias)

Unclear risk

"Randomization was performed at the pharmacy, and the bottles of immunoglobulin or placebo were wrapped in sealed opaque bags and brought to the patients' rooms. The entire IV set was covered by an opaque plastic bag to ensure that any possible fluid turbidity or frothing would not be evident to the investigators or patients" (Page 399)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All patients and evaluators were blinded to treatment" (Page 399)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"A relapse was confirmed only when the patient's symptoms were accompanied by objective changes on neurologic examination by the treating neurologist who was blind to the patient's treatment", and "Upon entry, and monthly thereafter, every patient underwent a neurologic examination by two examining neurologists, and an independent EDSS score was recorded by each" (Page 399)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 5.0% were lost to follow‐up (5.0% in immunoglobulins and 5.0% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Triton Biosciences and the role of the study sponsor was unclear

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of patients who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 3 months outcomes were reported incompletely, and disability worsening confirmed at 6 months was not assessed

ADVANCE 2014

Methods

RCT

Participants

Age: 18 to 65 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.5; prior use of any MS medication at any time prior to the start of study: 17%

Interventions

Peg‐interferon beta‐1a 125 μg subcutaneously once every 2 weeks for 12 months (n = 512)

Peg‐interferon beta‐1a 125 μg subcutaneously once every 4 weeks for 12 months (n = 500)

Placebo subcutaneously once every 2 weeks for 12 months (n = 500)

Outcomes

Relapse at 12 months

Notes

Funding: Biogen Idec

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned (1:1:1) to receive subcutaneous injections with pre‐filled syringes of placebo, peginterferon beta‐1a at a dose of 125 μg once every 2 weeks, or peginterferon beta‐1a 125 μg once every 4 weeks, stratified by site" (Page 658)

Allocation concealment (selection bias)

Low risk

"Randomisation was done by a centralised interactive voice response and web system. Placebo was a matched diluent, given with a matched pre‐filled syringe. Patients received either study drug or placebo every 2 weeks to maintain masking; those assigned to receive study drug every 4 weeks received alternate injections of placebo and peginterferon beta‐1a every 2 weeks" (Page 658)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All study management and site personnel, investigators, and patients were masked to treatment assignment" (Page 658)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Each site had separate examining and treating neurologists, thereby maintaining rater masking for all treatment groups" and "relapse was confirmed by the independent neurological evaluation committee" (Page 658)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 11.9% were lost‐to follow‐up (14.5% in peg‐interferon beta‐1a 125 μg every 2 weeks, 12.4% in peg‐interferon beta‐1a 125 μg every 4 weeks, and 8.8% in placebo), with some indication of the differences in reasons: adverse events of 4.8% in peg‐interferon beta‐1a 125 μg every 2 weeks, 4.7% in peg‐interferon beta‐1a 125 μg every 4 weeks, and 1.0% in placebo

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Biogen Idec, "Biogen Idec collected, analysed, and contributed to the interpretation of the data" (Page 659), and 5 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of patients who discontinued the study and in time to discontinuation, which they did not report

AFFIRM 2006

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; median disease duration 5 years (range, 0 to 34 years); mean EDSS 2.3; prior use of DMT not reported

Interventions

Natalizumab 300 mg by intravenous infusion once every 4 weeks for up to 116 weeks (n = 627)
Placebo (unspecified) (n = 315)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Biogen Idec, Inc. and Elan Pharmaceutica

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned to treatment that was stratified according to study site in blocks of three (two active, one placebo) with the use of a computer‐generated block randomization schedule" (Page 900)

Allocation concealment (selection bias)

Low risk

"A multidigit identification number, implemented by an interactive voice‐response system was used" (Page 900)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All study personnel, patients, sponsor personnel involved in the conduct of the study, and the investigator advisory committee were unaware of treatment assignments throughout the study", and "Treating neurologists were responsible for all aspects of patient care, including the management of adverse events and the treatment of relapsing disease" (Pages 900‐1)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Examining neurologists performed objective evaluation with use of the EDSS and neurologic examination during all study visits; they were not in contact with patients in any other capacity, so as to reduce the possibility of being unblinded by side effects or laboratory assessments", "Patients visited the clinic every 12 weeks for scoring on the EDSS", and "If a relapse was suspected, the patient was referred to the examining neurologist, who evaluated the patient within five days after the event" (Page 901)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 9.1% were lost‐to follow‐up (8.3% in natalizumab and 10.8% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Biogen Idec and Elan Pharmaceuticals, "Data were analyzed by Biogen Idec and Elan Pharmaceuticals" (Page 909) and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 3 months outcomes were reported incompletely

ALLEGRO 2012

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 9 years; mean EDSS 2.6; prior use of DMT at any time prior to the start of study: 39.0% (38.2% in laquinimod and 39.7% in placebo)

Interventions

Laquinimod 0.6 mg oral capsule once daily for 24 months (n = 550)

Placebo oral capsule once daily for 24 months (n = 556)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Teva Pharmaceutical Industries

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomization list, stratified according to study center, was computer‐generated" (Page 1002)

Allocation concealment (selection bias)

Low risk

"The subject was allocated a screening number by the investigator using an Interactive Voice Response System (IVRS)" (Page 44 of Protocol)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Patients and investigators were unaware of the study assignments" (Page 1002)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Neurologic assessments and general medical evaluations were conducted by two neurologists in order to minimize the possibility of unblinding: an examining neurologist assessed neurologic condition, and the treating neurologist determined whether a patient had a relapse", and "the treating neurologist was unaware of the study‐group assignment" (Page 1002)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 21.9% were lost‐to follow‐up (20.5% in laquinimod and 23.2% in placebo), with some indication of the differences in reasons: adverse event(s) in 7.6% in laquinimod and 5.0% in placebo

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "The sponsor designed and monitored the study" and "The data were collected and analyzed by the sponsor" (Page 1001)

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 6 months outcomes were reported incompletely, and no additional data were provided on request

BECOME 2009

Methods

RCT

Participants

Age: 18 to 55; definite RRMS or CIS; median time since MS onset 1 year; mean EDSS 2.0; all participants (except 1) were previously untreated patients

Interventions

Interferon beta‐1b (Betaseron) 250 μg subcutaneously every other day for 24 months (n = 36)

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 39)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Bayer Schering Pharma

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization was stratified by clinical site (Newark or Teaneck) and the presence of enhancement on screening MRI" (Page 1977)

Allocation concealment (selection bias)

Unclear risk

Nothing was said about allocation concealment

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients could not be blinded because of the characteristic injection reactions to IFN‐1b or GA" (Page 1977)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Subjective relapses that were confirmed by a blinded examining neurologist using worsening scores on either the Scripps Neurological Rating Scale (SNRS) or the Expanded Disability Status Scale (EDSS) were considered objective relapses" (Page 1977). However, it is not clear how and when the examining neurologist evaluated subjective relapses and EDSS scores

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 14.7% were lost‐to follow‐up (19.4% in interferon beta‐1b and 10.3% in glatiramer acetate), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "The BECOME study was supported by Bayer Schering Pharma, distributors of IFN‐1b, but was investigator‐initiated and remains the intellectual property of New Jersey Medical School/University of Medicine & Dentistry of New Jersey. The sponsor of the study was allowed to comment on data interpretation and had the opportunity to review and comment on the final manuscript prior to submission. The sponsor was not allowed to participate in any of the following phases of the study: conduct of the study, data collection, data management, data analysis, and preparation of the manuscript" (Page 1981)

‐ Relapse outcome was reported incompletely, and no additional data were provided on request

BEYOND 2009

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 5 years; mean EDSS 2.3; prior use of DMT not reported

Interventions

Interferon beta‐1b (Betaseron) 250 µg subcutaneous every other day for 24 months (n = 897)

Interferon beta‐1b (Betaseron) 500 µg subcutaneous every other day for 24 months (n = 899)

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 448)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Bayer HealthCare Pharmaceuticals

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Use of SAS‐based block randomisation with regional stratification" (Page 890)

Allocation concealment (selection bias)

Unclear risk

"Patients were randomly assigned in a 2:2:1 ratio ... by the central randomisation group..." (Page 890)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Physicians and patients were double‐blind to comparisons between the two doses of IFNß‐1b... Ibuprofen or acetaminophen were given at the same time as random assignment to IFNß‐1b, at least during the first 3 months, to reduce flu‐like symptoms. The treating physicians and the patients were therefore aware of treatment assignments" (Page 891)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The masked evaluating physicians did all neurological assessments and ascertained functional system and EDSS scores...The evaluating physicians were not involved in the care of patients and had no access to patient files or previous assessments", and "Patients covered their injection sites during neurological examination and did not discuss any adverse events with the evaluating physician" (Pages 891‐2)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 16.0% were lost‐to follow‐up (19.2% in interferon beta‐1b 500 µg, 12.6% in interferon beta‐1b 250 µg, and 16.5% in glatiramer acetate), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

Relapse and disability worsening outcomes were reported incompletely

Bornstein 1987

Methods

RCT

Participants

Age: 20 to 35 years; definite RRMS; mean disease duration 6 years; mean EDSS 3.1; prior use of DMT not reported

Interventions

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 25)

Placebo bacteriostatic saline subcutaneous daily for 24 months (n = 25)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: grants from the NINCDS and the NIH, Bethesda, Md

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"The random assignment of the first patient of a pair determined the assignment of both" (Page 409)

Allocation concealment (selection bias)

High risk

An open allocation schedule was used: "Treatment assignments were made known to the clinical assistant responsible for the production, labelling and distribution of medication" (Page 409)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"The patient's self evaluation of ... side effects were reported to the clinical assistant, who was not blinded to the treatment" (Page 409)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Patients visited the clinic every three months for two years. At each visit, a neurologist unaware of the patient's treatment group completed a neurologic examination and status evaluation" and "Patients were also seen at the time of suspected exacerbations ... the neurologist verified exacerbations on the basis of study criteria" (Page 409)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 4.0% were lost‐to follow‐up (0% in glatiramer acetate and 8.0% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Low risk

The study appears to be free of other sources of bias

BRAVO 2014

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; median disease duration 5 years; median EDSS 2.5; prior use of DMT at any time prior to the start of study: 7.4% (6.9% in laquinimod, 9.4% in interferon beta‐1a and 6.0% in placebo)

Interventions

Laquinimod 0.6 mg oral capsule once daily for 24 months (n = 434)

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 24 months (n = 447)

Placebo oral capsule once daily for 24 months (n = 450)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Teva Pharmaceutical Industries

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The computer‐generated randomization scheme prepared by the Teva Global Biostatistics Unit" (Page 775)

Allocation concealment (selection bias)

Unclear risk

"1:1:1 treatment assignment ratio stratified by study center, to laquinimod 0.6 mg capsule once‐daily, matching oral placebo, or IFNß‐1a IM 30 µg once‐weekly injection" (Page 775)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients and treating neurologists were blinded to oral treatment assignment (laquinimod or placebo), but not to IFNb‐1a IM assignment", and "All patients, including those receiving oral treatment, wore clothing and/or a robe that ensured coverage of all potential IM injection sites during examination and were instructed not to discuss adverse events (AEs), routes of administration, or treatment assignments with the examining neurologist" (Page 775)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The examining neurologist was blinded to all treatments", and "The examining neurologist performed an EDSS assessment for relapse confirmation within 7 days of symptom onset" (Page 775)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 18.1% were lost‐to follow‐up (18.7% in laquinimod, 15.4% in interferon beta‐1a, and 20.2% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "N. Sasson of Teva Pharmaceutical Industries provided statistical support for the manuscript" (Page 773), and 2 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

CAMMS223 2008

Methods

RCT

Participants

Age: 18 to 50; definite RRMS; median time since first relapse 1 year; mean EDSS 1.9; all participants were previously untreated patients

Interventions

Alemtuzumab 24 mg per day intravenously on 5 consecutive days during the first month and on 3 consecutive days at months 12 and 24 (n = 110)

Alemtuzumab 12 mg per day intravenously on 5 consecutive days during the first month and on 3 consecutive days at months 12 and 24 (n = 113)

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 36 months (n = 111)

All participants received 1 g of intravenous methylprednisolone for 3 days at baseline and at months 12 and 24

Outcomes

Relapse at 12, 24, and 36 months. Disability worsening at 24 and 36 months

Notes

Funding: Genzyme (a Sanofi company) and Bayer Schering Pharma

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Eligible patients were randomly assigned in a 1:1:1 ratio to receive alemtuzumab (at a dose of either 12 mg per day or 24 mg per day) or interferon beta‐1a with the use of the Pocock and Simon minimization algorithm to balance the study groups with regard to age (<30 years or ≥30 years), sex, and baseline EDSS score (<2.0 or ≥2.0)" (Page 1787)

Allocation concealment (selection bias)

Low risk

"Patients were allocated via an interactive voice response system (IVRS)" (Information provided on request by Genzyme)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients wore clothing that covered injection sites", and "Safety was assessed quarterly by the treating neurologist, who was aware of study‐group assignment" (Page 1787)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"EDSS scores were determined quarterly in a blinded fashion by a neurologist who also adjudicated possible relapses. Patients wore clothing that covered injection sites" (Page 1787). It is not clear how potential relapses were assessed

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 25.1% were lost to follow‐up (16.4% in alemtuzumab 24 mg, 18.6% in alemtuzumab 12 mg, and 40.5% in interferon beta‐1a), with some indication of the differences in reasons: adverse events of 0.01% in alemtuzumab 24 mg, 1.8% in alemtuzumab 12 mg, and 11.7% in interferon beta‐1a; and lack of benefit of 1.8% in alemtuzumab 24 mg, 1.8% in alemtuzumab 12 mg, and 14.4% in interferon beta‐1a

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes. Missing data not reported in the published paper were provided on request by Genzyme

Other bias

High risk

"Genzyme employees analyzed the data" (Page 1789), and 5 co‐authors of the published paper were affiliated to the pharmaceutical company

CARE‐MS I 2012

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; mean disease duration 2 years; mean EDSS 2.0; all participants were previously untreated patients

Interventions

Alemtuzumab 12 mg per day intravenously on 5 consecutive days at month 0 and 3 consecutive days at month 12 (n = 386)

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 24 months (n = 195)

Participants in both groups received 1 g per day of intravenous methylprednisolone on 3 consecutive days at baseline and at month 12. After a protocol amendment in January 2009, alemtuzumab patients received oral aciclovir 200 mg twice daily during alemtuzumab infusion and for 28 days thereafter as prophylaxis against herpes infection

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Genzyme (a Sanofi company)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"We randomly allocated patients in a 2:1 ratio" and "Randomisation was stratified by site" (Page 1820)

Allocation concealment (selection bias)

Low risk

"We randomly allocated patients using an interactive voice response system" (Page 1820)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Because both study drugs have adverse effects that precluded masking of patients and treating clinicians to treatment assignment, and because subcutaneous interferon beta 1a was available only in proprietary prefilled syringes that could not effectively be duplicated for placebo..." (Page 1820)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"We secured clinical data integrity by stringent clinical and MRI rater masking, and adjudication of relapses by a committee comprising six independent and masked neurologists. In the absence of a masked rater, unmasked raters could submit EDSS assessments" (Page 1820). Moreover, it is not clear how and when the committee evaluated potential relapses

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 7.1% were lost to follow‐up (4.9% in alemtuzumab 12 mg and 11.3% in interferon beta‐1a), with some indication of the differences in reasons: adverse events of 2.6% in alemtuzumab and 0% in interferon beta‐1a

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

"The study sponsor (Genzyme) was involved in the design and undertaking of the trial, data analysis and interpretation, writing of the manuscript, and the decision to submit the manuscript for publication. Bayer Schering Pharma participated in the design and oversight of the trial", "The sponsor did the statistical analyses" (Page 1822), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

CARE‐MS II 2012

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 5 years; mean EDSS 2.7; all patients were previously treated: "at least one relapse while on interferon beta or glatiramer after at least 6 months of treatment"

Interventions

Alemtuzumab 24 mg per day intravenously on 5 consecutive days at month 0 and 3 consecutive days at month 12 (n = 170; data presented for safety assessment only)

Alemtuzumab 12 mg per day intravenously on 5 consecutive days at month 0 and 3 consecutive days at month 12 (n = 436)

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 24 months (n = 231)

Participants in both groups received 1 g per day of intravenous methylprednisolone on 3 consecutive days at baseline and at month 12. After a protocol amendment in December 2008, alemtuzumab patients received oral aciclovir 200 mg twice daily during alemtuzumab infusion and for 28 days thereafter as prophylaxis against herpes infection

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Genzyme (a Sanofi company)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"2:1 randomisation allocation stratified by site" (Pages 1830‐1)

Allocation concealment (selection bias)

Low risk

"We randomly allocated patients with an interactive voice response system" (Page 1830)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Because both study drugs had adverse effects that precluded double‐blinding, and interferon beta 1a proprietary syringes could not effectively be duplicated for placebo..." (Page 1831)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Clinical data integrity was secured by stringent rater‐masking and independent adjudication of relapses. Raters, who were masked to treatment‐group assignment, did the EDSS assessments every 3 months and when a relapse was suspected" and "In the absence of a masked rater, unmasked raters could submit EDSS assessments" (Page 1831). Moreover, it is not clear how and when the raters evaluated potential relapses

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 11.4% were lost to follow‐up (4.6% in alemtuzumab 12 mg and 24.2% in interferon beta‐1a), with some indication of the differences in reasons: lack of benefit of 0% in alemtuzumab 12 mg and 2.6% in interferon beta‐1a

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "Genzyme (Sanofi) was involved in the design and undertaking of the trial, data analysis and interpretation, writing of the manuscript, and the decision to submit the manuscript for publication" (Page 1833), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ Sample size reported in the article was not that estimated in the protocol but calculated after an amendment in December 2008

CombiRx 2013

Methods

RCT

Participants

Age: 18 to 60 years; definite RRMS; mean disease duration 1 year; mean EDSS 2.0; all participants were previously untreated patients

Interventions

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week with matched placebo preparation for 36 months (n = 250)

Glatiramer acetate 20 mg subcutaneous daily with matched placebo preparation for 36 months (n = 259)

Outcomes

Relapse at 36 months. Disability worsening at 36 months

Notes

Funding: National Institutes of Health (NIH)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Participants were randomized via a computerized data entry system using a permuted block design within sites with block sizes of 6 and 12" (Page 328)

Allocation concealment (selection bias)

Low risk

"Participants were randomized via a computerized data entry system that masked treatment arm allocation" (Page 328)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Participants were randomized via a computerized data entry system that masked drug dispensing to participants and all site personnel for the entire duration of the trial period" (Page 328)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Treating clinician and an examining clinician were both blinded to treatment assignment", "confirmed progression was assessed by the blinded EDSS examiner and confirmed centrally", and "The designation of the type of relapse was determined centrally according to data entered onto a relapse assessment form and the change in EDSS" (Page 328‐329). The blinding of central commission was not reported

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 18.1% were lost to follow‐up (22.4% in interferon beta‐1a and 13.9% in glatiramer acetate; P value for proportion terminating early = 0.029), with some indication of the differences in reasons: adverse event(s) of 7.2% in interferon beta‐1a and 4.6% in glatiramer acetate

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of patients who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 6 months outcomes were reported incompletely, and no additional data were provided on request

Comi 2001

Methods

RCT

Participants

Age 18 to 50 years; definite RRMS; mean disease duration 8 years; mean EDSS 2.4; prior use of DMT not reported

Interventions

Glatiramer acetate 20 mg subcutaneous daily for 9 months (n = 119)

Placebo (not described) (n = 120)

Outcomes

Relapse at 9 months

Notes

Funding: Teva Pharmaceutical

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomization list, stratified by centers, was computer‐generated by the TEVA Statistical Data Management Department. Equal allocation of the two treatment groups was used" (Page 291)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"A treating neurologist was responsible for the overall medical management of the patient including safety monitoring ... All personnel were unaware of treatment allocation... both the treating neurologist and the patient were informed on the importance of not discussing safety issue with the examining neurologist" (Page 291)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"An examining neurologist was responsible for all scheduled neurological examinations and exacerbation follow‐up" (Page 291)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 5.9% were lost to follow‐up (5.9% in glatiramer acetate and 5.8% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Teva Pharmaceutical, and "Drs Stark, Gurevich, Kadosh, Zak, Pinchassi, and Ladkani are employees of Teva Pharmaceutical, Ltd., involved in trial design and execution, study management, database management, and statistical analysis" (Page 296)

‐ Relapse outcome was reported incompletely

CONFIRM 2012

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration (time since diagnosis) 5 years; mean EDSS 2.6; prior use of any MS medication at any time prior to the start of study: 40% to 41% across study groups

Interventions

Dimethyl fumarate 240 mg oral capsule 3 times daily for 24 months (n = 345)

Dimethyl fumarate 240 mg oral capsule 2 times daily for 24 months (n = 362)

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 350)

Placebo oral capsule 3 times daily for 24 months (n = 363)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Biogen Idec

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned in a 1:1:1:1 ratio to receive oral placebo, BG‐12 at a dose of 240 mg two times daily, BG‐12 at a dose of 240 mg three times daily, or subcutaneous daily injections of 20 mg of glatiramer acetate for 96 weeks" (Page 1088); and "The randomization was stratified by site" (Page 33 of Protocol)

Allocation concealment (selection bias)

Low risk

"Randomization took place across all study sites using a centralized Interactive Voice Response System (IVRS)" (Page 33 of Protocol)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients receiving glatiramer acetate were aware of their treatment assignment. All study management and site personnel, investigators, and patients were unaware of assignment to the BG‐12 and placebo groups", and "To ensure that the assignments to the BG‐12 and placebo groups would not be revealed, patients in those groups were instructed not to take the study medication within 4 hours before each study visit, since a flushing reaction is known to be more common with BG‐12" (Page 1088). Since flushing is a known side effect of dimethyl fumarate, patients were possibly not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"An independent neurologic evaluation committee, whose members were unaware of the study‐group assignments, provided confirmation of relapses of multiple sclerosis" and "examining neurologists and members of the independent neurologic evaluation committee were unaware of all study‐group assignments" (Page 1088)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 20.3% were lost to follow‐up (20.8% in dimethyl fumarate 240 mg 3 times daily, 20.7% in dimethyl fumarate 240 mg 2 times daily, 16.1% in glatiramer acetate, and 23.4% in placebo), with some indication of the differences in reasons: adverse events of 8.1% in dimethyl fumarate 240 mg 3 times daily, 6.1% in dimethyl fumarate 240 mg 2 times daily, 3.6% in glatiramer acetate, and 3.3% in placebo

Selective reporting (reporting bias)

High risk

The published report included all pre‐specified primary benefit outcomes. However, disability confirmed at 6 months was not reported in the published report, it was reported by the FDA in terms of survival probabilities

Other bias

High risk

‐ The study was sponsored by Biogen Idec, "data were analyzed by the sponsor" (Page 1088), and 6 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

DEFINE 2012

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration (time since diagnosis) 6 years; mean EDSS 2.4; prior use of DMT at any time prior to the start of study: 40.7% (40.4% in dimethyl fumarate 240 mg 3 times daily, 39.5% in dimethyl fumarate 240 mg 2 times daily, and 42.2% in placebo)

Interventions

Dimethyl fumarate 240 mg oral capsule 3 times daily for 24 months (n = 416)

Dimethyl fumarate 240 mg oral capsule 2 times daily for 24 months (n = 411)

Placebo oral capsule 3 times daily for 24 months (n = 410)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Biogen Idec

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned, in a 1:1:1 ratio, to receive BG‐12 at a dose of 240 mg twice daily, BG‐12 at a dose of 240 mg three times daily, or placebo. Randomization was performed centrally and was stratified according to site" (Page 1100)

Allocation concealment (selection bias)

Low risk

"Randomization was performed centrally" (Page 1100), and "Randomization took place across all study sites using a centralized Interactive Voice Response System (IVRS)" (Page 33 of Protocol)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Double‐blind", and "To ensure that the study‐group assignments would not be revealed, patients were instructed to take the assigned study drug at least 4 hours before study visits, in case patients in the BG‐12 groups had a side effect of flushing" (Page 1100). Since flushing is a known side effect of dimethyl fumarate, patients were possibly not blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"To maintain concealment of the study‐group assignments, each study center used separate examining and treating neurologists (all of whom remained unaware of the assignments throughout the trial). The examining neurologists conducted neurologic assessments, including assessment of the EDSS score, whereas the treating neurologists were responsible for all aspects of patient care, including the treatment of relapses and other disease symptoms" and "relapses were evaluated by an independent neurologic evaluation committee, whose members reviewed a standardized set of blinded clinical records (which did not include MRI data) from the treating and examining neurologists" (Page 1100)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 23.0% were lost to follow‐up (23.1% in dimethyl fumarate 240 mg 3 times daily, 23.4% in dimethyl fumarate 240 mg 2 times daily, and 22.7% in placebo), with some indication of the differences in reasons: adverse events of 8.7% in dimethyl fumarate 240 mg 3 times daily, 9.8% in dimethyl fumarate 240 mg 2 times daily, and 5.4% in placebo

Selective reporting (reporting bias)

High risk

The published report included all pre‐specified primary benefit outcomes. However, disability confirmed at 6 months was not reported in the published report, it was reported by the FDA in terms of survival probabilities

Other bias

High risk

The study was sponsored by Biogen Idec, "data were analyzed by the sponsor" (Page 1099), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

Etemadifar 2007

Methods

RCT

Participants

Age: 13 to 50 years; definite RRMS; mean disease duration not reported ("short duration"); mean EDSS 1.5; all participants were previously untreated patients

Interventions

Azathioprine 3 mg/kg body weight oral daily for 12 months (n = 47)

Interferons beta (Betaseron, Avonex, or Rebif) for 12 months (n = 47: 15 Betaseron 250 μg subcutaneously every other day, 19 Avonex 30 µg intramuscular once a week, 13 Rebif 44 µg subcutaneous 3 times a week)

Outcomes

Relapse at 12 months

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomized according to a preexisting list produced by a computer program that differed from a random number generator only in that it assigned equal numbers of patients into each treatment group" (Page 1724)

Allocation concealment (selection bias)

Unclear risk

"The first treatment group received IFNβ products regimen. The second group received AZA" (Page 1724)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"The trial was single blinded in that patients were aware but physicians who assessed the outcome were unaware of treatment type that the patient was receiving" (Page 1724)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The trial was single blinded in that patients were aware but physicians who assessed the outcome were unaware of treatment type that the patient was receiving", and "Two neurologists (ME and VS) who do not know which patients had received which treatment clinically evaluated all patients" (Page 1724‐5)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 6.4% were lost to follow‐up (6.4% in azathioprine and 6.4% in interferon beta), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Low risk

The study appears to be free of other sources of bias

EVIDENCE 2007

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.3; prior use of DMT not reported

Interventions

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 12 months (n = 339)

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 12 months (n = 338)

Outcomes

Relapse at 12 months

Notes

Funding: Serono Inc.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Computer‐generated scheme with block size of 6 followed by block size of 4" (Page 2033)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients and a treating physician who was not involved in end point assessment were aware of treatment assignments" (Page 2033)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Evaluating physicians who were blinded to the patients' treatment and symptoms performed all clinical exams" (Page 2033)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 10.6% were lost to follow‐up (11.8% in Rebif and 9.5% in Avonex), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Serono Inc., "The sponsor designed and implemented the study and managed the data" (Page 2047)

‐ Relapse outcome was reported incompletely

Fazekas 1997

Methods

RCT

Participants

Age: 15 to 64 years; definite RRMS; mean disease duration 7 years; mean EDSS 3.3; prior use of DMT not reported

Interventions

Immunoglobulins 0.15 to 0.20 g/kg body weight intravenously monthly for 24 months (n = 75)

Placebo intravenously monthly for 24 months (n = 75)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Sero‐Merieux (Vienna, Austria)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Centralised computer‐generated randomisation schedule with stratification by centre, age, sex, and deterioration rate" (Page 590)

Allocation concealment (selection bias)

Low risk

"Randomly and centrally allocated" and "Infusions of IVIg and placebo were identical in appearance and were stored in plastic bags for concealment during administration" (Page 590)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"At each monthly visit a neurologist who was aware of treatment allocation (treating physician) administered the study medication and asked the patient about any side‐effects" (Page 590)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Patients were assessed on the first day of treatment, every 6 months, and at the end of the 2‐year study by a different neurologist (assessing physician) who was unaware of treatment allocation", and "All patients were told to contact their centre as soon as there was any change in their condition. In such cases, the assessing physician examined the patient to confirm a possible relapse and to assess the severity of the disability" (Page 590)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 1.3% were lost to follow‐up (0% in immunoglobulins and 2.7% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Unclear risk

‐ The study was sponsored by Triton Biosciences and the role of the study sponsor was unclear

‐ Definition of sustained disability worsening was not clearly reported

Fazekas 2008

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 3 years; mean EDSS 2.0; prior use of DMT not reported

Interventions

Immunoglobulins 0.2 g/kg body weight intravenously monthly for 12 months (n = 45)
Immunoglobulins 0.4 g/kg body weight intravenously monthly for 12 months (n = 42)
Placebo intravenously monthly for 12 months (n = 41)

Outcomes

Relapse at 12 months

Notes

Funding: Bayer HealthCare AG

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The random code number was computer generated by the Statistics and Data System Department of Bayer" (Page 266)

Allocation concealment (selection bias)

Unclear risk

"Randomisation performed by an unblinded pharmacist who assigned code numbers from sealed envelopes in a sequential manner" (Page 266)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Considerable effort was made to achieve optimal blinding, including the provision that all patients received a total volume of 4 mL/kg body weight per infusion, which was adjusted by the addition of dextrose 5%" (Page 266)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Endpoints "assessed by an evaluating physician who was otherwise not involved in patient care" (Page 266)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 12.5% were lost to follow‐up (9.5% in immunoglobulins 0.4 g/kg, 17.8% in immunoglobulins 0.2 g/kg, and 9.8% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Bayer HealthCare AG and the role of the study sponsor was unclear. 3 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ Relapse outcome was reported incompletely

FREEDOMS 2010

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 8 years; mean EDSS 2.4; prior use of DMT at any time prior to the start of study: 40.9% (39.6% in fingolimod 1.25 mg, 42.6% in fingolimod 0.5 mg, and 40.4% in placebo)

Interventions

Fingolimod 1.25 mg oral capsule once daily for 24 months (n = 429)

Fingolimod 0.5 mg oral capsule once daily for 24 months (n = 425)

Placebo oral capsule once daily for 24 months (n = 418)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Novartis Pharma

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomly assigned, in a 1:1:1 ratio, to receive oral fingolimod capsules in a dose of 0.5 mg or 1.25 mg or matching placebo ... Randomization was performed ... with the use of stratification according to site, with a block size of six within each site" (Page 388)

Allocation concealment (selection bias)

Unclear risk

"Randomization was performed centrally, with the use of a validated system " (Page 388)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Double blind" (Page 388)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"To ensure that all assessments remained unbiased regarding the study‐group assignments (i.e., unaffected by awareness of them), an independent, specially trained and certified examining neurologist determined all the EDSS scores" (Page 388). "Relapses were verified by the examining neurologist within 7 days after the onset of symptoms" (Page 389)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 18.8% were lost to follow‐up (22.6% in fingolimod 1.25 mg, 13.2% in fingolimod 0.5 mg, and 20.6% in placebo), with some indication of the differences in reasons: unsatisfactory therapeutic effect 3.0% in fingolimod 1.25 mg, 1.4% in fingolimod 0.5 mg, and 6.0% in placebo; and abnormal laboratory values(s) 4.7% in fingolimod 1.25 mg, 2.1% in fingolimod 0.5 mg, and 0.2% in placebo

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Novartis Pharma, "data were analyzed by the sponsor" (Page 388), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

FREEDOMS II 2014

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 11 years; mean EDSS 2.4; prior use of DMT at any time prior to the start of study: 74.8% (77.6% in fingolimod 1.25 mg, 73.7% in fingolimod 0.5 mg, and 73.0% in placebo)

Interventions

Fingolimod 1.25 mg oral capsule once daily for 24 months (n = 370)

Fingolimod 0.5 mg oral capsule once daily for 24 months (n = 358)

Placebo oral capsule once daily for 24 months (n = 355)

"After review of data from the FREEDOMS and TRANSFORMS phase 3 studies, completed on Nov 12, 2009, after consultation with and at the recommendation of the data and safety monitoring board, we decided to stop the 1·25 mg dose. Patients on the high dose were subsequently switched to the 0·5 mg dose in a blinded manner" (Page 546)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Novartis Pharma

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"We randomly allocated patients (1:1:1; stratified by study centre) to receive oral fingolimod capsules in a dose of 0.5 mg or 1.25 mg or matching placebo, once daily for 24 months. The randomisation sequence was generated with an automated system under the supervision of the Novartis Drug Supply Management team" (Page 546)

Allocation concealment (selection bias)

Unclear risk

"To mask treatment allocation, both fingolimod and placebo were dispensed in hard gelatin capsules of identical colour and size and packed in identical bottles" (Page 546)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Patients, investigators, site personnel, independent evaluating physician, first dose administrator and all Novartis personnel were blinded to the study medication assignments from the time of randomisation until the database lock and data analysis for the double‐blind Treatment Phase was completed" (Appendix, Page 2)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The efficacy assessments (ie, confirmation of relapses, scheduled EDSS, ...) were done by an independent, specially trained, and certified assessor not otherwise involved in the treatment of patients)" (Page 546), "Patients were instructed not to discuss adverse events with the independent evaluating physician", "Another physician not otherwise involved in the care of the study patient monitored patients for 6 or more hours after administration of the first dose of the study drug to maintain blind for the known heart rate decrease with fingolimod upon first dose administration", "Clinical assessments were performed at screening and at randomization (baseline), and study visits were scheduled at 2 weeks and 1, 2, 3, 6, 9, 12, 15, 18, 21, and 24 months after randomization", and "In the case of MS relapse EDSS assessment was required at every unscheduled visit to confirm relapse" (Appendix, Page 2)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 28.2% were lost to follow‐up (32.2% in fingolimod 1.25 mg, 24.0% in fingolimod 0.5 mg, and 28.2% in placebo), with some indication of the differences in reasons: unsatisfactory therapeutic effect 2.7% in fingolimod 1.25 mg, 1.7% in fingolimod 0.5 mg, and 4.8% in placebo; and adverse events or abnormal laboratory values(s) 12.7% in fingolimod 1.25 mg, 10.1% in fingolimod 0.5 mg, and 5.1% in placebo

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Novartis Pharma, "The study sponsor participated in the design of the study, conduct of the study, data collection, data management, data analysis and interpretation, and preparation, review, and approval of the paper" (Page 550), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

GALA 2013

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 8 years; mean EDSS 2.8; prior use of DMT at any time prior to the start of study: 13.6% (13.6% in glatiramer acetate and 13.7% in placebo)

Interventions

Glatiramer acetate 40 mg subcutaneous 3 times a week for 12 months (n = 943)

Placebo subcutaneous 3 times a week for 12 months (n = 461)

Outcomes

Relapse at 12 months

Notes

Funding: Teva Pharmaceutical Industries

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Eligible patients were assigned to treatment groups in a 2:1 ratio (GA 40mg tiw or placebo) according to the randomization scheme produced. The randomization scheme used constrained blocks stratified by center" (Page 706)

Allocation concealment (selection bias)

Unclear risk

"Study drugs were packaged and labeled in a way that maintained the masked nature of the study; the appearance, shape, color, and smell were identical" (Page 706)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"The investigators, the sponsor, and any personnel involved in patients’ assessments, monitoring, analysis, and data management were blinded to treatment assignment" (Page 706)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Patients’ general medical assessments were performed separately from the neurological assessments by 2 neurologists or physicians. The examining neurologist/physician was responsible for all neurological assessments" and "All follow‐up neurological examinations were performed by the blinded examining neurologist" (Page 706‐7)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 8.2% were lost to follow‐up (8.9% in glatiramer acetate and 6.7% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "This study was funded by Teva Pharmaceutical Industries, Petah Tikva, Israel. All members of the clinical advisory board, the country principal investigators, the Data Monitoring Committee (DMC), and the MRI Reading Center were reimbursed for their specific services on a contractual basis by Teva Pharmaceutical Industries" (Page 711)

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report.

‐ Relapse outcome was reported incompletely, and no additional data were provided on request

Goodkin 1991

Methods

RCT

Participants

Age: 18 to 65 years; definite RRMS; mean disease duration 6 years; mean EDSS 3.5; prior use of DMT not reported

Interventions

Azathioprine 3.0 mg/kg body weight oral daily for 24 months (n = 30)
Placebo oral daily for 24 months (n = 29)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Wellcome Company

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomised by the statistician using random number tables" (Page 21)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Patients and personnel were blinded, "group PLC received indistinguishable placebo", and "whenever the treating physician made a dose change for an AZA patient, a similar dose change was simultaneously made for a matched placebo patient to preserve the blind" (Pages 20‐1)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Each patient had the same masked examining neurologist and unmasked treating neurologist for the duration of the study. Standardized neurologic examinations were recorded at study entry and at 6 month intervals by the examining neurologist unless the patient reported subjective worsening, in which case an examination was performed as soon as was practical" (Page 21)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 11.9% were lost to follow‐up (10.0% in azathioprine and 13.8% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Unclear risk

Definition of sustained disability worsening not clearly reported

IFNB MS Group 1993

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; mean disease duration (time since diagnosis) 4 years; mean EDSS 2.9; prior use of DMT not reported

Interventions

Interferon beta‐1b (Betaseron) 250 µg subcutaneous every other day for 24 months (n = 124)

Interferon beta‐1b (Betaseron) 50 µg subcutaneous every other day for 24 months (n = 125)

Placebo subcutaneous every other day for 24 months (n = 123)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Triton Biosciences, Inc., Alameda, CA and Berlex Laboratories Inc.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not described

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Each placebo vial contained only similar quantity of albumin and dextrose", "All personnel were blinded to treatment categories", and "One treating neurologist who knew about side effects, reviewed laboratory findings for toxicity, and was responsible for overall care" (Page 656)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"One neurologist who was not aware of drug side effects to do the periodic examinations" (Page 656). However, it is not clear how and when potential relapses and EDSS were assessed

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 9.1% were lost to follow‐up (7.3% in interferon beta‐1b 250 µg, 11.2% in interferon beta‐1b 50 µg, and 8.9% in placebo). Nothing was said about the reasons for study discontinuation

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Triton Biosciences and the role of the study sponsor was unclear

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Disability worsening confirmed at 3 months outcome was reported incompletely, and disability worsening confirmed at 6 months was not assessed

INCOMIN 2002

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; mean disease duration (time since diagnosis) 6 years; mean EDSS 2.0; all participants were previously untreated patients

Interventions

Interferon beta‐1b (Betaseron) 250 µg subcutaneous every other day for 24 months (n = 96)

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 24 months (n = 92)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: The Italian Ministry of Health and the Italian MS Society

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation followed computer‐generated random sequences of digits that were different for each centre and for each sex, to achieve centre and sex stratification" (Page 1454)

Allocation concealment (selection bias)

Low risk

"The codes were randomly assigned to treatments by an independent team of statisticians unaware of the patient’s clinical characteristics" (Page 1454)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"All clinical outcomes were assessed in an open‐label manner" (Page 1454)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"All clinical outcomes were assessed in an open‐label manner" (Page 1454)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 3.2% were lost to follow‐up (2.1% in interferon beta‐1b and 4.3% in interferon beta‐1a). Nothing was said about the reasons for study discontinuation

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Unclear risk

Relapse and disability worsening outcomes were reported incompletely

Johnson 1995

Methods

RCT

Participants

Age: 18 to 45 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.6; prior use of DMT not reported

Interventions

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 125)

Placebo (not described) (n = 126)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: Teva Pharmaceutical

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"A centralized randomization scheme was used" (Page 1270)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Treating neurologists were blinded" (Page 1270)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Examining neurologists were blinded" (Page 1270)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 14.3% were lost to follow‐up (15.2% in glatiramer acetate and 13.5% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 3 months outcomes were reported incompletely, and disability worsening confirmed at 6 months was not assessed

Koch‐Henriksen 2006

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 8 years; mean EDSS 2.9; prior use of DMT not reported

Interventions

Interferon beta‐1b (Betaseron) 250 µg subcutaneous every other day for 24 months (n = 158)

Interferon beta‐1a (Rebif) 22 µg subcutaneous once a week for 24 months (n = 143)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomization algorithm was adjusted to reduce deviations from a 50/50 result in each center" (Page 1057)

Allocation concealment (selection bias)

Low risk

"A central computerized randomization schedule assigned patients to treatment" (Page 1057)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Blinding was abandoned because it could not be maintained owing to the different administration schemes of the two study drugs" (Page 1057)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"Open‐label trial" (Page 1057)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 25.6% were lost to follow‐up (27.8% in interferon beta‐1b and 23.1% in interferon beta‐1a), with some indication of the differences in reasons: "The main cause of withdrawal in the IFN‐1b 250 g arm was side effects (24/158, 15.2%), and treatment failure was the most frequent cause in the IFN‐1a arm (15/143, 10.5%)" (Page 1057)

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ It is unclear if the study was sponsored

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse and disability worsening confirmed at 3 months outcomes were reported incompletely, and disability worsening confirmed at 6 months was not assessed

‐ Rebif at very low dose that is not used in clinical practice

Lewanska 2002

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 9 years; mean EDSS 3.0; prior use of DMT not reported

Interventions

Immunoglobulins 0.2 g/kg body weight intravenously monthly for 12 months (n = 17)
Immunoglobulins 0.4 g/kg body weight intravenously monthly for 12 months (n = 16)
Placebo intravenously monthly for 12 months (n = 18)

Outcomes

Relapse at 12 months

Notes

Funding: Supported by the KBN (State Research Committee)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The generation of allocation sequence was based on random‐number table" (Page 566)

Allocation concealment (selection bias)

Unclear risk

"Infusions of intravenous immunoglobulins and placebo were stored in identical opaque plastic bags for concealment during administration" (Page 566)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Not described

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Evaluating physician was unaware of the actual treatment allocation. Before entry to the study, and monthly thereafter during the study and 3 months after the end of the study, each patient was examined blindly by the same neurologist who was unaware of treatment allocation. Monitoring and recording of relapses, concomitant treatment, side‐effects or other medical events were documented throughout the study" (Page 566)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 3.9% were lost to follow‐up (6.3% in immunoglobulins 0.4 g/kg, 0% in immunoglobulins 0.2 g/kg, and 5.6% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report.

‐ Relapse outcome was reported incompletely

MAIN TRIAL

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 6 years; mean EDSS 1.9; prior use of DMT at any time prior to the start of study: 6.0% (6.5% in azathioprine and 5.5% in interferon beta)

Interventions

Azathioprine 3 mg/kg body weight oral daily for 24 months (n = 77)

Interferons beta (Betaseron, Avonex, or Rebif) for 24 months (n = 73: 5 Betaseron 250 μg subcutaneously every other day, 26 Avonex 30 µg intramuscular once a week, 35 Rebif 22 µg subcutaneous 3 times a week, 7 Rebif 44 µg subcutaneous 3 times a week)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: AIFA (Italian medicines agency)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were selected for AZA or IFNs using a randomization list (1:1 ratio), in blocks of four and stratified by disability score (EDSS≤3.5 or>3.5)"

Allocation concealment (selection bias)

Low risk

"Patients were selected for AZA or IFNs using a computer generated central randomization list"

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Single‐masked"

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"Patients were assessed by an un‐masked treating and a masked examining neurologist at their centers", and "The masked examining neurologist was responsible for the neurological examination and EDSS scoring at scheduled (every six months) and unscheduled visits, requested by the treating neurologist to confirm relapses". Relapse assessment was not blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 15.3% were lost to follow‐up (19.5% in azathioprine and 11.0% in interferon beta), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

Low risk

The study appears to be free of other sources of bias

Millefiorini 1997

Methods

RCT

Participants

Age: 18 to 45 years; definite RRMS; mean disease duration 5 years; mean EDSS 3.6; prior use of DMT not reported

Interventions

Mitoxantrone 8 mg/m² of body surface intravenously monthly for 12 months (total dosage of 96 mg/m² of body surface over 12 months) (n = 27)
Placebo intravenously monthly for 12 months (n = 24)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomized to MTX or placebo using a scheme stratified on age, sex and EDSS which resulted in eight different age/sex/EDSS strata. According to the study protocol, within each stratum the allocation of patients to treatment or placebo was balanced by using a block design of size eight" (Page 154)

Allocation concealment (selection bias)

Low risk

"Central allocation and the intravenous bag and tubing were black to ensure no differences between the treatment groups" (Page 154)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Treating physicians were not blinded. Unclear blinding of patients

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"Monitoring and recording of exacerbations, concomitant therapy or other medical events were documented throughout the study by a treating physician selected in each centre before the beginning of the study. The treating physician was not blinded to study treatment", and "In order to maintain blindness, the interaction of the EDSS physicians with the patient was strictly restricted to the neurological examination. The neurologist was not allowed to talk with the patient about adverse events, or any other issue which could potentially disclose the patient’s treatment" (Page 154)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

None were lost to follow‐up

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ It is unclear if this study was sponsored

‐ Definition of sustained disability worsening was not clearly reported

MSCRG 1996

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.4; all participants were previously untreated patients

Interventions

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 24 months (n = 158)

Placebo intramuscular once a week for 24 months (n = 143)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Biogen, Inc, Cambridge, MA

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Randomisation performed at statistical centre of Buffalo General Hospital, one of the participating centres (biased coin assignment used for sequence generation)" (Page 286)

Allocation concealment (selection bias)

Unclear risk

"schedule sent to each clinical centre, included patients were sequentially assigned the next ID number from the schedule"(Page 286)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"Personnel and participants were blinded to treatment status" (Page 286)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Evaluating physicians were blinded to treatment status" (Page 286)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 42.9% were lost to follow‐up (46.2% in interferon beta‐1a and 39.2% in placebo). The study stopped early for benefit without a formal‐stopping rule

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

The study was sponsored by Biogen and "Personnel of the study sponsor (Biogen) were involved in the conduct and data analysis" (Page 293)

OWIMS 1999

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.6; prior use of DMT not reported

Interventions

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 12 months (n = 98)

Interferon beta‐1a (Rebif) 22 µg subcutaneous 3 times a week for 12 months (n = 95)

Placebo subcutaneous 3 times a week for 12 months (n = 100)

Outcomes

Relapse at 12 months

Notes

Funding: Ares‐Serono International SA, Geneva, Switzerland

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation performed at Corporate Biometrics Department of Ares‐Serono (computer‐generated list)" (Page 680)

Allocation concealment (selection bias)

Low risk

"The randomization code for each patient was delivered to the investigator in sealed envelopes" (Page 680)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"If desired, patients could remain on blinded study medication for another 24 weeks", "Both active treatment and placebo were administered as ready‐to‐use solutions in a volume of 0.5 mL", and "To preserve blinding, patients were instructed to cover injection sites and to refrain from discussing any symptoms that might be in any way related to treatment when visiting the evaluating physician" (Page 681)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The evaluating physician was responsible for neurologic assessments, both at scheduled visits and during exacerbations. Throughout the study, the evaluating physician remained unaware of adverse event profiles and any changes in safety assessments" (Page 681)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 8.2% were lost to follow‐up (13.3% in interferon beta‐1a 44 µg, 8.4% in interferon beta‐1a 22 µg, and 3.0% in placebo), with some indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Ares‐Serono International SA, Geneva, Switzerland

‐ Relapse outcome was reported incompletely

PRISMS 1998

Methods

RCT

Participants

Age: 18 to 50 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.5; prior use of DMT: "Only 3% of patients had received previous immunosuppressive therapy"

Interventions

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 24 months (n = 184)

Interferon beta‐1a (Rebif) 22 µg subcutaneous 3 times a week for 24 months (n = 189)

Placebo subcutaneous 3 times a week for 24 months (n = 187)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Ares‐Serono International SA, Geneva, Switzerland

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation at Corporate Biometrics Department of Ares‐Serono (computer‐generated list, stratified by centre, equal allocation of the treatment groups by a block size of 6)" (Page 1499)

Allocation concealment (selection bias)

Low risk

"The study drug was packed accordingly to the randomisation list and delivered to the centres so that treatment allocation remained concealed" (Page 1499)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

"All personnel involved in the study were unaware of treatment allocation", and "All injection sites were covered up at neurological examinations to ensure that masking was not compromised because of local reactions" (Page 1499)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"All personnel involved in the study were unaware of treatment allocation", "Patients were assessed by two physicians. A “treating” neurologist was responsible for overall medical management of the patient, including treatment of any side‐effects, and an “assessing” neurologist was responsible for neurological assessments and follow‐up of relapses", and "All patients had a neurological assessment every 3 months. Additional assessments were done during relapses" (Page 1499)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 4.8% were lost to follow‐up (2.7% in interferon beta‐1a 44 µg, 6.3% in interferon beta‐1a 22 µg, and 5.3% in placebo), without indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Ares‐Serono International SA, Geneva, Switzerland

‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

‐ Relapse outcome was reported incompletely

REGARD 2008

Methods

RCT

Participants

Age: 18 to 60 years; definite RRMS; mean disease duration 6 years; mean EDSS 2.3; prior use of DMT not reported

Interventions

Interferon beta‐1a (Rebif) 44 µg subcutaneous 3 times a week for 24 months (n = 386)

Glatiramer acetate 20 mg subcutaneous daily for 24 months (n = 378)

Outcomes

Relapse at 24 months. Disability worsening at 24 months

Notes

Funding: EMD Serono and Pfizer

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Computer‐generated randomisation list stratified by centre" (Page 904)

Allocation concealment (selection bias)

Unclear risk

Not described

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Neither the patients nor the treating physicians were blinded to treatment" (Page 904)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The physicians who assessed patients ...were blinded to treatment and communicated with the patients only as needed to complete the EDSS, Kurtzke functional scale (KFS), and relapse assessments. Patients were asked not to discuss their treatment with the assessing physician and they covered their injection sites" (Page 904)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 3.3% were lost to follow‐up (5.2% in interferon beta‐1a and 1.3% in glatiramer acetate). Nothing was said about the reasons for study discontinuation

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ "The study protocol was drafted and developed by the study sponsors, EMD Serono and Pfizer, in conjunction with the investigator steering committee. Data management and analysis were done by the study sponsors" (Page 907), and 2 co‐authors of the published paper were affiliated to the pharmaceutical company

‐ Disability worsening confirmed at 6 months outcome was reported incompletely

SELECT 2013

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; median disease duration (since diagnosis) 3 years; mean EDSS 2.7; prior use of DMT at any time prior to the start of study: 23.7% (22.5% in daclizumab 300 mg, 25.5% in daclizumab 150 mg, and 24.0% in placebo)

Interventions

Daclizumab 300 mg subcutaneously once every 4 weeks for 12 months (n = 209)

Daclizumab 150 mg subcutaneously once every 4 weeks for 12 months (n = 208)

Placebo subcutaneously once every 4 weeks for 12 months (n = 204)

Outcomes

Relapse at 12 months

Notes

Funding: Biogen Idec and AbbVie Biotherapeutics Inc.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Patients were randomly assigned in a 1:1:1 ratio" (Page 2168)

Allocation concealment (selection bias)

Low risk

"Patients were randomly assigned via a centralised interactive voice response system" (Page 2168)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"All personnel and patients were masked to treatment assignment" (Page 2168)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Three members of an independent neurology assessment committee, consisting of multiple sclerosis neurologists who were masked to group assignment, adjudicated whether the protocol definition of relapse was satisfied" (Page 2168)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 7.1% were lost to follow‐up (5.7% in daclizumab 300 mg, 7.7% in daclizumab 150 mg, and 7.8% in placebo). Nothing was said about the reasons for study discontinuation

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Biogen Idec and AbbVie Biotherapeutics Inc, "The sponsor of the study provided assistance in manuscript preparation. The study was designed by the sponsor; the sponsor held and analysed data" (Page 2169), and 5 co‐authors of the published paper were affiliated to the pharmaceutical company
‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

TEMSO 2011

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 9 years; mean EDSS 2.7; prior use of DMT in the previous 2 years: 27.0% (28.4% in teriflunomide 14 mg, 27.9% in teriflunomide 7 mg, and 24.8% in placebo)

Interventions

Teriflunomide 14 mg oral capsule once daily for 25 months (n = 359)

Teriflunomide 7 mg oral capsule once daily for 25 months (n = 366)

Placebo oral capsule once daily for 25 months (n = 363)

Outcomes

Relapse at 12 and 24 months. Disability worsening at 24 months

Notes

Funding: Sanofi‐Aventis

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Eligible patients were randomly assigned (in a 1:1:1 ratio) to receive a once‐daily oral dose of placebo, 7 mg of teriflunomide, or 14 mg of teriflunomide for 108 weeks. Randomization was stratified according to the baseline EDSS score (≤3.5 or >3.5) and according to trial site, with a block size of 6." (Page 1294)

Allocation concealment (selection bias)

Low risk

"The treatment allocation was determined according to the randomization code provided by an interactive voice response system (IVRS)" (Page 74 of Medical Review of FDA)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Double blind" (Page 1294), and at Page 40 of the Protocol they described blinding, packaging and labeling ("Each medication kit was labeled with a two‐part tear‐off label..."). "Unblinding of 40 patients in TEMSO study, and the reasons provided do not appear to justify the need of unblinding" (Page 230 of Statistical Review of FDA)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"A treating neurologist at each site was responsible for evaluating patient eligibility, supervising the administration of study medication, recording and managing adverse events, assessing relapses, and monitoring safety assessments. An independent, specially trained and certified examining neurologist determined all the EDSS scores and performed all assessments of functional systems. Both treating and examining neurologists were unaware of treatment assignments; only the treating neurologist was aware of any side effects that could potentially be related to active therapy" (Pages 1294‐5), "Each episode of relapse was to be confirmed by the treating neurologist (unblinded), based on the objective assessments by an independent examining neurologist (blinded)" (Page 207 of Statistical Review of FDA) and "Patients were required to visit the study site within 7 days after the onset of a suspected relapse, for assessments by the examining neurologist" (Page 1295).

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 20.1% were lost to follow‐up (21.2% in teriflunomide 14 mg, 19.1% in teriflunomide 7 mg, and 20.1% in placebo). Nothing was said about the reasons for study discontinuation. "Some patients discontinued study at the time of blind broken, although it is not clear whether or not the discontinuation was due to unblinding" (Page 208 of Statistical Review of FDA)

Selective reporting (reporting bias)

High risk

The published report included all pre‐specified primary benefit outcomes. However, disability confirmed at 6 months was not reported in the published report, it was reported by the FDA in terms of survival probabilities

Other bias

High risk

‐ The study was sponsored by Sanofi‐Aventis, "data were analyzed by the sponsor" (Page 1294), and 3 co‐authors of the published paper were affiliated to the pharmaceutical company
‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

TENERE 2014

Methods

RCT

Participants

Age: 18 years and older; definite RRMS; mean disease duration 7 years; mean EDSS 2.1; prior use of DMT in the previous 2 years: 18.8% (11.7% in teriflunomide 14 mg, 21.1% in teriflunomide 7 mg, and 24.0% in interferon beta‐1a)

Interventions

Teriflunomide 14 mg oral capsule once daily for at least 12 months (n = 111)

Teriflunomide 7 mg oral capsule once daily for at least 12 months (n = 109)

Interferon beta‐1a (Rebif) 44 µg ("when the 44 μg dose was not tolerated, the dose was reduced to 22 μg") subcutaneous 3 times a week for at least 12 months (n = 104)

The study was completed 48 weeks after the last patient was randomised, resulting in a variable duration of follow‐up

Outcomes

Relapse at 12 months

Notes

Funding: Genzyme (a Sanofi company)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Patients were randomised 1:1:1 to teriflunomide 7 mg or 14 mg or IFNβ‐1a, and stratified by country (Americas, Eastern Europe, Western Europe and Africa) and baseline EDSS score (≤3.5 or >3.5)" (Page 706)

Allocation concealment (selection bias)

Low risk

"A phone interactive voice response system was used to randomize patients" (information provided on request by Genzyme)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Patients were randomised 1:1:1 to teriflunomide 7 mg or 14 mg (double‐blind) or IFNβ‐1a (open‐label)" (Page 706)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"The treating neurologist was responsible for relapse assessments, while an examining neurologist scored the EDSS. The examining neurologist remained blinded to treatment and associated AEs", and "Each relapse was confirmed by the treating neurologist based on the objective assessment of the examining neurologist" (Page 706)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, at 1 year 17.9% were lost to follow‐up (17.1% in teriflunomide 14 mg, 10.1% in teriflunomide 7 mg, and 26.9% in interferon beta‐1a) (data provided on request by Genzyme), with some indication of the differences in reasons

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes. Missing data not reported in the published paper were provided on request by Genzyme

Other bias

High risk

"This study was funded by Genzyme, a Sanofi company. Editorial support was provided by Meg Church, Fishawack Communications, Ltd, also funded by Genzyme, a Sanofi company" (Page 716), and 3 co‐authors of the published paper were affiliated to the pharmaceutical company

TOWER 2014

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 8 years; mean EDSS 2.7; prior use of DMT in the previous 2 years: 32.8% (33.9% in teriflunomide 14 mg, 30.1% in teriflunomide 7 mg, and 34.7% in placebo)

Interventions

Teriflunomide 14 mg oral capsule once daily for at least 12 months (n = 372)

Teriflunomide 7 mg oral capsule once daily for at least 12 months (n = 408)

Placebo oral capsule once daily for at least 12 months (n = 389)

The study was completed 48 weeks after the last patient was randomised, resulting in a variable duration of follow‐up

Outcomes

Relapse at 12 months

Notes

Funding: Genzyme (a Sanofi company)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation was done using a permuted‐block randomisation schedule with stratification according to study site and baseline EDSS score (≤3.5 or >3.5)" (Page 248)

Allocation concealment (selection bias)

Low risk

"Randomisation was done centrally, via an interactive voice recognition system that generated an allocation sequence" and "investigators used the allocation sequence to randomly assign eligible patients in a 1:1:1 ratio to receive once‐daily oral placebo, teriflunomide 7 mg, or teriflunomide 14 mg (identical in taste and appearance)" (Page 248)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Patients and individuals administering the interventions were masked to treatment assignment" (Page 248)

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Those assessing the outcomes were masked to treatment assignment" and "A treating neurologist was responsible for recording of adverse events, and assessment of relapses. An examining neurologist assigned EDSS scores at screening, randomisation, and every 12 weeks until the last treatment visit, and on any unscheduled visits for assessment of suspected relapse or disability worsening" (Page 248)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Overall, 29.8% were lost to follow‐up (30.6% in teriflunomide 14 mg, 29.2% in teriflunomide 7 mg, and 29.6% in placebo), with some indication of the differences in reasons: adverse events of 15.6% in teriflunomide 14 mg, 13.2% in teriflunomide 7 mg, and 6.7% in placebo; and lack of benefit of 5.4% in teriflunomide 14 mg, 7.4% in teriflunomide 7 mg, and 9.5% in placebo

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes

Other bias

High risk

‐ The study was sponsored by Genzyme, "data were analyzed by the sponsor" (Page 250), and 4 co‐authors of the published paper were affiliated to the pharmaceutical company
‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

TRASFORMS 2010

Methods

RCT

Participants

Age: 18 to 55 years; definite RRMS; mean disease duration 7 years; mean EDSS 2.2; prior use of DMT at any time prior to the start of study: 56.7% (58.5% in fingolimod 1.25 mg, 55.2% in fingolimod 0.5 mg, and 56.3% in interferon beta‐1a)

Interventions

Fingolimod 1.25 mg oral capsule once daily for 12 months (n = 426)

Fingolimod 0.5 mg oral capsule once daily for 12 months (n = 431)

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 12 months (n = 435)

Outcomes

Relapse at 12 months

Notes

Funding: Novartis Pharma

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomization was performed in blocks of six within each site and was stratified according to site" (Page 403)

Allocation concealment (selection bias)

Low risk

"Randomization was performed centrally" and "Study‐group assignments were performed with the use of an interactive voice‐response system" (Page 403)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"Capsules, syringes and packaging materials for active and placebo treatments were indistinguishable", "During the trial, patients, study personnel, steering‐committee members, and the study statistician were unaware of study‐group assignments and leukocyte counts", and "An independent physician monitored patients after the first dose of the oral study drug was administered and was instructed not to discuss heart‐rate changes with patients or study personnel" (Page 404)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

"At each site, a treating neurologist supervised medical management", "Patients were instructed to not to discuss adverse events with clinical evaluators", and "Potential relapses triggered an unscheduled visit and were confirmed by the treating neurologist on the basis of blinded examination by the examining neurologist" (Pages 403‐4)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Overall, 10.8% were lost to follow‐up (13.4% in fingolimod 1.25 mg, 7.7% in fingolimod 0.5 mg, and 11.3% in interferon beta‐1a), with few indications of the differences in reasons: unsatisfactory therapeutic effect of 0.7% in fingolimod 1.25 mg, 0.7% in fingolimod 0.5 mg, and 1.6% in interferon beta‐1a; adverse event(s) of 6.1% in fingolimod 1.25 mg, 2.1% in fingolimod 0.5 mg, and 2.1% in interferon beta‐1a; and abnormal laboratory values(s) of 0.9% in fingolimod 1.25 mg, 1.4% in fingolimod 0.5 mg, and 0.2% in interferon beta‐1a

Selective reporting (reporting bias)

Low risk

The published report included all pre‐specified primary benefit outcomes. Missing data not reported in the published paper were provided on request by Novartis Pharma

Other bias

High risk

‐ The study was sponsored by Novartis Pharma, "data were analyzed by the sponsor" (Page 403), and 5 co‐authors of the published paper were affiliated to the pharmaceutical company
‐ The primary benefit outcome measure for relapse (ARR) was strongly affected by differences among treatment groups both in the number of participants who discontinued the study and in time to discontinuation, which they did not report

ARR: annualised relapse rate
CIS: clinically isolated syndrome
DMT: disease modifying therapy
EDSS: Expanded Disability Status Scale
FDA: (US) Food and Drug Administration
MS: multiple sclerosis
RCT: randomised controlled trial
RRMS: relapsing‐remitting multiple sclerosis

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

ACT 2009

Study evaluating combination therapy (interferon beta‐1a combined with methotrexate, methylprednisolone, or both)

Ashtari 2011

Study on interferon beta‐1a versus methotrexate; methotrexate is not relevant to the review

ATAMS 2014

Study on atacicept versus placebo; atacicept is not relevant to the review

Calabrese 2012

Non‐randomised study

CHOICE 2010

Follow‐up of 6 months

Etemadifar 2006

Non‐randomised study

FORTE 2011

Study evaluating 2 doses of glatiramer acetate (40 mg compared to 20 mg) without a control group

Freedman 2012

Study evaluating combination therapy (interferon beta‐1a alone and combined with teriflunomide), with a follow‐up of 6 months

Havrdova 2009

Study evaluating combination therapy (interferon beta‐1a alone and combined with low‐dose azathioprine alone or low‐dose azathioprine and low‐dose corticosteroids)

Kappos 2006

Follow‐up of 6 months

The patients were possibly included in the FREEDOMS study

Kappos 2008

Follow‐up of 6 months

Kappos 2011

Follow‐up of 6 months

Khoury 2010

Study evaluating combination therapy (glatiramer acetate alone and combined with albuterol)

Knobler 1993

Follow‐up of 6 months

Saida 2012

Follow‐up of 6 months

SENTINEL 2006

Study evaluating combination therapy (natalizumab combined with interferon beta‐1a versus interferon beta‐1a alone)

Sorensen 2014

Follow‐up of 6 months

Characteristics of ongoing studies [ordered by study ID]

DECIDE

Trial name or title

Multicenter, double‐blind, randomized, parallel‐group, monotherapy, active‐control study to determine the efficacy and safety of daclizumab high yield process (DAC HYP) versus Avonex® (interferon β 1a) in patients with relapsing‐remitting multiple sclerosis

Methods

RCT

Participants

Inclusion criteria:

  • Aged 18 to 55 years old

  • Must have a confirmed diagnosis of relapsing remitting multiple sclerosis, and a cranial MRI demonstrating lesion(s) consistent with MS

  • Must have a baseline EDSS between 0.0 and 5.0

  • Male subjects and female subjects of childbearing potential must be willing to practice effective contraception during the study and be willing and able to continue contraception for 4 months after their last dose of study treatment

Exclusion criteria:

  • Known intolerance, contraindication to, or history of non compliance with Avonex 30 µg

  • History of treatment with daclizumab

  • History of malignancy

  • History of severe allergic or anaphylactic reactions

  • Known hypersensitivity to study drugs or their excipients

  • History of abnormal laboratory results indicative of any significant disease

  • History of human immunodeficiency virus (HIV) or other immunodeficient conditions

  • History of drug or alcohol abuse (as defined by the investigator) within the 2 years prior to randomisation

  • History of seizure disorder or unexplained blackouts or history of a seizure within 6 months prior to baseline

  • History of suicidal ideation or an episode of clinically severe depression (as determined by the investigator) within 3 months prior to day 1

  • A MS relapse that has occurred within the 50 days prior to randomisation and/or the subject has not stabilised from a previous relapse prior to randomisation

  • Known history of, or positive screening test result for, hepatitis C virus or hepatitis B virus

  • Varicella or herpes zoster virus infection or any severe viral infection within 6 weeks before screening

  • Exposure to varicella zoster virus within 21 days before screening

Interventions

Daclizumab 150 mg subcutaneously once every 4 weeks for 24 to 36 months

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 24 to 36 months

Outcomes

Primary outcome measures:

  • Annualised relapse rate (ARR) at 3 years.

Secondary outcome measures (time frame: 2 years):

  • Number of new or newly enlarging T2 hyperintense lesions on brain MRI

  • Proportion of subjects with sustained (for 3 months) disability worsening

  • Proportion of subjects who are relapse‐free

  • Proportion of subjects with a ≥ 7.5 point worsening from baseline in the MSIS‐29 physical score

Starting date

May 2010

Contact information

Biogen Idec

Notes

Sponsor: Biogen Idec

ClinicalTrials.gov Identifier: NCT01064401

NCT01247324

Trial name or title

A randomized, double‐blind, double‐dummy, parallel‐group study to evaluate the efficacy and safety of Ocrelizumab in comparison to Interferon Beta‐1a (Rebif®) in patients with relapsing multiple sclerosis

Methods

RCT

Participants

Inclusion criteria:

  • Aged 18 to 55 years old

  • Diagnosis of multiple sclerosis, in accordance with the revised McDonald criteria (2010)

  • At least 2 documented clinical attacks within the last 2 years prior to screening or 1 clinical attack in the years prior to screening (but not within 30 days prior to screening)

  • Neurologic stability for >/= 30 days prior to both screening and baseline

  • Expanded Disability Status Scale (EDSS) score 0 to 5.5

Exclusion criteria:

  • Primary progressive multiple sclerosis

  • Disease duration of more than 10 years in patients with EDSS </= 2.0 at screening

  • Contraindications for MRI

  • Known presence of other neurological disorders that may mimic multiple sclerosis

  • Pregnancy or lactation

  • Requirement for chronic treatment with systemic corticosteroids or immunosuppressants during the course of the study

  • History of or currently active primary or secondary immunodeficiency

  • History of severe allergic or anaphylactic reactions to humanised or murine monoclonal antibodies

  • Active infection, or history of or known presence of recurrent or chronic infection (e.g. hepatitis B or C, HIV, syphilis, tuberculosis)

  • History of progressive multifocal leukoencephalopathy

  • Contraindications to or intolerance of oral or intravenous corticosteroids

  • Contraindications to Rebif or incompatibility with Rebif use

Interventions

Ocrelizumab 600 mg intravenously every 24 weeks for 24 months

Interferon beta‐1a (Rebif) 8.8 µg (weeks 1 + 2)/22 µg (weeks 3 + 4)/44 µg (week 5 and following) subcutaneous 3 times a week for 24 months

Outcomes

Primary outcome measures:

  • Annualised Relapse Rate (ARR) at 2 years

Secondary outcome measures (time frame: 2 years):

  • Time to onset of sustained disability worsening for at least 3 months

  • Time to onset of sustained disability worsening for at least 6 months

  • Proportion of relapse‐free patients

  • Change in total T2 lesion volume as detected by brain MRI

  • Total number of new and/or enlarging T2 hyperintense lesions as detected by brain MRI

  • Change in Multiple Sclerosis Functional Composite Scale (MSFCS) score

  • Change in brain volume as detected by MRI

  • Safety: incidence of adverse events

  • Pharmacokinetics: exposure to ocrelizumab (area under the concentration ‐ time curve)

  • Immunogenicity: human anti‐human antibodies (HAHA) levels

Starting date

August 2011

Contact information

Hoffmann‐La Roche

Notes

Sponsor: Hoffmann‐La Roche

ClinicalTrials.gov Identifier: NCT01247324

NCT01412333

Trial name or title

A randomized, double‐blind, double‐dummy, parallel‐group study to evaluate the efficacy and safety of ocrelizumab in comparison to interferon beta‐1a (Rebif®) in patients with relapsing multiple sclerosis

Methods

RCT

Participants

Inclusion criteria:

  • Aged 18 to 55 years old

  • Diagnosis of multiple sclerosis, in accordance with the revised McDonald criteria (2010)

  • At least 2 documented clinical attacks within the last 2 years prior to screening or 1 clinical attack in the years prior to screening (but not within 30 days prior to screening)

  • Neurologic stability for >/= 30 days prior to both screening and baseline

  • Expanded Disability Status Scale (EDSS) score 0 to 5.5

Exclusion criteria:

  • Primary progressive multiple sclerosis

  • Disease duration of more than 10 years in patients with EDSS </= 2.0 at screening

  • Contraindications for MRI

  • Known presence of other neurological disorders which may mimic multiple sclerosis

  • Pregnancy or lactation

  • Requirement for chronic treatment with systemic corticosteroids or immunosuppressants during the course of the study

  • History of or currently active primary or secondary immunodeficiency

  • History of severe allergic or anaphylactic reactions to humanised or murine monoclonal antibodies

  • Active infection, or history of or known presence of recurrent or chronic infection (e.g. hepatitis B or C, HIV, syphilis, tuberculosis)

  • History of progressive multifocal leukoencephalopathy

  • Contraindications to or intolerance of oral or intravenous corticosteroids

  • Contraindications to Rebif or incompatibility with Rebif use

Interventions

Ocrelizumab 600 mg intravenously every 24 weeks for 24 months

Interferon beta‐1a (Rebif) 8.8 µg (weeks 1 + 2)/22 µg (weeks 3 + 4)/44 µg (week 5 and following) subcutaneous 3 times a week for 24 months

Outcomes

Primary outcome measures:

  • Annualised Relapse Rate (ARR) at 2 years

Secondary outcome measures (time frame: 2 years):

  • Time to onset of sustained disability worsening for at least 3 months

  • Time to onset of sustained disability worsening for at least 6 months

  • Proportion of relapse‐free patients

  • Change in total T2 lesion volume as detected by brain MRI

  • Total number of new and/or enlarging T2 hyperintense lesions as detected by brain MRI

  • Change in Multiple Sclerosis Functional Composite Scale (MSFCS) score

  • Change in brain volume as detected by MRI

  • Safety: incidence of adverse events

  • Pharmacokinetics: exposure to ocrelizumab (area under the concentration ‐ time curve)

  • Immunogenicity: human anti‐human antibodies (HAHA) levels

Starting date

September 2011

Contact information

Hoffmann‐La Roche

Notes

Sponsor: Hoffmann‐La Roche

ClinicalTrials.gov Identifier: NCT01412333

EDSS: Expanded Disability Status Scale
MRI: magnetic resonance imaging
MS: multiple sclerosis
RCT: randomised controlled trial

Data and analyses

Open in table viewer
Comparison 1. Treatment benefit within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisons for relapses over 12 months Show forest plot

29

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 1 Comparisons for relapses over 12 months.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 1 Comparisons for relapses over 12 months.

1.1 Interferon beta‐1a (Avonex) versus placebo

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.73, 1.05]

1.2 Interferon beta‐1a (Rebif) versus placebo

2

853

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.66, 1.19]

1.3 Glatiramer acetate versus placebo

4

2416

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.66, 0.95]

1.4 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.47, 0.66]

1.5 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.21, 0.74]

1.6 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.48, 0.82]

1.7 Teriflunomide versus placebo

2

2257

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.78, 0.95]

1.8 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.71, 0.88]

1.9 Pegylated interferon beta‐1a versus placebo

1

1512

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.78, 1.01]

1.10 Daclizumab versus placebo

1

621

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

1.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.61, 1.24]

1.12 Immunoglobulins versus placebo

3

219

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.47, 1.36]

1.13 Interferon beta‐1a (Rebif) versus interferon beta‐1a (Avonex)

1

677

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.80, 1.03]

1.14 Glatiramer acetate versus interferon beta‐1b (Betaseron)

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.48, 1.38]

1.15 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

2

244

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.49, 1.33]

1.16 Fingolimod versus interferon beta‐1a (Avonex)

1

1292

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.57, 0.79]

1.17 Teriflunomide versus interferon beta‐1a (Rebif)

1

324

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.69, 1.18]

1.18 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.14]

1.19 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.39, 0.55]

2 Comparisons for relapses over 24 months Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 2 Comparisons for relapses over 24 months.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 2 Comparisons for relapses over 24 months.

2.1 Interferon beta‐1b (Betaseron) versus placebo

1

372

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.81, 0.99]

2.2 Interferon beta‐1a (Avonex) versus placebo

2

1198

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.76, 1.04]

2.3 Interferon beta‐1a (Rebif) versus placebo

1

560

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.77, 0.92]

2.4 Glatiramer acetate versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.75, 0.98]

2.5 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.49, 0.64]

2.6 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.27, 0.80]

2.7 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.67, 0.78]

2.8 Teriflunomide versus placebo

1

1088

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.79, 0.98]

2.9 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.81, 0.97]

2.10 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.75, 0.99]

2.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.56, 1.05]

2.12 Immunoglobulins versus placebo

2

190

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.61, 0.90]

2.13 Interferon beta‐1a (Avonex) versus interferon beta‐1b (Betaseron)

1

188

Risk Ratio (M‐H, Random, 95% CI)

1.34 [1.06, 1.71]

2.14 Interferon beta‐1a (Rebif) versus interferon beta‐1b (Betaseron)

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.89, 1.11]

2.15 Glatiramer acetate versus interferon beta‐1b (Betaseron)

2

2319

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.47, 1.38]

2.16 Glatiramer acetate versus interferon beta‐1a (Rebif)

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.78, 1.09]

2.17 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.98, 1.21]

2.18 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.39, 0.65]

2.19 Laquinimod versus interferon beta‐1a (Avonex)

1

881

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.97, 1.32]

2.20 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.64, 1.16]

3 Comparisons for disability worsening over 24 months Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 3 Comparisons for disability worsening over 24 months.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 3 Comparisons for disability worsening over 24 months.

3.1 Interferon beta‐1b (Betaseron) versus placebo

1

372

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.72, 1.32]

3.2 Interferon beta‐1a (Avonex) versus placebo

2

1198

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.70, 1.09]

3.3 Interferon beta‐1a (Rebif) versus placebo

1

560

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.61, 0.96]

3.4 Glatiramer acetate versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.61, 1.09]

3.5 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.52, 0.80]

3.6 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.05, 0.83]

3.7 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.76, 0.99]

3.8 Teriflunomide versus placebo

1

1088

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.75, 1.01]

3.9 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.72, 0.90]

3.10 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.73, 0.95]

3.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.31, 1.34]

3.12 Immunoglobulins versus placebo

2

190

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.39, 1.24]

3.13 Interferon beta‐1a (Avonex) versus interferon beta‐1b (Betaseron)

1

188

Risk Ratio (M‐H, Random, 95% CI)

2.23 [1.29, 3.83]

3.14 Interferon beta‐1a (Rebif) versus interferon beta‐1b (Betaseron)

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.78, 1.25]

3.15 Glatiramer acetate versus interferon beta‐1b (Betaseron)

2

2319

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.86, 1.13]

3.16 Glatiramer acetate versus interferon beta‐1a (Rebif)

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.39, 0.85]

3.17 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.83, 1.22]

3.18 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.32, 0.54]

3.19 Laquinimod versus interferon beta‐1a (Avonex)

1

881

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.85, 1.33]

3.20 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.49, 1.23]

Open in table viewer
Comparison 2. Treatment acceptability within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisons for treatment discontinuation due to AEs over 12 months Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 1 Comparisons for treatment discontinuation due to AEs over 12 months.

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 1 Comparisons for treatment discontinuation due to AEs over 12 months.

1.1 Interferon beta‐1a (Avonex) 30 µg versus placebo

1

301

Risk Ratio (M‐H, Random, 95% CI)

3.17 [0.67, 15.00]

1.2 Interferon beta‐1a (Rebif) 22 µg versus placebo

1

195

Risk Ratio (M‐H, Random, 95% CI)

3.16 [0.13, 76.54]

1.3 Interferon beta‐1a (Rebif) 44 µg versus placebo

1

198

Risk Ratio (M‐H, Random, 95% CI)

11.22 [0.63, 200.27]

1.4 Glatiramer acetate 20 mg daily versus placebo

1

239

Risk Ratio (M‐H, Random, 95% CI)

1.51 [0.26, 8.89]

1.5 Glatiramer 40 mg three times per week versus placebo

1

1404

Risk Ratio (M‐H, Random, 95% CI)

2.36 [0.99, 5.65]

1.6 Teriflunomide 7 mg versus placebo

1

797

Risk Ratio (M‐H, Random, 95% CI)

2.06 [1.31, 3.24]

1.7 Teriflunomide 14 mg versus placebo

1

761

Risk Ratio (M‐H, Random, 95% CI)

2.51 [1.61, 3.91]

1.8 Pegylated interferon beta‐1a every 4 weeks versus placebo

1

1000

Risk Ratio (M‐H, Random, 95% CI)

2.78 [1.31, 5.89]

1.9 Pegylated interferon beta‐1a every 2 weeks versus placebo

1

1012

Risk Ratio (M‐H, Random, 95% CI)

2.82 [1.34, 5.96]

1.10 Daclizumab 150 mg versus placebo

1

412

Risk Ratio (M‐H, Random, 95% CI)

3.43 [0.72, 16.33]

1.11 Daclizumab 300 mg versus placebo

1

413

Risk Ratio (M‐H, Random, 95% CI)

4.39 [0.96, 20.08]

1.12 Immunoglobulins 0.2 g versus placebo

2

163

Risk Ratio (M‐H, Random, 95% CI)

2.14 [0.23, 19.96]

1.13 Immunoglobulins 0.4 g versus placebo

1

34

Risk Ratio (M‐H, Random, 95% CI)

3.35 [0.15, 76.93]

1.14 Interferon beta‐1a (Rebif) 44 µg versus interferon beta‐1a (Avonex) 30 µg

1

677

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.56, 1.97]

1.15 Fingolimod 0.5 mg versus interferon beta‐1a (Avonex) 30 µg

1

866

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.81, 2.54]

1.16 Fingolimod 1.25 mg versus interferon beta‐1a (Avonex) 30 µg

1

861

Risk Ratio (M‐H, Random, 95% CI)

2.36 [1.40, 3.98]

1.17 Teriflunomide 7 mg versus interferon beta‐1a (Rebif) 44 µg

1

213

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.19, 0.81]

1.18 Teriflunomide 14 mg versus interferon beta‐1a (Rebif) 44 µg

1

215

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

1.19 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

94

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.21, 4.70]

2 Comparisons for treatment discontinuation due to AEs over 24 months Show forest plot

23

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 2 Comparisons for treatment discontinuation due to AEs over 24 months.

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 2 Comparisons for treatment discontinuation due to AEs over 24 months.

2.1 Interferon beta‐1b (Betaseron) 50 µg versus placebo

1

248

Risk Ratio (M‐H, Random, 95% CI)

4.92 [0.58, 41.51]

2.2 Interferon beta‐1b (Betaseron) 250 µg versus placebo

1

247

Risk Ratio (M‐H, Random, 95% CI)

9.92 [1.29, 76.32]

2.3 Interferon beta‐1a (Avonex) 30 µg versus placebo

1

897

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.81, 2.54]

2.4 Interferon beta‐1a (Rebif) 22 µg versus placebo

1

376

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.61, 8.79]

2.5 Interferon beta‐1a (Rebif) 44 µg versus placebo

1

371

Risk Ratio (M‐H, Random, 95% CI)

3.05 [0.84, 11.08]

2.6 Glatiramer acetate 20 mg daily versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

1.74 [0.49, 6.13]

2.7 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.93, 2.53]

2.8 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

9.82 [0.57, 168.84]

2.9 Fingolimod 0.5 mg versus placebo

2

1556

Risk Ratio (M‐H, Random, 95% CI)

1.42 [0.89, 2.25]

2.10 Fingolimod 1.25 mg versus placebo

2

1572

Risk Ratio (M‐H, Random, 95% CI)

1.93 [1.48, 2.52]

2.11 Teriflunomide 7 mg versus placebo

1

729

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.77, 1.96]

2.12 Teriflunomide 14 mg versus placebo

1

722

Risk Ratio (M‐H, Random, 95% CI)

1.36 [0.86, 2.15]

2.13 Dimethyl fumarate 480 mg versus placebo

2

1546

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.91, 1.51]

2.14 Dimethyl fumarate 720 mg versus placebo

2

1534

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.93, 1.54]

2.15 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.96, 2.00]

2.16 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

5.8 [0.74, 45.26]

2.17 Immunoglobulins 0.15 to 0.20 g versus placebo

1

150

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.32, 28.19]

2.18 Interferon beta‐1a (Avonex) 30 µg versus interferon beta‐1b (Betaseron) 250 µg

1

188

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.02, 1.75]

2.19 Glatiramer acetate 20 mg daily versus interferon beta‐1b (Betaseron) 250 μg

2

1420

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.56, 2.53]

2.20 Glatiramer acetate 20 mg daily versus interferon beta‐1b (Betaseron) 500 μg

1

1347

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.37, 1.68]

2.21 Glatiramer acetate 20 mg daily versus interferon beta‐1a (Rebif) 44 µg

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.47, 1.52]

2.22 Dimethyl fumarate 480 mg versus glatiramer acetate 20 mg daily

1

722

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.84]

2.23 Dimethyl fumarate 720 mg versus glatiramer acetate 20 mg daily

1

705

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.85]

2.24 Alemtuzumab 12 mg versus interferon beta‐1a (Rebif) 44 µg

3

1472

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.22, 0.68]

2.25 Alemtuzumab 24 mg versus interferon beta‐1a (Rebif) 44 µg

2

625

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.10, 1.09]

2.26 Laquinimod versus interferon beta‐1a (Avonex) 30 µg

1

881

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.49, 1.45]

2.27 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

2.21 [0.90, 5.45]

Open in table viewer
Comparison 3. Treatment safety against placebo within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Treatment safety against placebo within pairwise comparisons, Outcome 1 Serious adverse events.

Comparison 3 Treatment safety against placebo within pairwise comparisons, Outcome 1 Serious adverse events.

1.1 Interferons beta (Avonex, Rebif or Betaseron) versus placebo

3

870

Risk Ratio (M‐H, Random, 95% CI)

1.26 [0.67, 2.37]

1.2 Glatiramer acetate versus placebo

2

490

Risk Ratio (M‐H, Random, 95% CI)

1.87 [0.73, 4.74]

1.3 Natalizumab versus placebo

1

939

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.81, 1.73]

1.4 Fingolimod versus placebo

2

1572

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.72, 1.30]

1.5 Teriflunomide versus placebo

1

718

Risk Ratio (M‐H, Random, 95% CI)

1.26 [0.87, 1.83]

1.6 Dimethyl fumarate versus placebo

2

1531

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.76, 1.55]

1.7 Pegylated interferon beta‐1a versus placebo

1

1012

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.57, 1.68]

1.8 Daclizumab versus placebo

1

413

Risk Ratio (M‐H, Random, 95% CI)

1.55 [0.77, 3.10]

1.9 Laquinimod versus placebo

2

1988

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.67, 1.41]

1.10 Immunoglobulins versus placebo

1

83

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.11, 3.70]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 2

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

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

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

Network plots of treatment comparisons for benefit and acceptability outcomes.
Figuras y tablas -
Figure 4

Network plots of treatment comparisons for benefit and acceptability outcomes.

Network meta‐analysis (NMA) estimates of treatment benefit against placebo: relapses over 12 and 24 months, and disability worsening over 24 months.CI: confidence interval; RR: risk ratio.
Figuras y tablas -
Figure 5

Network meta‐analysis (NMA) estimates of treatment benefit against placebo: relapses over 12 and 24 months, and disability worsening over 24 months.

CI: confidence interval; RR: risk ratio.

Network meta‐analysis (NMA) estimates of treatment acceptability against placebo: treatment discontinuation due to AEs over 12 and 24 months.AEs: adverse events; CI: confidence interval; RR: risk ratio.
Figuras y tablas -
Figure 6

Network meta‐analysis (NMA) estimates of treatment acceptability against placebo: treatment discontinuation due to AEs over 12 and 24 months.

AEs: adverse events; CI: confidence interval; RR: risk ratio.

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 12 months for each comparison: relapses and treatment discontinuation due to adverse events (AEs) over 12 months.Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate (risk ratio, RR) is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dacliz: daclizumab; Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Mitoxan: mitoxantrone; Nataliz: natalizumab; PegIFNß: pegylated interferon beta‐1a; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.
Figuras y tablas -
Figure 7

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 12 months for each comparison: relapses and treatment discontinuation due to adverse events (AEs) over 12 months.

Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate (risk ratio, RR) is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.

Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dacliz: daclizumab; Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Mitoxan: mitoxantrone; Nataliz: natalizumab; PegIFNß: pegylated interferon beta‐1a; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 24 months for each comparison: relapses and treatment discontinuation due to adverse events (AEs) over 24 months. Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate (risk ratio, RR) is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Laquin: laquinimod; Mitoxan: mitoxantrone; Nataliz: natalizumab; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.
Figuras y tablas -
Figure 8

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 24 months for each comparison: relapses and treatment discontinuation due to adverse events (AEs) over 24 months. Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate (risk ratio, RR) is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.

Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Laquin: laquinimod; Mitoxan: mitoxantrone; Nataliz: natalizumab; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.

Clustered ranking plot based on cluster analysis of surface under the cumulative ranking curve (SUCRA) values for benefit (relapses) and acceptability (treatment discontinuation due to AEs) over 24 months. Each colour represents a group of treatments that belong to the same cluster. Treatments lying in the upper right corner are more effective and acceptable than the other treatments.
Figuras y tablas -
Figure 9

Clustered ranking plot based on cluster analysis of surface under the cumulative ranking curve (SUCRA) values for benefit (relapses) and acceptability (treatment discontinuation due to AEs) over 24 months. Each colour represents a group of treatments that belong to the same cluster. Treatments lying in the upper right corner are more effective and acceptable than the other treatments.

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 24 months for each comparison: disability worsening and treatment discontinuation due to adverse events (AEs) over 24 months. Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Laquin: laquinimod; Mitoxan: mitoxantrone; Nataliz: natalizumab; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.
Figuras y tablas -
Figure 10

Network meta‐analysis (NMA) estimates of treatment benefit (lower triangle) and acceptability (upper triangle) over 24 months for each comparison: disability worsening and treatment discontinuation due to adverse events (AEs) over 24 months. Drugs are reported in order of primary benefit ranking. Comparisons should be read from left to right. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. A RR value below 1 favours the column‐defining treatment for lower triangle, and the row‐defining treatment for upper triangle. To obtain RRs for comparisons in the opposing direction, reciprocals should be taken. Significant results are bolded and underscored.

Alemtuz: alemtuzumab; Avonex: interferon beta‐1a (Avonex); Aza: azathioprine; Betaseron: interferon beta‐1b (Betaseron); Dimethyl: dimethyl fumarate; Fingolim: fingolimod; Glatir: glatiramer acetate; IFNß: interferons beta; Immunogl: immunoglobulins; Laquin: laquinimod; Mitoxan: mitoxantrone; Nataliz: natalizumab; Rebif: interferon beta‐1a (Rebif); Terifl: teriflunomide.

Clustered ranking plot based on cluster analysis of surface under the cumulative ranking curve (SUCRA) values for benefit (disability worsening) and acceptability (treatment discontinuation due to AEs) over 24 months. Each colour represents a group of treatments that belong to the same cluster. Treatments lying in the upper right corner are more effective and acceptable than the other treatments.
Figuras y tablas -
Figure 11

Clustered ranking plot based on cluster analysis of surface under the cumulative ranking curve (SUCRA) values for benefit (disability worsening) and acceptability (treatment discontinuation due to AEs) over 24 months. Each colour represents a group of treatments that belong to the same cluster. Treatments lying in the upper right corner are more effective and acceptable than the other treatments.

Inconsistency plots for relapses over 12 and 24 months and disability worsening over 24 months assuming loop‐specific heterogeneity estimates. RRR is calculated as the risk ratio for direct evidence over the risk ratio for indirect evidence in the loop and it is reported together with its 95% confidence interval (CI). RRR values close to one indicate the absence of evidence for disagreement between direct and indirect evidence.
Figuras y tablas -
Figure 12

Inconsistency plots for relapses over 12 and 24 months and disability worsening over 24 months assuming loop‐specific heterogeneity estimates. RRR is calculated as the risk ratio for direct evidence over the risk ratio for indirect evidence in the loop and it is reported together with its 95% confidence interval (CI). RRR values close to one indicate the absence of evidence for disagreement between direct and indirect evidence.

Inconsistency plots for acceptability over 12 and 24 months assuming loop‐specific heterogeneity estimates. RRR is calculated as the risk ratio for direct evidence over the risk ratio for indirect evidence in the loop and it is reported together with its 95% confidence interval (CI). RRR values close to one indicate the absence of evidence for disagreement between direct and indirect evidence.
Figuras y tablas -
Figure 13

Inconsistency plots for acceptability over 12 and 24 months assuming loop‐specific heterogeneity estimates. RRR is calculated as the risk ratio for direct evidence over the risk ratio for indirect evidence in the loop and it is reported together with its 95% confidence interval (CI). RRR values close to one indicate the absence of evidence for disagreement between direct and indirect evidence.

Study limitations distribution for each network estimate for pairwise comparisons versus placebo on relapses over 12 and 24 months and disability worsening over 24 months outcomes. Calculations are based on the contributions of direct evidence to the network estimates and the overall risks of bias considering our predefined criteria (allocation concealment, blinding of outcome assessor, and incomplete outcome data) within studies contributing to the direct evidence. The colours represent risk (green, low; yellow, moderate; red, high). The direct comparisons against placebo are described in the vertical axis.
Figuras y tablas -
Figure 14

Study limitations distribution for each network estimate for pairwise comparisons versus placebo on relapses over 12 and 24 months and disability worsening over 24 months outcomes. Calculations are based on the contributions of direct evidence to the network estimates and the overall risks of bias considering our predefined criteria (allocation concealment, blinding of outcome assessor, and incomplete outcome data) within studies contributing to the direct evidence. The colours represent risk (green, low; yellow, moderate; red, high). The direct comparisons against placebo are described in the vertical axis.

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for relapses over 12 months outcome. Rows correspond to NMA risk ratios of each treatment versus placebo (separated for mixed and indirect evidence) and the columns correspond to direct meta‐analysis risk ratios. The last row shows the number of included direct comparisons. The names of the treatment comparisons are shown in the first column. For example, for relapses over 12 months, information for the network estimate of interferon beta 1a (Avonex) versus placebo is derived from both direct and indirect evidence (generating a mixed estimate). Of this mixed network estimate, trials directly comparing interferon beta 1a (Avonex) versus placebo contribute 31.2% of the information to the network estimate of effect and trials directly comparing interferon beta 1a (Rebif) versus interferon beta 1a (Avonex) contribute 18.8% of the network estimated effect, etc. The contribution matrix shows how much each direct comparison in the network contributes to each network (mixed or indirect) estimate and to the entire network.
Figuras y tablas -
Figure 15

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for relapses over 12 months outcome. Rows correspond to NMA risk ratios of each treatment versus placebo (separated for mixed and indirect evidence) and the columns correspond to direct meta‐analysis risk ratios. The last row shows the number of included direct comparisons. The names of the treatment comparisons are shown in the first column. For example, for relapses over 12 months, information for the network estimate of interferon beta 1a (Avonex) versus placebo is derived from both direct and indirect evidence (generating a mixed estimate). Of this mixed network estimate, trials directly comparing interferon beta 1a (Avonex) versus placebo contribute 31.2% of the information to the network estimate of effect and trials directly comparing interferon beta 1a (Rebif) versus interferon beta 1a (Avonex) contribute 18.8% of the network estimated effect, etc. The contribution matrix shows how much each direct comparison in the network contributes to each network (mixed or indirect) estimate and to the entire network.

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for relapses over 24 months outcome.
Figuras y tablas -
Figure 16

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for relapses over 24 months outcome.

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for disability worsening over 24 months outcome.
Figuras y tablas -
Figure 17

Contribution matrix: percentage contribution of each direct estimate to the NMA estimates versus placebo for disability worsening over 24 months outcome.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 1 Comparisons for relapses over 12 months.
Figuras y tablas -
Analysis 1.1

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 1 Comparisons for relapses over 12 months.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 2 Comparisons for relapses over 24 months.
Figuras y tablas -
Analysis 1.2

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 2 Comparisons for relapses over 24 months.

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 3 Comparisons for disability worsening over 24 months.
Figuras y tablas -
Analysis 1.3

Comparison 1 Treatment benefit within pairwise comparisons, Outcome 3 Comparisons for disability worsening over 24 months.

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 1 Comparisons for treatment discontinuation due to AEs over 12 months.
Figuras y tablas -
Analysis 2.1

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 1 Comparisons for treatment discontinuation due to AEs over 12 months.

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 2 Comparisons for treatment discontinuation due to AEs over 24 months.
Figuras y tablas -
Analysis 2.2

Comparison 2 Treatment acceptability within pairwise comparisons, Outcome 2 Comparisons for treatment discontinuation due to AEs over 24 months.

Comparison 3 Treatment safety against placebo within pairwise comparisons, Outcome 1 Serious adverse events.
Figuras y tablas -
Analysis 3.1

Comparison 3 Treatment safety against placebo within pairwise comparisons, Outcome 1 Serious adverse events.

Summary of findings for the main comparison. Summary of findings for the main comparisons of treatment effects against placebo

Patient or population: patients with relapsing‐remitting multiple sclerosis (RRMS)

Settings: secondary healthcare centres

Intervention: any immunomodulators or immunosuppressants used for RRMS

Comparison: placebo

Intervention

Illustrative comparative risks*

Relative effect
(95% CI)

SUCRA

No of participants
(studies)#

Confidence in the evidence
(GRADE)

Reasons for downgrading
our confidence in the evidence°

Assumed risk with placebo

Corresponding risk with intervention
(95% CI)

CHANCE OF EXPERIENCING ONE OR MORE RELAPSES OVER 12 MONTHS

Alemtuzumab

Low

RR 0.40

(0.31 to 0.51)

97%

Moderate

Downgraded one level due to risk of bias ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains

41 per 100

16 per 100
(13 to 21)

High

89 per 100

36 per 100
(28 to 45)

Mitoxantrone

Low

RR 0.40

(0.20 to 0.76)

93%

51
(1 study)

Low

Downgraded two levels due to risk of bias ‐ the singular study contributing to this estimate at high risk of bias in blinding of outcome assessor domain

41 per 100

16 per 100
(8 to 31)

High

89 per 100

36 per 100
(18 to 68)

Natalizumab

Low

RR 0.56

(0.43 to 0.73)

85%

942
(1 study)

High

41 per 100

23 per 100
(18 to 30)

High

89 per 100

50 per 100
(38 to 65)

Fingolimod

Low

RR 0.63

(0.53 to 0.74)

80%

2355
(2 studies)

Low

Downgraded one level due to risk of bias and one level due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; I2 = 82% (P value = 0.02)

41 per 100

26 per 100
(22 to 30)

High

89 per 100

56 per 100
(47 to 66)

Dimethyl fumarate

Low

RR 0.78

(0.65 to 0.93)

55%

2307
(2 studies)

Moderate

Downgraded one level due to inconsistency ‐ wide predictive interval

41 per 100

32 per 100
(27 to 38)

High

89 per 100

69 per 100
(58 to 83)

Immunoglobulins

Low

RR 0.78

(0.61 to 1.00)

53%

219
(3 studies)

Very low

Downgraded one level due to risk of bias, two levels due to inconsistency, and one level due to imprecision ‐ the majority of studies at unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; I2 = 83% (P value = 0.003) and differences between pairwise and common τ2 (0.18 versus 0.01); wide CIs

41 per 100

32 per 100
(25 to 41)

High

89 per 100

69 per 100
(54 to 89)

Glatiramer acetate

Low

RR 0.80

(0.68 to 0.93)

52%

2416
(4 studies)

Moderate

Downgraded one level due to inconsistency ‐ wide predictive interval

41 per 100

33 per 100
(28 to 38)

High

89 per 100

71 per 100
(61 to 83)

Daclizumab

Low

RR 0.79

(0.61 to 1.02)

52%

621
(1 study)

Moderate

Downgraded one level due to imprecision ‐ wide CIs

41 per 100

32 per 100
(25 to 42)

High

89 per 100

70 per 100
(54 to 91)

Teriflunomide

Low

RR 0.84

(0.72 to 0.99)

42%

2257
(2 studies)

Low

Downgraded one level due to risk of bias and one level due to inconsistency ‐ the majority of studies at unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; wide predictive interval

41 per 100

34 per 100
(30 to 41)

High

89 per 100

75 per 100
(64 to 88)

Azathioprine

Low

RR 0.87

(0.58 to 1.31)

39%

59
(1 study)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and two levels due to imprecision ‐ the singular study contributing to this estimate at unclear risk of bias in allocation concealment domain; indirectness of population (one monocentric study); wide CIs

41 per 100

36 per 100
(24 to 54)

High

89 per 100

77 per 100
(52 to 100)

Interferon beta‐1a (Rebif)

Low

RR 0.87

(0.76 to 1.01)

36%

853
(2 studies)

Low

Downgraded one level due to risk of bias and one level due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; I2 = 88% (P value = 0.004)

41 per 100

36 per 100
(31 to 41)

High

89 per 100

77 per 100
(68 to 90)

Pegylated interferon beta‐1a

Low

RR 0.89

(0.70 to 1.13)

33%

1512
(1 study)

Low

Downgraded one level due to risk of bias and one level due to imprecision ‐ the singular study contributing to this estimate at unclear risk of bias in blinding of outcome assessor domain; wide CIs

41 per 100

36 per 100
(29 to 46)

High

89 per 100

79 per 100
(62 to 100)

Interferon beta‐1b (Betaseron)

Low

RR 0.98

(0.54 to 1.75)

27%

Very low

Downgraded one level due to risk of bias and two levels due to imprecision ‐ the majority of studies at unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; wide CIs

41 per 100

40 per 100
(22 to 72)

High

89 per 100

87 per 100
(48 to 100)

Interferon beta‐1a (Avonex)

Low

RR 0.93

(0.78 to 1.10)

25%

301
(1 study)

Moderate

Downgraded one level due to risk of bias ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains

41 per 100

38 per 100
(32 to 45)

High

89 per 100

83 per 100
(69 to 98)

Interferons beta (Avonex, Rebif or Betaseron)

Low

RR 1.05

(0.61 to 1.79)

20%

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and two levels due to imprecision ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; indirectness of population (one monocentric study contributing 50% to this estimate); wide CIs

41 per 100

43 per 100
(25 to 73)

High

89 per 100

93 per 100
(54 to 100)

CHANCE OF EXPERIENCING ONE OR MORE RELAPSES OVER 24 MONTHS

Alemtuzumab

Low

RR 0.46

(0.38 to 0.55)

96%

Moderate

Downgraded one level due to risk of bias ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains

57 per 100

26 per 100
(22 to 31)

High

85 per 100

39 per 100
(32 to 47)

Mitoxantrone

Low

RR 0.47

(0.27 to 0.81)

92%

51
(1 study)

Very low

Downgraded two levels due to risk of bias and one level due to inconsistency ‐ the singular study contributing to this estimate at high risk of bias in blinding of outcome assessor domain; wide predictive interval

57 per 100

27 per 100
(15 to 46)

High

85 per 100

40 per 100
(23 to 69)

Natalizumab

Low

RR 0.56

(0.47 to 0.66)

88%

942
(1 study)

High

57 per 100

32 per 100
(27 to 38)

High

85 per 100

48 per 100
(40 to 56)

Fingolimod

Low

RR 0.72

(0.64 to 0.81)

71%

2355
(2 studies)

Moderate

Downgraded one level due to risk of bias ‐ studies at unclear risk of bias in allocation concealment domain

57 per 100

41 per 100
(36 to 46)

High

85 per 100

61 per 100
(54 to 69)

Immunoglobulins

Low

RR 0.74

(0.60 to 0.91)

66%

190
(2 studies)

Moderate

Downgraded one level due to inconsistency ‐ wide predictive interval

57 per 100

42 per 100
(34 to 52)

High

85 per 100

63 per 100
(51 to 77)

Azathioprine

Low

RR 0.77

(0.55 to 1.07)

57%

59
(1 study)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and one level due to imprecision ‐ the singular study contributing to this estimate at unclear risk of bias in allocation concealment domain; indirectness of population (one monocentric study); wide CIs

57 per 100

44 per 100
(31 to 61)

High

85 per 100

65 per 100
(47 to 91)

Glatiramer acetate

Low

RR 0.83

(0.75 to 0.91)

48%

1024
(3 studies)

Moderate

Downgraded one level due to inconsistency ‐ wide predictive interval

57 per 100

47 per 100
(43 to 52)

High

85 per 100

71 per 100
(64 to 77)

Interferon beta‐1b (Betaseron)

Low

RR 0.85

(0.77 to 0.94)

42%

372
(1 study)

Very low

Downgraded one level due to risk of bias and two levels due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; wide predictive interval and inconsistent loops of evidence

57 per 100

48 per 100
(44 to 54)

High

85 per 100

72 per 100
(65 to 80)

Interferon beta‐1a (Rebif)

Low

RR 0.86

(0.77 to 0.95)

39%

560
(1 study)

Low

Downgraded one level due to risk of bias and one level due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; wide predictive interval

57 per 100

49 per 100
(44 to 54)

High

85 per 100

73 per 100
(65 to 81)

Interferons beta (Avonex, Rebif or Betaseron)

Low

RR 0.89

(0.56 to 1.42)

33%

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and one level due to imprecision ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; indirectness of population (one monocentric study contributing for 50% to this estimate); wide CIs

57 per 100

51 per 100
(32 to 81)

High

85 per 100

76 per 100
(48 to 100)

Teriflunomide

Low

RR 0.88

(0.75 to 1.03)

32%

1088
(1 study)

Very low

Downgraded two levels due to risk of bias and one level due to imprecision ‐ the singular study contributing to this estimate at high risk of bias in blinding of outcome assessor domain; wide CIs

57 per 100

50 per 100
(43 to 59)

High

85 per 100

75 per 100
(64 to 88)

Laquinimod

Low

RR 0.88

(0.79 to 0.99)

31%

1990
(2 studies)

Very low

Downgraded one level due to risk of bias and two levels due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; I2 = 66% (P value = 0.09), wide predictive interval and inconsistent loops of evidence

57 per 100

50 per 100
(45 to 56)

High

85 per 100

75 per 100
(67 to 84)

Dimethyl fumarate

Low

RR 0.89

(0.81 to 0.98)

30%

2307
(2 studies)

Moderate

Downgraded one level due to inconsistency ‐ wide predictive interval

57 per 100

51 per 100
(46 to 56)

High

85 per 100

76 per 100
(69 to 83)

Interferon beta‐1a (Avonex)

Low

RR 0.91

(0.82 to 1.02)

22%

1198
(2 studies)

Low

Downgraded one level due to risk of bias and one level due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; inconsistent loops of evidence

57 per 100

52 per 100
(47 to 58)

High

85 per 100

77 per 100
(70 to 87)

CHANCE OF DISABILITY GETTING WORSE OVER 24 MONTHS

Mitoxantrone

Low

RR 0.20

(0.05 to 0.84)

96%

51
(1 study)

Low

Downgraded one level due to indirectness and one level due to inconsistency ‐ surrogate outcome unclear; wide predictive interval

25 per 100

5 per 100
(1 to 21)

High

52 per 100

10 per 100
(3 to 44)

Alemtuzumab

Low

RR 0.35

(0.26 to 0.48)

94%

Low

Downgraded one level due to risk of bias and one level due to indirectness ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; surrogate outcome in the majority of studies contributing to this estimate

25 per 100

9 per 100
(6 to 12)

High

52 per 100

18 per 100
(14 to 25)

Natalizumab

Low

RR 0.64

(0.49 to 0.85)

74%

942
(1 study)

Moderate

Downgraded one level due to indirectness ‐ surrogate outcome

25 per 100

16 per 100
(12 to 21)

High

52 per 100

33 per 100
(25 to 44)

Azathioprine

Low

RR 0.64

(0.30 to 1.37)

64%

59
(1 study)

Very low

Downgraded one level due to risk of bias, two levels due to indirectness, and two levels due to imprecision ‐ the singular study contributing to this estimate at unclear risk of bias in allocation concealment domain; indirectness of population (one monocentric study) and surrogate outcome unclear; wide CIs

25 per 100

16 per 100
(8 to 34)

High

52 per 100

33 per 100
(16 to 71)

Glatiramer acetate

Low

RR 0.77

(0.64 to 0.92)

58%

1024
(3 studies)

Very low

Downgraded one level due to indirectness and two levels due to inconsistency ‐ surrogate outcome in the majority of studies contributing to this estimate; wide predictive interval and inconsistent loops of evidence

25 per 100

19 per 100
(16 to 23)

High

52 per 100

40 per 100
(33 to 48)

Immunoglobulins

Low

RR 0.70

(0.39 to 1.27)

56%

190
(2 studies)

Very low

Downgraded one level due to indirectness, one level due to inconsistency, and two levels due to imprecision ‐ surrogate outcome in the majority of studies contributing to this estimate; wide predictive interval; wide CIs

25 per 100

18 per 100
(10 to 32)

High

52 per 100

36 per 100
(20 to 66)

Interferon beta‐1b (Betaseron)

Low

RR 0.79

(0.65 to 0.97)

51%

372
(1 study)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and two levels due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; surrogate outcome in the majority of studies contributing to this estimate; wide predictive interval and inconsistent loops of evidence

25 per 100

20 per 100
(16 to 24)

High

52 per 100

41 per 100
(34 to 50)

Dimethyl fumarate

Low

RR 0.80

(0.67 to 0.94)

50%

2307
(2 studies)

Low

Downgraded one level due to indirectness and one level due to inconsistency ‐ surrogate outcome in the majority of studies contributing to this estimate; wide predictive interval

25 per 100

20 per 100
(17 to 23)

High

52 per 100

42 per 100
(35 to 49)

Interferons beta (Avonex, Rebif or Betaseron)

Low

RR 0.83

(0.34 to 2.07)

40%

Very low

Downgraded one level due to indirectness, one level due to inconsistency, and two levels due to imprecision ‐ indirectness of population and surrogate outcome unclear (one study contributing for 50% to this estimate); wide predictive interval; wide CIs

25 per 100

21 per 100
(9 to 52)

High

52 per 100

43 per 100
(18 to 100)

Interferon beta‐1a (Rebif)

Low

RR 0.86

(0.69 to 1.06)

36%

560
(1 study)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, one level due to inconsistency, and one level due to imprecision ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; surrogate outcome in the majority of studies contributing to this estimate; inconsistent loops of evidence; wide CIs

25 per 100

22 per 100
(17 to 26)

High

52 per 100

45 per 100
(36 to 55)

Fingolimod

Low

RR 0.86

(0.73 to 1.03)

34%

2355
(2 studies)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and one level due to imprecision ‐ studies at unclear risk of bias in allocation concealment domain; surrogate outcome; wide CIs

25 per 100

22 per 100
(18 to 26)

High

52 per 100

45 per 100
(38 to 54)

Laquinimod

Low

RR 0.87

(0.72 to 1.04)

34%

1990
(2 studies)

Low

Downgraded one level due to indirectness and one level due to imprecision ‐ surrogate outcome in the majority of studies contributing to this estimate; wide CIs

25 per 100

22 per 100
(18 to 26)

High

52 per 100

45 per 100
(37 to 54)

Teriflunomide

Low

RR 0.87

(0.69 to 1.10)

34%

1088
(1 study)

Low

Downgraded one level due to indirectness and one level due to imprecision ‐ surrogate outcome; wide CIs

25 per 100

22 per 100
(17 to 28)

High

52 per 100

45 per 100
(36 to 57)

Interferon beta‐1a (Avonex)

Low

RR 0.93

(0.77 to 1.13)

21%

1198
(2 studies)

Very low

Downgraded one level due to risk of bias, one level due to indirectness, and two levels due to inconsistency ‐ the majority of studies at high or unclear risk of bias in allocation concealment and/or blinding of outcome assessor domains; surrogate outcome in the majority of studies contributing to this estimate; I2 = 57% (P value = 0.13), and inconsistent loops of evidence

25 per 100

23 per 100
(19 to 28)

High

52 per 100

48 per 100
(40 to 59)

*The corresponding risk with intervention (and its 95% confidence interval) is based on the assumed risk with placebo and the relative effect of the intervention (and its 95% CI). Two values were chosen for the assumed risk with placebo, i.e. the second highest and second lowest placebo group risks in the included studies, defined as low and high assumed risk.

#No of Participants (studies) is not available when the nature of the evidence is indirect.

°We did not downgrade for reasons of reporting bias as insufficient studies contributed to network treatment estimates to draw meaningful conclusions.
CI: confidence interval; RR: risk ratio; SUCRA: surface under the cumulative ranking curve

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

Figuras y tablas -
Summary of findings for the main comparison. Summary of findings for the main comparisons of treatment effects against placebo
Table 1. Assessment of adverse events monitoring

Study

Risk of bias

Did the researchers actively monitor for adverse events (AEs) or did they simply provide spontaneous reporting of AEs that arose?

Risk of bias

Did the authors define serious AEs (SAEs) according to an accepted international classification and report the number of SAEs?

Achiron 1998

Unclear

Not reported

High

SAEs not reported

ADVANCE 2014

Unclear

Not reported

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

AFFIRM 2006

Low

"Treating neurologists were responsible for all aspects of patient care, including the management of adverse events". Participants"visited the clinic every 12 weeks for ... blood chemical and hematologic analyses, evaluation of adverse events..." (Page 901)

Unclear

Insufficient information on SAEs definition

ALLEGRO 2012

Low

"Safety assessments were performed at screening, at baseline, and every 3 months until month 24" (Page 1002)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

BECOME 2009

Low

"After the initial interim analysis failed to raise any safety concerns with the use of monthly triple dose gadolinium, all patients still in the study were offered the option of obtaining additional monthly MRI scans for a second year of treatment" (Page 1977)

High

SAEs not reported

BEYOND 2009

Low

"Clinic visits were scheduled every 3 months to assess ... safety, and tolerability. The occurrence of new neurological symptoms and adverse events was assessed by telephone, 6 weeks after each visit" (Page 891)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

Bornstein 1987

High

"Self‐evaluation reported to a clinical assistant" (Page 409)

High

SAEs not reported

BRAVO 2014

Low

"Patients were evaluated at 12 scheduled visits: months ‐1 (screening), 0 (baseline), 1, 2, 3, 6, 9, 12, 15, 18, 21, and 24. Safety assessments (laboratory measures, vital signs) were performed at all visits, and electrocardiograms (ECGs) were performed at months ‐1, 0, 1, 2, 3, 6, 12, 18, and 24/early termination" (Page 775)

Unclear

Insufficient information on SAEs definition

CAMMS223 2008

Low

"Safety was assessed quarterly by the treating neurologist, who was aware of study‐group assignment" (Page 1787), "Thyroid function and levels of antithyrotropinreceptor antibodies and lymphocyte subpopulations were measured quarterly at a central laboratory", and "All adverse events with an onset up to 36 months are reported. In addition, all serious adverse events and autoimmune‐associated disorders occurring before March 1, 2008, are listed" (Page 1788)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

CARE‐MS I 2012

Low

"To assess safety, we undertook monthly questionnaire follow‐up of patients, and did complete blood counts, serum creatinine, urinalysis, and microscopy monthly (every three months in patients in the interferon beta 1a group), and thyroid function tests every 3 months", "Circulating lymphocyte subsets were assessed every 3 months in all patients and 1 month after alemtuzumab administration. We screened for antialemtuzumab antibodies with a bridging ELISA before and at 1 month, 3 months, and 12 months after each dosing", and "We measured interferon beta 1a‐neutralising antibodies at baseline and at 24 months with a cytopathic effect inhibition assay" (Page 1821)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use).

CARE‐MS II 2012

Low

"To assess safety, we undertook monthly questionnaire follow‐up of patients, and did complete blood counts, serum creatinine, and urinalysis with microscopy monthly (every 3 months in patients in the interferon beta 1a group), and thyroid function tests every 3 months", "We assessed circulating lymphocyte subsets every 3 months in all patients and 1 month after every course of alemtuzumab. We screened for anti‐alemtuzumab antibodies with ELISA before and at 1 month, 3 months and 12 months after each dosing", and "We measured interferon beta 1a‐neutralising antibodies at baseline and at 24 months with a cytopathic effect inhibition assay" (Page 1832)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

CombiRx 2013

Low

"Safety was assessed by recording all adverse events, serious and nonserious" (Page 329)

Unclear

No information on SAE definition

Comi 2001

Unclear

"The treating physician monitored safety..." (Page 291)

Unclear

Insufficient information on SAEs definition

CONFIRM 2012

Low

"Throughout the course of the study, every effort was made to remain alert to possible adverse events (AEs)" and "Any AE or SAE experienced by the subject was recorded on the CRF, regardless of the severity of the event or its relationship to study treatment" (Pages 66‐7 of Protocol)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

DEFINE 2012

Low

"Study visits were scheduled every 4 weeks for safety assessments, including the monitoring of laboratory values" (Page 1100)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

Etemadifar 2007

Low

"Adverse events, vital signs and blood tests were monitored monthly" (Page 1724)

High

SAEs not reported

EVIDENCE 2007

High

"Adverse events were determined by spontaneous reporting and monthly laboratory testing during the comparative phase" (Page 2031)

Unclear

Insufficient information on SAEs definition

Fazekas 1997

Low

Participants "asked about safety monthly..." (Page 590)

High

SAEs not reported

Fazekas 2008

Unclear

Not reported

Unclear

Insufficient information on SAEs definition

FREEDOMS 2010

Low

"An independent data and safety monitoring board evaluated the safety" and "Study visits, including safety assessments, were scheduled at 2 weeks and 1, 2, 3, 6, 9, 12, 15, 18, 21, and 24 months after randomization" (Page 389)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

FREEDOMS II 2014

Low

"...safety assessments, were scheduled at 2 weeks and 1, 2, 3, 6, 9, 12, 15, 18, 21, and 24 months after randomization" (Appendix, Page 2)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

GALA 2013

Low

"Safety assessments included adverse events (AEs), standard clinical laboratory tests, vital signs, and electrocardiographic (ECG) measurements" (Page 707)

Unclear

No information on SAE definition

Goodkin 1991

High

"Side effect were reported to the treating neurologist every 6 months" (Page 21)

High

SAEs not reported

IFNB MS Group 1993

Low

"Treating neurologist reviewed side effects, laboratory findings for toxicity ..." (Page 656)

High

SAEs not reported

INCOMIN 2002

Low

"Safety assessments included adverse events, vital signs, physical examination, and concomitant medications. Patients underwent haematology and biochemical tests, including liver‐function tests, every 2 weeks for the first 8 weeks, and then every 3 months" (Page 1455)

High

SAEs not reported

Johnson 1995

Low

"The evaluating physician monitored safety every 3 month..." (Page 1270)

Unclear

Insufficient information on SAEs definition

Koch‐Henriksen 2006

Low

"Patients were interviewed about side effects and had routine blood tests including hematology and liver function tests every 3 months and thyroid tests and neutralizing antibodies every 6 months" (Page 1057)

High

SAEs not reported

Lewanska 2002

Unclear

"Laboratory safety examinations were made at the beginning and at the end of the study period" (Page 566)

Unclear

Insufficient information on SAEs definition

MAIN TRIAL

Low

"At scheduled (quarterly) and unscheduled (i.e., at the onset of new symptoms or complications) follow‐up visits the treating neurologist recorded symptoms, blood test results, clinical AEs and their management"

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

Millefiorini 1997

Low

"The safety of the treatment was assessed on the basis of adverse events volunteered by the patient either spontaneously or on questioning and monitoring of the main laboratory parameters" (Page 155)

Unclear

Insufficient information on SAEs definition

MSCRG 1996

Low

"Study visits were scheduled at baseline and every 6 months. Treating physicians reviewed toxicity test results, examined patients, and made all medical decision" (Page 286)

Unclear

Insufficient information on SAEs definition

OWIMS 1999

Unclear

"The treating physician recorded and treated AEs..." (Page 680)

Unclear

Insufficient information on SAEs definition

PRISMS 1998

Unclear

"A “treating” neurologist was responsible for overall medical management of the patient, including treatment of any side‐effects" (Page 1499)

Unclear

Insufficient information on SAEs definition

REGARD 2008

Unclear

"Adverse events (including pregnancy), withdrawals owing to adverse events, serious adverse events, and laboratory results were obtained for safety comparisons" (Page 905)

Unclear

Insufficient information on SAEs definition

SELECT 2013

Low

"Safety parameters were assessed at all visits" (Page 2168)

Unclear

No information on SAE definition

TEMSO 2011

Low

"A treating neurologist at each site was responsible for recording and managing adverse events and monitoring safety assessments" and "Safety was evaluated on the basis of adverse events reported by study participants or investigators. Laboratory tests were performed at the time of screening, at baseline, every 2 weeks for the first 24 weeks, and then every 6 weeks until study completion. Physical and neurologic examinations were performed at week 12 and then every 24 weeks. An abdominal ultrasonographic examination to asses for pancreatic abnormalities was performed before the study and then every 24 weeks, because of previous infrequent reports of pancreatitis associated with leflunomide use" (Pages 1294‐5)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

TENERE 2014

Low

"Safety and tolerability were assessed using AE reporting, vital signs and laboratory assessments. Adverse event reports were collected at randomisation, Weeks 2, 6, 12, 18, 24, 36 and every 12 weeks thereafter. Vital signs were documented at screening, randomisation and every 12 weeks thereafter; clinical laboratory results were assessed throughout the study. Adverse events and vital signs were also recorded during unscheduled relapse visits" (Page 707)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use) [information provided on request by Genzyme]

TOWER 2014

Low

"Safety was assessed through adverse event reporting (upon occurrence), clinical laboratory tests (every 2 weeks until week 24, then every 6 weeks while still on treatment), vital signs (at weeks 2 and 6, then every 6 weeks until week 24, then every 12 weeks while still on treatment), abdominal ultrasonography (at week 24, then every 24 weeks), and electrocardiography (at baseline and end of treatment)" (Page 248)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

TRASFORMS 2010

Low

"An independent data and safety monitoring board evaluated overall safety in the fingolimod phase 3 program" and "Safety assessments were conducted during screening, at baseline, and at months 1, 2, 3, 6, 9, and 12" (Page 404)

Low

Categorisation of SAEs conformed to ICH guidelines (International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use)

Figuras y tablas -
Table 1. Assessment of adverse events monitoring
Table 2. Subgroup analyses: network meta‐analysis estimates for relapse outcome over 24 months for the three best drugs based on moderate to high quality evidence

Intervention

Subgroup analysis by

Diagnostic criteria
RR (95% CI)

Previous treatments

RR (95% CI)

Definition of relapse

RR (95% CI)

Pre‐trial relapse rate

RR (95% CI)

Poser criteria

McDonald criteria

No

Yes

24‐hour definition

48‐hour definition

≥ 1 during the year

before randomisation

≥ 2 during the 2/3 years

before randomisation

Alemtuzumab

0.48 (0.33 to 0.68)

0.46 (0.28 to 0.76)

0.47 (0.27 to 0.79)

0.46 (0.27 to 0.78)

0.63 (0.48 to 0.81)

0.28 (0.16 to 0.49)

Natalizumab

0.56 (0.45 to 0.69)

0.70 (0.56 to 0.88)

0.63 (0.52 to 0.77)

0.68 (0.54 to 0.85)

Fingolimod

0.72 (0.63 to 0.83)

0.72 (0.65 to 0.80)

0.81 (0.67 to 0.97)

0.72 (0.60 to 0.87)

CI: confidence interval; NMA: network meta‐analysis; RR: risk ratio.

Figuras y tablas -
Table 2. Subgroup analyses: network meta‐analysis estimates for relapse outcome over 24 months for the three best drugs based on moderate to high quality evidence
Table 3. Sensitivity analyses: NMA estimates for relapse outcome over 24 months for the three best drugs based on moderate to high quality evidence

Intervention

Sensitivity analysis

Including only trials of low risk of bias
RR (95% CI)

Excluding studies that did not provide complete

and clear reporting of dropout data

RR (95% CI)

Excluding trials with a total sample size of

fewer than 50 randomised participants

RR (95% CI)

Alemtuzumab

0.47 (0.35 to 0.63)

0.46 (0.39 to 0.56)

Natalizumab

0.66 (0.54 to 0.81)

0.56 (0.47 to 0.66)

Fingolimod

0.72 (0.65 to 0.80)

0.72 (0.64 to 0.81)

CI: confidence interval; NMA: network meta‐analysis; RR: risk ratio.

Figuras y tablas -
Table 3. Sensitivity analyses: NMA estimates for relapse outcome over 24 months for the three best drugs based on moderate to high quality evidence
Comparison 1. Treatment benefit within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisons for relapses over 12 months Show forest plot

29

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Interferon beta‐1a (Avonex) versus placebo

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.73, 1.05]

1.2 Interferon beta‐1a (Rebif) versus placebo

2

853

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.66, 1.19]

1.3 Glatiramer acetate versus placebo

4

2416

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.66, 0.95]

1.4 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.47, 0.66]

1.5 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.21, 0.74]

1.6 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.48, 0.82]

1.7 Teriflunomide versus placebo

2

2257

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.78, 0.95]

1.8 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.71, 0.88]

1.9 Pegylated interferon beta‐1a versus placebo

1

1512

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.78, 1.01]

1.10 Daclizumab versus placebo

1

621

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.68, 0.92]

1.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.61, 1.24]

1.12 Immunoglobulins versus placebo

3

219

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.47, 1.36]

1.13 Interferon beta‐1a (Rebif) versus interferon beta‐1a (Avonex)

1

677

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.80, 1.03]

1.14 Glatiramer acetate versus interferon beta‐1b (Betaseron)

1

75

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.48, 1.38]

1.15 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

2

244

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.49, 1.33]

1.16 Fingolimod versus interferon beta‐1a (Avonex)

1

1292

Risk Ratio (M‐H, Random, 95% CI)

0.67 [0.57, 0.79]

1.17 Teriflunomide versus interferon beta‐1a (Rebif)

1

324

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.69, 1.18]

1.18 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

0.96 [0.80, 1.14]

1.19 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.39, 0.55]

2 Comparisons for relapses over 24 months Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Interferon beta‐1b (Betaseron) versus placebo

1

372

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.81, 0.99]

2.2 Interferon beta‐1a (Avonex) versus placebo

2

1198

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.76, 1.04]

2.3 Interferon beta‐1a (Rebif) versus placebo

1

560

Risk Ratio (M‐H, Random, 95% CI)

0.85 [0.77, 0.92]

2.4 Glatiramer acetate versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.75, 0.98]

2.5 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.56 [0.49, 0.64]

2.6 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.27, 0.80]

2.7 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.67, 0.78]

2.8 Teriflunomide versus placebo

1

1088

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.79, 0.98]

2.9 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.81, 0.97]

2.10 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.75, 0.99]

2.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.56, 1.05]

2.12 Immunoglobulins versus placebo

2

190

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.61, 0.90]

2.13 Interferon beta‐1a (Avonex) versus interferon beta‐1b (Betaseron)

1

188

Risk Ratio (M‐H, Random, 95% CI)

1.34 [1.06, 1.71]

2.14 Interferon beta‐1a (Rebif) versus interferon beta‐1b (Betaseron)

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.89, 1.11]

2.15 Glatiramer acetate versus interferon beta‐1b (Betaseron)

2

2319

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.47, 1.38]

2.16 Glatiramer acetate versus interferon beta‐1a (Rebif)

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.78, 1.09]

2.17 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.98, 1.21]

2.18 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.39, 0.65]

2.19 Laquinimod versus interferon beta‐1a (Avonex)

1

881

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.97, 1.32]

2.20 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

0.86 [0.64, 1.16]

3 Comparisons for disability worsening over 24 months Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 Interferon beta‐1b (Betaseron) versus placebo

1

372

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.72, 1.32]

3.2 Interferon beta‐1a (Avonex) versus placebo

2

1198

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.70, 1.09]

3.3 Interferon beta‐1a (Rebif) versus placebo

1

560

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.61, 0.96]

3.4 Glatiramer acetate versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

0.81 [0.61, 1.09]

3.5 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.52, 0.80]

3.6 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

0.20 [0.05, 0.83]

3.7 Fingolimod versus placebo

2

2355

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.76, 0.99]

3.8 Teriflunomide versus placebo

1

1088

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.75, 1.01]

3.9 Dimethyl fumarate versus placebo

2

2307

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.72, 0.90]

3.10 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.73, 0.95]

3.11 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.31, 1.34]

3.12 Immunoglobulins versus placebo

2

190

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.39, 1.24]

3.13 Interferon beta‐1a (Avonex) versus interferon beta‐1b (Betaseron)

1

188

Risk Ratio (M‐H, Random, 95% CI)

2.23 [1.29, 3.83]

3.14 Interferon beta‐1a (Rebif) versus interferon beta‐1b (Betaseron)

1

301

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.78, 1.25]

3.15 Glatiramer acetate versus interferon beta‐1b (Betaseron)

2

2319

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.86, 1.13]

3.16 Glatiramer acetate versus interferon beta‐1a (Rebif)

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.39, 0.85]

3.17 Dimethyl fumarate versus glatiramer acetate

1

1067

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.83, 1.22]

3.18 Alemtuzumab versus interferon beta‐1a (Rebif)

3

1582

Risk Ratio (M‐H, Random, 95% CI)

0.41 [0.32, 0.54]

3.19 Laquinimod versus interferon beta‐1a (Avonex)

1

881

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.85, 1.33]

3.20 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.49, 1.23]

Figuras y tablas -
Comparison 1. Treatment benefit within pairwise comparisons
Comparison 2. Treatment acceptability within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Comparisons for treatment discontinuation due to AEs over 12 months Show forest plot

13

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Interferon beta‐1a (Avonex) 30 µg versus placebo

1

301

Risk Ratio (M‐H, Random, 95% CI)

3.17 [0.67, 15.00]

1.2 Interferon beta‐1a (Rebif) 22 µg versus placebo

1

195

Risk Ratio (M‐H, Random, 95% CI)

3.16 [0.13, 76.54]

1.3 Interferon beta‐1a (Rebif) 44 µg versus placebo

1

198

Risk Ratio (M‐H, Random, 95% CI)

11.22 [0.63, 200.27]

1.4 Glatiramer acetate 20 mg daily versus placebo

1

239

Risk Ratio (M‐H, Random, 95% CI)

1.51 [0.26, 8.89]

1.5 Glatiramer 40 mg three times per week versus placebo

1

1404

Risk Ratio (M‐H, Random, 95% CI)

2.36 [0.99, 5.65]

1.6 Teriflunomide 7 mg versus placebo

1

797

Risk Ratio (M‐H, Random, 95% CI)

2.06 [1.31, 3.24]

1.7 Teriflunomide 14 mg versus placebo

1

761

Risk Ratio (M‐H, Random, 95% CI)

2.51 [1.61, 3.91]

1.8 Pegylated interferon beta‐1a every 4 weeks versus placebo

1

1000

Risk Ratio (M‐H, Random, 95% CI)

2.78 [1.31, 5.89]

1.9 Pegylated interferon beta‐1a every 2 weeks versus placebo

1

1012

Risk Ratio (M‐H, Random, 95% CI)

2.82 [1.34, 5.96]

1.10 Daclizumab 150 mg versus placebo

1

412

Risk Ratio (M‐H, Random, 95% CI)

3.43 [0.72, 16.33]

1.11 Daclizumab 300 mg versus placebo

1

413

Risk Ratio (M‐H, Random, 95% CI)

4.39 [0.96, 20.08]

1.12 Immunoglobulins 0.2 g versus placebo

2

163

Risk Ratio (M‐H, Random, 95% CI)

2.14 [0.23, 19.96]

1.13 Immunoglobulins 0.4 g versus placebo

1

34

Risk Ratio (M‐H, Random, 95% CI)

3.35 [0.15, 76.93]

1.14 Interferon beta‐1a (Rebif) 44 µg versus interferon beta‐1a (Avonex) 30 µg

1

677

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.56, 1.97]

1.15 Fingolimod 0.5 mg versus interferon beta‐1a (Avonex) 30 µg

1

866

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.81, 2.54]

1.16 Fingolimod 1.25 mg versus interferon beta‐1a (Avonex) 30 µg

1

861

Risk Ratio (M‐H, Random, 95% CI)

2.36 [1.40, 3.98]

1.17 Teriflunomide 7 mg versus interferon beta‐1a (Rebif) 44 µg

1

213

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.19, 0.81]

1.18 Teriflunomide 14 mg versus interferon beta‐1a (Rebif) 44 µg

1

215

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

1.19 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

94

Risk Ratio (M‐H, Random, 95% CI)

1.0 [0.21, 4.70]

2 Comparisons for treatment discontinuation due to AEs over 24 months Show forest plot

23

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Interferon beta‐1b (Betaseron) 50 µg versus placebo

1

248

Risk Ratio (M‐H, Random, 95% CI)

4.92 [0.58, 41.51]

2.2 Interferon beta‐1b (Betaseron) 250 µg versus placebo

1

247

Risk Ratio (M‐H, Random, 95% CI)

9.92 [1.29, 76.32]

2.3 Interferon beta‐1a (Avonex) 30 µg versus placebo

1

897

Risk Ratio (M‐H, Random, 95% CI)

1.43 [0.81, 2.54]

2.4 Interferon beta‐1a (Rebif) 22 µg versus placebo

1

376

Risk Ratio (M‐H, Random, 95% CI)

2.31 [0.61, 8.79]

2.5 Interferon beta‐1a (Rebif) 44 µg versus placebo

1

371

Risk Ratio (M‐H, Random, 95% CI)

3.05 [0.84, 11.08]

2.6 Glatiramer acetate 20 mg daily versus placebo

3

1024

Risk Ratio (M‐H, Random, 95% CI)

1.74 [0.49, 6.13]

2.7 Natalizumab versus placebo

1

942

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.93, 2.53]

2.8 Mitoxantrone versus placebo

1

51

Risk Ratio (M‐H, Random, 95% CI)

9.82 [0.57, 168.84]

2.9 Fingolimod 0.5 mg versus placebo

2

1556

Risk Ratio (M‐H, Random, 95% CI)

1.42 [0.89, 2.25]

2.10 Fingolimod 1.25 mg versus placebo

2

1572

Risk Ratio (M‐H, Random, 95% CI)

1.93 [1.48, 2.52]

2.11 Teriflunomide 7 mg versus placebo

1

729

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.77, 1.96]

2.12 Teriflunomide 14 mg versus placebo

1

722

Risk Ratio (M‐H, Random, 95% CI)

1.36 [0.86, 2.15]

2.13 Dimethyl fumarate 480 mg versus placebo

2

1546

Risk Ratio (M‐H, Random, 95% CI)

1.17 [0.91, 1.51]

2.14 Dimethyl fumarate 720 mg versus placebo

2

1534

Risk Ratio (M‐H, Random, 95% CI)

1.20 [0.93, 1.54]

2.15 Laquinimod versus placebo

2

1990

Risk Ratio (M‐H, Random, 95% CI)

1.39 [0.96, 2.00]

2.16 Azathioprine versus placebo

1

59

Risk Ratio (M‐H, Random, 95% CI)

5.8 [0.74, 45.26]

2.17 Immunoglobulins 0.15 to 0.20 g versus placebo

1

150

Risk Ratio (M‐H, Random, 95% CI)

3.0 [0.32, 28.19]

2.18 Interferon beta‐1a (Avonex) 30 µg versus interferon beta‐1b (Betaseron) 250 µg

1

188

Risk Ratio (M‐H, Random, 95% CI)

0.21 [0.02, 1.75]

2.19 Glatiramer acetate 20 mg daily versus interferon beta‐1b (Betaseron) 250 μg

2

1420

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.56, 2.53]

2.20 Glatiramer acetate 20 mg daily versus interferon beta‐1b (Betaseron) 500 μg

1

1347

Risk Ratio (M‐H, Random, 95% CI)

0.79 [0.37, 1.68]

2.21 Glatiramer acetate 20 mg daily versus interferon beta‐1a (Rebif) 44 µg

1

764

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.47, 1.52]

2.22 Dimethyl fumarate 480 mg versus glatiramer acetate 20 mg daily

1

722

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.84]

2.23 Dimethyl fumarate 720 mg versus glatiramer acetate 20 mg daily

1

705

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.80, 1.85]

2.24 Alemtuzumab 12 mg versus interferon beta‐1a (Rebif) 44 µg

3

1472

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.22, 0.68]

2.25 Alemtuzumab 24 mg versus interferon beta‐1a (Rebif) 44 µg

2

625

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.10, 1.09]

2.26 Laquinimod versus interferon beta‐1a (Avonex) 30 µg

1

881

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.49, 1.45]

2.27 Azathioprine versus interferons beta (Avonex, Rebif or Betaseron)

1

150

Risk Ratio (M‐H, Random, 95% CI)

2.21 [0.90, 5.45]

Figuras y tablas -
Comparison 2. Treatment acceptability within pairwise comparisons
Comparison 3. Treatment safety against placebo within pairwise comparisons

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Serious adverse events Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Interferons beta (Avonex, Rebif or Betaseron) versus placebo

3

870

Risk Ratio (M‐H, Random, 95% CI)

1.26 [0.67, 2.37]

1.2 Glatiramer acetate versus placebo

2

490

Risk Ratio (M‐H, Random, 95% CI)

1.87 [0.73, 4.74]

1.3 Natalizumab versus placebo

1

939

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.81, 1.73]

1.4 Fingolimod versus placebo

2

1572

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.72, 1.30]

1.5 Teriflunomide versus placebo

1

718

Risk Ratio (M‐H, Random, 95% CI)

1.26 [0.87, 1.83]

1.6 Dimethyl fumarate versus placebo

2

1531

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.76, 1.55]

1.7 Pegylated interferon beta‐1a versus placebo

1

1012

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.57, 1.68]

1.8 Daclizumab versus placebo

1

413

Risk Ratio (M‐H, Random, 95% CI)

1.55 [0.77, 3.10]

1.9 Laquinimod versus placebo

2

1988

Risk Ratio (M‐H, Random, 95% CI)

0.97 [0.67, 1.41]

1.10 Immunoglobulins versus placebo

1

83

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.11, 3.70]

Figuras y tablas -
Comparison 3. Treatment safety against placebo within pairwise comparisons