Scolaris Content Display Scolaris Content Display

Tratamiento con fármacos modificadores de la enfermedad para los pacientes con un primer episodio clínico indicativo de esclerosis múltiple

Collapse all Expand all

References

References to studies included in this review

Achiron 2004 {published data only}

Achiron A, Kishner I, Sarova‐Pinhas I, Raz H, Faibel M, Stern Y, et al. Intravenous immunoglobulin treatment following the first demyelinating event suggestive of multiple sclerosis: a randomized, double‐blind, placebo‐controlled trial. Archives of Neurology 2004;61(10):1515‐20. [PUBMED: 15477504]CENTRAL

ACISS 2010 {published data only}

Fazekas F, Baumhackl U, Berger T, Deisenhammer F, Fuchs S, Kristoferitsch W, et al. Decision‐making for and impact of early immunomodulatory treatment: the Austrian Clinically Isolated Syndrome Study (ACISS). European Journal of Neurology 2010;17(6):852‐60. [PUBMED: 20100231]CENTRAL

BENEFIT 2006 {published data only}

Kappos L, Polman CH, Freedman MS, Edan G, Hartung HP, Miller DH, et al. Treatment with interferon beta‐1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology 2006;67(7):1242‐9. [PUBMED: 16914693]CENTRAL

BENEFIT 2007 (3 years FU) {published data only}

Kappos L, Freedman MS, Polman CH, Edan G, Hartung HP, Miller DH, et al. Effect of early versus delayed interferon beta‐1b treatment on disability after a first clinical event suggestive of multiple sclerosis: a 3‐year follow‐up analysis of the BENEFIT study. Lancet 2007;370(9585):389‐97. [PUBMED: 17679016]CENTRAL

BENEFIT 2009 (5 years FU) {published data only}

Kappos L, Freedman MS, Polman CH, Edan G, Hartung HP, Miller DH, et al. Long‐term effect of early treatment with interferon beta‐1bafter a first clinical event suggestive of multiple sclerosis: 5‐year active treatment extension of the phase 3 BENEFIT trial. Lancet Neurology 2009;8(11):987–97. [PUBMED: 19748319]CENTRAL

BENEFIT 2014 (8.7 years FU) {published data only}

Edan G, Kappos L, Montalbán X, Polman C, Freedman M, Hartung H, et al. Long‐term impact of interferon beta‐1b in patients with CIS: 8‐year follow‐up of BENEFIT. Journal of Neurology, Neurosurgery, and Psychiatry 2014;85:1183–9. [PUBMED: 24218527]CENTRAL

BENEFIT 2016 (11 years FU) {published data only}

Kappos L, Edan G, Freedman MS, Montalbán X, Hartung HP, Hemmer B, et al. The 11‐year long‐term follow‐up study from the randomized BENEFIT CIS trial. Neurology 2016;87(10):978‐87. [PUBMED: 27511182]CENTRAL

CHAMPS 2000 {published data only}

CHAMPS Study Group. Baseline MRI characteristics of patients at high risk for multiple sclerosis: results from the CHAMPS trial. Controlled High‐Risk Subjects Avonex Multiple Sclerosis Prevention Study. Multiple Sclerosis 2002;8(4):330‐8. [PUBMED: 12166504]CENTRAL
Jacobs LD, Beck RW, Simon JH, Kinkel RP, Brownscheidle CM, Murray TJ, et al. Intramuscular interferon beta‐1a therapy initiated during a first demyelinating event in multiple sclerosis. CHAMPS Study Group. New England Journal of Medicine 2000;343(13):898‐904. [PUBMED: 11006365]CENTRAL

CHAMPS 2006 (5 years FU) {published data only}

Kinkel RP, Kollman C, O'Connor P, Murray TJ, Simon J, Arnold D, et al. IM interferon beta‐1a delays definite multiple sclerosis 5 years after a first demyelinating event. Neurology 2006;66(5):678‐84. [PUBMED: 16436649]CENTRAL

CHAMPS 2012 (10 years FU) {published data only}

Kinkel RP, Dontchev M, Kollman C, Skaramagas TT, O'Connor PW, Simon JH, et al. Association between immediate initiation of intramuscular interferon beta‐1a at the time of a clinically isolated syndrome and long‐term outcomes: a 10‐year follow‐up of the Controlled High‐Risk Avonex Multiple Sclerosis Prevention Study in Ongoing Neurological Surveillance. Archives of Neurology 2012;69(2):183‐90. [PUBMED: 21987393]CENTRAL
Simon JH, Kinkel RP, Kollman C, O'Connor P, Fisher E, You X, et al. Ten‐year follow‐up of the 'minimal MRI lesion' subgroup from the original CHAMPS Multiple Sclerosis Prevention Trial. Multiple Sclerosis 2015;21(4):415‐22. [PUBMED: 25344370]CENTRAL

ETOMS 2001 {published data only}

Comi G, Filippi M, Barkhof F, Durelli L, Edan G, Fernández O, et al. Effect of early interferon treatment on conversion to definite multiple sclerosis: a randomised study. Lancet 2001;357(9268):1576‐82. [PUBMED: 11377645]CENTRAL

GERONIMUS 2013 {published data only}

D'Alessandro R, Vignatelli L, Lugaresi A, Baldin E, Granella F, Tola MR, et al. Risk of multiple sclerosis following clinically isolated syndrome: a 4‐year prospective study. Journal of Neurology 2013;260(6):1583‐93. [PUBMED: 23377434]CENTRAL

Motamed 2007 {published data only}

Motamed MR, Najimi N, Fereshtehnejad SM. The effect of interferon‐beta‐1a on relapses and progression of disability in patients with clinically isolated syndromes (CIS) suggestive of multiple sclerosis. Clinical Neurology and Neurosurgery 2007;109(4):344‐9. [PUBMED: 17300863]CENTRAL

MSBASIS 2016 {published data only}

Spelman T, Meyniel C, Rojas JI, Lugaresi A, Izquierdo G, Grand'Maison F, et al. Quantifying risk of early relapse in patients with first demyelinating events: prediction in clinical practice. Multiple Sclerosis Journal 2016;1:1‐12. [PUBMED: 27885062]CENTRAL

ORACLE 2014 {published data only}

Leist TP, Comi G, Cree BA, Coyle PK, Freedman MS, Hartung HP, et al. Effect of oral cladribine on time to conversion to clinically definite multiple sclerosis in patients with a first demyelinating event (ORACLE MS): a phase 3 randomised trial. Lancet Neurology 2014;13(3):257‐67. [PUBMED: 24502830]CENTRAL

Pakdaman 2007 {published data only}

Pakdaman H, Sahraian MA, Fallah A, Pakdaman R, Ghareghozli K, Ghafarpour M, et al. Effect of early interferon beta‐1a therapy on conversion to multiple sclerosis in Iranian patients with a first demyelinating event. Acta Neurologica Scandinavica 2007;115(6):429‐31. [PUBMED: 17511854]CENTRAL

PRECISE 2009 {published data only}

Comi G, Martinelli V, Rodegher M, Moiola L, Bajenaru O, Carra A, et al. Effect of glatiramer acetate on conversion to clinically definite multiple sclerosis in patients with clinically isolated syndrome (PreCISe study): a randomised, double‐blind, placebo‐controlled trial. Lancet 2009;374(9700):1503‐11. [PUBMED: 19815268]CENTRAL

PRECISE 2013 (5 years FU) {published data only}

Comi G, Martinelli V, Rodegher M, Moiola L, Leocani L, Bajenaru O, et al. Effects of early treatment with glatiramer acetate in patients with clinically isolated syndrome. Multiple Sclerosis 2013;19(8):1074‐83. [PUBMED: 23234810]CENTRAL

REFLEX 2012 {published data only}

Comi G, De Stefano N, Freedman MS, Barkhof F, Polman CH, Uitdehaag BM, et al. Comparison of two dosing frequencies of subcutaneous interferon beta‐1a in patients with a first clinical demyelinating event suggestive of multiple sclerosis (REFLEX): a phase 3 randomised controlled trial. Lancet Neurology 2012;11(1):33‐41. [PUBMED: 22146409]CENTRAL

REFLEX 2016 (3 and 5 years FU) {published data only}

Comi G, De Stefano N, Freedman M, Barkhof F, Uitdehaag B, de Vos M, et al. Subcutaneous interferon β‐1a in the treatment of clinically isolated syndromes: 3‐year and 5‐year results of the phase III dosing frequency‐blind multicentre REFLEXION study. Journal of Neurology, Neurosurgery, and Psychiatry 2016;88(4):285‐94. [PUBMED: 28039317]CENTRAL

Tintore 2015 {published data only}

Tintore M, Rovira À, Río J, Otero‐Romero S, Arrambide G, Tur C, et al. Defining high, medium and low impact prognostic factors for developing multiple sclerosis. Brain 2015;138:1863‐74. [PUBMED: 25902415]CENTRAL

TOPIC 2014 {published data only}

Miller AE, Wolinsky JS, Kappos L, Comi G, Freedman MS, Olsson TP, et al. Oral teriflunomide for patients with a first clinical episode suggestive of multiple sclerosis (TOPIC): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Neurology 2014;13(10):977‐86. [PUBMED: 25192851]CENTRAL

References to studies excluded from this review

BENEFIT 2007 {published data only}

Barkhof F, Polman CH, Radue EW, Kappos L, Freedman MS, Edan G, et al. Magnetic resonance imaging effects of interferon beta‐1b in the BENEFIT study: integrated 2‐year results. Archives of Neurology 2007;64(9):1292‐8. [PUBMED: 17846268]CENTRAL

BENEFIT 2008 {published data only}

Polman C, Kappos L, Freedman MS, Edan G, Hartung HP, Miller DH, et al. Subgroups of the BENEFIT study: risk of developing MS and treatment effect of interferon beta‐1b. Journal of Neurology 2008;255(4):480‐7. [PUBMED: 18004635]CENTRAL

BENEFIT 2011 {published data only}

Hartung HP, Freedman MS, Polman CH, Edan G, Kappos L, Miller DH, et al. Interferon β‐1b‐neutralizing antibodies 5 years after clinically isolated syndrome. Neurology 2011;77(9):835‐43. [PUBMED: 21849647]CENTRAL

BENEFIT 2012 {published data only}

Penner IK, Stemper B, Calabrese P, Freedman MS, Polman CH, Edan G, et al. Effects of interferon beta‐1b on cognitive performance in patients with a first event suggestive of multiple sclerosis. Multiple Sclerosis 2012;18(10):1466‐71. [PUBMED: 22492127]CENTRAL

BENEFIT 2014a {published data only}

Ascherio A, Munger KL, White R, Köchert K, Simon KC, Polman CH, et al. Vitamin D as an early predictor of multiple sclerosis activity and progression. JAMA 2014;71(3):306‐14. [PUBMED: 24445558]CENTRAL

BENEFIT 2014b {published data only}

Nagtegaal GJ, Pohl C, Wattjes MP, Hulst HE, Freedman MS, Hartung HP, et al. Interferon beta‐1b reduces black holes in a randomised trial of clinically isolated syndrome. Multiple Sclerosis 2014;20(2):234‐42. [PUBMED: 23842212]CENTRAL

CHAMPIONS 2015 {published data only}

Simon JH, Kinkel RP, Kollman C, O'Connor P, Fisher E, You X, et al. Ten‐year follow‐up of the 'minimal MRI lesion' subgroup from the original CHAMPS Multiple Sclerosis Prevention Trial. Multiple Sclerosis 2015;21(4):415‐22. [PUBMED: 25344370 ]CENTRAL

CHAMPS 2001 {published data only}

CHAMPS Study Group. Interferon ‐1a for optic neuritis patients at high risk for multiple sclerosis. American Journal of Ophthalmology 2001;132(4):463‐71. [PUBMED: 11589865]CENTRAL

CHAMPS 2002a {published data only}

CHAMPS Study Group. Predictors of short‐term disease activity following a first clinical demyelinating event: analysis of the CHAMPS placebo group. Multiple Sclerosis 2002;8(5):405‐9. [PUBMED: 12356207]CENTRAL

CHAMPS 2002b {published data only}

Beck RW, Chandler DL, Cole SR, Simon JH, Jacobs LD, Kinkel RP, et al. Interferon beta‐1a for early multiple sclerosis: CHAMPS trial subgroup analyses. Annals of Neurology 2002;51(4):481‐90. [PUBMED: 11921054]CENTRAL

CHAMPS 2002c {published data only}

CHAMPS Study Group. Baseline MRI characteristics of patients at high risk for multiple sclerosis: results from the CHAMPS trial. Multiple Sclerosis 2002;8(4):330‐8. [PUBMED: 12166504]CENTRAL

CHAMPS 2003 {published data only}

O'Connor P. The effects of intramuscular interferon beta‐1a in patients at high risk for development of multiple sclerosis: a post hoc analysis of data from CHAMPS. Clinical Therapeutics 2003;25(11):2865‐74. [PUBMED: 14693310]CENTRAL

CHAMPS 2009 {published data only}

O'Connor P, Kinkel RP, Kremenchutzky M. Efficacy of intramuscular interferon beta‐1a in patients with clinically isolated syndrome: analysis of subgroups based on new risk criteria. Multiple sclerosis 2009;15(6):728‐34. [PUBMED: 19482863]CENTRAL

Curkendall 2011 {published data only}

Curkendall SM, Wang C, Johnson BH, Cao Z, Preblick R, Torres AM, et al. Potential health care cost savings associated with early treatment of multiple sclerosis using disease‐modifying therapy. Clinical Therapeutics 2011;33(7):914‐25. [PUBMED: 21684600]CENTRAL

ETOMS 2003 {published data only}

Barkhof F, Rocca M, Francis G, Van Waesberghe JH, Uitdehaag BM, Hommes OR, et al. Validation of diagnostic magnetic resonance imaging criteria for multiple sclerosis and response to interferon beta1a. Annals of Neurology 2003;53(6):718‐24. [PUBMED: 12783417]CENTRAL

Filippi 2004 {published data only}

Filippi M, Rovaris M, Inglese M, Barkhof F, De Stefano N, Smith S, et al. Interferon beta‐1a for brain tissue loss in patients at presentation with syndromes suggestive of multiple sclerosis: a randomised, double‐blind, placebo‐controlled trial. Lancet 2004;364(9444):1489‐96. [PUBMED: 15500893]CENTRAL

Kuhle 2015 {published data only}

Kuhle J, Disanto G, Dobson R, Adiutori R, Bianchi L, Topping J, et al. Conversion from clinically isolated syndrome to multiple sclerosis: a large multicentre study. Multiple Sclerosis 2015;21(8):1013‐24. [PUBMED: 25680984]CENTRAL

Lazzaro 2009 {published data only}

Lazzaro C, Bianchi C, Peracino L, Zacchetti P, Uccelli A. Economic evaluation of treating clinically isolated syndrome and subsequent multiple sclerosis with interferon beta‐1b. Neurological Sciences 2009;30(1):21‐31. [PUBMED: 19169625]CENTRAL

Meyniel 2012 {published data only}

Meyniel C, Spelman T, Jokubaitis VG, Trojano M, Izquierdo G, Grand'Maison F, et al. Country, sex, EDSS change and therapy choice independently predict treatment discontinuation in multiple sclerosis and clinically isolated syndrome. PLoS One 2012;7(6):e38661. [PUBMED: 22768046]CENTRAL

Moraal 2009 {published data only}

Moraal B, Pohl C, Uitdehaag BM, Polman CH, Edan G, Freedman MS, et al. Magnetic resonance imaging predictors of conversion to multiple sclerosis in the BENEFIT study. Archives of Neurology 2009;66(11):1345‐52. [PUBMED: 19901165]CENTRAL

Mowry 2009 {published data only}

Mowry EM, Pesic M, Grimes B, Deen SR, Bacchetti P, Waubant E. Clinical predictors of early second event in patients with clinically isolated syndrome. Journal of Neurology 2009;256(7):1061‐6. [PUBMED: 19252775]CENTRAL

MSBASIS 2015 {published data only}

Jokubaitis VG, Spelman T, Kalincik T, Izquierdo G, Grand'Maison F, Duquette P, et al. Predictors of disability worsening in clinically isolated syndrome. Annals of Clinical and Translational Neurology 2015;2(5):479‐91. [PUBMED: 26000321]CENTRAL

REFLEX 2014a {published data only}

Freedman MS, De Stefano N, Barkhof F, Polman CH, Comi G, Uitdehaag BM, et al. Patient subgroup analyses of the treatment effect of subcutaneous interferon β‐1a on development of multiple sclerosis in the randomized controlled REFLEX study. Journal of Neurology 2014;261(3):490‐9. [PUBMED: 24413638]CENTRAL

REFLEX 2014b {published data only}

De Stefano N, Comi G, Kappos L, Freedman MS, Polman CH, Uitdehaag BM, et al. Efficacy of subcutaneous interferon β‐1a on MRI outcomes in a randomised controlled trial of patients with clinically isolated syndromes. Journal of Neurology Neurosurgery and Psychiatry 2914;85(6):647‐53. [PUBMED: 24292999]CENTRAL

SWISS COHORT STUDY 2013 {published data only}

Gobbi C, Zecca C, Linnebank M, Müller S, You X, Meier R, et al. Swiss analysis of multiple sclerosis: a multicenter, non‐interventional, retrospective cohort study of disease‐modifying therapies. European Neurology 2013;70(1‐2):35‐41. [PUBMED: 23689307]CENTRAL

SWISS COHORT STUDY 2016 {published data only}

Disanto G, Benkert P, Lorscheider J, Mueller S, Vehoff J, Zecca C, et al. The Swiss Multiple Sclerosis Cohort‐Study (SMSC): a prospective Swiss wide investigation of key phases in disease evolution and new treatment options. PLoS One 2016;11(3):e0152347. [PUBMED: 27032105]CENTRAL

References to ongoing studies

NCT01013350 {unpublished data only}

NCT01013350. Prospective observational long‐term safety registry of multiple sclerosis patients who have participated in cladribine clinical trials (PREMIERE). (first received 11 November 2009). CENTRAL

NCT01371071 {published and unpublished data}

NCT01371071. Cohort study of clinically isolated syndrome and early multiple sclerosis (CIS‐COHORT) [Clinically isolated syndrome and newly diagnosed multiple sclerosis: diagnostic, prognostic and therapy ‐ response markers ‐ a Prospective Observational Study (Berlin CIS‐COHORT)]. clinicaltrials.gov/show/NCT01371071 (first received 8 June 2011). CENTRAL

Additional references

Active Biotech 2014

Active Biotech. Teva and Active Biotech remain committed to the development of NERVENTRA® (laquinimod) for multiple sclerosis following the negative opinion from the EMA's CHMP. www.activebiotech.com/press‐releases‐1?pressurl=http://cws.huginonline.com/A/1002/PR/201401/1756936.xml. (accessed 05 April 2017).

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. [PUBMED: 24934599]

Broadley 2014

Broadley SA, Barnett MH, Boggild M, Brew BJ, Butzkueven H, Heard R, et al. Therapeutic approaches to disease modifying therapy for multiple sclerosis in adults: an Australian and New Zealand perspective: part 3 treatment practicalities and recommendations. MS Neurology Group of the Australian and New Zealand Association of Neurologists. Journal of Clinical Neuroscience 2014;21(11):1857‐65. [PUBMED: 24993136]

Chun 2010

Chun J, Hartung HP. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clinical Neuropharmacology 2010;33(2):91‐101. [PUBMED: 20061941]

Claussen 2012

Claussen MC, Korn T. Immune mechanisms of new therapeutic strategies in MS: teriflunomide. Clinical Immunology 2012;142:49‐56. [PUBMED: 21367665]

Clerico 2008

Clerico M, Faggiano F, Palace J, Rice G, Tintorè M, Durelli L. Recombinant interferon beta or glatiramer acetate for delaying conversion of the first demyelinating event to multiple sclerosis. Cochrane Database of Systematic Reviews 2008, Issue 2. [DOI: 10.1002/14651858.CD005278.pub3]

Colombo 2014

Colombo C, Mosconi P, Confalonieri P, Baroni I, Traversa S, Hill SJ, et al. Web search behavior and information needs of people with multiple sclerosis: focus group study and analysis of online postings. Interactive Journal of Medical Research 2014;3(3):e12. [PUBMED: 25093374]

Colombo 2016

Colombo C, Filippini G, Synnot A, Hill S, Guglielmino R, Traversa S, et al. Development and assessment of a website presenting evidence‐based information for people with multiple sclerosis: the IN‐DEEP project. BMC Neurology 2016;16(1):30. [PUBMED: 26934873]

EMA 2006

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Tysabri. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000603/wapp/Post‐authorisation/human_wapp_000166.jsp&mid=WC0b01ac058001d128 (accessed 05 April 2017).

EMA 2011

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Gilenya. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002202/human_med_001433.jsp&mid=WC0b01ac058001d124 (accessed 05 April 2017).

EMA 2013a

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Aubagio. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002514/human_med_001645.jsp&mid=WC0b01ac058001d124 (accessed 05 April 2017).

EMA 2013b

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Lemtrada. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/003718/human_med_001678.jsp&mid=WC0b01ac058001d124 (accessed 05 April 2017).

EMA 2014a

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Tecfidera. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002601/human_med_001657.jsp&mid=WC0b01ac058001d124 (accessed 05 April 2017).

EMA 2014b

European Medicines Agency. Committee for proprietary medicinal products European public assessment report: Plegridy. www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002827/human_med_001782.jsp&mid=WC0b01ac058001d124 (accessed 05 April 2017).

EMA 2014c

European Medicines Agency. Refusal of the marketing authorisation for Nerventra (laquinimod). www.ema.europa.eu/docs/en_GB/document_library/Summary_of_opinion_‐_Initial_authorisation/human/002546/WC500160120.pdf (accessed 05 April 2017).

EMA 2015a

European Medicines Agency. Guideline on clinical investigation of medicinal products for the treatment of multiple sclerosis. Committee for Medicinal Products for Human Use (CHMP). EMA/CHMP/771815/2011, Rev. 2. www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2015/03/WC500185161.pdf (accessed 05 April 2017).

EMA 2015b

European Medicines Agency. Refusal of the marketing authorization for Movectro (cladribine): outcome of re‐examination. www.ema.europa.eu/docs/en_GB/document_library/Summary_of_opinion_‐_Initial_authorisation/human/001197/WC500101072.pdf (accessed 05 April 2017).

EMA 2016

EMA. Marketing authorisation for the medicinal product Zinbryta. www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Product_Information/human/003862/WC500210598.pdf (accessed 05 April 2017).

Faria 2015

Faria R, Hernandez Alava M, Manca A, Wailoo AJ. NICE DSU technical support document 17: the use of observational data to inform estimates of treatment effectiveness in technology appraisal: methods for comparative individual patient data. www.nicedsu.org.uk/TSD17%20‐%20DSU%20Observational%20data%20FINAL.pdf (accessed 05 April 2017).

FDA 2000

U.S. Food, Drug Administration. Mitoxantrone (Novantrone) Product Approval Information ‐ Licensing Action 2000. www.accessdata.fda.gov/drugsatfda_docs/nda/2000/21120.pdf_Novantrone_Approv.pdf (accessed 05 April 2017).

FDA 2006

U.S. Food, Drug Administration. FDA approves resumed marketing of Tysabri under a special distribution program. www.fda.gov/ohrms/dockets/ac/06/briefing/2006‐4208b1_02_01biogenbriefingmaterial.pdf (accessed 05 April 2017).

FDA 2010

U.S. Food, Drug Administration. Gilenya (Fingolimod) Product Approval Information 2010. www.accessdata.fda.gov/drugsatfda_docs/appletter/2010/022527s000ltr.pdf (accessed 05 April 2017).

FDA 2012

U.S. Food, Drug Administration. Aubagio (Teriflunomide) Product Approval Information 2012. www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/202992Orig1s000ltr.pdf (accessed 05 April 2017).

FDA 2012a

U.S. Food, Drug Administration. Avonex® (interferon beta‐1a) intramuscular injection. Prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2012/103628s5191lbl.pdf (accessed 05 April 2017).

FDA 2012b

U.S. Food, Drug Administration. Betaseron® (interferon beta‐1b). Prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2012/103471s5063s5067s5079s5088s5120s5124s5136s5138lbl.pdf (accessed 05 April 2017).

FDA 2013

U.S. Food, Drug Administration. Copaxone® (glatiramer acetate injection) solution for subcutaneous injection. www.accessdata.fda.gov/drugsatfda_docs/label/2013/020622s087lbl.pdf (accessed 05 April 2017).

FDA 2014a

U.S. Food, Drug Administration. Alemtuzumab (Lemtrada) Product Approval Information. Licensing action 2014. www.accessdata.fda.gov/drugsatfda_docs/label/2014/103948s5139lbl.pdf (accessed 05 April 2017).

FDA 2014b

U.S. Food, Drug Administration. Peginterferon beta‐1a (Plegridy) Product Approval Information. Licensing action 2014. www.accessdata.fda.gov/drugsatfda_docs/label/2014/125499lbl.pdf (accessed 05 April 2017).

FDA 2016

U.S. Food, Drug Administration. Zinbryta™ (daclizumab) injection, for subcutaneous use. Prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2016/761029s000lbl.pdf (accessed 13/04/2017).

FDA 2017

U.S. Food, Drug Administration. Ocrevus™ (ocrelizumab) injection, for intravenous use. Prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2017/761053lbl.pdf (accessed 13/04/2017).

Filippi 2016

Filippi M, Rocca MA, Ciccarelli O, De Stefano N, Evangelou N, Kappos L, et al. MRI criteria for the diagnosis of multiple sclerosis: MAGNIMS consensus guidelines. Lancet Neurology 2016;15(3):292‐303. [DOI: 10.1016/S1474‐4422(15)00393‐2]

Fox 2004

Fox E. Mechanism of action of mitoxantrone. Neurology 2004;12(12 Sppl 6):15‐8. [PUBMED: 15623664]

Freedman 2014

Freedman MS, Comi G, De Stefano N, Barkhof F, Polman CH, Uitdehaag BM, et al. Moving toward earlier treatment of multiple sclerosis: findings from a decade of clinical trials and implications for clinical practice. Multiple Sclerosis and Related Disorders 2014;3(2):147‐55. [PUBMED: 25878002]

Frischer 2009

Frischer JM, Bramow S, Dal‐Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain 2009;132:1175‐89. [PUBMED: 19339255]

Goodin 2002

Goodin DS, Frohman EM, Garmany GP, Halper J, Likosky WH, Lublin FD, et al. 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. [PUBMED: 11805241]

GRADE Working Group 2004

GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490‐4. [PUBMED: 15205295]

Higgins 2003

Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60. [PUBMED: 12958120]

Higgins 2008

Higgins JP, White IR, Wood AM. Imputation methods for missing outcome data in meta‐analysis of clinical trials. Clinical Trials 2008;5(3):225–39. [PUBMED: 18559412]

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. 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 handbook.cochrane.org.

Hill 2012

Hill S, Filippini G, Synnot A, Summers M, Beecher D, Colombo C, et al. Presenting evidence‐based health information for people with multiple sclerosis: the IN‐DEEP project protocol. BMC Medical Informatics and Decision Making 2012;12:20. [PUBMED: 22424304]

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(6):798‐808. [PUBMED: 21680782]

Jansen 2013

Jansen JP, Naci H. Is network meta‐analysis as valid as standard pairwise meta‐analysis? It all depends on the distribution of effect modifiers. BMC Medicine 2013;4(11):1‐8. [PUBMED: 23826681]

Kappos 2011

Kappos L, Calabresi PA, O'Connor P, Bar‐Or A, Barkhof F, Yin M, et al. Ocrelizumab in relapsing‐remitting multiple sclerosis: a phase 2, randomised, placebo‐controlled, multicentre trial. Lancet 2011;378(9805):1779‐87. [PUBMED: 22047971]

Kieseier 2011

Kieseier BC. The mechanism of action of interferon‐β in relapsing multiple sclerosis. CNS Drugs 2011;25(6):491‐502. [PUBMED: 21649449]

Kurtzke 1983

Kurtzke J. Rating neurologic impairment in multiple sclerosis: an Expanded Disability Status Scale (EDSS). Neurology 1983;33(11):1444‐52. [PUBMED: 6685237]

Leist 2011

Leist TP, Weissert R. Cladribine: mode of action and implications for treatment of multiple sclerosis. Clinical Neuropharmacology 2011;34(1):28‐35. [PUBMED: 21242742]

Linker 2011

Linker RA, Lee DH, Ryan S, Van Dam AM, Conrad R, Bista P, et al. Fumaric acid esters exert neuroprotective effects in neuroinflammation via activation of the Nrf2 antioxidant pathway. Brain 2011;134(3):678‐92. [PUBMED: 21354971]

Lycke 2015

Lycke J. Monoclonal antibody therapies for the treatment of relapsing‐remitting multiple sclerosis: differentiating mechanisms and clinical outcomes. Therapeutic Advances in Neurological Disorders 2015;8(6):274‐93. [PUBMED: 26600872]

Millard 2011

Millard M, Odde S, Neamati N. Integrin targeted therapeutics. Theranostics 2011;17(1):154‐88. [PUBMED: 21547158]

Murphy 2010

Murphy JA, Harris JA, Crannage AJ. Potential short‐term use of oral cladribine in treatment of relapsing‐remitting multiple sclerosis. Neuropsychiatric Disease and Treatment 2010;5(6):619‐25. [PUBMED: 20957121]

Naismith 2010

Naismith R, Piccio L, Lyons J, Lauber J, Tutlam N, Parks B, et al. Rituximab add‐on therapy for breakthrough relapsing multiple sclerosis: a 52‐week phase II trial. Neurology 2010;74(23):1860–7. [PUBMED: 20530322]

NHS England 2014

NHS England Clinical Reference Group for Neurosciences. Clinical commissioning policy: disease modifying therapies for patients with multiple sclerosis (MS). www.england.nhs.uk/wp‐content/uploads/2013/10/d04‐p‐b.pdf (accessed 13 April 2016).

Oh 2013

Oh J, O’Connor PW. An update of teriflunomide for treatment of multiple sclerosis. Therapeutics and Clinical Risk Management 2013;9:177‐90. [PUBMED: 23761970]

Pakpoor 2015

Pakpoor J, Disanto G, Altmann DR, Pavitt S, Turner BP, Marta M, et al. No evidence for higher risk of cancer in patients with multiple sclerosis taking cladribine. Neurology Neuroimmunology & Neuroinflammation 2015;2(6):e158. [PUBMED: 26468472]

Palace 2013

Palace J. Partnership and consent in MS treatment choice. Journal of the Neurological Sciences 2013;335:5‐8. [PUBMED: 24090756]

Polman 2011

Polman C, Reingold S, Banwell B, Clanet M, Cohen J, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Annals of Neurology 2011;69(2):292‐302. [PUBMED: 21387374]

Poser 1983

Poser CM, Paty DW, Scheinberg L, McDonald WI, Davis FA, Ebers GC, et al. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Annals of Neurology 1983;13:227–31. [PUBMED: 6847134]

RevMan 2017 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2017.

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. [PUBMED: 26062083]

Scalfari 2013

Scalfari A, Neuhaus A, Daumer M, Deluca G, Muraro P, Ebers G. Early relapses, onset of progression, and late outcome in multiple sclerosis. JAMA Neurology 2013;70(2):214‐22. [PUBMED: 23407713]

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(3):163–74. [PUBMED: 12706402]

Schmitz 2013

Schmitz S, Adams R, Walsh C. Incorporating data from various trial designs into a mixed treatment comparison model. Statistics in Medicine 2013;32(17):2935‐49. [PUBMED: 23440610]

Schunemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, et al. Chapter 12: Interpreting results and drawing conclusions. 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 handbook.cochrane.org.

Scolding 2015

Scolding N, Barnes D, Cader S, Chataway J, Chaudhuri A, Coles A, et al. Association of British Neurologists: revised (2015) guidelines for prescribing disease‐modifying treatments in multiple sclerosis. Practical Neurology 2015;15(4):273‐9. [PUBMED: 26101071]

Singh 2011

Singh JA, Wells GA, Christensen R, Tanjong Ghogomu E, Maxwell L, MacDonald JK, et al. Adverse effects of biologics: a network meta‐analysis and Cochrane overview. Cochrane Database of Systematic Reviews 2011, Issue 2. [DOI: 10.1002/14651858.CD008794]

Smith 2010

Smith B, Carson S, Fu R, McDonagh M, Dana T, Chan BKS, et al. Drug Class Review: Disease‐modifying Drugs for Multiple Sclerosis: Final Update 1 Report. 2010 Aug. Portland (OR): Oregon Health & Science University, 2010. [PUBMED: 21348046 ]

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. [PUBMED: 10369303]

Sterne 2016

Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, et al. ROBINS‐I: a tool for assessing risk of bias in non‐randomised studies of interventions. BMJ (Clinical Research Ed.)2016; Vol. 355:i4919. [PUBMED: 27733354]

Synnot 2014

Synnot AJ, Hill SJ, Garner KA, Summers MP, Filippini G, Osborne RH, et al. Online health information seeking: how people with multiple sclerosis find, assess and integrate treatment information to manage their health. Health Expectations 2016;19(3):727‐37. [PUBMED: 25165024]

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. The Journal of Clinical Investigation 2003;111:1133–45. [PUBMED: 12697733]

Tramacere 2015

Tramacere I, Del Giovane C, Salanti G, D'Amico R, Filippini G. Immunomodulators and immunosuppressants for relapsing‐remitting multiple sclerosis: a network meta‐analysis. Cochrane Database of Systematic Reviews 2015, Issue 9. [DOI: 10.1002/14651858.CD011381]

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. [PUBMED: 24731945]

Verde 2015

Verde PE, Ohmann C. Combining randomized and non‐randomized evidence in clinical research: a review of methods and applications. Research Synthesis Methods 2015;6(1):45‐62. [PUBMED: 26035469]

White 2008

White IR, Higgins JPT, Wood AM. Allowing for uncertainty due to missing data in meta‐analysis. 1. Two‐stage methods. Statistics in Medicine 2008;27(5):711‐27. [PUBMED: 17703496]

Wilms 2010

Wilms H, Sievers J, Rickert U, Rostami‐Yazdi M, Mrowietz U, Lucius R. Dimethyl fumarate 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):2‐8. [PUBMED: 20482831]

Wingerchuk 2014

Wingerchuk DM, Carter JL. Multiple sclerosis: current and emerging disease‐modifying therapies and treatment strategies. Mayo Clinic Proceedings 2014;89(2):225‐40. [PUBMED: 24485135]

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(5):604‐9. [PUBMED: 21532597]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Achiron 2004

Methods

RCT, parallel‐group. Recruitment period: March 1998‐March 2003. Countries: 1 (Israel). Centres: 1

Participants

N = 91. Women 74%. Age, mean (range): 34 years (15‐50 years)

Participants with a first neurological episode suggesting MS in the previous 3 months. They had positive brain MRI according to Fazekas criteria

Interventions

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 once every 6 weeks for a period of 12 months (N = 45)
Placebo (0.9% saline) intravenously monthly for 12 months (N = 46)

Outcomes

Conversion to CDMS, i.e. number of participants who experienced a second attack within 12 months

Notes

The study was supported by a research grant from Omrix Biopharmaceuticals, Tel‐Aviv, Israel, which also supplied the study drugs. The authors have no financial relationship to Omrix Biopharmaceuticals. All authors had full access to all the data and had the right to publish all the data. The data were analysed by an independent statistician (p 1519‐20)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A block‐stratified randomisation procedure (p 1516)

Allocation concealment (selection bias)

Unclear risk

"According to block‐stratified randomisation, participants were randomly assigned to each of the two treatment groups" (p 1516)

Other major baseline imbalance

Unclear risk

The study appears to be free of sources of bias related to major baseline imbalance (Table 1; p 1517)

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

"At the pharmacy, containers and tubing of IVIg or saline were wrapped in sealed opaque bags. The active treatment and placebo were administered intravenously in identical settings and regime" (p 1516)

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Each patient was evaluated by an examining neurologist who was unaware of the patient’s treatment assignment. Changes on neurological examination to determine whether a new relapse had occurred were based on the neurological examination performed by two evaluating neurologists both unaware of treatment assignment" (p 1516)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

One participant in the treated group and zero participants in the placebo group were lost to follow‐up (Fig. 1, p 1516)

Selective reporting (reporting bias)

High risk

Disability‐worsening that was included as a secondary outcome was not reported in the results

ACISS 2010

Methods

Cohort study. Austrian MS participating centres: 29. Participant recruitment started in September 2003 and terminated in December 2005. Final participant follow‐up visits in December 2007

Participants

N = 296. Women 72%. Age, mean (sd): 32.5 (9.5) years. Participants with newly diagnosed CIS. The diagnosis of a CIS was based on the presence of signs and symptoms compatible with MS without evidence for any other CNS disorder which might have caused them (Miller 2008). Monofocal presentation 80% (optic neuritis 29%; spinal cord syndrome 27%; brain stem 25%; other 19%). Multifocal presentation: 16%. EDSS , median (range): 2.0 (0‐6). Brain MRI, abnormal: 94% of participants. Oligoclonal antibodies in cerebrospinal fluid, positive: 82% of participants

Interventions

Treatments for 163 participants who completed 2 years of observation were:

N = 49 treated with DMTs within 3 months of CIS onset (early treatment). INF‐ß 1a intramuscular: 76%; INFß‐1a subcutaneous and INF‐ß 1b subcutaneous: 12% each; glatiramer acetate: 16%; others: 4%

N = 59 treated with DMTs between 3‐24 months of follow‐up (delayed treatment). IFNs: 76%; glatiramer acetate 20%; others: 4%

N = 55 never treated up to 24 months of follow‐up

Outcomes

Proportion of participants who converted to CDMS. Number of relapses over 2 years. EDSS (median and range). Quality of life assessed globally with a VAS, which ranged from 0 (worst) to 100 (best). It was recorded at baseline and through the follow‐up by the participant and the treating physician independently

Notes

No analysis was done to reduce confounding. The study organisation and monitoring were supported by Biogen Idec Austria

BENEFIT 2006

Methods

RCT, parallel‐group. Recruitment period: February 2002‐June 2003. Countries: 18 European countries, Israel and Canada. Centres: 98

Participants

N = 487. Women 71%; Age, mean (range): 31 years (18‐45 years). Participants with a a first neurological event suggestive of MS within 2 months after onset of the first event. Presentation: monofocal 52% (optic neuritis 17%; spinal cord syndrome 16%; brain stem or cerebellar syndrome 12%; other cerebral 7%), or multifocal 48%. They had at least two clinically silent lesions on their T2‐weighted brain MRI scan with a size of at least 3 mm, at least one of which being ovoid, periventricular, or infratentorial

Interventions

Interferon beta‐1b (Betaseron) 250 µg subcutaneous every other day for 24 months (N= 305)

Placebo subcutaneous every other day for 24 months (N = 182)

Corticosteroid treatment of first relapse: 71%

Participants who converted to CDMS during the double‐blind period were offered interferon beta‐1b 250 µg subcutaneously every other day for up to 5 years from randomisation. (BENEFIT 2007 (3 years FU)) (p 390)

Outcomes

Primary: time to conversion to CDMS represented by Kaplan‐Meier estimates of the cumulative percentage of participants with CDMS, defined by: 1) a relapse with clinical evidence of at least one CNS lesion, and if the first presentation was monofocal distinct from the lesion responsible for the CIS presentation, or 2) sustained progression by 1.5 points on the EDSS reaching a total EDSS score of 2.5 and confirmed at a consecutive visit 3 months later (slightly modified Poser criteria). The validity of CDMS diagnoses was confirmed by a central committee

Notes

Funded by Schering AG. Four co‐authors of Schering AG. Restriction description: any manuscript/abstract related to the study had to be submitted for review to the sponsor at least 90 days prior to publication (clinicalTrials.gov)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A minimization procedure with an element of chance was applied to minimize imbalance of treatment groups for (selected) factors with potential impact on the risk of developing definite MS: steroid use during the first clinical event; onset of the first event as monofocal vs multifocal by central assessment; number of T2 lesions on the screening MRI" (p 1243)

Allocation concealment (selection bias)

Low risk

Central randomisation in a 5:3 ratio (p 1243)

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance (table 1; p 1244)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"After CDMS confirmation, all evaluations foreseen per protocol for the month 24/end‐of‐study visit were performed. At this end‐of‐study visit—without breaking the randomisation code—participants were given the option of participating in the follow‐up study with open‐label interferon beta‐1b treatment" (p 1243). High risk of un‐blinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Participants were instructed to cover injection sites during the examination by the masked evaluation neurologist". The diagnosis of CDMS had to be confirmed by a central committee whose masking was not reported (p 1243)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Proportions and reasons of incomplete outcome data did not differ substantially across intervention groups. 437 (89.7%) of the 487 randomised participants completed the study (271 (88.8%) of 305 allocated to IFNB‐1b and 166 (91.2%) of 182 allocated to placebo). 34 (11.2%) IFNB‐1b participants did not complete the study: 13 did not receive IFNB‐1b and were not followed; 8 adverse event; 3 lost to follow‐up; 9 withdrawal by subject; 1 adverse event, then subject's withdrawal. 16 (8.8%) placebo did not complete the study: 6 did not receive placebo and were not followed; 2 lost to follow‐up; 7 withdrawal by subject; 1 fulfilled local definition and McDonald criteria

Selective reporting (reporting bias)

Low risk

The study protocol was not available but it is clear that the published reports included all expected outcomes, including those that were pre‐specified

BENEFIT 2007 (3 years FU)

Methods

OLE study of the BENEFIT 2006 (placebo‐controlled phase of 24 months). Centres: 97 of the original 98 BENEFIT study sites

Participants

N = 418. Women 71%. Age, median: 30 years

Interventions

N = 261/305 originally randomised to Interferon beta‐1b 250 µg subcutaneous every other day (early‐treatment group). Active treatment exposure: 36 months

N = 157/182 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 12 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 36 months. Annualised relapse rate

Notes

Multivariate Cox regressions for time to CDMS and time to McDonald MS (steroid use during the first clinical event, onset of disease (monofocal vs multifocal), age at screening, sex, and number of T2 lesions and gadolinium‐enhanced lesions at screening; time to confirmed EDSS progression was adjusted (as preplanned) for T2‐lesion volume at screening)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Low risk

Clinical characteristics were similar between the two groups.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

Low risk

18% and 21% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons of missing participants did not differ substantially across intervention groups.

Selective reporting (reporting bias)

High risk

Serious adverse events not reported

BENEFIT 2009 (5 years FU)

Methods

OLE study of the BENEFIT 2006 (placebo‐controlled phase of 24 months). Centres: 97 of the original 98 BENEFIT study sites

Participants

N = 418. Women 71%. Age, median: 30 years

Interventions

N = 261/305 originally randomised to Interferon beta‐1b 250 µg subcutaneous every other day (early‐treatment group). Active treatment exposure: 60 months

N = 157/182 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 36 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 60 months. Annualised relapse rate and proportions with relapses

Notes

Adjusted Cox proportional hazards regression for time to conversion (steroid use during the first clinical event, onset of disease (monofocal vs multifocal), age, sex, and number of T2 lesions and gadolinium‐enhancing lesions at screening)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

23% and 32% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

High risk

Treatment discontinuation for adverse events not reported

BENEFIT 2014 (8.7 years FU)

Methods

OLE study of the BENEFIT 2006 (placebo‐controlled phase of 24 months). Centres: 72 of the original 98 BENEFIT study sites

Participants

N = 284 recruited from 72 of the 97 initial centres in the BENEFIT RCT. Women 71%. Age, median: 30 years

Interventions

N = 178/305 originally randomised to Interferon beta‐1b 250 µg subcutaneous every other day (early‐treatment group). Active treatment exposure: 60 months

N = 106/182 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 36 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 8.7 years. Annualised relapse rate

Notes

Proportional hazards regression (covariates: randomised treatment, steroid use during the first clinical event, type of disease onset and categorised number of T2 lesions on BENEFITscreening MRI)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

48.5% and 44.5% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

High risk

Treatment discontinuation for adverse events not reported

BENEFIT 2016 (11 years FU)

Methods

Following the OLE study, a prospective, comprehensive, 11‐year, cross‐sectional reassessment of the BENEFIT 2006 (placebo‐controlled phase of 24 months). Centres: 66 of the original 98 BENEFIT study sites

Participants

N = 278 recruited from 66 of the 97 initial centres in the BENEFIT RCT. Women 70%. Age, median: 30 years

Interventions

N = 167/305 originally randomised to Interferon beta‐1b 250 µg subcutaneous every other day (early‐treatment group). Active treatment exposure: 60 months

N = 111/182 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 36 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 11 years. Annualised relapse rate

Notes

Proportional hazards regression for time‐to‐event outcomes and generalised linear regression models. Steroid use during first event (yes or no), multifocal or monofocal onset of disease, and number of T2 lesions at screening (2–4, 5–8, or ≥9) included as the standard set of covariates. An extended set of covariates that included number of gadolinium‐enhancing (Gd1) lesions at screening, age, and sex in addition to the standard covariates was used for analysis of time to CDMS, time to first relapse, and ARR

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Insufficient information

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

45% and 39% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

High risk

Treatment discontinuation for adverse events not reported

CHAMPS 2000

Methods

RCT, parallel‐group. Recruitment period: April 1996‐March 2000. Countries: 2 (USA and Canada). Centres: 50

Participants

N = 383. Women 75%. Age, mean (range): 33 years (18‐50 years). Participants with a first isolated, well‐defined neurologic event no more than 27 days before randomisation. Monofocal presentation: 70% (optic neuritis 50%; spinal cord syndrome 22%; brain stem or cerebellar syndrome 28%). They had 2 or more clinically silent lesions of the brain that were at least 3 mm in diameter on MRI scans (at least 1 lesion had to be periventricular or ovoid)

Interventions

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 18 months (N = 193)

Placebo intramuscular once a week for 18 months (N = 190)

All participants (100%) received corticosteroid treatment (18 days)

Acetaminophen (paracetamol) 650 mg before and after each injection during the first 6 months of treatment

Outcomes

Primary: conversion to CDMS as defined by: 1) a new clinical abnormality consistent with the participant's report of neurological or visual symptom distinct from that of the initial episode at study entry or: 2) worsening by 1.5 points on the EDSS confirmed at a consecutive visit 3 months later (slightly modified Poser criteria)

Notes

Funded by Biogen. Stopped after 18‐month interim analysis

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"To assign participants randomly in approximately equal numbers to the two treatment groups, we used a minimization procedure to minimize imbalance of treatment groups for (selected) factors the number of lesions on T2‐weighted MRI scans and the type of initial clinical event" (p 899)

Allocation concealment (selection bias)

Unclear risk

No information

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Unclear risk of unblinding because it is unclear if participants could have shifted to active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Each patient was examined by a treating and an examining neurologist, both of whom were unaware of the patient’s treatment assignment (p 899). Clinical outcomes were confirmed by a central end‐point committee whose members were unaware of the participants’ treatment assignments" (p 899)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Truncated. The study period was planned to be 3 years. It was stopped early after 18‐month interim analysis of efficacy. 177 participants (46% of the randomised) (80 treated and 97 placebo) had completed the study. At 3 years, 30 withdrawals + 83 interrupted = 113 (58.5%) in interferon group; 27 withdrawals + 66 interrupted = 93 (48.9%) in placebo group. (Fig. 1; p 902)

Selective reporting (reporting bias)

Low risk

The study protocol was not available but the published reports included all expected outcomes, including those that were pre‐specified

CHAMPS 2006 (5 years FU)

Methods

OLE study of the CHAMPS 2000 (placebo‐controlled phase of 36 months) Centres: 32 of the original 50 CHAMPS study sites

Participants

N = 203 recruited from 32 of the 50 initial centres in the CHAMPS trial. Women 77% (early treatment), 74% delayed treatment. Age, mean: 35 years

Interventions

N = 100/193 originally randomised to interferon beta‐1a (Avonex) 30 µg intramuscular once a week (early‐treatment group). Active treatment exposure: 60 months

N = 103/190 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 24 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 60 months. Annualised relapse rate

Notes

Multivariate model was used to adjust for recipient age, clinical centre, baseline brain MRI T2 lesion volume (log transformation), and the number of Gd lesions at baseline. Effect modification related to these factors was assessed with interaction terms in the model. Possible violations of the proportional hazards assumption were checked using time‐dependent variables

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Low risk

Demographic and clinical characteristics were similar between the 2 groups

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

50% and 51% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

High risk

Treatment discontinuation for adverse events and serious adverse events not reported

CHAMPS 2012 (10 years FU)

Methods

OLE study of the CHAMPS 2000 (placebo‐controlled phase of 36 months). Centres: 24 of the original 50 CHAMPS study sites

Participants

N = 155 recruited from 24 of the 50 initial centres in the CHAMPS trial. Women 74% (early treatment), 72% delayed treatment. Age, mean: 35 years

Interventions

N = 81/193 originally randomised to interferon beta‐1a (Avonex) 30 µg intramuscular once a week (early‐treatment group). Active treatment exposure: 120 months

N = 74/190 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 84 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 60 months. Annualised relapse rate

Notes

Multivariate model was used to adjust for recipient age, clinical centre, baseline brain MRI T2 lesion volume (log transformation), and the number of Gd lesions at baseline. Effect modification related to these factors was assessed with interaction terms in the model. Possible violations of the proportional hazards assumption were checked using time‐dependent variables

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Insufficient information about clinical characteristics of participants

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

65% and 69% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

High risk

Treatment discontinuation for adverse events not reported

ETOMS 2001

Methods

RCT, parallel‐group. Recruitment period: August 1995‐July 1997. 14 countries in Europe. Centres: 57

Participants

N = 309. Women 64%. Age, mean (range): 28 years (18‐40 years)

Participants with a first neurological episode suggesting multiple sclerosis in the previous 3 months. Monofocal (61%) or multifocal presentation. They had positive brain MRI for at least 4 white‐matter lesions on the T2‐weighted scans, or presence of at least 3 white‐matter lesions, if at least one was infratentorial or enhancing after gadolinium

Interventions

Interferon beta‐1a (Rebif ) 22 µg subcutaneous once a week for 24 months (N = 154)
Placebo subcutaneous once a week for 24 months (N = 155)

Steroid use at first clinical demyelinating event: 70%

After the conversion to CDMS, the investigator discussed with the participant the possibility of starting open‐label treatment with interferon beta‐1a once weekly until the completion of the trial (p 1577)

Outcomes

Primary: conversion to CDMS defined according to Poser diagnostic criteria

Notes

Funded by Serono. COI of authors not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The treatment was assigned according to a computer‐generated randomisation list stratified by centre" (p 1577)

Allocation concealment (selection bias)

Unclear risk

No information

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"After the occurrence of the second exacerbation, as stipulated in the protocol, the investigator discussed with the patient the possibility of starting open‐label treatment with interferon beta‐1a once weekly until the completion of the trial". (p 1577) High risk of unblinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"At each study site, a treating physician was responsible for the overall management of the patient, including safety monitoring. An evaluating physician was responsible for all scheduled neurological examinations and exacerbation follow‐up. Two members of the steering committee reviewed the documentation of all exacerbations and, by consensus, classified them as confirmed or unconfirmed" (p 1577)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Proportions and reasons of incomplete outcome data did not differ substantially across intervention groups. 141 (91.6%) of 154 participants in Interferon beta‐1a and 137 (88.4%) of 155 participants in the placebo group completed the study

Selective reporting (reporting bias)

Low risk

The study protocol is available and all of the study’s pre‐specified (primary and secondary) outcomes have been reported

GERONIMUS 2013

Methods

Prospective cohort study. Regione Emilia Romagna, Italy. MS participating centres: 22. Recruitment of participants from December 2004‐June 2007. Censoring date: 31 March 2010

Participants

N = 168. Women 69%. Age, mean (sd): 33.0 (8.0) years. Participants with first symptom suggestive of an inflammatory demyelinating disorder of the central nervous system in the preceding 6 months. Monofocal presentation 73% (optic neuritis 29%; brain stem/cerebellar 21%; cerebral or spinal 23%). Multifocal presentation: 27%. EDSS, median (range): 1.0 (0‐6.5). Participants with MRI positive for ≥ 3 Barkhof criteria: 60% of participants. Oligoclonal antibodies in cerebrospinal fluid, positive: 68% of participants

Interventions

N = 31 (18%) and N = 51 (30%) of participants were treated with disease‐modifying drugs before or after conversion to CDMS. N = 86 not treated.

N = 67 interferon; N = 9 glatiramer acetate; N =3 intravenous immunoglobulin; N =2 azathioprine; N = 1 mitoxantrone

N = 18 participants underwent at least one other treatment: natalizumab (10); glatiramer acetate (5); mitoxantrone (3); azathioprine (2); plasma exchange (1)

Outcomes

CDMS according to Poser criteria. Follow‐up 2 and 4 years

Notes

Multivariate analysis was done using the Cox proportional‐hazard regression model. Functional systems at onset were categorised as afferent (visual or sensitive or both), efferent (any of the others) or combined (afferent and efferent); Barkhof criteria were dichotomised as C3 of 4 (positive) versus B2 of 4 (negative). All the variables statistically significant in the univariate analysis for conversion to MS according to either McDonald criteria or CDMS criteria were simultaneously entered in the multivariate model, except the number of T2 lesions and GD positive lesions that are already included in Barkhof criteria. This study was supported with an unconditional grant by Biogen Idec.

Motamed 2007

Methods

RCT. Recruitment period: October 2002‐March 2005. Country: Iran. One centre

Participants

N = 25. Women 68%. Age, mean (range): 25 years (17‐39 years)

Participants with a first, isolated optic neuritis (32%), spinal cord (28%), brain stem (24%) or cerebellar (16%) syndrome, and which was confirmed on ophthalmologic or neurologic examination

Mean EDSS: 1.74 (SD = 0.76)

MRI scan judged to be positive according to McDonald criteria (revision of 2005)

Interventions

N = 11: interferon beta‐1a (Rebif) 22 µg subcutaneous 3 times a week for 21 months

N = 14: no disease‐modifying treatment

Outcomes

Worsening of disability measured by Kurtzke Expanded Disability Status Scale (EDSS) and numbers of new relapses during 21 months of follow‐up

Notes

Sponsor not reported. Potential conflicts of interest of authors not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information

Allocation concealment (selection bias)

Unclear risk

No information

Other major baseline imbalance

Unclear risk

Baseline MRI findings imbalance. (Table 1; p 346)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Absence of blinding. (p 348)

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Absence of blinding. (p 348)

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No information

Selective reporting (reporting bias)

Unclear risk

No information

MSBASIS 2016

Methods

Prospective cohort study (the MSBase Incident Study ‐ MSBasis) from an international MSBase Registry. MSBasis started in November 2004. Centres: 50. Countries: 22

Participants

N = 3296. Women 70.5%. Age, median (IQR): 31.6 (25.3‐39.3) years. Registry participants with a CIS with symptom onset less than 12 months from the enrolment date. Clinical presentation: optic pathways 22%; supratentorial 20.5%; brainstem 21.5%; spinal cord syndrome 26%). EDSS, median (IQR): 2.0 (1‐2.5). Abnormal T1 and T2 MRI scans were recorded in 47% and 96% of participants, respectively. Oligoclonal antibodies in cerebrospinal fluid, positive: 32% of participants

Interventions

N = 910 (28%) participants were treated with intramuscular IFNβ‐1a (42.7%), subcutaneous IFNβ‐1a (33.8%), IFNβ‐1b (18.4%), or glatiramer acetate (13.7%)

N = 2386 (72%) were not exposed to disease‐modifying drugs during follow‐up

Outcomes

Primary outcome: time to CDMS, i.e. individualised risk of clinical conversion to CDMS at 12 months. CDMS defined as examination evidence of a symptomatic second neurological episode attributable to demyelination of more than 24 hs' duration and more than 4 weeks from the initial attack (Poser criteria). N = 5378.70 person‐years contributed to outcome data

Notes

All models presented were adjusted for country to control for any residual inter‐country heterogeneity, for baseline and time‐varying factors.

The MSBasis study was supported by Merck Serono, between 2004 and 2009

ORACLE 2014

Methods

RCT, parallel‐group. Recruitment period: October 2008‐October 2010. Countries: 34 (Argentina (2), Austria (2), Belgium (4), Bosnia and Herzegovina (1), Bulgaria (9), Canada (1), Croatia (2), Czech Republic (6), Estonia (2), Finland (4), France (3), Georgia (3), Germany (2), India (6), Italy (19), Korea (5), Lebanon (1), Macedonia (1), Norway (2), Poland (9), Portugal (4), Romania (4), Russia (25), Serbia (3), Singapore (1), Spain (2), Sweden (3), Taiwan (3), Thailand (1), Turkey (2), Ukraine (3), United Arab Emirates (1), UK (1), USA (23). Centres: 160

Participants

N = 617. Women 65%. Age, mean (range): 32 years (18‐55 years). Presentation: monofocal 52%; multifocal 48%. Participants with a first clinical demyelinating event within 75 days before screening. They had an abnormal brain MRI consisting of at least two clinically silent T2‐weighted MRI lesions, at least one of which was ovoid, periventricular, or infratentorial, of at least 3 mm in diameter

Interventions

Cladribine cumulative dose: 3.5 mg/kg body weight oral for 22 months (N = 206)

Cladribine cumulative dose: 5.25 mg/kg body weight oral for 22 months (N = 205)

Placebo oral tablets (undefined) oral for 22 months (N = 206)

Corticosteroid treatment of first relapse: 66%

Participants who converted to CDMS during the double‐blind period entered the open‐label maintenance period and were offered open‐label treatment with subcutaneous interferon beta‐1a, 44 μg 3 times weekly. participants who did not convert to CDMS were eligible to enter the long‐term follow‐up without study drug until conversion to MS according to the 2005 McDonald criteria, when they were treated with open‐label cladribine 3.5 mg/kg under the original design, or with subcutaneous interferon beta‐1a 44 μg three times weekly after the protocol amendment due to the sponsor’s decision to terminate development of oral cladribine. Participants converting to CDMS during long‐term follow‐up received interferon beta‐1a 44 μg 3 times weekly. (p 258)

Outcomes

Primary: time to CDMS conversion represented by Kaplan‐Meier estimates of the cumulative percentage of participants with CDMS (time frame: baseline up to month 22) defined according to Poser criteria, i.e. the occurrence of a second attack or a sustained increase in the expanded disability status scale (EDSS) score.

Secondary: 1) time to develop MS conversion according to the revised McDonald Criteria (2005) represented by Kaplan‐Meier estimates of the cumulative percentage of participants with McDonald MS (time frame: baseline up to month 22); 2) number of participants with adverse events and serious adverse events (time frame: baseline up to month 22)

Notes

Funded by Merck Serono SA Geneva, a subsidiary of Merck KGaA, Darmstadt, Germany. Early termination following the sponsor's decision (October 2011) (p 258). The study was designed by members of the steering committee and the sponsor. Data were collected, analysed, and interpreted by the sponsor. All authors had access to the data and contributed to data analysis and interpretation (p 261)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation was done using a central web‐based randomisation system and was stratified by geographic region". (p 258)

Allocation concealment (selection bias)

Low risk

A central web‐based randomisation system. (p 258)

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"Participants who converted to CDMS during the double‐blind period entered the open‐label maintenance period and were offered open‐label treatment with subcutaneous interferon beta‐1a, 44 μg three times weekly". (p 258) High risk of unblinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"Masking was maintained using a two‐physician model (both doctors were masked). The treating physician supervised study medication administration, and recorded and treated adverse events and MS relapses. The evaluating physician assessed all neurological findings and relapses, and was additionally masked to patient laboratory data. For every patient, conversion to CDMS required confirmation and approval by a sponsor‐appointed, treatment‐blinded study adjudication committee". (p 258)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Truncated. The study period was planned to be 22 months. It was stopped early following the sponsor’s decision to stop the cladribine programme (Supplementary web‐appendix). 211 (34%) of 614 randomised participants (104 Cladribine 5.25 mg/kg; 131 Cladribine 3.5 mg/kg; and 104 placebo) completed the study (Fig. 3; p 262). The number excludes participants who converted to CDMS during the double‐blind period, and therefore left the double‐blind period to enter the open‐label maintenance

Selective reporting (reporting bias)

High risk

In the original protocol, an analysis of disability‐worsening was to be done in participants who had converted to CDMS but, owing to the early trial termination, the sponsor decided before database lock, and with an amended statistical analysis plan, not to analyse time to disability‐worsening. (p 260)

Pakdaman 2007

Methods

RCT, parallel‐group. Recruitment period: February 2002‐August 2005. Country: Iran. Centres: 4

Participants

N = 217. Women 68%. Age, range: 19‐50 years. Participants with a first optic neuritis (48%), spinal cord syndrome (24%), brain stem or cerebellar syndrome (22%) in the previous 3 months confirmed by neurologic examination. They had an abnormal brain MRI consisting of 2 or more clinically silent lesions that were at least 3 mm in diameter and at least 1 had to be periventricular or ovoid

Interventions

Interferon beta‐1a (Avonex) 30 µg intramuscular once a week for 36 months (N = 104 included in analysis)
Placebo (unspecified) for 36 months (N = 98 included in analysis)

Outcomes

Primary: time to conversion to CDMS as defined by the occurrence of a second exacerbation that was attributed to a part of central nervous system that differed from the initial episode at study entry

Secondary: time to second exacerbation

Notes

Sponsor not reported. Potential conflicts of interest of authors not reported

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information

Allocation concealment (selection bias)

Unclear risk

No information

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance (Table 1; p 430)

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Insufficient information. Only "double blind trial" is reported

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Of the 217 participants randomised, 202 (93%) completed the study; 104 received interferon beta 1a and 98 received placebo. Data on participants in whom CDMS did not occur were censored on the date they were last seen by the neurologist. (p 430)

Selective reporting (reporting bias)

High risk

Selective under‐reporting of data: conversion to CDMS was reported but with inadequate detail for the data

PRECISE 2009

Methods

RCT, parallel‐group. Recruitment period: January 2004‐January 2006. 16 countries worldwide, in 80 centres from the USA, Europe, Argentina, Australia, and New Zealand

Participants

N = 481. Women 67%. Age, mean (range): 31 years (18‐45 years). Monofocal presentation: 100%. Participants with one unifocal neurological event within 90 days after onset. They had positive brain MRI for at least 2 cerebral lesions on the T2‐weighted images of at least 6 mm in diameter

Interventions

Glatiramer acetate (Copaxone) 20 µg subcutaneous once a day for 36 months (N = 243)
Placebo subcutaneous once a day for 36 months (N = 238)

Steroid use at first clinical demyelinating event: 64%

Ibuprofen (400 mg) or paracetamol (acetaminophen) (1000 mg) prophylactically with each injection during the first 3 months of treatment.

All participants switched to active treatment with glatiramer acetate upon conversion to CDMS. (p 1506)

Outcomes

Primary: time to conversion to CDMS defined by: 1) a second event suggestive of MS lasting at least 48 h duration or: 2) worsening by 1.5 points on the EDSS confirmed at a consecutive visit 3 months later (slightly modified Poser criteria)

Notes

Funded by TEVA. The sponsor was involved in the study design, conduct, monitoring, data analysis, and writing of the report. The corresponding author had full access to all the data and had final responsibility for the decision to submit for publication (p 1507)

Restriction: should the investigator wish to publish the results of this study, he/she agrees to provide Teva with a manuscript for review 60 days prior to submission for publication. Teva retains the right to delete confidential information and to object to suggest publication and/or its timing (at the Company's sole discretion).

If Teva chooses to publish this study a copy will be provided to the investigator at least 30 days prior to the expected date of submission to the intended publisher (trial.gov)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"The randomisation scheme was produced by the sponsor of the study with a 1:1 assignment ratio. A SAS‐based blocks with block size of 4, stratified by centre was used". (p 1504)

Allocation concealment (selection bias)

Unclear risk

No information

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance. (table 2; p 1506)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The criterion to enter the prospectively planned open label study phase was either a second relapse or the end of the double‐blind phase, whichever came first. High risk of unblinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Unclear risk for blinding of clinical outcome assessment. "Treating and examining neurologists at the sites were masked to MRI results during the study. The unmasked statistician presented unmasked results to the Data Monitoring Committee, as per their request" (p 1504)

Incomplete outcome data (attrition bias)
All outcomes

High risk

Truncated. The study period was planned to be 3 years. Based on the results of a planned interim analysis of efficacy and on the recommendations of the data monitoring committee (unmasked), the trial was stopped early and all participants were switched to glatiramer acetate. At the time of the interim analysis, 230 (47.8%) of 481 randomised participants completed the study. 98 (40.3%) of 243 treated participants and 132 (55.5%) of 238 placebo completed the study. Proportion and reasons of incomplete data differed between the groups. 39 (16.0%) of 243 participants in the glatiramer group and 23 (8.8%) of 238 in the placebo group discontinued treatment early (table 1, p 1505) and the proportion of termination because of adverse events differed significantly between the two treatment groups

Selective reporting (reporting bias)

Low risk

The study protocol was available and all of the study’s pre‐specified primary outcomes were reported

PRECISE 2013 (5 years FU)

Methods

OLE study of the PRECISE 2009 (placebo‐controlled phase of 36 months). Centres: 80 of the original 80 PRECISE study sites

Participants

N = 409. Women 69% and 65% in the early and delayed groups, respectively. Age, median: 30 years

Interventions

N = 198/243 originally randomised to glatiramer acetate (early‐treatment group). Active treatment exposure: 60 months

N = 211/238 originally randomised to placebo (delayed‐treatment group). Active treatment exposure: 24 months

Outcomes

Time to CDMS and time to confirmed disability progression measured by EDSS scale at 60 months. Annualised relapse rate. Proportion of participants with relapses or disability progression

Notes

Risk of conversion to CDMS from a Cox’s proportional hazards model was assessed for early‐ and delayed‐treatment subgroups defined by demographics, characteristics of CIS (gender, age, presenting syndrome, steroid treatment for the initial attack) and MRI findings (disease dissemination and activity), at baseline

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to the OLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Clinical characteristics were similar between the two groups

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

33% and 47% of early and delayed treatment groups, respectively, dropped out. Proportions and reasons for missing participants differed substantially across intervention groups

Selective reporting (reporting bias)

Low risk

Prespecified outcome were reported including adverse events and serious adverse events

REFLEX 2012

Methods

RCT, parallel‐group. Recruitment period: November 2006‐August 2010. 28 countries in Europe and Canada. Centres: 78

Participants

N = 517. Women 64%. Age, mean (range): 31 years (18‐50 years). Participants with a single event suggestive of MS within 60 days before study entry. Presentation: monofocal 54%; multifocal 46%. They had at least two clinically silent lesions of 3 mm or more on T2‐weighted brain MRI scan, at least one of which was ovoid, periventricular, or infratentorial

Interventions

Interferon beta‐1a (Rebif ) 44 µg subcutaneous 3 times a week for 24 months (N = 171)
Interferon beta‐1a (Rebif ) 44 µg subcutaneous once a week and placebo subcutaneous 2 times a week for 24 months (N = 175)
Placebo subcutaneous 3 times a week for 24 months (N = 171)

Steroid use at first clinical demyelinating event: 71%

Ibuprofen (400 mg) or paracetamol (acetaminophen) (1000 mg) prophylactically with each injection during the first 3 months of treatment.

On conversion to CDMS, participants were switched to open‐label subcutaneous interferon beta‐1a at 44 μg 3 times a week until the end of the 24 months

Outcomes

Primary: time to conversion to MS according to the McDonald Criteria (2005) to 24 months

Secondary: time to conversion to CDMS defined by either a second attack or a 3‐month sustained increase (≥ to 1.5 points) in EDSS score (slightly modified Poser criteria)

Notes

Funded by Merck Serono. The study was designed by members of the steering committee and the sponsor. The sponsor collected the data, did the analysis, and was involved in the interpretation of the data. The data were available to all authors, and they contributed to the analysis and interpretation of the data. The steering committee was responsible for the final decision to submit this report for publication.

Restriction: sponsor has the right to publish any results communication in connection with the study. The PI shall submit any communications including study results to the sponsor for review 30 working days prior to communication submission. The sponsor can request the PI to modify or delete any sponsor's proprietary information. If the PI refuses the modification, the submission shall be postponed for 60 days from PI refusal, to provide the sponsor the opportunity to file a patent or seek legal remedies (trial.gov)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation. "Randomisation was stratified according to baseline factors: age (<30 years vs ≥30 years), steroid use for first event (yes vs no), classification of first event (monofocal vs multifocal), and at least one MRI gadolinium‐enhancing lesion (yes vs no)". (p 34)

Allocation concealment (selection bias)

Low risk

"The study centre dialled a centralised interactive voice response system to randomly assign participants in a 1:1:1 ratio" (p 34). "A treatment kit number, corresponding to the randomisation group, was allocated centrally to each patient for use only by that individual". (p 34)

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance. (table 1; p 36)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

"On conversion to CDMS, participants were switched to open‐label subcutaneous interferon beta‐1a at 44 μg three times a week until the end of the 24 months". (p 34) High risk of unblinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"A two‐physician (treating and assessing) model was used to assist with study masking. The treating physician was responsible for supervision of study drug administration and for recording adverse events and safety assessments. The assessing physician was not involved in the care of study participants and was exclusively responsible for all neurological assessments, beginning with the pre‐study assessment. Injection sites were covered before a patient saw the assessing physician to maintain masking". (p 34)

Incomplete outcome data (attrition bias)
All outcomes

Low risk

448 (87%) of the 517 randomised participants had completed the study: 146 (85%) of 171 participants in interferon beta‐1a 44 µg 3 times a week; 156 (89%) of 175 participants in interferon beta‐1a 44 µg once a week; 146 (85%) of 171 in placebo. Proportion and reasons of withdrawn from study did not differ between the groups: 26 (15.2%) of 171 participants in interferon beta‐1a 44 µg 3 times a week; 20 (11.4%) of 175 participants in interferon beta‐1a 44 µg once a week; and 26 (15.2) of 171 in placebo

Selective reporting (reporting bias)

Low risk

The study protocol was available and all of the study’s pre‐specified (primary and secondary) outcomes were reported

REFLEX 2016 (3 and 5 years FU)

Methods

OLE study of the REFLEX 2012 (placebo‐controlled phase of 24 months). Centres: 70 of the original 78 REFLEX study sites

Participants

N = 402. Women 61%, 62% and 62% in the 3 groups, respectively. Age, mean: 31.4 (SD 8.3) years

Interventions

N = 127/171 originally randomised to sc IFN β‐1a 44 mg tiw (early‐treatment group). Active treatment exposure: 60 months

N = 142/175 originally randomised to sc IFN β‐1a 44 μg qw (early‐treatment group). Active treatment exposure: 60 months

N = 133/171 in the delayed‐treatment arm (originally randomised to placebo). Active treatment exposure: 36 months

Outcomes

Time to CDMS conversion (defined in REFLEX 2012) from first randomisation to month 36; time to CDMS to month 60 (secondary end point). Proportion of participants remaining relapse‐free; time to confirmed disability‐worsening (increase of EDSS ≥ 1.0 point, confirmed during a visit 6 months later) and EDSS change from baseline

Notes

Probabilities of CDMS conversion, McDonald MS conversion and EDSS progression over time were determined for each treatment group in the form of cumulative incidence curves estimated using the non‐parametric Kaplan‐Meier method

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

No random assignment to theOLE groups

Allocation concealment (selection bias)

High risk

No allocation concealment to the OLE groups

Other major baseline imbalance

Unclear risk

Insufficient information about clinical characteristics of participants

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Unblinding

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Unblinding

Incomplete outcome data (attrition bias)
All outcomes

High risk

30%, 25%, and 29% dropped out respectively in early and delayed treatments groups with different reasons. Not reported reasons for discontinuation across the groups

Selective reporting (reporting bias)

Low risk

All expected outcomes were reported

Tintore 2015

Methods

Prospective cohort study. Study started in January 1995 and the database was locked on 15 March 2013

Participants

N = 1015. Women 68%. Age, mean (SD): 31.1(8.2) years. Monocentric. Participants with CIS that was suggestive of CNS demyelination and was not attributable to other diseases, with symptom onset within 3 months of the first clinical evaluation. Clinical topography at onset: optic neuritis 37%; spinal cord syndrome 26%; brain stem 27%; other 11%. EDSS , median (range): 2.0 (0‐6). Brain MRI available for 94% of participants and abnormal in 69% of them. Oligoclonal antibodies in cerebrospinal fluid available for 79% of participants and positive in 57% of them.

N = 1058 enrolled

‐ N = 43 (4%) excluded for various reasons: previous attack (N = 7), age over 50 (N = 4), exceeded entry window (N = 12), and alternative diagnosis (N = 20)

‐ N = 1015 included in analysis

‐ N = 7 (0.7%) died during follow‐up: car accident (N = 1), myocardial infarction (N = 1), pancreatic cancer (N = 1), meningitis as a complication of septoplasty (N = 1), septic shock in a participant with severe disability (N = 1), cardiogenic shock of unknown origin (N = 1) and acute leukemia in a participant who received mitoxantrone (N = 1)

Interventions

N = 388 (38.3%) of participants were on DMT at least once during follow‐up (IFNs or glatiramer acetate).

N = 174 (45%) of participants were on DMTs prior to conversion to clinical definite MS (early treatment)

N = 214 (55%) of participants were on DMTs after conversion to CDMS (delayed treatment)

N = 376 (97%) of participants were on DMTs prior to reaching an EDSS score of 3.0

N = 281(75.3%) of 375 participants with 3–4 Barkhof criteria at baseline received DMT, 143 (51%) of those prior CDMS (early treatment), and 273 (97%) prior to EDSS score of 3.0

The mean time to DMT was significantly shorter in the participants with CIS from 2002–2007 compared with those from 1995–2001 (15.2 months SD = 21 versus 41.5 months, SD = 38, P < 0.001)

Outcomes

CDMS and disability‐worsening (reaching EDSS score 3.0). Clinical follow‐up duration, mean (SD) (range): 81 (57) (0.3–220) months.

The participants were evaluated on a regular basis (every 3‐6 months or annually depending on each participant’s characteristics). The participants who did not attend two consecutive follow‐up visits were defined as ‘lost to follow‐up’

Notes

Multivariate Cox proportional hazards regression analyses for the time to conversion to CDMS or McDonald. Covariates including age, gender, clinical topography, oligoclonal bands, MRI criteria (Barkhof criteria and the number of lesions) and DMT onset prior to the diagnosis of CDMS or McDonald 2005 multiple sclerosis criteria, depending on the outcome, were considered. Possible interactions between age, gender, topographic characteristics, the presence of oligoclonal bands, the number of lesions and DMT were also evaluated. DMT was used in these models as a time‐dependent variable to take into account the date of treatment onset.

This work is independent of all the funding bodies, which have played no part in any of its stages.

TOPIC 2014

Methods

RCT, parallel‐group. Recruitment period: February 2008‐August 2012. 20 countries in Europe, USA, Canada, and Australia. Centres: 112

Participants

N = 618. Women 68%. Age, mean (range): 32 years (18‐55 years). Presentation: monofocal 59%; multifocal 41%. Participants with a first acute or subacute optic neuritis, spinal cord syndrome, brain stem or cerebellar syndrome occurring within 3 months before randomisation. They had an abnormal brain MRI consisting of at least two T2‐weighted MRI lesions of at least 3 mm in diameter

Interventions

Teriflunomide 14 mg oral capsule once daily for up to 25 months (N = 216)
Teriflunomide 7 mg oral capsule once daily for up to 25 months (N = 205)
Placebo oral capsule once daily for 25 months (N = 197)

Previous systemic corticosteroid treatment: 14%

Outcomes

Primary: conversion to CDMS as defined by the occurrence of a second relapse (Poser diagnostic criteria)

Secondary: time to relapse

Notes

Funder Sanofi, Genzyme. Data were obtained by the investigators and were analysed by the sponsor. Interpretation of the data was done by the sponsor and the authors. All authors had full access to, and take responsibility for, the veracity of study data.

TOPIC was stopped on Aug 10, 2012, because the 2010 revisions of the MCDonald diagnostic criteria enabled an earlier diagnosis of multiple sclerosis, in some cases at first clinical event. Re‐evaluation of the power calculation based on updated information from the teriflunomide clinical programme, especially from the TOWER study, indicated that sufficient power to detect a reduction in risk of relapse had already been achieved

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation was done centrally, by an interactive voice recognition system that generated an allocation sequence using a permuted‐block randomisation schedule (block size of six) with stratification by baseline monofocal or multifocal status". (p 978)

Allocation concealment (selection bias)

Low risk

"An independent company (ClinPhone, Perceptive Informatics, Nottingham, UK) ran and maintained the interactive voice recognition system for the duration of the study, under the responsibility of the study funder. After a screening phase (up to 4 weeks), investigators called the interactive voice recognition system to receive a random, masked treatment assignment for each patient". (p 978)

Other major baseline imbalance

Low risk

The study appears to be free of sources of bias related to major baseline imbalance. (table 1; p 980)

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Participants who had a relapse which defined CDMS, and had been treated for at least 24 weeks, could also enter the OLE study. (p 978) High risk of unblinding after shifting to open‐label active treatment during the randomised study

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"A treating neurologist at each site assessed participant eligibility, supervised study drug administration, and did the safety assessments. An independent examining neurologist was responsible for all functional system and EDSS assessments. Relapses indicating CDMS were confirmed by the treating neurologist based on the examining neurologist’s EDSS assessment" (p 979)

Incomplete outcome data (attrition bias)
All outcomes

High risk

At least 41% and 45% in the treated groups and 40% in the placebo group were lost to follow‐up (Fig. 2; p 980)

Selective reporting (reporting bias)

Low risk

The study protocol is not available, however the published reports included all expected outcomes

CDMS: clinically definite multiple sclerosis; CIS: clinically isolated syndrome; CNS: central nervous system; DMT: disease‐modifying treatment; EDSS: Expanded Disability Status Scale; h: hour; IFN: interferon; MRI: magnetic resonance imaging; MS: multiple sclerosis; N: number; OLE: open‐label extension; VAS: visual analogue scale

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

BENEFIT 2007

Subanalysis of BENEFIT 2006 to evaluate prognostic effect of MRI at baseline on conversion to CDMS

BENEFIT 2008

Subanalysis of BENEFIT 2006 trial to evaluate predictive effect of treatment in subgroups of participants

BENEFIT 2011

Subanalysis of BENEFIT 2006 trial to evaluate the frequency and consequences of neutralizing antibodies in subgroups of participants

BENEFIT 2012

Subanalysis of BENEFIT 2006 trial and open label extension (5 years) study to evaluate cognitive performance in subgroups of participants

BENEFIT 2014a

Subanalysis of BENEFIT 2006 trial to evaluate effect of Vitamin‐D on conversion to CDMS

BENEFIT 2014b

Reanalysis of BENEFIT 2006 trial and OLE (5 years) study (BENEFIT 2009 (5 years FU)) to evaluate predictive effect of treatment on persisting T1 hypointensities on MRI

CHAMPIONS 2015

Reanalysis of CHAMPS 2000 trial and OLE (10 years) study (CHAMPS 2012 (10 years FU)) to compare the 10‐year disease progression between subgroups of participants with different baseline MRI characteristics (low and higher T2 lesion counts)

CHAMPS 2001

Subanalysis of CHAMPS 2000 trial in participants with a first clinical attack of optic neuritis

CHAMPS 2002a

Subanalysis of CHAMPS 2000 trial in participants allocated to placebo group

CHAMPS 2002b

Subanalysis of CHAMPS 2000 trial to evaluate predictive effect of baseline characteristics on conversion to CDMS

CHAMPS 2002c

Subanalysis of CHAMPS 2000 trial to evaluate prognostic effect of MRI at baseline on conversion to CDMS

CHAMPS 2003

Subanalysis of CHAMPS 2000 trial to evaluate predictive effect of treatment in subgroups of high risk participants

CHAMPS 2009

Reanalysis of CHAMPS 2000 trial to evaluate predictive effect of treatment in subgroups of participants with different baseline risk of disease progression.

Curkendall 2011

Outcomes were not measured. A retrospective study using insurance claims data (2000–2008) of participants with a first clinical attack suggestive of MS. The objective of the study was to assess health care utilisation and expenditures associated with treating participants early with disease‐modifying drugs rather than delaying until participants met the full diagnostic criteria of MS

ETOMS 2003

Subanalysis of ETOMS 2001 trial to evaluate prognostic effect of MRI at baseline on conversion to CDMS

Filippi 2004

Subanalysis of ETOMS 2001 trial to evaluate prognostic effect of MRI at baseline on conversion to CDMS

Kuhle 2015

Cohort study. No treatment with disease‐modifying drugs

Lazzaro 2009

An open cohorts epidemiological model based on demographics of participants enrolled in the BENEFIT 2006 trial. The model arbitrarily started with 2000 CIS participants diagnosed according to Mc Donald criteria, i.e. MS

Meyniel 2012

Prospective cohort study. N = 125 (10%) participants initially treated with disease‐modifying drugs at the first clinical attack suggestive of MS were not reported separately from participants (1094; 88%) who converted to relapsing‐remitting MS at the time of their first disease‐modifying drugs commencement

Moraal 2009

Reanalysis of CHAMPS 2000 trial to assess the prognostic value of baseline MRI for conversion to CDMS over 3 years and the predictive effect of the intervention

Mowry 2009

Prospective cohort study. N = 9 (9%) participants who began disease‐modifying drugs within 1 year of their first clinical attack suggestive of MS were not reported separately from participants (96; 91%) who converted to relapsing‐remitting MS at the time of their first disease‐modifying drugs commencement

MSBASIS 2015

Prospective cohort study, a sub‐study of the MSBase Registry (an international online database on MS). N = 252 (19%) participants who began disease‐modifying drugs at their first clinical attack suggestive of MS were not reported separately from participants (1087; 81%) who converted to relapsing‐remitting MS at the time of their first disease‐modifying drugs commencement

REFLEX 2014a

Subanalysis of REFLEX 2012 trial to evaluate predictive effect of participants' baseline characteristics

REFLEX 2014b

Subanalysis of REFLEX 2012 trial to evaluate prognostic effect of MRI at baseline on conversion to CDMS

SWISS COHORT STUDY 2013

Cohort Study. N = 54 (10%) participants initially treated with disease‐modifying drugs at their first clinical attack suggestive of MS were not reported separately from participants (492; 90%) who converted to relapsing‐remitting MS at the time of their first disease‐modifying drugs commencement

SWISS COHORT STUDY 2016

Cohort study. Outcomes were not measured

CDMS: clinically definite multiple sclerosis; CIS: clinically isolated syndrome; MRI: magnetic resonance imaging; MS: multiple sclerosis; OLE: open‐label extension

Characteristics of ongoing studies [ordered by study ID]

NCT01013350

Trial name or title

Prospective observational long‐term safety registry of multiple sclerosis patients who have participated in cladribine clinical trials (PREMIERE)

Methods

Observational study

Participants

Estimated enrolment: 1190. Subjects with MS and had already participated in sponsor oral cladribine clinical development trials

Interventions

Cladribine

Outcomes

  • Number of participants with serious adverse drug reactions (SADRs)

  • Time to resolution of lymphopenia, among registry participants with persistent lymphopenia

  • Number of participants with all adverse events (adverse events)

Time frame: up to the end of the registry, which is planned for 2018, or 8 years after the participant's first enrolment into a cladribine clinical trial, whichever occurs first

Starting date

November 2009

Contact information

US Medical Information

Notes

ClinicalTrials.gov identifier: NCT01013350

NCT01371071

Trial name or title

Cohort study of clinically isolated syndrome and early multiple sclerosis (CIS‐COHORT)

Methods

Prospective cohort

Participants

Estimated enrolment: 200 participants with CIS within the last 6 months or diagnosis of MS within the last 2 years

Interventions

Immunomodulatory therapy

Outcomes

Primary: time (in days) until relapse during the observation period of 4 years

Starting date

January 2011

Contact information

Prof. Friedemann Paul [email protected]; Dr. Klemens Ruprecht [email protected]

Notes

ClinicalTrials.gov identifier: NCT01371071

CIS: clinically isolated syndrome; MS: multiple sclerosis

Data and analyses

Open in table viewer
Comparison 1. Active intervention versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Occurrence of at least one serious adverse event over 24 months Show forest plot

7

3385

Odds Ratio (M‐H, Random, 95% CI)

0.78 [0.60, 1.03]

Analysis 1.1

Comparison 1 Active intervention versus placebo, Outcome 1 Occurrence of at least one serious adverse event over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 1 Occurrence of at least one serious adverse event over 24 months.

1.1 Interferon beta‐1b (Betaseron) versus placebo

1

468

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.48, 2.11]

1.2 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

0.60 [0.28, 1.27]

1.3 Interferon beta‐1a (Rebif) versus placebo

2

823

Odds Ratio (M‐H, Random, 95% CI)

0.72 [0.35, 1.46]

1.4 Glatiramer acetate versus placebo

1

481

Odds Ratio (M‐H, Random, 95% CI)

0.55 [0.25, 1.17]

1.5 Teriflunomide versus placebo

1

614

Odds Ratio (M‐H, Random, 95% CI)

1.06 [0.59, 1.89]

1.6 Cladribine versus placebo

1

616

Odds Ratio (M‐H, Random, 95% CI)

0.77 [0.43, 1.37]

2 Occurrence of at least one serious adverse event over 36 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Active intervention versus placebo, Outcome 2 Occurrence of at least one serious adverse event over 36 months.

Comparison 1 Active intervention versus placebo, Outcome 2 Occurrence of at least one serious adverse event over 36 months.

2.1 Interferon beta‐1a (Avonex) versus placebo

1

202

Odds Ratio (M‐H, Random, 95% CI)

1.23 [0.44, 3.45]

3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months Show forest plot

5

2693

Odds Ratio (M‐H, Random, 95% CI)

2.43 [0.91, 6.49]

Analysis 1.3

Comparison 1 Active intervention versus placebo, Outcome 3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months.

3.1 Interferon beta‐1b (Betaseron) versus placebo

1

468

Odds Ratio (M‐H, Random, 95% CI)

21.54 [2.92, 159.08]

3.2 Interferon beta‐1a (Rebif) versus placebo

1

514

Odds Ratio (M‐H, Random, 95% CI)

0.73 [0.26, 2.10]

3.3 Glatiramer acetate versus placebo

1

481

Odds Ratio (M‐H, Random, 95% CI)

3.58 [1.16, 11.03]

3.4 Teriflunomide versus placebo

1

614

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.58, 1.81]

3.5 Cladribine versus placebo

1

616

Odds Ratio (M‐H, Random, 95% CI)

4.13 [1.44, 11.87]

4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Active intervention versus placebo, Outcome 4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months.

Comparison 1 Active intervention versus placebo, Outcome 4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months.

4.1 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

0.14 [0.02, 1.12]

5 Time to conversion to CDMS over 24 months Show forest plot

9

Hazard Ratio (Random, 95% CI)

0.53 [0.47, 0.60]

Analysis 1.5

Comparison 1 Active intervention versus placebo, Outcome 5 Time to conversion to CDMS over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 5 Time to conversion to CDMS over 24 months.

5.1 Interferon beta‐1b (Betaseron) versus placebo

1

Hazard Ratio (Random, 95% CI)

0.50 [0.36, 0.69]

5.2 Interferon beta‐1a (Avonex) versus placebo

1

Hazard Ratio (Random, 95% CI)

0.56 [0.38, 0.83]

5.3 Interferon beta‐1a (Rebif) versus placebo

2

Hazard Ratio (Random, 95% CI)

0.57 [0.43, 0.77]

5.4 Glatiramer acetate versus placebo

1

Hazard Ratio (Random, 95% CI)

0.55 [0.40, 0.76]

5.5 Teriflunomide versus placebo

1

Hazard Ratio (Random, 95% CI)

0.57 [0.38, 0.86]

5.6 Cladribine versus placebo

1

Hazard Ratio (Random, 95% CI)

0.38 [0.25, 0.58]

5.7 Any DMD vs no treatment

2

Hazard Ratio (Random, 95% CI)

0.48 [0.30, 0.78]

6 Time to conversion to CDMS over 12 months Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Active intervention versus placebo, Outcome 6 Time to conversion to CDMS over 12 months.

Comparison 1 Active intervention versus placebo, Outcome 6 Time to conversion to CDMS over 12 months.

6.1 Immunoglobulins versus placebo

1

Hazard Ratio (Random, 95% CI)

0.36 [0.15, 0.86]

7 Withdrawing from the study or discontinuing the drug for any reason over 24 months Show forest plot

6

2931

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.61, 1.62]

Analysis 1.7

Comparison 1 Active intervention versus placebo, Outcome 7 Withdrawing from the study or discontinuing the drug for any reason over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 7 Withdrawing from the study or discontinuing the drug for any reason over 24 months.

7.1 Interferon beta‐1b (Betaseron) versus placebo

1

487

Odds Ratio (M‐H, Random, 95% CI)

1.50 [0.95, 2.35]

7.2 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

1.09 [0.65, 1.81]

7.3 Interferon beta‐1a (Rebif) versus placebo

2

826

Odds Ratio (M‐H, Random, 95% CI)

0.52 [0.18, 1.44]

7.4 Teriflunomide versus placebo

1

618

Odds Ratio (M‐H, Random, 95% CI)

0.87 [0.59, 1.27]

7.5 Cladribine versus placebo

1

617

Odds Ratio (M‐H, Random, 95% CI)

2.30 [1.49, 3.56]

8 Withdrawing from the study or discontinuing the drug for any reason over 12 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Active intervention versus placebo, Outcome 8 Withdrawing from the study or discontinuing the drug for any reason over 12 months.

Comparison 1 Active intervention versus placebo, Outcome 8 Withdrawing from the study or discontinuing the drug for any reason over 12 months.

8.1 Immunoglobulins versus placebo

1

91

Odds Ratio (M‐H, Random, 95% CI)

2.15 [0.37, 12.35]

Open in table viewer
Comparison 2. Early versus delayed treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Time to conversion to CDMS at different follow‐up years Show forest plot

10

Hazard Ratio (Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Early versus delayed treatment, Outcome 1 Time to conversion to CDMS at different follow‐up years.

Comparison 2 Early versus delayed treatment, Outcome 1 Time to conversion to CDMS at different follow‐up years.

1.1 2‐4 years' follow‐up

5

Hazard Ratio (Random, 95% CI)

0.62 [0.48, 0.81]

1.2 5 years' follow‐up

4

Hazard Ratio (Random, 95% CI)

0.62 [0.53, 0.73]

1.3 8.7‐10 years' follow‐up

2

Hazard Ratio (Random, 95% CI)

0.65 [0.54, 0.79]

Study flow diagram.
 DMD: disease‐modifying drugs; OLEs: open label extension studies; RCTs: randomised controlled trials
Figures and Tables -
Figure 1

Study flow diagram.
DMD: disease‐modifying drugs; OLEs: open label extension studies; RCTs: randomised controlled trials

Review authors' judgements about each risk of bias item presented as percentages across all included studies and review authors' judgements about each risk of bias item for each included study
Figures and Tables -
Figure 2

Review authors' judgements about each risk of bias item presented as percentages across all included studies and review authors' judgements about each risk of bias item for each included study

Forest plot of comparison: treatment with disease‐modifying drugs compared with placebo. Random‐effects meta‐analysis results of proportion of participants with disability‐worsening over 24 months in RCT studies. We assumed in both groups that the odds of disability‐worsening in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7
Figures and Tables -
Figure 3

Forest plot of comparison: treatment with disease‐modifying drugs compared with placebo. Random‐effects meta‐analysis results of proportion of participants with disability‐worsening over 24 months in RCT studies. We assumed in both groups that the odds of disability‐worsening in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7

Network plot of comparisons and network meta‐analysis estimates for the proportion of participants who withdrew from the study because of adverse events in RCT studies. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a OR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a OR value larger 1 favours the row‐defining treatment. Significant results are in italic
Figures and Tables -
Figure 4

Network plot of comparisons and network meta‐analysis estimates for the proportion of participants who withdrew from the study because of adverse events in RCT studies. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a OR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a OR value larger 1 favours the row‐defining treatment. Significant results are in italic

Network plot of comparisons and network meta‐analysis estimates for the time to conversion to CDMS in RCT studies over 24 months. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a HR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a HR value larger than 1 favours the row‐defining treatment. Significant results are in italic
Figures and Tables -
Figure 5

Network plot of comparisons and network meta‐analysis estimates for the time to conversion to CDMS in RCT studies over 24 months. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a HR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a HR value larger than 1 favours the row‐defining treatment. Significant results are in italic

Network plot of comparisons and network meta‐analysis estimates for the proportion of participants who discontinued treatment and were followed up to the end of the study or who were lost to follow‐up for any reason in RCT studies. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a HR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a HR value larger 1 favours the row‐defining treatment
Figures and Tables -
Figure 6

Network plot of comparisons and network meta‐analysis estimates for the proportion of participants who discontinued treatment and were followed up to the end of the study or who were lost to follow‐up for any reason in RCT studies. The estimate is located at the intersection of the column‐defining treatment and the row‐defining treatment. In the lower triangle the comparisons should be read from left to right, a HR value less than 1 favours the column‐defining treatment. In the upper triangle the comparisons should be read from right to left, a HR value larger 1 favours the row‐defining treatment

Forest plot of comparison: early treatment compared with delayed treatment with disease‐modifying drugs. Random‐effects meta‐analysis results of proportions of participants with disability‐worsening at a maximum of 3 years, 5 years and 10 years of follow‐up in open‐label extension studies. We assumed in both groups that the odds of disability‐worsening in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7
Figures and Tables -
Figure 7

Forest plot of comparison: early treatment compared with delayed treatment with disease‐modifying drugs. Random‐effects meta‐analysis results of proportions of participants with disability‐worsening at a maximum of 3 years, 5 years and 10 years of follow‐up in open‐label extension studies. We assumed in both groups that the odds of disability‐worsening in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7

Forest plot of comparison: Early treatment compared with delayed treatment with disease‐modifying drugs. Random‐effects meta‐analysis results for proportion of participants with relapse over 5 years follow‐up in OLE studies. We assumed in both groups that the odds of relapses in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7
Figures and Tables -
Figure 8

Forest plot of comparison: Early treatment compared with delayed treatment with disease‐modifying drugs. Random‐effects meta‐analysis results for proportion of participants with relapse over 5 years follow‐up in OLE studies. We assumed in both groups that the odds of relapses in missing participants were 5.95 times the odds in the observed participants with 95% CI from 3 to 7

Comparison 1 Active intervention versus placebo, Outcome 1 Occurrence of at least one serious adverse event over 24 months.
Figures and Tables -
Analysis 1.1

Comparison 1 Active intervention versus placebo, Outcome 1 Occurrence of at least one serious adverse event over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 2 Occurrence of at least one serious adverse event over 36 months.
Figures and Tables -
Analysis 1.2

Comparison 1 Active intervention versus placebo, Outcome 2 Occurrence of at least one serious adverse event over 36 months.

Comparison 1 Active intervention versus placebo, Outcome 3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months.
Figures and Tables -
Analysis 1.3

Comparison 1 Active intervention versus placebo, Outcome 3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months.
Figures and Tables -
Analysis 1.4

Comparison 1 Active intervention versus placebo, Outcome 4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months.

Comparison 1 Active intervention versus placebo, Outcome 5 Time to conversion to CDMS over 24 months.
Figures and Tables -
Analysis 1.5

Comparison 1 Active intervention versus placebo, Outcome 5 Time to conversion to CDMS over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 6 Time to conversion to CDMS over 12 months.
Figures and Tables -
Analysis 1.6

Comparison 1 Active intervention versus placebo, Outcome 6 Time to conversion to CDMS over 12 months.

Comparison 1 Active intervention versus placebo, Outcome 7 Withdrawing from the study or discontinuing the drug for any reason over 24 months.
Figures and Tables -
Analysis 1.7

Comparison 1 Active intervention versus placebo, Outcome 7 Withdrawing from the study or discontinuing the drug for any reason over 24 months.

Comparison 1 Active intervention versus placebo, Outcome 8 Withdrawing from the study or discontinuing the drug for any reason over 12 months.
Figures and Tables -
Analysis 1.8

Comparison 1 Active intervention versus placebo, Outcome 8 Withdrawing from the study or discontinuing the drug for any reason over 12 months.

Comparison 2 Early versus delayed treatment, Outcome 1 Time to conversion to CDMS at different follow‐up years.
Figures and Tables -
Analysis 2.1

Comparison 2 Early versus delayed treatment, Outcome 1 Time to conversion to CDMS at different follow‐up years.

Summary of findings for the main comparison. Are disease‐modifying drugs for a first attack suggestive of multiple sclerosis (MS) effective and safe compared to placebo?

Patient: adults with first attack suggestive of MS
Setting: MS centres
Intervention: early disease‐modifying drug treatment
Comparison: placebo

Outcomes

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Quality of the evidence
(GRADE)

What happens

With placebo

With early disease‐modifying drugs treatment

Difference

Disability‐worsening

Proportion of participants with disability‐worsening, assessed by EDSS** during 24 months of treatment
Participants: N = 927
(2 RCTs)

OR 0.74
(0.49 to 1.14)

34.1%

27.7%

(20.2 to 37.1)

6.4% fewer (13.9 fewer to 3 more)

⊕⊝⊝⊝
Very lowa,b,c

The risk of disability‐worsening is less with disease‐modifying drugs than with placebo, but there is a lot of uncertainty in the effect

Relapse

Proportion of participants with relapse during 24 months of treatment
Participants: N = 618
(1 RCT)

OR 0.65
(0.38 to 1.12)

41.6%

31.7%

(21.3 to 44.4)

10.0% fewer (20.3 fewer to 2.8 more)

⊕⊝⊝⊝
Very lowa,c,d

The risk of relapse is less with disease‐modifying drugs than with placebo, but there is a lot of uncertainty in the effects

Occurrence of at least one serious adverse event Proportion of participants with at least one serious adverse event during 24 months of treatment
Participants: N = 3385
(7 RCTs)

OR 0.78
(0.60 to 1.03)

8.0%

6.3%
(5.0 to 8.2)

1.6% fewer
(3 fewer to 0.2 more)

⊕⊕⊝⊝
Lowa,e

Compared to placebo, disease‐modifying drugs were associated with less risk of serious adverse events

Withdrawls or drug discontinuation due to adverse events

during 24 months of treatment
Participants: N = 2693
(5 RCTs)

OR 2.43
(0.91 to 6.49)

3.5%

8.0%
(3.2 to 18.9)

4.6% more
(0.3 fewer to 15.4 more)

⊕⊝⊝⊝
Very lowa,f,g

Compared to placebo interferon beta 1‐b, glatiramer acetate, and cladribine were associated with higher risk of withdrawals due to adverse events

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

**EDSS: expanded disability status scale
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

aHigh risk of bias for blinding of participants and outcome assessment and incomplete outcome data.
bSurrogate outcome in both studies contributing to this estimate.
cThe confidence interval does not rule out a null effect or benefit.
dOnly one study contributed to this estimate.
eDefinition and methods of monitoring and detecting serious adverse events not reported in most trials.
fHigh heterogeneity (I² = 78%, P = 0.001) not explained; high subgroup differences (I² = 75%, P = 0.003).
gDefinition and methods of monitoring and detecting adverse events not reported in most trials.

Figures and Tables -
Summary of findings for the main comparison. Are disease‐modifying drugs for a first attack suggestive of multiple sclerosis (MS) effective and safe compared to placebo?
Summary of findings 2. Is early treatment with disease‐modifying drugs more efficacious and safer than delayed treatment?

Patient: adults with first attack suggestive of MS
Setting: MS centres
Intervention: early disease‐modifying drug treatment
Comparison: delayed disease‐modifying drug treatment; after the second attack or diagnosis with clinically definitive MS

Outcomes

Relative effect
(95% CI)

Anticipated absolute effects* (95% CI)

Quality of the evidence
(GRADE)

What happens

Without early disease‐modifying drug treatment

With early disease‐modifying drug treatment

Difference

Disability‐worsening

Proportion of participants with disability‐worsening at a maximum of five years' follow‐up (assessed by EDSS**)
Participants: N = 1868
(4 open‐label extension studies)

OR 0.88
(0.50 to 1.57)

40.2%

37.2%
(25.2 to 51.4)

3.0% fewer
(15 fewer to 11.1 more)

⊕⊝⊝⊝
Very lowa,b,c, d

No significant effect of early treatment compared to delayed treatment during five years' follow‐up; however there is a significant heterogeneity between the studies

Relapse

Proportion of participants with relapse at a maximum of five years' follow‐up
Participants: N = 1485
(3 open‐label extension studies)

OR 0.35
(0.26 to 0.48)

83.4%

63.8%
(56.7 to 70.7)

19.6% fewer
(26.7 fewer to 12.7 fewer)

⊕⊕⊝⊝
Lowa

Early treatment reduced the risk of relapses compared to delayed treatment during five years' follow‐up

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
**EDSS: expanded disability status scale
CI: Confidence interval; OR: Odds ratio

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

aHigh risk of bias for allocation concealment, blinding of outcome assessment and incomplete outcome data.
bSurrogate outcome in two out of four studies contributing to this estimate.
cHigh heterogeneity (I² = 67%, P = 0.03).
dThe confidence interval fails to exclude important benefit or important harms.

Figures and Tables -
Summary of findings 2. Is early treatment with disease‐modifying drugs more efficacious and safer than delayed treatment?
Table 1. Summary of characteristics of included studies

Type of intervention

Route

RCTs

N = 10

OLEs

N = 8

Cohort studies

N = 4

Interferon beta‐1b sc (Betaseron®)

sc

1

4 OLEs at a maximum follow‐up of 3, 5, 8.7, and 11 years

0

Interferon beta‐1a (Avonex®)

im

2

2 OLEs at a maximum follow‐up of 5 and 10 years

0

Interferon beta‐1a (Rebif®)

sc

3

1 OLE at a maximum follow‐up of 3 and 5 years

0

Glatiramer acetate sc

sc

1

1 OLE at a maximum follow‐up of 5 years

0

Cladribine os

os

1

0

0

Teriflunomide os

os

1

0

0

Immunoglobulins iv

iv

1

0

0

disease‐modifying drugs

0

0

follow‐up from 2 to 6 years

im: intramuscular; iv: intravenously; OLEs: open‐label extension studies; os: oral; RCTs: randomised controlled studies; sc: subcutaneous

Figures and Tables -
Table 1. Summary of characteristics of included studies
Table 2. Risk of bias in included cohort studies (ROBINS‐I)

ACISS 2010

Bias

Authors’ judgment

Support for judgement

Confounding

Serious

All known important domains were not appropriately controlled for

Selection of participants into the study

Low

All participants who would have been eligible for the target trial were likely included in the study and for each participant start of follow up and start of intervention likely coincided

Classification of interventions

Low

Intervention status was well defined and intervention definition was based on information collected at the time of intervention

Deviations from intended interventions

NI

No information was reported on whether there was deviation from the intended intervention

Missing data

Critical

There were critical differences between early, delayed or no treatment in participants with missing data and an appropriate analysis to address missing data was not done

Measurement of outcomes

Serious

The outcome measures were subjective and assessed by assessors aware of the intervention received by study participants. This judgment is applicable to all the three outcomes reported in the article

Selection of the reported result

Low

There was evidence that reported results corresponded to all intended outcomes and analyses

Overall bias

Critical

Study judged to be at critical risk of bias in one domain

GERONIMUS 2013

Bias

Authors’ judgment

Support for judgement

Confounding

Moderate

Confounding expected, all known important confounding domains appropriately measured and controlled for, and reliability and validity of measurement of important domains were sufficient, such that we do not expect serious residual confounding

Selection of participants into the study

Low

All participants who would have been eligible for the target trial were likely included in the study and for each participant start of follow up and start of intervention likely coincided

Classification of interventions

Serious

Intervention status was not well defined

Deviations from intended interventions

NI

No information was reported on whether there was deviation from the intended intervention

Missing data

Low

Data were reasonably complete

Measurement of outcomes

Serious

CDMS was assessed by assessors aware of the intervention received by study participants

Selection of the reported result

Low

There was evidence that reported results corresponded to all intended outcomes and analyses

Overall bias

Serious

Study judged to be at serious risk of bias in two domains, but not at critical risk of bias in any domain

MSBASIS 2016

Bias

Authors’ judgment

Support for judgement

Confounding

Serious

Important domains were not appropriately controlled for

Selection of participants into the study

Critical

Selection into the study was very strongly related to intervention and outcome and this could not be adjusted for in analyses

Classification of interventions

Serious

Intervention status was not well defined

Deviations from intended interventions

NI

No information was reported on whether there was deviation from the intended intervention

Missing data

NI

No information was reported on missing data

Measurement of outcomes

Serious

The outcome measures were subjective and they were assessed by assessors aware of the intervention received by study participants. Follow‐up duration not reported

Selection of the reported result

Low

There was evidence that reported results corresponded to all intended outcomes and analyses

Overall bias

Critical

Study judged to be at critical risk of bias in one domain

Tintore 2015

Bias

Authors’ judgment

Support for judgement

Confounding

Serious

Important domains were not appropriately controlled for

Selection of participants into the study

Low

All participants who would have been eligible for the target trial were likely included in the study and for each participant start of follow up and start of intervention likely coincided

Classification of interventions

Serious

Intervention status was not well defined

Deviations from intended interventions

NI

No information was reported on whether there was deviation from the intended intervention

Missing data

Serious

Reasons for missing data differed substantially across interventions, and the analysis is unlikely to have removed the risk of bias arising from the missing data

Measurement of outcomes

Serious

The outcome measures were subjective and assessed by assessors aware of the intervention received by study participants. This judgment is applicable to all outcomes reported in the article

Selection of the reported result

Low

There was evidence that reported results corresponded to all intended outcomes and analyses

Overall bias

Serious

Study judged to be at serious risk of bias in four domains, but not at critical risk of bias in any domain

ROBINS‐I is a tool to evaluate Risk Of Bias In Non‐randomised Studies ‐ of Interventions (Sterne 2016)

Figures and Tables -
Table 2. Risk of bias in included cohort studies (ROBINS‐I)
Table 3. Assessment of adverse events monitoring, definition and reporting of serious adverse events

Study

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

Did the authors define serious adverse events according to an accepted international classification and report the number of serious adverse events?

Achiron 2004

No information

No information

ACISS 2010

No information

No information

BENEFIT 2006

Yes, active monitoring.“Regular visits were scheduled for safety assessments at months 3, 6, 9, 12, 18, and 24”. (page 1243)

No information

BENEFIT 2007 (3 years FU)

No information

No information

BENEFIT 2009 (5 years FU)

No information

No information

BENEFIT 2014 (8.7 years FU)

No information

No information

BENEFIT 2016 (11 years FU)

No information

No information

CHAMPS 2000

No active monitoring. "Each center was instructed to report all adverse events during the first six months of treatment, but thereafter to report only serious adverse events". (page 899)

No information

CHAMPS 2006 (5 years FU)

No information

No information

CHAMPS 2012 (10 years FU)

No information

No information

ETOMS 2001

Yes, active monitoring. "Safety was assessed at 1, 6, 12, 18, 24 months". (page 1577)

Yes to both questions. "Serious adverse events were defined according to the guidelines of the International Conference on Harmonisation". (page 1580)

GERONIMUS 2013

No information

No information

Motamed 2007

Yes, active monitoring."Safety assessments were performed at the end of months 1, 2, 3, 9, 15, and 21 by a neurologist". (page 345)

No information

MSBASIS 2016

No information

No information

ORACLE 2014

Yes, active monitoring. "Adverse events and laboratory findings were recorded at study visits and at regularly scheduled interim visits" (page 259). "International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use". (page 258)

Yes to both questions. "International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use". (page 258)

Pakdaman 2007

No information

No information

PRECISE 2009

Unclear whether the researchers actively monitored for adverse events or they simply provided spontaneous reporting of adverse events

No information

PRECISE 2013 (5 years FU)

Unclear whether the researchers actively monitored for adverse events or they simply provided spontaneous reporting of adverse events

No information

REFLEX 2012

Yes, active monitoring. "Active monitoring by personnel was ensured via various testing". (page 34). "Adverse events were coded with the Medical Dictionary for Regulatory Activities (MedDRA) and analysed according to the preferred terms". (page 35)

Yes to both questions."Adverse events were coded with the Medical Dictionary for Regulatory Activities (MedDRA) and analysed according to the preferred terms". (page 35)

REFLEX 2016 (3 and 5 years FU)

Unclear. "Adverse events (adverse events) were monitored at months 25 and 27 and then every 3 months to the study end". (page 2)

No information

Tintore 2015

No information

No information

TOPIC 2014

Unclear. "Adverse events were reported by study participants or investigators throughout the study; investigators recorded all such events on case report forms". (page 979)

No information

Figures and Tables -
Table 3. Assessment of adverse events monitoring, definition and reporting of serious adverse events
Table 4. Outcome data from cohort studies

ACISS 2010

Early DMDs treatment (N = 49)

Delayed DMDs treatment (N = 57)

No treatment

(N = 52)

EDSS score over 24 months' follow‐up

Mean (SD)

Median (range)

Kruskal–Wallis H‐Test P value <0.001

1.2 (0.9)

1.5 (0‐3)

1.6 (1.2)

1.5 (0‐6)

0.8 (0.8)

1.0 (0‐3)

P value versus no treatment

0.016

< 0.001

NA

P value early versus delayed treatment (Wilcoxon matched pair test)

0.055

NA

NA

Relapses

Mean (SD)

Median (range)

Kruskal–Wallis H‐Test P value < 0.001

0.5 (0.8)

0.0 (0‐4)

1.0 (1.1)

1.0 (0‐4)

0.2 (0.5)

0.0 (0‐3)

P value versus no treatment

0.059

< 0.001

NA

P value early versus delayed treatment (Wilcoxon matched pair test)

0.01

NA

NA

Tintore 2015

Risk of attaining an EDSS score of 3.0 with early DMDs compared with delayed DMDs treatment.

Adjusted hazard ratio: 0.5 (95% CI 0.3 to 0.9)

Unadjusted hazard ratio: 1.1 (95% CI 0.7 to 1.9)

DMDs: disease‐modifying drugs. EDSS: expanded disability status scale; NA: not applicable; SD: standard deviation

Figures and Tables -
Table 4. Outcome data from cohort studies
Table 5. Time until the delayed treatment in open‐label extension studies

Study

Time until the delayed treatment after randomisation

BENEFIT 2006

Mean (SD): 1.5 (0.73) years

CHAMPS 2000

Median (interquartile range): 30 (24‐35) months

PRECISE 2009

Median (range): 29 (0.5 –38) months

REFLEX 2012

Data not reported

Figures and Tables -
Table 5. Time until the delayed treatment in open‐label extension studies
Table 6. Safety outcome data from open‐label extension studies

Interferon beta‐1b

Intramuscular interferon beta 1‐a (Avonex)

Subcutaneous interferon beta 1‐a (Rebif)

Glatiramer acetate

Participants

487

383

517

481

Serious adverse events ‐ number of participants

123

65

49

60

Discontinued treatment for any adverse events

Not reported

Not reported

20

71

Discontinued treatment or were lost to follow‐up for any reason

204

Not reported

146

192

Years of follow‐up

8.7

10

5

5

Figures and Tables -
Table 6. Safety outcome data from open‐label extension studies
Comparison 1. Active intervention versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Occurrence of at least one serious adverse event over 24 months Show forest plot

7

3385

Odds Ratio (M‐H, Random, 95% CI)

0.78 [0.60, 1.03]

1.1 Interferon beta‐1b (Betaseron) versus placebo

1

468

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.48, 2.11]

1.2 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

0.60 [0.28, 1.27]

1.3 Interferon beta‐1a (Rebif) versus placebo

2

823

Odds Ratio (M‐H, Random, 95% CI)

0.72 [0.35, 1.46]

1.4 Glatiramer acetate versus placebo

1

481

Odds Ratio (M‐H, Random, 95% CI)

0.55 [0.25, 1.17]

1.5 Teriflunomide versus placebo

1

614

Odds Ratio (M‐H, Random, 95% CI)

1.06 [0.59, 1.89]

1.6 Cladribine versus placebo

1

616

Odds Ratio (M‐H, Random, 95% CI)

0.77 [0.43, 1.37]

2 Occurrence of at least one serious adverse event over 36 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Interferon beta‐1a (Avonex) versus placebo

1

202

Odds Ratio (M‐H, Random, 95% CI)

1.23 [0.44, 3.45]

3 Withdrawing from the study or discontinuing the drug due to adverse events over 24 months Show forest plot

5

2693

Odds Ratio (M‐H, Random, 95% CI)

2.43 [0.91, 6.49]

3.1 Interferon beta‐1b (Betaseron) versus placebo

1

468

Odds Ratio (M‐H, Random, 95% CI)

21.54 [2.92, 159.08]

3.2 Interferon beta‐1a (Rebif) versus placebo

1

514

Odds Ratio (M‐H, Random, 95% CI)

0.73 [0.26, 2.10]

3.3 Glatiramer acetate versus placebo

1

481

Odds Ratio (M‐H, Random, 95% CI)

3.58 [1.16, 11.03]

3.4 Teriflunomide versus placebo

1

614

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.58, 1.81]

3.5 Cladribine versus placebo

1

616

Odds Ratio (M‐H, Random, 95% CI)

4.13 [1.44, 11.87]

4 Withdrawing from the study or discontinuing the drug due to adverse events over 12 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

0.14 [0.02, 1.12]

5 Time to conversion to CDMS over 24 months Show forest plot

9

Hazard Ratio (Random, 95% CI)

0.53 [0.47, 0.60]

5.1 Interferon beta‐1b (Betaseron) versus placebo

1

Hazard Ratio (Random, 95% CI)

0.50 [0.36, 0.69]

5.2 Interferon beta‐1a (Avonex) versus placebo

1

Hazard Ratio (Random, 95% CI)

0.56 [0.38, 0.83]

5.3 Interferon beta‐1a (Rebif) versus placebo

2

Hazard Ratio (Random, 95% CI)

0.57 [0.43, 0.77]

5.4 Glatiramer acetate versus placebo

1

Hazard Ratio (Random, 95% CI)

0.55 [0.40, 0.76]

5.5 Teriflunomide versus placebo

1

Hazard Ratio (Random, 95% CI)

0.57 [0.38, 0.86]

5.6 Cladribine versus placebo

1

Hazard Ratio (Random, 95% CI)

0.38 [0.25, 0.58]

5.7 Any DMD vs no treatment

2

Hazard Ratio (Random, 95% CI)

0.48 [0.30, 0.78]

6 Time to conversion to CDMS over 12 months Show forest plot

1

Hazard Ratio (Random, 95% CI)

Subtotals only

6.1 Immunoglobulins versus placebo

1

Hazard Ratio (Random, 95% CI)

0.36 [0.15, 0.86]

7 Withdrawing from the study or discontinuing the drug for any reason over 24 months Show forest plot

6

2931

Odds Ratio (M‐H, Random, 95% CI)

1.00 [0.61, 1.62]

7.1 Interferon beta‐1b (Betaseron) versus placebo

1

487

Odds Ratio (M‐H, Random, 95% CI)

1.50 [0.95, 2.35]

7.2 Interferon beta‐1a (Avonex) versus placebo

1

383

Odds Ratio (M‐H, Random, 95% CI)

1.09 [0.65, 1.81]

7.3 Interferon beta‐1a (Rebif) versus placebo

2

826

Odds Ratio (M‐H, Random, 95% CI)

0.52 [0.18, 1.44]

7.4 Teriflunomide versus placebo

1

618

Odds Ratio (M‐H, Random, 95% CI)

0.87 [0.59, 1.27]

7.5 Cladribine versus placebo

1

617

Odds Ratio (M‐H, Random, 95% CI)

2.30 [1.49, 3.56]

8 Withdrawing from the study or discontinuing the drug for any reason over 12 months Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 Immunoglobulins versus placebo

1

91

Odds Ratio (M‐H, Random, 95% CI)

2.15 [0.37, 12.35]

Figures and Tables -
Comparison 1. Active intervention versus placebo
Comparison 2. Early versus delayed treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Time to conversion to CDMS at different follow‐up years Show forest plot

10

Hazard Ratio (Random, 95% CI)

Subtotals only

1.1 2‐4 years' follow‐up

5

Hazard Ratio (Random, 95% CI)

0.62 [0.48, 0.81]

1.2 5 years' follow‐up

4

Hazard Ratio (Random, 95% CI)

0.62 [0.53, 0.73]

1.3 8.7‐10 years' follow‐up

2

Hazard Ratio (Random, 95% CI)

0.65 [0.54, 0.79]

Figures and Tables -
Comparison 2. Early versus delayed treatment