Scolaris Content Display Scolaris Content Display

Teriflunomid bei Multipler Sklerose

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Confavreux 2014 {published data only}

Comi G, Freedman MS, Kappos L, Miller A, Olsson TP, Wolinsky JS, et al. Effect of teriflunomide on lymphocyte and neutrophil counts in patients with relapsing multiple sclerosis: results from the TOWER study. Journal of the Neurological Sciences 2013;333:e376.
Confavreux C, O'Connor P, Comi G, Freedman MS, Miller AE, Olsson TP, et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Neurology 2014;13(3):247‐56.
Kappos L, Comi G, Confavreux C, Freedman MS, Miller AE, Olsson TP, et al. The efficacy and safety of teriflunomide in patients with relapsing MS: results from TOWER, a phase Ⅲ, placebo‐controlled study. Multiple Sclerosis 2012;18:50‐1.
Miller A, Kappos L, Comi G, Confavreux C, Freedman M, Olsson T, et al. Teriflunomide efficacy and safety in patients with relapsing multiple sclerosis: results from tower, a second, pivotal, phase 3 placebo‐controlled study. Neurology. 2013; Vol. 80.
Miller AE, Macdonell R, Comi G, Freedman MS, Kappos L, Maurer M, et al. Teriflunomide reduces relapses with sequelae and relapses leading to hospitalizations: results from the TOWER study. Journal of Neurology 2014;261(9):1781‐8.
Moses H, Freedman M, Kappos L, Miller A, Olsson T, Wolinsky J, et al. Pre‐defined subgroups analyses of tower, a placebo‐controlled phase 3 trial of teriflunomide in patients with relapsing multiple sclerosis. Proceedings of the American Academy of Neurology's 65th AAN Annual Meeting; 2013 Mar 16‐23; San Diego (CA).
O'Connor P, Lublin F, Wolinsky J, Comi G, Confavreux C, Freedman M, et al. Teriflunomide reduces relapse‐related sequelae, hospitalizations and corticosteroid use: a post‐HOC analysis of the phase 3 TOWER study. Proceedings of The American Academy of Neurology's 65th AAN Annual Meeting; 2013 Mar 16‐23; San Diego (CA).

Freedman 2012 {published data only}

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

NCT01252355 {published data only}

NCT01252355. Efficacy and Safety of Teriflunomide in Patients with Relapsing Multiple Sclerosis and Treated with Interferon‐beta (TERACLES). www.clinicaltrials.gov/ct2/show/results/NCT01252355 (accessed 12 January 2016).

O'Connor 2011 {published data only}

Comi G, Benzerdjeb H, Wang L, Truffinet P, O'Connor P. Effect of teriflunomide on lymphocyte and neutrophil levels in patients with relapsing multiple sclerosis: results from the TEMSO study. Journal of Neurological Science 2013;333(Suppl 1):e376.
Comi G, O'Connor P, Wolinsky J, Confavreux C, Kappos L, Olsson T, et al. Extension of a phase Ⅲ trial (TEMSO) of oral teriflunomide in multiple sclerosis with relapses: safety outcomes with up to 4 years of follow‐Up. Multiple Sclerosis 2011;17:S182‐3.
Freedman M, Wolinsky J, Comi G, Kappos L, Olsson T, Miller A, et al. Long‐term safety and efficacy of teriflunomide in patients with relapsing forms of multiple sclerosis in the TEMSO extension trial. Multiple Sclerosis 2013;19:225.
Freedman M, Wolinsky J, Comi G, Kappos L, Olsson T, Miller A, et al. Safety and efficacy of teriflunomide for up to 9 years in relapsing forms of multiple sclerosis: Update of the TEMSO extension trial. Neurology2014; Vol. 82, issue Suppl 10:3‐150.
Freedman MS, Delhay JL, Benamor M. Gastrointestinal symptoms infrequently lead to discontinuation of teriflunomide therapy. Multiple Sclerosis 2012;18:S13.
Miller A, Lublin F, O'Connor P, Taniou C, Dive‐Pouletty C. Impact of relapses with sequelae on disability, health‐related quality of life, and fatigue in a population with relapsing forms of multiple sclerosis using data from TEMSO, a pivotal phase Ⅲ teriflunomide trial. Neurology 2012;78:P07.082.
Miller A, Lublin F, O'Connor P, Wolinsky J, Comi G, Kappos L, et al. Effect of teriflunomide on relapses with sequelae and relapse leading to hospitalization in a population with relapsing forms of multiple sclerosis: results from the TEMSO study. Neurology 2012;78:S30.003.
Miller A, O'Connor P, Wolinsky J, Confavreux C, Comi G, Kappos L, et al. Efficacy of oral teriflunomide in multiple sclerosis with relapses: cognitive outcomes from a phase Ⅲ trial (TEMSO). Multiple Sclerosis 2011;17:S182.
Miller AE, O'Connor P, Wolinsky JS, Confavreux C, Kappos L, Olsson TP, et al. Pre‐specified subgroup analyses of a placebo‐controlled phase Ⅲ trial (TEMSO) of oral teriflunomide in relapsing multiple sclerosis. Multiple Sclerosis 2012;18(11):1625‐32.
O'Connor P, Briggs A, Carita P, Bego‐Le‐Bagousse G. Impact on health‐related quality of life of teriflunomide treatment by estimating utilities in patients with relapsing multiple sclerosis: results from TEMSO post hoc analysis. Journal of Neurology 2012;259:S107.
O'Connor P, Burrell A, Wang L, Wei L, Carita P, Zhang Q. Effect of teriflunomide on longitudinal patterns of relapse and relapse‐related hospitalization in multiple sclerosis. Multiple Sclerosis 2011;17:S414.
O'Connor P, Lublin F, Wolinsky J, Comi G, Kappos L, Freedman M, et al. Effect of teriflunomide on relapses leading to healthcare resource use: results from the TEMSO study. Multiple Sclerosis 2011;17:S95‐6.
O'Connor P, Wolinsky J, Confavreux C, Comi G, Kappos L, Olsson T, et al. Extension of a phase Ⅲ trial (TEMSO) of oral teriflunomide in multiple sclerosis with relapses: clinical and MRI data 5 years after initial randomization. Multiple Sclerosis 2011;17:S414‐5.
O'Connor P, Wolinsky JS, Confavreux C, Comi G, Kappos L, Olsson TP, et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. New England Journal of Medicine 2011;365(14):1293‐303.
O'Connor PW, Lublin FD, Wolinsky JS, Confavreux C, Comi G, Freedman MS, et al. Teriflunomide reduces relapse‐related neurological sequelae, hospitalizations and steroid use. Journal of Neurology 2013;260(10):2472‐80.
Wolinksy JS, Wang L, Truffinet P, Narayana PA, Nelson F, O'Connor P. Magnetic resonance imaging as a surrogate for clinical endpoints in multiple sclerosis. Multiple Sclerosis 2012;18:470‐1.
Wolinsky JS, Narayana PA, Nelson F, Datta S, O'Connor P, Confavreux C, et al. Magnetic resonance imaging outcomes from a phase Ⅲ trial of teriflunomide. Multiple Sclerosis 2013;19(10):1310‐9.

Vermersch 2014 {published data only}

Vermersch P, Czlonkowska A, Grimaldi LM, Confavreux C, Comi G, Kappos L, et al. A multicenter, randomized, parallel‐group, rater‐blinded study comparing the effectiveness and safety of teriflunomide and subcutaneous interferon beta‐1a in patients with relapsing multiple sclerosis. Multiple Sclerosis 2012;18:S9‐10.
Vermersch P, Czlonkowska A, Grimaldi LM, Confavreux C, Comi G, Kappos L, et al. Evaluation of patient satisfaction from the TENERE study: a comparison of teriflunomide and subcutaneous interferon beta‐1a in patients with relapsing multiple sclerosis. Journal of Neurology 2012;259:S38.
Vermersch P, Czlonkowska A, Grimaldi LM, Confavreux C, Comi G, Kappos L, et al. Teriflunomide versus subcutaneous interferon beta‐1a in patients with relapsing multiple sclerosis: a randomised, controlled phase 3 trial. Multiple Sclerosis 2014;20(6):705‐16.

References to studies excluded from this review

Miller 2014 {published data only}

Comi G, Miller AE, Wolinsky JS, Benamor M, Bauer D, Truffinet P, et al. The effect of teriflunomide on lymphocyte and neutrophil count in patients with a first clinical episode consistent with multiple sclerosis: results from the TOPIC study. European Journal of Neurology 2014;21:127‐8.
Miller A, Wolinsky J, Kappos L, Comi G, Freedman M, Olsson T, et al. TOPIC: efficacy and safety of once‐daily oral teriflunomide in patients with first clinical episode consistent with multiple sclerosis. Neurology. 2014; Vol. 82.
Miller A, Wolinsky J, Kappos L, Comi G, Freedman MS, Olsson T, et al. TOPIC main outcomes: efficacy and safety of once‐daily oral teriflunomide in patients with clinically isolated syndrome. Multiple Sclerosis 2013;19:25‐6.
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.
Turner B, Bauer D, Benamor M, Truffinet P, Miller A. Teriflunomide in early stage MS: results from TOPIC. Journal of Neurology, Neurosurgery and Psychiatry 2014;85:A40.
Wolinsky JS, Comi G, Kappos L, Bauer D, Truffinet P, Miller AE. Effect of teriflunomide on MRI activity in patients with early MS: outcomes from the phase 3 TOPIC study. European Journal of Neurology 2014;21:103.
Wolinsky JS, Truffinet P, Bauer D, Miller AE. Efficacy of teriflunomide in patients with early stage MS: analysis of the TOPIC study using 2010 McDonald diagnostic criteria. Multiple Sclerosis 2014;20:109‐10.
Wolinsky JS, Truffinet P, Bauer D, Miller AE. MRI outcomes in patients with early multiple sclerosis treated with teriflunomide: subgroup analyses from the TOPIC phase 3 study. Multiple Sclerosis 2014;20:84.

O'Connor 2006 {published data only}

Confavreux C, Li DK, Freedman MS, Truffinet P, Benzerdjeb H, Wang D, et al. Long‐term follow‐up of a phase 2 study of oral teriflunomide in relapsing multiple sclerosis: safety and efficacy results up to 8.5 years. Multiple Sclerosis 2012;18(9):1278‐89.
Confavreux C, O'Connor P, Freedman M, Benzerdjeb H, Wang D, Bar‐Or A. Long term safety and tolerability of teriflunomide in multiple sclerosis: 9‐year follow‐up of a phase Ⅱ study. Multiple Sclerosis 2011;17:S409‐10.
Jumah MA, Li DB, Yamout B, Truffinet P, Dukovic D, O'Connor PW. Long‐term clinical and magnetic resonance imaging outcomes from patients treated with teriflunomide: results from a phase 2 extension study. Multiple Sclerosis and Related Disorders 2014;3:755‐56.
Li D, O'Connor P, Confavreux C, Truffinet P, Wang D, Traboulsee A. Efficacy of teriflunomide in relapsing multiple sclerosis: phase Ⅱ extension study with 8‐year follow‐up. Multiple Sclerosis 2011;17:S183‐4.
Li DBK, Traboulsee AL, Truffinet P, Dukovic D, O'Connor PW. Long‐term MRI outcomes from patients treated with teriflunomide: results from a phase 2 extension study. Multiple Sclerosis 2014;20:102‐3.
O'Connor PW, Li D, Freedman MS, Bar‐Or A, Rice GP, Confavreux C, et al. A Phase Ⅱ study of the safety and efficacy of teriflunomide in multiple sclerosis with relapses. Neurology 2006;66(6):894‐900.

Bruneau 1998

Bruneau JM, Yea CM, Spinella‐Jaegle S, Fudali C, Woodward K, Robson PA, et al. Purification of human dihydro‐orotate dehydrogenase and its inhibition by A77 1726, the active metabolite of leflunomide. Biochemical Journal 1998;336(Pt 2):299‐303.

Cella 1996

Cella DF, Dineen K, Arnason B, Reder A, Webster KA, Karabatsos G, et al. Validation of the functional assessment of multiple sclerosis quality of life instrument. Neurology 1996;47(1):129‐39.

Cherwinski 1995

Cherwinski HM, McCarley D, Schatzman R, Devens B, Ransom JT. The immunosuppressant leflunomide inhibits lymphocyte progression through cell cycle by a novel mechanism. Journal of Pharmacology and Experimental Therapeutics 1995;272(1):460‐8.

Confavreux 2006

Confavreux C, Vukusic S. Natural history of multiple sclerosis: a unifying concept. Brain 2006;129(Pt 3):606‐16.

Deage 1998

Deage V, Burger D, Dayer JM. Exposure of T lymphocytes to leflunomide but not to dexamethasone favors the production by monocytic cells of interleukin‐1 receptor antagonist and the tissue‐inhibitor of metalloproteinases‐1 over that of interleukin‐1beta and metalloproteinases. European Cytokine Network 1998;9(4):663‐8.

DerSimonian 1986

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

Fischer 1999

Fischer JS, LaRocca NG, Miller DM, Ritvo PG, Andrews H, Paty D. Recent developments in the assessment of quality of life in multiple sclerosis (MS). Multiple Sclerosis 1999;5(4):251‐9.

Greene 1995

Greene S, Watanabe K, Braatz‐Trulson J, Lou L. Inhibition of dihydroorotate dehydrogenase by the immunosuppressive agent leflunomide. Biochemical Pharmacology 1995;50(6):861‐7.

Greenland 1985

Greenland S, Robins JM. Estimation of a common effect parameter from sparse follow‐up data. Biometrics 1985;41(1):55‐68.

Hadjigeorgiou 2013

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

Hamilton 1999

Hamilton LC, Vojnovic I, Warner TD. A771726, the active metabolite of leflunomide, directly inhibits the activity of cyclo‐oxygenase‐2 in vitro and in vivo in a substrate‐sensitive manner. British Journal of Pharmacology 1999;127(7):1589‐96.

Higgins 2011

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

Karampampa 2012

Karampampa K, Gustavsson A, Miltenburger C, Kindundu CM, Selchen DH. Treatment experience, burden, and unmet needs (TRIBUNE) in multiple sclerosis: the costs and utilities of MS patients in Canada. Journal of Population Therapeutics and Clinical Pharmacology 2012;19(1):11‐25.

Korn 2004

Korn T, Magnus T, Toyka K, Jung S. Modulation of effector cell functions in experimental autoimmune encephalomyelitis by leflunomide ‐ mechanisms independent of pyrimidine depletion. Journal of Leukocyte Biology 2004;76:950‐60.

Kurtzke 1983

Kurtzke JF. Rating neurological impairment in multiple sclerosis: an Expanded Disability Status Scale (EDSS). Neurology 1983;33:1444‐52.

Lublin 1996

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

Lublin 2014a

Lublin FD, Reingold SC, Cohen JA, Cutter GR, Sørensen PS, Thompson AJ. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology 2014;83(3):278‐86.

Lublin 2014b

Lublin FD. New multiple sclerosis phenotypic classification. European Neurology 2014;72 Suppl:1‐5.

Manna 1999

Manna SK, Aggarwal BB. Immunosuppressive leflunomide metabolite (A77 1726) blocks TNF‐dependent nuclear factor‐kappa B activation and gene expression. Journal of Immunology 1999;162(4):2095‐102.

Mantel 1959

Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. Journal of the National Cancer Institute 1959;22(4):719‐48.

McDonald 2001

McDonald WI, Compston A, Edan G, Goodkin D, Hartung HP, Lublin FD, et al. Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis. Annals of Neurology 2001;50(1):121‐7.

Merrill 2009

Merrill JE, Hanak S, Pu SF, Liang J, Dang C, Iglesias‐Bregna D, et al. Teriflunomide reduces behavioral, electrophysiological, and histopathological deficits in the Dark Agouti rat model of experimental autoimmune encephalomyelitis. Journal of Neurology 2009;256(1):89‐103.

O'Connell 2014

O'Connell K, Kelly SB, Fogarty E, Duggan M, Buckley L, Hutchinson M, et al. Economic costs associated with an MS relapse. Multiple Sclerosis and Related Disorders 2014;3(6):678‐83.

Oleen‐Burkey 2012

Oleen‐Burkey M, Castelli‐Haley J, Lage MJ, Johnson KP. Burden of a multiple sclerosis relapse: the patients perspective. Patient 2012;5(1):57‐69.

Parisé 2013

Parisé H, Laliberté F, Lefebvre P, Duh MS, Kim E, Agashivala N, et al. Direct and indirect cost burden associated with multiple sclerosis relapses: excess costs of persons with MS and their spouse caregivers. Journal of the Neurological Sciences 2013;330(1‐2):71‐7.

Polman 2005

Polman CH, Reingold SC, Edan G, Filippi M, Hartung HP, Kappos L, et al. Diagnostic criteria for multiple sclerosis: 2005 revisions to the "McDonald Criteria". Annals of Neurology 2005;58(6):840‐6.

Polman 2011

Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Annals of Neurology 2011;69(2):292‐302.

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(3):227‐31.

RevMan 2015 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2015.

Siemasko 1998

Siemasko K, Chong AS, Jäck HM, Gong H, Williams JW, Finnegan A. Inhibition of JAK3 and STAT6 tyrosine phosphorylation by the immunosuppressive drug leflunomide leads to a block in IgG1 production. Journal of Immunology 1998;160(4):1581‐8.

Tallantyre 2008

Tallantyre E, Evangelou N, Constantinescu CS. Spotlight on teriflunomide. International MS Journal 2008;15(2):62‐8.

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]

Tullman 2004

Tullman MJ, Oshinsky RJ, Lublin FD, Cutter GR. Clinical characteristics of progressive relapsing multiple sclerosis. Multiple Sclerosis 2004;10(4):451‐4.

Vickrey 1995

Vickrey BG, Hays RD, Harooni R, Myers LW, Ellison GW. A health‐related quality of life measure for multiple sclerosis. Quality of Life Research 1995;4(3):187‐206.

Ware 1992

Ware JE, Sherbourne CD. The MOS 36‐item short‐form health survey (SF‐36). I. Conceptual framework and item selection. Medical Care 1992;30(6):473‐83.

Xu 1995

Xu X, Williams JW, Bremer EG, Finnegan A, Chong AS. Inhibition of protein tyrosine phosphorylation in T cells by a novel immunosuppressive agent, leflunomide. Journal of Biological Chemistry 1995;270(21):12398‐403.

Xu 1996

Xu X, Williams JW, Gong H, Finnegan A, Chong AS. Two activities of the immunosuppressive metabolite of leflunomide, A77 1726. Inhibition of pyrimidine nucleotide synthesis and protein tyrosine phosphorylation. Biochemical Pharmacology 1996;52(4):527‐34.

Zagmutt 2015

Zagmutt FJ, Carroll CA. Meta‐analysis of adverse events in recent randomized clinical trials for dimethyl fumarate, glatiramer acetate and teriflunomide for the treatment of relapsing forms of multiple sclerosis. International Journal of Neuroscience 2015;125(11):798‐807.

Zeyda 2005

Zeyda M, Poglitsch M, Geyeregger R, Smolen JS, Zlabinger GJ, Hörl WH, et al. Disruption of the interaction of T cells with antigen‐presenting cells by the active leflunomide metabolite teriflunomide: involvement of impaired integrin activation and immunologic synapse formation. Arthritis and Rheumatism 2005;52(9):2730‐9.

References to other published versions of this review

He 2012

He D, Xu Z, Dong S, Zhang H, Zhou H, Wang L, Zhang S. Teriflunomide for multiple sclerosis. Cochrane Database of Systematic Reviews 2012, Issue 12. [DOI: 10.1002/14651858.CD009882.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Confavreux 2014

Methods

Randomized, double‐blind, placebo‐controlled, phase 3 trial (TOWER study)

Population included in data analyses: all randomly assigned participants who received at least 1 dose of study drug or placebo

Did not use the ITT analysis principle

Participants

Inclusion criteria: people aged 18‐55 years; with relapsing MS meeting 2005 McDonald criteria; with or without underlying progression; an EDSS score ≤ 5.5 points; at least 1 relapse in the previous year or at least 2 relapses in the previous 2 years, and no relapse in the 30 days before randomization

Key exclusion criteria: people with other relevant diseases; pregnant, breastfeeding or planning to conceive or father a child during the study; previously or concomitantly receiving cytokine therapy, IFNβ or glatiramer acetate within 3 months of randomization; ever using natalizumab or other immunosuppressive agents

1169 participants were randomly assigned in a 1:1:1 ratio to receive 3 different interventions. 1 participant in placebo group, 1 participant in teriflunomide 7 mg/day group and 2 participants in teriflunomide 14 mg/day group did not receive treatment

Baseline demographic and clinical characteristics were generally well balanced among the study groups
Summary of participant characteristics at baseline (placebo: G1 (n = 389), teriflunomide 7 mg/day: G2 (n = 408), teriflunomide 14 mg/day: G3 (n = 372)

Age (mean ± SD): G1 = 38.1 ± 9.1 years, G2 = 37.4 ± 9.4 years, G3 = 38.2 ± 9.4 years

Women: G1 = 273 (70%), G2 = 300 (74%), G3 = 258 (69%)

Race: white G1 = 318 (82%), G2 = 329 (81%), G3 = 313 (84%); Asian G1 = 60 (15%), G2 = 60 (15%), G3 = 49 (13%); black G1 = 7 (2%), G2 = 8 (2%), G3 = 7 (2%); other G1 = 4 (1%), G2 = 11 (3%), G3 = 3 (1%)

Region: Western Europe and Tunisia G1 = 121 (31%), G2 = 127 (31%), G3 = 120 (32%); Eastern Europe G1 = 117 (30%), G2 = 124 (30%), G3 = 116 (31%); America G1 = 84 (22%), G2 = 92 (23%), G3 = 81 (22%); Asia and Australia G1 = 67 (17%), G2 = 65 (16%), G3 = 55 (15%)

Time from first symptoms of MS (mean ± SD): G1 = 7.64 ± 6.70 years, G2 = 8.18 ± 6.75 years, G3 = 8.18 ± 6.73 years

Time since most recent relapse onset (mean ± SD): G1 = 5.29 ± 3.41 months, G2 = 5.18 ± 3.41 months, G3 = 5.33 ± 3.32 months

Relapses per participant: within previous year (mean ± SD): G1 = 1.4 ± 0.8, G2 = 1.4 ± 0.7, G3 = 1.4 ± 0.7; within previous 2 years: G1 = 2.1 ± 1.1, G2 = 2.1 ± 1.1, G3 = 2.1 ± 1.2

MS subtype: relapsing‐remitting G1 = 379 (97%), G2 = 393 (96%), G3 = 366 (99%); secondary progressive G1 = 4 (1%), G2 = 3 (1%), G3 = 2 (1%); progressive relapsing G1 = 6 (2%), G2 = 12 (3%), G3 = 2 (1%)

Use of MS medication in the previous 2 years: G1 = 135 (35%), G2 = 123 (30%), G3 = 126 (34%); IFNβ‐1a G1 = 59 (15%), G2 = 63 (15%), G3 = 64 (17%); glatiramer acetate G1 = 52 (13%), G2 = 47 (12%), G3 = 37 (10%); IFNβ‐1b G1 = 38 (10%), G2 = 27 (7%), G3 = 35 (9%)

EDSS total score (mean ± SD): G1 = 2.69 ± 1.36, G2 = 2.71 ± 1.39, G3 = 2.71 ± 1.35

FIS score (mean ± SD): G1 = 54.67 ± 37.89, G2 = 56.16 ± 38.20, G3 = 55.25 ± 38.26

Interventions

Experimental group 1: oral teriflunomide 7 mg once daily (n = 408)

Experimental group 2: oral teriflunomide 14 mg once daily (n = 372)

Control group: matching oral placebo once daily (n = 389)

Core treatment period: 48‐152 weeks (maximum treatment duration 173 weeks). Treatment duration was variable, ending 48 weeks after the last participant was included. The median duration of study treatment was similar across all groups (581 days (IQR 392‐756) in the placebo group vs. 556 days (IQR 385‐749) in the teriflunomide 7 mg group vs. 588 days (IQR 351‐765) in the teriflunomide 14 mg group)

Outcomes

Primary outcome:

  • annualized relapse rate (number of relapses per participant‐year)

Secondary outcomes:

  • time to 12 week sustained accumulation of disability

  • time to first relapse

  • proportion of participants free from relapses

  • proportion of participants free of accumulation of disability

  • change from baseline in EDSS score at week 48

  • change in FIS at week 48

  • change in SF‐36 scores at week 48

Relapse was defined as new or worsening clinical signs or symptoms lasting at least 24 hours without fever. Protocol‐defined relapses constituted an increase of either 1 point in at least 2 EDSS functional system scores, or 2 points in 1 EDSS functional system score (excluding bowel and bladder function, and cerebral function), or 0.5 points in total EDSS score from a previous clinically stable assessment

Sustained accumulation of disability was defined as an increase from baseline of at least 1 EDSS point (or ≥ 0.5 points when baseline EDSS score was > 5.5 points) that persisted for at least 12 weeks

Notes

The investigators obtained the data and the sponsor (Genzyme, owned by Sanofi‐Aventis) analyzed the data. Both the sponsor and the authors interpreted the data.

ClinicalTrials.gov number: NCT00751881

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "An interactive voice recognition system generated an allocation sequence using a permuted‐block randomisation schedule with stratification according to study site and baseline EDSS score (≤3.5 or > 3.5)"

Comment: sequence generation was adequate

Allocation concealment (selection bias)

Low risk

Quote: "Randomisation was done centrally, via an interactive voice recognition system"

Comment: allocation concealment was adequate

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients, individuals administering the interventions, and those assessing the outcomes were masked to treatment assignment" "The experimental drugs were identical in taste and appearance"

Comment: participants and personnel were blinded to the allocated interventions

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "Those assessing the outcomes were masked to treatment assignment." "A treating neurologist was responsible for assessment of patient eligibility, supervision of administration of study drug or placebo, recording of adverse events, and assessment of relapses." "An examining neurologist, certified in the Neurostatus system for consistent EDSS assessment, 12 assigned EDSS scores at screening, randomisation, and every 12 weeks until the last treatment visit, and on any unscheduled visits for assessment of suspected relapse or disability worsening"

Comment: treating neurologist who recorded adverse events was responsible for assessment of relapses, blinding of relapse assessment was probably not adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

According to the Figure 1 (Trial profile), overall, 348 (29.8%) participants withdrew from study (115 (29.6%) participants in placebo group, 119 (29.2%) participants in teriflunomide 7 mg/day group and 114 (30.6%) participants in teriflunomide 14 mg/day group)

Comment: to a large degree, missing data and reasons were not balanced between groups. The high overall dropout rate of
29.8% over a period of 48 weeks had potential impacts on the results, a high risk of attrition bias existed

Selective reporting (reporting bias)

Low risk

All listed outcomes were reported adequately. Risk of reporting bias was low

Other bias

High risk

Quote: "The study was sponsored by Genzyme." "Data were obtained by the investigators and analysed by the sponsor (Genzyme)," "Interpretation of the data was done by the sponsor and the authors," "4 co‐authors (TB, J‐LD, DD, and PT) of the published paper are all employees of Genzyme"
Comment: conflicts of interest probably existed. There was probably a high risk of bias

Freedman 2012

Methods

Phase 2 multicentre, placebo‐controlled, double‐blind, randomized study

Follow‐up period: 48 weeks

Population included in data analyses: all randomly assigned participants exposed to at least 1 dose of any study medication

Did not use the ITT analysis principle

Participants

Inclusion criteria: people aged 18‐55 years; with a diagnosis of MS as per the 2005 McDonald criteria; with a relapsing clinical course; with or without progression; having a score of ≤ 5.5 on the EDSS; with no relapse for 8 weeks; having a clinically stable condition for 4 weeks pre‐study; receiving a stable dose of IFNβ for at least 26 weeks before screening

Baseline demographic and disease characteristics were well balanced among the groups.

Summary of baseline characteristics of participants in the initial 24‐week study (placebo + IFNβ: G1 (n = 41), teriflunomide 7 mg + IFNβ: G2 (n = 37), teriflunomide 14 mg + INFβ: G3 (n = 38)

Age (mean ± SD): G1 = 39.2 ± 9.0 years, G2 = 41.4 ± 6.8 years, G3 = 39.6 ± 8.1 years

Women: G1 = 31 (75.6%), G2 = 25 (67.6%), G3 = 25 (65.8%)

White: G1 = 40 (97.6%), G2 = 34 (91.9%), G3 = 38 (100%)

MS subtype: relapsing‐remitting: G1 = 38 (92.7%), G2 = 30 (81.1%), G3 = 34 (89.5%); secondary progressive: G1 = 2 (4.9%), G2 = 2 (5.4%), G3 = 3 (7.9%); progressive relapsing: G1 = 1 (2.4%), G2 = 5 (13.5%), G3 = 1 (2.6%)

Number of relapse within the past 12 months (mean ± SD): G1 = 0.9 ± 0.9, G2 = 0.6 ± 0.8, G3 = 0.9 ± 0.8

Proportion of participants with ≥ 1 relapse in the past 12 months: G1 = 58.5%, G2 = 48.6%, G3 = 65.8%

EDSS score: mean ± SD G1 = 2.6 ± 1.3, G2 = 2.4 ± 1.4, G3 = 2.5 ± 1.6; median (range) G1 = 2.5 (0‐5.5), G2 = 2.0 (0‐5.5), G3 = 2.5 (0‐5.5)

Proportion with IFNβ neutralizing antibodies: < 20 titre G1 = 86.5%, G2 = 80.6%, G3 = 87.9%; 20‐640 titre: G1 = 10.8%, G2 = 11.1%, G3 = 9.1%; > 640 titre: G1 = 2.7%, G2 = 8.3%, G3 = 3.0%

The number of T1‐Gd lesions: 0: G1 = 77.5%, G2 = 78.4%, G3 = 78.9%; ≥ 1: G1 = 22.5%, G2 = 21.6%, G3 = 21.1%

Baseline state of IFNβ: high dose: G1 = 68.3%, G2 = 67.6%, G3 = 63.2%; low dose: G1 = 31.7%, G2 = 32.4%, G3 = 36.8%

Interventions

Experimental group 1: teriflunomide 7 mg/day orally added to IFNβ

Experimental group 2: teriflunomide 14 mg/day orally added to IFNβ

Control group: matching placebo orally added to IFNβ

The ongoing IFNβ regimens were IFN‐1a (Avonex; Biogen Idec, Cambridge, MA), 30 g IM once a week (classified as low‐dose), IFNβ‐1a (Rebif; EMD Serono Inc., Rockland, MA), 22 or 44 g SC 3 times per week (classified as low‐dose and high‐dose, respectively), and IFNβ‐1b (Betaseron; Bayer HealthCare Pharmaceuticals Inc, Montville, NJ), 0.25 mg SC every other day (classified as high‐dose)

Outcomes

Primary outcome:

  • safety and tolerability (treatment‐emergent adverse events occurring with an incidence > 10%)

Secondary outcomes:

  • annualized relapse rates

  • MRI activity: the total number of Gd‐enhancing T1 lesions and the total Gd‐enhancing T1 lesion volume per MRI scan

  • burden of disease: least squares mean change from baseline and least squares mean difference from placebo

  • mean number of unique active lesions per scan

  • volume of post‐Gd T1 hypointense lesions (black holes)

  • T2 lesion component volume change from baseline

  • atrophy change from baseline

  • white matter change from baseline

  • grey matter change from baseline

  • Z4 composite score change from baseline (the Z4 composite score integrated quantitative measures of volume of T1‐Gd lesions, burden of disease, volume of T1‐hypointense lesions and the proportion of total intracranial contents segmented as CSF. The Z4 score was defined as the sum of individual Z scores derived from each of these 4 parameters)

A relapse was defined as the appearance of a new clinical sign or symptom or worsening of a previous symptom that persisted for ≥ 24 hours in the absence of fever

Notes

NCT00489489 [24‐week study] and NCT00811395 [24‐week extension]. This study was sponsored by Sanofi‐Aventis

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The authors reported that randomization was stratified by country and IFN regimen (high‐dose or low‐dose). This was an international multicentre clinical trial, the random sequence generation was probably made by software

Allocation concealment (selection bias)

Low risk

The authors did not describe the method of allocation concealment. This was an international multicentre clinical trial, central randomization was probably used

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "A phase II multicenter, placebo controlled, double‐blind, randomised study comparing 2 doses of teriflunomide in patients with relapsing MS receiving a stable dose of IFNβ was conducted"

Comment: IFNβ regimens were diverse. It was not a truly double‐blind, double‐dummy study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "Each occurrence of relapse was confirmed by the treating neurologist, based on objective assessments by an independent evaluator who was blinded to treatment allocation." "Adverse events were reported by the patient or the investigator"

Comment: relapse was confirmed by the treating neurologist, who was also responsible for reporting. Blinding of outcome assessment was probably not adequate

Incomplete outcome data (attrition bias)
All outcomes

High risk

According to the study disposition, 118 participants were randomly assigned. 2 participants (1 in each teriflunomide group) were excluded before treatment because of protocol violations, leaving 116 exposed to study medication for 24 weeks. 86 participants entered the 24‐week extension phase. 75 participants completed 1 year. 43 (36.4%) participants withdrew from study at 48 weeks
Comment: the authors did not report the reasons why some participants were not rolled over into extension. Most of reasons for drop‐outs differed between groups. Missing data and reasons were not carefully recorded and did not balance between groups. The high overall rate of drop‐outs (36.4%) and the unbalanced reasons for drop‐outs contributed to a high risk of attrition bias

Selective reporting (reporting bias)

Low risk

All listed outcomes were reported adequately

Other bias

High risk

Quote: "The study funding was supported by Sanofi‐Aventis"

Comment: conflicts of interest probably existed. There was probably a high risk of bias

NCT01252355

Methods

Phase 3, multicentre, placebo‐controlled, double‐blind, randomized study (TERACLES study)

Treatment duration: 24‐108 weeks

ITT population: all randomized and treated participants. Participants were considered in the treatment group to which they were randomized regardless of the drug they actually received

Participants

Inclusion criteria: aged 18‐55 years; diagnosis of MS by McDonald's criteria; participant with relapsing forms of MS treated with IFNβ; stable dose of IFNβ for at least 6 months prior to randomization; disease activity in the 12 months prior to randomization and after first 3 months of IFNβ treatment (at least 1 relapse supported by EDSS or equivalent neurological examination, or, at least 1 brain or spinal cord MRI with at least 1 T1 Gd‐enhancing lesion)
Exclusion criteria: McDonald's criteria for MS diagnosis not met at time of screening visit; EDSS score > 5.5 at screening visit; not treated with a stable dose of IFNβ for ≥ 6 months prior to randomization or not tolerating IFNβ or not expected to remain on IFNβ for the duration of the study; a relapse within 30 days prior randomization; human immunodeficiency virus‐positive; prior or concomitant use of cladribine, mitoxantrone or other immunosuppressant agents such as azathioprine, cyclophosphamide, ciclosporin, methotrexate, mycophenolate or fingolimod in previous 6 months; prior use in the 3 months preceding randomization of cytokine therapy (except baseline IFNβ), glatiramer acetate or intravenous immunoglobulins, or concomitant use of these treatments; prior or concomitant use of natalizumab (Tysabri®) in previous 6 months; pregnant, breastfeeding or planning to become pregnant during the study

Baseline demographic and clinical characteristics were generally well balanced among the study groups

Summary of baseline characteristics of participants (placebo + IFNβ: G1 (n = 177), teriflunomide 7 mg + IFNβ: G2 (n = 178), teriflunomide 14 mg + INFβ: G3 (n = 179)

Age (mean ± SD): G1 = 38.3 ± 8.9 years, G2 = 38.7 ± 9.5 years, G3 = 37.7 ± 9.2 years

Women: G1 = 113 (63.8%), G2 = 125 (70.2%), G3 = 114 (63.7%)

Region of enrolment: America G1 = 33 (32.8%), G2 = 30 (16.9%), G3 = 37 (20.7%); Western Europe G1 = 86 (48.6%), G2 = 86 (48.3%), G3 = 79 (44.1%); Eastern Europe G1 = 51 (28.8%), G2 = 51 (28.7%), G3 = 56 (31.3%); Asia, Africa and Australia G1 = 7 (4.0%), G2 = 11 (6.2%), G3 = 7 (3.9%)

Time since first diagnosis of MS (mean ± SD): G1 = 7.0 ± 5.6 years, G2 = 6.6 ± 5.6 years, G3 = 6.8 ± 5.9 years

MS subtype: relapsing‐remitting: G1 = 174 (98.3%), G2 = 173 (97.2%), G3 = 175 (97.8%); secondary progressive: G1 = 2 (1.1%), G2 = 3 (1.7%), G3 = 4 (2.2%); progressive relapsing: G1 = 1 (0.6%), G2 = 2 (1.1%), G3 = 0 (0%)

Number of relapse within the past 12 months (median (range)): G1 = 1 (0‐4), G2 = 1 (0‐3), G3 = 1 (0‐4)

Number of relapse within the past 2 years (median (range)): G1 = 2 (0‐6), G2 = 2 (0‐8), G3 = 2 (0‐8)

Time since most recent MS relapse onset) (median (range)): G1 = 5.0 (1.0‐75.0) months, G2 = 5.0 (1.0‐36.0) months, G3 = 4.0 (1.0‐174.0) months

EDSS score (mean ± SD): G1 = 2.67 ± 1.25, G2 = 2.63 ± 1.37, G3 = 2.64 ± 1.18

Dose level of IFNβ based on IVRS: high dose: G1 = 120 (67.8%), G2 = 128 (71.9%), G3 = 120 (67.0%); low dose: G1 = 57 (32.2%), G2 = 50 (28.1%), G3 = 59 (33.0%)

Interventions

Experimental group 1: teriflunomide 7 mg once daily + IFNβ (any of the IFNβ) (n = 178)

Experimental group 2: teriflunomide 14 mg once daily + IFNβ (any of the IFNβ) (n = 179)

Control group: matching placebo (for teriflunomide) once daily + IFNβ (any of the IFNβ) (n = 177)

Outcomes

Primary outcome:

  • annualized relapse rate = total number of confirmed relapses that occurred during the treatment period divided by the total number of participant‐years treated. Each episode of relapse (appearance, or worsening of a clinical symptom that was stable for at least 30 days, that persisted for a minimum of 24 hours in the absence of fever) was confirmed by an increase in EDSS score or Functional System scores

Secondary outcomes:

  • number of Gd‐enhancing T1‐lesions as measured by brain MRI

  • time to 12‐week sustained disability progression as assessed by EDSS

  • volume of Gd‐enhancing T1‐lesions

  • change from baseline in total lesion volume

  • time to first confirmed relapse

  • change from baseline in FIS total score

  • change from baseline in Short Form Generic Health Survey‐36 Items, version 2 Summary Score

  • resource utilization when relapse

  • overview of adverse events

  • liver function

Notes

The recruitment initiated in January 2011 was discontinued in December 2012 following the decision of the Sponsor (Sanofi‐Aventis) to discontinue the study, the common treatment end date was defined as 28 February 2013 (treatment duration 24‐108 weeks)

846 participants were screened at 185 sites in 28 countries

ClinicalTrials.gov number: NCT01252355

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Randomisation was stratified by investigational site and Interferon‐β dose level (high/low)"

Comments: this was an international multicentre clinical trial, the random sequence generation was probably made by software

Allocation concealment (selection bias)

Low risk

Quote: "Assignment to groups was done centrally using an IVRS in a 1:1:1 ratio after confirmation of selection criteria"

Comments: allocation concealment was adequate

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Any of the IFNβ was used, as long as the IFNβ was approved for marketed use in the country where the participant was enrolled

Comments: IFNβ regimens were diverse. It was not a truly double‐blind, double‐dummy study

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Participant, investigator and outcomes assessor were blinded

Comments: unclear whether all outcome assessors were independent of the treating neurologist who recorded and managed adverse events

Incomplete outcome data (attrition bias)
All outcomes

High risk

Study was prematurely terminated by the sponsor. All participants did not complete the study

Comments: high risk of attrition bias existed

Selective reporting (reporting bias)

Low risk

All pre‐set outcomes in the protocol were described in the results

Comments: reporting bias was low

Other bias

High risk

Quote: "The study was sponsored by Sanofi‐Aventis"
Comment: conflicts of interest probably existed. There was probably a high risk of bias

O'Connor 2011

Methods

Phase 3, randomized, double‐blind, placebo‐controlled, parallel‐group study (TEMSO study)

Follow‐up period: 108 weeks

Population included in data analyses: people who underwent randomization and were exposed to study medication for at least 1 day

Did not use the ITT analysis principle

Participants

Inclusion criteria: people aged 18‐55 years; meeting the 2001 McDonald criteria for a diagnosis of MS; with a relapsing clinical course; with or without progression; having a score of ≤ 5.5 on the EDSS; having at least 2 clinical relapses in the previous 2 years or 1 relapse during the preceding year, but no relapses in the 60 days before randomization

Exclusion criteria: people with other systemic diseases; pregnant; planning to conceive during the trial period

All baseline characteristics were well matched among the groups

Summary of participant characteristics at baseline (placebo: G1 (n = 363), teriflunomide 7 mg: G2 (n = 366), teriflunomide 14 mg: G3 (n = 359)

Age (mean ± SD): G1 = 38.4 ± 9.0 years, G2 = 37.4 ± 9.0 years, G3 = 37.8 ± 8.2 years

Women: G1 = 275 (75.8%), G2 = 255 (69.7%), G3 = 225 (71.0%)

White: G1 = 356 (98.3%), G2 = 355 (97.3%), G3 = 347 (96.9%)

Region: Western Europe G1 = 167 (46.0%), G2 = 167 (45.6%), G3 = 170 (47.4%); Eastern Europe G1 = 114 (31.4%), G2 = 116 (31.7%), G3 = 108 (30.1%); Americas G1 = 82 (22.6%), G2 = 83 (22.7%), G3 = 81 (22.6%)

Time from first symptom of MS (mean ± SD): G1 = 8.6 ± 7.1 years, G2 = 8.8 ± 6.8 years, G3 = 8.7 ± 6.7 years

Relapse (mean ± SD): in previous year G1 = 1.4 ± 0.7, G2 = 1.4 ± 0.7, G3 = 1.3 ± 0.7; in previous 2 years G1 = 2.2 ± 1.0, G2 = 2.3 ± 1.2, G3 = 2.2 ± 1.0

MS subtype: relapsing‐remitting G1 = 329 (90.6%), G2 = 333 (91.0%), G3 = 333 (92.8%); secondary progressive G1 = 22 (6.1%), G2 = 17 (4.6%), G3 = 12 (3.3%); progressive relapsing G1 = 12 (3.3%), G2 = 16 (4.4%), G3 = 14 (3.9%)

Use of DMT in previous 2 years: G1 = 90 (24.8%), G2 = 102 (27.9%), G3 = 102 (28.4%); IFNβ‐1a G1 = 58 (16.0%), G2 = 74 (20.2%), G3 = 62 (17.3%); IFNβ‐1b G1 = 18 (5.0%), G2 = 22 (6.0%), G3 = 27 (7.5%); glatiramer acetate G1 = 36 (9.9%), G2 = 23 (6.3%), G3 = 43 (12.0%)

EDSS score (mean ± SD): G1 = 2.68 ± 1.34, G2 = 2.68 ± 1.34, G3 = 2.67 ± 1.24

FIS score (mean ± SD): G1 = 53.2 ± 37.9, G2 = 50.4 ± 35.6, G3 = 50.3 ± 35.9

Total lesion volume on MRI (mean ± SD): G1 = 19.34 ± 18.94 mL, G2 = 20.37 ± 20.59 mL, G3 = 18.08 ± 17.49 mL

Number of participants with Gd‐enhancing lesions: G1 = 137 (38.2%), G2 = 127 (35.3%), G3 = 125 (35.2%)

Number of Gd‐enhancing lesions on T1‐weighted images (mean ± SD): G1 = 1.66 ± 3.55, G2 = 1.50 ± 3.96, G3 = 1.81 ± 5.17

Volume of hypointense lesions on T1‐weighted images (mean ± SD): G1 = 3.26 ± 3.64, G2 = 3.35 ± 3.96, G3 = 2.91 ± 3.25

Brain parenchymal fraction (mean ± SD): G1 = 0.76 ± 0.02, G2 = 0.76 ± 0.02, G3 = 0.76 ± 0.02

Interventions

Experimental group 1: oral teriflunomide 7 mg once daily (n = 366)

Experimental group 2: oral teriflunomide 14 mg once daily (n = 359)

Control group: matching oral placebo once daily (n = 363)

Outcomes

Primary outcome:

  • annualized relapse rate (defined as the number of confirmed relapses per participant‐year). A relapse was defined as the appearance of a new clinical sign or symptom, or clinical worsening of a previous sign or symptom that had been stable for at least 30 days and that persisted for a minimum of 24 hours in the absence of fever

Secondary outcomes:

  • time to 12‐week sustained disability progression. Sustained disability progression was defined as an increase from baseline of at least 1.0 point in the EDSS score (or at least 0.5 points for participants with a baseline EDSS score > 5.5) that persisted for at least 12 weeks

  • total lesion volume

  • number of Gd‐enhancing lesions on T1‐weighted images

  • volume of hypointense lesion components on T1‐weighted images

  • number of unique active lesions (defined as the number of Gd‐enhancing lesions on T1‐weighted images or new or enlarged lesions on T2‐weighted images, without double counting)

  • brain atrophy

  • participant‐reported fatigue, assessed using the FIS

A relapse was defined as the appearance of a new clinical sign or symptom, or clinical worsening of a previous sign or symptom that had been stable for at least 30 days and that persisted for a minimum of 24 hours in the absence of fever. Confirmed relapses required an increase of 1 point in each of 2 EDSS functional‐system scores or of 2 points in 1 EDSS functional‐system score (excluding bowel and bladder function and cerebral function) or an increase of 0.5 points in the EDSS score from the previous clinically stable assessment

Sustained disability progression was defined as an increase from baseline of at least 1.0 point in the EDSS score (or at least 0.5 points for participants with a baseline EDSS score > 5.5) that persisted for at least 12 weeks

Notes

This was the Teriflunomide Multiple Sclerosis Oral (TEMSO) trial, sponsored by Sanofi‐Aventis. The investigators collected the data and the sponsor analysed the data

ClinicalTrials.gov number: NCT00134563

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The authors reported that randomization was stratified according to the baseline EDSS score (≤ 3.5 or > 3.5) and according to trial site, with a block size of 6. We wrote to the principal author for random sequence generation, the principal author described that the IVRS assigned the randomization number from a list that was loaded in the database

Comment: sequence generation was adequate

Allocation concealment (selection bias)

Low risk

The method of allocation concealment was not reported. We wrote to the principal author, the principal author offered central randomization via an IVRS

Comment: allocation concealment was adequate

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Treating neurologist was unaware of treatment assignments but was aware of any side effects that could potentially be related to active therapy"

The principal author described the study medication teriflunomide (7 mg and 14 mg) and placebo were supplied as identical tablets

Comment: participants and personnel were blinded to the allocated interventions

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "A treating neurologist at each site was responsible for recording and managing adverse events, assessing relapses, and monitoring safety assessments." "An independent, specially trained and certified examining neurologist determined all the EDSS scores and performed all assessments of functional systems." "The examining neurologists were unaware of treatment assignments, only the treating neurologist was aware of any side effects that could potentially be related to active therapy"

Comment: treating neurologist who recorded adverse events was responsible for assessment of relapses, blinding of relapse assessment was probably not adequate

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

According to the Figure 1, 104 participants in placebo group, 92 participants in teriflunomide 7 mg group and 96 participants in teriflunomide 14 mg group discontinued study treatment

Quote: "Of the patients who discontinued the study medication prematurely, 31, 22, and 20 patients in the placebo, teriflunomide 7 mg group, and higher‐dose teriflunomide groups, respectively, completed the planned follow‐up"

Comment: overall, 219 (20.1%) participants were lost‐to follow‐up (73 (20.1%) participants in placebo group, 70 (19.1%) participants in teriflunomide 7 mg group, 76 (21.2%) participants in teriflunomide 14 mg group). There was no sufficient information to understand the reasons for study discontinuation and their balance among the 3 groups

Selective reporting (reporting bias)

Low risk

All listed outcomes were reported adequately

Other bias

High risk

Quote: "The study was sponsored by Sanofi‐Aventis." "Data were collected by the investigators and analysed by the sponsor." "3 co‐authors (HB, PT and LW) of the published paper were affiliated to Sanofi‐Aventis"
Comment: conflicts of interest probably existed. There was probably a high risk of bias

Vermersch 2014

Methods

Phase 3, multicentre, parallel‐group, rater‐blinded study (TENERE study)

Population included in data analyses: efficacy analyses were conducted on the ITT population, which included all randomized participants. The safety analysis included all randomized participants exposed to study medication

Participants

Inclusion criteria: people aged≥ 18 years; meeting the 2005 McDonald criteria for a diagnosis of MS; having a relapsing clinical course; with or without progression; with an EDSS score ≤ 5.5 at screening; being relapse free for 30 days prior to randomization

Exclusion criteria: people with prior use of SC IFNβ‐1a, teriflunomide or leflunomide; prior or ongoing use of natalizumab, cladribine, mitoxantrone or other immunosuppressants; use of other interferons, glatiramer acetate, intravenous immunoglobulins or cytokine therapy within 3 months; having other relevant systemic illnesses; being pregnant, breast‐feeding, or both; planning to conceive

Baseline demographics and characteristics were balanced except for a lower DMT use in the past 2 years in the teriflunomide 14 mg group compared with the IFNβ‐1a group

Summary of participant characteristics at baseline (IFNβ‐1a: G1 (n = 104), teriflunomide 7 mg: G2 (n = 109), teriflunomide 14 mg: G3 (n = 111)

Age (mean ± SD): G1 = 37.0 ± 10.6 years, G2 = 35.2 ± 9.2 years, G3 = 36.8 ± 10.3 years

Female: G1 = 71 (68.3%), G2 = 70 (64.2%), G3 = 78 (70.3%)

White: G1 = 104 (100%), G2 = 109 (100%), G3 = 111 (100%)

Region: Eastern Europe G1 = 35 (33.7%), G2 = 39 (35.8%), G3 = 41 (36.9%); Western Europe and Africa G1 = 62 (59.6%), G2 = 62 (56.9%), G3 = 64 (57.7%); Americas G1 = 7 (6.7%), G2 = 8 (7.3%), G3 = 6 (5.4%)

Time since first symptom of MS (mean ± SD): G1 = 7.7 ± 7.6 years, G2 = 7.0 ± 6.9 years, G3 = 6.6 ± 7.6 years

Relapse (mean ± SD): within previous year G1 = 1.2 ± 1.0, G2 = 1.3 ± 0.8, G3 = 1.4 ± 0.8; within previous 2 years G1 = 1.7 ± 1.1, G2 = 1.7 ± 0.9, G3 = 1.7 ± 0.9

MS subtype: relapsing‐remitting G1 = 104 (100%), G2 = 109 (100%), G3 = 108 (97.3%); secondary progressive G1 = 0, G2 = 0, G3 = 1 (0.9%); progressive relapsing G1 = 0, G2 = 0, G3 = 2 (1.8%)

Use of DMT in previous 2 years: G1 = 25 (24.0%), G2 = 23 (21.1%), G3 = 13 (11.7%); IFNβ‐1a G1 = 6 (5.8%), G2 = 6 (5.5%), G3 = 3 (2.7%); IFNβ‐1b G1 = 10 (9.6%), G2 = 9 (8.3%), G3 = 5 (4.5%); glatiramer acetate G1 = 12 (11.5%), G2 = 10 (9.2%), G3 = 7 (6.3%)

Baseline EDSS score (mean ± SD): G1 = 2.0 ± 1.2, G2 = 2.0 ± 1.2, G3 = 2.3 ± 1.4

Baseline FIS score (mean ± SD): G1 = 34.2 ± 32.7, G2 = 39.5 ± 34.8, G3 = 42.5 ± 37.8

Interventions

Experimental group 1: oral teriflunomide 7 mg once daily (n = 109)

Experimental group 2: oral teriflunomide 14 mg once daily (n = 111)

Control group: IFNβ‐1a 44 μg, SC injection 3 times per week (n = 104)

Treatment duration: 48‐115 weeks. The study was completed 48 weeks after the last participant was randomized, resulting in a variable duration of follow‐up. Median duration of exposure was 60.1 weeks in the IFNβ‐1a group, 66.6 weeks in the teriflunomide 7 mg group and 64.2 weeks in the teriflunomide 14 mg group. Median duration of exposure for all treatment groups was 63.6 weeks; actual maximum exposure was 115 weeks in any group

Outcomes

Primary outcome:

  • time to failure, defined as first occurrence of confirmed relapse or permanent treatment discontinuation for any cause

Secondary outcomes:

  • annualized relapse rate (number of confirmed relapses during the treatment period per participant‐year)

  • changes in participant‐reported fatigue (using the FIS)

  • treatment satisfaction (using the Treatment Satisfaction Questionnaire for Medication (TSQM, version 1.4), with domains for effectiveness, adverse effects, convenience and global satisfaction

  • safety and tolerability

Relapse criteria required the appearance of a new clinical sign/symptom or clinical worsening of a previous sign/symptom (previously stable for at least 30 days) that persisted for at least 24 hours without fever

Notes

Study funded by Genzyme, a Sanofi company. Editorial support was provided by Meg Church, Fishawack Communications, Ltd, also funded by Genzyme

ClinicalTrials.gov number: NCT00883337

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

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

Comment: this was an international multicentre clinical trial, the random sequence generation was probably made by software

Allocation concealment (selection bias)

Low risk

Comment: this was an international multicentre clinical trial, central randomization was probably used

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Participants were randomized 1:1:1 to teriflunomide 7 mg or 14 mg (double‐blind) or IFNβ‐1a (open‐label)

Comment: control (IFNβ‐1a) group was open‐label

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "The treating neurologist was responsible for managing adverse events, and relapse and safety assessments, while an examining neurologist scored the Functional Systems and EDSS. The examining neurologist remained blinded to treatment and associated adverse events"

Comment: treating neurologist who managed adverse events was responsible for assessment of relapses, blinding of relapse assessment was probably not adequate

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The number and reasons of drop‐outs from the study were not reported in the published article

Comment: incomplete outcome data were unclear, the risk of attrition bias was unclear

Selective reporting (reporting bias)

Low risk

All listed outcomes were reported adequately

Other bias

High risk

Quote: "This study was funded by Genzyme, a Sanofi company. Editorial support was provided by Meg Church, Fishawack Communications, Ltd, also funded by Genzyme, a Sanofi company"

Comment: conflicts of interest probably existed. There was probably a high risk of bias

CSF: cerebrospinal fluid; DMT: disease‐modifying therapy; EDSS: Expanded Disability Status Scale; FIS: Fatigue Impact Scale; Gd: gadolinium; IFNβ: interferon beta; IM: intramuscular; ITT: intention‐to‐treat; IQR: interquartile range; IVRS: interactive voice response system; MS: multiple sclerosis; MRI: magnetic resonance imaging; n: number of participants; SC: subcutaneous; SD: standard deviation; SF‐36: 36‐item Short Form.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Miller 2014

Participants were diagnosed with a first clinical episode suggestive of MS rather than definite diagnosis of MS

O'Connor 2006

An RCT with a length of follow‐up shorter than 1 year

MS: multiple sclerosis; RCT: randomized controlled trial.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

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

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

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

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

Summary of findings for the main comparison. Teriflunomide compared to placebo for multiple sclerosis

Teriflunomide compared to placebo for multiple sclerosis

Patient or population: people with relapsing multiple sclerosis
Settings: US, Austria, France, Canada, Germany, UK, Sweden, Netherlands, Turkey, Poland, Chile, Ukraine, China, Italy, Australia, etc.
Intervention: teriflunomide at a dose of 14 mg orally once daily
Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Teriflunomide

Proportion of participants with at least 1 relapse at 1 year
Follow‐up: 1 year

394 per 1000

237 per 1000
(189 to 296)

RR 0.60
(0.48 to 0.75)

761
(1 study)

⊕⊕⊝⊝
lowa

This outcome was considered low, because we considered there were very serious limitation in study design and execution. The bias that influenced the validity of the results for this outcome included: the high risks of bias due to unblinded assessments for relapse and conflicts of interest (sensitivity analysis according to a likely‐case scenario showed a robustness for the results of this outcome, we considered that the high attrition bias did not influence the robustness of the results on relapse). Therefore, we downgraded the quality of evidence for this outcome by 2 levels

The proportion of participants with at least 1 relapse at 2 years
Follow‐up: 2 years

545 per 1000

436 per 1000
(376 to 507)

RR 0.80
(0.69 to 0.93)

722
(1 study)

⊕⊕⊝⊝
lowb

This outcome was considered low, because we considered there were very serious limitation in study design and execution. The bias that influenced the validity of the results for this outcome included: the high risks of bias due to unblinded assessments for relapse and conflicts of interest. (Sensitivity analysis according to a likely‐case scenario showed a robustness for the results of this outcome, we considered that the unclear attrition bias did not influence the robustness of the results on relapse.) Therefore, we downgraded the quality of evidence for this outcome by 2 levels

The proportion of participants with disability progression at 1 year
Follow‐up: 1 year

142 per 1000

78 per 1000
(51 to 119)

RR 0.55
(0.36 to 0.84)

761
(1 study)

⊕⊝⊝⊝
very lowc,e

This outcome was considered very low based on the following reasons:

  • we considered there were very serious limitation in study design and execution. The bias that influenced the validity of the results for this outcome included: the high risks of bias due to the high attrition bias and conflicts of interest. Sensitivity analysis according to a likely‐case scenario showed an unsteadiness for the results of this outcome, we considered that the high attrition bias influenced the robustness of the results on progression disability. Therefore, we downgraded the quality of evidence for this outcome by 2 levels

  • This outcome was an indirect outcome because disability progression was confirmed at 3 months of follow‐up. We had serious doubts about directness. Therefore, we downgraded the quality of evidence for this outcome by 1 level

The proportion of participants with disability progression at 2 years
Follow‐up: 2 years

273 per 1000

202 per 1000
(153 to 262)

RR 0.74
(0.56 to 0.96)

722
(1 study)

⊕⊝⊝⊝
very lowd,e

This outcome was considered very low based on the following reasons:

  • We considered there were very serious limitation in study design and execution. The bias that influenced the validity of the results for this outcome included: the high risks of bias due to unclear attrition bias and conflicts of interest. Sensitivity analysis according to a likely‐case scenario showed an unsteadiness for the results of this outcome, we considered that the unclear attrition bias influenced the robustness of the results on progression disability. Therefore, we downgraded the quality of evidence for this outcome by 2 levels

  • This outcome was an indirect outcome because disability progression was confirmed at 3 months of follow‐up. We had serious doubts about directness. Therefore, we downgraded the quality of evidence for this outcome by 1 level

The proportion of participants with diarrhoea at 2 years
Follow‐up: 2 years

89 per 1000

179 per 1000
(120 to 267)

RR 2.01
(1.35 to 3.00)

718
(1 study)

⊕⊕⊕⊝
moderatef

The follow‐up periods were diverse in Confavreux 2014 and O'Connor 2011 (at least 48 weeks (Confavreux 2014) and 108 weeks (O'Connor 2011)). Treatment duration of participants in Confavreux 2014 was variable, ending 48 weeks after the last participant was included (a maximum treatment duration of 173 weeks). Actually, the data on adverse events in Confavreux 2014 were not at 2 years. There was a heterogeneity in follow‐up period between the studies. Therefore, we did not combine the data on adverse events in Confavreux 2014 and O'Connor 2011

The proportion of participants with hair thinning at 2 years
Follow‐up: 2 years

33 per 1000

131 per 1000
(71 to 243)

RR 3.94
(2.13 to 7.30)

718
(1 study)

⊕⊕⊕⊝
moderatef

The follow‐up periods were diverse in Confavreux 2014 and O'Connor 2011 (at least 48 weeks (Confavreux 2014) and 108 weeks (O'Connor 2011)). Treatment duration of participants in Confavreux 2014 was variable, ending 48 weeks after the last participant was included (a maximum treatment duration of 173 weeks). Actually, the data on adverse events in Confavreux 2014 were not at 2 years. There was a heterogeneity in follow‐up period between the studies. Therefore, we did not combine the data on adverse events in Confavreux 2014 and O'Connor 2011

The proportion of participants with elevated ALT levels at 2 years
Follow‐up: 2 years

67 per 1000

143 per 1000
(90 to 226)

RR 2.14
(1.35 to 3.39)

718
(1 study)

⊕⊕⊕⊝
moderatef

The follow‐up periods were diverse in Confavreux 2014 and O'Connor 2011 (at least 48 weeks (Confavreux 2014) and 108 weeks (O'Connor 2011)). Treatment duration of participants in Confavreux 2014 was variable, ending 48 weeks after the last participant was included (a maximum treatment duration of 173 weeks). Actually, the data on adverse events in Confavreux 2014 were not at 2 years. There was a heterogeneity in follow‐up period between the studies. Therefore, we did not combine the data on adverse events in Confavreux 2014 and O'Connor 2011

*The basis for assumed risk is the placebo group risk. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ALT: alanine aminotransferase; CI: confidence interval; RR: risk ratio.

The assumed risk was defined as placebo group risk because only one study was evaluated.

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

a High risks of bias existed in Confavreux 2014 due to unblinded assessments for relapse and conflicts of interest.

b High risks of bias existed in O'Connor 2011 due to unblinded assessments for relapse and conflicts of interest.

c High risks of bias existed in Confavreux 2014 due to effects of the high attrition bias on progression disability and conflicts of interest.

d High risk of bias existed in O'Connor 2011 due to effects of the unclear attrition bias on progression disability and conflicts of interest.

e Serious indirectness existed in Confavreux 2014 or in O'Connor 2011 because disability progression was confirmed at 3 months of follow‐up.

f High risk of bias existed in O'Connor 2011 due to an unclear attrition bias and conflicts of interest.

Figuras y tablas -
Summary of findings for the main comparison. Teriflunomide compared to placebo for multiple sclerosis