Scolaris Content Display Scolaris Content Display

Interferón beta para la esclerosis múltiple progresiva secundaria

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Andersen 2004 (Nordic) {published data only}

Andersen O, Elovaara I, Farkkila M, Hansen HJ, Mellgren SI, Myhr KM, et al. Multicentre, randomised, double blind, placebo controlled, phase III study of weekly, low dose, subcutaneous interferon beta‐1a in secondary progressive multiple sclerosis. Journal of Neurology, Neurosurgery & Psychiatry 2004;75(5):706‐10.
Beiske AG, Naess H, Aarseth JH, Andersen O, Elovaara I, Farkkila M, et al. Health‐related quality of life in secondary progressive multiple sclerosis. Multiple Sclerosis 2007;13(3):386‐92.
Wu X, Dastidar P, Kuusisto H, Ukkonen M, Huhtala H, Elovaara I. Increased disability and MRI lesions after discontinuation of IFN‐b‐1in secondary progressive MS. Acta Neurol Scand 2005;112:242–7.
Wu X, Kuusisto H, Dastidar P, Huhtala H, Nikkari ST, Ukkonen M, et al. Once‐weekly 22lg subcutaneous IFN‐b‐1a in secondary progressive MS: a 3‐year follow‐up study on brain MRI measurements and serum MMP‐9 levels. Acta Neurol Scand 2007;116:43–8.

Cohen 2002 (IMPACT) {published data only}

Balcer LJ, Baier ML, Cohen JA, Kooijmans MF, Sandrock AW, Nano‐Schiavi ML, et al. Contrast letter acuity as a visual component for the Multiple Sclerosis Functional Composite. Neurology 2003;61(10):1367‐73.
Cadavid D, Tang Y, O’Neill G. Responsiveness of the Expanded Disability Status Scale (EDSS) to disease progression and therapeutic intervention in progressive forms of multiple sclerosis. Rev Neurol 2010;51(6):321‐9.
Cohen JA, Cutter GR, Fischer JS, Goodman AD, Heidenreich FR, Jak AJ, et al. Use of the Multiple Sclerosis Functional Composite as an Outcome Measure in a Phase 3 Clinical Trial. Archives of Neurology 2001;58(6):961‐7.
Cohen JA, Cutter GR, Fischer JS, Goodman AD, Heidenreich FR, Kooijmans MF, et al. Benefit of interferon beta‐1a on MSFC progression in secondary progressive MS. Neurology 2002;59(5):679‐87.
Masiello SA Director Office of Compliance and Biologics Quality Center for Biologics Evaluation and Research. CBER 01‐018 Warning Letter. Avonex (Interferon beta‐1a) for SPMS. Biogen Inc; March 29, 2001. FDA 2001 Food and Drug Administration Center for Biologlcs Evaluation and Research (accessed 18 May 2010).
Miller DM, Cohen JA, Kooijmans M, Tsao E, Cutter G, Baier M. Change in clinician‐assessed measures of multiple sclerosis and subject‐reported quality of life: results from the IMPACT study. Multiple Sclerosis 2006;12(2):180‐6.

North American SG 2004 {published data only}

FDA 2001 ‐ Unger, EF MD. Clinical Review of the June 28, 2000 Amendment to Supplemental BLA 98‐0737 ‐ ‐ STN103471 Chiron Corp.; Interferon β‐1b (Betaseron ®.Information covered in this review: June 28, 2000 – major amendment to supplement June 14, 2001 – Responses to FDA Complete Review Letter; Combined analyses of North American and European SPMS Studies. http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/ucm088275.pdf (accessed 6 June 2010).
Kappos L, Weinshenker B, Pozzilli C, Thompson AJ, Dahlke F, Beckmann K, et al. Interferon beta‐1b in secondary progressive MS: a combined analysis of the two trials. Neurology 2004;63(10):1779‐87.
The North American Study Group on Interferon beta‐1b in Secondary Progressive MS. Interferon beta‐1b in secondary progressive MS: Results from a 3‐year controlled. Neurology 2004;63:1788‐95.

SPECTRIMS 2001 {published data only}

Li DK, Zhao GJ, Paty DW, The University of British Columbia, MS/MRI Analysis Research Group, The SPECTRIMS Study Group. Randomized controlled trial of interferon‐beta‐1a in secondary progressive MS: MRI results. Neurology 2001;56(11):1505‐13.
Patten SB, Metz LM. Interferon beta1a and depression in secondary progressive MS: data from the SPECTRIMS Trial. Neurology 2002;59(5):744‐6.
Sandberg‐Wollheim M, Hommes OR, Hughes RA, Paty DW, Abdul‐Ahad AK. Recombinant human interferon beta in the treatment of relapsing‐remitting and secondary progressive multiple sclerosis. Multiple sclerosis (Houndmills, Basingstoke, England) 1995;1 Suppl 1:S48‐50.
Secondary Progressive Efficacy Clinical Trial of Recombinant Interferon‐beta‐1a in MS (SPECTRIMS) Study Group*. Randomized controlled trial of interferon‐beta‐1a in secondary progressive MS Clinical results. Neurology 2001;56:1496–504.
Sormani, MP, Stubinski B, Cornelisse P, Rocak S, Li D, De Stefano N. Magnetic Resonance Active Lesions As Individual‐Level Surrogate for Relapses in Multiple Sclerosis. Multiple Sclerosis   2010 Dec 9 [Epub ahead of print].

The European SG 1998 {published data only}

Anonymous. Interferon beta 1b and secondary progressive multiple sclerosis. Rev Prescr 2000;20(205):243‐5.
Anonymous. Placebo‐controlled multicentre randomised trial of interferon beta‐1 b in treatment of secondary progressive multiple sclerosis. Mezhdunarodnyi Meditsinskii Zhurnal 2000;nr:306‐16.
Barkhof F, van Waesberghe JH, Filippi M, Yousry T, Miller DH, Hahn D, et al. T(1) hypointense lesions in secondary progressive multiple sclerosis: effect of interferon beta‐1b treatment. Brain 2001;124((Pt 7)):1396‐402.
Brex PA, Molyneux PD, Smiddy P, Barkhof F, Filippi M, Yousry TA, et al. The effect of IFNbeta‐1b on the evolution of enhancing lesions in secondary progressive MS. Neurology 2001;57(12):2185‐90.
Brex PA, Molyneux PD, Smiddy P, Moseley IF, Thompson AJ, Miller DH, et al. The effect of interferon beta‐1b on the evolution of new enhancing lesions to hypointense T1 lesions in secondary progressive multiple sclerosis. Multiple Sclerosis 1999;5 Suppl 1:S9 [Abstract no: 31].
FDA. Clinical Review of the June 28, 2000 Amendment to Supplemental BLA 98‐0737 ‐ ‐ STN103471 Chiron Corp.; Interferon β‐1b (Betaseron ®.Information covered in this review: June 28, 2000 – major amendment to supplement June 14, 2001 – Responses to FDA Complete Review Letter; Combined analyses of North American and European SPMS Studies. http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/ucm088275.pdf December 13, 2001.
Freeman JA, Thompson AJ, Fitzpatrick R, Hutchinson M, Miltenburger C, Beckmann K, et al. Interferon‐beta1b in the treatment of secondary progressive MS: impact on quality of life. Neurology 2001;57(10):1870‐5.
Goodin SD. Interferon b treatment for multiple sclerosis. Lancet 1999;353:496.
Goodkin DE. Interferon b therapy for multiple sclerosis. Lancet 1998;352:1486–7.
Inglese M, van Waesberghe JH, Rovaris M, Beckmann K, Barkhof F, Hahn D, et al. The effect of interferon beta‐1b on quantities derived from MT MRI in secondary progressive MS. Neurology 2003;60(5):853‐60.
Johnson KP, Panitch HS. Interferon b treatment for multiple sclerosis. The Lancet 1999;353:494.
Kappos L, Polman C, Pozzilli C, Thompson A, Beckmann K, Dahlke F. Final analysis of the European multicenter trial on IFNbeta‐1b in secondary‐progressive MS. Neurology 2001;57(11):1969‐75.
Kappos L, Weinshenker B, Pozzilli C, Thompson AJ, Dahlke F, Beckmann K, et al. Interferon beta‐1b in secondary progressive MS: a combined analysis of the two trials. Neurology 2004;63(10):1779‐87.
Miller DH, Molyneux PD, Barker GJ, MacManus DG, Moseley IF, Wagner K. Effect of interferon‐beta1b on magnetic resonance imaging outcomes in secondary progressive multiple sclerosis: results of a European multicenter, randomized, double‐blind, placebo‐controlled trial. European Study Group on Interferon‐beta1b in secondary progressive multiple sclerosis. Annals of Neurology 1999;46(6):850‐9.
Miller DH, Molyneux PD, MacManus DG, Barker GJ, Wagner K. A double‐blind, placebo‐controlled trial of interferon A‐1b in secondary progressive multiple sclerosis: magnetic resonance imaging results. Annals of Neurology 1998;44(3):503‐4.
Miller DH, Polman C, Pozzilli C, Kappos L, Thompson AJ, Wagner K, et al. MRI protocol for the European trial of Beta interferon‐1b in secondary progressive multiple sclerosis. Journal of the Neurological Sciences 1997;150:S251.
Molineux PD, Kappos L, Polman C, Pozzilli C, Barkhof F, Filippi M, et al. The effect of interferon beta 1 b treatment on MRI measures of cerebral atrophy in secondary progressive multiple sclerosis. Brain 2000;123:2256‐63.
Molyneux PD, Barker GJ, Barkhof F, Beckmann K, Dahlke F, Filippi M, et al. Clinical‐MRI correlations in a European trial of interferon beta‐1b in secondary progressive MS. Neurology 2001;57(12):2191‐7.
Molyneux PD, Brex PA, Fogg C, Lewis S, Middleditch C, Barkhof F, et al. The precision of T1 hypointense lesion volume quantification in multiple sclerosis treatment trials: a multicenter study. Multiple Sclerosis 2000;6(4):237‐40.
Polman C, Kappos L, White R, Dahlke F, Beckmann K, Pozzilli C, et al. Neutralizing antibodies during treatment of secondary progressive MS with interferon beta‐1b. Neurology 2003;60(1):37‐43.
Polman CH, Dahlke F, Thompson AJ, Ghazi M, Kappos L, Miltenburger C, et al. Interferon beta‐1b in secondary progressive multiple sclerosis‐‐outline of the clinical trial. Multiple Sclerosis 1995;1 Suppl 1:S51‐4.
Polman CH, Kappos L, Dahlke F, Graf R, Beckmann K, Bogumil T, et al. Interferon beta‐1b treatment does not induce autoantibodies. Neurology 2005;64(6):996‐1000.
Sormani MP, Rovaris M, Comi G, Filippi M. A reassessment of the plateauing relationship between T2 lesion load and disability in MS. Neurology 2009;73:1538‐42.
The European Study Group on Interferon beta. Placebo‐controlled multicentre randomised trial of interferon beta‐1b in treatment of secondary progressive multiple sclerosis. Lancet 1998;352(9139):1491‐7.
Thompson AJ, Kappos L, Polman CH, Pozzilli C, Dahlke E, The European Study Group on Interferon beta. Interferon beta‐1b delays progression of disability in secondary progressive multiple sclerosis: final results of the European multicentre study. Multiple Sclerosis 1998;4(4):392.
Zimmermann C, Walther EU, Goebels N, Lienert C, Kappos L, Hartung HP, et al. Interferon beta‐1b for treatment of secondary chronic progressive multiple sclerosis. Nervenarzt 1999;70(8):759‐63.

References to studies excluded from this review

Arnason 1999 {published data only}

Arnason, BGW, Jacobs G, Hanlon M, Harding B, Noronha A.B.C, Auty A, et al. TNF neutralization in MS: Results of a randomized, placebo‐controlled multicenter study. Neurology 1999;53(3):457‐65.

Bar‐Or 2007 {published data only}

Bar‐Or A, Vollmer T, Antel J, Arnold DL, Bodner CA, Campagnolo D, et al. Induction of Antigen‐Specific Tolerance in Multiple Sclerosis after Immunization With DNA Encoding Myelin Basic Protein in a Randomized, Placebo‐Controlled Phase 1/2 Trial. Archives of Neurology 2007;64(10):1407‐15.

Baum 2006 {published data only}

Baum K, Mannitol Formulation Study Group. Safety and tolerability of a 'refrigeration‐free' formulation of interferon beta‐1b‐‐results of a double‐blind, multicentre, comparative study in patients with relapsing‐remitting or secondary progressive multiple sclerosis. The Journal of international medical research 2006;34(1):1‐12.

Boiko2001 {published data only}

Boiko A, Skurkovich S, Buglak A, Alekseeva, T, Smirnova N, Skurkovich B, et al. Randomized, placebo‐controlled trial comparing short course of anti‐ifn? And anti‐tnfa showed only anti‐ifn? Effective in secondary progressive ms . Journal of Neurosurgery  2001;187 (suppl 1):S456 .

Buhse 2006 {published data only}

Buhse M. Efficacy of EMLA cream to reduce fear and pain associated with interferon beta‐1a injection in patients with multiple sclerosis. The Journal of neuroscience nursing : journal of the American Association of Neuroscience Nurses 2006;38(4):222‐6.

Christodoulou 2006 {published data only}

Christodoulou C, Melville P, Scherl WF, MacAllister WS, Elkins LE, Krupp LB. Effects of donepezil on memory and cognition in multiple sclerosis. Journal of the neurological sciences. 2006;245(1‐2):127‐36.

Fernandez 2002 {published data only}

Fernandez O, Guerrero M, Mayorga C, Munoz L, Lean A, Luque G, et al. Combination therapy with interferon beta‐1b and azathioprine in secondary progressive multiple sclerosis. A two‐year pilot study. Journal of Neurology 2002;249(8):1058‐62.

Goodkin 1997 {published data only}

Goodkin DE, Kinkel RP, Weinstock‐Guttman B, Medendorp VanderBurg, Gogol DM, Perryman JE, et al. A randomized, double‐masked, dose‐comparison, phase II study of bimonthly intravenous methyprednisolone (IVMP) to modify progression of disability in patients with secondary progressive multiple sclerosis (SPMS). Neurology 1997;48 Suppl 2(3):A339‐40.

Goodkin 2000a {published data only}

Goodkin DE, Shulman M, Winkelhake J, Waubant E, Andersson P, Stewart T, et al. A phase I trial of solubilized DR2:MBP84‐102 (AG284) in multiple sclerosis. Neurology 2000;54 (7):1414.

Hoogervorst 2002 {published data only}

Hoogervorst EL, Polman CH, Barkhof F. Cerebral volume changes in multiple sclerosis patients treated with high‐dose intravenous methylprednisolone. Multiple Sclerosis 2002;8(5):415‐9.

Leary 2003 {published data only}

Leary SM, Miller DH, Stevenson VL, Brex PA, Chard DT, Thompson AJ. Interferon beta‐1a in primary progressive MS: an exploratory, randomized, controlled trial. Neurology 2003;60(1):44‐51.

Montalban 2009 {published data only}

Montalban X, Sastre‐Garriga J, Tintoré M, Brieva L, Aymerich FX, Río J, et al. progressive and transitional multiple sclerosisA single‐center, randomized, double‐blind, placebo‐controlled study of interferon beta‐1b on primary. Multiple Sclerosis 2009;15(10):1195–205.

Patti 1999 {published data only}

Patti F, L'Episcopo MR, Cataldi ML, Reggio A. Natural interferon‐beta treatment of relapsing‐remitting and secondary‐progressive multiple sclerosis patients. A two‐year study. Acta Neurologica Scandinavica 1999;100(5):283‐9.

Rio 2007 {published data only}

Río J, Tintoré M, Nos C, Téllez N, Galán I, Pelayo R, et al. Interferon beta in secondary progressive multiple sclerosis. Daily clinical practice. Journal of Neurology 2007;254:849–53.

Skurkovich 2001 {published data only}

Skurkovich S, Boiko A, Beliaeva I, Buglak A, Alekseeva T, Smirnova N, et al. Randomized study of antibodies to IFN‐gamma and TNF‐alpha in secondary progressive multiple sclerosis. Multiple Sclerosis 2001;7(5):277‐84.

Smith 2005 {published data only}

Smith DR, Weinstock‐Guttman B, Cohen JA, Wei X, Gutmann C, Bakshi R, et al. A randomized blinded trial of combination therapy with cyclophosphamide in patients‐with active multiple sclerosis on interferon beta. Multiple Sclerosis 2005;11(5):573‐82.

Tubridy 1999 {published data only}

Tubridy N, Behan PO, Capildeo R, Chaudhuri A, Forbes R, Hawkins CP, et al. The effect of anti‐alpha4 integrin antibody on brain lesion activity in MS. The UK Antegren Study Group. Neurology 1999;53(3):466‐72.

Zavalishin 2003 {published data only}

Zavalishin IA, Gusev EI, Iakhno NN, Skoromets AA, Ko AN, Demina TL, et al. Results of a multicenter study of Rebif‐22 mcg administration in Russia. Zhurnal nevrologii i psikhiatrii imeni S.S. Korsakova / Ministerstvo zdravookhraneniia i meditsinskoi promyshlennosti Rossiiskoi Federatsii, Vserossiiskoe obshchestvo nevrologov [i] Vserossiiskoe obshchestvo psikhiatrov (ZH NEVROL PSIKHIATR) 2003;Spec No 2:73‐8.

Additional references

Amato 2006

Amato MP, Valentina Zipolia, Portaccio E. Multiple sclerosis‐related cognitive changes: A review of cross‐sectional and longitudinal studies. Journal of the Neurological Sciences 2006;245(1‐2):41‐6.

Calabrese 2009

Calabrese M, Agosta F, Rinaldi F, Mattisi I, Grossi P, Favaretto A, et al. Cortical Lesions and Atrophy Associated With Cognitive Impairment in Relapsing‐Remitting Multiple Sclerosis. Archives of Neurology 2009;66(9):1144‐50.

Compston 2002

Compston A, Coles A. Multiple Sclerosis. Lancet 2002;359(9313):1221‐31. [MEDLINE: 11955556]

Confavreux 2000

Confavreux C, Vukusic S, Moreau T, Adeleine P. Relapses and progression of disability in multiple sclerosis. New England Journal of Medicine 2000;343(20):1430‐8.

Confavreux 2003

Confavreux C, Vukusic S, Adeleine P. Early clinical predictors and progression of irreversible disability in multiple sclerosis: an amnesic process. Brain 2003;126:770‐82.

Confavreux 2006

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

Cottrell 1999

Cottrell DA, Rice GPA, Hader W, BaskervilleJ, Koopman WJ, Ebers GC. The natural history of multiple sclerosis: a geographically based study. 5. The clinical features and natural history of primary progressive multiple sclerosis. Brain 1999;122:625‐39.

Der Simonian 1986

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

Duque 2008

Duque B, Sepulcre J, Bejarano B, Samaranch L, Pastor P, Villoslada P. Memory decline evolves independently of disease activity in MS. Multiple Sclerosis 2008;14(7):947‐53.

Ebers 2000

Ebers GC, Koopman WJ, Hader W, Sadovnick AD, Kremenchutzky M, Mandalfino P, et al. The natural history of multiple sclerosis: a geographically based study: 8: familial multiple sclerosis. Brain 2000;123(Pt 3):641‐9.

Ebers 2008

Ebers GC, Heigenhauser L, Daumer M, Lederer C, Noseworthy JH. Disability as an outcome in MS clinical trials. Neurology 2008;71(9):624‐31.

Ebers 2009

Ebers GC, Reder AT, Traboulsee A, Li D, Langdon D, Goodin DS, et al. Long‐term follow‐up of the original interferon‐beta1b trial in multiple sclerosis: Design and lessons from a 16‐year observational study. Clinical Therapeutics 2009;31(8):1724‐36.

Ebers 2010

Ebers GC, Traboulsee A, Li D, Langdon D, Reder AT, Goodin DS, et al. Analysis of clinical outcomes according to original treatment groups 16 years after the pivotal IFNB‐1b trial. Journal of Neurology Neurosurgery & Psychiatry 2010;81:907‐12.

Egger 1997

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

EMEA 2000

The European Agency for the Evaluation of Medical Products. PRESS RELEASE. CPMP/4305/00 18 December 2000.

EMEA 2006

European Medicines Agency. Press Release. Doc. Ref. EMEA/152608/2006 28 April 2006.

EMEA 2008

Committee for medicinal products for human use summary of positive opinion for Extavia. International Nonproprietary Name (INN): interferon beta‐1b. Novartis Europharm Ltd.; London, 20 March 2008. Doc. Ref. EMEA/CHMP/138637/2008. http://data.ellispub.com/pdf/EN/2008/EMEA/Extavia_13863908en.pdf (17/02/2011).

Esposito 2010

Esposito F, Radaelli M, Martinelli V, Sormani MP, Martinelli Boneschi F, Moiola L, et al. Comparative study of mitoxantrone efficacy profile in patients with relapsing–remitting and secondary progressive multiple sclerosis. Multiple Sclerosis 2010;16(12):1490‐9.

FDA 2000

Center for Drug Evaluation and Research. Approval letter for Novantrone (mitoxantrone hydrochloride) injection for Multiple Sclerosis: Immunex Corporation. FDA U.S. Food & Drug Administration; 1 March 2000. Application Number NDA 21‐120. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/21120.pdf_Novantrone_Approv.pdf (accessed 31 May 2011).

FDA 2001

Unger, EF. Interferon 1b (Betaseron) treatment of secondary progressive forms of Multiple Sclerosis. Clinical Review of the June 28, 2000 Amendment to Supplemental BLA 98‐0737 ‐‐ STN103471: Chiron Corp. FDA U.S. Food & Drug Administraion; 13 December 2001. http://www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/ucm088275.pdf (accessed 6 June 2010).

FDA 2009

Center for Drug Evaluation and Research. Approval letter for Interferon Beta 1b, label product with the proprietary name Extavia: Novartis Pharmaceuticals Corporation. FDA U.S. Food & Drug Administration; 14 August 2009. Application Number 125290. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2009/0125290s000approv.pdf (accessed 26 May 2011).

Filippini 2003

Filippini, Munari L, Ebers GC, D'Amico R, Rice GPA. Interferons in relapsing remitting multiple sclerosis. The Lancet 2003;361(9371):545‐52.

Frisher 2009

Frisher 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.

Glass 2010

Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell 2010;140(6):918‐34.

Higgins 2009

Higgins JPT, Altman DG (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.2 [updated September 2009]. The Cochrane Collaboration, 2009. Available from www.cochrane‐handbook.org.

Kappos 2001

Kappos L, Polman C, Pozzilli C, Thompson A, Beckmann K, Dahlke F. Final analysis of the European multicenter trial on IFNbeta‐1b in secondary‐progressive MS. Neurology 2001;57(11):1969‐75.

Kappos 2004

Kappos L, Weinshenker B, Pozzilli C, Thompson AJ, Dahlke F, Beckmann K, et al. Interferon beta‐1b in secondary progressive MS: a combined analysis of the two trials. Neurology 2004;63(10):1779‐87. [PUBMED: 15557490]

Kappos 2009

Kappos L, Freedman MS, Polman CH, Edan G, Hartung HP, Miller DH, et al. Long‐term effect of early treatment with interferon beta‐1b after 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.

Koch 2010

Koch M, Kingwell E, Rieckmann P, Tremlett H, UBC MS Clinic Neurologists. The natural history of secondary progressive multiple sclerosis . J Neurol Neurosurg Psychiatry 2010;81:1039‐43.

Koch‐Henriksen 2006

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

Kremenchutzky 2006

Kremenchutzky M, Rice GPA, Baskerville DM, Wingerchuk DM, Ebers GC. The natural history of multiple sclerosis: a geographically based study 9: Observations on the progressive phase of the disease. Brain 2006;129(3):584‐94.

Kurtzke 1983

Kurtzke JF. Rating neurological impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 1983;33(11):1444‐52. [MEDLINE: 84040263]

Lassmann 2009

Lassmann H. Axonal and neuronal pathology in multiple sclerosis:what have we learnt from animal models. Experimental Neurology 2009 Oct 17 [Epub ahead of print]. [DOI: doi:10.1016/j.expneurol.2009.10.009]

Li 2001

Li DK, Zhao GJ, Paty DW, The University of British Columbia, MS/MRI Analysis Research Group, The SPECTRIMS Study Group. Randomized controlled trial of interferon‐beta‐1a in secondary progressive MS: MRI results. Neurology 2001;56(11):1505‐13.

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(4):907‐11.

Lublin 2003

Lublin FD, Baier M, Cutter G. Effect of relapses on development of residual deficit in multiple sclerosis. Neurology 2003;61(11):1528‐32.

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. [MEDLINE: 11456302]

Miller 1999

Miller DH, Molyneux PD, Barker GJ, MacManus DG, Moseley IF, Wagner K. Effect of interferon‐beta1b on magnetic resonance imaging outcomes in secondary progressive multiple sclerosis: results of a European multicenter, randomized, double‐blind, placebo‐controlled trial. European Study Group on Interferon‐beta1b in secondary progressive multiple sclerosis. Annals of Neurology 1999;46(6):850‐9.

Miller 2006

Miller DM, Cohen JA, Kooijmans M, Tsao E, Cutter G, Baier M. Change in clinician‐assessed measures of multiple sclerosis and subject‐reported quality of life: results from the IMPACT study. Multiple Sclerosis 2006;12(2):180‐6.

Molyneux 2000b

Molyneux PD, Kappos L, Polamn C, Pozzilli C, Barkhof F, Filippi M, et al. The effect of interferon beta 1 b treatment on MRI measures of cerebral atrophy in secondary progressive multiple sclerosis. Brain 2000;123:2256‐63.

Nikfar 2010

Nikfar S, Rahimi R, Abdollahi M. A Meta‐Analysis of the Efficacy and Tolerability of Interferon‐Beta in Multiple Sclerosis, Overall and by Drug and Disease Type. Clinical Therapy 2010;32:1871‐88.

Noseworthy 2000

Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple Sclerosis. New England Journal of Medicine 2000;343(13):938‐52.

Oliver 2011

Oliver BJ, Kohli E, Kasper LH. Interferon therapy in relapsing‐remitting multiple sclerosis: A systematic review and meta‐analysis of the comparative trials. Journal of the Neurological Sciences 2011;302(1‐2):96‐105.

Patrucco 2009

Patrucco L, Rojas JI, Cristiano E. Long Term Effect of Interferon‐Beta on Disease Severity in Relapsing‐Remitting Multiple Sclerosis Patients. Rev Neurol  2010;50:529‐32.

Paty 1998

Paty DW, Ebers GC. Multiple Sclerosis. Philadelphia: FA Davis Co, 1998.

Polman 1995

Polman CH, Dahlke F, Thompson AJ, Ghazi M, Kappos L, Miltenburger C, et al. Interferon beta‐1b in secondary progressive multiple sclerosis‐‐outline of the clinical trial. Multiple Sclerosis 1995;1 Suppl 1:S51‐4.

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. [PUBMED: 16283615]

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. [MEDLINE: 83202737]

Rice 2001

Rice GPA, Incorvaia B, Munari L, Ebers G, Polman C, D'Amico R, et al. Interferon in relapsing‐remitting multiple sclerosis. Cochrane Database of Systematic Reviews 2001, Issue 4. [DOI: 10.1002/14651858.CD002002]

Seewann 2009

Seewann A, Vrenken H, van der Valk P, Blezer EL, Castelijns JA, Polman CH, et al. Diffusely abnormal white matter in chronic multiple sclerosis: imaging and histopathologic analysis. Archives of Neurology 2009;66(5):601‐9.

Tekok‐Kilic 2007

Tekok‐Kilic A, Benedict RH, Weinstock‐Guttman B, Dwyer MG, Carone D, Srinivasaraghavan B, et al. Independent contributions of cortical gray matter atrophy and ventricle enlargement for predicting neuropsychological impairment in multiple sclerosis. Neuroimage 2007;36(4):1294‐300. [PUBMED: 17524670]

Tremlett 2006

Tremlett H, Paty D, Devonshire V. Disability progression in multiple sclerosis is slower than previously reported. Neurology 2006;66:172‐7.

Tremlett 2009

Tremlett H, Yousefi M, Devonshire V, Rieckmann P, Zhao Y on behalf of the UBC Neurologists. Impact of multiple sclerosis relapses on progression diminishes with time. Neurology 2009;73(20):1616‐23.

Troiano 2007

Trojano M, Pellegrini F, Fuiani A, Paolicelli D, Zipoli V, Zimatore GB, et al. New Natural History of Interferon‐beta –Treated Relapsing Multiple Sclerosis. Ann Neurol 2007;61:300–6.

Ukkonen 2009

Ukkonen M, Vahvelainen T, Hämäläinen P, Dastidar P, Elovaara I. Cognitive dysfunction in primary progressive multiple sclerosis: a neuropsychological and MRI study. Multiple Sclerosis 2009;15:1055‐61.

Weinshenker 1989

Weinshenker BG, Bass B, Rice GP, Noseworthy J, Carriere W, Baskerville J, et al. The natural history of multiple sclerosis:a geographically based study. 2. Predictive value of the early clinical course. Brain 1989;112(Pt 6):1419‐28.

Weinshenker 1989a

Weinshenker BG, Bass B, Rice GPA, Noseworthy J, Carriere W, Baskerville J, et al. The natural history of multiple sclerosis: a geographically based study I Clinical course and disability. Brian 1989;112:133‐46.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Andersen 2004 (Nordic)

Methods

Randomized, double‐blind, placebo controlled two arm trial.

It was a multicentre study (32): Denmark (4 centres, 48 patients), Finland (6 centres, 123 patients), Norway (12 centres, 120 patients) and Sweden (10 centres, 80 patients).

Participants

371 patients with SP MS: 188 IFN and 183 placebo‐treated.

Interventions

IFN beta‐1a (Rebif; Serono), 22 ug subcutaneously once weekly versus placebo (unspecified) for 36 months.

Outcomes

Primary: The time to progression on the EDSS, defined as an increase from baseline by at least 1.0 point (or 0.5 points if
the baseline EDSS score was 5.5 or higher) and confirmed at two consecutive scheduled visits separated by 6 months.

Secondary:Time to progression on the RFSS (Regional Functional System Score).

Tertiary: The proportion of progression free patients, time to first exacerbation, proportion of exacerbation free patients, MS related hospitalisation rate, ambulation index and arm index.

Notes

Serono personnel were involved in the study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

No information.

Allocation concealment (selection bias)

Unclear risk

"equal allocation (page 707)".

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only 83% of randomised patients completed about 3 years.

Lost to follow‐up:

‐ 21% for treated patients

‐ 16% for placebo

More AE and patients' decision in the treated group

The median time on treatment was 35.2 months (mean 32.0) for placebo and 35.0 months (mean 31.1) for IFN.

Other bias

High risk

The study was terminated during the third year by the Steering Committee based on negative results from the SPECTRIMS study.

Blinding of participants and personnel (performance bias)

Unclear risk

"Patients were instructed to cover injection sites."

No blinding questionnaire was conducted at study termination.

Blinding of outcome assessment (detection bias)

Unclear risk

"double blind" is reported, without any other comments

Placebo is not described.

No blinding questionnaire was conducted at study termination.

Cohen 2002 (IMPACT)

Methods

Randomized, double‐blind, placebo‐controlled, two‐arm trial.

It was a multicentre study (42): 31 in the United States, four in Canada, and seven in Europe.

Participants

436 patients with SPMS: 217 IFN beta 1a (Avonex) and 219 placebo (unspecified) patients.

Interventions

Weekly IM injections of IFNb‐1a (60 ug) versus placebo (unspecified) for 24 months.

Outcomes

Primary: Baseline to month 24 change in the MS Functional Composite (MSFC), MSFC Z‐score change.

Others :

Clinical outcomes: number of patients with EDSS worsening or stable, time to EDSS worsening, defined as a 1.0 step increase for baseline EDSS 3.5 to 5.5 and 0.5 step increase for baseline EDSS 6.0 to 6.5 sustained for 3 months. Relapse rate, Quality of life (The MS Quality of Life Inventory (MSQLI), and depression (The Beck Depression Inventory).

MRI Outcome: Total volumes of T2‐hyperintense lesions and Gd‐enhancing lesions.

Notes

Biogen, Inc. supported this study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The contract research organization computer generated two minimization schemes, one for North America and one for Europe and Israel (page 680).

Allocation concealment (selection bias)

Unclear risk

Not reported any comments on this aspect.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Lost to follow‐up 52 patients (P 23; T 29 ) /436 (12 %).

Reasons for withdrawal were reported.

Other bias

High risk

In FDA’s letters of July 23, 1998, December 6, 1999, and March 8,2000, Biogen was informed that the proposed primary endpoint, the Multiple Sclerosis Functional Composite (MSFC) had not been validated as a clinical efficacy outcome measurement and therefore, was not appropriate for use as a primary efficacy endpoint in a Phase 3 study (FDA 2001; Cohen 2002 (IMPACT)).

Blinding of participants and personnel (performance bias)

Unclear risk

Patients were instructed to cover injection sites.

No blinding questionnaire was conducted at study termination.

Blinding of outcome assessment (detection bias)

Low risk

The examining technician administered the MSFC, and the examining neurologist determined the EDSS during all scheduled study visits.Neither the examining technician nor the examining neurologist was involved with any other aspect of subject care, and neither had access to the results of prior examinations or to clinical information that might compromise blinding.

North American SG 2004

Methods

Randomized, double‐blind, placebo‐controlled, four‐arm trial.

It was a multicentre study, performed in United States (31 centers) and Canada (4).

Participants

939 patients with SPMS: 631 treated with IFN beta‐1b (Betaferon, 2 arms with different dosages) and 308 placebo‐treated.

Interventions

Subcutaneous injection every other day of IFN‐1b 250 ug (8.0 million international units [MIU]), IFN‐1b 160ug (5 MIU)/m2 body surface area (mean administered dose 220 ug) or one of two placebo treatments, for 36 months.

Outcomes

Primary: The time to progression on the EDSS, defined as an increase from baseline by more than 1.0 point (or 0.5 points if the baseline EDSS score was 6.0 to 6.5 ) and confirmed at two consecutive scheduled visits separated by 6 months.

Secondary :

Clinical : Mean EDSS change, annual relapse rate, change in composite neuropsychological test score (Rao Brief Repeatable Battery).

MRI: absolute change in T2‐weighted lesion area (assessed annually)from baseline to end point,annual new active lesion rate (new, recurrent, and newly enlarging or enhancing lesions per year on study) (Monthly scanning for Frequent MRI substudy population, n=163).

Tertiary :

Clinical: proportion of subjects with confirmed disease progression, relapse‐related endpoints, social handicap (Environmental Status Scale), Quality of Life (MSQLI), depression (Beck Depression Inventory).
MRI : Absolute and % change in annual T2 lesion area, other measures of disease activity (FDA 2001).

Notes

Funded by Berlex Laboratories, Montville, NJ.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Each block allocated 2 subjects to fixed‐dose Betaseron, 2 to Body Surface Area (BSA)‐adjusted Betaseron, 1 to fixed placebo, and 1 to BSA‐adjusted placebo. Each site received an adequate number of blocks, based on projected subject recruitment, to ensure sequential subject numbering within each site. The randomisation schedule was generated by the Biostatistics and Data Management Group of Berlex Laboratories (Richmond, CA) using an SAS program (Cary, NC).

Allocation concealment (selection bias)

Low risk

The biostatistician and supporting programmers were the only individuals with access to the randomisation codes.The active study drug was indistinguishable from placebo in appearance, smell and colour, and labels and packages for active study agent and placebo were indistinguishable.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only 72% of randomised patients were included in analysis (FDA 2001 page 26).

Other bias

High risk

The Independent Data and Safety Monitoring Board recommended early termination of the trial based on the results of a planned interim analysis indicating that continuing the trial was unlikely to change the results.The study initiated in August 2, 1995, interrupted November 22, 1999. The last patient enrolled on April 1, 1997. The final patient visit occurred on November 15, 1999.

Blinding of participants and personnel (performance bias)

High risk

Patients and treating physicians were more likely to guess treatment allocation correctly due to side effects. 

Blinding of outcome assessment (detection bias)

Low risk

Separate treating and examining physicians were employed.

A questionnaire to evaluate the success of blinding was made: results suggest a substantial maintenance of blinding for the critical evaluation physicians (FDA 2001).

SPECTRIMS 2001

Methods

Randomized, double‐blind, placebo‐controlled, three‐arm trial.

It was a multicentre study (22) performed in Europe, Canada and Australia.

Participants

618 patients with SPMS: 413 treated with IFN beta‐1a (2 arms with different dosages= 204 Rebif 44ug and 209 Rebif 22ug) and 205 placebo (unspecified) patients.

Interventions

Subcutaneous injection, three times weekly of IFN‐1a (Rebif) at 2 different dosages (22 and 44 ug) or placebo (unspecified) treatments, for 36 months.

Outcomes

Primary: The time to progression on the EDSS, defined as an increase from baseline by more than 1.0 point (or 0.5 points if baseline EDSS was < 5.5), confirmed at two consecutive scheduled visits separated by 3 months.

Secondary :

Clinical : proportion of patients progressing, exacerbation count, time to first exacerbation, time between first and second exacerbations, number of moderate and severe exacerbations, number of steroid courses, number of hospitalizations, and Integrated Disability Status Score (IDSS, defined by area under an EDSS time‐curve adjusted for baseline).

MRI: lesion burden, T2 activity (New T2, recurrent T2, newly enlarging T2, and persistently enlarging T2 lesions) enhancing lesions and Combined Unique (CU) activity analysis: (Monthly scanning for Frequent MRI substudy population, n=283).

Notes

Funded by Serono International SA, Geneva, Switzerland.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generated randomisation list provided by Serono, stratified by center; treatments were equally allocated with a block size of six.

Allocation concealment (selection bias)

Low risk

The block size was not revealed to the investigators.The manufacturer labelled containers of study medication with patient identification numbers based on the randomisation list, and patients received the medication labelled with their numbers.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Lost to follow‐up 47/618 patients (44 ug = 14, 22 ug =14, Placebo= 19) (8 %).

Other bias

Low risk

Blinding of participants and personnel (performance bias)

Unclear risk

Patients were instructed to cover injection sites and to discuss only neurologic matters during neurologic evaluations.

Blinding questionnaire was conducted at study termination.

Blinding of outcome assessment (detection bias)

Low risk

Solutions of IFNb‐1a and placebo were physically indistinguishable, and packaging and labelling were prepared to preserve blinding neurologic assessments. Separate treating and examining physicians were employed. Patients were instructed to cover injection sites and to discuss only neurologic matters during neurologic evaluations.

Evaluating physicians guessed treatment assignments correctly for 29% of patients on active treatment and 26% of patients on placebo.

The European SG 1998

Methods

Randomized, double‐blind, placebo‐controlled, two‐arm trial. It was a multicentre study (32) performed in European countries.

Participants

718 patients with SP MS: 360 treated with IFN beta 1b (Betaferon) and 358 placebo‐treated.

Interventions

Subcutaneous injection every other day of IFN‐1b 250 ug (8.0 million international units [MIU]), or placebo (unspecified) treatments, for 36 months.

Outcomes

Primary: The time to progression on the EDSS, defined as an increase from baseline by more than 1.0 point (or 0.5 points if baseline EDSS was 6), confirmed at two consecutive scheduled visits separated by 3 months, not obtained during relapses.

Secondary :

Clinical : proportion of patients with confirmed progression,Time to becoming wheelchair‐bound,Proportion of patients becoming wheelchair bound,Change in EDSS from baseline,Annual relapse rate,Time to first relapse, Proportion of patients with moderate or severe relapse.

Tertiary :

MS‐related steroid use and hospital admissions. Montgomery Asberg Depression RatingScale (MADRS) to assess mood changes and suicidal risk.

MRI :Percentage change in annual T2 lesion volume. (Monthly scanning for Frequent MRI substudy population, n=125)T1‐weighted gadolinium‐enhanced scans in months 0–6 and 18–24.

Notes

This study was funded by Schering AG, Berlin.

The cognitive function evaluation was included in the protocol Polman 1995 but results were not retrieved in the primary study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Central randomisation schedule assigned placebo or interferon ‐1b to blocks of six patients in a 1/1 ratio.

Allocation concealment (selection bias)

Low risk

Access to the code was strictly limited according to study protocol.

Incomplete outcome data (attrition bias)
All outcomes

High risk

1/4 of randomised patients were lost to follow‐up.

Due to early study termination, at 33 months primary outcome was not available for 187/718 patients (26%) of the randomised patients; placebo= 97/358 (27%), treated= 90/360 (25%).

Is not clear which data were used for analysis.

Other bias

High risk

In November 20, 1997, a planned interim analysis was done after all patients had been in the study for at least 2 years.The results gave evidence of efficacy and led to a recommendation to the early termination of the study. The study has been initiated in September 1994 and stopped in March 1998. EDSS information at month 33 was available for three‐fourths of patients in both treatment groups in the final data set, compared with 40% in the interim analysis (Kappos 2001, page 1971).

Blinding of participants and personnel (performance bias)

High risk

54·3% of 304 patients guessed correctly that they were on placebo and 65·6% of 308 that they were on active treatment.

Blinding of outcome assessment (detection bias)

Unclear risk

Interferon beta‐1b was indistinguishable from placebo.

Separate treating and examining physicians were employed. Separate designated EDSS raters performed only the standardized neurologic tests EDSS physicians received no potentially unmasking information from the treating physicians, and were allowed to speak to patients only as necessary to carry out neurological tests. During EDSS assessments all potential injection sites were covered.

Blinding questionnaire was conducted at study termination; EDSS physicians guessed correctly for only 54 (18.6%) of 291 of patients on placebo and 65 (20.8%) of 312 patients on interferon ‐1b. "The data at study termination were collected under double‐blind conditions and comprise the data collected after database lock for the interim analysis until the beginning of the open‐label extension phase" (Kappos 2001 page 1970). It is unclear if the 3 years evaluation was double blind or open for the early study termination.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Arnason 1999

Treatment is Lenercept

Bar‐Or 2007

Treatment is BHT‐3009, a tolerizing DNA vaccine encoding full‐length human myelin basic protein

Baum 2006

Randomized, double‐blind, parallel group, comparative trial evaluating the safety and tolerability of two formulations of IFNb (IFNbeta‐1b‐G or the refrigeration‐free formulation (IFNbeta‐1b‐M) in patients with relapsing‐remitting or secondary progressive MS.

Boiko2001

Patients with secondary progressive MS in a randomized, double‐blind, placebo‐controlled study evaluating short course of antibodies (AB) to IFN‐gamma or AB to tumour necrosis factor (TNF)‐alpha

Buhse 2006

Relapsing remitting MS patients randomised to receive EMLA cream or placebo cream 2 hr before IFNβ‐1a injection once weekly

Christodoulou 2006

Treatment is donepezil

Fernandez 2002

Prospective open label evaluating combination therapy with interferon beta 1 b and azathioprine in secondary progressive MS.

Goodkin 1997

A randomized, double‐masked, dose‐comparison, phase II study of intravenous methyprednisolone in patients with secondary progressive multiple sclerosis.

Goodkin 2000a

The treatment is complex of human leukocyte antigen‐DR2 with myelin basic protein84‐102 (AG284).

Hoogervorst 2002

MS patients suffered from an acute relapse , treated with intravenous methylprednisolone, participating in a double‐blind, placebo‐controlled,
randomized, parallel‐group, multicentre, phase II study evaluating the safety, tolerability and MRI effects of oral interferon beta‐1a.

Leary 2003

Double‐blind, placebo‐controlled trial of two doses of interferon ‐1a in primary progressive MS patients.

Montalban 2009

study on the effect of interferon beta in primary progressive MS.

Patti 1999

Treatment is natural interferon beta derived from human fibroblasts (Ares‐Serono@).

Rio 2007

Open‐label, non‐randomised, observational study.

Skurkovich 2001

Double‐blind, placebo‐controlled, study. In secondary progressive MS randomized to three groups (antibodies to TNF‐a, to IFN‐g; and placebo).

Smith 2005

Combination therapy with pulse cyclophosphamide given with methylprednisolone
and interferon beta (IFNb)‐1a in remitting‐relapsing MS patients during IFNb monotherapy.

Tubridy 1999

A double‐blind, placebo‐controlled trial in patients with relapsing‐remitting and secondary progressive MS randomized to infusions of anti‐4 integrin antibody (natalizumab; Antegren) or placebo.

Zavalishin 2003

Multicenter study evaluating the effect of Rebif 22 in relapsing remitting and secondary progressive MS.

Data and analyses

Open in table viewer
Comparison 1. Disability progression

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Sustained (6 months) EDSS increase after 3 years Show forest plot

3

2026

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.82, 1.16]

Analysis 1.1

Comparison 1 Disability progression, Outcome 1 Sustained (6 months) EDSS increase after 3 years.

Comparison 1 Disability progression, Outcome 1 Sustained (6 months) EDSS increase after 3 years.

2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Disability progression, Outcome 2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses.

Comparison 1 Disability progression, Outcome 2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses.

2.1 In patients with pre study relapses

3

1106

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.75, 1.09]

2.2 In patients without pre‐study relapses

3

903

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.83, 1.33]

3 Sustained (3 months) EDSS increase Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Disability progression, Outcome 3 Sustained (3 months) EDSS increase.

Comparison 1 Disability progression, Outcome 3 Sustained (3 months) EDSS increase.

3.1 After 2 years

2

1054

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.81, 1.08]

3.2 After 3 years

2

1336

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.80, 0.97]

4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Disability progression, Outcome 4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients.

Comparison 1 Disability progression, Outcome 4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients.

4.1 low age and disease duration

2

1336

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.83, 1.02]

4.2 high age and disease duration

3

1739

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.90, 1.18]

Open in table viewer
Comparison 2. Adverse Events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of patients with Serious AEs Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.83, 1.19]

Analysis 2.1

Comparison 2 Adverse Events, Outcome 1 Total number of patients with Serious AEs.

Comparison 2 Adverse Events, Outcome 1 Total number of patients with Serious AEs.

2 Patients who had discontinuated for AEs (including SAEs) * Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

2.62 [1.92, 3.57]

Analysis 2.2

Comparison 2 Adverse Events, Outcome 2 Patients who had discontinuated for AEs (including SAEs) *.

Comparison 2 Adverse Events, Outcome 2 Patients who had discontinuated for AEs (including SAEs) *.

3 Deaths Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.58, 3.42]

Analysis 2.3

Comparison 2 Adverse Events, Outcome 3 Deaths.

Comparison 2 Adverse Events, Outcome 3 Deaths.

4 Patients who done or attempted suicide Show forest plot

4

2441

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.36, 1.95]

Analysis 2.4

Comparison 2 Adverse Events, Outcome 4 Patients who done or attempted suicide.

Comparison 2 Adverse Events, Outcome 4 Patients who done or attempted suicide.

5 Allergy/Rash (number of events) Show forest plot

2

1657

Risk Ratio (M‐H, Random, 95% CI)

1.57 [0.99, 2.48]

Analysis 2.5

Comparison 2 Adverse Events, Outcome 5 Allergy/Rash (number of events).

Comparison 2 Adverse Events, Outcome 5 Allergy/Rash (number of events).

6 Cutaneous necrosis * Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 Adverse Events, Outcome 6 Cutaneous necrosis *.

Comparison 2 Adverse Events, Outcome 6 Cutaneous necrosis *.

6.1 Number of events

2

1336

Risk Ratio (M‐H, Random, 95% CI)

33.02 [4.57, 238.79]

6.2 Number of patients

2

1557

Risk Ratio (M‐H, Random, 95% CI)

18.76 [3.72, 94.61]

7 Injection site reactions * Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.7

Comparison 2 Adverse Events, Outcome 7 Injection site reactions *.

Comparison 2 Adverse Events, Outcome 7 Injection site reactions *.

7.1 Number of events

2

1657

Risk Ratio (M‐H, Random, 95% CI)

3.84 [3.11, 4.74]

7.2 Number of patients

3

1425

Risk Ratio (M‐H, Random, 95% CI)

1.60 [0.84, 3.07]

8 Patients with psychiatric disorders Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.94, 1.18]

Analysis 2.8

Comparison 2 Adverse Events, Outcome 8 Patients with psychiatric disorders.

Comparison 2 Adverse Events, Outcome 8 Patients with psychiatric disorders.

9 Patients with headache Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.97, 1.60]

Analysis 2.9

Comparison 2 Adverse Events, Outcome 9 Patients with headache.

Comparison 2 Adverse Events, Outcome 9 Patients with headache.

10 Patients with influenza like syndrome * Show forest plot

4

2364

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.01, 2.07]

Analysis 2.10

Comparison 2 Adverse Events, Outcome 10 Patients with influenza like syndrome *.

Comparison 2 Adverse Events, Outcome 10 Patients with influenza like syndrome *.

11 Patients with myalgia Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.93, 1.83]

Analysis 2.11

Comparison 2 Adverse Events, Outcome 11 Patients with myalgia.

Comparison 2 Adverse Events, Outcome 11 Patients with myalgia.

12 Patients with fatigue/asthenia Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.98, 1.22]

Analysis 2.12

Comparison 2 Adverse Events, Outcome 12 Patients with fatigue/asthenia.

Comparison 2 Adverse Events, Outcome 12 Patients with fatigue/asthenia.

13 Patients with leucopenia * Show forest plot

3

1921

Risk Ratio (M‐H, Random, 95% CI)

2.25 [1.06, 4.75]

Analysis 2.13

Comparison 2 Adverse Events, Outcome 13 Patients with leucopenia *.

Comparison 2 Adverse Events, Outcome 13 Patients with leucopenia *.

14 Patients with liver dysfunction Show forest plot

2

1310

Risk Ratio (M‐H, Random, 95% CI)

1.69 [0.97, 2.95]

Analysis 2.14

Comparison 2 Adverse Events, Outcome 14 Patients with liver dysfunction.

Comparison 2 Adverse Events, Outcome 14 Patients with liver dysfunction.

Open in table viewer
Comparison 3. Relapses' outcomes

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of patients with at least one relapse during follow Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Relapses' outcomes, Outcome 1 Number of patients with at least one relapse during follow.

Comparison 3 Relapses' outcomes, Outcome 1 Number of patients with at least one relapse during follow.

1.1 After 2 years

1

436

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.54, 0.95]

1.2 After 3 years

4

2639

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.84, 0.97]

2 Relapse rate Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Relapses' outcomes, Outcome 2 Relapse rate.

Comparison 3 Relapses' outcomes, Outcome 2 Relapse rate.

2.1 After 2 years

1

436

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.20, ‐0.04]

2.2 After 3 years

3

1752

Mean Difference (IV, Random, 95% CI)

‐0.16 [‐0.21, ‐0.10]

Open in table viewer
Comparison 4. MRI: role of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of patients with combined lesions at different times (6, 9 and 24 months) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.1

Comparison 4 MRI: role of treatment, Outcome 1 Number of patients with combined lesions at different times (6, 9 and 24 months).

Comparison 4 MRI: role of treatment, Outcome 1 Number of patients with combined lesions at different times (6, 9 and 24 months).

1.1 6 months

1

125

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.52, 0.95]

1.2 9 months

1

264

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.42, 0.62]

1.3 24 months

1

109

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.33, 0.80]

2 Mean absolute change of T2 lesion load Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 MRI: role of treatment, Outcome 2 Mean absolute change of T2 lesion load.

Comparison 4 MRI: role of treatment, Outcome 2 Mean absolute change of T2 lesion load.

2.1 1 year

2

1022

Mean Difference (IV, Random, 95% CI)

‐1.74 [‐3.25, ‐0.23]

2.2 2 years

2

956

Mean Difference (IV, Random, 95% CI)

‐2.56 [‐5.08, ‐0.05]

2.3 3 years

1

567

Mean Difference (IV, Random, 95% CI)

‐4.87 [‐6.22, ‐3.52]

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.

Comparison 1 Disability progression, Outcome 1 Sustained (6 months) EDSS increase after 3 years.
Figuras y tablas -
Analysis 1.1

Comparison 1 Disability progression, Outcome 1 Sustained (6 months) EDSS increase after 3 years.

Comparison 1 Disability progression, Outcome 2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses.
Figuras y tablas -
Analysis 1.2

Comparison 1 Disability progression, Outcome 2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses.

Comparison 1 Disability progression, Outcome 3 Sustained (3 months) EDSS increase.
Figuras y tablas -
Analysis 1.3

Comparison 1 Disability progression, Outcome 3 Sustained (3 months) EDSS increase.

Comparison 1 Disability progression, Outcome 4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients.
Figuras y tablas -
Analysis 1.4

Comparison 1 Disability progression, Outcome 4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients.

Comparison 2 Adverse Events, Outcome 1 Total number of patients with Serious AEs.
Figuras y tablas -
Analysis 2.1

Comparison 2 Adverse Events, Outcome 1 Total number of patients with Serious AEs.

Comparison 2 Adverse Events, Outcome 2 Patients who had discontinuated for AEs (including SAEs) *.
Figuras y tablas -
Analysis 2.2

Comparison 2 Adverse Events, Outcome 2 Patients who had discontinuated for AEs (including SAEs) *.

Comparison 2 Adverse Events, Outcome 3 Deaths.
Figuras y tablas -
Analysis 2.3

Comparison 2 Adverse Events, Outcome 3 Deaths.

Comparison 2 Adverse Events, Outcome 4 Patients who done or attempted suicide.
Figuras y tablas -
Analysis 2.4

Comparison 2 Adverse Events, Outcome 4 Patients who done or attempted suicide.

Comparison 2 Adverse Events, Outcome 5 Allergy/Rash (number of events).
Figuras y tablas -
Analysis 2.5

Comparison 2 Adverse Events, Outcome 5 Allergy/Rash (number of events).

Comparison 2 Adverse Events, Outcome 6 Cutaneous necrosis *.
Figuras y tablas -
Analysis 2.6

Comparison 2 Adverse Events, Outcome 6 Cutaneous necrosis *.

Comparison 2 Adverse Events, Outcome 7 Injection site reactions *.
Figuras y tablas -
Analysis 2.7

Comparison 2 Adverse Events, Outcome 7 Injection site reactions *.

Comparison 2 Adverse Events, Outcome 8 Patients with psychiatric disorders.
Figuras y tablas -
Analysis 2.8

Comparison 2 Adverse Events, Outcome 8 Patients with psychiatric disorders.

Comparison 2 Adverse Events, Outcome 9 Patients with headache.
Figuras y tablas -
Analysis 2.9

Comparison 2 Adverse Events, Outcome 9 Patients with headache.

Comparison 2 Adverse Events, Outcome 10 Patients with influenza like syndrome *.
Figuras y tablas -
Analysis 2.10

Comparison 2 Adverse Events, Outcome 10 Patients with influenza like syndrome *.

Comparison 2 Adverse Events, Outcome 11 Patients with myalgia.
Figuras y tablas -
Analysis 2.11

Comparison 2 Adverse Events, Outcome 11 Patients with myalgia.

Comparison 2 Adverse Events, Outcome 12 Patients with fatigue/asthenia.
Figuras y tablas -
Analysis 2.12

Comparison 2 Adverse Events, Outcome 12 Patients with fatigue/asthenia.

Comparison 2 Adverse Events, Outcome 13 Patients with leucopenia *.
Figuras y tablas -
Analysis 2.13

Comparison 2 Adverse Events, Outcome 13 Patients with leucopenia *.

Comparison 2 Adverse Events, Outcome 14 Patients with liver dysfunction.
Figuras y tablas -
Analysis 2.14

Comparison 2 Adverse Events, Outcome 14 Patients with liver dysfunction.

Comparison 3 Relapses' outcomes, Outcome 1 Number of patients with at least one relapse during follow.
Figuras y tablas -
Analysis 3.1

Comparison 3 Relapses' outcomes, Outcome 1 Number of patients with at least one relapse during follow.

Comparison 3 Relapses' outcomes, Outcome 2 Relapse rate.
Figuras y tablas -
Analysis 3.2

Comparison 3 Relapses' outcomes, Outcome 2 Relapse rate.

Comparison 4 MRI: role of treatment, Outcome 1 Number of patients with combined lesions at different times (6, 9 and 24 months).
Figuras y tablas -
Analysis 4.1

Comparison 4 MRI: role of treatment, Outcome 1 Number of patients with combined lesions at different times (6, 9 and 24 months).

Comparison 4 MRI: role of treatment, Outcome 2 Mean absolute change of T2 lesion load.
Figuras y tablas -
Analysis 4.2

Comparison 4 MRI: role of treatment, Outcome 2 Mean absolute change of T2 lesion load.

Summary of findings for the main comparison. INTERFERONS for secondary progressive multiple sclerosis

INTERFERONS for secondary progressive multiple sclerosis

Patient or population: patients with secondary progressive multiple sclerosis
Settings:
Intervention: INTERFERONS

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Control

INTERFERONS

Sustained (6 months) EDSS increase after 3 years

Study population

RR 0.98
(0.82 to 1.16)

2026
(3)

HIGH

RCTs low risk of bias

Heterogeneity probably due to different clinical characteristics of patients

410 per 1000

402 per 1000
(336 to 475)

Moderate

372 per 1000

365 per 1000
(305 to 432)

Sustained (3 months) EDSS increase after 3 years

Study population

RR 0.88
(0.8 to 0.97)

1336
(2)

HIGH

579 per 1000

510 per 1000
(463 to 562)

Moderate

594 per 1000

523 per 1000
(475 to 576)

Sustained (3 or 6 months) EDSS increase at 3 years in patients with pre study relapses

Study population

RR 0.9
(0.75 to 1.09)

1106
(3)

HIGH

465 per 1000

419 per 1000
(349 to 507)

Moderate

388 per 1000

349 per 1000
(291 to 423)

Number of patients with at least one relapse during follow‐up ‐ after 3 years

Study population

RR 0.91
(0.84 to 0.97)

2639
(4)

HIGH

530 per 1000

482 per 1000
(445 to 514)

Moderate

509 per 1000

463 per 1000
(428 to 494)

*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. 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).
CI: Confidence interval; RR: Risk ratio;

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

Figuras y tablas -
Summary of findings for the main comparison. INTERFERONS for secondary progressive multiple sclerosis
Table 1. Summary of baseline characteristics of included studies

Study Name

Intervention

Number of patients

% of female

Age ‐ mean (SD)

Baseline EDSS ‐ mean (SD)

Disease duration ‐ mean (SD)

Pre‐study change* in EDSS ‐ mean (SD)

Pre‐study progression duration* ‐ mean (SD)

Pre‐study number of relapses* ‐ mean (SD)

Pre‐study % of patients without relapse*

Andersen 2004 (Nordic)

Rebif 22µg weekly

188

60

 45.1 (nr)

4.7 (nr)

 14.2 (nr)

nr

 4.8 (nr)

1.7 (0.4)

40

Placebo

183

60

46.4 (nr)

5.0 (nr)

.14.4 (nr)

nr

6.1 (nr)

1.6 (0.4)

34

Cohen 2002 (IMPACT)

Avonex 60µg weekly

217

64

47.2 (8.2)

5.2 (1.1)

16.2 (9.0)

nr

nr

1.5 (2.1)

37

Placebo

219

64

47.9 (7.7)

5.2 (1.1)

16.7 (9.0)

nr

nr

1.3 (2.1)

44

North American SG 2004

Betaferon 250µg, every other day  

317

66

46.1 (8.0)

5.2 (1.1)

14.6 (7.8)

1.7 (0.9)

4.0 (3.3)

0.8 (1.1)

54

Betaferon 160µg/m2, every other day

314

61

46.8 (8.3)

5.1 (1.2)

14.5 (8.7)

1.7 (0.9)

4.1 (3.5)

0.9 (1.6)

55

Placebo 

308

60

47.6 (8.2)

5.1 +1.1

14.9 (8.3)

1.7 (0.9)

4.1 (3.5)

0.8 (1.2)

56

SPECTRIMS 2001

Rebif 44µg, three times weekly

204

67

42.6 (7.3)

5.3 + 1.1

12.9 (6.9)

1.5 (0.8)

3.7 (2.7)

0.9 (1.3)

52

Rebif 22µg, three times weekly

209

62

43.1 (7.2)

5.5 (1.1)

13.3 (7.4)

1.6 (0.9)

4.2 (3.1)

0.9 (1.4)

54

Placebo 

205

60

42.7 (6.8)

5.4 (1.1)

13.7 (7.2)

1.7 (1.0)

4.1 (3.2)

0.9 (1.2)

52

The European SG 1998

Betaferon 250µg, every other day  

360

58·1

 41·1 (7·2)

5·1 (1·1)

12.8 (6.6)

1.5 (nr)

3·8 (2·7)

1.7 (0.85)

32

Placebo

358

64·2

40·9 (7·2)

5·2 (1·1)

13.4 (7.5)

3·8 (3·4)

28

* Pre‐study length of observation: 4 years (Andersen 2004 (Nordic)) ; 3 years (Cohen 2002 (IMPACT) ; 2 years (North American SG 2004 ; SPECTRIMS 2001 ; The European SG 1998)

Figuras y tablas -
Table 1. Summary of baseline characteristics of included studies
Comparison 1. Disability progression

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Sustained (6 months) EDSS increase after 3 years Show forest plot

3

2026

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.82, 1.16]

2 Sustained (3 or 6 months) EDSS increase at 3 years in patients with or without pre‐study relapses Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 In patients with pre study relapses

3

1106

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.75, 1.09]

2.2 In patients without pre‐study relapses

3

903

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.83, 1.33]

3 Sustained (3 months) EDSS increase Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 After 2 years

2

1054

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.81, 1.08]

3.2 After 3 years

2

1336

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.80, 0.97]

4 Sustained (3 or 6 months') EDSS increase according to pre‐study clinical characteristics of patients Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 low age and disease duration

2

1336

Risk Ratio (M‐H, Fixed, 95% CI)

0.92 [0.83, 1.02]

4.2 high age and disease duration

3

1739

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.90, 1.18]

Figuras y tablas -
Comparison 1. Disability progression
Comparison 2. Adverse Events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Total number of patients with Serious AEs Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.83, 1.19]

2 Patients who had discontinuated for AEs (including SAEs) * Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

2.62 [1.92, 3.57]

3 Deaths Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.41 [0.58, 3.42]

4 Patients who done or attempted suicide Show forest plot

4

2441

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.36, 1.95]

5 Allergy/Rash (number of events) Show forest plot

2

1657

Risk Ratio (M‐H, Random, 95% CI)

1.57 [0.99, 2.48]

6 Cutaneous necrosis * Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 Number of events

2

1336

Risk Ratio (M‐H, Random, 95% CI)

33.02 [4.57, 238.79]

6.2 Number of patients

2

1557

Risk Ratio (M‐H, Random, 95% CI)

18.76 [3.72, 94.61]

7 Injection site reactions * Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 Number of events

2

1657

Risk Ratio (M‐H, Random, 95% CI)

3.84 [3.11, 4.74]

7.2 Number of patients

3

1425

Risk Ratio (M‐H, Random, 95% CI)

1.60 [0.84, 3.07]

8 Patients with psychiatric disorders Show forest plot

5

3082

Risk Ratio (M‐H, Random, 95% CI)

1.05 [0.94, 1.18]

9 Patients with headache Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.24 [0.97, 1.60]

10 Patients with influenza like syndrome * Show forest plot

4

2364

Risk Ratio (M‐H, Random, 95% CI)

1.45 [1.01, 2.07]

11 Patients with myalgia Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.30 [0.93, 1.83]

12 Patients with fatigue/asthenia Show forest plot

3

1746

Risk Ratio (M‐H, Random, 95% CI)

1.09 [0.98, 1.22]

13 Patients with leucopenia * Show forest plot

3

1921

Risk Ratio (M‐H, Random, 95% CI)

2.25 [1.06, 4.75]

14 Patients with liver dysfunction Show forest plot

2

1310

Risk Ratio (M‐H, Random, 95% CI)

1.69 [0.97, 2.95]

Figuras y tablas -
Comparison 2. Adverse Events
Comparison 3. Relapses' outcomes

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of patients with at least one relapse during follow Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 After 2 years

1

436

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.54, 0.95]

1.2 After 3 years

4

2639

Risk Ratio (M‐H, Random, 95% CI)

0.91 [0.84, 0.97]

2 Relapse rate Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 After 2 years

1

436

Mean Difference (IV, Random, 95% CI)

‐0.12 [‐0.20, ‐0.04]

2.2 After 3 years

3

1752

Mean Difference (IV, Random, 95% CI)

‐0.16 [‐0.21, ‐0.10]

Figuras y tablas -
Comparison 3. Relapses' outcomes
Comparison 4. MRI: role of treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Number of patients with combined lesions at different times (6, 9 and 24 months) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 6 months

1

125

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.52, 0.95]

1.2 9 months

1

264

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.42, 0.62]

1.3 24 months

1

109

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.33, 0.80]

2 Mean absolute change of T2 lesion load Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

2.1 1 year

2

1022

Mean Difference (IV, Random, 95% CI)

‐1.74 [‐3.25, ‐0.23]

2.2 2 years

2

956

Mean Difference (IV, Random, 95% CI)

‐2.56 [‐5.08, ‐0.05]

2.3 3 years

1

567

Mean Difference (IV, Random, 95% CI)

‐4.87 [‐6.22, ‐3.52]

Figuras y tablas -
Comparison 4. MRI: role of treatment