Scolaris Content Display Scolaris Content Display

Alemtuzumab bei Multipler Sklerose

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

CAMMS223 {published data only}

Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, Margolin DH, et al. Alemtuzumab vs. interferon beta‐1a in early multiple sclerosis. New England Journal of Medicine 2008;359(17):1786‐801.
Coles AJ, Fox E, Vladic A, Gazda SK, Brinar V, Selmaj KW, et al. Alemtuzumab more effective than interferon β‐1a at 5‐year follow‐up of CAMMS223 clinical trial. Neurology 2012;78:1069–78.
Coles AJ, Fox E, Vladic A, Gazda SK, Brinar V, Selmaj KW, et al. Alemtuzumab versus interferon beta‐1a in early relapsing‐remitting multiple sclerosis: post‐hoc and subset analyses of clinical efficacy outcomes. Lancet Neurology 2011;10(4):338‐48.
Daniels GH, Vladic A, Brinar V, Zavalishin I, Valente W, Oyuela P, et al. Alemtuzumab‐related thyroid dysfunction in a phase 2 trial of patients with relapsing‐remitting multiple sclerosis. Journal of Clinical Endocrinology and Metabolism  2014;99(1):80‐9.

CARE‐MS I {published data only}

Arnold D, Brinar V, Cohen J, Coles A, Confavreux C, Fisher E, et al. Effect of alemtuzumab vs. RebifTM on brain MRI measurements: results of CARE‐MS I, a phase 3 study. Neurology 2012;78(1):S11.006.
Cohen JA, Coles AJ, Arnold DL, Confavreux C, Fox EJ, Hartung HP, et al. Alemtuzumab versus interferon beta 1a as first‐line treatment for patients with relapsing‐remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet 2012;380(9856):1819‐28.
Coles A, Brinar V, Arnold D, Cohen J, Confavreux C, Fox E, et al. Efficacy and safety results from comparison of alemtuzumab and RebifTM efficacy in multiple sclerosis I (CARE‐MS I): a phase 3 study in relapsing‐remitting treatment‐naive patients. Neurology 2012;78(Suppl 1):S01.006.
Fox E, Arnold D, Brinar V, Cohen J, Coles A, Confavreux C, et al. Relapse outcomes with alemtuzumab vs. RebifTM in treatment‐naive relapsing‐remitting multiple sclerosis (CARE‐MS I): secondary and tertiary endpoints. Neurology 2012;78(Suppl 1):PD5.004. [DOI: 10.1212/WNL.78.1_MeetingAbstracts.PD5.004]
Giovannoni G, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox HP, et al. Disease activity‐free status in comparison of alemtuzumab and RebifTM efficacy in multiple sclerosis I (CARE‐MS I) phase 3 study. Journal of Neurology 2012;259(Suppl 1):47.
Habek M, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Thyroid autoimmunity in comparison of Alemtuzumab and RebifTM efficacy in multiple sclerosis studies I and II. Journal of Neurology 2012;259(Suppl 1):66.
Havrdova E, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Infections Phase 3 study: comparison of alemtuzumab and RebifTM efficacy in multiple sclerosis I (CARE‐MS I). Neurology 2012;78(Suppl 1):S41.007.
Lycke J, Arnold DL, Cohen JA, Coles AJ, Confavreux C, Fox EJ, et al. Adverse event profile of alemtuzumab over time in treatment‐naive patients with early, active relapsing‐remitting multiple sclerosis (RRMS; CARE‐MS I study). Journal of the Neurological Sciences 2013;333(Suppl 1):e374‐5.
Lycke J, Arnold DL, Cohen JA, Coles AJ, Fox EJ, Hartung HP, et al. Adverse event profile of alemtuzumab in active relapsing remitting multiple sclerosis patients who participated in the CARE‐MS studies: three‐year follow‐up. 29th Congress or the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS), 2‐5 October 2013, Copenhagen, Denmark. 2013.
Miller T, Arnold D, Cohen J, Coles A, Confavreux C, Fox E, et al. Detection, incidence, and management of thyroid autoimmunity in comparison of alemtuzumab and RebifTM in multiple sclerosis (CARE‐MS) I and II. Neurology 2013;80(1):P01.173.
Selmaj K, Arnold DL, Brinar V, Cohen J, Coles AJ, Confavreux C, et al. Incidence of autoimmunity in a phase 3 trial: comparison of alemtuzumab and RebifTM in multiple sclerosis I (CARE‐MS I). Neurology 2012;78(1):S41.006.
Selmaj K, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Alemtuzumab improves patient‐reported quality of life in relapsing‐remitting multiple sclerosis: CARE‐MS I and II phase 3 trials. Journal of Neurology 2012;259(Suppl 1):S65‐6.

CARE‐MS II {published data only}

Arnold DL, Cohen J, Coles AJ, Confavreux C, Fisher E, Fox EJ, et al. Effect of alemtuzumab vs. Rebif® on brain MRI measurements. Multiple Sclerosis 2012;18(4):397.
Arroyo R, Arnold DL, Cohen JA, Coles AJ, Confavreux C, Fox EJ, et al. Alemtuzumab improves quality of life compared to SC IFNB‐1a in CARE‐MS II. Journal of Neurology 2013;260:S121‐2.
Barkhof F, Fisher E, Palmer J, Margolin DH, Arnold DL. Alemtuzumab demonstrates improvement in MRI outcomes across baseline subgroups versus subcutaneous interferon beta‐1a in relapsing‐remitting multiple sclerosis patients who relapsed on prior therapy. European Journal of Neurology 2014;21:126‐7.
Brinar V, Arnold DL, Cohen J, Coles AJ, Fox EJ, Hartung HP, et al. Alemtuzumab improves expanded disability status scale (EDSS) via effects on functional systems: CARE‐MS II. Multiple Sclerosis. Multiple Sclerosis Journal 2013;19(S1):283‐4.
Coles AJ, Twyman CL, Arnold DL, Cohen JA, Confavreux C, Fox EJ, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease‐modifying therapy: a randomised controlled phase 3 trial. Lancet 2012;380:1829‐39.
Confavreux C, Twyman CL, Arnold D, Cohen J, Coles AJ, Fox EJ, et al. Efficacy of alemtuzumab in relapsing remitting multiple sclerosis (RRMS) patients with highly active disease despite therapy. European Journal of Neurology 2012;19(Suppl 1):458.
Fernandez O, Arnold DL, Cohen JA, Coles AJ, Confavreux C, Fox EJ, et al. Alemtuzumab improves disability by month 6 independent of relapse history in relapsing‐remitting multiple sclerosis patients: CARE‐MS II 20620. Journal of Neurology 2013;260:S14.
Fisher E, Barkhof F, Cohen JA, Fox EJ, Selmaj KW, Margolin DH, et al. Alemtuzumab improves MRI outcomes in relapsing‐remitting multiple sclerosis patients who relapsed on prior therapy: three‐year follow‐up of CARE‐MS II. Multiple Sclerosis Journal 2014;20(Suppl 1):67.
Giovannoni G, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox E, et al. Disability improvement with alemtuzumab vs. interferon b‐ 1a in relapsing‐remitting multiple sclerosis patients who relapsed on prior therapy (CARE‐MS II). Multiple Sclerosis 2012;18(S1):419.
Giovannoni G, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox E, et al. Multiple sclerosis: clinical trials outcomes disability improvement with alemtuzumab vs. interferon beta‐1a in relapsing‐remitting multiple sclerosis patients who experienced disease activity while on prior therapy (CARE‐MS II). Neurology 2013;80(Suppl 1):P07.120.
Habek M, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Thyroid autoimmunity in comparison of alemtuzumab and RebifTM efficacy in multiple sclerosis studies I and II. Journal of Neurology 2012;259(Suppl 1):S66.
Hartung H, Vollmer T, Arnold D, Cohen J, Coles A, Confavreux C, et al. Alemtuzumab reduces ms disease activity in active relapsing‐remitting multiple sclerosis patients who had disease activity on prior therapy. Neurology 2013;80(Suppl 1):P07.093.
Hartung HP, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Disability outcomes for alemtuzumab in RRMS patients who relapsed on prior therapy: CARE‐MS II. Journal of Neurology 2012;259(Suppl 1):S47‐8.
Havrdova E, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Safety of alemtuzumab in relapsing‐remitting multiple sclerosis patients who relapsed on prior therapy (CARE‐MS II). Multiple Sclerosis 2012;18(4):235.
Lycke J, Arnold DL, Cohen JA, Coles AJ, Fox EJ, Hartung HP, et al. Adverse event profile of alemtuzumab in active relapsing remitting multiple sclerosis patients who participated in the CARE‐MS studies: three‐year follow‐up. 29th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS), 2‐5 October 2013, Copenhagen, Denmark 2013;19(S1):487‐8.
Miller T, Arnold D, Cohen J, Coles A, Confavreux C, Fox E, et al. Detection, incidence, and management of thyroid autoimmunity in comparison of alemtuzumab and RebifTM in multiple sclerosis (CARE‐MS) I and II. Neurology 2013;80(1):P01.173.
Moreau T, Margolin DH, Kasten L, Singer B. Alemtuzumab improves quality of life in relapsing remitting multiple sclerosis patients who relapsed on prior therapy: 3‐year follow‐up of CARE‐MS II. Multiple Sclerosis 2014;20(1):86.
Selmaj K, Arnold DL, Cohen J, Coles AJ, Confavreux C, Fox EJ, et al. Alemtuzumab improves patient‐reported quality of life in relapsing‐remitting multiple sclerosis: CARE‐MS I and II phase 3 trials. Journal of Neurology 2012;259(Suppl 1):S65‐6.
Wray S, Arnold D, Cohen J, Coles A, Confavreux C, Fox E, et al. Comparison of infection risk with alemtuzumab and sc IFNB‐1a in patients with multiple sclerosis who experienced disease activity while on prior therapy (CARE‐MS II). Neurology 2013;80(1):P01.172.

Additional references

CADTH 2013

Canadian Agency for Drugs and Technologies in Health. Management of relapsing‐remitting multiple sclerosis. http://www.ncbi.nlm.nih.gov/books/NBK169748/ (accessed 11 March 2016).

Coles 1999a

Coles AJ, Wing M, Smith S, Coraddu F, Greer S, Taylor C, et al. Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet 1999;354:1691‐5.

Coles 1999b

Coles AJ, Wing MG, Molyneux P, Paolillo A, Davie CM, Hale G, et al. Monoclonal antibody treatment exposes three mechanisms underlying the clinical course of multiple sclerosis. Annals of Neurology 1999;46:296‐304.

Coles 2006

Coles AJ, Cox A, Le Page E, Jones J, Trip SA, Deans J, et al. The window of therapeutic opportunity in multiple sclerosis: evidence from monoclonal antibody therapy. Journal of Neurology 2006;253:98‐108.

Cossburn 2011

Cossburn M, Pace AA, Jones J, Ali R, Ingram G, Baker K, et al. Autoimmune disease after alemtuzumab treatment for multiple sclerosis in a multicenter cohort. Neurology 2011;77(6):573‐9.

EMA 2013

European Medicines Agency. Lemtrada ‐ Committee for Medicinal Products for Human Use (CHMP) ‐ Assessment report. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_‐_Public_assessment_report/human/003718/WC500150522.pdf (accessed 11 March 2016).

FDA 2001

US Food, Drug Administration. Campath (alemtuzumab) Product Approval Information ‐ Application number BLA 103948/0. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/103948_0000_Campath_AprvbleLtr2.pdf (accessed 11 March 2016).

FDA 2014

US Food, Drug Administration. Alemtuzumab (Lemtrada) Product Approval Information. Licensing Action 2014. http://www.accessdata.fda.gov/drugsatfda_docs/appletter/2014/103948Orig1s5139ltr.pdf (accessed 11 March 2016).

FDA 2015

US Food, Drug Administration. Risk Evaluation and Mitigation Strategies (REMS). Lemtrada 2015. http://www.accessdata.fda.gov/scripts/cder/rems/index.cfm?event=IndvRemsDetails.page&REMS=340 (accessed 11 March 2016).

Filippini 2013

Filippini G, Del Giovane C, Vacchi L, D'Amico R, Di Pietrantonj C, Beecher D, et al. Immunomodulators and immunosuppressants for multiple sclerosis: a network meta‐analysis. Cochrane Database of Systematic Reviews 2013, Issue 6. [DOI: 10.1002/14651858.CD008933.pub2]

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;5:251‐9.

Genzyme 2013

Peripheral and Central Nervous System Drugs Advisory Committee. Alemtuzumab Advisory Committee Briefing Document. BLA 103948. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/PeripheralandCentralNervousSystemDrugsAdvisoryCommittee/UCM374188.pdf (accessed 11 March 2016).

Gilleece 1993

Gilleece M, Dexter T. Effect of campath‐1h antibody on human hematopoietic progenitors in vitro. Blood 1993;82:807–12.

Gomez‐Almaguer 2012

Gomez‐Almaguer D, Jaime‐Perez J, Ruiz‐Arguelles G. Antibodies in the treatment of aplastic anaemia. Archivum Immunologiae et Therapiae Experimentalis 2012;60:99‐106.

GRADEpro 2008 [Computer program]

GRADE Working Group. GRADEpro. Version 3.2 for Windows. GRADE Working Group, 2008.

Gray 2004

Gray O, McDonnell GV, Forbes RB. Methotrexate for multiple sclerosis. Cochrane Database of Systematic Reviews 2004, Issue 2. [DOI: 10.1002/14651858.CD003208.pub2]

Hawkins 1999

Hawkins SA, McDonnell GV. Benign multiple sclerosis? Clinical course, long term follow up, and assessment of prognostic factors. Journal of Neurology Neurosurgery and Psychiatry 1999;67:148‐52.

Healy 2013

Healy BC, Engler D, Glanz B, Musallam A, Chitnis T. Assessment of definitions of sustained disease progression in relapsing‐remitting multiple sclerosis. Multiple Sclerosis International 2013;2013:189624. [PUBMED: 23555057]

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. www.cochrane‐handbook.org.

Hill‐Cawthorne 2012

Hill‐Cawthorne GA, Button T, Tuohy O, Jones JL, May K, Somerfield J, et al. Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis. Journal of Neurology, Neurosurgery and Psychiatry 2012;83(3):298‐304.

Hirst 2008

Hirst CL, Pace A, Pickersgill TP, Jones R, McLean BN, Zajicek JP, et al. Campath1‐H treatment in patients with aggressive relapsing remitting multiple sclerosis. Journal of Neurology 2008;255:231‐8.

Keating 2002

Keating M, Flinn I, Jain V, Binet J, Hillmen P, Byrd J, et al. Therapeutic role of alemtuzumab (Campath‐1H) in patients who have failed fludarabine: results of a large international study. Blood 2002;99:3554–61.

Krupp 1989

Krupp LB, LaRocca NG, Muir‐Nash J, Steinberg AD. The fatigue severity scale. Application to patients with multiple sclerosis and systemic lupus erythematosus. Archives of Neurology 1989;46:1121–3.

Kurtzke 1983

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

Li 1999

Li DK, Paty DW, the UBC MS/MRI Analysis Research Group and the PRISMS Study Group. Magnetic resonance imaging results of the PRISMS trial: a randomised, double‐blind, placebo‐controlled study of interferon‐beta la in relapsing–remitting multiple sclerosis. Annals of Neurology 1999;46:197‐206.

Lockwood 2003

Lockwood C, Hale G, Waldmann H, Jayne D. Remission induction in Behcet’s disease following lymphocyte depletion by the anti‐CD52 antibody CAMPATH‐1H. Rheumatology 2003;42:1539‐44.

Lublin 1996

Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. Neurology 1996;46:907‐11.

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;50:121‐7.

Multiple Sclerosis International Federation 2010

Multiple Sclerosis International Federation (MSIF). Global Economic Impact of Multiple Sclerosis. http://www.msif.org/wp‐content/uploads/2014/09/Global_economic_impact_of_MS.pdf (accessed 11 March 2016).

Noseworthy 2000

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

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, McDonal WI, Davis FA, Ebers GC. New diagnostic criteria for multiple sclerosis: guidelines for research protocols. Annals of Neurology 1983;13:227‐31.

Rao 2012

Rao SP, Sancho J, Campos‐Rivera J, Boutin PM, Severy PB, Weeden T, et al. Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS One 2012;7(6):e39416.

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.

Rieckmann 2009

Rieckmann P. Concepts of induction and escalation therapy in multiple sclerosis. Journal of Neurosurgical Sciences 2009;277(Suppl 1):42‐5.

Scolding 2015

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

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.

Waldmann 2005

Waldmann H, Hale G. Campath: from concept to clinic. Philosophical transactions of the Royal Society B: Biological Sciences 2005;360:1707–11.

Weissenbacher 2010

Weissenbacher A, Boesmueller C, Brandacher G, Oellinger R, Pratschke J, Schneeberger S. Alemtuzumab in solid organ transplantation and in composite tissue allotransplantation. Immunotherapy 2010;2:783‐90.

Xia 1991

Xia M, Tone M, Packman L, Hale G, Waldmann H. Characterization of the Campath‐1 (CDW2) antigen: biochemical analysis and cDNA cloning reveal an unusually small peptide backbone. European Journal of Immunology 1991;21:1677‐84.

References to other published versions of this review

Riera 2014

Riera R, Porfirio G, Migliorini CR, Torloni MR. Alemtuzumab for multiple sclerosis. Cochrane Database of Systematic Reviews 2014, Issue 7. [DOI: 10.1002/14651858.CD011203]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

CAMMS223

Methods

  • CAMMS223 (primary reference)

  • Multicentric, phase II, randomised clinical trial

  • 49 centres in Europe and the United States

  • Randomisation ratio 1:1:1, stratification to balance the study groups with regard to age (< 30 years or ≥ 30 years), gender and baseline EDSS scores (< 2.0 or ≥ 2.0)

  • N = 334/available for analysis = 333

  • Treatment duration: 24 months

  • Follow‐up duration: 36 months

Participants

Inclusion criteria

  • Diagnosis of relapsing–remitting MS based on the McDonald criteria

  • Onset of symptoms no more than 36 months before the time of screening; at least 2 clinical episodes during the previous 2 years; a score of 3 or less on the EDSS, which ranges from 0 to 10, with higher scores indicating greater disability; and one or more enhancing lesions, as seen on at least one of up to 4 monthly cranial magnetic resonance imaging (MRI) scans.

Exclusion criteria

  • Previous disease‐modifying treatments

  • History of clinically significant autoimmunity

  • Presence of serum antithyrotropin‐receptor antibodies

Interventions

Main interventions 

  • Alemtuzumab 12 mg administered IV, once a day for 5 consecutive days at the first month and for 3 consecutive days at months 12 and 24 (the latter at the treating physicians' discretion if the CD4+ T‐cell count was ≥ 100 × 106 cells/L) (n = 113/available for analysis = 112)

  • Alemtuzumab 24 mg administered IV, once a day for 5 consecutive days at the first month and for 3 consecutive days at months 12 and 24 (the latter at the treating physicians' discretion if the CD4+ T‐cell count was ≥ 100 × 106 cells/L) (n = 110/available for analysis = 110)

Comparator

  • Interferon beta‐1a (44 μg) administered subcutaneously 3 times weekly after dose escalation (n = 111/available for analysis = 111)

All participants received 1 g of intravenous methylprednisolone for 3 days at baseline and at months 12 and 24, coinciding with infusion cycles as premedication for those receiving alemtuzumab. Some participants also received antihistamines or antipyretics at the investigators' discretion

Outcomes

Primary outcome measure

  • Rate of participants with sustained accumulation of disability: Disability was assessed according to the ordinal EDSS score. A sustained accumulation of disability was defined as an increase of at least 1.5 points for participants with a baseline score of 0 and of at least 1.0 point for participants with a baseline score of 1.0 or more; all scores were confirmed twice during a 3‐ and 6‐month period. The onset of a sustained level of disability was timed to the first recorded increase in the EDSS score aside from relapse.

  • Rate of relapse: Relapse was defined as new or worsening symptoms with an objective change in neurologic examination attributable to multiple sclerosis that lasted for at least 48 hours, that were present at normal body temperature, and that were preceded by at least 30 days of clinical stability.

Secondary outcome measures

  • Proportion of participants who did not have a relapse (proportion of participants with relapse‐free survival)

  • Changes in lesion burden (as seen on T2‐weighted MRI)

  • Brain volume (as measured by the Losseff method on T1‐weighted MRI8)

  • Adverse effects, including the following measures:

      • Thyroid function and levels of antithyrotropin receptor antibodies and lymphocyte subpopulations were measured quarterly at a central laboratory

      • Serum‐binding antibodies against alemtuzumab were measured with the use of a validated enzyme‐linked immunosorbent assay (ELISA) at BioAnaLab

      • Immune thrombocytopenia by single confirmed platelet count of fewer than 50,000 per microlitre without clumping or a platelet count of more than 50,000 but fewer than 100,000 per microlitre on at least 2 consecutive occasions during a period of at least 1 month, with normal haemoglobin, neutrophil and eosinophil counts; an absence of splenomegaly; and a normal peripheral‐blood smear (apart from thrombocytopenia).

All adverse events with an onset up to 36 months were reported. In addition, all serious adverse events and autoimmune‐associated disorders occurring before 1 March 2008, were listed. A subsequent adverse event of Burkitt's lymphoma not associated with Epstein–Barr virus (EBV) was also included in this report.

Notes

  • The effectiveness of blinding was assessed at the end‐of‐study visit.

  • Participants with an increased level of disability could be discontinued from the study.

  • There was no active monitoring for progressive multifocal leukoencephalopathy.

  • Preplanned interim analyses were performed when most participants had completed at least 1 year and 2 years with a prespecified alpha spending function. Disclosure of these results formed part of safety announcements by the sponsor in September 2005 and 2006. After the interim analyses, P values of less than 0.016 and 0.004 were considered to have statistical significance for the rates of sustained disability and relapse, respectively.

  • In September 2005, the data and safety monitoring board recommended suspension of alemtuzumab treatments after receiving reports of 3 cases of immune thrombocytopenic purpura, including 1 death. All safety and efficacy assessments proceeded as planned and participants who were receiving interferon beta‐1a continued to receive the drug. At the time of dose suspension, only 2 eligible participants (1%) had not received the second cycle of alemtuzumab at month 12, whereas 155 participants (75%) were precluded from receiving the third cycle of alemtuzumab at month 24.

  • More participants discontinued interferon beta‐1a than alemtuzumab, principally because of a lack of efficacy and adverse events, so that only 59% of the original group of participants receiving interferon beta‐1a completed the 36‐month study, as compared with 83% of participants receiving alemtuzumab. At the end of the study review, 90% and 91% of raters remained unaware of assignments to the group receiving interferon beta‐1a and the group receiving alemtuzumab, respectively.

  • Funding: Genzyme

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Eligible participants were randomly assigned in a 1:1:1 with the use of the Pocock and Simon minimisation algorithm.

Allocation concealment (selection bias)

Unclear risk

No information available to allow a judgement

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Both study drugs have adverse effects that precluded masking of participants and treating clinicians to treatment assignment

Blinding of outcome EDSS assessment
EDSS

Low risk

"EDSS scores were determined quarterly in a blinded fashion by a neurologist who also adjudicated possible relapses".

Blinding of outcome assessment (detection bias)
All outcomes, except EDSS

Low risk

"MRI scans were performed annually and interpreted by a neuroradiologist who was unaware of assignments to study groups".

Blinding of safety outcome assessment

High risk

"Safety was assessed quarterly by the treating neurologist, who was aware of study‐group assignment".

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Number and reasons of withdrawals were similar in the interventions and control arms.

Selective reporting (reporting bias)

Low risk

Other bias

Low risk

CARE‐MS I

Methods

  • CARE‐MS I (primary reference)

  • Multicentric, phase III, randomised clinical trial

  • 101 academic medical centres and clinical practices in 16 countries

  • Randomisation ratio 2:1; stratification by site

  • N = 581/available for analysis = 563

  • Treatment duration: 12 months

  • Follow‐up duration: 24 months

Participants

Inclusion criteria

  • 18 to 50 years of age

  • Diagnosis of relapsing‐remitting multiple sclerosis fulfilling the McDonald criteria

  • Disease duration of up to 5 years

  • At least 2 relapses in the previous 2 years and at least 1 in the previous year

  • Expanded disability status scale (EDSS) 10 scores of 3.0 or lower

  • Cranial abnormalities on MRI attributable to multiple sclerosis

Exclusion criteria

  • Progressive disease course

  • Previous multiple sclerosis disease therapy (apart from corticosteroids)

  • Previous immunosuppressive, investigational or monoclonal antibody therapy

  • Clinically significant autoimmunity other than multiple sclerosis

Interventions

Main interventions 

  • Alemtuzumab 12 mg administered IV, once a day for 5 consecutive days at baseline and for 3 consecutive days at 12 months (n = 386/available for analysis = 376)

After a protocol amendment in January 2009, alemtuzumab participants received oral aciclovir 200 mg twice daily during alemtuzumab infusion and for 28 days thereafter as prophylaxis against herpes infection.

Comparator

  • Interferon beta‐1a 44 μg given subcutaneously 3 times weekly after dose titration (n = 195/available for analysis = 187)

Participants in both groups received 1 g per day of intravenous methylprednisolone on 3 consecutive days at baseline and at month 12. Concomitant treatment with an antipyretic or antihistamine drug was allowed, at the discretion of the treating neurologist.

Outcomes

Primary outcome measures

  • Relapse rate, defined as new or worsening neurological symptoms attributable to MS, lasting at least 48 hours, without pyrexia, after at least 30 days of clinical stability, with an objective change on neurological examination assessed by a masked rater. The relapse adjudication panel decided the status of suspected relapses on the basis of the protocol definition and their masked review of all data collected by the site, including whether there was an objective change corresponding to current relapse symptoms (1 point on 2 functional system scales or 2 points on 1 functional system scale or increase in the EDSS score).

  • Sustained accumulation of disability, defined as an increase from baseline of at least 1 EDSS point (or ≥1.5 points if baseline EDSS score was 0) confirmed over 6 months.

Secondary outcome measure (24 months)

  • Rate of relapse‐free participants

  • Change in EDSS

  • Number of participants with new or enlarging T2‐hyperintense lesions on magnetic resonance imaging

  • Change in multiple sclerosis functional composite (MSFC)

  • Freedom from clinical disease activity, defined as absence both of relapses and sustained accumulation of disability

  • Freedom from MRI disease activity, defined as absence both of gadolinium‐enhancing lesions and new or enlarging T2‐hyperintense lesions

  • Adverse events, actively searched by a monthly questionnaire follow‐up of participants, complete blood counts, serum creatinine, urinalysis and microscopy monthly (every 3 months for participants in the interferon beta‐1a group), and thyroid function tests every 3 months

Notes

  • Raters completed a questionnaire assessing quality of the masking at each EDSS assessment

  • All participants who received at least one dose of study drug were included in the efficacy and safety analyses according to treatment assignment ("modified ITT analysis")

  • Funding: Genzyme. The study sponsor was involved in the design and undertaking of the trial, data analysis and interpretation, writing of the manuscript, and the decision to submit the manuscript for publication. Bayer Schering Pharma participated in the design and oversight of the trial. Clinical investigators collaborated with the sponsor to design and oversee the trial. The sponsor did the statistical analyses.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"We randomly allocated patients using an interactive voice response system"

Allocation concealment (selection bias)

Low risk

Use of an interactive voice response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

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

Blinding of outcome EDSS assessment
EDSS

High risk

"In the absence of a masked rater, unmasked raters could submit EDSS assessments"

Blinding of outcome assessment (detection bias)
All outcomes, except EDSS

Low risk

"Stringent clinical and MRI rater masking, and adjudication of relapses by a committee comprising six independent and masked neurologists"

Blinding of safety outcome assessment

Low risk

"Stringent clinical and MRI rater masking, and adjudication of relapses by a committee comprising six independent and masked neurologists"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only participants who received at least one dose of study drugs were included in the efficacy and safety analyses according to treatment assignment (modified ITT analysis)

Selective reporting (reporting bias)

Low risk

Other bias

Low risk

CARE‐MS II

Methods

  • CARE‐MS II (primary reference)

  • Multicentric, phase III, randomised clinical trial

  • 194 centres around the world

  • N = 798/available for analysis = 667

  • Randomisation ratio 1:2:2

  • Treatment duration: 12 months

  • Follow‐up: 24 months

Participants

Inclusion criteria:

  • 18 to 55 years of age

  • Diagnosis of relapsing‐remitting MS fulfilling the McDonald diagnostic criteria

  • Disease duration of 10 years or less

  • At least 2 relapses in the previous 2 years, with at least 1 in the previous year

  • At least 1 relapse while on interferon beta or glatiramer acetate after at least 6 months of treatment

  • Expanded Disability Status Scale (EDSS) scores of 5.0 or less

  • Cranial and spinal MRI lesions fulfilling protocol‐defined criteria (MRI scan demonstrating white matter lesions attributable to MS)

Exclusion criteria:

  • Previous treatment with alemtuzumab

  • Previous treatment with any investigational drug (i.e. a medication that is not approved at any dose or for any indication)

  • Treatment with natalizumab, methotrexate, azathioprine or cyclosporine in the past 6 months

  • Previous treatment with mitoxantrone, cyclophosphamide, cladribine, rituximab or any other immunosuppressive, or cytotoxic therapy (other than steroid treatment)

  • Any progressive form of MS

  • Any disability acquired from trauma or another illness that could interfere with evaluation of disability due to MS

  • Major systemic disease that cannot be treated or adequately controlled by therapy

  • Active infection or high risk of infection

  • Autoimmune disorder (other than MS)

  • Impaired hepatic or renal function

  • History of malignancy, except basal skin cell carcinoma

  • Medical, psychiatric, cognitive or other conditions that compromise the patient's ability to understand the patient information, to give informed consent, to comply with the trial protocol or to complete the study

  • Known bleeding disorder

  • Women of childbearing potential with a positive serum pregnancy test, pregnant or lactating

  • Current participation in another clinical study or previous participation in CAMMS323 (NCT00530348)

  • Previous hypersensitivity reaction to any immunoglobulin product

  • Known allergy or intolerance to interferon beta, human albumin or mannitol

  • Intolerance of pulsed corticosteroids, especially a history of steroid psychosis

  • Inability to self administer subcutaneous (SC) injections or receive SC injections from caregiver

  • Inability to undergo MRI with gadolinium administration

  • Unwilling to use a reliable and acceptable contraceptive method throughout the study period (fertile participants only)

Interventions

Main Interventions 

  • Alemtuzumab 12 mg per day administered IV, once a day for 5 consecutive days at Month 0 and 12 mg per day administered through IV, once a day for 3 consecutive days at Month 12 (n = 436, available for analysis = 426)

  • Alemtuzumab  24 mg per day administered IV, once a day for 5 consecutive days at Month 0 and 24 mg per day administered IV, once a day for 3 consecutive days at Month 12 (n = 173, available for analysis = 170)

 Alemtuzumab was administered in 2 annual cycles, once at the beginning of the study and again 1 year later

 

Comparator

  • Interferon beta‐1a (Rebif®) 44 µg administered 3 times weekly by subcutaneous self injections (n = 231, available for analysis = 202)

 

Outcomes

 Primary outcome measure 

  • Time to Sustained Accumulation of Disability (SAD) (time frame: 2 years)

  • Relapse rate (time frame: 2 years). Relapse was defined as new or worsening symptoms with an objective changes in neurologic examination attributable to multiple sclerosis that lasted for at least 48 hours, that were present at normal body temperature and that were preceded by at least 30 days of clinical stability.

Secondary outcome measures

  • Rate of participants who are relapse‐free at year 2 (time frame: 2 years)

  • Change from baseline in EDSS (Expanded Disability Status Scale) (time frame: 2 years)

  • Acquisition of disability as measured by change from baseline in Multiple Sclerosis Functional Composite (MSFC) (time frame: 2 years)

  • Number of participants with new or enlarging T2‐hyperintense lesions in magnetic resonance

  • Adverse effects

  • Quality of life: assessed by Functional Assessment of Multiple Sclerosis (FAMS; scale 0 to 176 for total score); Medical Outcomes Study 36‐Item Short‐Form Survey (SF‐36; scale 1 to 100; healthy population mean = 50; administered annually in the core study); and EuroQol in 5 Dimensions visual analogue scale (EQ‐5D VAS; scale 0 to 100).

Co‐primary endpoints were relapse rate and time to 6‐month sustained accumulation of disability, comparing alemtuzumab 12 mg and interferon beta‐1a in all participants who received at least one dose of study drug.

Notes

  • The 24 mg per day group was discontinued to aid recruitment, but data were included for safety assessments. The decision to close recruitment into the alemtuzumab 24 mg arm was made by the Neurology Steering Committee and Genzyme management without review of safety or efficacy data from this study.

  • Raters completed a questionnaire assessing quality of the masking at each EDSS assessment.

  • Funding: Genzyme (Sanofi) and Bayer ‐ Schering Pharma.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"We randomly allocated patients with an interactive voice response system in a 2:2:1 scheme"

Allocation concealment (selection bias)

Low risk

Use of an interactive voice response system

Blinding of participants and personnel (performance bias)
All outcomes

High risk

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

Blinding of outcome EDSS assessment
EDSS

High risk

"In the absence of a masked rater, unmasked raters could submit EDSS assessments"

Blinding of outcome assessment (detection bias)
All outcomes, except EDSS

Low risk

"Raters were masked to treatment‐group assignment"

Blinding of safety outcome assessment

Low risk

"Raters were masked to treatment‐group assignment"

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Selective reporting (reporting bias)

High risk

Efficacy outcomes for alemtuzumab 24 mg were not provided.

The results from some previously planned outcomes were not provided (i.e. quality of life)

Other bias

Low risk

EDSS: Expanded Disability Status Scale
ITT: intention‐to treat
IV: intravenous
MRI: magnetic resonance imaging
MS: multiple sclerosis
SC: subcutaneous
VAS: visual analogue scale

Data and analyses

Open in table viewer
Comparison 1. Alemtuzumab 12 mg versus interferon beta‐1a

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse‐free survival Show forest plot

2

Hazard Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 1 Relapse‐free survival.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 1 Relapse‐free survival.

1.1 24‐month analysis

2

Hazard Ratio (Fixed, 95% CI)

0.50 [0.41, 0.60]

2 Sustained disease progression‐free survival Show forest plot

3

Hazard Ratio (Fixed, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 2 Sustained disease progression‐free survival.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 2 Sustained disease progression‐free survival.

2.1 24‐month analysis

2

Hazard Ratio (Fixed, 95% CI)

0.62 [0.44, 0.87]

2.2 36‐month analysis

1

Hazard Ratio (Fixed, 95% CI)

0.25 [0.11, 0.57]

3 Number of participants with at least one adverse event Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 3 Number of participants with at least one adverse event.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 3 Number of participants with at least one adverse event.

3.1 24‐month analysis

2

1248

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [1.01, 1.06]

3.2 36‐month analysis

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.98, 1.02]

4 Change in EDSS score Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 4 Change in EDSS score.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 4 Change in EDSS score.

4.1 24‐month analysis

2

1199

Mean Difference (IV, Random, 95% CI)

‐0.20 [‐0.60, 0.20]

4.2 36‐month analysis

1

223

Mean Difference (IV, Random, 95% CI)

‐0.7 [‐1.04, ‐0.36]

5 Number of participants with new or enlarging T2‐hyperintense lesions Show forest plot

2

1238

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.59, 0.91]

Analysis 1.5

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 5 Number of participants with new or enlarging T2‐hyperintense lesions.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 5 Number of participants with new or enlarging T2‐hyperintense lesions.

6 Number of dropouts Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 6 Number of dropouts.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 6 Number of dropouts.

6.1 24‐months analysis

2

1248

Risk Ratio (M‐H, Fixed, 95% CI)

0.31 [0.23, 0.41]

6.2 36‐months analysis

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.57, 1.14]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

'Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

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

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.1 Relapse‐free survival.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.1 Relapse‐free survival.

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.2 Sustained disease progression‐free survival.
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.2 Sustained disease progression‐free survival.

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.2 Rate of participants with at least one adverse event.
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 Alemtuzumab 12 mg versus interferon beta‐1a, outcome: 1.2 Rate of participants with at least one adverse event.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 1 Relapse‐free survival.
Figuras y tablas -
Analysis 1.1

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 1 Relapse‐free survival.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 2 Sustained disease progression‐free survival.
Figuras y tablas -
Analysis 1.2

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 2 Sustained disease progression‐free survival.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 3 Number of participants with at least one adverse event.
Figuras y tablas -
Analysis 1.3

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 3 Number of participants with at least one adverse event.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 4 Change in EDSS score.
Figuras y tablas -
Analysis 1.4

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 4 Change in EDSS score.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 5 Number of participants with new or enlarging T2‐hyperintense lesions.
Figuras y tablas -
Analysis 1.5

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 5 Number of participants with new or enlarging T2‐hyperintense lesions.

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 6 Number of dropouts.
Figuras y tablas -
Analysis 1.6

Comparison 1 Alemtuzumab 12 mg versus interferon beta‐1a, Outcome 6 Number of dropouts.

Summary of findings for the main comparison. Alemtuzumab 12 mg compared to interferon beta‐1a for multiple sclerosis

Alemtuzumab 12 mg compared to interferon beta‐1a for multiple sclerosis

Patient or population: patients with multiple sclerosis
Settings: outpatients
Intervention: alemtuzumab 12 mg
Comparison: interferon beta‐1a

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Interferon beta‐1a

Alemtuzumab 12 mg

Relapse‐free survival
Follow‐up: 24 months

Not estimated

Not estimated

HR 0.50

(0.41 to 0.60)

1248
(2 studies)

⊕⊕⊕⊝
moderate5

Sustained disease progression‐free survival
Follow‐up: 24 months

Not estimated

Not estimated

HR 0.62

(0.44 to 0.87)

1191
(2 studies)

⊕⊕⊕⊝
moderate2

Number of participants with at least one adverse event

Study population

RR 1.04
(1.01 to 1.06)

1248
(2 studies)

⊕⊕⊕⊝
moderate1

94 per 100

98 per 100
(95 to 100)

Moderate

94 per 100

98 per 100
(95 to 99)

Change in EDSS score
Follow‐up: 24 months

The mean change in EDSS score in the intervention groups was
0.2 lower
(0.6 lower to 0.2 higher)

1199
(2 studies)

⊕⊝⊝⊝
very low2,3

Number of participants with new or enlarging T2‐hyperintense lesions
Follow‐up: 24 months

69 per 100

51 per 100
(41 to 63)

RR 0.74
(0.59 to 0.91)

1238
(2 studies)

⊕⊕⊕⊝
moderate4

Dropouts
Follow‐up: 24 months

Study population

RR 0.31
(0.23 to 0.41)

1248
(2 studies)

⊕⊕⊝⊝
low1,5

24 per 100

8 per 100
(6 to 10)

Moderate

24 per 100

7 per 100
(5 to 10)

*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; HR: hazard 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.

1Participants and personnel were not blinded and this outcome could be affected by this fact.
2Participants, personnel and outcome assessors were not blinded and this outcome could be affected by this fact.
3High heterogeneity; I2 = 88%.
4High heterogeneity; I2 = 80%.
5Low number of events (fewer than 300).

Figuras y tablas -
Summary of findings for the main comparison. Alemtuzumab 12 mg compared to interferon beta‐1a for multiple sclerosis
Comparison 1. Alemtuzumab 12 mg versus interferon beta‐1a

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Relapse‐free survival Show forest plot

2

Hazard Ratio (Fixed, 95% CI)

Subtotals only

1.1 24‐month analysis

2

Hazard Ratio (Fixed, 95% CI)

0.50 [0.41, 0.60]

2 Sustained disease progression‐free survival Show forest plot

3

Hazard Ratio (Fixed, 95% CI)

Subtotals only

2.1 24‐month analysis

2

Hazard Ratio (Fixed, 95% CI)

0.62 [0.44, 0.87]

2.2 36‐month analysis

1

Hazard Ratio (Fixed, 95% CI)

0.25 [0.11, 0.57]

3 Number of participants with at least one adverse event Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 24‐month analysis

2

1248

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [1.01, 1.06]

3.2 36‐month analysis

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.98, 1.02]

4 Change in EDSS score Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 24‐month analysis

2

1199

Mean Difference (IV, Random, 95% CI)

‐0.20 [‐0.60, 0.20]

4.2 36‐month analysis

1

223

Mean Difference (IV, Random, 95% CI)

‐0.7 [‐1.04, ‐0.36]

5 Number of participants with new or enlarging T2‐hyperintense lesions Show forest plot

2

1238

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.59, 0.91]

6 Number of dropouts Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 24‐months analysis

2

1248

Risk Ratio (M‐H, Fixed, 95% CI)

0.31 [0.23, 0.41]

6.2 36‐months analysis

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

0.81 [0.57, 1.14]

Figuras y tablas -
Comparison 1. Alemtuzumab 12 mg versus interferon beta‐1a