Efficacy and safety of ofatumumab in recently diagnosed, treatment-naive patients with multiple sclerosis: Results from ASCLEPIOS I and II

Jutta Gärtner, Stephen L Hauser, Amit Bar-Or, Xavier Montalban, Jeffrey A Cohen, Anne H Cross, Kumaran Deiva, Habib Ganjgahi, Dieter A Häring, Bingbing Li, Ratnakar Pingili, Krishnan Ramanathan, Wendy Su, Roman Willi, Bernd Kieseier, Ludwig Kappos, Jutta Gärtner, Stephen L Hauser, Amit Bar-Or, Xavier Montalban, Jeffrey A Cohen, Anne H Cross, Kumaran Deiva, Habib Ganjgahi, Dieter A Häring, Bingbing Li, Ratnakar Pingili, Krishnan Ramanathan, Wendy Su, Roman Willi, Bernd Kieseier, Ludwig Kappos

Abstract

Background: In the phase III ASCLEPIOS I and II trials, participants with relapsing multiple sclerosis receiving ofatumumab had significantly better clinical and magnetic resonance imaging (MRI) outcomes than those receiving teriflunomide.

Objectives: To assess the efficacy and safety of ofatumumab versus teriflunomide in recently diagnosed, treatment-naive (RDTN) participants from ASCLEPIOS.

Methods: Participants were randomized to receive ofatumumab (20 mg subcutaneously every 4 weeks) or teriflunomide (14 mg orally once daily) for up to 30 months. Endpoints analysed post hoc in the protocol-defined RDTN population included annualized relapse rate (ARR), confirmed disability worsening (CDW), progression independent of relapse activity (PIRA) and adverse events.

Results: Data were analysed from 615 RDTN participants (ofatumumab: n = 314; teriflunomide: n = 301). Compared with teriflunomide, ofatumumab reduced ARR by 50% (rate ratio (95% confidence interval (CI)): 0.50 (0.33, 0.74); p < 0.001), and delayed 6-month CDW by 46% (hazard ratio (HR; 95% CI): 0.54 (0.30, 0.98); p = 0.044) and 6-month PIRA by 56% (HR: 0.44 (0.20, 1.00); p = 0.049). Safety findings were manageable and consistent with those of the overall ASCLEPIOS population.

Conclusion: The favourable benefit-risk profile of ofatumumab versus teriflunomide supports its consideration as a first-line therapy in RDTN patients.ASCLEPIOS I and II are registered at ClinicalTrials.gov (NCT02792218 and NCT02792231).

Keywords: Relapsing multiple sclerosis; neurofilament light chain; no evidence of disease activity; progression independent of relapse activity; recently diagnosed; treatment-naive.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship and/or publication of this article: J.G., in the past 3 years, has received fees for lectures and consultancy fees from Bayer, Biogen, Merck, Novartis and Sanofi, as well as funding for a research project from Novartis. S.L.H. has received personal compensation from Alector, Annexon Biosciences, Bionure and Neurona Therapeutics; he has also received travel reimbursement from F. Hoffmann-La Roche and Novartis for CD20-related meetings and presentations. A.B.-O. has participated as a speaker in meetings sponsored by and received consulting fees and/or grant support from Atara Biotherapeutics, Biogen Idec, Celgene/Receptos, Janssen/Actelion, MedImmune, Merck/EMD Serono, Novartis, Roche/Genentech and Sanofi Genzyme. X.M. has received speaking fees and travel expenses for participation in scientific meetings, has been a steering committee member for clinical trials, or has participated in advisory boards for clinical trials in the past years with Actelion, Alexion Pharmaceuticals, Bayer, Biogen, Celgene, EMD Serono, EXCEMED, Genzyme, Immunic, MedDay, Merck, the MS International Federation, Mylan, NervGen Pharma, the National Multiple Sclerosis Society, Novartis, Roche, Sanofi Genzyme, Teva Pharmaceuticals and TG Therapeutics. J.A.C. has received personal compensation for consulting from Adamas Pharmaceuticals, Atara Biotherapeutics, Bristol Myers Squibb, Convelo Therapeutics, MedDay and Mylan, and for serving as an editor of the Multiple Sclerosis Journal. A.H.C. has consulted for Biogen, Celgene, EMD Serono, Genentech/Roche, Novartis and TG Therapeutics. K.D. has received personal compensation for speaker activities from Novartis and Sanofi. In the past 3 years, L.K.’s institution (University Hospital of Basel) has received steering committee, advisory board and consultancy fees used exclusively for research support in the department, as well as support of educational activities, from Actelion, Allergan, Almirall, Baxalta, Bayer, Biogen, Celgene/Receptos, CSL Behring, Desitin, Eisai, EXCEMED, F. Hoffmann-La Roche, Genzyme, Japan Tobacco, Merck, Minoryx Therapeutics, Novartis, Pfizer, Sanofi Aventis, Santhera Pharmaceuticals and Teva Pharmaceuticals, and license fees for Neurostatus-UHB products. Research at the MS Center in Basel has been supported by grants from Bayer, Biogen, the European Union, Inno-Suisse, Novartis, the Swiss MS Society, the Swiss National Research Foundation and Roche research foundations. D.A.H., K.R., R.W. and B.K. are employees of Novartis Pharma AG, Basel, Switzerland. B.L., R.P. and W.S. are employees of Novartis Pharmaceutical Corporation, East Hanover, NJ, USA.

Figures

Figure 1.
Figure 1.
Kaplan–Meier estimates of percentage of patients with disability worsening confirmed at: (a) 3 and (b) 6 months. Disability worsening confirmed at 3 or 6 months was defined as an increase from baseline in the Expanded Disability Status Scale (EDSS) score (on a scale from 0 to 10.0, with higher scores indicating worse disability) that was sustained for at least 3 or 6 months. For patients with a baseline EDSS score of 0, an increase in the EDSS score of at least 1.5 points was required; for patients with a baseline EDSS score of 1.0 to 5.0, the criterion was an increase of at least 1.0 points; and for patients with a baseline EDSS score of at least 5.5 points, the criterion was an increase of at least 0.5 points.
Figure 2.
Figure 2.
Kaplan–Meier analyses of time to PIRA in RDTN participants: (a) time to 3mPIRA in participants without confirmed relapses on study, (b) time to 6mPIRA in participants without confirmed relapses on study, (c) time to 3mPIRA in participants without confirmed relapses on study or before 3mCDW and (d) time to 6mPIRA in participants without confirmed relapses on study or before 6mCDW. 3mCDW: 3-month confirmed disability worsening; 3mPIRA: 3-month progression independent of relapse activity; 6mCDW: 6 month confirmed disability worsening; 6mPIRA: 6-month progression independent of relapse activity; PIRA: progression independent of relapse activity; RDTN: recently diagnosed, treatment-naive.
Figure 3.
Figure 3.
Empirical lesion incidence maps for Gd+T1 lesions in all RDTN participants: (a) at baseline (N = 615); (b) 12 months after initiation of treatment with ofatumumab (N = 314); and (c) 12 months after initiation of treatment with teriflunomide (N = 301). Gd+, gadolinium-enhancing; RDTN: recently diagnosed, treatment-naive.
Figure 4.
Figure 4.
Proportion of RDTN participants with injection-related systemic reactions following the first 10 injections in the study. RDTN: recently diagnosed, treatment-naive. Only Common Terminology Criteria for Adverse Events grades that were observed in the data are shown. Only reactions/symptoms occurring within 24 hours after injections are included (i.e. time to onset of reaction ⩽ 24 hours). As teriflunomide is taken as an oral medication, injection-related systemic reactions in participants treated with teriflunomide are in response to placebo injections.

References

    1. Filippi M, Bar-Or A, Piehl F, et al.. Multiple sclerosis. Nat Rev Dis Primers 2018; 4(1): 43.
    1. Koch-Henriksen N, Sørensen PS. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurol 2010; 9(5): 520–532.
    1. Scott TF. Understanding the impact of relapses in the overall course of MS; refinement of the 2 stage natural history model. J Neuroimmunol 2017; 305: 162–166.
    1. Kappos L, Butzkueven H, Wiendl H, et al.. Greater sensitivity to multiple sclerosis disability worsening and progression events using a roving versus a fixed reference value in a prospective cohort study. Mult Scler 2018; 24(7): 963–973.
    1. Kappos L, Wolinsky JS, Giovannoni G, et al.. Contribution of relapse-independent progression vs relapse-associated worsening to overall confirmed disability accumulation in typical relapsing multiple sclerosis in a pooled analysis of 2 randomized clinical trials. JAMA Neurol 2020; 77: 1132–1140.
    1. University of California San Francisco MS-EPIC Team, Cree BAC, Hollenbach JA, et al.. Silent progression in disease activity-free relapsing multiple sclerosis. Ann Neurol 2019; 85(5): 653–666.
    1. Pfeifenbring S, Bunyan RF, Metz I, et al.. Extensive acute axonal damage in pediatric multiple sclerosis lesions. Ann Neurol 2015; 77(4): 655–667.
    1. Barro C, Benkert P, Disanto G, et al.. Serum neurofilament as a predictor of disease worsening and brain and spinal cord atrophy in multiple sclerosis. Brain 2018; 141(8): 2382–2391.
    1. De Stefano N, Giorgio A, Battaglini M, et al.. Assessing brain atrophy rates in a large population of untreated multiple sclerosis subtypes. Neurology 2010; 74(23): 1868–1876.
    1. Dahlke F, Arnold DL, Aarden P, et al.. Characterisation of MS phenotypes across the age span using a novel data set integrating 34 clinical trials ( cohort): Age is a key contributor to presentation. Mult Scler 2021; 27(13): 2062–2076.
    1. Andravizou A, Dardiotis E, Artemiadis A, et al.. Brain atrophy in multiple sclerosis: Mechanisms, clinical relevance and treatment options. Auto Immun Highlights 2019; 10(1): 7.
    1. Banwell B, Arnold DL, Tillema JM, et al.. MRI in the evaluation of pediatric multiple sclerosis. Neurology 2016; 87(9 Suppl 2): S88–S96.
    1. Traboulsee AL, Cornelisse P, Sandberg-Wollheim M, et al.. Prognostic factors for long-term outcomes in relapsing-remitting multiple sclerosis. Mult Scler J Exp Transl Clin 2016; 2: 2055217316666406.
    1. Fisher KS, Cuascut FX, Rivera VM, et al.. Current advances in pediatric onset multiple sclerosis. Biomedicines 2020; 8(4): 71.
    1. Gärtner J, Chitnis T, Ghezzi A, et al.. Relapse rate and MRI activity in young adult patients with multiple sclerosis: A post hoc analysis of phase 3 fingolimod trials. Mult Scler J Exp Transl Clin 2018; 4(2): 2055217318778610.
    1. Ghezzi A, Baroncini D, Zaffaroni M, et al.. Pediatric versus adult MS: Similar or different? Mult Scler Demyelinating Disord 2017; 2(1): 5.
    1. Merkel B, Butzkueven H, Traboulsee AL, et al.. Timing of high-efficacy therapy in relapsing-remitting multiple sclerosis: A systematic review. Autoimmun Rev 2017; 16(6): 658–665.
    1. Rotstein DL, Healy BC, Malik MT, et al.. Evaluation of no evidence of disease activity in a 7-year longitudinal multiple sclerosis cohort. JAMA Neurol 2015; 72(2): 152–158.
    1. Engelberts PJ, Voorhorst M, Schuurman J, et al.. Type I CD20 antibodies recruit the B cell receptor for complement-dependent lysis of malignant B cells. J Immunol 2016; 197(12): 4829–4837.
    1. Kesimpta® prescribing information, (2020, accessed 15 December 2020).
    1. Kesimpta® summary of product characteristics, (2021, accessed 23 April 2021).
    1. Hauser SL, Bar-Or A, Cohen JA, et al.. Ofatumumab versus teriflunomide in multiple sclerosis. N Engl J Med 2020; 383(6): 546–557.
    1. Samjoo IA, Worthington E, Drudge C, et al.. Comparison of ofatumumab and other disease-modifying therapies for relapsing multiple sclerosis: A network meta-analysis. J Comp Eff Res 2020; 9: 1255–1274.
    1. Havrdová E, Arnold DL, Bar-Or A, et al.. No evidence of disease activity (NEDA) analysis by epochs in patients with relapsing multiple sclerosis treated with ocrelizumab vs interferon beta-1a. Mult Scler J Exp Transl Clin 2018; 4(1): 2055217318760642.
    1. US Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE), (2017, accessed 13 November 2020).
    1. De Stefano N, Silva DG, Barnett MH. Effect of fingolimod on brain volume loss in patients with multiple sclerosis. CNS Drugs 2017; 31(4): 289–305.
    1. Radue EW, Barkhof F, Kappos L, et al.. Correlation between brain volume loss and clinical and MRI outcomes in multiple sclerosis. Neurology 2015; 84(8): 784–793.
    1. Krieger SC, Cook K, De Nino S, et al.. The topographical model of multiple sclerosis: A dynamic visualization of disease course. Neurol Neuroimmunol Neuroinflamm 2016; 3(5): e279.
    1. University of California San Francisco MS-EPIC Team, Cree BA, Gourraud PA, et al.. Long-term evolution of multiple sclerosis disability in the treatment era. Ann Neurol 2016; 80(4): 499–510.
    1. Fisher E, Rudick RA, Simon JH, et al.. Eight-year follow-up study of brain atrophy in patients with MS. Neurology 2002; 59(9): 1412–1420.
    1. Bar-Or A, Grove RA, Austin DJ, et al.. Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: The MIRROR study. Neurology 2018; 90(20): e1805–e1814.
    1. La Mantia L, Di Pietrantonj C, Rovaris M, et al.. Interferons-beta versus glatiramer acetate for relapsing-remitting multiple sclerosis. Cochrane Database Syst Rev 2016; 11: CD009333.
    1. Cross AH, Naismith RT. Established and novel disease-modifying treatments in multiple sclerosis. J Intern Med 2014; 275(4): 350–363.
    1. Lebrun-Frenay C, Moulignier A, Pierrot-Deseilligny C, et al.. Five-year outcome in the copaxone observatory: A nationwide cohort of patients with multiple sclerosis starting treatment with glatiramer acetate in France. J Neurol 2019; 266(4): 888–901.

Source: PubMed

3
Sottoscrivi