Five years of ocrelizumab in relapsing multiple sclerosis: OPERA studies open-label extension

Stephen L Hauser, Ludwig Kappos, Douglas L Arnold, Amit Bar-Or, Bruno Brochet, Robert T Naismith, Anthony Traboulsee, Jerry S Wolinsky, Shibeshih Belachew, Harold Koendgen, Victoria Levesque, Marianna Manfrini, Fabian Model, Stanislas Hubeaux, Lahar Mehta, Xavier Montalban, Stephen L Hauser, Ludwig Kappos, Douglas L Arnold, Amit Bar-Or, Bruno Brochet, Robert T Naismith, Anthony Traboulsee, Jerry S Wolinsky, Shibeshih Belachew, Harold Koendgen, Victoria Levesque, Marianna Manfrini, Fabian Model, Stanislas Hubeaux, Lahar Mehta, Xavier Montalban

Abstract

Objective: To assess over 3 years of follow-up the effects of maintaining or switching to ocrelizumab (OCR) therapy on clinical and MRI outcomes and safety measures in the open-label extension (OLE) phase of the pooled OPERA: I/II studies in relapsing multiple sclerosis.

Methods: After 2 years of double-blind, controlled treatment, patients continued OCR (600 mg infusions every 24 weeks) or switched from interferon (IFN)-β-1a (44 μg 3 times weekly) to OCR when entering the OLE phase (3 years). Adjusted annualized relapse rate, time to onset of 24-week confirmed disability progression (CDP)/improvement (CDP), brain MRI activity (gadolinium-enhanced and new/enlarging T2 lesions), and percentage brain volume change were analyzed.

Results: Of patients entering the OLE phase, 88.6% completed year 5. The cumulative proportion with 24-week CDP was lower in patients who initiated OCR earlier vs patients initially receiving IFN-β-1a (16.1% vs 21.3% at year 5; p = 0.014). Patients continuing OCR maintained and those switching from IFN-β-1a to OCR attained near complete and sustained suppression of new brain MRI lesion activity from years 3-5. Over the OLE phase, patients continuing OCR exhibited less whole brain volume loss from double-blind study baseline vs those switching from IFN-β-1a (-1.87% vs -2.15% at year 5; p < 0.01). Adverse events were consistent with past reports and no new safety signals emerged with prolonged treatment.

Conclusion: Compared with patients switching from IFN-β-1a, earlier and continuous OCR treatment up to 5 years provided sustained benefit on clinical and MRI measures of disease progression.

Classification of evidence: This study provides Class III evidence that earlier and continuous treatment with OCR provided sustained benefit on clinical and MRI outcomes of disease activity and progression compared with patients switching from IFN-β-1a. The study is rated Class III because of the initial treatment randomization disclosure that occurred after inclusion in OLE.

Clinical trial identifiers: NCT01247324/NCT01412333.

Copyright © 2020 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Figures

Figure 1. Patient disposition in the pooled…
Figure 1. Patient disposition in the pooled OPERA I and OPERA II intent-to-treat (ITT) population
Percentages in parentheses are of the ITT population. aClinical cutoff date: February 5, 2018; patients entering the open-label extension (OLE) phase who completed the double-blind period (DBP): interferon (IFN)-β-1a 623/660 (94.4%) and ocrelizumab (OCR) 702/726 (96.7%). b88.4%, and c88.7%, of patients who entered the OLE completed year 3. SFU = safety follow-up.
Figure 2. Annualized relapse rate (ARR) in…
Figure 2. Annualized relapse rate (ARR) in double-blind controlled treatment phase (DBP) study years 1 and 2 and open-label extension (OLE) years 1–3
Estimates are from analysis based on generalized estimating equation Poisson regression model with repeated measurements using unstructured covariance matrix, adjusted by randomized treatment, study, baseline Expanded Disability Status Scale (<4.0 vs ≥4.0), geographic region (United States vs rest of the world), year, and treatment-by-year interaction. Log-transformed exposure time is included as an offset variable. aThe total number of relapses for all patients in the treatment group divided by the total patient-years of exposure to that treatment. bDBP year 1 and DBP year 2 data include the intention-to-treat population (number of patients available); year 3 (OLE year 1), year 4 (OLE year 2), and year 5 (OLE year 3) data include the OLE ITT population (number of patients available). Clinical cutoff date: February 5, 2018. IFN = interferon; OCR = ocrelizumab.
Figure 3. Time to onset of confirmed…
Figure 3. Time to onset of confirmed disability progression (CDP) and confirmed disability improvement (CDI) for at least 24 weeks during double-blind controlled treatment phase (DBP) and open-label extension (OLE) periods
Time to onset of (A) CDP and (B) CDI. Curves show Kaplan-Meier estimates of the proportion of patients with disability progression/improvement events relative to the original double-blind treatment period baseline throughout double-blind and OLE periods. Without imputation. Intention-to-treat (ITT) population. Pooled OPERA I and OPERA II population; DBP clinical cutoff dates: April 2, 2015, and May 12, 2015, respectively; OLE clinical cutoff date: February 5, 2018. Data shown up to week 240, the last visit all ongoing patients completed. HR = hazard ratio; IFN = interferon; OCR = ocrelizumab.
Figure 4. Total number of T1 gadolinium…
Figure 4. Total number of T1 gadolinium (Gd)-enhancing lesions and new or enlarging T2 lesions in the double-blind controlled treatment phase (DBP) and open-label extension (OLE)
(A) T1 Gd-enhancing lesions and (B) new or enlarging T2 lesions in the DBP and OLE. aDBP week 24, DBP year 1, and DBP year 2 data include the intention-to-treat (ITT) population; year 3 (OLE year 1), year 4 (OLE year 2), and year 5 (OLE year 3) data include the OLE ITT population; clinical cutoff date: February 5, 2018. bUnadjusted rate. IFN = interferon; OCR = ocrelizumab.
Figure 5. Percentage change from baseline and…
Figure 5. Percentage change from baseline and annualized, in whole brain volume (WBV), cortical gray matter volume (CGMV), and white matter volume (WMV) in the double-blind controlled treatment phase (DBP) and open-label extension (OLE)
Percentage change (A.a, B.a, C.a) from baseline and (A.b, B.b, C.b) annualized, in (A) WBV, (B) CGMV, and (C) WMV in the DBP and OLE mixed-effect model of repeated measures (MMRM) plot, intention-to-treat (ITT) population. Pooled OPERA [Rebif] I and OPERA II (clinical cutoff date: February 5, 2018); p values shown for difference in adjusted means. Graph includes patients with assessment at baseline and at least 1 postbaseline value. Estimates are from analysis based on MMRM using unstructured covariance matrix: percentage change = baseline brain volume + geographic region (United States vs rest of the world) + baseline Expanded Disability Status Scale (<4.0 vs ≥4.0) + study + week + treatment + treatment × week (repeated values over week) + baseline brain volume × week. aThe bars represent the annualized change in WBV, CGMV, or WMV occurring during the time period delineated by the axis label and the preceding 24 or 48 weeks. BL = baseline; CI = confidence interval; IFN = interferon; OCR = ocrelizumab.

References

    1. DiLillo DJ, Hamaguchi Y, Ueda Y, et al. . Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice. J Immunol 2008;180:361–371.
    1. Klein C, Lammens A, Schäfer W, et al. . Epitope interactions of monoclonal antibodies targeting CD20 and their relationship to functional properties. MAbs 2013;5:22–33.
    1. Hauser SL, Bar-Or A, Comi G, et al. . Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med 2017;376:221–234.
    1. Polman CH, Reingold SC, Banwell B, et al. . Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 2011;69:292–302.
    1. Smith SM, Zhang Y, Jenkinson M, et al. . Accurate, robust, and automated longitudinal and cross-sectional brain change analysis. Neuroimage 2002;17:479–489.
    1. Nakamura K, Guizard N, Fonov VS, Narayanan S, Collins DL, Arnold DL. Jacobian integration method increases the statistical power to measure gray matter atrophy in multiple sclerosis. Neuroimage Clin 2014;4:10–17.
    1. Goodin DS, Reder AT, Bermel RA, et al. . Relapses in multiple sclerosis: relationship to disability. Mult Scler Relat Disord 2016;6:10–20.
    1. Harding K, Williams O, Willis M, et al. . Clinical outcomes of escalation vs early intensive disease-modifying therapy in patients with multiple sclerosis. JAMA Neurol 2019;76:536–541.
    1. Brown JWL, Coles A, Horakova D, et al. . Association of initial disease-modifying therapy with later conversion to secondary progressive multiple sclerosis. JAMA 2019;321:175–187.
    1. Naegelin Y, Naegelin P, von Felten S, et al. . Association of rituximab treatment with disability progression among patients with secondary progressive multiple sclerosis. JAMA Neurol 2019;76:274–281.
    1. He A, Merkel B, Zhovits L, et al. . Early start of high-efficacy therapies improves disability outcomes over 10 years. Presented at the 34th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS); October 10–12, 2018; Berlin. Poster P919.
    1. Battaglini M, Giorgio A, Luchetti L, et al. . Dynamics of pseudo-atrophy in relapsing-remitting MS patients treated with interferon beta-1a as assessed by monthly brain MRI. Presented at the 34th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS); October 10–12, 2018; Berlin. Poster P798.
    1. De Stefano N, Stromillo ML, Giorgio A, et al. . Establishing pathological cut-offs of brain atrophy rates in multiple sclerosis. J Neurol Neurosurg Psychiatry 2016;87:93–99.
    1. Hauser SL, Kappos L, Montalban X, et al. . Safety of ocrelizumab in multiple sclerosis: updated analysis in patients with relapsing and primary progressive multiple sclerosis. Presented at the 34th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS); October 10–12, 2018; Berlin, Germany. Poster P1229.
    1. Nørgaard M, Veres K, Didden EM, Wormser D, Magyari M. Multiple sclerosis and cancer incidence: a Danish nationwide cohort study. Mult Scler Relat Disord 2019;28:81–85.
    1. Hauser SL, Kappos L, Montalban X, et al. . Safety of ocrelizumab in multiple sclerosis: updated analysis in patients with relapsing and primary progressive multiple sclerosis. Presented at the 14th World Congress of Neurology Meeting; October 27–31, 2019; Dubai, UAE.
    1. Martínez-Yélamos S, Martínez-Yélamos A, Martín Ozaeta G, Casado V, Carmona O, Arbizu T. Regression to the mean in multiple sclerosis. Mult Scler 2006;12:826–829.
    1. Ziemssen T, De Stefano N, Sormani MP, Van Wijmeersch B, Wiendl H, Kieseier BC. Optimizing therapy early in multiple sclerosis: an evidence-based view. Mult Scler Relat Disord 2015;4:460–469.

Source: PubMed

3
Subscribe