Efficacy of Epratuzumab, an Anti-CD22 Monoclonal IgG Antibody, in Systemic Lupus Erythematosus Patients With Associated Sjögren's Syndrome: Post Hoc Analyses From the EMBODY Trials

Jacques-Eric Gottenberg, Thomas Dörner, Hendrika Bootsma, Valérie Devauchelle-Pensec, Simon J Bowman, Xavier Mariette, Holger Bartz, Marga Oortgiesen, Anthony Shock, Willem Koetse, Catrinel Galateanu, Sabine Bongardt, William A Wegener, David M Goldenberg, Guy Meno-Tetang, Gordana Kosutic, Caroline Gordon, Jacques-Eric Gottenberg, Thomas Dörner, Hendrika Bootsma, Valérie Devauchelle-Pensec, Simon J Bowman, Xavier Mariette, Holger Bartz, Marga Oortgiesen, Anthony Shock, Willem Koetse, Catrinel Galateanu, Sabine Bongardt, William A Wegener, David M Goldenberg, Guy Meno-Tetang, Gordana Kosutic, Caroline Gordon

Abstract

Objective: EMBODY 1 (ClinicalTrials.gov identifier: NCT01262365) and EMBODY 2 (ClinicalTrials.gov identifier: NCT01261793) investigated the efficacy and safety of epratuzumab, a CD22-targeted humanized monoclonal IgG antibody, in patients with systemic lupus erythematosus (SLE). The studies showed no significant difference from placebo in primary or secondary clinical outcome measures but did demonstrate B cell-specific immunologic activity. The aim of this post hoc analysis was to determine whether epratuzumab had a different clinical efficacy profile in SLE patients with versus those without an associated diagnosis of Sjögren's syndrome (SS).

Methods: The efficacy and safety of epratuzumab were compared between 2 patient subpopulations randomized in EMBODY 1 and 2: SLE patients with and those without a diagnosis of associated SS. British Isles Lupus Assessment Group (BILAG) total score, BILAG-based Combined Lupus Assessment (BICLA) clinical response to treatment, biologic markers (including B cells, IgG, IgM, and IgA), and safety were assessed.

Results: A total of 1,584 patients were randomized in the EMBODY 1 and EMBODY 2 trials; 113 patients were anti-SSA positive and had a diagnosis of associated SS, and 1,375 patients (86.8%) had no diagnosis of associated SS (918 patients were randomized to receive epratuzumab and 457 to receive placebo). For patients with associated SS, but not those without associated SS, a higher proportion of patients receiving epratuzumab achieved a BICLA response and a reduction from baseline in BILAG total score. B cell reduction was faster in patients with associated SS. The sensitivity of B cells to epratuzumab as measured by the mean concentration producing 50% of the maximum B cell count depletion was lower for patients with associated SS (9.5 μg/ml) versus the total EMBODY population (87.1 μg/ml). No difference in the frequency of adverse events in those receiving placebo was reported.

Conclusion: Patients with SLE and associated SS treated with epratuzumab showed improvement in SLE disease activity, which was associated with bioactivity, such as decreases in B cell number and IgM level.

© 2018 The Authors. Arthritis & Rheumatology published by Wiley Periodicals, Inc. on behalf of American College of Rheumatology.

Figures

Figure 1
Figure 1
A–D, British Isles Lupus Assessment Group (BILAG)–based Combined Lupus Assessment (BICLA) response rate (A), change in total BILAG score from baseline (B), change in physician's global assessment (PGA) from baseline (C), and change in Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) from baseline (D) in SLE patients without associated Sjögren's syndrome (SS; non‐aSjS patients) receiving placebo or epratuzumab (Emab). E–H, BICLA response rate (E), change in total BILAG score from baseline (F), change in PGA from baseline (G), and change in SLEDAI from baseline (H) in SLE patients with associated SS receiving placebo or epratuzumab. Analyses were conducted using the full analysis set. Values are the median percent change from baseline. For total BILAG score, PGA, and SLEDAI, statistical testing was conducted only at weeks 24 and 48. Nonresponder imputation was used for the BICLA response. The last observation carried forward method was used to impute missing values for the total BILAG score, PGA, and SLEDAI. Light asterisk = P < 0.05 for epratuzumab 1,200 mg every other week (QOW) versus placebo; dark asterisk = P < 0.05 for epratuzumab 600 mg every week (QW) versus placebo.
Figure 2
Figure 2
A and B, Percentage change from baseline in CD19+ cells from patients with systemic lupus erythematosus (SLE) without associated Sjögren's syndrome (SS; non‐aSjS patients) (A) and patients with SLE with associated SS (B) receiving placebo or epratuzumab (Emab). C, Pharmacokinetics/pharmacodynamics modeling of B cell reduction following intravenous epratuzumab. Dotted lines represent the 5th, 25th, 75th, and 95th percentiles, respectively, of the observed epratuzumab average steady‐state plasma drug concentration during multiple‐dose administration; broken line represents the 50th percentile. The shaded area shows the 95% confidence interval based on 500 bootstrap sampling with replacement available for all the “All data” model only. QOW = every other week; QW = every week. Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.40425/abstract.
Figure 3
Figure 3
A and B, Median percent change from baseline in IgM (A) and IgG (B) serum concentrations in patients with systemic lupus erythematosus (SLE) without associated Sjögren's syndrome (SS; non‐aSjS) receiving placebo or epratuzumab (Emab). C and D, Median percent change from baseline in IgM (C) and IgG (D) serum concentrations in patients with SLE with associated SS receiving placebo or epratuzumab. Analyses were conducted using the full analysis set and observed data. Values are the median (25th–75th percentile) percent change from baseline. QOW = every other week; QW = every week. Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.40425/abstract.
Figure 4
Figure 4
A and B, Mean change from baseline in anti‐SSA (A) and anti‐SSB (B) in patients with systemic lupus erythematosus (SLE) without associated Sjögren's syndrome (SS; non‐aSjS) receiving placebo or epratuzumab (Emab). C and D, Mean change from baseline in anti‐SSA (C) and anti‐SSB (D) in patients with SLE with associated SS receiving placebo or epratuzumab. Analyses were conducted using the full analysis set and observed data. Values are the mean (95% confidence interval) change from baseline. QOW = every other week; QW = every week. Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.40425/abstract.

References

    1. Fox RI. Sjogren's syndrome. Lancet 2005;366:321–31.
    1. Youinou P, Devauchelle‐Pensec V, Pers JO. Significance of B cells and B cell clonality in Sjögren's syndrome. Arthritis Rheum 2010;62:2605–10.
    1. Salomonsson S, Jonsson MV, Skarstein K, Brokstad KA, Hjelmström P, Wahren‐Herlenius M, et al. Cellular basis of ectopic germinal center formation and autoantibody production in the target organ of patients with Sjögren's syndrome. Arthritis Rheum 2003;48:3187–201.
    1. Risselada AP, Looije MF, Kruize AA, Bijlsma JW, van Roon JA. The role of ectopic germinal centers in the immunopathology of primary Sjögren's syndrome: a systematic review. Semin Arthritis Rheum 2013;42:368–76.
    1. Ambrus JL, Suresh L, Peck A. Multiple roles for B‐lymphocytes in Sjögren's syndrome. J Clin Med 2016;5:E87.
    1. Manoussakis MN, Georgopoulou C, Zintzaras E, Spyropoulou M, Stavropoulou A, Skopouli FN, et al. Sjögren's syndrome associated with systemic lupus erythematosus: clinical and laboratory profiles and comparison with primary Sjögren's syndrome. Arthritis Rheum 2004;50:882–91.
    1. Fox PC. Autoimmune diseases and Sjögren's syndrome. Ann N Y Acad Sci 2007;1098:15–21.
    1. Patel R, Shahane A. The epidemiology of Sjögren's syndrome. Clin Epidemiol 2014;6:247.
    1. Choi J, Kim ST, Craft J. The pathogenesis of systemic lupus erythematosus: an update. Curr Opin Immunol 2012;24:651–7.
    1. Mariette X, Gottenberg JE. Pathogenesis of Sjögren's syndrome and therapeutic consequences. Curr Opin Rheumatol 2010;22:471–7.
    1. Looney RJ, Anolik JH, Campbell D, Felgar RE, Young F, Arend LJ, et al. B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose‐escalation trial of rituximab. Arthritis Rheum 2004;50:2580–9.
    1. Shirota Y, Yarboro C, Fischer R, Pham TH, Lipsky P, Illei GG. Impact of anti‐interleukin‐6 receptor blockade on circulating T and B cell subsets in patients with systemic lupus erythematosus. Ann Rheum Dis 2013;72:118–28.
    1. Stohl W, Hiepe F, Latinis KM, Thomas M, Scheinberg MA, Clarke A, et al. Belimumab reduces autoantibodies, normalizes low complement levels, and reduces select B cell populations in patients with systemic lupus erythematosus. Arthritis Rheum 2012;64:2328–37.
    1. Lumb S, Fleischer SJ, Wiedemann A, Daridon C, Maloney A, Shock A, et al. Engagement of CD22 on B cells with the monoclonal antibody epratuzumab stimulates the phosphorylation of upstream inhibitory signals of the B cell receptor. J Cell Commun Signal 2016;10:143–51.
    1. Carnahan J, Wang P, Kendall R, Chen C, Hu S, Boone T, et al. Epratuzumab, a humanized monoclonal antibody targeting CD22: characterization of in vitro properties. Clin Cancer Res 2003;9:3982s–90s.
    1. Rossi D, Spina V, Bomben R, Rasi S, Dal‐Bo M, Bruscaggin A, et al. Association between molecular lesions and specific B‐cell receptor subsets in chronic lymphocytic leukemia. Blood 2013;121:4902–5.
    1. Fleischer V, Sieber J, Fleischer SJ, Shock A, Heine G, Daridon C, et al. Epratuzumab inhibits the production of the proinflammatory cytokines IL‐6 and TNF‐α, but not the regulatory cytokine IL‐10, by B cells from healthy donors and SLE patients. Arthritis Res Ther 2015;17:185.
    1. Jacobi AM, Goldenberg DM, Hiepe F, Radbruch A, Burmester GR, Dorner T. Differential effects of epratuzumab on peripheral blood B cells of patients with systemic lupus erythematosus versus normal controls. Ann Rheum Dis 2008;67:450–7.
    1. Wallace DJ, Kalunian K, Petri MA, Strand V, Houssiau FA, Pike M, et al. Efficacy and safety of epratuzumab in patients with moderate/severe active systemic lupus erythematosus: results from EMBLEM, a phase IIb, randomised, double‐blind, placebo‐controlled, multicentre study. Ann Rheum Dis 2014;73:183–90.
    1. Wallace DJ, Hobbs K, Clowse ME, Petri M, Strand V, Pike M, et al. Long‐term safety and efficacy of epratuzumab in the treatment of moderate‐to‐severe systemic lupus erythematosus: results from an open‐label extension study. Arthritis Care Res (Hoboken) 2016;68:534–43.
    1. Clowse ME, Wallace DJ, Furie RA, Petri MA, Pike MC, Leszczyński P, et al. Efficacy and safety of epratuzumab in moderately to severely active systemic lupus erythematosus: results from two phase III, randomized, double‐blind, placebo‐controlled trials. Arthritis Rheumatol 2017;69:362–75.
    1. Wallace DJ, Strand V, Furie R, Petri M, Kalunian K, Pike M, et al. Evaluation of treatment success in systemic lupus erythematosus clinical trials: development of the British Isles Lupus Assessment Group‐Based Composite Lupus Assessment endpoint [abstract]. Arthritis Rheum 2011;63 Suppl:S885.
    1. Steinfeld SD, Tant L, Burmester GR, Teoh NK, Wegener WA, Goldenberg DM, et al. Epratuzumab (humanised anti‐CD22 antibody) in primary Sjogren's syndrome: an open‐label phase I/II study. Arthritis Res Ther 2006;8:R129.
    1. Hochberg MC, for the Diagnostic and Therapeutic Criteria Committee of the American College of Rheumatology . Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus [letter]. Arthritis Rheum 1997;40:1725.
    1. Yee CS, Farewell V, Isenberg DA, Griffiths B, Teh LS, Bruce IN, et al. The BILAG‐2004 index is sensitive to change for assessment of SLE disease activity. Rheumatology (Oxford) 2009;48:691–5.
    1. Isenberg DA, Rahman A, Allen E, Farewell V, Akil M, Bruce IN, et al. BILAG 2004: development and initial validation of an updated version of the British Isles Lupus Assessment Group's disease activity index for patients with systemic lupus erythematosus. Rheumatology (Oxford) 2005;44:902–6.
    1. Gladman DD, Ibanez D, Urowitz MB. Systemic Lupus Erythematosus Disease Activity Index 2000. J Rheumatol 2002;29:288–91.
    1. Yee CS, Cresswell L, Farewell V, Rahman A, Teh LS, Griffiths B, et al. Numerical scoring for the BILAG‐2004 index. Rheumatology (Oxford) 2010;49:1665–9.
    1. Hansen A, Lipsky PE, Dorner T. B cells in Sjogren's syndrome: indications for disturbed selection and differentiation in ectopic lymphoid tissue. Arthritis Res Ther 2007;9:218.
    1. Ambrosi A, Wahren‐Herlenius M. Update on the immunobiology of Sjogren's syndrome. Curr Opin Rheumatol 2015;27:468–75.
    1. Dass S, Bowman SJ, Vital EM, Ikeda K, Pease CT, Hamburger J, et al. Reduction of fatigue in Sjogren syndrome with rituximab: results of a randomised, double‐blind, placebo‐controlled pilot study. Ann Rheum Dis 2008;67:1541–4.
    1. Pijpe J, van Imhoff GW, Spijkervet FK, Roodenburg JL, Wolbink GJ, Mansour K, et al. Rituximab treatment in patients with primary Sjögren's syndrome: an open‐label phase II study. Arthritis Rheum 2005;52:2740–50.
    1. Devauchelle‐Pensec V, Mariette X, Jousse‐Joulin S, Berthelot JM, Perdriger A, Puechal X, et al. Treatment of primary Sjogren syndrome with rituximab: a randomized trial. Ann Intern Med 2014;160:233–42.
    1. Bowman SJ, Everett CC, O'Dwyer JL, Emery P, Pitzalis C, Ng WF, et al. Randomized controlled trial of rituximab and cost‐effectiveness analysis in treating fatigue and oral dryness in primary Sjögren's syndrome. Arthritis Rheumatol 2017;69:1440–50.
    1. Mariette X, Seror R, Quartuccio L, Baron G, Salvin S, Fabris M, et al. Efficacy and safety of belimumab in primary Sjogren's syndrome: results of the BELISS open‐label phase II study. Ann Rheum Dis 2015;74:526–31.
    1. Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE, et al, and the European Study Group on Classification Criteria for Sjögren's Syndrome . Classification criteria for Sjögren's syndrome: a revised version of the European criteria proposed by the American‐European Consensus Group. Ann Rheum Dis 2002;61:554–8.

Source: PubMed

3
S'abonner