Skip to main content

EDITORIAL article

Front. Med., 21 September 2022
Sec. Rheumatology
This article is part of the Research Topic Advance In B-cell therapies For The Treatment of Rheumatic and Musculoskeletal Diseases View all 10 articles

Editorial: Advance in B-cell therapies for the treatment of rheumatic and musculoskeletal diseases

  • 1Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
  • 2National Institute for Health and Care Research (NIHR) Leeds Biomedical Research Centre, Leeds Teaching Hospitals National Health Service (NHS) Trust, Leeds, United Kingdom
  • 3Immunology Core Facility, Gemelli Science Technological Park, GSTeP, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
  • 4Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)—Università Cattolica del Sacro Cuore, Rome, Italy
  • 5Rheumatology Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden

In this Research Topic of Frontiers in Medicine, our aim was to highlight advances in B-cell therapies in various rheumatic and musculoskeletal diseases (RMD) to further refine their use in clinical trial and routine practice. The two most evaluated strategies for B-cell blockade for the treatment of rheumatic and musculoskeletal diseases (RMD) over the last three decades are B-cell depletion and inhibition of B-cell survival factors i.e., B-cell-activating factor (BAFF) and/or A proliferation-inducing ligand (APRIL). Rituximab is a chimeric anti-CD20 monoclonal antibody (mAb). The depth of B-cell depletion and clinical responses may vary, implying potential pathogenic or pharmacodynamic differences between subgroups of patients. Recent data have supported the efficacy of reduced rituximab dose and the different retreatment strategies in its licensed indications; rheumatoid arthritis (RA), granulomatosis with polyangiitis/microscopic polyangiitis and pemphigus vulgaris, although long-term data are still needed to establish the optimal approach (15). Despite failure of rituximab in meeting its primary endpoints when investigated in systemic lupus erythematosus (SLE) (6, 7) and primary Sjogren's syndrome (pSS) (8, 9), it may still be used in refractory cases based on clinical effectiveness from case series (1013). Belimumab, a BAFF-inhibitor, is licensed for patients aged ≥5 years with active, autoantibody-positive SLE who are receiving standard therapy and those aged ≥18 with active lupus nephritis who are receiving standard therapy. Its action on both B-cells and non-B-cells may have contributed to the success of belimumab trials. There remains an unmet need for mechanistic and clinical studies concerning stratification of patients who would respond best to both rituximab and belimumab to aid personalized therapy.

In RA, the optimal retreatment paradigm for rituximab has not been fully determined. Three strategies are commonly used (3). (i) Fixed retreatment e.g., 2 × 1,000 gm infusions administered every 6 months. Nevertheless, regular retreatment may risk overtreatment in some patients and increases the risk of infections associated with secondary hypogammaglobulinaemia (14). (ii) Retreatment can also be employed based on treat-to-target approach, in line with the European League Against Rheumatism (EULAR) recommendations for RA management whereby target of treatment is remission [DAS28 <2.6, Simple Disease Activity Index (SDAI) <3.3 or Clinical Disease Activity Index (CDAI) <2.8 or at least low disease activity (LDA)] (15). (iii) Retreatment-on-clinical relapse or “on demand.” Inherent to this is a degree of instability, with potential clinical implications, such as more short-term corticosteroid use that can be potentially detrimental to long-term outcomes. This could be improved by identifying clinical and biomarkers of imminent relapse. Kim et al. used data from the Korean Rheumatology Biologics registry (KOBIO) and patients who were treated at the Ajou University Hospital, Suwon South Korea. Eighty-two patients were enrolled and those who responded were treated on demand. The mean time-to-retreatment was 16 months. In multivariable analysis, factors associated with longer time-to-retreatment were concomitant use of 2 or more csDMARD and concomitant use of corticosteroid (16). The latter should be interpreted with caution since there was no consistent association with time-to-retreatment when concomitant daily oral prednisolone dose was evaluated. At 5 years, the rituximab retention rate was 72% which was a good outcome from therapeutic perspective. Since some patients appear to be refractory to B-cell depleting therapy in RA, another therapeutic option is through the Janus kinase (JAK) inhibition. Moura and Fonseca discussed in a narrative review article that currently available JAK inhibitors (tofacitinib, baricitinib, upadacitinib, peficitinib, filgotinib, and decernotinib) can affect B-cell activation, proliferation and differentiation and could be beneficial in the pre-clinical or early phase of RA (17).

In pSS, Pavlych et al. conducted a retrospective observational cohort study to compare the effectiveness of rituximab originator (MabTherara®) and rituximab biosimilar (Truxima®) in patients with a disease duration of <5 years and a systemic moderate–high activity [as defined by The EULAR Sjögren's syndrome (SS) disease activity index (ESSDAI) ≥5 points]. Nine and eight patients were treated with the originator and the biosmilar, respectively. At 48 weeks, the mean ESSDAI score was significantly reduced compared to pre-rituximab score in all patients and there was no difference in the change in ESSDAI score from baseline between both treatment arms. Disappointingly, there was no improvement observed in the change in a patient-reported outcome, the EULAR Sjogren's Syndrome Patient Reported Index (ESSPRI) in all patients at weeks 24 and 48 from baseline and between the treatment groups (18).

In SLE, Wise and Stohl wrote a narrative review article and discussed the outcome disparities in randomized controlled trials (RCTs) between rituximab and belimumab. Failure of rituximab in meeting its primary endpoint could be attributed to its poor trial design and to a degree its biological effect i.e., plasma cells do not express CD20 and thus, are insensitive to rituximab. In contrast, in addition to a better trial design including the use of a new composite primary endpoint, the SLE Responder Index (SRI-4) and adequate sample size, plasma cells express B-cell maturation antigen (BCMA) and TNF receptor superfamily member 13b (TACI) of which both are inhibited by belimumab, thus abate ongoing pathogenic autoantibody production by plasma cells (19). These factors could influence the success of belimumab in five RCTs. Post-hoc analysis of belimumab RCTs and real-world observational studies identified characteristics of patients would most likely to respond to belimumab including those with high disease activity [the SLE Disease Activity Index 2000 (SLEDAI-2K) ≥10], anti-dsDNA positivity, low complement levels, polyarthritis, non-smoking status, and lack of significant end organ damage (2022). Plüß et al. reported case series of seven patients who were treated with belimumab for non-approved SLE features (renal = 6 and neuropsychiatric = 1). Following therapy with belimumab, proteinuria was markedly improved in all patients and one patient with dysarthria and ataxia improved (23). Belimumab plus standard therapy has since approved for active lupus nephritis following a positive outcome in a phase III RCT (24). Nevertheless, it is important to note that the effect size of belimumab over its comparator in RCTs overall was rather modest (ranging from 9.7 to 14%), as well as an RCT in patients of black African ancestry failed to meet its primary endpoint at 52 weeks (25). Another subgroup of patient who may not respond well to belimumab is those who develop secondary non-depletion non response (2NDNR) to rituximab which is associated with anti-rituximab antibodies (26). Hassan et al. conducted an observational cohort study and compared the effectiveness of switching those with 2NDNR to rituximab to either belimumab (N = 8) or an alternative humanized anti-CD20 agent (N = 6; ocrelizumab = 3, ofatumumab = 2, obinutuzumab = 1). All patients in the latter group achieved SRI-4 response while only 1/8 patient in the former group met SRI-4 response. Moreover, 2/8 patients in the former developed lupus nephritis including one de novo Class II and V nephritis (27). This study suggests that patients who developed 2NDNR to rituximab should be switched within the same biologic class i.e., humanized or type 2 anti-CD20 mAbs.

In a narrative review article, Parodis et al. discussed other promising strategies to improve B-cell blockade in SLE including plasma cell inhibition using proteasome inhibitor such as bortezomib, the next generation anti-CD20 mAbs including obinutuzumab, targeting B-cell intracellular signaling through inhibition of Bruton's Tyrosine Kinase (BTK) and the use of chimeric auto-antigen receptor (CAAR) T-cells that have been genetically engineered to kill human autoreactive B-cells (28). They also discussed since BAFF level rose following treatment with rituximab, combining therapy of rituximab and belimumab would be a logical approach. This was supported by the results from BEAT-LUPUS RCT where add on add-on belimumab was superior over rituximab alone in prolonging the time-to-severe SLE flare and in reducing anti-dsDNA antibody titres (29). Another alternative is sequential therapy for which Petricca et al. reported a case report of a patient with severe and refractory lupus nephritis and bullous pemphigus who responded to treatment with rituximab, followed by belimumab (30).

Finally from B-cell biomarker perspective, You et al. compared peripheral blood mononuclear cells of 57 SLE patients and 50 healthy controls using flow cytometry. They found that double negative B-cells (DN Bcells; CD19+CD27–lgD–) were associated with lupus nephritis and positively correlated with proteinuria level. The proportion of lupus nephritis patients who achieved remission following a therapy was higher in SLE patients with low DN Bcells than in patients with high pre-treatment DN Bcells rates; 83 and 25% respectively, thus could be a promising prognostic biomarker in lupus nephritis (31).

We thank the contributing authors for shedding light on the advance in B-cell therapies in our Research Topic, with the ultimate goal of improving the care of patients with RMD. Future research agenda will build on this progress and should focus on better biomarkers that may allow prediction of active disease, prognosis and/or response to therapy through the application of new technologies and clinical efficacy of novel B-cell-targeted therapies with stratification of therapy to disease manifestation and their long-term safety particularly in terms of the risk of severe infection and major cardiovascular events.

Author contributions

MM wrote the first draft of the Editorial, which was then reviewed by SA and KC who revised it critically for important intellectual content and final approval of the version published. All authors have agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Acknowledgments

MM is a Senior Research Fellow and was funded/supported by the Wellcome Trust Institutional Strategic Support Fund Fellowship (204825/Z/16/Z). This article/paper/report also presents independent research funded/supported by the NIHR Leeds Biomedical Research Center.

Conflict of interest

MM has received consultancy fees from Aurinia Pharmaceuticals and UCB. KC received consultancy fees from Eli Lilly, AbbVie, and Pfizer.

The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Author disclaimer

The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care.

References

1. Verhoef LM, den Broeder N, Thurlings RM, van der Laan WH, van der Weele W, Kok MR, et al. Ultra-low doses of rituximab for continued treatment of rheumatoid arthritis (REDO study): a randomised controlled non-inferiority trial. Lancet Rheumatol. (2019) 1:e145–e53. doi: 10.1016/S2665-9913(19)30066-9

CrossRef Full Text | Google Scholar

2. Mariette X, Rouanet S, Sibilia J, Combe B, Le Loët X, Tebib J, et al. Evaluation of low-dose rituximab for the retreatment of patients with active rheumatoid arthritis: a non-inferiority randomised controlled trial. Ann Rheum Dis. (2014) 73:1508–14. doi: 10.1136/annrheumdis-2013-203480

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Buch MH, Smolen JS, Betteridge N, Breedveld FC, Burmester G, Dörner T, et al. Updated consensus statement on the use of rituximab in patients with rheumatoid arthritis. Ann Rheum Dis. (2011) 70:909–20. doi: 10.1136/ard.2010.144998

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Charles P, Perrodeau É, Samson M, Bonnotte B, Néel A, Agard C, et al. Long-term rituximab use to maintain remission of antineutrophil cytoplasmic antibody–associated vasculitis. Ann Intern Med. (2020) 173:179–87. doi: 10.7326/M19-3827

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Tieu J, Smith R, Basu N, Brogan P, D'Cruz D, Dhaun N, et al. Rituximab for maintenance of remission in ANCA-associated vasculitis: expert consensus guidelines. Rheumatology. (2020) 59:e24–32. doi: 10.1093/rheumatology/kez632

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Merrill JT, Neuwelt CM, Wallace DJ, Shanahan JC, Latinis KM, Oates JC, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. (2010) 62:222–33. doi: 10.1002/art.27233

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. (2012) 64:1215–26. doi: 10.1002/art.34359

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Devauchelle-Pensec V, Mariette X, Jousse-Joulin S, Berthelot JM, Perdriger A, Puéchal X, et al. Treatment of primary Sjögren syndrome with rituximab: a randomized trial. Ann Intern Med. (2014) 160:233–42. doi: 10.7326/M13-1085

PubMed Abstract | CrossRef Full Text | Google Scholar

9. 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. doi: 10.1002/art.40093

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Fanouriakis A, Kostopoulou M, Alunno A, Aringer M, Bajema I, Boletis JN, et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. BMJ. (2019) 78:736-45. doi: 10.1136/annrheumdis-2019-eular.3117

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Ramos-Casals M, Soto MJ, Cuadrado MJ, Khamashta MA. Rituximab in systemic lupus erythematosus: a systematic review of off-label use in 188 cases. Lupus. (2009) 18:767–76. doi: 10.1177/0961203309106174

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Gottenberg J-E, Cinquetti G, Larroche C, Combe B, Hachulla E, Meyer O, et al. Efficacy of rituximab in systemic manifestations of primary Sjögren's syndrome: results in 78 patients of the AutoImmune and Rituximab registry. Ann Rheum Dis. (2013) 72:1026–31.

PubMed Abstract | Google Scholar

13. Pepple S, Arnold J, Vital EM, Rawstron AC, Pease CT, Dass S, et al. Predicting sustained clinical response to rituximab in moderate to severe systemic manifestations of primary Sjögren syndrome. ACR Open Rheumatol. (2022) 4:689–99. doi: 10.1002/acr2.11466

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Md Yusof MY, Vital EM, McElvenny DM, Hensor EMA, Das S, Dass S, et al. Predicting severe infection and effects of hypogammaglobulinemia during therapy with rituximab in rheumatic and musculoskeletal diseases. Arthritis Rheumatol. (2019) 71:1812–23. doi: 10.1002/art.40937

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, Emery P, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. (2016) 75:3–15. doi: 10.1136/annrheumdis-2015-207524

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Kim JW, Jung JY, Shin K, Suh CH, Kim HA. Factors determining retreatment time interval of rituximab in Korean patients with rheumatoid arthritis. Front Med. (2021) 8:765535. doi: 10.3389/fmed.2021.765535

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Moura RA, Fonseca JE. JAK inhibitors and modulation of B cell immune responses in rheumatoid arthritis. Front Med. (2020) 7:607725. doi: 10.3389/fmed.2020.607725

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Pavlych V, Di Muzio C, Alunno A, Carubbi F. Comparison of rituximab originator with CT-P10 biosimilar in patients with primary Sjögren's syndrome: a retrospective analysis in a real-life setting. Front Med. (2020) 7:534. doi: 10.3389/fmed.2020.00534

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Wise LM, Stohl W. Belimumab and rituximab in systemic lupus erythematosus: a tale of two B cell-targeting agents. Front Med. (2020) 7:303. doi: 10.3389/fmed.2020.00303

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Parodis I, Sjöwall C, Jönsen A, Ramsköld D, Zickert A, Frodlund M, et al. Smoking and pre-existing organ damage reduce the efficacy of belimumab in systemic lupus erythematosus. Autoimmun Rev. (2017) 16:343–51. doi: 10.1016/j.autrev.2017.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

21. van Vollenhoven RF, Petri MA, Cervera R, Roth DA Ji BN, Kleoudis CS, et al. Belimumab in the treatment of systemic lupus erythematosus: high disease activity predictors of response. Ann Rheum Dis. (2012) 71:1343–9. doi: 10.1136/annrheumdis-2011-200937

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Iaccarino L, Andreoli L, Bocci EB, Bortoluzzi A, Ceccarelli F, Conti F, et al. Clinical predictors of response and discontinuation of belimumab in patients with systemic lupus erythematosus in real life setting. Results of a large, multicentric, nationwide study. J Autoimmunity. (2018) 86:1–8. doi: 10.1016/j.jaut.2017.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Plüß M, Tampe B, Niebusch N, Zeisberg M, Müller GA, Korsten P. Clinical efficacy of routinely administered belimumab on proteinuria and neuropsychiatric lupus. Front Med. (2020) 7:222. doi: 10.3389/fmed.2020.00222

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Furie R, Rovin BH, Houssiau F, Malvar A, Teng YKO, Contreras G, et al. Two-year, randomized, controlled trial of belimumab in lupus nephritis. N Engl J Med. (2020) 383:1117–28. doi: 10.1056/NEJMoa2001180

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ginzler E, Guedes Barbosa LS, D'Cruz D, Furie R, Maksimowicz-McKinnon K, Oates J, et al. Phase III/IV, randomized, fifty-two-week study of the efficacy and safety of belimumab in patients of Black African ancestry with systemic lupus erythematosus. Arthritis Rheumatol. (2022) 74:112–23. doi: 10.1002/art.41900

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Md Yusof MY, Shaw D, El-Sherbiny YM, Dunn E, Rawstron AC, Emery P, et al. Predicting and managing primary and secondary non-response to rituximab using B-cell biomarkers in systemic lupus erythematosus. Ann Rheum Dis. (2017) 76:1829–36. doi: 10.1136/annrheumdis-2017-211191

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Hassan SU, Md Yusof MY, Emery P, Dass S, Vital EM. Biologic sequencing in systemic lupus erythematosus: after secondary non-response to rituximab, switching to humanised anti-CD20 agent is more effective than belimumab. Front Med. (2020) 7:498. doi: 10.3389/fmed.2020.00498

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Parodis I, Stockfelt M, Sjöwall C. B cell therapy in systemic lupus erythematosus: from rationale to clinical practice. Front Med. (2020) 7:316. doi: 10.3389/fmed.2020.00316

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Shipa M, Embleton-Thirsk A, Parvaz M, Santos LR, Muller P, Chowdhury K, et al. Effectiveness of belimumab after rituximab in systemic lupus erythematosus: a randomized controlled trial. Ann Intern Med. (2021) 174:1647–57. doi: 10.7326/M21-2078

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Petricca L, Gigante MR, Paglionico A, Costanzi S, Vischini G, Di Mario C, et al. Rituximab followed by belimumab controls severe lupus nephritis and bullous pemphigoid in systemic lupus erythematosus refractory to several combination therapies. Front Med. (2020) 7:553075. doi: 10.3389/fmed.2020.553075

PubMed Abstract | CrossRef Full Text | Google Scholar

31. You X, Zhang R, Shao M, He J, Chen J, Liu J, et al. Double negative B cell is associated with renal impairment in systemic lupus erythematosus and acts as a marker for nephritis remission. Front Med. (2020) 7:85. doi: 10.3389/fmed.2020.00085

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: B lymphocyte, biological DMARDs, rheumatoid arthritis, Sjögren's syndrome, systemic lupus erythematosus

Citation: Md Yusof MY, Alivernini S and Chatzidionysiou K (2022) Editorial: Advance in B-cell therapies for the treatment of rheumatic and musculoskeletal diseases. Front. Med. 9:1020859. doi: 10.3389/fmed.2022.1020859

Received: 16 August 2022; Accepted: 05 September 2022;
Published: 21 September 2022.

Edited and reviewed by: João Eurico Fonseca, University of Lisbon, Portugal

Copyright © 2022 Md Yusof, Alivernini and Chatzidionysiou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Md Yuzaiful Md Yusof, y.yusof@leeds.ac.uk

These authors have contributed equally to this work and share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.