Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2023

Open Access 01-12-2023 | Ocrelizumab | Research

Extended interval dosing of ocrelizumab modifies the repopulation of B cells without altering the clinical efficacy in multiple sclerosis

Authors: Carla Rodriguez-Mogeda, Zoë Y. G. J. van Lierop, Susanne M. A. van der Pol, Loet Coenen, Laura Hogenboom, Alwin Kamermans, Ernesto Rodriguez, Jack van Horssen, Zoé L. E. van Kempen, Bernard M. J. Uitdehaag, Charlotte E. Teunissen, Maarten E. Witte, Joep Killestein, Helga E. de Vries

Published in: Journal of Neuroinflammation | Issue 1/2023

Login to get access

Abstract

Background

Recent studies suggest that extended interval dosing of ocrelizumab, an anti-B cell therapy, does not affect its clinical effectiveness in most patients with multiple sclerosis (MS). However, it remains to be established whether certain B cell subsets are differentially repopulated after different dosing intervals and whether these subsets relate to clinical efficacy.

Methods

We performed high-dimensional single-cell characterization of the peripheral immune landscape of patients with MS after standard (SID; n = 43) or extended interval dosing (EID; n = 37) of ocrelizumab and in non-ocrelizumab-treated (control group, CG; n = 28) patients with MS, using mass cytometry by time of flight (CyTOF).

Results

The first B cells that repopulate after both ocrelizumab dosing schemes were immature, transitional and regulatory CD1d+ CD5+ B cells. In addition, we observed a higher percentage of transitional, naïve and regulatory B cells after EID in comparison with SID, but not of memory B cells or plasmablasts. The majority of repopulated B cell subsets showed an increased migratory phenotype, characterized by higher expression of CD49d, CD11a, CD54 and CD162. Interestingly, after EID, repopulated B cells expressed increased CD20 levels compared to B cells in CG and after SID, which was associated with a delayed repopulation of B cells after a subsequent ocrelizumab infusion. Finally, the number of/changes in B cell subsets after both dosing schemes did not correlate with any relapses nor progression of the disease.

Conclusions

Taken together, our data highlight that extending the dosing interval of ocrelizumab does not lead to increased repopulation of effector B cells. We show that the increase of CD20 expression on B cell subsets in EID might lead to longer depletion or less repopulation of B cells after the next infusion of ocrelizumab. Lastly, even though extending the ocrelizumab interval dosing alters B cell repopulation, it does not affect the clinical efficacy of ocrelizumab in our cohort of patients with MS.
Appendix
Available only for authorised users
Literature
1.
go back to reference Compston A, Coles A. Multiple sclerosis. The Lancet. 2008;372(9648):1502–17. Compston A, Coles A. Multiple sclerosis. The Lancet. 2008;372(9648):1502–17.
2.
go back to reference Portaccio E, et al. Progression is independent of relapse activity in early multiple sclerosis: a real-life cohort study. Brain. 2022;145(8):2796–805.PubMed Portaccio E, et al. Progression is independent of relapse activity in early multiple sclerosis: a real-life cohort study. Brain. 2022;145(8):2796–805.PubMed
3.
go back to reference Kappos L, 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(9):1132–40.PubMed Kappos L, 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(9):1132–40.PubMed
4.
go back to reference Cencioni MT, et al. B cells in multiple sclerosis—from targeted depletion to immune reconstitution therapies. Nat Rev Neurol. 2021;17(7):399–414.PubMed Cencioni MT, et al. B cells in multiple sclerosis—from targeted depletion to immune reconstitution therapies. Nat Rev Neurol. 2021;17(7):399–414.PubMed
5.
go back to reference Klein C, et al. Epitope interactions of monoclonal antibodies targeting CD20 and their relationship to functional properties. MAbs. 2013;5(1):22–33.PubMedPubMedCentral Klein C, et al. Epitope interactions of monoclonal antibodies targeting CD20 and their relationship to functional properties. MAbs. 2013;5(1):22–33.PubMedPubMedCentral
6.
go back to reference Hauser SL, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med. 2016;376(3):221–34.PubMed Hauser SL, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med. 2016;376(3):221–34.PubMed
7.
go back to reference Montalban X, et al. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. 2017;376(3):209–20.PubMed Montalban X, et al. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. 2017;376(3):209–20.PubMed
8.
go back to reference Baker D, et al. Memory B cells are major targets for effective immunotherapy in relapsing multiple sclerosis. EBioMedicine. 2017;16:41–50.PubMedPubMedCentral Baker D, et al. Memory B cells are major targets for effective immunotherapy in relapsing multiple sclerosis. EBioMedicine. 2017;16:41–50.PubMedPubMedCentral
9.
go back to reference Baker D, et al. CD19 B cell repopulation after ocrelizumab, alemtuzumab and cladribine: implications for SARS-CoV-2 vaccinations in multiple sclerosis. Mult Scler Relat Disord. 2022;57: 103448.PubMed Baker D, et al. CD19 B cell repopulation after ocrelizumab, alemtuzumab and cladribine: implications for SARS-CoV-2 vaccinations in multiple sclerosis. Mult Scler Relat Disord. 2022;57: 103448.PubMed
10.
go back to reference Zabalza A, et al. COVID-19 in multiple sclerosis patients: susceptibility, severity risk factors and serological response. Eur J Neurol. 2021;28(10):3384–95.PubMed Zabalza A, et al. COVID-19 in multiple sclerosis patients: susceptibility, severity risk factors and serological response. Eur J Neurol. 2021;28(10):3384–95.PubMed
11.
go back to reference van Lierop ZY, et al. Personalized B-cell tailored dosing of ocrelizumab in patients with multiple sclerosis during the COVID-19 pandemic. Mult Scler. 2022;28(7):1121–5.PubMed van Lierop ZY, et al. Personalized B-cell tailored dosing of ocrelizumab in patients with multiple sclerosis during the COVID-19 pandemic. Mult Scler. 2022;28(7):1121–5.PubMed
12.
go back to reference Rolfes L, et al. Ocrelizumab extended interval dosing in multiple sclerosis in times of COVID-19. Neurol Neuroimmunol Neuroinflamm. 2021;8(5): e1035.PubMedPubMedCentral Rolfes L, et al. Ocrelizumab extended interval dosing in multiple sclerosis in times of COVID-19. Neurol Neuroimmunol Neuroinflamm. 2021;8(5): e1035.PubMedPubMedCentral
13.
go back to reference van Kempen ZL, et al. Extended dosing of monoclonal antibodies in multiple sclerosis. Mult Scler. 2022;28(13):2001–9.PubMed van Kempen ZL, et al. Extended dosing of monoclonal antibodies in multiple sclerosis. Mult Scler. 2022;28(13):2001–9.PubMed
14.
go back to reference Bagwell CB, et al. Automated data cleanup for mass cytometry. Cytometry A. 2020;97(2):184–98.PubMed Bagwell CB, et al. Automated data cleanup for mass cytometry. Cytometry A. 2020;97(2):184–98.PubMed
15.
go back to reference Meskas J, et al. flowCut: an R package for automated removal of outlier events and flagging of files based on time versus fluorescence analysis. Cytometry A. 2023;103(1):71–81.PubMed Meskas J, et al. flowCut: an R package for automated removal of outlier events and flagging of files based on time versus fluorescence analysis. Cytometry A. 2023;103(1):71–81.PubMed
16.
go back to reference Crowell HL et al. An R-based reproducible and user-friendly preprocessing pipeline for CyTOF data. F1000Res. 2020; 9. Crowell HL et al. An R-based reproducible and user-friendly preprocessing pipeline for CyTOF data. F1000Res. 2020; 9.
17.
go back to reference Van Gassen S, et al. CytoNorm: a normalization algorithm for cytometry data. Cytometry A. 2020;97(3):268–78.PubMed Van Gassen S, et al. CytoNorm: a normalization algorithm for cytometry data. Cytometry A. 2020;97(3):268–78.PubMed
18.
go back to reference McInnes L, Healy J, Melville J. Umap: Uniform manifold approximation and projection for dimension reduction. arXiv preprint arXiv:1802.03426, 2018. McInnes L, Healy J, Melville J. Umap: Uniform manifold approximation and projection for dimension reduction. arXiv preprint arXiv:​1802.​03426, 2018.
19.
go back to reference Stassen SV, et al. PARC: ultrafast and accurate clustering of phenotypic data of millions of single cells. Bioinformatics. 2020;36(9):2778–86.PubMedPubMedCentral Stassen SV, et al. PARC: ultrafast and accurate clustering of phenotypic data of millions of single cells. Bioinformatics. 2020;36(9):2778–86.PubMedPubMedCentral
20.
go back to reference Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Annal Statis. 2001;29(4):1165–88. Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Annal Statis. 2001;29(4):1165–88.
21.
go back to reference Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Statis Soc Ser B (Methodolog). 1995;57(1):289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Statis Soc Ser B (Methodolog). 1995;57(1):289–300.
22.
go back to reference Luster AD, Alon R, von Andrian UH. Immune cell migration in inflammation: present and future therapeutic targets. Nat Immunol. 2005;6(12):1182–90.PubMed Luster AD, Alon R, von Andrian UH. Immune cell migration in inflammation: present and future therapeutic targets. Nat Immunol. 2005;6(12):1182–90.PubMed
23.
go back to reference Rodriguez-Mogeda C, et al. Breaching brain barriers: B cell migration in multiple sclerosis. Biomolecules. 2022;12(6):800.PubMedPubMedCentral Rodriguez-Mogeda C, et al. Breaching brain barriers: B cell migration in multiple sclerosis. Biomolecules. 2022;12(6):800.PubMedPubMedCentral
24.
go back to reference Nagasawa T. Microenvironmental niches in the bone marrow required for B-cell development. Nat Rev Immunol. 2006;6(2):107–16.PubMed Nagasawa T. Microenvironmental niches in the bone marrow required for B-cell development. Nat Rev Immunol. 2006;6(2):107–16.PubMed
25.
go back to reference Laidlaw BJ, Cyster JG. Transcriptional regulation of memory B cell differentiation. Nat Rev Immunol. 2021;21(4):209–20.PubMed Laidlaw BJ, Cyster JG. Transcriptional regulation of memory B cell differentiation. Nat Rev Immunol. 2021;21(4):209–20.PubMed
26.
go back to reference Dorcet G, et al. Early B cells repopulation in multiple sclerosis patients treated with rituximab is not predictive of a risk of relapse or clinical progression. J Neurol. 2022;269(10):5443–53.PubMedPubMedCentral Dorcet G, et al. Early B cells repopulation in multiple sclerosis patients treated with rituximab is not predictive of a risk of relapse or clinical progression. J Neurol. 2022;269(10):5443–53.PubMedPubMedCentral
27.
go back to reference Nissimov N, et al. B cells reappear less mature and more activated after their anti-CD20-mediated depletion in multiple sclerosis. Proc Natl Acad Sci. 2020;117(41):25690–9.PubMedPubMedCentral Nissimov N, et al. B cells reappear less mature and more activated after their anti-CD20-mediated depletion in multiple sclerosis. Proc Natl Acad Sci. 2020;117(41):25690–9.PubMedPubMedCentral
28.
go back to reference Palanichamy A, et al. Rituximab efficiently depletes increased CD20-expressing T cells in multiple sclerosis patients. J Immunol. 2014;193(2):580–6.PubMed Palanichamy A, et al. Rituximab efficiently depletes increased CD20-expressing T cells in multiple sclerosis patients. J Immunol. 2014;193(2):580–6.PubMed
29.
go back to reference Weiss N, et al. The blood–brain barrier in brain homeostasis and neurological diseases. Biochim Biophys Acta. 2009;1788(4):842–57.PubMed Weiss N, et al. The blood–brain barrier in brain homeostasis and neurological diseases. Biochim Biophys Acta. 2009;1788(4):842–57.PubMed
30.
go back to reference Rojas OL, et al. Recirculating intestinal IgA-producing cells regulate neuroinflammation via IL-10. Cell. 2019;176(3):610-624.e18.PubMedPubMedCentral Rojas OL, et al. Recirculating intestinal IgA-producing cells regulate neuroinflammation via IL-10. Cell. 2019;176(3):610-624.e18.PubMedPubMedCentral
31.
go back to reference Shen P, et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature. 2014;507(7492):366–70.PubMedPubMedCentral Shen P, et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature. 2014;507(7492):366–70.PubMedPubMedCentral
32.
go back to reference Chekol Abebe E, et al. The role of regulatory B cells in health and diseases: a systemic review. J Inflamm Res. 2021;14:75–84.PubMedPubMedCentral Chekol Abebe E, et al. The role of regulatory B cells in health and diseases: a systemic review. J Inflamm Res. 2021;14:75–84.PubMedPubMedCentral
33.
go back to reference Yanaba K, et al. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity. 2008;28(5):639–50.PubMed Yanaba K, et al. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity. 2008;28(5):639–50.PubMed
34.
go back to reference Chen Y et al. IL-10-producing CD1dhiCD5+ regulatory B cells may play a critical role in modulating immune homeostasis in silicosis patients. Front Immunol. 2017; 8. Chen Y et al. IL-10-producing CD1dhiCD5+ regulatory B cells may play a critical role in modulating immune homeostasis in silicosis patients. Front Immunol. 2017; 8.
35.
go back to reference Pavlasova G, Mraz M. The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy. Haematologica. 2020;105(6):1494–506.PubMedPubMedCentral Pavlasova G, Mraz M. The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy. Haematologica. 2020;105(6):1494–506.PubMedPubMedCentral
36.
go back to reference Kläsener K, et al. CD20 as a gatekeeper of the resting state of human B cells. Proc Natl Acad Sci. 2021;118(7): e2021342118.PubMedPubMedCentral Kläsener K, et al. CD20 as a gatekeeper of the resting state of human B cells. Proc Natl Acad Sci. 2021;118(7): e2021342118.PubMedPubMedCentral
37.
go back to reference Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discovery. 2021;20(3):179–99.PubMed Lee DSW, Rojas OL, Gommerman JL. B cell depletion therapies in autoimmune disease: advances and mechanistic insights. Nat Rev Drug Discovery. 2021;20(3):179–99.PubMed
38.
go back to reference Gingele S et al. Ocrelizumab depletes CD20+ T cells in multiple sclerosis patients. Cells. 2018; 8(1). Gingele S et al. Ocrelizumab depletes CD20+ T cells in multiple sclerosis patients. Cells. 2018; 8(1).
39.
go back to reference Quendt C, et al. Proinflammatory CD20(+) T cells are differentially affected by multiple sclerosis therapeutics. Ann Neurol. 2021;90(5):834–9.PubMed Quendt C, et al. Proinflammatory CD20(+) T cells are differentially affected by multiple sclerosis therapeutics. Ann Neurol. 2021;90(5):834–9.PubMed
40.
go back to reference Ochs J, et al. Proinflammatory CD20(+) T cells contribute to CNS-directed autoimmunity. Sci Transl Med. 2022;14(638):eabi6432. Ochs J, et al. Proinflammatory CD20(+) T cells contribute to CNS-directed autoimmunity. Sci Transl Med. 2022;14(638):eabi6432.
41.
go back to reference Garcia A, et al. Immune profiling reveals the T-cell effect of ocrelizumab in early relapsing-remitting multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2023;10(3):e200091.PubMedPubMedCentral Garcia A, et al. Immune profiling reveals the T-cell effect of ocrelizumab in early relapsing-remitting multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2023;10(3):e200091.PubMedPubMedCentral
42.
go back to reference Mathias A, et al. Ocrelizumab impairs the phenotype and function of memory CD8+ T cells. A 1-year longitudinal study in patients with multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2023;10(2):e200084.PubMedPubMedCentral Mathias A, et al. Ocrelizumab impairs the phenotype and function of memory CD8+ T cells. A 1-year longitudinal study in patients with multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2023;10(2):e200084.PubMedPubMedCentral
43.
go back to reference Laurent SA, et al. Effect of ocrelizumab on B- and T-cell receptor repertoire diversity in patients with relapsing multiple sclerosis from the randomized phase III OPERA Trial. Neurol Neuroimmunol Neuroinflamm. 2023;10(4):e200118.PubMedPubMedCentral Laurent SA, et al. Effect of ocrelizumab on B- and T-cell receptor repertoire diversity in patients with relapsing multiple sclerosis from the randomized phase III OPERA Trial. Neurol Neuroimmunol Neuroinflamm. 2023;10(4):e200118.PubMedPubMedCentral
44.
go back to reference Allen C, et al. Extended interval dosing of ocrelizumab reduces risk of developing hypogammaglobulinaemia. J Neurol Neurosurg Psychiatry. 2022;93(9): e2. Allen C, et al. Extended interval dosing of ocrelizumab reduces risk of developing hypogammaglobulinaemia. J Neurol Neurosurg Psychiatry. 2022;93(9): e2.
45.
go back to reference Shirah BH, Algahtani H. Personalized half-dose ocrelizumab in selected patients with relapsing-remitting multiple sclerosis. Mult Scler Relat Disord. 2023;71:104349. Shirah BH, Algahtani H. Personalized half-dose ocrelizumab in selected patients with relapsing-remitting multiple sclerosis. Mult Scler Relat Disord. 2023;71:104349.
46.
go back to reference Baker D, et al. The ocrelizumab phase II extension trial suggests the potential to improve the risk: benefit balance in multiple sclerosis. Mult Scler Relat Disord. 2020;44: 102279.PubMed Baker D, et al. The ocrelizumab phase II extension trial suggests the potential to improve the risk: benefit balance in multiple sclerosis. Mult Scler Relat Disord. 2020;44: 102279.PubMed
47.
go back to reference Baker D, et al. COVID-19 vaccine-readiness for anti-CD20-depleting therapy in autoimmune diseases. Clin Exp Immunol. 2020;202(2):149–61.PubMedPubMedCentral Baker D, et al. COVID-19 vaccine-readiness for anti-CD20-depleting therapy in autoimmune diseases. Clin Exp Immunol. 2020;202(2):149–61.PubMedPubMedCentral
48.
go back to reference Zanghì A, et al. Is it time for ocrelizumab extended interval dosing in relapsing remitting MS? Evidence from An Italian Multicenter Experience During the COVID-19 Pandemic. Neurotherapeutics. 2022;19(5):1535–45.PubMedPubMedCentral Zanghì A, et al. Is it time for ocrelizumab extended interval dosing in relapsing remitting MS? Evidence from An Italian Multicenter Experience During the COVID-19 Pandemic. Neurotherapeutics. 2022;19(5):1535–45.PubMedPubMedCentral
49.
go back to reference Du Pasquier RA, Pinschewer DD, Merkler D. Immunological mechanism of action and clinical profile of disease-modifying treatments in multiple sclerosis. CNS Drugs. 2014;28(6):535–58.PubMedPubMedCentral Du Pasquier RA, Pinschewer DD, Merkler D. Immunological mechanism of action and clinical profile of disease-modifying treatments in multiple sclerosis. CNS Drugs. 2014;28(6):535–58.PubMedPubMedCentral
50.
go back to reference Thompson SAJ, et al. B-cell reconstitution and BAFF after alemtuzumab (Campath-1H) treatment of multiple sclerosis. J Clin Immunol. 2010;30(1):99–105.PubMed Thompson SAJ, et al. B-cell reconstitution and BAFF after alemtuzumab (Campath-1H) treatment of multiple sclerosis. J Clin Immunol. 2010;30(1):99–105.PubMed
Metadata
Title
Extended interval dosing of ocrelizumab modifies the repopulation of B cells without altering the clinical efficacy in multiple sclerosis
Authors
Carla Rodriguez-Mogeda
Zoë Y. G. J. van Lierop
Susanne M. A. van der Pol
Loet Coenen
Laura Hogenboom
Alwin Kamermans
Ernesto Rodriguez
Jack van Horssen
Zoé L. E. van Kempen
Bernard M. J. Uitdehaag
Charlotte E. Teunissen
Maarten E. Witte
Joep Killestein
Helga E. de Vries
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2023
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-023-02900-z

Other articles of this Issue 1/2023

Journal of Neuroinflammation 1/2023 Go to the issue