Vol 54, No 6 (2020)
Invited Review Article
Published online: 2020-10-19

open access

Page views 2664
Article views/downloads 2197
Get Citation

Connect on Social Media

Connect on Social Media

B cell targeting therapies in MS patients during the SARS-CoV-2 pandemic — when immunosuppression meets infection?

Marcin P. Mycko1
Pubmed: 33073348
Neurol Neurochir Pol 2020;54(6):490-501.

Abstract

Introduction. Research into the mechanisms of autoimmune demyelination have highlighted B cells in this process. Therapies targeting this population were a recent addition to the multiple sclerosis (MS) drugs portfolio. The SARS-CoV-2 pandemic and the risk of severe COVID-19 have challenged the safety of B cell depletion in MS patients.

State of the art. Selective depletion of B cells by monoclonal antibodies as monotherapy in MS has been shown to profoundly suppress disease activity among relapsing-remitting MS patients. Furthermore ocrelizumab, a humanised anti-CD20 monoclonal antibody, was the first licensed therapy in primary progressive MS. Based on the concept of the role of B cells in MS, many therapeutic approaches are emerging as novel ways to treat autoimmune demyelination. However, during the SARS-CoV-2 pandemic, a conservative approach toward limiting immune suppression in MS patients has been proposed.

Clinical implications. Emerging evidence does not support the notion of increased susceptibility among MS patients to the SARS-CoV-2 infection, or any predisposition toward greater severity of COVID-19. This also does not appear to be the case for MS patients undergoing B cell depletion therapies. Thus, any decision to withhold immune suppression in MS patients during the SARS-CoV-2 pandemic is probably incorrect. MS therapeutic decision-making should focus on the danger of poorly controlled autoimmune demyelination rather than perceived risks from COVID-19.

Future directions. The current pandemic highlights the need to develop more selective and safer methods of immunomodulation in MS. B cells represent several functionally different populations. Further research into the different role of these cells during autoimmune demyelination should yield better, safer strategies to control the encephalitogenic process.

Article available in PDF format

View PDF Download PDF file

References

  1. Karlsson EK, Kwiatkowski DP, Sabeti PC. Natural selection and infectious disease in human populations. Nat Rev Genet. 2014; 15(6): 379–393.
  2. Casanova JL. Human genetic basis of interindividual variability in the course of infection. Proc Natl Acad Sci U S A. 2015; 112(51): E7118–E7127.
  3. Geginat J, Paroni M, Pagani M, et al. The Enigmatic Role of Viruses in Multiple Sclerosis: Molecular Mimicry or Disturbed Immune Surveillance? Trends Immunol. 2017; 38(7): 498–512.
  4. Bach JF. The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol. 2018; 18(2): 105–120.
  5. Steri M, Orrù V, Idda ML, et al. Overexpression of the Cytokine BAFF and Autoimmunity Risk. N Engl J Med. 2017; 376(17): 1615–1626.
  6. Thompson A, Baranzini S, Geurts J, et al. Multiple sclerosis. The Lancet. 2018; 391(10130): 1622–1636.
  7. Cotsapas C, Mitrovic M, Cotsapas C, et al. Multiple sclerosis. Handb Clin Neurol. 2018; 148(6): 723–730.
  8. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science. 2019; 365(6460).
  9. Mechelli R, Umeton R, Policano C, et al. International Multiple Sclerosis Genetics Consortium, Wellcome Trust Case Control Consortium,2. A "candidate-interactome" aggregate analysis of genome-wide association data in multiple sclerosis. PLoS One. 2013; 8(5): e63300.
  10. Zurawska A, Mycko MP, Selmaj KW. Circular RNAs as a novel layer of regulatory mechanism in multiple sclerosis. J Neuroimmunol. 2019; 334: 576971.
  11. Mycko MP, Baranzini SE. microRNA and exosome profiling in multiple sclerosis. Mult Scler. 2020; 26(5): 599–604.
  12. Cwiklinska H, Cichalewska-Studzinska M, Selmaj KW, et al. The Heat Shock Protein HSP70 Promotes Th17 Genes' Expression via Specific Regulation of microRNA. Int J Mol Sci. 2020; 21(8).
  13. Sospedra M, Martin R. Immunology of Multiple Sclerosis. Semin Neurol. 2016; 36(2): 115–127.
  14. Hemmer B, Kerschensteiner M, Korn T. Role of the innate and adaptive immune responses in the course of multiple sclerosis. Lancet Neurol. 2015; 14(4): 406–419.
  15. Jelcic I, Nimer FAl, Wang J, et al. Memory B Cells Activate Brain-Homing, Autoreactive CD4+ T Cells in Multiple Sclerosis. immuneACCESS. .
  16. Mycko MP, Waldner H, Anderson DE, et al. Cross-reactive TCR responses to self antigens presented by different MHC class II molecules. J Immunol. 2004; 173(3): 1689–1698.
  17. Harkiolaki M, Holmes SL, Svendsen P, et al. T cell-mediated autoimmune disease due to low-affinity crossreactivity to common microbial peptides. Immunity. 2009; 30(3): 348–357.
  18. Glatigny S, Bettelli E. Experimental Autoimmune Encephalomyelitis (EAE) as Animal Models of Multiple Sclerosis (MS). Cold Spring Harbor Perspectives in Medicine. 2018; 8(11): a028977.
  19. Burkett PR, Meyer zu Horste G, Kuchroo VK. Pouring fuel on the fire: Th17 cells, the environment, and autoimmunity. J Clin Invest. 2015; 125(6): 2211–2219.
  20. Bettelli E, Korn T, Oukka M, et al. Induction and effector functions of TH17 cells. Nature. 2008; 453(7198): 1051–1057.
  21. Hiltensperger M, Korn T. The Interleukin (IL)-23/T helper (Th)17 Axis in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Cold Spring Harb Perspect Med. 2018; 8(1).
  22. Bielekova B, Sung MH, Kadom N, et al. Expansion and functional relevance of high-avidity myelin-specific CD4+ T cells in multiple sclerosis. J Immunol. 2004; 172(6): 3893–3904.
  23. Cao Y, Goods BA, Raddassi K, et al. Functional inflammatory profiles distinguish myelin-reactive T cells from patients with multiple sclerosis. Sci Transl Med. 2015; 7(287): 287ra74.
  24. Galli E, Hartmann FJ, Schreiner B, et al. GM-CSF and CXCR4 define a T helper cell signature in multiple sclerosis. Nat Med. 2019; 25(8): 1290–1300.
  25. Lee Y, Collins M, Kuchroo VK. Unexpected targets and triggers of autoimmunity. J Clin Immunol. 2014; 34 Suppl 1: S56–S60.
  26. van Oosten BW, Lai M, Hodgkinson S, et al. Treatment of multiple sclerosis with the monoclonal anti-CD4 antibody cM-T412: results of a randomized, double-blind, placebo-controlled, MR-monitored phase II trial. Neurology. 1997; 49(2): 351–357.
  27. van Oosten BW, Lai M, Hodgkinson S, et al. A phase II trial of anti-CD4 antibodies in the treatment of multiple sclerosis. Mult Scler. 1996; 1(6): 339–342.
  28. Havrdová E, Belova A, Goloborodko A, et al. Activity of secukinumab, an anti-IL-17A antibody, on brain lesions in RRMS: results from a randomized, proof-of-concept study. J Neurol. 2016; 263(7): 1287–1295.
  29. Juryńczyk M, Walczak A, Jurewicz A, et al. Immune regulation of multiple sclerosis by transdermally applied myelin peptides. Ann Neurol. 2010; 68(5): 593–601.
  30. Magliozzi R, Cross AH. Can CSF biomarkers predict future MS disease activity and severity? Mult Scler. 2020; 26(5): 582–590.
  31. Hartung HP, Graf J, Aktas O, et al. Diagnosis of multiple sclerosis: revisions of the McDonald criteria 2017 - continuity and change. Curr Opin Neurol. 2019; 32(3): 327–337.
  32. Eggers EL, Michel BA, Wu H, et al. Clonal relationships of CSF B cells in treatment-naive multiple sclerosis patients. JCI Insight. 2017; 2(22).
  33. Hauser SL. The Charcot Lecture | beating MS: a story of B cells, with twists and turns. Mult Scler. 2015; 21(1): 8–21.
  34. Sabatino JJ, Pröbstel AK, Zamvil SS. B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci. 2019; 20(12): 728–745.
  35. Thi Cuc B, Pohar J, Fillatreau S. Understanding regulatory B cells in autoimmune diseases: the case of multiple sclerosis. Curr Opin Immunol. 2019; 61: 26–32.
  36. Chen D, Ireland SJ, Remington G, et al. CD40-Mediated NF-kappaB Activation in B Cells Is Increased in Multiple Sclerosis and Modulated by Therapeutics. J Immunol. 2016; 197: 4257–65.
  37. Li R, Rezk A, Miyazaki Y, et al. Canadian B cells in MS Team. Proinflammatory GM-CSF-producing B cells in multiple sclerosis and B cell depletion therapy. Sci Transl Med. 2015; 7(310): 310ra166.
  38. Stein J, Xu Q, Jackson KC, et al. Intrathecal B Cells in MS Have Significantly Greater Lymphangiogenic Potential Compared to B Cells Derived From Non-MS Subjects. Front Neurol. 2018; 9: 554.
  39. Giacomini E, Rizzo F, Etna MP, et al. Thymosin-α1 expands deficient IL-10-producing regulatory B cell subsets in relapsing-remitting multiple sclerosis patients. Mult Scler. 2018; 24(2): 127–139.
  40. Brod SA. In MS: Immunosuppression is passé. Mult Scler Relat Disord. 2020; 40: 101967.
  41. Hauser SL, Waubant E, Arnold DL, et al. HERMES Trial Group. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008; 358(7): 676–688.
  42. Hauser SL, Bar-Or A, Comi G, et al. OPERA I and OPERA II Clinical Investigators. Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis. N Engl J Med. 2017; 376(3): 221–234.
  43. Bar-Or A, Grove RA, Tolson JM, et al. Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: The MIRROR study. Neurology. 2018; 90(20): e1805–e1814.
  44. Fox E, Lovett-Racke AE, Gormley M, et al. A phase 2 multicenter study of ublituximab, a novel glycoengineered anti-CD20 monoclonal antibody, in patients with relapsing forms of multiple sclerosis. Mult Scler. 2020 [Epub ahead of print]: 1352458520918375.
  45. Agius MA, Klodowska-Duda G, Maciejowski M, et al. Safety and tolerability of inebilizumab (MEDI-551), an anti-CD19 monoclonal antibody, in patients with relapsing forms of multiple sclerosis: Results from a phase 1 randomised, placebo-controlled, escalating intravenous and subcutaneous dose study. Mult Scler. 2019; 25(2): 235–245.
  46. Kappos L, Hartung HP, Freedman MS, et al. ATAMS Study Group. Atacicept in multiple sclerosis (ATAMS): a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Neurol. 2014; 13(4): 353–363.
  47. Montalban X, Hauser SL, Kappos L, et al. ORATORIO Clinical Investigators. Ocrelizumab versus Placebo in Primary Progressive Multiple Sclerosis. N Engl J Med. 2017; 376(3): 209–220.
  48. Crofford LJ, Nyhoff LE, Sheehan JH, et al. The role of Bruton's tyrosine kinase in autoimmunity and implications for therapy. Expert Rev Clin Immunol. 2016; 12(7): 763–773.
  49. Haselmayer P, Camps M, Liu-Bujalski L, et al. Efficacy and Pharmacodynamic Modeling of the BTK Inhibitor Evobrutinib in Autoimmune Disease Models. J Immunol. 2019; 202(10): 2888–2906.
  50. Montalban X, Arnold DL, Weber MS, et al. Evobrutinib Phase 2 Study Group. Placebo-Controlled Trial of an Oral BTK Inhibitor in Multiple Sclerosis. N Engl J Med. 2019; 380(25): 2406–2417.
  51. https://www.sanofi.com/en/media-room/press-releases/2020/2020-02-06-07-00-00 (Setember 5th, 2020.).
  52. Rip J, Van Der Ploeg EK, Hendriks RW, et al. The Role of Bruton's Tyrosine Kinase in Immune Cell Signaling and Systemic Autoimmunity. Crit Rev Immunol. 2018; 38(1): 17–62.
  53. Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019; 17(3): 181–192.
  54. Amanat F, Krammer F. SARS-CoV-2 Vaccines: Status Report. Immunity. 2020; 52(4): 583–589.
  55. de Wit E, van Doremalen N, Falzarano D, et al. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol. 2016; 14(8): 523–534.
  56. Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell. 2020; 181(5): 1036–1045.e9.
  57. Netea MG, Giamarellos-Bourboulis EJ, Domínguez-Andrés J, et al. Trained Immunity: a Tool for Reducing Susceptibility to and the Severity of SARS-CoV-2 Infection. Cell. 2020; 181(5): 969–977.
  58. Tay MZ, Poh CM, Rénia L, et al. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020; 20(6): 363–374.
  59. Mehta P, McAuley DF, Brown M, et al. HLH Across Speciality Collaboration, UK. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020; 395(10229): 1033–1034.
  60. Vardhana SA, Wolchok JD. The many faces of the anti-COVID immune response. J Exp Med. 2020; 217(6).
  61. Cao Y, Guo X, Sun W, et al. Potent Neutralizing Antibodies against SARS-CoV-2 Identified by High-Throughput Single-Cell Sequencing of Convalescent Patients' B Cells. Cell. 2020 (182): 73–84 e16.
  62. Ni L, Cheng ML, Feng Y, et al. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity. 2020 (52): 971–977 e3.
  63. Vabret N, Britton GJ, Gruber C, et al. Sinai Immunology Review Project. Immunology of COVID-19: Current State of the Science. Immunity. 2020; 52(6): 910–941.
  64. Akkaya M, Kwak K, Pierce SKB. cell memory: building two walls of protection against pathogens. Nat Rev Immunol. 2020; 20: 229–38.
  65. Ciotti JR, Valtcheva MV, Cross AH. Effects of MS disease-modifying therapies on responses to vaccinations: A review. Mult Scler Relat Disord. 2020 [Epub ahead of print]; 45: 102439.
  66. Willis MD, Robertson NP. Multiple sclerosis and the risk of infection: considerations in the threat of the novel coronavirus, COVID-19/SARS-CoV-2. J Neurol. 2020; 267(5): 1567–1569.
  67. Luna G, Alping P, Burman J, et al. Infection Risks Among Patients With Multiple Sclerosis Treated With Fingolimod, Natalizumab, Rituximab, and Injectable Therapies. JAMA Neurol. 2020; 77(2): 184–191.
  68. Hartung HP, Aktas O. COVID-19 and management of neuroimmunological disorders. Nat Rev Neurol. 2020; 16(7): 347–348.
  69. Sormani M. An Italian programme for COVID-19 infection in multiple sclerosis. The Lancet Neurology. 2020; 19(6): 481–482.
  70. Ciampi E, Uribe-San-Martin R, Cárcamo C. COVID-19 pandemic: The experience of a multiple sclerosis centre in Chile. Mult Scler Relat Disord. 2020; 42: 102204.
  71. Rejdak K, Grieb P. Adamantanes might be protective from COVID-19 in patients with neurological diseases: multiple sclerosis, parkinsonism and cognitive impairment. Mult Scler Relat Disord. 2020; 42: 102163.
  72. D'Antiga L. Coronaviruses and Immunosuppressed Patients: The Facts During the Third Epidemic. Liver Transpl. 2020; 26(6): 832–834.
  73. Baker D, Amor S, Kang AS, et al. The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID-19 pandemic. Mult Scler Relat Disord. 2020; 43: 102174.
  74. Brownlee W, Bourdette D, Broadley S, et al. Treating multiple sclerosis and neuromyelitis optica spectrum disorder during the COVID-19 pandemic. Neurology. 2020; 94(22): 949–952.
  75. Baker D, Roberts CAK, Pryce G, et al. COVID-19 vaccine-readiness for anti-CD20-depleting therapy in autoimmune diseases. Clin Exp Immunol. 2020 [Epub ahead of print].
  76. Ziemssen T, Bar-Or A, Arnold DL, et al. P 2 Effect of ocrelizumab on humoral immunity markers in the phase iii, double-blind, double-dummy, IFNβ-1a–controlled OPERA I and OPERA II studies. Clinical Neurophysiology. 2017; 128(10): e326–e327.
  77. Stokmaier DWK, Chognot C, et al. Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis. Presented at the American Academy of Neurology Annual Meeting in Los Angeles, CA. AAN Oral Presentation #S3. 6002; 2018.
  78. 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. Multiple Sclerosis and Related Disorders. 2018; 26: 264.
  79. Derfuss T, Weber M, Hughes R, et al. P36 Serum immunoglobulin levels and risk of serious infections in the pivotal phase III trials of ocrelizumab in multiple sclerosis and their open-label extensions. Clinical Neurophysiology. 2020; 131(4): e196.
  80. Moreno Torres I, García-Merino A. Anti-CD20 monoclonal antibodies in multiple sclerosis. Expert Rev Neurother. 2017; 17(4): 359–371.
  81. Midaglia L, Mora L, Mulero P, et al. [Rituximab: its efficacy, effectiveness and safety in the treatment of multiple sclerosis]. Rev Neurol. 2018; 66(1): 25–32.
  82. Meca-Lallana V, Aguirre C, Cardeñoso L, et al. COVID-19 in 7 multiple sclerosis patients in treatment with ANTI-CD20 therapies. Mult Scler Relat Disord. 2020 [Epub ahead of print]; 44: 102306.
  83. Louapre C, Collongues N, Stankoff B, et al. Covisep investigators. Clinical Characteristics and Outcomes in Patients With Coronavirus Disease 2019 and Multiple Sclerosis. JAMA Neurol. 2020 [Epub ahead of print].
  84. Milo R, Milo R. Therapeutic strategies targeting B-cells in multiple sclerosis. Autoimmun Rev. 2016; 15(7): 714–718.
  85. He A, Merkel B, Brown JW, et al. MSBase study group. Timing of high-efficacy therapy for multiple sclerosis: a retrospective observational cohort study. Lancet Neurol. 2020; 19(4): 307–316.
  86. 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(5): 536–541.
  87. Hawker K, O'Connor P, Freedman MS, et al. OLYMPUS trial group. Rituximab in patients with primary progressive multiple sclerosis: results of a randomized double-blind placebo-controlled multicenter trial. Ann Neurol. 2009; 66(4): 460–471.
  88. Novi G, Mikulska M, Briano F, et al. COVID-19 in a MS patient treated with ocrelizumab: does immunosuppression have a protective role? Mult Scler Relat Disord. 2020; 42: 102120.
  89. Hughes R, Pedotti R, Koendgen H. COVID-19 in persons with multiple sclerosis treated with ocrelizumab - A pharmacovigilance case series. Mult Scler Relat Disord. 2020; 42: 102192.
  90. Montero-Escribano P, Matías-Guiu J, Gómez-Iglesias P, et al. Anti-CD20 and COVID-19 in multiple sclerosis and related disorders: A case series of 60 patients from Madrid, Spain. Mult Scler Relat Disord. 2020; 42: 102185.
  91. Ghajarzadeh M, Mirmosayyeb O, Barzegar M, et al. Favorable outcome after COVID-19 infection in a multiple sclerosis patient initiated on ocrelizumab during the pandemic. Mult Scler Relat Disord. 2020; 43: 102222.
  92. Wurm H, Attfield K, Iversen AK, et al. Recovery from COVID-19 in a B-cell-depleted multiple sclerosis patient. Mult Scler. ; 2020: 1352458520943791.
  93. Louapre C, Maillart E, Roux T, et al. Patients with MS treated with immunosuppressive agents: Across the COVID-19 spectrum. Rev Neurol (Paris). 2020; 176(6): 523–525.
  94. Parrotta E, Kister I, Charvet L, et al. COVID-19 outcomes in MS: Observational study of early experience from NYU Multiple Sclerosis Comprehensive Care Center. Neurol Neuroimmunol Neuroinflamm. 2020; 7(5).
  95. Safavi F, Nourbakhsh B, Azimi AR. B-cell depleting therapies may affect susceptibility to acute respiratory illness among patients with multiple sclerosis during the early COVID-19 epidemic in Iran. Mult Scler Relat Disord. 2020; 43: 102195.
  96. Suwanwongse K, Shabarek N. Benign course of COVID-19 in a multiple sclerosis patient treated with Ocrelizumab. Mult Scler Relat Disord. 2020; 42: 102201.
  97. Conte WL. Attenuation of antibody response to SARS-CoV-2 in a patient on ocrelizumab with hypogammaglobulinemia. Mult Scler Relat Disord. 2020; 44: 102315.
  98. Lucchini M, Bianco A, Del Giacomo P, et al. Is serological response to SARS-CoV-2 preserved in MS patients on ocrelizumab treatment? A case report. Mult Scler Relat Disord. 2020 [Epub ahead of print]; 44: 102323.
  99. Thornton JR, Harel A. Negative SARS-CoV-2 antibody testing following COVID-19 infection in Two MS patients treated with ocrelizumab. Mult Scler Relat Disord. 2020 [Epub ahead of print]; 44: 102341.
  100. Barzegar M, Mirmosayyeb O, Ghajarzadeh M, et al. Characteristics of COVID-19 disease in multiple sclerosis patients. Mult Scler Relat Disord. 2020 [Epub ahead of print]; 45: 102276.



Neurologia i Neurochirurgia Polska