Hostname: page-component-76fb5796d-wq484 Total loading time: 0 Render date: 2024-04-26T20:26:48.898Z Has data issue: false hasContentIssue false

B-Cell-Directed Therapies: A New Era in Multiple Sclerosis Treatment

Published online by Cambridge University Press:  16 May 2022

Panagiotis Kanatas
Affiliation:
First Department of Neurology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece Eginiteion Hospital, Athens, Greece
Ioannis Stouras
Affiliation:
First Department of Neurology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
Leonidas Stefanis
Affiliation:
First Department of Neurology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece Eginiteion Hospital, Athens, Greece
Panos Stathopoulos*
Affiliation:
First Department of Neurology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece Eginiteion Hospital, Athens, Greece
*
Corresponding author: Panos Stathopoulos, Eginiteion Hospital, V. Sofias 72, 115 28 Athens, Greece. Email: pmstathopoulos@gmail.com
Rights & Permissions [Opens in a new window]

Abstract:

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system (CNS) that often progresses to severe disability. Previous studies have highlighted the role of T cells in disease pathophysiology; however, the success of B-cell-targeted therapies has led to an increased interest in how B cells contribute to disease immunopathology. In this review, we summarize evidence of B-cell involvement in MS disease mechanisms, starting with pathology and moving on to review aspects of B cell immunobiology potentially relevant to MS. We describe current theories of critical B cell contributions to the inflammatory CNS milieu in MS, namely (i) production of autoantibodies, (ii) antigen presentation, (iii) production of proinflammatory cytokines (bystander activation), and (iv) EBV involvement. In the second part of the review, we summarize medications that have targeted B cells in patients with MS and their current position in the therapeutic armamentarium based on clinical trials and real-world data. Covered therapeutic strategies include the targeting of surface molecules such as CD20 (rituximab, ocrelizumab, ofatumumab, ublituximab) and CD19 (inebilizumab), and molecules necessary for B-cell activation such as B cell activating factor (BAFF) (belimumab) and Bruton’s Tyrosine Kinase (BTK) (evobrutinib). We finally discuss the use of B-cell-targeted therapeutics in pregnancy.

Résumé :

RÉSUMÉ :

Les traitements ciblant les lymphocytes B dans la sclérose en plaques : nouvelle ère thérapeutique en vue.

La sclérose en plaques (SP) est une maladie auto-immune chronique démyélinisante du système nerveux central (SNC) qui aboutit souvent à une grande incapacité. Le rôle des lymphocytes T dans la physiopathologie de la maladie a déjà été mis en évidence dans des études antérieures, mais les bons résultats des traitements ciblant les lymphocytes B ont suscité de l’intérêt pour le rôle de ces derniers dans l’immunopathologie de la maladie. Aussi présenterons-nous dans l’article de synthèse des données probantes qui font ressortir l’action des lymphocytes B dans les mécanismes d’évolution de la SP, depuis la maladie elle-même jusqu’aux éléments immunobiologiques des lymphocytes B potentiellement associés à la SP. Dans la première partie, il sera question des théories existantes sur le rôle fondamental que jouent les lymphocytes B dans le milieu inflammatoire du SNC, dans la SP, à savoir i) la production d’autoanticorps; ii) la présentation d’antigènes; iii) la production de cytokines pro-inflammatoires (activation de voisinage); iv) le rôle du virus d’Epstein-Barr. Dans la seconde partie, nous présenterons un résumé des médicaments qui ciblent les lymphocytes B chez les patients atteints de la SP, et discuterons de leur place dans l’arsenal thérapeutique de la maladie d’après les résultats d’essais cliniques et des données réelles. Les stratégies thérapeutiques traitées dans l’article porteront notamment sur la prise pour cible des molécules présentes à la surface des lymphocytes telles que la CD20 (par le rituximab, l’ocrélizumab, l’ofatumumab ou l’ublituximab) et la CD19 (par l’inébilizumab), ainsi que sur la prise pour cible des molécules nécessaires à l’activation des lymphocytes B tel que le facteur d’activation des lymphocytes B (BAFF, en anglais) (par le bélimumab), et à l’inhibition de la tyrosine-kinase de Bruton (par l’évobrutinib). Enfin, il sera question de l’emploi des traitements ciblant les lymphocytes B chez les femmes enceintes.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of Canadian Neurological Sciences Federation

Introduction

Multiple sclerosis (MS) is a chronic autoimmune demyelinating inflammatory disease of the central nervous system (CNS), in the majority of cases gradually leading to progressive, severe disability if left untreated. MS is the leading cause of non-traumatic disability among young adults in the developed world. It is most often diagnosed between 20 and 40 years of age and affects women and men at a ratio of approximately 2:1. Reference Yeshokumar, Narula and Banwell1Reference Filippi, Bar-Or and Piehl3 The clinical course of MS can be characterized as (i) clinically isolated syndrome (CIS), (ii) relapsing-remitting (RRMS), (iii) primary progressive (PPMS), or (iv) secondary progressive (SPMS). Each of the above MS categories can be further subcategorized as either active or inactive, based on both the clinical relapse rate and MRI findings (new T2 lesions and/or active, gadolinium-enhancing lesions-GdELs). Further, progressive forms can be subcategorized as actively progressive or stable. Reference Lublin4

Significant progress in understanding MS pathophysiology has been accomplished in the past decades. Two hundred and thirty-three genetic variants have been identified as risk factors for MS, 32 of which refer to the major histocompatibility complex family (MHC). 5 Prominent among the many risk variants, MHC Class II DR15 molecule entails mechanistically relevant susceptibility to the disease rather than just being a genetic marker. Reference O’Connor, Bar-Or and Hafler6 Additional genetic variants associated with the disease refer to other genes of the immune system, such as genes involved in T-cell activation and proliferation (IL-2, IL-7R), tumor necrosis factor-alpha (TNF-α)-related pathways, and vitamin D metabolic pathways (GC, CYP24A1). Reference Filippi, Bar-Or and Piehl3,Reference De Jager, Jia and Wang7Reference Mokry, Ross and Ahmad11

In MS, myelin is phagocytosed by CD68-positive macrophages, while immune cells including B and T cells seem to be activated in the periphery and to express adherence molecules that enable them to cross blood-brain barrier (BBB) in order to participate in the formation of MS lesions. Accumulating evidence suggests an important contribution of CD4+ T cells to disease pathophysiology. Reference Ota, Matsui, Milford, Mackin, Weiner and Hafler12 Often being present from the beginning and increasing in quantity as disease progresses, axonal degeneration is regarded as a correlate of disability progression. Reference Bjartmar, Yin and Trapp13

MS was long considered mainly T-cell-mediated; however, intrathecal IgG synthesis, Reference Bonnan14 a hallmark of MS, supports B-cell involvement. Reference Cepok, Rosche and Grummel15,Reference Kowarik, Cepok and Sellner16 The T-cell-dominated view of MS pathogenesis was further challenged by the remarkable efficiency of CD20+ B-cell depletion in eliminating inflammatory activity in patients with MS. In this review, we aim to shed light on the key role B cells play in the pathogenesis of MS and present current advances in MS treatment strategies based on promising and effective B-cell-targeted therapeutic regimens.

B Cells in MS Pathology

Histological studies of active MS lesions have demonstrated that B cells can reside in the perivascular space and the CSF but also within the parenchyma. Reference Krumbholz, Derfuss, Hohlfeld and Meinl17 Moreover, ectopic lymphoid follicles are found primarily in the intrameningeal spaces. Reference Kivisäkk, Imitola and Rasmussen18 and are associated with subpial cortical demyelination in patients with SPMS. Reference Moreno Torres and García-Merino19,Reference Lassmann20 In addition, four histopathological patterns have been proposed for the classification of acute MS plaques. Type I lesions (15% of MS patients) are characterized by a T-cell and activated microglia inflammatory environment without immunoglobulin deposition and complement activation. On the contrary, type II lesions (58% of MS patients) develop in an inflammatory milieu with immunoglobulin production and complement activation. Demyelination in type III lesions (26% of MS patients) is accompanied by oligodendrocyte apoptosis without immunoglobulin deposition or complement activation. Finally, in type IV lesions (rare; 1% of MS patients) inflammatory modulators result in nonapoptotic death of oligodendrocytes in the white matter surrounding the plaque due to metabolic disorganization processes. Reference Gh Popescu, Pirko and Lucchinetti21 However, it is important to note that IgG deposits in MS histopathological specimens are not specific for MS Reference Barnett, Parratt, Cho and Prineas22 and that no disease-characterizing autoantibodies have been defined to date. Nevertheless, pattern II has been linked to better response to plasma exchange. Reference Keegan, König and McClelland23

Potential Roles of B Cells in MS Pathophysiology

Several studies have explored potential roles of B cells in the development of MS: antibody production, antigen presentation, and secretion of pro- and anti-inflammatory mediators are three prominent research directions that have been explored. Reference Gasperi, Stüve and Hemmer24,Reference Häusser-Kinzel and Weber25 In addition, clear epidemiological associations of B-lymphotropic Epstein-Barr virus (EBV) infection to MS have led to the explorations of its pathophysiological relevance. Reference Guan, Jakimovski, Ramanathan, Weinstock-Guttman and Zivadinov26

Autoantibodies

Autoreactive B cells that escape peripheral tolerance checkpoint selection could target antigens of the CNS and cause autoimmune inflammation; however, no consistent B-cell antigen that is specific for MS and that causes demyelination has been identified to date despite numerous attempts. Intrathecal oligoclonal bands, a hallmark of MS diagnosis found in up to 95% of patients, Reference Abdelhak, Hottenrott and Mayer27,Reference Thouvenot28 are not specific for MS (found also in e.g. meningitis and subacute sclerosing panencephalitis) and have been found to target intracellular antigens in patients with MS. Reference Brändle, Obermeier and Senel29,Reference Willis, Stathopoulos, Chastre, Compton, Hafler and O’Connor30 In addition, detection of intrathecal IgM synthesis has been associated with onset of relapses and a more aggressive disease course. Reference Villar, Masjuan and González-Porqué31 Similarly to antibodies of oligoclonal bands, B cells of MS lesions have been found to target intracellular antigens. Reference Willis, Stathopoulos, Chastre, Compton, Hafler and O’Connor30 Antibodies previously thought to be present in MS such as antibodies against myelin oligodendrocyte glycoprotein (MOG) rather characterize a distinct disease entity (MOG-antibody disease) that encompasses pediatric acquired demyelinating syndrome, recurrent optic neuritis, acute disseminated encephalomyelitis, and neuromyelitis optica without anti-aquaporin four autoantibodies. A minority of MS patients harbor antibodies against a variety of antigens (some of them cell surface proteins) such as contactin-2, Reference Boronat, Sepúlveda and Llufriu32 OMGP, Reference Gerhards, Pfeffer and Lorenz33 and other peptide and lipid antigens. Reference Yeste and Quintana34,Reference Kanter, Narayana and Ho35

It must be noted that autoantibodies can also be responsible for the activation and chemotaxis of CD4+ T cells. The opsonization of myelin antigens, even at low concentrations, enhances the presentative competence of resident antigen-presenting cells (APCs), such as macrophages and dendritic cells, leading to increased recruitment of effector T cells and, consequently, aggravation of the disease severity, as explained in more detail below. Reference Trotter, DeJong and Smith36Reference Getahun, Dahlström, Wernersson and Heyman38

B Cells as Antigen-Presenting Cells

B cells are efficient APCs and express MHC class II and costimulatory molecules, such as CD40, CD80, and CD86. Reference Häusser-Kinzel and Weber25,Reference Kinzel and Weber39 They can capture soluble and membrane-tethered antigens via their B-cell receptor (BCR) and present them to T cells in an up-to-a 10.000 times more efficient way compared to myeloid APCs. Reference Ancau, Berthele and Hemmer40 Evidence from experimental autoimmune encephalitis, a rodent model simulating "efferent" MS pathophysiology, opposes the hypothesis that the antigen-presenting function of B cells is central to the pathophysiology of MS. Specifically, MOG-specific B cells may initiate CNS inflammation and, consequently, the symptomatic onset of the disease, but do not affect either the proliferation or the molecular profile (i.e. secreted cytokines, activation markers) of MOG-specific T cells in the spleen and the draining lymph nodes. Reference Flach, Litke and Strauss37

On the other hand, a recently published report focusing on human leukocyte antigen (HLA)-DR15, which is the major genetic risk factor for MS, addresses how the immunopeptidomes presented by both DR15 allomorphs, DR2a and DR2b, on different APCs in the thymus, peripheral blood, and brain – including B cells – could affect autoimmune T cells. The results showed that DR2a and DR2b immunopeptidomes on B cells are significantly skewed toward HLA-DR-self peptides (HLA-DR-SPs) – compared to monocytes – which are consequently presented to autoreactive CD4+ T cells. These T cells responded robustly to individual and pooled HLA-DR-SPs in MS patients, compared to healthy donors, suggesting that DR2a and DR2b could jointly shape an autoreactive T-cell repertoire in MS. Reference Wang, Jelcic and Mühlenbruch41

B Cells as a Source of Cytokines

Physiologically, B cells can be a source of both proinflammatory and anti-inflammatory (regulatory) cytokines. B cells of RRMS patients however feature a profile that is skewed towards an abnormally hyperactive proinflammatory response. In mice, high levels of B-cell-secreted IL-6 can foster the differentiation of Th17 cells, while preventing the generation of T regulatory cells. Reference Bettelli, Carrier and Gao42Reference Korn, Mitsdoerffer and Croxford44 In MS patients, B-cell production of lymphotoxin alpha (LT-α), TNF-α, and granulocyte macrophage-colony stimulating factor (GM-CSF) appears elevated, forging a chronically inflammatory milieu within the CNS. Reference Moreno Torres and García-Merino19,Reference Häusser-Kinzel and Weber25,Reference Li, Rezk and Miyazaki45 At the same time, anti-inflammatory, regulatory cytokines that are produced by B cells, such as IL-35, Reference Shen, Roch and Lampropoulou46 but also TGF-β1 and IL-10, are instrumental in controlling inflammation in experimental models of MS. Reference Arneth47

EBV in MS

Among the infectious factors examined, the B-lymphotropic EBV has been shown to confer increased risk of developing MS, via, as yet, unclear mechanisms. Reference Morandi, Jagessar, ‘t Hart and Gran48 Ninety-six percent of the general population is positive for IgG antibodies against EBV (indicating past infection), while in MS patients this percentage is almost 100%. Reference Guan, Jakimovski, Ramanathan, Weinstock-Guttman and Zivadinov26 Moreover, a prospective cohort study of 955 incident MS patients showed a 97% of EBV (but not other viruses) seroconversion before development of the disease, significantly higher compared to controls. Reference Kjetil, Marianna and H.B.49 Studies and experiments have shaped four major theories about EBV’s role in the pathogenesis of MS: the cross-reactivity hypothesis, Reference Lang, Jacobsen and Ikemizu50 the bystander damage hypothesis, Reference Angelini, Serafini and Piras51 the αβ-crystallin hypothesis, Reference Pender and Burrows52 and the EBV-infected autoreactive B-cell hypothesis. Reference Pender53 Cellular and CSF findings however only partially match the pathophysiology of MS as far as the first three hypotheses are concerned. One important finding is the recent demonstration of molecular mimicry between EBV transcription factor EBNA1 and CNS protein glial cell adhesion molecule (GlialCAM), leading to the production of cross-reactive antibodies with higher affinity towards an intracellular GlialCAM epitope. Reference Lanz, Brewer and Ho54 The fourth hypothesis postulates that EBV-infected autoreactive B cells accumulate in the target organ and orchestrate the disease by producing antibodies and stimulating T cells due to a defect in their elimination by the antiviral CD8+ T cells. Moreover, the EBV antiapoptotic protein BHRF1, produced by both latently and lytically infected cells, inhibits B-cell apoptosis, Reference McCarthy, Hazlewood, Huen, Rickinson and Williams55 resulting in immortalization of the autoreactive B cells it infects. In support of this hypothesis, substantial EBV persistence in B and plasma cells as well as meningeal B-cell lymphoid follicles of all MS cases examined was reported, Reference Serafini, Rosicarelli and Franciotta56 but could not be reproduced in multiple independent replication studies. Reference Sargsyan, Shearer and Ritchie57Reference Torkildsen, Stansberg and Angelskår60 Overall, the epidemiological associations remain; however, no underlying biological mechanism has been consequently supported by experimental data.

Anti-B-cell Agents as a Therapeutic Strategy

Most anti-B-cell agents are monoclonal antibodies (mAbs); however, small molecules have also emerged as promising agents and have better CNS penetrance (Table 1). B-cell-depleting antibodies can be categorized in 1st-, 2nd-, or 3rd-generation. 1st-generation monoclonal antibodies (mAbs) can be either fully murine (suffix: -omab) or chimeric (65% human, suffix: -ximab), while 2nd-generation ones can be humanized (>90% human, suffix: -zumab) or even fully human (suffix: -mumab). 3rd-generation mAbs consist of a modified Fc region, chimeric, or humanized. Immunogenicity in theory ranges from higher in 1st-generation mAbs to lower in 2nd- and 3rd-generation ones. Reference Whittam, Tallantyre and Jolles61

Table 1: A summary of medicines targeting B cells that have been used in MS

Anti-CD20 mAbs

CD20 is a 33-37kDa transmembrane protein, which spans the membrane four times, thus consisting of two extracellular loops and intracellular C- and N-termini. Although some T cells with CD20 surface expression can be found in all lymphatic organs, are often CD8-positive, can be myelin-specific, Reference Schuh, Berer and Mulazzani62Reference Sabatino, Wilson, Calabresi, Hauser, Schneck and Zamvil64 and may correlate positively with disease severity, Reference von Essen, Ammitzbøll and Hansen65 CD20 serves a more important role on B cells. The molecule is not expressed throughout the entirety of the B-cell line of differentiation, but only in pre-B cells and mature B cells, with stem cells and the majority of antibody-secreting cells being CD20-negative (Figure 1). Reference Payandeh, Bahrami and Hoseinpoor66Reference Tedder, Streuli, Schlossman and Saito68 Physiologically, CD20 plays a key role as regulator of calcium influx in the signaling pathways that lead to B-cell differentiation into antibody-secreting plasma cells Reference Santos and Lima69 and its presence on the surface of most, but not all, B cells makes it an attractive target for monoclonal antibody-based therapy. B cells targeted by anti-CD20 monoclonal antibodies are eliminated via three main mechanisms: programmed cell death / apoptosis, complement-dependent cytotoxicity (CDC), or antibody-dependent cellular cytotoxicity (ADCC) processes. Reference Maloney70 Evidence from animal studies shows that anti-CD20 antibody-mediated B-cell depletion may be incomplete in lymph node germinal centers. Reference Schroder, Azimzadeh, Wu, Price, Atkinson and Pierson71 Moreover, cerebrospinal fluid B cells seem to be less affected than peripheral B cells by intravenous rituximab (the first anti-CD20 monoclonal antibody) administration, although the drug itself can be detected in a very low concentration (up to 1000 times lower than in the periphery) behind the BBB. Reference Piccio, Naismith and Trinkaus72Reference Monson, Cravens, Frohman, Hawker and Racke75 Of note, the limited access of anti-CD20 mAbs to the CNS due to their relatively high molecular weight could, at least to some extent, be overcome with intrathecal (IT) administration of anti-CD20 mAbs. The four main antibodies evaluated for anti-CD20 MS therapy are analyzed below.

Figure 1: Expression of cell surface antigens throughout B-cell maturation. CD19 is expressed in all stages of B-cell development, with the exception of stem cells and the majority of plasma cells. CD20 is not present on plasma cells, most plasmablasts, pro-B cells, and stem cells. BAFF-receptor (BAFF-R) is expressed on both immature and mature B cells in the germinal center, as well as memory B cells and late plasmablasts. Transmembrane activator and CAML interactor (TACI) and B-cell maturation antigen (BCMA) are expressed on germinal center B cells, memory cells, and antibody-secreting cells.

Rituximab

Rituximab is a 1st-generation chimeric monoclonal antibody (IgG1), engineered by fusing a murine Fab with a human Fc domain. Reference Whittam, Tallantyre and Jolles61 Its elimination half-time is estimated at around 20 days; Reference Bar-Or, Calabresi and Arnold76 it may, however, vary according to sex, body weight, and renal function. Reference Ng, Bruno, Combs and Davies77 Rituximab depletes B cells via ADCC and CDC and has been found to be extremely effective in patients with RRMS. A landmark 48-week, phase 2, double-blind, placebo-controlled study convincingly highlighted the efficacy of rituximab monotherapy in reducing gadolinium-enhanced lesions in patients with RRMS (n = 104). Reference Hauser, Arnold and Vollmer78 In addition, a retrospective observational study of 808 patients with RRMS revealed absence of rebound disease activity upon rituximab cessation, Reference Juto, Fink, al Nimer and Piehl79 whereas rebound activity has been reported with natalizumab Reference lo Re, Capobianco and Ragonese80 and fingolimod cessation. Reference Hatcher, Waubant, Nourbakhsh, Crabtree-Hartman and Graves81,Reference Sacco, Emming, Gobbi, Zecca and Monticelli82

In regard to progressive forms, a phase 3 (n = 439 PPMS patients), double-blind and placebo-controlled trial concluded in 2009 that CD20+ B-cell depletion can slow disease progression in a subgroup of younger patients with PPMS, particularly those with inflammatory lesions (GdELs), as rituximab-treated patients had less increase in T2 lesions and confirmed disease progression was delayed in the subgroup with GdELs. Reference Hawker, O’Connor and Freedman83 Overall, however, the study was negative. Moreover, a large observational, retrospective study from the Swedish MS registry included 822 patients (557 RRMS, 198 SPMS, and 67 PPMS) and confirmed both rituximab’s safety as well as its efficacy in reducing GdELs; GdELs went from 26.2% (pretreatment) to 4.6% in the pooled post-treatment cohort. Reference Salzer, Svenningsson and Alping84 Interestingly, disability remained constant in RRMS patients but increased in SPMS and more so in PPMS patients. The question of whether disability progression differs in treated and untreated patients was tackled by a retrospective cohort study of 88 SPMS patients. This study resulted in significantly lower Expanded Disability Status Scale scores (p < 0.001) and delayed disease progression (p = 0.02) in the rituximab-treated group in comparison to the matched control group. Reference Naegelin, Naegelin and von Felten85 It should be noted however that the rituximab-treated group included more patients with radiologic activity, which may have driven the difference between the two groups.

In clinical practice, rituximab is widely used as an off-label treatment for the management of RRMS, as well as active SPMS, as its safety profile is acceptable, well-characterized, Reference Luna, Alping and Burman86,Reference Alping, Askling and Burman87 and the efficacy evident, despite the lack of phase III trials. Reference Yamout, El-Ayoubi, Nicolas, Kouzi, Khoury and Zeineddine88 The drug is generally well-tolerated by patients all throughout the MS type spectrum, and the main adverse effects are mild to moderate infusion-related reactions (IRRs), typically with the first dose, as well as mild to moderate infections. No cases of progressive multifocal leukoencephalopathy (PML) due to John Cunningham virus, which is mostly seen with natalizumab treatment, Reference Ghajarzadeh, Azimi, Valizadeh, Sahraian and Mohammadifar89,Reference Erickson and Garcea90 have been recorded in MS patients treated with rituximab, and the frequency of PML in non-neurologic patients treated with rituximab seems to range around 1:4000; however, usually these patients have received multiple immunosuppressants. Reference Clifford, Ances and Costello91,Reference Kapoor, Mahadeshwar and Hui-Yuen92 Finally, an added advantage of rituximab is its relatively low cost (biosimilars are also available); however, its off-label prescription is complex and time-consuming for physicians. While open questions remain about optimal dosing and frequency strategies, a common tactic is 2 × 500 or 1000 mg, in a 14-day period, and repeat dosing of 500–1000 mg every 6 months or yearly. Reference Whittam, Tallantyre and Jolles61

Ocrelizumab

Ocrelizumab, an IgG1 immunoglobulin, is a 2nd-generation recombinant humanized anti-CD20 mAb. Reference Syed93 The drug has a terminal elimination half-time of around 26 days, which is not affected by mild renal or hepatic impairment. 94 Compared to rituximab, ocrelizumab mobilizes in vitro lower CDC, but higher ADCC action Reference Klein, Lammens and Schäfer95 and as a humanized molecule is expected to be less immunogenic than rituximab with lower titles of neutralizing anti-drug antibodies. Reference Vugmeyster, Beyer and Howell96,Reference Sorensen and Blinkenberg97

In OPERA I (n = 821 patients) and OPERA II (n = 835 patients), two phase 3, double-blind trials published in 2017, ocrelizumab was associated with a lower annualized relapse rate (by 46–47%) and an impressive reduction of the mean number of GdELs (by 94%) over a 96-week time period compared to interferon beta-1a (p < 0.001). The drug effectively depleted CD19 B cells (CD19 B cells serve as index of B-cell count in anti-CD20 treatment) within 2 weeks (which is when CD19 cells were measured). Reference Hauser, Bar-Or and Comi98 ORATORIO, a phase 3, double-blind, placebo-controlled trial, examined ocrelizumab’s efficacy in managing PPMS progression. Results from 732 patients revealed that ocrelizumab was associated with lower rates of clinical and MRI progression than placebo. Because in this study the effect was driven by a fraction of PPMS that had evident MRI inflammation, EMA has approved the drug only in inflammatory PPMS, whereas other agencies such as the FDA and Swissmedic have not applied this restriction. Reference Montalban, Hauser and Kappos99

The most common adverse effects of ocrelizumab include mild and manageable IRRs, like pruritus, rashes, throat irritations, and flushing, but their severity and frequency decrease with the number of infusions. Generally, mild to moderate infections occur in 30% of patients, but severe ones are relatively rare. Other adverse events such as extremity pain, diarrhea, and peripheral edema may also occur in rare cases. Reference Rommer and Zettl100,Reference Rommer, Dudesek, Stüve and Zettl101

Ocrelizumab is administered intravenously according to a fixed dosing schedule, as approved based on the phase 3 studies. An initial dose of 600 mg is divided in 2 × 300 mg with a 2-week time interval. Subsequent doses of 600 mg are given in a single infusion once every 6 months. 94 Interestingly, a post hoc analysis from ORATORIO, where patients with lower body weight (and respectively higher ocrelizumab dose per kg) suffered less progression of deficits, prompted a currently ongoing clinical trial that examines the safety and efficacy of higher than standard ocrelizumab doses (1200 mg for body weights <75 kg, or 1800 mg for body weights >75 kg) in PPMS. Reference Gibiansky, Petry and Mercier102,103

Ofatumumab

Ofatumumab is a 2nd-generation, fully human IgG1 mAb Reference Florou, Katsara, Feehan, Dardiotis and Apostolopoulos104 that depletes circulating CD20 B cells via ADCC Reference Masoud, McAdoo, Bedi, Cairns and Lightstone105 and CDC. 106 Two identically designed, double-blind, phase 3 clinical trials, ASCLEPIOS I and II, compared the efficacy of subcutaneously administered ofatumumab to that of oral teriflunomide, the oral pyrimidine synthesis inhibitor. The trials enrolled 1882 patients in total in 1.6 years, and their results indicated a statistically significant advantage of ofatumumab over teriflunomide in suppressing both new relapses and GdEL activity (the latter by 94–97%). Side effects were reported to be mild to moderate and included injection-related reactions, headache, and infections (in 51.6% of patients treated with ofatumumab) such as nasopharyngitis, upper respiratory, and urinary tract infection. Reference Hauser, Bar-Or and Cohen107 Consequently, the FDA approved ofatumumab as a therapy for RRMS, CIS, and active SPMS in the form of an auto-injector pen, while the EMA for relapsing, active MS. 106 Ofatumumab was approved for subcutaneous use at a dose of 20 mg/0.4 mL once per week for the first 3 weeks of treatment and once monthly thereafter. 108

Ublituximab

Ublituximab is a 3rd-generation anti-CD20 glycoengineered chimeric IgG1 mAb that exerts its action primarily via ADCC, which is facilitated by defucosylation of its Fc region and thereby increased affinity for FcγRIIIa. Reference Whittam, Tallantyre and Jolles61 A 48-week, placebo-controlled, phase 2 trial of ublituximab in 45 RRMS patients established that 150 mg iv on day 1 and 450–600 mg on day 15 and week 24 were able to efficiently deplete B cells within 4 weeks (which is when B cells were measured); moreover, 74% of patients achieved no evidence of disease activity status (NEDA), that is had no relapses, no radiological disease activity, and no progression of disability. Similarly to the other CD20 agents, adverse effects comprised mild to moderate IRRs and upper respiratory infections, influenza, nasopharyngitis, sinusitis, and fungal infections. Reference Fox, Lovett-Racke and Gormley109 In follow-up, two double-blind, phase 3 trials [ULTIMATE I (NCT03277261) and II (NCT03277248)] will assess ublituximab’s efficacy and safety compared to teriflunomide in 880 patients with RRMS. Reference Mealy and Levy110,111

Anti-CD19 mAbs

CD19 belongs to the Ig superfamily and along with CD21, CD82, and CD225 contributes to the formation of a multimolecular signal-transduction complex that ultimately leads to the activation of PI-3 kinase. Reference Tedder112 Compared to CD20, CD19 is expressed on B cells of earlier developmental stages as well as in more antibody-secreting cells and is thus an appealing therapeutic target (Figure 1). Reference Chen, Gallagher, Monson, Herbst and Wang113 In addition to having a broader expression during B-cell stages of development and differentiation, CD19, unlike CD20, is selectively expressed on B cells and not T cells. Reference Schuh, Berer and Mulazzani62 A phase 1 study assessing the pharmacokinetic (intravenous and subcutaneous) profile of inebilizumab, a humanized afucosylated IgG1κ anti-CD19 mAb, Reference Herbst, Wang and Gallagher114 has been conducted in patients with relapsing MS with positive results, 115 but no phase III trials for MS are currently known to be underway.

Atacicept

Atacicept is a human recombinant fusion protein, consisting of a human IgG Fc portion and the extracellular domain of TACI receptor that binds both BAFF and a proliferation-inducing ligand (APRIL). Reference Magliozzi, Marastoni and Calabrese116 The drug therefore competes for BAFF and APRIL binding with native TACI, which is both membrane-bound and soluble, Reference Hoffmann, Kuhn and Laurent117 as well as, to a lesser extent, with the other receptors of the BAFF-APRIL system (BAFF-R and BCMA). Reference Benson, Dillon and Castigli118,Reference Hartung and Kieseier119 After improving rheumatoid arthritis and systemic lupus erythematosus (SLE), Reference van Vollenhoven, Kinnman, Vincent, Wax and Bathon120 atacicept was tried in MS.

Subcutaneous atacicept was evaluated in a 36-week, phase 2, double-blind, and placebo-controlled trial in 255 patients with relapsing MS. The trial was prematurely terminated when an increase in inflammatory disease activity was noticed despite immunoglobulin and naïve B-cell decrease, which led to the suspension of every atacicept trial in MS. Reference Hartung and Kieseier119,Reference Kappos, Hartung and Freedman121,122 Another 36-week, phase 2, double-blind, and placebo-controlled atacicept trial in 34 patients with unilateral optic neuritis as clinical isolated syndrome also showed disease exacerbation, with a significantly higher proportion of patients converting to clinically definite MS compared with placebo. Reference Sergott, Bennett and Rieckmann123 As atacicept effectively depletes naive B cells and induces a transient but marked increase in memory B cells (especially class-switched ones), Reference Hoffmann, Kuhn and Laurent117,Reference Lühder and Gold124,Reference Jelcic, al Nimer and Wang125 possible reasons why atacicept aggravated MS include elimination of regulatory naïve B cells and enhancement of pathogenic memory B-cell function. Reference Baker, Pryce, James, Schmierer and Giovannoni126,Reference Baker, Marta, Pryce, Giovannoni and Schmierer127

Belimumab

Belimumab is a human IgG1λ recombinant monoclonal antibody directed against BAFF that prevents BAFF from interacting with its three receptors on the surface of B cells, thereby reducing B-cell survival, differentiation, and antibody production. Reference Halpern, Lappin and Zanardi128,Reference Dubey, Handu, Dubey, Sharma, Sharma and Ahmed129 Interestingly, belimumab administration does not result in overt immunosuppression. 130 While being moderately effective and FDA-approved for the treatment of SLE since 2011, it failed in myasthenia gravis, Reference Hewett, Sanders and Grove131 a disease mediated by pathogenic autoantibodies. Reference Stohl and Hilbert132 A phase 2, open-label trial of subcutaneous belimumab in addition to ocrelizumab (standard dose) in 40 patients with RRMS was scheduled to start within 2021. 130

Evobrutinib

Evobrutinib is a small molecule drug that binds permanently to and deactivates Bruton’s Tyrosine Kinase (BTK). BTK is an integral part of the BCR signaling cascade that affects B-cell activation and is essential for B-cell maturation and their ultimate, terminal differentiation into memory or plasma cells. Of interest, BTK is involved in the entry of B cells into follicular structures. Knockout or absence of BTK results in lack of B-cell activation, moreover almost complete lack of peripheral B and plasma cells and low circulating immunoglobulin. Reference Dingjan, Middendorp, Dahlenborg, Maas, Grosveld and Hendriks133Reference Torke and Weber136 Importantly, about 75% of the CNS cells that express BTK are microglial, while BTK expression levels in the brain increase after demyelination. Reference Martin, Aigrot and Grenningloh137 As evobrutinib can bypass the BBB and enter the CNS, it can affect microglial cells and B cells within the CNS.

Evobrutinib was the first BTK inhibitor (BTKI) to be tested as a monotherapy in relapsing MS. Reference Becker, Martin and Mitchell138 In a double-blind, phase II trial (n = 267), evobrutinib was tested against placebo and dimethyl fumarate. The results showed that patients who received 75 mg of daily evobrutinib had significantly fewer GdELs during weeks 12 through 24 than those who received placebo (1.69 ± 4.69 against 3.85 ± 5.44, p = 0.005), while adverse effects were minimal (e.g. nasopharyngitis, alanine aminotransferase, and aspartate aminotransferase level elevation). 139,Reference Montalban, Arnold and Weber140 Evobrutinib is now being advanced to phase III evaluation, along with several other BTKI (some of them with reversible BTK binding); fenebrutinib, ibrutinib, and tolebrutinib. 141

While CD19/20 B-cell depletion has shown tremendous efficacy in reducing clinical and radiological MS activity, it raises several safety concerns on humoral deficiency with long-term usage in addition to a reduced response to vaccination. Reference Luna, Alping and Burman86,Reference Nazi, Kelton and Larché142,Reference Bar-Or, Calkwood and Chognot143 These disadvantages could possibly be avoided with inhibition of B-cell activation and maturation with small molecules such as BTKIs. Reference Torke and Weber136,Reference Dolgin144 In contrast with antibody-based B-cell depletion, BTKIs do not destroy or lastingly minimize the frequency of peripheral B cells, but seem to prevent the development of pathogenic B cells. Reference Torke, Pretzsch and Häusler145 Their effect on disease activity does not seem to be as impressive as that of anti-B-cell antibodies, and they cannot control the pathogenic properties of B cells as rapidly; however, they are smaller in size, can penetrate the CNS, target microglia, and might therefore have a better effect on disability progression. Reference Boschert, Crandall and Pereira146

B-Cell-Targeted Therapies and Pregnancy

As MS largely affects female patients with childbearing potential, the utilization of B-cell-targeted therapies in women of childbearing age deserves special mention. Reference Tisovic and Amezcua147,Reference Wallin, Culpepper and Campbell148 Rituximab-associated B-cell depletion persists long after the drug’s elimination, which occurs approximately 3 months after the last infusion. Thus, conception can be considered safe 3 months after the last infusion without significant risk of fetal exposure. But even if a woman conceives before rituximab’s effective elimination, IgG1 subclass mAbs cannot cross the placenta barrier during the first trimester, resulting in low chance of fetal exposure. Reference Das, Damotte and Gelfand149 Importantly, rituximab administration and concurrent B-cell depletion have not been linked to increased risk of adverse pregnancy outcomes compared with the expected incidence in population. Reference Smith, Hellwig, Fink, Lyell, Piehl and Langer-Gould150 Also, infants breastfed under anti-CD20 treatment had normal B-cell counts, and no negative impact on health and development was attributed to breastfeeding in the 1-year follow-up period. Reference Ciplea, Langer-Gould and de Vries151 Although data regarding ocrelizumab administration in this population group are limited, it is reasonable to apply the same principles as with rituximab. One additional advantage of CD20 depletion in terms of family planning is that discontinuation of therapy is not associated with a rebound phenomenon, as has been observed with natalizumab. In that regard, a cohort study regarding the safety of anti-C20 mAbs rituximab and ocrelizumab during the last 12 months before or during pregnancy concluded that the drugs are effective in controlling disease in women with RRMS, during and partly after pregnancy. However, B-cell monitoring is essential both for the newborn and for the mother after delivery, and larger studies are required to assess their safety profile and to establish the best time to restart the therapy after delivery. Reference Kümpfel, Thiel and Meinl152 Recent recommendations suggest prioritization of MS management and conception postponement in cases of highly active MS and contraception for up to 4 months after ocrelizumab administration.Reference Wiendl, Gold and Berger 153

Conclusion

The therapeutic criterion underlines that B cells not only participate in the pathogenesis of MS but can act as the orchestrators of the inflammatory processes. As shown by clinical trials and real-world data, B-cell-targeting agents (in particular CD20-depleting agents) have established a new era in MS therapeutics and immunotherapy in general, considering their remarkable efficacy and safety profile. Long-term safety, especially increased risk of infection with slowly but gradually decreasing total serum immunoglobulin levels, remains a significant concern that has a limiting effect on anti-CD20 usage in clinical practice. Regular monitoring of immunoglobulin levels (e.g. before each follow-up infusion) can help timely detection of a decrease and lowers the risk of infection due to associated immunodeficiency. Reference Keystone, Fleischmann and Emery154Reference van Vollenhoven, Emery and Bingham156 Future studies will further inform on long-term effects of CD20-targeting medications, on the use of oral BTKI agents and determine the new therapeutic algorithm that will likely move more towards induction rather than escalation.

Acknowledgements

Figure was created with BioRender.com. The publication of the article in OA mode was financially supported by HEAL-Link.

Funding

PK, IS, and LS received no funding in relation to the present topic. PS is supported by the Onassis Foundation.

Conflict of Interest

PK and IS declare no conflicts of interest. LS is the site investigator in the trials MUSETTE (BN42082) and GAVOTTE (BN42083), sponsored by F. Hoffmann La-Roche Ltd. PS has received a travel grant from Sanofi and research funding by the Onassis Foundation.

Statement of Authorship

Conceptualization: PS.

Drafting: PS, PK, IS.

Editing: PS, PK, IS, LS.

References

Yeshokumar, AK, Narula, S, Banwell, B. Pediatric multiple sclerosis. Curr Opin Neurol. 2017;30:21621. DOI 10.1097/WCO.0000000000000452.CrossRefGoogle ScholarPubMed
Thompson, AJ, Baranzini, SE, Geurts, J, Hemmer, B, Ciccarelli, O. Multiple sclerosis. Lancet. 2018;391:162236. DOI 10.1016/S0140-6736(18)30481-1.CrossRefGoogle ScholarPubMed
Filippi, M, Bar-Or, A, Piehl, F, et al. Multiple sclerosis. Nat Rev Dis Primers. 2018;4:43. DOI 10.1038/s41572-018-0041-4.CrossRefGoogle ScholarPubMed
Lublin, FD. New multiple sclerosis phenotypic classification. Eur Neurol. 2014;72 Suppl 1:15. DOI 10.1159/000367614.CrossRefGoogle Scholar
International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science (1979). 2019;365:eaav7188. DOI 10.1126/science.aav7188.Google Scholar
O’Connor, KC, Bar-Or, A, Hafler, DA. The neuroimmunology of multiple sclerosis: possible roles of T and B lymphocytes in immunopathogenesis. J Clin Immunol. 2001;21:8192. DOI 10.1023/a:1011064007686.CrossRefGoogle Scholar
De Jager, PL, Jia, X, Wang, J, et al. Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet. 2009;41:77682. DOI 10.1038/ng.401.CrossRefGoogle ScholarPubMed
Olsson, T, Barcellos, LF, Alfredsson, L. Interactions between genetic, lifestyle and environmental risk factors for multiple sclerosis. Nat Rev Neurol. 2017;13:2536. DOI 10.1038/nrneurol.2016.187.CrossRefGoogle ScholarPubMed
Cotsapas, C, Mitrovic, M. Genome-wide association studies of multiple sclerosis. Clin Transl Immunol. 2018;7:e1018. DOI 10.1002/cti2.1018.CrossRefGoogle ScholarPubMed
Beecham, AH, Patsopoulos, NA, Xifara, DK, et al. Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet. 2013;45:135360. DOI 10.1038/ng.2770.Google ScholarPubMed
Mokry, LE, Ross, S, Ahmad, OS, et al. Vitamin D and risk of multiple sclerosis: a mendelian randomization study. PLoS Med. 2015;12:e1001866. DOI 10.1371/journal.pmed.1001866.CrossRefGoogle ScholarPubMed
Ota, K, Matsui, M, Milford, EL, Mackin, GA, Weiner, HL, Hafler, DA. T-Cell recognition of an immuno-dominant myelin basic protein epitope in multiple sclerosis. Nature. 1990;346:1837. DOI 10.1038/346183a0.CrossRefGoogle Scholar
Bjartmar, C, Yin, X, Trapp, BD. Axonal pathology in myelin disorders. J Neurocytol. 1999;28:38395.CrossRefGoogle ScholarPubMed
Bonnan, M. Intrathecal IgG synthesis: a resistant and valuable target for future multiple sclerosis treatments. Mult Scler Int. 2015;2015:296184. DOI 10.1155/2015/296184.Google ScholarPubMed
Cepok, S, Rosche, B, Grummel, V, et al. Short-lived plasma blasts are the main B cell effector subset during the course of multiple sclerosis. Brain. 2005;128:166776. DOI 10.1093/brain/awh486.CrossRefGoogle Scholar
Kowarik, MC, Cepok, S, Sellner, J, et al. CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation. J Neuroinflammation. 2012;9:93.CrossRefGoogle Scholar
Krumbholz, M, Derfuss, T, Hohlfeld, R, Meinl, E. B cells and antibodies in multiple sclerosis pathogenesis and therapy. Nat Rev Neurol. 2012;8:61323. DOI 10.1038/nrneurol.2012.203.CrossRefGoogle ScholarPubMed
Kivisäkk, P, Imitola, J, Rasmussen, S, et al. Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann Neurol. 2009;65:45769. DOI 10.1002/ana.21379.CrossRefGoogle ScholarPubMed
Moreno Torres, I, García-Merino, A. Anti-CD20 monoclonal antibodies in multiple sclerosis. Expert Rev Neurother. 2017;17:35971. DOI 10.1080/14737175.2017.1245616.CrossRefGoogle ScholarPubMed
Lassmann, H. Pathogenic mechanisms associated with different clinical courses of multiple sclerosis. Front Immunol. 2018;9:3116. DOI 10.3389/fimmu.2018.03116.CrossRefGoogle ScholarPubMed
Gh Popescu, BF, Pirko, I, Lucchinetti, CF. Pathology of multiple sclerosis: where do we stand? Continuum (Minneap Minn). 2013;19:90121.Google Scholar
Barnett, MH, Parratt, JDE, Cho, ES, Prineas, JW. Immunoglobulins and complement in postmortem multiple sclerosis tissue. Ann Neurol. 2009;65:3246. DOI 10.1002/ana.21524.CrossRefGoogle ScholarPubMed
Keegan, M, König, F, McClelland, R, et al. Relation between humoral pathological changes in multiple sclerosis and response to therapeutic plasma exchange. Lancet. 2005;366:57982. DOI 10.1016/S0140-6736(05)67102-4.CrossRefGoogle ScholarPubMed
Gasperi, C, Stüve, O, Hemmer, B. B cell-directed therapies in multiple sclerosis. Neurodegener Dis Manag. 2016;6:3747. DOI 10.2217/nmt.15.67.CrossRefGoogle ScholarPubMed
Häusser-Kinzel, S, Weber, MS. The role of B cells and antibodies in multiple sclerosis, neuromyelitis optica, and related disorders. Front Immunol. 2019;10:201. DOI 10.3389/fimmu.2019.00201.CrossRefGoogle ScholarPubMed
Guan, Y, Jakimovski, D, Ramanathan, M, Weinstock-Guttman, B, Zivadinov, R. The role of Epstein-Barr virus in multiple sclerosis: from molecular pathophysiology to in vivo imaging. Neural Regen Res. 2019;14:37386.Google ScholarPubMed
Abdelhak, A, Hottenrott, T, Mayer, C, et al. CSF profile in primary progressive multiple sclerosis: re-exploring the basics. PLOS ONE. 2017;12:e0182647.CrossRefGoogle ScholarPubMed
Thouvenot, E. Multiple sclerosis biomarkers: helping the diagnosis? Rev Neurol (Paris). 2018;174:36471. DOI 10.1016/j.neurol.2018.04.002.CrossRefGoogle ScholarPubMed
Brändle, SM, Obermeier, B, Senel, M, et al. Distinct oligoclonal band antibodies in multiple sclerosis recognize ubiquitous self-proteins. Proc Natl Acad Sci U S A. 2016;113:78649. DOI 10.1073/pnas.1522730113.CrossRefGoogle ScholarPubMed
Willis, SN, Stathopoulos, P, Chastre, A, Compton, SD, Hafler, DA, O’Connor, KC. Investigating the antigen specificity of multiple sclerosis central nervous system-derived immunoglobulins. Front Immunol. 2015;6:600. DOI 10.3389/fimmu.2015.00600.CrossRefGoogle ScholarPubMed
Villar, LM, Masjuan, J, González-Porqué, P, et al. Intrathecal IgM synthesis predicts the onset of new relapses and a worse disease course in MS. Neurology. 2002;59:5559. DOI 10.1212/wnl.59.4.555.CrossRefGoogle Scholar
Boronat, A, Sepúlveda, M, Llufriu, S, et al. Analysis of antibodies to surface epitopes of contactin-2 in multiple sclerosis. J Neuroimmunol. 2012;244:1036. DOI 10.1016/j.jneuroim.2011.12.023.CrossRefGoogle ScholarPubMed
Gerhards, R, Pfeffer, LK, Lorenz, J, et al. Oligodendrocyte myelin glycoprotein as a novel target for pathogenic autoimmunity in the CNS. Acta Neuropathol Commun. 2020;8:207. DOI 10.1186/s40478-020-01086-2.CrossRefGoogle ScholarPubMed
Yeste, A, Quintana, FJ. Antigen microarrays for the study of autoimmune diseases. Clin Chem. 2013;59:103644. DOI 10.1373/clinchem.2012.194423.CrossRefGoogle Scholar
Kanter, JL, Narayana, S, Ho, PP, et al. Lipid microarrays identify key mediators of autoimmune brain inflammation. Nat Med. 2006;12:13843. DOI 10.1038/nm1344.CrossRefGoogle ScholarPubMed
Trotter, J, DeJong, LJ, Smith, ME. Opsonization with antimyelin antibody increases the uptake and intracellular metabolism of myelin in inflammatory macrophages. J Neurochem. 1986;47:77989. DOI 10.1111/j.1471-4159.1986.tb00679.x.Google ScholarPubMed
Flach, AC, Litke, T, Strauss, J, et al. Autoantibody-boosted T-cell reactivation in the target organ triggers manifestation of autoimmune CNS disease. Proc Natl Acad Sci U S A. 2016;113:33238. DOI 10.1073/pnas.1519608113.CrossRefGoogle ScholarPubMed
Getahun, A, Dahlström, J, Wernersson, S, Heyman, B. IgG2a-mediated enhancement of antibody and T cell responses and its relation to inhibitory and activating Fc gamma receptors. J Immunol. 2004;172:526976. DOI 10.4049/jimmunol.172.9.5269.CrossRefGoogle Scholar
Kinzel, S, Weber, MS. B cell-directed therapeutics in multiple sclerosis: rationale and clinical evidence. CNS Drugs. 2016;30:113748. DOI 10.1007/s40263-016-0396-6.CrossRefGoogle ScholarPubMed
Ancau, M, Berthele, A, Hemmer, B. CD20 monoclonal antibodies for the treatment of multiple sclerosis: up-to-date. Expert Opin Biol Ther. 2019;19:82943. DOI 10.1080/14712598.2019.1611778.CrossRefGoogle ScholarPubMed
Wang, J, Jelcic, I, Mühlenbruch, L, et al. HLA-DR15 molecules jointly shape an autoreactive T cell repertoire in multiple sclerosis. Cell. 2020;183:12641281.e20. DOI 10.1016/j.cell.2020.09.054.CrossRefGoogle ScholarPubMed
Bettelli, E, Carrier, Y, Gao, W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:2358. DOI 10.1038/nature04753.CrossRefGoogle ScholarPubMed
Molnarfi, N, Schulze-Topphoff, U, Weber, MS, et al. MHC class II-dependent B cell APC function is required for induction of CNS autoimmunity independent of myelin-specific antibodies. J Exp Med. 2013;210:292137. DOI 10.1084/jem.20130699.CrossRefGoogle ScholarPubMed
Korn, T, Mitsdoerffer, M, Croxford, AL, et al. IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells. Proc Natl Acad Sci U S A. 2008;105:184605. DOI 10.1073/pnas.0809850105.CrossRefGoogle ScholarPubMed
Li, R, Rezk, A, Miyazaki, Y, et al. Proinflammatory GM-CSF-producing B cells in multiple sclerosis and B cell depletion therapy. Sci Transl Med. 2015;7:310ra166.CrossRefGoogle ScholarPubMed
Shen, P, Roch, T, Lampropoulou, V, et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature. 2014;507:36670. DOI 10.1038/nature12979.CrossRefGoogle ScholarPubMed
Arneth, BM. Impact of B cells to the pathophysiology of multiple sclerosis. J Neuroinflammation. 2019;16:128. DOI 10.1186/s12974-019-1517-1.CrossRefGoogle Scholar
Morandi, E, Jagessar, SA, ‘t Hart, BA, Gran, B. EBV infection empowers human B cells for autoimmunity: role of autophagy and relevance to multiple sclerosis. J Immunol. 2017;199:43548. DOI 10.4049/jimmunol.1700178.CrossRefGoogle Scholar
Kjetil, B, Marianna, C, H.B., C, et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science (1979). 2022;375:296301. DOI 10.1126/science.abj8222.Google Scholar
Lang, HLE, Jacobsen, H, Ikemizu, S, et al. A functional and structural basis for TCR cross-reactivity in multiple sclerosis. Nat Immunol. 2002;3:9403. DOI 10.1038/ni835.CrossRefGoogle ScholarPubMed
Angelini, DF, Serafini, B, Piras, E, et al. Increased CD8+ T cell response to Epstein-Barr virus lytic antigens in the active phase of multiple sclerosis. PLoS Pathog. 2013;9:e1003220.CrossRefGoogle ScholarPubMed
Pender, MP, Burrows, SR. Epstein-Barr virus and multiple sclerosis: potential opportunities for immunotherapy. Clin Transl Immunol. 2014;3:e27.CrossRefGoogle ScholarPubMed
Pender, MP. Infection of autoreactive B lymphocytes with EBV, causing chronic autoimmune diseases. Trends Immunol. 2003;24:5848. DOI 10.1016/j.it.2003.09.005.CrossRefGoogle ScholarPubMed
Lanz, Tv, Brewer, RC, Ho, PP, et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature. 2022;603:3217. DOI 10.1038/s41586-022-04432-7.CrossRefGoogle ScholarPubMed
McCarthy, NJ, Hazlewood, SA, Huen, DS, Rickinson, AB, Williams, GT. The Epstein-Barr virus gene BHRF1, a homologue of the cellular oncogene Bcl-2, inhibits apoptosis induced by gamma radiation and chemotherapeutic drugs. Adv Exp Med Biol. 1996;406:8397. DOI 10.1007/978-1-4899-0274-0_9.CrossRefGoogle ScholarPubMed
Serafini, B, Rosicarelli, B, Franciotta, D, et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J Exp Med. 2007;204:2899912. DOI 10.1084/jem.20071030.CrossRefGoogle ScholarPubMed
Sargsyan, SA, Shearer, AJ, Ritchie, AM, et al. Absence of Epstein-Barr virus in the brain and CSF of patients with multiple sclerosis. Neurology. 2010;74:112735. DOI 10.1212/WNL.0b013e3181d865a1.CrossRefGoogle ScholarPubMed
Peferoen, LAN, Lamers, F, Lodder, LNR, et al. Epstein Barr virus is not a characteristic feature in the central nervous system in established multiple sclerosis. Brain. 2010;133:e137.CrossRefGoogle Scholar
Willis, SN, Stadelmann, C, Rodig, SJ, et al. Epstein-Barr virus infection is not a characteristic feature of multiple sclerosis brain. Brain. 2009;132:331828. DOI 10.1093/brain/awp200.CrossRefGoogle Scholar
Torkildsen, Ø., Stansberg, C, Angelskår, SM, et al. Upregulation of immunoglobulin-related genes in cortical sections from multiple sclerosis patients. Brain Pathol. 2010;20:720–9. DOI 10.1111/j.1750-3639.2009.00343.x.Google ScholarPubMed
Whittam, DH, Tallantyre, EC, Jolles, S, et al. Rituximab in neurological disease: principles, evidence and practice. Pract Neurol. 2019;19:5. DOI 10.1136/practneurol-2018-001899.CrossRefGoogle ScholarPubMed
Schuh, E, Berer, K, Mulazzani, M, et al. Features of human CD3+CD20+ T cells. J Immunol. 2016;197:11117. DOI 10.4049/jimmunol.1600089.CrossRefGoogle ScholarPubMed
Palanichamy, A, Jahn, S, Nickles, D, et al. Rituximab efficiently depletes increased CD20-expressing T cells in multiple sclerosis patients. J Immunol. 2014;193:580–6. DOI 10.4049/jimmunol.1400118.CrossRefGoogle ScholarPubMed
Sabatino, JJ Jr, Wilson, MR, Calabresi, PA, Hauser, SL, Schneck, JP, Zamvil, SS. Anti-CD20 therapy depletes activated myelin-specific CD8(+) T cells in multiple sclerosis. Proc Natl Acad Sci U S A. 2019;116:258007. DOI 10.1073/pnas.1915309116.CrossRefGoogle ScholarPubMed
von Essen, MR, Ammitzbøll, C, Hansen, RH, et al. Proinflammatory CD20+ T cells in the pathogenesis of multiple sclerosis. Brain. 2019;142:12032. DOI 10.1093/brain/awy301.CrossRefGoogle ScholarPubMed
Payandeh, Z, Bahrami, AA, Hoseinpoor, R, et al. The applications of anti-CD20 antibodies to treat various B cells disorders. Biomed Pharmacother. 2019;109:241526. DOI 10.1016/j.biopha.2018.11.121.CrossRefGoogle ScholarPubMed
Dalakas, MC. B cells in the pathophysiology of autoimmune neurological disorders: a credible therapeutic target. Pharmacol Ther. 2006;112:5770. DOI 10.1016/j.pharmthera.2006.03.005.CrossRefGoogle ScholarPubMed
Tedder, TF, Streuli, M, Schlossman, SF, Saito, H. Isolation and structure of a CDNA encoding the B1 (CD20) cell-surface antigen of human B lymphocytes. Proc Natl Acad Sci U S A. 1988;85:20812. DOI 10.1073/pnas.85.1.208.CrossRefGoogle ScholarPubMed
Santos, MAO, Lima, MM. CD20 role in pathophysiology of Hodgkin’s disease. Rev Assoc Med Bras (1992). 2017;63:8103. DOI 10.1590/1806-9282.63.09.810.CrossRefGoogle ScholarPubMed
Maloney, DG. Anti-CD20 antibody therapy for B-cell lymphomas. N Engl J Med. 2012;366:200816. DOI 10.1056/NEJMct1114348.CrossRefGoogle ScholarPubMed
Schroder, C, Azimzadeh, AM, Wu, G, Price, JO, Atkinson, JB, Pierson, RN. Anti-CD20 treatment depletes B-cells in blood and lymphatic tissue of cynomolgus monkeys. Transpl Immunol. 2003;12:1928.CrossRefGoogle ScholarPubMed
Piccio, L, Naismith, RT, Trinkaus, K, et al. Changes in B- and T-lymphocyte and chemokine levels with rituximab treatment in multiple sclerosis. Arch Neurol. 2010;67:70714. DOI 10.1001/archneurol.2010.99.CrossRefGoogle Scholar
Ramwadhdoebe, TH, van Baarsen, LGM, Boumans, MJH, et al. Effect of rituximab treatment on T and B cell subsets in lymph node biopsies of patients with rheumatoid arthritis. Rheumatology (Oxford), 58:107585. DOI 10.1093/rheumatology/key428.Google Scholar
Petereit, HF, Rubbert-Roth, A. Rituximab levels in cerebrospinal fluid of patients with neurological autoimmune disorders. Mult Scler J. 2008;15:18992. DOI 10.1177/1352458508098268.Google ScholarPubMed
Monson, NL, Cravens, PD, Frohman, EM, Hawker, K, Racke, MK. Effect of rituximab on the peripheral blood and cerebrospinal fluid B cells in patients with primary progressive multiple sclerosis. Arch Neurol. 2005;62:25864. DOI 10.1001/archneur.62.2.258.Google ScholarPubMed
Bar-Or, A, Calabresi, PAJ, Arnold, D, et al. Rituximab in relapsing-remitting multiple sclerosis: a 72-week, open-label, phase I trial. Ann Neurol. 2008;63:395400. DOI 10.1002/ana.21363.CrossRefGoogle ScholarPubMed
Ng, CM, Bruno, R, Combs, D, Davies, B. Population pharmacokinetics of rituximab (anti-CD20 monoclonal antibody) in rheumatoid arthritis patients during a phase II clinical trial. J Clin Pharmacol. 2005;45:792801. DOI 10.1177/0091270005277075.CrossRefGoogle ScholarPubMed
Hauser, SL, Arnold, DL, Vollmer, T, et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358:67688.CrossRefGoogle ScholarPubMed
Juto, A, Fink, K, al Nimer, F, Piehl, F. Interrupting rituximab treatment in relapsing-remitting multiple sclerosis; no evidence of rebound disease activity. Mult Scler Relat Dis. 2020;37:101468. DOI 10.1016/j.msard.2019.101468.CrossRefGoogle ScholarPubMed
lo Re, M, Capobianco, M, Ragonese, P, et al. Natalizumab discontinuation and treatment strategies in patients with multiple sclerosis (MS): a retrospective study from two Italian MS centers. Neurol Ther. 2015;4:14757. DOI 10.1007/s40120-015-0038-9.CrossRefGoogle ScholarPubMed
Hatcher, SE, Waubant, E, Nourbakhsh, B, Crabtree-Hartman, E, Graves, JS. Rebound syndrome in patients with multiple sclerosis after cessation of fingolimod treatment. JAMA Neurol. 2016;73:7904. DOI 10.1001/jamaneurol.2016.0826.Google ScholarPubMed
Sacco, R, Emming, S, Gobbi, C, Zecca, C, Monticelli, S. Rebound of disease activity after fingolimod withdrawal: immunological and gene expression profiling. Mult Scler Relat Dis. 2020;40:101927. DOI 10.1016/j.msard.2020.101927.CrossRefGoogle ScholarPubMed
Hawker, K, O’Connor, P, Freedman, MS, et al. Rituximab in patients with primary progressive multiple sclerosis: results of a randomized double-blind placebo-controlled multicenter trial. Ann Neurol. 2009;66:46071. DOI 10.1002/ana.21867.CrossRefGoogle ScholarPubMed
Salzer, J, Svenningsson, R, Alping, P, et al. Rituximab in multiple sclerosis. Neurology. 2016;87:207481. DOI 10.1212/WNL.0000000000003331.CrossRefGoogle ScholarPubMed
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;76:27481. DOI 10.1001/jamaneurol.2018.4239.CrossRefGoogle ScholarPubMed
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;77:18491. DOI 10.1001/jamaneurol.2019.3365.CrossRefGoogle ScholarPubMed
Alping, P, Askling, J, Burman, J, et al. Cancer risk for fingolimod, natalizumab, and rituximab in multiple sclerosis patients. Ann Neurol. 2020;87:68899. DOI 10.1002/ana.25701.CrossRefGoogle ScholarPubMed
Yamout, BI, El-Ayoubi, NK, Nicolas, J, Kouzi, Yel, Khoury, SJ, Zeineddine, MM. Safety and efficacy of rituximab in multiple sclerosis: a retrospective observational study. J Immunol Res. 2018;2018:19. DOI 10.1155/2018/9084759.CrossRefGoogle ScholarPubMed
Ghajarzadeh, M, Azimi, A, Valizadeh, Z, Sahraian, MA, Mohammadifar, M. Efficacy and safety of rituximab in treating patients with multiple sclerosis (MS): a systematic review and meta-analysis. Autoimmun Rev. 2020;19:102585. DOI 10.1016/j.autrev.2020.102585.CrossRefGoogle ScholarPubMed
Erickson, KD, Garcea, RL. Viral replication centers and the DNA damage response in JC virus-infected cells. Virology. 2019;528:198206. DOI 10.1016/j.virol.2018.12.014.CrossRefGoogle ScholarPubMed
Clifford, DB, Ances, B, Costello, C, et al. Rituximab-associated progressive multifocal leukoencephalopathy in rheumatoid arthritis. Arch Neurol. 2011;68:115664. DOI 10.1001/archneurol.2011.103.CrossRefGoogle ScholarPubMed
Kapoor, T, Mahadeshwar, P, Hui-Yuen, J, et al. Prevalence of progressive multifocal leukoencephalopathy (PML) in adults and children with systemic lupus erythematosus. Lupus Sci Med. 2020;7:e000388. DOI 10.1136/lupus-2020-000388.CrossRefGoogle ScholarPubMed
Syed, YY. Ocrelizumab: a review in multiple sclerosis. CNS Drugs. 2018;32:88390.CrossRefGoogle ScholarPubMed
Ocrevus | European Medicines Agency.Available at: https://www.ema.europa.eu/en/medicines/human/EPAR/ocrevus; accessed December 1, 2021.Google Scholar
Klein, C, Lammens, A, Schäfer, W, et al. Epitope interactions of monoclonal antibodies targeting CD20 and their relationship to functional properties. MAbs. 2013;5:2233. DOI 10.4161/mabs.22771.CrossRefGoogle ScholarPubMed
Vugmeyster, Y, Beyer, J, Howell, K, et al. Depletion of B cells by a humanized anti-CD20 antibody PRO70769 in macaca fascicularis. J Immunother. 2005;28:2129. DOI 10.1097/01.cji.0000155050.03916.04.CrossRefGoogle ScholarPubMed
Sorensen, PS, Blinkenberg, M. The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects. Ther Adv Neurol Disord. 2016;9:4452. DOI 10.1177/1756285615601933.CrossRefGoogle ScholarPubMed
Hauser, SL, Bar-Or, A, Comi, G, et al. Ocrelizumab versus interferon beta-1a in relapsing multiple sclerosis. N Engl J Med. 2017;376:22134. DOI 10.1056/NEJMoa1601277.CrossRefGoogle ScholarPubMed
Montalban, X, Hauser, SL, Kappos, L, et al. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. 2017;376:20920. DOI 10.1056/NEJMoa1606468.CrossRefGoogle ScholarPubMed
Rommer, PS, Zettl, UK. Managing the side effects of multiple sclerosis therapy: pharmacotherapy options for patients. Expert Opin Pharmacother. 2018;19:48398.CrossRefGoogle ScholarPubMed
Rommer, PS, Dudesek, A, Stüve, O, Zettl, UK. Monoclonal antibodies in treatment of multiple sclerosis. Clin Exp Immunol. 2014;175:37384. DOI 10.1111/cei.12197.CrossRefGoogle ScholarPubMed
Gibiansky, E, Petry, C, Mercier, F, et al. Ocrelizumab in relapsing and primary progressive multiple sclerosis: pharmacokinetic and pharmacodynamic analyses of OPERA I, OPERA II and ORATORIO. Br J Clin Pharmacol. 2021;87:251120. DOI 10.1111/bcp.14658.CrossRefGoogle ScholarPubMed
A study to evaluate the efficacy, safety and pharmacokinetics of a higher dose of ocrelizumab in adults with primary progressive multiple sclerosis (PPMS). ClinicalTrials. Gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04548999; accessed May 18, 2021.Google Scholar
Florou, D, Katsara, M, Feehan, J, Dardiotis, E, Apostolopoulos, V. Anti-CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatumumab. Brain Sci. 2020;10:758. DOI 10.3390/brainsci10100758.CrossRefGoogle ScholarPubMed
Masoud, S, McAdoo, SP, Bedi, R, Cairns, TD, Lightstone, L. Ofatumumab for B cell depletion in patients with systemic lupus erythematosus who are allergic to rituximab. Rheumatology (Oxford). 2018;57:115661. DOI 10.1093/rheumatology/key042.CrossRefGoogle ScholarPubMed
Ofatumumab. Am J Health Syst Pharm. 2020;77:20258. DOI 10.1093/ajhp/zxaa322.CrossRefGoogle Scholar
Hauser, SL, Bar-Or, A, Cohen, JA, et al. Ofatumumab versus teriflunomide in multiple sclerosis. N Engl J Med. 2020;383:54657. DOI 10.1056/NEJMoa1917246.CrossRefGoogle ScholarPubMed
Kesimpta (Ofatumumab SC) dosing, indications, interactions, adverse effects, and more. Available at: https://reference.medscape.com/drug/kesimpta-ofatumumab-sc-4000083#0; accessed January 23, 2017.Google Scholar
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. 2021;27:4209. DOI 10.1177/1352458520918375.CrossRefGoogle ScholarPubMed
Mealy, MA, Levy, M. A pilot safety study of ublituximab, a monoclonal antibody against CD20, in acute relapses of neuromyelitis optica spectrum disorder. Medicine. 2019;98:e15944. DOI 10.1097/MD.0000000000015944.CrossRefGoogle ScholarPubMed
Study to Assess the Efficacy and Safety of Ublituximab in Participants With Relapsing Forms of Multiple Sclerosis (RMS) (ULTIMATE II) (NCT03277248). Available online: https://clinicaltrials.gov/ct2/show/NCT03277248 Google Scholar
Tedder, TF. CD19: a promising B cell target for rheumatoid arthritis. Nat Rev Rheumatol. 2009;5:5727.CrossRefGoogle ScholarPubMed
Chen, D, Gallagher, S, Monson, NL, Herbst, R, Wang, Y. Inebilizumab, a B cell-depleting anti-CD19 antibody for the treatment of autoimmune neurological diseases: insights from preclinical studies. J Clin Med. 2016;5:107. DOI 10.3390/jcm5120107.CrossRefGoogle Scholar
Herbst, R, Wang, Y, Gallagher, S, et al. B-cell depletion in vitro and in vivo with an afucosylated anti-CD19 antibody. J Pharmacol Exp Ther. 2010;335:21322. DOI 10.1124/jpet.110.168062.CrossRefGoogle ScholarPubMed
Safety and tolerability study of MEDI-551, a B-cell depleting agent, to treat relapsing forms of multiple sclerosis.Available at: https://clinicaltrials.gov/ct2/show/NCT01585766 Google Scholar
Magliozzi, R, Marastoni, D, Calabrese, M. The BAFF/APRIL system as therapeutic target in multiple sclerosis. Expert Opin Ther Targets. 2020;24:113545. DOI 10.1080/14728222.2020.1821647.CrossRefGoogle ScholarPubMed
Hoffmann, FS, Kuhn, P-H, Laurent, SA, et al. The immunoregulator soluble TACI is released by ADAM10 and reflects B cell activation in autoimmunity. J Immunol. 2015;194:54252. DOI 10.4049/jimmunol.1402070.CrossRefGoogle ScholarPubMed
Benson, MJ, Dillon, SR, Castigli, E, et al. Cutting edge: the dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol. 2008;180:36559. DOI 10.4049/jimmunol.180.6.3655.CrossRefGoogle ScholarPubMed
Hartung, H-P, Kieseier, BC. Atacicept: targeting B cells in multiple sclerosis. Ther Adv Neurol Disord. 2010;3:20516. DOI 10.1177/1756285610371146.CrossRefGoogle ScholarPubMed
van Vollenhoven, RF, Kinnman, N, Vincent, E, Wax, S, Bathon, J. Atacicept in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase II, randomized, placebo-controlled trial. Arthritis Rheum. 2011;63:178292. DOI 10.1002/art.30372.CrossRefGoogle Scholar
Kappos, L, Hartung, H-P, Freedman, MS, et al. Atacicept in Multiple Sclerosis (ATAMS): a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Neurol. 2014;13:35363. DOI 10.1016/S1474-4422(14)70028-6.CrossRefGoogle ScholarPubMed
A phase 2 study of atacicept in subjects with relapsing multiple sclerosis (ATAMS). Available at: https://clinicaltrials.gov/ct2/show/NCT00642902 Google Scholar
Sergott, RC, Bennett, JL, Rieckmann, P, et al. Results from a phase II randomized trial of the B-cell-targeting agent atacicept in patients with optic neuritis. J Neurol Sci. 2015;351:1748. DOI 10.1016/j.jns.2015.02.019.CrossRefGoogle ScholarPubMed
Lühder, F, Gold, R. Trial and error in clinical studies: lessons from ATAMS. Lancet Neurol. 2014;13:3401. DOI 10.1016/S1474-4422(14)70050-X.CrossRefGoogle ScholarPubMed
Jelcic, I, al Nimer, F, Wang, J, et al. Autoreactive CD4+ T cells in multiple sclerosis. Cell. 2018;175:85100.e23. DOI 10.1016/j.cell.2018.08.011.CrossRefGoogle ScholarPubMed
Baker, D, Pryce, G, James, LK, Schmierer, K, Giovannoni, G. Failed B cell survival factor trials support the importance of memory B cells in multiple sclerosis. Eur J Neurol. 2020;27:2218.CrossRefGoogle ScholarPubMed
Baker, D, Marta, M, Pryce, G, Giovannoni, G, Schmierer, K. Memory B cells are major targets for effective immunotherapy in relapsing multiple sclerosis. EBioMedicine. 2017;16:4150. DOI 10.1016/j.ebiom.2017.01.042.CrossRefGoogle ScholarPubMed
Halpern, WG, Lappin, P, Zanardi, T, et al. Chronic administration of belimumab, a BLyS antagonist, decreases tissue and peripheral blood B-lymphocyte populations in cynomolgus monkeys: pharmacokinetic, pharmacodynamic, and toxicologic effects. Toxicol Sci. 2006;91:58699. DOI 10.1093/toxsci/kfj148.CrossRefGoogle ScholarPubMed
Dubey, AK, Handu, SS, Dubey, S, Sharma, P, Sharma, KK, Ahmed, QM. Belimumab: first targeted biological treatment for systemic lupus erythematosus. J Pharmacol Pharmacother. 2011;2:3179. DOI 10.4103/0976-500X.85930.CrossRefGoogle ScholarPubMed
Addition of belimumab to B-cell depletion in relapsing-remitting multiple sclerosis. ClinicalTrials. Gov. Available at: https://clinicaltrials.gov/ct2/show/study/NCT04767698; accessed May 18, 2021.Google Scholar
Hewett, K., Sanders, D.B., Grove, R.A., et al. Randomized study of adjunctive belimumab in participants with generalized myasthenia gravis. Neurology. 2018;90:e1425e1434. DOI 10.1212/WNL.0000000000005323.CrossRefGoogle ScholarPubMed
Stohl, W, Hilbert, DM. The discovery and development of belimumab: the anti-BLyS-lupus connection. Nat Biotechnol. 2012;30:6977. DOI 10.1038/nbt.2076.CrossRefGoogle ScholarPubMed
Dingjan, GM, Middendorp, S, Dahlenborg, K, Maas, A, Grosveld, F, Hendriks, RW. Bruton’s tyrosine kinase regulates the activation of gene rearrangements at the lambda light chain locus in precursor B cells in the mouse. J Exp Med. 2001;193:116978. DOI 10.1084/jem.193.10.1169.CrossRefGoogle ScholarPubMed
Middendorp, S, Dingjan, GM, Hendriks, RW. Impaired precursor B cell differentiation in Bruton’s tyrosine kinase-deficient mice. J Immunol. 2002;168:2695703. DOI 10.4049/jimmunol.168.6.2695.CrossRefGoogle ScholarPubMed
Nomura, K, Kanegane, H, Karasuyama, H, et al. Genetic defect in human X-linked agammaglobulinemia impedes a maturational evolution of pro-B cells into a later stage of pre-B cells in the B-cell differentiation pathway. Blood. 2000;96:6107.Google ScholarPubMed
Torke, S, Weber, MS. Inhibition of bruton’s tyrosine kinase as a novel therapeutic approach in multiple sclerosis. Expert Opin Investig Drugs. 2020;29:114350. DOI 10.1080/13543784.2020.1807934.CrossRefGoogle ScholarPubMed
Martin, E, Aigrot, M-S, Grenningloh, R, et al. Bruton’s tyrosine kinase inhibition promotes myelin repair. Adv Exp Med Biol. 2020;5:12333. DOI 10.3233/bpl-200100.Google ScholarPubMed
Becker, A, Martin, EC, Mitchell, DY, et al. Safety, Tolerability, Pharmacokinetics, Target Occupancy, and Concentration-QT Analysis of the Novel BTK Inhibitor Evobrutinib in Healthy Volunteers. Clin Transl Sci. 2020 Mar;13(2):325-336. DOI 10.1111/cts.12713 CrossRefGoogle ScholarPubMed
A study of efficacy and safety of M2951 in participants with relapsing multiple sclerosis. Available at: https://www.clinicaltrials.gov/ct2/show/NCT02975349?term=evobrutinib&cond=Multiple+Sclerosis&draw=2&rank=5 Google Scholar
Montalban, X, Arnold, DL, Weber, MS, et al. Placebo-controlled trial of an oral BTK inhibitor in multiple sclerosis. N Engl J Med. 2019;380:240617. DOI 10.1056/NEJMoa1901981.CrossRefGoogle ScholarPubMed
Study of evobrutinib in participants with relapsing multiple sclerosis (RMS) (EvolutionRMS 2). Available at: https://www.clinicaltrials.gov/ct2/show/NCT04338061?term=evobrutinib&cond=Multiple+Sclerosis&draw=2&rank=1 Google Scholar
Nazi, I, Kelton, JG, Larché, M, et al. The effect of rituximab on vaccine responses in patients with immune thrombocytopenia. Blood. 2013;122:194653. DOI 10.1182/blood-2013-04-494096.CrossRefGoogle ScholarPubMed
Bar-Or, A, Calkwood, JC, Chognot, C, et al. Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis. Neurology. 2020;95:e1999e2008. DOI 10.1212/WNL.0000000000010380.CrossRefGoogle ScholarPubMed
Dolgin, E. BTK blockers make headway in multiple sclerosis. Nat Biotechnol. 2021;39:35. DOI 10.1038/s41587-020-00790-7.CrossRefGoogle ScholarPubMed
Torke, S, Pretzsch, R, Häusler, D, et al. Inhibition of Bruton’s tyrosine kinase interferes with pathogenic B-cell development in inflammatory CNS demyelinating disease. Acta Neuropathol. 2020;140:53548. DOI 10.1007/s00401-020-02204-z.CrossRefGoogle ScholarPubMed
Boschert, U, Crandall, T, Pereira, A, et al. T cell mediated experimental CNS autoimmunity induced by PLP in SJL mice is modulated by evobrutinib (M2951) a novel Bruton’s tyrosine kinase inhibitor. Mult Scler J. 2017;23:327.Google Scholar
Tisovic, K, Amezcua, L. Women’s health: contemporary management of MS in pregnancy and post-partum. Biomedicines. 2019;7:32. DOI 10.3390/biomedicines7020032.CrossRefGoogle ScholarPubMed
Wallin, MT, Culpepper, WJ, Campbell, JD, et al. The prevalence of MS in the United States: a population-based estimate using health claims data. Neurology. 2019;92:e1029e1040. DOI 10.1212/WNL.0000000000007035.CrossRefGoogle ScholarPubMed
Das, G, Damotte, V, Gelfand, JM, et al. Rituximab before and during pregnancy: a systematic review, and a case series in MS and NMOSD. Neurol Neuroimmunol Neuroinflamm. 2018;5:e453. DOI 10.1212/NXI.0000000000000453.CrossRefGoogle Scholar
Smith, JB, Hellwig, K, Fink, K, Lyell, DJ, Piehl, F, Langer-Gould, A. Rituximab, MS, and pregnancy. Neurol Neuroimmunol Neuroinflamm. 2020;7, 10.1212/NXI.0000000000000734.CrossRefGoogle ScholarPubMed
Ciplea, AI, Langer-Gould, A, de Vries, A, et al. Monoclonal antibody treatment during pregnancy and/or lactation in women with MS or neuromyelitis optica spectrum disorder. Neurol Neuroimmunol Neuroinflamm. 2020;7:e723. DOI 10.1212/NXI.0000000000000723.CrossRefGoogle ScholarPubMed
Kümpfel, T, Thiel, S, Meinl, I, et al. Anti-CD20 therapies and pregnancy in neuroimmunologic disorders: a cohort study from Germany. Neurol Neuroimmunol Neuroinflamm. 2021;8:e913. DOI 10.1212/NXI.0000000000000913.CrossRefGoogle ScholarPubMed
Wiendl, H, Gold, R, Berger, T, et al. Multiple Sclerosis Therapy Consensus Group (MSTCG): position statement on disease-modifying therapies for multiple sclerosis (white paper). Ther Adv Neurol Disord. 2021;14:175628642110396. DOI 10.1177/17562864211039648.CrossRefGoogle Scholar
Keystone, E, Fleischmann, R, Emery, P, et al. Safety and efficacy of additional courses of rituximab in patients with active rheumatoid arthritis: an open-label extension analysis. Arthritis Rheum. 2007;56:3896908. DOI 10.1002/art.23059.CrossRefGoogle ScholarPubMed
Stathopoulos, P, Dalakas, MC. Evolution of anti-B cell therapeutics in autoimmune neurological diseases. Neurotherapeutics. 2022;112:57. DOI 10.1007/s13311-022-01196-w.Google Scholar
van Vollenhoven, RF, Emery, P, Bingham, CO, et al. Longterm safety of patients receiving rituximab in rheumatoid arthritis clinical trials. J Rheumatol. 2010;37:55867. DOI 10.3899/jrheum.090856.CrossRefGoogle ScholarPubMed
Figure 0

Table 1: A summary of medicines targeting B cells that have been used in MS

Figure 1

Figure 1: Expression of cell surface antigens throughout B-cell maturation. CD19 is expressed in all stages of B-cell development, with the exception of stem cells and the majority of plasma cells. CD20 is not present on plasma cells, most plasmablasts, pro-B cells, and stem cells. BAFF-receptor (BAFF-R) is expressed on both immature and mature B cells in the germinal center, as well as memory B cells and late plasmablasts. Transmembrane activator and CAML interactor (TACI) and B-cell maturation antigen (BCMA) are expressed on germinal center B cells, memory cells, and antibody-secreting cells.