ArticlePDF AvailableLiterature Review

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

Authors:

Abstract and Figures

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.
Content may be subject to copyright.
Review Article
B-Cell-Directed Therapies: A New Era in Multiple Sclerosis Treatment
Panagiotis Kanatas1
,
2, Ioannis Stouras1, Leonidas Stefanis1
,
2and Panos Stathopoulos1
,
2
1First Department of Neurology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece and 2Eginiteion Hospital, Athens, Greece
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 mol-
ecules such as CD20 (rituximab, ocrelizumab, ofatumumab, ublituximab) and CD19 (inebilizumab), and molecules necessary for B-cell acti-
vation such as B cell activating factor (BAFF) (belimumab) and Brutons Tyrosine Kinase (BTK) (evobrutinib). We finally discuss the use of
B-cell-targeted therapeutics in pregnancy.
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 lintérêt pour le rôle de ces derniers dans limmunopathologie de la maladie. Aussi
présenterons-nous dans larticle de synthèse des données probantes qui font ressortir laction des lymphocytes B dans les mécanismes
dévolution de la SP, depuis la maladie elle-même jusquaux 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 inflam-
matoire du SNC, dans la SP, à savoir i) la production dautoanticorps; ii) la présentation dantigènes; iii) la production de cytokines pro-inflam-
matoires (activation de voisinage); iv) le le du virus dEpstein-Barr. Dans la secondepartie, nous présenterons un résumé des médicaments qui
ciblent les lymphocytes B chez les patients atteints de la SP, et discuterons de leur place dans larsenal thérapeutique de la maladie daprès les
résultats dessais cliniques et des données réelles. Les stratégies thérapeutiques traitées dans larticle porteront notamment sur la prise pour cible
des molécules présentes à la surface des lymphocytes telles que la CD20 (par le rituximab, locrélizumab, lofatumumab ou lublituximab) et la
CD19 (par linébilizumab), ainsi quesur la prise pour cible des molécules cessaires à lactivation des lymphocytes B tel que le facteur dactivation
des lymphocytes B (BAFF, en anglais) (par le bélimumab), et à linhibition de la tyrosine-kinase de Bruton (par lévobrutinib). Enfin, il sera
question de lemploi des traitements ciblant les lymphocytes B chez les femmes enceintes.
Keywords: Multiple sclerosis; B cells; Monoclonal antibodies; CD20; CD19
(Received 7 January 2022; final revisions submitted 15 April 2022; date of acceptance 3 May 2022)
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 dis-
ability 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 2:1.13The 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 cat-
egories 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.4
Significant progress in understanding MS pathophysiology has
been accomplished in the past decades. Two hundred and thirty-
Corresponding author: Panos Stathopoulos, Eginiteion Hospital, V. Sofias 72, 115 28 Athens, Greece. Email: pmstathopoulos@gmail.com
Cite this article: Kanatas P, Stouras I, Stefanis L, and Stathopoulos P. B-Cell-Directed Therapies: A New Era in Multiple Sclerosis Treatment. The Canadian Journal of Neurological
Sciences https://doi.org/10.1017/cjn.2022.60
© The Author(s), 2022. Published by Cambridge University Press on behalf of Canadian Neurological Sciences Federat ion. 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.
The Canadian Journal of Neurological Sciences (2022), 110
doi:10.1017/cjn.2022.60
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
three genetic variants have been identified as risk factors for MS, 32
of which refer to the major histocompatibility complex family
(MHC).5Prominent among the many risk variants, MHC Class
II DR15 molecule entails mechanistically relevant susceptibility
to the disease rather than just being a genetic marker.6
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 path-
ways (GC, CYP24A1).3,711
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 CD4þT cells to disease pathophysiol-
ogy.12 Often being present from the beginning and increasing in
quantity as disease progresses, axonal degeneration is regarded
as a correlate of disability progression.13
MS was long considered mainly T-cell-mediated; however,
intrathecal IgG synthesis,14 a hallmark of MS, supports B-cell
involvement.15,16 The T-cell-dominated view of MS pathogenesis
was further challenged by the remarkable efficiency of CD20þ
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-tar-
geted 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.17 Moreover, ectopic lymphoid follicles
are found primarily in the intrameningeal spaces.18 and are asso-
ciated with subpial cortical demyelination in patients with
SPMS.19,20 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 accom-
panied by oligodendrocyte apoptosis without immunoglobulin
deposition or complement activation. Finally, in type IV lesions
(rare; 1% of MS patients) inflammatory modulators result in non-
apoptotic death of oligodendrocytes in the white matter surround-
ing the plaque due to metabolic disorganization processes.21
However, it is important to note that IgG deposits in MS histopa-
thological specimens are not specific for MS22 and that no disease-
characterizing autoantibodies have been defined to date.
Nevertheless, pattern II has been linked to better response to
plasma exchange.23
Potential Roles of B Cells in MS Pathophysiology
Several studies have explored potential roles of B cells in the devel-
opment of MS: antibody production, antigen presentation, and
secretion of pro- and anti-inflammatory mediators are three
prominent research directions that have been explored.24,25 In
addition, clear epidemiological associations of B-lymphotropic
Epstein-Barr virus (EBV) infection to MS have led to the explora-
tions of its pathophysiological relevance.26
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 hall-
mark of MS diagnosis found in up to 95% of patients,27,28 are
not specific for MS (found also in e.g. meningitis and subacute scle-
rosing panencephalitis) and have been found to target intracellular
antigens in patients with MS.29,30 In addition, detection of intrathe-
cal IgM synthesis has been associated with onset of relapses and a
more aggressive disease course.31 Similarly to antibodies of oligo-
clonal bands, B cells of MS lesions have been found to target intra-
cellular antigens.30 Antibodies previously thought to be present in
MS such as antibodies against myelin oligodendrocyte glycopro-
tein (MOG) rather characterize a distinct disease entity (MOG-
antibody disease) that encompasses pediatric acquired demyelin-
ating syndrome, recurrent optic neuritis, acute disseminated
encephalomyelitis, and neuromyelitis optica without anti-aqua-
porin four autoantibodies. A minority of MS patients harbor anti-
bodies against a variety of antigens (some of them cell surface
proteins) such as contactin-2,32 OMGP,33 and other peptide and
lipid antigens.34,35
It must be noted that autoantibodies can also be responsible for
the activation and chemotaxis of CD4þT cells. The opsonization
of myelin antigens, even at low concentrations, enhances the pre-
sentative 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.3638
B Cells as Antigen-Presenting Cells
B cells are efficient APCs and express MHC class II and costimu-
latory molecules, such as CD40, CD80, and CD86.25,39 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.40
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 sympto-
matic onset of the disease, but do not affect either the proliferation
or the molecular profile (i.e. secreted cytokines, activation mark-
ers) of MOG-specific T cells in the spleen and the draining lymph
nodes.37
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 pre-
sented 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
CD4þT cells. These T cells responded robustly to individual
and pooled HLA-DR-SPs in MS patients, compared to healthy
2The Canadian Journal of Neurological Sciences
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
donors, suggesting that DR2a and DR2b could jointly shape an
autoreactive T-cell repertoire in MS.41
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 abnor-
mally 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.4244 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.19,25,45 At the same time, anti-inflammatory, regulatory
cytokines that are produced by B cells, such as IL-35,46 but also
TGF-β1 and IL-10, are instrumental in controlling inflammation
in experimental models of MS.47
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.48 Ninety-six percent of the general pop-
ulation is positive for IgG antibodies against EBV (indicating past
infection), while in MS patients this percentage is almost 100%.26
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.49 Studies and experiments have shaped four major
theories about EBVs role in the pathogenesis of MS: the cross-
reactivity hypothesis,50 the bystander damage hypothesis,51 the
αβ-crystallin hypothesis,52 and the EBV-infected autoreactive
B-cell hypothesis.53 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.54
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 CD8þT cells. Moreover, the EBV anti-
apoptotic protein BHRF1, produced by both latently and lytically
infected cells, inhibits B-cell apoptosis,55 resulting in immortaliza-
tion of the autoreactive B cells it infects. In support of this hypoth-
esis, substantial EBV persistence in B and plasma cells as well as
meningeal B-cell lymphoid follicles of all MS cases examined
was reported,56 but could not be reproduced in multiple indepen-
dent replication studies.5760 Overall, the epidemiological associa-
tions 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); how-
ever, 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-gener-
ation 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-and3
rd-generation ones.61
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.
Le Journal Canadien Des Sciences Neurologiques 3
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
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,6264 and may
correlate positively with disease severity,65 CD20 serves a more
important role on B cells. The molecule is not expressed through-
out 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).6668
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 cells69 and its presence on the sur-
face 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.70 Evidence from animal studies
shows that anti-CD20 antibody-mediated B-cell depletion may
be incomplete in lymph node germinal centers.71 Moreover, cer-
ebrospinal 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.7275 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.
Rituximab
Rituximab is a 1st-generation chimeric monoclonal antibody (IgG1),
engineered by fusing a murine Fab with a human Fc domain.61 Its
elimination half-time is estimated at around 20 days;76 it may,
however, vary according to sex, body weight, and renal function.77
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).78
In addition, a retrospective observational study of 808 patients with
RRMS revealed absence of rebound disease activity upon rituximab
cessation,79 whereas rebound activity has been reported with nata-
lizumab80 and fingolimod cessation.81,82
In regard to progressive forms, a phase 3 (n =439 PPMS
patients), double-blind and placebo-controlled trial concluded in
2009 that CD20þB-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.83 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 rituximabs
safety as well as its efficacy in reducing GdELs; GdELs went from
26.2% (pretreatment) to 4.6% in the pooled post-treatment
cohort.84 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.85 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,86,87 and the
efficacy evident, despite the lack of phase III trials.88 The drug is
Table 1: A summary of medicines targeting B cells that have been used in MS
Drug Type Target molecule Clinical trials Pregnancy Dosing
Rituximab Chimeric IgG1 Large extracellular loop
of CD20 molecule
Phase 1; Phase 2 (Hermes);
Phase 3 (Olympus).
Not FDA-approved
Last infusion 3.5
months prior to
conception
Intravenous dose of 2 ×1000 mg in
a 2-week period
Ocrelizumab Humanized
glycosylated IgG1
Large extracellular loop
of CD20 molecule
Phase 2; Phase 3 (OPERA I &
II, ORATORIO). FDA-approved
Last infusion 6
months prior to
conception
Intravenous doses of 600 mg twice
a year
Ofatumumab Fully human IgG1 Large & small
extracellular loop of
CD20 molecule
Phase 2b (MIRROR); Phase 3
(ASCLEPIOS I & II).
FDA-approved
N/A 20 mg/0.4 mL once per week for
the first 3 weeks, once a month
thereafter
Ublituximab Glycoengineered
chimeric IgG1
Large extracellular loop
of CD20 molecule
Phase 2 (NEDA); Phase 3
(ULTIMATE I & II). Not yet
FDA-approved
N/A N/A
MEDI-551 Glycoengineered,
humanized, fucozylated
IgG1κ
CD19 Phase 1; Phase 2/3
(N-MOmmentum).
FDA-approved
N/A Intravenous infusions of 300 mg
every 6 months.
Atacicept Human recombinant
fusion protein (Fc
IgG þTACI)
BAFF-APRIL Phase 2 (ATAMS & ANOS).
Failed
N/A
Evobrutinib Small molecule drug BTK Phase 2; Phase 3.
Not FDA-approved
N/A 75 mg dose daily
4The Canadian Journal of Neurological Sciences
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
generally well-tolerated by patients all throughout the MS type
spectrum, and the main adverse effects are mild to moderate infu-
sion-related reactions (IRRs), typically with the first dose, as well as
mild to moderate infections. No cases of progressive multifocal leu-
koencephalopathy (PML) due to John Cunningham virus, which is
mostly seen with natalizumab treatment,89,90 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.91,92 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 5001000 mg every 6 months
or yearly.61
Ocrelizumab
Ocrelizumab, an IgG1 immunoglobulin, is a 2nd-generation
recombinant humanized anti-CD20 mAb.93 The drug has a termi-
nal 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
action95 and as a humanized molecule is expected to be less
immunogenic than rituximab with lower titles of neutralizing
anti-drug antibodies.96,97
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 4647%) and an impressive reduction of the mean number of
GdELs (by 94%) over a 96-week time period compared to inter-
feron 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 mea-
sured).98 ORATORIO, a phase 3, double-blind, placebo-con-
trolled trial, examined ocrelizumabs efficacy in managing
PPMS progression. Results from 732 patients revealed that ocre-
lizumab was associated with lower rates of clinical and MRI pro-
gression than placebo. Because in this study the effect was driven
by a fraction of PPMS that had evident MRI inflammation, EMA
hasapprovedthedrugonlyininflammatoryPPMS,whereas
otheragenciessuchastheFDAand Swissmedic have not applied
this restriction.99
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 num-
ber 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.100,101
Ocrelizumab is administered intravenously according to a fixed
dosing schedule, as approved based on the phase 3 studies. An ini-
tial 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 respec-
tively 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.102,103
Ofatumumab
Ofatumumab is a 2nd-generation, fully human IgG1 mAb104 that
depletes circulating CD20 B cells via ADCC105 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 signifi-
cant advantage of ofatumumab over teriflunomide in suppressing
both new relapses and GdEL activity (the latter by 9497%). Side
effects were reported to be mild to moderate and included injec-
tion-related reactions, headache, and infections (in 51.6% of
patients treated with ofatumumab) such as nasopharyngitis, upper
respiratory, and urinary tract infection.107 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 subcutane-
ous 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 chi-
meric IgG1 mAb that exerts its action primarily via ADCC, which
is facilitated by defucosylation of its Fc region and thereby
increased affinity for FcγRIIIa.61 A 48-week, placebo-controlled,
phase 2 trial of ublituximab in 45 RRMS patients established that
150 mg iv on day 1 and 450600 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 evi-
dence 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.109 In follow-up,
two double-blind, phase 3 trials [ULTIMATE I (NCT03277261)
and II (NCT03277248)] will assess ublituximabs efficacy and
safety compared to teriflunomide in 880 patients with RRMS.110,111
Anti-CD19 mAbs
CD19 belongs to the Ig superfamily and along with CD21, CD82,
and CD225 contributes to the formation of a multimolecular sig-
nal-transduction complex that ultimately leads to the activation of
PI-3 kinase.112 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).113 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.62 A phase 1 study assessing the
pharmacokinetic (intravenous and subcutaneous) profile of inebi-
lizumab, a humanized afucosylated IgG1κanti-CD19 mAb,114 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).116 The drug therefore competes for BAFF and APRIL
binding with native TACI, which is both membrane-bound and
soluble,117 as well as, to a lesser extent, with the other receptors
Le Journal Canadien Des Sciences Neurologiques 5
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
of the BAFF-APRIL system (BAFF-R and BCMA).118,119 After
improving rheumatoid arthritis and systemic lupus erythematosus
(SLE),120 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 sus-
pension of every atacicept trial in MS.119,121,122 Another 36-week,
phase 2, double-blind, and placebo-controlled atacicept trial in
34 patients with unilateral optic neuritis as clinical isolated syn-
drome also showed disease exacerbation, with a significantly
higher proportion of patients converting to clinically definite
MS compared with placebo.123 As atacicept effectively depletes
naive B cells and induces a transient but marked increase in
memory B cells (especially class-switched ones),117,124,125 possible
reasons why atacicept aggravated MS include elimination of regu-
latory naïve B cells and enhancement of pathogenic memory B-cell
function.126,127
Belimumab
Belimumab is a human IgG1λrecombinant 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.128,129
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,131 a disease mediated by pathogenic autoanti-
bodies.132 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 Brutons Tyrosine Kinase (BTK). BTK is an inte-
gral 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.133136 Importantly, about
75% of the CNS cells that express BTK are microglial, while
BTK expression levels in the brain increase after demyelination.137
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.138 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 amino-
transferase level elevation).139,140 Evobrutinib is now being
advanced to phase III evaluation, along with several other BTKI
(some of them with reversible BTK binding); fenebrutinib, ibruti-
nib, and tolebrutinib.141
While CD19/20 B-cell depletion has shown tremendous effi-
cacy 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.86,142,143
These disadvantages could possibly be avoided with inhibition
of B-cell activation and maturation with small molecules such as
BTKIs.136,144 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 patho-
genic B cells.145 Their effect on disease activity does not seem to be
as impressive as that of anti-B-cell antibodies, and they cannot con-
trol 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.146
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 child-
bearing age deserves special mention.147,148 Rituximab-associated
B-cell depletion persists long after the drugs elimination, which
occurs approximately 3 months after the last infusion. Thus, con-
ception can be considered safe 3 months after the last infusion
without significant risk of fetal exposure. But even if a woman con-
ceives before rituximabs effective elimination, IgG1 subclass mAbs
cannot cross the placenta barrier during the first trimester, result-
ing in low chance of fetal exposure.149 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.150 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.151 Although data
regarding ocrelizumab administration in this population group are
limited, it is reasonable to apply the same principles as with ritux-
imab. 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 natalizu-
mab. 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 con-
trolling disease in women with RRMS, during and partly after preg-
nancy. 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.152 Recent recommenda-
tions suggest prioritization of MS management and conception
postponement in cases of highly active MS and contraception
for up to 4 months after ocrelizumab administration.153
Conclusion
The therapeutic criterion underlines that B cells not only partici-
pate 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.154156
6The Canadian Journal of Neurological Sciences
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
Future studies will further inform on long-term effects of CD20-
targeting medications, on the use of oral BTKI agents and deter-
mine 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
1. Yeshokumar AK, Narula S, Banwell B. Pediatric multiple sclerosis. Curr
Opin Neurol. 2017;30:21621. DOI 10.1097/WCO.0000000000000452.
2. Thompson AJ, Baranzini SE, Geurts J, Hemmer B, Ciccarelli O. Multiple
sclerosis. Lancet. 2018;391:162236. DOI 10.1016/S0140-6736(18)30481-1.
3. 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.
4. Lublin FD. New multiple sclerosis phenotypic classification. Eur Neurol.
2014;72 Suppl 1:15. DOI 10.1159/000367614.
5. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis
genomic map implicates peripheral immune cells and microglia in suscep-
tibility. Science (1979). 2019;365:eaav7188. DOI 10.1126/science.aav7188.
6. OConnor 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.
7. De Jager PL, Jia X, Wang J, et al. Meta-analysis of genome scans and rep-
lication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis sus-
ceptibility loci. Nat Genet. 2009;41:77682. DOI 10.1038/ng.401.
8. Olsson T, Barcellos LF, Alfredsson L. Interactions between genetic, life-
style and environmental risk factors for multiple sclerosis. Nat Rev
Neurol. 2017;13:2536. DOI 10.1038/nrneurol.2016.187.
9. Cotsapas C, Mitrovic M. Genome-wide association studies of multiple
sclerosis. Clin Transl Immunol. 2018;7:e1018. DOI 10.1002/cti2.1018.
10. 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.
11. Mokry LE, Ross S, Ahmad OS, et al. Vitamin D and risk of multiple scle-
rosis: a mendelian randomization study. PLoS Med. 2015;12:e1001866.
DOI 10.1371/journal.pmed.1001866.
12. 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.
13. Bjartmar C, Yin X, Trapp BD. Axonal pathology in myelin disorders. J
Neurocytol. 1999;28:38395.
14. 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.
15. 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.
16. 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.
17. 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.
18. 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.
19. Moreno Torres I, García-MerinoA. Anti-CD20 monoclonal antibodies in
multiple sclerosis. Expert Rev Neurother. 2017;17:35971. DOI 10.1080/
14737175.2017.1245616.
20. Lassmann H. Pathogenic mechanisms associated with different clinical
courses of multiple sclerosis. Front Immunol. 2018;9:3116. DOI 10.
3389/fimmu.2018.03116.
21. Gh Popescu BF, Pirko I, Lucchinetti CF. Pathology of multiple sclerosis:
where do we stand? Continuum (Minneap Minn). 2013;19:90121.
22. 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.
23. 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.
24. Gasperi C, Stüve O, Hemmer B. B cell-directed therapies in multiple scle-
rosis. Neurodegener Dis Manag. 2016;6:3747. DOI 10.2217/nmt.15.67.
25. Häusser-Kinzel S, Weber MS. The role of B cells and antibodies in multi-
ple sclerosis, neuromyelitis optica, and related disorders. Front Immunol.
2019;10:201. DOI 10.3389/fimmu.2019.00201.
26. 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.
27. Abdelhak A, Hottenrott T, Mayer C, et al. CSF profile in primary
progressive multiple sclerosis: re-exploring the basics. PLOS ONE.
2017;12:e0182647.
28. Thouvenot E. Multiple sclerosis biomarkers: helping the diagnosis? Rev
Neurol (Paris). 2018;174:36471. DOI 10.1016/j.neurol.2018.04.002.
29. Brändle SM, Obermeier B, Senel M, et al. Distinct oligoclonal band anti-
bodies in multiple sclerosis recognize ubiquitous self-proteins. Proc Natl
Acad Sci U S A. 2016;113:78649. DOI 10.1073/pnas.1522730113.
30. Willis SN, Stathopoulos P, Chastre A, Compton SD, Hafler DA, OConnor
KC. Investigating the antigen specificity of multiple sclerosis central
nervous system-derived immunoglobulins. Front Immunol. 2015;6:600.
DOI 10.3389/fimmu.2015.00600.
31. 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.
32. 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.
33. Gerhards R, Pfeffer LK, Lorenz J, et al. Oligodendrocyte myelin glycopro-
tein as a novel target for pathogenic autoimmunity in the CNS. Acta
Neuropathol Commun. 2020;8:207. DOI 10.1186/s40478-020-01086-2.
34. Yeste A, Quintana FJ. Antigen microarrays for the study of autoimmune
diseases. Clin Chem. 2013;59:103644. DOI 10.1373/clinchem.2012.
194423.
35. 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.
36. Trotter J, DeJong LJ, Smith ME. Opsonization with antimyelin antibody
increases the uptake and intracellular metabolism of myelin in inflamma-
tory macrophages. J Neurochem. 1986;47:77989. DOI 10.1111/j.1471-
4159.1986.tb00679.x.
37. Flach AC, Litke T, Strauss J, et al. Autoantibody-boosted T-cell reactiva-
tion in the target organ triggers manifestation of autoimmune CNS dis-
ease. Proc Natl Acad Sci U S A. 2016;113:33238. DOI 10.1073/pnas.
1519608113.
38. Getahun A, Dahlström J, Wernersson S, Heyman B. IgG2a-mediated
enhancement of antibody and T cell responses and its relation to inhibi-
tory and activating Fc gamma receptors. J Immunol. 2004;172:526976.
DOI 10.4049/jimmunol.172.9.5269.
Le Journal Canadien Des Sciences Neurologiques 7
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
39. 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.
40. 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.
41. Wang J, Jelcic I, Mühlenbruch L, et al. HLA-DR15 molecules jointly shape
an autoreactive T cell repertoire in multiple sclerosis. Cell. 2020;183:1264
1281.e20. DOI 10.1016/j.cell.2020.09.054.
42. 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.
43. Molnarfi N, Schulze-Topphoff U, Weber MS, et al. MHC class II-depen-
dent 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.
44. Korn T, Mitsdoerffer M, Croxford AL, et al. IL-6 controls Th17 immunity
in vivo by inhibiting the conversion of conventional T cells into Foxp3þ
regulatory T cells. Proc Natl Acad Sci U S A. 2008;105:184605. DOI 10.
1073/pnas.0809850105.
45. 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.
46. Shen P, Roch T, Lampropoulou V, et al. IL-35-producing B cells are criti-
cal regulators of immunity during autoimmune and infectious diseases.
Nature. 2014;507:36670. DOI 10.1038/nature12979.
47. Arneth BM. Impact of B cells to the pathophysiology of multiple sclerosis.
J Neuroinflammation. 2019;16:128. DOI 10.1186/s12974-019-1517-1.
48. 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.
49. 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.
50. 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.
51. Angelini DF, Serafini B, Piras E, et al. Increased CD8þT cell response to
Epstein-Barr virus lytic antigens in the active phase of multiple sclerosis.
PLoS Pathog. 2013;9:e1003220.
52. Pender MP, Burrows SR. Epstein-Barr virus and multiple sclerosis: poten-
tial opportunities for immunotherapy. Clin Transl Immunol. 2014;3:e27.
53. 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.
54. 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.
55. 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 chemothera-
peutic drugs. Adv Exp Med Biol. 1996;406:8397. DOI 10.1007/978-1-
4899-0274-0_9.
56. 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.
57. 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.
58. 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.
59. 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.
60. Torkildsen Ø., Stansberg C, Angelskår SM, et al. Upregulation of
immunoglobulin-related genes in cortical sections from multiple sclerosis
patients. Brain Pathol. 2010;20:7209. DOI 10.1111/j.1750-3639.2009.
00343.x.
61. Whittam DH, Tallantyre EC, Jolles S, et al. Rituximab in neurological dis-
ease: principles, evidence and practice. Pract Neurol. 2019;19:5. DOI 10.
1136/practneurol-2018-001899.
62. Schuh E, Berer K, Mulazzani M, et al. Features of human
CD3þCD20þT cells. J Immunol. 2016;197:11117. DOI 10.4049/
jimmunol.1600089.
63. Palanichamy A, Jahn S, Nickles D, et al. Rituximab efficiently depletes
increased CD20-expressing T cells in multiple sclerosis patients.
J Immunol. 2014;193:5806. DOI 10.4049/jimmunol.1400118.
64. Sabatino JJ Jr, Wilson MR, Calabresi PA, Hauser SL, Schneck JP, Zamvil
SS. Anti-CD20 therapy depletes activated myelin-specific CD8(þ) T cells
in multiple sclerosis. Proc Natl Acad Sci U S A. 2019;116:258007. DOI 10.
1073/pnas.1915309116.
65. von Essen MR, Ammitzbøll C, Hansen RH, et al. Proinflammatory
CD20þT cells in the pathogenesis of multiple sclerosis. Brain.
2019;142:12032. DOI 10.1093/brain/awy301.
66. 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.
67. 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.
68. Tedder TF, Streuli M, Schlossman SF, Saito H. Isolation and structure of a
CDNA encoding the B1 (CD20) cell-surface antigen of human B lympho-
cytes. Proc Natl Acad Sci U S A. 1988;85:20812. DOI 10.1073/pnas.
85.1.208.
69. Santos MAO, Lima MM. CD20 role in pathophysiology of Hodgkins dis-
ease. Rev Assoc Med Bras (1992). 2017;63:8103. DOI 10.1590/1806-
9282.63.09.810.
70. Maloney DG. Anti-CD20 antibody therapy for B-cell lymphomas. N Engl
J Med. 2012;366:200816. DOI 10.1056/NEJMct1114348.
71. 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.
72. 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.
73. 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.
74. 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.
75. 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.
76. Bar-Or A, Calabresi PAJ, Arnold D, et al. Rituximab in relapsing-remit-
ting multiple sclerosis: a 72-week, open-label, phase I trial. Ann Neurol.
2008;63:395400. DOI 10.1002/ana.21363.
77. 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.
78. 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.
79. 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.
80. lo Re M, Capobianco M, Ragonese P, et al. Natalizumab discontinuation
and treatment strategies in patients with multiple sclerosis (MS): a retro-
spective study from two Italian MS centers. Neurol Ther. 2015;4:14757.
DOI 10.1007/s40120-015-0038-9.
8The Canadian Journal of Neurological Sciences
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
81. 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.
82. 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.
83. Hawker K, OConnor 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;66:460
71. DOI 10.1002/ana.21867.
84. Salzer J, Svenningsson R, Alping P, et al. Rituximab in multiple sclerosis.
Neurology. 2016;87:207481. DOI 10.1212/WNL.0000000000003331.
85. Naegelin Y, Naegelin P, von Felten S, et al. Association of rituximab treat-
ment with disability progression among patients with secondary
progressive multiple sclerosis. JAMA Neurol. 2019;76:27481. DOI 10.
1001/jamaneurol.2018.4239.
86. 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.
87. Alping P, Askling J, Burman J, et al. Cancer risk for fingolimod, natalizu-
mab, and rituximab in multiple sclerosis patients. Ann Neurol.
2020;87:68899. DOI 10.1002/ana.25701.
88. 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.
89. 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.
90. 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.
91. 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.
92. Kapoor T, Mahadeshwar P, Hui-Yuen J, et al. Prevalence of progressive
multifocal leukoencephalopathy (PML) in adults and children with sys-
temic lupus erythematosus. Lupus Sci Med. 2020;7:e000388. DOI 10.
1136/lupus-2020-000388.
93. Syed YY. Ocrelizumab: a review in multiple sclerosis. CNS Drugs.
2018;32:88390.
94. Ocrevus | European Medicines Agency.Available at: https://www.ema.
europa.eu/en/medicines/human/EPAR/ocrevus; accessed December 1, 2021.
95. 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.
96. Vugmeyster Y, Beyer J, Howell K, et al. Depletion of B cells by a human-
ized anti-CD20 antibody PRO70769 in macaca fascicularis. J
Immunother. 2005;28:2129. DOI 10.1097/01.cji.0000155050.03916.04.
97. 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.
98. 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.
99. 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.
100. Rommer PS, Zettl UK. Managing the side effects of multiple sclerosis
therapy: pharmacotherapy options for patients. Expert Opin
Pharmacother. 2018;19:48398.
101. 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.
102. Gibiansky E, Petry C, Mercier F, et al. Ocrelizumab in relapsing and pri-
mary progressive multiple sclerosis: pharmacokinetic and pharmacody-
namic analyses of OPERA I, OPERA II and ORATORIO. Br J Clin
Pharmacol. 2021;87:251120. DOI 10.1111/bcp.14658.
103. 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.
104. Florou D, Katsara M, Feehan J, Dardiotis E, Apostolopoulos V. Anti-
CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatu-
mumab. Brain Sci. 2020;10:758. DOI 10.3390/brainsci10100758.
105. 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.
106. Ofatumumab. Am J Health Syst Pharm. 2020;77:20258. DOI 10.1093/
ajhp/zxaa322.
107. 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.
108. 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.
109. Fox E, Lovett-Racke AE, Gormley M, etal. 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.
110. 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.00000000
00015944.
111. 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
112. Tedder TF. CD19: a promising B cell target for rheumatoid arthritis. Nat
Rev Rheumatol. 2009;5:5727.
113. 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.
114. 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.
115. Safety and tolerability study of MEDI-551, a B-cell depleting agent, totreat
relapsing forms of multiple sclerosis.Available at: https://clinicaltrials.gov/
ct2/show/NCT01585766
116. 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.
117. Hoffmann FS, Kuhn P-H, Laurent SA, et al. The immunoregulator solu-
ble TACI is released by ADAM10 and reflects B cell activation in
autoimmunity. J Immunol. 2015;194:54252. DOI 10.4049/jimmunol.
1402070.
118. 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.
119. Hartung H-P, Kieseier BC. Atacicept: targeting B cells in multiple sclero-
sis. Ther Adv Neurol Disord. 2010;3:20516. DOI 10.1177/175628
5610371146.
120. 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.
121. 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.
Le Journal Canadien Des Sciences Neurologiques 9
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
122. A phase 2 study of atacicept in subjects with relapsing multiple sclerosis
(ATAMS). Available at: https://clinicaltrials.gov/ct2/show/NCT00642902
123. Sergott RC, Bennett JL, Rieckmann P, et al. Results from a phase II ran-
domized 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.
124. 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.
125. Jelcic I, al Nimer F, Wang J, et al. Autoreactive CD4þT cells in multiple
sclerosis. Cell. 2018;175:85100.e23. DOI 10.1016/j.cell.2018.08.011.
126. 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.
127. Baker D, Marta M, Pryce G, Giovannoni G, Schmierer K. Memory B cells
are major targets for effective immunotherapy in relapsing multiple scle-
rosis. EBioMedicine. 2017;16:4150. DOI 10.1016/j.ebiom.2017.01.042.
128. Halpern WG, Lappin P, Zanardi T, et al. Chronic administration of beli-
mumab, 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.
129. Dubey AK, Handu SS, Dubey S, Sharma P, Sharma KK, Ahmed QM.
Belimumab: first targeted biological treatment for systemic lupus erythe-
matosus. J Pharmacol Pharmacother. 2011;2:3179. DOI 10.4103/0976-
500X.85930.
130. 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.
131. Hewett K., Sanders D.B., Grove R.A., et al. Randomized study of adjunc-
tive belimumab in participants with generalized myasthenia gravis.
Neurology. 2018;90:e1425e1434. DOI 10.1212/WNL.0000000000
005323.
132. 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.
133. Dingjan GM, Middendorp S, Dahlenborg K, Maas A, Grosveld F,
Hendriks RW. Brutons 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.
134. Middendorp S, Dingjan GM, Hendriks RW. Impaired precursor B cell dif-
ferentiation in Brutons tyrosine kinase-deficient mice. J Immunol.
2002;168:2695703. DOI 10.4049/jimmunol.168.6.2695.
135. Nomura K, Kanegane H, Karasuyama H, et al. Genetic defectin 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.
136. Torke S, Weber MS. Inhibition of brutons tyrosine kinase as a novel
therapeutic approach in multiple sclerosis. Expert Opin Investig Drugs.
2020;29:114350. DOI 10.1080/13543784.2020.1807934.
137. Martin E, Aigrot M-S, Grenningloh R, et al. Brutons tyrosine kinase
inhibition promotes myelin repair. Adv Exp Med Biol. 2020;5:12333.
DOI 10.3233/bpl-200100.
138. 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
139. 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=MultipleþSclerosis&draw=2&
rank=5
140. 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.
141. Study of evobrutinib in participants with relapsing multiple sclerosis
(RMS) (EvolutionRMS 2). Available at: https://www.clinicaltrials.gov/
ct2/show/NCT04338061?term=evobrutinib&cond=MultipleþSclerosis&
draw=2&rank=1
142. 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.
143. Bar-Or A, Calkwood JC, Chognot C, et al. Effect of ocrelizumab on vac-
cine responses in patients with multiple sclerosis. Neurology. 2020;95:
e1999e2008. DOI 10.1212/WNL.0000000000010380.
144. Dolgin E. BTK blockers make headway in multiple sclerosis. Nat
Biotechnol. 2021;39:35. DOI 10.1038/s41587-020-00790-7.
145. Torke S, Pretzsch R, Häusler D, et al. Inhibition of Brutons 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.
146. 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 Brutons tyrosine kinase inhibitor. Mult Scler J.
2017;23:327.
147. Tisovic K, Amezcua L. Womens health: contemporary management of
MS in pregnancy and post-partum. Biomedicines. 2019;7:32. DOI 10.
3390/biomedicines7020032.
148. 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.00000000000
07035.
149. Das G, Damotte V, Gelfand JM, et al. Rituximab before and during preg-
nancy: a systematic review, and a case series in MS and NMOSD. Neurol
Neuroimmunol Neuroinflamm. 2018;5:e453. DOI 10.1212/NXI.
0000000000000453.
150. 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.
151. Ciplea AI, Langer-Gould A, de Vries A, et al. Monoclonal antibody treat-
ment during pregnancy and/or lactation in women with MS or neuromye-
litis optica spectrum disorder. Neurol Neuroimmunol Neuroinflamm.
2020;7:e723. DOI 10.1212/NXI.0000000000000723.
152. 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.
153. 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.
154. Keystone E, Fleischmann R, Emery P, et al. Safety and efficacy of addi-
tional courses of rituximab in patients with active rheumatoid arthritis:
an open-label extension analysis. Arthritis Rheum. 2007;56:3896908.
DOI 10.1002/art.23059.
155. Stathopoulos P, Dalakas MC. Evolution of anti-B cell therapeutics in auto-
immune neurological diseases. Neurotherapeutics. 2022;112:57. DOI 10.
1007/s13311-022-01196-w.
156. van Vollenhoven RF, Emery P, Bingham CO 3rd, et al. Longterm safety
of patients receiving rituximab in rheumatoid arthritis clinical trials.
J Rheumatol. 2010;37:55867. DOI 10.3899/jrheum.090856.
10 The Canadian Journal of Neurological Sciences
https://doi.org/10.1017/cjn.2022.60 Published online by Cambridge University Press
... Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disorder of the central nervous system (CNS) [1][2][3][4][5][6] with a prevalence exceeding two million people worldwide [3]. It is a leading cause of nontraumatic physical disability in young adults [3,6]. ...
... Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disorder of the central nervous system (CNS) [1][2][3][4][5][6] with a prevalence exceeding two million people worldwide [3]. It is a leading cause of nontraumatic physical disability in young adults [3,6]. In recent years, the classic paradigm of MS as a primarily T-cell-driven disease has been challenged by evidence that revealed B-cells playing a larger role in pathogenesis than previously understood [1,2,6], leading to the development of selective B-cell depleting disease-modifying therapies (DMTs) that specifically target CD20. ...
... It is a leading cause of nontraumatic physical disability in young adults [3,6]. In recent years, the classic paradigm of MS as a primarily T-cell-driven disease has been challenged by evidence that revealed B-cells playing a larger role in pathogenesis than previously understood [1,2,6], leading to the development of selective B-cell depleting disease-modifying therapies (DMTs) that specifically target CD20. These new therapies have proven to be highly efficacious in reducing MS disease activity. ...
Article
Full-text available
Multiple sclerosis (MS) has a global prevalence exceeding two million people and is a leading cause of non-traumatic physical disability. MS can be treated with ocrelizumab, an anti-CD20 monoclonal antibody. West Nile virus (WNV) is the most common cause of mosquito-borne viral encephalitis in North America. It can lead to neuroinvasive WNV disease (WNND) affecting the brain and peripheral nervous system, especially in immunocompromised patients, such as those being treated with ocrelizumab for MS. WNND is exceedingly rare and reported in less than 1% of cases of WNV. It has been established that inpatient rehabilitation improves functional outcomes in patients with MS and those with WNND. However, the inpatient rehabilitation outcomes in patients diagnosed with both WNND and MS have not been reported. In this study, we aimed to examine the rehabilitation outcomes of MS patients on ocrelizumab diagnosed with WNND. We performed a retrospective chart review of patients with MS treated with ocrelizumab, who were diagnosed with WNND and admitted to a single facility. Rehabilitation outcomes were assessed using functional independence measure (FIM) scores on admission and discharge. Three patients met the inclusion criteria; two in acute rehab, and one in the long-term acute care hospital (LTACH). Both patients admitted to acute inpatient rehabilitation showed an improvement in FIM scores from admission to discharge, one patient from 9 to 16 and the other from 14 to 54. However, the patient admitted to the LTACH had no improvement in FIM score from admission to discharge. Patients admitted to acute rehab were ultimately discharged home, while the patient admitted to the LTACH required discharge to a subacute rehabilitation facility. Based on our findings, intense and prolonged comprehensive inpatient rehabilitation is associated with improved functional outcomes and increased likelihood of discharge to home in this population suffering from both central and peripheral nervous system involvement due to MS and WNND.
... While MS was traditionally considered a Tcell-mediated disease, the presence of intrathecal IgG synthesis, a hallmark of MS, strongly suggests the involvement of B cells (Cepok et al., 2005;Kowarik et al., 2012). The predominant perception of MS pathogenesis being T-cell-driven was challenged by the notable efficacy of CD20 + B-cell depletion in halting inflammatory activity in MS patients (Kanatas et al., 2023). Moreover, recent studies have shown that B cells act as antigen-presenting cells in the periphery, interacting with activated T cells which then migrate to the CNS and contribute to neural damage (Jelcic et al., 2018). ...
Article
Full-text available
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system that primarily affects young adults, predominantly females. This was partially attributed to sex differences in immunity, which are influenced by changes in sex hormones occurring during women‘s life, among other factors. Furthermore, MS patients experience significant improvement in their symptoms during pregnancy when levels of female sex-hormones significantly increase. This phenomenon was attributed to immune adaptations occurring during gestation which are regulated by paternal antigens and sex hormones. The human chorionic gonadotropin (hCG) was shown to have strong immunosuppressive abilities. We aimed to analyze here the capacity of the hCG to regulate pro- and anti-inflammatory cytokine production by PBMC from MS patients. PBMC isolated from 17 MS patients receiving IFNβ1a treatment were cultured with or without recombinant or urinary hCG. Cytokine production in the supernatants was assessed using a CBA array and cytokine production by lymphocytes and expression of co-stimulatory molecules in B-lymphocytes were evaluated by flow cytometry. hCG reduced the production of TNF by PBMC from MS patients while lowering the percentages of TNF producing T cells and diminishing the production of TNF by B cells. hCG significantly boosted the production of IL-10 by regulatory T cells and CD19 high B cells from MS patients. Furthermore, hCG treatment lowered the percentages of CD80+CD86+ expressing B cells within PBMC from MS patients. Overall, our results described a novel and not yet explored mechanisms of action of hCG in the context of MS.
... (6) Expanded Treatment Options: The advent of BCDT has broadened treatment options for autoimmune diseases, moving beyond broad immunosuppressive agents with systemic side effects. These therapies offer a more targeted approach, directly addressing the underlying autoimmune response, thus presenting new avenues for managing autoimmune diseases and improving patient care [2,7,[63][64][65]. ...
Article
Full-text available
It has been rediscovered in the last fifteen years that B-cells play an active role in autoimmune etiology rather than just being spectators. The clinical success of B-cell depletion therapies (BCDTs) has contributed to this. BCDTs, including those that target CD20, CD19, and BAFF, were first developed to eradicate malignant B-cells. These days, they treat autoimmune conditions like multiple sclerosis and systemic lupus erythematosus. Particular surprises have resulted from the use of BCDTs in autoimmune diseases. For example, even in cases where BCDT is used to treat the condition, its effects on antibody-secreting plasma cells and antibody levels are restricted, even though these cells are regarded to play a detrimental pathogenic role in autoimmune diseases. In this Review, we provide an update on our knowledge of the biology of B-cells, examine the outcomes of clinical studies employing BCDT for autoimmune reasons, talk about potential explanations for the drug’s mode of action, and make predictions about future approaches to targeting B-cells other than depletion.
... Molecular mimicry has been described for several EBV proteins and human proteins, and epitope spreading and bystander activation/damage may also play important roles (3,4,(10)(11)(12). CD20 antibodies, the most recent addition to the therapeutic management of MS, specifically target the major B cell populations harboring active or latent EBV, and other drugs may also target different stages of EBV infection (13). Altogether, further elucidation of the biological mechanisms underlying the role of EBV in MS highly likely will reveal important insights into the pathogenesis of MS. ...
... Furthermore, the mechanism and the pathogenesis of MS have been identified and confirmed in the EAE [13]. However, the model has certain limitations, such as the involvement of B cells, which is well established in MS, whereas the involvement of B cells is not required with the MOG 35-55 EAE model [14,15]. Also, the induction of EAE is induced artificially, not spontaneity like the MS, which makes the model imperfect in studying the etiology of the disease. ...
Article
Full-text available
To investigate the effect of the therapeutic treatment of the immunopeptide, peptide inhibitor of trans-endothelial migration (PEPITEM) on the severity of disease in a mouse model of experimental autoimmune encephalomyelitis (EAE) as a model for human multiple sclerosis (MS), a series of experiments were conducted. Using C57BL/6 female mice, we dosed the PEPITEM in the EAE model via IP after observing the first sign of inflammation. The disease was induced using MOG35-55 and complete Freund’s adjuvants augmented with pertussis toxin. The EAE score was recorded daily until the end of the experiment (21 days). The histological and immunohistochemistry analysis was conducted on the spinal cord sections. A Western blot analysis was performed to measure the protein concentration of MBP, MAP-2, and N-Cadherin, and ELISA kits were used to measure IL-17 and FOXP3 in the serum and spinal cord lysate. The therapeutic treatment with PEPITEM reduced the CNS infiltration of T cells, and decreased levels of the protein concertations of MBP, MAP-2, and N-Cadherin were observed, in addition to reduced concertations of IL-17 and FOXP3. Using PEPITEM alleviated the severity of the symptoms in the EAE model. Our study revealed the potential of PEPITEM to control inflammation in MS patients and to reduce the harmful effects of synthetic drugs.
... It is expressed during several stages of B-cell maturation (Figure 1), i.e. in pre-B cells and mature B cells, not including stem cells and plasma cells, and is therefore an attractive target for monoclonal antibody-based therapy [33]. B cells targeted by CD20specific monoclonal antibodies are eliminated via three main mechanisms: programmed cell death/apoptosis, complement-dependent cytotoxicity (CDC), or antibodydependent cellular cytotoxicity (ADCC) [34]. Rituximab is a 1st generation chimeric monoclonal antibody (IgG1κ), engineered by fusing a murine Fab with a human Fc domain [35]. ...
Article
Full-text available
Myasthenia gravis (MG) is an autoimmune disorder characterized by muscle weakness and fatigue, mediated in the majority of cases by IgG1 autoantibodies targeting the acetylcholine receptor (AChR). As AChR autoantibodies have been shown to be pathogenic, therapies targeting B cells have been applied in patients with AChR MG for more than a decade. Recently, a phase 2 trial of the CD20-targeting agent, rituximab, in AChR MG unfortunately failed to meet its primary endpoint. Converging data however from non-randomized clinical series, some of which with more participants than the phase 2 trial, support efficacy of rituximab in AChR MG, especially early onset disease. In this opinion article, we summarize both clinical data and mechanistic principles on the use of CD20 depletion therapy in AChR MG, which we believe lend support to the argument that CD20 depletion can still be a useful therapeutic strategy for patients with AChR MG.
Article
Full-text available
Esta pesquisa investiga a progressão das terapias emergentes no âmbito do tratamento da esclerose múltipla (EM), enfatizando a importância dessas abordagens no manejo desta intricada condição neurológica. As terapias emergentes desempenham uma função crucial na busca por alternativas mais eficazes e focalizadas, ultrapassando as abordagens convencionais de modulação imunológica e controle sintomático. A análise integral destas opções objetiva proporcionar aos profissionais de saúde uma compreensão aprofundada das estratégias mais recentes e inovadoras, levando em consideração fatores como a progressão da patologia e a qualidade de vida dos pacientes. A determinação das terapias emergentes na EM é multifacetada, influenciada por variáveis como o perfil genético do paciente, estágio da doença e resposta a tratamentos anteriores. Nesse contexto, a avaliação comparativa de distintas abordagens, tais como imunoterapias avançadas, terapias celulares e intervenções genéticas, torna-se imperativa para garantir a eficácia do tratamento e minimizar possíveis efeitos adversos. Ademais, o tratamento da EM com terapias emergentes representa um desafio complexo que exige uma abordagem integrada. Esta pesquisa explora diversas modalidades terapêuticas, desde intervenções farmacológicas até métodos não farmacológicos, com o intuito de fornecer discernimentos valiosos para otimizar o gerenciamento da EM e aprimorar a qualidade de vida dos pacientes pós-tratamento. A compreensão das complexidades e sutilezas destas terapias é crucial para assegurar um cuidado abrangente e personalizado aos pacientes com EM, ressaltando a importância de uma abordagem holística na gestão dessa condição neurodegenerativa. Por fim, este artigo almeja consolidar informações atualizadas sobre terapias emergentes na esclerose múltipla, fornecendo uma análise crítica da literatura científica disponível. O propósito é contribuir para o contínuo desenvolvimento das práticas clínicas nessa área, promovendo a eficácia dos procedimentos e aprimorando a qualidade de vida para os pacientes que enfrentam esta desafiadora condição neurológica.
Article
Ublituximab (BRIUMVITM), an anti-CD20 immunoglobulin G1 monoclonal antibody, is a promising new treatment option for patients with relapsing forms of multiple sclerosis (MS). Ublituximab is approved in the USA and the EU for the treatment of adult patients with relapsing forms of MS. Following a starting dose of 150 mg infused over 4 h, ublituximab is conveniently administered twice-yearly as a short (1 h) intravenous infusion. In phase 3 clinical trials in patients with relapsing MS, ublituximab was more effective than oral teriflunomide at reducing annualized relapse rates and numbers of brain lesions over a period of 96 weeks. However, ublituximab did not result in a significantly lower risk of worsening of disability. Ublituximab had an acceptable tolerability profile in clinical trials. The most commonly reported adverse events were infusion-related reactions (IRRs), which occurred in almost half of ublituximab recipients. However, the majority of IRRs were of mild to moderate severity, occurred after the first dose and decreased in frequency with subsequent dosing.
Article
Full-text available
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
Article
Full-text available
Multiple sclerosis (MS) is a heterogenous autoimmune disease in which autoreactive lymphocytes attack the myelin sheath of the central nervous system (CNS). B lymphocytes in the cerebrospinal fluid (CSF) of MS patients contribute to inflammation and secrete oligoclonal immunoglobulins1,2. Epstein-Barr virus (EBV) infection has been linked to MS epidemiologically, but its pathological role remains unclear3. Here we demonstrate high-affinity molecular mimicry between the EBV transcription factor EBNA1 and the CNS protein GlialCAM, and provide structural and in-vivo functional evidence for its relevance. A cross-reactive CSF-derived antibody was initially identified by single-cell sequencing of the paired-chain B cell repertoire of MS blood and CSF, followed by protein microarray-based testing of recombinantly expressed CSF-derived antibodies against MS-associated viruses. Sequence analysis, affinity measurements, and the crystal structure of the EBNA1-peptide epitope in complex with the autoreactive Fab fragment allowed for tracking the development of the naïve EBNA1-restricted antibody to a mature EBNA1/GlialCAM cross-reactive antibody. Molecular mimicry is facilitated by a post-translational modification of GlialCAM. EBNA1 immunization exacerbates the mouse model of MS and anti-EBNA1/GlialCAM antibodies are prevalent in MS patients. Our results provide a mechanistic link for the association between MS and EBV, and could guide the development of novel MS therapies.
Article
Full-text available
Multiple sclerosis is a complex, autoimmune-mediated disease of the central nervous system characterized by inflammatory demyelination and axonal/neuronal damage. The approval of various disease-modifying therapies and our increased understanding of disease mechanisms and evolution in recent years have significantly changed the prognosis and course of the disease. This update of the Multiple Sclerosis Therapy Consensus Group treatment recommendation focuses on the most important recommendations for disease-modifying therapies of multiple sclerosis in 2021. Our recommendations are based on current scientific evidence and apply to those medications approved in wide parts of Europe, particularly German-speaking countries (Germany, Austria, and Switzerland).
Article
Full-text available
Objective To report pregnancy outcomes and disease activity (DA) in women with MS, neuromyelitis optica spectrum disorders (NMOSDs), and other neuroimmunologic diseases (ONID) after treatment with rituximab (RTX)/ocrelizumab (OCR) 12 months before or during pregnancy. Methods Data were collected in the German MS and pregnancy registry and centers from the Neuromyelitis Optica Study Group. Sixty-eight known outcomes of 88 pregnancies from 81 women (64 MS, 10 NMOSD, and 7 ONID) were included and stratified in 3 exposure groups: >6M-group = RTX/OCR >6 but ≤12 months before the last menstrual period (LMP) (n = 8); <6M group = RTX/OCR <6 months before the LMP (n = 47); preg group = RTX/OCR after the LMP (n = 13). Results Pregnancy outcomes were similar between groups, but significantly more preterm births (9.8% vs 45%) occurred after exposure during pregnancy. Overall, 2 major congenital abnormalities (3.3%), both in the preg group, were observed. Three women had severe infections during pregnancy. All women with MS (35) and 12/13 women with NMOSD, RTX/OCR exposure before the LMP and known pregnancy outcomes after gestational week 22 were relapse free during pregnancy. Five of 29 (17.2%) women with relapsing-remitting MS (RRMS) and 1 of 12 (8.3%) with NMOSD and at least 6 months postpartum follow-up experienced a relapse postpartum. Duration of RTX/OCR and early retreatment but not detection of B-cells were possible predictors for postpartum relapses in patients with RRMS/NMOSD. Conclusions Although RTX/OCR might be an interesting option for women with RRMS/NMOSD who plan to become pregnant to control DA, more data on pregnancy outcomes and rare risks are needed.
Article
Full-text available
Aims Ocrelizumab is a humanized monoclonal antibody that selectively targets CD20‐positive B cells and is indicated for treatment of patients with relapsing forms of multiple sclerosis (RMS) or primary progressive multiple sclerosis (PPMS). The pharmacokinetics and pharmacodynamics of ocrelizumab in patients with RMS or PPMS were assessed. Methods A population pharmacokinetic model was developed based on data from the Phase II study and the Phase III studies OPERA I and OPERA II in patients with RMS. Data from the ORATORIO Phase III study in patients with PPMS became available after model finalization and was used for external model evaluation. Results The ocrelizumab serum concentration vs time course was accurately described by a 2‐compartment model with time‐dependent clearance. Body weight was found to be the main covariate. The area under the concentration–time curve over the dosing interval was estimated to be 26% higher for patients with RMS weighing <60 kg and 21% lower for patients weighing >90 kg when compared with the 60–90 kg group. The terminal half‐life of ocrelizumab was estimated as 26 days. The extent of B‐cell depletion in blood, as the pharmacodynamic marker, was greater with increasing ocrelizumab exposure. Conclusion The pharmacokinetics of ocrelizumab was described with pharmacokinetic parameters typical for an immunoglobulin G1 monoclonal antibody, with body weight as the main covariate. The pharmacokinetics and B‐cell depletion in blood were comparable across the RMS and PPMS trials, and the extent of blood B‐cell depletion was greater with higher exposure.
Article
Full-text available
Autoimmune disorders of the central nervous system (CNS) comprise a broad spectrum of clinical entities. The stratification of patients based on the recognized autoantigen is of great importance for therapy optimization and for concepts of pathogenicity, but for most of these patients, the actual target of their autoimmune response is unknown. Here we investigated oligodendrocyte myelin glycoprotein (OMGP) as autoimmune target, because OMGP is expressed specifically in the CNS and there on oligodendrocytes and neurons. Using a stringent cell-based assay, we detected autoantibodies to OMGP in serum of 8/352 patients with multiple sclerosis, 1/28 children with acute disseminated encephalomyelitis and unexpectedly, also in one patient with psychosis, but in none of 114 healthy controls. Since OMGP is GPI-anchored, we validated its recognition also in GPI-anchored form. The autoantibodies to OMGP were largely IgG1 with a contribution of IgG4, indicating cognate T cell help. We found high levels of soluble OMGP in human spinal fluid, presumably due to shedding of the GPI-linked OMGP. Analyzing the pathogenic relevance of autoimmunity to OMGP in an animal model, we found that OMGP-specific T cells induce a novel type of experimental autoimmune encephalomyelitis dominated by meningitis above the cortical convexities. This unusual localization may be directed by intrathecal uptake and presentation of OMGP by meningeal phagocytes. Together, OMGP-directed autoimmunity provides a new element of heterogeneity, helping to improve the stratification of patients for diagnostic and therapeutic purposes.
Article
Full-text available
Until recently, in the pathogenesis of Multiple Sclerosis (MS), the contribution of B cells has been largely underestimated, and the disease was considered a T-cell-mediated disorder. However, newer evidence shows that B cells play a crucial role in the pathogenesis of MS via antigen-driven autoantibody responses and through the cross regulation of T-helper cells. As B cells express the surface molecule CD20 at all points of differentiation, it provides a specific target for monoclonal antibodies, and the development and clinical testing of anti-CD20 antibody treatments for MS have been successful. After some observations, some small clinical trials found positive effects for the first anti-CD20 therapeutic rituximab in MS; newer agents have been specifically evaluated, resulting in the development of ocrelizumab and ofatumumab. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, was approved in March 2017 by the Food and Drug Administration (FDA) and is also the first proven therapy to reduce disability progression in primary progressive MS. This is particularly significant considering that disease-modifying treatment options are few for both primary and secondary progressive MS. Ofatumumab, a fully human anti-CD20 monoclonal antibody, that binds a distinct epitope, has been further investigated in phase 3 trials for relapsing forms of MS. In this review, we discuss in detail these two anti-CD20 agents and their advent for treatment of MS.
Article
Full-text available
The HLA-DR15 haplotype is the strongest genetic risk factor for multiple sclerosis (MS), but our understanding of how it contributes toMSis limited. Because autoreactive CD4+ T cells and B cells as antigen-presenting cells are involved in MS pathogenesis, we characterized the immunopeptidomes of the two HLA-DR15 allomorphs DR2a and DR2b of human primary B cells and monocytes, thymus, and MS brain tissue. Self-peptides from HLA-DR molecules, particularly from DR2a and DR2b themselves, are abundant on B cells and thymic antigen-presenting cells. Furthermore, we identified autoreactive CD4+ T cell clones that can cross-react with HLA-DR-derived self-peptides (HLA-DR-SPs), peptides from MS-associated foreign agents (Epstein-Barr virus and Akkermansia muciniphila), and autoantigens presented by DR2a and DR2b. Thus, both HLA-DR15 allomorphs jointly shape an autoreactive T cell repertoire by serving as antigen-presenting structures and epitope sources and by presenting the same foreign peptides and autoantigens to autoreactive CD4+ T cells in MS.
Article
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system of unknown etiology. We tested the hypothesis that MS is caused by Epstein-Barr virus (EBV) in a cohort comprising more than 10 million young adults on active duty in the US military, 955 of whom were diagnosed with MS during their period of service. Risk of MS increased 32-fold after infection with EBV but was not increased after infection with other viruses, including the similarly transmitted cytomegalovirus. Serum levels of neurofilament light chain, a biomarker of neuroaxonal degeneration, increased only after EBV seroconversion. These findings cannot be explained by any known risk factor for MS and suggest EBV as the leading cause of MS.
Article
By targeting both arms of the immune system, not just B cells, brain-penetrant inhibitors of Bruton’s tyrosine kinase could improve on anti-CD20 therapy for patients with multiple sclerosis.