You are on page 1of 16

666741

research-article2016
TAN0010.1177/1756285616666741Therapeutic Advances in Neurological DisordersS Bittner, T Ruck

Therapeutic Advances in Neurological Disorders Review

Targeting B cells in relapsingremitting


Ther Adv Neurol Disord

2017, Vol. 10(1) 5166

multiple sclerosis: from pathophysiology DOI: 10.1177/


1756285616666741

to optimal clinical management


The Author(s), 2016.
Reprints and permissions:
http://www.sagepub.co.uk/
journalsPermissions.nav

Stefan Bittner*, Tobias Ruck*, Heinz Wiendl, Oliver M. Grauer and Sven G. Meuth

Abstract: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease that


is caused by an autoimmune response against central nervous system (CNS) structures.
Traditionally considered a T-cell-mediated disorder, the contribution of B cells to the
pathogenesis of MS has long been debated. Based on recent promising clinical results from
CD20-depleting strategies by three therapeutic monoclonal antibodies in clinical phase II
and III trials (rituximab, ocrelizumab and ofatumumab), targeting B cells in MS is currently
attracting growing interest among basic researchers and clinicians. Many questions about
the role of B and plasma cells in MS remain still unanswered, ranging from the role of
specific B-cell subsets and functions to the optimal treatment regimen of B-cell depletion
and monitoring thereafter. Here, we will assess our current knowledge of the mechanisms
implicating B cells in multiple steps of disease pathology and examine current and future
therapeutic approaches for the treatment of MS.

Keywords: B lymphocytes, dosage regime, monitoring, multiple sclerosis, ocrelizumab,


ofatumumab, rituximab

Introduction different stages of MS pathology still remain Correspondence to:


Stefan Bittner, MD
Multiple sclerosis (MS) is a chronic inflammatory largely unclear. Several currently approved thera- Department of Neurology,
demyelinating disease of the central nervous sys- pies for MS have at least a partial effect on B cells. University of Mainz, Mainz,
Germany
tem (CNS). The complex pathogenesis of MS is Novel therapies addressing B cells either make use stefan.bittner@
still incompletely understood and, while MS has of anti-cell surface receptor directed antibodies unimedizin-mainz.de
long been considered a classical T-cell-mediated resulting in cell depletion or aim at B-cell signal- Sven G. Meuth, MD, PhD
Department of Neurology,
autoimmune disorder, intensive research has ing pathways. Three different monoclonal anti- University of Mnster,
implicated an involvement of nearly all cell types bodies against CD20-positive B cells (rituximab, Mnster, Germany
sven.meuth@ukmuenster.
of the immune system and CNS [Hafler et al. ocrelizumab and ofatumumab) have shown over- de
2005]. A role of the humoral immune system was all promising effects in clinical phase II and III tri- Tobias Ruck, MD
suggested based on data from histological stain- als. CD20 is not expressed in haematopoietic stem Heinz Wiendl, MD
Oliver M. Grauer, MD, PhD
ings of MS lesions and the presence of intrathecal cell and plasma cells. B-cell-depleting antibodies Department of Neurology,
immunoglobulin production [OBrien etal. 2010]. have a differential effect on different B-cell sub- University of Mnster,
Mnster, Germany
However, T cells have traditionally dominated our sets and recovery after depletion determines not *equal contribution
view of MS pathophysiology based on data from only treatment efficacy, but also treatment dura-
animal models, mainly experimental autoimmune tion and side effects. Therefore, the dosage and
encephalomyelitis (EAE), that biased pathoge- mode of application, treatment intervals and
netic concepts towards T helper cells. The contri- monitoring strategies are critical factors that
bution of the B cells to MS pathology has been may determine the therapeutic success of B-cell-
reassessed in recent years due to new findings targeting approaches. Here, we discuss the patho-
from basic research and pivotal case reports on the physiological rationale of targeting B cells and
beneficial effect of B-cell-depleting therapies summarize current therapeutic approaches with a
[Monson etal. 2005; Stuve etal. 2005]. The pre- special focus on clinical administration regimens
cise mechanisms by which B cells are involved in and monitoring strategies.

http://tan.sagepub.com 51
Therapeutic Advances in Neurological Disorders 10(1)

Evidence for B-cell involvement in the protein (MBP), myelin oligodendrocyte glycopro-
pathophysiology of MS tein (MOG) or myelin-associated glycoprotein
There is growing evidence of an additional involve- (MAG)], no single predominant antigen structure
ment of humoral immunity in MS pathogenesis: A for autoantibody responses in MS could be estab-
seminal study in 1950 first reported the presence lished (see e.g. [Owens etal. 2009] for details on
of intrathecal immunoglobulin synthesis in MS this topic). A possible interpretation of these
patients [Kabat et al. 1950]. Oligoclonal bands results is that an individual antibody response can
(OCBs) are distinct protein bands that can be be of differing affinity with different antigenic tar-
detected in the immunoglobulin region by isoelec- gets which might change over time by epitope
trofocusing and immunoblot assay. Their pres- spreading. Recently, it was reported that myelin-
ence in cerebrospinal fluid (CSF) but not in serum specific antibodies produced by autoreactive B
shows that synthesis of immunoglobulins has cells after activation in the periphery accumulate
occurred within the CNS. While OCBs are not in antigen-presenting cells in the CNS and signifi-
specific for MS, they are found in nearly 70% of cantly enhanced the activation of invading effector
patients with clinically isolated syndrome and T cells [Flach etal. 2016]. Studies of MS pathol-
nearly 90% of patients with clinically definite MS ogy have shown the presence of ectopic lymphoid
[Boster etal. 2010; Dobson etal. 2013]. The pres- follicles resembling germinal centers containing B
ence of OCBs has been used as a diagnostic tool in cells, T cells and antigen-presenting cells in the
patients with suspected relapsingremitting MS meninges of secondary progressive MS [Serafini
(RRMS) in the past (McDonald Criteria etal. 2004; Magliozzi etal. 2010] leading to the
2001/2005). While they are not included in the suggestion that meningeal inflammation perpet-
current revised McDonald Criteria 2010 for uated by lymphoid-like structures are a driven
RRMS due to an increasing value of magnetic force of cortical neuronal damage especially in
resonance imaging (MRI) findings, they are still later disease phases [Dendrou etal. 2015; Haider
valid for the diagnosis of primary progressive MS etal. 2016]. These follicles also harbor short-lived
(PPMS) and for differential diagnosis [Polman plasmablasts and plasma cells producing class-
etal. 2011]. Two studies of OCBs in patients with switched immunoglobulins that contribute to
MS demonstrated that the absence of OCBs was the compartmentalization of humoral responses in
associated with a benign disease course while a the CNS [Corcione et al. 2004; Michel et al.
high number of OCBs correlated with a worse dis- 2015].
ease course [Zeman etal. 1996; Villar etal. 2002].
Using microarray approaches to investigate char- Recent studies applying deep sequencing of IgG
acteristics of MS lesions, another study showed heavy chain variable region genes (IgG-VH) in B
that samples from acute lesions displayed signifi- cells from MS patients revealed the ability of bidi-
cantly elevated levels of immunoglobulin tran- rectional B-cell exchange between the CNS and
scripts compared to chronic silent lesions [Lock peripheral immune system [Blauth et al. 2015].
et al. 2002]. Various independent histological Different studies investigated the VH repertoire
analysis of CNS lesions from MS patients revealed in the peripheral blood, cervical lymph nodes,
the presence of B cells, plasma cells and immuno- meninges, CNS parenchyma and CSF [Von
globulins [Esiri, 1977; Prineas and Wright, 1978]. Budingen etal. 2012; Palanichamy etal. 2014a;
Prominent immunoglobulin reactivity with depo- Stern etal. 2014]. A common observation of all
sitions of IgG antibodies and C9neo complement studies were overlapping clonal B-cell popula-
is characteristic for myelin destruction in type II tions common to peripheral and CNS compart-
plaques according to the classification by ments. The patterns suggest that B cells are able
Luccinetti and colleagues [Lucchinetti et al. to travel back and forth across the bloodbrain
2000]. Immunoglobulin stainings associated with barrier and commonly reenter germinal centers
degenerating myelin and myelin degradation (in the meninges or cervical lymph nodes) to
products within macrophages at the active edge of undergo further somatic hypermutations. These
MS plaques argues for an active role in demyeli- findings change our view of lymphocytic surveil-
nating lesions. Data from EAE studies support lance of CNS tissue and underlines that B-cell
the notion that IgG antibodies facilitate contact trafficking is an important topic for future research
between myelin and macrophages leading to and therapy strategies.
myelin phagocytosis [Epstein etal. 1983; Moore
and Raine, 1988]. Despite continuous research The three putative biological roles of B cells are
effort on putative CNS antigens [e.g. myelin basic antibody production, antigen presentation and

52 http://tan.sagepub.com
S Bittner, T Ruck et al.

production of immunoregulatory cytokines. The express more IL-10 and less pro-inflammatory
later has led to the recognition of different B-cell cytokines, including tumor necrosis factor
subtypes producing either proinflammatory or (TNF)-, IL-6 and GM-CSF when compared
regulatory cytokines (regulatory B cells and B with pretreatment B cells [Pers et al. 2008; Li
effector cells). Abnormal antibodies and OCBs et al. 2015a]. In line with this, another recent
present in the spinal fluid of MS patients are not study also showed an elevation of IL10-producing
significantly reduced following effective B-cell regulatory B cells after therapy with fingolimod
depletion. Study results therefore indicate that [Grutzke etal. 2015]. However, CD20-mediated
peripheral antibodies titers do not impact relapse B-cell depletion is also associated with an activa-
rate, Expanded Disability Status Scale (EDSS) tion of monocytes/myeloid cells [Lehmann-Horn
changes or MRI parameters on a short-term lev- et al. 2011]. This underlines that a pan-B-cell
els. The effects of B-cell-modulating therapies on depletion might also lead to negative effects as
antibodies in CNS lesions themselves is an regulatory B cells are depleted as well. Further
unknown topic. This is especially relevant in the studies are expected to expand our knowledge on
light of recent studies showing that CNS-reactive the effect of different B-cell-directed therapies on
autoantibodies are capable of initiating encepha- cytokine-producing B cells in MS. Accumulating
litogenic immune responses by opsonization of evidence suggests that CD20 is also expressed on
endogenous antigens. These are then recognized a subset of T cells (CD3+CD20dim T cells), that
by otherwise inactive myeloid antigen-presenting is increased in MS patients. While the patho-
cells [Kinzel etal. 2016]. In the light of newly dis- physiological relevance of CD20-expressing T
covered functional lymphatic vessels lining the cells remains to be determined, a depletion of
dural sinuses [Louveau etal. 2015] and a regular these T cells has been shown under treatment
bidirectional trafficking of B cells between brain with rituximab thus potentially contributing to
and periphery (see above), this study points the overall clinical effect of CD20-depleting ther-
towards novel and relevant B-cell effector path- apies [Palanichamy etal. 2014b].
ways. Recent studies also elaborated the impor-
tance of antibody-independent functions of B B cells can per se act as potent antigen-presenting
cells in MS. In a recent study, a subtype of mem- cells and thereby activate T cells. A role of anti-
ory B cells that produces the proinflammatory gen-presenting B cells in MS pathology has been
cytokine granulocyte macrophage colony-stimu- suggested by some studies [Weber et al. 2010;
lating factor (GM-CSF) was found to be more Brimnes etal. 2014], while the relevance of this
frequent and active in the blood of MS patients mechanism is still far from understood. In sum-
compared with controls [Li etal. 2015b]. These B mary, these studies suggest that B cells play a role
cells were especially able to switch myeloid cells in modulating T-cell function driving MS pathol-
(and subsequently T cells) to a proinflammatory ogy and that antibody binding can directly medi-
phenotype. After depletion with rituximab, ate demyelinating processes within MS lesions.
repopulated B cells showed a reduction in the
number of GM-CSF-producing B cells.
B cells as therapeutic targets in MS:
There is also growing evidence that a subset of currently approved therapies
regulatory B cells (Bregs) that display an immu- While there is much clinical and experimental
noregulatory potential play an important role data for a role of B cells in the pathogenesis of
determining the safety and effectiveness of B-cell MS, a direct effect of MS therapies on B cells
targeting therapies [Shen and Fillatreau, 2015]. would provide further support for this hypothesis.
These Bregs maintain peripheral and central Currently approved therapeutic approaches for
immunological tolerance by secreting immu- MS therapy have mostly broad immune-modula-
noregulatory cytokines, especially interleukin tory effects and follow either a general shaping of
(IL)-10 and IL-35 [Fillatreau et al. 2002; Shen the immune system towards anti-inflammatory
etal. 2014]. IL-10 and IL-35 are predominantly immune responses by multifaceted targets (e.g.
provided by distinct sets of regulatory plasma interferons, glatiramer acetate) or result in a broad
cells, leading to a protective effect in animal depletion of different immune cells (e.g. mitox-
models of MS. Clinical experiences showed that antrone, alemtuzumab). More recently, the field of
Bregs with increased expression of CD38 and neuroimmunotherapy is changing as novel thera-
CD5 are predominately present after rituximab pies aim at a selective modulation of distinct molec-
treatment and these B cells, when activated, ular pathways (e.g. natalizumab). Interestingly,

http://tan.sagepub.com 53
Therapeutic Advances in Neurological Disorders 10(1)

nearly all current MS therapies have the potential inhibits dihydro-orotate dehydrogenase leading
to affect B cells while the contribution of these bio- to a reduction in proliferation of activated T and
logical effects compared with other potential mode B lymphocytes [Bar-Or etal. 2014b]. Finally, tar-
of actions are often difficult to quantify. These dif- geting of CD52 by alemtuzumab selectively
ficulties and the lack of clinically approved B-cell- depletes CD52-bearing immune cells (i.e. T and
specific interventions for MS might have led to B lymphocytes and to a lower degree also mono-
an underestimation of B-cell-specific therapeutic cytes, macrophages and eosinophil granulocytes)
effects (see also Krumbholz et al. [2012] and rapidly after infusion by antibody-dependent and
Longbrake and Cross [2016] for more details). complement-dependent cytolysis. The slow
Treatment with -interferons (IFN) signifi- repopulation of these cell populations is believed
cantly reduces the number of circulating CD80+ to rebalance immunological responses mediating
B cells in patients with active disease correlating long-term beneficial therapeutic effects. B lym-
with clinical amelioration [Genc et al. 1997; phocytes recover early after 3 months showing an
Liu et al. 2001]. IFN therapy also reduces the overshoot to approximately 150% of baseline lev-
expression of CD40, a marker necessary for B-cell els after 12 months. B-cell reconstitution is asso-
differentiation [Liu et al. 2001], as well as the ciated with a shift towards nave B cells and a
expression of MHC II inhibiting the ability of B long-term memory B-cell lymphopenia
cells to stimulate antigen-specific T-cell responses [Thompson etal. 2010; Heidt etal. 2012].
[Jiang et al. 1995]. Glatiramer acetate has been
shown to alter the function of antigen-presenting
cells (including B cells) in order to shift the phe- Preclinical therapies targeting B cells in MS
notype from TH1 to TH2 phenotypes [Neuhaus Some of the most compelling lines of evidence
et al. 2001; Chofflon, 2005] and brain antigen- for B-cell involvement in MS pathogenesis comes
specific B-cell responses have been proposed to from recent clinical trials that may ultimately
correlate with glatiramer acetate response in MS lead to the approval of B-cell-specific therapies in
patients [Rovituso etal. 2015]. Apart from these the near future. Multiple strategies have been
classical broad immunomodulatory drugs, effects proposed for the modulation of B-cell popula-
on B cells have also been shown for novel immu- tions in MS patients (see Figure 1). CD20 is a
notherapeutics. Natalizumab was initially devel- transmembrane molecule that can be found on
oped to block T cell adherence to the inflamed nave and memory B cells, but not on stem cells
bloodbrain barrier via an inhibition of the or fully differentiated plasma cells [Maloney,
molecular interaction between 41-integrin 2012]. Depletion of CD20-expressing B cells is
(VLA-4) on T lymphocytes and VCAM-1 on the common mode of action of rituximab, ocreli-
endothelial cells. Memory B cells also express zumab and ofatumumab. An overview of cur-
high levels VLA-4 and it has been proposed that rently ongoing clinical trials targeting B cells is
natalizumab treatment affects directly the migra- provided in Table 1.
tion of B cells into the CNS. However, B cell
reduction in the CSF might also be a secondary
effect to reduced T cells in the CNS [Alter etal. Rituximab
2003; Stuve et al. 2006]. Fingolimod acts as a Rituximab is a chimeric monoclonal antibody
functional antagonist of the sphingosine-1-phos- leading to depletion of CD20+ B cells. This
phate (S1P) receptor, necessary for lymphocyte established B-cell targeting therapy has been
egress from secondary lymphoid structures. As a approved for the treatment of different lympho-
consequence, nave and central memory T cells mas, rheumatoid arthritis (RA) and antineutro-
are trapped within the lymphatic tissue. However, phil cytoplasmic antibody (ANCA)-associated
a significant drop also of B cells can be observed vasculitis in Europe and has been shown to be
in the peripheral blood [Kowarik et al. 2011]. effective in different additional autoimmune dis-
Additionally, circulating B cells showed sup- orders such as systemic lupus erythematodes or
pressed pro-inflammatory properties with lower Sjgrens syndrome [Tokunaga et al. 2007;
levels of CD80, lower TFN levels and higher lev- Gurcan et al. 2009]. In MS, four clinical trials
els of IL-10 [Miyazaki et al. 2014]. Recently, have so far been completed [Barun and Bar-Or,
these findings have been underlined by the find- 2012]. Two smaller open-label phase I/II
ing of a significant increase in the proportion of trials showed good tolerability and a significant
regulatory B cells upon fingolimod treatment reduction in gadolinium (Gd)-enhanced lesions
[Grutzke et al. 2015]. Teriflunomide selectively [Bar-Or etal. 2008; Naismith etal. 2010]. In a

54 http://tan.sagepub.com
S Bittner, T Ruck et al.

Figure 1. Pathways of B-cell differentiation and characteristic cell surface markers. Targeting of B-cell
subtypes by MEDI-551, rituximab, ofatumumab, ocrelizumab and atacicept are shown. See the text for details
on the therapeutics.

randomized, placebo-controlled phase II trial in shown a depletion in the CSF and in the perivas-
104 patients with RRMS (HERMES trial), cular spaces as revealed by histological stainings
patients treated with rituximab showed a 91% [Martin Mdel et al. 2009]. These findings are
reduction of Gd-enhanced lesions and a reduc- especially relevant in light of the particular rele-
tion of relapse rates by 49% after 48 weeks vance of B cells in meningeal compartments and
[Hauser et al. 2008]. In contrast, a phase II/III B-cell trafficking routes in and out of the CNS
trial in 439 patients with PPMS (OLYMPUS (see above). However, direct evidence from a
trial) failed to show a reduction in confirmed dis- therapeutic effect on MS lesions has so far not
ease progression after 96 weeks [Hawker et al. been reported. Based on the observation of ger-
2009]. However, when looking at subgroups minal center-like structures in the meninges of
of patients younger than 50 years or with progressive MS patients, intrathecal administra-
Gd-enhanced lesions at baseline, there was a sig- tion of low doses of rituximab has been investi-
nificant effect. Despite promising effects in the gated, demonstrating a sustained depletion of
HERMES trial and subgroups in the OLYMPUS peripheral B cells, but only a moderate decline of
trial, no further phase III trials necessary for reg- CSF B cells [Svenningsson etal. 2015]. Further
ulatory approval were launched for rituximab. smaller trials are ongoing focusing on the intrath-
Instead, clinical research focused on newer-gen- ecal administration or the combined systemic and
eration anti-CD20 mAbs ocrelizumab and ofatu- intrathecal use of rituximab [ClinicalTrials.gov
mumab. While these fully humanized antibodies identifier: NCT01212094] (Table 1). Of note,
might prove superior over a chimeric antibody serious adverse events have been reported in
especially concerning safety, tolerability and bio- patients treated with rituximab in different disor-
logical properties, it has been suggested that ders, including cases of progressive multifocal
licensing issues have had an additional influence leukoencephalopathy (PML), a rare viral infec-
on this decision [Buttmann, 2010; Gasperi etal. tion of the CNS caused by JC virus. While no
2016; Milo, 2016]. PML cases have been reported in MS patients
receiving rituximab, the number of individuals
Rituximab treatment has shown not only to affect with MS treated with rituximab is relatively small.
peripheral, but also centrally localized B cells.
Animal studies have shown that anti-CD20 treat-
ment depleted B cells within the CNS of mice Ocrelizumab
with disease systems [Weber et al. 2010]. Ocrelizumab is an anti-CD20 antibody causing
Investigations of rituximab-treated patients have B-cell depletion by complement-dependent lysis

http://tan.sagepub.com 55
Table 1. Currently ongoing or recently terminated clinical trials using B-cell-directed strategies in multiple sclerosis.
Drug Mode of Phase ClinicalTrials. Study rationale Patients Control Primary endpoint Duration Status
action gov identifier group

Rituximab Depletion of 2 NCT00097188 Safety and efficacy of 2 1 g intravenous 104 Placebo Total count of gadolinium- 2 years Hauser etal. [2008]
CD20 B cells rituximab on patients with RRMS enhancing lesions in MRI
2/3 NCT00087529 Efficacy of intravenous repeating doses of 439 Placebo Time to confirmed disease 4 years Hawker etal.
rituximab on patients with PPMS progression (increase in EDSS) [2009]
1/2 NCT01212094 Safety and efficacy of combined systemic and 44 Placebo Brain atrophy progression 2 years Active, not
intrathecal treatment in patients with SPMS recruiting
1 NCT02253264 Safety and efficacy of intrathecal treatment 12 None Number of serious adverse 1 year Recruiting
in patients with progressive MS showing events
meningeal inflammation
2 NCT02545959 Efficacy of intrathecal treatment on CSF 12 Methyl- Change in osteopontin level 1 year Recruiting
markers in patients with progressive MS prednisolone in CSF
2 NCT01719159 Safety and efficacy of intrathecal treatment 30 None Number of participants with 1 year Recruiting
Therapeutic Advances in Neurological Disorders 10(1)

in patients with progressive MS adverse events


Ocrelizumab Depletion of 2 NCT00676715 Dose finding study to evaluate the efficacy 220 Placebo, Total count of gadolinium- 24 weeks Kappos etal.
CD20 B cells and safety of 2 dose regimens of ocrelizumab IFN- 1a i.m. enhancing lesions in MRI [2011]
in patients with RRMS
3 NCT01412333 Efficacy and safety of ocrelizumab in patients 835 IFN- 1a s.c. Annualized relapse rate by 2 96 weeks Completed, results
with RRMS years not published yet
3 NCT01247324 Efficacy and safety of ocrelizumab in patients 821 IFN- 1a s.c. Annualized relapse rate by 2 96 weeks Completed, results
with RRMS years not published yet
3 NCT01194570 Efficacy and safety of ocrelizumab in patients 732 Placebo Time to onset of sustained Up to 5.5 Completed, results
with PPMS disability progression years not published yet
Ofatumumab Depletion of 2 NCT00640328 Safety of escalating doses of intravenous 38 Placebo Safety 24 weeks Sorensen etal.
CD20 B cells ofatumumab in patients with RRMS [2014]
2 NCT01457924 Safety and efficacy of subcutaneous doses of 232 Placebo Cumulative number of new 24 weeks Completed, results
ofatumumab in patients with RRMS Gd-enhanced lesions in cMRI not published yet
(up to week 12)
MEDI-551 Depletion of 1 NCT01585766 Safety and tolerability of i.v. and s.c. MEDI- 56 None Safety and tolerability 170 days Ongoing, not
CD19 B cells 551 in patients with RRMS recruiting
VAY736 Blockade of 2 NCT02038049 Safety, tolerability and efficacy of VAY736 Unknown None Cumulative number of new 16 weeks Finished, awaiting
BAFF-R Gd-enhanced lesions results.

Literature search was performed on www.clinicaltrials.gov in September 2015. All published, finished (but unpublished) and continuing studies are listed.
CSF, cerebrospinal fluid; EDSS, Expanded Disability Status Scale; Gd, gadolinium; MS, multiple sclerosis; PPMS, primary progressive multiple sclerosis; RRMS, relapsingremitting multiple sclerosis; SPMS,
secondary progressive multiple sclerosis.

56 http://tan.sagepub.com
S Bittner, T Ruck et al.

and antibody-dependent cytotoxicity [Gasperi Higher rates of upper respiratory tract and naso-
etal. 2016]. In contrast to rituximab, a chimeric pharyngitis infections were observed (approxi-
antibody, ocrelizumab is a humanized IgG1 mately 15% versus 10%) but especially no
antibody with decreased immunogenicity. opportunistic infections were reported. However,
Ocrelizumab binds to a different but overlapping PML has been reported in patients treated with
epitope than rituximab. Compared with rituxi- anti-CD20 antibodies for other indications. In
mab in vitro, ocrelizumab demonstrated increased contrast to experiences from alemtuzumab
antibody-dependent cellular cytotoxicity and (depleting B and T lymphocytes), no cases of
reduced complement-dependent cytotoxicity, secondary autoimmunity were reported so far.
which might additionally lead to reduced immu-
nogenicity and increased tolerability [Oflazoglu In parallel, the efficacy of ocrelizumab was
and Audoly, 2010]. Development of ocrelizumab tested in a placebo-controlled phase III trial
in RA and systemic lupus erythematosus (SLE) (ORATORIO) in patients with PPMS. A total of
were terminated since phase III trials did not 488 patients received 600 mg ocrelizumab every
demonstrate additional benefit of ocrelizumab 24 weeks compared with placebo (244 patients)
over existing therapies [Mysler etal. 2013; Emery and primary endpoint was confirmed disability
etal. 2014]. Serious infects were more frequent in progression after 12 weeks of treatment. Secondary
both RA and SLE trials, however, ocrelizumab endpoints included confirmed disability progres-
was used as a combination therapy together with sion after 24 weeks of treatment, timed walked
methotrexate or glucocorticoids and mycopheno- test, T2-weighted lesion volume and loss of total
late mofetil. brain volume. Ocrelizumab treatment resulted in
a 24% lower risk for confirmed disability progres-
For MS, ocrelizumab has been used as a mono- sion after 12 weeks and a 25% lower risk after 24
therapy. A phase II randomized, placebo-con- weeks. At 120 weeks, patients had a 29% reduc-
trolled trial in patients with RRMS showed a tion in walking time. T2-weighted lesion volumes
significant reduction of annualized relapse rates were 3.4% lower than at baseline and brain vol-
(ARRs) and Gd-enhanced lesions in the ocreli- ume loss was 17.5% lower than in patients who
zumab groups compared to both placebo and received placebo. Ocrelizumab was therefore the
IFNb-1a treatment [Kappos etal. 2011]. Due to first treatment ever to show clinical efficacy in
one death due to acute-onset thrombotic micro- patients with PPMS. However, the occurrence of
angiopathy in the higher dose arm (2000 mg i.v.), side effects in the ORATORIO trial has raised
following phase III trials focused on the lower- questions about its safety profile. Specifically, the
dose regimen (600 mg i.v.). Results of two phase frequency of malignancies was higher in patients
III trials (OPERA I and II [ClinicalTrials.gov treated with ocrelizumab compared with placebo
identifiers: NCT01247324 and NCT01412333]) (11 versus 2). If basal cell carcinomas were
in RRMS patients and one phase III trial excluded (as they are sometimes considered not to
(ORATORIA [ClinicalTrials.gov identifier: be malignancies), the rate was still 8 to 1. This is
NCT01194570]) in PPMS patients were recently considerably higher than in pooled OPERAI/II
reported at the ECTRIMS congress 2015 data (2 versus 4 malignancies) for yet unknown
[Sorensen and Blinkenberg, 2016]. In the OPERA reasons. When looking at the type of malignancy,
I and II trials, 600 mg ocrelizumab i.v. adminis- breast cancer occurred altogether in 6 ocrelizumab
tered every 24 weeks to 1656 patients led to a sig- patients versus 0 in placebo groups. It is so far
nificant reduction of ARRs (~47%) compared unclear, whether there is indeed a link between
with IFN-1a treatment after 96 weeks. ocrelizumab and an increased risk for malignan-
Ocrelizumab also reduced the risk of confirmed cies which would also not have been suspected
disability progression by 40% after 12 and 24 from experiences with rituximab.
weeks of treatment. Moreover, a drastic effect
was observed for reduction of Gd-enhanced T1 Of note, different clinical trials in patients with
lesions by 9495%. Most common adverse events PPMS have been shown to vary concerning the
associated with ocrelizumab were infusion-related proportion of patients with Gd-enhanced lesions
reactions in 34% versus 10%. Most infusion- at baseline ranging from 14.1% in the PROMiSe
related reactions were mild-to-moderate in sever- trial (glatiramer acetate) to 24.5% in the
ity and decreased with number of treatment OLYMPUS trial (rituximab) and 27.5% in the
cycles. In the OPERAI/II trials, serious adverse ORATORIA trial raising concerns about a puta-
events were comparable with the IFN-1a group. tive bias towards more inflammatory active

http://tan.sagepub.com 57
Therapeutic Advances in Neurological Disorders 10(1)

patients. While in the ORATORIA trial, the safe and effective. CD20 is detected on pre-B
number of patients with Gd-enhanced lesions cells, immature B cells, nave B cells and mature
was quite comparable in the placebo versus ocreli- B cells. However, both plasmablasts and plasma
zumab arms (24.7% versus 27.5%), the actual cells are spared by these therapeutic approaches.
number of Gd-enhanced lesions was twice as high Therefore, the depletion of CD19-expressing
in the active arm as in placebo (0.6 versus 1.2). cells might offer potential advantages with regard
Nevertheless, a subgroup analysis presented at to efficacy by additionally depleting antibody-
the ACTRIMS congress (02/2016) showed that producing plasmablasts, but potentially with a
ocrelizumab showed no significant differences in higher risk of infections. Mature plasma cells are
efficacy in patients with and without T1 not directly affected by anti-CD19 treatment
Gd-enhanced lesions at baseline. However, as the [Halliley et al. 2015]. The most advanced com-
study was not powered to show statistical signifi- pound in clinical trials targeting CD19 is MEDI-
cance between subgroups, no definite conclu- 551, a humanized monoclonal antibody.
sions can be drawn as to which patients might MEDI-551 is currently been tested in a phase I
benefit most from ocrelizumab treatment. trial and no results have been released so far
[ClinicalTrials.gov identifier: NCT01585766].

Ofatumumab
Ofatumumab is a fully humanized anti-CD20 Atacicept and VAY736
antibody binding to a different epitope than rituxi- Atacicept is a human recombinant fusion protein
mab (and ocrelizumab) resulting in a pronounced with an Ig Fc domain that combines the binding
complement-mediated cytotoxicity in vitro [Klotz portions of receptors for both BLyS/BAFF
and Wiendl, 2013]. Ofatumumab has a higher (B-Lymphocyte Stimulator or B-Cell activating
binding affinity to CD20 compared with rituxi- Factor of the TNF Family) and APRIL (A
mab and it binds to an additional antigenic deter- Proliferation-Inducing Ligand). BAFF and
minant. It is currently approved for the treatment APRIL are two TNF family ligands regulating B
of chronic lymphatic leukemia. However, whether lymphocyte and plasma-cell survival, antibody
this differences translate into distinct clinical dif- isotype switching, antibody responses and T-cell
ferences in patients with MS is currently unknown costimulation via binding to different receptors
[Zhang, 2009]. In a placebo-controlled phase I/II (see e.g. Schneider [2005] for a detailed review
trial in 38 patients with RRMS treated with on this topic). Atacicept binds soluble BAFF
two intravenous administrations of 100, 300 or and APRIL molecules thereby preferentially
700 mg ofatumumab, a significant reduction of impairing mature B cells and plasma cells with
Gd-enhanced lesions and new and enlarged T2 less impact on progenitor cells memory cells
lesions was observed after 24 weeks in all doses [Edwards and Cambridge, 2006]. Treatment
[Sorensen etal. 2014]. Treatment was not associ- with this blocking antibody causes the arrest of
ated with any unexpected safety concerns and was splenic B-cell development at the immature tran-
generally well tolerated. A subsequent phase II sitional T1 stage [Boster et al. 2010] and sup-
placebo-controlled in 232 patients (MIRROR presses the production of autoreactive antibodies
trial) assessed the efficacy of subcutaneously in mouse models of RA and SLE. However, a
administrated ofatumumab (3, 30 and 60 mg phase II trial in patients with MS who received
every 4 weeks) over 6 months [ClinicalTrials.gov weekly subcutaneous doses of atacicept (25, 75 or
identifier NCT01457924]. The cumulative num- 150 mg, ATAMS study) was terminated due to
ber of new T1 Gd-enhanced lesions was reduced an increase of clinical disease activity [Kappos
by 65%. A dose-dependent CD19 B-cell deple- etal. 2014]. ARRs more than doubled in all ataci-
tion, reconstitution rate and effect on MRI activity cept groups compared to placebo (ARR placebo:
was seen across regimens [Bar-Or et al. 2014a]. 0.38; ARR atacicept: 0.790.98). Of note, MRI
Phase III trials have been announced but no findings did not reflect these differences as mean
further details are available yet. numbers of Gd-enhanced T1 lesions per scan
were similar in all groups. The differences
between atacicept and anti-CD20 drugs points
MEDI-551 towards a complex role of B cells in the patho-
In MS and neuromyelitis optica (NMO), there physiology of MS. An additional expression for
are substantial data from clinical trials that B-cell BAFF and APRIL has for example been described
depletion with anti-CD20 antibodies is relatively not only on B cells but also on T cells. One other

58 http://tan.sagepub.com
S Bittner, T Ruck et al.

possible explanation might have to do with the additional reduction of Gd-enhanced brain lesions
specific B-cell subsets preferentially targeted by [Naismith et al. 2010]. The RIVITALISE trial
atacicept. It has been hypothesized that atacicept [ClinicalTrials.gov identifier: NCT01212094]
might preferentially intervene with regulatory evaluated the efficacy of intrathecally adminis-
B-cell subtypes possessing high expression levels tered rituximab compared with placebo in SPMS
of APRIL [Stuve etal. 2014]. following the hypothesis of compartmentalized
inflammation in the progressive phase of MS. 2%
Another current approach is VAY736, a human of serum concentrations were measured in the
monoclonal antibody targeting BAFF-receptor, CSF representing a 20-fold increase in bioavaila-
that is currently been tested in a phase II trial bility of serum values achievable without intrathe-
[ClinicalTrials.gov identifier: NCT02038049]. cal dosing [Komori etal. 2016]. However, the trial
Results are awaited for 2016. was prematurely terminated due to not meeting
efficacy endpoints during the interim analysis. In
contrast to commonly applied regimens, the same
Dosing of B-cell-depleting therapies: study combined only 200 mg i.v. rituximab on day
experiences from rituximab 1 and day 15 with intrathecally administered
Experiences from B-cell-depleting therapies have rituximab. The authors demonstrated that higher
underlined the importance of an optimal treat- rituximab concentrations paradoxically decreased
ment regimen to balance clinical safety and effi- the rituximab concentration on the surface of B
cacy. Phase III trials for ocrelizumab used slightly cells, due to facilitated internalization of CD20/
different regimens: for OPERA I/II, 600 mg i.v. rituximab complexes in vitro [Komori etal. 2016].
were applied every 6 months (first cycle: twice 300 Therefore, the 200 mg dose was considered suffi-
mg i.v. 2 weeks apart, then single dose 600 mg). cient to saturate CD20 leading to long-lasting
For ORATORIA, a continued splitting of the dos- depletion of B cells from peripheral circulation
age in 300 mg i.v., 2 weeks apart, was chosen for [Komori et al. 2016]. However, greater surface
every 6-month cycle. It is however unclear, binding does not mean more-efficient depletion
whether this treatment regimen is optimal and and the study did not investigate B-cell depletion
whether individual patients responses should be in vivo for the different rituximab dosages to allow
taken into account. Available studies from closely definite conclusions. Nevertheless, the duration of
related rituximab in MS and other indications depletion is dose-dependent as shown in a study
(e.g. NMOSD) might help to give further insights. by Greenberg and colleagues investigating B-cell
repopulation dynamics in NMO and MS patients.
Rituximab is currently approved for the treatment In patients given 100 mg twice the CD19 positive
of CD20-positive non-Hodgkin lymphomas, B-cell population was greater than 2% in a mean
CD20-positive chronic lymphocytic leukemia of 99 days (range 43172), for the 1000 mg dose
(CLL), RA and ANCA-associated systemic vas- in 184 days (range 52288) [Greenberg et al.
culitis; however its off-label use extends to various 2012]. Higher dosages might be able to penetrate
other autoimmune disorders. In MS it is used B-cell-containing secondary lymphoid tissues in
more often than expected for an unapproved drug. higher magnitude and therefore may be critical to
The Swedish register of neurological diseases allo- delay reconstitution.
cates 14% of MS therapies in adults to rituximab
(August 2015, see http://www.neuroreg.se) [Salzer In RA several trials and cohort studies suggested
etal. 2016]. Alongside the variety of indications, comparable efficacy of high-dose (1000 mg
there are many different dosing and application twice) and low-dose (500 mg twice or 1000 mg
strategies. In MS rituximab is commonly adminis- once) rituximab treatment. A meta-analysis of
tered intravenously either at a dose of 1000 mg on four randomized trials showed non-inferiority
days 1 and 15, or 375 mg/m2 in 4-weekly doses of the low dose for the most composite measures
every 612 months [Kim etal. 2013; Melzer and of disease activity and patient-reported out-
Meuth, 2014]. Concomitant steroids, antihista- comes. However, some trends were found for
minic and antipyretic drugs are used to reduce long-term clinical outcomes and early withdrawal
infusion-associated adverse reactions. Apart from due to lack of efficacy favoring high-dose rituxi-
the usage as monotherapy, rituximab was also mab [Bredemeier etal. 2014]. In immune throm-
tested as add-on therapy (4-weekly doses of bocytopenia a multicenter retrospective study
375 mg/m2, added on to IFNs or glatiramer ace- compared rituximab 1000 mg on days 1 and 15
tate) in patients with RRMS demonstrating an with the standard regimen of 4-weekly doses of

http://tan.sagepub.com 59
Therapeutic Advances in Neurological Disorders 10(1)

375 mg/m including 107 patients. A total of the last rituximab infusion [Greenberg et al.
22/61 patients (36%) treated with the standard 2012]. Interestingly, patients with early repopu-
regimen and 23/46 (50%) with the 1000 mg lation did not always show the same repopulation
twice dosing reached the definition for treatment dynamic in repeated infusion courses emphasiz-
response without being significantly different ing the necessity of individual monitoring strate-
[Mahevas etal. 2013]. So far there have been no gies. Greenberg and colleagues suggested
randomized controlled trials in MS comparing monitoring of the B-cell population by analyzing
efficacy and safety of the different dosing strate- CD19 positive cell counts as it has been estab-
gies. To the current knowledge both commonly lished for RA before [Dass etal. 2008]. In con-
administered dosing regimens of rituximab seem trast to previously defined limits of B-cell
to be feasible to induce long-term B-cell deple- depletion with CD19 counts below 0.01*109/l,
tion in MS. Higher doses of rituximab could be this study set a CD19 percentage of 2% of all
associated with more prolonged B-cell depletion. peripheral blood mononuclear cells (PBMCs) as
However, the individual variety in magnitude cut-off for retreatment, which was based on the
and duration of B-cell depletion demonstrates observation of a significant rise of CD19 counts
the importance of a close monitoring in B-cell- when reaching this limit [Pellkofer etal. 2011]. A
targeted therapies. subsequent study by Kim and colleagues found
that CD19 counts as appropriate monitoring tool
for rituximab therapy. In 30 NMO patients 65%
Monitoring of rituximab therapy of relapses or 60% respectively occurred in
patients with CD19 counts <0.5% or <0.01
Retreatment 109/l of PBMCs [Kim et al. 2013]. In RA the
Rituximab is an expensive treatment with unclear return of disease activity correlated with higher
long-term safety of repeated infusions. numbers of memory B cells [Leandro etal. 2006]
Monitoring strategies guiding when and how to and in NMO their depletion was associated with
retreat patients might help to minimize unneces- clinical response to rituximab [Roll etal. 2008].
sary exposure to the drug and therefore to opti- Therefore, Kim and colleagues suggested evalua-
mize the individual efficacy and safety and also tion of the reemerging CD27+ memory B-cell
allow for significant cost savings. In RA patients frequency to decide on retreatment. PBMCs
are regularly monitored for disease activity using were obtained every 6 weeks throughout the first
different clinical composite measures and rituxi- year, every 8 weeks throughout the second year
mab treatment is adjusted to achieve predefined and every 10 weeks thereafter to evaluate lym-
goals [Emery et al. 2011]. However, in MS the phocytes subsets. Patients were retreated with
clinical and radiological signs of disease activity one additional infusion of rituximab whenever
do not present gradually and are no clearly the frequency of CD27+ memory B cells exceeded
defined tools to continuously monitor disease 0.05% of PBMCs in the first 2 years or 0.1%
activity or treatment effects. Of note, outcome in thereafter. Control disease activity was main-
RA are distinct from MS and rituximab is often tained in almost all patients with longer retreat-
discontinued unless there is evidence of contin- ment intervals after 2 years than during the initial
ued activity and is often used in conjunction with 2-year study (about 8 versus 5 months, respec-
other medications. tively) and the cumulative dose of rituximab was
much lower when compared with a calculated
Therefore, initial treatment strategies in NMO or fixed treatment interval of every 6 months. Of
MS followed uncontrolled fixed intervals. The note, the reemergence of CD27+ memory B cells
every-6-month rituximab dosing strategy was was not necessarily proportional to the reconsti-
based on data suggesting that most patients show tution of CD19+ B cells and might exceed the
B-cell depletion for approximately 6 months limits even with low CD19 counts [Kim et al.
[Roll et al. 2006; Greenberg et al. 2012]. 2011, 2013]. In a phase II trial of ocrelizumab in
However, there are high interindividual varia- RRMS long-lasting memory B-cell repopulation
tions of B-cell repopulation as stated above and coincided with sustained suppression of MS dis-
therefore these uncontrolled dosing regimens ease activity indicating that memory B-cell counts
might not be adequate to control disease activity might also be a tool to guide retreatment strate-
in many patients. In a cohort study of NMO and gies in MS for B-cell-targeted therapies. Even
MS patients, 5 of 29 (17%) patients demon- after multiple rituximab cycles memory B-cell
strated B-cell repopulation before 6 months after repopulation follows comparable patterns

60 http://tan.sagepub.com
S Bittner, T Ruck et al.

supporting its use in long-term monitoring [Roll Precautions/adverse events


etal. 2015]. However, with analyzing the CD27+ Before rituximab initiation all patients should be
memory B-cell pool relapses can still be observed screened for HBV, HCV and HIV infection. All
in NMO patients during periods where memory vaccines following the respective guidelines
B cells were lower than the therapeutic target as should be completed at least 46 weeks prior to
has been shown in a follow-up study of 100 ritux- therapy start. After rituximab administration
imab-treated NMO patients [Kim et al. 2015] CBC with differential and platelets should be
demanding new and more developed monitoring done in weekly to monthly intervals with higher
strategies in NMO, MS and other indications. frequencies in patients with cytopenias. Frequent
Moreover, there is no randomized clinical trial adverse events are infusion-associated reactions
comparing the different surveillance and fixed- within 24 h after the first infusion and mild to
dose strategies to draw definite conclusions for moderate infections (nasopharyngitis, upper res-
rituximab use. piratory tract infections, urinary tract infections
and sinusitis). Infrequent, but severe adverse
Based on the expression of IgD, IgM and CD27 events are toxic epidermal necrolysis (Lyell syn-
the memory B-cell pool can be further subdivided drome), StevensJohnson syndrome and PML.
into CD27+IgM+IgD+, IgM-only, IgD-only, In RA the PML risk is estimated at 1 per 25,000
CD27+IgMIgD (CD27+, class-switched), and treated individuals [Clifford etal. 2011]. In addi-
CD27 IgMIgD memory B cells (CD27, class- tion, hypogammaglobinemia has been reported in
switched) [Wu et al. 2011]. CD27 memory B rituximab-treated patient cohorts. In a 5-year fol-
cells represent 14% of all peripheral B cells cor- low up of 41 NMO patients 53%, 29%, and 10%
responding to approximately 25% of IgG+ blood had low IgM, IgG, and IgA levels, respectively
B cells and questioning CD27 as exclusive mem- [Kim etal. 2015]. However, infection rates were
ory B-cell marker [Fecteau et al. 2006]. In RA not increased with low IgM or IgG levels. In con-
both the number of CD27+IgD or CD27IgD trast, in a cohort of lymphoma patients 6.6%
class-switched memory B cells has been shown to patients needed treatment with intravenous
correlate with response to rituximab therapy immunoglobulins for recurrent infections [Casulo
[Moller etal. 2009; Tony etal. 2015]. Other tools et al. 2013]. Therefore, monitoring of serum
such as markers for late-stage B-cell lineage plas- immunoglobulin concentrations might be needed
mablasts and fragment c gamma receptor 3A in patients receiving rituximab, along with other
(FCGR3A) gene polymorphisms have been sug- therapies for oncologic indications.
gested to predict efficacy of B-cell-targeted thera-
pies and might help to identify eligible patients
[Owczarczyk etal. 2011; Kim etal. 2015]. Conclusion
Recent clinical trial results showed that antibody-
In MS rituximab was shown to deplete B cells mediated depletion of B cells using anti-CD20
from the CSF [Cross etal. 2006] and might also antibody, significantly reduced new CNS inflam-
be able to diminish B cells in the brain tissue mation and relapses in MS patients and progres-
[Martin Mdel etal. 2009] opening new opportu- sion in those with gradually worsening disease.
nities for monitoring. The evaluation of B-cell- Marketing authorization for ocrelizumab as a
related biomarkers such as IL-6, BAFF (B-cell novel treatment option for MS is expected to be
activating factor), APRIL (A proliferation-induc- pursued in the near future opening up new ques-
ing ligand) and soluble CD21 or repopulation tions for both researchers and clinicians: Which
dynamics of different B cell subsets in the CSF pathways and specific B-cell subtypes exactly
might be more suitable to predict treatment drive disease pathology? How are other parts of
response and the need for retreatment due to the the immune system affected by antigen presenta-
close proximity to the inflammatory responses in tion and release of inflammatory mediators by B
MS. In accordance, rituximab has been shown to cells in the periphery and the CNS compartment?
normalize IL-6 and GM-CSF levels secreted by B What is the optimal dosing strategy and what
cells isolated from RRMS patients in vitro [Barr monitoring parameters do best reflect treatment
et al. 2012; Li et al. 2015]. With the upcoming success and indicate the need for retreatment?
variety of B-cell-targeted therapies for MS new Growing clinical experience will contribute to an
monitoring strategies are urgently needed to assessment of adverse events providing informa-
improve patient selection and guidance and there- tion where to place B-cell-depleting therapies in
fore therapeutic efficacy and safety. the growing armamentarium of MS treatments.

http://tan.sagepub.com 61
Therapeutic Advances in Neurological Disorders 10(1)

Funding its mechanism of action in multiple sclerosis. Drugs


The author(s) received no financial support for 74: 659674.
the research, authorship, and/or publication of Barr, T., Shen, P., Brown, S., Lampropoulou, V.,
this article. Roch, T., Lawrie, S. etal. (2012) B cell depletion
therapy ameliorates autoimmune disease through
Conflict of interest statement ablation of IL-6-producing B cells. J Exp Med 209:
The author(s) declared the following potential 10011010.
conflicts of interest with respect to the research, Barun, B. and Bar-Or, A. (2012) Treatment of
authorship, and/or publication of this article: SB multiple sclerosis with anti-CD20 antibodies. Clin
has received honoraria for lecturing, travel Immunol 142: 3137.
expenses for attending meetings and financial
Blauth, K., Owens, G. and Bennett, J. (2015) The ins
research support from Bayer Schering AG, Biogen
and outs of B cells in multiple sclerosis. Front Immunol
Idec, Merck Serono, Novartis and TEVA. TR has 6: 565.
received travel expenses and financial research
support from Genzyme and has received hono- Boster, A., Ankeny, D. and Racke, M. (2010) The
raria for lecturing from Genzyme, Biogen and potential role of B cell-targeted therapies in multiple
Teva. OG has received honoraria for lecturing and sclerosis. Drugs 70: 23432356.
travel expenses for attending meetings from Roche Bredemeier, M., De Oliveira, F. and Rocha,
and Bristol-Myers-Squibb. HW has received hon- C. (2014) Low- versus high-dose rituximab for
oraria for lecturing, travel expenses for attending rheumatoid arthritis: a systematic review and meta-
meetings from Bayer Health Care, Biogen Idec/ analysis. Arthritis Care Res 66: 228235.
Elan Corporation, Lilly, Lundbeck Merck Serono, Brimnes, M., Hansen, B., Nielsen, L., Dziegiel, M.
Novartis, Sanofi Aventis, and TEVA Neuroscience; and Nielsen, C. (2014) Uptake and presentation of
has served/serves as a consultant for Biogen Idec, myelin basic protein by normal human B cells. PloS
Merck Serono, Novartis Pharma Sanofi-Aventis; one 9: e113388. doi: 10.1371/journal.pone.0113388.
and receives research support from Bayer Schering
Buttmann, M. (2010) Treating multiple sclerosis with
Pharma, Biogen Idec/Elan Corporation, Merck monoclonal antibodies: a 2010 update. Expert Rev
Serono, Novartis, Novo Nordisk and Sanofi- Neurother 10: 791809.
Aventis. SGM has received honoraria for lecturing
and travel expenses for attending meetings and Casulo, C., Maragulia, J. and Zelenetz, A. (2013)
has received financial research support from Incidence of hypogammaglobulinemia in patients
Bayer, Bayer Schering, Biogen Idec, Genzyme, receiving rituximab and the use of intravenous
immunoglobulin for recurrent infections. Clin
Merck Serono, MSD, Novartis, Novo Nordisk,
Lymphoma Myeloma Leuk 13: 106111.
Sanofi-Aventis and Teva.
Chofflon, M. (2005) Mechanisms of action for
treatments in multiple sclerosis: does a heterogeneous
disease demand a multi-targeted therapeutic
References approach? BioDrugs 19: 299308.
Alter, A., Duddy, M., Hebert, S., Biernacki, K., Prat,
A., Antel, J. etal. (2003) Determinants of human B Clifford, D., Ances, B., Costello, C., Rosen-
cell migration across brain endothelial cells. J Immunol Schmidt, S., Andersson, M., Parks, D. etal.
170: 44974505. (2011) Rituximab-associated progressive multifocal
leukoencephalopathy in rheumatoid arthritis. Arch
Bar-Or, A., Calabresi, P., Arnold, D., Markowitz,
Neurol 68: 11561164.
C., Shafer, S., Kasper, L. etal. (2008) Rituximab
in relapsing-remitting multiple sclerosis: a 72-week, Corcione, A., Casazza, S., Ferretti, E., Giunti, D.,
open-label, phase I trial. Ann Neurol 63: 395400. Zappia, E., Pistorio, A. etal. (2004) Recapitulation of
B cell differentiation in the central nervous system of
Bar-Or, A., Grove, R., Austin, A., Tolson,
patients with multiple sclerosis. Proc Natl Acad Sci U S
J., Vanmeter, S., Lewis, E. etal. (2014a) The
A 101: 1106411069.
MIRROR study: a randomized, double-blind,
placebo-controlled, parallel-group, dose-ranging Cross, A., Stark, J., Lauber, J., Ramsbottom, M.
study to investigate the safety and MRI efficacfy of and Lyons, J. (2006) Rituximab reduces B cells and
subcutaneous ofatumumab in subjects with relapsing- T cells in cerebrospinal fluid of multiple sclerosis
remitting multiple sclerosis (RRMS). Neurology patients. J Neuroimmunol 180: 6370.
82(Suppl. 10): I71.007.
Dass, S., Rawstron, A., Vital, E., Henshaw, K.,
Bar-Or, A., Pachner, A., Menguy-Vacheron, F., Mcgonagle, D. and Emery, P. (2008) Highly sensitive
Kaplan, J. and Wiendl, H. (2014b) Teriflunomide and B cell analysis predicts response to rituximab therapy

62 http://tan.sagepub.com
S Bittner, T Ruck et al.

in rheumatoid arthritis. Arthritis Rheum 58: (2012) Rituximab dosing and monitoring strategies in
29932999. neuromyelitis optica patients: creating strategies for
therapeutic success. Mult Scler 18: 10221026.
Dendrou, C., Fugger, L. and Friese, M. (2015)
Immunopathology of multiple sclerosis. Nat Rev Grutzke, B., Hucke, S., Gross, C., Herold, M.,
Immunol 15: 545558. Posevitz-Fejfar, A., Wildemann, B. etal. (2015)
Fingolimod treatment promotes regulatory phenotype
Dobson, R., Ramagopalan, S., Davis, A. and
and function of B cells. Ann Clin Transl Neurol 2:
Giovannoni, G. (2013) Cerebrospinal fluid oligoclonal
119130.
bands in multiple sclerosis and clinically isolated
syndromes: a meta-analysis of prevalence, prognosis Gurcan, H., Keskin, D., Stern, J., Nitzberg, M.,
and effect of latitude. J Neurol Neurosurg Psychiatry 84: Shekhani, H. and Ahmed, A. (2009) A review of the
909914. current use of rituximab in autoimmune diseases. Int
Immunopharmacol 9: 1025.
Edwards, J. and Cambridge, G. (2006) B-cell
targeting in rheumatoid arthritis and other Hafler, D., Slavik, J., Anderson, D., Oconnor, K.,
autoimmune diseases. Nat Rev Immunol 6: 394403. De Jager, P. and Baecher-Allan, C. (2005) Multiple
sclerosis. Immunol Rev 204: 208231.
Emery, P., Mease, P., Rubbert-Roth, A., Curtis,
J., Muller-Ladner, U., Gaylis, N. etal. (2011) Haider, L., Zrzavy, T., Hametner, S., Hoftberger, R.,
Retreatment with rituximab based on a treatment- Bagnato, F., Grabner, G. etal. (2016) The topograpy
to-target approach provides better disease control of demyelination and neurodegeneration in the
than treatment as needed in patients with rheumatoid multiple sclerosis brain. Brain 139: 807815.
arthritis: a retrospective pooled analysis. Rheumatology
Halliley, J., Tipton, C., Liesveld, J., Rosenberg, A.,
50: 22232232.
Darce, J., Gregoretti, I. etal. (2015) Long-lived
Emery, P., Rigby, W., Tak, P., Dorner, T., Olech, plasma cells are contained within the CD19(-)
E., Martin, C. etal. (2014) Safety with ocrelizumab Cd38(hi)Cd138(+) subset in human bone marrow.
in rheumatoid arthritis: results from the ocrelizumab Immunity 43: 132145.
phase III program. PloS one 9: e87379. doi: 10.1371/
Hauser, S., Waubant, E., Arnold, D., Vollmer, T.,
journal.pone.0087379.
Antel, J., Fox, R. etal. (2008) B-cell depletion with
Epstein, L., Prineas, J. and Raine, C. (1983) rituximab in relapsing-remitting multiple sclerosis. N
Attachment of myelin to coated pits on macrophages Engl J Med 358: 676688.
in experimental allergic encephalomyelitis. J Neurol
Hawker, K., OConnor, P., Freedman, M.,
Sci 61: 341348.
Calabresi, P., Antel, J., Simon, J. etal. (2009)
Esiri, M. (1977) Immunoglobulin-containing cells in Rituximab in patients with primary progressive
multiple-sclerosis plaques. Lancet 2: 478. multiple sclerosis: results of a randomized double-
blind placebo-controlled multicenter trial. Ann
Fecteau, J., Cote, G. and Neron, S. (2006) A new
Neurol 66: 460471.
memory CD27-IgG+ B cell population in peripheral
blood expressing VH genes with low frequency of Heidt, S., Hester, J., Shankar, S., Friend, P.
somatic mutation. J Immunol 177: 37283736. and Wood, K. (2012) B cell repopulation after
alemtuzumab induction-transient increase in
Fillatreau, S., Sweenie, C., Mcgeachy, M., Gray, D.
transitional B cells and long-term dominance of naive
and Anderton, S. (2002) B cells regulate autoimmunity
B cells. Am J Transplant 12: 17841792.
by provision of IL-10. Nat Immunol 3: 944950.
Jiang, H., Milo, R., Swoveland, P., Johnson, K.,
Flach, A., Litke, T., Strauss, J., Haberl, M., Gomez,
Panitch, H. and Dhib-Jalbut, S. (1995) Interferon
C., Reindl, M. etal. (2016) Autoantibody-boosted
beta-1b reduces interferon gamma-induced antigen-
T-cell reactivation in the target organ triggers
presenting capacity of human glial and B cells. J
manifestation of autoimmune CNS disease. Proc Natl
Neuroimmunol 61: 1725.
Acad Sci U S A 113: 33233328 .
Kabat, E., Freedman, D., Murray, J. and Knaub, V.
Gasperi, C., Stuve, O. and Hemmer, B. (2016) B cell-
(1950) A study of the crystalline albumin, gamma
directed therapies in multiple sclerosis. Neurodegener
globulin and total protein in the cerebrospinal fluid of
Dis Manag 6: 3747.
100 cases of multiple sclerosis and in other diseases.
Genc, K., Dona, D. and Reder, A. (1997) Increased Am J Med Sci 219: 5564.
CD80(+) B cells in active multiple sclerosis and
Kappos, L., Hartung, H., Freedman, M., Boyko,
reversal by interferon beta-1b therapy. J Clin Invest
A., Radu, E., Mikol, D. etal. (2014) Atacicept in
99: 26642671.
multiple sclerosis (ATAMS): a randomised, placebo-
Greenberg, B., Graves, D., Remington, G., controlled, double-blind, phase II trial. Lancet Neurol
Hardeman, P., Mann, M., Karandikar, N. etal. 13: 353363.

http://tan.sagepub.com 63
Therapeutic Advances in Neurological Disorders 10(1)

Kappos, L., Li, D., Calabresi, P., OConnor, P., Li, R., Rezk, A., Miyazaki, Y., Hilgenberg, E.,
Bar-Or, A., Barkhof, F. etal. (2011) Ocrelizumab Touil, H., Shen, P. etal. (2015b) Proinflammatory
in relapsing-remitting multiple sclerosis: a phase II, GM-CSF-producing B cells in multiple sclerosis and
randomised, placebo-controlled, multicentre trial. B cell depletion therapy. Sci Transl Med 7: 310ra166.
Lancet 378: 17791787. doi: 10.1126/scitranslmed.aab4176.
Kim, S., Huh, S., Lee, S., Joung, A. and Kim, H. Liu, Z., Pelfrey, C., Cotleur, A., Lee, J. and Rudick,
(2013) A 5-year follow-up of rituximab treatment in R. (2001) Immunomodulatory effects of interferon
patients with neuromyelitis optica spectrum disorder. beta-1a in multiple sclerosis. J Neuroimmunol 112:
JAMA Neurol 70: 11101117. 153162.
Kim, S., Jeong, I., Hyun, J., Joung, A., Jo, H., Hwang, Lock, C., Hermans, G., Pedotti, R., Brendolan, A.,
S. etal. (2015) Treatment outcomes with rituximab Schadt, E., Garren, H. etal. (2002) Gene-microarray
in 100 patients with neuromyelitis optica: influence of analysis of multiple sclerosis lesions yields new targets
FCGR3A polymorphisms on the therapeutic response validated in autoimmune encephalomyelitis. Nat Med
to rituximab. JAMA Neurol 72: 989995. 8: 500508.

Kim, S., Kim, W., Li, X., Jung, I. and Kim, H. Longbrake, E. and Cross, A. (2016) Effect of multiple
(2011) Repeated treatment with rituximab based on sclerosis disease-modifying therapies on B cells and
the assessment of peripheral circulating memory B humoral immunity. JAMA Neurol 73: 219225.
cells in patients with relapsing neuromyelitis optica Louveau, A., Smirnov, I., Keyes, T., Eccles, J.,
over 2 years. Arch Neurol 68: 14121420. Rouhani, S., Peske, J. etal. (2015) Structural
Kinzel, S., Lehmann-Horn, K., Torke, S., Hausler, and functional features of central nervous system
D., Winkler, A., Stadelmann, C. etal. (2016) Myelin- lymphatic vessels. Nature 523: 337341.
reactive antibodies initiate T cell-mediated CNS Lucchinetti, C., Bruck, W., Parisi, J., Scheithauer,
autoimmune disease by opsonization of endogenous B., Rodriguez, M. and Lassmann, H. (2000)
antigen. Acta Neuropathol 132: 4358. Heterogeneity of multiple sclerosis lesions:
Klotz, L. and Wiendl, H. (2013) Monoclonal implications for the pathogenesis of demyelination.
antibodies in neuroinflammatory diseases. Expert Opin Ann Neurol 47: 707717.
Biol Ther 13: 831846. Magliozzi, R., Howell, O., Reeves, C., Roncaroli, F.,
Komori, M., Lin, Y., Cortese, I., Blake, A., Ohayon, Nicholas, R., Serafini, B. etal. (2010) A gradient of
J., Cherup, J. etal. (2016) Insufficient disease neuronal loss and meningeal inflammation in multiple
inhibition by intrathecal rituximab in progressive sclerosis. Ann Neurol 68: 477493.
multiple sclerosis. Ann Clin Transl Neurol 3: 166 Mahevas, M., Ebbo, M., Audia, S., Bonnotte, B.,
179. Schleinitz, N., Durand, J. etal. (2013) Efficacy and
Kowarik, M., Pellkofer, H., Cepok, S., Korn, T., safety of rituximab given at 1,000 mg on days 1 and
Kumpfel, T., Buck, D. etal. (2011) Differential effects 15 compared to the standard regimen to treat adult
of fingolimod (FTY720) on immune cells in the CSF immune thrombocytopenia. Am J Hematol 88: 858861.
and blood of patients with MS. Neurology 76: 1214 Maloney, D. (2012) Anti-CD20 antibody therapy for
1221. B-cell lymphomas. N Engl J Med 366: 20082016.
Krumbholz, M., Derfuss, T., Hohlfeld, R. and Martin Mdel, P., Cravens, P., Winger, R., Kieseier,
Meinl, E. (2012) B cells and antibodies in multiple B., Cepok, S., Eagar, T. etal. (2009) Depletion of
sclerosis pathogenesis and therapy. Nat Rev Neurol B lymphocytes from cerebral perivascular spaces by
8: 613623. rituximab. Arch Neurol 66: 10161020.
Leandro, M., Cambridge, G., Ehrenstein, M. and Melzer, N. and Meuth, S. (2014) Disease-modifying
Edwards, J. (2006) Reconstitution of peripheral blood therapy in multiple sclerosis and chronic inflammatory
B cells after depletion with rituximab in patients with demyelinating polyradiculoneuropathy: common
rheumatoid arthritis. Arthritis Rheum 54: 613620. and divergent current and future strategies. Clin Exp
Lehmann-Horn, K., Schleich, E., Hertzenberg, D., Immunol 175: 359372.
Hapfelmeier, A., Kumpfel, T., von Bubnoff, N. Michel, L., Touil, H., Pikor, N., Gommerman,
etal. (2011) Anti-CD20 B-cell depletion enhances J., Prat, A. and Bar-Or, A. (2015) B Cells in the
monocyte reactivity in neuroimmunological disorders. multiple sclerosis central nervous system: trafficking
J Neuroinflammation 8: 146. and contribution to CNS-compartmentalized
Li, R., Rezk, A., Healy, L., Muirhead, G., Prat, A., inflammation. Front Immunol 6: 636.
Gommerman, J. etal. (2015a) Cytokine-defined B cell Milo, R. (2016) Therapeutic strategies targeting
responses as therapeutic targets in multiple sclerosis. B-cells in multiple sclerosis. Autoimmun Rev 15:
Front Immunol 6: 626. 714718.

64 http://tan.sagepub.com
S Bittner, T Ruck et al.

Miyazaki, Y., Niino, M., Fukazawa, T., Takahashi, Palanichamy, A., Jahn, S., Nickles, D., Derstine, M.,
E., Nonaka, T., Amino, I. etal. (2014) Suppressed Abounasr, A., Hauser, S. etal. (2014b) Rituximab
pro-inflammatory properties of circulating B cells efficiently depletes increased CD20-expressing T
in patients with multiple sclerosis treated with cells in multiple sclerosis patients. J Immunol 193:
fingolimod, based on altered proportions of B-cell 580586.
subpopulations. Clin Immunol 151: 127135.
Pellkofer, H., Krumbholz, M., Berthele, A., Hemmer,
Moller, B., Aeberli, D., Eggli, S., Fuhrer, M., Vajtai, B., Gerdes, L., Havla, J. etal. (2011) Long-term
I., Vogelin, E. etal. (2009) Class-switched B cells follow-up of patients with neuromyelitis optica after
display response to therapeutic B-cell depletion in repeated therapy with rituximab. Neurology 76:
rheumatoid arthritis. Arthritis Res Ther 11: R62. 13101315.
Monson, N., Cravens, P., Frohman, E., Hawker, Pers, J., Daridon, C., Bendaoud, B., Devauchelle, V.,
K. and Racke, M. (2005) Effect of rituximab on the Berthou, C., Saraux, A. etal. (2008) B-cell depletion
peripheral blood and cerebrospinal fluid B cells in and repopulation in autoimmune diseases. Clin Rev
patients with primary progressive multiple sclerosis. Allergy Immunol 34: 5055.
Arch Neurol 62: 258264.
Polman, C., Reingold, S., Banwell, B., Clanet, M.,
Moore, G. and Raine, C. (1988) Immunogold Cohen, J., Filippi, M. etal. (2011) Diagnostic criteria
localization and analysis of IgG during immune- for multiple sclerosis: 2010 revisions to the McDonald
mediated demyelination. Lab Invest 59: 641648. criteria. Ann Neurol 69: 292302.
Mysler, E., Spindler, A., Guzman, R., Bijl, M., Prineas, J. and Wright, R. (1978) Macrophages,
Jayne, D., Furie, R. etal. (2013) Efficacy and safety lymphocytes, and plasma cells in the perivascular
of ocrelizumab in active proliferative lupus nephritis: compartment in chronic multiple sclerosis. Lab Invest
results from a randomized, double-blind, phase III 38: 409421.
study. Arthritis Rheum 65: 23682379.
Roll, P., Dorner, T. and Tony, H. (2008) Anti-CD20
Naismith, R., Piccio, L., Lyons, J., Lauber, J., therapy in patients with rheumatoid arthritis:
Tutlam, N., Parks, B. etal. (2010) Rituximab predictors of response and B cell subset regeneration
add-on therapy for breakthrough relapsing multiple after repeated treatment. Arthritis Rheum 58: 1566
sclerosis: a 52-week phase II trial. Neurology 74: 1575.
18601867.
Roll, P., Mahmood, Z., Muhammad, K.,
Neuhaus, O., Farina, C., Wekerle, H. and Hohlfeld, Feuchtenberger, M., Dorner, T. and Tony, H. (2015)
R. (2001) Mechanisms of action of glatiramer acetate Long-term repopulation of peripheral B-cell subsets
in multiple sclerosis. Neurology 56: 702708. after single and repeated rituximab infusions in
patients with rheumatoid arthritis. Clin Exp Rheumatol
OBrien, K., Gran, B. and Rostami, A. (2010)
33: 347353.
T-cell based immunotherapy in experimental
autoimmune encephalomyelitis and multiple sclerosis. Roll, P., Palanichamy, A., Kneitz, C., Dorner, T. and
Immunotherapy 2: 99115. Tony, H. (2006) Regeneration of B cell subsets after
transient B cell depletion using anti-CD20 antibodies
Oflazoglu, E. and Audoly, L. (2010) Evolution of
in rheumatoid arthritis. Arthritis Rheum 54: 2377
anti-CD20 monoclonal antibody therapeutics in
2386.
oncology. MAbs 2: 1419.
Rovituso, D., Duffy, C., Schroeter, M., Kaiser,
Owczarczyk, K., Lal, P., Abbas, A., Wolslegel, K.,
C., Kleinschnitz, C., Bayas, A. etal. (2015) The
Holweg, C., Dummer, W. etal. (2011) A plasmablast
brain antigen-specific B cell response correlates
biomarker for nonresponse to antibody therapy to
with glatiramer acetate responsiveness in relapsing-
CD20 in rheumatoid arthritis. Sci Transl Med 3:
remitting multiple sclerosis patients. Sci Rep 5: 14265.
101ra192. doi: 10.1126/scitranslmed.3002432.
doi: 10.1038/srep14265.
Owens, G., Bennett, J., Lassmann, H., OConnor,
Salzer, J., Lycke, J., Wickstrom, R., Naver, H.,
K., Ritchie, A., Shearer, A. etal. (2009) Antibodies
Piehl, F. and Svenningsson, A. (2016) Rituximab
produced by clonally expanded plasma cells in
in paediatric onset multiple sclerosis: a case series.
multiple sclerosis cerebrospinal fluid. Ann Neurol 65:
Journal of Neurology 263: 322326.
639649.
Schneider, P. (2005) The role of APRIL and BAFF
Palanichamy, A., Apeltsin, L., Kuo, T., Sirota, M.,
in lymphocyte activation. Curr Opin Immunol 17:
Wang, S., Pitts, S. etal. (2014a) Immunoglobulin
282289.
class-switched B cells form an active immune
axis between CNS and periphery in multiple Serafini, B., Rosicarelli, B., Magliozzi, R., Stigliano,
sclerosis. Sci Transl Med 6: 248ra106. doi: 10.1126/ E. and Aloisi, F. (2004) Detection of ectopic B-cell
scitranslmed.3008930. follicles with germinal centers in the meninges of

http://tan.sagepub.com 65
Therapeutic Advances in Neurological Disorders 10(1)

patients with secondary progressive multiple sclerosis. Rapid depletion of B lymphocytes by ultra-low-
Brain Pathol 14: 164174. dose rituximab delivered intrathecally. Neurol
Neuroimmunol Neuroinflamm 2: e79. doi: 10.1212/
Shen, P. and Fillatreau, S. (2015) Antibody-
NXI.0000000000000079.
independent functions of B cells: a focus on cytokines.
Nat Rev Immunol 15: 441451. Thompson, S., Jones, J., Cox, A., Compston, D. and
Coles, A. (2010) B-cell reconstitution and BAFF after
Shen, P., Roch, T., Lampropoulou, V., OConnor,
alemtuzumab (Campath-1H) treatment of multiple
R., Stervbo, U., Hilgenberg, E. etal. (2014) IL-35-
sclerosis. J Clin Immunol 30: 99105.
producing B cells are critical regulators of immunity
during autoimmune and infectious diseases. Nature Tokunaga, M., Saito, K., Kawabata, D., Imura, Y.,
507: 366370. Fujii, T., Nakayamada, S. etal. (2007) Efficacy of
rituximab (anti-CD20) for refractory systemic lupus
Sorensen, P. and Blinkenberg, M. (2016) The
erythematosus involving the central nervous system.
potential role for ocrelizumab in the treatment
Ann Rheum Dis 66: 470475.
of multiple sclerosis: current evidence and future
prospects. Ther Adv Neurol Disord 9: 4452. Tony, H., Roll, P., Mei, H., Blumner, E., Straka,
A., Gnuegge, L. etal. (2015) Combination of B cell
Sorensen, P., Lisby, S., Grove, R., Derosier, F.,
biomarkers as independent predictors of response
Shackelford, S., Havrdova, E. etal. (2014) Safety
in patients with rheumatoid arthritis treated with
and efficacy of ofatumumab in relapsing-remitting
rituximab. Clin Exp Rheumatol 33: 887894.
multiple sclerosis: a phase II study. Neurology 82:
573581. Villar, L., Masjuan, J., Gonzalez-Porque, P., Plaza,
J., Sadaba, M., Roldan, E. etal. (2002) Intrathecal
Stern, J., Yaari, G., Vander Heiden, J., Church,
IgM synthesis in neurologic diseases: relationship with
G., Donahue, W., Hintzen, R. etal. (2014) B cells
disability in MS. Neurology 58: 824826.
populating the multiple sclerosis brain mature in
the draining cervical lymph nodes. Sci Transl Med 6: Von Budingen, H., Kuo, T., Sirota, M., Van Belle,
248ra107. doi: 10.1126/scitranslmed.3008879. C., Apeltsin, L., Glanville, J. etal. (2012) B cell
exchange across the blood-brain barrier in multiple
Stuve, O., Cepok, S., Elias, B., Saleh, A., Hartung,
sclerosis. J Clin Invest 122: 45334543.
H., Hemmer, B. etal. (2005) Clinical stabilization and
effective B-lymphocyte depletion in the cerebrospinal Weber, M., Prodhomme, T., Patarroyo, J., Molnarfi,
fluid and peripheral blood of a patient with fulminant N., Karnezis, T., Lehmann-Horn, K. etal. (2010)
relapsing-remitting multiple sclerosis. Arch Neurol 62: B-cell activation influences T-cell polarization and
16201623. outcome of anti-CD20 B-cell depletion in central
nervous system autoimmunity. Ann Neurol 68: 369383.
Stuve, O., Marra, C., Jerome, K., Cook, L., Cravens,
P., Cepok, S. etal. (2006) Immune surveillance in Wu, Y., Kipling, D. and Dunn-Walters, D. (2011)
multiple sclerosis patients treated with natalizumab. The relationship between CD27 negative and positive
Ann Neurol 59: 743747. B cell populations in human peripheral blood. Front
Immunol 2: 81.
Stuve, O., Warnke, C., Deason, K., Stangel, M.,
Kieseier, B., Hartung, H. etal. (2014) CD19 as Zeman, A., Kidd, D., Mclean, B., Kelly, M., Francis,
a molecular target in CNS autoimmunity. Acta D., Miller, D. etal. (1996) A study of oligoclonal
Neuropathol 128: 177190. band negative multiple sclerosis. J Neurol Neurosurg
Visit SAGE journals online
http://tan.sagepub.com Psychiatry 60: 2730.
Svenningsson, A., Bergman, J., Dring, A., Vagberg,
SAGE journals M., Birgander, R., Lindqvist, T. etal. (2015) Zhang, B. (2009) Ofatumumab. MAbs 1: 326331.

66 http://tan.sagepub.com

You might also like