Accumulating evidence has designated B cells as central players in the pathogenesis of immune diseases. In the late 1990s, anti-CD20 monoclonal antibodies were developed for the treatment of B-cell non-Hodgkin lymphomas, offering the opportunity to efficiently deplete the B-cell compartment for therapeutic immunointerventions. Several studies have since established the beneficial effect of this drug on the course of a wide range of immune diseases. However, paradoxically, it has also been reported that rituximab sometimes worsens the symptoms of the very same conditions. The explanation that reconciles such apparently conflicting results has recently emerged from basic studies, which demonstrate that (1) B cells are also endowed with immune-regulatory properties and (2) the opposing contributions of B cells may overlap during the course of the disease. Caution should therefore be exercised when considering B-cell depletion because the therapeutic effect will depend on the relative contributions of the opposing B-cell activities at the time of the drug administration.

Historically, the role of B lymphocytes in the pathogenesis of immune diseases has been associated mainly with their capacity to produce harmful antibodies after differentiation into plasma cells. This conception was based on seminal experiments that demonstrated that the mere transfer of antibodies was sufficient to recapitulate the symptoms of myasthenia gravis, Graves disease, Goodpasture disease, among others.1  In contrast to these diseases, other autoimmune conditions, in which the role of antibodies has not been recognized, have traditionally been termed “T cell–mediated” diseases. Among the latter, type 1 diabetes, in which T lymphocytes are crucial for the destruction of the β islets, has long been considered as archetypal. However, recent investigations in nonobese diabetic mice have shown that more than 50%2  of the lymphocytes infiltrating islets of Langerhans are B cells3  and that these B cells are critically necessary for the development of diabetes.4  Another clue that B cells exert pathogenic roles through “antibody-independent” mechanistic pathways came from genetically modified lupus-prone mice. Although B-cell depletion leads to abrogation of the disease in this model, transgenic mice, whose B cells cannot secrete immunoglobulin, still developed nephritis.5  Thus, in many immune diseases, even including those not driven by antibodies, B cells have been demonstrated to play an essential pathogenic role.

Among the antibody-independent pathogenic roles of B cells, accumulating evidence points to their capacity to present antigen.6  Upon recognition of specific antigen, the B-cell membrane is reorganized resulting in the aggregation of B-cell receptor in an immunologic synapse that functions as a platform for internalization of the complex.7  Internalized antigen is degraded and subsequently exposed on the B-cell surface in association with major histocompatibility complex molecules for presentation to T cells. This surface presentation of antigen, in the presence of various costimulatory molecules, elicits the T-cell assistance required for B-cell maturation, which in turn allows B cells to drive optimal T-cell activation and differentiation into memory subsets8  (Figure 1 left panel). Of note, B cells are endowed with unique properties as antigen-presenting cells because they have an antigen-specific receptor, allowing extraction and presentation of antigen, even if it is membrane tethered9  or present in limiting quantities.10  Furthermore, B cells also have the capacity to clonally expand, thereby becoming the numerically dominant antigen-presenting cells.

Figure 1

Janus-faced B cells. Janus, the Roman god of beginnings and endings, is most often depicted as having 2 faces facing opposite directions. Like him, B cells can either elicit (right panel) or terminate (left panel) an immune response, after their activation by combination of ligation of the B-cell receptor, CD40, and Toll-like receptors (TLRs). (Left panel) B cells present the antigen along with costimulatory signals, leading to the activation and the proliferation of T effectors. In turn, activated T cells provide CD40 ligand (CD40L) for the differentiation of B cells into antibody-producing plasma cells. The cytokines produced by B cells may participate into the polarization of T effectors. Finally, B cells play a critical role for the development of T-cell memory. (Right panel) B cells regulate immune response by provision of IL10 that suppresses the activation and the expansion of T effectors directly, and indirectly through the differentiation of T regulatory cells and the suppression of dendritic cell function.

Figure 1

Janus-faced B cells. Janus, the Roman god of beginnings and endings, is most often depicted as having 2 faces facing opposite directions. Like him, B cells can either elicit (right panel) or terminate (left panel) an immune response, after their activation by combination of ligation of the B-cell receptor, CD40, and Toll-like receptors (TLRs). (Left panel) B cells present the antigen along with costimulatory signals, leading to the activation and the proliferation of T effectors. In turn, activated T cells provide CD40 ligand (CD40L) for the differentiation of B cells into antibody-producing plasma cells. The cytokines produced by B cells may participate into the polarization of T effectors. Finally, B cells play a critical role for the development of T-cell memory. (Right panel) B cells regulate immune response by provision of IL10 that suppresses the activation and the expansion of T effectors directly, and indirectly through the differentiation of T regulatory cells and the suppression of dendritic cell function.

Close modal

In addition to antigen presentation, activated B cells also produce a wide range of cytokines and chemokines that modulate the maturation, migration, and function of other immune effectors.11,12  In particular, B cells have been shown to play a critical role in lymphoid neogenesis,13  that is, the process by which ectopic functional lymphoid structures (“tertiary lymphoid tissues”) appear de novo during chronic inflammation.14,15  Several studies16-18  have demonstrated that tertiary lymphoid tissues are permissive microenvironments for the induction of immune responses and have led to the hypothesis that lymphoid neogenesis may contribute to the exacerbation of a wide range of chronic inflammatory diseases.19 

Given the multiple pathogenic roles attributed to B lymphocytes, therapeutic strategies that aim at depleting this cell population were expected to be beneficial in a wide range of immune diseases.

Pioneer attempts to deplete B cells in the 1980s relied on xenogenic polyclonal antibodies directed against the surface immunoglobulin receptor.20  Finally, it was only in the late 1990s that progress achieved in the treatment of lymphoproliferative disorders offered clinicians the opportunity to efficiently target the B-cell compartment for therapeutic immunointerventions.

CD20 is a transmembrane protein that functions as a Ca-permeable cation channel, whose expression is restricted to B cells from the pre-B-cell to the immunoblast stage.21  Rituximab, a chimeric monoclonal antibody composed of human immunoglobulin G1 kappa antibody with variable regions isolated from a murine anti-CD20 clone (IDEC-2B8),22  was initially introduced for the treatment of B-cell non-Hodgkin lymphomas.23  A single course of rituximab successfully depleted peripheral human B lymphocytes for periods ranging from 3 months to more than 1 year through mechanisms involving Fc- and complement-dependent killing as well as other signals inducing apoptosis24  (Figure 2 top panel). Several studies have highlighted the influence of circulatory dynamics and microenvironment on the efficiency of rituximab depletion.25,26  As a result, the reduction of B-cell numbers after rituximab appears to be less complete in secondary lymphoid tissues than in peripheral blood, and variations in the extent and kinetics of the depletion have been reported among B-cell subsets.1 

Figure 2

Mechanisms of action of anti-CD20 antibodies. (Top panel) Apoptosis of malignant B-cell clones occurs upon cross-linking of rituximab-CD20 complexes in the lipid rafts. This activates signaling pathways involving the Src kinases and their regulatory molecules. Complement-mediated cytolysis involves the ability of anti-CD20 immunoglobulin G1 bound to their antigen to bind C1 and trigger the classical complement pathway. Antibody-dependent cell cytotoxicity requires interaction between the Fc portion of rituximab and appropriate receptors on effector cells. (Bottom panel) Another possible mechanism by which rituximab could promote the destruction of malignant B-cell clones is the restoration of the protective antitumoral response. The destruction of the nonmalignant B cells endowed with immune-regulatory properties could facilitate the development of antitumoral T-cell clones.

Figure 2

Mechanisms of action of anti-CD20 antibodies. (Top panel) Apoptosis of malignant B-cell clones occurs upon cross-linking of rituximab-CD20 complexes in the lipid rafts. This activates signaling pathways involving the Src kinases and their regulatory molecules. Complement-mediated cytolysis involves the ability of anti-CD20 immunoglobulin G1 bound to their antigen to bind C1 and trigger the classical complement pathway. Antibody-dependent cell cytotoxicity requires interaction between the Fc portion of rituximab and appropriate receptors on effector cells. (Bottom panel) Another possible mechanism by which rituximab could promote the destruction of malignant B-cell clones is the restoration of the protective antitumoral response. The destruction of the nonmalignant B cells endowed with immune-regulatory properties could facilitate the development of antitumoral T-cell clones.

Close modal

The most frequent hematologic malignancy is non-Hodgkin lymphoma, 85% of which are of B-cell origin. Rituximab has been approved for the treatment of B-cell non-Hodgkin lymphoma in 1997. Since then, several randomized, phase 3 trials have reported significant survival benefits associated with rituximab, in combination with chemotherapy, in patients with diffuse large B-cell lymphoma and follicular lymphoma. Furthermore, these benefits have been demonstrated for rituximab in combination with a wide variety of chemotherapy regimens, across many patient subtypes, and in different treatment and disease settings (for a recent review please see Hagemeister27 ), leading to the conclusion that anti-CD20 monoclonal antibodies represent one of the most important advance in the treatment of B-cell lymphoma in the past 30 years.

In chronic lymphocytic leukemia, the leukemic counterpart of small lymphocytic lymphoma, the addition of rituximab to fludarabine-based chemotherapy has significantly increased complete response and progression-free survival rates for both untreated and relapsed or refractory chronic lymphocytic leukemia.27 

The association between autoimmune diseases and hematologic malignancies has long been recognized. Although autoimmune conditions associated with hematologic malignancies can affect any organ, they seem to predominantly target blood constituents.28  The pathophysiology of these autoimmune manifestations is complex and remains incompletely understood. Autoimmune anemia, for example, either can result from monoclonal antibodies produced by the lymphoma clone (ie, cold hemagglutinin disease) or can be the consequence of polyclonal autoantibodies produced by the residual nonmalignant B cells in chronic lymphocytic leukemia. Several reports suggest that treatments, in particular chlorambucil and fludarabine, might trigger the onset of autoimmune manifestations.29,30  In contrast, rituximab has been shown to be an effective treatment for chronic lymphocytic leukemia–associated autoimmune hemolytic anemia,28,31  and for mixed cryoglobulinemia and cold agglutinins secondary to non-Hodgkin lymphoma.32 

These observations along with the favorable safety profile of the rituximab in patients treated for hematologic malignancies33  have catalyzed the application of the drug in primary immune diseases. Several clinical studies have since established the beneficial effect of B-cell depletion on the course of a wide range of immune diseases.

Rheumatoid arthritis, a multisystem disorder that predominantly causes inflammation in synovial joints, is the autoimmune condition in which the effect of rituximab has been the most studied.34  In the pivotal phase 2a study, approximately 80% of patients with active rheumatoid arthritis (despite methotrexate treatment) showed clinical benefit after rituximab administration—a percentage similar to the one obtained with anti–tumor necrosis factor agents.35  Furthermore, a recent phase 3 trial has reported that even patients with an inadequate response to anti–tumor necrosis factor agents showed significant improvement after rituximab therapy,36  a result that prompted the Food and Drug Administration to approve the drug for the treatment of refractory rheumatoid arthritis.

In chronic idiopathic thrombocytopenic purpura, an acquired hemorrhagic condition associated with accelerated platelet consumption due to antiplatelet autoantibodies, a phase 2 study has reported a 40% good response rate to rituximab.37 

Based on previous encouraging case reports, a phase 2 controlled clinical trial has recently been conducted in multiple sclerosis. In patients with relapsing-remitting multiple sclerosis, a single course of rituximab reduced inflammatory brain lesions and clinical relapses for 48 weeks.38 

Numerous small, open-label studies and isolated cases have also reported favorable outcomes after rituximab administration in a wide range of autoimmune diseases, including vasculitis,39  autoantibody-associated neuropathies,40  Graves disease,41  myasthenia gravis,42  myositis,43  blistering skin disorders,44,45  mixed cryoglobulinemia,46  thrombotic thrombocytopenic purpura,47  Sjögren syndrome,48  and anti–factor VIII syndrome.49  There are ongoing phase 2 and 3 trials of rituximab for each of these autoimmune disorders.50 

Finally, recent advances in solid organ transplantation have unraveled new mechanisms of allograft damage. Unexpected clusters of CD20+ B cells have been discovered in rejected grafts,18,51  and C4d deposition, indicating classic complement pathway activation, is now routinely seen in refractory rejection.52  Rituximab has therefore emerged as a rational choice for therapy in transplantation to abrogate B cell–mediated events.53,54 

Given the central role of B cells, autoantibodies, and immune complexes in the pathophysiology of systemic lupus erythematosus (SLE), it was widely anticipated that anti-CD20 would be efficient in treating SLE. However, despite the successes reported in some open-label preliminary studies,55,56  rituximab failed to meet its primary and secondary end points in 2 large controlled trials of nonrenal SLE57  and renal lupus nephritis.58  These disappointing results are sometimes attributed to the fact that the highly heterogeneous clinical presentation of SLE makes it difficult to devise quantitative measures of response to therapy. However, the latter justification is an unlikely explanation for either the paradoxical exacerbation of the clinical condition59-61  or the onset of new immune diseases (including psoriasis,62,63  vasculitis,64-66  interstitial pneumonitis,67-69  and autoimmune cytopenia70,71 ) that are sometimes observed after rituximab administration. One could argue that these small nonrandomized studies focus on a very rare adverse effect of rituximab, because this problem was not identified in the huge cohort of patients treated for hematologic malignancies.33  We rather favor the alternative explanation that rituximab-induced autoimmune complications are underreported. It is indeed a well-established fact that the quality and quantity of drug safety reporting are inadequate,72  a matter that seems even more common for drugs with excellent efficacy outcomes.72  One can indeed conceive the reluctance of clinicians to report side effects of a drug that has changed the standard of care for patients suffering from life-threatening diseases such as non-Hodgkin lymphoma or chronic lymphocytic leukemia. Furthermore, the widely recognized association between autoimmune diseases and hematologic malignancies already discussed makes very difficult the formal incrimination of the drug for these patients. The same concern also exists for patients receiving rituximab for an autoimmune disease, the occurrence of distinct autoimmune conditions in the same patients being a very common feature.73  Thus, the current lack of a consistent demonstration of the increased incidence of autoimmune phenomena after rituximab administration does not exclude the possibility that a carefully conducted meta-analysis might still be able to detect this adverse event.

In this context, the results of the study recently published by Clatworthy et al74  are particularly interesting because they clearly establish the “paradoxical” immune stimulatory effect of rituximab. Indeed, this randomized controlled trial, which compared rituximab with an anti-CD25 monoclonal antibody as induction therapy in patients undergoing renal transplantation, had to be suspended because the authors observed a 6-fold increased incidence of acute cellular rejection in the rituximab group (83% vs 14%).

Based on the worsening of autoimmunity observed in chronic lymphocytic leukemia patients receiving fludarabine,29,30  a drug that induces a profound lymphopenia, it is tempting to speculate that this paradoxical immune stimulation is rather a general feature after B-cell depletion than a specific adverse effect of rituximab.

Finally, the ambivalence of B-cell depletion on the course of immune diseases has been recently demonstrated in experimental autoimmune encephalomyelitis.75  Experimental autoimmune encephalomyelitis (EAE) is an autoimmune central nervous system disease that can be induced in certain susceptible murine strains after immunization with myelin to model human multiple sclerosis. Whereas CD20 antibody–mediated B-cell depletion during EAE disease progression dramatically reduced the symptoms, the same treatment given before EAE induction substantially exacerbated the disease (Figure 3). Interestingly, administration of the drug at other time points had no significant effect (Figure 3).

Figure 3

B-cell depletion has ambivalent effects on the course of immune diseases. The figure is a schematic representation of the findings of Matsushita et al.75  During the course of EAE, B cells play opposite overlapping roles. Depending on the timing of anti-CD20 antibody administration, the net clinical effect can be deleterious, neutral, or beneficial. Dashed lines indicate the severity of EAE in untreated control animals.

Figure 3

B-cell depletion has ambivalent effects on the course of immune diseases. The figure is a schematic representation of the findings of Matsushita et al.75  During the course of EAE, B cells play opposite overlapping roles. Depending on the timing of anti-CD20 antibody administration, the net clinical effect can be deleterious, neutral, or beneficial. Dashed lines indicate the severity of EAE in untreated control animals.

Close modal

Altogether, these data demonstrate that it is more difficult than anticipated to forecast the effect of B-cell depletion on the course of an immune disease: the same therapy can lead to opposite outcomes depending on the timing of its administration.65 

The explanation that reconciles such apparently conflicting results has recently emerged from basic studies that demonstrate an immune-regulatory role of B cells.

The first clue that B cells can regulate immune responses was provided by seminal in vivo experimental studies by Shimamura et al, demonstrating that adoptive transfer of antigen-activated B cells could induce tolerance in naive mice through the induction of suppressor T cells.76  Despite these data, the role of B cells in the regulation of immune diseases remained overlooked for another decade, until Wolf et al reported that mice lacking B cells suffered an unusually severe and chronic form of EAE.77  Although spontaneous recovery is the norm in wild-type mice, the authors observed that genetically B cell–deficient mice of the same background experienced a greater variation in disease onset and severity, and failed to recover completely. After this seminal contribution, independent groups made consistent observations in other experimental models of autoimmune diseases, including collagen-induced arthritis78  and a model of spontaneous colitis, pathologically reminiscent of human ulcerative colitis.79  Interestingly, whereas the pathogenic T-cell response involves the same T helper 1 (Th1) cells and Th17 proinflammatory T-cell populations in EAE and collagen-induced arthritis, the colitis model differs in that the inflammation appears to be driven by Th2 cells. Thus the B-cell compartment has the capacity to control organ-specific inflammation that may be driven by Th1, Th2, or Th17 effectors.

Dissection of the underlying mechanisms revealed that B cells limit immune disease progression by providing interleukin-10 (IL-10)75,78,80-82  that, in turn, directly suppresses the differentiation of pathogenic T cells, promotes the development of regulatory T cells,83  and constrains dendritic cell functions84  (Figure 1 right panel).

The recent demonstration that IL-10–producing B cells exist in humans, and that B cells from patients with multiple sclerosis85  and lupus86  produce decreased amounts of IL10, suggests a general role of B cells in clinical immune homeostasis.

B cells can be stimulated to produce IL-10 by a combination of ligation of the B-cell receptor by the antigen and of CD40 by CD40 ligand. Some reports also point to a critical nonredundant role of certain Toll-like receptors (TLR2 and TLR4) in driving the regulatory activity in B cells.87 

The involvement of the B-cell receptor, CD40, and TLRs in the regulatory function of B cells raises a conceptual difficulty. Indeed, these signals are the very same as the ones involved in the activation of B cells in most immune responses. One hypothesis would therefore be the existence of a peculiar “B-reg” subset, endowed with the unique function to regulate immune processes. In the mouse, B cells are classically divided into B-1 cells that reside in pleural and peritoneal cavities and B-2 cells that populate secondary lymphoid organs. Peritoneal B-1 cells, known to produce particularly large amounts of IL10 after stimulation, have been ascribed with regulatory function in some studies.88,89  However, because this subset was excluded from the transfer experiments demonstrating the regulatory role of B cells,78,80,90  it is likely that B-2 cells also contain a subset regulating immune diseases.

Interestingly, a rare population of splenic B cells characterized by a unique phenotype that associates features from B-1 (expression of CD5) and B-2 (high expression of CD1d, like the marginal zone B cells) cells has recently been reported to play a critical role in the regulation of murine EAE.82  However, the B-cell subsets involved in suppression of other experimental immune diseases do not have exactly the same phenotype.81,91-94 

Ambiguity therefore remains regarding the B-cell subpopulation(s) involved in the regulation of immune responses. An alternative hypothesis is that the immune-suppressive activity is not a unique property of a single B-cell subset but is perhaps exerted by different B-cell subsets, depending on the integration of available signals in the microenvironment.

Collectively, these studies demonstrate that Janus-faced B cells play both pathogenic and regulatory activities in immunopathogenesis (Figure 1) and that these opposing contributions may overlap during the course of the disease. Consequently, the therapeutic effect of B-cell depletion depends on the relative contributions of the opposing B-cell activities at the time of drug administration (Figure 3). It would be of utmost importance to define simple criteria or reliable markers that would help to predict whether administration of rituximab would be beneficial for the patient. Although there are clues about directions that should be taken in the future, there is no easy answer to this question yet. The recent identification of a human B-cell subset with regulatory properties in the peripheral blood of lupus patients86  represents an important step but will likely be insufficient because this subset appears to be qualitatively rather than quantitatively deficient in these patients.86  A functional assay, which would measure the response of T cells to a normalized stimulation in presence or absence of the B cells, could be theoretically useful but its development would require an important amount of work, the limitations of functional assay for routine immunomonitoring being well known (minor changes of test conditions potentially having a major impact on the test results, necessity to work with freshly isolated cells, etc).

Of note, immune stimulation resulting from the removal of B-cell regulation could also be used in a therapeutic perspective. This strategy could be particularly useful in cancer, where B cells have been shown to inhibit the induction of T cell–dependent protective antitumor immunity.95  Accordingly, the administration of anti-CD20 antibodies slowed the growth of nonhematopoietic solid tumors (not expressing CD20) and enhanced the efficiency of a tumor vaccine in a murine model.96  It is thus conceivable that the mode of action of rituximab in hematologic malignancies also relies in part on such a mechanism (Figure 2 bottom panel).

The optimization of B-cell depletion strategies for the treatment of immune diseases therefore not only should focus on the identification of new targets and the development of more depleting drugs, but also requires a better understanding of the complex temporal interplay between pathogenic and regulatory B cells. Instead of a mere depletion of the B-cell compartment, future therapy should instead attempt to reset the regulatory balance—to which B cells can clearly contribute.

This work was supported by grants from the CENTAURE Transplantation Research Network and the Hospices Civils de Lyon.

Contribution: O.T., E.M., and T.D. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Olivier Thaunat, Service de Transplantation Rénale et d'Immunologie Clinique, Hôpital Edouard Herriot, 5 place d'Arsonval, 69437 Lyon Cedex 03, France; e-mail: olivier.thaunatpastu@free.fr.

1
Martin
 
F
Chan
 
AC
B cell immunobiology in disease: evolving concepts from the clinic.
Annu Rev Immunol
2006
, vol. 
24
 (pg. 
467
-
496
)
2
Green
 
EA
Flavell
 
RA
Tumor necrosis factor-alpha and the progression of diabetes in non-obese diabetic mice.
Immunol Rev
1999
, vol. 
169
 (pg. 
11
-
22
)
3
Brodie
 
GM
Wallberg
 
M
Santamaria
 
P
Wong
 
FS
Green
 
EA
B-cells promote intra-islet CD8+ cytotoxic T-cell survival to enhance type 1 diabetes.
Diabetes
2008
, vol. 
57
 
4
(pg. 
909
-
917
)
4
Serreze
 
DV
Chapman
 
HD
Varnum
 
DS
, et al. 
B lymphocytes are essential for the initiation of T cell-mediated autoimmune diabetes: analysis of a new “speed congenic” stock of NOD. Ig mu null mice.
J Exp Med
1996
, vol. 
184
 
5
(pg. 
2049
-
2053
)
5
Chan
 
OT
Hannum
 
LG
Haberman
 
AM
Madaio
 
MP
Shlomchik
 
MJ
A novel mouse with B cells but lacking serum antibody reveals an antibody-independent role for B cells in murine lupus.
J Exp Med
1999
, vol. 
189
 
10
(pg. 
1639
-
1648
)
6
Crawford
 
A
Macleod
 
M
Schumacher
 
T
Corlett
 
L
Gray
 
D
Primary T cell expansion and differentiation in vivo requires antigen presentation by B cells.
J Immunol
2006
, vol. 
176
 
6
(pg. 
3498
-
3506
)
7
Batista
 
FD
Iber
 
D
Neuberger
 
MS
B cells acquire antigen from target cells after synapse formation.
Nature
2001
, vol. 
411
 
6836
(pg. 
489
-
494
)
8
Linton
 
PJ
Harbertson
 
J
Bradley
 
LM
A critical role for B cells in the development of memory CD4 cells.
J Immunol
2000
, vol. 
165
 
10
(pg. 
5558
-
5565
)
9
Batista
 
FD
Harwood
 
NE
The who, how and where of antigen presentation to B cells.
Nat Rev Immunol
2009
, vol. 
9
 
1
(pg. 
15
-
27
)
10
Kakiuchi
 
T
Chesnut
 
RW
Grey
 
HM
B cells as antigen-presenting cells: the requirement for B cell activation.
J Immunol
1983
, vol. 
131
 
1
(pg. 
109
-
114
)
11
Lund
 
FE
Cytokine-producing B lymphocytes-key regulators of immunity.
Curr Opin Immunol
2008
, vol. 
20
 
3
(pg. 
332
-
338
)
12
Moulin
 
V
Andris
 
F
Thielemans
 
K
Maliszewski
 
C
Urbain
 
J
Moser
 
M
B lymphocytes regulate dendritic cell (DC) function in vivo: increased interleukin 12 production by DCs from B cell-deficient mice results in T helper cell type 1 deviation.
J Exp Med
2000
, vol. 
192
 
4
(pg. 
475
-
482
)
13
Takemura
 
S
Klimiuk
 
PA
Braun
 
A
Goronzy
 
JJ
Weyand
 
CM
T cell activation in rheumatoid synovium is B cell dependent.
J Immunol
2001
, vol. 
167
 
8
(pg. 
4710
-
4718
)
14
Drayton
 
DL
Liao
 
S
Mounzer
 
RH
Ruddle
 
NH
Lymphoid organ development: from ontogeny to neogenesis.
Nat Immunol
2006
, vol. 
7
 
4
(pg. 
344
-
353
)
15
Kratz
 
A
Campos-Neto
 
A
Hanson
 
MS
Ruddle
 
NH
Chronic inflammation caused by lymphotoxin is lymphoid neogenesis.
J Exp Med
1996
, vol. 
183
 
4
(pg. 
1461
-
1472
)
16
Hjelmström
 
P
Lymphoid neogenesis: de novo formation of lymphoid tissue in chronic inflammation through expression of homing chemokines.
J Leukoc Biol
2001
, vol. 
69
 
3
(pg. 
331
-
339
)
17
Schröder
 
AE
Greiner
 
A
Seyfert
 
C
Berek
 
C
Differentiation of B cells in the nonlymphoid tissue of the synovial membrane of patients with rheumatoid arthritis.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 
1
(pg. 
221
-
225
)
18
Thaunat
 
O
Field
 
AC
Dai
 
J
, et al. 
Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
41
(pg. 
14723
-
14728
)
19
Aloisi
 
F
Pujol-Borrell
 
R
Lymphoid neogenesis in chronic inflammatory diseases.
Nat Rev Immunol
2006
, vol. 
6
 
3
(pg. 
205
-
217
)
20
Cooper
 
MD
Kearney
 
JF
Gathings
 
WE
Lawton
 
AR
Effects of anti-Ig antibodies on the development and differentiation of B cells.
Immunol Rev
1980
, vol. 
52
 (pg. 
29
-
53
)
21
Riley
 
JK
Sliwkowski
 
MX
CD20: a gene in search of a function.
Semin Oncol
2000
, vol. 
27
 
6 suppl 12
(pg. 
17
-
24
)
22
Grillo-Lopez
 
AJ
Rituximab: an insider's historical perspective.
Semin Oncol
2000
, vol. 
27
 
6 suppl 12
(pg. 
9
-
16
)
23
Maloney
 
DG
Liles
 
TM
Czerwinski
 
DK
, et al. 
Phase I clinical trial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC-C2B8) in patients with recurrent B-cell lymphoma.
Blood
1994
, vol. 
84
 
8
(pg. 
2457
-
2466
)
24
Maloney
 
DG
Mechanism of action of rituximab.
Anticancer Drugs
2001
, vol. 
12
 
suppl 2
(pg. 
S1
-
S4
)
25
Gong
 
Q
Ou
 
Q
Ye
 
S
, et al. 
Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy.
J Immunol
2005
, vol. 
174
 
2
(pg. 
817
-
826
)
26
Thaunat
 
O
Patey
 
N
Gautreau
 
C
, et al. 
B cell survival in intragraft tertiary lymphoid organs after rituximab therapy.
Transplantation
2008
, vol. 
85
 
11
(pg. 
1648
-
1653
)
27
Hagemeister
 
F
Rituximab for the treatment of non-Hodgkin's lymphoma and chronic lymphocytic leukaemia.
Drugs
2010
, vol. 
70
 
3
(pg. 
261
-
272
)
28
Hamblin
 
TJ
Autoimmune complications of chronic lymphocytic leukemia.
Semin Oncol
2006
, vol. 
33
 
2
(pg. 
230
-
239
)
29
Braess
 
J
Reich
 
K
Willert
 
S
, et al. 
Mucocutaneous autoimmune syndrome following fludarabine therapy for low-grade non-Hodgkin's lymphoma of B-cell type (B-NHL).
Ann Hematol
1997
, vol. 
75
 
5-6
(pg. 
227
-
230
)
30
Dearden
 
C
Wade
 
R
Else
 
M
, et al. 
The prognostic significance of a positive direct antiglobulin test in chronic lymphocytic leukemia: a beneficial effect of the combination of fludarabine and cyclophosphamide on the incidence of hemolytic anemia.
Blood
2008
, vol. 
111
 
4
(pg. 
1820
-
1826
)
31
D'Arena
 
G
Laurenti
 
L
Capalbo
 
S
, et al. 
Rituximab therapy for chronic lymphocytic leukemia-associated autoimmune hemolytic anemia.
Am J Hematol
2006
, vol. 
81
 
8
(pg. 
598
-
602
)
32
Bauduer
 
F
Rituximab: a very efficient therapy in cold agglutinins and refractory autoimmune haemolytic anaemia associated with CD20-positive, low-grade non-Hodgkin's lymphoma.
Br J Haematol
2001
, vol. 
112
 
4
(pg. 
1085
-
1086
)
33
Davis
 
TA
Grillo-Lopez
 
AJ
White
 
CA
, et al. 
Rituximab anti-CD20 monoclonal antibody therapy in non-Hodgkin's lymphoma: safety and efficacy of re-treatment.
J Clin Oncol
2000
, vol. 
18
 
17
(pg. 
3135
-
3143
)
34
Edwards
 
JC
Cambridge
 
G
B-cell targeting in rheumatoid arthritis and other autoimmune diseases.
Nat Rev Immunol
2006
, vol. 
6
 
5
(pg. 
394
-
403
)
35
Edwards
 
JC
Szczepanski
 
L
Szechinski
 
J
, et al. 
Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis.
N Engl J Med
2004
, vol. 
350
 
25
(pg. 
2572
-
2581
)
36
Cohen
 
SB
Emery
 
P
Greenwald
 
MW
, et al. 
Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks.
Arthritis Rheum
2006
, vol. 
54
 
9
(pg. 
2793
-
2806
)
37
Godeau
 
B
Porcher
 
R
Fain
 
O
, et al. 
Rituximab efficacy and safety in adult splenectomy candidates with chronic immune thrombocytopenic purpura: results of a prospective multicenter phase 2 study.
Blood
2008
, vol. 
112
 
4
(pg. 
999
-
1004
)
38
Hauser
 
SL
Waubant
 
E
Arnold
 
DL
, et al. 
B-cell depletion with rituximab in relapsing-remitting multiple sclerosis.
N Engl J Med
2008
, vol. 
358
 
7
(pg. 
676
-
688
)
39
Specks
 
U
Fervenza
 
FC
McDonald
 
TJ
Hogan
 
MC
Response of Wegener's granulomatosis to anti-CD20 chimeric monoclonal antibody therapy.
Arthritis Rheum
2001
, vol. 
44
 
12
(pg. 
2836
-
2840
)
40
Levine
 
TD
Pestronk
 
A
IgM antibody-related polyneuropathies: B-cell depletion chemotherapy using Rituximab.
Neurology
1999
, vol. 
52
 
8
(pg. 
1701
-
1704
)
41
Heemstra
 
KA
Toes
 
RE
Sepers
 
J
, et al. 
Rituximab in relapsing Graves' disease, a phase II study.
Eur J Endocrinol
2008
, vol. 
159
 
5
(pg. 
609
-
615
)
42
Zaja
 
F
Russo
 
D
Fuga
 
G
Perella
 
G
Baccarani
 
M
Rituximab for myasthenia gravis developing after bone marrow transplant.
Neurology
2000
, vol. 
55
 
7
(pg. 
1062
-
1063
)
43
Lambotte
 
O
Kotb
 
R
Maigne
 
G
Blanc
 
FX
Goujard
 
C
Delfraissy
 
JF
Efficacy of rituximab in refractory polymyositis.
J Rheumatol
2005
, vol. 
32
 
7
(pg. 
1369
-
1370
)
44
Ahmed
 
AR
Spigelman
 
Z
Cavacini
 
LA
Posner
 
MR
Treatment of pemphigus vulgaris with rituximab and intravenous immune globulin.
N Engl J Med
2006
, vol. 
355
 
17
(pg. 
1772
-
1779
)
45
Joly
 
P
Mouquet
 
H
Roujeau
 
JC
, et al. 
A single cycle of rituximab for the treatment of severe pemphigus.
N Engl J Med
2007
, vol. 
357
 
6
(pg. 
545
-
552
)
46
Zaja
 
F
De Vita
 
S
Mazzaro
 
C
, et al. 
Efficacy and safety of rituximab in type II mixed cryoglobulinemia.
Blood
2003
, vol. 
101
 
10
(pg. 
3827
-
3834
)
47
Fakhouri
 
F
Vernant
 
JP
Veyradier
 
A
, et al. 
Efficiency of curative and prophylactic treatment with rituximab in ADAMTS13-deficient thrombotic thrombocytopenic purpura: a study of 11 cases.
Blood
2005
, vol. 
106
 
6
(pg. 
1932
-
1937
)
48
Pijpe
 
J
van Imhoff
 
GW
Spijkervet
 
FK
, et al. 
Rituximab treatment in patients with primary Sjogren's syndrome: an open-label phase II study.
Arthritis Rheum
2005
, vol. 
52
 
9
(pg. 
2740
-
2750
)
49
Wiestner
 
A
Cho
 
HJ
Asch
 
AS
, et al. 
Rituximab in the treatment of acquired factor VIII inhibitors.
Blood
2002
, vol. 
100
 
9
(pg. 
3426
-
3428
)
50
Levesque
 
MC
Translational mini-review series on B cell-directed therapies: recent advances in B cell-directed biological therapies for autoimmune disorders.
Clin Exp Immunol
2009
, vol. 
157
 
2
(pg. 
198
-
208
)
51
Thaunat
 
O
Patey
 
N
Morelon
 
E
Michel
 
JB
Nicoletti
 
A
Lymphoid neogenesis in chronic rejection: the murderer is in the house.
Curr Opin Immunol
2006
, vol. 
18
 
5
(pg. 
576
-
579
)
52
Colvin
 
RB
Smith
 
RN
Antibody-mediated organ-allograft rejection.
Nat Rev Immunol
2005
, vol. 
5
 
10
(pg. 
807
-
817
)
53
Becker
 
YT
Becker
 
BN
Pirsch
 
JD
Sollinger
 
HW
Rituximab as treatment for refractory kidney transplant rejection.
Am J Transplant
2004
, vol. 
4
 
6
(pg. 
996
-
1001
)
54
Becker
 
YT
Samaniego-Picota
 
M
Sollinger
 
HW
The emerging role of rituximab in organ transplantation.
Transpl Int
2006
, vol. 
19
 
8
(pg. 
621
-
628
)
55
Leandro
 
MJ
Cambridge
 
G
Edwards
 
JC
Ehrenstein
 
MR
Isenberg
 
DA
B-cell depletion in the treatment of patients with systemic lupus erythematosus: a longitudinal analysis of 24 patients.
Rheumatology (Oxford)
2005
, vol. 
44
 
12
(pg. 
1542
-
1545
)
56
Looney
 
RJ
Anolik
 
JH
Campbell
 
D
, et al. 
B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose-escalation trial of rituximab.
Arthritis Rheum
2004
, vol. 
50
 
8
(pg. 
2580
-
2589
)
57
Merrill
 
JT
Neuwelt
 
CM
Wallace
 
DJ
, et al. 
Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial.
Arthritis Rheum
2010
, vol. 
62
 
1
(pg. 
222
-
233
)
58
Favas
 
C
Isenberg
 
DA
B-cell-depletion therapy in SLE: what are the current prospects for its acceptance?
Nat Rev Rheumatol
2009
, vol. 
5
 
12
(pg. 
711
-
716
)
59
Goetz
 
M
Atreya
 
R
Ghalibafian
 
M
Galle
 
PR
Neurath
 
MF
Exacerbation of ulcerative colitis after rituximab salvage therapy.
Inflamm Bowel Dis
2007
, vol. 
13
 
11
(pg. 
1365
-
1368
)
60
Shaikh
 
A
Habermann
 
TM
Fidler
 
ME
Kumar
 
S
Leung
 
N
Acute renal failure secondary to severe type I cryoglobulinemia following rituximab therapy for Waldenstrom's macroglobulinemia.
Clin Exp Nephrol
2008
, vol. 
12
 
4
(pg. 
292
-
295
)
61
Suzuki
 
K
Nagasawa
 
H
Kameda
 
H
, et al. 
Severe acute thrombotic exacerbation in two cases with anti-phospholipid syndrome after retreatment with rituximab in phase I/II clinical trial for refractory systemic lupus erythematosus.
Rheumatology (Oxford)
2009
, vol. 
48
 
2
(pg. 
198
-
199
)
62
Dass
 
S
Vital
 
EM
Emery
 
P
Development of psoriasis after B cell depletion with rituximab.
Arthritis Rheum
2007
, vol. 
56
 
8
(pg. 
2715
-
2718
)
63
Mielke
 
F
Schneider-Obermeyer
 
J
Dorner
 
T
Onset of psoriasis with psoriatic arthropathy during rituximab treatment of non-Hodgkin lymphoma.
Ann Rheum Dis
2008
, vol. 
67
 
7
(pg. 
1056
-
1057
)
64
Dereure
 
O
Navarro
 
R
Rossi
 
JF
Guilhou
 
JJ
Rituximab-induced vasculitis.
Dermatology
2001
, vol. 
203
 
1
(pg. 
83
-
84
)
65
Kandula
 
P
Kouides
 
PA
Rituximab-induced leukocytoclastic vasculitis: a case report.
Arch Dermatol
2006
, vol. 
142
 
2
(pg. 
246
-
247
)
66
Kim
 
MJ
Kim
 
HO
Kim
 
HY
Park
 
YM
Rituximab-induced vasculitis: a case report and review of the medical published work.
J Dermatol
2009
, vol. 
36
 
5
(pg. 
284
-
287
)
67
Liu
 
X
Hong
 
XN
Gu
 
YJ
Wang
 
BY
Luo
 
ZG
Cao
 
J
Interstitial pneumonitis during rituximab-containing chemotherapy for non-Hodgkin lymphoma.
Leuk Lymphoma
2008
, vol. 
49
 
9
(pg. 
1778
-
1783
)
68
Tonelli
 
AR
Lottenberg
 
R
Allan
 
RW
Sriram
 
PS
Rituximab-induced hypersensitivity pneumonitis.
Respiration
2009
, vol. 
78
 
2
(pg. 
225
-
229
)
69
Wagner
 
SA
Mehta
 
AC
Laber
 
DA
Rituximab-induced interstitial lung disease.
Am J Hematol
2007
, vol. 
82
 
10
(pg. 
916
-
919
)
70
Jourdan
 
E
Topart
 
D
Richard
 
B
Jourdan
 
J
Sotto
 
A
Severe autoimmune hemolytic anemia following rituximab therapy in a patient with a lymphoproliferative disorder.
Leuk Lymphoma
2003
, vol. 
44
 
5
(pg. 
889
-
890
)
71
Voog
 
E
Morschhauser
 
F
Solal-Celigny
 
P
Neutropenia in patients treated with rituximab.
N Engl J Med
2003
, vol. 
348
 
26
(pg. 
2691
-
2694
discussion 2691–2694
72
Ioannidis
 
JP
Lau
 
J
Completeness of safety reporting in randomized trials: an evaluation of 7 medical areas.
JAMA
2001
, vol. 
285
 
4
(pg. 
437
-
443
)
73
Fridkis-Hareli
 
M
Immunogenetic mechanisms for the coexistence of organ-specific and systemic autoimmune diseases.
J Autoimmune Dis
2008
, vol. 
5
 pg. 
1
  
74
Clatworthy
 
MR
Watson
 
CJ
Plotnek
 
G
, et al. 
B-cell-depleting induction therapy and acute cellular rejection.
N Engl J Med
2009
, vol. 
360
 
25
(pg. 
2683
-
2685
)
75
Matsushita
 
T
Yanaba
 
K
Bouaziz
 
JD
Fujimoto
 
M
Tedder
 
TF
Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression.
J Clin Invest
2008
, vol. 
118
 
10
(pg. 
3420
-
3430
)
76
Shimamura
 
T
Hashimoto
 
K
Sasaki
 
S
Feedback suppression of the immune response in vivo: I, immune B cells induce antigen-specific suppressor T cells.
Cell Immunol
1982
, vol. 
68
 
1
(pg. 
104
-
113
)
77
Wolf
 
SD
Dittel
 
BN
Hardardottir
 
F
Janeway
 
CA
Experimental autoimmune encephalomyelitis induction in genetically B cell-deficient mice.
J Exp Med
1996
, vol. 
184
 
6
(pg. 
2271
-
2278
)
78
Mauri
 
C
Gray
 
D
Mushtaq
 
N
Londei
 
M
Prevention of arthritis by interleukin 10-producing B cells.
J Exp Med
2003
, vol. 
197
 
4
(pg. 
489
-
501
)
79
Mizoguchi
 
A
Mizoguchi
 
E
Smith
 
RN
Preffer
 
FI
Bhan
 
AK
Suppressive role of B cells in chronic colitis of T cell receptor alpha mutant mice.
J Exp Med
1997
, vol. 
186
 
10
(pg. 
1749
-
1756
)
80
Fillatreau
 
S
Sweenie
 
CH
McGeachy
 
MJ
Gray
 
D
Anderton
 
SM
B cells regulate autoimmunity by provision of IL-10.
Nat Immunol
2002
, vol. 
3
 
10
(pg. 
944
-
950
)
81
Mizoguchi
 
A
Mizoguchi
 
E
Takedatsu
 
H
Blumberg
 
RS
Bhan
 
AK
Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation.
Immunity
2002
, vol. 
16
 
2
(pg. 
219
-
230
)
82
Yanaba
 
K
Bouaziz
 
JD
Haas
 
KM
Poe
 
JC
Fujimoto
 
M
Tedder
 
TF
A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses.
Immunity
2008
, vol. 
28
 
5
(pg. 
639
-
650
)
83
Sun
 
JB
Flach
 
CF
Czerkinsky
 
C
Holmgren
 
J
B lymphocytes promote expansion of regulatory T cells in oral tolerance: powerful induction by antigen coupled to cholera toxin B subunit.
J Immunol
2008
, vol. 
181
 
12
(pg. 
8278
-
8287
)
84
Fillatreau
 
S
Gray
 
D
Anderton
 
SM
Not always the bad guys: B cells as regulators of autoimmune pathology.
Nat Rev Immunol
2008
, vol. 
8
 
5
(pg. 
391
-
397
)
85
Duddy
 
M
Niino
 
M
Adatia
 
F
, et al. 
Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis.
J Immunol
2007
, vol. 
178
 
10
(pg. 
6092
-
6099
)
86
Blair
 
PA
Norena
 
LY
Flores-Borja
 
F
, et al. 
CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients.
Immunity
2010
, vol. 
32
 
1
(pg. 
129
-
140
)
87
Lampropoulou
 
V
Hoehlig
 
K
Roch
 
T
, et al. 
TLR-activated B cells suppress T cell-mediated autoimmunity.
J Immunol
2008
, vol. 
180
 
7
(pg. 
4763
-
4773
)
88
Carroll
 
MC
Prodeus
 
AP
Linkages of innate and adaptive immunity.
Curr Opin Immunol
1998
, vol. 
10
 
1
(pg. 
36
-
40
)
89
Silverman
 
GJ
Srikrishnan
 
R
Germar
 
K
, et al. 
Genetic imprinting of autoantibody repertoires in systemic lupus erythematosus patients.
Clin Exp Immunol
2008
, vol. 
153
 
1
(pg. 
102
-
116
)
90
Wei
 
B
Velazquez
 
P
Turovskaya
 
O
, et al. 
Mesenteric B cells centrally inhibit CD4+ T cell colitis through interaction with regulatory T cell subsets.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
6
(pg. 
2010
-
2015
)
91
Blair
 
PA
Chavez-Rueda
 
KA
Evans
 
JG
, et al. 
Selective targeting of B cells with agonistic anti-CD40 is an efficacious strategy for the generation of induced regulatory T2-like B cells and for the suppression of lupus in MRL/lpr mice.
J Immunol
2009
, vol. 
182
 
6
(pg. 
3492
-
3502
)
92
Evans
 
JG
Chavez-Rueda
 
KA
Eddaoudi
 
A
, et al. 
Novel suppressive function of transitional 2 B cells in experimental arthritis.
J Immunol
2007
, vol. 
178
 
12
(pg. 
7868
-
7878
)
93
Gray
 
M
Miles
 
K
Salter
 
D
Gray
 
D
Savill
 
J
Apoptotic cells protect mice from autoimmune inflammation by the induction of regulatory B cells.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
35
(pg. 
14080
-
14085
)
94
Lenert
 
P
Brummel
 
R
Field
 
EH
Ashman
 
RF
TLR-9 activation of marginal zone B cells in lupus mice regulates immunity through increased IL-10 production.
J Clin Immunol
2005
, vol. 
25
 
1
(pg. 
29
-
40
)
95
Qin
 
Z
Richter
 
G
Schuler
 
T
Ibe
 
S
Cao
 
X
Blankenstein
 
T
B cells inhibit induction of T cell-dependent tumor immunity.
Nat Med
1998
, vol. 
4
 
5
(pg. 
627
-
630
)
96
Kim
 
S
Fridlender
 
ZG
Dunn
 
R
, et al. 
B-cell depletion using an anti-CD20 antibody augments antitumor immune responses and immunotherapy in nonhematopoetic murine tumor models.
J Immunother
2008
, vol. 
31
 
5
(pg. 
446
-
457
)
Sign in via your Institution