Next Article in Journal
Novel Diels–Alder Type Adducts from Morus alba Root Bark Targeting Human Monoamine Oxidase and Dopaminergic Receptors for the Management of Neurodegenerative Diseases
Previous Article in Journal
Full-Length Transcriptome Sequencing and Different Chemotype Expression Profile Analysis of Genes Related to Monoterpenoid Biosynthesis in Cinnamomum porrectum
Previous Article in Special Issue
Multiple Functions of B Cells in the Pathogenesis of Systemic Lupus Erythematosus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

B Cell Abnormalities in Systemic Lupus Erythematosus and Lupus Nephritis—Role in Pathogenesis and Effect of Immunosuppressive Treatments

Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(24), 6231; https://doi.org/10.3390/ijms20246231
Submission received: 28 October 2019 / Revised: 2 December 2019 / Accepted: 3 December 2019 / Published: 10 December 2019

Abstract

:
Abnormalities in B cells play pivotal roles in the pathogenesis of systemic lupus erythematosus (SLE) and lupus nephritis (LN). Breach in central and peripheral tolerance mechanisms generates autoreactive B cells which contribute to the pathogenesis of SLE and LN. Dysregulation of B cell transcription factors, cytokines and B cell–T cell interaction can result in aberrant B cell maturation and autoantibody production. These immunological abnormalities also lead to perturbations in circulating and infiltrating B cells in SLE and LN patients. Conventional and novel immunosuppressive medications confer differential effects on B cells which have important clinical implications. While cyclophosphamide and mycophenolate mofetil (MMF) showed comparable clinical efficacy in active LN, MMF induction was associated with earlier reduction in circulating plasmablasts and plasma cells. Accumulating evidence suggests that MMF maintenance is associated with lower risk of disease relapse than azathioprine, which may be explained by its more potent and selective suppression of B cell proliferation. Novel therapeutic approaches targeting the B cell repertoire include B cell depletion with monoclonal antibodies binding to cell surface markers, inhibition of B cell cytokines, and modulation of costimulatory signals in B cell–T cell interaction. These biologics, despite showing improvements in serological parameters and proteinuria, did not achieve primary endpoints when used as add-on therapy to standard treatments in active LN patients. Other emerging treatments such as calcineurin inhibitors, mammalian target of rapamycin inhibitors and proteasome inhibitors also show distinct inhibitory effects on the B cell repertoire. Advancement in the knowledge on B cell biology has fueled the development of new therapeutic strategies in SLE and LN. Modification in background treatments, study endpoints and selective recruitment of subjects showing aberrant B cells or its signaling pathways when designing future clinical trials may better elucidate the roles of these novel therapies for SLE and LN patients.

1. Introduction

Systemic lupus erythematosus (SLE) is an autoimmune disease with abnormal interplay between innate and adaptive immunity, breach of immune tolerance, production of autoantibodies, and immunological insult to multiple organ systems. Kidney involvement is common among patients with SLE, and the presence of lupus nephritis (LN) significantly increased the risk of renal failure and patient mortality [1,2]. The management of LN is challenging because of substantial patient variability in disease course and response to treatment [3,4]. Such patient heterogeneity may be related to the complexity of LN pathogenesis. Genetic predispositions, abnormalities in lymphocytes, aberrant complement activation, autoantibody production, and perturbed cytokine milieu all contribute to the pathogenesis of SLE and LN [5,6,7]. In this context, abnormalities in B cells is a key player in SLE and LN pathogenesis as autoantibodies are important for diagnosis and the changes in autoantibodies level may also show correlations with clinical disease activity [5,6,7,8,9,10]. B cells also show important immunological functions pertinent to SLE and LN pathogenesis such as presentation of autoantigens and secretion of proinflammatory cytokines. Therefore, the study of B lymphocytes can potentially unravel important pathogenic mechanisms of SLE/LN and thus help develop more specific therapies to improve treatment efficacy and tolerability. The following discussion will highlight the B cell abnormalities which are relevant to SLE and LN pathogenesis and also the effect of immunosuppressive medications on the B cell repertoire.

2. Normal Development and Homeostasis of B Lymphocytes

The development of B cells begins in the bone marrow. Hematopoietic stem cells commit to the lymphoid lineage and become common lymphoid precursors (CLP) [11]. CLP differentiate into pre-B cells and pro-B cells, which will then undergo immunoglobulin heavy and light chain gene rearrangement to form B cell receptors (BCR) and surface Immunoglobulin M (IgM) to become immature B cells. These stages of B cell development within the bone marrow are antigen-independent. Immature B cells exit the bone marrow to complete further maturation and differentiation in peripheral secondary lymphoid organs such as the spleen or lymph nodes, whereby B cells can be activated by either T cell-dependent or -independent pathways [12]. Within the secondary lymphoid organs, immature B cells migrate towards the lymphoid follicles. The majority of naïve B cells move into the germinal center (follicular B cells) while a small number of naïve B cells remain in the marginal zone. Follicular B cells, after cognate interaction with T helper cells, will undergo somatic hypermutation (SHM) and class switch recombination (CSR) to enhance the affinity of immunoglobulins [11,12]. Follicular B cells can further differentiate into memory B cells when the antigenic stimulation is relatively weak but into long-lived plasma cells when there are strong antigenic stimuli. Memory B cells usually remain in a dormant state, but can rapidly differentiate into antibody-producing effector cells when re-challenged with previously encountered antigens. Long-lived plasma cells mostly migrate back to the bone marrow and are responsible for antibody production, although a small proportion will remain in peripheral tissues [13]. In contrast, naïve B cells which remain in the marginal zone will become short-lived plasma cells.
The homeostasis of B cells is tightly orchestrated by various transcription factors and cytokines., The early B cell factor (EBF) promotes CLP to commit to the B cell lineage and initiates immunoglobulin heavy chain rearrangement [14]. Paired box protein -5 (PAX5) is a master regulator in B cell development at different stages, and regulates gene transcription by recruiting chromatin-remodeling, histone-modifying, and basal transcription factor complexes to its target genes [12]. PAX5 shows dual effects on early B cell development, which include the suppression of B-lineage-inappropriate genes and activation B cell-specific genes [15]. PAX5 is also involved in V(D)J recombination, B cell signaling, as well as adhesion and migration of B lymphocytes [15]. BACH2 is another signature B cell transcription factor with instrumental roles in B lymphocyte development starting early from the CLP stage. broad complex-tramtrack-bric a brac and Cap’n’collar homology 1 (BACH2), with broad complex-tramtrack-bric a brac and Cap’n’collar homology 2 (BACH1) as an auxiliary, suppress ‘myeloid genes’ in pre-pro-B cells by binding to their regulatory regions to promote development of B lymphocytes [16]. BACH2 expression is also critical in determining the fate of B cells during germinal center reaction and can interact with BCL-6 and repress lymphocyte-induced maturation protein-1 (BLIMP-1) transcription to prevent premature plasma cell differentiation [17]. Murine splenic B cells in the absence of BACH2 show increased propensity to differentiate into plasma cells, and such differentiation is regulated via both BLIMP-1-dependent and -independent pathways [18]. On the other hand, B lymphocytes which show increased BACH2 and decreased BLIMP-1 in the germinal center will preferentially differentiate into memory B cells [14].
The survival, maturation and differentiation of B lymphocytes are also affected by various B cell-related cytokines such as B-cell activating factor (BAFF), interleukin-6 (IL-6) and interleukin (IL-21) [19]. Both BAFF (also known as B lymphocytes stimulator, BLys) and a proliferation-inducing ligand (APRIL) belong to the TNF ligand superfamily and are actively secreted by dendritic cells, macrophages, and neutrophils. BAFF and APRIL are key survival factors for B lymphocytes and have potent stimulatory effects on B cell proliferation and antibody production [20]. BAFF can bind to three types of receptors: BAFF receptor, transmembrane activator, and calcium modulator and cyclophilin ligand interactor (TACI) and B-cell maturation antigen (BCMA), while APRIL can only bind to TACI and BCMA [19,21,22]. Most B cell subsets express all three types of receptors but plasma cells only show BCMA on their cell surface [19,21]. These variations in receptor expression may account for the differential response of B cell subsets to biologics targeting B cell survival factors. IL-6 is a proinflammatory cytokine produced by monocytes, fibroblasts, and endothelial cells, and to a lesser extent B and T cells and resident renal cells [19]. IL-6 promotes the maturation of naïve B cells into memory or plasma cells, differentiation of follicular T helper cells (TFH), formation of germinal center, and production of antibodies (including autoantibodies) [19,23]. IL-21 is a cytokine produced by activated T helper cells (especially Th2, Th17, and TFH cells), and exerts strong driving effects on plasma cell proliferation and differentiation as well as immunoglobulin production [19]. In this context, IL-21 works synergistically with CD40L to induce differentiation of naïve and memory B cells into antibody-producing plasma cells [24]. The role of these B cell-related cytokines in SLE and LN pathogenesis and the implications on treatment will be further discussed in the following sections.

3. Abnormalities in B Cell Tolerance and Regulation—Role in SLE and LN Pathogenesis

The development and survival of B cells which react against self-antigens are prevented by both central and peripheral tolerance mechanisms [14]. Failure of these tolerance mechanisms will result in generation of autoreactive B cells which contribute to the pathogenesis of SLE and LN. Abnormality in BCR signaling is one important mechanism for the breakdown of central tolerance. Deficiency of surrogate light chains (SLC) was associated with development of autoreactive B cells, possibly as a result of the selection advantage of BCR expressing self-recognizing IgH chains [14]. Impaired BCR signaling in B cells will also lead to failure to trigger apoptosis of early B cells which recognize self-antigens [14,25].
Defects in peripheral B cell tolerance are also pertinent to B cell autoimmunity. SLE patients show aberrant and increased expression of recombination-activating genes (RAG) in peripheral B cells, which can lead to mutation of BCR and thus development of autoreactive B lymphocytes [26]. Aberrant T cell-B cell interaction (e.g., shorted interaction time in the germinal center) results in inadequate anergic signals to autoreactive B cells and hence enhancing their survival [27,28]. Increased BCR-mediated signaling can also lower the activation threshold of peripheral B lymphocytes and promote cellular phenotypes characteristic of SLE [29]. Lupus B cells also showed augmented SHM and CSR [30,31,32,33,34], which contribute to increased pathogenicity of plasma cells. Other immunological anomalies relevant to B cell autoreactivity in SLE include increased plasma cell differentiation and survival, upregulated Toll-like receptor (TLR) signaling and increased expression of key B cell cytokines such as BAFF, IL-6, and IL-21 [13,35,36,37,38,39,40]. Indeed, transgenic mice overexpressing BAFF exhibited increased number of plasma cells in secondary lymphoid tissues, presence of autoantibodies, and also increased circulating immune complexes and immunoglobulin deposition in the kidneys [41]. Raised blood levels of BAFF has been observed in NZB/W F1 and MRL/lpr mice at the onset of disease [22], and treatment with soluble TACI-Ig mitigated the development of proteinuria and improved survival of NZB/W F1 mice [22]. Deletion of TACI receptor in transgenic mice overexpressing BAFF inhibited immune activation, diminished immunoglobulins production and conferred long-term protection from progressive glomerulonephritis for up to 12 months in these mice [42]. Elevated circulating BAFF levels have been observed in patients with SLE, which correlated with anti-dsDNA autoantibody levels and SLEDAI scores [43].
Interleukin-6 (IL-6) is a proinflammatory cytokine and its strong pathogenic significance in SLE and LN has been demonstrated by both animal and human studies. B lymphocytes isolated from SLE patients secrete high amount of IL-6 which can bind to the IL-6 receptor of other B cells to promote their terminal differentiation, and thus forming a positive IL-6 feedback loop [44]. Treatment with polyclonal anti-IL-6 or anti-IL-6 receptor monoclonal antibodies could inhibit IL-6 binding and suppressed total IgG and IgG anti-ssDNA antibody secretion in lupus B cells [44]. In a murine SLE model, B cell-derived IL-6 could induce TFH differentiation and initiate germinal center formation [45]. Treatment of lupus prone NZB/W F1 mice with IL-6 exacerbated glomerulonephritis [46], whilst treatment with anti-IL-6 monoclonal antibodies in NZB/W F1 mice ameliorated kidney manifestations and reduced circulating anti-dsDNA autoantibodies titers [47,48]. Active LN patients showed elevated urinary levels of IL-6 compared with patients in remission [49], and renal biopsies obtained from LN patients also showed increased IL-6 expression in the glomerular and tubular regions [50].
IL-21 is a key driver of plasma cell differentiation and proliferation and thus has important pathogenic relevance in SLE. B lymphocytes isolated from SLE patients, when stimulated with autologous CD3+ T lymphocytes and IL-21, showed prominent increase in IgG production whereas treatment with Fc fusion protein against IL-21 receptor (IL-21R) would inhibit the differentiation of B lymphocytes into plasma cells [51]. BXSB-Yaa lupus-prone mice showed higher circulating IL-21 and its mRNA transcripts compared with wild-type mice [52], and deletion of IL-21R would abrogate characteristic lupus phenotypes such as autoantibodies production and glomerulonephritis in these mice [53]. Treatment of MRL/lpr mice with IL-21R.Fc fusion protein reduced anti-dsDNA autoantibody titers and lymph node enlargement, and also alleviated renal and dermatological lesions [54]. SLE patients showed raised serum IL-21 levels, and population-based case-control association analysis demonstrated that genetic polymorphisms in the IL-21 (rs907715) and IL-21R gene (rs2221903) were associated with escalated risk of SLE in European-American patients [55,56].
Toll-like receptors (TLR) play pivotal roles in B cell activation and also contribute to the pathogenesis of SLE and LN. In this context, TLR-7 and TLR-9 are potent inducers of Type I interferon response and show more pathogenic relevance in SLE and LN [57]. TLR-7 is expressed on different B cell subpopulations and a previous study showed that autophagy in B cells was a trigger for TLR-7-dependent autoantibody production [58,59]. BCR-driven uptake of immune complexes stimulates TLR-7 and -9 in B cells and promotes RNA- and DNA-autoantibodies production [39,60,61,62,63]. TLR-9 signaling in B lymphocytes is also essential for generation of autoantibodies against DNA in mice and enhances the differentiation of autoantibody-producing B cells and plasma cells in human [64,65]. TLR-9 mRNA expression was also increased in PBMCs isolated from SLE patients and correlated with severity of LN and anti-DNA antibody titers [66].

4. Perturbations in Circulating and Infiltrating B Cell Subsets—Role in SLE and LN Pathogenesis

Abnormalities in the tolerance and regulatory mechanisms of the B cell repertoire in SLE and LN can result in perturbations in the B lymphocyte subsets and their immune responsiveness. B cells which secrete autoantibodies against glomerular antigens have been isolated from nephritic MRL/lpr mice, and MRL/lpr and NZB/W F1 mice with established nephritis also exhibited increased intra-renal B cells and plasma cells infiltration [67,68]. In NZB/W F1 mice, intra-renal plasma cells showed comparable number and immunoglobulin secretion as the plasma cells residing in the bone marrow, and the degree of intra-renal plasma cell infiltration also correlated with anti-dsDNA autoantibodies titers [68]. The pathogenic contribution of the B cell repertoire in LN extends beyond autoantibodies production. While glomerulonephritis can still develop in MRL/lpr mice which were genetically manipulated to become incapable of antibody secretion, severe nephritic lesions do not occur in MRL/lpr mice with B-cell deficiency and such observation might be related to impaired development of activated cytotoxic and helper T cells in these mice [69,70]. In SLE patients, there was an increase in the frequency of peripheral plasma cells and memory B cells but diminished number of circulating naïve B cells [71,72]. Such alterations in the B cell subsets profiles have several important implications on disease behavior in SLE and LN patients. Memory B cells have reduced FcγRIIb expression and thus lower reactivation threshold [72,73]. The low proliferation rates in memory B cells also make them less susceptible to conventional immunosuppressive medications which are cell-cycle dependent, and thus become more readily reactivated during disease relapse [71]. Patients with active SLE have elevated number of circulating plasma cells, which shows positive relationship with serum levels of immunoglobulin and anti-dsDNA autoantibodies and disease activity scores [71,74]. Furthermore, the degree of intra-renal plasma cell infiltration also correlates with renal histological activity and chronicity indices in LN patients [68].

5. Effect of Immunosuppressive Treatments on B Cells and Implications on the Choice of Therapies in Lupus nephritis

Treatment for LN is generally governed by histopathological findings and the severity of renal disease. Immunosuppressive treatment is indicated in patients presenting with active severe LN—focal or diffuse proliferative (Class III or IV) LN, or severe membranous (Class V) LN. Clinical management is often divided into the induction and maintenance phases. The induction phase entails the use of high-dose immunosuppressive therapies for approximately four to six months, with the goal to abate active renal inflammation and limit kidney parenchymal damage. The maintenance phase refers to the application of continuous low-dose immunosuppression after induction therapy, to consolidate response and to prevent disease relapse. Given its instrumental roles in SLE and LN pathogenesis, the B cell repertoire is therefore an attractive therapeutic target in SLE and LN (Figure 1) and the effect of immunosuppressive treatments on B cells have important clinical implications (Table 1).

5.1. Conventional Immunosuppressive Treatments for SLE and LN

The current standard-of-care induction treatments for active severe LN are high-dose corticosteroids combined with either cyclophosphamide (CYC) or mycophenolate (MMF), and the maintenance therapies are low-dose corticosteroids coupled with either MMF or azathioprine (AZA) [75,76,77,78]. The current data show that CYC or MMF induction conferred similar short-term efficacy in patients with active proliferative LN [79,80,81,82]. Interestingly, MMF induction was associated with earlier reduction of circulating plasmablasts and plasma cells compared with CYC [83]. Also, CYC induction showed preferential depletion of less mature B cells such as naïve B cells and pre-switched memory B cells compared with MMF [83]. Whether these effects on B cell subsets can account for the better comparative efficacy of MMF in some high-risk ethnic groups (e.g., Afro-Americans) remain speculative. Other investigators have reported that CYC or MMF induction in active SLE patients was associated with different T cell subset profiles but not the B cell subpopulations after 4 weeks of treatment [84]. More importantly, neither CYC nor MMF had significant effects on class-switched memory B cells which are typically resting and non-proliferating during active LN and therefore these memory B cells can be activated to trigger disease relapse in patients who have apparently responded well to induction treatments [83]. Accumulating evidence has demonstrated MMF maintenance is associated with lower risk of disease relapse compared with azathioprine (AZA) maintenance [85,86,87]. Indeed, data from in vitro studies suggested that the AZA dose required for the suppression of humoral response in mice was significantly higher than that required for the inhibition of cellular immunity [88,89]. It has also been demonstrated that SLE patients receiving MMF maintenance showed lower number of circulating plasmablasts but higher number of naïve and transitional B cells compared with patients receiving AZA maintenance [90]. The authors also showed that MMF could profoundly suppress the proliferation of B cells from healthy individuals in vitro, but there was no comparison with AZA, and no information on lupus B cells [90]. Our preliminary results showed decreased miRNA-148a and increased BACH1, BACH2, and PAX5 expression in B cells from LN patients receiving MMF maintenance treatment compared with B cells from AZA-treated patients [91,92]. The B cells from MMF-treated LN patients also showed reduced cell proliferation upon ex vivo stimulation compared with B cells from patients receiving AZA maintenance [92]. These differences in B cell subset profiles, cellular signatures, and proliferative capacity provide a rationale for the relatively lower relapse rate in patients receiving MMF maintenance observed clinically.

5.2. Biologics and Emerging Therapies or SLE and LN

Anti-CD20 monoclonal antibody is an established treatment for B cell malignancy and has also been used in various autoimmune conditions [93,94]. CD20 is a cell surface marker expressed on different B cell subpopulations, and therefore anti-CD20 treatment can lead to profound depletion of B lymphocytes which usually last for at least 6 months [95]. The mechanisms of B cell depletion with anti-CD20 monoclonal antibodies include antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity, and enhanced apoptosis of B cells [96]. Treatment of MRL/lpr mice with anti-CD20 monoclonal antibodies resulted in decline of anti-dsDNA autoantibodies and amelioration of renal lesions [97]. Also, administration of anti-CD20 monoclonal antibodies in NZB/W F1 mice could delay the onset of disease and retard progression of glomerulonephritis [98]. Rituximab is a chimeric monoclonal antibody against CD20, which was initially developed for the treatment of B cell lymphomas [93]. The effect of rituximab treatment on B cells is quite variable between SLE patients [99,100,101]. Previous studies showed that rituximab treatment could lead to profound B cell depletion as early as 2 weeks after administration, and B cells repopulate to approximately 65% of the baseline levels after 12 months [102,103]. Results from the LUNAR trial showed that rituximab as add-on therapy to standard induction treatments did not improve renal outcomes although the rituximab group showed more improvement in the levels of complement and anti-dsDNA and proteinuria [104]. The failure to achieve additional renal benefit may be related to the potent background immunosuppressive treatments, and that anti-CD20 treatment only deplete circulating B lymphocytes but not tissue B cells and plasma cells which do not express CD20. While the clinical response to anti-CD20 treatment could be quite variable between SLE patients, monitoring of B cell biomarkers might help predict treatment response and disease course. Persistent presence of B cells was associated with poor therapeutic response, and early repopulation of memory B cells and plasmablasts was related to early disease relapse [101]. Lower pre-treatment plasmablasts were predictive of complete B cell depletion, and B cell depletion at 6 weeks increased the odds of major clinical response [105]. Other investigators also reported that faster repopulation of B cells was associated with early relapse, and patients who relapsed with high anti-dsDNA levels showed increased percentage of circulating IgD-CD27hi plasmablasts whereas those with low anti-dsDNA levels was accompanied by a higher percentage of circulating IgD-CD27- B cells [106]. Nevertheless, anti-CD20 monoclonal antibodies have roles in refractory or frequently relapsing SLE, severe extra-renal lupus complications and steroid minimization [107,108,109,110]. The ACR guidelines also recommends that rituximab treatment in LN patients who fails to improve or worsens after 6 months of standard induction therapies, or after the patient has failed both CYC or MMF treatments [75]. The latest EULAR guideline 2019 suggests that rituximab is to be considered in SLE patients who have severe renal or extra-renal (mainly hematological and neuropsychiatric) disease refractory to other immunosuppressants and/or belimumab, or in patients with contraindications to other immunosuppressive drugs [111]. One recent phase 2 study (NOBILITY, NCT02550652) demonstrated that obinutuzumab, a new Type 2 monoclonal antibody with enhanced ADCC, in combination with corticosteroids and MMF met its primary and secondary efficacy endpoints for active proliferative LN, and a phase 3 clinical trial will be conducted shortly to verify these encouraging observations [112]. Depletion of B lymphocytes can also be achieved by targeting other B cell surface molecules such as CD19 (XmAb5871) [113] and CD22 (epratuzumab) [18,114,115], and further clinical data are required to establish their roles in SLE and LN patients. Proteosome inhibitors (PI) can effectively deplete plasma cells and hence are potentially useful treatments in SLE and LN patients. Preliminary clinical data showed that bortezomib could significantly deplete peripheral and bone marrow plasma cells and improved proteinuria and serological parameters in active LN patients who were refractory to standard induction treatments [116,117]. However, there was a high rate of treatment discontinuation due to adverse reactions. Bortezomib is currently being tested in a Phase 2 trial for SLE (NCT02102594) and ixazomib (a second-generation oral PI with less neurological side effects) is being investigated in LN patients (NCT02176486). The application of chimeric antigen receptor T cells (CAR-T) technology can potentially confer more profound and sustained B cell depletion, and is shown to be highly effective in a murine lupus model [118].
Inhibition of B cell survival factors is another therapeutic approach in SLE and LN. Treatment of NZB/W F1 mice with a soluble TACI-Ig or BAFF receptor-Ig fusion protein could suppress proteinuria and improve survival [22,119]. Belimumab is a humanized IgG1γ monoclonal antibody against soluble BAFF. One prospective cohort study showed that belimumab treatment was associated with early reduction of both naïve and autoimmunity-associated B cells (CD11c + CD21-) at 3 months, but with little impact on class-switched memory B cells and plasma cells over a follow-up duration of 3 years [120]. Results from two randomized controlled trials (RCT) (BLISS-52 and BLISS-76) showed that belimumab treatment was effective in active SLE patients, but patients with moderate to severe nephritis were excluded from these two studies [121,122]. Post hoc analysis of the pooled data from these two RCTs suggested that patients treated with belimumab showed reduced risk of renal flare [123], and therefore another RCT (BLISS-LN; NCT01639339) is underway to investigate the use of belimumab in active LN patients. Substantial and continual reduction of B lymphocyte subsets was observed beyond 76 weeks of belimumab treatment, with 80–90% reduction for naïve plasmacytoid B cells, 70–75% decrease for CD19+/CD20+ B cells, and 50–60% drop for plasma cells respectively [124]. Memory B cells showed a biphasic response to belimumab, with a rapid increase through week 8, possibly as a result of transient redistribution of these cells from lymphoid tissues to the circulation, then followed by a gradual decline [124]. Whether the lower risk of renal flares in patients receiving belimumab was associated with these changes in B cell repertoire remains speculative. Although data on belimumab in SLE and LN appeared to be quite encouraging, clinical trials on other anti-BAFF therapies showed conflicting results. Tabalumab is a monoclonal antibody targeting both soluble and membrane-bound BAFF. The results from two Phase 3 RCTs (ILLUMINATE-1 and -2) demonstrated that tabalumab treatment did not reach the primary endpoints in active non-renal SLE patients [125,126]. Post hoc analysis of the pooled data from ILLUMINATE-1 and -2 also did not reveal any benefits on renal outcomes [127]. Furthermore, the inhibition of B cell growth factors in addition to standard induction therapies should also be used with caution, as one previous Phase 2/3 study on atacicept (a human recombinant fusion protein of TACI and the Fc portion of IgG1) had early discontinuation due to significant risk of hypogammaglobulinemia and infections [128]. Other new biologics targeting the BAFF/APRIL axis include blisibimod (NCT01395745), RC18 (TACI-antibody-fusion protein; NCT02885610), AMG570 (bispecific peptibody against BAFF), and ICOS ligand (NCT02618967). In this regard, data from a phase 3 randomized, double-blind, placebo-controlled trial suggested that blisibimod did not meet its SRI-6 endpoint but there was significantly higher proportion of patients in the treatment arm achieved partial renal response and corticosteroids taper [129].
Interruption of B cell–T cell interaction represents another novel therapeutic strategy in SLE and LN. Abatacept is a fusion protein of CTLA-4 linked to the Fc portion of IgG1 and can selectively intervene the CD28-CD80/86 costimulatory signal, and thereby attenuate B cell–T cell interactions. In vitro studies showed that abatacept treatment would decrease CD80/CD86 expression and impair memory formation in human B lymphocytes [130]. Inhibition of the co-stimulatory molecule has been shown to be effective in murine models of LN [131,132,133]. Abatacept is currently approved for the treatment of rheumatoid arthritis and juvenile inflammatory arthritis [134]. Previous studies in RA patients demonstrated that treatment with abatacept was associated with selective reduction of memory B cells, and higher basal memory B cells counts were predictive of good treatment response [135]. However, active Class III or IV LN patients treated with abatacept on a background of corticosteroids and MMF showed improvements in serological markers and proteinuria but did not achieve the primary study endpoint of improving renal outcomes [136]. The negative result might be related to the potent background therapy and stringent definitions of complete renal response [137]. Nevertheless, abatacept treatment may potentially confer long-term benefits in LN patients because the selective depletion of memory B cells may decrease the risk of subsequent renal relapses. Blockade of CD40/CD40L pathway also represents another innovative approach to modulate B cell–T cell interaction and a humanized anti-CD40 monoclonal antibody (CFZ533) is now being studied in LN patients (NCT03610516). While anti-IL-6 treatment was effective in murine LN [47,48], in a phase 2 RCT anti-IL-6 treatment (PF-04236921) did not achieve the primary efficacy endpoint in patients with non-renal SLE and the higher dose (200 mg) cohort was discontinued due to safety issues [138].
Calcineurin inhibitors (CNI) and mammalian target of rapamycin (mTOR) inhibitors are emerging treatments for SLE and LN. Calcineurin inhibitors show specific suppressive actions on T lymphocytes and hence do not affect B lymphocytes directly. Instead, the effects of CNI on B cells are mediated through T cell-dependent manners such as inhibition of TFH and germinal center reactions [139,140,141]. Tacrolimus (TAC) is a CNI which has most clinical data on the management of LN to date, and current experience suggests that TAC when used in dual or triple immunosuppression showed at least comparable efficacy as CYC- or MMF-based induction and with earlier reduction of proteinuria [142,143,144,145]. A recent phase 2 study reported that a new CNI voclosporin, when combined with corticosteroids and MMF, resulted in higher renal response rates in active LN patients compared with placebo [146]. There is little clinical data on the effect of CNI on B cell biology in SLE and LN patients. Sirolimus (RAPA) is an mTOR inhibitor with suppressive effects on both B and T lymphocytes. One in vitro study showed that RAPA treatment could effectively inhibit human memory B cells proliferation and plasma cell differentiation [140]. Another recent study reported that pathogenic memory B cells isolated from treatment-naïve SLE patients showed increased mTORC1 activation, and the proliferation of these memory B cells can be significantly suppressed by RAPA treatment in vitro [147]. Furthermore, treatment of NZB/W F1 mice with RAPA could prevent development of nephritis and alleviate established nephritis, which was also accompanied by reduction in intra-renal B cell infiltration [148,149]. Whilst clinical data on the use of RAPA in patients with SLE and LN continue to emerge [150,151,152], its effect on B lymphocytes remains to be fully understood.

6. Future Directions and Concluding Remarks

The B cell repertoire shows pathogenic significance in SLE and LN, and therefore targeting B cells and plasma cells presents an appealing therapeutic approach in SLE and LN. Increasing understanding of B cell biology in SLE and LN has propelled the development of novel and more specific treatments. While many drug development programs for B cell-targeted therapies have been apparently unsuccessful either due to failure to achieve primary endpoints or because of excessive adverse events, future studies are still warranted. In this regard, attention to study design such as modification in background therapies or study endpoints, or more selective recruitment of subjects based on aberrant B cell signatures may confer a higher chance of success and inform the selection of patients for these novel therapies.
Within the bone marrow, deficiency in surrogate light chain (SLC) and impaired BCR signaling at the stage of pre-/pro-B cells will enhance survival of autoreactive B cells which contribute to the pathogenesis of systemic lupus erythematosus and lupus nephritis. In the secondary lymphoid organs (e.g., spleen or lymph nodes), naïve B cells will interact with follicular T helper (TFH) cells and further differentiate into memory B cells or plasma cells. At this stage, mechanisms which augmented the pathogenicity of B cell repertoire in SLE and LN include enhanced RAG expression and BCR signaling, increased somatic hypermutation (SHM) and class-switch recombination (CSR), aberrations in B cell transcription factors (e.g., BACH2, BLIMP1) and increased expression of B cell-related cytokines (e.g., BAFF, IL-6 and IL-21). Ways to affect the B cell repertoire include disruption of DNA synthesis (e.g., cyclophosphamide, mycophenolate mofetil, azathioprine), direct cell depletion by binding to cell surface molecules (e.g., rituximab, obinutuzumab), neutralization of B cell survival factors (e.g., belimumab, atacicept), interruption of B cell–T cell interaction (e.g., abatacept), inhibition of T cell (tacrolimus) activation and mTOR signaling (sirolimus), and inhibition of proteasome in plasma cells (e.g., bortezomib, ixazomib).
AZA, azathioprine; BAFF, B cell activating factor; BAFF-R, BAFF receptor; BCR, B cell receptor; BCMA, B-cell maturation antigen; CLP, common progenitor precursors; CSR, class-switch recombination; CYC, cyclophosphamide; mTOR, mammalian target of rapamycin; MMF, mycophenolate mofetil; RAPA, sirolimus; SHR, somatic hypermutation; TAC, tacrolimus; TACI, transmembrane activator and calcium modulator and cyclophilin ligand interactor TCR, T cell receptor; TFH, follicular T helper cells.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yap, D.Y.; Tang, C.S.; Ma, M.K.; Lam, M.F.; Chan, T.M. Survival analysis and causes of mortality in patients with lupus nephritis. Nephrol. Dial. Transplant. 2012, 27, 3248–3254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bernatsky, S.; Boivin, J.F.; Joseph, L.; Manzi, S.; Ginzler, E.; Gladman, D.D.; Urowitz, M.; Fortin, P.R.; Petri, M.; Barr, S.; et al. Mortality in systemic lupus erythematosus. Arthritis Rheum. 2006, 54, 2550–2557. [Google Scholar] [CrossRef] [PubMed]
  3. Yap, D.Y.; Chan, T.M. Lupus Nephritis in Asia: Clinical Features and Management. Kidney Dis. 2015, 1, 100–109. [Google Scholar] [CrossRef] [PubMed]
  4. Isenberg, D.; Appel, G.B.; Contreras, G.; Dooley, M.A.; Ginzler, E.M.; Jayne, D.; Sanchez-Guerrero, J.; Wofsy, D.; Yu, X.; Solomons, N. Influence of race/ethnicity on response to lupus nephritis treatment: The ALMS study. Rheumatology 2010, 49, 128–140. [Google Scholar] [CrossRef] [Green Version]
  5. Yap, D.Y.; Lai, K.N. Pathogenesis of renal disease in systemic lupus erythematosus-the role of autoantibodies and lymphocytes subset abnormalities. Int. J. Mol. Sci. 2015, 16, 7917–7931. [Google Scholar] [CrossRef]
  6. Yung, S.; Chan, T.M. Anti-dsDNA antibodies and resident renal cells-heir putative roles in pathogenesis of renal lesions in lupus nephritis. Clin. Immunol. 2017, 185, 40–50. [Google Scholar] [CrossRef]
  7. Bagavant, H.; Fu, S.M. Pathogenesis of kidney disease in systemic lupus erythematosus. Curr. Opin. Rheumatol. 2009, 21, 489–494. [Google Scholar] [CrossRef] [Green Version]
  8. Esdaile, J.M.; Abrhamowicz, M.; Joseph, L.; MacKenzie, T.; Li, Y.; Danoff, D. Laboratory tests as predictors of disease exacerbations in systemic lupus erythematosus. Why some tests fail. Arthritis Rheum. 1996, 39, 370–378. [Google Scholar] [CrossRef]
  9. Esdaile, J.M.; Joseph, L.; Abrahamowicz, M.; Li, Y.; Danoff, D.; Clarke, A.E. Routine immunologic tests in systemic lupus erythematosus: Is there a need for more studies? J. Rheumatol. 1996, 23, 1891–1896. [Google Scholar]
  10. Moroni, G.; Radice, A.; Giammarresi, G.; Qualini, S.; Gallelli, B.; Leoni, A.; Li Vecchi, M.; Messa, P.; Sinico, R.A. Are laboraotry tests useful for monitoring the activity of lupus nephritis? A 6-year prospective study in a cohort of 228 patients with lupus nephritis. Ann. Rheum. Dis. 2009, 68, 234–237. [Google Scholar] [CrossRef]
  11. Pieper, K.; Grimbacher, B.; Eibel, H. B-cell biology and development. J. Allergy Clin. Immunol. 2013, 131, 959–971. [Google Scholar] [CrossRef]
  12. Dorner, T.; Radbruch, A.; Burmester, G.R. B-cell-directed therapies for autoimmune disease. Nat. Rev. Rheumatol. 2009, 5, 433–441. [Google Scholar] [CrossRef] [PubMed]
  13. Malkiel, S.; Barlev, A.N.; Atisha-Fregoso, Y.; Suurmond, J.; Diamond, B. Plasma Cell Differentiation Pathways in Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 427. [Google Scholar] [CrossRef] [PubMed]
  14. Karrar, S.; Cunninghame Graham, D.S. Abnormal B Cell Development in Systemic Lupus Erythematosus: What the Genetics Tell Us. Arthritis Rheumatol. 2018, 70, 496–507. [Google Scholar] [CrossRef] [PubMed]
  15. Medvedovic, J.; Ebert, A.; Tagoh, H.; Busslinger, M. Pax5: A master regulator of B cell development and leukemogenesis. Adv. Immunol. 2011, 111, 179–206. [Google Scholar] [CrossRef]
  16. Itoh-Nakadai, A.; Hikota, R.; Muto, A.; Kometani, K.; Watanabe-Matsui, M.; Sato, Y.; Kobayashi, M.; Nakamura, A.; Miura, Y.; Yano, Y.; et al. The transcription repressors Bach2 and Bach1 promote B cell development by repressing the myeloid program. Nat. Immunol. 2014, 15, 1171–1180. [Google Scholar] [CrossRef]
  17. Huang, C.; Geng, H.; Boss, I.; Wang, L.; Melnick, A. Cooperative transcriptional repression by BCL6 and BACH2 in germinal center B-cell differentiation. Blood 2014, 123, 1012–1020. [Google Scholar] [CrossRef]
  18. Muto, A.; Ochiai, K.; Kimura, Y.; Itoh-Nakadai, A.; Calame, K.L.; Ikebe, D.; Tashiro, S.; Igarashi, K. Bach2 represses plasma cell gene regulatory network in B cells to promote antibody class switch. EMBO J. 2010, 29, 4048–4061. [Google Scholar] [CrossRef] [Green Version]
  19. Yap, D.Y.; Lai, K.N. The role of cytokines in the pathogenesis of systemic lupus erythematosus-from bench to bedside. Nephrology 2013, 18, 243–255. [Google Scholar] [CrossRef]
  20. Mackay, F.; Schneider, P. Cracking the BAFF code. Nat. Rev. Immunol. 2009, 9, 491–502. [Google Scholar] [CrossRef] [Green Version]
  21. Almaani, S.; Rovin, B.H. B-cell therapy in lupus nephritis: An overview. Nephrol. Dial. Transplant. 2019, 34, 22–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Gross, J.A.; Johnston, J.; Mudri, S.; Enselman, R.; Dillon, S.R.; Madden, K.; Xu, W.; Parrish-Novak, J.; Foster, D.; Lofton-Day, C.; et al. TACI and BCMA are receptors for a TNF homologue implicated in B-cell autoimmune disease. Nature 2000, 404, 995–999. [Google Scholar] [CrossRef] [PubMed]
  23. Tackey, E.; Lipsky, P.E.; Illei, G.G. Rationale for interleukin-6 blockade in systemic lupus erythematosus. Lupus 2004, 13, 339–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ding, B.B.; Bi, E.; Chen, H.; Yu, J.J.; Ye, B.H. IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp-1 in human B cells. J. Immunol. 2013, 190, 1827–1836. [Google Scholar] [CrossRef] [PubMed]
  25. Meffre, E. The establishment of early B cell tolerance in humans: Lessons from primary immunodeficiency diseases. Ann. N. Y. Acad. Sci. 2011, 1246, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Girschick, H.J.; Grammer, A.C.; Nanki, T.; Vazquez, E.; Lipsky, P.E. Expression of recombination activating genes 1 and 2 in peripheral B cells of patients with systemic lupus erythematosus. Arthritis Rheum. 2002, 46, 1255–1263. [Google Scholar] [CrossRef]
  27. Sinai, P.; Dozmorov, I.M.; Song, R.; Schwartzberg, P.L.; Wakeland, E.K.; Wulfing, C. T/B-cell interactions are more transient in response to weak stimuli in SLE-prone mice. Eur. J. Immunol. 2014, 44, 3522–3531. [Google Scholar] [CrossRef] [Green Version]
  28. Davis, D.M. Mechanisms and functions for the duration of intercellular contacts made by lymphocytes. Nat. Rev. Immunol. 2009, 9, 543–555. [Google Scholar] [CrossRef]
  29. Nashi, E.; Wang, Y.; Diamond, B. The role of B cells in lupus pathogenesis. Int. J. Biochem. Cell Biol. 2010, 42, 543–550. [Google Scholar] [CrossRef] [Green Version]
  30. Tipton, C.M.; Fucile, C.F.; Darce, J.; Chida, A.; Ichikawa, T.; Gregoretti, I.; Schieferl, S.; Hom, J.; Jenks, S.; Feldman, R.J.; et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nat. Immunol. 2015, 16, 755–765. [Google Scholar] [CrossRef] [Green Version]
  31. Boes, M.; Schmidt, T.; Linkemann, K.; Beaudette, B.C.; Marshak-Rothstein, A.; Chen, J. Accelerated development of IgG autoantibodies and autoimmune disease in the absence of secreted IgM. Proc. Natl. Acad. Sci. USA 2000, 97, 1184–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. William, J.; Euler, C.; Christensen, S.; Shlomchik, M.J. Evolution of autoantibody responses via somatic hypermutation outside of germinal centers. Science 2002, 297, 2066–2070. [Google Scholar] [CrossRef] [PubMed]
  33. Odegard, J.M.; Marks, B.R.; DiPlacido, L.D.; Poholek, A.C.; Kono, D.H.; Dong, C.; Flavell, R.A.; Craft, J. ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity. J. Exp. Med. 2008, 205, 2873–2886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Herlands, R.A.; Christensen, S.R.; Sweet, R.A.; Hershberg, U.; Shlomchik, M.J. T cell-independent and toll-like receptor-dependent antigen-driven activation of autoreactive B cells. Immunity 2008, 29, 249–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Luo, J.; Niu, X.; Liu, H.; Zhang, M.; Chen, M.; Deng, S. Up-regulation of transcription factor Blimp1 in systemic lupus erythematosus. Mol. Immunol. 2013, 56, 574–582. [Google Scholar] [CrossRef]
  36. Guimaraes, P.M.; Scavuzzi, B.M.; Stadtlober, N.P.; Franchi Santos, L.; Lozovoy, M.A.B.; Iriyoda, T.M.V.; Costa, N.T.; Reiche, E.M.V.; Maes, M.; Dichi, I.; et al. Cytokines in systemic lupus erythematosus: Far beyond Th1/Th2 dualism lupus: Cytokine profiles. Immunol. Cell Biol. 2017, 95, 824–831. [Google Scholar] [CrossRef]
  37. Salazar-Camarena, D.C.; Ortiz-Lazareno, P.C.; Cruz, A.; Oregon-Romero, E.; Machado-Contreras, J.R.; Munoz-Valle, J.F.; Orozco-Lopez, M.; Marin-Rosales, M.; Palafox-Sanchez, C.A. Association of BAFF, APRIL serum levels, BAFF-R, TACI and BCMA expression on peripheral B-cell subsets with clinical manifestations in systemic lupus erythematosus. Lupus 2016, 25, 582–592. [Google Scholar] [CrossRef]
  38. Hoyer, B.F.; Moser, K.; Hauser, A.E.; Peddinghaus, A.; Voigt, C.; Eilat, D.; Radbruch, A.; Hiepe, F.; Manz, R.A. Short-lived plasmablasts and long-lived plasma cells contribute to chronic humoral autoimmunity in NZB/W mice. J. Exp. Med. 2004, 199, 1577–1584. [Google Scholar] [CrossRef] [Green Version]
  39. Nickerson, K.M.; Christensen, S.R.; Shupe, J.; Kashgarian, M.; Kim, D.; Elkon, K.; Shlomchik, M.J. TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J. Immunol. 2010, 184, 1840–1848. [Google Scholar] [CrossRef] [Green Version]
  40. Christensen, S.R.; Shupe, J.; Nickerson, K.; Kashgarian, M.; Flavell, R.A.; Shlomchik, M.J. Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity 2006, 25, 417–428. [Google Scholar] [CrossRef] [Green Version]
  41. Mackay, F.; Woodcock, S.A.; Lawton, P.; Ambrose, C.; Baetscher, M.; Schneider, P.; Tschopp, J.; Browning, J.L. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J. Exp. Med. 1999, 190, 1697–1710. [Google Scholar] [CrossRef] [PubMed]
  42. Arkatkar, T.; Jacobs, H.M.; Du, S.W.; Li, Q.Z.; Hudkins, K.L.; Alpers, C.E.; Rawlings, D.J.; Jackson, S.W. TACI deletion protects against progressive murine lupus nephritis induced by BAFF overexpression. Kidney Int. 2018, 94, 728–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Petri, M.; Stohl, W.; Chatham, W.; McCune, W.J.; Chevrier, M.; Ryel, J.; Recta, V.; Zhong, J.; Freimuth, W. Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus. Arthritis Rheum. 2008, 58, 2453–2459. [Google Scholar] [CrossRef] [PubMed]
  44. Kitani, A.; Hara, M.; Hirose, T.; Harigai, M.; Suzuki, K.; Kawakami, M.; Kawaguchi, Y.; Hidaka, T.; Kawagoe, M.; Nakamura, H. Autostimulatory effects of IL-6 on excessive B cell differentiation in patients with systemic lupus erythematosus: Analysis of IL-6 production and IL-6R expression. Clin. Exp. Immunol. 1992, 88, 75–83. [Google Scholar] [CrossRef]
  45. Arkatkar, T.; Du, S.W.; Jacobs, H.M.; Dam, E.M.; Hou, B.; Buckner, J.H.; Rawlings, D.J.; Jackson, S.W. B cell-derived IL-6 initiates spontaneous germinal center formation during systemic autoimmunity. J. Exp. Med. 2017, 214, 3207–3217. [Google Scholar] [CrossRef]
  46. Ryffel, B.; Car, B.D.; Gunn, H.; Roman, D.; Hiestand, P.; Mihatsch, M.J. Interleukin-6 exacerbates glomerulonephritis in (NZB x NZW) F1 mice. Am. J. Pathol. 1994, 144, 927–937. [Google Scholar]
  47. Finck, B.K.; Chan, B.; Wofsy, D. Interleukin 6 promotes murine lupus in NZB/NZW F1 mice. J. Clin. Investig. 1994, 94, 585–591. [Google Scholar] [CrossRef] [Green Version]
  48. Liang, B.; Gardner, D.B.; Griswold, D.E.; Bugelski, P.J.; Song, X.Y. Anti-interleukin-6 monoclonal antibody inhibits autoimmune responses in a murine model of systemic lupus erythematosus. Immunology 2006, 119, 296–305. [Google Scholar] [CrossRef]
  49. Tsai, C.Y.; Wu, T.H.; Yu, C.L.; Lu, J.Y.; Tsai, Y.Y. Increased excretions of beta2-microglobulin, IL-6, and IL-8 and decreased excretion of Tamm-Horsfall glycoprotein in urine of patients with active lupus nephritis. Nephron 2000, 85, 207–214. [Google Scholar] [CrossRef]
  50. Herrera-Esparza, R.; Barbosa-Cisneros, O.; Villalobos-Hurtado, R.; Avalos-Diaz, E. Renal expression of IL-6 and TNFalpha genes in lupus nephritis. Lupus 1998, 7, 154–158. [Google Scholar] [CrossRef]
  51. Nakou, M.; Papadimitraki, E.D.; Fanouriakis, A.; Bertsias, G.K.; Choulaki, C.; Goulidaki, N.; Sidiropoulos, P.; Boumpas, D.T. Interleukin-21 is increased in active systemic lupus erythematosus patients and contributes to the generation of plasma B cells. Clin. Exp. Rheumatol. 2013, 31, 172–179. [Google Scholar] [PubMed]
  52. Ozaki, K.; Spolski, R.; Ettinger, R.; Kim, H.P.; Wang, G.; Qi, C.F.; Hwu, P.; Shaffer, D.J.; Akilesh, S.; Roopenian, D.C.; et al. Regulation of B cell differentiation and plasma cell generation by IL-21, a novel inducer of Blimp-1 and Bcl-6. J. Immunol. 2004, 173, 5361–5371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Bubier, J.A.; Sproule, T.J.; Foreman, O.; Spolski, R.; Shaffer, D.J.; Morse, H.C., 3rd; Leonard, W.J.; Roopenian, D.C. A critical role for IL-21 receptor signaling in the pathogenesis of systemic lupus erythematosus in BXSB-Yaa mice. Proc. Natl. Acad. Sci. USA 2009, 106, 1518–1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Herber, D.; Brown, T.P.; Liang, S.; Young, D.A.; Collins, M.; Dunussi-Joannopoulos, K. IL-21 has a pathogenic role in a lupus-prone mouse model and its blockade with IL-21R.Fc reduces disease progression. J. Immunol. 2007, 178, 3822–3830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Sawalha, A.H.; Kaufman, K.M.; Kelly, J.A.; Adler, A.J.; Aberle, T.; Kilpatrick, J.; Wakeland, E.K.; Li, Q.Z.; Wandstrat, A.E.; Karp, D.R.; et al. Genetic association of interleukin-21 polymorphisms with systemic lupus erythematosus. Ann. Rheum. Dis. 2008, 67, 458–461. [Google Scholar] [CrossRef] [Green Version]
  56. Webb, R.; Merrill, J.T.; Kelly, J.A.; Sestak, A.; Kaufman, K.M.; Langefeld, C.D.; Ziegler, J.; Kimberly, R.P.; Edberg, J.C.; Ramsey-Goldman, R.; et al. A polymorphism within IL21R confers risk for systemic lupus erythematosus. Arthritis Rheum. 2009, 60, 2402–2407. [Google Scholar] [CrossRef] [Green Version]
  57. Devarapu, S.K.; Anders, H.J. Toll-like receptors in lupus nephritis. J. Biomed. Sci. 2018, 25, 35. [Google Scholar] [CrossRef] [Green Version]
  58. Celhar, T.; Hopkins, R.; Thornhill, S.I.; De Magalhaes, R.; Hwang, S.H.; Lee, H.Y.; Yasuga, H.; Jones, L.A.; Casco, J.; Lee, B.; et al. RNA sensing by conventional dendritic cells is central to the development of lupus nephritis. Proc. Natl. Acad. Sci. USA 2015, 112, E6195–E6204. [Google Scholar] [CrossRef] [Green Version]
  59. Weindel, C.G.; Richley, L.J.; Bolland, S.; Mehta, A.J.; Kearney, J.F.; Huber, B.T. B cell autophagy mediates TLR7-dependent autoimmunity and inflammation. Autophagy 2015, 11, 1010–1024. [Google Scholar] [CrossRef] [Green Version]
  60. Applequist, S.E.; Wallin, R.P.; Ljunggren, H.G. Variable expression of toll-like receptor in murine innate and adaptive immune cell lines. Int. Immunol. 2002, 14, 1065–1074. [Google Scholar] [CrossRef] [Green Version]
  61. Lau, C.M.; Broughton, C.; Tabor, A.S.; Akira, S.; Falvell, R.A.; Mamula, M.J.; Christensen, S.R.; Shlomchik, M.J.; Viglianti, G.A.; Rifkin, I.R.; et al. RNA-associated autoantigens activate B cells by combined B cell antigen receptor/toll-like receptor 7 engagement. J. Exp. Med. 2005, 202, 1171–1177. [Google Scholar] [CrossRef]
  62. Leadbetter, E.A.; Rifkin, I.R.; Hohlbaum, A.M.; Beadudette, B.C.; Schlomchik, M.J.; Marshak-Rothstein, A. Chromatin-IgG complexes activate B cells by dual engagement of IgM and toll-like receptors. Nature 2002, 416, 603–607. [Google Scholar] [CrossRef]
  63. Boule, M.W.; Broughton, C.; Mackay, F.; Akira, S.; Marshak-Rothstein, A.; Rifkin, I.R. Toll-like receptor 9-dependent and independent dendritic cell activation by chromatin-immunoglobulin G complexes. J. Exp. Med. 2004, 199, 1631–1640. [Google Scholar] [CrossRef] [Green Version]
  64. Jackson, S.W.; Scharping, N.E.; Kolhatkar, N.S.; Khim, S.; Schwartz, M.A.; Li, Q.Z.; Hudkins, K.L.; Alpers, C.E.; Liggitt, D.; Rawlings, D.J. Opposing impact of B cell-intrinsic TLR7 and TLR9 signals on autoantibody repertoire and systemic inflammation. J. Immunol. 2014, 192, 25–32. [Google Scholar] [CrossRef] [Green Version]
  65. Capolunghi, F.; Roasado, M.M.; Cascioli, S.; Girolami, E.; Bordasco, S.; Vivarelli, M.; Ruggiero, B.; Cortis, E.; Insalaco, A.; Fanto, N.; et al. Pharmacological inhibition of TLR9 activation blocks autoantibody production in human B cells from SLE patients. Rheumatol. Oxf. 2010, 49, 2281–2289. [Google Scholar] [CrossRef] [Green Version]
  66. Chauhan, S.K.; Singh, W.; Rai, R.; Rai, M.; Rai, G. Distinct autoantibody profiles in systemic lupus erythematosus patients are selectively associated with TLR7 and TLR9 upregulation. J. Clin. Immunol. 2013, 33, 954–964. [Google Scholar] [CrossRef]
  67. Sekine, H.; Watanabe, H.; Gilkeson, G.S. Enrichment of anti-glomerular antigen antibody-producing cells in the kidneys of MRL/MpJ-Fas(lpr) mice. J. Immunol. 2004, 172, 3913–3921. [Google Scholar] [CrossRef] [Green Version]
  68. Espeli, M.; Bokers, S.; Giannico, G.; Dickinson, H.A.; Bardsley, V.; Fogo, A.B.; Smith, K.G. Local renal autoantibody production in lupus nephritis. J. Am. Soc. Nephrol. 2011, 22, 296–305. [Google Scholar] [CrossRef] [Green Version]
  69. Chan, O.T.; Hannum, L.G.; Haberman, A.M.; Madaio, M.P.; Shlomchik, M.J. 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, 189, 1639–1648. [Google Scholar] [CrossRef]
  70. Chan, O.T.; Madaio, M.P.; Shlomchik, M.J. B cells are required for lupus nephritis in the polygenic, Fas-intact MRL model of systemic autoimmunity. J. Immunol. 1999, 163, 3592–3596. [Google Scholar]
  71. Odendahl, M.; Jacobi, A.; Hansen, A.; Feist, E.; Hiepe, F.; Burmester, G.R.; Lipsky, P.E.; Radbruch, A.; Dorner, T. Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus. J. Immunol. 2000, 165, 5970–5979. [Google Scholar] [CrossRef] [Green Version]
  72. Dorner, T.; Jacobi, A.M.; Lee, J.; Lipsky, P.E. Abnormalities of B cell subsets in patients with systemic lupus erythematosus. J. Immunol. Methods 2011, 363, 187–197. [Google Scholar] [CrossRef]
  73. Tiller, T.; Tsuiji, M.; Yurasov, S.; Velinzon, K.; Nussenzweig, M.C.; Wardemann, H. Autoreactivity in human IgG+ memory B cells. Immunity 2007, 26, 205–213. [Google Scholar] [CrossRef] [Green Version]
  74. Jacobi, A.M.; Odendahl, M.; Reiter, K.; Bruns, A.; Burmester, G.R.; Radbruch, A.; Valet, G.; Lipsky, P.E.; Dorner, T. Correlation between circulating CD27high plasma cells and disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2003, 48, 1332–1342. [Google Scholar] [CrossRef]
  75. Hahn, B.H.; McMahon, M.A.; Wilkinson, A.; Wallace, W.D.; Daikh, D.I.; Fitzgerald, J.D.; Karpouzas, G.A.; Merrill, J.T.; Wallace, D.J.; Yazdany, J.; et al. American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res. 2012, 64, 797–808. [Google Scholar] [CrossRef] [Green Version]
  76. Bertsias, G.K.; Tektonidou, M.; Amoura, Z.; Aringer, M.; Bajema, I.; Berden, J.H.; Boletis, J.; Cervera, R.; Dorner, T.; Doria, A.; et al. Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis. Ann. Rheum. Dis. 2012, 71, 1771–1782. [Google Scholar] [CrossRef]
  77. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulonephritis Work Group. KDIGO Clinical Practice Guidelines for Glomerulonephritis. Kidney Int. Suppl. 2012, 2, 139–274. [Google Scholar]
  78. Mok, C.C.; Yap, D.Y.; Navarra, S.V.; Liu, Z.H.; Zhao, M.H.; Lu, L.; Takeuchi, T.; Avihingsanon, Y.; Yu, X.Q.; Lapid, E.A.; et al. Overview of lupus nephritis management guidelines and perspective from Asia. Nephrology 2014, 19, 11–20. [Google Scholar] [CrossRef]
  79. Chan, T.M.; Li, F.K.; Tang, C.S.; Wong, R.W.; Fang, G.X.; Ji, Y.L.; Lau, C.S.; Wong, A.K.; Tong, M.K.; Chan, K.W.; et al. Efficacy of mycophenolate mofetil in patients with diffuse proliferative lupus nephritis. N. Engl. J. Med. 2000, 343, 1156–1162. [Google Scholar] [CrossRef] [Green Version]
  80. Ginzler, E.M.; Dooley, M.A.; Aranow, C.; Kim, M.Y.; Buyon, J.; Merrill, J.T.; Petri, M.; Gilkeson, G.S.; Wallace, D.J.; Weisman, M.H.; et al. Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N. Engl. J. Med. 2005, 353, 2219–2228. [Google Scholar] [CrossRef] [Green Version]
  81. Contreras, G.; Pardo, V.; Leclercq, B.; Lenz, O.; Tozman, E.; O’Nan, P.; Roth, D. Sequential therapies for proliferative lupus nephritis. N. Engl. J. Med. 2004, 350, 971–980. [Google Scholar] [CrossRef] [PubMed]
  82. Appel, G.B.; Contreras, G.; Dooley, M.A.; Ginzler, E.M.; Isenberg, D.; Jayne, D.; Li, L.S.; Mysler, E.; Sanchez-Guerrero, J.; Solomons, N.; et al. Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J. Am. Soc. Nephrol. 2009, 20, 1103–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Fassbinder, T.; Saunders, U.; Mickholz, E.; Jung, E.; Becker, H.; Schluter, B.; Jacobi, A.M. Differential effects of cyclophosphamide and mycophenolate mofetil on cellular and serological parameters in patients with systemic lupus erythematosus. Arthritis Res. Ther. 2015, 17, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhao, L.; Jiang, Z.; Jiang, Y.; Ma, N.; Wang, K.; Zhang, Y. Changes in immune cell frequencies after cyclophosphamide or mycophenolate mofetil treatments in patients with systemic lupus erythematosus. Clin. Rheumatol. 2012, 31, 951–959. [Google Scholar] [CrossRef]
  85. Dooley, M.A.; Jayne, D.; Ginzler, E.M.; Isenberg, D.; Olsen, N.J.; Wofsy, D.; Eitner, F.; Appel, G.B.; Contreras, G.; Lisk, L.; et al. Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N. Engl. J. Med. 2011, 365, 1886–1895. [Google Scholar] [CrossRef] [Green Version]
  86. Yap, D.Y.; Ma, M.K.; Mok, M.M.; Tang, C.S.; Chan, T.M. Long-term data on corticosteroids and mycophenolate mofetil treatment in lupus nephritis. Rheumatology 2013, 52, 480–486. [Google Scholar] [CrossRef] [Green Version]
  87. Yap, D.Y.H.; Tang, C.; Ma, M.K.M.; Mok, M.M.Y.; Chan, G.C.W.; Kwan, L.P.Y.; Chan, T.M. Longterm Data on Disease Flares in Patients with Proliferative Lupus Nephritis in Recent Years. J. Rheumatol. 2017, 44, 1375–1383. [Google Scholar] [CrossRef]
  88. Rollinghoff, M.; Schrader, J.; Wagner, H. Effect of azathioprine and cytosine arabinoside on humoral and cellular immunity in vitro. Clin. Exp. Immunol. 1973, 15, 261–269. [Google Scholar]
  89. Gorski, A.; Korczak-Kowalska, G.; Nowaczyk, M.; Paczek, L.; Gaciong, Z. The effect of azathioprine on terminal differentiation of human B lymphocytes. Immunopharmacology 1983, 6, 259–266. [Google Scholar] [CrossRef]
  90. Eickenberg, S.; Mickholz, E.; Jung, E.; Nofer, J.R.; Pavenstadt, H.J.; Jacobi, A.M. Mycophenolic acid counteracts B cell proliferation and plasmablast formation in patients with systemic lupus erythematosus. Arthritis Res. Ther. 2012, 14, R110. [Google Scholar] [CrossRef] [Green Version]
  91. Yap, D.Y.; Lee, P.; Tam, C.; Yam, I.; Yung, S. B cell subsets and signatures in lupus nephritis patients receiving mycophenolate or azathioprine maintenance. J. Am. Soc. Nephrol. 2018, 29, 336. [Google Scholar]
  92. Yap, D.Y.; Lee, P.; Tam, C.; Yam, I.; Yung, S.; Chan, T.M. Proliferation and changes in cellular signatures in memory B cells from lupus nephritis patients receiving mycophenolate or azathioprine maintenance. J. Am. Soc. Nephrol. 2019, 30, 63. [Google Scholar]
  93. Salles, G.; Barrett, M.; Foa, R.; Maurer, J.; O’Brien, S.; Valente, N.; Wenger, M.; Maloney, D.G. Rituximab in B-Cell Hematologic Malignancies: A Review of 20 Years of Clinical Experience. Adv. Ther. 2017, 34, 2232–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Gopal, A.K.; Press, O.W. Clinical applications of anti-CD20 antibodies. J. Lab. Clin. Med. 1999, 134, 445–450. [Google Scholar] [CrossRef]
  95. Chen, D.R.; Cohen, P.L. Living life without B cells: Is repeated B-cell depletion a safe and effective long-term treatment plan for rheumatoid arthritis? Int. J. Clin. Rheumtol. 2012, 7, 159–166. [Google Scholar] [CrossRef]
  96. Boross, P.; Leusen, J.H. Mechanisms of action of CD20 antibodies. Am. J. Cancer Res. 2012, 2, 676–690. [Google Scholar]
  97. Ahuja, A.; Shupe, J.; Dunn, R.; Kashgarian, M.; Kehry, M.R.; Shlomchik, M.J. Depletion of B cells in murine lupus: Efficacy and resistance. J. Immunol. 2007, 179, 3351–3361. [Google Scholar] [CrossRef]
  98. Bekar, K.W.; Owen, T.; Dunn, R.; Ichikawa, T.; Wang, W.; Wang, R.; Barnard, J.; Brady, S.; Nevarez, S.; Goldman, B.I.; et al. Prolonged effects of short-term anti-CD20 B cell depletion therapy in murine systemic lupus erythematosus. Arthritis Rheum. 2010, 62, 2443–2457. [Google Scholar] [CrossRef] [Green Version]
  99. Albert, D.; Dunham, J.; Khan, S.; Stansberry, J.; Kolasinski, S.; Tsai, D.; Pullman-Mooar, S.; Bamack, F.; Striebich, C.; Looney, R.J.; et al. Variability in the biological response to anti-CD20 B cell depletion in systemic lupus erythematosus. Ann. Rheum. Dis. 2008, 67, 1724–1731. [Google Scholar] [CrossRef]
  100. Gomez Mendez, L.M.; Cascino, M.D.; Garg, J.; Katsumoto, T.R.; Brakeman, P.; Dall’Eram, M.; Looney, R.J.; Rovin, B.; Deagone, L.; Brunetta, P. Peripheral Blood B Cell Depletion after Rituximab and Complete Response in Lupus Nephritis. Clin. J. Am. Soc. Nephrol. 2018, 13, 1502–1509. [Google Scholar] [CrossRef] [Green Version]
  101. Vital, E.M.; Dass, S.; Buch, M.H.; Henshaw, K.; Pease, C.T.; Martin, M.F.; Ponchel, F.; Rawstron, A.C.; Emery, P. B cell biomarkers of rituximab responses in systemic erythematosus. Arthritis Rheum. 2011, 63, 3038–3047. [Google Scholar] [CrossRef] [PubMed]
  102. Looney, R.J.; Anolik, J.H.; Campbell, D.; Felgar, R.E.; Young, F.; Arend, L.J.; Sloand, J.A.; Rosenblatt, J.; Sanz, I. B cell depletion as a novel treatment for systemic lupus erythematosus: A phase I/II dose-escalation trial of rituximab. Arthritis Rheum. 2004, 50, 2580–2589. [Google Scholar] [CrossRef] [PubMed]
  103. Merrill, J.T.; Neuwelt, C.M.; Wallace, D.J.; Shanahan, J.C.; Latinis, K.M.; Oates, J.C.; Utset, T.O.; Gordon, C.; Isenberg, D.A.; Hsieh, H.J.; 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, 62, 222–233. [Google Scholar] [CrossRef] [PubMed]
  104. Rovin, B.H.; Furie, R.; Latinis, K.; Looney, R.J.; Fervenza, F.C.; Sanchez-Guerrero, J.; Maciuca, R.; Zhang, D.; Garg, J.P.; Brunetta, P.; et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: The Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012, 64, 1215–1226. [Google Scholar] [CrossRef] [PubMed]
  105. Md Yusof, M.Y.; Shaw, D.; El-Sherbiny, Y.M.; Dunn, E.; Rawstron, A.C.; Emery, P.; Vital, E.M. Predicting and managing primary and secondary non-response to rituximab using B-cell biomarkers in systemic lupus erythematosus. Ann. Rheum. Dis. 2017, 76, 1829–1836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lazarus, M.N.; Turner-Stokes, T.; Chavele, K.M.; Isenberg, D.A.; Ehrenstein, M.R. B-cell numbers and phenotype at clinical relapse following rituximab therapy differ in SLE patients according to anti-dsDNA antibody levels. Rheumatology 2012, 51, 1208–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Weidenbusch, M.; Rommele, C.; Schrottle, A.; Anders, H.J. Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol. Dial. Transplant. 2013, 28, 106–111. [Google Scholar] [CrossRef] [Green Version]
  108. Diaz-Lagares, C.; Croca, S.; Sangle, S.; Vital, E.M.; Catapano, F.; Martinez-Berriotxoa, A.; Garcia-Hernandez, F.; Callejas-Rubio, J.L.; Rascon, J.; D’Cruz, D.; et al. Efficacy of rituximab in 164 patients with biopsy-proven lupus nephritis: Pooled data from European cohorts. Autoimmun. Rev. 2012, 11, 357–364. [Google Scholar] [CrossRef]
  109. Condon, M.B.; Ashby, D.; Pepper, R.J.; Cook, H.T.; Levy, J.B.; Griffith, M.; Cairns, T.D.; Lightstone, L. Prospective observational single-centre cohort study to evaluate the effectiveness of treating lupus nephritis with rituximab and mycophenolate mofetil but no oral steroids. Ann. Rheum. Dis. 2013, 72, 1280–1286. [Google Scholar] [CrossRef]
  110. Pepper, R.; Griffith, M.; Kirwan, C.; Levy, J.; Taube, D.; Pusey, C.; Lightstone, L.; Cairns, T. Rituximab is an effective treatment for lupus nephritis and allows a reduction in maintenance steroids. Nephrol. Dial. Transplant. 2009, 24, 3717–3723. [Google Scholar] [CrossRef] [Green Version]
  111. Fanouriakis, A.; Kostopoulou, M.; Alunno, A.; Aringer, M.; Bajema, I.; Boletis, J.N.; Cervera, R.; Doria, A.; Gordon, C.; Govoni, M.; et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann. Rheum. Dis. 2019, 78, 736–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Rovin, B.; Aroca Martinez, G.; Alvarez, A.; Fragoso Ioyo, H.E.; Zuta, A.E.; Furie, R.; Brunetta, P.; Schindler, T.; Hassan, I.; Cascino, M.; et al. A Phase 2 Randomized, Controlled Study of Obinutuzumab with Mycophenolate and Corticosteroids in Proliferative Lupus Nephritis. In Proceedings of the American Society of Nephrology Kidney Week 2019, FR-OR136, Washington, DC, USA, 5–10 November 2019. [Google Scholar]
  113. Merrill, J.T.; June, J.; Koumpouras, F.; Machua, W.; Khan, M.F.; Askanase, A.D.; Sheikh, S.Z.; Khosroshahi, A.; Foster, P.; Zack, D.J. Results of a phase 2, double-blind, randomized, placebo-controlled study of a reversible B cell inhibitor, XmAb5871, in systemic lupus erythematosus. Lupus Sci. Med. 2019, 6, A1–A227. [Google Scholar]
  114. Wallace, D.J.; Gordon, C.; Strand, V.; Hobbs, K.; Petri, M.; Kalunian, K.; Houssiau, F.; Tak, P.P.; Isenberg, D.A.; Kelley, L.; et al. Efficacy and safety of epratuzumab in patients with moderate/severe flaring systemic lupus erythematosus: Results from two randomized, double-blind, placebo-controlled, multicentre studies (ALLEVIATE) and follow-up. Rheumatology 2013, 52, 1313–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Clowse, M.E.; Wallace, D.J.; Furie, R.A.; Petri, M.A.; Pike, M.C.; Leszczyński, P.; Neuwelt, C.M.; Hobbs, K.; Keiserman, M.; Duca, L.; et al. Efficacy and Safety of Epratuzumab in Moderately to Severely Active Systemic Lupus Erythematosus: Results from Two Phase III Randomized, Double-Blind, Placebo-Controlled Trials. Arthritis Rheumatol. 2017, 69, 362–375. [Google Scholar]
  116. Alexander, T.; Sarfert, R.; Klotsche, J.; Kuhl, A.A.; Rubbert-Roth, A.; Lorenz, H.M.; Rech, J.; Hoyer, B.F.; Cheng, Q.; Waka, A.; et al. The proteasome inhibitor bortezomib depletes plasma cells and ameliorates clinical manifestations of refractory systemic lupus erythematosus. Ann. Rheum. Dis. 2015, 74, 1474–1478. [Google Scholar] [CrossRef]
  117. Zhang, H.; Liu, Z.; Huang, L.; Hou, J.; Zhou, M.; Huang, X.; Hu, W.; Liu, Z. The short-term efficacy of bortezomib combined with glucocorticoids for the treatment of refractory lupus nephritis. Lupus 2017, 26, 952–958. [Google Scholar] [CrossRef] [PubMed]
  118. Kansal, R.; Richardson, N.; Neeli, I.; Khawaja, S.; Chamberlain, D.; Ghani, M.; Ghani, Q.U.; Balazs, L.; Beranova-Giorgianni, S.; Giorgianni, F.; et al. Sustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupus. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef]
  119. Kayagaki, N.; Yan, M.; Seshasayee, D.; Wang, H.; Lee, W.; French, D.M.; Grewal, I.S.; Cochran, A.G.; Gordon, N.C.; Yin, J.; et al. BAFF/BLyS receptor 3 binds the B cell survival factor BAFF ligand through a discrete surface loop and promotes processing of NF-kappaB2. Immunity 2002, 17, 515–524. [Google Scholar] [CrossRef] [Green Version]
  120. Ramskold, D.; Parodis, I.; Lakshmikanth, T.; Sippl, N.; Khademi, M.; Chen, Y.; Zickert, A.; Mikes, J.; Achour, A.; Amara, K.; et al. B cell alterations during BAFF inhibition with belimumab in SLE. EBioMedicine 2019, 40, 517–527. [Google Scholar] [CrossRef] [Green Version]
  121. Navarra, S.V.; Guzman, R.M.; Gallacher, A.E.; Hall, S.; Levy, R.A.; Jimenez, R.E.; Li, E.K.; Thomas, M.; Kim, H.Y.; Leon, M.G.; et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: A randomised, placebo-controlled, phase 3 trial. Lancet 2011, 377, 721–731. [Google Scholar] [CrossRef]
  122. Furie, R.; Petri, M.; Zamani, O.; Cervera, R.; Wallace, D.J.; Tegzova, D.; Sanchez-Guerrero, J.; Schwarting, A.; Merrill, J.T.; Chatham, W.W.; et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011, 63, 3918–3930. [Google Scholar] [CrossRef] [PubMed]
  123. Dooley, M.A.; Houssiau, F.; Aranow, C.; D’Cruz, D.P.; Askanase, A.; Roth, D.A.; Zhong, Z.J.; Cooper, S.; Freimuth, W.W.; Ginzler, E.M.; et al. Effect of belimumab treatment on renal outcomes: Results from the phase 3 belimumab clinical trials in patients with SLE. Lupus 2013, 22, 63–72. [Google Scholar] [CrossRef] [PubMed]
  124. Stohl, W.; Hiepe, F.; Latinis, K.M.; Thomas, M.; Scheinberg, M.A.; Clarke, A.; Aranow, C.; Wellborne, F.R.; Abud-Mendoza, C.; Hough, D.R.; et al. Belimumab reduces autoantibodies, normalizes low complement levels, and reduces select B cell populations in patients with systemic lupus erythematosus. Arthritis Rheum. 2012, 64, 2328–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Isenberg, D.A.; Petri, M.; Kalunian, K.; Tanaka, Y.; Urowitz, M.B.; Hoffman, R.W.; Morgan-Cox, M.; Iikuni, N.; Silk, M.; Wallace, D.J. Efficacy and safety of subcutaneous tabalumab in patients with systemic lupus erythematosus: Results from ILLUMINATE-1, a 52-week, phase III, multicentre, randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016, 75, 323–331. [Google Scholar] [CrossRef] [PubMed]
  126. Merrill, J.T.; van Vollenhoven, R.F.; Buyon, J.P.; Furie, R.A.; Stohl, W.; Morgan-Cox, M.; Dickson, C.; Anderson, P.W.; Lee, C.; Berclaz, P.Y.; et al. Efficacy and safety of subcutaneous tabalumab, a monoclonal antibody to B-cell activating factor, in patients with systemic lupus erythematosus: Results from ILLUMINATE-2, a 52-week, phase III, multicentre, randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016, 75, 332–340. [Google Scholar] [CrossRef] [Green Version]
  127. Rovin, B.H.; Dooley, M.A.; Radhakrishnan, J.; Ginzler, E.M.; Forrester, T.D.; Anderson, P.W. The impact of tabalumab on the kidney in systemic lupus erythematosus: Results from two phase 3 randomized, clinical trials. Lupus 2016, 25, 1597–1601. [Google Scholar] [CrossRef]
  128. Ginzler, E.M.; Wax, S.; Rajeswaran, A.; Copt, S.; Hillson, J.; Ramos, E.; Singer, N.G. Atacicept in combination with MMF and corticosteroids in lupus nephritis: Results of a prematurely terminated trial. Arthritis Res. Ther. 2012, 14, R33. [Google Scholar] [CrossRef] [Green Version]
  129. Merrill, J.T.; Shanahan, W.R.; Scheinberg, M.; Kalunian, K.C.; Wofsy, D.; Martin, R.S. Phase III trial results with blisibimod, a selective inhibitor of B-cell activating factor, in subjects with systemic lupus erythematosus (SLE): Results from a randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2018, 77, 883–889. [Google Scholar] [CrossRef]
  130. Lorenzetti, R.; Janowska, I.; Smulski, C.R.; Frede, N.; Henneberger, N.; Walter, L.; Schleyer, M.T.; Huppe, J.M.; Staniek, J.; Salzer, U.; et al. Abatacept modulates CD80 and CD86 expression and memory formation in human B-cells. J. Autoimmun. 2019, 101, 145–152. [Google Scholar] [CrossRef]
  131. Daikh, D.I.; Finck, B.K.; Linsley, P.S.; Hollenbaugh, D.; Wofsy, D. Long-term inhibition of murine lupus by brief simultaneous blockade of the B7/CD28 and CD40/gp39 costimulation pathways. J. Immunol. 1997, 159, 3104–3108. [Google Scholar]
  132. Daikh, D.I.; Wofsy, D. Cutting edge: Reversal of murine lupus nephritis with CTLA4Ig and cyclophosphamide. J. Immunol. 2001, 166, 2913–2916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Schiffer, L.; Sinha, J.; Wang, X.; Huang, W.; von Gersdorff, G.; Schiffer, M.; Madaio, M.P.; Davidson, A. Short term administration of costimulatory blockade and cyclophosphamide induces remission of systemic lupus erythematosus nephritis in NZB/W F1 mice by a mechanism downstream of renal immune complex deposition. J. Immunol. 2003, 171, 489–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Keating, G.M. Abatacept: A review of its use in the management of rheumatoid arthritis. Drugs 2013, 73, 1095–1119. [Google Scholar] [CrossRef] [PubMed]
  135. Gazeau, P.; Alegria, G.C.; Devauchelle-Pensec, V.; Jamin, C.; Lemerle, J.; Bendaoud, B.; Brooks, W.H.; Saraux, A.; Cornec, D.; Renaudineau, Y. Memory B Cells and Response to Abatacept in Rheumatoid Arthritis. Clin. Rev. Allergy Immunol. 2017, 53, 166–176. [Google Scholar] [CrossRef]
  136. Furie, R.; Nicholls, K.; Cheng, T.T.; Houssiau, F.; Burgos-Vargas, R.; Chen, S.L.; Hillson, J.L.; Meadows-Shropshire, S.; Kinaszczuk, M.; Merrill, J.T. Efficacy and safety of abatacept in lupus nephritis: A twelve-month, randomized, double-blind study. Arthritis Rheumatol. 2014, 66, 379–389. [Google Scholar] [CrossRef]
  137. Wofsy, D.; Hillson, J.L.; Diamond, B. Abatacept for lupus nephritis: Alternative definitions of complete response support conflicting conclusions. Arthritis Rheum. 2012, 64, 3660–3665. [Google Scholar] [CrossRef] [Green Version]
  138. Wallace, D.J.; Strand, V.; Merrill, J.T.; Popa, S.; Spindler, A.J.; Eimon, A.; Petri, M.; Smolen, J.S.; Wajdula, J.; Christensen, J.; et al. Efficacy and safety of an interleukin 6 monoclonal antibody for the treatment of systemic lupus erythematosus: A phase II dose-ranging randomised controlled trial. Ann. Rheum. Dis. 2017, 76, 534–542. [Google Scholar] [CrossRef] [Green Version]
  139. Heidt, S.; Roelen, D.L.; Eijsink, C.; Eikmans, M.; van Kooten, C.; Claas, F.H.; Mulder, A. Calcineurin inhibitors affect B cell antibody responses indirectly by interfering with T cell help. Clin. Exp. Immunol. 2010, 159, 199–207. [Google Scholar] [CrossRef]
  140. Traitanon, O.; Mathew, J.M.; La Monica, G.; Xu, L.; Mas, V.; Gallon, L. Differential Effects of Tacrolimus versus Sirolimus on the Proliferation, Activation and Differentiation of Human B Cells. PLoS ONE 2015, 10, e0129658. [Google Scholar] [CrossRef] [Green Version]
  141. Wallin, E.F.; Hill, D.L.; Linterman, M.A.; Wood, K.J. The Calcineurin Inhibitor Tacrolimus Specifically Suppresses Human T Follicular Helper Cells. Front. Immunol. 2018, 9, 1184. [Google Scholar] [CrossRef] [Green Version]
  142. Chen, W.; Tang, X.; Liu, Q.; Chen, W.; Fu, P.; Liu, F.; Liao, Y.; Yang, Z.; Zhang, J.; Chen, J.; et al. Short-term outcomes of induction therapy with tacrolimus versus cyclophosphamide for active lupus nephritis: A multicenter randomized clinical trial. Am. J. Kidney Dis. 2011, 57, 235–244. [Google Scholar] [CrossRef] [PubMed]
  143. Mok, C.C.; Ying, K.Y.; Yim, C.W.; Siu, Y.P.; Tong, K.H.; To, C.H.; Ng, W.L. Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: A randomised controlled trial and long-term follow-up. Ann. Rheum. Dis. 2016, 75, 30–36. [Google Scholar] [CrossRef] [PubMed]
  144. Bao, H.; Liu, Z.H.; Xie, H.L.; Hu, W.X.; Zhang, H.T.; Li, L.S. Successful treatment of class V+IV lupus nephritis with multitarget therapy. J. Am. Soc. Nephrol. 2008, 19, 2001–2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, Z.; Zhang, H.; Liu, Z.; Xing, C.; Fu, P.; Ni, Z.; Chen, J.; Lin, H.; Liu, F.; He, Y.; et al. Multitarget therapy for induction treatment of lupus nephritis: A randomized trial. Ann. Intern. Med. 2015, 162, 18–26. [Google Scholar] [CrossRef] [PubMed]
  146. Rovin, B.H.; Solomons, N.; Pendergraft, W.F.; Dooley, M.A.; Tumlin, J.; Romero-Diaz, J.; Lysenko, L.; Navarra, S.V.; Huizinga, R.B.; Aura-Lv Study Group. A randomized, controlled double-blind study comparing the efficacy and safety of dose-ranging voclosporin with placebo in achieving remission in patients with active lupus nephritis. Kidney Int. 2019, 95, 219–231. [Google Scholar] [CrossRef]
  147. Wu, C.; Fu, Q.; Guo, Q.; Chen, S.; Goswami, S.; Sun, S.; Li, T.; Cao, X.; Chu, F.; Chen, Z.; et al. Lupus-associated atypical memory B cells are mTORC1-hyperactivated and functionally dysregulated. Ann. Rheum. Dis. 2019, 78, 1090–1100. [Google Scholar] [CrossRef] [Green Version]
  148. Lui, S.L.; Yung, S.; Tsang, R.; Zhang, F.; Chan, K.W.; Tam, S.; Chan, T.M. Rapamycin prevents the development of nephritis in lupus-prone NZB/W F1 mice. Lupus 2008, 17, 305–313. [Google Scholar] [CrossRef]
  149. Lui, S.L.; Tsang, R.; Chan, K.W.; Zhang, F.; Tam, S.; Yung, S.; Chan, T.M. Rapamycin attenuates the severity of established nephritis in lupus-prone NZB/W F1 mice. Nephrol. Dial. Transplant. 2008, 23, 2768–2776. [Google Scholar] [CrossRef] [Green Version]
  150. Yap, D.Y.; Ma, M.K.; Tang, C.S.; Chan, T.M. Proliferation signal inhibitors in the treatment of lupus nephritis: Preliminary experience. Nephrology 2012, 17, 676–680. [Google Scholar] [CrossRef]
  151. Lai, Z.W.; Kelly, R.; Winans, T.; Marchena, I.; Shadakshari, A.; Yu, J.; Dawood, M.; Garcia, R.; Tily, H.; Francis, L.; et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: A single-arm, open-label, phase 1/2 trial. Lancet 2018, 391, 1186–1196. [Google Scholar] [CrossRef]
  152. Yap, D.Y.H.; Tang, C.; Chan, G.C.W.; Kwan, L.P.Y.; Ma, M.K.M.; Mok, M.M.Y.; Chan, T.M. Longterm Data on Sirolimus Treatment in Patients with Lupus Nephritis. J. Rheumatol. 2018, 45, 1663–1670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic diagram showing the B cell abnormalities in systemic lupus erythematosus and the potential therapeutic targets.
Figure 1. Schematic diagram showing the B cell abnormalities in systemic lupus erythematosus and the potential therapeutic targets.
Ijms 20 06231 g001
Table 1. The effect of currently available immunosuppressive medications on B cells.
Table 1. The effect of currently available immunosuppressive medications on B cells.
DrugsMechanisms of ActionEffect on B Cells
CYCDisrupts DNA replication and thus confers cytotoxic effect on actively proliferating cells including lymphocytesPreferentially depletes less mature B cells (e.g., naïve B and pre-switched memory B cells) compared with MMF.
Little effect on class-switched memory B cells
MMFInhibits IMPDH and therefore selectively blocks de novo purine synthesis in B and T lymphocytesEarlier reduction of circulating plasmablasts compared with CYC but with little effect on class-switched memory B cells
More potent than AZA in suppressing naïve and memory B cell proliferation
AZAConverts to 6-mercaptopurine and interferes with DNA replication and purine synthesis in lymphocytesAnimal data shows higher AZA dose required to suppress humoral immunity than that required to suppress cellular immunity
TACInhibits IL-2 production and thus T cell activation and proliferationInhibits TFH and GC formation, thereby impairs B cell maturation and antibody production
RAPAInhibits the activation of mTOR signals in lymphocytesSuppresses proliferation of different B cell subsets (especially memory B cell with ↑mTORC1 activation).
Blocks differentiation of B cells into plasma cells.
↓intra-renal B cell infiltration in murine LN models
RituximabBinds to CD20 on B cells, leading to ADCC, CDC and ↑apoptosis of B cellsProfoundly depletes different B subsets except plasma cells within 2 weeks.
B cell reconstitution occurs at approximately 6–9 months
BelimumabInhibits BAFF and hence survival and maturation of B cellsSustained reduction in naïve plasmacytoid B cells (80–90%), CD19+/CD20+ B cells (70–75%) and plasma cells (50–60%).
AbataceptInterruption of co-stimulatory signal for B cell–T cell interactionPreferentially suppresses memory B cells
ADCC, antibody-dependent cell-mediated cytotoxicity; AZA, azathioprine; BAFF, B cell activating factor; CDC, complement-dependent cytotoxicity; CYC, cyclophosphamide; GC, germinal center; IL-2, interleukin-2; IMPDH, inosine monophosphate dehydrogenase; MMF, mycophenolate mofetil, mTORC, mammalian target of rapamycin; RAPA, sirolimus; TAC, tacrolimus; TFH, follicular T helper cells. ↑: increase; ↓: decrease.

Share and Cite

MDPI and ACS Style

Yap, D.Y.H.; Chan, T.M. B Cell Abnormalities in Systemic Lupus Erythematosus and Lupus Nephritis—Role in Pathogenesis and Effect of Immunosuppressive Treatments. Int. J. Mol. Sci. 2019, 20, 6231. https://doi.org/10.3390/ijms20246231

AMA Style

Yap DYH, Chan TM. B Cell Abnormalities in Systemic Lupus Erythematosus and Lupus Nephritis—Role in Pathogenesis and Effect of Immunosuppressive Treatments. International Journal of Molecular Sciences. 2019; 20(24):6231. https://doi.org/10.3390/ijms20246231

Chicago/Turabian Style

Yap, Desmond Y. H., and Tak Mao Chan. 2019. "B Cell Abnormalities in Systemic Lupus Erythematosus and Lupus Nephritis—Role in Pathogenesis and Effect of Immunosuppressive Treatments" International Journal of Molecular Sciences 20, no. 24: 6231. https://doi.org/10.3390/ijms20246231

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop