Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 1.
Published in final edited form as: Immunol Rev. 2019 Sep 28;292(1):102–119. doi: 10.1111/imr.12810

BAFF inhibition in SLE – is tolerance restored?

Shaun W Jackson 1,2, Anne Davidson 3
PMCID: PMC6935406  NIHMSID: NIHMS1050839  PMID: 31562657

Summary:

The B cell activating factor (BAFF) inhibitor, belimumab, is the first biologic drug approved for the treatment of SLE, and exhibits modest, but durable, efficacy in decreasing disease flares and organ damage. BAFF and its homolog APRIL are TNF-like cytokines that support the survival and differentiation of B cells at distinct developmental stages. BAFF is a crucial survival factor for transitional and mature B cells that acts as rheostat for the maturation of low-affinity autoreactive cells. In addition, BAFF augments innate B cell responses via complex interactions with the B cell receptor (BCR) and Toll like receptor (TLR) pathways. In this manner, BAFF impacts autoreactive B cell activation via extrafollicular pathways and fine tunes affinity selection within germinal centers (GC). Finally, BAFF and APRIL support plasma cell survival, with differential impacts on IgM- and IgG-producing populations. Therapeutically, BAFF and combined BAFF/APRIL inhibition delays disease onset in diverse murine lupus strains, although responsiveness to BAFF inhibition is model dependent, in keeping with heterogeneity in clinical responses to belimumab treatment in humans. In this review, we discuss the mechanisms whereby BAFF/APRIL signals promote autoreactive B cell activation, discuss whether altered selection accounts for therapeutic benefits of BAFF inhibition, and address whether new insights into BAFF/APRIL family complexity can be exploited to improve human lupus treatments.

Keywords: SLE, B cells, BAFF, selection, tolerance

1. Introduction

Systemic lupus erythematosus (SLE – lupus) is a severe autoimmune disease in which loss of B cell tolerance to ubiquitous nuclear antigens causes autoantibody-mediated organ damage. Despite the continuing acquisition of new knowledge regarding lupus pathogenesis, numerous biologic drugs have failed to achieve a significant clinical response in rationally designed trials using large patient cohorts 1. Therefore, much remains to be learned regarding the mechanisms underlying autoreactive B cell activation and strategies to restore B cell tolerance in lupus patients.

The association of high levels of BAFF with lupus susceptibility was fueled by the observation that two independently derived strains of BAFF transgenic mice manifested B cell hyperplasia, anti-dsDNA antibodies and a lupus-like illness with renal immune complex deposits. BAFF levels were found to be elevated in murine lupus models and disease was significantly attenuated by BAFF inhibition 24. Once it was established that patients with SLE also exhibit increased serum BAFF 5, 6, therapeutics that target BAFF and its receptors were developed. This work culminated in the FDA approval of the human monoclonal anti-BAFF antibody, belimumab, for the treatment of SLE - the first approved application of a biologic drug in human lupus.

Despite the reproducible clinical efficacy of belimumab in several trials and convincing longitudinal data showing that it decreases lupus flare rates over long periods of time 7, clinical efficacy is only moderate and not all patients respond 8, 9. Thus, many questions remain regarding the mechanism of action of belimumab and about the best strategy to target BAFF, its homologous cytokine APRIL, and the three BAFF family receptors in order to maximize clinical benefit. In this review, we summarize recent insights into the impact of BAFF and APRIL on B cell subsets in SLE, focusing on our own data, and speculate about the potential immune mechanisms underlying BAFF’s roles in lupus pathogenesis.

2. The complexity of the BAFF-APRIL system (Figure 1)

Figure 1: BAFF, APRIL, and their receptors and splice variants:

Figure 1:

Both BAFF and APRIL multimerize: BAFF multimerizes into 60-mers via its flap region whereas APRIL multimerizes on cells surfaces by binding to proteoglycans. TACI and BCMA both shed soluble forms that can act as decoy receptors and human TACI has an intracellular short form that preferentially interacts with TLR9. Therapeutic approaches are displayed in purple boxes. Specific targeting of the flap region of BAFF should prevent multimer formation that is required for optimal signaling through all three receptors. This is more likely to target membrane BAFF-TACI and BAFF-BCMA interactions and may help to preserve B cell numbers while allowing the continued function of the soluble decoy receptors 13. Given relatively restricted expression on B cells, BAFF family receptors are potential targets for chimeric antigen receptor (CAR) T cell cancer therapies. Specifically, anti-BAFF-R 143 and anti-BCMA 144 CARs are being developed for the treatment of B cell leukemia/lymphoma and multiple myeloma, respectively, while APRIL-expressing CARs target TACI and BCMA expressing cells 145. Although currently restricted to oncology trials, these therapies could potentially be used to target B cell subsets in SLE.

2.1. The BAFF family cytokines and receptors

BAFF:

BAFF is a type II transmembrane protein produced predominantly by stromal and myeloid cells that initially assembles as a homotrimer on the cell membrane but is subsequently cleaved by furin protease to release the soluble trimeric cytokine 10. BAFF trimers can also multimerize via a loop region of 10 amino acids (flap) to form 60-mers 11, 12. BAFF has three receptors that are expressed predominantly by B cells, BAFF-R, TACI and BCMA. While trimeric BAFF induces signaling via BAFF-R, TACI is only activated by the higher-order multimers. The flap region is not required for binding to any known receptor but it is necessary for optimal signaling after binding of the soluble BAFF trimer to BAFF-R, suggesting that a degree of multimerization is required even for BAFF-R activation 13. Alternate BAFF forms include BAFF-APRIL heterotrimers and a splice variant called ΔBAFF, which is inefficiently cleaved from the cell surface and serves as an antagonist via heteromultimerization with BAFF 14. As detailed below, a human polymorphism of the BAFF gene that increases BAFF expression by ~1.5–2 fold is associated with a modest increase in susceptibility to certain autoimmune diseases, including SLE 15.

APRIL:

APRIL is a type II transmembrane protein produced by multiple cell types including myeloid cells, epithelial cells, and tumor cells 16. APRIL is cleaved within the Golgi apparatus and does not usually appear on the cell membrane 17. However, the APRIL gene is located adjacent to the TWEAK gene such that hybrid molecules comprising the cytoplasmic domain of TWEAK and the extracellular domain of APRIL may be generated and expressed on the cell surface 18. APRIL also appears to have a membrane-bound splice isoform 19. APRIL binds to BCMA, TACI, and to surface heparin sulfate proteoglycan (HSPG), which serves to multimerize APRIL and thereby allow downstream signaling following receptor ligation. Additional HSPG binding to TACI facilitates APRIL-TACI-HSPG interactions, resulting in complex impacts on BAFF family receptor activation. While APRIL deficient mice have normal immune development, class switching to IgA and maintenance of serum IgA levels is highly APRIL-dependent 20, 21. In addition, APRIL supports the survival of long-loved plasma cells (LLPC) in the bone marrow, especially during neonatal development 21.

BAFF family receptors:

BAFF-R, TACI, and BCMA are the three known surface receptors for BAFF and APRIL. Each receptor exhibits a unique expression pattern on distinct B cell subsets, in part accounting for differential functional roles during an immune response. In addition, alternative splice isoforms of BCMA and TACI have been reported 22. All three of the BAFF/APRIL receptors can be shed from the cell surface by the activity of γ-secretase (BCMA) or ADAM proteases (BAFF-R and TACI) 23, 24. Shedding of BAFF-R and TACI is dependent on receptor ligation, B cell subset, B cell activation state, and, for BAFF-R, the coexpression of TACI 25. Importantly, constitutive cleavage of BCMA and TACI generates functional decoy receptors, whereas BAFF-R shedding does not appear to generate a decoy but rather regulates B cell survival via modulation of surface BAFF-R expression 26.

BAFF-R:

BAFF-R expression is first observed at the transitional stage of B cell maturation. However, low-level BAFF-R expression has been observed on bone marrow B cells, consistent with evidence that BAFF-R engagement mediates positive selection of developing B cells prior to the transitional type 2 (T2) stage, especially those lacking BCR engagement, thereby skewing the emerging repertoire against autoreactivity 27, 28. In contrast, BAFF/BAFF-R interactions are critical for T2 selection and naïve B cell survival. In these cells, BAFF-R and BCR signals cooperate to reinforce each signaling pathway and inhibit apoptosis 2931. Unlike other BAFF/APRIL receptors, BAFF-R signals through the alternative NFκB pathway, for which BCR engagement provides the essential intermediary p100 29, 32, 33. BAFF-R ligation also activates the phosphoinositide-3-kinase-dependent signaling cascade to enhance protein synthesis and mediate metabolic reprogramming in order to facilitate cell survival (reviewed in 26). Importantly, since immature and anergic B cells exhibit lower surface BCR expression, these cells exhibit an increased requirement for BAFF-R mediated survival signals. In this manner, BAFF functions as a rheostat for naïve B cell selection at the transitional stage.

As described in detail below, BAFF-R is downregulated on GC B cells, but is re-expressed on memory B cells 25. While the function of BAFF-R in memory B cells remains incompletely defined, IgM+ memory B cells exhibit partial BAFF/BAFF-R dependence, whereas IgG+ memory B cells require neither BAFF nor APRIL for their survival 34, 35. Finally, BAFF-R is also expressed on a subset of T cells and may function to modulate T cell activation and cytokine production (reviewed 36).

TACI:

TACI is expressed by all mature peripheral B cells including marginal zone, B1 B cells, and plasma cells. Engagement with BAFF and APRIL multimers promotes TACI signaling via classical NFκB, Mek and Jnk/p38 pathways to counteract apoptosis, drive immunoglobulin class switch recombination, and promote antibody production. The interaction of TACI with APRIL preferentially supports IgA responses 21, 37. TACI is required for T-independent responses but appears dispensable for the initiation of T dependent responses 37, 38. Nevertheless, TACI serves to maintain BLIMP-1 expression and thus supports the differentiation and survival of long-lived plasma cells 39. While TACI signals promote T cell-independent B cell activation, TACI deficient mice exhibit B cell hyperplasia and mild autoimmunity, suggesting a potential negative regulatory role in B cell activation 37, 38. In addition to a direct regulatory function, loss of surface TACI or of the soluble decoy receptor may account for B cell hyperplasia because of reciprocally increased serum BAFF and APRIL levels 4042. In contrast to mice, humans express both short and long isoforms of TACI. Functionally, short isoform TACI is induced by TLR9 agonists, has a higher binding affinity for both BAFF and APRIL, promotes plasma cell development and is preferentially expressed intracellularly, whereas the long form is preferentially membrane bound and expressed by resting cells 43.

BCMA:

BCMA is expressed predominantly by antibody-secreting cells and mediates plasma cell survival, particularly in the bone marrow, making it an excellent therapeutic target for plasma cell depleting strategies. Like TACI, BCMA signals though the classical NFκB pathway. It interacts preferentially to APRIL in mice, but in humans can also bind BAFF. Human BCMA is expressed in the Golgi apparatus and on the cell surface, while soluble BCMA can function as a decoy receptor. In addition, alternate splice variants of unknown function have been identified (reviewed 44).

2.2. Interactions between BAFF/APRIL and TLR signaling pathways – Figure 2

Figure 2: Crosstalk between endosomal TLRs and BAFF/APRIL:

Figure 2:

Excessive load of cell debris, oxidized DNA and/or nucleic acid containing immune complexes can trigger endosomal TLRs. (A) B cells that internalize DNA activate TLR9 and its adapter MyD88 that directly binds to and enhances TACI activity. In a high BAFF environment mature TLR9 stimulated B cells are protected from apoptosis and differentiate to plasma cells. In marginal zone B cells MyD88 enhances plasmablast differentiation through an mTOR pathway. (B) Excess RNA activates TLR7 in pDCs resulting in the release of Type I IFN that induces myeloid DCs to secrete BAFF. RNA recognizing B cells internalize RNA into endosomes through the BCR; activation of TLR7 in these cells induces expression of TACI which in turn amplifies expression of TLR7. In a high BAFF environment autoreactive B cells will therefore preferentially survive and differentiate into plasma cells secreting more autoantibodies.

BAFF is an essential component of the innate immune response. Type I interferons (IFNs) induce BAFF secretion by myeloid DC and neutrophils 45, to complement constitutive expression by radiation resistant stromal cells in lymphoid tissues. Functionally, BAFF upregulates Toll-like receptor (TLR) expression in B cells, and cooperates with dual BCR/TLR signals to drive T independent B cell differentiation. Moreover, B cell activation in response to engagement of endosomal TLRs by nucleic acid-containing autoantigens drives the upregulation of BAFF family receptors, in particular TACI 46, 47. As described below, humoral autoimmunity in BAFF transgenic (BAFF-Tg) mice is T cell independent but requires signals through the TLR adaptor molecule MyD88 and TACI, confirming the relevance of this crosstalk in driving breaks in B cell tolerance 4851. In addition, whereas combined BCR/TLR9 signaling induces B cell proliferation followed by apoptotic cell death, the provision of excess BAFF allows these cells to differentiate into short-lived plasma cells with pathogenic potential 52. Finally, TACI, in particular the short human isoform, can enter endosomal compartments where it associates with the TLR adapter molecule MyD88 through a domain separate from the TLR TIR domain to mediate enhanced NFκB signals that drive plasma cell differentiation. By enhancing the production of the short TACI isoform, TLR9 signals amplify this response 43. Marginal zone B cells express high levels of TACI and in these cells TLR9 and TACI recruit both MyD88 and mTOR to facilitate T-independent class switching and plasma cell differentiation 49. These interactions have important implications for autoimmunity because cross talk between type 1 IFN, TLR, BCR, and BAFF signaling preferentially supports the survival, class-switch recombination, and plasma cell differentiation of B cells recognizing nuclear autoantigens.

3. Regulation of B cell tolerance by BAFF and its receptors – Figure 3

Figure 3: The effect of BAFF inhibition on B cell development:

Figure 3:

Stages of B cell development are shown. The red bars indicate known checkpoints for autoreactivity in the bone marrow (1), at the early transitional stage (2), at GC entry (3) and at post-GC differentiation to memory and plasma cells (4). Green arrows indicate the B cell developmental stages at which BAFF inhibition influences B cell development at the T1 to T2 stage (5), follicular vs marginal zone decision point (6), GC entry (7), GC T dependent affinity maturation (8), plasma cell survival (9) or promotes cell anergy or death (10). X indicates inhibition. B1 B cells, memory B cells and age-associated B cells are BAFF independent.

B cell receptors are generated in the bone marrow by random recombination of immunoglobulin heavy and light chain gene segments to yield a broad repertoire capable of recognizing diverse pathogens. To counter the inevitable generation of self-reactive B cells via random VDJ recombination, overlapping tolerance mechanisms restrain autoreactive B cells, including receptor editing, clonal deletion and the induction of functional unresponsiveness or anergy 53. Importantly, excessive purging of self-reactive clones from the B cell compartment would promote “gaps” in the pre-immune repertoire which could be exploited by pathogens. To counter this problem, coordinated positive and negative selection mechanisms ensure a diverse B cell repertoire balancing breadth of response with risk of autoimmunity. Chief among these positive selection mechanisms, serum BAFF levels regulate B cell development and homeostasis 10. In murine models, either Baff or BAFF-R deletion results loss of peripheral B cells beyond the transitional type 1 stage, indicating a critical role for BAFF/BAFF-R signals in regulating in mature B cell survival and the size of the naïve B cell compartment 54. An additional function of BAFF may be to preferentially enhance the generation of IL10-producing regulatory cells 5557, however this effect appears to be lost in the setting of inflammation 58.

In addition to regulating B cell survival, serum BAFF levels modulate the relative autoreactivity of the naive B cell repertoire. Initial studies indicated that transgenic BAFF overexpression (BAFF-Tg) results in spontaneous humoral autoimmunity, suggesting that excess BAFF broadly disrupts B cell tolerance 3. However, analysis of autoreactive BCR transgenic models indicated that the deletion of high-affinity autoreactive B cells via central tolerance is not impacted by BAFF overexpression, a finding consistent with limited BAFF-R expression on developing BM B cells. Moreover, whereas excess BAFF promotes the survival and follicular entry of high-affinity anergic B cells, the introduction of competing non-autoreactive B cells during development curtails the maturation of these autoreactive B cell clones. Rather, supraphysiologic BAFF exerts a relatively nuanced impact on the stringency of transitional B cell selection, by exerting context specific effects on the survival of lower affinity self-reactive B cells 5961.

As detailed above, BAFF-R signals integrate with tonic BCR signaling pathways to modulate transitional B cell selection in competitive environments 6264. Since autoreactive B cells downregulate surface BCR, they exhibit greater dependence on BAFF survival signals. Thus, in settings of reduced available BAFF, these cells are more likely to be deleted or anergized than their non-autoreactive counterparts. In contrast, increased serum BAFF enhances the survival and maturation of these lower affinity self-reactive clones. As an example of this dichotomy, the Brink group investigated the effects of excess BAFF on autoreactive B cell survival using the Hen Egg Lysozyme (HEL) transgenic system. In this model, whereas BAFF failed to rescue high-affinity anti-HEL B cells from central deletion, it enhanced the survival, maturation and follicular entry of lower affinity autoreactive B cells. However, if intercellular competition for available BAFF was provided by non-autoreactive B cells, these same cells could no longer be rescued by excess BAFF and were anergized or deleted. In the same competitive environment, B cells expressing the anti-HEL heavy chain alone (i.e. exhibiting lower affinity HEL binding) matured to the follicular compartment but were excluded from the marginal zone. When excess BAFF was provided these cells were then able to reconstitute the marginal zone 59, 65. These studies are highly instructive because they provide a framework for interpreting studies of the effect of BAFF excess or deficiency on naive B cell fate in immunoglobulin transgenic mice based on two major variables, namely affinity of the immunoglobulin for autoantigen and the degree of competition for BAFF provided by non-autoreactive B cells.

Importantly, affinity for antigen is not the only predictor of BAFF dependence since other autoreactive transgenes behave differently. The Manser group has studied two heavy chain knock-in mice that produce large numbers of follicular (FO) B cells that are either weakly or strongly autoreactive with nuclear autoantigens. In these models, BAFF excess expanded the MZ pool without allowing either highly- or weakly-autoreactive B cells to outcompete non-autoreactive cells in the mature compartment 66. The reasons for these conflicting data remain incompletely understood, but may relate to the chemical nature, abundance or valence of the relevant self-antigen in these different transgenic models. In sum, these experiments show that BCR signals and available BAFF levels influence autoreactive B cell survival. However, BAFF is not the only molecule needed for the survival or depletion of autoreactive B cells and in a competitive environment not all autoreactive B cells are equally dependent on the availability of BAFF.

A potential consequence of this general model in which BAFF functions as a rheostat for autoreactive B cell survival is that increases in serum BAFF are predicted to increase both the breadth of the pre-immune B cell repertoire and the relative self-reactivity of naïve B cells, resulting in a trade-off between autoimmune risk and enhanced pathogen responses. Consistent with this idea, a genetic study of elevated rates of autoimmunity in the Sardinia population identified a novel insertion–deletion variant in the 3′ UTR of human TNFSF13B (encoding BAFF) 15. Variant TNFSF13B (BAFF-var) exhibited reduced microRNA degradation, resulting in 1.5–2 fold increase in circulating BAFF levels, B cell expansion and an elevated risk of SLE and multiple sclerosis (MS). Although a detailed analysis of the naïve B cell repertoire in Sardinian subjects has not yet been performed, murine modeling predicts that BAFF-var carriers likely express a B cell repertoire enriched for low-affinity autoreactive B cell specificities. Strikingly, this autoimmune risk variant showed evidence of positive selection within the Sardinian population, an observation the authors attributed to the selective pressure of malaria, a pathogen endemic in the region until the 1950s 67. In keeping with this hypothesis, BAFF-Tg mice are protected from lethal malaria infection 68 and human B cells expressing the intrinsically autoreactive VH4–34 heavy chain family are expanded in the memory compartment of malaria patients 69, suggesting a link between relaxed B cell developmental selection and robust pathogen responses.

3.1. BAFF-R and TACI signals integrate to promote autoimmunity in BAFF transgenic mice

Given human data linking increased serum BAFF with the pathogenesis of SLE, the immune mechanisms whereby BAFF modulates B cell tolerance has been of considerable interest. Despite central and peripheral tolerance mechanisms, a substantial proportion of naïve B cells express autoreactive BCRs even at physiologic BAFF levels 70. Thus, BAFF-driven expansion of low-affinity autoreactive B cells is unlikely the sole contributor to humoral autoimmunity observed in high BAFF settings. Rather, signals downstream of BAFF-family receptors integrate with antigen receptor and innate signals to drive breaks in B cell tolerance at later stages of B cell development.

Murine models of transgenic BAFF overexpression (BAFF-Tg) exhibit spontaneous humoral autoimmunity characterized by class-switched autoantibodies and plasmablast expansion in the absence of CD4+ T cell help 71. As detailed above, BAFF-R signals drive B cell expansion and the survival of low-affinity autoreactive B cells, suggesting that BAFF-R is the predominant receptor responsible for B cell tolerance breaks. In contrast, separate groups, including our own, used independently-generated BAFF-Tg and Taci−/− murine strains to uncover a critical role for TACI signals in BAFF-Tg autoimmunity 50, 51, 72, 73. These data were particular surprising given that TACI was initially proposed as a negative regulator of BAFF signaling 38, 74, and highlight the complexity of BAFF family biology.

Based on these findings, we sought to identify the B cell subset that is the target of TACI-dependent activation in BAFF-Tg mice. Since the splenic marginal zone (MZ) is both enriched for autoreactive specificities and the number of MZ B cells is increased in BAFF-Tg mice 3, MZ B cells were initially predicted to promote BAFF-driven SLE 10. Consistent with this idea, MZ-like cells have been found in inflamed salivary glands of BAFF-Tg mice 75. However, BAFF-Tg mice deficient in lymphotoxin-β (resulting in disordered splenic architecture and loss of MZ B cells) as well as genetically or surgically splenectomized BAFF-Tg mice (resulting in loss of both MZ B cells and peritoneal B1a B cells) develop immune-complex glomerulonephritis 76, 77, findings suggesting that neither MZ B cells nor B1a B cells are required for BAFF-mediated lupus nephritis.

To address these controversies, we examined the effect of increased BAFF on surface TACI expression by B cell developmental subsets. Surprisingly, whereas TACI is typically limited to mature B cells at physiological BAFF levels, we observed a striking expansion of TACI-expressing transitional B cells in BAFF-Tg mice. We confirmed that this TACI+ transitional subset derived from bone fide transitional cells, using a fate-mapping approach. In settings of BAFF excess, TACI expression marked a subset of transitional B cells exhibiting cellular proliferation, activation marker expression, and a BCR repertoire enriched for autoreactivity. In keeping with a direct contribution of this subset to BAFF-driven autoimmunity, TACI+ transitional B cells expressed activation-induced cytidine deaminase (AID) and Blimp1 required for somatic hypermutation, class-switch recombination, and plasma cell differentiation, respectively 73. Finally, sorted TACI+ transitional B cells spontaneously produced class-switched autoantibodies ex vivo, whereas no antibody was generated by BAFF-Tg follicular mature (FM) and MZ B cells. Importantly, whereas our data strongly suggest that transitional B cells are a prominent source of pathogenic autoantibodies in BAFF-Tg mice, these findings do not preclude additional contributions of alternate B cell subsets to BAFF-driven autoimmunity. Indeed, a recent study identified a role for peritoneal B1b B cells in the autoantibody repertoire of BAFF-Tg mice 78, data in keeping with prominent TACI expression by peritoneal B1 B cells.

We next examined the additional B cell signals driving transitional B cell TACI expression and BAFF-driven breaks in tolerance. Integration of BCR and TLR signals enhances B cell activation, and autoimmunity in BAFF-Tg mice is abrogated in the absence of the TLR signaling adaptor, MyD88 71. In addition, as described above, prior studies have shown that BCR and TLR activation ex vivo increases sTACI levels on mature B cells 46, and that TACI also associates intracellularly with MyD88 in endosomes. In keeping with these data, deletion of the BCR signaling adaptor Bruton’s tyrosine kinase (BTK) or MyD88 similarly protected BAFF-Tg mice from progressive glomerulonephritis. However, these signaling pathways exerted distinct impacts on surface TACI levels and autoantibody production. Whereas loss of BCR signals abrogated transitional TACI expression and blocked autoantibody production across immunoglobulin isotypes, surface TACI was maintained in Myd88−/−.BAFF-Tg mice which manifested a specific reduction in class-switched, but not IgM, autoantibodies 72.

Of the Myd88-dependent TLRs, the endosomal receptors TLR7 and TLR9 have been implicated in the production of RNA- or DNA-associated autoantibody specificities, respectively. Although original studies reported high-titer anti-double stranded DNA (dsDNA) autoantibodies in BAFF over-expression models 3, 71, we noted a marked enrichment for RNA-associated autoantibodies in BAFF-Tg mice, with relatively low-titer anti-dsDNA autoantibodies observed using the stringent Crithidia luciliae assay for native DNA reactivity. Consistent with these findings, TLR7 deletion abrogated class-switched autoantibodies in BAFF-Tg mice. Moreover, TACI deletion resulted in a relatively specific reduction in BAFF-driven RNA-associated autoantibodies, including against RNP and Sm antigens. Strikingly, loss of these specificities correlated with a specific decrease in endocapillary, but not mesangial, immune complex deposits within inflamed glomeruli of aged BAFF-Tg mice 51.

Importantly, whereas TACI expression is critical for BAFF-driven autoimmunity, BAFF-R signals exerted complementary roles in facilitating breaks in B cells tolerance. Specifically, the expansion of cycling TACI+ transitional B cells and production of class-switched autoantibodies was absent in BAFF-R−/−.BAFF-Tg mice 72. Although the mechanisms whereby BAFF-R and TACI signals integrate to break B cell tolerance have not been completely defined, we hypothesize that BAFF-R signals both facilitate the survival of low-affinity autoreactive transitional B cells and promote the expansion of autoreactive clones that have engaged self-antigen. Notably, this model requires that BAFF-R signals impact B cell biology earlier in development as described above. Alternatively, BAFF-R and TACI ligation could enhance B cell TLR mediated signals 49 and thereby facilitate class-switched autoantibody production.

In summary, our data demonstrate that multiple signaling pathways, including BCR, TLR, TACI and BAFF-R signals, integrate to promote class-switched autoantibodies in high BAFF settings. It remains to be determined whether modest BAFF excess such as that observed in Sardinian individuals or in the setting of SLE are sufficient to break tolerance in the transitional compartment and drive T cell independent autoimmunity.

3.2. Distinct contribution of EF and GC pathways to lupus pathogenesis

During a protective or pathogenic humoral response, antibody-secreting plasma cells can be generated by either extrafollicular (EF) or germinal center (GC)-dependent B cell activation pathways. After infectious challenge, EF B cell activation produces an initial wave of relatively low-affinity protective antibody, which is followed by high-affinity, durable antibodies derived from GCs. Functionally, EF-derived plasma cells are believed to represent a short-lived population residing in the splenic red pulp or lymph node medullary cords, while GC-derived plasma cells are maintained long-term in bone marrow survival niches 79. Importantly, both EF and GC pathways have been implicated as sources for pathogenic autoantibodies in humoral autoimmune diseases, in particular SLE. Specifically, EF B cell activation underlies the generation of somatically mutated plasma cells in the MRL.Faslpr80 and BAFF-Tg lupus-prone stains 71. In addition, immunophenotyping and VH-family clonal analysis has uncovered an in vivo developmental link between expanded active naïve B cells (aNAV; defined as CD19+IgD+CD27CXCR5), double-negative 2 (DN2; CD19+IgDCD27CXCR5) B cells and circulating plasma cells in lupus patients 81, 82; findings implicating EF B cell activation as important contributor to pathogenic autoantibodies in human SLE. Mechanisms for regulation of autoimmunity generated via this EF pathway remain poorly understood.

In addition to EF B cell activation, dysregulated GCs represent an alternate source for class-switched autoantibodies in SLE. Murine studies have shown that the entry of non-anergic autoreactive B cells into the GC is usually tightly regulated, perhaps by the availability of T cell help 83. In addition, those B cells that acquire autoreactivity as a result of somatic mutation are censored before they reach the memory compartment 8487. Despite these data, evidence from both animal and human studies implicates dysregulated GCs as an important site for B cell tolerance breaks. For example, the development of spontaneous GCs is frequently observed in murine lupus models and human subjects can develop ectopic GCs within inflamed target organs 8890. Moreover, a subset of autoantibody specificities can persist despite anti-CD20 B cell depletion, implicating GC-derived CD20neg long-lived plasma cells in their genesis. Finally, SLE is characterized by a prolonged period of pre-clinical autoimmunity during which patients sequentially accumulate autoantibody specificities in a manner consistent with epitope spreading within an ongoing dysregulated GC response 91.

Based on these observations, the contribution of BAFF signals to autoreactive GC formation is particularly relevant. Notably, the GC is a relatively BAFF-poor environment. While BAFF-R occupation by BAFF occurs in the GC light zone, B cells in the dark zone downregulate BAFF-R, most likely as a result of receptor cleavage 25. Despite these data, Baff−/− mice exhibit a defect in the formation of a mature follicular dendritic cell (FDC) network and are characterized by premature GC termination 92, 93. In addition, evidence suggests that T follicular helper (Tfh) cells regulate light zone affinity selection of GC B cells by producing BAFF. Although T cell-derived BAFF was not required for GC formation, loss of T cell-intrinsic BAFF production reduced the emergence of somatically mutated high-affinity B cell clones following candidate antigen immunization 94. Notably, TACI is profoundly downregulated on GC B cells in an IL-21 dependent manner. By limiting the sequestration of free BAFF, decreased TACI expression may thereby allow reduced concentrations of locally-generated BAFF to modulate GC selection via interaction with BAFF-R 94. These data are compatible with the general model wherein higher affinity B cells present more antigen to Tfh cells and thereby receive enhanced survival signals, including BAFF. These findings suggest that increased serum BAFF and therapeutic BAFF inhibition could reciprocally modulate GC selection via either enhancing selection or deletion of autoreactive clones.

Importantly, recent studies suggest that, as in mice, the drive towards either the GC or EF pathway in SLE may be patient dependent, resulting in differences in the accumulation of autoreactive class switched memory cells vs. plasma cells in individual lupus patients 95. Cumulatively, these murine and human data indicate that significant complexity underlies lupus pathogenesis with both EF and GC-dependent B cell activation pathways contributing to pathogenic autoantibody production in SLE. More importantly, this complexity may explain contradictory data from human clinical trials. Since the various BAFF-family cytokines and receptors have distinct roles in different B cell activation pathways, therapeutic targeting of a specific pathway may result in modest or inconsistent benefit based on the patient population studied.

3.3. Roles for individual BAFF family cytokines and receptors in lupus pathogenesis

The study of murine SLE strains rendered genetically deficient in specific BAFF family cytokines and receptors has informed our understanding of lupus pathogenesis. However, the at times contradictory nature of these data based on the specific lupus-prone strain studied has complicated the interpretation of human clinical trials of BAFF blockade in SLE. As detailed above, the potential generation of lupus-associated autoantibodies via distinct B cell activation pathways may account in part for these conflicting results. Viewed through this lens, individual BAFF cytokines and/or receptors could facilitate disease pathogenesis in individual patients and may explain the heterogeneity in clinical responses to BAFF inhibition.

The interpretation of murine genetic models is further complicated by fluctuations in serum BAFF and APRIL levels conferred by changes in the availability of surface receptors and the ability of shed forms of TACI and BCMA to act as decoy receptors. Thus, deficiency of either BAFF-R or TACI increases the circulating levels of their respective ligands and genetic deletion of either TACI or BCMA results in the loss of the regulatory effect of the soluble decoy receptors. Despite these caveats, we will briefly summarize the available data regarding the impact of genetic targeting of both BAFF-family cytokines and the three known receptors in murine SLE, with the goal of informing the interpretation of human clinical trials of BAFF blockade (Table 1).

Table 1:

The effect of deficiency of BAFF family members in murine lupus models

Deficiency Mouse strain Result Reference
BAFF-R MRL.Faslpr1 Profound B cell depletion
No reduction in autoreactive plasma cells or effect on disease progression
102
NZM2328 2 101
TACI MRL.Faslpr Decreases autoantibodies
Improves outcome
51
Nba2.Yaa 3 99
BAFF Tg 50, 51, 73
NZM2328 No effect on autoantibodies, plasma cells or mortality 101
BCMA Nba2.Yaa Increases plasma cells, enhances autoantibodies and exacerbates disease 103
NZM2328 Increased spleen size
No effect on autoantibodies, plasma cells or mortality
101
APRIL NZM2328 No effect on autoantibodies, plasma cells or mortality 100
Nba2.Yaa Decreased autoantibodies and nephritis 99
BAFF NZM2328 Delay in autoantibodies and failure to progress from glomerulonephritis to ESRD 97
BAFF/APRIL NZM2328 Same as BAFF deficiency 98
BCMA/TACI Nba2.Yaa Same as TACI deficiency 99
BAFF-R/TACI NZM2328 Protective 98
BAFF-R/BCMA NZM2328 Protective 98
TACI/BCMA NZM2328 Increases plasma cells, enhances autoantibodies and exacerbates disease 98
1

Loss of Fas, expansion of double negative T cells, EF B cell expansion and short-lived plasma cells

2

Hyperactivity of B cells, enhanced GC formation and long-lived plasma cells

3

Nba2 locus confers B cell hyperreactivity. Yaa translocation confers an extra copy of TLR7

BAFF:

BAFF deletion significantly attenuates autoimmunity in NZM2328 lupus-prone mice, as manifested by reduced glomerular pathology and delayed proteinuria even if the mice are challenged with Type I IFN 96. Despite a profound reduction in circulating mature B cells, aged Baff−/−.NZM2328 mice still developed class-switched IgG+ anti-nuclear antibodies. However, glomerular immune deposits in Baff−/−.NZM2328 mice were predominantly IgG1 subclass, rather than the complement-fixing IgG2a in Baff-sufficient NZM2328 animals. Consistent with these findings, BAFF deletion prevented glomerular C3 complement deposition and limited progressive nephritis in aged Baff−/−.NZM2328 mice 97. APRIL may partially compensate for lack of BAFF in this model, a conclusion supported by loss of serum autoantibodies in dual-deficient Baff−/−.April−/−.NZM2328 mice 98. The cellular mechanisms underlying BAFF-independent autoantibody generation have not yet been defined, however GCs are reduced, but not absent, in Baff-deficient NZM2328 mice. In addition, peritoneal B1 and splenic transitional B cells are relatively preserved, implicating these B cell subpopulations as potential autoantibody sources.

APRIL:

The impact of APRIL deletion is strain dependent, with attenuation of autoantibody production and lupus nephritis in Nba2.Yaa mice 99, but no impact on disease development in NZM2328 mice 100. The immune mechanisms accounting for this discrepancy have not been addressed but may relate the differential requirements for TACI in these respective models (see below).

BAFF-R:

Given the critical role for BAFF in B cell homeostasis including the requirement for BAFF-R signals in B cell maturation beyond the transitional stage, BAFF-R deletion was predicted to inhibit lupus development in murine models. Indeed, as described above, BAFF-R deficiency ameliorated autoantibody production in BAFF-Tg mice. However, lack of BAFF-R signals did not prevent humoral autoimmunity in either MRL.Faslpr or NZM2328 mice despite profound B cell depletion 101, 102. These murine lupus models differ in several important immunologic characteristics. In the MRL.Faslpr strain, FAS deficiency promotes predominantly EF B cell activation resulting in the generation of short-lived TACI+ plasma cells. In contrast, NZM2328 mice develop spontaneous GCs generating long-lived plasma cells. These data suggest that increased stringency of naïve B cell selection in BAFF-R−/− mice is not sufficient to prevent breaks in B cell tolerance in either the EF or GC pathway. One caveat is that increased available BAFF in the setting of BAFF-R deletion may induce supraphysiologic TACI signals in these models. In keeping with this hypothesis, dual BAFF-R and TACI deletion prevented disease in NZM2328 mice 98.

TACI:

In addition to critical roles for TACI in BAFF-Tg autoimmunity 50, 51, TACI deletion limited disease progression in both the MRL.Faslpr and Nba2.Yaa 99 lupus models. Notably, in each of these strains, autoantibody secreting plasma cells are believed to be generated predominantly by an EF activation pathway 95. In contrast, TACI deletion exerted no impact on serum autoantibodies, plasma cell numbers and lifespan in NZM2328 lupus-prone mice, a model characterized by spontaneous GC formation and long-lived plasma cell expansion 101. Importantly, these data are consistent with the known requirement for TACI signals in T cell-independent, but not T-cell dependent, antibody responses. Most importantly, these conflicting animal models suggest that the therapeutic benefit of TACI blockade may be limited to a subset of human lupus patients in whom disease is driven by a TACI-dependent B cell activation program.

BCMA:

BCMA, like TACI, contributes to plasma cell maintenance, but surprisingly deficiency of BCMA increases the number of plasma cells, enhances autoantibody production and exacerbates disease in several lupus models 44, 103. Several mechanisms have been proposed to account for these unexpected data. First, BCMA has been proposed to act in a T cell-intrinsic manner to restrain autoreactive T follicular helper cell (TFH) expansion, resulting in expanded GCs and increased autoantibodies in BCMA-null lupus models 44. Second, BCMA deletion may impact lupus pathogenesis via loss of a soluble decoy protein. Consistent with this hypothesis, dual TACI and BCMA deficiency in NZM2328 mice results in prominent B cell expansion, increased plasma cells, high-titer autoantibodies and exacerbated disease 98, an outcome attributed to increased serum BAFF in the setting of combined decoy receptor deletion. These data are also consistent with a model in which plasma cell survival in inflammatory settings can be independent of both BCMA and TACI. The protective effect of double BAFF-R/BCMA deficiency in NZM2328 mice 98, leaving TACI as the only functional receptor, is difficult to explain given the limited impact of BAFF-R deletion alone and the exacerbating effect of BCMA deficiency.

4. Pharmacologic targeting of BAFF family cytokines and receptors

It is clear from the above studies that the BAFF cytokine family plays an important role in the pathogenesis of lupus, but that heterogeneity in responses to BAFF inhibition should be expected based on the variety of B cell developmental pathways that contribute to lupus pathogenesis. In addition, an important distinction between the genetic models described above and treatment of human SLE is that targeted therapies are provided to patients with established disease. Thus, in addition to roles in driving breaks in tolerance, an important therapeutic consideration is the requirement for BAFF-family cytokines in the long-term maintenance of autoreactive memory B cells and plasma cells.

Therapeutic approaches to BAFF family blockade include the use of antibodies against specific cytokines or receptors, or the construction of receptor fusion proteins that target BAFF, or both BAFF and APRIL. In this regard, BAFF-R-Ig blocks BAFF binding to BAFF-R and TACI, whereas TACI-Ig blocks the interaction of BAFF and APRIL with all three receptors. Clinically, there has been much debate about the utility of targeting BAFF vs. targeting both BAFF and APRIL, targeting APRIL alone or using newer approaches that spare the regulatory decoy proteins of TACI and BCMA (Figure 1).

Among the agents currently in development for the treatment of human SLE, anti-BAFF and variants of BAFF-R that act as pure BAFF antagonists all have a similar mechanism of action in preventing BAFF interactions with its receptors. Of these, the human anti-BAFF antibody belimumab has been the most effective in human clinical trials and is the only agent with FDA approval for the treatment of SLE 8, 9, 104. In addition, dual targeting of BAFF and APRIL using TACI-Ig (atacicept) completed phase 2 studies 105. Recent studies have shown that belimumab binds to and inhibits the activity of BAFF trimers but not multimers whereas atacicept can bind to both trimers and multimers 12. Atacicept also antagonizes the activity of APRIL and of BAFF-APRIL heterotrimers. Both belimumab and atacicept bind to and antagonize the activity of membrane bound BAFF 106 but do not mediate complement or antibody dependent cytotoxicity in vitro.

5. Insights into the impact of BAFF family inhibition from murine models

To inform the design and interpretation of human clinical trials, our group has examined the role of BAFF and its receptors in B cell selection and disease progression in three different mouse models of SLE. Importantly, the various therapeutic approaches resulted in distinct impacts on each model tested, highlighting the complexity of lupus pathogenesis and predicting similar heterogeneity of clinical response in human studies. To briefly summarize the murine models evaluated, NZB/W F1 mice are a classic murine lupus model in which T cell-driven immune responses result in the GC-driven production of somatically mutated high affinity IgG anti-DNA antibodies that deposit in the kidneys and initiate a proliferative glomerulonephritis with abundant interstitial inflammatory infiltrates reminiscent of human Class IV lupus nephritis. Similarly, NZM2410 mice are an inbred strain derived from NZB and NZW lineage, which also generate T cell-dependent anti-DNA autoantibodies, although renal disease in the model is primarily glomerulosclerosis with limited interstitial inflammation. Finally, NZW/BXSB male mice express an additional copy of TLR7 via the Yaa translocation, produce both GC and EF derived autoantibodies and develop severe glomerulonephritis and interstitial renal disease. Autoantibodies in these mice are predominantly directed towards RNA-associated and phospholipid autoantigens 107112.

5.1. Is there a difference between targeting BAFF or APRIL alone compared with dual targeting?

We first asked whether targeting BAFF or both BAFF and APRIL differentially impacted autoimmune responses and longevity in NZB/W F1 mice. Whereas delivery of BAFF-R-Ig or TACI-Ig to pre-nephritic mice using an adenoviral vector delayed disease onset by ~6 months, neither agent was sufficient to reverse established disease. However, the addition of a brief two-week course of costimulatory antagonist CTLA4-Ig synergized with both BAFF-R-Ig and TACI-Ig to limit renal damage, proteinuria, and enhance survival 111, 113. Notably, a related study confirmed TACI-Ig-mediated protection from progressive nephritis, but showed no effect on disease with BAFF-R-Ig treatment 114, findings that contrast with the robust impacts of even short-term BAFF-R-Ig in our hands. Potential explanations for this discrepancy include differences in dose or protein structure of the BAFF-R-Ig used in each study. Mechanistically, both BAFF-R-Ig and TACI-Ig resulted in a marked reduction in T2, marginal zone and follicular B cells, resulting in a secondary decrease in the accumulation of splenic class switched B cells, memory T cells and dendritic cells. In contrast, the number of splenic B1 B cells was unchanged. Moreover, despite prolonged B cell depletion, spontaneous GCs still formed, resulting in the generation of high-affinity somatically-mutated anti-DNA autoantibodies shortly after treatment discontinuation.

Plasma cell survival in mice depends on signals via BCMA or TACI that can be delivered by either BAFF or APRIL ligation. Notably, in the NZB/W model, IgM and IgG plasma cells exhibited distinct requirements for BAFF family survival signals. Short course TACI-Ig treatment markedly reduced serum IgM levels for several months, whereas BAFF-R-Ig exerted minimal impacts on IgM titers 113. These findings suggested that BAFF and APRIL together are required for the terminal differentiation and maintenance of IgM plasma cells. In contrast, IgG plasma cells are more resistant to the effects of TACI-Ig. Treatment with BAFF-R-Ig minimally affected serum IgG. In keeping with compensatory roles for BAFF and APRIL in plasma cell survival, a specific antibody to APRIL only modestly decreased autoantibody titers in aged NZB/W mice without affecting overall survival 16, 115. Consistent with these results, long-term TACI-Ig treatment reduced splenic and bone marrow plasma cells in NZB/W mice, resulting in reduced anti-dsDNA titers and protection from proteinuria 114.

5.2. How does strain influence the response to BAFF/APRIL inhibition?

We next determined whether other lupus strains would be similarly responsive to administration of adenoviral vectors expressing BAFF-R-Ig or TACI-Ig. NZM2410 mice are characterized by the expansion of splenic plasma cells secreting IgG1 autoantibodies. This strain was highly responsive to short-term BAFF-R-Ig and TACI-Ig, even in animals treated after the onset of proteinuria. Similar to NZB/W mice, short-term exposure to TACI-Ig profoundly depleted serum IgM but induced only a temporary decrease in total IgG1 which was followed by the rapid return of class-switched autoantibodies 110.

NZM2410 mice manifest rapid progression of glomerular damage with predominant glomerulosclerosis and minimal interstitial inflammation. Despite glomerular IgG deposition in both BAFF-R-Ig and TACI-Ig treated animals, BAFF family blockade significantly attenuated renal damage and protected against proteinuria. Surprisingly, BAFF-R-Ig treated animals developed significant interstitial infiltrates, but lacked the tubular and endothelial damage associated with ESRD in this model suggesting that an additional checkpoint in the progression of renal damage is regulated by BAFF, perhaps via direct modulation of intra-renal myeloid cells 110, 116.

In the MRL.Faslpr model, class-switched autoantibodies are produced by predominantly short-lived plasma cells generated via a T cell-dependent EF pathway. As predicted, TACI-Ig treatment resulted in a rapid decrease in both IgM and IgG autoantibodies and prolonged survival, without impacting T cell activation or renal interstitial infiltrates. Therefore, in MRL.Faslpr mice, plasma cell expansion requires continued BAFF and APRIL signals to maintain IgM and IgG plasma cells 117.

Finally, we studied the impact of BAFF inhibition in NZW/BXSB mice in which the Yaa translocation promotes TLR7-dependent anti-RNA and anti-cardiolipin autoantibody generation. Notably, both BAFF-R-Ig and TACI-Ig treatment limited proteinuria and attenuated the cardiac and renal damage characteristic of this model. However, protection from these disease manifestations occurred in the absence of any effect of serum autoantibody titers, glomerular IgG deposition or immune thrombocytopenia. Continued antibody-mediated platelet destruction despite BAFF/APRIL inhibition indicates no treatment-induced loss of autoantibody pathogenicity. Rather, BAFF inhibition likely attenuated the inflammatory response to autoantibody deposition in target organs 112.

5.3. What is the effect of Type I IFN on response to BAFF/APRIL inhibition?

SLE is characterized by a Type I interferon (IFN) signature. Since type 1 IFN is known to induce feed-forward BAFF production by myeloid dendritic cells, we asked whether BAFF inhibition could protect NZB/W mice from type 1 IFN-accelerated autoimmunity. Continuous TACI-Ig initiated at the time of IFN treatment delayed disease onset and prolonged lifespan of IFN-accelerated NZB/W mice but lacked efficacy if TACI-Ig was delayed until after autoantibody formation. In this IFN-accelerated model, TACI-Ig still induced profound IgM depletion but exerted no impact on IgG autoantibodies, findings consistent with abundant GCs and IgG+ plasma cells in treated animals 118. Together, these data show that Type 1 IFN modulates the splenic microenvironment to support IgG plasma cell survival in the absence of BAFF/APRIL signals, perhaps by promoting B cell TLR7 expression or increasing expression of cytokines known to support B cell differentiation and survival. Moreover, these findings indicated that differences in the ability of BAFF/APRIL to support IgM vs. IgG plasma cells is not due either to differences in the microenvironment or intrinsic differences between short-lived and long-lived plasma cells, since IgG plasma cells in the IFN-induced model persist mainly in the spleen as a short-lived population. Despite conserved glomerular IgG deposition, renal inflammation was attenuated in the TACI-Ig treated mice. These findings correlated with a reduction in inflammatory mediators produced by renal myeloid cells, including TNF, CXCL13 and CCL5 119. Thus, TACI-Ig either decreased the relative pathogenicity of deposited autoantibodies, or BAFF/APRIL inhibition exerted B cell-extrinsic effects on renal inflammation.

5.4. Does BAFF inhibition alter B cell selection?

An important question is whether BAFF inhibition restores tolerance in the naïve, EF, or GC B cell repertoire in SLE. To address this question, we utilized NZB/W mice bearing a germline heavy chain (D42) that was derived from an anti-dsDNA hybridoma. Despite the potential for diverse light chain pairing with D42, the majority of IgG anti-DNA autoantibodies in 6-month-old NZB/W mice derive from D42 paired with the VκRF light chain, the original hybridoma heavy/light chain combination which confers high affinity dsDNA binding without additional somatic hypermutation.

To study autoreactive B cell selection and survival in competition with wild-type cells, we established mixed BM chimeras which were treated with TACI-Ig from 3 days post-transplant to facilitate B cell reconstitution in a BAFF/APRIL poor environment. Consistent with the models described above, high-affinity D42/VκRF B cells were deleted in the BM at the pre-B / immature B cell stage. In contrast, lower affinity D42 B cells were deleted between the T1 and T2 stages, a process which was enhanced by TACI-Ig treatment. Using single cell PCR analysis, we observed no preferential effect of TACI-Ig on the light chain repertoire of D42 cells. Thus, our data show that competition with non-autoreactive B cells is sufficient for deletion of high affinity D42/VκRF B cells, while TACI-Ig broadly impairs the survival of lower affinity D24 B cells. However, TACI-Ig did not prevent GC formation or selection of D42/VκRF B cells into the GC and plasma cell compartments, resulting in equivalent anti-DNA IgG autoantibodies in treated and untreated chimeras despite profound B cell depletion 120.

Since D42/VκRF DNA reactivity does not require somatic mutation, we performed similar experiments using 3H9 heavy chain knock-in mice. In this model, the 3H9 heavy chain can pair with multiple light chains to encode autoreactive and non-autoreactive specificities, and somatic mutation improves the affinity of self-reactive 3H9 autoantibodies. Deletion of 3H9 B cells occurs predominantly at the pre-B cell stage in this strain, with 3H9 cells surviving BM deletion exhibiting reduced surface IgM expression. Using BAFF-R-Ig to inhibit BAFF in this study, we observed prolonged proteinuria-free survival in both NZB/W and NZW/BXSB 3H9 chimeras. However, BAFF-R-Ig had no effect on 3H9 B cell deletion, IgM downregulation or selection of the naïve B cell repertoire in either strain 108.

Despite central and peripheral tolerance mechanisms, 3H9 B cells can be positively selected into the GC compartment. DNA and cardiolipin specificity of 3H9-derived autoantibodies is facilitated by pairing with Vκ5–43 and Vκ5–48 light chains that confer either low or moderate autoreactivity, respectively. For this reason, we asked whether BAFF-R-Ig alters the selection and mutational frequency of these heavy/light chain combinations. In the NZB/W model, the relative representation of 3H9/Vκ5–43 and 3H9/Vκ5–48 GC B cells was not impacted by BAFF-R-Ig treatment, but the lower affinity Vκ5–43 encoded light chains expressed fewer mutations and were underrepresented in the plasma cell compartment following BAFF inhibition. In contrast, BAFF-R-Ig treated NZW/BXSB mice exhibited a ~4 fold reduction in the higher affinity 3H9.Vκ5–48 clones in the GC and plasma cell compartment, compared with Vκ5–43-expressing B cells, with no difference in somatic mutation rates. Altered GC selection correlated with modest reductions in anti-cardiolipin titers in NZW/BXSB 3H9 chimeras, events perhaps explained by BAFF-dependent modulation of TLR7 signals in this Yaa-expressing strain 108.

Together, these data confirm the variable impacts of BAFF inhibition on the selection of naïve and activated B cells in lupus models. High affinity autoreactive B cells are deleted in the BM without further regulation by BAFF inhibition. Although cells escaping BM deletion may be subject to regulation by BAFF at the transitional stage, this process does not apply equally to all autoreactive specificities. Moreover, in spite of tolerance mechanisms, a subset of high affinity self-reactive B cells within the naïve compartment can enter autoimmune GCs, undergo somatic hypermutation and differentiate into pathogenic plasma cells producing class-switched autoantibodies. BAFF inhibition has modest, strain-specific impacts on GC selection at this stage. Whereas only lower affinity GC B cells are regulated by BAFF in NZB/W mice, the selection of pathogenic anti-cardiolipin expressing B cells is modestly impacted by BAFF inhibition in NZW/BXSB mice. Thus, these data indicate that reduced serum BAFF can exert a context-dependent impact on B cell selection, somatic mutation and plasma cell differentiation within autoimmune GCs.

5.5. Summary of therapeutic studies in murine lupus models

Together, these studies provide several important insights into BAFF/APRIL inhibition in SLE that are relevant to the treatment of human patients. First, there is considerable heterogeneity between murine strains with respect to responsiveness to treatment with either selective BAFF or dual BAFF/APRIL blockade. These distinctions are likely applicable to the diverse immunologic underpinnings of human lupus and may account for variable treatment responses in lupus clinical trials. Second, BAFF/APRIL inhibition is more effective when initiated early in the disease course, with reduced effectiveness after immune complex deposition and the onset tissue damage. Third, increased Type I IFN may confer resistance to BAFF/APRIL inhibition. Fourth, there are differences between blocking BAFF alone vs. combined BAFF/APRIL inhibition. Most prominent amongst these is the sustained decrease in serum IgM induced by TACI-Ig, which may increase infectious complications following TACI-Ig treatment. In contrast, TACI-Ig treatment exhibited greater efficacy than BAFF-R-Ig in certain models, including in MRL.Faslpr mice. Fifth, there is a modest and strain specific effect of BAFF inhibition on GC B cell selection that may modulate the pathogenicity of the emerging autoantibodies. Sixth, plasma cells required both BAFF and APRIL for long-term survival but the effect of BAFF/APRIL inhibition on IgG plasma cells is variable between models. Finally, BAFF inhibition appears to confer a component of protection from renal damage that is independent of direct impacts on B cells.

6. Therapeutic targeting of BAFF in humans

Given the variable effects of BAFF inhibition in mouse models it is likely that responses in humans will be heterogeneous and can only be evaluated in a clinical trial setting. It is therefore useful to compare the known effects of belimumab and atacicept in humans. Readers are referred to a recent comprehensive review that details the design and clinical outcomes of the human trials 104.

6.1. Selective BAFF inhibition

Because of the mechanistic similarity of selective BAFF blocking agents, we will only consider the anti-BAFF monoclonal antibody belimumab here. Belimumab has consistently shown a benefit when added to standard of care therapy in moderately active SLE, particularly in those patients with higher disease activity, increased anti-dsDNA antibodies and low serum complement 121. Similar to mouse studies using BAFF-R-Ig, belimumab induces a profound reduction in naïve B cells and transitional B cells past the T1 stage, with lesser impacts on class switched memory cells, B1 cells and plasma cells 122, 123.

Belimumab has an excellent long-term safety profile, and effects on serum total immunoglobulin titers are minimal. Although naïve B cells are depleted by ~95%, BAFF inhibition is associated with a temporary increase in memory B cells, perhaps because of expansion or peripheral mobilization following naïve B cell depletion. Nevertheless, memory B cells decreased substantially with prolonged belimumab treatment (by ~70% reduction after 7 years), likely because of reduced replacement from the naïve compartment. Similarly, circulating plasma cells are relatively resistant to short-term BAFF inhibition, but are reduced by ~70% in chronically-treated patients 122, 123. This long-term reduction in memory B cells and plasma cells is associated with a 40–60% decrease in autoantibody titers, without increased infectious complications

Despite similar B cell depletion, not all treated subjects respond to belimumab. To identify predictors of belimumab response, PBMC gene expression was assessed in patients treated with the BAFF inhibitor tabalumab. Whereas ~75% of patients exhibited a Type 1 IFN signature at baseline, this did not change either with disease activity or in response to one year of treatment with tabalumab, and thus lacked efficacy as a clinical biomarker. In addition, serum BAFF levels could not be used to predict treatment response 124.

While all three major belimumab clinical trials in general SLE met their primary endpoints, it is unclear whether this therapy will confer a similar benefit in lupus nephritis. Clinical trials in lupus nephritis are complex given the requirement for new therapies to be tested without withdrawal of standard immunosuppression, such as mycophenolic acid or cyclophosphamide. Therapeutic B cell depletion is accompanied by a homeostatic increase in circulating BAFF, which could result in two potential consequences. First, increased BAFF may alter the stringency of negative selection and allow the survival of low affinity autoreactive B cells, as described earlier. Second, BAFF may facilitate a niche for long-lived plasma cells in secondary lymphoid organs and thereby contribute to disease relapse 125. For these reasons, the combination of B cell depletion with BAFF antagonists has been proposed as a useful strategy for severe lupus manifestations including lupus nephritis 126. Unfortunately, initial data from the CALIBRATE trial, in which belimumab was added to a Rituximab, cyclophosphamide and prednisone induction regimen, did not show any clinical benefit at 48 weeks 127. Nevertheless, several longer term studies have shown reduced SLE flare rates and slower progression of tissue damage in lupus patients taking belimumab 7. Thus, continued clinical experience with belimumab and other BAFF family inhibitors will be needed to define the therapeutic role of these agents in lupus nephritis.

6.2. Dual BAFF/APRIL inhibition

As described above, there are several reasons to predict that dual BAFF and APRIL inhibition may confer greater benefits to targeting BAFF alone. These include blocking BAFF/APRIL interactions with TACI, greater inhibition of signaling by BAFF multimers, and a more profound impact on plasma cell survival and other APRIL contributions to lupus pathogenesis. However, dual BAFF/APRIL inhibition may result in increased infectious complications because of reduced IgM responses and impacts in TACI-dependent innate signaling pathway mediated via endosomal TLRs. Clinical studies using the human TACI-Ig fusion protein atacicept are ongoing, but an initial study using two atacicept doses in SLE failed to meet the primary endpoint of reduction in SRI-4. However, several secondary endpoints were achieved, including an improvement in serological markers and reduced steroid requirement. As expected from murine models, serum IgM decreased by 70% whereas total IgG and autoantibody titers were only reduced by 30% 105. These changes in immunoglobulin titers were not associated with increased infectious complications. Loss of protective immunity was evaluated in atacicept treated rheumatoid arthritis patients and varied from 0–25% depending on the antigen, reflecting the modest loss in total IgG 128. Thus, there does not appear to be either a markedly increased infection risk or major therapeutic benefit of BAFF/APRIL blockade over BAFF inhibition alone. However, it is possible that a subset of individuals may respond better to a particular therapy.

6.3. Does BAFF inhibition alter B cell selection in humans?

Our mouse studies suggest considerable variability in the effects of BAFF inhibition on the regulation of B cell tolerance. To address this question in humans, we analyzed the B cell repertoire of lupus patients treated with belimumab for more than 7 years, a duration sufficient to allow complete turnover of the naïve B cell repertoire 122.

Naïve B cells:

Repertoire differences between transitional and mature naïve B cells reflect negative selection at the transitional checkpoint. Using next generation sequencing, we first asked whether belimumab treatment alters normal transitional selection. Surprisingly, we observed no impact of belimumab on VH family usage within the naïve repertoire compared with matched SLE controls. Furthermore, we observed no differences in pre- and post-belimumab naïve repertoire in the subset of patients were longitudinal samples were available. Prior studies have indicated that ~40% of naïve B cells from lupus patients are autoreactive, with a repertoire skewed towards VH4–34, 4–39 and 3–21 usage and more acidic CDR3 regions 70. However, an in-depth analysis of multiple parameters of the B cell repertoire (including VDJ usage, D length, charge, and related biophysical characteristics) uncovered no additional differences between belimumab treated subjects and disease controls. Finally, VH4–34 is an intrinsically-autoreactive heavy chain gene that is censored in the GC and plasma cell compartments of healthy subjects, but enriched among lupus plasma cells, especially during disease flares 87. Despite this putative role in lupus pathogenesis, we observed no preferential loss of VH4–34 in belimumab treated patients compared with SLE controls. Thus, despite profound depletion of the naïve compartment, BAFF inhibition resulted in no preferential loss or enrichment of specific VH genes among remaining naïve B cells 122.

Activated B cells and plasma cells:

Examination of the repertoire of mutated CD27 “activated” naïve B cells uncovered a loss of VH4–34 sequences between the naïve and activated, and the naïve and plasmablast stages, in belimumab treated subjects, but not lupus controls 122. Although the plasmablast compartment exhibited abundant clonal expansion in SLE, no differences were noted between patients with or without BAFF inhibition. These findings are provocative, since, together with mouse studies, they suggest an alternate mechanism for BAFF inhibition beyond impacts on B cell selection at the T1-T3 transition. One potential mechanism is anergy induction in autoreactive naïve B cells such that they are unlikely to be activated via either EF or GC pathways. This model is consistent studies from the Diamond group showing that anergic B cells are infrequent in lupus patients, but similar in number in belimumab treated subjects and healthy controls 129. An alternate explanation could be modulated endosomal TLR/TACI interaction in activated B cells resulting in reduced survival in low BAFF settings.

Overall, our findings are consistent with animal studies showing a limited impact of BAFF inhibition on the naïve B cell repertoire in the presence of physiologic competition from non-autoreactive B cells. As in mice, a modest effect of belimumab on the antigen-activated autoreactive immunoglobulin gene repertoire is apparent, but it is unclear whether these events are responsible for declining autoantibody titers or whether loss of autoantibodies reflects chronic B cell depletion and the loss of plasma cells over time.

7. Other effects of BAFF inhibition on inflammation

Since BAFF receptors are expressed by T cells and myeloid cells it is possible that BAFF inhibition alters the activation of these cells in the inflammatory environment. In T cells, roles for BAFF are controversial since both activating and inhibitory BAFF-R effects have been reported 130. BAFF supports the survival and differentiation of monocytes and enhances the activation of human myeloid DCs. TACI deficient macrophages skew towards M2 differentiation, and thus induced less inflammation with reduced infection clearance 131, 132. In murine arthritis, BAFF-silenced synovial dendritic cells remained immature and were unable to support T helper type 17 (Th17) differentiation 133.

BAFF inhibition may also impact inflammation by modulating B cell regulatory function. For example, an intriguing new study showed that TACI-dependent production of the regulatory cytokine IL-10 by IgA+ plasma cells protected against experimental autoimmune encephalomyelitis (EAE) in BAFF-Tg mice 134. In a separate study, local APRIL production by infiltrating macrophages induced IL-10 production by astrocytes to limit multiple sclerosis (MS) lesions 135. Together, these mechanisms may account for the unanticipated increase in disease flares in the atacicept MS clinical trials 136, and suggest caution with dual BAFF/APRIL inhibition in other inflammatory diseases. Similarly, whereas global and B cell-specific BAFF-R deletion protected against atherosclerosis, BAFF neutralization increased and BAFF overexpression limited atheroma formation in mouse models 137, 138. Mechanistically, BAFF-dependent TACI signals in atheroma M1 macrophages decreased the production of the pro-atherogenic chemokine CXCL10, resulting in accelerated atherosclerosis after global and myeloid-specific TACI deletion 137. Since elevated serum BAFF levels are associated with increased atherosclerosis risk in human SLE 139, the clinical relevance of these animal models to human lupus is unclear. However, these studies emphasize the need for long-term studies of cardiovascular event rates in belimumab treated patients. In this context, protective effect of BAFF inhibition on adiposity and insulin resistance may also be relevant 140, 141.

Finally, multiple mouse studies have demonstrated decreased renal inflammation in BAFF-deficient and BAFF-R-Ig/TACI-Ig-treated lupus models in excess of impacts on serum autoantibody titers. In the NZB/W model, our studies have shown protection from progressive lupus nephritis that could not be explained by a reduction in the autoantibody levels or reduced glomerular immune complex formation. Rather, gene profiling studies in NZM2410 mice indicated that progression from proteinuria to ESRD correlated with podocyte loss, tubular and endothelial dysfunction, and metabolic derangements, which was limited in BAFF-R-Ig treated animals despite ongoing glomerular immune complex deposits and inflammatory infiltrates 142. How BAFF inhibition mediated this effect and whether a similar therapeutic benefit will be observed in human clinical trials has not yet been determined.

8. Concluding remarks

Much has been learned about BAFF, APRIL, and their receptors since their initial discoveries two decades ago, but the full therapeutic benefits of targeting this family have yet to be achieved. In addition to clinical successes of belimumab in lupus trials, the recent genetic association of variants impacting BAFF levels with SLE and MS risk confirms the importance of this cytokine in human autoimmunity. Recent data have increased our understanding of how BAFF interacts with both the BCR and TLR pathways to regulate B cell survival, activation and differentiation via EF- and GC-dependent pathways. These data expand BAFF’s role beyond its essential ability to support naïve B cell survival to a complex one integrating innate and adaptive B cell responses during humoral autoimmunity. In this model, increased serum BAFF levels associated with inflammatory states and following therapeutic B cell depletion may relax the stringency of B cell selection within both the naïve and activated B cell compartments. Selective BAFF or combined BAFF/APRIL inhibition appears safe in human studies, with chronic BAFF blockade decreasing lupus flare rates and progressive organ damage over time. However, the clinical response to BAFF inhibition is heterogenous in both murine models and human lupus patients, emphasizing the need for improved therapeutic approaches and mechanistic biomarkers that can identify patients likely to respond to specific therapies.

Acknowledgements:

This work was supported by the National Institutes of Health under award numbers: K08AI112993 (SWJ), 1R03AI109324-01 (AD), RO1 AR064811-01 (AD) and NIH R21 AR070540 (AD). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Additional support provided by the American College of Rheumatology (ACR) Rheumatology Research Foundation (RRF) Career Development K Supplement (SWJ); by the Arthritis National Research Foundation (ANRF) Eng Tan Scholar Award (SWJ); and by a Lupus Research Alliance, Novel Research Grant (SWJ).

References

  • 1.Dall’Era M, Bruce IN, Gordon C, Manzi S, McCaffrey J and Lipsky PE. Current challenges in the development of new treatments for lupus. Annals of the rheumatic diseases 2019. [DOI] [PubMed]
  • 2.Moore PA, Belvedere O, Orr A, et al. BLyS: member of the tumor necrosis factor family and B lymphocyte stimulator. Science 1999; 285: 260–3. [DOI] [PubMed] [Google Scholar]
  • 3.Mackay F, Woodcock SA, Lawton P, et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med 1999; 190: 1697–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Gross JA, Dillon SR, Mudri S, et al. TACI-Ig neutralizes molecules critical for B cell development and autoimmune disease. impaired B cell maturation in mice lacking BLyS. Immunity 2001; 15: 289–302. [DOI] [PubMed] [Google Scholar]
  • 5.Bosello S, Youinou P, Daridon C, et al. Concentrations of BAFF correlate with autoantibody levels, clinical disease activity, and response to treatment in early rheumatoid arthritis. J Rheumatol 2008; 35: 1256–64. [PubMed] [Google Scholar]
  • 6.Petri M, Stohl W, Chatham W, et al. BLyS plasma concentrations correlate with disease activity and levels of anti-dsDNA autoantibodies and immunoglobulins (IgG) in a SLE patient observational study. Arthritis and Rheumatism 2003; 48: S655 (abstract). [Google Scholar]
  • 7.Urowitz MB, Ohsfeldt RL, Wielage RC, Kelton KA, Asukai Y and Ramachandran S. Organ damage in patients treated with belimumab versus standard of care: a propensity score-matched comparative analysis. Annals of the rheumatic diseases 2019; 78: 372–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Furie R, Petri M, Zamani O, 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: 3918–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Navarra SV, Guzman RM, Gallacher AE, et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet 2011; 377: 721–31. [DOI] [PubMed] [Google Scholar]
  • 10.Mackay F and Schneider P. Cracking the BAFF code. Nat Rev Immunol 2009; 9: 491–502. [DOI] [PubMed] [Google Scholar]
  • 11.Liu Y, Xu L, Opalka N, Kappler J, Shu HB and Zhang G. Crystal structure of sTALL-1 reveals a virus-like assembly of TNF family ligands. Cell 2002; 108: 383–94. [DOI] [PubMed] [Google Scholar]
  • 12.Vigolo M, Chambers MG, Willen L, et al. A loop region of BAFF controls B cell survival and regulates recognition by different inhibitors. Nature communications 2018; 9: 1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Shin W, Lee HT, Lim H, et al. BAFF-neutralizing interaction of belimumab related to its therapeutic efficacy for treating systemic lupus erythematosus. Nature communications 2018; 9: 1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gavin AL, Ait-Azzouzene D, Ware CF and Nemazee D. DeltaBAFF, an alternate splice isoform that regulates receptor binding and biopresentation of the B cell survival cytokine, BAFF. J Biol Chem 2003; 278: 38220–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Steri MFICGOMP, University of Milan do Porto-Hospital Santo A, Unit for Multidisciplinary Research in A, et al. Overexpression of the Cytokine BAFF and Autoimmunity Risk. N Engl J Med 2017; 376: 1615–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Baert L, Manfroi B, Casez O, Sturm N and Huard B. The role of APRIL - A proliferation inducing ligand - In autoimmune diseases and expectations from its targeting. J Autoimmun 2018; 95: 179–90. [DOI] [PubMed] [Google Scholar]
  • 17.Lopez-Fraga M, Fernandez R, Albar JP and Hahne M. Biologically active APRIL is secreted following intracellular processing in the Golgi apparatus by furin convertase. EMBO reports 2001; 2: 945–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pradet-Balade B, Medema JP, Lopez-Fraga M, et al. An endogenous hybrid mRNA encodes TWE-PRIL, a functional cell surface TWEAK-APRIL fusion protein. Embo J 2002; 21: 5711–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Furuya T, Koga M, Hikami K, Kawasaki A and Tsuchiya N. Effects of APRIL (TNFSF13) polymorphisms and splicing isoforms on the secretion of soluble APRIL. Modern rheumatology 2012; 22: 541–9. [DOI] [PubMed] [Google Scholar]
  • 20.Varfolomeev E, Kischkel F, Martin F, et al. APRIL-deficient mice have normal immune system development. Mol Cell Biol 2004; 24: 997–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Castigli E, Scott S, Dedeoglu F, et al. Impaired IgA class switching in APRIL-deficient mice. Proc Natl Acad Sci U S A 2004; 101: 3903–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Smirnova AS, Andrade-Oliveira V and Gerbase-Delima M. Identification of new splice variants of the genes BAFF and BCMA. Mol Immunol 2008; 45: 1179–83. [DOI] [PubMed] [Google Scholar]
  • 23.Schuh E, Musumeci A, Thaler FS, et al. Human Plasmacytoid Dendritic Cells Display and Shed B Cell Maturation Antigen upon TLR Engagement. Journal of immunology (Baltimore, Md : 1950) 2017; 198: 3081–8. [DOI] [PubMed] [Google Scholar]
  • 24.Meinl E, Thaler FS and Lichtenthaler SF. Shedding of BAFF/APRIL Receptors Controls B Cells. Trends Immunol 2018; 39: 673–6. [DOI] [PubMed] [Google Scholar]
  • 25.Smulski CR, Kury P, Seidel LM, et al. BAFF- and TACI-Dependent Processing of BAFFR by ADAM Proteases Regulates the Survival of B Cells. Cell Rep 2017; 18: 2189–202. [DOI] [PubMed] [Google Scholar]
  • 26.Smulski CR and Eibel H. BAFF and BAFF-Receptor in B Cell Selection and Survival. Frontiers in immunology 2018; 9: 2285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rowland SL, Leahy KF, Halverson R, Torres RM and Pelanda R. BAFF receptor signaling aids the differentiation of immature B cells into transitional B cells following tonic BCR signaling. J Immunol 2010; 185: 4570–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tussiwand R, Rauch M, Fluck LA and Rolink AG. BAFF-R expression correlates with positive selection of immature B cells. Eur J Immunol 2012; 42: 206–16. [DOI] [PubMed] [Google Scholar]
  • 29.Stadanlick JE, Kaileh M, Karnell FG, et al. Tonic B cell antigen receptor signals supply an NF-kappaB substrate for prosurvival BLyS signaling. Nat Immunol 2008; 9: 1379–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Henley T, Kovesdi D and Turner M. B-cell responses to B-cell activation factor of the TNF family (BAFF) are impaired in the absence of PI3K delta. Eur J Immunol 2008; 38: 3543–8. [DOI] [PubMed] [Google Scholar]
  • 31.Mackay F, Figgett WA, Saulep D, Lepage M and Hibbs ML. B-cell stage and context-dependent requirements for survival signals from BAFF and the B-cell receptor. Immunol Rev 2010; 237: 205–25. [DOI] [PubMed] [Google Scholar]
  • 32.Claudio E, Brown K, Park S, Wang H and Siebenlist U. BAFF-induced NEMO-independent processing of NF-kappa B2 in maturing B cells. Nat Immunol 2002; 3: 958–65. [DOI] [PubMed] [Google Scholar]
  • 33.Almaden JV, Tsui R, Liu YC, et al. A pathway switch directs BAFF signaling to distinct NFkappaB transcription factors in maturing and proliferating B cells. Cell Rep 2014; 9: 2098–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Scholz JL, Crowley JE, Tomayko MM, et al. BLyS inhibition eliminates primary B cells but leaves natural and acquired humoral immunity intact. Proc Natl Acad Sci U S A 2008; 105: 15517–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Benson MJ, Dillon SR, Castigli E, et al. Cutting edge: the dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol 2008; 180: 3655–9. [DOI] [PubMed] [Google Scholar]
  • 36.Schneider P The role of APRIL and BAFF in lymphocyte activation. Curr Opin Immunol 2005; 17: 282–9. [DOI] [PubMed] [Google Scholar]
  • 37.von Bulow GU, van Deursen JM and Bram RJ. Regulation of the T-independent humoral response by TACI. Immunity 2001; 14: 573–82. [DOI] [PubMed] [Google Scholar]
  • 38.Seshasayee D, Valdez P, Yan M, Dixit VM, Tumas D and Grewal IS. Loss of TACI causes fatal lymphoproliferation and autoimmunity, establishing TACI as an inhibitory BLyS receptor. Immunity 2003; 18: 279–88. [DOI] [PubMed] [Google Scholar]
  • 39.Tsuji S, Cortesao C, Bram RJ, Platt JL and Cascalho M. TACI deficiency impairs sustained Blimp-1 expression in B cells decreasing long-lived plasma cells in the bone marrow. Blood 2011; 118: 5832–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mackay F and Schneider P. TACI, an enigmatic BAFF/APRIL receptor, with new unappreciated biochemical and biological properties. Cytokine Growth Factor Rev 2008; 19: 263–76. [DOI] [PubMed] [Google Scholar]
  • 41.Laurent SA, Hoffmann FS, Kuhn PH, et al. gamma-Secretase directly sheds the survival receptor BCMA from plasma cells. Nature communications 2015; 6: 7333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hoffmann FS, Kuhn PH, Laurent SA, et al. The immunoregulator soluble TACI is released by ADAM10 and reflects B cell activation in autoimmunity. J Immunol 2015; 194: 542–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Garcia-Carmona Y, Ting AT, Radigan L, et al. TACI Isoforms Regulate Ligand Binding and Receptor Function. Frontiers in immunology 2018; 9: 2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Coquery CM and Erickson LD. Regulatory roles of the tumor necrosis factor receptor BCMA. Critical reviews in immunology 2012; 32: 287–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yasuda K, Richez C, Maciaszek JW, et al. Murine dendritic cell type I IFN production induced by human IgG-RNA immune complexes is IFN regulatory factor (IRF)5 and IRF7 dependent and is required for IL-6 production. J Immunol 2007; 178: 6876–85. [DOI] [PubMed] [Google Scholar]
  • 46.Treml LS, Carlesso G, Hoek KL, et al. TLR stimulation modifies BLyS receptor expression in follicular and marginal zone B cells. J Immunol 2007; 178: 7531–9. [DOI] [PubMed] [Google Scholar]
  • 47.Katsenelson N, Kanswal S, Puig M, Mostowski H, Verthelyi D and Akkoyunlu M. Synthetic CpG oligodeoxynucleotides augment BAFF- and APRIL-mediated immunoglobulin secretion. Eur J Immunol 2007; 37: 1785–95. [DOI] [PubMed] [Google Scholar]
  • 48.He B, Santamaria R, Xu W, et al. The transmembrane activator TACI triggers immunoglobulin class switching by activating B cells through the adaptor MyD88. Nat Immunol 2010; 11: 836–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sintes J, Gentile M, Zhang S, et al. mTOR intersects antibody-inducing signals from TACI in marginal zone B cells. Nature communications 2017; 8: 1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Figgett WA, Deliyanti D, Fairfax KA, Quah PS, Wilkinson-Berka JL and Mackay F. Deleting the BAFF receptor TACI protects against systemic lupus erythematosus without extensive reduction of B cell numbers. Journal of autoimmunity 2015; 61: 9–16. [DOI] [PubMed] [Google Scholar]
  • 51.Arkatkar T, Jacobs HM, Du SW, et al. TACI deletion protects against progressive murine lupus nephritis induced by BAFF overexpression. Kidney international 2018; 94: 728–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sindhava VJ, Oropallo MA, Moody K, et al. A TLR9-dependent checkpoint governs B cell responses to DNA-containing antigens. J Clin Invest 2017; 127: 1651–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nemazee D Mechanisms of central tolerance for B cells. Nat Rev Immunol 2017; 17: 281–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Thompson JS, Bixler SA, Qian F, et al. BAFF-R, a newly identified TNF receptor that specifically interacts with BAFF. Science 2001; 293: 2108–11. [DOI] [PubMed] [Google Scholar]
  • 55.Yang M, Sun L, Wang S, et al. Novel function of B cell-activating factor in the induction of IL-10-producing regulatory B cells. J Immunol 2010; 184: 3321–5. [DOI] [PubMed] [Google Scholar]
  • 56.Ponnuswamy P, Joffre J, Herbin O, et al. Angiotensin II synergizes with BAFF to promote atheroprotective regulatory B cells. Sci Rep 2017; 7: 4111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Matsushita T, Kobayashi T, Mizumaki K, et al. BAFF inhibition attenuates fibrosis in scleroderma by modulating the regulatory and effector B cell balance. Science advances 2018; 4: eaas9944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ma N, Zhang Y, Liu Q, et al. B cell activating factor (BAFF) selects IL-10(−)B cells over IL-10(+)B cells during inflammatory responses. Mol Immunol 2017; 85: 18–26. [DOI] [PubMed] [Google Scholar]
  • 59.Thien M, Phan TG, Gardam S, et al. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity 2004; 20: 785–98. [DOI] [PubMed] [Google Scholar]
  • 60.Lesley R, Xu Y, Kalled SL, et al. Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF. Immunity 2004; 20: 441–53. [DOI] [PubMed] [Google Scholar]
  • 61.Hondowicz BD, Alexander ST, Quinn WJ 3rd, et al. The role of BLyS/BLyS receptors in anti-chromatin B cell regulation. Int Immunol 2007; 19: 465–75. [DOI] [PubMed] [Google Scholar]
  • 62.Schweighoffer E, Vanes L, Nys J, et al. The BAFF Receptor Transduces Survival Signals by Co-opting the B Cell Receptor Signaling Pathway. Immunity 2013; 38: 475–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hobeika E, Levit-Zerdoun E, Anastasopoulou V, et al. CD19 and BAFF-R can signal to promote B-cell survival in the absence of Syk. EMBO J 2015; 34: 925–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jellusova J, Miletic AV, Cato MH, et al. Context-specific BAFF-R signaling by the NF-kappaB and PI3K pathways. Cell Rep 2013; 5: 1022–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gardam S and Brink R. Non-Canonical NF-kappaB Signaling Initiated by BAFF Influences B Cell Biology at Multiple Junctures. Front Immunol 2014; 4: 509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Nikbakht N, Migone TS, Ward CP and Manser T. Cellular competition independent of BAFF/B lymphocyte stimulator results in low frequency of an autoreactive clonotype in mature polyclonal B cell compartments. J Immunol 2011; 187: 37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tognotti E Program to eradicate malaria in Sardinia, 1946–1950. Emerg Infect Dis 2009; 15: 1460–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liu XQ, Stacey KJ, Horne-Debets JM, et al. Malaria infection alters the expression of B-cell activating factor resulting in diminished memory antibody responses and survival. Eur J Immunol 2012; 42: 3291–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hart GT, Akkaya M, Chida AS, et al. The Regulation of Inherently Autoreactive VH4–34-Expressing B Cells in Individuals Living in a Malaria-Endemic Area of West Africa. J Immunol 2016; 197: 3841–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E and Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science 2003; 301: 1374–7. [DOI] [PubMed] [Google Scholar]
  • 71.Groom JR, Fletcher CA, Walters SN, et al. BAFF and MyD88 signals promote a lupuslike disease independent of T cells. J Exp Med 2007; 204: 1959–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Du SW, Jacobs HM, Arkatkar T, Rawlings DJ and Jackson SW. Integrated B Cell, Toll-like, and BAFF Receptor Signals Promote Autoantibody Production by Transitional B Cells. Journal of immunology (Baltimore, Md : 1950) 2018; 201: 3258–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jacobs HM, Thouvenel CD, Leach S, et al. Cutting Edge: BAFF Promotes Autoantibody Production via TACI-Dependent Activation of Transitional B Cells. J Immunol 2016; 196: 3525–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yan M, Wang H, Chan B, et al. Activation and accumulation of B cells in TACI-deficient mice. Nat Immunol 2001; 2: 638–43. [DOI] [PubMed] [Google Scholar]
  • 75.Groom J, Kalled SL, Cutler AH, et al. Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjogren’s syndrome. J Clin Invest 2002; 109: 59–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fletcher CA, Sutherland AP, Groom JR, et al. Development of nephritis but not sialadenitis in autoimmune-prone BAFF transgenic mice lacking marginal zone B cells. Eur J Immunol 2006; 36: 2504–14. [DOI] [PubMed] [Google Scholar]
  • 77.Fletcher CA, Groom JR, Woehl B, Leung H, Mackay C and Mackay F. Development of autoimmune nephritis in genetically asplenic and splenectomized BAFF transgenic mice. J Autoimmun 2011; 36: 125–34. [DOI] [PubMed] [Google Scholar]
  • 78.Fairfax KA, Tsantikos E, Figgett WA, et al. BAFF-driven autoimmunity requires CD19 expression. Journal of autoimmunity 2015; 62: 1–10. [DOI] [PubMed] [Google Scholar]
  • 79.Nutt SL, Hodgkin PD, Tarlinton DM and Corcoran LM. The generation of antibody-secreting plasma cells. Nat Rev Immunol 2015; 15: 160–71. [DOI] [PubMed] [Google Scholar]
  • 80.William J, Euler C, Christensen S and Shlomchik MJ. Evolution of autoantibody responses via somatic hypermutation outside of germinal centers. Science 2002; 297: 2066–70. [DOI] [PubMed] [Google Scholar]
  • 81.Tipton CM, Fucile CF, Darce J, et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nat Immunol 2015; 16: 755–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jenks SA, Cashman KS, Zumaquero E, et al. Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus. Immunity 2018; 49: 725–39 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.DeFranco AL. Germinal centers and autoimmune disease in humans and mice. Immunol Cell Biol 2016; 94: 918–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shlomchik MJ, Luo W and Weisel F. Linking signaling and selection in the germinal center. Immunol Rev 2019; 288: 49–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Haberman AM, Gonzalez DG, Wong P, Zhang TT and Kerfoot SM. Germinal center B cell initiation, GC maturation, and the coevolution of its stromal cell niches. Immunol Rev 2019; 288: 10–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Suan D, Sundling C and Brink R. Plasma cell and memory B cell differentiation from the germinal center. Curr Opin Immunol 2017; 45: 97–102. [DOI] [PubMed] [Google Scholar]
  • 87.Cappione A 3rd, Anolik JH, Pugh-Bernard A, et al. Germinal center exclusion of autoreactive B cells is defective in human systemic lupus erythematosus. J Clin Invest 2005; 115: 3205–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vinuesa CG, Sanz I and Cook MC. Dysregulation of germinal centres in autoimmune disease. Nat Rev Immunol 2009; 9: 845–57. [DOI] [PubMed] [Google Scholar]
  • 89.Aloisi F and Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol 2006; 6: 205–17. [DOI] [PubMed] [Google Scholar]
  • 90.Domeier PP, Schell SL and Rahman ZS. Spontaneous germinal centers and autoimmunity. Autoimmunity 2017; 50: 4–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Arbuckle MR, James JA, Kohlhase KF, Rubertone MV, Dennis GJ and Harley JB. Development of anti-dsDNA autoantibodies prior to clinical diagnosis of systemic lupus erythematosus. Scand J Immunol 2001; 54: 211–9. [DOI] [PubMed] [Google Scholar]
  • 92.Kalled SL. Impact of the BAFF/BR3 axis on B cell survival, germinal center maintenance and antibody production. Semin Immunol 2006; 18: 290–6. [DOI] [PubMed] [Google Scholar]
  • 93.Rahman ZS, Rao SP, Kalled SL and Manser T. Normal Induction but Attenuated Progression of Germinal Center Responses in BAFF and BAFF-R Signaling-Deficient Mice. J Exp Med 2003; 198: 1157–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Goenka R, Matthews AH, Zhang B, et al. Local BLyS production by T follicular cells mediates retention of high affinity B cells during affinity maturation. J Exp Med 2014; 211: 45–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Suurmond J, Atisha-Fregoso Y, Barlev AN, et al. Patterns of ANA+ B cells for SLE patient stratification. JCI insight 2019; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Jacob N, Guo S, Mathian A, et al. B Cell and BAFF dependence of IFN-alpha-exaggerated disease in systemic lupus erythematosus-prone NZM 2328 mice. J Immunol 2011; 186: 4984–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Jacob CO, Pricop L, Putterman C, et al. Paucity of clinical disease despite serological autoimmunity and kidney pathology in lupus-prone New Zealand mixed 2328 mice deficient in BAFF. J Immunol 2006; 177: 2671–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Jacob CO, Yu N, Sindhava V, et al. Differential Development of Systemic Lupus Erythematosus in NZM 2328 Mice Deficient in Discrete Pairs of BAFF Receptors. Arthritis Rheumatol 2015; 67: 2523–35. [DOI] [PubMed] [Google Scholar]
  • 99.Tran NL, Schneider P and Santiago-Raber ML. TACI-dependent APRIL signaling maintains autoreactive B cells in a mouse model of systemic lupus erythematosus. European journal of immunology 2017; 47: 713–23. [DOI] [PubMed] [Google Scholar]
  • 100.Jacob CO, Guo S, Jacob N, et al. Dispensability of APRIL to the development of systemic lupus erythematosus in NZM 2328 mice. Arthritis Rheum 2012; 64: 1610–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Jacob CO, Yu N, Guo S, et al. Development of systemic lupus erythematosus in NZM 2328 mice in the absence of any single BAFF receptor. Arthritis Rheum 2013. [DOI] [PMC free article] [PubMed]
  • 102.Ju ZL, Shi GY, Zuo JX and Zhang JW. Unexpected development of autoimmunity in BAFF-R-mutant MRL-lpr mice. Immunology 2007; 120: 281–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jiang C, Loo WM, Greenley EJ, Tung KS and Erickson LD. B cell maturation antigen deficiency exacerbates lymphoproliferation and autoimmunity in murine lupus. J Immunol 2011; 186: 6136–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Oon S, Huq M, Godfrey TA and Mmnuea Nikpour Systematic review, and meta-analysis of steroid-sparing effect, of biologic agents in randomized, placebo-controlled phase 3 trials for systemic lupus erythematosus. Seminars in arthritis and rheumatism 2018; 48: 221–39. [DOI] [PubMed] [Google Scholar]
  • 105.Merrill JT, Wallace DJ, Wax S, et al. Efficacy and Safety of Atacicept in Patients With Systemic Lupus Erythematosus. Arthritis Rheumatol 2018; 70: 266–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kowalczyk-Quintas C, Chevalley D, Willen L, Jandus C, Vigolo M and Schneider P. Inhibition of Membrane-Bound BAFF by the Anti-BAFF Antibody Belimumab. Front Immunol 2018; 9: 2698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Rottman JB and Willis CR. Mouse models of systemic lupus erythematosus reveal a complex pathogenesis. Vet Pathol 2010; 47: 664–76. [DOI] [PubMed] [Google Scholar]
  • 108.Boneparth A, Woods M, Huang W, Akerman M, Lesser M and Davidson A. The Effect of BAFF Inhibition on Autoreactive B-Cell Selection in Murine Systemic Lupus Erythematosus. Mol Med 2016; 22: 173–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Moisini I, Huang W, Bethunaickan R, et al. The Yaa locus and IFN-alpha fine-tune germinal center B cell selection in murine systemic lupus erythematosus. J Immunol 2012; 189: 4305–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ramanujam M, Bethunaickan R, Huang W, Tao H, Madaio MP and Davidson A. Selective blockade of BAFF for the prevention and treatment of systemic lupus erythematosus nephritis in NZM2410 mice. Arthritis Rheum 2010; 62: 1457–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ramanujam M, Wang X, Huang W, et al. Mechanism of action of transmembrane activator and calcium modulator ligand interactor-Ig in murine systemic lupus erythematosus. J Immunol 2004; 173: 3524–34. [DOI] [PubMed] [Google Scholar]
  • 112.Kahn P, Ramanujam M, Bethunaickan R, et al. Prevention of murine antiphospholipid syndrome by BAFF blockade. Arthritis Rheum 2008; 58: 2824–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ramanujam M, Wang X, Huang W, et al. Similarities and differences between selective and nonselective BAFF blockade in murine SLE. J Clin Invest 2006; 116: 724–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Haselmayer P, Vigolo M, Nys J, Schneider P and Hess H. A mouse model of systemic lupus erythematosus responds better to soluble TACI than to soluble BAFFR, correlating with depletion of plasma cells. Eur J Immunol 2017; 47: 1075–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Huard B, Tran NL, Benkhoucha M, Manzin-Lorenzi C and Santiago-Raber ML. Selective APRIL blockade delays systemic lupus erythematosus in mouse. PLoS One 2012; 7: e31837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bethunaickan R, Berthier CC, Zhang W, et al. Identification of stage-specific genes associated with lupus nephritis and response to remission induction in (NZB x NZW)F1 and NZM2410 mice. Arthritis Rheumatol 2014; 66: 2246–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Liu W, Szalai A, Zhao L, et al. Control of spontaneous B lymphocyte autoimmunity with adenovirus-encoded soluble TACI. Arthritis Rheum 2004; 50: 1884–96. [DOI] [PubMed] [Google Scholar]
  • 118.Liu Z, Bethunaickan R, Huang W, Ramanujam M, Madaio MP and Davidson A. IFN-alpha confers resistance of systemic lupus erythematosus nephritis to therapy in NZB/W F1 mice. Journal of immunology (Baltimore, Md : 1950) 2011; 187: 1506–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Liu Z, Bethunaickan R, Huang W, Ramanujam M, Madaio MP and Davidson A. IFN-{alpha} Confers Resistance of Systemic Lupus Erythematosus Nephritis to Therapy in NZB/W F1 Mice. J Immunol 2011; 187: 1506–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Huang W, Moisini I, Bethunaickan R, et al. BAFF/APRIL inhibition decreases selection of naive but not antigen-induced autoreactive B cells in murine systemic lupus erythematosus. J Immunol 2011; 187: 6571–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.van Vollenhoven RF, Petri MA, Cervera R, et al. Belimumab in the treatment of systemic lupus erythematosus: high disease activity predictors of response. Ann Rheum Dis 2012; 71: 1343–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Huang W, Quach TD, Dascalu C, et al. Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients. JCI insight 2018; 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Furie RA, Wallace DJ, Aranow C, et al. Long-Term Safety and Efficacy of Belimumab in Patients With Systemic Lupus Erythematosus: A Continuation of a Seventy-Six-Week Phase III Parent Study in the United States. Arthritis Rheumatol 2018. [DOI] [PMC free article] [PubMed]
  • 124.Hoffman RW, Merrill JT, Alarcon-Riquelme MM, et al. Gene Expression and Pharmacodynamic Changes in 1,760 Systemic Lupus Erythematosus Patients From Two Phase III Trials of BAFF Blockade With Tabalumab. Arthritis & rheumatology (Hoboken, NJ) 2017; 69: 643–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Thai LH, Le Gallou S, Robbins A, et al. BAFF and CD4(+) T cells are major survival factors for long-lived splenic plasma cells in a B-cell-depletion context. Blood 2018; 131: 1545–55. [DOI] [PubMed] [Google Scholar]
  • 126.Teng YKON, Bruce IN, Diamond B, et al. Phase III, multicentre, randomised, double-blind, placebo-controlled, 104-week study of subcutaneous belimumab administered in combination with rituximab in adults with systemic lupus erythematosus (SLE): BLISS-BELIEVE study protocol. BMJ open 2019; 9: e025687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Aranow C, Dall’Era M, Byron M, et al. Phase 2 trial of induction therapy with antiCD20 (rituximab) followed by maintenance therapy with anti-BAFF (belimumab) in patients with active lupus nephritis. Annals of the rheumatic diseases 2018: FRI0305.
  • 128.Genovese MC, Kinnman N, de La Bourdonnaye G, Pena Rossi C and Tak PP. Atacicept in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor antagonist therapy: Results of a phase II, randomized, placebo-controlled, dose-finding trial. Arthritis & Rheumatism 2011; 63: 1793–803. [DOI] [PubMed] [Google Scholar]
  • 129.Malkiel S, Jeganathan V, Wolfson S, et al. Checkpoints for Autoreactive B Cells in the Peripheral Blood of Lupus Patients Assessed by Flow Cytometry. Arthritis Rheumatol 2016; 68: 2210–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Bloom DDW-M, Reshetylo S, Nytes C, Goodsett CT and Hematti PW-M. Blockade of BAFF Receptor BR3 on T Cells Enhances Their Activation and Cytotoxicity. J Immunother 2018; 41: 213–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.An Y, Zhou Y, Bi L, et al. Combined immunosuppressive treatment (CIST) in lupus nephritis: a multicenter, randomized controlled study. Clinical rheumatology 2018. [DOI] [PubMed]
  • 132.Allman WR, Dey R, Liu L, et al. TACI deficiency leads to alternatively activated macrophage phenotype and susceptibility to Leishmania infection. Proceedings of the National Academy of Sciences of the United States of America 2015; 112: E4094–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Lai Kwan Lam Q, King Hung Ko O, Zheng BJ and Lu L. Local BAFF gene silencing suppresses Th17-cell generation and ameliorates autoimmune arthritis. Proc Natl Acad Sci U S A 2008; 105: 14993–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Rojas OL, Probstel AK, Porfilio EA, et al. Recirculating Intestinal IgA-Producing Cells Regulate Neuroinflammation via IL-10. Cell 2019; 176: 610–24 e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Baert L, Benkhoucha M, Popa N, et al. A proliferation-inducing ligand-mediated anti-inflammatory response of astrocytes in multiple sclerosis. Annals of neurology 2019. [DOI] [PubMed]
  • 136.Sergott RC, Bennett JL, Rieckmann P, et al. ATON: results from a Phase II randomized trial of the B-cell-targeting agent atacicept in patients with optic neuritis. Journal of the neurological sciences 2015; 351: 174–8. [DOI] [PubMed] [Google Scholar]
  • 137.Tsiantoulas D, Sage AZM, Goderle L, et al. B Cell-Activating Factor Neutralization Aggravates Atherosclerosis. Circulation 2018; 138: 2263–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Jackson SW, Scharping NE, Jacobs HM, Wang S, Chait A and Rawlings DJ. Cutting Edge: BAFF Overexpression Reduces Atherosclerosis via TACI-Dependent B Cell Activation. J Immunol 2016; 197: 4529–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Theodorou E, Nezos A, Antypa EG, et al. B-cell activating factor and related genetic variants in lupus related atherosclerosis. J Autoimmun 2018; 92: 87–92. [DOI] [PubMed] [Google Scholar]
  • 140.Liu L, Inouye KE, Allman WR, et al. TACI-Deficient Macrophages Protect Mice Against Metaflammation and Obesity-Induced Dysregulation of Glucose Homeostasis. Diabetes 2018; 67: 1589–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Wang Q, Racine JJ, Ratiu JJ, et al. Transient BAFF Blockade Inhibits Type 1 Diabetes Development in Nonobese Diabetic Mice by Enriching Immunoregulatory B Lymphocytes Sensitive to Deletion by Anti-CD20 Cotherapy. J Immunol 2017; 199: 3757–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bethunaickan R, Berthier CC, Zhang W, Kretzler M and Davidson A. Comparative Transcriptional Profiling of 3 Murine Models of SLE Nephritis Reveals Both Unique and Shared Regulatory Networks. PLoS One 2013; 8: e77489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Turazzi N, Fazio G, Rossi V, et al. Engineered T cells towards TNFRSF13C (BAFFR): a novel strategy to efficiently target B-cell acute lymphoblastic leukaemia. British journal of haematology 2018; 182: 939–43. [DOI] [PubMed] [Google Scholar]
  • 144.Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. The New England journal of medicine 2019; 380: 1726–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Lee L, Draper B, Chaplin N, et al. An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma. Blood 2018; 131: 746–58. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES