Skip to main content
  • Review
  • Published:

Cytokine disturbances in systemic lupus erythematosus

Abstract

The pathogenesis of systemic lupus erythematosus (SLE) is complex, and the resulting disease manifestations are heterogeneous. Cytokine dysregulation is pervasive, and their protein and gene expression profiles may serve as markers of disease activity and severity. Importantly, biologic agents that target specific cytokines may represent novel therapies for SLE. Four cytokines (IL-6, TNFα, IFNα, and BLyS) are being evaluated as therapeutic targets in SLE. The present review will examine the roles of each of these cytokines in murine and human SLE, and will summarize results from clinical trials of agents that target these cytokines.

Introduction

Cytokines collectively play key roles in the regulation of systemic inflammation, local tissue damage, and immunomodulation. Not surprisingly, cytokines often play direct roles in disease pathogenesis, including that of systemic lupus erythematosus (SLE). To the chagrin of both investigator and practitioner, the effects of cytokines are pleiotropic and include both synergistic and antagonistic effects on other cytokines - thereby introducing tremendous complexity that has often led to diametrically opposing conclusions in different studies. In the present review, we focus on four cytokines that have received great attention either as candidate biomarkers for disease activity and/or as candidate targets of novel biologic agents.

Interleukin-6

General biology of IL-6

IL-6 is a pleiotropic 26 kDa protein produced by a panoply of cell types, and affects the function of an equally broad spectrum of cell types. Of greatest relevance to SLE is the ability of IL-6 to promote activation and/or differentiation of cells central to the development of systemic autoimmunity and the attendant pathologic inflammatory responses, including T cells, B cells, macrophages, and neutrophils [1].

IL-6 signal transduction occurs via coordinated interactions between the 80 kDa IL-6 binding chain (IL-6 receptor (IL-6R), IL-6R α chain, CD126) and the 130 kDa signal transducing chain (gp130, IL-6R β chain, CD130). IL-6 can bind to membrane IL-6R, thereby inducing homodimerization of gp130 and leading to activation of gp130-associated JAK1 and tyrosine phosphorylation of gp130. Alternatively (and not mutually exclusively), IL-6R can be enzymatically cleaved from the cell surface, thereby producing soluble IL-6R. Soluble IL-6R can then bind circulating IL-6 and form IL-6/IL-6R complexes that act on cells expressing gp130 [1].

IL-6 in murine systemic lupus erythematosus

Studies in multiple murine SLE models point to an essential role for the IL-6 pathway in SLE. Genetic deficiency of IL-6 in MRL/lpr mice delays disease development, reduces CD4+ and CD8+ T-cell numbers, decreases glomerular IgG and C3 deposition, downregulates renal parenchymal vascular cell adhesion molecule-1 (VCAM-1) expression, and diminishes kidney macrophage infiltration [2]. This last effect may be especially important, in as much as a close association between activated renal macrophages and disease has been documented in (NZB × NZW)F1 (BWF) mice [3].

Additional important observations have been made with BWF mice. Exogenous IL-6 increases IgG anti-DNA autoantibody production by B cells isolated from clinically affected BWF mice, whereas neutralization of IL-6 via either addition of an anti-IL-6 mAb or macrophage depletion decreases production of such autoantibodies [4–6]. Building on these ex vivo findings, administration of human IL-6 to 6-month-old female BWF mice promoted accelerated membranoproliferative glomerulonephritis associated with marked upregulation of mesangial MHC class II antigens and glomerular inter-cellular adhesion molecule-1 (ICAM-1) expression. Treatment with cyclosporin inhibited the development of glomerulonephritis, distinguishing the effects of IL-6 on systemic inflammation from its direct effect on renal mesangial cells [7]. More impressive were the significant decreases in mortality, progression of proteinuria, and anti-dsDNA antibody levels in BWF mice chronically treated from 3 months of age with anti-IL-6 mAb or anti-IL6 receptor antibody [8, 9].

IL-6 is also associated with SLE in other murine SLE models. In pristine-induced SLE, renal disease was milder, and high levels of IgG anti-single-strand DNA, anti-dsDNA, and anti-chromatin antibodies were absent in IL-6-deficient mice [10]. In JunBΔep mice, development of an SLE-like phenotype (including skin lesions and immune complex glomerulonephritis) was linked to increased epidermal IL-6 secretion that arose from the specific loss of epidermal JunB. Intercrosses with IL-6-deficient mice abrogated the SLE phenotype [11].

IL-6 in human systemic lupus erythematosus

As in murine SLE, serum levels of IL-6 are elevated in human SLE and have correlated with disease activity or anti-dsDNA levels in some studies [12–14]. The increased frequency of IL-6-producing peripheral blood mononuclear cells correlates with disease severity/activity and treatment response [15, 16]. Of note, SLE B cells express IL-6R spontaneously and produce great amounts of IL-6 [17, 18]. Moreover, autoreactive T-cell clones from SLE patients also produce large amounts of IL-6, and thereby promote B-cell activation and autoantibody production [19]. Indeed, the copious spontaneous production of immunoglobulin by SLE B cells can be enhanced by exogenous IL-6 and diminished by neutralizing antibodies to IL-6 [13].

Aside from its systemic effects, IL-6 is closely linked with specific disease manifestations. Elevated cerebral spinal fluid levels of IL-6 are found in SLE patients with psychosis and may afford an effective measure of its diagnosis [20]. In SLE nephritis patients, urinary levels of IL-6 are elevated, correlate with titers of anti-dsDNA antibodies, and decrease following treatment [14, 21]. Local expression of IL-6 was documented in kidney tissue of SLE nephritis patients [22], arising at least in part from mesangial cells [23].

Therapeutic blockade of IL-6 in systemic lupus erythematosus

Given the considerable evidence pointing to a contributory role for IL-6 in SLE pathogenesis, the a priori likelihood that therapeutic targeting of this pathway in SLE will be efficacious is high. Tocilizumab, a mouse-human chimeric anti-human anti-IL-6R mAb, is already US Food and Drug Administration approved for the treatment of patients with rheumatoid arthritis. Tocilizumab effectively inhibits IL-6 signal transduction by its ability to bind both mIL-6R (preventing IL-6 binding to cell-surface mIL-6R/gp130 complex) and sIL-6R (preventing formation of IL-6/sIL-6R complex, which could bind to cell-surface gp130).

Although initial results suggest that tocilizumab can control SLE disease activity, an increased risk of infections may limit its ultimate clinical utility. In an open-label phase I dosage-escalation study (Table 1), 16 patients with mild to moderate disease activity (presence of chronic glomerulonephritis or extrarenal SLE Disease Activity Index - SELENA modification score of 3 to 10) received one of three doses of tocilizumab (2, 4, or 8 mg/kg) every other week for 12 weeks [24]. Disease activity showed improvement, with a decrease of ≥4 points in activity scores for eight of 15 evaluable patients. Additionally, arthritis improved in all seven patients who had arthritis at baseline, and completely resolved in four of them. Th ere were also significant decreases in the frequency of circulating plasma cells and in levels of anti-dsDNA antibodies. Tocilizumab also led to a similar decline in levels of C3 and the complement activation products, iC3b and C5b-9 (terminal activation complex). Tocilizumab treatment led to dose-related decreases in the absolute neutrophil count, however, with two patients experiencing severe neutropenia (absolute neutrophil count of 500 to 1,000/μl). The decline in neutrophil counts probably had clinical consequences - 16 infections developed in 11 patients, 10 of whom required systemic antibiotics or antivirals. Additional studies are clearly needed before tocilizumab or other IL-6 antagonists can be considered viable therapeutics in human SLE.

Table 1 Cytokine-targeting agents and their clinical trials in SLE

Tumor necrosis factor alpha

General biology of TNFα

TNFα is expressed as a 26 kDa transmembrane protein that can be cleaved by TNFα-converting enzyme to release a 17 kDa soluble protein [25]. Like IL-6, TNFα is produced by a variety of cells, including activated macrophages and monocytes as well as B cells, T cells, keratinocytes, renal cells, and fibroblasts.

Depending upon the conditions, TNFα can trigger either proinflammatory or anti-inflammatory pathways by engaging one or both of two distinct transmembrane receptors: the type I, p55 tumor necrosis factor receptor (TNFR1), and the type II, p75 TNFR (TNFR2) [26]. TNFR1 may trigger apoptotic and anti-inflammatory signals through Fas-associated death domain (FADD) and activation of the caspase cascade. Conversely, it may also trigger anti-apoptotic and proinflammatory responses through recruitment of TNF receptor-associated factor 2 (TRAF-2), which activates NF-κB, c-Jun NH2-terminal kinase (JNK), and mitogen-activated protein (MAP) kinase. Binding of TNFα to TNFR2 also leads to recruitment of TRAF2 and promotes proinflammatory responses. The TNF/TNFR system can thus serve dually both as a potent proinflammatory mediator and as a key immune modulator as a function of the immunological setting [25, 26].

TNFα in murine systemic lupus erythematosus

Several studies point to a pathogenic role for TNFα in murine SLE. TNFα expression is increased in the sera and kidneys of MRL/lpr mice and correlates with disease activity [27, 28]. Indeed, TNF antagonism/inhibition in such mice has clinical benefit. Furthermore, in C3H.SW mice with induced SLE, treatment with anti-TNFα mAb results in decreased development of anti-DNA antibodies, proteinuria, and glomerular immune-complex deposition [29, 30].

In sharp contrast, the administration of recombinant TNFα or TNFα-inducing agents to BWF mice is clearly beneficial [31–33]. Moreover, TNFα deficiency in other SLE mice has resulted in disease induction/exacerbation. Autoimmunity and fatal SLE nephritis develop in NZB mice when crossed with TNFα-deficient mice [34]. Similarly, TNFα deficiency in mice bearing a mixed B6/129 genetic background (H-2b/b) develop IgG2b and IgG3 anti-DNA antibodies, glomerular IgG deposition, and alterations in glomerular structure resembling SLE nephritis [35].

These often conflicting effects of TNFα in different murine SLE models underscore this cytokine's dual proinflammatory and immunoregulatory roles. These conflicting effects are also disease stage specific and may reflect the cell type expressing the receptors as well as differential expression of the receptors themselves by a given cell type. Although the underlying molecular and cellular mechanisms remain largely enigmatic, at least part of TNF's functional duality might segregate at the level of the two TNFα receptors [35, 36]. Indeed, in anti-GBM antibody-induced glomerulonephritis, TNFR2 deficiency is protective, whereas TNFR1-deficient mice develop proteinuria and renal pathology similar to those in wild-type controls [37]. In addition, TNFR1 deficiency greatly accelerates lymphoadenopathy, autoantibody production, and mortality in C57BL/6-lpr mice [38]. In NZM2328 mice singly deficient in one TNFR, however, signaling through the other receptor can compensate and permit persistence of the wild-type clinical and pathological phenotypes. Deletion of both receptors, with complete abrogation of TNFα signaling, results in significantly accelerated disease [39].

TNFα in human systemic lupus erythematosus

As in animal models, the evidence has been conflicting regarding the role of TNFα in human SLE. Some studies have shown that serum TNFα levels are elevated in SLE patients and correlate with disease activity [40, 41]. Other studies, however, have shown otherwise. One study demonstrated that elevated TNFα plasma levels do not correlate with SLE disease activity, and another study showed that TNFα levels were actually higher in patients with inactive disease, suggesting a protective role for TNFα in SLE [42, 43]. Differences in patient characteristics, assays for TNFα, and study designs probably each contribute to the somewhat inchoate picture that emerges, highlighting the pleiotropic and oftentimes unpredictable nature of TNFα.

In addition to systemic production of TNFα, local production of TNFα may contribute to SLE. Studies of local production of TNFα in the kidneys have documented upregulated TNFα protein and gene expression in renal biopsy tissue from 52% of SLE nephritis patients [22]. Local upregulated TNFα protein and gene expression within the resident glomerular, tubular, and mesangial cells has also been demonstrated [23].

The effects of systemically produced TNFα may be dissociated from the effects of locally produced TNFα. For example, DR3-positive or DR4-positive SLE patients, who genetically are high TNFα producers in response to appropriate induction, are not predisposed to nephritis. In contrast, DR2 and DQw1-positive SLE patients, who genetically are low TNFα producers following induction, have an increased incidence of SLE nephritis [44]. As another example, the TNFα adaptor proteins TNF receptor type 1-associated DEATH domain (TRADD), FADD, receptor interacting protein-1 (RIP-1), and TRAF-2 are downregulated in SLE peripheral blood mononuclear cells, and their decreased expression correlates with disease activity [45]. The downregulation of TRADD and FADD (which can recruit the caspase cascade) may thus promote an anti-apoptotic effect that leads to survival of autoreactive cells. Of note, TRADD, RIP-1, and TRAF-2 (which can activate the NF-κB pathway) are upregulated, rather than downregulated, in renal tissue from SLE nephritis patients, suggesting that TNFα may promote immunoregulatory functions systemically but may mediate inflammation locally [46].

Therapeutic blockade of TNFα in systemic lupus erythematosus

Use of TNFα antagonists is well established in the treatment of rheumatoid arthritis, juvenile idiopathic arthritis, psoriatic arthritis, chronic inflammatory bowel disease, and ankylosing spondylitis [47]. Consistent with the competing proinflammatory and anti-inflammatory effects of TNFα, use of TNFα antagonists in multiple sclerosis patients has led to immune activation and disease exacerbation [48]. Moreover, TNFα antagonists have triggered the development of autoantibodies, neuroinflammatory disease, or SLE-like features in some patients with rheumatoid arthritis or chronic inflammatory bowel disease [49–51]. Indeed, depending on the disease, two-thirds of previously antinuclear antibody (ANA)-negative patients became ANA-positive after initiation of TNFα-antagonist therapy. A recent report described six rheumatoid arthritis patients who developed active SLE associated with major organ involvement and life-threatening manifestations and required additional treatment beyond cessation of the TNFα-antagonist regimen [52].

Nevertheless, TNFα-antagonist therapy has been successful in some SLE patients. Six patients with moderately active SLE (three with nephritis, two with arthritis refractory to other therapies, and one with both joint and renal involvement) were given four infusions of 300 mg doses of infliximab, in combination with immunosuppression with azathioprine or methotrexate [53]. Patients with joint involvement experienced remission of arthritis, which relapsed 8 to 11 weeks after the last infliximab infusion. In the four patients with nephritis, proteinuria decreased by >60% within 8 weeks. Of note, anti-dsDNA and anti-cardiolipin antibodies increased in four patients. A number of long-term studies also revealed modest clinical benefit, but there were several adverse events, including deep-vein thrombosis, infections, central nervous system lymphoma, and Legionella pneumonia [54, 55].

The need to evaluate safety and efficacy of TNFα blockade in SLE prompted two large randomized phase II and phase III trials with etanercept and infliximab (Table 1), but both studies (NCT00447265 and NCT00368264) were terminated prematurely (although this may have had more to do with corporate business matters than with medical matters). As with neutralization of IL-6, additional studies with TNFα antagonists will be needed before use of such agents becomes a routinely viable option in human SLE.

Type I interferons

General biology of type I interferon

Type I interferon represent a large family of cytokines that includes many IFNα subtypes, a single IFNβ, and multiple IFNω subtypes that exert their expansive biological properties through engagement of a common heterodimeric receptor (type I interferon receptor (IFNAR)) composed of IFNAR1 and IFNAR2 subunits. These broad effects include activating dendritic cells; promoting proliferation, survival, and differentiation of monocytes into antigen-presenting cells and B cells into plasma cells; stimulating the Th1 pathway and preventing apoptosis of activated cytotoxic T cells; and suppressing regulatory T cells, enhancing natural killer cell activity, and modulating each of these cells' respective cytokine production and signaling responses.

Within the type I interferon family, the IFNα subtypes are arguably the biologically most important in general, and this certainly is the case for SLE. The major producers of IFNα are plasmacytoid dendritic cells, although virtually any cell type can elaborate interferon [56]. IFNα is produced both in response to exogenous stimuli, such as bacterial and viral pathogens, and to endogenous stimuli, such as self-nucleic acids and nucleic acid-containing immune complexes, via Toll-like receptor (TLR)-dependent and TLR-independent path-ways. Among the TLR-dependent pathways, engagement of TLR7 and TLR8 by single-strand RNA-containing complexes and engagement of TLR9 by dsDNA-containing complexes may be especially relevant to SLE pathogenesis [56].

Type I interferon in murine lupus

Numerous studies have collectively demonstrated the complexity of the type I interferon/IFNAR axis in the pathogenesis of murine SLE. In several strains of mice with SLE-related autoimmune phenotypes, type I interferon/IFNAR signaling promotes autoantibody production and development of renal disease. Indeed, genetic deletion of the Ifnar1 gene prevents the development of severe clinical and pathological disease in SLE-prone NZB and NZM2328 mice [57, 58]. Similarly, up-regulation or administration of IFNα to BWF mice, (NZWxBXSB)F1 mice, B6.Sle123 mice, or NZM2328 mice markedly accelerates nephritis and death [59–61], and in BWF mice is associated with unabated expansion of short-lived plasma cells [62]. Nevertheless, type I interferon/IFNAR signaling is not always deleterious. IFNAR-deficient MRL/lpr mice develop more severe disease than do their IFNAR-sufficient counterparts [63], an observation that may relate to the ability of IFNβ to ameliorate disease manifestations in MRL/lpr mice. Importantly, these counterintuitive effects of type I interferon are not limited to MRL/lpr mice, in that blockade of type I interferon in B57BL/6 or B6.Sle2 mice led to increased, rather than decreased, levels of IgG autoantibodies [64].

In addition to studies that directly manipulated type I interferon and/or IFNAR expression, studies focused on TLR (especially TLR7 and TLR9) have also pointed to a vital role for IFNα in murine SLE. Interestingly, TLR7/9 doubly-deficient MRL/lpr mice display significantly reduced ANA titers, proteinuria, and kidney disease [65]. In agreement with these results, treatment of BWF mice with a dual inhibitor of TLR7 and TLR9 demonstrated significant reductions in interferon produced by plasmacytoid dendritic cells; decreased proteinuria; reduced glomerulonephritis, serum autoantibody levels, and end-organ damage; and increased survival [66]. Indeed, administration of bacterial or viral TLR ligands to SLE-prone mice leads to increased IFNα production and disease exacerbation, whereas disease is not exacerbated in similarly treated IFNAR-deficient SLE-prone mice [67].

Type I interferon and human systemic lupus erythematosus

Serum type I interferon activity is increased in SLE patients and their first-degree relatives, and ages of peak type I interferon are associated with ages of peak SLE incidence [68, 69]. Type I interferon levels have correlated with disease activity in a number of cross-sectional studies, although longitudinal correlations could not be established in other studies [70–72]. The use of elevated serum levels of type I interferon-regulated chemokines as biomarkers for disease activity was suggested in early studies [73] and was later replicated and validated by associations between elevated transcript levels of these chemokines with disease activity and organ damage [74, 75]. By relying on expression of type I interferon-inducible genes rather than on circulating type I interferon protein levels (which are often barely detectable or not detectable), most SLE patients have been demonstrated to unequivocally express type I interferon-inducible genes in peripheral blood mononuclear cells. This so-called interferon signature has been convincingly correlated with active disease, renal manifestations, and increased damage index [70, 76, 77]. Furthermore, the interferon gene signature has been found in glomerular and synovial tissue, suggesting local organ involvement of type I interferon [78, 79]. Indeed, plasmacytoid dendritic cells - the main producers of IFNα - accumulate in the glomeruli of SLE nephritis patients and can also be found in cutaneous lesions where they promote continuous IFNα release [80, 81].

In terms of potential interferon contribution to lymphocyte autoreactivity in SLE, interferon not only promotes B-cell activation, antibody production, and class switching, but can also decrease B-cell selectivity for CpG-rich DNA, thereby activating TLR9, and can also allow stimulation of B cells by non-CpG DNA [82, 83]. Moreover, interferon can potentially promote survival of autoreactive B cells, since it can prevent B-cell apoptosis and enhance proliferation even in the absence of mitogenic stimuli [84].

T-cell function is also modulated by interferon in SLE. Ample literature has demonstrated dysfunction of regulatory T cells in SLE, with SLE regulatory T cells being inefficient suppressors of inflammation and T-cell proliferation [85, 86]. Part of this dysfunction appears to be due to interferon production by antigen-presenting cells [87].

The ex vivo and in vitro findings that suggest a role for type I interferon in SLE pathogenesis are supported by the development of SLE or SLE-like syndromes (malar rash, oral ulcers, photosensitivity, renal involvement, and anti-Sm and anti-dsDNA antibodies) following administration of IFNα [88, 89]. Occasionally, IFNα-induced SLE has led to life-threatening multiorgan involvement [90].

Recent investigations into the genetics of SLE have also revealed a strong connection between the type I interferon pathway and SLE. The gene coding for the transcription factor IRF5 was the first gene involved in IFNα production to be associated with SLE susceptibility [91]. Further evidence of genetic association between SLE and IRF5 and for interaction between IRF5 and TYK2, a Janus kinase involved in cytokine signaling that binds to IFNAR, has also been demonstrated [92]. Moreover, IRF5 risk loci are also associated with high serum type I interferon activity and the development of autoantibodies to dsDNA and RNA-binding proteins. [93]. Subsequently, the related IRF7 has also been associated with SLE [94]. Furthermore, the STAT4 gene, which codes for a protein that interacts with the cytoplasmic portion of IFNAR, is strongly associated with SLE. STAT4 risk loci increase sensitivity to IFNα, are associated with a more severe phenotype that includes anti-dsDNA antibodies and renal involvement, and interact with IRF5 susceptibility loci [95–97]. Another SLE susceptibility gene codes for IRAK1, which is involved in TLR-triggered signal transduction [98].

IFNα, independent of its effects on the immune system, may be especially important in promoting atherosclerotic disease in SLE. The dysfunction of circulating endothelial progenitor cells and myelomonocytic circulating angiogenic cells in SLE appears to be mediated by IFNα, in as much as neutralization of IFNα restores a normal endothelial progenitor cell/circulating angiogenic cell phenotype [99]. Indeed, SLE patients with robust interferon signatures have decreased endothelial function [100], and interferon signatures correlate with carotid intima media thickness [101].

Therapeutic blockade of type I interferon in systemic lupus erythematosus

The fact that type I interferon exacerbates SLE in some mouse strains but ameliorates SLE in others [59–61, 63] raises the possibility that markedly divergent clinical responses to type I interferon antagonists might emerge among individual SLE patients. Experience to date with antagonists against type I interferon in SLE is very limited (Table 1). Treatment of SLE patients (n = 62) in a phase I trial with anti-IFNα mAb sifalimumab (MEDI-545) in single escalating intravenous doses of 0.3, 1.0, 3.0, 10.0, or 30.0 mg/kg led to dose-dependent reversal of the interferon signature in both the blood and skin, downregulation of several cytokines, and reduced disease activity without any increase in serious infections [102]. Results from a phase I trial with the anti-IFNα mAb rontalizumab (rhuMab IFN-α) in SLE patients (n = 32) replicated this dose-dependent reduction in the interferon signature [103]. Two phase II trials are currently ongoing to evaluate the effects of anti-IFNα in SLE (NCT01031836 and NCT00657189), but as yet it will be some time before the utility of type I interferon antagonists in SLE is known.

B-lymphocyte stimulator

General biology of BLyS

BLyS (also known as BAFF) is a cytokine that is essential for survival of most B cells beyond the transitional 1 stage [104]. BLyS binds to three receptors: BCMA, TACI, and BR3 (also known as BAFFR). Two of these receptors (BCMA and TACI), but not the third (BR3), also bind APRIL, a cytokine closely related to BLyS that shares some, but not all, of its biological properties. Whereas survival of plasma cells, which express TACI and BCMA, is supported by either BLyS or APRIL, the survival of preplasmablast mature B cells, which express much BR3 but little TACI and essentially no BCMA, is supported only by BLyS. Of note, memory B cells appear to be independent of both BLyS and APRIL.

In addition to affecting B-cell survival/function, BLyS can affect other cell types that express BLyS receptors. Specifically, BR3 is expressed on T cells, although its role in T-cell signaling/stimulation is controversial [104]. Moreover, dendritic cells also express BLyS receptors, and BLyS-stimulated dendritic cells upregulate co-stimulatory molecules and produce inflammatory cytokines and chemokines such as IL-1, IL-6, CCL2, and CCL5.

BLyS in murine systemic lupus erythematosus

There is an irrefutable link between BLyS and murine SLE. Constitutive overexpression of BLyS in BLyS-transgenic nonautoimmune-prone mice leads to SLE-like features, including elevated levels of multiple autoantibodies (including anti-dsDNA), circulating immune complexes, and glomerular immunoglobulin deposition [105]. Moreover, BLyS overexpression accelerates the development of SLE-like features in mice that bear an autoimmune diathesis but otherwise do not develop overt SLE [106]. On the other hand, SLE-prone NZM2328 mice genetically deficient in BLyS are largely spared from overt disease (severe proteinuria and premature death), although the lifelong absence of BLyS does not protect them from ultimately developing serological autoimmunity and renal pathology [107]. Most importantly, treatment of BWF mice, (NZMxBXSB)F1 mice, MRL/lpr mice, or NZM2410 mice with either TACI-Ig (which neutralizes both BLyS and APRIL) or BR3-Ig (which selectively neutralizes BLyS) is effective at preventing clinical disease and ameliorating renal injury [108]. Intriguingly, IFNα-driven exaggerated disease in several SLE-prone mouse strains is associated with increases in serum BLyS levels [59–61], and IFNα-driven exaggerated disease is completely blocked in BLyS-deficient NZM2328 mice [109], indicating that BLyS is a vital contributor to the IFNα-driven pathogenic pathway in SLE.

BLyS in human systemic lupus erythematosus

Not only is BLyS associated with murine SLE, but is also associated with human SLE. Circulating BLyS levels are elevated in as many as 50% of SLE patients [110], and disease activity correlates with blood leukocyte expression of BLyS mRNA [111]. Aberrant expression of BLyS on multiple immune cells has been observed in SLE. Although BLyS is largely expressed in myeloid lineage cells, activated B cells can also express both BLyS and APRIL. In SLE patients, B cells and plasma cells express high levels of BLyS and APRIL mRNA, which correlate with disease activity and levels of anti-dsDNA antibodies [112].

In addition to the biologic studies summarized above, investigations into genetic susceptibility loci in SLE have revealed associations between polymorphisms in BLyS and APRIL genes with human SLE [113, 114]. In light of the results from murine SLE models, from ex vivo studies of human SLE leukocytes, from BLyS serum analyses, and from the genetics of SLE, BLyS has emerged as a highly attractive cytokine target in SLE.

Therapeutic blockade of BLyS in human systemic lupus erythematosus

The greatest experience to date with BLyS antagonists (Table 1) has accrued with belimumab, a fully human IgG1λ mAb that binds and neutralizes soluble BLyS [115]. Belimumab was shown to be safe in a randomized, double-blind, placebo-controlled phase I trial of SLE, in which the prevalence of adverse events was no different between belimumab-treated and placebo-treated patients [116]. Of note, only modest reductions in peripheral blood B cells were observed among belimumab-treated patients. No clinical efficacy was demonstrated in this phase I trial, but the small number of patients (n = 70) and the very brief treatment schedules (single infusion or two infusions 3 weeks apart) and follow-up period (12 weeks after final infusion) precluded demonstration of clinical benefit.

Disappointingly, the subsequent phase II trial (n = 449) failed to meet its co-primary endpoints [117]. Extensive post hoc analysis, however, led to a novel composite index of clinical response (SLE responder index) [118] and demonstrated significantly increased clinical response among belimumab-treated patients at 52 weeks among the patients who were seropositive (ANA titer ≥1:80 and/or positive for anti-dsDNA antibodies) at entry. Using this novel SLE responder index, two separate large randomized, double-blind, placebo-controlled phase III trials (n = 865 and n = 819, respectively) of belimumab in seropositive SLE patients each met their primary endpoints (increased percentage of responders at 52 weeks) [119, 120]. Importantly, analysis of the combined 1,864 SLE patients in both trials pointed to reductions in disease activity and prevention of worsening across vital internal organ systems [121]. Although questions remain regarding the durability of the clinical response, a US Food and Drug Administration advisory panel in November 2010 recommended approval for belimumab in the treatment of SLE, and final approval by the US Food and Drug Administration was given on 9 March 2011.

Concern has been raised regarding the ostensibly lower clinical efficacy of belimumab in human SLE in comparison with the ostensibly more robust clinical efficacy of BLyS antagonists in murine SLE. This difference in clinical response between human and murine SLE may be more apparent than real. First, the clinical response in the phase III human trials was based on a composite of several instruments each rooted in multiple organ systems [118]. In contrast, clinical response in the murine trials was simply the absence of pre-moribund proteinuria and death. (Belimumab in all likelihood will prove to be very effective in preventing pre-moribund proteinuria and death; however, it will take many years of follow-up to formally prove this point.) Second, human SLE patients received standard-of-care therapy in addition to belimumab in the human clinical trials, whereas murine SLE patients in the murine clinical trials received no treatment other than a BLyS antagonist. (Human SLE patients treated with belimumab alone would undoubtedly do much better than untreated patients, but such a human trial would be entirely unethical.)

In addition to belimumab, several other BLyS antagonists are undergoing clinical evaluation in SLE. The one furthest advanced in clinical evaluation is atacicept, a fusion protein between one of the BLyS receptors (TACI) and the Fc portion of IgG. Favorable safety and tolerability were demonstrated in a randomized, double-blind, placebo-controlled phase I trial (n = 49) [122]. Dose-dependent reductions in peripheral blood B cells and in circulating immunoglobulin levels were noted, but clinical efficacy could not be demonstrated due to the limited treatment and limited follow-up period. Of concern, an increased risk of severe infections was observed in a subsequent trial involving patients with SLE nephritis who were concurrently taking mycophenolate mofetil and corticosteroids (NCT00573157). As a consequence, this trial was prematurely terminated. Nevertheless, a separate phase II/III trial of atacicept in SLE has recently been initiated (NCT00624338). Whether atacicept achieves clinical success from efficacy and safety standpoints remains to be determined.

A third BLyS antagonist being tested in clinical trials is A-623 (previously known as AMG 623), a fusion between the Fc portion of IgG and a peptide sequence selected for its ability to bind with high affinity to BLyS. In a double-blind, placebo-controlled phase I trial, SLE patients received a single dose (n = 54) or four weekly doses (n = 63) of escalating doses of AMG 623 or matching placebo [123]. A dose-independent decrease in naive and total peripheral blood B cells was accompanied by an increase in memory B cells - an observation that has now also been made in patients treated with atacicept or belimumab [122, 124]. Clinical responses were not reported, so the relevance of the disparate changes among B-cell subsets to clinical parameters remains unknown. A phase II trial of A-623 in SLE had been initiated but was suspended due to 'structural failure identified in some product vials', but the trial was recently resumed (NCT01162681).

A fourth BLyS antagonist in clinical development for SLE is LY2127399, a mAb that binds both soluble and membrane BLyS [125]. Two phase III trials in SLE are planned (NCT01205438 and NCT01196091), but neither has yet started recruiting patients. It remains to be determined whether neutralization of soluble plus membrane BLyS (as with LY2127399) will have greater therapeutic efficacy than neutralization of soluble BLyS alone (as with belimumab).

Concluding remarks

Cytokine-targeted therapy may prove effective in the treatment of SLE and offer less toxic options in comparison with 'conventional' therapies such as glucocorticoids or cytotoxics. In addition to the four cytokines reviewed above, there are many more cytokines - including IFNγ, IL-1, and IL-17 - that have been implicated in SLE. As has become painfully evident, cytokine biology is extremely complex - and increasingly so in a complex disease such as SLE. Continued investigations of cytokine pathways in vivo in animal models, ex vivo in human SLE tissues, and in the genetics of SLE will reveal the roles of additional cytokines in disease pathogenesis and should offer additional novel targets for treatment.

Note

Autoimmune Basis of Rheumatic Diseases

This article is part of a series on Systemic lupus erythematosus, edited by David Pisetsky, which can be found online at http://arthritis-research.com/series/lupus

This series forms part of a special collection of reviews covering major autoimmune rheumatic diseases, available at: http://arthritis-research.com/series/abrd

Abbreviations

ANA:

antinuclear antibody

APRIL:

a proliferation-inducing ligand

BAFF:

B-cell activating factor of the TNF family

BCMA:

B-cell maturation antigen

BR3:

BLyS receptor 3

BLyS:

B-lymphocyte stimulator

BWF:

(NZB × NZW)F1

dsDNA:

double-strand DNA

FADD:

Fas-associated death domain

ICAM-1:

intercellular adhesion molecule-1

IFN:

interferon

IFNAR:

type I interferon receptor

IL:

interleukin

IL-6R:

IL-6 receptor

mAb:

monoclonal antibody

MHC:

major histocompatibility complex

NF:

nuclear factor

RIP-1:

receptor interacting protein-1

SLE:

systemic lupus erythematosus

TACI:

transmembrane activator and calcium modulator and cyclophilin ligand interactor

Th:

T-helper type

TLR:

Toll-like receptor

TNF:

tumor necrosis factor

TNFR:

tumor necrosis factor receptor

TRAF:

TNF receptor-associated factor

TRADD:

TNF receptor type 1-associated DEATH domain

VCAM-1:

vascular cell adhesion molecule-1.

References

  1. Naka T, Nishimoto N, Kishimoto T: The paradigm of IL-6: from basic science to medicine. Arthritis Res. 2002, 4 (Suppl 3): S233-S242. 10.1186/ar565.

    PubMed Central  PubMed  Google Scholar 

  2. Cash H, Relle M, Menke J, Brochhausen C, Jones SA, Topley N, Galle PR, Schwarting A: Interleukin 6 (IL-6) deficiency delays lupus nephritis in MRL-Faslpr mice: the IL-6 pathway as a new therapeutic target in treatment of autoimmune kidney disease in systemic lupus erythematosus. J Rheumatol. 2010, 37: 60-70. 10.3899/jrheum.090194.

    CAS  PubMed  Google Scholar 

  3. Schiffer L, Bethunaickan R, Ramanujam M, Huang W, Schiffer M, Tao H, Madaio MP, Bottinger EP, Davidson A: Activated renal macrophages are markers of disease onset and disease remission in lupus nephritis. J Immunol. 2008, 180: 1938-1947.

    PubMed Central  CAS  PubMed  Google Scholar 

  4. Alarcón-Riquelme ME, Möller G, Fernández C: Age-dependent responsiveness to interleukin-6 in B lymphocytes from a systemic lupus erythematosus-prone (NZB × NZW)F1 hybrid. Clin Immunol Immunopathol. 1992, 62: 264-269. 10.1016/0090-1229(92)90101-S.

    PubMed  Google Scholar 

  5. Mihara M, Fukui H, Koishihara Y, Saito M, Ohsugi Y: Immunologic abnormality in NZB/W F1 mice. Thymus-independent expansion of B cells responding to interleukin-6. Clin Exp Immunol. 1990, 82: 533-537.

    PubMed Central  CAS  PubMed  Google Scholar 

  6. Alarcón-Riquelme ME, Möller G, Fernández C: Macrophage depletion decreases IgG anti-DNA in cultures from (NZB × NZW)F1 spleen cells by eliminating the main source of IL-6. Clin Exp Immunol. 1993, 91: 220-225.

    PubMed Central  PubMed  Google Scholar 

  7. Ryffel B, Car BD, Gunn H, Roman D, Hiestand P, Mihatsch MJ: Interleukin-6 exacerbates glomerulonephritis in (NZB × NZW)F1 mice. Am J Pathol. 1994, 144: 927-937.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Finck BK, Chan B, Wofsy D: Interleukin 6 promotes murine lupus in NZB/NZW F1 mice. J Clin Invest. 1994, 94: 585-591. 10.1172/JCI117373.

    PubMed Central  CAS  PubMed  Google Scholar 

  9. Mihara M, Takagi N, Takeda Y, Ohsugi Y: IL-6 receptor blockage inhibits the onset of autoimmune kidney disease in NZB/W F1 mice. Clin Exp Immunol. 1998, 112: 397-402. 10.1046/j.1365-2249.1998.00612.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  10. Richards HB, Satoh M, Shaw M, Libert C, Poli V, Reeves WH: Interleukin 6 dependence of anti-DNA antibody production: evidence for two pathways of autoantibody formation in pristane-induced lupus. J Exp Med. 1998, 188: 985-990. 10.1084/jem.188.5.985.

    PubMed Central  CAS  PubMed  Google Scholar 

  11. Pflegerl P, Vesely P, Hantusch B, Schlederer M, Zenz R, Janig E, Steiner G, Meixner A, Petzelbauer P, Wolf P, Soleiman A, Egger G, Moriggl R, Kishimoto T, Wagner EF, Kenner L: Epidermal loss of JunB leads to a SLE phenotype due to hyper IL-6 signaling. Proc Natl Acad Sci USA. 2009, 106: 20423-20428. 10.1073/pnas.0910371106.

    PubMed Central  CAS  PubMed  Google Scholar 

  12. Gröndal G, Gunnarsson I, Rönnelid J, Rogberg S, Klareskog L, Lundberg I: Cytokine production, serum levels and disease activity in systemic lupus erythematosus. Clin Exp Rheumatol. 2000, 18: 565-570.

    PubMed  Google Scholar 

  13. Linker-Israeli M, Deans RJ, Wallace DJ, Prehn J, Ozeri-Chen T, Klinenberg JR: Elevated levels of endogenous IL-6 in systemic lupus erythematosus. A putative role in pathogenesis. J Immunol. 1991, 147: 117-123.

    CAS  PubMed  Google Scholar 

  14. Peterson E, Robertson AD, Emlen W: Serum and urinary interleukin-6 in systemic lupus erythematosus. Lupus. 1996, 5: 571-575. 10.1177/096120339600500603.

    CAS  PubMed  Google Scholar 

  15. Hagiwara E, Gourley MF, Lee S, Klinman DK: Disease severity in patients with systemic lupus erythematosus correlates with an increased ratio of interleukin-10:interferon-gamma-secreting cells in the peripheral blood. Arthritis Rheum. 1996, 39: 379-385. 10.1002/art.1780390305.

    CAS  PubMed  Google Scholar 

  16. Esposito P, Balletta MM, Procino A, Postiglione L, Memoli B: Interleukin-6 release from peripheral mononuclear cells is associated to disease activity and treatment response in patients with lupus nephritis. Lupus. 2009, 18: 1329-1330. 10.1177/0961203309106183.

    PubMed  Google Scholar 

  17. Kitani A, Hara M, Hirose T, Norioka K, Harigai M, Hirose W, Suzuki K, Kawakami M, Kawagoe M, Nakamura H: Heterogeneity of B cell responsiveness to interleukin 4, interleukin 6 and low molecular weight B cell growth factor in discrete stages of B cell activation in patients with systemic lupus erythematosus. Clin Exp Immunol. 1989, 77: 31-36.

    PubMed Central  CAS  PubMed  Google Scholar 

  18. Nagafuchi H, Suzuki N, Mizushima Y, Sakane T: Constitutive expression of IL-6 receptors and their role in the excessive B cell function in patients with systemic lupus erythematosus. J Immunol. 1993, 151: 6525-6534.

    CAS  PubMed  Google Scholar 

  19. Takeno M, Nagafuchi H, Kaneko S, Wakisaka S, Oneda K, Takeba Y, Yamashita N, Suzuki N, Kaneoka H, Sakane T: Autoreactive T cell clones from patients with systemic lupus erythematosus support polyclonal autoantibody production. J Immunol. 1997, 15: 3529-3538.

    Google Scholar 

  20. Hirohata S, Kanai Y, Mitsuo A, Tokano Y, Hashimoto H, NPSLE Research Subcommittee: Accuracy of cerebrospinal fluid IL-6 testing for diagnosis of lupus psychosis. A multicenter retrospective study. Clin Rheumatol. 2009, 28: 1319-1323. 10.1007/s10067-009-1226-8.

    PubMed  Google Scholar 

  21. Iwano M, Dohi K, Hirata E, Kurumatani N, Horii Y, Shiiki H, Fukatsu A, Matsuda T, Hirano T, Kishimoto T: Urinary levels of IL-6 in patients with active lupus nephritis. Clin Nephrol. 1993, 40: 16-21.

    CAS  PubMed  Google Scholar 

  22. Herrera-Esparza R, Barbosa-Cisneros O, Villalobos-Hurtado R, Avalos-Díaz E: Renal expression of IL-6 and TNFα genes in lupus nephritis. Lupus. 1998, 7: 154-158. 10.1191/096120398678919949.

    CAS  PubMed  Google Scholar 

  23. Malide D, Russo P, Bendayan M: Presence of tumor necrosis factor alpha and interleukin-6 in renal mesangial cells of lupus nephritis patients. Hum Pathol. 1995, 26: 558-564. 10.1016/0046-8177(95)90253-8.

    CAS  PubMed  Google Scholar 

  24. Illei GG, Shirota Y, Yarboro CH, Daruwalla J, Tackey E, Takada K, Fleisher T, Balow JE, Lipsky PE: Tocilizumab in systemic lupus erythematosus: data on safety, preliminary efficacy, and impact on circulating plasma cells from an open-label phase I dosage-escalation study. Arthritis Rheum. 2010, 62: 542-552. 10.1002/art.27221.

    PubMed Central  CAS  PubMed  Google Scholar 

  25. MacEwan DJ: TNF ligands and receptors - a matter of life and death. Br J Pharmacol. 2002, 135: 855-875. 10.1038/sj.bjp.0704549.

    PubMed Central  CAS  PubMed  Google Scholar 

  26. McDevitt H, Munson S, Ettinger R, Wu A: Multiple roles for tumor necrosis factor-alpha and lymphotoxin alpha/beta in immunity and autoimmunity. Arthritis Res. 2002, 4 (Suppl 3): S141-S152. 10.1186/ar570.

    PubMed Central  PubMed  Google Scholar 

  27. Boswell JM, Yui MA, Burt DW, Kelley VE: Increased tumor necrosis factor and IL-1 beta gene expression in the kidneys of mice with lupus nephritis. J Immunol. 1988, 141: 3050-3054.

    CAS  PubMed  Google Scholar 

  28. Yokoyama H, Kreft B, Kelley VR: Biphasic increase in circulating and renal TNF-α in MRL-lpr mice with differing regulatory mechanisms. Kidney Int. 1995, 47: 122-130. 10.1038/ki.1995.14.

    CAS  PubMed  Google Scholar 

  29. Edwards CK, Zhou T, Zhang J, Baker TJ, De M, Long RE, Borcherding DR, Bowlin TL, Bluethmann H, Mountz JD: Inhibition of superantigen-induced proinflammatory cytokine production and inflammatory arthritis in MRL-lpr/lpr mice by a transcriptional inhibitor of TNF-α. J Immunol. 1996, 157: 1758-1772.

    CAS  PubMed  Google Scholar 

  30. Segal R, Dayan M, Zinger H, Mozes E: Suppression of experimental systemic lupus erythematosus (SLE) in mice via TNF inhibition by an anti-TNFα monoclonal antibody and by pentoxiphylline. Lupus. 2001, 10: 23-31. 10.1191/096120301675275538.

    CAS  PubMed  Google Scholar 

  31. Jacob CO, McDevitt HO: Tumour necrosis factor-alpha in murine autoimmune 'lupus' nephritis. Nature. 1988, 331: 356-358. 10.1038/331356a0.

    CAS  PubMed  Google Scholar 

  32. Gordon C, Ranges GE, Greenspan JS, Wofsy D: Chronic therapy with recombinant tumor necrosis factor-alpha in autoimmune NZB/NZW F1 mice. Clin Immunol Immunopathol. 1989, 52: 421-434. 10.1016/0090-1229(89)90157-8.

    CAS  PubMed  Google Scholar 

  33. Gordon C, Wofsy D: Effects of recombinant murine tumor necrosis factor-alpha on immune function. J Immunol. 1990, 144: 1753-1758.

    CAS  PubMed  Google Scholar 

  34. Kontoyiannis D, Kollias G: Accelerated autoimmunity and lupus nephritis in NZB mice with an engineered heterozygous deficiency in tumor necrosis factor. Eur J Immunol. 2000, 30: 2038-2047. 10.1002/1521-4141(200007)30:7<2038::AID-IMMU2038>3.0.CO;2-K.

    CAS  PubMed  Google Scholar 

  35. Kollias G, Kontoyianni D: Role of TNF/TNFR in autoimmunity: specific TNF receptor blockade may be advantageous to anti-TNF treatments. Cytokine Growth Factor Rev. 2002, 13: 315-321. 10.1016/S1359-6101(02)00019-9.

    CAS  PubMed  Google Scholar 

  36. Vielhauer V, Mayadas TN: Functions of TNF and its receptors in renal disease: distinct roles in inflammatory tissue injury and immune regulation. Semin Nephrol. 2007, 27: 286-308. 10.1016/j.semnephrol.2007.02.004.

    CAS  PubMed  Google Scholar 

  37. Vielhauer V, Stavrakis G, Mayadas TN: Renal cell-expressed TNF receptor 2, not receptor 1, is essential for the development of glomerulonephritis. J Clin Invest. 2005, 115: 1199-1209.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. Zhou T, Edwards CK, Yang P, Wang Z, Bluethmann H, Mountz JD: Greatly accelerated lymphadenopathy and autoimmune disease in lpr mice lacking tumor necrosis factor receptor I. J Immunol. 1996, 156: 2661-2665.

    CAS  PubMed  Google Scholar 

  39. Jacob N, Yang H, Pricop L, Liu Y, Gao X, Zheng SG, Wang J, Gao HX, Putterman C, Koss MN, Stohl W, Jacob CO: Accelerated pathological and clinical nephritis in systemic lupus erythematosus-prone New Zealand Mixed 2328 mice doubly deficient in TNF receptor 1 and TNF receptor 2 via a Th17-associated pathway. J Immunol. 2009, 182: 2532-2541. 10.4049/jimmunol.0802948.

    PubMed Central  CAS  PubMed  Google Scholar 

  40. Studnicka-Benke A, Steiner G, Petera P, Smolen JS: Tumour necrosis factor alpha and its soluble receptors parallel clinical disease and autoimmune activity in systemic lupus erythematosus. Br J Rheumatol. 1996, 35: 1067-1074. 10.1093/rheumatology/35.11.1067.

    CAS  PubMed  Google Scholar 

  41. Gabay C, Cakir N, Moral F, Roux-Lombard P, Meyer O, Dayer JM, Vischer T, Yazici H, Guerne PA: Circulating levels of tumor necrosis factor soluble receptors in systemic lupus erythematosus are significantly higher than in other rheumatic diseases and correlate with disease activity. J Rheumatol. 1997, 24: 303-308.

    CAS  PubMed  Google Scholar 

  42. Zhu LJ, Landolt-Marticorena C, Li T, Yang X, Yu XQ, Gladman DD, Urowitz MB, Fortin PR, Wither JE: Altered expression of TNF-α signaling pathway proteins in systemic lupus erythematosus. J Rheumatol. 2010, 37: 1658-1666. 10.3899/jrheum.091123.

    CAS  PubMed  Google Scholar 

  43. Gómez D, Correa PA, Gómez LM, Cadena J, Molina JF, Anaya JM: Th1/Th2 cytokines in patients with systemic lupus erythematosus: is tumor necrosis factor alpha protective?. Semin Arthritis Rheum. 2004, 33: 404-413. 10.1016/j.semarthrit.2003.11.002.

    PubMed  Google Scholar 

  44. Jacob CO, Fronek Z, Lewis GD, Koo M, Hansen JA, McDevitt HO: Heritable major histocompatibility complex class II-associated differences in production of tumor necrosis factor alpha: relevance to genetic predisposition to systemic lupus erythematosus. Proc Natl Acad Sci USA. 1990, 87: 1233-1237. 10.1073/pnas.87.3.1233.

    PubMed Central  CAS  PubMed  Google Scholar 

  45. Zhu L, Yang X, Chen W, Li X, Ji Y, Mao H, Nie J, Yu X: Decreased expressions of the TNF-α signaling adapters in peripheral blood mononuclear cells (PBMCs) are correlated with disease activity in patients with systemic lupus erythematosus. Clin Rheumatol. 2007, 26: 1481-1489. 10.1007/s10067-006-0531-8.

    PubMed  Google Scholar 

  46. Zhu L, Yang X, Ji Y, Chen W, Guan W, Zhou SF, Yu X: Up-regulated renal expression of TNF-α signaling adapter proteins in lupus glomerulonephritis. Lupus. 2009, 18: 116-127. 10.1177/0961203308094764.

    CAS  PubMed  Google Scholar 

  47. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen JS, Leeb B, Breedveld FC, Macfarlane JD, Bijl H: Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet. 1994, 344: 1105-1110. 10.1016/S0140-6736(94)90628-9.

    CAS  PubMed  Google Scholar 

  48. Lenercept Multiple Sclerosis Study Group; University of British Columbia MS/MRI Analysis Group: TNF neutralization in MS: results of a randomized, placebo-controlled multicenter study. Neurology. 1999, 53: 457-465.

    Google Scholar 

  49. Feldmann M, Brennan FM, Maini RN: Role of cytokines in rheumatoid arthritis. Annu Rev Immunol. 1996, 14: 397-440. 10.1146/annurev.immunol.14.1.397.

    CAS  PubMed  Google Scholar 

  50. Sicotte NL, Voskuhl RR: Onset of multiple sclerosis associated with anti-TNF therapy. Neurology. 2001, 57: 1885-1888.

    CAS  PubMed  Google Scholar 

  51. Sandborn WJ, Hanauer SB: Antitumor necrosis factor therapy for inflammatory bowel disease: a review of agents, pharmacology, clinical results, and safety. Inflamm Bowel Dis. 1999, 5: 119-133.

    CAS  PubMed  Google Scholar 

  52. Soforo E, Baumgartner M, Francis L, Allam F, Phillips PE, Perl A: Induction of systemic lupus erythematosus with tumor necrosis factor blockers. J Rheumatol. 2010, 37: 204-205. 10.3899/jrheum.081312.

    CAS  PubMed  Google Scholar 

  53. Aringer M, Graninger WB, Steiner G, Smolen JS: Safety and efficacy of tumor necrosis factor alpha blockade in systemic lupus erythematosus: an openlabel study. Arthritis Rheum. 2004, 50: 3161-3169. 10.1002/art.20576.

    CAS  PubMed  Google Scholar 

  54. Matsumura R, Umemiya K, Sugiyama T, Sueishi M, Umibe T, Ichikawa K, Yoshimura M: Anti-tumor necrosis factor therapy in patients with difficult-to-treat lupus nephritis: a prospective series of nine patients. Clin Exp Rheumatol. 2009, 27: 416-421.

    CAS  PubMed  Google Scholar 

  55. Aringer M, Houssiau F, Gordon C, Graninger WB, Voll RE, Rath E, Steiner G, Smolen JS: Adverse events and efficacy of TNF-α blockade with infliximab in patients with systemic lupus erythematosus: long-term follow-up of 13 patients. Rheumatology (Oxford). 2009, 48: 1451-1454. 10.1093/rheumatology/kep270.

    CAS  Google Scholar 

  56. Baccala R, Hoebe K, Kono DH, Beutler B, Theofilopoulos AN: TLR-dependent and TLR-independent pathways of type I interferon induction in systemic autoimmunity. Nat Med. 2007, 13: 543-551. 10.1038/nm1590.

    CAS  PubMed  Google Scholar 

  57. Santiago-Raber M-L, Baccala R, Haraldsson KM, Choubey D, Stewart TA, Kono DH, Theofilopoulos AN: Type-I interferon receptor deficiency reduces lupus-like disease in NZB mice. J Exp Med. 2003, 197: 777-788. 10.1084/jem.20021996.

    PubMed Central  CAS  PubMed  Google Scholar 

  58. Agrawal H, Jacob N, Carreras E, Bajana S, Putterman C, Turner S, Neas B, Mathian A, Koss MN, Stohl W, Kovats S, Jacob CO: Deficiency of type I IFN receptor in lupus-prone New Zealand mixed 2328 mice decreases dendritic cell numbers and activation and protects from disease. J Immunol. 2009, 183: 6021-6029. 10.4049/jimmunol.0803872.

    PubMed Central  CAS  PubMed  Google Scholar 

  59. Mathian A, Weinberg A, Gallegos M, Banchereau J, Koutouzov S: IFN-α induces early lethal lupus in preautoimmune (New Zealand Black × New Zealand White) F1 but not in BALB/c mice. J Immunol. 2005, 174: 2499-2506.

    CAS  PubMed  Google Scholar 

  60. Fairhurst AM, Mathian A, Connolly JE, Wang A, Gray HF, George TA, Boudreaux CD, Zhou XJ, Li QZ, Koutouzov S, Banchereau J, Wakeland EK: Systemic IFN-α drives kidney nephritis in B6.Sle123 mice. Eur J Immunol. 2008, 38: 1948-1960. 10.1002/eji.200837925.

    PubMed Central  CAS  PubMed  Google Scholar 

  61. Ramanujam M, Kahn P, Huang W, Tao H, Madaio MP, Factor SM, Davidson A: Interferon-alpha treatment of female (NZW × BXSB)F(1) mice mimics some but not all features associated with the Yaa mutation. Arthritis Rheum. 2009, 60: 1096-1101. 10.1002/art.24414.

    PubMed Central  CAS  PubMed  Google Scholar 

  62. Mathian A, Gallegos M, Pascual V, Banchereau J, Koutouzov S: Interferon-α induces unabated production of short-lived plasma cells in preautoimmune lupus-prone (NZB × NZW)F1 mice but not in BALB/c mice. Eur J Immunol. 2011, 41: 863-872. 10.1002/eji.201040649.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. Hron JD, Peng SL: Type I IFN protects against murine lupus. J Immunol. 2004, 173: 2134-2142.

    CAS  PubMed  Google Scholar 

  64. Li J, Liu Y, Xie C, Zhu J, Kreska D, Morel L, Mohan C: Deficiency of type I interferon contributes to Sle2-associated component lupus phenotypes. Arthritis Rheum. 2005, 52: 3063-3072. 10.1002/art.21307.

    CAS  PubMed  Google Scholar 

  65. Nickerson KM, Christensen SR, Shupe J, Kashgarian M, Kim D, Elkon K, Shlomchik MJ: TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J Immunol. 2010, 184: 1840-1848. 10.4049/jimmunol.0902592.

    PubMed Central  CAS  PubMed  Google Scholar 

  66. Barrat FJ, Meeker T, Chan JH, Guiducci C, Coffmann RL: Treatment of lupusprone mice with a dual inhibitor of TLR7 and TLR9 leads to reduction of autoantibody production and amelioration of disease symptoms. Eur J Immunol. 2007, 37: 3582-3586. 10.1002/eji.200737815.

    CAS  PubMed  Google Scholar 

  67. Theofilopoulos AN, Baccala R, Beutler B, Kono DH: Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005, 23: 307-336. 10.1146/annurev.immunol.23.021704.115843.

    CAS  PubMed  Google Scholar 

  68. Niewold TB, Hua J, Lehman TJ, Harley JB, Crow MK: High serum IFN-α activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007, 8: 492-502. 10.1038/sj.gene.6364408.

    PubMed Central  CAS  PubMed  Google Scholar 

  69. Niewold TB, Adler JE, Glenn SB, Lehman TJ, Harley JB, Crow MK: Age- and sex-related patterns of serum interferon-alpha activity in lupus families. Arthritis Rheum. 2008, 58: 2113-2119. 10.1002/art.23619.

    PubMed Central  CAS  PubMed  Google Scholar 

  70. Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK: Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005, 52: 1491-1503. 10.1002/art.21031.

    CAS  PubMed  Google Scholar 

  71. Dall'era MC, Cardarelli PM, Preston BT, Witte A, Davis JC: Type I interferon correlates with serological and clinical manifestations of SLE. Ann Rheum Dis. 2005, 64: 1692-1697. 10.1136/ard.2004.033753.

    PubMed Central  PubMed  Google Scholar 

  72. Landolt-Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J, Gladman DD, Urowitz M, Fortin PR, Wither J: Lack of association between the interferon-alpha signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann Rheum Dis. 2009, 68: 1440-1446. 10.1136/ard.2008.093146.

    CAS  PubMed  Google Scholar 

  73. Bauer JW, Baechler EC, Petri M, Batliwalla FM, Crawford D, Ortmann WA, Espe KJ, Li W, Patel DD, Gregersen PK, Behrens TW: Elevated serum levels of interferon-regulated chemokines are biomarkers for active human systemic lupus erythematosus. PLoS Med. 2006, 3: e491-10.1371/journal.pmed.0030491.

    PubMed Central  PubMed  Google Scholar 

  74. Fu Q, Chen X, Cui H, Guo Y, Chen J, Shen N, Bao C: Association of elevated transcript levels of interferon-inducible chemokines with disease activity and organ damage in systemic lupus erythematosus patients. Arthritis Res Ther. 2008, 10: R112-10.1186/ar2510.

    PubMed Central  PubMed  Google Scholar 

  75. Bauer JW, Petri M, Batliwalla FM, Koeuth T, Wilson J, Slattery C, Panoskaltsis-Mortari A, Gregersen PK, Behrens TW, Baechler EC: Interferon-regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: a validation study. Arthritis Rheum. 2009, 60: 3098-3107. 10.1002/art.24803.

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW: Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci USA. 2003, 100: 2610-2615. 10.1073/pnas.0337679100.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, Pascual V: Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med. 2003, 197: 711-723. 10.1084/jem.20021553.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. Peterson KS, Huang JF, Zhu J, D'Agati V, Liu X, Miller N, Erlander MG, Jackson MR, Winchester RJ: Characterization of heterogeneity in the molecular pathogenesis of lupus nephritis from transcriptional profiles of laser-captured glomeruli. J Clin Invest. 2004, 113: 1722-1733.

    PubMed Central  CAS  PubMed  Google Scholar 

  79. Nzeusseu Toukap A, Galant C, Theate I, Maudoux AL, Lories RJ, Houssiau FA, Lauwerys BR: Identification of distinct gene expression profiles in the synovium of patients with systemic lupus erythematosus. Arthritis Rheum. 2007, 56: 1579-1588. 10.1002/art.22578.

    CAS  PubMed  Google Scholar 

  80. Tucci M, Quatraro C, Lombardi L, Pellegrino C, Dammacco F, Silvestris F: Glomerular accumulation of plasmacytoid dendritic cells in active lupus nephritis: role of interleukin-18. Arthritis Rheum. 2008, 58: 251-262. 10.1002/art.23186.

    CAS  PubMed  Google Scholar 

  81. Farkas L, Beiske K, Lund-Johansen F, Brandtzaeg P, Jahnsen FL: Plasmacytoid dendritic cells (natural interferon-alpha/beta-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am J Pathol. 2001, 159: 237-243. 10.1016/S0002-9440(10)61689-6.

    PubMed Central  CAS  PubMed  Google Scholar 

  82. Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, Banchereau J: Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity. 2003, 19: 225-234. 10.1016/S1074-7613(03)00208-5.

    CAS  PubMed  Google Scholar 

  83. Le Bon A, Thompson C, Kamphuis E, Durand V, Rossmann C, Kalinke U, Tough DF: Cutting edge: enhancement of antibody responses through direct stimulation of B and T cells by type I IFN. J Immunol. 2006, 176: 2074-2078.

    CAS  PubMed  Google Scholar 

  84. Ruuth K, Carlsson L, Hallberg B, Lundgren E: Interferon-alpha promotes survival of human primary B-lymphocytes via phosphatidylinositol 3-kinase. Biochem Biophys Res Commun. 2001, 284: 583-586. 10.1006/bbrc.2001.5025.

    CAS  PubMed  Google Scholar 

  85. Suen JL, Li HT, Jong YJ, Chiang BL, Yen JH: Altered homeostasis of CD4(+) FoxP3(+) regulatory T-cell subpopulations in systemic lupus erythematosus. Immunology. 2009, 127: 196-205. 10.1111/j.1365-2567.2008.02937.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  86. Venigalla RK, Tretter T, Krienke S, Max R, Eckstein V, Blank N, Fiehn C, Ho AD, Lorenz HM: Reduced CD4+, CD25-T cell sensitivity to the suppressive function of CD4+, CD25high, CD127-/low regulatory T cells in patients with active systemic lupus erythematosus. Arthritis Rheum. 2008, 58: 2120-2130. 10.1002/art.23556.

    PubMed  Google Scholar 

  87. Yan B, Ye S, Chen G, Kuang M, Shen N, Chen S: Dysfunctional CD4+, CD25+ regulatory T cells in untreated active systemic lupus erythematosus secondary to interferon-alpha-producing antigen-presenting cells. Arthritis Rheum. 2008, 58: 801-812. 10.1002/art.23268.

    CAS  PubMed  Google Scholar 

  88. Rönnblom LE, Alm GV, Oberg KE: Possible induction of systemic lupus erythematosus by interferon-alpha treatment in a patient with a malignant carcinoid tumour. J Intern Med. 1990, 227: 207-210. 10.1111/j.1365-2796.1990.tb00144.x.

    PubMed  Google Scholar 

  89. Niewold TB, Swedler WI: Systemic lupus erythematosus arising during interferon-alpha therapy for cryoglobulinemic vasculitis associated with hepatitis C. Clin Rheumatol. 2005, 24: 178-181. 10.1007/s10067-004-1024-2.

    PubMed  Google Scholar 

  90. Niewold TB: Interferon alpha-induced lupus: proof of principle. J Clin Rheumatol. 2008, 14: 131-132. 10.1097/RHU.0b013e318177627d.

    PubMed Central  PubMed  Google Scholar 

  91. Sigurdsson S, Nordmark G, Göring HH, Lindroos K, Wiman AC, Sturfelt G, Jönsen A, Rantapää-Dahlqvist S, Möller B, Kere J, Koskenmies S, Widén E, Eloranta ML, Julkunen H, Kristjansdottir H, Steinsson K, Alm G, Rönnblom L, Syvänen AC: Polymorphisms in the tyrosine kinase 2 and interferon regulatory factor 5 genes are associated with systemic lupus erythematosus. Am J Hum Genet. 2005, 76: 528-537. 10.1086/428480.

    PubMed Central  CAS  PubMed  Google Scholar 

  92. Hellquist A, Järvinen TM, Koskenmies S, Zucchelli M, Orsmark-Pietras C, Berglind L, Panelius J, Hasan T, Julkunen H, D'Amato M, Saarialho-Kere U, Kere J: Evidence for genetic association and interaction between the TYK2 and IRF5 genes in systemic lupus erythematosus. J Rheumatol. 2009, 36: 1631-1638. 10.3899/jrheum.081160.

    CAS  PubMed  Google Scholar 

  93. Niewold TB, Kelly JA, Flesch MH, Espinoza LR, Harley JB, Crow MK: Association of the IRF5 risk haplotype with high serum interferon-alpha activity in systemic lupus erythematosus patients. Arthritis Rheum. 2008, 58: 2481-2487. 10.1002/art.23613.

    PubMed Central  CAS  PubMed  Google Scholar 

  94. International Consortium for Systemic Lupus Erythematosus Genetics (SLEGEN), Harley JB, Alarcón-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP, Moser KL, Tsao BP, Vyse TJ, Langefeld CD, Nath SK, Guthridge JM, Cobb BL, Mirel DB, Marion MC, Williams AH, Divers J, Wang W, Frank SG, Namjou B, Gabriel SB, Lee AT, Gregersen PK, Behrens TW, Taylor KE, Fernando M, Zidovetzki R, Gaffney PM, Edberg JC, Rioux JD, Ojwang JO, et al: Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet. 2008, 40: 204-210. 10.1038/ng.81.

    Google Scholar 

  95. Kariuki SN, Kirou KA, MacDermott EJ, Barillas-Arias L, Crow MK, Niewold TB: Cutting edge: autoimmune disease risk variant of STAT4 confers increased sensitivity to IFN-alpha in lupus patients in vivo. J Immunol. 2009, 182: 34-38.

    PubMed Central  CAS  PubMed  Google Scholar 

  96. Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM, Tian C, Chung SA, Nititham J, Hom G, Kao AH, Demirci FY, Kamboh MI, Petri M, Manzi S, Kastner DL, Seldin MF, Gregersen PK, Behrens TW, Criswell LA: Specificity of the STAT4 genetic association for severe disease manifestations of systemic lupus erythematosus. PLoS Genet. 2008, 4: e1000084-10.1371/journal.pgen.1000084.

    PubMed Central  PubMed  Google Scholar 

  97. Abelson AK, Delgado-Vega AM, Kozyrev SV, Sánchez E, Velázquez-Cruz R, Eriksson N, Wojcik J, Linga Reddy MV, Lima G, D'Alfonso S, Migliaresi S, Baca V, Orozco L, Witte T, Ortego-Centeno N, AADEA Group, Abderrahim H, Pons-Estel BA, Gutiérrez C, Suárez A, González-Escribano MF, Martin J, Alarcón-Riquelme ME: STAT4 associates with systemic lupus erythematosus through two independent effects that correlate with gene expression and act additively with IRF5 to increase risk. Ann Rheum Dis. 2009, 68: 1746-1753. 10.1136/ard.2008.097642.

    CAS  PubMed  Google Scholar 

  98. Jacob CO, Zhu J, Armstrong DL, Yan M, Han J, Zhou XJ, Thomas JA, Reiff A, Myones BL, Ojwang JO, Kaufman KM, Klein-Gitelman M, McCurdy D, Wagner-Weiner L, Silverman E, Ziegler J, Kelly JA, Merrill JT, Harley JB, Ramsey-Goldman R, Vila LM, Bae SC, Vyse TJ, Gilkeson GS, Gaffney PM, Moser KL, Langefeld CD, Zidovetzki R, Mohan C: Identification of IRAK1 as a risk gene with critical role in the pathogenesis of systemic lupus erythematosus. Proc Natl Acad Sci USA. 2009, 106: 6256-6261. 10.1073/pnas.0901181106.

    PubMed Central  CAS  PubMed  Google Scholar 

  99. Denny MF, Thacker S, Mehta H, Somers EC, Dodick T, Barrat FJ, McCune WJ, Kaplan MJ: Interferon-α promotes abnormal vasculogenesis in lupus: a potential pathway for premature atherosclerosis. Blood. 2007, 110: 2907-2915. 10.1182/blood-2007-05-089086.

    PubMed Central  CAS  PubMed  Google Scholar 

  100. Lee PY, Li Y, Richards HB, Chan FS, Zhuang H, Narain S, Butfiloski EJ, Sobel ES, Reeves WH, Segal MS: Type I interferon as a novel risk factor for endothelial progenitor cell depletion and endothelial dysfunction in systemic lupus erythematosus. Arthritis Rheum. 2007, 56: 3759-3769. 10.1002/art.23035.

    CAS  PubMed  Google Scholar 

  101. Zhao W, Somers E, McCune WJ, Kaplan MJ: Type I interferon signatures are associated with vascular risk and atherosclerosis in systemic lupus erythematosus [abstract]. Arthritis Rheum. 2009, 59: S582-

    Google Scholar 

  102. Yao Y, Richman L, Higgs BW, Morehouse CA, de los Reyes M, Brohawn P, Zhang J, White B, Coyle AJ, Kiener PA, Jallal B: Neutralization of interferon-alpha/beta-inducible genes and downstream effect in a phase I trial of an anti-interferon-alpha monoclonal antibody in systemic lupus erythematosus. Arthritis Rheum. 2009, 60: 1785-1796. 10.1002/art.24557.

    CAS  PubMed  Google Scholar 

  103. McBride JM, Wallace DJ, Yao Z, Morimoto A, Jiang J, Maciuca R, McLean I, Drappa J: Dose-dependent modulation of interferon regulated genes with administration of single and repeat doses of rontalizumab in a phase I, placebo controlled, double blind, dose escalation study in SLE. Arthritis Rheum. 2009, 60 (Suppl): S775-S776.

    Google Scholar 

  104. Mackay F, Schneider P: Cracking the BAFF code. Nat Rev Immunol. 2009, 9: 491-502. 10.1038/nri2572.

    CAS  PubMed  Google Scholar 

  105. Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, Tschopp J, Browning JL: Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999, 190: 1697-1710. 10.1084/jem.190.11.1697.

    PubMed Central  CAS  PubMed  Google Scholar 

  106. Stohl W, Xu D, Kim KS, Koss MN, Jorgensen TN, Deocharan B, Metzger TE, Bixler SA, Hong YS, Ambrose CM, Mackay F, Morel L, Putterman C, Kotzin BL, Kalled SL: BAFF overexpression and accelerated glomerular disease in mice with an incomplete genetic predisposition to systemic lupus erythematosus. Arthritis Rheum. 2005, 52: 2080-2091. 10.1002/art.21138.

    CAS  PubMed  Google Scholar 

  107. Jacob CO, Pricop L, Putterman C, Koss MN, Liu Y, Kollaros M, Bixler SA, Ambrose CM, Scott ML, Stohl W: 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: 2671-2680.

    PubMed Central  CAS  PubMed  Google Scholar 

  108. Ramanujam M, Bethunaickan R, Huang W, Tao H, Madaio MP, Davidson A: Selective blockade of BAFF for the prevention and treatment of systemic lupus erythematosus nephritis in NZM2410 mice. Arthritis Rheum. 2010, 62: 1457-1468.

    PubMed Central  CAS  PubMed  Google Scholar 

  109. Jacob N, Guo S, Mathian A, Koss MN, Gindea S, Putterman C, Jacob CO, Stohl W: B cell and BAFF dependence of IFN-α-exaggerated disease in systemic lupus erythematosus-prone NZM 2328 mice. J Immunol. 2011, 186: 4984-4993. 10.4049/jimmunol.1000466.

    PubMed Central  CAS  PubMed  Google Scholar 

  110. Stohl W, Looney RJ: B cell depletion therapy in systemic rheumatic diseases: different strokes for different folks?. Clin Immunol. 2006, 121: 1-12. 10.1016/j.clim.2006.03.010.

    CAS  PubMed  Google Scholar 

  111. Collins CE, Gavin AL, Migone TS, Hilbert DM, Nemazee D, Stohl W: B lymphocyte stimulator (BLyS) isoforms in systemic lupus erythematosus: disease activity correlates better with blood leukocyte BLyS mRNA levels than with plasma BLyS protein levels. Arthritis Res Ther. 2006, 8: R6-10.1186/ar1855.

    PubMed Central  PubMed  Google Scholar 

  112. Chu VT, Enghard P, Schürer S, Steinhauser G, Rudolph B, Riemekasten G, Berek C: Systemic activation of the immune system induces aberrant BAFF and APRIL expression in B cells in patients with systemic lupus erythematosus. Arthritis Rheum. 2009, 60: 2083-2093. 10.1002/art.24628.

    CAS  PubMed  Google Scholar 

  113. Lee YH, Ota F, Kim-Howard X, Kaufman KM, Nath SK: APRIL polymorphism and systemic lupus erythematosus (SLE) susceptibility. Rheumatology (Oxford). 2007, 46: 1274-1276. 10.1093/rheumatology/kem093.

    CAS  Google Scholar 

  114. Kawasaki A, Tsuchiya N, Fukazawa T, Hashimoto H, Tokunaga K: Analysis on the association of human BLYS (BAFF, TNFSF13B) polymorphisms with systemic lupus erythematosus and rheumatoid arthritis. Genes Immun. 2002, 3: 424-429. 10.1038/sj.gene.6363923.

    CAS  PubMed  Google Scholar 

  115. Baker KP, Edwards BM, Main SH, Choi GH, Wager RE, Halpern WG, Lappin PB, Riccobene T, Abramian D, Sekut L, Sturm B, Poortman C, Minter RR, Dobson CL, Williams E, Carmen S, Smith R, Roschke V, Hilbert DM, Vaughan TJ, Albert VR: Generation and characterization of LymphoStat-B, a human monoclonal antibody that antagonizes the bioactivities of B lymphocyte stimulator. Arthritis Rheum. 2003, 48: 3253-3265. 10.1002/art.11299.

    CAS  PubMed  Google Scholar 

  116. Furie R, Stohl W, Ginzler EM, Becker M, Mishra N, Chatham W, Merrill JT, Weinstein A, McCune WJ, Zhong J, Cai W, Freimuth W, Belimumab Study Group: Biologic activity and safety of belimumab, a neutralizing anti-B-lymphocyte stimulator (BLyS) monoclonal antibody: a phase I trial in patients with systemic lupus erythematosus. Arthritis Res Ther. 2008, 10: R109-10.1186/ar2506.

    PubMed Central  PubMed  Google Scholar 

  117. Wallace DJ, Stohl W, Furie RA, Lisse JR, McKay JD, Merrill JT, Petri MA, Ginzler EM, Chatham WW, McCune WJ, Fernández V, Chevrier MR, Zhong ZJ, Freimuth WW: A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus. Arthritis Rheum. 2009, 61: 1168-1178. 10.1002/art.24699.

    PubMed Central  CAS  PubMed  Google Scholar 

  118. Furie RA, Petri MA, Wallace DJ, Ginzler EM, Merrill JT, Stohl W, Chatham WW, Strand V, Weinstein A, Chevrier MR, Zhong ZJ, Freimuth WW: Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum. 2009, 61: 1143-1151. 10.1002/art.24698.

    PubMed Central  CAS  PubMed  Google Scholar 

  119. Navarra S, Guzman R, Gallacher A, Levy RA, Li EK, Thomas M, Jimenez R, Leon M, Hall S, Lan JL, Nasonov E, Tanasescu C, Kim HY, Pineda L, Zhong ZJ, Freimuth W, Petri MA, BLISS-52 Study Group: Belimumab, a BLyS-specific inhibitor, reduced disease activity, flares and prednisone use in patients with active SLE: efficacy and safety results from the phase 3 BLISS-52 study [abstract]. Arthritis Rheum. 2009, 60: 3859-

    Google Scholar 

  120. Furie R, Zamani O, Wallace D, Tegzová D, Petri M, Merrill JT, Chatham W, Stohl W, Schwarting A, Cooper S, Zhong ZJ, Freimuth W, Hough D, van Vollenhoven RF, BLISS-76 Study Group: Belimumab, a BLyS-specific inhibitor, reduced disease activity and severe flares in seropositive SLE patients: BLISS-76 study results through wk 76. Arthritis Rheum. 2010, 62: S606-

    Google Scholar 

  121. Manzi S, Sanchez-Guerrero J, Merrill JT, Furie RA, Gladman D, Navarra S, Ginzler EM, D'Cruz D, Doria A, Cooper S, Zhong ZJ, Hough D, Freimuth W, Petri M, BLISS-52 and BLISS-76 Study Groups: Belimumab, a BLyS-specific inhibitor, reduced disease activity across multiple organ domains: combined efficacy results from the phase 3 BLISS-52 and -76 studies. Arthritis Rheum. 2010, 62: S607-

    Google Scholar 

  122. Dall'Era M, Chakravarty E, Wallace D, Genovese M, Weisman M, Kavanaugh A, Kalunian K, Dhar P, Vincent E, Pena-Rossi C, Wofsy D: Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial. Arthritis Rheum. 2007, 56: 4142-4150. 10.1002/art.23047.

    PubMed  Google Scholar 

  123. Stohl W, Merrill JT, Looney RJ, Buyon J, Wallace DJ, Weisman M, Ginzler EM, Cooke B, Holloway D, Kuchimanchi K, Cheah TC, Rasmussen E, Ferbas J, Belouski SS, Zack DJ: Phase 1a single- and phase 1b multiple-dose studies of AMG 623 (an anti-BAFF peptibody) in systemic lupus erythematosus (SLE). Arthritis Rheum. 2008, 58: S565-

    Google Scholar 

  124. Stohl W, Hiepe F, Thomas M, Scheinberg M, Clarke A, Aranow C, Jimenez R, Wellborne F, Abud-Mendoza C, Hough D, Pineda L, Migone TS, Freimuth W, Chatham W, BLISS-52 and BLISS-76 Study Groups: Belimumab, a BLyS-specific inhibitor, significantly reduced autoantibodies, normalized low complement, and reduced selected B-cell populations in patients with seropositive systemic lupus erythematosus (SLE): the phase 3 BLISS studies [abstract]. Arthritis Rheum. 2010, 62: S480-10.1002/acr.20038.

    Google Scholar 

  125. Kikly K, Manetta J, Smith H, Wierda D, Witcher D: Characterization of LY2127399, a neutralizing antibody for BAFF [abstract]. Arthritis Rheum. 2009, 60: 693-

    Google Scholar 

Download references

Acknowledgements

The present work was supported in part by a grant from the National Institutes of Health (R01 AR059103) to WS.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to William Stohl.

Additional information

Competing interests

WS has conducted clinical trials in systemic lupus erythematosus with belimumab (sponsored by Human Genome Sciences). NJ has no competing interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jacob, N., Stohl, W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res Ther 13, 228 (2011). https://doi.org/10.1186/ar3349

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/ar3349

Keywords