Review

Targeting B cells to treat systemic erythematosus

B cells play a central role in the pathogenesis of systemic lupus erythematosus. Of late, there has been growing interest in utilizing B cells as targets for the development of new treatments for this frequently devastating disease. In addition to producing (auto)antibodies, B cells are involved in a variety of autoantibody-independent pathogenic mechanisms, such as effector functions, cytokine production and costimulation. Therefore, depleting B cells or inhibiting their actions can suppress the immune hyperactivity that is characteristic of systemic lupus erythematosus. This article examines the latest advances in novel B-cell-directed therapies for patients with systemic lupus erythematosus.

1 KEYWORDS: B‑cell depletion n B‑cell -directed therapies n B‑lymphocyte stimulator n Arlene T Tieng , CD20 n CD22 n costimulation n systemic lupus erythematosus Gisele Zandman-Goddard2,3 & Elena Peeva†1 Systemic lupus erythematosus (SLE) is a chronic are hyperactive and display a variety of abnor- 1Rheumatology Division, Department of Medicine, Albert Einstein College of autoimmune disorder that can induce a variety malities; lupus B cells have increased calcium Medicine, Bronx, NY, USA of nonspecific constitutional symptoms such as influx upon B‑cell receptor engagement, reduced 2Department of Medicine, fatigue, malaise, fever and weight loss, but also levels of Lyn kinase and increased CD45 in lipid Wolfson Medical Center, Holon, Israel 3Sackler Faculty of Medicine, Tel-Aviv can involve various organs such as the skin, rafts, and decreased expression of FcgRIIB on University, Tel Aviv, Israel - + † joints, heart, kidneys, nervous, hematologic IgM CD27 memory cells [7]. Author for correspondence: Arthritis Clinic, Montefiore Hospital, and musculoskeletal systems. Lupus nephritis is Despite the multitude of studies testing vari- DTC Bldg 440,111 E 210th St, Bronx, the key predictor of poor outcome. Some of the ous therapies, there are only a few drugs that are NY 10467, USA Tel.: +1 718 920 5584 SLE manifestations have an unpredictable clini- approved for treatment of SLE [8]. The current Fax: +1 718 798 3029 cal course and can be life-threatening. Genetics treatments are nonspecific immunosuppressants [email protected] seem to play an important role in lupus severity that can induce multiple side effects including as ethnicity predisposes people more to renal and severe infections and ovarian failure. The ratio- neuropsychiatric damage than socioeconomic nale for developing B‑cell-directed therapies for factors [1]. Furthermore, SLE manifestations are SLE is based on the aforementioned critical role usually more severe in African and Asian patients of B cells in the pathogenesis of lupus. The aim is than in those of European ancestry [1]. Compared to develop novel focused drugs that will be devoid with the general population, SLE patients have a of the significant side effects of the broad-spec- three- to five-fold increased mortality [2]. trum immunosuppressants. Better specificity and The fundamental immunologic characteris- improved efficacy are the major characteristics tic of SLE is a broad dysregulation of immune expected of the new drugs, with the goal being responses that includes hyperactivity of CD4+ amelioration of SLE disease activity and preven- helper T cells and B cells, leading to the forma- tion of sequels without significant side effects. tion of autoantibodies [3,4]. Antinuclear anti­ This article will highlight the newest treatments bodies (ANAs), the hallmark autoantibodies that target B cells in lupus (Figure 1). Despite the in SLE, are directed against dsDNA and other fact that some of the trials that target B cells have nuclear components [5]. Although historically, the been disappointing, they have provided valuable production of autoantibodies has been thought of information that can help improve the design of as the only mechanism of B‑cell-mediated effects, future trials. this notion is now archaic, as B cells have been found to exert a handful of other functions, such Therapies directed to B‑cell surface as the presentation of autoantigens to T cells, reg- markers CD20 & CD22 ulation of dendritic cell differentiation, mainte- Rituximab is the most commonly used mono- nance of lymphoid organization, and secretion clonal antibody for rapid B-cell depletion. of cytokines that can affect and It is a chimeric antibody directed against the lymphopoesis, and play other effector roles in B‑cell-specific surface marker CD20, which is immune responses [2,6]. B cells in SLE patients expressed on immature and mature B cells, but

10.2217/IJR.10.95 © 2010 Future Medicine Ltd Int. J. Clin. Rheumatol. (2010) 5(6), 627–636 ISSN 1758-4272 627 Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

IL-10 B-N10

Tocilizumab IL-6 AMG 557

B7RP-1/ICOS

Epratuzumab CD22

B7/CTLA4 Rituximab CD20 SBI-087 T cell Abatacept

Belimumab BAFF BAFF

Atacicept

APRIL

Figure 1. Presentation of the novel B‑cell-directed therapies for systemic lupus erythematosus. APRIL: A proliferation-inducing ligand; BAFF: B‑cell-activating factor; ICOS: Inducible costimulator.

not on early pre-B cells or plasma cells (Table 1). clinical response, but the difference did not Rituximab depletes CD20+ B cells through anti- reach statistical significance (29.6 vs 28.4%). body-dependent cell-mediated and complement- However, the decrease and increase in the levels mediated cytotoxicity [9]. Rituximab has been of anti-dsDNA autoantibodies and complement, approved for the treatment of non-Hodgkin’s respectively, were significantly different between B‑cell lymphoma and, more recently, rheuma- the rituximab- and the placebo-treated patients. toid arthritis. In case reports and case series, Furthermore, patients with baseline thrombo- rituximab has benefited SLE patients with cytopenia had improved platelet counts when refractory vasculitic ulcers, shrinking lung syn- treated with rituximab [18]. It is also worthwhile drome, refractory thrombotic thrombocytopenic noting that the subgroup ana­lysis showed that purpura, bilateral retinal vasculitis and recur- the African–American and Hispanic patients, rent enteritis [10–14]. Although open-label trials who comprised approximately a third of the also implied the efficacy of rituximab in SLE study population, did have significant benefit [15,16], two moderate-sized, randomized, pla- from rituximab (33.8 vs 15.7%). In addition, an cebo-controlled trials of rituximab in extrarenal ad hoc ana­lysis demonstrated that the subgroup and renal lupus (Exploratory Phase II/III SLE of patients who received rituximab on back- Evaluation of Rituximab [EXPLORER] and ground therapy with methotrexate had improved Lupus Nephritis Assessment with Rituximab mean global British Isles Lupus Assessment [LUNAR] trials, respectively) disappointingly Group (BILAG) scores compared with placebo. failed to meet their primary and secondary end Serious adverse events occurred at a similar per- points. The EXPLORER trial was a 52‑week- centage of patients in both the rituximab and long study that randomly assigned 257 SLE placebo groups (37.9 vs 36.4%). patients with moderate to severe disease activity The LUNAR trial was a Phase III, random- in a 2:1 ratio to rituximab or placebo [17]. The ized, double‑blind, placebo-controlled, multi- patients were administered intravenous infu- center study in which 144 patients with active sions as two 1000‑mg doses on days 1, 15, 168 proliferative nephritis (class III/IV) were ran- and 182, as well as daily prednisone (as per trial domly assigned in a 1:1 ratio to rituximab or regimen) and their baseline immunosuppressive placebo, in the same dose and regimen that was regimen, which could have included azathio- administered in EXPLORER [19]. Background prine, mycophenolate mofetil or methotrexate. therapy of steroids and mycophenolate mofetil Compared with placebo, slightly more patients was permitted. Patients who took prednisone at in the rituximab group showed a major or partial a dose greater than 20 mg daily for more than

628 Int. J. Clin. Rheumatol. (2010) 5(6) future science group Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

2 weeks prior to screening were excluded. The The fact that EXPLORER and LUNAR primary end point, assessed at 52 weeks, was the studies did not reach their end points has pro- percentage of patients who achieved a complete voked multiple discussions regarding their study or partial renal response. There were numerically designs, end points and background immuno- more responders in the rituximab group than in suppressive therapies [20,21]. Concerns regarding the placebo group (57 vs 46%), but the differ- glucocorticoids and other immunosuppressants ence did not reach statistical significance in the masking rituximab’s effects have been expressed. end points – complete or partial renal responses In addition, some lupologists believe that the at week 52. Despite this, the rituximab group variability caused by SLE heterogeneity may displayed a significantly greater improvement have had a negative impact on the results of the in the levels of anti-dsDNA and complement rituximab studies and advocate that new clini- than the placebo group. The numbers of serious cal trials focus on patient subsets with specific adverse events were similar between the treat- organ system involvement and/or specific lupus ment and placebo groups, except that the ritux- autoantibody profile. Furthermore, additional imab group had more neutropenia, ­leukopenia research has shown that the following factors and hypotension. are closely connected to the therapeutic effect (or

Table 1. Novel B‑cell-directed therapies for systemic lupus erythematosus. Drug Class Target Method Study Outcomes Ref. Rituximab Chimeric CD20 B‑cell Phase II/III Failed to meet [17,19] monoclonal depletion (EXPLORER), primary end points antibody Phase III (LUNAR) Ocrelizumab Humanized CD20 B‑cell Phase III Terminated due to [29] monoclonal depletion (BEGIN), infections antibody Phase III (BELONG) SBI-087 Humanized CD20 B‑cell Phase I Currently recruiting [30] fusion protein depletion Epratuzumab Humanized CD22 B‑cell Phase III Improved BILAG but [35] monoclonal reduction (SL0003, terminated early due antibody SL0004) to manufacturing problems Phase IIb BILAG improvement (EMBLEM) in all affected body systems Abatacept CTLA4 Ig B7 Blockade of Phase IIb, Did not meet primary [36] fusion protein costimulation Phase II end point, currently (ACCESS) recruiting AMG 557 Fully human B7RP-1 Blockade of Phase I Enrolling participants monoclonal costimulation by invitation only antibody Fully human BLys Blockade of Phase III Improved the SRI [44,106] monoclonal cytokines (BLISS-52 antibody and BLISS-76) Atacicept Recombinant BAFF Blockade of Phase II, Halted owing [52] fusion protein and cytokines Phase II/III to infection, APRIL (APRIL SLE) actively recruiting Humanized a‑chain Blockade of Phase I Improved SLAM and [56] monoclonal of the cytokines mSELENA-SLEDAI antibody IL-6 receptor B-N10 Murine IL-10 Blockade of Open pilot Improved [59] monoclonal cytokines MEX-SLEDAI antibody ACCESS: Abatacept and Cyclophosphamide Combination Therapy for Lupus Nephritis; APRIL: A proliferation-inducing ligand; BAFF: B‑cell-activating factor; BILAG: British Isles Lupus Assessment Group; BLyS: B lymphocyte stimulator; EXPLORER: Exploratory Phase II/III SLE Evaluation of Rituximab; LUNAR: Lupus Nephritis Assessment with Rituximab; SLE: Systemic lupus erythematosus; SLEDAI: Systemic Lupus Erythematosus Disease Activity Index; SRI: SLE responder index.

future science group www.futuremedicine.com 629 Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

lack of) of rituximab: serum levels of the treat- steroids and cyclophosphosphamide prior to the ment antibody, FcgRIIIa genotype, phenotype two rounds of rituximab 375 mg/m2/week for of the circulating B cells and the B‑cell subset 4 weeks [27]. It is uncertain whether rituximab emerging after the completion of treatment [22,23], itself led to the development of progressive multi- indicating that the prediction of response is very focal leukoencephalopathy, since it has been indi- complex and includes a high number of variables. cated that severe and SLE Studies with lupus prone mice have demon- alone may be associated with a predisposition to strated that even without secreting antibodies, progressive multifocal leukoencephalopathy [28]. B cells significantly influenced the course of The experience with anti-CD20 therapies other lupus [24,25]. These results suggest that therapies than rituximab is rather scarce, and includes ocrel- targeting B cells, but not affecting plasma cells izumab and SBI-087. Ocrelizumab, a humanized and/or autoantibodies, might be an effective against CD20, underwent a treatment for SLE. However, the lack of CD20 Phase III clinical trial (BEGIN) for active extra- expression on the surface of plasma cells has not renal SLE [101] and Phase III trial (BELONG) for been excluded as a possible factor implicated in class III or IV lupus nephritis [29,102], but the latter the failure of the randomized controlled trials study was terminated due to serious and oppor- (RCTs) with rituximab. Concerns regarding the tunistic infections. The reasons for the increased lack of effect of rituximab on antibody secret- infection rates with ocrelizumab, but not ritux- ing cells have led to the consideration of utilizing imab, are not clear. SBI-087 is a novel therapeutic rituximab along with agents affecting antibody agent that targets CD20; it is a humanized small secretion. Nevertheless, combination treatments modular immuno­pharmaceutical that consists of

of rituximab and anti-B lymphocyte stimula- single-chain variable regions (VL and VH) that tor (BLyS) or anti-IL‑10 monoclonal antibody bind CD20 [30]. These single chains are fused ­therapies have not been explored. by a human IgG1 hinge domain to the constant The ana­lysis of prospective data from the regions of human IgG1 heavy domains (CH2 nationwide French Autoimmunity and Rituximab and CH3). In an open-label study, this anti- (AIR) registry showed good clinical efficacy of CD20 fusion protein was administered at doses rituximab in the treatment of SLE patients [26]. of 0.5 mg/kg intravenous, 25 mg subcutaneous The AIR patients (n = 136) had articular, cuta- or 75 mg subcutaneous to 18 patients with con- neous, hematologic and renal manifestations of trolled SLE, and was generally well tolerated [31]. SLE. More than half of the patients were treated SBI-087 is currently in a Phase I trial for SLE + with two doses of 1 g rituximab, and less than [103]. This study plans to enroll 24 ANA patients half were treated with four doses of 375 mg/m2 with SLE to assess safety and tolerability as pri- rituximab. Rituximab was added to a stable back- mary outcome measures, and pharmacokinetics ground regimen of immuno­suppressive agents, and pharmacodynamics as ­secondary end points. and the mean prednisone dosage was 30 mg/day. Epratuzumab is a humanized monoclonal In the entire cohort, the mean follow-up duration antibody against the B‑cell surface was 18.6 months from the last rituximab infu- CD22. CD22 is expressed on a subset of mature sion. The mean Systemic Lupus Erythematosus B cells, but disappears when B cells differentiate Disease Activity Index (SLEDAI) score decreased into plasma cells. This molecule is involved in significantly at 6 months. The rate of severe B‑cell receptor (BCR) signaling and can exert infections was less than that reported in the both stimulatory or inhibitory effects on the EXPLORER study. However, the AIR registry BCR signal transduction [32,33]. does not meet the rigor of a randomized clini- Epratuzumab downregulates hyperactive cal trial; the data are observational and reflect B cells by inhibiting BCR signaling and by induc- anecdotal experience, and therefore do not allow ing rapid internalization of CD22 [34]. In addi- for reliable conclusions. In any case, the AIR reg- tion, epratuzumab causes approximately 40% istry data are positive and the two major RCTs reduction in the B cells in peripheral blood. The with rituximab have reported negative findings, results of the initial Phase III RCTs, SL0003 and reopening the debate on clinical benefit of B‑cell SL0004, suggested clinically meaningful efficacy depletion in SLE. of epratuzumab in SLE patients at week 12, but Of the serious adverse events associated with both trials were terminated early due to problems this treatment, it is noteworthy to mention that with manufacturing the . two SLE patients who received rituximab later on More recently, a dose-ranging Phase IIb study developed progressive multifocal leukoencepha- with epratuzumab, EMBLEM™, was completed. lopathy. One of the patients was treated with As reported at EULAR 2010, epratuzumab

630 Int. J. Clin. Rheumatol. (2010) 5(6) future science group Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

demonstrated clinically meaningful improve- B7-related protein‑1. A Phase I study is currently ments in patients with moderate-to-severe SLE. enrolling SLE patients to compare AMG 557 Statistically significant response rates to epratu- with placebo [105]. zumab were seen in the 600 mg/week group and in the combined group of all patients who Therapies that block received a cumulative dose of 2400 mg during B‑cell-stimulating cytokines the 12‑week treatment. Compared with placebo, The cytokine BLyS, which is also known as epratuzumab 600 mg/week provided greater B‑cell-activating factor or BAFF, belongs to the BILAG improvement in all affected body sys- tumor necrosis family. BLyS is an essential fac- tems, but the efficacy was especially prominent tor in the differentiation, homeostasis, selection in cardiorespiratory and neuro­psychiatric sys- and survival of B cells [39]. SLE patients have tems [35]. Two Phase III studies of epratuzumab elevated circulating BLyS levels and rising BLyS for the treatment of patients with moderate and concentrations can predict increases in SLE severe lupus will be initiated in the second half disease activity [40]. In the Phase I trial, belim- of 2010. umab, a recombinant, fully human monoclonal antibody that binds to soluble human BLyS, Therapies that block costimulation was found to be biologically active in vivo and Costimulation plays a role in T‑cell activation, was safely administered to and well tolerated in and also in the multistep process of antibody patients with mild-to-moderate SLE [41]. The production. One of the costimulatory pathways Phase II study randomized 449 SLE patients involves the CD28 and cytotoxic T lymphocyte with a SELENA-SLEDAI score of at least four antigen 4 (CTLA4) receptors on T cells and their to receive 1, 4 or 10 mg/kg of belimumab or coligands B7.1 and B7.2 on antigen-presenting placebo on days 0, 14 and 28, and then every cells, including B cells. CTLA4 binds the B7 28 days in addition to standard of care therapy molecules with greater affinity than CD28 and [42]. This trial did not meet its co-primary effi- delivers an ­inhibitory signal to the activated cacy end points of changes in SELENA-SLEDAI T cell. score or time to first flare during 52 weeks of Abatacept, a CTLA4 Ig fusion protein, acts treatment. The inclusion criteria for this study as a blocker of costimulation. An exploratory, required SLE patients to have a history of auto- Phase IIb trial randomized 175 SLE patients antibodies such as ANAs or anti-dsDNAs, with polyarthritis, discoid lesions, pleuritis but they were not mandated to be positive at and/or pericarditis in a 2:1 ratio to abatacept screening. Thus, at baseline, only 71.3% of these (10 mg/kg) or placebo [36]. Therapy was given patients had an ANA titer of 1:80 or greater and in addition to prednisone 30 mg/day for a 50% had anti-dsDNA antibodies. The subset of month, which was then tapered. The primary seronegative patients included in the study could end point was the proportion of patients with have confounded the evaluation of belimumab’s new BILAG A/B score flare in any organ sys- effect on the reduction of SLE disease activity tem after the beginning of the steroid taper. [43]. When serologically active patients (ANAs This study did not meet its primary or second- ≥1:80 by HEp-2 cell immunofluorescence ary end points. A Phase II study (Abatacept and and/or anti-dsDNA antibodies ≥30 IU/ml) were Cyclophosphamide Combination Therapy for retrospectively evaluated, the belimumab group Lupus Nephritis [ACCESS]) sponsored by the had a statistically larger percentage of responders National Institute of Allergy and Infectious than the placebo group at 52 and 56 weeks. Two Diseases is currently recruiting patients with large anti-BLyS Phase III trials (BLISS‑52 and active proliferative lupus nephritis to deter- ‑76) treated 865 and 819 patients, respectively, mine whether abatacept in combination with and reported favorable clinical responses com- cyclophosphamide is more effective than cyclo­ pared with placebo. The primary efficacy end phosphamide alone [104]. The primary outcome point in both trials was the SLE responder index, measure is the proportion of patients who defined as improvement in four or more points achieve a complete response at 24 weeks. in SELENA-SLEDAI score without clinically Closely related to CD28 is a molecule called significant worsening in the physician’s global inducible co-stimulator [37]. Inducible co- assessment and BILAG. BLISS‑52 random- stimulator interacts with B7-related protein-1, ized seropositive SLE patients to beliumumab which is expressed on B cells. B7-related pro- 1 or 10 mg/kg or placebo on days 0, 14 and tein‑1 costimulates T cells [38]. AMG 557 is a 28 and then every 28 days for 48 weeks on top fully human monoclonal antibody that binds to of standard of care therapy [44]. Belimumab

future science group www.futuremedicine.com 631 Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

significantly reduced SLE disease activity, and in the mature and total B cell numbers. Mild flare rates and use of prednisone. BLISS‑76, a injection site reactions were observed more fre- Phase III, 76‑week study, also evaluated belim- quently in the atacicept-treated patients than the umab 1 mg/kg or 10 mg/kg against placebo [106]. placebo-treated patients. Another Phase Ib study The study enrolled 275 patients in the placebo randomized 24 patients with mild-to-moderate arm 271 patients in the belimumab 1 mg/kg SLE (5:1) to receive intravenous atacicept, sin- arm, and 273 patients in the belimumab 10 mg/ gle or multiple doses, or placebo and followed kg arm. Only patients with positive serology them for 6 weeks [50]. Treatment with atacicept (ANA and anti-DNA antibodies) were included resulted in a reduction of B cells in the periph- in the study. At week 52, the response rates eral blood (40% from baseline in the single dose for placebo, belimumab 1 and 10 mg/kg were group and 55% reduction from baseline in the 33.6, 40.6 and 43.2%, respectively. Compared multiple dose group) and immunoglobulin lev- with placebo, only the 10 mg/kg dose showed els, and again only mild injection site reactions ­significantly improved SLE responder index. were reported. Despite this favorable safety pro- AMG 623 is a peptide-Fc fusion protein, file of atacicept monotherapy, a Phase II trial which binds both cell surface-expressed BLyS using atacicept in combination with mycopheno- and soluble BLyS. AMG 623 was administered late mofetil for active lupus nephritis was halted subcutaneously or intravenously against placebo because of increased serious infection rates [52]. to 54 and 63 SLE patients in Phase IA and IB Nonetheless, a Phase II/III study (APRIL SLE) studies, respectively [45]. A dose-independent is actively recruiting SLE patients who recently decrease in the percentage of naive B cells was experienced a flare. The study will compare the detectable, and the safety profile was compara- effects of atacicept 75 or 150 mg subcutaneously ble between the AMG 623 and placebo groups. versus placebo over a period of 52 weeks [107]. Further studies are needed to evaluate therapeu- The primary outcome measure will assess the tic effects of AMG 623 on lupus disease activ- proportion of patients with a new flare as defined ity and to elucidate whether AMG 623’s abil- by a BILAG score of A or B. ity to bind both surface and soluble BLyS will The overproduction of IL‑10 and ‑6 in SLE- translate into better efficacy than the efficacy inspired studies to determine if blockade of these of ­belimumab, which binds only soluble BLyS. cytokines can have therapeutic benefit[53] . B cells B‑cell-activating factor mediates its effects have been shown to produce IL‑6 [54]. In SLE through three different receptors: BAFF recep- patients, IL‑6 also appears to activate B cells, tor, B-cell maturation antigen and transmem- and thus plays a role in the production of anti- brane activator and calcium modulator and DNA antibodies. In particular, IL‑6 contributes cyclophilin-ligand interactor (TACI). Related to the generation of the IgG anti-DNA antibod- to BAFF is another member of the TNF ligand ies, which are known for their pathogenic poten- superfamily called a proliferation-inducing tial [55]. In an open-label Phase I pilot study with ligand (APRIL; also known as TNF ligand tocilizumab, a humanized monoclonal antibody superfamily member 13A, TALL-1 and TRDL- against the IL‑6 receptor, 16 SLE patients with 1). APRIL functions as a soluble factor and binds moderately active lupus were given this therapy to B-cell maturation antigen and TACI, but does in doses of 2, 4 or 8 mg/kg intravenously every not bind to the BAFF receptor. APRIL is involved 2 weeks for a total of seven infusions in addi- in T‑cell-independent type II antigen responses tion to prednisone [56]. At the end of 14 weeks, [46] and activation of B cells [47]. In addition, there was a significant improvement in modified APRIL can complex with BLyS into heterotri- SELENA-SLEDAI and Systemic Lupus Activity mers, which can promote survival, selection and Measure (SLAM) scores. Although the primary differentiation of B cells into plasma cells [48]. adverse event was dose-related neutropenia, these Circulating levels of APRIL/BLyS heterotrimers results seem promising and can lead to future are frequently increased in patients with SLE [49]. controlled trials of tocilizumab in SLE patients. Atacicept (TACI-Ig) is a recombinant fusion IL‑10 plays a critical role in immunoglobulin protein that binds both BAFF and APRIL, and production [57] and SLE patients have increased inhibits their effects on B cells [50]. A Phase Ib serum levels of IL‑10 [58]. An open pilot study trial included 47 patients with mild-to-moder- of six SLE patients who received daily infusions ate SLE who were treated with subcutaneous of B‑N10, an anti-IL‑10 monoclonal antibody, atacicept or placebo in a 3:1 ratio [51]. Atacicept for 21 days showed that blocking IL‑10 may administration was associated with dose-depen- have beneficial therapeutic effect in SLE. After dent reductions in the immunoglobulin levels 6 months, five patients were clinically inactive,

632 Int. J. Clin. Rheumatol. (2010) 5(6) future science group Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

as measured by Mexican (MEX)-SLEDAI scores The serological status of patients is another of less than three [59]. The only adverse event was important parameter that needs to be consid- chills during an infusion on day 16. However, ered in lupus trials. In the Phase II studies of there are no ongoing trials with this treatment. belimumab, a large number of seronegative SLE patients were included, which may have skewed Conclusion the results. However, the recent Phase III study Systemic lupus erythematosus is consid- with belimumab that included only ANA+ ered a proto­typical . patients met its primary end point and, thus Autoantibodies, the trademark of SLE, are pro- far, is the only bright spot among the clinical duced by hyperactive B cells that recognize a lupus trials in recent years. By the end of 2010, plethora of nuclear such as DNA, Ro, we may witness the approval of belimumab as a La, Sm and ribosomal P protein. The produc- new drug for SLE. The prospect of a new lupus tion of pathogenic autoantibodies is not the only drug heralds an exciting time for rheumatolo- mechanism by which autoreactive B cells induce gists and holds the promise for development of and perpetuate inflammation in SLE. B cells novel B‑cell-directed therapies for patients with also secrete proinflammatory cytokines, increase SLE. Well-designed clinical trials focused on costimulation and activate autoreactive T cells. specific clearly defined SLE subsets are needed Furthermore, lupus B cells are characterized by to better understand the therapeutic potential a variety of abnormalities including increased of B‑cell depleting and anticytokine therapies calcium influx upon BCR engagement, reduced in lupus. levels of Lyn kinase in lipid rafts and decreased expression of FcgRIIB in IgM-CD27+ memory Future perspective cells. The array of autoreactive B‑cell functions Despite multiple studies exploring the patho- implicated in the pathogenesis of SLE has led genesis of SLE, researchers have been unable to to the emergence of B‑cell-directed therapeutic exploit this knowledge to develop new therapies approaches for lupus. for lupus for more than 40 years. In that same Over the past decade, the growing interest in vein, the majority of recent randomized clinical B cells evolved after open-label studies produced trials with B‑cell-directed therapies have also promising results. The RCTs have inspired even yielded negative results. However, many lessons more curiosity, despite the fact that the majority can be learnt from the failed clinical studies. of them did not reach their end points. It has been Flaws in trial designs, end points and/or the suggested that in the rituximab and abatacept background immunosuppressive therapies have RCTs, the heterogeneous nature of SLE created been considered as the possible reasons for the a challenge for successful results since one organ poor success of these trials. Patient stratifica- system may improve while another worsens. tion based on characteristics such as genetic fac- Many of the lupus researchers believe that tors, specific organ involvement, disease stage, the failure of EXPLORER and LUNAR studies autoantibody patterns or approximate severity with rituximab was more attributable to flaws in scores may also help develop focused clinical the study design than to the ineffectiveness of trials with a better chance for success. Within the drug [21]. Multiple factors related to the end the coming decade, clinical trials involving points, patient population and background anti- B‑cell-depleting and cytokine-blocking thera- inflammatory therapy have been considered as pies are expected to result in novel treatments possible contributors to the failure of these stud- for SLE patients whose disease manifestations ies. The use of high doses of oral glucocorticoids are refractory to the current standard of care and other immunosuppressants such as myco- management. phenolate mofetil has been heavily scrutinized since these background therapies may already Financial & competing interests disclosure have been aggressive enough to treat active SLE E Peeva participated as an investigator in the epratuzumab without rituximab. The heterogeneity of the dis- SL0003 and SL0004 studies sponsored by Immunomedics ease relating to genetic differences among lupus and UCB. The authors have no other relevant affiliations patients, differences in disease activity, specific or financial involvement with any organization or entity organ system involvement, particular symptoms with a financial interest in or financial conflict with the and duration of the symptoms, and the pres- subject matter or materials discussed in the manuscript ence or absence of serological parameters have apart from those disclosed. also been considered as factors that could have No writing assistance was utilized in the production of affected the results of these studies. this manuscript.

future science group www.futuremedicine.com 633 Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

Executive summary ƒƒ Over the past decade B cells have become a major therapeutic target in lupus. Open-label studies, randomized controlled trials and registries on B‑cell-directed treatments have provided inconsistent data. ƒƒ Complete B‑cell depletion with the chimeric anti-CD20 monoclonal antibody rituximab in systemic lupus erythematosus (SLE) did not meet the primary and secondary end points in the Exploratory Phase II/III SLE Evaluation of Rituximab (EXPLORER) and Lupus Nephritis Assessment with Rituximab (LUNAR) trials, but the French registry Autoimmunity and Rituximab (AIR) showed good clinical efficacy of rituximab in lupus reopening the debate on clinical benefit of anti-CD20 therapy in SLE. Studies with epratuzumab, a humanized anti- CD22 monoclonal antibody, demonstrated clinically meaningful improvements in patients with SLE. ƒƒ A clinical trial evaluating the costimulation blocker abatacept in extrarenal lupus did not meet its primary end point, but another trial is currently recruiting patients to investigate the effects of abatacept in lupus nephritis. ƒƒ B‑cell-stimulating cytokines have also been targeted. Belimumab, a monoclonal antibody directed to a B‑lymphocyte stimulator, decreased lupus activity as measured by SLE responder index, and may soon receive US FDA approval to treat SLE. Atacicept, a transmembrane activator and calcium modulator and cyclophilin-ligand interactor immunoglobulin, which targets cytokines B‑lymphocyte stimulator and APRIL, has been shown to reduce immunoglobulin levels and B‑cell numbers, and currently there is an ongoing trial with atacicept recruiting SLE patients with disease flares. IL‑6 and ‑10 are also therapeutic targets in SLE. An open-label Phase I pilot study with tocilizumab, a humanized monoclonal antibody against the IL‑6 receptor, demonstrated beneficial therapeutic effects in lupus patients with moderately active disease. ƒƒ Although some of the randomized controlled trials with B‑cell-directed therapies have yielded disappointing results, they have provided valuable information. Applying the lessons learnt from these trials can help design better clinical studies in SLE.

Bibliography 9 Taylor RP, Lindorfer MA: Immuno­ 17 Merrill JT, Neuwelt CM, Wallace DJ et al.: Papers of special note have been highlighted as: therapeutic mechanisms of anti-CD20 Efficacy and safety of rituximab in n of interest monoclonal antibodies. Curr. Opin. Immunol. moderately-to-severely active systemic lupus nn of considerable interest 20(4), 444–449 (2008). erythematosus: the randomized, double- blind, Phase II/III systemic lupus 1 Borchers AT, Naguwa SM, Shoenfeld Y, 10 Lui NL, Thumboo J, Fong KY: A case of erythematosus evaluation of rituximab Gershwin ME: The geoepidemiology of refractory vasculitic ulcers in a systemic trial. Arthritis Rheum. 62(1), 222–233 systemic lupus erythematosus. Autoimmun. lupus erythematosus patient responding (2010). Rev. 9(5), A277–A287 (2010). to rituximab and hyperbaric oxygen therapy. Int. J. Rheum. Dis. 12(4), 366–369 nn Exploratory Phase II/III SLE Evaluation 2 Ahmed S, Anolik JH: B-cell biology and (2009). related therapies in systemic lupus of Rituximab (EXPLORER), a Phase III erythematosus. Rheum. Dis. Clin. North Am. 11 Benham H, Garske L, Vecchio P, Eckert BW: study, found no differences between 36(1), 109–130, viii–ix (2010). Successful treatment of shrinking lung placebo and rituximab in the primary and syndrome with rituximab in a patient with secondary end points in systemic lupus 3 Mok MY: The immunological basis of systemic lupus erythematosus. J. Clin. B-cell therapy in systemic lupus erythematosus (SLE) patients who were Rheumatol. 16(2), 68–70 (2010). erythematosus. Int. J. Rheum. Dis. 13(1), treated with aggressive background 3–11 (2010). 12 Niaz FA, Aleem A: Response to rituximab in immunosuppressive therapy. a refractory case of thrombotic 4 Hostmann A, Jacobi AM, Mei H, Hiepe F, 18 Sanz I, Lee FE: B cells as therapeutic targets thrombocytopenic purpura associated with Dorner T: Peripheral B cell abnormalities and in SLE. Nat. Rev. Rheumatol. 6(6), 326–337 systemic lupus erythematosus. Saudi J. Kidney disease activity in systemic lupus (2010). Dis. Transpl. 21(1), 109–112 (2010). erythematosus. Lupus 17(12), 1064–1069 19 Furie R, Looney RJ, Rovin B: Efficacy and (2008). 13 Hickman RA, Denniston AK, Yee CS, safety of rituximab in subjects with active Toescu V, Murray PI, Gordon C: Bilateral 5 Tan EM: Antinuclear antibodies: diagnostic proliferative lupus nephritis (LN): results retinal vasculitis in a patient with systemic markers for autoimmune diseases and probes from the randomized, double-blind Phase III lupus erythematosus and its remission with for cell biology. Adv. Immunol. 44, 93–151 lunar study. Presented at: Program and rituximab therapy. Lupus 19(3), 327–329 (1989). Abstracts of the American College of (2010). Rheumatology Scientific Meeting. PA, USA, 6 Jacob N, Stohl W: Autoantibody-dependent 14 Oh JS, Kim YG, Lee SG, Lee CK, Yoo B: 17–21 October 2009. and autoantibody-independent roles for B Successful treatment of recurrent lupus cells in systemic lupus erythematosus: past, 20 Bruce IN: Re-evaluation of biologic therapies enteritis with rituximab. Lupus 19(2), present, and future. Autoimmunity 43(1), in systemic lupus erythematosus. Curr. Opin. 220–222 (2010). 84–97 (2010). Rheumatol. 22(3), 273–277 (2010). 15 Leandro MJ, Edwards JC, Cambridge G, 7 Jenks SA, Sanz I: Altered B cell receptor 21 Favas C, Isenberg DA: B-cell-depletion Ehrenstein MR, Isenberg DA: An open study signaling in human systemic lupus therapy in SLE – what are the current of B lymphocyte depletion in systemic lupus erythematosus. Autoimmun. Rev. 8(3), prospects for its acceptance? Nat. Rev. erythematosus. Arthritis Rheum. 46(10), 209–213 (2009). Rheumatol. 5(12), 711–716 (2009). 2673–2677 (2002). 8 Merrill JT, Erkan D, Buyon JP: Challenges in 22 Anolik JH, Campbell D, Felgar RE et al.: 16 Tanaka Y, Yamamoto K, Takeuchi T et al.: bringing the bench to bedside in drug The relationship of FcgRIIIa genotype to A multicenter Phase I/II trial of rituximab for development for sle. Nat. Rev. Drug Discov. degree of B cell depletion by rituximab in the refractory systemic lupus erythematosus. 3(12), 1036–1046 (2004). treatment of systemic lupus erythematosus. Mod. Rheumatol. 17(3), 191–197 (2007). Arthritis Rheum. 48(2), 455–459 (2003).

634 Int. J. Clin. Rheumatol. (2010) 5(6) future science group Review Tieng, Zandman-Goddard & Peeva Targeting B cells to treat systemic lupus erythematosus Review

23 Sutter JA, Kwan-Morley J, Dunham J 33 Poe JC, Fujimoto M, Jansen PJ, Miller AS, belimumab in patients with active systemic et al.: A longitudinal ana­lysis of SLE Tedder TF: CD22 forms a quaternary lupus erythematosus. Arthritis Rheum. 61(9), patients treated with rituximab (anti-CD20): complex with SHIP, Grb2, and Shc. 1168–1178 (2009). factors associated with B lymphocyte A pathway for regulation of B lymphocyte 43 Wallace DJ: New therapies in systemic lupus recovery. Clin. Immunol. 126(3), 282–290 antigen receptor-induced calcium flux. J. Biol. erythematosus – trials, troubles and (2008). Chem. 275(23), 17420–17427 (2000). tribulations – working towards a solution: 24 Chan O, Shlomchik MJ: A new role for 34 Carnahan J, Wang P, Kendall R et al.: Part 2 – the politically incorrect version. B cells in systemic autoimmunity: B cells Epratuzumab, a humanized monoclonal Lupus 18(2), 101–103 (2009). promote spontaneous T cell activation in antibody targeting CD22: characterization of 44 Navarra S, Guzman R, Gallacher A, Levy R, MRL-lpr/lpr mice. J. Immunol. 160(1), 51–59 in vitro properties. Clin. Cancer Res. 9(10 Pt Li E, Thomas M: Belimumab, a BLYS- (1998). 2), 3982S–3990S (2003). specific inhibitor, reduced disease activity, n A set of candidate biomarkers for 35 Epratuzumab demonstrates clinically flares and prednisone use in patients with lymphocyte monitoring in rituximab-treated meaningful improvements in patients with active SLE: efficacy and safety results from patients with autoimmune disease moderate to severe systemic lupus the Phase 3 BLISS-52 study. Presented at: is described. erythematosus (SLE): results from Program and abstracts of the American College EMBLEM™, a Phase IIb study. Presented at: of Rheumatology Scientific Meeting. PA, USA 25 Chan OT, Hannum LG, Haberman AM, EULAR. Rome, Italy, 16–19 June 2010 17–21 October, 2009. Madaio MP, Shlomchik MJ: A novel mouse (Poster SAT0212). with B cells but lacking serum antibody nn In a Phase III clinical trial, belimumab reveals an antibody-independent role for nn A Phase IIb study with the anti-CD22 demonstrated therapeutic effect in SLE B cells in murine lupus. J. Exp. Med. 189(10), monoclonal antibody epratuzumab patients with active disease. 1639–1648 (1999). provided benefits in SLE patients with 45 Stohl W, Merrill JT: Phase 1a single- and 26 Terrier B, Amoura Z, Ravaud P et al.: Safety moderate-to-severe disease. Phase 1b multiple-dose studies of AMG 623 and efficacy of rituximab in systemic lupus 36 Merrill JT, Burgos-Vargas R, Westhovens R (an anti-BAFF peptibody) in systemic lupus erythematosus: results from 136 patients from et al.: The efficacy and safety of abatacept in erythematosus (SLE). Arthritis Rheum. (58), the french AIR registry. Arthritis Rheum. patients with non-life-threatening S565 (2008). 62(8), 2458–2466 (2010). manifestations of SLE: results of a 12‑month 46 Mackay F, Ambrose C: The TNF family 27 Harris HE: Progressive multifocal exploratory study. Arthritis Rheum. 62(10), members BAFF and APRIL: the growing leucoencephalopathy in a patient with 3077–3087 (2010). complexity. Cytokine Growth Factor Rev. systemic lupus erythematosus treated with nn The primary and secondary end points 14(3–4), 311–324 (2003). rituximab. Rheumatology (Oxford) 47(2), were not met in the Phase IIb study 47 Planelles L, Medema JP, Hahne M, 224–225 (2008). with abatacept, but improvements in Hardenberg G: The expanding role of APRIL 28 Molloy ES, Calabrese LH: Progressive several exploratory measures suggest in cancer and immunity. Curr. Mol. Med. multifocal leukoencephalopathy: a national some efficacy in SLE patients with 8(8), 829–844 (2008). estimate of frequency in systemic lupus non-life-threatening disease. 48 Roschke V, Sosnovtseva S, Ward CD et al.: erythematosus and other rheumatic 37 Coyle AJ, Lehar S, Lloyd C et al.: The BLyS and APRIL form biologically active diseases. Arthritis Rheum. 60(12), 3761–3765 CD28-related molecule ICOS is required for heterotrimers that are expressed in patients (2009). effective T cell-dependent immune responses. with systemic immune-based rheumatic 29 Hutas G: Ocrelizumab, a humanized Immunity 13(1), 95–105 (2000). diseases. J. Immunol. 169(8), 4314–4321 monoclonal antibody against CD20 for (2002). 38 Yoshinaga SK, Whoriskey JS, Khare SD et al.: inflammatory disorders and B-cell T-cell co-stimulation through B7RP-1 and 49 Stohl W: Systemic lupus erythematosus and malignancies. Curr. Opin. Investig. Drugs ICOS. Nature 402(6763), 827–832 (1999). its ABCs (APRIL/BLyS complexes). Arthritis 9(11), 1206–1215 (2008). Res. Ther. 12(2), 111 (2010). 39 Cancro MP, D’cruz DP, Khamashta MA: 30 Mack GS: CD20 blockers eye crowded The role of B lymphocyte stimulator (BLyS) 50 Pena-Rossi C, Nasonov E, Stanislav M et al.: rheumatology market. Nat. Biotechnol. in systemic lupus erythematosus. J. Clin. An exploratory dose-escalating study 26(10), 1053–1054 (2008). Invest. 119(5), 1066–1073 (2009). investigating the safety, tolerability, 31 Fleischmann R, Cohen S, Pardo P : pharmacokinetics and pharmacodynamics of et al. 40 Petri M, Stohl W, Chatham W et al.: intravenous atacicept in patients with systemic B cell depletion in subjections with controlled Association of plasma B lymphocyte lupus erythematosus. Lupus 18(6), 547–555 systemic lupus erythematosus (SLE) after stimulator levels and disease activity in (2009). intravenous or subcutaneous administration systemic lupus erythematosus. Arthritis of SBI-087. Ann. Rheum. Dis. 69(Suppl. 3), Rheum. 58(8), 2453–2459 (2008). 51 Dall’Era M, Chakravarty E, Wallace D 550 (2010). et al.: Reduced B lymphocyte and 41 Furie R, Stohl W, Ginzler EM et al.: n immunoglobulin levels after atacicept Effects of a novel subcutaneous Biologic activity and safety of belimumab, a treatment in patients with systemic lupus anti-CD20 treatment on B cells in neutralizing anti-B-lymphocyte stimulator erythematosus: results of a multicenter, SLE patients with controlled disease (BLYS) monoclonal antibody: a Phase I trial Phase IB, double-blind, placebo-controlled, are described. in patients with systemic lupus dose-escalating trial. Arthritis Rheum. 56(12), 32 Sato S, Tuscano JM, Inaoki M, Tedder TF: erythematosus. Arthritis Res. Ther. 10(5), 4142–4150 (2007). CD22 negatively and positively regulates R109 (2008). 52 Dall’Era M, Wofsy D: Systemic lupus signal transduction through the B lymphocyte 42 Wallace DJ, Stohl W, Furie RA et al.: erythematosus clinical trials – an interim antigen receptor. Semin. Immunol. 10(4), A Phase II, randomized, double-blind, ana­lysis. Nat. Rev. Rheumatol. 5(6), 348–351 287–297 (1998). placebo-controlled, dose-ranging study of (2009).

future science group www.futuremedicine.com 635 Review Tieng, Zandman-Goddard & Peeva

53 Mellor-Pita S, Citores MJ, Castejon R et al.: 57 Llorente L, Zou W, Levy Y et al.: Role of 103 Clinical Trials Monocytes and T lymphocytes contribute to a 10 in the B lymphocyte http://clinicaltrials.gov/ct2/show/ predominance of interleukin‑6 and hyperactivity and autoantibody production NCT00714116?term=SBI- interleukin‑10 in systemic lupus of human systemic lupus erythematosus. 087+AND+lupus&rank=1 erythematosus. Cytometry B Clin. Cytom. J. Exp. Med. 181(3), 839–844 (1995). 104 Clinical Trials 76B(4), 261–270 (2009). 58 Uhm WS, Na K, Song GW et al.: Cytokine http://clinicaltrials.gov/ct2/show/NCT00774 54 Kitani A, Hara M, Hirose T et al.: balance in kidney tissue from lupus nephritis 852?term=abatacept+AND+cyclophosphami Autostimulatory effects of IL-6 on excessive patients. Rheumatology (Oxford) 42(8), de+AND+lupus&rank=1 B cell differentiation in patients with systemic 935–938 (2003). 105 Clinical Trials lupus erythematosus: analysis of IL-6 59 Llorente L, Richaud-Patin Y, García-Padilla C http://clinicaltrials.gov/ct2/show/ production and IL-6R expression. Clin. Exp. et al.: Clinical and biologic effects of NCT00774943?term=AMG+557&rank=1 Immunol. 88(1), 75–83 (1992). anti-interleukin-10 monoclonal antibody 106 Human Genome Sciences and 55 Nagafuchi H, Suzuki N, Mizushima Y, administration in systemic lupus GlaxoSmithKline announce positive results in Sakane T: Constitutive expression of IL-6 erythematosus. Arthritis Rheum. 43(8), second of two Phase 3 trials of Benlysta™ in receptors and their role in the excessive B cell 1790–1800 (2000). systemic lupus erythematosus (April, 2010) function in patients with systemic lupus www.gsk.com/media/ erythematosus. J. Immunol. 151(11), pressreleases/2010/2010_pressrelease_10039. 6525–6534 (1993). „ „Websites htm 56 Illei GG, Shirota Y, Yarboro CH et al.: 101 Clinical Trials 107 Clinical Trials Tocilizumab in systemic lupus erythematosus: http://clinicaltrials.gov/ct2/show/NCT00539 http://clinicaltrials.gov/ct2/show/NCT0062 data on safety, preliminary efficacy, and 838?term=ocrelizumab+AND+lupus&ra 4338?term=atacicept+AND+lupus&rank=2 impact on circulating plasma cells from an nk=2 open-label Phase I dosage-escalation study. 102 Clinical Trials Arthritis Rheum. 62(2), 542–552 (2010). http://clinicaltrials.gov/ct2/show/NCT00626 nn In a Phase I study, SLE patients show 197?term=ocrelizumab+AND+lupus&ra promising clinical and serologic responses nk=1 to tocilizumab.

636 Int. J. Clin. Rheumatol. (2010) 5(6) future science group