Elsevier

Journal of Autoimmunity

Volume 54, November 2014, Pages 60-71
Journal of Autoimmunity

Review
Novel approaches to the development of targeted therapeutic agents for systemic lupus erythematosus

https://doi.org/10.1016/j.jaut.2014.06.002Get rights and content

Abstract

Systemic lupus erythematosus (SLE) is a chronic multisystem disease in which various cell types and immunological pathways are dysregulated. Current therapies for SLE are based mainly on the use of non-specific immunosuppressive drugs that cause serious side effects. There is, therefore, an unmet need for novel therapeutic means with improved efficacy and lower toxicity. Based on recent better understanding of the pathogenesis of SLE, targeted biological therapies are under different stages of development. The latter include B-cell targeted treatments, agents directed against the B lymphocyte stimulator (BLyS), inhibitors of T cell activation as well as cytokine blocking means. Out of the latter, Belimumab was the first drug approved by the FDA for the treatment of SLE patients. In addition to the non-antigen specific agents that may affect the normal immune system as well, SLE-specific therapeutic means are under development. These are synthetic peptides (e.g. pConsensus, nucleosomal peptides, P140 and hCDR1) that are sequences of conserved regions of molecules involved in the pathogenesis of lupus. The peptides are tolerogenic T-cell epitopes that immunomodulate only cell types and pathways that play a role in the pathogenesis of SLE without interfering with normal immune functions. Two of the peptides (P140 and hCDR1) were tested in clinical trials and were reported to be safe and well tolerated. Thus, synthetic peptides are attractive potential means for the specific treatment of lupus patients. In this review we discuss the various biological treatments that have been developed for lupus with a special focus on the tolerogenic peptides.

Introduction

Systemic lupus erythematosus (SLE, lupus) is a chronic multisystem disease of unknown etiology [1], [2]. It affects all races though it is more common among African-Americans, Hispanics and Asians [1]. SLE is much more prevalent in females as compared to males (9:1 ratio) [2]. The prevalence of SLE is about 1:1000 females but it is increasing constantly due to both, a better early diagnosis and a better survival of lupus patients [1], [2]. Dysregulation of both, the innate and the adaptive immune systems play a role in SLE with the production of variety of autoantibodies and pro-inflammatory cytokines, impaired T cell function and enhanced apoptosis [1], [2], [3]. The clinical spectrum of lupus varies, ranging from mild mucocutaneous/musculoskeletal manifestations to life-threatening renal or neurologic disease [1], [2]. Disease activity and lupus organ involvement fluctuate along the time with flares and remissions (either spontaneous or induced by treatment) [1], [2]. Although the precise etiology of SLE is not fully defined yet, genetic, hormonal and environmental factors appear to play a role in the pathogenesis and course of the disease [3], [4].

The current "standard" treatment of SLE includes anti-malarial agents (mainly Hydroxychloroquine, HCQ), corticosteroids, immunoglobulins (IVIG) and cytotoxic immunosuppressive agents [1], [2], [5]. HCQ was shown to be effective in the treatment of mild to moderate mucocutaneous and musculoskeletal manifestations of lupus [5]. Corticosteroids, given orally or intravenously, are effective for almost all lupus related manifestations. However, the long-term adverse effects of the latter agents limit their usage. Recently, Thamer et al. demonstrated that a dose of as low as 6 mg prednisone per day increases the corticosteroids-induced organ damage by 50% [6]. Thus, a steroid sparing therapeutic approach is mandatory [7]. Other immunosuppressive agents such as Cyclophosphamide, Azathioprine, Methotrexate and Mycophenolate Mofetil were shown to be effective in the treatment of moderate to severe lupus manifestations (e.g. renal or CNS involvement) but these agents also have significant short and long term adverse effects [7]. Moreover, although the above treatment modalities are quite effective they are not specific for lupus and the control of disease activity with those agents remains suboptimal. Thus, in spite of treatment, lupus patients have active lupus related flares in substantial fractions of their life [7]. Therefore, there is an unmet need for alternative nontoxic effective and more lupus specific therapeutic approaches.

Based on the knowledge of the different dysregulated innate and adaptive immunological pathways involved in the pathogenesis of SLE, attempts have been made to develop biological therapies against targets that play a role in lupus. The various therapeutic means are at different stages of development. In general, they could be divided into non-specific and SLE specific means. The non-specific approaches could be further categorized as cell depleting agents and immunomodulatory means. In the present article we review the various biological therapeutic agents with a special focus on lupus antigen-specific therapeutic means.

Section snippets

B-cell targeted treatments

B cell activation with excess generation of immunoglobulins and autoantibodies is the hallmark of SLE [1], [2], [8]. Therefore, biological agents that target B cells and reduce their activity have been developed as potential candidates for the treatment of lupus [8].

BLyS targeted therapy

B lymphocyte stimulator (BLyS), also known as B cell activating factor (BAFF), is a 285 amino acid transmembrane protein (expressed on the surface of monocytes, macrophages, dendritic and activated T cells) that belongs to the tumor necrosis factor ligand superfamily [24], [25], [26]. Following a cleavage by purin protease, soluble active BLyS (17Kd protein) is released into the circulation [26]. The soluble BLyS can bind to three receptors (BCMR – B cell maturation antigen, TACI –

Inhibition of T-cell activation

The activation of T cells requires at least two independent signals. The first signal results from the engagement of the MHC complex and the antigen with the T cell receptor. The second activation signal is an antigen-independent event which involves an interaction of co-stimulatory molecules such as CD28 (expressed on T cells) and CD80/CD86 (expressed on antigen presenting cells such as DC, macrophages or B cells). Another co-stimulatory pathway is the interaction between CD40 (on B cells) and

Tocilizumab (anti IL-6 receptor mAb)

IL-6 is a multifactorial pro-inflammatory cytokine that was shown to play a role in the pathogenesis and treatment of murine lupus [52], [53]. Moreover, elevated levels of IL-6 were found in sera of active lupus patients [54]. Tocilizumab (Fig. 1; 8) is a fully humanized mAb against the IL-6 receptor that prevents binding of IL-6 to both, membrane and soluble receptors. A small phase I trial (16 lupus patients) suggested that Tocilizumab is safe and beneficial in SLE [55]. Further controlled

Eculizumab (Anti-C5 mAb)

The early complement components are essential for immune complex clearance whereas activation of the late components were shown to be associated with lupus exacerbations [66]. Eculizumab is a humanized IgG2/4 mAb directed against the complement protein C5, blocking the formation of the complement terminal membrane attack complex [66], [67]. This mAb was shown to ameliorate murine lupus [67]. A phase I trial with Eculizumab in 24 lupus patients demonstrated good safety profile but no clear

Therapeutic approaches for the specific treatment of SLE

The rationale behind attempting the development of antigen specific therapy for lupus as well as for other autoimmune diseases is to achieve the mean that will modulate only the disease specific pathogenic cells and pathways and will leave the rest of the immune system intact. Thus, in contrast to the current standard of care for lupus that suppresses large portions of the immune system, and unlike many of the newly therapeutic approaches that introduce agents that deplete large parts of the

Conclusions

The major progress made in recent years towards the understanding of the pathogenesis and development of SLE resulted in attempts to develop novel biological agents for the treatment of the disease. In this review we discussed a variety of biological therapies that are under different stages of development. The latter target different cell populations and molecules either by their depletion or by blocking inflammatory processes. Indeed, one of these biological agents, namely, Belimumab, that

References (133)

  • A.E. Majeski et al.

    Mechanisms of chaperone-mediated autophagy

    Int J Biochem Cell Biol

    (2004)
  • A. Rahman et al.

    Systemic lupus erythematosus

    N Engl J Med

    (2008)
  • G.C. Tsokos

    Systemic lupus erythematosus

    N Engl J Med

    (2011)
  • D. Wofsy

    Recent progress in conventional and biologic therapy for systemic lupus erythematosus

    Ann Rheum Dis

    (2013)
  • M. Thamer et al.

    Prednisone, lupus activity, and permanent organ damage

    J Rheumatol

    (2009)
  • A. Lateef et al.

    Unmet medical needs in systemic lupus erythematosus

    Arthritis Res Ther

    (2012)
  • J.H. Anolik

    B cell biology: implications for treatment of systemic lupus erythematosus

    Lupus

    (2013)
  • P. Emery et al.

    The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial

    Arthritis Rheum

    (2006)
  • B. Duxbury et al.

    Rituximab in systemic lupus erythematosus: an updated systematic review and meta-analysis

    Lupus

    (2013)
  • R.J. Looney et al.

    B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose-escalation trial of rituximab

    Arthritis Rheum

    (2004)
  • T. Turner-Stokes et al.

    The efficacy of repeated treatment with B-cell depletion therapy in systemic lupus erythematosus: an evaluation

    Rheumatol Oxf

    (2011)
  • B. Terrier et al.

    Club Rhumatismes et Inflammation. Safety and efficacy of rituximab in systemic lupus erythematosus:results from 136 patients from the French AutoImmunity and Rituximab registry

    Arthritis Rheum

    (2010)
  • B.H. Rovin et al.

    Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study

    Arthritis Rheum

    (2012)
  • J.T. Merrill et al.

    Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial

    Arthritis Rheum

    (2010)
  • V. Reddy et al.

    B-cell depletion in SLE: clinical and trial experience with rituximab and ocrelizumab and implications for study design

    Arthritis Res Ther

    (2013)
  • G.K. Bertsias et al.

    European league against Rheumatism and European Renal Association-European Dialysis and Transplant Association. Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and paediatric lupus nephritis

    Ann Rheum Dis

    (2012)
  • M. Aringer et al.

    Current state of evidence on 'off-label' therapeutic options for systemic lupus erythematosus, including biological immunosuppressive agents, in Germany, Austria and Switzerland—a consensus report

    Lupus

    (2012)
  • D.J. Wallace et al.

    Epratuzumab for systemic lupus erythematosus

    Lupus

    (2013)
  • T. Dörner et al.

    Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus

    Arthritis Res Ther

    (2006)
  • D.J. Wallace et al.

    Efficacy and safety of epratuzumab in patients with moderate/severe active systemic lupus erythematosus: results from EMBLEM, a phase IIb, randomised, double-blind, placebo-controlled, multicentre study

    Ann Rheum Dis

    (2014)
  • W. Stohl

    Biologic differences between various inhibitors of the BLyS/BAFF pathway: should we expect differences between belimumab and other inhibitors in development?

    Curr Rheumatol Rep

    (2012)
  • P. Schneider et al.

    BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth

    J Exp Med

    (1999)
  • M. Batten et al.

    BAFF mediates survival of peripheral immature B lymphocytes

    J Exp Med

    (2000)
  • A.G. Rolink et al.

    BAFF is a survival and maturation factor for mouse B cells

    Eur J Immunol

    (2002)
  • M. Ota et al.

    Regulation of the B cell receptor repertoire and self-reactivity by BAFF

    J Immunol

    (2010)
  • F. Mackay et al.

    Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations

    J Exp Med

    (1999)
  • C.O. Jacob et al.

    Paucity of clinical disease despite serological autoimmunity and kidney pathology in lupus-prone New Zealand mixed 2328 mice deficient in BAFF

    J Immunol

    (2006)
  • M. Petri et al.

    Association of plasma B lymphocyte stimulator levels and disease activity in systemic lupus erythematosus

    Arthritis Rheum

    (2008)
  • G.S. Cheema et al.

    Elevated serum B lymphocyte stimulator levels in patients with systemic immune-based rheumatic diseases

    Arthritis Rheum

    (2001)
  • R. Furie et al.

    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)
  • D.J. Wallace et al.

    A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus

    Arthritis Rheum

    (2009)
  • R. Furie et al.

    A phase III, randomized,placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus

    Arthritis Rheum

    (2011)
  • B.H. Hahn

    Belimumab for systemic lupus erythematosus

    N Engl J Med

    (2013)
  • J.T. Merrill et al.

    Long-term safety profile of belimumab plus standard therapy in patients with systemic lupus erythematosus

    Arthritis Rheum

    (2012)
  • D.J. Wallace et al.

    BLISS-52 and -76, and LBSL02 Study Groups. Safety profile of belimumab: pooled data from placebo-controlled phase 2 and 3 studies in patients with systemic lupus erythematosus

    Lupus

    (2013)
  • J. Zhang et al.

    Cutting edge: a role for B lymphocyte stimulator in systemic lupus erythematosus

    J Immunol

    (2001)
  • M. Dall'Era et al.

    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)
  • E.M. Ginzler et al.

    Atacicept in combination with MMF and corticosteroids in lupus nephritis: results of a prematurely terminated trial

    Arthritis Res Ther

    (2012)
  • A.Y. Hoi et al.

    Abatacept in the treatment of lupus

    Expert Opin Biol Ther

    (2012)
  • B.K. Finck et al.

    Treatment of murine lupus with CTLA4Ig

    Science

    (1994)
  • Cited by (39)

    • Autoantibodies in systemic lupus erythematosus: From immunopathology to therapeutic target

      2022, Journal of Autoimmunity
      Citation Excerpt :

      The anti-Id antibodies could mimic the non-protein antigens that inhibit the antibody binding to the target antigen and suppress antibody response [132,133]. The CDR regions of the Id bearing antibodies have been extracted for the peptide synthesis, targeting the pathogenic antibody binding [134]. These peptides, including hCDR1 (GYYWSWIRQPPGKGEEWIG) and pConsensus (FIEWNKLRFRQGLEW), were shown to downregulate the production of autoantibodies and proinflammatory cytokines [130,135].

    • Indoleamine-2,3-dioxygenase in murine and human systemic lupus erythematosus: Down-regulation by the tolerogeneic peptide hCDR1

      2018, Clinical Immunology
      Citation Excerpt :

      As a result of the down regulation of those cytokines, gene expression of IDO (shown to be pro-inflammatory in lupus) decreases significantly. On the other hand, treatment with hCDR1 up-regulates TGF-β expression and production that leads to elevated expression of FOXP3 and, as shown previously, to the induction of functional regulatory T cells that play a crucial role in SLE disease amelioration [13,14,18,50]. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

    • The tolerogenic peptide hCDR1 immunomodulates cytokine and regulatory molecule gene expression in blood mononuclear cells of primary Sjogren's syndrome patients

      2018, Clinical Immunology
      Citation Excerpt :

      These latter effects were associated with clinical amelioration of the patients. Thus, hCDR1 reverses the global immune dysregulation in those lupus patients [34,33,27]. In view of the various immunomodulatory effects of hCDR1 in murine and human SLE and the potential similarity in the pathogenic mechanisms between pSS and SLE, we were interested in determining the potential beneficial effects of this peptide on pSS patients.

    • Structure-Function in Antibodies to Double-Stranded DNA

      2018, Structural Biology in Immunology: Structure and Function of Novel Molecules of Immunologic Importance
    View all citing articles on Scopus
    1

    Present address: Rheumatology Institute, Sourasky Medical Center, Tel-Aviv, Israel.

    View full text