Elsevier

Autoimmunity Reviews

Volume 10, Issue 1, November 2010, Pages 22-26
Autoimmunity Reviews

Review
A novel tolerogenic peptide, hCDR1, for the specific treatment of systemic lupus erythematosus

https://doi.org/10.1016/j.autrev.2010.07.004Get rights and content

Abstract

Treatment with a tolerogenic peptide, hCDR1, designed for the specific treatment of systemic lupus erythematosus (SLE) ameliorated the serological and kidney-related clinical manifestations in murine models of induced and spontaneous lupus. Furthermore, hCDR1 reduced brain pathology and improved behavior parameters in mice with central nervous system manifestations. The beneficial effects were associated with a diminished production of pathogenic cytokines (e.g. IFN-γ, IL-10, and IL-1β) and with increased production of the immunosuppressive cytokine, TGF-β. Treatment with hCDR1 up-regulated CD4 and CD8 regulatory T cells (Tregs) that played a key role in the ameliorating effects of hCDR1. Reduction of T cell apoptosis by hCDR1 contributed to the beneficial effects of hCDR. Moreover, treatment with hCDR1 down-regulated B cell maturation and autoreactive B cell survival by diminishing the B cell activating factor (BAFF/BLyS). Finally, hCDR1 suppressed in vivo gene expression of pathogenic cytokines, apoptosis and BLyS and up-regulated immunosuppressive molecules in peripheral blood lymphocytes of SLE patients. The latter was associated with clinical amelioration. Thus, treatment with hCDR1 leads to a cascade of events that culminate in the down-regulation of SLE-associated autoreactive T and B cells and in the clinical amelioration of lupus. hCDR1 is therefore a candidate for the specific treatment of SLE patients.

Introduction

Systemic lupus erythematosus (SLE) is a complex disease characterized by the generation of autoantibodies and clinical involvement of multiple organs. T and B cells play a role in the development of SLE. Current therapies for SLE are mainly based on the use of non-specific immunosuppressive drugs and have serious side effects [1]. In spite of intensive efforts, it has been more than 50 years since a new treatment for lupus has been approved. Thus there is an unmet need for novel therapeutic means that will be specific, with improved efficacy and lower toxicity than the currently available therapies for SLE.

As a potential candidate for the specific treatment of SLE patients, we have designed and synthesized a tolerogenic peptide, designated hCDR1 [2], that is based on the sequence of the heavy chain complementarity determining region (CDR)1 of a human monoclonal anti-DNA antibody [3]. hCDR1 was tested for its ability to ameliorate SLE manifestations in spontaneous and induced models of SLE in mice. Administration of hCDR1 (25–50 μg/mouse) subcutaneously [4], once a week for 10 weeks resulted in a significant amelioration of the serological (e.g. reduced autoantibody production) and renal (e.g. diminished proteinuria levels and decreased IgG and complement C3 complex depositions in the glomeruli) manifestations that developed either in the SLE-prone (NZB × NZW)F1 mice, in mice with experimental SLE induced with the anti-DNA autoantibody that bears the 16/6 idiotype (Id) [4], [5] or in severe combined immunodeficient (SCID) mice that were engrafted with peripheral blood mononuclear cells (PBMC) of SLE patients and developed an SLE-like disease [6]. More recently we showed that treatment with hCDR1 down-regulated disease manifestations associated with central nervous system (CNS) lupus, as evidenced by diminished brain pathology (e.g. cell infiltration, immune complex deposits and gliosis) and improved behavior parameters (e.g. anxiety and memory deficit) [7]. Thus, tolerogenic administration of low doses of hCDR1 ameliorated the serological, renal and neurological manifestations of SLE in murine models.

Section snippets

Cytokines

Cytokines have been shown to play an important role in the abnormal immune regulation observed in SLE-afflicted mice and in SLE patients [8], [9]. The beneficial effects of hCDR1 were associated with down-regulated expression and secretion of the proinflammatory and pathogenic cytokines interleukin (IL)-1β, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-10. In contrast, the immunosuppressive cytokine transforming growth factor (TGF)-β was up-regulated following treatment with hCDR1,

The effects of treatment with hCDR1 on the B cell compartment in mice with SLE

B cells play an important role in the pathogenesis of SLE. They produce autoantibodies that mediate tissue injury, they function as antigen presenting cells (APCs) that present epitopes of self antigens to autoreactive T cells, and they produce soluble mediators involved in the organization of lymphoid tissues and in the initiation and perpetuation of inflammatory processes [31], [32]. B cell activating factor (BAFF, also known as BLyS) is a member of the TNF superfamily that regulates B cell

The effects of treatment with hCDR1 on experimental SLE in pigs

We established a mild SLE-like disease in pigs by their immunization with a monoclonal anti-DNA autoantibody. The disease was manifested by the production of dsDNA and nuclear antigen-specific antibodies, by elevated levels of erythrocyte sedimentation rates and by immune complex depositions that were observed in the kidney sections [38]. Weekly treatment injections (for 10 weeks) with various doses of hCDR1 down-regulated the observed SLE-related manifestations. Furthermore, the treatment

In vitro effects of hCDR1 on gene expression and Tregs in PBMC of lupus patients

In view of the various immunomodulatory effects of hCDR1 in the murine models of lupus, it was of interest to determine the effects of the peptide on PBMC obtained from SLE patients. To this end PBMC of 11 lupus patients and 5 gender- and age-matched healthy controls were cultured with hCDR1 or the control scrambled peptide and gene expression as well as the presence of Tregs were assessed. hCDR1 down-regulated significantly IL-1β, IFN-γ and IL-10 gene expression. Furthermore, it up-regulated

Concluding remarks

The tolerogenic peptide, hCDR1, was demonstrated to ameliorate SLE-associated manifestations in a number of experimental models as well as in patients with lupus. Table 1 lists the SLE systems in which the beneficial effects of hCDR1 were studied. The mechanism of action of hCDR1 is demonstrated schematically in Fig. 1. In SLE, T and B cells are highly activated. As shown in Fig. 1, following treatment with hCDR1, two subsets of Tregs are induced, namely, CD8 and CD4 Tregs. The induction of CD8

Take-home messages

  • The ameliorative effects of hCDR1 on SLE were demonstrated in a number of animal models and in patients affected with the disease.

  • Treatment with the tolerogenic peptide, hCDR1, results in the inhibition of SLE-associated responses via the peptide-specific induction of CD8 and CD4 Tregs.

  • Induction of tolerance by hCDR1 results in the up-regulation of inhibitory/regulatory molecules and in better controlled signaling pathways.

  • Treatment with hCDR1 affects the T and B cell compartments.

  • Treatment of

References (40)

  • B.H. Hahn

    Overview of pathogenesis of systemic lupus erythematosus

  • Z.M. Sthoeger et al.

    Modulation of autoreactive responses of peripheral blood lymphocytes of patients with systemic lupus erythemathosus by peptides based on human and murine anti-DNA autoantibodies

    Clin Exp Immunol

    (2003)
  • A. Waisman et al.

    The pathogenic human monoclonal anti-DNA that induces experimental systemic lupus erythematosus in mice is encoded by a VH4 gene segment

    Int Immunol

    (1995)
  • D. Luger et al.

    A peptide based on the complementarity determining region 1 of a human monoclonal autoantibody ameliorates spontaneous and induced lupus manifestations in correlation with cytokine immunomodulation

    J Clin Immunol

    (2004)
  • M. Blank et al.

    The story of 16/6 idiotype and systemic lupus erythematosus

    Isr Med Assoc J

    (2008)
  • N. Mauermann et al.

    Amelioration of lupus manifestations by a peptide based on the complementarity determining region 1 of an autoantibody in severe combined immunodeficient (SCID) mice engrafted with peripheral blood lymphocytes of systemic lupus erythematosus (SLE) patients

    Clin Exp Immunol

    (2004)
  • S. Lapter et al.

    Amelioration of brain pathology and behavioral dysfunction of mice with systemic lupus erythematosus following treatment with a tolerogenic peptide

    Arthritis Rheum

    (2009)
  • D.A. Horowitz et al.

    The cytokine network in the pathogenesis of systemic lupus erythematosus and possible therapeutic implications

    Springer Semin Immunopathol

    (1994)
  • G.S. Dean et al.

    Cytokines and systemic lupus erythematosus

    Ann Rheum Dis

    (2000)
  • C. Haas et al.

    IFN-gamma receptor deletion prevents autoantibody production and glomerulonephritis in lupus-prone (NZBxNZW)F1 mice

    J Immunol

    (1998)
  • Cited by (0)

    View full text