Treatment of lupus patients with a tolerogenic peptide, hCDR1 (Edratide): Immunomodulation of gene expression

https://doi.org/10.1016/j.jaut.2009.03.009Get rights and content

Abstract

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulation of cytokines, apoptosis, and B- and T-cell functions. The tolerogenic peptide, hCDR1 (Edratide), ameliorated the clinical manifestations of murine lupus via down-regulation of pro-inflammatory cytokines and apoptosis, up-regulation of the immunosuppressive cytokine TGF-β, and the induction of regulatory T-cells. In the present study, gene expression was determined in peripheral blood mononuclear cells of 9 lupus patients that were treated for 26 weeks with either hCDR1 (five patients), or placebo (four patients). Disease activity was assessed by SLEDAI-2K and the BILAG scores. Treatment with hCDR1 significantly down-regulated the mRNA expression of the pathogenic cytokines IL-1β, TNF-α, IFN-γ, and IL-10, of BLyS (B-lymphocyte stimulator) and of the pro-apoptotic molecules caspase-3 and caspase-8. In contrast, the treatment up-regulated in vivo gene expression of both TGF-β and FoxP3. Furthermore, hCDR1 treatment resulted in a significant decrease in SLEDAI-2K (from 8.0 ± 2.45 to 4.4 ± 1.67; P = 0.02) and BILAG (from 8.2 ± 2.7 to 3.6 ± 2.9; P = 0.03) scores. Thus, the tolerogenic peptide hCDR1, immunomodulates, in vivo, the expression of genes that play a role in SLE, consequently restoring the global immune dysregulation of lupus patients. Hence, hCDR1 has a potential role as a novel disease-specific treatment for lupus patients.

Introduction

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of autoantibodies and by impaired functions of B- and T-cells, accompanied by systemic clinical manifestations [1], [2]. Previous studies demonstrated the role of cytokines [3], [4], apoptosis [5], [6], and dysfunction of regulatory T-cells [7], [8] in the pathogenesis of murine and human SLE. For a specific treatment of SLE, our laboratory designed a peptide, designated hCDR1 [9], which is based on the sequence of the complementarity-determining region (CDR)1 of a human anti-DNA monoclonal antibody that bears the major idiotype designated 16/6Id [10], [11]. hCDR1 was shown to ameliorate the serological and clinical manifestations of induced or spontaneously developed lupus in mice [12]. The beneficial effects of hCDR1 were associated with the down-regulation of the pathogenic cytokines IL-1β, IFN-γ, and IL-10 [12], and the up-regulation of the immunosuppressive cytokine TGF-β [13]. Treatment with hCDR1 also diminished significantly the production of the B-cell stimulator (BLyS, BAFF) in lupus-prone mice [14]. Furthermore, treatment of SLE-afflicted mice with hCDR1 resulted in reduced apoptosis rates of T-cells [15], [16], [17], and in specific induction of CD4+CD25+Foxp3+ regulatory cells [13], [18].

We have previously shown that hCDR1 down-regulated, in vitro, autoreactive T-cell responses of peripheral blood mononuclear cells (PBMC) of SLE patients in association with an increased production of TGF-β [9]. Moreover, we established a human model of SLE by transferring PBMC of lupus patients into severe combined immunodeficient mice [19]. Treatment with hCDR1 ameliorated the serological (human anti-DNA antibodies) and clinical lupus-related manifestations in the latter model of human SLE [20]. Furthermore, we have recently shown that in vitro incubation of PBMC of lupus patients with hCDR1, but not with a control peptide, down-regulated gene expression of IL-1β, TNF-α, IFN-γ, IL-10, and caspase-3 and up-regulated the expression of TGF-β, FoxP3, the anti-apoptotic molecule Bcl-xL and of the negative regulators Foxj1 and Foxo3a [21]. In addition, the in vitro incubation with hCDR1 resulted in an increase of CD4+CD25+FoxP3+ functional regulatory T-cells [21].

It has been of great interest to determine whether hCDR1 functions via similar mechanisms of actions when administered to SLE patients. The present study was therefore aimed at determining the in vivo effects of hCDR1 on human SLE. To this end, blood samples were taken from 9 lupus patients that were treated with hCDR1 or with the vehicle for a period of 26 weeks. Gene expression of the pathogenic cytokines (IL-1β, TNF-α, IFN-γ, and IL-10), pro-apoptotic molecules (caspase-3, caspase-8), BLyS, and immunoregulatory TGF-β and FoxP3 molecules was determined by real-time RT-PCR. The results indicate that hCDR1 immunomodulated in vivo the expression of the above genes in SLE patients in a manner similar to that demonstrated for SLE-afflicted mice.

Section snippets

Patients

The data presented here are of nine SLE patients from two Israeli Medical Centers (Kaplan, Rehovot; Rabin, Petah-Tikva). These patients participated in a large double-blinded clinical trial. Included are all patients from the two medical centers who completed the study and from whom blood samples were taken at least twice (before treatment initiation and at week 24) for mRNA preparation. We present here the effect of treatment with hCDR1 on cytokine, apoptosis, BLyS, and regulatory gene

Patients

The mean (±SD) age of our nine patients (one male, eight females) at the time of the study was 46.4 ± 10.5 (range 29–61) years. All had antinuclear antibodies and anti-dsDNA autoantibodies in their sera at study entry. Hypocomplementemia (either C3 or C4) was observed in five patients. The main SLE-related clinical manifestations at study entry were skin involvement (6/9), arthritis (5/9), alopecia (4/9), thrombocytopenia and lymphopenia (2/9), mucosal ulcers (2/9), and pleuritis (1/9). The mean

Discussion

The main finding of the present study is that the tolerogenic peptide hCDR1 is capable of suppressing, in vivo, gene expression of pro-inflammatory cytokines, apoptosis, and of B-cell stimulation. Furthermore, in vivo treatment with hCDR1 up-regulated immunosuppressive and regulatory molecules in PBMC of SLE patients. To the best of our knowledge, this is the first report demonstrating in vivo immunomodulation of gene expression in SLE patients by a specific peptide previously shown to

Acknowledgement

The clinical trial was conducted by Teva Pharmaceutical Industries, Ltd., and the blood samples used in our study were of patients that participated in this trial. However, the study reported here was performed independently at the Weizmann Institute of Science.

References (38)

  • S. Sakaguchi

    Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses

    Annu Rev Immunol

    (2004)
  • S. Paust et al.

    Regulatory T cells and autoimmune disease

    Immunol Rev

    (2005)
  • Z.M. Sthoeger et al.

    Modulation of autoreactive responses of peripheral blood lymphocytes of patients with systemic lupus erythematosus 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)
  • M. Blank et al.

    The story of the 16/6 idiotype and systemic lupus erythematosus

    Isr Med Assoc J

    (2008)
  • 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)
  • A. Sharabi et al.

    A peptide based on the complementarity-determining region 1 of an autoantibody ameliorates lupus by up-regulating CD4+CD25+ cells and TGF-beta

    Proc Natl Acad Sci U S A

    (2006)
  • Parameswaran R, David HB, Sharabi A, Zinger H, Mozes E. B-cell activating factor (BAFF) plays a role in the mechanism...
  • A. Sharabi et al.

    The role of apoptosis in the ameliorating effects of a CDR1-based peptide on lupus manifestations in a mouse model

    J Immunol

    (2007)
  • Cited by (51)

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

      2018, Clinical Immunology
      Citation Excerpt :

      We have further determined the expression levels of the IDO gene in lupus patients that were treated with hCDR1 (5) or vehicle (4) in a Phase II clinical trial by weekly subcutaneous injections [20]. The inclusion and exclusion criteria, characterization of patients and clinical outcome were described previously [20]. Blood samples were collected from the patients in PAXgene tubes prior and following 24 weeks of treatment for the preparation of mRNA.

    • 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 :

      Furthermore, a randomized double-blind placebo-controlled trial indicating that IL-1β inhibition affected favorably fatigue in patients with pSS [42] and studies showing the role of TNF-α in the NOD model of SS [43] suggest the involvement of both cytokines in the pathogenesis of pSS. We previously demonstrated that hCDR1 significantly down-regulated the levels of the BLyS (BAFF), that was shown to play a role in the pathogenesis of lupus, in both murine models and patients with SLE [27,30,34]. Elevated levels of circulating BLyS as well as an up-regulation of BLyS expression in inflamed salivary glands were reported in pSS patients and were shown to correlate with the elevated secretion of autoantibodies such as anti-Ro (SS-A) and anti-La (SS-B), known hallmarks of the disease [16,18].

    • Autoantigenesis and Antigen-Based Therapy and Vaccination in SLE

      2013, Dubois' Lupus Erythematosus and Related Syndromes: Eighth Edition
    View all citing articles on Scopus
    View full text