Elsevier

Clinical Immunology

Volume 143, Issue 3, June 2012, Pages 210-221
Clinical Immunology

REVIEW
Emerging therapies for systemic lupus erythematosus — Focus on targeting interferon-alpha

https://doi.org/10.1016/j.clim.2012.03.005Get rights and content

Abstract

Current therapies for systemic lupus erythematosus (SLE), a debilitating, potentially lethal, multifactorial systemic autoimmune disease, are limited to suppressing disease activity and are associated with multiple adverse effects. Recent advances in basic and translational sciences have elucidated a crucial role for the interferon-alpha (IFNα) pathway in the pathogenesis of this enigmatic disease. The so-called “type I interferon signature” has emerged as a major risk factor for disease activity of SLE. Multiple genes encoding for molecules within the type I interferon pathway have been associated with SLE in genome wide association studies. In addition, innate immune receptors are thought to be triggered by either endogenous and/or exogenous stimuli that lead to hypersecretion of IFNα. We review the multiple emerging treatment strategies targeting IFNα-related pathways. These include monoclonal antibodies against IFNα, anti-IFNα antibody-inducing vaccines, and inhibitors of Toll-like receptors. We also summarize the current status of these pharmaceutical agents in early clinical trials.

Highlights

► Several anti-IFNα monoclonal antibodies are being tested in early clinical trials. ► Active immunotherapy with a kinoid leads to a sustained anti-IFNα immune response. ► Novel TLR7/TLR9 inhibitors are expected to attenuate stimulation of IFNα secretion. ► Here we summarize these novel approaches to target the IFNα pathway in SLE patients.

Introduction

Systemic lupus erythematosus (SLE) is a chronic multisystem autoimmune disease with a wide range of clinical manifestations and a pathogenesis whose details have remained relatively elusive. Dysregulation of adaptive immune responses in SLE leads to autoantibody production and immune complex deposition in various tissues [1], [2]. Clinical manifestations commonly appear in the skin, kidney, musculoskeletal, and hematologic systems, but SLE can also affect the lungs, central nervous system, serous membranes and virtually every other organ system of the body [1], [3]. The disease is responsible for significant morbidity and mortality, with most recent studies showing a 10-year survival of approximately 70–90% [4], [5]. Both genetic and environmental factors have been linked to SLE [2], [6]. The genetic risk of developing SLE is generally thought to result from the aggregate effects of multiple polymorphisms (although rare single gene mutations also cause SLE-like disease) [7]. Environmental triggers include smoking [8], UV light [9], various medications [10], and possibly certain viruses [2].

Current therapies for SLE are generally lacking in effectiveness and/or safety, and include primarily nonspecific immunomodulatory, immunosuppressive or cytotoxic agents. These therapies inhibit broadly inflammatory mediators or pathways, including those that are not particularly relevant to SLE pathogenesis. Antimalarial agents and nonsteroidal anti-inflammatory drugs (NSAIDs) remain the first-line drugs for mild disease. Corticosteroids are the primary therapy for more serious disease or one that is resistant to first-line agents, as well as during a lupus flare. Other systemic treatments targeting inflammation include cyclophosphamide, mycophenolate mofetil, and azathioprine. Less commonly used immunosuppressive agents include methotrexate, cyclosporine, tacrolimus, and leflunomide [11], [12]. All of these therapies have a broad range of nonspecific effects and are associated with considerable toxicities [11], [12]. More recently developed biologic therapies have been studied in SLE patients and B cell targeted therapy appears to provide some benefit. Belimumab (an inhibitor of the molecule “B Lymphocyte Stimulator,” or BLyS) was recently given FDA-approval for use in treating SLE, the first drug in over 40 years to achieve this status [13]. The original FDA-approved disease-modifying drug for SLE, hydroxychloroquine, an antimalarial agent, has a lengthy track record in the treatment of lupus and has been shown to have an impact on survival [14]. Antimalarial agents have a variety of effects that may be relevant to their therapeutic benefit in SLE, including interference with Toll-like receptor (TLR) signaling pathways that induce interferon-alpha (IFNα) production [15]. Additional evidence has also implicated IFNα in SLE pathogenesis, heightening interest in the development of novel pharmaceutical agents that specifically target the IFNα pathway. The role of IFNα in disease pathogenesis and the current state of development of therapies targeting IFNα are discussed below.

Section snippets

Pathogenesis of SLE

A poor understanding of the pathogenesis of SLE has hampered the development of new therapies directed at the underlying disease process. SLE involves immune dysregulation at the interface between the innate and adaptive immune systems with both endogenous and exogenous triggers contributing to evolution of disease and induction of disease flares, e.g. viral infections, UV light exposure and certain drugs. Basic research has led to the widely held view that defective clearance of apoptotic

Therapeutic Targets for SLE

Evidence for the IFNα pathway in the pathogenesis of lupus has highlighted IFNα and many related signaling molecules as attractive therapeutic targets. Several clinical trials are now underway investigating monoclonal antibodies against IFNα (discussed below) [97], [98], [99]. Other therapeutic approaches under investigation include vaccination against IFNα using an IFNα-kinoid [100], [101], inhibition of TLRs [102], blockade of INFARs [103], and inhibition of JAK/STAT signaling [104]. IFNα

Summary

In summary, evidence from gene expression profiling in SLE patients, as well as from animal and in vitro models, has stimulated the development of anti-cytokine therapy for this disease. Given the remarkable success of targeting cytokines with biologic drugs to treat other autoimmune disease, especially TNF in rheumatoid arthritis, this approach seems reasonable, and we may soon see the emergence of a new class of effective drugs for SLE. Limited early data using sifalimumab has shown reduced

Conflict of Interest Statement

The authors declare no conflict of interest.

Acknowledgments

This work was supported by a grant from the National Institutes of Health (K08 AI095318 to M.A.K.).

References (140)

  • B.J. Barnes et al.

    Global and distinct targets of IRF-5 and IRF-7 during innate response to viral infection

    J. Biol. Chem.

    (2004)
  • R. Salloum et al.

    Interferon regulatory factors in human lupus pathogenesis

    Transl. Res.

    (2011)
  • D.B. Stetson et al.

    Trex1 prevents cell-intrinsic initiation of autoimmunity

    Cell

    (2008)
  • D. Rajagopal et al.

    Plasmacytoid dendritic cell-derived type I interferon is crucial for the adjuvant activity of Toll-like receptor 7 agonists

    Blood

    (2010)
  • S. Sigurdsson et al.

    Polymorphisms in the tyrosine kinase 2 and interferon regulatory factor 5 genes are associated with systemic lupus erythematosus

    Am. J. Hum. Genet.

    (2005)
  • C.J. Lessard et al.

    Identification of a systemic lupus erythematosus susceptibility locus at 11p13 between PDHX and CD44 in a multiethnic study

    Am. J. Hum. Genet.

    (2011)
  • R.G. Lahita

    Systemic Lupus Erythematosus

    (1999)
  • G.C. Tsokos

    Systemic lupus erythematosus

    N. Engl. J. Med.

    (2011)
  • R. Cervera et al.

    Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients

    Medicine

    (2003)
  • J. Trager et al.

    Mortality and causes of death in systemic lupus erythematosus

    Curr. Opin. Rheumatol.

    (2001)
  • A. Jonsen et al.

    Gene–environment interactions in the aetiology of systemic lupus erythematosus

    Autoimmunity

    (2007)
  • K.L. Moser et al.

    Recent insights into the genetic basis of systemic lupus erythematosus

    Genes Immun.

    (2009)
  • J.F. Simard et al.

    Exposure to maternal smoking and incident SLE in a prospective cohort study

    Lupus

    (2009)
  • M. Bijl et al.

    Ultraviolet light and cutaneous lupus

    Lupus

    (2006)
  • E. Ballestar et al.

    The epigenetic face of systemic lupus erythematosus

    J. Immunol.

    (2006)
  • M. Wong et al.

    Lupus, the current therapeutic approaches

    Drugs Today (Barc.)

    (2011)
  • G.S. Alarcon et al.

    Effect of hydroxychloroquine on the survival of patients with systemic lupus erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA L)

    Ann. Rheum. Dis.

    (2007)
  • S. Sun et al.

    TLR7/9 antagonists as therapeutics for immune-mediated inflammatory disorders

    Inflamm. Allergy Drug Targets

    (2007)
  • L.E. Munoz et al.

    SLE—a disease of clearance deficiency?

    Rheumatology (Oxford)

    (2005)
  • M. Herrmann et al.

    Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus

    Arthritis Rheum.

    (1998)
  • A. Rahman et al.

    Systemic lupus erythematosus

    N. Engl. J. Med.

    (2008)
  • D. Koffler et al.

    Immunological studies concerning the nephritis of systemic lupus erythematosus

    J. Exp. Med.

    (1967)
  • J.J. Hooks et al.

    Immune interferon in the circulation of patients with autoimmune disease

    N. Engl. J. Med.

    (1979)
  • E.C. Baechler et al.

    Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • L. Bennett et al.

    Interferon and granulopoiesis signatures in systemic lupus erythematosus blood

    J. Exp. Med.

    (2003)
  • T.B. Niewold et al.

    High serum IFN-alpha activity is a heritable risk factor for systemic lupus erythematosus

    Genes Immun.

    (2007)
  • T.B. Niewold et al.

    Interferon alpha in systemic lupus erythematosus

    J. Biomed. Biotechnol.

    (2010)
  • B. Yan et al.

    Dysfunctional CD4+, CD25 + regulatory T cells in untreated active systemic lupus erythematosus secondary to interferon-alpha-producing antigen-presenting cells

    Arthritis Rheum.

    (2008)
  • K.B. Elkon et al.

    Type I interferon and systemic lupus erythematosus

    J. Interferon Cytokine Res.

    (2011)
  • G.M. Barton et al.

    Toll-like receptor signaling pathways

    Science

    (2003)
  • R. Baccala et al.

    TLR-dependent and TLR-independent pathways of type I interferon induction in systemic autoimmunity

    Nat. Med.

    (2007)
  • M. Swiecki et al.

    Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance

    Immunol. Rev.

    (2010)
  • K. Takeda et al.

    Toll-like receptors

    Annu. Rev. Immunol.

    (2003)
  • C.G. Horton et al.

    Targeting Toll-like receptors for treatment of SLE

    Mediators Inflamm.

    (2010)
  • T. Kawai et al.

    Toll-like receptor and RIG-I-like receptor signaling

    Ann. N. Y. Acad. Sci.

    (2008)
  • T. Kawai et al.

    Interferon-alpha induction through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6

    Nat. Immunol.

    (2004)
  • M.Y. Balkhi et al.

    Functional regulation of MyD88-activated interferon regulatory factor 5 by K63-linked polyubiquitination

    Mol. Cell. Biol.

    (2008)
  • K. Honda et al.

    Role of a transductional–transcriptional processor complex involving MyD88 and IRF-7 in Toll-like receptor signaling

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • K. Honda et al.

    IRF-7 is the master regulator of type-I interferon-dependent immune responses

    Nature

    (2005)
  • B. Barnes et al.

    On the role of IRF in host defense

    J. Interferon Cytokine Res.

    (2002)
  • Cited by (72)

    • Moving Forward With Biologics in Lupus Nephritis

      2019, Advances in Chronic Kidney Disease
      Citation Excerpt :

      Autoantibodies and self-nucleic acids are known stimulators of TLR 7 and 9 on B-cells and dendritic cells resulting in IFN production.79,80 Type I IFNs support adaptive immune responses, promote B-cell maturation and T-cell activation, and are a major driver of SLE inflammation.81,82 IFN-α given to lupus-prone mice accelerates disease, and overexpression of type I IFN correlates with anti-dsDNA titers and LN activity in humans.83-85

    • Therapeutic advances in the treatment of SLE

      2019, International Immunopharmacology
    • Inflammation in systemic immune diseases: Role of TLR9 signaling and the resultant oxidative stress in pathology of lupus

      2017, Immunity and Inflammation in Health and Disease: Emerging Roles of Nutraceuticals and Functional Foods in Immune Support
    • Vaccination to Treat Noninfectious Diseases: Surveying the Opportunities

      2017, Immunopotentiators in Modern Vaccines: Second Edition
    View all citing articles on Scopus
    View full text