Reviews and feature article
Psoriasis pathogenesis and the development of novel targeted immune therapies

https://doi.org/10.1016/j.jaci.2017.07.004Get rights and content

Psoriasis is caused by a complex interplay between the immune system, psoriasis-associated susceptibility loci, autoantigens, and multiple environmental factors. Over the last 2 decades, research has unequivocally shown that psoriasis represents a bona fide T cell–mediated disease primarily driven by pathogenic T cells that produce high levels of IL-17 in response to IL-23. The discovery of the central role for the IL-23/type 17 T-cell axis in the development of psoriasis has led to a major paradigm shift in the pathogenic model for this condition. The activation and upregulation of IL-17 in prepsoriatic skin produces a “feed forward” inflammatory response in keratinocytes that is self-amplifying and drives the development of mature psoriatic plaques by inducing epidermal hyperplasia, epidermal cell proliferation, and recruitment of leukocyte subsets into the skin. Clinical trial data for mAbs against IL-17 signaling (secukinumab, ixekizumab, and brodalumab) and newer IL-23p19 antagonists (tildrakizumab, guselkumab, and risankizumab) underscore the central role of these cytokines as predominant drivers of psoriatic disease. Currently, we are witnessing a translational revolution in the treatment and management of psoriasis. Emerging bispecific antibodies offer the potential for even better disease control, whereas small-molecule drugs offer future alternatives to the use of biologics and less costly long-term disease management.

Section snippets

Psoriasis as a T cell–mediated autoimmune condition

The requirement of the immune response for the development of psoriasis was suggested by early observations that characteristic skin lesions contained increased numbers of inflammatory cellular infiltrates.9, 10 Additionally, patients with psoriasis undergoing bone marrow transplantations11 or treatment with immunosuppressive agents, such as cyclosporine12, 13 and methotrexate,14 experienced dramatic improvements in their inflammatory skin lesions. Subsequent experiments found that the

The pivotal role of IL-17 in updated disease models of psoriasis

For many years, psoriasis was primarily characterized as a TH1-driven disease based on increased production of IFN-γ by CD4+ T cells found in psoriatic tissues compared with low production of cytokines that define the TH2 T-cell subset (ie, IL-4, IL-5, and IL-13). However, with the characterization of a novel TH17 cell subset discovered in the experimental autoimmune encephalomyelitis mouse model commonly used to study multiple sclerosis,42 the field of dermatology research experienced a major

TNF antagonists

TNF is a potent proinflammatory cytokine made by a variety of immune cells found in the skin, including T cells, keratinocytes, DCs, and macrophages. Its upregulation in the skin of patients with psoriasis has been well characterized.59, 60 The clinical efficacy of multiple TNF antagonists (eg, adalimumab, etanercept, and infliximab) underscore the importance of this cytokine in the promotion and maintenance of psoriatic skin lesions, although the percentage of patients experiencing dramatic

Recently approved and novel therapeutic agents for psoriasis

The discovery of the T17 cell population and elucidation of the broad effects of the IL-23/T17 cell signaling axis on keratinocytes and infiltrating immune cells in the skin has largely shaped the current disease model of psoriasis. Therefore psoriatic disease is best understood as a patterned response to chronic activation of the IL-23/T17 cell pathway. For this reason, current therapeutic strategies are now focused on the development of novel agents that disrupt IL-23 or IL-17 cytokine

Conclusions and future directions

The success of the IL-17 and IL-23 antagonists for the treatment of psoriatic disease has resulted in a modification of the primary clinical outcome being used in clinical trials. The treatment target has been moving to increased clearing of disease, such that PASI90 and PASI100 responses are now considered better targets than the PASI75 response. Still, the mere existence of patients with psoriasis whose symptoms do not improve with IL-17 or IL-23 blockade highlights the complexity of this

References (85)

  • A. Chiricozzi et al.

    Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis

    J Invest Dermatol

    (2011)
  • M.B. Teunissen et al.

    Interleukin-17 and interferon-gamma synergize in the enhancement of proinflammatory cytokine production by human keratinocytes

    J Invest Dermatol

    (1998)
  • C.Q. Wang et al.

    IL-17 and TNF synergistically modulate cytokine expression while suppressing melanogenesis: potential relevance to psoriasis

    J Invest Dermatol

    (2013)
  • E.G. Harper et al.

    Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis

    J Invest Dermatol

    (2009)
  • A.S. Haider et al.

    Insights into gene modulation by therapeutic TNF and IFNgamma antibodies: TNF regulates IFNgamma production by T cells and TNF-regulated genes linked to psoriasis transcriptome

    J Invest Dermatol

    (2008)
  • L.C. Zaba et al.

    Resident and “inflammatory” dendritic cells in human skin

    J Invest Dermatol

    (2009)
  • D. Thaci et al.

    Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate to severe plaque psoriasis: CLEAR, a randomized controlled trial

    J Am Acad Dermatol

    (2015)
  • A. Blauvelt et al.

    Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-to-severe plaque psoriasis up to 1 year: results from the CLEAR study

    J Am Acad Dermatol

    (2017)
  • I.B. McInnes et al.

    Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial

    Lancet

    (2015)
  • C.E. Griffiths et al.

    Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials

    Lancet

    (2015)
  • H. Sofen et al.

    Guselkumab (an IL-23-specific mAb) demonstrates clinical and molecular response in patients with moderate-to-severe psoriasis

    J Allergy Clin Immunol

    (2014)
  • J.G. Krueger et al.

    Anti-IL-23A mAb BI 655066 for treatment of moderate-to-severe psoriasis: Safety, efficacy, pharmacokinetics, and biomarker results of a single-rising-dose, randomized, double-blind, placebo-controlled trial

    J Allergy Clin Immunol

    (2015)
  • A. Blauvelt et al.

    Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial

    J Am Acad Dermatol

    (2017)
  • K. Reich et al.

    Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: results from the phase III, double-blind, placebo- and active comparator-controlled VOYAGE 2 trial

    J Am Acad Dermatol

    (2017)
  • J. de Wit et al.

    RORgammat inhibitors suppress T(H)17 responses in inflammatory arthritis and inflammatory bowel disease

    J Allergy Clin Immunol

    (2016)
  • F.O. Nestle et al.

    Psoriasis

    N Engl J Med

    (2009)
  • E.A. Brezinski et al.

    Economic burden of psoriasis in the United States: a systematic review

    JAMA Dermatol

    (2015)
  • J.M. Gelfand et al.

    The risk of mortality in patients with psoriasis: results from a population-based study

    Arch Dermatol

    (2007)
  • J.D. Bos et al.

    Predominance of “memory” T cells (CD4+, CDw29+) over “naive” T cells (CD4+, CD45R+) in both normal and diseased human skin

    Arch Dermatol Res

    (1989)
  • J.D. Bos et al.

    Immunocompetent cells in psoriasis. In situ immunophenotyping by monoclonal antibodies

    Arch Dermatol Res

    (1983)
  • D.J. Eedy et al.

    Clearance of severe psoriasis after allogenic bone marrow transplantation

    BMJ

    (1990)
  • C.N. Ellis et al.

    Cyclosporine improves psoriasis in a double-blind study

    JAMA

    (1986)
  • C.E. Griffiths et al.

    Clearance of psoriasis with low dose cyclosporin

    Br Med J (Clin Res Ed)

    (1986)
  • T.J. Ryan et al.

    Systemic corticosteroids and folic acid antagonists in the treatment of generalized pustular psoriasis. Evaluation and prognosis based on the study of 104 cases

    Br J Dermatol

    (1969)
  • J.C. Chang et al.

    CD8+ T cells in psoriatic lesions preferentially use T-cell receptor V beta 3 and/or V beta 13.1 genes

    Proc Natl Acad Sci U S A

    (1994)
  • S.L. Gottlieb et al.

    Response of psoriasis to a lymphocyte-selective toxin (DAB389IL-2) suggests a primary immune, but not keratinocyte, pathogenic basis

    Nat Med

    (1995)
  • J.R. Abrams et al.

    CTLA4Ig-mediated blockade of T-cell costimulation in patients with psoriasis vulgaris

    J Clin Invest

    (1999)
  • G.G. Krueger et al.

    Development and use of alefacept to treat psoriasis

    J Am Acad Dermatol

    (2003)
  • M. Lebwohl et al.

    A novel targeted T-cell modulator, efalizumab, for plaque psoriasis

    N Engl J Med

    (2003)
  • E.M. Farber et al.

    Natural history of psoriasis in 61 twin pairs

    Arch Dermatol

    (1974)
  • F. Brandrup et al.

    Psoriasis in monozygotic twins: variations in expression in individuals with identical genetic constitution

    Acta Derm Venereol

    (1982)
  • A. Tiilikainen et al.

    Psoriasis and HLA-Cw6

    Br J Dermatol

    (1980)
  • Cited by (619)

    • Small-molecule agents for treating skin diseases

      2024, European Journal of Medicinal Chemistry
    View all citing articles on Scopus

    J.E.H. is supported in part by Rockefeller University CTSA award grant no. KL2TR001865 from the National Center for Advancing Translational Sciences (NCATS)/National Institutes of Health (NIH) Clinical and Translational Science Award (CTSA) program. J.E.H. and J.G.K. are supported in part by grant no. UL1TR001866 from the NCATS/NIH CTSA program.

    Disclosure of potential conflict of interest: J. G. Krueger has received personal fees from Novartis, Pfizer, Lilly, Boehringer, BiogenIdec, Janssen, AbbVie, Escalier, Acros, Roche, Valeant, Allergan, and Aurigene and has received grants paid to his institution from Novartis, Pfizer, Amgen, Boehringer, Innovaderm, BMS, EMD Serono, AbbVie, Paraxel, Leo Pharma, Vitae, Kineta, Regeneron, and Novan. The rest of the authors declare that they have no relevant conflicts of interest.

    Terms in boldface and italics are defined in the glossary on page 646.

    These authors contributed equally to this work.

    View full text