Skip to main content
Top

08-03-2016 | Psoriatic arthritis | Review | Article

Beyond TNF Inhibitors: New Pathways and Emerging Treatments for Psoriatic Arthritis

Journal: Drugs

Authors: Ennio Lubrano, Fabio Massimo Perrotta

Publisher: Springer International Publishing

Abstract

Psoriatic arthritis (PsA) is a chronic inflammatory disease characterized by psoriasis, synovitis, enthesitis, spondylitis and association with other extra-articular manifestations. Chronic inflammation of involved tissues possibly leads to structural damage and to a reduction in function and quality of life. The treatment of PsA dramatically changed with the introduction of anti-tumor necrosis factor (TNF)-α drugs, which have been shown to reduce the symptoms and signs of the disease, and slow radiographic progression. However, some patients do not respond to anti-TNFα or have a loss of response. Recently, the discovery of new pathogenic mechanisms have made possible the development of new drugs that target pro-inflammatory cytokines, such as interleukin (IL)-12, IL-23 and IL-17, or interfere with cellular pathways involved in skin, joint and entheseal inflammation. New molecules, namely ustekinumab, secukinumab, and apremilast have shown efficacy and safety over the various components of the disease in randomized clinical trials. These drugs have been recently approved for the treatment of PsA and included in new treatment recommendations. Other molecules are currently being tested in phase III clinical trials and are potential new treatment options for PsA. The aim of this review is to update the new pathways involved in the development of the disease and the emerging treatments for PsA beyond TNFα inhibition.
Literature
1.
Gladman DD, Antoni C, Mease P, Clegg DO, Nash P. Psoriatic arthritis: epidemiology, clinical features, course and outcome. Ann Rheum Dis. 2005;64(2):14–7.
2.
McHugh NJ, Balachrishnan C, Jones SM. Progression of peripheral joint disease in psoriatic arthritis: a 5-yr prospective study. Rheumatology (Oxford). 2003;42:778–83.PubMedCrossRef
3.
Veale D. Psoriatic arthritis: recent progress in pathophysiology and drug development. Arthritis Res Ther. 2013;15:224.PubMedPubMedCentralCrossRef
4.
Ritchlin CT, Kavanaugh A, Gladman DD, Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA), et al. Treatment recommendations for psoriatic arthritis. Ann Rheum Dis. 2009;68:1387–94.PubMedPubMedCentralCrossRef
5.
D’Angelo S, Palazzi C, Olivieri I. Psoriatic arthritis: treatment strategies using biologic agents. Reumatismo. 2012;64:113–21.PubMed
6.
Atteno M, Peluso R, Costa L, et al. Comparison of effectiveness and safety of infliximab, etanercept, and adalimumab in psoriatic arthritis patients who experienced an inadequate response to previous disease-modifying antirheumatic drugs. Clin Rheumatol. 2010;29(4):399–403.PubMedCrossRef
7.
Fénix-Caballero S, Alegre-del Rey EJ, Castaño-Lara R, Puigventós-Latorre F, Borrero-Rubio JM, López-Vallejo JF. Direct and indirect comparison of the efficacy and safety of adalimumab, etanercept, infliximab and golimumab in psoriatic arthritis. J Clin Pharm Ther. 2013;38(4):286–93.PubMedCrossRef
8.
Gomez-Reino JJ, Carmona L, BIOBADASER Group. Switching TNFα antagonists in patients with chronic arthritis: an observational study of 488 patients over a four-year period. Arthritis Res Ther. 2006;8(1):R29.PubMedPubMedCentralCrossRef
9.
Collamer AN, Guerrero KT, Henning JS, Battafarano DF. Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy: a literature review and potential mechanisms of action. Arthritis Rheum. 2008;59(7):996–1001.PubMedCrossRef
10.
Dörner T, Kay J. Biosimilars in rheumatology: current perspectives and lessons learnt. Nat Rev Rheumatol. 2015;11(12):713–24.PubMedCrossRef
11.
Smith JA, Colbert RA. Review: the interleukin-23/interleukin-17 axis in spondyloarthritis pathogenesis: Th17 and beyond. Arthritis Rheumatol. 2014;66(2):231–41.PubMedPubMedCentralCrossRef
12.
Spadaro A, Montepaone M, Lubrano E. A novel biological target for the treatment of psoriatic arthritis. Immunotherapy. 2014;6(5):515–8.PubMedCrossRef
13.
Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3(2):133–46.PubMedCrossRef
14.
Oppmann B, Lesley R, Blom B, et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity. 2000;13(5):715–25.PubMedCrossRef
15.
Cargill M, Schrodi SJ, Chang M, et al. A large-scale genetic association study confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes. Am J Hum Genet. 2007;80:273–90.PubMedPubMedCentralCrossRef
16.
Barnas JL, Ritchlin CT. Etiology and pathogenesis of psoriatic arthritis. Rheum Dis Clin North Am. 2015;41(4):643–63.PubMedCrossRef
17.
Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129(6):1339–50.PubMedCrossRef
18.
Jandus C, Bioley G, Rivals JP, Dudler J, Speiser D, Romero P. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum. 2008;58:2307–17.PubMedCrossRef
19.
Lee Y. The role of interleukin-17 in bone metabolism and inflammatory skeletal diseases. BMB Rep. 2013;46(10):479–83.PubMedPubMedCentralCrossRef
20.
Toichi E, Torres G, McCormick TS, et al. An anti-IL-12p40 antibody down-regulates type 1 cytokines, chemokines, and IL-12/IL-23 in psoriasis. J Immunol. 2006;1177(7):4917–26.CrossRef
21.
Davari P, Leo MS, Kamangar F, Fazel N. Ustekinumab for the treatment of psoriatic arthritis: an update. Clin Cosmet Investig Dermatol. 2014;7:243–9.PubMedPubMedCentral
22.
Griffiths CE, Strober BE, van de Kerkhof P, ACCEPT Study Group, et al. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med. 2010;362(2):118–28.PubMedCrossRef
23.
McInnes IB, Kavanaugh A, Gottlieb AB, et al. PSUMMIT 1 Study Group. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet. 2013;382(9894):780–9.PubMedCrossRef
24.
Ritchlin C, Rahman P, Kavanaugh A, PSUMMIT 2 Study Group, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour necrosis factor therapy: 6-month and 1-year results of the phase 3, multicentre, double-blind, placebo controlled, randomized PSUMMIT 2 trial. Ann Rheum Dis. 2014;73(6):990–9.PubMedPubMedCentralCrossRef
25.
Papp K, Gottlieb AB, Naldi L, et al. Safety Surveillance for Ustekinumab and Other Psoriasis Treatments From the Psoriasis Longitudinal Assessment and Registry (PSOLAR). J Drugs Dermatol. 2015;14(7):706–14.PubMed
26.
Kavanaugh A, Ritchlin C, Rahman P, PSUMMIT-1 and 2 Study Groups, et al. Ustekinumab, an anti-IL-12/23 p40 monoclonal antibody, inhibits radiographic progression in patients with active psoriatic arthritis: results of an integrated analysis of radiographic data from the phase 3, multicentre, randomised, double-blind, placebo-controlled PSUMMIT-1 and PSUMMIT-2 trials. Ann Rheum Dis. 2014;73(6):1000–6.PubMedPubMedCentralCrossRef
27.
Kavanaugh A, Puig L, Gottlieb AB, PSUMMIT I Study Group, et al. Maintenance of clinical efficacy and radiographic benefit through 2 years of ustekinumab therapy in patients with active psoriatic arthritis: results from the PSUMMIT 1 trial. Arthritis Care Res Hoboken. 2015;19:26097039 (Epub ahead of print).
28.
Gordon KB, Langley RG, Gottlieb AB, et al. A phase III, randomized, controlled trial of the fully human IL-12/23 mAb briakinumab in moderate-to-severe psoriasis. J Invest Dermatol. 2012;132(2):304–14.PubMedCrossRef
29.
McInnes IB, Mease PJ, Kirkham B, FUTURE 2 Study Group, 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;386(9999):1137–4630.PubMedCrossRef
30.
Mease PJ, McInnes IB, Kirkham B, FUTURE 1 Study Group, et al. Secukinumab inhibition of interleukin-17A in patients with psoriatic arthritis. N Engl J Med. 2015;373(14):1329–39.PubMedCrossRef
31.
Gossec L, Smolen JS, Ramiro S, et al. European League Against Rheumatism (EULAR) recommendations for the management of psoriatic arthritis with pharmacological therapies: 2015 update. Ann Rheum Dis. 2016;75(3):499–510.PubMedCrossRef
32.
Coates LC, Kavanaugh A, Mease PJ, et al. Group for research and assessment of psoriasis and psoriatic arthritis: treatment recommendations for psoriatic arthritis 2015. Arthritis Rheumatol. 2016. doi:10.​1002/​art.​39573.
33.
Papp KA, Leonardi C, Menter A, et al. Brodalumab, an anti–interleukin-17-receptor antibody for psoriasis. N Engl J Med. 2012;366:1181–9.PubMedCrossRef
34.
Mease PJ, Genovese MC, Greenwald MW, et al. Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med. 2014;370(24):2295–306.PubMedCrossRef
35.
Leonardi C, Matheson R, Zachariae C, et al. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl J Med. 2012;366:1190–9.PubMedCrossRef
36.
Conti M, Beavo J. Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu Rev Biochem. 2007;76:481–511.PubMedCrossRef
37.
Houslay MD, Adams DR. PDE4 cAMP phosphodiesterases: modular enzymes that orchestrate signalling cross-talk, desensitization and compartmentalization. Biochem J. 2003;370(1):1–18.PubMedPubMedCentralCrossRef
38.
Houslay MD, Schafer P, Zhang KY. Keynote review: phosphodiesterase-4 as a therapeutic target. Drug Discov Today. 2005;10(22):1503–19.PubMedCrossRef
39.
Jin SL, Ding SL, Lin SC. Phosphodiesterase 4 and its inhibitors in inflammatory diseases. Chang Gung Med J. 2012;35(3):197–210.PubMed
40.
Jimenez JL, Punzón C, Navarro J, Muñoz-Fernández MA, Fresno M. Phosphodiesterase 4 inhibitors prevent cytokine secretion by T lymphocytes by inhibiting nuclear factor-kappaB and nuclear factor of activated T cells activation. J Pharmacol Exp Ther. 2001;299(2):753–9.PubMed
41.
Schett G, Sloan VS, Stevens RM, Schafer P. Apremilast: a novel PDE4 inhibitor in the treatment of autoimmune and inflammatory diseases. Ther Adv Musculoskelet Dis. 2010;2(5):271–8.PubMedPubMedCentralCrossRef
42.
Liu J, Chen M, Wang X. Calcitonin gene-related peptide inhibits lipopolysaccharide-induced interleukin-12 release from mouse peritoneal macrophages, mediated by the cAMP pathway. Immunology. 2000;101(1):61–7.PubMedPubMedCentralCrossRef
43.
Schafer PH, Parton A, Gandhi AK, et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br J Pharmacol. 2010;159(4):842–55.PubMedPubMedCentralCrossRef
44.
Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015;73(1):37–49.PubMedCrossRef
45.
Paul C, Cather J, Gooderham M, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe plaque psoriasis over 52 weeks: a phase III, randomized, controlled trial (ESTEEM 2). Br J Dermatol. 2015 (Epub ahead of print).
46.
Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomised, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020–6.PubMedPubMedCentralCrossRef
47.
Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Longterm (52-week) results of a phase III randomized, controlled trial of apremilast in patients with psoriatic arthritis. J Rheumatol. 2015;42(3):479–88.PubMedCrossRef
48.
Ungprasert P, Thongprayoon C, Davis JM 3rd. Indirect comparisons of the efficacy of subsequent biological agents in patients with psoriatic arthritis with an inadequate response to tumor necrosis factor inhibitors: a meta-analysis. Clin Rheumatol. 2016 (Epub ahead of print).
49.
Mease PJ. Psoriatic arthritis treatment update. Bull NYU Hosp Joint Dis. 2012;70:167–71.
50.
Cohen JD. Successful treatment of psoriatic arthritis with rituximab. Ann Rheum Dis. 2008;67(11):1647–8.PubMedCrossRef
51.
Sarzi-Puttini P, Atzeni F. New biological treatments for psoriatic arthritis. Isr Med Assoc J. 2014;16(10):643–5.PubMed
52.
Ogata A, Kumanogoh A, Tanaka T. Pathological role of interleukin-6 in psoriatic arthritis. Arthritis. 2012;2012:713618.PubMedPubMedCentralCrossRef
53.
Mease PJ, Gottlieb AB, Berman A et al. A phase IIb, randomized, double-blind, placebo-controlled, dose-ranging, multicenter study to evaluate the efficacy and safety of clazakizumab, an anti-IL-6 monoclonal antibody, in adults with active psoriatic arthritis. ACR/ARHP 2014 Annual Meeting. Abstract Number: 952.
54.
Shetty A, Hanson R, Korsten P, et al. Tocilizumab in the treatment of rheumatoid arthritis and beyond. Drug Des Devel Ther. 2014;8:349–64.PubMedPubMedCentral
55.
Murakami M, Nishimoto N. The value of blocking IL-6 outside of rheumatoid arthritis: current perspective. Curr Opin Rheumatol. 2011;23(3):273–7.PubMedCrossRef
56.
Nishimoto N. Clinical studies in patients with Castleman’s disease, Crohn’s disease, and rheumatoid arthritis in Japan. Clin Rev Allergy Immunol. 2005;28(3):221–30.PubMedCrossRef
57.
Ogata A, Umegaki N, Katayama I, Kumanogoh A, Tanaka T. Psoriatic arthritis in two patients with an inadequate response to treatment with tocilizumab. Joint Bone Spine. 2012;79(1):85–7.PubMedCrossRef
58.
Sieper J, Porter-Brown B, Thompson L, Harari O, Dougados M. Assessment of short-term symptomatic efficacy of tocilizumab in ankylosing spondylitis: results of randomised, placebo-controlled trials. Ann Rheum Dis. 2014;3(1):95–100.CrossRef
59.
Lekpa FK, Poulain C, Wendling D, Club Rhumatismes et Inflammation, et al. Is IL-6 an appropriate target to treat spondyloarthritis patients refractory to anti-TNF therapy? A multicenter retrospective observational study. Arthritis Res Ther. 2012;14(2):R53.PubMedPubMedCentralCrossRef
60.
Pieper J, Herrath J, Raghavan S, Muhammad K, Vollenhoven Rv, Malmström V. CTLA4-Ig (abatacept) therapy modulates T cell effector functions in autoantibody-positive rheumatoid arthritis patients. BMC Immunol. 2013;14:34.PubMedPubMedCentralCrossRef
61.
VicenteRabaneda EF, Herrero-Beaumont G, Castañeda S. Update on the use of abatacept for the treatment of rheumatoid arthritis. Expert Rev Clin Immunol. 2013;9(7):599–621.CrossRef
62.
Mease P, Genovese MC, Gladstein G, et al. Abatacept in the treatment of patients with psoriatic arthritis: results of a six-month, multicenter, randomized, double-blind, placebo-controlled, phase II trial. Arthritis Rheum. 2011;63(4):939–48.PubMedCrossRef
63.
Ghoreschi K, Laurence A, O’She J. Janus kinases in immune cell signaling. Immunol Rev. 2009;228:273–87.PubMedPubMedCentralCrossRef
64.
Eriksen KW, Lovato P, Skov L, et al. Increased sensitivity to interferon-alpha in psoriatic T cells. J Invest Dermatol. 2005;125(5):936–44.PubMedCrossRef
65.
Rácz E, Prens EP, Kurek D, et al. Effective treatment of psoriasis with narrow-band UVB phototherapy is linked to suppression of the IFN and Th17 pathways. J Invest Dermatol. 2011;131(7):1547–58.PubMedCrossRef
66.
Kontzias A, Kotlyar A, Laurence A, Changelian P, O’Shea JJ. Jakinibs: a new class of kinase inhibitors in cancer and autoimmune disease. Curr Opin Pharmacol. 2012;12(4):464–70.PubMedPubMedCentralCrossRef
67.
Burmester GR, Blanco R, Charles-Schoeman C, ORAL Step investigators, et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet. 2013;381(9865):451–60.PubMedCrossRef
68.
Lubrano E, Perrotta FM, Marchesoni A, et al. Remission in non radiographic axial spondyloarthritis treated with anti-tumor necrosis factor-α drugs: an Italian multicenter study. J Rheumatol. 2015;42:258–63.PubMedCrossRef
69.
Spadaro A, Lubrano E, Marchesoni A, et al. Remission in ankylosing spondylitis treated with anti-TNF-α drugs: a national multicentre study. Rheumatology (Oxford). 2013;52:1914–9.PubMedCrossRef
70.
Lubrano E, Perrotta FM, Kavanaugh A. An overview of low disease activity and remission in psoriatic arthritis. Clin Exp Rheumatol. 2015;33(5):51–4.
71.
Lubrano E, Soriano E, FitzGerald O. Can traditional disease-modifying anti-rheumatic drugs be withdrawn or tapered in psoriatic arthritis? Clin Exp Rheumatol. 2013;31:S54–8.PubMed
72.
Antoni CE, Kavanaugh A, Kirkham B, et al. Sustained benefits of infliximab therapy for dermatologic and articular manifestations of psoriatic arthritis: results from the infliximab multinational psoriatic arthritis controlled trial (IMPACT). Arthritis Rheum. 2005;52(4):1227–36.PubMedCrossRef
73.
Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment of psoriatic arthritis: safety, efficacy, and effect on disease progression. Arthritis Rheum. 2004;50(7):2264–72.PubMedCrossRef
74.
Mease PJ, Gladman DD, Ritchlin CT, Adalimumab Effectiveness in Psoriatic Arthritis Trial Study Group, et al. Adalimumab for the treatment of patients with moderately to severely active psoriatic arthritis: results of a double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2005;52(10):3279–89.PubMedCrossRef
75.
Kavanaugh A, McInnes I, Mease P, et al. Golimumab, a new human tumor necrosis factor alpha antibody, administered every four weeks as a subcutaneous injection in psoriatic arthritis: twenty-four-week efficacy and safety results of a randomized, placebo-controlled study. Arthritis Rheum. 2009;60(4):976–86.PubMedCrossRef
76.
Mease PJ, Fleischmann R, Deodhar AA, et al. Effect of certolizumab pegol on signs and symptoms in patients with psoriatic arthritis: 24-week results of a Phase 3 double-blind randomised placebo-controlled study (RAPID-PsA). Ann Rheum Dis. 2014;73(1):48–55.PubMedPubMedCentralCrossRef
77.
Maska L, Anderson J, Michaud K. Measures of functional status and quality of life in rheumatoid arthritis: Health Assessment Questionnaire Disability Index (HAQ), Modified Health Assessment Questionnaire (MHAQ), Multidimensional Health Assessment Questionnaire (MDHAQ), Health Assessment Questionnaire II (HAQ-II), Improved Health Assessment Questionnaire (Improved HAQ), and Rheumatoid Arthritis Quality of Life (RAQoL). Arthritis Care Res (Hoboken). 2011;63(11):S4–13.PubMedCrossRef