935
Views
18
CrossRef citations to date
0
Altmetric
Reviews

Novel systemic therapies for the treatment of psoriasis

&

Bibliography

  • Papers of special note have been highlighted as either of interest (*) or of considerable interest (**) to readers.
  • Parisi R, Symmons D, Griffiths CE. (On behalf of IMPACT team). Global epidemiology of psoriasis: a systematic review of incidence and prevalence. J Invest Dermatol. 2013;133(2):377–385.
  • Wu Y, Mills D, Bala M. Psoriasis: cardiovascular risk factors and other disease comorbidities. J Drugs Dermatol. 2008;7(4):373–377.
  • Kim N, Thrash B, Menter A. Comorbidities in psoriasis patients. Semin Cutan Med Surg. 2010;29(1):10–15.
  • Ficco HM, Citera G, Cocco JA. Prevalence of psoriatic arthritis in psoriasis patients according to newer classification criteria. Clin Rheumatol. 2014;33(10):1489–1493.
  • Kurd SK, Troxel AB, Crits-Christoph P, et al. The risk of depression, anxiety and suicidality in patients with psoriasis: a population-based cohort study. Arch Dermatol. 2010;146(8):891–895.
  • Sterry W, Strober BE, Menter A. International Psoriasis Council. Obesity in psoriasis: the metabolic, clinical and therapeutic implications. Report of an interdisciplinary conference and review. Br J Dermatol. 2007;157(4):649–655.
  • Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and hypertension: a systematic review and meta-analysis of observational studies. J Hypertens. 2013;31(3):433–442. discussion 442-3.
  • Horreau C, Pouplard C, Brenaut E, et al. Cardiovascular morbidity and mortality in psoriasis and psoriatic arthritis: a systematic literature review. J Eur Acad Dermatol Venereol. 2013;27(Suppl 3):12–29.
  • Parisi R, Rutter MK, Lunt M, et al. Psoriasis and the risk of major cardiovascular events: cohort study using the clinical practice research datalink. J Invest Dermatol. 2015. [ Epub ahead of print]. DOI:10.1038/jid.2015.87.

* Discusses the link between psoriasis and major cardiovascular events.

  • Heller MM, Wong JW, Nguyen TV, et al. Quality-of-life instruments: evaluation of the impact of psoriasis on patients. Dermatol Clin. 2012;30(2):281–291.
  • Vanderpuye-Orgle J, Zhao Y, Lu J. Evaluating the economic burden of psoriasis in the United States. J Am Acad Dermatol. 2015;72:961–967.
  • Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370(9583):263–271.
  • Tsoi LC, Spain SL, Ellinghaus E, et al. Enhanced meta-analysis and replication studies identify five new psoriasis susceptibility loci. Nat Commun. 2015;6:7001.
  • Yin X, Low HQ, Wang L. Genome-wide meta-analysis identifies multiple novel associations and ethnic heterogeneity of psoriasis susceptibility. Nat Commun. 2015;6:6916.
  • Chandran V. The genetics of psoriasis and psoriatic arthritis. Clin Rev Allergy Immunol. 2013;44(2):149–156.
  • Menter A, Gottlieb A, Feldman SR, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008;58(5):826–850.
  • Albanesi C, De Pita O, Girolomoni G. Resident skin cells in psoriasis: a special look at the pathogenetic functions of keratinocytes. Clin Dermatol. 2007;25:581–588.
  • Girolomoni G, Mrowietz U, Paul C. Psoriasis: rationale for targeting interleukin-17. Br J Dermatol. 2012;167(4):717–724.
  • Lynde C, Poulin Y, Vender R. Interleukin 17A: toward a new understanding of psoriasis pathogenesis. J Am Acad Dermatol, 2014;71:141–150.

** Discussion on the rationale for targeting IL-17 in psoriasis.

  • Lowes M, Russell C, Martin D, et al. The IL-23/T17 pathogenic axis in psoriasis is amplified by keratinocyte responses. Trends Immunol. 2013;34(4):174–181.
  • Gaffen SL. Structure and signaling in the IL-17 receptor family. Nat Rev Immunol. 2009;9:556–567.
  • Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol. 2009;129:2175–2183.
  • Palanivel JA, Macbeth AE, Chetty NC, et al. An insight into JAK-STAT signalling in dermatology. Clin Exp Dermatol. 2014;39(4):513–518.

* Explains the roles of JAK/STAT pathway and possible risks of inhibition.

  • Rawlings JS, Rosler KM, Harrison AD. The JAK/STAT signalling pathway. J Cell Sci. 2004;117:1281–1283.
  • Serezani CH, Ballinger MN, Aronoff DM, et al. Cyclic AMP: master regulator of innate immune cell function. Am J Respir Cell Mol Biol. 2008;39:127–132.
  • Tasken K, Aandahl EM. Localized effects of cAMP mediated by distinct routes of protein kinase A. Physiol Rev. 2004;84:137–167.
  • Sanz MJ, Cortijo J, Morcillo EJ. PDE4 inhibitors as new anti-inflammatory drugs: effects on cell trafficking and cell adhesion molecules expression. Pharmacol Ther. 2005;106(3):269–297.
  • Schafer PH. Targeting PDE4 with apremilast, an orally available specific PDE4 inhibitor, in clinical trials for the treatment of psoriasis and arthritis. Presented at the International Congress on Autoimmunity; 2012 May 9–13; Granada, Spain.
  • Schafer P, Robert M. Novel systemic drugs for psoriasis: mechanism of action for apremilast, a specific inhibitor of PDE4. J Am Acad Dermatol. 2013;68(6):1041–1042.
  • Schafer P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol. 2012;83:1583–1590.
  • Novartis Press Release. Novartis Cosentyx ™ receives positive CHMP opinion for first-line treatment of moderate-to-severe psoriasis patients. 2014 Nov 21.
  • Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaque psoriasis – results of two phase 3 trials. N Engl J Med. 2014;371(4):326–338.
  • Mrowietz U, Leonardi C, Girolomoni G, et al. Secukinumab retreatment-as-needed versus fixed interval maintenance regimen for moderate to severe plaque psoriasis: a randomized, double-blind, non-inferiority trial (SCULPTURE). J Am Acad Dermatol. 2015;73(1):27–36.e1. Epub 2015 May 14. DOI:10.1016/j.jaad.2015.04.011.
  • Thaçi D, Blauvelt A, Reich K. 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. [ Epub ahead of print]. pii: S0190-9622(15)01683-7. DOI:10.1016/j.jaad.2015.05.013.

* Paper that demonstrates the efficacy of secukinumab and its superiority to ustekinumab.

  • Leonardi C, Matheson R, Zachariae C, et al. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl J Med. 2012;366:1190–1199.
  • Gordon K, Leonardi C, Lebwohl M, et al. A 52-week, open-label study of the efficacy and safety of ixekizumab, an anti-interleukin-17A monoclonal antibody, in patients with chronic plaque psoriasis. J Am Acad Dermatol. 2014;71:1176–1182.
  • Saeki H, Nakagawa H, Ishii T, et al. Efficacy and safety of open-label ixekizumab treatment in Japanese patients with moderate-to-severe plaque psoriasis, erythrodermic psoriasis and generalized pustular psoriasis. J Eur Acad Dermatol Venereol. 2015;29(6):1148–1155.
  • Griffiths CE, Reich K, Lebwohl M, 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. [ Epub ahead of print]. pii: S0140-6736(15)60125-8. DOI:10.1016/S0140-6736(15)60125-8.
  • Papp K, Leonardi C, Menter A, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl J Med. 2012;366:1181–1189.
  • Papp K, Menter A, Strober B, et al. Efficacy and safety of brodalumab in subpopulations of patients with difficult-to-treat moderate-to-severe plaque psoriasis. J Am Acad Dermatol. 2015;72(3):436–439.e1.
  • Papp K, Reich K, Leonardi C, et al. Efficacy and safety of brodalumab in patients with moderate to severe plaque psoriasis: results of amagine-1, a phase 3, randomized, double-blind, placebo-controlled study through week 12. Poster session (275): The 73rd Annual Meeting of the American Academy of Dermatology; 2015 Mar 20–24; San Francisco, CA.
  • Papp K, Leonardi C, Menter A, et al. Safety and efficacy of brodalumab for psoriasis after 120 weeks of treatment. J Am Acad Dermatol. 2014;71:1183–1190.
  • Amgen Press release. Amgen to Terminate Participation in Co-development and Commercialization of Brodalumab. 2015 May 22.
  • Lebwohl M, Strober B, Menter A, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med. 2015;373(14):1318–1328.
  • Papp K, Reich K, Leonardi C, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe psoriasis: results of a phase III, randomized, controlled trial (Efficacy and safety trial evaluating the effects of apremilast in psoriasias [ESTEEM] 1). J Am Acad Dermatol. 2015;73(1):37–49.
  • Paul C, Crowley J, Cather J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe psoriasis: 16-week results of a phase 3 randomized, controlled trial (ESTEEM 2). Poster session: The American Academy of Dermatology 72nd Annual Meeting; 2014 Mar 21–25; Denver, CO.
  • Korman N, Poulin Y, Griffiths CEM, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe plaque psoriasis: pooled 16-week efficacy in patient subgroups (ESTEEM 1 and 2). Poster session (1099): The 73rd Annual Meeting of the American Academy of Dermatology; 2015 Mar 20–24; San Francisco, CA.
  • 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;73:1020–1026.
  • 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;42:479–488.
  • Celgene Corporation press release. Oral Otezla (apremilast) approved by the U.S. Food and Drug Administration for the treatment of patients with moderate to severe plaque psoriasis. 2014 Sept 23.
  • Reich K, Soung J, Gooderham M, et al. Efficacy and safety of apremilast or etanercept compared with placebo in patients with moderate to severe psoriasis. Late-breaking research: therapeutic (F010): Presented at the 73rd Annual Meeting of the American Academy of Dermatology; 2015 Mar 20–24; San Francisco, CA.
  • Papp KA, Menter A, Strober B, et al. Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: a phase 2b randomized placebo-controlled dose-ranging study. Br J Dermatol. 2012;167:668–677.
  • Mamolo C, Harness J, Tan H, et al. Tofacitinib (CP-690,550), an oral Janus kinase inhibitor, improves patient-reported outcomes in a phase 2b, randomized, double-blind, placebo-controlled study in patients with moderate-to-severe psoriasis. J Eur Acad Dermatol Venereol. 2014;28(2):192–203.
  • Bachelez H, van de Kerkhof PC, Strohal R, et al. Tofacitinib versus etanercept or placebo in moderate-to-severe chronic plaque psoriasis: a phase 3 randomised non-inferiority trial. Lancet. 2015;386:552–561.
  • Strober B, Buonanno M, Clark JD, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol. 2013;169(5):992–999.
  • Papp KA, Menter MA, Abe M, et al. Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: results from two, randomised, placebo-controlled, phase 3 trials. Br J Dermatol. 2015. [ Epub ahead of print]. DOI:10.1111/bjd.14018.
  • Van Roon EN, van de Laar MA. Methotrexate bioavailability. Clin Exp Rheumatol. 2010;28(5 Suppl 61):S27–S32.
  • Dervieux T, Zablocki R, Kremer J. Red blood cell methotrexate polyglutamates emerge as a function of dosage intensity and route of administration during pulse methotrexate therapy in rheumatoid arthritis. Rheumatology (Oxford). 2010;49:2337–2345.
  • Rutkowska-Sak L, Rell-Bakalarska M, Lisowska B. Oral vs subcutaneous low-dose methotrexate treatment in reducing gastrointestinal side effects. Reumatologia. 2009;47:207–211.
  • Litjens NH, Nibbering PH, Barrois AJ, et al. Beneficial effects of fumarate therapy in psoriasis vulgaris patients coincide with downregulation of type 1 cytokines. Br J Dermatol. 2003;148:444–451.
  • Arbiser JL. Fumarate esters as angiogenesis inhibitors: key to action in psoriasis? J Invest Dermatol. 2011;131(6):1189–1191.
  • ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). Identifier NCT01815723, a randomised, double blind, double dummy, active comparator and placebo controlled confirmative non-inferiority trial of FP187 compared to Fumaderm® in moderate to severe plaque psoriasis; [cited 2015 Jul 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01815723.
  • Blauvelt A, Lebwohl M, Bissonnette R. IL-23/IL-17A dysfunction phenotypes inform possible clinical effects from anti-IL-17A therapies. J Invest Dermatol. 2015. [ Epub ahead of print]. DOI:10.1038/jid.2015.144.

** Looks at theoretical risks of IL-23 and IL-17 blockade including possible infective risks.

  • Bustamante J, Boisson-Dupuis S, Jouanguy E, et al. Novel primary immunodeficiencies revealed by the investigation of paediatric infectious diseases. Curr Opin Immunol. 2008;20(1):39–48.
  • Akahoshi M, Nakashima H, Miyake K, et al. Influence of interleukin-12 receptor β1 polymorphisms on tuberculosis. Hum Genet. 2003;112(3):237–243.
  • Godinez I, Keestra AM, Spees A, et al. The IL-23 axis in Salmonella gastroenteritis. Cell Dmicrobiol. 2011;13(11):1639–1647.
  • Ma CS, Chew GY, Simpson N, et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med. 2008;205:1551–1557.
  • Yang B, Kang H, Fung A, et al. The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm. 2014;2014:623759.
  • He D, Li H, Yusuf N, et al. IL-17 promotes tumor development through the induction of tumor promoting microenvironments at tumor sites and myeloid-derived suppressor cells. J Immunol. 2010;184(5): 2281–2288.
  • Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9(11):798–809.
  • Grivennikov SI, Karin M. Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010;21(1):11–19.
  • Alshaker HA, Matalka KZ. IFN-γ, IL-17 and TGF-β involvement in shaping the tumor microenvironment: the significance of modulating such cytokines in treating malignant solid tumors. Cancer Cell Int. 2011;11:33.
  • Wu JJ, Poon KY, Channual JC, et al. Association between tumor necrosis factor inhibitor therapy and myocardial infarction risk in patients with psoriasis. Arch Dermatol, 2012;148(11):1244–1250. DOI:10.1001/archdermatol.2012.2502. Erratum in: Arch Dermatol 2013 Feb;149(2):158. DOI: http://dx.doi.org./10.1001/jamadermatol.2013.3334.
  • Ahlehoff O, Skov L, Gislason G, et al. Cardiovascular disease event rates in patients with severe psoriasis treated with systemic anti-inflammatory drugs: a Danish real-world cohort study. J Intern Med. 2013;273(2):197–204. DOI:10.1111/j.1365-2796.2012.02593.x.
  • Ryan C, Leonardi CL, Krueger JG, et al. Association between biologic therapies for chronic plaque psoriasis and cardiovascular events: a meta-analysis of randomized controlled trials. JAMA. 2011;306(8): 864–871.
  • Abuabara K, Lee H, Kimball AB. The effect of systemic psoriasis therapies on the incidence of myocardial infarction: a cohort study. Br J Dermatol. 2011;165(5):1066–1073.
  • Dommasch ED, Troxel AB, Gelfand JM. Major cardiovascular events associated with anti-IL 12/23 agents: a tale of two meta-analyses. J Am Acad Dermatol. 2013;68(5):863–865.
  • Tzellos T, Kyrgidis A, Zouboulis CC. Re-evaluation of the risk for major adverse cardiovascular events in patients treated with anti-IL-12/23 biological agents for chronic plaque psoriasis: a meta-analysis of randomized controlled trials. J Eur Acad Dermatol Venereol. 2013;27(5):622–627.
  • Puig L. Cardiovascular risk and psoriasis: the role of biologic therapy. Actas Dermosifiliogr. 2012;103(10):853–862.
  • Simon T, Taleb S, Danchin N. Circulating levels of interleukin-17 and cardiovascular outcomes in patients with acute myocardial infarction. Eur Heart J. 2013;34(8):570–577.
  • Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–186.
  • Shelton RC, Miller AH. Inflammation in depression: is adiposity a cause? Dialogues Clin Neurosci. 2011;13(1):41–53.
  • Hunter HJ, Hinz R, Gerhard A, et al. Brain inflammation and psoriasis: a [11C]-(R)-PK11195 positron emission tomography study. Br J Dermatol. 2015. [ Epub ahead of print]. DOI:10.1111/bjd.13788.
  • 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):976–986.
  • Kavanaugh A, McInnes I, Mease P, et al. Clinical efficacy, radiographic and safety findings through 5 years of subcutaneous golimumab treatment in patients with active psoriatic arthritis: results from a long-term extension of a randomised, placebo-controlled trial (the GO-REVEAL study). Ann Rheum Dis. 2014;73(9):1689–1694.
  • National Cancer Institute. The surveillance, epidemiology, and end results (SEER) database; 2004 [cited 2013 Jul 22]. Available from: http://seer.cancer.gov/statistics.
  • Reich K, Ortonne JP, Gottlieb AB, et al. Successful treatment of moderate to severe plaque psoriasis with the PEGylated Fab’ certolizumab pegol: results of a phase II randomized, placebo-controlled trial with a re-treatment extension. Br J Dermatol. 2012;167(1):180–190.
  • Gordon KB, Duffin KC, Bissonnette R, et al, A phase 2 trial of guselkumab versus adalimumab for plaque psoriasis. N Engl J Med. 2015;373(2): 136–144.
  • Papp K, Thaçi D, Reich K. Tildrakizumab (MK-3222), an anti- IL-23p19 monoclonal antibody, improves psoriasis in a phase 2b randomized placebo- controlled trial. Br J Dermatol. 2015. [ Epub ahead of print]. DOI:10.1111/bjd.13932.
  • Krueger JG, Ferris LK, Menter A, 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 Mar 11. [Epub ahead of print]. pii: S0091-6749(15)00108-6 DOI:10.1016/j.jaci.2015.01.018.
  • Papp KA, et al. Efficacy and safety of different dose regimens of a novel selective IL-23p19 inhibitor (BI-655066) compared with ustekinumab in patients with moderate-to-severe plaque psoriasis. Abstract F010.2. The 73rd Annual Meeting of the American Academy of Dermatology; 2015 March 20–24; San Francisco, CA.
  • ClinicalTrials.gov. Bethesda (MD): National Library of Medicine (US). Identifier NCT02349451, a phase 2 study to investigate the safety, tolerability and efficacy of ABT-122 in subjects with active psoriatic arthritis who have an inadequate response to methotrexate [cited 2015 Sept 25]. Available from: https://clinicaltrials.gov/ct2/show/NCT02349451.
  • Vaclavkova A, Chimenti S, Arenberger P, et al. Oral ponesimod in patients with chronic plaque psoriasis: a randomised, double-blind, placebo-controlled phase 2 trial. Lancet. 2014;384(9959): 2036–2045.
  • Strober BE, Armour K, Romiti R, et al. Biopharmaceuticals and biosimilars in psoriasis: what the dermatologist needs to know. J Am Acad Dermatol. 2012;66(2):317–322.
  • Feagan BG, Choquette D, Ghosh S, et al. The challenge of indication extrapolation for infliximab biosimilars. Biologicals. 2014;42(4):177–183. Epub 2014 Jun 21.
  • Jung YS, Park DI, Kim YH. Efficacy and safety of CT-P13, a biosimilar of infliximab, in patients with inflammatory bowel disease: a retrospective multicenter study. J Gastroenterol Hepatol. 2015. [ Epub ahead of print]. DOI:10.1111/jgh.12997.
  • McKeage K. A review of CT-P13: an infliximab biosimilar. BioDrugs. 2014;28(3):313–321.
  • ClinicalTrials.gov. A service of the U.S. National Institutes of Health; 2014 [cited 2015 July 10]. Available from: https://clinicaltrials.gov/.
  • Nikamo P, Lysell J, Ståhle M. Association with genetic variants in the IL-23 and NF-κB pathways discriminates between mild and severe psoriasis skin disease. J Invest Dermatol. 2015;135(8):1969–1976. Epub 2015 Mar 19. DOI:10.1038/jid.2015.103.
  • Talamonti M, Botti E, Galluzzo M, et al. Pharmacogenetics of psoriasis: HLA-Cw6 but not LCE3B/3C deletion nor TNFAIP3 polymorphism predisposes to clinical response to interleukin 12/23 blocker ustekinumab. Br J Dermatol. 2013;169(2):458–463.
  • Julià M, Guilabert A, Lozano F, et al. The role of Fcγ receptor polymorphisms in the response to anti–tumor necrosis factor therapy in psoriasis A pharmacogenetic study. JAMA Dermatol. 2013;149(9):1033–1039.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.