1932

Abstract

The discovery of the biological functions of the interleukin-23/-17 axis led to the identification of IL-23 and IL-17 as important participants in the pathogenesis of several immune-mediated diseases. Therapeutic agents targeting these cytokines and/or their receptors have now been developed as potential treatment strategies for common immune-mediated diseases. Anti-IL-17 and anti-IL-12/-23 regimens appear particularly effective in psoriasis, with promising results in spondyloarthropathies also emerging. Overall, these agents appear well tolerated, with adverse-event rates that are commensurate with those in other biologic treatment programs. The strategic utility of these new agents, however, remains uncertain, and further studies will be required to determine their place in the context of existing conventional and biologic immune-modifying agents.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-051914-021944
2016-01-14
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/med/67/1/annurev-med-051914-021944.html?itemId=/content/journals/10.1146/annurev-med-051914-021944&mimeType=html&fmt=ahah

Literature Cited

  1. Johansen C, Usher PA, Kjellerup RB. 1.  et al. 2009. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br. J. Dermatol. 160:319–24 [Google Scholar]
  2. Hirota K, Duarte JH, Veldhoen M. 2.  et al. 2011. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12:255–63 [Google Scholar]
  3. Harrington LE, Hatton RD, Mangan PR. 3.  et al. 2005. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat. Immunol. 6:1123–32 [Google Scholar]
  4. Gaffen SL, Jain R, Garg AV, Cua DJ. 4.  2014. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat. Rev. Immunol. 14:585–600Authoritative review describing the regulation of the IL-23/-17 axis. [Google Scholar]
  5. Frleta M, Siebert S, McInnes IB. 5.  2014. The interleukin-17 pathway in psoriasis and psoriatic arthritis: disease pathogenesis and possibilities of treatment. Curr. Rheumatol. Rep. 16:414 [Google Scholar]
  6. Zhu S, Qian Y. 6.  2012. IL-17/IL-17 receptor system in autoimmune disease: mechanisms and therapeutic potential. Clin. Sci. 122:487–511 [Google Scholar]
  7. Griffin GK, Newton G, Tarrio ML. 7.  et al. 2012. IL-17 and TNF-alpha sustain neutrophil recruitment during inflammation through synergistic effects on endothelial activation. J. Immunol. 188:6287–99 [Google Scholar]
  8. Suzuki E, Mellins ED, Gershwin ME. 8.  et al. 2014. The IL-23/IL-17 axis in psoriatic arthritis. Autoimmun. Rev. 13:496–502Excellent review of the IL-23/-17 axis with focus on psoriatic arthritis. [Google Scholar]
  9. Parham C, Chirica M, Timans J. 9.  et al. 2002. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 and a novel cytokine receptor subunit, IL-23R. J. Immunol. 168:5699–708 [Google Scholar]
  10. Ciofani M, Madar A, Galan C. 10.  et al. 2012. A validated regulatory network for Th17 cell specification. Cell 151:289–303 [Google Scholar]
  11. Weigle N, McBane S. 11.  2013. Psoriasis. Am. Fam. Phys. 87:626–33 [Google Scholar]
  12. Elliott M, Benson J, Blank M. 12.  et al. 2009. Ustekinumab: lessons learned from targeting interleukin-12/23p40 in immune-mediated diseases. Ann. N.Y. Acad. Sci. 1182:97–110 [Google Scholar]
  13. Bowes J, Barton A. 13.  2010. The genetics of psoriatic arthritis: lessons from genome-wide association studies. Discov. Med. 10:177–83 [Google Scholar]
  14. Hueber W, Patel DD, Dryja T. 14.  et al. 2010. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci. Trans. Med. 2:52ra72 [Google Scholar]
  15. Rich P, Sigurgeirsson B, Thaci D. 15.  et al. 2013. Secukinumab induction and maintenance therapy in moderate-to-severe plaque psoriasis: a randomized, double-blind, placebo-controlled, phase II regimen-finding study. Br. J. Dermatol. 168:402–11 [Google Scholar]
  16. Papp KA, Langley RG, Sigurgeirsson B. 16.  et al. 2013. Efficacy and safety of secukinumab in the treatment of moderate-to-severe plaque psoriasis: a randomized, double-blind, placebo-controlled phase II dose-ranging study. Br. J. Dermatol. 168:412–21 [Google Scholar]
  17. Langley RG, Elewski BE, Lebwohl M. 17.  et al. 2014. Secukinumab in plaque psoriasis—results of two phase 3 trials. N. Engl. J. Med. 371:326–38 [Google Scholar]
  18. Leonardi C, Matheson R, Zachariae C. 18.  et al. 2012. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N. Engl. J. Med. 366:1190–99 [Google Scholar]
  19. Zhu B, Edson-Heredia E, Cameron GS. 19.  et al. 2013. Early clinical response as a predictor of subsequent response to ixekizumab treatment: results from a phase II study of patients with moderate-to-severe plaque psoriasis. Br. J. Dermatol. 169:1337–41 [Google Scholar]
  20. Gordon KB, Leonardi CL, Lebwohl M. 20.  et al. 2014. 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. 71:1176–82 [Google Scholar]
  21. Papp KA, Leonardi C, Menter A. 21.  et al. 2012. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N. Engl. J. Med. 366:1181–89 [Google Scholar]
  22. Gordon KB, Kimball AB, Chau D. 22.  et al. 2014. Impact of brodalumab treatment on psoriasis symptoms and health-related quality of life: use of a novel patient-reported outcome measure, the Psoriasis Symptom Inventory. Br. J. Dermatol. 170:705–15 [Google Scholar]
  23. Papp K, Menter A, Strober B. 23.  et al. 2015. Efficacy and safety of brodalumab in subpopulations of patients with difficult-to-treat moderate-to-severe plaque psoriasis. J. Am. Acad. Dermatol. 72:436–39e1 [Google Scholar]
  24. Papp K, Leonardi C, Menter A. 24.  et al. 2014. Safety and efficacy of brodalumab for psoriasis after 120 weeks of treatment. J. Am. Acad. Dermatol. 71:1183–90e3 [Google Scholar]
  25. Krueger GG, Langley RG, Leonardi C. 25.  et al. 2007. A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N. Engl. J. Med. 356:580–92 [Google Scholar]
  26. Leonardi CL, Kimball AB, Papp KA. 26.  et al. 2008. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 371:1665–74 [Google Scholar]
  27. Papp KA, Langley RG, Lebwohl M. 27.  et al. 2008. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 371:1675–84 [Google Scholar]
  28. Lebwohl M, Papp K, Han C. 28.  et al. 2010. Ustekinumab improves health-related quality of life in patients with moderate-to-severe psoriasis: results from the PHOENIX 1 trial. Br. J. Dermatol. 162:137–46 [Google Scholar]
  29. Reich K, Schenkel B, Zhao N. 29.  et al. 2011. Ustekinumab decreases work limitations, improves work productivity, and reduces work days missed in patients with moderate-to-severe psoriasis: results from PHOENIX 2. J. Dermatol. Treat. 22:337–47 [Google Scholar]
  30. Kimball AB, Gordon KB, Fakharzadeh S. 30.  et al. 2012. Long-term efficacy of ustekinumab in patients with moderate-to-severe psoriasis: results from the PHOENIX 1 trial through up to 3 years. Br. J. Dermatol. 166:861–72 [Google Scholar]
  31. Kimball AB, Papp KA, Wasfi Y. 31.  et al. 2013. Long-term efficacy of ustekinumab in patients with moderate-to-severe psoriasis treated for up to 5 years in the PHOENIX 1 study. J. Eur. Acad. Dermatol. Venereol. 27:1535–45 [Google Scholar]
  32. Langley RG, Lebwohl M, Krueger GG. 32.  et al. 2014. Long-term efficacy and safety of ustekinumab, with and without dosing adjustment, in patients with moderate-to-severe psoriasis: results from the PHOENIX 2 study through 5 years of follow-up. Br. J. Dermatol. 172:1371–83 [Google Scholar]
  33. Paul C, Puig L, Kragballe K. 33.  et al. 2014. Transition to ustekinumab in patients with moderate-to-severe psoriasis and inadequate response to methotrexate: a randomized clinical trial (TRANSIT). Br. J. Dermatol. 170:425–34 [Google Scholar]
  34. Griffiths CE, Strober BE, van de Kerkhof P. 34.  et al. 2010. Comparison of ustekinumab and etanercept for moderate-to-severe psoriasis. N. Engl. J. Med. 362:118–28 [Google Scholar]
  35. Patsatsi A, Kyriakou A, Sotiriadis D. 35.  2013. Ustekinumab in nail psoriasis: an open-label, uncontrolled, nonrandomized study. J. Dermatol. Treat. 24:96–100 [Google Scholar]
  36. Rigopoulos D, Gregoriou S, Makris M, Ioannides D. 36.  2011. Efficacy of ustekinumab in nail psoriasis and improvement in nail-associated quality of life in a population treated with ustekinumab for cutaneous psoriasis: an open prospective unblinded study. Dermatology 223:325–29 [Google Scholar]
  37. Au SC, Goldminz AM, Kim N. 37.  et al. 2013. Investigator-initiated, open-label trial of ustekinumab for the treatment of moderate-to-severe palmoplantar psoriasis. J. Dermatol. Treat. 24:179–87 [Google Scholar]
  38. Bissonnette R, Nigen S, Langley RG. 38.  et al. 2014. Increased expression of IL-17A and limited involvement of IL-23 in patients with palmo-plantar (PP) pustular psoriasis or PP pustulosis; results from a randomised controlled trial. J. Eur. Acad. Dermatol. Venereol. 28:1298–305 [Google Scholar]
  39. Meng Y, Dongmei L, Yanbin P. 39.  et al. 2014. Systematic review and meta-analysis of ustekinumab for moderate to severe psoriasis. Clin. Exp. Dermatol. 39:696–707 [Google Scholar]
  40. Signorovitch JE, Betts KA, Yan YS. 40.  et al. 2015. Comparative efficacy of biological treatments for moderate-to-severe psoriasis: a network meta-analysis adjusting for cross-trial differences in reference arm response. Br. J. Dermatol. 172:504–12 [Google Scholar]
  41. Wilder EG, Patel M, Hebeler K, Menter A. 41.  2014. Ustekinumab treatment for psoriasis in 119 patients maintained on therapy for a minimum of one year: a review. J. Drugs Dermatol. 13:905–10 [Google Scholar]
  42. Sofen H, Smith S, Matheson RT. 42.  et al. 2014. Guselkumab (an IL-23-specific mAb) demonstrates clinical and molecular response in patients with moderate-to-severe psoriasis. J. Allergy Clin. Immunol. 133:1032–40 [Google Scholar]
  43. Kopp T, Riedl E, Bangert C. 43.  et al. 2015. Clinical improvement in psoriasis with specific targeting of interleukin-23. Nature 521:222–26 [Google Scholar]
  44. Kimball AB, Gordon KB, Langley RG. 44.  et al. 2008. Safety and efficacy of ABT-874, a fully human interleukin 12/23 monoclonal antibody, in the treatment of moderate to severe chronic plaque psoriasis: results of a randomized, placebo-controlled, phase 2 trial. Arch. Dermatol. 144:200–7 [Google Scholar]
  45. Strober BE, Crowley JJ, Yamauchi PS. 45.  et al. 2011. Efficacy and safety results from a phase III, randomized controlled trial comparing the safety and efficacy of briakinumab with etanercept and placebo in patients with moderate to severe chronic plaque psoriasis. Br. J. Dermatol. 165:661–68 [Google Scholar]
  46. Reich K, Langley RG, Papp KA. 46.  et al. 2011. A 52-week trial comparing briakinumab with methotrexate in patients with psoriasis. N. Engl. J. Med. 365:1586–96 [Google Scholar]
  47. Gordon KB, Langley RG, Gottlieb AB. 47.  et al. 2012. A phase III, randomized, controlled trial of the fully human IL-12/23 mAb briakinumab in moderate-to-severe psoriasis. J. Investig. Dermatol. 132:304–14 [Google Scholar]
  48. Langley RG, Papp K, Gottlieb AB. 48.  et al. 2013. Safety results from a pooled analysis of randomized, controlled phase II and III clinical trials and interim data from an open-label extension trial of the interleukin-12/23 monoclonal antibody, briakinumab, in moderate to severe psoriasis. J. Eur. Acad. Dermatol. Venereol. 27:1252–61 [Google Scholar]
  49. Raychaudhuri SP, Raychaudhuri SK, Genovese MC. 49.  2012. IL-17 receptor and its functional significance in psoriatic arthritis. Mol. Cell. Biochem. 359:419–29 [Google Scholar]
  50. Celis R, Planell N, Fernandez-Sueiro JL. 50.  et al. 2012. Synovial cytokine expression in psoriatic arthritis and associations with lymphoid neogenesis and clinical features. Arthritis Res. Ther. 14:R93 [Google Scholar]
  51. Felson DT, Anderson JJ, Boers M. 51.  et al. 1995. American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum. 38:727–35 [Google Scholar]
  52. Gottlieb A, Menter A, Mendelsohn A. 52.  et al. 2009. Ustekinumab, a human interleukin 12/23 monoclonal antibody, for psoriatic arthritis: randomised, double-blind, placebo-controlled, crossover trial. Lancet 373:633–40 [Google Scholar]
  53. McInnes IB, Kavanaugh A, Gottlieb AB. 53.  et al. 2013. 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 382:780–89 [Google Scholar]
  54. Ritchlin C, Rahman P, Kavanaugh A. 54.  et al. 2014. 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, randomised PSUMMIT 2 trial. Ann. Rheum. Dis. 73:990–99 [Google Scholar]
  55. Kavanaugh A, Ritchlin C, Rahman P. 55.  et al. 2014. 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. 73:1000–6 [Google Scholar]
  56. McInnes IB, Sieper J, Braun J. 56.  et al. 2014. Efficacy and safety of secukinumab, a fully human anti-interleukin-17A monoclonal antibody, in patients with moderate-to-severe psoriatic arthritis: a 24-week, randomised, double-blind, placebo-controlled, phase II proof-of-concept trial. Ann. Rheum. Dis. 73:349–56 [Google Scholar]
  57. McInnes IB MP, Kirkham B, Kavanaugh A. 57.  et al. 2014. Secukinumab, a human anti-interleukin-17A monoclonal antibody, improves active psoriatic arthritis: 24-week efficacy and safety data from a phase 3 randomized, multicenter, double-blind, placebo-controlled study using subcutaneous dosing. Arthritis Rheumatol. 66:3529–40 [Google Scholar]
  58. Mease PJ MI, Kirkham B, Kavanaugh A. 58.  et al. 2014. Secukinumab, a human anti–interleukin-17A monoclonal antibody, improves active psoriatic arthritis and inhibits radiographic progression: efficacy and safety data from a phase 3 randomized, multicenter, double-blind, placebo-controlled study. Arthritis Rheumatol. 66:3529–40 [Google Scholar]
  59. Mease PJ, Genovese MC, Greenwald MW. 59.  et al. 2014. Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N. Engl. J. Med. 370:2295–306 [Google Scholar]
  60. Smith JA, Colbert RA. 60.  2014. Review: the interleukin-23/interleukin-17 axis in spondyloarthritis pathogenesis: Th17 and beyond. Arthritis Rheumatol. 66:231–41 [Google Scholar]
  61. Reveille JD. 61.  2012. Genetics of spondyloarthritis—beyond the MHC. Nat. Rev. Rheumatol. 8:296–304 [Google Scholar]
  62. Shen H, Goodall JC, Hill Gaston JS. 62.  2009. Frequency and phenotype of peripheral blood Th17 cells in ankylosing spondylitis and rheumatoid arthritis. Arthritis Rheum. 60:1647–56 [Google Scholar]
  63. Colbert RA, Tran TM, Layh-Schmitt G. 63.  2014. HLA-B27 misfolding and ankylosing spondylitis. Mol. Immunol. 57:44–51 [Google Scholar]
  64. Sherlock JP, Joyce-Shaikh B, Turner SP. 64.  et al. 2012. IL-23 induces spondyloarthropathy by acting on ROR-γt+ CD3+CD4–CD8– entheseal resident T cells. Nat. Med. 18:1069–76Discovery in mouse spondylarthropathy model of a new T cell subset sensitive to IL-23. [Google Scholar]
  65. Baeten D, Baraliakos X, Braun J. 65.  et al. 2013. Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial. Lancet 382:1705–13 [Google Scholar]
  66. Poddubnyy D, Hermann KG, Callhoff J. 66.  et al. 2014. Ustekinumab for the treatment of patients with active ankylosing spondylitis: results of a 28-week, prospective, open-label, proof-of-concept study (TOPAS). Ann. Rheum. Dis. 73:817–23 [Google Scholar]
  67. Baraliakos X, Braun J, Laurent DD. 67.  et al. 2011. Interleukin-17A blockade with secukinumab reduces spinal inflammation in patients with ankylosing spondylitis as early as week 6, as detected by magnetic resonance imaging. Arthritis Rheum. 63:Suppl. 102486D [Google Scholar]
  68. Lubberts E, Koenders MI, Oppers-Walgreen B. 68.  et al. 2004. Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. Arthritis Rheum. 50:650–59 [Google Scholar]
  69. Malfait AM, Butler DM, Presky DH. 69.  et al. 1998. Blockade of IL-12 during the induction of collagen-induced arthritis (CIA) markedly attenuates the severity of the arthritis. Clin. Exp. Immunol. 111:377–83 [Google Scholar]
  70. Kotake S, Udagawa N, Takahashi N. 70.  et al. 1999. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J. Clin. Invest. 103:1345–52 [Google Scholar]
  71. Honorati MC, Meliconi R, Pulsatelli L. 71.  et al. 2001. High in vivo expression of interleukin-17 receptor in synovial endothelial cells and chondrocytes from arthritis patients. Rheumatology 40:522–27 [Google Scholar]
  72. Shahrara S, Pickens SR, Dorfleutner A, Pope RM. 72.  2009. IL-17 induces monocyte migration in rheumatoid arthritis. J. Immunol. 182:3884–91 [Google Scholar]
  73. Melis L, Vandooren B, Kruithof E. 73.  et al. 2010. Systemic levels of IL-23 are strongly associated with disease activity in rheumatoid arthritis but not spondyloarthritis. Ann. Rheum. Dis. 69:618–23 [Google Scholar]
  74. Genovese MC, Durez P, Richards HB. 74.  et al. 2013. Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study. Ann. Rheum. Dis. 72:863–69 [Google Scholar]
  75. Genovese MC, Durez P, Richards HB. 75.  et al. 2014. One-year efficacy and safety results of secukinumab in patients with rheumatoid arthritis: phase II, dose-finding, double-blind, randomized, placebo-controlled study. J. Rheumatol. 41:414–21 [Google Scholar]
  76. Genovese MC, Greenwald M, Cho CS. 76.  et al. 2014. A phase II randomized study of subcutaneous ixekizumab, an anti-interleukin-17 monoclonal antibody, in rheumatoid arthritis patients who were naive to biologic agents or had an inadequate response to tumor necrosis factor inhibitors. Arthritis Rheumatol. 66:1693–704 [Google Scholar]
  77. Martin DA, Churchill M, Flores-Suarez L. 77.  et al. 2013. A phase Ib multiple ascending dose study evaluating safety, pharmacokinetics, and early clinical response of brodalumab, a human anti-IL-17R antibody, in methotrexate-resistant rheumatoid arthritis. Arthritis Res. Ther. 15:R164 [Google Scholar]
  78. Duerr RH, Taylor KD, Brant SR. 78.  et al. 2006. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314:1461–63 [Google Scholar]
  79. Neurath MF, Fuss I, Kelsall BL. 79.  et al. 1995. Antibodies to interleukin 12 abrogate established experimental colitis in mice. J. Exp. Med. 182:1281–90 [Google Scholar]
  80. Kobayashi T, Okamoto S, Hisamatsu T. 80.  et al. 2008. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn's disease. Gut 57:1682–89 [Google Scholar]
  81. Hueber W, Sands BE, Lewitzky S. 81.  et al. 2012. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 61:1693–700 [Google Scholar]
  82. Colombel JF, Sendid B, Jouault T, Poulain D. 82.  2013. Secukinumab failure in Crohn's disease: the yeast connection?. Gut 62:800–1 [Google Scholar]
  83. Targan SR FB, Vermeire S, Panaccione R. 83.  et al. 2012. A randomized, double-blind, placebo-controlled study to evaluate the safety, tolerability, and efficacy of AMG 827 in subjects with moderate to severe Crohn's disease. Gastroenterology 143:e26 (Abstr.) [Google Scholar]
  84. Sandborn WJ, Feagan BG, Fedorak RN. 84.  et al. 2008. A randomized trial of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology 135:1130–41 [Google Scholar]
  85. Sandborn WJ, Gasink C, Gao LL. 85.  et al. 2012. Ustekinumab induction and maintenance therapy in refractory Crohn's disease. N. Engl. J. Med. 367:1519–28 [Google Scholar]
  86. Ringkowski S, Thomas PS, Herbert C. 86.  2014. Interleukin-12 family cytokines and sarcoidosis. Front. Pharmacol. 5:233 [Google Scholar]
  87. Puel A, Cypowyj S, Bustamante J. 87.  et al. 2011. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 332:65–68 [Google Scholar]
  88. Filipe-Santos O, Bustamante J, Chapgier A. 88.  et al. 2006. Inborn errors of IL-12/23- and IFN-gamma-mediated immunity: molecular, cellular, and clinical features. Semin. Immunol. 18:347–61 [Google Scholar]
  89. Tsai TF, Ho V, Song M. 89.  et al. 2012. The safety of ustekinumab treatment in patients with moderate-to-severe psoriasis and latent tuberculosis infection. Br. J. Dermatol. 167:1145–52 [Google Scholar]
  90. Adelzadeh L, Jourabchi N, Wu JJ. 90.  2014. The risk of herpes zoster during biological therapy for psoriasis and other inflammatory conditions. J. Eur. Acad. Dermatol. Venereol. 28:846–52 [Google Scholar]
  91. Papp KA, Griffiths CE, Gordon K. 91.  et al. 2013. Long-term safety of ustekinumab in patients with moderate-to-severe psoriasis: final results from 5 years of follow-up. Br. J. Dermatol. 168:844–54 [Google Scholar]
  92. Ryan C, Leonardi CL, Krueger JG. 92.  et al. 2011. Association between biologic therapies for chronic plaque psoriasis and cardiovascular events: a meta-analysis of randomized controlled trials. JAMA 306:864–71 [Google Scholar]
  93. Tzellos T, Kyrgidis A, Zouboulis CC. 93.  2013. 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. 27:622–27 [Google Scholar]
  94. Dommasch ED, Troxel AB, Gelfand JM. 94.  2014. Counterpoint: a tale of two meta-analyses revisited. J. Am. Acad. Dermatol. 70:381–83 [Google Scholar]
  95. Gottlieb AB, Kalb RE, Langley RG. 95.  et al. 2014. Safety observations in 12095 patients with psoriasis enrolled in an international registry (PSOLAR): experience with infliximab and other systemic and biologic therapies. J. Drugs Dermatol. 13:1441–48 [Google Scholar]
  96. Reich K, Langley RG, Lebwohl M. 96.  et al. 2011. Cardiovascular safety of ustekinumab in patients with moderate to severe psoriasis: results of integrated analyses of data from phase II and III clinical studies. Br. J. Dermatol. 164:862–72 [Google Scholar]
  97. Fischer JA, Hueber AJ, Wilson S. 97.  et al. 2015. Combined inhibition of tumor necrosis factor alpha and interleukin-17 as a therapeutic opportunity in rheumatoid arthritis: development and characterization of a novel bispecific antibody. Arthritis Rheumatol. 67:51–62Interesting study demonstrating promising results of antibodies against both IL-17 and TNFα. [Google Scholar]
  98. Solt LA, Kumar N, Nuhant P. 98.  et al. 2011. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature 472:491–94 [Google Scholar]
  99. Mease PJ. 99.  2015. Inhibition of interleukin-17, interleukin-23 and the TH17 cell pathway in the treatment of psoriatic arthritis and psoriasis. Curr. Opin. Rheumatol. 27:127–33 [Google Scholar]
  100. Patel DD, Lee DM, Kolbinger F, Antoni C. 100.  2013. Effect of IL-17A blockade with secukinumab in autoimmune diseases. Ann. Rheum. Dis. 72:Suppl. 2) ii116–23 [Google Scholar]
/content/journals/10.1146/annurev-med-051914-021944
Loading
/content/journals/10.1146/annurev-med-051914-021944
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error