1,496
Views
24
CrossRef citations to date
0
Altmetric
Review

Biological therapy targeting the IL-23/IL-17 axis in inflammatory bowel disease

, , &
Pages 31-47 | Received 30 Jun 2016, Accepted 04 Nov 2016, Published online: 18 Nov 2016

References

  • Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007 Jul;448(7152):427–434. DOI:10.1038/nature06005
  • Lee JC. Predicting the course of IBD: light at the end of the tunnel? Dig Dis. 2012;30(Suppl 1):95–99. DOI:10.1159/000341132
  • Nielsen OH. New strategies for treatment of inflammatory bowel disease. Front Med (Lausanne). 2014;1:3.
  • Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol. 2003 Jul;3(7):521–533. DOI:10.1038/nri1132
  • Mannon PJ, Fuss IJ, Mayer L, et al. Anti-interleukin-12 antibody for active Crohn’s disease. N Engl J Med. 2004 Nov;351(20):2069–2079. DOI:10.1056/NEJMoa033402
  • Hunter CA. New IL-12-family members: IL-23 and IL-27, cytokines with divergent functions. Nat Rev Immunol. 2005 Jul;5(7):521–531. DOI:10.1038/nri1648
  • Duerr RH, Taylor KD, Brant SR, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006 Dec;314(5804):1461–1463. DOI:10.1126/science.1135245
  • Hue S, Ahern P, Buonocore S, et al. Interleukin-23 drives innate and T cell-mediated intestinal inflammation. J Exp Med. 2006 Oct;203(11):2473–2483. DOI:10.1084/jem.20061099
  • Kullberg MC, Jankovic D, Feng CG, et al. IL-23 plays a key role in Helicobacter hepaticus-induced T cell-dependent colitis. J Exp Med. 2006 Oct;203(11):2485–2494. DOI:10.1084/jem.20061082
  • Ahern PP, Izcue A, Maloy KJ, et al. The interleukin-23 axis in intestinal inflammation. Immunol Rev. 2008 Dec;226:147–159.
  • Maloy KJ. The Interleukin-23 /Interleukin-17 axis in intestinal inflammation. J Intern Med. 2008 Jun;263(6):584–590. DOI:10.1111/j.1365-2796.2008.01950.x
  • Kobayashi T, Okamoto S, Hisamatsu T, et al. IL23 differentially regulates the Th1/Th17 balance in ulcerative colitis and Crohn’s disease. Gut. 2008 Dec;57(12):1682–1689. DOI:10.1136/gut.2007.135053
  • Abraham C, Cho J. Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflamm Bowel Dis. 2009 Jul;15(7):1090–1100. DOI:10.1002/ibd.20894
  • Sarra M, Pallone F, Macdonald TT, et al. IL-23/IL-17 axis in IBD. Inflamm Bowel Dis. 2010 Oct;16(10):1808–1813. DOI:10.1002/ibd.21248
  • Shen W, Durum SK. Synergy of IL-23 and Th17 cytokines: new light on inflammatory bowel disease. Neurochem Res. 2010 Jun;35(6):940–946. DOI:10.1007/s11064-009-0091-9
  • Fitzpatrick LR. Novel pharmacological approaches for inflammatory bowel disease: targeting key intracellular pathways and the IL-23/IL-17 axis. Int J Inflam. 2012;2012:1–8. DOI:10.1155/2012/389404
  • Geremia A, Jewell DP. The IL-23/IL-17 pathway in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2012 Apr;6(2):223–237. DOI:10.1586/egh.11.107
  • Yang J, Sundrud MS, Skepner J, et al. Targeting Th17 cells in autoimmune diseases. Trends Pharmacol Sci. 2014 Oct;35(10):493–500. DOI:10.1016/j.tips.2014.07.006
  • Jiang W, Su J, Zhang X, et al. Elevated levels of Th17 cells and Th17-related cytokines are associated with disease activity in patients with inflammatory bowel disease. Inflamm Res. 2014 Nov;63(11):943–950. DOI:10.1007/s00011-014-0768-7
  • Maxwell JR, Zhang Y, Brown WA, et al. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity. 2015 Oct;43(4):739–750. DOI:10.1016/j.immuni.2015.08.019
  • Davies JM, Abreu MT. The innate immune system and inflammatory bowel disease. Scand J Gastroenterol. 2015 Jan;50(1):24–33. DOI:10.3109/00365521.2014.966321
  • Cătană CS, Berindan Neagoe I, Cozma V, et al. Contribution of the IL-17/IL-23 axis to the pathogenesis of inflammatory bowel disease. World J Gastroenterol. 2015 May;21(19):5823–5830. DOI:10.3748/wjg.v21.i19.5823
  • Duvallet E, Semerano L, Assier E, et al. Interleukin-23: a key cytokine in inflammatory diseases. Ann Med. 2011 Nov;43(7):503–511. DOI:10.3109/07853890.2011.577093
  • Blanco P, Palucka AK, Pascual V, et al. Dendritic cells and cytokines in human inflammatory and autoimmune diseases. Cytokine Growth Factor Rev. 2008 Feb;19(1):41–52. DOI:10.1016/j.cytogfr.2007.10.004
  • Noack M, Miossec P. Th17 and regulatory T cell balance in autoimmune and inflammatory diseases. Autoimmun Rev. 2014 Jun;13(6):668–677. DOI:10.1016/j.autrev.2013.12.004
  • Zhang Z, Hinrichs DJ, Lu H, et al. After interleukin-12p40, are interleukin-23 and interleukin-17 the next therapeutic targets for inflammatory bowel disease? Int Immunopharmacol. 2007 Apr;7(4):409–416. DOI:10.1016/j.intimp.2006.09.024
  • Sakaguchi S, Miyara M, Costantino CM, et al. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010 Jul;10(7):490–500. DOI:10.1038/nri2785
  • Globig AM, Hennecke N, Martin B, et al. Comprehensive intestinal T helper cell profiling reveals specific accumulation of IFN-gamma+IL-17+coproducing CD4+ T cells in active inflammatory bowel disease. Inflamm Bowel Dis. 2014 Dec;20(12):2321–2329. DOI:10.1097/MIB.0000000000000210
  • Mucida D, Park Y, Kim G, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007 Jul 13;317(5835):256–260.
  • Ueno A, Jijon H, Chan R, et al. Increased prevalence of circulating novel IL-17 secreting Foxp3 expressing CD4+ T cells and defective suppressive function of circulating Foxp3+ regulatory cells support plasticity between Th17 and regulatory T cells in inflammatory bowel disease patients. Inflamm Bowel Dis. 2013 Nov;19(12):2522–2534. DOI:10.1097/MIB.0b013e3182a85709
  • McGeachy MJ, Bak-Jensen KS, Chen Y, et al. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 2007 Dec;8(12):1390–1397. DOI:10.1038/ni1539
  • Ramesh R, Kozhaya L, McKevitt K, et al. Pro-inflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids. J Exp Med. 2014 Jan 13;211(1):89–104.
  • Fuss IJ, Becker C, Yang Z, et al. Both IL-12p70 and IL-23 are synthesized during active Crohn’s disease and are down-regulated by treatment with anti-IL-12 p40 monoclonal antibody. Inflamm Bowel Dis. 2006 Jan;12(1):9–15.
  • O’Connor W, Kamanaka M, Booth CJ, et al. A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol. 2009 Jun;10(6):603–609. DOI:10.1038/ni.1736
  • Panaccione R, Sandborn WJ, Gordon GL, et al. Briakinumab for treatment of Crohn’s disease: results of a randomized trial. Inflamm Bowel Dis. 2015 Jun;21(6):1329–1340. DOI:10.1097/MIB.0000000000000366
  • Benson JM, Sachs CW, Treacy G, et al. Therapeutic targeting of the IL-12/23 pathways: generation and characterization of ustekinumab. Nat Biotechnol. 2011 Jul;29(7):615–624. DOI:10.1038/nbt.1903
  • Sandborn WJ, Feagan BG, Fedorak RN, et al. A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn’s disease. Gastroenterology. 2008 Oct;135(4):1130–1141. DOI:10.1053/j.gastro.2008.07.014
  • Elliott M, Benson J, Blank M, et al. Ustekinumab: lessons learned from targeting interleukin-12/23p40 in immune-mediated diseases. Ann N Y Acad Sci. 2009 Dec;1182:97–110.
  • Toedter GP, Blank M, Lang Y, et al. Relationship of C-reactive protein with clinical response after therapy with ustekinumab in Crohn’s disease. Am J Gastroenterol. 2009 Nov;104(11):2768–2773. DOI:10.1038/ajg.2009.454
  • Su C, Lichtenstein GR, Krok K, et al. A meta-analysis of the placebo rates of remission and response in clinical trials of active Crohn’s disease. Gastroenterology. 2004 May;126(5):1257–1269.
  • Sandborn WJ, Gasink C, Gao LL, et al. Ustekinumab induction and maintenance therapy in refractory Crohn’s disease. N Engl J Med. 2012 Oct;367(16):1519–1528. DOI:10.1056/NEJMoa1203572
  • Rutgeerts P, Gasink C, Blank M, et al. A multicentre, double-blind, placebo-controlled phase 3 study of ustekinumab, a human IL-12/23p40 mab, in moderate-to-severe CD refractory to anti-tumour necrosis factor alpha: UNITI-1. Congress of ECCO. Amsterdam;2016.
  • Feagan B A multicenter, randomized, double-blind, placebo-controlled phase 3 study of ustekinumab, a human monoclonal antibody to IL-12/23P40, in patients with moderately- to severely-active Crohn’s Disease who are naive or not refractory to anti-TNFa: results from the UNITI-2 study.ACG Meeting 2015. Honolulu, Hawai, USA; 2015.
  • Gasink C, Lang Y, Jacobstein D, et al. Assessment of serum C-reactive protein, faecal lactoferrin and faecal calprotectin in patients with Crohn’s disease: results from the UNITI-1 and UNITI-2 Ustekinumab induction studies. 11th Congress of ECCO. Amsterdam; 2016.
  • Sandborn W, Feagan B, Gasink C, et al. A phase 3, randomized, multicenter, double-blind, placebo-controlled, study of ustekinumab maintenance therapy in moderate-severe crohn’s disease patients: results from IM-UNITI. DDW 2016. San Diego; 2016. DOI:10.1016/S0016-5085(16)30623-0
  • Clinical trials database. [cited 2016 April 6]. Available from: www.clinicaltrials.gov
  • Kopylov U, Afif W, Cohen A, et al. Subcutaneous ustekinumab for the treatment of anti-TNF resistant Crohn’s disease–the McGill experience. J Crohns Colitis. 2014 Nov;8(11):1516–1522. DOI:10.1016/j.crohns.2014.06.005
  • Battat R, Kopylov U, Bessissow T, et al. Association of ustekinumab trough concentrations with clinical, biochemical and endoscopic outcomes in Crohn’s disease. DDW 2016. San Diego; 2016.
  • Wils P, Bouhnik Y, Michetti P, et al. Subcutaneous ustekinumab provides clinical benefit for two-thirds of patients with crohn’s disease refractory to anti-tumor necrosis factor agents. Clin Gastroenterol Hepatol. 2016 Feb;14(2):242–250. DOI:10.1016/j.cgh.2015.09.018
  • Ginard D, Khorrami S, Marin I, et al. Effectiveness and safety of ustekinumab asrescue therapy in multi-drug resistant Crohn’s disease. 10th Congress of ECCO. Barcelona; 2015.
  • Herrera C, Jimenez C, Casellas F, et al. Ustekinumab in super-refractory Crohn’s disease. 10th Congress of ECCO. Barcelona; 2015.
  • Harris KA, Horst S, Gadani A, et al. Patients with refractory crohn’s disease successfully treated with ustekinumab. Inflamm Bowel Dis. 2016 Feb;22(2):397–401. DOI:10.1097/MIB.0000000000000624
  • Simon EG, Ghosh S, Iacucci M, et al. Ustekinumab for the treatment of Crohn’s disease: can it find its niche? Therap Adv Gastroenterol. 2016 Jan;9(1):26–36. DOI:10.1177/1756283X15618130
  • Raine T, Kaser A. Seventeen in Crohn’s disease: less prime than we thought? Gut. 2012 Dec;61(12):1653–1654. DOI:10.1136/gutjnl-2012-302525
  • Khanna R, Feagan BG. Ustekinumab for the treatment of Crohn’s disease. Immunotherapy. 2013 Aug;5(8):803–815. DOI:10.2217/imt.13.81
  • Lebwohl M, Leonardi C, Griffiths CE, et al. Long-term safety experience of ustekinumab in patients with moderate-to-severe psoriasis (Part I of II): results from analyses of general safety parameters from pooled Phase 2 and 3 clinical trials. J Am Acad Dermatol. 2012 May;66(5):731–741. DOI:10.1016/j.jaad.2011.06.011
  • Gordon KB, Papp KA, Langley RG, et al. Long-term safety experience of ustekinumab in patients with moderate to severe psoriasis (Part II of II): results from analyses of infections and malignancy from pooled phase II and III clinical trials. J Am Acad Dermatol. 2012 May;66(5):742–751. DOI:10.1016/j.jaad.2011.06.041
  • Khanna R, Preiss JC, MacDonald JK, et al. Anti-IL-12/23p40 antibodies for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2015;5:CD007572.
  • Adedokun OJ, Xu Z, Gasink C, et al. Pharmacokinetics and exposure-response relationships of intravenously administered ustekinumab during induction treatment in patients with Crohn’s Disease: results from the UNITI-1 and UNITI-2 studies. 11th Congress of ECCO. Amsterdam; 2016.
  • Badat Y, Meissner WG, Laharie D. Demyelination in a patient receiving ustekinumab for refractory Crohn’s disease. J Crohns Colitis. 2014 Sep;8(9):1138–1139. DOI:10.1016/j.crohns.2014.02.004
  • Ehmann LM, Tillack-Schreiber C, Brand S, et al. Malignant melanoma during ustekinumab therapy of Crohn’s disease. Inflamm Bowel Dis. 2012 Jan;18(1):E199–E200. DOI:10.1002/ibd.21877
  • Janssen Biotech Inc. Stelara solution for injecting PRESCRIBING INFORMATION. [cited 2016 April 6]. Available from: http://www.janssen-pi.co.uk/stelara
  • Janssen Biotech Inc. Stelara prescribing information. [cited 2016 April 6]. Available from: http://www.stelarahcp.com/pdf/PrescribingInformation.pdf
  • 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 Aug;306(8):864–871. DOI:10.1001/jama.2011.1211
  • Battat R, Kopylov U, Bessissow T, et al. Association of ustekinumab through concentrations with clinical, biochemical and endoscopic outcome. 11th Congress of ECCO. Amsterdam; 2016.
  • Galli-Novak E, Mook SC, Büning J, et al. Successful pregnancy outcome under prolonged ustekinumab treatment in a patient with Crohn’s disease and paradoxical psoriasis. J Eur Acad Dermatol Venereol. 2015 Nov. DOI:10.1111/jdv.13499
  • Andrulonis R, Ferris LK. Treatment of severe psoriasis with ustekinumab during pregnancy. J Drugs Dermatol. 2012 Oct;11(10):1240.
  • Fotiadou C, Lazaridou E, Sotiriou E, et al. Spontaneous abortion during ustekinumab therapy. J Dermatol Case Rep. 2012 Dec;6(4):105–107. DOI:10.3315/jdcr.2012.1116
  • Alsenaid A, Prinz JC. Inadvertent pregnancy during ustekinumab therapy in a patient with plaque psoriasis and impetigo herpetiformis. J Eur Acad Dermatol Venereol. 2016 Mar;30(3):488–490. DOI:10.1111/jdv.12872
  • Rocha K, Piccinin MC, Kalache LF, et al. Pregnancy during ustekinumab treatment for severe psoriasis. Dermatology. 2015;231(2):103–104. DOI:10.1159/000380880
  • Sheeran C, Nicolopoulos J. Pregnancy outcomes of two patients exposed to ustekinumab in the first trimester. Australas J Dermatol. 2014 Aug;55(3):235–236. DOI:10.1111/ajd.12214
  • JanssenMD. Stelara prescribing information. [cited 2016 April 6]. Available from: http://www.stelarahcp.com/pdf/PrescribingInformation.pdf
  • Billiet T, Rutgeerts P, Ferrante M, et al. Targeting TNF-α for the treatment of inflammatory bowel disease. Expert Opin Biol Ther. 2014 Jan;14(1):75–101. DOI:10.1517/14712598.2014.858695
  • Scherl EJ, Kumar S, Warren RU. Review of the safety and efficacy of ustekinumab. Therap Adv Gastroenterol. 2010 Sep;3(5):321–328. DOI:10.1177/1756283X10374216
  • Ghetie V, Ward ES. Transcytosis and catabolism of antibody. Immunol Res. 2002;25(2):97–113. DOI:10.1385/IR:25:2:097
  • Billiet T, Dreesen E, Cleynen I, et al. A genetic variation in the neonatal fc-receptor affects anti-tnf drug concentrations in inflammatory bowel disease. Am J Gastroenterol. 2016;111:1438–1445. DOI:10.1038/ajg.2016.306
  • Wang Y, Tian Z, Thirumalai D, et al. Neonatal Fc receptor (FcRn): a novel target for therapeutic antibodies and antibody engineering. J Drug Target. 2014 May;22(4):269–278. DOI:10.3109/1061186X.2013.875030
  • Schoch A, Kettenberger H, Mundigl O, et al. Charge-mediated influence of the antibody variable domain on FcRn-dependent pharmacokinetics. Proc Natl Acad Sci U S A. 2015 May 12;112(19):5997–6002.
  • Ginard D, Khorrami S, Marin I, et al. Effectiveness and safety of ustekinumab as rescue therapy in multi-drug resistant Crohn’s disease. 10th Congress of ECCO. Barcelona; 2015.
  • Kaser A. Not all monoclonals are created equal - lessons from failed drug trials in Crohn’s disease. Best Pract Res Clin Gastroenterol. 2014 Jun;28(3):437–449. DOI:10.1016/j.bpg.2014.04.005
  • Cleynen I, van Moerkercke W, Billiet T, et al. Characteristics of skin lesions associated with anti-tumor necrosis factor therapy in patients with inflammatory bowel disease: a cohort study. Ann Intern Med. 2016 Jan;164(1):10–22. DOI:10.7326/M15-0729
  • Tillack C, Ehmann LM, Friedrich M, et al. Anti-TNF antibody-induced psoriasiform skin lesions in patients with inflammatory bowel disease are characterised by interferon-γ-expressing Th1 cells and IL-17A/IL-22-expressing Th17 cells and respond to anti-IL-12/IL-23 antibody treatment. Gut. 2014 Apr;63(4):567–577. DOI:10.1136/gutjnl-2012-302853
  • Burakoff R, Barish CF, Riff D, et al. A phase 1/2A trial of STA 5326, an oral interleukin-12/23 inhibitor, in patients with active moderate to severe Crohn’s disease. Inflamm Bowel Dis. 2006 Jul;12(7):558–565. DOI:10.1097/01.ibd.0000225337.14356.31
  • Sands BE, Jacobson EW, Sylwestrowicz T, et al. Randomized, double-blind, placebo-controlled trial of the oral interleukin-12/23 inhibitor apilimod mesylate for treatment of active Crohn’s disease. Inflamm Bowel Dis. 2010 Jul;16(7):1209–1218. DOI:10.1002/ibd.21159
  • Singh S, Kroe-Barrett RR, Canada KA, et al. Selective targeting of the IL23 pathway: generation and characterization of a novel high-affinity humanized anti-IL23A antibody. MAbs. 2015;7(4):778–791. DOI:10.1080/19420862.2015.1032491
  • Sands B, Chen J, Penney J. A randomized, double-blind placebo-controlled phase 2a induction study of MEDI2070 (anti-p19 antibody) in patients with active Crohn’s disease who have failed anti-TNF antibody therapy. J Crohn’s Colitis. 2015;9(Suppl 1):S15–S16. DOI:10.1093/ecco-jcc/jju027.025
  • 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 Jul;136(1):116–24.e7. DOI:10.1016/j.jaci.2015.01.018
  • Feagan B, Sandborn W, Panes J, et al. Efficacy and safety of induction therapy with the selective IL-23 inhibitor risankizumab (BI 655066) in patients with moderate-to-severe 
Crohn’s disease: results of a randomized, double-blind, placebo controlled Phase II study. DDW 2016. San Diego; 2016.
  • Merck. [cited 2016 April 6]. Available from: http://www.merck.com/licensing/our-partnership/sunpharma_partnership.html
  • Papp K, Thaçi D, Reich K, et al. Tildrakizumab (MK-3222), an anti-interleukin-23p19 monoclonal antibody, improves psoriasis in a phase IIb randomized placebo-controlled trial. Br J Dermatol. 2015 Oct;173(4):930–939. DOI:10.1111/bjd.13932
  • Gordon KB, Duffin KC, Bissonnette R, et al. A phase 2 trial of guselkumab versus adalimumab for plaque psoriasis. N Engl J Med. 2015 Jul;373(2):136–144. DOI:10.1056/NEJMoa1501646
  • Sabat R, Witte E, Witte K, et al. IL-22 and IL-17: an overview. In: Quesniaux V, Ryffel B, Padova F, editors IL-17, IL-22 and their producing cells: role in inflammation and autoimmunity progress in inflammation research. Basel: Springer 2013:p.11–35.
  • Korn T, Bettelli E, Oukka M, et al. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. DOI:10.1146/annurev.immunol.021908.132710
  • Seiderer J, Elben I, Diegelmann J, et al. Role of the novel Th17 cytokine IL-17F in inflammatory bowel disease (IBD): upregulated colonic IL-17F expression in active Crohn’s disease and analysis of the IL17F p.His161Arg polymorphism in IBD. Inflamm Bowel Dis. 2008 Apr;14(4):437–445. DOI:10.1002/ibd.20339
  • Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 2003 Jan;52(1):65–70.
  • Targan S, Feagan B, Vermeire S, et al. 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. 2012;143(3):e26.
  • Hueber W, Patel DD, Dryja T, et al. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2010 Oct;2(52):52ra72. DOI:10.1126/scitranslmed.3001107
  • Hueber W, Sands BE, Lewitzky S, et al. 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. 2012 Dec;61(12):1693–1700. DOI:10.1136/gutjnl-2011-301668
  • Puel A, Cypowyj S, Bustamante J, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science. 2011 Apr;332(6025):65–68. DOI:10.1126/science.1200439
  • Zhang H, Xia B, Li J, et al. Expression and clinical significance of IL-17 and IL-17 receptor in ulcerative colitis. J Huazhong Univ Sci Technol Med Sci. 2016 Feb;36(1):37–40. DOI:10.1007/s11596-016-1538-2

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.