817
Views
38
CrossRef citations to date
0
Altmetric
Review

Adverse events in IBD therapy: the 2018 update

, &
Pages 1183-1191 | Received 13 May 2018, Accepted 24 Oct 2018, Published online: 15 Nov 2018

References

  • Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. The Lancet. 2017;390(10114):2769–2778.
  • Yu H, MacIsaac D, Wong JJ, et al. Market share and costs of biologic therapies for inflammatory bowel disease in the USA. Aliment Pharmacol Ther. 2018;47(3):364–370.
  • Prefontaine E, Sutherland LR, Macdonald JK, et al. Azathioprine or 6-mercaptopurine for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2009;1:CD000067.
  • Timmer A, McDonald JW, Tsoulis DJ, et al. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2012;9:CD000478.
  • Kennedy NA, Rhatigan E, Arnott ID, et al. A trial of mercaptopurine is a safe strategy in patients with inflammatory bowel disease intolerant to azathioprine: an observational study, systematic review and meta-analysis. Aliment Pharmacol Ther. 2013;38(10):1255–1266.
  • Lichtenstein GR, Abreu MT, Cohen R, et al. American gastroenterological association institute medical position statement on corticosteroids, immunomodulators, and infliximab in inflammatory bowel disease. Gastroenterology. 2006;130(3):935–939.
  • Chaparro M, Ordas I, Cabre E, et al. Safety of thiopurine therapy in inflammatory bowel disease: long-term follow-up study of 3931 patients. Inflamm Bowel Dis. 2013;19(7):1404–1410.
  • Dubinsky MC, Feldman EJ, Abreu MT, et al. Thioguanine: a potential alternate thiopurine for IBD patients allergic to 6-mercaptopurine or azathioprine. Am J Gastroenterol. 2003;98(5):1058–1063.
  • Mayo JM, Colmenarejo MB, Vaquerizo PJ, et al. Hypersensitivity reaction to azathioprine in a patient with ulcerative colitis. Infrequent manifestations. Inflamm Bowel Dis. 2004;10(5):700.
  • Ansari A, Arenas M, Greenfield SM, et al. Prospective evaluation of the pharmacogenetics of azathioprine in the treatment of inflammatory bowel disease. Aliment Pharmacol Ther. 2008;28(8):973–983.
  • Bidinger JJ, Sky K, Battafarano DF, et al. The cutaneous and systemic manifestations of azathioprine hypersensitivity syndrome. J Am Acad Dermatol. 2011;65(1):184–191.
  • Costantino G, Furfaro F, Belvedere A, et al. Thiopurine treatment in inflammatory bowel disease: response predictors, safety, and withdrawal in follow-up. J Crohns Colitis. 2012;6(5):588–596.
  • Meggitt SJ, Anstey AV, Mohd Mustapa MF, et al. British association of dermatologists’ guidelines for the safe and effective prescribing of azathioprine 2011. Br J Dermatol. 2011;165(4):711–734.
  • Aberra FN, Lichtenstein GR. Methods to avoid infections in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2005;11(7):685–695.
  • Toruner M, Loftus EV Jr., Harmsen WS, et al. Risk factors for opportunistic infections in patients with inflammatory bowel disease. Gastroenterology. 2008;134(4):929–936.
  • Fraser AG, Orchard TR, Jewell DP. The efficacy of azathioprine for the treatment of inflammatory bowel disease: a 30 year review. Gut. 2002;50(4):485–489.
  • Wroblova K, Kolorz M, Batovsky M, et al. Gene polymorphisms involved in manifestation of leucopenia, digestive intolerance, and pancreatitis in azathioprine-treated patients. Dig Dis Sci. 2012;57(9):2394–2401.
  • Lennard L, Van Loon JA, Weinshilboum RM. Pharmacogenetics of acute azathioprine toxicity: relationship to thiopurine methyltransferase genetic polymorphism. Clin Pharmacol Ther. 1989;46(2):149–154.
  • Gearry RB, Barclay ML, Burt MJ, et al. Thiopurine S-methyltransferase (TPMT) genotype does not predict adverse drug reactions to thiopurine drugs in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2003;18(4):395–400.
  • Hindorf U, Lindqvist M, Hildebrand H, et al. Adverse events leading to modification of therapy in a large cohort of patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2006;24(2):331–342.
  • Dubinsky MC, Lamothe S, Yang HY, et al. Pharmacogenomics and metabolite measurement for 6-mercaptopurine therapy in inflammatory bowel disease. Gastroenterology. 2000;118(4):705–713.
  • Shaye OA, Yadegari M, Abreu MT, et al. Hepatotoxicity of 6-mercaptopurine (6-MP) and Azathioprine (AZA) in adult IBD patients. Am J Gastroenterol. 2007;102(11):2488–2494.
  • Sparrow MP, Hande SA, Friedman S, et al. Allopurinol safely and effectively optimizes tioguanine metabolites in inflammatory bowel disease patients not responding to azathioprine and mercaptopurine. Aliment Pharmacol Ther. 2005;22(5):441–446.
  • Belaiche J, Desager JP, Horsmans Y, et al. Therapeutic drug monitoring of azathioprine and 6-mercaptopurine metabolites in Crohn disease. Scand J Gastroenterol. 2001;36(1):71–76.
  • Vernier-Massouille G, Cosnes J, Lemann M, et al. Nodular regenerative hyperplasia in patients with inflammatory bowel disease treated with azathioprine. Gut. 2007;56(10):1404–1409.
  • Seksik P, Mary JY, Beaugerie L, et al. Incidence of nodular regenerative hyperplasia in inflammatory bowel disease patients treated with azathioprine. Inflamm Bowel Dis. 2011;17(2):565–572.
  • Rasmussen HH, Fonager K, Sorensen HT, et al. Risk of acute pancreatitis in patients with chronic inflammatory bowel disease. A Danish 16-year nationwide follow-up study. Scand J Gastroenterol. 1999;34(2):199–201.
  • Bermejo F, Lopez-Sanroman A, Taxonera C, et al. Acute pancreatitis in inflammatory bowel disease, with special reference to azathioprine-induced pancreatitis. Aliment Pharmacol Ther. 2008;28(5):623–628.
  • Siegel CA, Marden SM, Persing SM, et al. Risk of lymphoma associated with combination anti-tumor necrosis factor and immunomodulator therapy for the treatment of Crohn’s disease: a meta-analysis. Clin Gastroenterol Hepatol. 2009;7(8):874–881.
  • Long MD, Herfarth HH, Pipkin CA, et al. Increased risk for non-melanoma skin cancer in patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2010;8(3):268–274.
  • Peyrin-Biroulet L, Khosrotehrani K, Carrat F, et al. Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for inflammatory bowel disease. Gastroenterology. 2011;141(5):1621–28 e1-5.
  • Long MD, Martin CF, Pipkin CA, et al. Risk of melanoma and nonmelanoma skin cancer among patients with inflammatory bowel disease. Gastroenterology. 2012;143(2):390–9 e1.
  • Khan N, Abbas AM, Lichtenstein GR, et al. Risk of lymphoma in patients with ulcerative colitis treated with thiopurines: a nationwide retrospective cohort study. Gastroenterology. 2013;145(5):1007–15 e3.
  • Afif W, Sandborn WJ, Faubion WA, et al. Risk factors for lymphoma in patients with inflammatory bowel disease: a case-control study. Inflamm Bowel Dis. 2013;19(7):1384–1389.
  • Beaugerie L, Brousse N, Bouvier AM, et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Lancet. 2009;374(9701):1617–1625.
  • Wu Y, Ghaly S, Kerr S, et al. UV exposure and skin type are more important than thiopurine exposure for non-melanoma skin cancer risk in IBD. Gastroenterology. 2017;152(5):S576.
  • Bourrier A, Carrat F, Colombel JF, et al. Excess risk of urinary tract cancers in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Aliment Pharmacol Ther. 2016;43(2):252–261.
  • Wahed M, Louis-Auguste JR, Baxter LM, et al. Efficacy of methotrexate in Crohn’s disease and ulcerative colitis patients unresponsive or intolerant to azathioprine/mercaptopurine. Aliment Pharmacol Ther. 2009;30(6):614–620.
  • Saibeni S, Bollani S, Losco A, et al. The use of methotrexate for treatment of inflammatory bowel disease in clinical practice. Dig Liver Dis. 2012;44(2):123–127.
  • Vena GA, Cassano N, Iannone F. Update on subcutaneous methotrexate for inflammatory arthritis and psoriasis. Ther Clin Risk Manag. 2018;14:105–116.
  • Feagan BG, Rochon J, Fedorak RN, et al. Methotrexate for the treatment of Crohn’s disease. The North American Crohn’s study group investigators. N Engl J Med. 1995;332(5):292–297.
  • Alfadhli AA, McDonald JW, Feagan BG. Methotrexate for induction of remission in refractory Crohn’s disease. Cochrane Database Syst Rev. 2005;1:CD003459.
  • Parker R, Dixit A, Fraser A, et al. Clinical experience of methotrexate in Crohn’s disease: response, safety and monitoring of treatment. Postgrad Med J. 2010;86(1014):208–211.
  • Seinen ML, Ponsioen CY, de Boer NK, et al. Sustained clinical benefit and tolerability of methotrexate monotherapy after thiopurine therapy in patients with Crohn’s disease. Clin Gastroenterol Hepatol. 2013;11(6):667–672.
  • Hochberg M, Gravallese E, Silman A, et al. Rheumatology. 7th. Philadelphia: Elsevier, Inc; 2019.
  • Dhir V, Sandhu A, Kaur J, et al. Comparison of two different folic acid doses with methotrexate–a randomized controlled trial (FOLVARI Study). Arthritis Res Ther. 2015;17:156.
  • Kameyama S, Kase Y, Kurihara S, et al. Influence of high-dose folic acid on methotrexate efficacies and safety in Japanese rheumatoid arthritis patients. Drug Res (Stuttg). 2017;67(12):705–709.
  • Herfarth HH, Kappelman MD, Long MD, et al. Use of methotrexate in the treatment of inflammatory bowel diseases. Inflamm Bowel Dis. 2016;22(1):224–233.
  • Mori S, Hidaka M, Kawakita T, et al. Factors associated with myelosuppression related to low-dose methotrexate therapy for inflammatory rheumatic diseases. PLoS One. 2016;11(4):e0154744.
  • Guidelines for monitoring drug therapy in rheumatoid arthritis. American college of rheumatology ad hoc committee on clinical guidelines. Arthritis Rheum. 1996;39(5):723–731.
  • Margagnoni G, Papi V, Aratari A, et al. Methotrexate-induced pneumonitis in a patient with Crohn’s disease. J Crohns Colitis. 2010;4(2):211–214.
  • Berends MA, Snoek J, de Jong EM, et al. Liver injury in long-term methotrexate treatment in psoriasis is relatively infrequent. Aliment Pharmacol Ther. 2006;24(5):805–811.
  • Roenigk HH Jr., Auerbach R, Maibach H, et al. Methotrexate in psoriasis: consensus conference. J Am Acad Dermatol. 1998;38(3):478–485.
  • Skalko RG, Gold MP. Teratogenicity of methotrexate in mice. Teratology. 1974;9(2):159–163.
  • Buckley LM, Bullaboy CA, Leichtman L, et al. Multiple congenital anomalies associated with weekly low-dose methotrexate treatment of the mother. Arthritis Rheum. 1997;40(5):971–973.
  • Hyoun SC, Obican SG, Scialli AR. Teratogen update: methotrexate. Birth Defects Res A Clin Mol Teratol. 2012;94(4):187–207.
  • Hospira, I. Methotrexate Injection, USP. IL: Lake Forest; 2011.
  • Beghin D, Cournot MP, Vauzelle C, et al. Paternal exposure to methotrexate and pregnancy outcomes. J Rheumatol. 2011;38(4):628–632.
  • Johns DG, Rutherford LD, Leighton PC, et al. Secretion of methotrexate into human milk. Am J Obstet Gynecol. 1972;112(7):978–980.
  • Herfarth H, Barnes EL, Jackson S, et al. Methotrexate is not superior to placebo in maintaining remission in patients with ulcerative colitis: results from the MERIT-UC study. Journal of Crohns & Colitis. 2018;12:S300–S1.
  • Carbonnel F, Colombel JF, Filippi J, et al. Methotrexate is not superior to placebo for inducing steroid-free remission, but induces steroid-free clinical remission in a larger proportion of patients with ulcerative colitis. Gastroenterology. 2016;150(2):380–388.
  • Targan SR, Hanauer SB, van Deventer SJ, et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s disease cA2 study group. N Engl J Med. 1997;337(15):1029–1035.
  • Hanauer SB, Feagan BG, Lichtenstein GR, et al. Maintenance infliximab for Crohn’s disease: the ACCENT I randomised trial. Lancet. 2002;359(9317):1541–1549.
  • Colombel JF, Sandborn WJ, Rutgeerts P, et al. Adalimumab for maintenance of clinical response and remission in patients with Crohn’s disease: the CHARM trial. Gastroenterology. 2007;132(1):52–65.
  • Colombel JF, Sandborn WJ, Rutgeerts P, et al. Comparison of two adalimumab treatment schedule strategies for moderate-to-severe Crohn’s disease: results from the CHARM trial. Am J Gastroenterol. 2009;104(5):1170–1179.
  • Sandborn WJ, Feagan BG, Stoinov S, et al. Certolizumab pegol for the treatment of Crohn’s disease. N Engl J Med. 2007;357(3):228–238.
  • Rutgeerts P, Sandborn WJ, Feagan BG, et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005;353(23):2462–2476.
  • Sandborn WJ, van Assche G, Reinisch W, et al. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142(2):257–65 e1-3.
  • Sandborn WJ, Feagan BG, Marano C, et al. Subcutaneous golimumab induces clinical response and remission in patients with moderate to severe ulcerative colitis. Gastroenterology. 2013.
  • Sandborn WJ, Feagan BG, Marano C, et al. Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2013.
  • Colombel JF, Loftus EV Jr., Tremaine WJ, et al. The safety profile of infliximab in patients with Crohn’s disease: the mayo clinic experience in 500 patients. Gastroenterology. 2004;126(1):19–31.
  • Marehbian J, Arrighi HM, Hass S, et al. Adverse events associated with common therapy regimens for moderate-to-severe Crohn’s disease. Am J Gastroenterol. 2009;104(10):2524–2533.
  • Sands BE, Blank MA, Patel K, et al. Long-term treatment of rectovaginal fistulas in Crohn’s disease: response to infliximab in the ACCENT II Study. Clin Gastroenterol Hepatol. 2004;2(10):912–920.
  • Schreiber S, Khaliq-Kareemi M, Lawrance IC, et al. Maintenance therapy with certolizumab pegol for Crohn’s disease. N Engl J Med. 2007;357(3):239–250.
  • Loras C, Gisbert JP, Minguez M, et al. Liver dysfunction related to hepatitis B and C in patients with inflammatory bowel disease treated with immunosuppressive therapy. Gut. 2010;59(10):1340–1346.
  • Katz LH, Fraser A, Gafter-Gvili A, et al. Lamivudine prevents reactivation of hepatitis B and reduces mortality in immunosuppressed patients: systematic review and meta-analysis. J Viral Hepat. 2008;15(2):89–102.
  • European Association For The Study Of The, L. EASL clinical practice guidelines: management of chronic hepatitis B. J Hepatol. 2009;50(2):227–242.
  • Lok AS, McMahon BJ. Chronic hepatitis B: update 2009. Hepatology. 2009;50(3):661–662.
  • Xie X, Li F, Chen JW, et al. Risk of tuberculosis infection in anti-TNF-alpha biological therapy: from bench to bedside. J Microbiol Immunol Infect. 2014;47(4):268–274.
  • Kalyoncu U, Karadag O, Akdogan A, et al. Pneumocystis carinii pneumonia in a rheumatoid arthritis patient treated with adalimumab. Scand J Infect Dis. 2007;39(5):475–478.
  • Abreu C, Magro F, Vilas-Boas F, et al. Listeria infection in patients on anti-TNF treatment: report of two cases and review of the literature. J Crohns Colitis. 2013;7(2):175–182.
  • Ford AC, Peyrin-Biroulet L. Opportunistic infections with anti-tumor necrosis factor-alpha therapy in inflammatory bowel disease: meta-analysis of randomized controlled trials. Am J Gastroenterol. 2013;108(8):1268–1276.
  • Schaible TF. Long term safety of infliximab. Can J Gastroenterol. 2000;14(Suppl C):29C–32C.
  • Cheifetz A, Smedley M, Martin S, et al. The incidence and management of infusion reactions to infliximab: a large center experience. Am J Gastroenterol. 2003;98(6):1315–1324.
  • Lichtenstein GR, Feagan BG, Cohen RD, et al. Serious infection and mortality in patients with Crohn’s disease: more than 5 years of follow-up in the TREAT registry. Am J Gastroenterol. 2012;107(9):1409–1422.
  • Lecluse LL, Piskin G, Mekkes JR, et al. Review and expert opinion on prevention and treatment of infliximab-related infusion reactions. Br J Dermatol. 2008;159(3):527–536.
  • Picoraro J, Winberry G, Siegel CA, et al. Premedication use before infliximab administration: a cross-sectional analysis. Inflamm Bowel Dis. 2017;23(1):174–180.
  • Nash P, Vanhoof J, Hall S, et al. Randomized crossover comparison of injection site pain with 40 mg/0.4 or 0.8 mL formulations of adalimumab in patients with rheumatoid arthritis. Rheumatol Ther. 2016;3(2):257–270.
  • Rahier JF, Buche S, Peyrin-Biroulet L, et al. Severe skin lesions cause patients with inflammatory bowel disease to discontinue anti-tumor necrosis factor therapy. Clin Gastroenterol Hepatol. 2010;8(12):1048–1055.
  • Kip KE, Swoger JM, Grandinetti LM, et al. Tumor necrosis factor alpha antagonist-associated psoriasis in inflammatory diseases: an analysis of the FDA adverse event reporting system. Inflamm Bowel Dis. 2013;19(6):1164–1172.
  • Collamer AN, Guerrero KT, Henning JS, et al. 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.
  • Ko JM, Gottlieb AB, Kerbleski JF. Induction and exacerbation of psoriasis with TNF-blockade therapy: a review and analysis of 127 cases. J Dermatolog Treat. 2009;20(2):100–108.
  • Sarzi-Puttini P, Atzeni F, Capsoni F, et al. Drug-induced lupus erythematosus. Autoimmunity. 2005;38(7):507–518.
  • Beigel F, Schnitzler F, Paul Laubender R, et al. Formation of antinuclear and double-strand DNA antibodies and frequency of lupus-like syndrome in anti-TNF-alpha antibody-treated patients with inflammatory bowel disease. Inflamm Bowel Dis. 2011;17(1):91–98.
  • Katz U, Zandman-Goddard G. Drug-induced lupus: an update. Autoimmun Rev. 2010;10(1):46–50.
  • Thomas CW Jr., Weinshenker BG, Sandborn WJ. Demyelination during anti-tumor necrosis factor alpha therapy with infliximab for Crohn’s disease. Inflamm Bowel Dis. 2004;10(1):28–31.
  • Enayati PJ, Papadakis KA. Association of anti-tumor necrosis factor therapy with the development of multiple sclerosis. J Clin Gastroenterol. 2005;39(4):303–306.
  • Kaltsonoudis E, Voulgari PV, Konitsiotis S, et al. Demyelination and other neurological adverse events after anti-TNF therapy. Autoimmun Rev. 2014;13(1):54–58.
  • Kemanetzoglou E, Andreadou E. CNS demyelination with TNF-alpha blockers. Curr Neurol Neurosci Rep. 2017;17(4):36.
  • Kwon HJ, Cote TR, Cuffe MS, et al. Case reports of heart failure after therapy with a tumor necrosis factor antagonist. Ann Intern Med. 2003;138(10):807–811.
  • Inc., A. HUMIRA injection [package insert]. Chicago, IL: AbbVie Inc; 2013.
  • Jannsen biotech, I. Remicade (infliximab) [Prescribing information]. NJ: Bridgewater; 2013.
  • UCB, I. Cimzia injection [package insert]. Smyrna, GA; 2013.
  • Singh S, Nagpal SJ, Murad MH, et al. Inflammatory bowel disease is associated with an increased risk of melanoma: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2014;12(2):210–218.
  • Wolfe F, Michaud K. The effect of methotrexate and anti-tumor necrosis factor therapy on the risk of lymphoma in rheumatoid arthritis in 19,562 patients during 89,710 person-years of observation. Arthritis Rheum. 2007;56(5):1433–1439.
  • Herrinton LJ, Liu L, Weng X, et al. Role of thiopurine and anti-TNF therapy in lymphoma in inflammatory bowel disease. Am J Gastroenterol. 2011;106(12):2146–2153.
  • Mariette X, Matucci-Cerinic M, Pavelka K, et al. Malignancies associated with tumour necrosis factor inhibitors in registries and prospective observational studies: a systematic review and meta-analysis. Ann Rheum Dis. 2011;70(11):1895–1904.
  • Lopez-Olivo MA, Tayar JH, Martinez-Lopez JA, et al. Risk of malignancies in patients with rheumatoid arthritis treated with biologic therapy: a meta-analysis. JAMA. 2012;308(9):898–908.
  • Wadstrom H, Frisell T, Askling J. Malignant neoplasms in patients with rheumatoid arthritis treated with tumor necrosis factor inhibitors, tocilizumab, abatacept, or rituximab in clinical practice: a nationwide cohort study from Sweden. JAMA Intern Med. 2017;177(11):1605–1612.
  • Lemaitre M, Kirchgesner J, Rudnichi A, et al. Association between use of thiopurines or tumor necrosis factor antagonists alone or in combination and risk of lymphoma in patients with inflammatory bowel disease. JAMA. 2017;318(17):1679–1686.
  • Narula N, Charleton D, Marshall JK. Meta-analysis: peri-operative anti-TNFalpha treatment and post-operative complications in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2013;37(11):1057–1064.
  • Syed A, Cross RK, Flasar MH. Anti-tumor necrosis factor therapy is associated with infections after abdominal surgery in Crohn’s disease patients. Am J Gastroenterol. 2013;108(4):583–593.
  • Ali T, Yun L, Rubin DT. Risk of post-operative complications associated with anti-TNF therapy in inflammatory bowel disease. World J Gastroenterol. 2012;18(3):197–204.
  • Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369(8):699–710.
  • Sandborn WJ, Feagan BG, Rutgeerts P, et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease. New England J Med. 2013;369(8):711–721.
  • Christensen B, Colman RJ, Micic D, et al. Vedolizumab as induction and maintenance for inflammatory bowel disease: 12-month effectiveness and safety. Inflamm Bowel Dis. 2018;24(4):849–860.
  • Colombel J-F, Sands BE, Rutgeerts P, et al. The safety of vedolizumab for ulcerative colitis and Crohn’s disease. Gut. 2017;66(5):839–851.
  • Feagan BG, Sandborn WJ, Gasink C, et al. Ustekinumab as induction and maintenance therapy for Crohn’s disease. New England J Med. 2016;375(20):1946–1960.
  • Papp KA, Griffiths CE, Gordon K, et al. Long-term safety of ustekinumab in patients with moderate-to-severe psoriasis: final results from 5 years of follow-up. Br J Dermatol. 2013;168(4):844–854.
  • 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):706–714.
  • Kawalec P, Mocko P, Malinowska-Lipien I, et al. Efficacy and safety of ustekinumab in the induction therapy of TNF-alpha-refractory Crohn’s disease patients: a systematic review and meta-analysis. J Comp Eff Res. 2017;6(7):601–612.
  • Siegel CA. Lost in translation: helping patients understand the risks of inflammatory bowel disease therapy. Inflamm Bowel Dis. 2010;16(12):2168–2172.
  • Johnson FR, Hauber B, Ozdemir S, et al. Are gastroenterologists less tolerant of treatment risks than patients? Benefit-risk preferences in Crohn’s disease management. J Manag Care Pharm. 2010;16(8):616–628.
  • Siegel CA. Making therapeutic decisions in IBD: the role of patients. Curr Opin Gastroenterol. 2009;25(4):334–338.
  • Munkholm P, Langholz E, Davidsen M, et al. Intestinal cancer risk and mortality in patients with Crohn’s disease. Gastroenterology. 1993;105(6):1716–1723.
  • Afif W, Loftus EV Jr., Faubion WA, et al. Clinical utility of measuring infliximab and human anti-chimeric antibody concentrations in patients with inflammatory bowel disease. Am J Gastroenterol. 2010;105(5):1133–1139.
  • Sandborn WJ. Crohn’s disease evaluation and treatment: clinical decision tool. Gastroenterology. 2014;147(3):702–705.
  • Dassopoulos T, Cohen RD, Scherl EJ, et al. Ulcerative colitis care pathway. Gastroenterology. 2015;149(1):238–245.
  • O’Shea JJ, Kontzias A, Yamaoka K, et al. Janus kinase inhibitors in autoimmune diseases. Ann Rheum Dis. 2013;72(2):2012–202576.
  • Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376(18):1723–1736.
  • Cohen SB, Tanaka Y, Mariette X, et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis. 2017;76(7):1253–1262.
  • Sands BE, Panés J, Higgins PDR, et al. 14 Post-hoc analysis of tofacitinib Crohn’s disease phase 2 induction efficacy in subgroups with baseline endoscopic or biomarker evidence of inflammation. Gastroenterology. 2018;154(1):S81.
  • De Vries LCS, Wildenberg ME, De Jonge WJ, et al. The future of Janus kinase inhibitors in inflammatory bowel disease. Journal of Crohn’s and Colitis. 2017;11(7):885–893.
  • Vermeire S, Schreiber S, Petryka R, et al. Clinical remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet. 2017;389(10066):266–275.

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.